Open Access

Identification of differentially expressed proteins involved in fetal scarless wound healing using a rat model of cleft lip

  • Authors:
    • Yu Yan
    • Hong Liu
    • Jiarong Yi
    • Zizi Chen
    • Jia Chen
    • Jianfei Zhang
    • Kewa Gao
    • Siqi He
    • Aijun Wang
    • Ping Jin
    • Feng Hu
    • Jianda Zhou
  • View Affiliations

  • Published online on: June 22, 2021     https://doi.org/10.3892/mmr.2021.12235
  • Article Number: 596
  • Copyright: © Yan 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

In early pregnancy, fetal skin wounds can heal quickly and undergo a transition period from scarless healing to scar formation. The aim of the present study was to identify potential biomarkers associated with scarless repair of cleft lips, in order to determine the intrinsic factors leading to scar formation in embryonic tissue. A stable model of cleft lip was established using microsurgery by constructing a wedge‑shaped cleft lip‑like defect in fetal rats at gestational age (GA) 16.5 and GA18.5. The GA16.5 and GA18.5 groups were used to model scarless healing and scar formation, respectively. The fetuses were returned to the uterus following surgery, then removed 72 h after the procedure. Macroscopic observation of the cleft defect and histological examination were carried out. Reverse transcription‑quantitative (RT‑q) PCR and parallel reaction monitoring (PRM) were used to detect mRNA and protein expression levels, respectively. The upper‑left lip completely healed 72 h after surgery in the GA16.5 group of fetal rats. However, this was not the case in the GA18.5 group. Histological examination indicated new follicles visible under the epidermis of the scarless group (GA16.5). Scarring was visible on the upper‑left cleft lip wound of the fetal rats in the GA18.5 group. The expression of some growth and pro‑inflammatory factors, including TNF‑α, were also different between two groups. Label‑free quantification was used to identified differentially expressed proteins and five differentially expressed proteins (Smad4, Fabp5, S100a4, S100a8 and S100a9) were identified. The relative expression of these molecules at the mRNA and protein levels were measured using RT‑qPCR and PRM. These molecules may represent potential biomarkers for the scarless repair of fetal rat cleft lip wounds.

Introduction

The cleft lip is a very common congenital oral and maxillofacial malformation, often accompanied by cleft palate and alveolar cleft. Although surgical repair techniques are continuously being improved, numerous patients still experience inevitable secondary scar formation after surgery. In recent years, with the development of prenatal diagnosis and treatment technology (1), intrauterine surgery has made it possible to correct developmental deformities, such as a cleft lip.

The concept of scarless healing was first proposed by Burrington (2) in 1971. It was later observed that fetal skin wounds that occur during early pregnancy can heal quickly and restore intact skin barrier functions. In contrast, fetal skin damage that occurs in the third trimester of pregnancy can result in the formation of scar tissue similar to that of an adult (3). Therefore, the different manifestations of scarless healing of mammalian fetal wounds are related to the gestational age of the fetus (4). Dang et al (5) and Longaker et al (6) demonstrated that this transition period from scarless healing to scar formation occurred between day 16.5 of gestational age (GA) and GA18.5 in rats and mice, which have a gestation period of ~21.5 days. Lorenz et al (7) and Cass et al (8) suggested that when 1–2 mm incisions are inflicted on fetal rats, the transition period of scarless healing to healing with scar formation was still between 16.5 (GA16.5) and 18.5 days (GA18.5).

This phenotypic difference in fetal wounds has inspired further examination of the specific underlying mechanisms. Initially, it was hypothesized that the reason for early scar repair was that the fetus developed in amniotic fluid, which is rich in growth factors and extracellular matrix (ECM) components (9,10). Previous studies typically utilized large animal models to study the presence of scars following repair (11,12). However, only a few studies have reported the use of a fetal rat cleft lip wound model to establish the effectiveness of surgical repair at different gestational ages. Moreover, due to the short gestation period of rats, the experimental cycle can be shortened, and the experiment can therefore be repeated.

Given the importance of this process, the present study, screened out several specific markers of early fetal scarless repair. The present study aimed to gain insight into the occurrence and mechanisms of scarless repair, and to identify new clinical targets for the prevention and treatment of scars.

Materials and methods

Animals

A total of 36 SPF-grade adult Sprague-Dawley (SD) rats (female; mean weight, 250 g; age, 12 weeks) were obtained from the Third Xiangya Hospital of Central South University Animal Experiment Center (Hunan, China) and divided into two groups that received surgery once their fetuses reached GA16.5 or GA18.5, respectively (n=18 each). The following housing conditions were implemented: A temperature between 25±2°C, relative humidity of 55±15%, ventilation rate of 10–20 times per hour, time-controlled artificial lighting (12-h day-night cycle) and ad libitum access to food and water. The experiments were supervised throughout and were performed in accordance with animal experimentation ethics.

Preliminary study on different repair modes applicable to fetal rats with artificial cleft lip wounds

Fetal rats located away from the uterine horn were selected to prevent subsequent abortion, as described previously (13). In the current study, rats were anesthetized with 30 mg/kg pentobarbital sodium intraperitoneally before surgery. A wedge-shaped cleft-like defect was created on the upper-left lip of the fetal rats. The upper-right lip did not receive any treatment and was used as a control condition. The fetal rats were then returned to the uterus. Fetal rats from the GA16.5 and GA18.5 groups were then removed three days post-surgery as previously described (4) (i.e., at GA19.5 or GA21.5, respectively). All fetuses and rats were euthanized using carbon dioxide (30% volume displaced/min). Death was confirmed using cervical dislocation. A total of three fetal rats were obtained from each pregnant rat, for a total of 54 fetal rats from both GA16.5 and GA18.5 groups, and the survival rate was calculated. Tissue samples from the surgical site on the upper-left lip and asymmetrical sections from the upper-right lip were collected from the fetal rats for histological examination, including hematoxylin and eosin (H&E) staining, Masson's Trichrome staining and type-I collagen immunohistochemical staining as previously described (1416).

The upper-left lip tissue samples from the GA16.5 group were defined as group 1, whereas the upper-right lip tissue samples from the GA16.5 group were defined as group 2. In addition, the upper-left lip tissue samples from the GA18.5 group were defined as group 3, whereas the upper-right lip tissue samples from the GA18.5 group were defined as group 4. Each subgroup included 27 samples. Protein expression was compared between group 1 and 2, group 3 and 4, as well as group 3 and 1. Label-free quantification PRM was performed as previously described (17) and was used to detect the differentially expressed proteins among the different groups. MaxQuant 1.5.6 (https://www.maxquant.org) and Perseus 1.4 (https://www.maxquant.org/perseus/) were used to analyze the results of label-free quantification PRM: Volcano plots were generated for differentially expressed proteins: Y-axis, -log10(P-value); x-axis: log2(ratio). The points distributed outside the two vertical borders and above the horizontal border represented the proteins with significant differences; proteins with at least a 1.5-fold change in expression and P<0.05 were considered significant. Subsequently, bioinformatics analysis, including GO and KEGG pathway analysis, was performed to identify differentially expressed proteins (18).

