You are on page 1of 15

3D-Printed Poly(-caprolactone) Scaffold

Augmented With Mesenchymal Stem Cells


for Total Meniscal Substitution
A 12- and 24-Week Animal Study in a Rabbit Model
Zheng-Zheng Zhang,* MD, PhD, Shao-Jie Wang,*y MD, PhD, Ji-Ying Zhang,* BS,
Wen-Bo Jiang,z PhD, Ai-Bing Huang,* MD, PhD, Yan-Song Qi,* MD, PhD,
Jian-Xun Ding,§ PhD, Xue-Si Chen,§ PhD, Dong Jiang,*|| MD, and Jia-Kuo Yu,*|| MD, PhD
Investigation performed at the Institute of Sports Medicine of Peking University Third Hospital,
Beijing, China

Background: Total meniscectomy leads to knee osteoarthritis in the long term. The poly(e-caprolactone) (PCL) scaffold is a prom-
ising material for meniscal tissue regeneration, but cell-free scaffolds result in relatively poor tissue regeneration and lead to joint
degeneration.
Hypothesis: A novel, 3-dimensional (3D)–printed PCL scaffold augmented with mesenchymal stem cells (MSCs) would offer ben-
efits in meniscal regeneration and cartilage protection.
Study Design: Controlled laboratory study.
Methods: PCL meniscal scaffolds were 3D printed and seeded with bone marrow–derived MSCs. Seventy-two New Zealand
White rabbits were included and were divided into 4 groups: cell-seeded scaffold, cell-free scaffold, sham operation, and total
meniscectomy alone. The regeneration of the implanted tissue and the degeneration of articular cartilage were assessed by gross
and microscopic (histological and scanning electron microscope) analysis at 12 and 24 weeks postoperatively. The mechanical
properties of implants were also evaluated (tensile and compressive testing).
Results: Compared with the cell-free group, the cell-seeded scaffold showed notably better gross appearance, with a shiny white
color and a smooth surface. Fibrochondrocytes with extracellular collagen type I, II, and III and proteoglycans were found in both
seeded and cell-free scaffold implants at 12 and 24 weeks, while the results were significantly better for the cell-seeded group at
week 24. Furthermore, the cell-seeded group presented notably lower cartilage degeneration in both femur and tibia compared
with the cell-free or meniscectomy group. Both the tensile and compressive properties of the implants in the cell-seeded group
were significantly increased compared with those of the cell-free group.
Conclusion: Seeding MSCs in the PCL scaffold increased its fibrocartilaginous tissue regeneration and mechanical strength, pro-
viding a functional replacement to protect articular cartilage from damage after total meniscectomy.
Clinical Relevance: The study suggests the potential of the novel 3D PCL scaffold augmented with MSCs as an alternative me-
niscal substitution, although this approach requires further improvement before being used in clinical practice.
Keywords: knee; meniscus; stem cell therapy; tissue engineering

Lesions in the meniscus are the most frequently recorded meniscectomy, but long-term results of such transplants
orthopaedic diagnosis.14 More than 1.5 million people across are unsatisfactory given poor tissue remodeling and are con-
the United States and Europe receive partial or total menis- troversial due to issues of compatibility and risk of disease
cectomy annually.37 However, the abnormal physiological transmission.18,19 Moreover, no total meniscal substitute
stress placed on articular cartilage postoperatively often has been approved by the Food and Drug Administration.
leads to knee osteoarthritis and related morbidity.15,26 Tissue engineering, which aims to regenerate damaged
Meniscal allograft transplant provides an alternative to tissue, offers a potential strategy for meniscal replacement.
Recently, numerous materials have been investigated as
scaffolds to construct meniscal implants. However, tissue-
based materials or extracellular matrix (ECM) compo-
The American Journal of Sports Medicine, Vol. 45, No. 7
DOI: 10.1177/0363546517691513 nents, such as tendon,23 small intestinal submucosa,8
Ó 2017 The Author(s) and collagen meniscal implants,43 have been shown to

