Journal of
Medicinal Plants Research

  • Abbreviation: J. Med. Plants Res.
  • Language: English
  • ISSN: 1996-0875
  • DOI: 10.5897/JMPR
  • Start Year: 2007
  • Published Articles: 3835

Full Length Research Paper

Effect of ethanol extract of plant mixture on hair regeneration in human dermal papilla cells and C57BL/6J mice

Bo Huang
  • Bo Huang
  • College of Food Science and Engineering, Liaoning Medical University, Jinzhou 121000, China.
  • Google Scholar
Bueom-Goo Kang
  • Bueom-Goo Kang
  • Department of Food Science and Nutrition, College of Natural Sciences, Hallym University, Chuncheon, Gangwon-do, 200-702, Republic of Korea.
  • Google Scholar
Zhiqiang Wang
  • Zhiqiang Wang
  • Department of Food Science and Nutrition, College of Natural Sciences, Hallym University, Chuncheon, Gangwon-do, 200-702, Republic of Korea.
  • Google Scholar
Soon Sung Lim
  • Soon Sung Lim
  • 2Department of Food Science and Nutrition, College of Natural Sciences, Hallym University, Chuncheon, Gangwon-do, 200-702, Republic of Korea. 3Institute of Natural Medicine, Hallym University, Chuncheon, 200-702, Republic of Korea. 4Institute of Korean Nutrition, Hallym University, Chuncheon, 200-702, Republic of Korea.
  • Google Scholar


  •  Received: 07 January 2014
  •  Accepted: 16 November 2015
  •  Published: 03 December 2015

 ABSTRACT

This study determined the effect of 30% ethanol extract of plant mixture (EPM: Glycyrrhiza uralensis, Angelica gigas, Acorus calamus, Cnidium officinale, Panax ginseng, Camellia sinensis, Salvia miltiorrhiza, Zanthoxylum schinifolium, Carthamus tinctorius, Prunus persica and Scrophularia buergeriana) on promotion of hair growth in human hair dermal papilla cells and C57BL/6J mice. EPM significantly increased the proliferation of human hair dermal papilla cells in a dose- and time- dependent manner (p < 0.05 and p < 0.01). EPM also enhanced mRNA and protein levels of growth factors such as IGF-1, VEGF, KGF and HGF. Moreover, photographical and histological observation showed that application of EPM resulted in the early onset and prolongation of the anagen phase of the hair cycle. In addition, the EPM was revealed to possess potent inhibitory effect on the 5α-reductase activity. Taken together, these results suggest that EPM has hair growth promoting potential and can be used for hair growing products.

Key words: Hair growth, extract of plant mixture, growth factors, steroid 5α-reductase.


 INTRODUCTION

Hair growth is controlled by a unique repetitive cycle comprising a hair fiber production phase (anagen), a brief regression phase (catagen) and a resting period (telogen) (Stenn et al., 1996; Paus and Cotsarelis, 1999). The regulatory mechanism of the hair cycle has not yet  been fully understood as several factors were implicated to exert their specific roles in hair growth control. Androgens are known to cause regression and balding on the scalp in genetically disposed individuals. Testosterone and dihydrotestosterone (DHT), which is formed from testosterone by the action of 5α-reductase, are two major androgens, and DHT has been considered more potent to trigger hair growth or hair loss (Russell and Wilson, 1994). The main role of 5α-reductase is to metabolize testosterone into a more potent potent androgen, DHT, which can bind firmly to androgen receptors with greater affinity and slower dissociation rate than testosterone. Molecular cloning studies have characterized two genes that encode two isoenzymes, 5αR1 and 5αR2. The former exists predominantly in the skin, whereas the latter exists in the prostate (Liu and Yamauchi, 2008). Human hair dermal papilla cells (HHDPCs) are a group of specialized fibroblasts within the hair follicle bulb; these cells play an essential role in the control of hair growth through the secretion of diffusible proteins such as insulin-like growth factor-1 (IGF-1), vascular endothelial growth factor (VEGF), keratinocyte growth factor (KGF) and hepatocyte growth factor (HGF) (Philpott and Kealey, 1994; Itami et al., 1995; Lachgar et al., 1996; Fujie et al., 2001).
 
