You are on page 1of 12

Acta Biomaterialia 54 (2017) 333–344

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Full length article

Human lung fibroblast-derived matrix facilitates vascular


morphogenesis in 3D environment and enhances skin wound healing
Ping Du a, Muhammad Suhaeri a,b, Sang Su Ha a,b, Seung Ja Oh a,b, Sang-Heon Kim a,b, Kwideok Park a,b,⇑
a
Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
b
Dept. of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea

a r t i c l e i n f o a b s t r a c t

Article history: Extracellular matrix (ECM) is crucial to many aspects of vascular morphogenesis and maintenance of vas-
Received 22 September 2016 culature function. Currently the recapitulation of angiogenic ECM microenvironment is still challenging,
Received in revised form 23 March 2017 due mainly to its diverse components and complex organization. Here we investigate the angiogenic
Accepted 24 March 2017
potential of human lung fibroblast-derived matrix (hFDM) in creating a three-dimensional (3D) vascular
Available online 27 March 2017
construct. hFDM was obtained via decellularization of in vitro cultured human lung fibroblasts and ana-
lyzed via immunofluorescence staining and ELISA, which detect multiple ECM macromolecules and
Keywords:
angiogenic growth factors (GFs). Human umbilical vein endothelial cells (HUVECs) morphology was more
Human lung fibroblast-derived matrix
(hFDM)
elongated and better proliferative on hFDM than on gelatin-coated substrate. To prepare 3D construct,
Collagen hydrogel hFDM is collected, quantitatively analyzed, and incorporated in collagen hydrogel (Col) with HUVECs.
Human umbilical vein endothelial cells Capillary-like structure (CLS) formation at 7 day was significantly better with the groups containing
(HUVECs) Vascular morphogenesis higher doses of hFDM compared to the Col group (control). Moreover, the group (Col/hFDM/GFs) with
Skin wound healing both hFDM and angiogenic GFs (VEGF, bFGF, SDF-1) showed the synergistic activity on CLS formation
and found much larger capillary lumen diameters with time. Further analysis of hFDM via angiogenesis
antibody array kit reveals abundant biochemical cues, such as angiogenesis-related cytokines, GFs, and
proteolytic enzymes. Significantly up-regulated expression of VE-cadherin and ECM-specific integrin sub-
units was also noticed in Col/hFDM/GFs. In addition, transplantation of Col/hFMD/GFs with HUVECs in
skin wound model presents more effective re-epithelialization, many regenerated hair follicles, better
transplanted cells viability, and advanced neovascularization. We believe that current system is a very
promising platform for 3D vasculature construction in vitro and for cell delivery toward therapeutic
applications in vivo.

Statement of Significance

Functional 3D vasculature construction in vitro is still challenging due to the difficulty of recapitulating
the complex angiogenic extracellular matrix (ECM) environment. Herein, we present a simple and prac-
tical method to create an angiogenic 3D environment via incorporation of human lung fibroblast-derived
matrix (hFDM) into collagen hydrogel. We found that hFDM offers a significantly improved angiogenic
microenvironment for HUVECs on 2D substrates and in 3D construct. A synergistic effect of hFDM and
angiogenic growth factors has been well confirmed in 3D condition. The prevascularized 3D collagen con-
structs also facilitate skin wound healing. We believe that current system should be a convenient and
powerful platform in engineering 3D vasculature in vitro, and in delivering cells for therapeutic purposes
in vivo.
Ó 2017 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction

Neovascularization that includes vasculogenesis and


angiogenesis is a complex process that involves different cell types,
⇑ Corresponding author at: Center for Biomaterials, Korea Institute of Science and multiple extracellular matrix (ECM) components, and growth
Technology (KIST), Seoul 02792, Republic of Korea.
factors (GFs) [1,2]. Promotion of vascularization is not only critical
E-mail address: kpark@kist.re.kr (K. Park).

http://dx.doi.org/10.1016/j.actbio.2017.03.035
1742-7061/Ó 2017 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
334 P. Du et al. / Acta Biomaterialia 54 (2017) 333–344

for construction of viable engineered tissue constructs in vitro, but immunofluorescence and ELISA, respectively. The effect of hFDM
also important for the treatment of various pathological on HUVECs vascular morphogenesis was comprehensively investi-
conditions, i.e., wound, ulcer, myocardial infarction, and peripheral gated for up to 7 days in 3D constructs that were built incorporat-
vascular diseases [3–5]. Upon the significance of vascularization in ing HUVECs and different doses of hFDM or angiogenic GFs into
tissue engineering, many approaches have been adopted to create collagen hydrogel. Moreover, to figure out the underlying mecha-
three-dimensional (3D) vasculatures. Common strategies for vas- nism of significantly advanced HUVECs vasculogenesis, an angio-
culature construction are mostly relied on materials, GFs, and cell genic factors profiling of hFDM was carried out, along with the
types. Natural hydrogels, such as fibrin [6], collagen, and MatrigelÒ study of gene expression related to cell-ECM interactions (inte-
are very promising materials for 3D vasculature formation [7]. grins) and cell-cell contacts (VE-cadherins). In addition, therapeu-
However, these materials have a weak mechanical property and tic effect of such 3D constructs was evaluated using murine skin
are easily degradable or deformed. To overcome these shortcom- wound model. To our best knowledge, this study is the first report
ings, synthetic polymer materials were frequently modified using that clearly demonstrates the pro-angiogenic effects of hFDM in 3D
fibronectin (Fn), collagen (Col), laminin (Ln), or other ECM-like environment on HUVECs vascular morphogenesis in vitro and ther-
components to enhance cell-matrix interactions that would trigger apeutic efficacy in vivo. Current system should be a convenient and
specific signaling pathways for angiogenesis [8]. Diverse angio- an effective platform in engineering 3D vasculature of angiogenic
genic GFs, i.e., vascular endothelial growth factor (VEGF), basic model in vitro, and in delivering cells for therapeutic purposes.
fibroblast growth factor (bFGF), angiopoietin, and stromal derived
factor (SDF)-a1 have also been employed to promote vascular cell
2. Materials and method
survival, proliferation and vascular morphogenesis [9,10]. Selec-
tion of proper cell types, such as endothelial and perivascular cells,
2.1. Preparation of human fibroblasts-derived matrix (hFDM)
or human mesenchymal stem cells are other strategies to create a
fully vascularized construct [11].
Human lung fibroblasts (hFB) (WI-38; ATCCÒ-CCL-75TM, Manas-
Previously mentioned approaches of neovascularization are
sas, VA) were cultured in Dulbecco’s Modified Eagle’s Medium
appreciable but still far short of recapitulating the in vivo process
(DMEM) supplemented with 10% fetal bovine serum (FBS),
of vasculature formation. One of the critical issues is to establish
100 U/mL penicillin, and 100 mg/mL streptomycin (Invitrogen,
a natural angiogenic ECM microenvironment in 3D condition.
Carlsbad, CA). For the preparation of hFDM, hFB are seeded at a
ECM is a physical entity of the extracellular domain that is mainly
density of 2  104/cm2 onto cell culture dishes (100 mm diameter)
comprised of macromolecular proteins, polysaccharides, and other
or gelatin-coated glass coverslips, and cultivated for 7–10 days
constituents. ECM includes structural elements (Col, fibrillins, fibu-
until confluence. The culture medium was supplemented with
lins and elastin), cell adhesive proteins (Fn, Ln, vitronectin, and
50 mg/mL ascorbic acid 2-phosphate (A4403; Sigma) to enhance
tenascins), proteoglycans (chondroitin sulfate and heparin sulfate),
cell-derived ECM deposition. The medium was changed every 2
non-proteoglycans, and matrix metalloproteinase [12,13]. Bio-
or 3 days. For decellularization, the confluent cells were treated
chemical cues in ECM are also important in initiating and in regu-
briefly with a detergent solution containing 0.25% Triton X-100
lating vascular cells morphogenesis. ECM is known as a reservoir of
and 50 mM NH4OH (Sigma-Aldrich) [22]. After the samples were
diverse GFs. Among many ECM candidates, we have been investi-
washed with phosphate buffered saline (PBS), the mixture of
gating cell-derived ECM (CDM) as an analog of natural ECM
50 U/mL DNase I and 2.5 mL/mL RNase A (Invitrogen) was added
in vivo. In fact, CDM shares similar constituents with endothelial
and incubated at 37 °C for 1–2 h. The resultant decellularized
basement membrane (BM) through which vascular cells interact
ECM was named hFDM. The hFDM on the culture dishes were used
with pericytes or smooth muscle cells [14]. For this reason, CDM
as prepared or collected in corning tubes with gentle pipetting. All
have been utilized to investigate a fundamental of ECs–matrix
the hFDMs were stored at 4 °C prior to further use. Meanwhile
interactions that may lead to a neovascular or pathological path-
hFDM on gelatin-coated coverglass was gently washed with PBS
way [15]. For example, studies have demonstrated that CDMs from
and used as a control for comparative analysis on 2D condition.
human lung fibroblast [16] or from a co-culture of dermal fibrob-
lasts with breast cancer-cell line [17] supported human umbilical
vein endothelial cells (HUVECs) vascular morphogenesis in vitro. 2.2. Compositional analysis of hFDM
Other reports have suggested the promotion of ECs sprouting and
lumen formation in the presence of fibroblasts via deposition of 2.2.1. BCA assay
ECM components, and via secretion of angiogenic GFs and prote- To quantitatively determine the total protein contents on hFDM
olytic enzymes [18–20]. Our previous studies reported that mouse before and after decellularization, samples prepared in 100 mm
fibroblast-derived matrix (FDM) and preosteoblast-derived matrix diameter Petri dish were analyzed using BCA assay kit (23225;
(PDM) are an excellent 2D platform for HUVECs adhesion, prolifer- Thermo Fisher Scientific). After several rinsing with PBS, hFB sam-
ation, and vascular morphogenesis, compared to Fn- or gelatin- ples before and after decellularization were solubilized in RIPA cell
coated substrates [21]. Interestingly chondrocyte-derived matrix lysis buffer (R0278; Sigma) with EDTA-free protease inhibitors
was very poor in the formation of capillary-like structure. These cocktail (78415 and 1862495; Thermo Scientific), and then col-
studies set a good example in demonstrating the importance of lected in the 1.7 mL centrifuge tubes, respectively. They were then
providing HUVECs with appropriate ECM niche for vascular mor- suspended using pipette, rocked gently for 30 min at 2–8 °C, and
phogenesis. Vascular structure on these 2D environments is how- centrifuged at 14,000g for 5 min. The supernatants were trans-
ever stable only for a very short period (less than 3 days). And ferred into a new tube and subjected to BCA assay immediately
there are practical issues due to dimensional limitation of 2D envi- or stored at 80 °C as aliquots. Total protein amount of each sam-
ronment. Accordingly, advanced strategies combining 3D construct ple was normalized to the surface area of culture dishes and thus
and CDM are necessary to create more stable and robust vascula- the decellularized hFDM concentration was denoted as mg/cm2.
tures for a longer period time.
In this study, we hypothesize that ECM obtained from decellu- 2.2.2. Enzyme linked immunoassay (ELISA)
larized human lung fibroblasts can facilitate HUVECs vasculogene- To determine the amount of GFs in hFDM (before and after
sis in 3D environment. To test our hypothesis, we first identified decellularization), the same samples obtained from Section 2.2.1
major ECM macromolecules and angiogenic GFs within hFDM by were analyzed using VEGF ELISA kit (DVE00; R&D system) and
P. Du et al. / Acta Biomaterialia 54 (2017) 333–344 335

