Open Access

Erythroid differentiation regulator 1 promotes wound healing by inducing the production of C‑C motif chemokine ligand 2 via the activation of MAP kinases in vitro and in vivo

  • Authors:
    • Byung‑Cheol Lee
    • Jisun Song
    • Arim Lee
    • Daeho Cho
    • Tae Sung Kim
  • View Affiliations

  • Published online on: October 19, 2020     https://doi.org/10.3892/ijmm.2020.4762
  • Pages: 2185-2193
  • Copyright: © Lee et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The erythroid differentiation regulator 1 (Erdr1) protein has been studied for its role in various inflammatory skin diseases, including skin cancer, actinic keratosis and psoriasis. However, the therapeutic effects of Erdr1 on wound repair and its underlying mechanisms remain unknown. The present study aimed to investigate the effects of Erdr1 on wound healing in vitro and in vivo. The results demonstrated that treatment with recombinant Erdr1 enhanced wound healing in vivo and in vitro. In addition, Erdr1 increased the proliferation and migration of human dermal fibroblasts (HDFs). Notably, Erdr1 significantly induced the production of the chemoattractant C‑C motif chemokine ligand 2 (CCL2) and recruited immune cells involved in wound healing. Treatment with recombinant Erdr1 induced the activation of the ERK1/1, p38 and JNK1/2 mitogen‑activated protein (MAP) kinases. Treatment with specific inhibitors for MAP kinase inhibitors markedly suppressed cell proliferation and migration, and inhibited the production of CCL2 in HDFs. Furthermore, the inhibition of CCL2 with a neutralizing antibody significantly suppressed the recombinant Erdr1‑induced proliferation and migration of HDFs. The wound healing activity of Erdr1 was comparable to that of epidermal growth factor. Taken together, these results demonstrated that Erdr1 promoted the proliferation and migration of HDFs and exhibited potent wound healing properties mediated by CCL2. Therefore, the results of the present study suggested that Erdr1 may be a potential therapeutic target for promoting wound healing.

Introduction

The processes that induce the restoration of injured tissues immediately after wound healing comprise three interrelated dynamic phases, namely, the inflammatory, proliferative and regeneration phases (1,2). A number of studies have demonstrated that the primary resident fibroblasts in the dermis are the key players that maintain skin homeostasis during wound healing (3-5). Dermal fibroblasts migrate to the wound site and are induced to proliferate by a variety of growth factors, including platelet-derived growth factor (PDGF), epidermal growth factor (EGF) and transforming growth factor β (3,6,7). Therefore, the migration and proliferation of fibroblasts need to be precisely regulated during wound healing.

Previous studies have reported that a variety of chemokines are expressed at the wound site, indicating the important role of chemokines in recruiting inflammatory cells to the wound site (6,8-10). Notably, the monocyte chemoattractant protein-1, also referred to as C-C motif chemokine ligand 2 (CCL2), is one of the chemokines that mediate the infiltration of monocytes and macrophages during wound healing (11-15). Additionally, treatment with CCL2 improves wound healing by enhancing the recruitment of myeloid cells in db/db mice, a Toll-like receptor 3-deficient mouse model in which the secretion of CCL2 is impaired (16,17). Therefore, understanding the role of CCL2 is important for aiding wound healing and developing novel therapeutic agents.

The erythroid differentiation regulator 1 (Erdr1) is a highly conserved autocrine factor that can induce the synthesis of hemoglobin in both human and murine erythroleukemia (18). Erdr1 is expressed in various tissues, including the placenta, liver, brain, lung, intestine, bone marrow, thymus, sebaceous glands, vessels, nerves, normal human epidermis and human keratinocytes (18-20). A number of studies have reported that Erdr1 exhibits anticancer effects in various types of cancer, including gastric cancer and melanoma. For example, Erdr1 is an antimetastatic factor that is negatively regulated by IL-18 via downregulation of the expression of heat shock protein 90 in melanoma (21). Additionally, recombinant Erdr1 has been reported to suppress the invasiveness and motility of gastric cancer cells via the JNK pathway (19). Erdr1 exhibits a therapeutic potential for various inflammatory diseases, including psoriasis, rosacea, hair loss disorders and rheumatoid arthritis (20,22-26). We therefore hypothesized that Erdr1 may promote the migration and proliferation of fibroblasts involved in wound healing. The mechanisms underlying the therapeutic effects of Erdr1 in wound healing are yet to be elucidated. The present study aimed to investigate the effects of Erdr1 on wound healing.

Materials and methods

Mice and cell culture

Female BALB/c mice (7-week-old; weight, 20-22 g) were purchased from orient Bio, Inc. All experiments were performed following the ethical guidelines of the Korea university Institutional Animal Care and use Committee (Seoul, Korea; approval no. KUIACUC-2018-0045). Human dermal fibroblasts (HDFs; Biosolution Co., Ltd.) were cultured in a mixture (3:1) of DMEM (Thermo Fisher scientific, Inc) and F-12K with 10% fetal bovine serum (Capricorn scientific GmbH), 100 u/ml penicillin and 0.1 mg/ml streptomycin (Thermo Fisher scientific, Inc.) in an incubator at 37°C with 5% CO2. The passage number was <13 for all experiments.

