You are on page 1of 12

Acta Biomaterialia 92 (2019) 160–171

Contents lists available at ScienceDirect

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

Full length article

A collagen scaffold loaded with human umbilical cord-derived


mesenchymal stem cells facilitates endometrial regeneration and
restores fertility
Liaobing Xin a,b,c,1, Xiaona Lin a,c,1, Yibin Pan a,c, Xiaowen Zheng b, Libing Shi a,c, Yanling Zhang a,c, Lie Ma b,c,⇑,
Changyou Gao b, Songying Zhang a,c,⇑
a
Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
b
MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
c
Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, Hangzhou 310016, China

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

Article history: In women of reproductive age, severe injuries to the endometrium are often accompanied by endometrial
Received 22 November 2018 scar formation or intrauterine adhesions (IUAs), which can result in infertility or miscarriage. Although
Received in revised form 17 April 2019 many approaches have been used to treat severe IUAs, high recurrence rates and endometrial thinning
Accepted 6 May 2019
have limited therapeutic efficiency. In this study, a collagen scaffold (CS) loaded with human umbilical
Available online 7 May 2019
cord-derived mesenchymal stem cells (UC-MSCs) was fabricated and applied for endometrial regenera-
tion. The CS/UC-MSCs promoted human endometrial stromal cell proliferation and inhibited apoptosis
Keywords:
in vitro through paracrine effects. In a model of endometrial damage, transplantation with the CS/UC-
Collagen scaffold
Umbilical cord-derived mesenchymal stem
MSCs maintained normal luminal structure, promoted endometrial regeneration and collagen remodel-
cells ing, induced intrinsic endometrial cell proliferation and epithelium recovery, and enhanced the expres-
Endometrial regeneration sion of estrogen receptor a and progesterone receptor. An improved ability of the regenerated
Intrauterine adhesion endometrium to receive embryos was confirmed. Together, our results indicate that the CS/UC-MSCs pro-
Fertility restoration moted endometrial structural reconstruction and functional recovery. Topical administration of the CS/
UC-MSCs after trans-cervical resection of adhesions might prevent re-adhesion, promote endometrium
regeneration and improve pregnancy outcomes for patients with severe IUAs.

Statement of Significance

Intrauterine adhesions due to severe endometrium injuries happen frequently in clinic and become one
of the crucial reasons for women’s infertility or miscarriage. Therefore, how to regenerate the damaged
endometrium is a big challenge. In this study, a collagen scaffold (CS) loaded with human umbilical
cord-derived mesenchymal stem cells (UC-MSCs) was fabricated and applied for endometrium regener-
ation. Herein, UC-MSCs, known for low immunogenicity and high proliferative potential, exhibit promis-
ing potential for endometrium regeneration; and collagen scaffolds provide suitable physical support. It
was proved that transplantation with CS/UC-MSCs promoted endometrial regeneration and fertility
restoration. It suggested that topical administration of CS/UC-MSCs in uterus could be a promising strat-
egy for patients suffering severe intrauterine adhesion and infertility.
Ó 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction

Human endometrium is a dynamic and regenerative tissue


⇑ Corresponding authors at: Key Laboratory of Reproductive Dysfunction Man- composed of epithelial cells, stromal cells, vascular smooth cells,
agement of Zhejiang Province, No. 3 Qingchun East Road, Jianggan District, and vascular endothelial cells. The endometrium can be struc-
Hangzhou 310016, China. turally divided into two zones: the upper functional layer and
E-mail addresses: liema@zju.edu.cn (L. Ma), zhangsongying@zju.edu.cn
lower basal layer. During a menstrual cycle, the functional layer
(S. Zhang).
1
These authors contributed equally.
is shed while the permanent basal layer regenerates a brand-new

https://doi.org/10.1016/j.actbio.2019.05.012
1742-7061/Ó 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171 161

functional layer according to fluctuating levels of estrogen and pro- articular cartilage [29], periodontal tissue [30], and brain [31]. In
gesterone. However, intrauterine adhesions (IUAs), which result the reproductive field, most studies have focused on regeneration
from repeated curettage, infections and intrauterine operations, of the uterine wall or even the entire uterus. For example, an engi-
can cause damage to the endometrial basal layer [1,2]. Subsequent neered uterine tissue was constructed by encapsulating human
failure of re-epithelization, inflammation and fibrotic tissue depo- endometrial stromal cells, epithelial cells and smooth muscle cells
sition lead to shrinking of the uterine cavity and endometrium, into a collagen/Matrigel scaffold in vitro [32]. In addition, the uter-
inactive glands and poorly vascularized stroma. A meta-analysis ine horns of rats were reconstructed by the transplantation of stem
showed that approximately 19% of women experiencing miscar- cell-loaded CS grafts after partial excisions [33–35]. However,
riages were diagnosed IUAs within 12 months. Among these regeneration of an entire uterus is a major challenge because of
women, 58.1%, 28.2% and 13.7% had mild, moderate and severe the complexity of uterine tissue. Moreover, the primary cause of
IUAs, respectively [1]. Importantly, severe IUAs may be associated IUA is not disorder of the uterus or uterine walls, but damage to
with infertility, including menstrual disorders, spontaneous abor- the endometrium. In this study, we aimed to develop a CS loaded
tion and pregnancy complications [3,4]. Many strategies, including with UC-MSCs for endometrial regeneration. We demonstrated
hysteroscopy, hormone therapy, intrauterine devices and antibi- that the CS/UC-MSCs promoted endometrial stromal cells prolifer-
otics, have been used for treatment of IUAs [5–7]. However, ation and inhibited apoptosis in vitro via paracrine effects. The effi-
endometrial regeneration remains a significant challenge. Current cacies of the CS/UC-MSCs in restoring the structure and function of
treatments can temporarily normalize the shape and volume of the endometrium were assessed in a rat model of endometrial
the uterine cavity but cannot restore the structure and function damage. Finally, regenerative outcomes were assessed using a fer-
of the endometrium, leading to IUA reformation. The recurrence tility test.
rate of severe IUAs after hysteroscopy is up to 62.5% [3,8], and
the pregnancy rate following surgical treatment is only 33.3% [9].
2. Materials and methods
Multiple approaches have been proposed to prevent recurrence
of IUAs after surgery, such as application of hyaluronic acid gel in
2.1. UC-MSC monoculture, flow cytometric analysis and multipotent
humans [10], transplantation of freeze-dried amnion grafts in
differentiation analysis
humans [11], or transplantation of oral mucosal epithelial cell
sheets in rats [12]. These interventions can prevent the recurrence
Frozen UC-MSCs at passage 3 were purchased from Boyalife
of IUAs to some degree, but do not restore a functional endome-
Group Ltd. (China). After thawing, the cells were seeded in 10-cm
trium. Therefore, the question of how to reconstruct an endome-
culture dishes (1  106 cells/dish) in Dulbecco’s modified Eagle’s
trium with normal morphology and function is a vital issue for
medium/F12 (DMEM/F12, Gino Biological, Hangzhou, China) sup-
IUA treatment.
plemented with 10% (v/v) fetal bovine serum (FBS, SA112.02, Cell-
Several medications such as aspirin [13–15] and granulocyte
max, Beijing, China), 100 U/mL penicillin (Gibco), and 100 mg/mL
colony stimulating factor [16] are recommended to promote
streptomycin (Gibco). When UC-MSCs reached 80–100% conflu-
human endometrial regeneration by increasing blood perfusion.
ence, cells were subcultured in quadruplicate. UC-MSCs harvested
However, due to damaged revascularization and ischemia in severe
at passage 4–6 were used in this study.
IUA lesions, the clinical effects of these medications are limited
The phenotype and multipotent differentiation of UC-MSCs
[17].
were confirmed (Fig. S1). For flow cytometric analysis, UC-MSCs
It has become evident that endometrial stem/progenitor cells in
(1  106) were incubated with 1 mg each of fluorescein
both the functional and basal layers are responsible for endometrial
isothiocynate(FITC)-conjugated anti-human CD73 (Biolegend,
regeneration [18–20]. In IUAs, the numbers and function of endome-
344016, USA), phycoerythrin (PE)-conjugated anti-human HLA-
trial stem/progenitor cells are diminished due to the lack of func-
DR (Beckman, IM1639, USA), anti-human CD34 (Beckman,
tional endometrium [3]. Recently, cell therapy toward uterus have
A07776, USA), anti-human CD45 (Beckman, A07783, USA), anti-
been popular in treating endometrial dysfunction [20]. Mesenchy-
human CD19 (Beckman, A07769, USA), anti-human CD11b (Beck-
mal stem cells (MSCs) have emerged as a promising cell type for tis-
man, IM2581U, USA), PE/CF594-conjugated anti-human CD90
sue regeneration since they are characterized by multipotent
(Beckman, IM3703, USA), and PE/Cy7-conjugated anti-human
differentiation, high expansive potential, and immunomodulatory
CD105 (Biolegend, 323218, USA) antibodies for 30 min in the dark.
capability. Depending on the cellular source, MSCs can be classified
After washing twice with phosphate-buffered saline (PBS), the
as bone marrow-derived, umbilical cord-derived, adipose-derived,
samples were analyzed using a flow cytometer (Beckman Coulter,
and human menstrual blood-derived MSCs. Umbilical cord-
Fullerton, CA, USA). For osteogenic differentiation analysis, UC-
derived MSCs (UC-MSCs) have been regarded as a promising source
MSCs (5  103 cells/cm2) were treated with 0.1 lM dexametha-
for cell-based therapies because of their easy collection, low
sone (Sigma-Aldrich, St Louis, MO, USA), 10 mM b-glycerol phos-
immunogenicity, and high proliferative potential [21]. Applications
phate (Sigma-Aldrich, St Louis, MO, USA) and 50 lg/mL ascorbic
of UC-MSCs include bone defects [22], renal diseases [23], and
acid (Sigma-Aldrich, St Louis, MO, USA). After incubating for
peripheral nerve injuries [24]. However, few studies have reported
15 days, positive induction was observed by Alizarin Red staining.
the application of UC-MSCs to promote endometrial repair.
For analysis of adipogenic and chondroblast differentiation, cells
The main obstacle to treatment with MSCs is their low local
were incubated in adipogenic differentiation medium (Biological
persistence and utilization rate in the endometrium [25]. Only
Industries, Israel) or chondrogenic differentiation medium (Biolog-
0.59% or 0.65% of engrafted cells were observed around small
ical Industries, Israel) for 15 days according to the manufacturer’s
endometrial vessels following intrauterine infusion or tail vein
instructions. Positive induction was observed by oil red staining
injection in a murine model [26]. As critical components of tissue
and Alcian Blue staining.
engineering materials, porous scaffolds play an essential role in tis-
sue repair and regeneration. Collagen, a natural biomaterial and
the main component of the extracellular matrix, has been widely 2.2. Human endometrial stromal cell (HESC) isolation, culture and
utilized for tissue engineering scaffolds. A collagen scaffold (CS) immunofluorescence analysis
can provide a suitable physical support and microenvironment
for transplanted stem cells [27,28]. CSs loaded with stem cells have HESCs were obtained from fresh endometrial specimens as pre-
been applied for regeneration of many tissues and organs including viously described [36]. Informed consent was obtained from all
162 L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171

