Skip to main content
Dermato-endocrinology logoLink to Dermato-endocrinology
. 2011 Jan-Mar;3(1):32–36. doi: 10.4161/derm.3.1.14661

Neurotrophins in healthy and diseased skin

Francesca Truzzi 1, Alessandra Marconi 1, Carlo Pincelli 1,
PMCID: PMC3051851  PMID: 21519407

Abstract

Neurotrophins (NT) belong to a family of structurally and functionally related proteins that, depending on the tissue context and the receptors involved, promote either neuronal cell survival and differentiation or cell death. NT, and in particular NGF, were first identified as neurotrophic factors supporting the synthesis and development of sensory neurons in the central and peripheral nervous system. It is now widely accepted that NT also act as growth factors in non-neuronal cells, including the skin. In the skin, most cell types are able to secrete and/or to respond to stimulation by NT, creating a unique network of molecular signaling in the cutaneous microenvironment. Moreover, many skin diseases have been associated with an involvement of a number of neural factors including NT, but less attention has been given to the role of NT as growth factors in the development of skin pathologies. This review summarizes currently data on the expression and function of NT and their receptors in several cell types in the skin. Moreover it focuses on the role of the skin NT network in two cutaneous conditions, melanoma and psoriasis where NT are clearly involved.

Key words: neurotrophins, Trk, p75NTR, keratinocytes, melanocytes, fibroblasts, melanoma, psoriasis

Neurotrophins and Their Receptors

Neurotrophins (NT) belong to a group of functionally and structurally related proteins that were first identified as promoters for neuronal survival. Subsequently, it was demonstrated that they regulate many aspects of neuronal development and function, like synapse formation and synaptic plasticity.1 Nerve growth factor (NGF) was the first NT to be discovered during a search for survival factors.2 Brain-derived neurotrophic factor (BDNF) was the second NT to be characterized. It has been identified as a survival factor for several neuronal populations not responsive to NGF.3 These two proteins revealed conserved features of the sequences, leading to isolation of clones encoding additional members of this family.

Nowadays, we know that four NT are expressed in mammals: NGF, BDNF, NT-3 NT-4. NT play a critical role in developmental neurobiology. They play an essential role in cellular interactions, in controlling cell survival and differentiation.4 NGF is internalized by receptor-dependent mechanisms and transported along axons in membranous vesicles and energy-dependent processes.5

The effects of neurotrophins are mediated by two classes of cell-surface receptors, a family of tyrosine kinase receptors called Trks (TrkA, TrkB and TrkC) and the p75 neurotrophin receptor (NTR). Whereas p75NTR binds all neurotrophins with low affinity and specificity, Trk receptors bind neurotrophins with higher affinity and specificity. While TrkA binds NGF and NT-3, TrkB binds BDNF, NT-3, NT-4/5. TrkC only binds NT-3.

Trks are tyrosine kinase receptors, activated by ligand-induced formation of noncovalently associated receptor dimers. Several different mutation types in the trkA gene have been identified in congenital insensitivity to pain with anhydrosis (CIPA) patients.6 The phenotypes associated with CIPA are believed to result in large part from loss of NGF-dependent neurons, including nociceptive sensory and sympathetic neurons, during embryogenesis.

A dominant mutation in TrkB (Y722C) that impairs TrkB kinase signalling has recently been described in a patient with severe hyperphagic obesity and severe impairments in nociception, learning and memory.7

NT dimerize the Trk receptors, resulting in activation through transphosphrylation of the kinases present in their cytoplasmic domains. NT interact with these receptors at the membrane-proximal immunoglobulin-like domain. The three-dimensional structures of this domain in each of the Trk receptors have been solved.8 Expression of a specific Trk receptor confers responsiveness to the NT to which it binds. On the other hand the isoform of TrkA including an insert is also activated by NT-3 in addition to NGF,9 while the similar isoform of TrkB is activated by NT-3 and NT-4 in addition to BDNF.10 Thirty-six novel isoforms of TrkB proteins with unique properties have been described recently. This suggests high complexity in the synthesis, regulation and function of this important NT receptor, emphasizing the need for further study of these novel TrkB variants.11

TrkC has several characteristics of a tumor suppressor, and its expression in tumors has often been associated with good prognosis. TrkC was recently demonstrated to be a dependence receptor, transducing different positive signals in the presence of ligand but inducing apoptosis in the absence of ligand. Indeed, the TrkC ligand NT-3 (NT-3) is upregulated in a large fraction of aggressive human neuroblastomas (NB) and it blocks TrkC-induced apoptosis of human NB cell lines.12,13

p75NTR was the first NT receptor to be identified as a low-affinity receptor for NGF, but it was subsequently shown to bind each of the neurotrophins with a similar affinity.14,15 p75NTR is a member of the tumour necrosis factor receptor superfamily with an extra-cellular domain containing four cysteine-rich motifs, a single transmembrane domain and a cytoplasmic domain that includes a ‘death’ domain, similar to those present in other members of this family.16 Although this receptor does not contain a catalytic motif, it interacts with several protein that transmit important signals for regulating neuronal survival and differentiation as well as synaptic plasticity. The three-dimensional structure of the extracellular domain of p75NTR in association with an NGF dimer indicates that each of the four cysteine-rich repeats participates in binding to NGF.16 The binding of NGF to p75NTR may result in dissociation of p75NTR multimers and is compatible with the possibility that Trk and p75NTR monomers simultaneously bind the same neurotrophin dimer. A gene related to p75NTR, named NRH-2, has recently been identified. The product of this gene lacks the extracellular cysteine-rich repeats present in p75NTR and fails to bind NGF, but it is able to interact and influence the ligand-binding properties of TrkA.17

