Fact-checked by Grok 2 weeks ago

Trk receptor

The Trk receptors, also known as tropomyosin receptor kinases (TRKs), constitute a family of three high-affinity receptor tyrosine kinases—TrkA (encoded by NTRK1), TrkB (NTRK2), and TrkC (NTRK3)—that bind specific to mediate critical signaling for neuronal survival, differentiation, growth, and throughout the . These receptors play indispensable roles in embryonic development, ensuring proper , target innervation, and prevention of in diverse neuronal populations, while in adulthood, they support functions such as learning, formation, , and neural repair. Structurally, each Trk receptor is a single-pass featuring an extracellular ligand-binding domain composed of a leucine-rich , two cysteine-rich domains, and two immunoglobulin-like (Ig-like) domains, which confer specificity for ; a hydrophobic ; and an intracellular juxtamembrane region followed by a domain that shares approximately 72–78% sequence identity across family members. Ligand specificity is as follows: TrkA binds primarily to (NGF), with lower affinity for (NT-3); TrkB binds brain-derived neurotrophic factor (BDNF) and NT-4, also interacting weakly with NT-3; and TrkC binds exclusively to NT-3 at high affinity. generates isoforms, including kinase-deficient variants that modulate signaling, particularly for TrkB and TrkC, which lack the domain and act as dominant negatives. Upon neurotrophin binding at the cell surface or in endosomes, Trk receptors undergo dimerization and autophosphorylation on key intracellular tyrosine residues (e.g., Y490, Y785 in TrkA), recruiting adaptor proteins like Shc, FRS2, and IRS to activate three major downstream cascades: the /extracellular signal-regulated kinase (ERK)/ (MAPK) pathway, which promotes neuronal differentiation and proliferation; the (PI3K)/Akt pathway, essential for cell survival and anti-apoptotic effects; and the C-γ (PLC-γ) pathway, which mobilizes intracellular calcium to influence synaptic transmission and plasticity. Trk signaling often synergizes with the low-affinity p75 neurotrophin receptor (p75NTR), a member of the tumor necrosis factor receptor superfamily, to fine-tune ligand affinity and elicit diverse outcomes, such as enhanced survival for TrkA–NGF complexes or apoptosis promotion in the absence of Trk activation. In neuronal development, TrkA supports the survival and target innervation of sensory and sympathetic neurons, with genetic knockout studies showing 70–90% loss of neurons; TrkB regulates hippocampal and cortical neuron maturation, dendritic arborization, and (LTP) via BDNF; and TrkC ensures proprioceptive neuron viability and synaptic strengthening through NT-3. Beyond the central and peripheral nervous systems, Trk expression extends to non-neuronal cells like immune cells and tumors, where aberrant activation via gene fusions drives oncogenesis, though their primary physiological domain remains neurotrophin-mediated neural .

Discovery and Nomenclature

Historical discovery

The discovery of the Trk receptor family originated from studies on oncogenes in human cancers. In 1982, Mariano Barbacid and colleagues identified a transforming gene, later termed trk, during gene transfer experiments using DNA from a human colon carcinoma biopsy; this oncogene was found to promote morphological transformation in NIH 3T3 cells. The transforming activity was traced to a chimeric gene fusing sequences from the non-muscle tropomyosin gene to a novel protein tyrosine kinase domain. In 1986, detailed molecular characterization by Barbacid's group revealed that the v-trk oncogene (as it was initially called, though derived from human tumor DNA rather than a virus) resulted from a somatic rearrangement in the colon carcinoma, replacing the extracellular domain of a with sequences, thereby activating its kinase activity constitutively. This work led to the identification of the corresponding cellular proto-oncogene, c-trk (now known as NTRK1), which encodes a 140-kDa transmembrane expressed primarily in neural tissues. Subsequent efforts expanded the family. In 1989, Rüdiger Klein and colleagues cloned trkB (NTRK2) from mouse brain cDNA using low-stringency hybridization to the trk domain, identifying it as a related receptor abundantly expressed during neural development. TrkC (NTRK3) was cloned in by Frank Lamballe, Klein, and Barbacid through PCR amplification of degenerate based on conserved motifs from trkA and trkB, revealing another neural-specific receptor. The functional linkage to neurotrophin signaling emerged in 1991, when David Kaplan, Dionisio Martin-Zanca, and Luis Parada demonstrated that nerve growth factor (NGF) specifically induced tyrosine phosphorylation and autophosphorylation of the TrkA , establishing TrkA as its high-affinity receptor. Concurrently, Klein et al. showed NGF binding to TrkA-transfected cells with high affinity, confirming its role in neuronal survival and differentiation. Key milestones followed: in 1991, Sophie Soppet, Mariano Barbacid, and colleagues identified (BDNF) and (NT-3) as ligands for TrkB, with BDNF showing high specificity. By 1991, Lamballe et al. solidified NT-3 as the primary ligand for TrkC, completing the -Trk pairing framework.

Origin of the name

The abbreviation "Trk" for the receptor family stands for "tropomyosin receptor kinase," a name derived from the initial identification of the proto-oncogene through its oncogenic fusion form, v-trk, which combined sequences from the gene with a domain in a colon . This fusion, reported in 1986, revealed a novel with transforming potential, leading to the characterization of the normal cellular proto-oncogene (trk) in 1989 as a expressed in neural tissues. The official human , established by the in the early 1990s following the of the respective genes around 1991, designates them as NTRK1 (for TrkA), NTRK2 (for TrkB), and NTRK3 (for TrkC), where "NTRK" reflects their function as neurotrophic tyrosine receptor kinases. This standardization clarified the distinction from the viral v-trk and emphasized their roles in neurotrophin signaling, with the protein products conventionally named TrkA, TrkB, and TrkC using uppercase "Trk" to denote the cellular receptors, while lowercase "trk" is retained for the oncogene in .

Molecular Structure and Types

General structure

The Trk receptors, also known as neurotrophic tyrosine receptor kinases (NTRKs), are single-pass transmembrane proteins belonging to the (RTK) superfamily. Their overall topology consists of an extracellular (ECD) responsible for ligand binding, a single transmembrane helix that anchors the receptor in the plasma membrane, an intracellular juxtamembrane region, and a cytoplasmic that transduces signals upon activation. The ECD is composed of several structural motifs, including two cysteine-rich clusters flanking three leucine-rich repeats (LRRs) and followed by two immunoglobulin-like (Ig-like) domains, often referred to collectively as five domains (–D5) that facilitate receptor dimerization. The intracellular portion features a juxtamembrane segment and a catalytic domain containing an activation loop with conserved residues, such as Y674 in TrkA, which are critical for kinase activity.55117-1/pdf) Mature Trk receptors typically exhibit a molecular weight of approximately 140–145 , reflecting extensive post-translational of the ECD. The kinase domains across TrkA, TrkB, and TrkC subtypes display high of approximately 72–78% across the family members, underscoring their shared catalytic mechanisms, whereas ECD lengths vary due to differences in isoform expression. Post-translational modifications are prominent, with multiple N- sites in the ECD—such as the four conserved sites in TrkA—essential for proper folding, trafficking, and surface localization. The intracellular domain harbors several tyrosine phosphorylation sites, including those in the juxtamembrane and regions, which serve as platforms for downstream effectors. Subtype-specific variations, such as differences in ECD composition, are detailed in subsequent sections on individual Trk receptors.

TrkA (NTRK1)

The NTRK1 gene, located on human 1q23.1, encodes the TrkA receptor protein. The full-length isoform of TrkA comprises 796 amino acids and features a single , an extracellular ligand-binding region with leucine-rich motifs and immunoglobulin-like domains, and an intracellular domain essential for . This structure enables TrkA to function as a in neuronal development and maintenance. Alternative splicing of NTRK1 transcripts produces two primary full-length isoforms, TrkA-I (790 amino acids) and TrkA-II (796 amino acids), which differ by the inclusion of a 6-amino-acid sequence in the extracellular domain of TrkA-II. Both isoforms retain the complete intracellular kinase domain, enabling autophosphorylation and downstream signaling upon ligand binding. TrkA-II predominates in neuronal tissues, while TrkA-I is more common in non-neuronal cells. Additionally, truncated, kinase-deficient isoforms lacking the kinase domain exist and may act as dominant negatives by sequestering ligands or forming non-functional heterodimers with full-length receptors. These isoforms exhibit tissue-specific expression, with TrkA-II predominating in the brain. TrkA expression is prominent in peripheral sensory and sympathetic neurons, where it supports survival and differentiation during development. It is also highly expressed in central neurons, contributing to their maturation and target innervation in regions like the . A defining feature of TrkA is its high-affinity binding site for (NGF), with a in the picomolar range, which distinguishes it from other Trk receptors. In the context of , TrkA activation leads to sensitization of nociceptors via of specific intracellular tyrosines, such as Y785, which serves as a docking site for C-γ (PLCγ) and initiates downstream pathways enhancing neuronal excitability. Rare germline mutations in NTRK1, often affecting the domain, are linked to with anhidrosis (CIPA), an autosomal recessive disorder characterized by loss of pain perception and thermoregulatory sweating due to impaired TrkA signaling. These variants, including missense changes like R190W, disrupt receptor maturation or autophosphorylation, underscoring TrkA's critical role in function.

TrkB (NTRK2)

The TrkB receptor, encoded by the NTRK2 gene located on chromosome 9q21.33, produces a full-length consisting of 838 that functions as a transmembrane receptor. This gene spans approximately 427 kb and generates multiple isoforms through , including the catalytically active full-length TrkB (TrkB.FL) and truncated variants such as TrkB.T1 and TrkB.Shc, which lack the intracellular domain and often exert dominant-negative effects by heterodimerizing with full-length receptors to inhibit signaling. TrkB.T1, the most prevalent truncated isoform, features a short intracellular of 23 and is widely expressed in the , where it modulates BDNF availability and receptor trafficking. Similarly, TrkB.Shc binds Shc adaptor proteins to influence downstream pathways independently of activity. TrkB exhibits high expression levels throughout the (CNS), particularly in the and , where it supports neuronal survival and plasticity, as well as in motor neurons and the , contributing to axonal maintenance and visual circuit refinement. In the , TrkB signaling is essential for activity-dependent synaptic strengthening, while in motor neurons, it promotes postnatal survival against injury-induced degeneration. Retinal ganglion cells express TrkB to regulate dendritic arborization and synaptic connectivity during development. Unlike other Trk family members, TrkB preferentially binds (BDNF) and neurotrophin-4/5 (NT-4/5) with high affinity, initiating dimerization via its extracellular domain (ECD), which comprises leucine-rich motifs, cysteine-rich clusters, and two immunoglobulin-like C2-type domains that facilitate ligand specificity. These structural elements enable TrkB to mediate synaptic scaling, a homeostatic process adjusting synaptic strength; truncated isoforms like TrkB.T1 fine-tune this by limiting full-length receptor activation during prolonged BDNF exposure, preventing in cortical networks. Dysregulation of TrkB expression or signaling is implicated in neuropsychiatric and metabolic disorders, with reduced TrkB levels associated with heightened vulnerability to through impaired hippocampal and responsiveness. In models, TrkB disrupts serotonin-mediated behaviors and , underscoring its role in treatment resistance. Similarly, diminished TrkB activity contributes to by altering and regulation, as evidenced in genetic models where TrkB mutations lead to hyperphagia and fat accumulation without primary feeding circuit defects. These associations highlight TrkB's therapeutic potential, though isoform-specific functions complicate interventions.

