Inhibitory effects of TGP on KGF‑induced hyperproliferation of HaCaT cells via suppression of the p38 MAPK/NF‑κB p65 pathway

  • Authors:
    • Weirong Pang
    • Xiaoming Qi
    • Chunmei Cao
    • Shuosheng Zhang
  • View Affiliations

  • Published online on: June 15, 2018     https://doi.org/10.3892/mmr.2018.9177
  • Pages: 2207-2215
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Psoriasis is a chronic inflammatory skin disease, primarily caused by overgrowth and abnormal differentiation of epidermal keratinocytes. Studies have suggested that keratinocyte growth factor (KGF) may be involved in the regulation of differentiation and development of keratinocytes. Total glucosides of peony (TGP) have been widely used for the treatment of psoriasis. The present study aimed to determine whether the therapeutic effect of TGP on psoriasis is mediated by modulation of the p38 mitogen‑activated protein kinase (p38 MAPK)/nuclear factor (NF)‑κB p65 signaling pathways. Cell proliferation was evaluated by CCK‑8 and cell cycle was assessed by flow cytometry assay. Protein and mRNA expression of genes were determined by western blot and reverse transcription‑quantitative polymerase chain reaction, respectively. The results of the present study demonstrated that KGF can promote proliferation of HaCaT cells in a dose‑dependent manner. In addition, it was demonstrated that TGP may suppress the hyperproliferation of HaCaT cells stimulated by KGF by inducing arrest of the cell cycle at the G1 phase. The expression levels of the proinflammatory cytokines interleukin (IL)‑22 and vascular endothelial growth factor (VEGF) were markedly elevated in cells treated with KGF, whereas they were downregulated in cells treated with TGP. Furthermore, combination treatments with p38 MAPK inhibitor SB203580 and KGF, or TGP and KGF suppressed the mRNA and protein expression levels of IL‑22 and VEGF, compared with cells treated with KGF alone. Furthermore, the expression profiles of phosphorylated‑p38 MAPK and NF‑κB p65 were similar to those of IL‑22 and VEGF. The results of the present study suggested that the therapeutic effect of TGP on psoriasis may be mediated by modulation of the p38 MAPK/NF‑κB p65 signaling pathway. The results of the present study contribute to the understanding of the role of TGP in the treatment of psoriasis. The present study provides insights suggesting that p38 MAPK may be a novel regulatory signaling pathway for the treatment of psoriasis.

Introduction

Psoriasis is a chronic inflammatory skin disease characterized by erythema, pimples, patches and silver scales (1). It has been demonstrated that psoriasis is a disorder resulting from a combination of mutations in multiple genes and interaction between numerous other factors, including trauma, infection and drugs (2). The primary pathological features of psoriasis include overgrowth and abnormal differentiation of epidermal keratinocytes, expansion of dermal papillary capillaries and infiltration of inflammatory cells, among which keratinocyte dysfunction serves a role in the pathogenesis of psoriasis (2,3).

Keratinocytes are able to secrete a number of cytokines, resulting in a local inflammatory response (4,5). Upregulation of cytokine expression further stimulates the keratinocytes and aggravates the inflammatory response of the skin (4,6). A complex immune network formed by keratinocytes and cytokines serves a role in psoriasis. Previous studies demonstrated that psoriasis is initiated when keratinocytes are subjected to a variety of stressors, including infection, pregnancy and trauma (4,7,8). These stressors result inactivation of the immune system, secretion of a number of immune-associated cytokines and chemokines, and induction of inflammatory responses (4). Cytokines are additionally produced by activated T cells and dendritic cells (9). Therefore, the formation of psoriatic lesions results from an interaction between the activation of skin T cells and stimulation of keratinocytes (10).

Keratinocyte growth factor (KGF) is a member of the fibroblast growth factor family, and is additionally termed fibroblast growth factor-7 (11). KGF is a paracrine growth factor secreted by various interstitial cells, including fibroblasts, endothelial cells, smooth muscle cells and dendritic epidermal T cells (1113). KGF interacts with a specific receptor, fibroblast growth factor receptor 2 IIIb on epidermal cells (13). Although KGF is secreted by interstitial cells, it targets epidermal cells. A previous study demonstrated that the expression of KGF was markedly increased following trauma in mice and humans (14). In addition, cytological studies have demonstrated that KGF is able to specifically promote the proliferation and migration of keratinocytes (14,15). These studies suggested that KGF may serve a role in wound healing and in promoting the proliferation of keratinocytes (15).

