Fact-checked by Grok 2 weeks ago

Neurotransmitter

Neurotransmitters are endogenous chemicals that transmit signals across a , from a (or from a to a muscle or ) to a target , enabling communication throughout the . They are synthesized within s and stored in synaptic vesicles at the presynaptic terminal, where they are released in response to an via calcium-dependent . Upon release into the synaptic cleft, neurotransmitters diffuse across the narrow gap and bind to specific receptors on the postsynaptic , either directly altering permeability (ionotropic receptors) or triggering intracellular signaling cascades (metabotropic receptors), thereby modulating the target cell's electrical activity or biochemical state. This process underlies essential neural functions, including , inhibition, and of synaptic transmission. The major classes of neurotransmitters are broadly categorized by chemical structure and function, with , monoamines, peptides, and others playing distinct roles in brain activity. Excitatory neurotransmitters, such as glutamate—the principal excitatory transmitter in the —promote neuronal depolarization and are crucial for , learning, and formation. In contrast, inhibitory neurotransmitters like gamma-aminobutyric acid (), which accounts for about 40% of inhibitory neurotransmission in the brain, and , the primary inhibitory transmitter in the , hyperpolarize postsynaptic neurons to dampen activity and maintain neural balance. Modulatory neurotransmitters, including monoamines such as (involved in reward, , and ), norepinephrine (regulating , , and responses), and serotonin (influencing , , and ), often act via G-protein-coupled receptors to fine-tune broader neural circuits. serves diverse roles, from facilitating at neuromuscular junctions to modulating and autonomic functions. Neurotransmitters are essential for coordinating complex processes like , , emotional , and cognitive functions, with their dysregulation implicated in various neurological conditions. After release, they are rapidly cleared from the synaptic cleft through into the presynaptic , enzymatic degradation, or diffusion, ensuring precise temporal control of signaling. More than 100 neurotransmitters have been identified, though the core ones—, , , serotonin, norepinephrine, and —dominate synaptic communication in the mammalian . Their discovery and study, beginning with in the early 20th century, have revolutionized , highlighting their role in both normal and therapeutic targets for disorders like and .

Definition and Fundamentals

Chemical Composition

Neurotransmitters are endogenous chemicals that transmit signals across a , from a presynaptic to a postsynaptic or target such as a muscle or . These signaling molecules enable communication in the by being released into the synaptic cleft and binding to specific receptors on the target . Neurotransmitters are broadly classified into several chemical categories based on their molecular structures and properties, including small-molecule neurotransmitters (such as , monoamines, , and purines), neuropeptides, and gaseous molecules like . Small-molecule neurotransmitters typically have low molecular weights (around 100–200 ) and include like glutamate (C5H9NO4), the principal excitatory neurotransmitter, and gamma-aminobutyric acid (, C4H9NO2), a major inhibitory neurotransmitter derived from glutamate. Monoamines, another subgroup of small molecules, encompass catecholamines such as (C8H11NO2) and norepinephrine (C8H11NO3), which feature a characteristic ring with hydroxyl groups derived from , and indolamines like serotonin (C10H12N2O), derived from . Neuropeptides are larger, consisting of short chains of 3 to 36 , such as or the , with molecular weights often exceeding 3000 . Gaseous neurotransmitters, exemplified by (NO, molecular weight 30 ), are unconventional due to their instability and lack of vesicular storage. The physicochemical properties of neurotransmitters, including and , critically influence their across the synaptic cleft and with receptors. Most small-molecule neurotransmitters, such as and monoamines, are polar and water-soluble, enabling their dissolution in the aqueous environment of the and storage in synaptic vesicles. Their moderate lipophilicity, arising from aromatic rings in monoamines, facilitates binding to lipid-embedded receptors while preventing excessive membrane penetration. In contrast, gaseous neurotransmitters like are highly lipophilic, allowing rapid through cell membranes without reliance on transporters or vesicles. Neuropeptides, being peptides, exhibit amphiphilic properties with hydrophilic backbones and variable side-chain s that affect their solubility and receptor affinity. For classical neurotransmitters, a substance must meet specific criteria related to its localization and functional role in synaptic transmission. It must be synthesized or actively taken up by the presynaptic and stored in synaptic vesicles. Upon presynaptic , it is released in a calcium-dependent manner into the synaptic cleft. When applied exogenously to the postsynaptic cell, it must mimic the natural response by binding to specific receptors and altering the target's electrical or biochemical properties. Finally, mechanisms must exist to inactivate or remove the substance from the synapse after signaling. Unconventional neurotransmitters, such as gaseous molecules, fulfill analogous roles through modified mechanisms, including on-demand synthesis and diffusion without vesicular storage.

Functions in Neural Signaling

Neurotransmitters serve as chemical messengers that bridge the gap between presynaptic s and postsynaptic cells at chemical synapses, enabling the transmission of signals across the synaptic cleft through their release and binding to receptors on the target cell. This process is triggered by action potentials arriving at the presynaptic terminal, leading to calcium influx and vesicular of neurotransmitters, which then diffuse to activate postsynaptic responses. In this way, neurotransmitters facilitate the propagation of electrical signals in the form of action potentials from one neuron to another or to effector cells like muscle or gland cells. The effects of neurotransmitters on postsynaptic cells can be classified as excitatory, inhibitory, or modulatory, depending on the type of receptor activated and the resulting change in . Excitatory neurotransmitters, such as glutamate, typically open channels that allow influx of cations like sodium, causing and increasing the likelihood of generation in the postsynaptic . Inhibitory neurotransmitters, including gamma-aminobutyric acid () and , promote hyperpolarization by facilitating chloride influx or potassium efflux, thereby reducing neuronal excitability and preventing excessive firing. Modulatory neurotransmitters, often monoamines like or serotonin, exert longer-lasting influences by altering the excitability of neural circuits through second-messenger systems, contributing to processes such as learning and emotional regulation without directly driving rapid synaptic transmission. Beyond immediate synaptic transmission, neurotransmitters play crucial roles in synaptic integration, where multiple inputs are summed spatially and temporally to determine whether a postsynaptic fires an , and in network oscillations that synchronize activity across regions for coordinated functions like and . For instance, inhibition helps generate rhythmic oscillations in cortical networks, while modulation can enhance theta rhythms in the . These mechanisms ensure efficient information processing and maintain the balance of excitation and inhibition in neural circuits. In addition to synaptic actions, neurotransmitters participate in non-synaptic transmission, such as volume transmission, where they diffuse over larger distances to influence multiple target cells via paracrine signaling, particularly for neuromodulators like monoamines and neuropeptides. This mode of signaling, distinct from the precise, point-to-point synaptic release, allows for broader regulation of neural activity and is mediated by extrasynaptic receptors, enabling slower, more diffuse effects on circuit dynamics. The fundamental roles of neurotransmitters in neural signaling exhibit remarkable evolutionary conservation, appearing in simple such as nematodes, where acetylcholine functions as an excitatory transmitter at neuromuscular junctions, and extending to complex mammalian systems with conserved pathways for glutamate, , and biogenic amines across bilaterians. This conservation underscores their ancient origins in secretory signaling predating synaptic structures, adapting over time to support diverse neural functions from basic locomotion to higher cognition.

Biosynthetic and Metabolic Processes

Synthesis Mechanisms

Neurotransmitters are primarily synthesized from precursors in the of presynaptic s or at their terminals, where specific enzymes catalyze the conversion processes. These syntheses occur in specialized types, such as neurons for catecholamines or neurons for serotonin, ensuring targeted production for synaptic transmission. The enzymes involved are typically transported from the cell body to the terminals via , allowing for on-demand synthesis close to release sites. For catecholamines like dopamine and norepinephrine, synthesis begins with the amino acid tyrosine, which is hydroxylated to L-3,4-dihydroxyphenylalanine (L-DOPA) by the enzyme tyrosine hydroxylase (TH), followed by decarboxylation of L-DOPA to dopamine by aromatic L-amino acid decarboxylase (AADC, also known as DOPA decarboxylase). Dopamine is then further converted to norepinephrine by dopamine β-hydroxylase in noradrenergic neurons. This pathway is localized in catecholaminergic neurons, such as those in the substantia nigra for dopamine. Serotonin (5-hydroxytryptamine) is synthesized from the , first hydroxylated to 5-hydroxytryptophan (5-HTP) by (TPH), and then decarboxylated to serotonin by AADC. TPH exists in two isoforms, TPH1 and TPH2, with TPH2 being the predominant form in the brain's neurons of the . The inhibitory neurotransmitter (GABA) is produced from the excitatory glutamate through by glutamic acid (GAD), which has two isoforms: GAD65 and GAD67. This is confined to neurons throughout the , with GAD67 primarily responsible for basal GABA production in the cytoplasm. Acetylcholine synthesis involves the esterification of choline with acetyl-coenzyme A () by the enzyme (), occurring in neurons such as those in the and motor neurons. is generated from glucose metabolism and requires ATP for its formation via in mitochondria. Synthesis rates are tightly regulated to match neuronal activity and demand. serves as the rate-limiting enzyme in catecholamine production, subject to feedback inhibition by end-product catecholamines like , which bind to its regulatory domain to reduce activity. Similarly, TPH is rate-limiting for serotonin synthesis and experiences feedback inhibition by serotonin. GAD activity for is regulated post-translationally, including through , while levels are controlled transcriptionally in response to neuronal signaling. These processes require specific cofactors, such as () for and TPH in monoamine synthesis, which acts as a hydrogen donor and is essential for enzymatic function. Localization to particular types ensures specificity, with deficiencies in these pathways linked to neurological disorders. Following synthesis, neurotransmitters are briefly stored in vesicles prior to release.

