Fact-checked by Grok 2 weeks ago

Angiotensin-converting enzyme 2

Angiotensin-converting 2 (ACE2) is a zinc-dependent carboxypeptidase that functions as a key regulator in the renin- (RAS), primarily by hydrolyzing angiotensin II (Ang II) to angiotensin-(1-7) (Ang-(1-7)), thereby counteracting the vasoconstrictive and pro-inflammatory effects of (ACE). Discovered in through screening of human expressed sequence tags and a cDNA library, ACE2 is a type I transmembrane consisting of an extracellular catalytic domain homologous to ACE, a transmembrane , and a short intracellular C-terminal tail fused to a collectrin-like domain that facilitates its localization to the plasma membrane. Beyond its role in the RAS, ACE2 also cleaves other peptides such as apelin-13, [des-Arg⁹]-bradykinin, and amyloid-β, contributing to cardioprotection, anti-fibrotic effects, and potential . ACE2 is widely expressed on the surface of endothelial and epithelial cells, with high levels in the lungs, , heart, kidneys, and vascular , where it promotes intestinal absorption via interaction with neutral transporters and helps maintain . Its expression and activity are tightly regulated at multiple levels, including transcriptional control by factors like hepatocyte nuclear factor 1α (HNF1α), posttranscriptional modulation by microRNAs such as miR-421, and posttranslational shedding by ADAM17 sheddase, which releases a soluble form (sACE2) into circulation. Dysregulation of ACE2 has been implicated in various pathologies; for instance, reduced ACE2 activity is associated with , , , and acute lung injury, while its upregulation may offer therapeutic benefits in these conditions. Notably, ACE2 serves as the functional receptor for severe acute respiratory syndrome coronaviruses, binding the of SARS-CoV and to facilitate viral entry into host cells, a discovery that has heightened its relevance in infectious research and development. This interaction underscores ACE2's dual role in both physiological protection against RAS overactivation and vulnerability to viral pathogens, influencing outcomes in where viral binding may downregulate membrane-bound ACE2, exacerbating and organ damage. Ongoing research explores ACE2 modulators as potential therapies for cardiovascular diseases and as strategies to mitigate viral entry without compromising its protective functions.

Molecular Structure and Genetics

Protein Structure

Angiotensin-converting enzyme 2 (ACE2) is a single-pass type I transmembrane comprising 805 in humans. The protein features an N-terminal (residues 1–18) that directs its translocation to the , followed by a large extracellular domain (residues 19–740), a single transmembrane (residues 741–761), and a short cytoplasmic tail (residues 762–805). The extracellular domain encompasses the functional core of ACE2, divided into a zinc metallopeptidase domain (residues 19–615) responsible for catalytic activity and a C-terminal collectrin-like domain (residues 616–740) that mediates homodimerization. Key structural elements include the zinc-binding motif (HEMGH) within the active site of the peptidase domain, which coordinates a catalytic zinc ion essential for peptidase function, and seven N-linked glycosylation sites (at Asn53, Asn90, Asn103, Asn322, Asn432, Asn546, and Asn690) that contribute to protein stability and trafficking. Crystal structures of the ACE2 ectodomain, such as the apo form (PDB ID: 1R42), reveal a clam-shell-like with two lobes forming a deep substrate-binding groove; the peptidase domain consists of a central eight-stranded β-sheet flanked by α-helices, including 20 helical segments overall. Additional structures, like the complex with SARS-CoV-2 receptor-binding domain and neutral amino acid transporter B(0)AT1 (PDB ID: 6M17), highlight dimerization interfaces and conformational flexibility in the collectrin-like domain. In comparison to angiotensin-converting enzyme 1 (ACE1), ACE2 possesses a single catalytic domain, whereas somatic ACE1 has two homologous domains (N- and C-terminal); the peptidase domains of ACE2 and ACE1 share approximately 42% sequence identity.

Gene and Expression Regulation

The ACE2 gene is located on the X chromosome at position Xp22.2 in humans, spanning approximately 89 kb of genomic DNA and consisting of 22 exons interrupted by 21 introns. This structure encodes the full-length ACE2 protein, a monocarboxypeptidase involved in the renin-angiotensin system. The gene's X-linked position contributes to its inheritance patterns and potential for sex-specific expression differences. ACE2 exhibits strong evolutionary across mammalian at both the and protein sequence levels, reflecting its essential physiological roles in cardiovascular and renal . Key polymorphisms, such as rs2285666 located in intron 3, influence expression levels; the A can increase ACE2 mRNA expression by up to 50% through effects on or enhancer activity. Transcriptional regulation of ACE2 is mediated by promoters and enhancers responsive to various stimuli. upregulates ACE2 expression via (ESR1), promoting protective effects in tissues like the airway and myocardium. In contrast, cytokines such as interferon-gamma (IFN-γ) downregulate ACE2 in certain inflammatory contexts, potentially exacerbating viral susceptibility. Additionally, -inducible factor 1-alpha (HIF-1α) modulates ACE2 during hypoxic conditions; while acute may upregulate it, chronic exposure often leads to downregulation via HIF-1α binding to -responsive elements. Post-transcriptional control further fine-tunes ACE2 levels. MicroRNAs, including miR-421, bind the 3'- of ACE2 mRNA to suppress its , reducing protein abundance in conditions like or . generates truncated variants, such as deltaACE2 (dACE2), which lacks the and is induced by interferons or viral infection, potentially altering receptor function without full enzymatic activity. Due to its X-linked location, ACE2 expression shows sex-based differences, with higher levels often observed in males across tissues like the lungs and heart, partly because the gene may escape X-inactivation in females. This dimorphism contributes to increased male susceptibility to diseases involving ACE2 dysregulation, such as severe COVID-19.

