您的位置: 首页 > 2025年3月 第2卷 第1期 > 文字全文
2023年7月 第38卷 第7期11
目录

A review of corneal nerve and limbal stem cell

A review of corneal nerve and limbal stem cell

来源期刊: Eye Science | 2025年3月 第2卷 第1期 68-84 发布时间:2025-02-25 收稿时间:2025/4/9 11:47:37 阅读量:108
作者:
关键词:
CorneaLimbal epithelial stem cellsLESCs Cornea nerve Biological molecules
CorneaLimbal epithelial stem cellsLESCs Cornea nerve Biological molecules
DOI:
10.12419/es24071605
Received date:
2024-02-24 
Revised date:
2024-03-11 
Accepted date:
2024-11-16 
Published online:
2025-02-25 

The cornea is a transparent tissue that serves as the main refractive element of the eye ball.Limbal epithelial stem cells(LESCs), residing in the basal epithelial layer of the Palisades of Vogt located in the corneal limbus between cornea and scleral, are believed to be crucial for the continuous turnover of the corneal epithelium. The proliferation, migration, and differentiation of the LESCs are modulated by unique physical and chemical futures contained within the microenvironment known as the limbal niche. This niche, composed of nerve terminals, cells, extracellular matrix, vasculature, and signaling molecules, is the home for processe such as proliferation, migration and differentiation. Corneal nerve terminals possess special anatomical structures in the limbal region and basal epithelial cells, and they demonstrate pivotal biological effects in the regulation of the LESC function and corneal epithelium homeostasis. Biological molecules such as neuropeptides, neurotransmitters, and neurotrophic factors play a crucial role in modulating the LESCs phenotype responsible for corneal epithelium homeostasis. This paper will review recent studies on how  these nerve derived molecules function in this process and provide clear orientations for future research. 
The cornea is a transparent tissue that serves as the main refractive element of the eye ball.Limbal epithelial stem cells(LESCs), residing in the basal epithelial layer of the Palisades of Vogt located in the corneal limbus between cornea and scleral, are believed to be crucial for the continuous turnover of the corneal epithelium. The proliferation, migration, and differentiation of the LESCs are modulated by unique physical and chemical futures contained within the microenvironment known as the limbal niche. This niche, composed of nerve terminals, cells, extracellular matrix, vasculature, and signaling molecules, is the home for processe such as proliferation, migration and differentiation. Corneal nerve terminals possess special anatomical structures in the limbal region and basal epithelial cells, and they demonstrate pivotal biological effects in the regulation of the LESC function and corneal epithelium homeostasis. Biological molecules such as neuropeptides, neurotransmitters, and neurotrophic factors play a crucial role in modulating the LESCs phenotype responsible for corneal epithelium homeostasis. This paper will review recent studies on how  these nerve derived molecules function in this process and provide clear orientations for future research. 

HIGHTLIGHTS

1.Critical Discoveries and Outcomes

The anatomical structure and biological functions of corneal nerves and limbal stem cells are described, and the biological role of nerve-derived molecules is summarized. How corneal nerves play a role in regulating corneal epithelial homeostasis is intended to provide clinical feasibility for future related applications.

2.Methodological Innovations

Limbal epithelial stem cells (LESCs), residing in the basal epithelial layer of the Palisades of Vogt found in the corneal limbus  located between cornea and scleral, Corneal nerve terminals possess special anatomical structures in the limbal region and basal epithelial cells, and they demonstrate pivotal biological effects in the regulation of the LESC function and corneal epithelium homeostasis.

3. Prospective Applications and Future Directions

Biological molecules such as neuropeptides, neurotransmitters, and neurotrophic factors play a crucial role in modulating the LESCs phenotype responsible for corneal epithelium homeostasis.

The cornea is a transparent tissue that serves as the main refractive element of the eye ball, and consists of five layers:  the epithelium, Bowman’s layer, stroma, Descemet’s membrane, and endothelium[1,2]. Limbal epithelial stem cells (LESCs), residing in the basal epithelial layer of the Palisades of Vogt found in the corneal limbus  located between cornea and scleral,  are believed to be crucial for the continuously turnover of corneal epithelium[3-5]. The proliferation, migration, and differentiation of the LESCs are modulated by unique physical and chemical futures contained within the microenvironment known as limbal niche, which composed of nerve terminals, cells, extracellular matrix, vasculature and signaling molecules[5,6]. The maintenance of LESCs and corneal epithelial homeostasis has been found to be orchestrated by core transcription regulatory circuitries (CRCs), with RUNX1 and SMAD3 being required for the maintenance of corneal epithelial identity and homeostasis[7]. Current data suggest that LESCs may comprise two coexisting populations, termed the “outer” and “inner” limbus, localized in separate and well-defined sub-compartments[8]. The primitive population of quiescent outer LESCs participates in wound healing and boundary formation and is  regulated by T cells, which serve as a niche. In contrast, the inner peri-corneal limbus hosts active LESCs that maintain corneal epithelial homeostasis[9].Another study reported that corneal stem cells  are not only located in the limbus but also distributed throughout the entire ocular surface in other mammals[10]. However, this does not explain the phenomena that damage to limbal area, such as chemical burn, surgeries, and contact lens use, leading to the limbal stem cell deficiency (LSCD), usually results in the invasion of conjunctival epithelium onto the corneal surface and corneal neovascularization in human[11]. Therefore, most researchers still focus on the view that corneal epithelial stem cells  reside in the limbal area.

LESCs are considered to be a group of cells exhibiting the special characteristics of mitotic inactivity and high proliferative potential, which is activated during the transient amplifying stage. They divide asymmetrically to produce one stem cells and one transient amplifying cell ( TAC, a cell capable of multiple but limited cellular division) to  maintain the stem cell population. TACs migrate to the central cornea along the basement membrane and responsible for the turnover of the epithelium[12]. The balance of this process is modulated within the limbal niche. Most research groups have focused on the studying cells (e.g. mesenchymal stem cells), extracellular matrix, and biological factors (e.g. hemoderived factors, soluble factors/cocktails) that function to maintain the LESCs population and reconstruct the limbal niche based on these theories[13-19]. However, increasing evidences has demonstrated that nerve endings play an important role in the regulation of the epithelial stem cell niche[20-22].

Corneal nerves plays a critical role in the regulation of corneal epithelial homeostasis by  stimulating  proliferation, regeneration, differentiation, and possibly migration[23]. Malfunction of nerves, such as in diabetes mellitus, ocular herpes simplex, neoplasia, and ophthalmic surgery, typically leads to the neurotropic keratopathy, which exhibits symptoms including persistent corneal epithelial defects, stromal opacification, stromal melting, and neovascularization. This phenomenon has been demonstrated through a series of  experimental models involving cornea denervation[24-26]. Furthermore,  growing evidence has shown  a close correlation between corneal basal cell density (BCD) and nerve parameters in  models of LSCD. Increasing severity of LSCD is accompanied by a decline in the density of the central corneal sub-basal nerve plexus density (SND)[27-29]. Moreover, in an animal model of ocular denervation, the number and function of corneal stem/progenitor cells were significantly reduced[30,31]. Biological molecules, such as neuropeptides, neurotransmitters and neurotrophic factors, play pivotal role in modulating the phenotype of LESCs, which are responsible for corneal epithelium homeostasis. This paper will review recent studies on how does these nerve-derived molecules function in this process and provide clear directions for future research.

ANATOMY OF LESCs NICHE AND CORNEA NERVE

 LESCs niche and their biological functions

LESCs are considered to reside in the special area of  the limbus, defined as the limbal palisades of Vogt (POV) . Within this area, numerous limbal epithelial crypts (LECs) are found, which extend radially from the limbal palisades into the conjunctival stroma[32]. As reported, LECs were uniformly distributed around the corneal circumference and are considered to be the potential stem cell repositories[33]. The anatomy of POV and LECs, together with cellular elements (e.g. immune cells, mesenchymal stem cells, and melanocytes), vascular and nerve terminals,  extracellular matrix (ECM), and signaling molecules, comprises a niche that is related to the maintenance of the stem cell functions for corneal epithelial homeostasis[34,35]. The process of corneal epithelium turnover can be summarized as follows: LESC located in the basal epithelial layer of LEC divide symmetrically into two identical cells or asymmetrically into one LESC and a TAC; then, the TACs are divided into post-mitotic cells (PMCs) as they migrate centripetally and  differentiate into terminally differentiated cells (TDCs)[36](Fig 1).

Figure 1 Hypothetical scheme of limbal stem cell niche[36]

Fig 1 Hypothetical scheme of limbal stem cell niche36

TDC, terminally differentiated cell; PMC, post-mitotic cells; TAC, transient amplifying cell; LESC, limbal epithelial stem cell; MSC, mesenchymal stem cell.

 Nerves in the cornea

The cornea is innervated by both the sensory and autonomic nervous systems. The human cornea receives most of its sensory innervation from two or three long ciliary nerves, which originate from the nasociliary nerve, a branch of the first (ophthalmic) division of the trigeminal nerve. Before reaching the corneoscleral limbus, nerves fibers repeatedly bundle  together and anastomose extensively with branches of the short ciliary nerves, distributing uniformly around the corneal circumference and entering the cornea radially from all directions[37,38].Upon entering the corneoscleral limbus, the parent nerve gives rise to highly tortuous nerves that ultimately terminate in CNE, which are  encapsulated compact structure often referred to as “receptors” or “ corpuscles” (Fig 2).The CNEs are predominantly arranged in clusters, located in a close proximity to the limbal POV, and completely surrounded by limbal epithelial crypts (LECs), which are considered as stem cell niches (Fig 3)[21]. This interesting and unique  structure suggests that CNE may play a critical role in the  maintenance of limbal stem cell niche.

Figure 2 The whole-mount acetylcholinesterase-stained cornea

Fig 2 The whole-mount acetylcholinesterase-stained cornea

(A,B) CNE (black arrows) distributed around limbal POV (arrowheads). (C) Nerve fibres in the stroma (arrowheads). White arrows point to CNE in the peripheral cornea. (D) The deep stroma nerve gives rise to tortuous nerve branches (arrows). The tortuous nerves terminate in CNE as seen in C. Bar=100 μm (A,B), 500 μm (C), 100 μm (D), 10 μm (E), 25 μm (F), 120 μm (G) and 100 μm (H) [21]

Figure 3 Acetylcholinesterase prestained sections of limbus demonstrating CNE (arrows and arrowheads) completely wrapped by LECs. Bar=50 μm (A–D) [21]

Fig 3 Acetylcholinesterase prestained sections of limbus demonstrating CNE (arrows and arrowheads) completely wrapped by LECs. Bar=50 μm (A–D) 21
Next, the stromal nerve bundle extends into the cornea stroma and gives rise to small stromal nerves that anastomosed frequently to form a moderately dense plexus, occupying approximately the anterior one-half of the peripheral stroma and one-third of the central stroma. These nerve fibers branch into the narrow band of the anterior stroma, penetrate the Bowman’s membrane, and give off sub-basal nerves that coursed parallel to the ocular surface and below the basal epithelium, known as intraepithelial corneal basal nerves(ICBNs)[39]. Notably, the density and length of ICBN fibers steadily decline with age[40,41]. Interestingly, most of the perforation sites are observed in the mid-peripheral corneal compared to the central and peripheral zones, and characterized by bulb-like thickenings from which the sub-basal nerves arose[42].The direction of the sub-basal nerves immediately after penetrating  Bowman’s membrane is mainly vertical, and then spread to form a spiral-like assemblage of long, curvilinear or vortex-like patterns, with a whorl-like arrangement when viewed in its entirety[43,44]. This observation aligns with the hypothesis that the migration of  basal epithelial cells from the limbus to central cornea in a whorl-like fashion is directed by chemotropic molecules or electromagnetic cues, which may be related with the nerves[45-47]. There is conclusive visual evidence to support the existence of a physical and anatomical association  between nerves and epithelia within the cornea[41]. The intraepithelial nerve terminals, which branch from the sub-basal nerves distributed abundantly throughout all layers of the corneal epithelium, and with special structures of “bulbous nerve endings” in the basal epithelial cell layers[48] (Fig 4) . Moreover, nerve terminals in the basal epithelial cell layers generally extended parallelly to the parent sub-basal nerves fibers, and with relatively infrequent branches.

Figure 4. Subbasal nerves in the peripheral cornea. Arrows, bulbous nerve endings in the basal epithelial cell layer[48]

Fig 4. Subbasal nerves in the peripheral cornea. Arrows, bulbous nerve endings in the basal epithelial cell layer48
In addition to sensory nerves, the mammalian cornea also receives autonomic nerves, including sympathetic nerve fibers originating from the superior cervical ganglion and the parasympathetic fibers originating from the ciliary ganglion[22]. However, there are significant differences in the density of  autonomic innervation among different species[49].