White peony is the dry root of plants in the Ranunculaceae (crowfoot family), and its primary therapeutic ingredient is a group of substances collectively termed total glucosides of peony (TGP), which include peony bitter, hydroxyl paeoniflorin, benzoyl and paeoniflorin, which accounts for >90% of all the ingredients (16,17). Clinical application and pharmacological studies have demonstrated that TGP exhibit anti-inflammatory, analgesic and immune regulatory effects, protect the liver and vascular endothelium against inflammation (17,18). TGP is used for the treatment of a variety of autoimmune diseases, including psoriasis (19), systemic lupus erythematosus (19,20) and ankylosing spondylitis (21). A number of studies have suggested that TGP is involved in numerous aspects of autoimmune processes (1). The expression levels of interleukin (IL)-2 and −4 are reduced and elevated, respectively, in dinitrochlorobenzene-induced chronic dermatitis-eczema models (22). TGP are involved in the G protein-adenylate cyclase-cyclic adenosine monophosphate (cAMP) signal transduction pathway (23). It has additionally been demonstrated that TGP may downregulate the expression of inflammatory cytokines, including tumor necrosis factor-α (TNF-α), IL-6, IL-8 and interferon (IFN)-α (24).

A previous study reported that KGF may regulate the expression of the proinflammatory mediators TNF-α and IL-1 to stimulate inflammatory responses via activation of p38 mitogen-activated protein kinase (p38 MAPK) (25). The therapeutic effects of TGP may be associated with the regulation of p38 MAPK. Hyperproliferative HaCaT cells exhibit characteristics of keratinocytes in psoriatic lesions, and are therefore used as in vitro models of psoriasis in drug research (26). Therefore, in order elucidate the underlying mechanism of action of TGP in the treatment of psoriasis, the effects of TGP on the proliferation of HaCaT cells induced by KGF were determined. The p38 MAPK signaling pathway was selected to elucidate the molecular mechanism of TGP.

Materials and methods

Cell culture

The immortal human keratinocyte line HaCaT was purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The HaCaT cells were cultured in Dulbecco's modified Eagle medium (DMEM; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 5% fetal bovine serum, 100 U/ml penicillin and 100 µg/ml streptomycin, and were incubated in a cell culture chamber at 37°C in a humidified atmosphere containing 5% CO2. Cells were grown to 80% confluence, the culture medium was removed and washed with PBS, and the cells were digested using 0.25% trypsin. Cells in the logarithmic growth phase were used for the subsequent experiments.

Cell experiments

Confluent cultures of HaCaT cells were incubated with DMEM supplemented with 0, 1.25, 2.5, 5, 10, 20 and 40 ng/ml KGF (Prospec-Tany TechnoGene, Ltd., East Brunswick, NJ, USA) for 24, 48 and 72 h at 37°C. The cell hyperproliferation model of psoriasis was established in HaCaT cells via treatment with KGF (10 ng/ml) for 24 h at 37°C and confirmed using a Cell Counting Kit-8 (CCK-8) assay. To investigate the effect of TGP on hyperproliferative HaCaT cells induced by KGF, cell proliferation and the cell cycle were detected for these cells following treatment with different concentrations of TGP: i) TGP-low (L; 10 mg/l); ii) TGP-medium (M; 50 mg/l); and iii) TGP-high (H; 120 mg/l) or 48 h at 37°C. Subsequently, to determine the role of TGP, cells were divided into five groups: i) Model, cells treated with 10 ng/ml KGF; ii) TGP, cells treated with 120 mg/lTGP; iii) control, cells with no treatment; iv) KGF-IN [cells treated with 10 ng/ml KGF+10 µm SB203580 (Calbiochem; Merck KGaA)]; and v) KGF-TGP (cells treated with 10 ng/ml KGF+120 mg/lTGP). Following incubation for 48 h at 37°C, cells were harvested and used for subsequent experimentation as described below.

Detection of cell proliferation activity by CCK-8 assay

To determine alterations in cell proliferation, HaCaT cells from all groups were seeded in 96-well plates at a density of 1×105 cells/well. Cell proliferation was detected using a CCK-8 kit (Dojindo Molecular Technologies, Inc., Kumamoto, Japan), according to the manufacturer's instructions. A total of 20 µl CCK-8 was added and cells were incubated at 37°C for 4 h. The optical density values were measured at a wavelength of 450 nm using an ELISA reader (BioTek Instruments, Inc., Winooski, VT, USA), according to the manufacturer's protocol.

Cell cycle detection by flow cytometry assay

HaCaT cells collected from all groups were washed with PBS and centrifuged at 500 × g for 5 min at 4°C. Cells were adjusted to a density of 1×106 and re-suspended. Propidium iodide (400 µl) and RNA-seA (100 µl; both from Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) were subsequently added to the cells. Following staining for 30 min at room temperature, the DNA content was determined using a FACSCalibur system and analyzed using CellQuest software (version 3.3; BD Biosciences, Franklin Lakes, NJ, USA).

