Fact-checked by Grok 2 weeks ago

Vesicular monoamine transporter

The vesicular monoamine transporter (VMAT), also known as SLC18, is a family of membrane proteins that actively transport monoamine neurotransmitters—including , serotonin, norepinephrine, and —from the neuronal into synaptic vesicles, enabling their storage and regulated release during . These transporters function as proton antiporters, coupling the influx of one monoamine molecule with the efflux of two protons, powered by an generated by the vesicular H⁺-. VMATs are essential for maintaining neurotransmitter , protecting cytosolic monoamines from oxidative degradation, and preventing toxicity in monoaminergic neurons. Two main isoforms exist: VMAT1 (SLC18A1), primarily expressed in peripheral neuroendocrine cells such as adrenal chromaffin and enterochromaffin cells, and VMAT2 (SLC18A2), the predominant form in neurons, sympathetic neurons, and mast cells. VMAT2 exhibits higher substrate affinity and uniquely transports , distinguishing it functionally from VMAT1, which is more selective for catecholamines in non-neuronal tissues. Structurally, both isoforms are integral membrane glycoproteins of approximately 70 kDa, featuring 12 transmembrane α-helices organized into N- and C-terminal bundles that undergo conformational changes between outward- and inward-facing states to facilitate transport. Recent cryo-electron studies have resolved the VMAT2 in multiple states, revealing key binding sites for substrates like in a central and conserved aspartate residues (e.g., Asp399, Asp426) critical for proton coupling. VMATs play a pivotal role in physiological processes, including mood regulation, motor control, cognition, and immune responses, by sequestering monoamines away from degradative enzymes and enabling quantal release at synapses. Dysregulation or inhibition of VMATs is implicated in neurological and psychiatric disorders; for instance, VMAT2 deficiencies contribute to through impaired dopamine packaging, while selective VMAT2 inhibitors like are used therapeutically for hyperkinetic such as Huntington's . Pharmacological modulation of VMATs also underlies the actions of drugs like , which depletes monoamine stores and models , highlighting their therapeutic potential and risks in treating conditions involving monoamine imbalance.

Overview

Definition and physiological role

Vesicular monoamine transporters (VMATs) are integral membrane proteins belonging to the solute carrier family 18 (SLC18) that actively sequester cytoplasmic monoamines—including dopamine, norepinephrine, serotonin, and histamine—from the cytosol into synaptic or secretory vesicles. This process occurs in monoaminergic neurons and neuroendocrine cells, where VMATs function as antiporters, exchanging vesicular protons for the positively charged monoamine substrates. The transport is driven by the proton motive force (\Delta \mu_{H^+}), an electrochemical gradient generated by the vacuolar-type H⁺-ATPase (V-ATPase) that acidifies the vesicle interior. Specifically, the transport rate depends on the pH gradient (\DeltapH) and membrane potential (\Delta \psi) components of \Delta \mu_{H^+}, with VMATs typically exchanging two protons for one monoamine molecule to achieve accumulation ratios exceeding 10,000-fold against the concentration gradient. Physiologically, VMATs play a critical role in protecting monoamines from enzymatic degradation in the by (MAO), which would otherwise oxidize them and generate leading to cellular toxicity. By packaging these neurotransmitters into vesicles, VMATs enable their quantal release via upon neuronal stimulation, ensuring precise and regulated delivery into the synaptic cleft for effective . Furthermore, VMAT activity contributes to maintaining intravesicular pH and osmotic balance, preventing vesicle swelling, while modulating cytosolic monoamine concentrations to avoid toxicity or inhibitory feedback on biosynthetic enzymes like . Disruption of VMAT function impairs monoaminergic signaling and is implicated in neurological and psychiatric disorders, including , , and substance use disorders, where altered vesicular storage affects dynamics and neuronal vulnerability. VMAT2 serves as the primary isoform in the and peripheral sympathetic neurons, supporting in , noradrenergic, and neurons, whereas VMAT1 predominates in endocrine tissues such as the .

Types of VMATs

There are two primary isoforms of the vesicular monoamine transporter (VMAT), known as VMAT1 and VMAT2, which are encoded by distinct genes and exhibit differences in tissue expression, substrate affinity, and pharmacological sensitivity. These isoforms share a common role in monoamine sequestration but are adapted to specific physiological contexts, with VMAT1 predominantly functioning in peripheral tissues and VMAT2 in the . VMAT1, encoded by the SLC18A1 gene located on chromosome 8p21.3, is primarily expressed in peripheral neuroendocrine cells, including adrenal chromaffin cells and enterochromaffin cells of the gastrointestinal tract. It exhibits lower affinity for monoamine substrates such as catecholamines compared to VMAT2, making it suitable for environments with higher substrate concentrations in the periphery, though it shows poor transport efficiency for . The protein consists of 12 transmembrane domains typical of the , contributing to its role in vesicular packaging of monoamines in these non-neuronal endocrine settings. In contrast, VMAT2, encoded by the SLC18A2 gene on chromosome 10q25.3, is the predominant isoform in the , where it is expressed in , , and noradrenergic neurons, as well as in histamine-containing cells such as . VMAT2 demonstrates higher affinity for key monoamine neurotransmitters, including , serotonin, and norepinephrine, facilitating efficient uptake in low-concentration synaptic environments; it also transports with moderate efficiency, supporting storage in mast cell granules. Like VMAT1, it features 12 conserved transmembrane domains but is distinguished by enhanced sensitivity to inhibitors such as , which binds with higher affinity to VMAT2. The two isoforms share approximately 62% amino acid sequence identity in humans, with high conservation in the core transmembrane regions that form the pathway, but notable divergence in the N- and C-terminal tails. These terminal differences influence protein trafficking, vesicular targeting, and regulatory interactions, contributing to their tissue-specific expression and functional specialization. Functionally, VMAT2's greater sensitivity to inhibition—compared to VMAT1—highlights its distinct pharmacological profile, while both isoforms maintain antiport activity driven by proton gradients, though VMAT2's higher substrate affinity underscores its critical role in central .

Substrates and specificity

Monoamine neurotransmitters

The primary substrates of vesicular monoamine transporters (VMATs) are monoamine neurotransmitters, including , norepinephrine, epinephrine, , and , which are essential for synaptic transmission in the central and peripheral nervous systems. These molecules are synthesized in the neuronal and rapidly sequestered into synaptic vesicles by VMATs to protect them from degradation by cytosolic enzymes such as (MAO). Monoamines are chemically classified based on their core structures: catecholamines, which include , norepinephrine, and epinephrine, derived from and featuring a ring; indolamines, represented by serotonin; and imidazoleamines, such as . At physiological , all these monoamines exist predominantly in their protonated (cationic) forms due to the basic nature of their groups, which is critical for high-affinity recognition and binding by VMATs during transport. Biosynthesis of these monoamines occurs via enzymatic pathways in the . is produced from through two sequential steps: by (TH), the rate-limiting enzyme, to form , followed by by (AADC). is synthesized from by dopamine β-hydroxylase (DBH), which requires ascorbic acid as a cofactor and occurs within vesicles in some cell types. Epinephrine is then formed from norepinephrine by phenylethanolamine N-methyltransferase (PNMT), primarily in adrenal chromaffin cells. is derived from via by (TPH), the rate-limiting step, and subsequent by AADC. is generated from through by (HDC). VMAT-mediated sequestration into vesicles not only concentrates these neurotransmitters for release but also shields them from MAO-mediated oxidative degradation in the . In addition to these core monoamines, VMATs can transport trace amines such as phenylethylamine () and , with affinities comparable to those of the primary catecholamines such as and norepinephrine.

Substrate recognition and selectivity

The vesicular monoamine transporters (VMATs) recognize their substrates through specific interactions between the protonated group of monoamines and conserved aspartate residues in the transmembrane helices, such as Asp33 in 1 (TM1) and Asp399 in TM10 of VMAT2. These aspartates form salt bridges or hydrogen bonds with the positively charged nitrogen, facilitating initial binding in a cytoplasmic-facing conformation. Additionally, hydrophobic pockets lined by aromatic residues, including Tyr341 in TM8 and Tyr433 in TM11, accommodate the or rings of substrates via π–π stacking interactions, contributing to the overall specificity for biogenic amines. VMAT2 exhibits higher affinity for serotonin compared to dopamine, norepinephrine, and , with Km values of approximately 0.3 μM for serotonin and 1 μM for , while norepinephrine shows similar affinity to and has lower affinity around 40 μM (measured as ). In contrast, VMAT1 displays reduced selectivity for , with an exceeding 300 μM and no effective transport, whereas its Km for serotonin is about 0.5 μM, highlighting isoform-specific differences driven by variations in transmembrane residues like Tyr434 and Asp461. These rankings underscore VMAT2's broader role in monoamine handling and VMAT1's specialization in peripheral tissues. Selectivity is modulated by the acidic vesicular (optimal around 5.5–6.0), which protonates substrates and aspartate residues like Asp33 and Glu313, inducing allosteric conformational changes that favor binding and prevent simultaneous proton and occupancy. among substrates occurs at shared binding sites, with higher-affinity monoamines like serotonin outcompeting others at low concentrations, while proton gradients enhance overall efficiency. VMATs also weakly transport non-monoamine substrates such as the MPP+, with a Km of approximately 9 μM for VMAT2, allowing sequestration into vesicles and potential protection against , though at much lower efficiency than native monoamines.