NERVE DERIVED BIOLOGICAL MOLECULES

Substance P

Substance P (SP), an 11-amino acid peptide encoded by the Tachykinin Precursor 1 (TAC1) gene, belongs to the tachykinin neuropeptide family and is highly expressed in both the peripheral and central nervous systems. It is secreted by  specific neuronal cells as well as non-neuronal cell type, such as  epithelial cells and immune cells, and exerts its functions by binding to the G protein-coupled neurokinin receptors (NKRs), which are expressed by many cell types in the body. There are three types of NKRs: NK1R, NK2R, and NK3R. Among members of the NKR family, neurokinin receptor 1(NK1R) has the highest affinity for SP[50], and is expressed in corneal epithelial cells and keratocytes[51]. The SP-NK1R signaling pathway is believed to mediate a diverse set of pathways, including those involved in corneal neovascularization, inflammation,cell proliferation, migration, apoptosis, stem cell mobilization, and has profound implications for wound healing and inflammatory modulation[50]. NK1R is proven to be expressed on the cell membrane of corneal epithelial stem cells, supporting the key role of SP and nerves in stem cell pathophysiology. It is reported that SP ablation or NK1R blockade significantly enhances epithelial wound healing and corneal transparency[52]. Additionally, excessive expression of SP can induce senescence and exhaustion of residual stem cells through activation of NK1R, which is associated with LSCD. SP aggravates corneal LSCD through stimulation of the mammalian target of rapamycin(mTOR) pathway, eventually leading to accelerated corneal epithelial cell senescence. It is identified that the mTOR signaling, as a key driver of stem cell activity, participates in corneal epithelial cell proliferation and differentiation[53].It is widely reported that SP is a key mediator of neurogenic inflammation through the enhancement of the microvascular permeability, vasodilatation, and plasma extravasation[54]. However,  SP plays an indispensable role during wound healing process by functioning as an injury-inducible messenger[55] and exerts an activity in maintaining corneal epithelium homeostasis[56]. It has been revealed that the effect of SP on epithelial wound closure is time and concentration dependent[57]. In addition, Romina hypothesized that an early, controlled release of SP is beneficial, while excessive amounts may impair healing[58]. The motility of many cells types is enhanced by SP through different signaling pathways, such as mitogen-activated protein kinases (MAPKs), phosphoinositide 3-kinase-Akt, protein kinase C and epidermal growth factor receptor (EGFR) pathways[56,59]. In a diabetic model, SP was found to promote corneal epithelial wound healing by stimulating the reactivation of Akt, EGFR and Sirt1,thereby alleviating the epithelial lesions[56].The corneal sensitivity of diabetic mice was also restored by the activity of SP-NK1R signaling pathway, which was compatible with the decrease in the corneal nerve terminal density observed in NK1R knockout mouse[60]. Unlike the diabetic model, SP needed to act synergistically with insulin-like growth factor-1(IGF-1) to promote corneal epithelial defect closure. This combined effect of SP/IGF-1 was mediated by NK-1R, but not by NK2R and NK3R[61].  Moreover, the SP-NK1R interaction has been shown to inhibit the apoptosis of the corneal epithelial cells in vitro[62]as well as regulate the expression of E-cadherin and ZO-1 tight junction proteins in the corneal epithelium[63,64], and exhibited an important role in the maintenance of the corneal epithelium homeostasis.

Several reports have demonstrated that SP can enhance  corneal wound healing both by regulating the proliferation, migration and differentiation of LESCs as well as the mobilization of the MSCs[64-68]. Furthermore, additional studies should be conducted to evaluate this hypothesis.

Calcitonin gene-related peptide(CGRP)  

CGRP, a 37-amino-acid neuropeptide expressed by trigeminal ganglion neurons and corneal nerves, is widely distributed in the nervous system alongside  SP. Previous studies have investigated the expression of CGRP in the cornea sensory nerve across in a wide range of animal species. CGRP plays a crucial role in immune reactions by regulating immune cells such as macrophages, neutrophils, dendritic cells[69]. In a study of corneal  inflammation induced by Pseudomonas aeruginosa (P. aeruginosa), corneal sensory neuron activation, macrophage recruitment, and up-expression of CGRP  were observed. The synergistic effect between sensory neurons and macrophages promotes the release of CGRP. CGRP inhibits corneal inflammation and promotes the transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway[70].

After being applied topically to the ocular surface in a corneal epithelial  wound model, CGRP was found to enhance the healing rate of corneal epithelial cells through a cAMP-dependent effect on  actin filament polymerization, which is involved in the cell migration[71]. Recent research has demonstrated the CGRP derived from trigeminal ganglion sensory neurons and corneal nerves  promotes corneal angiogenesis. Moreover, blocking the signaling of nerve-derived CGRP inhibits corneal angiogenesis[72]. Paradoxically, in a model of corneal injury induced by SM, the expression of CGRP increased while the number of limbal stem cell declined and nerve  damage occurred. The report speculated that CGRP might be responsible for changes in the LESC niche , as it has been shown to interact with corneal epithelial cells in vitro to release interleukin-8, which acts as a chemoattractant  for neutrophils into the inflammation area, a process related to the “neurogenic inflammation” [73]. In a study of corneal injury induced by lacrimal gland excision, the expression of CGRP was observed in  isolectin B4 (IB4) -positive corneal neurons. This response may initially facilitate axonal regeneration;however, the increased expression of CGRP is also likely to  contribute to persistent corneal pain[74]. Furthermore, CGRP’s role in neuroplasticity and hypersensitivity explains its close association with photophobia, a common symptom in  dry eye disease , migraine and traumatic brain injury[75].  Further data should be collected to evaluate the precise role that CGRP plays in maintenance corneal epithelial homeostasis.

Melanocyte-stimulating hormones (MSH)

MSH,a family of peptide hormones and neuropeptides consisting of α-MSH, β-MSH, and γ-MSH, is generated from different cleavages of the proopiomelanocortin(POMC) protein. It is mainly secreted by melanocytes, neurons or anterior lobe of the pituitary gland, and functions to stimulate the production and release of melanin in skin and hair, suppress appetite, and contribute to sexual arousal in the hypothalamus[76]. α-MSH, a 13-amino-acid peptide, is  widely distributed in the skin and cornea[77]. It is also well-known for its roles in metabolic regulation, neuroprotection, inflammation suppression, and anti-angiogenic effects[78-81], mediated through binding to the melanocortin receptors, which consist of 5 subtypes (MC1R-MC5R) belonging to the G protein-coupled receptor(GPCR) family[76].In a study of scopolamine-induced  dry eye  syndrome,  α-MSH was found to restore ocular surface functions by upregulating EGFR expression in the JAK-STAT signaling pathway[77].

Melanocytes,  one of the most important cells in the LESC niche, are responsible for  pigmentation in the perilimbal area, constructing an anti-oxidant system for LESC protection[82,83]. The α-MSH/MC1R signaling pathway plays vital roles in bulge melanocyte stem cells and melanin synthesis[84]. Additionally, α-MSH/MC1R signaling is critical for corneal endothelial cell  function and graft survival after corneal transplantation[85]. Therefore, corneal nerve might play a role in the LESC niche environment through the regulation of α-MSH/MC1R signaling pathway.

Acetylcholine (ACh)

Acetylcholine, a choline molecule  acetylated at the oxygen atom, is synthesized mainly in certain neurons by the enzyme choline acetyltransferase from the compounds choline and acetyl-CoA. It is regarded as a classical neurotransmitter that is widely distributed in central nervous system, neuromuscular junctions, and autonomic nervous system[86]. However, increasing evidence has demonstrated that ACh is also synthesized by a majority of human cells, such as cornea epithelium[87], and exerts its functions in cellular process (e.g. proliferation, differentiation, adhesion, migration, secretion, etc) , anti-inflammation, and stimulating wound healing (e.g. skin, cornea) by binding to nicotinic and muscarinic receptors(nAChRs and mAChRs, respectively)[88-92]. nAChRs on nerve endings in the nose initiate a reflex arc resulting in instantaneous tear secretion, which represents a novel approach to treat dry eye disease by increasing endogenous tear production[93,94]. Furthermore, the M3 receptor plays a pivotal role in tear production, and its absence leads to ocular surface changes typical of dry eye disease in advanced age[95].

nAChRs (α1-10, β1-4, γ, δ and ε subunits) and mAChRs (M1-M5) have been found to be expressed in many types of cells, including corneal epithelial cells. The nAChRs are directly linked to icon channels that mediate the influx of Na+ and Ca2+ and efflux of K+, and they can also activate the downstream signaling pathways by modulating the activities of protein kinases and phosphatases[96]. Activation of the nAChR and mAChRs pathways promotes epithelial cell division and re-epithelialization after corneal abrasion[97,98]. This biological function, mediated by the cytotransmitter ACh-based signal transduction, is not only regulated by the cholinergic nervous system, but also by the autocrine/paracrine in the corneal epithelium[99]. Nevertheless, the role of ACh and its downstream signaling pathways  in the process of corneal epithelium re-epithelialization remains unknown. The processes and mechanisms of cholinergic modulations in stem cells are  complex, and further studies are needed to  elucidate the mechanisms underlying this biological function and to determine whether it plays a role in regulating corneal epithelial stem cell functions and corneal epithelium homeostasis.

Catecholamines

Catecholamines, including epinephrine (E), norepinephrine (NE)and dopamine(DA), are primarily synthesized by the chromaffin cells of the adrenal medulla and the postganglionic fibers of the sympathetic nervous system. These compounds originate from the amino acid tyrosine. The catecholamine-secreting cells initially convert tyrosine to L-Dihydroxyphenylalanine(L-DOPA), which is subsequently transformed into DA. DA is then further metabolized into  NE by dopamine β-hydroxylase or into E by phenylethanolamine N-methyltransferase[100]. NE and E bind to the 7-transmembrane, G protein-coupled receptors known as adrenoceptors, which comprise three major types: α1, α2 and β1-3. Adrenoceptors are widely expressed in both the central nervous system and peripheral tissues, and they exert their functions on cellar processes primarily through cAMP-PKA downstream pathways.

In the cornea, catecholamines and adrenoceptors are believed to play a role in the proliferation of corneal epithelial cells. NE, the principal neurotransmitter produced and released by sympathetic nerves, is significantly more abundant than E in rabbit and human corneas, while DA is found to be the most prevalent catecholamine in the central and intermediate segments of the cornea[101]. It has been reported that NE is directly or indirectly associated with the onset and exacerbation of  corneal inflammation[58,102,103]. Human corneal limbal epithelial cell sheets cultured on carboxymethyl cellulose (CMC)-DA-coated substrates have been  safely transplanted in a rabbit animal model of LSCD[104]. Notably, there is no evidence to suggest  that the E is present in the human cornea. The α2 adrenoceptors are primarily located in the intercellular spaces of the epithelial basal layer, particularly in the peripheral anterior cornea[105]. The concentration gradient of NE decreases from the periphery to the center of the cornea[101]. Both E and α2 adrenoceptor exert their effects on corneal epithelial cells by regulating the cAMP-PKA pathway, which plays a crucial role in corneal wound healing and overall homeostasis[106,107]. Furthermore, activating the DA receptors (D1/D2) expressed in corneal epithelial cells and endothelial cells in the wounded cornea increases the nerve growth factor (NGF) and corneal nerve density, thereby enhancing corneal epithelial healing[108,109]. This suggests that DA may have indirect function in maintaining corneal epithelium homeostasis through the NGF signaling pathways.

Ciliary neurotrophic factor (CNTF)

CNTF is a protein  encoded by the CNTF gene in humans. It belongs to the interleukin 6(IL-6) family cytokines, which includes IL-6, IL-11, CNTF, leukemia inhibitory factor (LIF), oncostatin M (OSM), cardiotrophin 1 (CT-1), cardiotrophin-like cytokine (CLC), and IL-27[110]. When induced pluripotent stem cells(iPSCs) respond to the electrical activity of their microenvironment, they increase endogenous CNTF release to enhance neuronal differentiation and facilitate the rapid conversion of  iPSCs. CNTF can induce adipose-derived mesenchymal stem cells (AMSCs) to exit the cell cycle, undergo mitosis, and initiate the expression of neuron markers[111]. CNTF has been shown to improve the generation of cornea nerve fibers[112], but both CNTF and its receptor,  CNTFRα,  are hardly expressed in normal human cornea[113,114]. Interestingly, in mouse models, CNTF has been found to improve  corneal epithelial wound healing and promote the proliferation of corneal epithelial stem/progenitor cells by the activation of STAT3 signaling pathway,  a transcription factor that regulates cellular processes including apoptosis, proliferation, migration, and survival[115]. However, the mechanism underlying this biological function, how CNTF regulates the STAT3 signaling pathway and whether there are any differences in CNTF/ CNTFR expression in cornea epithelium among different species remains unknown.    

Nerve growth factor (NGF)

NGF is a well-known neurotrophic factor and neuropeptide taht is widely distributed in the nervous system and major cell types throughout the body. It plays an important role in development and survival of sensory neurons[116], as well as in the cellular process such as proliferation, differentiation, survival, and apoptosis. It also regulates  the immune system[117], accelerates wound healing in the skin and cornea[118,119], and exhibits insulinotropic, angiogenic[120], and antioxidant properties[121].

