Fact-checked by Grok 2 weeks ago

TRPV1

Transient Receptor Potential Vanilloid 1 (TRPV1) is a non-selective cation belonging to the transient (TRP) family of channels, primarily expressed in y neurons of the dorsal root and trigeminal ganglia, where it functions as a polymodal detecting noxious heat above 43°C, (the pungent compound in chili peppers), protons (low ), and endogenous ligands such as . As a with each subunit comprising approximately 839 (e.g., 838 in ), six transmembrane helices (S1–S6), an intracellular N-terminal rich in repeats, and a C-terminal , TRPV1 forms a central pore that permits influx of monovalent and divalent cations, particularly calcium, leading to membrane and initiation of nociceptive signaling. First cloned in 1997 from dorsal root ganglia as the receptor (initially termed VR1), its identification marked a pivotal advance in understanding molecular mechanisms of transduction. TRPV1's activation integrates multiple stimuli relevant to tissue damage and : thermal gating occurs at temperatures exceeding physiological norms, while chemical activation by or binds to a specific intracellular vanilloid-binding domain, inducing conformational changes that open the ; proton sensitivity enhances gating under acidic conditions, such as during . Endogenous modulation includes phosphorylation by (PKC) or A (), which sensitizes the , and interactions with like 12-HPETE that potentiate activity, contributing to in states. Beyond primary sensory neurons, TRPV1 is expressed in non-neuronal tissues including the , , , and , where it regulates diverse processes such as , insulin secretion, and vascular function. Physiologically, TRPV1 serves as a critical in the pain pathway, converting environmental and endogenous noxious signals into electrical impulses that propagate to the , thereby eliciting protective responses like withdrawal reflexes; its role extends to inflammatory and , where upregulation amplifies sensory . In thermoregulation, TRPV1 activation in hypothalamic neurons helps maintain core body temperature, while in metabolic contexts, it influences and prevents visceral fat accumulation. Pathologically, aberrant TRPV1 activity contributes to conditions like , , , and even neurodegenerative diseases through impaired calcium and . Due to its central role in , TRPV1 has emerged as a prime therapeutic target; agonists like are used topically for desensitization in , while antagonists (e.g., or clinical candidates like NEO6860) aim to block without affecting normal sensation, though challenges include thermoregulatory side effects such as . Recent structural insights from cryo-electron microscopy, including 2024 structures of human TRPV1 with antagonists, have elucidated and inhibition , facilitating rational to modulate specific gating modes. Ongoing research as of 2025 explores TRPV1's involvement in cancer progression (e.g., sensitizing tumors to ), immune modulation, , and therapies, underscoring its broader implications in neuroimmune interactions.

Molecular Structure

Overall Architecture

TRPV1 is a tetrameric , with each subunit comprising six transmembrane helices (S1–S6), an intracellular amino (N)-terminal domain, and an intracellular carboxy (C)-terminal domain. The S5–S6 helices and the intervening re-entrant pore loop (S5–S6 linker) form the central pore domain, while the S1–S4 helices constitute a voltage-sensor-like domain that contributes to the channel's peripheral architecture. This overall fold aligns with the canonical of tetrameric cation channels, as revealed by the first high-resolution cryo-EM of rat TRPV1 at 3.4 Å in 2013. Subsequent refinements have achieved resolutions below 3 Å, including ligand-bound states, enabling detailed visualization of conformational dynamics, with resolved as of 2024. Recent 2024 have further revealed inhibitory sites involving the S2–S3 transmembrane segments, aiding efforts. The capsaicin-binding pocket is located within the transmembrane region, primarily involving residues from the S3–S4 helices of one subunit and the S4–S5 linker of the adjacent subunit, forming a interaction site that accommodates agonists like through hydrophobic and hydrogen-bonding interactions. On the extracellular side, the double-knot toxin (DkTx) binds at the periphery of the pore domain, interacting with residues in the S1–S2 and S3–S4 linkers across adjacent subunits in a counterclockwise manner, which stabilizes the open conformation. These binding sites highlight the channel's modular design for polymodal activation, with the site positioned externally to modulate pore accessibility. The cytoplasmic N-terminus features a conserved ankyrin repeat domain (ARD) consisting of approximately six ankyrin repeats, which is evolutionarily preserved across TRPV subfamily members and links to the first transmembrane helix via a short linker. This ARD contributes to the of the intracellular , as observed in multiple cryo-EM structures where it adopts an elongated, solenoid-like fold. The C-terminal domain, in contrast, includes a TRP box motif adjacent to S6 and calmodulin-binding sites, forming inter-subunit contacts that support tetrameric .

Functional Domains

The N-terminal region of TRPV1 features an ankyrin repeat domain (ARD) consisting of six ankyrin repeats, each comprising a characteristic 33-amino-acid motif that forms pairs of antiparallel α-helices connected by β-hairpin loops, creating a concave ligand-binding surface essential for integrity and regulation. This ARD contributes to protein stability by maintaining structural folding and facilitating proper assembly, as deletion of the domain impairs overall function and trafficking. Additionally, the ARD serves as a multiligand-binding site, where residues in repeats 1–3 (e.g., R115, K155, K160 for groups and Y199, Q202 for ) enable ATP binding to prevent desensitization (), while overlapping sites allow Ca²⁺-dependent binding to promote it, thereby modulating sensitivity to stimuli. The TRP domain, located immediately C-terminal to the S6 transmembrane segment, encompasses a conserved helical that plays a key role in allosteric gating and interactions. This domain includes the conserved TRP box IWKLQR (residues 691–696 in rat TRPV1), which positions near the intracellular leaflet to sense phosphoinositide like PIP₂, negatively regulating basal channel activity and fine-tuning responses to activators by influencing conformational changes during gating. within this domain, such as at E692 or R701, disrupt the coupling between sensory inputs and opening, underscoring its pivotal role in coordinating -dependent modulation and efficient channel activation. In the , a -binding site (residues approximately 779–838) enables Ca²⁺- interaction to facilitate desensitization following prolonged activation, distinct from the N-terminal site by its lower affinity and role in terminating responses to or heat. Adjacent to this, a coiled-coil domain (residues 683–721) promotes tetramerization by mediating subunit-subunit interactions, ensuring stable quaternary assembly necessary for functional channel formation, as isolated expression of this domain suffices for oligomerization . TRPV1 contains several phosphorylation sites that serve as modular points for sensitization by kinases, particularly (PKC). Notable PKC consensus sites include S502 in the C-terminal linker region and S800 in the distal , where enhances channel sensitivity to agonists like and protons by shifting activation thresholds and reducing desensitization rates, thereby amplifying nociceptive signaling during . Specific residues within the transmembrane domains critically define by diverse stimuli. For binding, Y511 in the S3-S4 linker forms part of the pocket, where its hydroxyl group hydrogen-bonds with the ligand's , enabling recognition and stabilizing the open state; to abolishes without affecting overall structure. Proton involves acidic residues like E600 on the extracellular side of S5 and D576 near the pore turret, which become protonated at low to induce conformational shifts that lower the heat threshold and potentiate gating. Heat sensing, while distributed across the pore domain, is particularly influenced by residues such as R491 in S3 and interactions involving Y511, which couple thermal energy to movements that widen the lower gate for .

Gene and Expression

Genomic Organization

The human TRPV1 gene is located on the short arm of chromosome 17 at position 17p13.2, spanning approximately 45 kb from genomic coordinates 3,565,446 to 3,609,411 (GRCh38.p14 , on the complementary strand). It consists of 19 s, with the coding sequence initiating in exon 2, and produces multiple mRNA transcripts through promoter usage and splicing events. The canonical transcript encodes a 839-amino-acid protein, while other isoforms arise from variations in the or exon inclusion. Alternative splicing of the TRPV1 pre-mRNA generates several isoforms, including the full-length VR.1 variant and truncated or modified forms such as VR.5′sv, TRPV1β, and TRPV1var. VR.5′sv, for instance, results from the use of an alternative 5′ exon and lacks to and heat, potentially acting as a dominant-negative regulator when co-expressed with VR.1. TRPV1β incorporates an additional in the 5′ region, leading to a shorter , while TRPV1var retains 5, producing a frameshift and premature that may influence channel trafficking or stability. These variants contribute to tissue-specific expression and functional diversity of TRPV1 channels. The promoter region upstream of the TRPV1 gene contains regulatory elements, including Sp1/Sp4 binding sites, that mediate transcriptional activation in response to (NGF). NGF, via TrkA receptor signaling and downstream activation of p38 MAPK, enhances TRPV1 promoter activity, increasing mRNA levels in sensory neurons during inflammatory conditions. Additional elements responsive to inflammatory signals, such as those involving pathways, further modulate transcription to adapt channel expression to pathological states. Single nucleotide polymorphisms (SNPs) in the TRPV1 gene, such as rs222747 (c.945A>G, p.Ile315Met) located in exon 5 within the ankyrin repeat domain, have been linked to variations in pain sensitivity and capsaicin-induced irritation. Individuals carrying the G allele exhibit altered thermal and chemical nociception thresholds. Evolutionarily, TRPV1 is highly conserved among mammals, reflecting its fundamental role in thermosensation and nociception; the orthologous Trpv1 gene in mice resides on chromosome 11, sharing >80% sequence identity with the human counterpart. This conservation extends to other vertebrates, with syntenic relationships preserved across species.

Tissue and Cellular Distribution

TRPV1 is primarily expressed in primary sensory neurons, particularly small- and medium-diameter nociceptors within the dorsal root ganglia (DRG) and , where it is found in approximately 50-80% of these neurons based on immunohistochemical and analyses. These neurons project to peripheral tissues involved in detecting noxious stimuli, with TRPV1 localization often co-occurring with markers like (CGRP) and . Beyond neuronal tissues, TRPV1 exhibits non-neuronal expression in various cell types, including epithelial cells of , urinary , and , as confirmed by RNA sequencing and studies. It is also present in immune cells such as macrophages and T lymphocytes, where it contributes to cellular signaling, and in vascular endothelial cells, particularly in response to inflammatory cues. TRPV1 expression undergoes developmental regulation, with postnatal upregulation observed in rodent DRG neurons mediated by nerve growth factor (NGF) signaling through the TrkA receptor, leading to increased mRNA and protein levels by postnatal day 14-21. Species-specific differences are notable, such as higher TRPV1 expression in human bladder urothelium compared to rodents, as revealed by comparative qPCR and analyses. Recent quantitative data from single-cell and in the 2020s have identified low but detectable TRPV1 expression in regions, including the and , often in neurons and , with transcript levels varying by up to 2-5 fold across cell clusters.