History

Discovery and early studies

The discovery of the vesicular monoamine transporter (VMAT) emerged from investigations into the mechanism of , an known to deplete monoamine stores in tissues. In the mid-1950s, and Nils-Åke Hillarp demonstrated that administration caused a profound depletion of catecholamines, including adrenaline, from the in rabbits, indicating that monoamines were stored in a releasable, vesicular form rather than freely diffusible within cells. This observation suggested the involvement of a carrier-mediated uptake process into storage organelles, as did not affect cytoplasmic monoamine levels but specifically targeted vesicular pools. By the early 1960s, studies provided direct evidence for ATP-dependent monoamine uptake into vesicular fractions. Norman Kirshner isolated a particulate fraction from bovine that exhibited stimulated uptake of catecholamines in the presence of Mg²⁺ and ATP, with inhibiting this process by over 90% at micromolar concentrations, confirming the specificity of the transport mechanism. Extending these findings to the , 1970s experiments using subcellular preparations from and adrenal tissue demonstrated -sensitive, ATP-driven accumulation of tritium-labeled into synaptic vesicles, establishing VMAT activity as a conserved process across peripheral and central monoaminergic systems. Subcellular fractionation techniques further localized VMAT to vesicles during this period. Tomas Hökfelt employed and histochemical methods to visualize monoamine storage in small granular vesicles within peripheral and central neurons, supporting their role as primary storage sites. Similarly, Floyd Bloom used to confirm noradrenaline localization in autonomic nerve ending vesicles, reinforcing the vesicular compartmentalization of monoamines. These efforts laid the groundwork for later purification attempts in the early , where partial isolation of the reserpine-binding protein from bovine chromaffin granules marked the onset of molecular characterization. A key mechanistic insight came in 1980, when Linda Toll and B. Dixon Howard showed that VMAT-mediated of epinephrine into chromaffin granules relied on a generated by a vesicular Mg²⁺-ATPase, rather than direct by the transporter itself; dissipating the gradient with proton ionophores abolished uptake, while blocked it independently. This proton antiport model, initially proposed for catecholamines, highlighted the energy coupling essential for vesicular loading and provided a foundational framework for understanding VMAT function.

Key milestones and researchers

The of the vesicular monoamine transporter genes marked a pivotal advancement in understanding monoamine storage mechanisms. In 1992, two independent groups isolated cDNAs encoding VMAT2: Liu et al. used expression in PC12 cells derived from to identify a reserpine-sensitive transporter from tissue, while Erickson et al. employed a similar expression strategy in CV-1 fibroblasts to the gene from a library. Shortly thereafter, in 1994, Gasnier et al. VMAT1 from bovine adrenal chromaffin granules using degenerate based on conserved sequences, revealing the first isoform primarily associated with peripheral endocrine tissues. Functional validation through genetic models further elucidated VMAT2's essential role in monoamine . In the late 1990s, Fon et al. generated VMAT2 mice via targeted disruption, demonstrating that homozygous deficiency leads to profound depletion of catecholamines and serotonin, resulting in neonatal within hours of birth due to and autonomic dysfunction. Heterozygous mice survived but exhibited reduced vesicular storage capacity, highlighting VMAT2's dosage-sensitive function in neuronal signaling. Key researchers advanced insights into substrate selectivity and transporter regulation during this period. Randy Blakely and Gary Rudnick contributed seminal studies on VMAT's interaction with monoamines and , elucidating how structural motifs influence recognition and proton-coupled efficiency through biochemical assays and . Their work, including analyses of and isoform differences, established foundational models for VMAT's specificity toward catecholamines versus . In the 2000s, isoform-specific expression mapping refined the understanding of VMAT distribution across tissues. Weihe and Eiden's comprehensive neuroanatomical surveys using and demonstrated VMAT1's predominance in peripheral endocrine cells, such as adrenal chromaffin cells, while VMAT2 was mapped to central monoaminergic neurons, including in the and serotonergic , underscoring their complementary roles in systemic versus neural monoamine handling. Post-2010s structural investigations provided mechanistic depth to VMAT . A 2013 study by Yaffe et al. identified critical hinge points in VMAT2's alternating access mechanism using accessibility mapping, revealing how proton gradients drive conformational changes for substrate translocation. The brought high-resolution cryo-EM structures that resolved VMAT binding sites. In 2024, Schapira et al. reported a 3.1 structure of human VMAT2 bound to , illuminating the inhibitor's occlusion in the central and substrate entry pathways. Subsequent 2024 structures by Ma et al. captured human VMAT2 in multiple conformations with monoamine substrates including and serotonin, detailing the binding pocket's residues for selectivity and enabling targeted for neurological disorders.

Molecular structure

Overall topology and domains

Vesicular monoamine transporters (VMATs), members of the 18 (SLC18), exhibit a canonical major facilitator superfamily (MFS) fold characterized by 12 transmembrane-spanning α-helices (TM1–TM12). These helices are organized into two pseudosymmetrical bundles: an N-terminal (NTD) comprising TM1–TM6 and a C-terminal (CTD) comprising TM7–TM12, which facilitate the rocker-switch mechanism for alternating access between cytosolic and luminal sides. The N- and C-termini are both oriented toward the , consistent with the inward-facing topology in physiological vesicular membranes, while a large extracellular loop (ECL1) between TM1 and TM2 extends into the vesicle lumen, often featuring sites that contribute to protein stability. The core architecture includes a central cavity formed primarily by TM6–TM8, which serves as the site for coordinated proton and exchange in the cycle. Conserved acidic residues, such as Asp399 and Glu312, are critical for proton binding and are preserved across the SLC18 family to support the electrochemical gradient-driven transport. This domain organization underscores the structural conservation within MFS transporters, enabling VMATs to couple vesicular acidification to monoamine sequestration. VMAT1 and VMAT2 isoforms display high sequence conservation in their core transmembrane helices, with approximately 62% identity, reflecting shared functional architecture despite tissue-specific expression. Differences arise primarily in peripheral regions, including N-terminal sites in the TM1–TM2 loop of VMAT1 and C-terminal dileucine motifs (e.g., or sequences) in VMAT2 that mediate endocytic trafficking. Evidence from biochemical and cryo-EM studies suggests VMATs can assemble into dimers within lipid membranes, though the functional implications of this oligomerization remain unclear and may be context-dependent.

Binding sites and ligand interactions

The substrate binding site in the vesicular monoamine transporter 2 (VMAT2), often referred to as the reserpine-sensitive site, is situated within the central hydrophilic cavity of the protein, primarily involving residues from transmembrane helices (TMs) TM1, TM4, TM5, TM7, TM8, TM10, and TM11. Cryo-electron microscopy (cryo-EM) structures at resolutions of 3.4–3.6 Å reveal that the protonated group of substrates like or is coordinated by negatively charged residues such as Asp399, which forms a , and Tyr341, facilitating recognition of the positively charged moiety. Aromatic residues, including Phe334 and Tyr341, engage in π-π stacking interactions with the substrate's or ring, contributing to specificity for monoamine neurotransmitters. These interactions position the substrate's hydroxyl group toward Glu312, enabling proton-coupled transport in the lumen-facing conformation. Inhibitor binding sites differ slightly from the substrate site, with tetrabenazine (TBZ) occupying a central pocket in the outward-open or occluded conformation, lined by TMs TM1, TM4, TM5, TM7, TM9, TM11, and TM12. At 3.1 Å resolution, the TBZ structure shows its tertiary amine interacting with Glu312 and Lys138, while hydrophobic moieties contact aromatic residues like Trp318 and Phe429, stabilizing a conformation that prevents substrate access and transporter cycling. Reserpine, in contrast, binds the lumen-facing site in a cytosol-accessible manner, engaging Asp399, Glu312, Tyr341, and Val232, as captured in a 3.7 Å cryo-EM structure using a Y422C mutant to trap the state; this high-affinity interaction effectively locks the transporter without forming a covalent adduct, though the slow dissociation rate mimics irreversibility. Recent structural studies from 2023–2025, including those by Wang et al. and others, have elucidated these sites across multiple conformational states (inward-open, outward-open, occluded, and lumen-facing) at resolutions of 3.1–3.7 Å, highlighting ligand-induced gating mechanisms where or triggers rearrangements, such as in TM2 and TM7 for TBZ. Additional 2025 cryo-EM structures at 3.14–3.31 Å resolutions capture VMAT2 in cytoplasmic-open states bound to and lumenal-facing states with serotonin (interacting with Asp399, Asn305, Tyr341), (with Glu312, Tyr433), and (with Asp399, Asn305 but not Glu312), revealing amphetamine's role in promoting monoamine exchange and release rather than direct gradient dissipation. Affinities reflect pharmacological potency, with exhibiting a Ki of approximately 1–2 nM and TBZ a Ki of ~100 nM for VMAT2. Selectivity between VMAT isoforms arises from steric hindrance in VMAT1's , where bulkier residues (e.g., corresponding to Phe429 in VMAT2) reduce TBZ affinity by over 100-fold compared to VMAT2.