The role of p75NTR is still controversial: in the presence of Trk, p75NTR increases high-affinity NT binding, thereby enhancing the ability of Trk to promote survival.18 By contrast, in the absence of Trk, p75NTR can induce apoptosis, via its own signal transduction, by interacting with a mounting number of downstream molecules.19 Recently, it has been shown that the proform of NT proNGF binds p75NTR, in association with its co-receptor sortilin, but not Trk.20 More specifically, sortilin, a member of the vps-10 protein family, binds the “pro” region of NGF, whereas p75NTR binds mature NGF. The p75NTR-sortilin complex couples with proNGF to induce apoptosis.21

Neurotrophins and the Skin

NT and in particular NGF were first identified as neurotrophic factors supporting the synthesis and development of sensory neurons, that in turn extend their dendrites into the skin, thus playing an essential role in skin innervation. Indeed, null mutations of genes from the NGF family of NT and their receptors (NTRs) lead to loss/reduction of specific neurons in sensory ganglia; conversely, cutaneous overexpression of NT results in skin hyperinnervation and increase in the number of sensory neurons innervating the skin.22 In addition, NGF synthesized in the epidermis, is retrogradelly transported to the ganglia to stimulate the release of neuropeptides in the skin, thus favoring cutaneous neurogenic inflammation.23 Subsequently, it became clear that NGF possesses a number of biological effects also on non-neuronal cells. In particular, Pincelli and co-workers first demonstrated that normal human keratinocytes synthesize and release NGF that can act as a growth factor for these cells.24 Later, expression and function of NGF and other NT was shown in other skin cells, definitely indicating that NT play very important functions other than supporting skin innervation. In this review, we focus on NT as growth factors in most cutaneous cells and report the concept of a rich NT network in the skin.

Keratinocytes.

Human keratinocytes synthesize and secrete biologically active NGF,25,26 BDNF, NT-3 and NT-4.27,28 NGF is secreted at highest levels as compared to the other NT, whereas NT-3 and NGF upregulate each other's secretion in human keratinocytes. NT-3 release is augmented by UVA, whereas NGF expression is downregulated by UVB irradiation.29,30 In human skin, NGF is released in increasing amounts by proliferating keratinocytes, whereas secretion ends in more differentiated cells.24 NGF is able to induce proliferation in human keratinocytes,31 and it can either stimulate or inhibit murine epidermal and hair follicle keratinocyte proliferation in situ.32 Human keratinocytes express the high affinity receptors TrkA and TrkC, but not the functional form of the TrkB.28 Inhibiting TrkA phosphorylation with the natural alkaloid k252a, in absence of exogenous NGF, reduces keratinocyte proliferation.33 This indicates that endogenous NGF autocrinally sustains keratinocyte proliferation. Keratinocytes transfected with NGF proliferate to a greater extent as compared to mock cells,29 while increasing amounts of NT-3 antibody inhibits keratinocyte proliferation.28 NT together with their receptors TrkA and TrkB are able to stimulate mouse keratinocyte proliferation in ex-vivo cultured skin explants.27,32 NT also modulate susceptibility to apoptosis in the epidermis. While UVB downregulates both NGF and TrkA and induce apoptosis in human keratinocytes,29 autocrine NGF protects these cells from UVB-induced apoptosis.29 Because normal human keratinocytes typically lack TrkB expression, NT may exert different functions in this system, by binding p75NTR alone. In this context, p75NTR acts as a proapoptotic receptor. This is exemplified by BDNF and NT4, which induce a higher rate of apoptosis in normal human keratinocytes overexpressing p75NTR, as compared to mock-transfected cells. On the other hand, p75NTR siRNA-transfected keratinocytes fail to undergo cell death after administration of NT4 (Truzzi F, et al. unpublished data).

Interestingly, NGF is mostly expressed and released by stem keratinocytes (KSC),28 and blocking TrkA inhibits the proliferation of this keratinocyte subpopulation.28 This seems to indicate that a NGF-TrkA autocrine loop, by acting as both a mitogenic and survival factor, contributes to the maintenance of the so-called “stemness” in keratinocytes. On the other hand, p75NTR is almost exclusively expressed in the more differentiated transit amplifying (TA) cells, and it is barely detected in KSC. TA cells have been shown to be more susceptible than KSC to apoptosis.34 Therefore, p75NTR being predominantly expressed in TA cells seems to be consistent with the pro-apoptotic role of this receptor in human keratinocytes (Truzzi F, et al. submitted). Taken together, these results point to a critical role of NT and their receptors in epidermal homeostasis.

Melanocytes.

