Fact-checked by Grok 2 weeks ago

Angiotensin II receptor

The angiotensin II receptors are a class of G protein-coupled receptors (GPCRs) that bind the octapeptide hormone angiotensin II, serving as key mediators in the renin-angiotensin-aldosterone system (RAAS) to regulate , fluid and balance, vascular tone, and cardiovascular . These receptors, primarily the type 1 (AT1R) and type 2 (AT2R) subtypes, exhibit distinct but sometimes opposing physiological effects, with AT1R driving most vasoconstrictive and pro-inflammatory actions while AT2R promotes and tissue protection. The existence of specific receptors for was first demonstrated in the 1970s through radioligand binding studies. The AT1 receptor was cloned in 1991 from vascular cells, revealing its role as the primary mediator of angiotensin II's effects in adults. The AT2 receptor was cloned in 1993, noted for its distinct expression and functions. was standardized in the mid-1990s by an international consensus, designating the subtypes as AT1 and AT2 to reflect their pharmacological profiles, replacing earlier designations like type I/III and type II.

Introduction

Definition and physiological role

Angiotensin II receptors are a class of G-protein-coupled receptors (GPCRs) that bind angiotensin II (Ang II), the primary bioactive of the renin-angiotensin-aldosterone (RAAS), a hormonal cascade essential for cardiovascular and renal . These receptors, predominantly the AT1 and AT2 subtypes, transduce Ang II signals across membranes to elicit diverse cellular responses in target organs such as the vasculature, kidneys, and adrenal glands. As members of the class A (rhodopsin-like) GPCR superfamily, they exhibit evolutionary conservation across vertebrates, reflecting their fundamental role in physiological adaptation to environmental stressors like dehydration and hemorrhage. Ang II itself is an octapeptide produced through sequential enzymatic cleavage: angiotensinogen is first converted to the decapeptide I by renin, which is then hydrolyzed to Ang II by (), mainly in the pulmonary and renal tissues. Upon binding to its receptors, Ang II initiates signaling cascades that primarily activate Gq/11 proteins, leading to activation, production, and subsequent intracellular calcium mobilization. This receptor-mediated activation ensures precise control over Ang II's effects, preventing indiscriminate hormonal action and allowing tissue-specific responses. The core physiological roles of angiotensin II receptors center on maintaining , fluid volume, and balance. Through AT1 receptor activation, they promote to elevate vascular tone and systemic pressure, while also stimulating aldosterone release from the to enhance and water retention. In the , receptor signaling modulates glomerular filtration and tubular reabsorption, fine-tuning volume. Notably, the two main subtypes exhibit partially counter-regulatory functions: AT1 drives pressor and growth-promoting effects, whereas AT2 often opposes these by promoting and , contributing to overall system balance.

Historical discovery and nomenclature

The discovery of the renin-angiotensin-aldosterone system (RAAS), of which angiotensin II (Ang II) receptors serve as key mediators, traces back to 1898 when Robert Tigerstedt and Per Bergman identified renin as a pressor substance in extracts that elevated upon injection. Subsequent work in by Irvine Page and colleagues revealed that renin acted on a —later termed angiotensinogen—to produce a heat-stable vasopressor factor, setting the stage for understanding Ang II as the system's primary effector. By the early 1950s, Leonard Skeggs and his team purified and characterized two forms of angiotensin: the inactive decapeptide angiotensin I and the active octapeptide Ang II, confirming a enzymatic conversion process involving a converting , which highlighted the receptors' role in transducing Ang II's physiological effects such as and aldosterone release. Receptor studies advanced in the 1970s with the development of radiolabeled Ang II for binding assays, enabling the identification of specific high-affinity sites in vascular, adrenal, and renal tissues. These assays, pioneered by researchers like Pierre Meyer and colleagues, demonstrated receptor heterogeneity and downregulation by Ang II itself, as reported in studies from 1976 onward. Pharmacological differentiation in the 1980s using peptide analogs revealed two distinct subtypes: AT1, predominant in adult tissues and mediating classic RAAS actions, and AT2, more abundant in fetal tissues with opposing effects. The molecular era began with the cloning of the AT1 receptor in 1991 by Koji Sasaki and colleagues from bovine adrenal cortex cDNA, followed by the AT2 receptor cloning in 1993 by Mitsuo Kambayashi's group from rat pheochromocytoma cells, establishing both as G protein-coupled receptors (GPCRs) within the rhodopsin-like family. Nomenclature evolved from generic "angiotensin receptors" to the standardized AT1 and AT2 designations proposed in 1991 by the Committee of the Council for High Research, chaired by F. Bumpus, based on pharmacological profiles and molecular sequences. Early proposals in the introduced AT3 and subtypes for atypical binding sites—AT3 for certain non-mammalian or variant responses, and for high-affinity Ang IV (Ang II[3-8]) sites—but subsequent reclassification showed AT3 lacked distinct molecular identity, while was identified as insulin-regulated (IRAP), a non-GPCR , rather than a true Ang II receptor. Key pharmacological milestones included the synthesis of losartan in 1986 by scientists and others, the first selective non-peptide AT1 , which facilitated subtype validation and led to its clinical approval in 1995. More recently, cryo-electron microscopy (cryo-EM) structures in the 2020s, such as the 2023 resolution of the human AT1R-Gq-Sar1-Ang II complex at 2.9 Å by Dan Zhang and colleagues, have provided atomic-level insights into ligand-receptor interactions, building on earlier efforts.

