Bradykinin is a linear nonapeptide with the amino acid sequence Arg¹-Pro²-Pro³-Gly⁴-Phe⁵-Ser⁶-Pro⁷-Phe⁸-Arg⁹, belonging to the kinin family of peptides and serving as a key mediator in the kallikrein-kinin system.[1] It is generated through the proteolytic cleavage of high-molecular-weight kininogen (HMWK) by plasma kallikrein or, indirectly, from low-molecular-weight kininogen via tissue kallikrein to form lysyl-bradykinin (kallidin), which is then converted to bradykinin.[2] This short-lived peptide exerts potent vasodilatory effects, increases vascular permeability, and sensitizes pain receptors, contributing to the cardinal signs of inflammation such as redness, swelling, heat, and pain.[3]Bradykinin primarily acts through two G protein-coupled receptors: the constitutive B₂ receptor, which is widely expressed and mediates most physiological responses, and the inducible B₁ receptor, which is upregulated during inflammation and tissueinjury.[3] Physiologically, it promotes blood flow to tissues, supports wound healing, and exhibits anticoagulant properties by stimulating prostacyclin release from endothelial cells.[2] However, dysregulation of bradykinin signaling underlies several pathological conditions, including hereditary angioedema due to C1 esterase inhibitor deficiency, where unchecked production leads to severe swelling, and adverse effects from angiotensin-converting enzyme (ACE) inhibitors, which prolong bradykinin half-life by inhibiting its degradation.[2]Recent research has highlighted bradykinin's role in inflammatory and infectious diseases, such as its contribution to the "bradykinin storm" hypothesized in severe COVID-19 cases, where excessive accumulation drives vascular leakage, hypoxia, and acute respiratory distress syndrome.[3] Therapeutically, bradykinin receptor antagonists like icatibant target B₂ receptors to manage acute attacks in hereditary angioedema, underscoring the peptide's clinical significance.[3]
Molecular Structure and Properties
Chemical Composition
Bradykinin is a linear nonapeptide composed of nine amino acids in the sequence Arg¹-Pro²-Pro³-Gly⁴-Phe⁵-Ser⁶-Pro⁷-Phe⁸-Arg⁹, where the arginine residues at both termini contribute to its charged nature. This sequence was first elucidated through biochemical analysis of plasma kinins derived from bovine blood. The peptide's structure features a combination of hydrophobic phenylalanine residues and hydrophilic polar groups, which influence its interactions in biological systems.The empirical formula of bradykinin is C₅₀H₇₃N₁₅O₁₁, reflecting the atomic composition from its amino acid building blocks, and its molecular weight is 1,060.228 g/mol. These properties position bradykinin as a small, bioactive molecule within the kinin family.Bradykinin demonstrates high solubility in water, exceeding 40 mg/mL at room temperature, owing to its polar amino acid residues including arginine, serine, and glycine that facilitate hydrogen bonding with solvent molecules. It maintains stability in aqueous environments at physiological pH around 7.4, allowing it to function effectively in bodily fluids prior to enzymatic processing. In contrast to related kinins, bradykinin lacks an N-terminal lysine residue present in kallidin (also known as Lys-bradykinin), a decapeptide analog that shares the core sequence but exhibits distinct metabolic origins and receptor affinities.
