Fact-checked by Grok 2 weeks ago

Low-affinity nerve growth factor receptor

The low-affinity nerve growth factor receptor (LNGFR), also known as p75 neurotrophin receptor (p75NTR) or CD271, is a transmembrane glycoprotein belonging to the tumor necrosis factor receptor superfamily that serves as a low-affinity binder for all mammalian neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and NT-4/5, while also interacting with their proneurotrophin precursors and modulating high-affinity signaling via Trk receptors. Encoded by the NGFR gene on chromosome 17q21.33, it consists of 427 amino acids, featuring an extracellular domain with four cysteine-rich repeats for ligand binding, a single transmembrane domain, and an intracellular death domain that facilitates interactions with signaling adaptors but lacks intrinsic enzymatic activity. This receptor plays a dual role in neuronal biology, promoting survival, axonal growth, and synaptic plasticity when co-expressed with Trk receptors, while independently driving apoptosis and growth cone collapse in contexts of injury or disease. Structurally, p75NTR exists in full-length and truncated isoforms, with the full-length form spanning six exons and enabling diverse signaling outcomes through associations with co-receptors like sortilin (for pro-apoptotic binding) or the Nogo-66 receptor (for inhibitory signaling). Its ligand-binding extracellular region exhibits low nanomolar affinity for mature (Kd ≈ 10-9 M) but higher affinity for when paired with sortilin, allowing it to fine-tune responses based on local ligand availability. Downstream, p75NTR activates pathways such as for cell survival, JNK/ for , and RhoA for cytoskeletal regulation, often in opposition or with Trk-mediated PI3K/Akt and MAPK survival signals. Expression of p75NTR is dynamically regulated, peaking during embryonic development in neurons, Schwann cells, and derivatives to support and , then declining in adulthood except in select populations like neurons and melanocytes. Post-injury, such as in peripheral nerve damage or trauma, its levels surge in reactive , immune cells, and regenerating axons, aiding remyelination and while potentially exacerbating degeneration if pro-apoptotic ligands predominate. Physiologically, it is essential for proper development, as NGFR knockout mice exhibit defects in sensory and sympathetic innervation, and it also functions beyond as a receptor for pathogens like . In pathology, p75NTR contributes to neurodegenerative conditions including , where it mediates amyloid-β-induced toxicity and tau hyperphosphorylation in neurons, and via motoneuron ; it also marks cancer stem cells in and influences pain signaling through NGF sensitization. Therapeutic strategies targeting p75NTR, such as small-molecule inhibitors like LM11A-31, have shown promise in preclinical models for , reducing and enhancing repair without disrupting Trk pathways.

Receptor family and nomenclature

Family classification

The low-affinity nerve growth factor receptor, also known as p75NTR, is classified within the superfamily, a group of type I transmembrane proteins characterized by their roles in regulating cell survival, proliferation, and apoptosis. Specifically, p75NTR belongs to the death receptor subfamily of this superfamily, distinguished by the presence of an intracellular death domain (DD) that facilitates pro-apoptotic signaling upon binding. This DD, a approximately 90-amino-acid module, shares structural and functional with those found in other death receptors, enabling interactions with adaptor proteins like TRADD to initiate activation cascades. Like prototypical TNFR family members such as TNFR1 (also known as p55 or CD120a) and (CD95), p75NTR features an extracellular region composed of four cysteine-rich domains (CRDs) that mediate ligand binding through disulfide-bonded modules. These CRDs exhibit sequence and structural conservation with those in TNFR1, allowing trimerization upon ligand engagement, yet p75NTR is unique in its specificity for neurotrophins—such as (NGF), (BDNF), and (NT-3)—rather than cytokines like TNF-α that bind other TNFRs. This neurotrophin selectivity arises from adaptations in the CRD architecture, setting p75NTR apart while preserving the superfamily's core signaling motifs. p75NTR demonstrates high evolutionary across mammals, with orthologs in humans and (e.g., and ) sharing over 90% sequence identity, particularly in the transmembrane and intracellular domains critical for signaling. This conservation underscores its fundamental role in developmental processes and suggests minimal functional divergence since the divergence of mammalian lineages. In contrast to the high-affinity receptors of the Trk family (TrkA, TrkB, TrkC), which are receptor with intracellular kinase domains that autophosphorylate upon binding, p75NTR lacks enzymatic activity and relies on adaptor-mediated pathways, highlighting its distinct mechanistic class despite overlapping ligand recognition.

Historical naming and discovery

The low-affinity nerve growth factor receptor was first characterized in 1979 through binding studies demonstrating two distinct classes of NGF binding sites on chick embryo sensory ganglia cells. Subsequent studies in 1980 on PC12 cells further identified it as a 75 kDa protein exhibiting low-affinity binding to (NGF). These early experiments demonstrated two classes of NGF binding sites on PC12 cells, with the low-affinity site corresponding to the 75 kDa receptor, distinguishing it from higher-affinity sites observed in neuronal populations. The receptor was molecularly cloned in 1986 by the laboratory led by Moses Chao and David Johnson, who isolated the human cDNA from a cell library and confirmed its expression as a 75 kDa transmembrane capable of binding NGF with low affinity (Kd ≈ 10-9 M). This effort provided the first sequence information, revealing structural features such as cysteine-rich extracellular domains and establishing it as the primary low-affinity NGF receptor. Initially termed the "low-affinity nerve growth factor receptor" (LNGFR) to reflect its specific association with NGF, the nomenclature evolved in 1990 to p75 neurotrophin receptor (p75NTR) after studies showed it binds additional neurotrophins, including (BDNF), underscoring its role as a pan-neurotrophin receptor. This redesignation highlighted its membership in the tumor necrosis factor receptor (TNFR) superfamily and its broader ligand specificity beyond NGF. Key milestones in p75NTR research include its linkage to apoptosis in the 1990s, with seminal work demonstrating that p75NTR activation in the absence of Trk co-receptors promotes neuronal cell death via pathways involving caspases and JNK signaling. In the 2000s, the receptor's role expanded to include high-affinity binding to proneurotrophins (e.g., proNGF and proBDNF), which trigger pro-apoptotic signaling through sortilin co-receptors, contrasting with survival-promoting effects of mature neurotrophins. Alternative names for p75NTR include NGFR (nerve growth factor receptor), p75NGFR, and CD271 (cluster of differentiation 271), the latter assigned based on its expression on neural crest-derived cells.

Molecular structure

Domain architecture

The low-affinity nerve growth factor receptor, also known as p75NTR, is a type I belonging to the receptor (TNFR) superfamily, characterized by a modular domain architecture that facilitates recognition and intracellular signaling. The extracellular region comprises approximately 230 organized into four tandem cysteine-rich domains (CRDs), designated CRD1 through CRD4, each featuring six conserved residues that form three intramolecular bonds to stabilize the structure. Among these, CRD2 and CRD3 are particularly critical for interactions with , forming the primary binding interface while CRD1 and CRD4 contribute to overall specificity and receptor conformation. Crystal structures of the extracellular domain, such as the 2.4 Å resolution complex with (PDB: 1SG1) and the 2.6 Å resolution complex with (PDB: 3BUK), reveal an elongated, asymmetric arrangement of these CRDs that supports dimeric binding. Spanning the plasma membrane is a single α-helical transmembrane domain of about 21 amino acids, which promotes receptor dimerization through hydrophobic interactions and a conserved cysteine residue that can form intermolecular disulfide bonds.35610-6/fulltext) This domain links the extracellular and intracellular regions, enabling signal transmission across the membrane. The intracellular region consists of a 155-amino acid tail that includes a juxtamembrane segment, a chopper domain, and a C-terminal death domain (DD) of approximately 80 residues (residues 339–417 in human p75NTR), which adopts a six-α-helical bundle fold to recruit signaling adaptors. NMR and crystal structures of the DD (e.g., PDB: 4F42) highlight its propensity for both symmetric disulfide-linked dimers and asymmetric non-covalent oligomers via interfaces involving hydrophobic residues. The core polypeptide chain of p75NTR has a calculated molecular weight of 45 kDa, but extensive N- and in the extracellular region increases the apparent size to approximately 75 kDa in its mature form. Dimerization is further stabilized by interfaces in both the transmembrane and intracellular domains, influencing receptor activation states.35610-6/fulltext)