Molecular Structure

Overall architecture

Angiotensin II receptors belong to the (GPCR) superfamily and exhibit the canonical architecture characteristic of class A (rhodopsin-like) GPCRs, consisting of seven transmembrane alpha-helices (TM1–TM7) that form a bundle embedded in the . These receptors feature an extracellular N-terminal domain, an intracellular C-terminal tail, three extracellular loops (ECL1–ECL3) connecting the transmembrane helices on the outer side, and three intracellular loops (ICL1–ICL3) on the cytoplasmic side that facilitate interactions with intracellular signaling partners. This topological arrangement positions the ligand-binding site within the transmembrane bundle and enables across the membrane upon activation. Key conserved structural features include the orthosteric binding pocket located deep within the TM bundle, primarily formed by residues from TM2, TM3, TM6, and TM7, which accommodates the peptide ligand . G-protein coupling occurs primarily through the ICL2 and ICL3 regions, as well as the C-terminal tail, allowing recruitment of heterotrimeric G proteins such as Gq/11 for AT1R and Gi/o for AT2R. Additionally, angiotensin II receptors possess the potential for dimerization, with interfaces involving TM1, TM4, and TM5, which can influence receptor trafficking and signaling efficacy. The primary sequence of angiotensin II receptors comprises approximately 350–363 , resulting in a calculated molecular weight of around 40–41 for the unglycosylated form. Post-translational modifications, including N-linked sites on the extracellular (e.g., at Asn residues in the first 30 ), contribute to receptor maturation, stability, and surface expression, often increasing the apparent molecular weight to 60–100 in glycosylated forms. Structural determination has advanced significantly in recent years, beginning with the 2015 room-temperature of the AT1 receptor in complex with the ZD7155 at 2.9 Å (PDB: 4YAY), which confirmed the 7TM helical bundle and revealed the ligand-binding pocket's conformation in the inactive state. Subsequent cryo-electron microscopy (cryo-EM) studies in the have resolved active conformations, such as the AT1 receptor bound to II and protein (e.g., PDB: 7F6G at 3.2 Å), delineating both orthosteric and allosteric sites involved in peptide recognition and G-protein engagement. For the AT2 receptor, such as the 2019 inactive-state structure in complex with an analog (PDB: 6DO1) and with II (PDB: 5XJ1) reveal a similar 7TM but with distinct features, including a different ligand-binding pose and repositioning of 8 that may inhibit canonical coupling. These high-resolution structures have provided a foundational template for understanding the overall shared by angiotensin II receptor subtypes.

Ligand binding and activation mechanisms

The orthosteric binding pocket of the angiotensin II receptor, a (GPCR), is primarily formed by transmembrane helices TM2, TM3, TM6, and TM7, along with the extracellular loop 2 (ECL2), which contributes to recognition and subtype selectivity. Angiotensin II (Ang II), the endogenous , binds in a manner that positions its C-terminal carboxylate group deep within this pocket, forming critical hydrogen bonds with key residues such as Arg167 in ECL2 and Asn111 in TM3, stabilizing the interaction and facilitating receptor engagement. These interactions, conserved across subtypes like AT1 and AT2, enable the peptide's to extend toward the extracellular surface while the core sequence anchors within the . Upon binding, the receptor undergoes a conformational shift from an inactive state, characterized by an open extracellular vestibule, to an active state with a closed extracellular region and an open intracellular crevice for coupling. This is driven by the outward movement of TM6 by approximately 14 Å, a hallmark GPCR that disrupts ionic locks and repositions residues like the NPxxY in TM7, propagating the signal intracellularly. In AT1 receptors, Ang II induces this transition through direct contacts with the toggle switch residue Trp253^{6.48}, promoting helix rearrangements essential for efficacy. Allosteric modulation fine-tunes efficacy and selectivity at the angiotensin II receptor; for instance, binding in the transmembrane core stabilizes inactive conformations, reducing Ang II potency, while intracellular sodium ions allosterically inhibit by preserving ionic interactions in the inactive state. Biased ligands, such as TRV027, exploit these sites to preferentially activate β-arrestin pathways over signaling, altering the conformational landscape without fully engaging the orthosteric pocket. The binding affinity of Ang II to the receptor typically exhibits a (K_d) in the range of 0.5-2 nM, reflecting high potency suitable for physiological concentrations. This affinity can be modeled using the Langmuir binding isotherm, which describes fractional receptor (θ) as a function of concentration [L]: \theta = \frac{[L]}{K_d + [L]} This equation derives from the under equilibrium conditions, where θ approaches 1 at saturating [L], providing a basis for dose-response analyses in receptor .

