Neuroendocrinology Of The Skin

  • November 2019
  • PDF

This document was uploaded by user and they confirmed that they have the permission to share it. If you are author or own the copyright of this book, please report to us by using this DMCA report form. Report DMCA


Overview

Download & View Neuroendocrinology Of The Skin as PDF for free.

More details

  • Words: 28,804
  • Pages: 31
0163-769X/00/$03.00/0 Endocrine Reviews 21(5): 457– 487 Copyright © 2000 by The Endocrine Society Printed in U.S.A.

Neuroendocrinology of the Skin* ANDRZEJ SLOMINSKI

AND

JACOBO WORTSMAN

Department of Pathology (A.S.), University of Tennessee, Memphis, Tennessee 38163; and Department of Medicine (J.W.), Southern Illinois University, Springfield, Illinois ABSTRACT The classical observations of the skin as a target for melanotropins have been complemented by the discovery of their actual production at the local level. In fact, all of the elements controlling the activity of the hypothalamus-pituitary-adrenal axis are expressed in the skin including CRH, urocortin, and POMC, with its products ACTH, ␣-MSH, and ␤-endorphin. Demonstration of the corresponding receptors in the same cells suggests para- or autocrine mechanisms of action. These findings, together with the demonstration of cutaneous production of numerous other hormones including vitamin D3, PTHrelated protein (PTHrP), catecholamines, and acetylcholine that share regulation by environmental stressors such as UV light, un-

derlie a role for these agents in the skin response to stress. The endocrine mediators with their receptors are organized into dermal and epidermal units that allow precise control of their activity in a field-restricted manner. The skin neuroendocrine system communicates with itself and with the systemic level through humoral and neural pathways to induce vascular, immune, or pigmentary changes, to directly buffer noxious agents or neutralize the elicited local reactions. Therefore, we suggest that the skin neuroendocrine system acts by preserving and maintaining the skin structural and functional integrity and, by inference, systemic homeostasis. (Endocrine Reviews 21: 457– 487, 2000)

I. Introduction II. Structure of the Skin A. Developmental biology B. Anatomy and histology C. Physiology III. Skin as a Target for Neuroendocrine Signals A. CRH and urocortin receptors (CRH-R) B. Melanocortin receptors (MC-R) C. Opioid receptors D. GH receptor (GH-R) E. PRL and LH/CG receptors (LH/CG-R) F. Neurokinin receptors (NK-R) G. Calcitonin gene-related peptide receptor (CGRP-R) H. Vasoactive intestinal peptide receptor (VIP-R) I. Neutrophin (NT) receptors J. Miscellaneous neuropeptide receptors K. PTH and PTH-related protein (PTHrP) receptors L. Vitamin D receptor (VDR) M. Glucocorticoid and mineralocorticoid receptors N. Androgen and estrogen receptors O. Thyroid hormone receptors P. Cholinergic receptors Q. Adrenergic receptors R. Glutamate receptors S. Serotonin receptors T. Histamine receptors U. Miscellaneous receptors IV. Skin as a Source of Hormones and Neurotransmitters A. PTHrP

V. VI.

VII.

VIII.

B. Hypothalamic and pituitary hormones C. Neuropeptides and neurotrophins D. Neurotransmitters/neurohormones E. Thyroid hormones F. Sex steroid hormones G. Other steroid hormones Molecular and Structural Basis for the Organizational Integration of Neuroendocrine Elements of the Skin Regulation of Cutaneous Neuroendocrine System A. Solar radiation B. Hair cycle C. Cytokines D. Degradation or inactivation of hormones and neurotransmitters Regulation of Cutaneous Vitamin D Production A. Vitamin D3 production B. Precutaneous regulation C. Cutaneous regulation D. Postcutaneous regulation E. General comments Final Comments and Future Directions I. Introduction

T

he skin is the largest body organ and functions as a metabolically active biological barrier separating internal homeostasis from the external environment. Depending on anatomic localization and environmental influences, the skin shows remarkable functional and structural diversity (1– 4), since it is continuously exposed to fluctuating external information represented by solar and thermal radiation, mechanical energy, changes in humidity, and/or chemical and biological insults. The maintenance of skin structural integrity is therefore critical and must be served by rapid mechanisms to restore the barrier properties of the epidermis when disrupted by external trauma. Maintenance

Address reprint requests to: Andrzej Slominski, M.D., Ph.D., Department of Pathology, RM576-BMH Main, University of Tennessee, 899 Madison Avenue, Memphis, Tennessee 38163. E-mail: [email protected] * This work was supported by grants from National Science Foundation (NSF) (IBN-9604364, IBN-9896030, and IBN-9405242), and American Cancer Society, Illinois Division (no. 99 –51) to A.S., and internal funding from the Department of Pathology, University of Tennessee.

457 Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

458

SLOMINSKI AND WORTSMAN

of organ, and hence systemic homeostasis, requires a special cutaneous property, the capability to recognize and integrate appropriate signals with a high degree of specificity. Such sensory mechanism must be widely distributed, efficiently self-regulated in intensity and field of activity, and endowed with the capability of differentiating environmental noise from biologically relevant signals (5, 6). To some extent, these mechanisms are represented by the skin immune system, activated by biological insults or trauma (7); and in humans, by the pigmentary system, activated or modified by solar radiation (8 –10). However, as presented in this work, the main component in this critical skin function is the level of activity of the local neuroendocrine system. Over the last decade, it has become increasingly apparent that the skin, particularly the epidermis, has powerful metabolic and endocrine capabilities (11, 12). For example, the skin synthesizes vitamin D, which enters the circulation and, upon activation, exerts profound metabolic and endocrine effects (13, 14). Resident skin cells also synthesize and release the hormones parathyroid hormone-related protein (PTHrP) (15), POMC-derived MSH, ACTH, and ␤-endorphin peptides (5, 16, 17), the CRH and urocortin peptides (18, 19), the neurotransmitters catecholamines and acetylcholine (20, 21), and precursors to biogenic amines (9, 21–23). While production of some of these factors is not constitutive, it does respond to specific inductive stimuli. The skin is also a site for activation of steroid hormones such as the conversion of testosterone to 5␣-dihydrotestosterone or to estradiol, or the conversion of T4 to T3 (4, 11, 12). These locally generated hormones and neurotransmitters can act in a paracrine or autocrine fashion. Moreover, the presence of numerous nerve endings and a rich vascular network provide additional mechanisms for the expression of neuroendocrine functions, e.g., transmission of regulatory signals to the global or central systems via the vascular system, or through the afferent neural network. This emerging concept, of skin as a neuroendocrine organ, is a relatively new addition to the field of cutaneous biology; it combines concepts from immunology, endocrinology, and neurobiology to unravel the multidirectional communications between brain, the endocrine and immune systems, and peripheral organs (24 –28). In this regard, the skin has a unique role because of its location, size, and relative functional diversity. Moreover, cutaneous signals sent to neuroendocrine centers may play modulatory roles, although peripheral intraorgan or intersystemic communications are also necessary to maintain global and local homeostasis. We will presently review evidence on the production of hormones and neurotransmitters by the skin and on the expression of the corresponding receptors. Cutaneous regulation of neuroendocrine communication will be analyzed and its function discussed within the context of organ homeostasis. Data on the experimental characterization of receptors for neurohormones in skin cells will be reviewed, including regulation of expression, ligand production, and characterization of signal transduction pathways. Pertinent data will be included with the understanding that the mere presence of a substance in a culture system of skin cells does not necessarily imply that the substance has physiological relevance in vivo. Special attention will be given to intraorgan communication and to the potential systemic or central effects of skin-produced factors acting on

Vol. 21, No. 5

sensory nerves, or skin-activated circulating cells, or through direct release into the circulation as neurohormones or mediators. This review will end by setting the stage for future basic and clinical research. II. Structure of the Skin A. Developmental biology

The epithelial skin structures, e.g., epidermis, hair follicle, and sebaceous, apocrine, and eccrine glands, all derive from the embryonal outer epithelium, which originates from ectoderm (29). Nonkeratinocytic cells of the epidermis and hair follicle that include melanocytes and Merkel cells are also of ectodermal origin, but melanocytes migrate to the epidermis from the neural crest (1, 3, 4, 29). Cell populations of mesodermal origin comprise the Langerhans cells and the T lymphocytes, which include the T␥␦ type expressed in mouse epidermis and hair follicle and the sparse, mostly T␣␤ cells expressed in human epidermis (7). All of the dermal components are of mesodermal origin, with the exception of nerves and specialized sensory receptors that develop from the ectoderm (1, 3, 4, 7, 29). The dermal cellular populations include fibroblasts/fibrocytes/myofibroblasts, adipocytes, monocytes/macrophages, mast cells, Langerhans cells, T lymphocytes, dendrocytes, smooth muscle cells, and vascular and lymphatic endothelial cells. Fibrocytes arise by differentiation of stellate mesenchymal cells present in the primordial dermis, whereas adipocytes differentiate from subdermal mesenchymal cells that surround newly formed blood vessels. Macrophages, mast cells, Langerhans cells, and dendrocytes migrate to the skin from the bone marrow. Formation of the adnexal structures results from precise mesenchymal epithelial interactions producing down growth of primordial adnexal structures to reach the reticular dermis and subcutis (1, 3, 4, 29 –31). The multidirectional interaction between cells of ectodermal and mesodermal origin results in a cohesive unified skin structure that, nevertheless, maintains a degree of heterogeneity expressed by marked regional differences (1, 4, 29). In the context of this review it must be noted that brain, peripheral nervous system, retina, and medulla of adrenal gland are also of ectodermal origin, whereas olfactory epithelium and olfactory nerves, anterior lobe of hypophysis, and epithelial elements of the mammary gland all derive from the outer epithelium (29). Mesodermal structures include the immune system, endothelium of blood vessels, adrenal cortex, and gonadal epithelium and stroma (7, 29). These embryologic associations may determine the potential capability for resident skin cells to produce molecules similar to their close or distant relatives. Thus, cellular lineage may predict neuroendocrine functional activity. B. Anatomy and histology

The skin is composed of two main compartments: the epidermis with the adnexal epithelial structures and the dermis with the nonepithelial elements of adnexa (1– 4). While not a skin component, the subcutaneous fat is closely related to the skin anatomically and functionally. Structure and

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

thickness of both epidermis and dermis vary according to anatomic location; thus, the average thickness of the epidermis is 0.1 mm, but in the acral areas is up to 1.6 mm thick. The latter regions contain thick cornified and granular layers and numerous eccrine units and nerve endings but lack folliculosebaceous-apocrine units. In contrast, facial skin contains numerous vellus follicles with prominent sebaceous glands; skin in the axilla and groins is characterized by numerous apocrine glands, back skin has very thick reticular dermis, and scalp skin contains large terminal hair follicles routed deep into the subcutaneous fat. In furry animals, terminal hair follicles cover most of the body, serving as insulating cover and as touch organs (30 –32). The basal membrane zone separates the epidermis and epithelial adnexal structures from the dermis. Beneath the basement membrane is a thin zone of adventitial dermis that comprises the papillary dermis, between the epidermal folds and the periadnexal dermis surrounding adnexal structures. The papillary dermis is characterized by thin collagen bundles interspersed with elastic fibers, frequent fibrocytes, abundant matrix, and a rich vascular network composed predominantly of capillaries. The reticular dermis is composed predominantly of thick collagen bundles and elastic fibers and a lower concentration of stromal matrix, with comparatively fewer fibrocytes; there are also blood vessels, and adipocytes that extend upward from the subcutaneous fat. The skin immune system is composed of resident, recruited, and recirculating cell populations (7). The resident population is constitutively expressed in the skin under physiological conditions. This is represented by keratinocytes, fibroblasts, vascular and lymphatic endothelial cells, mast cells, tissue macrophages (histiocytes), T lymphocytes, and dendritic cells. The recruited population comprises monocytes, basophilic, neutrophilic, and eosinophilic granulocytes, as well as mast cells and T and B lymphocytes. The recirculating cell population is represented by dendritic cells, natural killer cells, and T lymphocytes. Recruited or recirculating cells reach the skin via circulation. The vasculature is arranged into a superficial (subpapillary) plexus, located in the upper reticular dermis, and a deep plexus positioned in the lower reticular dermis (1– 4). These plexuses are connected by communicating blood vessels, which are most numerous in the upper dermis and around folliculosebaceous and eccrine units. The vascular network provides rich capillary supply for the dermal papillae and periadnexal dermis. A lymphatic network accompanies the vascular bed, although it is a functionally separate entity. The skin contains an extensive neural network represented by cholinergic and adrenergic nerves and by myelinated and unmyelinated sensory fibers (1, 3, 4, 33, 34). The autonomic nerves supply arterioles, glomus bodies, hair erector muscles, and apocrine and eccrine glands. The terminal endings of sensory fibers are either surrounded by histologically distinctive structures, such as Pacini and Meissner’s corpuscles, Ruffini organs, Merkel disks, and mucocutaneous end organs, or supply directly individual Merkel cells. A rich network of free sensory endings surround and penetrate hair follicles, pilosebaceous units, eccrine and apocrine glands, papillary dermis, and epidermis. The sensory and autonomic

459

networks do show regional differences according to anatomic sites and also have topographical specificity by distributing into well defined areas called dermatomes. The torso, extremities, posterior scalp, and neck are supplied by sensory nerves arising from dorsal root ganglia of the spinal cord, whereas face, most of the scalp, and upper anterior neck are innervated by trigeminal nerve branches. C. Physiology (1–22, 30 –35)

The most important function of the skin, determined by its location at the interface between external and internal environments, is that of physical barrier. This is established in the epidermis by a precisely regulated gradient of keratinocyte differentiation stages, which forms a highly impermeable protein-lipid layer at the outer-most segment. This layer prevents the destruction of living keratinocytes by environmental factors and reduces or minimizes water evaporation, maintaining a liquid environment to preserve skin structural integrity in the face of frequent mechanical trauma. The epidermal pigmentary system protects the skin against the damaging effect of solar radiation in humans and, in conjunction with the follicular pigmentary system, determines hair and skin color that play an important role in social communication and camouflage in many mammalian species. The epidermal and dermal immune elements provide defense against biological insults, and they are also involved in the integration of the response to foreign and self-antigens through interactions with the central immune system. Immune responses are involved in the reaction to viral or microbial infections, or to cancer development; dysregulated immune responses may be pathogenic in autoimmune diseases. The adnexal organs are epidermally derived structures that extend into dermis and subcutis. Their functional role is pleiotropic by participating in the formation of hair shafts from hair follicle, serving protective, thermoregulatory, and sensory (touch) functions, as well as being involved in social communication. The secretion of eccrine, apocrine, and sebaceous glands is important for thermoregulation, for preservation of the integrity of the physical barrier, for regulation of electrolyte balance, and for secretion of the pheromones and odorant-affecting behavior. The dermis, in addition to its structural role, is involved in mechanical protection and thermoregulation via its rich vascular network. The skin also provides the sensory reception for touch, pressure, vibration, temperature, pain, and pleasure through a neural network comprised of specialized receptors and free nerve endings. Finally, the skin, as a regulator of metabolism, transforms various hormones and can also inactivate potentially harmful substances of exogenous or endogenous origin. III. Skin as a Target for Neuroendocrine Signals

Skin resident and circulating immune cells express receptors for neuropeptides and neurotransmitters identical to those expressed in the central neuroendocrine systems. Examples of those receptors and their expression sites are listed in Table 1. Clinical observations made in diverse endocrine disorders associated with cutaneous changes also confirm that the skin is a target for hormones, neurohormones, and neurotransmitters (3– 6, 12, 16 –21, 35–38). That the skin is

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

460

SLOMINSKI AND WORTSMAN

Vol. 21, No. 5

TABLE 1. Selected hormone and neurotransmitter receptors expressed in keratinocytes and melanocytes Cell type

Receptor repertoire

Keratinocytes

CRH-R1, MC1-R, ␮- and ␨-opioid-R, PRL-R, LH/CG-R, GH-R, CGRP-R, VIP-R, neurokinin-R, class II PTH/PTHrP-R, vitamin D-R, androgen-R, estrogen-R, glucocorticoid-R, mineralocorticoid-R, muscarinic-R, nicotinic-R, adrenoreceptors, glutamate-R, gastrin-releasing peptide-R, NPY-R, purinoreceptors, H1 and H2 histamine-R, somatostatin-R (?), bombesin-R, (?) CRH-R1, MC1-R, LH/CG-R, GH-R, CGRP-R, VIP-R, vitamin D-R, androgen-R estrogen-R, glucocorticoidR, adrenoreceptors, muscarinic-R, H2 histamine-R

Melanocytes

R, Receptor; (?), possible expression.

also a target of neural responses is supported by studies showing neural contributions to the etiology and clinical manifestations of inflammatory skin diseases and vitiligo (3, 4, 33, 34, 38, 39). A. CRH and urocortin receptors (CRH-R)

This group comprises the G protein-coupled membranebound CRH-R1 and CRH-R2 receptors (5, 40, 41), whose gene expression was recently documented in human and rodent skin (5, 18, 40, 42– 46). CRH-R1 expression has been detected in epidermal and follicular keratinocytes, melanocytes, and mast cells (5, 44, 46 – 49), and it is possible that these cells may coexpress CRH-R2 (5, 44, 46, 50, 51). While specific CRH binding sites were additionally seen in dermal fibroblasts, endothelial cells, and smooth muscle of blood vessels (5, 40, 44, 46, 52–54), it remains to be tested whether these binding sites represent CRH-R1, CRH-R2, or coexpression of both receptors. Signal transduction through cutaneous CRH receptors is linked to stimulation of cAMP production and increase of cytosolic Ca levels (46, 47, 50, 51). It remains to be tested whether other pathways coupled to different receptor subtypes specific for skin cells are also activated (46, 50). CRH and urocortin have a recognized role in skin pathophysiology through their actions on the skin immune system (5, 40, 48, 55–59). Thus, in the periphery, CRH can act as a proinflammatory agent (48, 55, 56) and, together with urocortin, induces degranulation of mast cells (48, 57). However, antiinflammatory effects have been also demonstrated in models of tissue injury, e.g., in thermally injured skin where local injection of CRH has an antiedema effect independent of hypothalamus-pituitary-adrenal (HPA) axis function, and in doxorubicin-induced eye lid inflammation that is reduced in severity by pretreatment of the eyelid with CRH (58 – 60). In addition, CRH has antinociceptive activity and accelerates wound healing (58 – 60). CRH and urocortin also inhibit proliferation of keratinocytes (51) and either stimulate or inhibit melanoma cell proliferation depending on culture conditions (Refs. 46 and 49 and A. Slominski and B. Zbytek, unpublished data). B. Melanocortin receptors (MC-R)

The membrane-bound G protein-coupled melanocortin receptors of type 1, 2, and 5 (MC1-R, MC2-R, MC5-R) have been identified in the skin (5, 8, 17, 61– 69). MC1-R was detected in melanocytes, keratinocytes, sebocytes, fibroblasts, endothelial cells, Langerhans cells, and dermal immune cells, while MC5-R was detected in the epithelial cells of eccrine, apocrine, and sebaceous glands (5, 8, 17, 61– 64). Although expression of the MC2-R gene has been detected in human

and mouse skin (65, 66), precise cell compartment(s) assignment will require further testing; possible expression sites include adipocytes, keratinocytes, and melanocytes (63– 66). The MC receptors are activated by ACTH, and by ␣-, ␤and ␥-MSH; ligand affinity varies according to receptor type and mammalian species (5, 8, 17, 61, 70). Signal transduction through MC1-R, MC2-R, and MC5-R has been linked to activation of adenylate cyclase (5, 8, 17, 61, 67– 69). The best recognized phenotypic effect of the POMCderived ACTH and MSH peptides is the stimulation of melanogenesis and its switching from pheo- to eumelanogenesis (5, 8, 16, 17, 61, 67–72), which can also be documented clinically (3–5, 8, 67, 72). There is a general agreement that ACTH, ␣-MSH, and ␤-MSH have the strongest melanogenic activity (8, 16, 67– 69). ␥-MSH peptides have low intrinsic melanogenic activity in human normal melanocytes and rodent malignant melanocytes (70, 71), similar to findings in frog and lizard melanophores (8, 67, 68, 73). However, it is possible that selected ␥-MSH peptides such as ␥2 and ␥3 could still modulate pigmentation indirectly, modifying cellular responses to the other melanotropins (71). Studies in cultured normal and malignant melanocytes show that MSH and ACTH, acting via cAMP-dependent pathways (5, 8, 16, 17, 61, 67– 69, 72, 74 –79), stimulate the expression and activity of enzymatic, structural, and regulatory proteins involved in melanogenesis (5, 8, 67– 69). Depending on species and cellular genotype, MSH and ACTH can inhibit or stimulate proliferation of malignant melanocytes (8, 67– 69, 72, 74 –78). However, most authors agree that in normal human melanocytes, ACTH and MSH act as stimulators of cell proliferation (8, 69, 79), dendrite production (5, 8, 67– 69, 74), and melanocyte migration (8) and decrease expression of intercellular adhesion molecule-1 (ICAM-1) (80). Epidermal, adnexal, vascular, and dermal structures represent additional targets for POMC peptides (5, 17, 35, 61– 64). Thus, ␣-MSH can modify proliferation and differentiation of keratinocytes as well as their immune activity and regulate activity of dermal fibroblast (5, 17, 64, 81, 82). In endothelial cells, ␣-MSH may play a crucial role decreasing their adherence and the transmigration of inflammatory cells, a prerequisite step for immune and inflammatory reactions. ␣-MSH and ACTH have strong immunomodulating activity in the skin that results in an overall immunosuppressive effect (5, 17, 64, 82). For example, ␣-MSH acts as antagonist to interleukin-1 (IL-1) suppressing production of proinflammatory cytokines while it induces production of the immunosuppressive cytokine IL-10. ␣-MSH is also capable of suppressing accessory molecule expression on antigen-presenting cells and may thereby serve as one of the

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

signals responsible for anergy or tolerance induction (17, 64, 82). In addition to the regulation of hair pigmentation, ␣-MSH and ACTH have other actions on adnexal structures (5, 17, 35, 62, 83, 84). For example, in mink and mice, ACTH acts as inducer of anagen development (85, 86), whereas in mouse anagen skin it induces premature onset of catagen (83). ACTH and ␣-MSH also influence sebaceous gland function (35, 84): ␣-MSH specifically stimulates sebum secretion and lipogenesis in cutaneous sebaceous glands, enhancing wax and sterol ester biosynthesis, and stimulating production and release of female sex attractant odors and of male aggression-promoting pheromones (by specialized preputial glands) (35). ␣-MSH and perhaps ACTH may be important in overall rodent skin thermoregulation, by preventing overwetting of hairs, and in behavior regulation through its action on nonspecialized and specialized sebaceous glands (35, 84). It is likely that MSH and ACTH peptides also affect function of human sebaceous glands. C. Opioid receptors

␮-Opioid receptors, which bind with high-affinity ␤endorphin, were detected in cultured human epidermal keratinocytes (87). Further investigations using in situ hybridization and immunocytochemistry on skin biopsy specimens showed that the receptors are localized to keratinocytes in the epidermis and outer root sheath of the hair follicles, to the peripheral epithelial cells in sebaceous glands, and to the secretory component in sweat glands (87). The related ␨opioid receptor that binds enkephalins with high affinity has also been detected in human and mouse epidermal keratinocytes (88). Met-enkephalin has been shown to inhibit proliferation of mouse epidermal keratinocytes in vivo, in a circadian pattern (88), and both met- and leu-enkephalins can inhibit differentiation of human keratinocytes in vitro (89). In addition, ␤-endorphin and enkephalins have antinociceptive and immunomodulatory properties (7, 24, 25, 27, 28).