Storage and Release Dynamics

Neurotransmitters are stored in synaptic vesicles within the presynaptic terminal, where they are concentrated against their s by specific vesicular transporters powered by a proton (ΔμH⁺) generated by vacuolar H⁺-ATPases. For monoamines such as , norepinephrine, and serotonin, the vesicular monoamine transporters (VMAT1 and VMAT2) facilitate uptake primarily driven by the gradient (ΔpH), exchanging two protons for one monoamine , achieving concentrations up to 100,000-fold higher than in the . Glutamate, the primary excitatory neurotransmitter, is loaded into vesicles via vesicular glutamate transporters (VGLUT1-3), which rely mainly on the component (Δψ) of the gradient, with ions modulating activity to optimize filling. Inhibitory neurotransmitters like and are transported by the vesicular GABA transporter (VGAT), utilizing both ΔpH and Δψ, often with cotransport, ensuring efficient packaging for release. Synaptic vesicles exist in distinct types tailored to their cargo. Small synaptic vesicles (40-60 nm in diameter), which appear clear in electron micrographs, primarily store classical small-molecule neurotransmitters like glutamate, , and monoamines, enabling rapid, high-frequency release at active zones. In contrast, large dense-core vesicles (90-250 nm), identifiable by their electron-dense cores, package neuropeptides such as or enkephalins, often alongside small molecules in some neurons, and support slower, activity-dependent release from extrasynaptic sites. These differences in size and composition influence release kinetics, with small vesicles recycling quickly via clathrin-mediated to sustain ongoing signaling. Upon arrival of an , neurotransmitter release is triggered by calcium influx through voltage-gated calcium channels (e.g., P/Q- or N-type) clustered at the active zone, raising local Ca²⁺ concentrations to micromolar levels within microseconds. This Ca²⁺ binds to synaptotagmin-1, the primary Ca²⁺ sensor, inducing a conformational change that promotes assembly of the SNARE complex—comprising syntaxin-1, SNAP-25 on the plasma membrane, and synaptobrevin-2 (VAMP2) on the vesicle—driving rapid through membrane fusion. Release occurs in quanta, with each vesicle representing one quantum; spontaneous miniature end-plate potentials (MEPPs) reflect single-vesicle fusion events, while evoked release involves multiple quanta whose number follows statistics. The probability of release is modulated by readily releasable, recycling, and reserve vesicle pools, with Ca²⁺ enhancing recruitment from these pools to fine-tune synaptic strength. Co-release of multiple neurotransmitters from the same vesicle adds complexity to signaling, particularly in neurons packaging a fast-acting with a modulatory . For instance, central excitatory neurons often co-store glutamate and neuropeptides like cholecystokinin in small synaptic vesicles or dense-core vesicles, allowing simultaneous release to elicit both rapid ionotropic and prolonged metabotropic effects. This co-packaging is facilitated by compatible vesicular transporters and gradients, enabling diverse postsynaptic responses without requiring separate vesicle populations.

Inactivation and Clearance

After neurotransmitter release into the synaptic cleft, rapid inactivation and clearance are essential to terminate signaling, prevent receptor overstimulation, and maintain synaptic homeostasis. Enzymatic degradation represents a primary mechanism for terminating the action of several neurotransmitters. For acetylcholine (ACh), acetylcholinesterase (AChE) hydrolyzes it into choline and acetate within milliseconds in the synaptic cleft. Monoamine neurotransmitters, such as dopamine and serotonin, undergo oxidative deamination by monoamine oxidase (MAO), primarily within presynaptic terminals or glial cells, converting them into inactive metabolites. Reuptake via specific transporter proteins provides another key clearance pathway, allowing neurotransmitters to be recycled back into the presynaptic . The (DAT) facilitates rapid uptake of from the synaptic cleft, while the (SERT) performs a similar function for serotonin. These sodium-dependent transporters operate on timescales of milliseconds to seconds, depending on concentration gradients and transporter density. For gaseous neurotransmitters like nitric oxide (NO), clearance occurs primarily through passive diffusion away from the synapse due to its lipophilic nature and lack of specific transporters or enzymes for rapid degradation. NO diffuses freely across cell membranes, with its signaling terminated by reactions with superoxide or hemoglobin, leading to a half-life of seconds in biological tissues. Glial cells, particularly , play a crucial role in clearing excitatory like glutamate to prevent . express excitatory transporters (EAATs), such as EAAT1 and EAAT2, which mediate sodium- and potassium-dependent uptake of glutamate from the synaptic cleft into glial cells for conversion to . This process helps regulate extracellular glutamate levels and supports neuronal supply for resynthesis. Recycling of cleared components enables efficient neurotransmitter replenishment. For instance, choline produced from AChE-mediated is reuptaken by the high-affinity choline transporter (CHT1) into presynaptic terminals, where it is reused in ACh synthesis via . Similarly, reuptaken monoamines like are repackaged into vesicles by the (VMAT2). The timescales of clearance vary by neurotransmitter type and mechanism. Small-molecule neurotransmitters, such as and glutamate, are typically cleared within milliseconds through enzymatic degradation or , ensuring precise temporal control of signaling. In contrast, neuropeptides rely mainly on and extracellular , resulting in slower clearance over seconds to minutes, which contributes to their prolonged modulatory effects.

Molecular Mechanisms of Action

Receptor Interactions

Neurotransmitters exert their effects primarily through to specific receptors on the postsynaptic or other target sites, initiating rapid or modulated responses in neural signaling. Receptors are broadly classified into two main types: ionotropic and metabotropic. Ionotropic receptors function as ligand-gated channels, allowing direct upon neurotransmitter , which leads to fast synaptic transmission. For instance, the N-methyl-D-aspartate (, activated by glutamate, permits influx of sodium and calcium s, generating fast excitatory postsynaptic potentials (EPSPs) essential for synaptic excitation. In contrast, metabotropic receptors are G-protein-coupled receptors (GPCRs) that indirectly influence channels or cellular processes via intermediary signaling molecules, resulting in slower, modulatory effects. The D2 exemplifies this class, coupling to Gi/o proteins to inhibit and modulate neuronal excitability over longer timescales. Binding specificity between neurotransmitters and their receptors is governed by precise molecular interactions, characterized by affinity constants that quantify the strength of association. The (Kd) typically ranges from nanomolar to micromolar levels, reflecting high selectivity; for example, binds to nicotinic receptors with a Kd of approximately 160 μM. This specificity ensures that only neurotransmitters or structurally similar ligands effectively , minimizing off-target effects in diverse neural circuits. binding often stabilizes the receptor in an active conformation, whereas antagonists lock it in an inactive state, directly influencing synaptic efficacy. Receptors are strategically localized to optimize signal transmission and regulation. Postsynaptic densities (PSDs) are specialized protein scaffolds in excitatory synapses where ionotropic receptors, such as and NMDA types for glutamate, cluster to enhance sensitivity to released neurotransmitters and facilitate rapid postsynaptic responses. Conversely, presynaptic autoreceptors, often metabotropic, reside on the presynaptic terminal and detect spillover neurotransmitter to provide , thereby inhibiting further release and maintaining synaptic ; for example, presynaptic D2 autoreceptors on terminals reduce secretion in response to elevated extracellular levels. Allosteric modulation fine-tunes receptor function by binding at sites distinct from the orthosteric neurotransmitter site, altering binding affinity or . Positive allosteric modulators enhance the receptor's response to the endogenous , while negative modulators diminish it. A prominent example is the action of benzodiazepines on _A receptors, where they bind at an allosteric site on the γ2 subunit interface, increasing GABA affinity and potentiating influx for enhanced inhibition. Such allows for nuanced control of synaptic strength without directly competing with the neurotransmitter. From an evolutionary perspective, neurotransmitter receptor families exhibit remarkable conservation across species, reflecting ancient origins in early metazoans. Ionotropic glutamate receptors and GPCR families share core structural motifs preserved across diverse phyla, underscoring their fundamental role in neural function.

Signal Transduction Pathways

Upon binding to their receptors, neurotransmitters initiate intracellular signal transduction pathways that convert extracellular signals into cellular responses, primarily through two major classes: ionotropic and metabotropic receptors. Ionotropic receptors function as ligand-gated ion channels, enabling direct and rapid ion flux across the postsynaptic membrane to alter neuronal excitability. In ionotropic pathways, neurotransmitter binding directly opens the pore, allowing specific ions to flow and generate immediate postsynaptic potentials. For excitatory transmission, such as with glutamate binding to receptors, sodium (Na⁺) and potassium (K⁺) influx causes membrane depolarization, facilitating initiation. In contrast, inhibitory neurotransmitters like or activate receptors that permit (Cl⁻) influx, leading to hyperpolarization and reduced neuronal firing. These pathways operate on a timescale, enabling fast synaptic transmission essential for precise neural communication. Metabotropic pathways, mediated by G-protein-coupled receptors (GPCRs), involve indirect signaling through heterotrimeric G proteins that dissociate upon receptor activation to modulate intracellular effectors. Different G protein subtypes direct distinct cascades: Gs proteins stimulate adenylyl cyclase to increase cyclic AMP (cAMP) levels, activating protein kinase A (PKA); Gq proteins activate phospholipase C, producing inositol trisphosphate (IP₃) and diacylglycerol (DAG), which release intracellular calcium (Ca²⁺) and activate protein kinase C (PKC); and Gi proteins inhibit adenylyl cyclase or open potassium (K⁺) channels, promoting hyperpolarization. Examples include norepinephrine acting via β-adrenergic receptors (Gs-coupled) to enhance excitability or serotonin via 5-HT1 receptors (Gi-coupled) to suppress it. These slower processes, lasting seconds to minutes, allow for neuromodulation and broader cellular adjustments. Signal amplification in these pathways occurs through enzymatic cascades, particularly in metabotropic routes, where second messengers like activate kinases such as , which phosphorylate ion channels, pumps, or transcription factors to propagate and intensify the signal. For instance, can phosphorylate voltage-gated calcium channels to modulate neurotransmitter release probability. This multistep amplification enables a single receptor activation to elicit widespread effects within the . Crosstalk between neurotransmitter pathways and other signaling systems, such as those involving like (BDNF), integrates synaptic activity with trophic support, where receptors (e.g., TrkB) can enhance G-protein cascades to fine-tune neuronal survival and excitability. Overall, ionotropic pathways drive rapid, localized responses, while metabotropic ones provide sustained modulation, together enabling the nervous system's dynamic information processing.