Experimental verification of tissue repair proteins in fetal rats with artificial cleft lip wounds

The mRNA levels of the differentially expressed molecules were assessed using reverse transcription-quantitative (RT-q) PCR, as previously described (19). Differentially expressed levels of proteins were detected by label-free quantification PRM as previously described (17).

Statistical analysis

GraphPad Prism 8.0 (GraphPad Software, Inc.) and SPSS 22.0 (IBM Corp.) were used to perform calculations and carry out statistical analysis. Student's t-test was used to compare differences between two groups. The experimental data from each group were analyzed for congruence of variance before the t-test were applied. The FDR values were within 0.01 in the comparisons. Mixed ANOVA followed by Sidak's post hoc test was used to analyze the differences between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Gross observation

All fetal rats were observed before delivery. The nasolabial cleft was first observed before surgery and images were captured to facilitate the observation of changes in the fetal rats from the GA16.5 and GA18.5 groups. We observed the same area again 72 h post-surgery to identify differences. The cuneiform tissue of the upper-left lip was removed by microsurgery to create a cleft lip wound. The changes in the fetal rats were observed macroscopically. In the GA16.5 group, the upper-left cleft lip wound completely healed 72 h after surgery (i.e., GA19.5) and the continuity of the upper lip tissue was restored. Only a slight depression was observed in the surgical area. The upper-left lip tissue was nearly symmetrical with that of the right side. However, in the GA18.5 group, the cleft lip wound was not completely healed 72 h after surgery (i.e., GA21.5); a clear scar was observed in the surgical area, and the upper lip was asymmetrical on both sides due to wound contracture (Fig. 1).

Histological analysis

In the GA16.5 group 72 h after surgery, the tissue of the upper-left lip wound demonstrated complete regeneration when observed under the microscope (Figs. 24). The results of H&E staining demonstrated complete epithelialization of the upper-left lip, and the structure of new follicles was detected under the epidermis. Compared with the normal skin of the upper-right lip, a slight depression in the cleft part of the upper-left lip and thickening of the skin was noted, whereas inflammatory cell infiltration and neovascularization were not apparent (Fig. 2). Masson's Trichrome staining revealed collagen fibers under the epidermis, demonstrating a fine reticular and emerging follicular structure (Fig. 3). Immunohistochemical analysis indicated no obvious difference in the amount of type-I collagen in the upper-left cleft lip area and the rest of the upper lip (Fig. 4).

In the GA18.5 group, the position of the wound was easily identified by a distinct scar on the upper-left lip. H&E staining demonstrated that partial epithelialization occurred in the upper-left cleft lip area. Compared with the normal skin of the upper-right lip, the upper-left lip displayed a clear scar, new capillary formation around the wound and increased fibroblast proliferation and ECM volume, whereas structural components of hair follicles were not observed under the epidermis (Fig. 5). Masson's Trichrome staining demonstrated the absence of new follicular structure and the presence of dense collagen fibers under the epidermis (Fig. 6). Immunohistochemical analysis in the upper-left cleft lip wound demonstrated an increase in type-I collagen expression and fiber density, as well as a more compact structure and absence of adnexal skin (Fig. 7), compared with normal upper lip tissue.

Immunohistochemical analysis of cell proliferation markers was also carried out. Compared with GA16.5 fetal rats, the expression of Ki67 and CD31 slightly increased in the GA18.5 group following surgery. By contrast, the expression of CK10 decreased in the GA18.5 group, compared with the GA16.5 group (Fig. 8).

RT-qPCR analysis of inflammatory factors

The relative mRNA expression levels of the pro-inflammatory factors TNF-α, IL-10 and TGF-β were evaluated in the two groups of fetal rats. The mRNA levels of TNF-α and IL-10 were significantly higher in GA18.5 rats, compared with GA16.5 rats. Furthermore, the mRNA expression levels of TGF-β were significantly reduced in the GA18.5 group (Fig. 9).

Protein identification and differential protein screening

Compared with group 1, 57 differentially expressed proteins were identified in group 2, of which 37 were upregulated and 20 were downregulated. A comparison of groups 3 and 4 revealed 312 differentially expressed proteins, of which 171 were upregulated and 141 were downregulated. Lastly, compared with group 1,289 differentially expressed proteins were identified in group 3, of which 151 were upregulated and 138 were downregulated. Only 50 differentially expressed proteins and their multiple variations were upregulated or downregulated between all groups (Tables IIV). The distribution of the differentially expressed proteins among the selected samples is presented as volcano plots (Figs. 10,11,12).

Table I.

Comparison of differentially expressed protein numbers between samples.

Table I.

Comparison of differentially expressed protein numbers between samples.

SampleDifferentially expressed proteins, nUpregulated proteins, nDownregulated protein, n
Group 1 vs. Group 2573720
Group 3 vs. Group 4312171141
Group 3 vs. Group 1312151138

[i] Group 1, upper-left lip without scar repair group at 72 h after modeling in GA16.5 rats; Group 2, upper-right lip normal group at 72 h after modeling in GA16.5 rats; Group 3, upper-left lip scar repair group at 72 h after modeling in GA18.5 rats; Group 4, upper-right lip normal group at 72 h after modeling in GA18.5 rats.

Table IV.

Differential protein expression in group 3 and group 1.

Table IV.

Differential protein expression in group 3 and group 1.