Function and mechanism

Transport process

The vesicular monoamine transporter (VMAT) facilitates the uptake of monoamines from the cytosol into the acidic lumen of synaptic vesicles through an alternating access mechanism, a conserved rocker-switch model common to major facilitator superfamily transporters. In this process, VMAT alternates between a cytosol-open (inward-facing) conformation, where the central binding site is accessible from the cytoplasmic side, and a lumen-open (outward-facing) conformation, exposing the site to the vesicular interior. Recent cryo-electron microscopy structures have resolved human VMAT2 in outward-open, occluded, and inward-open states, revealing a central substrate-binding cavity and key interactions driving the cycle. The antiport mechanism exchanges two protons outward for each monoamine molecule inward, driven by the proton motive force established by . In the outward-facing conformation, of key residues such as Asp399 (in transmembrane 10) and Asp426 (in 11) at the low luminal (~5.5–6.0) occurs, with Asp399 disrupting interactions (e.g., with dopamine's ) to promote release into the and trigger the switch to the inward-facing state. Protons are then released to the ( ~7.2), facilitating rebinding of cytosolic monoamines (protonated form) to the now-accessible site via electrostatic interactions. A subsequent conformational change returns the transporter to the outward-facing state, releasing the monoamine into the acidic while preventing reverse due to pH-sensitive proton-binding sites involving conserved aspartates and tyrosines. The overall efficiency of VMAT-mediated transport relies on a stoichiometry of one monoamine exchanged for two protons per transport cycle, ensuring vectorial uptake against a steep (up to 10,000-fold accumulation). This coupling maintains directionality, as the sensitivity of proton-binding sites—such as those involving conserved tyrosines and aspartates—prevents reverse transport under physiological conditions, with low luminal favoring substrate trapping and high cytosolic promoting rebinding of protons for cycle continuation. The proton motive force comprises a (ΔpH ≈ 1–1.5 units) and a (Δψ ≈ 60–80 mV, lumen positive).

Kinetics and energy coupling

The vesicular monoamine transporters (VMATs), particularly VMAT2, exhibit Michaelis-Menten kinetics in their substrate uptake, characterized by saturable transport rates that follow a hyperbolic relationship between substrate concentration and velocity. For uptake by VMAT2 in synaptic vesicles, representative kinetic parameters include a K_m of approximately 0.3–0.5 μM and a V_{\max} ranging from 35 to 114 pmol/min/mg protein, depending on the vesicular preparation and assay conditions. These values indicate high-affinity binding for , with transport efficiency optimized for physiological cytoplasmic concentrations. The Hill coefficient is approximately 1, consistent with non-cooperative, single-site binding and by excess substrate or structurally similar monoamines. VMAT-mediated transport is tightly coupled to the proton (\Delta \mu H^+) established by vesicular , which drives antiport of monoamines into the vesicle in for protons. This energy dependence is evident from the dissipation of the proton gradient during active uptake, quantifiable through of acridine orange , a pH-sensitive that reports on intravesicular acidification. Protonophores such as FCCP abolish transport by collapsing the \Delta \mathrm{pH} and \Delta \psi components of the gradient, confirming the essential role of \Delta \mu H^+ without direct effects on the transporter protein itself. Transport activity is modulated by environmental factors, with optimal rates observed between 25°C and 37°C, reflecting physiological relevance in neuronal environments. The vesicular interior maintains an acidic of approximately 5.5, which is critical for substrate and efficient antiport; deviations from this pH impair uptake velocity. The temperature coefficient Q_{10} for VMAT2-mediated dopamine transport is around 2, indicating moderate temperature sensitivity typical of membrane-bound carrier proteins, where rates roughly double with a 10°C increase in the physiological range.

Cellular localization and expression

Tissue and subcellular distribution

Vesicular monoamine transporters (VMATs) exist in two main isoforms, VMAT1 and VMAT2, which exhibit distinct tissue distributions reflecting their roles in peripheral and central monoamine handling, respectively. VMAT2 is predominantly expressed in the , particularly in monoaminergic neurons of the (dopaminergic), raphe nuclei (), and (noradrenergic), as well as in peripheral sites such as and mast cells. In contrast, VMAT1 shows high expression in peripheral neuroendocrine tissues, including the adrenal medulla's chromaffin cells, enterochromaffin cells of the , and renal proximal tubules, with notably lower levels in the . At the subcellular level, both isoforms localize exclusively to intracellular vesicles, with no detectable presence on the plasma membrane. VMAT2 is primarily associated with synaptic vesicles in central and peripheral neurons, facilitating monoamine storage for synaptic release, while in endocrine and immune cells like mast cells, it resides in dense-core secretory vesicles. VMAT1, similarly confined to vesicular compartments, is enriched in large dense-core vesicles of neuroendocrine cells, such as those in the and enterochromaffin cells, supporting bulk monoamine secretion. Immunoelectron microscopy studies have confirmed this vesicular localization for VMAT2 in secretory granules of monoaminergic neurons and peripheral cells, underscoring its role in compartmentalized storage rather than surface transport. Regional variations in VMAT2 expression within the further highlight its impact on monoamine dynamics, particularly . VMAT2 density is markedly higher in the compared to the —often by over 100-fold—enabling efficient packaging in nigrostriatal terminals, as evidenced by recent quantitative studies. A analysis of VMAT expression patterns reinforced these gradients, showing elevated VMAT2 in striatal dopaminergic terminals versus cortical regions, which influences regional homeostasis and release . These distribution differences between isoforms and regions ensure specialized monoamine sequestration tailored to neuronal versus endocrine functions.

Genetic and regulatory control

The vesicular monoamine transporters are encoded by two distinct genes: SLC18A1, which codes for VMAT1 and spans approximately 38 kb with 17 exons, and SLC18A2, which encodes VMAT2 and consists of 16 exons across about 38 kb on chromosome 10q25.3. The promoters of both genes contain binding sites for transcription factors such as Sp1 and CREB, which mediate basal transcription and activity-dependent regulation, respectively; for instance, CREB phosphorylation enhances VMAT2 promoter activity in response to stimuli like gastrin in enterochromaffin cells. VMAT gene expression is dynamically regulated by environmental and pharmacological factors. Chronic exposure to upregulates SLC18A2 (VMAT2) mRNA in neurons through feedback mechanisms involving the (DAT), leading to increased vesicular storage capacity as an adaptive response to elevated cytosolic monoamines. In contrast, VMAT2 expression is downregulated in preclinical models of , such as those with progressive VMAT2 deficiency, contributing to impaired packaging and heightened in nigrostriatal terminals. Sex-specific differences also influence expression; for example, modulates VMAT2 levels in the , with females exhibiting higher VMAT2 activity and protection against toxin-induced loss compared to males. Epigenetic modifications further fine-tune VMAT expression, particularly in contexts of substance use. In addiction models, such as chronic exposure, increased at the SLC18A2 promoter enhances transcriptional activity in reward-related brain regions like the , promoting VMAT2 upregulation and monoamine . Genetic polymorphisms in SLC18A2 contribute to inter-individual variability in expression; the rs363224 , for instance, is an intronic variant associated with altered VMAT2 expression, where the low-expression AA genotype correlates with reduced VMAT2 levels and protection against . Developmentally, VMAT2 expression in monoaminergic neurons follows a temporal , with low levels during embryogenesis and a postnatal peak that coincides with synaptic maturation and neurotransmitter system refinement in regions like the and . This surge supports the establishment of vesicular storage capacity essential for adult monoamine signaling.