Three-Dimensional Structure
Bradykinin adopts a predominantly random coil conformation in aqueous solution, reflecting its inherent flexibility as a small nonapeptide hormone, as evidenced by nuclear magnetic resonance (NMR) spectroscopy studies that show averaged signals indicative of rapid motional averaging without stable secondary structure elements.[4] This flexibility allows the peptide to sample a range of conformations, which is crucial for its interactions with diverse biological targets. In the absence of stabilizing environments, such as membranes or receptors, bradykinin lacks rigid helical or sheet structures, prioritizing dynamic adaptability over fixed folding.[5]NMR investigations, including two-dimensional techniques like NOESY, combined with distance geometry and restrained molecular dynamics, have identified transient secondary structure features, notably a possible type II β-turn centered at the Pro³-Gly⁴-Phe⁵ sequence in solution or under mildly constraining conditions.[6] This turn involves hydrogen bonding between the carbonyl of Pro³ and the amide of Phe⁵, contributing to a folded motif in a subset of conformations. Additionally, in membrane-mimetic environments like SDS micelles or phospholipid vesicles, a β-turn-like structure emerges at residues Ser⁶-Pro⁷-Phe⁸-Arg⁹, stabilizing the C-terminal region.[7] These insights highlight how environmental factors induce localized folding from the default disordered state.The peptide's conformational dynamics involve an equilibrium between extended and more compact states, with the N- and C-termini exhibiting high flexibility—marked by rapid tumbling of Arg¹ and Arg⁹—while a central hydrophobic core forms transiently through interactions among Phe⁵, Pro⁷, and Phe⁸ residues.[4] In solution, NMR relaxation data and Monte Carlo simulations reveal multiple conformational families, including type I and distorted type II β-turns, underscoring the peptide's ability to shift populations in response to solvent or lipid interactions without a single dominant structure.[4] No high-resolution X-ray crystallographic structures of free bradykinin exist due to its flexibility, but NMR-derived models provide atomic-level details of these dynamics.[5]While native bradykinin lacks common post-translational modifications like phosphorylation or glycosylation, studies on synthetic variants demonstrate that such alterations can rigidify the structure; for instance, phosphorylation at Ser⁶ in analogs stabilizes β-turns and alters the hydrophobic core, potentially influencing activity, though these effects are not observed in the unmodified peptide.[8]
Biosynthesis and Metabolism
Synthesis Pathways
Bradykinin is generated endogenously through the kinin-kallikrein system (KKS), a proteolytic cascade that liberates kinins from precursor proteins known as kininogens, playing a central role in vascular and inflammatory responses.[3] The system involves serine proteases called kallikreins that cleave kininogens to release active peptides, with bradykinin specifically produced as a nonapeptide (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg).[9] This process occurs primarily in plasma and tissues, contributing to the localized and systemic actions of kinins.[3]The key precursors for bradykinin synthesis are high-molecular-weight kininogen (HMWK) and low-molecular-weight kininogen (LMWK), both encoded by the KNG1 gene on chromosome 3q26 through alternative splicing of a common pre-mRNA.[3]HMWK, a 120 kDa glycoprotein synthesized mainly in the liver, circulates in plasma bound to prekallikrein and factor XI, featuring a 56 kDa light chain that harbors the bradykinin moiety.[3] In contrast, LMWK, approximately 70 kDa, is also liver-derived but more prevalent in tissues, with a shorter 4 kDa light chain containing a lysine-extended bradykinin sequence.[3] These structural differences dictate their substrate specificity in distinct KKS pathways.[9]The plasma pathway of bradykinin synthesis begins with the activation of the contact system in the coagulation cascade, where exposure to negatively charged surfaces triggers factor XII (Hageman factor) to autoactivate into factor XIIa.[9] Factor XIIa then converts plasma prekallikrein (a 88 kDa zymogen) to active plasma kallikrein, which proteolytically cleaves HMWK at specific arginine-serine bonds to liberate free bradykinin.[9] This pathway links kinin generation to hemostasis and is amplified by high-molecular-weight multimers of HMWK serving as cofactors.[3]In the tissue pathway, glandular or tissue kallikreins—serine proteases encoded by a cluster of 15 KLK genes on chromosome 19q13—predominantly act on LMWK to release kallidin (lysyl-bradykinin), a decapeptide intermediate.[3] Kallidin is then rapidly converted to bradykinin by the action of aminopeptidase B, which removes the N-terminal lysine residue.