Post-translational modifications and oligomerization

The low-affinity nerve growth factor receptor, also known as p75NTR, undergoes N-linked glycosylation at asparagine 60 within the first cysteine-rich domain (CRD1) of its extracellular region. This modification introduces complex glycoforms, such as fucosylated paucomannosidic structures, which maintain the receptor in a monomeric state in solution and contribute to proper complex formation with ligands. Glycosylation at this site enhances ligand binding affinity, with the glycosylated form exhibiting a dissociation constant (Kd) of approximately 2.1 nM for nerve growth factor (NGF), compared to a roughly 100-fold lower affinity in unglycosylated variants. Additionally, O-linked glycosylation occurs at multiple sites in the juxtamembrane stalk domain, influencing receptor trafficking and apical sorting in polarized cells by facilitating association with specific membrane compartments.95442-4) Palmitoylation of p75NTR occurs post-translationally via linkage at 279 in the juxtamembrane region. This modification enhances receptor stability by promoting its partitioning into lipid rafts and modulating interactions with the membrane environment. Substitution of 279 with abolishes palmitoylation, confirming its specificity and role in anchoring the receptor to the plasma membrane, which indirectly supports dimer stability. Oligomerization of p75NTR is constitutive, with the receptor primarily forming homodimers through its , facilitated by a GxxxG-like (AXXXG, residues 262-266). This motif enables helix-helix interactions that stabilize dimers independently of extracellular or intracellular domains, as evidenced by energy transfer () and cross-linking studies showing surface dimerization. A transmembrane at position 257 further promotes covalent dimerization via bonds, enhancing overall assembly. binding induces higher-order clusters, including trimers, which coexist with monomers and dimers at the cell surface to regulate receptor dynamics. Ubiquitination targets p75NTR for and lysosomal , primarily through residues in the intracellular . The E3 ligase TRAF6 mediates K63-linked polyubiquitination at 247, 280, and 283, promoting receptor and subsequent in a ligand-dependent manner. Similarly, c-Cbl acts as an ligase following of the receptor, directing ubiquitination and turnover to control surface levels. Phosphorylation of serine and threonine residues in the intracellular tail modulates adaptor protein binding for p75NTR. () targets serine 304, influencing receptor localization and interactions with downstream effectors. The serine/threonine-rich C-terminal region, including sites like serine 308, undergoes that recruits adaptors such as RIP2 upon ligand stimulation. These modifications alter binding affinity for intracellular partners without intrinsic kinase activity in p75NTR itself.

Ligands and binding properties

Interactions with mature neurotrophins

The low-affinity nerve growth factor receptor, also known as p75NTR, binds all four mature (NGF), (BDNF), (NT-3), and neurotrophin-4 (NT-4)—with characteristically low affinity. These interactions are mediated by dissociation constants (Kd) in the nanomolar range, approximately 10-9 M, as exemplified by Kd values of 2.1 × 10-9 M for NGF and 8 × 10-10 M for NT-3. In contrast, the high-affinity bind their preferred mature neurotrophins with picomolar affinities, around 10-11 M, such as 1.8 × 10-11 M for TrkC with NT-3. The primary binding interface for mature resides in the cysteine-rich domains 2 through 4 (CRD2-4) of the p75NTR extracellular domain, where these domains form the key contact surfaces for engagement. The resulting complex adopts a 2:2 , with a p75NTR dimer binding a dimeric molecule, as confirmed by structural analyses of the p75NTR-NGF complex. This dimeric arrangement supports efficient capture and receptor clustering on the cell surface. Co-expression of p75NTR with enhances the overall affinity and specificity of mature binding, forming ternary complexes such as NGF-p75NTR-, which accelerates association rates and promotes pro-survival signaling. For instance, p75NTR increases 's selectivity for NGF while reducing non-specific binding to other like NT-3. Beyond binding, p75NTR facilitates the internalization of mature into signaling endosomes and their axonal , as demonstrated by p75NTR-mediated uptake and trafficking of BDNF in neurons.

Interactions with proneurotrophins

The low-affinity nerve growth factor receptor (p75NTR) binds with high affinity to , including proNGF, proBDNF, and proNT-3, exhibiting dissociation constants (Kd) approximately 10-10 M when co-expressed with the sortilin co-receptor, which markedly enhances binding compared to mature . This interaction occurs primarily through the cysteine-rich domain 1 (CRD1) of p75NTR's extracellular region, where the pro-domains of expose specific epitopes absent in their cleaved, mature counterparts, enabling selective recognition and dimerization for receptor engagement. The structural basis involves a symmetric between proNGF and p75NTR, stabilized by calcium ions and sortilin, which positions the to trigger intracellular signaling. Proneurotrophins represent the biosynthetic precursors of , synthesized as pre-pro forms and cleaved intracellularly or extracellularly by or proconvertases; however, uncleaved proneurotrophins remain dimeric and serve as potent ligands for p75NTR, driving pro-apoptotic cascades rather than signals associated with forms. This binding induces rapid recruitment of sortilin, forming a ternary complex that activates downstream effectors like JNK and , culminating in neuronal . In cellular models, such as sympathetic neurons or hippocampal cultures, proNGF and proBDNF at subnanomolar concentrations elicit specifically via this p75NTR-sortilin pathway, underscoring the mechanistic distinction from low-affinity interactions. p75NTR expression is particularly elevated in the developing , where proneurotrophins play a critical role in to refine neural connectivity and eliminate excess neurons during circuit sculpting. For instance, in the and , high levels of p75NTR coincide with proneurotrophin activity, promoting in target-dependent neurons post-innervation. A 2023 review in the affirms proneurotrophins as the predominant ligands for p75NTR in the adult brain, especially in regions affected by neurodegeneration, where they contribute to ongoing apoptotic regulation and pathological cell loss. This tissue-specific dynamic highlights p75NTR's evolutionarily conserved function in balancing survival and death during neural maturation and .

Biological functions

Role in neuronal development and survival

The low-affinity nerve growth factor receptor, p75NTR, exhibits dynamic expression patterns during neuronal development, peaking in the embryonic (CNS) and (PNS) before declining postnatally. In the developing CNS and PNS, p75NTR is highly expressed in neurons and glial cells from embryonic stages through early postnatal periods, coinciding with phases of axon growth, target innervation, and . This expression diminishes in adulthood, with low levels persisting in select regions such as the and , reflecting its transition from a developmental to a more restricted maintenance role. p75NTR promotes the survival of sensory neurons during embryogenesis by forming a complex with (NGF) and the high-affinity receptor TrkA, which enhances NGF binding affinity and sensitizes neurons to low concentrations of the ligand. This trimeric NGF-TrkA-p75NTR complex facilitates pro-survival signaling in developing sensory and sympathetic neurons, accelerating outgrowth and preventing in response to limited NGF availability in target tissues. Studies on embryonic sensory ganglia demonstrate that co-expression of p75NTR with TrkA increases neuronal responsiveness to NGF, ensuring appropriate survival rates during early innervation phases. p75NTR contributes to axonal branching and target innervation in sensory systems, as evidenced by deficits in p75NTR knockout mice, which exhibit reduced outgrowth, impaired branching, and sensory neuron loss. In these mutants, spinal and trigeminal sensory nerves show diminished elongation and collateral formation, leading to incomplete target innervation and up to 50% fewer neurons in affected ganglia. For instance, gustatory axons in the display disrupted branching patterns, resulting in fewer and sensory deficits, underscoring p75NTR's role in refining peripheral projections during development. In the retina, p75NTR participates in naturally occurring cell death to balance neuron numbers during development, particularly influencing retinal ganglion cell (RGC) populations in rat models. Seminal studies on postnatal rat retinas reveal that p75NTR expression in RGCs and Müller glia modulates apoptosis, with blockade or absence leading to altered RGC survival rates and disrupted laminar organization. This process ensures precise neuron matching to target demands, preventing over-innervation in the optic nerve and superior colliculus. p75NTR aids in refining topographic maps in the visual and somatosensory systems by supporting axon guidance and repulsion mechanisms essential for precise connectivity. In the visual system, p75NTR mediates ephrin-A reverse signaling in cortical interneurons and subplate neurons, promoting axon repulsion and map alignment in the superior colliculus and visual cortex during late embryonic and early postnatal refinement. Similarly, in somatosensory pathways, p75NTR influences barrel cortex map formation through modulated growth cone dynamics, with knockouts showing coarse projections and delayed topographic organization.