Physiological regulation

Trafficking and vesicular cycle

Vesicular monoamine transporters (VMAT1 and VMAT2) are synthesized on ribosomes and inserted into the () membrane as polytopic proteins, where they undergo initial folding and N-glycosylation before trafficking to the Golgi apparatus for further processing and sorting. In the trans-Golgi network (TGN), VMATs are selectively packaged into constitutive secretory vesicles for delivery to synaptic vesicles (SVs) or into the regulated secretory pathway for incorporation into immature large dense-core vesicles (LDCVs) in monoaminergic neurons. Once sorted, VMAT-containing vesicles are transported axonally along to presynaptic terminals, ensuring targeted localization in distal neurites. A dileucine-like motif in the C-terminal cytoplasmic tail of both VMAT1 and VMAT2 binds the adaptor protein complex AP-2, facilitating clathrin-mediated endocytosis and retrieval from the plasma membrane during the vesicular lifecycle. This motif is essential for efficient in both PC12 cells and neurons, preventing excessive surface exposure and supporting rapid recycling. Following biosynthesis, newly synthesized VMATs traffic to immature secretory vesicles at the TGN, where they integrate into forming LDCVs or SV precursors via signals in their N-glycosylated lumenal loop and C-terminal domains. Vesicular maturation involves acidification by the V-ATPase proton pump, which not only enables monoamine uptake by VMATs but also drives the remodeling of immature vesicles through removal of mannose-6-phosphate receptors and other proteins via AP-1- and PACS-1-dependent budding. An acidic cluster in the VMAT2 C-terminus, when phosphorylated by casein kinase II (CKII), retains the transporter in maturing LDCVs by promoting interactions that resist removal during this process; dephosphorylation shifts VMAT2 toward constitutive pathways and small SVs. Similar PKA-mediated regulation influences trafficking of both VMAT1 and VMAT2 through interactions with sorting complexes. Post-exocytosis, VMATs recycle from the plasma membrane through clathrin/AP-2-mediated endocytosis into early endosomes, from which they are resorted into newly forming SVs or LDCVs at the nerve terminal, maintaining vesicular pools for sustained neurotransmitter release. Phosphorylation events regulate VMAT trafficking dynamics; for instance, CKII-mediated phosphorylation of the C-terminal serines enhances retention in LDCVs and overall vesicular incorporation, while protein kinase A (PKA) influences sorting complexes to modulate surface-to-vesicle distribution for both isoforms. Synaptic activity, such as depolarization-induced exocytosis, increases VMAT2 shuttling from surface pools to recycling endosomes and nascent vesicles, potentially via co-trafficking with glutamate transporters like VGLUT2, thereby amplifying monoamine packaging capacity during heightened neurotransmission. As of 2024, studies have shown that tricyclic and tetracyclic antidepressants upregulate VMAT2 activity by promoting its trafficking to synaptic vesicles and increasing protein expression, potentially enhancing monoamine storage in response to pharmacological modulation. Under cellular stress conditions, such as exposure, VMAT2 undergoes ubiquitination, marking it for lysosomal degradation via the endolysosomal pathway, which reduces transporter levels and impairs monoamine storage; this contrasts with basal proteasomal degradation and highlights stress-specific quality control mechanisms.

Post-translational modifications

(VMAT2) is subject to N-linked primarily at multiple sites within the large intraluminal between transmembrane domains 1 and 2. These modifications are crucial for proper , exit from the (), and maturation into the fully glycosylated form observed at approximately 120 kDa, as opposed to the immature 75 kDa species retained in the . facilitates trafficking to appropriate vesicular compartments, such as large dense-core vesicles, and supports overall transport function; inhibition of glycosylation or mutation of these sites leads to mislocalization and reduced monoamine uptake activity by up to 50%. VMAT1 undergoes analogous N- in its luminal , essential for its stability and targeting in peripheral neuroendocrine cells. Phosphorylation occurs at serine residues 512 and 514 in the C-terminal of VMAT2, mediated by casein kinase II (CKII), with potential involvement of casein kinase I (CKI). This enhances the maximum velocity (Vmax) of serotonin uptake by about 40%, thereby increasing transport efficiency and promoting association with large dense-core vesicles rather than small synaptic vesicles. Phospho-mimetic mutations (serine to ) further elevate uptake rates, while non-phosphorylatable substitutions decrease activity and alter vesicular targeting. Although (PKA) does not directly phosphorylate VMAT2, it regulates trafficking through interactions with associated proteins like syntaxin 1A, indirectly influencing localization and function; PKA similarly modulates VMAT1 trafficking. N-terminal sites may also modulate VMAT2 activity, with phospho-mimetic changes reducing methamphetamine-stimulated release and potentially mimicking aspects of amphetamine-induced by altering vesicular dynamics. Palmitoylation on specific cysteine residues contributes to VMAT2 membrane stability and integration into lipid rafts, though detailed sites and mechanisms remain less characterized compared to other modifications. residues, including those forming bonds (e.g., Cys117-Cys324), support structural integrity essential for function, but S-palmitoylation's role appears auxiliary for rather than direct of . Ubiquitination of VMAT2, predominantly K48-linked polyubiquitination, is not prominent under basal conditions but occurs in pathways, targeting for proteasomal breakdown to regulate and prevent accumulation of misfolded forms. This process is evident following proteasomal inhibition, where ubiquitinated VMAT2 accumulates, leading to and reduced stability; lysosomal plays a minimal role basally.

Pharmacology and inhibition

Inhibitor classes and mechanisms

Vesicular monoamine transporter (VMAT) inhibitors are classified primarily into reserpinoids, tetrabenazine-like compounds, and lobeline analogs, each distinguished by their structural features and interactions with VMAT isoforms, particularly VMAT2. Reserpinoids, derived from the , act as irreversible inhibitors by covalently to a lumenal-facing site within the transporter's central cavity, effectively trapping VMAT in a substrate-bound conformation that prevents further monoamine uptake. This mechanism depletes vesicular stores over time due to the irreversible of the inhibition, contrasting with reversible classes. Tetrabenazine-like inhibitors, including (TBZ) and its derivatives such as and , represent a class of reversible, non-competitive inhibitors that bind to a distinct cytoplasmic-facing site, stabilizing VMAT2 in an occluded or outward-open conformation. This binding, involving key residues like Phe135 and Tyr433 through π-stacking interactions, hinders the conformational switch necessary for translocation, thereby blocking monoamine packaging without directly competing at the site. TBZ exhibits high selectivity for VMAT2 over VMAT1, due to specific hydrophobic interactions at residues like Val232. In contrast, reserpinoids like are non-selective, inhibiting both VMAT1 and VMAT2 with similar affinities (Ki ≈ 170 nM for VMAT2), leading to broader monoamine depletion across neuronal and non-neuronal tissues. Lobeline analogs, such as lobelane and GZ-793A, function as partial agonists or antagonists at VMAT2, interacting primarily at the substrate-binding site to modulate monoamine release in a concentration-dependent manner. These compounds inhibit VMAT2 function non-competitively by reducing the cytosolic dopamine available for reverse transport, with lobeline showing moderate affinity (IC50 ≈ 0.9 μM) and potential for decreasing methamphetamine-evoked dopamine efflux. Competitive inhibition is exemplified by amphetamine-like substrates such as fenfluramine, which bind to the central substrate site and displace endogenous monoamines, acting as alternative substrates that promote their efflux from vesicles. Additionally, some inhibitors, including certain amphetamine derivatives, exert allosteric effects by modulating proton binding sites, disrupting the proton gradient essential for VMAT's antiport mechanism without directly occluding the substrate pathway. The therapeutic potential of these inhibitors varies with dosage and selectivity; low doses of VMAT2-selective agents like TBZ provide targeted depletion for treating hyperkinetic disorders such as associated with , while high doses of non-selective reserpinoids cause extensive monoamine depletion, potentially inducing depressive symptoms through profound loss.

Specific pharmacological agents

, an derived from the plant Rauwolfia serpentina, acts as an irreversible of VMAT2 by to its following proton translocation from the vesicular lumen, thereby preventing monoamine uptake into synaptic vesicles and leading to their cytosolic depletion through enzymatic degradation. This depletion of such as , norepinephrine, and serotonin underlies reserpine's historical use as an antihypertensive agent in the mid-20th century, where it effectively lowered by reducing sympathetic tone. However, reserpine's clinical application declined due to its association with severe depressive symptoms, including , observed in approximately 23% of treated non-psychiatric (hypertensive) patients across historical studies, which contributed to early insights into monoamine hypotheses of . Tetrabenazine (TBZ), a non-indole reversible , selectively targets VMAT2 with high affinity, stabilizing the transporter in a lumen-facing occluded conformation and thereby inhibiting monoamine sequestration into vesicles, which reduces synaptic release without broadly depleting cytosolic stores. Approved by the FDA in 2008 for the treatment of associated with , TBZ provides symptomatic relief by modulating hyperkinetic movements linked to excessive activity. Its derivatives, and , offer improved pharmacokinetics with once-daily dosing and reduced peak-trough fluctuations; was FDA-approved in 2017 for both Huntington's and , while received approval in 2017 specifically for and in 2023 for associated with . These agents similarly reduce vesicular loading but exhibit lower risks of and compared to TBZ due to their selective VMAT2 inhibition and slower . Amphetamines, including and , function as substrate analogs of VMAT2, entering synaptic vesicles where they become protonated in the acidic , dissipate the proton gradient essential for transport, and induce reverse transport (efflux) of monoamines into the to facilitate non-vesicular release via plasma membrane transporters. This mechanism amplifies extracellular monoamine levels, contributing to the psychostimulant effects of these drugs. indirectly modulates VMAT2 function through its primary blockade of the (), leading to elevated cytosolic that overwhelms vesicular capacity and is associated with reduced VMAT2 density in chronic users, as evidenced by decreased binding in studies. Among other agents, serves as a weak, serotonin-preferring of VMAT2-mediated , to the with modest and showing slight selectivity over VMAT1, though its primary clinical role remains as a antagonist. Recent investigations, including a 2024 , highlight the potential of established VMAT2 like derivatives in treating psychotic symptoms, suggesting antipsychotic efficacy with a lower risk of compared to traditional agents, though dedicated trials for novel VMAT2-targeted compounds in remain ongoing.