Physiological Function

Ion Channel Properties

TRPV1 operates as a non-selective cation , allowing permeation of monovalent ions such as Na⁺ and K⁺, as well as divalent cations like Ca²⁺, with a ratio of P_Ca/P_Na ≈ 9.6. This high calcium permeability contributes to significant Ca²⁺ influx upon activation, which plays a key role in downstream signaling in sensory neurons. The channel's ion selectivity arises from its tetrameric pore structure, which lacks stringent discrimination typical of selective channels like voltage-gated sodium channels. In single-channel recordings, TRPV1 exhibits a conductance of approximately 50–100 , varying with the driving force and ionic composition; for instance, outward currents show a slope conductance of ~100 between +20 and +60 , while inward currents are lower at ~35 . The current-voltage relationship demonstrates pronounced rectification, where currents at positive potentials are larger than at negative ones due to voltage-dependent gating and possible asymmetric ion permeation. This rectification ensures robust under physiological conditions where shifts toward positive values during activation. TRPV1 displays weak but measurable voltage dependence, with channel opening favored at depolarized (positive) membrane potentials, where the activation curve shifts to enhance open probability. The channel's gating can be described by a Boltzmann function for steady-state open probability:
P_o = \frac{1}{1 + \exp\left(-\frac{zF(V - V_{1/2})}{RT}\right)}
where z is the gating valence (~1–2), F is Faraday's constant, R is the gas constant, T is temperature, V is membrane potential, and V_{1/2} is the half-activation voltage (typically ~0 to +100 mV without agonists).
Temperature sensitivity is a hallmark of TRPV1, with an activation threshold of ~43°C and an exceptionally high Q_{10} value of ≈20–25, reflecting steep changes in open probability over narrow ranges. This gating integrates with voltage dependence, as rising shifts V_{1/2} toward more hyperpolarized potentials, promoting even at resting membrane voltages. The adapted for effects modulates V_{1/2} as a function of , underscoring the coupled thermo-voltage mechanism.00252-4)

Sensitization Mechanisms

Sensitization of TRPV1 enhances its responsiveness to stimuli such as heat and , contributing to inflammatory through various molecular mechanisms. These processes primarily involve post-translational modifications and signaling cascades that lower the activation threshold of the channel. Acute sensitization occurs rapidly, within seconds to minutes, often via direct or alterations, while sensitization develops over hours to days through , particularly in inflammatory contexts.00908-X) One key mechanism is (PKC)-mediated , which targets specific serine residues on TRPV1 to increase sensitivity. at Ser502 and Ser800 by PKC, particularly the ε isoform, enhances responses to and protons, as well as heat-evoked currents, without altering basal activity. This modification is activated downstream of Gq-coupled receptors and (PLC), leading to diacylglycerol production and PKC translocation to the . Studies using phosphorylation-deficient mutants (S502A/S800A) demonstrate reduced sensitization, confirming the functional importance of these sites in nociceptive signaling.00413-7) Another pathway involves lipid modulation, notably the depletion of (PIP2) and activation of (PLA2). PIP2 acts as a tonic inhibitor of TRPV1; its by PLC during inflammatory signaling removes this inhibition, thereby sensitizing the channel to agonists. Concurrently, PLA2 activation generates (AA) and subsequent metabolites like 12- or 15-hydroperoxyeicosatetraenoic acid (HPETE), which directly bind and potentiate TRPV1 activity. These lipid changes contribute to acute observed in inflamed tissues.00345-7) Nerve growth factor (NGF) signaling via its receptor TrkA induces chronic sensitization through p38 mitogen-activated protein kinase (MAPK)-dependent transcriptional upregulation of TRPV1 expression. NGF binding to TrkA activates PLC and p38 MAPK, promoting increased TRPV1 mRNA and protein levels in dorsal root ganglion neurons, which sustains hyperalgesia during prolonged inflammation. Inhibition of p38 MAPK blocks this upregulation and reduces heat hypersensitivity in vivo.00908-X) Bradykinin and ATP further promote sensitization by engaging Gq-coupled receptors, such as bradykinin B2 and P2Y receptors, respectively, which trigger PLC activation, PKC phosphorylation, and PIP2 hydrolysis. Bradykinin enhances TRPV1 currents and thermal responses via PKC-dependent pathways, while ATP similarly potentiates channel activity through Gq signaling, amplifying pain signaling in acute inflammatory settings.00413-7)

Desensitization Processes

TRPV1 undergoes several desensitization processes that attenuate its activity following prolonged or repeated stimulation, serving as protective mechanisms against excessive activation. These include acute and forms, which differ in timescale, molecular basis, and functional consequences. Acute desensitization occurs rapidly during continuous exposure, while desensitization involves longer-term reduction in availability at the plasma membrane. refers to the diminished response to successive brief applications, contributing to overall . Acute desensitization of TRPV1 is primarily Ca²⁺-dependent and mediated by () binding to the . Upon channel by agonists like , Ca²⁺ influx elevates intracellular Ca²⁺ levels, enabling Ca²⁺/ to bind a 35-amino acid segment (residues 767–801) in the , which stabilizes a closed conformation and inhibits further . This binding shifts the voltage dependence of the channel rightward, with the half- voltage (V₁/₂) moving from approximately -122 mV under alone to +40 mV in the presence of Ca²⁺/, thereby reducing the open probability and promoting pore closure. The process is fast, occurring within seconds, and is essential for limiting Ca²⁺ overload in sensory neurons. Tachyphylaxis represents a rapid form of desensitization observed during repeated short pulses of agonists, characterized by pore closure that is largely independent of Ca²⁺ influx. This phenomenon arises from agonist-induced conformational changes that directly close the , often linked to depletion of (PIP₂) in the plasma membrane, which normally stabilizes the open state. Unlike acute desensitization, persists in Ca²⁺-free extracellular solutions, though it is slower and less pronounced, highlighting its role in immediate response attenuation without relying on secondary Ca²⁺ signaling. Intracellular ATP or PIP₂ supplementation can prevent or reverse , underscoring the involvement of modulation. Chronic desensitization involves the downregulation of surface TRPV1 expression through agonist-induced , a Ca²⁺-dependent process requiring by (PKC). Prolonged exposure triggers β-arrestin-1 and -2 recruitment to phosphorylated TRPV1, facilitating clathrin-mediated and subsequent trafficking to lysosomes for . This reduces the number of functional channels on the surface by up to 70–80% within 10–30 minutes, contributing to sustained loss of responsiveness. Inhibitors of or clathrin heavy chain block this , confirming the pathway's specificity. Recovery from desensitization occurs over minutes to hours and involves distinct routes for acute and chronic forms. For acute and tachyphylactic desensitization, recovery requires Ca²⁺ influx and activation of Ca²⁺/calmodulin-dependent protein kinase II (CaMKII) and (), which promote TRPV1 reinsertion into the plasma membrane via vesicular trafficking, restoring currents to near-baseline levels within 10–30 minutes. In contrast, recovery from chronic desensitization is slower, relying on protein synthesis to replenish channels after lysosomal , with full restoration taking 1–24 hours depending on stimulus intensity. Recycling of endocytosed TRPV1 back to the surface can partially contribute in milder cases, but degradation predominates during strong activation. These desensitization processes play a key role in pain adaptation by modulating excitability. Repeated exposure induces partial desensitization initially, allowing transient pain relief through temporary channel inactivation, but prolonged or high-dose application leads to complete desensitization via and defunctionalization of sensory neurons, providing longer-term analgesia. This contrasts with mechanisms that enhance TRPV1 responsiveness under inflammatory conditions.

Ligands and Modulators

Agonists

TRPV1, a polymodal , is activated by various agonists that bind to specific sites or influence gating mechanisms, leading to cation influx and cellular . Exogenous vanilloids represent the primary class of chemical agonists, mimicking the of and by engaging an intracellular binding pocket. These compounds have been instrumental in elucidating TRPV1's role in , with potency measured by half-maximal effective concentration (EC50) values in functional assays such as or patch-clamp . Capsaicin, the active component of chili peppers, is the prototypical , activating the channel with an EC50 of approximately 0.7 μM in TRPV1-expressing cells. It binds to the intracellular site, primarily interacting with residues tyrosine 511 (Y511) and serine 512 (S512) in the S3-S4 transmembrane region, stabilizing the open conformation through hydrogen bonding and hydrophobic interactions. This binding induces a conformational change that couples to the channel's domain, facilitating ion permeation. Prolonged exposure to leads to desensitization, a property exploited in topical analgesics for pain relief. Resiniferatoxin (RTX), derived from the resin spurge plant , is an ultrapotent vanilloid agonist with an EC50 of about 1 nM, roughly 1,000 times more potent than due to enhanced affinity at the same intracellular site involving Y511 and S512. RTX's high potency arises from its extended structure, which forms additional stabilizing interactions within the binding pocket, resulting in robust channel activation even at nanomolar concentrations. Beyond its role in , RTX is utilized in imaging studies to label TRPV1-expressing cells, such as in autoradiography for visualizing nociceptors in tissues. Other natural vanilloids, such as from and gingerols (e.g., 6-gingerol) from ginger, also activate TRPV1, albeit with lower potency than (EC50 values in the low micromolar range). These compounds bind similarly to the intracellular vanilloid pocket, eliciting pungent sensations and contributing to the thermogenic effects of spices; for instance, induces calcium influx in TRPV1-transfected cells, while gingerols modulate channel gating to produce mild heat-like responses. Their dietary presence underscores TRPV1's evolutionary role in detecting irritants. In addition to chemical agonists, TRPV1 responds to protons as a non-vanilloid activator, with direct channel opening occurring at extracellular below 6.4, corresponding to a of about 5.4. Protonation targets the extracellular residue 600 (E600) in the extracellular linker between transmembrane segments S5 and the pore helix, neutralizing negative charges and promoting conformational shifts toward the open state; mutations at E600 abolish this sensitivity. This mechanism integrates acidic environments, such as those in inflamed tissues, with TRPV1 activation. Heat serves as a non-chemical , activating TRPV1 at temperatures exceeding 42°C through polymodal gating that converges on shared intracellular activation pathways. Elevated temperatures increase the channel's open probability by enhancing molecular vibrations in the voltage-sensing-like , integrating with or proton signals to lower the activation threshold; this thermosensitivity is tuned for detecting noxious in sensory neurons.