1497
1498 Zhang et al The American Journal of Sports Medicine

possess unsatisfactory mechanical properties for in vivo Study Design


application.50 Synthetic polymers, such as polyurea,32,46
poly(e-caprolactone) (PCL),24,25,30 and polyglycolide,1 have Seventy-two skeletally mature, male New Zealand White
also been investigated for preparing meniscal scaffolds. rabbits weighing 3.0 kg were used for animal experiments.
However, polyurea is thought to potentially release carci- Forty-eight animals underwent total medial meniscectomy
nogenic compounds upon degradation.45 Polyglycolide scaf- of the left knees and were divided into 2 groups. Twenty-
fold with a porosity of 95% demonstrated successful four of the left knees received implantation by means of
fibrochondrocyte seeding but had insufficient mechanical PCL scaffolds seeded with MSCs (cell-seeded group), and
properties after 7 weeks of culture.1 PCL is a promising 24 others received cell-free scaffolds (cell-free group). As
material for meniscal tissue engineering given its superior a control, the remaining 24 rabbits underwent total medial
mechanics, bioactivity, and material processability.33 In meniscectomy of the right knees without implantation
addition, PCL loses its molecular weight in vivo consider- (Meni group). The left knees of the Meni group received
ably more slowly than do other aliphatic polyesters,27 sham operation involving exposure of the medial joint
and the scaffold could maintain initial mechanical support and closure in layers (sham group). Twelve rabbits were
until adequate tissue ingrowth occurs. randomly selected to be sacrificed at the 12-week and 24-
As previously reported, a 3-dimensional (3D) PCL scaf- week time points. At each time point, 6 knees from each
fold fabricated by fused deposition modeling (FDM) was group were collected for synovial fluid testing, followed
used to create tissue-engineered meniscus.51 Although by histological evaluation for implant regeneration and
cell-free scaffolds showed potential in fibrocartilaginous articular cartilage degeneration. Six other knees under-
tissue regeneration, joint degeneration occurred in the went biomechanical testing of implants and microscopic
early stage after implantation. Compared with strategies observation of cartilage surface by means of scanning elec-
that used seeding cells, cell-free augmentation resulted tron microscope (SEM).
in inadequate ECM secretion and insufficient mechanical
strength of implants, which might be the reason for joint Fabrication of 3D PCL Scaffolds
degeneration.21 Therefore, we determined that further ani-
mal studies were necessary to compare cell-seeded scaf- Five skeletally mature, male New Zealand White rabbit
folds with cell-free scaffolds in terms of fibrocartilaginous legs were scanned by magnetic resonance imaging (3.0-T
tissue regeneration, biomechanical properties, and carti- Signa HDxt; GE Healthcare) (512 3 512 pixels bitmap
lage protection for total meniscal replacement. image, 0.31-mm resolution). The magnetic resonance imag-
In this context, the 3D-printed PCL scaffolds were seeded ing data were imported into a Mimics software system
with mesenchymal stem cells (MSCs); these are seed cells (version 17.0; Materialise), and then a 3D reconstruction
used in meniscal tissue engineering that are promising for model of the meniscus was generated to calculate its size,
their significant healing capacity in a meniscal defect model, including the posterior horn width, anterior horn width,
but they have not been used for total meniscal replacement.16 anterior horn length, posterior horn length, posterior
Next, total meniscal substitution was performed in a rabbit horn to anterior horn distance, peripheral horn thickness,
model. The fibrocartilaginous formation (histology and immu- and peripheral horn width. For the rabbit medial meniscal
nohistochemistry), the mechanical strength, and the chondro- scaffold, a wedge-shaped arc disk was selected as a typical
protective effect of the implants were evaluated and compared model (Figure 1, A-C). As previously described regarding
with those of the cell-free scaffold and the native meniscus. the fabrication process, PCL (average molecular weight
Our hypothesis was that the 3D-printed PCL scaffold seeded 74,600 g mol-1 and melting point 52.9°C) (Changchun Sino-
with MSCs would offer some benefits in meniscal regenera- Biomaterials) was melted and extruded through a heated
tion, mechanical strength, and chondroprotection. The results metal nozzle, which was controlled by FDM software. For
of the present study might offer an alternative for total menis- the scaffold parameters, 215 6 23 mm pore size with
cal substitution, providing further insight into clinical treat- 300 mm road width (diameter of the printed fiber) and
ment for meniscal lesions. 200 mm space was determined to be optimal.51