Clinical significance

Role in neurological disorders

Dysfunction of the (VMAT2) plays a significant role in several neurological disorders, particularly those involving . In (PD), reduced VMAT2 binding in the , as measured by (PET) imaging with [¹⁸F]dihydrotetrabenazine (DTBZ), strongly correlates with the loss of dopamine-producing neurons in the . This decline in VMAT2 density serves as a reliable for assessing dopaminergic degeneration and monitoring disease progression, with longitudinal studies showing detectable changes over 2 years in PD patients. VMAT2 PET imaging thus provides an measure of nigrostriatal integrity, independent of symptomatic fluctuations. In (HD), VMAT2 inhibitors like (TBZ) effectively treat by reversibly blocking VMAT2, which reduces vesicular uptake and depletes presynaptic stores in the , thereby attenuating hyperkinetic movements. A 2024 systematic and of clinical trials confirmed TBZ's , with response rates up to 75% in long-term studies and mean reductions of 3-5 points in total maximal scores compared to placebo. This therapeutic approach highlights VMAT2's role in modulating striatal release, offering symptomatic relief without altering progression. Rare genetic mutations in the SLC18A2 gene encoding VMAT2 are associated with severe neurological syndromes, including a form of monoamine vesicular that presents with infantile-onset parkinsonism-dystonia and autonomic instability. Case reports from the 2010s describe infantile VMAT2 deficiency leading to profound , hypotonia, and lethality in early childhood due to or systemic complications. These mutations disrupt monoamine packaging, underscoring VMAT2's essential role in early neurodevelopment. At the mechanistic level, loss-of-function in VMAT2 results in impaired sequestration of into synaptic vesicles, causing its accumulation in the where it undergoes auto-oxidation to form reactive quinones and . This promotes mitochondrial dysfunction, including complex I inhibition and energy failure, which exacerbates neuronal toxicity and contributes to neurodegeneration in disorders like . Such cytosolic buildup thus links VMAT2 deficits to broader cellular damage in monoaminergic systems.

Implications in psychiatric conditions and addiction

Vesicular monoamine transporter 2 (VMAT2) dysfunction contributes to mood dysregulation in psychiatric conditions through impaired monoamine storage and release. Heterozygous VMAT2 mutant mice exhibit a depressive-like characterized by increased immobility in forced swim and tail suspension tests, without accompanying anxiety-like behaviors, suggesting a specific role in mood instability. Pharmacological inhibition of VMAT2 by depletes vesicular monoamine stores, precipitating depressive symptoms that mimic those in and bipolar depression, highlighting VMAT2's necessity for maintaining monoaminergic tone. Elevated VMAT2 binding has been observed in the and ventral brainstem of individuals with type I, potentially reflecting compensatory adaptations to underlying monoamine imbalances. In , particularly , reduced VMAT2 expression disrupts presynaptic packaging, contributing to hyperdopaminergic states in mesolimbic pathways. Postmortem analyses of schizophrenic tissue reveal significantly decreased VMAT2 mRNA levels in the and , alongside reduced protein availability, which may impair vesicular sequestration and exacerbate psychotic symptoms. A 2024 and indicates that VMAT2 inhibitors exhibit efficacy in preclinical models, potentially offering a therapeutic avenue with a lower risk of compared to traditional antagonists. This approach modulates release indirectly, preserving receptor sensitivity while mitigating associated with chronic use. VMAT2 plays a central role in addiction by facilitating drug-induced monoamine efflux and subsequent neuroadaptations. Amphetamines and MDMA reverse VMAT2-mediated transport, promoting dopamine and serotonin efflux from vesicles into the cytosol and enhancing synaptic release, which amplifies reward signaling in the nucleus accumbens. This mechanism underlies the acute reinforcing effects of these substances, as VMAT2 inhibition increases cytosolic monoamine availability for reversal through plasma membrane transporters. Chronic exposure to stimulants like methamphetamine downregulates VMAT2 expression and function in the striatum, leading to depleted vesicular stores, tolerance to drug effects, and heightened vulnerability to relapse. In animal models, VMAT2 knockdown via heterozygous knockout enhances behavioral sensitization to cocaine, increasing locomotor responses and promoting self-administration behaviors that model addiction escalation. Human positron emission tomography imaging corroborates this, showing reduced striatal VMAT2 availability in individuals with cocaine use disorder, correlating with greater addiction severity and relapse risk.

Current research

Structural and mechanistic advances

Recent advances in cryo-electron microscopy (cryo-EM) have provided high-resolution structures of human VMAT2 in multiple conformational states, elucidating key aspects of its transport cycle. In 2024, Wang et al. reported structures of VMAT2 in the apo lumen-facing state at 3.6 Å resolution, serotonin-bound lumen-facing state at 3.57 Å, tetrabenazine-bound occluded state at 3.37 Å, and reserpine-bound cytosol-facing state at 3.7 Å, revealing the proton-driven alternating access mechanism and inhibitor binding modes. Similarly, the same year, Schvartz et al. determined a 3.1 Å structure of VMAT2 bound to tetrabenazine in an outward-open conformation, highlighting inhibitor-induced stabilization of the cytoplasmic-facing state. These structural data have been complemented by mechanistic studies on recognition and proton coupling. Im et al. (2024) resolved VMAT2 structures in the apo outward-facing state at 3.05 Å, -bound outward-facing state at 2.90 Å, and tetrabenazine-bound occluded state at 3.18 Å, demonstrating that engages a side-pocket via hydrogen bonds with Glu312 and Ser338, a with Asp399, and π-π stacking with Tyr341 and Tyr433. The study further identified Asp399 and Asp426 as critical protonatable residues that couple binding to proton antiport, with hierarchical facilitating conformational transitions during the transport cycle. Computational modeling has enhanced understanding of VMAT2 dynamics beyond static structures. (MD) simulations in the Wang et al. study, using and 200-ns trajectories, predicted conformational shifts from lumenal- to cytoplasmic-open states upon serotonin and , identifying flexible loops involved in substrate occlusion. Additional MD analyses by Schvartz et al. explored tetrabenazine-bound states over 1 μs, revealing allosteric pockets near the proton pathway that could serve as targets for novel modulators by stabilizing intermediate conformations. Comparative structural analyses of VMAT isoforms have clarified substrate selectivity differences. Ye et al. (2024) obtained eight cryo-EM structures of VMAT1 at resolutions ranging from 2.9 to 3.4 Å, including unbound, monoamine-bound (, noradrenaline, serotonin, ), and reserpine-bound states, showing a wrist-and-fist-shaped binding pocket with polar residues like Glu320 and Ser346 that accommodate histamine's ring via specific hydrogen bonds, unlike in VMAT2. This structural divergence explains VMAT1's preferential transport of histamine in peripheral tissues compared to VMAT2's bias toward catecholamines and serotonin in the . In 2025, further cryo-EM structures advanced the understanding of VMAT2 function. Liu et al. reported an ensemble of high-resolution structures of human VMAT2 in three distinct states bound to multiple substrates and inhibitors, providing insights into substrate and drug inhibition mechanisms. Additionally, Wu et al. presented structures of VMAT2 bound to serotonin, , tetrabenazine, and valbenazine, elucidating detailed inhibition and processes.