Recently, the Food and Drug Administration (FDA) has approved two drugs- finasteride (inhibitor of 5α-reductase) and minoxidil (anti-hypertensive), for the treatment of hair loss in men. However, these drugs have limited therapeutic uses owing to their unsatisfactory cure rates (D'Amico and Roehrborn, 2007). Lately, several studies have shown that hair growth is affected by hair follicle inflammation, hair follicle oxidation and hair follicle keratinocyte apoptosis (Peters et al., 2006). There is little published data regarding the anti-inflammatory, anti-oxidative and anti-apoptotic properties, and hair growth effects of Glycyrrhiza uralensis (Shin et al., 2008), Angelica gigas (Lee et al., 2003), Acorus calamus (Sandeep and Nair, 2010), Cnidium officinale (Onishi et al., 1998), Panax ginseng (Matsuda et al., 2003), Camellia sinensis (Hsu, 2005), Salvia miltiorrhiza (Yagi and Takeo, 2003), Zanthoxylum schinifolium (Tsai et al., 2000), Carthamus tinctorius (Akihisa et al., 1996), Prunus persica (Valero et al., 2012) and Scrophularia buergeriana (Jeong et al., 2008). In the present study, whether EPM affects proliferation and up-regulation of HHDPCs growth factors was investigated. Furthermore, topical application of EPM promoted hair regeneration in C57BL/6J mice. 


 MATERIALS AND METHODS

All the plant mixtures were obtained from Deakwang, Korea. About 250 g of plant mixture [G. uralensis (root), A. gigas (root), A. calamus (root), C. officinale (root), P. ginseng (root), C. sinensis (leaf), S. miltiorrhiza (root), Z. schinifolium (bark), C. tinctorius (flower), P. persica (seed) and S. buergeriana (root) (1:1:1:1:1:1:1:1:1:1:2.5) were crushed and 2000 ml of 30% ethanol was mixed at 65°C for 120 min. The extract was then concentrated in a vacuum evaporator. The weight of the resulting residue was determined as 50 g, and it was then dissolved in 30% ethanol for subsequent treatment for in vivo study. EPM for in vitro study were then filtered to sterilize using 0.20 μm-filter units (ADVANTEC, Toyo Roshi Kaisha, Japan).
 
Preparation of rat microsomes
 
The method of preparation of rat microsomal suspension has been reported elsewhere (Liu et al., 2006). Two male Sprague-Dawley rats were killed. The livers were removed and rinsed with cold normal saline solution. Specimens were then minced with scissors and homogenized in a solution composed of Krebs-Ringer phosphate buffer (pH 7.2). The homogenate was then centrifuged three times at 900 x g, 4°C for 10 min each time. All the supernatants were collected. The resulting supernatants containing microsomal particles were tested for soluble protein by the Lowry method (Lowry et al., 1951) and kept at -80°C until use.
 
