MedPath

Acetylcholine Advanced Drug Monograph

Published:Aug 22, 2025

Generic Name

Acetylcholine

Brand Names

Miochol

Drug Type

Small Molecule

Chemical Formula

C7H16NO2

CAS Number

51-84-3

Acetylcholine (DB03128): A Comprehensive Monograph on its Physiology, Pharmacology, and Clinical Significance

Section 1: Introduction and Historical Perspective

1.1 Overview of Acetylcholine

Acetylcholine (ACh) holds a unique and foundational position in the annals of neuroscience and pharmacology. As the first neurotransmitter to be chemically identified, it serves as the archetypal molecule for understanding chemical communication within the nervous system.[1] It is a primary chemical messenger, a small molecule neurotransmitter that operates ubiquitously throughout both the central and peripheral nervous systems. Its physiological roles are vast and critical, mediating an expansive array of processes that range from the direct control of voluntary muscle contraction at the neuromuscular junction to the complex modulation of higher cognitive functions such as memory, learning, attention, and arousal within the brain.[3]

This monograph explores the central paradox of acetylcholine: despite its vital and widespread endogenous functions, its application as a therapeutic agent is exceptionally narrow and specialized. This limitation is a direct consequence of its pharmacokinetic profile, which is perfectly tuned for its role as a transient, local signaling molecule but renders it unsuitable for systemic administration. The very properties that make it an efficient neurotransmitter—specifically, its extremely rapid breakdown by cholinesterases—prevent it from achieving sustained, targeted effects when introduced as an exogenous drug.[1] This report will delve into the multifaceted nature of acetylcholine, examining its history, biochemistry, pharmacology, clinical applications, and role in pathology to provide a comprehensive understanding of this pivotal neurochemical.

1.2 The Discovery of Chemical Neurotransmission: A Paradigm Shift

The discovery of acetylcholine was not merely the identification of a new biological molecule; it was a revolutionary event that fundamentally altered the scientific understanding of how the nervous system functions. It provided the first concrete evidence for chemical neurotransmission, shifting the prevailing paradigm from a purely electrical model of nerve communication to a more nuanced electrochemical one. This conceptual leap laid the groundwork for the entire field of modern neuropharmacology.

The journey to this discovery began at the turn of the 20th century when scientific consensus held that nerve signals were conveyed primarily, if not exclusively, through direct electrical impulses.[8] The definitive challenge to this view came from the elegant and now-famous experiments conducted by the German pharmacologist Otto Loewi in 1921. As the story is often told, the idea for the experiment came to Loewi in a dream.[9] In his laboratory, he isolated two frog hearts, keeping them viable in separate chambers filled with a saline solution. The first heart had its vagus nerve intact, while the second was denervated. When Loewi electrically stimulated the vagus nerve of the first heart, its beat slowed, a known physiological response. The crucial step was what followed: he collected the perfusion fluid from this first heart and applied it to the second. The beat of the second, unconnected heart also slowed down, proving that a soluble chemical substance, released by the nerve stimulation, was responsible for mediating the effect.[2] Loewi initially named this unknown chemical messenger "Vagusstoff," meaning "Vagus substance" in German.[9]

Contemporaneously, the English physiologist Sir Henry Dale was conducting extensive research on the physiological effects of various compounds. As early as 1914, Dale had identified the powerful physiological actions of acetylcholine, noting its ability to mimic the effects of parasympathetic nerve stimulation.[8] Following Loewi's demonstration, the collaborative efforts of the scientific community, including Dale's own subsequent work, confirmed that Loewi's "Vagusstoff" was, in fact, acetylcholine. Dale's research further elucidated the role of acetylcholine as a neurotransmitter at numerous sites, including autonomic ganglia and the neuromuscular junction, solidifying its importance in the nervous system.[8]

The profound significance of their collective work was formally recognized in 1936 when Otto Loewi and Sir Henry Dale were jointly awarded the Nobel Prize in Physiology or Medicine.[2] Their discovery provided the first tangible proof that chemical agents could mediate physiological processes between neurons and their target cells. This established a new biological target for pharmacological intervention: the neurotransmitter receptor. If a naturally occurring chemical could produce a physiological effect by binding to a receptor, it followed that synthetic chemicals—drugs—could be designed to mimic, block, or otherwise modulate that effect. Consequently, the entire modern enterprise of developing pharmaceuticals that target neurotransmitter systems, including antidepressants, antipsychotics, and treatments for neurodegenerative diseases, has its conceptual roots in the discovery of acetylcholine.

Section 2: Physicochemical Properties and Molecular Identification

A comprehensive understanding of acetylcholine's biological function begins with its fundamental chemical and physical identity. As a small, quaternary ammonium compound, its structure dictates its behavior in biological systems, particularly its high polarity and charge. These characteristics are responsible for its excellent solubility in aqueous environments like the extracellular space and its inability to passively diffuse across lipid membranes, such as the blood-brain barrier.[1] This section consolidates the key identification and property data for acetylcholine from multiple chemical and pharmacological databases to provide a single, authoritative reference.

Identifier/PropertyValue / DescriptionSource(s)
Common NameAcetylcholine1
DrugBank IDDB031287
CAS Number51-84-31
FDA UNIIN9YNS0M02X1
SynonymsACh, Choline acetate, O-Acetylcholine, (2-acetoxyethyl)trimethylammonium, Ethanaminium, 2-(acetyloxy)-N,N,N-trimethyl-1
Chemical FormulaC7​H16​NO2​12
Molecular Weight146.209 g/mol13
Canonical SMILESO=C(OCCN+(C)C)C
InChIKeyOIPILFWXSMYKGL-UHFFFAOYSA-N
Melting Point85-87 °C
Boiling Point≈265.84°C (estimate)
SolubilityHighly soluble in water and ethanol; poorly soluble in ether
LogP-2.17
ATC CodeS01EB09

Section 3: Endogenous Acetylcholine: Synthesis, Release, and Inactivation

The lifecycle of acetylcholine within a cholinergic neuron is a tightly regulated and remarkably efficient process, optimized for the speed and precision required of a primary neurotransmitter. This cycle, encompassing synthesis, packaging, release, and rapid degradation, ensures the high fidelity of cholinergic signaling. The very efficiency of this system, however, is what makes exogenous acetylcholine a poor candidate for a systemic therapeutic drug.