Protein IDGene nameProtein nameP-valueFold change
Q6jhy3SmgcSubmandibular gland protein c precursor0.001753.286
D3zge2MpoMyeloperoxidase precursor0.000271.832
Q62714Np4Neutrophil antibiotic peptide np-40.000244.647
D3zmi6Olfm4Olfactomedin-4 precursor0.000189.639
D3zy96NgpNeutrophilic granule protein precursor0.014130.459
B2ryb8Itgb2Integrin beta 2 precursor0.00244.874
G3v8l7ItgamIntegrin alpha-m precursor0.01232.024
Q6ig02Krt2Keratin, type ii cytoskeletal 2 epidermal0.01126.577
Q63015Csap1Common salivary protein 1 precursor0.00326.571
P50115S100a8S100 calcium binding protein a80.00225.933
P50116S100a9S100 calcium binding protein a90.00119.538
Q9jkb7GdaGuanine deaminase0.02319.076
D3zd07Fmo9Flavin containing monooxygenase 9 pseudogene0.00317.428
Q5u1y2Rac2Ras-related c3 botulinum toxin substrate 20.01112.859
O54854Klk6Kallikrein-6 precursor0.00010.614
Q5u2v4Plbd1Phospholipase b-like 10.00210.508
Q6pdv1Lyz1Lysozyme c-1 precursor0.0028.631
Q4g075Serpinb1aLeukocyte elastase inhibitor a0.0018.260
Q6ldz3PtprcReceptor-type tyrosine-protein phosphatase c0.0238.077
Q9wuq4SlpiSecretory leukocyte peptidase inhibitor precursor0.0077.093
Q9erl1CybbCytochrome b-245, beta polypeptide0.0097.056
E0a3n4Serpina3nSerine protease inhibitor a3n0.0137.049
G3v6k1Tcn2Transcobalamin-2 precursor0.0176.577
P14669Anxa3Annexin a30.0105.005
P22985XdhXanthine dehydrogenase/oxidase0.0034.802
Q91zn1Coro1aCoronin-1a0.0014.694
P05982Nqo1Nad(p)h quinone dehydrogenase 10.0014.334
P23640Rab27aRas-related protein rab-27a0.0193.830
Q6ifu9Krt16Keratin, type i cytoskeletal 160.0013.797
Q62894Ecm1Extracellular matrix protein 10.0393.655
Q5×i38Lcp1Plastin-20.0033.469
P07150Anxa1Annexin a10.0033.446
Q78zr5HopxHomeodomain-only protein0.0023.376
P01015AgtAngiotensinogen angiotensin-1 angiotensin-2 angiotensin-30.0103.300
Q6axy8Dhrs1 Dehydrogenase/reductase sdr family member 10.0063.287
Q91w30Akr1b8Aldose reductase-related protein 20.0073.244
P32755Hpd 4-hydroxyphenylpyruvate dioxygenase0.0243.149
Q499n7Ptpn6Tyrosine-protein phosphatase non-receptor type 60.0383.025
G3v755Sprr1aCornifin-a0.0023.006
Q5×fv4Fabp4Fatty acid-binding protein, adipocyte0.0242.910
B1wbv8Pld4Phospholipase d40.0112.909
D3zpf9Serpinb12Serpin b120.0382.880
Q4qqv6Lsp1Lymphocyte specific 10.0012.665
P29524Serpinb2Plasminogen activator inhibitor 2 type a0.0012.653
O55162Lypd3Ly6/plaur domain-containing protein 30.0042.551
D4a5u3Tgm3Protein-glutamine gamma-glutamyltransferase e protein0.0332.547
D3zsh7Col17a1Collagen alpha-1(xvii) chain0.0022.485
D3zjk2Serpinb3aProtein serpinb3a0.0382.445
Q6ie17Stfa2l2Stefin-30.0052.439
Q5u206Calml3Calmodulin-like protein 30.0132.429
Q4v885Colec12Collectin-120.0170.547
D3zqi1Gpx7Glutathione peroxidase 7 precursor0.0360.541
D3z9m5Fkbp7Peptidyl-prolyl cis-trans isomerase fkbp7 precursor0.0140.530
O88201Clec11aC-type lectin domain family 11 member a0.0470.529
D3zrd3Pde6dRetinal rod rhodopsin-sensitive cgmp 3′,5′-cyclic phosphodiesterase subunit delta0.0090.528
P21807PrphPeripherin0.0080.527
G3v6m4Capn6Calpain-60.0240.514
D3zg88Sssca1Sjogren syndrome/scleroderma autoantigen 1 homolog0.0340.502
Q2eja0Yap1Yorkie homolog0.0020.494
Q3b7u1Maged2Melanoma-associated antigen d20.0030.492
O35276Nrp2Neuropilin-20.0150.491
D3zun5Pofut2Gdp-fucose protein o-fucosyltransferase 2 precursor0.0180.490
P70583; d4a6v3DutDeoxyuridine 5′-triphosphate nucleotidohydrolase0.0210.489
P19527NeflNeurofilament light polypeptide0.0260.479
M0r649Exoc4Exocyst complex component 40.0310.466
Q99pd6Tgfb1i1Transforming growth factor beta-1-induced transcript 1 protein0.0480.466
P54001P4ha1Prolyl 4-hydroxylase subunit alpha-10.0190.461
D3zt07Sept5Septin-50.0460.452
P12839; g3v7s2NefmNeurofilament medium polypeptide0.0200.444
B5df50Galnt2Polypeptide n-acetylgalactosaminyltransferase 20.0380.436
D3zuq0Rilpl1Rilp-like protein 10.0410.418
D4a8h3Uba6Ubiquitin-like modifier-activating enzyme 60.0240.415
D4a9u4ElnElastin0.0410.409
D4ad75Dpy19l1Protein dpy-19 homolog 10.0140.408
Q6p7d4Cyp20a1Cytochrome p450 20a10.0070.406
Q5×i28Raver1Ribonucleoprotein ptb-binding 10.0450.398
P09117Aldoc Fructose-bisphosphate aldolase c0.0040.396
D3zct5Pald1Paladin0.0040.395
A1l1k3Anapc5Anaphase-promoting complex subunit 50.0280.392
P62966Crabp1Cellular retinoic acid-binding protein 10.0160.384
Q569b7Rwdd4Rwd domain-containing protein 40.0400.384
Q5hze4Mri1 Methylthioribose-1-phosphate isomerase0.0110.376
F1lqz3Kif3aKinesin family member 3a0.0110.376
O88752Hbe1Hemoglobin, epsilon 10.0330.375
Q5u1z0Rab3gap2Rab3 gtpase-activating protein non-catalytic subunit0.0030.373
A1a5r1Rbfox1Fox-1 homolog c0.0330.366
D4a845Rpa3Replication protein a 14 kda subunit0.0210.366
D3zwc6Sntb1 Beta-1-syntrophin0.0030.365
G3v8m1Pold1Dna polymerase delta catalytic subunit0.0030.353
P23565Ina Alpha-internexin0.0390.352
Q4klk9Ssu72Rna polymerase ii subunit a c-terminal domain phosphatase ssu720.0250.349
F1mah6Cdh11Cadherin 110.0070.326
Q6ayg3PrunePrune homolog (drosophila) (ec:3.6.1.1)0.0010.278
P04638Apoa2Apolipoprotein a-ii0.0100.270
Q9z2z8Dhcr7 7-dehydrocholesterol reductase0.0000.221
Q10758Krt8Keratin, type ii cytoskeletal 80.0020.214
Q812d3Ppil3Peptidyl-prolyl cis-trans isomerase-like 30.0000.188
G3v8r3HbzHemoglobin, zeta0.0040.128
B5dfl9Sestd1Sec14 and spectrin domains 10.0000.065
Q9eph1A1bg Alpha-1b-glycoprotein0.0160.033

[i] Group 1, upper-left lip of fetus at 72 h after modeling in GA16.5 rats; Group 3, upper-left lip of fetus at 72 h after modeling in GA18.5 rats. GA, gestational age.