Harvest, Culture, and Seeding of MSCs


METHODS
Bone marrow–derived MSCs were isolated from 3-month-
All experimental protocols involving animals were approved old New Zealand White rabbits. Isolation, cultivation,
by the local institutional animal care and use committee. and trilineage differentiation potential assays of MSCs

||
Address correspondence to Jia-Kuo Yu, MD, PhD, or Dong Jiang, MD, Institute of Sports Medicine, Peking University Third Hospital, No. 49 North
Garden Road, Haidian, Beijing, China 100191 (email: yujiakuo@126.com; bysyjiangdong@126.com).
*Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, P.R. China.
y
Department of Joint Surgery, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, P.R. China.
z
Clinical Translational R&D Center of 3D Printing Technology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine,
Shanghai, P.R. China.
§
Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P.R. China.
One or more of the authors has declared the following potential conflict of interest or source of funding: This study was supported by the National Nat-
ural Scientific Foundation of China (Grant Nos. 51273004, 31200725, 31670982, 81630056).
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1499

Figure 1. (A) Anatomic reconstruction model of rabbit menisci in left knee. (B) A typical model of 3D medial meniscal scaffold. (C)
3D-printed poly(e-caprolactone) (PCL) scaffold seeded with mesenchymal stem cells (scale bar represents 10 mm). (D) PCL scaf-
fold (black arrow) implanted between femur and tibia, with medial collateral ligament (green arrow) reserved.

were performed as previously described.13 MSCs in the tissue, and skin were closed with No. 3-0 Vicryl sutures (Ethi-
third passage were then seeded on the scaffold (5 3 106 con). For the Meni group, total resection of the medial menis-
cells per scaffold) by means of a centrifugal method.52 cus was performed only in the right knees; the left knees,
Briefly, the scaffold was placed at the bottom of a 1.5-mL serving as sham controls, underwent a procedure involving
centrifugal tube, and 100.0 mL of concentrated cell solution exposure of the medial joint and closure in layers. Animals
was added. The tube was centrifuged at 500 rpm for 1 min- were allowed unlimited movement after the operation. The
ute and then turned over as 1 loop. The process was antibiotic prophylaxis was continued for 3 days. At each
repeated 3 consecutive times. The seeded scaffolds were time point, the randomly selected animals were sacrificed
cultured under Dulbecco’s modified eagle medium (DMEM) via pentobarbital sodium.
for 24 hours before implantation.
Synovial Fluid Collection and Analysis
Surgical Procedure
At each time point, synovial fluid was collected via a 2-mL
Antibiotic prophylaxis was administered preoperatively by syringe with an 18-gauge needle, as previously reported.17
means of intramuscular penicillin (400,000 U). The The supernatants were assayed for interleukin 1 (IL-1) and
implantation procedure was performed as previously tumor necrosis factor a (TNF-a) by means of standard
reported.20 Briefly, after anesthesia and routine prepara- enzyme-linked immunosorbent assay (ELISA) kits (Rabbit
tion, the knee was approached through a medial parapatel- IL-1 ELISA Kit, I079SC; Rabbit TNF-a ELISA Kit, T103SC;
lar incision. A total meniscectomy was performed by Hermes Criterion Biotechnology).
resecting the medial meniscus sharply along the periphery
and detaching it from its anterior and posterior junction. Evaluation of Implants
Care was taken not to injure the medial collateral liga-
ment, which is important for the postoperative stability Half of the knees of the experimental group were excised,
of the knee joint (Figure 1D). Both the anterior and poste- and then the exposed femoral condyles and the tibial sur-
rior horns and periphery of the scaffolds were reattached to face with the implants in place were photographed. Three
the respective root attachments and appropriate adjacent researchers who were blinded to the experimental groups
synovium with absorbable No. 4-0 sutures (Ethicon). For evaluated the menisci by using the Gross Evaluation of
the posterior root, attachment of the medial meniscus is Meniscus Implant Score.24 This score includes 9 parame-
adjacent to the posterior cruciate ligament.6 We used ters: implant integration, implant position, horn position,
a self-made threading apparatus and extracapsular knot- shape, presence of tears in the implant, implant surface,
ting technique to fix the posterior horn of the scaffold with implant size, tissue quality, and condition of the synovia.
the ligamentous structures. The joint capsule, periarticular In this system, each parameter is scored from 1 to 3 based
1500 Zhang et al The American Journal of Sports Medicine

