442
Views
66
CrossRef citations to date
0
Altmetric
Review

Sex steroids and cutaneous wound healing: the contrasting influences of estrogens and androgens

&
Pages 276-288 | Received 10 Nov 2006, Accepted 10 Jan 2007, Published online: 03 Jul 2009

References

  • Martin P. Wound healing – aiming for perfect skin regeneration. Science 1997; 276: 75–81
  • Ashcroft G S, Mills S J. Androgen receptor-mediated inhibition of cutaneous wound healing. J Clin Invest 2002; 110: 615–624
  • Ashcroft G S, Horan M A, Ferguson M WJ. Aging alters inflammatory and endothelial cell adhesion molecule profiles during human cutaneous wound healing. Lab Invest 1998; 78: 47–58
  • Ashcroft G S, Kielty C M, Horan M A, Ferguson M WJ. Age-related changes in the temporal and spatial distributions of fibrillin and elastin mRNAs and proteins in acute cutaneous wounds of healthy humans. J Pathol 1997; 183: 80–89
  • Taylor R J, Taylor A D, Smyth J V. Using an artificial neural network to predict healing times and risk factors for venous leg ulcers. J Wound Care 2002; 11: 101–105
  • Wysocki A B, Staiano-Cioco, Grinnell F. Wound fluid from chronic leg ulcers contains elevated levels of metalloproteinases MMP-2 and MMP-9. J Invest Dermatol 1993; 101: 64–68
  • Nikolaevich K N, Ivanovich S J, Victorovich S S. Major reproduction hormones as regulators of cell-to-cell interactions in humoral immune responses. Brain Behav Immun 1991; 5: 149–161
  • Kocar I H, Yesilova Z, Ozata M, Turan M, Sengul A, Ozdemir I. The effect of testosterone replacement treatment on immunological features of patients with Klinefelter's syndrome. Clin Exp Immunol 2000; 121: 448–452
  • Offner P J, Moore E E, Biffl W L. Male gender is a risk factor for major infections after surgery. Arch Surg 1999; 134: 935–938
  • McCashland T M, Brand R, Lyden E, de Garmo P. Gender differences in colorectal polyps and tumors. Am J Gastroenterol 2001; 96: 882–886
  • Verthelyi D. Sex hormones as immunomodulators in health and disease. Int Immunopharmacol 2001; 1: 983–993
  • D'Agostino P, Milano S, Barbera C, et al. Sex hormones modulate inflammatory mediators produced by macrophages. Ann N Y Acad Sci 1999; 876: 426–429
  • Shuster S, Black M M, McVitie E. The influence of age and sex on skin thickness, skin collagen and density. Br J Dermatol 1975; 93: 639–643
  • Branchet M C, Boisnic S, Frances C, Robert A M. Skin thickness changes in normal aging skin. Gerontology 1990; 36: 28–35
  • Brincat M, Kabalan S, Studd J W, Moniz C F, de Trafford J, Montgomery J. A study of the decrease of skin collagen content, skin thickness, and bone mass in the postmenopausal woman. Obstet Gynecol 1987; 70: 840–845
  • Brincat M, Moniz C J, Studd J W, et al. Long-term effects of the menopause and sex hormones on skin thickness. Br J Obstet Gynaecol 1985; 92: 256–259
  • Gilchrest B A. In vitro assessment of keratinocyte aging. J Invest Dermatol 1983; 81: 184–19S
  • West M D, Pereira-Smith O M, Smith J R. Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Exp Cell Res 1989; 184: 138–147
  • El-Domyati M, Attia S, Saleh F, et al. Intrinsic aging vs. photoaging: a comparative histopathological, immunohistochemical, and ultrastructural study of skin. Exp Dermatol 2002; 11: 398–405
  • Jacobsen E, Billings J K, Frantz R A, Kinney C K, Stewart M E, Downing D T. Age-related changes in sebaceous wax ester secretion rates in men and women. J Invest Dermatol 1985; 85: 483–485
  • Braverman I M, Fonferko E. Studies in cutaneous aging. II. The microvasculature. J Invest Dermatol 1982; 78: 444–448
  • Herrick S, Ashcroft G, Ireland G, Horan M, McCollum C, Ferguson M. Up-regulation of elastase in acute wounds of healthy aged humans and chronic venous leg ulcers are associated with matrix degradation. Lab Invest 1997; 77: 281–288