patients prior to endometrium tissue collection. The procedures UC-MSCs (a 0.5  0.5 cm CS seeded with 2  105 UC-MSCs) was
were approved by the ethics committee of Sir Run Run Shaw placed on top of the HESC-seeded plate. HESCs cultured in
Hospital, Zhejiang University. In brief, endometrial specimens were DMEM/F12 containing a blank insert and HESCs cultured in
collected by biopsy from women with normal menstrual cycles and DMEM/F12 containing 10% FBS were used as negative and positive
rinsed with Hank’s Balanced Salt Solution (HBSS, Invitrogen, USA) controls, respectively. The medium was refreshed every 24 h. A
on ice. The samples were minced to <1 mm3 and incubated in CCK-8 assay (Dojindo, Shanghai, China) was used to evaluate the
DMEM/F12 containing 0.25% (w/v) collagenase at 37 °C for proliferation of HESCs according to the manufacturer’s protocol.
90 min. After digestion, stromal cells were obtained by passage Three samples from each group were assessed in the CCK-8 assay
through a 40-mm nylon cell strainer (BD Biosciences) and washed (n = 3).
twice with HBSS. The stromal cells were seeded in 35-mm plates For cell apoptosis assays, HESCs were seeded into 6 well plates
(1.5  105 cells/well) in DMEM/F12 supplemented with 10% FBS, and grown to 80% confluence. An insert containing a CS/UC-MSCs
100 U/mL penicillin (Gibco) and 100 lg/mL streptomycin (Gibco). (a 2  2 cm collagen scaffold seeded with 1  10 6 UC-MSCs) was
The cells were passaged until near confluence and cells were col- placed in each well using a procedure similar to the cell prolifera-
lected at passage 4–6 for subsequent experiments. tion assay. A PE Annexin V Apoptosis Detection Kit I (559763, BD
HESC phenotypes were confirmed by immunofluorescence Biosciences, USA) was used to measure HESC apoptosis after 48 h
analysis (Fig. S1C). Cells were fixed in 4% (v/v) paraformaldehyde co-culture according to the manufacturer’s instructions. In brief,
for 10 min, permeabilized with 0.2% (v/v) Triton X-100 for 100 mL of HESCs (1  106 cells/mL) were incubated with 5 mL of
10 min, blocked with 2% (w/v) bovine serum albumin (BSA, Gibco) phycoerythrin and 5 mL of 7-amino-actinomycin (7-AAD) for
for 1 h and stained with rabbit anti-vimentin (1:100, ab92547, 15 min in the dark. Three samples from each group were evaluated
Abcam, Cambridge, UK) or mouse anti-cytokeratin (1:100, in this assay (n = 3) and analyzed using a flow cytometer (BD
ab215838, Abcam, Cambridge, UK) antibodies diluted in 2% BSA LSRFortessa TM Cell Analyzer, USA).
overnight at 4 °C. Sections were incubated with Alexa Fluor 488-
conjugated goat anti-rabbit IgG (1:300, ab150077, Abcam, Cam- 2.6. Enzyme-linked immunosorbent assay (ELISA)
bridge, UK) and DyLight 549-conjugated goat anti-mouse IgG
(1:200, GAM5492, MultiSciences, Hangzhou, China) diluted in Vascular endothelial growth factor (VEGF-A), transforming
PBS for 1 h at room temperature. Nuclei were stained with 40 , 6- growth factor (TGF-b1), and platelet-derived growth factor
diamidino-2-phenylindole (DAPI) (20 lg/mL, Roche, Germany) (PDGF-BB) concentrations in the co-culture system were measured
and observed using a fluorescence microscope (Leica, Germany). by ELISA. The same trans-well co-culture system was used as for
the cell proliferation assay and culture medium was collected after
2.3. Fabrication of CS/UC-MSCs 24 h co-culture. The medium was centrifuged at 800 rpm for 5 min.
The supernatant was collected and analyzed using a VEGF-A ELISA
Collagen type I was obtained from bovine tendon by tryptic Kit (Elabscience, E-EL-H0111c, Wuhan, China), a TGF-b1 ELISA Kit
digestion and acid dissolution as previously described [37]. The CS (Elabscience, E-EL-H0110c, Wuhan, China) and a PDGF-BB ELISA
was fabricated as previously described [38,39]. Briefly, the collagen Kit (Elabscience, E-EL-H1577c, Wuhan, China) according to the
solution (0.5% w/v) was injected into a polytetrafluoroethylene manufacturer’s protocols. Concentrations of VEGF-A, TGF- b1, and
mold, frozen at 20 °C for 12 h and then lyophilized (Biocool, China) PDGF-BB were quantified and expressed in pg/mL. Four samples
for 24 h to obtain CSs. The CSs were treated by thermal dehydration from each group were assessed in this assay (n = 4).
crosslinking at 105 °C under vacuum (Jinghong, China) for 15 h. To
fabricate the CS/UC-MSCs, 50 lL of UC-MSCs (1  107 cells/mL) were 2.7. Rat model of endometrial damage and transplantation of CS/UC-
placed onto a sterilized CS segment (2.5 cm  0.5 cm) and further MSCs
cultured in DMEM/F12-10% FBS for 3 h.
Animal experiments were approved by the committee for ani-
2.4. Characterization of CS/UC-MSCs mal ethics, Zhejiang University, and were monitored by the ethics
committee of Sir Run Run Shaw Hospital, Zhejiang University.
The CS/UC-MSCs were fixed with 4% paraformaldehyde for 24 h, Female Sprague-Dawley (SD) rats (200–230 g, 7–9 weeks) were
dehydrated with graded ethanol, embedded in paraffin and sec- purchased from the Experimental Animal Center of Zhejiang Pro-
tioned at 5 lm thickness. Sections were stained with hematoxylin vince (China) and housed in a controlled environment at 22 °C with
and eosin (HE). The distribution of UC-MSCs within CSs after 12 h a 12 h/12 h light/dark cycle. The animals were allowed to acclima-
culture was observed under an optical microscope (Nikon Eclipse tize for 1 week prior to the experiments.
80i, Japan). The microstructures of CSs and UC-MSC morphology A rat model of endometrial damage was established as
within the scaffolds after 12 h were observed using scanning elec- described previously [12]. Briefly, all rats were anesthetized by
tron microscopy (SEM, Hitachi Model S-520, Japan). The SEM spec- intraperitoneal injection of 2% sodium pentobarbital
imens were fixed with 2.5% (v/v) glutaraldehyde for 24 h at 4 °C, (0.3 mL/100 g), and the uterine horns were exposed via an abdom-
post-fixed with 1% osmium tetroxide, dehydrated in graded etha- inal incision. A 3-cm longitudinal incision was made in the uterus
nol, dried under a critical point drier, and coated with gold. The to expose the inner endometrium. The endometrium was scraped
proliferative behavior of UC-MSCs in CSs and tissue culture using a T10 scalpel blade until the surface of the uterus was rough
polystyrenes (TCPSs) was evaluated using a cell counting kit-8 and bleeding. After washing with PBS (pH 7.4), a CS/UC-MSCs
(CCK-8) assay. Three samples from each group at different time (2.5 cm  0.5 cm) was transplanted onto the damaged endometrial
intervals were assessed in the CCK-8 assay (n = 3). surface using a plastic device as previous described [12]. The
uterus was stitched using 6–0 absorbable sutures.
2.5. Effect of CS/UC-MSCs on HESC proliferation and apoptosis in vitro
2.8. In vivo tracing of implanted CS/UC-MSCs
A transwell co-culture system (Corning, NY) was used in this
study. For the cell proliferation assay, HESCs (passage < 6) were To trace implanted CS/UC-MSCs, 18 rats (36 uterine horns)
seeded in a 24 well plate (1  105 cells/well) containing DMEM/ were used. At various time intervals (1, 3, 5, 7, 15 and 30 days
F12 and cultured for 2 h. A 0.4-lm pore insert containing a CS/ post-transplantation), 3 rats (6 uterine horns) were sacrificed and
L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171 163