In human skin, melanocytes reside at the dermo-epidermal junction and in the hair matrix. During skin development, melanoblasts, which derive from neural crest, migrate into the skin and differentiate into melanocytes. Together with other paracrine signalling molecules, NT are important for melanocytes migration, viability and differentiation.35,36 Normal human melanocytes seem to be a target of the NT skin network, because they express all the NT receptors both in vitro and in vivo.37 Normal human melanocytes also express p75NTR, which is upregulated by different stimuli, like UV irradiation.38 NT influence melanocytes in paracrine fashion: for example, in vitro NGF is chemotactic for melanocytes and stimulate their dendrite formation.26 NGF is implicated in melanocyte survival,39 migration and dendricity,26 and its synthesis and secretion are enhanced by UV irradiation.40 Moreover, when melanocytes are irradiated with UV, NGF reduces apoptosis through upregulation of the anti-apoptotic Bcl-2 protein in vivo.30 From these data it is possibile to conclude that NT are important for protection of UV-induced oxidative stress and apoptosis in melanocytes. Melanocytes produce all NT, while when these factors are added to the cultures, they fail to stimulate cell proliferation.37 When melanocytes are maintained in growth factor-depleted medium, NGF and NT-3 promote melanocyte survival.41 Both NT3 and NT4 secretion promotes the synthesis of tyrosinase and tyrosinase-related peptide (TRP)-1, critical enzyme of melanin biosynthesis.37 NT-3, NT-4 or NGF significantly increases the melanin content when stimulated with endotelin-1.37

Human melanocytes express Trk receptors.37,41 Interestingly, phorbol-12-tetradecantate-13-acetate, a strong activator of protein kinase C, induces the expression of TrkA41 and decrease the expression of TrkC, suggesting that NGF and NT3 mediate different signals through their specific high affinity receptors. Melanocytes expess also the NT low affinity receptor p75NTR, which expression is upregulated after TPA treatment.41

Fibroblasts.

NT and their receptors are expressed both in dermal fibroblasts and in the more differentiated myofibroblasts. p75NTR and TrkB are expressed at higher levels in myofibroblasts than in fibroblasts, which in contrast express higher levels of TrkA. Dermal fibroblast and myofibroblasts secrete NGF and NT3 at higher levels than NT4 and BDNF. Exogenous NT per se fail to stimulate dermal fibroblast proliferation, possibly because endogenous production of NT is sufficient to normal cell activity and survival. Interestingly, NT also promote fibroblast differentiation into myofibroblasts, by inducing α-SMA expression, indicating that NT could have a functional role in the fibro-myofibroblast system (Palazzo E, et al. manuscript in preparation). It has been shown that NGF induce fibroblast-like keratinocyte differentiation into myofibroblasts,42 their contraction in 3D collagen matrix42 and the expression of MMP-9 (Metalloprotease-9) in keratoconjunctivitis-derived fibroblasts,43 and different works show the applicative possibility of this NGF function.4446 It has been demonstrated that all NT promote fibroblast migration while NGF and BDNF promote their contractile activity. Therefore NGF and BDNF, produced by dermal and epidermal cells, could be key regulators of the biomechanical properties in the dermis.

Other skin cells.

Skin mast cells express functional TrkA and produce NGF.47 They also express p75NTR mRNA and protein.48 NGF has been shown to play a collaborative action on mediator release from residential tissue mast cells in rat skin by in vivo extravasation assay. This result implies that the activation pathway in mast cells may occur in the pathophysiological condition. Activation of mast cells induces the release of histamine, leukotrienes and produces inflammatory cytokines, resulting in the recruitment and activation of circulating leukocytes to the area of allergic and non allergic inflammation.49

Merkel cells are sensory cells of neural crest origin. NT have been investigated during development in mice finding that neither NT-3 nor TrkC and p75NTR are expressed by Merkel cells in the murine whisker. At the time of birth, however, Merkel cells are immunoreactive for NT-3, TrkC and p75NTR. In TrkC null and NT-3 null mice, Merkel cells differentiate initially, but undergo apoptosis perinatally. These results show that NT-3 signaling is not required for the differentiation of Merkel cells, but that it is essential for their postnatal survival.50 Merkel cell of whisker follicles of NT3 null newborns exhibited decreased immunoreactivity for cytokeratin 8 and contained apoptotic bodies that were positive for cleaved caspase-3. It was suggested that perinatal apoptosis is responsible for the loss of Merkel cells lacking innervation in NT3 null mice. TrkB was shown to be important for Merkel cells development, as mice lacking TrkB lost 32% of neurons and this neuronal loss was associated with the absence of Meissner corpuscles and reduction of hair follicle-associated sensory nerve endings and Merkel cells.51 Using mice lacking p75NTR, indicates that Merkel cells require p75NTR during the late postnatal development.52

Neurotrophins in Diseased Skin

Many skin diseases have been associated with an involvement of a number of neural factors including NT. There is a huge body of literature on the increased nerve density in several dermatoses which has lead many investigators to conclude that augmented sprouting of nerves and upregulation of neuropeptide release are involved in the pathogenesis of certain cutaneous conditions. It remains to be defined whether hyperinnervation and the release of neural substances associated with neurogenic inflammation are primary phenomena or just amplification events in skin diseases.

