Collagen peptides from soft‑shelled turtle induce calpain‑1 expression and regulate inflammatory cytokine expression in HaCaT human skin keratinocytes
- Authors:
- Published online on: May 8, 2018 https://doi.org/10.3892/ijmm.2018.3659
- Pages: 1168-1180
Abstract
Introduction
Collagen is a ubiquitous structural protein. There are more than 20 different types of collagen, with specific functions in each tissue (1,2). These proteins have important roles in the maintenance of the extracellular matrix environment (3-7). Certain studies have demonstrated that collagen regulates cell proliferation or apoptosis (8,9). In this decade, collagens of marine origin (e.g., fish, sponges and mollusks) have been considered a useful resource due to their high availability (10-17). These collagens have been widely used as functional foods or dietary supplements. Collagen has also been used for skin substitutes and drug delivery vehicles (18-23).
Recently, collagen peptides (CPs), derived from chemical and enzymatic collagen hydrolysis (24,25), have been increasingly used as functional materials, due to their various bioactivities and high bioavailability (26,27). Several studies have demonstrated the beneficial effects of CPs. For instance, CPs derived from fish skin were demonstrated to have several protective effects on skin photo-aging and wound healing, as they improved moisture retention and repaired endogenous collagen and elastin protein fibers (28-32). Therefore, CPs are considered a useful material for the development of cosmetics, pharmaceuticals and medical products.
Previous studies by our group reported that tissue from soft-shelled turtle, Pelodiscus sinensis, may be a useful alternative source of collagen (33). Due its ability to induce keratinocytes to enter the epithelial-mesenchymal transition (EMT), which facilitates wound healing, this collagen may be a useful component of pharmaceuticals and medical products (34). Furthermore, CPs from soft-shelled turtle may have beneficial effects on skin. However, due to differences in habitat environments, collagen from soft-shelled turtle may differ greatly from collagen from mammalian or marine sources, in terms of physicochemical properties, amino acid composition and physiological functions. Therefore, further research is required prior to the use of CPs derived from soft-shelled turtle tissue in commercial products.
In the present study, a shotgun liquid chromatography/mass spectrometry (LC/MS)-based global proteomic analysis of human keratinocytes treated with CPs was performed to examine the functional effects of CPs on human skin. A total of 211 differentially expressed proteins was identified in keratinocytes treated with CPs compared with untreated keratinocytes. It was investigated whether any of these proteins may be involved in the induction of inflammatory factors in human skin.
Materials and methods
Chemicals and reagents
The highest-grade chemicals and reagents available were purchased from Wako Pure Chemical Industries (Osaka, Japan). Emperor tissue, a soft tissue in the region around the shell of soft-shelled turtles (P. sinensis), was provided by Shin-uoei, Inc. (Osaka, Japan).
Collagen extraction
Collagen extraction was performed as described in a previous study (33). In brief, emperor tissue was treated with 0.1 M formic acid at a ratio of 1:10 (w/v) for 24 h for demineralization. The sample was then treated with 0.1 M NaOH at a ratio of 1:10 (w/v) for 3 days to remove non-collagenous proteins, including endogenous proteases. The NaOH solution was changed every day. Finally, the sample was incubated with 0.03 M citric acid for 24 h. After the incubation, the solution was centrifuged at 6,500 x g for 20 min at 4°C, and the supernatant was collected. This collagen solution was used in the subsequent experiments.
Tryptic digestion of collagen
The extracted collagen solution was subjected to proteolytic activation with bovine pancreatic trypsin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) in 100 mM ammonium bicarbonate buffer (pH 8.0). The collagen was incubated with trypsin at a trypsin/collagen ratio of 1:100 (w:w) at 37°C. At each indicated time-point, reaction solutions were quickly removed and heated to 100°C to terminate trypsin digestion.
Tricine-SDS-PAGE
The molecular weights of the tryptic digestion products were determined with tricine-SDS-PAGE, as described previously (35). For comparison, molecular weight markers ranging from 3.5 to 42 kDa (Wako Pure Chemical Industries) were used. The electrophoresed gel was stained with Coomassie brilliant blue at room temperature for 1 h.