uteri were harvested (n = 6). For HE staining, the uteri were fixed The percentage positive area in histological and immunohistologi-
with 4% paraformaldehyde overnight and sectioned at 5-lm thick- cal staining was measured using Image-J software (NIH, USA). Data
ness. For immunofluorescence tracing of UC-MSCs labeled with were presented as means ± standard deviation. Multiple compar-
CM-Dil (C7000, Molecular Probes, Invitrogen, Eugene, USA) isons were conducted using one-way analysis of variance (SPSS
[33,35,40,41], the samples were frozen in liquid nitrogen for 17.0). The 2 test was used for analyzing the results of fertility
cryo-sectioning (Leica, Germany). Cell nuclei were stained with tests. Statistical significance was assumed for p < 0.05 (* and **
DAPI (20 lg/mL, Roche, Germany), and observed using a fluores- indicate p < 0.05 and p < 0.01, respectively).
cence microscope (Leica, Germany).

2.9. Histological and immunohistological analysis 3. Results

72 rats (144 uterine horns) were assigned randomly to four 3.1. Characterization of CS/UC-MSCs
groups: (i) the sham group (sham, n = 18 uterine horns), (ii) the
natural repair group (NR, n = 36 uterine horns), (iii) the CS group After thawing, UC-MSCs exhibited a shuttle-like morphology.
(CS, n = 36 uterine horns), and (iv) the CS/UC-MSCs group (CS/ UC-MSCs were positive for CD73, CD90, and CD105, and negative
UC-MSCs, n = 54 uterine horns). Rats with damaged uteri trans- for CD34, CD45, CD19, CD11b, and HLA-DR. UC-MSCs were able
planted with a CS/UC-MSCs were referred to as the CS/UC-MSCs to differentiate into osteoblasts, adipocytes and chondrocytes
group. Rats transplanted with a CS or receiving no treatment were (Fig. S1A, B, D).
referred to as the CS group or the NR group, respectively. Rats with The macroscopic appearance and microscopic structure of CSs
intact uteri without excision after abdominal incision were with or without seeding with UC-MSCs were displayed in Fig. 1.
referred to as the sham group. Samples with a length of 0.5 cm Before seeding with UC-MSCs, the CS showed a sheet-like mor-
were harvested from uterine wound sites near the cervix at each phology (Fig. 1A). HE staining and SEM showed that the CS had a
time interval. The sections were stained with HE and Masson tri- porous structure with a pore size of 100–200 mm (Fig. 1B, C). There
chrome using conventional protocols. The thickness of the regener- were no obvious differences between the gross views of the CS and
ated endometrium was measured as the maximum vertical the CS/UC-MSCs after culture in complete medium (red color) for
distance from the luminal epithelium to the outer smooth muscle 12 h (Fig. 1D). As shown by HE staining, UC-MSCs attached well
including the uterine cavity or half of the maximum linear distance to the CS (Fig. 1E). A large number of cells with shuttle-like mor-
between the smooth muscle excluding the uterine cavity under a phology were observed on the CS/UC-MSCs surface by SEM
magnification of 40  . For immunohistochemistry, sections were (Fig. 1, F). The CCK-8 assay demonstrated that the UC-MSCs prolif-
deparaffinized and rinsed with water. Antigen retrieval was per- erated well on both CS and TCPS. Moreover, after 72 h incubation,
formed in 10 mM sodium citrate using a microwave. The slides cells on the CS showed higher proliferative activity than those on
were washed in PBS containing 0.1% Tween 20, blocked in 5% TCPS, which may be attributed to the three-dimensional structure
BSA for 1 h at room temperature, and incubated with primary anti- of the CS (Fig. 1, G) (n = 3).
bodies diluted in 5% BSA overnight at 4 °C. The primary antibodies
used were anti-Ki67 (1:200, ab15580, Abcam, Cambridge, UK),
anti-pan-cytokeratin (1:150, ab215838, Abcam, Cambridge, UK), 3.2. Paracrine effects of CS/UC-MSCs on HESCs
anti-estrogen receptor a (ERa) (1:200, ab79413, Abcam, Cam-
bridge, UK), anti-progesterone receptor (PR) (1:100, ab2765, The paracrine effects of CS/UC-MSCs on the proliferation and
Abcam, Cambridge, UK), anti-VEGF-A (1:200, ab46154, Abcam, apoptosis of HESCs were evaluated using a trans-well model. HESCs
Cambridge, UK), anti-TGFb1 (1:200, ab125287, Abcam, Cambridge, were positive for vimentin and negative for cytokeratin, and the
UK), anti-PDGF-BB antibodies (1:100, orb101747, Biorbyt, Cam- purity of HESCs was nearly 100% (Fig. S1, C). As shown in Fig. 1H,
bridge, UK). Endogenous peroxidase was blocked with 0.3% (v/v) the results of the CCK-8 assay demonstrated that the viabilities
H2O2 for 10 min. After washing in PBS containing 0.1% Tween 20, of HESCs cultured in different conditions increased with culture
the slides were reacted with horseradish peroxidase-conjugated time, indicating that all these cells could proliferate under the
goat anti-mouse/rabbit secondary antibody (GK500711, Genetech, three conditions tested. However, three different proliferation pro-
Shanghai, China) for 1 h at room temperature according to the files were observed. At days 1 and 2, the three groups showed sim-
manufacturer’s instructions. ilar CCK-8 values. After culturing for 3 days, the CCK-8 values of the
HESCs in the CS/UC-MSCs group (0.90 ± 0.04) were higher than
2.10. Fertility test those of the negative control group (0.60 ± 0.09) (p < 0.05, n = 3),
albeit still lower than those of the positive control group
A total of 60 rats (120 uterine horns) were used in the fertility (1.10 ± 0.04) (p < 0.05, n = 3). Analysis of HESC apoptosis under dif-
test. The right uterine horn of each rat was treated as described ferent culture conditions is shown in Fig. 1, I. Co-culture with CS/
above and assigned randomly to the CS/UC-MSCs group (n = 20 UC-MSCs decreased the rate of HESC apoptosis (31 ± 4%) after
uterine horns), the CS group (n = 20 uterine horns) or the NR group 48 h compared with the negative control group (62 ± 2%)
(n = 20 uterine horns). The left uterine horn of each rat was left (p < 0.01, n = 3) (Fig. 1, I). No significant difference was found
intact as the control group (Con) (n = 60 uterine horns). At day between the CS/UC-MSCs group and the positive control group.
60 post-treatment, the rats were mated with male SD rats for Higher levels of VEGF-A, TGF-b1, and PDGF-BB in the super-
4 days. The first day of mating was regarded as gestational day 0. natants of co-cultures from the CS/UC-MSCs group were detected
At day 18–20 after gestation, the uteri were exposed under anes- by ELISA than in those from the negative control group (Fig. 1J, K,
thesia to confirm the presence of embryos, and the embryos larger L). The concentration of VEGF-A in culture supernatants from the
than 1 cm in diameter were counted. CS/UC-MSCs group (440 ± 50 pg/mL) was higher than that in the
negative control group (15.2 ± 0.9 pg/mL) (p < 0.01, n = 4). The con-
2.11. Statistical analysis centration of TGF-b1 in culture supernatants from the CS/UC-MSCs
group (84 ± 4 pg/mL) was higher than that in the negative control
Two independent observers performed endometrial thickness group (66 ± 5 pg/mL) (p < 0.05, n = 4). The concentration of PDGF-
measurement, and the average was used for subsequent analysis. BB in culture supernatants from the CS/UC-MSCs group
164 L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171

Fig. 1. Characterization and paracrine effects of CS/UC-MSCs. (A, D) Macroscopic observation of CS (A) and CS/UC-MSCs (D). (B, E) HE staining of CS (B) and CS/UC-MSCs (E).
(C, F) SEM images of CS (C) and CS/UC-MSCs (F). (G) CCK-8 assay of UC-MSCs cultured on CSs (CS/UC-MSCs) and TCPS (UC-MSCs) for various time periods. (H) CCK-8 assay of
HESC proliferation in a CS/UC-MSCs co-culture system. HESCs cultured in DMEM/F12 with a blank insert and HESCs cultured in DMEM/F12 containing 10% FBS were used as
negative and positive controls, respectively. (I) Statistical analysis of the percentage of Annexin V and 7-AAD positive HESCs in the CS/UC-MSCs co-culture system measured
by flow cytometric analysis. (J, K, L) Statistical analysis of the concentrations of VEGF-A(J), TGF-b1(K), and PDGF-BB(L) in the CS/UC-MSCs co-culture system measured by
ELISA.