Measurement of steroid 5α-reductase inhibitory activity
 
5α-Reductase assay was performed according to the method of Matsuda et al. (2001) with some modifications. The 0.75 ml reaction solutions each contained 0.05 ml of various plant extracts in dimethyl sulfoxide (DMSO) solution, 0.05 ml of freshly prepared 500 μg/ml testosterone solution in 50% ethanol solution, 0.2 ml of microsomal suspension, 0.08 ml of 0.77 mg/ml NADPH in 0.01 M Tris-HCl buffer (pH 7.2), and 0.37 ml of 2 mM dithiothreitol water solution; the reaction solutions were then incubated at 37°C for 30 min. The reactions were then stopped by adding 0.82 ml dichloromethane, followed by 0.1 mg/ml p-hydroxybenzoic acid n-hexyl ester (as an internal standard for HPLC). Samples were shaken for 60 s, and then centrifuged at 400 xg for 10 min. The methylene chloride (MC) phase was decanted and evaporated till dry. The residue was redissolved in 500 μl methanol. A 20 μl aliquot was analyzed for remaining testosterone content using high pressure liquid chromatography (HPLC). Samples were injected into an analytical Hypersil®-Gold column (Thermo Scientific, USA, part No 25005-154630) 150 x 4.6 mm i.d. with 5 μm internal particle size, using testosterone (> 98% pure) as a standard. A isocratic elution mode was applied, in which the 65% methanol water solution (v/v) were used as the mobile phase with a flow rate of 1 ml/min and was detected under UV absorbance at 242 nm. The temperature of the column was maintained at 40°C. To determine inhibitory activity, two control reactions must be completed: firstly, a complete reaction (rxn) containing 0.2% by the volume of the 50% ethanol instead of the extract; secondly, an enzyme blank (control) in which 5.0 ml dichloromethane is added before the addition of NADPH, so that the conversion of testosterone into DHT does not occur due to the enzyme denaturation. The percent inhibition was calculated using peak area ratio (r) of testosterone/internal standard following the equation:
 
Inhibition (%) = [(rsample - rrxn)/(rctrl - rrxn)] x 100
 
Cells culture and proliferation assay
 
HHDPCs were purchased from ScienCell (Calsbad, CA) as primary cells and grown in mesenchymal stem cell medium (MSCM) supplemented with 5% fetal bovine serum (FBS), 1% penicillin-streptomycin, and 1% mesangial cell growth supplement (MSCGS) in humidified environment at 37°C in 5% CO2. The cell proliferation assay were performed using MTS [3-(4, 5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; MTS] assay kit (Promega, Madison, USA). Briefly, HHDPCs were seeded into 96-well plates (1.5 x 104 cells/well) and treated with various concentrations of EPM (0, 0.05, 0.1, 0.2, 0.5 and 1 μg/ml) and minoxidil (1 μM) for 24, 48 and 72 h, respectively. After incubation, 20 μl/well of MTS solution was incubated for 1 h at 37°C in a humidified 5% CO2 atmosphere. The optical density at 490 nm was measured three times using an ELISA reader (Bio-Tek, Vermont).
 
Animals
 
All animal experiment procedures were as per the guidelines approved by the Institutional Animal Care and Use Committees (IACUC) of Hallym University (Hallym-2012-70-1). Five 5-week-old male C57BL/6J mice (n = 5) purchased from Central Lab Animal (SLC, Japan) were housed individually in stainless steel cages in a room with controlled temperature (22 ± 2°C), relative humidity (50 ± 10%), and 12 h light-dark cycle. Mice were maintained on a commercial pellet diet and fresh tap water. After acclimatization for 7 days, all mice were anesthetized with an intraperitoneal injection of zoletil (Virbac, Carros, France). Mice were randomly allocated into three experimental groups of five animals each and provided with one of the following treatments: 30% ethanol as vehicle for negative control group, 5% minoxidil for positive control group, and 1 mg/ml EPM for experimental group. About 200 μl sample was topically applied to all mice every day for 30 days.
 
Histological analysis
 
Individual back dermal tissue samples were fixed in 4% paraformaldehyde (Sigma-Aldrich, MO, USA), and the tissues were then dehydrated and embedded in paraffin using an automatic tissue processor (Leica ASP 300; Leica Microsystems, Wetzler, Germany), sectioned to 5 μm thickness with a microtome, stained with hematoxylin and eosin, and then examined under light microscope (Olympus CX 31; Olympus Tokyo, Japan) for studying histological morphology.
 