Bioinformatics analysis

Gene ontology (GO) enrichment analysis was performed on the differentially expressed proteins, and their properties were generally described as biological process (BP), molecular function (MF) or cellular component (CC). The first 10 GO enrichment results from each group are displayed in Fig. 13. The results demonstrated that 73, 542 and 376 differentially expressed proteins were significantly enriched between groups 1 and 2, 3 and 4 and 3 and 1, respectively. The results of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis identified the possible pathway related to the differentially expressed proteins between groups (Fig. 14).

In addition, the interaction network of the differentially expressed proteins that regulate wound repair were analyzed. Examples of the interaction networks of the differentially expressed proteins involved in wound repair are as follows: i) Smad4, Tgf1i1, Ptpn6 and Hmox1 in group 1 and 2; ii) S100a9, Fgg, Anxa1, Fgb, Plg and S100a8 in group 3 and 4; and iii) CD36, S100a9, S100a8, Cd9Fgg, Anxa1, Fgb, Plg and S100a8 in group 3 and 1 (Fig. 15).

RT-qPCR analysis of possible target protein in cleft lip repair

RNA was extracted from tissue samples with TRIzol® reagent and the quality was checked using gel electrophoresis. Relative mRNA levels were analyzed using RT-qPCR (Fig. 16). The relative mRNA expression levels of Smad4 were significantly higher in group 2, compared with group 1 (P<0.05). Moreover, the relative mRNA expression levels of Fabp5 were significantly lower in groups 4 and 1, compared with group 3 (P<0.05). Additionally, the relative mRNA expression levels of S100a4 were significantly lower in group 4, compared with group 3 (P<0.05). S100a8 and S100a9 were significantly higher in group 3, compared with in groups 1 and 4 (P<0.05).

Immunofluorescence results

Immunofluorescence staining of Smad4, Fabp5, S100a4, S100a8 and S100a9 was performed on samples from both the GA16.5 and GA18.5 groups 72 h post-surgery. The expression levels of all five proteins increased in GA18.5 compared to GA16.5, and the differences were statistically significant (P<0.05; Fig. 17).

PRM analysis of differential protein expression

The differences in multiple variations of Smad4 expression were compared between groups 1 and 2. The panel reaction monitoring calculated this difference as 0.557, indicating downregulation in group 1 compared with in group 2 (P=0.043) (Table II). In contrast, no statistically significant differences were observed between groups 3 and 4. The difference in multiple variations of Fabp5 between groups 3 and 4 was calculated as 2.91, indicating upregulation in group 3 compared with in group 4 (P=0.024) (Table III). Additionally, the expression levels of Fabp5 were upregulated (P=0.01) in group 3 compared with in group 1; however, the difference between the variations present in groups 1 and 2 was not statistically significant. The difference in the multiple variations of S100a4 and S100a8 between groups 3 and 4 was calculated as 2.897 and 92.828, respectively, indicating an upregulation of the expression levels of both proteins in group 3 (P=0.001 and P=0.002, respectively) (Table III). Furthermore, the difference in the multiple variations of S100a8 between groups 3 and 1 was 25.933, which indicates upregulation in group 3 (P=0.002) (Table IV). However, the differences were not statistically significant between groups 1 and 2. The difference in the multiple variations of S100a9 was 30.191 and 19.538 between groups 3 and 4 and groups 3 and 1, respectively, suggesting upregulation in group 3 (P=0.0004 and P=0.001, respectively) (Tables III and IV). In contrast, the difference in the multiple variations of S100a9 between groups 1 and 2 was not statistically significant (Tables IIIV).

Table II.

Differential protein expression in group 1 and group 2.

Table II.

Differential protein expression in group 1 and group 2.

Protein IDGene nameProtein nameP-valueFold-change
G3V8R3HbzHemoglobin, zeta0.0047.788
B2RYS8Ndufb8NADH dehydrogenase0.0005.024
Q920P6AdaAdenosine deaminase0.0384.831
O88752Hbe1Hemoglobin, epsilon 10.0034.801
Q499N7Ptpn6Tyrosine-protein phosphatase non-receptor type 60.0034.700
Q4FZU2Krt6aKeratin 6A0.0014.044
P06762Hmox1Heme oxygenase 10.0113.902
Q6IFU9Krt16Keratin, type I cytoskeletal 160.0063.613
Q99PD6Tgfb1i1Transforming growth factor beta-1-induced transcript 1 protein0.0043.590
Q6P7S1Asah1Acid ceramidase0.0293.167
Q63066Hbg1Hemoglobin, gamma A0.0023.082
Q10758Krt8Keratin, type II cytoskeletal 80.0042.901
Q6AYQ4Tmem109Transmembrane protein 1090.0482.710
Q9Z2Q7Stx8Syntaxin-80.0352.660
G3V9M8Fam50aProtein fam50a0.0342.515
M0R9Y3Nup43Nucleoporin 430.0052.461
B2GVB9Fermt3Fermitin family homolog 30.0182.425
G3V8HOlfml3Olfactomedin-like protein 3 precursor0.0452.383
D4A531Polr2iRna polymerase ii subunit i0.0492.290
Q68FS1Nubp2Cytosolic Fe-S cluster assembly factor0.0282.221
D3ZLS5Hectd1Hect domain e3 ubiquitin protein ligase 10.0332.112
D4A0M2NxnNucleoredoxin0.0332.056
Q6IE17Stfa2l2Stefin-30.0071.969
P27139Ca2Carbonic anhydrase 20.0321.957
D3ZF44LOC684499Protein LOC6844990.0151.940
Q6LDZ3PtprcReceptor-type tyrosine-protein phosphatase C0.0071.878
Q5XI38Lcp1Plastin-20.0181.843
Q5PPG2LgmnLegumain precursor0.0151.820
P06765Pf4Platelet factor 40.0501.708
Q9R1T3CtszCathepsin Z0.0111.707
Q5U1Y2Rac2Ras-related C3 botulinum toxin substrate 20.0211.669
Q5U2V4Plbd1Phospholipase B-like 10.0281.630
Q9EPX0Hspb8Heat shock protein beta-80.0051.603
O35532Msmo1Methylsterol monooxygenase 10.0371.592
Q91ZN1Coro1aCoronin-1A0.0131.586
O88201Clec11aC-type lectin domain family 11 member A0.0251.547
Q5U329Slc4a1Band 3 anion transport protein0.0271.512
Q496Z5PrphPeripherin0.0410.626
P19527NeflNeurofilament light polypeptide0.0400.608
Q9ESI7DcxNeuronal migration protein doublecortin0.0030.597
Q6AY98Ube2e2Ubiquitin conjugating enzyme e2 e20.0460.577
Q7TSX7Nr3c1;grGlucocorticoid receptor0.0260.560
O70437Smad4Mothers against decapentaplegic homolog 40.0430.557
F1M754Map4k4Mitogen-activated protein kinase kinase kinase kinase 40.0220.526
D4A2Z8Dhx36Probable ATP-dependent RNA helicase DHX360.0090.522
P31430Dpep1Dipeptidase 10.0100.513
Q6AXY8Dhrs1 Dehydrogenase/reductase SDR family member 10.0190.495
D4A414Cox15COX15 homolog0.0310.476
D4ABV5Calm1Calmodulin 10.0120.473
D3ZRN3Actbl2Beta-actin-like protein 20.0480.413
Q8CGS4Chmp3Charged multivesicular body protein 30.0220.410
D3ZHA7Myl6bMyosin light chain 6b0.0110.390
P70541Eif2b3Translation initiation factor eif-2B subunit gamma0.0020.324
D3ZX50Krtap11-1Uncharacterized protein0.0370.287
D3ZD07Fmo9Flavin containing monooxygenase 9 pseudogene0.0070.277
Q6IFX1Krt24Keratin, type I cytoskeletal 240.0120.050
Q6IG02Krt2Keratin, type II cytoskeletal 20.0080.021