Synaptic Plasticity Effects

Neurotransmitters play a central role in Hebbian plasticity, a form of synaptic strengthening or weakening based on correlated pre- and postsynaptic activity, often summarized by the principle "neurons that fire together wire together." Long-term potentiation (LTP) and long-term depression (LTD) represent key manifestations of this process, where high-frequency stimulation induces persistent increases or decreases in synaptic efficacy, respectively. In LTP, glutamate release activates NMDA receptors, allowing calcium influx upon coincident presynaptic and postsynaptic depolarization, which triggers intracellular cascades leading to enhanced synaptic transmission. This calcium-dependent signaling promotes the insertion of AMPA receptors into the postsynaptic membrane via trafficking mechanisms involving exocytosis and stabilization at the synapse. Conversely, in LTD, moderate calcium levels through NMDA receptors facilitate AMPA receptor endocytosis, reducing synaptic strength. Glutamate is pivotal in hippocampal LTP, where its binding to NMDA receptors during theta-burst or high-frequency stimulation initiates the calcium influx essential for trafficking and long-lasting synaptic enhancement. Dopamine, acting as a neuromodulator, contributes to reward-based by gating plasticity in circuits like the and , where phasic release during unexpected rewards strengthens synapses via D1/D5 receptor activation and enhancement of LTP-like processes. This modulation integrates motivational signals with Hebbian rules, enabling associative learning. Homeostatic scaling provides a complementary to Hebbian , maintaining overall by globally adjusting synaptic strengths in response to activity changes, often through balanced of excitatory glutamate and inhibitory . Reduced activity triggers multiplicative upregulation of surface expression at glutamatergic and increased clustering, compensating to restore firing rates. Conversely, heightened activity downscales these synapses via receptor removal, preventing runaway excitation or silencing. This form of operates on slower timescales than Hebbian mechanisms, ensuring long-term circuit . Excessive glutamate release can lead to pathological plasticity through , where prolonged activation causes overwhelming calcium influx, activating proteases, lipases, and endonucleases that damage cellular components and induce neuronal death. This process underlies synaptic weakening and circuit dysfunction in conditions of unchecked excitatory . Experimental evidence for these effects derives from hippocampal slice preparations, where frequency-dependent protocols reveal LTP induction with high-frequency stimulation (e.g., 100 Hz tetani) and with low-frequency stimulation (1-5 Hz), as measured by field excitatory postsynaptic potentials. Blocking NMDA receptors with antagonists like APV abolishes LTP in these slices, confirming the calcium influx pathway, while manipulations of trafficking mimic or occlude changes. Such studies demonstrate how neurotransmitter dynamics directly sculpt adaptive synaptic modifications.

Historical and Methodological Foundations

Key Discoveries

In the early , the debate between electrical and chemical modes of neural transmission began to favor the latter through experiments with , a paralytic . French physiologist demonstrated in the 1850s that curare blocks neuromuscular transmission specifically at the junction between nerve and muscle, without affecting nerve conduction itself, providing early evidence for a chemical intermediary in synaptic signaling. A pivotal breakthrough occurred in 1921 when conducted his famous frog heart experiments, proving chemical . By stimulating the of one frog heart and transferring the perfusate to a second heart, Loewi observed slowed beating in the recipient, attributing the effect to a released substance he termed "vagusstoff," later identified as . This work, shared with Henry Dale in the 1936 , established the chemical basis of synaptic transmission. The mid-20th century saw the identification of additional key neurotransmitters. In 1946, Ulf von Euler isolated norepinephrine from sympathetic nerves and confirmed its role as the primary transmitter in the , earning him a share of the 1970 for elucidating its storage and release mechanisms.64948-3/pdf) Concurrently, in 1948, Maurice Rapport and colleagues at the isolated a vasoconstrictive factor from blood serum, naming it serotonin (5-hydroxytryptamine), which was soon recognized as a neurotransmitter modulating and other functions. The 1950s and 1960s expanded the roster to include transmitters. Japanese pharmacologist Takashi Hayashi reported in 1954 that sodium glutamate excites the , injecting it into the of dogs to induce rhythmic movements, proposing glutamate as an excitatory transmitter. Independently, Austrian-born physiologist Ernst Florey identified an inhibitory factor in nervous systems during the 1950s, which collaborative work in 1957 chemically confirmed as gamma-aminobutyric acid (), the brain's chief inhibitory neurotransmitter. Later decades revealed more diverse transmitters, including and gases. In the early , Susan Leeman's team purified and sequenced , an 11-amino-acid peptide first described in the 1930s, establishing it as the archetypal involved in pain transmission and inflammation. By the 1990s, (NO) emerged as a unconventional gaseous transmitter; Furchgott, , and Ferid Murad's discoveries of NO's role in vascular relaxation and signaling earned the 1998 , with Ignarro specifically highlighting its neuronal functions.

Techniques for Identification

Classical techniques for identifying neurotransmitters primarily involved bioassays that assessed physiological responses to candidate substances extracted from neural tissue. For instance, preparations, such as frog rectus abdominis muscle assays, measured contractions induced by to confirm its transmitter role through dose-response similarities. Pharmacological mimics further validated identifications by replicating or blocking effects; eserine (physostigmine), an , prolonged acetylcholine-mediated responses in bioassays, distinguishing it from other candidates like adrenaline. Biochemical approaches provide direct quantification and structural confirmation of neurotransmitters in tissues or fluids. (HPLC), often coupled with electrochemical detection (HPLC-ECD), separates and detects small-molecule transmitters like and serotonin with nanomolar sensitivity, enabling analysis of microdialysate samples from brain regions. (MS), typically integrated with liquid chromatography (LC-MS/MS), offers high specificity for simultaneous identification of multiple transmitters, including like glutamate, achieving picomolar limits of detection without derivatization. For neuropeptides, radioimmunoassays (RIA) employ specific antibodies to quantify low-abundance transmitters like in neural extracts, providing quantitative measures of content and release. Electrophysiological methods record synaptic events to infer neurotransmitter involvement through receptor-mediated currents. Patch-clamp techniques, in whole-cell or voltage-clamp configurations, measure postsynaptic potentials or currents evoked by synaptic , identifying transmitters via (e.g., bicuculline-sensitive currents for ). enables selective activation of transmitter-specific neurons using , allowing paired electrophysiological recordings to confirm release dynamics, such as dopamine-induced currents in target cells. Imaging techniques visualize neurotransmitter dynamics in living tissue with spatiotemporal precision. Fluorescent sensors, including genetically encoded indicators like GRAB (GPCR-activation-based) for , bind transmitters and undergo conformational changes that increase fluorescence, enabling real-time monitoring of release in behaving animals with sub-second resolution. Calcium indicators such as , while primarily tracking presynaptic calcium influx, indirectly reveal transmitter release probability during synaptic events. Genetic approaches use engineered models to verify neurotransmitter roles by disrupting synthesis, storage, or release. mice lacking (VMAT2) exhibit depleted monoamine storage, confirming its essentiality for and serotonin vesicular loading through reduced evoked release and behavioral deficits. Such models, combined with rescue experiments, distinguish transmitter-specific functions from compensatory mechanisms.

Classification and Diversity

Major Classes

Neurotransmitters are classified into several major categories based on their and , including small-molecule neurotransmitters (such as , biogenic amines, , and purines), neuropeptides, and gaseous molecules. Small-molecule neurotransmitters encompass biogenic amines and , which mediate fast synaptic transmission at the majority of (CNS) synapses. Biogenic amines, such as , serotonin, and histamine, are involved in modulating mood, arousal, and reward pathways and are estimated to operate at 5-10% of synapses. include the excitatory neurotransmitter glutamate, which serves as the primary excitatory agent at approximately 80% of CNS synapses, and the inhibitory neurotransmitters and , which together account for 20-30% of synapses, with predominant in the and more common in the . Acetylcholine functions as a key small-molecule neurotransmitter primarily at neuromuscular junctions in the , where it triggers , and in autonomic ganglia, facilitating transmission in both sympathetic and parasympathetic pathways. Neuropeptides represent a diverse class of over 100 identified types, often acting as neuromodulators with slower onset and longer duration compared to small molecules; examples include , which alleviate pain by binding to receptors, and orexins, which promote and . Gaseous neurotransmitters, such as (NO) and (CO), differ from classical transmitters by being freely diffusible, non-vesicular messengers involved in to modulate and . Purines, notably (ATP), often serve as co-transmitters alongside other small molecules, exerting excitatory effects in sensory neurons, autonomic ganglia, and certain CNS circuits.