Preparation of recombinant proteins

The recombinant mouse Erdr1 protein was prepared as previously reported (19,21). Briefly, the Erdr1-pCMv-SPORT6 plasmid was purchased from open Biosystems, Inc. The region of the coding sequence was transferred into the bacterial expression plasmid pET22B (Merck KGaA). The 177 amino acid encoded Erdr1 protein was expressed in the Escherichia coli Top10 system (Invitrogen; Thermo Fisher scientific, Inc.), purified using a Ni-NTA purification system according to the manufacturer's instructions (Invitrogen; Thermo Fisher scientific, Inc.), quantified by Pierce™ BCA protein assay kit (Thermo Fisher scientific, Inc.), separated by 10% SDS-PAGE and visualized by Coomassie blue staining (sigma-Aldrich; Merck KGaA) (purity >95%). The endotoxin level of the purified protein (<0.1 EU/ml) was measured using the LAL system (Associated of Cape Cod International, Inc.). The recombinant human EGF protein (purity >98%) was purchased from PeproTech, Inc.

Reagents and antibodies

Antibodies against tubulin (cat. no. sc-69969), JNK1/2 (cat. no. sc-571), phosphorylated (p-) JNK1/2 (cat. no. sc-6254), ERK1/2 (cat. no. sc-153), p-ERK1/2 (cat. no. sc-7383) and p38 (cat. no. sc-535) were purchased from Santa Cruz Biotechnology, Inc. Rabbit-HRP (cat. no. 7074), mouse-HRP (cat. no. 7076) and p-p38 (cat. no. 9216) antibodies were purchased from Cell signaling Technology, Inc. Human CCL2 ELISA pair set (cat. no. SEK10134) was purchased from Sino Biological, Inc. Pluronic® F-127 (cat. no. P2443) for the preparation of 22% (w/v) hydrogel was purchased from sigma Aldrich; Merck KGaA and dissolved in saline overnight on the rotator at 4°C. Inhibitors for ERK (u0126; cat. no. 662005), p38 (SB203580; cat. no. S8307) and JNK (sP600125; cat. no. S5567) were purchased from Merck KGaA.

Proliferation assay

HDFs were seeded and pre-cultured into 96-well plates at a density of 5×103 cells/well for 24 h at 37°C in a 5% CO2 incubator. Subsequently, the cells were cultured without serum for 16 h at 37°C and pre-treated with 1, 10 or 25 µg/ml mitomycin-C (MMC) or PBs for 1 h at 37°C. HDFs were washed and treated with 1, 10 or 50 ng/ml Erdr1 or EGF as a positive control for 24 h at 37°C in serum-free culture medium. To evaluate the effects of the CCL2-neutralizing antibody (Sino Biological, Inc.) on cell proliferation, HDFs were treated with 50 ng/ml Erdr1 or PBs for 24 h following pretreatment with 1, 10 or 30 µg/ml CCL2-neutralizing antibody. After 24 h, the cells were treated with 20 µl of 100 µg/ml 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) solution for 2 h, and the medium was removed. The precipitated formazan was dissolved in 200 µl DMSO, and the absorbance at 560 nm was determined by a microplate reader.

In vitro migration assay

HDFs were seeded into 12-well plates at a density of 8×104 cells/well and pre-cultured for 24 h, followed by culture with serum-free medium for 16 h at 37°C in 5% Co2 incubator. The monolayers of HDFs at 80-90% confluency were scratched with a 200-µl pipette tip, and the plates were washed with PBs to remove the cell debris. HDFs were pre-treated with 1, 10 or 25 ng/ml MMC for 1 h at 37°C, washed and stimulated with 50 ng/ml Erdr1 or 50 ng/ml EGF. To evaluate the effects of the CCL2-neutralizing antibody on cell migration, HDFs were treated with 50 ng/ml Erdr1 or PBs for 24 h following pretreatment with 1, 10 or 30 µg/ml CCL2-neutralizing antibody. After 24 h, the cells were fixed with 4% paraformaldehyde for 10 min, permeabilized with 0.1% Triton x-100 in PBs for 5 min and incubated with Rhodamine phalloidin for 1 h at room temperature. Fluorescence images (magnification, ×100) were acquired using an olympus I× 71 fluorescence microscope (olympus Corporation).