Tissue Distribution and Localization

Primary Locations in the Human Body

ACE2 exhibits high expression in several key organs and tissues in the human body, particularly those involved in cardiovascular and respiratory function. Notable sites include the lungs, where it is predominantly found in alveolar epithelial cells; the small intestine, with strong presence in enterocytes; the kidneys, specifically in proximal tubule cells; the heart, in cardiomyocytes and endothelial cells; and broadly across vascular endothelium. These patterns are supported by both RNA sequencing and proteomic analyses, highlighting ACE2's role in epithelial and endothelial barriers. Quantitative data from the GTEx database, derived from RNA-seq across multiple donors, further delineates these expression levels using median transcripts per million (TPM). The shows the highest median expression at approximately 55 TPM (varying by subregion: ~15 TPM, ~13 TPM), followed by the testis at 37 TPM, (primarily ) at 7 TPM, heart (atrial and ventricular) at around 7 TPM, and lungs at approximately 2 TPM, establishing a hierarchy where gastrointestinal and reproductive tissues lead, followed by renal and cardiac, with pulmonary expression low but present in specific cells. Moderate expression is observed in (~9 TPM), while liver, , and show low expression (<1 TPM). These distributions are corroborated by proteomic studies confirming protein-level consistency in enterocytes, renal tubules, cardiomyocytes, and endothelial cells. Developmentally, ACE2 expression is generally low in fetal tissues, with significant levels restricted primarily to the intestine and kidney, and minimal presence in the fetal lung or placenta. Postnatally, expression increases in response to physiological demands, such as maturation of the , reaching adult levels in organs like the lung and heart. This ontogenetic shift underscores adaptive regulation tied to organ development. Species differences in ACE2 distribution are particularly evident in the lungs, where human expression is higher than in rodents, contributing to the limited utility of standard rodent models for studying respiratory pathologies involving ACE2. In mice, pulmonary ACE2 mRNA levels are notably lower (often <1 TPM), necessitating transgenic humanized models for accurate replication of human-like infection dynamics.

Cellular and Subcellular Localization

Angiotensin-converting enzyme 2 (ACE2) is predominantly expressed as a membrane-bound glycoprotein anchored to the plasma membrane via a single transmembrane domain, with its extracellular ectodomain facing the extracellular space. In polarized epithelial cells, such as those in the lung alveoli and intestinal epithelium, ACE2 is primarily localized to the apical membrane, where it co-localizes with apical markers like glycoprotein gp135 and is associated with tight junction proteins, facilitating its positioning at the brush border. This apical distribution positions ACE2 optimally for interaction with luminal substrates and pathogens in these barrier tissues. A soluble form of ACE2 is generated through ectodomain shedding mediated by the disintegrin and metalloproteinase 17 () protease, which cleaves the protein near the transmembrane domain, releasing the catalytically active ectodomain into the extracellular space and circulation as circulating soluble ACE2. This shedding process is regulated and can be enhanced by inflammatory signals or ligand binding, contributing to variable levels of soluble ACE2 in plasma. Subcellularly, newly synthesized ACE2 undergoes post-translational modifications, including N-linked glycosylation in the endoplasmic reticulum and further processing in the Golgi apparatus, before being trafficked via vesicles to the plasma membrane. Once at the surface, ACE2 can undergo endocytic recycling, allowing it to be internalized and returned to the membrane, maintaining its surface expression. Immunohistochemical studies have confirmed ACE2's membrane localization in epithelial cells, showing strong apical staining and co-localization with tight junctions in respiratory and intestinal epithelia, while in neuronal cells, it exhibits a more intracellular, cytoplasmic distribution, often perinuclear. The localization of ACE2 is dynamic; upon binding to ligands such as the , it undergoes clathrin-mediated endocytosis, leading to internalization and potential lysosomal degradation or altered shedding rates, which can reduce surface levels. This trafficking regulation influences ACE2's availability for enzymatic function and receptor roles.

Biochemical Function

Enzymatic Activity and Mechanism

Angiotensin-converting enzyme 2 (ACE2) is classified as a zinc-dependent carboxypeptidase (EC 3.4.17.23) that catalyzes the hydrolysis of peptide bonds at the C-terminal end of substrates. This enzymatic activity is central to its role in processing bioactive peptides within the renin-angiotensin system. The active site of ACE2 contains a zinc ion (Zn²⁺) coordinated by the imidazole rings of histidine residues His383 and His387, as well as the carboxylate group of Glu402, forming a classic HEXXH zinc-binding motif typical of gluzincin metallopeptidases. In the catalytic mechanism, the coordinated Zn²⁺ polarizes the carbonyl group of the scissile peptide bond, facilitating nucleophilic attack by a water molecule; Glu402 acts as a proton shuttle, abstracting a proton from the water to generate the hydroxide nucleophile and later donating it to the departing amine group. This general acid-base catalysis ensures efficient C-terminal residue removal, with the process being strictly exopeptidase in nature. Kinetic parameters for ACE2 activity toward angiotensin II (Ang II) include a Michaelis constant (K_m) of approximately 2 μM and a turnover number (k_cat) of about 210 min⁻¹ (3.5 s⁻¹), yielding a catalytic efficiency (k_cat/K_m) of 1.8 × 10⁶ M⁻¹ s⁻¹. The enzyme exhibits an optimal pH range of 6.5–7.5, with activity enhanced by monovalent anions such as chloride and sharply declining outside this range. ACE2 is inhibited by metal chelators like EDTA, which sequesters the essential Zn²⁺ cofactor, confirming its metalloprotease nature. ACE2 forms homodimers through its C-terminal collectrin-like domain, which promotes proper orientation of the peptidase domains and increases substrate affinity compared to the monomeric form. Beyond its catalytic function, ACE2 plays a non-enzymatic structural role in facilitating neutral amino acid transport; its collectrin-like domain interacts with the transporter B⁰AT1 (SLC6A19) at the apical membrane of intestinal epithelial cells, stabilizing surface expression and enhancing transport efficiency independently of peptidase activity, as demonstrated by catalytically inactive mutants. This interaction mirrors the function of collectrin (TMEM27) in the kidney but is tissue-specific to the gut for ACE2.