[i] Group 1: Upper-left lip of fetus at 72 h after modeling in GA16.5 rats; Group 2, upper-right lip of fetus at 72 h after modeling in GA16.5 rats. GA, gestational age.

Table III.

Differential protein expression in group 3 and group 4.

Table III.

Differential protein expression in group 3 and group 4.

Protein IDGene nameProtein nameP-valueFold change
D3ZGE2Mpo Myeloperoxidase0.000377.923
Q62714Np4Neutrophil antibiotic peptide NP-40.000231.771
D3ZY96NgpNeutrophilic granule protein precursor0.010226.724
P50115S100a8S100 Calcium Binding Protein A80.00192.828
Q7TP54Fam65bProtein FAM65B0.00077.718
D3ZMI6Olfm4Olfactomedin-4 precursor0.00163.833
D4A081Setdb1Histone-lysine N-methyltransferase SETDB10.00047.032
Q9JI30ItgamIntegrin alpha-M precursor0.01139.489
B2RYB8Itgb2Integrin beta 2 precursor0.00334.443
P50116S100a9S100 Calcium Binding Protein A90.00030.191
Q920P6AdaAdenosine deaminase0.02827.433
Q499N7Ptpn6Tyrosine-protein phosphatase non-receptor type 60.00323.157
Q9ERL1CybbCytochrome b-245, beta polypeptide0.00221.484
Q9JKB7GdaGuanine deaminase0.01920.202
Q5U1Y2Rac2Ras-related C3 botulinum toxin substrate 20.00719.291
Q6IFU9Krt16Keratin, type I cytoskeletal 160.00113.524
O54854Klk6Kallikrein-6 precursor0.00011.605
B2GVB9Fermt3Fermitin family homolog 30.01211.176
Q5PQW8Gbp2Interferon-induced guanylate-binding protein 20.01910.854
Q6LDZ3PtprcReceptor-type tyrosine-protein phosphatase C0.01510.626
Q4G075Serpinb1aLeukocyte elastase inhibitor A0.0019.051
Q6PDV1Lyz1Lysozyme C-1 precursor0.0029.049
Q6IE17Stfa2l2Stefin-30.0008.930
Q5U2V4Plbd1Phospholipase B-like 10.0038.669
Q91ZN1Coro1aCoronin-1A0.0018.199
P14669Anxa3Annexin A30.0088.100
Q9R0D6Tcn2Transcobalamin-2 precursor0.0147.286
Q4QQV6Lsp1Lymphocyte specific 10.0046.785
P06768Rbp2Retinol-binding protein 20.0066.051
Q5XI38Lcp1Plastin-20.0015.841
Q91W30Akr1b8Aldo-Keto Reductase Family 1 Member B80.0015.492
Q63015Csap1Common salivary protein 1 precursor0.0015.454
P31720C1qaComplement C1q subcomponent subunit A0.0085.339
G3V904Pld4Phospholipase D40.0054.857
D4ADD7Glrx5 Glutaredoxin-related protein 50.0024.782
P22985XdhXanthine dehydrogenase/oxidase0.0034.221
P06866HpHaptoglobin Haptoglobin alpha chain Haptoglobin beta chain0.0023.945
B2RYS9Trmt112Uncharacterized protein0.0163.827
P23640Rab27aRas-related protein Rab-27A0.0173.774
P06762Hmox1Heme oxygenase 10.0083.769
Q9WUQ4SlpiSecretory leukocyte peptidase inhibitor precursor0.0153.710
P07150Anxa1Annexin A10.0033.449
D3ZX79Ly6g6cLymphocyte antigen 6 complex G6C precursor0.0183.236
O88752Hbe1Hemoglobin, epsilon 10.0293.211
Q9R1T3CtszCathepsin Z0.0023.195
D3ZJH9Me2NAD-dependent malic enzyme, mitochondrial0.0342.926
P05942S100a4S100 Calcium Binding Protein A40.0022.897
Q5XJW6CfhComplement factor H precursor0.0082.891
O54892Hk2Hexokinase-20.0072.876
Q6P7D4Cyp20a1Cytochrome P450 20A10.0130.490
D3ZWC6Sntb1 beta-1-syntrophin0.0250.490
Q62997Gfra1GDNF family receptor alpha-10.0430.488
P02600Myl1Myosin light chain 1/30.0110.482
O35878Hspb2Heat shock protein beta-20.0050.481
P17209Myl4Myosin light chain 40.0040.475
D4A8H3Uba6Ubiquitin-like modifier-activating enzyme 60.0310.471
A1L1K3Anapc5Anaphase-promoting complex subunit 50.0460.470
D3ZTW9ExogNuclease EXOG0.0280.467
D4A3D2Smyd1SET and MYND domain-containing protein 10.0040.465
P04466MylpfMyosin regulatory light chain 20.0090.464
P12847Myh3Myosin-30.0070.461
P13413Tnni1Troponin I0.0010.460
D4A4Y2Hsd17b14 17-beta-hydroxysteroid dehydrogenase 140.0330.455
P23928CryabAlpha-crystallin B chain0.0200.454
Q7TNB2Tnnt1Troponin T0.0020.451
D3ZCD7Tp53rkTP53-regulating kinase0.0040.450
P00564CkmCreatine kinase M-type0.0370.450
Q80W59HrcSarcoplasmic reticulum histidine-rich calcium-binding protein precursor0.0190.445
P50463Csrp3Cysteine and glycine-rich protein 30.0130.444
Q5XIG1Ldb3Ldb3 protein0.0170.443
D3ZUB7Anapc4Anaphase-promoting complex subunit 40.0300.442
Q64578Atp2a1ATPase, Ca++ transporting, cardiac muscle, fast twitch 10.0320.442
Q6P792Fhl1Four and a half LIM domains protein 10.0130.431
Q8K4F2Alox15bArachidonate 15-lipoxygenase B0.0250.428
M0RBL8Tceal6Protein LOC6799740.0030.427
P51868Casq2Calsequestrin-2 precursor0.0080.425
B4F789Apobec2Probable C->U-editing enzyme APOBEC-20.0080.421
P16290Pgam2Phosphoglycerate mutase 20.0140.418
Q9Z2J4NexnNexilin0.0020.412
Q9QYU4Crym Thiomorpholine-carboxylate dehydrogenase0.0170.411
D3ZUQ0Rilpl1RILP-like protein 10.0060.409
D4A2H6Rbfox3Fox-1 homolog C0.0370.408
D3ZVM5Hspa12bHeat shock 70 kDa protein 12B0.0380.406
O54747Pold1DNA polymerase delta catalytic subunit0.0010.403
P52481Cap2Adenylyl cyclase-associated protein 20.0070.396
Q63544Sncg Gamma-synuclein0.0040.381
Q496Z5PrphPeripherin0.0010.376
P07483Fabp3Fatty acid-binding protein, heart0.0110.357
P23565Ina Alpha-internexin0.0050.332
D4ADS4Mgst3Microsomal glutathione S-transferase 30.0240.328
P19527NeflNeurofilament light polypeptide0.0060.326
P12839NefmNeurofilament medium polypeptide0.0040.326
B2RZ77DptDermatopontin precursor0.0240.320
Q6AYG3PrunePrune homolog0.0170.320
G3V7K1Myom2Myomesin 20.0250.299
G3V6V5Atp1b4Protein ATP1B40.0050.272
Q9Z2Z8Dhcr7 7-dehydrocholesterol reductase0.0000.270
P19633Casq1 Calsequestrin-10.0210.201
D3ZX18Myoz2Myozenin-20.0010.198
Q812D3Ppil3Peptidyl-prolyl cis-trans isomerase-like 30.0000.179