Therapeutic developments and clinical trials

Vesicular monoamine transporter 2 (VMAT2) inhibitors have advanced significantly in clinical applications for , with demonstrating robust efficacy in phase III trials. The KINECT-HD phase III study, completed prior to 2025, showed that treatment led to a statistically significant reduction in severity, with an approximate 40% improvement in Unified Huntington's Disease Rating Scale total maximal scores compared to placebo over 12 weeks. Long-term follow-up data from 2025, including three-year analyses from the KINECT-HD2 open-label extension, confirmed sustained reductions and a favorable safety profile, with treatment-emergent adverse events primarily mild to moderate. Additionally, from 2025 pediatric cohorts with hyperkinetic , including Huntington's, indicated that VMAT2 inhibitors like are well tolerated, with as the most common affecting about 10% of patients and low discontinuation rates. In the realm of therapy, VMAT2 inhibitors are emerging as potential adjuncts for due to their ability to modulate presynaptic release without strong postsynaptic receptor . A 2024 and of clinical data on VMAT2 inhibitors, including and analogs, found evidence of effects in psychotic disorders, with improvements in positive symptoms and a potentially lower risk of compared to traditional . Phase 1 trials of novel VMAT2 inhibitors, such as Neurocrine's NBI-1140675, are underway as of 2025, building on earlier investigations into selective VMAT2 modulation for , though specific analogs like those derived from pathways remain in preclinical exploration. For addiction therapies targeting dependence, preclinical research has focused on VMAT2 upregulators to counteract drug-induced depletion of vesicular stores. Studies in models demonstrate that genetic overexpression of VMAT2 protects against by enhancing packaging and reducing cytosolic leakage, without amplifying rewarding effects. However, no VMAT2-targeted therapies have received regulatory approval for use disorder as of 2025, with ongoing efforts emphasizing inhibitors like lobeline analogs to block 's interaction with VMAT2. VMAT2 PET imaging ligands serve as promising biomarkers for early detection and , with ligands like 18F-AV-133 enabling quantification of nigrostriatal degeneration. Clinical trials from 2023 to 2025 have validated 18F-AV-133's sensitivity in monitoring disease progression, showing a detectable annual decline in striatal binding of approximately 5-7% over two years in patients, providing class IV evidence for its utility in tracking neuron loss. These studies underscore 18F-AV-133's role in identifying prodromal and evaluating therapeutic responses, with automated analysis pipelines enhancing reproducibility.