By contrast, less attention has been given to the role of NT as growth factors in the development of skin pathologies. Here, we will review two cutaneous conditions, melanoma and psoriasis where NT are clearly involved.

Melanoma.

Morphologically, melanomas can be subdivided in several subtypes, such as epithelioid, pleomorphic spindle cell and desmoplastic melanoma. Iwamoto et al. found that there was no detectable p75NTR immunostaining in melanocytes of the normal epidermis, whereas 13 of 14 benign nevi showed detectable p75NTR. This was primarily within the spindled nevocytic structures within the dermis.53 Interestingly, p75NTR is highly expressed in desmoplastic melanomas and spindle cell melanomas and weakly expressed in the epithelioid melanomas.54 In a study by Huttenbach et al. only 33% of their desmoplastic melanomas stained positive with p75NTR,55 while more recently Lazova56 showed p75NTR staining to be more diffuse and intense as compared with S100, as reported previously by Kanik et al.

Recently Boiko et al. showed that p75NTR (CD271), which is considered a neural crest stem cell marker, allows the identification and isolation of melanoma cancer stem cells. Indeed, tumors derived from transplanted human CD271+ melanoma cells were capable of metastasizing in vivo.58 Marchetti et al. using melanoma cell lines, observed that NGF/p75NTR signaling promotes the survival of melanoma cells.59 They also observed the presence of NGF and NT-3 in tumor adjacent tissues at the invasive front of melanoma brain metastases, which might indicate a paracrine activation of p75NTR and TrkC in melanoma cells by NGF and NT-3 produced by nearby glial cells. Besides promoting melanoma cell survival, neurotrophins also induce the expression in melanoma cells of heparanase, an important enzyme for local invasion and metastasis, that cleave heparan sulfate chains of proteoglycans, thus modifying the extracellular matrix of tumor cells.60 Moreover, Shonukan et al. established that neurotrophins are chemotactic for melanoma cells, triggered by p75NTR-mediated dephosphorylation of the actin-bundling protein fascin.61 In addition, TrkA is expressed by primary and metastatic melanomas and is associated with poor clinical outcome.62

Melanoma originates from melanocytes. Whereas melanocytes seem to benefit from the cutaneous neurotrophic network mainly as paracrine targets, once malignant transformation occurs, melanoma cells acquire self-renewal capabilities mediated also by autocrine NT loops. Indeed all NT, particularly NT-3 and NT-4, have been detected in conditioned medium of different melanoma cell lines.63 Recent results show that melanoma cells proliferate through autocrine NT stimulation. K252a significantly reduces melanoma cell proliferation by inhibiting Trk phosphorylation. Proliferation is significantly reduced when endogenous NT are removed from the culture medium by soluble Trk/Fc receptors. NT appeared to be important for melanoma cell migration in vitro, with special respect for metastatic cell lines. The migratory phenotype is necessarily dependent on the presence of both the high- and low-affinity NT receptors. Cells treated with p75NTR small interfering RNA (p75NTR siRNA) fail to respond to NT stimulation. Similarly, the administration of K252a blocks melanoma cell migration, confirming that NT stimulate melanoma cell migration and invasion cooperation of the low- and high-affinity receptors.63

Psoriasis.

Psoriasis is a chronic-relapsing inflammatory disease characterized by keratinocyte hyperproliferation and reduced apoptosis, leading to an increased epidermal turnover. Interestingly, NGF that is both a mitogen and a survival factor for keratinocytes, is overexpressed in psoriatic lesions as well as in psoriatic keratinocytes,64,65 and its high-affinity receptor TrkA, that is located only in basal keratinocytes in healthy skin, is expressed throughout all epidermal layers in psoriasis.66 On the other hand, p75NTR that plays a proapoptotic role in keratinocytes, is absent in psoriatic keratinocytes. The rate of apoptosis in psoriatic TA cells is significantly lower as compared to TA cells from normal epidermis. Interestingly, TA cells from psoriatic skin express lower levels of p75NTR as compared to TA from normal skin. Thus, absence of p75NTR in TA cells could account for the low keratinocyte apoptosis in psoriasis. In fact, infection of psoriatic TA with p75NTR restores apoptosis (Truzzi F, et al. submitted). Taken together, these results suggest that, under normal conditions, there is a balance between Trk and p75NTR functions, that allows epidermal homeostasis. On the contrary, in psoriasis, NGF and Trk upregulation associated with reduced p75NTR expression result in increased keratinocyte proliferation and reduced apoptosis, thus favoring epidermal thickness, a typical feature of this dermatosis. Interestingly, k252a, that has been shown to improve psoriasis in vivo in the SCID mouse model,67 is now in Phase II clinical trial as a novel topical treatment for psoriasis.

In conclusion, NTs influence various cellular functions in normal and diseased skin. In the last decade, substantial progress has been made in the understanding of the molecular mechanisms involved in NT signaling. Additional studies are necessary to evaluate such molecular pathways in cells from healthy and pathologic skin. A translation of the relevant data from the field of cutaneous neurobiology into clinically-oriented research will unravel novel therapeutic strategies for the treatment of skin diseases, by targeting the skin NT network.