Role in non-neuronal development and tissue

The low-affinity nerve growth factor receptor, p75NTR, plays a pivotal role in regulating osteoblast and maintaining mass beyond its neuronal functions. Studies have shown that p75NTR influences skeletal by modulating the of mesenchymal precursors into osteoblasts, thereby supporting trabecular formation. In p75NTR mice, postnatal skeletal development is impaired, resulting in overall growth retardation and significantly reduced trabecular volume, as evidenced by microcomputed tomography analyses revealing decreased bone mineral density and altered architecture. These findings underscore p75NTR's essential function in NGF-responsive JNK signaling pathways that drive osteogenic processes during skeletal maturation. p75NTR is also critical for postnatal craniofacial , where it facilitates and formation through NGF signaling. Disruption of p75NTR in models leads to craniofacial dysmorphology, including shortened snouts and reduced thickness, due to impaired and of cranial neural crest-derived mesenchymal cells. Specifically, p75NTR deficiency results in decreased expression of osteogenic markers such as and Osterix in craniofacial tissues, highlighting its role in coordinating mesenchymal condensation and during early postnatal stages. These effects are mediated by p75NTR's interaction with proneurotrophins, which briefly promote pro-differentiation signals in non-neuronal ectomesenchymal cells. In epithelial-mesenchymal interactions, p75NTR maintains tissue integrity in and by regulating cyclic remodeling processes. During morphogenesis and cycling, p75NTR expression in dermal papilla cells and modulates apoptosis-driven , ensuring balanced epithelial-mesenchymal signaling for follicle regeneration. Loss of p75NTR function disrupts these interactions, leading to abnormal follicle and altered progression, as observed in models where follicular downgrowth is accelerated. p75NTR contributes to immune cell homeostasis by modulating survival and activation, particularly in subsets. Activation of p75NTR on plasmacytoid dendritic cells and B lymphocytes limits the expansion of immature CD21^lo^ B cell populations, thereby preserving immune balance during inflammatory responses. In p75NTR-deficient models, enhanced survival of IgM memory B cells disrupts maturation processes, indicating p75NTR's regulatory role in selection and preventing aberrant immune activation. Expression of p75NTR extends to various non-neuronal tissues, including and salivary glands, where it supports maintenance and secretory functions. In , p75NTR is present on mesenchymal stem cells, influencing their potential in hematopoietic niches. Similarly, in salivary glands, p75NTR expression correlates with acinar and ductal cell , potentially aiding in tissue repair and fluid secretion regulation.

Receptor interactions

Co-receptor complexes with sortilin

Sortilin, encoded by the SORT1 gene, functions as a type I transmembrane co-receptor that specifically binds the pro-domains of , such as proNGF and proBDNF, to facilitate their interaction with p75NTR. This binding is calcium-dependent and occurs through sortilin's luminal Vps10p , which recognizes the positively charged pro-peptides with higher (Kd ≈ 770 nM for proNGF) compared to mature . The p75NTR-sortilin complex forms a ternary assembly with proneurotrophins in a dimeric , where the mature domain of proNGF engages p75NTR while the pro-domain interacts with sortilin, resulting in a high-affinity complex (Kd ≈ 140 nM). This ternary complex potently induces in neurons by promoting pro-apoptotic signaling, distinct from the survival-promoting actions of mature . Structural studies from the , including a 3.75 Å of the proNGF-p75NTR complex revealing a 2:2 symmetric binding mode, indicate that sortilin's Vps10p domain interacts with the cysteine-rich domain (CRD) of p75NTR, stabilizing the assembly through a ten-helix interface and charge-balanced contacts in the receptor stalk region. In the , p75NTR and sortilin are co-expressed in neuronal populations of the and , where the complex contributes to activity-dependent via pro-apoptotic mechanisms during development and in response to injury. Mutations in SORT1, such as the K302E variant identified in (AD) patients, impair sortilin function and ligand binding in the Vps10p domain, leading to dysregulated p75NTR signaling and exacerbated neuronal degeneration in AD models.

Crosstalk with Trk receptors

The low-affinity receptor (p75NTR) modulates signaling through high-affinity tropomyosin receptor kinase () receptors, particularly TrkA and TrkB, by forming heterocomplexes that enhance neuronal responsiveness to . This crosstalk increases the binding affinity of TrkA for (NGF) and TrkB for (BDNF) by 10- to 100-fold, primarily through allosteric enhancement mediated by the intracellular domain of p75NTR. The association rate for ligand binding is accelerated up to 25-fold in co-expressing cells, allowing detection of low concentrations that would otherwise be insufficient for Trk activation alone. Physical interactions between p75NTR and occur via their transmembrane domains and have been confirmed through co-immunoprecipitation and cross-linking studies, enabling reciprocal regulation of downstream pathways. In synaptic contexts, p75NTR facilitates the recruitment of to trans-synaptic clusters, promoting localized signaling that supports assembly and maintenance.00556-8) This physical association is crucial for efficient transduction across synapses, where p75NTR acts as a scaffold to position for optimal encounter. The effects of this crosstalk vary by form: mature like NGF and BDNF, when bound to p75NTR-Trk complexes, amplify pro-survival signals such as PI3K-Akt pathway activation, enhancing neuronal differentiation and growth. In contrast, proneurotrophins compete for p75NTR binding, inhibiting Trk phosphorylation and shifting toward inhibitory outcomes, though this modulation fine-tunes Trk specificity without fully blocking survival under physiological conditions. During development, p75NTR-Trk crosstalk is vital for sympathetic survival, particularly in target tissues with limited availability. Neurons from p75NTR null mice display hyporesponsiveness to Trk ligands, with reduced TrkA activation and downstream signaling (e.g., ERK and Akt ) at low NGF concentrations, leading to impaired neurite outgrowth and increased neuronal loss. Crosses with TrkA null mice further reveal that p75NTR absence rescues developmental sympathetic death by ~80% in early postnatal stages, underscoring its role in balancing Trk-dependent survival against basal pro-apoptotic tones. Beyond development, this interplay extends to , where p75NTR-TrkA complexes in dorsal root ganglia contribute to pain sensitization by heightening excitability in response to inflammatory cues.

Binding to Nogo-66 receptor (NgR1)

The low-affinity nerve growth factor receptor, known as p75NTR, functions as a co-receptor for the Nogo-66 receptor 1 (NgR1), a glycosylphosphatidylinositol-anchored protein lacking intracellular signaling domains, to transduce inhibitory signals from myelin-associated molecules that restrict axonal growth. This association enables the complex to bind and signal in response to Nogo-66, myelin-associated glycoprotein (MAG), and myelin glycoprotein (OMgp), thereby promoting collapse and limiting neurite extension in neurons encountering debris. Within the NgR1-p75NTR complex, the death domain (DD) of p75NTR recruits intracellular signaling effectors, which is critical for downstream activation of RhoA and the consequent cytoskeletal rearrangements that inhibit axonal outgrowth (detailed further in the section on cytoskeletal regulation via RhoA). assembly typically involves additional partners such as LINGO-1 to facilitate from the extracellular ligands to intracellular pathways. In the context of central nervous system injury, the p75NTR-NgR1 complex mediates myelin-induced inhibition of neurite outgrowth, contributing to the failure of axonal regeneration; experimental blockade of this , including through p75NTR knockout or interference with complex formation, enhances axonal regrowth in models. p75NTR exhibits high expression levels in and is upregulated in neurons following injury, positioning it to amplify the inhibitory effects of myelin-associated signals in the damaged environment. Studies from the 2020s have further elucidated the p75NTR-NgR1 complex's involvement in (RGC) inhibition after injury, where myelin-derived ligands via this pathway hinder regeneration, and targeted disruptions promote RGC and regrowth.

Signaling pathways

Pro- signaling via

The low-affinity nerve growth factor receptor (p75NTR), upon binding mature such as (NGF), initiates pro-survival signaling primarily through the nuclear factor kappa B () pathway in developing neurons. This pathway is activated when ligand binding induces oligomerization of p75NTR, facilitating the recruitment of tumor necrosis factor receptor-associated factor 6 (TRAF6) to the intracellular death domain (DD) of the receptor. TRAF6 then interacts with downstream kinases, including transforming growth factor beta-activated kinase 1 (TAK1) and the (IKK) complex, leading to phosphorylation and degradation of the inhibitory protein IκB, which releases the heterodimer (typically p65/ and p50) for nuclear translocation. Once in the nucleus, binds to specific promoter regions to drive transcription of target genes that promote cell survival and . Key transcriptional targets of this NF-κB activation include anti-apoptotic factors such as B-cell lymphoma 2 (Bcl-2), which inhibits mitochondrial outer membrane permeabilization, and superoxide dismutase 2 (SOD2), a mitochondrial that mitigates oxidative damage in neurons. These targets collectively enhance neuronal resilience against stressors like and trophic factor deprivation, underscoring the pathway's role in maintaining cellular during development. Experimental studies have demonstrated the protective effects of this signaling axis; for instance, overexpression of p75NTR in cultured neurons increases activity and confers resistance to oxidative stress-induced , as evidenced by reduced in response to exposure. Similarly, in models of amyloid-β toxicity, p75NTR-mediated activation via TRAF6 recruitment suppresses neuronal death, highlighting its relevance in neuroprotective contexts. The pathway can be summarized as: p75NTR → TRAF6 → TAK1/IKK → (p65/p50) → gene expression (e.g., , ).