(36 ± 8 pg/mL) was higher than that in the negative control group (Fig. 2 B1 and B5). With longer time post-transplantation, the num-
(5.4 ± 0.4 pg/mL) (p < 0.05, n = 4). ber of CM-Dil-labeled UC-MSCs decreased (Fig. 2 B2–B4).

3.3. In vivo tracing of CS/UC-MSCs 3.4. Gross views of harvested uteri

As shown in Fig. S2, a rat model of endometrial damage was At day 15 post-surgery, hydrometra and structural deformation
established via a scraping treatment and demonstrated by histo- of the wounded sections were observed in the NR group (Fig. 3, B)
logical staining. The stromal layer of the damaged endometrium and the CS group (Fig. 3, C). However, the CS/UC-MSCs group
became thinner compared to a normal endometrium with a com- exhibited hydrometra only at the ends of uteri, as well as mild
plete luminal epithelial layer and stromal layer. Moreover, hyperemia and edema (Fig. 3, D). At 30 days post-transplantation,
immunohistochemical analysis showed that pan-cytokeratin posi- severe stenosis of the uterine cavity near the wounded section
tive epithelial cells were found on the normal endometrial luminal was observed in the NR group (Fig. 3, F) and the CS group (Fig. 3,
surface, but no positive cells were observed following scraping G). By contrast, in the CS/UC-MSCs group, hyperemia was observed
treatment (Fig. S2C–F). at the lesion site without uterine atrophy (Fig. 3, H). At 60 days
To track the transplanted CS/UC-MSCs in vivo, HE staining of post-transplantation, normal-seeming uteri were harvested in the
uteri was performed at each time interval (Fig. 2, A). At 1 and CS/UC-MSCs group (Fig. 3, L), similar to those of the sham group
3 days post-transplantation, CSs with porous structures could still (Fig. 3 A, E, I). However, the uteri of the CS group and the NR group
be observed in the wound region (Fig. 2 A1, A2). At 5 and 7 days showed severe uterine atrophy (Fig. 3, J, K).
post-transplantation, no obvious porous structure was observed,
probably due to the fusion of the CS with surrounding tissues 3.5. HE and Masson’s trichrome staining
(Fig. 2 A3, A4).
To track the transplanted UC-MSCs in the regenerated endome- Restoration of the endometrium following different treatments
trium, immunofluorescence of uteri at each time interval was ana- was assessed by HE staining. At 15 days post-surgery, the NR group
lyzed (Fig. 2, B). At 3 days post-transplantation, a number of CM- and the CS group exhibited endometrial regeneration without
Dil-labeled UC-MSCs were observed in the wounded endometrium luminal structures (Fig. 4 A2, A3). However, histological observa-
L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171 165

Fig. 2. Tracing of the implanted CS/UC-MSCs in vivo. (A) HE staining of uteri after CS/UC-MSCs transplantation for 1 day (A1), 3 days (A2), 5 days(A3) and 7 days (A4). (B)
Immunofluorescence of uteri after CS/UC-MSCs transplantation for 3 days (B1, B5), 7 days (B2), 15 days (B3) and 30 days (B4). UC-MSCs were labeled with CM-Dil.

Fig. 3. Morphology of uteri following different treatments for 15 days (A–D), 30 days (E–H) and 60 days (I–L). Treatments included the sham group (Sham) (A, E, I), the natural
repair group (NR) (B, F, J), the CS group (CS) (C, G, K) and the CS/UC-MSCs group (CS/UC-MSCs) (D, H, L).

tion of the CS/UC-MSCs group showed endometrial regeneration epithelium and secretory glands in the stromal layer of the CS/
with apparent luminal structures (Fig. 4, A4). The thickness of UC-MSCs group (Fig. 4, A8) By contrast, no luminal structures
the regenerated endometrium in the CS/UC-MSCs group and disordered glands were observed in the NR group and the CS
(710 ± 60 mm) was significantly greater than that in the NR group group (Fig. 4 A6, A7). The regenerated endometrium in the CS/
(560 ± 20 mm) (p < 0.01, n = 6) and the CS group (530 ± 10 mm) UC-MSCs group (620 ± 20 mm) was thicker than in the NR group
(p < 0.01, n = 6) (Fig. 4 A2, A3). At 30 days post-surgery, the struc- (420 ± 50 mm) (p < 0.01, n = 6) or the CS group (430 ± 30 mm)
ture of the endometrium appeared well organized with luminal (p < 0.01, n = 6) (Fig. 4, B). At 60 days post-transplantation, the
166 L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171

Fig. 4. Effects of different treatments on endometrial regeneration and collagen remodeling. (A) HE staining of uteri after different treatments for 15 days (A1–A4), 30 days
(A5–A8) and 60 days (A9–A12) in the sham group (sham) (A1, A5, A9), the natural repair group (NR) (A2, A6, A10), the CS group (CS) (A3, A7, A11) and the CS/UC-MSCs group
(CS/UC-MSCs) (A4, A8, A12). Inserts are the corresponding overview pictures with lower magnification, and the magnified regions are marked with black squares. (B)
Statistical analysis of endometrial thickness after different treatments for 15 days, 30 days and 60 days. (C) Collagen staining of uteri using Masson trichrome after different
treatments for 15 days (C1–C4), 30 days (C5–C8) and 60 days (C9–C12) in the sham group (sham) (C1, C5, C9), the natural repair group (NR) (C2, C6, C10), the CS group (CS)
(C3, C7, C11) and the CS/UC-MSCs group (CS/UC-MSCs) (C4, C8, C12). Inserts are the corresponding overview pictures with lower magnification, and the magnified regions are
marked with black squares. (D) Statistical analysis of the percentages of collagen positive staining after different treatments for 15 days, 30 days and 60 days.

CS/UC-MSCs group showed normal-seeming endometrium (Fig. 4, ing the menstrual phase [42]. Early rapid re-epithelialization is
A12). However, the NR group and the CS group exhibited severe critical for subsequent endometrial recovery after endometrial
IUAs (Fig. 4 A10, A11). The thickness of the endometrium in the damage. Ki67 and cyto-keratin are markers of cellular proliferation
CS/UC-MSCs group (610 ± 30 mm) was greater than that in the NR and epithelium recovery, respectively (Fig. 5).
group (170 ± 20 mm) (p < 0.01, n = 6) and the CS group The number of Ki67-positive cells increased in all groups as
(220 ± 20 mm) (p < 0.01, n = 6) (Fig. 4, B). time elapsed after surgery. However, at 15 days post-
To evaluate collagen remodeling in the reconstructed endome- transplantation, the CS/UC-MSCs group showed significant upregu-
trium after transplanting CS/UC-MSCs, Masson’s trichrome stain- lation in the percentage of Ki67 positive areas (15 ± 2%) (Fig. 5 A4,
ing was performed (Fig. 4, C) and areas of collagen staining were B) compared with the NR group (5.9 ± 0.8%) (p < 0.01, n = 6) (Fig. 5
analyzed quantitatively. At 15 days post-transplantation, the CS/ A2, B) and the CS group (6.7 ± 0.6%) (p < 0.01, n = 6) (Fig. 5 A3, B).
UC-MSCs group (43 ± 3%) showed lower collagen deposition than These proliferating cells were mainly located in the shallow layer
the NR group (70 ± 3%) (p < 0.01, n = 6) and the CS group of the endometrium (Fig. 5 A4, B). At day 30, a higher percentage
(62 ± 3%) (p < 0.01, n = 6) (Fig. 4 C2–C4, D). At 30 days post- of Ki67 positive areas were observed in the CS/UC-MSCs group
transplantation, collagen deposition was severe in the NR group (17 ± 2%) (Fig. 5 A8, B) compared with the NR group (9 ± 2%)
and the CS group. Lower collagen staining was detected in the (p < 0.01, n = 6) (Fig. 5 A6, B) and the CS group (11 ± 2%)
CS/UC-MSCs group (38 ± 5%) compared with the NR group (p < 0.01, n = 6) (Fig. 5 A7, B). At day 60, the percentage of Ki67 pos-
(73 ± 2%) (p < 0.01, n = 6) and the CS group (74 ± 2%) (p < 0.01, itive areas in the CS/UC-MSCs group (21 ± 2%) (Fig. 5 A12, B) was
n = 6) (Fig. 4 C6–C8, D), reflecting mild fibrosis of wounds treated similar to that in the Sham group (24 ± 2%) (p > 0.05, n = 6)
with CS/UC-MSCs. At 60 days post-transplantation, dense endome- (Fig. 5 A1, A5, A9, B) but was still higher than that in the NR group
trial fibrosis was confirmed in the NR group and CS group (Fig. 4 (9 ± 1%) (p < 0.01, n = 6) (Fig. 5 A10, B) and the CS group (11 ± 1%)
C10, C11). However, collagen deposition in the CS/UC-MSCs group (p < 0.01, n = 6) (Fig. 5 A11, B).
(41 ± 2%) was similar to that of the sham group (41 ± 3%) (p greater The number of pan-cytokeratin positive cells increased in the
than 0.05, n = 6), and much lower than that of the NR group CS/UC-MSCs group over time after surgery. However, the number
(75 ± 2%) (p < 0.01, n = 6) and the CS group (74 ± 2%) (p < 0.01, of pan-cytokeratin positive cells in the NR and CS groups remained
n = 6) (Fig. 4 C9–C12, D). low (Fig. 5 C, D). At 15 days post-transplantation, the CS/UC-MSCs
group (20 ± 1%) (Fig. 5 C4, D) showed obvious upregulation in the
3.6. Immunohistochemical staining of Ki67 and pan-cytokeratin percentage of pan-cytokeratin positive areas compared with the
NR group (4 ± 1%) (p < 0.01, n = 6) (Fig. 5 C2, D) and the CS group
Studies have shown that the initial repair of endometrial (2 ± 1%) (p < 0.01, n = 6) (Fig. 5 C3, D). At 30 days post-
epithelium is associated with shedding of endometrial tissue dur- transplantation, the percentage of pan-cytokeratin positive areas
L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171 167