Substrates, Products, and Interactions

Angiotensin-converting enzyme 2 (ACE2) primarily catalyzes the hydrolysis of angiotensin II (Ang II) to angiotensin-(1-7) [Ang-(1-7)] by removing the C-terminal phenylalanine residue, thereby counterbalancing the effects of the classical renin-angiotensin system pathway. This conversion is a key biochemical transformation mediated by ACE2's carboxypeptidase activity. In addition to Ang II, ACE2 processes angiotensin I (Ang I) into Ang-(1-9) through the removal of a single C-terminal leucine residue, although with lower catalytic efficiency compared to the Ang II reaction. Other notable substrates include apelin-13, which is cleaved to apelin-12; kinetensin; des-Arg⁹-bradykinin; and amyloid-β peptides, all of which undergo C-terminal hydrolysis. ACE2 demonstrates a substrate preference for peptides featuring hydrophobic residues, such as proline, leucine, or phenylalanine, at the penultimate C-terminal position, facilitating selective cleavage. The products of ACE2-mediated hydrolysis play significant roles in physiological signaling. Ang-(1-7), the principal product from Ang II, functions as an endogenous ligand for the Mas receptor (MasR), activating downstream pathways that promote vasodilation, anti-inflammatory responses, and cardioprotection. In contrast, Ang-(1-9) acts primarily as an intermediate peptide, which can be further degraded by neutral endopeptidase or ACE to yield Ang-(1-7), thereby contributing indirectly to MasR activation. These transformations highlight ACE2's role in generating bioactive heptapeptides that oppose angiotensin II-mediated effects. Beyond enzymatic substrates, ACE2 engages in key protein-protein interactions that influence its localization and function. Allosteric modulation of ACE2 activity occurs through binding of small molecules, such as the selective inhibitor MLN-4760, which targets a site distinct from the catalytic zinc-binding domain to potently suppress enzymatic function.

Physiological Roles

Role in the Renin-Angiotensin-Aldosterone System

Angiotensin-converting enzyme 2 (ACE2) serves as a critical component of the renin-angiotensin-aldosterone system (RAS), acting as a carboxypeptidase that hydrolyzes angiotensin II (Ang II) to angiotensin-(1-7) [Ang-(1-7)], thereby counteracting the vasoconstrictive and pro-inflammatory effects of Ang II mediated through the angiotensin type 1 receptor (AT1R). This enzymatic activity positions ACE2 as a negative regulator of the classical RAS pathway, which involves angiotensinogen conversion to Ang I by renin, followed by ACE-mediated formation of Ang II. By degrading Ang II with approximately 400-fold higher catalytic efficiency than its action on Ang I to produce Ang-(1-9), ACE2 helps maintain RAS homeostasis, particularly in cardiovascular and renal tissues. In the broader RAS pathway, ACE2 integrates with the ACE/Ang II axis to prevent excessive Ang II accumulation; its deficiency results in elevated Ang II levels, leading to hypertension and cardiac hypertrophy, as demonstrated in ACE2 knockout mice that exhibit exacerbated pressure overload responses and impaired myocardial contractility. These models underscore ACE2's role in balancing the system, where loss of function amplifies the pathological effects of the classical pathway, such as increased blood pressure and ventricular remodeling. The ACE2/Ang-(1-7)/Mas receptor (MasR) axis forms a protective arm of the RAS, with Ang-(1-7) binding to MasR to promote vasodilation through nitric oxide release, while exerting anti-fibrotic and anti-inflammatory effects that oppose Ang II-induced tissue damage. This pathway mitigates fibrosis by inhibiting transforming growth factor-β signaling and reduces inflammation via suppression of pro-inflammatory cytokines, contributing to cardioprotection in models of heart failure. Feedback mechanisms further regulate ACE2 within the RAS; Ang II upregulates ACE2 expression in cardiac fibroblasts through AT1R and ERK-MAPK pathways, potentially serving as a compensatory response to elevated Ang II levels. Additionally, aldosterone influences ACE2 activity via mineralocorticoid receptors, where blockade of these receptors increases ACE2 expression and activity, suggesting aldosterone normally suppresses ACE2 to promote RAS imbalance.

Roles in Cardiovascular, Renal, and Other Systems

In the cardiovascular system, ACE2 contributes to endothelial protection by counteracting angiotensin II-mediated vasoconstriction and promoting vasodilation through the generation of angiotensin-(1-7), which enhances nitric oxide production in endothelial cells. This enzymatic activity also reduces oxidative stress by limiting reactive oxygen species formation in vascular tissues, thereby preventing endothelial dysfunction and supporting vascular integrity. Furthermore, ACE2 attenuates cardiac hypertrophy by mitigating angiotensin II-induced signaling pathways that promote cardiomyocyte growth, maintaining balanced cardiac remodeling under physiological conditions. In relation to atherosclerosis prevention, ACE2 overexpression has been shown to inhibit inflammatory responses in endothelial cells, reducing monocyte adhesion and early plaque formation while preserving arterial wall homeostasis. Within the renal system, ACE2 regulates glomerular filtration by modulating local angiotensin II levels in podocytes and tubular cells, ensuring appropriate sodium reabsorption and filtration barrier function. Its expression in podocytes specifically supports cellular integrity and reduces oxidative stress, which helps maintain the structural stability of the glomerular filtration barrier essential for efficient ultrafiltration. Through the ACE2/angiotensin-(1-7)/Mas receptor axis, ACE2 further promotes anti-inflammatory effects in renal tissues, counteracting potential disruptions to podocyte architecture and preserving overall renal hemodynamics. In the pulmonary system, ACE2 maintains alveolar fluid balance by facilitating electrolyte and liquid homeostasis in alveolar epithelial cells, preventing excessive fluid accumulation through balanced regulation of sodium transport and vascular permeability. It also exerts anti-inflammatory actions in the airways by degrading pro-inflammatory and producing anti-fibrotic , which supports epithelial cell survival and reduces cytokine-mediated inflammation in lung tissues. These functions collectively ensure proper alveolar gas exchange and airway patency under normal physiological states. Beyond these systems, ACE2 plays roles in other organs, including the gut, where it acts as a chaperone for the neutral amino acid transporter , facilitating the absorption of dietary amino acids such as tryptophan and maintaining intestinal homeostasis. In the brain, ACE2 provides neuroprotection by activating the angiotensin-(1-7)/Mas axis, which inhibits neuronal apoptosis and oxidative damage, while its expression in cerebrovascular endothelial cells helps preserve blood-brain barrier integrity against permeability challenges. Recent insights from 2025 highlight ACE2's involvement in the pancreas, where low-level expression in β-cells enhances glucose-stimulated insulin secretion and supports overall glucose homeostasis by modulating local renin-angiotensin system balance. Circulating soluble ACE2, derived from ectodomain shedding, enables multi-organ interplay by systemically degrading angiotensin II and generating angiotensin-(1-7), thereby modulating inflammation across tissues and contributing to coordinated physiological responses. This soluble form briefly references its contribution to renin-angiotensin-aldosterone system balance without altering core pathway mechanics.