Receptor Subtypes

AT1 Receptor

The AT1 receptor, also known as the angiotensin II type 1 receptor, is encoded by the AGTR1 gene located on the long arm of human at position 3q21-25. This gene spans approximately 45 kb and consists of five exons, with multiple transcription initiation sites contributing to its regulation. In humans, AGTR1 produces a single isoform of the AT1 receptor, whereas exhibit resulting in two highly homologous subtypes: AT1A (Agtr1a) and AT1B (Agtr1b), which share approximately 94-95% sequence identity but display tissue-specific expression patterns. These isoforms in arise from distinct chromosomal loci—Agtr1a on and Agtr1b on —allowing for nuanced physiological responses in experimental models. Structurally, the human AT1 receptor is a seven-transmembrane comprising 359 , with a calculated of approximately 41 prior to post-translational modifications such as N-glycosylation at three sites (Asn4, Asn176, and Asn188), which facilitate proper folding and plasma membrane trafficking. The receptor's extracellular and intracellular flank the transmembrane helices (TM1-TM7), with key ligand-interaction sites in the helical bundle. A notable residue for subtype selectivity and binding is Lys199 in transmembrane helix 5 (TM5), which forms salt bridges with the acidic moieties of non-peptide antagonists, contributing to the receptor's high affinity for these compounds over the AT2 subtype. This structural feature underscores the AT1 receptor's distinct pharmacological profile within the angiotensin receptor family. The AT1 receptor exhibits widespread tissue distribution, with particularly high expression in cardiovascular and renal tissues critical to the renin-angiotensin-aldosterone system (RAAS). In the vasculature, it is abundantly present in cells, where it mediates contractile responses; in the , expression is prominent in juxtaglomerular cells, proximal tubules, and glomeruli; and in the , it drives aldosterone secretion. Cardiac myocytes and fibroblasts also express high levels, supporting its role in myocardial remodeling, while hepatic expression influences and . In the , AT1 receptors are enriched in circumventricular organs such as the , organum vasculosum of the , and , which lack a blood-brain barrier and facilitate neuroendocrine integration of circulating II signals. The AT1 receptor accounts for approximately 80-90% of the physiological effects of angiotensin II, including vasoconstriction, aldosterone release, and sodium retention, making it the primary mediator of RAAS-dependent . It demonstrates particular sensitivity to non-peptide antagonists such as losartan and , which bind with high selectivity (often >10,000-fold over AT2) due to interactions at sites like Lys199, enabling effective without agonist activity. This pharmacological distinction has positioned AT1 as the key therapeutic target for modulating angiotensin II actions in clinical settings.

AT2 Receptor

The angiotensin II type 2 receptor (AT2R), encoded by the AGTR2 gene located on the at Xq23, is a single-isoform protein consisting of 363 . The gene spans approximately 5 kb with three exons, where the third exon encodes the full receptor sequence. As a member of the seven-transmembrane G-protein-coupled receptor superfamily, AT2R shares about 34% overall sequence with the AT1 receptor, though the s exhibit greater conservation to support similar binding topology, while the extracellular and intracellular domains diverge significantly. Structurally, AT2R features unique residues such as Phe308 in the seventh , which contributes to its specific interactions with angiotensin II (Ang II). AT2R expression is developmentally regulated, with high levels predominant in fetal tissues including the , , and , where it plays a key role during . In adults, expression declines markedly and is restricted to specific sites such as the brain (particularly the medulla and ), adrenal , uterine , kidney tubules, and atretic ovaries, with notably low abundance in the vasculature compared to AT1R. This pattern underscores its counterbalancing role to AT1R, often opposing growth-promoting effects in a tissue-specific manner. Distinctive features of AT2R include its higher binding affinity for Ang II, with a dissociation constant (Kd) of approximately 0.45 nM, enabling sensitive detection of the ligand. In adult tissues, AT2R typically constitutes 10-20% of total Ang II receptors, reflecting its minor but functionally significant presence. Unlike AT1R, AT2R is resistant to blockade by common AT1 receptor antagonists such as losartan, allowing selective preservation of its effects. Additionally, AT2R exhibits potential for homo- and heterodimerization with AT1R, which may modulate receptor trafficking and signaling efficiency in co-expressing cells. The angiotensin II receptor family extends beyond the canonical AT1 and AT2 subtypes to include non-canonical receptors historically associated with angiotensin II fragments or alternative RAAS components, though many remain hypothetical or reclassified. The AT3 receptor, proposed in early 1990s reports based on unique binding pharmacology in certain tissues, has not been cloned or molecularly identified, and current evidence attributes such observations to artifacts, experimental variability, or functional heterodimers of AT1 and AT2 receptors. These early findings suggested a third subtype with distinct signaling, but subsequent studies failed to confirm its existence as a separate entity. In contrast, the AT4 receptor has been definitively identified as insulin-regulated aminopeptidase (IRAP), a type II transmembrane zinc metallopeptidase encoded by the LNPEP gene on chromosome 5q15.2. IRAP selectively binds angiotensin IV (Ang IV, the Ang II 3-8 fragment) with high affinity, acting as an enzyme inhibitor rather than a classical GPCR. It is widely distributed in tissues including the brain (particularly hippocampus), kidney, and heart, where it localizes to intracellular vesicles and plasma membranes. Ang IV binding to IRAP mediates renal effects such as increased cortical blood flow and reduced sodium transport in proximal tubules, contributing to local hemodynamic regulation within the kidney. In the brain, IRAP facilitates cognitive processes, including learning and memory enhancement, by modulating neuronal activity in regions like the hippocampus. The Mas receptor, a G protein-coupled receptor encoded by the MAS1 gene on chromosome 7p21.3, serves as the primary receptor for angiotensin (1-7), a counter-regulatory peptide in the RAAS that opposes many Ang II actions. Expressed prominently in the kidney, heart, and brain, Mas shares limited sequence homology (approximately 19% with AT2 and less with AT1) but couples to similar G protein pathways. Activation of Mas by Ang (1-7) promotes vasodilation through endothelial nitric oxide synthase stimulation and nitric oxide release, supporting vasoprotective effects. Recent 2020s research has explored the (pro)renin receptor, encoded by ATP6AP2, as a potential functional analog to the hypothetical AT3 in non-canonical RAAS signaling, particularly in prorenin-mediated pathways independent of Ang II generation. This receptor, a single-pass , influences renal and cardiovascular responses but lacks direct Ang II binding, highlighting extensions of RAAS beyond traditional subtypes.