Structural and Functional Criteria

Neurotransmitters are defined by a set of established structural and functional criteria that ensure their role as specific signaling molecules in synaptic transmission. These criteria, originally outlined by John C. Eccles in his seminal 1964 work on synaptic physiology, require that a candidate substance be synthesized and stored in the presynaptic , released in a calcium-dependent manner upon presynaptic , bind to specific receptors on the postsynaptic cell to elicit a physiological response, mimic the natural synaptic effect when applied exogenously, and possess a mechanism for rapid inactivation or clearance from the . These standards, refined in subsequent neuroscientific literature, emphasize the substance's localization within presynaptic terminals and its Ca²⁺-evoked release, typically validated through techniques like for presence and microdialysis for release dynamics. Functionally, neurotransmitters exhibit diverse modes of action that influence synaptic communication speed and scope. Ionotropic receptors mediate fast, point-to-point transmission by directly gating channels, resulting in rapid excitatory or inhibitory postsynaptic potentials within milliseconds, as seen with glutamate acting on receptors. In contrast, metabotropic receptors trigger slower, diffuse modulation through G-protein-coupled signaling cascades, often leading to prolonged changes in cellular excitability via second messengers like , exemplified by norepinephrine's effects on β-adrenergic receptors. This allows neurotransmitters to support both precise, localized signaling and broader network modulation, with functional efficacy typically requiring synaptic concentrations exceeding 1 μM to activate postsynaptic responses effectively. Structurally, neurotransmitters vary in , which dictates their , mechanisms, and properties. Polar molecules, such as derivatives like glutamate and , are hydrophilic and rely on specific transmembrane transporters (e.g., EAATs for glutamate) for uptake and due to limited passive across bilayers. Non-polar or lipophilic substances, including gaseous transmitters like , exhibit greater membrane permeability and diffuse freely without dedicated transporters, enabling volume transmission over larger distances. These structural features ensure targeted delivery and prevent nonspecific spillover, with polar neurotransmitters often stored in vesicles for regulated release. Controversies arise with substances like endocannabinoids (e.g., ), which function as retrograde messengers synthesized postsynaptically and diffusing backward to inhibit presynaptic release via CB1 receptors, thus deviating from classical anterograde criteria. Unlike traditional neurotransmitters, they lack vesicular storage and Ca²⁺-dependent from presynaptic terminals, challenging strict classification while highlighting expanded definitions of synaptic signaling.

Neurotransmitter Systems in the Nervous System

Central Nervous System Roles

In the (CNS), neurotransmitters play essential roles in modulating neural circuits within the and , facilitating processes such as , , and emotional regulation. Glutamate serves as the primary excitatory neurotransmitter, predominantly released by pyramidal neurons in the and . These neurons utilize ionotropic receptors like and to mediate synaptic transmission, enabling (LTP), a key mechanism underlying learning and formation. For instance, activation in hippocampal CA1 pyramidal neurons allows calcium influx that triggers signaling cascades for synaptic strengthening, as demonstrated in studies of tasks. Complementing excitation, gamma-aminobutyric acid () acts as the main inhibitory neurotransmitter, primarily through that constitute about 10-15% of hippocampal neurons. These target principal cells at somatic, dendritic, or axonal sites, providing fast phasic inhibition via GABA_A receptors and slower tonic inhibition to maintain excitation-inhibition balance and prevent hyperexcitability. Subtypes such as parvalbumin-expressing basket cells deliver precise perisomatic inhibition to synchronize network oscillations, while somatostatin-expressing cells modulate distal dendritic inputs, both critical for averting in cortical and hippocampal circuits. Dopamine, a key modulatory neurotransmitter, operates through distinct pathways originating in the . The , arising from neurons, projects to the and , influencing reward processing, , and by modulating in limbic regions. In contrast, the from pars compacta neurons innervates the dorsal striatum, regulating voluntary motor control and habit formation within circuits. Serotonin, released by neurons in the , exerts widespread influence on mood-regulating areas through projections across the . Dorsal raphe serotonergic neurons promote antidepressant-like effects by reducing passive coping behaviors in stress models, while median raphe neurons contribute to anxiety modulation via inputs to the and . This serotonergic modulation fine-tunes emotional responses, with balanced activity between dorsal and median raphe subpopulations essential for adaptive behavior. Neurotransmitter systems integrate in structures like the and to ensure coordinated motor function. In the , dopaminergic inputs from the interact with and signals to balance direct and indirect pathways, facilitating smooth movement initiation and vigor. Cerebellar circuits, receiving inputs via thalamic relays, refine these signals for precise coordination and error correction, with climbing fiber activity encoding adjustments that prevent . This interplay maintains overall CNS stability, underscoring the reciprocal modulation between excitatory, inhibitory, and modulatory transmitters.

Peripheral and Non-Neuronal Roles

Neurotransmitters play crucial roles beyond the central nervous system, influencing peripheral organs, sensory pathways, and even non-neuronal tissues through autonomic, enteric, and other signaling mechanisms. In the autonomic nervous system, acetylcholine (ACh) serves as the primary neurotransmitter in the parasympathetic branch, where it binds to muscarinic receptors—particularly M2 subtypes in the heart—to slow heart rate and reduce contractility, promoting rest and digestion. Conversely, norepinephrine acts as the main postganglionic neurotransmitter in the sympathetic nervous system, facilitating the "fight-or-flight" response by increasing heart rate, vasoconstriction in certain vascular beds, and mobilization of energy reserves through adrenergic receptors. In the (ENS), which governs gastrointestinal function independently of central control, serotonin (5-HT) is a key modulator, with approximately 95% of the body's serotonin synthesized and stored in enterochromaffin cells of the gut. This neurotransmitter regulates gut by activating 5-HT receptors on enteric neurons and , enhancing and to facilitate and propulsion of contents. Dysregulation of enteric serotonin signaling can alter patterns, underscoring its essential role in maintaining gastrointestinal . Non-neuronal cells also utilize neurotransmitters for intercellular communication. Glutamate, traditionally known as an excitatory neurotransmitter, functions as an immunomodulator in immune cells such as T lymphocytes, where it binds to glutamate receptors to influence activation, proliferation, and release, thereby shaping adaptive immune responses. Similarly, (NO), a gaseous neurotransmitter produced by endothelial cells in blood vessels, induces by activating in cells, relaxing vessels to regulate blood flow and prevent . These actions highlight neurotransmitters' broader paracrine effects in non-neural contexts. In peripheral sensory systems, , a neurotransmitter, is released from nociceptors—primary afferent neurons detecting —and transmits nociceptive signals to the via neurokinin-1 receptors, contributing to the perception of inflammatory and . This release also promotes neurogenic inflammation by inducing plasma and immune cell at peripheral sites. Inter-tissue communication further extends these roles, as seen in the gut-brain where serotonin from the ENS signals via vagal afferents to influence central autonomic regulation, modulating mood, appetite, and stress responses through bidirectional neural pathways.

Pharmacological and Therapeutic Implications

Drug Modulation Strategies

Drug modulation strategies aim to therapeutically alter neurotransmitter levels or activity by targeting various stages of their lifecycle, from and to release and , without directly interacting with receptors. These approaches enhance or diminish synaptic to restore balance in neurotransmitter systems, often addressing deficiencies or excesses associated with neurological conditions. By intervening at presynaptic mechanisms, such strategies can increase extracellular neurotransmitter availability, thereby influencing downstream signaling pathways. One primary method involves reuptake inhibitors, which block the presynaptic transporters responsible for clearing neurotransmitters from the synaptic cleft, thereby elevating their concentrations. For instance, selective serotonin reuptake inhibitors (SSRIs) target the () on the presynaptic , preventing serotonin and allowing greater accumulation in the . This inhibition prolongs signaling, a central to their therapeutic effects. Similarly, inhibitors of other monoamine transporters, such as norepinephrine or , operate on analogous principles to sustain elevated levels of these neurotransmitters. Enzyme inhibitors represent another key strategy, focusing on preventing the of neurotransmitters to maintain higher cytosolic pools available for release. inhibitors () irreversibly bind to the enzyme (), which catalyzes the oxidative of monoamines like serotonin, norepinephrine, and . By blocking this breakdown, MAOIs increase the intracellular concentrations of these neurotransmitters, enhancing their vesicular packaging and subsequent synaptic release. This approach is particularly relevant for monoaminergic systems, where enzymatic limits transmitter availability. Vesicular modulators target the storage of neurotransmitters within synaptic vesicles, altering the reserves available for . Reserpine, a classic example, inhibits the (VMAT), which normally sequesters monoamines into vesicles using a . By to VMAT and blocking this uptake, reserpine causes depletion of vesicular stores, leading to reduced neurotransmitter release upon and eventual cytosolic leakage, which can be degraded or reverse-transported. This mechanism disrupts monoamine transmission, historically used to model depletion states. Release enhancers promote the expulsion of neurotransmitters into the by manipulating transporter dynamics or vesicular trafficking. Amphetamines act as substrates for plasma membrane transporters like the (), inducing a reversal of their normal uptake function through conformational changes and events. This reverse transport effluxes (and other monoamines) from the to the , independent of vesicular release, thereby acutely boosting synaptic levels. Such actions amplify and noradrenergic signaling. Clinical strategies often combine these tactics to balance neurotransmitter systems, particularly by augmenting precursor availability to replenish depleted pools. Levodopa serves as a precursor that crosses the blood-brain barrier via large neutral transporters and is decarboxylated by into within neurons. This loading approach increases cytosolic for vesicular storage and release, compensating for synthetic deficits in affected pathways. Integration of these strategies allows for tailored modulation, optimizing therapeutic outcomes while minimizing off-target effects.