Pro-apoptotic signaling via JNK and caspases

The pro-apoptotic signaling of the low-affinity nerve growth factor receptor (p75NTR), also known as p75 receptor, is prominently mediated by the c-Jun N-terminal (JNK) pathway and subsequent activation, particularly in response to binding of proneurotrophins such as proNGF and proBDNF. These ligands preferentially engage p75NTR to induce in contexts like target-deprived neurons, where mature are absent or insufficient to counter the death signal. This pathway contrasts with pro-survival effects elicited by mature neurotrophins and is enhanced when p75NTR forms complexes with co-receptors like sortilin, which increases proneurotrophin affinity and signaling potency. Upon proneurotrophin binding, p75NTR oligomerizes, recruiting tumor necrosis factor receptor-associated factors (TRAFs), particularly TRAF6, to its intracellular domain; this interaction activates the JNK signaling cascade in a ligand-dependent manner. Activated JNK, predominantly the JNK3 isoform in neurons, then phosphorylates the BH3-only protein BimEL at serine 65, enhancing its pro-apoptotic function by promoting dissociation from anti-apoptotic members and translocation to mitochondria, where it activates Bax to induce release. Concurrently, JNK phosphorylates c-Jun, driving transcriptional upregulation of Bim via AP-1 sites in the Bim promoter, thereby amplifying the pool of pro-apoptotic Bim proteins. This JNK-BimEL axis is essential for p75NTR-induced neuronal death, as demonstrated by genetic knockdown of Bim or expression of a non-phosphorylatable BimEL (S65A), which significantly attenuates . Parallel to the JNK-mediated intrinsic pathway, the death domain (DD) of p75NTR facilitates apoptosis through caspase activation, primarily involving initiator caspase-9 leading to effector caspases such as caspase-3 in certain models. Although caspase-8 activation has been reported in some cellular contexts, it is not universal and the DD remains indispensable for this cascade, as its deletion abolishes apoptosis induction. Mature neurotrophins inhibit this pro-apoptotic axis by competing for p75NTR binding and diverting signaling toward survival pathways, underscoring the ligand-specificity of death induction. The role of p75NTR in was first established in the 1990s through studies showing its necessity for naturally occurring neuronal during , such as in sympathetic neurons deprived of target-derived NGF. More recent investigations, including 2023 analyses, have refined the specificity of proneurotrophins in activating this JNK-caspase axis, highlighting its relevance in injury-induced neurodegeneration and immune-mediated synaptic remodeling.

Cytoskeletal regulation via RhoA

The low-affinity nerve growth factor receptor (p75NTR), in complex with the Nogo-66 receptor (NgR1), facilitates the activation of in response to myelin-associated inhibitory ligands such as Nogo-A, myelin-associated glycoprotein (MAG), and oligodendrocyte myelin glycoprotein (OMgp). This complex formation leads to the release of Rho-guanosine diphosphate dissociation inhibitor (RhoGDI) from , allowing the to adopt its active GTP-bound state, which is essential for transducing repulsive signals in neuronal processes. Additionally, downstream effectors like ROCK1 and ROCK2 are recruited following activation, amplifying the inhibitory cascade on axonal extension. Activated RhoA-GTP propagates signaling through multiple pathways that regulate cytoskeletal dynamics, primarily via effectors such as and mammalian diaphanous (mDia). phosphorylates myosin light chain and inhibits myosin light chain , promoting actomyosin contractility, while mDia induces actin polymerization; together, these actions drive the contraction and disassembly of the cytoskeleton in , resulting in their and inhibition of neurite outgrowth. A parallel branch involves RhoA-mediated activation of LIM kinase (LIMK), which phosphorylates , preventing its severing activity on filamentous actin (F-actin) and thereby stabilizing the cytoskeleton in a rigid, non-dynamic state that halts forward protrusion. This detailed pathway—p75NTR engagement leading to RhoGDI displacement, RhoA-GTP formation, and subsequent LIMK/ phosphorylation—underlies the precise control of F-actin turnover essential for remodeling. In the context of axon guidance, the p75NTR-RhoA axis mediates repulsive cues, notably through semaphorin signaling, where plexin receptors cooperate with p75NTR to activate RhoA and induce growth cone collapse, ensuring accurate pathfinding during neural development. Pharmacological inhibition of this pathway, such as with C3 transferase (C3 toxin), which ADP-ribosylates and inactivates RhoA, blocks p75NTR-dependent growth cone collapse in models of axonal regeneration, highlighting its therapeutic potential for overcoming inhibitory environments in the .

Role in diseases

Neurodegenerative disorders

In (AD), the low-affinity nerve growth factor receptor (p75NTR) is upregulated in neurons of the , contributing to their selective degeneration. This upregulation, coupled with increased levels of proNGF—a proneurotrophin ligand that binds with high affinity to p75NTR—shifts signaling toward pro-apoptotic pathways, promoting retrograde axonal degeneration and loss of these neurons critical for and . Recent clinical evidence from a phase 2a trial demonstrates that modulating p75NTR with the small-molecule ligand LM11A-31 reduces markers of and neurodegeneration in patients with mild to moderate AD, suggesting therapeutic potential in attenuating disease progression. In (), p75NTR expression is elevated in vulnerable s, where it promotes through JNK-mediated signaling, particularly in models harboring mutations. This receptor facilitates retrograde degenerative signals in SOD1G93A mouse models, exacerbating death triggered by mutant SOD1-linked oxidative stress and imbalances. Blocking p75NTR signaling has been shown to delay loss in these models, highlighting its role in ALS pathogenesis. In (HD), p75NTR levels are elevated in the of mutant -expressing models, such as HdhQ111 mice, where signaling is dysregulated by effects of mutant to exacerbate neuronal toxicity. This elevation disrupts the balance between pro-survival and pro-apoptotic pathways, including altered activation, contributing to striatal vulnerability and synaptic dysfunction. Small-molecule modulation of p75NTR normalizes these signaling imbalances and reduces mutant -induced pathology in preclinical HD models. A common theme across these neurodegenerative disorders is the accumulation of proneurotrophins, such as proNGF and proBDNF, which preferentially bind p75NTR in the absence of sufficient co-activation, driving a shift from pro-survival to pro-apoptotic signaling and accelerating neuronal loss. This dysregulation is particularly pronounced in protein-misfolding diseases like , ALS, and HD, where proneurotrophin buildup correlates with p75NTR-mediated JNK and caspase activation. Therapeutic strategies targeting p75NTR in AD models, including blockade of proNGF signaling, have demonstrated by preventing proNGF-induced cholinergic degeneration and tau hyperphosphorylation in preclinical settings. These approaches, often combined with small-molecule modulators, underscore p75NTR as a viable target for mitigating neurodegeneration without broadly disrupting .

Other neurological and developmental diseases

In (), the low-affinity nerve growth factor receptor (p75NTR) forms a complex with the Nogo-66 receptor (NgR1) that mediates inhibitory signaling from myelin-associated glycoproteins, thereby suppressing axonal regeneration and extension. This interaction activates downstream RhoA pathways, which collapse cytoskeletons and limit functional recovery post-injury. Administration of p75NTR antagonists, such as recombinant p75NTR-ED-Fc fusion proteins, blocks this inhibition, enhancing axonal sprouting, remyelination, and locomotor improvements in rodent models. Similarly, small-molecule inhibitors like EVT901 disrupt p75NTR oligomerization, reducing glial scarring and promoting behavioral recovery without affecting regenerative sprouting directly. In pain disorders, p75NTR expressed in dorsal root ganglia (DRG) neurons sensitizes nociceptors through crosstalk with (BDNF) and its high-affinity receptor TrkB, amplifying neuronal excitability and contributing to chronic . This sensitization involves p75NTR-mediated facilitation of BDNF-TrkB signaling, which upregulates ion channels like and promotes following peripheral . Pharmacological suppression of p75NTR, using antisense or blocking antibodies, attenuates TrkB and reverses mechanical , thermal , and cold in injury models. Mutations or deficiencies in p75NTR have been linked to developmental syndromes involving craniofacial dysmorphisms, as demonstrated in 2025 studies using models. p75NTR−/− mice exhibit significantly reduced postnatal skull length at day 7, with 15–20% decreases in calvarial bone volumes and trabecular thickness by day 28, alongside shape alterations in nasal, parietal, and occipital bones. These morphological changes, including a shortened cranium and tapered nasal structure, arise from impaired proliferation and regulation, suggesting p75NTR mutations contribute to congenital craniofacial malformations in humans. In retinal degeneration, p75NTR plays a critical role in promoting photoreceptor , particularly through binding pro-nerve growth factor (proNGF) released by activated , which exacerbates cell loss during degenerative processes. Elevated proNGF levels in models like rd10 and RhoP23H mice correlate with increased p75NTR signaling, inducing TNFα secretion and neurotoxic that drive outer nuclear layer thinning. Treatment with p75NTR antagonists, such as THX-B, reduces TUNEL-positive photoreceptor death by up to 60% in retinal explants and preserves layer thickness by mitigating microglial activation and release. Schizophrenia models reveal altered p75NTR signaling in the , where upregulated receptor expression during disrupts parvalbumin-positive maturation and connectivity. This dysregulation promotes pro-apoptotic pathways and , reducing oligodendrocyte density in layer 5 and impairing integrity, which correlates with cognitive deficits like reduced flexibility. In preclinical models, p75NTR restores prefrontal activity patterns and behavioral outcomes, highlighting its role in schizophrenia-like neuropathology.