[i] Group 3, upper-left lip of fetus at 72 h after modeling in GA18.5 rats; Group 4, upper-right lip of fetus at 72 h after modeling in GA18.5 rats. GA, gestational age.

Discussion

In recent decades, various animal models of congenital cleft lip have been successfully established through surgical induction (4,20). It has been suggested that intrauterine cleft lip repair can effectively improve this defect and reduce the impact of scars on normal facial development after birth. Thus, it also provides a new way for the effective repair of congenital cleft lips. In the present study, pregnant SD rats were used to establish a fetal rat model of cleft lip wound at two time points, GA16.5 and GA18.5. The different pregnancy models were induced by using two different repair methods of a cleft lip wound of the fetus (21,22). The exact gestational age is particularly important for the results of the repair of cleft lip in the fetal rats. Thus, the use of a rat model provides an added advantage in that the exact time of conception can be replicated, thereby minimizing differences between groups.

The present findings confirmed the hypothesis that fetal rat defects can be regenerated during early pregnancy without scar formation (23). It was also demonstrated that fetal rat defects could not be completely regenerated in late pregnancy and resulted in scarring (24). Furthermore, the expression of pro-inflammatory factors was different between the two groups. However, these observations were only made at one time point (72 h) after constructing cleft lip wounds in fetal rats. Future studies are needed to examine samples collected at different time points following surgery. Another shortcoming of this study entails the lack of comparison between the cleft lip wound repairs of fetal rats at different ages, such as the fetus in the early stages of pregnancy, or in newborn and/or adult rats. Label-free quantitative proteomics were used to examine proteins that play important roles in the postoperative repair process of fetal cleft lip. Protein expression was examined in four groups of samples. In addition, bioinformatics analysis was conducted to identify potential biological markers, providing a theoretical reference and methodological basis for the examination of relevant mechanisms underlying fetal intrauterine scar repair. However, further studies are required to determine whether any one protein or several proteins, plays a key role in wound healing.

Smad4 belongs to the family of Smad proteins and is a common mediator in the signal transduction processes of the TGF-β family (25). TGF-β expression can lead to fibroblast proliferation and ECM deposition (26,27). The present findings indicated that the mRNA and protein expression levels of Smad4 were downregulated in the scar-free repair group.

Furthermore, the mRNA and protein expression levels of Fabp5 were upregulated in the scar formation group. Therefore, it may be hypothesized that Fabp5 could be involved in the fibrosis of the fetal cleft lip wound, which may be mediated by the TGF-β signaling pathway (2830).

S100a4 is a member of the S100 calcium-binding protein family, and its expression is associated with various non-neoplastic diseases, such as chronic obstructive pulmonary disease and cardiac hypertrophy (3135). The present study demonstrated that the mRNA and protein expression levels of S100a4 were upregulated in the scar formation group, which may be associated with scar repair of fetal rat cleft lip wounds.

S100a8 is also a member of the S100 calcium-binding protein family (3642). mRNA and protein expression levels of S100a8 were significantly upregulated in the scar repair group in the present study, indicating a potential role for S100a8 in the process of fetal cleft lip wound healing.

Current reports frequently associate S100a9, a member of the calcium-binding protein family S100, with infectious diseases, immune diseases and tumors, such as non-small cell lung adenocarcinoma (4345). mRNA and protein expression levels of S100a9 were significantly upregulated in the scar formation group. Therefore, we speculated that S100a9 may play an important role in the process of fetal wound healing. However, whether the reduced expression levels of Fabp5, S100a4, S100a8 and S100a9 in the third trimester of pregnancy would reduce or worsen scar formation remains unclear. Further functional testing and regulatory studies are required to confirm the role of these five differentially expressed proteins in fetal wound repair.

The cleft lip is a very common congenital condition that often leaves life-long scarring. The present study identified five differentially expressed proteins, namely Smad4, Fabp5, S100a4, S100a8 and S100a9, that may be potential biomarkers of the scarless repair process in fetal rat cleft lip wounds. These findings may facilitate the discovery of new clinical targets for the prevention and treatment of scars. However, the role of these proteins in fetal wound repair and potential underlying mechanisms require further examination.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the National Science and Basic Resources Survey Special Foundation, China (grant no. 2017FY101204), the Technology Innovation Guide Program of Hunan Province, China (grant no. 2017SK50124), the Science and Technology Major Project of Hunan Province, China (grant no. 2019SK1010) and the Science and Technology Major Project of Hunan Province, China (grant no. 2019SK1015).

