Skip to main content
Advances in Wound Care logoLink to Advances in Wound Care
. 2018 Jan 1;7(1):1–10. doi: 10.1089/wound.2017.0763

Pathway Analysis of Gene Expression of E14 Versus E18 Fetal Fibroblasts

Michael S Hu 1,,2, Mimi R Borrelli 1, Michael Januszyk 1, Anna Luan 1, Samir Malhotra 1, Graham G Walmsley 1, Wan Xing Hong 1, Ruth Tevlin 1, Geoffrey C Gurtner 1, Michael T Longaker 1,,2, Hermann P Lorenz 1,,*
PMCID: PMC5770085  PMID: 29344429

Abstract

Objective: Fetuses early in gestation heal skin wounds without forming scars. The biological mechanisms behind this process are largely unknown. Fibroblasts, however, are cells known to be intimately involved in wound healing and scar formation. We examined fibroblasts in different stages of development to characterize differences in gene expression that may result in the switch from regenerative wound repair to repair with scarring.

Approach: Fibroblasts were isolated and cultured from the back skin of BALB/c wild-type mouse fetuses at embryonic day (E)14 and E18 (n = 10). The fibroblast total RNA was extracted, and microarray analysis was conducted using chips containing 42,000 genes. Significance analysis of microarrays was performed to identify genes with greater than twofold expression difference and a false discovery rate of less than two. Identified genes subsequently underwent enrichment analysis to detect differentially expressed pathways.

Results: Two hundred seventy-five genes were differentially expressed between E14 and E18 in fetal fibroblasts. Thirty genes were significantly downregulated and 245 genes were significantly upregulated at E18 compared with E14. Ingenuity pathway analysis identified the top 20 signaling pathways differentially activated in fetal fibroblasts between the E18 and E14 time points.

Innovation: To our knowledge, this work represents the first instance where differentially expressed genes and signaling pathways between fetal fibroblasts at E14 and E18 have been studied.

Conclusion: The genes and pathways identified here potentially underlie the mechanism behind the transition from fetal wound healing via regeneration to wound healing by repair, and may prove to be key targets for future therapeutics.

Keywords: : wound healing, scarless repair, regeneration, microarray


graphic file with name fig-2.jpg

Hermann P. Lorenz, MD

Introduction

The physiological response to cutaneous injury is a complex and tightly regulated process. Cutaneous injury initiates an acute inflammatory response, followed by a rapid migration of progenitor cells to the wound site. Multipotent cells differentiate into numerous cell types that rapidly form new blood vessels and epithelium. Concurrently, fibroblasts proliferate at the wound site, synthesize collagen, and differentiate into myofibroblasts, which act to contract the wounded skin. Remodeling of the wounded tissue continues far beyond the initial injury, and scarring is characterized by the excess accumulation of extracellular matrix (ECM). The organization of the newly synthesized ECM never recapitulates that found in uninjured skin, resulting in a 20–30% decrease in overall tensile strength in the repaired, compared to normal, skin tissue.1

Interestingly, Rowlatt described the ability of the human fetus, early in gestation, to repair cutaneous wounds without forming a scar.2 Further work has subsequently confirmed these findings in both animal models and in human fetuses.3 The finding of scarless wound healing in human skin suggests there is potential that adult skin can be stimulated to function as fetal skin, and heal via regeneration rather than fibrosis. The mechanisms of scarless healing, however, require further characterization before such knowledge can be applied for therapeutic benefit.

There are numerous differences documented between fetal and adult wound healing phenotypes. Scarless wound healing is age dependent; there is a distinct switch from regenerative to scarring healing around 24 weeks of gestation in human embryos4 and around gestational age 18 days (embryonic day [E]18) in mice.5 Scar-free healing is also dependent on the size of the wound, with larger wounds healing via scarring at earlier gestational ages.4 In vitro, fetal skin fibroblasts are able to simultaneously proliferate and synthesize collagen, whereas adult skin fibroblasts proliferate before they synthesize collagen, suggesting a central role of fibroblasts in the mechanism of scarless wound healing.6 This study focused on the difference in fibroblast function at different ages of gestation. A microarray transcriptional profiling comparison was conducted on fetal fibroblasts harvested from mouse fetuses at E14 and E18 to detect any deviations in transcriptomes between scarless and scarring repair. The aim was to identify novel pathways involving fibroblasts that promote regenerative repair and scarless healing.

Clinical Problem Addressed

Scarring is the expected outcome of the human adult wound healing process and is a significant medical issue that can substantially reduce patients' quality of life. Numerous physiological and psychological consequences result from scarring, either the result of trauma or surgery.7 Facial scars are often cosmetically displeasing and result in substantial psychosocial distress. Keloid scars can cause immense pain and severe itching, while hypertrophic scars can lead to contracture, erosion of skeletal structure, and potentially lifelong disability.8 Scars and their associated consequences surmount to enormous economic cost estimated in the tens of billions of dollars.9

Materials and Methods

Animals

BALB/c wild-type mice at 6 weeks of age were purchased from Charles River Laboratories (Wilmington, MA). After acclimation for at least 1 week, male and female mice were bred overnight. Every day, the female mice were checked and the day of vaginal plug was determined to be E0.5 for gestational timing. All animal procedures were performed in accordance with National Institutes of Health (NIH) guidelines according to university-approved protocols. Mice were closely monitored by the Stanford Administrative Panel on Laboratory Animal Care (APLAC).

Primary cell culture

Gestational age E14 and E18 pregnant mice were sacrificed with CO2 and cervical dislocation. Fetal mice were surgically removed using sterile technique. Under a dissecting microscope, E14 (n = 10) and E18 (n = 10) fetal mouse dorsal skin was collected and pooled for fibroblast isolation and primary cell culture under sterile conditions.