Western blot analysis

HaCaT cell lysates from all groups were extracted using lysis Triton X-100 buffer (250 mM HEPES, 50 mM MgCl2, 10 mM EGTA and 5% Triton X-100). Protein concentration was determined using a bicinchoninic acid protein assay kit (Pierce; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Proteins (10 µg) were resolved on 10% SDS-PAGE gels and transferred onto polyvinylidene fluoride membranes. Membranes were blocked with 5% bovine serum albumin (BSA; Sigma-Aldrich; Merck KGaA) in TBS with 0.1% Tween-20 for 1 h at room temperature, and incubated with anti-β-actin (ab8226; 1:2,000), anti-IL-22, anti-VEGF (ab9570; 1:1,000), anti-p38 MAPK (ab31828; 1:1,000), anti-phosphorylated (p)-p38 MAPK (ab4822; 1:1,000) and anti-NF-κB p65 (ab16502; 1:1,000) (all from Abcam) antibodies at 4°C overnight. β-actin was used as a loading control. Following washing with PBS, blots were incubated with horseradish peroxidase-conjugated secondary antibodies (Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at a concentration of 1:2,000–1:5,000 at room temperature for 1 h. Blots were visualized using chemiluminescence (GE Healthcare Life Sciences, Little Chalfont, UK) and developed on film (Kodak, Rochester, NY, USA), according to the manufacturer's instructions. ImageJ software version 1.42 (National Institutes of Health, Bethesda, MD, USA) was used for densitometry analysis.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay

Total RNA was isolated using an RNeasy mini kit (Qiagen, Inc., Valencia, CA, USA), according to the manufacturer's instructions. A total of 1 µg total RNA was reverse transcribed to cDNA using a PrimeScriptH RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). Reverse transcription reaction conditions were: 30°C for 10 min, 42°C for 30 min, 99°C 5 min and 4°C for 5 min. qPCR was performed using SYBR Green PCR master mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) with a SYBR Green detection system [FS Universal SYBR Green Master (Rox); Roche Applied Science, Penzberg, Germany]. Thermocycling procedures consisted of 50°C for 2 min, 95°C for 10 min and 40 cycles of amplification reactions at 95°C for 15 sec, and at 60°C for 1 min. All samples were analyzed in triplicate and assessed by melting curve analysis, and the PCR products obtained were analyzed by 1% (w/v) agarose gel electrophoresis. The expression levels of IL-22 and VEGF were normalized to β-actin and calculated using the 2−ΔΔCq method (27). The following primer sequences were used: IL-22, 5′-TGAGTGAGCGCTGCTATCTG-3′ (forward), 5′-TGTGCTTAGCCTGTTGCTGA-3′ (reverse); VEGF, 5′-CTGTCTAATGCCCTGGAGCC-3′ (forward), 5′-ACGCGAGTCTGTGTTTTTGC-3′ (reverse); β-actin, 5′-GCAGGAGTATGACGAGTCCG-3′ (forward), 5′-AACAACGCATCTCATATTTGGAA-3′ (reverse).

Statistical analysis

Data are presented as the mean ± standard deviation. All samples were analyzed in triplicate. Statistical comparisons between different groups were performed using SPSS software (version 20; IBM Corp., Armonk, NY, USA). One-way analysis of variance followed by Bonferroni post hoc pairwise comparison was used for the evaluation of differences between groups. P<0.05 was considered to indicate a statistically significant difference.

Results

HaCaT cell proliferation under different concentrations of KGF

To investigate the response of HaCaT cells to KGF, proliferation activity was determined in the presence of different concentrations of KGF. Fig. 1 presents the alterations in cell morphology following treatment with 0–40 ng/ml KGF for 24 h. Cell densities markedly increased in cells treated with 0–10 ng/ml KGF, and marginally increased in the presence of KGF at 10–40 ng/ml. Cells treated with 5–20 ng/ml KGF appeared more elongated compared with cells treated with 0–2.5 ng/ml KGF. The viability of HaCaT cells in the presence of KGF was determined at different time-points (24, 48 and 72 h) and concentrations (0, 1.25, 2.5, 5, 10, 20 and 40 ng/ml). Cell viability was notably elevated following treatment with KGF in a time- and dose- dependent manner. Cell viability significantly increased in cells treated with a dose of >10 ng/ml of KGF, compared with the control.