TrkC (NTRK3)

The TrkC receptor, also known as neurotrophic receptor type 3 (NTRK3), is encoded by the NTRK3 located on human chromosome 15q25.3. The full-length isoform of the TrkC protein comprises 839 amino acids, featuring an extracellular ligand-binding domain, a single transmembrane region, and an intracellular domain characteristic of the Trk family. This structure enables TrkC to function primarily as the high-affinity receptor for (NT-3), facilitating signaling critical for neuronal differentiation and survival. NTRK3 undergoes to generate multiple isoforms, primarily through mechanisms that produce both -active and truncated variants. -active isoforms include those without inserts and those with a 14-amino-acid insertion (K14) or a 25-amino-acid insertion () within the domain, while truncated forms lack the kinase domain entirely, resulting in non-signaling receptors that may modulate availability.90360-3) These variable kinase inserts notably alter substrate specificity; for instance, insert-containing isoforms exhibit reduced autophosphorylation at key sites and impaired binding to effectors like phospholipase Cγ and Shc, thereby modifying downstream signaling efficiency compared to insert-free variants.90396-0/fulltext) TrkC expression is predominantly observed in proprioceptive sensory neurons of the dorsal root ganglia, cerebellar granule cells, and select hematopoietic cells such as mast cells and precursors, particularly during embryonic and early postnatal . In adults, TrkC levels are markedly lower across these tissues, reflecting a developmental downregulation consistent with its role in rather than maintenance. This expression profile underscores TrkC's specificity for proprioceptive and cerebellar functions, distinguishing it from other Trk receptors. Genetic alterations in NTRK3, including point mutations and gene fusions such as ETV6-NTRK3, have been implicated in , where high TrkC expression often correlates with favorable prognosis in sonic hedgehog-driven subtypes.

Ligands and Receptor Activation

Neurotrophins as ligands

The constitute a family of structurally related growth factors that act as primary ligands for Trk receptors, comprising (NGF), (BDNF), (NT-3), and neurotrophin-4 (NT-4, also known as NT-4/5). These proteins function as non-covalently associated homodimers, with each subunit approximately 13-14 kDa in size, and share a conserved formed by three bonds stabilizing antiparallel β-strands. This fold is critical for their stability and in supporting neuronal development and function. The discovery of NGF in the 1950s by Stanley Cohen and marked the inception of research; they identified it as a growth-promoting factor isolated from 180 tumors and extracts, which induced dramatic neurite outgrowth in sensory and . BDNF was subsequently purified in 1982 by Yves-Alain Barde, Detlev Edgar, and Hans Thoenen from porcine brain, revealing a distinct neurotrophic factor that supported survival of embryonic chick neurons. NT-3 emerged in 1990 through cloning efforts by Peter C. Maisonpierre and colleagues, who demonstrated its sequence homology to NGF and BDNF and its unique expression pattern in the developing . NT-4 was identified in 1991 by Fredrik Hallböök, Carlos F. Ibáñez, and Håkan Persson as a novel family member abundantly expressed in ovary, further expanding the repertoire. Neurotrophins are initially synthesized as proneurotrophins, larger precursor proteins that undergo proteolytic cleavage—typically by or proprotein convertases in the trans-Golgi network or secretory vesicles—to yield the , active forms. The neurotrophins are then packaged into vesicles, stored, and released from neurons and target tissues in a regulated, activity-dependent manner, allowing precise spatiotemporal control over their signaling. In general, exhibit low-affinity binding to the p75 neurotrophin receptor (p75NTR) and high-affinity binding to Trk receptors (TrkA, TrkB, or TrkC), a dual-receptor essential for neuronal survival, maintenance, and during and adulthood. At the structural level, the interaction with the extracellular domain of Trk receptors involves specific β-hairpin loops (particularly loops 1, 2, and 4) in the that contact conserved patches on the receptor's immunoglobulin-like domains, facilitating recognition and downstream activation.

Ligand-receptor specificity and cross-talk

The Trk receptors display a high degree of ligand specificity, with (NGF) binding exclusively and with high affinity to TrkA (Kd ≈ 10^{-11} M), (BDNF) and neurotrophin-4 (NT-4) binding preferentially to TrkB, and neurotrophin-3 (NT-3) binding primarily to TrkC. This selectivity arises from interactions primarily involving the fifth immunoglobulin-like domain (D5) of the Trk extracellular region and specific residues on the neurotrophins, as revealed by crystallographic studies of the NGF-TrkA complex. NT-3 also exhibits low-affinity binding to TrkA and TrkB, typically at concentrations 100-fold higher than those required for TrkC activation. Cross-talk between and non-cognate Trk receptors occurs under certain conditions; for example, NT-3 can activate TrkA and TrkB signaling pathways at elevated levels, while BDNF weakly activates TrkC in specific neuronal contexts. assays on neuronal cultures and recombinant receptors have quantified this promiscuity, showing NT-3 affinities for TrkA and TrkB in the range of 10^{-9} to 10^{-10} M, compared to picomolar values for the preferred pairs. Structural models, such as those derived from of TrkB-NT-4 complexes, further illustrate how subtle variations in loops and receptor patches enable such cross-interactions while maintaining overall specificity. These interactions have functional implications, providing redundancy in neural development by allowing alternative ligands to support neuronal survival and differentiation when primary pathways are disrupted. In pathological states, cross-talk can be dysregulated; for instance, NT-3 engagement of TrkA on nociceptive neurons contributes to enhanced signaling in neuropathic conditions. Experimental evidence from models confirms that this overlap ensures developmental compensation, such as NT-3 partially substituting for NGF in TrkA-dependent processes. Binding affinities are modulated by environmental factors, including and ; extracellular acidification (e.g., pH 6.5) promotes TrkA mobilization to the plasma membrane, enhancing NGF responsiveness, while higher ionic conditions like 0.15 M KCl influence complex stability in simulations.

Regulation of Trk Signaling

Role of p75NTR

The p75 neurotrophin receptor (p75NTR), also known as NGFR, belongs to the (TNF) receptor superfamily and functions as a low-affinity pan- receptor. It is a type I transmembrane characterized by an extracellular (ECD) comprising four cysteine-rich repeats that mediate binding, a single-span transmembrane , and an intracellular (ICD) featuring a death responsible for recruiting downstream signaling adaptors. This structural architecture enables p75NTR to form both homodimers and heterocomplexes, facilitating diverse signaling outcomes in neuronal contexts. p75NTR interacts directly with Trk receptors through both extracellular and intracellular domains, forming heterocomplexes that modulate Trk signaling. These interactions occur via the transmembrane domains and are stabilized upon binding, as demonstrated by co-immunoprecipitation and studies in transfected cells and neurons. When co-expressed with TrkA, p75NTR dramatically increases the binding affinity for mature (NGF) by approximately 100-fold compared to TrkA alone, thereby sensitizing cells to low concentrations of this . This enhancement arises from allosteric changes in the receptor complex that accelerate association rates while slowing dissociation. In its role as a co-receptor, p75NTR refines Trk ligand specificity by suppressing non-preferred interactions, such as reducing TrkA responsiveness to while amplifying selectivity. This modulation promotes pro-survival signaling through TrkA, including of downstream pathways that support neuronal differentiation and maintenance in developing sympathetic and sensory neurons. Conversely, p75NTR can independently trigger in the absence of Trk , particularly through to proneurotrophins like proNGF and proBDNF, often in complex with co-receptor sortilin (SORT1), which engage the death domain to recruit factors such as TRAF6 and activate JNK-mediated cascades. p75NTR is prominently co-expressed with Trk receptors in developing neurons, including those in the peripheral nervous system and , where it peaks during periods of axonal growth and target innervation. Beyond affinity modulation, p75NTR regulates Trk trafficking by influencing and ; for instance, it promotes rapid of TrkA-bound NGF complexes, ensuring efficient signal propagation from distal axons to cell bodies. This trafficking control is evident in hippocampal and cortical neurons, where p75NTR limits ubiquitination of surface Trk receptors, thereby prolonging ligand-induced activation. Key insights into these functions emerged from 1990s studies using p75NTR knockout mice, which revealed altered Trk responses and neuronal phenotypes. In p75NTR-null mice, sympathetic neurons exhibit reduced naturally occurring but display aberrant sensory innervation patterns, with excessive branching due to diminished TrkA specificity for NGF. These animals also show impaired survival in the , highlighting p75NTR's role in fine-tuning Trk-dependent trophic support during development. Such findings underscored the dual modulatory capacity of p75NTR, balancing survival and apoptotic cues in Trk-expressing populations.