Wound healing assay in vivo

Mouse dorsal hair was shaved and wiped with 70% ethanol before wounding. The mice were anesthetized with 65% N2, 30% O2 and 5% isoflurane and maintained with 2% isoflurane while a 5-mm wound puncture was performed. HG was not used for delivering the compounds to the wound sites after 4 days. The wounds were directly treated with saline, 1 µg EGF or 1 µg Erdr1 in 22% hydrogel (HG) once a day for 5 days. The wound closure was monitored by capturing images at the same position with a fixed height stand to ensure consistent image size. Measurement of wound area in the images was performed using ImageJ software (version 1.45; National Institutes of Health). The analysis of area measurement was performed by one person who was blinded to the grouping of the mice and the sampling day for each wound being measured to avoid measurement bias, and the percentage of wound size was calculated as follows (4,27): wound closure (%)=(wound area on day 0-wound area on day of post-wounding)/wound area on day 0×100%.

Reverse transcription (RT)-PCR

HDFs were stimulated with 1, 10, 25, 50 or 100 ng/ml Erdr1 or EGF for 1, 3, 6, 9 or 12 h, and total RNA was extracted using a RiboEx total RNA kit (GeneAll Biotechnology Co., Ltd.). Total RNA (1 µg) was reverse-transcribed into cDNA by the AccuPower® RT Premix kit (BioNeer Corporation) according to the manufacturer's instructions. The cDNAs were amplified with specific primers by PCR using Genie™ 32 Thermal Block (BioNeer Corporation). The specific primer sequences were as follows: CCL2, sense, 5′-CCTTGCCTTGCTGCTCTACC-3′ and antisense, 5′-CCTATGTGCTGGCCTTGGTG-3′; and GAPDH sense, 5′-ACATCAAGAAGGTGGTGAAG-3′ and antisense, 5′-ATTCAAGAGAGTAGGGAGGG-3′. The PCR products were separated on 1.5% agarose gels and stained using the StainingSTAR solution (Dynebio, Inc.).

ELISA

Human dermal fibroblasts were seeded at 2×105 cells/well into 6-well plates and cultured overnight in serum-free medium for 24 h at 37°C in a 5% Co2 incubator. The cells were subsequently treated with 1, 10 or 50 ng/ml EGF or Erdr1 for 24 h at 37°C. CCL2 protein levels were determined using a CCL2 matched ELISA kit (cat. no. SEK10134; Sino Biological, Inc.) according to the manufacturer's protocol. The absorbance at 450 nm was measured by a microplate reader.

Western blot analysis

Whole cell lysates were prepared using RIPA lysis and extraction buffer (Thermo Fisher scientific, Inc.), and the protein concentration was quantified by BCA assay kit (Thermo Fisher scientific, Inc.). A total of 30 µg of samples per lane were separated on 10% SDS-PAGE gels and transferred to a PVDF membrane. The membrane was blocked with 5% skim milk (Neogen Corporation) in 1× TBS-T (0.1% Tween-20) buffer. The membrane was washed twice with 1× TBS and incubated with the primary antibodies (1:1,000) in 5% BSA for 2 h at room temperature (sigma-Aldrich; Merck KGaA)-TBS buffer. The membrane was washed six times with TBS-T buffer and incubated for 1 h at room temperature with the corresponding HRP-conjugated mouse or rabbit IgG secondary antibodies (1:5,000) in 5% skim milk-TBS. The target proteins were visualized using Amersham ECL Prime western blotting Detection Regent (Cytiva).

Histologic analysis of wounds

Mice (n=5 mice/group) were sacrificed and skin tissues were harvest at day 6 post-wounding. Cross-sections (5 µm) of paraffin-embedded skin tissues were stained with hematoxylin for 5 min and eosin for 2 min (H&E). Stained tissues were mounted with mounting media (Thermo Fisher scientific, Inc.) and observed under light microscopy (x100 magnification). The granulation tissue was identified by the presence of tissue matrix, immune cells, vascular tissue and fibroblasts, and the formation of granulation tissue area was defined as the area between underneath the neo-epithelium and above the subcutaneous fat tissue. The relative granulation tissue area ratios were calculated on the digital images using ImageJ based on the wound closure area, area of dermis and epidermis.

Statistical analysis

The data are presented as the mean ± standard deviation. All experiments were repeated at least three times independently. Statistical differences between the control and the treated groups were assessed by one-way analysis of variance followed by Tukey's post hoc test using SPSS 22 software (IBM Corp.). P<0.05 was considered to indicate a statistically significant difference.

Results

Erdr1 promotes wound repair in vivo

To examine the effects of Erdr1 on wound closure, the wounds were treated with Erdr1-, EGF-, or saline-HG and analyzed. As demonstrated in Fig. 1A, the sizes of wounds in the Erdr1-HG or EGF-HG-treated mice were decreased between days 4 and 10 compared with those in the mice that were treated with saline-HG. The wound closure of the mice treated with Erdr1-HG or saline-HG was ~100% on day 10 (Fig. 1B). As previous studies have reported that the formation of granulation tissue in the early stage of a wound is an important step for supporting re-epithelialization and dermal reconstitution (28,29), the tissues from the wound site were collected on day 6 and stained with H&E for histological analysis; the results demonstrated that the formation of granulation tissues in the group that was treated with Erdr1-HG was similar to that of the group treated with EGF-HG and higher compared with that in the group treated with saline-HG (Fig. 1C). These results demonstrated that the promotion of wound healing of the Erdr1-HG-treated group was similar to that of EGF-HG treated group.