3.1 Biosynthesis Pathway

The synthesis of acetylcholine occurs within the presynaptic terminal of cholinergic neurons and depends on two key precursors: choline and acetyl coenzyme A (acetyl-CoA).

The first and rate-limiting step in this pathway is the availability and uptake of choline. Choline is not synthesized de novo in neurons and must be obtained from the extracellular fluid. It is sourced from the diet, with rich sources including egg yolks, liver, and legumes, and is also produced endogenously by the liver. From the plasma, choline is transported across the blood-brain barrier and into the nerve terminal via a high-affinity, sodium-dependent choline transporter (CHT). A significant portion of the required choline is also derived from the recycling of choline produced after the breakdown of acetylcholine in the synaptic cleft.

The second precursor, acetyl-CoA, is generated within the neuron's mitochondria primarily through the metabolism of glucose. Once both precursors are present in the cytoplasm of the axon terminal, the enzyme choline acetyltransferase (CAT) catalyzes their condensation to form acetylcholine. The presence of CAT is considered the definitive biochemical marker for identifying a neuron as cholinergic.

3.2 Vesicular Packaging and Synaptic Release

Following its synthesis in the cytosol, acetylcholine is actively transported into synaptic vesicles. This packaging process is mediated by the vesicular acetylcholine transporter (VAChT), an antiporter that exchanges protons from inside the vesicle for cytosolic acetylcholine. This mechanism concentrates acetylcholine within the vesicles to high levels, protecting it from enzymatic degradation and preparing it for quantal release into the synapse.

The release of acetylcholine is a tightly controlled, calcium-dependent process known as exocytosis. The arrival of an action potential at the axon terminal causes depolarization of the presynaptic membrane, which triggers the opening of voltage-gated Ca2+ channels. The resulting rapid influx of calcium ions is the critical signal for vesicle fusion. This process is orchestrated by a complex of proteins known as SNAREs. The calcium sensor protein, synaptotagmin, located on the vesicle membrane, binds the incoming

Ca2+ and triggers the final fusion of the vesicle with the presynaptic membrane, releasing its contents into the synaptic cleft. This release mechanism is the target of several potent neurotoxins. Botulinum toxin, for example, prevents ACh release by cleaving SNARE proteins, leading to flaccid paralysis, whereas the venom of the black widow spider causes a massive, uncontrolled release of ACh, leading to severe muscle spasms and paralysis.

3.3 Enzymatic Degradation: The Key to Temporal Precision

Once released, acetylcholine diffuses across the synaptic cleft to bind with its receptors on the postsynaptic membrane. Its action is extremely brief, a necessity for ensuring discrete and rapid signaling. This temporal precision is achieved through its rapid inactivation by the enzyme acetylcholinesterase (AChE).

AChE is present at high concentrations in the synaptic cleft, where it hydrolyzes acetylcholine into its inactive constituents, choline and acetate, with remarkable efficiency. Each molecule of AChE can degrade approximately 25,000 molecules of acetylcholine per second, a rate that approaches the limit of diffusion. This near-instantaneous degradation prevents the neurotransmitter from lingering in the synapse, which would otherwise lead to prolonged receptor activation and a loss of signal fidelity. Much of the choline generated by this hydrolysis is then transported back into the presynaptic terminal for the synthesis of new acetylcholine, completing the cycle.

This entire lifecycle, optimized for precise, transient, local signaling, directly explains why systemic administration of acetylcholine as a drug is therapeutically unviable. If introduced into the bloodstream, it would be hydrolyzed by cholinesterases in the plasma and tissues long before it could reach and exert a sustained, meaningful effect on target organs. This inherent limitation has driven the development of two alternative therapeutic strategies: the use of more stable synthetic analogues (cholinergic agonists) that are resistant to hydrolysis, and the use of drugs that inhibit AChE (acetylcholinesterase inhibitors) to amplify the effects of endogenous acetylcholine.

Section 4: The Cholinergic Receptors: Gateways of Action

The diverse and profound physiological effects of acetylcholine are not inherent to the molecule itself but are determined by the specific receptors to which it binds. The cholinergic receptor system is broadly divided into two distinct superfamilies: nicotinic and muscarinic receptors. These two receptor classes differ fundamentally in their structure, signaling mechanisms, and physiological roles, and their differential distribution throughout the body is the key to understanding acetylcholine's pleiotropic actions.

4.1 Nicotinic Acetylcholine Receptors (nAChRs): Fast Ionotropic Transmission

Nicotinic receptors are members of the "cys-loop" superfamily of ligand-gated ion channels, responsible for fast, excitatory synaptic transmission.

Structure and Function: Structurally, nAChRs are pentameric proteins, meaning they are composed of five individual protein subunits that assemble symmetrically to form a central, water-filled pore that spans the cell membrane. The binding of two acetylcholine molecules to specific sites on the extracellular portion of the receptor induces a rapid conformational change, or "twist," in the subunits. This change opens the ion channel, allowing for the rapid influx of cations—primarily sodium (

Na+) and, in some cases, calcium (Ca2+)—down their electrochemical gradients. This influx of positive charge causes a rapid depolarization of the postsynaptic membrane, generating an excitatory postsynaptic potential (EPSP). This direct link between ligand binding and channel opening makes the nicotinic response extremely fast, occurring on a millisecond timescale.

Subtypes and Locations: Nicotinic receptors are broadly classified into two main subtypes based on their location and subunit composition:

  • Muscular (Nm) Receptors: These receptors are found exclusively at the neuromuscular junction, the specialized synapse between motor neurons and skeletal muscle fibers. Their activation by acetylcholine is the essential final step in the pathway for all voluntary muscle movement, triggering the depolarization that leads to muscle contraction.
  • Neuronal (Nn) Receptors: These receptors are more diverse and are widely distributed. They are found in the autonomic ganglia, where they mediate synaptic transmission for both the sympathetic and parasympathetic nervous systems. They are also located on the chromaffin cells of the adrenal medulla, where their activation triggers the release of adrenaline and norepinephrine into the bloodstream. Within the central nervous system, Nn receptors are found in numerous brain regions and are involved in a wide range of functions, including cognitive processes and neurotransmitter release.