Other regulatory mechanisms

Trk receptors undergo ligand-induced primarily through clathrin-mediated pathways, where binding of such as NGF to TrkA recruits adaptor proteins like AP-2 and , facilitating receptor into early endosomes. This process is essential for attenuating surface signaling and initiating intracellular trafficking. In neuronal axons, internalized Trk receptors form signaling endosomes that are transported along by motors, enabling long-distance transmission of survival and growth signals to the cell body; disruption of this retrograde transport impairs -dependent neuronal maintenance. Alternative splicing of Trk genes produces truncated isoforms, such as TrkB.T1, which lack the intracellular domain but retain the extracellular ligand-binding region. These isoforms act as dominant-negative regulators by sequestering like BDNF, thereby preventing their interaction with full-length Trk receptors and inhibiting downstream signaling pathways such as MAPK and PI3K. For instance, overexpression of TrkB.T1 in neuronal cultures reduces BDNF-induced of full-length TrkB and suppresses neurite outgrowth, highlighting their role in fine-tuning trophic responses during and in adulthood. Post-translational modifications, particularly ubiquitination, control Trk receptor stability and localization. Upon binding, TrkA is multi-monoubiquitinated by E3 ligases such as Nedd4-2, which recognizes a PPXY and targets the receptor for lysosomal , thereby limiting prolonged signaling and promoting receptor turnover. Similarly, TRAF6 mediates K63-linked polyubiquitination of TrkA, enhancing without immediate and modulating pain sensitivity in sensory neurons. Palmitoylation contributes to Trk receptor membrane targeting, though its precise role in Trk-specific localization remains less characterized compared to ubiquitination. Negative feedback mechanisms dephosphorylate activated Trk receptors to terminate signaling. Protein phosphatases, such as PTPσ (PTPRS), bind directly to TrkA and TrkC, dephosphorylating key residues and suppressing neurotrophin-induced activation; for example, PTPσ overexpression in PC12 cells attenuates NGF-dependent neurite outgrowth. MicroRNAs also regulate Trk expression at the transcriptional level; miR-128 targets the 3' of the truncated TrkC isoform (T-TrkC), reducing its levels and thereby enhancing full-length TrkC signaling, as observed in cells where miR-128 overexpression promotes cell survival via upregulation. Extracellular matrix components influence Trk receptor organization and activity. Laminin-1, a basement membrane glycoprotein, binds to monosialoganglioside GM1 in lipid rafts, inducing rapid clustering of TrkA receptors on the neuronal plasma membrane and potentiating NGF signaling for neurite outgrowth. This ECM-mediated clustering enhances TrkA colocalization with β1 integrins, activating downstream pathways like Akt and MAPK independently of direct ligand binding.

Downstream Signaling Pathways

Receptor activation and dimerization

Upon binding of dimeric ligands to the extracellular domain of Trk receptors, the receptors undergo dimerization, primarily forming homodimers that bring the intracellular kinase domains into proximity for . This is facilitated by the ligand's bivalent nature, which promotes a 2:2 of two ligand molecules and two receptor extracellular domains, as revealed by crystal structures of the TrkA extracellular domain in complex with . Co-expression with p75NTR can enhance dimer stability through high-avidity heterocomplexes, although homodimers predominate in ligand-induced . Dimerization triggers conformational changes that propagate across the , involving of the transmembrane helices and reorientation of the juxtamembrane regions, which enable asymmetric of the kinase domains. Structural analyses, including NMR spectroscopy of the TrkB , confirm that these helical rotations stabilize an active conformation, with the intracellular domains adopting an asymmetric arrangement where one kinase acts as the primary activator. The activated subsequently autophosphorylate key residues in the activation loop and juxtamembrane regions, such as Y490 and Y785 in TrkA, which are essential for kinase activity and signal propagation. occurs rapidly, with detectable on Trk receptors within seconds of exposure in cellular assays. Sustained activation is often maintained through endocytic trafficking and in neurons.

MAPK/ERK pathway

Upon ligand-induced dimerization and autophosphorylation of Trk receptors, specific residues in the intracellular , such as Y490 in TrkA, serve as sites for adaptor proteins Shc and FRS2. These adaptors are phosphorylated by the activated Trk and subsequently recruit the Grb2- via their SH2 domains. , a , then promotes the exchange of GDP for GTP on , yielding active Ras-GTP. Active Ras-GTP recruits and activates , which in turn phosphorylates and activates MEK1/2. MEK1/2 subsequently dual-phosphorylates ERK1/2 on and residues within the TEY motif, fully activating the terminal kinases of this cascade. Activated ERK1/2 translocates from the to the , where it transcription factors such as CREB and Elk-1. This enhances their activity, leading to the of immediate early genes like c-Fos that drive cellular responses including and . The duration of ERK —transient for or sustained for —is influenced by the specific adaptors recruited, with FRS2 contributing to prolonged signaling via additional pathways like Rap1-B-Raf. The MAPK/ERK pathway is most robustly activated by TrkA and TrkB receptors, playing an essential role in neurotrophin-mediated neuronal , whereas TrkC activation is weaker and more context-dependent. In TrkA-expressing sympathetic neurons and PC12 cells, this pathway is critical for NGF-induced . Scaffold proteins such as KSR1 enhance the efficiency of the cascade by assembling Raf, MEK, and ERK into a multiprotein complex at the plasma membrane, facilitating sequential and spatial organization. is provided by dual-specificity phosphatases (DUSPs), which dephosphorylate ERK1/2 to terminate signaling and prevent overstimulation. Experimentally, inhibition of ERK activation with pharmacological agents like PD98059 blocks Trk-mediated neuronal survival and neurite outgrowth in PC12 cells, underscoring the pathway's necessity for these responses.

PI3K/Akt pathway

Upon ligand-induced activation, Trk receptors autophosphorylate at specific intracellular tyrosine residues, enabling recruitment of adaptor proteins including IRS-1/IRS-2 and Gab1. These adaptors recruit and activate phosphoinositide 3-kinase (PI3K), which phosphorylates phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3). PIP3 then recruits phosphoinositide-dependent kinase 1 (PDK1) and Akt (also known as protein kinase B) to the plasma membrane, where PDK1 phosphorylates Akt at T308, and subsequent phosphorylation at S473 by mTORC2 fully activates Akt. Activated Akt promotes cell survival and growth through multiple downstream effectors. It phosphorylates and inactivates pro-apoptotic proteins such as BAD and Bax, preventing mitochondrial release and activation; BAD is sequestered by 14-3-3 proteins in its phosphorylated state. Additionally, Akt activates complex 1 (), which enhances protein synthesis via of targets like S6 kinase and 4E-BP1, supporting neuronal growth and maintenance. The PI3K/Akt pathway is particularly prominent in TrkB and TrkC signaling, where it drives neuronal survival in response to BDNF and NT-3, respectively; for instance, BDNF-TrkB activation via IRS-2 adaptors sustains hippocampal and viability. Isoform-specific adaptors, such as Gab1 for TrkA and IRS proteins preferentially for TrkB, contribute to nuanced pathway activation across Trk subtypes. This pathway exhibits synergistic crosstalk with the MAPK/ERK cascade, where concurrent activation amplifies neuronal survival signals beyond individual pathway effects. Conversely, the pathway is negatively regulated by the phosphatase , which dephosphorylates PIP3 to PIP2, limiting Akt activation; in Trk contexts, p75NTR can induce PTEN expression to suppress PI3K/Akt-mediated survival. Pharmacological studies demonstrate the pathway's essentiality: PI3K inhibitors like LY294002 induce Trk-dependent in neurotrophin-supported neurons, such as NGF-dependent sympathetic neurons, by blocking Akt activation and allowing BAD-mediated .

PLCγ/PKC pathway

Upon ligand-induced dimerization and autophosphorylation of Trk receptors, residue 785 (Y785) in the C-terminal tail of TrkA (or the homologous Y816 in TrkB and Y795 in TrkC) becomes phosphorylated, serving as a primary docking site for the of C-γ1 (PLCγ1). This recruitment facilitates PLCγ1 autophosphorylation at Y783 by the Trk domain, activating its enzymatic activity to hydrolyze membrane-bound (PIP₂) into inositol 1,4,5-trisphosphate (IP₃) and diacylglycerol (DAG). IP₃ diffuses to the , where it binds and opens IP₃ receptors (IP₃Rs), triggering Ca²⁺ release from intracellular stores and generating oscillatory Ca²⁺ signals that modulate neuronal excitability. Concurrently, DAG remains membrane-associated and recruits, along with elevated Ca²⁺, various (PKC) isoforms (such as PKCε in neuronal cells), leading to their activation through conformational changes and translocation to the plasma membrane. Activated PKC phosphorylates downstream targets, including the myristoylated alanine-rich C-kinase substrate (MARCKS), at serine residues within its effector domain, which disrupts MARCKS' electrostatic interactions with filaments and PIP₂. This phosphorylation promotes MARCKS translocation from the membrane to the , facilitating cytoskeleton remodeling and membrane protrusion essential for neurite extension and motility. The Ca²⁺ oscillations mediated by IP₃Rs further amplify these effects by coordinating local Ca²⁺ transients that drive cytoskeletal dynamics. Among Trk subtypes, TrkA exhibits the strongest PLCγ1 activation due to optimal docking geometry at Y785, whereas TrkC shows weaker coupling attributable to naturally occurring kinase domain inserts (e.g., 14- or 25-amino-acid sequences) that sterically hinder PLCγ1 binding and without affecting overall receptor autophosphorylation. TrkB displays intermediate efficiency, supporting robust signaling in central neurons. This pathway underpins key physiological processes, including enhancement of synaptic vesicle release through PKC-dependent phosphorylation of exocytotic machinery components, such as Munc13 and SNAP-25, which increases fusion probability at presynaptic terminals. In sensory neurons, TrkA-PLCγ1-PKC signaling contributes to pain hypersensitivity by sensitizing transient receptor potential vanilloid 1 (TRPV1) channels via DAG/PKC-mediated phosphorylation, amplifying nociceptive responses to thermal and inflammatory stimuli. Evidence for its role in development comes from studies showing that disruption of PLCγ1-TrkA interaction, as in Y785F mutants or PLCγ1-deficient models, significantly impairs NGF-induced neurite outgrowth in PC12 cells and primary neurons, reducing process elongation by up to 50% without affecting cell survival.