Antagonists

TRPV1 antagonists are compounds that inhibit the channel's activity by to specific sites, thereby blocking permeation and reducing responses to stimuli such as , protons, and capsaicin-like agonists. These inhibitors are classified as competitive, which target the vanilloid site, or non-competitive, which act at the pore or allosteric sites. Early antagonists like served as tools to characterize TRPV1 , while later pharmaceutical developments aimed at relief but encountered physiological side effects. Capsazepine, the first identified competitive of TRPV1, was developed as a synthetic analog of capsaicin and exhibits an of approximately 0.56 μM for inhibiting capsaicin-induced currents. It binds directly to the vanilloid site within the transmembrane domains 3 and 4 of the channel, preventing activation without affecting heat or proton responses at low concentrations. This compound has been instrumental in dissecting TRPV1 , though its modest potency and partial in some assays limited therapeutic advancement. Pharmaceutical efforts in the 2000s produced high-affinity TRPV1 antagonists such as from , with sub-nanomolar potency against activation ( ~1 nM). and similar compounds like AMG9810 advanced to clinical trials for management, demonstrating efficacy in reversing inflammatory in preclinical models at doses as low as 0.3 mg/kg. However, these antagonists induced significant in phase I trials, with body temperatures rising to 39–40.2°C in about one-third of participants due to blockade of TRPV1-mediated , leading to program discontinuation. Natural antagonists include such as , which interacts with TRPV1 to antagonize channel activation, potentially through binding sites that inhibit responses, as shown in studies and functional assays. Another example is ruthenium red, a non-competitive pore blocker that occludes the ion conduction pathway of TRPV1 with high affinity ( ~10 nM), preventing cation influx regardless of the activation modality; cryo-EM structures reveal it coordinates with residues in the selectivity filter to stabilize a closed conformation. These natural compounds offer scaffolds for developing less toxic inhibitors but often lack selectivity. Allosteric modulators of TRPV1 include protons and divalent cations, which can inhibit channel activity under specific conditions. At neutral , protons exert modulatory effects by interacting with extracellular residues, reducing sensitization without direct activation, while divalent cations like Mg²⁺ and Ca²⁺ can block the pore at millimolar concentrations, competing with monovalent ions for permeation sites. These endogenous-like modulators highlight opportunities for indirect inhibition strategies. As of , topical TRPV1 antagonists like ACD440 Gel are in Phase 2a clinical trials for painful neuropathy, showing potential efficacy in reducing evoked pain without systemic . Development of TRPV1 antagonists faces significant challenges, particularly in achieving selectivity over related TRP channels like TRPV2–4 and , which share structural homology and could lead to off-target effects on cardiovascular or respiratory functions. advances have aided in designing site-specific blockers, but balancing potency with minimal and species differences in trial outcomes remains a hurdle for clinical translation.

Endogenous Regulators

Endogenous regulators of TRPV1 encompass a variety of physiological molecules produced within the body that modulate channel activity, often in response to cellular signaling or environmental changes. These include endocannabinoids, fatty acid-derived metabolites, ions such as protons and divalent cations, and membrane lipids like (PIP2). Such regulation fine-tunes TRPV1's role in sensory transduction, particularly in and , by altering channel gating, , or desensitization thresholds. Endocannabinoids, such as (AEA) and N-arachidonoyl (NADA), act as endogenous agonists of TRPV1, binding to the intracellular capsaicin-binding site to directly activate the channel. elicits calcium influx through TRPV1 with an of approximately 11 μM in sensory neurons, functioning as a that contributes to thermal during . N-Arachidonoyl similarly activates TRPV1 with higher potency, achieving an of around 857 nM, and promotes pain signaling by enhancing channel-mediated calcium entry in nociceptive neurons. These endocannabinoids are synthesized on demand via pathways and play a dual role in modulating pain through both TRPV1 and receptors. Fatty acid metabolites generated through the (PLA2) pathway also potently regulate TRPV1, often sensitizing the channel to thermal or chemical stimuli. For instance, 12-hydroperoxyeicosatetraenoic acid (12-HPETE), a product of , activates TRPV1 by binding to lipid-sensing regions, lowering the heat activation threshold and contributing to inflammatory . Similarly, (LTB4), another derived from the PLA2- cascade, directly gates TRPV1 in sensory and non-neuronal cells, such as pancreatic duct epithelia, with implications for tissue and . These metabolites accumulate during cellular stress, amplifying TRPV1 responses to sustain nociceptive signaling. Protons and divalent cations represent key ionic endogenous modulators of TRPV1, influencing gating through extracellular and interactions. Acidic shifts, as occur in ischemic or inflamed tissues (pH ~6.4–5.4), protonate specific residues like Glu600 (in the S5- linker) and Glu648 (in the outer region) in the extracellular domains, potentiating TRPV1 activation and shifting its voltage dependence toward hyperpolarization. Divalent cations, particularly Mg²⁺, exert a biphasic effect: at millimolar concentrations (~10 mM), they potentiate TRPV1 by stabilizing conformational changes that lower the threshold, while also acting as voltage-dependent blockers of the open , reducing inward at depolarized potentials. This modulation is critical for TRPV1's response to physiological ionic fluctuations during tissue damage. Phosphatidylinositol 4,5-bisphosphate (PIP2), a basal , negatively regulates TRPV1 activity by interacting with the proximal C-terminal region, including positively charged residues like Lys710, thereby inhibiting heat- and capsaicin-induced gating. Depletion of PIP2, often via activation during Gq-coupled receptor signaling, removes this tonic inhibition, sensitizing TRPV1 to agonists and contributing to enhanced in inflammatory states. This lipid-channel interaction underscores PIP2's role in maintaining TRPV1's excitability under homeostatic conditions. Recent studies from 2023–2025 highlight omega-3 polyunsaturated fatty acids, such as (DHA) and (EPA), as modulators of TRPV1, potentially mitigating . For example, EPA attenuates TRPV1 overexpression in brain regions in a fibromyalgia model, reducing . DHA and EPA can activate TRPV1 in a protein kinase C-dependent manner, with DHA showing greater efficacy, though direct channel modulation requires prior sensitization.

Physiological Roles

In Sensory Perception

TRPV1 channels are primarily expressed in the peripheral terminals of small-diameter nociceptive primary afferent neurons, including unmyelinated C-fibers and thinly myelinated Aδ fibers, enabling the detection of noxious thermal, chemical, and acidic stimuli. These channels open in response to temperatures exceeding 43°C, the pungent compound from peppers, and extracellular protons (low ), leading to cation influx, membrane depolarization, and generation that signals potential tissue damage to the . This polymodal activation positions TRPV1 as a critical molecular transducer for initiating perception from diverse environmental threats. Upon activation, TRPV1-expressing afferents convey nociceptive signals via their central projections to the superficial laminae (I and II) of the dorsal horn, where they synapse with second-order projection neurons and local to facilitate processing and transmission to supraspinal centers. In this relay, TRPV1-mediated calcium entry enhances glutamate release from primary afferent terminals, amplifying synaptic efficacy and contributing to the encoding of intensity and quality in the dorsal horn circuitry. Under inflammatory conditions, sensitization of TRPV1—through by kinases activated by proinflammatory mediators like , , and protons—lowers its activation threshold, resulting in thermal where innocuous warmth evokes and noxious heat is perceived as more intense. TRPV1 engages in polymodal integration with other transient receptor potential (TRP) channels, such as , which is co-expressed in a subset of these sensory neurons and detects cold temperatures and chemical irritants like . This co-expression allows synergistic enhancement of neuronal excitability; for instance, TRPA1 activation can potentiate TRPV1 currents, broadening the sensory repertoire to encompass multifaceted noxious inputs like those from or mechanical insults. Genetic ablation studies underscore TRPV1's essential role, as TRPV1-null mice display markedly reduced nocifensive behaviors to acute (>50°C) and injection, along with attenuated thermal in models of , without altering responses to mechanical or cold stimuli.

In Non-Neuronal Systems

TRPV1 channels are expressed in , where their activation by heat stimuli triggers the release of proinflammatory cytokines, contributing to barrier responses and . Specifically, heat exposure above 43°C activates TRPV1 in human epidermal , leading to increased expression and secretion of TNF-α through calcium influx and downstream signaling pathways such as NFATc1. Similarly, TRPV1-mediated responses in promote IL-1β release, which is involved in heat-induced inflammatory cascades that support and immune activation, though excessive activation can exacerbate conditions like . In the bladder urothelium, TRPV1 functions as a for stretch and acidic environments, playing a key role in modulating the micturition reflex. Urothelial TRPV1 detects distension, facilitating ATP release that activates purinergic signaling to afferent , thereby helping regulate voiding thresholds and prevent overdistension. Acidic conditions (pH <6.5) directly gate TRPV1 channels in urothelial cells, enhancing sensory feedback during irritation and contributing to the coordination of normal bladder function, as evidenced by reduced ATP release and altered voiding patterns in TRPV1-deficient models. TRPV1 expression in vascular endothelial cells regulates vasodilation and vascular permeability, supporting hemodynamic homeostasis and tissue perfusion. Activation of endothelial TRPV1 by ligands such as anandamide induces calcium influx, which stimulates nitric oxide (NO) production via eNOS phosphorylation, promoting endothelium-dependent vasodilation in arteries like the mesenteric and cerebral vessels. Additionally, TRPV1 modulates endothelial barrier integrity; its stimulation enhances vascular permeability through cytoskeletal rearrangements, while in the blood-brain barrier, it can reduce permeability to limit edema during injury. In the gut epithelium, TRPV1 contributes to mucosal protection against environmental toxins and is implicated in the pathophysiology of irritable bowel syndrome (IBS). Epithelial TRPV1 activation by capsaicin or acidic toxins increases mucosal blood flow via CGRP release from associated sensory fibers, enhancing barrier integrity and mitigating damage from irritants like ethanol or excess acid in the stomach and intestines. In IBS, upregulated TRPV1 in colonic epithelium and submucosal layers correlates with visceral hypersensitivity, where bile acids and inflammatory mediators sensitize the channel, leading to altered motility and pain without overt inflammation. In pancreatic beta cells, TRPV1 is activated by endogenous ligands like anandamide, leading to calcium influx that promotes insulin secretion and contributes to glucose homeostasis. Similarly, in adipocytes, TRPV1 expression helps regulate energy balance by influencing lipolysis and preventing excessive visceral fat accumulation, with knockout models showing metabolic alterations. Recent studies from 2024–2025 indicate that peripheral TRPV1 activation can disrupt sleep via airway hypersensitivity.

Clinical Significance

Pain and Analgesia

TRPV1 plays a central role in the transduction of noxious stimuli, contributing to the perception of pain across various modalities. In neuropathic pain, TRPV1 acts as a "pain switch" through its sensitization, which heightens pain signaling in response to nerve injury, and desensitization, which can provide relief upon prolonged activation. In inflammatory pain, TRPV1 is sensitized by endogenous mediators such as protons and lipids, amplifying nociceptive responses in affected tissues. For migraine, TRPV1 is expressed in trigeminal nociceptive neurons, where its activation promotes neurogenic inflammation and headache pathogenesis, positioning it as a potential therapeutic target. Genetic variations in the TRPV1 gene influence individual sensitivity. Single-nucleotide polymorphisms (SNPs), such as those at positions affecting responsiveness, have been linked to altered thresholds for burning and thermal hypersensitivity. For instance, the TRPV1 1911 A>G polymorphism modulates sensory responses to , resulting in reduced heat sensitivity in variant carriers. These polymorphisms contribute to inter-individual differences in and response to analgesics. Desensitization-based analgesia exploits TRPV1's capacity for functional refractoriness following sustained . This mechanism involves calcium influx through TRPV1 channels, leading to channel and , which temporarily silences activity and reduces transmission. The duration of this analgesia varies by potency, dose, and application site, often lasting from weeks to months, providing prolonged relief without continuous dosing. A key example is the 8% topical patch Qutenza, approved by the FDA in 2009 for managing associated with . Applied for 60 minutes up to four times, it induces localized desensitization, significantly reducing scores for up to three months in clinical use. In , particularly (TMJ) disorders, TRPV1 activation in sensory afferents exacerbates inflammatory . Recent 2025 studies highlight TRPV1's role in TMJ pain modulation, with targeted agonists and antagonists showing promise in preclinical models for alleviating joint hypersensitivity and improving function. TRPV1 antagonists, while effective in blocking pathways, often induce as an on-target in clinical trials. This occurs through disruption of TRPV1-mediated in the and , leading to elevated core body temperature that can limit dosing and tolerability. In early-phase trials, such as with AMG517 targeting the site, transient occurred in approximately one-third of participants, prompting design of next-generation antagonists to mitigate this issue.