4.2 Muscarinic Acetylcholine Receptors (mAChRs): Slower, Modulatory Metabotropic Action

In contrast to the fast, direct action of nicotinic receptors, muscarinic receptors mediate slower, longer-lasting, and more modulatory responses. They belong to the superfamily of G-protein coupled receptors (GPCRs).

Structure and Function: Muscarinic receptors are single polypeptide chains that traverse the cell membrane seven times. Instead of forming an ion channel themselves, they are coupled to intracellular signaling proteins known as G-proteins. When acetylcholine binds to a muscarinic receptor, it causes a conformational change that activates the associated G-protein. The activated G-protein then initiates a downstream cascade of intracellular second messengers, which ultimately leads to the cellular response, such as the opening or closing of separate ion channels or changes in enzyme activity. This indirect mechanism of action results in a response that is significantly slower in onset and longer in duration compared to that of nicotinic receptors.

Subtypes and Signaling: There are five distinct subtypes of muscarinic receptors, designated M1 through M5. They are often grouped based on the type of G-protein they couple to and their resulting signaling pathway :

  • M1, M3, and M5 Receptors: These subtypes typically couple to G-proteins of the Gq​/11 family. Activation of this pathway stimulates the enzyme phospholipase C (PLC), which in turn hydrolyzes a membrane lipid to produce two second messengers: inositol triphosphate (IP3​) and diacylglycerol (DAG). IP3​ triggers the release of Ca2+ from intracellular stores, while DAG activates protein kinase C (PKC). These pathways are generally considered excitatory.
  • M2 and M4 Receptors: These subtypes couple to G-proteins of the Gi​/o family. Activation of this pathway inhibits the enzyme adenylyl cyclase, leading to a decrease in intracellular levels of cyclic AMP (cAMP). Additionally, the dissociated βγ subunit of the G-protein can directly activate certain types of potassium (K+) channels, leading to an efflux of potassium and hyperpolarization of the cell membrane. These pathways are generally considered inhibitory.

Locations: Muscarinic receptors are widely distributed throughout the body, mediating the majority of acetylcholine's effects in the parasympathetic nervous system and the CNS. M1 receptors are prominent in the CNS and secretory glands; M2 receptors are the primary subtype in the heart; M3 receptors are found in smooth muscle and glands; and M4 and M5 receptors are located primarily within the CNS.

4.3 Receptor Binding Affinity and Modulation

The interaction between acetylcholine and its receptors is a dynamic process influenced by the precise molecular architecture of the binding site. The affinity of a receptor for acetylcholine—a measure of how tightly it binds—is determined by specific amino acid residues within the binding pocket, with aromatic residues like tyrosine and tryptophan playing a particularly important role. Different receptor subtypes exhibit distinct affinities for acetylcholine and other cholinergic ligands. For instance, certain neuronal nAChR subtypes have a higher affinity for nicotine than for acetylcholine itself. Furthermore, the affinity of a receptor for its agonist is not static; it can change as the receptor transitions between its resting (closed) and active (open) conformational states, a fundamental principle that underlies the efficiency of signal transduction. This differential affinity across receptor subtypes is a key focus of pharmacological research, as it allows for the development of subtype-selective drugs that can target specific physiological processes while minimizing off-target effects.

Section 5: System-Wide Pharmacodynamics

The widespread distribution of the various cholinergic receptor subtypes results in acetylcholine exerting a vast and complex array of physiological effects across virtually every organ system. The overall effect of cholinergic stimulation is a manifestation of the integrated responses of these different receptors. A core principle that emerges is that the action of acetylcholine is entirely context-dependent, determined not by the molecule itself, but by the specific receptor and tissue it engages. This explains how the same neurotransmitter can be excitatory in one location (e.g., skeletal muscle) and inhibitory in another (e.g., the heart).

Central Nervous System: Within the CNS, acetylcholine acts as a critical neuromodulator, influencing the activity of broad neuronal networks. Cholinergic neurons originating in key areas like the basal forebrain (specifically the nucleus basalis of Meynert) and the brainstem project throughout the cortex and limbic structures. Through a complex interplay of nicotinic and muscarinic receptor activation, acetylcholine plays an indispensable role in higher cognitive functions, including learning, memory formation and retrieval, attention, and arousal. It is also instrumental in promoting and maintaining rapid eye movement (REM) sleep.

Peripheral Nervous System:

  • Somatic Nervous System: Acetylcholine's role here is singular and absolute. It is the sole neurotransmitter released by motor neurons at the neuromuscular junction to activate nicotinic (Nm) receptors on skeletal muscle fibers. As such, it is essential for initiating all forms of voluntary movement.
  • Autonomic Nervous System: Acetylcholine is a key player in the autonomic nervous system, which regulates involuntary bodily functions. It serves as the neurotransmitter at all preganglionic synapses (in both the sympathetic and parasympathetic divisions) and at all postganglionic effector junctions of the parasympathetic nervous system. This makes it the dominant neurotransmitter of the parasympathetic, or "rest-and-digest," system.

Cardiovascular System: The cardiovascular effects of acetylcholine are primarily mediated by M2 muscarinic receptors located in the heart. Vagal nerve stimulation releases acetylcholine, which acts on the sinoatrial (SA) and atrioventricular (AV) nodes. This leads to a decrease in heart rate (negative chronotropy), a reduction in the force of atrial contraction (negative inotropy), and a slowing of the electrical conduction velocity through the AV node (negative dromotropy). Acetylcholine also promotes generalized vasodilation, contributing to a decrease in blood pressure.

Respiratory System: In the respiratory tract, activation of M3 receptors on the smooth muscle of the bronchioles leads to bronchoconstriction (narrowing of the airways). Stimulation of M3 receptors on glandular cells also increases the secretion of tracheobronchial mucus.

Gastrointestinal System: Acetylcholine is a major stimulator of the digestive system. Through M3 receptor activation, it increases the tone and peristaltic contractions of the stomach and intestines, propelling food through the GI tract. It also stimulates the secretion of saliva and digestive juices and promotes the relaxation of gastrointestinal sphincters.

Genitourinary System: In the urinary system, acetylcholine acting on M3 receptors causes contraction of the detrusor muscle in the bladder wall and relaxation of the trigone and sphincter muscles, thereby increasing voiding pressure and promoting urination. In the male reproductive system, it is involved in mediating penile erection.