References

  1. [1]
    Vesicular Monoamine Transporters: Structure-Function ... - PMC - NIH
    Vesicular monoamine transporters (VMAT) are responsible for the uptake of cytosolic monoamines into synaptic vesicles in monoaminergic neurons.
  2. [2]
    Neurotransmitter recognition by human vesicular monoamine ...
    Sep 16, 2024 · Human vesicular monoamine transporter 2 (VMAT2), a member of the SLC18 family, plays a crucial role in regulating neurotransmitters in the brain.
  3. [3]
    The ins and outs of vesicular monoamine transporters - PMC
    VMAT is a transporter essential for life. VMAT mediates packaging of the monoamines serotonin, dopamine, norepinephrine, and histamine from the neuronal ...
  4. [4]
    SLC18: Vesicular neurotransmitter transporters for monoamines and ...
    Their potential role in cellular detoxication aside, the major biological function for SLC18 family members is the storage and release of neurotransmitters (Fig ...
  5. [5]
    The ins and outs of vesicular monoamine transporters
    Apr 17, 2018 · The H+-coupled vesicular monoamine transporter (VMAT) is a transporter essential for life. VMAT mediates packaging of the monoamines serotonin, ...
  6. [6]
    Biochemistry and Molecular Biology of the Vesicular Monoamine ...
    Nov 1, 1994 · The vesicular monoamine transporter (VMAT) is sensitive to both components of the proton-motive force ΔμH the pH gradient and the ...
  7. [7]
    SLC18A1 solute carrier family 18 member A1 [ (human)] - NCBI
    Sep 9, 2025 · Polymorphism in VMAT1 gene on chromosome 8p is associated with schizophrenia. Our findings support the hypothesis that anxiety-related ...
  8. [8]
    Individual Residues Contribute to Multiple Differences in Ligand ...
    Functional analysis has previously shown that VMAT2 has a higher affinity than VMAT1 for monoamine neurotransmitters as well as the inhibitor tetrabenazine. The ...
  9. [9]
    p54219 · vmat1_human - UniProt
    Expressed primarily in neuroendocrine tissues. Highly expressed in chromaffin cells of the adrenal medulla (at protein level). Detected in peripheral ...
  10. [10]
    SLC18A2 solute carrier family 18 member A2 [ (human)] - NCBI
    Aug 19, 2025 · Chromogranin A and vesicular monoamine transporter 2 are localized in protein bodies of human locus coeruleus neurons. These results may ...
  11. [11]
    Transport and inhibition mechanism for VMAT2-mediated synaptic ...
    Jan 2, 2024 · Both VMAT2 and VMAT1 have a similar affinity for serotonin, but VMAT2 exhibits a preference for catecholamines (e.g., DA, NE, epinephrine) with ...
  12. [12]
    Expression and function of the rat vesicular monoamine transporter 2
    Feb 20, 2008 · VMAT1 is insensitive to the noncompetitive inhibitor tetra- benazine while VMAT2 shows nanomolar affinity. In addition,. VMAT1 displays two ...
  13. [13]
    Structural mechanisms for VMAT2 inhibition by tetrabenazine - eLife
    Mar 22, 2024 · VMAT2 and VMAT1 share 62% sequence identity but have distinct substrate specificity and pharmacological properties (Erickson et al., 1996).<|control11|><|separator|>
  14. [14]
    Chimeric Vesicular Monoamine Transporters Identify Structural ...
    In contrast to substrate affinity and tetrabenazine sensitivity, reserpine inhibits serotonin transport by VMAT1 with slightly greater potency than ...<|separator|>
  15. [15]
    The vesicular monoamine transporter 2 - PMC - NIH
    VMATs primarily transport monoamines (dopamine, serotonin, norepinephrine and histamine), but also sequester toxicants into vesicles, shunting them away from ...
  16. [16]
    A Review of Neurotransmitters Sensing Methods for Neuro ... - MDPI
    They are classified into three groups: catecholamines, indolamines and imidazoleamines. The three known catecholamines (dopamine, norepinephrine and ...
  17. [17]
  18. [18]
    The Biogenic Amines - Neuroscience - NCBI Bookshelf
    Norepinephrine (also called noradrenaline) synthesis requires dopamine β-hydroxylase, which catalyzes the production of norepinephrine from dopamine (see Figure ...
  19. [19]
    VMAT2: a dynamic regulator of brain monoaminergic neuronal ...
    VMAT2 is the CNS vesicular transporter for not only the biogenic amines DA, NE, EPI, 5-HT, and HIS, but likely also for the trace amines TYR, PEA, and ...
  20. [20]
  21. [21]
  22. [22]
  23. [23]
    [PDF] Arvid Carlsson - Nobel Lecture
    We demonstrated the depletion of catecholamines from the adrenal medulla of rabbits following treatment with reserpine (Carlsson and Hillarp 1956). This was ...Missing: vesicular | Show results with:vesicular
  24. [24]
    Uptake of catecholamines by a particulate fraction of the adrenal ...
    The uptake was stimulated three- to five-fold by Mg(++) and adenosine triphosphate. Reserpine (1 x 10(-5)M) caused a 90 percent inhibition of uptake; ...Missing: dependent | Show results with:dependent
  25. [25]
    Cloning and expression of a rat brain L-glutamate transporter
    Here we use an antibody against a glial L-glutamate transporter from rat brain to isolate a complementary DNA clone encoding this transporter.Missing: VMAT2 Liu
  26. [26]
    Cloning and functional expression of a tetrabenazine ... - PubMed
    Unlike rat VMAT1, bovine VMAT2 appears to be expressed both in the adrenal medulla and the brain, as judged by Northern analysis. After modification/deletion of ...Missing: Liu | Show results with:Liu
  27. [27]
    Knockout of the vesicular monoamine transporter 2 gene ... - PubMed
    Knockout of the vesicular monoamine transporter 2 gene results in neonatal death and supersensitivity to cocaine and amphetamine · Authors · Affiliation.Missing: Fon et al 1990s lethality
  28. [28]
    Vesicular and Plasma Membrane Transporters for Neurotransmitters
    Vesicular and Plasma Membrane Transporters for Neurotransmitters. Randy D Blakely ... This high apparent affinity may contribute to the robust VMAT activity ...
  29. [29]
    p-Chloroamphetamine induces serotonin release through serotonin ...
    Gary Rudnick, Walter Sandtner. Serotonin transport in the 21st century ... Studies with Human Neuroblastoma Cells Transfected with a Rat Serotonin Transporter.
  30. [30]
    Chemical neuroanatomy of the vesicular amine transporters
    Dec 1, 2000 · The proteins expressed by all of these cell populations that make them 'amine handling' include enzymes for synthesizing amines, plasma membrane ...
  31. [31]
    Identification of molecular hinge points mediating alternating access ...
    Transport of monoamines (serotonin, dopamine, histamine, adrenaline, and noradrenaline) is carried out by the vesicular monoamine transporter (VMAT) family, ...
  32. [32]
    Engineering of a mammalian VMAT2 for cryo-EM analysis results in ...
    Aug 2, 2024 · We present two cryo-EM structures of VMAT2, with a frog VMAT2 adopting a canonical MFS fold and an engineered sheep VMAT2 adopting a non-canonical fold.
  33. [33]
    Structural insights into vesicular monoamine storage and drug ...
    This structure provides direct evidence that VMAT1 binds to amphetamine in a similar manner to monoamines, confirming that amphetamine is an actual VMAT ...
  34. [34]
  35. [35]
    Trafficking of Vesicular Neurotransmitter Transporters - PMC
    For VMAT2, a dileucine motif (IL) encoded in the C-terminus is required for its efficient endocytosis in PC12 cells as well as in hippocampal neurons (44, 95) ( ...
  36. [36]
    8JTC: Human VMAT2 complex with reserpine - RCSB PDB
    Nov 29, 2023 · Ki: min: 1, max: 630 (nM) from 4 assay(s). Kd: 8 (nM) from 1 assay(s). IC50: min: 1.3, max: 100 (nM) from 12 assay(s). Experimental Data ...
  37. [37]
    Emulating proton-induced conformational changes in the vesicular ...
    Nov 7, 2016 · Here we show that two potent inhibitors of VMAT2, tetrabenazine and reserpine, bind to different conformations of the protein.Missing: DXXND | Show results with:DXXND
  38. [38]
    Identification of molecular hinge points mediating alternating access ...
    Vesicular monoamine transporter 2 (VMAT2) catalyzes transport of monoamines into storage vesicles in a process that involves exchange of the charged monoamine ...
  39. [39]
    Structural mechanisms for VMAT2 inhibition by tetrabenazine - PMC
    Sep 5, 2023 · The cytosolic facing side of VMAT2 is characterized primarily by the unstructured N- and C- termini along with a 20-residue loop that connects ...Missing: luminal | Show results with:luminal
  40. [40]
  41. [41]
  42. [42]
  43. [43]
  44. [44]
    Structures and General Transport Mechanisms by the Major ...
    Apr 22, 2021 · In this review, we explore the structure, function, regulation, dynamics, oligomerization, and complexes of the major facilitator superfamily ( ...
  45. [45]
    Localization and Expression of VMAT2 Aross Mammalian Species
    Feb 18, 2014 · This chapter provides an update on the neuroanatomical distribution of VMAT2 across various mammalian species, including human, primate, pig, rat, and mouse.Missing: subcellular | Show results with:subcellular
  46. [46]
    Immunochemical localization of vesicular monoamine transporter 2 ...
    The antibody successfully binds to and labels VMAT2, showing specific protein expression in regions that correspond to monoamine production and release ...
  47. [47]
    Differential expression of two vesicular monoamine transporters
    We have detected expression of vesicular monoamine transporter 1 (VMAT1) in adrenal chromaffin cells but not in neural cells.Missing: Hökfelt Bloom fractionation
  48. [48]
    Vesicular monoamine transporter 1 mediates dopamine secretion in ...
    ... VMAT-1 is the predominant isoform expressed in isolated proximal tubule cells. These results were confirmed by immunohistochemistry analysis of rat renal ...
  49. [49]
    Vesicular monoamine transporter (VMAT) regional expression and ...
    VMATs are key regulators of neurotransmitter release responsible for controlling numerous physiological, cognitive, emotional, and behavioral functions.
  50. [50]
    Immunoelectron microscopic localization of VMAT2 protein to ...
    VMAT2 resides on the membrane of secretory vesicles in monoaminergic neurons of the nervous system and in a variety of secretory cells in the gastrointestinal, ...
  51. [51]
    Distribution of vesicular monoamine transporter 2 protein in human ...
    The levels of VMAT2 protein in the cerebellar cortex and the widespread cerebral neocortical areas were all at least 100-fold lower than those in striatum.Missing: Helander 2023
  52. [52]
    Vesicular monoamine transporter (VMAT) regional ... - Cell Press
    Nov 15, 2023 · VMAT2 expression is also high in mast cells, leading to the accu- mulation of serotonin and histamine in variable ratios, and providing ...
  53. [53]
    Variations in the vesicular monoamine transporter 1 gene (VMAT1 ...
    Two different isoforms of the transporter are known, VMAT1 and VMAT2, both encoded by different genes (Peter et al, 1993).
  54. [54]
    Gastrin Induces Expression and Promoter Activity of the Vesicular ...
    Combined mutations of the CRE and either one of the adjacent Sp1 sites lead to a complete derogation of the basal promoter activity. ... VMAT-2 via Sp1 and CREB.
  55. [55]
    SLC18A2 Gene - VMAT2 Antibody - GeneCards
    3 by Ensembl. SLC18A2 Gene in genomic location: bands according to Ensembl, locations ... gene (or VAT2) maps to chromosome 10q25. Both of these sites occur ...
  56. [56]
    Expression of dopamine transporter and vesicular monoamine ...
    Thus, repeated amphetamine administration exerts modest and regionally selective effects on DAT and VMAT2 mRNA expression in subpopulations of midbrain dopamine ...
  57. [57]
    Acquired dysregulation of dopamine homeostasis reproduces ...
    Nov 13, 2020 · This model demonstrates that a progressive acquired loss of VMAT2 expression in adulthood is sufficient to induce Parkinson's disease-associated ...
  58. [58]
    Oestrogens Prevent Loss of Dopamine Transporter (DAT) and ...
    Jul 1, 2005 · MPTP induced a loss of DAT and VMAT2 specific binding in the striatum and substantia nigra, as well as a decrease of VMAT2 mRNA in the ...