human FGF-basic ELISA development kit (900-K08; Peprotech), density is 2  106 cell/mL. All the procedures were carried out on
respectively. The concentration of two GFs was also normalized ice. Once mixed thoroughly, 30 mL mixture of HUVECs, Col, and dif-
to the surface area of the culture dishes and therefore each GF ferent doses of hFDM was loaded into 96-well half-well plate
amount is denoted as pg/cm2 [23]. (3696; Corning) (n = 6, each group) and incubated at 37 °C for
30 min until polymerization. After then, 80 mL EGM-2 medium
2.2.3. Angiogenesis antibody array was added into each sample. HUVECs morphology in four test
Angiogenesis related factors were screened using a sandwich groups was observed at different time points. To visualize different
immunoassay array kit (ARY007; Proteome ProfilerTM Human doses of hFDM, hFDM was incorporated into Col without HUVECs
Angiogenesis Array Kit, R&D system) following the manufacturer’s and stained by Fn. And for a vasculature formation within the con-
instructions. In brief, antibody-coated membrane was first blocked structs, samples were fixed on day 3 and 7, and subjected to F-actin
with supplied block buffer and then protein sample (300 mg hFDM) and DAPI staining.
was added, along with certain amount of detection antibody in
array buffer to make a total volume 1.5 mL. They were incubated 2.5. Incorporation of hFDM and angiogenic growth factors in 3D
overnight at 2–8 °C on a rocking platform shaker. On the next constructs
day, the membrane was first washed with 1x wash buffer 3 times
(10 min each time), and then incubated with diluted streptavidin- To investigate a synergistic effect of both hFDM and growth fac-
HRP for 30 min at room temperature on the shaker. After 3 times tors (GFs), 3D constructs containing HUVECs (2  106 cell/mL) and
rinsing for 10 min each time, membrane was reacted with 1 mL angiogenic GFs were prepared as follows: Col alone (Col), Col with
prepared Chemi Reagent mix for 4 min. Chemiluminescence was GFs (Col/GFs), Col with hFDM (Col/hFDM), and Col with both hFDM
examined via LAS 3000 imaging system (Fuji), and the intensity and GFs (Col/hFDM/GFs). The GFs are human vascular endothelial
of the positive dots was recorded using Multi gauge v3.0 software growth factor (VEGF) (cyt-260, Prospec, Israel), recombinant
(Fuji Film). Quantitative data are obtained after each dot was nor- human fibroblast growth factor basic-2 (bFGF) (cyt-218), and
malized on a scale ranging from negative control (0%) to positive recombinant human stromal cell-derived factor-1 alpha (SDF-1a)
control value (100%). (chm-262). The final concentration of all three GFs was 200 ng/
mL [25], respectively and hFDM was used at 250 mg/mL. These con-
2.3. Huvecs culture, adhesion, and proliferation on 2D substrates structs were cultivated for up to 7 days. The same protocols were
applied for the analysis as described in the Section 2.4.
Human umbilical vein endothelial cells (HUVECs) (passage 3 to
6) (C2517A; Lonza, Walkersville, MD) are cultured in endothelial 2.6. Immunofluorescence (IF) staining
cell growth medium (EGM-2 BulletKit, CC-3162) at 37 °C under
5% CO2 humidified atmosphere. For cell adhesion and proliferation, For hFB, hFDM, and HUVECs staining on 2D substrates, samples
HUVECs suspended in EGM-2 medium or endothelial cell basal on glass coverslips were fixed in 4% paraformaldehyde (pH 7.4) for
medium (EBM-2) (CC-3156; no GFs, no cytokines, no other supple- 10 min and washed by PBS three times. They were permeabilized
ments) are seeded on the gelatin-coated coverslips (control) and in 0.2% Triton-X 100 for 5 min, blocked with 3% bovine serum albu-
hFDM in 12-well plates (5  104 cells/well) (n = 4, each group), min (BSA) for 1 h, then incubated with primary antibodies over-
respectively. Cell attachment at 3 h and cell proliferation at 24 night at 4 °C, washed again with PBS, and incubated with
and 72 h on these substrates were examined in different medium secondary antibodies for 1 h at room temperature. These samples
conditions using cell counting kit-8 (CCK-8) (CK04; Dojindo, Japan) were washed three times and mounted onto microscope slides
assay. Briefly, 10% CCK-8 solution diluted in culture medium was using vectashiedÒ mounting medium with DAPI (H-1200; Vector
added to each sample and incubated at 37 °C for 2 h. Aliquots of Laboratories, INC., CA) for nucleic labeling. There is little chance
each sample (100 mL) were transferred to 96-well plate and the of non-specific binding of the secondary antibody after IF staining
absorbance was measured at 450 nm wavelength using multiskan (Fig. S1). For capillary-like structure (CLS) observation in 3D con-
microplate reader (Thermo Scientific, Rockford, IL). Cell structs, the same protocol was applied as mentioned before but
proliferation assay was performed in triplicate. In addition, more extensive washing and longer incubation with 0.2% Triton-
HUVECs morphology was visualized via CD31 and F-actin X 100 for 30 min and 3% BSA for 2 h. DAPI (46190; thermo scien-
immunofluorescence (IF) staining or examined under phase- tific) was co-stained with the samples to label nuclei. Once the
contrast microscope. Moreover, to investigate cell-matrix interac- staining procedures were completed, samples were washed thor-
tions on two different substrates, HUVECs cultured in EGM-2 or oughly and transferred to coverglass bottom dishes (200350; SPL
EBM-2 medium (n = 4, each group) for 24 h were fixed and sub- Life Sciences). Fluorescence images of CLS in the 3D constructs
jected to IF staining of integrin a5, Fn, and F-actin, respectively. were taken using a confocal microscope (LSM 700, Carl Zeiss) with
consecutive z-stack images at 400 magnifications (20–80 iterations
2.4. Construction of HUVECs 3D culture model with an interval thickness of 1 mm). To identify the luminal struc-
ture and capillary thickness, 3D reconstruction of CLS was per-
The obtained hFDM from Section 2.1 was re-suspended in 5 mL formed via Imaris software (Imaris 7.7.2). The CLS formation was
autoclaved distilled water (DW) by moderate agitation. After cen- quantitatively evaluated via the number of segments, total capil-
trifugation at 3000 rpm, the supernatant was removed and this lary length, respectively. The number of ECs per 100 lm capillary
process was repeated twice, then a mass of hFDM was collected. length was also determined using Imaris software, based on the
To test hFDM effect on HUVECs morphogenesis in 3D environment, DAPI-positive stains of capillary networks. In addition, the number
different amounts of hFDM were incorporated into collagen hydro- of ECs per capillary diameter was counted using the cross-section
gel (Col) to have final concentrations of hFDM at 0, 100, 250 and images of lumen space.
500 mg/mL, where they were designated as 0 hFDM, 100 hFDM, Primary antibodies and dilution ratios used in this study are
250 hFDM, and 500 hFDM, respectively. A final concentration of summarized as follows: mouse anti-Fn (SC-8422; Santa Cruz
Col at 2.5 mg/mL was prepared [24] using rat tail collagen type I Biotechnology, 1: 200), rabbit anti-Fn (SC-9068, 1: 200), goat
(354236; CorningÒ) diluted in 1  M199 (11150059) and anti-Ln (SC-16590, 1: 100), rabbit polyclonal anti-collagen type I
10  M199 medium (11825015; GibcoTM) with phenol red. The pH (ab34710, 1: 300; Abcam), rabbit polyclonal anti-CD31 (ab28364,
was adjusted to 7.4 using 1 N NaOH (Sigma) and the final cell 1: 100), and mouse anti-a-smooth muscle actin (A2547;
336 P. Du et al. / Acta Biomaterialia 54 (2017) 333–344