Agonists and Antagonists

Agonists are substances that bind to neurotransmitter receptors and activate them, mimicking the effects of endogenous neurotransmitters to elicit a biological response. These compounds can be classified based on their degree of activation relative to the natural . Full agonists produce the maximum possible response upon receptor binding, equivalent to that of the endogenous neurotransmitter. For instance, acts as a full agonist at nicotinic acetylcholine receptors (nAChRs), binding to the α4β2 subtype and opening the to allow cation influx, thereby depolarizing the postsynaptic membrane. Partial agonists, in contrast, bind to the same site but elicit a submaximal response even at full receptor occupancy, often due to a lower capacity for conformational change in the receptor. exemplifies this at mu-opioid receptors, where it activates the receptor to a lesser extent than full agonists like , resulting in ceiling effects on analgesia and respiratory depression. Inverse agonists bind to the same receptor but stabilize an inactive conformation, reducing basal activity in constitutively active receptors; functions as an inverse at 5-HT2A serotonin receptors, decreasing receptor signaling even in the absence of agonist stimulation. Antagonists inhibit neurotransmitter effects by binding to receptors without activating them, thereby preventing or reducing the action of agonists. Competitive antagonists bind reversibly to the orthosteric site, competing directly with the neurotransmitter or for binding; their effects can be overcome by increasing agonist concentration. serves as a classic competitive antagonist at mu-opioid receptors, rapidly displacing opioids like to reverse overdose effects by blocking G-protein coupling and downstream inhibition of . Non-competitive antagonists, however, bind to an allosteric site or irreversibly to the orthosteric site, altering receptor function without direct competition; their inhibition persists regardless of agonist concentration. exemplifies non-competitive antagonism at NMDA glutamate receptors, entering the pore to block calcium influx in an open-channel state, thereby disrupting excitatory transmission. Selectivity in agonists and antagonists refers to the preference for specific receptor subtypes, minimizing off-target effects. Many compounds target subtypes within neurotransmitter families, such as adrenergic receptors. , a non-selective beta-blocker, antagonizes both β1- and β2-adrenergic receptors by binding to the catecholamine site, inhibiting norepinephrine-mediated increases in and bronchodilation. In contrast, selective agents like metoprolol primarily target β1-receptors in cardiac tissue, reducing cardiovascular effects while sparing β2-mediated pulmonary functions. Receptor binding kinetics distinguish between affinity, which measures the strength of ligand-receptor association (quantified by the dissociation constant K_d), and efficacy, which reflects the ligand's ability to stabilize the active receptor state and produce a response (often denoted as \epsilon). High-affinity ligands bind tightly but may have low efficacy if they fail to induce conformational changes, as seen in partial agonists. These properties determine potency (EC_{50}, the concentration for half-maximal effect) and therapeutic windows, with slow dissociation kinetics prolonging blockade in antagonists. Chronic exposure to agonists can lead to receptor desensitization, a side effect where prolonged activation reduces receptor responsiveness through mechanisms like by kinases (e.g., GRKs) and subsequent binding, uncoupling the receptor from G-proteins. This is prominent in β2-adrenergic receptors with long-acting agonists, contributing to diminished bronchodilation over time. Desensitization may also involve receptor internalization and downregulation, exacerbating tolerance in systems like signaling.

Pathophysiological Associations

Imbalances and Disorders

Imbalances in neurotransmitter systems arise from disruptions in , release, , or receptor function, leading to either excess or deficiency that contributes to neurological and psychiatric disorders. Excess neurotransmitter activity, such as glutamate-mediated , occurs when prolonged activation of glutamate receptors overwhelms cellular calcium , triggering neuronal death; this mechanism is prominent in ischemic stroke, where rapid glutamate release during energy failure exacerbates injury. Conversely, deficiencies, exemplified by dopamine loss in , result from degeneration of dopaminergic neurons in the , reducing striatal levels and impairing . These imbalances highlight how neurotransmitter dysregulation can directly impair neural signaling and circuit integrity. Many disorders involve multifactorial interactions across neurotransmitter systems rather than isolated deficits. For instance, in , dysregulated serotonin modulation of release in mesolimbic pathways can amplify hyperactivity, contributing to psychotic symptoms through altered prefrontal and striatal signaling. Such interactions underscore the interconnected nature of neurotransmitter networks, where compensatory changes in one system may exacerbate vulnerabilities in another, complicating disease etiology. Diagnosis of neurotransmitter imbalances often relies on direct measurement techniques. (CSF) analysis quantifies neurotransmitter metabolites like homovanillic acid (for ) or (for serotonin), providing insights into turnover; this method is particularly valuable for identifying primary neurotransmitter disorders. (PET) imaging of transporters, using radioligands such as [11C]raclopride for , visualizes binding potential and endogenous release dynamics , enabling non-invasive assessment of system integrity across brain regions. Epidemiologically, neurotransmitter imbalances are linked to high-prevalence conditions; for example, dysregulation of serotonin systems is implicated in , which has a lifetime of approximately 12-16% in the general population, with reduced serotonin signaling observed in a significant subset of cases through CSF studies. These associations emphasize the broad impact of neurotransmitter-related pathologies. Recent post-2020 research has revealed the gut microbiome's role in modulating neurotransmitter production, particularly serotonin, where commensal bacteria influence host metabolism and activity to produce up to 90% of peripheral serotonin, potentially affecting central levels via the gut-brain axis and contributing to mood disorders. This insight highlights emerging environmental factors in neurotransmitter .

Specific Neurotransmitter Dysfunctions

Dysfunctions in dopamine neurotransmission are central to several neurological and psychiatric disorders. In , the progressive loss of dopaminergic neurons in the leads to dopamine depletion in the , resulting in motor symptoms such as bradykinesia, rigidity, and . This nigral degeneration disrupts the balance between direct and indirect pathways in the , causing hypoactivity in motor-facilitating circuits and contributing to slowed movements. In contrast, is associated with hyperactivity in the mesolimbic dopamine pathway, particularly from the to the , which underlies positive symptoms like hallucinations and delusions. This hyperdopaminergic state is thought to arise from dysregulated dopamine release and receptor sensitivity, amplifying aberrant salience attribution to internal stimuli. Serotonin dysregulation contributes to mood and anxiety disorders through altered firing and autoregulatory mechanisms in the . In , reduced serotonergic neuron firing in the diminishes serotonin release across projection areas like the and , impairing mood regulation and leading to symptoms such as persistent sadness and . This hypoactivity may stem from enhanced negative feedback via 5-HT1A autoreceptors, limiting serotonin synthesis and transmission. For anxiety disorders, impaired serotonin autoregulation, particularly involving 5-HT1A somatodendritic autoreceptors, results in excessive serotonergic inhibition of neurons, disrupting adaptive responses to stress and manifesting as heightened worry, panic, and autonomic arousal. Such dysregulation can lead to unbalanced excitation in limbic regions like the . Glutamate, the primary excitatory neurotransmitter, is implicated in neurodegenerative and seizure disorders via excitotoxic mechanisms. In epilepsy, excessive glutamate release and impaired uptake cause hyperexcitability in neuronal networks, particularly through overactivation of NMDA and receptors, leading to synchronized firing, , and potential neuronal damage. This imbalance overwhelms inhibitory control, propagating ictal activity across brain regions like the and . In , amyloid-beta peptides induce changes in function, including enhanced NMDA receptor activity and reduced trafficking, which promote synaptic dysfunction, calcium overload, and progressive cognitive decline such as memory loss. These alterations exacerbate pathology and neuronal loss in affected areas. Acetylcholine (ACh) deficits underlie neuromuscular and cognitive impairments in autoimmune and degenerative conditions. is primarily caused by autoantibodies against postsynaptic nicotinic ACh receptors at the , leading to receptor blockade, internalization, and complement-mediated destruction, which impairs and results in fluctuating weakness, fatigue, and ptosis. This autoimmune attack reduces the number of functional receptors by up to 70-80%, disrupting endplate potentials. In , selective loss of neurons in the , particularly the of Meynert, causes ACh depletion in cortical and hippocampal targets, contributing to attentional deficits, memory impairment, and overall cognitive decline. This hypofunction correlates with and pathology, amplifying synaptic failure. GABAergic dysfunction disrupts inhibitory control, contributing to sleep and substance use disorders. In insomnia, reduced GABA levels or impaired GABA_A receptor function in sleep-regulating circuits, such as the ventrolateral preoptic nucleus, fail to suppress wake-promoting neurons in the arousal centers, leading to prolonged sleep latency, fragmented sleep, and daytime fatigue. This inhibitory shortfall heightens cortical excitability during intended rest periods. In addiction, chronic exposure to substances like alcohol or benzodiazepines induces tolerance through downregulation of GABA_A receptors, particularly α1-containing subtypes, in reward pathways like the ventral tegmental area, necessitating higher doses for effect and increasing vulnerability to dependence and withdrawal symptoms such as anxiety and seizures. This adaptive decrease in receptor density and sensitivity underlies the diminished inhibitory tone.