References

  • 1.Huang EJ, Reichardt LF. Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem. 2003;72:609–642. doi: 10.1146/annurev.biochem.72.121801.161629. [DOI] [PubMed] [Google Scholar]
  • 2.Levi-Montalcini R. The nerve growth factor 35 years later. Science. 1987;237:1154–1162. doi: 10.1126/science.3306916. [DOI] [PubMed] [Google Scholar]
  • 3.Barde YA, Edgar D, Thoenen H. Purification of a new neurotrophic factor from mammalian brain. EMBO J. 1982;1:549–553. doi: 10.1002/j.1460-2075.1982.tb01207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Segal RA. Selectivity in neurotrophin signaling: theme and variations. Annu Rev Neurosci. 2003;26:299–330. doi: 10.1146/annurev.neuro.26.041002.131421. [DOI] [PubMed] [Google Scholar]
  • 5.Thoenen H, Barde YA. Physiology of nerve growth factor. Physiol Rev. 1980;60:1284–1335. doi: 10.1152/physrev.1980.60.4.1284. [DOI] [PubMed] [Google Scholar]
  • 6.Indo Y. Molecular basis of congenital insensitivity to pain with anhidrosis (CIPA): mutations and polymorphisms in TRKA (NTRK1) gene encoding the receptor tyrosine kinase for nerve growth factor. Hum Mutat. 2001;18:462–471. doi: 10.1002/humu.1224. [DOI] [PubMed] [Google Scholar]
  • 7.Yeo GS, Connie Hung CC, Rochford J, Keogh J, Gray J, Sivaramakrishnan S, et al. A de novo mutation affecting human TrkB associated with severe obesity and developmental delay. Nat Neurosci. 2004;7:1187–1189. doi: 10.1038/nn1336. [DOI] [PubMed] [Google Scholar]
  • 8.Ultsch MH, Wiesmann C, Simmons LC, Henrich J, Yang M, Reilly D, et al. Crystal structures of the neurotrophin-binding domain of TrkA, TrkB and TrkC. J Mol Biol. 1999;290:149–159. doi: 10.1006/jmbi.1999.2816. [DOI] [PubMed] [Google Scholar]
  • 9.Clary DO, Reichardt LF. An alternatively spliced form of the nerve growth factor receptor TrkA confers an enhanced response to neurotrophin 3. Proc Natl Acad Sci USA. 1994;91:11133–11137. doi: 10.1073/pnas.91.23.11133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Strohmaier C, Carter BD, Urfer R, Barde YA, Dechant G. A splice variant of the neurotrophin receptor trkB with increased specificity for brain-derived neurotrophic factor. EMBO J. 1996;15:3332–3337. [PMC free article] [PubMed] [Google Scholar]
  • 11.Luberg K, Wong J, Weickert CS, Timmusk T. Human TrkB gene: novel alternative transcripts, protein isoforms and expression pattern in the prefrontal cerebral cortex during postnatal development. J Neurochem. 2010;113:952–964. doi: 10.1111/j.1471-4159.2010.06662.x. [DOI] [PubMed] [Google Scholar]
  • 12.Bouzas-Rodriguez J, Cabrera JR, Delloye-Bourgeois C, Ichim G, Delcros JG, Raquin MA, et al. Neurotrophin-3 production promotes human neuroblastoma cell survival by inhibiting TrkC-induced apoptosis. J Clin Invest. 2010;120:850–858. doi: 10.1172/JCI41013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Nikoletopoulou V, Lickert H, Frade JM, Rencurel C, Giallonardo P, Zhang L, et al. Neurotrophin receptors TrkA and TrkC cause neuronal death whereas TrkB does not. Nature. 2010;467:59–63. doi: 10.1038/nature09336. [DOI] [PubMed] [Google Scholar]
  • 14.Rodriguez-Tébar A, Dechant G, Barde YA. Binding of brain-derived neurotrophic factor to the nerve growth factor receptor. Neuron. 1990;4:487–492. doi: 10.1016/0896-6273(90)90107-q. [DOI] [PubMed] [Google Scholar]
  • 15.Frade JM, Barde YA. Nerve growth factor: two receptors, multiple functions. Bioessays. 1998;20:137–145. doi: 10.1002/(SICI)1521-1878(199802)20:2<137::AID-BIES6>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  • 16.He XL, Garcia KC. Structure of nerve growth factor complexed with the shared neurotrophin receptor p75. Science. 2004;304:870–875. doi: 10.1126/science.1095190. [DOI] [PubMed] [Google Scholar]
  • 17.Murray SS, Perez P, Lee R, Hempstead BL, Chao MV. A novel p75 neurotrophin receptor-related protein, NRH2, regulates nerve growth factor binding to the TrkA receptor. J Neurosci. 2004;24:2742–2749. doi: 10.1523/JNEUROSCI.3960-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dechant G, Barde YA. The neurotrophin receptor p75(NTR): novel functions and implications for diseases of the nervous system. Nat Neurosci. 2002;5:1131–1136. doi: 10.1038/nn1102-1131. [DOI] [PubMed] [Google Scholar]