Pathological and Clinical Significance

Involvement in Viral Infections

Angiotensin-converting enzyme 2 (ACE2) serves as a critical receptor for the entry of several into host cells, primarily through binding of the viral spike protein's S1 subunit to the peptidase domain of ACE2. This interaction was first established for (SARS-CoV), where the spike protein binds ACE2 with high affinity, facilitating viral attachment and subsequent membrane fusion. Key residues in ACE2, such as lysine 353 (K353) and tyrosine 385 (Y385), form essential contacts within the receptor-binding motif of the SARS-CoV spike, enabling species-specific tropism and efficient entry into human airway epithelial cells. Similarly, , the causative agent of , exploits the same receptor, with its receptor-binding domain (RBD) engaging ACE2 via conserved interfaces involving K353, which forms a salt bridge with glutamic acid 484 in the viral spike, underscoring the structural conservation that allows cross-reactivity between these . The entry mechanism of SARS-CoV-2 into cells involves ACE2-mediated attachment followed by proteolytic priming of the spike protein, which can occur via two primary pathways. At the plasma membrane, transmembrane serine protease 2 (TMPRSS2) cleaves the spike at the S1/S2 boundary, promoting direct fusion; alternatively, if TMPRSS2 activity is low, the virus undergoes clathrin-mediated endocytosis, where endosomal acidification activates cathepsin L to perform the cleavage, releasing the S2 subunit for membrane fusion. This dual-route dependency enhances SARS-CoV-2's infectivity across diverse cell types expressing ACE2, such as alveolar epithelial cells and endothelial cells. Beyond SARS coronaviruses, human coronavirus NL63 (HCoV-NL63), an alphacoronavirus causing mild respiratory illness, also utilizes ACE2 as its receptor, with its spike binding a distinct but overlapping region of the peptidase domain, highlighting ACE2's broader role in coronavirus pathogenesis. Viral engagement of ACE2 triggers downstream dysregulation, notably the shedding of ACE2 from the cell surface via ADAM17-mediated cleavage, which generates soluble ACE2 (sACE2) but depletes membrane-bound ACE2. This "ACE2 shedding paradox" reduces production of the protective peptide angiotensin-(1-7) [Ang-(1-7)], tipping the renin-angiotensin system toward proinflammatory angiotensin II signaling and exacerbating lung injury and cytokine storm in severe cases. Post-2020 research on SARS-CoV-2 variants reveals altered ACE2 interactions; for instance, the Omicron subvariants (BA.1 and BA.2) exhibit higher binding affinity to ACE2 compared to the ancestral strain, driven by mutations like Q493R and N501Y that stabilize the RBD-ACE2 interface, contributing to enhanced transmissibility despite partial immune evasion. More recent variants, such as JN.1 as of 2025, continue to evolve with mutations maintaining or enhancing ACE2 binding, influencing ongoing COVID-19 dynamics and long-term sequelae. In long COVID, persistent ACE2 dysregulation, including sustained downregulation and vascular shedding, is linked to ongoing endothelial dysfunction and symptoms like fatigue and microvascular thrombosis, as evidenced by elevated circulating sACE2 levels in affected individuals months post-infection.

Implications in Cardiovascular, Metabolic, and Other Diseases

Angiotensin-converting enzyme 2 (ACE2) deficiency exacerbates cardiovascular pathologies by disrupting the balance of the renin-angiotensin system (RAS), leading to elevated angiotensin II (Ang II) levels that promote inflammation, fibrosis, and vascular contraction. In hypertension models, ACE2 knockout mice exhibit accelerated blood pressure elevation and increased cardiac hypertrophy, while ACE2 overexpression attenuates these effects by enhancing Ang-(1-7) production. Similarly, in heart failure, ACE2 loss in murine models results in systolic dysfunction, reduced ejection fraction, and worsened fibrosis, whereas recombinant human ACE2 (rhACE2) administration improves cardiac function by mitigating oxidative stress. Polymorphisms in the ACE2 gene, such as those in the promoter region, have been associated with increased risk of myocardial infarction in population studies, particularly in hypertensive individuals, highlighting genetic contributions to disease susceptibility. In metabolic disorders, reduced ACE2 expression in type 2 diabetes mellitus (T2DM) contributes to insulin resistance by impairing the ACE2/Ang-(1-7)/Mas axis, which normally enhances insulin signaling and glucose uptake in skeletal muscle and adipocytes. Studies in db/db mice demonstrate that ACE2 deficiency decreases β-cell mass and proliferation, leading to impaired glucose-stimulated insulin secretion and elevated fasting glucose, while Ang-(1-7) infusion restores β-cell function and improves glycemic control. Recent 2024-2025 reviews emphasize ACE2's role in glucose homeostasis, noting that activation of this axis reduces hepatic gluconeogenesis and promotes thermogenesis in brown adipose tissue via Akt/FOXO1 and PKA pathways in high-fat diet models. ACE2 dysregulation promotes renal injury, with loss accelerating acute kidney injury (AKI) in sepsis through unchecked Ang II-mediated vasoconstriction and inflammation. In ischemia-reperfusion AKI models, ACE2 knockout mice display exacerbated tubular damage, increased immune cell infiltration, and higher creatinine levels compared to wild-type controls, underscoring ACE2's protective role in maintaining glomerular filtration. For chronic kidney disease (CKD), diminished ACE2 expression drives progression via enhanced fibrosis, as evidenced by increased extracellular matrix deposition and TGF-β signaling in ACE2-deficient diabetic nephropathy models. The ACE2/Ang-(1-7)/Mas axis counteracts this by suppressing oxidative stress and epithelial-mesenchymal transition in proximal tubules. Beyond these systems, ACE2 influences other diseases through anti-inflammatory and anti-proliferative mechanisms. In Alzheimer's disease, reduced brain ACE2 levels correlate with neuroinflammation and cognitive decline, as ACE2 activation via small-molecule agonists like diminazene aceturate decreases amyloid-β accumulation and tau pathology in mouse models by modulating the RAS axis. In cancer, ACE2 exerts tumor-suppressive effects by inhibiting angiogenesis; for instance, ACE2 overexpression in renal cell carcinoma cells reduces vascular endothelial growth factor (VEGF) expression and tumor growth in xenografts, with higher ACE2 correlating to improved patient survival in multiple tumor types. Post-COVID-19 sequelae involve long-term endothelial dysfunction linked to ACE2 shedding and downregulation, leading to persistent vascular inflammation and increased cardiovascular risk, as observed in cohort studies showing elevated Ang II and reduced nitric oxide bioavailability up to two years post-infection. Plasma soluble ACE2 levels serve as an emerging biomarker for disease outcomes, with elevated concentrations predicting adverse events in heart failure patients, including hospitalization and mortality, independent of traditional markers like BNP. In CKD, higher soluble ACE2 correlates with faster eGFR decline and fibrosis progression in 2025 longitudinal data, reflecting ongoing RAS imbalance.