461

hyperpigmentation, eccrine and apocrine hyperhidrosis, increased sebum secretion, growth of pedunculated fibromas, and thickening and hardening of the nails (1–3, 36, 37, 68). Transgenic mice overexpresssing GH show skin overgrowth with increased dermal thickness, significant dermal fibrosis, and replacement of subcutaneous adipose tissue by fibrous tissue (97). E. PRL and LH/CG receptors (LH/CG-R)

Receptors for the pituitary hormones PRL, LH, and human CG (hCG) are also expressed in the skin (98 –103). PRL receptors have been localized in rat epidermal and follicular keratinocytes by in situ hybridization (98) and in ovine dermal papilla fibroblasts and follicular keratinocytes with a radioligand binding assay (99). PRL binding sites exhibiting high affinity for the ligand were identified in membrane preparation from mink skin; the highest concentration of binding sites was found during the winter fur growth cycle (100). PRL stimulation of cultured human keratinocytes proliferation has been linked to the expression of high-affinity PRL binding sites on the cell surface (101). PRL can directly and indirectly modulate the hair growth, shedding, and molting cycle of furry animals, whereas in humans hyperprolactinemia has been associated with hirsutism (3, 31, 104 – 106). It has also been proposed that PRL participates in the regulation of sebaceous gland activity, since acne vulgaris can be associated with idiopathic hyperprolactinemia in the absence of altered androgen concentrations (3, 106). PRL has potent immunomodulatory properties (107), suggesting that it can also regulate functions in the skin immune system. Normal human skin also contains the mRNAs for the LH/CG-R and a 66-kDa protein capable of binding 125I-hCG (102). These receptors were detected in the epidermis, inner and outer root sheaths of the anagen hair follicle, sebaceous glands, and eccrine glands (103). Testing for expression of FSH receptors in normal human skin found it to be below the level of detectability (103).

D. GH receptor (GH-R)

The GH-R has been detected in human and rodent skin in epidermis, hair follicle, eccrine glands, dermal fibroblasts, adipocytes, and in Schwann and muscle cells (90 –93). Transcription of the GH-R gene has also been detected in cultured human melanocytes (91). These findings suggest that epidermal, adnexal, and dermal cell populations can be direct targets for GH. For example, GH can stimulate differentiation of rat sebocytes and modify melanocyte proliferation (94, 95). However, the phenotypic effects could also arise from an indirect effect such as the stimulation of cutaneous cells to produce insulin growth factor-1 (IGF-1). The cutaneous phenotypic effects of GH have been thoroughly described in patients with acromegaly, whose skin thickness increases considerably and acquires a doughy texture (1–3, 36, 37, 68). This effect is accompanied by increased fibroblasts activity and dermal glycosaminoglycans deposition that promotes water retention (96). Additional cutaneous signs of acromegaly are acanthosis nigricans, hypertrichosis with exception of the beard region,

F. Neurokinin receptors (NK-R)

Expression of the G protein-coupled neurokinin receptors NK-1R, NK-2R, and NK-3R has been reported in human and rodent skin (38, 108); however, others have detected only NK1-R in extracts from human skin (109). Either substance P (SP) or neurokinins A and B (NKA and NKB) could activate these receptors, through signal transduction pathways involving adenylate cyclase and phospholipases C and A2 (33, 34, 38). Human and rodent keratinocytes and endothelial cells express NK-1R to NK-3R, and mast cells, fibroblasts, and Langerhans cells express NK-1R (38). Activation of these receptors stimulates proliferation of keratinocytes, fibroblasts, and endothelial cells and neovascularization (4, 7, 33, 34, 38, 108 –113). NKA and SP stimulate mast cells release of histamine and tumor necrosis factor-␣ (TNF␣), and keratinocyte and endothelial cell function with production and release of proinflammatory cytokines, and expression of adhesion molecules (7, 33, 34, 38, 108 –115). SP stimulates hair growth in rodents (116).

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

462

SLOMINSKI AND WORTSMAN

Vol. 21, No. 5

G. Calcitonin gene-related peptide receptor (CGRP-R)

J. Miscellaneous neuropeptide receptors

There is functional evidence that the G protein-coupled receptor CGRP-R is expressed in skin cells (3, 4, 7, 33, 38, 108, 110 –114, 117–120). CGRP is a potent vasodilator of small and large vessels, at least partly through direct activation of arteriolar smooth muscle cell receptors. CGRP also increases vascular permeability, producing dermal edema through indirect activation of mast cells or through stimulation of nitric oxide (NO) production by endothelial cells with consequent vasodilatation (4, 33, 38, 108, 114). CGRP stimulates endothelial cell, keratinocyte, and melanocyte proliferation (117, 119). Stimulation of keratinocyte proliferation has been linked to direct activation of adenylyl cyclase activity (117), and CGRP induces keratinocyte production and release of promelanogenic factors (120). CGRP modifies the activity of the skin immune system (7, 38, 110 –114, 118).

Somatostatin is known to affect skin immune functions and basal secretion of histamine and, thus, immune cells and keratinocytes probably express the corresponding receptors (38, 110 –112, 133). Similarly, the inhibition of cAMP production by neuropeptide Y (NPY) in human keratinocytes, the stimulation of keratinocyte DNA synthesis by bombesin, and the acceleration of skin wound healing by TSH (12, 33, 38, 110 –112, 117) suggest that specific receptors for these hormones may be also expressed in skin. There are, in fact, data showing that the gene coding for the TSH receptor is expressed in adipocytes and fibroblasts (134). Receptors for bombesin and somatostatin have been shown in dermal fibroblasts (33, 38, 111, 112).

H. Vasoactive intestinal peptide receptor (VIP-R)

VIP-Rs are present in skin, and their activity is linked to G protein-coupled stimulation of adenylyl cyclase activity with cAMP production (3, 4, 7, 33, 38, 108, 110 –113, 117, 118). VIP biding sites have been characterized in malignant human melanocytes (121), where VIP stimulates cAMP production (122).VIP also stimulates keratinocyte proliferation and sweat production (4, 38, 117). Peptide histidine-methionine (PHM) and GH releasing factor (GFR) have stimulatory effects on keratinocyte cAMP production and cell proliferation, thought to be mediated through the VIP-R (38, 117). Indirectly, VIP also participates in the wheal and flare reaction through the activation of mast cells histamine release, or through NO-induced vasodilatation (3, 4, 7, 33, 38, 108, 111, 112). I. Neurotrophin (NT) receptors

Both human and rodent skin express transmembrane receptor proteins of the tyrosine kinase (Trk) and p75 panneurotrophin (p75NTR) families, which show high and low affinity for neuron growth factor (NGF), respectively (33, 38, 108, 123–131). The high-affinity receptors for NGF and NT4 include TrkA and TrkB; TrkC serves as a high-affinity receptor for NT3, which also binds, but with low affinity, to TrkA and TrkB. The receptors for the Trk family and for p75NTR are expressed in epidermal and follicular keratinocytes, epidermal melanocytes, specialized dermal fibroblasts, mast cells, immunocytes, and cutaneous nerves (7, 33, 38, 108, 123–130). NGF stimulates melanocyte dendrite formation and prolongs melanocyte survival after UV damage (123, 124). NGF and other neurotrophins can regulate keratinocyte proliferation and differentiation (123, 125, 128 – 130), functions that in the mouse appear to be coordinated with the hair cycle (128). Lastly, NGF and other neurotrophins can act as mast cells secretagogs and can modulate dermal fibroblasts and dermal immune cells function (7, 33, 38, 108, 123–130). NGF and neurotrophins may have a physiological role in hair cycle and hair follicle morphogenesis (128 –130, 132), whereas NGF may protect human keratinocytes from UVB-induced apoptosis (125).

K. PTH and PTH-related protein (PTHrP) receptors

Dermal fibroblasts express class 1 PTH/PTHrP receptors, which respond to PTH or the PTHrP signal by increasing cAMP, production of cytokines and keratinocyte growth factor (KGF) (135, 136); however, the same receptor is not detected in keratinocytes (135, 137–139). Nevertheless, PTHrP has direct epidermal biological activity stimulating keratinocyte proliferation and differentiation both in vitro and in vivo, and hair follicle formation (15, 140, 141). The keratinocyte intracellular activation pathway differs from that stimulated by class I receptors; PTHrP, while producing intracellular calcium accumulation and protein kinase C stimulation, does not stimulate adenylate cyclase; instead it activates the phospholipase C pathway (138, 139, 142, 143). These PTH/PTHrP class II keratinocyte receptors have been partially characterized and found to also respond to PTH (138, 139, 143). PTHrP can indirectly regulate functional activity of the epidermis through the stimulation of KGF production by dermal fibroblasts (136). Furthermore, PTHrP can play a role during wound healing, helping restore epidermal homeostasis (144). Studies on the PTHrP knockout mouse model, and in mice overexpressing the peptide in the skin, document an important role of PTHrP on epidermal function and hair formation (15, 141). L. Vitamin D receptor (VDR)

Receptors for the active form of vitamin D [1,25-dihydroxyvitamin D (1,25-(OH)2D or calcitriol] are expressed in human and rodent epidermal and follicular keratinocytes (13, 145–151). In the mouse, hair cycle-dependent VDR expression has been reported: it is stronger in mid and late anagen and in catagen and weaker in the telogen and early anagen phases of hair growth (146). VDRs serve as targets mediating calcitriol induction of keratinocyte differentiation and inhibition of cell proliferation (13, 148). Because of these properties, vitamin D derivatives are being used therapeutically (topically) in psoriasis (3, 13, 14). The presence of alopecia in some forms of vitamin D-resistant rickets with decreased expression of VDR in dermal papilla cells indicates a role in hair growth (13, 150). Some authors have identified VDRs in human melanocytes and observed a modulatory effect of 1,25-(OH) 2D on melanogenesis (8, 151). This observation has not been confirmed universally (152). Skin

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

immune cells may also express VDRs because of the finding of constitutive immunosuppressive activity for 1,25-(OH) 2D (153). Patients with mutations of VDR present with hypocalcemia, rickets, and significant cutaneous involvement expressed as sparse body hair and, sometimes, total alopecia that includes the eyebrows and eyelashes (150). In the latter subjects, VDR gene mutations result in premature stop signals or abnormal DNA binding and marked resistance to calcitriol therapy (150). Experiments performed on mice treated with topical calcitriol show normal hair regrowth after chemotherapy-induced alopecia (147). M. Glucocorticoid and mineralocorticoid receptors

Glucocorticoid receptors (GRs) are members of the superfamily of trans-acting transcriptional factors and are widely expressed in all skin compartments (3, 4, 12, 31, 154 –157). More specifically, GRs are expressed in epidermal and follicular keratinocytes, epithelial cells of eccrine and apocrine glands, sebocytes, melanocytes, immune cells of epidermis and dermis, dermal fibroblasts, and smooth muscle (154 – 157); activation of these receptors regulates or modulates specific functions in the corresponding cells. The role of glucocorticoids is best emphasized by the skin changes associated with hypercortisolism (3, 36, 37). In such states there are alterations in body fat distribution, general atrophy of the skin, impairment of wound healing, easy bruisability, mild acanthosis nigricans, acne, hirsutism, and alopecia. A glucocorticoid direct inhibitory effect on hair growth has been well documented in animal models (31). It must also be emphasized that glucocorticoids, whether administered topically or orally, are potent drugs used in the treatment of inflammatory skin diseases (3). Most recently, mineralocorticoid receptors (MRs) have been detected in keratinocytes of the epidermis and hair follicle and in sweat and sebaceous glands of human skin (158). The same cutaneous structures also expressed 11␤hydroxysteroid dehydrogenase (11HSD), which converts glucocorticoids to their inactive metabolites, thereby allowing the binding of aldosterone to MRs at much lower prevailing levels (158, 159). N. Androgen and estrogen receptors

Sex steroid receptors belong to the superfamily of transacting transcriptional factors, similar to glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) (36, 68). They are widely distributed in all skin compartments, and their density and expression level vary depending on anatomic site and gender (3, 4, 12, 160 –173). The well recognized androgen effects on hair growth and sebaceous gland functions are related to expression of the corresponding androgen receptors (ARs) in epithelial cells of those adnexal structures and in specialized dermal papilla fibroblasts that regulate hair morphogenesis (161–165). ARs are also expressed in other adnexal structures, in epidermal keratinocytes and melanocytes, dermal fibroblasts, and resident and circulating cells of the skin immune system (3, 4, 12, 31, 160 –165, 169). Similar to ARs, estrogen receptors (ERs) are also expressed in the epidermal, adnexal, and dermal compartments of the

463

skin (3, 4, 12, 166 –168, 170 –173). ERs have been detected variably, depending on the sensitivity method and presence or absence of pathology, in epithelial cells of epidermis, hair follicle, sebaceous, eccrine and apocrine glands, in melanocytes, and in dermal fibroblasts. Thus, both estrogens and androgens regulate hair growth, sebaceous gland function, proliferation and differentiation of epithelial cells of the epidermis and adnexa, functional activity of dermal fibroblasts and fibrocytes, wound healing, and skin immune cells activity. There are also data showing that androgens and estrogens can modulate proliferation and melanogenesis in cultured melanocytes (169 –171). Lastly, transgenic male mice overexpressing GH show overgrowth of the skin that is androgen dependent, e.g., it is not observed in females or in castrated males (97). Clinical signs of androgen excess include acne, hirsutism, and androgenic alopecia (3, 36, 37, 163, 164). Acne results from follicular hyperkeratinization, increased sebum production, and from the release of lipases and proinflammatory mediators by Propionicum acnes. In these conditions, androgens [mainly dihydrotestosterone (DHT) and to a lesser degree testosterone] mediate the increased sebum production and follicular hyperkeratinization (3, 37, 163, 164). Hirsutism and androgenic alopecia are associated with increased production of DHT within the dermal papilla of androgen-responsive hair follicles of the face, chest, genital skin, and scalp (3, 12, 31, 37, 163). Conversely, in males with androgen deficiency, the skin remains thin and fine; sebaceous and apocrine glands and sexual hair follicles remain dormant; beard, axillary, and pubic hair do not develop and neither does androgenic alopecia; and there is also a general decrease in skin pigmentation (3, 37, 163). Increased estrogen levels, for example during pregnancy, can lead to hyperpigmentation of nipples, areolae, genital skin, and facial skin (3). The latter, known as melasma, is exacerbated by sun exposure (3). In addition, preexisting nevi and ephelides darken, and telangiectasia, spider angioma, and palmar erythema may develop (3, 37). The presence of actual ERs in malignant melanocytes has been questioned (174). However, studies with normal cultured melanocytes have demonstrated both the presence of receptors and phenotypic effects on cell proliferation (171). Nevertheless, the reports are truly conflicting as regards the estrogen effect on melanogenesis. Thus, while some have reported stimulation of tyrosinase activity and melanin synthesis by estrogens (170), others have shown an opposite inhibitory effect (171). These contradictory results indicate the need for additional work on the role of estrogens in melanocyte functions. O. Thyroid hormone receptors

The skin is a recognized target for T3 (3, 4, 12, 31, 36, 37, 157, 164, 175, 176). This hormone is involved in the process of epidermal differentiation and increases its responsiveness to growth factors. It also participates in the function of sebaceous, eccrine, and apocrine glands, in hair growth, and in the production of proteo- and glycosaminoglycans by dermal fibroblasts. All of these effects are probably mediated by interactions with the specific thyroid hormone receptors

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

464

SLOMINSKI AND WORTSMAN

(TRs) that serve as transcriptional regulators. In fact, thyroid hormone receptors (c-erb-A) were detected by RT-PCR in human skin (177), and c-erbA␤ and c-erbA␣ mRNAs were detected in dermal fibroblasts, consistent with T3 binding to fibroblast nuclear extracts (178). Since T3 may have an inhibitory effect on melanogenesis in malignant melanocytes, it is likely that TR is also expressed in melanocytes (179). A potential role for thyroid hormones in the regulation of skin function is suggested by its changes in hyper- and hypothyroidism (3, 36, 37). In the former, the skin changes include erythema, palmoplantar hyperhidrosis, acropathy, and infiltrative dermopathy. Graves’ disease also may be associated with generalized pruritus, chronic urticaria, alopecia areata, vitiligo, and diffuse skin pigmentation. In hypothyroidism, the skin is cool, dry with pasty appearance; the epidermis is thin and hyperkeratotic; alopecia may develop, and there is diffuse myxedema. In contrast to the pretibial myxedema present in hyperthyroidism, the generalized myxedema of hypothyroidism is reversible with thyroid hormone therapy (37). P. Cholinergic receptors

Grando and associates (20, 180 –182) found that human keratinocytes express both nicotinic and muscarinic receptors in a differentiation-dependent manner. Specifically, human keratinocytes were found to express the ␣3, ␣5, ␣6, ␣7, ␤1, ␤2, and ␤4 nicotinic receptor subunits (20, 180, 181). Immunocytochemistry studies further showed that the number and subunit composition varies according to stage of epidermal keratinocyte differentiation (20, 181). The nicotinic receptors on keratinocytes represent functional ion channels mediating the influx of Na⫹ and Ca⫹2, and the efflux of K⫹, being thus essential for keratinocyte viability (20, 180, 181). Activation of nicotinic receptors stimulates keratinocyte motility and differentiation (20, 180, 181). Muscarinic receptors of several subtypes have been detected in vitro and in vivo in epidermal keratinocytes (20, 181, 182). The subtypes expressed include the m1, m3, m4, and m5 types, with both timing and level of expression being dependent on keratinocyte differentiation stage (20, 181, 182). Muscarinic receptors have been characterized also in malignant human melanocytes (183, 184), and there is strong evidence for their expression in normal melanocytes (Grando et al., unpublished observation). Q. Adrenergic receptors

Radioligand binding studies have shown cutaneous adrenergic receptors, with epidermal keratinocytes and eccrine epithelial cells expressing predominantly the ␤2 adrenoreceptors (4, 21, 185–187). In situ binding assays have further identified ␣1- adrenoreceptors in the epidermis (188). Stimulation of ␤-adrenergic receptors in epidermal keratinocytes results in increased cAMP production, calcium influx, and stimulation of keratinocyte differentiation (3, 4, 21, 187). ␤-Receptors are expressed in inflammatory cells of the dermis (185), thus explaining the ␤2-adrenoreceptor agonists inhibition of proinflammatory TNF␣ release (189). ␣- And ␤-adrenoreceptors are expressed in dermal blood vessels,

Vol. 21, No. 5

and their activation induces vasoconstriction and decreases vascular permeability (4, 190, 191). Since the melanoma cell phenotype can be modified by adrenergic agonists, it is possible that normal mammalian melanocytes may express adrenergic receptors, similar to pigment cells of other vertebrates (8, 66, 192). The ␤1, ␤2, and ␤3 adrenoceptors are also present on adipocytes (193). R. Glutamate receptors

Immunocytochemical studies performed on rat skin demonstrated the presence of the G protein-coupled metabotropic receptors of the ionotropic glutamate-gated ion channels such as the N-methyl-d-aspartate (NMDA) and ␣-amino-3-hydroxy5-methyl-4-isoxazole propionate (AMPA) types of glutamate receptors in basal epidermal keratinocytes (194). In addition to receptor expression, the specific glutamate transporters have been also detected. Thus, EAAC1 was found in basal keratinocytes, GLT-1 in suprabasal keratinocytes, and the AMPA-type receptor clustering protein, GRIP, in basal keratinocytes (194). In the same rat model, epidermal expression levels of the NMAD receptor and the EAAC1 glutamate transporter were significantly related to wound healing and embryogenesis. Cultured human keratinocytes have shown expression of mRNAs for the NMDAR1 subunit and for GRIP (194). S. Serotonin receptors

The potential presence of serotonin receptors in the skin is suggested by the local effects of serotonin, e.g., pro-edema, vasodilatory, proinflammatory, and pruritogenic (3, 4, 34, 115, 195, 196). In mouse skin, serotonin-induced vascular permeability is mediated by the activation of 5-hydroxytryptamine type 1 (HT1) and HT2 receptors (197). The pruritogenic effect of serotonin may be mediated through direct activation of HT3 receptors or indirectly through mast cells (7, 27, 34, 198). Cutaneous expression of 5-HT2A receptors was detected in unmyelinated axons at the dermal-epidermal junction and in the nerve endings of Pacinian corpuscles (199). Because of serotonin proinflammatory activity, it is likely that cells of the skin immune system will also express the HT receptors generally found in the immune cells (7, 27). Such mechanism would explain the initiation of T celldependent contact sensitivity by serotonin released from human platelets (200), and also the release of prostaglandin E2 from rat skin in vitro (201). Since serotonin can stimulate epidermal keratinocyte proliferation in organ culture, these cells may also express HT receptors (202). T. Histamine receptors

After its release from mast cells, basophils, and platelets, histamine has pleiotropic phenotypic effects in the skin through interactions with H1, H2, and H3 receptors (3, 7, 115, 203). Histamine’s most prominent cutaneous effects are on the local vascular and immune systems, supporting the use of antihistamine drugs for the treatment of pruritus, urticaria, and angioedema (3, 7, 115, 195). Histamine receptors are expressed in the dermal compartment on immunocytes, endothelial cells, blood vessels, smooth muscle, fibroblasts, and nerve endings (3, 7, 115, 195, 203), whereas in the epi-

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

dermis, H1 and H2 receptors are expressed on keratinocytes (204 –207). Epidermal melanocytes express H2 receptors (208). Activation of keratinocyte H2 receptors affect proliferation and differentiation via activation of the adenylate cyclase/phospholipase C pathway with associated increases in intracellular calcium levels (204, 205). In the same cell system, activation of the H1 receptor enhances UVB-induced IL-6 production (206), whereas H1 receptor antagonists inhibit ICAM-1 expression (207). Activation of the H2 receptors on melanocytes stimulates melanogenesis (208). Thus, both the dermal and epidermal compartments are clear targets for histamine, regulating cellular functions not directly connected with the previously described proinflammatory effects of this mediator. Of great interest is the proposed role for the mast cell as a coordinator of immune, neural, and endocrine activity on the central level and peripheral organs (115, 203); in this context the cutaneous actions of histamine through specific receptors would also be addressed at coordinating the local cutaneous neuroendocrine system responses (203). U. Miscellaneous receptors

A number of studies suggest that epidermal keratinocytes express purinoreceptors that, when activated by adenosine or adenine nucleotides, will stimulate cAMP and IP3 production, respectively (209, 210). Purinoreceptor activation inhibits keratinocyte proliferation (211). Functionally active adrenomedullin receptors (AM-R), which are G-protein linked and coupled to adenylyl cyclase activity, have been identified in epithelial cells of epidermis, hair follicle, sebaceous and eccrine glands, and in melanoma cells (212). AM binding sites have been characterized in cultured keratinocytes and in melanoma cells; in the latter system AM stimulates DNA synthesis (212). Calcium sensing receptors identical to those found in the parathyroid glands have been identified in cultured normal human keratinocytes (213). There is also experimental data suggesting the existence of receptors for l-tyrosine and l-DOPA (214), since both ltyrosine and l-DOPA can act as regulators of melanogenesis (10, 23), and l-DOPA can suppress lymphocyte activity (215). Finally, the modulatory effect of melatonin on keratinocyte proliferation and inhibition of melanogenesis suggests that melatonin receptors are expressed also in mammalian skin (216 –218). Nevertheless, the evidence is based solely on the detection of melatonin binding sites, while specific receptors for melatonin remain to be characterized. In lower vertebrates, skin melatonin receptors are well characterized, and melatonin is recognized to play an important role in skin pigmentation, acting as a lightening agent (8, 67, 68).