  • 19.Wang JJ, Tasinato A, Ethell DW, Testa MP, Bredesen DE. Phosphorylation of the common neurotrophin receptor p75 by p38beta2 kinase affects NFkappaB and AP-1 activities. J Mol Neurosci. 2000;15:19–29. doi: 10.1385/JMN:15:1:19. [DOI] [PubMed] [Google Scholar]
  • 20.Nykjaer A, Lee R, Teng KK, Jansen P, Madsen P, Nielsen MS, et al. Sortilin is essential for proNGF-induced neuronal cell death. Nature. 2004;427:843–848. doi: 10.1038/nature02319. [DOI] [PubMed] [Google Scholar]
  • 21.Kaplan DR, Miller FD. Neurobiology: a move to sort life from death. Nature. 2004;427:798–799. doi: 10.1038/427798a. [DOI] [PubMed] [Google Scholar]
  • 22.Montaño JA, Pérez-Piñera P, García-Suárez O, Cobo J, Vega JA. Development and neuronal dependence of cutaneous sensory nerve formations: Lessons from neurotrophins. Microsc Res Tech. 2010;73:513–529. doi: 10.1002/jemt.20790. [DOI] [PubMed] [Google Scholar]
  • 23.Davis BM, Fundin BT, Albers KM, Goodness TP, Cronk KM, Rice FL. Overexpression of nerve growth factor in skin causes preferential increases among innervation to specific sensory targets. J Comp Neurol. 1997;387:489–506. doi: 10.1002/(sici)1096-9861(19971103)387:4<489::aid-cne2>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 24.Pincelli C, Fantini F, Giannetti A. Nerve growth factor and the skin. Int J Dermatol. 1994;33:308–312. doi: 10.1111/j.1365-4362.1994.tb01058.x. [DOI] [PubMed] [Google Scholar]
  • 25.Di Marco E, Marchisio PC, Bondanza S, Franzi AT, Cancedda R, De Luca M. Growth-regulated synthesis and secretion of biologically active nerve growth factor by human keratinocytes. J Biol Chem. 1991;266:21718–21722. [PubMed] [Google Scholar]
  • 26.Yaar M, Grossman K, Eller M, Gilchrest BA. Evidente for nerve growth factor-mediated paracrine effects in human epidermis. J Cell Biol. 1991;115:821–828. doi: 10.1083/jcb.115.3.821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Botchkarev VA, Metz M, Botchkareva NV, Welker P, Lommatzsch M, Renz H, et al. Brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin-4 act as “epitheliotrophins” in murine skin. Lab Invest. 1999;79:557–572. [PubMed] [Google Scholar]
  • 28.Marconi A, Terracina M, Fila C, Franchi J, Bonté F, Romagnoli G, et al. Expression and function of neurotrophins and their receptors in cultured human keratinocytes. J Invest Dermatol. 2003;121:1515–1521. doi: 10.1111/j.1523-1747.2003.12624.x. [DOI] [PubMed] [Google Scholar]
  • 29.Marconi A, Vaschieri C, Zanoli S, Giannetti A, Pincelli C. Nerve growth factor protects human keratinocytes from ultraviolet-B-induced apoptosis. J Invest Dermatol. 1999;113:920–927. doi: 10.1046/j.1523-1747.1999.00773.x. [DOI] [PubMed] [Google Scholar]
  • 30.Stefanato CM, Yaar M, Bhawan J, Philips TJ, Kosmadaki MG, Botchkarev V, et al. Modulations of nerve growth factor and Bcl-2 in ultraviolet-irradiated human epidermis. J Cutan Pathos. 2003;30:351–357. doi: 10.1034/j.1600-0560.2003.00065.x. [DOI] [PubMed] [Google Scholar]
  • 31.Di Marco E, Mathor M, Bondanza S, Cutuli N, Marchisio PC, Cancedda R, et al. Nerve growth factor binds to normal human keratinocytes through high and low affinity receptors and stimulates their growth by a novel autocrine loop. J Biol Chem. 1993;268:22838–22846. [PubMed] [Google Scholar]
  • 32.Paus R, Lüftl M, Czarnetzki BM. Nerve growth factor modulates keratinocyte proliferation in murine skin organ culture. Br J Dermatol. 1994;130:174–180. doi: 10.1111/j.1365-2133.1994.tb02896.x. [DOI] [PubMed] [Google Scholar]
  • 33.Pincelli C, Haake AR, Benassi L, Grassilli E, Magnoni C, Ottani D, et al. Autocrine nerve growth factor protects human keratinocytes from apoptosis through its high affinity receptor (TRK): a role for BCL-2. J Invest Dermatol. 1997;109:757–764. doi: 10.1111/1523-1747.ep12340768. [DOI] [PubMed] [Google Scholar]
  • 34.Tiberio R, Marconi A, Fila C, Fumelli C, Pignatti M, Krajewski S, et al. Keratinocytes enriched for stem cells are protected from anoikis via an integrin signaling pathway in a Bcl-2 dependent manner. FEBS Lett. 2002;524:139–144. doi: 10.1016/s0014-5793(02)03040-5. [DOI] [PubMed] [Google Scholar]
  • 35.Pincelli C, Yaar M. Nerve growth factor: its significance in cutaneous biology. J Invetig Dermatol Symp Proc. 1997;2:61–68. doi: 10.1038/jidsymp.1997.8. [DOI] [PubMed] [Google Scholar]