Therapeutic and Research Developments

Recombinant Human ACE2

Recombinant human ACE2 (rhACE2), also known as human recombinant soluble ACE2 (hrsACE2), was first developed in 2007 as a soluble ectodomain construct comprising residues 1-740 of the native protein to facilitate its secretion and enzymatic function outside the cell membrane. This form was produced by transient transfection of HEK293 cells using an expression construct that fused a signal peptide to the extracellular domain, allowing for purification from cell culture supernatant via affinity chromatography. The resulting protein mimics the catalytic domain of membrane-bound ACE2 while being amenable to systemic administration. The recombinant protein retains the core enzymatic activity of native ACE2, hydrolyzing angiotensin II (Ang II) to angiotensin-(1-7) with a Michaelis constant (Km) of approximately 5 μM, comparable to that reported for the endogenous enzyme. Its plasma half-life is about 10 hours following intravenous (IV) infusion, enabling transient elevation of ACE2 activity to modulate the renin-angiotensin system (RAS). Administered IV, rhACE2 has demonstrated good tolerability in preclinical models and early human studies, with rapid distribution to tissues expressing high ACE2 levels, such as the lungs and kidneys. Early clinical evaluation occurred in a phase I/II trial for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), initiated around 2012 and reporting results in 2017, where IV rhACE2 (GSK2586881) at doses up to 0.8 mg/kg was safe and well-tolerated over 3 days. The trial observed a dose-dependent reduction in circulating Ang II levels by up to 35% without significant adverse effects, supporting its potential to restore RAS balance in inflammatory lung conditions. No serious immunogenicity was noted in this short-term study. During the COVID-19 pandemic, hrsACE2 (APN01) advanced to phase II trials starting in 2020, including a randomized, placebo-controlled study in moderate-to-severe cases completed by late 2020 and an extension into 2022 evaluating nebulized delivery. These trials confirmed safety and tolerability at doses of 0.4 mg/kg IV twice daily for 7 days, with evidence of spike protein neutralization in vitro and ex vivo, reducing viral entry into cells by competing for receptor binding. Additionally, APN01 restored RAS equilibrium by lowering Ang II and increasing angiotensin-(1-7), correlating with improved oxygenation in some patients, though broader efficacy endpoints like mortality reduction were not conclusively met. Post-2022 analyses highlighted persistent challenges, including the protein's short half-life necessitating frequent dosing and potential immunogenicity from repeated administration, which could limit long-term use despite no acute antibody responses in initial cohorts.

Modulators, Inhibitors, and Emerging Therapies

Angiotensin-converting enzyme 2 (ACE2) inhibitors have been primarily developed for research purposes to modulate the (RAS) and investigate ACE2's role in various pathologies. MLN-4760 is a highly selective small-molecule inhibitor that binds to the active site of ACE2 with high affinity (IC50 ≈ 0.44 nM), effectively blocking its enzymatic activity toward substrates like II. This compound has been widely used in preclinical studies to dissect ACE2-specific effects, such as in models of pulmonary injury where it attenuates lipopolysaccharide-induced by inhibiting ACE2-mediated conversion of II to angiotensin-(1-7). In contrast, , a classical , exhibits weak inhibitory activity against ACE2 (IC50 > 10 μM) and is not selective, but it has been employed in comparative studies to differentiate ACE from ACE2 contributions in glomerular injury and acute lung . These inhibitors highlight the potential for targeted RAS modulation, though clinical translation remains limited due to off-target effects and lack of disease-specific indications. Direct activators of ACE2 enzymatic activity are not currently available, but indirect modulation through upregulation of ACE2 expression has been achieved via (PPARγ) agonists. Pioglitazone, a thiazolidinedione-class drug, increases ACE2 protein levels in tissues such as liver, adipose, and , thereby enhancing the protective Ang-(1-7)/Mas receptor axis in models of non-alcoholic and . This upregulation occurs through PPARγ-mediated transcriptional activation, offering a strategy to counterbalance pathological overactivation without directly altering enzymatic kinetics. Emerging therapies targeting ACE2 include approaches for overexpression in , where preclinical studies using (AAV)-mediated delivery of the ACE2 gene have demonstrated reduced cardiac , , and improved in rodent models of pressure overload-induced failure. Additionally, neutralizing monoclonal antibodies that disrupt the ACE2-spike protein interaction have been developed post-COVID-19, with broad-spectrum candidates like CV3-1 binding to conserved epitopes on the SARS-CoV-2 spike receptor-binding domain to prevent entry while preserving ACE2 function. These antibodies, elicited or refined from vaccine-induced responses, show promise against variants and other coronaviruses in preclinical assays. Recent preclinical studies as of 2024 have explored engineered variants of recombinant human ACE2 with enhanced stability and antiviral activity, demonstrating reduced infection in cellular and animal models. As of 2025, the clinical pipeline for ACE2-targeted interventions includes soluble ACE2 infusions for (ARDS), building on phase II trials of recombinant human ACE2 (e.g., APN01) that demonstrated safety and preliminary efficacy in reducing ventilator days and in COVID-19-associated ARDS patients. In preclinical models, diminazene aceturate serves as an ACE2 activator, lowering and mitigating vascular remodeling by enhancing Ang-(1-7) production, with ongoing studies exploring its translation to clinical use. Key challenges in ACE2 modulator development involve balancing cardioprotective benefits against the risk of facilitating entry, as enhanced ACE2 expression or activity could increase to and related pathogens in at-risk populations. Furthermore, ACE2's X-linked genetic location necessitates consideration of sex-specific dosing, as females exhibit higher baseline expression due to incomplete X-chromosome inactivation, potentially requiring adjusted therapeutic levels to avoid over- or under-modulation in sex-biased contexts.