Exocrine Glands: Acetylcholine is a potent secretagogue, stimulating secretion from all glands that receive parasympathetic innervation. This includes the stimulation of tears from lacrimal glands, saliva from salivary glands, sweat from exocrine sweat glands, and various digestive secretions, primarily through M1 and M3 receptor activation.

Ocular System: In the eye, acetylcholine produces two key effects via M3 receptor activation. It contracts the sphincter pupillae muscle of the iris, causing the pupil to constrict (miosis). It also contracts the ciliary muscle, which relaxes the zonular fibers and allows the lens to become more convex, a process known as accommodation, which is necessary for focusing on near objects.

Section 6: Clinical Application in Ophthalmic Surgery

Despite its ubiquitous physiological importance, the therapeutic use of acetylcholine as an administered drug is confined to a single, highly specialized application: as an intraocular solution to induce rapid miosis during ophthalmic surgery. This clinical niche exists precisely because of the pharmacokinetic properties that make acetylcholine unsuitable for any other use. Its inherent instability and rapid local metabolism, which preclude systemic administration, become advantageous in the controlled environment of the eye, providing a powerful, transient, and localized effect with minimal systemic risk.

6.1 Indication and Therapeutic Rationale

The commercially available formulation of acetylcholine for ophthalmic use is Miochol-E (acetylcholine chloride). Its sole indication is to obtain rapid and profound miosis (constriction of the pupil) in seconds during anterior segment surgical procedures. These procedures include cataract extraction (specifically, after the artificial intraocular lens has been delivered), penetrating keratoplasty (corneal transplant), and iridectomy.

The therapeutic rationale for inducing miosis in these settings is multifaceted. A rapidly constricted pupil provides immediate protection for the delicate vitreous face after the natural lens is removed, reducing the risk of vitreous loss. It also facilitates the placement of corneo-scleral sutures by pulling the iris away from the surgical site, thereby reducing the hazard of accidental iris incarceration by the sutures or instruments.

6.2 Dosage, Administration, and Pharmacokinetics

Preparation and Dosage: Miochol-E is supplied as a two-part system: a vial containing 20 mg of lyophilized acetylcholine chloride powder with mannitol as an excipient, and an ampoule containing 2 mL of a sterile electrolyte diluent. The two components must be aseptically combined immediately before use to reconstitute a 1% (10 mg/mL) isotonic solution. This requirement for immediate reconstitution is critical because aqueous solutions of acetylcholine are highly unstable and degrade rapidly. The typical adult dose is 0.5 to 2 mL of the reconstituted solution.

Administration: The solution is administered via intraocular instillation. Using a suitable atraumatic cannula, the surgeon gently irrigates the solution into the anterior chamber of the eye. The instillation should be gentle and directed parallel to the face of the iris and tangential to the pupillary border to avoid trauma to ocular structures.

Pharmacokinetics and Onset of Action: When applied directly to the iris in the anterior chamber, acetylcholine acts on muscarinic receptors on the pupillary sphincter muscle, causing constriction to begin within seconds. The miotic effect is potent but short-lived, typically persisting for only about 10 minutes. This transient action is due to the rapid hydrolysis of acetylcholine by acetylcholinesterase, which is present in ocular tissues. This rapid local metabolism forms a "pharmacokinetic firewall," confining the drug's potent effect to the target organ and preventing significant amounts from entering systemic circulation. In contrast, topical application of acetylcholine to the intact eye (as eye drops) is completely ineffective, as cholinesterase in the cornea and tear film destroys the molecule far more rapidly than it can penetrate into the anterior chamber.

6.3 Safety Profile and Adverse Events

The safety profile of intraocular acetylcholine is generally favorable, a direct result of its localized action and rapid inactivation.

Ocular Adverse Events: The most frequently reported adverse effects are localized to the eye itself. These include transient corneal edema (swelling), corneal clouding, and, infrequently, corneal decompensation, which may affect vision post-operatively.

Systemic Adverse Events: Systemic absorption is minimal and adverse events related to it are rare. However, if significant absorption were to occur, the effects would be predictable extensions of acetylcholine's systemic pharmacology. These include bradycardia (slow heart rate), hypotension (low blood pressure), flushing, breathing difficulties, and sweating.

Contraindications and Precautions: The primary contraindication is a known hypersensitivity to acetylcholine chloride or any other component of the formulation. Caution is warranted when using the drug in patients with pre-existing conditions that could be dangerously exacerbated by systemic cholinergic stimulation, such as severe asthma, acute heart failure, peptic ulcer disease, or urinary tract obstruction, even though the risk of systemic effects is low. For the drug to be effective, any anatomical hindrances to miosis, such as synechiae (adhesions), must be surgically released prior to administration.

Overdose Management: In the unlikely event of an overdose leading to significant systemic effects, specific antidotes should be readily available. Atropine sulfate (0.5 to 1 mg), a competitive muscarinic antagonist, can be administered intramuscularly or intravenously to counteract the muscarinic symptoms. Epinephrine may also be used to overcome severe cardiovascular or bronchoconstrictor responses.

Section 7: Acetylcholine in Neurological Disease

Dysfunction of the cholinergic system is a central pathological feature in several of the most significant neurological disorders affecting humanity. The specific nature of the cholinergic disruption—whether it is a deficiency of the neurotransmitter, a blockade of its receptors, or a relative excess due to imbalance with another system—defines the disease's clinical presentation and dictates the therapeutic strategy. The study of acetylcholine's role in Alzheimer's disease, myasthenia gravis, and Parkinson's disease provides a powerful illustration of clinical neuropharmacology.

7.1 Alzheimer's Disease: The Cholinergic Hypothesis

The "cholinergic hypothesis" was one of the earliest and most enduring theories proposed to explain the cognitive symptoms of Alzheimer's disease. This hypothesis posits that a significant portion of the cognitive decline seen in the disease is a direct result of a deficiency in cholinergic neurotransmission.

