Content » Vol 96, Issue 5

Review

The Role of Neuromediators and Innervation in Cutaneous Wound Healing

Mohammed ASHRAFI1,2, Mohamed BAGUNEID2 and Ardeshir BAYAT1,2

1Plastic and Reconstructive Surgery Research, Centre for Dermatological Research, Institute of Inflammation and Repair, University of Manchester, 2University Hospital South Manchester NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK

The skin is densely innervated with an intricate network of cutaneous nerves, neuromediators and specific receptors which influence a variety of physiological and disease processes. There is emerging evidence that cutaneous innervation may play an important role in mediating wound healing. This review aims to comprehensively examine the evidence that signifies the role of innervation during the overlapping stages of cutaneous wound healing. Numerous neuropeptides that are secreted by the sensory and autonomic nerve fibres play an essential part during the distinct phases of wound healing. Delayed wound healing in diabetes and fetal cutaneous regeneration following wounding further highlights the pivotal role skin innervation and its associated neuromediators play in wound healing. Understanding the mechanisms via which cutaneous innervation modulates wound healing in both the adult and fetus will provide opportunities to develop therapeutic devices which could manipulate skin innervation to aid wound healing. Key words: wound healing; skin innervation; neuromediators; neuropeptides; fetal wound healing.

Accepted Dec 15, 2015; Epub ahead of print Dec 17, 2015

Acta Derm Venereol 2016; 96: XX–XX.

Ardeshir Bayat PhD, Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology (MIB), 131 Princess Street, Manchester M1 7DN, UK. E-mail: ardeshir.bayat@manchester.ac.uk

Wound healing involves a set of highly regulated overlapping mechanisms that utilise cellular, molecular and chemical components. It begins at the moment of injury and continues for months or years to produce a healed wound that is similar to uninjured skin in terms of function and appearance. The central nervous system-skin interactions involve a number of neuromediators, cytokines, hormones and other effector molecules (1). Therefore stimuli received at the skin interface can influence the nervous system at both a local and central level; the brain can modulate skin function in physiological and pathophysiological states and the skin can alter the pharmacology of the central nervous system by releasing large amounts of neuropeptides (2).

The aim of this review is to provide a detailed update on normal skin innervation and its role in the different phases of wound healing. Furthermore, it describes the effect of cutaneous denervation on wounds in animal models and human clinical observations. Finally, the role of innervation in fetal wound healing which is regenerative rather than reparative is discussed.

SKIN INNERVATION

Skin contains a complex network of sensory nerve fibres (Fig. 1). The innervated skin is a vital barrier with direct contact to the central nervous system. These nerves are crucial in influencing physiological and pathophysiological cutaneous functions (3). Mechanoreceptors, thermoreceptors and nociceptors are found in the epidermis and dermis. Mechanoreceptors in the epidermis are Merkel discs and free nerve endings. Mechanoreceptors in the dermis are Ruffini, Meissner, Pacinian corpuscles and free nerve endings (4, 5).

4609fig1.tif

Fig. 1. Cross section outlining the distribution of the cutaneous nervous system in the skin. MC: Meissner’s corpuscles; MD: Merkel disc; PC: Pacinian corpuscle; RC: Ruffini’s corpuscle; AN: autonomic nerves.

Two distinct groups of nerve fibres are found in the skin. Cutaneous sensory nerve fibres are in close contact with dermal blood vessels, mast cells, fibroblasts, keratinocytes and Langerhans cells in the epidermis (6–9). Cutaneous sensory nerves are classified according to diameter and speed of impulse as Aβ, Aδ and C nerve fibres. Aβ fibres are fast and large whereas C fibres are slow and small. Aδ fibres constitute 80% of primary sensory nerves emerging from the dorsal root ganglia, whereas C fibres make up 20% of the primary nerves (10, 11). Aβ and Aδ are myelinated by accompanying Schwann cells. These sensory nerves, which extend throughout all layers of the skin (12), transfer signals from mechanoreceptors, thermoreceptors and nociceptors to their origin in the dorsal root ganglia. Mechanical stimuli are detected via mechanoreceptors associated with sensory corpuscles through Aβ fibres or with Aδ fibres; temperature via the thermoreceptors through Aδ and C fibres; and pain via nociceptors through Aδ and C fibres (3). From there, sensations like pain, burning and itching are forwarded to specific areas in the brain. The second group of nerves comprises the autonomic nerve fibres which constitute only a minority of cutaneous nerve fibres compared with sensory nerves. They are restricted to the dermis and are involved in regulating blood circulation, lymphatic function and the regulation of skin appendages (sweat glands, apocrine glands and hair follicles) (13, 14).

Sensory nerves in the skin are able to release neuromediators such as neuropeptides which signal to the skin (15, 16). In un-stimulated nerves, neuromediators are barely detectable within the skin tissues. However on direct stimulation a significant increase of regulatory neuromediators can be detected in vitro and in vivo (3). Neuropeptides are a family of extracellular messengers, which act as neurotransmitters, hormones or paracrine factors (17). The majority consist of a group of small peptides that exert their effects by interacting with members of a superfamily of G protein-coupled receptors (3). Numerous neuropeptides are expressed and released from sensory as well as autonomic cutaneous nerves, including calcitonin gene related peptide (CGRP), substance P, neurokinin A, and vasoactive intestinal peptide (VIP) (13). In addition, cutaneous cells themselves such as keratinocytes, micro-vascular endothelial cells, merkel cells, fibroblasts or leukocytes are capable of releasing neuropeptides (18, 19). The presence of neuropeptides in free nerve fibre endings and the proximity of these endings to a variety of cells in the skin seem to associate the cutaneous nervous system not merely for its role in sensation but in other biological actions as well, namely wound healing (20).

WOUND HEALING PHASES AND ROLE OF NEUROMEDIATORS

Wound healing involves a range of processes that operate in a systematic and timely manner to repair the skin’s integrity and function. Wound healing can be divided into 3 overlapping stages: inflammation, proliferation and remodelling. There is increasing evidence that cutaneous innervation may play an important role in mediating normal wound healing. Numerous neuropeptides that are secreted by the sensory and autonomic nerve fibres play an essential part during the distinct phases of wound healing (Table I). The role of skin innervation and neuropeptides in the different phases of wound healing are discussed below.