Functions and Signaling

AT1 Receptor functions and pathways

The AT1 receptor primarily couples to the Gq/11 protein family upon activation by angiotensin II, leading to the stimulation of (PLC), which hydrolyzes (PIP2) into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 subsequently binds to IP3 receptors on the , triggering intracellular calcium (Ca²⁺) release, while DAG activates (PKC), which phosphorylates downstream targets to amplify signaling. This Gq/11-PLC-IP3/DAG pathway forms the core of acute AT1-mediated responses, such as rapid cellular excitation. In addition to Gq/11 coupling, the AT1 receptor engages G12/13 proteins to activate RhoA guanine nucleotide exchange factors, promoting RhoA-mediated cytoskeletal reorganization and formation. G protein-independent signaling occurs via β-arrestin recruitment, which scaffolds and activates (MAPK)/extracellular signal-regulated kinase (ERK) pathways, contributing to sustained proliferative signals. The receptor also activates /signal transducer and activator of transcription (JAK/STAT) pathways through direct JAK2 association, driving transcription for and . Furthermore, AT1 stimulation upregulates NADPH oxidase subunits (e.g., Nox1 and p22phox), generating (ROS) that propagate oxidative signaling. These signaling cascades underpin key physiological effects of AT1 activation. In vascular smooth muscle cells, the Gq/11-mediated Ca²⁺ surge induces myosin light chain phosphorylation, resulting in potent vasoconstriction that elevates blood pressure. In the adrenal , AT1 signaling, particularly via β-arrestin and ERK, stimulates aldosterone biosynthesis and secretion, enhancing sodium retention and potassium excretion. Renal AT1 receptors in the and collecting ducts promote sodium reabsorption through modulation of ion transporters, contributing to fluid volume . In cardiomyocytes, chronic AT1 activation via MAPK/ERK and ROS pathways induces hypertrophic gene expression, leading to cardiac remodeling. Centrally, AT1 receptors in the and drive and antidiuretic hormone (ADH) release, integrating behavioral and endocrine responses to . The dose-dependent nature of Ca²⁺ signaling exemplifies the pathway's sensitivity to angiotensin II levels, with intracellular Ca²⁺ concentration ([Ca²⁺]ᵢ) rising in response to IP3 production. AT1 receptors dominate angiotensin II effects in the adult renin-angiotensin-aldosterone (RAAS), mediating most of its cardiovascular and renal actions, with AT2 providing counter-regulatory . Biased at AT1 offers therapeutic nuance; for instance, the β-arrestin-biased TRV027, developed in the , preferentially activates ERK without strong /11 coupling, potentially mitigating while preserving contractility.

AT2 Receptor functions and pathways

The angiotensin II type 2 receptor (AT2R) primarily signals through coupling to inhibitory G proteins (Gi/o), which inhibits adenylyl cyclase activity and thereby reduces intracellular cyclic AMP (cAMP) levels, counteracting proliferative signals. Additionally, AT2R activation stimulates protein phosphatase 2A (PP2A), promoting dephosphorylation of target proteins and inhibiting mitogen-activated protein kinase (MAPK) pathways, such as extracellular signal-regulated kinase (ERK), to suppress cell growth and inflammation. A key vasodilatory mechanism involves stimulation of nitric oxide synthase (NOS), leading to nitric oxide (NO) production and subsequent elevation of cyclic guanosine monophosphate (cGMP) via guanylyl cyclase, ultimately contributing to vascular smooth muscle relaxation. These pathways often occur independently of β-arrestin recruitment, allowing sustained signaling without rapid desensitization. Physiologically, AT2R activation induces in endothelial and vascular cells through the NO-cGMP , opposing the vasoconstrictive effects of the AT1R. In the , it promotes anti-proliferative and pro-apoptotic effects, reducing vascular remodeling and . Renal AT2R stimulation enhances by increasing sodium excretion and renal blood flow, partly via NO-mediated inhibition of Na+/K+-. During fetal development, AT2R plays a crucial role in tissue morphogenesis, particularly in the and , where it supports neuronal and organ maturation through phosphatase-dependent pathways. In the heart, it exerts anti-fibrotic effects by decreasing deposition and accumulation. Beyond core signaling, AT2R engages in pathway , including of the PTEN/PI3K axis to limit pro-survival signals and enhancement of (BMP) signaling to promote tissue repair. Unique aspects of AT2R include its upregulation following , such as in or , where it facilitates regeneration and via NO-cGMP modulation. As an X-linked gene, AT2R exhibits sex differences, with females often displaying amplified protective effects due to interactions. Recent 2025 research highlights how selective agonists like compound 21 (C21) amplify these neuroprotective functions, enhancing in the ventricular-subventricular zone post-.

Clinical Significance

Role in diseases

Dysregulation of angiotensin II receptors, particularly the AT1 subtype, plays a central role in the of various diseases through imbalances in the renin-angiotensin-aldosterone system (RAAS), which contributes to the majority of cases. Overactivation of AT1 receptors promotes , , and tissue remodeling, while reduced AT2 receptor activity may exacerbate these effects by diminishing counter-regulatory protection. In , AT1 receptor overexpression in vascular tissues leads to sustained and elevated . High salt intake, for instance, induces vascular AT1 receptor mRNA and protein density increases of up to 160%, enhancing II-mediated pressor responses. Genetic variants such as the AGTR1 A1166C polymorphism (rs5186) are associated with increased risk of and greater , with the C linked to enhanced receptor function and disease susceptibility in multiple populations. In cardiovascular diseases, AT1 receptor signaling drives pathological cardiac and in , where sustained stimulation upregulates growth factors like transforming growth factor-β, promoting myocardial remodeling and systolic dysfunction. This contributes to adverse outcomes in conditions like , where AT1 activation exacerbates plaque formation through inflammatory pathways. In contrast, AT2 receptors exert protective effects against ischemia-reperfusion injury by suppressing immune responses and reducing infarct size in myocardial tissue. Renal diseases involving angiotensin II receptors include , where AT1 activation causes damage and glomerular injury via upregulation of receptors for and increased . This leads to and progressive kidney dysfunction. AT2 receptors, however, provide renoprotection by promoting anti-inflammatory cytokine production, such as interleukin-10, and limiting in models of . Beyond these, AT1 receptors contribute to brain inflammation in , where their activation amplifies microglial responses and amyloid-β neurotoxicity, accelerating cognitive decline. In cancer, AT1 signaling promotes tumor by stimulating expression and endothelial cell proliferation, supporting tumor growth in models like breast and . Polymorphisms in angiotensin II receptor genes, including AGTR1 A1166C, are linked to increased risk, with certain alleles associated with heightened maternal vascular sensitivity and placental dysfunction. AT2 receptor agonists, such as Compound 21 (C21), have demonstrated potential in improving respiratory function in patients with lung involvement, as shown in a 2021 phase 2 trial.