NGF has been found to orgiginate from both corneal nerves and limbal basal epithelium, where it promotes sustained corneal epithelium wound healing, the recovery of corneal sensitivity and cornea nerve regeneration[122,123]. As a rhNGF, Cenegermin has shown beneficial effects on neurotrophic keratopathy, with effects persisting several months after  an 8-week course of  topical rhNGF treatment[124]. NGF has been found to extend the lifespan of LESCs in vitro, promoting their proliferation, colony-forming efficiency, and maintenance of   phenotype ( as evidenced by ΔNp63α and ABCG2 expression)[1]. The NGF/TrkA signaling pathway promotes the migration and proliferation of limbal epithelial cells by activating  various signaling pathways, including phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (Akt), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (Erk), and phospholiase C-γ (PLC-γ)[125].Therefore, NGF is likely to play an important role in regulating the LESC niche. Recently, topical recombinant human nerve growth factor (rhNGF) has been  demonstrated to be a safe and effective agent for promoting the healing of moderate-to-severe neurotrophic keratitis[126,127]. The expression of microRNAs  in human epithelial corneal cells after time-dependent rhNGF treatment has also been analyzed, revealing a potential strong impact of miRNAs  on the modulation of specific NGF-induced cellular responses[128]. Hsa-miR-143-3p has been shown to play a role in the maintenance of CESCs through downregulating of key proteins involved in Wnt and MAPK signaling[129]. However, the mechanism by which NGF regulates the LESC phenotype requires further investigation.

 Melanocyte-stimulating hormones (MSH)

MSH,a family of peptide hormones and neuropeptides comprising α-MSH, β-MSH, and γ-MSH, originates from different cleavages of the proopiomelanocortin(POMC) protein. It is mainly secreted by melanocytes, neurons or the anterior lobe of the pituitary gland, and  it functions to stimulate the production and release of melanin in skin and hair, suppress appetite, and contribute to sexual arousal while in the hypothalamus[76]. α-MSH, a 13-amino-acid peptide, is found widely distributed in skin and cornea[77]. It is also well-recognized for its roles in metabolic regulation, neuroprotection, inflammation suppression, and anti-angiogenic activities[78-81], mediated through binding to the melanocortin receptors, which consist of 5 subtypes (MC1R-MC5R) belonging to the G protein-coupled receptor(GPCR) family[76].In a study of scopolamine-induced  dry eye  syndrome,  α-MSH restored ocular surface functions by upregulating EGFR expression via the JAK-STAT signaling pathway[77].

Melanocytes,  one of the most important LESC niche cells, are responsible for the pigmentation in POV, thereby establishing  an antioxidant system for LESC protection[82,83]. The α-MSH/MC1R signaling pathway plays vital roles in bulge melanocyte stem cells and melanin synthesis[84]. Furthermore, α-MSH/MC1R signaling is critical for corneal endothelial cells  function and graft survival after corneal transplantation[85]. Thus, corneal nerve might play a role in LESC niche environment through the regulation of α-MSH/MC1R signaling pathway.

A brief introduction, expression site, and possible regulatory role of the seven nerve-derived biological molecules mentioned above have been summarized in Table 1 for reference.

Table 1 A brief introduction, expression site, and possible regulatory role of the seven nerve derived biological molecules

Nerve derived

 biological molecules

Brief introduction

Expression site

Possible regulatory role

Substance P

11-aminoacid peptid,belongs to the tachykinin neuropeptide family

highly expressed in the peripheral and central nervous systems

SP-NK1R signal pathway:involved in corneal neovascularization, inflammation,cell proliferation, migration, apoptosis, stem cells mobilization and wound healing and inflammatory modulation.

mTOR pathway:accelerated corneal epithelial cell senescence.

Akt, EGFR and Sirt1 pathway:promoted the corneal epithelial wound healing,alleviated the epithelial lesions.

SP/IGF-1:promoted corneal epithelial defect closure.

Nanog and Wnt/ β-catenin signaling pathway:improve the corneal wound healing both by regulating the proliferation,migration and differentiation of LESCs and the mobilization of the MSCs.

 

Calcitoningene-related peptide(CGRP)

37-amino-acid neuropeptide

widely distributed in the nervous system in accordance with SP

PI3K/Akt and p38MAPK signaling pathway:homed of human umbilical cord mesenchymal stem cells (HUMSCs) ,inhibited corneal inflammation and promoted the transformation of macrophages to the M2 phenotype.

nerve-derived CGRP signaling pathway:corneal angiogenesis.

Melanocyte-stimulating hormones (MSH)

 


a family of peptide hormones and neuropeptides consisting of α-MSH, β-MSH, and γ-MSH, is generated from different cleavages of the proopiomelanocortin(POMC) protein
.

widely distributed in skin and cornea

JAK-STAT signaling pathway:restored ocular surface functions by upregulating EGFR expression.

α-MSH/MC1R signaling pathway:critical for corneal endothelial cells function and graft survival after corneal transplantation.

Acetylcholine (ACh)

 

choline molecule that has been acetylated at the oxygen atom,synthesized mainly in certain neurons by the enzyme choline acetyltransferase from the compounds choline and acetyl-CoA

widely distributed in central nervous system, neuromuscular and autonomic nervous system

nAChRs directly linked to icon channels:mediate the influx of Na+ and Ca2+ and efflux of K+.

downstream signaling pathway:activated by modulating the activities of protein kinases and phosphatases.

M1,M3 and M5 receptors acoupled with Gq proteins:activated protein kinase C (PKC) by elevating intracellular Ca2+ and diacylglycerol.

M2,M4 receptors coupled with Gi/o proteins: inhibited protein kinase A by diminishing adenylyl cyclase activity.

nAChR and mAChRs pathway:promoted the epithelial cells division and re-epithelialization after corneal abrasion,modulating the intestinal crypt stem cells proliferation and differentiation by acting on intracellular pathways such as Wnt or trans-activate EGER.

Catecholamines

including epinephrine (E), norepinephrine (NE)and dopamine(DA), are produced mainly by the chromaffin cells of the adrenal medulla and the postganglionic fibers of the sympathetic nervous system

 

largely expressed both in the central nervous system and in peripheral tissues

cAMP-PKA downstream pathway:exerted their functions on cell process,corneal wound healing and overall homeostasis.

NDF signaling pathway:ctivated the DA receptor (D1/D2) that expressed in corneal epithelial cells and endothelial cells in the wounded cornea increased the NDF impression and corneal nerve density.

Ciliary neurotrophic factor(CNTF)

protein that in humans encoded by the CNTF gene,belongs to the interleukin 6(IL-6) family cytokines consisting of IL-6, IL-11, CNTF, leukemia inhibitory factor (LIF), oncostatin M (OSM), cardiotrophin 1 (CT-1), cardiotrophin-like cytokine (CLC), and IL-27

 widely distributed in neurons, glial and Schwann cells throughout the central and peripheral nervous system

induced pluripotent stem cells(iPSCs)’s response:increase endogenous CNTF release to enhance neuronal differentiation and the iPSCs’ rapid conversion

CNTFRα:permitting the recruitment of glycoprotein 130 kDa(gp130) and LIF receptor membrane spanning signal transducing units

CNTF:improve the corneal epithelial wound healing, and promote the proliferation of corneal epithelial stem/progenitor cells

 Nerve growth factor (NGF)

neurotrophic factor and neuropeptide

widely distributed in nervous system and major groups of cells in the body

NGF/TrkA signal pathway:promote the migration and proliferation of limbal epithelial cells by inducing the activation of various signaling pathways, including phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (Akt) and mitogen activated protein kinase (MAPK)/extracellular signal-regulated kinase (Erk) and phospholiase C-γ (PLC-γ)

p75NTR:bind to and increase the affinity of TrkA for NGF as well as modulating its downstream signaling pathways,and downregulated upon the LESCs differentiation

Wnt and MAPK signaling pathway:Hsa-miR-143-3p in the maintenance of CESCs through downregulation of key proteins involved in Wnt and MAPK signaling

SUMMARY AND CONCLUSION

Corneal epithelium homeostasis is regulated by a complex biological process. Although some researchers suggest that corneal epithelial stem cells reside the entire ocular surface, most studies indicate that LESCs are responsible for the turnover of  corneal epithelium. The niche in the limbal POV is the site where LESCs undergo processes such as proliferation, migration and differentiation. LESCs may comprise at least two types of cells: one that remains quiescent during corneal epithelial wounding, and another that becomes proliferative in response to such wounding. Corneal nerve terminals possess unique  anatomy structures in the limbal POV and basal epithelial cells, and they exert crucial biological effects in the regulation of the LESC function and corneal epithelium homeostasis. A major group of neuro-derived biological molecules participate in this process through indirect modulation of the niche environment or direct regulation of LESCs functions.Substance P acts through the SP-NK1R signaling pathway, mTOR pathway, Akt, EGFR, and Sirt1 pathways, as well as the Nanog and Wnt/β-catenin signaling pathways. Calcitonin gene-related peptide (CGRP) is involved in the PI3K/Akt and p38 MAPK signaling pathways, as well as in nerve-derived CGRP signaling pathways. Melanocyte-stimulating hormones (MSH) function through the JAK-STAT signaling pathway and the α-MSH/MC1R signaling pathway. Acetylcholine (ACh) operates through nAChRs, which are directly linked to ion channels, and downstream signaling pathways, including M1, M3, and M5 receptors coupled with Gq proteins, and M2 and M4 receptors coupled with Gi/o proteins. Additionally, the nAChR and mAChRs pathways are involved. Catecholamines function through the cAMP-PKA downstream pathway and the NDF signaling pathway. Ciliary neurotrophic factor (CNTF) induces a response in induced pluripotent stem cells (iPSCs) through CNTFRα and CNTF. Finally, nerve growth factor (NGF) operates through the NGF/TrkA signaling pathway, p75NTR, and the Wnt and MAPK signaling pathways.Some of these neuro-derived molecules have been used clinically. For example, rhNGF has been successfully used for treatment of neurotrophic keratitis. Moreover, elucidating how corneal nerve exert their specific functions in regulating  corneal epithelium homeostasis, especially in modulation of LESCs process, is important for the reconstruction of LESC niche and ocular surface. This review may provide some directions for future investigations on this topic and establish their clinical feasibility.  

Correction notice

None

Acknowledgements

None

Author Contributions

(I) Conception and design: Liqun Chen
(II) Administrative support: Xianglong Yi
(III) Provision of study materials or patients:  : Liqun Chen
(IV) Collection and assembly of data: : Liqun Chen
(V) Data analysis and interpretation: : Liqun Chen
(VI) Manuscript writing: All authors
(VII) Final approval of manuscript: All authors

Funding

None

Conflict of Interests

None of the authors has any conflicts of interest to disclose.All authors have declared in the completed the ICMJE uniform disclosure form.

Patient consent for publication

None

Ethical Statement

None

Provenance and Peer Review

This article was a standard submission to our journal. The article has undergone peer review with our anonymous review system.