Inflammation and Immunity

TRPV1 channels are expressed on , where their activation induces calcium influx that activates the , leading to the processing and release of pro-inflammatory interleukin-1β (IL-1β). This calcium-dependent mechanism enhances macrophage inflammatory responses during innate immune activation. Additionally, TRPV1-mediated calcium entry promotes phagocytic activity in these cells by facilitating cytoskeletal rearrangements and engulfment of pathogens or debris, thereby supporting clearance in inflammatory environments. In adaptive immunity, TRPV1 is functionally expressed on CD4+ T cells, where it contributes to T cell receptor (TCR)-induced calcium signaling essential for cell activation, migration toward inflamed tissues, and cytokine production. Specifically, TRPV1 deficiency in CD4+ T cells impairs nuclear factor of activated T cells (NFAT) and nuclear factor kappa B (NF-κB) activation, resulting in reduced secretion of interferon-γ (IFN-γ) and other cytokines such as IL-2 and IL-17A upon stimulation. This role positions TRPV1 as a regulator of T cell-mediated adaptive responses in inflammatory conditions. TRPV1 also participates in innate immunity by influencing function, particularly through interactions with (LTB4), a potent chemoattractant. LTB4 activates TRPV1 on sensory neurons and potentially on neutrophils themselves, promoting calcium influx that drives and recruitment to sites of . This mechanism amplifies neutrophil infiltration and , exacerbating acute inflammatory responses. Recent 2024 studies highlight TRPV1's role as a neuroimmune bridge in , the brain's resident macrophages, where it modulates chronic inflammation. Microglia-specific TRPV1 deficiency accelerates glial activation, , and T cell infiltration in models of APOE4-related , underscoring its protective function against persistent . TRPV1 activation in suppresses inflammatory pathways, such as CaMKII/NRF2/SIRT3 signaling, thereby mitigating chronic inflammatory damage and neuronal dysfunction. In autoimmune diseases like (), TRPV1 expression on synovial fibroblasts and immune cells drives by enhancing production, including IL-6 and IL-8, in response to neuropeptides. TRPV1 knockout models show reduced synovial from early disease stages, indicating its contribution to joint destruction and immune dysregulation in RA pathogenesis. Targeting TRPV1 thus emerges as a potential strategy for modulating autoimmune inflammatory responses.

Cancer and Cell Death

TRPV1 is overexpressed in various cancers, including , , and malignancies, often correlating with advanced disease stages and poor patient prognosis. In , elevated TRPV1 expression increases with higher Gleason grades, serving as a prognostic indicator of tumor progression and reduced survival. Similarly, in , intracellular aggregation of TRPV1 is linked to lower overall survival rates among patients. For , high TRPV1 expression is observed in advanced stages, contrasting with reduced levels in low-grade tumors, suggesting its association with aggressive disease and unfavorable outcomes. Activation of TRPV1 by agonists induces calcium influx, leading to overload in mitochondria and subsequent in cancer cells. This mechanism involves excessive Ca²⁺ entry through the , disrupting mitochondrial and triggering pro-apoptotic pathways, as demonstrated in models of , , and other carcinomas. In TRPV1-expressing tumor cells, this Ca²⁺-dependent process promotes without affecting non-cancerous cells lacking the . Capsaicin, a prototypical , exhibits antitumor effects by suppressing proliferation through activation of -dependent pathways. In and cells, capsaicin or TRPV1 overexpression enhances p53 activity, leading to arrest and while inhibiting growth. This selective highlights TRPV1's potential as a target for inducing in p53-responsive tumors. Recent 2025 research has explored agonists derived from odorous plants, such as from peppers and from ginger, which activate TRPV1 to prevent cancer initiation and progression in preclinical models. Despite these antitumor roles, TRPV1 can paradoxically promote tumor progression in certain contexts, such as by enhancing angiogenesis. Channel activation regulates vascular endothelial growth factor (VEGF) expression and endothelial cell function, fostering blood vessel formation that supports tumor growth and metastasis. This dual functionality underscores the context-dependent nature of TRPV1 in oncogenesis.

Neurodegeneration and Neuroinflammation

TRPV1, a non-selective cation channel, has been implicated in the progression of neurodegenerative diseases through its modulation of calcium influx, which influences neuronal excitability, glial , and inflammatory cascades in the . In models of (AD), upregulation of TRPV1 expression occurs in response to amyloid-β (Aβ) peptides, leading to enhanced channel that exacerbates tau hyperphosphorylation and aggregation. This Aβ-induced TRPV1 promotes microglial metabolic reprogramming and lipid accumulation, contributing to synaptic dysfunction and cognitive decline, as demonstrated in 3×Tg-AD mouse models where TRPV1 antagonists or genetic modulation ameliorated tau pathology. In (), TRPV1 agonists such as have shown neuroprotective effects on neurons in the . Activation of astrocytic TRPV1 triggers the release of ciliary neurotrophic factor (CNTF), which rescues nigral neurons from degeneration in MPTP-induced models, restoring signaling and reducing motor deficits. Studies from 2023 highlight that (1 mg/kg, intraperitoneal) inhibits glial-mediated and , increasing tyrosine hydroxylase-positive neurons and preventing α-synuclein-induced toxicity in rodent models. Regarding mitochondrial dysfunction, TRPV1 knockout in neurodegeneration models attenuates by preventing excessive calcium entry into mitochondria, which otherwise leads to (ROS) overproduction and activation. In cellular assays and models, TRPV1 elevates mitochondrial calcium levels, promoting and energy deficits, whereas knockout variants exhibit reduced ROS and preserved mitochondrial integrity. This protective effect of TRPV1 underscores its , where chronic activation contributes to bioenergetic failure in vulnerable neurons. TRPV1 also influences vascular aspects of brain aging, where capsaicin-mediated activation mitigates in cerebral vessels. In 2024 investigations using aged rat models, (doses of 5-10 mg/kg) improved endothelial production and reduced vascular stiffness by modulating TRPV1-dependent , thereby alleviating age-related hypoperfusion that exacerbates neurodegeneration. These findings suggest TRPV1 agonism as a strategy to preserve blood-brain barrier integrity during vascular aging. Emerging evidence links hypothalamic TRPV1 to -wake regulation, particularly through its role in thermosensory and processes. In models, TRPV1-expressing neurons in the dorsomedial and paraventricular hypothalamic nuclei modulate transitions. In the context of , TRPV1 briefly ties to central immune responses by facilitating calcium-dependent activation in , amplifying release that bridges peripheral signals to , though detailed immune mechanisms are addressed elsewhere.

Protein Interactions

Binding Partners

TRPV1, a non-selective cation primarily expressed in sensory neurons, interacts with several proteins that modulate its localization, activity, and through direct binding. These interactions often occur at specific structural domains, such as the intracellular N- and C-termini, influencing gating, desensitization, and membrane trafficking. β-Arrestin-2 binds to the C-terminal domain of TRPV1, acting as a scaffold to promote desensitization by recruiting phosphodiesterase PDE4D5, which hydrolyzes and facilitates of TRPV1 at key serine residues. Calmodulin associates with both the N-terminal ankyrin repeat domain and the C-terminal region of TRPV1 in a calcium-dependent manner, with distinct affinities to mediate rapid desensitization following calcium influx through the . This inhibits further opening, providing a mechanism to prevent in sensory neurons. Src interacts with TRPV1 to phosphorylate residues, enhancing sensitization in response to ; this is crucial for amplifying TRPV1 activity under inflammatory conditions. TRPV1 forms functional heterodimers with , another co-expressed in sensory neurons, through associations involving their transmembrane domains; this heteromerization results in channels responsive to TRPV1 agonists like , heat, and protons, as well as PKC sensitization relevant to , but insensitive to TRPA1-specific stimuli such as cold and , thereby modulating polymodal detection capabilities. Mass spectrometry-based interactomics in neurons has revealed numerous TRPV1 binding partners that vary with states, including vesicle trafficking proteins like Vti1b, which stabilizes TRPV1 at the during to promote . These studies underscore the dynamic nature of the TRPV1 interactome in neuronal contexts. Additionally, TRPV1 interacts with A-kinase anchoring protein 79/150 (AKAP79/150) at the C-terminal domain, which scaffolds (PKA) and (PKC) to facilitate phosphorylation-dependent .

Signaling Pathways

Activation of the transient receptor potential vanilloid 1 (TRPV1) channel primarily initiates signaling through influx of calcium ions (Ca²⁺), which serves as a key second messenger to trigger diverse downstream cascades. This Ca²⁺ entry depolarizes cells and activates various effectors, including kinases and transcription factors, leading to cellular responses such as sensitization, , and modulation of . In immune cells, particularly CD4⁺ T lymphocytes, TRPV1-mediated Ca²⁺ influx contributes to the nuclear factor of activated T cells (NFAT) pathway, promoting gene transcription essential for immune activation. Upon stimulation, TRPV1 facilitates sustained Ca²⁺ entry, which activates to dephosphorylate NFAT, enabling its translocation to the nucleus and upregulation of proinflammatory like IFN-γ and IL-17A. Studies in Trpv1-deficient T cells demonstrate reduced NFAT nuclear localization and cytokine production, highlighting TRPV1's role in enhancing immune responses. TRPV1 activation also engages the /extracellular signal-regulated kinase (MAPK/ERK) pathway, which underlies channel sensitization and . In sensory neurons, Ca²⁺ influx from TRPV1 stimulates ERK via upstream kinases like 3-kinase (PI3K), amplifying TRPV1 responsiveness to stimuli and contributing to . This pathway extends to non-neuronal cells, where sustained ERK activation promotes , as observed in models of where TRPV1 agonists enhance ERK signaling to drive cell growth. Furthermore, TRPV1-induced Ca²⁺ entry promotes (ROS) production through mitochondrial Ca²⁺ uptake, disrupting mitochondrial function and amplifying . The influxed Ca²⁺ is taken up by mitochondria via the mitochondrial calcium uniporter, leading to membrane depolarization and ROS generation from the , which can exacerbate cellular damage in stressed environments. This mechanism has been evidenced in fibroblasts and neurons, where blocking TRPV1 reduces mitochondrial Ca²⁺ overload and subsequent ROS levels. TRPV1 exhibits crosstalk with (CB1), influencing analgesic signaling through shared cellular compartments in sensory neurons. Activation of CB1 can desensitize TRPV1 via G-protein-coupled inhibition of , reducing levels and attenuating Ca²⁺ responses to TRPV1 agonists, thereby mediating analgesia in pain models. This interaction is particularly relevant in dorsal root ganglia, where co-localization allows CB1 ligands to modulate TRPV1 hyperactivity. Recent findings from 2024 link TRPV1 to the in , where TRPV1-driven Ca²⁺ signaling activates NLRP3 assembly in and macrophages. In models of , TRPV1 upregulation enhances Ca²⁺-dependent NLRP3 priming and activation, leading to IL-1β release and neuroinflammatory cascades; inhibiting TRPV1 suppresses this pathway and mitigates . This underscores TRPV1's emerging role in inflammasome-mediated neuroimmune responses.