Effect of TGP treatment on cell hyperproliferation triggered by KGF

One of the primary pathological characteristics of psoriasis is overgrowth and abnormal differentiation of epidermal keratinocytes (4,6). The present study investigated the inhibitory effect of treatment with TGP on the hyperproliferation of HaCaT cells. The cell viability of the model group treated with 10 ng/ml KGF (detected using a CCK-8 assay) significantly increased compared with the control group. However, additional treatment with TGP can reduce the excessive cell proliferation caused by KGF (Fig. 2). Cell hyperproliferation triggered by KGF was markedly inhibited in the TGP-H group, compared with the model group. The effect of TGP treatment on cell hyperproliferation induced by KGF was additionally validated in HaCaT cells obtained from CLS Cell Lines Service GmbH (Eppelheim, Germany; data not shown).

Treatment with TGP suppresses the hyperproliferation of HaCaT cells stimulated by KGF by triggering cell cycle arrest at the G1 phase

To determine the mechanism underlying TGP-mediated inhibition of KGF-induced hyperproliferation, the cell cycle was investigated by flow cytometry. The effect of different concentrations of TGP on cells exposed to KGF was investigated (Fig. 3). The results demonstrated that among cells treated with KGF, fewer cells were in the G0/G1-phase and an increased percentage was in the S-phase, compared with the control group, indicating that KGF treatment stimulated the proliferation of HaCaT cells (Fig. 3A, B and F). However, the KGF-induced proliferation of HaCaT cells was inhibited by increasing concentrations of TGP. Among cells treated with TGP, an increased number of G0/G1-phase cells and a lower number of S-phase cells was determined, compared with the model group, suggesting that treatment with TGP suppressed the hyperproliferation of HaCaT cells stimulated by KGF by inducing G1 arrest (Fig. 3B-F).

Expression of inflammatory cytokines induced by KGF is suppressed by treatment with TGP

A previous study indicated that KGF can simulate the expression of cytokines to promote cell proliferation via activation of p38 MAPK (25). To confirm this function of KGF, the p38 MAPK inhibitor SB203580 was used to treat HaCaT cells. In addition, the effect of treatment with TGP on HaCaT cells was assessed. The results revealed that the expression levels of the inflammatory cytokines IL-22 and VEGF in HaCaT cells were markedly elevated in the model group, compared with the control (Fig. 4). The expression levels of these cytokines decreased following treatment with KGF and SB203580, and following treatment with TGP, compared with the control. Furthermore, the expression levels of IL-22 and VEGF were suppressed in cells treated with KGF and TGP, compared with the model group.

Activation of p38 MAPK is inhibited in cells treated with TGP

Based on the results of a previous study (25), the authors of the present study hypothesized that TGP may induce a similar inhibitory effect to SB203580 on p38 MAPK. Therefore, the expression of p38 MAPK and p-p38 MAPK was determined by western blot analysis. The expression level of p-p38 MAPK was significantly elevated following treatment with KGF compared with the control. By contrast, following treatment with TGP, the expression level of p-p38 MAPK was markedly inhibited compared with the control (Fig. 5). Furthermore, compared with the model group, the expression level of p-p38 MAPK was suppressed by combination treatment with KGF and SB203580, or KGF and TGP (Fig. 5B). However, the expression level of p38 MAPK was approximately equal in all groups. The ratio of p-p38 MAPK to p38 MAPK exhibited a similar pattern to the levels of p-p38 MAPK, since the expression of p38 MAPK was similar in all groups. The inhibitory effect of TGP on p38 MAPK activation was additionally validated in HaCaT cells obtained from CLS Cell Lines Service GmbH (data not shown).

Activation of NF-κB p65 in cells is suppressed by treatment with TGP

Expression of inflammatory cytokinesis regulated by p38 MAPK/NF-κB p65 pathway. Therefore, the present study further determined the expression levels of NF-κB p65 (Fig. 6). The NF-κB p65 protein expression level in cells treated with KGF was upregulated compared with the control. By contrast, treatment with TGP resulted in a downregulation of the expression levels of NF-κB p65. NF-κB p65 protein expression levels were downregulated in cells with combination treatment using KGF and SB203580 or KGF and TGP compared with the model group. The above results were additionally validated in HaCaT cells obtained from CLS Cell Lines Service GmbH (data not shown).

Discussion

The present study aimed to determine whether p38 MAPK may be involved in the therapeutic effect mediated by TGP in an in vitro model of psoriasis. Consistent with previous studies (2426), the results of the present study demonstrated that KGF is associated with the activation of p38 MAPK, which was confirmed by the use of the p38 MAPK inhibitor SB203580. Similar to SB203580, TGP exhibited an apparent inhibitory effect on the activation of p38 MAPK. Furthermore, the expression of IL-22 and VEGF protein and mRNA was suppressed by treatment with TGP, which further supported the hypothesis that TGP may induce a similar inhibitory effect to SB203580 on p38 MAPK.