Sigma-Aldrich, 1: 100). Secondary antibodies are Alexa FluorÒ 488 every 2 or 3 days. Digital photographs were taken to record the
F(ab0 ) fragment of goat anti-mouse IgG (A-11017), Dylight 488 of wound healing process during 14 days. Wound area is quantified
goat polyclonal to rabbit IgG-H&L (ab96899), Alexa FluorÒ 594- using ImageJ software (NIH, Bethesda, MD) in which wound heal-
conjugated donkey anti-goat IgG (Jackson Immunoresearch, West ing is expressed as a ratio of the wound area divided by the initial
Grove, PA), and Alexa FluorÒ 647-conjugated goat polyclonal to wound area at specific time points.
mouse IgG-H&L (ab150115). All the secondary antibodies were
diluted in 1% BSA at 1: 500. F-actin cytoskeleton of HUVECs was 2.9. Histological and IF staining
labeled by tetramethylrhodamine isothiocyanate (TRITC)-
conjugated phalloidin (A415; Life Technologies, 1: 200). After transplantation for 14 days, samples in wound areas were
harvested, cut into halves, and fixed in 4% paraformaldehyde. Half
2.7. Real-time quantitative polymerase chain reaction (q-PCR) tissue was embedded into optimum cutting medium (OCT) for
cryosection to identify the transplanted HUVECs. The other half
Total mRNA of HUVECs cultured in the 3D constructs of Col, Col/ was embedded in paraffin wax, and sliced into 6 mm thick sections
GFs, Col/hFDM, and Col/hFDM/GFs was extracted at day 1, 3, and 5 for Hematoxylin and Eosin (H&E) and Masson’s Trichrome staining,
(n = 4), respectively using RNeasy kit (Qiagen) following the man- respectively. For paraffin slices staining, the sections were first
ufacturer’s instruction. Concentration and purity of the isolated deparaffinized, rehydrated, and then microwaved in 10 mM citric
mRNA were determined using a NanoDrop ND-1000 spectropho- acid buffer (pH 6.0) for antigen retrieval. Histological images were
tometer (Thermo Fisher Scientific, Wilmington, DE). 500 ng mRNA obtained under a light microscope (Zeiss Axio Vert.A1, Germany).
per sample was added to PrimeScriptTM RT reagent Kit (RR037A; IF stainings of angiogenic vessel markers (CD31, a-SMA) were also
Takara) to a total volume of 10 mL for cDNA synthesis at 37 °C for performed using the same procedures as described in the
15 min and RTase inactivation at 85 °C for 5 s. The cDNA product Section 2.6.
(1 mL), 10 pmol of each reverse and forward primer in 2 mL sterile
DW, 10 mL of SYBR Green real-time PCR mix (RR420A; Takara),
2.10. Statistical analysis
0.4 mL of ROX reference dye, and 6.6 mL DW were mixed in PCR
reaction tubes (Applied biosystems) to a total volume of 20 mL.
Statistical analysis of current data was performed using one-
The reaction reagents were mixed thoroughly, centrifuged briefly,
way analysis of variance (ANOVA), with Tukey’s post hoc multiple
and then placed in the q-PCR instrument (7500 Real-Time PCR Sys-
comparisons (GraphPad Prism 5). All the data represent the mean
tem; Applied biosystems), where the reaction conditions are initial
value and standard deviation. Statistically significant difference
denaturation and DNA polymerase activation at 95 °C for 5 min,
was marked as ⁄(p < 0.05), ⁄⁄(p < 0.01), or ⁄⁄⁄(p < 0.001).
followed by 40 cycles of denaturation at 95 °C for 10 s, and anneal-
ing/extension at 58 °C for 34 s. Target genes, their primer
sequences, and melting curve analysis are summarized in the 3. Results
supplementary data (Fig. S2). The sequences of primer sets of
VE-cadherin and integrin subunits are designed using the primer 3.1. hFDM composition and biochemical factors
express 3.0 software (Applied Biosystems) based on human gene
sequences from GeneBank. Glyceraldehyde 3-phosphate dehydro- According to the IF staining of hFDM before and after decellular-
genase (GAPDH) is used as a housekeeping gene. ization, macromolecular ECM components (Fn, Ln, and Col I) were
detected in both samples and they showed a fibrillar structure
2.8. Animal study: Mouse excisional wound splinting model (Fig. 1A). The clearance of nuclei was also confirmed in decellular-
ized hFDM (Fig. 1A, bottom). Based on BCA assay, while total pro-
Animal study was performed in accordance with the Korea tein content of hFDM (before decellularization) was
Institute of Science and Technology Animal Care and Use Commit- 8.36 ± 1.64 mg/cm2, that of hFDM (after decellularization) was
tee Guidelines. 8-week-old male BALB/C nude mice were housed 2.73 ± 0.55 mg/cm2 (Fig. 1B). Regarding the source of measured pro-
and cared in a temperature- and humidity-controlled animal room. teins, it is notable that previous results indicate that proteins con-
5 mm full thickness wounds were created symmetrically on the tained in the FBS are barely adsorbed onto the hFDM. From
dorsal skin using sterile disposable biopsy punches (33–35, Inte- quantitative analysis of GFs, 43.27 ± 9.46 pg/cm2 VEGF and
gra) after induced anesthesia via intraperitoneal injection of Zoletil 190.02 ± 11.27 pg/cm2 bFGF were measured before decellulariza-
and Rompun (1 mL/kg) as previously described [26]. Mice were tion, whereas 6.36 ± 0.73 pg/cm2 VEGF and 22.73 ± 2.36 pg/cm2
randomized into four groups treated with different samples, bFGF were found in hFDM (Fig. 1C and D). These results suggest
respectively (n = 3 animals per group, 2 wounds per animal): that significant gap of proteins and GFs amount is mainly due to
HUVECs in Col (Col), HUVECs in Col with GFs (Col/GFs), HUVECs the loss of intracellular components during decellularization pro-
in Col with hFDM (Col/hFDM), and HUVECs in Col with hFDM cess. hFDM mass collected from cell culture dishes was used for
and GFs (Col/hFDM/GFs). hFDM and GFs followed the same con- biochemical analysis. This hFDM mass is also utilized later in build-
centrations as shown in the Section 2.5. Before transplantation, ing a 3D construct that is composed of HUVECs, hFDM, and colla-
HUVECs were pre-labeled using a red fluorescent cell tracker gen hydrogel (Col).
(PKH-26, Sigma). 3D constructs (200 mL per sample) were prepared
in 48-well plate with HUVECs density of 2  106 cells/mL. All the 3.2. Huvecs morphology and proliferation on hFDM
samples were cultured in EGM-2 medium for 3 days to allow vas-
culatures formation before implantation. To prevent a self-healing Upon the identification of multiple ECM macromolecules (Fn,
by wound contraction, all the wounds post-transplantation were Col, Ln) and angiogenic GFs (VEGF and bFGF), HUVECs behavior
splinted by silicone 5 mm diameter washer (Invitrogen), secured on 2D hFDM was examined, along with the test of gelatin-coated
with tissue adhesive glue (1469SB; 3 M VetbondTM), and treated coverslips. Cultured in endothelial growth medium (EGM-2;
with an interrupted black silk 5–0 suture (SK526; Teleflex Medical marked ‘‘ + ”), HUVECs on gelatin-coated substrates exhibit a nor-
OEM, Korea). Wounds were covered with Tegaderm sterile dress- mal cobble stone like morphology on day 2, whereas these on
ing (3 M Healthcare, St Paul, MN) and subsequently with self- hFDM show an elongated morphology (Fig. 2A and inset). Cultured
adhering elastic bandages (3 M Healthcare), which were changed in endothelial basal medium (EBM-2; marked ‘‘”) without any
P. Du et al. / Acta Biomaterialia 54 (2017) 333–344 337