[3] Tissue kallikreins are expressed in various organs, such as the pancreas, salivary glands, and kidneys, facilitating localized kinin production for tissue-specific functions like repair and fluid balance.[3] Unlike the plasma route, this pathway operates independently of coagulation factors and is more prominent in extravascular sites.[9]Regulation of bradykinin synthesis integrates with broader physiological controls, particularly the coagulation cascade for the plasma pathway, where inhibitors like C1-esterase inhibitor modulate factor XII and kallikrein activity to prevent excessive kinin release.[9] Hormonal influences further fine-tune the system; for instance, estrogen elevates plasma levels of factor XII, plasma kallikrein, and bradykinin in females, likely via enhanced gene transcription, thereby increasing KKS activity during reproductive cycles.[10] This estrogen-mediated upregulation contributes to variations in kininogen expression and enzymatic efficiency across physiological states.[10]
Degradation Mechanisms
Bradykinin exhibits a remarkably short half-life in human plasma, typically around 30 seconds, due to rapid enzymatic hydrolysis that prevents prolonged vasoactive effects and maintains homeostasis.[11] This swift degradation is mediated primarily by membrane-bound and soluble peptidases in the vasculature, particularly in the lungs and kidneys, where bradykinin circulates and exerts its physiological actions. The process involves sequential cleavage at specific peptide bonds, resulting in inactive fragments that are further metabolized or excreted.The primary enzyme responsible for bradykinin catabolism is angiotensin-converting enzyme (ACE, also known as kininase II), a zinc metallopeptidase expressed on endothelial cells. ACE hydrolyzes the Pro⁷-Phe⁸ bond in the bradykinin sequence (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg), releasing the C-terminal dipeptide Phe-Arg and generating the heptapeptide bradykinin-(1-7), which is biologically inactive.[12] This cleavage accounts for the majority of bradykinin inactivation in plasma, underscoring ACE's central role in kinin system regulation.[13]Neutral endopeptidase (NEP, or neprilysin), another zinc-dependent endopeptidase, contributes significantly to bradykinin degradation, especially in tissues beyond the pulmonary circulation. NEP cleaves multiple internal bonds, including Phe⁵-Ser⁶ and Ser⁶-Pro⁷, producing fragments such as bradykinin-(1-4) and bradykinin-(1-7) that diminish its activity.[13]Aminopeptidase P (APP), a proline-specific exopeptidase, acts at the N-terminus by hydrolyzing the Arg¹-Pro² bond, yielding bradykinin-(2-9) and rendering the peptide inactive for further processing by other enzymes.[14] These enzymes collectively ensure efficient clearance, with their activities varying by tissue and physiological state.Inhibition of these degradative pathways alters bradykinin dynamics; for instance, ACE inhibitors, such as enalapril, extend its plasma half-life by up to 12-fold by blocking the Pro⁷-Phe⁸ cleavage, thereby elevating local concentrations and potentiating effects like vasodilation.[11] This mechanism highlights the therapeutic implications of targeting bradykinin metabolism while also explaining associated risks in clinical use.[13]
Physiological Functions
Cellular and Tissue Effects
Bradykinin exerts diverse effects on cellular and tissue levels, primarily influencing vascular and smooth muscle functions as well as sensory responses. It promotes vasodilation by relaxing arteriolar smooth muscle, which increases blood flow and contributes to hypotension through the release of nitric oxide (NO) and prostaglandins from endothelial cells.[2][9] This relaxation enhances tissue perfusion but can lead to systemic drops in blood pressure when bradykinin levels rise. Additionally, bradykinin induces contraction in non-vascular smooth muscles, notably in the bronchial airways and gastrointestinal tract, altering airflow resistance and gut motility.[2][9]In sensory tissues, bradykinin contributes to pain and inflammatory responses by sensitizing nociceptors, particularly through pathways involving prostaglandin E2 (PGE2), which amplifies heat and mechanical stimuli detection.[15] This sensitization heightens pain perception without directly causing tissue damage. On vascular endothelium, bradykinin increases permeability by inducing contractions that form intercellular gaps between endothelial cells, facilitating plasma extravasation and localized edema formation.[16][17]These effects exhibit dose-dependency, with low concentrations predominantly eliciting vasodilation and permeability increases, while higher doses can shift toward contractile responses in certain vascular beds, such as afferent arterioles.[18][19] This biphasic nature allows bradykinin to fine-tune physiological adjustments based on local concentrations.