Fibroblast primary cell culture was conducted by mincing tissue and treating it with 0.25% trypsin/ethylenediaminetetraacetic acid (EDTA) in 37°C with mild agitation for 10 min. Mouse embryonic fibroblast culture medium consisting of Dulbecco's modified Eagle's medium, GlutaMAX supplement (Thermo Fisher Scientific, Waltham, MA), 10% fetal bovine serum (Omega Scientific, Tarzana, CA), 0.1 mM 2-mercaptoethanol (Sigma, St. Louis, MO), and 1% penicillin/streptomycin (Thermo Fisher Scientific) was used for cell culture. Cells were cultured at 37°C in a humidified incubator with 5% CO2. All experiments used passage one fibroblasts from male and female fetuses.

RNA extraction and amplification

RNA was extracted using the TRIzol protocol (Invitrogen, Carlsbad, CA) according to the manufacturer's instructions. Subsequently, the MessageAmp antisense RNA (aRNA) kit (Ambion, Austin, TX) was used to amplify 1 μg of extracted RNA from each group. To compare the different arrays between gestational time points, universal mouse RNA was amplified in individual reaction mixtures of 1 μg aliquots simultaneously, with amplifications in the experimental sample and utilized as an internal amplification control.

Preparation of fluorescent complementary DNA probes

Two micrograms of random hexamer and 4 μg of RNA were heated at 65°C for 10 min. Samples were then reverse transcribed with 2 mM of each deoxyribose nucleotide triphosphate, 1 × first-strand buffer, 0.5 μL RNAse inhibitor, 200 U superscript II, 10 mM dithiothreitol, and either 3 μL Cy3-deoxyribose uridine triphosphate (dUTP) (for experimental samples) or Cy5-dUTP (for universal mouse control samples) at 42°C for 1 h in a 30 μL total reaction volume. To increase the reaction, 200 U superscript II was added to the mixture and samples were incubated at 42°C for 1 h. Fluorescent Cy3- or Cy5-labeled probes were washed with Tris-EDTA (TE) buffer (10 mM Tris, 1 mM EDTA) through a microcon mini column (Millipore, Billerica, MA), treated with 450 μL TE buffer, and the inverted mini column was spun into a new tube. Microarray chips were immediately hybridized with probes.

Pretreatment of microarray chips

The Stanford Microarray Database Center was used to print microarray chips with 42,000 specific complementary DNAs (cDNAs) printed onto lysine-coated slides. Sequences and accession numbers can be accessed at http://genome-www5.stanford.edu//index.shtml, where single accession numbers from GenBank represent chosen cDNAs. Microarray chips were rehydrated before hybridization by being held down quickly over distilled water. Microarray chips were then quickly snap-dried over a 100°C heating block and DNA was crosslinked using a UV crosslinker (300 mJ).

Microarray hybridization

Fluorescent-labeled probes were heated at 100°C for 2 min, denatured, and then incubated at 37°C for 20 min. The recovered probe hybridization mixture with a volume of 32 μL, 6.8 μL of 20 × saline sodium citrate (SSC), and 1.2 μL of 10% sodium dodecyl sulfate (SDS) was placed onto microarray slides that were prewarmed. Coverslips were applied and slides were placed into a sealed moisture chamber at 65°C for 16 h to hybridize. Slides were then immediately washed with 1 × SSC in 0.03% SDS, with 0.5% SSC twice, and with 0.06% SSC twice. After the washes, slides were centrifuged at 84 g for 2 min and immediately scanned. An Axon microarray scanner (Molecular Devices, Sunnyvale, CA) was used for scanning slides.

Microarray data analysis

GenePix Pro 4.0 software (Molecular Devices) was used to analyze scanned slides. Densitometry data for gene identification and analysis were uploaded into the Stanford Microarray Database. The log (base 2) of red/green normalized ratio (mean) was found and filtered based on a regression correlation of 0.6. Each gene was centered to the median and was only included in the final analysis if genes passed >80% good data filter criterion. Pearson correlation was used to cluster genes. Subsequently, genes between E14 and E18 with significant differences were selected using significance analysis of microarrays (SAM). By utilizing a set of gene-specific t tests, SAM is able to identify gene expression changes that are statistically significant. The analysis assigns a score to each individual gene dependent on the expression change based on the standard deviation of the gene repeated measurements. Permutations of repeated measurements are used to determine the false discovery rate (FDR) for genes equivalent to chance. Only genes that had both an FDR less than two and at least twofold expression difference were selected.

Functional analysis of differentially expressed genes

As previously described by Jovov et al.,10 network and pathway analyses of probes were performed using Ingenuity Pathway Analysis (IPA; www.ingenuity.com, Ingenuity Systems, Redwood City, CA) between significantly regulated genes to identify functional connections. The significance of networks was calculated by IPA's integrated Ingenuity algorithm, which calculates p-values using the right-tailed Fisher's exact test for each canonical pathway. The association between a subset of genes from the whole experimental data set and a related function/pathway is evaluated to be due to random association.

Results

Differential gene expression between scarless E14 and scarring E18 fibroblasts

SAM identified a total of 275 genes that were differentially expressed, with at least a twofold difference, in fetal fibroblasts between E14 and E18. Of these genes, 30 were significantly downregulated (Table 1) and 245 genes were significantly upregulated at the E18 compared with the E14 time point (Table 2).

Table 1.