Physiological Functions

Neural development and survival

Trk receptors play a pivotal role in neural development by promoting the , , and of neuronal precursors during embryogenesis. such as (NGF), (BDNF), and (NT-3) bind to their cognate Trk receptors—TrkA, TrkB, and TrkC, respectively—to initiate signaling cascades that regulate these processes. Expression of Trk receptors peaks during embryonic stages, coinciding with critical periods of and neuronal maturation in the central and peripheral nervous systems. In early neural development, TrkC signaling via NT-3 supports the proliferation of , which give rise to diverse neuronal lineages including sensory and autonomic neurons. NT-3 acts as a for cultured cells, enhancing their division in a serum-free medium and contributing to the expansion of the progenitor pool before . Similarly, TrkA by NGF is essential for the survival of sympathetic neuron precursors; in the absence of this signaling, developing sympathetic neurons undergo , leading to substantial cell loss. Target-derived prevent in post-mitotic neurons through mediated by Trk receptors. Upon binding at terminals, activated Trk receptors are internalized into signaling endosomes and transported retrogradely to the cell body, where they inhibit pro-apoptotic pathways such as those involving PI3K/Akt. This mechanism ensures the survival of neurons that successfully innervate their targets during the period of naturally occurring in embryogenesis. TrkB signaling induced by BDNF promotes neuronal , particularly the arborization of dendrites in cortical neurons. In developing , BDNF enhances dendritic growth of pyramidal neurons, increasing branch complexity and length in an activity-dependent manner. This effect is mediated through TrkB activation, which coordinates cytoskeletal remodeling and changes essential for dendritic maturation. Genetic studies in mice underscore the quantitative impact of Trk signaling on neuronal survival. TrkA-null mice exhibit near-complete loss of sympathetic neurons and approximately 80% loss of sensory neurons by birth. TrkC-deficient mice show about 30–50% depletion of proprioceptive neurons in sensory ganglia, while TrkB knockouts lead to losses of up to 70% in certain populations. These phenotypes highlight the non-redundant roles of individual Trk receptors in maintaining neuronal numbers during . In humans, defects in Trk signaling contribute to neurodevelopmental disorders such as , where mutations in the MECP2 gene lead to impaired BDNF/TrkB phosphorylation and reduced neuronal survival and differentiation. Mouse models of demonstrate deficits in TrkB activation, resulting in abnormal dendritic arborization and motor impairments that can be partially rescued by TrkB agonists.

and target innervation

Trk receptors play a crucial role in by enabling neurons to sense and respond to gradients of their cognate , directing axonal growth toward appropriate targets. For instance, TrkA activation by (NGF) gradients guides sympathetic axons to their peripheral targets, such as sweat glands and blood vessels, through chemotactic attraction at the . Similarly, TrkC signaling in response to (NT-3) gradients serves as a chemoattractant for proprioceptive axons, facilitating their precise navigation to muscle spindles and other sensory targets in the limbs. Local activation of Trk receptors at the promotes directed advance by modulating the . Upon binding, Trk receptors trigger rapid signaling that reorganizes and dynamics, advancing the toward higher ligand concentrations; this process involves pathways such as PLCγ, which links receptor activation to cytoskeletal changes for enhanced motility. In compartmentalized cultures of sympathetic neurons, localized NGF application to distal axons induces TrkA-dependent turning and extension, underscoring the receptor's role in sensing independent of signals. Target innervation is refined through competition among axons for limited neurotrophins at shared targets, ensuring precise matching and elimination of excess projections. In dorsal root ganglion (DRG) neurons expressing TrkA, competition for target-derived NGF limits hyperinnervation, promoting selective stabilization of appropriate connections while excess axons retract due to insufficient trophic support. This competitive mechanism, observed in both sensory and sympathetic systems, balances neuronal numbers with target capacity during development. In the , TrkB signaling via (BDNF) contributes to the refinement of retinogeniculate projections, including the formation of columns. BDNF/TrkB activity supports activity-dependent segregation of retinal inputs in the , where disruptions in signaling lead to blurred eye-specific territories and altered topography. Genetic studies in Trk mutants highlight the receptors' essential functions in and innervation. TrkC-null mice exhibit severe defects in proprioceptive pathfinding, resulting in a profound loss of limb innervation and absence of muscle sensory endings, as proprioceptive neurons fail to reach their targets without NT-3/TrkC-mediated guidance. Similarly, targeted disruptions in TrkA signaling impair sympathetic targeting to glands and organs, demonstrating the receptors' non-redundant roles in establishing peripheral connectivity.

Synaptic plasticity and function

Trk receptors, particularly TrkB and TrkC, play essential roles in by modulating synapse formation, strengthening, and maintenance in both developing and mature neural circuits. Binding of neurotrophins such as (BDNF) to TrkB or (NT-3) to TrkC activates downstream pathways that regulate structural and functional changes at synapses, including dynamics and [long-term potentiation](/page/Long-term_p potentiation) (LTP). These processes are critical for learning, , and circuit refinement, with Trk signaling integrating activity-dependent cues to adapt synaptic efficacy. In the , TrkB activation by BDNF is required for the and of LTP, a cellular correlate of learning, primarily through the MAPK/ERK pathway. Genetic disruption of TrkB in mice impairs hippocampal LTP, as TrkB recruits phospholipase Cγ (PLCγ) to phosphorylate calcium/calmodulin-dependent protein kinase IV (CaMKIV), enhancing synaptic strengthening. Similarly, long-term depression () is altered in TrkB-deficient models, underscoring bidirectional regulation of synaptic weight by TrkB signaling. Retrograde transport of BDNF-TrkB endosomes further promotes density and stability in hippocampal neurons, with ERK1/2 activation necessary for BDNF-induced spine formation and maturation. TrkC signaling via NT-3 contributes to , particularly at mossy fiber-CA3 synapses in the , where it enhances presynaptic organization and terminal maturation. Lack of TrkC impairs mossy fiber , leading to reduced clustering and altered excitatory . At peripheral synapses, TrkB maintains integrity and function; inhibiting TrkB kinase activity disrupts cycling and at adult diaphragm s, highlighting its role in sustaining synaptic structure. Activity-dependent BDNF release, such as during seizures, upregulates TrkB expression to facilitate plasticity in regions like the and . Seizure-induced BDNF elevates TrkB mRNA levels transiently, promoting adaptive synaptic remodeling and potentially contributing to epileptogenesis through enhanced LTP-like mechanisms. In adult circuits, TrkA supports maintenance in the , preserving synaptic inputs to the ; TrkA antagonism withdraws cholinergic terminals, impairing cognitive function. In models, TrkB and TrkC deficits exacerbate synaptic loss and plasticity impairments, with amyloid-β toxicity disrupting BDNF-TrkB signaling and reducing LTP. Supporting evidence comes from experiments where BDNF infusion restores LTP in TrkB mutant mice, rescuing synaptic deficits and demonstrating the sufficiency of exogenous BDNF-TrkB activation for plasticity recovery. These findings emphasize Trk receptors' therapeutic potential in plasticity-related disorders.

Role in pain and sensory processing

TrkA receptors, activated by (NGF), play a central role in by sensitizing transient receptor potential vanilloid 1 ()-expressing sensory fibers to thermal and chemical stimuli. NGF binding to TrkA triggers intracellular signaling cascades, including (PI3K), which rapidly increases TRPV1 membrane expression and enhances its responsiveness, thereby lowering the threshold for heat-evoked pain in nociceptive dorsal root ganglion neurons. This sensitization is mediated through TrkA-dependent phosphorylation and trafficking of TRPV1 to the plasma membrane, contributing to acute inflammatory . In inflammatory conditions, such as or tissue injury, NGF and TrkA expression are upregulated in affected tissues and sensory neurons, amplifying pain signaling via sustained TRPV1 activation and release of pro-nociceptive mediators like . TrkC receptors, bound by neurotrophin-3 (NT-3), are essential for mechanosensation, particularly in low-threshold mechanoreceptors and proprioceptive pathways. NT-3/TrkC signaling supports the development and function of D-hair low-threshold s in , which detect gentle touch, and is critical for the survival and differentiation of muscle spindle afferents that mediate . In muscle spindles, NT-3 derived from intrafusal fibers activates TrkC on proprioceptive sensory neurons, ensuring proper synaptic connectivity to motoneurons and precise sensing of limb position and movement. Disruptions in NT-3/TrkC signaling lead to reduced mechanoreceptor populations and impaired tactile discrimination, highlighting their role in non-noxious sensory processing. In central pain processing, TrkB receptors, activated by (BDNF), modulate within the dorsal horn. BDNF/TrkB signaling enhances synaptic transmission between primary afferents and second-order neurons, promoting central sensitization and mechanical or thermal following peripheral injury. Upregulation of BDNF and TrkB in the and dorsal root ganglia contributes to persistent states, such as those induced by nerve ligation, by facilitating excitatory and inhibiting inhibitory . Pathophysiological dysregulation of Trk receptors underlies disorders, with TrkA antagonism emerging as a therapeutic strategy. Selective TrkA inhibitors or neutralizing antibodies block NGF/TrkA signaling, reducing sensitization and alleviating in preclinical models of without affecting non-noxious sensation. Phase III clinical trials of NGF-blocking monoclonal antibodies, such as , demonstrated significant pain reduction and improved function in patients with pain, though development was discontinued in 2021 due to concerns over joint safety risks. These interventions highlight TrkA's potential as a target for non-opioid analgesics in inflammatory and conditions.

Role in Cancer and Therapeutics

Oncogenic activations and fusions

Oncogenic activations of Trk receptors in cancer predominantly arise from chromosomal rearrangements that generate , leading to constitutive activity independent of binding. These fusions typically involve the 5' portion of a partner gene, which provides a dimerization motif, fused to the 3' domain of NTRK1 (encoding TrkA), NTRK2 (TrkB), or NTRK3 (TrkC), thereby promoting oligomerization and autophosphorylation of the receptor. The first such fusion, TPM3-NTRK1, was identified in 1986 in a colon cancer sample, marking the initial recognition of NTRK rearrangements as oncogenic drivers. Among NTRK1 fusions, common examples include TPM3-NTRK1, LMNA-NTRK1, and TPR-NTRK1, which occur in various solid tumors such as and , with reported frequencies of approximately 1-5% in cancers and less than 1% in lung cancers. NTRK2 fusions are rarer and include partners like AFAP1-NTRK2 or QKI-NTRK2, primarily observed in tumors or sarcomas at frequencies below 0.5% pan-cancer. For NTRK3, the ETV6-NTRK3 fusion is particularly prevalent, accounting for up to 75-100% of cases in secretory breast carcinoma and nearly 90% in .455118-3/fulltext) Overall, NTRK fusions exhibit a pan-cancer prevalence of about 0.3%, though this rises dramatically in certain rare tumor types, such as over 90% in mammary analogue secretory and infantile . These alterations drive tumorigenesis by sustaining signaling through pathways like MAPK/ERK, independent of ligands. Detection of NTRK fusions relies on methods such as (FISH) for specific rearrangements like ETV6-NTRK3, or RNA sequencing to identify novel partners, with next-generation sequencing (NGS) panels becoming standard in the 2020s for comprehensive profiling in clinical settings. using pan-TRK antibodies serves as a screening tool, though confirmatory molecular testing is essential due to potential non-specificity.45992-9/fulltext)

Trk receptors in tumorigenesis

Trk receptors contribute to tumorigenesis through mechanisms independent of gene fusions, including , overexpression, and activating point , which lead to ligand-independent or enhanced signaling. These alterations result in constitutive activation of downstream pathways such as MAPK/ERK and PI3K/Akt, promoting oncogenic behaviors in various malignancies. Overexpression of Trk receptors, often driven by autocrine or paracrine loops, is observed in multiple tumor types and correlates with aggressive phenotypes. In , overexpression of TrkB, particularly in high-risk cases with MYCN , drives uncontrolled through constitutive of MAPK and PI3K pathways, leading to tumor growth and poor prognosis. Similarly, TrkA overexpression in and cancers enhances proliferative signaling via these pathways, contributing to tumor progression. Activating , such as the G595R substitution in the NTRK1 kinase domain, are rare but functionally oncogenic, mimicking binding and sustaining in colorectal and other cancers. The TrkB/BDNF axis promotes invasion and metastasis by inducing epithelial-mesenchymal transition () in and cancers, facilitating tumor , anoikis resistance, and distant spread through activation of downstream effectors including PI3K/Akt and PLCγ/PKC signaling. In , this axis upregulates EMT markers like and downregulates E-cadherin, enhancing metastatic potential to sites such as the . cells with elevated TrkB exhibit increased invasiveness via similar paracrine BDNF stimulation. High TrkC levels correlate with increased microvessel density in primitive neuroectodermal tumors (PNET), suggesting a role in tumor angiogenesis. Activated Trk receptors confer chemotherapy resistance in ovarian cancer, where TrkB/BDNF signaling via PI3K/Akt inhibits apoptosis and enhances survival under drug stress, as seen in platinum-resistant cases with elevated receptor expression. NTRK1 amplification is rare in colorectal cancers (<1%) but can drive resistance and progression when present, amplifying oncogenic signaling.