Fig. 5. Effects of different treatments on cell proliferation and epithelial recovery. (A) Immunohistochemical staining of Ki67 for cell proliferation after different treatments
for 15 days (A1–A4), 30 days (A5–A8) and 60 days (A9–A12) in the sham group (sham) (A1, A5, A9), the natural repair group (NR) (A2, A6, A10), the CS group (CS) (A3, A7, A11)
and the CS/UC-MSCs group (CS/UC-MSCs) (A4, A8, A12). Inserts are the corresponding overview pictures with lower magnification, and the magnified regions are marked with
black squares. (B) Statistical analysis of the percentages of Ki67 positive areas after different treatments for 15 days, 30 days and 60 days. (C) Immunohistochemical staining
of pan-cytokeratin after different treatments for 15 days (C1–C4), 30 days (C5–C8) and 60 days (C9–C12) in the sham group (sham) (C1, C5, C9), the natural repair group (NR)
(C2, C6, C10), the CS group (CS) (C3, C7, C11) and the CS/UC-MSCs group (CS/UC-MSCs) (C4, C8, C12). Inserts are the corresponding overview pictures with lower
magnification, and the magnified regions are marked with black squares. (D) Statistical analysis of the percentages of pan-cytokeratin positive areas after different treatments
for 15 days, 30 days and 60 days.

in the CS/UC-MSCs group (24 ± 2%) (p < 0.01, n = 6) (Fig. 5 C8, D) areas compared with the NR group (5 ± 1%) (p < 0.01, n = 6) (Fig. 6
was higher than that in the NR group (4 ± 1%) (p < 0.01, n = 6) A10, B) and the CS group (4 ± 1%) (p < 0.01, n = 6) (Fig. 6 A11, B).
(Fig. 5 C6, D) and the CS group (4 ± 2%) (p < 0.01, n = 6) (Fig. 5 C7, The results of PR expression analysis were similar to those for
D). At day 60, the CS/UC-MSCs group (26 ± 1%) (Fig. 5 C12, D) ERa. PR positive cells were mainly located in the epithelium
had similar numbers of pan-cytokeratin positive areas as the SHAM (Fig. 6, C). At 15 days post-transplantation, the CS/UC-MSCs group
group (33 ± 2%) (p > 0.05, n = 6) (Fig. 5 C1, C5, C9, D), and both were showed upregulation in the percentage of PR positive areas
significantly higher than the NR group (5 ± 2%) (p < 0.01, n = 6) (12 ± 1%) (Fig. 6 C4, D) compared with the NR group (3.4 ± 0.6%)
(Fig. 5 C10, D) and the CS group (4 ± 1%) (p < 0.01, n = 6) (Fig. 5 (p < 0.01, n = 6) (Fig. 6 C2, D) and the CS group (3.1 ± 0.5%)
C11, D). (p < 0.01, n = 6) (Fig. 6 C3, D). At 30 days post-transplantation,
the percentage of PR positive areas in the CS/UC-MSCs group
3.7. Immunohistochemical staining of ERa and PR (17 ± 1%) (p < 0.01, n = 6) (Fig. 6 C8, D) was higher than in the NR
group (4.7 ± 0.7%) (p < 0.01, n = 6) (Fig. 6 C6, D) and the CS group
Following endometrial re-epithelialization, the functional layer (4.2 ± 0.6%) (p < 0.01, n = 6) (Fig. 6 C7, D). At day 60, the numbers
of endometrium increased in size and differentiated under the of PR-positive areas in the CS/UC-MSCs group (19 ± 2%) (Fig. 6
influence of circulating estrogen and progesterone. The expression C12, D) were similar to those of the Sham group (23 ± 3%)
of ERa and PRs is critical for endometrial regeneration. (p > 0.05, n = 6) (Fig. 6 C1, C5, C9, D), and both were significantly
As shown in Fig. 6, ERa positive cells were mainly located in the higher than the NR group (4.9 ± 0.7%) (p < 0.01, n = 6) (Fig. 6 C10,
epithelium (Fig. 6, A). At 15 days post-transplantation, the CS/UC- D) and the CS group (4.5 ± 0.5%) (p < 0.01, n = 6) (Fig. 6 C11, D).
MSCs group showed obvious upregulation in the percentage of
ERa positive areas (13 ± 2%) (Fig. 6 A4, B) compared with that in
the NR group (2.1 ± 0.4%) (p < 0.01, n = 6) (Fig. 6 A2, B) and the 3.8. Immunohistochemical staining of VEGF-A, TGF-b1, and PDGF-BB
CS group (2.7 ± 0.7%) (p < 0.01, n = 6) (Fig. 6 A3, B). At 30 days
post-transplantation, the percentage of ERa positive areas in the Growth factors participate in endometrial regeneration via
CS/UC-MSCs group (13 ± 1%) (p < 0.01, n = 6) (Fig. 6 A8, B) was autocrine and/or paracrine effects between endometrial stromal
higher than that in the NR group (2.4 ± 0.6%) (p < 0.01, n = 6) cells and epithelial cells. They are also involved in mediating the
(Fig. 6 A6, B) and the CS group (3.4 ± 0.6%) (p < 0.01, n = 6) (Fig. 6 effects of estrogen on proliferation and progesterone on differenti-
A7, B). At day 60, the CS/UC-MSCs group (15.3 ± 0.9%) (Fig. 6 A12, ation [43]. Immunohistochemical staining for VEGF-A, TGF-b1, and
B) showed a significant increase in the percentage of ERa positive PDGF-BB was performed at 15 days post-surgery (Fig. S3).
168 L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171

Fig. 6. Effects of different treatments on expression of ERa and PR. (A) Immunohistochemical staining of ERa after different treatments for 15 days (A1–A4), 30 days (A5–A8)
and 60 days (A9–A12) in the sham group (sham) (A1, A5, A9), the natural repair group (NR) (A2, A6, A10), the CS group (CS) (A3, A7, A11) and the CS/UC-MSCs group (CS/UC-
MSCs) (A4, A8, A12). Inserts are the corresponding overview pictures with lower magnification, and the magnified regions are marked with black squares. (B) Statistical
analysis of the percentages of ERa positive areas after different treatments for 15 days, 30 days and 60 days. (C) Immunohistochemical staining of PR after different
treatments for 15 days (C1–C4), 30 days (C5–C8) and 60 days (C9–C12) in the sham group (sham) (C1, C5, C9), the natural repair group (NR) (C2, C6, C10), the CS group (CS)
(C3, C7, C11) and the CS/UC-MSCs group (CS/UC-MSCs) (C4, C8, C12). Inserts are the corresponding overview pictures with lower magnification, and the magnified regions are
marked with black squares (D) Statistical analysis of the percentages of PR positive areas after different treatments for 15 days, 30 days and 60 days.