Therapeutics and pharmacology

Angiotensin II receptor blockers (ARBs), such as losartan and , are non-peptide competitive antagonists that selectively bind to the AT1 receptor subtype, producing surmountable blockade by preventing -induced and aldosterone release. Losartan, the first ARB approved by the FDA in 1995, is indicated for , with reduced , and , where it reduces and slows renal progression. , approved subsequently, shares similar indications, including post-myocardial infarction left ventricular dysfunction and , demonstrating comparable efficacy to inhibitors in lowering and cardiovascular events with better tolerability. Common side effects of ARBs include , , and , particularly in patients with renal impairment or volume depletion, necessitating monitoring of and renal function. Agonists targeting the AT2 receptor represent an emerging therapeutic class, countering the pathological effects of AT1 activation through anti-inflammatory and antifibrotic mechanisms. , a selective non-peptide AT2 receptor , has advanced to phase II clinical trials in the 2020s for , where it improved lung function and reduced fibrosis markers in early studies, and for acute ischemic stroke, showing neuroprotective effects in preclinical models. As of August 2025, the phase 2b ASPIRE trial (NCT06588686) for is ongoing, with full enrollment expected by the first half of 2026; phase 2a results demonstrated a mean increase in of 216 mL over 36 weeks with no treatment-related serious adverse events. Unlike peptide-based AT2 ligands, C21 offers oral and high selectivity, avoiding with AT1 or Mas receptors, though its development focuses on conditions like and without broad approval as of 2025. Beyond subtype-specific agents, direct infusion (Giapreza) was FDA-approved in 2017 for adults with septic or other refractory to catecholamines, acting as a potent vasoconstrictor via both AT1 and AT2 receptors to rapidly increase . Mas receptor agonists, such as the non-peptide AVE 0991, mimic angiotensin-(1-7) to activate the counter-regulatory arm of the renin--aldosterone system, exhibiting cardioprotective and renoprotective effects in preclinical models of and ischemia without AT1 . Dual AT1/AT2 modulators are under investigation for enhanced efficacy in and cardiovascular disorders by targeting both receptor subtypes. Pharmacodynamically, ARBs exert at the orthosteric binding site of the AT1 receptor, shifting the II dose-response curve rightward without altering the maximum response, as quantified by the K_b. The potency is expressed as \mathrm{pA_2} = -\log_{10}(K_b), where K_b is derived from Schild plot analysis, plotting \log(\mathrm{DR} - 1) against the negative logarithm of concentration, yielding a near unity for competitive . Recent advancements include studies on biased signaling at the AT1 receptor, with 2025 research elucidating gating mechanisms for biased agonism that promote distinct signaling pathways. Combinations such as , an angiotensin receptor-neprilysin inhibitor approved for , pair valsartan's AT1 blockade with inhibition to enhance natriuretic peptides, outperforming monotherapy in reducing hospitalizations.