Table I. The role of neuromediators in cutaneous wound healing

Neuromediator

Wound healing action

BDNF

Re-epithelialisation/keratinocyte proliferation

Wound contraction

Nerve regeneration

CGRP

Vasodilatation

Release of pro-inflammatory cytokines

Re-epithelialisation/keratinocyte proliferation

Granulation tissue formation/fibroblast proliferation

Angiogenesis

Wound contraction

Collagen maturation and remodelling

CRH

Release of pro-inflammatory cytokines

Angiogenesis

SP

Vasodilatation

Polymorphonuclear cell infiltration

Release of pro-inflammatory cytokines

Re-epithelialisation/keratinocyte proliferation

Granulation tissue formation/fibroblast proliferation

Angiogenesis

Collagen maturation and remodelling

Nerve regeneration

NPY

Vasoconstriction

Angiogenesis

Gal

Vasoconstriction

Release of pro-inflammatory cytokines

Re-epithelialisation/keratinocyte proliferation

Nerve regeneration

GRP

Re-epithelialisation/keratinocyte proliferation

Angiogenesis

NGF

Re-epithelialisation/keratinocyte proliferation

Angiogenesis

Wound contraction

Fibroblast differentiation into myofibroblasts

Collagen maturation and remodelling

Nerve regeneration

NKA

Vasodilatation

Polymorphonuclear cell infiltration

Re-epithelialisation/keratinocyte proliferation

Granulation tissue formation/fibroblast proliferation

Angiogenesis

Nerve regeneration

NT-3

Nerve regeneration

PACAP

Vasodilatation

Polymorphonuclear cell infiltration

Re-epithelialisation/keratinocyte proliferation

VIP

Vasodilatation

Release of pro-inflammatory cytokines

Re-epithelialisation/keratinocyte proliferation (-ve effect)

Angiogenesis

Collagen deposition

Nerve regeneration

BDNF: brain-derived neurotrophic factor; CGRP: calcitonin gene related peptide; CRH: corticotropin-releasing hormone; SP: Substance P; NPY: Neuropeptide Y; Gal: Galanoin; GRP: gastrin releasing peptide; NGF: nerve growth factor; NKA: neurokinin A ; NT-3: Neurotrophin-3; PACAP: pituitary adenylate cyclase activating peptide ; VIP: vasoactive intestinal peptide.

Inflammatory phase

After a cutaneous injury, haemostasis which lasts a couple of hours produces a fibrin plug. The aggregated platelets release pro-inflammatory mediators such as cytokines and growth factors. These chemotactic signals recruit neutrophils which are the principal cellular components in the activation of the inflammatory phase of wound healing (21). Neutrophils act as chemo-attractants for other cells that are involved in the inflammatory phase (22), including modulating the expression of macrophages (23). Macrophages enter the wound and support the ongoing process by phagocytosis of pathogens and cell debris (24, 25) as well as by the secretion of growth factors, chemokines and cytokines (26, 27). The inflammatory phase can last between hours and days. Numerous neuropeptides released from the cutaneous innervation have been shown to activate vital processes during the inflammatory phase of wound healing (Fig. 2). Substance P (SP) appears to play a major role in the inflammatory phase; however other neuropeptides have also been identified and are reviewed.

4609fig2.tif

Fig. 2. Inflammatory phase. Overview outlining the role of neuromediators on the different wound healing actions during the inflammatory phase.

SP is produced in the dorsal root ganglion of the spinal cord and distributed to the dorsal horn of the spinal cord and the nerve endings of the sensory neurons in the dermis and epidermis (17, 28). SP is also detected on human keratinocytes, endothelial cells and fibroblasts (29). The actions of SP are regulated through a neurokinin G-protein coupled receptor, NK-1R, which is expressed in neurons and peripheral tissues (30). SP acts on the vasculature, cutaneous epithelium and connective tissue. Nitric oxide is essential for adequate wound healing (31). SP stimulates vasodilatation and micro-vascular permeability through increasing nitric oxide release and through direct effects on endothelial cells (32). SP up-regulates expression of adhesion molecules on endothelial cells, monocyte chemotaxis and inflammatory cell activity (33–35). SP also modulates the synthesis and release of pro-inflammatory cytokines such as interleukins, transforming growth factor-α (TGF-α) and tumour necrosis factor-α; key components during the inflammatory phase of wound healing (36).

Neutral endopeptidase (NEP) is a zinc metalloprotease which competes with NK-1R and inhibits the actions of SP through enzymatic degradation (37). The interactions of SP and NEP are key components of inflammatory signalling in wound repair (20).

Neurokinin A is a bio-active tachykinin released into the skin after injury and activates, preferentially through neurokinin-2 receptor NK-2R, cutaneous target cells such as keratinocytes and dermal endothelial cells thereby modulating skin inflammation during wound healing (30). Immunohistochemistry studies have demonstrated corticotropin-releasing hormone (CRH) is found in sensory cutaneous nerves (38). CRH acts as a pro-inflammatory mediator and induces skin mast cell degranulation, thereby increasing vascular permeability and the release of pro-inflammatory cytokines (3). CRH has also been shown to enhance angiogenesis in the skin (39).

CGRP is a vasodilator and enhances plasma extravasation (40) and can stimulate angiogenesis. CGRP has been shown to increase the inflammatory response of other mediators such as SP (41). Activin is a member of the TGF-β superfamily which increases on wounding and has been shown to up-regulate CGRP expression in innervating sensory neurons (42), highlighting its regulatory role in wound healing. Nerve growth factor (NGF) has been shown to increase the release of CGRP from peripheral nerve terminals into peripheral tissue underlining its role as a modulator of the inflammatory phase of wound healing (43).

Kim & Pomeranz (44) showed that sympathetic nervous system stimulation accelerates wound healing at the dermal and epidermal levels through its proposed action on the process of neurogenic inflammation. Peripheral nerve fibres express α1-adrenoceptors and their expression increases after nerve injury possibly boosting neurogenic inflammation (45).

Proliferative phase

The early inflammatory phase is succeeded by a proliferative phase lasting several weeks. It is characterised by the invasion of macrophages and fibroblasts into the wounded region and formation of granulation tissue. Macrophages induce fibroblasts to produce an extracellular matrix (ECM) of type-3 collagen that provides a structural framework for endothelial cells, angiogenesis and wound contraction (Fig. 3).

4609fig3.tif

Fig. 3. Proliferative phase. Overview outlining the role of neuromediators on the different wound healing actions during the proliferative phase.

SP has potent proliferative effects on fibroblasts, keratinocytes and endothelial cells by stimulating DNA synthesis (46, 47). It also stimulates angiogenesis through possible nitric oxide mediation (48, 49). SP plays a crucial role in the granulation tissue remodelling process by promoting the proliferation and the migration of dermal fibroblasts (50) and by stimulating the expression of epidermal growth factor and its associated receptor (50, 51).

NGF is a polypeptide neurotrophin found in neurons of the central and peripheral nervous system as well as in a variety of cells including fibroblasts, epithelial cells, keratinocytes and immune cells (52). NGF has an essential role in differentiation, function and survival of sensory and autonomic nerves (53). NGF also has anti-inflammatory properties (54). NGF has been postulated to promote the proliferation of local immature cells in wounds, blood vessel formation and neurite overgrowth (55). NGF has been shown in animal studies and in a human case study to promote angiogenesis and epithelial healing (30, 55). Neurokinin A stimulates the release of NGF in the epidermis (56).

Other important neuropeptides identified to play a role in the proliferative phase include gastrin releasing peptide (GRP), CGRP, galanin, VIP and pituitary adenylate cyclase activating peptide (PACAP). GRP is widely distributed in the central and peripheral nervous system and although its implications in wound healing are not certain, some studies have shown it could promote migration, proliferation of keratinocytes and angiogenesis (57). CGRP stimulates the proliferation and migration of keratinocytes (58). Galanin is a peptide released from afferent nerves which signals through G-protein coupled receptors and has anti-proliferative effects in tissue (59). On the contrary an in vitro study identified galanin induced the up-regulation of NGF (60). VIP has been shown to act as a growth factor for proliferation of keratinocytes and as a modulator of their migration (61) as well as inducing histamine release by mast cells causing vasodilatation (62). VIP may be involved in the re-innervation of wounded tissue as it has been shown to promote sciatic nerve regeneration in rat sciatic nerve after transection (63). PACAP is found in sensory cutaneous nerves (64). It is a member of the VIP peptide family and is a potent vasodilatator (65). It is postulated that C-fibres release PACAP in response to neuronal activation which in turn leads vasodilatation and extravasation. PACAP is involved in cutaneous inflammation by releasing histamine from mast cells and it also promotes human keratinocyte proliferation (66).