References

  1. [1]
    Physiology, Neurotransmitters - StatPearls - NCBI Bookshelf
    Neurotransmitters are endogenous chemicals that allow neurons to communicate with each other throughout the body. They enable the brain to provide a variety ...Introduction · Function · Mechanism · Pathophysiology
  2. [2]
    What Defines a Neurotransmitter? - Neuroscience - NCBI Bookshelf
    As briefly described in the preceding chapter, neurotransmitters are chemical signals released from presynaptic nerve terminals into the synaptic cleft.
  3. [3]
    Neurotransmitters—Key Factors in Neurological and ...
    Neurotransmitters are molecules that amplify, transmit, and convert signals in cells, having an essential role in information transmission throughout the ...
  4. [4]
    Neurotransmitter - an overview | ScienceDirect Topics
    Neurotransmitters are chemical substances made by the neuron specifically to transmit a message [1]. Different types of neurotransmitters have been identified.
  5. [5]
    The Biogenic Amines - Neuroscience - NCBI Bookshelf
    There are five established biogenic amine neurotransmitters: the three catecholamines—dopamine, norepinephrine (noradrenaline), and epinephrine (adrenaline)—andMissing: glutamate GABA
  6. [6]
    Two Major Categories of Neurotransmitters - Neuroscience - NCBI
    Small-molecule transmitters can be subdivided into acetylcholine, the amino acids, purines, and biogenic amines.Missing: structures nitric oxide
  7. [7]
    GABA Neurons and the Mechanisms of Network Oscillations - NIH
    GABA neuron-mediated fast synaptic inhibition occurs when an interneuron fires an action potential (or spike) that propagates down its axon and triggers GABA ...
  8. [8]
    Mechanisms of neuromodulatory volume transmission - Nature
    May 24, 2024 · In this review, we focus on the discussion of volume transmission as vesicular transmission that is not restricted to a synaptic contact. A ...
  9. [9]
    molecular insights into origins of neurotransmitters - Journals
    Feb 8, 2021 · This paper summarizes some older and novel insights about evolutionary aspects of neurotransmitter functions. The narrative is not designed to ...Introduction: behaviour is a... · Why are there so many... · Volume transmission in...
  10. [10]
    Neurotransmitter Synthesis - Neuroscience - NCBI Bookshelf - NIH
    The synthesis, packaging, secretion, and removal of neurotransmitters. (A) The life cycle of transmitter agents entails (1) neurotransmitter synthesis, (2) ...Missing: PDF | Show results with:PDF
  11. [11]
    Tyrosine Hydroxylase and Regulation of Dopamine Synthesis - PMC
    Tyrosine hydroxylase is the rate-limiting enzyme of catecholamine biosynthesis; it uses tetrahydrobiopterin and molecular oxygen to convert tyrosine to DOPA.
  12. [12]
    Tryptophan Metabolic Pathways and Brain Serotonergic Activity
    Thus, the rate limiting step in the biosynthesis of 5-HT is the hydroxylation of Trp which is catalyzed by the enzyme tryptophan hydroxylase (TPH) (Figure 1).<|control11|><|separator|>
  13. [13]
    Glutamic Acid Decarboxylase 65: A Link Between GABAergic ...
    Mar 25, 2014 · The rate of GABA synthesis is controlled through the enzyme glutamic acid decarboxylase (GAD), which catalyzes the conversion of glutamic acid ...
  14. [14]
    Choline Acetyltransferase - an overview | ScienceDirect Topics
    Choline acetyltransferase catalyzes the synthesis of acetylcholine from choline and acetyl coenzyme A in cholinergic nerve terminals, a process that depends on ...
  15. [15]
    Vesicular and Plasma Membrane Transporters for Neurotransmitters
    In this article, we focus on the mechanisms responsible for packaging transmitter into synaptic vesicles and for reuptake from the extracellular space that both ...
  16. [16]
    Packaging Neurotransmitters - Neuroscience - NCBI Bookshelf - NIH
    Neuropeptides, in contrast, are packaged into larger synaptic vesicles that range from 90 to 250 nm in diameter. These vesicles are electron-dense in electron ...Missing: peptides | Show results with:peptides
  17. [17]
    Neurotransmitter release is triggered by a calcium-induced ...
    Jun 21, 2024 · The Ca 2+ sensor synaptotagmin-1 triggers neurotransmitter release together with the neuronal SNARE complex formed by syntaxin-1, SNAP25 and synaptobrevin.
  18. [18]
    Bernard Katz, quantal transmitter release and the foundations ... - NIH
    Using the then-new technique of intracellular microelectrode recording, Katz and colleagues recorded the postsynaptic responses – termed the end-plate potential ...
  19. [19]
    Dual-transmitter neurons: Functional implications of co-release and ...
    Here we consider recent examples of co-transmission as well as co-release – the packaging of multiple neurotransmitters into a single vesicle.
  20. [20]
    Neurotransmitter Release and Removal - Neuroscience - NCBI - NIH
    The mechanisms by which neurotransmitters are removed vary but always involve diffusion in combination with reuptake into nerve terminals or surrounding glial ...
  21. [21]
    Physiology, Acetylcholine - StatPearls - NCBI Bookshelf
    Acetylcholine derives from two constituents, choline, and an acetyl group, the latter derived from the coenzyme acetyl-CoA.[2] Choline is naturally present in ...
  22. [22]
    The Impact of Neurotransmitters on the Neurobiology of ...
    Oct 19, 2023 · The signal termination and degradation mechanisms for DA transmission include enzymatic processing by monoamine oxidase (MAO-B), which is ...
  23. [23]
    Allosteric modulation of serotonin and dopamine transporters
    We present here a comparative study of the structural dynamics and ligand-binding properties of two MATs, dopamine transporter (DAT) and serotonin transporter ...
  24. [24]
    Allosteric Modulation of Neurotransmitter Transporters as a ...
    Neurotransmitter transporters (NTTs) are involved in the fine-tuning of brain neurotransmitter homeostasis. As such, they are implicated in a plethora of ...
  25. [25]
    Nitric Oxide: Physiological Functions, Delivery, and Biomedical ...
    Nitric oxide (NO) is a gaseous molecule that has a central role in signaling pathways involved in numerous physiological processes.
  26. [26]
    Glutamate transporters, EAAT1 and EAAT2, are potentially important ...
    Excitatory amino acid transporter (EAAT) 1 and EAAT2 are Na+-dependant glutamate transporters expressed predominantly in glia cells of the central nervous ...
  27. [27]
    Astrocytes Maintain Glutamate Homeostasis in the CNS by ...
    Feb 20, 2019 · Astrocytes contribute to glutamate homeostasis in the CNS, by maintaining the balance between their opposing functions of glutamate uptake and release.
  28. [28]
    Synthesis, Storage and Release of Acetylcholine - NCBI - NIH
    At least half of the choline used in ACh synthesis is thought to come directly from recycling of released ACh, hydrolyzed to choline by cholinesterase.
  29. [29]
    Functional consequences of neuropeptide and small-molecule co ...
    Colocalization of small-molecule and neuropeptide transmitters is common throughout the nervous system of all animals.
  30. [30]
    A fast synaptic potential mediated by NMDA and non-NMDA receptors
    Previous physiological data have suggested the involvement of glutamate receptors in this fast excitatory postsynaptic potential (EPSP), the chemical component ...
  31. [31]
    Dopamine Receptor - an overview | ScienceDirect Topics
    Dopamine receptors belong to a metabotropic G-protein-coupled receptor super-family that is prominent in the vertebrate central nervous system.
  32. [32]
    Agonist binding, agonist affinity and agonist efficacy at G protein ...
    Agonist affinity may be estimated in terms of the dissociation constant for agonist binding to a receptor using ligand binding or functional assays. It has ...
  33. [33]
    Receptor Affinity - an overview | ScienceDirect Topics
    Agonists preferentially bind to high-affinity receptor states, which are functional and capable of activating secondary signaling cascades, while antagonists do ...
  34. [34]
    Neurotransmitter Dynamics - The Dynamic Synapse - NCBI - NIH
    Most excitatory and inhibitory neurotransmitter receptors are concentrated at the postsynaptic density (PSD) facing pre-synaptic terminals containing the ...
  35. [35]
    Presynaptic Receptor - an overview | ScienceDirect Topics
    These receptors may function as autoreceptors, responding to neurotransmitter released from the same terminal to provide negative feedback, or as ...Classification and Molecular... · Mechanisms of Presynaptic...
  36. [36]
    GABA Receptor Positive Allosteric Modulators - StatPearls - NCBI
    Feb 28, 2024 · GABA receptor-positive allosteric modulators, encompassing benzodiazepines and barbiturates, are pivotal in addressing diverse medical conditions.Continuing Education Activity · Indications · Mechanism of Action · Adverse Effects
  37. [37]
    Identification of Neurotransmitter Receptor Genes Under ...
    From this data set, we identified 141 kinds of neuroreceptor ortholog pairs between humans and rodents (mouse or rat (or both)). These orthologs covered almost ...Results · Orthologous Gene Pairs... · Discussion<|control11|><|separator|>
  38. [38]
    Evolution of neuronal types and families - PMC - PubMed Central
    Mar 1, 2019 · Solving nervous system evolution requires cross-species ... Families of related neuronal types are conserved across large evolutionary distances.
  39. [39]
    Two Families of Postsynaptic Receptors - Neuroscience - NCBI - NIH
    The two families are ionotropic receptors, directly linked to ion channels, and metabotropic receptors, which use G-proteins to affect channels.
  40. [40]
    Examples of Neuronal Signal Transduction - Neuroscience - NCBI
    Three important signal transduction pathways can illustrate some of the roles of intracellular signal transduction processes in the nervous system.
  41. [41]
    Neurotransmitter signaling through heterotrimeric G proteins - NCBI
    Neurotransmitters signal via G protein coupled receptors (GPCRs) to modulate activity of neurons and muscles.Introduction · Neurotransmitters and... · The mechanism of signaling...
  42. [42]
    17.11: Signal Transduction - Biology LibreTexts
    May 27, 2022 · Once formed, cAMP binds to and activates protein kinase A (PKA), setting off a phosphorylation cascade that leads to a physiological response.
  43. [43]
    Neurotransmitters and neurotrophins collaborate to influence brain ...