Receptor Interactions
Bradykinin primarily exerts its effects through two G-protein-coupled receptors: the B2 receptor (BDKRB2), which is constitutively expressed in most tissues, and the B1 receptor (BDKRB1), which is inducible and upregulated under inflammatory conditions such as tissue injury or exposure to cytokines like IL-1β and TNF-α.[3][20] The B2 receptor binds bradykinin with high affinity (Kd ≈ 0.4–1 nM), while the B1 receptor preferentially binds des-Arg⁹-bradykinin (Kd ≈ 1–3 nM), a metabolite of bradykinin generated by carboxypeptidase N.[21][22] These receptors are seven-transmembrane proteins encoded by genes on chromosome 14 (both BDKRB1 and BDKRB2).[23] and their activation transduces signals via distinct G-protein pathways.[23]The B2 receptor predominantly couples to Gq/11 proteins, activating phospholipase C (PLC) to hydrolyze phosphatidylinositol 4,5-bisphosphate into inositol 1,4,5-trisphosphate (IP₃) and diacylglycerol, which mobilizes intracellular Ca²⁺ and activates protein kinase C, leading to downstream effects like nitric oxide release.[24] It can also couple to Gi/o proteins in certain contexts, inhibiting adenylyl cyclase. In contrast, the B1 receptor primarily signals through Gq/11, similarly increasing Ca²⁺ via PLC-IP₃, but also engages Gi/o to modulate MAPK and NF-κB pathways, promoting pro-inflammatory responses.[3][25] These signaling cascades are well-characterized in seminal studies, such as those by Regoli et al., which established the pharmacological distinction between B1 and B2 subtypes.[26]Receptor desensitization occurs primarily through phosphorylation by G-protein-coupled receptor kinases (GRKs) at C-terminal serine and threonine residues, followed by β-arrestin recruitment, which facilitates clathrin-mediated internalization.[24] For the B2 receptor, this leads to rapid tachyphylaxis (homologous desensitization within minutes) and either lysosomal degradation or recycling to the plasma membrane, depending on exposure duration.[3] The B1 receptor exhibits less pronounced desensitization, with agonist-independent internalization and sustained surface expression during inflammation.[3]Genetic variations in the BDKRB2 gene, particularly the -9/+9 insertion/deletion polymorphism in the 3' untranslated region (rs5810761), influence receptor expression levels and functional responses, such as altered nitric oxide production and inflammatory cytokine release in conditions like osteoarthritis.[23][27] The +9/+9 genotype is associated with enhanced receptor stability and heightened signaling, impacting vascular tone and pain sensitivity, as demonstrated in high-impact genetic association studies.[28][29]
Pathophysiological Roles
Associated Disorders
Dysregulation of bradykinin activity is central to several disorders characterized by systemic imbalances, primarily manifesting as excessive vascular permeability and edema. Hereditary angioedema (HAE) due to C1-inhibitor (C1-INH) deficiency represents the prototypical condition, where insufficient C1-INH leads to uncontrolled activation of the kallikrein-kinin system and overproduction of bradykinin.[30] This autosomal dominant disorder affects approximately 1 in 50,000 individuals worldwide, with symptoms including recurrent episodes of non-pitting subcutaneous or submucosal edema, often affecting the face, extremities, genitals, or gastrointestinal tract, and accompanied by severe abdominal pain due to intestinal wall swelling.[31] In HAE, bradykinin excess directly increases endothelial permeability without urticaria or pruritus, distinguishing it from histamine-mediated reactions.[32]Angiotensin-converting enzyme (ACE) inhibitor-induced angioedema and cough arise from impaired bradykinin degradation, as ACE normally metabolizes bradykinin alongside angiotensin II.[33] This accumulation occurs in 0.1-0.7% of patients on ACE inhibitors, presenting as orofacial or laryngeal edema similar to HAE,[34] and a dry, persistent cough in approximately 1.5-11% of cases, particularly in certain populations due to bradykinin receptor polymorphisms.[35] The cough mechanism involves bradykinin sensitization of airway sensory nerves, leading to heightened tussive reflexes.