전체메뉴
검색
Article Search

JMB Journal of Microbiolog and Biotechnology

QR Code QR Code

Research article


References

  1. Kaufman KD, Olsen EA, Whiting D, Savin R, DeVillez R, Bergfeld W, et al. 1998. Finasteride in the treatment of men with androgenetic alopecia. J. Am. Acad. Dermatol. 39: 578-589.
    Pubmed CrossRef
  2. Choi N, Shin S, Song SU, Sung J-H. 2018. Minoxidil promotes hair growth through stimulation of growth factor release from adipose-derived stem cells. Int. J. Mol. Sci. 19: 691.
    Pubmed PMC CrossRef
  3. Sica DA. 2004. Minoxidil: an underused vasodilator for resistant or severe hypertension. J. Clin. Hypertens. 6: 283-287.
    Pubmed PMC CrossRef
  4. Hirshburg JM, Kelsey PA, Therrien CA, Gavino AC, Reichenberg JS. 2016. Adverse effects and safety of 5-alpha reductase inhibitors (finasteride, dutasteride): a systematic review. J. Clin. Aesthet. Dermatol. 9: 56-62.
  5. Martino PA, Heitman N, Rendl M. 2021. The dermal sheath: an emerging component of the hair follicle stem cell niche. Exp. Dermatol. 30: 512-521.
    Pubmed PMC CrossRef
  6. Yang CC, Cotsarelis G. 2010. Review of hair follicle dermal cells. J. Dermatol. Sci. 57: 2-11.
    Pubmed PMC CrossRef
  7. Jahoda C, Horne K, Oliver R. 1984. Induction of hair growth by implantation of cultured dermal papilla cells. Nature 311: 560-562.
    Pubmed CrossRef
  8. Elliott K, Messenger AG, Stephenson TJ. 1999. Differences in hair follicle dermal papilla volume are due to extracellular matrix volume and cell number: implications for the control of hair follicle size and androgen responses. J. Investig. Dermatol. 113: 873-877.
    Pubmed CrossRef
  9. Taghiabadi E, Nilforoushzadeh MA, Aghdami N. 2020. Maintaining hair inductivity in human dermal papilla cells: a review of effective methods. Skin Pharmacol. Physiol. 33: 280-292.
    Pubmed CrossRef
  10. Kubanov A, Gallyamova YA, Korableva O, Kalinina P. 2017. The role of the VEGF, KGF, EGF, and TGF-Β1Growth factors in the pathogenesis of telogen effluvium in women. Biomed. Pharmacol. J. 10: 191-198.
    CrossRef
  11. Hardy MH. 1992. The secret life of the hair follicle. Trends Genet. 8: 55-61.
    Pubmed CrossRef
  12. Kishimoto J, Burgeson RE, Morgan BA. 2000. Wnt signaling maintains the hair-inducing activity of the dermal papilla. Genes Dev. 14: 1181-1185.
    CrossRef
  13. Collins CA, Kretzschmar K, Watt FM. 2011. Reprogramming adult dermis to a neonatal state through epidermal activation of β-catenin. Development 138: 5189-5199.
    Pubmed PMC CrossRef
  14. Huelsken J, Vogel R, Erdmann B, Cotsarelis G, Birchmeier W. 2001. β-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell 105: 533-545.
    Pubmed CrossRef
  15. Deng Z, Chen M, Liu F, Wang Y, Xu S, Sha K, et al. 2022. Androgen receptor-mediated paracrine signaling induces regression of blood vessels in the dermal papilla in androgenetic alopecia. J. Investig. Dermatol. 142: 2088-2099.e2089.
    Pubmed CrossRef
  16. Dejana E. 2010. The role of wnt signaling in physiological and pathological angiogenesis. Circ. Res. 107: 943-952.
    Pubmed CrossRef
  17. Houschyar KS, Borrelli MR, Tapking C, Popp D, Puladi B, Ooms M, et al. 2020. Molecular mechanisms of hair growth and regeneration: current understanding and novel paradigms. Dermatology 236: 271-280.
    Pubmed CrossRef
  18. Kim H, Choi N, Kim DY, Kim SY, Song SY, Sung JH. 2021. TGF-β2 and collagen play pivotal roles in the spheroid formation and antiaging of human dermal papilla cells. Aging (Albany NY) 13: 19978.
    Pubmed PMC CrossRef
  19. Williams R, Pawlus AD, Thornton MJ. 2020. Getting under the skin of hair aging: the impact of the hair follicle environment. Exp. Dermatol. 29: 588-597.
    Pubmed CrossRef
  20. Pyun HB, Kim M, Park J, Sakai Y, Numata N, Shin JY, et al. 2012. Effects of collagen tripeptide supplement on photoaging and epidermal skin barrier in UVB-exposed hairless mice. Prev. Nutr. Food Sci. 17: 245.
    Pubmed PMC CrossRef
  21. Kim DU, Chung HC, Choi J, Sakai Y, Lee BY. 2018. Oral intake of low-molecular-weight collagen peptide improves hydration, elasticity, and wrinkling in human skin: a randomized, double-blind, placebo-controlled study. Nutrients 10: 826.
    Pubmed PMC CrossRef
  22. Lee MH, Kim HM, Chung HC, Kim DU, Lee JH. 2021. Low-molecular-weight collagen peptide ameliorates osteoarthritis progression through promoting extracellular matrix synthesis by chondrocytes in a rabbit anterior cruciate ligament transection model. J. Microbiol. Biotechnol. 31: 1401-1408.
    Pubmed PMC CrossRef
  23. Frantz C, Stewart KM, Weaver VM. 2010. The extracellular matrix at a glance. J. Cell Sci. 123: 4195-4200.
    Pubmed PMC CrossRef
  24. Tang Y, Luo B, Deng Z, Wang B, Liu F, Li J, et al. 2016. Mitochondrial aerobic respiration is activated during hair follicle stem cell differentiation, and its dysfunction retards hair regeneration. PeerJ. 4: e1821.
    Pubmed PMC CrossRef
  25. Lemasters JJ, Ramshesh VK, Lovelace GL, Lim J, Wright GD, Harland D, et al. 2017. Compartmentation of mitochondrial and oxidative metabolism in growing hair follicles: a ring of fire. J. Investig. Dermatol. 137: 1434-1444.
    Pubmed PMC CrossRef
  26. Ohyama M. 2019. Use of human intra-tissue stem/progenitor cells and induced pluripotent stem cells for hair follicle regeneration. Inflamm. Regen. 39: 4.
    Pubmed PMC CrossRef
  27. Fukuyama M, Tsukashima A, Kimishima M, Yamazaki Y, Okano H, Ohyama M. 2021. Human iPS cell-derived cell aggregates exhibited dermal papilla cell properties in in vitro three-dimensional assemblage mimicking hair follicle structures. Front. Cell Dev. Biol. 9: 590333.
    Pubmed PMC CrossRef
  28. Kwack MH, Jang YJ, Won GH, Kim MK, Kim JC, Sung YK. 2019. Overexpression of alkaline phosphatase improves the hairinductive capacity of cultured human dermal papilla spheres. J. Dermatol. Sci. 95: 126-129.
    Pubmed CrossRef
  29. Nicu C, Wikramanayake TC, Paus R. 2020. Clues that mitochondria are involved in the hair cycle clock: MPZL3 regulates entry into and progression of murine hair follicle cycling. Exp. Dermatol. 29: 1243-1249.
    Pubmed CrossRef
  30. Vidali S, Chéret J, Giesen M, Haeger S, Alam M, Watson RE, et al. 2016. Thyroid hormones enhance mitochondrial function in human epidermis. J. Investig. Dermatol. 136: 2003-2012.
    Pubmed CrossRef
  31. Van Beek N, Bodo E, Kromminga A, Gáspár E, Meyer K, Zmijewski MA, et al. 2008. Thyroid hormones directly alter human hair follicle functions: anagen prolongation and stimulation of both hair matrix keratinocyte proliferation and hair pigmentation. J. Clin. Endocrinol. Metab. 93: 4381-4388.
    Pubmed CrossRef
  32. Choi N, Choi J, Kim JH, Jang Y, Yeo JH, Kang J, et al. 2018. Generation of trichogenic adipose-derived stem cells by expression of three factors. J. Dermatol. Sci. 92: 18-29.
    Pubmed CrossRef
  33. Peus D, Pittelkow MR. 1996. Growth factors in hair organ development and the hair growth cycle. Dermatol. Clin. 14: 559-572.
    Pubmed CrossRef
  34. Alexandrescu DT, Kauffman CL, Dasanu CA. 2009. Persistent hair growth during treatment with the EGFR inhibitor erlotinib. Dermatol. Online J. 15: 4.
    CrossRef
  35. Yano K, Brown LF, Detmar M. 2001. Control of hair growth and follicle size by VEGF-mediated angiogenesis. J. Clin. Investig. 107: 409-417.
    Pubmed PMC CrossRef
  36. Gentile P. 2019. Autologous cellular method using micrografts of human adipose tissue derived follicle stem cells in androgenic alopecia. Int. J. Mol. Sci. 20: 3446.
    Pubmed PMC CrossRef
  37. Back SH, Yoon JB, Sim WY, Haw CR. 1999. Effects of vaseular endothelial growth factors on hair growth in vitro. Korean J. Dermatol. 37: 23-30.
  38. Katzer T, Leite Junior A, Beck R, da Silva C. 2019. Physiopathology and current treatments of androgenetic alopecia: going beyond androgens and anti‐androgens. Dermatol. Ther. 32: e13059.
    Pubmed CrossRef
  39. Paus R, Cotsarelis G. 1999. The biology of hair follicles. New Eng. J. Med. 341: 491-497.
    Pubmed CrossRef
  40. Kumar N, Rungseevijitprapa W, Narkkhong NA, Suttajit M, Chaiyasut C. 2012. 5α-reductase inhibition and hair growth promotion of some Thai plants traditionally used for hair treatment. J. Ethnopharmacol. 139: 765-771.
    Pubmed CrossRef
  41. Müller-Röver S, Foitzik K, Paus R, Handjiski B, van der Veen C, Eichmüller S, et al. 2001. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J. Investig. Dermatol. 117: 3-15.
    Pubmed CrossRef
  42. Piccolo M, Ferraro MG, Maione F, Maisto M, Stornaiuolo M, Tenore GC, et al. 2019. Induction of hair keratins expression by an annurca apple-based nutraceutical formulation in human follicular cells. Nutrients 11: 3041.
    Pubmed PMC CrossRef
  43. An SY, Kim HS, Kim SY, Van SY, Kim HJ, Lee JH, et al. 2022. Keratin-mediated hair growth and its underlying biological mechanism. Commun. Biol. 5: 1270.
    Pubmed PMC CrossRef