Therapeutic Applications

Agonist-Based Strategies

Agonist-based strategies for TRPV1 leverage the channel's activation to induce desensitization of nociceptive neurons or promote pro-apoptotic effects in targeted cells, offering therapeutic potential in and beyond. High-dose , a prototypical TRPV1 agonist, is administered via patches or intra-articular injections to achieve prolonged analgesia through selective defunctionalization of TRPV1-expressing sensory afferents. The 8% patch (Qutenza) has demonstrated efficacy in reducing intensity in clinical settings, with studies confirming its benefits for conditions like and . For , the CADOR study, a Phase III trial protocol published in 2025, aims to evaluate a single 60-minute application of 8% versus low-dose controls for digital osteoarthritis pain over 60 days. Intra-articular injections of high-dose (RTX; e.g., 0.05-0.15 µg doses in formulations like RTX-GRT7039) have provided sustained relief in knee , with Phase III trials ongoing as of 2025 (e.g., NCT05248386) demonstrating pain reduction lasting up to 12 weeks without systemic opioids. These approaches exploit the agonist's ability to cause calcium influx and subsequent degeneration of TRPV1-positive nerve terminals, minimizing signaling. Resiniferatoxin (RTX), an ultrapotent agonist approximately 1,000 times more potent than , enables site-specific delivery for cancer by targeting TRPV1-expressing tumor cells or innervating nerves. Intrathecal or of RTX ablates TRPV1-positive sensory neurons, providing long-lasting analgesia in advanced cancer patients to , as evidenced by a 2025 NIH I where low-dose RTX reduced worst intensity by 38% and consumption by 57% for months while preserving non-nociceptive sensations. In preclinical models, RTX conjugates or direct application to TRPV1-overexpressing cancer cells induce calcium overload and , potentially shrinking tumor volume; for instance, studies in and models showed RTX-mediated defunctionalization of TRPV1+ afferents and direct , reducing and tumor burden simultaneously. Preclinical investigations suggest potential for intravesical RTX in , with site-specific delivery via achieving localized effects without widespread neuronal loss. Herbal-derived TRPV1 agonists, such as derivatives from officinale, offer milder activation for anti-inflammatory applications by modulating immune responses without severe desensitization. 6- and related compounds bind TRPV1 to inhibit pro-inflammatory release (e.g., TNF-α and IL-6) in macrophages and endothelial cells, contributing to ginger's traditional use in reducing and gastrointestinal inflammation. and animal studies confirm that gingerols at micromolar concentrations activate TRPV1 to suppress signaling, yielding anti-inflammatory effects comparable to low-dose but with fewer acute sensations, as detailed in seminal work on natural TRP modulators. Despite these advances, agonist-based therapies face challenges including initial pain flare from acute TRPV1 activation and potential cardiovascular effects. Application of high-dose or RTX often triggers intense burning pain due to rapid calcium influx and release, necessitating pre-treatment with local anesthetics in up to 80% of patients to manage this transient exacerbation lasting 30-60 minutes. Cardiovascular side effects, such as transient or from reflex sympathetic activation, occur in 10-20% of cases, particularly with systemic exposure, though these are generally mild and resolve within hours. In 2025, research on and TRPV1 highlights potential for mitigating vascular aging through vasodilatory and anti-senescence effects, improving vascular stiffness in aging models.

Antagonist-Based Strategies

Antagonist-based strategies for TRPV1 aim to block channel activation, thereby reducing signaling and related pathologies without the desensitization associated with agonists. These approaches primarily target - or proton-induced activation to provide analgesia, while efforts focus on minimizing off-target effects like . Small-molecule antagonists have been the most explored class, with clinical translation challenged by selectivity issues. NEO6860 represents a prototypical small-molecule TRPV1 designed for selectivity, inhibiting capsaicin-evoked activation while sparing heat and pH responses to avoid and heat insensitivity. In a Phase II proof-of-concept trial for (2018), NEO6860 demonstrated modest effects, reducing pain scores compared to baseline, but failed to outperform or naproxen in efficacy endpoints. The trial highlighted an unfavorable safety profile, including higher exposure levels than in Phase I and increased adverse events relative to controls, leading to discontinuation in the due to on-target tolerability concerns despite its selective design. Monoclonal antibodies targeting the extracellular domain of TRPV1 offer a promising for management, leveraging their specificity and long half-life to achieve sustained inhibition without penetrating the blood-brain barrier extensively. has focused on modality-selective antibodies that block binding while preserving heat-gated function, using to screen for extracellular epitopes accessible . These antibodies demonstrated selective antagonism in and assays, reducing nociceptive responses in pain models without inducing , positioning them as candidates for clinical evaluation in neuropathic and inflammatory pain conditions. Allosteric inhibitors of TRPV1 have been investigated to disrupt pro-tumor signaling in cancers where channel activation promotes and , such as in and tumors. By modulating non-competitive sites, these inhibitors aim to suppress calcium influx that drives oncogenic pathways without fully ablating physiological responses. In preclinical models, allosteric blockade reduced tumor growth and by inhibiting TRPV1-mediated Ca²⁺ signaling in the , highlighting potential adjunctive roles in cancer therapy. Recent advancements in 2024-2025 have introduced selective TRPV1 antagonists for specialized indications like and neurodegeneration. For , JTS-653, a selective , alleviated arthritic and in preclinical rodent models by blocking TRPV1 sensitization in trigeminal neurons, with a 2025 review underscoring its potential for clinical translation in temporomandibular disorders. In neurodegeneration, , a TRPV1 , attenuated astrocyte activation and in models of , mitigating neuronal damage via TRPV1-independent pathways while preserving cognitive function. The safety profile of TRPV1 antagonists has improved through biased allosteric modulation, which decouples analgesia from thermoregulatory disruptions. Conventional antagonists often induce hyperthermia by blocking proton-gated TRPV1 in the central nervous system, but biased compounds like PSFL2874 avoid binding to the S4-S5 linker region, preserving core body temperature regulation while effectively relieving inflammatory pain in murine models. This approach enables safer long-term use by maintaining heat sensitivity and minimizing cardiovascular risks.

History and Discovery

Initial Identification

The initial identification of TRPV1, originally termed the vanilloid receptor 1 (VR1), occurred through expression cloning from a . In 1997, Michael J. Caterina and colleagues employed a calcium imaging-based screening strategy in HEK293 cells to isolate clones that conferred responsiveness to , the pungent component of peppers known to activate nociceptive sensory neurons. This approach identified a single cDNA encoding VR1, a protein predicted to form a non-selective cation channel with six transmembrane domains and intracellular ankyrin repeats, homologous to members of the transient (TRP) family. Upon in HEK293 cells and oocytes, VR1 was characterized as a ligand-gated that mediates robust calcium influx in response to , with an EC₅₀ of approximately 710 nM, and is antagonized by (IC₅₀ ≈ 280 nM). Electrophysiological recordings revealed that VR1 currents exhibit desensitization upon repeated application in the presence of extracellular calcium, reducing responses by up to 87% over multiple exposures, a phenomenon absent in calcium-free conditions. Notably, VR1 demonstrated sensitivity to noxious , activating at temperatures around 45°C to produce inward currents comparable to those elicited by , and its responses to were potentiated up to fivefold by acidic conditions ( 6.3), though low alone did not sufficiently activate the . These findings established VR1 as a molecular of chemical and signals. The nomenclature evolved in 2002 when VR1 was redesignated as TRPV1 to reflect its membership in the vanilloid subfamily of TRP channels, as part of a unified system for the TRP superfamily.

Key Milestones

The study of , the active compound responsible for the pungency of peppers, dates back to the 19th century, with early observations of its physiological effects laying the groundwork for understanding TRPV1 function. In 1878, Endre Hõgyes reported that capsaicin administration in dogs induced and degeneration of s, hinting at its selective action on peripheral nerves. By the 1940s, Nicholas Jancsó demonstrated that systemic capsaicin caused selective degeneration of small-diameter sensory neurons and long-term desensitization to pain, establishing capsaicin-sensitive afferents as a distinct population involved in . These findings, further elaborated in the 1960s by János Szolcsányi and colleagues, revealed capsaicin's role in neurogenic and sensory nerve excitation, prompting searches for its molecular target. A pivotal advancement occurred in 1994 with the synthesis and identification of , the first competitive antagonist of 's excitatory effects on sensory neurons, which facilitated pharmacological dissection of capsaicin-sensitive pathways years before receptor cloning. The molecular era began in 1997 when Michael Caterina, Mark Schumacher, and David Julius cloned the capsaicin receptor, named vanilloid receptor 1 (VR1) or TRPV1, from rat dorsal root ganglia using expression cloning in HEK293 cells; they showed it to be a heat-activated, non-selective cation channel expressed in nociceptive neurons, integrating and noxious (>43°C) stimuli to trigger signaling. This discovery, published in , provided the first molecular explanation for capsaicin-induced burning and thermal nociception. In 1998, Makoto Tominaga and colleagues extended these insights by demonstrating TRPV1's sensitization by protons (low pH), linking it to inflammatory conditions like . Subsequent milestones illuminated TRPV1's structure and broader roles. In 1999, TRPV1 was shown to respond to endogenous lipids like anandamide, expanding its function beyond exogenous ligands to include endocannabinoid signaling in pain modulation. The channel's tetrameric structure was resolved in 2013 by Maofu Liao, Erhu Cao, David Julius, and Yifan Cheng using cryo-electron microscopy at 3.4 Å resolution, revealing key domains for ligand binding, heat gating, and ion permeation, which accelerated drug design efforts. Clinical translation advanced with the development of TRPV1 antagonists entering trials in the mid-2000s, such as SB-705498, which demonstrated efficacy in pain models but faced challenges like hyperthermia. The field's impact was recognized in 2021 when David Julius and Ardem Patapoutian received the Nobel Prize in Physiology or Medicine for discovering TRPV1 and other thermosensory receptors, underscoring their role in decoding temperature and touch.