Reverse transcription-polymerase chain reaction (RT-PCR)
 
Total RNA was extracted from the HHDPCs and mouse back dermal tissue using the EASY-BLUE total RNA extraction kit (Intron Bio, Beverly, MA). cDNA synthesis was performed with 1 μg of total RNA, oligo (15)dT primers and reverse transcriptase in a total volume of 50 μl. PCR reactions were performed in a total volume of 20 μl comprising 2 μl of cDNA product, 0.2 mM of each dNTP, 20 pmol of each primer, and 0.8 unit of Taq polymerase. Oligonucleotide primer sequences used in the experiments are shown Table 1. The RT-PCR products were electrophoresed in 1% agarose gels under 100 V and stained with 0.5 μg/ml ethidium bromide. Scanning densitometry was performed using an i-MAXTM Gel Image Analysis system (Core-Bio, Seoul, Korea).
 
 
Protein extraction and Western blot
 
HHDPCs and mouse back dermal tissue were homogenized in lysis buffer. Total proteins (30 μg) were separated using 8% SDS-polyacrylamide gel electrophoresis, transferred to polyvinylidene difluoride membranes (Millipore, Billerica, MA), and then hybridized overnight at 4°C with 1:1000 diluted IGF-1, VEGF, KGF, HGF and Actin primary antibodies (Santa Cruz Biotechnology,  Santa  Cruz, CA). After incubation with 1:2000 diluted horseradish-peroxidase-conjugated goat anti-rabbit or donkey anti-rabbit immunoglobulin G secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA) for 1 h at room temperature, immunoreactive proteins were visualized by an enhanced chemiluminescent solution (Amersham, Uppsala, Sweden) and quantified using a densitometric analysis.
 
Statistical analysis
 
The results are represented as the mean ± S.E. The findings of the control and treated groups were compared using ANOVA variance analysis and their significance was analyzed using Student’s t-test. Differences of p < 0.05 were considered statistically significant. 


 RESULTS

EPM induction of HHDPCs proliferation
 
EPM increased the proliferation of HHDPCs in a dose and time-dependent manner (Figure 1). Compared with the control, in the HHDPCs, the proliferation rates were increased by 31% on adding 1 μg/ml of EPM and by 36% on adding 1 μM of minoxidil after 72 h. The result suggests that none of the EPM inhibited cell viability at concentrations shown to be effective with minoxidil.
 
 
EPM expression of growth factors in HHDPCs
 
Insulin-like growth factor-1 (IGF-1), vascular endothelial growth factor (VEGF), keratinocyte growth factor (KGF) and hepatocyte growth factor (HGF) have been previously identified as key mediators of hair growth and generation (Peus and Pittelkow, 1996; Yano et al., 2001; Peters et al., 2005). Therefore, the expression of these growth factors in the presence and absence of EPM were compared by using RT-PCR and Western blot analysis. As shown in Figure 2 EPM was observed to increase the expression of growth factors (IFG-1, VEGF, KGF and HGF) in HHDPCs. EPM treatment for 24 h with various concentrations increased the expression of growth factor mRNA (Figure 2A) and protein (Figure 2B).
 
 
Effect of EPM on 5α-reductase activity
 
Conversion of testosterone to DHT is an important factor in hair loss. To determine whether EPM could inhibit 5α-reductase activity, the 5α-reductase activities were examined using microsomes from rat liver. As shown in Figure 3, EPM inhibited 5α-reductase activities by 37.1, 59.4 and 78.8% in a dose-dependent manner at concentrations of 0.1, 0.5, and 1 μg/ml. Finasteride, a positive control inhibited 5α-reductase activities by 89% at a concentration of 1 mg/ml.
 
 
Effect of EPM on hair regeneration in C57BL/6J mice
 
The dorsal hair in C57BL/6J mice is known to have a time-synchronized hair growth cycle (Park et al., 2011). Depilated mice skin in telogen phase is pink and darkens along with anagen initiation and then turns gray (Paus et al., 1990). As shown in Figure 4, all mice in EPM groups and minoxidil group exhibited light gray skin from 1 week after depilation, and their hair shafts were visible at 2 week after depilation. Three weeks after depilation, the back dermal tissue was in mature anagen phase in all of the mice. Histologic studies showed that EPM markedly increased the number and morphology of hair follicles as compared to the control group (Figure 5). This result clearly shows that EPM induces early onset of anagen and stimulates hair growth in C57BL/6J mice. 
 