Erdr1 induces HDF proliferation and migration

To investigate the effects of Erdr1 on the proliferation and migration of HDFs, MTT and wound healing assays were performed. As demonstrated in Fig. 2A, 25-100 ng/ml Erdr1 significantly increased the proliferation of HDFs by ~25% compared with that of the PBs-treated control group. The proliferation of the HDFs that were treated with Erdr1 was comparable to that of the HDFs that were treated with 50 ng/ml EGF in the positive control group. To exclude the effects of Erdr1 on the proliferation of HDFs while evaluating the Erdr1-induced migration of HDF wound healing, subsequent assays were performed in the presence of MMC to block cell proliferation. As demonstrated in Fig. 2B, 25 µg/ml MMC completely inhibited cell proliferation, which was not observed at concentrations of 1 and 10 µg/ml. In addition, the proliferation of HDFs induced by 50 ng/ml EFG or 50 ng/ml Erdr1 was decreased by MMC (Fig. 2B).

The results of the wound healing assay demonstrated that Erdr1 increased the migration of HDFs (Fig. 2C). To further examine the effects of Erdr1 on the migration of HDFs, the wound healing assay was performed in the presence of MMC; as presented in Fig. 2D, 1 and 10 µg/ml MMC did not inhibit the migration of Erdr1-treated HDFs. However, 25 µg/ml MMC slightly inhibited the Erdr1-induced migration of HDFs (Fig. 2D). These results suggested that Erdr1 may promote wound healing by inducing the proliferation and migration of HDFs.

Erdr1 increases the production of CCL2 in HDFs

To examine the effects of Erdr1 on the expression of CCL2, HDFs were cultured with various concentrations of Erdr1 or EGF (positive control) for different durations. The mRNA and protein levels of CCL2 were determined using RT-PCR and ELISA, respectively. As presented in Fig. 3A, treatment with 50 and 100 ng/ml Erdr1 increased the mRNA expression levels of CCL2. In addition, the mRNA expression of CCL2 induced by 50 ng/ml Erdr1 appeared to increase over time (Fig. 3B). In order to confirm the effects of Erdr1 on the production of CCL2, ELISA was performed to detect the production of CCL2 in HDFs following stimulation with Erdr1 or EGF. As demonstrated in Fig. 3C, following HDF stimulation with Erdr1, the secretion of CCL2 significantly increased compared with that in the control cells. These results suggested that the Erdr1-induced production of CCL2 may serve a crucial role in wound healing in HDFs.

Erdr1 induces CCL2 production via the MAP kinase pathway in HDFs

Previous studies have demonstrated that the MAP kinase signaling pathway serves an important role in the migration and proliferation of skin fibroblasts and keratinocytes (5,30-33). Therefore, the effects of Erdr1 on the activation of the ERK1/2, p38 and JNK1/2 MAP kinases in HDFs were assessed in the present study. As demonstrated in Fig. 4A, treatment with Erdr1 induced the phosphorylation of ERK1/2, p38 and JNK1/2, and the phosphorylation of MAP kinases induced by Erdr1 at 5 min was comparable to that in the EGF-treated HDFs (Fig. 4A).

In order to further investigate the role of the Erdr1-induced phosphorylation of ERK1/2, p38, and JNK1/2 in the proliferation and migration of HDFs, specific inhibitors of ERK1/2 (u0126), p38 (sB203580) and JNK1/2 (sP600125) were used. As presented in Fig. 4B and C, the proliferation and migration of HDFs induced by Erdr1 were significantly inhibited by the specific inhibitors of ERK1/2, p38 and JNK1/2 MAP kinases. To determine whether these MAP kinases were involved in the Erdr1-induced production of CCL2, HDFs were pretreated with specific MAP kinase inhibitors, and the production of CCL2 protein was measured by ELISA. The results demonstrated that the MAP kinase inhibitors significantly inhibited the production of CCL2 in Erdr1-treated HDFs compared with that in the Erdr1-HG and DMSO-treated group (Fig. 4D), consistent with the results of the proliferation and migration assays. Taken together, these results suggested that the Erdr1-induced production of CCL2 enhanced the proliferation and migration of HDFs by phosphorylating ERK1/2, p38 and JNK1/2.

Involvement of CCL2 in Erdr1-mediated wound healing

To determine the role of the Erdr1-induced production of CCL2 in wound healing, HDFs were treated with a CCL2-neutralizing antibody. As presented in Fig. 5A, the CCL2-neutralizing antibody inhibited the proliferation of HDFs in the Erdr1-treated group. Furthermore, the migration of the cells treated with only CCL2-neutralizing antibody did not increase compared with that in the negative control (PBs-treated) group (Fig. 5B). Taken together, these results suggested that Erdr1 promoted wound healing by inducing the production of CCL2 via the activation of the ERK1/2, p38 and JNK1/2 MAP kinase signaling pathways (Fig. 5C).