Development and use of Trk inhibitors

The development of Trk inhibitors has focused on targeting oncogenic NTRK fusions across various solid tumors, leading to the approval of first-generation agents that marked a shift toward tumor-agnostic therapies. (LOXO-101; Vitrakvi) received accelerated FDA approval in November 2018 and full approval in April 2025 for adult and pediatric patients with solid tumors harboring NTRK gene fusions, irrespective of tumor , based on integrated phase I/II trial data demonstrating rapid and durable responses. followed with FDA approval in August 2019 for similar indications in patients aged 12 years and older, distinguished by its additional inhibition of ROS1 and ALK kinases and enhanced penetration, which addresses intracranial disease progression. These first-generation inhibitors are classified as type I agents, binding competitively to the ATP site in the active (DFG-in) conformation of Trk kinases, with exemplifying high selectivity for TrkA, TrkB, and TrkC ( values <1 nM). Second-generation Trk inhibitors emerged to overcome acquired , with selitrectinib (LOXO-195) designed as a next-generation type I that maintains ATP-competitive binding but exhibits improved potency against common mutations, such as solvent-front alterations. Selitrectinib binds wild-type Trk kinases with low nanomolar affinity ( <1 nM) and retains activity against mutants like TRKA G595R ( ~40 nM), enabling its use in patients progressing on first-line therapy. Repotrectinib (TPX-0005, Augtyro), another second-generation agent, shares type I characteristics but incorporates a macrocyclic for broader kinase coverage and superior brain penetration, receiving FDA accelerated approval in June 2024 for NTRK fusion-positive solid tumors in adults and children aged 12 and older. Resistance to Trk inhibitors arises through on-target mechanisms, such as the TRKA G595R solvent-front mutation, which sterically hinders type I inhibitor binding and has been identified in up to 30% of resistant cases post-larotrectinib or entrectinib treatment. Off-target resistance involves bypass pathway activation, including co-amplifications of MET or hotspot mutations in KRAS and BRAF, leading to sustained MAPK signaling despite Trk inhibition. In the 2020s, combination strategies have been pursued, such as co-targeting with MEK inhibitors (e.g., binimetinib) to block convergent ERK/MAPK pathway reactivation, showing preclinical synergy in restoring sensitivity to Trk inhibition in mutation-bearing models. Clinically, Trk inhibitors have achieved an overall response rate (ORR) of approximately 75% in NTRK fusion-positive tumors across phase I/II trials, with median duration of response exceeding 2 years in responsive patients. Approvals extend to pediatric sarcomas, where has demonstrated high efficacy in NTRK fusion-driven entities like , enabling durable remissions and supporting its use as frontline therapy in children under 18. For brain metastases, ongoing trials as of 2025 highlight repotrectinib's intracranial ORR of around 80% in NTRK-positive cases, building on its CNS-penetrant profile to address unmet needs in metastatic disease. Key challenges include class-specific toxicities from Trk inhibition, with reported in over 50% of patients and in up to 40%, both linked to on-target effects on neuronal Trk signaling and often manageable with dose adjustments. Successful implementation requires biomarker-driven selection, primarily through next-generation sequencing (NGS) to detect NTRK fusions at low prevalence (0.1-1% across solid tumors), ensuring precise patient stratification.