References

  1. [1]
    ACE2 Cell Biology, Regulation, and Physiological Functions - PMC
    Angiotensin-converting enzyme 2 (ACE2), discovered as a homologue of ACE, acts as its physiological counterbalance providing homeostatic regulation of ...
  2. [2]
    Understanding SARS-CoV-2 interaction with the ACE2 receptor and ...
    ACE2 is postulated to serve as a major entry receptor for SARS-CoV-2 in human cells, as it does for SARS-CoV. Many infected individuals develop COVID-19 with ...
  3. [3]
    ACE2 - Angiotensin-converting enzyme 2 - Homo sapiens (Human)
    Converts angiotensin I to angiotensin 1-9, a nine-amino acid peptide with anti-hypertrophic effects in cardiomyocytes, and angiotensin II to angiotensin 1-7, ...
  4. [4]
    Structural basis for the recognition of SARS-CoV-2 by full-length ...
    The ACE2-B0AT1 complex is assembled as a dimer of heterodimers, with the collectrin-like domain of ACE2 mediating homodimerization. The RBD is recognized by the ...
  5. [5]
    ACE2 and ACE: structure-based insights into mechanism, regulation ...
    Both ACE and ACE2 are members of the zinc metalloprotease family and contain the HEXXH + E (downstream glutamate residue) zinc binding motif. As described above ...
  6. [6]
    Dual nature of human ACE2 glycosylation in binding to SARS-CoV ...
    Apr 26, 2021 · The extracellular domain of the ACE2 receptor has seven N-glycosylation sites (N53, N90, N103, N322, N432, N546, and N690) and several O- ...
  7. [7]
    1R42: Native Human Angiotensin Converting Enzyme ... - RCSB PDB
    Feb 3, 2004 · The angiotensin-converting enzyme (ACE)-related carboxypeptidase, ACE2, is a type I integral membrane protein of 805 amino acids that contains one HEXXH + E ...Missing: 6M17 | Show results with:6M17
  8. [8]
    Ectodomain shedding of angiotensin converting enzyme 2 in human ...
    The human ACE2 crystal structure revealed it contains 20 α-helical segments (58). ... A: surface representation of ACE2 protein (pdb id 1R42) with the 2 catalytic ...<|separator|>
  9. [9]
    6M17: The 2019-nCoV RBD/ACE2-B0AT1 complex - RCSB PDB
    Mar 11, 2020 · Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. · Total Structure Weight: 394.14 kDa · Atom Count: 25,344 · Modeled ...Missing: 1R42 | Show results with:1R42
  10. [10]
    A Novel Angiotensin-Converting Enzyme–Related ...
    The metalloprotease catalytic domains of ACE2 and ACE are 42% identical, and comparison of the genomic structures indicates that the two genes arose through ...Missing: identity | Show results with:identity
  11. [11]
    Gene ResultACE2 angiotensin converting enzyme 2 [ (human)] - NCBI
    Sep 9, 2025 · ACE2 gene polymorphisms are associated with elevated pulmonary artery pressure in congenital heart diseases. Association between angiotensin ...Missing: Xp22. 39.6 kb exons
  12. [12]
    Analysis of ACE2 Gene-Encoded Proteins Across Mammalian Species
    The major finding of our predictive analysis suggested ACE2 gene-encoded proteins to be highly homologous across mammals.
  13. [13]
    Genetic Association of ACE2 rs2285666 Polymorphism With COVID ...
    Sep 24, 2020 · It was unanimously shown that a polymorphism rs2285666 present in ACE2, varied significantly among European and Asians (Asselta et al., 2020; ...Missing: conservation mammals
  14. [14]
  15. [15]
    ACE2 in chronic disease and COVID-19: gene regulation and post ...
    Aug 22, 2023 · These results indicate that DYRK1A increases ACE2 mRNA levels by promoting chromatin accessibility, leading to enhancement of SARS-CoV-2 ...Missing: ESR1 | Show results with:ESR1
  16. [16]
    Role of HIF-1α in the regulation ACE and ACE2 expression in ...
    We conclude that ACE is directly regulated by HIF-1α, whereas ACE2 is regulated in a bidirectional way during hypoxia and may play a protective role during the ...
  17. [17]
    Angiotensin-converting enzyme 2 is subject to post-transcriptional ...
    The ability of miR-421, an miRNA implicated in the development of thrombosis, to down-regulate ACE2 expression was subsequently confirmed by Western blot ...
  18. [18]
    Interferons and viruses induce a novel truncated ACE2 isoform and ...
    Oct 19, 2020 · We report the discovery of a novel, transcriptionally independent truncated isoform of ACE2, which we designate as deltaACE2 (dACE2).
  19. [19]
  20. [20]
    Men more vulnerable to COVID-19: explained by ACE2 on the X ...
    Jun 24, 2020 · The mere occurrence of ACE2 on the X chromosome could also be important in explaining why men would suffer more from ACE2-related diseases than women.
  21. [21]
    The protein expression profile of ACE2 in human tissues
    ACE2 expression was mainly observed in enterocytes, renal tubules, gallbladder, cardiomyocytes, male reproductive cells, placental trophoblasts, ductal cells, ...
  22. [22]
    Tissue expression of ACE2 - Summary - The Human Protein Atlas
    Membranous expression in ciliated cells in nasal mucosa, bronchus, and fallopian tube. Expressed in endothelial cells and pericytes in many tissues.Missing: database | Show results with:database
  23. [23]
    A comprehensive investigation of the mRNA and protein level of ...
    Jun 18, 2020 · Therefore, the distribution of ACE2 in human tissues may indicate the susceptibility of different human organs to SARS-CoV-2 infection [10].
  24. [24]
    The protein expression profile of ACE2 in human tissues - PMC - NIH
    The highest expression was observed in the intestinal tract, followed by kidney, testis, gallbladder and heart. A few tissues showed a lower expression of < 5 ...
  25. [25]
    assessing the expression of ACE2/TMPRSS2 in the human fetus ...
    ACE2 is present at significant levels only in the fetal intestine and kidney, and is not expressed in the fetal lung. The placenta also does not co‐express the ...<|separator|>
  26. [26]
    The SARS-CoV-2 receptor ACE2 expression of maternal-fetal ...