 
 
Effect of EPM on growth factors in back dermal tissue C57BL/6J mice
 
Parallel to in vitro assay, gene and protein expressions responsible for growth factors in the back dermal tissue were examined. As shown in panels A and B of Figure 6, gene and protein expressions of IGF-1, VEGF, KGF and HGF increased in EPM-treated mice as compared to those in control mice. 
 


 DISCUSSION

Androgens regulate hair growth, and 5α-reductase plays a pivotal role in the action of androgens on target organs. In the preliminary study on screening plants with 5α-reductase inhibition effect, extract of S. buergeriana showed the most potent 5α-reductase inhibition activity on high performance liquid chromatography (HPLC) assay (data not shown). On the basis of our previous results, we decided to explore whether EPM of G. uralensis, A. gigas, A. calamus, C. officinale, P. ginseng, C. sinensis, S. miltiorrhiza, Z. schinifolium, C. tinctorius, P. persica and S. buergeriana (1:1:1:1:1:1:1:1:1:1:2.5) has hair growth effects in HHDPCs and C57BL/6J mice. In the meantime, EPM did not  effect  toxicity  (data  not shown). Our results demonstrated that EPM has an outstanding hair growth promoting effect as well as a regulatory role on the expression of growth factors and inhibitory effect on 5α-reductase.
 
The mesenchyme-derived dermal papilla (DP) cells play a pivotal role in hair growth regulation. The morphology of DP cells can be altered through the hair growth cycle, being highest in volume in the growing phase and lowest in the resting phase. In the present study, the mesenchyme-derived DP cells are well characterized to have a regulatory role in hair growth, and it is believed that diffusible factors like IGF-1 and KGF from the DP could directly affect follicular epithelium (Itami et al., 1995). Moreover, in IGF-1 transgenic animals, hair elongation was significantly higher than that in their littermates (Roh et al., 2002). KGF, which belongs to the fibroblast growth factor (FGF) family, is an important paracrine mediator of proliferation and differentiation in a wide variety of epithelial cells (Danilenko, 1999). VEGF is a marker of angiogenesis, stimulating hair growth by facilitating the supply of nutrients to the hair follicle, increasing follicular diameter (Gnann et al., 2013). The regulatory role of KGF in hair growth is well implicated in transgenic mice models in which KGF directly affects the development of hair follicles (Werner et al., 1994). Accordingly, the regulation of growth factors like IGF-1 and KGF in DP cells is fundamental to the control of hair follicle growth. In this study, we haveshown that EPM increased levels of IGF-1, VEGF, KGF, and HGF mRNA and protein in the HHDPCs and C57BL/6J mice. These results suggest that EPM may have positive effects on the hair growth promotion, partly through the regulation of growth factors in the HHDPCs and C57BL/6J mice.
 
In all animals, including humans, two different 5α-reductase isozymes have been characterized. Although type I is predominant in scalp, type II has been identified to have a crucial role in the hair growth regulation. The important role of type II 5α-reductase is supported by a well-documented male pseudohermaphroditism where type II 5α-reductase is deficient and shows no androgenetic alopecia (Hoffmann and Happle, 2000). Moreover, the FDA USA approved the use of finasteride, an inhibitor of type II 5α-reductase, in men with androgenetic alopecia. Thus, it is noteworthy that EPM has a potent inhibitory effect on type II 5α-reductase activity.
 
Overall, it has been demonstrated that EPM is capable of preventing hair loss via proliferation of HHDPCs, inhibition of 5α-reductase activity and putative molecular regulatory roles, thus suggesting that EPM may be a good candidate for promoting hair growth. 


 CONFLICT OF INTERESTS

The authors have not declared any conflict of interests.


 ACKNOWLEDGEMENTS

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2012R1A1A2008842) and Priority Research Centers Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (NRF-2009-0094071).