Activation and antagonism of the sympathetic nervous system via adrenoceptors during the proliferative phase of wound healing has been studied. Α1-adrenoceptors influence growth cycles via mitogen activated protein kinase signalling pathways thus regulating cellular proliferation after injury (67). Activation of α1β-adrenoceptors and the β2-adrenoceptors mediates the proliferation and migration of fibroblasts (68). Whereas, β1/β2-adrenoceptor blockade accelerates human keratinocyte migration and re-epithelialisation in wound healing models (69). The production and remodelling of connective tissues in the skin are influenced by the balance of several essential cytokines, including TGF-β and insulin like growth factor-1 (IGF-1) (70). Studies have shown TGF-β stimulates overall ECM formation (71) and IGF-1 plays a regulatory role in enhancing proliferation of fibroblasts and modulates chemotaxis (72). Sympathetic stimulation through α-adrenoceptors elevates the production of TGF-β1 and IGF-1 in skin fibroblasts. These results imply that the sympathetic nervous system contributes to the modulation of cytokine secretion, ECM production and fibroblast migration in the skin (73). Neuropeptide Y (NPY) is widely distributed in the central and peripheral nervous system. In the skin the expression of NPY was detected in sympathetic nerve fibres in the deep and superficial dermis (74). NPY directly stimulates endothelial cell proliferation and migration, however its role as an angiogenic factor is debatable (75, 76).

Remodelling phase

The remodelling phase is characterised by proliferative cell apoptosis, ECM adjustment and organisation and replacement of type-3 with type-1 collagen (Fig. 4). The remodelling phase can last between weeks to years. Very little is known of the function of cutaneous innervation or neuropeptides in the remodelling phase.

4609fig4.tif

Fig. 4. Remodelling phase. Overview outlining the role of neuromediators on the different wound healing actions during the remodelling phase.

Altun et al. (77) demonstrated significantly higher number of nerve fibres in normotrophic scars in comparison to hypertrophic scars suggesting a regulatory role for skin nerves in the remodelling phase of wound healing. Sensory nerve fibres regenerate into the repaired epidermis and dermis in response to neuropeptides (78). SP induces human dermal micro-vascular endothelial cells to produce NGF in vitro which in turn is required for nerve fibre regeneration following cutaneous injury (79). NGF has also been suggested to accelerate tissue remodelling (55). Neurotrophin-3 (NT-3) is a neurotrophic growth factor expressed by sensory and sympathetic nerves which is essential for growth, proliferation, and maintenance of nerves (80). Likewise brain-derived neurotrophic factor (BDNF) is required for the postnatal survival or functional maturation of sensory neurons. BDNF and their receptors are expressed by keratinocytes, fibroblasts and myofibroblasts, and promote their proliferation and differentiation (81, 82).

SP has been shown to influence the process of wound collagen degradation by increasing the matrix matalloproteinase-2 activity in fibroblasts (83). Fujiwara et al. (84) showed direct neuronal contact in vitro accelerates differentiation of fibroblasts into myofibroblasts which in turn secrete collagen fibres and induce wound contraction. Cheret et al. (85) showed SP, CGRP and VIP modulate matrix metalloproteinase activities and affect collagen-1 and collagen-3 productions during skin wound healing.

WOUND HEALING IN DENERVATED SKIN

Delayed wound healing in denervated skin further highlights the significance of cutaneous innervation in wound repair. Denervated skin is not simply associated with wounding but has a major role to play in the delayed healing process. This has been investigated experimentally in human and animal models and observed clinically.

Experimental denervation

Experimentally induced denervation supports the key role of skin innervation in wound healing. Vanilloid receptor-1 is expressed on sensory Aδ and C-fibres; and capsaicin is an agonist which causes denervation of these fibres. Capsaicin induced sensory denervation results in delayed re-epithelialisation, absence of vasodilatation and plasma protein extravasation indicating that cutaneous nerves are responsible for the initial two phases of wound healing (86, 87). Chemical sympathectomy with 6-hydroxydopamine delays wound healing by its effect on the inflammatory phase (88).

Denervated skin has been shown to exhibit delayed wound contraction and reduced micro-vascular response (89, 90). Fukai et al. (91) showed both wound contraction and epithelisation were delayed in denervated skin of mice by 17% and 25% respectively. Engin et al. (89) showed delayed wound contraction and loss of neuropeptide secretion from nerve endings in denervated tissue. The lack of neuropeptides in denervated skin may be the cause of deficiencies seen in the wound healing process. This hypothesis is enhanced by the finding that SP levels are decreased in denervated tissue and exogenous administration of SP promotes wound healing in mice (92–94). Likewise, capsaicin induced denervation lowers NGF transport (95) and differing levels of exogenous NGF accelerates wound closure, epithelisation and wound contraction (96, 97). Also, CGRP immune-reactive nerves are reduced in wounded tissue following denervation (92) and the role of CGRP in wound healing have been detailed above.

Diabetes mellitus

Diabetes is an important risk factor for the development and persistence of chronic wounds and all phases of wound healing are affected. Neuropathy is a possible cause of delayed wound healing in diabetes. There are fewer nerves in the epidermis and dermis in the skin of diabetic humans and mice (93). Animal studies have shown that diabetic mice have slower wound healing compared with controls and have delayed inflammatory cell migration (98). Neuropathy itself hinders the rate of cutaneous nerve regeneration (99) and Cheng et al. (100) showed that diabetic mice lack the capabilities to regenerate nerves. Neuropeptide expression and functions are affected in diabetes. Reduced SP and NGF positive nerve fibres are observed in diabetic patients and rats (101, 102); as well as reduced NGF and SP levels in the skin and serum (102, 103). This could also account for the decrease in angiogenesis seen in this sub-group of patients (104). NPY levels in diabetic skin are reduced (105). NPY binding to Y2 receptors is important in angiogenesis and as a result deletion of the Y2 receptor in diabetic mice results in the blockage of NPY induced angiogenesis and delays wound healing (75). NEP regulates the biologic action of SP. NEP levels are increased in diabetic wounds which may contribute to the deficient neuro-inflammatory signalling and wound healing in diabetics by decreasing the neuropeptide levels (106). The expression, release and action of CGRP is decreased in diabetes resulting in reduced CGRP mediated vasodilatation (107).

Paraplegic patients

The clinical observation that paraplegic and quadriplegic patients present with impaired wound closure below the level of the spinal cord lesion suggests the participation of the nervous system in wound healing (108). Below the level of injury there is interruption of spinal vasomotor pathways resulting in a loss of tone in the vascular bed below the lesion and a state of generalised vasodilation (109). The denervated tissue is therefore unable to mount an inflammatory response (110) and the lack of tissue perfusion due to a chronic state of hypotension leads to the inability to deliver essential components to the wound (111, 112). This leads to lack of oxygen delivery to the wound which is vital for numerous steps of the wound healing cascade including angiogenesis, keratinocyte and fibroblast proliferation, collagen production and remodelling (113, 114). Further observations show wound healing is delayed in elderly humans (115). Khalil et al. (116) showed older rats heal substantially more slowly than those wounded at a younger age. All phases of wound healing are affected by age including delayed inflammatory infiltration into the wound, angiogenesis and collagen production and remodelling. A reduction in nerve endings as the skin ages could be a causative factor as to the delayed wound healing seen with ageing (117). Another human example is the delayed cutaneous wound healing that is apparent in those with leprosy induced neuropathy (118).