References

  1. [1]
    The capsaicin receptor: a heat-activated ion channel in the pain ...
    Oct 23, 1997 · Capsaicin, the main pungent ingredient in 'hot' chilli peppers, elicits a sensation of burning pain by selectively activating sensory neurons.Vr1 And Vanilloid Receptor... · Vr1 Ion Channel Has High Ca... · Vanilloid Receptor Activated...
  2. [2]
    TRP (transient receptor potential) ion channel family - Nature
    Jul 5, 2023 · TRPV1 is a heat-activated cation channel modulated by inflammatory mediators and contributes to acute and chronic pain. About 30% of the TRPA1- ...
  3. [3]
    Structure of the TRPV1 ion channel determined by electron cryo ...
    Dec 4, 2013 · Here we exploit advances in electron cryo-microscopy to determine the structure of a mammalian TRP channel, TRPV1, at 3.4 Å resolution.
  4. [4]
    The capsaicin receptor: a heat-activated ion channel in the pain ...
    Capsaicin, the main pungent ingredient in 'hot' chilli peppers, elicits a sensation of burning pain by selectively activating sensory neurons.
  5. [5]
  6. [6]
  7. [7]
  8. [8]
    The Ankyrin Repeats of TRPV1 Bind Multiple Ligands and Modulate ...
    Jun 21, 2007 · We describe the structure of the cytosolic ankyrin repeat domain (ARD) of TRPV1 and identify a multiligand-binding site important in regulating channel ...
  9. [9]
    Structural Insights into the Function of TRP Channels - NCBI
    The TRPV1 N-terminus is important for calmodulin regulation of TRPV1 [36] ... Identification of a tetramerization domain in the C-terminus of the vanilloid ...The Transmembrane Region · Cytosolic Domains · Ankyrin Repeats
  10. [10]
    Distinct properties of Ca2+–calmodulin binding to N- and C-terminal ...
    Two CaM-binding sites are identified in TRPV1: the N-terminal ankyrin repeat domain (ARD) and a short distal C-terminal (CT) segment. Here, we present the ...
  11. [11]
    The Integrity of the TRP Domain Is Pivotal for Correct TRPV1 ...
    Collectively, our findings imply that the integrity of the TRP domain in TRPV1 is important for fine-tuning stimuli sensing and gate opening, and that even ...
  12. [12]
    Identification of a Tetramerization Domain in the C Terminus of the ...
    TRPV1 (transient receptor potential vanilloid receptor subtype 1) is a member of the TRP channel family gated by vanilloids, protons, and heat.
  13. [13]
    S800 is a polymodal sensitization site - PMC - PubMed Central - NIH
    Our data suggest that TRPV1 S800 is an important site for PKC-induced hypersensitivity both in HEK293 cells and sensory neurons. These results were obtained by ...
  14. [14]
    Activation of NMDA receptors leads to phosphorylation of TRPV1 ...
    ... TRPV1 phosphorylation [30-32]. PKC phosphorylates TRPV1 at S502, S800 [23, 32-33], and threonine (T) 704, whereas PKA activation results in the phosphorylation ...
  15. [15]
    Capsaicin Interaction with TRPV1 Channels in a Lipid Bilayer
    In particular, mutating a tyrosine residue on the cytoplasmic side of the S3 helix (Y511) (Fig. 1 B) to alanine removes capsaicin sensitivity while the channel ...
  16. [16]
    A molecular perspective on identifying TRPV1 thermosensitive ...
    In this review, we summarize molecular studies of TRPV1 thermosensing, focusing on the cross-talk between heat and other activation modes.Introduction · The Trpv1 Ion Channel Is... · Abbreviations
  17. [17]
    TRPV1 transient receptor potential cation channel subfamily ... - NCBI
    Sep 5, 2025 · The protein encoded by this gene is a receptor for capsaicin and is a non-selective cation channel that is structurally related to members of the TRP family of ...Missing: IWxxIW | Show results with:IWxxIW
  18. [18]
    Entry - *602076 - TRANSIENT RECEPTOR POTENTIAL CATION ...
    The initiation ATG is located within exon 2. Cortright et al. (2001) determined that the human TRPV1 gene contains 18 exons and spans at least 32 kb.
  19. [19]
  20. [20]
    TRPV1 splice variants: structure and function - PMC - PubMed Central
    The N - terminal intracellular region is distinguished by three ankyrin repeat domains that participate in channel stability, activation and modulation. The ...Trpv1 Splice Variants... · 3. Trpv1: Structure And... · 4. Trpv1 Splice Variants
  21. [21]
    TRPV1 splice variants: structure and function - PubMed - NIH
    Jun 1, 2010 · This review is focused on the structural and functional consequence of TRPV1 splice variants: VR.5'sv, TRPV1b/beta and TRPV1var.
  22. [22]
    Transcription factors Sp1 and Sp4 regulate TRPV1 gene expression ...
    Jun 6, 2011 · ... NGF in primary sensory neurons after inflammation ... TRPV1 utilizes a dual promoter system that is positively regulated by nerve growth factor.
  23. [23]
    TRPV1-polymorphism 1911 A>G alters capsaicin-induced sensory ...
    (2) Capsaicin stimulation induces less warm hypoesthesia and less heat pain sensitivity in TRPV1 variant carriers. Thus, our results are not in line with ...
  24. [24]
    Effect of single-nucleotide polymorphisms in TRPV1 on burning pain ...
    We detected 6 single-nucleotide polymorphisms and 11 single-nucleotide polymorphisms related to burning pain and capsaicin sensitivity, respectively.
  25. [25]
    Dynamic evolution of transient receptor potential vanilloid (TRPV ...
    Nov 1, 2022 · The conservation of synteny confirms orthology of TRPV1 across chondrichthyans, sarcopterygians and actinopterygians. Some of the ...
  26. [26]
    Beyond Neuronal Heat Sensing: Diversity of TRPV1 Heat-Capsaicin ...
    The purpose of the present review is to provide a clear-cut distinction between neurogenic TRPV1 effects in various tissues consequent to its activation in ...
  27. [27]
    The role of TRPV1 in different subtypes of dorsal root ganglion ...
    We found that TRPV1 expression increased in distinct small DRG neurons. As shown in Fig. 4, TRPV1 increased within CGRP positive neurons from day 1 to day 28 ( ...
  28. [28]
    PMC - PubMed Central
    May 24, 2024 · The expression of TRPV1 has been confirmed in various immune cells, including T lymphocytes, macrophages, natural killer cells, dendritic ...Missing: vascular | Show results with:vascular
  29. [29]
    Endothelial TRPV1 as an Emerging Molecular Target to Promote ...
    TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis.
  30. [30]
    Regulation of transient receptor potential cation channel subfamily ...
    The present study investigated the underlying molecular mechanisms by which TRPV1 was regulated by nerve growth factor (NGF) signaling pathways in colonic ...<|separator|>
  31. [31]
    Molecular and Functional Study of Transient Receptor Potential ...
    Nov 10, 2020 · Our present study reveals an interspecies difference between Rat and Human. Functional contributions of TRPV1-4 subtype expression were not identical in rat ...
  32. [32]
    Interaction between TRPV1-expressing neurons in the hypothalamus
    Our study demonstrated the functional expression of TRPV1 in the dorsomedial hypothalamic nucleus and paraventricular nucleus in adult mice.Missing: seq 2020s
  33. [33]
    Carcinogenesis and Metastasis: Focus on TRPV1-Positive Neurons ...
    Besides the hypothalamus, TRPV1 is expressed in the hippocampus and midbrain and the expression is not only found in neurons but also in glial cells and ...Missing: RNA- seq
  34. [34]
    Protein kinase C phosphorylation sensitizes but does not activate ...
    Protein kinase C (PKC) modulates the function of the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1).
  35. [35]
    TRPV1 Receptors and Signal Transduction - NCBI
    TRPV1 is a TRP ion channel activated by capsaicin, heat, and low pH, acting as a molecular transducer for pain, and has six transmembrane domains.INTRODUCTION · TRPV1 ACTIVATORS · MODULATION OF TRPV1...
  36. [36]
  37. [37]
    The activation mechanism of rat vanilloid receptor 1 by capsaicin ...
    Electrophysiological studies of expressed VR1 receptors show that they are activated independently by capsaicin (EC50 = 0.7 μM), acid (pH50 = 5.4), and elevated ...
  38. [38]
    mechanism of TRPV1 channel activation by capsaicin | Protein & Cell
    Jan 2, 2017 · The success of cryo-EM study of TRPV1 structure has marked a new era of structural biology for membrane proteins. For TRP channels, structures ...
  39. [39]
    resiniferatoxin | Ligand Activity Charts
    TRPV1/Vanilloid receptor in Human [ChEMBL: CHEMBL4794] [GtoPdb: 507] ... -, 8.4, pEC50, 3.98, nM, EC50, Eur J Pharmacol (2001) 417: 51-8 [PMID:11301059].
  40. [40]
    Activation of transient receptor potential vanilloid 1 (TRPV1) by ...
    Resiniferatoxin (RTX) is an ultrapotent agonist that activates TRPV1, a non-selective cation channel, and is the most potent known agonist.
  41. [41]
    The pungent substances piperine, capsaicin, 6-gingerol and ... - NIH
    In addition, they have been shown to be modulated by several TRP agonists. We investigated whether the pungent substances piperine, capsaicin, 6-gingerol and ...
  42. [42]
    Uncoupling Proton Activation of Vanilloid Receptor TRPV1
    Nov 21, 2007 · TRPV1, a pain receptor involved in nociception at the peripheral nerve terminals, can be activated by a range of physical and chemical stimuli.Materials And Methods · Results · Specificity To Proton Gating<|control11|><|separator|>
  43. [43]
    Temperature-induced opening of TRPV1 ion channel is stabilized by ...
    Caterina MJ, et al. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature. 1997;389:816–824. doi: 10.1038/39807. [DOI] [PubMed] ...Missing: paper | Show results with:paper<|separator|>
  44. [44]
    TRPV1: A Potential Drug Target for Treating Various Diseases - PMC
    TRPV1 is an ion channel on sensory neurons, activated by heat, protons, and capsaicin, and is a potential drug target for conditions like chronic pain and ...2. Endogenous Trpv1 Agonists · 3.1. Role Of Trpv1 In... · 4. Trpv1 Antagonists<|control11|><|separator|>
  45. [45]
  46. [46]
    Ligand-Binding Sites in Vanilloid-Subtype TRP Channels - PMC
    May 16, 2022 · In TRPV1, two copies of the agonist DkTx bind to the extracellular side of the channel in a two-fold symmetrical manner, with each knot of this ...
  47. [47]
    Pharmacological blockade of the vanilloid receptor TRPV1 elicits ...
    AMG 517 caused hyperthermia by inducing tail skin vasoconstriction and increasing thermogenesis, which suggests that TRPV1 regulates vasomotor tone and ...
  48. [48]
    Investigation of selected 13 FDA approved drugs for TRPV1 ... - RJPT
    Hence its can be concluded that Rutin and glimepiride exhibited best possible interaction by antagonizing the effect of TRPV1 receptor so can be consider for ...
  49. [49]
    Molecular details of ruthenium red pore block in TRPV channels
    Jan 15, 2024 · TRPV channels are blocked by ruthenium red (RR), a universal pore blocker for a wide array of cation channels. Here we use cryo-electron microscopy to reveal ...Missing: natural flavonoids
  50. [50]