Genes downregulated in E14 fibroblasts

Symbol Gene Name
Arf4 ADP-ribosylation factor 4
Mfge8 Milk fat globule-EGF factor 8 protein
Tmem147 Transmembrane protein 147
Mtg1 Mitochondrial GTPase 1 homologue (Saccharomyces cerevisiae)
Galnt10 UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 10
Crmp1 Collapsin response mediator protein 1
Rpl27 Ribosomal protein L27
Timp3 Tissue inhibitor of metalloproteinase 3
1500003O03Rik RIKEN cDNA 1500003O03 gene
Dpm1 Dolichol-phosphate (beta-D) mannosyltransferase 1
Csnk1a1 Casein kinase 1, alpha 1
Pgk1 Phosphoglycerate kinase 1
Pdcd6 Programmed cell death 6
Hdgf Hepatoma-derived growth factor
Lama2 Laminin, alpha 2
Rab10 RAB10, member RAS oncogene family
Tmem206 Transmembrane protein 206
Ubqln1 Ubiquilin 1
Myadm Myeloid-associated differentiation marker
Mat2a Methionine adenosyltransferase II, alpha
Parn poly(A)-specific ribonuclease (deadenylation nuclease)
Rae1 RAE1 RNA export 1 homologue (Schizosaccharomyces pombe)
Seh1l SEH1-like (S. cerevisiae)
Rpl3 Ribosomal protein L3
Sub1 SUB1 homologue (S. cerevisiae)
Nrbp2 Nuclear receptor binding protein 2
Ppp3ca Protein phosphatase 3, catalytic subunit, alpha isoform
Mtmr6 Myotubularin-related protein 6
Odc1 Ornithine decarboxylase, structural 1
Ttc12 Tetratricopeptide repeat domain 12

Table 2.