References

  1. [1]
    Angiotensin Receptors: Structure, Function, Signaling and Clinical ...
    Two well characterized receptors are angiotensin type 1 receptor (AT1 receptor) and type 2 receptor (AT2 receptor). They respond to the octapeptide hormone ...
  2. [2]
    Structural insights into ligand recognition and activation of ...
    G protein-coupled angiotensin II receptors, AT1R and AT2R, are integral components of the renin–angiotensin system (RAS) that regulates blood pressure and fluid ...
  3. [3]
    Role of the Angiotensin Type 2 Receptor in the Regulation of Blood ...
    Abstract—The renin-angiotensin system is a major physiological regulator of body fluid volume, electrolyte balance, and arterial pressure.
  4. [4]
    Evolutionary information helps understand distinctive features of the ...
    Feb 24, 2022 · We analyze the evolution of two G protein-coupled receptors, AT1 and AT2, which bind the angiotensin II peptide and are important regulators of the ...Results · Properties Of D2. 50 In At1... · Sodium Binding Mode In The...<|control11|><|separator|>
  5. [5]
    Angiotensin: What It Is, Causes & Function - Cleveland Clinic
    Angiotensin is a hormone that helps regulate blood pressure by constricting (narrowing) blood vessels and triggering water and salt (sodium) intake.
  6. [6]
    Angiotensin II Receptor Physiology Using Gene Targeting
    Angiotensin II regulates blood pressure through its actions as a potent vasoconstrictor, its ability to modulate sodium reabsorption by the kidney, and by ...
  7. [7]
    Mechanism of Hormone Peptide Activation of a GPCR: Angiotensin ...
    The endocrine octa-peptide angiotensin II (AngII) activates AT 1 R signaling in our bodies which maintains physiological blood pressure, electrolyte balance, ...
  8. [8]
    Structural insights into GPCR signaling activated by peptide ligands
    Jul 8, 2025 · We examine representative GPCRs, such as the angiotensin II type 1 receptor ... GPCRs share a conserved seven-transmembrane (7TM) domain ...
  9. [9]
    Advances in the allostery of angiotensin II type 1 receptor
    Jun 17, 2023 · G protein-coupled receptors (GPCRs) are the largest family of membrane receptors with seven transmembrane (TM) α-helices. GPCR-tageting ...
  10. [10]
    AGTR1 - Type-1 angiotensin II receptor - Homo sapiens (Human)
    Receptor for angiotensin II, a vasoconstricting peptide, which acts as a key regulator of blood pressure and sodium retention by the kidney.
  11. [11]
    AGTR2 - Type-2 angiotensin II receptor - Homo sapiens (Human)
    Receptor for angiotensin II, a vasoconstricting peptide (PubMed:28379944, PubMed:29967536, PubMed:31899086, PubMed:8185599).
  12. [12]
    N-Linked Glycosylation Is Required for Optimal AT 1a Angiotensin ...
    These findings suggest that glycosylation enhances receptor stability, possibly by protecting nascent receptors from proteolytic degradation.
  13. [13]
    7F6G: Cryo-EM structure of human angiotensin receptor AT1R in ...
    Mar 29, 2023 · We present a cryo-electron microscopy (cryo-EM) structure of the human AT 1 R in complex with a balanced agonist, Sar 1 -AngII, and G q protein at 2.9 Å ...Missing: 2020s | Show results with:2020s
  14. [14]
  15. [15]
  16. [16]
  17. [17]
    K D values for conversion of angiotensin I and angiotensin II in the...
    In homogenates of human left ventricle, (125)I-[Sar(1),Ile(8)]angiotensin II bound with sub-nanomolar affinity, with a corresponding K(D) of 0.42+/-0.09 nM, a B ...
  18. [18]
    Angiotensin II type 1 receptor polymorphisms and susceptibility to ...
    The AGTR1 gene is located on chromosome 3q21–25 and is more than 55 Kb long. Four transcription initiation sites have been described. AGTR1 is composed of five ...Missing: rodents | Show results with:rodents
  19. [19]
    Type 1 angiotensin receptor pharmacology: Signaling beyond G ...
    The important cardiovascular actions of AngII, including regulation of arterial blood pressure and water-salt balance, are predominantly mediated by the AT1R in ...Type 1 Angiotensin Receptor... · 3.1. Gpcr Phosphorylation... · 3.1. 1. G Protein Receptor...
  20. [20]
    Angiotensin II type 1 receptor blockers: Class effects vs. Molecular ...
    The Kd values of AT1 receptor binding were determined by 125I-[Sar1, Ile8]Ang II-binding experiments under equilibrium conditions, and binding kinetics values ...
  21. [21]
    Structure of the Angiotensin Receptor Revealed by Serial ... - NIH
    Angiotensin II type 1 receptor (AT1R) is a G protein-coupled receptor that serves as a primary regulator for blood pressure maintenance.
  22. [22]
    Novel Functions for Angiotensin-Converting Enzymes Review
    The AT1 receptor is predominantly expressed in the kidneys, adrenal glands, vascular smooth muscle cells and the heart, and it is to this receptor that the.
  23. [23]
    Localization and function of angiotensin AT 1 receptors
    AT1A receptors are expressed predominantly in the vascular smooth muscle, liver, lung, and kidney, whereas the AT1B receptors occur mainly in the adrenal and ...
  24. [24]
    Brain-Selective Overexpression of Angiotensin (AT1) Receptors ...
    Several AT1-rich sites lack a blood-brain-barrier, eg, subfornical organ, organum vasculosum of the lamina terminalis, area postrema, and median preoptic ...
  25. [25]
    The Biology of Angiotensin II Receptors - ScienceDirect.com
    In the human a single AT1 receptor protein mediates virtually all the effects of angiotensin II, suggesting that tissue specificity of angiotensin II must be ...<|separator|>
  26. [26]
    The angiotensin II type 1 receptor and receptor-associated proteins
    Sep 1, 2001 · Cloning of the AT1 receptor. Numerous previous attempts to purify the AT1 receptor failed because of its instability and minute quantities ...
  27. [27]
    300034 - ANGIOTENSIN II RECEPTOR, TYPE 2; AGTR2 - OMIM