    Extracellular Cations Sensitize and Gate Capsaicin Receptor ...
    May 25, 2005 · We demonstrate here that cations gate TRPV1 directly and that this mechanism may contribute to the nociceptive response to elevated ionic strength.
  51. [51]
    Divalent cations activate TRPV1 through promoting conformational ...
    Divalent cations Mg2+ and Ba2+ selectively and directly potentiate transient receptor potential vanilloid type 1 heat activation by lowering the activation ...
  52. [52]
    Advances in TRP channel drug discovery: from target validation to ...
    Sep 15, 2021 · Albeit at much lower levels, TRPV1 is also expressed in various brain nuclei and in non-neuronal cells. Fig. 2: TRPV1 in the pain pathway: ...
  53. [53]
    TRPV1: Structure, Endogenous Agonists, and Mechanisms - PMC
    May 12, 2020 · The Transient Receptor Potential Vanilloid 1 (TRPV1) channel is a polymodal protein with functions widely linked to the generation of pain.<|control11|><|separator|>
  54. [54]
    Modulation of trigeminal sensory neuron activity by the dual ...
    AEA (EC(50)=11.0 microM), NADA (EC(50)=857 nM) and arachidonyl-2-chloroethylamide ACEA (EC(50)=14.0 microM) each evoked calcitonin gene-related peptide (CGRP) ...
  55. [55]
    TRP channels interaction with lipids and its implications in disease
    Endogenous TRPV1 lipid ligands also include LOX metabolites such as 12-(S)-hydroperoxyeicosa-5Z,8Z,10E,14Z-tetraeonic acid 12-(S)HPETE, 15-(S)HPETE and ...
  56. [56]
    Leukotriene B4 Mediates Inflammation via TRPV1 in Duct ...
    We tested the hypothesis that leukotriene B4 (LTB4) mediates pancreatic inflammation in rats via activation of the Transient Receptor Potential Vanilloid 1 ...
  57. [57]
    The role of endogenous molecules in modulating pain through ...
    Apr 24, 2013 · In summary, TRPV1 is regulated by heat, exogenous chemicals (e.g. spices), voltage, and endogenous compounds including acid, bases, ions, gases ...
  58. [58]
    Divalent cations potentiate TRPV1 channel by lowering the heat ...
    Dec 16, 2013 · We estimated that, at saturating capsaicin concentrations and +80 mV, the mouse TRPV1 activation reaches an open probability of ∼80%. Based ...
  59. [59]
    Localization of the PIP2 Sensor of TRPV1 Ion Channels - PMC - NIH
    It has recently been shown that depletion of PIP2 in cells leads to TRPV1 channel deactivation (10, 11), indicating that PIP2 is an activator rather than an ...
  60. [60]
    TRPV1 Channels Are Intrinsically Heat Sensitive and Negatively ...
    Feb 20, 2013 · Phosphoinositide lipids function as important regulatory factors for many types of membrane ion channels and transporters, including members of ...<|control11|><|separator|>
  61. [61]
    Dual Regulation of TRPV1 by Phosphoinositides
    Jun 27, 2007 · The membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2 or PIP2] regulates many ion channels.
  62. [62]
    Eicosapentaenoic Acid Modulates Transient Receptor Potential V1 ...
    Mar 1, 2024 · The present results provide insights into the role of the TRPV1 signaling pathway for fibromyalgia and its potential as a clinical target.
  63. [63]
    Polyunsaturated fatty acids and their endocannabinoid-related ...
    Mar 24, 2025 · Polyunsaturated fatty acids and their endocannabinoid-related metabolites activity at human TRPV1 and TRPA1 ion channels expressed in HEK-293 cells.
  64. [64]
    Emerging Role of Spinal Cord TRPV1 in Pain Exacerbation - PMC
    A series of recent reports on the spinal cord synapses have provided evidence that TRPV1 plays an important role in synaptic transmission in the pain pathway.
  65. [65]
    Inflammatory Pain: The Cellular Basis of Heat Hyperalgesia - PMC
    Knockout studies have shown that mice lacking the TRPV1 receptor fail to develop thermal hyperalgesia during inflammation [18,25].
  66. [66]
    Sensitization of small‐diameter sensory neurons is controlled by ...
    Nov 21, 2019 · However, the co-expression and interaction of TRPA1 with TRPV1 proved to be the most critical for differential sensitization of sensory neurons.
  67. [67]
    TRPV1: Role in Skin and Skin Diseases and Potential Target for ...
    Jun 7, 2021 · In skin, keratinocytes, sebocytes, nociceptors, and several immune cells express TRPV1, making it an attractive focus area for treating wounds.
  68. [68]
  69. [69]
    TRPs in bladder diseases - PMC - PubMed Central - NIH
    This review attempts to provide an overview of the current knowledge of TRP proteins and their possible role in bladder function and disease.
  70. [70]
  71. [71]
    Recent Developments in Vascular Endothelial Cell Transient ...
    Oct 28, 2005 · For example, activation of TRPV1 causes endothelium-dependent vascular dilation, and it also increases vascular permeability. Another ...
  72. [72]
  73. [73]
    TRP channels in the digestive system - PMC - NIH
    Through this type of mechanism TRPV1-positive sensory nerve fibres are able to protect the oesophageal, gastric and intestinal mucosa from a variety of ...
  74. [74]
  75. [75]
    TRPV1: a new target for treatment of visceral pain in IBS? - PMC - NIH
    TRPV1 is an IBS drug target with high potential, if its upregulation in this disorder reflects a causal implication rather than just a marker of enhanced ...Missing: epithelium | Show results with:epithelium
  76. [76]
  77. [77]
    The roles of TRPV1 receptors in nervous system with a special ...
    Jan 26, 2025 · TRPV1 receptors, which are distributed at all levels of the nervous system, regulate nerve reflexes and sensory transduction in response to sleep disturbances.
  78. [78]
  79. [79]
    The dual role of TRPV1 in peripheral neuropathic pain
    Sep 9, 2024 · TRPV1 plays a dual role in peripheral NeuP, acting as a “switch” for pain through its sensitization and desensitization processes. In CIPN and ...
  80. [80]
    Role of TRPV1 in Health and Disease - Xia & He Publishing Inc.
    Likewise, there is substantial evidence revealing that NGF produced after inflammation and/or tissue injury has an impact on a regulatory region located ...Introduction · Trpv1 Structure · Trpv1 In Health<|control11|><|separator|>
  81. [81]
    TRPA1 and other TRP channels in migraine
    Aug 13, 2013 · In particular, the TRP vanilloid 1 (TRPV1) and the TRP ankyrin 1 (TRPA1) are expressed in nociceptive neurons, which also express the sensory ...Review · Trpa1, Pain And Neurogenic... · Trpa1, Trpv1, And Migraine
  82. [82]
    TRP Channels: Recent Development in Translational Research and ...
    In headache and pain research, TRPV1 is a very promising target for the development of new analgesics for the treatment of inflammatory and neuropathic pain.Trp Channels: Recent... · 1. Migraine And Other... · 1.2. Migraine
  83. [83]
    Polymorphisms in gene encoding TRPV1-receptor involved in pain ...
    Dec 24, 2009 · The aim of our study was to investigate the genetic variation of TRPV1 in Dutch patients with chronic pancreatitis and healthy controls.
  84. [84]
    Calcium-dependent desensitization of vanilloid receptor TRPV1
    This review summarizes the current state of knowledge concerning the mechanisms that underlie the acute capsaicin-induced Ca2+-dependent desensitization of ...
  85. [85]
    Agonist- and Ca2+-dependent Desensitization of TRPV1 Channel ...
    Depending on the vanilloid, dose, and administration site, nociceptor refractoriness may last from minutes up to months, suggesting the contribution of ...
  86. [86]
    FDA approves treatment for pain relief of post-herpetic neuralgia
    Nov 18, 2009 · The FDA has approved the approval of a capsaicin 8% medicated skin patch (Qutenza, Neurogesx) to treat pain associated with post-herpetic ...
  87. [87]
    [PDF] QUTENZA® (capsaicin) patch - accessdata.fda.gov
    The recommended dose of QUTENZA for neuropathic pain associated with postherpetic neuralgia is a single, 60-minute application of up to four patches. 2.
  88. [88]
    TRPV1-target drugs for the treatment of orofacial pain - Frontiers
    Apr 23, 2025 · This review focuses on the role of Transient Receptor Potential Vanilloid 1 (TRPV1) channels in modulating orofacial pain and new ligands targeting this ...
  89. [89]
    Transient receptor potential channels in dental inflammation and ...
    Jan 30, 2025 · Activation of TRPV1 in these nerve endings might contribute to pain sensation associated with TMJ disorders. Acute temporomandibular disorder ...Research Article · 2.1. Pulpitis · 2.2. Dentin Hypersensitivity
  90. [90]
    Hyperthermia induced by transient receptor potential vanilloid-1 ...
    However, during the in-vivo testing of TRPV1 antagonists, adverse effects on body temperature (Tb), primarily hyperthermia, were repeatedly observed in animal ...
  91. [91]
    TRPV1 analgesics disturb core body temperature via a biased ...
    Mar 15, 2024 · All four antagonists effectively blocked the activation of TRPV1 currents by capsaicin (30 nM for AMG9810, 3 nM for AMG517, 100 nM for SB366791, ...Missing: capsazepine | Show results with:capsazepine
  92. [92]
    TRPV1 channel mediates NLRP3 inflammasome-dependent ...
    Dec 14, 2021 · These data suggested that the TRPV1 channel in microglia can regulate NLRP3 inflammasome activation and consequently mediate neuroinflammation.
  93. [93]
    TRPV1: The key bridge in neuroimmune interactions - ScienceDirect
    The TRPV1 protein is encoded by the TRPV1 gene located on chromosome 17p13 and has a molecular weight of 95 KD. It is a tetrameric protein consisting of ...
  94. [94]
    What is the evidence for the role of TRP channels in inflammatory ...
    Nov 25, 2015 · This review discusses the roles of TRP channels in inflammatory and immune cell function and provides an overview of the effects of inflammatory and immune TRP ...Trpm2 Channels · Trpc1, Trpc3, Trpc5 And... · Trp Channels In Immune And...
  95. [95]
    Transient Receptor Potential Vanilloid1 (TRPV1) Channel Opens ...
    In this review, we analyze recent data on the expression and functions of TRPV1 in T cells and T cell-mediated inflammatory diseases. The data showed that TRPV1 ...
  96. [96]
    key elements in the mediation of leukotriene B4-induced itch by ...
    We investigated the role of TRPV1 and TRPA1 in mediating leukotriene B4 (LTB4)-induced itch. LTB4 is a potent chemoattractant for leukocytes released from the ...
  97. [97]
  98. [98]
    TRPV1 Suppresses Microglial Inflammatory Activation to Ameliorate ...
    Sep 11, 2025 · TRPV1-KO microglia, but not neurons, promoted inflammation and neuronal damage via CaMKII/NRF2/SIRT3 downregulation. Conclusions. These findings ...
  99. [99]
    Neuropeptides activate TRPV1 in rheumatoid arthritis fibroblast-like ...
    TRPV1 activation may act as an inducer of inflammatory signals or cooperate with neuropeptides to increase cytokine production during RA, as demonstrated in ...
  100. [100]
    TDAG8, TRPV1, and ASIC3 involved in establishing hyperalgesic ...