Role in cancer

Expression and function in cancer stem cells

The low-affinity nerve growth factor receptor (p75NTR, also known as CD271) is upregulated in cancer stem cells (CSCs) across multiple tumor types, including , , and , where it identifies a subpopulation of tumor-initiating cells with enhanced stem-like properties. In , CD271+ cells exhibit self-renewal capacity and tumor propagation potential, though their expression can be unstable in patient-derived samples. Similarly, in , p75NTR marks invasive CSCs that contribute to treatment resistance, while in , its overexpression supports CSC survival under stress conditions. p75NTR promotes CSC self-renewal by upregulating pluripotency factors such as , Nanog, and , while inhibiting differentiation through downstream activation of signaling. This mechanism maintains stemness and enables tumor propagation, as seen in and hypopharyngeal CSCs where p75NTR defines populations capable of serial sphere formation and in vivo tumorigenesis. In CSCs, p75NTR exerts a pro-survival role via an autocrine loop involving (NGF), which stimulates receptor cleavage and enhances invasion, proliferation, and resistance to therapies independently of . This loop is particularly active in p75NTR-expressing cells, promoting aggressive behavior without additional exogenous NGF. Studies from the 2020s demonstrate that p75NTR knockdown impairs CSC sphere formation and reduces chemoresistance in models of hypopharyngeal, , and squamous cell carcinomas, highlighting its functional necessity for stem-like maintenance. For instance, silencing CD271 decreases spheroid proliferation and sensitizes cells to DNA-damaging agents by altering repair pathways. In , p75NTR expression correlates with favorable prognosis, particularly when co-expressed with TrkA, as it enhances differentiation and reduces tumor aggressiveness.

Therapeutic targeting in

The low-affinity nerve growth factor receptor, p75NTR, exhibits context-dependent roles in cancer, acting as either a tumor suppressor or promoter, which influences therapeutic strategies aimed at its modulation in . In cancers where p75NTR drives progression, such as , , and (RCC), targeting its signaling or processing has shown promise in preclinical models to inhibit tumor , , and . Conversely, in tumors like gastric cancer where it suppresses growth, strategies to enhance p75NTR activity are under exploration, though inhibition remains the predominant focus due to its pro-oncogenic effects in many solid tumors. One key therapeutic approach involves inhibiting the proteolytic processing of p75NTR, particularly its intramembrane cleavage by gamma-secretase, which generates intracellular domains that promote invasion. In multiforme (GBM), p75NTR expression correlates with tumor aggressiveness, and gamma-secretase inhibitors like DAPT or Compound X significantly reduce invasion (p < 0.001) and localize tumor growth in intracranial mouse models, extending median survival from 28 to 42 days (p < 0.0001). This strategy targets brain tumor-initiating cells, where p75NTR processing is enriched, offering potential for treating invasive gliomas resistant to standard therapies. Small-molecule modulators of p75NTR's represent another emerging strategy, particularly for . The derivative NSC49652 binds to residues in the p75NTR transmembrane helix (e.g., Ile252, Pro253), inducing oligomerization and JNK-dependent in cell lines like A375, with an of 10 μM. In xenograft models, (200 mg/kg) reduced tumor volume by 60% and improved survival (median 36 vs. 28 days), highlighting its efficacy against p75NTR-driven proliferation and potential to overcome , such as to , mediated by the p75NTR carboxyl-terminal fragment. In RCC, p75NTR is overexpressed in clear cell tumors and correlates with higher Fuhrman grade (p = 0.001) and worse prognosis, promoting pro-BDNF-induced AKT/ERK activation for cell survival and . Blocking p75NTR with neutralizing antibodies or silencing decreases viability by 40-50% and in 786-O cells, positioning it as a target for antibody-based therapies to curb in advanced disease. Similarly, in , p75NTR contributes to chemoresistance; its inhibition sensitizes cells to , suggesting combination potential. Despite these advances, challenges persist due to p75NTR's dual functionality; for instance, forced expression suppresses tumor growth in xenografts, indicating that agonists might benefit suppressor contexts. Ongoing prioritizes selective inhibitors to exploit its pro-apoptotic pathways while minimizing off-target effects in neural tissues. As of November 2025, CD271-targeted near-infrared photoimmunotherapy has shown potential for local depletion of cancer stem cells in preclinical models.