    They identified 2 missense mutations: G21V and I53F (300034.0006). Patients with the AGTR2 sequence variants had severe/profound mental retardation, epileptic ...300034 - ANGIOTENSIN II ...
  28. [28]
    Angiotensin II Type 2 Receptor: A Target for Protection Against ...
    Jun 15, 2021 · This review focuses on the most recent findings on the beneficial effects of AT 2 R by summarizing both gene knockout studies as well as pharmacological ...
  29. [29]
    Functional Reconstitution of the Angiotensin II Type 2 Receptor and ...
    The apparent Kd values for Ang II were ⬇0.45 and ⬇0.78 nmol/L, and the calculated Bmax was 627 and 280 fmol/mg protein for AT2 wild-type and R142A-AT2 receptors ...
  30. [30]
    Ligand-independent signals from angiotensin II type 2 receptor ... - NIH
    ... (AT2) receptor by angiotensin II (Ang II) is . ... The expression level of AT2 receptor in normal adult tissues is ∼10–20 fmol/mg protein.
  31. [31]
    AT 2 receptor - IUPHAR/BPS Guide to PHARMACOLOGY
    The IUPHAR/BPS Guide to Pharmacology. AT 2 receptor - Angiotensin receptors. Detailed annotation on the structure, function, physiology, pharmacology and ...
  32. [32]
    Angiotensin Receptors Heterodimerization and Trafficking
    Aug 3, 2020 · This review is focused on angiotensin receptors and how their biological function is influenced by trafficking and interaction with others receptors.
  33. [33]
    Hypertension Compendium - American Heart Association Journals
    Mar 13, 2015 · the AT4 receptor (please note that AT3 receptors do not exist, the 4 refers to Ang IV). ... The (pro)renin receptor/ATP6AP2 is essential for ...
  34. [34]
    LNPEP leucyl and cystinyl aminopeptidase [ (human)] - NCBI
    Sep 9, 2025 · Insulin-regulated aminopeptidase (IRAP)/AT4 receptors are involved in neither the regulation of RBF or CBF nor in the handling of renal sodium.
  35. [35]
    AT4 receptor is insulin-regulated membrane aminopeptidase
    Here, we explore the potential mechanisms by which Ang IV binding to IRAP leads to the facilitation of learning and memory.Missing: gene SLC3A2
  36. [36]
    Local expression of AP/AngIV/IRAP and effect of AngIV on glucose ...
    Aug 26, 2015 · 11.3). IRAP is expressed in various organs such as brain, adrenal gland, kidney, lung and heart. There are data ...
  37. [37]
    The angiotensin IV/AT4 receptor - PubMed
    In the kidney Ang IV increases renal cortical blood flow and decreases Na(+) transport in isolated renal proximal tubules. The AT(4) receptor has recently been ...
  38. [38]
    Angiotensin-(1–7) is an endogenous ligand for the G protein ... - PNAS
    AT1 and AT2 specific 125I-[Sar-1, Ile-8]Ang II binding was similar in both WT and Mas-deficient kidneys (t = 0.98 and 0.46, respectively, df = 10). Fig. 1.Missing: homology | Show results with:homology
  39. [39]
    Angiotensin II and Angiotensin Receptors 1 and 2—Multifunctional ...
    The classic angiotensin receptors also include the 7-transmembrane receptors but have only 8% and 19% sequential homology with Mas protein, respectively. The ...
  40. [40]
    Significance of angiotensin 1–7 coupling with MAS1 receptor and ...
    Evidence for a functional interaction of the angiotensin‐(1‐7) receptor Mas with AT1 and AT2 receptors in the mouse heart. Hypertension 46: 937–942. [DOI] ...Missing: homology | Show results with:homology
  41. [41]
    Angiotensin-(1-7) through receptor Mas mediates endothelial nitric ...
    Our findings demonstrate that Ang-(1-7), through Mas, stimulates eNOS activation and NO production via Akt-dependent pathways.
  42. [42]
    ANG II-independent prorenin/(pro)renin receptor signaling pathways ...
    The (pro)renin receptor (PRR, also called ATP6AP2) ... Pivotal role of the renin/prorenin receptor in angiotensin II production and cellular responses to renin.
  43. [43]
    AT1 receptor signaling pathways in the cardiovascular system - PMC
    The AT1 receptor is predominantly expressed in various tissues throughout the cardiovascular system including vascular smooth muscle, endothelium, heart and ...Missing: distribution liver
  44. [44]
  45. [45]
  46. [46]
  47. [47]
  48. [48]
    Angiotensin II Type 2 Receptor: A Target for Protection Against ...
    Apr 12, 2021 · This review focuses on the most recent findings on the beneficial effects of AT 2 R by summarizing both gene knockout studies as well as pharmacological ...
  49. [49]
  50. [50]
    AT2 RECEPTOR ACTIVITIES AND PATHOPHYSIOLOGICAL ...
    These receptors have similar affinity to Ang II, but share a nucleic acid sequence homology of only 34% (1–3). Although the AT1R activities are known for many ...At2 Receptor Activities And... · At2r Structure, Regulation... · At2r Agonists And Vascular...
  51. [51]
    Renin-angiotensin system as an emerging target to modulate adult ...
    Jul 1, 2025 · Activation of AT2R with the non-peptide agonist C21 increased the proliferation and generation of neuroblasts in the V-SVZ both in rats and mice ...
  52. [52]
    Blood pressure regulation by the angiotensin type 1 receptor in the ...
    Reduction of proximal tubule AT1a receptors led to lower BPs, whereas overexpression generally caused increased BPs. Summary: AT1a receptors in the proximal ...
  53. [53]
    Salt induces vascular AT1 receptor overexpression in vitro and in vivo
    High salt intake caused an increase of AT1 receptor mRNA and AT1 receptor density to approximately 160% compared with control levels. Northern analysis revealed ...
  54. [54]
    Association of angiotensin II type 1 receptor gene polymorphism ...
    These results suggest that the AGTR1 A1166C polymorphism is associated with essential hypertension and carotid atherosclerosis in a Chinese population.
  55. [55]
    Angiotensin II Type 1 Receptor rs5186 Gene Variant Predicts ...
    The gain-of-function rs5186 A1166C variant in angtiotensin receptor type 1 (AGTR1) gene has been linked to hypertension, cardiovascular disease and metabolic ...