Related articles in JMB

More Related Articles

Article

Research article

J. Microbiol. Biotechnol. 2024; 34(1): 17-28

Published online January 28, 2024 https://doi.org/10.4014/jmb.2308.08013

Copyright © The Korean Society for Microbiology and Biotechnology.

Low Molecular Weight Collagen Peptide (LMWCP) Promotes Hair Growth by Activating the Wnt/GSK-3β/β-Catenin Signaling Pathway

Yujin Kim1,2†, Jung Ok Lee1†, Jung Min Lee1, Mun-Hoe Lee3, Hyeong-Min Kim3, Hee-Chul Chung3, Do-Un Kim3, Jin-Hee Lee3, and Beom Joon Kim1,2*

1Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
2Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Republic of Korea
3Health Food Research and Development, NEWTREE Co., Ltd., Seoul 05604, Republic of Korea

Correspondence to:Beom joon Kim,        beomjoon74@gmail.com

Yujin Kim and Jung Ok Lee contributed equally to this work.

Received: August 9, 2023; Revised: September 5, 2023; Accepted: September 21, 2023

Abstract

Low molecular weight collagen peptide (LMWCP) is a collagen hydrolysate derived from fish. We investigated the effects of LMWCP on hair growth using human dermal papilla cells (hDPCs), human hair follicles (hHFs), patch assay, and telogenic C57BL/6 mice, while also examining the underlying mechanisms of its action. LMWCP promoted proliferation and mitochondrial potential, and the secretion of hair growth-related factors, such as EGF, HB-EGF, FGF-4, and FGF-6 in hDPCs. Patch assay showed that LMWCP increased the neogeneration of new HFs in a dose-dependent manner. This result correlated with an increase in the expression of dermal papilla (DP) signature genes such as, ALPL, SHH, FGF7, and BMP-2. LMWCP upregulated phosphorylation of glycogen synthase kinase-3β (GSK-3β) and β-catenin, and nuclear translocation of β-catenin, and it increased the expression of Wnt3a, LEF1, VEGF, ALP, and β-catenin. LMWCP promoted the growth of hHFs and increased the expression of β-catenin and VEGF. Oral administration of LMWCP to mice significantly stimulated hair growth. The expression of Wnt3a, β-catenin, PCNA, Cyclin D1, and VEGF was also elevated in the back skin of the mice. Furthermore, LMWCP increased the expression of cytokeratin and Keratin Type I and II. Collectively, these findings demonstrate that LMWCP has the potential to increase hair growth via activating the Wnt/β-catenin signaling pathway.

Keywords: LMWCP (low molecular weight collagen peptide), &beta,-catenin, human dermal papilla cells (hDPCs), VEGF, Wnt3a

Introduction

Alopecia is on the rise regardless of gender and age. Currently, there are two drugs approved by the United States Food and Drug Administration (US FDA): Finasteride and Minoxidil (MNX)[1]. Finasteride promotes a decrease in the concentration of dihydrotestosterone (DHT), which induces apoptosis in human dermal papilla cells (hDPCs) [1]. MNX enhances the nutrient supply to hair follicles through vasodilation [2]. However, these drugs have side effects such as allergic contact dermatitis and itching. Furthermore, discontinuation of MNX leads to the recurrence of alopecia, while prolonged use of finasteride can cause sexual dysfunction [3, 4]. Therefore, there is a significant demand for new hair loss treatment options with fewer side effects and easier accessibility.

Human hair follicles (hHFs) are composed of epidermal (epithelial) and dermal (mesenchymal) compartments, and their communication is crucial in the morphogenesis and growth of HFs [5, 6]. hDPCs play a pivotal role in the regulation of growth, formation, and cycling of hHF mainly through reciprocal interactions with surrounding epithelial cells [7, 8]. Growth factors, including insulin-like growth factor-1 (IGF-1), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), and keratinocyte growth factor (KGF), secreted from hDPCs stimulate keratinocytes to proliferate and differentiate into the hair shaft during the anagen phase [9, 10].

Wnt/β-catenin signaling pathway is important in regulating the growth cycle of hair follicles [11, 12]. Its activation promotes the proliferation and migration of hair follicle stem-cells, hair matrix cells, and hDPCs [13, 14], thereby inducing the transition of hHFs from telogen to anagen. In addition, this pathway can promote angiogenesis and provide a nutrient-rich environment for hHFs growth [15, 16]. Therefore, targeting this pathway represents a potential approach for hair-loss prevention and treatment.

hHFs undergo repetitive cycles of growth (anagen), regression (catagen), and rest (telogen) [17]. The volume of dermal papilla (DP) in hHFs is greatly affected by the amount of collagen during the anagen phase. Collagen is essential for increasing hair thickness [18], making collagen an important factor in maintaining normal hair growth. However, skin aging promotes the cleavage of collagen through the activation of matrix metalloproteinases (MMPs), and this collagen loss is not completely replenished by de novo collagen synthesis [19]. Therefore, supplementing external collagen can help maintain healthy hair during aging.