Genes upregulated in E14 fibroblasts

Symbol Gene Name Symbol Gene Name
Lpar4 Lysophosphatidic acid receptor 4 Pex19 Peroxisomal biogenesis factor 19
Ankrd17 Ankyrin repeat domain 17 Pikfyve Phosphoinositide kinase, FYVE finger containing
C330019G07Rik RIKEN cDNA C330019G07 gene Plac9 Placenta specific 9
Cstf2t Cleavage stimulation factor, 3′ pre-RNA subunit 2, tau Dstyk Dual serine/threonine and tyrosine protein kinase
Golt1b Golgi transport 1 homologue B (S. cerevisiae) Fcgbp Fc fragment of IgG binding protein
Adprh ADP-ribosylarginine hydrolase Pdgfa Platelet-derived growth factor, alpha
Ifrg15 Interferon alpha responsive gene Txndc16 Thioredoxin domain containing 16
4632428 N05Rik RIKEN cDNA 4632428 N05 gene Gatm Glycine amidinotransferase (L-arginine:glycine amidinotransferase)
Slc35a1 Solute carrier family 35 (CMP-sialic acid transporter), member 1 Foxn2 Forkhead box N2
Gsk3b Glycogen synthase kinase 3 beta Mrpl49 Mitochondrial ribosomal protein L49
Mepce Methylphosphate capping enzyme Rbm34 RNA binding motif protein 34
Ficd FIC domain containing Bfar Bifunctional apoptosis regulator
Ptges Prostaglandin E synthase Mdh2 Malate dehydrogenase 2, NAD (mitochondrial)
BC016495 cDNA sequence BC016495 Wrnip1 Werner helicase interacting protein 1
Myo10 Myosin X Thap7 THAP domain containing 7
Pbk PDZ binding kinase Map1lc3b Microtubule-associated protein 1 light chain 3 beta
Tex19.1 Testis expressed gene 19.1 Hoxb2 Homeobox B2
Mocs1 Molybdenum cofactor synthesis 1 Mphosph10 M-phase phosphoprotein 10 (U3 small nucleolar ribonucleoprotein)
Snap25 Synaptosomal-associated protein 25 Cyba Cytochrome b-245, alpha polypeptide
Adam10 A disintegrin and metallopeptidase domain 10 Slc9a6 Solute carrier family 9 (sodium/hydrogen exchanger), member 6
Idh3a Isocitrate dehydrogenase 3 (NAD+) alpha Pepd Peptidase D
Phtf2 Putative homeodomain transcription factor 2 Tmsb15l Thymosin beta 15b like
Hist1 h2ae Histone cluster 1, H2ae Gadd45 g Growth arrest and DNA-damage-inducible 45 gamma
Stmn3 Stathmin-like 3 Cct6a Chaperonin containing Tcp1, subunit 6a (zeta)
Ift74 Intraflagellar transport 74 homologue (Chlamydomonas) Smurf2 SMAD-specific E3 ubiquitin protein ligase 2
Mettl21a Methyltransferase-like 21A Nol4 Nucleolar protein 4
Il2 Interleukin 2 Ppp1r2 Protein phosphatase 1, regulatory (inhibitor) subunit 2
Ppp3cc Protein phosphatase 3, catalytic subunit, gamma isoform 4833439 L19Rik RIKEN cDNA 4833439 L19 gene
Ndufa13 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 13 Tnfrsf12a Tumor necrosis factor receptor superfamily, member 12a
Gzmb Granzyme B Spire1 Spire homologue 1 (Drosophila)
Ccr5 Chemokine (C-C motif) receptor 5 Acadsb Acyl-coenzyme A dehydrogenase, short/branched chain
Mbd1 Methyl-CpG binding domain protein 1 Stat1 Signal transducer and activator of transcription 1
Atp7a ATPase, Cu++ transporting, alpha polypeptide Notch4 Notch gene homologue 4 (Drosophila)
Psmc2 Proteasome (prosome, macropain) 26S subunit, ATPase 2 Cdc42ep5 CDC42 effector protein (Rho GTPase binding) 5
Cenph Centromere protein H Coro1a Coronin, actin binding protein 1A
Mllt3 Translocated to, 3 Map2k2 Mitogen-activated protein kinase kinase 2
Zfp868 Zinc finger protein 868 Wnt4 Wingless-related MMTV integration site 4
4930528F23Rik RIKEN cDNA 4930528F23 gene Agpat3 1-acylglycerol-3-phosphate O-acyltransferase 3
Khdc1b KH domain containing 1B Sh3d19 SH3 domain protein D19
Tm4sf4 Transmembrane 4 superfamily member 4 Wdr54 WD repeat domain 54
Mcm5 Minichromosome maintenance-deficient 5, cell division cycle 46 (S. cerevisiae) Adal Adenosine deaminase-like
Fank1 Fibronectin type 3 and ankyrin repeat domains 1 Rpl19 Ribosomal protein L19
Vegfc Vascular endothelial growth factor C Aasdhppt Aminoadipate-semialdehyde dehydrogenase-phosphopantetheinyl transferase
Parp16 Poly (ADP-ribose) polymerase family, member 16 Cdh3 Cadherin 3
Ubxn8 UBX domain protein 8 Rsph9 Radial spoke head 9 homologue (Chlamydomonas)
Dll1 Delta-like 1 (Drosophila) Hao2 Hydroxyacid oxidase 2
Eef1b2 Eukaryotic translation elongation factor 1 beta 2 Syf2 SYF2 homologue, RNA splicing factor (S. cerevisiae)
Abhd14a Abhydrolase domain containing 14A Pcp4l1 Purkinje cell protein 4-like 1
Ergic2 ERGIC and golgi 2 Appl1 Adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 1
Josd2 Josephin domain containing 2 Cdk5rap3 CDK5 regulatory subunit associated protein 3
Cyth1 Cytohesin 1 Tmem41b Transmembrane protein 41B
Atp4a ATPase, H+/K+ exchanging, gastric, alpha polypeptide G3bp2 GTPase activating protein (SH3 domain) binding protein 2
Unkl Unkempt-like (Drosophila) Slc35b2 Solute carrier family 35, member B2
Ndufaf1 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, assembly factor 1 Lmbrd1 LMBR1 domain containing 1
Fam57a Family with sequence similarity 57, member A Uckl1 Uridine-cytidine kinase 1-like 1
Prkag1 Protein kinase, AMP-activated, gamma 1 noncatalytic subunit Scnm1 Sodium channel modifier 1
Phip Pleckstrin homology domain interacting protein Hspbp1 HSPA (heat shock 70 kDa) binding protein, cytoplasmic cochaperone 1
Akap13 A kinase (PRKA) anchor protein 13 1810009J06Rik RIKEN cDNA 1810009J06 gene
Odz3 Odd Oz/ten-m homologue 3 (Drosophila) Ilk Integrin linked kinase
Lrat Lecithin-retinol acyltransferase (phosphatidylcholine-retinol-O-acyltransferase) Cyp26a1 cytochrome P450, family 26, subfamily a, polypeptide 1
Fam20c Family with sequence similarity 20, member C Fdx1l Ferredoxin 1-like
Zfp821 Zinc finger protein 821 Ndufa11 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 11
8030474K03Rik RIKEN cDNA 8030474K03 gene Cct8l1 Chaperonin containing TCP1, subunit 8 (theta)-like 1
Dock7 Dedicator of cytokinesis 7 Rbbp6 Retinoblastoma binding protein 6
Rab3a RAB3A, member RAS oncogene family Nppb Natriuretic peptide type B
5430437P03Rik RIKEN cDNA 5430437P03 gene Ppat Phosphoribosyl pyrophosphate amidotransferase
Lysmd4 LysM, putative peptidoglycan-binding, domain containing 4 Lrch2 Leucine-rich repeats and calponin homology (CH) domain containing 2
Zcchc3 Zinc finger, CCHC domain containing 3 Paqr5 Progestin and adipoQ receptor family member V
Ucp1 Uncoupling protein 1 (mitochondrial, proton carrier) Csnk2b Casein kinase 2, beta polypeptide
Ckap2l Cytoskeleton-associated protein 2-like Egf Epidermal growth factor
Atp7a ATPase, Cu++ transporting, alpha polypeptide Acsl6 Acyl-CoA synthetase long-chain family member 6
BC016423 cDNA sequence BC016423 Gosr2 Golgi SNAP receptor complex member 2
Tipin Timeless interacting protein Myo16 Myosin XVI
Cnot1 CCR4-NOT transcription complex, subunit 1 Sult1d1 Sulfotransferase family 1D, member 1
AI314180 Expressed sequence AI314180 Setd3 SET domain containing 3
Hspb7 Heat shock protein family, member 7 (cardiovascular) Slc30a4 Solute carrier family 30 (zinc transporter), member 4
Prpsap1 Phosphoribosyl pyrophosphate synthetase-associated protein 1 Plekhf2 Pleckstrin homology domain containing, family F (with FYVE domain) member 2
Sft2d1 SFT2 domain containing 1 Tgif2 TGFB-induced factor homeobox 2
Crym Crystallin, mu Dynll2 Dynein light chain LC8-type 2
Edn2 Endothelin 2 Ccs Copper chaperone for superoxide dismutase
Senp6 SUMO/sentrin-specific peptidase 6 Car10 Carbonic anhydrase 10
Mbd4 Methyl-CpG binding domain protein 4 5-Sep Septin 6
Rpl3 Ribosomal protein L3 Mycbp2 MYC binding protein 2
1700001J03Rik RIKEN cDNA 1700001J03 gene Vps41 Vacuolar protein sorting 41 (yeast)
Angptl1 Angiopoietin-like 1 Lefty1 Left right determination factor 1
Camsap1 Calmodulin-regulated spectrin-associated protein 1 Dcbld1 Discoidin, CUB and LCCL domain containing 1
Dhx16 DEAH (Asp-Glu-Ala-His) box polypeptide 16 Mpp1 Membrane protein, palmitoylated
1190007F08Rik RIKEN cDNA 1190007F08 gene Kcnab3 Potassium voltage-gated channel, shaker-related subfamily, beta member 3
Cdkn2b Cyclin-dependent kinase inhibitor 2B (p15, inhibits CDK4) Stoml2 Stomatin (Epb7.2)-like 2
Fam195b Family with sequence similarity 195, member B Steap3 STEAP family member 3
Muc2 Mucin 2 Tspan31 Tetraspanin 31
Stat6 Signal transducer and activator of transcription 6 Ankrd1 Ankyrin repeat domain 1 (cardiac muscle)
Med19 Mediator of RNA polymerase II transcription, subunit 19 homologue (yeast) Ormdl1 ORM1-like 1 (S. cerevisiae)
Gas5 Growth arrest-specific 5 1110008F13Rik RIKEN cDNA 1110008F13 gene
Tbl2 Transducin (beta)-like 2 Oma1 OMA1 homologue, zinc metallopeptidase (S. cerevisiae)
Ehd3 EH-domain containing 3 Cda Cytidine deaminase
Cntln Centlein, centrosomal protein Slc6a1 Solute carrier family 6 (neurotransmitter transporter, GABA), member 1
Myh8 Myosin, heavy polypeptide 8, skeletal muscle, perinatal Ccdc17 Coiled-coil domain containing 17
Srp14 Signal recognition particle 14 Lsm6 LSM6 homologue, U6 small nuclear RNA associated (S. cerevisiae)
Asnsd1 Asparagine synthetase domain containing 1 D2Ertd750e DNA segment, Chr 2, ERATO Doi 750, expressed
Zfp692 Zinc finger protein 692 Tgm1 Transglutaminase 1, K polypeptide
Fam187b Family with sequence similarity 187, member B Galntl5 UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase-like 5
Efna2 Ephrin A2 Mobkl2a MOB1, Mps one binder kinase activator-like 2A (yeast)
Prpf40a PRP40 pre-mRNA processing factor 40 homologue A (yeast) Il10ra Interleukin 10 receptor, alpha
Zyx Zyxin Pja1 Praja1, RING-H2 motif containing
Calb1 Calbindin 1 Cd83 CD83 antigen
Asb1 Ankyrin repeat and SOCS box-containing 1 Stra13 Stimulated by retinoic acid 13
Zfp105 Zinc finger protein 105 1700028P14Rik RIKEN cDNA 1700028P14 gene
Trmt2a TRM2 tRNA methyltransferase 2 homologue A (S. cerevisiae) 1700012B15Rik RIKEN cDNA 1700012B15 gene
Celf4 CUGBP, Elav-like family member 4 Chid1 Chitinase domain containing 1
1110034A24Rik RIKEN cDNA 1110034A24 gene Chgb Chromogranin B
Acbd5 Acyl-Coenzyme A binding domain containing 5 Kctd14 Potassium channel tetramerization domain containing 14
Gtf3c6 General transcription factor IIIC, polypeptide 6, alpha Rab11fip1 RAB11 family interacting protein 1 (class I)
Myc Myelocytomatosis oncogene Aldh3b1 Aldehyde dehydrogenase 3 family, member B1
Rrp12 Ribosomal RNA processing 12 homologue (S. cerevisiae) Tpd52l1 Tumor protein D52-like 1
Mapk10 Mitogen-activated protein kinase 10 2210012G02Rik RIKEN cDNA 2210012G02 gene
Pde6d Phosphodiesterase 6D, cGMP-specific, rod, delta Tspan3 Tetraspanin 3
Mtus2 Microtubule-associated tumor suppressor candidate 2 Rbm39 RNA binding motif protein 39
Pon1 Paraoxonase 1 Bin1 Bridging integrator 1
Sfxn1 Sideroflexin 1 Smc3 Structural maintenance of chromosomes 3
Pak2 p21 protein (Cdc42/Rac)-activated kinase 2 Dnttip1 Deoxynucleotidyltransferase, terminal, interacting protein 1
Etv6 ETS variant gene 6 (TEL oncogene) Dcaf4 DDB1- and CUL4-associated factor 4