Discussion

Wound healing is an intricate, interactive biological process (34,35). Successful wound healing involves inflammation, tissue formation and remodeling (2,5,36). The various behaviors of skin cells, including the migration and proliferation of fibroblasts, are crucial events for wound contraction (2,5,36,37). Additionally, dermal fibroblasts serve an important role in process of wound healing via extracellular modulation (2,5,36,37). The migration and proliferation of dermal fibroblasts serve an essential role in tissue regeneration and repair; fibroblasts and keratinocytes are involved in the proliferation, migration and deposition of the extracellular matrix as well as the formation of granulation tissue (2,6,8). Erdr1 is expressed in various tissues and cells, including the normal human epidermis, CD4+ T cells, skin tumors and human keratinocytes (18-20,38). Our previous study demonstrated that the sequence of human Erdr1 identified from the cDNA from human tissues exhibited 100% homology with the sequence of Erdr1 from murine tissues (21). However, the sequence of human Erdr1 was not identified in the present study; the sequence of human Erdr1 will be analyzed in future studies. Our recent study reported that Erdr1 is a potential therapeutic agent for inflammatory skin diseases including psoriasis (25), rosacea (26) and hair loss disorders (39); therefore, we hypothesized in the present study that Erdr1 may also promote wound healing.

In the present study, topical administration of Erdr1-HG enhanced the healing of skin wounds and induced complete wound closure after 10 days of treatment in vivo. It was observer that tissue repair was more rapid in the wounds that were treated with Erdr1 compared with that in the wounds treated with saline, and the wound healing effects of Erdr1 were comparable to those of EGF-HG in vivo. Notably, in the present study, the skin wounds in vivo were treated with saline, Erdr1 or EGF in HG at a final concentration of 20% (w/v); HG was used in these experiments as it has been used in several studies for delivering therapeutic agents to wound sites (40-42). However, HG was unable to penetrate the wound site owing to the formation of a fibrin clot, which inhibited the penetration of the compounds when the skin wounds were treated with saline, Erdr1 or EGF after 4 days of inflicting the wounds. Therefore, HG was not used for delivering the compounds to the wound sites after 4 days. It is necessary to modify the composition of the HG to improve the penetrative efficiency and targeted delivery of the compounds to the wound site in future studies.

It has been demonstrated that the role of the MAP kinase pathway in inflammatory skin diseases is mediated by inflammatory cytokines, such as IL-22 and IL-23, and several cellular functions, such as fibroblast and keratinocyte migration (5,31). The results of the present study demonstrated that the ERK, p38 and JNK pathways served an important role in promoting the migration and proliferation of HDFs, and were consistent with the results of several previous studies (5,28,31). Therefore, the role of the MAP kinase pathway in the migration, proliferation and CCL2 production was evaluated in the present study. The results demonstrated that Erdr1 served an important role in the migration and proliferation of HDFs via the activation of the ERK1/2, p38 and JNK1/2 signaling pathways. Although the levels of p-ERK1/2 and p-p38 began to decrease or disappeared after 5 min of EGF and Erdr1 treatments, this did not represent a reduction in CCL2 production. In addition, certain MAPK pathway inhibitors suppressed Erdr1-induced cell proliferation, CCL2 production and migration at 24 h.

Chemokines are chemoattractants that recruit monocytes, neutrophils and lymphocytes by binding to G-protein-coupled receptors (6,14,17). CCL2 is one of the key chemokines that regulate the migration and infiltration of monocytes and macrophages (43,44). Recent studies have demonstrated that several cytokines are expressed at the site of the wound and serve an essential role in the recruitment of inflammatory cells to the wound site (6,11,14). Furthermore, the expression of chemokine receptors on the resident cells suggests that chemokines contribute to re-epithelialization, tissue remodeling, and angiogenesis (10). In particular, MCP-1/CCL2 is a major chemoattractant that participates in the infiltration of inflammatory cells, including monocytes and macrophages, to the wound site (9-11,13,14,16,17,44). These studies suggest that CCL2 is directly involved in wound healing. The production of CCL2 is induced by oxidative stress, cytokines or growth factors in a variety of cell types, including endothelial, epithelial, smooth muscle and mesangial cells, astrocytes, monocytes, microglial cells and fibroblasts (45). It has been demonstrated that CCL2 is involved in wound healing and various diseases, including multiple sclerosis, rheumatoid arthritis, atherosclerosis, insulin-resistant diabetes (46-48).

A previous study has reported that treatment with a CCL-2-neutralizing antibody suppresses the number of macrophages at the wound site (15). In the present study, treatment with the CCL2-neutralizing antibody decreased in the migration and proliferation of HDFs. Furthermore, Erdr1 significantly induced the production of CCL2 by activating ERK1/2, p38 and JNK1/2 in HDFs.