Pathophysiology: Post-mortem studies of brains from patients with Alzheimer's disease consistently reveal a profound and selective loss of cholinergic neurons. The most severe degeneration occurs in the nucleus basalis of Meynert, a region in the basal forebrain that provides the primary cholinergic innervation to the entire cerebral cortex and hippocampus. These cortical and hippocampal regions are critical for memory, learning, and attention. The loss of these neurons leads to a dramatic reduction in the levels of both acetylcholine and its synthesizing enzyme, choline acetyltransferase (CAT), in these target areas. This cholinergic deficit is strongly correlated with the severity of dementia. In addition to the loss of presynaptic neurons, a significant reduction in the number of nicotinic acetylcholine receptors in the brain has also been reported.

Therapeutic Strategy: As a direct consequence of this understanding, the primary pharmacological strategy for the symptomatic treatment of mild to moderate Alzheimer's disease is to enhance the function of the remaining cholinergic neurons. This is achieved through the use of acetylcholinesterase inhibitors (AChEIs), such as donepezil, rivastigmine, and galantamine. These drugs work by reversibly inhibiting the AChE enzyme, thereby slowing the breakdown of acetylcholine in the synaptic cleft. This increases the concentration and prolongs the action of the acetylcholine that is still being released, amplifying the diminished cholinergic signal and leading to modest but clinically meaningful improvements in cognitive function for some patients.

7.2 Myasthenia Gravis: An Autoimmune Attack on the Neuromuscular Junction

Myasthenia gravis (MG) is the archetypal autoimmune disease of the neuromuscular junction, characterized by a direct assault on the postsynaptic components of the cholinergic system.

Pathophysiology: In MG, the immune system mistakenly produces antibodies that target and attack the nicotinic (Nm) acetylcholine receptors on the surface of skeletal muscle fibers. These antibodies disrupt neuromuscular transmission in three ways: by directly blocking the acetylcholine binding site, by accelerating the internalization and degradation of the receptors, and by damaging the postsynaptic membrane via complement activation. The net result is a marked reduction in the number of functional ACh receptors at the neuromuscular junction. Consequently, the end-plate potentials generated by acetylcholine release are often too small to trigger a muscle action potential, leading to a failure of muscle contraction. This manifests clinically as fluctuating and fatigable weakness of voluntary muscles.

Therapeutic Strategy: The therapeutic approach is aimed at improving the efficiency of neuromuscular transmission. The first-line symptomatic treatment for MG is the administration of AChE inhibitors, most commonly pyridostigmine. By inhibiting the breakdown of acetylcholine, these drugs increase the amount and duration of the neurotransmitter in the synaptic cleft. This increased concentration enhances the probability that acetylcholine molecules will find and activate the few remaining functional receptors, thereby improving muscle strength and reducing fatigue. This is often combined with immunosuppressive therapies to target the underlying autoimmune cause.

7.3 Parkinson's Disease: A Dopamine-Acetylcholine Imbalance

While Parkinson's disease is primarily defined by the loss of dopamine-producing neurons, the cholinergic system plays a critical and opposing role in the pathophysiology of its motor symptoms.

Pathophysiology: The cardinal motor symptoms of Parkinson's disease—tremor, rigidity, and bradykinesia—arise from the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta. These neurons project to the striatum, a key brain region for motor control. Within the striatum, there is a delicate functional balance between the actions of dopamine (which is largely inhibitory) and acetylcholine (which is largely excitatory, released by local interneurons). The profound loss of dopamine in Parkinson's disease disrupts this balance, leading to a state of relative cholinergic overactivity or dominance. This unopposed cholinergic influence is thought to contribute significantly to the motor symptoms, particularly tremor and rigidity.

Therapeutic Strategy: Based on this model of imbalance, one of the earliest pharmacological treatments for Parkinson's disease involved the use of anticholinergic drugs (muscarinic receptor antagonists), such as trihexyphenidyl and benztropine. By blocking the action of acetylcholine in the striatum, these drugs help to restore the functional balance between the dopaminergic and cholinergic systems. While they are less effective for bradykinesia, they can be particularly useful in alleviating the resting tremor associated with the disease.

Section 8: Toxicology: The Cholinergic Crisis

A cholinergic crisis is a medical emergency that arises from the excessive, widespread stimulation of cholinergic receptors throughout the body. This condition, also known as cholinergic toxidrome, represents an extreme exaggeration of acetylcholine's normal physiological effects and can be life-threatening if not promptly recognized and treated. Understanding the normal pharmacodynamics of acetylcholine is the key to predicting, identifying, and rationally managing every symptom of this toxic state.

8.1 Etiology and Pathophysiology

Cholinergic toxicity is caused by an overabundance of acetylcholine at its receptors. While this can be caused by overdose of direct-acting cholinergic agonists, the most common and severe cases result from poisoning with substances that inhibit the acetylcholinesterase (AChE) enzyme. By blocking the primary mechanism of acetylcholine degradation, these inhibitors cause a massive accumulation of the neurotransmitter at all cholinergic synapses—muscarinic and nicotinic, in both the central and peripheral nervous systems. The most common sources of AChE inhibitors are organophosphate and carbamate insecticides, which are a major cause of poisoning worldwide, and highly potent chemical warfare nerve agents like sarin and VX.

8.2 Clinical Manifestations (The Toxidrome)

The signs and symptoms of a cholinergic crisis are a direct and predictable consequence of receptor overstimulation and are typically divided into muscarinic, nicotinic, and central effects.

Muscarinic Effects: Overstimulation of muscarinic receptors on parasympathetic end-organs produces a classic set of symptoms. These are often remembered by the mnemonics DUMBELS or SLUDGEM :

  • Diaphoresis (sweating) / Diarrhea
  • Urination (incontinence)
  • Miosis (pinpoint pupils)
  • Bronchospasm and Bronchorrhea (excessive bronchial secretions)
  • Emesis (vomiting)
  • Lacrimation (tearing)
  • Salivation (excessive drooling)

The combination of bronchospasm and massive fluid secretion into the airways (bronchorrhea) is particularly dangerous and is a primary cause of respiratory failure. Other major muscarinic effects include severe bradycardia and hypotension.

Nicotinic Effects: Overstimulation of nicotinic receptors has effects at the autonomic ganglia and, most critically, at the neuromuscular junction. This leads to initial muscle fasciculations (involuntary twitching) followed by profound muscle weakness and ultimately flaccid paralysis. Paralysis of the diaphragm and other respiratory muscles is a major contributor to death from cholinergic poisoning.