    Apr 16, 2020 · Our results revealed that ACE2 was highly expressed in maternal-fetal interface cells including stromal cells and perivascular cells of decidua, ...
  27. [27]
    Comparative genetic analysis of the novel coronavirus (2019-nCoV ...
    Feb 24, 2020 · To analyze the distribution of eQTLs for ACE2, we used the Genotype Tissue Expression (GTEx) database (https://www.gtexportal.org/home/datasets) ...Missing: body | Show results with:body
  28. [28]
    Comparative analysis of ACE2 protein expression in rodent, non ...
    Feb 24, 2021 · ACE2 protein expression demonstrates a significant gradient between upper and lower respiratory tract in humans and is scarce in the lung.
  29. [29]
    The ACE2 Receptor for Coronavirus Entry Is Localized at Apical Cell ...
    Here we explored the expression and localization of ACE2 and its association with transmembrane and tight junction proteins in epithelial tissues and cultured ...Missing: immunohistochemistry | Show results with:immunohistochemistry
  30. [30]
    The soluble catalytic ectodomain of ACE2 a biomarker of cardiac ...
    Jan 6, 2021 · ACE2 membrane-anchored protein at the catalytically active ectodomain undergoes shedding by the ADAM17, forming a soluble form of ACE2. ACE ...
  31. [31]
    ACE2 Nascence, trafficking, and SARS-CoV-2 pathogenesis - NIH
    In the Golgi, ACE2 undergoes further modifications and packaging and is then transported to the plasma membrane by vesicular transport (Fig. 3a) [116]. As ...
  32. [32]
    Sequences in the Cytoplasmic Tail Contribute to the Intracellular ...
    ... ACE2 surface expression, indicating endocytic recycling of this receptor. The S protein has been shown to inhibit the endocytic recycling of ACE2 by ...
  33. [33]
    Differential Expression of Neuronal ACE2 in Transgenic Mice with ...
    These results suggest that ACE2 is localized to the cytoplasm of neuronal cells in the brain, and that ACE2 levels appear highly regulated by other components ...Missing: tight junctions perinuclear dynamic
  34. [34]
    ACE2 and ACE: structure-based insights into mechanism, regulation ...
    Nov 4, 2020 · In addition, N-domain Asn131 and C-domain Asn155 are on a short loop between the β1 and β2 strands and, while this loop is two residues longer ...
  35. [35]
  36. [36]
  37. [37]
    Angiotensin Converting Enzyme 2, Angiotensin-(1-7), and Receptor ...
    However, the main substrate for ACE2 is Ang II, which is converted into Ang-(1-7) [7]. Consequently, ACE2 plays a pivotal role in the balance between both RAS ...
  38. [38]
    ACE and ACE2: a tale of two enzymes | European Heart Journal
    Although there are many peptidases which can hydrolyse Ang II to Ang-(1–7), ACE2 appears to be the major pathway for Ang-(1–7) formation from Ang II in the ...
  39. [39]
    Hydrolysis of Biological Peptides by Human Angiotensin-converting ...
    The catalytic activity of human soluble ACE2 was found to have a pH optimum of 6.5 in the presence of 1.0 m NaCl. There is a sharp pH dependence under acidic ...Missing: enzymatic | Show results with:enzymatic
  40. [40]
    ACE2, a new regulator of the renin–angiotensin system - PMC
    In addition to effects on the angiotensin peptides, ACE2 cleaves the C-terminal residue of the peptides des-Arg9-bradykinin, neurotensin 1–13 and kinetensin [4] ...
  41. [41]
    ACE2/ANG-(1–7)/Mas pathway in the brain: the axis of good - PMC
    In this review, we will summarize the role of the ACE2/ANG-(1–7)/Mas receptor, focusing on the central nervous system with respect to cardiovascular diseases.
  42. [42]
    Angiotensin-(1-7) and Mas receptor in the brain
    ACE2 also acts on Ang I, although with an enzymatic efficiency 400 times lower, generating Ang-(1-9), which can be cleaved to Ang-(1-7) by ACE or NEP [13]. Both ...Missing: intermediate | Show results with:intermediate
  43. [43]
    A SARS-CoV-2 protein interaction map reveals targets for drug ...
    Apr 30, 2020 · We identify 66 druggable human proteins or host factors targeted by 69 compounds (of which, 29 drugs are approved by the US Food and Drug Administration)
  44. [44]
    Osmotic Adaptation by Na+-Dependent Transporters and ACE2
    This review provides a comprehensive summary of the role of ACE2 in the assembly of Na + -dependent transporters of glucose, imino and neutral amino acids.
  45. [45]
    Role and Interaction Between ACE1, ACE2 and Their Related ...
    This paper reviews the role and interaction between ACE1, ACE2 and their related genes in cardiovascular disorders.
  46. [46]
    Allosteric Binders of ACE2 Are Promising Anti-SARS-CoV-2 Agents
    Jun 22, 2022 · In the enzymatic assay, MLN-4760 showed a dose-dependent inhibition of ACE2 activity with a half-maximum inhibitory concentration (IC50) of 1.50 ...Results and Discussion · Materials and Methods · Author Information · References
  47. [47]
    A human homolog of angiotensin-converting enzyme ... - PubMed
    A human homolog of angiotensin-converting enzyme. Cloning and functional expression as a captopril-insensitive carboxypeptidase. J Biol Chem. 2000 Oct 27;275(43): ...
  48. [48]
    Angiotensin-converting enzyme 2 is an essential regulator of heart ...
    Jun 20, 2002 · Targeted disruption of ACE2 in mice results in a severe cardiac contractility defect, increased angiotensin II levels, and upregulation of ...
  49. [49]
    Deletion of Angiotensin-Converting Enzyme 2 Accelerates Pressure ...
    Apr 1, 2006 · ... ACE2 is much higher than that of angiotensin I to angiotensin 1-9. Crackower et al reported that 3-month-old mice lacking ACE2 (ACE2−/y mice) ...
  50. [50]
    Angiotensin-(1–7) and Vascular Function | Hypertension
    Dec 4, 2017 · Ang-(1–7) induces vasodilatory, anti-inflammatory, antifibrotic, antiangiogenic, and antihypertensive effects by binding to its G-protein– ...
  51. [51]
    ACE2, angiotensin-(1-7) and Mas receptor axis in inflammation and ...