The results of the present study have several clinical limitations. First, Erdr1 is a novel factor for wound healing that significantly increased the CCL2 production in HDFs. Thus, we suggest that Erdr1 may be used as a therapeutic agent with other growth factors such as EGF for synergistic improvement of wound repair. Second, undefined receptors of Erdr1 should be investigated for increasing the therapeutic potential of Erdr1 to promote tissue repair. Third, it would be interesting to assess the effect of topical application of Erdr1 in chronic/diabetic wounds in future studies.

In conclusion, the results of the present study suggested that Erdr1 accelerated wound healing by increasing the production of CCL2, which induced the migration and proliferation of fibroblasts via the activation of the ERK1/2, p38 and JNK1/2 signaling pathways in vitro. Therefore, Erdr1 may serve as a potential therapeutic target for wound healing and for the development of therapeutic agents for topical treatment of wounds.

Acknowledgments

No applicable.

Funding

This study was supported by the Creative Materials Discovery Program through the National Research Foundation of Korea (grant no. 2016M3D1A1021387) and the National Research Foundation of Korea (grant no. NRF-2018R1A2B6008434), and a grant from Kine Sciences.

Availability of data and materials

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

Authors' contributions

BCL conceived and designed the experiments. BCL, JS and AL performed the experiments. BCL and TSK wrote the manuscript. BCL, DC and TSK analyzed data. BCL and TSK were responsible for overall study design and supervised the project. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All experiments were performed following the ethical guidelines of the Korea university Institutional Animal Care and use Committee (Seoul, Korea; approval no. KUIACUC-2018-0045).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Singer AJ and Clark RA: Cutaneous wound healing. N Engl J Med. 341:738–746. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Darby IA, Laverdet B, Bonte F and Desmouliere A: Fibroblasts and myofibroblasts in wound healing. Clin Cosmet Investig Dermatol. 7:301–311. 2014.PubMed/NCBI

3 

Walraven M, Gouverneur M, Middelkoop E, Beelen RH and Ulrich MM: Altered TGF-ß signaling in fetal fibroblasts: What is known about the underlying mechanisms? Wound Repair Regen. 22:3–13. 2014. View Article : Google Scholar

4 

Lee BC, Song J, Lee A, Cho D and Kim TS: Visfatin promotes wound healing through the activation of ERK1/2 and JNK1/2 pathway. Int J Mol Sci. 19:36422018. View Article : Google Scholar :

5 

Kanazawa S, Fujiwara T, Matsuzaki S, Shingaki K, Taniguchi M, Miyata S, Tohyama M, Sakai Y, Yano K, Hosokawa K and Kubo T: bFGF regulates PI3-kinase-Rac1-JNK pathway and promotes fibroblast migration in wound healing. PLoS One. 5:e122282010. View Article : Google Scholar : PubMed/NCBI

6 

Barrientos S, Stojadinovic O, Golinko MS, Brem H and Tomic-Canic M: Growth factors and cytokines in wound healing. Wound Repair Regen. 16:585–601. 2008. View Article : Google Scholar

7 

Lichtman MK, Otero-Vinas M and Falanga V: Transforming growth factor beta (TGF-ß) isoforms in wound healing and fibrosis. Wound Repair Regen. 24:215–222. 2016. View Article : Google Scholar

8 

Raja, Sivamani K, Garcia MS and Isseroff RR: Wound re-epithelialization: Modulating keratinocyte migration in wound healing. Front Biosci. 12:2849–2868. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Ding J and Tredget EE: The role of chemokines in fibrotic wound healing. Adv Wound Care (New Rochelle). 4:673–686. 2015. View Article : Google Scholar

10 

Rees PA, Greaves NS, Baguneid M and Bayat A: Chemokines in wound healing and as potential therapeutic targets for reducing cutaneous scarring. Adv Wound Care (New Rochelle). 4:687–703. 2015. View Article : Google Scholar

11 

DiPietro LA, Polverini PJ, Rahbe SM and Kovacs EJ: Modulation of JE/MCP-1 expression in dermal wound repair. Am J Pathol. 146:868–875. 1995.PubMed/NCBI

12 

Wetzler C, Kampfer H, Stallmeyer B, Pfeilschifter J and Frank S: Large and sustained induction of chemokines during impaired wound healing in the genetically diabetic mouse: Prolonged persistence of neutrophils and macrophages during the late phase of repair. J Invest Dermatol. 115:245–253. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Low QE, Drugea I A, Duffner LA, Quinn DG, Cook DN, Rollins BJ, Kovacs EJ and DiPietro LA: Wound healing in MIP-1alpha(-/-) and MCP-1(-/-) mice. Am J Pathol. 159:457–463. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Engelhardt E, Toksoy A, Goebeler M, Debus S, Brocker EB and Gillitzer R: Chemokines IL-8, GROalpha, mcp-1, IP-10, and Mig are sequentially and differentially expressed during phase-specific infiltration of leukocyte subsets in human wound healing. Am J Pathol. 153:1849–1860. 1998. View Article : Google Scholar : PubMed/NCBI