Matrix-assisted laser desorption-time of flight/mass spec- trometry (MALDI-TOF/MS)
CPs were applied onto the MALDI target plate (Shimadzu, Kyoto, Japan) with 10 mg/ml of α-cyano-4-hydroxy cinnamic acid (Sigma-Aldrich; Merck KGaA) in 50% acetonitrile and 0.05% trifluoroacetic acid. The mass spectra of the CPs were determined with an AXIMA Confidence (Shimadzu) in reflector mode. Prior to acquiring the peptide mass spectrum of the sample, the system was calibrated with a ProteoMass Peptide & Protein MALDI-MS Calibration Kit (cat. no. MSCAL1-1KT; Bradykinin fragment 1-7, 757.3997; P14R, 1,533.8582; insulin oxidized B-chain, 3,494.6513; Sigma-Aldrich; Merck KGaA).
Cell culture
HaCaT immortalized human keratinocytes were purchased from CLS Cell Lines Service GmbH (Eppelheim, Germany). The cells were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in an atmosphere containing 5% CO2 at 37°C.
Cell growth assay
Cells were plated at a density of 5×103 cells/well in a 96-well plate and grown in culture medium. The medium was changed the next day, and different concentrations of CPs were added. After 72-h treatments, the cells were incubated with the WST-8 cell counting reagent (Wako Pure Chemical Industries), and the optical density of the culture solution was measured at 450 nm with an ELISA plate reader.
Protein preparation
HaCaT cells were plated in a 60-mm dish at a density of 2×105 cells per dish and grown in culture medium. The medium was changed the next day and CPs were added. After 72-h treatments, the cells were solubilized in urea lysis buffer (7 M urea, 2 M thiourea, 5% CHAPS, 1% Triton X-100). The protein concentration was measured with the Bio-Rad Protein Assay (cat. no. 5000006JA; Bio-Rad Laboratories, Hercules, CA, USA).
In-solution trypsin digestion
The gel-free digestion method was applied as described previously (36). In brief, 10 μg protein extract from each sample was chemically reduced by adding 45 mM dithiothreitol and 20 mM tris(2-carboxyethyl)phos-phine. Subsequently, the protein was alkylated with 100 mM iodoacetamide. After the alkylation, the samples were digested with mass spectrometry grade trypsin gold (Promega Corp., Madison, WI, USA) at 37°C for 24 h. Next, the digests were purified with PepClean C-18 Spin Columns (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol.
Liquid chromatography tandem MS (LC-MS/MS) analysis for protein identification
Peptide samples (~2 μg) were injected into a peptide L-trap column (Chemicals Evaluation and Research Institute, Tokyo, Japan) with an HTC PAL autosampler (CTC Analytics, Zwingen, Switzerland). The peptides were separated further in a Paradigm MS4 (AMR Inc., Tokyo, Japan) with a reverse-phase C18-column (L-column, 3-μm-diameter gel particles and 120 Å pore size, 0.2×150 mm, Chemicals Evaluation and Research Institute). The mobile phase consisted of 0.1% formic acid in water (solution A) and acetonitrile (solution B). The column flow rate was 1 μl/min with a concentration gradient of 5% B to 40% B over 120 min. Gradient-eluted peptides were analyzed with an LTQ ion-trap mass spectrometer (Thermo Fisher Scientific, Inc.). The results were acquired in a data-dependent manner, where MS/MS fragmentation was performed on the two most intense peaks of each full MS scan.
All MS/MS spectral data were entered into a search for comparisons against the SwissProt Homo sapiens database with the Mascot tool (version 2.4.01; Matrix Science, London, UK). The search criteria were as follows: Enzyme, trypsin; with the following allowances: Up to two missed cleavage peptides; mass tolerance, ±2.0 kDa; MS/MS tolerance, ±0.8 kDa; and cysteine carbamidomethylation and methionine oxidation modifications.
Semiquantitative analysis of identified proteins
The fold-change in expression was calculated as the log2 of the ratio of protein abundances (Rsc), evaluated by spectral counting (37). For comparison, the relative amounts of identified proteins were calculated using the normalized spectral abundance factor (NSAF) (38). Differential expression of proteins were considered significant when the Rsc was >1 or <-1, which corresponded to fold-changes of >2 or <0.5, respectively.
Bioinformatics
The function of proteins that exhibited a significant change in expression with CP treatment was investigated. These sequences were processed by examining their functional annotations in the Database for Annotation, Visualization, and Integrated Discovery (DAVID) version 6.8 (http://david.abcc.ncifcrf.gov/home.jsp) (39-41).