    Many studies have now shown that Ang-(1-7) by acting via Mas receptor exerts inhibitory effects on inflammation and on vascular and cellular growth mechanisms.Missing: intermediate | Show results with:intermediate
  52. [52]
    Regulation of angiotensin converting enzyme II ... - ScienceDirect.com
    Ang II upregulates ACE2 expression in HCF cells via the AT1R and the ERK–MAPK pathway. In HCF cells treated with Ang II, the amount of ACE2 mRNA was markedly ...
  53. [53]
  54. [54]
    ACE2/Ang-(1-7)/Mas1 axis and the vascular system - NIH
    Jan 29, 2021 · (2015) ACE2 and Ang-(1-7) protect endothelial cell function and prevent early atherosclerosis by inhibiting inflammatory response. Inflamm ...
  55. [55]
    Angiotensin II and Cardiovascular Disease - PubMed Central - NIH
    Jul 1, 2025 · ... ACE2 provides further cardiovascular protection by lowering oxidative stress, inflammation, and abnormal cell growth. Bearing these in mind ...
  56. [56]
    Upregulation of Angiotensin (1-7)-Mediated Signaling Preserves ...
    Animal studies show that ACE2 overexpression improves endothelial function in hypertension (43), attenuates the progression of atherosclerosis (29, 59), and ...
  57. [57]
    Role of Angiotensin II in the Development of Nephropathy and ... - NIH
    ACE2 has been localized to podocytes, and in db/db diabetic mice glomerular expression of ACE2 is reduced whereas glomerular ACE expression is increased [42].
  58. [58]
    Diabetic Nephropathy: Novel Molecular Mechanisms and ... - NIH
    In this regard, the selective overexpression of human ACE2 in the podocytes attenuated the development of nephropathy in mice with streptozotocin-induced type ...
  59. [59]
    Ang-(1-7) attenuates podocyte injury induced by high glucose in vitro
    Activation of the ACE2/Ang-(1-7)/MasR axis plays a protective role by reducing oxidative stress, inflammation, cell proliferation, and fibrosis (30), which ...
  60. [60]
    COVID-19: angiotensin-converting enzyme 2 (ACE2) expression ...
    Jan 3, 2021 · Angiotensin-converting enzyme 2 (ACE2) is not only an enzyme but also a functional receptor on cell surfaces through which SARS-CoV-2 enters the ...
  61. [61]
    Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19 - PMC
    Jul 13, 2020 · Several studies have revealed the protective effects of the ACE2/angiotensin-(1–7)/MAS axis in the lungs. It alleviates lung inflammation, ...
  62. [62]
    Role of Angiotensin Converting Enzyme-2 and its modulation in ...
    This review aims at probing the role of ACE2 and its therapeutic modulation in pulmonary and extra pulmonary diseases, including COVID-19.<|control11|><|separator|>
  63. [63]
    ACE2 links amino acid malnutrition to microbial ecology ... - Nature
    Jul 25, 2012 · Our results identify ACE2 as a key regulator of dietary amino acid homeostasis, innate immunity, gut microbial ecology, and transmissible susceptibility to ...
  64. [64]
    Neuroprotective Mechanisms of the ACE2–Angiotensin-(1-7)
    Accumulating evidence has demonstrated that activation of the ACE2–Ang-(1-7)–Mas axis can exert profound neuroprotective effects in both ischemic and ...
  65. [65]
    MOLECULAR DISSECTION OF THE ROLE OF ACE2 IN GLUCOSE ...
    Jul 1, 2025 · ACE2 plays a pivotal role in diabetes by regulating glucose homeostasis in a tissue-specific manner. In the pancreas, ACE2 was shown to promote insulin ...
  66. [66]
    SARS-CoV-2, ACE2 expression, and systemic organ invasion - PMC
    Angiotensin-converting enzyme 2 (ACE2) is a membrane-bound enzyme, which both SARS-CoV and SARS-CoV-2 use as a receptor for host cellular entry (1, 3, 4).
  67. [67]
  68. [68]
  69. [69]
  70. [70]
  71. [71]
  72. [72]
    Angiotensin‐converting enzyme 2 catalytic activity in human plasma ...
    An expression construct for secreted soluble ACE2 was generated by fusing the interleukin‐3 signal sequence and a Flag tag to the N‐terminus of the ...Assay For Ace2 Activity · Assay For Ace Activity · Results
  73. [73]
    Human Recombinant ACE2 Protein, aa18-740 (HEK293-expressed)
    Human Recombinant ACE2 Protein, aa18-740 is expressed in HEK293 cells and is a type I transmembrane protein involved in regulating angiotensin.Missing: 2007 soluble ectodomain residues
  74. [74]
    New mass spectrometric assay for angiotensin-converting enzyme 2 ...
    A novel assay was developed for evaluation of mouse angiotensin-converting enzyme (ACE) 2 and recombinant human ACE2 (rACE2) activity.
  75. [75]
    A pan-SARS-CoV-2-specific soluble angiotensin-converting enzyme ...
    This is due to the fact that soluble dimeric human ACE2 has a plasma half-life of only 10 h (44), which is not optimal in an infection setting requiring both ...
  76. [76]
    The Safety, Tolerability, PK and PD of GSK2586881 in Patients With ...
    This is an early phase (phase IIa), randomized, multi-center study in subjects with acute lung injury (ALI) or acute respiratory distress syndrome (ARDS).
  77. [77]
    A pilot clinical trial of recombinant human angiotensin-converting ...
    Sep 7, 2017 · We postulated that repleting ACE2 using GSK2586881, a recombinant form of human angiotensin-converting enzyme 2 (rhACE2), could attenuate acute lung injury.
  78. [78]
    APN01 – Our work on a potential drug candidate for COVID-19 ...
    APN01 has the potential to prevent cells from infection with the SARS-CoV-2 virus, reduce injury to multiple organs and additionally to treat the inflammatory ...Missing: 2020-2022 | Show results with:2020-2022
  79. [79]
    Phase I dose-escalation study to assess the safety, tolerability ...
    Feb 19, 2024 · In COVID-19 patients, 0.4 mg·kg−1 i.v. APN01 administered twice daily for 7 days was found to be safe and well tolerated, with the proportion ...Missing: hrsACE2 outcomes
  80. [80]
    Tolerability of soluble ACE2 (APN01) administered via nebulizer
    We hypothesized that aerosol administration of clinical grade soluble human recombinant ACE2 (APN01) will neutralize SARS-CoV-2 in the airways.