    The material presented indicates that cross- talk between neurotransmitters and neurotrophins may be a mechanism common to the development of multiple neuronal ...
  44. [44]
    Frequency-dependent involvement of NMDA receptors in ... - Nature
    Jul 17, 1986 · Collingridge, G. L., Kehl, S. J. & McLennan, H. J. Physiol., Lond. 334, 19–31 (1983). Article CAS Google Scholar. Coan, E. J. & Collingridge, ...
  45. [45]
    AMPA Receptor Trafficking and Synaptic Plasticity - Annual Reviews
    Mar 1, 2002 · Here we review the growing literature that supports a critical role for AMPA receptor trafficking in LTP and LTD, focusing on the roles proposed for specific ...
  46. [46]
    Role of Dopamine Neurons in Reward and Aversion: A Synaptic ...
    Jun 3, 2015 · In this review, we will discuss how changes in synaptic plasticity of dopamine neurons may affect dopamine release, as well as behavioral ...
  47. [47]
    Dopamine: The Neuromodulator of Long-Term Synaptic Plasticity ...
    Dopamine Triggers Heterosynaptic Plasticity in the Hippocampus and Regulates Cognitive Processes. Together with its extensive role in motor control, reward and ...
  48. [48]
    Homeostatic Synaptic Plasticity: Local and Global Mechanisms for ...
    It is widely agreed that synaptic scaling involves postsynaptic changes in receptor accumulation (Turrigiano 2008), but under some circumstances additional ...
  49. [49]
    Keeping Your Brain in Balance: Homeostatic Regulation of Network ...
    Aug 8, 2024 · Keeping Your Brain in Balance: Homeostatic Regulation of Network Function. Wei Wen1 and Gina G. Turrigiano1. View Affiliations ...
  50. [50]
    Excitotoxicity and N‐methyl‐D‐Aspartate receptors - Olney - 1989
    Not only have endogenous EAA such as glutamate and aspartate become recognized as the leading neurotransmitter candidates at the majority of excitatory synapses ...Missing: excess | Show results with:excess
  51. [51]
    Glutamate, a Neurotoxic Transmitter - John W. Olney, 1989
    Olney JW: Brain lesions, obesity and other disturbances in mice treated with monosodium glutamate. Science 1969;164: 719-721.
  52. [52]
    In Vitro Investigation of Synaptic Plasticity - PMC - NIH
    Here, we focus on two in vitro synaptic plasticity models that rely on acute slices from the rodent brain: field-recording LTP in the hippocampal CA1 region ( ...
  53. [53]
    Discovery of the First Neurotransmitter Receptor: The Acetylcholine ...
    This review retraces the history of the difficulties and successes in the identification of the nicotinic acetylcholine receptor, the first neurotransmitter ...
  54. [54]
    Claude Bernard on the action of curare - PMC - NIH
    “In June 1844 I made my first experiment with curare: I inserted under the skin of the back of a frog a small piece of dry curare, and observed the animal.
  55. [55]
    Otto Loewi – Facts - NobelPrize.org
    In 1921 Otto Loewi stimulated the heart of a frog with electrical impulses and had it pump a small amount of nutrient solution. When the fluid was ...
  56. [56]
    Otto Loewi (1873–1961): Dreamer and Nobel laureate - PMC - NIH
    He named the inhibitory factor 'vagusstoff', which is known today as acetylcholine. Loewi's dream thus led to the discovery that the primary language of nerve ...
  57. [57]
    [PDF] U. S. von Euler - Nobel Lecture
    One of our early findings was that vigorous stimulation of the adrenergic nerves to the spleen did not appreciably lower the noradrenaline content of the organ ...
  58. [58]
    The Discovery of Serotonin and its Role in Neuroscience - Nature
    Aug 1, 1999 · 1948b). However, the chemical structure was still unknown. In the fall of 1948, Maurice Rapport left The Cleveland Clinic for a position at ...
  59. [59]
    EFFECTS OF SODIUM GLUTAMATE ON THE NERVOUS SYSTEM
    Volume 3 (1954) Issue 4; /; Article overview. EFFECTS OF SODIUM GLUTAMATE ON THE NERVOUS SYSTEM. TAKASHI HAYASHI. Author information. TAKASHI HAYASHI.
  60. [60]
    Synaptic chemistry in single neurons: GABA is identified as an ...
    Jun 1, 2005 · Interest in GABA as an inhibitory neurotransmitter was initiated by Bazemore, Elliott, and Florey, who isolated inhibitory substances from brain ...Missing: Erwin | Show results with:Erwin
  61. [61]
    A half century of γ-aminobutyric acid - Sage Journals
    Nov 27, 2019 · The discovery of GABA in the nervous system was first made in ... Florey was sceptical as to whether GABA was actually a neurotransmitter.Missing: Erwin | Show results with:Erwin
  62. [62]
    Nitric oxide is not just blowing in the wind - PMC - PubMed Central
    In 1998, Robert Furchgott, Louis Ignarro and Ferid Murad were awarded the Nobel Prize in Physiology or Medicine for 'for their discoveries concerning nitric ...
  63. [63]
    The Nobel Prize in Physiology or Medicine 1998 - Press release
    When Furchgott and Ignarro presented their conclusions at a conference in ... Nitric oxide protects the heart, stimulates the brain, kills bacteria, etc.Missing: neurotransmitter | Show results with:neurotransmitter
  64. [64]
    Review of recent advances in analytical techniques for the ...
    Techniques most commonly used for the measurement of neurotransmitters include microelectrodes, biosensors, liquid chromatography (LC) and capillary ...
  65. [65]
    Neuropeptide Functions and Regulation - Basic Neurochemistry
    Radioimmunoassay is a second method for neuropeptide detection which provides a quantitative measure of release or content. Sometimes passive immunization has ...
  66. [66]
    An Update of the Classical and Novel Methods Used for Measuring ...
    Detection of neurotransmitters can be done by fluorescence, optical density, electrochemistry or other detection systems more sophisticated. Although the ...
  67. [67]
    Whole-cell patch clamp electrophysiology to study ionotropic ...
    Here we will discuss commonly used protocols and techniques for performing whole-cell patch clamp recordings and exploring AMPA and NMDA receptor mediated ...
  68. [68]
    Studying Synaptic Connectivity and Strength with Optogenetics and ...
    We seek to provide practical insights into the methods used to study neural circuits and synapses, by combining optogenetics and patch-clamp electrophysiology.
  69. [69]
    Genetically encoded fluorescent sensors for imaging neuronal ...
    This review summarizes recent progress in the development and delivery of genetically encoded fluorescent sensors, and highlights example applications in the ...
  70. [70]
    Pushing the frontiers: tools for monitoring neurotransmitters and ...
    Here, we review a range of non-genetically encoded methods (including electrophysiological methods, microdialysis and electrochemical methods) and genetically ...
  71. [71]
    VMAT2 knockout mice: Heterozygotes display reduced ... - NIH
    VMAT2 knockout mice: Heterozygotes display reduced amphetamine-conditioned reward, enhanced amphetamine locomotion, and enhanced MPTP toxicity.
  72. [72]
    Identification of neurotransmitters and co-localization of transmitters ...
    A more recent method for identifying the major transmitter is through the use of in situ hybridization (ISH) for definitive markers of glutamatergic ...
  73. [73]
    Overview of neuropeptides: awakening the senses? - PMC
    Classical small molecule transmitters, such as glutamate, are stored in clear synaptic vesicles, while peptides are stored in dense core vesicles. The ...
  74. [74]
    Protective Role and Functional Engineering of Neuropeptides in ...
    Feb 16, 2023 · There are two types of orexin, orexin-A ... There are three types of endorphins, with beta-endorphins being the most studied and prevalent.
  75. [75]
    Gases as biological messengers: nitric oxide and carbon monoxide ...
    NO and CO have been recognized as putative neurotransmitters. These two novel messenger molecules have greatly expanded the criteria for candidacy of a ...
  76. [76]
    Carbon monoxide and nitric oxide as coneurotransmitters in the ...
    Nitric oxide (NO) and carbon monoxide (CO) seem to be neurotransmitters in the brain. The colocalization of their respective biosynthetic enzymes, neuronal NO ...
  77. [77]
    Box A, Criteria That Define a Neurotransmitter - Neuroscience - NCBI Bookshelf
    ### Criteria for a Substance to Be Considered a Neurotransmitter
  78. [78]
    Neurotransmitter - an overview | ScienceDirect Topics
    Neurotransmitters may be biogenic amines (e.g., acetylcholine, dopamine, norepinephrine, epinephrine, serotonin, histamine), amino acids (e.g., glutamate, GABA) ...
  79. [79]
    Endocannabinoid System: Chemical Characteristics and Biological ...
    Endocannabinoids, unlike classical neurotransmitters, are considered atypical messengers because of the modulation of information from postsynaptic terminals to ...
  80. [80]
    Endocannabinoid signaling and synaptic function - PMC
    Retrograde signaling is the principal mode by which endocannabinoids mediate short- and long-term forms of plasticity at both excitatory and inhibitory synapses ...
  81. [81]
    Overview of Glutamatergic Neurotransmission in the Nervous System
    The authors of the following articles will review the preclinical and clinical evidence for aberrant glutamatergic neurotransmission in neuropsychiatric ...
  82. [82]
    Hippocampal GABAergic Inhibitory Interneurons
    Sep 27, 2017 · In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10–15% of the total neuronal population.
  83. [83]
    Dopaminergic Neurons and Brain Reward Pathways
    Dopamine released from the nigrostriatal pathway modulates corticostriatal transmission in medium spiny neurons expressing dopamine D1 or D2 receptors, which ...
  84. [84]
    Activity of raphé serotonergic neurons controls emotional behaviors
    Thus we find a critical role of serotonergic neuronal activity in emotional regulation and uncover opposing roles of median and dorsal raphe function.
  85. [85]
    Cerebellar Contributions to the Basal Ganglia Influence Motor ...
    Nov 9, 2022 · Both the cerebellum and basal ganglia are implicated in diverse functions, such as motor, cognitive, reward, and emotional processing.
  86. [86]
    Autonomic and endocrine control of cardiovascular function - PMC
    In relation to the cardiovascular system the parasympathetic nervous system has two different kinds of muscarinic receptors: the M2 and M3 receptors (Table 1).
  87. [87]
    Physiology, Noradrenergic Synapse - StatPearls - NCBI Bookshelf
    One major role it is involved in is the body's “fight or flight” response. During states of stress or anxiety, norepinephrine and epinephrine are released and ...
  88. [88]