on the condition of the meniscal implants. The Gross Eval- midpoint of the medial femoral condyle, as described previ-
uation of Meniscus Implant Score is an evaluation system ously.20 The slides of the femur and tibia were stained with
for meniscal implants that is based on previous litera- H&E and TB and graded blindly according to the Mankin
ture.20,24,25,51 For scoring, each meniscal implant was cut grading system.34
at the level of the medial collateral ligament. The speci-
mens were cut to produce blocks exposing their wedge-
Biomechanical Analyses
shaped profile, which showed outer and inner regions
and superior and inferior surfaces of the regenerated The biomechanical properties of specimens were assessed by
menisci in histological sections. For each implant (total of means of a materials testing machine (AG-IS; Shimadzu).31
6 implants for each group), inner, intermediate, and outer The specimens were prepared as previously described.35,44
zones were evaluated. Blocks of meniscal implant were Briefly, in tensile testing, the samples (100 mm thick)
scored for the presence of residual scaffold, foreign body were cut into rectangular shapes from the peripheral edge
response, cellularity, blood vessel ingrowth, fibrosis, carti- of each meniscus, leaving a 3-mm gauge length in the
laginous matrix, integration of the implant with the joint approximate circumferential direction for testing. The sam-
capsule, and inflammatory cell infiltrate. The above fea- ples could not be cut into ‘‘dogbone’’ shapes owing to their
tures were scored as being either present or absent. Then small size. The sample was tested to failure at a rate of
the implant was dissected from the tibia and fixed in 0.06 mm/s. Multiple samples from each meniscus or implant
10% (vol/vol) neutral buffered formalin (Sigma Diagnos- were tested. The elastic modulus was analyzed from the lin-
tics). All samples were then dehydrated and embedded in ear portion of the stress-strain curve.10 Given the small size
paraffin. Next, 6-mm-thick cross sections were cut and of the medial rabbit meniscus, only the posterior portion
stained with hematoxylin and eosin (H&E) and toluidine could be cut for sampling in confined compression testing.
blue (TB, positive for proteoglycans). In addition, sections The samples were loaded into a custom-made jig.35 The
were treated by an immunohistochemistry procedure creep response of the specimen under the step force was
with labeling of collagen I and II (Calbiochem) and picro- monitored until equilibrium (defined as slope \1 3
sirius red (PR, staining to distinguish collagen I and III). 10-6 mm/s, at least 7200 s). Then the test force was automat-
The sections of implants were analyzed by blinded ically removed and the recovery phase began until equilib-
researchers according to the meniscal histology scoring rium (defined as slope \1 3 10-6 mm/s, at least 3600 s).
system.24 Moreover, immunohistochemical analyses were Overall, the creep indenter yielded the creep and recovery
used to conduct semi-quantitative study of the differences deformation of each specimen in response to a 0.02-N step
of collagen contents within the implants.53 Briefly, 10 dig- load.44 The aggregate modulus and permeability were calcu-
ital images were captured by an Olympus BX-51 micro- lated by using Mow’s biphasic theory.38
scope. Integrated optical density (IOD) value and area of
each microimage were measured with Image-Pro Plus 6.0
software (Media Cybernetics).47 The descriptors of relative Statistical Analyses
density (IOD per area) were measured to semi-quantify the All statistical data were expressed as mean 6 standard
deposition of collagen I and II. deviation (SD). Significance of the results was determined
with analysis of variance (ANOVA) or Mann-Whitney test
Evaluation of Joint Cartilages and repeated-measures tests with Bonferroni correction.
All data analyses were performed via SPSS statistical soft-
The cartilages of the femur and tibia were macroscopically ware (version 15.0; SPSS Inc). P \ .05 was considered sta-
evaluated according to the criteria of the International tistically significant, and P \ .01 and P \ .001 were
Cartilage Repair Society (ICRS) cartilage lesion classifica- considered highly significant.
tion.3 Three researchers who were blinded to the experi-
mental groups analyzed the cartilage of the medial
femoral condyle and the tibial plateau. The mean scores RESULTS
of the researchers were then calculated.
For microscopic observation of the cartilage surface, Gross Observation
specimens were assessed by JSM5600LV SEM (JEOL
USA). Briefly, samples were trimmed without disturbing All of the rabbits had regained their normal gait patterns
the cartilage surface, fixed immediately in 10 mL of 25% at 2 weeks postoperatively. No significant weight changes
glutaraldehyde for 1 day at 4°C, dehydrated in a graded or complications were seen. No signs of intra-articular
ethanol series, and finally subjected to critical point drying hemorrhage, swelling, or inflammation were seen in the
for complete dehydration. The surface of the cartilage was joints. The synovial fluid was clear postmortem.
coated with a 5-nm layer of gold and then viewed. At the 12-week time point, all the scaffolds formed
For histological evaluation, osteochondral specimens meniscal tissue in vivo (Figure 2). The cell-seeded scaffold
were fixed in 10% (vol/vol) neutral buffered formalin and became better integrated to the joint, with no sign of tears
decalcified in 10% ethylenediaminetetraacetic acid (Titri- and gap formation. Most of the implants maintained their
plex III; Merck). At 2 to 3 weeks, the osteochondral speci- original shape and size, while only 1 implant was slightly
mens were sectioned in the coronal plane at the midpoint smaller than the original construct. The new meniscal tis-
of the tibial plateau and in the sagittal plane at the sue formed with the cell-free scaffold showed significantly
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1501