465

IV. Skin as a Source of Hormones and Neurotransmitters

Hormones and neurotransmitters produced by epidermal and adnexal structures and dermal cells are listed in Table 2. Neuropeptides released by cutaneous nerve endings or produced by skin are listed in Table 3. The production of vitamin D is covered separately because it is synthesized in the skin, and its systemic effects have been well characterized. A. PTHrP

PTHrP is a protein made ubiquitously throughout the body and expressed most intensely in embryonal and fetal tissues (15). Its gene is encoded in chromosome 12 and, when expressed and processed in human keratinocytes, yields three or more isoforms (5, 137). Three transcripts that have been best characterized are 139, 141, and 173 amino acids long (137). The N-terminal region of PTHrP shows a high degree of homology with PTH, with which it shares 8 or 9 of the first residues. Likewise, PTHrP can bind to the classic bone and renal receptors for PTH (type I), producing hypercalcemia (219). In the skin, PTHrP is highly expressed in the granular layer of the epidermis and outer root sheath of the hair follicle and at much lower levels in basal keratinocytes and melanocytes (15, 220). Pathologically, PTHrP is frequently expressed in squamous carcinomas and in their corresponding cutaneous form (15, 143). Expression of PTHrP has also been reported in metastatic melanoma (221, 222). In addition, during wound healing PTHrP is produced by granulation tissue cells that include myofibroblasts and infiltrating macrophages (144). It is of interest that PTHrP is the main cause for the syndrome of humoral hypercalcemia of malignancy (HHM) and that it is produced predominantly by squamous cell tumors (223). However, cutaneous squamous cell carcinomas, which have a high incidence of about 39/100,000, may have resulted in only a few documented cases of HHM (223). The reason for this discrepancy is unclear but may be related to lower levels of expression, release of mostly inactive fragments, or of inability of PTHrP molecules to cross the basement membranes and reach the systemic circulation (219). Presumably, such a barrier is broken in some cases of advanced squamous cell carcinoma of the skin that develop hypercalcemia. HHM associated with increased serum levels of PTHrP has also been described in metastatic human melanoma (221); and, in at least one case, clear evidence is provided that melanoma cells themselves have been the source of PTHrP (222). The reported patient had no signs of bone metastases, and PTHrP immunoreactivity was detected in melanoma cells on autopsy specimens but not in a biopsy

TABLE 2. Selected hormones and neurotransmitters produced in the skin Compartment

Hormones and neurotransmitter repertoire

Epidermis Dermis and adnexal structures

a

Vitamin D, PTHrP, androgens, T3, L-DOPA, catecholamines, acetylcholine, serotonin, glutamate, aspartate, CRH, urocortin, ␣-, ␤-, ␥-MSH, ACTH, ␤-endorphin, enkephalins, TRH Vitamin D, PTHrP, estrogens, androgens, L-DOPA, serotonin, glutamate, aspartate, CRH, urocortin, ␣-, ␤-, ␥-MSH, ACTH, ␤-endorphin, enkephalins, GH, histamine catecholamines,a acetylcholinea

In the dermis, catecholamines and acetylcholine originate predominantly from cutaneous nerve endings.

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

466

SLOMINSKI AND WORTSMAN

Vol. 21, No. 5

TABLE 3. Selected neuropeptides generated in the skin Source

Neuropeptide

Resident and circulating skin cells

Gastrin-releasing peptide, somatostatin, NPY, atrial natriuretic peptide, PHM/PHI, galanin, neurokinins, substance P, neurotensin, CGRP, VIP, bradikinin, cholecystokinin, endothelins, CRH, urocortin, ␣-, ␤-, ␥-MSH, ACTH, ␤-endorphin, enkephalins Substance P, neurokinins, neurotensin, CGRP, VIP, somatostatin, NPY, atrial natriuretic peptide, gastrin-releasing peptide PHM/PHI, bradikinin, galanin, cholecystokinin, endothelins, ␣-, ␥-MSH, ␤-endorphin, CRH, urocortin, dynorphin, enkephalins

Nerve endings

specimen of the melanoma obtained before onset of hypercalcemia (222). B. Hypothalamic and pituitary hormones

1. CRH and related urocortin peptide. The skin is one of the organs producing all the peptides hormones that are central components of the HPA, the main mediator of the systemic response to stress (5, 6, 9, 16 –19, 25, 28, 36, 56). Among the hormones involved in this classic neuroendocrine pathway are hypothalamic CRH and, more recently, the related peptide urocortin (18, 19). In the skin, CRH gene expression has been detected in cultured human keratinocytes and melanocytes with actual production of the peptide, as shown by RIA and RP-HPLC; furthermore, the CRH antigen has been localized in situ to epidermis, hair follicle, nerve bundles, and dermal blood vessels (18, 42– 44, 46, 49). In mouse skin, which does not express the CRH gene, high concentrations of CRH have been detected by RIA, and chemical identification of CRH was documented by RP-HPLC analysis (44, 46). Since the CRH immunoreactivity was localized to epidermal and follicular keratinocytes and in nerve bundles (43, 44), it was postulated that CRH is imported to the mouse skin by cutaneous nerve endings (43, 44, 46); alternatively, mouse skin could express a related gene, with high homology to CRH (46). We recently observed expression of the urocortin gene in mouse skin and also detected the actual urocortin peptide (19). Urocortin tissue levels were highest in telogen skin and decreased progressively during hair cycle to the lowest level in late anagen (19). This pattern is opposite to the hair cycleassociated production of CRH as determined in the same model (44). Expression of the urocortin gene has also been documented in whole human skin, human keratinocytes, human melanocytes, and in hamster melanoma cells (19). Similar to CRH, expression of urocortin peptide was detected in situ in the epidermis, hair follicle, sweat glands, melanocytic nevi, smooth muscle, and wall of blood vessels (19). The reported expression of CRH and urocortin in lymphocytes (224, 225) strongly suggests that cells of the skin immune system may also contribute to the cutaneous pool of those peptides. Therefore, the hormone products that initiate HPA activation at the central level are also readily available in the skin. 2. POMC. There is a large body of data documenting expression of POMC gene in whole human and rodent skin and in cultured skin cells that include keratinocytes, melanocytes, dermal fibroblasts, and endothelial cells, Langerhans cells, monocytes/macrophages, T lymphocytes, and leukocytes (5, 16, 17, 64, 83).

a. Human skin. The POMC peptides ACTH, ␣-MSH, and ␤-MSH and ␤-endorphin peptides have been detected by immunocytochemistry in normal and pathological melanocytes, keratinocytes, Langerhans cells, and mononuclear dermal inflammatory cells (5, 17, 226, 227). Studies with RPHPLC and Western blotting in cultured human melanocytes and keratinocytes showed multiple forms of those peptides such as ACTH 1–10, acetyl-ACTH 1–10, ACTH 1–17, ACTH 1–39, desacetyl-␣-MSH, ␣-MSH, and ␤-endorphin (5, 17, 228 –230). Human dermal endothelial cells and fibroblasts did not only produce, but also released, ␣-MSH and ACTH immunoreactivity into the medium (17, 64, 82, 108, 231). The other POMC peptide, ␤-MSH, was detected by immunocytochemistry in human skin in epidermal and follicular keratinocytes, malignant keratinocytes, melanoma cells, and dermal inflammatory mononuclear cells (5). ␥3-MSH has also been detected in keratinocytes, melanoma cells, neutrophils, and in nerve endings (5, 232). b. Rodent skin. ACTH, ␣-MSH, and ␤-MSH antigens were detected in epidermal and follicular keratinocytes of mouse skin (5, 17, 83), and ACTH and ␣-MSH were also detected in nerve bundles and smooth muscle; ␤-endorphin has been identified only in the sebaceous glands (5, 83). Cultured murine and hamster melanoma cells expressed the ACTH, ␣-MSH, ␤-endorphin, and ␥3-MSH antigens (5, 16). As regards the POMC gene expression, the transcription of shorter and longer POMC mRNA forms has been identified in epidermal and dermal cells (5, 17). This pattern was accompanied by translation of a 30-kDa POMC precursor and its subsequent processing to ACTH, ␣-MSH, ␤-MSH, ␤-endorphin, and ␥3-MSH peptides (5, 17). Detection of the processing enzymes PC1 and PC2 convertases in human and rodent skin indicates that processing of POMC in skin is similar to that in the hypothalamus and pituitary (17, 108, 232a). It must be noted that one group has reported only 80% homology between the human cutaneous POMC mRNA and its pituitary counterpart (233); however, subsequent analysis of the reported sequence by others showed contamination with murine pituitary POMC cDNA (234). Therefore, the information above is convincing evidence of POMC peptide production in the skin and its processing to ␣-MSH, ACTH, and ␤-endorphin-related peptides. Definition of local production of other products of POMC processing will require additional research. 3. Other pituitary hormones. Studies with in vitro systems have shown that human dermal fibroblasts express PRL mRNA 150 kb longer than the pituitary form (235). However, the fibroblasts synthesized and secreted PRL peptide, immunologically and electrophoretically identical to pituitary PRL

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

(235). The data are also consistent with earlier observations of PRL production by normal human connective tissue (236), and with detection of PRL immunoreactivity in sweat glands (237, 238). Expression of the human GH gene has also been detected by RT-PCR in cultured dermal fibroblasts (239); human endothelial cells express the PRL gene (240); and human immune cells produce both PRL and GH (25, 241– 243). Our most recent studies indicate restricted expression of GH gene in the dermal compartment, that failed to detect production of PRL mRNA in whole human skin (243a). Therefore, actual GH and PRL production by the main cutaneous cell compartments has yet to be determined. C. Neuropeptides and neurotrophins

1. Enkephalins. Met-enkephalins (Met-E) and leu-enkephalins (Leu-E), which are products of the larger protein precursor proenkephalin A (PEA) (36, 68), are also produced by mammalian skin (88, 244 –249). Met-E immunoreactivity has been detected in normal human skin and is increased in areas affected by psoriasis (88, 244, 245). The corresponding antigen is located in epidermal keratinocytes and in inflammatory infiltrate components such as T lymphocytes, macrophages, and leukocytes. Met-E has been also detected in the keratinocytes of basal, spinous, and granular layers of human and murine epidermis (88, 245, 250). The detection of PEA mRNA in lesional psoriatic skin further supports local production of the Met-E peptide (244). The cellular source expressing PEA could be mesenchymal dermal cells (248, 249) or immune cells including mast cells (24, 25), since regulated PEA mRNA expression and production of final enkephalin peptides has been detected in rodent skin mesenchymal cells (249), and circulating immune cells express the PEA gene (251). The Met-E peptide has been also detected in epidermal Merkel cells and Langerhans cells (246, 247). Upon further review of the data, it appears that cell type and conditions necessary for expression of the PEA gene remain to be determined. Similar research is needed to determine the cellular source of pro-dynorphin-related peptides, the presence of which has been reported in mammalian skin (252). 2. Nonopioid neuropeptides. Mammalian skin expresses a variety of neuropeptides that include tachykinins SP and NKA, CGRP, VIP, NPY, somatostatin (SOM), galanin, atrial natriuretic peptide (ANP), peptide histidine methionine/peptide histidine-isoleucinamide (PHM/PHI), bradykinin, cholecystokinin (CKK), and gastrin-releasing peptide (GRP) (4, 7, 33, 34, 38, 108 –114). In normal human skin, the most abundant of these peptides are SP, CGRP, VIP, and NPY, although detectable but lower levels of NKA, SOM, and ANP are also present. Neuropeptides are synthesized by nerve cells and released predominantly by unmyelinated afferent C fibers characterized as C-polymodal nociceptors (C-PNN) and by small myelinated A␦-fibers (33, 34, 38, 108). To a lesser extent, autonomic efferent nerves also release the neuropeptides (4, 33, 34). In general, nerves penetrating into the epidermis contain SP, NKA, and CGRP, whereas those innervating dermal structures contain SP, CGRP, VIP, and NKA (33). The neuropeptides are synthesized in dorsal root ganglia, where they are processed and sorted in the Golgi network and then

467

migrate, within dense core vesicles through retrograde axonal transport, to nerve endings in the skin (34). The skin concentration of neuropeptides varies by anatomical site, reflecting, probably, regional differences in innervation (4, 33, 253). It is generally accepted that afferent or efferent nerves are the main source for the cutaneous neuropeptides listed above. An additional source of cutaneous neuropeptides is their synthesis and secretion by resident and circulating skin cells, present in inflamed or even normal skin (4, 7, 24, 25, 27, 28, 38, 110, 111, 115, 254, 255). For example, Merkel cells express antigens that are recognized by antibodies against CGRP, SP, NKA, VIP, PHI, NPY, SOM, and galanin (38, 111, 112, 255). Likewise, Langerhans cells express CGRP, SP, GRP, VIP, SOM, and NKA antigens (110, 254). It remains to be tested whether expression of those antigens is connected to actual transcription and translation of the corresponding genes. Several immunocytochemical studies have reported the presence of VIP, SOM, SP, CGRP, NPY, and NKA in dermal and epidermal immune cells from skin affected with psoriasis, urticaria pigmentosa, allergic dermatitis, and, in some cases, uninvolved normal skin (4, 7, 38, 110, 111, 254, 255). NPY has been additionally detected in epidermal and follicular keratinocytes of normal skin and SOM in basal epidermal keratinocytes of atopic dermatitis skin (38, 133, 254). Thus, skin cells definitively can produce neuropeptides; however, the conditions necessary for such production and the precise identity of the producing cells remain to be defined. 3. Neurotrophins. The skin can produce the neurotrophins NGF, NT-3, NT-4, and brain-derived neurotrophic factor (BDNF) (7, 123–125, 128 –130, 256 –258). NGF is synthesized and secreted by keratinocytes, Merkel cells, and dermal fibroblasts and mast cells (7, 123, 128, 256 –258). In human skin, production of NT3 has been detected in dermal fibroblasts (124), whereas in the mouse it is more widely expressed since NT3 is found in epidermal and follicular keratinocytes of developing skin, and in adult animals it is detected in keratinocytes of hair follicle and DP fibroblasts (128 –130). Hair follicle keratinocytes can synthesize NT4 and NGF (128 –130), and dermal Schwann cells can synthesize NGF, NT-3, and NT-4. Locally produced neurotensin can induce mast cell degranulation (203). It may be speculated that the pleomorphism of neurotrophin cutaneous expression could be related to their significance in regeneration, a functional capability vital for the maintenance of homeostasis. D. Neurotransmitters/neurohormones

1. Acetylcholine. Cultured human keratinocytes can synthesize, secrete, and degrade acetylcholine (180, 181, 259). Keratinocyte acetylcholine is synthesized by choline acetyltransferase from acetyl coenzyme A and choline; in turn, acetylcholine is hydrolyzed by acetylcholinesterase to acetate and choline. Activities of both enzymes, choline acetyltransferase and acetylcholinenesterase, have been detected and characterized in homogenates of cultured keratinocytes. Immunolocalization studies have shown that choline acetyltransferase is consistently present in all layers of the human

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

468

SLOMINSKI AND WORTSMAN

epidermis, while acetylcholinesterase is restricted to basal keratinocytes (180, 181, 259). Acetylcholinesterase activity has been also detected in situ in epidermal melanocytes (260). In addition to local synthesis, acetylcholine is also released by cholinergic nerve endings supplying dermal structures (34). 2. Catecholamines. The human epidermis has the capability to synthesize the catecholamines dopamine, norepinephrine, and epinephrine (21, 22, 187, 261–263), which is consistent with previous findings of phenylethanolamine-Nmethyl transferase immunoreactivity in human epidermal keratinocytes (264). The synthetic activity of cutaneous catecholamines resides predominantly in keratinocytes that express biopterin-dependent tyrosine hydroxylase and phenylethanolamine-N-methyl transferase (21, 187, 261, 262). Catecholamine production takes place in human and rodent melanoma cells (265, 266), which suggests that normal melanocytes may also produce catecholamines (267). Catecholamines can be inactivated directly in the epidermis by the enzymes monoamine oxidase (MAO) and by catechol-methyl transferase, the enzyme already characterized in keratinocytes and melanocytes (187, 268, 269). l-Tyrosine, a precursor for both catecholamines and for melanin, is also synthesized in human keratinocytes and melanocytes from l-phenylalanine by phenylalanine hydroxylase (20, 21, 187, 262, 270). Moreover, phenylalanine hydroxylase and tyrosine hydroxylase activities are dependent on the cofactor 6BH4, which is also synthesized and recycled by human keratinocytes and melanocytes (21, 22, 187, 261). Lymphocytes may also represent an additional source of catecholamines (271). It has been proposed that l-tyrosine and its hydroxylation product l-DOPA would have hormone- and neurotransmitterlike roles (23, 214, 272), with melanocytes being the main site of cutaneous l-DOPA production through the tyrosine hydroxylase activity of tyrosinase (8, 273). l-DOPA produced by melanocytes can, in fact, be released into the extracellular environment. As for norepinephrine, an important cutaneous source is its dermal release from adrenergic nerve fibers (4, 33, 34). 3. Other neurohormones. Serotonin may be also synthesized in the mammalian skin, since rodent mast cells can synthesize serotonin, although this property is not shared by human mast cells (7, 27, 115, 195). Serotonin has also been detected in Merkel cells and human melanocytes and melanoma cells (266, 274, 275). In rodent skin serotonin can be transformed into N-acetylserotonin (NAS), and the responsible enzyme arylalkylamine N-acetyltransferase, together with its gene, are correspondingly expressed (196, 276, 277). In hamster skin NAS can be further metabolized to melatonin and 5methoxytryptamine (278). Finally, the neurotransmitters glutamate and aspartate have been detected by immunocytochemistry in epidermal keratinocytes and in dermal and epidermal dendritic immunocytes (279). E. Thyroid hormones

Human skin may be an extrathyroid site of conversion of T4 into the more active T3 (12, 280 –282). This metabolic trans-

Vol. 21, No. 5

formation would occur in the epidermal keratinocytes through the action of type 2 deiodination pathway (280, 281), with efficiency inversely related to the serum T4 (12). The human epidermis is also the site of T3 deiodination to 3,3⬘diiodothyronine (12). More recent data suggest, however, that at least rodent skin expresses only deiodinase type 3, which catalyzes the 5-deiodination of thyroid hormones (283–285). Since the cutaneous expression of three deiodinases in rodent skin changes during embryonal development, this area needs to be reexamined with modern methods, as it applies to human skin (284, 285). F. Sex steroid hormones

The skin can transform the steroids dehydroepiandrosterone (DHEA) and its sulfate (DHEA-S) into active androgens and estrogens (4, 160, 286 –288). Specifically, enzymatic activity corresponding to 3␤-hydroxysteroid dehydrogenase/⌬5–⌬4 isomerase (3␤-HSD) has been localized to the sebaceous glands and, to a lesser degree, in hair follicles, epidermis, and eccrine glands, while 17␤-hydroxysteroid dehydrogenase (17␤-HSD) has been localized to follicular and epidermal keratinocytes (287–291). 3␤-HSD converts DHEA into 4-androstenedione, and 5-androstene-3␤,17␤-diol into testosterone, while 17␤-HSD converts DHEA into 5-androstene-3␤,17␤-diol, 4-androstenedione into testosterone, and androstanedione into DHT (4, 36, 160). Testosterone is also converted into DHT through the action of a 5␣-reductase, detected in dermal and dermal papilla fibroblasts, follicular and epidermal keratinocytes, and sebaceous and apocrine glands (4, 160, 164, 286, 289 –297). There are two isozymic forms of the 5␣-reductase, but the skin expresses predominantly the type I in a highly specific cellular and regional distribution (290 –296). Nevertheless, cutaneous expression of 5␣-reductase type 2 has been also reported, but at much lower levels; this form has been immunodetected in hair follicles of human scalp (295, 296). The skin immune system can also convert DHEA into 5-androstene-3␤,17␤-diol and into 5-androstene-3␤,7␤,17␤-triol. Cutaneous conversion of testosterone into estradiol is mediated by an aromatase expressed in dermal fibroblasts and adipocytes, but not in keratinocytes (4). However, in keratinocytes 17␤-HSD can transform 17␤-estradiol into estrone or estrone into 17␤estradiol (286). G. Other steroid hormones

The presence of 17␤-HSD indicates that skin can dehydrogenate pregnenolone into progesterone, although the reaction does not proceed in cultured keratinocytes (286). The skin expresses genes for cytochromes P450SCC, P450c17, and P450c21 (66). Immunocytochemistry localization of the antigens for cytochrome P450SCC and P450C17 showed the former in epidermal keratinocytes and eccrine glands and the latter in epidermal and follicular keratinocytes, and in sebaceous and eccrine glands (102). These findings, together with the expression of the ACTH and of the MC2-R gene, suggest that the skin could potentially synthesize glucocorticoids (66). In fact, early studies have shown that whole human skin can metabolize progesterone (PROG) (4), while

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

human keratinocytes can transform DOC into 5␣-dihydroDOC (286). We have recently reported that skin-derived malignant melanocytes can indeed metabolize exogenous PROG to DOC, corticosterone, and 18OHDOC, and that it can also metabolize DOC to corticosterone and 18OHDOC (Fig. 1) (298). A more physiological preparation, whole skin from the rat, can transform PROG into DOC and metabolize DOC to corticosterone-like and to 11-dehydrocorticosteronelike molecular species (Fig. 2) (299).

V. Molecular and Structural Basis for the Organizational Integration of Neuroendocrine Elements of the Skin

It is apparent from the massive amount of data summarized in this work that the skin function extends beyond that of static barrier organ. Thus, in addition to separating the external environment from internal homeostasis, the skin also exerts important endocrine and exocrine activities (cf. Fig. 3). The well characterized exocrine function is performed by the adnexal structures that comprise eccrine, apocrine, and sebaceous glands and hair follicles. These are important in strengthening the epidermal barrier, in thermoregulation, and in the defense against microorganisms (3, 4, 7, 11). Formation of the hair shaft together with the secretions of eccrine, apocrine, and sebaceous glands also plays an important role in social communication. Furthermore, at least in furry animals, the skin plays an important role in behavioral regulation, ascribed to the exocrine activity of the specialized and nonspecialized sebaceous glands that produce and secrete odorants and pheromones (35). As regards the newly recognized endocrine function of the skin, this is performed by cells compartmentally arranged into endocrine units (Fig. 4). These units are composed of cells of epithelial, neural crest, mesenchymal, and bone marrow origin that form the epidermal, dermal, and adnexal structures (cf. Figure 3). As listed in Tables 1 and 2, these cutaneous cells and adnexal structures can concomitantly produce hormones and express the corresponding receptors (see Section III), indicating that the predominant mechanisms of interaction within the different cutaneous compartments are auto- and paracrine in nature. For example, the common skin stressor, UV radiation, stimulates epidermal production and secretion of the POMC-derived MSH and ACTH peptides. In turn, these peptides interact with MC receptors on melanocytes, keratinocytes, and Langerhans cells (LC) to modify their functional activity and increase cutaneous melanin pigmentation (5, 9, 17, 64) and also generate local antiinflammatory and immunosuppressive effects (17, 64, 108). Immunosuppression, mediated predominantly by ␣-MSH, is expressed as functional antagonism to IL-1, down-regulation of accessory molecules expression on antigen presenting cells, and stimulation of IL-10 secretion (17, 64, 82, 108). At the dermal level these immunosuppressive effects include modulation of local cytokine production, and inhibition of endothelial cells expression of adhesion molecules necessary for inflammatory cells transmigration through the capillary network (7, 17, 64, 82, 108). Dermally produced ␣-MSH and ACTH modify hair pigmentation and sebaceous gland func-

469

tion, whereas ACTH modifies hair growth (5, 16, 35, 61, 83, 86). Cutaneously produced CRH and urocortin affect epidermal proliferation of keratinocytes and melanocytes (18, 19, 46, 51), and in the dermis these peptides can modify local immune responses and act as vasodilators and mast cell segregators (40, 48, 56 – 60, 203). There is important neural representation in the skin, and one of its components is the epidermal cholinergic system (20). Its neurotransmitter, acetylcholine, is produced by the keratinocytes in which, through interactions with muscarinic and nicotinic receptors expressed predominantly in the basal or suprabasal epidermal layers, keratinocyte proliferation, migration, and differentiation are regulated (20, 181, 259). Acetylcholine availability is determined by the local concentration of acetylcholinesterase, which is highest in the basal layer and decreases gradually along the vertical axis to reach its lowest levels below the stratum corneum (20, 181). The adrenergic system, represented by the catecholamines produced by keratinocytes, interacts with adrenergic receptors expressed on the same cells to regulate their phenotype (21, 187, 300). The local production of the nonessential amino acid l-tyrosine from l-phenylalanine by melanocytes and keratinocytes ensures its availability for catecholamine synthesis and melanogenesis in the epidermal unit, independent of systemic supply (21, 22, 261). Both l-DOPA, a product of local l-tyrosine hydroxylation, and catecholamines can potentially modify cutaneous immune responses (215, 271). As regards sex hormones actions, testosterone by itself, or after conversion to DHT in keratinocytes and dermal fibroblasts modifies hair growth and sebaceous glands function (4, 163, 164). Dermally produced estradiol (4) can affect function of adnexal structures and the wound healing process (172, 173). As discussed in the section on the vitamin D receptor, the active vitamin D3 metabolite 1,25-(OH)2D3 inhibits keratinocytes proliferation and stimulates their differentiation via interaction with the VDR (13, 14). Evidence for the epidermal conversion of vitamin D into 1,25-(OH) 2D3 (301) would further support local (auto- and paracrine) mechanisms of action. PTHrP produced in the epidermis and hair follicle has a similar effect on keratinocytes through interaction with a receptor different from the classic class 1 PTH/PTHrP receptor (15, 302). Both vitamin 1,25-(OH) 2D3 and PTHrP can affect hair growth (13–15, 302). Notwithstanding their overwhelming local action, hormones produced in the skin can also enter superficial and deep vascular dermal plexuses, or vessels supplying adnexal structures, with long distance effects. Such a true endocrine role is most apparent in the case of hormones and cytokines produced in the dermis that have rather free access, by diffusion to local capillary vessels. Limiting factors for this diffusion are the distance between production site and vasculature, adhesiveness to the extracellular matrix, and local rate of degradation. In contrast, epidermally produced hormones and cytokines must penetrate the basement membrane (BM) before traversing the extracellular matrix (EM) of the papillary dermis; therefore, the permeability barrier and the adhesiveness to the BM and EM limit their access to the most superficial capillary vessels. An example of the relative restriction posed by the dermal-epidermal junction is the

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

470

SLOMINSKI AND WORTSMAN

Vol. 21, No. 5

FIG. 1. Metabolism of progesterone to DOC, corticosterone, and 18OHDOC in malignant melanocytes. A, TLC separation of 14C-progesterone metabolites produced by melanoma cells. B, RP-HPLC identification of 3H-18OHDOC as a metabolite of 3H-DOC. Upper panel, UV detection of 18OHDOC standard; lower panel, radioactivity in eluted fractions with arrow indicating 3H-18OHDOC peak. C, Enzyme-linked immunosorbent assay (ELISA) identification of corticosterone (B) in RP-HPLC-separated fractions. Upper panel, UV detection of corticosterone (B) standard; lower panel, immunoreactive corticosterone (arrow) in media from human melanoma cells incubated with progesterone. Control represents media from cells cultured without progesterone added. DOC, 11-Deoxycorticosterone; B, corticosterone; Aldo, aldosterone; 11dehydro-B, 11-dehydrocorticosterone; 18ODOC, 18-hydroxycorticosterone. [Reproduced with permission from A. Slominski et al.: FEBS Lett 445:364 –366, 1999 (298). Experimental conditions are detailed in Ref. 298.]

rarity of patients showing systemic effect (hypercalcemia) from cutaneous PTHrP, a hormone produced abundantly in the epidermis (15, 302). Still, epidermally produced vitamin D (14), urocanic acid (303, 304), and, perhaps, PTHrP do enter the systemic circulation and are able to modify the functional

activity of distant organs, providing evidence for endocrine effects by factors produced in the epidermis. Urocanic acid produced in the stratum corneum of the epidermis can also enter the systemic circulation and have immunosuppressive effects (4, 123, 303, 304). As mentioned above, epidermal

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

471

FIG. 2. Active steroidogenesis in normal rat skin. Both panels show TLC separation of steroid products. The right lanes marked 14C-PROG (A) or 14C-DOC (B) show radioactive steroid intermediates generated in rat skin. The left lanes marked controls represent nonenzymatic transformation of 14C-PROG (A) or 14C-DOC (B) incubated in culture media only. Experimental conditions are detailed in Ref. 299. PROG, Progesterone; DOC, 11-deoxycorticosterone; B, corticosterone; Aldo, aldosterone; A, 11-dehydrocorticosterone; 18OHB, 18-hydroxycorticosterone. [Reproduced with permission from: A. Slominski et al.: Biochim Biophys Acta 1474:1– 4, 2000 (299). © Elsevier Science.]

PTHrP may play predominantly para- or autocrine roles (15, 141). It could, however, have a distant effect after release by pathological conditions (skin cancer) (222, 223). Cutaneous neuroendocrine elements are therefore tightly organized and arranged into epidermal and dermal endocrine units, as determined by the physical separation between those compartments (Figs. 4 and 5). These units, which become fully expressed in a field-restricted stress-dependent manner, have broad bidirectional communications. This is accomplished through soluble factors able to penetrate the basement membrane and, also, through sensory nerve endings connecting epidermis and dermal structures (Figs. 4 and 5). Sensory nerve fibers provide anterograde or retrograde transmission of impulses through axon reflexes with release of neuropeptides at epidermal or dermal nerve endings (33, 34, 38). The latter intracutaneous communication mechanism

represents therefore a high-speed and extremely specific connection for the transfer of information, sensed externally or internally, to specific target cells. Target cell activation may then be mediated by neuropeptides (cf. Table 3 and Section IV.C) synthesized and released predominantly by the unmyelinated C fibers described as C-polymodal nociceptors (C-PNN), or by myelinated A␦-fibers (33, 34, 38). There are specific roles for each of the different neuropeptides released by afferent nerve endings in the functional regulation of epidermal barrier properties, skin immune activity, vascular activity, hair growth, and adnexal functions. Those topics, as well as their mechanism of action, have been discussed in comprehensive reviews on the subject (5, 7, 33, 34, 38, 56, 110 –112, 128, 203). It is apparent from neuroimmunocytochemistry studies that nerve subpopulations containing different neuropeptides, such as SP, NKA, CGRP, VIP, SOM,

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

472

SLOMINSKI AND WORTSMAN

Vol. 21, No. 5

FIG. 3. Localization of the cellular components of the exocrine and endocrine skin units. The epidermis is represented by the upper layer, separated from the dermis by the basement membrane. The listed cell types have the capability to produce hormones, neural mediators, and cytokines; they also express the corresponding receptors indicating auto- and paracrine mechanisms of action.

NPY, PHM, enkephalins, CRH, and ␥- and ␣-MSH, do enter cutaneous structures. The presence of this neural network with fibers penetrating all the vital layers of the epidermis and branching into dermal structures, adnexa, Merkel cells, epidermal Langerhans cells, melanocytes, and dermal mast cells provides, therefore, the support for a dual role for those cells, as effectors and regulators (33, 34, 38, 110, 114, 115, 118, 119, 128, 203, 300, 305–313). Within this local branching neural network, disturbances of local homeostasis expressed as production of chemical mediators can be sensed in a specific manner and transmitted to local subunits to counteract noxious stimuli or protect against further damage. The neural branches can be also activated directly by neurohormones and bioactive peptides produced locally, such as histamine, eicosanoids, or NO (33, 34, 38, 112, 203, 312, 314); or, by physicochemical agents such as changes in pH, cation, and free radical concentration (cf. Ref. 34). Indirect neural modulation may be provided by the cytokine networks that modify the chemical environment surrounding the nerve endings. As compared with this neural mechanism, humoral communication, dependent on local diffusion, results in a much slower response. The special role of autonomic nerves in the cutaneous neuroendocrine organization is limited by the predominant dermal ending of their fibers. Thus, they can only regulate function in the dermal endocrine unit although neurotransmitters released in the proximity of the basement membrane could potentially diffuse into the epidermal compartment. It is also possible that autonomic nerves could enter the epi-

dermis; however, this hypothesis still requires experimental investigation. The bidirectional interaction between skin elements and local neural network is perhaps best illustrated by the changes in murine skin during the hair cycle, which are dependent on appropriate dermal and adnexal innervation (128, 300, 311). In this process, the hair cycle-associated tissue remodeling is accompanied by a tightly regulated sprouting and regression of specific afferent and efferent nerve fibers that form a neural and neurotransmitter network. Its cutaneous expression is highly specific and tightly determined by the actual phase of the hair cycle (128, 300, 311). The epidermal and dermal endocrine units with their bidirectional communication pathways, which proceed via soluble mediators or via antidromic axon reflexes through the nerve branches that link both compartments, combine to form the skin neuroendocrine organization (Fig. 5). In general, this neuroendocrine organization functions to coordinate the epidermal and dermal changes necessary for reinforcing the physical barrier and maintaining its structural integrity. To implement these objectives, it modulates sensory reception, melanin pigment production and distribution, activity of the local immune system, vascular functions, thermoregulation, exocrine secretion, and metabolic transformation of prohormones or hormones into other molecules of different biological activity. The skin neuroendocrine system is thus continuously sensing environmental components and, when activation threshold levels are reached, a reaction is triggered with

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

FIG. 4. Diagram demonstrating the proposed flow of information between dermal and epidermal endocrine units and their systemic connections. For functional purposes, cells with endocrine capability become activated in response to environmental stimuli in a stressdependent manner, organized according to anatomic compartment of residence. They communicate by humoral signals (central arrows) and sensory nerve endings (arrows on the left). The efferent neural regulation is provided by the autonomic nerves (right), and also by sensory nerves (left) via antidromic conduction.

production of specific biological factors. Some of these factors may be released to the extracellular compartment to activate sensory nerve endings, directly enter the circulation, or activate circulating immune cells. The sum of these actions sets the optimal mode for dealing with deleterious environmental changes (Fig. 6). Humoral signals that could directly enter the circulation include cytokines and hormones and vitamin D3. The latter represents a marker for a cutaneous action in response to an environmental component (UV-B), which results in well defined systemic effects on calcium homeostasis (14). An analogous example in amphibians is the skin regulation of pituitary function through TRH and skin peptide tyrosine-tyrosine (SPPY) (315, 316). Environmental factor(s) determine concentrations of TRH and SPPY in frog skin, which are higher than in any neuroendocrine organ; skin TRH reaches the pituitary to stimulate production and release of PRL and ␣-MSH, while SPPY inhibits production of ␣-MSH (315, 316). In mammals, there are other local hormonal factors that could potentially enter systemic circulation after UV radiation exposure or in pathological conditions. Among those are POMC-derived ␣-MSH and ␤endorphin (5, 9, 108, 230), met-enkephalin (245), PTHrP (15, 302), or DHT, estradiol, and T3 (4, 11, 12). Skin cytokines can also directly affect the functional activity of distant immune and nonimmune organs (7, 11, 17, 27, 108), whereas circulating cytokines are known to affect hypothalamo-pituitary axis function (24 –28). Thus, IL-1, IL-6, interferons, and TNF␣ can access brain and pituitary to up-regulate production and release of selected hypothalamic and pituitary hormones.

473

FIG. 5. Organization and function of the skin neuroendocrine system. The epidermal and dermal endocrine units modulate functional activity of epidermal and dermal compartments. Endocrine units coordinate their actions by humoral signals (central arrows) and/or by the local neural network (connecting arrow on the left).

Cytokines may have similar action on the regulation of adrenal gland function (28, 317). Intermediates of melanogenesis l-DOPA and products of its metabolism in the melanocytes might also have systemic effects when, under pathological conditions, they are released into the circulation (10, 23, 318). Many epidermally or dermally derived molecules are incapable of migrating because of insignificant concentration in the vicinity of blood vessels; however, a systemic effect is still theoretically possible. Thus, direct stimulation of dermal, adnexal, or subcutaneous cellular components could secondarily result in the production of biological mediators with definite systemic effects. An example of this amplification mechanism is represented by the cytokines IL-1 and TNF␣, which can stimulate leptin production by adipose tissue in the deep dermis and/or subcutis (319, 320). In this manner, small amounts of cutaneous cytokines could affect feeding behavior and energy balance (319). Another example is the activation of skin immune cells that can enter the circulation and have distant immunological or regulatory effect (7, 27). Similar to the humoral model of communication in the cutaneous endocrine system, with potential cytokine-mediated stimulation of the HPA axis, the cutaneous neural signaling system could also activate central nervous system pathways. The latter connections have the advantages of being more rapid with higher specificity (Fig. 6). Thus, changes in the skin physicochemical environment generated by physical, chemical, or biological trauma, UV radiation, or local disease processes could be sensed by afferent nerve ending, and thence transmitted via the spinal cord to the brain. However, before the information is sent to the brain,

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

474

SLOMINSKI AND WORTSMAN

Vol. 21, No. 5

the spinal cord to other organs without ever reaching the higher centers. To summarize, the skin can generate rapid (neural) or slow (humoral) moving signals to induce responses at the general or systemic level or at the organspecific level. These responses are designed to counteract the damaging effect of environmental insults or to adjust the homeostatic system to the optimal mode that would buffer environmental noxious agents most efficiently. VI. Regulation of Cutaneous Neuroendocrine System

There are number of environmental and intrinsic factors that regulate the cutaneous neuroendocrine system activity level. The most prominent environmental factor affecting the skin is solar radiation, particularly within the UVA and UVB wavelengths. Temperature, humidity, and concentrations of chemical and biological agents represent other factors. Some internal factors affecting the skin neuroendocrine system are changes in the physicochemical microenvironment or in biological modifiers; these may be generated in reaction to environmental signals or result from local cyclic biological rhythm associated with hair cycling, or from local or general disease processes. Of the endocrine factors produced by the skin the most important is vitamin D, which is not only a regulator of the calcium metabolism but has other systemic effects (13, 14, 145). For example, epidemiological evidence suggests that sunlight deprivation with associated reduction in the circulating levels of vitamin D3 derivatives may result in increased incidence of the carcinomas of breast, colon, and prostate (145). A. Solar radiation FIG. 6. Systemic effects of skin neuroendocrine system products. In response to noxious stimuli, the skin endocrine system mounts a progressive, intensity-dependent, highly coordinated response. In the case of high-magnitude stimuli, the generated signals travel through humoral or neural pathways to reach the central nervous system, immune system, and other organs.

it may be modulated by the local cutaneous neuroendocrine units through the direct activation of nerve receptors by neurohormones and neurotransmitters, or by neuropeptides (cf. Tables 2 and 3), histamine, eicosanoids, NO, and other proinflammatory mediators (5, 7, 27, 33, 34, 108, 113, 203, 314). Alternatively, stress released cytokines and proinflammatory biological modifiers could affect signal type and neural sensor availability through indirect mechanisms, e.g., activation of other cells to produce and release factors activating afferent receptors. In this context, mast cells, melanocytes, Langerhans cells, and Merkel cells could be particularly important because of their close contact with nerve endings (33, 34, 110, 111, 119, 203, 305–308). Secondary changes in hydrogen ions, cations, free radical and NO concentrations or eicosanoids produced by cytokine-activated keratinocytes or immune cells could have similar effects on sensory nerve endings. In the visceral organs the cytokine IL-1, IL-6, and TNF␣ signals can be potentially transmitted through an indirect mechanism via the vagus nerve to the central nervous system (26, 28, 321). Lastly, upon leaving the skin, some afferent neural signals may also be relayed from

UV light is a form of electromagnetic energy that includes the wavelength between 100 to 400 nm of the solar spectrum. Although it includes vacuum UV, UVC, UVB, and UVA, only the 290 – 400 nm wavelengths that comprise UVA and UVB reach the surface of the earth, because of the partial absorption by atmosphere. UVB (290 –320 nm) interacts very efficiently with the skin, inducing sunburn and pigmentation (3, 4, 9). UVA (320 – 400 nm) has better penetration through the atmosphere but lower efficiency in inducing erythema and melanogenesis. It is classified as UVA1 (320 –340 nm) and UVA2 (340 – 400 nm), and it has been proposed that the photobiological mechanism of action for UVA1 is similar to that of UVB; the effects of UVA2 would involve distinctive oxygen-dependent photochemistry. The cutaneous effects of UV radiation are dependent on the penetration and absorption of the particular wavelength. In human skin UVB is absorbed predominantly by stratum corneum and to a lesser degree by the epidermis. The very small fraction of UVB that reaches the dermis, however, has significant biological effects inducing immediate and delayed erythema (3, 4, 9). Transmission of UVA through epidermis of white skin is high, resulting in approximately 50% of energy reaching the dermis. UVA has only 1/1,000 of UVB biological activity, but it also contributes to the cutaneous actions of solar radiation (3, 4, 9). Thus, it has a major effect in aging of the skin, it has a more limited role in the induction of skin cancer, and it does not produce burning of the skin. In general, the biological

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

responses to solar radiation are dependent on individual susceptibility determined by skin pigmentation, prior exposure to UV radiation (decreases the threshold for subsequent responses), region affected, radiation field size, and environmental conditions (3, 4, 9). There is, nevertheless, a high degree of precision and predictability of the cutaneous response to UV, demonstrating that mechanisms have evolved to transform some of the solar energy into a catalyst activating a recording system (9). Such a recording role could be served by the local neuroendocrine system, whose activation is designed to buffer or counteract the damaging effect of UV. 1. CRH-POMC system. UVB stimulates production of CRH peptide in normal melanocytes without noticeable changes in CRH mRNA levels, suggesting posttranscriptional regulation (322). UVR can stimulate/induce POMC gene expression in the skin in human and rodent normal and malignant keratinocytes and melanocytes maintained in cell culture (5, 9, 17, 64, 108, 230, 323). The stimulation of ACTH, ␣-MSH, ␤-LPH, and ␤-endorphin production and secretion in response to UVB is dose dependent in normal and malignant epidermal cells, and in dermal endothelial cells; POMC mRNA production is correspondingly increased (17, 64, 108, 323). UVA can also stimulate POMC gene expression with subsequent MSH and ACTH production in human keratinocytes and endothelial cells (17, 64, 108). This stimulation of POMC gene expression and production of POMC peptides are observed after a 10-h latency period; production becomes significant at 10 –24 h (17). Of interest, humans and horses exposed to sunlight exhibit increases in the circulating levels of ␣-MSH and ACTH, and experimental whole-body exposure to UVR in humans increases ␤-LPH and ␤-endorphin serum levels (5). In the case of ␤-LPH the response is abrogated by UV-absorbing topical sun blockers, implicating mediation by a photoreaction (324). UVB can also up-regulate expression of MC1-R on normal and malignant cultured melanocytes and keratinocytes (5, 17, 323). This UVB up-regulation of MSH receptors expression is associated in melanocytes, with increased responsiveness to MSH in terms of stimulation of melanogenesis, as shown in both cell culture and in vivo conditions (75, 323, 325). These experimental findings are consistent with the effects of exogenous MSH and ACTH in humans, which cause increased skin pigmentation affecting predominantly the sun-exposed areas. Clinically, the similarly increased skin pigmentation of patients with Addison’s disease is most striking in the sun-exposed areas. These experimental and clinical observations led Pawelek and colleagues (9, 75, 325) to propose that the effects of UVB on cutaneous melanogenesis do not represent random (unrelated) events but, instead, a highly coordinated sequence in which expression of MSH receptors and local production of POMC-derived MSH and ACTH peptides are important intermediate steps. 2. Immunoregulatory molecules. UV radiation influences the immune system at both local and systemic levels with the net effect being immunosuppressive (7, 17, 64, 82, 108, 114, 326, 327). The mechanism for this action can be either direct absorption of light energy by cells of skin immune system that include resident and nonresident (circulating cells) or

475

indirect through UV-induced activation of nonimmune cells in epidermis and dermis with release of cytokines and chemical mediators (7, 9, 17, 64, 82, 108, 114, 326 –333). Important in this regard is trans-urocanic acid (UCA) which, after absorption of UVB, or to lesser degree of UVA energy, isomerizes into cis-UCA in the stratum corneum (303, 304). cis-UCA acts as a potent local and systemic immunomodulator and immunosuppressor (7, 303, 304). Keratinocytes stimulated by UVR can produce and secrete the cytokines IL-1, IL-6, IL-8, IL-10, IL-12, IL-15, TNF␣, and macrophage inhibitory factor (MIF), eicosanoids, basic fibroblast growth factor (bFGF), IGF-I, transforming growth factor (TGF-␣), and endothelins (7, 17, 108, 114, 326 –333). This effect is rapid (within 1–3 h) and predominantly mediated by UVB, although UVA also stimulates IL-10 and, to a lesser degree, IL-6 production. UVR also switches the local cytokines and mediators release profile of nonepithelial components of epidermis and dermis including lymphocytes, macrophages, mast cells, endothelial cells, and melanocytes. The cytokines IL-1, IL-6, TNF␣, and MIF exert local activity, which does not preclude systemic effects upon entering the circulation. 3. Neuropeptides, neurotrophins, and neurotransmitters. UVA, but not UVB, irradiation increases the skin levels of metenkephalin, and multiple whole-body UVA exposure can also increase the plasma level of the peptide (245). For its part, UVB induces release of CGRP, SP, and NKA from cutaneous sensory nerves (38, 108). CGRP appears to have immunosuppressive properties, while SP and NKA enhance cutaneous neuroinflammation (38, 108, 114). UVB also stimulates NO production by keratinocytes, melanocytes, and NO release from sensory nerve endings (9, 108, 314). Also, UVB induces production and release of the neutrophin NGF by epidermal keratinocytes (123, 256). Schallreuter et al. (334, 335) have shown that UVB enhances tetrahydrobiopterin production and phenylalanine hydroxylase activity, with net increase in the epidermal supply of l-tyrosine. l-Tyrosine is a precursor for both catecholamine biosynthesis and melanogenesis. Stimulation of melanogenesis by UVB is associated with increased production of the biologically active products l-DOPA, dihydroxyindole (DHI), and DHI carboxylic acid (9, 123). B. Hair cycle

Hair growth and the cyclic activity of the hair follicle are timed by a “biological clock,” which in rodents changes periodically the physiology and morphology of the entire skin (30, 31). In mice, the expression of POMC gene and production of the POMC peptides ␤-endorphin, ACTH, and ␣-MSH are synchronized with hair follicle cycle (5). POMC production is lowest in telogen (resting phase), increases during anagen (growing phase), and decreases in catagen (involution phase). These changes correlate closely with the local expression of the MC1 gene (65). The intracutaneous concentration of CRH and expression of CRH-R1 exhibit similar changes coupled with the hair cycle, being highest during anagen and lowest during the catagen and telogen phases (44). A similar phenomenon has been described for

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

476

SLOMINSKI AND WORTSMAN

SP, with maximal levels occurring in early anagen and minimal levels occurring in catagen skin (116). Thus, the biological clock regulating the cyclic activity of hair follicles appears to regulate simultaneously the local production or release of neuropeptides and the expression of the corresponding receptors. The hair cycle is associated with striking changes, which are qualitative in distribution and quantitative in expression levels of the neutrophins NT-3 and NGF and their corresponding receptors in the skin of the C57 BL/6 mouse (128 – 132). Furthermore, the pattern of sensory and sympathetic innervation of different cutaneous structures including the hair follicle itself, shows significant hair cycle-dependent changes (128, 300, 311). The changes in adrenergic innervation are accompanied by specific patterns of follicular ␤2adrenergic receptor expression. Therefore, the whole cutaneous neural network involved in the regulation of hair growth undergoes cyclic changes (128, 300, 311), which can potentially affect the function of other cutaneous structures, sensory skin responsiveness, and transmission of afferent signals to the spinal cord. Because of the extent and magnitude of the hair cycle-dependent changes, it is likely that these are regulated within the skin itself, under the control of the “biological clock” governing hair cycle.

Vol. 21, No. 5

context peptide hormones and neuropeptides can be degradated by neutral endopeptidases (NEP) and angiotensin converting enzyme (ACE), which are present in dermal fibroblasts, endothelial cells, and keratinocytes (34, 38, 108, 337). NEP activity is not static, but can be stimulated by proinflammatory cytokines, by factors raising intracellular cAMP, and glucocorticoids (38); in addition, its cutaneous pattern of expression changes during wound healing (337). Other proteolytic enzymes such as mast cells-derived tryptase or chymase can degrade neuropeptides, effectively attenuating their activity (7, 34, 38, 108, 115, 203). Catecholamines and other biogenic amines can be inactivated directly in the epidermis, by the action of MAO and/or by catechol-methyl transferase (21, 268, 269); the latter enzyme has been already characterized in keratinocytes and melanocytes. Acetylcholine is degraded to acetate and choline by epidermal acetylcholinesterase (20, 259). The skin is, in addition, a well recognized site for the transformation of glucocorticoids and sex hormones to molecular forms with higher or lower hormonal activity, or overtly inactive (4, 11, 160, 297). VII. Regulation of Cutaneous Vitamin D Production A. Vitamin D3 production

C. Cytokines

Similar to its effects at the central level, the proinflammatory IL-1 has significant local stimulatory/inductory effects on POMC gene expression and production of POMC peptides in normal and malignant epidermal melanocytes and keratinocytes, dermal endothelial cells, and circulating immune cells that include macrophages (5, 17, 64, 108, 230). Another cytokine, TNF␣, stimulates production of POMC mRNA in normal dermal fibroblasts, while TGF-␤ inhibits it in keratinocytes and normal dermal fibroblasts, but not in keloid fibroblasts (231). TNF␣ also stimulates production of the POMC products ␤-endorphin and ACTH peptides (336). Many cytokines can up-regulate expression of the MC-1 gene and of functional cell surface MSH receptors in normal and malignant melanocytes (5, 17, 64, 323). Those include Il-1␣, IL-1␤, endothelin-1 (ET-1), adult T cell leukemia-derived factor/thioredoxin (ADF/TRX), INF-␣, INF-␤, INF-␥, (Bu)2cAMP, and the hormones ␣-MSH, ␤-MSH, and ACTH. IL-1 can also stimulate MC-1 receptor expression in normal and malignant human keratinocytes and in human dermal microvascular endothelial cells (HDMEC) (17, 64, 230). Conversely, TNF␣ inhibits MC1 expression in melanocytes. Thus, selected cytokines regulate precisely (“fine-tuning”) the level of expression of POMC and MC1-R. The roles of cytokines in the cutaneous regulation of epidermal cholinergic system, production of catecholamines, steroid synthesis and metabolism, and synthesis of neuropeptides CRH, urocortin, and enkephalins remain to be investigated. D. Degradation or inactivation of hormones and neurotransmitters

One important mechanism regulating the availability of locally produced hormones is their degradation in situ. In this

The recognition that the skin could react with an invisible component of sunlight to make a factor indispensable for the mineralization of bone vitamin D represents a remarkable scientific accomplishment (14). Vitamin D3, or cholecalciferol, is formed from the precursor steroid 7-dehydrocholesterol (7-DHC), which is normally concentrated to the plasma membrane of basal epidermal keratinocytes (80% of skin content). Upon stimulation with sunlight photons of UVB light (290 –310 nm wavelength), 7-DHC undergoes photolysis with breakage of the 9,10-carbon bond generating the thermolabile intermediate previtamin D3. At normal skin temperature previtamin D3 molecules convert into vitamin D3 through internal rearrangement (Fig. 7). Vitamin D3 enters the circulation bound with high specificity to vitamin D binding protein (DBP) to exert its systemic actions (338). When newly formed previtamin D3 and vitamin D3 continue to be irradiated, they convert into additional steroids such as lumisterol, pyrocalciferol, and tachysterol, which are practically devoid of vitamin D3 activity (14). Although light and temperature are the only variables involved in the biosynthetic process, they do not account entirely for the rates of biosynthesis observed in tissues. Thus, irradiation of 7-DHC dissolved in isotropic organic solvents such as hexane generates only one tenth of the amount observed in similarly irradiated skin (14). The explanation for this discrepancy was obtained in experiments with artificial liposomes, which uncovered the crucial role played by plasma membrane phospholipids in the kinetics of the reaction. Thus, through amphipatic interactions, previtamin D3 remains stabilized in its “cholesterol like” cZc– conformation, the only one leading to conversion to vitamin D3. Furthermore, the fastest rate of previtamin D3 isomerization was seen in association with phospholipids containing 18 carbon atoms in the hydrocarbon chain (339).

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

477

FIG. 7. Cutaneous synthesis of vitamin D3. Under exposure to UVB, cutaneous 7-dehydrocholesterol molecules undergo photolysis to the secosteroid product cZc-previtamin D3 (this cis-conformer is the only form that can be converted into vitamin D3). Thermal energy produces intramolecular rearrangement of cZc-previtamin D3 with a 1,7-sigmatropic hydrogen transfer from C-19 to C-9, to yield vitamin D3. [Modified from X. Q. Tian and M. F. Holick: J Biol Chem 274:4174 – 4179, 1999 (339), by permission from the Journal of Biological Chemistry. © The American Society for Biochemistry and Molecular Biology.]

Once formed in the skin, vitamin D is translocated to the circulation over a period of 30 h or longer. It reaches peripheral tissues bound with high specificity (98%) to vitamin D binding protein (also known as group-specific component or GC). This is in contrast to orally derived vitamin D2 (ergocholecalciferol), which circulates only 50% bound to vitamin D binding protein, with the remaining fraction being bound to cholesterol containing lipoproteins (14). Vitamin D is metabolized (activated) to 25-hydroxyvitamin D (in the liver) and to 1,25 dihydroxyvitamin D (in the kidney) (14). According to the level of action, the physiological and pathological factors that modify the systemic supply of cutaneous vitamin D3 can be classified into precutaneous, cutaneous, and postcutaneous. B. Precutaneous regulation

1. Environment dependent. This category includes geographic latitude. Thus, as the earth experiences its seasonal tilting during fall and winter, sunlight arrives at an angle at the more polar latitudes crossing the atmosphere almost tangentially during winter, and lessening transmission of UVB wavelengths. For example, during winter in Boston, Massachusetts (latitude 42.2 oN) these wavelengths disappear (339), and a similar situation is observed in the Southern Hemisphere, where vitamin D3 photosynthesis is extremely low during winter in Cape Town, South Africa (latitude 35oS), but almost unchanged throughout the year in Johannesburg (26oS)(340). Another environmental variable is the time of day, since vitamin D3 synthesis is maximal at midday, with only very small amounts being formed before 0800 h or after 1700 h. In fact, in Cape Town, only negligible amounts of previtamin D3 are formed in the winter before 0010 h and after 1500 h. In general, latitude, season of the year, and time of day affect the cutaneous photosynthetic process in a highly coordinated, mutually dependent manner (14). 2. Environment independent. a. Clothing. Garments provide significant protection against the damaging effects of solar light that include erythema, accelerated aging, and development of skin cancer. Experiments on transmission of UVB light through different fabrics showed significant effects on the photosynthesis of vitamin D3 (341). Of fabrics with similar thread density, black wool had the highest light absorption coefficient followed,

respectively, by black polyester, black cotton, white cotton, white wool, and white polyester. All of the fabrics produced significant attenuation of the shorter wavelengths (UVB), and studies in volunteers wearing garments made of the same materials showed complete obliteration of the normal serum vitamin D3 photosynthetic response to one MED (minimal erythema dose) of UVB (341). This absence of vitamin D serum response persisted after whole-body irradiation was increased to the equivalent of 6 MEDs. In addition, regular street clothing, even that worn during summer, also produced significant suppression of the vitamin D3 response to UVB (341). b. Sunscreens. These agents prevent skin penetration of solar radiation. Thus, PABA (para-aminobenzoic acid), the common active ingredient of sunscreens, has an absorption spectrum that overlaps the spectrum responsible for the photosynthesis of vitamin D3 (342). As would be expected, application of PABA to skin pieces blocked the conversion of 7-DHC to previtamin D3 that normally follows exposure to simulated sunlight. Moreover, in healthy volunteers, coverage of the whole body with sunscreens abolished the serum vitamin D3 response to UVB light delivered in a phototherapy unit (342). Also patients with photodependent cutaneous disorders such as skin cancer, who must use sunscreen chronically, have lower serum levels of 25-hydroxyvitamin D as compared with matched controls (343). Nevertheless, it must be noted that these lower 25-hydroxyvitamin D levels have not been associated with secondary hyperparathyroidism or metabolic bone disease (344). C. Cutaneous regulation

1. Regional (anatomical) activity of solar radiation. The segmental body contributions to the supply of vitamin D3 were evaluated in healthy individuals who had sunscreens applied to selected areas of the body before UVB irradiation (345). Significant and almost equivalent serum vitamin D3 increases occurred after selective irradiation of either trunk, legs, or the entire body. UVB exposure of only the head and neck or arms produced a lesser rise in vitamin D3 serum levels, which did not reach statistical significance (345). 2. Race-related skin pigmentation. Melanin is not a “neutral density” light filter, but exhibits varying absorption coefficients, with maximal absorption for the shorter wavelengths

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

478

SLOMINSKI AND WORTSMAN

of the spectrum (⬃ 300 nm) (346). Thus, melanin has a significant effect on the synthesis of vitamin D3. As would be expected, the highest vitamin D3 response to UVB is seen in white individuals, followed by Orientals (East Asians) and Indians (South Asians) and is extremely attenuated in blacks (African Americans) (347). This race effect is also associated with lower prevailing levels of 25-hydroxyvitamin D, although the serum concentrations of 1,25-dihydroxyvitamin D are similar among race groups. The latter results from the intrinsic properties of the renal enzyme l␣-hydroxylase, which can compensate for wide differences in availability of 25-hydroxyvitamin D (348). Asian Indian individuals, who have additional peculiarities in their vitamin D metabolism, are particularly sensitive to the development of clinical vitamin D deficiency, rickets and osteomalacia, when living in areas with low levels of ambient sunlight (349). The UVB light threshold that produced measurable synthesis of vitamin D3 was 18 mjoules/cm2 in a population of white subjects with similar cutaneous photosensitivity (skin type III of the Fitzpatrick-Pathak classification; 1 MED⫽30 mjoules/cm2), although every dose tested was associated with blood levels higher than baseline (350). Serum GC is another factor involved in the availability of vitamin D3 that could be race dependent. Thus, anthropologists have identified a large number of variants, each related to different human populations. By isoelectric focusing, the observed GC suballele frequencies appear to correlate with skin pigmentation, but electrophoretically identical variants were also found in populations widely differing genetically and geographically (351). From the functional point of view, there is no evidence for differences in vitamin D binding among those variants. Moreover, the serum concentration of GC is similar across race groups that include blacks, whites, Orientals, and Asian Indians (347). 3. Suntanning. In the setting of suntanning, vitamin D3 formation is affected in a complex manner. As already mentioned, both previtamin D3 and vitamin D3 are photosensitive substrates that, if irradiated continuously, undergo further conversion to inactive metabolites while still in the skin (14). Nevertheless, measurements performed in tanned white subjects showed elevated vitamin D3 serum levels with correspondingly higher serum 25-hydroxyvitamin D concentration (352). Acute exposure to UVB radiation resulted, however, in attenuated serum vitamin D3 response, presumably the result of acquired cutaneous melanization (352). 4. Aging. Elderly individuals have lower serum levels of 25-hydroxyvitamin D as compared with their younger counterparts. This aging effect is due to progressive decrease in epidermal 7– dehydrocholesterol substrate content (353). As would be expected, acute irradiation with UVB in older subjects results in blunted serum vitamin D3 responses (354). 5. Cutaneous disease. The only cutaneous disorders in which vitamin D3 formation has been systematically studied are the epidermal disease, psoriasis (355), and the connective tissue disease, progressive systemic sclerosis (356). The latter, although predominantly a dermal disorder, is often associated with epidermal atrophy. Acute irradiation experiments in these two groups of patients did show similar vitamin D3

Vol. 21, No. 5

responses in patients and controls. Moreover, the responses were unrelated to the extent of cutaneous involvement. These results probably reflect the small fraction of irradiated body surface required to sustain the normal vitamin D3 requirements. A small fraction of vitamin D3 formation does occur in the dermis (⬍10%), which has a much lower 7-DHC content than the epidermis and is less exposed to the lowpenetrance shorter light wavelengths responsible for vitamin D3 formation. As mentioned above, the dermal disease, progressive systemic sclerosis, even when widespread, does not interfere with the vitamin D3 response to UVB (356). D. Postcutaneous regulation

1. Obesity. Overweight appears to represent the only postcutaneous factor interfering with vitamin D3 photosynthesis. Obese individuals have lower serum 25-hydroxyvitamin D levels than lean controls and also have blunted response to UVB. Oral absorption of vitamin D2 is also decreased in obesity. In vitro experiments of irradiation of skin pieces from obese and lean individuals showed similar epidermal 7-DHC content and in vitro response to UVB. These results are suggestive of defective translocation of the vitamin into the circulation, or defective plasma transport (357). Of note, obesity is also associated with increases in plasma FFA, which can displace vitamin D3 from plasma vitamin D binding protein (358). E. General comments

An important consideration on the physiology of vitamin D3 synthesis is that it represents a mostly biophysical reaction. Thus, the serum vitamin D3 response to UVB is not altered by the oral administration of pharmacological doses of vitamin D2 (359) or of 1,25-dihydroxyvitamin D (360). There are nevertheless local regulatory mechanisms that can influence the process. When exposure to sunlight is excessive, inactivation of previtamin D3 and vitamin D3 itself are well known consequences. Moreover, when high irradiance levels are sustained, melanocyte activity is enhanced, resulting in tanning and blunted response to acute UVB exposure. The opposite situation, decreased exposure to UVB with reduced vitamin D3 production, can be compensated, at least partly, by enhanced activity of the renal 1␣-hydroxylase enzyme (348). Lastly, there has been some controversy regarding the significance of findings in acute vs. chronic studies evaluating the vitamin D response to UVB. Within this context, it must be noted that acute studies are performed under more stringent conditions, i.e., mostly during the winter, to prevent the interference of ambient sunlight, which involves exposure of the whole body to UVB, and require a phototherapy unit that must be continuously calibrated. It is then apparent that biological significance is better evaluated in larger populations. Thus, small acute responses that do not reach statistical significance, such as those observed after selective irradiance of the upper extremities or head and neck, or after subthreshold doses of UVB, may still result in normal vitamin D3 levels, when irradiation is continued through much longer periods. A similar explanation would be operative in

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

black individuals, who show lack of response to acute UVB irradiance, yet also exhibit the normal seasonal variance (UVB dependent) in 25-hydroxyvitamin D serum levels (361).

VIII. Final Comments and Future Directions

The present work provides a conceptual framework for the organization of the cutaneous neuroendocrine system into epidermal and dermal units. Further, it develops an understanding of the integrated pattern of regulation of those endocrine units, and of the exquisite coordination in expression of their functional effects to achieve the best strategy for dealing with the continuous interactions between skin and environmental agents. Among the recently uncovered regulatory mechanisms are a myriad of precise interactions with receptors for hormones, neurotransmitters, and neuropeptides, e.g., hypothalamic, pituitary, and steroid hormones, PTH and PTHrP, vitamin D, neuropeptides, neurotrophins, and with nicotinic, muscarinic, adrenergic, serotonin, and glutamate neurotransmitter receptors, all widely expressed in cutaneous compartments. Surprisingly, the source of ligands for these receptors is the skin itself, sometimes the same histological compartment, or even the same cells. Thus, the conclusive documentation now available demonstrates that in addition to vitamin D3, the skin produces PTHrP, CRH, urocortin, POMC-derived peptides, enkephalins, catecholamines, acetylcholine, and neurotrophins. The skin is also involved in steroidogenesis and sex hormone conversion. Intriguing experimental data showing interconversion of thyroid hormones, however, require further confirmation. Existing data also justify further studies on cutaneous production and metabolism of neurohormones serotonin and melatonin and the neurotransmitters aspartate and glutamate. Rapid and efficient communication between the different cutaneous compartments is provided by the existing rich sensory innervation with its reservoir of neuropeptides and neurotransmitters, affording a high degree of specificity. A physiological role for this system is underscored by its effects on systemic immunity, by the production of vitamin D3 with its action on calcium homeostasis and bone mineralization, and by the generation of afferent neurogenic signals connecting the skin with the rest of the body. Concomitant functional studies have helped further clarify its function by disclosing that cutaneous neuroendocrine system activity can be effectively modified by exposure to common environmental factors such as solar radiation, by intrinsic signals such as those associated with hair cycling, by biological modifiers (cytokines/chemokines), or by local or systemic pathological conditions. This field represents, therefore, a fertile ground for future studies on cutaneous biology, including the application of more advanced methodology to confirm previous findings; the determination of other hormonal factors that could be produced by the skin; and the further definition of existent or yet-to-be-discovered regulatory pathways. Moreover, the potential clinical implications cannot be overlooked, as this area opens the possibility for multiple points of interaction on ongoing pathological processes. Possible pathological tar-

479

get conditions include not only inflammatory diseases, but benign hyperproliferative skin disorders, vasculopathic and autoimmune reactions, disorders of pigmentation and hair cycling, and malignant processes such as melanoma development and epidermal carcinogenesis. Specifically, locally produced POMC peptides ACTH and ␣-MSH can affect skin functions by enhancing melanogenesis, stimulating hair growth and sebaceous gland functions, and attenuating inflammatory responses. CRH, in addition to its local vasodilatory and proinflammatory effects, may also inhibit proliferation of epidermal keratinocytes. Vitamin D is already used in the therapy of psoriasis, and glucocorticoids are drugs of choice in the treatment of inflammatory skin disorders. Finally, the multidirectional communication between skin, endocrine, immune, and central nervous systems suggests that the skin may be an important regulator of global homeostasis acting as a sensor for external or internal disturbances, and as an effector/producer of humoral or neural signals sent to other coordinating centers. In this context, the possibility of pathological consequences for dysregulation in this cutaneous neuroendocrine system poses a powerful challenge that can only be addressed with a strong, coordinated, and multidisciplinary approach. Acknowledgments We thank Drs. C. Gomez-Sanchez, O. Johansson, W. B. Malarkey, J. Orloff, M. F. Holick, S. Asa, D. L. St. Germain, S. Grando, T. Smith, E. Wei, and L. Y. Matsuoka for their help in the preparation of the manuscript. The excellent secretarial work of Ms. LuEllen Giera is acknowledged.

References 1. Ackerman B, Chongchitnant Sanchez J, Gou Y, Bennin B, Reichel M, Randall MB 1997 Histologic Diagnosis of Inflammatory Skin Diseases, ed 2. Williams & Wilkins, Baltimore 2. Weedon D, Strutton G 1997 Skin Pathology. Churchill Livingstone, New York 3. Fitzpatrick TB, Eisen AZ, Wolff K, Freedberg IM, Austen KF 1993 Dermatology in General Medicine. McGraw Hill, New York 4. Goldsmith LA 1991 Physiology, Biochemistry, and Molecular Biology of the Skin. Oxford University Press, New York 5. Slominski A, Wortsman J, Paus R, Luger T, Salomon S 2000 Corticotropin releasing hormone and proopiomelanocortin involvement in the cutaneous response to stress. Physiol Rev 80: 979 –1020 6. Slominski A, Mihm MC 1996 Potential mechanism of skin response to stress. Int J Dermatol 35:849 – 851 7. Bos JD 1997 Skin Immune System (SIS). CRC Press, Boca Raton, FL 8. Nordlund JJ, Boissy RE, Hearing VJ, King RA, Ortonne JP 1998 The Pigmentary System: Physiology and Pathophysiology. Oxford University Press, New York 9. Slominski A, Pawelek J 1998 Animals under the sun: effects of ultraviolet radiation on mammalian skin. Clin Dermatol 16:503–515 10. Slominski A, Paus R, Schaderdorf D 1993 Melanocytes are sensory and regulatory cells in the epidermis. J Theor Biol 164:103–120 11. Milstone LM, Edelson RL 1988 Endocrine, metabolic and immunologic functions of keratinocytes. Ann NY Acad Sci 548:1–366 12. Grando SA 1993 Physiology of endocrine skin interrelations. J Am Acad Dermatol 28:981–992 13. Kragballe K, Fogh K, Larsen CG 1996 Vitamin D: actions and applications in dermatology. J Invest Dermatol Symp Proc 1:1–114 14. Holick MF 1994 McCollum award lecture, 1994: vitamin D–new horizons for the 21st century. Am J Clin Nutr 60:619 – 630 15. Philbrick WM, Wysolmerski JJ, Galbraith S, Holt E, Orloff JJ,

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

480

16. 17. 18. 19. 20. 21. 22.

23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38.

39. 40. 41. 42.

SLOMINSKI AND WORTSMAN Yang KH, Vasavada RC, Weir EC, Broadus AE, Stewart AF 1996 Defining the roles of parathyroid hormone-related protein in normal physiology. Physiol Rev 76:127–173 Slominski A, Paus R, Wortsman J 1993 On the potential role of proopiomelanocortin in skin physiology and pathology. Mol Cell Endocrinol 93:C1–C6 Luger T, Paus R, Slominski A, Lipton J 1999 Cutaneous neuromodulation: the proopiomelanocortin system. Ann NY Acad Sci 885:1– 479 Slominski A, Ermak G, Mazurkiewicz JE, Baker J, Wortsman J 1998 Characterization of corticotropin releasing hormone (CRH) in human skin. J Clin Endocrinol Metab 83:1020 –1024 Slominski A, Roloff B, Curry J, Dahiya M, Szczesniewski A, Wortsman J 2000 The skin produces urocortin. J Clin Endocrinol Metab 85:815– 823 Grando SA, Horton RM 1997 The keratinocyte cholinergic system with acetylcholine as an epidermal cytotransmitter. Curr Opin Dermatol 4:462–268 Schallreuter KU, Lemke KR, Pittelkow MR, Wood JM, Korner C, Malik R 1995 Catecholamines in human keratinocyte differentiation. J Invest Dermatol 104:953–957 Schallreuter KU, Wood JM, Pittelkow MR, Guˆtlich M, Lemke R, Rodl W, Swanson NN, Hitzemann K, Ziegler I 1994 Regulation of melanin biosynthesis in the human epidermis by tetrahydrobiopterin. Science 263:1444 –1446 Slominski A, Paus R 1990 Are L-tyrosine and L-dopa hormone-like bioregulators? J Theor Biol 143:123–138 Blalock JE 1989 Molecular basis for bidirectional communication between the immune and the neuroendocrine systems. Physiol Rev 69:1–32 Besedovsky HO, Del Rey A 1996 Immune-neuro-endocrine interactions: facts and hypotheses. Endocr Rev 17:64 –102 Pennisi E 1997 Tracing molecules that make the brain-body connection. Science 275:930 –931 Plotnikoff NP, Faith RE, Murgo AJ, Good RA 1999 Cytokines: Stress and Immunity. CRC Press, Boca Raton, FL Turnbull AV, Rivier CL 1999 Regulation of the hypothalamicpituitary-adrenal axis by cytokines: actions and mechanisms of action. Physiol Rev 79:1–71 Gilbert SF 1997 Developmental Biology, ed 5. Sinauer Assoc Inc, Sunderland, MA Montagna W, Ellis RA 1958 The Biology of Hair Growth. Academic Press, New York Paus R 1996 Control of the hair cycle and hair diseases as cycling disorders. Curr Opin Dermatol 3:248 –258 Halata Z 1990 Touch organs in the hairy and glabrous skin of some mammals (an ultrastructural comparison). Neth J Zool 40:329 –351 Rossi R, Johansson O 1998 Cutaneous innervation and the role of neuronal peptides in cutaneous inflammation: a minireview. Eur J Dermatol 8:299 –306 Belmonte C, Cervero F 1996 Neurobiology of Nociceptors. Oxford University Press, New York Thody AJ, Shuster S 1989 Control and function of sebaceous glands. Physiol Rev 69:383– 416 Feling P, Baxter JD, Frohman LA 1995 Endocrinology and Metabolism, ed 3. McGraw-Hill, New York Thiboutot DM 1995 Dermatological manifestations of endocrine disorders. J Clin Endocrinol Metab 80:3082–3087 Scholzen T, Armstrong CA, Bunnett NW, Luger TA, Olerud JE, Ansel JC 1998 Neuropeptides in the skin: interactions between the neuroendocrine and the skin immune systems. Exp Dermatol 7:81–96 Al Abadie MS, Kent GG, Gawkrodger DJ 1994 The relationship between stress and the onset and exacerbation of psoriasis and other skin conditions. Br J Dermatol 130:199 –203 Perrin MH, Vale W 1999 Corticotropin releasing factor receptors and their ligand family. Ann NY Acad Sci 885:312–328 Aguilera G 1998 Corticotropin releasing hormone, receptor regulation and the stress response. Trends Endocrinol 9:329 –336 Slominski A, Ermak G, Hwang J, Chakraborty A, Mazurkiewicz J, Mihm M 1995 Proopiomelanocortin, corticotropin releasing hormone and corticotropin releasing hormone receptor genes are expressed in human skin. FEBS Lett 374:113–116

Vol. 21, No. 5

43. Slominski A, Ermak G, Hwang J, Mazurkiewicz J, Corliss D, Eastman A 1996 The expression of proopiomelanocortin (POMC) and of corticotropin releasing hormone receptor (CRH-R) genes in mouse skin. Biochim Biophys Acta 1289:247–251 44. Roloff B, Fechner K, Slominski A, Furkert J, Botchkarev VA, Bulfone-Paus S, Zipper J, Krause K, Paus R 1998 Hair cycledependent expression of corticotropin releasing hormone (CRH) and CRH receptors (CRH-R) in murine skin. FASEB J 12:287–297 45. Arbiser JI, Karalis K, Viswanathan A, Koike C, Anand-Apte B, Flynn E, Zetter B, Majzoub JA 1999 Corticotropin-releasing hormone stimulates angiogenesis and epithelial tumor growth in the skin. J Invest Dermatol 113:838 – 842 46. Slominski A, Botchkarev V, Choudhry M, Fazal N, Fechner K, Furkert J, Krause E, Roloff B, Sayeed M, Wei E, Zbytek B, Zipper J, Wortsman J, Paus R 1999 Cutaneous expression of CRH and CRH-R: is there a “skin stress system”? Ann NY Acad Sci 885: 287–311 47. Fazal N, Slominski A, Choudhry MA, Wei ET, Sayeed MM 1998 Effect of CRF and related peptides on calcium signaling in human and rodent melanoma cells. FEBS Lett 435:187–190 48. Theoharides T, Singh L, Boucher W, Pang X, Letourneau R, Webster E, Chrousos G 1998 Corticotropin-releasing hormone induces skin mast cell degranulation and increased vascular permeability, a possible explanation for its proinflammatory effects. Endocrinology 139:403– 413 49. Funasaka Y, Sato H, Chakraborty AK, Ohashi A, Chrousos GP, Ichihashi M 1999 Expression of proopiomelanocortin, corticotropin-releasing hormone (CRH), and CRH receptor in melanoma cells, nevus cells, and normal melanocytes. J Invest Dermatol Symp Proc 4:105–109 50. Kiang JG 1997 Corticotropin-releasing factor-like peptides increase cytosolic [Ca2⫹] in human epidermoid A-431 cells. Eur J Pharmacol 329:237–244 51. Slominski A, Roloff B, Zbytek B, Wei ET, Fechner K, Curry J, Wortsman J 2000 Corticotropin releasing hormone (CRH) and related petides can act as bioregulatory factors in human keratinocytes. In Vitro Cell Dev Biol 36:211–216 52. Fleisher-Berkovich S, Danon A 1995 Effect of corticotropinreleasing factor on prostaglandin synthesis in endothelial cells and fibroblasts. Endocrinology 136:4068 – 4072 53. Fleisher-Berkovich Rimon SG, Danon A 1998 Corticotropin releasing factor modulates interleukin-1-induced prostaglandin synthesis in fibroblasts: receptor binding and effects of antagonists. Regul Pept 77:121–126 54. Rohde E, Furdert J, Fechner K, Beyermann M, Mulvany MJ, Richter RM, Denef C, Bienert M, Berger H 1996 Corticotropin releasing hormone (CRH) receptors in the mesenteric small arteries of rats resemble the (2)-subtype. Biochem Pharmacol 52:829 – 833 55. Chrousos GP 1995 The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med 332:1351–1362 56. Karalis K, Muglia LJ, Bae D, Hilderbrand H, Majzoub JA 1997 CRH and the immune system. J Neuroimmunol 72:131–136 57. Singh LK, Boucher W, Pang X, Letourneau R, Seretakis D, Green M, Theoharides TC 1999 Potent mast cell degranulation and vascular permeability triggered by urocortin through activation of corticotropin releasing hormone receptors. J Pharmacol Exp Ther 288:1349 –135 58. Wei ET, Thomas HA 1993 Anti-inflammatory peptide agonists. Annu Rev Pharmacol Toxicol 33:91–108 59. Wei ET, Gao GC, Thomas HA 1993 Peripheral anti-inflammatory actions of corticotropin-releasing factor. Ciba Found Symp 172: 258 –268; discussion 268 –267 60. Schafer M, Mousa SA, Stein C 1997 Corticotropin-releasing factor in antinociception and inflammation. Eur J Pharmacol 323:1–10 61. Cone RD, Lu D, Koppula S, Vage DI, Klungland H, Boston B, Chen W, Orth DN, Pouton C, Kesterson RA 1996 The melanocortin receptors: agonists, antagonists, and the hormonal control of pigmentation. Recent Prog Horm Res 51:287–317; discussion 318 62. Van Der Kraan M, Adan RA, Entwistle ML, Gispen WH, Burbach JP, Tatro JB 1998 Expression of melanocortin-5 receptor in secretory epithelia supports a functional role in exocrine and endocrine glands. Endocrinology 139:2348 –2355 63. Boston BA, Cone RD 1996 Characterization of melanocortin re-

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

64.

65.

66. 67. 68. 69. 70.

71. 72. 73.

74. 75.

76.

77.

78. 79.

80.

81.

82. 83. 84.

NEUROENDOCRINOLOGY OF THE SKIN

ceptor subtype expression in murine adipose tissues and in the 3T3–L1 cell line. Endocrinology 137:2043–2050 Luger TA, Scholzen T, Brzoska T, Becher E, Slominski A, Paus R 1998 Cutaneous immunomodulation and coordination of skin stress responses by ␣-melanocyte-stimulating hormone. Ann NY Acad Sci 840:381–394 Ermak G, Slominski A 1997 Production of POMC, CRH-R1, MC-1, and MC-2- receptor mRNA and expression of tyrosinase gene in relation to hair cycle and dexamethasone treatment in the C57BL/6 mouse skin. J Invest Dermatol 108:160 –165 Slominski A, Ermak G, Mihm M 1996 ACTH receptor, CYP11A1, CYP17 and CYP21A2 genes are expressed in skin. J Clin Endocrinol Metab 81:2746 –2749 Eberle AN 1988 The Melanotropins: Chemistry, Physiology and Mechanism of Action. Karger, New York Hadley M 1996 Endocrinology. Prentice Hall, Englewood Cliffs, NJ Siegrist W, Eberle AN 1995 Melanocortin and their implication in melanoma. Trends Endocrinol Metab 6:115–120 Suzuki I, Cone RD, Im S, Nordlund J, Abdel-Malek ZA 1996 Binding of melanotropin hormones to the melanocortin receptor MC1R on human melanocytes stimulates proliferation and melanogenesis. Endocrinology 137:1627–1633 Slominski A, Costantino R, Wortsman J, Paus R, Ling N 1992 Melanotropic activity of ␥ MSH peptides in melanoma cells. Life Sci 50:1103–1108 Lerner AB 1993 The discovery of the melanotropins: a history of pituitary endocrinology. Ann NY Acad Sci 680:1–12 Ling N, Ying S, Minick S, Guillemin R 1979 Synthesis and biological activity of four ␥-melanotropin peptides derived from the cryptic region of the adrenocorticotropin/␤-lipotropin precursor. Life Sci 25:1773–1780 Moellmann G, Slominski A, Kuklinska E, Lerner AB 1988 Regulation of melanogenesis in melanocytes. Pigment Cell Res [Suppl 1]:79 – 87 Pawelek JM, Chakraborty AL, Osber MP, Orlow SJ, Min KK, Rosenzweig KE, Bolognia JL 1992 Molecular cascades in UVinduced melanogenesis: a central role for melanotropins? Pigment Cell Res 5:348 –356 Pawelek JM, Fleischmann R, McLane J, Guillette B, Emanuel J, Korner A, Bergstrom A, Murray M 1984 Studies on growth and pigmentation of Cloudman S91 melanoma cells. Pigment Cell Res 6:521–533 Slominski A, Moellmann G, Kuklinska E 1989 MSH inhibits growth in a line of amelanotic hamster melanoma cells and induces increases in cAMP levels and tyrosinase activity without inducing melanogenesis. J Cell Sci 92:551–559 Pawelek JM, Halaban R, Christie G 1975 Melanoma cells with a cyclic AMP growth requirement. Nature 258:539 –540 Abdel-Malek Z, Swope VB, Suzuki I, Akcali MD, Harriger ST, Boyce K, Urabe K, Hearing VJ 1995 Mitogenic and melanogenic stimulation of normal human melanocytes by melanotropic peptides. Proc Natl Acad Sci USA 92:1789 –1793 Hedley SJ, Gawkroder DJ, Weetman AP, Morandini R, Boeynaems J-M, Ghanem G, Neil SM 1998 ␣-Melanocyte stimulating hormone inhibits tumour necrosis factor- ␣ stimulated intercellular adhesion molecule-1 expression in normal cutaneous human melanocytes and in melanoma cell lines. Br J Dermatol 138:536 –543 Kiss M, Wlaschek M, Brenneisen P, Michel G, Hommel C, Lange T, Peus D, Kemeny L, Dobozy A, Scharffetter-Kochanek K, Ruzicka T 1995 ␣-Melanocyte stimulating hormone induces collagenase/matrix metalloproteinase-1 in human dermal fibroblasts. J Biol Chem 376:425– 430 Luger TA, Scholzen T, Grabbe S 1997 The role of ␣-melanocytestimulating hormone in cutaneous biology. J Invest Dermatol Symp Proc 2:87–93 Paus R, Botchkarev VA, Botchkareva NV, Fechner K, Furkert J, Roloff B, Slominski A 1999 The skin POMC system (SPS): leads and lessons from the hair follicle. Ann NY Acad Sci 885:350 –363 Chen W, Kelly MA, Opitz-Araya X, Thomas RE, Low MJ, Cone RD 1997 Exocrine gland dysfunction in MC5-R-deficient mice: evidence for coordinated regulation of exocrine gland function by melanocortin peptides. Cell 91:789 –798

481

85. Paus R, Maurer M, Slominski A, Czarnetzki B 1994 Mast cell involvement in murine hair growth. Dev Biol 163:230 –240 86. Rose J 1998 Adrenocorticotropic hormone (ACTH) but not ␣melanocyte stimulating hormone (␣-MSH) as a mediator of adrenalectomy induced hair growth in mink. J Invest Dermatol 110: 456 – 457 87. Bigliardi PL, Bigliardi-Qi M, Buchner S, Rufli T 1998 Expression of ␮-opiate receptor in human epidermis and keratinocytes. J Invest Dermatol 111:297–301 88. Zagon IS, Wu Y, McLaughlin PJ 1996 The opioid growth factor, met-enkephalin, and the ␨ opioid receptor are present in human and mouse skin and tonically act to inhibit DNA synthesis in the epidermis. J Invest Dermatol 106:490 – 497 89. Nissen JB, Kragballe K 1997 Enkephalins modulate differentiation of normal human keratinocytes in vitro. Exp Dermatol 6:222–229 90. Oakes SR, Haynes KM, Waters MJ, Herington AC, Werther GA 1992 Demonstration and localization of growth hormone receptor in human skin and skin fibroblasts. J Clin Endocrinol Metab 75: 1368 –1373 91. Tavakkol A, Elder JT, Griffiths CE, Cooper KD, Talwar H, Fisher GJ, Keane KM, Foltin SK, Voorhees JJ 1992 Expression of growth hormone receptor, insulin-like growth factor 1 (IGF-1) IGF-1 receptor mRNA and proteins in human skin. J Invest Dermatol 99: 343–349 92. Lobie PE, Breipohl W, Lincoln DT, Garcia-Aragon J, Waters MJ 1990 Localization of the growth hormone receptor/binding protein in skin. J Endocrinol 126:467– 471 93. Lobie PE, Garcia-Aragon J, Wang BS, Baumbach WR, Waters MJ 1992 Cellular localization of the growth hormone binding protein in the rat. Endocrinology 130:3057–3065 94. Deplewski D, Rosenfield RL 1999 Growth hormone and insulinlike growth factors have different effects on sebaceous cell growth and differentiation. Endocrinology 140:4089 – 4094 95. Edmondson SR, Russo VC, McFarlane AC, Wraight CJ, Werther GA 1999 Interactions between growth hormone, insulin-like growth factor I, and basic growth factor in melanocyte growth. J Clin Endocrinol Metab 84:1638 –1644 96. Matsuoka LY, Wortsman J, Kupchella CE, Eng A, Dietrich JE 1982 Histochemical characterization of the cutaneous involvement of acromegaly. Arch Intern Med 142:1820 –1823 97. Wanke R, Milz S, Rieger N, Ogiolda L, Renner-Muller I, Brem G, Hermanns W, Wolf E 1999 Overgrowth of skin in growth hormone transgenic mice depends on the presence of male gonads. J Invest Dermatol 113:967–971 98. Ouhtit A, Morel G, Kelly PA 1993 Visualization of gene expression of short and long forms of prolactin receptor in the rat. Endocrinology 133:135–144 99. Choy VJ, Nixon AJ, Pearson AJ 1995 Localization of receptors for prolactin in ovine skin. J Endocrinol 144:143–151 100. Rose J, Garwood T, Jaber B 1995 Prolactin receptor concentration in the skin of mink during the winter fur growth cycle. J Exp Zool 271:205–210 101. Girolomoni G, Phillips JT, Bergstresser PR 1993 Prolactin stimulates proliferation of cultured human keratinocytes. J Invest Dermatol 101:275–279 102. Pabon JE, Bird JS, Li X, Huang ZH, Lei ZM, Sanfilippo JS, Yussman MA, Rao CV 1996 Human skin contains luteninizing hormone/chorionic gonadotropin receptors. J Clin Endocrinol Metab 81:2738 –2741 103. Venencie PY, Meduri G, Pissard S, Jolivet A, Loosefelt H, Milgrom E, Misrahi M 1999 Luteinizing hormone/human chorionic gonadotrophin receptors in various epidermal structures. Br J Dermatol 141:438 – 446 104. Dicks P, Russel AJF, Lincoln GA 1994 The role of prolactin in the reactivation of hair follicles in relation to moulting in cashmere goats. J Endocrinol 143:441– 448 105. Rufaut NW, Pearson AJ, Nixon AJ, Wheeler TT, Wilkins RJ 1999 Identification of differentially expressed genes during a wool follicle growth cycle induced by prolactin. J Invest Dermatol 113: 865– 872 106. Paus R 1991 Does prolactin play a role in skin biology and pathology? Med Hypotheses 36:33– 42 107. Clevenger CV, Freier DO, Kline JB 1998 Prolactin receptor signal

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

482

108.

109.

110. 111. 112. 113. 114.

115. 116. 117.

118. 119. 120.

121.

122.

123. 124.

125.

126.

127. 128.

SLOMINSKI AND WORTSMAN transduction in cells of the immune system. J Endocrinol 157: 187–197 Scholzen TE, Brzoska T, Kalden D-H, O’Reilly FO, Armstrong CA, Luger TA, Ansel JC 1999 Effect of ultraviolet light on the release of neuropeptides and neuroendocrine hormone in the skin: mediators of photodermatitis and cutaneous inflammation. J Invest Dermatol Symp Proc 4:55–59 Bianchi B, Matucci Danesi A, Rossi R, Ipponi P, Giannotti B, Johansson O, Cappugi P 1999 Characterization of [3H] substance P binding sites in human skin. J Eur Acad Dermatol Venereol 12:6 –10 Misery L 1998 Langerhans cells in the neuro-immuno-cutaneous system. J Neuroimmunol 89:83– 87 Misery L 1997 Skin, immunity and the nervous system. Br J Dermatol 137:843– 850 Eedy DJ 1993 Neuropeptides in skin. Br J Dermatol 128:597– 605 O’Sullivan RL, Lipper G, Lerner EA 1998 The neuro-immunocutaneous-endocrine network: relationship of mind and skin. Arch Dermatol 134:1431–1435 Streilein JW, Alard P, Niizeki H 1997 A new concept of skinassociated lymphoid tissue (salt): UVB light impaired cutaneous immunity reveals a prominent role for cutaneous nerves. Keio J Med 48:22–27 Theoharides TC 1996 The mast cell: a neuroendocrine master player. Int J Tissue React 18:1–21 Paus R, Heinzelman T, Schulz KD, Furtkert J, Fechner K, Czarnetzki BM 1994 Hair growth induction by substance P. Lab Invest 71:134 –140 Takahashi KT, Nakanishi S, Imamura S 1993 Direct effects of cutaneous neuropeptides on adenylyl cyclase activity and proliferation in a keratinocyte cell line: stimulation of cyclic AMP formation by CGRP and VIP/PHM, and inhibition by NPY through G protein-coupled receptors. J Invest Dermatol 101:646 – 651 Torii H, Tamaki K, Granstein RD 1999 The effect of neuropeptides/hormones on Langerhans cells. J Dermatol Sci 20:21–28 Hara M, Toyoda M, Yaar M, Bhawan J, Avila EM, Penner IR, Gilchrest BA 1996 Innervation of melanocytes in human skin. J Exp Med 184:1385–1395 Toyoda M, Luo Y, Makino T, Matsui C, Morohashi M 1999 Calcitonin gene-related peptide upregulates melanogenesis and enhances melanocyte dendricity via induction of keratinocytederived melanotropic factors. J Invest Dermatol Symp Proc 4: 116 –125 Luis J, Martin JM, El Battari A, Reynier M, Marvaldi J, Pichon J 1989 A human melanoma-derived cell line (IGR 39) with a very high number of vasoactive-intestinal-peptide (VIP) receptors. 1. Molecular characterization of the binding site. Eur J Biochem 180: 429 – 433 Luis J, Martin JM, El Battari A, Reynier M, Marvaldi J, Pichon J 1989 A human melanoma-derived cell line (IGR 39) with a very high number of vasoactive-intestinal-peptide (VIP) receptors. 2. Effect of VIP on cAMP production and on cell-surface VIP-binding sites. Eur J Biochem 180:435– 439 Gilchrest BA, Park H-Y, Eller MS, Yaar M 1996 Mechanisms of ultraviolet light-induced pigmentation. Photochem Photobiol 63:1–10 Yaar M, Eller MS, DiBenedetto P, Reenstra WR, Zhai S, McQuaid T, Archambault M, Gilchrest BA 1994 The trk family of receptors mediates nerve growth factor and neurotrophin-3 effects in melanocytes. J Clin Invest 94:1550 –1562 Pincelli C, Hakke AR, Bernassi L, Grassili E, Magnoni C, Ottani D, Polakowska R, Franceschi C, Gianneti A 1997 Autocrine nerve growth factor protects human keratinocytes from apoptosis through its high affinity receptor (trk): a role for bcl-2. J Invest Dermatol 109:757–764 Fantini F, Johansson O 1992 Expression of growth-associated protein 43 and nerve growth factor receptor in human skin: a comparative immunohistochemical investigation. J Invest Dermatol 99:734 –741 Liang Y, Johnansson O 1998 Light and electron microscopic demonstration of the p75 nerve growth factor receptor in normal human cutaneous nerve fibers: new vistas. J Invest Dermatol 1998:114 –118 Paus R, Peters EMJ, Eichenmu¨eller S, Botchkarev VA 1997 Neural

129.

130.

131. 132.

133.

134. 135.

136.

137.

138.

139.

140. 141.

142.

143.

144. 145. 146. 147.

Vol. 21, No. 5

mechanisms of hair growth control. J Invest Dermatol Symp Proc 2:61– 68 Botchkarev VA, Welker P, Albers KM, Botchkareva NV, Metz M, Lewin GR, Bulfone-Paus S, Peters EMV, Lindner G, Paus R 1998 A new role for neurotrophin-3 involvement in the regulation of hair follicle regression (catagen). Am J Pathol 153:785–799 Botchkarev VA, Botchkareva NV, Albers KM, van der Veen C, Lewin GR, Paus R 1998 Neurotrophin-3 involvement in the regulation of the hair follicle morphogenesis. J Invest Dermatol 111: 279 –285 Botchakarev VA, Kief S, Paus R 1999 Overexpression of brainderived neurotrophic factor increase Merkel cell number in murine skin. J Invest Dermatol 113:691– 692 Botchkareva NV, Botchkarev VA, Metz M, Silos-Santiago I, Paus R 1999 Retardation of hair follicle development by the deletion of trkC, high-affinity neurotrophin-3 receptor. J Invest Dermatol 113: 425– 427 Johannsson O, Hilleges M, Toomas T, Marcusson JA, Wetterberg L 1994 Somatostatin immunoreactive cells in lesional psioriatic human skin during peptide T treatment. Acta Derm Venereol 74: 106 –109 Endo T, Ohta K, Harahuchi K, Onaya T 1995 Cloning and functional expression of thyrotropin receptor cDNA from rat fat cells. J Biol Chem 270:10833–10837 Hanafin NM, Chen TC, Heinrich G, Segre GV, Holick MF 1995 Cultured human fibroblasts and not cultured human keratinocytes express a PTH/PTHrP receptor mRNA. J Invest Dermatol 105:133–137 Blomme EAG, Sugimoto Y, Lin YC, Capen CC, Rosol TJ 1998 Parathyroid hormone-related protein is a positive regulator of keratinocyte growth factor expression by normal dermal fibroblasts. Mol Cell Endocrinol 152:189 –197 Sharpe GR, Dillon JP, Durham B Gallagher JA, Fraser WD 1998 Human keratinocytes express transcripts for three isoforms of parathyroid hormone related protein (PTHrP), but not for the parathyroid hormone/PTHrP receptor; effects of 1,25 (OH)2 vitamin D3. Br J Dermatol 138:944 –951 Orloff JJ, Ganz MB, Nathanson MH, Moyer MS, Kats Y, Mitnick M, Behal A, Gasalla-Herraiz J, Isales CM 1996 A midregion parathyroid hormone-related peptide mobilizes cytosolic calcium and stimulates formation of inositol triphosphate in a squamous carcinoma cell line. Endocrinology 137:5376 –5385 Orloff JJ, Kats Y, Urena P, Schipani E, Vasavada RC, Philbrick WM, Behal A, Abou-Samra A-B, Segre GV, Juppner H 1995 Further evidence for a novel receptor for amino-terminal parathyroid hormone-related protein on keratinocytes and squamous carcinoma cell lines. Endocrinology 135:3016 –3023 Holick MF, Ray S, Chen TC, Tian X, Persons KS 1994 A parathyroid antagonist stimulates epidermal proliferation and hair growth in mice. Proc Natl Acad Sci USA 91:8014 – 8016 Wysolmerski JJ, Broadus AE, Zhou J, Fuchs E, Milstone LM, Phillbrick WM 1994 Overexpression of parathyroid hormonerelated protein in the skin of transgenic mice interferes with hair follicle development. Proc Natl Acad Sci USA 91:1133–1137 Whitfield JF, Chakravarthy BR, Durkin JP, Isaacs RJ, Jouishomme H, Sikorska M, Williams RE, Rixon RH 1992 Parathyroid hormone stimulates protein kinase C but not adenylate cyclase in mouse epidermal keratinocytes. J Cell Physiol 150:299 –303 Orloff JJ, Ganz MB, Ribaudo AE, Burtis WJ, Reiss M, Milstone LM, Stewart AF 1992 Analysis of PTHrP binding and signal transduction mechanism in benign and malignant squamous cells. Am J Physiol 262:E599 –E607 Blomme EAG, Zhou H, Kartsogiannis V, Capen CC, Rosol TJ 1999 Spatial and temporal expression of parathyroid hormone-related protein during wound healing. J Invest Dermatol 112:788 –795 Studzinski GP, Moore DC 1995 Sunlight-can it prevent as well as cause cancer? Cancer Res 55:4014 – 4022 Reichrath J, Schilli M, Kerber A, Bahmer FA, Czarnetzki BM, Paus R 1994 Hair follicle expression of 1,25-dihydroxyvitamin D3 receptors during the murine hair cycle. Br J Dermatol 131:477– 482 Paus R, Schilli MB, Handjiski B, Menrad A, Henz BM, Plonka P 1996 Topical calcitirol enhances normal hair regrowth but does not

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

148. 149. 150. 151.

152. 153. 154. 155.

156. 157.

158.

159.

160. 161.

162.

163. 164. 165. 166. 167.

168. 169.

NEUROENDOCRINOLOGY OF THE SKIN

prevent chemotherapy-induced alopecia in mice. Cancer Res 56: 4438 – 4443 Ratnam AV, Bikle DD, Cho J-K 1999 1,25 dihydroxyvitamin D3 enhances the calcium response of keratinocytes. J Cell Physiol 178:188 –196 Feichrath J, Komradt J, Zhe XH, Hong X, Tilgen W, Holick M 1999 Analysis of 1,25 dihydroxyvitamin D3 receptors (VDR) in basal cell carcinomas. Am J Pathol 155:583–589 Malloy PJ, Pike JW, Feldman D 1999 The vitamin D receptor and the syndrome of hereditary 1,25-dihydroxyvitamin D-resistant rickets. Endocr Rev 20:156 –198 Ranson M, Posen S, Mason RS 1988 Human melanocytes as a target tissue for hormones: in vitro studies with 1 ␣25, dihydroxyvitamin D3, ␣-melanocyte stimulating hormone, and ␤-estradiol. J Invest Dermatol 91:593–598 Mansur CP, Gordon PR, Ray S, Holick MF, Gilchrest BA 1988 Vitamin D, its precursors and metabolites do not affect melanization of cultured human melanocytes. J Invest Dermatol 91:16 –21 Muller K, Bendtzen K 1996 1,25-Dihydroxyvitamin D3 as a natural regulator of human immune functions. J Invest Dermatol Symp Proc 1:68 –71 Serres M, Viac J, Schmidt D 1996 Glucocorticoid receptor localization in human epidermal cells. Arch Dermatol Res 288:140 –146 Karstila T, Rechardt L, Honkaniemi J, Gustafsson JA, Wikstroms AC, Karppinen A, Peltohuikko M 1994 Immunocytochemical localization of glucocorticoid receptor in rat skin. Histochemistry 102:305–309 Budunova IV, Carbajal S, Kang H, Viaje A, Slaga TG 1997 Altered glucocorticoid receptor expression and function during mouse carcinogenesis. Mol Carcinog 18:177–185 Kumuves LG, Hanley K, Jiang Y, Elias PM, Williams ML, Feingold KR 1998 Ligands and activators of nuclear hormone receptors regulate epidermal differentiation during fetal rat skin development. J Invest Dermatol 111:429 – 433 Kenough S, Lombers M, Delahaye F, Eugene E, Bonvalet J-P, Farman N 1994 Human skin as target for aldosterone: coexpression of mineralocorticoid receptors and 11 36-hydroxysteroid dehydrogenase. J Clin Endocrinol Metab 79:1334 –1341 Hirasawa G, Sasano H, Suzuki T, Takeyama j Muramatu Y, Fukushima K, Hiwatashi N, Toyota T, Nagura H, Krozowski ZS 1999 11 ␤-hydroxysteroid dehydogenase type 2 and mineralocortoid receptor in human fetal development. J Clin Endocrinol Metab 84:1453–1458 Takayasu S 1979 Metabolism and action of androgen in the skin. Int J Dermatol 18:681– 692 Kimura N, Mizok A, Oonuma T, Sasano H, Nagura H 1993 Immunocytochemical localization of androgen receptor with polyclonal antibody in paraffin-embedded human tissues. J Histocherm Cytochem 41:671– 678 Choudhry R, Hodgins MB, Van der Kwast TH, Brinkman AO, Boersma WJA 1992 Localization of androgen receptors in human skin by immunohistochemistry: implications for the hormonal regulation of hair growth, sebaceous glands and sweat glands. J Endocrinol 133:467– 475 Randall VA 1994 Androgens and human hair growth. Clin Endocrinol (Oxf) 40:439 – 457 Orfanos CE, Happle R 1990 Hair and Hair Diseases. SpringerVerlag, New York Wilson CM, McPhaul MJ 1996 A and B forms of the androgen receptor are expressed in a variety of human tissues. Mol Cell Endocrinol 120:51–57 Offidani A, Campanati A 1999 Papillary hidradenoma: immunohistochemical analysis of steroid receptor profile with a focus on apocrine differentiation. J Clin Pathol 52:829 – 832 Uotinen N, Puustinen R, Pasanen S, Manninene T, Kivineva M, Syvala H, Tuohima P, Ylokomi T 1999 Distribution of progesterone receptor in female mouse tissues. Gen Comp Endocrinol 115: 429 – 441 Wallace ML, Smoller BR 1998 Estrogen and progesterone receptors in androgenic alopecia vs. alopecia areata. Am J Dermatopathol 20:160 –163 Tadokoro T, Itami S, Hosokawa K, Terashi H, Takayasu S 1997

170. 171. 172.

173. 174. 175. 176. 177.

178.

179. 180.

181. 182.

183.

184. 185. 186. 187. 188. 189. 190. 191.

483

Human genital melanocytes as androgen target cells. J Invest Dermatol 109:513–517 McLeod SD, Ranson M, Mason RS 1994 Effects of estrogens in human melanocytes in vitro. J Steroid Biochem Mol Biol 49:9 –14 Jee S-H, Lee S-Y, Chiu H-C, Chang C-C, Chen TJ 1994 Effects of estrogen and estrogen receptor in normal human melanocytes. Biochem Biophys Res Commun 199:1407–1412 Ashcroft GS, Greenwell-Wild T, Horan MA, Wahl SM, Ferguson MWJ 1999 Topical estrogen accelerates cutaneous wound healing in aged humans associated with an altered inflammatory response. Am J Pathol 155:1137–1146 Oh HS, Smart RC 1996 An estrogen receptor pathway regulates the telogen-anagen hair follicle transition and influences epidermal cell proliferation. Proc Natl Acad Sci USA 93:12525–125230 Zava DT, Goldhirsch A 1983 Estrogen receptor in malignant melanoma: fact or artefact? Eur Cancer Clin Oncol 19:1151–1159 Holt PJ 1978 In vitro responses of the epidermis to triiodothyronine. J Invest Dermatol 71:202–204 Holt PJ, Marks R 1977 The epidermal response to change in thyroid status. J Invest Dermatol 68:299 –301 Torma H, Rollman O, Vahlquist A 1993 Detection of messenger transcripts for retinoic acid, vitamin D3, and thyroid hormone (c-erb-A) nuclear receptors in human skin using reverse transcription and polymerase chain reaction. Acta Dermatol Venereol 73: 102–107 Klann RC, Torres B, Menke JB, Holbrook CT, Bercu BB, Usala SJ 1993 Competetive polymerase chain reaction quantitiation of c-erbA␤1, c-erbA␣1, and c-erbA␣2 messenger ribonucleic acid levels in normal, heterozygous, and homozygous fibroblasts of kindreds with thyroid hormone resistance. J Clin Endocrinol Metab 77:969 –975 Smith TJ, Kline EL 1988 Multi-hormonal regulation of tyrosinase expression in B16/C3 melanoma cells in culture. Prog Clin Biol Res 262:241–255 Grando S, Horton RM, Mauro TM, Kist DA, Lee TX, Dahl MV 1996 Activation of keratinocyte nicotinic cholinergic receptors stimulates calcium influx and enhances cell differentiation. J Invest Dermatol 107:412– 418 Grando S 1997 Biological function of keratinocyte cholinergic receptors. J Invest Dermatol Symp Proc 2:41– 48 Ndoye A, Buchli R, Greenberg B, Nguyen VT, Zia S, Rodriguez JG, Webber RJ, Lawry MA, Grando SA 1998 Identification and mapping of keratinocyte muscarinic acetylcholine receptor subtypes in human epidermis. J Invest Dermatol 111:410 – 416 Kohn EC, Alessandro R, Probst J, Jacobs W, Brilley E, Felder CC 1996 Identification and molecular characterization of a m5 muscarinic receptor in A2058 human melanoma cells. Coupling to inhibition of adenylyl cyclase and stimulation of phospholipase A2. J Biol Chem 271:17476 –17484 Noda S, Lammerling-Koppel M, Oettling G, Drews U 1998 Characterization of muscarinic receptors in the human melanoma cell line SK-Mel-28 via calcium mobilization. Cancer Lett 13:107–14 Steinkraus V, Mak JCW, Pichlmeier U, Mensing H, Ring J, Barnes PJ 1996 Autoradiographic manning of ␤ adrenoceptors in human skin. Arch Dermatol Res 288:549 –553 Steinkraus V, Steinfath M, Korner C, Mensing H 1992 Binding of ␤-adrenergic receptors in human skin. J Invest Dermatol 111: 470 – 480 Schallreuter KU, Wood JM, Lemke R, LePoole C, Das P, Westerhof W, Pittelkow MR, Thody A 1992 Production of catecholamines in the human epidermis. Biochem Biophys Res Commun 189:72–78 Drummond PD, Skipworth S, Finch PM 1996 ␣(1)-Adrenoceptors in normal and hyperalgesic human skin. Clin Sci 91:73–77 Bissonnette EY, Befus AD 1997 Anti-inflammatory effect of ␤(2)-agonists-inhibition of TNF-␣ release from human mast cells. J Allergy Clin Immunol 100:825– 831 Ding ZQ, Jiang MZ, Li SH, Zhang YF 1995 Vascular barrierenhancing effect of an endogenous ␤-adrenergic agonist. Inflammation 19:1– 8 Harada K, Ohaski K, Fujimura A, Kumagai Y, Ebihara A 1996 Effect of ␣ (1)-adrenoceptor antagonist, prazosin and urapidil, on a finger skin vasoconstrictor response to cold stimulation. Eur Clin Pharmacol 49:371–375

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

484

SLOMINSKI AND WORTSMAN

192. Deoliveira AR, Castrucci AML, Viscounti MA 1996 Cellular signaling in vertebrate pigment cells. Braz J Med Biol Res 29:1743–1749 193. Arner P 1995 The ␤3 adrenergic receptor. A cause and cure of obesity? N Engl J Med 261:25–33 194. Genever PG, Maxfield SJ, Kennovin GD, Maltman J, Bowgen CJ, Raxworthy MJ, Skerry TM 1999 Evidence for a novel glutamatemediated signaling pathway in keratinocytes. J Invest Dermatol 112:337–342 195. Askenase PW 1977 Role of basophils, mast cells, and vasomines in hypersensitivity reactions with a delayed time course. Prog Allergy 23:199 –320 196. Tachibana T, Taniguchi S, Furukawa F, Miwa S, Imamura S 1990 Serotonin metabolism in the Arthus reaction. J Invest Dermatol 94:120 –125 197. Fujji E, Irie K, Uchida Y, Tsukahara F, Muraki T 1994 Possible role of nitric oxide in 5-hydroxytryptamine-induced increase in vascular permeability in mouse skin. Naunyn Schmiedebergs Arch Pharmacol 350:361–364 198. Weisshaar E, Ziethen B, Gollmick H 1997 Can a serotonin type 3 (5-HT3) receptor antagonist reduce experimentally-induced itch? Inflamm Res 46:412– 416 199. Carlton SM, Coggeshall RE 1997 Immunohistochemical localization of 5-HT2A receptors in peripheral sensory axons in rat glabrous skin. Brain Res 763:271–275 200. Matsuda H, Ushio H, Geba GP, Askenase PW 1997 Human platelets can initiate T cell-dependent contact sensitivity through local serotonin release mediated by IgE antibodies. J Immunol 158:2891– 2897 201. Sauer SK, Schafer D, Kress M, Reeh PW 1998 Stimulated prostaglandin E-2 release from rat skin, in vitro. Life Sci 62:2045–2055 202. Maurer M, Opitz M, Henz BM, Paus R 1997 The mast cell products histamine and serotonin stimulate and TNF-␣ inhibits the proliferation of murine epidermal keratinocytes in situ. J Dermatol Sci 16:79 – 84 203. Singh LK, Pang XZ, Alexacos N, Latourneau R, Theoharides TC 1999 Acute immobilization stress triggers skin mast cell degranulation via corticotropin releasing hormone, neurotensin and substance P: a link to neurologic skin disorders. Brain Behav Immun 13:225–239 204. Koizumi H, Ohkawara A 1999 H2 histamine receptor-mediated increase in intracellular Ca2⫹ in cultured human keratinocytes. J Dermatol Sci 21:127–132 205. Koizumi H, Shimizu T, Nishino H, Ohkawara A 1988 CISurocanic acid attenuates histamine receptor-mediated activation of adenylate cyclase and increase intracellular Ca2⫹. Arch Dermatol Res 290:264 –269 206. Shinoda S, Kameyoshi Y, Hide M, Morita E, Yamamoto S 1998 Histamine enhances UVB-induced IL-6 production by human keratinocytes. Arch Dermatol Res 290:429 – 434 207. Albanesi C, Pastore S, Fanalesbelasio E, Girolomoni G 1998 Cetirizine and hydrocortisone differentially regulate ICAM-1 expression and chemokine release in cultured human keratinocytes. Clin Exp Allergy 28:101–109 208. Yoshida M, Takahashi Y, Inoue S 2000 Histamine induces melanogenesis and morphologic changes by protein kinase A activation via H-2 receptors in human normal melanocytes. J Invest Dermatol 114:334 –342 209. Iizuka H, Adachi K, Halprin KM, Levine V 1976 Adenosine and adenine nucleotide stimulation of skin (epidermal) adenylate cyclase. Biochim Biophys Acta 444:685– 693 210. Pillai S, Bikle DD 1992 Adenosine triphosphate stimulates phsophoinositide metabolism, mobilizes intracellular calcium, and inhibits terminal differentiation of human epidermal keratinocytes. J Clin Invest 90:42–51 211. Cook P, Ashton N, Pittelkow M 1995 Adenosine and adenine nucelotides inhibit the autonomous and epidermal growth factormediated proliferation of cultured human keratinocytes. J Invest Dermatol 104:976 –981 212. Martinez A, Elsasser TH, Muro-Cacho C, Moody TW, Miller MJ, Macri CJ, Cuttitta F 1997 Expression of adrenomedullin and its receptor in normal and malignant human skin: a potential pluripotent in the integument. Endocrinology 138:5597–5604 213. Tu C-L, Oda Y, Bikle D 1999 Effects of a calcium receptor activator

Vol. 21, No. 5

on the cellular response to calcium in human keratinocytes. J Invest Dermatol 113:340 –345 214. Slominski A, Paus R 1994 Towards defining receptors for Ltyrosine and L-DOPA. Mol Cell Endocrinol 99:C7–C11 215. Slominski A, Paus R, Mihm M 1998 Inhibition of melanogenesis as an adjuvant strategy for the treatment of melanotic melanomas. Selective review and hypothesis. Anticancer Res 18:3709 –3716 216. Slominski A, Pruski D 1993 Melatonin inhibits proliferation and melanogenesis in rodent melanoma cells. Exp Cell Res 206:189 –294 217. Slominski A, Chassalerris N, Mazurkiewicz J, Paus R 1994 Murine skin as a target for melatonin bioregulation. Exp Dermatol 3:45– 40 218. Valverde P, Benedito E, Solano F, Oaknin S, Lozano JA, GarciaBorron JC 1995 Melatonin antagonizes ␣-melanocyte stimulating hormone enhancement of melanogenesis in mouse melanoma cells by blocking the hormone-induced accumulation of the c locus tyrosinase. Eur J Biochem 232:257–263 219. Wysolmerski JJ, Vasavada R, Foley J, Weir EC, Burtis WJ, Kukreja SC, Guise TA, Broadus AE, Phillbrick WM 1996 Transactivation of the PTHrP gene in squamous carcinoma predicts the occurrence of hypercalcemia in athymic mice. Cancer Res 56:1043–1049 220. Atillasoy EJ, Burtis WJ, Milstone LM 1991 Immunohistochemical localization of parathyroid hormone-related protein (PTHRP) in normal human skin. J Invest Dermatol 96:277–280 221. Kageshita T, Matsui T, Hirai S, Fukuda Y, Ono T 1999 Hypercalcemia in melanoma patients associated with increase levels of parathyroid hormone-related protein. Melanoma Res 9:69 –73 222. Yeung SCJ, Eton O, Burton DW, Deftos LJ, Vassilopoulousellin R, Gagel RF 1998 Hypercalcemia due to parathyroid hormonerelated protein secretion by melanoma. Horm Res 49:288 –291 223. Crespo M, Sopena B, Orloff JJ, Cameselle Teijeiro JF, Dann P, Andrade MA, Freire M, de la Fuente J, Martinez-Vazquez C 1999 Immunohistochemical detection of parathyroid hormone-related protein in a cutaneous squamous cell carcinoma causing hypercalcemia of malignancy. Arch Pathol Lab Med 123:725–730 224. Bamberger CM, Wald CM, Bamberger AM, Ergun S, Beil FU, Schulte HM 1998 Human lymphocytes produce urocortin, but not corticotropin-releasing hormone. J Clin Endocrinol Metab 83:708 –711 225. Brouxhon SM, Prasad SA, Joseph SA, Felten DL, Bellinger DL 1998 Localization of corticotropin-releasing factor in primary and secondary lymphoid organs of the rat. Brain Behav Immun 12: 107–122 226. Slominski AJ, Wortsman AJ, Mazurkiewicz JE, Matsuoka L, Dietrich J, Lawrence K, Gorbini A, Paus R 1993 Detection of the proopiomelanocortin-derived antigens in normal and pathologic human skin. J Lab Clin Med 122:658 – 666 227. Nagahama M, Funasaka Y, Fernandez-Frez ML, Ohaski A, Chakraborty AK, Ueda M, Ichihashi M 1998 Immunoreactivity of ␣-melanocyte-stimulating hormone, adrenocorticotrophic hormone and ␤-endorphin in cutaneous malignant melanoma and benign melanocytic naevi. Br J Dermatol 138:981–985 228. Wakamatsu KA, Graham A, Cook D, Thody AJ 1997 Characterization of ACTH peptides in human skin and their activation of melanocortin-1 receptor. Pigment Cell Res 10:288 –297 229. Slominski A 1998 Identification of ␤-endorphin, ␣-MSH and ACTH peptides in cultured human melanocytes, melanoma and squamous cell carcinoma cells by RP-HPLC. Exp Dermatol 7: 213–216 230. Wintzen M, Yaar M, Burback JP, Gilchrest BA 1996 Proopiomelanocortin gene product regulation in keratinocytes. J Invest Dermatol 106:673– 678 231. Teofoli P, Motoki K, Lotti TM, Uitto J, Mauviel A 1997 Proopiomelanocortin (POMC) gene expression by normal skin and keloid fibroblasts in culture: modulation by cytokines. Exp Dermatol 6:111–115 232. Johansson O, Ljungberg A, Han SW, Vaalasti A 1991 Evidence for ␥-melanocyte stimulating hormone containing nerves and neutrophilic granulocytes in the human skin by indirect immunofluorescence. J Invest Dermatol 96:852– 856 232a.Mazurkiewicz JE, Corliss D, Slominski A 2000 Spatiotemporal expression, distribution, and processing of POMC and POMCderived peptides in murine skin. J Histochem Cytochem 48:905–914

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

233. Can G, Abdel-Malek Porter-Gill PA, Gill P, Boyce S, Graboqski GA, Nordlund J, Farooqui J 1998 Identification and sequencing of a putative variant of proopiomelanocortin in human epidermis and epidermal cells in culture. J Invest Dermatol 111:485– 491 234. Ancans J, Thody AJ, Wood JM, Beazley WD, Schallreuter KU 1999 Human epidermal proopiomelanocortin (POMC) cDNA variant is identical to mouse POMC cDNA. J Invest Dermatol 112:516 –517 235. Richards RG, Hartman SM 1996 Human dermal fibroblast cells express prolactin in vitro. J Invest Dermatol 106:1250 –1255 236. Chapitis J, Betz LM, Brumsted JR, Gibson M, Kuslis ST, GeorgeMcDaniel H, Riddick DH 1989 Observation of production of immunoactive prolactin by normal human connective tissue in cell culture. In Vitro Cell Dev Biol 25:564 –570 237. Robertseon MT, Alho HR, Martin AA 1989 Localization of prolactin-like immunoreactivity in grafted human sweat glands. J Histochem Cytochem 37:625– 628 238. Walker AM, Robertson MT, Jones CJ 1989 Distribution of a prolactin-like material in human eccrine sweat glands. J Invest Dermatol 93:50 –53 239. Palmetshofer A, Zechner D, Luger TA, Barta A 1995 Splicing variants of the human growth hormone mRNA: detection of pituitary, mononuclear cells and dermal fibroblasts. Mol Cell Endocrinol 113:225–234 240. Clapp C, Lopez-Gomez FJ, Nava G, Corbacho A, Torner L, Macotela Y, Duenas Z, Ochoa A, Noris G, Acosta E, Garay E, Martinez de la Escalara G 1998 Expression of prolactin mRNA and of prolactin-like proteins in endothelial cells: evidence for autocrine effects. J Endocrinol 158:137–144 241. Clevenger CV, Freier DO, Kline JB 1998 Prolactin receptor signal transduction in cells of the immune system. J Endocrinol 157: 187–197 242. Wu H, Devi R, Malarkey WB 1996 Expression and localization of prolactin mRNA in the human immune system. Endocrinology 137:349 –353 243. Wu H, Devi R, Malarkey WB 1996 Localization of growth hormone messenger RNA in the human immune system. J Clin Endocrinol Metab 81:1278 –1282 243a.Slominski A, Malarkey WB, Wortsman J, Asa S, Carlson A, Human skin expresses growth hormone (GH), but not the prolactin (PRL) gene. J Lab Clin Med, in press 244. Nissen JB, Lund M, Stengaard-Pedersens K, Kragballe K 1997 Enkephalin-like immunoreactivity in human skin is found selectively in fractions of CD68 positive dermal cells: increase of enkephalin positive cells in lesional psoriasis. Arch Dermatol Res 289:265–271 245. Nissen JB, Avarch WW, Hansen ES, Stengaard-Pedersens K, Kragballe K 1998 Increased levels of enkephalin following natural sunlight (combined with salt water bathing at the Dead Sea) and ultraviolet A irradiation. Br J Dermatol 139:1012–1019 246. Cheng Chew SB, Leung PY 1991 Immunocytochemical evidence of a met-enkephalin-like substance in the dense-core granules of mouse Merkel cells. Cell Tissue Res 265:611– 614 247. Vega JA, Hernandez LC, Del Valle ME, Dobovy P, Bengoechea ME, Perez-Casas A 1990 Localization of met-enkephalin like immunoreactivity in the glabrous skin of the cat rhinarium. Eur J Morphol 28:69 –78 248. Keshet E, Polakiewicz RD, Itin A, Ornoy A, Rosen H 1989 Proenkephalin A is expressed in mesodermal lineages during organogenesis. EMBO J 8:2917–2923 249. Polakiewicz RD, Behar OZ, Comb MJ, Rosen H 1992 Regulation of proenkephalin expression in cultured skin mesenchymal cells. Mol Endocrinol 6:399 – 407 250. Nissen JB, Avrach WW, Hansen ES, Stengaard-Pedersen K, Kragbelle K 1999 Decrease in enkephalin levels in psoriatic lesions after calcipotriol and mometasone furoate treatment. Dermatology 198: 11–17 251. Carr DJJ 1991 The role of endogenous opioids and their receptors in the immune system. Proc Soc Exp Biol Med 198:710 –720 252. Weihe E, McKnight AT, Corbett AD, Hartschuh Reinercker M, Kosterlitz HW 1983 Characterization of opioid peptides in guinea pig heart and skin. Life Sci [Suppl 1)]33:711–714 253. Wallengren J, Ekman R, Sundler F 1987 Occurrence and distribution of neuropeptides in the human skin. An immunocytochem-

254.

255. 256.

257.

258. 259. 260. 261.

262.

263. 264. 265. 266. 267. 268.

269.

270.

271. 272. 273.

274.

275.

485

ical study on normal skin and blister fluid from inflamed skin. Acta Derm Venereol 67:185–192 Johansson O, Liu P-Y, Han S-W, Lindberger M, Ljungdahl A, Wahl-Gren C-F 1995 An immunohistochemical study of neuroactive substances in the skin of atopic dermatitis. Eur J Dermatol 5:516 –523 Fantani F, Johansson O 1995 Neurochemical markers in human cutaneous Merkel cells. An immunocytochemical investigation. Exp Dermatol 4:365–371 DiMarco E, Marchisio PC, Bondaza S, Franzi AT, Cancedda R, DeLuca M 1991 Growth-regulated synthesis and secretion of biologically active nerve growth factor by human kerantinocytes. J Biol Chem 266:21718 –21722 Hattori A, Iwaski S, Murase K, Tsujimoto M, Sate M, Hayashi K, Kohno M 1994 Tumor necrosis factor is markedly synergistic with interleukin 1 and interferon-␥ in stimulating the production of nerve growth factor in fibroblasts. FEBS Lett 340:177–180 Vos P, Stark F, Pittman RN 1991 Merkel cells in vitro production of nerve growth factor and selective interactions with sensory neurons. Dev Biol 144:281–300 Grando S, Kist DA, Qi M, Dhal MV 1993 human keratinocytes synthesize, secrete, and degrade acetylcholine. J Invest Dermatol 101:32–36 Iyengar B 1989 Modulation of melanocytic activity by acetylcholine. Acta Anat (Basel) 136:139 –141 Schallreuter KU, Wood JM, Ziegler I, Lemke KR, Pittelkow MR, Lindsey NJ, Gutlich 1994 Defective tetrahydrobiopterin and catecholamine biosynthesis in the depigmentation disorder vitiligo. Biochim Biophys Acta 1226:181–192 Chang YT, Mues G, Pittelkow MR, Hyland K 1996 Cultured human keratinocytes as a peripheral source of mRNA for tyrosine hydroxylase and aromatic L-amino acid decarboxylase. J Inherit Metab Dis 19:239 –242 Chang YT, Hyland K, Mues G, Marsh JL 1997 Human hair follicles as a peripheral source of tyrosine hydroxylase and aromatic Lamino acid decarboxylase mRNA. Neurosci Lett 222:210 –212 Johansson O, Olsson A, Enhamre A, Hammar H, Goldstein M 1987 Phenylethanolamine N-methyltransferase-like imunoreactivity in psoriasis. Acta Derm Venereol 67:1–7 Freed WJ, Adinolfi AM, Laskin JD, Geller HM 1989 Transplantation of B16/C3 melanoma cells into the brains of rats and mice. Brain Res 486:349 –362 McEwan M, Parsons PG 1987 Inhibition of melanization in human melanoma cells by a serotonin uptake inhibitor. J Invest Dermatol 89:82– 86 Iyengar B, Misra RS 1988 Neural differentiation of melanocytes in vitiliginous skin. Acta Anat (Basel) 133:62– 65 Schallreuter KU, Wood JM, Pittelkow MR, Buttner G, Swanson N, Korner C, Ehrke C 1996 Increased monoamine oxidase a activity in the epidermis of patients with vitiligo. Arch Dermatol Res 288: 14 –18 Smit NPM, Pavel S, Kammeyer A, Westerhof W 1990 Determination of catechol O-methyltransferase activity in relation to melanin metabolism using high performance liquid chromatography with flourimetric detection. Anal Biochem 190:286 –291 Schallreuter KU, Wood JM 1999 The importance of L-phenylalanine transport and its autocrine turnover to L-tyrosine for melanogenesis in human epidermal melanocytes. Biochem Biophys Res Commun 262:423– 428 Musso NR, Brenci S, Indiveri F, Lotti G 1996 L-tyrosine and nicotine induce synthesis of L-dopa and norepinephrine in human lymphocytes. J Neuroimmunol 74:117–120 Misu Y, Goshima Y, Ueda H, Okamura H 1996 Neurobiology of L-DOPAergic systems. Prog Neurobiol 49:415– 454 Slominski A, Moellmann G, Kuklinska E 1989 L-tyrosine, L-dopa and tyrosinase as positive regulators of the subcellular apparatus of melanogenesis in Bomirski Ab amelanotic melanoma. Pigment Cell Res 2:109 –116 Johansson O, Lie P-Y, Bondesson L, Nordlind K, Olsson MJ, Lontz W, Verhofstad A, Liang Y, Gangi S 1998 A serotonin-like immunoreactivity is present in human cutaneous melanocytes. J Invest Dermatol 111:1010 –1014 Garcia-Caballero T, Gallego R, Roson E, Basanta D, Morel G,

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

486

276.

277. 278.

279. 280.

281. 282. 283. 284. 285. 286. 287.

288.

289.

290.

291. 292.

293.

294.

295.

SLOMINSKI AND WORTSMAN Beiras A 1989 Localization of serotonin-like immunoreactivity in the Merkel cells of pig snout skin. Anat Rec 225:267–271 Gaudet S, Slominski A, Etminan M, Pruski D, Paus R, Namboodiri MAA 1993 Identification and characterization of two isozymic forms of arylamine N-acetyltransferase in Syrian hamster skin. J Invest Dermatol 101:650 – 656 Debiecrychter M, Land SJ, King CM 1996 Histological localization of messenger RNAs for rat acetyltransferases that acetylate serotonin and genotoxic arylamines. Cancer Res 56:1517–1525 Slominski A, Baker J, Rosano T, Guisti LW, Ermak G, Grande M, Gaudet SJ 1996 Metabolism of serotonin to N-acetylserotonin, melatonin and 5-hydroxytryptamine in hamster skin culture. J Biol Chem 271:12281–12286 Nordlind K, Johansson O, Liden S, Hokfelt T 1993 Glutamate- and aspartate-like immunoreactivities in human normal and inflamed skin. Virchow Arch B Cell Pathol 64:75– 82 Kaplan MM, Pan C, Gordon PR, Lee J-K, Gilchrest BA 1988 Human epidermal keratinocytes in culture convert thyroxine to 3,5,3⬘-triidothyronine by type II iodothyronine deiodination. A novel endocrine function of the skin. J Clin Endocrinol Metab 66:815– 822 Huang T, Chopra IJ, Beredo A, Solomon DH, Chua Teco GN 1985 Skin is an active site of inner ring monodeiodiation of thyroxine to 3,3⬘,5⬘-triiodothyronine. Endocrinology 117:2106 –2113 Kaplan MM, Gordon PR, Pan C, Lee J-K, Gilchrest BA 1988 Keratinocytes convert thyroxine to triidothyronine. Ann NY Acad Sci 548:56 – 65 Croteau W, Whittemore SL, Schneider MJ, St Germain DL 1995 Cloning and expression of a cDNA for a mammalian type III iodothyronine deiodinase. J Biol Chem 270:16569 –16575 Bates JM, St Germain DL, Galton VA 1999 Expression profiles of the three iodothyronine deiodinases, D1, D2, D3, in the developing rat. Endocrinology 140:844 – 851 St Germain DL 1999 Development effects of thyroid hormone: the role of deiodinases in regulatory control. Biochem Soc Trans 27: 83– 88 Milewich L, Shaw CB, Sontheimer RD 1988 Steroid metabolism by epidermal keratinocytes. Ann NY Acad Sci 548:66 – 84 Couet J, Martel C, Labrie Y, Luo SQ, Simard J, Labrie F 1994 Opposite effects of prolactin and corticosterone on the expression and activity of 3-␤-hydroxysteroid dehydrogenase/␦5 – ␦ 4 isomerase in rat skin. J Invest Dermatol 103:60 – 64 Dumont M, Luu-The V, Dupont E, Pelletier G, Labrie F 1992 Characterization, expression, and immunohistochemical localization of 3␤-hydroxysteroid dehydrogenase/␦ 5 – ␦ 4 isomerase in human skin. J Invest Dermatol 99:415– 421 Ando Y, Yamaguchi Y, Hamada K, Yoshikawa K, Itami S 1999 Expression of mRNA for androgen receptor, 5␣-reductase and 17␤hydroxysteroid dehydrogenase in human dermal papilla cells. Br J Dermatol 141:840 – 845 Thiboutot D, Knaggs H, Gilliland K, Lin G 1998 Activity of 5-␣-reductase and 17-␤-hydroxysteroid dehdrogenase in the infrainfundibulum of subjects with and without acne vulgaris. Dermatology 196:38 – 42 Eicheler W, Happle R, Hoffmann R 1998 5-␣-Reductase activity in the human hair follicle concentrates in the dermal papilla. Arch Dermatol Res 290:126 –132 Chen W, Zouboulis CC, Fritsch M, Blume-Peytavi U, Kodelja V, Goerdt S, Luu-The V, Orfanos CE 1998 Evidence of heterogeneity and quantitative difference of the type 1 5␣-reductase expression in cultured human skin cells-evidence of its presence in melanocytes. J Invest Dermatol 110:84 – 89 Thiboutout D, Harris G, Iles V, Climis G, Gilliland K, Hagari S 1995 Activity of the Type I 5␣-reductase exhibits regional differences in isolated sebaceous glands and whole skin. J Invest Dermatol 105:209 –214 Wahe M, Antonipillai I, Horton R 1993 Effects of transforming growth factor ␤ and epidermal growth factor on steroid 5 ␣reductase activity in genital skin fibroblasts. Mol Cell Endocrinol 98:55–59 Sato T, Sonoda T, Itami S, Takayasu 1998 Predominance of type I 5␣-reductase in apocrine sweat glands of patients with excessive

296.

297. 298.

299. 300.

301. 302. 303. 304. 305.

306.

307.

308. 309. 310. 311. 312. 313. 314. 315.

316. 317.

Vol. 21, No. 5

of abnormal odour derived from apocrine sweat (osmidrosis). Br J Dermatol 139:806 – 810 Bayne EK, Flanagan J, Einstein M, Ayala J, Chang B, Azzolina B, Whiting DA, Mumford RA, Thiboutot D, Singer LI, Harris G 1999 Immunohistochemical localization of types 1 and 2 5␣-reductase in human scalp. Br J Dermatol 141:481– 491 Hsia SL 1971 Potential in exploring the biochemistry of human skin. Essays Biochem 7:1–38 Slominski A, Gomez-Sanchez Celso Foecking MF, Wortsman J 1999 Metabolism of progesterone to DOC, corticosterone and 18OHDOC in cultured human melanoma cells. FEBS Lett 445: 364 –366 Slominski A, Gomez-Sanchez Celso Foecking MF, Wortsman J 2000 Active steroidogenesis in the normal rat skin. Biochim Biophys Acta 1474:1– 4 Botchkarev VA, Peters EMJ, Botchkareva NV, Maurer M, Paus P 1999 Hair cycle-dependent changes in adrenergic skin innervation, and hair growth modulation by adrenergic drugs. J Invest Dermatol 113:878 – 887 Bikle DD, Pillai S 1988 Vitamin D metabolite production and function in keratinocytes. Ann NY Acad Sci 548:27– 44 Strewler GJ 2000 The physiology of parathyroid hormone-related protein. N Engl J Med 342:177–185 Mohammad T, Morrison H, HogenEsch H 1999 Urocanic acid photochemistry and photobiology. Photochem Photobiol 69: 115–135 Webber LJ, Whang E, DeFabo EC 1997 The effects of UVA-I (340 – 400 nm), UVA-II (320 –340 nm) and UVA-I⫹II on the photoisomerization of urocanic acid in vitro. Photochem Photobiol 66:484 – 492 Johansson O, Fantini F, Hu H 1999 Neuronal structural protein, transmitters, transmitter enzymes and neuropeptides in human Meissner’s corpuscles: a reappraisal using immunohistochemistry. Arch Dermatol 291:419 – 424 Botchkarev VA, Eichenmu¨eller S, Peters EM, Pietsch P, Johansson O, Maurer M, Paus R 1997 A simple immnoflouresecence technique for simultaneous visualization of mast cells and nerve fibers reveals selectivity and hair cycle-dependent changes in mast –nerve fiber contacts in murine skin. Arch Dermatol Res 289:292–302 Egan CL, Viglione-Schneck MJ, Walsh LJ, Green B, Trojanowski JQ, Whitaker-Menezes D, Murphy GF 1998 Characterization of unmyeliniated axons uniting epidermal and dermal immune cells in primate and murine skin. J Cutan Pathol 25:20 –29 Pasche F, Merot Y, Carraux P, Saurat J-H 1990 Relationship between Merkel cells and nerve endings during embryogenesis in the mouse epidermis. J Invest Dermatol 95:247–251 Hilliges M, Wang L, Johnasson O 1995 Ultrastructural evidence for nerve fibers within all vital layers of the human epidermis. J Invest Dermatol 104:134 –137 Kennedy WR, Wendelschafer-Crabb G 1993 The innervation of human epidermis. J Neurol Sci 115:184 –190 Botchkarev VA, Eichenmu¨eller S, Johansson O, Paus R 1997 Hair cycle-dependent plasticity of skin hair follicle innervation in normal skin. J Comp Neurol 386:379 –395 Johansson O, Virtanen M, Hilleges M 1995 Histaminergic nerves demonstrated in the skin. A new direct mode of neurogenic inflammation? Exp Dermatol 4:93–96 Hsieh S-T, Lin W-M 1999 Modulation of keratinocyte proliferation by skin innervation. J Invest Dermatol 113:579 –586 Bruch-Gerharz D, Ruzicka T, Kolb-Bachofen V 1998 Nitric oxide in human skin; current status and future prospects. J Invest Dermatol 110:1–7 Mor A, Chartel N, Vaudry H, Nicolas P 1994 Skin peptide tyrosinetyrosine, a member of the pancreatic polypeptide family: isolation, structure, synthesis, and endocrine activity. Proc Natl Acad Sci USA 91:10295–10299 Vaudry H, Chartrel N, Desrues L, Galas L, Kikuyama S, Mor A, Nicolas P, Tonon MC 1999 The pituitary-skin connection in amphibians. Ann NY Acad Sci 885:41–56 Bornstein SR, Chrousos GP 1999 Adrenocorticotropin (ACTH)and non-ACTH-mediated regulation of the adrenal cortex: neural and immune imputs. J Clin Endocrinol Metab 84:1729 –1736

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

October, 2000

NEUROENDOCRINOLOGY OF THE SKIN

318. Ito S 1992 Melanin-related metabolites as markers of melanoma: a review. J Dermatol 19:802– 805 319. Langhans W, Hrupka B 1999 Interleukins and tumor necrosis factor as inhibitors of food intake. Neuropeptides 33:415– 424 320. Sewter CP, Digby JE, Blows F, Prins J, O’Rahilly S 1999 Regulation of tumour necrosis factor-␣ release from human adipose tissue in vitro. J Endocrinol 163:33–38 321. Gaykema RPA, Dijkstra I, Tilders FJH 1995 Subdiaphragmatic vagotomy suppresses endotoxin-induced activation of hypothalmic corticotropin-releasing hormone neurons and ACTH secretion. Endocrinology 136:4717– 4721 322. Slominski A, Baker J, Ermak G, Chakraborty A, Pawelek J 1996 UVB stimulates production of corticotropin releasing factor (CRF) by human melanocytes. FEBS Lett 399:175–176 323. Chakraborty AK, Funasaka Y, Slominski A, Bolognia J, Sodi S, Ichihashi M, Pawelek JM 1999 UV light and MSH receptors. Ann NY Acad Sci 885:41–56; 885:100 –116 324. Belon PE 1985 UVA exposure and pituitary secretion: variations of human lipotropin concentrations (␤LPH) after UVA exposure. Photochem Photobiol 42:327–329 325. Bolognia J, Murray M, Pawelek J 1989 UVB-induced melanogenesis may be mediated through the MSH-receptor system. J Invest Dermatol 92:651– 656 326. Duthie MS, Kimber I, Norval M 1999 The effects of ultraviolet radiation on the human immune system. Br J Dermatol 140:995– 1009 327. Kondo S 1999 The roles of keratinocyte-derived cytokines in the epidermis and their possible response to UVA-irradiation. J Invest Dermatol Symp Proc 4:177–183 328. Ko¨ck A, Schwarz T, Kirnbauer R, Urbanski A, Perry P, Ansel JC, Luger TA 1990 Human keratinocytes are a source for tumor necrosis factor ␣ evidence for synthesis and release upon stimulation with endotoxin or ultraviolet light. J Exp Med 172:1609 –1614 329. Grewe M, Gyufko K, Krutmann J 1995 Interleukin-10 production by cultured human keratinocytes: regulation by ultraviolet B and ultraviolet A1 radiation. J Invest Dermatol 104:3– 6 330. Skov L, Hansen H, Allen M, Villadsen L, Norval M, Barker JNWN, Simon J, Baadsgaard O 1998 Contrasting effects of ultraviolet A1 and ultraviolet B exposure on the induction of tumour necrosis factor-␣ in human skin. Br J Dermatol 138:216 –220 331. Shimizu T, Abe R, Ohkawars A, Nishihira J 1999 Ultraviolet B radiation upregulates the production of macrophage migration inhibitory factor (MIF) in human epidermal keratinocytes. J Invest Dermatol 112:210 –215 332. Werth VP, Zhang W 1999 Wavelength-specific synergy between ultraviolet radiation and interleukin-1 ␣ in the regulation of matrixrelated genes: mechanistic role for tumor necrosis factor-␣. J Invest Dermatol 113:196 –201 333. Scholzen T, Hartmeyer M, Fastrich M, Brzoska T, Becher E, Schwarz T, Luger TA 1998 Ultraviolet light and interleukin-10 modulate expression of cytokines by transformed human dermal microvascular endothelial cells (HMEC-1). J Invest Dermatol 111: 50 –56 334. Schallreuter KU, Wood JM, Korner C, Harle KM, Schulzdouglas V, Werner ER 1998 6-tetrahydrobiopterin functions as a UVB-light switch for de novo melanogenesis. Biochim Biophys Acta 1382: 339 –344 335. Schallreuter KU, Schulzdouglas V, Bunz A, Beazley W, Korner C 1997 Pteridines in the control of pigmentation. J Invest Dermatol 109:31–35 336. Teofoli P, Frezzolini A, Puddu P, DePita O, Mauviel A, Lotti T 1999 The role of proopiomelanocortin-derived peptides in skin fibroblast and mast cell functions. Ann NY Acad Sci 885:268 –276 337. Olerud JE, Usi ML, Seckin D, Chiu DS, Haycox CL, Song I-S, Ansel JC, Bunnett NW 1999 Neutral endopeptidase expression and distribution in human skin and wounds. J Invest Dermatol 112: 873– 881 338. Haddad JG, Matsuoka LY, Hollis BW, Hu YZ, Wortsman J 1993 Human plasma transport of vitamin D3 after its endogenous synthesis. J Clin Invest 91:2552–2555

487

339. Tian XQ, Holick MF 1999 A liposomal model that mimics the cutaneous production of vitamin D3. J Biol Chem 274:4174 – 4179 340. Pettifor JM, Moodley GP, Hough FS, Koch H, Chen T, Lu Z, Holick MF 1996 The effect of season and latitude on in vitro vitamin D formation by sunlight in South Africa. S Afr Med J 86:1270 –1272 341. Matsuoka LY, Wortsman J, Dannenberg MJ, Hollis BW, Lu Z, Holick MF 1992 Clothing prevents ultraviolet-B radiation dependent photosynthesis of vitamin D3. J Clin Endocrinol Metab 75: 1099 –1103 342. Matsuoka LY, Ide L, Wortsman J, MacLaughlin JA, Holick MF 1987 Sunscreens suppress cutaneous vitamin D3 synthesis. J Clin Endocrinol Metab 64:1165–1168 343. Matsuoka LY, Wortsman J, Hanifan N, Holick MF 1988 Chronic sunscreen use decreases circulating concentrations of 25-hydroxyvitamin D. Arch Dermatol 124:1802–1804 344. Farrerons J, Barnadas M, Rodriguez J, Renau A, Yoldi B, LopezNavidad A, Moragas J 1998 Clinically prescribed sunscreen (sun protection factor 15) does not decrease serum vitamin D concentration sufficiently either to induce changes in parathyroid function or in metabolic markers. Br J Dermatol 139:422– 427 345. Matsuoka LY, Wortsman J, Hollis BW 1990 Use of topical sunscreen for the evaluation of regional synthesis of vitamin D3. J Am Acad Dermatol 22:772–775 346. Anderson RR, Parrish JA 1981 The optics of human skin. J Invest Dermatol 77:13–19 347. Matsuoka LY, Wortsman J, Haddad JG, Kolm P, Hollis BW 1991 Racial pigmentation and the cutaneous synthesis of vitamin D3. Arch Dermatol 127:536 –538 348. Matsuoka LY, Wortsman J, Chen TC, Holick MF 1995 Compensation for the interracial variance in the cutaneous synthesis of vitamin D3. J Lab Clin Med 126:452– 457 349. Awumey EMK, Mitra DA, Hollis BW, Kumar R, Bell NH 1998 Vitamin D metabolism is altered in Asian Indians in the southern United States: a clinical research center study. J Clin Endocrinol Metab 83:169 –173 350. Matsuoka LY, Wortsman J, Haddad JG, Hollis BW 1989 In vivo threshold for cutaneous synthesis of vitamin D3. J Lab Clin Med 114:301–305 351. Kamboh MI, Ferrell RE 1986 Ethnic variation in vitamin D binding protein (GC): a review of isoelectric focusing studies in human populations. Hum Genet 72:281–293 352. Matsuoka LY, Wortsman J, Hollis BW 1990 Suntanning and cutaneous synthesis of vitamin D3. J Lab Clin Med 116:87–90 353. MacLaughlin J, Holick MF 1985 Aging decreases the capacity of human skin to produce vitamin D3. J Clin Invest 76:1536 –1538 354. Holick MF, Matsuoka LY, Wortsman J 1989 Age, vitamin D, and solar ultraviolet. Lancet 2:1104 –1105 355. Matsuoka LY, Wortsman J, Haddad JG, Hollis BW 1990 Cutaneous formation of vitamin D in psoriasis. Arch Dermatol 126:1107– 1108 356. Matsuoka LY, Dannenberg MJ, Wortsman J, Hollis BW, Jimenez SA, Varga J 1991 Cutaneous vitamin D3 formation in progressive systemic sclerosis. J Rheumatol 18:1196 –1198 357. Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF 2000 Decreased bioavailablity of vitmain D in obesity. Am J Clin Nutr 72:690 – 693 358. Calvo M, Enu JM 1989 Relations between vitamin D and fatty acid binding property of vitamin D-binding protein. Biochem Biophys Res Commun 163:14 –17 359. Matsuoka LY, Wortsman J, Haddad JG, Hollis BW 1992 Elevation of blood vitamin D2 levels does not impede the release of vitamin D3 from the skin. Metabolism. 41:1257–1260 360. Matsuoka LY, Wortsman J, Hollis BW 1988 Lack of effect of exogenous calcitriol on the cutaneous production of vitamin D3. Metabolism 66:451– 453 361. Harris SS, Dawson-Hughes B 1998 Seasonal changes in plasma 25-hydroxyvitamin D concentration of young American black and white women. Am J Clin Nutr 67:1232–1236

Downloaded from edrv.endojournals.org at India:Endo Jnls Sponsored on May 22, 2007

Related Documents

Histology Of The Skin
November 2019 29
Assessmnet Of The Skin
April 2020 22
Skin
November 2019 41
Skin
December 2019 43