Western blot analysis
Total protein (5 μg) that had been extracted from CP-treated cells was added to each well of an SDS-PAGE gel and electrophoresis was performed under reducing conditions. The separated proteins were transferred to polyvinylidene fluoride membranes (Merck KGaA) for 30 min at 15 V. After blocking in TBS-Tween-20 (0.1%) buffer with 5% skimmed milk for 2 h at room temperature, the membranes were incubated with an anti-calpain 1 antibody (1:1,000 dilution; cat. no. 2556; Cell Signaling Technology, Inc., Beverly, MA, USA) at 4°C overnight. The membranes were then washed and incubated with horseradish peroxidase-conjugated anti-rabbit immunoglobulin (Ig)G antibody (cat. no. A106PU; American Qualex, San Clemente, CA, USA) at room temperature for 1 h. The blots were washed and visualized with SuperSignal West Dura Extended Duration substrate (Thermo Fisher Scientific, Inc.). The bands were analyzed with the myECL Imager system (version 2.0; Thermo Fisher Scientific, Inc.). Next, the membranes were stripped by Restore Western Blot Stripping buffer (Thermo Fisher Scientific, Inc.), and the same membranes were re-probed with an anti-β-actin antibody (1:5,000 dilution; cat. no. sc-47778; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) at 4°C overnight, which served as the protein loading control. The intensities of calpain-1 and β-actin were quantified with myImageAnalysis software (version 2.0; Thermo Fisher Scientific, Inc.). The relative quantities of calpain-1 over β-actin were used to evaluate calpain-1 expression under different conditions. All western blot analyses were performed as three independent experiments.
Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)
Total RNA was extracted from HaCaT cells with the GenElute Mammalian Total RNA Miniprep kit (cat. no. RTN70-1KT; Sigma-Aldrich; Merck KGaA). Complementary (c)DNA was synthesized with the High Capacity cDNA Reverse Transcription kit (cat. no. 4368814; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. To measure the expression levels of interleukin (IL)-1α, IL-6, IL-8 and tumor necrosis factor (TNF)-α, PCR amplification was performed in the 7500 system (Applied Biosystems; Thermo Fisher Scientific, Inc.). Primers and TaqMan probes for detecting IL-1α (assay ID, Hs00174092_m1), IL-6 (assay ID, Hs00985639_m1), IL-8 (assay ID, Hs00174103_m1), TNF-α (assay ID, Hs01113624_g1) and 18S ribosomal (r)RNA (assay ID, Hs03928990_g1) were supplied with the TaqMan Gene Expression Assay (Applied Biosystems; Thermo Fisher Scientific, Inc.). The relative gene expression was calculated via the ΔΔCq method (42-46). The ΔΔCq method uses the normalized ΔCq value of each sample, which was calculated with 18S rRNA as the endogenous control gene. The ΔΔCq value is the difference between treated and control samples. Finally, the fold-change was determined as 2−ΔΔCq. Gene expression was evaluated in triplicate.
Statistical analysis
All data are presented as the mean ± standard error of the mean. The data were analyzed by one-way analysis of variance followed by Dunnett's test or the unpaired Student's t-test for two groups. P<0.05 was considered to indicate a signifi-cant difference. Computations were performed with GraphPad Prism version 5.1 (GraphPad Software Inc., La Jolla, CA, USA).
Results
Tryptic digestion of collagen from soft-shelled turtle
Collagen extracted from soft-shelled turtle was digested with trypsin to obtain CPs with molecular weights of <3.5 kDa. The collagen digestion was monitored by extracting samples at different time-points. The samples were separated on a 15% tricine-SDS-PAGE gel (Fig. 1A). After 1 h of trypsin digestion, bands that corresponded to collagen or CPs at around 42 kDa were observed (Fig. 1A; black square). After 96 h, these bands completely disappeared (Fig. 1A). The molecular weight distribution of the digested CPs was evaluated using MALDI-TOF/MS. The results indicated that the collagen was digested to CPs with a molecular weight of <4.0 kDa, and most CPs had mass-to-charge (m/z) ratios of 800-2,500 (Fig. 1B).