VEGF-A, TGF-b1, and PDGF-BB positive cells were mainly regenerate functional endometrium for the implantation of
located in the epithelium. The CS/UC-MSCs group showed obvious embryos and promote fertility restoration.
upregulation in the percentage of VEGF-A positive areas (20 ± 4%)
(Fig. S3 A4, B) compared with the NR group (10 ± 1%) (p < 0.05,
n = 6) (Fig. S3 A2, B) and the CS group (8 ± 2%) (p < 0.05, n = 6) 4. Discussion
(Fig. S3 A3, B). Similarly to VEGF-A, TGF-b1 expression in the CS/
UC-MSCs group (10 ± 1%) (Fig. S3 A8, C) was higher than in the In this study, we fabricated a CS/UC-MSCs and demonstrated
NR group (4 ± 2%) (p < 0.05, n = 6) (Fig. S3 A6, C) and the CS group that the scaffold facilitated endometrial regeneration and fertility
(3 ± 1%) (p < 0.05, n = 6) (Fig. S3 A7, C). Expression of PDGF-BB in restoration. Thicker endometrium, obvious luminal structures,
the CS/UC-MSCs group (14 ± 3%) (Fig. S3 A12, D) was also higher and better collagen remodeling were observed in the CS/UC-
than in the NR group (5 ± 1%) (p < 0.05, n = 6) (Fig. S3 A10, D) MSCs group. These findings could be attributed to the proliferation
and the CS group (5 ± 1%) (p < 0.05, n = 6) (Fig. S3 A11, D). of intrinsic endometrial cells and epithelial reconstruction.
Normally, IUAs are characterized by severe fibrosis and lack of
3.9. Fertility restoration normal endometrium. Hysteroscopy and subsequent intrauterine
physical barriers are the main conventional methods used clini-
At gestational day 18–20, pregnant uteri and the corresponding cally to treat IUAs. These intrauterine devices may prevent re-
embryos were collected and the results are displayed in Fig. 7. The adhesion to some degree. However, due to low biocompatibility
pregnancy rate and the rate of implantation of embryos with diam- and amenorrhea associated with compression of the endometrium,
eters larger than 1 cm were summarized in Table 1. In the control they may cause inflammatory reactions and be harmful for
group, the pregnancy rate and the embryo implantation rate was endometrial regeneration [44]. Therefore, a degradable and active
95.0% and 90.4%, respectively. In the NR group, no implanted scaffold is expected to overcome these limitations.
embryos were observed (Fig. 7, C). In the CS group, only one preg- In our study, we used a CS loaded with stem cells, which has
nancy was observed among 20 uteri, and only one embryo larger proven efficacy in tissue regeneration [29–31]. We fabricated the
than 1 cm was identified (Fig. 7 D, E). However, in the CS/UC- CS by freeze-drying and thermal treatment crosslinking. The scaf-
MSCs group, the pregnancy rate was 45.0% and the rate of embryo fold exhibited a porous structure, which was suitable for cell adhe-
implantation was 30.3% (Fig. 7 F, G and Table 1). The CS/UC-MSCs sion and nutrient or oxygen delivery. As UC-MSCs exhibit many
group exhibited improved fertility compared with the NR group advantages including their non-invasive collection, high prolifera-
and the CS group. These results implied that the CS/UC-MSCs could tion potential, and mutilineage differentiation, these cells have
L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171 169

Fig. 7. Effects of different treatments on fertility restoration. Gross views of pregnant uteri and corresponding embryos from the control group (Con) (A, B), the CS group (CS)
(D, E) and the CS UC-MSCs group (CS/UC-MSCs) (F, G). (C) Gross view of uteri from the natural repair group (NR).

Table 1
Reproductive outcomes over 60 days following different treatments.

Variable Control NR CS CS/UC-MSCs


Total number of uterine horns 60 20 20 20
Number (percentage) of pregnant uterine horns 57 (95.0%) 0 (0)a 1 (5.0%)b 9 (45.0%)
Total number of embryos 417 0 3 33
Number (percentage) of embryos larger than 1 cm 377 (90.4%) 0 (0)c 1 (33.3%)d 10 (30.3%)
a
p < 0.01 NR group versus CS/UC-MSCs group.
b
p < 0.01 CS group versus CS/UC-MSCs group.
c
p < 0.01 NR group versus CS/UC-MSCs group.
d
p > 0.05 CS group versus CS/UC-MSCs group.

been regarded as an ideal source for the fabrication of CS/stem cell epithelialization and vascularization [55]. TGFbs are suggested to
constructs [45–49]. We showed that the porous CS provided an induce proliferation and regulate immune responses in the endo-
ideal physical support for the attachment and proliferation of metrium [56,57]. PDGFs promote stromal cell proliferation via
UC-MSCs. autocrine effects at the proliferative stage [54,58].
In vitro, we established a co-culture system and showed that the In animal experiments, we developed an endometrium scraping
CS/UC-MSCs promoted HESC proliferation and inhibited apoptosis. model in rats and confirmed that scraping ultimately led to severe
Similarly, many studies have also demonstrated that UC-MSCs IUAs and infertility. This model was quite similar to the induction
inhibited apoptosis, promoted angiogenesis, and modulated of IUAs in humans, and this model may be useful in further
immune responses by secreting a variety of soluble factors, includ- research. By tracing the CS/UC-MSCs in vivo, no obvious porous
ing VEGF-A, insulin-like growth factor 1, TGF-b, and hepatocyte material was observed at 7 days post-transplantation. Similarly,
growth factor [50–53]. In the present study, higher levels of we observed that the intensity of labeled UC-MSCs in uteri was
VEGF-A, TGF-b1, and PDGF-BB were observed in the CS/UC-MSCs high during the first 7 days post-transplantation and then faded
group compared with the negative control group. From these over time. Thus, CS/UC-MSCs transplantation might create an
results, we concluded that the proliferation and decreased apopto- appropriate environment for tissue repair during the early stages
sis of HESCs might relate to paracrine effects of CS/UC-MSCs. It following endometrial damage.
should be pointed out that immune modulation of MSCs might We observed normal morphology, thicker endometrium and
be another important effect involved in endometrial regeneration better collagen remodeling after CS/UC-MSCs transplantation. By
and should be investigated further. We found in vivo that the site contrast, atrophied uteri, thin endometrium and severe collagen
adjacent to CS/UC-MSCs transplantation in uteri displayed higher deposition were observed in the NR and CS groups.
levels of VEGF-A, TGF-b1, and PDGF-BB compared with the NR Re-epithelialization of endometrium is critical for scar-free
group and the CS group. These growth factors have important roles repair with menstrual breakdown in humans. Epithelial repair
in endometrial regeneration [54]. VEGF is vital for endometrial re- may be associated with resurfacing of epithelial cells from glands
170 L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171