Fig. 1. hFDM characterization. Macromolecular ECM components (Col I, Fn, Ln) of human fibroblast (hFB) before or after decellularization were identified by IF staining (A).
Scale bar is 100 mm. Once total protein contents of hFB before or after decellularization were measured using BCA assay, the normalized protein amounts are indicated as mg/
cm2 (B). Total VEGF and bFGF contents were quantified via Elisa and the normalized values of VEGF (C) and bFGF (D) are marked in pg/cm2. Each experiment was performed in
triplicate. Statistically significant difference is denoted as **(p < 0.01) or ***(p < 0.001).

Fig. 2. HUVECs morphology and proliferation on 2D substrates. Phase-contrast images show HUVECs morphology on gelatin-coated coverslips and hFDM at 48 h in EGM-2
medium (A) and in EBM-2 medium (B), respectively (Scale bar = 200 mm). Insets are HUVECs co-stained via CD 31(green) and F-actin (red) at 24 h (Scale bar = 50 mm). HUVECs
morphology is quantitatively compared by measuring cell aspect ratio using image J software (C). Cell proliferation assay at 3, 24 and 72 h, respectively (D). HUVECs cultured
in EGM-2 (with supplements) is denoted (+), and in EBM-2 medium (without any supplements) is denoted (). Statistically significant difference is indicated as *(p < 0.05),
**
(p < 0.01) or ***(p < 0.001). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
338 P. Du et al. / Acta Biomaterialia 54 (2017) 333–344

supplements, HUVECs on gelatin have an abnormal morphology (Fig. 4A). As compared with Col itself, incorporation of hFDM pro-
with much lesser cell density, but those on hFDM show more elon- moted HUVECs to form a capillary-like structure (CLS) in 3D con-
gated cell morphology than those on hFDM in EGM-2 (Fig. 2B and struct as shown by F-actin staining at 3 days (Fig. 4B). This effect
inset). When HUVECs morphology is quantified by measuring cell was even more obvious at 7 days, showing that higher doses of
aspect ratio using image J, it showed the largest aspect ratio at hFDM were more effective in CLS formation in general (Fig. 4B, bot-
hFDM () condition (Fig. 2C). Furthermore, when HUVECs prolifer- tom). The presence of hFDM in 3D construct (no HUVECs) is visu-
ation in different medium conditions were compared, cells number alized via Fn staining (Fig. 4B, inset). The CLS formation is
on each substrate was similar at the early time point (3 h). How- quantitatively compared via two parameters: number of segments
ever, HUVECs proliferation on hFDM was better than that on gela- and total segment length. On day 3, the number of segments in all
tin in both EGM-2 (+) and EBM-2 () medium at 24 and 72 h, hFDM-incorporated groups is significantly higher than that with-
respectively (Fig. 2D). Significant increase of cells number on hFDM out hFDM (Col) and that of 250 hFDM and 500 hFDM group is
in EBM-2 medium within 24 h but little change of cells number on about twice that of 100 hFDM (Fig. 4C). This trend was similar at
gelatin in the same condition was notable. 7 days. The total capillary length of hFDM-incorporated constructs
was more than five times longer than Col on day 3, and the trend
3.3. Observation of cell-matrix interactions was maintained until day 7 (Fig. 4D). The quantitative difference
between 250 hFDM and 500 hFDM is not statistically significant.
Cell-matrix interactions play a very critical role in regulating When the extent of CLS formation in a larger scale at 7 days was
HUVECs vascular network formation. Fn is one of the most impor- further examined by tile-scanned large dimensional images (mm
tant ECM components and ECs actively interact with surrounding scale), they supported more active CLS formation with higher dose
environment by either breaking down existing ECM or secreting of hFDM and showed the homogeneous distribution of CLS through
new Fn [27]. Therefore, Fn staining of hFDM and F-actin staining the whole 3D construct (Fig. S3). In addition, the stability of CLS
of HUVECs can visualize how matrix fibers of hFDM interact with with time is very critical and thus each group was stained using
HUVECs. HUVECs appeared to be embedded inside the matrix toluidine blue O on day 14 (Fig. S4). The results showed that few
fibers of hFDM as indicated via z-stack confocal microscope images CLS was detected in Col group, whereas CLS in hFDM-
and showed largely elongated cell morphology (Fig. 3A). More incorporated constructs was maintained for 14 days and the den-
elongated cell morphology was noticed on hFDM in EBM-2 med- sity of CLS was directly correlated with hFDM doses.
ium than in EGM-2 medium, but a hollow luminal structure was
undetectable in both conditions. In addition, it was notable that 3.5. Huvecs vascular morphogenesis in 3D construct: Effect of hFDM
newly synthesized Fn by HUVECs was found on gelatin-coated sub- and GFs
strate at 24 h and more positive signals of Fn were observed in
EGM-2 medium than in EBM-2 medium (Fig. 3B). HUVECs can also Since the results in 3.4 indicated similar effects between
synthesize new Fn on the hFDM but the difference of Fn itself syn- 250 hFDM and 500 hFDM, we selected 250 hFDM as an optimal
thesized on either gelatin or hFDM is unknown at this time. dose and combined it with angiogenic GFs to investigate their syn-
ergistic activities on vascular morphogenesis in 3D construct. On
3.4. Huvecs vascular morphogenesis in 3D construct: Effect of hFDM day 3, comparable level of CLS formation was observed between
dose Col/GFs and Col/hFDM, and the highest level was found in Col/
hFDM/GFs. The same trend was witnessed at 7 days but Col/hFDM
The HUVECs behaviors on 2D hFDM demonstrate its supportive showed much more effectiveness than Col/GFs (Fig. 5A). When seg-
role for HUVECs adhesion, proliferation, and capillary-like mor- ments number and total capillary length were quantitatively eval-
phology. Next step is the examination of hFDM effect in 3D envi- uated on day 3, Col/GFs, Col/hFDM, and Col/hFDM/GFs were
ronment by combining different doses of hFDM with Col significantly better in CLS formation than Col, and the best was