Functional pathway analysis

Of the 245 genes upregulated in E18 compared with E14 fetal fibroblasts identified using microarray analysis (Fig. 1A), IPA identified 20 functional pathways represented by these genes (Fig. 1B). The top five pathways identified were associated with colorectal cancer (CRC) metastasis signaling, hepatic fibrosis/hepatic stellate cell activation, pancreatic adenocarcinoma signaling, platelet-derived growth factor (PDGF) signaling, and endometrial cancer signaling. From the 30 genes downregulated in E18 compared with E14 fibroblasts, IPA identified 20 functional pathways represented by these genes (Fig. 1C). The five most significant functional pathways identified were putrescine biosynthesis III, s-adenosyl-l-methionine biosynthesis, d-myo-inositol (1,4,5,6)-tetrakisphosphate biosynthesis, d-myo-inositol (3,4,5,6)-tetrakisphosphate biosynthesis, and dolichyl-diphosphooligosaccharide biosynthesis.

Figure 1.

Figure 1.

Microarray analysis of E14 and E18 fibroblasts. (A) Hierarchical clustering of upregulated and downregulated genes from fetal fibroblasts at E14 versus E18. Individual genes cluster according to the dendrogram on the left and are visually represented in the heat map on the right; blue indicates downregulation and yellow indicates upregulation. (B) Canonical pathways identified via IPA that were significantly enriched for among genes observed to be substantially upregulated. (C) Canonical pathways identified via IPA that were significantly enriched for among genes observed to be substantially downregulated. E14, embryonic day 14; E18, embryonic day 18; IPA, Ingenuity Pathway Analysis.