Cytotoxicity of CPs to HaCaT cells
To examine the possible cytotoxic effects of CPs on HaCaT cells, it was assessed whether the cell growth rate was affected when the cells were grown in culture medium containing CPs at concentrations of 0.1-100 μg/ml. The results indicated that CPs did not inhibit the HaCaT cell growth rate at any of the tested concentrations (Fig. 2). Therefore, the CPs were used at a concentration of 100 μg/ml in the subsequent experiments.
Identification and semi-quantitative comparison of differen- tially expressed proteins in CP-treated HaCaT cells
Next, the potential effect of CPs on cells in the basal layer of the skin was investigated by treating HaCaT cells with CPs. To determine the molecular profile of proteins that are regulated by CPs, a shotgun proteomics approach was used. A label-free semi-quantitative method based on spectral counting was utilized to evaluate the proteins expressed in HaCaT cells. The Rsc values were calculated for proteins that had been identified in CP-treated HaCaT cells and untreated cells. A positive value indicated increased expression by CP-treatment and a negative value indicated reduced expression by CP-treatment (Fig. 3; light grey area). For each protein that had been identified in CP-treated HaCaT cells and untreated cells, the NSAF value was also calculated (Fig. 3; black bars; peptides, grey bar; control). Proteins with a >1 and <-1 Rsc value were considered candidate CP-regulated proteins.
Based on this semi-quantitative procedure, a total of 211 proteins that were differentially expressed with CP treatment were identified (Table I). The expression of housekeeping proteins, including β-actin and GAPDH, was not altered by CP treatment.
Functional annotation of proteins regulated by CPs
A gene ontology (GO) analysis of the candidate CP-regulated proteins was then performed. GO terms associated with 'pathway' (Fig. 4A) and 'molecular function' (Fig. 4B) were searched for in DAVID. The search focused on proteins classified as 'protein processing in endoplasmic reticulum' (Table II).
Table IIDifferentially expressed proteins categorized as 'protein processing in endoplasmic reticulum' in a Gene Ontology analysis in human skin keratinocytes. |
Effect of CP on calpain-1 expression in HaCaT cells
To confirm that CP treatment altered caipain-1 expression, caipain-1 protein levels were examined in CP-treated HaCaT cells. It was identified that caipain-1 expression was signifi-cantly increased with CP treatment compared with that in control cells (Fig. 5).
Effect of CP treatment on the expression of inflammatory factors in HaCaT cells
To investigate whether the CP-induced increase in calpain-1 expression was associated with the expression of inflammatory cytokines in HaCaT cells, expression levels of IL-1α, IL-6, IL-8 and TNF-α were examined (Fig. 6). Although CP-treated HaCaT cells tended to exhibit increases in IL-1α expression (Fig. 6A), the change was not significant (P=0.0781). However, IL-6 expression was significantly decreased in HaCaT cells treated with CP, and by contrast, IL-8 expression was significantly increased with CP treatment (Fig. 6B and C). Finally, CPs had no effect on TNF-α expression (Fig. 6D).
Discussion
In the present study, a gel-free LC-MS-based proteomics approach was used to examine the functional effects of soft-shelled turtle CPs on human skin cells. A total of 211 proteins that exhibited >2-fold changes in expression after CP treatment were successfully identified in HaCaT cells, based on a semi-quantitative method of spectral counting. To examine the roles of these identified proteins, a GO analysis was performed. The study focused on the functions of proteins classified as 'protein processing in endoplasmic reticulum', as they have important roles in the synthesis of correctly folded proteins and in the degradation of misfolded proteins. The function of calpain-1, which is a member of this pathway, was also examined.
To validate the spectral counting results, a western blot analysis was performed to examine whether calpain-1 expression is increased in HaCaT cells with CP treatment. Calpain-1 is a calcium-dependent intracellular cysteine protease (47,48). It has an important role in various biological processes, including cell proliferation, cell migration, apoptosis and cytoskeletal remodeling (49,50). Therefore, calpain is considered a therapeutic target for disorders involving inflammation, wound healing and tumor progression. A previous study reported that downregulation of calpain-1 expression in IgE-activated mast cells led to a reduced expression of cytokines, including IL-6 and TNF-α. Thus, they concluded that calpain-1 may regulate IgE-mediated allergic inflammation (51). Another study indicated that downregulation of calpain-1 expression in lung fibroblast cells also reduced the expression of cytokines, including IL-6, IL-8, and TNF-α (52). Furthermore, calpain-1 knockout mice exhibited impaired bactericidal activity in an acute bacterial peritonitis model, due to a reduction in IL-1α production (53). Therefore, calpain-1 is considered a key factor in the immune response through its regulation of inflammation. Accordingly, it was hypothesized that CP-induced increases in calpain-1 expression in HaCaT cells may affect the expression of inflammatory cytokines in keratinocytes. The present results support this hypothesis. Therefore, CP treatment may regulate the immune system in the setting of skin wounds.