Fig. 3. Cell-matrix interactions. 3D reconstructed confocal microscope z-stack images (24 h) exhibit that HUVECs are embedded in the matrix fibers of hFDM in EGM-2 and
EBM-2 medium, respectively. hFDM was stained using IF of Fn (green), and HUVECs were via F-actin (red) and DAPI (blue) staining (A). Cell-matrix interactions on gelatin-
coated substrate and hFDM in different medium conditions were visualized at 24 h via Fn staining (green), and HUVECs were stained by F-actin (magenta) (B). Scale bar is
50 lm. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)
P. Du et al. / Acta Biomaterialia 54 (2017) 333–344 339

Fig. 4. hFDM dose effect on HUVECs vasculogenesis in 3D constructs. Schematic shows HUVECs incorporated in Col with or without hFDM (A). HUVECs morphology on day 3
and 7 was visualized by F-actin (red), DAPI (blue) staining, and hFDM was detected via IF of Fn (green) (inset) (B). The Fn signal in the inset is derived from hFDM, not HUVECs
and indicates the presence of hFDM and its distribution in the construct. CLS formation in each group was quantitatively compared using two parameters: number of
segments (C) and total capillary lengths (D). CLS formation is progressively improved upon hFDM-dose dependent manner. Scale bar is 200 mm. Statistically significant
difference among the test groups is indicated as *(p < 0.05), **(p < 0.01) or ***(p < 0.001).

Fig. 5. Effect of both hFDM and GFs on HUVECs vasculogenesis in 3D constructs. HUVECs morphology on day 3 and 7 was examined using F-actin (red), DAPI (blue) staining,
and the presence of hFDM was spotted via IF of Fn (green) (A). Scale bar is 200 mm. Insets are the schematic of hFDM and/or GFs and HUVECs incorporated in Col. CLS
formation in each group was quantitatively compared via two parameters: number of segments (B) and total capillary length (C). Statistically significant differences are
marked as *(p < 0.05), **(p < 0.01) or ***(p < 0.001). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

Col/hFDM/GFs (Fig. 5B and C). The decreasing trend of total length than that of Col and Col/GFs. The difference between Col/
capillary length was obvious on day 7, especially with the hFDM and Col/hFDM/GFs was also statistically significant on day
GFs-containing groups, Col/GFs and Col/hFDM/GFs. And the 7. Although the exact mechanism is unclear at this time, it is likely
hFDM-containing groups were much better with total capillary that hFDM plays a role in stabilizing the bioactivity of GFs
340 P. Du et al. / Acta Biomaterialia 54 (2017) 333–344

compared to Col/GFs group that lacks hFDM and thus loses GFs improving the lumen diameter with time. In addition when the
efficacy faster. In addition, more analysis of DAPI-positive stains capillary luminal space was visualized by cross-sectioned 3D
of Fig. 5A shows that there was a significant increase of ECs num- reconstructed confocal microscope images (Fig. S6A), there is little
ber per 100 lm capillary length for Col/hFDM/GFs at 7 day com- change of the cell number per capillary diameter with time except
pared to that on day 3 (Fig. S5). the Col group (Fig. S6B).

3.6. hFDM and GFs significantly increase capillary lumen diameter 3.7. Angiogenic profiling of hFDM

To better understand the quality of capillary structure, IF Upon the confirmation of solid angiogenic potential of hFDM,
images of CLS were taken under confocal microscope at higher res- we have investigated the hFDM further using 55 angiogenesis-
olution. Accumulated z-stack images exhibit different level of cap- related antibodies array kit (Fig. 7A). According to the intensity of
illary structures on each group (Fig. 6A). When the diameter of positive dots, most abundant angiogenesis-related cytokines
lumen was quantitatively analyzed, the diameter in both Col/hFDM detected in hFDM are coagulation factor III, dipeptidyl peptidase
(8.46 ± 2.65 mm) and Col/hFDM/GFs (8.52 ± 2.03 mm) was signifi- IV (DPPIV), FGF-basic, hepatocyte growth factors (HGF), serpin E1
cantly larger than that of either Col (5.23 ± 3.26 mm) or Col/GFs (plasminogen activator inhibitor-1), tissue inhibitor of
(6.06 ± 1.74 mm) at 3 days (Fig. 6B). It was interesting to see that metalloproteinase-1 (TIMP-1), thrombospondin-1 (TSP-1), uroki-
the luminal structure on day 7 was dramatically regressed in the nase plasminogen activator (uPA). The intensity of these dots is
Col group but that of Col/hFDM/GFs was still sustainable and the more than 50% of the positive signals (Fig. 7B). The relatively abun-
size even increased to a maximum diameter of 68.05 mm (Fig. 6C), dant components are amphiregulin, endostatin, FGF-acidic, FGF-7,
much better than that of Col/GFs and Col/hFDM. The average VEGF and their intensity is 30 to 50% to the positive ones. The assay
lumen diameter at 7 days in Col/hFDM/GFs is 27.78 ± 16.61 mm, results demonstrate the angiogenic activity of hFDM and strongly
which is three times larger than that at 3 days. This may explain support the early data in this study.
the results of Col/hFDM/GFs (Fig. 5C) that showed significantly
reduced total capillary lengths at 7 days. The lumen diameters in 3.8. VE-cadherin and integrin receptors expression
Col/GFs and Col/hFDM slightly decreased with time. This result
suggests that the combined hFDM and GFs can be synergistic in Cell-cell contacts between ECs are crucial for vasculature for-
stimulating capillary remodeling and accordingly significantly mation. VE-cadherin is one of the cell-cell adhesion molecules that

Fig. 6. hFDM and GFs enhance capillary lumen diameters with time. CLS on day 3 and 7 was visualized via F-actin (red) and DAPI (blue) staining, and the entity of hFDM was
identified via IF of Fn (green) (A). Scale bar is 50 mm. CLS lumen diameters in each group were quantitatively determined on day 3 (B) and 7 (C), respectively. Statistically
significant difference is indicated as *(p < 0.05), **(p < 0.01) or ***(p < 0.001). (For interpretation of the references to colour in this figure legend, the reader is referred to the web
version of this article.)
P. Du et al. / Acta Biomaterialia 54 (2017) 333–344 341