    Aug 21, 2017 · TRPV1 regulates synovial inflammation from the beginning of disease, but TRPV1 deficiency affected RA-associated pain beginning at the later ...
  101. [101]
    The role of TRPV1 in RA pathogenesis: worthy of attention - PMC
    The TRPV1 antagonist capsazepine significantly attenuates TRPV1 expression and SP release (126). Consistent with this, CAP treatment activates the TRPV1 ...
  102. [102]
    [PDF] Function and mechanism of action of the TRPV1 channel in the ...
    May 11, 2024 · Increased TRPV1 expression often indicates poor prognosis. TRPV1 expression correlates with the tumor grade of prostate cancer and increases ...
  103. [103]
    Intracellular aggregated TRPV1 is associated with lower survival in ...
    Oct 15, 2018 · Of clinical relevance, the expression of the nonclassical pattern is associated with worse prognosis and lower survival, suggesting that TRPV1 ...Missing: bladder | Show results with:bladder
  104. [104]
    Immunohistochemical expression of transient receptor potential ...
    Mar 19, 2025 · High expression of TRPV1 was seen in the advanced stage of bladder cancer, while decreased expression was seen in low-grade bladder cancer. Very ...Missing: poor | Show results with:poor
  105. [105]
    Capsaicin induces mitochondrial dysfunction and apoptosis in ...
    Aug 6, 2020 · Thus, our study demonstrated that capsaicin induced mitochondrial calcium overload and apoptosis in ATC cells through a TRPV1-mediated pathway.
  106. [106]
    Calcium influx, oxidative stress, and apoptosis induced by TRPV1 in ...
    Feb 14, 2023 · We demonstrated that the triggering of transient receptor potential vanilloid 1 inhibits cell growth and promotes apoptosis of chronic myeloid leukemia cells.
  107. [107]
    Downregulated TRPV1 Expression Contributes to Melanoma ...
    Forwardly, the activation of TRPV1 by either capsaicin treatment or TRPV1 overexpression remarkably attenuated melanoma proliferation by activating p53 and ...
  108. [108]
    TRPV1 Induced Apoptosis of Colorectal Cancer Cells by Activating ...
    Apr 30, 2019 · Then, we found that the TRVP1 agonist capsaicin treatment inhibited CRC growth and induced apoptosis by activating P53. Subsequent mechanistic ...
  109. [109]
    New options for targeting TRPV1 receptors for cancer treatment
    Feb 11, 2025 · TRPV1 expression promotes drug resistance in NSCLC. High TRPV1 expression is associated with poor prognosis in LUAD.
  110. [110]
    New options for targeting TRPV1 receptors for cancer treatment
    Feb 10, 2025 · Weber LV et al. found that the expression of TRPV1 in breast cancer tissues and cells was significantly higher than that of healthy breast ...
  111. [111]
    Inflammation, Cancer and Immunity—Implication of TRPV1 Channel
    (73) who demonstrated that TRPV1 is involved in the uptake of anandamide, the endogenous cannabinoid and agonist of TRPV1 implicated in tumor-angiogenesis, by ...
  112. [112]
    Impact of TRPV1 on Pathogenesis and Therapy of ... - MDPI
    It has been shown that TRPV1 is involved in microglia-induced inflammation [8,10]. Studies have indicated that the stimulation of TRPV1 may exacerbate ...
  113. [113]
    The TRPV1-PKM2-SREBP1 axis maintains microglial lipid ... - Nature
    Jan 14, 2025 · Our study suggests that TRPV1-PKM2-SREBP1 axis regulation of microglia lipid metabolism could be a therapeutic approach to alleviate the consequences of AD.
  114. [114]
    TRP channels: Role in neurodegenerative diseases and therapeutic ...
    Aβ triggers the activation of neuroinflammatory processes, directed by activated-glial cells that produce inflammatory cytokines, which activate TRPV1. All ...Review Article · 4.1. Alzheimer's Disease · 4.2. Parkinson's Disease
  115. [115]
    Capsaicin, The Vanilloid Receptor TRPV1 Agonist in Neuroprotection
    Jul 26, 2023 · Several experimental and clinical studies provided evidence that capsaicin protected against ischaemic or excitotoxic cerebral neuronal injury ...
  116. [116]
    TRPV1 on astrocytes rescues nigral dopamine neurons in ... - PubMed
    Here we report that transient receptor potential vanilloid 1 (TRPV1) on astrocytes mediates endogenous production of ciliary neurotrophic factor (CNTF).
  117. [117]
    TRPV1-Mediated Microglial Autophagy Attenuates Alzheimer's ...
    TRPV1 agonist capsaicin decreased amyloid and phosphorylated tau pathology and reversed memory deficits by promoting microglia activation, metabolism, and ...
  118. [118]
    Capsaicin and TRPV1: A Novel Therapeutic Approach to Mitigate ...
    We propose that capsaicin may serve as the medication with the potential to slow the progress of vascular aging and could constitute a new strategy to treat ...
  119. [119]
    Interaction between TRPV1-expressing neurons in the hypothalamus
    Jan 1, 2019 · Our study demonstrated the functional expression of TRPV1 in the dorsomedial hypothalamic nucleus and paraventricular nucleus in adult mice.
  120. [120]
    Sleep deficiency exacerbates periodontal inflammation via ... - PNAS
    Jun 9, 2025 · Our findings unveil a critical neuron-vessel-immune axis that exacerbates periodontitis during sleep deficiency and suggest potential therapeutic strategies.
  121. [121]
    The physiological role of TRP channels in sleep and circadian rhythm
    Apr 27, 2024 · TRP channels provide evidence of an ability to create, regulate and modify our sleep and circadian rhythm in a wide array of physiological and ...
  122. [122]
    β-Arrestin-2 Desensitizes the Transient Receptor Potential Vanilloid ...
    β-Arrestin-2 functions as a scaffold protein to mediate TRPV1 desensitization in multiple cell models.
  123. [123]
    calmodulin binding to N- and C-terminal regulatory regions of the ...
    Oct 29, 2012 · Two CaM-binding sites are identified in TRPV1: the N-terminal ankyrin repeat domain (ARD) and a short distal C-terminal (CT) segment. Here, we ...Missing: Src | Show results with:Src
  124. [124]
    Role of Src Kinase in Regulating Protein Kinase C Mediated ...
    Aug 4, 2022 · This study aimed to explore the relationship between Src kinase and PKC phosphorylation in the sensitization of TRPV1. We demonstrate that ...Missing: Ca2+ | Show results with:Ca2+
  125. [125]
    Direct Evidence for Functional TRPV1/TRPA1 Heteromers - PubMed
    There is evidence for a TRPV1-TRPA1 interaction that is predominantly calcium-dependent, and it has been suggested that the two proteins might form a ...
  126. [126]
    Vti1b promotes TRPV1 sensitization during inflammatory pain - LWW
    We show that the vesicle transport through interaction with t-SNAREs homolog 1B (Vti1b) protein promotes TRPV1 sensitization upon inflammation in cell culture.
  127. [127]
    The ion channel TRPV1 regulates the activation and ...
    We observed that Trpv1−/− CD4+ T cells produced lower pan-cytokine levels (IFN-γ, IL-17A, IL-2, IL-10, IL-4 and TNF) after anti-CD3+28 stimulation compared to ...Missing: migration | Show results with:migration
  128. [128]
    Phosphatidylinositol 3-Kinase Activates ERK in Primary Sensory ...
    Sep 22, 2004 · Phosphatidylinositol 3-Kinase Activates ERK in Primary Sensory Neurons and Mediates Inflammatory Heat Hyperalgesia through TRPV1 Sensitization.
  129. [129]
    Vanilloid receptor TRPV1-mediated phosphorylation of ERK in ...
    Aug 5, 2008 · We conclude that TRPV1, expressed by neurons in the articular afferent pathway, contributes to the pathogenesis of arthritis via an ERK-mediated pathway.
  130. [130]
    [PDF] A Noxious Signal Transducer That Affects Mitochondrial Function
    Nov 24, 2020 · The evidence described above suggests that TRPV1 activation causes mitochondrial Ca2+ unbalance, disrupting the mitochondrial membrane potential ...
  131. [131]
    Cannabinoid-based drugs targeting CB1 and TRPV1, the ...
    Sep 6, 2015 · In this review, we focus on CB 1 and transient receptor potential vanilloid 1 (TRPV1)-mediated effects on RA since most anti-inflammatory mechanisms induced by ...
  132. [132]
    CB1 receptor‐dependent desensitisation of TRPV1 channels ...
    Sep 6, 2020 · We have reported that the dipyrone metabolite 4‐aminoantipyrine (4‐AA) reduces PGE 2 ‐induced pain‐related behaviour through cannabinoid CB 1 receptors.
  133. [133]
    TRPV1 modulated NLRP3 inflammasome activation via calcium in ...
    TRPV1 is a non-selective calcium channel that is involved in the pathology of neuroinflammation, but its role in SAH has not been revealed.
  134. [134]
    NEO6860, modality-selective TRPV1 antagonist - PAIN Reports
    NEO6860 exposure was ∼1.6 times higher compared with previous phase I. In this study, NEO6860 safety profile was less favorable than naproxen or placebo.
  135. [135]
    NEO6860, modality-selective TRPV1 antagonist - PubMed Central
    NEO6860 is a TRPV1 antagonist when activated by capsaicin but not by heat or pH, developed to relieve pain without the adverse events reported with non–modality ...
  136. [136]
    A new class of modality selective antagonistic TRPV1 antibodies for ...
    Feb 27, 2020 · We set out to develop monoclonal antibodies that inhibit capsaicin activation of TRPV1 while avoiding inhibiting heat activation and with ...Missing: domain | Show results with:domain
  137. [137]
    Unraveling TRPV1's Role in Cancer: Expression, Modulation ... - NIH
    Oct 7, 2024 · Higher TRPV1 mRNA expression is an independent risk factor for poor prognosis. ... Bladder cancer and urothelial impairment: The role of TRPV1 ...
  138. [138]
    TRPV1-target drugs for the treatment of orofacial pain - PMC
    Apr 24, 2025 · This review focuses on the role of Transient Receptor Potential Vanilloid 1 (TRPV1) channels in modulating orofacial pain and new ligands targeting this ...
  139. [139]
    Capsazepine Inhibits Astrocyte Activation and Attenuates ...
    May 19, 2025 · Capsazepine (CZP) mitigates neuroinflammation by suppressing astrocyte activation via a TRPV1-independent pathway.
  140. [140]
    The Mysteries of Capsaicin-Sensitive Afferents - Frontiers
    TRPV1 is activated by heat (Caterina et al., 1997) with a threshold of just above 40 °C (Zhang et al., 2018): this is not far from the human heat pain ...
  141. [141]
    Forty years in capsaicin research for sensory pharmacology and ...
    Hence, capsaicin research led to the discovery of the first temperature-gated ion channel gated by noxious heat, protons, vanilloids and endogenous ligands as ...Review · Abstract · Capsaicin Desensitization...
  142. [142]
    The discovery of capsazepine, the first competitive ... - PubMed
    The discovery of capsazepine, the first competitive antagonist of the sensory neuron excitants capsaicin and resiniferatoxin. J Med Chem. 1994 Jun 24;37(13): ...Missing: TRPV1 date
  143. [143]
    Press release: The Nobel Prize in Physiology or Medicine 2021
    Oct 4, 2021 · David Julius' discovery of TRPV1 was the breakthrough that allowed us to understand how differences in temperature can induce electrical signals ...Missing: milestones | Show results with:milestones