References

  1. [1]
    The p75 neurotrophin receptor: at the crossroad of neural repair and ...
    The p75 neurotrophin receptor (p75 NTR) has emerged as a potential target for pharmacological control of neurotrophin activity.
  2. [2]
    Entry - *162010 - NERVE GROWTH FACTOR RECEPTOR; NGFR
    Nerve growth factor receptor (NGFR) is also referred to as p75(NTR) because of its molecular mass and its ability to bind at low affinity not only NGF.
  3. [3]
    The Nerve Growth Factor Receptor (NGFR/p75NTR) - PubMed Central
    The Nerve growth factor receptor (NGFR/p75 NTR) represents a low-affinity receptor for all known mammalians neurotrophins (ie, proNGF, NGF, BDNF, NT-3 e NT-4/5)
  4. [4]
    Low-Affinity Nerve Growth Factor Receptor - ScienceDirect.com
    A low-affinity nerve growth factor (NGF) receptor is a type of receptor that is induced in various neural cells in response to injuries, and its exact function ...
  5. [5]
    Implication of the neurotrophin receptor p75NTR in vascular diseases
    The p75 neurotrophin receptor (p75NTR) is a member of TNF-α receptor superfamily that bind all neurotrophins, mainly regulating their pro-apoptotic actions.Missing: classification | Show results with:classification
  6. [6]
    p75NTR: A study in contrasts | Cell Death & Differentiation - Nature
    May 15, 1998 · The p75NTR binds each of the neurotrophins with low nanomolar affinity whereas the three trk receptors show strong binding preferences for ...
  7. [7]
    p75 Neurotrophin Receptor Expression on Adult Human ...
    Feb 15, 1998 · The p75 neurotrophin receptor (p75NTR), also a member of the TNF-R superfamily, contains a death domain sequence in its cytoplasmic region, and ...
  8. [8]
    p75NTR Mediates Neurotrophin-Induced Apoptosis of Vascular ...
    ... p55 TNF receptor (TNF-R1) and FAS. Like the TNF and FAS receptors, p75NTR has a cysteine-rich extracellular domain and an intracellular domain that lacks ...Missing: comparison | Show results with:comparison
  9. [9]
    Structural Characterization of the p75 Neurotrophin Receptor
    Aug 7, 2025 · p75NTR like TNF-R1 and Fas-R contain an extracellular domain with four cysteine-rich domains (CRD) and a death domain (DD) in the ...
  10. [10]
    Structural basis of death domain signaling in the p75 neurotrophin ...
    Dec 8, 2015 · ... human forms of the respective proteins. Human and rat p75NTR DD share more than 90% sequence identity—including all functionally relevant ...
  11. [11]
    A Novel p75 Neurotrophin Receptor-Related Protein, NRH2 ...
    Mar 17, 2004 · The Ig-like domains of Trk receptors and the cysteine-rich repeats of p75NTR are involved in binding to the neurotrophins.Missing: TNFR1 | Show results with:TNFR1
  12. [12]
    Nerve growth factor receptors on PC12 cells - PubMed
    The pheochromocytoma PC12 cell possesses specific cell surface receptors that bind nerve growth factor (NGF) with two different affinities.Missing: 1977 | Show results with:1977
  13. [13]
    Gene ResultNGFR nerve growth factor receptor [ (human)] - NCBI
    Aug 19, 2025 · Here, the authors unveil nerve growth factor receptor (NGFR, p75NTR or CD271) as a novel p53 inactivator.
  14. [14]
    UniProt
    Insufficient relevant content. The provided URL (https://www.uniprot.org/uniprotkb/P08138/entry) only contains a confirmation prompt for real users and general website information (cookies, privacy notice, help), with no specific details on p75NTR domain architecture, extracellular domains, transmembrane, intracellular regions, lengths, molecular weight, or crystal structures.
  15. [15]
    Structure of nerve growth factor complexed with the ... - PubMed
    We have determined the 2.4 A crystal structure of the prototypic neurotrophin, nerve growth factor (NGF), complexed with the extracellular domain of p75.Missing: p75NTR Vilar
  16. [16]
    1SG1: Crystal Structure of the Receptor-Ligand Complex between ...
    Jun 1, 2004 · We have determined the 2.4 A crystal structure of the prototypic neurotrophin, nerve growth factor (NGF), complexed with the extracellular domain of p75.Missing: p75NTR | Show results with:p75NTR
  17. [17]
    3BUK: Crystal Structure of the Neurotrophin-3 and p75NTR ...
    Jul 15, 2008 · Here we report the 2.6-A resolution crystal structure of neurotrophin-3 (NT-3) complexed to the ectodomain of glycosylated p75(NTR).<|control11|><|separator|>
  18. [18]
    Revising the mechanism of p75NTR activation - Nature
    Aug 13, 2020 · The ICD of p75NTR contains the chopper domain (residues 277–308), linker region (309–338) and a death domain (DD) (residues 339–417). P75NTR ...Missing: architecture | Show results with:architecture
  19. [19]
    4F42: Neurotrophin p75NTR intracellular domain - RCSB PDB
    May 15, 2013 · Total Structure Weight: 11.03 kDa · Atom Count: 700 · Modeled Residue Count: 84 · Deposited Residue Count: 94 · Unique protein chains: 1.Missing: architecture | Show results with:architecture
  20. [20]
    Structural Characterization of the Self-Association of the Death ...
    The neurotrophin receptor p75NTR conveys multiple signals via its intracellular death domain. However, how the death domain is activated and interacts with ...Missing: classification | Show results with:classification<|control11|><|separator|>
  21. [21]
    NGFR Gene - GeneCards | TNR16 Protein
    Aliases for NGFR Gene · GeneCards Symbol: NGFR · Nerve Growth Factor Receptor · TNFRSF16 · P75NTR · CD271 · Tumor Necrosis Factor Receptor Superfamily Member 16 · Low- ...
  22. [22]
  23. [23]
  24. [24]
  25. [25]
  26. [26]
  27. [27]
    Binding of neurotrophin‐3 to its neuronal receptors and interactions ...
    Mar 1, 1992 · Neurotrophin‐3 (NT‐3) has low‐affinity (Kd = 8 × 10(−10) M), as well as high‐affinity receptors (Kd = 1.8 × 10(−11) M) on embryonic chick ...
  28. [28]
    A p75NTR Pivoting Paradigm Propels Perspicacity - ScienceDirect
    CRD2, CRD3, and CRD4 and the cleft between CRD1 and CRD2 contain the binding surfaces that allow p75NTR to bind neurotrophin. To date, a functional role has ...
  29. [29]
    High-affinity TrkA and p75 neurotrophin receptor complexes - PMC
    It is also widely accepted that p75 directly influences Trk in such a way that the latter receptor forms a high-affinity binding site for its cognate ligand, ...
  30. [30]
    Retrograde Transport of Neurotrophins from the Eye to the Brain in ...
    May 1, 1996 · Antibodies specific for chicken p75NTRwere kindly provided by Gisela Weskamp (ChEX antibody; Weskamp and Reichardt, 1991) and Hideaki Tanaka ( ...
  31. [31]
  32. [32]
  33. [33]
    Novel functions and implications for diseases of the nervous system
    Aug 10, 2025 · This review describes recent advances in identifying the molecular components involved in p75NTR signal transduction and suggests that p75NTR ...<|separator|>
  34. [34]
    NGF Binding to p75 Enhances the Sensitivity of Sensory and ...
    To clarify the role of the common neurotrophin receptor p75 in modulating the survival response of sensory and sympathetic neurons to NGF at different ...
  35. [35]
    Differential Modulation of Neuron Survival during Development by ...
    To correlate NGF responsiveness with receptor expression, we measured p75 and TrkA mRNA levels during sensory neuron development. We found that p75 and TrkA ...
  36. [36]
    p75 Is Important for Axon Growth and Schwann Cell Migration ...
    Oct 15, 2000 · (1993) p75-deficient trigeminal sensory neurons have an altered response to NGF but not to other neurotrophins. Neuron 11:565–574. OpenUrl ...
  37. [37]
    Neurotrophin Binding to the p75 Receptor Modulates Rho Activity ...
    These results indicate that p75 NTR modulates in a ligand-dependent fashion the activity of intracellular proteins known to regulate actin assembly.Results · Activation Of Rho By P75 · Experimental ProceduresMissing: redesignated | Show results with:redesignated
  38. [38]
    The neurotrophin receptor p75 regulates gustatory axon branching ...
    Jun 24, 2014 · Likewise, since innervation to the tongue was disrupted, p75 mice also had a loss of taste buds. In all cases with neurotrophin knockout mice ...
  39. [39]
    Study of the Role of the Low-Affinity Neurotrophin Receptor p75 in ...
    Study of the Role of the Low-Affinity Neurotrophin Receptor p75 in Naturally Occurring Cell Death During Development of the Rat Retina. Dev Neurosci. 2001;23 ...
  40. [40]
    Study of the Role of the Low-Affinity Neurotrophin Receptor p75 in ...
    Feb 27, 2002 · Study of the Role of the Low-Affinity Neurotrophin Receptor p75 in Naturally Occurring Cell Death during Development of the Rat Retina Available.
  41. [41]
    p75NTR mediates ephrin-A reverse signaling required for axon ...
    ... topographic map development, visual system development. Introduction. Axonal ... At neonatal stages, prior to map refinement, the projections labeled in ...
  42. [42]
    p75 Neurotrophin Receptor Activation Regulates the Timing of the ...
    During development, patterned neural activity instructs topographic map refinement. ... Interestingly, activation of p75NTR onto PV cells in adult visual ...
  43. [43]
    p75 NTR regulates postnatal skeletal development via NGF ...
    Jun 13, 2025 · p75NTR has emerged as a key regulator of skeletal development and bone homeostasis. To define its role, we characterized skeletal phenotypes in ...
  44. [44]
    p75NTR regulates postnatal skeletal development via NGF ...
    Jun 13, 2025 · p75NTR has emerged as a key regulator of skeletal development and bone homeostasis. To define its role, we characterized skeletal phenotypes ...
  45. [45]