[36]In allergic reactions, bradykinin contributes to anaphylaxis by amplifying vascular permeability alongside histamine, promoting fluid extravasation in severe systemic responses.[37] This synergistic effect exacerbates hypotension and edema during mast cell degranulation triggered by allergens.[38]Bradykinin exhibits paradoxical effects in hypertension regulation, acting as a potent vasodilator to lower blood pressure under normal conditions, yet reduced bradykinin activity is associated with elevated blood pressure in experimental and clinical hypertension models.[39] In the context of ACE inhibition, enhanced bradykinin signaling contributes to antihypertensive efficacy but can precipitate adverse vascular events in susceptible individuals.[40]
Role in Specific Diseases
Bradykinin has been implicated in the pathophysiology of COVID-19 through dysregulation of the kallikrein-kinin system, potentially contributing to a "bradykinin storm" characterized by excessive vascular permeability, inflammation, and cytokine release in the lungs.[41] This hypothesis, often referred to as the kallikrein-kinin paradox, arises from impaired bradykinin degradation, leading to heightened B2 receptor signaling and endothelial dysfunction, which may exacerbate severe respiratory symptoms.[42] Studies, including those up to 2025, have linked this mechanism to the renin-angiotensin system's imbalance during SARS-CoV-2 infection, though the exact contributions remain debated due to overlapping pathways like complement activation.[43] A 2025 post-hoc analysis of the ICAT-COVID clinical trial of the bradykinin antagonist icatibant suggested potential benefits in reducing pneumonia severity and mortality in severe cases.[44]In various cancers, bradykinin promotes tumor progression by enhancing angiogenesis and facilitating metastasis, primarily through activation of B2 receptors on endothelial and stromal cells. For instance, in prostate cancer, tissue kallikrein-generated bradykinin stimulates B2 receptor signaling to support vascularization and tumor growth in both cell lines and patient tissues.[45] Similarly, in ovarian cancer, bradykinin contributes to lymphangiogenesis and invasion by modulating stromal interactions, underscoring its role in metastatic spread.[46] These effects are mediated by both B1 and B2 receptors, with antagonists showing potential to inhibit angiogenic responses in preclinical models.[47]Bradykinin links to metabolic syndrome through its influence on inflammatory pathways that drive insulin resistance and obesity. In obese models, bradykinin exerts proinflammatory effects in adipose tissue, promoting cytokine release and macrophage infiltration that impair insulin signaling.[48] Conversely, it can enhance insulin sensitivity in skeletal muscle and inhibit hepatic gluconeogenesis, highlighting context-dependent actions via B2 receptors.[49] The kallikrein-kinin system's activation in brown adipose tissue further ties bradykinin to thermogenesis deficits, exacerbating obesity-related metabolic dysfunction.[50]In neurological disorders, bradykinin contributes to pain mechanisms, particularly in migraine and neuropathic pain. During migraine attacks, bradykinin sensitizes trigeminal sensory neurons, amplifying prostaglandin-mediated inflammation and central nociceptive signaling.[51] In neuropathic pain, kinins via B1 and B2 receptors induce hyperalgesia and allodynia by enhancing neuronal excitability and inflammatory cascades in peripheral nerves.[52]Estrogen modulation of bradykinin signaling in trigeminal ganglia may further explain sex differences in migraine susceptibility.[53]Recent studies from 2023 to 2025 have explored bradykinin's role in long COVID and post-viral syndromes, with evidence suggesting persistent kallikrein-kinin dysregulation contributes to symptoms like fatigue and endothelial dysfunction.[54] Autoantibodies targeting G protein-coupled receptors, including those in the kinin pathway, have been associated with prolonged inflammation in long COVID patients.[55] However, research gaps remain, as direct causal links to post-viral syndromes are still emerging and require further validation beyond acute infection models.