  • Ashcroft G S, Horan M A, Herrick S H, Tarnuzzer R W, Schultz G S, Ferguson M WJ. Age-related differences in the temporal and spatial regulation of matrix metalloproteinases (MMPs) in normal skin and acute cutaneous wounds of healthy humans. Cell Tissue Res 1997; 290: 581–591
  • Ashcroft G S, Herrick S E, Tarnuzzer R W, Horan M A, Schultz G S, Ferguson M WJ. Human ageing impairs injury-induced in vivo expression of tissue inhibitor of matrix metalloproteinases (TIMP)-1 and -2 proteins and mRNA. J Pathol 1997; 183: 169–176
  • Swift M E, Burns A L, Gray K L, DiPietro L A. Age-related alterations in the inflammatory response to dermal injury. J Invest Dermatol 2001; 117: 1027–1035
  • Holt D R, Kirk S J, Regan M C, Hurson M, Lindblad W J, Barbul A. Effect of age on wound healing in healthy human beings. Surgery 1992; 112: 293–297
  • Ashcroft G S, Horan M A, Ferguson M W. The effects of ageing on wound healing: immunolocalisation of growth factors and their receptors in a murine incisional model. J Anat 1997; 190: 351–365
  • Ashcroft G S, Horan M A, Ferguson M W. Aging is associated with reduced deposition of specific extracellular matrix components, an upregulation of angiogenesis, and an altered inflammatory response in a murine incisional wound healing model. J Invest Dermatol 1997; 108: 430–437
  • Swift M E, Kleinman H K, DiPietro L A. Impaired wound repair and delayed angiogenesis in aged mice. Lab Invest 1999; 79: 1479–1487
  • Sandblom P H, Peterson P, Muren A. Determination of the tensile strength of the healing wound as a clinical test. Acta Chirug Scand 1953; 105: 252–257
  • Lindstedt E, Sandblom P. Wound healing in man: tensile strength of healing wounds in some patient groups. Ann Surg 1975; 181: 842–846
  • Barrett-Connor E. Epidemiology and the menopause: a global overview. Int J Fertil Menopausal Stud 1993; 38: 6–14
  • Chakravarti S, Collins W P, Forecast J D, Newton J R, Oram D H, Studd J W. Hormonal profiles after the menopause. Br Med J 1976; 2: 784–787
  • Vermeulen A. The hormonal activity of the postmenopausal ovary. J Clin Endocrinol Metab 1976; 42: 247–253
  • Endoh A, Kristiansen S B, Casson P R, Buster J E, Hornsby P J. The zona reticularis is the site of biosynthesis of dehydroepiandrosterone and dehydroepiandrosterone sulfate in the adult human adrenal cortex resulting from its low expression of 3β-hydroxysteroid dehydrogenase. J Clin Endocrinol Metab 1996; 81: 3558–3565
  • Longcope C. Adrenal and gonadal androgen secretion in normal females. Clin Endocrinol Metab 1986; 15: 213–228
  • Labrie F, Belanger A, Cusan L, Gomez J L, Candas B. Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. J Clin Endocrinol Metab 1997; 82: 2396–2402
  • Morley J E, Kaiser F E, Perry H M, 3rd, et al. Longitudinal changes in testosterone, luteinizing hormone, and follicle-stimulating hormone in healthy older men. Metabolism 1997; 46: 410–413
  • Abraham G E. Ovarian and adrenal contribution to peripheral androgens during the menstrual cycle. J Clin Endocrinol Metab 1974; 39: 340–346
  • Davison S, Bell R, Donath S, Montalto J, Davis S. Androgen levels in adult females: changes with age, menopause and oophorectomy. J Clin Endocrinol Metab 2005; 90: 3847–3853
  • Burger H G, Dudley E C, Cui J, Dennerstein L, Hopper J L. A prospective longitudinal study of serum testosterone, dehydroepiandrosterone sulfate, and sex hormone-binding globulin levels through the menopause transition. J Clin Endocrinol Metab 2000; 85: 2832–2838
  • Jiroutek M R, Chen M H, Johnston C C, Longcope C. Changes in reproductive hormones and sex hormone-binding globulin in a group of postmenopausal women measured over 10 years. Menopause 1998; 5: 90–94
  • Labrie F, Luu-The V, Labrie C, Pelletier G, El-Alfy M. Intracrinology and the skin. Horm Res 2000; 54: 218–229