  • 36.Yaar M. Neurotrophins in skin. In: Sieber-Blum M, editor. Neurotrophins and the neural crest. Boston-London: CRC Press; 1999. pp. 117–140. [Google Scholar]
  • 37.Marconi A, Panza MC, Bonnet-Duquennoy M, Lazou K, Kurfurst R, Truzzi F, et al. Espression and function of neurotrophins and their receptors in human melanocytes. Int J Cosmet Sci. 2006;28:255–261. doi: 10.1111/j.1467-2494.2006.00321.x. [DOI] [PubMed] [Google Scholar]
  • 38.Peacocke M, Yaar M, Mansur CP, Chao MV, Gilchrest BA. Induction of nerve growth factor receptor on cultured human melanocytes. Proc Natl Acad Sci USA. 1988;85:5282–5286. doi: 10.1073/pnas.85.14.5282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhai S, Yaar M, Doyle SM, Gilchrest BA. Nerve growth factor rescues pigment cells from ultraviolet-induced apoptosis by upregulating BCL-2 levels. Exp Cell Res. 1996:224–335. doi: 10.1006/excr.1996.0143. [DOI] [PubMed] [Google Scholar]
  • 40.Tron VA, Coughlin MD, Jang DE, Stanisz J, Sauder DN. Expression and modulation of nerve growth factor in murine keratinocytes (PAM 212) J Clin Invest. 1990;85:1085. doi: 10.1172/JCI114539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yaar M, Eller MS, DiBenedetto P, Reenstra WR, Zhai S, McQuaid T, et al. The trk family of receptors mediates nerve growth factor and neurotrophin-3 effects in melanocytes. Clin Invest. 1994;94:1550–1562. doi: 10.1172/JCI117496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Micera A, Vigneti E, Pickholtz D, Reich R, Pappo O, Bonini S, et al. Nerve growth factor displays stimulatory effects on human skin and lung fibroblasts, demonstrating a direct role for this factor in tissue repair. Proc Natl Acad Sci USA. 2001;98:6162–6167. doi: 10.1073/pnas.101130898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Micera A, Lambiase A, Stampachiacchiere B, Sgrulletta R, Normando EM, Bonini S, et al. Nerve growth factor has a modulatory role on human primary fibroblast cultures derived from vernal keratoconjunctivitis-affected conjunctiva. Mol Vis. 2007;13:981–987. [PMC free article] [PubMed] [Google Scholar]
  • 44.Landi F, Aloe L, Russo A, Cesari M, Onder G, Bonini S, et al. Topical Treatment of Pressure Ulcers with Nerve Growth Factor. A Randomized Clinical Trial. Ann Int Med. 2003;139:635–641. doi: 10.7326/0003-4819-139-8-200310210-00006. [DOI] [PubMed] [Google Scholar]
  • 45.Aloe L, Tirassa P, Lambiase A. The topical application of nerve growth factor as a pharmacological tool for human corneal and skin ulcers. Pharmacol Res. 2008;57:253–258. doi: 10.1016/j.phrs.2008.01.010. [DOI] [PubMed] [Google Scholar]
  • 46.Sun W, Lin H, Chen B, Zhao W, Zhao Y, Xiao Z, et al. Collagen scaffolds loaded with collagen-binding NGFbeta accelerate ulcer healing. J Biomed Mater Res A. 2010;92:887–895. doi: 10.1002/jbm.a.32445. [DOI] [PubMed] [Google Scholar]
  • 47.Marshall JS, Gomi K, Blennerhassett MG, Bienenstock J. Nerve growth factor modifies the expression of inflammatory cytokines by mast cells via a prostanoiddependent mechanism. J Immunol. 1999;162:4271–4276. [PubMed] [Google Scholar]
  • 48.Fischer TC, Lauenstein HD, Serowka F, Pilzner C, Groneberg DA, Welker P. Pan-neurotrophin receptor p75NTR expression is strongly induced in lesional atopic mast cells. Clin Exp Allergy. 2008;38:1168–1173. doi: 10.1111/j.1365-2222.2008.02994.x. [DOI] [PubMed] [Google Scholar]
  • 49.Kawamoto K, Aoki J, Tanaka A, Itakura A, Hosono H, Arai H, et al. Nerve growth factor activates mast cells through the collaborative interaction with lysophosphatidylserine expressed on the membrane surface of activated platelets. J Immunol. 2002;168:6412–6419. doi: 10.4049/jimmunol.168.12.6412. [DOI] [PubMed] [Google Scholar]
  • 50.Szeder V, Grim M, Kucera J, Sieber-Blum M. Neurotrophin-3 signaling in mammalian Merkel cell development. Dev Dyn. 2003;228:623–629. doi: 10.1002/dvdy.10403. [DOI] [PubMed] [Google Scholar]
  • 51.Perez-Pinera P, García-Suarez O, Germanà A, Díaz-Esnal B, de Carlos F, Silos-Santiago I, et al. Characterization of sensory deficits in TrkB knockout mice. Neurosci Lett. 2008;433:43–47. doi: 10.1016/j.neulet.2007.12.035. [DOI] [PubMed] [Google Scholar]