in the basal layer [59]. In our study, we demonstrated that the CS/ Acknowledgments
UC-MSCs increased cell proliferation in the endometrium by Ki67
immunostaining, similar to the research of bone mesenchymal The authors thank Boyalife Group Ltd. for help with flow cytometry
stem cells(BMSCs)’ promoting proliferation effect observed in mur- and multipotent differentiation analysis of UC-MSCs. This study
ine uterus after IUA [26]. These proliferating cells on the luminal was financially supported by the National Key Research Program
surface were positive for pan-cytokeratin, which indicated better of China (2018YFC1004800, 2017YFA0104900), Wu Jieping medi-
epithelium recovery following CS/UC-MSCs transplantation. cal fund (320.6755.15028), the National Natural Science Founda-
After re-epithelialization, ERa and PR expression is induced in tion of China (51673167), and the Key Research and
endometrial cells by rising estrogen and progesterone levels. As a Development Program of Zhejiang Province (2017C03022).
result, regeneration of endometrial mucosa and revascularization
occurs in the proliferative stage [60–62]. ERa and PR, as markers
Author disclosure statement
of endometrial receptivity, are critical targets for clinical IUA treat-
ment. Due to the lack of functional endometrium in IUAs, the
The authors have no conflicts of interest to declare.
effects of estrogen and progesterone supplement therapy are not
satisfactory. In this study, the CS/UC-MSCs remarkably upregulated
the expression of ERa and PR. Moreover, ERa and PR were mainly Appendix A. Supplementary data
expressed in the regenerated epithelium. These results indicated
that the regeneration of endometrial glands and luminal epithe- Supplementary data to this article can be found online at
lium played a vital role in recovery of endometrial function. Estro- https://doi.org/10.1016/j.actbio.2019.05.012.
gen or progesterone could bind to ERa or PR, which activated
downstream signal transduction and triggered the expression to References
maintain uterine function [63]. These results implicated that sup-
[1] A.B. Hooker, M. Lemmers, A.L. Thurkow, M.W. Heymans, B.C. Opmeer, H.A.
plementation with estrogen and progesterone following CS/UC- Brölmann, B.W. Mol, J.A. Hulme, Systematic review and meta - analysis of
MSCs transplantation might lead to better outcomes in clinical intrauterine adhesions after miscarriage: prevalence, risk factors and long -
treatment. term reproductive outcome, Hum. Reprod. Update 20 (2012) 262–278.
[2] E.A. Tonguc, T. Var, N. Yilmaz, S. Batioglu, Intrauterine device or estrogen
We showed that endometrial thickness and collagen deposition treatment after hysteroscopic uterine septum resection, Int. J. Gynecol. Obstet.
did not change significantly from day 15 to day 60 post- 109 (2010) 226–229.
transplantation, indicating that primary structural remodeling [3] D. Yu, Y.M. Wong, Y. Cheong, E. Xia, T.C. Li, Asherman syndrome–one century
later, Fertil. Steril. 89 (2008) 759–779.
was almost complete within 15 days. However, the gradually
[4] R. Deans, J. Abbott, Review of intrauterine adhesions, J. Minim Invas Gyn. 17
increased expression of Ki67, pan-cytokeratin, ERa, and PR as time (2010) 555–569.
elapsed post-transplantation probably indicated that the recovery [5] C.M. March, Management of Asherman’s syndrome Online, Reprod. Biomed. 23
of precise structures (such as the glandular epithelium of the endo- (2011) 87–92.
[6] M.M. Hanstede, E. van der Meij, L. Goedemans, M.H. Emanuel, Results of
metrium) and their functions may require longer time. Therefore, centralized Asherman surgery, 2003–2013, Fertil. Steril. 104 (2015) 1561–
we performed the fertility test at 60 days post-surgery. The preg- 1568.
nancy capacity is the gold standard to confirm the function of [7] S. Preutthipan, V. Linasmita, Reproductive outcome following hysteroscopic
lysis of intrauterine adhesions: a result of 65 cases at Ramathibodi Hospital, J.
regenerated endometrium. We found that pregnancy rates in the Pak. Med. Assoc. 83 (2000) 42–46.
CS/UC-MSCs group were higher than in the CS group and the NR [8] R. Szymański, P. Kamiński, L. Marianowski, Electroresectoscopy in submucous
group, indicating the functional recovery of regenerated endome- fibroids, intrauterine adhesions and uterine malformation treatment, Ginekol.
Pol. 71 (2000) 1031–1035.
trium after transplantation with CS/UC-MSCs. Early implantation [9] K.K. Roy, J. Baruah, J.B. Sharma, S. Kumar, G. Kachawa, N. Singh, Reproductive
sites can be evaluated using implantation-specific assay such as outcome following hysteroscopic adhesiolysis in patients with infertility due
Evans-blue staining on gestational day 4.5–5.5, but embryonic to Asherman’s syndrome, Arch. Gynecol. Obstet. 281 (2010) 355–361.
[10] A.B. Hooker, R. de Leeuw, P.M. van de Ven, E.A. Bakkum, A.L. Thurkow, N.E.A.
development is still unclear during pregnancy. In this study, Vogel, H.A.A.M. van Vliet, M.Y. Bongers, M.H. Emanuel, A.E.M. Verdonkschot, H.
embryos were observed on gestational day 18–20 and those larger A.M. Brölmann, J.A.F. Huirne, Prevalence of intrauterine adhesions after the
than 1 cm were deemed as well-developed embryos. Our results application of hyaluronic acid gel after dilatation and curettage in women with
at least one previous curettage: short-term outcomes of a multicenter,
indicated that the rate of implantation of embryos larger than
prospective randomized controlled trial, Fertil. Steril. 107 (2017) 1223–1231.
1 cm in the CS/UC-MSCs group was higher than in the NR group, [11] L. Gan, H. Duan, F.Q. Sun, Q. Xu, Y.Q. Tang, S. Wang, Efficacy of freeze-dried
which can be attributed to the better-organized epithelium and amnion graft following hysteroscopic adhesiolysis of severe intrauterine
improved responsiveness to sex hormones of the regenerated adhesions, Int. J. Gynaecol. Obstet. 137 (2017) 116–122.
[12] G. Kuramoto, S. Takagi, K. Ishitani, T. Shimizu, T. Okano, H. Matsui, Preventive
endometrium in the CS/UC-MSCs group. effect of oral mucosal epithelial cell sheets on intrauterine adhesions, Hum.
Reprod. 30 (2015) 406–416.
5. Conclusions [13] J. Zhao, Q. Zhang, Y. Wang, Y. Li, Rat bone marrow mesenchymal stem cells
improve regeneration of thin endometrium in rat, Fertil. Steril. 101 (2014)
587–604.
A CS/UC-MSCs was fabricated and applied for endometrial [14] Y. Chen, L. Liu, Y. Luo, M. Chen, Y. Huan, R. Fang, Effects of aspirin and
regeneration. In vitro studies showed that the CS/UC-MSCs pro- intrauterine balloon on endometrial repair and reproductive prognosis in
patients with severe intrauterine adhesion:a prospective cohort study, Biomed
moted HESC proliferation and inhibited apoptosis through para- Res. Int. 2017 (2017) 8526104.
crine effects. The transplanted CS/UC-MSCs maintained luminal [15] Y. Chi, P. He, L. Lei, Y. Lan, J. Hu, Y. Meng, L. Hu, Transdermal estrogen gel and
structures, and promoted endometrium regeneration and collagen oral aspirin combination therapy improves fertility prognosis via the
promotion of endometrila receptivity in moderate to severe intrauterine
remodeling in a rat model of endometrial damage. The regenerated adhesion, Mol. Med. Rep. 17 (2018) 6337–6344.
endometrium expressed ERa and PR and the ability of the regener- [16] X. Lin, Y. Zhang, Y. Pan, S. He, Y. Dai, B. Zhu, C. Wei, L. Xin, W. Xu, C. Xiang, S.
ated endometrium to receive embryos was confirmed. Together, Zhang, Endometrial stem cell-derived G-CSF attenuates endometrial fibrosis
via sonic hedgehog transcriptional activator Gli2, Biol. Reprod. 98 (2018) 480–
these results indicated that the CS/UC-MSCs could facilitate the
490.
reconstruction of endometrium structure and function. The topical [17] Y. Chen, Y. Chang, S. Yao, Role of angiogenesis in endometrial repair of patients
transplantation of CS/UC-MSCs after trans-cervical resection of with severe intrauterine adhesion, Int. J. Exp. Pathol. 6 (2013) 1343–1350.
adhesions offers a novel strategy to prevent re-adhesion, promote [18] H. Masuda, Y. Matsuzaki, E. Hiratsu, M. Ono, T. Nagashima, T. Kajitani, T. Arase,
H. Oda, H. Uchida, H. Asada, M. Ito, N. Yoshimura, T. Maruyama, H. Okano,
endometrium regeneration and improve pregnancy outcomes in Stem cell-like properties of the endometrial side population: implication in
the clinic. endometrial regeneration, PLoS One 5 (2010) e10387.
L. Xin et al. / Acta Biomaterialia 92 (2019) 160–171 171