were found between the 2 groups in total scores at week 24


(P 5 .013).
At each time point, the meniscus was intact in the sham
control group. Moreover, no sign of joint degradation was
observed. However, various degrees of cartilage damage
were observed in the other operated joints, especially in
the Meni group.

Histological Evaluation of Implants


The histological evaluation of implants (Table 2) revealed no
evidence of a significant inflammatory cell infiltration in the
implants. At week 12, a small number of lymphocytes were
found, mostly within new tissue, while foreign body reaction
(macrophages, foreign body giant cells) sometimes appeared
around the polymer scaffolds. The persistence of scaffold
residuals together with a consequent foreign body reaction
was present in both the cell-seeded and the cell-free implant
groups. Moreover, the foreign body reaction localized in
some areas of the implants, where hypocellularity was evi-
dent. Specially, the cell-seeded group showed a general
trend for a reduction of lymphocytes and foreign body giant
cells from 12 to 24 weeks with the absorption of the scaffold
and the new tissue regeneration. At each time point, all
implants showed extensive vascularization in the outer
regions of the new meniscal tissue.
The main differences observed between the cell-seeded
and cell-free groups were related to fibrosis and cartilage
metaplasia (Figure 3). At 12 weeks, new cartilaginous
matrix deposition in the tip region of the implant was
observed in 8 cases (5 cell-seeded and 3 cell-free), among
which 5 cases (3 cell-seeded and 2 cell-free) presented car-
tilaginous staining also at the central region of the new
meniscal tissue. At 24 weeks, there were more cases show-
ing cartilaginous matrix deposition in the cell-seeded
group, and most cases revealed staining at the tip and cen-
tral regions. Otherwise, cell phenotype appeared different
between groups. Interestingly, in the cell-seeded group,
a small number of round cells around the cartilage islands
presented in the inner region of the implant (Figure 3,
Aiii), which exhibited similar cell morphologic characteris-
tics based on the observation of native meniscus in the
sham group (Figure 3, Ciii). However, in the cell-free
Figure 2. Macroscopic observations of joints at 12 and 24 group, a large number of spindle-shaped cells with elon-
weeks after operation. Meniscal tissue excised from the tibial gated nuclei were surrounded by fibrous tissue in each
plateau is shown on the right. Scale bars represent 10 mm. region of the implants (Figure 3, Bi-Biii). At 24 weeks,
Meni, group that underwent total medial meniscectomy with- the various cell phenotypes within implants at different
out implantation of right knees. zones were also observed in the cell-seeded group. It was
apparent that outer zone cells had fusiform-shaped fibro-
blast-like cells, while cells in the inner portion appeared
less integration with the joint compared with the seeded as circular fibrochondrocytic shapes that were embedded
scaffold (P 5 .02; Table 1), although no significant difference in cartilage islands (Figure 3D), which was similar to the
was found for total score between these 2 groups (P 5 .108). native meniscus in the sham group (Figure 3F).
After 24 weeks, the gross appearance of all meniscal tis-
sues from the cell-seeded or the cell-free groups was nor- Immunohistochemical, PR, and TB
mal with a shiny white color and a smooth surface. No Evaluation of Implants
obvious synovial hypertrophy or implant tears were found
in either group. The new meniscal tissue formed with the Immunohistochemistry and PR staining were carried out to
cell-free scaffold had an inferior shape, especially in the explore the collagen component and type of the implants. In
anterior horn (P 5 .025). Moreover, significant differences PR staining, red and birefringent fibers represented collagen
1502 Zhang et al The American Journal of Sports Medicine