Discussion

Previous work conducted by our laboratory analyzed the differences in the transcriptomes of fetal keratinocytes and fibroblasts between E16 and E18. At the time of this research, however, it was difficult to efficiently culture E14 fetal fibroblasts.11 Using a larger number of E14 fetal mice and a smaller culture plating surface in this study, we were able to successfully culture E14 fetal fibroblasts. The microarray analysis and signal pathway analysis conducted at the E14 time point in this study revealed a number of significant gene expression changes that occur during the transition period from scarless healing to healing with scarring. A recent investigation by our laboratory compared gene expression in fetal (E17) and adult mouse wounds and identified 178 genes upregulated and 13 genes downregulated in fetal compared with adult wounds. A selection of down- and upregulated pathways was identified (unpublished observations). Together with the findings presented here, this research contributes to unraveling the mechanism of fetal regenerative cutaneous wound repair. In the following sections, we characterize in greater detail some of the notable pathways differentially activated in E14 fibroblasts versus E18 fibroblasts.

CRC metastasis signaling

SAM revealed a significant increase in CRC metastasis signaling in E18 compared with E14 fetal fibroblasts. CRC tumorigenesis is well characterized and results from the accumulation of genetic mutations. Inflammation is a known environmental factor strongly associated with genetic mutations in cells of the colonic mucosa, which predisposes it to developing CRC. The association between CRC tumorigenesis and inflammation is demonstrated by the increased risk of CRC in patients with extensive and long-term colitis.12 A number of proinflammatory factors are activated by the network of CRC signaling,13 including TNFα, interleukin (IL)-8, IL-6, and VEGF, which are all found to be elevated in the serum of patients with CRC.14 Strong evidence identifies transforming growth factor-β (TGFβ) as one of the key factors promoting inflammation during tumorigenesis.15 Interestingly, inflammation and TGFβ have also been demonstrated to play an immediate role in the fibrotic response to cutaneous injury, and TGFβ expression is significantly elevated in the fibroblasts present in keloid and hypertrophic scars.16 It is highly likely, therefore, that the observed upregulation of CRC metastasis signaling in fetal fibroblasts at E18 compared with E14 is associated with increased levels of proinflammatory factors, including TGFβ, which ultimately contribute to scar formation. Consequently, targeted inhibition of one or more proinflammatory factors upregulated in scar-forming fetal fibroblasts could significantly improve the wound repair process in adult cutaneous wounds.

Hepatic fibrosis/hepatic stellate cell activation

A significant upregulation of hepatic fibrosis/hepatic stellate cell activation pathway in fetal fibroblasts at E18 compared with E14 was found. This pathway involves activation of the hepatic stellate cell following liver injury, the principal effector of hepatic fibrogenesis, which becomes highly proliferative and synthesizes a fibrotic matrix that is rich in type I collagen.17 Hepatic fibrosis can arise secondary to a number of factors causing liver injury, including viral hepatitis or alcohol abuse. Regardless of the precise etiology, hepatic fibrosis is ultimately characterized by an increase in ECM deposition and formation of a hepatic scar. Immediately following liver injury, there is a marked transition of hepatic stellate cells from a quiescent to a robust, activated state. This response is elicited by neighboring Kupffer, endothelial, and injured hepatocyte cells, which release potent reactive oxygen intermediates (ROI) that exert paracrine stimulation of stellate cells.18 Excessive accumulation of reactive oxygen species (ROS) in wounds significantly impairs wound healing and causes substantial tissue damage.19 Our findings here suggest a differential stellate cell activation pathway between the E14 and E18 time points in fetal fibroblasts, and this could be partially caused by increased ROS in the wound bed at E18. Accordingly, methods that eliminate accumulated ROS in wound beds may enhance the regenerative capability of skin.

In addition to activating stellate cells, endothelial cells also activate plasmin. Plasmin stimulates the conversion of latent TGFβ1 to an active fibrogenic form.20 TGFβ, as mentioned, plays a major role in the fibrotic response to injury and the formation of scars. Therefore, targeted inhibition of the conversion of TGFβ1 into its more active fibrogenic form will likely enhance the regenerative properties of skin once beyond the E14 time point.

PDGF signaling

Our microarray analysis demonstrated significantly increased expression of PDGF in E18 compared with E14 fetal fibroblasts. Higher levels of PDGF have been documented both in adult wounds compared with embryonic wounds, and in uninjured adult skin compared with uninjured embryonic skin.21,22 No previous work has specifically compared the differential activation of PDGF signaling between early scar-free fetal skin and later scar-forming fetal skin. The functional pathway analysis here suggests that increased PDGF signaling contributes to the loss of the regenerative healing capability between the E14 and E18 time points. This finding is further supported by the role of PDGF in mediating proliferation and differentiation of fibroblasts. PDGF expression also leads to subsequent upregulation of TGFβ1 receptors, which have a major role in the fibrotic response to tissue injury.

Putrescine biosynthesis III

Microarray analyses showed greater downregulation of putrescine biosynthesis in E18 compared with E14 fetal fibroblasts. Putrescine has been identified to play a significant role in cell proliferation in response to injury, and is required for the completion of DNA synthesis.23 Putrescine is also suggested to be an integral precursor for the development of complex polyamines, which are major regulatory factors of mammalian tissues and influence both growth and signal transduction.24 The formation of new tissue, through cellular proliferation and migration of cells to the wound site, is a key part of wound healing. Downregulation of the putrescine biosynthesis III pathway at E18 may impair signaling, impact the regulation of cellular proliferation, and ultimately increase the amount of new tissue formed, leading to the formation of a scar. Further exploration of the putrescine biosynthesis III pathway could reveal a target for drug therapy that could potentially rescue regenerative capability at scar-forming time points.