[19] C.E. Gargett, H. Masuda, Adult stem cells in the endometrium, Mol. Hum. [41] A. Mirsaidi, K. Genelin, J.R. Vetsch, S. Stanger, F. Theiss, R.A. Lindtner, B. von
Reprod. 11 (2010) 818–834. Rechenberg, M. Blauth, R. Müller, G.A. Kuhn, S. Hofmann Boss, H.L. Ebner, P.J.
[20] C.E. Gargett, L. Ye, Endometrial reconstruction from stem cells, Fertil. Steril. 98 Richards, Therapeutic potential of adipose-derived stromal cells in age-related
(2012) 11–20. osteoporosis, Biomaterials 35 (2014) 7326–7335.
[21] D.C. Ding, Y.H. Chang, W.C. Shyu, S.Z. Lin, Human umbilical cord mesenchymal [42] P. Henriet, H.P. Gaide Chevronnay, E. Marbaix, The endocrine and paracrine
stem cells: a new era for stem cell therapy, Cell Transplant. 24 (2015) 339– control of menstruation, Mol. Cell. Endocrinol. 358 (2012) 197–207.
347. [43] L.C. Giudice, Growth factors and growth modulators in human uterine
[22] W. Chen, J. Liu, N. Manuchehrabadi, M.D. Weir, Z. Zhu, H.H. Xu, Umbilical cord endometrium: their potential relevance to reproductive medicine, Fertil.
and bone marrow mesenchymal stem cell seeding on macro-porous calcium Steril. 61 (1994) 1–17.
phosphate for bone regeneration in rat cranial defects, Biomaterials 34 (2013) [44] J. Eriksen, C. Kaestel, The incidence of uterine atresia after post-partum
9917–9925. curettage.A follow-up examination of 141 patients, Dan. Med. Bull. 7 (1960)
[23] X. Peng, H. Xu, Y. Zhou, B. Wang, Y. Yan, X. Zhang, M. Wang, S. Gao, W. Zhu, W. 50–51.
Xu, H. Qian, Human umbilical cord mesenchymal stem cells attenuate [45] D.L. Troyer, M.L. Weiss, Wharton’s jelly-derived cells are a primitive stromal
cisplatin-induced acute and chronic renal injury, Exp. Biol. Med. 238 (2013) cell population, Stem. Cells 26 (2008) 591–599.
960–970. [46] M.L. Weiss, C. Anderson, S. Medicetty, K.B. Seshareddy, R.J. Weiss, I.
[24] Z.Y. Guo, X. Sun, X.L. Xu, Q. Zhao, J. Peng, Y. Wang, Human umbilical cord VanderWerff, D. Troyer, K.R. McIntosh, Immune properties of human
mesenchymal stem cells promote peripheral nerve repair via paracrine umbilical cord Wharton’s jelly-derived cells, Stem. Cells 26 (2008) 2865–2874.
mechanisms, Neural. Regen. Res. 10 (2015) 651–658. [47] A. Can, S. Karahuseyinoglu, Concise review: human umbilical cord stroma with
[25] F. Alawadhi, H. Du, H. Cakmak, H.S. Taylor, Bone Marrow-derived stem cell regard to the source of fetus-derived stem cells, Stem. Cells 25 (2007) 2886–
(BMDSC) transplantation improves fertility in a murine model of Asherman’s 2895.
syndrome, PLoS One 9 (2014) e96662. [48] D. Baksh, R. Yao, R.S. Tuan, Comparison of proliferative and multilineage
[26] I. Cervello, C. Gil-Sanchis, X. Santamaria, S. Cabanillas, A. Diaz, A. Faus, A. differentiation potential of human mesenchymal stem cells derived from
Pellicer, C. Simon, Human CD133+ bone marrow-derived stem cells promote umbilical cord and bone marrow, Stem. Cells 25 (2007) 1384–1392.
endometrial proliferation in a murine model of Asherman syndrome, Fertil. [49] T. Li, M. Xia, Y. Gao, Y. Chen, Y. Xu, Human umbilical cord mesenchymal stem
Steril. 104 (2015) 1552–1560. cells: an overview of their potential in cell-based therapy, Expert. Opin.
[27] V. Serpooshan, M. Zhao, S.A. Metzler, K. Wei, P.B. Shah, A. Wang, M. Mahmoudi, Environ. Biol. 15 (2015) 1293–1306.
A.V. Malkovskiy, J. Rajadas, M.J. Butte, D. Bernstein, P. Ruiz-Lozano, The effect [50] P. Kuchroo, V. Dave, A. Vijayan, C. Viswanathan, D. Ghosh, Paracrine factors
of bioengineered acellular collagen patch on cardiac remodeling and secreted by umbilical cord-derived mesenchymal stem cells induce
ventricular function post myocardial infarction, Biomaterials 34 (2013) angiogenesis in vitro by a VEGF-independent pathway, Stem. Cells. Dev. 24
9048–9055. (2015) 437–450.
[28] F. Boccafoschi, J. Habermehl, S. Vesentini, D. Mantovani, Biological [51] C. Shen, P. Lie, T. Miao, M. Yu, Q. Lu, T. Feng, J. Li, T. Zu, X. Liu, H. Li, Conditioned
performances of collagen-based scaffolds for vascular tissue engineering, medium from umbilical cord mesenchymal stem cells induces migration and
Biomaterials 26 (2005) 7410–7417. angiogenesis, Mol. Med. Rep. 12 (2015) 20–30.
[29] Y.Y. Li, H.W. Cheng, K.M. Cheung, D. Chan, B.P. Chan, Mesenchymal stem cell- [52] A.I. Arno, S. Amini-Nik, P.H. Blit, M. Al-Shehab, C. Belo, E. Herer, C.H. Tien, M.G.
collagen microspheres for articular cartilage repair: cell density and Jeschke, Human Wharton’s jelly mesenchymal stem cells promote skin wound
differentiation status, Acta Biomater. 10 (2014) 1919–1929. healing through paracrine signaling, Stem Cell Res. Ther. 5 (2014) 28.
[30] Z. Liu, X. Yin, Q. Ye, W. He, M. Ge, X. Zhou, J. Hu, S. Zou, Periodontal [53] F. Shang, S. Liu, L. Ming, R. Tian, F. Jin, Y. Ding, Y. Zhang, H. Zhang, Z. Deng, Y.
regeneration with stem cells-seeded collagen-hydroxyapatite scaffold, J. Jin, Human umbilical cord MSCs as new cell sources for promoting periodontal
Biomater. Appl. 31 (2016) 121–131. regeneration in inflammatory periodontal defect, Theranostics 7 (2017) 4370–
[31] J. Guan, Z. Zhu, R.C. Zhao, Z. Xiao, C. Wu, Q. Han, L. Chen, W. Tong, J. Zhang, Q. 4382.
Han, J. Gao, M. Feng, X. Bao, J. Dai, R. Wang, Transplantation of human [54] C.E. Gargett, R.W. Chan, K.E. Schwab, Hormone and growth factor signaling in
mesenchymal stem cells loaded on collagen scaffolds for the treatment of endometrial renewal: role of stem/progenitor cells, Mol. Cell. Endocrinol. 288
traumatic brain injury in rats, Biomaterials 34 (2013) 5937–5946. (2008) 22–29.
[32] S.H. Lv, H.B. Wang, H. Liu, H.P. Wang, Q.X. Lin, D.X. Li, Y.X. Song, C.M. Duan, L.X. [55] X. Fan, S. Krieg, C.J. Kuo, S.J. Wiegand, M. Rabinovitch, M.L. Druzin, R.M.
Feng, C.Y. Wang, Reconstruction of engineered uterine tissues containing Brenner, L.C. Giudice, N.R. Nayak, VEGF blockade inhibits angiogenesis and re-
smooth muscle layer in collagen/matrigel scaffold in vitro, Tissue Eng. Part A epithelialization of endometrium, FASEB J. 22 (2008) 3571–3580.
15 (2009) 1611–1618. [56] C.O. Omwandho, L. Konrad, G. Halis, F. Oehmke, H.R. Tinneberg, Role of TGF-
[33] L. Ding, X. Li, H. Sun, J. Su, N. Lin, B. Peault, T. Song, J. Yang, J. Dai, Y. Hu, betas in normal human endometrium and endometriosis, Hum. Reprod. 25
Transplantation of bone marrow mesenchymal stem cells on collagen scaffolds (2010) 101–109.
for the functional regeneration of injured rat uterus, Biomaterials 35 (2014) [57] R.L. Jones, C. Stoikos, J.K. Findlay, L.A. Salamonsen, TGF-beta superfamily
4888–4900. expression and actions in the endometrium and placenta, Reproduction 132
[34] T. Song, X. Zhao, H. Sun, X. Li, N. Lin, L. Ding, J. Dai, Y. Hu, Regeneration of (2006) 217–232.
uterine horns in rats using collagen scaffolds loaded with human embryonic [58] N. Chegini, M.J. Rossi, B.J. Masterson, Platelet-derived growth factor (PDGF),
stem cell-derived endometrium-like cells, Tissue Eng. Part A 21 (2015) 353– epidermal growth factor (EGF), and EGF and PDGF beta-receptors in human
361. endometrial tissue: localization and in vitro action, Endocrinology 130 (1992)
[35] L. Xu, L. Ding, L. Wang, Y. Cao, H. Zhu, J. Lu, X. Li, T. Song, Y. Hu, J. Dai, Umbilical 2373–2385.
cord-derived mesenchymal stem cells on scaffolds facilitate collagen [59] W.C. Okulicz, R. Scarrell, Estrogen receptor a and progesterone receptor in the
degradation via upregulation of MMP-9 in rat uterine scars, Stem Cell Res. rhesus endometrium during the late secretory phase and menses, Proc. Soc.
Ther. 8 (2017) 84. Exp. Biol. Med. 218 (1998) 316–321.
[36] J. Li, Y. Dai, H. Zhu, Y. Jiang, S. Zhang, Endometriotic mesenchymal stem cells [60] A. Ferenczy, G. Bertrand, M.M. Gelfand, Proliferation kinetics of human
significantly promote fibrogenesis in ovarian endometrioma through the Wnt/ endometrium during the normal menstrual cycle, Am. J. Obstet. Gynecol.
b-catenin pathway by paracrine production of TGF-b1 and Wnt1, Hum. 133 (1979) 859–867.
Reprod. 31 (2016) 1224–1235. [61] H.A. Padykula, L.G. Coles, W.C. Okulicz, S.I. Rapaport, J.A. McCracken, N.W. King
[37] L. Ma, C.Y. Gao, Z.W. Mao, J. Zhou, J.C. Shen, X. Hu, C. Han, Collagen/chitosan Jr, C. Longcope, I.R. Kaiserman-Abramof, The basalis of the primate
porous scaffolds with improved biostability for skin tissue engineering, endometrium: a bifunctional germinal compartment, Biol. Reprod. 40 (1989)
Biomaterials 24 (2003) 4833–4841. 681–690.
[38] F.J. O’Brien, B.A. Harley, I.V. Yannas, L.J. Gibson, The effect of pore size on cell [62] R.M. Brenner, O.D. Slayden, W.H. Rodgers, H.O.D. Critchley, R. Carroll, X.J. Nie,
adhesion in collagen-GAG scaffolds, Biomaterials 26 (2005) 433–441. K. Mah, Immunocytochemical assessment of mitotic activity with an antibody
[39] I.V. Yannas, E. Lee, D.P. Orgill, E.M. Skrabut, G.F. Murphy, Synthesis and to phosphorylated histone H3 in the macaque and human endometrium, Hum.
characterization of a model extracellular matrix that induces partial Reprod. 18 (2003) 1185–1193.
regeneration of adult mammalian skin, Proc. Natl. Acad. Sci. U.S.A 86 (1989) [63] R.W. Su, M.R. Strug, J.W. Jeong, L. Miele, A.T. Fazleabas, Aberrant activation of
933–937. canonical Notch1 signaling in the mouse uterus decreases progesterone
[40] J. Lin, W. Zhang, J.J. Zhao, A.H. Kwart, C. Yang, D. Ma, X. Ren, Y.T. Tai, K.C. receptor by hypermethylation and leads to infertility, Proc. Natl. Acad. Sci. U.S.
Anderson, R.I. Handin, N.C. Munshi, A clinically relevant in vivo zebrafish A 113 (2016) 2300–2305.
model of human multiple myeloma to study preclinical therapeutic efficacy,
Blood 128 (2016) 249–252.

You might also like