Data Availability Statement

None

Open Access Statement

This is an Open Access article distributed in accordance with the Creative Commons AttributionNonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license).
1、Kolli S, Bojic S, Ghareeb AE, et al. The role of nerve growth factor in maintaining proliferative capacity, colony-forming efficiency, and the limbal stem cell phenotype. Stem Cells. 2019, 37(1): 139-149. DOI: 10.1002/stem.2921. Kolli S, Bojic S, Ghareeb AE, et al. The role of nerve growth factor in maintaining proliferative capacity, colony-forming efficiency, and the limbal stem cell phenotype. Stem Cells. 2019, 37(1): 139-149. DOI: 10.1002/stem.2921.
2、Notara M, Lentzsch A, Coroneo M, et al. The role of limbal epithelial stem cells in regulating corneal (lymph)angiogenic privilege and the micromilieu of the limbal niche following UV exposure. Stem Cells Int. 2018, 2018: 8620172. DOI: 10.1155/2018/8620172. Notara M, Lentzsch A, Coroneo M, et al. The role of limbal epithelial stem cells in regulating corneal (lymph)angiogenic privilege and the micromilieu of the limbal niche following UV exposure. Stem Cells Int. 2018, 2018: 8620172. DOI: 10.1155/2018/8620172.
3、Moreno IY, Parsaie A, Gesteira TF, et al. Characterization of the limbal epithelial stem cell niche. Invest Ophthalmol Vis Sci. 2023, 64(13): 48. DOI: 10.1167/iovs.64.13.48. Moreno IY, Parsaie A, Gesteira TF, et al. Characterization of the limbal epithelial stem cell niche. Invest Ophthalmol Vis Sci. 2023, 64(13): 48. DOI: 10.1167/iovs.64.13.48.
4、Chuephanich P, Supiyaphun C, Aravena C, et al. Characterization of the corneal subbasal nerve plexus in limbal stem cell deficiency. Cornea. 2017, 36(3): 347-352. DOI: 10.1097/ico.0000000000001092. Chuephanich P, Supiyaphun C, Aravena C, et al. Characterization of the corneal subbasal nerve plexus in limbal stem cell deficiency. Cornea. 2017, 36(3): 347-352. DOI: 10.1097/ico.0000000000001092.
5、DelMonte DW, Kim T. Anatomy and physiology of the cornea. J Cataract Refract Surg. 2011, 37(3): 588-598. DOI: 10.1016/j.jcrs.2010.12.037.DelMonte DW, Kim T. Anatomy and physiology of the cornea. J Cataract Refract Surg. 2011, 37(3): 588-598. DOI: 10.1016/j.jcrs.2010.12.037.
6、Bonnet C, González S, Roberts JS, et al. Human limbal epithelial stem cell regulation, bioengineering and function. Prog Retin Eye Res. 2021, 85: 100956. DOI: 10.1016/j.preteyeres.2021.100956. Bonnet C, González S, Roberts JS, et al. Human limbal epithelial stem cell regulation, bioengineering and function. Prog Retin Eye Res. 2021, 85: 100956. DOI: 10.1016/j.preteyeres.2021.100956.
7、Li M, Huang H, Li L, et al. Core transcription regulatory circuitry orchestrates corneal epithelial homeostasis. Nat Commun. 2021, 12: 420. DOI: 10.1038/s41467-020-20713-z. Li M, Huang H, Li L, et al. Core transcription regulatory circuitry orchestrates corneal epithelial homeostasis. Nat Commun. 2021, 12: 420. DOI: 10.1038/s41467-020-20713-z.
8、Altshuler A, Amitai-Lange A, Tarazi N, et al. Discrete limbal epithelial stem cell populations mediate corneal homeostasis and wound healing. Cell Stem Cell. 2021, 28(7): 1248-1261.e8. DOI: 10.1016/j.stem.2021.04.003.Altshuler A, Amitai-Lange A, Tarazi N, et al. Discrete limbal epithelial stem cell populations mediate corneal homeostasis and wound healing. Cell Stem Cell. 2021, 28(7): 1248-1261.e8. DOI: 10.1016/j.stem.2021.04.003.
9、Song Z, Chen B, Tsai CH, et al. Differentiation trajectory of limbal stem and progenitor cells under normal homeostasis and upon corneal wounding. Cells. 2022, 11(13): 1983. DOI: 10.3390/cells11131983. Song Z, Chen B, Tsai CH, et al. Differentiation trajectory of limbal stem and progenitor cells under normal homeostasis and upon corneal wounding. Cells. 2022, 11(13): 1983. DOI: 10.3390/cells11131983.
10、Majo F, Rochat A, Nicolas M, et al. Oligopotent stem cells are distributed throughout the mammalian ocular surface. Nature. 2008, 456: 250-254. DOI: 10.1038/nature07406.Majo F, Rochat A, Nicolas M, et al. Oligopotent stem cells are distributed throughout the mammalian ocular surface. Nature. 2008, 456: 250-254. DOI: 10.1038/nature07406.
11、Moshirfar M, Masud M, Harvey DH, et al. The multifold etiologies of limbal stem cell deficiency: a comprehensive review on the etiologies and additional treatment options for limbal stem cell deficiency. J Clin Med. 2023, 12(13): 4418. DOI: 10.3390/jcm12134418. Moshirfar M, Masud M, Harvey DH, et al. The multifold etiologies of limbal stem cell deficiency: a comprehensive review on the etiologies and additional treatment options for limbal stem cell deficiency. J Clin Med. 2023, 12(13): 4418. DOI: 10.3390/jcm12134418.
12、Verma S, Lin X, Coulson-Thomas VJ. The potential reversible transition between stem cells and transient-amplifying cells: the limbal epithelial stem cell perspective. Cells. 2024, 13(9): 748. DOI: 10.3390/cells13090748. Verma S, Lin X, Coulson-Thomas VJ. The potential reversible transition between stem cells and transient-amplifying cells: the limbal epithelial stem cell perspective. Cells. 2024, 13(9): 748. DOI: 10.3390/cells13090748.
13、 Galindo S, de la Mata A, López-Paniagua M, et al. Subconjunctival injection of mesenchymal stem cells for corneal failure due to limbal stem cell deficiency: state of the art. Stem Cell Res Ther. 2021, 12(1): 60. DOI: 10.1186/s13287-020-02129-0. Galindo S, de la Mata A, López-Paniagua M, et al. Subconjunctival injection of mesenchymal stem cells for corneal failure due to limbal stem cell deficiency: state of the art. Stem Cell Res Ther. 2021, 12(1): 60. DOI: 10.1186/s13287-020-02129-0.
14、Mei H, Gonzalez S, Deng SX. Extracellular matrix is an important component of limbal stem cell niche. J Funct Biomater. 2012, 3(4): 879-894. DOI: 10.3390/jfb3040879. Mei H, Gonzalez S, Deng SX. Extracellular matrix is an important component of limbal stem cell niche. J Funct Biomater. 2012, 3(4): 879-894. DOI: 10.3390/jfb3040879.
15、Okumura Y, Inomata T, Fujimoto K, et al. Biological effects of stored platelet-rich plasma eye-drops in corneal wound healing. Br J Ophthalmol. 2023, 108(1): 37-44. DOI: 10.1136/bjo-2022-322068. Okumura Y, Inomata T, Fujimoto K, et al. Biological effects of stored platelet-rich plasma eye-drops in corneal wound healing. Br J Ophthalmol. 2023, 108(1): 37-44. DOI: 10.1136/bjo-2022-322068.
16、Chen J, Chen P, Backman LJ, et al. Ciliary neurotrophic factor promotes the migration of corneal epithelial stem/progenitor cells by up-regulation of MMPs through the phosphorylation of Akt. Sci Rep. 2016, 6: 25870. DOI: 10.1038/srep25870. Chen J, Chen P, Backman LJ, et al. Ciliary neurotrophic factor promotes the migration of corneal epithelial stem/progenitor cells by up-regulation of MMPs through the phosphorylation of Akt. Sci Rep. 2016, 6: 25870. DOI: 10.1038/srep25870.
17、Chen J, Lan J, Liu D, et al. Ascorbic acid promotes the stemness of corneal epithelial stem/progenitor cells and accelerates epithelial wound healing in the cornea. Stem Cells Transl Med. 2017, 6(5): 1356-1365. DOI: 10.1002/sctm.16-0441. Chen J, Lan J, Liu D, et al. Ascorbic acid promotes the stemness of corneal epithelial stem/progenitor cells and accelerates epithelial wound healing in the cornea. Stem Cells Transl Med. 2017, 6(5): 1356-1365. DOI: 10.1002/sctm.16-0441.
18、Eslani M, Putra I, Shen X, et al. Cornea-derived mesenchymal stromal cells therapeutically modulate macrophage immunophenotype and angiogenic function. Stem Cells. 2018, 36(5): 775-784. DOI: 10.1002/stem.2781. Eslani M, Putra I, Shen X, et al. Cornea-derived mesenchymal stromal cells therapeutically modulate macrophage immunophenotype and angiogenic function. Stem Cells. 2018, 36(5): 775-784. DOI: 10.1002/stem.2781.
19、Volatier T, Cursiefen C, Notara M. Current advances in corneal stromal stem cell biology and therapeutic applications. Cells. 2024, 13(2): 163. DOI: 10.3390/cells13020163.Volatier T, Cursiefen C, Notara M. Current advances in corneal stromal stem cell biology and therapeutic applications. Cells. 2024, 13(2): 163. DOI: 10.3390/cells13020163.
20、Huang S, Kuri P, Aubert Y, et al. Lgr6 marks epidermal stem cells with a nerve-dependent role in wound re-epithelialization. Cell Stem Cell. 2021, 28(9): 1582-1596.e6. DOI: 10.1016/j.stem.2021.05.007. Huang S, Kuri P, Aubert Y, et al. Lgr6 marks epidermal stem cells with a nerve-dependent role in wound re-epithelialization. Cell Stem Cell. 2021, 28(9): 1582-1596.e6. DOI: 10.1016/j.stem.2021.05.007.
21、Al-Aqaba MA, Anis FS, Mohammed I, et al. Nerve terminals at the human corneoscleral limbus. Br J Ophthalmol. 2018, 102(4): 556-561. DOI: 10.1136/bjophthalmol-2017-311146. Al-Aqaba MA, Anis FS, Mohammed I, et al. Nerve terminals at the human corneoscleral limbus. Br J Ophthalmol. 2018, 102(4): 556-561. DOI: 10.1136/bjophthalmol-2017-311146.
22、Al-Aqaba MA, Dhillon VK, et al. Corneal nerves in health and disease. Prog Retin Eye Res. 2019, 73: 100762. DOI: 10.1016/j.preteyeres.2019.05.003.Al-Aqaba MA, Dhillon VK, et al. Corneal nerves in health and disease. Prog Retin Eye Res. 2019, 73: 100762. DOI: 10.1016/j.preteyeres.2019.05.003.
23、Caro-Magdaleno M, Alfaro-Juárez A, Montero-Iruzubieta J, et al. In vivo confocal microscopy indicates an inverse relationship between the sub-basal corneal plexus and the conjunctivalisation in patients with limbal stem cell deficiency. Br J Ophthalmol. 2019, 103(3): 327-331. DOI: 10.1136/bjophthalmol-2017-311693.Caro-Magdaleno M, Alfaro-Juárez A, Montero-Iruzubieta J, et al. In vivo confocal microscopy indicates an inverse relationship between the sub-basal corneal plexus and the conjunctivalisation in patients with limbal stem cell deficiency. Br J Ophthalmol. 2019, 103(3): 327-331. DOI: 10.1136/bjophthalmol-2017-311693.
24、Khilji M, Tanveer S, Khan FZ, et al. Neurotrophic Keratopathy and Topical Insulin Therapy: A Case Report. Cureus. 2023, 15(9): e46242. DOI: 10.7759/cureus.46242.Khilji M, Tanveer S, Khan FZ, et al. Neurotrophic Keratopathy and Topical Insulin Therapy: A Case Report. Cureus. 2023, 15(9): e46242. DOI: 10.7759/cureus.46242.
25、NaPier E, Camacho M, McDevitt TF, et al. Neurotrophic keratopathy: current challenges and future prospects. Ann Med. 2022, 54(1): 666-673. DOI: 10.1080/07853890.2022.2045035. NaPier E, Camacho M, McDevitt TF, et al. Neurotrophic keratopathy: current challenges and future prospects. Ann Med. 2022, 54(1): 666-673. DOI: 10.1080/07853890.2022.2045035.
26、 Surico PL, Narimatsu A, Forouzanfar K, et al. Effects of diabetes mellitus on corneal immune cell activation and the development of keratopathy. Cells. 2024, 13(6): 532. DOI: 10.3390/cells13060532. Surico PL, Narimatsu A, Forouzanfar K, et al. Effects of diabetes mellitus on corneal immune cell activation and the development of keratopathy. Cells. 2024, 13(6): 532. DOI: 10.3390/cells13060532.