    Novel aspects of enteric serotonergic signaling in health and brain ...
    Approximately 95% of the body's serotonin ... Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks.
  89. [89]
    Serotonergic Mechanisms Regulating the GI Tract - PubMed Central
    5-HT has been found to modulate enteric nervous system (ENS) development and neurogenesis, motility, secretion, inflammation, sensation, and epithelial ...
  90. [90]
    Role of glutamate on T-cell mediated immunity - PubMed
    Recent studies have shown that glutamate not only has a role as neurotransmitter, but also as an important immunomodulator. In this regard, several glutamate ...
  91. [91]
    Vascular nitric oxide: formation and function - PMC - PubMed Central
    NO plays a versatile role in the vasculature, maintaining vascular tone, inhibiting or reversing platelet activity, and preventing endothelial dysfunction.Missing: neurotransmitter | Show results with:neurotransmitter
  92. [92]
    Substance P and pain chronicity - PMC - PubMed Central
    SP is an 11-amino acid neuropeptide that preferentially activates the neurokinin-1 receptor (NK1R). It transmits nociceptive signals via primary afferent fibers ...
  93. [93]
    Interaction of the Vagus Nerve and Serotonin in the Gut–Brain Axis
    This review synthesizes the current knowledge on serotonin signaling, vagal nerve pathways, and central autonomic regulation.
  94. [94]
    Nicotinic Acetylcholine Receptors and Nicotine Addiction: A Brief ...
    Nicotine binds to neuronal nicotinic acetylcholine receptors (nAChRs), which come in a diverse collection of subtypes. The nAChRs that contain the α4 and β2 ...
  95. [95]
    CPNP 2019 Annual Meeting Poster Abstracts - PMC - PubMed Central
    May 13, 2019 · When administered concomitantly with full opioid agonists, buprenorphine will antagonize the full agonist, resulting in suboptimal analgesia.
  96. [96]
    The behavioral pharmacology and therapeutic potential of lorcaserin ...
    Although 5-HT2A receptor antagonists (e.g., ketanserin, MDL100907), and inverse agonists (e.g., pimavanserin) appear to be more effective at reducing the ...Missing: types | Show results with:types
  97. [97]
    In vivo pharmacological resultant analysis reveals noncompetitive ...
    In vivo, naltrexone, naloxone, CTAP and CTOP appear to be selective μ-opioid receptor antagonists in non-dependent subjects (Gulya et al., 1988; Kramer et al., ...Results · Antagonists Alone · Discussion
  98. [98]
    Ketamine Toxicity - StatPearls - NCBI Bookshelf
    Ketamine's main site action is a non-competitive antagonist of the N-methyl-D-aspartate (NMDA) glutamate receptor, though it exhibits effects on a myriad of ...
  99. [99]
    Beta Blockers - StatPearls - NCBI Bookshelf
    Beta-blockers vary in their specificity towards different receptors, and accordingly, the effects produced depend on the type of receptor(s) blocked as well as ...
  100. [100]
    Selective Beta-1 Blockers - StatPearls - NCBI Bookshelf
    The cardio-selective beta-1-blockers include atenolol, betaxolol, bisoprolol, esmolol, acebutolol, metoprolol, and nebivolol.Continuing Education Activity · Indications · Mechanism of Action · Adverse Effects
  101. [101]
    Molecular determinants of drug–receptor binding kinetics
    molecular size, conformational fluctuations, ...
  102. [102]
    Agonist efficacy and receptor desensitization: from partial truths to a ...
    One risk of long-term exposure of a G protein-coupled receptor (GPCR) agonist is desensitization of the response, most commonly via receptor phosphorylation and ...Missing: side | Show results with:side
  103. [103]
    Agonist-Directed Desensitization of the β2-Adrenergic Receptor - PMC
    Apr 26, 2011 · However, prolonged or repeated use of current β2-agonist drugs leads to loss of their effects, a pervasive phenomenon termed tachyphylaxis, ...Missing: side | Show results with:side
  104. [104]
    Glutamate excitotoxicity: Potential therapeutic target for ischemic ...
    May 24, 2022 · In this review, we highlight recent findings on glutamate release, reuptake mechanisms, NMDAR and its downstream cellular signaling pathways in post-ischemic ...
  105. [105]
    Dopamine and Parkinson's Disease - NCBI - NIH
    Neurons of the substantia nigra communicate with neurons of the basal ganglia by liberating the neurotransmitter dopamine (DA).
  106. [106]
    Serotonin-dopamine interaction and its relevance to schizophrenia
    Conclusions: Serotonergic modulation of dopaminergic function provides a viable mechanism for enhancing therapeutics in schizophrenia, but much remains unclear.
  107. [107]
    Clinical Use of CSF Neurotransmitters - PubMed
    Cerebrospinal fluid pterins and monoamine metabolites are diagnostic in a range of primary neurotransmitter disorders, including disorders of biogenic amine ...
  108. [108]
    PET neurochemical imaging modes - PMC - PubMed Central - NIH
    The most important principle for measuring endogenous neurotransmitter level changes with PET is radioligand sensitivity to competition with the endogenous ...
  109. [109]
    Major Depressive Disorder - StatPearls - NCBI Bookshelf
    It has a lifetime prevalence of about 5 to 17 percent, with the average being 12 percent. The prevalence rate is almost double in women than in men.[5] This ...
  110. [110]
    What has serotonin to do with depression? - PMC - PubMed Central
    Jun 4, 2015 · The best evidence that serotonin plays a role in the pathophysiology of depression comes from studies of “tryptophan depletion”.Missing: prevalence | Show results with:prevalence
  111. [111]
    Microbiota–gut–brain axis and its therapeutic applications ... - Nature
    Feb 16, 2024 · Studies have shown that gut microbiota is capable of producing or stimulating the production of neurotransmitters, including serotonin, dopamine ...
  112. [112]
    Parkinson's Disease: Etiology, Neuropathology, and Pathogenesis
    The movement disorder arises due to the loss of dopaminergic neurons of the substantia nigra pars compacta, with the pathological hallmark being intracellular ...Parkinson's Disease... · Etiology · Lewy Body Pathology
  113. [113]
    Determinants of dopaminergic neuron loss in Parkinson's disease
    The cardinal motor symptoms of Parkinson's disease (PD) are caused by the death of dopaminergic neurons in the substantia nigra pars compacta (SNc).
  114. [114]
    Schizophrenia, Dopamine and the Striatum - PubMed Central - NIH
    Sep 28, 2018 · The mesolimbic hypothesis has been a central dogma of schizophrenia for decades, positing that aberrant functioning of midbrain dopamine ...
  115. [115]
    The Dopamine Hypothesis of Schizophrenia: Version III—The Final ...
    Mar 26, 2009 · The hypothesis that dopamine and dopaminergic mechanisms are central to schizophrenia, and particularly psychosis, has been one of the most ...Missing: hyperactivity | Show results with:hyperactivity
  116. [116]
    Revisiting the role of raphe and serotonin in neuropsychiatric ... - NIH
    The monoaminergic hypothesis of depression posits that this illness results from a deficit in serotonin (5-HT), noradrenaline, and dopamine signaling in the ...
  117. [117]
    Increased Serotonin-1A (5-HT1A) Autoreceptor Expression ... - NIH
    The serotonin-1A (5-HT1A) somatodendritic autoreceptor inhibits the firing of raphe serotonin neurons to negatively regulate the serotonin system (4).
  118. [118]
    Regulation of dorsal raphe nucleus function by serotonin ...
    Serotonin 1B Autoreceptors Originating in the Caudal Dorsal Raphe Nucleus Reduce Expression of Fear and Depression-Like Behavior. Biol Psychiatry. 2011;69 ...5 Influence Of 5-Ht... · 6. Role Of 5-Ht... · 6.3 Stress And 5-Ht...
  119. [119]
    Glutamatergic Mechanisms Associated with Seizures and Epilepsy
    Seizures elevate extracellular glutamate—the main excitatory neurotransmitter of the brain—which leads to aberrant neuronal signaling and connectivity.
  120. [120]
    The Role of Glutamate Receptors in Epilepsy - PMC - PubMed Central
    Glutamate plays a role in both pre- and post-synaptic excitatory neurotransmission resulting in cellular and network hyperactivity, and underlies the formation ...
  121. [121]
    Amyloid β, Glutamate, Excitotoxicity in Alzheimer's Disease
    A rise in glutamate would activate synaptic NMDA receptors, subsequently inducing a desensitization of the receptors with ultimately synaptic depression. A ...Glutamate And Synaptic... · Synaptic Plasticity In... · Glutamate Hypothesis And...
  122. [122]
    Glutamatergic receptor expression changes in the Alzheimer's ...
    This study aimed to characterize the expression of specific glutamate receptor subunits in post‐mortem human brain tissue using immunohistochemistry and ...
  123. [123]
    Autoimmune myasthenia gravis: emerging clinical and biological ...
    In most cases, it is caused by pathogenic autoantibodies directed towards the skeletal muscle acetylcholine receptor (AChR).
  124. [124]
    Mechanisms of acetylcholine receptor loss in myasthenia gravis
    The fundamental abnormality affecting the neuromuscular junctions of myasthenic patients is a reduction of available AChRs, due to an autoimmune attack ...
  125. [125]
    The cholinergic system in the pathophysiology and treatment of ...
    The cholinergic hypothesis of Alzheimer's disease centres on the progressive loss of limbic and neocortical cholinergic innervation. Neurofibrillary ...
  126. [126]
    Cholinergic system during the progression of Alzheimer's disease
    Alzheimer's disease (AD) is characterized by a progressive phenotypic downregulation of markers within cholinergic basal forebrain (CBF) neurons.
  127. [127]
    The Role of GABA in Primary Insomnia - PMC - PubMed Central
    GABA, the primary inhibitory neurotransmitter in the central nervous system (CNS), plays a role in the etiology of chronic insomnia.
  128. [128]
    GABA mechanisms and sleep - PubMed
    It has been shown that GABA(B) receptor antagonists increase brain-activated behavioral states (waking and paradoxical sleep: dreaming stage). Recently, a ...
  129. [129]
    The Role of GABAA Receptors in the Development of Alcoholism
    GABA A receptors undergo allosteric modulation by ethanol, anesthetics, benzodiazepines and neurosteroids and have been implicated in the acute as well as the ...
  130. [130]
    The role of GABAA receptors in the acute and chronic effects of ...
    The effects of chronic ethanol administration are influenced by adaptations in GABA A receptor function, expression, trafficking, and subcellular localization.<|control11|><|separator|>