In the present GO analysis in the category molecular function, several proteins classified as 'cadherin binding involved in cell-cell adhesion' exhibited altered expression with CP treatment. A previous study by our group reported that in HaCaT cells, these proteins were changed by treatment with collagen derived from soft-shelled turtle. Furthermore, it was observed that changing the expression of these proteins enhanced the wound healing properties of HaCaT cells by inducing EMT (34). In the present study, it was revealed that CP treatment was associated with >2-fold increases in ceramide synthase 2 expression in HaCaT cells compared with that in control cells (Table I). Ceramide is an epidermal sphingolipid that has important roles in maintaining the skin barrier and supporting wound healing processes (54-56). The present result is similar to previous ones reported for collagen derived from soft-shelled turtles and for CPs derived from other origins (25,31,32,57). It is therefore suggested that CPs from soft-shelled turtles may facilitate wound healing.
Although the present study suggested that CPs from soft-shelled turtles may be a useful material for pharmaceuticals and medical products, it remains elusive whether these CPs affect the wound healing processes in keratinocytes in vivo. Of note, certain CP-regulated inflammatory cytokines including IL-6 and IL-8 were not been investigated to determine their potential effects on the wound healing process in keratinocytes. Further in vitro and in vivo studies are necessary to clarify the effect of CPs from soft-shelled turtle on wound healing and the role of CP-regulated inflammatory cytokines in keratinocytes. In addition, it is also necessary to investigate the correlation between the expression of calpain-1 and inflammatory cytokines in CP-treated keratinocytes.
In conclusion, the present shotgun LC/MS-based global proteomic analysis revealed that CP treatment regulated the expression of inflammatory cytokines in HaCaT cells and induced the expression of proteins associated with cell-cell adhesion and skin barrier maintenance. Therefore, CPs from soft-shelled turtles may provide significant benefits in maintaining the biological environment of the skin. These peptides may be a useful material for pharmaceuticals and medical products.
Funding
This study was supported in part by a Grant-in-Aid for Scientific Research (C) from the Japanese Society for the Promotion of Science to T.Y. (grant no. 15K09054).
Availability of data and materials
All data generated or analyzed during this study are included in this published article.
Authors' contributions
TY and AT designed the study and analyzed the data. TY and SN performed the experimental work. TY drafted the manuscript. KM contributed conduct the literature review. AT critically evaluated the study and final version of the manuscript. All authors participated in discussion of the study and gave final approval.
Ethical approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Abbreviations:
MALDI-TOF/MS |
matrix-assisted laser desorptiontime of flight/mass spectrometry |
LC-MS/MS |
liquid chromatography tandem mass spectrometry |
NSAF |
normalized spectral abundance factor |
RT-PCR |
reverse transcription-polymerase chain reaction |
IL |
interleukin |
TNF |
tumor necrosis factor |
Acknowledgments
The authors are grateful to Mr. Takashi Aboshi (Shin-uoei, Inc.) for providing the soft-shelled turtle tissue used in the present study.