Fig. 7. Angiogenesis related factors analysis. Angiogenesis-related factors in hFDM were analyzed using a sandwich-based human angiogenesis antibody array kit (A). The
positive signals (individual dots) were normalized to the dot intensity of negative (0%; F23 and F24) or positive control values (100%; A1, A2, A23, A24, F1, and F2) (B). The
quantitative data are obtained from three biological replicates.

play a crucial role in the control of vasculature permeability and the integrin receptors of Ln, nidogen, Col IV and Fn are significantly
integrity [28]. When the gene expression level of VE-cadherin in up-regulated with Col/hFDM and Col/hFDM/GFs on day 5 (Fig. 8B).
each group was examined, it was intriguing that VE-cadherin This result suggests a possibility that macromolecular components
expression steadily increased with time only in Col/hFDM/GFs in hFDM may actively interact with counterpart integrin receptors
group (Fig. 8A). While the level of VE-cadherin was similar to each in promoting HUVECs vasculogenesis by up-regulation of the
other on day 1, it gradually climbed for Col/hFDM/GFs and reached related integrin receptors.
the level to which the difference was statistically significant at
5 days compared with the other groups. This result provides an 3.9. Therapeutic effect of pre-vascularized 3D construct for skin wound
implication related to the establishment of cell-cell contacts for
the creation and maintenance of vasculatures. Upon the results Previously tested 3D constructs with HUVECs are examined for
of larger capillary lumen formation in Col/hFDM/GFs on day 7 their therapeutic effect via skin wound model. The wounds treated
(Fig. 6C), it is plausible to reason that VE-cadherin may have by Col/GFs, Col/hFDM, and Col/hFDM/GFs appeared to have a fully
involved in the process of vasculatures remodeling to reshape recovered skin at 14 days whereas those treated by Col exhibited
CLS with larger diameters [29]. To investigate whether macro- an incomplete recovery (Fig. 9A). When the extent of wound heal-
molecular proteins in hFDM are related to HUVECs vasculogenesis, ing was quantitatively evaluated by wound occupied area (%) on
expression of specific integrin receptors was monitored with time. day 3, 5, 7 and 14, it presented that Col/hFDM/GFs was faster
On day 1, all the integrin subunits a1, 2, 3, 5, 6 and b1 presented a and more effective in wound healing compared to the other groups
similar expression level among four test groups (Fig. S7). However (Fig. 9B). As the wound regeneration was further investigated by

Fig. 8. Gene expression analysis of VE-cadherin and integrins in 3D constructs. VE-cadherin expression was examined with HUVECs cultured in different 3D constructs on day
1, 3, and 5 (A). Integrin subunits, a1, a2, a3, a5, a6, and b1 expression level was also compared on day 5 (B). Statistically significant differences are marked as *(p < 0.05),
**
(p < 0.01) or ***(p < 0.001).
342 P. Du et al. / Acta Biomaterialia 54 (2017) 333–344

Fig. 9. Therapeutic effect of transplanted 3D constructs in skin wound model. Gross images exhibit skin wound healing process during 14 days (A). Initial wound size is 5 mm.
Wound size is quantitatively analyzed at different time points (B). Skin regeneration in the wound area was evaluated by H&E staining on day 14 (C). Scale bar is 500 mm.
Arrow heads (blue) indicate the neoepidermis and asterisks (blue) denote regenerated hair follicles. Arrows (red) mark the border of initial wound area. Moreover,
arrowheads (blue) indicate the neo-epidermis and this area is shown in a higher magnification (D). Scale bar is 100 mm. Statistically significant differences are marked as
*
(p < 0.05), **(p < 0.01) or ***(p < 0.001).

H&E staining, the wound treated with Col was poorly Numerous biochemical cues and biomaterials have been used to
re-epithelialized but the one treated by Col/hFDM/GFs formed a construct angiogenic 3D environments. In this study, when hFDM
smooth epidermis, with many regenerated hair follicles that were was selected and intensively investigated for its angiogenic poten-
barely detectable in Col/GFs and Col/hFDM (Fig. 9C). Higher magni- tial, the overall results suggested solid angiogenic activities of
fication images in the neo-epidermis areas support this finding hFDM in 3D environment. According to the compositional and bio-
(Fig. 9D). In addition, Masson’s Trichrome staining also confirmed chemical analysis, hFDM reserves multiple GFs, proteolytic
the advanced therapeutic effect of Col/hFDM/GFs with higher col- enzymes, and ECM macromolecules, all of which play a crucial role
lagen deposition, and signs of neovascularization via the presence in regulating vascular cells behavior. The combination of hFDM and
of erythrocytes (Fig. S8). Moreover, the transplanted 3D constructs GFs demonstrates significant improvements of HUVECs vascular
in the wound model was further evaluated for neovascularization morphogenesis, more robust CLS formation, and much effective
via immunostaining of angiogenic markers: CD31 and a-SMA. wound healing in vivo.
The results show that neovessels were detected in all the groups With an effort to understand critical factors that would stimu-
but they were more abundant in Col/hFDM/GFs group (Fig. 10A). late HUVECs vasculogenic activity, the role of each component in
As the blood vessel density (#/mm2) and wound occupied area hFDM needs to be addressed. First of all, according to the quantifi-
(%) were quantitatively analyzed, higher vessel density was obvi- cation of GFs contents in hFDM (Fig. 1C and D), the highest dose of
ous in the wounds treated by Col/hFDM (54 ± 25) and Col/hFDM/ hFDM (500 mg/mL) is estimated to contain about 1.2 ng/mL VEGF
GFs (58 ± 23) better than Col (26 ± 12) or Col/GFs (33 ± 10) and 4.2 ng/mL bFGF, respectively. Compared with the soluble GFs
(Fig. 10B). A similar trend was observed with blood vessels occu- (VEGF, bFGF, and SDF-1a) concentrations (200 ng/mL, each) used
pied area, in which Col/hFDM/GFs showed much larger percentage in this study, the amount of those GFs in hFDM seems negligible
than the other groups (Fig. 10C). To investigate the potential mech- to be effective. Other biochemical factors as shown in Fig. 7, how-
anism of Col/hFDM/GFs on such a therapeutic efficacy in the ever may explain HUVECs vasculogenesis in 3D construct. Among
wound area, the HUVECs labeled via PKH-26 fluorescent dye before these factors, FGF-basic, FGF-acidic, FGF-7, VEGF, HGF are very
transplantation are tracked down at14 day. While PKH-26 labeled important proangiogenic GFs [30]. Coagulation factor III, known
HUVECs are observed in all the groups, the most positive signals as a tissue factor maintains the vascular integrity of tissue by ini-
were noticed in both Col/hFDM and Col/hFDM/GFs (Fig. S9). In tiating the coagulation cascade after vessel injury [31]. Amphireg-
addition, neovessel stained by a-SMA (green) was also detected ulin is an EGF family GFs, highly expressed in mammary epithelial
in Col/hFDM/GFs group. However a direct link between trans- cells, and clinical studies have suggested it plays some roles in
planted HUVECs and host vessels was unidentified. human cancer progression [32]. Both TSP-1 and endostatin are
reported as anti-angiogenic agents [33,34].
In addition, we postulate that proteolytic enzymes in hFDM
4. Discussion may have played a certain role in facilitating HUVECs migration
to form vasculatures via ECM remodeling. Proteolytic ECM degra-
The extracellular microenvironments of vascular cells contain a dation by stromal cells has shown to increase neovascularization
variety of instructive cues. Each component has a specific role in by either releasing sequestered angiogenic factors or providing
regulating cell behaviors and in maintaining a vascular system. physical guidance cues [35]. Current data of human angiogenesis
P. Du et al. / Acta Biomaterialia 54 (2017) 333–344 343

Fig. 10. Neovascularization of transplanted 3D constructs in skin wound model. New vessel formation in the wound areas was investigated via CD31 and a-SMA staining,
respectively (A). Neovascularization was quantitatively evaluated using vessels density per mm2 (B) and vessels occupied area in percentage (C). Scale bar is 50 mm.
Statistically significant differences are marked as *(p < 0.05), **(p < 0.01) or ***(p < 0.001).