  56. [56]
    Sustained AT 1 R stimulation induces upregulation of growth factors ...
    Dec 15, 2022 · Our data support the concept whereby sustained AT 1 R stimulation contributes to the development of myocardial fibrosis and hypertrophy.
  57. [57]
    Pathophysiology of Angiotensin II-Mediated Hypertension, Cardiac ...
    Dec 4, 2024 · Heart failure is a complex syndrome characterized by cardiac hypertrophy, fibrosis, and diastolic/systolic dysfunction.
  58. [58]
    Neuroprotective effect of angiotensin II type 2 receptor during ...
    These results indicate that AT2R activation suppresses immune and inflammatory responses, and protects against cerebral ischemia/reperfusion injury. Keywords: ...
  59. [59]
    Protection of the myocardium against ischemia/reperfusion injury by ...
    Protection of the myocardium against ischemia/reperfusion injury by angiotensin-(1-9) through an AT2R and Akt-dependent mechanism · Authors · Affiliations.
  60. [60]
    Angiotensin II upregulates RAGE expression on podocytes - PubMed
    Since intraglomerular ANG II levels are increased in diabetic nephropathy, this interaction may have pathophysiological consequences for podocyte injury and ...Missing: damage | Show results with:damage
  61. [61]
    The effects of angiotensin-II receptor blockers on podocyte damage ...
    Jan 26, 2011 · The aim of the study was to determine in a rat model of streptozotocin-induced diabetic nephropathy the expression of: WT-1 (for podocyte loss ...
  62. [62]
    Angiotensin AT2 Receptor is Anti-inflammatory and Reno-Protective ...
    Apr 15, 2021 · Our data suggest that the involvement of IL-10 in AT2R-mediated anti-inflammation and reno-protection against LPS is complex.
  63. [63]
    Proximal tubule angiotensin AT2 receptors mediate an ... - PubMed
    We hypothesized that AT2R activation is renoprotective by directly increasing the levels of anti-inflammatory cytokine IL-10 in the kidney via nitric oxide (NO) ...
  64. [64]
    Angiotensin II AT1 receptor blockade ameliorates brain inflammation
    Dec 8, 2010 · Our results demonstrate that systemic administration of the centrally acting angiotensin II AT(1) receptor blocker (ARB) candesartan to normotensive rats ...
  65. [65]
    Inhibition of angiotensin II receptor 1 limits tumor-associated ...
    Sep 22, 2009 · Together, these results show that blockage of AT1 receptor signaling may be a promising anti-tumor strategy, interfering with angiogenesis ...
  66. [66]
    Candesartan attenuates angiogenesis in hepatocellular carcinoma ...
    Angiotensin II type 1 receptor (AT1R) was reported to express in many types of tumors, promoting tumor growth and angiogenesis. We herein examined AT1R ...
  67. [67]
    The Gene Variants of Maternal/Fetal Renin-Angiotensin System in ...
    Jul 11, 2017 · Our findings demonstrate that fetal ACE I/D, ACE G2350A, AGT M235T, and AT1R A1166C polymorphisms may play significant roles in PE development among pregnant ...<|separator|>
  68. [68]
    COVID-19 lung disease shares driver AT2 cytopathic features with ...
    In the aftermath of Covid-19, some patients develop a fibrotic lung disease, i.e., post-COVID-19 lung disease (PCLD), for which we currently lack insights ...
  69. [69]
    Angiotensin II Receptor Blockers (ARB) - StatPearls - NCBI Bookshelf
    May 5, 2025 · ARBs are a class of medications that selectively inhibit the binding of angiotensin II to the angiotensin type 1 receptor.
  70. [70]
    [PDF] COZAAR® (losartan potassium) tablets - accessdata.fda.gov
    See full prescribing information for COZAAR. COZAAR® (losartan potassium) tablets, for oral use. Initial U.S. Approval: 1995. WARNING: FETAL TOXICITY. See full ...
  71. [71]
    Valsartan: Uses, Interactions, Mechanism of Action | DrugBank Online
    Valsartan is indicated for the treatment of hypertension to reduce the risk of fatal and nonfatal cardiovascular events, primarily strokes and myocardial ...Identification · Pharmacology · Interactions · Products
  72. [72]
    Safety, Efficacy and Pharmacokinetics of C21 in Subjects With IPF
    A phase of research to describe clinical trials that gather more information about a drug's safety and effectiveness by studying different populations and ...Missing: AT2 | Show results with:AT2
  73. [73]
    [PDF] Vicore Pharma
    Aug 20, 2025 · The company's lead program, buloxibutid (C21), is a first-in-class oral small molecule angiotensin II type 2 (AT2) receptor agonist, which.
  74. [74]
    [PDF] 4199496 This label may not be the latest approved by FDA. For ...
    GIAPREZA increases blood pressure in adults with septic or other distributive shock. 2. DOSAGE AND ADMINISTRATION. 2.1. Preparation. Parenteral drug products ...
  75. [75]
    Nonpeptide AVE 0991 is an angiotensin-(1-7) receptor Mas agonist ...
    It has been described recently that the nonpeptide AVE 0991 (AVE) mimics the effects of angiotensin-(1-7) [Ang-(1-7)] in bovine endothelial cells.
  76. [76]
    Balanced affinity AT1/AT2 receptor nonpeptide binding ... - PubMed
    These data suggest that AT1-selective and dual receptor nonpeptides share overlapping but distinct binding pockets on the AT1 receptor. These findings may lead ...
  77. [77]
    Angiotensin II dose–effect curves and Schild regression plots ... - NIH
    The 'Schild regression' method is based on the principle of assessing the rightward shift of agonist dose–effect curves in the presence of different doses/ ...
  78. [78]
    Biased Agonists of the Type 1 Angiotensin II Receptor Promote ...
    Sep 20, 2025 · Here, we examine how various biased agonists influence the recruitment of β-arrestins 1 and 2 induced by the angiotensin II type 1 receptor at ...
  79. [79]
    Sacubitril-Valsartan - StatPearls - NCBI Bookshelf
    Sacubitril-valsartan is the first agent approved in a new class of drugs called angiotensin receptor neprilysin inhibitor (ARNI).