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

YY, FH and JZho conceived and designed research. YY and FH performed animal experiments and staining. YY, HL, PJ, FH, JY, KG, SH and JZha performed PCR and label-free quantification PRM. YY, FH, JC, JY, ZC, AW and JZha analyzed data. YY and FH prepared figures. YY drafted the manuscript. FH and JZho edited and revised the manuscript. YY, FH, HL AW, PJ and JZho approved the final version of the manuscript. FH and JZho confirmed the authenticity of all of the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study followed the regulations stipulated by the People's Republic of China regarding the Management of Experimental Animals and was approved by The Animal Experiment Management and Medical Ethics Sub-committee of The Third Xiangya Hospital of Central South University, Hunan, China (approval no. 2014-S168).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

VanKoevering KK, Morrison RJ, Prabhu SP, Torres MF, Mychaliska GB, Treadwell MC, Hollister SJ and Green GE: Antenatal three-dimensional printing of aberrant facial anatomy. Pediatrics. 136:e1382–e1385. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Burrington JD: Wound healing in the fetal lamb. J Pediatr Surg. 6:523–528. 1971. View Article : Google Scholar : PubMed/NCBI

3 

Beanes SR, Hu FY, Soo C, Dang CM, Urata M, Ting K, Atkinson JB, Benhaim P, Hedrick MH and Lorenz HP: Confocal microscopic analysis of scarless repair in the fetal rat: Defining the transition. Plast Reconstr Surg. 109:160–170. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Walmsley GG, Hu MS, Hong WX, Maan ZN, Lorenz HP and Longaker MT: A mouse fetal skin model of scarless wound repair. J Vis Exp. 16:522972015.

5 

Dang CM, Beanes SR, Lee H, Zhang X, Soo C and Ting K: Scarless fetal wounds are associated with an increased matrix metalloproteinase-to-tissue-derived inhibitor of metalloproteinase ratio. Plast Reconstr Surg. 111:2273–2285. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Longaker MT, Whitby DJ, Adzick NS, Crombleholme TM, Langer JC, Duncan BW, Bradley SM, Stern R, Ferguson MW and Harrison MR: Studies in fetal wound healing, VI. Second and early third trimester fetal wounds demonstrate rapid collagen deposition without scar formation. J Pediatr Surg. 25:63–69. 1990. View Article : Google Scholar : PubMed/NCBI

7 

Lorenz HP, Whitby DJ, Longaker MT and Adzick NS: Fetal wound healing. The ontogeny of scar formation in the non-human primate. Ann Surg. 217:391–396. 1993. View Article : Google Scholar : PubMed/NCBI

8 

Cass D, Bullard KM, Sylvester KG, Yang EY, Longaker MT and Adzick NS: Wound size and gestational age modulate scar formation in fetal wound repair. J Pediatr Surg. 32:411–415. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Longaker MT and Adzick NS: The biology of fetal wound healing: A review. Plast Reconstr Surg. 87:788–798. 1991. View Article : Google Scholar : PubMed/NCBI

10 

Armstrong JR and Ferguson MW: Ontogeny of the skin and the transition from scar-free to scarring phenotype during wound healing in the pouch young of a marsupial, Monodelphis domestica. Dev Biol. 169:242–260. 1995. View Article : Google Scholar : PubMed/NCBI

11 

Stern M, Dodson TB, Longaker MT, Lorenz HP, Harrison MR and Kaban LB: Fetal cleft lip repair in lambs: Histologic characteristics of the healing wound. Int J Oral Maxillofac Surg. 22:371–374. 1993. View Article : Google Scholar : PubMed/NCBI

12 

Longaker MT, Dodson TB and Kaban LB: A rabbit model for fetal cleft lip repair. J Oral Maxillofac Surg. 48:714–719. 1990. View Article : Google Scholar : PubMed/NCBI

13 

Oberg KC, Evans ML, Nguyen T, Peckham NH, Kirsch WM and Hardesty RA: Intrauterine repair of surgically created defects in mice (lip incision model) with a microclip: Preamble to endoscopic intrauterine surgery. Cleft Palate Craniofac J. 32:129–137. 1995. View Article : Google Scholar : PubMed/NCBI

14 

Hu F, Yan Y, Wang CW, Liu Y, Wang JJ, Zhou F, Zeng QH, Zhou X, Chen J, Wang AJ and Zhou JD: Article effect and mechanism of ganoderma lucidum polysaccharides on human fibroblasts and skin wound healing in mice. Chin J Integr Med. 25:203–209. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Xue YN, Yan Y, Chen ZZ, Chen J, Tang FJ, Xie HQ, Tang SJ, Cao K, Zhou X, Wang AJ and Zhou JD: LncRNA TUG1 regulates FGF1 to enhance endothelial differentiation of adipose-derived stem cells by sponging miR-143. J Cell Biochem. 120:19087–19097. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Guo H, Chen T, Liang Z, Fan L, Shen Y and Zhou D: iTRAQ and PRM-based comparative proteomic profiling in gills of white shrimp Litopenaeus vannamei under copper stress. Chemosphere. 263:1282702021. View Article : Google Scholar : PubMed/NCBI

17 

Chen PS, Li YP and Ni HF: Morphology and evaluation of renal fibrosis. Adv Exp Med Biol. 1165:17–36. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Luo Y, Yan Y, Zhang S and Li Z: Computational approach to investigating key GO terms and KEGG pathways associated with CNV. Biomed Res Int. 2018:84068572018. View Article : Google Scholar : PubMed/NCBI

19 

Dong X, Landford WN, Hart J, Risolino M, Kaymakcalan O, Jin J, Toyoda Y, Ferretti E, Selleri L and Spector JA: Toward microsurgical correction of cleft lip ex utero through restoration of craniofacial developmental programs. Plast Reconstr Surg. 140:75–85. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Stelnicki EJ, Lee S, Hoffman W, Lopoo J, Foster R, Harrison MR and Longaker MT: A long-term, controlled-outcome analysis of in utero versus neonatal cleft lip repair using an ovine model. Plast Reconstr Surg. 104:607–615. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Harling TR, Stelnicki EJ, Hedrick MH and Longaker MT: In utero models of craniofacial surgery. World J Surg. 27:108–116. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Mast BA, Haynes JH, Krummel TM, Diegelmann RF and Cohen IK: In vivo degradation of fetal wound hyaluronic acid results in increased fibroplasia, collagen deposition, and neovascularization. Plast Reconstr Surg. 89:503–509. 1992. View Article : Google Scholar : PubMed/NCBI

23 

Frantz FW, Diegelmann RF, Mast BA and Cohen IK: Biology of fetal wound healing: Collagen biosynthesis during dermal repair. J Pediatr Surg. 27:945–949. 1992. View Article : Google Scholar : PubMed/NCBI