  • Shin M H, Rhie G E, Park C H, et al. Modulation of collagen metabolism by the topical application of dehydroepiandrosterone to human skin. J Invest Dermatol 2005; 124: 315–323
  • Sourla A, Richard V, Labrie F, Labrie C. Exclusive androgenic effect of dehydroepiandrosterone in sebaceous glands of rat skin. J Endocrinol 2000; 166: 455–462
  • Thornton M J, Taylor A H, Mulligan K, et al. The distribution of estrogen receptor β is distinct to that of estrogen receptor α and the androgen receptor in human skin and the pilosebaceous unit. J Invest Dermatol Symp Proc 2003; 8: 100–103
  • Callens A, Vaillant L, Lecomte P, Berson M, Gall Y, Lorette G. Does hormonal skin aging exist? A study of the influence of different hormone therapy regimens on the skin of postmenopausal women using non-invasive measurement techniques. Dermatology 1996; 193: 289–294
  • Conrad F, Ohnemus U, Bodo E, Bettermann A, Paus R. Estrogens and human scalp hair growth – still more questions than answers. J Invest Dermatol 2004; 122: 840–842
  • Schmidt J B, Binder M, Macheiner W, Kainz C, Gitsch G, Bieglmayer C. Treatment of skin ageing symptoms in perimenopausal females with estrogen compounds. A pilot study. Maturitas 1994; 20: 25–30
  • Maheux R, Naud F, Rioux M, et al. A randomized, double-blind, placebo-controlled study on the effect of conjugated estrogens on skin thickness. Am J Obstet Gynecol 1994; 170: 642–649
  • Varila E, Rantala I, Oikarinen A, et al. The effect of topical oestradiol on skin collagen of postmenopausal women. Br J Obstet Gynaecol 1995; 102: 985–989
  • Henry F, Piérard-Franchimont C, Cauwenbergh G, Piérard G E. Age-related changes in facial skin contours and rheology. J Am Geriatr Soc 1997; 45: 220–222
  • Punnonen R, Vaajalahti P, Teisala K. Local oestriol treatment improves the structure of elastic fibers in the skin of postmenopausal women. Ann Chir Gynaecol Suppl 1987; 202: 39–41
  • Kariya Y, Moriya T, Suzuki T, et al. Sex steroid hormone receptors in human skin appendage and its neoplasms. Endocr J 2005; 52: 317–325
  • Shuster S, Hinks W M, Thody A J. Effect of sex and age at gonadectomy on the sebaceous response to progesterone. J Endocrinol 1977; 73: 67–70
  • Bläuer M, Vaalasti A, Pauli S L, Ylikomi T, Joensuu T, Tuohimaa P. Location of androgen receptor in human skin. J Invest Dermatol 1991; 97: 264–268
  • Choudhry R, Hodgins M B, Van der Kwast T H, Brinkmann A O, Boersma W J. Localization of androgen receptors in human skin by immunohistochemistry: implications for the hormonal regulation of hair growth, sebaceous glands and sweat glands. J Endocrinol 1992; 133: 467–475
  • Liang T, Hoyer S, Yu R, et al. Immunocytochemical localization of androgen receptors in human skin using monoclonal antibodies against the androgen receptor. J Invest Dermatol 1993; 100: 663–666
  • Ebling F JG. Hair follicles and associated glands are androgen targets. J Clin Endocrinol Metab 1986; 15: 319–339
  • Messenger A G. The control of hair growth: an overview. J Invest Dermatol 1993; 101: 4–9S
  • Pochi P E, Strauss J S. Endocrinologic control of the development and activity of the human sebaceous gland. J Invest Dermatol 1974; 62: 191–201
  • Tammi R, Santti R. Morphometric analysis of human epidermis treated with testosterone and dehydroepiandrosterone in organ culture. Arch Dermatol Res 1989; 281: 417–423
  • Black M M, Shuster S, Bottoms E. Osteoporosis, skin collagen and androgen. Br Med J 1970; 4: 773–774
  • O'Connor C A, Cernak I, Vink R. Both estrogen and progesterone attenuate edema formation following diffuse traumatic brain injury in rats. Brain Res 2005; 1062: 171–174
  • Rusa R, Alkayed N J, Crain B J, et al. 17β-Estradiol reduces stroke injury in estrogen-deficient female animals. Stroke 1999; 30: 1665–1670
  • Dubal D B, Zhu H, Yu J, et al. Estrogen receptor α, not β, is a critical link in estradiol-mediated protection against brain injury. Proc Natl Acad Sci USA 2001; 98: 1952–1957