antibody array demonstrate that DPPIV and uPA are the most HUVECs adhesion, proliferation, and vascular morphogenesis. The
abundant enzymes in hFDM (Fig. 7). Previous study found out that up-regulation of integrin subunit genes of HUVECs in Col/hFDM/
DPPIV and uPA was implicated in remodeling of ECM to enhance GFs (Fig. 8B) suggests a possibility that the macromolecular com-
cell adhesion, proliferation and migration [36,37]. In addition, ponents in hFDM may actively interact with counterpart integrin
TIMP-1 and serpin E1 are also proteolytic enzymes involved in receptors during HUVECs vasculogenesis. Interestingly, much
the angiogenic processes via modulation of ECM remodeling. If this higher expression level of integrin a3 in Col/hFDM/GFs than Col/
hypothesis is true, human lung fibroblasts (hFB) conditioned med- GFs suggests the positive effect of interactions between integrin
ium (CM) may have some effects on HUVECs vasculogenesis since a3 and integrin a3 specific ECM molecules (nidogen and laminin)
human lung fibroblasts secrete these enzymes into culture med- and this may play a role in facilitating vasculature formation and
ium. To test this hypothesis, HUVECs were cultured in Col, with remodeling into large vessels in 3D construct. The presence of
the addition of CM inside or outside the Col or both sides. Com- nidogen was not confirmed in this study.
pared with the Col group, CM did not show any positive effects Currently we have demonstrated the great potential of hFDM as
on HUVECs vasculogenic behaviors on day 2 (Fig. S10). This result a key constituent of angiogenic 3D construct in vitro. For in vivo
suggests that proteolytic enzymes in CM are insufficient or inactive study, we find that Col/hFDM/GFs group with the highest level of
to develop a capillary formation. It would be very interesting to fig- cell engraftment promotes skin wound healing much better than
ure out the most active constituent in the hFDM. Our previous other groups. This result is partly explained by an indirect, para-
work examined 3D construct containing Fn (instead of hFDM), crine effect of transplanted cells as reported in the previous litera-
GFs, Col, and HUVECs but we could not observe a nicely formed tures [44,45]. Angiogenic factors secreted by the transplanted
capillary structure (data not shown). HUVECs, and those contained in hFDM can play a major role in
The macromolecular components of hFDM can also improve encouraging neovessel formation in the wound area and thus
cell-matrix interactions and concurrently trigger angiogenic sig- result in the improvement of wound healing. Regarding the impor-
naling. Besides cell-cell contacts, cell adhesion to specific compo- tance of functional vessel formation in vivo, however current study
nents of ECM is considered critical for vascular morphogenesis is unable to find direct evidences whether the transplanted
[38]. Many studies suggest that ECs-matrix coupling through the HUVECs are linked to the host vasculature. We consider some
cell surface receptors initiates the matrix-integrin-cytoskeletal sig- other strategies to improve our system by delivering more number
nal transduction, which causes simultaneous angiogenic sprouting of ECs or adding perivascular cells. Since perivascular cells and
[39]. Specific interactions between extracellular components and multiple BM macromolecules stabilize native blood vessels, cur-
integrins are essential in ECs vascular morphogenesis in Col [40]. rent platform lacks such cell type. For more mature and functional
The major integrins responsible for such interactions are a1b1 vasculature construction, HUVECs and pericytes co-culture system
(Ln/ collagen IV), a2b1 (Col I), a3b1 (nidogen/Ln), a5b1 (Fn), and can be a next stop. Further in-depth study including hFDM-
a6b1 (Ln) [41]. In particular, binding between Fn and its receptor mediated intracellular signaling should shed a light on the mecha-
integrin a5b1 is an important step in the angiogenesis process of nism of HUVECs vasculogenesis in 3D environment. hFDM can find
ECs [42,43]. Higher integrin a5 expressions of HUVECs on 2D various applications with other biomaterials to develop a new plat-
hFDM (Fig. 3B) support the crucial role of Fn in promoting form for functional vasculature formation.
344 P. Du et al. / Acta Biomaterialia 54 (2017) 333–344