TGP have been used to efficiently treat psoriasis (19). Even though the mechanism underlying the therapeutic effect of TGP on psoriasis has been previously investigated, and it has been demonstrated to involve the G protein-adenylate cyclase-cAMP signal transduction pathway, it remains to be completely elucidated (22,23). In addition, in a previous study, the proliferation of epidermal cells was affected by KGF, epidermal growth factor, nerve growth factor, TGF, TNF-α and other cytokines (28). However, KGF is the most potent and specific cytokine that promotes keratinocyte proliferation (14,15,29). Therefore, the present study aimed to determine the effect of TGP on the KGF-induced hyperproliferation of HaCaT cells. Consistent with other studies, in the present study, KGF enhanced the proliferation of HaCaT cells. However, this effect of KGF was suppressed following treatment with TGP. Cell cycle analysis demonstrated that among cells treated with KGF and TGP, an elevated number of cells was in the G0/G1-phase and a decreased number of cells was in the S-phase, compared with cells treated with KGF alone. The above results indicated that TGP may suppress the hyperproliferation of HaCaT cells stimulated by KGF, by triggering G1-phase arrest.

It has been hypothesized that the abnormal infiltration of T lymphocytes (primarily CD4+ T lymphocytes) serves a role in the pathogenesis of psoriasis (4,10). Elevated levels of T helper (Th)1-type lymphocytes is the predominant response observed in patients with pancreatic diseases, and Th1 type cytokines, including TNF-α, interferon-γ and IL-2, contribute to the aggravation of psoriasis (30). Th2 type cytokines, including IL-4 and −10, serve a protective role instead (31). Previous studies suggested that psoriasis is a disease based on Th17/Th1 cell responses and hypothesized that these induced Th-17+/IFN-γ+ cells may be the cause of psoriasis (32,33). However, psoriasis is a multi-gene genetic disease associated with a number of factors, including trauma, infection, and the underlying mechanisms of psoriasis remain to be completely elucidated (2,34). A number of previous studies demonstrated that the interaction between IL-23, −17 and −22, and other cytokines, serves a role in the development of psoriasis (3537). Elevated mRNA and protein expression levels of IL-21, −22, −17 and −23 were detected in skin tissue with psoriatic lesions, and IL-23 may induce lesions similar to human psoriasis in mouse skin, including epidermal hyperplasia erythema, leukocyte infiltration and keratosis (35,36). Furthermore, inflammation and acanthosis of the dermal layer have been hypothesized to be associated with cytokine IL-22 and C-C chemokine receptor type 6 (38). The present study demonstrated that the mRNA and protein expression levels of IL-22 were upregulated in cells treated with KGF, whereas they were inhibited in model cells following treatment with TGP or the p38 MAPK inhibitor SB203580.

During the development of psoriasis, numerous vascular-derived cytokines secreted by epidermal cells are involved in angiogenesis, including VEGF (39). In psoriatic lesions, keratinocytes are the primary source of angiogenic cytokines (4,6). Previous studies demonstrated that the mRNA and protein expression levels of VEGF in patients with psoriatic lesions were markedly increased (40,41). Furthermore, it was demonstrated that serum levels of VEGF in patients with severe psoriasis were elevated and served a role in the progression of psoriasis (42). The results of the present study demonstrated that the protein and mRNA expression levels of VEGF were increased in cells treated with KGF. Similar to the levels of IL-22, the expression of VEGF protein and mRNA was suppressed following treatment with TGP. Consistent with these results, treatment of model cells with the p38 MAPK inhibitor SB203580 or TGP additionally inhibited the expression of VEGF.

A previous study demonstrated that KGF simulates p38 MAPK to trigger stress fiber formation in human prostate DU145 cells (43). In addition, a study revealed that the production of VEGF was upregulated following treatment with KGF, and fibroblast growth factor-1 and −2 via the MAPK and p38 MAPK pathways (44). Furthermore, the results of the present study revealed that the expression levels of VEGF protein and mRNA were elevated in cells treated with KGF, while these levels were suppressed by combined treatment with KGF and p38 MAPK inhibitor SB203580, indicating that the expression of VEGF may be associated with the p38 MAPK signaling pathway. Therefore, it may be hypothesized that KGF induces p38 MAPK activation to promote the upregulation of VEGF, and the results of the present study are consistent with this hypothesis. The results of the present study additionally demonstrated that the levels of p-p38 MAPK were associated with the expression levels of IL-22 and VEGF. In addition, similar to the p38 MAPK inhibitor SB20358, TGP inhibited p-p38 MAPK expression levels. The above results suggested that TGP were able to downregulate the expression of proinflammatory factors, including IL-22 and VEGF, by inhibiting the activation of p38 MAPK. Subsequently, alterations in the p38 MAPK/NF-κB p65 signaling pathway were determined. The results indicated that the expression level of NF-κB p65 was markedly elevated and reduced following treatment with KGF and TGP, respectively. Furthermore, subsequent experiments revealed that the expression levels of NF-κB p65 were decreased following treatment of model cells with the p38 MAPK inhibitor SB203580 or TGP, compared with the model group. The above results suggested that the therapeutic effect of TGP on psoriasis may be mediated by the regulation of the p38 MAPK/NF-κB p65 signaling pathway.