 ABBREVIATIONS

EPM, Extract of plant mixture; DHT, dihydrotestosterone; HHDPCs, human hair dermal papilla cells; IGF-1, insulin-like growth factor-1; VEGF, vascular endothelial growth factor; KGF, keratinocyte growth factor; HGF, hepatocyte growth factor; HPLC, high performance liquid chromatography.



 REFERENCES

D'Amico AV, Roehrborn CG (2007). Effect of 1 mg/day finasteride on concentrations of serum prostate-specific antigen in men with androgenic alopecia: A randomized controlled trial. Lancet Oncol. 8(1):21-25.
Crossref

 

Danilenko DM (1999). Preclinical and early clinical development of keratinocyte growth factor, an epithelial-specific tissue growth factor. Toxicol. Pathol. 27(1):64-71.
Crossref

 
 

Fujie T, Katoh S, Oura H, Urano Y, Arase S (2001). The chemotactic effect of a dermal papilla cell-derived factor on outer root sheath cells. J. Dermatol. Sci. 25(3):206-212.
Crossref

 
 

Gnann LA, Castro RF, Azzalis LA, Feder D, Perazzo FF, Pereira EC, Rosa PC, Junqueira VB, Rocha KC, Machado CD, Paschoal FC, Deabreu LC, Valenti VE, Fonseca FL (2013). Hematological and hepatic effects of vascular epidermal growth factor (VEGF) used to stimulate hair growth in an animal model. BMC Dermatol. 13:15.
Crossref

 
 

Hoffmann R, Happle R (2000). Current understanding of androgenetic alopecia. Part i: Etiopathogenesis. Eur. J. Dermatol. 10(4):319-327.

 
 

Hsu S (2005). Green tea and the skin. J. Am. Acad. Dermatol. 52(6):1049-1059.
Crossref

 
 

Itami S, Kurata S, Takayasu S (1995). Androgen induction of follicular epithelial cell growth is mediated via insulin-like growth factor-i from dermal papilla cells. Biochem. Biophys. Res. Commun. 212(3):988-994.
Crossref

 
 

Jeong EJ, Lee KY, Kim SH, Sung SH, Kim YC (2008). Cognitive-enhancing and antioxidant activities of iridoid glycosides from scrophularia buergeriana in scopolamine- treated mice. Eur. J. Pharmacol. 588(1):78-84.
Crossref

 
 

Lachgar S, Moukadiri H, Jonca F, Charveron M, Bouhaddioui N, Gall Y, Bonafe JL, Plouet J (1996). Vascular endothelial growth factor is an autocrine growth factor for hair dermal papilla cells. J. Invest. Dermatol. 106(1):17-23.
Crossref

 
 

Lee S, Lee YS, Jung SH, Shin KH, Kim BK, Kang SS (2003). Anti-tumor activities of decursinol angelate and decursin from angelica gigas. Arch. Pharm. Res. 26(9):727-730.
Crossref

 
 

Liu J, Kurashiki K, Shimizu K, Kondo R (2006). Structure-activity relationship for inhibition of 5alpha-reductase by triterpenoids isolated from ganoderma lucidum. Bioorg. Med. Chem. 14(24):8654-8660.
Crossref

 
 

Liu S, Yamauchi H (2008). Different patterns of 5α-reductase expression, cellular distribution, and testosterone metabolism in human follicular dermal papilla cells. Biochem. Biophys. Res. Commun. 368(4):858-64.
Crossref

 
 

Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951). Protein measurement with the folin phenol reagent. J. Biol. Chem. 193(1):265-75.

 
 

Matsuda H, Sato N, Yamazaki M, Naruto S, Kubo M (2001). Testosterone 5alpha-reductase inhibitory active constituents from anemarrhenae rhizoma. Biol. Pharm. Bull. 24(5):586-587.
Crossref

 
 

Onishi Y, Yamaura T, Tauchi K, Sakamoto T, Tsukada K, Nunome S, Komatsu Y, Saiki I (1998). Expression of the anti-metastatic effect induced by Juzen-taiho-to is based on the content of Shimotsu-to constituents. Biol. Pharm. Bull. 21(7):761-5.
Crossref

 
 