TABLE 1
Gross Evaluation of Meniscal Implant Scorea

P Valuesb

Parameter Cell-Seeded, 12 wk Cell-Free, 12 wk Cell-Seeded, 24 wk Cell-Free, 24 wk a b


c
Integration 2.0 (1-3) 1.2 (1-2) 2.2 (2-3) 1.5 (1-2) .020 .067
Implant position 2.0 (1-3) 1.7 (1-2) 2.3 (2-3) 1.8 (1-3) .654 .397
Horn position 2.2 (1-3) 1.7 (1-3) 2.5 (2-3) 2.0 (1-3) .403 .403
Shape 2.0 (1-3) 1.7 (1-2) 2.7 (2-3) 1.7 (1-2) .606 .025c
Tears 2.7 (2-3) 2.2 (2-3) 2.8 (2-3) 2.3 (2-3) .149 .149
Surface 1.3 (1-2) 1.5 (1-2) 2.3 (2-3) 1.7 (1-2) .830 .078
Size 1.7 (1-2) 1.5 (1-2) 2.2 (2-3) 1.7 (1-3) .864 .292
Tissue 2.2 (1-3) 1.8 (1-3) 2.3 (2-3) 1.8 (1-3) .746 .524
Synovia 2.3 (2-3) 2.0 (1-3) 2.3 (2-3) 2.0 (1-3) .685 .685
Total score 18.3 (14-21) 14.7 (11-19) 21.7 (20-23) 16.2 (11-22) .108 .013c

a
Each parameter is scored from 1 to 3 based on the condition of the meniscal implant. Scores are provided as mean (range).
b
a represents cell-seeded 12 wk versus cell-free 12 wk; b represents cell-seeded 24 wk versus cell-free 24 wk.
c
Statistically significant (P \ .05).

TABLE 2
Histological Features of Implantsa

Feature Cell-Seeded, 12 wk Cell-Free, 12 wk Cell-Seeded, 24 wk Cell-Free, 24 wk

Residual scaffold 6 (100) 6 (100) 6 (100) 6 (100)


Foreign body reaction 6 (100) 6 (100) 4 (66.7) 6 (100)
Hypocellular areas 4 (66.7) 6 (100) 4 (66.7) 5 (83.3)
Blood vessels 6 (100) 6 (100) 6 (100) 6 (100)
Fibrosis 3 (50) 6 (100) 1 (16.7) 5 (83.3)
Cartilage metaplasia
Tip 5 (83.3) 3 (50) 6 (66.7) 3 (50)
Central 3 (50) 2 (33.3) 5 (83.3) 2 (33.3)
Integration
Good 5 (83.3) 3 (50) 6 (100) 4 (66.7)
Poor 1 (16.7) 3 (50) 0 (0) 2 (33.3)
Inflammatory infiltrate
Lymphocytes 6 (100) 6 (100) 4 (66.7) 6 (100)
Plasma 0 0 0 0
Neutrophils 0 0 0 0

a
The features were scored as being either present or absent for each implant (total of 6 implants for each group). Data are provided as n
(%).