References
Gelse K, Pöschl E and Aigner T: Collagens-structure, function, and biosynthesis. Adv Drug Deliv Rev. 55:1531–1546. 2003. View Article : Google Scholar : PubMed/NCBI | |
Myllyharju J and Kivirikko KI: Collagens, modifying enzymes and their mutations in humans, flies and worms. Trends Genet. 20:33–43. 2004. View Article : Google Scholar | |
Birk DE and Trelstad RL: Extracellular compartments in tendon morphogenesis: Collagen fibril, bundle, and macroaggregate formation. J Cell Biol. 103:231- 2401986. View Article : Google Scholar | |
Adachi E and Hayashi T: Anchoring of epithelia to underlying connective tissue: Evidence of frayed ends of collagen fibrils directly merging with meshwork of lamina densa. J Electron Microsc (Tokyo). 43:264–271. 1994. | |
Park KH and Bae YH: Phenotype of hepatocyte spheroids in Arg- GLY- Asp (RGD) containing a thermo- reversible extracellular matrix. Biosci Biotechnol Biochem. 66:1473- 14782002. View Article : Google Scholar | |
Liu B, Weinzimer SA, Gibson TB, Mascarenhas D and Cohen P: Type Ialpha collagen is an IGFBP- 3 binding protein. Growth Hormone IGF Res. 13:89- 972003. View Article : Google Scholar | |
Di Lullo GA, Sweeney SM, Korkko J, Ala-Kokko L and San Antonio JD: Mapping the ligand-binding sites and disease-associated mutations on the most abundant protein in the human, type I collagen. J Biol Chem. 277:4223–4231. 2002. View Article : Google Scholar | |
Saby C, Buache E, Brassart-Pasco S, El Btaouri H, Courageot MP, Van Gulick L, Garnotel R, Jeannesson P and Morjani H: Type I collagen aging impairs discoidin domain receptor 2-mediated tumor cell growth suppression. Oncotarget. 7:24908–24927. 2016. View Article : Google Scholar : PubMed/NCBI | |
Maquoi E, Assent D, Detilleux J, Pequeux C, Foidart JM and Noël A: MT1-MMP protects breast carcinoma cells against type I collagen-induced apoptosis. Oncogene. 31:480–493. 2012. View Article : Google Scholar | |
Muralidharan N, Jeya Shakila R, Sukumar D and Jeyasekaran G: Skin, bone and muscle collagen extraction from the trash fish, leather jacket (Odonus niger) and their characterization. J Food Sci Technol. 50:1106–1113. 2013. View Article : Google Scholar : | |
Wang Y and Regenstein JM: Effect of EDTA, HCl, and citric acid on Ca salt removal from Asian (silver) carp scales prior to gelatin extraction. J Food Sci. 74:C426–C431. 2009. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Zhan CL, Cai QF, Du CH, Liu GM, Su WJ and Cao MJ: Expression and characterization of common carp (Cyprinus carpio) matrix metalloproteinase-2 and its activity against type I collagen. J Biotechnol. 177:45- 522014. View Article : Google Scholar : PubMed/NCBI | |
Benjakul S, Thiansilakul Y, Visessanguan W, Roytrakul S, Kishimura H, Prodpran T and Meesane J: Extraction and characterisation of pepsin-solubilised collagens from the skin of bigeye snapper (Priacanthus tayenus and Priacanthus macracanthus). J Sci Food Agric. 90:132–138. 2010. View Article : Google Scholar : PubMed/NCBI | |
Nalinanon S, Benjakul S and Kishimura H: Collagens from the skin of arabesque greenling (Pleurogrammus azonus) solubilized with the aid of acetic acid and pepsin from albacore tuna (Thunnus alalunga) stomach. J Sci Food Agric. 90:1492–1500. 2010. View Article : Google Scholar : PubMed/NCBI | |
Tziveleka LA, Ioannou E, Tsiourvas D, Berillis P, Foufa E and Roussis V: Collagen from the marine sponges Axinella cannabina and Suberites carnosus: Isolation and morphological, biochemical, and biophysical characterization. Mar Drugs. 15:E1522017. View Article : Google Scholar : PubMed/NCBI | |
Pallela R, Venkatesan J, Janapala VR and Kim SK: Biophysicochemical evaluation of chitosan- hydroxyap-atite-marine sponge collagen composite for bone tissue engineering. J Biomed Mater Res A. 100:486- 4952012. | |
Coelho RCG, Marques ALP, Oliveira SM, Diogo GS, Pirraco RP, Moreira-Silva J, Xavier JC, Reis RL, Silva TH and Mano JF: Extraction and characterization of collagen from Antarctic and Sub-Antarctic squid and its potential application in hybrid scaffolds for tissue engineering. Mater Sci Eng C Mater Biol Appl. 78:787–795. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gorell ES, Leung TH, Khuu P and Lane AT: Purified type I collagen wound matrix improves chronic wound healing in patients with recessive dystrophic epidermolysis bullosa. Pediatr Dermat. 32:220–225. 2015. View Article : Google Scholar | |