  • Xing D, Miller A, Novak L, Rocha R, Chen Y F, Oparil S. Estradiol and progestins differentially modulate leukocyte infiltration after vascular injury. Circulation 2004; 109: 234–241
  • Ashcroft G S, Dodsworth J, Van Boxtel E, et al. Estrogen accelerates cutaneous wound healing associated with an increase in TGF-β1 levels. Nature Med 1997; 3: 1209–1215
  • Roberts A B, Sporn M B, Assoian R K, et al. Transforming growth factor type β: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci USA 1986; 83: 4167–4172
  • Shah M, Foreman D M, Ferguson M W. Neutralising antibody to TGF-β1,2 reduces cutaneous scarring in adult rodents. J Cell Sci 1994; 107: 1137–1157
  • Ashcroft G S, Greenwell-Wild T, Horan M A, Wahl S M, Ferguson M WJ. Topical estrogen accelerates cutaneous wound healing in aged humans associated with an altered inflammatory response. Am J Pathol 1999; 155: 1137–1146
  • Lundgren D. Influence of estrogen and progesterone on exudation, inflammatory cell migration and granulation tissue formation in preformed cavities. Scand J Plast Reconstr Surg 1973; 7: 10–14
  • Ashcroft G S, Mills S J, Lei K, et al. Estrogen modulates cutaneous wound healing by downregulating macrophage migration inhibitory factor. J Clin Invest 2003; 111: 1309–1318
  • Hardman M J, Waite A, Zeef L, Burow M, Nakayama T, Ashcroft G S. Macrophage migration inhibitory factor. A central regulator of wound healing. Am J Pathol 2005; 167: 1561–1574
  • Ashcroft G S, Mills S J, Flanders K C, et al. Role of Smad3 in the hormonal modulation of in vivo wound healing responses. Wound Repair Regen 2003; 11: 468–473
  • Pirilä E, Ramamurthy N, Maisi P, et al. Wound healing in ovariectomized rats: effects of chemically modified tetracycline (CMT-8) and estrogen on matrix metalloproteinases −8, −13 and type I collagen expression. Curr Med Chem 2001; 8: 281–294
  • Pirilä E, Parikka M, Ramamurthy N S, et al. Chemically modified tetracycline (CMT-8) and estrogen promote wound healing in ovariectomized rats: effects on matrix metalloproteinase-2, membrane type 1 matrix metalloproteinase, and laminin-5 γ2-chain. Wound Repair Regen 2002; 10: 38–51
  • Ashworth J J, Smyth J V, Pendleton N, et al. The dinucleotide (CA) repeat polymorphism of estrogen receptor beta but not the dinucleotide (TA) repeat polymorphism of estrogen receptor alpha is associated with venous ulceration. J Steroid Biochem Mol Biol 2005; 97: 266–270
  • Nyman S, Lindhe J, Zederfeldt B. Influence of estrogen and progesterone on tissue regeneration in oophorectomized rabbits. Acta Chir Scand 1971; 137: 131–139
  • Nyman S, Lindhe J, Zederfeldt B. The vascularity of wounded areas in estradiol and progesterone treated female rabbits. A microangiographic study. Acta Chir Scand 1971; 137: 631–637
  • Lundgren D. Influence of estrogen and progesterone on vascularization of granulation tissue in preformed cavities. A microangiographic study. Scand J Plast Reconstr Surg 1973; 7: 85–90
  • Grady D, Rubin S M, Petitti D B, et al. Hormone therapy to prevent disease and prolong life in postmenopausal women. Ann Intern Med 1992; 117: 1016–1037
  • Roof R L, Duvdevani R, Stein D G. Gender influences outcome of brain injury: progesterone plays a protective role. Brain Res 1993; 607: 333–336
  • Shear D A, Galani R, Hoffman S W, Stein D G. Progesterone protects against necrotic damage and behavioral abnormalities caused by traumatic brain injury. Exp Neurol 2002; 178: 59–67
  • He J, Evans C O, Hoffman S W, Oyesiku N M, Stein D G. Progesterone and allopregnanolone reduce inflammatory cytokines after traumatic brain injury. Exp Neurol 2004; 189: 404–412
  • Kuebler J F, Yokoyama Y, Jarrar D, et al. Administration of progesterone after trauma and hemorrhagic shock prevents hepatocellular injury. Arch Surg 2003; 138: 727–734