References

  1. [1]
    Tropomyosin receptor kinase (TRK) biology and the role of NTRK ...
    Nov 18, 2019 · This review describes the normal function and physiology of TRK receptors and the biology behind NTRK gene fusions and how they act as oncogenic drivers in ...
  2. [2]
    Neurotrophins: Roles in Neuronal Development and Function - PMC
    Neurotrophins activate two different classes of receptors, the Trk family of receptor tyrosine kinases and p75NTR, a member of the TNF receptor superfamily.
  3. [3]
    Insights into Current Tropomyosin Receptor Kinase (TRK) Inhibitors
    In this role, the TRK family regulates cell differentiation, proliferation, survival, and pain. There are three members of the TRK family: TRKA (encoded by ...
  4. [4]
    A human oncogene formed by the fusion of truncated tropomyosin ...
    Feb 27, 1986 · Published: 27 February 1986. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Dionisio ...
  5. [5]
    Molecular and biochemical characterization of the human trk proto ...
    Mitra G., Martin-Zanca D., Barbacid M. Identification and biochemical characterization of p70TRK, product of the human TRK oncogene. Proc Natl Acad Sci U S ...
  6. [6]
    a Signal Transducing Receptor for Nerve Growth Factor - Science
    The trk proto-oncogene encodes a 140-kilodalton, membrane-spanning protein tyrosine kinase (p140prototrk) that is expressed only in neural tissues.
  7. [7]
    Molecular and biochemical characterization of the human trk proto ...
    Molecular analysis of the human trk oncogene, a transforming gene isolated ... Authors. D Martin-Zanca , R Oskam, G Mitra, T Copeland, M Barbacid ...Missing: 1982 | Show results with:1982
  8. [8]
    p04629 · ntrk1_human - UniProt
    Miscellaneous. Trk also stands for tropomyosin-related kinase since it was first isolated as an oncogenic protein which was the result of a fusion between the ...
  9. [9]
    Mechanism of activation of the human trk oncogene - PubMed - NIH
    The human trk oncogene was generated by a genetic rearrangement that replaced the extracellular domain of the normal trk tyrosine kinase receptor.Missing: 1982 | Show results with:1982
  10. [10]
  11. [11]
  12. [12]
  13. [13]
    Assembly of Neuronal Connectivity by Neurotrophic Factors and ...
    Aug 8, 2016 · The extracellular domains of the Trk receptors contain two cysteine-rich clusters flanking three leucine-rich repeat (LRR) motives, followed by ...
  14. [14]
    Molecular determinants of signal transduction in tropomyosin ...
    Oct 6, 2025 · Trk receptors display sequence and structural homology across their approximately 800 amino acid length.
  15. [15]
    Mechanisms of Activation of Receptor Tyrosine Kinases: Monomers ...
    TrkA, TrkB and TrkC share significant sequence homology and a conserved domain organization comprising, from the N-terminus to C-terminus, an extracellular ...
  16. [16]
    A function–structure model for NGF‐activated TRK - EMBO Press
    In a biologically based study of TRK activation loop tyrosines, we found that mutation of either Tyr670, Tyr674 or Tyr675 significantly impairs NGF‐inducible ...
  17. [17]
    Molecular Kinetics of Nerve Growth Factor Receptor Trafficking and ...
    Pulse-chase experiments indicate that TrkA is synthesized as a 110-kDa N-glycosylated precursor that leads to the mature 140-kDa form of the receptor with a ...
  18. [18]
    Comparison of tyrosine kinase domain properties ... - Portland Press
    Oct 29, 2020 · The Trks share a high degree of homology and utilize overlapping signaling pathways, yet their signaling is associated with starkly different ...
  19. [19]
    TrkA glycosylation regulates receptor localization and activity - Watson
    Apr 9, 1999 · The human nerve growth factor receptor (TrkA) contains four potential N-glycosylation sites that are highly conserved within the Trk family ...
  20. [20]
    Novel transcripts reveal a complex structure of the human TRKA ...
    Nov 18, 2015 · The transmembrane domain of TRKA is encoded by exon 11 and the intracellular tyrosine kinase domain by exons 13…17 [19, 20]. The glycosylation ...
  21. [21]
    Regulation of neurotrophin receptor (Trk) signaling - Frontiers
    Tyrosine residues at positions 670, 674, and 675 are located in the activation loop of the human TrkA tyrosine kinase domain. Tyrosine 490 forms a PTB ...Missing: Y516 | Show results with:Y516
  22. [22]
    NTRK1 neurotrophic receptor tyrosine kinase 1 [ (human)] - NCBI
    Sep 5, 2025 · This gene encodes a member of the neurotrophic tyrosine kinase receptor (NTKR) family. This kinase is a membrane-bound receptor that, upon neurotrophin binding ...
  23. [23]
    NTRK1 Gene - Neurotrophic Receptor Tyrosine Kinase 1 - GeneCards
    This gene encodes a member of the neurotrophic tyrosine kinase receptor (NTKR) family. This kinase is a membrane-bound receptor that, upon neurotrophin ...
  24. [24]
    Tissue-specific alternative splicing generates two isoforms of the ...
    In this report, we show that alternative splicing results in the production of two distinct trkA isoforms in both rats and humans.
  25. [25]
    mKlf7, a potential transcriptional regulator of TrkA nerve growth ...
    Apr 1, 2001 · Using in situ hybridization, we demonstrate that mKlf7 is coexpressed with TrkA in sensory and sympathetic neurons during embryogenesis and in ...
  26. [26]
    A Role for TrkA during Maturation of Striatal and Basal Forebrain ...
    Oct 15, 1997 · Our findings suggest that TrkA signaling is required for the normal maturation and possibly the survival of both basal forebrain and striatal ...
  27. [27]
    TrkA Receptor - an overview | ScienceDirect Topics
    TrkA binds NGF with high affinity ( K D = 10 −11 M), and upon binding dimerizes to form a symmetric 2:2 signalling complex with the NGF dimer. The NGF/TrkA ...
  28. [28]
    effect of TRKA (NTRK1) missense mutations on autophosphorylation ...
    The short C-terminal tail of 15 amino acids includes a conserved tyrosine residue (–785Tyr in TRKA) which is responsible for binding of PLCγ-1 (29,30).
  29. [29]
    Mutations in the TRKA/NGF receptor gene in patients with ... - Nature
    Aug 1, 1996 · Congenital insensitivity to pain with anhidrosis (CIPA; MIM 256800) is an autosomal-recessive disorder characterized by recurrent episodes ...
  30. [30]
    Structural analysis of TrkA mutations in patients with congenital ...
    Apr 26, 2022 · To investigate the relevance of the TrkA-PLCγ interaction in pain signaling, we synthesized a peptide based on the region of TrkA that anchors ...Missing: Y785 | Show results with:Y785
  31. [31]
    Tropomyosin Receptor Kinase B - an overview | ScienceDirect Topics
    Both TrkB and p75NTR are single-transmembrane receptors. The human NTRK2 gene encodes TrkB (Trk2) of 838 aa residues. The human NGFR gene encodes p75NTR of 427 ...
  32. [32]
    TrkB Truncated Isoform Receptors as Transducers and ... - Frontiers
    Thus, for a long time, the main functions attributed to these truncated receptor isoforms were of acting as dominant-negative inhibitors of the full-length ...
  33. [33]
    Evidence that truncated TrkB isoform, TrkB-Shc can regulate ...
    In this study, we analyzed the biological function of the TrkB-Shc receptor in response to exogenous BDNF treatment. In experiments transiently overexpressing ...
  34. [34]
    TrkB Receptor Signalling: Implications in Neurodegenerative ... - MDPI
    TrkB is expressed in both the central (CNS) and peripheral nervous systems (PNS). In the CNS, a high TrkB expression is observed in cerebral cortex, hippocampus ...
  35. [35]
    TrkB Receptor Signaling Regulates Developmental Death Dynamics ...
    Nov 5, 2003 · ... TrkB signaling is required for postnatal survival of CNS neurons and protects hippocampal and motor neurons from axotomy-induced cell death.
  36. [36]
    neurotrophic receptor tyrosine kinase 2 | Type VII RTKs
    Gene and Protein Information Click here for help. Species, TM, AA, Chromosomal Location, Gene Symbol, Gene Name, Reference. Human, 1, 822, 9q21.33, NTRK2 ...
  37. [37]
    Experience‐dependent regulation of TrkB isoforms in rodent visual ...
    Feb 17, 2009 · These isoforms are important components of BDNF signaling ... Critical periods for experience-dependent synaptic scaling in visual cortex.
  38. [38]
    TrkB Agonists Ameliorate Obesity and Associated Metabolic ...
    Mice (5) or humans (6) with a hypomorphic or loss-of-function allele of the tyrosine kinase receptor trkB display excessive appetite, reduced energy expenditure ...Missing: seminal | Show results with:seminal
  39. [39]
    NTRK3 neurotrophic receptor tyrosine kinase 3 [ (human)] - NCBI
    Sep 9, 2025 · Low TRKC is associated with low vincristine and lomustine response in medulloblastoma. Mesenchymal stem cells modified by adenovirus carrying ...
  40. [40]
    NTRK3 Gene - Neurotrophic Receptor Tyrosine Kinase 3 - GeneCards
    Protein attributes for NTRK3 Gene. Size: 839 amino acids; Molecular mass: 94428 Da. Protein existence level: PE1. Quaternary structure: Exists in a dynamic ...
  41. [41]
    q16288 · ntrk3_human - UniProt
    Receptor tyrosine kinase involved in nervous system and probably heart development. Upon binding of its ligand NTF3/neurotrophin-3, NTRK3 autophosphorylates ...Missing: full- | Show results with:full-
  42. [42]
    Molecular cloning of the cDNA for human TrkC (NTRK3 ... - PubMed
    We cloned one human splice variant that has a nucleic acid sequence identical to the rat isoform with an insert of 14 amino acids. The human trkC cDNA also has ...Missing: full- length
  43. [43]
    TrkC Isoforms with Inserts in the Kinase Domain Show Impaired ...
    We show that the TrkC kinase insert isoforms fail to transduce signals to known substrates, induce immediate early gene transcription, or efficiently ...
  44. [44]
    trkC, a receptor for neurotrophin-3, is widely expressed in the ...
    Jun 1, 1993 · Here we present a developmental expression study of trkC, which encodes a receptor for neurotrophin-3 (NT-3). Like the related genes, trk and ...
  45. [45]
    Developmental expression of trkC, the neurotrophin-3 receptor, in ...
    Jan 1, 1994 · In later embryonic development, trkC is expressed in various structures of the CNS including the caudatoputamen, septal nuclei, cerebellum, and ...Missing: hematopoietic | Show results with:hematopoietic
  46. [46]
    Expression of Neurotrophins and their Receptors in Human Bone ...
    Certain hematopoietic cells were found to be immunoreactive for only one member of the Trk family receptors. For example, neutrophil precursors expressed both ...Missing: cerebellum | Show results with:cerebellum
  47. [47]
    Roles of TrkC Signaling in the Regulation of Tumorigenicity and ...
    TrkC and TrkC fusion proteins are associated with a variety of human cancers and play essential roles in the progression and metastasis of human cancers.
  48. [48]
    Expression of the neurotrophin receptor TrkC is linked to a ... - PNAS
    The level of trkC expression was highly variable, with a more than 50-fold difference between the highest and lowest values. By Kaplan-Meier analysis, patients ...
  49. [49]
    Neurotrophin Signaling in Medulloblastoma - PMC - PubMed Central
    High TrkC mRNA expression is frequent in SHH MB [121]. Our analysis showed NTRK3 to be increased across all 4 subtypes of SHH tumors (Figure 4A). Evaluation of ...
  50. [50]
    The ETV6-NTRK3 chimeric tyrosine kinase suppresses TGF ... - PNAS
    TrkC is frequently overexpressed in human cancers including pancreatic and prostate carcinoma, neuroblastoma, and medulloblastoma (25, 26). Moreover, recent ...
  51. [51]
    Neurotrophin-3: a Neurotrophic Factor Related to NGF and BDNF
    BARDE, Y.A., PURIFICATION OF A NEW NEUROTROPHIC FACTOR FROM MAMMALIAN BRAIN, EMBO JOURNAL 1: 549 (1982). Crossref · PubMed · Web of Science · Google Scholar.
  52. [52]
    Differential expression of nerve growth factor receptors ... - PNAS
    The ability of these cells to respond to NGF implies that some biological responses may be uncoupled from high-affinity binding. Responsiveness to NT-3.
  53. [53]
    High-affinity TrkA and p75 neurotrophin receptor complexes - PMC
    The Trk family has three members, namely TrkA, TrkB, and TrkC. Each of these has preferred neurotrophin binding partner(s); TrkA binds preferentially to NGF, ...<|control11|><|separator|>
  54. [54]
    Targeting Neurotrophins to Specific Populations of Neurons - MDPI