Central Nervous System (CNS) Effects: Because acetylcholine is also a neurotransmitter in the brain, its excess can cross the blood-brain barrier (in the case of lipid-soluble poisons) and cause a range of CNS effects. These can include anxiety, confusion, headache, and drowsiness, progressing in severe cases to convulsions (seizures), coma, and central respiratory depression.

Death in a cholinergic crisis is typically due to respiratory failure, resulting from the lethal combination of central respiratory depression, bronchoconstriction, overwhelming airway secretions, and paralysis of the respiratory muscles.

8.3 Principles of Management

The management of a cholinergic crisis is a medical emergency that focuses on three main principles: supportive care, decontamination, and the administration of specific pharmacological antidotes.

  1. Supportive Care: The immediate priority is the "ABCs" of resuscitation: maintaining a patent Airway, ensuring adequate Breathing, and supporting Circulation. Aggressive suctioning of the airway is often required to clear the copious secretions. Patients with respiratory distress or paralysis will require endotracheal intubation and mechanical ventilation.
  2. Decontamination: In cases of topical exposure (e.g., from pesticides), it is critical to remove all of the patient's clothing and thoroughly wash their skin and eyes to prevent further absorption of the toxin. Healthcare providers must wear appropriate personal protective equipment to avoid becoming contaminated themselves.
  3. Pharmacological Antidotes: Two primary antidotes are used, each targeting a different aspect of the poisoning:
  • Atropine: This drug is a competitive antagonist at muscarinic receptors. It is given in large and repeated intravenous doses to block the life-threatening muscarinic effects, particularly the excessive airway secretions and severe bradycardia. Atropine does not affect the nicotinic symptoms (muscle weakness).
  • Pralidoxime (2-PAM) or other Oximes: These drugs are cholinesterase reactivators. They work by binding to the organophosphate-inhibited AChE enzyme and removing the phosphate group, thereby restoring the enzyme's function. This is particularly important for reversing the nicotinic effects, especially the paralysis of respiratory muscles. Oximes must be administered early after exposure to be effective.
  • Benzodiazepines: Drugs like diazepam are used to control seizures and reduce the neuronal damage associated with them.

Section 9: Investigational and Diagnostic Frontiers

While the therapeutic application of acetylcholine is limited, its use in clinical research and as a sophisticated diagnostic agent is expanding. By leveraging its well-understood physiological effects, clinicians and researchers can use acetylcholine not as a treatment, but as a pharmacological probe to unmask underlying pathology or investigate physiological processes.

9.1 Diagnostic Use in Cardiology: The Acetylcholine Provocation Test

A prominent and increasingly recognized application of acetylcholine is in the diagnostic evaluation of coronary artery function. The intracoronary acetylcholine provocation test is a specialized procedure used to identify coronary vasomotor disorders, such as vasospastic angina and coronary microvascular dysfunction, in patients who present with symptoms of myocardial ischemia (e.g., chest pain) but have non-obstructive coronary arteries on standard angiography (a condition known as INOCA or MINOCA).

Rationale and Procedure: The test is based on the differential response of healthy versus dysfunctional coronary arteries to acetylcholine. In a healthy coronary artery, acetylcholine binds to muscarinic receptors on the endothelial cells, stimulating the release of nitric oxide (NO), a potent vasodilator. This leads to relaxation of the underlying smooth muscle and an increase in coronary blood flow. However, in patients with endothelial dysfunction, this NO-mediated vasodilation is impaired. Instead, acetylcholine can act directly on muscarinic receptors on the coronary smooth muscle cells, causing paradoxical vasoconstriction. During the procedure, which is performed in a cardiac catheterization lab, incremental doses of acetylcholine are infused directly into the left and right coronary arteries while coronary angiography and ECG are continuously monitored. A positive test for epicardial spasm is defined as a significant focal or diffuse narrowing of a major coronary artery accompanied by the patient's typical chest pain and ischemic ECG changes. A positive test for microvascular spasm is diagnosed when the patient experiences chest pain and ECG changes without significant epicardial constriction.

Safety and Efficacy: Despite initial concerns about safety, multiple large-scale studies and a meta-analysis involving over 70,000 patients have established that the acetylcholine provocation test is a safe procedure when performed by experienced operators. The rate of major complications (e.g., fatal events, shock, sustained ventricular arrhythmias) is exceedingly low, reported at 0% to 0.5%. Minor, transient complications are more common (occurring in about 9% of patients in one study) but are generally manageable. These include transient bradycardia or atrioventricular block (especially during right coronary artery infusion), which typically resolves quickly, and paroxysmal atrial fibrillation. The test has significant diagnostic and prognostic value. It can definitively identify a cause for symptoms in a large proportion of INOCA/MINOCA patients and, importantly, a positive test is an independent predictor of future major adverse cardiovascular events, allowing for targeted therapy and risk stratification.

9.2 Other Areas of Clinical Research

Clinical trial registries indicate that acetylcholine has been employed as an investigational tool in a variety of other research contexts, further highlighting its utility as a pharmacological probe. While these do not represent approved clinical uses, they demonstrate the breadth of research into cholinergic mechanisms.

  • Type 1 Diabetes Mellitus: Acetylcholine was used in a completed clinical trial (NCT01097551) designed to investigate endothelial dysfunction, a common complication of diabetes. Its vasodilatory properties were likely used to assess the functional integrity of the vascular endothelium in these patients.
  • Sickle Cell Anemia: In a completed Phase 2 trial (NCT00009581), acetylcholine was included in a study focused on nitric oxide pathways to improve blood flow in patients with sickle cell disease. This again points to its use in assessing vascular reactivity.
  • Chronic Kidney Disease: A completed trial (NCT02987465) used acetylcholine to help evaluate the effects of standard erythropoiesis-stimulating agents on forearm blood flow in patients with anemia associated with chronic kidney disease, another setting where vascular function is often compromised.

In these diverse research settings, acetylcholine is not being investigated as a potential therapy. Instead, its well-characterized effect on the vascular endothelium is being harnessed as a standardized challenge to measure and quantify the health or dysfunction of the cardiovascular system in various disease states.