Therapeutic Applications
Pharmacological Targets
Bradykinin activity can be modulated pharmacologically through targeting enzymes involved in its synthesis and degradation, as well as its specific receptors. Key degradative enzymes include angiotensin-converting enzyme (ACE) and neutral endopeptidase (NEP), both of which cleave bradykinin to inactive fragments, thereby regulating its bioavailability.[13] Inhibition of ACE, a zinc metalloprotease, prevents bradykinin breakdown and elevates its levels, contributing to vasodilatory and inflammatory effects observed with ACE inhibitors.[56] Similarly, NEP degrades bradykinin alongside other vasoactive peptides, and its inhibition—often via dual ACE/NEP inhibitors—enhances bradykinin-mediated responses, though this approach requires careful management to avoid excessive accumulation.[57] Enhancing degradation through these enzymes is less commonly targeted but can be explored via recombinant enzymes or potentiators in research settings to mitigate bradykinin excess.[58]Bradykinin exerts its effects primarily via two G protein-coupled receptors, B1 and B2, with B2 being constitutively expressed and mediating most physiological actions like vasodilation.[42] Selective antagonists for these receptors represent major pharmacological targets for dampening bradykinin signaling. For the B2 receptor, icatibant (HOE-140), a stable peptide analog of bradykinin, acts as a potent, competitive antagonist that blocks B2-mediated vascular permeability and pain pathways without significant agonist activity.[59] B1 receptor antagonists, such as des-Arg¹⁰-HOE-140, target the inducible B1 receptor, which is upregulated in inflammation and contributes to chronic pain and hypersensitivity; this compound is a modified derivative of HOE-140 with enhanced B1 selectivity.[60]Agonists of bradykinin receptors have limited clinical application and are primarily used in research to study vasodilation and endothelial function. Synthetic B2 receptor agonists, such as [Hyp³]-bradykinin derivatives, mimic bradykinin's ability to induce endothelium-dependent vasodilation via nitric oxide release, aiding investigations into cardiovascular responses.[61]Upstream inhibition of the kallikrein-kinin pathway provides another target by reducing bradykinin generation from high-molecular-weight kininogen. Plasma kallikrein inhibitors, exemplified by ecallantide (DX-88), a recombinant peptide, selectively block kallikrein activity to limit bradykinin production, particularly in conditions involving pathway overactivation.[62]Developing selective modulators faces challenges due to off-target effects in the multi-receptor kinin system, where antagonists like icatibant can inadvertently inhibit enzymes such as aminopeptidase N, potentially altering unrelated peptide processing.[63] Additionally, achieving high selectivity between B1 and B2 receptors is complicated by structural similarities, leading to cross-reactivity that may exacerbate side effects in inflammatory contexts.[64]
Clinical Uses and Drugs
ACE inhibitors, such as captopril and enalapril, are widely used to treat hypertension by inhibiting angiotensin-converting enzyme (ACE), which indirectly increases bradykinin levels and contributes to vasodilation.[65] A common side effect is a dry, non-productive cough occurring in 5–20% of patients, attributed to elevated bradykinin accumulation in the lungs.[66] This cough typically resolves upon discontinuation of the drug.[67]Bradykinin B2 receptor antagonists like icatibant (Firazyr) are approved for the treatment of acute attacks of hereditary angioedema (HAE) in adults aged 18 years and older.[68] The recommended dose is 30 mg administered subcutaneously in the abdominal area, with additional doses possible at intervals of at least 6 hours if needed, up to a maximum of three doses in 24 hours.[69] Self-administration is permitted following proper training.[70]C1-inhibitor replacements, such as Berinert (C1 esterase inhibitor, human), are used for short-term prophylaxis against HAE attacks, particularly before medical procedures or surgery.[71] The dosage for short-term prophylaxis is 10–20 international units per kg body weight, administered intravenously 1 hour before the procedure or within 6 hours prior.[71]Berinert is also indicated for acute treatment of HAE attacks at a dose of 20 international units per kg intravenously.[72]Investigational expansions for icatibant (Firazyr) include potential broader applications in HAE management beyond acute attacks, though no new indications have been approved as of 2025.[73]Neprilysin (NEP) inhibitors, such as sacubitril in combination with valsartan (Entresto), are approved for heart failure with reduced ejection fraction and elevate bradykinin levels by inhibiting its degradation, contributing to cardioprotective effects.[74] Despite preclinical interest, no bradykinin-specific therapies for COVID-19 have advanced to approval from trials conducted between 2023 and 2025, with research remaining focused on hypotheses rather than clinical outcomes.[75]Post-2023 approvals in the HAE space include garadacimab (Andembry), a monoclonal antibody targeting factor XIIa upstream in the bradykinin pathway, approved in June 2025 for once-monthly subcutaneous prophylaxis in adults and pediatric patients aged 12 years and older.[76] Additionally, sebetralstat (Ekterly), an oral plasma kallikrein inhibitor affecting bradykinin generation, was approved in July 2025 as the first on-demand treatment for acute HAE attacks in patients aged 12 years and older.[77] In August 2025, donidalorsen (Dawnzera), an antisense oligonucleotide targeting prekallikrein to reduce bradykinin production, was approved for subcutaneous prophylactic treatment to prevent HAE attacks in adults and adolescents aged 12 years and older.[78]