27、Bhattacharya P, Edwards K, Harkin D, et al. Central corneal basal cell density and nerve parameters in ocular surface disease and limbal stem cell deficiency: a review and meta-analysis. Br J Ophthalmol. 2020, 104(12): 1633-1639. DOI: 10.1136/bjophthalmol-2019-315231. Bhattacharya P, Edwards K, Harkin D, et al. Central corneal basal cell density and nerve parameters in ocular surface disease and limbal stem cell deficiency: a review and meta-analysis. Br J Ophthalmol. 2020, 104(12): 1633-1639. DOI: 10.1136/bjophthalmol-2019-315231.
28、Bonnet%20C%2C%20et%20al.%20Cell%20Morphology%20as%20an%20In%20Vivo%20Parameter%20for%20the%20Diagnosis%20of%20Limbal%20Stem%20Cell%20Deficiency.%20Cornea.%202022%2C%2041(8)%3A995-1001.%20DOI%3A%2010.1097%2FICO.0000000000002955.%C2%A0Bonnet%20C%2C%20et%20al.%20Cell%20Morphology%20as%20an%20In%20Vivo%20Parameter%20for%20the%20Diagnosis%20of%20Limbal%20Stem%20Cell%20Deficiency.%20Cornea.%202022%2C%2041(8)%3A995-1001.%20DOI%3A%2010.1097%2FICO.0000000000002955.%C2%A0
29、Wang LY, Wei ZY, Cao K, et al. In vivo confocal microscopic characteristics of limbal stem cell deficiency. Zhonghua Yan Ke Za Zhi. 2020, 56(6): 447-455. DOI: 10.3760/cma.j.cn112142-20191221-00660. Wang LY, Wei ZY, Cao K, et al. In vivo confocal microscopic characteristics of limbal stem cell deficiency. Zhonghua Yan Ke Za Zhi. 2020, 56(6): 447-455. DOI: 10.3760/cma.j.cn112142-20191221-00660.
30、Li Y, Ma X, Li J, et al. Corneal denervation causes epithelial apoptosis through inhibiting NAD+ biosynthesis. Invest Ophthalmol Vis Sci. 2019, 60(10): 3538-3546. DOI: 10.1167/iovs.19-26909. Li Y, Ma X, Li J, et al. Corneal denervation causes epithelial apoptosis through inhibiting NAD+ biosynthesis. Invest Ophthalmol Vis Sci. 2019, 60(10): 3538-3546. DOI: 10.1167/iovs.19-26909.
31、Ueno H, Ferrari G, Hattori T, et al. Dependence of corneal stem/progenitor cells on ocular surface innervation. Invest Ophthalmol Vis Sci. 2012, 53(2): 867-872. DOI: 10.1167/iovs.11-8438.Ueno H, Ferrari G, Hattori T, et al. Dependence of corneal stem/progenitor cells on ocular surface innervation. Invest Ophthalmol Vis Sci. 2012, 53(2): 867-872. DOI: 10.1167/iovs.11-8438.
32、Yazdanpanah G, Jabbehdari S, Djalilian AR. Limbal and corneal epithelial homeostasis. Curr Opin Ophthalmol. 2017, 28(4): 348-354. DOI: 10.1097/icu.0000000000000378.Yazdanpanah G, Jabbehdari S, Djalilian AR. Limbal and corneal epithelial homeostasis. Curr Opin Ophthalmol. 2017, 28(4): 348-354. DOI: 10.1097/icu.0000000000000378.
33、Amin S, Jalilian E, Katz E, et al. The limbal niche and regenerative strategies. Vision (Basel). 2021, 5(4): 43. DOI: 10.3390/vision5040043.Amin S, Jalilian E, Katz E, et al. The limbal niche and regenerative strategies. Vision (Basel). 2021, 5(4): 43. DOI: 10.3390/vision5040043.
34、 Grieve K, Ghoubay D, Georgeon C, et al. Three-dimensional structure of the mammalian limbal stem cell niche. Exp Eye Res, 2015, 140: 75-84. DOI: 10.1016/j.exer.2015.08.003. Grieve K, Ghoubay D, Georgeon C, et al. Three-dimensional structure of the mammalian limbal stem cell niche. Exp Eye Res, 2015, 140: 75-84. DOI: 10.1016/j.exer.2015.08.003.
35、Sun D, Shi WY, Dou SQ. Single-cell RNA sequencing in cornea research: Insights into limbal stem cells and their niche regulation. World J Stem Cells. 2023, 15(5): 466-475. DOI: 10.4252/wjsc.v15.i5.466.Sun D, Shi WY, Dou SQ. Single-cell RNA sequencing in cornea research: Insights into limbal stem cells and their niche regulation. World J Stem Cells. 2023, 15(5): 466-475. DOI: 10.4252/wjsc.v15.i5.466.
36、Yazdanpanah G, Haq Z, Kang K, et al. Strategies for reconstructing the limbal stem cell niche. Ocul Surf. 2019, 17(2): 230-240. DOI: 10.1016/j.jtos.2019.01.002.Yazdanpanah G, Haq Z, Kang K, et al. Strategies for reconstructing the limbal stem cell niche. Ocul Surf. 2019, 17(2): 230-240. DOI: 10.1016/j.jtos.2019.01.002.
37、Medeiros CS, Santhiago MR. Corneal nerves anatomy, function, injury and regeneration. Exp Eye Res. 2020, 200: 108243. DOI: 10.1016/j.exer.2020.108243.Medeiros CS, Santhiago MR. Corneal nerves anatomy, function, injury and regeneration. Exp Eye Res. 2020, 200: 108243. DOI: 10.1016/j.exer.2020.108243.
38、Yang AY, Chow J, Liu J. Corneal Innervation and Sensation: The Eye and Beyond. Yale J Biol Med. 2018, 91(1): 13-21.Yang AY, Chow J, Liu J. Corneal Innervation and Sensation: The Eye and Beyond. Yale J Biol Med. 2018, 91(1): 13-21.
39、Stepp%20MA%2C%20Pal-Ghosh%20S%2C%20Downie%20LE%2C%20et%20al.%20Corneal%20epithelial%20%E2%80%9Cneuromas%E2%80%9D%3A%20a%20case%20of%20mistaken%20identity%3F.%20Cornea.%202020%2C%2039(7)%3A%20930-934.%20DOI%3A%2010.1097%2Fico.0000000000002294.%20Stepp%20MA%2C%20Pal-Ghosh%20S%2C%20Downie%20LE%2C%20et%20al.%20Corneal%20epithelial%20%E2%80%9Cneuromas%E2%80%9D%3A%20a%20case%20of%20mistaken%20identity%3F.%20Cornea.%202020%2C%2039(7)%3A%20930-934.%20DOI%3A%2010.1097%2Fico.0000000000002294.%20
40、Wang C, Fu T, Xia C, et al. Changes in mouse corneal epithelial innervation with age. Invest Ophthalmol Vis Sci. 2012, 53(8): 5077. DOI: 10.1167/iovs.12-9704. Wang C, Fu T, Xia C, et al. Changes in mouse corneal epithelial innervation with age. Invest Ophthalmol Vis Sci. 2012, 53(8): 5077. DOI: 10.1167/iovs.12-9704.
41、Tuck H, Park M, Carnell M, et al. Neuronal-epithelial cell alignment: a determinant of health and disease status of the cornea. Ocul Surf. 2021, 21: 257-270. DOI: 10.1016/j.jtos.2021.03.007. Tuck H, Park M, Carnell M, et al. Neuronal-epithelial cell alignment: a determinant of health and disease status of the cornea. Ocul Surf. 2021, 21: 257-270. DOI: 10.1016/j.jtos.2021.03.007.
42、Al-Aqaba MA, Fares U, Suleman H, et al. Architecture and distribution of human corneal nerves. Br J Ophthalmol. 2010, 94(6): 784-789. DOI: 10.1136/bjo.2009.173799. Al-Aqaba MA, Fares U, Suleman H, et al. Architecture and distribution of human corneal nerves. Br J Ophthalmol. 2010, 94(6): 784-789. DOI: 10.1136/bjo.2009.173799.
43、Badian RA, Lagali N. The inferocentral whorl region and its directional patterns in the corneal sub-basal nerve plexus: a review. Exp Eye Res. 2024, 244: 109926. DOI: 10.1016/j.exer.2024.109926. Badian RA, Lagali N. The inferocentral whorl region and its directional patterns in the corneal sub-basal nerve plexus: a review. Exp Eye Res. 2024, 244: 109926. DOI: 10.1016/j.exer.2024.109926.
44、Stache N, Sterenczak KA, Sperlich K, et al. Assessment of dynamic corneal nerve changes using static landmarks by in vivo large-area confocal microscopy: a longitudinal proof-of-concept study. Quant Imaging Med Surg. 2022, 12(10): 4734-4746. DOI: 10.21037/qims-22-15. Stache N, Sterenczak KA, Sperlich K, et al. Assessment of dynamic corneal nerve changes using static landmarks by in vivo large-area confocal microscopy: a longitudinal proof-of-concept study. Quant Imaging Med Surg. 2022, 12(10): 4734-4746. DOI: 10.21037/qims-22-15.
45、Bhattacharya S, Mukherjee A, Pisano S, et al. The biophysical property of the limbal niche maintains stemness through YAP. Cell Death Differ. 2023, 30(6): 1601-1614. DOI: 10.1038/s41418-023-01156-7. Bhattacharya S, Mukherjee A, Pisano S, et al. The biophysical property of the limbal niche maintains stemness through YAP. Cell Death Differ. 2023, 30(6): 1601-1614. DOI: 10.1038/s41418-023-01156-7.
46、 Collinson JM, Chanas SA, Hill RE, et al. Corneal development, limbal stem cell function, and corneal epithelial cell migration in the Pax6(+ /-) mouse. Invest Ophthalmol Vis Sci. 2004, 45(4): 1101-1108. DOI: 10.1167/iovs.03-1118. Collinson JM, Chanas SA, Hill RE, et al. Corneal development, limbal stem cell function, and corneal epithelial cell migration in the Pax6(+ /-) mouse. Invest Ophthalmol Vis Sci. 2004, 45(4): 1101-1108. DOI: 10.1167/iovs.03-1118.
47、Collinson JM, Morris L, Reid AI, et al. Clonal analysis of patterns of growth, stem cell activity, and cell movement during the development and maintenance of the murine corneal epithelium. Dev Dyn. 2002, 224(4): 432-440. DOI: 10.1002/dvdy.10124.Collinson JM, Morris L, Reid AI, et al. Clonal analysis of patterns of growth, stem cell activity, and cell movement during the development and maintenance of the murine corneal epithelium. Dev Dyn. 2002, 224(4): 432-440. DOI: 10.1002/dvdy.10124.
48、Marfurt CF, Cox J, Deek S, et al. Anatomy of the human corneal innervation. Exp Eye Res. 2010, 90(4): 478-492. DOI: 10.1016/j.exer.2009.12.010. Marfurt CF, Cox J, Deek S, et al. Anatomy of the human corneal innervation. Exp Eye Res. 2010, 90(4): 478-492. DOI: 10.1016/j.exer.2009.12.010.
49、Marfurt CF, Ellis LC. Immunohistochemical localization of tyrosine hydroxylase in corneal nerves. J Comp Neurol. 1993, 336(4): 517-531. DOI: 10.1002/cne.903360405. Marfurt CF, Ellis LC. Immunohistochemical localization of tyrosine hydroxylase in corneal nerves. J Comp Neurol. 1993, 336(4): 517-531. DOI: 10.1002/cne.903360405.
50、Singh RB, Naderi A, Cho W, et al. Modulating the tachykinin: Role of substance P and neurokinin receptor expression in ocular surface disorders. Ocul Surf. 2022, 25: 142-153. DOI: 10.1016/j.jtos.2022.06.007. Singh RB, Naderi A, Cho W, et al. Modulating the tachykinin: Role of substance P and neurokinin receptor expression in ocular surface disorders. Ocul Surf. 2022, 25: 142-153. DOI: 10.1016/j.jtos.2022.06.007.
51、%20S%C5%82oniecka%20M%2C%20Le%20Roux%20S%2C%20Zhou%20Q%2C%20et%20al.%20Substance%20P%20enhances%20keratocyte%20migration%20and%20neutrophil%20recruitment%20through%20interleukin-8.%20Mol%20Pharmacol.%202016%2C%2089(2)%3A%20215-225.%20DOI%3A%2010.1124%2Fmol.115.101014.%20%20S%C5%82oniecka%20M%2C%20Le%20Roux%20S%2C%20Zhou%20Q%2C%20et%20al.%20Substance%20P%20enhances%20keratocyte%20migration%20and%20neutrophil%20recruitment%20through%20interleukin-8.%20Mol%20Pharmacol.%202016%2C%2089(2)%3A%20215-225.%20DOI%3A%2010.1124%2Fmol.115.101014.%20
52、Lasagni Vitar R, Triani F, Barbariga M, et al. Substance P/neurokinin-1 receptor pathway blockade ameliorates limbal stem cell deficiency by modulating mTOR pathway and preventing cell senescence. Stem Cell Rep. 2022, 17(4): 849-863. DOI: 10.1016/j.stemcr.2022.02.012. Lasagni Vitar R, Triani F, Barbariga M, et al. Substance P/neurokinin-1 receptor pathway blockade ameliorates limbal stem cell deficiency by modulating mTOR pathway and preventing cell senescence. Stem Cell Rep. 2022, 17(4): 849-863. DOI: 10.1016/j.stemcr.2022.02.012.