    p75 neurotrophin receptor regulates craniofacial growth and ...
    Mar 17, 2025 · Recent studies by Wang et al. also showed that p75NTR knockout mice have reduced alveolar bone mass and less osteogenic differentiation of ...
  46. [46]
    p75 neurotrophin receptor regulates craniofacial growth and ...
    In this study, we investigate the role of p75 neurotrophin receptor (p75NTR) in craniofacial development by comparing wild-type (p75NTR+/+) mice ...Missing: Frontiers | Show results with:Frontiers
  47. [47]
    [PDF] p75 neurotrophin receptor regulates craniofacial growth ... - Frontiers
    Mar 18, 2025 · In this study, we investigate the role of p75 neurotrophin receptor (p75NTR) in craniofacial development by comparing wild-type (p75NTR+/+) mice ...
  48. [48]
    A role for p75 neurotrophin receptor in the control of apoptosis ...
    Oct 1, 2000 · In contrast, apoptotic cell death mediated via p75NTR signaling does not occur if p75NTR is coexpressed with high-affinity Trk receptors (35–39) ...
  49. [49]
    A Role for p75 Neurotrophin Receptor in the Control of Hair Follicle ...
    p75NTR plays an important role during HF morphogenesis, functioning as a receptor that negatively controls HF development.
  50. [50]
    [PDF] Expression and function of P75 neurotrophin receptor in the immune ...
    In vivo studies performed with p75NTR exon 4 knockout mice indicate a modulatory role in selection, survival and maturation process of IgM memory B cells and/or ...
  51. [51]
    Role of p75 neurotrophin receptor in stem cell biology
    The aim of this review is to overview what is known about p75NTR in stem cell biology. Based upon the well-known and diverse signaling functions of p75NTR ...Missing: history paper
  52. [52]
    NGFR protein expression summary - The Human Protein Atlas
    A summary of the overall protein expression profile across the analyzed normal tissues based on knowledge-based annotation, presented in the Tissue resource. " ...
  53. [53]
    Molecular and Structural Insight into proNGF Engagement of ...
    For proNGF/p75NTR complexes, 2:1 and 2:2 complexes would be 79.3 and 97.8 kDa, respectively, and we measure molecular species with a number-averaged molar mass ...
  54. [54]
    Mapping of the Interaction Site between Sortilin and the p75 ... - NIH
    These results further show that proBDNF and proNT3 bind with very high affinity to sortilin and p75NTR, similar to what we previously have reported for proNGF ( ...
  55. [55]
    Finding memo: versatile interactions of the VPS10p-Domain ...
    Nov 18, 2022 · Crystal structure of the ligand-free form of the Vps10 ectodomain of dimerized Sortilin at acidic pH. FEBS Lett. 2018;592:2647–57. Article ...Missing: CRD1 | Show results with:CRD1
  56. [56]
  57. [57]
  58. [58]
    TrkA Mediates Developmental Sympathetic Neuron Survival in Vivo ...
    Crosses of p75NTR-/- and TrkA-/- mice indicate that the coincident absence of p75NTR substantially rescues TrkA-/- sympathetic neurons from developmental death ...Missing: null hyporesponsiveness
  59. [59]
    NGF in Neuropathic Pain: Understanding Its Role and Therapeutic ...
    NGF interacts with its receptor TrkA and p75NTR to activate intracellular signaling pathways associated with nociception and the emergence of allodynia and ...Ngf In Neuropathic Pain... · 3. Neuropathic Pain: A... · 3.2. Role Of Ngf In...
  60. [60]
    A p75(NTR) and Nogo receptor complex mediates ... - PubMed
    We conclude that p75(NTR) is a co-receptor of NgR for MAG signaling and a potential therapeutic target for promoting nerve regeneration.
  61. [61]
  62. [62]
    The p75 receptor transduces the signal from myelin-associated ...
    Thus, p75NTR and GT1b may form a receptor complex for MAG to transmit the inhibitory signals in neurons. Keywords: p75; neurotrophin; myelin-associated ...
  63. [63]
    MAG, myelin and overcoming growth inhibition in the CNS - Frontiers
    But given that NgR1 is a GPI-linked cell surface protein, additional partners are needed to transmit an intracellular signal inhibiting axonal outgrowth.<|control11|><|separator|>
  64. [64]
    p75 Neurotrophin Receptor Expression on Adult Human ...
    In the current study, we have found expression of p75NTR on cultured adult CNS-derived human OLs but not on other glial cells. TrkA was not detected on these ...Missing: NgR1 | Show results with:NgR1
  65. [65]
    p75 Neurotrophin Receptor: A Double-Edged Sword in Pathology ...
    The low-affinity nerve growth factor receptor p75NTR is a major neurotrophin receptor involved in manifold and pleiotropic functions in the developing and ...
  66. [66]
    (PDF) Synaptic or Non-synaptic? Different Intercellular Interactions ...
    Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration ... p75(NTR) and Nogo receptor complex medi-. ates repulsive ...
  67. [67]
    p75NTR signals via NF-kB - Reactome
    The NF-kB pathway is an important pro-survival signalling pathway activated by mature NGF, but not BDNF or NT-3, through p75NTR. It is unclear whether TRKA ...Missing: TRAF6 IKK
  68. [68]
    A Prosurvival Function for the p75 Receptor Death Domain ...
    Aug 15, 2001 · Together with the above considerations, the role of TRAF6 may be to mediate both p75-induced NF-κB and JNK activation. Indeed, NGF gives an ...
  69. [69]
    Neurotrophin Signaling through the p75 Receptor Is Deficient in traf6
    Our results demonstrate that TRAF6 is required for p75-mediated activation of NFκB and JNK; moreover, this signal mediator is necessary for the receptor- ...
  70. [70]
    Immune activation of the p75 neurotrophin receptor - Frontiers
    Broadly the TNFR family can be divided into three groups; (1) death receptors (2) TRAF-motif containing receptors or (3) decoy receptors.Missing: classification | Show results with:classification
  71. [71]
    Nerve Growth Factor-Dependent Activation of NF-κB Contributes to ...
    Dec 15, 1998 · We also demonstrate that overexpression of NF-κB is sufficient to protect sympathetic neurons from apoptosis caused by NGF deprivation. These ...
  72. [72]
    Roles for NF-κB in nerve cell survival, plasticity, and disease - Nature
    Jan 6, 2006 · Signaling through the p75NTR has been alternatively linked to both pro-survival and pro-death functions. A possible explanation lies in the ...
  73. [73]
    p75-Mediated NF-κB Activation Enhances the Survival Response of ...
    NF-κB activation, promoted by p65/p50, enhances survival of sensory neurons to NGF. NGF also activates NF-κB, and p75-mediated pathway is involved.
  74. [74]
    TRAF6 and p62 inhibit amyloid β-induced neuronal death through ...
    Sep 24, 2012 · p75NTR increased NF-κB activity by recruiting TRAF6/p62, which thereby mediated cell survival. These findings indicate that TRAF6/p62 abrogated ...Traf6 And P62 Inhibit... · 3. Results And Discussion · 3.4. Traf6/p62 Potentiates...<|control11|><|separator|>
  75. [75]
    Understanding Proneurotrophin Actions: Recent Advances and ...
    At the molecular level, mature neurotrophins interact with two distinct receptors: p75NTR and Trk (Kaplan and Miller, 2000; Huang and Reichardt, 2003; Segal, ...
  76. [76]
    Characterization of the c-Jun N-Terminal Kinase-Bim EL Signaling ...
    Oct 6, 2004 · In addition, we demonstrated that BimEL and JNK-induced phosphorylation of BimEL at serine 65 are required for p75NTR-induced apoptosis.
  77. [77]
    p75 Neurotrophin Receptor-mediated Apoptosis in Sympathetic ...
    During the development of the sympathetic nervous system, the p75 neurotrophin receptor (p75NTR) has a dual function: promoting survival together with TrkA ...Missing: hyporesponsiveness | Show results with:hyporesponsiveness
  78. [78]
    Apoptosis Induced by p75NTR Overexpression Requires Jun ...
    Together, these results show that p75NTR induces apoptosis through an intrinsic death pathway that results in mitochondrial cytochrome c release and caspase 9 ...
  79. [79]
  80. [80]
    Characterization of a p75(NTR) apoptotic signaling pathway using a ...
    Sep 7, 2001 · We conclude that p75(NTR) signals apoptosis by similar mechanisms as other death receptors but uses different adaptors and apical caspases.Missing: RIPK1 FADD
  81. [81]
    The p75 Neurotrophin Receptor Mediates Neuronal Apoptosis and ...
    These data indicate that p75NTR can signal to mediate apoptosis, and that this mechanism is essential for naturally occurring sympathetic neuron death.
  82. [82]
    Deathtouch: The Long and Selective Reach of Proneurotrophin ...
    Oct 26, 2023 · They identified that cortical neurons near the injured site contained higher levels of p75NTR and showed evidence of apoptosis. Importantly, ...
  83. [83]
    Mechanisms of neuronal growth cone guidance - PubMed Central
    The unbound p75NTR receptor signals to RhoA inducing LIMK mediated phosphorylation of ADF/cofilin (pAC) to reduce filopodial length and number. This ...
  84. [84]
    Modulation of Semaphorin3A Activity by p75 Neurotrophin Receptor ...
    Sema3A's activity as a powerful negative regulator of axon growth (Togashi et al., 2006) and guidance has been well documented (Taniguchi et al., 1997; Ulupinar ...Missing: ependymin | Show results with:ependymin
  85. [85]
    Axon growth inhibition by RhoA/ROCK in the central nervous system
    RhoA/ROCK activation, in turn, activates downstream effectors, which regulate cytoskeletal reorganization such as growth cone collapse and neurite outgrowth ...
  86. [86]
    Role of Nerve Growth Factor Receptors in Cholinergic Basal ...