History and Research
Discovery
Bradykinin was discovered in 1948 by Brazilian physiologists Maurício Rocha e Silva, Wilson T. Beraldo, and Gastão Rosenfeld at the University of São Paulo's Institute of Physiology. Their research focused on the proteolytic effects of snake venoms, particularly from the Bothrops jararaca species, which they incubated with canine plasma globulin. This interaction released a potent substance that caused slow, sustained contractions in isolated smooth muscle preparations, such as guinea pig ileum, and induced hypotension when injected into dogs.[79][80]The researchers named the compound "bradykinin," derived from the Greek words "bradys" (slow) and "kinin" (from "kinein," meaning to move), reflecting its characteristic slow onset of contraction in bioassays compared to other kinins like kallidin. Early characterization relied on these biological assays: contractions were measured on isolated rabbit uterus and guinea pig ileum, while systemic effects were assessed via blood pressure drops in anesthetized dogs, confirming its hypotensive and spasmogenic properties. The discovery was detailed in a seminal 1949 publication, which described bradykinin as a novel polypeptide released not only by snake venom but also by trypsin from plasma precursors.[79][81]Subsequent efforts in the 1950s focused on isolating bradykinin from both plasma and snake venom sources, yielding milligram quantities for further study. Its complete amino acid sequence—a nonapeptide (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg)—was determined in 1960 through enzymatic degradation and chromatographic analysis of ox blood-derived material by D.F. Elliott, G.P. Lewis, and E.W. Horton. This structural elucidation confirmed bradykinin's identity across species and laid the groundwork for synthetic production.
Key Developments
In the 1970s and 1980s, pharmacological studies characterized bradykinin receptors, distinguishing the constitutive B2 receptor from the inducible B1 receptor based on agonist selectivity and tissue distribution.[82] Concurrently, research elucidated key components of the kinin-kallikrein system, including the roles of plasma kallikrein in cleaving high-molecular-weight kininogen to generate bradykinin and the regulatory functions of C1 esterase inhibitor.[83] These advancements built on earlier discoveries, providing a framework for understanding bradykinin's vasodilatory and inflammatory effects across physiological systems.[84]The 1990s marked progress in therapeutic targeting, with the cloning of B1 and B2 receptor genes enabling detailed molecular studies—human B2 in 1992, murine B2 in 1993, and human B1 in 1994.[85][86] Early bradykinin antagonists, such as [D-Arg0, Hyp3, Thi5,8, D-Phe7]-bradykinin, emerged in the mid-1980s but saw refined development into more potent peptide analogs during the decade.[87] Additionally, the link between bradykinin accumulation and angiotensin-converting enzyme (ACE) inhibitor-induced cough was established, attributing the side effect to impaired bradykinin degradation sensitizing airway nerves.From the 2000s to 2010s, bradykinin's role in hereditary angioedema (HAE) was confirmed through direct measurement of elevated plasma levels during attacks, validating its mediation via unchecked kallikrein activity in C1 inhibitor-deficient patients. This paved the way for targeted therapies, culminating in the European Medicines Agency approval of icatibant, a selective B2 receptor antagonist, in 2008 for acute HAE treatment. No direct Nobel Prize has been awarded for kinin research, though foundational work on related vasoactive peptides influenced cardiovascular physiology awards.In the 2020s, a bradykinin storm hypothesis emerged for severe COVID-19 complications, proposing that SARS-CoV-2 disrupts ACE2 and des-Arg9-bradykinin signaling, leading to vascular leakage and fibrosis independent of cytokine storms. Ongoing research explores bradykinin's pro-tumorigenic effects, such as B1 receptor-driven macrophage polarization in tumor microenvironments and enhanced cancer cell migration via IL-6 induction.[88] Therapeutic advancements continued with approvals in 2025 of sebetralstat (oral kallikrein inhibitor), garadacimab (anti-FXIIa antibody), and donidalorsen (factor XII antisense) for HAE management, targeting bradykinin production. Deucrictibant, an oral B2 antagonist, advanced in clinical trials for both prophylactic and acute treatment of bradykinin-mediated angioedema as of 2025. However, novel B1/B2 modulators for non-HAE indications like cancer and inflammation remain limited, as highlighted in recent reviews.