Section 10: Conclusion and Future Directions

Acetylcholine, the "Vagusstoff" of Otto Loewi's foundational experiments, remains a molecule of immense scientific and clinical importance more than a century after its discovery. This monograph has detailed its multifaceted nature, tracing its journey from a historical curiosity to the cornerstone of our modern understanding of chemical neurotransmission. Its discovery not only revealed the mechanism of nerve-to-cell communication but also gave birth to the field of neuropharmacology, providing the first rational target for drugs designed to modulate the nervous system.

A central theme that has emerged is the profound duality of acetylcholine's character. It is an indispensable endogenous messenger, with a lifecycle perfectly optimized for the rapid, precise, and localized signaling required for everything from muscle contraction to conscious thought. Yet, this very efficiency—specifically its near-instantaneous enzymatic degradation—is its greatest liability as a systemic therapeutic agent. This inherent pharmacokinetic limitation has confined its direct clinical use to the highly specialized niche of intraocular administration, where its instability becomes a key safety feature, creating a "pharmacokinetic firewall" that localizes its potent effects and minimizes systemic toxicity.

The clinical significance of acetylcholine extends far beyond its limited use as a drug. Dysfunction within the cholinergic system lies at the heart of several major neurological disorders. The cholinergic deficits in Alzheimer's disease, the autoimmune attack on nicotinic receptors in myasthenia gravis, and the relative cholinergic overactivity in Parkinson's disease have driven the development of entire classes of modern pharmaceuticals—the acetylcholinesterase inhibitors and anticholinergic agents—that indirectly manipulate the cholinergic system to provide critical symptomatic relief for millions of patients.

Looking forward, the story of acetylcholine continues to evolve. While its direct therapeutic potential remains limited, research into more stable and receptor-subtype-selective cholinergic agonists and antagonists continues, holding promise for more targeted treatments for a range of conditions. Perhaps most compelling is the expanding role of acetylcholine itself as a sophisticated diagnostic tool. Its use in the intracoronary provocation test exemplifies a clever repurposing of basic pharmacology, using the molecule's physiological effects not for treatment, but as a precise probe to unmask hidden pathology. This ensures that acetylcholine, the first neurotransmitter, will remain at the forefront of both fundamental neuroscience and innovative clinical practice for the foreseeable future.