D-myo-inositol (3,4,5,6)-tetrakisphosphate biosynthesis

Microarray analysis revealed that d-myo-inositol (3,4,5,6)-tetrakisphosphate biosynthesis was downregulated in E18 compared with E14 fetal fibroblasts. D-myo-inositol (3,4,5,6)-tetrakisphosphate is part of the inositol phosphate family, which are intracellular signaling molecules regulating the crucial functions of cell growth, apoptosis, and cell differentiation. Previous work has tentatively identified the target of d-myo-inositol (3,4,5,6)-tetrakisphosphate as a plasma membrane Ca2+-activated chloride channel.25 Interestingly, blockage of chloride channels severely reduces the rate of wound healing.26 Downregulation of chloride channel function at E18 may reduce healing capability in fetal wounds and prolong the formation of new tissue and scars. D-myo-inositol (3,4,5,6)-tetrakisphosphate is also involved in mediating the ability of remodeling complexes to induce transcription in phosphate-responsive genes.27 This finding suggests that reduction in d-myo-inositol (3,4,5,6)-tetrakisphosphate activity may contribute to epigenetic silencing and consequently the loss of regenerative healing capability observed at E18. Further work with d-myo-inositol (3,4,5,6)-tetrakisphosphate biosynthesis could reveal a novel target to induce regenerative healing mechanisms.

Innovation

Previous work has identified the key time point during fetal development when the ability to heal without scar is lost. However, the mechanism driving the transition from scarless, regenerative fetal wound healing to scar-forming adult wound repair is still largely unknown. Our work significantly narrows the search to a set of key genes and pathways that drive the scar-free healing mechanism. Further detailed examination of these genes and signaling pathways will give us a better understanding of the mechanism behind scarless wound healing and potentially identify targets for future therapies.

Key Findings.

  •  Thirty genes were significantly downregulated in E18 fetal fibroblasts compared with E14 fetal fibroblasts.

  •  Enrichment analysis revealed over 20 pathways were downregulated in E18 fetal fibroblasts compared with E14 fetal fibroblasts, including putrescine biosynthesis III, s-adenosyl-l-methionine biosynthesis, d-myo-inositol (1,4,5,6)-tetrakisphosphate biosynthesis, d-myo-inositol (3,4,5,6)-tetrakisphosphate biosynthesis, and dolichyl-diphosphooligosaccharide biosynthesis.

  •  Two hundred forty-five genes were significantly upregulated in E18 fetal fibroblasts compared with E14 fetal fibroblasts.

  •  Enrichment analysis revealed over 20 pathways upregulated in E18 fetal fibroblasts compared with E14 fetal fibroblasts, including CRC metastasis signaling, hepatic fibrosis/hepatic stellate cell activation, pancreatic adenocarcinoma signaling, PDGF signaling, and endometrial cancer signaling.

Abbreviations and Acronyms

aRNA

antisense RNA

cDNA

complementary DNA

CRC

colorectal cancer

dUTP

deoxyribose uridine triphosphate

E14

embryonic day 14

E18

embryonic day 18

ECM

extracellular matrix

EDTA

ethylenediaminetetraacetic acid

FDR

false discovery rate

IL

interleukin

IPA

Ingenuity Pathway Analysis

NIH

National Institutes of Health

PDGF

platelet-derived growth factor

ROI

reactive oxygen intermediates

ROS

reactive oxygen species

SAM

significance analysis of microarrays

SDS

sodium dodecyl sulfate

SSC

saline sodium citrate

TE

Tris-EDTA

TGFβ

transforming growth factor-β

Acknowledgments and Funding Sources

This work was supported, in part, by an NIH grant R01 GM087609 (to H.P.L.), a gift from Ingrid Lai and Bill Shu in honor of Anthony Shu (to H.P.L.), and the Hagey Laboratory for Pediatric Regenerative Medicine and Children's Surgical Research Program (to M.T.L. and H.P.L.). Additional funding was provided by the American Society of Maxillofacial Surgeons (ASMS)/Maxillofacial Surgeons Foundation (MSF) Research Grant Award (to M.S.H., M.T.L., and H.P.L.), the Sarnoff Cardiovascular Research Foundation (to W.X.H.), the California Institute for Regenerative Medicine (CIRM) Clinical Fellow training grant TG2-01159 (to M.S.H.), and the Stanford University School of Medicine Transplant and Tissue Engineering Fellowship Award (to M.S.H.).

Author Disclosure and Ghostwriting

No competing financial interests exist. The content of this article was expressly written by the authors listed. No ghostwriters were used to write this article.

About the Authors

Michael S. Hu, MD, MPH, MS, is a postdoctoral fellow at Stanford interested in stem cell biology and regenerative medicine. He is pursuing a career in craniofacial plastic surgery. Mimi R. Borrelli, MBBS, MSs, BSc, is a postdoctoral scholar at Stanford. Michael Januszyk, MD, PhD, is a resident in plastic surgery at UCLA. Anna Luan, MD, MS, is a resident in plastic surgery at Stanford. Samir Malhotra, BS, is a premedical undergraduate student at Stanford. Graham G. Walmsley, MD, PhD, is a stem cell scientist working in venture capital. Wan Xing Hong, MD, is a resident in general surgery at Stanford. Ruth Terlin, MB, BCh, BAO, MRCSI, MD, is a resident in plastic surgery at Stanford. Geoffrey C. Gurtner, MD, is a Professor of Surgery and Vice Chairman for Research in the Department of Surgery at Stanford. Michael T. Longaker, MD, MBA, is a Professor of Surgery and Bioengineering at Stanford. He is the Director of Research for the Program in Regenerative Medicine, Children's Surgical Research, and Division of Plastic and Reconstructive Surgery. His extensive research experience includes wound healing, tissue engineering, and developmental/stem cell biology. H. Peter Lorenz, MD, is a Professor and Chief of Plastic Surgery at the Lucile Packard Children's Hospital at Stanford. His clinical interests are in craniofacial surgery, pediatric plastic surgery, and reconstructive and cosmetic surgery. His laboratory group is studying mechanisms underlying scarless skin healing and the function of progenitor cells during wound repair/regeneration.