24 

Wilgus TA: Regenerative healing in fetal skin: A review of the literature. Ostomy Wound Manage. 53:16–33. 2007.PubMed/NCBI

25 

Hu HH, Chen DQ, Wang YN, Feng YL, Cao G, Vaziri ND and Zhao YY: New insights into TGF-beta/Smad signaling in tissue fibrosis. Chem Biol Interac. 292:76–83. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Honardoust D, Ding J, Varkey M, Shankowsky HA and Tredget EE: Deep dermal fibroblasts refractory to migration and decorin-induced apoptosis contribute to hypertrophic scarring. J Burn Care Res. 33:668–677. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Wu C, Jiang J, Boye A, Jiang Y and Yang Y: Compound Astragalus and Salvia miltiorrhiza extract suppresses rabbits' hypertrophic scar by modulating the TGF-β/Smad signal. Dermatology. 229:363–368. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Furuhashi M, Ogura M, Matsumoto M, Yuda S, Muranaka A, Kawamukai M, Omori A, Tanaka M, Moniwa N, Ohnishi H, et al: Serum FABP5 concentration is a potential biomarker for residual risk of atherosclerosis in relation to cholesterol efflux from macrophages. Sci Rep. 7:2172017. View Article : Google Scholar : PubMed/NCBI

29 

Yeung DC, Wang Y, Xu A, Cheung SC, Wat NM, Fong DY, Fong CH, Chau MT, Sham PC and Lam KS: Epidermal fatty-acid-binding protein: A new circulating biomarker associated with cardio-metabolic risk factors and carotid atherosclerosis. Eur Heart J. 29:2156–2163. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Song J, Zhang H, Wang Z, Xu W, Zhong L, Cao J, Yang J, Tian Y, Yu D, Ji J, et al: The role of FABP5 in radiation-induced human skin fibrosis. Radiat Res. 189:177–186. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Fei F, Qu J, Li C, Wang X, Li Y and Zhang S: Role of metastasis-induced protein S100A4 in human non-tumor pathophysiologies. Cell Biosci. 7:642017. View Article : Google Scholar : PubMed/NCBI

32 

Grotterød I, Maelandsmo GM and Boye K: Signal transduction mechanisms involved in S100A4-induced activation of the transcription factor NF-kappaB. BMC Cancer. 10:2412010. View Article : Google Scholar

33 

Schneider M, Kostin S, Strøm CC, Aplin M, Lyngbaek S, Theilade J, Grigorian M, Andersen CB, Lukanidin E, Lerche Hansen J and Sheikh SP: S100A4 is upregulated in injured myocardium and promotes growth and survival of cardiac myocytes. Cardiovasc Res. 75:40–50. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Tomcik M, Palumbo-Zerr K, Zerr P, Avouac J, Dees C, Sumova B, Distler A, Beyer C, Cerezo LA, Becvar R, et al: S100A4 amplifies TGF-β-induced fibroblast activation in systemic sclerosis. Ann Rheum Dis. 74:1748–1755. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Zhao YX, Ho CK, Xie Y, Chen YH, Li HZ, Zhang GY and Li QF: Calcimycin suppresses S100A4 expression and inhibits the stimulatory effect of transforming growth factor β1 on Keloid fibroblasts. Ann Plast Surg. 81:163–169. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Donato R: Intracellular and extracellular roles of S100 proteins. Microsc Res Tech. 60:540–551. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Lin H, Andersen GR and Yatime L: Crystal structure of human S100A8 in complex with zinc and calcium. BMC Struct Biol. 16:82016. View Article : Google Scholar : PubMed/NCBI

38 

Bouzidi F and Doussiere J: Binding of arachidonic acid to myeloid-related proteins (S100A8/A9) enhances phagocytic NADPH oxidase activation. Biochem Biophys Res Commun. 325:1060–1065. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Gebhardt C, Németh J, Angel P and Hess J: S100A8 and S100A9 in inflammation and cancer. Biochem Pharmacol. 72:1622–1631. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Basso D, Bozzato D, Padoan A, Moz S, Zambon CF, Fogar P, Greco E, Scorzeto M, Simonato F, Navaglia F, et al: Inflammation and pancreatic cancer: Molecular and functional interactions between S100A8, S100A9, NT-S100A8 and TGFβ1. Cell Commun Signal. 26:12–20. 2014.PubMed/NCBI

41 

Shabani F, Farasat A, Mahdavi M and Gheibi N: Calprotectin (S100A8/S100A9): A key protein between inflammation and cancer. Inflamm Res. 67:801–812. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Yaundong L, Dongyan W, Lijun H and Zhibo X: Effects of downregulation of S100A8 protein expression on cell cycle and apoptosis of fibroblasts derived from hypertrophic scars. Aesthet Surg J. 34:160–167. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Hessian PA, Edgeworth J and Hogg N: MRP-8 and MRP-14, two abundant Ca (2+)-binding proteins of neutrophils and monocytes. J Leukoc Biol. 53:197–204. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Markowitz J and Carson WE III: Review of S100A9 biology and its role in cancer. Biochim Biophys Acta. 1835:100–109. 2013.PubMed/NCBI

45 

Zhong A, Xu W, Zhao J, Xie P, Jia S, Sun J, Galiano RD, Mustoe TA and Hong SJ: S100A8 and S100A9 are induced by decreased hydration in the epidermis and promote fibroblast activation and fibrosis in the dermis. Am J Pathol. 186:109–122. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2021
Volume 24 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
Yan Y, Liu H, Yi J, Chen Z, Chen J, Zhang J, Gao K, He S, Wang A, Jin P, Jin P, et al: Identification of differentially expressed proteins involved in fetal scarless wound healing using a rat model of cleft lip. Mol Med Rep 24: 596, 2021
APA
Yan, Y., Liu, H., Yi, J., Chen, Z., Chen, J., Zhang, J. ... Zhou, J. (2021). Identification of differentially expressed proteins involved in fetal scarless wound healing using a rat model of cleft lip. Molecular Medicine Reports, 24, 596. https://doi.org/10.3892/mmr.2021.12235
MLA
Yan, Y., Liu, H., Yi, J., Chen, Z., Chen, J., Zhang, J., Gao, K., He, S., Wang, A., Jin, P., Hu, F., Zhou, J."Identification of differentially expressed proteins involved in fetal scarless wound healing using a rat model of cleft lip". Molecular Medicine Reports 24.2 (2021): 596.
Chicago
Yan, Y., Liu, H., Yi, J., Chen, Z., Chen, J., Zhang, J., Gao, K., He, S., Wang, A., Jin, P., Hu, F., Zhou, J."Identification of differentially expressed proteins involved in fetal scarless wound healing using a rat model of cleft lip". Molecular Medicine Reports 24, no. 2 (2021): 596. https://doi.org/10.3892/mmr.2021.12235