15 

Dipietro LA, Reintjes MG, Low QE, Levi B and Gamelli RL: Modulation of macrophage recruitment into wounds by monocyte chemoattractant protein-1. Wound Repair Regen. 9:28–33. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Wood S, Jayaraman V, Huelsmann EJ, Bonish B, Burgad D, Sivaramakrishnan G, Qin S, DiPietro LA, Zloza A, Zhang C and Shafikhani SH: Pro-inflammatory chemokine CCL2 (MCP-1) promotes healing in diabetic wounds by restoring the macrophage response. PLoS One. 9:e915742014. View Article : Google Scholar : PubMed/NCBI

17 

Lin Q, Fang D, Fang J, Ren X, Yang X, Wen F and Su SB: Impaired wound healing with defective expression of chemokines and recruitment of myeloid cells in TLR3-deficient mice. J Immunol. 186:3710–3717. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Dormer P, Spitzer E, Frankenberger M and Kremmer E: Erythroid differentiation regulator (EDR), a novel, highly conserved factor I. Induction of haemoglobin synthesis in erythroleukaemic cells. Cytokine. 26:231–242. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Jung MK, Houh YK, Ha S, Yang Y, Kim D, Kim TS, Yoon SR, Bang SI, Cho BJ, Lee WJ, et al: Recombinant Erdr1 suppresses the migration and invasion ability of human gastric cancer cells, SNU-216, through the JNK pathway. Immunol Lett. 150:145–151. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Lee YB, Kim HJ, Jung HY, Park YG, Kim SY, Cho BK, Cho D and Park HJ: Downregulation of erythroid differentiation regulator 1 as a novel marker of skin tumors. Int J Dermatol. 53:723–730. 2014. View Article : Google Scholar

21 

Jung MK, Park Y, Song SB, Cheon SY, Park S, Houh Y, Ha S, Kim HJ, Park JM, Kim TS, et al: Erythroid differentiation regulator 1, an interleukin 18-regulated gene, acts as a metastasis suppressor in melanoma. J Invest Dermatol. 131:2096–2104. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Houh YK, Kim KE, Park HJ and Cho D: Roles of erythroid differentiation regulator 1 (Erdrl) on inflammatory skin diseases. Int J Mol Sci. 17:20592016. View Article : Google Scholar

23 

Kim KE, Kim S, Park S, Houh Y, Yang Y, Park SB, Kim S, Kim D, Hur DY, Kim S, et al: Therapeutic effect of erythroid differentiation regulator 1 (Erdrl) on collagen-induced arthritis in DBA/1J mouse. Oncotarget. 7:76354–76361. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Kim KE, Houh Y, Lee J, Kim S, Cho D and Park HJ: Downregulation of erythroid differentiation regulator 1 (Erdrl) plays a critical role in psoriasis pathogenesis. Exp Dermatol. 25:570–572. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Kim KE, Houh Y, Park HJ and Cho D: Therapeutic effects of erythroid differentiation regulator 1 on imiquimod-induced psoriasis-like skin inflammation. Int J Mol Sci. 17:2442016. View Article : Google Scholar : PubMed/NCBI

26 

Kim M, Kim KE, Jung HY, Jo H, Jeong SW, Lee J, Kim CH, Kim H, Cho D and Park HJ: Recombinant erythroid differentiation regulator 1 inhibits both inflammation and angiogenesis in a mouse model of rosacea. Exp Dermatol. 24:680–685. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Asai J, Takenaka H, Hirakawa S, Sakabe JI, Hagura A, Kishimoto S, Maruyama K, Kajiya K, Kinoshita S, Tokura Y and Katoh N: Topical simvastatin accelerates wound healing in diabetes by enhancing angiogenesis and lymphangiogenesis. Am J Pathol. 181:2217–2224. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Matsubayashi Y, Ebisuya M, Honjoh S and Nishida E: ERK activation propagates in epithelial cell sheets and regulates their migration during wound healing. Curr Biol. 14:731–735. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Coutelle O, Hornig-Do HT, Witt A, Andree M, Schiffmann LM, Piekarek M, Brinkmann K, Seeger JM, Liwschitz M, Miwa S, et al: Embelin inhibits endothelial mitochondrial respiration and impairs neoangiogenesis during tumor growth and wound healing. EMBO Mol Med. 6:624–639. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Satish L, Babu M, Tran KT, Hebda PA and Wells A: Keloid fibroblast responsiveness to epidermal growth factor and activation of downstream intracellular signaling pathways. Wound Repair Regen. 12:183–192. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Wahedi HM, Park YU, Moon EY and Kim SY: Juglone ameliorates skin wound healing by promoting skin cell migration through Rac1/Cdc42/PAK pathway. Wound Repair Regen. 24:786–794. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Chrissouli S, Pratsinis H, Velissariou V, Anastasiou A and Kletsas D: Human amniotic fluid stimulates the proliferation of human fetal and adult skin fibroblasts: The roles of bFGF and PDGF and of the ERK and Akt signaling pathways. Wound Repair Regen. 18:643–654. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Jiang WG, Ye L, Patel G and Harding KG: Expression of WAVEs, the WASP (Wiskott-Aldrich syndrome protein) family of verprolin homologous proteins in human wound tissues and the biological influence on human keratinocytes. Wound Repair Regen. 18:594–604. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Aya KL and Stern R: Hyaluronan in wound healing: Rediscovering a major player. Wound Repair Regen. 22:579–593. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Eming SA, Martin P and Tomic-Canic M: Wound repair and regeneration: Mechanisms, signaling, and translation. Sci Transl Med. 6:265sr2662014. View Article : Google Scholar