Shevchenko RV, Sibbons PD, Sharpe JR and James SE: Use of a novel porcine collagen paste as a dermal substitute in full-thickness wounds. Wound Repair Regen. 16:198–207. 2008. View Article : Google Scholar : PubMed/NCBI | |
Wollina U, Meseg A and Weber A: Use of a collagen-elastin matrix for hard to treat soft tissue defects. Int Wound J. 8:291–296. 2011. View Article : Google Scholar : PubMed/NCBI | |
Barhoumi A, Salvador-Culla B and Kohane DS: NIR-triggered drug delivery by collagen-mediated second harmonic generation. Adv Healthc Mater. 4:1159–1163. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wallace DG and Rosenblatt J: Collagen gel systems for sustained delivery and tissue engineering. Adv Drug Deliv Rev. 55:1631–1649. 2003. View Article : Google Scholar : PubMed/NCBI | |
Friess W: Collagen-biomaterial for drug delivery. Eur J Pharm Biopharm. 45:113–136. 1998. View Article : Google Scholar : PubMed/NCBI | |
Song H, Zhang S, Zhang L and Li B: Effect of orally administered collagen peptides from bovine bone on skin aging in chronologically aged mice. Nutrients. 9:E12092017. View Article : Google Scholar : PubMed/NCBI | |
Hu Z, Yang P, Zhou C, Li S and Hong P: Marine collagen peptides from the skin of Nile Tilapia (Oreochromis niloticus): Characterization and wound healing evaluation. Mar Drugs. 15:E1022017. View Article : Google Scholar : PubMed/NCBI | |
Zhuang Y, Hou H, Zhao X, Zhang Z and Li B: Effects of collagen and collagen hydrolysate from jellyfish (Rhopilema esculentum) on mice skin photoaging induced by UV irradiation. J Food Sci. 74:H183- H1882009. View Article : Google Scholar | |
Zague V: A new view concerning the effects of collagen hydroly-sate intake on skin properties. Arch Dermatol Res. 300:479–483. 2008. View Article : Google Scholar : PubMed/NCBI | |
Fan J, Zhuang Y and Li B: Effects of collagen and collagen hydrolysate from jellyfish umbrella on histological and immunity changes of mice photoaging. Nutrients. 5:223–233. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hou H, Li B, Zhang Z, Xue C, Yu G, Wang J, Bao Y, Bu L, Sun J, Peng Z and Su S: Moisture absorption and retention properties, and activity in alleviating skin photodamage of collagen polypeptide from marine fish skin. Food Chem. 135:1432- 14392012. View Article : Google Scholar | |
Song H, Meng M, Cheng X, Li B and Wang C: The effect of collagen hydrolysates from silver carp (Hypophthalmichthys molitrix) skin on UV-induced photoaging in mice: Molecular weight affects skin repair. Food Funct. 8:1538–1546. 2017. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Wang J, Ding Y, Dai X and Li Y: Oral administration of marine collagen peptides from Chum Salmon skin enhances cutaneous wound healing and angiogenesis in rats. J Sci Food Agric. 91:2173–2179. 2011.PubMed/NCBI | |
Wang J, Xu M, Liang R, Zhao M, Zhang Z and Li Y: Oral administration of marine collagen peptides prepared from chum salmon (Oncorhynchus keta) improves wound healing following cesarean section in rats. Food Nutr Res. 59:264112015. View Article : Google Scholar : PubMed/NCBI | |
Yamamoto T, Uemura K, Sawashi Y, Mitamura K and Taga A: Optimization of method to extract collagen from 'Emperor' tissue of soft-shelled turtles. J Oleo Sci. 65:169–175. 2016. View Article : Google Scholar | |
Yamamoto T, Nakanishi S, Mitamura K and Taga A: Shotgun label- free proteomic analysis for identification of proteins in HaCaT human skin keratinocytes regulated by the administration of collagen from soft- shelled turtle. J Biomed Mater Res B Appl Biomater. 2017. View Article : Google Scholar | |
Schägger H: Tricine- SDS- PAGE. Nat Protoc. 1:16–22. 2006. View Article : Google Scholar | |
Bluemlein K and Ralser M: Monitoring protein expression in whole-cell extracts by targeted label- and standard-free LC-MS/MS. Nat Protoc. 6:859–869. 2011. View Article : Google Scholar : PubMed/NCBI | |