53、Wang Y, Gao G, Wu Y, et al. S100A4 silencing facilitates corneal wound healing after alkali burns by promoting autophagy via blocking the PI3K/Akt/mTOR signaling pathway. Invest Ophthalmol Vis Sci. 2020, 61(11): 19. DOI: 10.1167/iovs.61.11.19. Wang Y, Gao G, Wu Y, et al. S100A4 silencing facilitates corneal wound healing after alkali burns by promoting autophagy via blocking the PI3K/Akt/mTOR signaling pathway. Invest Ophthalmol Vis Sci. 2020, 61(11): 19. DOI: 10.1167/iovs.61.11.19.
54、Bignami F, Rama P, Ferrari G. Substance P and its inhibition in ocular inflammation. Curr Drug Targets. 2016, 17(11): 1265-1274. DOI: 10.2174/1389450116666151019100216. Bignami F, Rama P, Ferrari G. Substance P and its inhibition in ocular inflammation. Curr Drug Targets. 2016, 17(11): 1265-1274. DOI: 10.2174/1389450116666151019100216.
55、Kowtharapu BS, Murín R, Jünemann AGM, et al. Role of corneal stromal cells on epithelial cell function during wound healing. Int J Mol Sci. 2018, 19(2): E464. DOI: 10.3390/ijms19020464.Kowtharapu BS, Murín R, Jünemann AGM, et al. Role of corneal stromal cells on epithelial cell function during wound healing. Int J Mol Sci. 2018, 19(2): E464. DOI: 10.3390/ijms19020464.
56、Yang L, Di G, Qi X, et al. Substance P promotes diabetic corneal epithelial wound healing through molecular mechanisms mediated via the neurokinin-1 receptor. Diabetes. 2014, 63(12): 4262-4274. DOI: 10.2337/db14-0163.Yang L, Di G, Qi X, et al. Substance P promotes diabetic corneal epithelial wound healing through molecular mechanisms mediated via the neurokinin-1 receptor. Diabetes. 2014, 63(12): 4262-4274. DOI: 10.2337/db14-0163.
57、Redkiewicz P. The regenerative potential of substance P. Int J Mol Sci. 2022, 23(2): 750. DOI: 10.3390/ijms23020750. Redkiewicz P. The regenerative potential of substance P. Int J Mol Sci. 2022, 23(2): 750. DOI: 10.3390/ijms23020750.
58、Lasagni Vitar RM, Fonteyne P, Chaabane L, et al. A hypothalamic-controlled neural reflex promotes corneal inflammation. Invest Ophthalmol Vis Sci. 2021, 62(13): 21. DOI: 10.1167/iovs.62.13.21.Lasagni Vitar RM, Fonteyne P, Chaabane L, et al. A hypothalamic-controlled neural reflex promotes corneal inflammation. Invest Ophthalmol Vis Sci. 2021, 62(13): 21. DOI: 10.1167/iovs.62.13.21.
59、Sun J, Ramnath RD, Tamizhselvi R, et al. Role of protein kinase C and phosphoinositide 3-kinase-Akt in substance P-induced proinflammatory pathways in mouse macrophages. FASEB J. 2009, 23(4): 997-1010. DOI: 10.1096/fj.08-121756. Sun J, Ramnath RD, Tamizhselvi R, et al. Role of protein kinase C and phosphoinositide 3-kinase-Akt in substance P-induced proinflammatory pathways in mouse macrophages. FASEB J. 2009, 23(4): 997-1010. DOI: 10.1096/fj.08-121756.
60、Gaddipati S, Rao P, Jerome AD, et al. Loss of neurokinin-1 receptor alters ocular surface homeostasis and promotes an early development of herpes stromal keratitis. J Immunol. 2016, 197(10): 4021-4033. DOI: 10.4049/jimmunol.1600836.Gaddipati S, Rao P, Jerome AD, et al. Loss of neurokinin-1 receptor alters ocular surface homeostasis and promotes an early development of herpes stromal keratitis. J Immunol. 2016, 197(10): 4021-4033. DOI: 10.4049/jimmunol.1600836.
61、Woronkowicz M, Roberts H, Skopiński P. The role of insulin-like growth factor (IGF) system in the corneal epithelium homeostasis-from limbal epithelial stem cells to therapeutic applications. Biology (Basel). 2024, 13(3): 144. DOI: 10.3390/biology13030144. Woronkowicz M, Roberts H, Skopiński P. The role of insulin-like growth factor (IGF) system in the corneal epithelium homeostasis-from limbal epithelial stem cells to therapeutic applications. Biology (Basel). 2024, 13(3): 144. DOI: 10.3390/biology13030144.
62、Yang L, Sui W, Li Y, et al. Substance P inhibits hyperosmotic stress-induced apoptosis in corneal epithelial cells through the mechanism of Akt activation and reactive oxygen species scavenging via the neurokinin-1 receptor. PLoS One. 2016, 11(2): e0149865. DOI: 10.1371/journal.pone.0149865. Yang L, Sui W, Li Y, et al. Substance P inhibits hyperosmotic stress-induced apoptosis in corneal epithelial cells through the mechanism of Akt activation and reactive oxygen species scavenging via the neurokinin-1 receptor. PLoS One. 2016, 11(2): e0149865. DOI: 10.1371/journal.pone.0149865.
63、Ko JA, Yanai R, Nishida T. Up-regulation of ZO-1 expression and barrier function in cultured human corneal epithelial cells by substance P. FEBS Lett. 2009, 583(12): 2148-2153. DOI: 10.1016/j.febslet.2009.05.010. Ko JA, Yanai R, Nishida T. Up-regulation of ZO-1 expression and barrier function in cultured human corneal epithelial cells by substance P. FEBS Lett. 2009, 583(12): 2148-2153. DOI: 10.1016/j.febslet.2009.05.010.
64、 Puri S, Kenyon BM, Hamrah P. Immunomodulatory role of neuropeptides in the cornea. Biomedicines. 2022, 10(8): 1985. DOI: 10.3390/biomedicines10081985. Puri S, Kenyon BM, Hamrah P. Immunomodulatory role of neuropeptides in the cornea. Biomedicines. 2022, 10(8): 1985. DOI: 10.3390/biomedicines10081985.
65、Aerts-Kaya F, et al. Neurological Regulation of the Bone Marrow Niche. Adv Exp Med Biol. 2020, 1212: 127-153. DOI: 10.1007/5584_2019_398 .Aerts-Kaya F, et al. Neurological Regulation of the Bone Marrow Niche. Adv Exp Med Biol. 2020, 1212: 127-153. DOI: 10.1007/5584_2019_398 .
66、Mu C, Hu Y, Hou Y, et al. Substance P-embedded multilayer on titanium substrates promotes local osseointegration via MSC recruitment. J Mater Chem B. 2020, 8(6): 1212-1222. DOI: 10.1039/c9tb01124b.Mu C, Hu Y, Hou Y, et al. Substance P-embedded multilayer on titanium substrates promotes local osseointegration via MSC recruitment. J Mater Chem B. 2020, 8(6): 1212-1222. DOI: 10.1039/c9tb01124b.
67、Jung J, Jeong J, Hong HS. Substance P improves MSC-mediated RPE regeneration by modulating PDGF-BB. Biochem Biophys Res Commun. 2019, 515(4): 524-530. DOI: 10.1016/j.bbrc.2019.05.186. Jung J, Jeong J, Hong HS. Substance P improves MSC-mediated RPE regeneration by modulating PDGF-BB. Biochem Biophys Res Commun. 2019, 515(4): 524-530. DOI: 10.1016/j.bbrc.2019.05.186.
68、Cheng P, Sun X, Yin D, et al. Nanog down-regulates the Wnt signaling pathway via β-catenin phosphorylation during epidermal stem cell proliferation and differentiation. Cell Biosci. 2015, 5: 5. DOI: 10.1186/2045-3701-5-5.Cheng P, Sun X, Yin D, et al. Nanog down-regulates the Wnt signaling pathway via β-catenin phosphorylation during epidermal stem cell proliferation and differentiation. Cell Biosci. 2015, 5: 5. DOI: 10.1186/2045-3701-5-5.
69、Yin M, Li C, Peng XD, et al. Expression and role of calcitonin gene-related peptide in mouse Aspergillus fumigatus keratitis. Int J Ophthalmol. 2019, 12(5): 697-704. DOI: 10.18240/ijo.2019.05.01.Yin M, Li C, Peng XD, et al. Expression and role of calcitonin gene-related peptide in mouse Aspergillus fumigatus keratitis. Int J Ophthalmol. 2019, 12(5): 697-704. DOI: 10.18240/ijo.2019.05.01.
70、Yuan K, et al. Sensory nerves promote corneal inflammation resolution via CGRP mediated transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway. Int Immunopharmacol. 2022, 102: 108426. DOI: 10.1016/j.intimp.2021.108426 .Yuan K, et al. Sensory nerves promote corneal inflammation resolution via CGRP mediated transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway. Int Immunopharmacol. 2022, 102: 108426. DOI: 10.1016/j.intimp.2021.108426 .
71、Mikulec AA, Tanelian DL. CGRP increases the rate of corneal re-epithelialization in an in vitro whole mount preparation. J Ocul Pharmacol Ther. 1996, 12(4): 417-423. DOI: 10.1089/jop.1996.12.417. Mikulec AA, Tanelian DL. CGRP increases the rate of corneal re-epithelialization in an in vitro whole mount preparation. J Ocul Pharmacol Ther. 1996, 12(4): 417-423. DOI: 10.1089/jop.1996.12.417.
72、Zhu S, Zidan A, Pang K, et al. Promotion of corneal angiogenesis by sensory neuron-derived calcitonin gene-related peptide. Exp Eye Res. 2022, 220: 109125. DOI: 10.1016/j.exer.2022.109125.Zhu S, Zidan A, Pang K, et al. Promotion of corneal angiogenesis by sensory neuron-derived calcitonin gene-related peptide. Exp Eye Res. 2022, 220: 109125. DOI: 10.1016/j.exer.2022.109125.
73、Kadar T, Dachir S, Cohen M, et al. Prolonged impairment of corneal innervation after exposure to sulfur mustard and its relation to the development of delayed limbal stem cell deficiency. Cornea. 2013, 32(4): e44-50. DOI: 10.1097/ico.0b013e318262e885. Kadar T, Dachir S, Cohen M, et al. Prolonged impairment of corneal innervation after exposure to sulfur mustard and its relation to the development of delayed limbal stem cell deficiency. Cornea. 2013, 32(4): e44-50. DOI: 10.1097/ico.0b013e318262e885.
74、Sullivan C, et al. Evidence for a phenotypic switch in corneal afferents after lacrimal gland excision. Exp Eye Res. 2022, 218: 109005. DOI: 10.1016/j.exer.2022.109005.Sullivan C, et al. Evidence for a phenotypic switch in corneal afferents after lacrimal gland excision. Exp Eye Res. 2022, 218: 109005. DOI: 10.1016/j.exer.2022.109005.
75、Diel RJ, Mehra D, Kardon R, et al. Photophobia: shared pathophysiology underlying dry eye disease, migraine and traumatic brain injury leading to central neuroplasticity of the trigeminothalamic pathway. Br J Ophthalmol. 2021, 105(6): 751-760. DOI: 10.1136/bjophthalmol-2020-316417.Diel RJ, Mehra D, Kardon R, et al. Photophobia: shared pathophysiology underlying dry eye disease, migraine and traumatic brain injury leading to central neuroplasticity of the trigeminothalamic pathway. Br J Ophthalmol. 2021, 105(6): 751-760. DOI: 10.1136/bjophthalmol-2020-316417.
76、Weirath NA, Haskell-Luevano C. Recommended tool compounds for the melanocortin receptor (MCR) G protein-coupled receptors (GPCRs). ACS Pharmacol Transl Sci. 2024, 7(9): 2706-2724. DOI: 10.1021/acsptsci.4c00129.Weirath NA, Haskell-Luevano C. Recommended tool compounds for the melanocortin receptor (MCR) G protein-coupled receptors (GPCRs). ACS Pharmacol Transl Sci. 2024, 7(9): 2706-2724. DOI: 10.1021/acsptsci.4c00129.
77、Chu C, Huang Y, Ru Y, et al. α-MSH ameliorates corneal surface dysfunction in scopolamine-induced dry eye rats and human corneal epithelial cells via enhancing EGFR expression. Exp Eye Res. 2021, 210: 108685. DOI: 10.1016/j.exer.2021.108685. Chu C, Huang Y, Ru Y, et al. α-MSH ameliorates corneal surface dysfunction in scopolamine-induced dry eye rats and human corneal epithelial cells via enhancing EGFR expression. Exp Eye Res. 2021, 210: 108685. DOI: 10.1016/j.exer.2021.108685.
78、Nohara K, Zhang Y, Waraich RS, et al. Early-life exposure to testosterone programs the hypothalamic melanocortin system. Endocrinology. 2011, 152(4): 1661-1669. DOI: 10.1210/en.2010-1288. Nohara K, Zhang Y, Waraich RS, et al. Early-life exposure to testosterone programs the hypothalamic melanocortin system. Endocrinology. 2011, 152(4): 1661-1669. DOI: 10.1210/en.2010-1288.