  • Suparto I H, Koudy Williams J, Fox J L, Vinten-Johansen J. A comparison of two progestins on myocardial ischemia-reperfusion injury in ovariectomized monkeys receiving estrogen therapy. Coron Artery Dis 2005; 16: 301–308
  • Nyman S, Lindhe J, Zederfeldt B. Impaired wound healing in progesterone-treated rabbits. Acta Chir Scand 1973; 139: 415–419
  • Nyman S, Lindhe J, Zederfeldt B, Solvell L. Vascular permeability in wounded areas of estradiol and progesterone treated female rabbits. Acta Chir Scand 1971; 137: 709–714
  • Mills S J, Ashworth J J, Gilliver S C, Hardman M J, Ashcroft G S. The sex steroid precursor DHEA accelerates cutaneous wound healing via the estrogen receptors. J Invest Dermatol 2005; 125: 1053–1062
  • Padgett D A, Loria R M. Endocrine regulation of murine macrophage function: effects of dehydroepiandrosterone, androstenediol, and androstenetriol. J Neuroimmunol 1998; 84: 61–68
  • Gilliver S C, Wu F, Ashcroft G S. Regulatory roles of androgens in cutaneous wound healing. Thromb Haemost 2003; 90: 978–985
  • van den Beld A W, de Jong F H, Grobbee D E, Pols H A, Lamberts S W. Measures of bioavailable serum testosterone and estradiol and their relationships with muscle strength, bone density, and body composition in elderly men. J Clin Endocrinol Metab 2000; 85: 3276–3282
  • Gilliver S C, Ashworth J J, Mills S J, Hardman M J, Ashcroft G S. Androgens modulate the inflammatory response during acute wound healing. J Cell Sci 2006; 119: 722–732
  • Saika S, Ikeda K, Yamanaka O, et al. Loss of tumor necrosis factor α potentiates transforming growth factor β-mediated pathogenic tissue response during wound healing. Am J Pathol 2006; 168: 1848–1860
  • Ashcroft G S, Yang X, Glick A B, et al. Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response. Nat Cell Biol 1999; 1: 260–6
  • Nitsch S M, Wittmann F, Angele P, et al. Physiological levels of 5α-dihydrotestosterone depress wound immune function and impair wound healing following trauma-hemorrhage. Arch Surg 2004; 139: 157–63
  • Crowley L V, Kriss P, Rettura G, Nakao K, Levenson S M. Effects of testosterone propionate and environmental temperature on nitrogen balance and wound healing of rats with and without femoral fracture. J Trauma 1977; 17: 446–453
  • Rasmussen F. Effect of androgens and anabolic steroids on rabbit urinary bladder and ureter. Biochemical and morphological studies on intact tissues and healing wounds. Acta Endocrinol (Copenh) 1968; 57: 414–426
  • Park K M, Kim J I, Ahn Y, Bonventre A J, Bonventre J V. Testosterone is responsible for enhanced susceptibility of males to ischemic renal injury. J Biol Chem 2004; 279: 52282–52292
  • Fortepiani L A, Yanes L, Zhang H, Racusen L C, Reckelhoff J F. Role of androgens in mediating renal injury in aging SHR. Hypertension 2003; 42: 952–955
  • Sakemi T, Toyoshima H, Morito F. Testosterone eliminates the attenuating effect of castration on the progressive glomerular injury in hypercholesterolemic male Imai rats. Nephron 1994; 67: 469–476
  • Brown T J, Khan T, Jones K J. Androgen induced acceleration of functional recovery after rat sciatic nerve injury. Restor Neurol Neurosci 1999; 15: 289–295
  • Yang S H, Perez E, Cutright J, et al. Testosterone increases neurotoxicity of glutamate in vitro and ischemia-reperfusion injury in an animal model. J Appl Physiol 2002; 92: 195–201
  • Shamberger R C, Thistlethwaite P A, Thibault L E, Talbot T L, Brennan M F. The effect of testosterone propionate on wound healing in normal and castrate rats. J Surg Res 1982; 33: 58–68
  • Pearce C W, Foot N C, Jordan G L, Jr., Law S W, Wantz G E, Jr. The effect and interrelation of testosterone, cortisone, and protein nutrition on wound healing. Surg Gynecol Obstet 1960; 111: 274–284

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.