Old WM, Meyer-Arendt K, Aveline-Wolf L, Pierce KG, Mendoza A, Sevinsky JR, Resing KA and Ahn NG: Comparison of label-free methods for quantifying human proteins by shotgun proteomics. Mol Cell Proteomics. 4:1487–1502. 2005. View Article : Google Scholar : PubMed/NCBI | |
Zybailov B, Coleman MK, Florens L and Washburn MP: Correlation of relative abundance ratios derived from peptide ion chromatograms and spectrum counting for quantitative proteomic analysis using stable isotope labeling. Anal Chem. 77:6218–6224. 2005. View Article : Google Scholar : PubMed/NCBI | |
Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI | |
Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinfor-matics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar | |
Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar | |
Parikh P, Bai H, Swartz MF, Alfieris GM and Dean DA: Identification of differentially regulated genes in human patent ductus arteriosus. Exp Biol Med (Maywood). 241:2112- 21182016. View Article : Google Scholar | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar | |
Carbotti G, Nikpoor AR, Vacca P, Gangemi R, Giordano C, Campelli F, Ferrini S and Fabbi M: IL-27 mediates HLA class I up-regulation, which can be inhibited by the IL-6 pathway, in HLA-deficient small cell lung cancer cells. J Exp Clin Cancer Res. 36:1402017. View Article : Google Scholar : PubMed/NCBI | |
Adnan M, Morton G and Hadi S: Analysis of rpoS and bolA gene expression under various stress-induced environments in planktonic and biofilm phase using 2(−ΔΔCT) method. Mol Cell Biochem. 357:275- 2822011. View Article : Google Scholar | |
Soejima M and Koda Y: TaqMan-based real-time polymerase chain reaction for detection of FUT2 copy number variations: Identification of novel Alu-mediated deletion. Transfusion. 51:762–769. 2011. View Article : Google Scholar | |
Goll DE, Thompson VF, Li H, Wei W and Cong J: The calpain system. Physiol Rev. 83:731–801. 2003. View Article : Google Scholar : PubMed/NCBI | |
Momeni HR: Role of calpain in apoptosis. Cell J. 13:65–72. 2011.PubMed/NCBI | |
Glading A, Lauffenburger DA and Wells A: Cutting to the chase: Calpain proteases in cell motility. Trends Cell Biol. 12:46- 542002. View Article : Google Scholar : PubMed/NCBI | |
Sorimachi H, Ishiura S and Suzuki K: Structure and physiological function of calpains. Biochem J. 328:721- 7321997. View Article : Google Scholar | |
Wu Z, Chen X, Liu F, Chen W, Wu P, Wieschhaus AJ, Chishti AH, Roche PA, Chen WM and Lin TJ: Calpain-1 contributes to IgE-mediated mast cell activation. J Immunol. 192:5130–5139. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yin G, Zeng Q, Zhao H, Wu P, Cai S, Deng L and Jiang W: Effect and mechanism of calpains on pediatric lobar pneumonia. Bioengineered. 8:374- 3822017. View Article : Google Scholar : | |
Kumar V, Everingham S, Hall C, Greer PA and Craig AW: Calpains promote neutrophil recruitment and bacterial clearance in an acute bacterial peritonitis model. Eur J Immunol. 44:831–841. 2014. View Article : Google Scholar : PubMed/NCBI | |
Kim H, Kim J, Park J, Kim SH, Uchida Y, Holleran WM and Cho Y: Water extract of gromwell (Lithospermum erythrorhizon) enhances migration of human keratinocytes and dermal fibroblasts with increased lipid synthesis in an in vitro wound scratch model. Skin Pharmacol Physiol. 25:57–64. 2012. View Article : Google Scholar | |
Amen N, Mathow D, Rabionet M, Sandhoff R, Langbein L, Gretz N, Jäckel C, Gröne HJ and Jennemann R: Differentiation of epidermal keratinocytes is dependent on glucosylceramide: Ceramide processing. Hum Mol Genet. 22:4164- 41792013. View Article : Google Scholar | |
Meckfessel MH and Brandt S: The structure, function, and importance of ceramides in skin and their use as therapeutic agents in skin-care products. J Am Acad Dermatol. 71:177–184. 2014. View Article : Google Scholar : PubMed/NCBI | |
Gangwar M, Gautam MK, Ghildiyal S, Nath G and Goel RK: Mallotus philippinensis Muell. Arg fruit glandular hairs extract promotes wound healing on different wound model in rats. BMC Complement Altern Med. 15:1232015. View Article : Google Scholar : PubMed/NCBI |