In conclusion, TGP may suppress the KGF-induced hyperproliferation of HaCaT cells, which may be involved in the downregulation of the expression of proinflammatory factors, including IL-22 and VEGF. The results of the present study indicated that the p38 MAPK/NF-κB p65 pathway may be associated with psoriasis, and may be effectively suppressed by treatment with TGP.

Acknowledgements

Not applicable.

Funding

Not applicable.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

WP and CC contributed equally to this study and analyzed and interpreted data relating to the study. XQ and SZ designed the study, implemented experiments and wrote the paper. All authors read and approved the final study.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors declare that they have no conflict of interest.

References

1 

Bagel J: Topical therapies for the treatment of plaque psoriasis. Cutis. 84 4 Suppl:S3–S13. 2009.

2 

Dedhar S, Hannigan G, Hunt DWC, Tao JS and Fazli L: Treatment of inflammatory diseases including psoriasis. US Patent 37863429 B2. Filed November 30, 2001; issued January 4. 2011.

3 

Xia JX, Mei XL, Zhu WJ, Li X, Jin XH, Mou Y, Yu K, Wang YY and Li QF: Effect of FGF10 monoclonal antibody on psoriasis-like model in guinea pigs. Int J Clin Exp Pathol. 7:2219–2228. 2014.PubMed/NCBI

4 

Feliciani C, Gupta AK and Sauder DN: Keratinocytes and cytokine/growth factors. Crit Rev Oral Biol Med. 7:300–318. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Zeng X, Zhao J, Wu X, Shi H, Liu W, Cui B, Yang L, Ding X and Song P: PageRank analysis reveals topologically expressed genes correspond to psoriasis and their functions are associated with apoptosis resistance. Mol Med Rep. 13:3969–3976. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Carter EL, O'Herrin S, Woolery C and Longley Jack B: Epidermal stem cell factor augments the inflammatory response in irritant and allergic contact dermatitis. J Inv Dermatol. 128:1861–1863. 2008. View Article : Google Scholar

7 

Cather JC, Latremouille-Viau D, Horn EJ and Bao Y: Psoriasis is significantly associated with lower rates of pregnancy and live births. J Am Acad Dermatol. 66:AB2002012.

8 

Thomas J, Jacobson GA, Narkowicz CK, Peterson GM, Burnet H and Sharpe C: Toenail onychomycosis: An important global disease burden. J Clin Pharm Ther. 35:497–519. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Kubo T, Hatton RD, Oliver J, Liu X, Elson CO and Weaver CT: Regulatory T cell suppression and anergy are differentially regulated by proinflammatory cytokines produced by TLR-activated dendritic cells. J Immunol. 173:7249–7258. 2004. View Article : Google Scholar : PubMed/NCBI

10 

de Boer OJ, van der Loos CM, Hamerlinck F, Bos JD and Das PK: Reappraisal of in situ immunophenotypic analysis of psoriasis skin: Interaction of activated HLA-DR+ immunocompetent cells and endothelial cells is a major feature of psoriatic lesions. Arch Dermatol Res. 286:87–96. 1994. View Article : Google Scholar : PubMed/NCBI

11 

Strain AJ, Mcguinness G, Rubin JS and Aaronson SA: Keratinocyte growth factor and fibroblast growth factor action on DNA synthesis in rat and human hepatocytes: Modulation by heparin. Exp Cell Res. 210:253–259. 1994. View Article : Google Scholar : PubMed/NCBI

12 

Yamamoto T, Matsuda Y, Kawahara K, Naito Z and Ishiwata T: Keratinocyte growth factor stimulates growth of MIA PaCa-2 cells through extracellular signal-regulated kinase phosphorylation. Oncol Lett. 3:307–310. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Higashiyama S, Abraham JA, Miller J, Fiddes JC and Klagsbrun M: A heparin-binding growth factor secreted by macrophage-like cells that is related to EGF. Science. 251:936–939. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Li M: KGF-1 and KGF receptor expression in human periodontal disease and in vitro microwounding-associated-ligand-independent KGFR activation. 2007.http://hdl.handle.net/2429/418