I, and green and thin fibers with weak birefringence repre- while the matrix increased at week 24 (Figure 6A). The value
sented collagen III. The proteoglycan content of the tissues of IOD per area was higher in the cell-seeded group than the
was detected by means of TB staining. As shown by PR stain- cell-free group at week 24 (P \ .001).
ing, the native menisci in the sham group showed strong As shown in Figure 5, the native menisci in the sham
staining for collagen I (Figure 4). In the cell-seeded group, control group demonstrated strong TB staining in the
the implants at week 12 formed a matrix with partial collagen inner and central regions, whereas the implants in the
I staining. At 24 weeks, the cell-seeded and sham groups cell-free scaffolds at weeks 12 and 24 showed only slight
showed large amounts of red fibers (collagen I), especially in proteoglycan staining. In the cell-seeded group, TB stain-
intermediate and outer regions, compared with the cell-free ing appeared stronger at week 24 than at week 12. At
group. Consistent with the PR staining, the cell-free group each time point, stronger collagen II staining was observed
revealed weaker staining than the other groups in immuno- in the cell-seeded group compared with the cell-free group,
histochemical images, which was also indicated by the rela- which is presented by IOD per area value (Figure 6B).
tive density of IOD per area value. The relative density of
IOD per area value was used for semi-quantitative analysis Evaluation of Joint Cartilage Degradation
of the collagen component in immunohistochemistry. The pro-
tein expression of collagen I in the seeded scaffold was lower According to the histopathological observation of articular
at week 12 compared with the native meniscus (P \ .001), cartilage and the scoring systems used (Figure 7), the
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1503

Figure 3. Macroscopic and histological images (general view and high-power magnification) of meniscus tissue. Gross views of
implants or native menisci are shown on the left. Insert boxes identify the region analyzed by histology. General views of outer,
intermediate, and inner zones of implants or native menisci (hematoxylin and eosin staining) are shown in the middle (scale bars 5
100 mm). High-power magnification images of meniscus tissue are shown on the right (scale bars 5 25 mm). Fibroblast-like cells
(black arrows) and chondrocyte-like cells (asterisks) were observed simultaneously in the implants of the cell-seeded group and
the sham group.

sham group showed the lowest grade of degenerative SEM was used to assess microscopic changes in the sur-
changes, with a mean value of 0.17 to 0.5 for the Mankin faces of articular cartilage (Figure 8). Articular surfaces of
score, and some specimens showed only cartilage surface blocks taken from the sham group did not differ signifi-
irregularities. In contrast, the femoral condyle and tibial cantly from normal cartilage.42 A relatively uniform area
plateau revealed complete disorganization of the cartilage without cracks was observed, and some of the residues
and severe reduction of TB staining in the total meniscec- noted may be attributable to synovial fluid crystals or
tomy group at 12 weeks. The highest mean Mankin and bone fragments. In contrast, in the Meni group, the femoral
ICRS scores at each time point were found in the Meni condyle and tibial plateau showed deep cracks and lacunae
group, and the cartilage damage progressed with time. on the surface enmeshed within frayed fibrils. The cell-free
The cell-free scaffold groups revealed diffuse chondrocyte scaffold groups also revealed superficial cracks and large
cloning and clefts to the transitional or radial zone with amounts of uniformly fibrous surfaces. In the cell-seeded
slight reduction of TB staining, which indicated less severe group, the articular cartilage surface appeared disrupted,
cartilage degradation than that of the Meni group. How- with a hillocky appearance, at 12 weeks. At 24 weeks, few
ever, the degree of cartilage damage increased with time. superficial cracks were noted in the cartilage surface.
Compared with the Meni and cell-free groups, the cell- To quantify inflammatory factors, synovial fluid was
seeded group fared significantly better based on ICRS collected to analyze IL-1 and TNF-a (Figure 9). Similar
and Mankin scores. Surface irregularities with diffuse to the tendency reported above, the greatest number of
hypercellularity were displayed. Moreover, no significant cytokines was observed in the Meni group at 12 weeks.
changes appeared at each time point. Although IL-1 and TNF-a levels in the cell-seeded group
1504 Zhang et al The American Journal of Sports Medicine

Figure 4. Representative picrosirius red (PR) and immunohistochemical staining for collagen I (Col I) of regenerated and native
menisci. Crassi red fiber represents Col I in PR-stained images. Scale bars represent 50 mm.