79、Daini E, Vandini E, Bodria M, et al. Melanocortin receptor agonist NDP-α-MSH improves cognitive deficits and microgliosis but not amyloidosis in advanced stages of AD progression in 5XFAD and 3xTg mice. Front Immunol. 2023, 13: 1082036. DOI: 10.3389/fimmu.2022.1082036. Daini E, Vandini E, Bodria M, et al. Melanocortin receptor agonist NDP-α-MSH improves cognitive deficits and microgliosis but not amyloidosis in advanced stages of AD progression in 5XFAD and 3xTg mice. Front Immunol. 2023, 13: 1082036. DOI: 10.3389/fimmu.2022.1082036.
80、Bock F, Onderka J, Braun G, et al. Identification of novel endogenous anti(lymph)angiogenic factors in the aqueous humor. Invest Ophthalmol Vis Sci. 2016, 57(15): 6554. DOI: 10.1167/iovs.15-18526. Bock F, Onderka J, Braun G, et al. Identification of novel endogenous anti(lymph)angiogenic factors in the aqueous humor. Invest Ophthalmol Vis Sci. 2016, 57(15): 6554. DOI: 10.1167/iovs.15-18526.
81、Li C, Wu M, Gu L, et al. α- MSH plays anti-inflammatory and anti-fungal role in Aspergillus fumigatus keratitis. Curr Eye Res. 2022, 47(3): 343-351. DOI: 10.1080/02713683.2021.2006235. Li C, Wu M, Gu L, et al. α- MSH plays anti-inflammatory and anti-fungal role in Aspergillus fumigatus keratitis. Curr Eye Res. 2022, 47(3): 343-351. DOI: 10.1080/02713683.2021.2006235.
82、Kistenmacher%20S%2C%20Schw%C3%A4mmle%20M%2C%20Martin%20G%2C%20et%20al.%20Enrichment%2C%20characterization%2C%20and%20proteomic%20profiling%20of%20small%20extracellular%20vesicles%20derived%20from%20human%20limbal%20mesenchymal%20stromal%20cells%20and%20melanocytes.%20Cells.%202024%2C%2013(7)%3A%20623.%20DOI%3A%2010.3390%2Fcells13070623.%20Kistenmacher%20S%2C%20Schw%C3%A4mmle%20M%2C%20Martin%20G%2C%20et%20al.%20Enrichment%2C%20characterization%2C%20and%20proteomic%20profiling%20of%20small%20extracellular%20vesicles%20derived%20from%20human%20limbal%20mesenchymal%20stromal%20cells%20and%20melanocytes.%20Cells.%202024%2C%2013(7)%3A%20623.%20DOI%3A%2010.3390%2Fcells13070623.%20
83、Dziasko MA, Tuft SJ, Daniels JT. Limbal melanocytes support limbal epithelial stem cells in 2D and 3D microenvironments. Exp Eye Res. 2015, 138: 70-79. DOI: 10.1016/j.exer.2015.06.026. Dziasko MA, Tuft SJ, Daniels JT. Limbal melanocytes support limbal epithelial stem cells in 2D and 3D microenvironments. Exp Eye Res. 2015, 138: 70-79. DOI: 10.1016/j.exer.2015.06.026.
84、 Li H, Hou L. Regulation of melanocyte stem cell behavior by the niche microenvironment. Pigment Cell Melanoma Res. 2018, 31(5): 556-569. DOI: 10.1111/pcmr.12701. Li H, Hou L. Regulation of melanocyte stem cell behavior by the niche microenvironment. Pigment Cell Melanoma Res. 2018, 31(5): 556-569. DOI: 10.1111/pcmr.12701.
85、Lu%C5%BEnik%20Marzidov%C5%A1ek%20Z%2C%20Blanco%20T%2C%20Sun%20Z%2C%20et%20al.%20The%20neuropeptide%20alpha-melanocyte%E2%80%93stimulating%20hormone%20is%20critical%20for%20corneal%20endothelial%20cell%20protection%20and%20graft%20survival%20after%20transplantation.%20Am%20J%20Pathol.%202022%2C%20192(2)%3A%20270-280.%20DOI%3A%2010.1016%2Fj.ajpath.2021.10.016.%20Lu%C5%BEnik%20Marzidov%C5%A1ek%20Z%2C%20Blanco%20T%2C%20Sun%20Z%2C%20et%20al.%20The%20neuropeptide%20alpha-melanocyte%E2%80%93stimulating%20hormone%20is%20critical%20for%20corneal%20endothelial%20cell%20protection%20and%20graft%20survival%20after%20transplantation.%20Am%20J%20Pathol.%202022%2C%20192(2)%3A%20270-280.%20DOI%3A%2010.1016%2Fj.ajpath.2021.10.016.%20
86、Mashimo M, Moriwaki Y, Misawa H, et al. Regulation of immune functions by non-neuronal acetylcholine (ACh) via muscarinic and nicotinic ACh receptors. Int J Mol Sci. 2021, 22(13): 6818. DOI: 10.3390/ijms22136818.Mashimo M, Moriwaki Y, Misawa H, et al. Regulation of immune functions by non-neuronal acetylcholine (ACh) via muscarinic and nicotinic ACh receptors. Int J Mol Sci. 2021, 22(13): 6818. DOI: 10.3390/ijms22136818.
87、S%C5%82oniecka%20M%2C%20Danielson%20P.%20Acetylcholine%20decreases%20formation%20of%20myofibroblasts%20and%20excessive%20extracellular%20matrix%20production%20in%20an%20in%20vitro%20human%20corneal%20fibrosis%20model.%20J%20Cell%20Mol%20Med.%202020%2C%2024(8)%3A%204850-4862.%20DOI%3A%2010.1111%2Fjcmm.15168.%20S%C5%82oniecka%20M%2C%20Danielson%20P.%20Acetylcholine%20decreases%20formation%20of%20myofibroblasts%20and%20excessive%20extracellular%20matrix%20production%20in%20an%20in%20vitro%20human%20corneal%20fibrosis%20model.%20J%20Cell%20Mol%20Med.%202020%2C%2024(8)%3A%204850-4862.%20DOI%3A%2010.1111%2Fjcmm.15168.%20
88、Kurzen H, Wessler I, Kirkpatrick CJ, et al. The non-neuronal cholinergic system of human skin. Horm Metab Res. 2007, 39(2): 125-135. DOI: 10.1055/s-2007-961816. Kurzen H, Wessler I, Kirkpatrick CJ, et al. The non-neuronal cholinergic system of human skin. Horm Metab Res. 2007, 39(2): 125-135. DOI: 10.1055/s-2007-961816.
89、Takahashi T. Roles of nAChR and Wnt signaling in intestinal stem cell function and inflammation. Int Immunopharmacol. 2020, 81: 106260. DOI: 10.1016/j.intimp.2020.106260.Takahashi T. Roles of nAChR and Wnt signaling in intestinal stem cell function and inflammation. Int Immunopharmacol. 2020, 81: 106260. DOI: 10.1016/j.intimp.2020.106260.
90、Tracey KJ. Reflex control of immunity. Nat Rev Immunol. 2009, 9(6): 418-428. DOI: 10.1038/nri2566. Tracey KJ. Reflex control of immunity. Nat Rev Immunol. 2009, 9(6): 418-428. DOI: 10.1038/nri2566.
91、Grando SA, Pittelkow MR, Schallreuter KU. Adrenergic and cholinergic control in the biology of epidermis: physiological and clinical significance. J Investig Dermatol. 2006, 126(9): 1948-1965. DOI: 10.1038/sj.jid.5700151. Grando SA, Pittelkow MR, Schallreuter KU. Adrenergic and cholinergic control in the biology of epidermis: physiological and clinical significance. J Investig Dermatol. 2006, 126(9): 1948-1965. DOI: 10.1038/sj.jid.5700151.
92、S%C5%82oniecka%20M%2C%20Backman%20LJ%2C%20Danielson%20P.%20Acetylcholine%20enhances%20keratocyte%20proliferation%20through%20muscarinic%20receptor%20activation.%20Int%20Immunopharmacol.%202015%2C%2029(1)%3A%2057-62.%20DOI%3A%2010.1016%2Fj.intimp.2015.05.039.%20S%C5%82oniecka%20M%2C%20Backman%20LJ%2C%20Danielson%20P.%20Acetylcholine%20enhances%20keratocyte%20proliferation%20through%20muscarinic%20receptor%20activation.%20Int%20Immunopharmacol.%202015%2C%2029(1)%3A%2057-62.%20DOI%3A%2010.1016%2Fj.intimp.2015.05.039.%20
93、Pflugfelder SC, Cao A, Galor A, et al. Nicotinic acetylcholine receptor stimulation: a new approach for stimulating tear secretion in dry eye disease. Ocul Surf. 2022, 25: 58-64. DOI: 10.1016/j.jtos.2022.05.001.Pflugfelder SC, Cao A, Galor A, et al. Nicotinic acetylcholine receptor stimulation: a new approach for stimulating tear secretion in dry eye disease. Ocul Surf. 2022, 25: 58-64. DOI: 10.1016/j.jtos.2022.05.001.
94、Wirta D, Vollmer P, Paauw J, et al. Efficacy and safety of OC-01 (varenicline solution) nasal spray on signs and symptoms of dry eye disease the ONSET-2 phase 3 randomized trial. Ophthalmology. 2022, 129(4): 379-387. DOI: 10.1016/j.ophtha.2021.11.004. Wirta D, Vollmer P, Paauw J, et al. Efficacy and safety of OC-01 (varenicline solution) nasal spray on signs and symptoms of dry eye disease the ONSET-2 phase 3 randomized trial. Ophthalmology. 2022, 129(4): 379-387. DOI: 10.1016/j.ophtha.2021.11.004.
95、Musayeva A, Jiang S, Ruan Y, et al. Aged mice devoid of the M3 muscarinic acetylcholine receptor develop mild dry eye disease. Int J Mol Sci. 2021, 22(11): 6133. DOI: 10.3390/ijms22116133. Musayeva A, Jiang S, Ruan Y, et al. Aged mice devoid of the M3 muscarinic acetylcholine receptor develop mild dry eye disease. Int J Mol Sci. 2021, 22(11): 6133. DOI: 10.3390/ijms22116133.
96、Grando SA. Connections of nicotine to cancer. Nat Rev Cancer. 2014, 14: 419-429. DOI: 10.1038/nrc3725.Grando SA. Connections of nicotine to cancer. Nat Rev Cancer. 2014, 14: 419-429. DOI: 10.1038/nrc3725.
97、Chernyavsky AI, Galitovskiy V, Grando SA. Molecular mechanisms of synergy of corneal muscarinic and nicotinic acetylcholine receptors in upregulation of E-cadherin expression. Int Immunopharmacol. 2015, 29(1): 15-20. DOI: 10.1016/j.intimp.2015.04.036.Chernyavsky AI, Galitovskiy V, Grando SA. Molecular mechanisms of synergy of corneal muscarinic and nicotinic acetylcholine receptors in upregulation of E-cadherin expression. Int Immunopharmacol. 2015, 29(1): 15-20. DOI: 10.1016/j.intimp.2015.04.036.
98、 Xue Y, et al. The mouse autonomic nervous system modulates inflammation and epithelial renewal after corneal abrasion through the activation of distinct local macrophages. Mucosal Immunol. 2018, 11(5):1496-1511. DOI: 10.1038/s41385-018-0031-6. Xue Y, et al. The mouse autonomic nervous system modulates inflammation and epithelial renewal after corneal abrasion through the activation of distinct local macrophages. Mucosal Immunol. 2018, 11(5):1496-1511. DOI: 10.1038/s41385-018-0031-6.
99、Chernyavsky AI, Galitovskiy V, Shchepotin IB, et al. The acetylcholine signaling network of corneal epithelium and its role in regulation of random and directional migration of corneal epithelial cells. Investig Ophthalmol Vis Sci. 2014, 55(10): 6921-6933. DOI: 10.1167/iovs.14-14667. Chernyavsky AI, Galitovskiy V, Shchepotin IB, et al. The acetylcholine signaling network of corneal epithelium and its role in regulation of random and directional migration of corneal epithelial cells. Investig Ophthalmol Vis Sci. 2014, 55(10): 6921-6933. DOI: 10.1167/iovs.14-14667.
100、Maestroni GJM. Adrenergic modulation of hematopoiesis. J Neuroimmune Pharmacol. 2020, 15(1): 82-92. DOI: 10.1007/s11481-019-09840-7. Maestroni GJM. Adrenergic modulation of hematopoiesis. J Neuroimmune Pharmacol. 2020, 15(1): 82-92. DOI: 10.1007/s11481-019-09840-7.
101、Figueira L, Ferreira C, Janeiro C, et al. Concentration gradient of noradrenaline from the periphery to the centre of the cornea - A clue to its origin. Exp Eye Res. 2018, 168: 107-114. DOI: 10.1016/j.exer.2018.01.008. Figueira L, Ferreira C, Janeiro C, et al. Concentration gradient of noradrenaline from the periphery to the centre of the cornea - A clue to its origin. Exp Eye Res. 2018, 168: 107-114. DOI: 10.1016/j.exer.2018.01.008.
102、 Li J, Ma X, Zhao L, et al. Extended contact lens wear promotes corneal norepinephrine secretion and pseudomonas aeruginosa infection in mice. Invest Ophthalmol Vis Sci. 2020, 61(4): 17. DOI: 10.1167/iovs.61.4.17. Li J, Ma X, Zhao L, et al. Extended contact lens wear promotes corneal norepinephrine secretion and pseudomonas aeruginosa infection in mice. Invest Ophthalmol Vis Sci. 2020, 61(4): 17. DOI: 10.1167/iovs.61.4.17.