    Apr 1, 2005 · These studies have provided unique insights into the role that NGF receptor impairment plays in CBF neuronal dysfunction during the prodromal stages of AD.Ngf And The Survival Of... · Ngf Receptor Alterations In... · Pro-Ngf And Nucleus Basalis...
  87. [87]
    ProNGF elicits retrograde axonal degeneration of basal forebrain ...
    Sep 24, 2024 · To investigate whether p75NTR mediated the retrograde, proNGF-induced cell death in rat BFCNs, we pretreated the axons with a p75NTR ligand– ...
  88. [88]
    p75 neurotrophin receptor modulation in mild to moderate Alzheimer ...
    May 17, 2024 · ... neurotrophin receptor (p75NTR) ... The ADNI was launched in 2003 as a public–private partnership, clinical trial-like natural history study, led by ...
  89. [89]
    Mitochondrial Superoxide Production and Nuclear Factor Erythroid 2 ...
    Jul 18, 2007 · Induction of p75NTR renders motor neurons vulnerable to NGF-induced apoptosis. p75NTR has been implicated in motor neuron death occurring in ...
  90. [90]
    Retrograde apoptotic signaling by the p75 neurotrophin receptor
    The receptor p75 has also been implicated in retrograde degenerative signaling in motor neurons of superoxide dismutase 1 mutant mice (SOD1G93A), which undergo ...Missing: mutations | Show results with:mutations
  91. [91]
    Inhibition of motor neuron death in vitro and in vivo by a p75 ...
    Feb 1, 2016 · The p75 neurotrophin receptor (p75NTR; also known as NGFR) can mediate neuronal apoptosis in disease or following trauma, and facilitate ...
  92. [92]
    [PDF] p75NTR in Huntington's disease: beyond the basal ganglia
    Dec 17, 2015 · Interestingly, such increase was reversed in double mutant HdhQ7/Q111:p75+/- mice suggesting a direct link between aberrant p75NTR activity,.
  93. [93]
  94. [94]
    small molecule p75NTR ligand normalizes signalling and reduces ...
    The p75NTR ligand also decreased inflammation, increased striatal and hippocampal dendritic spine density, and improved motor performance and cognition in R6/2 ...Missing: orthologs | Show results with:orthologs
  95. [95]
    ProNGF and Neurodegeneration in Alzheimer's Disease - Frontiers
    ProNGF controls survival and death of NGF-dependent neurons in the basal forebrain, depending on the balance of TrkA and p75NTR. This is particularly important ...Missing: upregulated | Show results with:upregulated
  96. [96]
    Mediated Apoptosis via Neurotrophin Receptor-Interacting Factor in ...
    Sep 24, 2008 · The p75 neurotrophin receptor (p75NTR) can mediate many diverse cellular functions such as survival, apoptosis, and axonal growth. These ...
  97. [97]
    p75NTR Regulates Aβ Deposition by Increasing Aβ Production But ...
    Feb 9, 2011 · Our data suggest that p75NTR plays a critical role in regulating Aβ levels by both increasing Aβ production and attenuating its aggregation, and ...Aβ Preparation · Inhibition Of Aβ... · The Levels Of P75ntr In The...
  98. [98]
    Ameliorative Effects of p75NTR-ED-Fc on Axonal Regeneration and ...
    Nov 15, 2014 · Therefore, the p75NTR antagonist, such as recombinant p75NTR protein or its homogenates may block the inhibitory effects of myelin and promote ...
  99. [99]
    Suppression of the p75 Neurotrophin Receptor in Uninjured ...
    Nov 15, 2006 · The p75 neurotrophin receptor (p75NTR) has been implicated in diverse neuronal responses, including survival, cell death, myelination, and ...
  100. [100]
    Brain-derived neurotrophic factor contributes to activity-induced ...
    Nov 2, 2023 · BDNF, synthesized in the primary DRG neurons, may act as an autocrine or paracrine signal to activate the pre-synaptic TrkB, or p75NTR, ...
  101. [101]
    Suppression of the p75 Neurotrophin Receptor in Uninjured ...
    Our results show that p75NTR induced in undamaged DRG neurons facilitates TrkA signaling and contributes to heat and cold hyperalgesia. Keywords: p75 ...
  102. [102]
  103. [103]
    p75NTR antagonists attenuate photoreceptor cell loss in murine ...
    Jul 13, 2017 · Retinal explants treated with p75NTR antagonists showed significantly reduced photoreceptor cell death ... Photoreceptor degeneration is ...
  104. [104]
    The potential role of the p75 receptor in schizophrenia
    The purpose of this mini-review is to characterise the potential signalling mechanisms of the p75 receptor respective to neuropathological changes prevailing ...
  105. [105]
    p75NTR modulation prevents cellular, cortical activity and cognitive ...
    In summary, MPH-induced p75NTR upregulation causes reduced PV expression, abnormal brain activity and cognitive deficits in adult mice, and modulation of p75NTR ...
  106. [106]
    A Focus on the p75 Neurotrophin Receptor (p75NTR/CD271 ...
    Herein we review the expanding role that p75NTR plays in glioma progression with an emphasis on how p75NTR may contribute to the treatment refractory nature of ...
  107. [107]
    Glioma invasion mediated by the p75 neurotrophin receptor ...
    Using a novel invasive human glioma model, we previously identified the neurotrophin receptor p75 NTR (aka CD271) as a mediator of glioma invasion.
  108. [108]
    Decoding the Role of CD271 in Melanoma - PMC - PubMed Central
    The nerve growth factor receptor CD271 (p75NTR, NGFR) was identified in 1968, functionally described in 1986, and associated with melanoma aggressiveness and ...
  109. [109]
    Overexpression of p75(NTR) increases survival of breast cancer ...
    Aug 6, 2025 · While p75NTR supports tumor development in some of cancer types including glioma and breast cancer [89, 90], p75NTR also behaves as a tumor ...
  110. [110]
    CD271 Expression on Patient Melanoma Cells Is Unstable and ...
    This study presents evidence that calls into question a putative biomarker of melanoma stem cells, undermining an earlier rationale to explore its.
  111. [111]
    Cell Subpopulations Overexpressing p75NTR Have Tumor-initiating ...
    Cancer biology studies have shown that p75NTR-expressing melanoma cells have the ability to self-renew and cause tumor growth (29). In esophageal carcinomas, ...
  112. [112]
    Cancer stem cells-driven tumor growth and immune escape
    Furthermore, p75NTR regulates the expression of pluripotency transcription factors, including Sox2, Nanog, and MYC, promotes CSCs self-renewal in breast cancer, ...
  113. [113]
    p75NTR promotes survival of breast cancer resistant cells by ... - NIH
    In breast cancer, the neurotrophin receptor p75NTR is a critical factor that promotes resistance to the drug, however, its mechanism of action remains ...Missing: SOD2 neuroprotection
  114. [114]
    CD271 regulates the proliferation and motility of hypopharyngeal ...
    Jul 29, 2016 · CD271 (p75 neurotrophin receptor) plays both positive and negative roles in cancer development, depending on the cell type.
  115. [115]
    The p75 Neurotrophin Receptor Is a Central Regulator of Glioma ...
    ... glioblastoma multiforme;GFP, green fluorescent protein;NGF, nerve growth ... autocrine loop. We assessed the expression of several neurotrophins and ...
  116. [116]
    The p75 Neurotrophin Receptor Is a Central Regulator of Glioma ...
    Expression of p75NTR caused a significant increase in migration and invasion in vitro (Figure 3E and 3F). Treatment of these cells with NGF had no further ...Missing: CSCs | Show results with:CSCs
  117. [117]
    p75 Neurotrophin Receptor Cleavage by α- and γ-Secretases Is ...
    These results show that neurotrophins are secreted by BTICs and may activate receptors in an autocrine and/or paracrine fashion to mediate BTIC invasion, ...Knockdown Of P75ntr... · Ngf Triggers P75ntr Cleavage... · P75ntr, α-Secretase, And...Missing: loop CSCs
  118. [118]
    RELA is required for CD271 expression and stem-like ... - Nature
    Oct 22, 2022 · CD271 (also referred to as nerve growth factor receptor or p75NTR) is expressed on cancer stem cells in hypopharyngeal cancer (HPC) and ...
  119. [119]
    CD271 promotes proliferation and migration in bladder cancer
    Nov 28, 2023 · CD271 knockdown decreased proliferation in both adherent and spheroid cultures, and vice versa when CD271 was overexpressed in bladder cancer ...
  120. [120]
    CD271 mRNA/hnRNPA2B1 complex promotes proliferation ... - NIH
    CD271 critical for sphere formation in squamous cell carcinoma cells. We previously reported that CD271 defines cancer stem‐like cells in hypopharyngeal cancer ...
  121. [121]
    The role of the cancer stem cell marker CD271 in DNA damage ...
    Jan 23, 2017 · We demonstrate that expression of CD271 is responsible for genes associated with DNA repair and drug response.A Cd271-Associated Gene... · Knock-Down Of Cd271... · Cd271 Is Induced In A...<|control11|><|separator|>
  122. [122]
    Mechanisms involved in selecting and maintaining neuroblastoma ...
    NB cell responses to NT/TrkA activation are optimized by CD271/p75NTR, and TrkA/CD271/p75NTR co-expression in NB carries a good prognosis[16,69,148]. The ...
  123. [123]
    Neurotrophin and Neurotrophin Receptor Involvement in Human ...
    NBs that express TrkA but not CD271/p75NTR may have a worse prognosis, as they require higher NT concentrations for TrkA activation and signalling and would ...
  124. [124]
    Neurotrophin Trk Receptors: New Targets for Cancer Therapy
    In various cancers, however, activated p75NTR induces variable effects, from inhibition to stimulation of cell proliferation, dependent on their direct or ...
  125. [125]
    A comprehensive p75 neurotrophin receptor gene network ... - Nature
    Sep 11, 2020 · P75 neurotrophic receptor (p75NTR) is an important receptor for the role of neurotrophins in modulating brain plasticity and apoptosis.
  126. [126]
  127. [127]