15 

Putnins EE, Firth JD, Lohachitranont A, Uitto VJ and Larjava H: Keratinocyte growth factor (KGF) promotes keratinocyte cell attachment and migration on collagen and fibronectin. Cell Adhes Commun. 7:211–221. 1999. View Article : Google Scholar : PubMed/NCBI

16 

Wang K, Wu Y-G, Su J, Zhang JJ, Zhang P and Qi XM: Total glucosides of paeony regulates JAK2/STAT3 activation and macrophage proliferation in diabetic rat kidneys. Am J Chin Med. 40:521–536. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Harada M: Pharmacological studies on herb paeony root. IV. Analysis of therapeutic effects of paeony- and licorice-containing frequent prescriptions in Chinese medicine and comparison with effects of experimental pharmacological tests. Yakugaku Zasshi. 89:899–908. 1969.(In Japanese). View Article : Google Scholar : PubMed/NCBI

18 

Zhang ZG: Antianxiety and sleep disorder improving use of albiflorin or metabolite thereof in. Patent EP 2644198 A1. Filed November 9, 2011; issued October 2. 2013.

19 

Li Y, Cheng H, Zuo XB, Sheng YJ, Zhou FS, Tang XF, Tang HY, Gao JP, Zhang Z, He SM, et al: Association analyses identifying two common susceptibility loci shared by psoriasis and systemic lupus erythematosus in the Chinese Han population. J Med Genet. 50:812–818. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Zhang HF, Xiao WG and Hou P: Clinical study of total glucosides of paeony in patients with systemic lupus erythematosus. Zhongguo Zhong Xi Yi Jie He Za Zhi. 31:476–479. 2011.(In Chinese). PubMed/NCBI

21 

Wang SL, Wang JP and Bian H: Clinical observation on total glucosides of paeony combined with sulfasalazine in treatment of ankylosing spondylitis. Zhongguo Zhong XI Yi Jie He Za Zhi. 27:217–219. 2007.(In Chinese). PubMed/NCBI

22 

Chang Y, Wei W, Zhang L and Xu HM: Effects and mechanisms of total glucosides of paeony on synoviocytes activities in rat collagen-induced arthritis. J Ethnopharmacol. 121:43–48. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Jia XY, Chang Y, Sun XJ, Wu HX, Wang C, Xu HM, Zhang L, Zhang LL, Zheng YQ, Song LH and Wei W: Total glucosides of paeony inhibit the proliferation of fibroblast-like synoviocytes through the regulation of G proteins in rats with collagen-induced arthritis. Int Immunopharmacol. 18:1–6. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Zhou Y, Li J, Kong F, Zhang H, Fang X, Chen Z, Wang G and Li X and Li X: Clinical and immunological consequences of total glucosides of paeony treatment in Sjögren's syndrome: A randomized controlled pilot trial. Int Immunopharmacol. 39:314–319. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Westra J, Harmsen MC, van Rijswijk MH and Limburg PC: Effect of P38 mapkinase inhibitor RWJ-67657 on proinflammatory mediators produced by IL-1β-and/or TNFα-stimulated rheumatoid synovial fibroblasts. Arthritis Res. 4 Suppl 1:S452002. View Article : Google Scholar

26 

Micallef L, Belaubre F, Pinon A, Jayat-Vignoles C, Delage C, Charveron M and Simon A: Effects of extracellular calcium on the growth-differentiation switch in immortalized keratinocyte HaCaT cells compared with normal human keratinocytes. Exp Dermatol. 18:143–151. 2009. View Article : Google Scholar : PubMed/NCBI

27 

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

28 

Wang B, Amerio P and Sauder DN: Role of cytokines in epidermal Langerhans cell migration. J Leukoc Biol. 66:33–39. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Yang K, Yin J, Sheng B, Wang Q, Han B, Pu A, Yu M, Sun L, Xiao W and Yang H: AhR-E2F1-KGFR signaling is involved in KGF-induced intestinal epithelial cell proliferation. Mol Med Rep. 15:3019–3026. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Nestle FO, Turka LA and Nickoloff BJ: Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lymphocytes and induction of Th1 type cytokines. J Clin Invest. 94:202–209. 1994. View Article : Google Scholar : PubMed/NCBI

31 

de Jong R, Bezemer AC, Zomerdijk TP, Van dp KT, Ottenhoff TH and Nibbering PH: Selective stimulation of T helper 2 cytokine responses by the anti-psoriasis agent monomethylfumarate. Eur J Immunol. 26:2067–2074. 1996. View Article : Google Scholar : PubMed/NCBI