5. Conclusions [17] A.C. Hielscher, C. Qiu, S. Gerecht, Breast cancer cell-derived matrix supports
vascular morphogenesis, Am. J. Physiol. Cell Physiol. 302 (2012) C1243–C1256.
[18] F. Berthod, L. Germain, N. Tremblay, F.A. Auger, Extracellular matrix deposition
In summary, one of the major findings of this study is that by fibroblasts is necessary to promote capillary-like tube formation in vitro, J.
hFDM is not just an assembly of ECM macromolecules but contains Cell. Physiol. 207 (2006) 491–498.
[19] A.C. Newman, M.N. Nakatsu, W. Chou, P.D. Gershon, C.C.W. Hughes, The
angiogenesis-related factors, such as GFs, proteolytic enzymes, and
requirement for fibroblasts in angiogenesis: fibroblast-derived matrix proteins
others. Therefore, hFDM offers a great angiogenic microenviron- are essential for endothelial cell lumen formation, Mol. Biol. Cell 22 (2011)
ment for HUVECs vascular morphogenesis in 3D environment. 3791–3800.
[20] S. Kim, H. Lee, M. Chunga, N.L. Jeon, Engineering of functional, perfusable 3D
Incorporation of GFs and hFDM into 3D construct supports their
microvascular networks on a chip, Lab Chip 13 (2013) 1489–1500.
synergistic activities via much advanced angiogenic parameters [21] P. Du, R. Subbiah, J.H. Park, K. Park, Vascular morphogenesis of human
and significantly enlarged capillary diameters with time for up to umbilical vein endothelial cells on cell-derived macromolecular matrix
7 days. In addition, up-regulated gene expression of cell-cell adher- microenvironment, Tissue Eng. Part A 20 (2014) 2365–2377.
[22] M.D. Amatangelo, D.E. Bassi, A.J. Klein-Szanto, E. Cukierman, Stroma-derived
ent molecule, VE-cadherin and ECM specific integrin receptors are three-dimensional matrices are necessary and sufficient to promote
also notable, especially with Col/hFDM/GFs. Another benefit of Col/ desmoplastic differentiation of normal fibroblasts, Am. J. Pathol. 167 (2005)
hFMD/GFs is certified via skin wound model that shows effective 475–488.
[23] M.C. Prewitz, F.P. Seib, M. von Bonin, J. Friedrichs, A. Stißel, C. Niehage, K.
re-epithelialization, nearly complete regeneration of hair follicles, Müller, K. Anastassiadis, C. Waskow, B. Hoflack, M. Bornhäuser, C. Werner,
better transplanted cells viability, and much improved neovascu- Tightly anchored tissue-mimetic matrices as instructive stem cell
larization. We believe that current platform is very promising in microenvironments, Nat. Methods 10 (2013) 788–799.
[24] A.N. Stratman, K.M. Malotte, R.D. Mahan, M.J. Davis, G.E. Davis, Pericyte
the construction of 3D vasculature in vitro and as a cell delivery recruitment during vasculogenic tube assembly stimulates endothelial
carrier for therapeutic applications in vivo. basement membrane matrix formation, Blood 114 (2009) 5091–5101.
[25] A.O. Smith, S.L. Bowers, A.N. Stratman, G.E. Davis, Hematopoietic stem cell
cytokines and fibroblast growth factor-2 stimulate human endothelial cell-
Acknowledgements pericyte tube co-assembly in 3D fibrin matrices under serum-free defined
conditions, PLoS One 8 (2013) e85147–e85166.
[26] X. Wang, J. Ge, E.E. Tredget, Y. Wu, The mouse excisional wound splinting
This work was supported by National Research Foundation of
model, including applications for stem cell transplantation, Nat. Protoc. 8
Korea (NRF) grant (No. 2015R1 A2A2A04004469) from the Min- (2013) 302–309.
istry of Science, ICT and Future Planning, and by Korea Health [27] J.T. Daub, R.M. Merks, A cell-based model of extracellular-matrix-guided
Industry Development Institute (KHIDI), funded by the Ministry endothelial cell migration during angiogenesis, Bull. Math. Biol. 75 (2013)
1377–1399.
of Health and Welfare (HI16C0133), Republic of Korea. [28] E.S. Harris, W.J. Nelson, VE-cadherin: at the front, center, and sides of
endothelial cell organization and function, Curr. Opin. Cell Biol. 22 (2010) 651–
658.
Appendix A. Supplementary data [29] S. Yang, J. Graham, J.W. Kahn, E.A. Schwartz, M.E. Gerritsen, Functional roles
for PECAM-1 (CD31) and VE-cadherin (CD144) in tube assembly and lumen
formation in three-dimensional collagen gels, Am. J. Pathol. 155 (1999) 887–
Supplementary data associated with this article can be found, in 895.
the online version, at http://dx.doi.org/10.1016/j.actbio.2017.03. [30] K. Xie, D. Wei, S. Huang, Transcriptional anti-angiogenesis therapy of human
035. pancreatic cancer, Cytokine Growth Factor Rev. 17 (2006) 147–156.
[31] J.E. Bluff, N.J. Brown, M.W. Reed, C.A. Staton, Tissue factor, angiogenesis and
tumour progression, Breast Cancer Res. 10 (2008) 204–214.
References [32] B. Busser, L. Sancey, E. Brambilla, J.L. Coll, A. Hurbin, The multiple roles of
amphiregulin in human cancer, Bba-Rev Cancer 2011 (1816) 119–131.
[1] S.M. Weis, D.A. Cheresh, Tumor angiogenesis: molecular pathways and [33] J.E. Nor, R.S. Mitra, M.M. Sutorik, D.J. Mooney, V.P. Castle, P.J. Polverini,
therapeutic targets, Nat. Med. 17 (2011) 1359–1370. Thrombospondin-1 induces endothelial cell apoptosis and inhibits
[2] N.P. Fam, S. Verma, M. Kutryk, D.J. Stewart, Clinician guide to angiogenesis, angiogenesis by activating the caspase death pathway, J. Vasc. Res. 37
Circulation 108 (2003) 2613–2618. (2000) 209–218.
[3] A. Lesman, J. Koffler, R. Atlas, Y.J. Blinder, Z. Kam, S. Levenberg, Engineering [34] M.S. O’Reilly, T. Boehm, Y. Shing, N. Fukai, G. Vasios, W.S. Lane, E. Flynn, J.R.
vessel-like networks within multicellular fibrin-based constructs, Biomaterials Birkhead, B.R. Olsen, J. Folkman, Endostatin: an endogenous inhibitor of
32 (2011) 7856–7869. angiogenesis and tumor growth, Cell 88 (1997) 277–285.
[4] E.C. Novosel, C. Kleinhans, P.J. Kluger, Vascularization is the key challenge in [35] Y.H. Song, S.H. Shon, M. Shan, A.D. Stroock, C. Fischbach, Adipose-derived stem
tissue engineering, Adv. Drug Deliv. Rev. 63 (2011) 300–311. cells increase angiogenesis through matrix metalloproteinase-dependent
[5] F.A. Auger, L. Gibot, D. Lacroix, The Pivotal role of vascularization in tissue collagen remodeling, Integr. Biol. 8 (2016) 205–215.
engineering, Annu. Rev. Biomed. Eng. 15 (2013) 177–200. [36] G. Ghersi, Q. Zhao, M. Salamone, Y. Yeh, S. Zucker, W.T. Chen, The protease
[6] V.W.M. van Hinsbergh, A. Collen, P. Koolwijk, Role of fibrin matrix in complex consisting of dipeptidyl peptidase IV and seprase plays a role in the
angiogenesis, Ann. N. Y. Acad. Sci. 936 (2001) 426–437. migration and invasion of human endothelial cells in collagenous matrices,
[7] Y.J. Blinder, D.J. Mooney, S. Levenberg, Engineering approaches for inducing Cancer Res. 66 (2006) 4652–4661.
blood vessel formation, Curr. Opin. Chem. Eng. 3 (2014) 56–61. [37] M.J. Duffy, The urokinase plasminogen activator system: role in malignancy,
[8] H. Shin, S. Jo, A.G. Mikos, Biomimetic materials for tissue engineering, Curr. Pharm. Des. 10 (2004) 39–49.
Biomaterials 24 (2003) 4353–4364. [38] R.O. Hynes, Cell-matrix adhesion in vascular development, J. Thromb.
[9] G.E. Davis, D.J. Kim, C.X. Meng, P.R. Norden, K.R. Speichinger, M.T. Davis, A.O. Haemost. 5 (2007) 32–40.
Smith, S.L. Bowers, A.N. Stratman, Control of vascular tube morphogenesis and [39] G.E. Davis, K.J. Bayless, A. Mavila, Molecular basis of endothelial cell
maturation in 3D extracellular matrices by endothelial cells and pericytes, morphogenesis in three-dimensional extracellular matrices, Anat. Rec. 268
Methods Mol. Biol. 1066 (2013) 17–28. (2002) 252–275.
[10] P. Carmeliet, Manipulating angiogenesis in medicine, J. Intern. Med. 255 [40] G. Serini, D. Valdembri, F. Bussolino, Integrins and angiogenesis: a sticky
(2004) 538–561. business, Exp. Cell Res. 312 (2006) 651–658.
[11] C.J. Kirkpatrick, S. Fuchs, R.E. Unger, Co-culture systems for vascularization– [41] G.E. Davis, D.R. Senger, Endothelial extracellular matrix: biosynthesis,
learning from nature, Adv. Drug Deliv. Rev. 63 (2011) 291–299. remodeling, and functions during vascular morphogenesis and neovessel
[12] F.T. Bosman, I. Stamenkovic, Functional structure and composition of the stabilization, Circ. Res. 97 (2005) 1093–1107.
extracellular matrix, J Pathol. 200 (2003) 423–428. [42] A. Hielscher, K. Ellis, C. Qiu, J. Porterfield, S. Gerecht, Fibronectin deposition
[13] J.K. Kular, S. Basu, R.I. Sharma, The extracellular matrix: structure, participates in extracellular matrix assembly and vascular morphogenesis,
composition, age-related differences, tools for analysis and applications for PLoS One 11 (2016) e0147600–e0147627.
tissue engineering, J. Tissue Eng. 5 (2014) 1–17. [43] S. Astrof, R.O. Hynes, Fibronectins in vascular morphogenesis, Angiogenesis 12
[14] M. Aumailley, Structure and supramolecular organization of basement (2009) 165–175.
membranes, Kidney Int. Suppl. 49 (1995) S4–S7. [44] J.S. Choi, J.D. Kim, H.S. Yoon, Y.W. Cho, Full-thickness skin wound healing using
[15] X. Yang, H.A. Scott, F. Monickaraj, J. Xu, S. Ardekani, C.F. Nitta, A. Cabrera, P.G. human placenta-derived extracellular matrix containing bioactive molecules,
McGuire, U. Mohideen, A. Das, K. Ghosh, Basement membrane stiffening Tissue Eng. Part A 19 (2013) 329–339.
promotes retinal endothelial activation associated with diabetes, FASEB J. 30 [45] M.P. Morissette, A. Maux, V. Laterreur, D. Mayrand, V.L. Gagné, V.J. Moulin, J.
(2016) 601–611. Fradette, Enhancing repair of full-thickness excisional wounds in a murine
[16] P.A. Soucy, L.H. Romer, Endothelial cell adhesion, signaling, and model: impact of tissue-engineered biological dressings featuring human
morphogenesis in fibroblast-derived matrix, Matrix Biol. 28 (2009) 273–283. differentiated adipocytes, Acta Biomater. 22 (2015) 39–49.

You might also like