Works cited

  1. Acetylcholine | 51-84-3 - ChemicalBook, accessed August 22, 2025, https://www.chemicalbook.com/ChemicalProductProperty_EN_CB0895802.htm
  2. 3.5: Neurotransmitters- Acetylcholine - Medicine LibreTexts, accessed August 22, 2025, https://med.libretexts.org/Sandboxes/admin/Introduction_to_Neuroscience_(Hedges)/03%3A_Neuronal_Communication/3.05%3A_Neurotransmitters-_Acetylcholine
  3. Physiology, Acetylcholine - StatPearls - NCBI Bookshelf, accessed August 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK557825/
  4. Acetylcholine (ACh): What It Is, Function & Deficiency - Cleveland Clinic, accessed August 22, 2025, https://my.clevelandclinic.org/health/articles/24568-acetylcholine-ach
  5. How Acetylcholine Functions in Your Body - Verywell Mind, accessed August 22, 2025, https://www.verywellmind.com/what-is-acetylcholine-2794810
  6. Acetylcholine | Definition, Function, & Facts | Britannica, accessed August 22, 2025, https://www.britannica.com/science/acetylcholine
  7. Acetylcholine: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed August 22, 2025, https://go.drugbank.com/drugs/DB03128
  8. Sir Henry Dale – Facts - NobelPrize.org, accessed August 22, 2025, https://www.nobelprize.org/prizes/medicine/1936/dale/facts/
  9. Otto Loewi - Wikipedia, accessed August 22, 2025, https://en.wikipedia.org/wiki/Otto_Loewi
  10. Otto Loewi (1873–1961): Dreamer and Nobel laureate - PMC, accessed August 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4291908/
  11. Henry Dale and the discovery of acetylcholine - PubMed, accessed August 22, 2025, https://pubmed.ncbi.nlm.nih.gov/16731499/
  12. Acetylcholine - CAS Common Chemistry, accessed August 22, 2025, https://commonchemistry.cas.org/detail?cas_rn=51-84-3
  13. Acetylcholine - SIELC Technologies, accessed August 22, 2025, https://sielc.com/compound-acetylcholine
  14. Acetylcholine Neurotransmission (Section 1, Chapter 11) Neuroscience Online, accessed August 22, 2025, https://nba.uth.tmc.edu/neuroscience/m/s1/chapter11.html
  15. Miochol™-E (acetylcholine chloride intraocular solution) 20 mg/2 mL (10 mg/mL) Sterile 1:100 with Electrolyte Diluent FOR INT - Bausch + Lomb, accessed August 22, 2025, https://www.bausch.com/globalassets/pdf/packageinserts/pharma/miochol-e-acetylcholine-chloride-intraocular-solution.pdf
  16. Miochol-E: Package Insert / Prescribing Information - Drugs.com, accessed August 22, 2025, https://www.drugs.com/pro/miochol-e.html
  17. miochol-e (acetylcholine chloride) powder for solution for intraocular irrigation 1 name of the medicine - Medsinfo, accessed August 22, 2025, https://medsinfo.com.au/api/documents/Miochol-E_PI?format=pdf
  18. Acetylcholinesterase Inhibitors: Pharmacology and Toxicology - PMC - PubMed Central, accessed August 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3648782/
  19. Acetylcholine receptor - Wikipedia, accessed August 22, 2025, https://en.wikipedia.org/wiki/Acetylcholine_receptor
  20. Nicotinic acetylcholine receptor - Wikipedia, accessed August 22, 2025, https://en.wikipedia.org/wiki/Nicotinic_acetylcholine_receptor
  21. www.ncbi.nlm.nih.gov, accessed August 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK557825/#:~:text=In%20the%20somatic%20nervous%20system,neurons%20and%20affecting%20voluntary%20movements.
  22. High-affinity Binding of [3H]Acetylcholine to Muscarinic Cholinergic Receptors' - Journal of Neuroscience, accessed August 22, 2025, https://www.jneurosci.org/content/jneuro/5/6/1577.full.pdf
  23. The α-bungarotoxin binding site on the nicotinic acetylcholine receptor: Analysis using a phage–epitope library | PNAS, accessed August 22, 2025, https://www.pnas.org/doi/10.1073/pnas.94.12.6054
  24. Functional differences between neurotransmitter binding sites of muscle acetylcholine receptors | PNAS, accessed August 22, 2025, https://www.pnas.org/doi/10.1073/pnas.1414378111
  25. Action of nicotine and analogs on acetylcholine receptors having mutations of transmitter-binding site residue αG153, accessed August 22, 2025, https://rupress.org/jgp/article/141/1/95/43197/Action-of-nicotine-and-analogs-on-acetylcholine
  26. Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior - PMC - PubMed Central, accessed August 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3466476/
  27. Miochol®-E - Bausch + Lomb, accessed August 22, 2025, https://www.bausch.com/products/rx-pharmaceuticals/other-rx/
  28. Miochol-E Drug / Medicine Information - News-Medical, accessed August 22, 2025, https://www.news-medical.net/drugs/Miochol-E.aspx
  29. Acetylcholine Ophthalmic Dosage Guide + Max Dose, Adjustments ..., accessed August 22, 2025, https://www.drugs.com/dosage/acetylcholine-ophthalmic.html
  30. ACETYLCHOLINE IN CATARACT SURGERY*, accessed August 22, 2025, https://bjo.bmj.com/content/bjophthalmol/50/7/429.full.pdf
  31. NDA 20-213/S-010 Page 3 MIOCHOL -E (acetylcholine chloride intraocular solution) 1:100 with Electrolyte Diluent Prescribing Info - accessdata.fda.gov, accessed August 22, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2006/020213s010lbl.pdf
  32. Miochol E, (acetylcholine) dosing, indications, interactions, adverse ..., accessed August 22, 2025, https://reference.medscape.com/drug/miochol-e-acetylcholine-343661
  33. Acetylcholine (Professional Patient Advice) - Drugs.com, accessed August 22, 2025, https://www.drugs.com/ppa/acetylcholine.html
  34. Acetylcholine: Eye Surgery Uses, Warnings, Side Effects, Dosage - MedicineNet, accessed August 22, 2025, https://www.medicinenet.com/acetylcholine/article.htm
  35. acetylcholine chloride intraocular solution - medtigo, accessed August 22, 2025, https://medtigo.com/drug/acetylcholine-chloride-intraocular-solution/
  36. bausch+lomb, accessed August 22, 2025, https://mellanarkiv-offentlig.vgregion.se/alfresco/s/archive/stream/public/v1/source/available/sofia/rhs8136-160155217-365/native/Bipacksedel%20product-information-miochol-canada%20747236.pdf
  37. Acetylcholine: Side Effects, Uses, Dosage, Interactions, Warnings, accessed August 22, 2025, https://www.rxlist.com/acetylcholine/generic-drug.htm
  38. Miochol-E - NPS MedicineWise, accessed August 22, 2025, https://www.nps.org.au/medicine-finder/miochol-e-powder-for-intraocular-irrigation
  39. Acetylcholine: What it is, function, and links with health, accessed August 22, 2025, https://www.medicalnewstoday.com/articles/326638
  40. A Systematic Review on Drugs Acting as Nicotinic Acetylcholine Receptor Agonists in the Treatment of Dementia - MDPI, accessed August 22, 2025, https://www.mdpi.com/2073-4409/13/3/237
  41. Acetylcholinesterase inhibitor - Wikipedia, accessed August 22, 2025, https://en.wikipedia.org/wiki/Acetylcholinesterase_inhibitor
  42. www.ncbi.nlm.nih.gov, accessed August 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK557825/#:~:text=Acetylcholine%20(ACh)%20is%20clinically%20significant,and%20myasthenia%20gravis%20(MG).&text=MG%20is%20an%20autoimmune%20disorder,skeletal%20muscles%20after%20repeated%20use.
  43. Cholinergic Crisis - StatPearls - NCBI Bookshelf, accessed August 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK482433/
  44. Cholinergic Toxicity - StatPearls - NCBI Bookshelf, accessed August 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK539783/
  45. www.ncbi.nlm.nih.gov, accessed August 22, 2025, https://www.ncbi.nlm.nih.gov/books/NBK539783/#:~:text=Excess%20acetylcholine%20in%20the%20brain,%2C%20coma%2C%20and%20respiratory%20depression.
  46. Acetylcholine Use in Modern Cardiac Catheterization Laboratories: A Systematic Review - PMC - PubMed Central, accessed August 22, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8880288/
  47. Safety and prognostic relevance of acetylcholine testing in patients with stable myocardial ischaemia or myocardial infarction and non-obstructive coronary arteries | EuroIntervention, accessed August 22, 2025, https://eurointervention.pcronline.com/article/safety-and-prognostic-relevance-of-acetylcholine-testing-in-patients-with-stable-myocardial-ischaemia-or-myocardial-infarction-and-non-obstructive-coronary-arteries
  48. Clinical Usefulness, Angiographic Characteristics, and Safety Evaluation of Intracoronary Acetylcholine Provocation Testing Among 921 Consecutive White Patients With Unobstructed Coronary Arteries | Circulation - American Heart Association Journals, accessed August 22, 2025, https://www.ahajournals.org/doi/10.1161/circulationaha.113.004096
  49. Definition of Investigational Medicinal Products (IMPs) and use of Auxiliary Medicinal Products (AMPs) - Public Health - European Commission, accessed August 22, 2025, https://health.ec.europa.eu/document/download/24aabd1d-931c-4799-a956-1347e9efc69e_en
  50. Type 1 Diabetes Mellitus Completed Phase Trials for Acetylcholine (DB03128) - DrugBank, accessed August 22, 2025, https://go.drugbank.com/indications/DBCOND0029465/clinical_trials/DB03128?phase=&status=completed
  51. Acetylcholine Completed Phase 2 Trials for Sickle Cell Anemia Treatment - DrugBank, accessed August 22, 2025, https://go.drugbank.com/drugs/DB03128/clinical_trials?conditions=DBCOND0028117&phase=2&purpose=treatment&status=completed
  52. Chronic Kidneys Disease Completed Phase Trials for Acetylcholine (DB03128) - DrugBank, accessed August 22, 2025, https://go.drugbank.com/indications/DBCOND0068430/clinical_trials/DB03128?phase=&status=completed

Published at: August 22, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.