  • 52.Kinkelin I, Stucky CL, Koltzenburg M. Postnatal loss of Merkel cells, but not of slowly adapting mechanoreceptors in mice lacking the neurotrophin receptor p75. Eur J Neurosci. 1999;11:3963–3969. doi: 10.1046/j.1460-9568.1999.00822.x. [DOI] [PubMed] [Google Scholar]
  • 53.Iwamoto S, Burrows RC, Agoff SN, Piepkorn M, Bothwell M, Schmidt R. The p75 neurotrophin receptor, relative to other Schwann cell and melanoma markers, is abundantly Expressed in spindled melanomas. Am J Dermatopathol. 2001;23:288–294. doi: 10.1097/00000372-200108000-00002. [DOI] [PubMed] [Google Scholar]
  • 54.Iwamoto S, Odland PB, Piepkorn M, Bothwell M. Evidence that the p75 neurotrophin receptor mediates perineural spread of desmoplastic melanoma. J Am Acad Dermatol. 1996;35:725–731. doi: 10.1016/s0190-9622(96)90728-8. [DOI] [PubMed] [Google Scholar]
  • 55.Huttenbach Y, Prieto VG, Reed JA. Desmoplastic and spindle cell melanomas express protein markers of the neural crest but not of later committed stages of Schwann cell differentiation. J Cutan Pathol. 2002;29:562–568. doi: 10.1034/j.1600-0560.2002.290909.x. [DOI] [PubMed] [Google Scholar]
  • 56.Lazova R, Tantcheva-Poor I, Sigal AC. p75 nerve growth factor receptor staining is superior to S100 in identifying spindle cell and desmoplastic melanoma. J Am Acad Dermatol. 2010;63:852–858. doi: 10.1016/j.jaad.2009.11.688. [DOI] [PubMed] [Google Scholar]
  • 57.Kanik AB, Yaar M, Bhawan J. p75 nerve growth factor receptor staining helps identify desmoplastic and neurotropic melanoma. J Cutan Pathol. 1996;23:205–210. doi: 10.1111/j.1600-0560.1996.tb01468.x. [DOI] [PubMed] [Google Scholar]
  • 58.Boiko AD, Razorenova OV, van de Rijn M, Swetter SM, Johnson DL, Ly DP, et al. Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature. 2010;466:133–137. doi: 10.1038/nature09161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Marchetti D, Denkins Y, Reiland J, Greiter-Wilke A, Galjour J, Murry B, et al. Brainmetastatic melanoma: a neurotrophic perspective. Pathol Oncol Res. 2003;9:147–158. doi: 10.1007/BF03033729. [DOI] [PubMed] [Google Scholar]
  • 60.Walch ET, Albino AP, Marchetti D. Correlation of overexpression of the low-affinity p75 neurotrophin receptor with augmented invasion and heparanase production in human malignant melanoma cells. Int J Cancer. 1999;82:112–120. doi: 10.1002/(sici)1097-0215(19990702)82:1<112::aid-ijc19>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
  • 61.Shonukan O, Bagayogo I, McCrea P, Chao M, Hempstead B. Neurotrophin-induced melanoma cell migration is mediated through the actin-bundling protein fascin. Oncogene. 2003;22:3616–3623. doi: 10.1038/sj.onc.1206561. [DOI] [PubMed] [Google Scholar]
  • 62.Flørenes VA, Maelandsmo GM, Holm R, Reich R, Lazarovici P, Davidson B. Expression of activated TrkA protein in melanocytic tumors: relationship to cell proliferation and clinical outcome. Am J Clin Pathol. 2004;122:412–420. doi: 10.1309/CHFH-EYAT-44WW-P7J3. [DOI] [PubMed] [Google Scholar]
  • 63.Truzzi F, Marconi A, Lotti R, Dallaglio K, French LE, Hempstead BL, et al. Neurotrophins and their receptors stimulate melanoma cell proliferation and migration. J Invest Dermatol. 2008;128:2031–2040. doi: 10.1038/jid.2008.21. [DOI] [PubMed] [Google Scholar]
  • 64.Fantini F, Magnoni C, Bracci-Laudiero L, Pincelli C. Nerve growth factor is increased in psoriatic skin. J Invest Dermatol. 1995;105:854–855. doi: 10.1111/1523-1747.ep12326689. [DOI] [PubMed] [Google Scholar]
  • 65.Raychaudhuri SP, Jiang WY, Farber EM. Psoriatic keratinocytes express high levels of nerve growth factor. Acta Derm Venereol. 1998;78:84–86. doi: 10.1080/000155598433368. [DOI] [PubMed] [Google Scholar]
  • 66.Pincelli C. Nerve growth factor and keratinocytes: a role in psoriasis. Eur J Dermatol. 2000;10:85–90. [PubMed] [Google Scholar]
  • 67.Raychaudhuri SP, Sanyal M, Weltman H, Kundu-Raychaudhuri S. K252a, a high-affinity nerve growth factor receptor blocker, improves psoriasis: an in vivo study using the severe combined immunodeficient mouse-human skin model. J Invest Dermatol. 2004;122:812–819. doi: 10.1111/j.0022-202X.2003.12602.x. [DOI] [PubMed] [Google Scholar]

Articles from Dermato-endocrinology are provided here courtesy of Taylor & Francis

RESOURCES