    We focus on nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3), and their effects on populations of neurons within ...
  55. [55]
    Trk Receptors and Neurotrophin Cross-Interactions - PubMed Central
    May 3, 2017 · The Trk binding sites on some neurotrophins are predicted from their known structures, such as NT-3 (Gong et al., 2008), NT-4 and NT-4/BDNF ...
  56. [56]
    Selective activation and down-regulation of Trk receptors ... - PubMed
    We found that NT3 activates TrkC at concentrations lower than those of BDNF needed to activate TrkB. Downstream of Trk activation, the changes in gene ...
  57. [57]
    Specific High-Affinity Receptors for Neurotrophin-3 on Sympathetic ...
    These sympathetic neurons (which can be rescued by the addition of NGF) display high-affinity receptors for NT-3. (Kd of 1.8 x lo-'* M) that cannot be ...<|control11|><|separator|>
  58. [58]
    The Crystal Structure of TrkB-d5 in Complex with Neurotrophin-4/5
    Although helical conformations are teractions between TrkB and NGF, potentially explaining the much-reduced affinity of NGF for TrkB compared to observed for ...
  59. [59]
    Neurotrophins and Neuropathic Pain: Role in Pathobiology - PMC
    NT-3 may also influence TrkA-positive neurons [157,158], but a functional role for NT-3-TrkA signalling remains to be elucidated. 15. Pronociceptive Effects ...
  60. [60]
    NGF and Neurotrophin-3 Both Activate TrkA on Sympathetic ...
    These data suggest that NGF and NT-3 differentially regulate the TrkA receptor both with regard to activation time course and downstream targets, leading to ...
  61. [61]
    Extracellular pH and Neuronal Depolarization Serve as Dynamic ...
    May 8, 2013 · Mobilization of trkA to the membrane at pH 6.5 was abolished in neurons treated with the acid-sensitive ion channel blocker, amiloride. While ...
  62. [62]
    Revising the mechanism of p75NTR activation - Nature
    Aug 13, 2020 · p75NTR is a type I transmembrane protein and is composed of an extracellular ligand-binding domain (ECD), a single-span helical transmembrane ...
  63. [63]
    Structural basis of death domain signaling in the p75 neurotrophin ...
    Dec 8, 2015 · Here we report structures of complexes formed by the DD of p75 neurotrophin receptor (p75 NTR ) with RhoGDI, for activation of the RhoA pathway.
  64. [64]
    Structural Model for p75NTR–TrkA Intracellular Domain Interaction
    In the present work, we demonstrate by Förster resonance energy transfer that p75NTR and TrkA are physically associated through their intracellular (IC) domains ...Missing: seminal papers
  65. [65]
    Interaction between the transmembrane domains of neurotrophin ...
    We provide a structural model of the p75–TrkA receptor complex necessary for neuronal development stabilized by TMD interactions.
  66. [66]
    Dynamic nature of the p75 neurotrophin receptor in response to ...
    Heterodimerization of p75NTR with Trk receptors increases the affinity of the mature neurotrophin/Trk interaction by about 100-fold and enhances pro ...
  67. [67]
    High-affinity TrkA and p75 neurotrophin receptor complexes
    Jan 16, 2022 · The TrkA receptor consists of five extracellular domains (D1–D5), an α-helical transmembrane domain, and an intracellular segment containing a ...
  68. [68]
    the endocytic trafficking of p75NTR is driven by TrkA and regulates ...
    It is becoming apparent that p75NTR has a fundamental role in modulating TrkA function. p75NTR affects both the ligand affinity and the ligand-induced ...
  69. [69]
    p75 neurotrophin receptor reduces ligand‐induced ... - EMBO Press
    These results indicate that p75NTR may prolong cell‐surface Trk‐dependent signalling events by negatively regulating receptor ubiquitination.
  70. [70]
    Transgenic Mice Expressing the Intracellular Domain of the p75 ...
    Sep 15, 1997 · p75NTR−/− mice show reductions in NGF-mediated survival (Davies et al., 1993), a trkA-dependent response, consistent with the hypothesis that ...
  71. [71]
    Mechanisms of Neurotrophin Trafficking via Trk Receptors - PMC - NIH
    Mar 27, 2018 · Neurotrophins bind Trk receptor tyrosine kinases in axon terminals to promote endocytosis of ligand-bound phosphorylated receptors into signaling endosomes.
  72. [72]
    TrkB Truncated Isoform Receptors as Transducers and ... - PMC - NIH
    Mar 7, 2022 · However, Trk genes by alternative splicing can produce a wide array of other isoforms. For many of these isoforms the pattern of expression ...
  73. [73]
    A Review on Ubiquitination of Neurotrophin Receptors
    Mar 14, 2017 · Here we review what is currently known regarding the ubiquitination of neurotrophin receptors and its physiological and pathological relevance.Missing: palmitoylation | Show results with:palmitoylation
  74. [74]
    Overexpression of miR-128 specifically inhibits the truncated isoform ...
    Dec 10, 2010 · Overexpression of miR-128 specifically inhibits the truncated isoform of NTRK3 and upregulates BCL2 in SH-SY5Y neuroblastoma cells · Authors.
  75. [75]
    Binding of laminin-1 to monosialoganglioside GM1 in lipid rafts is ...
    Laminin-1-induced clustering of TrkA in DRG cells observed by electromicroscopy ... Ganglioside GM1 binds to the Trk protein and regulates receptor function.
  76. [76]
    Crystal structures of the neurotrophin-binding domain of TrkA, TrkB ...
    Jul 2, 1999 · The crystal structures of the ligand binding domain of TrkA, TrkB, and TrkC were solved and refined to high resolution.Missing: cryo- EM
  77. [77]
    Structural basis of the transmembrane domain dimerization and ...
    Our findings indicate that the transmembrane and juxtamembrane regions of TRKA play key roles in its dimerization and activation by NGF.
  78. [78]
    Structural basis for the transmembrane signaling and antidepressant ...
    Oct 29, 2024 · In the present study, we employ NMR spectroscopy to solve the structure of the TrkB dimeric transmembrane domain in the lipid environment.
  79. [79]
    Dissecting the Roles of Tyrosines 490 and 785 of TrkA Protein in the ...
    There are only two established phosphotyrosine-docking sites (Tyr-490 and Tyr-785 on TrkA) that are known to be directly involved in signal transduction.
  80. [80]
    Rapid Retrograde Tyrosine Phosphorylation of trkA and Other ...
    Other proximal proteins, including several apparently localized in cell bodies, displayed increased tyrosine phosphorylation within 5–15 min. However, no ...<|control11|><|separator|>
  81. [81]
    Trk Receptors Function As Rapid Retrograde Signal Carriers in the ...
    Sep 15, 1997 · In mature nerves, activated Trk receptors function as rapid retrograde signal carriers to execute remote responses to target-derived neurotrophins.
  82. [82]
    Neurotrophin-regulated signalling pathways - PMC - PubMed Central
    Similar to Shc, Frs2 is phosphorylated by active Trk receptors. ... Frs2 also activates the Grb2–SOS–Ras pathway. Frs2 also couples Trk signalling to Src ...
  83. [83]
    Inhibition of MAP Kinase Kinase Blocks the Differentiation of PC-12 ...
    We show here that PD98059 can completely inhibit the activation of MAP kinase by NGF in PC-12 cells, presumably due to the blockade of its upstream activator.
  84. [84]
    Role of Kinase Suppressor of Ras-1 in Neuronal Survival Signaling ...
    Oct 17, 2007 · Scaffolding proteins including kinase suppressor of Ras-1 (KSR1) determine specificity of signaling by extracellular signal-regulated kinase ...
  85. [85]
  86. [86]
    Regulation of neuronal survival by the serine-threonine protein ...
    These findings suggest that in the developing nervous system, Akt is a critical mediator of growth factor-induced neuronal survival.Missing: PI3K Trk
  87. [87]
    Roles of the PI3K/AKT/mTOR signalling pathways in ...
    Apr 1, 2020 · Inhibition of any part of the RAS/PI3K/AKT pathway significantly reduces the survival of cultured sympathetic neurons in the presence of nerve ...The Pi3k/akt/mtor Signalling... · Akt Target Proteins · Pi3k/akt/mtor Inhibitors
  88. [88]
    Phosphatidylinositol 3-Kinase and Akt Protein Kinase Are ...
    Apr 15, 1998 · These results demonstrate that PI 3-kinase and Akt are both necessary and sufficient for the survival of NGF-dependent sympathetic neurons.
  89. [89]
    Neurotrophin-regulated signalling pathways - Journals
    Consequently, in the presence of Trk signalling, activation of the NF-κB pathway makes a synergistic contribution to neuronal survival. Interactions between ...Neurotrophin-Regulated... · 2. Neurotrophins And Their... · 6. P75 Neurotrophin...<|control11|><|separator|>
  90. [90]
    ProNGF Induces PTEN via p75NTR to Suppress Trk-Mediated ...
    Nov 17, 2010 · Here, we demonstrate that induction of the PTEN phosphatase is an important mechanism involved in this cross talk by regulating Akt activation.Prongf Increases Pten In Bf... · Pten Inhibition Allows... · The Role Of Pten In...
  91. [91]
    Tyrosine 785 is a major determinant of Trk‐‐substrate interaction.
    Mar 1, 1993 · Furthermore, a tyrosine‐phosphorylated synthetic C‐terminal peptide competitively inhibited Trk cytoplasmic domain association with PLC gamma.
  92. [92]
    PLC-gamma1 signalling - Reactome Pathway Database
    PLC-gamma is activated through the phosphorylation by TrkA receptor kinase and this form hydrolyses PIP2 to generate inositol tris-phosphate (IP3) and ...Missing: PLCγ | Show results with:PLCγ
  93. [93]
    Analysis of mitogen-activated protein kinase activation by naturally ...
    The inserts also block the ability of TrkC to form a high-affinity complex with Shc and phospholipase Cγ(PLCγ) and the activation of PtdIns 3-kinase, and ...Missing: variations | Show results with:variations
  94. [94]
    Muscle Contraction Regulates BDNF/TrkB Signaling to Modulate ...
    Here, we investigated the hypothesis that nerve-induced muscle activity regulates BDNF/TrkB signaling pathway to modulate synaptic function via activation of ...
  95. [95]
    Trk signaling regulates neural precursor cell proliferation and ...
    Dec 15, 2007 · Here, we asked whether neurotrophin-mediated signaling through Trk tyrosine kinase receptors is important for embryonic cortical precursor cell ...Missing: seminal | Show results with:seminal
  96. [96]
    Characterization of an NGF–P-TrkA Retrograde-Signaling Complex ...
    Oct 1, 1999 · NGF promotes growth and survival of neurons via activation of the receptor tyrosine kinase TrkA. We used compartmentalized cultures of ...
  97. [97]
    Neurotrophins regulate dendritic growth in developing visual cortex
    On both apical and basal dendrites, the effects of the TrkB receptor ligands, BDNF and NT-4, were distinct. The spectrum of neurotrophic actions and the laminar ...Missing: arborization | Show results with:arborization
  98. [98]
    Characterization of sensory deficits in TrkB knockout mice
    Mice lacking TrkB lost 32% of neurons, corresponding to the intermediate-sized and neurocalcin-positive ones. This neuronal lost was accomplished by the absence ...
  99. [99]
    A TrkB Small Molecule Partial Agonist Rescues TrkB ...
    Feb 1, 2012 · These data provide new insight into the role of BDNF signaling deficits in the pathophysiology of RTT and highlight TrkB as a possible ...
  100. [100]
    The trkA Receptor Mediates Growth Cone Turning toward a ...
    However, roles for NGF in axonal guidance are suggested by the early production of neurotrophins in targets of DRG and sympathetic axons (Ebendal and Persson, ...
  101. [101]
    A Chemoattractant Role for NT-3 in Proprioceptive Axon Guidance
    Our results show that survival factor NT-3 acts as a short-distance axon guidance molecule for muscle sensory afferents as they approach their proper targets.
  102. [102]
    A Model for Neuronal Competition During Development - PMC - NIH
    Apr 18, 2008 · We report that developmental competition between sympathetic neurons for survival is critically dependent on a sensitization process initiated by target ...
  103. [103]
    The sympathetic nervous system in development and disease - PMC
    During late embryonic and neonatal stages, there is excessive axonal branching in final target fields and competition for target-derived NGF eliminates neurons ...Axon Growth And Target... · Fig. 2 |. Axon Growth And... · Neuron Survival<|control11|><|separator|>
  104. [104]
    Effects of trkB knockout on topography and ocular segregation of ...