The low molecular weight collagen peptide (LMWCP) is derived from a skin of pangasius hypophthalmus, and a collagen hydrolysate containing 3% Gly-Pro-Hyp and 15% tripeptide [20]. According to the previous studies, LMWCP has various health benefits, including wrinkle reduction, increased hydration and elasticity, and cartilage regeneration [21, 22]. Interestingly, Hyunju et al. revealed that collagen plays pivotal roles in spheroid formation and anti-aging of hDPCs [18]. Furthermore, Christian et al. reported that collagen crosslinking due to photoaging in the scalp increases with age, impacting the remodeling of the hHFs during early anagen as it moves downwards in the dermis [23]. These results indicate that collagen is a key regulator for hair growth and hair health.

Considering the above data, LMWCP, which is mainly composed of collagen, can play a positive role in regulating and maintaining hair growth. Thus, in this study, we investigated the effect of LMWCP on hair growth and explored the mechanism underlying its hair growth-promoting activity using the anagen induction assay in telogenic C57BL/6 mice, as well as patch assays, hHF organ cultures, and hDPCs.

Materials and Methods

LMWCP and Dermal Papilla Cell Culture

LMWCP supplied by NEWTREE Co., Ltd., South Korea was prepared by spray drying the gelatin hydrolysate obtained by enzymatic degradation of gelatin derived from a skin of pangasius hypophthalmus using a protease and was standardized based on Gly-Pro-Hyp (3%) and tripeptide (≥15%) contents. hDPCs were purchased from PromoCell (Germany). The cells were maintained in an incubator with 5% CO2 at 37°C using a follicle dermal papilla cell growth medium kit (PromoCell) and subcultured upon reaching 70-80% confluency.

Cell Viability Assay

hDPCs were seeded in 96-well plates and cultured to reach a confluency of 90%. After 24 h, the cells were treated with 0, 0.1, 0.3, 1, and 3 mg/ml of LMWCP for 24 h. Cell viability was quantified using a WST-8 assay kit (QuantiMax, Biomax, Korea). Absorbance was measured at 450 nm using a microplate spectrophotometer (SpectraMax 340; Molecular Devices, Inc., USA).

Mitochondrial Bioenergetics Assessment

Mitochondrial membrane potential was measured using a JC-1 mitochondrial membrane potential assay kit (Abcam, UK). Briefly, hDPCs treated with LMWCP or MNX (Sigma-Aldrich, USA) were stained with 1 μM JC-1 solution. Fluorescence intensities from JC-1 aggregate and monomer forms were measured at 590 nm (535 nm excitation) and 530 nm (475 nm excitation), respectively, using a microplate spectrophotometer (SpectraMax 340; Molecular Devices, Inc., USA). Mitochondrial membrane potential (ΔΨm) was visualized by capturing fluorescence images using a fluorescence microscope (DMi8, Leica, Germany).

Immuno Blot Assay

Total cellular proteins from hDPCs were collected and lysed in RIPA buffer (Thermo Fisher Scientific, USA). Protein samples (30 μg) were analyzed using western western blot assay with the following antibodies; Cytokeratin antibody (Sigma-Aldrich); VEGF and β-actin antibody (Santa Cruz Biotechnology, USA); Alkaline phosphatase (ALP) antibody (Thermo Fisher Scientific); Cyclin D1, p-GSK-3β (Ser9), GSK-3β, p-β-catenin (Ser675), p-β-catenin (Ser33/37/Thr41), total β-catenin, p-PKA(Thr197), PKA, LEF1, CDK2, p-AKT(Ser473), AKT, β-actin, and proliferating cell nuclear antigen (PCNA) antibody (Cell Signaling Technology Inc. USA); type I + II hair keratin antibody (Progen Biotechnical GmbH, Germany); Cyclin E, CDK6, and Wnt3a antibody (Abcam, UK). Protein bands were visualized using a ChemiDoc MP Imaging System (Bio-Rad Laboratories, Inc., USA). The resulting blots were analyzed using NIH Image J software (Bethesda, USA).

Immunocytochemistry (ICC)

The cells were fixed with 4% paraformaldehyde (PFA) for 30 min, washed with PBS (phosphate-buffered saline), blocked with 3% BSA (bovine serum albumin) and 0.2% Triton X-100 in PBS at room temperature (RT) for 1 h, and incubated with primary antibodies overnight at 4°C. After washing with PBS, the cells were incubated with anti-rabbit IgG-FITC secondary antibodies (Santa Cruz Biotechnology) in the dark at RT for 1 h. The cell nuclei were counter-stained with 4',6-Diamidino-2-Phenylindole, dihydrochloride (DAPI) (Immuno Bioscience Corp., USA), and the cells were observed using confocal microscopy (LSM 880, Zeiss, Germany).

Quantitative RT-PCR (qRT-PCR) Analysis

Total RNA was extracted using the TRIzol reagent (Invitrogen, USA). cDNA synthesis was performed using Prime Script TM RT Master Mix (Takara, Japan). Quantitative PCR was performed using qPCR 2X PreMIX SYBR (Enzynomics, Korea) on a CFX96 Touch Real-Time PCR Detection System (Bio-Rad). Gene expression levels were calculated and reported as cycle threshold (Ct) values using the ΔCt quantification method. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used for normalization. The primers used for qPCR are summarized in Table 1.

Table 1 . Primer sequences used for quantification of gene expression..

GenePrimer sequence (5'→ 3')
Human EGFFCAGGGAAGATGACCACCACT
RCAGTTCCCACCACTTCAGGT
Human HB-EGFFACAAGGAGGAGCACGGGAAAAG
RCGATGACCAGCAGACAGACAGATG
Human FGF-4FGGGAGTCTACAGACAGCAAG
RGAGCCTAGGGTGTGGTTTA
Human FGF-6FGGGAGTCTACAGACAGCAAG
RGAGCCTAGGGTGTGGTTTA
Human ALPLFATTGACCACGGGCACCAT
RCTCCACCGCCTCATGCA
Human SHHFGCGCCAGCGGAAGGTAT
RCCGGTGTTTTCTTCATCCTTAAA
Human FGF7FATCAGGACAGTGGCAGTTGGA
RAACATTTCCCCTCCGTTGTGT
Human BMP-2FGAGGTCCTGAGCGAGTTCGA
RTCTCTGTTTCAGGCCGAACA


Table 2 . Antibodies used for Western blot analysis..

AntibodiesProduct codeCompany
Anti-MITFMAB3747-ISigma-Aldrich (MO, USA)
Anti-Calnexinab22595Abcam (Cambridge, UK)
Anti-ALIXab275377Abcam (Cambridge, UK)
Anti-CD63ab134045Abcam (Cambridge, UK)
Anti-tyrosinaseab180753Abcam (Cambridge, UK)
Anti-p-MITFab59201Abcam (Cambridge, UK)
Anti-TRP-1sc-58437Santa Cruz Biotechnology (CA, USA)
Anti-TRP-2sc-25544Santa Cruz Biotechnology (CA, USA)
Anti-Rab27asc-22756Santa Cruz Biotechnology (CA, USA)
Anti-β-actinsc-47778Santa Cruz Biotechnology (CA, USA)
Anti-p-CREB#9198Cell Signaling Technology Inc. (Beverly, MA)
Anti-CREB#9197Cell Signaling Technology Inc. (Beverly, MA)
Anti-p-ERK#9101Cell Signaling Technology Inc. (Beverly, MA)
Anti-ERK#9102Cell Signaling Technology Inc. (Beverly, MA)
Anti-p-AKT#4060Cell Signaling Technology Inc. (Beverly, MA)
Anti-AKT#4691Cell Signaling Technology Inc. (Beverly, MA)
Anti-p-β-catenin#4176Cell Signaling Technology Inc. (Beverly, MA)
Anti-β-catenin#8480Cell Signaling Technology Inc. (Beverly, MA)
Anti-Myosin-Va#3402Cell Signaling Technology Inc. (Beverly, MA)
Anti-MLPH10338-1-APProteinTech Group (IL, USA)


Growth Factor Antibody Array

A human growth factor antibody array membrane kit (Abcam) was used to measure changes in the profiles of the growth factors in hDPCs following LMWCP treatment. Briefly, hDPCs (5 × 105 cells/well) were seeded in 6-well plates and cultured overnight. Cells were treated with 3 mg/ml of LMWCP for 24 h, and the culture supernatants were collected for growth factor analysis. Fresh medium and culture supernatant from non-treated cells were used as blank and control, respectively. The resulting blots were analyzed under identical conditions using a chemiluminescence EZ-capture (Atto, USA).

Human HF Organ Culture

All hair follicles were obtained from Dankook University Hospital (ethical approval number 2019M-008). The isolated anagen follicles were cultured in 500 μl of Williams E medium (Gibco, USA) at 37°C with 5% CO2. After 24 h, the hHFs were cultured with 1 or 3 mg/ml LMWCP or MNX (50 μM) for 8 days. The pictures of the hair follicles were obtained using a stereo microscope (Zeiss). On day 8, each hHF was evaluated as either in anagen VI (score 1), early catagen (score 2), mid-catagen (score 3), or late catagen (score 4), and the anagen/catagen ratio was calculated for each group. hHFs elongation was analyzed using ImageJ (version 1.52a). The hHFs were then fixed in 10% formalin.

Histology and Immunohistochemistry (IHC)

Dorsal skin tissues from each mouse were fixed with 10% formalin, embedded in paraffin, and then cut into sections that were stained with hematoxylin and eosin (H&E). For IHC, the sliced sections were incubated with primary antibodies. The stained slides were photographed using a slide scanner (Pannoramic MIDI; 3DHISTECH Ltd, Hungary) and observed using Case Viewer software. The number of hHFs was counted on a cropped image in a fixed area (1 × 1 mm).

Patch Assay

Truncal skin was removed from newborn C57BL/6 mice and rinsed in Dulbeccós phosphate-buffered saline (DPBS). The skin was washed with a povidone-iodine solution and incubated with Collagenase/Dispase (2.5 mg/ml; Roche, Switzerland) overnight at 4°C. Afterward, dermal cells and epidermal cells were isolated, and 0.25%trypsin-EDTA was added to each cell population, followed by incubation at 37°C for 2 h. The cells were centrifuged at 2,000 rpm for 20 min at 4°C. A cell mixture of 1 × 106 dermal cells and 5 × 105 epidermal cells were re-suspended in DMEM-F12 medium (Hyclone) and injected (26-gauge needle) into the hypodermis of BALB/c nude mice. The dorsal skin of the mice was monitored and photographed using a digital camera for 14 days.

Hair Regeneration Model

C57BL/6 mice (six-week-old, male) were purchased from Saeron Bio Inc. (Korea) and acclimated for 1 week. The mice were maintained at 23 ± 2°C and 50 ± 10% humidity with a 12-h light/12-h dark cycle. All animal experiments were conducted according to the Principles of Laboratory Animal Care established by the National Institutes of Health (NIH) and were approved by the Chung-Ang University Institutional Animal Care and Use Committee (IACUC No. A2022018). The mice were randomly divided into four groups: normal control (n = 7), LMWCP 615 mg/kg (n = 7), LMWCP 820 mg/kg (n = 7), and MNX 3% (n = 7). LMWCP was administered once a day for two weeks through oral administration, while MNX was administered topically. To compare the growth rate, the dorsal skin was photographed using a digital camera on days 0, 10, and 13 after depilation using hair removal cream, and the ratio growth area/total area was calculated using ImageJ software. The dorsal skin was removed for histological analysis after euthanizing the mice on days 0, 10, and 13.

Statistical Analysis

All data are reported as the mean ± standard deviation (SD) of at least three independent experiments. The data were analyzed using one-way analysis of variance (ANOVA) followed by a Bonferroni post hoc test. All statistical analyses were performed using the GraphPad Prism 7.0 software (GraphPad Software Inc., USA). Differences with p values lower than 0.05 were considered statistically significant and indicated with the following symbols: *, p < 0.05; **, p < 0.01; ***, p < 0.001; and ****, p < 0.0001.

Results

LMWCP Increases Proliferation and Mitochondrial Potential of hDPCs

LMWCP significantly increased the proliferation of hDPCs by 10–30% at concentrations of 0, 0.1, 0.3, 1, and 3 mg/ml (Fig. 1A), and it also upregulated the expression of PCNA (proliferating cell nuclear antigen), a cellular marker for proliferation (Fig. 1B). Mitochondrial β-oxidation favorably impacts hair growth in vitro [24, 25]. Mitochondrial membrane potential increased by 83% and 85% upon LMWCP treatment at concentrations of 1 and 3mg/ml, respectively (Fig. 1C). Highly activated mitochondrial membrane potential was visualized using fluorescence microscopy, where red dots were enhanced while green dots were reduced by LMWCP in cultured hDPCs (Fig. 1D). LMWCP also increased the protein levels of cyclin D1, cyclin E, cyclin-dependent kinase 2 (CDK2), and cyclin-dependent kinase 6 (CDK6) in a dose-dependent manner (Fig. 1E). To investigate the effect of LMWCP on hair growth factor secretion in hDPCs, we performed a human growth factor antibody array analysis. LMWCP significantly increased the secretion of EGF, HB-EGF, FGF-4, and FGF-6, which are known to stimulate hair growth (Fig. 1F) [17]. Additionally, using qRT-PCR, we confirmed that the expression levels of these factors were significantly increased in LMWCP-treated hDPCs (Fig. 1G).

Figure 1. Effect of the LMWCP on proliferation and cellular energy metabolism in hDPCs. (A) Proliferation of hDPCs was assessed after LMWCP treatment (0, 0.1, 0.3, 1, and 3 mg/ml) for 24 h. (B) The expression of PCNA after treatment with a LMWCP for 24 h. (C) JC-1 aggregates (A590)/monomer (A530) ratio of DPCs treated with LMWCP (0, 1, and 3 mg/ml) and MNX (1 μM) for 24 h. (D) JC-1 monomer form (green) and aggregate form (red) were detected using fluorescent microscopy. (E) The expression of cyclin D1, cyclin E, CDK2, and CDK6 after treatment with LMWCP for 24 h. (E) Cultured media from hDPCs treated with either vehicle, growth media, or LMWCP (0, 0.3, 1, and 3 mg/ml) for 48 h was used for analysis using the growth factor antibody array. (F) The mRNA expression levels of EGF, HB-EGF, FGF-4, and FGF-6 in hDPCs treated with LMWCP (3 mg/ml) for 1 h were analyzed by qPCR (n = 3). The results are shown as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.

LMWCP Activates Wnt /GSK-3β/β-Catenin Signaling Pathway

GSK-3β/β-catenin signaling is necessary for the regulation of diverse biological events, including cell proliferation, hair growth, and hair regeneration [11, 12]. Upon LMWCP treatment, the levels of p-Akt (Ser473) and the phosphorylation of glycogen synthase kinase-3b (GSK-3b) on Ser9 increased in a dose-dependent manner. One other hands, LMWCP decreased the phosphorylation of β-catenin on Ser33/37/Thr41(Fig. 2A). At the same time, LMWCP treatment also increased the phosphorylation of PKA on Thr197 and β-catenin on Ser675 (Fig. 2B), leading to less proteosomal degradation and more nuclear translocation and activation of β-catenin. We confirmed that LMWCP increased the expression levels of wnt family member 3a (Wnt3a), β-catenin, lymphoid Enhancer Binding Factor 1(LEF1), and vascular endothelial growth factor (VEGF) in a dose-dependent manner (Fig. 2C). By ICC, we observed that the increased translocation of β-catenin to the nucleus in LMWCP-treated hDPCs compared to controls (Fig. 2D). These results indicate that LMWCP activates the Wnt-AKT-GSK-3β/β-catenin signaling pathway.

Figure 2. Effect of LMWCP on the Wnt-AKT-GSK-3β/β-catenin pathway. (A) hDPCs treated with LMWCP (0, 0.3, 1, and 3 mg/ml) for 1 h were lysed and analyzed for p-AKT(ser473), AKT, p-GSK(Ser9), GSK, p-β-catenin(Ser675), p-β-catenin (Ser33/37/Thr41), β-catenin, PKA, p-PKA (Thr 197), and β-actin. (B) hDPCs treated with a LMWCP (0, 0.3, 1, and 3 mg/ml) for 24 h were analyzed for Wnt3a, LEF1, β-catenin, VEGF, and β-actin. (C) Expression of β-catenin was analyzed by ICC. Representative data from three independent experiments are shown. The results are shown as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.

LMWCP Increases Hair Inductivity of hDPCs

The tendency of DPCs to aggregate is associated with inductivity of hair growth [26, 27]. We evaluated the effect of LMWCP on hair inductivity using a three-dimensional (3D) spheroid culture. LMWCP promoted the aggregation of hDPCs spheres after 2 days compared to the control (Fig. S1). Next, we investigated the expression levels of DP signature genes after LMWCP treatment using qRT-PCR. The expression levels of ALPL, Sonic hedgehog (SHH), fibroblast growth factor-7 (FGF-7), and bone morphogenetic protein-2 (BMP-2) were significantly elevated in LMWCP-treated hDPCs compared with the control group (Fig. 3A). Particularly, the expression of ALP, which improves the hair inductivity of hDPCs [28], was upregulated in a dose dependent manner after LMWCP treatment (Fig. 3B). To confirm the effect of LMWCP on new hair inductivity in ex vivo, we conducted a patch assay. LMWCP-treated mixed epidermis and dermis cells exhibited promoted new HFs induction compared to the vehicle-treated group (Fig. 3C and 3D).

Figure 3. Effect of LMWCP on potential hair inductivity of hDPCs. (A) The mRNA expression levels of ALPL, SHH, FGF-7, and BMP-2 were analyzed by qPCR (n = 3). (B) hDPCs treated with LMWCP for 24 h were analyzed using western blotting for ALP expression. (C, D) Patch assay. At 2 weeks, nude mice were euthanized, and newly generated hair follicles on the back skin were counted using H&E staining. Scale bar, 200 μm. Bar graph shows the number of hHFs in back skin. Results are presented as the mean ± SD of data from three independent experiments. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.

LMWCP Enhances hHF Growth in an ex vivo Model

HFs are organs containing DPCs. hHFs treated with LMWCP (1 and 3 mg/ml) exhibited longer growth compared to the control hHFs at day 8, similar to the hHFs treated with MNX (50 μM) (Fig. 4A). After treatment with LMWCP, hair cycle of hHFs in 8 days was analyzed using cycle scoring criteria of the HFs (Fig. S2). LMWCP increased the number of hHFs in anagen stage, indicating that LMWCP prolonged the anagen phage of hair cycle in hHFs compared to the vehicle control (Fig. 4B). In addition, IHC staining showed that LMWCP increased the expression levels of β-catenin and VEGF in hHFs compared with the vehicle control (Fig. 4C).

Figure 4. Effect of LMWCP on hair elongation in a hHF organ culture model. The hHFs (8 hair follicles/group) were treated with LMWCP (0, 1, and 3 mg/ml) or MNX (50 μM) for 8 days. (A) HFs length was analyzed under a stereomicroscope on days 0, 2, 4, 6, and 8. The relative length of each hair shaft was measured using the ImageJ software. (B) After 10days of culture, the HFs phase was assessed following the hair cycle scoring criteria. Representative images of the HFs for each experimental group are shown, as well as the calculated ratios of the hair cycle phases. (C) H&E staining and IHC staining of β- catenin and VEGF. The results are shown as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.

LMWCP Accelerates Hair Growth in Telogenic C57BL/6 Mice

Depilated mice (7 weeks old) were orally injected with either the vehicle (saline) or LMWCP (615 and 820 mg/kg, referred to as LMWCP 615 and LMWCP 820, respectively) every day for 13 days (Fig. 5A). Mouse skin color score index measurements (Fig. S3) on day 10 showed the highest score in the order of MNX, LMWCP 615, and LMWCP 820 groups (Fig. 5B and 5D). After 13 days, the area of hair regrowth was significantly higher (p<0.001) in the LMWCP 615 and LMWCP 820 groups, along with the MNX group, compared to the negative control group (Fig. 5B and 5E). The thickness of the interfollicular whole skin also significantly increased in LMWCP-treated and MNX-treated mice (p<0.05) (Fig. 5C and 5F). Analysis of representative longitudinal (HF number) and transverse (hair growth phase) sections confirmed that the number of total HFs and anagen HFs were elevated in the LMWCP‐treated groups compared to the vehicle group (Fig. 5C, 5G, and 5H). The increases in the protein levels of Wnt3a, β-catenin, PCNA, cyclin D1, and VEGF were confirmed through western blot analysis (Fig. 5I). Furthermore, we confirmed that β-catenin and VEGF significantly increased in the back skin of the mice using IHC (Fig. S4). Collectively, these results indicate that LMWCP promotes hair growth by stimulating the transition from telogen phase to anagen phase.

Figure 5. Effect of the LMWCP on anagen induction in 7-week-old female C57BL/6 mice. (A) Timetable of experimental treatments and sample collection. (B) Representative photographs of mouse back skin on days 0, 10, and 13. (C) Representative images of H&E-stained longitudinal and transverse sections of the skin of each mouse on day 13. Scale bar, 200 μm. (D) Skin color scores for 10 days. (E) Hair growth area on the back skin observed for 13 days. (F) Hair dermis thickness, (G) HF number, and (H) anagen/telogen ratios on day 13. (I) The expression levels of Wnt3a, β-catenin, PCNA, cyclin D1, and VEGF on the dorsal skin at day 13. The results are expressed as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.

LMWCP Increases the Expression of Keratins

Keratin is a primary component of hair. Throughout the keratinization process, numerous keratins organize into protein filaments to participate in the assembly of the hair shaft within follicle bulbs [29]. We investigated whether a LMWCP affects the expression of keratin in hDPCs and back skin of mice. The cytokeratin total levels were evaluated using a broad-spectrum anti-pan-cytokeratin antibody. LMWCP significantly increased the cytokeratin levels in those cells and tissues compared to the controls (Fig. 6A and 6B). As shown in Fig. 6C, following the LMWCP treatment, it was confirmed that the expression of cytokeratins increased throughout the hair follicle. Next, to explore the biological effects of the LMWCP on the hair keratin expression, both Type I and II hair keratins were determined by Western blot analysis. The expression of Type I and II hair keratins accelerated in the dorsal skin administrated with the LMWCP (Fig. 6D). Overall, these results suggest that the LMWCP can promote hair growth by increasing keratin production.

Figure 6. LMWCP increases the expression of keratin in hDPCs and the dorsal skin of mice. (A) Western blotting analysis showing the total cytokeratin levels in hDPCs treated with 0, 0.3, 1, and 3 mg/ml of LMWCP for 24 h. (B–D) Western blotting and IHC analysis showing the total cytokeratin and Type I + II hair keratin levels in the dorsal skin of the exposed LMWCP (615 and 820 mg/kg) and MNX treatment and control mice after 13 days. Representative western blotting and IHC images from four independent experiments are shown. β-actin was used as a loading control. Scale bar, 400 μm. The results are expressed as the mean ± standard deviation. **, p < 0.01; ****, p < 0.0001 compared to the control group.

Discussion

Currently, there is growing interest in hair loss prevention and hair health. LMWCP has received special attention as it is an essential component of skin and hair, and it is considered a safe natural ingredient.

Mitochondria play an important role in follicle regeneration, as mitochondrial aerobic respiration is activated during hair follicle stem cell differentiation, and its dysfunction retards hair regeneration [24, 25]. Moreover, the stimulation of mitochondrial function prolongs anagen phase, enhances hair follicle keratinocyte proliferation, and modifies intra-follicular keratin expression [29-31], indicating that mitochondrial activity play an essential role in maintaining hair growth. In the present study, we observed that LMWCP increased not only hDPCs proliferation but also mitochondrial potential (Fig. 1A and 1C). These findings suggest that the increase in hDPCs proliferation in response to LMWCP treatment may be associated with activating mitochondrial potential.

hDPCs secrete several growth factors on the HFs to promote hHFs’ growth [9]. Any changes in the distribution of the relevant growth factor receptors and their expression levels can cause abnormalities in the growth and development of hair follicles [32, 33]. As shown in Fig. 1F, we found that LMWCP stimulated the secretion of EGF. The EGF interacts with the epidermal growth factor receptor (EGFR) in the outer root sheath (ORS) of mature hHFs, which induces DNA synthesis in ORS cells and differentiates hair bulb cells into ORS cells. In addition, EGF plays an inhibitory role in hair follicle formation during the initial stages of hair follicle growth [34]. Using ex vivo and in vivo models (Figs. 4A and 5B), we found that LMWCP increased the growth of both hHFs and mouse hair. These growth-stimulating effects may be mediated with the increased secretion of growth factors acting on the hHFs.

Next, we observed that LMWCP promoted the aggregation of hDPCs (Fig. S1) and upregulated the expression of DP signature genes such as ALPL, SHH, FGF7, and BMP-2 (Fig. 3A). Hyunju et al. reported that collagne13A1 (Col13A1) and collagne15A1 (Col15A1) induce the spheroid formation of hDPCs. This collagen expression is downregulated in aged HFs, and aged hDPCs are difficult to aggregate. Blocking COL13A1 and COL15A1 expression using small interfering RNA has been found to reduce aggregation and induce senescence of hDPCs in vitro [18]. These findings indicate that collagen plays pivotal roles in spheroid formation and support our finding that LMWCP induces hair growth by increasing hDPCs’ hair inductivity.

Cyclic hair growth depends on the induction of angiogenesis to meet the increased nutritional needs of hair follicles during the anagen phase of rapid cell division [35, 36]. VEGF, as an autocrine growth factor for hDPCs, can stimulate the proliferation and migration of hDPCs [37]. Interestingly, LMWCP induces a dose-dependent increase in VEGF expression in hDPCs (Fig. 2B). In addition, VEGF expression was upregulated in human HFs and mouse hair shafts in response to LMWCP treatment (Figs. 4C, 5I, and S4). These results suggest that LMWCP could promote the supplement of nutrients by increasing angiogenesis during hair growth.

Hair growth could be regulated by modulating the hair cycle, for example by prolonging the anagen phase or promoting the telogen-to-anagen transition [38, 39]. C57BL/6 mice possess melanocytes only in the hair follicles. and melanin synthesis occurs with the hair growth cycle [40]. Thus, change of the hair growth cycle can be easily identified by simply monitoring the transition of the skin color from pink (no hair) to black (fully grown hair)[41]. As shown in Fig. 5B and 5D, the skin score was higher in mice receiving 820 mg/kg LMWCP, indicating that LMWCP induces the telogen-anagen transition earlier. The results demonstrated that LMWCP increases hair growth by stimulating telogen transition of hair cycle.

Hair keratins constitute up to 95% of the hair structure and contribute to the mechanical strength of the cells [42]. Recently, Seong et al. found that keratin is critical for condensation of hDPCs and generation of a P-cadherin-expressing cell population (hair germ) from outer root sheath cells [43], indicating that keratin can promote the hair growth by increasing the hair inductivity of hDPCs. In our study, LMWCP increased the expression of cytosolic keratin in hDPCs and the dorsal skin of mice (Fig. 6A-6C). In addition, the expression levels of hair keratin type I and type II were increased in the dorsal skin of LMWCP-treated mice (Fig. 6D). These results indicate that LMWCP could play a crucial role in increasing hair growth by increasing the expression of keratin.

Conclusion

The results of the present study provide the first evidence that LMWCP contributes to the growth and cycling of hair through the Wnt-AKT-GSK-3β/β-catenin signaling pathway. LMWCP enhances new hair formation by increasing the secretion of growth factors and promoting hair inductivity. Moreover, we observed that oral administration of LMWCP during the telogen phase accelerated the onset of the anagen phase and increased the expression of VEGF and β-catenin. Collectively, LMWCP can be used as a supplement to alleviate the symptoms of hair loss.

Supplemental Materials

Acknowledgments and Funding

This research was supported by NEWTREE Co., Ltd.

Author Contributions

Yu-jin Kim: Methodology, data curation, investigation, formal analysis, and writing. Jung Ok Lee: Writing, reviewing, editing, and supervision. Mun-Hoe Lee: Investigation and data curation. Hyeong-Min Kim: Investigation and data curation. Hee-Chul Chung: Investigation and data curation. Do-Un Kim: Investigation and data curation. Jin-Hee Lee: Investigation and data curation. Beom Joon Kim: Conceptualization, methodology, research, and project administration.

Ethics Approval and Consent to Participate

The Committee on the Ethics of Animal Experiments at Chung-Ang University approved all animal experiments.

Conflict of Interest

The authors have no financial conflicts of interest to declare.

Fig 1.

Figure 1.Effect of the LMWCP on proliferation and cellular energy metabolism in hDPCs. (A) Proliferation of hDPCs was assessed after LMWCP treatment (0, 0.1, 0.3, 1, and 3 mg/ml) for 24 h. (B) The expression of PCNA after treatment with a LMWCP for 24 h. (C) JC-1 aggregates (A590)/monomer (A530) ratio of DPCs treated with LMWCP (0, 1, and 3 mg/ml) and MNX (1 μM) for 24 h. (D) JC-1 monomer form (green) and aggregate form (red) were detected using fluorescent microscopy. (E) The expression of cyclin D1, cyclin E, CDK2, and CDK6 after treatment with LMWCP for 24 h. (E) Cultured media from hDPCs treated with either vehicle, growth media, or LMWCP (0, 0.3, 1, and 3 mg/ml) for 48 h was used for analysis using the growth factor antibody array. (F) The mRNA expression levels of EGF, HB-EGF, FGF-4, and FGF-6 in hDPCs treated with LMWCP (3 mg/ml) for 1 h were analyzed by qPCR (n = 3). The results are shown as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.
Journal of Microbiology and Biotechnology 2024; 34: 17-28https://doi.org/10.4014/jmb.2308.08013

Fig 2.

Figure 2.Effect of LMWCP on the Wnt-AKT-GSK-3β/β-catenin pathway. (A) hDPCs treated with LMWCP (0, 0.3, 1, and 3 mg/ml) for 1 h were lysed and analyzed for p-AKT(ser473), AKT, p-GSK(Ser9), GSK, p-β-catenin(Ser675), p-β-catenin (Ser33/37/Thr41), β-catenin, PKA, p-PKA (Thr 197), and β-actin. (B) hDPCs treated with a LMWCP (0, 0.3, 1, and 3 mg/ml) for 24 h were analyzed for Wnt3a, LEF1, β-catenin, VEGF, and β-actin. (C) Expression of β-catenin was analyzed by ICC. Representative data from three independent experiments are shown. The results are shown as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.
Journal of Microbiology and Biotechnology 2024; 34: 17-28https://doi.org/10.4014/jmb.2308.08013

Fig 3.

Figure 3.Effect of LMWCP on potential hair inductivity of hDPCs. (A) The mRNA expression levels of ALPL, SHH, FGF-7, and BMP-2 were analyzed by qPCR (n = 3). (B) hDPCs treated with LMWCP for 24 h were analyzed using western blotting for ALP expression. (C, D) Patch assay. At 2 weeks, nude mice were euthanized, and newly generated hair follicles on the back skin were counted using H&E staining. Scale bar, 200 μm. Bar graph shows the number of hHFs in back skin. Results are presented as the mean ± SD of data from three independent experiments. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.
Journal of Microbiology and Biotechnology 2024; 34: 17-28https://doi.org/10.4014/jmb.2308.08013

Fig 4.

Figure 4.Effect of LMWCP on hair elongation in a hHF organ culture model. The hHFs (8 hair follicles/group) were treated with LMWCP (0, 1, and 3 mg/ml) or MNX (50 μM) for 8 days. (A) HFs length was analyzed under a stereomicroscope on days 0, 2, 4, 6, and 8. The relative length of each hair shaft was measured using the ImageJ software. (B) After 10days of culture, the HFs phase was assessed following the hair cycle scoring criteria. Representative images of the HFs for each experimental group are shown, as well as the calculated ratios of the hair cycle phases. (C) H&E staining and IHC staining of β- catenin and VEGF. The results are shown as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.
Journal of Microbiology and Biotechnology 2024; 34: 17-28https://doi.org/10.4014/jmb.2308.08013

Fig 5.

Figure 5.Effect of the LMWCP on anagen induction in 7-week-old female C57BL/6 mice. (A) Timetable of experimental treatments and sample collection. (B) Representative photographs of mouse back skin on days 0, 10, and 13. (C) Representative images of H&E-stained longitudinal and transverse sections of the skin of each mouse on day 13. Scale bar, 200 μm. (D) Skin color scores for 10 days. (E) Hair growth area on the back skin observed for 13 days. (F) Hair dermis thickness, (G) HF number, and (H) anagen/telogen ratios on day 13. (I) The expression levels of Wnt3a, β-catenin, PCNA, cyclin D1, and VEGF on the dorsal skin at day 13. The results are expressed as the mean ± standard deviation. *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001 compared to the control group.
Journal of Microbiology and Biotechnology 2024; 34: 17-28https://doi.org/10.4014/jmb.2308.08013

Fig 6.

Figure 6.LMWCP increases the expression of keratin in hDPCs and the dorsal skin of mice. (A) Western blotting analysis showing the total cytokeratin levels in hDPCs treated with 0, 0.3, 1, and 3 mg/ml of LMWCP for 24 h. (B–D) Western blotting and IHC analysis showing the total cytokeratin and Type I + II hair keratin levels in the dorsal skin of the exposed LMWCP (615 and 820 mg/kg) and MNX treatment and control mice after 13 days. Representative western blotting and IHC images from four independent experiments are shown. β-actin was used as a loading control. Scale bar, 400 μm. The results are expressed as the mean ± standard deviation. **, p < 0.01; ****, p < 0.0001 compared to the control group.
Journal of Microbiology and Biotechnology 2024; 34: 17-28https://doi.org/10.4014/jmb.2308.08013

Table 1 . Primer sequences used for quantification of gene expression..

GenePrimer sequence (5'→ 3')
Human EGFFCAGGGAAGATGACCACCACT
RCAGTTCCCACCACTTCAGGT
Human HB-EGFFACAAGGAGGAGCACGGGAAAAG
RCGATGACCAGCAGACAGACAGATG
Human FGF-4FGGGAGTCTACAGACAGCAAG
RGAGCCTAGGGTGTGGTTTA
Human FGF-6FGGGAGTCTACAGACAGCAAG
RGAGCCTAGGGTGTGGTTTA
Human ALPLFATTGACCACGGGCACCAT
RCTCCACCGCCTCATGCA
Human SHHFGCGCCAGCGGAAGGTAT
RCCGGTGTTTTCTTCATCCTTAAA
Human FGF7FATCAGGACAGTGGCAGTTGGA
RAACATTTCCCCTCCGTTGTGT
Human BMP-2FGAGGTCCTGAGCGAGTTCGA
RTCTCTGTTTCAGGCCGAACA

Table 2 . Antibodies used for Western blot analysis..

AntibodiesProduct codeCompany
Anti-MITFMAB3747-ISigma-Aldrich (MO, USA)
Anti-Calnexinab22595Abcam (Cambridge, UK)
Anti-ALIXab275377Abcam (Cambridge, UK)
Anti-CD63ab134045Abcam (Cambridge, UK)
Anti-tyrosinaseab180753Abcam (Cambridge, UK)
Anti-p-MITFab59201Abcam (Cambridge, UK)
Anti-TRP-1sc-58437Santa Cruz Biotechnology (CA, USA)
Anti-TRP-2sc-25544Santa Cruz Biotechnology (CA, USA)
Anti-Rab27asc-22756Santa Cruz Biotechnology (CA, USA)
Anti-β-actinsc-47778Santa Cruz Biotechnology (CA, USA)
Anti-p-CREB#9198Cell Signaling Technology Inc. (Beverly, MA)
Anti-CREB#9197Cell Signaling Technology Inc. (Beverly, MA)
Anti-p-ERK#9101Cell Signaling Technology Inc. (Beverly, MA)
Anti-ERK#9102Cell Signaling Technology Inc. (Beverly, MA)
Anti-p-AKT#4060Cell Signaling Technology Inc. (Beverly, MA)
Anti-AKT#4691Cell Signaling Technology Inc. (Beverly, MA)
Anti-p-β-catenin#4176Cell Signaling Technology Inc. (Beverly, MA)
Anti-β-catenin#8480Cell Signaling Technology Inc. (Beverly, MA)
Anti-Myosin-Va#3402Cell Signaling Technology Inc. (Beverly, MA)
Anti-MLPH10338-1-APProteinTech Group (IL, USA)

References

  1. Kaufman KD, Olsen EA, Whiting D, Savin R, DeVillez R, Bergfeld W, et al. 1998. Finasteride in the treatment of men with androgenetic alopecia. J. Am. Acad. Dermatol. 39: 578-589.
    Pubmed CrossRef
  2. Choi N, Shin S, Song SU, Sung J-H. 2018. Minoxidil promotes hair growth through stimulation of growth factor release from adipose-derived stem cells. Int. J. Mol. Sci. 19: 691.
    Pubmed KoreaMed CrossRef
  3. Sica DA. 2004. Minoxidil: an underused vasodilator for resistant or severe hypertension. J. Clin. Hypertens. 6: 283-287.
    Pubmed KoreaMed CrossRef
  4. Hirshburg JM, Kelsey PA, Therrien CA, Gavino AC, Reichenberg JS. 2016. Adverse effects and safety of 5-alpha reductase inhibitors (finasteride, dutasteride): a systematic review. J. Clin. Aesthet. Dermatol. 9: 56-62.
  5. Martino PA, Heitman N, Rendl M. 2021. The dermal sheath: an emerging component of the hair follicle stem cell niche. Exp. Dermatol. 30: 512-521.
    Pubmed KoreaMed CrossRef
  6. Yang CC, Cotsarelis G. 2010. Review of hair follicle dermal cells. J. Dermatol. Sci. 57: 2-11.
    Pubmed KoreaMed CrossRef
  7. Jahoda C, Horne K, Oliver R. 1984. Induction of hair growth by implantation of cultured dermal papilla cells. Nature 311: 560-562.
    Pubmed CrossRef
  8. Elliott K, Messenger AG, Stephenson TJ. 1999. Differences in hair follicle dermal papilla volume are due to extracellular matrix volume and cell number: implications for the control of hair follicle size and androgen responses. J. Investig. Dermatol. 113: 873-877.
    Pubmed CrossRef
  9. Taghiabadi E, Nilforoushzadeh MA, Aghdami N. 2020. Maintaining hair inductivity in human dermal papilla cells: a review of effective methods. Skin Pharmacol. Physiol. 33: 280-292.
    Pubmed CrossRef
  10. Kubanov A, Gallyamova YA, Korableva O, Kalinina P. 2017. The role of the VEGF, KGF, EGF, and TGF-Β1Growth factors in the pathogenesis of telogen effluvium in women. Biomed. Pharmacol. J. 10: 191-198.
    CrossRef
  11. Hardy MH. 1992. The secret life of the hair follicle. Trends Genet. 8: 55-61.
    Pubmed CrossRef
  12. Kishimoto J, Burgeson RE, Morgan BA. 2000. Wnt signaling maintains the hair-inducing activity of the dermal papilla. Genes Dev. 14: 1181-1185.
    CrossRef
  13. Collins CA, Kretzschmar K, Watt FM. 2011. Reprogramming adult dermis to a neonatal state through epidermal activation of β-catenin. Development 138: 5189-5199.
    Pubmed KoreaMed CrossRef
  14. Huelsken J, Vogel R, Erdmann B, Cotsarelis G, Birchmeier W. 2001. β-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell 105: 533-545.
    Pubmed CrossRef
  15. Deng Z, Chen M, Liu F, Wang Y, Xu S, Sha K, et al. 2022. Androgen receptor-mediated paracrine signaling induces regression of blood vessels in the dermal papilla in androgenetic alopecia. J. Investig. Dermatol. 142: 2088-2099.e2089.
    Pubmed CrossRef
  16. Dejana E. 2010. The role of wnt signaling in physiological and pathological angiogenesis. Circ. Res. 107: 943-952.
    Pubmed CrossRef
  17. Houschyar KS, Borrelli MR, Tapking C, Popp D, Puladi B, Ooms M, et al. 2020. Molecular mechanisms of hair growth and regeneration: current understanding and novel paradigms. Dermatology 236: 271-280.
    Pubmed CrossRef
  18. Kim H, Choi N, Kim DY, Kim SY, Song SY, Sung JH. 2021. TGF-β2 and collagen play pivotal roles in the spheroid formation and antiaging of human dermal papilla cells. Aging (Albany NY) 13: 19978.
    Pubmed KoreaMed CrossRef
  19. Williams R, Pawlus AD, Thornton MJ. 2020. Getting under the skin of hair aging: the impact of the hair follicle environment. Exp. Dermatol. 29: 588-597.
    Pubmed CrossRef
  20. Pyun HB, Kim M, Park J, Sakai Y, Numata N, Shin JY, et al. 2012. Effects of collagen tripeptide supplement on photoaging and epidermal skin barrier in UVB-exposed hairless mice. Prev. Nutr. Food Sci. 17: 245.
    Pubmed KoreaMed CrossRef
  21. Kim DU, Chung HC, Choi J, Sakai Y, Lee BY. 2018. Oral intake of low-molecular-weight collagen peptide improves hydration, elasticity, and wrinkling in human skin: a randomized, double-blind, placebo-controlled study. Nutrients 10: 826.
    Pubmed KoreaMed CrossRef
  22. Lee MH, Kim HM, Chung HC, Kim DU, Lee JH. 2021. Low-molecular-weight collagen peptide ameliorates osteoarthritis progression through promoting extracellular matrix synthesis by chondrocytes in a rabbit anterior cruciate ligament transection model. J. Microbiol. Biotechnol. 31: 1401-1408.
    Pubmed KoreaMed CrossRef
  23. Frantz C, Stewart KM, Weaver VM. 2010. The extracellular matrix at a glance. J. Cell Sci. 123: 4195-4200.
    Pubmed KoreaMed CrossRef
  24. Tang Y, Luo B, Deng Z, Wang B, Liu F, Li J, et al. 2016. Mitochondrial aerobic respiration is activated during hair follicle stem cell differentiation, and its dysfunction retards hair regeneration. PeerJ. 4: e1821.
    Pubmed KoreaMed CrossRef
  25. Lemasters JJ, Ramshesh VK, Lovelace GL, Lim J, Wright GD, Harland D, et al. 2017. Compartmentation of mitochondrial and oxidative metabolism in growing hair follicles: a ring of fire. J. Investig. Dermatol. 137: 1434-1444.
    Pubmed KoreaMed CrossRef
  26. Ohyama M. 2019. Use of human intra-tissue stem/progenitor cells and induced pluripotent stem cells for hair follicle regeneration. Inflamm. Regen. 39: 4.
    Pubmed KoreaMed CrossRef
  27. Fukuyama M, Tsukashima A, Kimishima M, Yamazaki Y, Okano H, Ohyama M. 2021. Human iPS cell-derived cell aggregates exhibited dermal papilla cell properties in in vitro three-dimensional assemblage mimicking hair follicle structures. Front. Cell Dev. Biol. 9: 590333.
    Pubmed KoreaMed CrossRef
  28. Kwack MH, Jang YJ, Won GH, Kim MK, Kim JC, Sung YK. 2019. Overexpression of alkaline phosphatase improves the hairinductive capacity of cultured human dermal papilla spheres. J. Dermatol. Sci. 95: 126-129.
    Pubmed CrossRef
  29. Nicu C, Wikramanayake TC, Paus R. 2020. Clues that mitochondria are involved in the hair cycle clock: MPZL3 regulates entry into and progression of murine hair follicle cycling. Exp. Dermatol. 29: 1243-1249.
    Pubmed CrossRef
  30. Vidali S, Chéret J, Giesen M, Haeger S, Alam M, Watson RE, et al. 2016. Thyroid hormones enhance mitochondrial function in human epidermis. J. Investig. Dermatol. 136: 2003-2012.
    Pubmed CrossRef
  31. Van Beek N, Bodo E, Kromminga A, Gáspár E, Meyer K, Zmijewski MA, et al. 2008. Thyroid hormones directly alter human hair follicle functions: anagen prolongation and stimulation of both hair matrix keratinocyte proliferation and hair pigmentation. J. Clin. Endocrinol. Metab. 93: 4381-4388.
    Pubmed CrossRef
  32. Choi N, Choi J, Kim JH, Jang Y, Yeo JH, Kang J, et al. 2018. Generation of trichogenic adipose-derived stem cells by expression of three factors. J. Dermatol. Sci. 92: 18-29.
    Pubmed CrossRef
  33. Peus D, Pittelkow MR. 1996. Growth factors in hair organ development and the hair growth cycle. Dermatol. Clin. 14: 559-572.
    Pubmed CrossRef
  34. Alexandrescu DT, Kauffman CL, Dasanu CA. 2009. Persistent hair growth during treatment with the EGFR inhibitor erlotinib. Dermatol. Online J. 15: 4.
    CrossRef
  35. Yano K, Brown LF, Detmar M. 2001. Control of hair growth and follicle size by VEGF-mediated angiogenesis. J. Clin. Investig. 107: 409-417.
    Pubmed KoreaMed CrossRef
  36. Gentile P. 2019. Autologous cellular method using micrografts of human adipose tissue derived follicle stem cells in androgenic alopecia. Int. J. Mol. Sci. 20: 3446.
    Pubmed KoreaMed CrossRef
  37. Back SH, Yoon JB, Sim WY, Haw CR. 1999. Effects of vaseular endothelial growth factors on hair growth in vitro. Korean J. Dermatol. 37: 23-30.
  38. Katzer T, Leite Junior A, Beck R, da Silva C. 2019. Physiopathology and current treatments of androgenetic alopecia: going beyond androgens and anti‐androgens. Dermatol. Ther. 32: e13059.
    Pubmed CrossRef
  39. Paus R, Cotsarelis G. 1999. The biology of hair follicles. New Eng. J. Med. 341: 491-497.
    Pubmed CrossRef
  40. Kumar N, Rungseevijitprapa W, Narkkhong NA, Suttajit M, Chaiyasut C. 2012. 5α-reductase inhibition and hair growth promotion of some Thai plants traditionally used for hair treatment. J. Ethnopharmacol. 139: 765-771.
    Pubmed CrossRef
  41. Müller-Röver S, Foitzik K, Paus R, Handjiski B, van der Veen C, Eichmüller S, et al. 2001. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J. Investig. Dermatol. 117: 3-15.
    Pubmed CrossRef
  42. Piccolo M, Ferraro MG, Maione F, Maisto M, Stornaiuolo M, Tenore GC, et al. 2019. Induction of hair keratins expression by an annurca apple-based nutraceutical formulation in human follicular cells. Nutrients 11: 3041.
    Pubmed KoreaMed CrossRef
  43. An SY, Kim HS, Kim SY, Van SY, Kim HJ, Lee JH, et al. 2022. Keratin-mediated hair growth and its underlying biological mechanism. Commun. Biol. 5: 1270.
    Pubmed KoreaMed CrossRef