INFLUENCE OF PAIN ON WOUND HEALING

Cutaneous wounds are inevitably associated with pain. Common pain mediators released on wounding are SP, CGRP, nitric oxide and TNF-α. In acute wound healing, a short period of pain mediator release promotes the inflammatory phase and also enhances fibroblast collagen production, cellular migration and re-epithelialisation (119,120). However, in chronic pain conditions a resultant sustained release of these mediators creates an imbalance and may impede wound healing. SP is responsible for transmission of pain from sensory neurons to the central nervous system. The initial role of SP as a mediator of wound healing has been described above and the depleted levels of SP noted in diabetic patients may contribute to impaired cutaneous wound healing that is evident in this sub-group (121, 122). However prolonged pain and over-expression of SP could lead to a chronic inflammatory wound state (123). Similarly CGRP is proposed to contribute to pain transmission and has a key role to play in wound healing. However, as with SP, over-expression is detrimental to wound healing (124). TNF-α and nitric oxide are two important pain mediators which have crucial roles in wound healing. However, increased levels leads to chronic wound inflammation as seen in non healing wounds with impaired collagen production and anti-proliferative effects on fibroblasts (120, 123, 125). These findings are corroborated in a clinical study by McGuire et al. (126) who reported persistent post-surgery pain was an important predictor of healing time. They showed those that reported milder levels of pain post surgery experienced faster healing compared with those who reported more intense pain (126).

FETAL WOUND HEALING AND INNERVATION

Post natal cutaneous wound healing restores tissue integrity through fibrosis and scarring at a cost of regenerating the normal tissue architecture. Fetal cutaneous wounds made at certain developmental stages show complete regeneration. However, when fetuses are wounded later in gestation, dermal structures do not regenerate and a histologically demonstrable scar is formed (127, 128).

Kishi et al. (129) showed nerve regeneration in early gestational fetal wound healing was similar to unwounded skin. This re-innervation during healing appears to be by both collateral sprouting from intact nerves in the base of the wound and by regeneration of divided axons at the wound peripheries (130). However this regenerative capacity was lost as the wounding occurred at a later gestational age (131). This highlights the importance of cutaneous innervation in wound healing, specifically regeneration. In certain amphibious species such as newts, regeneration of amputated limbs is nerve dependant with regenerative ability lost upon denervation (131). Fetal cutaneous regeneration following wounding is also reported to be disrupted by denervation (132). Stelnicki et al. (132) showed that in fetal lambs whose limbs were denervated, incisional wounds appeared to scar, and open wounds failed to heal. The related dependence of scarless fetal repair and peripheral nerve function has also been shown in fetal mice where transection of intercostal nerves leads to loss of skin wound regeneration (129).

Previous studies (as explored in preceding sections) have focused on the role of neuromediators and their receptors in relation to adult cutaneous healing after injury. Similarly, during fetal injury this interaction of neuromediators may modulate the fetal scarless repair mechanisms in response to injury. SP and CGRP are undetectable during wound healing at early gestational age (130, 133). Zhang & Ren (134) showed SP expression altered from undetectable in early fetal skin to increasing expression with gestational term. Xie et al. (135) showed that fetal rabbit wounds healed more rapidly in comparison to adults without the formation of a scar. They showed the expression of SP and CGRP in the wound was decreased in the early stages of wound healing compared to fetal controls and remained at all times at levels equivalent to or below controls. These low levels of neuropeptide expression may contribute to scarless wound healing.

CONCLUSION AND FUTURE PERSPECTIVES

The skin is a densely innervated organ and the above clearly suggests that the cutaneous nervous system and its associated neuromediators are not solely responsible for sensory neurotransmissions to the central nervous system but play a major role in skin homeostasis and during all phases of wound healing. Delayed wound healing in clinical and experimental studies of denervated tissue have further supported this concept. This is highlighted with the imbalance of neuromediators found in denervated disease states such as diabetes, where delayed wound healing is common.

From this review it becomes apparent that pharmacological and mechanical methods of cutaneous re-innervation are an important avenue that needs to be explored further. One possibility is the use of electrical stimulation (ES) which has been shown to have beneficial effects in wound healing (136, 137). ES has been shown to enhance nerve regeneration (138, 139), however the mechanism by which it affects nerve growth and function is less understood. The functions of sensory nerves involved in wound healing have been shown to be improved by ES (140). A randomised controlled trial in humans showed ES improved factors known to reflect C-fibre function (141). The improvement in C-fibre function was associated with nearly doubling of the rate of healing in the actively stimulated group compared to the sham group. ES has been shown to improve regeneration of peripheral sensory axons both in vitro and in vivo (142–145). Another possible mechanism is that the use of ES may enhance the activity of neuromediators involved in wound healing (139, 140, 146). However the limited evidence available that shows the direct effect of electrical stimulation on skin innervation needs further clarification and verification.

Finally, the differing roles of cutaneous innervation and neuromediators between post natal and fetal wound healing may provide the opportunities to develop therapeutic technologies that could manipulate the central nervous system-skin peripheral nervous system network to not only aid in the management of delayed wound healing but also to allow cutaneous regeneration which would provide scarless healing.

ACKNOWLEDGEMENT

We would like to thank Helen Carruthers for graphical support of this paper.

The authors declare no conflict of interest.

REFERENCES

1. O’Sullivan RL, Lipper G, Lerner EA. The neuro-immuno-cutaneous-endocrine network: relationship of mind and skin. Arch Dermatol 1998; 134: 1431–1435.

2. Brazzini B, Ghersetich I, Hercogova J, Lotti T. The neuro-immuno-cutaneous-endocrine network: relationship between mind and skin. Dermatol Ther 2003; 16: 123–131.

3. Roosterman D, Goerge T, Schneider SW, Bunnett NW, Steinhoff M. Neuronal control of skin function: the skin as a neuroimmunoendocrine organ. Physiol Rev 2006; 86: 1309–1379.

4. Albrecht FLRPJ. The Senses: A Comprehensive Reference. Vol. 6. Cutaneous Mechanisms of Tactile Perception: Morphological and Chemical Organization of the Innervation to the Skin. San Diego: Academic Press; 2008. pp. 1–32.

5. Iggo A, Andres KH. Morphology of cutaneous receptors. Annu Rev Neurosci 1982; 5: 1–31.

6. Dalsgaard CJ, Rydh M, Haegerstrand A. Cutaneous innervation in man visualized with protein gene product 9.5 (PGP 9.5) antibodies. Histochemistry 1989; 92: 385–390.

7. Torii H, Hosoi J, Asahina A, Granstein RD. Calcitonin-gene-related peptide and Langerhans cell function. J Investig Dermatol Symp Proc 1997; 2: 82–86.

8. Hilliges M, Wang L, Johansson O. Ultrastructural evidence for nerve fibers within all vital layers of the human epidermis. J Invest Dermatol 1995; 104: 134–137.