36 

Rosinczuk J, Taradaj J, Dymarek R and Sopel M: Mechanoregulation of wound healing and skin homeostasis. Biomed Res Int. 2016:39434812016. View Article : Google Scholar : PubMed/NCBI

37 

Rosique RG, Rosique MJ and Farina Junior JA: Curbing inflammation in skin wound healing: A review. Int J Inflam. 2015:3162352015. View Article : Google Scholar : PubMed/NCBI

38 

Soto R, Petersen C, Novis CL, Kubinak JL, Bell R, Stephens WZ, Lane TE, Fujinami RS, Bosque A, O'Connell RM and Round JL: Microbiota promotes systemic T-cell survival through suppression of an apoptotic factor. Proc Natl Acad Sci USA. 114:5497–5502. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Woo YR, Hwang S, Jeong SW, Cho DH and Park HJ: Erythroid differentiation regulator 1 as a novel biomarker for hair loss disorders. Int J Mol Sci. 18:3162017. View Article : Google Scholar :

40 

Zhang M and Zhao X: Alginate hydrogel dressings for advanced wound management. Int J Biol Macromol. 162:1414–1428. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Francesko A, Petkova P and Tzanov T: Hydrogel dressings for advanced wound management. Curr Med Chem. 25:5782–5797. 2018. View Article : Google Scholar

42 

Dhaliwal K and Lopez N: Hydrogel dressings and their application in burn wound care. Br J Community Nurs. 23(Suppl 9): S24–S27. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Lu B, Rutledge BJ, Gu L, Fiorillo J, Lukacs NW, Kunkel SL, North R, Gerard C and Rollins BJ: Abnormalities in monocyte recruitment and cytokine expression in monocyte chemoattractant protein 1-deficient mice. J Exp Med. 187:601–608. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Werner S and Grose R: Regulation of wound healing by growth factors and cytokines. Physiol Rev. 83:835–870. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Craig MJ and Loberg RD: CCL2 (Monocyte Chemoattractant Protein-1) in cancer bone metastases. Cancer Metastasis Rev. 25:611–619. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Standiford TJ, Kunkel SL, Phan SH, Rollins BJ and Strieter RM: Alveolar macrophage-derived cytokines induce monocyte chemoattractant protein-1 expression from human pulmonary type II-like epithelial cells. J Biol Chem. 266:9912–9918. 1991.PubMed/NCBI

47 

Kusano KF, Nakamura K, Kusano H, Nishii N, Banba K, Ikeda T, Hashimoto K, Yamamoto M, Fujio H, Miura A, et al: Significance of the level of monocyte chemoattractant protein-1 in human atherosclerosis. Circ J. 68:671–676. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Sartipy P and Loskutoff DJ: Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci USA. 100:7265–7270. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2020
Volume 46 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee BC, Song J, Lee A, Cho D and Kim TS: Erythroid differentiation regulator 1 promotes wound healing by inducing the production of C‑C motif chemokine ligand 2 via the activation of MAP kinases in vitro and in vivo. Int J Mol Med 46: 2185-2193, 2020
APA
Lee, B., Song, J., Lee, A., Cho, D., & Kim, T.S. (2020). Erythroid differentiation regulator 1 promotes wound healing by inducing the production of C‑C motif chemokine ligand 2 via the activation of MAP kinases in vitro and in vivo. International Journal of Molecular Medicine, 46, 2185-2193. https://doi.org/10.3892/ijmm.2020.4762
MLA
Lee, B., Song, J., Lee, A., Cho, D., Kim, T. S."Erythroid differentiation regulator 1 promotes wound healing by inducing the production of C‑C motif chemokine ligand 2 via the activation of MAP kinases in vitro and in vivo". International Journal of Molecular Medicine 46.6 (2020): 2185-2193.
Chicago
Lee, B., Song, J., Lee, A., Cho, D., Kim, T. S."Erythroid differentiation regulator 1 promotes wound healing by inducing the production of C‑C motif chemokine ligand 2 via the activation of MAP kinases in vitro and in vivo". International Journal of Molecular Medicine 46, no. 6 (2020): 2185-2193. https://doi.org/10.3892/ijmm.2020.4762