    We conclude that the normal topographic organization of uncrossed retinal projections depends upon trkB signaling, whereas the segregation of crossed and ...
  105. [105]
    Rapid Regulation of Brain-Derived Neurotrophic Factor mRNA ... - NIH
    Thus, blockade of retinal activity in one eye leads to a selective downregulation of BDNF mRNA within ocular dominance columns corresponding to that eye. BDNF ...
  106. [106]
    Targeted deletion of all isoforms of the trkC gene suggests the use of ...
    Neuronal counts of sensory ganglia in the trkC mutant mice reveal less severe losses than those in NT-3 null mutant mice, strongly suggesting that NT-3, in vivo ...
  107. [107]
    Article A Neurotrophin Signaling Cascade Coordinates Sympathetic ...
    Interestingly, both genetic and biochemical analyses have shown that NT-3, like NGF, promotes survival of sympathetic neurons via the receptor tyrosine kinase ...
  108. [108]
    Mechanism of TrkB-mediated hippocampal long-term potentiation
    Our results provide genetic evidence that TrkB mediates hippocampal plasticity via recruitment of PLCgamma, and by subsequent phosphorylation of CaMKIV and ...Missing: seminal paper
  109. [109]
    ERK1/2 Activation Is Necessary for BDNF to Increase Dendritic ...
    We demonstrated recently that BDNF/TrkB signaling increases dendritic spine density in hippocampal CA1 pyramidal neurons.
  110. [110]
    TrkB kinase activity maintains synaptic function and structural ...
    Inhibiting TrkB kinase activity for 7 days resulted in significant, yet reversible, impairments in neuromuscular transmission at diaphragm NMJs. Neuromuscular ...
  111. [111]
    The Role of Brain-Derived Neurotrophic Factor in Epileptogenesis
    Nov 25, 2021 · The hypothesis suggested that the expression of BDNF can suppress epileptogenesis by a combined receptor-ligand pathway; and the TrkB binding to ...
  112. [112]
    A nerve growth factor mimetic TrkA antagonist causes withdrawal of ...
    These observations support the notion that endogenously produced NGF acting through TrkA receptors is involved in the maintenance of the cholinergic phenotype ...
  113. [113]
    Regulation of BDNF-TrkB Signaling and Potential Therapeutic ...
    ... TrkB and interacts with it through the immunoglobulin constant 2 (Ig-C2) domain [1,2]. BDNF is widely distributed in the cortical areas, hippocampus, visual ...
  114. [114]
    NGF rapidly increases membrane expression of TRPV1 heat-gated ...
    Here we show that NGF, acting on the TrkA receptor, activates a signalling pathway in which PI3 kinase plays a crucial early role.
  115. [115]
    Signalling pathways involved in the sensitisation of mouse ... - NIH
    2003) The sensitisation caused by NGF is mediated by the TrkA receptor, because in p75NTR-null mice NGF can still induce thermal hyperalgesia, indicating that ...Missing: neurite outgrowth
  116. [116]
    NGF as a mediator of inflammatory pain - Journals
    In a wide variety of experimental inflammatory conditions NGF levels are rapidly increased in the inflamed tissue. 3. The high-affinity NGF receptor, trkA ...
  117. [117]
    Nerve Growth Factor Signaling and Its Contribution to Pain - PMC
    May 26, 2020 · TrkA has a higher affinity for mature NGF than for proNGF and activates neurotrophic signaling., P75NTR has a higher affinity for proNGF and can ...Missing: Y785 | Show results with:Y785
  118. [118]
    Muscle Spindle-Derived Neurotrophin 3 Regulates Synaptic ...
    May 1, 2002 · Proprioceptive afferents express the tyrosine kinase C (trkC) receptor, and fetal muscles express NT3, the major trkC ligand. The null mutation ...
  119. [119]
    Neurotrophin-3 Promotes the Differentiation of Muscle Spindle ...
    Together, our results indicate that NT3 signaling is both necessary and sufficient for the development of the proprioceptive phenotype, even in the absence of ...<|control11|><|separator|>
  120. [120]
    TrkB Signaling Is Required for Both the Induction and Maintenance ...
    The hyperalgesia is presumed to arise from a central sensitization of spinal cord dorsal horn circuits secondary to the intense afferent discharge induced by ...
  121. [121]
    Up-regulation of dorsal root ganglia BDNF and trkB receptor ... - PMC
    Brain-derived neurotrophic factor (BDNF) plays a neuromodulatory role in spinal cord dorsal horn via the post-synaptic tyrosine protein kinase B (trkB) receptor ...
  122. [122]
    Selective inhibition of tropomyosin-receptor-kinase A (TrkA) reduces ...
    May 4, 2016 · NGF blockade has reduced arthritis pain in clinical trials. We explored the mechanisms within the joint which may contribute to the analgesic ...
  123. [123]
    NERVE GROWTH FACTOR (NGF) BLOCKADE FOR THE ... - NIH
    NGF blockade continues to represent a promising new approach for the treatment of OA pain, but the actual benefits and risks remain to be fully elucidated.
  124. [124]
    A review of NTRK fusions in cancer - PMC - PubMed Central
    The prevalence of NTRK gene fusion is under 1% in solid tumors, but is highly seen in rare tumors such as mammary secretory carcinoma, mammary analogue ...
  125. [125]
    Systematic review of NTRK 1/2/3 fusion prevalence pan-cancer and ...
    Mar 13, 2023 · Specific NTRK fusions have been found at a high prevalence in a handful of rare cancer types, but otherwise are widely dispersed and uncommon ...
  126. [126]
    Genomic context of NTRK1/2/3 fusion-positive tumours from a large ...
    Jul 20, 2021 · This study aimed to interrogate a large real-world database of comprehensive genomic profiling data to describe the genomic landscape and prevalence of NTRK ...
  127. [127]
    NTRK gene fusions as novel targets of cancer therapy across ...
    The NTRK2 gene is mapped on chromosome 9q22.17 and contains 24 exons,8 coding for a protein of 822 amino acid residues (TrkB receptor). ... The NTRK3 gene is ...
  128. [128]
  129. [129]
    Regulating Tumorigenicity and Cancer Metastasis through TRKA ...
    This review summarizes the characteristics and research progress of TRKA, focusing on the regulatory role of the TRKA signal pathway in different tumors. In ...
  130. [130]
    Nerve growth factor & TrkA as novel therapeutic targets in cancer
    NGF and its receptors TrkA & p75NTR were recognized to be overexpressed in the overwhelming majority of human solid cancers.
  131. [131]
    Expression of the TrkA or TrkB receptor tyrosine kinase alters the ...
    TrkA enhances differentiation and inhibits proliferation and angiogenesis of neuroblastoma ... Non-canonical PI3K-Cdc42-Pak-Mek-Erk Signaling Promotes ...
  132. [132]
    Neurotrophins and their receptors in breast cancer - ScienceDirect
    The tyrosine kinase receptor TrkB, which binds BDNF and NT-4/5, is regarded ... Lagadec et al. TrkA overexpression enhances growth and metastasis of breast cancer ...The Neurotrophins And Their... · Bdnf, Nt-4/5, With P75 And... · Prongf Stimulates Breast...Missing: EMT | Show results with:EMT
  133. [133]
    NTRK insights: best practices for pathologists | Modern Pathology
    Sep 16, 2021 · The first tropomyosin-receptor kinase (TRK) fusion protein was found in 1982 in a colorectal adenocarcinoma cell line. NTRK1–3 fusions have now ...
  134. [134]
    Estradiol induces BDNF/TrkB signaling in triple-negative breast ...
    Feb 22, 2019 · Estradiol induces BDNF/TrkB signaling in triple-negative breast cancer to promote brain metastases ... BDNF/TRKB axis provokes EMT ...
  135. [135]
    Activation of BDNF/TrkB pathway promotes prostate cancer ...
    In vitro studies showed that stimulation with rhBDNF or overexpression of TrkB in PCa cells promoted cell migration, invasion, and anoikis resistance.
  136. [136]
    Perineural invasion-associated biomarkers for tumor development
    BDNF and its receptor TrkB are upregulated in PC tissues. Activation of the BDNF/TrkB pathway can promote PC progression by inducing an EMT-like transition ...
  137. [137]
    Angiogenic profile of childhood primitive neuroectodermal brain ...
    Our results indicate that all PNET/MB tested produce a wide range of angiogenic factors that are, individually or together, likely to play a direct role in PNET ...
  138. [138]
    Up-Regulation of miR-204 Enhances Anoikis Sensitivity in Epithelial ...
    High expression levels of TrkB and BDNF are thought to be associated with ovarian cancer, through phosphatidylinositol-3-kinase pathway, leading to metastasis ...
  139. [139]
    Prevalence and clinico-genomic characteristics of patients with TRK ...
    Aug 11, 2023 · The most frequent NTRK amplification occurred in NTRK1 (92%, 79/86), followed by NTRK3 (7%, 6/86), and NTRK2 (1%, 1/86). The frequency of NTRK ...
  140. [140]
    FDA approves larotrectinib for solid tumors with NTRK gene fusions
    Dec 14, 2018 · On November 26, 2018, the Food and Drug Administration granted accelerated approval to larotrectinib (VITRAKVI, Loxo Oncology Inc.
  141. [141]
    Entrectinib for the Treatment of NTRK gene Fusion Solid Tumors
    Abstract. The FDA-approved entrectinib on August 15, 2019, for the treatment of adult and pediatric patients 12 years of age and older with solid tumors.
  142. [142]
    Definition of Trk inhibitor LOXO-101 - NCI Dictionary of Cancer Terms
    Trk inhibitor LOXO-101 blocks the TRK protein, which may help keep cancer cells from growing and may kill them. It is a type of tyrosine kinase inhibitor.Missing: selitrectinib | Show results with:selitrectinib
  143. [143]
    TRK inhibitors in TRK fusion-positive cancers - Annals of Oncology
    The first-generation TRK inhibitors, larotrectinib and entrectinib, were granted landmark, tumour-agnostic regulatory approvals for the treatment of these ...Trk Inhibitors In Trk... · Trk Inhibitor Resistance · Trk Inhibitor SafetyMissing: history | Show results with:history<|separator|>
  144. [144]
    Selective type II TRK inhibitors overcome xDFG mutation mediated ...
    Nov 8, 2023 · Currently, two generations TRK inhibitors have been developed. The representative second-generation inhibitors selitrectinib and repotrectinib ...
  145. [145]
    TRK xDFG Mutations Trigger a Sensitivity Switch from Type I to II ...
    The second-generation TRK inhibitors selitrectinib (LOXO-195, a selective TRK ... classified as type I inhibitors and are canonical ATP competitors. In ...
  146. [146]
    BPI-28592 as a novel second generation inhibitor for NTRK fusion ...
    Sep 11, 2024 · The first-generation TRK inhibitors larotrectinib (LOXO-101, Vitrakvi) and entrectinib (Rozlytrek) have shown high response rates in NTRK ...
  147. [147]
    FDA grants accelerated approval to repotrectinib for solid tumors
    Jun 13, 2024 · FDA grants accelerated approval to repotrectinib for adult and pediatric patients with NTRK gene fusion-positive solid tumors. Resources for ...<|separator|>
  148. [148]
    Resistance to TRK inhibition mediated by convergent MAP kinase ...
    A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors.Missing: confusion | Show results with:confusion
  149. [149]
    TRK inhibitors in TRK fusion-positive cancers - Annals of Oncology
    Specifically, MET amplification,. BRAFV600E mutation or hotspot mutations involving KRAS have been shown to emerge in tumour and/or plasma samples from patients ...
  150. [150]
    Efficacy and safety of larotrectinib as first-line treatment for patients ...
    May 23, 2025 · The study included 101 adult and paediatric patients with treatment-naïve metastatic/unresectable TRK fusion cancer. •. The ORR was 77% (95% CI ...
  151. [151]
    Discontinuation and Resumption of Larotrectinib in Pediatric ...
    Feb 6, 2025 · The study included 91 patients (aged < 18 years) receiving larotrectinib in two clinical trials, with infantile fibrosarcoma in 49, other soft- ...
  152. [152]
    Repotrectinib Demonstrates Durable Efficacy in TKI-Naive and
    Sep 10, 2025 · Repotrectinib showed a cORR of 79% in TKI-naive and 41% in TKI-pretreated ROS1-positive NSCLC patients. · The median DOR was 36.8 months for TKI- ...
  153. [153]
    Characterization of On-Target Adverse Events Caused by TRK ...
    TRK inhibition-related AEs including weight gain, dizziness, and withdrawal pain occur in a substantial proportion of patients receiving TRK inhibitors.
  154. [154]
    NTRK gene fusion testing and management in lung cancer
    Weight gain, dizziness, paraesthesias and withdrawal pain could all be considered 'on-target' AEs secondary to the inhibition of TRKA/B/C that play post ...