9. Reilly DM, Ferdinando D, Johnston C, Shaw C, Buchanan KD, Green MR. The epidermal nerve fibre network: characterization of nerve fibres in human skin by confocal microscopy and assessment of racial variations. Br J Dermatol 1997; 137: 163–170.

10. Alvarez FJ, Fyffe RE. Nociceptors for the 21st century. Curr Rev Pain 2000; 4: 451–458.

11. Lawson SN. Phenotype and function of somatic primary afferent nociceptive neurones with C-, Adelta- or Aalpha/beta-fibres. Exp Physiol 2002; 87: 239–244.

12. Hosoi J, Murphy G, Egan C, Lerner E, Grabbe S, Asahina A, et al. Regulation of Langerhans cell function by nerves containing calcitonin generelated peptide. Nature 1993; 363: 159–163.

13. Sternini C. Organization of the peripheral nervous system: autonomic and sensory ganglia. J Investig Dermatol Symp Proc 1997; 2: 1–7.

14. Vetrugno R, Liguori R, Cortelli P, Montagna P. Sympathetic skin response: basic mechanisms and clinical applications. Clin Auton Res 2003; 13: 256–270.

15. Holzer P. Local effector functions of capsaicin-sensitive sensory nerve endings: involvement of tachykinins, calcitonin gene-related peptide and other neuropeptides. Neuroscience 1988; 24: 739–768.

16. Lembeck F, Holzer P. Substance P as neurogenic mediator of antidromic vasodilation and neurogenic plasma extravasation. Naunyn Schmiedebergs Arch Pharmacol 1979; 310: 175–183.

17. Schaffer M, Beiter T, Becker HD, Hunt TK. Neuropeptides: mediators of inflammation and tissue repair? Arch Surg 1998; 133: 1107–1116.

18. Leung MS, Wong CC. Expressions of putative neurotransmitters and neuronal growth related genes in Merkel cell-neurite complexes of the rats. Life Sci 2000; 66: 1481–1490.

19. Wang H, Xing L, Li W, Hou L, Guo J, Wang X. Production and secretion of calcitonin gene-related peptide from human lymphocytes. J Neuroimmunol 2002; 130: 155–162.

20. Ansel JC, Kaynard AH, Armstrong CA, Olerud J, Bunnett N, Payan D. Skin-nervous system interactions. J Invest Dermatol 1996; 106: 198–204.

21. Evers LH, Bhavsar D, Mailander P. The biology of burn injury. Exp Dermatol 2010; 19: 777–783.

22. Szpaderska AM, Egozi EI, Gamelli RL, DiPietro LA. The effect of thrombocytopenia on dermal wound healing. J Invest Dermatol 2003; 120: 1130–1137.

23. Daley JM, Reichner JS, Mahoney EJ, Manfield L, Henry WL Jr, Mastrofrancesco B, et al. Modulation of macrophage phenotype by soluble product(s) released from neutrophils. J Immunol 2005; 174: 2265–2272.

24. Tziotzios C, Profyris C, Sterling J. Cutaneous scarring: Pathophysiology, molecular mechanisms, and scar reduction therapeutics Part II. Strategies to reduce scar formation after dermatologic procedures. J Am Acad Dermatol 2012; 66: 13–24.

25. Profyris C, Tziotzios C, Do Vale I. Cutaneous scarring: Pathophysiology, molecular mechanisms, and scar reduction therapeutics Part I. The molecular basis of scar formation. J Am Acad Dermatol 2012; 66: 1–10.

26. Frank S, Kämpfer H, Wetzler C, Stallmeyer B, Pfeilschifter J. Large induction of the chemotactic cytokine RANTES during cutaneous wound repair: a regulatory role for nitric oxide in keratinocyte-derived RANTES expression. Biochem J 2000; 347: 265–273.

27. Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev 2003; 83: 835–870.

28. Ralevic V, Milner P, Hudlicka O, Kristek F, Burnstock G. Substance P is released from the endothelium of normal and capsaicin treated rat hind-limb vasculature in vivo, by increased flow. Circ Res 1990; 66: 1178–1183.

29. Liu JY, Hu JH, Zhu QG, Li FQ, Sun HJ. Substance P receptor expression in human skin keratinocytes and fibroblasts. Br J Dermatol 2006; 155: 657–662.

30. Emanueli C, Salis MB, Pinna A, Graiani G, Manni L, Madeddu P. Nerve growth factor promotes angiogenesis and arteriogenesis in ischemic hindlimb. Circulation 2002; 106: 2257–2262.

31. Frank S, Kampfer H, Wetzler C, Pfeilschifter J. Nitric oxide drives skin repair: novel functions of an established mediator. Kidney Int 2002; 61: 882–888.

32. Baraniuk JN, Kowalski ML, Kaliner MA. Relationships between permeable vessels, nerves, and mast cells in rat cutaneous neurogenic inflammation. J Appl Physiol 1990; 68: 2305–2311.

33. Lindsey KQ, Caughman SW, Olerud JE, Bunnett NW, Armstrong CA, Ansel JC. Neural regulation of endothelial cell-mediated inflammation. J Investig Dermatol Symp Proc 2000; 5: 74–78.

34. Helme RD, Eglezos A, Hosking CS. Substance P induces chemotaxis of neutrophils in normal and capsaicin-treated rats. Immunol Cell Biol 1987; 65: 267–269.

35. Lambert RW, Granstein RD. Neuropeptides and Langerhans cells. Exp Dermatol 1998; 7: 73–80.

36. Luger TA, Lotti T. Neuropeptides: role in inflammatory skin diseases. J Eur Acad Dermatol Venereol 1998; 10: 207–211.

37. Okamoto A, Lovett M, Payan DG, Bunnett NW. Interactions between neutral endopeptidase (EC 3.4.24.11) and the substance P (NK1) receptor expressed in mammalian cells. Biochem J 1994; 299: 683–693.

38. Slominski A, Wortsman J. Neuroendocrinology of the skin. Endocr Rev 2000; 21: 457–487.

39. Arbiser JL, Karalis K, Viswanathan A, Koike C, Anand-Apte B, Flynn E, et al. Corticotropin-releasing hormone stimulates angiogenesis and epithelial tumor growth in the skin. J Invest Dermatol 1999; 113: 838–842.

40. Gamse R, Saria A. Potentiation of tachykinin-induced plasma protein extravasation by calcitonin gene-related peptide. Eur J Pharmacol 1985; 114: 61–66.

41. Cruwys SC, Kidd BL, Mapp PI, Walsh DA, Blake DR. The effects of calcitonin gene-related peptide on formation of intra-articular oedema by inflammatory mediators. Br J Pharmacol 1992; 107: 116–119.

42. Cruise BA, Xu P, Hall AK. Wounds increase activin in skin and a vasoactive neuropeptide in sensory ganglia. Dev Biol 2004; 271: 1–10.

43. Bowles WR, Sabino M, Harding-Rose C, Hargreaves KM. Chronic nerve growth factor administration increases the peripheral exocytotic activity of capsaicin-sensitive cutaneous neurons. Neurosci Lett 2006; 403: 305–308.

44. Kim LR, Pomeranz B. The sympathomimetic agent, 6-hydoxydopamine, accelerates cutaneous wound healing. Eur J Pharmacol 1999; 376: 257–264.