Park S, Shin WS, Ho J (2011). Fructus panax ginseng extract promotes hair regeneration in c57bl/6 mice. J. Ethnopharmacol. 138(2):340-344.
Crossref

 
 

Paus R, Cotsarelis G (1999). The biology of hair follicles. N. Engl. J. Med. 341(7):491-7.
Crossref

 
 

Paus R, Stenn KS, Link RE (1990). Telogen skin contains an inhibitor of hair growth. Br. J. Dermatol. 122(6):777-784.
Crossref

 
 

Peters EM, Arck PC, Paus R (2006). Hair growth inhibition by psychoemotional stress: A mouse model for neural mechanisms in hair growth control. Exp. Dermatol. 15(1):1-13.
Crossref

 
 

Peters EM, Hansen MG, Overall RW, Nakamura M, Pertile P, Klapp BF, Arck PC, Paus R (2005). Control of human hair growth by neurotrophins: Brain-derived neurotrophic factor inhibits hair shaft elongation, induces catagen, and stimulates follicular transforming growth factor beta2 expression. J. Invest. Dermatol. 124(4):675-685.
Crossref

 
 

Peus D, Pittelkow MR (1996). Growth factors in hair organ development and the hair growth cycle. Dermatol. Clin. 14(4):559-572.
Crossref

 
 

Philpott MP, Kealey T (1994). Effects of egf on the morphology and patterns of DNA synthesis in isolated human hair follicles. J. Invest. Dermatol. 102(2):186-191.
Crossref

 
 

Roh SS, Kim CD, Lee MH, Hwang SL, Rang MJ, Yoon YK (2002). The hair growth promoting effect of sophora flavescens extract and its molecular regulation. J. Dermatol. Sci. 30(1):43-49.
Crossref

 
 

Russell DW, Wilson JD (1994). Steroid 5 alpha-reductase: Two genes/two enzymes. Annu. Rev. Biochem. 63:25-61.
Crossref

 
 

Sandeep D, Nair CK (2010). Protection of DNA and membrane from gamma-radiation induced damage by the extract of acorus calamus linn.: An in vitro study. Environ. Toxicol. Pharmacol. 29(3):302-307.
Crossref

 
 

Shin EM, Zhou HY, Guo LY, Kim JA, Lee SH, Merfort I, Kang SS, Kim HS, Kim S, Kim YS (2008). Anti-inflammatory effects of glycyrol isolated from glycyrrhiza uralensis in lps-stimulated raw264.7 macrophages. Int. Immunopharmacol. 8(11):1524-1532.
Crossref

 
 

Stenn KS, Combates NJ, Eilertsen KJ, Gordon JS, Pardinas JR, Parimoo S, Prouty SM (1996). Hair follicle growth controls. Dermatol. Clin. 14(4):543-558.
Crossref

 
 

Tsai IL, Lin WY, Teng CM, Ishikawa T, Doong SL, Huang MW, Chen YC, Chen IS (2000). Coumarins and antiplatelet constituents from the root bark of zanthoxylum schinifolium. Planta. Med. 66(7):618-623.
Crossref

 
 

Valero Y, Colina J, Ineichen E (2012). Effect of processing on the antioxidant capacity of the plum (Prunus domestica). Arch. Latinoam. Nutr. 62(4):363-369.

 
 

Werner S, Smola H, Liao X, Longaker MT, Krieg T, Hofschneider PH, Williams LT (1994). The function of kgf in morphogenesis of epithelium and reepithelialization of wounds. Science 266(5186):819-822.
Crossref

 
 

Yagi A, Takeo S (2003). Anti-inflammatory constituents, aloesin and aloemannan in aloe species and effects of tanshinon vi in salvia miltiorrhiza on heart]. Yakugaku Zasshi. 123(7): 517-532.
Crossref

 
 

Yano K, Brown LF, Detmar M (2001). Control of hair growth and follicle size by vegf-mediated angiogenesis. J. Clin. Invest. 107(4):409-417.
Crossref

 

 




          */?>