  59. [59]
    Sex differences in dopamine release regulation in the striatum - PMC
    Females show lower expression of D1 receptors on GABAergic MSNs as well higher DAT expression and increased VMAT2 activity, indicating greater regulation of ...
  60. [60]
    hVMAT2: A Target of Individualized Medication for Parkinson's ...
    Specifically, the epigenetic activity of hVMAT2 could be regulated more by DNA methylation than by histone acetylation in human dopaminergic neurons. DNA ...
  61. [61]
    Putative Role of Pharmacogenetics to Elucidate the Mechanism of ...
    VMAT2 transports and concentrates monoamines (catecholamines, serotonin ... The low-expression AA genotype of SCL18A2 rs363224 variant appeared to be ...
  62. [62]
    Association study of the vesicular monoamine transporter gene ...
    We further found the rs363224 marker to interact with the putative functional D2 receptor rs6277 (C957T) polymorphism (p = 0.001), supporting the dopamine ...Missing: variability | Show results with:variability
  63. [63]
    Trafficking of Vesicular Neurotransmitter Transporters - Fei - 2008
    Aug 13, 2008 · For VMAT2, a dileucine motif (IL) encoded in the C-terminus is required for its efficient endocytosis in PC12 cells as well as in ...
  64. [64]
    [PDF] mechanisms of vesicular monoamine transporter-2 degradation
    Nov 7, 2016 · Figure 1. Human VMAT2 sequence and putative transmembrane domains. The amino acid human VMAT2 sequence from the N-terminus starting at the ...
  65. [65]
    Glycosylation of a Vesicular Monoamine Transporter: A Mutation in a ...
    Pro43 is a highly conserved residue located at the beginning of the large loop in which all the potential glycosylation sites are found. A Pro43Leu mutant ...
  66. [66]
  67. [67]
    Phosphorylation of a vesicular monoamine transporter by casein ...
    Mar 7, 1997 · Purified CKI and CKII phosphorylate the wild-type carboxyl terminus of VMAT2, but not a double mutant with both serines 512 and 514 replaced by ...Missing: PKA | Show results with:PKA
  68. [68]
    Protein kinase A affects trafficking of the vesicular monoamine ...
    These results suggest that protein kinase A affects the formation and/or composition of VMAT trafficking complexes. Publication types. Research Support, U.S. ...Missing: PKC | Show results with:PKC
  69. [69]
    N-terminus regulation of VMAT2 mediates methamphetamine ... - NIH
    These data support the conclusion that the VMAT2 N-terminus regulates the sequestration of monoamine as well as its METH-triggered efflux from storage sites in ...Missing: feedback | Show results with:feedback
  70. [70]
    The Vesicular Monoamine Transporter-2 - PMC - NIH
    Reserpine produces a long-lasting depletion of monoaminergic content from vesicles that express VMAT2. The use of reserpine as a first-line antihypertensive was ...Missing: Ki | Show results with:Ki
  71. [71]
    Structural mechanisms for VMAT2 inhibition by tetrabenazine - PMC
    The cytosolic facing side of VMAT2 is characterized primarily by the unstructured N- and C- termini along with a 20-residue loop that connects the two halves, ...
  72. [72]
    Lobelane Inhibits Methamphetamine-Evoked Dopamine Release ...
    Lobeline interacts with nicotinic receptor subtypes, dopamine transporters (DATs), and vesicular monoamine transporters (VMAT2s). Methamphetamine inhibits VMAT2 ...
  73. [73]
    Interaction of Amphetamines and Related Compounds at the ... - DOI
    The major purpose of this study was to determine the mode of interaction (VMAT2 substrate or inhibitor) for a wide range of test compounds. Toward this end ...
  74. [74]
    Vesicular monoamine transporter - Wikipedia
    The vesicular monoamine transporter (VMAT) is a transport protein integrated into the membranes of synaptic vesicles of presynaptic neurons.
  75. [75]
    reserpine [TUSOM | Pharmwiki] - TMedWeb
    Aug 1, 2022 · A competitive inhibitor of the monoamine vesicular monoamine uptake transporters (VMAT1 & VMAT2). · Binds to the same site on VMAT1 & 2 as ...
  76. [76]
    The effects of reserpine on depression: A systematic review - PMC
    We systematically reviewed evidence on effects of reserpine on depressive and related symptoms (eg anxiety, suicidal ideation).
  77. [77]
    Vesicular Monoamine Transporter 2 (VMAT2) Inhibitors - NCBI - NIH
    Apr 2, 2019 · The vesicular monoamine transporter type 2 (VMAT2) inhibitors are agents that cause a depletion of neuroactive peptides such as dopamine in nerve terminals.Missing: D119 | Show results with:D119
  78. [78]
    Unique and overlapping mechanisms of valbenazine ... - Nature
    Apr 23, 2025 · In 2017, the Food and Drug Administration (FDA) approved valbenazine and deutetrabenazine, two vesicular monoamine transporter 2 (VMAT2) ...
  79. [79]
    New generation VMAT2 inhibitors induced parkinsonism - PMC - NIH
    In 2017, valbenazine and deutetrabenazine were FDA approved for the treatment of tardive syndrome. Valbenazine and deutetrabenazine are VMAT-2 inhibitors and ...
  80. [80]
    In Vivo Evidence for Low Striatal Vesicular Monoamine Transporter ...
    Jan 1, 2012 · The results of this in vivo PET study confirm previous in vitro reports of low VMAT2 availability in the striatum of cocaine abusers. It also ...
  81. [81]
    Meta-analysis and systematic review of vesicular monoamine ...
    Jan 19, 2024 · VMAT-2 inhibitors may have antipsychotic activity and may offer promise for treatment of psychosis with the potential for a reduced risk of TD.
  82. [82]
    Correlation of Parkinson Disease Severity and 18F-DTBZ Positron ...
    18 F-DTBZ PET is a potential imaging biomarker for measuring dopaminergic degeneration in vivo and monitoring the severity of disease.
  83. [83]
    Using 18F-AV-133 VMAT2 PET Imaging to Monitor Progressive ...
    This study provides Class IV evidence that VMAT2 PET can detect patients with Parkinson disease and quantify progression over a 2-year window.
  84. [84]
    18F-DTBZ PET tracks dopaminergic degeneration in patients with ...
    May 20, 2014 · “Our findings suggest that 18F-DTBZ PET imaging is a potential imaging biomarker for measuring dopaminergic degeneration in vivo and monitoring ...
  85. [85]
    Efficacy and safety of vesicular monoamine transporter 2 inhibitors ...
    Sep 23, 2025 · Conclusion: This study suggests that three VMAT2 inhibitors are effective in ameliorating chorea symptoms in patients with Huntington's disease.Missing: 2024 | Show results with:2024
  86. [86]
    Efficacy and Safety of Tetrabenazine in Reducing Chorea and ...
    Oct 14, 2024 · This systematic review investigated TBZ's efficacy in reducing chorea and improving motor function in HD patients. The review answered the ...
  87. [87]
    Brain Dopamine–Serotonin Vesicular Transport Disease and Its ...
    Jan 30, 2013 · We describe a disease encompassing infantile-onset movement disorder (including severe parkinsonism and nonambulation), mood disturbance, autonomic instability ...
  88. [88]
    Brain monoamine vesicular transport disease caused by ...
    Oct 31, 2022 · Brain monoamine vesicular transport disease is an infantile-onset movement disorder that mimics cerebral palsy. In 2013, the homozygous ...Missing: lethal | Show results with:lethal
  89. [89]
    Reduced Vesicular Storage of Dopamine Causes Progressive ...
    Jul 25, 2007 · ... disease (PD) (Jenner, 2003). This suggests that VMAT2 may be an important determinant in dopamine-related toxicity and reduction in its ...
  90. [90]
    Acquired dysregulation of dopamine homeostasis reproduces ...
    Nov 13, 2020 · ... toxicity was specific to the loss of VMAT2 ... Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson's disease.Results · Striatal Dopamine... · Induction Of Lrrk2 Activity
  91. [91]
    Vmat2 Heterozygous Mutant Mice Display a Depressive-Like ... - NIH
    Importantly, treatment with reserpine, an irreversible inhibitor of the VMAT, leads to depletion of vesicular monoamine stores and it can precipitate depressive ...
  92. [92]
    Vesicular monoamine transporter concentrations in bipolar disorder ...
    Aug 6, 2025 · In humans, changes in VMAT2 level have been correlated with depression, bipolar disorder ... Single-nucleotide polymorphisms upstream of VMAT2 ...<|separator|>
  93. [93]
    Putative presynaptic dopamine dysregulation in schizophrenia ... - NIH
    Jan 17, 2017 · A decrease in VMAT2 gene expression in the substantia nigra may contribute to reduced VMAT2 protein and suggests less efficient vesicular ...
  94. [94]
    Meta-analysis and systematic review of vesicular monoamine ...
    Jan 19, 2024 · VMAT-2 inhibitors may have antipsychotic activity and may offer promise for treatment of psychosis with the potential for a reduced risk of TD.Missing: classes | Show results with:classes<|control11|><|separator|>
  95. [95]
    VMAT-2 Inhibitors Potentially Help Psychosis - CARLAT PUBLISHING
    May 1, 2025 · VMAT-2 inhibitors may improve psychotic symptoms, but we'll wait for confirmation as this was a secondary analysis.
  96. [96]
    Vesicular Monoamine Transporter 2 and the Acute and Long-Term ...
    Thus, VMAT2 is a molecular target for a number of psychostimulant drugs, including methamphetamine and MDMA. We investigated the effects of depressed VMAT2 ...Missing: hyperpronation | Show results with:hyperpronation
  97. [97]
    Vesicular Transport Regulates Monoamine Storage and Release ...
    To assess the role of exocytotic release in signaling by monoamines, we have disrupted the neuronal vesicular monoamine transporter 2 (VMAT2) gene.
  98. [98]
    VMAT2 knockout mice: Heterozygotes display reduced ... - PNAS
    Amphetamines dissipate proton gradients across the membranes of synaptic vesicles, disrupt VMAT2 function, enhance cytoplasmic monoamine concentrations, and ...
  99. [99]
    In vivo evidence for reduced striatal vesicular monoamine ... - NIH
    The results of this in vivo PET study confirm previous in vitro reports of lower VMAT2 availability in the striatum of cocaine abusers.
  100. [100]
    Safety and efficacy of valbenazine for the treatment of chorea ...
    Interpretation: In individuals with Huntington's disease, valbenazine resulted in improvement in chorea compared with placebo and was well tolerated. Continued ...Missing: 2025 | Show results with:2025
  101. [101]
    Neurocrine Biosciences Presents New Three-Year Data ...
    Oct 6, 2025 · Once-daily INGREZZA treatment was generally well tolerated and showed early and sustained improvements in chorea severity ...
  102. [102]
    Real-World Experiences with VMAT2 Inhibitors in Pediatric ... - PMC
    Jun 16, 2025 · Our data also suggests that VMAT2 inhibitors are well tolerated with somnolence as the most common side effect; only 10.1% of patients ...
  103. [103]
    Neurocrine initiates trial of VMAT2 inhibitor in healthy adults
    Mar 6, 2025 · Neurocrine Biosciences has commenced the Phase I clinical trial of its oral, small molecule VMAT2 inhibitor, NBI-1140675 in healthy adults.
  104. [104]
    Increased Vesicular Monoamine Transporter 2 (VMAT2
    Mar 9, 2015 · These results demonstrate that elevated VMAT2 protects against METH toxicity without enhancing the rewarding effects of the drug.<|separator|>
  105. [105]
    Effects of VMAT2 inhibitors lobeline and GZ-793A on ... - Europe PMC
    These results provide further preclinical support for the development of VMAT2 inhibitors as therapeutic agents for the treatment of METH abuse. While the ...
  106. [106]
    Using 18 F-AV-133 VMAT2 PET Imaging to Monitor ... - PubMed
    Nov 27, 2023 · This study provides Class IV evidence that VMAT2 PET can detect patients with Parkinson disease and quantify progression over a 2-year window.Missing: ligands 2023-2025 prognostic value
  107. [107]
    Automated VMAT2 [18F] AV-133 PET analysis in Parkinson's disease
    Sep 8, 2023 · We discuss our latest work to evaluate a fully automated image analysis pipeline to process vesicular monoamine transporters type 2 (VMAT2) ...Missing: ligands prognostic