45. Drummond PD, Drummond ES, Dawson LF, Mitchell V, Finch PM, Vaughan CW, et al. Upregulation of α1-adrenoceptors on cutaneous nerve fibres after partial sciatic nerve ligation and in complex regional pain syndrome type II. Pain 2014; 155: 606–616.

46. Kahler CM, Reinisch N, Wiederman CJ. Interaction of SP with epidermal growth factor and fibroblast growth factor in cyclooxygenase-dependent proliferation of human skin fibroblasts. J Cell Physiol 1996; 166: 601–608.

47. Tanaka T, Danno K, Ikai K, Imamura S. Effects of substance P and substance K on the growth of cultured keratinocytes. J Invest Dermatol 1988; 90: 399–401.

48. Fan TP, Hu DE, Guard S, Gresham GA, Watling KJ. Stimulation of angiogenesis by substance P and interleukin-1 in the rat and its inhibition by NK1 or interleukin-1 receptor antagonists. Br J Pharmacol 1993; 110: 43–49.

49. Muangman P, Tamura RN, Muffley LA, Isik FF, Scott JR, Xie C, et al. Substance P enhances wound closure in nitric oxide synthase knockout mice. J Surg Res 2009; 153: 201–209.

50. Ziche M, Morbidelli L, Pacini M, Dolara P, Maggi CA. NK1-receptors mediate the proliferative response of human fibroblasts to tachykinins. Br J Pharmacol 1990; 100: 11–14.

51. Lai X, Wang Z, Wei L, Wang L. Effect of substance P released from peripheral nerve ending on endogenous expression of epidermal growth factor and its receptor in wound healing. Chin J Traumatol 2002; 5: 176–179.

52. Aloe L, Tirassa P, Bracci-Laudiero L. Nerve growth factor in neurological and non-neurological diseases: basic findings and emerging pharmacological prospectives. Curr Pharm Des 2001; 7: 113–123.

53. Sofroniew MV, Howe CL, Mobley WC. Nerve growth factor signaling, neuroprotection, and neural repair. Annu Rev Neurosci 2001; 24: 1217–1281.

54. Banks BE, Vernon CA, Warner JA. Nerve growth factor has anti-inflammatory activity in the rat hindpaw oedema test. Neurosci Lett 1984; 47: 41–45.

55. Aloe L. Nerve growth factor, human skin ulcers and vascularization. Our experience. Prog Brain Res 2004; 146: 515–522.

56. Burbach GJ, Kim KH, Zivony AS, Kim A, Aranda J, Wright S, et al. The neurosensory tachykinins substance P and neurokinin A directly induce keratinocyte nerve growth factor. J Invest Dermatol 2001; 17: 1075–1082.

57. Baroni A, Perfetto B, Canozo N, Braca A, Farina E, Melito A, et al. Bombesin: a possible role in wound repair. Peptides 2008; 29: 1157–1166.

58. Seike M, Ikeda M, Morimoto A, Matsumoto M, Kodama H. Increased synthesis of calcitonin gene-related peptide stimulates keratinocyte proliferation in murine UVB-irradiated skin. J Dermatol Sci 2002; 28: 135–143.

59. El-Salhy M, Tjomsland V, Theodorsson E. Effects of triple treatment with octreotide, galanin and serotonin on a human pancreas cancer cell line in xenografts. Histol Histopathol 2005; 20: 745–752.

60. Dallos A, Kiss M, Polyánka H, Dobozy A, Kemény L, Husz S. Effects of the neuropeptides substance P, calcitonin gene-related peptide, vasoactive intestinal polypeptide and galanin on the production of nerve growth factor and inflammatory cytokines in cultured human keratinocytes. Neuropeptides 2006; 40: 251–263.

61. Wollina U, Knoll B, Prufer K. Vasoactive intestinal peptide (VIP) supports migration and growth of human keratinocytes on polyurethane foils. J Dermatol Sci 1993; 6: 55.

62. González C, Barroso C, Martín C, Gulbenkian S, Estrada C. Neuronal nitric oxide synthase activation by vasoactive intestinal peptide in bovine cerebral arteries. J Cereb Blood Flow Metab 1997; 17: 977–984.

63. Rayan GM, Johnson C, Pitha J, Cahill S, Said S. Vasoactive intestinal peptide and nerve growth factor effects on nerve regeneration. J Okla State Med Assoc 1995; 88: 337–341.

64. Moller K, Zhang YZ, Hakanson R, Luts A, Sjolund B, Uddman R, et al. Pituitary adenylate cyclase activating peptide is a sensory neuropeptide: immunocytochemical and immunochemical evidence. Neuroscience 1993; 57: 725–732.

65. Warren JB, Larkin SW, Coughlan M, Kajekar R, Williams TJ. Pituitary adenylate cyclase activating polypeptide is a potent vasodilator and oedema potentiator in rabbit skin in vivo. Br J Pharmacol 1992; 106: 331–334.

66. Granoth R, Fridkin M, Gozes I. VIP and the potent analog, stearyl-Nle(17)-VIP, induce proliferation of keratinocytes. FEBS Lett 2000; 475: 78–83.

67. Piascik MT, Perez DM. alpha1-Adrenergic receptors: new insights and directions. J Pharmacol Exp Ther 2001; 298: 403–410.

68. Gonzalez-Cabrera PJ, Shi T, Yun J, McCune DF, Rorabaugh BR, Perez DM. Differential regulation of the cell cycle by alpha1-adrenergic receptor subtypes. Endocrinology 2004; 145: 5157–5167.

69. Pullar CE, Rizzo A, Isseroff RR. Beta-Adrenergic receptor antagonists accelerate skin wound healing: evidence for a catecholamine synthesis network in the epidermis. J Biol Chem 2006; 281: 21225–21235.

70. Makrantonaki E, Vogel K, Fimmel S, Oeff M, Seltmann H, Zouboulis CC. Interplay of IGF-I and 17beta-estradiol at age-specific levels in human sebocytes and fibroblasts in vitro. Exp Gerontol 2008; 43: 939–946.

71. Clark RA. Biology of dermal wound repair. Dermatol Clin 1993; 11: 647–666.

72. Freinkel RK, Woodley DT. The biology of the skin. Parthenon, New York, 2001.

73. Liao MH, Liu SS, Peng IC, Tsai FJ, Huang HH. The stimulatory effects of alpha1-adrenergic receptors on TGF-beta1, IGF-1 and hyaluronan production in human skin fibroblasts. Cell Tissue Res 2014; 357: 681–693.

74. Polak JM, Bloom SR. Regulatory peptides — the distribution of two newly discovered peptides: PHI and NPY. Peptides 1984; 5: 79–89.

75. Ekstrand AJ, Cao R, Bjorndahl M, Nystrom S, Jonsson-Rylander A-C, Hassani H, et al. Deletion of neuropeptide Y (NPY) 2 receptor in mice results in blockage of NPY-induced angiogenesis and delayed wound healing. Proc Natl Acad Sci U S A 2003; 100: 6033–6038.

76. Jain M, LoGerfo FW, Guthrie P, Pradhan L. Effect of hyperglycemia and neuropeptides on interleukin-8 expression and angiogenesis in dermal microvascular endothelial cells. J Vasc Surg 2011; 53: 1654–1660.