103、Ma X, Wang Q, Song F, et al. Corneal epithelial injury-induced norepinephrine promotes Pseudomonas aeruginosa keratitis. Exp Eye Res. 2020, 195: 108048. DOI: 10.1016/j.exer.2020.108048. Ma X, Wang Q, Song F, et al. Corneal epithelial injury-induced norepinephrine promotes Pseudomonas aeruginosa keratitis. Exp Eye Res. 2020, 195: 108048. DOI: 10.1016/j.exer.2020.108048.
104、Lee H, Lee JH, Hong S, et al. Transplantation of human corneal limbal epithelial cell sheet harvested on synthesized carboxymethyl cellulose and dopamine in a limbal stem cell deficiency. J Tissue Eng Regen Med. 2021, 15(2): 139-149. DOI: 10.1002/term.3159. Lee H, Lee JH, Hong S, et al. Transplantation of human corneal limbal epithelial cell sheet harvested on synthesized carboxymethyl cellulose and dopamine in a limbal stem cell deficiency. J Tissue Eng Regen Med. 2021, 15(2): 139-149. DOI: 10.1002/term.3159.
105、Figueira L, Janeiro C, Ferreirinha F, et al. Regulation of corneal noradrenaline release and topography of sympathetic innervation: Functional implications for adrenergic mechanisms in the human cornea. Exp Eye Res. 2018, 174: 121-132. DOI: 10.1016/j.exer.2018.05.023. Figueira L, Janeiro C, Ferreirinha F, et al. Regulation of corneal noradrenaline release and topography of sympathetic innervation: Functional implications for adrenergic mechanisms in the human cornea. Exp Eye Res. 2018, 174: 121-132. DOI: 10.1016/j.exer.2018.05.023.
106、Ghoghawala SY, Mannis MJ, Pullar CE, et al. Beta2-adrenergic receptor signaling mediates corneal epithelial wound repair. Invest Ophthalmol Vis Sci. 2008, 49(5): 1857-1863. DOI: 10.1167/iovs.07-0925.Ghoghawala SY, Mannis MJ, Pullar CE, et al. Beta2-adrenergic receptor signaling mediates corneal epithelial wound repair. Invest Ophthalmol Vis Sci. 2008, 49(5): 1857-1863. DOI: 10.1167/iovs.07-0925.
107、Grueb M, Bartz-Schmidt KU, Rohrbach JM. Adrenergic regulation of cAMP/protein kinase A pathway in corneal epithelium and endothelium. Ophthalmic Res. 2008, 40(6): 322-328. DOI: 10.1159/000150446.Grueb M, Bartz-Schmidt KU, Rohrbach JM. Adrenergic regulation of cAMP/protein kinase A pathway in corneal epithelium and endothelium. Ophthalmic Res. 2008, 40(6): 322-328. DOI: 10.1159/000150446.
108、 Zhang X, Muddana S, Kumar SR, et al. Topical pergolide enhance corneal nerve regrowth following induced corneal abrasion. Invest Ophthalmol Vis Sci. 2020, 61(1): 4. DOI: 10.1167/iovs.61.1.4. Zhang X, Muddana S, Kumar SR, et al. Topical pergolide enhance corneal nerve regrowth following induced corneal abrasion. Invest Ophthalmol Vis Sci. 2020, 61(1): 4. DOI: 10.1167/iovs.61.1.4.
109、Cavallotti C, Pescosolido N, Artico M, et al. Localization of dopamine receptors in the rabbit cornea. Cornea. 1999, 18(6): 721-728. DOI: 10.1097/00003226-199911000-00016. Cavallotti C, Pescosolido N, Artico M, et al. Localization of dopamine receptors in the rabbit cornea. Cornea. 1999, 18(6): 721-728. DOI: 10.1097/00003226-199911000-00016.
110、Rose-John S. Interleukin-6 family cytokines. Cold Spring Harb Perspect Biol. 2018, 10(2): a028415. DOI: 10.1101/cshperspect.a028415. Rose-John S. Interleukin-6 family cytokines. Cold Spring Harb Perspect Biol. 2018, 10(2): a028415. DOI: 10.1101/cshperspect.a028415.
111、Oh B, Wu YW, Swaminathan V, et al. Modulating the electrical and mechanical microenvironment to guide neuronal stem cell differentiation. Adv Sci. 2021, 8(7): 2002112. DOI: 10.1002/advs.202002112. Oh B, Wu YW, Swaminathan V, et al. Modulating the electrical and mechanical microenvironment to guide neuronal stem cell differentiation. Adv Sci. 2021, 8(7): 2002112. DOI: 10.1002/advs.202002112.
112、Reichard M, Hovakimyan M, Guthoff RF, et al. In vivo visualisation of murine corneal nerve fibre regeneration in response to ciliary neurotrophic factor. Exp Eye Res. 2014, 120: 20-27. DOI: 10.1016/j.exer.2013.12.015. Reichard M, Hovakimyan M, Guthoff RF, et al. In vivo visualisation of murine corneal nerve fibre regeneration in response to ciliary neurotrophic factor. Exp Eye Res. 2014, 120: 20-27. DOI: 10.1016/j.exer.2013.12.015.
113、Qi H, Chuang EY, Yoon KC, et al. Patterned expression of neurotrophic factors and receptors in human limbal and corneal regions. Mol Vis. 2007, 13: 1934-1941. Qi H, Chuang EY, Yoon KC, et al. Patterned expression of neurotrophic factors and receptors in human limbal and corneal regions. Mol Vis. 2007, 13: 1934-1941.
114、Chung ES, Lee KH, Kim M, et al. Expression of neurotrophic factors and their receptors in keratoconic cornea. Curr Eye Res. 2013, 38(7): 743-750. DOI: 10.3109/02713683.2013.774421. Chung ES, Lee KH, Kim M, et al. Expression of neurotrophic factors and their receptors in keratoconic cornea. Curr Eye Res. 2013, 38(7): 743-750. DOI: 10.3109/02713683.2013.774421.
115、Zhou Q, Chen P, Di G, et al. Ciliary neurotrophic factor promotes the activation of corneal epithelial stem/progenitor cells and accelerates corneal epithelial wound healing. Stem Cells. 2015, 33(5): 1566-1576. DOI: 10.1002/stem.1942.Zhou Q, Chen P, Di G, et al. Ciliary neurotrophic factor promotes the activation of corneal epithelial stem/progenitor cells and accelerates corneal epithelial wound healing. Stem Cells. 2015, 33(5): 1566-1576. DOI: 10.1002/stem.1942.
116、Levi-Montalcini R. The nerve growth factor 35 years later. Science. 1987, 237(4819): 1154-1162. DOI: 10.1126/science.3306916.Levi-Montalcini R. The nerve growth factor 35 years later. Science. 1987, 237(4819): 1154-1162. DOI: 10.1126/science.3306916.
117、Chen H, Zhang J, Dai Y, et al. Nerve growth factor inhibits TLR3-induced inflammatory cascades in human corneal epithelial cells. J Inflamm (Lond). 2019, 16: 27. DOI: 10.1186/s12950-019-0232-0. Chen H, Zhang J, Dai Y, et al. Nerve growth factor inhibits TLR3-induced inflammatory cascades in human corneal epithelial cells. J Inflamm (Lond). 2019, 16: 27. DOI: 10.1186/s12950-019-0232-0.
118、 Lambiase A, Manni L, Bonini S, et al. Nerve growth factor promotes corneal healing: structural, biochemical, and molecular analyses of rat and human corneas. Invest Ophthalmol Vis Sci. 2000, 41(5):1063-9. Lambiase A, Manni L, Bonini S, et al. Nerve growth factor promotes corneal healing: structural, biochemical, and molecular analyses of rat and human corneas. Invest Ophthalmol Vis Sci. 2000, 41(5):1063-9.
119、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(4): 253-258. DOI: 10.1016/j.phrs.2008.01.010. 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(4): 253-258. DOI: 10.1016/j.phrs.2008.01.010.
120、Fink DM, Connor AL, Kelley PM, et al. Nerve growth factor regulates neurolymphatic remodeling during corneal inflammation and resolution. PLoS One. 2014, 9(11): e112737. DOI: 10.1371/journal.pone.0112737. Fink DM, Connor AL, Kelley PM, et al. Nerve growth factor regulates neurolymphatic remodeling during corneal inflammation and resolution. PLoS One. 2014, 9(11): e112737. DOI: 10.1371/journal.pone.0112737.
121、Park JH, Kang SS, Kim JY, et al. Nerve growth factor attenuates apoptosis and inflammation in the diabetic cornea. Invest Ophthalmol Vis Sci. 2016, 57(15): 6767. DOI: 10.1167/iovs.16-19747. Park JH, Kang SS, Kim JY, et al. Nerve growth factor attenuates apoptosis and inflammation in the diabetic cornea. Invest Ophthalmol Vis Sci. 2016, 57(15): 6767. DOI: 10.1167/iovs.16-19747.
122、 Di G, Qi X, Zhao X, et al. Corneal epithelium-derived neurotrophic factors promote nerve regeneration. Invest Ophthalmol Vis Sci. 2017, 58(11): 4695-4702. DOI: 10.1167/iovs.16-21372. Di G, Qi X, Zhao X, et al. Corneal epithelium-derived neurotrophic factors promote nerve regeneration. Invest Ophthalmol Vis Sci. 2017, 58(11): 4695-4702. DOI: 10.1167/iovs.16-21372.
123、 Qi H, Li DQ, Shine HD, et al. Nerve growth factor and its receptor TrkA serve as potential markers for human corneal epithelial progenitor cells. Exp Eye Res. 2008, 86(1): 34-40. DOI: 10.1016/j.exer.2007.09.003. Qi H, Li DQ, Shine HD, et al. Nerve growth factor and its receptor TrkA serve as potential markers for human corneal epithelial progenitor cells. Exp Eye Res. 2008, 86(1): 34-40. DOI: 10.1016/j.exer.2007.09.003.
124、Pedrotti E, Bonacci E, Chierego C, et al. Eight months follow-up of corneal nerves and sensitivity after treatment with cenegermin for neurotrophic keratopathy. Orphanet J Rare Dis. 2022, 17(1): 63. DOI: 10.1186/s13023-022-02237-5. Pedrotti E, Bonacci E, Chierego C, et al. Eight months follow-up of corneal nerves and sensitivity after treatment with cenegermin for neurotrophic keratopathy. Orphanet J Rare Dis. 2022, 17(1): 63. DOI: 10.1186/s13023-022-02237-5.
125、Hong%20J%2C%20et%20al.%20NGF%20promotes%20cell%20cycle%20progression%20by%20regulating%20D-type%20cyclins%20via%20PI3K%2FAkt%20and%20MAPK%2FErk%20activation%20in%20human%20corneal%20epithelial%20cells.%20Mol%20Vis.%202012%2C%2018%3A%20758-764.%C2%A0%20Hong%20J%2C%20et%20al.%20NGF%20promotes%20cell%20cycle%20progression%20by%20regulating%20D-type%20cyclins%20via%20PI3K%2FAkt%20and%20MAPK%2FErk%20activation%20in%20human%20corneal%20epithelial%20cells.%20Mol%20Vis.%202012%2C%2018%3A%20758-764.%C2%A0%20
126、Ting DSJ. Re: bonini et al. phase 2 randomized, double-masked, vehicle-controlled trial of recombinant human nerve growth factor for neurotrophic keratitis (ophthalmology. 2018;125: 1332-1343). Ophthalmology. 2019, 126(2): e14-e15. DOI: 10.1016/j.ophtha.2018.09.017. Ting DSJ. Re: bonini et al. phase 2 randomized, double-masked, vehicle-controlled trial of recombinant human nerve growth factor for neurotrophic keratitis (ophthalmology. 2018;125: 1332-1343). Ophthalmology. 2019, 126(2): e14-e15. DOI: 10.1016/j.ophtha.2018.09.017.
127、Pedrotti E, Bonetto J, Cozzini T, et al. Cenegermin in pediatric neurotrophic keratopathy. Cornea. 2019, 38(11): 1450-1452. DOI: 10.1097/ico.0000000000002112.Pedrotti E, Bonetto J, Cozzini T, et al. Cenegermin in pediatric neurotrophic keratopathy. Cornea. 2019, 38(11): 1450-1452. DOI: 10.1097/ico.0000000000002112.
128、Compagnoni C, Zelli V, Bianchi A, et al. microRNAs expression in response to rhNGF in epithelial corneal cells: focus on neurotrophin signaling pathway. Int J Mol Sci. 2022, 23(7): 3597. DOI: 10.3390/ijms23073597. Compagnoni C, Zelli V, Bianchi A, et al. microRNAs expression in response to rhNGF in epithelial corneal cells: focus on neurotrophin signaling pathway. Int J Mol Sci. 2022, 23(7): 3597. DOI: 10.3390/ijms23073597.
129、Kalaimani L, Devarajan B, Namperumalsamy VP, et al. Hsa-miR-143-3p inhibits Wnt-β-catenin and MAPK signaling in human corneal epithelial stem cells. Sci Rep. 2022, 12(1): 11432. DOI: 10.1038/s41598-022-15263-x.Kalaimani L, Devarajan B, Namperumalsamy VP, et al. Hsa-miR-143-3p inhibits Wnt-β-catenin and MAPK signaling in human corneal epithelial stem cells. Sci Rep. 2022, 12(1): 11432. DOI: 10.1038/s41598-022-15263-x.
上一篇
下一篇
目录

点击右上角菜单,浏览器打开下载

我知道了