32 

Zaba LC, Fuentesduculan J, Eungdamrong NJ, Abello MV, Novitskaya I, Pierson KC, Gonzalez J, Krueger JG and Lowes MA: Psoriasis is characterized by accumulation of immunostimulatory and Th1/Th17 cell-polarizing myeloid dendritic cells. J Invest Dermatol. 129:79–88. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Kanai Y, Satoh T, Igawa K and Yokozeki H: Impaired expression of Tim-3 on Th17 and Th1 cells in psoriasis. Acta Derm Venereol. 92:367–371. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Harden JL, Krueger JG and Bowcock AM: The immunogenetics of psoriasis: A comprehensive review. J Autoimmun. 64:66–73. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Works MG, Yin F, Yin CC, Yiu Y, Shew K, Tran TT, Dunlap N, Lam J, Mitchell T, Reader J, et al: Inhibition of TYK2 and JAK1 ameliorates imiquimod-induced psoriasis-like dermatitis by inhibiting IL-22 and the IL-23/IL-17 axis. J Immunol. 193:3278–3287. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Raychaudhuri SP: Role of IL-17 in psoriasis and psoriatic arthritis. Clin Rev Allergy Immunol. 44:183–193. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Qin S, Wen J, Bai XC, Chen TY, Zheng RC, Zhou GB, Ma J, Feng JY, Zhong BL and Li YM: Endogenous n-3 polyunsaturated fatty acids protect against imiquimod-induced psoriasis-like inflammation via the IL-17/IL-23 axis. Mol Med Rep. 9:2097–2104. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Zheng Y, Danilenko DM, Valdez P, Kasman I, Eastham-Anderson J, Wu J and Ouyang W: Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature. 445:648–651. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Shibuya M: Involvement of the VEGF-Flt-receptor family in angiogenesis. Nihon Yakurigaku Zasshi. 107:119–131. 1996. View Article : Google Scholar : PubMed/NCBI

40 

Kwon YW, Kim KW, Jang HS, Kim MB, Oh CK and Kwon KS: Insulin-like growth factor-II (IGF-II) induces vascular endothelial growth factor (VEGF) expression through IGFR-1 and ERK1/2 activation in psoriasis. Korean J Dermatol. 42:728–734. 2004.

41 

Verdes D, Popescu R, Gotia SR, Gotia L, Filimon MN and Karabitsakos T: VEGF and VEGFRs expression in psoriasis lesions. Curr Opinion Biotechnol. 22:S1142011. View Article : Google Scholar

42 

Meki AR and Al-Shobaili H: Serum vascular endothelial growth factor, transforming growth factor β1 and nitric oxide levels in patients with psoriasis vulgaris: Their correlation to disease severity. J Clin Lab Anal. 28:496–501. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Mehta PB, Robson CN, Neal DE and Leung HY: Keratinocyte growth factor activates p38 MAPK to induce stress fibre formation in human prostate DU145 cells. Oncogene. 20:5359–5365. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Aimond F, Rauzier JM, Bony C and Vassort G: Simultaneous activation of p38 MAPK and p42/44 MAPK by ATP stimulates the K+ current ITREK in cardiomyocytes. J Biol Chem. 275:39110–39116. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 18 Issue 2

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
Pang W, Qi X, Cao C and Zhang S: Inhibitory effects of TGP on KGF‑induced hyperproliferation of HaCaT cells via suppression of the p38 MAPK/NF‑κB p65 pathway. Mol Med Rep 18: 2207-2215, 2018
APA
Pang, W., Qi, X., Cao, C., & Zhang, S. (2018). Inhibitory effects of TGP on KGF‑induced hyperproliferation of HaCaT cells via suppression of the p38 MAPK/NF‑κB p65 pathway. Molecular Medicine Reports, 18, 2207-2215. https://doi.org/10.3892/mmr.2018.9177
MLA
Pang, W., Qi, X., Cao, C., Zhang, S."Inhibitory effects of TGP on KGF‑induced hyperproliferation of HaCaT cells via suppression of the p38 MAPK/NF‑κB p65 pathway". Molecular Medicine Reports 18.2 (2018): 2207-2215.
Chicago
Pang, W., Qi, X., Cao, C., Zhang, S."Inhibitory effects of TGP on KGF‑induced hyperproliferation of HaCaT cells via suppression of the p38 MAPK/NF‑κB p65 pathway". Molecular Medicine Reports 18, no. 2 (2018): 2207-2215. https://doi.org/10.3892/mmr.2018.9177