77. Altun V, Hakvoort TE, van Zuijlen PP, van der Kwast TH, Prens EP. Nerve outgrowth and neuropeptide expression during the remodeling of human burn wound scars. A 7-month follow-up study of 22 patients. Burns 2001; 27: 717–722.

78. Dunnick C, Gibran N, Heimbach DM. Substance P has a role in neurogenic mediation of burn wound healing. J Burn Care Rehabil 1996; 17: 390–396.

79. Gibran NS, Tamura R, Tsou R, Isik FF. Human dermal microvascular endothelial cells produce nerve growth factor: implications for wound repair. Shock 2003; 19: 127–130.

80. Liang Y, Marcusson JA, Johansson O. Light and electron microscopic immunohistochemical observations of p75 nerve growth factor receptor-immunoreactive dermal nerves in prurigo nodularis. Arch Dermatol Res 1999; 291: 14–21.

81. Marconi A, Terracina M, Fila C, Franchi J, Bonté F, Romagnoli G, et al. Expression and function of neurotrophins and their receptors in cultured human keratinocytes. J. Invest. Dermatol 2003; 121: 1515–1521.

82. Palazzo E, Marconi A, Truzzi F, Dallaglio K, Petrachi T, Humbert P, et al. Role of neurotrophins on dermal fibroblast survival and differentiation. J Cell Physiol 2012; 227: 1017–1025.

83. Hecker-Kia A, Kolkenbrock H, Orgel D, Zimmerman B, Sparmann M, Ulbrich N. Substance P induces the secretion of gelatinase A from human synovial fibroblasts. Eur J Clin Chem Clin Biochem 1997; 35: 655–660.

84. Fujiwara T, Kubo T, Kanazawa S, Shingaki K, Taniguchi M, Matsuzaki S, et al. Direct contact of fibroblasts with neuronal processes promotes differentiation to myofibroblasts and induces contraction of collagen matrix in vitro. Wound Repair Regen 2013; 21: 588–594.

85. Cheret J, Lebonvallet N, Buhe V, Carre JL, Misery L, Le Gall-Ianotto C. Influence of sensory neuropeptides on human cutaneous wound healing process. J Dermatol Sci 2014; 74: 193–203.

86. Smith PG, Liu M. Impaired cutaneous wound healing after sensor denervation in developing rats: effects on cell proliferation and apoptosis. Cell Tissue Res 2002; 307: 281–291.

87. Jansco N, Jancso-Gabor A, Szolcsanyi J. Direct evidence for neurogenic inflammation and its prevention by denervation and by pretreatment with capsaicin. Br J Pharmacol Chemother 1967; 31: 138–151.

88. Souza BR, Cardoso JF, Amadeu TP, Desmoulière A, Costa AM. Sympathetic denervation accelerates wound contraction but delays reepithelialization in rats. Wound Repair Regen 2005; 13: 498–505.

89. Engin C, Demirkan F, Ayhan S, Atabay K, Baran NK. Delayed effect of denervation on wound contraction in rat skin. Plast Reconstr Surg 1996; 98: 1063–1067.

90. Carr RW, Delaney CA, Westerman RA, Roberts RG. Denervation impairs cutaneous microvascular function and blister healing in the rat hindlimb. Neuroreport 1993; 4: 467–470.

91. Fukai T, Takeda A, Uchinuma E. Wound healing in denervated rat skin. Wound Repair Regen 2005; 13: 175–180.

92. Chiang HY, Chen CT, Chien HF, Hsieh ST. Skin denervation, neuropathology, and neuropathic pain in a laser-induced focal neuropathy. Neurobiol Dis 2005; 18: 40–53.

93. Gibran NS, Jang YC, Isik FF, Greenhalgh DG, Muffley LA, Underwood RA, et al. Diminished neuropeptide levels contribute to impaired cutaneous healing response associated with diabetes mellitus. J Surg Res 2002; 108: 122–128.

94. Ishikawa S, Takeda A, Akimoto M, Kounoike N, Uchinuma E, Uezono Y. Effects of neuropeptides and their local administration to cutaneous wounds in sensory-impaired areas. J Plast Surg Hand Surg 2014; 48: 143–147.

95. Miller MS, Buck SH, Sipes IG, Yamamura HI, Burks TF. Regulation of substance P by nerve growth factor: disruption by capsaicin. Brain Res 1982; 250: 193–196.

96. Pornprom M, Muffley LA, Anthony JP, Spenny ML, Underwood RA, Olerud JE, et al. Nerve growth factor accelerates wound healing in diabetic mice. Wound Repair Regen 2004; 12: 44–52.

97. Li AK, Koroly MJ, Schattenkerk ME, Malt RA, Young M. Nerve growth factor: acceleration of the rate of wound healing in mice. Proc Natl Acad Sci U S A 1980; 77: 4379–4381.

98. Fahey TJ, Sadaty A, Jones WG 2nd, Barber A, Smoller B, Shires GT. Diabetes impairs the late inflammatory response to wound healing. J Surg Res 1991; 50: 308–313.

99. Polydefkis M, Hauer P, Sheth S, Sirdofsky M, Griffin JW, McArthur JC. The time course of epidermal nerve fibre regeneration: studies in normal controls and in people with diabetes, with and without neuropathy. Brain 2004; 127: 1606–1615.

100. Cheng C, Singh V, Krishnan A, Kan M, Martinez JA, Zochodne DW. Loss of innervation and axon plasticity accompanies impaired diabetic wound healing. PLoS One 2013; 8: e75877.

101. Lindberger M, Schroder HD, Schultzberg M, Kristensson K, Persson A, Ostman J, et al. Nerve fibre studies in skin biopsies in peripheral neuropathies. I. Immunohistochemical analysis of neuropeptides in diabetes mellitus. J Neurol Sci 1989; 93: 289–296.

102. Anand P, Terenghi G, Warner G, Kopelman P, Williams-Chestnut RE, Sinicropi DV. The role of endogenous nerve growth factor in human diabetic neuropathy. Nat Med 1996; 2: 703–707.

103. Faradji V, Sotelo J. Low serum levels of nerve growth factor in diabetic neuropathy. Acta Neurol Scand 1990; 81: 402–406.

104. Galkowska H, Olszewski WL, Wojewodzka U, Rosinski G, Karnafel W. Neurogenic factors in the impaired healing of diabetic foot ulcers. J Surg Res 2006; 134: 252–258.

105. Wallengren J, Badendick K, Sundler F, Håkanson R, Zander E. Innervation of the skin of the forearm in diabetic patients: relation to nerve function. Acta Derm Venereol 1995; 75: 37–42.

106. Antezana M, Sullivan SR, Usui M, Gibran N, Spenny M., Larsen J, et al. Neutral endopeptidase activity is increased in the skin of subjects with diabetic ulcers. J Invest Dermatol 2002; 119: 1400–1404.

107. Sheykhzade M, Dalsgaard GT, Johansen T, Nyborg NC. The effect of long-term streptozotocin-induced diabetes on contractile and relaxation responses of coronary arteries: selective attenuation of CGRP-induced relaxations. Br J Pharmacol 2000; 129: 1212–1218.

108. Basson MD, Burney RE. Defective wound healing in patients with paraplegia and quadriplegia. Surg Gynecol Obstet 1982; 155: 9–12.

109. Claus-Walker J, Halstead LS. Metabolic and endocrine changes in spinal cord injury. Part II. Section 1. Consequences of partial decentralization of the autonomic nervous system. Arch Phys Med Rehabil 1982; 63: 569–575.