References

  • 1.Larson BJ, Longaker MT, Lorenz HP. Scarless fetal wound healing: a basic science review. Plast Reconstr Surg 2010;126:1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rowlatt U. Intrauterine wound healing in a 20 week human fetus. Virchows Arch 1979;381:353–361 [DOI] [PubMed] [Google Scholar]
  • 3.Adzick NS, Longaker MT. Scarless fetal healing. Therapeutic implications. Ann Surg 1992;215:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cass DL, Bullard KM, Sylvester KG, Yang EY, Longaker MT, Adzick NS. Wound size and gestational age modulate scar formation in fetal wound repair. J Pediatr Surg 1997;32:411–415 [DOI] [PubMed] [Google Scholar]
  • 5.Colwell AS, Krummel TM, Longaker MT, Lorenz HP. An in vivo mouse excisional wound model of scarless healing. Plast Reconstr Surg 2006;117:2292–2296 [DOI] [PubMed] [Google Scholar]
  • 6.Lo DD, Zimmermann AS, Nauta A, Longaker MT, Lorenz HP. Scarless fetal skin wound healing update. Birth Defects Res C Embryo Today 2012;96:237–247 [DOI] [PubMed] [Google Scholar]
  • 7.Brown B, McKenna S, Siddhi K, McGrouther D, Bayat A. The hidden cost of skin scars: quality of life after skin scarring. J Plast Reconstr Aesthet Surg 2008;61:1049–1058 [DOI] [PubMed] [Google Scholar]
  • 8.Alster TS, Tanzi EL. Hypertrophic scars and keloids. Am J Clin Dermatol 2003;4:235–243 [DOI] [PubMed] [Google Scholar]
  • 9.Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 2008;453:314–321 [DOI] [PubMed] [Google Scholar]
  • 10.Jovov B, Araujo-Perez F, Sigel CS, et al. Differential gene expression between African American and European American colorectal cancer patients. PLoS One 2012;7:e30168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hu MS, Januszyk M, Hong WX, et al. Gene expression in fetal murine keratinocytes and fibroblasts. J Surg Res 2014;190:344–357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 2004;287:G7–G17 [DOI] [PubMed] [Google Scholar]
  • 13.Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420:860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Klampfer L. Cytokines, inflammation and colon cancer. Curr Cancer Drug Targets 2011;11:451–464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bierie B, Moses HL. Transforming growth factor beta (TGF-β) and inflammation in cancer. Cytokine Growth Factor Rev 2010;21:49–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Leask A, Abraham DJ. TGF-β signaling and the fibrotic response. FASEB J 2004;18:816–827 [DOI] [PubMed] [Google Scholar]
  • 17.Reeves HL, Friedman SL. Activation of hepatic stellate cells—a key issue in liver fibrosis. Front Biosci 2002;7:d808–d826 [DOI] [PubMed] [Google Scholar]
  • 18.Friedman SL. The cellular basis of hepatic fibrosis—mechanisms and treatment strategies. N Engl J Med 1993;328:1828–1835 [DOI] [PubMed] [Google Scholar]
  • 19.Rasik AM, Shukla A. Antioxidant status in delayed healing type of wounds. Int J Exp Pathol 2000;81:257–263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Friedman SL. Cytokines and fibrogenesis. In: Seminars in liver disease. Semin Liver Dis 1999;2:129–140 [DOI] [PubMed] [Google Scholar]
  • 21.Ferguson MW, O'Kane S. Scar-free healing: from embryonic mechanisms to adult therapeutic intervention. Philos Trans R Soc Lond B Biol Sci 2004;359:839–850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Song H, Chai J, Lin Z, et al. A comparative study of PDGF and EGF expression in skin wound healing between human fetal and adult. Zhonghua Zheng Xing Wai Ke Za Zhi 2003;19:199–202 [PubMed] [Google Scholar]
  • 23.Sunkara PS, Rao PN, Nishioka K. Putrescine biosynthesis in mammalial cells: essential for DNA synthesis but not for mitosis. Biochem Biophys Res Commun 1977;74:1125–1133 [DOI] [PubMed] [Google Scholar]
  • 24.Coleman CS, Guirong H. Putrescine biosynthesis in mammalian tissues. Biochem J 2004;379:849–855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ismailov II, Fuller CM, Berdiev BK, Shlyonsky VG, Benos DJ, Barrett KE. A biologic function for an “orphan” messenger: D-myo-inositol 3, 4, 5, 6-tetrakisphosphate selectively blocks epithelial calcium-activated chloride channels. Proc Natl Acad Sci U S A 1996;93:10505–10509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fuchigami T, Matsuzaki T, Ihara S. Possible roles of ENaC and Cl− channel in wound closure in Xenopus laevis embryos. Zoolog Sci 2011;28:703–711 [DOI] [PubMed] [Google Scholar]
  • 27.Steger DJ, Haswell ES, Miller AL, Wente SR, O'shea EK. Regulation of chromatin remodeling by inositol polyphosphates. Science 2003;299:114–116 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Advances in Wound Care are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES