Palmitoyl‑RGD promotes the expression of dermal‑epidermal junction components in HaCaT cells

  • Authors:
    • Joo Hyuck Lim
    • Jung Soo Bae
    • Seung Ki Lee
    • Dong Hun Lee
  • View Affiliations

  • Published online on: August 29, 2022     https://doi.org/10.3892/mmr.2022.12836
  • Article Number: 320
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

With age, the dermal‑epidermal junction (DEJ) becomes thinner and production of its protein components decreases; this may be associated with increased fragility and wrinkling of skin. Topical treatment with palmitoyl‑Arg‑Gly‑Asp (PAL‑RGD) improves facial wrinkles, skin elasticity and dermal density in humans. In the present study, the effect of PAL‑RGD on expression of DEJ components, such as laminin and collagen, was assessed. Human HaCaT keratinocytes were treated with PAL‑RGD. The protein expression levels of laminin‑332, collagen IV and collagen XVII were examined by western blotting. Reverse transcription-quantitative PCR was used to analyze laminin subunit (LAM)A3, LAMB3, LAMC2, collagen type IV α 1 chain (COL4A1) and COL17A1 mRNA expression levels. Western blot analysis showed that the expression levels of proteins comprising the DEJ, including laminin α3, β3 and γ2 and collagen IV and XVII demonstrated a significant dose‑dependent increase following PAL‑RGD treatment. Furthermore, PAL‑RGD treatment significantly enhanced LAMA3, LAMB3, LAMC2, COL4A1 and COL17A1 mRNA expression levels. PAL‑RGD may enhance the DEJ by inducing the expression of laminin‑332, collagen IV and collagen XVII.

Introduction

The dermal-epidermal junction (DEJ), located between the epidermis and dermis, provides a specific niche that mediates numerous signals such as MAPK in addition to providing structural support to keratinocytes (1,2). The DEJ serves an important role in skin cohesion, resistance to mechanical stress and exchange of the signals between the dermis and epidermis (3). As skin ages, the wavelike structure of the DEJ becomes thinner and appears flattened with a loss of rete ridges (46). Furthermore, previous studies have reported that production of protein components of the DEJ, including collagen IV, VII and XVII, nidogen, integrin β4 and laminin-332, decreases with age (69). Laminins are the most abundant glycoproteins of the basement membrane (BM) extracellular matrix (ECM) and are key for supporting tissue architecture and stability (10). Laminins are involved in skin reepithelization and wound healing via regulation of adhesion, proliferation, migration, apoptosis and differentiation (11). Collagen IV is a primary component of anchoring fibrils, which provide mechanical support for keratinocytes (12). Collagen XVII, another structural component of anchoring fibrils, serves important roles in assembly and function of the cell-matrix adhesion structure, signal transduction and keratinocyte differentiation (13). Based on the key roles of the DEJ in skin homeostasis, modulation of DEJ components has been suggested as a potential strategy to mitigate skin aging (1416).

Our previous study demonstrated that palmitoyl-Arg-Gly-Asp (PAL-RGD) decreases the appearance of human facial wrinkles, skin elasticity and dermal density via stimulation of procollagen synthesis and inhibition of matrix metalloproteinase (MMP)-1 expression in human dermal fibroblasts (17). Although collagen degradation within the dermis is considered the primary cause of wrinkles, biological changes in the epidermis and DEJ, such as decreased collagen IV, collagen VII, collagen XVII, integrin β4, and laminin-332, are also reported to affect wrinkle formation (6). In the present study, the effects of PAL-RGD on expression of DEJ components, including laminin-332, collagen IV and collagen XVII in HaCaT keratinocytes was investigated.

Materials and methods

Reagents

PAL-RGD was purchased from Celltrion Chemical Research Institute. MTT was purchased from Sigma-Aldrich (Merck KGaA). Dulbecco's Modified Eagle's Medium (DMEM), penicillin, streptomycin, PBS, fetal bovine serum (FBS) and trypsin/EDTA were purchased from Gibco (Thermo Fisher Scientific, Inc.).

Cell culture

The human keratinocyte HaCaT cell line, kindly gifted by Dr Norbert Fusenig of the German Cancer Research Center (Heidelberg, Germany), was cultured in DMEM supplemented with 10% FBS and 1% penicillin/streptomycin at 37°C in a humidified atmosphere with 5% CO2.

Cell viability assay

Cell viability was determined using MTT assay. Briefly, HaCaT keratinocytes were seeded into a 24-well plate at 5×104 cells/well and incubated at 37°C for 24 h. Following serum starvation at 37°C for 24 h, cells were treated with PAL-RGD (0, 1, 5, 10, 15 and 20 µg/ml) for 48 h in fresh serum-free DMEM at 37°C. MTT was added and cells were incubated at 37°C for 4 h to allow reduction of the MTT reagent to formazan dissolved with DMSO. The absorbance was measured at 570 nm using a VersaMax microplate reader (Molecular Devices, LLC).

Western blotting

HaCaT cells were seeded into a 6-well plate at 1×106 cells/well and incubated at 37°C for 24 h. Following serum starvation at 37°C for 24 h, the cells were treated with PAL-RGD (0.0, 5.0, 7.5, 10.0, 12.5 and 15.0 µg/ml) at 37°C for 48 h in fresh serum-free DMEM. Cell lysates were prepared using PRO-PREP Protein Extraction Solution (Intron Biotechnology, Inc.) and conditioned medium was collected. Protein concentration was determined with the BCA method (Pierce Biotechnology). The 20 µg protein from cell lysate and conditioned media were separated on 4–15% Mini-PROTEAN® TGX Precast Gels (Bio-Rad Laboratories, Inc.) and transferred to a nitrocellulose membrane. The membranes were blocked with 5% skimmed milk in Tris-buffered saline for 1 h at room temperature and incubated with primary antibodies overnight at 4°C against laminin α3 (1:500; cat. no. ab151715; Abcam), laminin β3 (1:500; cat. no. sc-133178; Santa Cruz Biotechnology, Inc.), laminin γ2 (1:500; cat. no. MAB19562; MilliporeSigma), collagen type IV (COL4; 1:500; cat. no. AB769; MilliporeSigma), COL17 (1:500; cat. no. ab184996; Abcam) and β-tubulin (1:1,000; cat. no. SC-9104; Santa Cruz Biotechnology, Inc.). After rinsing with TBS + 0.05% Tween-20, the membranes were incubated at room temperature for 2 h with horseradish peroxidase-conjugated anti-goat (cat. no. #605-4302), anti-mouse (cat. no. #610-4302) and anti-rabbit antibodies (all 1:5,000, #611-1302, all Rockland Immunochemicals, Inc.). Membranes were developed with ECL solution (SuperSignal West Femto Maximum Sensitivity Substrate, Thermo Fisher Scientific, Inc.). Signals were visualized using the ChemiDoc XRS system (Bio-Rad Laboratories, Inc.) and analyzed with Image Lab 5.0 (Bio-Rad Laboratories, Inc.).

Reverse transcription-quantitative (RT-q)PCR

Cells were incubated with PAL-RGD (0.0, 5.0, 7.5, 10.0, 12.5 and 15.0 µg/ml) for 2, 4, 8 and 24 h. Total RNA was isolated from cells using RNeasy Mini Kit (Qiagen GmbH) according to the manufacturer's protocol. First-strand complementary DNA (cDNA) synthesis using 1 µg total RNA was performed using SuperScript IV First-Strand Synthesis System (Thermo Fisher Scientific, Inc.). qPCR was performed using SYBR Green and StepOnePlus Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). The final reaction mixture contained 10 ng cDNA, 100 nmol each primer, 10 µl Power SYBR Green PCR Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) and RNase-free water to a final volume of 20 µl. PCR was performed an initial denaturation step at 95°C for 10 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min. Dissociation curve was generated to verify the specificity of each reaction. Data were analyzed using the 2−ΔΔCq method and expressed as percent change in gene expression relative to 36B4 (18). The primer sequences are presented in Table I.

Table I.

Primer sequences used for reverse transcription-quantitative PCR.

Table I.

Primer sequences used for reverse transcription-quantitative PCR.

GeneSequence, 5′-3′
LAMA3F: CAATTCTGAAGACCCCAGGA
R: TCCAAGACCCAAAGATCAGG
LAMB3F: CAGATTGGGTTGGAATGCTT
R: CCCAGCTTCCTTGACTTGAG
LAMC2F: GGCTGGTCTTACTGGAGCAG
R: TATGGCAGCTTCACTGTTGC
Col4A1F: CTGGTCCAAGAGGATTTCCA
R: TCATTGCCTTGCACGTAGAG
COL17A1F: TCTGCCTACAGCAACGTGAC
R: CTCACGGCTTGACAGCAATA
36B4F: TCGACAATGGCAGCATCTAC
R: TGATGCAACAGTTGGGTAGC

[i] LAM, laminin subunit; COL4A1, collagen type IV α 1 chain; COL17A1, collagen type XVII α 1 chain; F, forward; R, reverse.

Statistical analysis

Data are presented as the mean ± SD of ≥3 independent experiments. Statistical analysis was performed using SAS 9.4 (SAS Institute, Inc.) software. Statistical significance was calculated using one-way ANOVA followed by two-tailed post hoc Dunnett's multiple comparisons test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of PAL-RGD on viability in HaCaT cells

MTT assay was performed to determine optimal PAL-RGD concentration for treatment of cells. PAL-RGD did not induce a significant effect on cell viability at concentrations <15 µg/ml. The cell viability significantly decreased to 67% only at 20 µg/ml Pal-RGD (Fig. 1). For subsequent experiments, the maximum concentration used was 15 µg/ml.

Effect of PAL-RGD on DEJ protein expression in HaCaT cells

HaCaT cells were treated with PAL-RGD for 48 h. Total protein extracted from cell lysate and conditioned media was analyzed using western blotting. PAL-RGD dose-dependently increased the expression of DEJ proteins (Fig. 2). Treatment with 15 µg/ml PAL-RGD significantly increased protein expression of laminin α3 (167%), laminin β3 (160%), laminin γ2 (209%), collagen IV (141%) and collagen XVII (156%) compared with the control.

Effect of PAL-RGD on laminin subunit (LAM)A3, LAMB3 and LAMC2 mRNA expression in HaCaT cells

Treatment with PAL-RGD enhanced mRNA expression levels of LAMA3, LAMB3 and LAMC2 in HaCaT cells (Fig. 3). LAMA3 mRNA expression increased in a dose-dependent manner at 2, 4 and 8 h, then decreased to the basal level at 24 h. LAMA3 mRNA expression levels significantly increased at 4 h for all concentrations of PAL-RGD compared with the control. LAMB3 mRNA expression was increased in a dose-dependent manner at 2 h prior to dropping to the basal level at 8 h. The 2 h increase of LAMB3 mRNA expression was significant from 7.5 to 15 µg/ml of PAL-RGD used compared with the control. Furthermore, mRNA expression of LAMC2 significantly increased 2 h after PAL-RGD treatment. At 2 h increase of LAMC3 mRNA expression was significantly for all concentrations of PAL-RGD used compared with the control. The maximum increases in LAMA3, LAMB3 and LAMC2 in response to 15 µg/ml PAL-RGD treatment were significant by 201% (4 h, P<0.0001), 146% (2 h, P=0.0003) and 460% (4 h, P<0.0001) respectively, compared with the control.

Effect of PAL-RGD on COL4 α 1 chain (COL4A1) and COL17A1 mRNA expression in HaCaT cells

The mRNA expression of COL4A1 and COL17A1 was increased by PAL-RGD treatment in HaCaT cells (Fig. 4). Although unchanged following exposure to PAL-RGD for 2 and 8 h, mRNA expression levels increased in a dose-dependent manner at 24 h. Treatment with 12.5 µg/ml PAL-RGD significantly induced COL4A1 and COL17A1 mRNA levels by 146 and 136%, respectively. The mRNA expression of COL4A1 and COL17A1 in HaCaT cells treated with 15 µg/ml PAL-RGD significantly increased by 153 and 136%, respectively, compared with the control.

Discussion

In the present study, PAL-RGD treatment increased both mRNA and protein expression levels of DEJ components, such as laminin-332, collagen IV and collagen XVII, in HaCaT keratinocytes. The mRNA expression levels of laminin-332 components increased before 8 h and returned to normal at 24 h; however, protein expression levels in media were significantly increased at 48 h. Laminin-332 is a secreted protein that matures via specific proteolytic processing, which may contribute to the time lag between mRNA expression in cells and protein accumulation in media (19) Previous studies have reported that transforming growth factor-β (TGF-β) upregulates expression of LAMA3, LAMB3, LAMC2, COL4A1 and COL17A1 (2022). PAL-RGD may induce TGF-β signaling; further studies are required to investigate the signaling pathway underlying the PAL-RGD-mediated upregulation of laminin-332, collagen IV and collagen XVII.

Laminins are glycoproteins composed of three chains (α, β and γ). At least 15 different isoforms exist with a combination of five distinct α, three β and three γ subunits (23,24). The functional domains of LAMs serve distinct roles, such as assembly into trimeric molecules, binding to other ECM molecules and interaction with cell surface receptors. Laminins are involved in various cellular functions, such as cell adhesion, migration, proliferation and differentiation (25). In particular, laminin-332 activates the signaling through integrin α3β1 and α6β4 (26). The binding of laminin-332 and integrin α3β1 activates the mitogen-activated protein kinase signaling pathway, which regulates keratinocyte proliferation (27). Moreover, the interaction between laminin-332 and integrin α3β1 regulates Src kinase signaling through focal adhesion kinase, promoting polarized lamellipodium extension, which regulates keratinocyte migration (28). The interaction between laminin-332 and integrin α6β4 mediates assembly of the hemidesmosome cytoskeleton and recruitment of adaptor proteins Shc/growth factor receptor-bound protein 2 (29). Therefore, PAL-RGD may promote proliferation and migration of keratinocytes by increasing laminin-332 expression.

Collagen IV is a non-fibrillar collagen that constitutes ~50% of the BM, forming a structural scaffold for interactions between BM components, such as laminin networks, perlecan and proteoglycans, during BM assembly (30,31). Other extracellular molecules, including growth factors, laminins, proteoglycans and nidogens, are attached to the scaffolds, where they serve various biological roles, such as cell adhesion, migration, tissue regeneration, wound healing, immobilization of growth factors and enzymes and molecular sieving (32,33). Given these features of collagen and related molecules, PAL-RGD may strengthen BM scaffolds by promoting synthesis of collagen IV.

Collagen XVII is a hemidesmosomal transmembrane protein involved in epidermal-dermal attachment (34). Congenital collagen XVII deficiency results in junctional epidermolysis bullosa and acquired autoimmunity to collagen XVII leads to bullous pemphigoid (35). Furthermore, collagen XVII binds to laminin-332 in hemidesmosomes and collagen IV in the ECM, linking cytoplasmic structural components with ECM (36). Based on these functions of collagen XVII, PAL-RGD may enhance BM assembly by inducing collagen XVII synthesis.

In summary, the present study suggested that PAL-RGD may serve a role in strengthening the DEJ by promoting expression of laminin-332, collagen IV and collagen XVII in keratinocytes. Together with previous studies demonstrating that PAL-RGD decreases facial wrinkles by increasing collagen I and decreasing MMP-1, this compound could improve skin aging by increasing production of components of the DEJ and BM. A limitation of the present study was lack of data on the biological effect of PAL-RGD on the function of the DEJ. Further studies are needed to determine the biological effect of PAL-RGD on the function of the DEJ.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JHL conceived and designed the study, performed experiments, collected the data and wrote the manuscript. JSB designed the study, performed experiments and revised the manuscript. JHL and JSB confirm the authenticity of all the raw data. SKL conceived the study and revised the manuscript. DHL interpreted the data and revised the manuscript. All authors discussed the results. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Burgeson RE and Christiano AM: The dermal-epidermal junction. Curr Opin Cell Biol. 9:651–658. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Roig-Rosello E and Rousselle P: The human epidermal basement membrane: A shaped and cell instructive platform that aging slowly alters. Biomolecules. 10:16072020. View Article : Google Scholar : PubMed/NCBI

3 

Marionnet C, Vioux-Chagnoleau C, Pierrard C, Sok J, Asselineau D and Bernerd F: Morphogenesis of dermal-epidermal junction in a model of reconstructed skin: Beneficial effects of vitamin C. Exp Dermatol. 15:625–633. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Lavker RM, Zheng P and Dong G: Aged skin: A study by light, transmission electron, and scanning electron microscopy. J Invest Dermatol. 88 (3 Suppl):44s–51s. 1987. View Article : Google Scholar : PubMed/NCBI

5 

Vázquez F, Palacios S, Alemañ N and Guerrero F: Changes of the basement membrane and type IV collagen in human skin during aging. Maturitas. 25:209–215. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Langton AK, Halai P, Griffiths CE, Sherratt MJ and Watson RE: The impact of intrinsic ageing on the protein composition of the dermal-epidermal junction. Mech Ageing Dev. 156:14–16. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Mondon P, Hillion M, Peschard O, Andre N, Marchand T, Doridot E, Feuilloley MG, Pionneau C and Chardonnet S: Evaluation of dermal extracellular matrix and epidermal-dermal junction modifications using matrix-assisted laser desorption/ionization mass spectrometric imaging, in vivo reflectance confocal microscopy, echography, and histology: Effect of age and peptide applications. J Cosmet Dermatol. 14:152–160. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Contet-Audonneau JL, Jeanmaire C and Pauly G: A histological study of human wrinkle structures: Comparison between sun-exposed areas of the face, with or without wrinkles, and sun-protected areas. Br J Dermatol. 140:1038–1047. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Varlet BL, Chaudagne C, Barré P, Sauvage C, Berthouloux B, Meybeck A, Dumas M, Saunois A and Bonté F: Age-related functional and structural changes in human dermo-epidermal junction components. Journal of Investigative Dermatology Symposium Proceedings. Vol. 3. Elsevier; pp. 172–179. 1998, View Article : Google Scholar : PubMed/NCBI

10 

Ishihara J, Ishihara A, Fukunaga K, Sasaki K, White MJV, Briquez PS and Hubbell JA: Laminin heparin-binding peptides bind to several growth factors and enhance diabetic wound healing. Nat Commun. 9:21632018. View Article : Google Scholar : PubMed/NCBI

11 

Iorio V, Troughton LD and Hamill KJ: Laminins: Roles and utility in wound repair. Adv Wound Care (New Rochelle). 4:250–263. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Chung HJ and Uitto J: Type VII collagen: The anchoring fibril protein at fault in dystrophic epidermolysis bullosa. Dermatol Clin. 28:93–105. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Watanabe M, Natsuga K, Nishie W, Kobayashi Y, Donati G, Suzuki S, Fujimura Y, Tsukiyama T, Ujiie H, Shinkuma S, et al: Type XVII collagen coordinates proliferation in the interfollicular epidermis. Elife. 6:e266352017. View Article : Google Scholar : PubMed/NCBI

14 

Jeong S, Yoon S, Kim S, Jung J, Kor M, Shin K, Lim C, Han HS, Lee H, Park KY, et al: Anti-wrinkle benefits of peptides complex stimulating skin basement membrane proteins expression. Int J Mol Sci. 21:732019. View Article : Google Scholar : PubMed/NCBI

15 

Amano S: Possible involvement of basement membrane damage in skin photoaging. Journal of Investigative Dermatology Symposium Proceedings. Vol. 14. Elsevier; pp. 2–7. 2009, View Article : Google Scholar : PubMed/NCBI

16 

Kusumaningrum N, Oh JH, Lee DH, Shin CY, Jang JH, Kim YK and Chung JH: Topical treatment with a cathepsin G inhibitor, β-keto-phosphonic acid, blocks ultraviolet irradiation-induced basement membrane damage in hairless mouse skin. Photodermatol Photoimmunol Photomed. 35:148–156. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Bae JS, Kim JM, Kim JY, Choi CH, Kim JY, Moon WK, Lee MS, Moon SH, Lim JH, Park SJ, et al: Topical application of palmitoyl-RGD reduces human facial wrinkle formation in Korean women. Arch Dermatol Res. 309:665–671. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Peters BP, Hartle RJ, Krzesicki RF, Kroll TG, Perini F, Balun JE, Goldstein IJ and Ruddon RW: The biosynthesis, processing, and secretion of laminin by human choriocarcinoma cells. J Biol Chem. 260:14732–1442. 1985. View Article : Google Scholar : PubMed/NCBI

20 

Korang K, Christiano AM, Uitto J and Mauviel A: Differential cytokine modulation of the genes LAMA3, LAMB3, and LAMC2, encoding the constitutive polypeptides, alpha 3, beta 3, and gamma 2, of human laminin 5 in epidermal keratinocytes. FEBS Lett. 368:556–558. 1995. View Article : Google Scholar : PubMed/NCBI

21 

Feru J, Delobbe E, Ramont L, Brassart B, Terryn C, Dupont-Deshorgue A, Garbar C, Monboisse JC, Maquart FX and Brassart-Pasco S: Aging decreases collagen IV expression in vivo in the dermo-epidermal junction and in vitro in dermal fibroblasts: Possible involvement of TGF-β1. Eur J Dermatol. 26:350–360. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Huilaja L, Hurskainen T, Autio-Harmainen H, Hofmann SC, Sormunen R, Räsänen J, Ilves M, Franzke CW, Bruckner-Tuderman L and Tasanen K: Pemphigoid gestationis autoantigen, transmembrane collagen XVII, promotes the migration of cytotrophoblastic cells of placenta and is a structural component of fetal membranes. Matrix Biol. 27:190–200. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Sugawara K, Tsuruta D, Ishii M, Jones JC and Kobayashi H: Laminin-332 and −511 in skin. Exp Dermatol. 17:473–480. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Miner JH and Yurchenco PD: Laminin functions in tissue morphogenesis. Annu Rev Cell Dev Biol. 20:255–284. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Tsuruta D, Kobayashi H, Imanishi H, Sugawara K, Ishii M and Jones JC: Laminin-332-integrin interaction: A target for cancer therapy? Curr Med Chem. 15:1968–1975. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Kariya Y and Gu J: Roles of laminin-332 and alpha6beta4 integrin in tumor progression. Mini Rev Med Chem. 9:1284–1291. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Gonzales M, Haan K, Baker SE, Fitchmun M, Todorov I, Weitzman S and Jones JC: A cell signal pathway involving laminin-5, alpha3beta1 integrin, and mitogen-activated protein kinase can regulate epithelial cell proliferation. Mol Biol Cell. 10:259–270. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Choma DP, Milano V, Pumiglia KM and DiPersio CM: Integrin alpha3beta1-dependent activation of FAK/Src regulates Rac1-mediated keratinocyte polarization on laminin-5. J Invest Dermatol. 127:31–40. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Mainiero F, Pepe A, Wary KK, Spinardi L, Mohammadi M, Schlessinger J and Giancotti F: Signal transduction by the alpha 6 beta 4 integrin: Distinct beta 4 subunit sites mediate recruitment of Shc/Grb2 and association with the cytoskeleton of hemidesmosomes. EMBO J. 14:4470–4481. 1995. View Article : Google Scholar : PubMed/NCBI

30 

Pöschl E, Schlötzer-Schrehardt U, Brachvogel B, Saito K, Ninomiya Y and Mayer U: Collagen IV is essential for basement membrane stability but dispensable for initiation of its assembly during early development. Development. 131:1619–1628. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Brown KL, Cummings CF, Vanacore RM and Hudson BG: Building collagen IV smart scaffolds on the outside of cells. Protein Sci. 26:2151–2161. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Good MC, Zalatan JG and Lim WA: Scaffold proteins: Hubs for controlling the flow of cellular information. Science. 332:680–686. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Trappmann B, Gautrot JE, Connelly JT, Strange DG, Li Y, Oyen ML, Cohen Stuart MA, Boehm H, Li B, Vogel V, et al: Extracellular-matrix tethering regulates stem-cell fate. Nat Mater. 11:642–649. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Nishimura M, Nishie W, Shirafuji Y, Shinkuma S, Natsuga K, Nakamura H, Sawamura D, Iwatsuki K and Shimizu H: Extracellular cleavage of collagen XVII is essential for correct cutaneous basement membrane formation. Hum Mol Genet. 25:328–339. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Natsuga K, Watanabe M, Nishie W and Shimizu H: Life before and beyond blistering: The role of collagen XVII in epidermal physiology. Exp Dermatol. 28:1135–1141. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Kamaguchi M, Iwata H, Nishie W, Toyonaga E, Ujiie H, Natsuga K, Kitagawa Y and Shimizu H: The direct binding of collagen XVII and collagen IV is disrupted by pemphigoid autoantibodies. Lab Invest. 99:48–57. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2022
Volume 26 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lim JH, Bae JS, Lee SK and Lee DH: Palmitoyl‑RGD promotes the expression of dermal‑epidermal junction components in HaCaT cells. Mol Med Rep 26: 320, 2022
APA
Lim, J.H., Bae, J.S., Lee, S.K., & Lee, D.H. (2022). Palmitoyl‑RGD promotes the expression of dermal‑epidermal junction components in HaCaT cells. Molecular Medicine Reports, 26, 320. https://doi.org/10.3892/mmr.2022.12836
MLA
Lim, J. H., Bae, J. S., Lee, S. K., Lee, D. H."Palmitoyl‑RGD promotes the expression of dermal‑epidermal junction components in HaCaT cells". Molecular Medicine Reports 26.4 (2022): 320.
Chicago
Lim, J. H., Bae, J. S., Lee, S. K., Lee, D. H."Palmitoyl‑RGD promotes the expression of dermal‑epidermal junction components in HaCaT cells". Molecular Medicine Reports 26, no. 4 (2022): 320. https://doi.org/10.3892/mmr.2022.12836