110. Rappl LM. Physiological changes in tissues denervated by spinal cord injury tissues and possible effects on wound healing. Int. Wound J 2008; 5: 435–444.

111. Ramos MV, Freed MM, Kayne HL. Resting blood pressures of spinal cord injured patients. Sci Dig 1981; 3: 19–25.

112. Hunt TK, Conolly WB, Aronson SB, Goldstein P. Anaerobic metabolism and wound healing: an hypothesis for the initiation and cessation of collagen synthesis in wounds. Am J Surg 1978; 135: 328–332.

113. Knighton DR, Silver IA, Hunt TK. Regulation of wound-angiogenesis: effect of oxygen gradients and inspired oxygen concentration. Surgery 1981; 90: 262–270.

114. Claus-Walker J, Halstead LS. Metabolic and endocrine changes in spinal cord injury. Part I. The nervous system before and after transaction of the spinal cord. Arch Phys Med Rehabil 1981; 62: 595–601.

115. Gerstein AD, Phillips TJ, Rogers GS, Gilchrest BA. Wound healing and aging. Dermatol Clin 1993; 11: 749–757.

116. Khalil Z, Ralevic V, Bassirat M, Dusting GJ, Helme RD. Effects of ageing on sensory nerve function in rat skin. Brain Res 1994; 641: 265–272.

117. Sgonc R, Gruber J. Age-related aspects of cutaneous wound healing: a mini-review. Gerontology 2013; 59: 159–164.

118. Cabalar M, Yayla V, Ulutas S, Senadim S, Oktar A.C. The clinical & neurophysiological study of leprosy. Pak J Med Sci 2014; 30: 501–506.

119. Widgerow AD, Kalaria S. Pain mediators and wound healing--establishing the connection. Burns 2012; 38: 951–959.

120. Garg UC, Hassid A. Nitric oxide-generating vasodilators inhibit mitogenesis and proliferation of BALB/C 3T3 fibroblasts by cyclic GMP-independent mechanism. Biochem Biophys Res Commun 1990; 171: 474–479.

121. Scott JR, Muangman P, Gibran NS. Making sense of hypertrophic scar: a role for nerves. Wound Repair Regen 2007; 15: 27–31.

122. Sio SWS, Puthia MK, Lu J, Moochhala S, Bhatia M. The neuropeptide substance P is a critical mediator of burn induced acute lung injury. J Immunol 2008; 180: 8333–8341.

123. Enoch S, Harding K. Wound bed preparation: the science behind the removal of barriers to healing. Wounds 2003; 15: 213–229.

124. Brain SD, Grant AD. Vascular actions of calcitonin gene related peptide and adrenomedullin. Physiol Rev 2004; 84: 903–934.

125. Rizk M, Witte MB, Barbul A. Nitric oxide and wound healing. World J Surg 2004; 28: 301–306.

126. McGuire L, Heffner K, Glaser R, Needleman B, Malarkey W, Dickinson S, et al. Pain and wound healing in surgical patients. Ann Behav Med 2006; 31: 165–172.

127. Colwell AS, Longaker MT, Lorenz HP. Mammalian fetal organ regeneration. Adv Biochem Eng Biotechnol 2005; 93: 83–100.

128. Kishi K, Nakajima H, Tajima S. Differential responses of collagen and glycosaminoglycan syntheses and cell proliferation to exogenous transforming growth factor beta 1 in the developing mouse skin fibroblasts in culture. Br J Plast Surg 1999; 52: 579–582.

129. Kishi K, Ohyama K, Satoh H, Kubota Y, Tanaka T, Imanishi N, et al. Mutual dependence of murine fetal cutaneous regeneration and peripheral nerve regeneration. Wound Repair Regen 2006; 14: 91–99.

130. Henderson J, Terenghi G, Ferguson MW. The reinnervation and revascularisation pattern of scarless murine fetal wounds. J Anat 2011; 218: 660–667.

131. Egar MW. Accessory limb production by nerve-induced cell proliferation. Anat Rec 1988; 221: 550–564.

132. Stelnicki EJ, Doolabh V, Lee S, Levis C, Baumann FG, Longaker MT, et al. Nerve dependency in scarless fetal wound healing. Plast Reconstr Surg 2000; 105: 140–147.

133. Terenghi G, Sundaresan M, Moscoso G, Polak JM. Neuropeptides and a neuronal marker in cutaneous innervation during human foetal development. J Comp Neurol 1993; 328: 595–603.

134. Zhang HS, Ren HT. Correlation of substance P and mast cells with scarless wound healing in fetal skin. Journal of clinical and rehabilitative tissue engineering research 2007; 14: 2648–2651.

135. Xie J, Lai XN, Wang Z, Wang L, Xiang D, Huang Z, et al. Changes in neuropeptide substance P and calcitonin gene-related peptide in scarless wound healing in fetal rabbit skin. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue 2005; 17: 76–79.

136. Feedar JA, Kloth LC, Gentzkow GD. Chronic dermal ulcer healing enhanced with monophasic pulsed electrical stimulation. Phys Ther 1991: 71: 639–649.

137. Gentzkow GD, Pollack SV, Kloth LC, Stubbs HA. Improved healing of pressure ulcers using dermapulse, a new electrical stimulation device. Wounds 1991: 3: 158–170.

138. Singh B, Xu QG, Franz CK, Zhang R, Dalton C, Gordon T, et al. Accelerated axon outgrowth guidance, and target reinnervation across nerve transection gaps following a brief electrical stimulation paradigm. J Neurosurg 2012; 116: 498–512.

139. Kao CH, Chen JJ, Hsu YM, Bau DT, Yao CH, Chen YS. High-frequency electrical stimulation can be a complementary therapy to promote nerve regeneration in diabetic rats. PLoS One 2013; 8: e79078.

140. Khalil Z, Merhi M. Effects of aging on neurogenic vasodilator responses evoked by transcutaneous electrical nerve stimulation: relevance to wound healing. J Gerontol A Biol Sci Med Sci 2000; 55: 257–263.

141. Ogrin R, Darzins P, Khalil Z. The use of sensory nerve stimulation and compression bandaging to improve sensory nerve function and healing of chronic venous leg ulcers. Curr Aging Sci 2009; 2: 72–80.

142. Geremia NM, Gordon T, Brushart TM, Al-Majed AA, Verge VM. Electrical stimulation promotes sensory neuron regeneration and growth-associated gene expression. Exp Neurol 2007; 205: 347–359.

143. Nguyen HT, Sapp S, Wei C, Chow JK, Nguyen A, Coursen J, et al. Electric field stimulation through a biodegradable polypyrrole-co-polycaprolactone substrate enhances neural cell growth. J Biomed Mater Res A 2014; 102: 2554–2564.

144. Brushart TM, Jari R, Verge V, Rohde C, Gordon T. Electrical stimulation restores the specificity of sensory axon regeneration. Exp Neurol 2005; 194: 221–229.

145. Wood MD, Willits RK. Applied electric field enhances DRG neurite growth: influence of stimulation media, surface coating and growth supplements. J Neural Eng 2009 Aug; 6: 046003.

146. Merhi M, Helme R, Khalil Z. Age-related changes in sympathetic modulation of sensory nerve activity in rat skin. Inflamm Res 1998; 47: 239–244.