were identical to those of the sham group at 12 weeks, larger than that of the cell-free group at week 24 (P 5
these inflammatory factors increased progressively with .0423).
time. At 24 weeks postoperatively, the amount of either Confined compressive creep showed that the implants in
cytokine in the cell-seeded group was greater than that both the cell-seeded and the cell-free groups were 20% to
in the sham group while lower than that in the cell-free 40% as stiff in compression as the native meniscus (Figure
or Meni group. 10B). Moreover, the value of aggregate modulus in the
cell-seeded group was higher than that of the cell-free group
at 24 weeks (P \ .0001). The permeability of the scaffolds
Evaluation of Biomechanical Properties of Implant was significantly reduced after implantation (Figure 10D).
At 12 weeks, there were no differences between groups.
At 12 and 24 weeks, the biomechanical properties of native
meniscus were compared with those of the sham group,
and no differences were found. In tensile testing, the ten- DISCUSSION
sile modulus of the native meniscus was significantly
higher than that of the cell-seeded or cell-free group at The purpose of this study was to investigate the fibrocarti-
each time point (Figure 10A). In addition, the value of laginous regeneration, mechanical properties, and carti-
the tensile modulus in the cell-seeded group was higher lage protection capacity of a novel 3D-printed PCL
than that of the cell-free group at 24 weeks (P 5 .019). scaffold augmented with MSCs after implantation as
The ultimate tensile strength of the implants in the cell- a full meniscal substitution in a rabbit model. To our
seeded and cell-free groups at weeks 12 and 24 was approx- knowledge, this is the first study to examine the possibility
imately half that of the native medial menisci (Figure of regenerating the whole meniscus in a total meniscec-
10C). The ultimate strength in the cell-seeded group was tomy animal model by implanting MSCs-seeded scaffolds
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1505

Figure 5. Representative toluidine blue (TB) and immunohistochemical staining for collagen II (Col II) of regenerated and native
menisci. Scale bars represent 50 mm.

Figure 6. Immunohistochemical analyses of native meniscus and implants. Values for integrated optical density (IOD) per area of
(A) collagen I (Col I) and (B) collagen II (Col II) were larger in the cell-seeded group compared with the cell-free group, similar to the
native meniscus at 24 weeks (***P \ .001).

directly into the knee. Throughout 24 weeks, significantly In this study, a PCL scaffold fabricated by FDM was
better tissue formation, lower cartilage degeneration, and used to create tissue-engineered meniscus. Compared
retained mechanical strength were found in the cell-seeded with traditional processes such as particle/salt leaching11
scaffolds. The results indicated that the 3D-printed PCL and electrospinning,2 FDM is a rapid prototyping tech-
scaffold seeded with MSCs might be an alternative for nique that can produce a 3D scaffold with complete control
meniscal replacement. of geometric parameters such as pore size, porosity, and
1506 Zhang et al The American Journal of Sports Medicine

Figure 7. (A) Hematoxylin and eosin (H&E) and toluidine blue (TB) staining of articular cartilage surfaces in the femoral condyle
and tibial plateau (scale bars 5 100 mm). As indicated by (B) International Cartilage Repair Society (ICRS) and (C) Mankin scores,
the cell-seeded group presented lower cartilage degeneration in both the femur and tibia compared with cell-free group or Meni
group (*P \ .05, **P \ .01, ***P \ .001).
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1507

Figure 8. Scanning electron microscope (SEM) images of articular cartilage surfaces in femoral condyle and tibial plateau (scale
bars 5 5 mm).

Figure 9. Synovial fluid assessment. Levels of (A) interleukin 1 (IL-1) and (B) tumor necrosis factor a (TNF-a) in synovial fluid were
lower in the cell-seeded group compared with cell-free group or Meni group (*P \ .05, **P \ .01, ***P \ .001).

pore interconnection size.28,48,49 Moreover, PCL is a prom- or less degradation-resistant foreign macromolecular
ising material because of its superior bioactivity and material appeared as previously reported.36,41 We consider
mechanical characteristics.50 At week 24, better fibrocarti- that this low degree of lymphocyte infiltration combined
laginous formation coupled with lower foreign body reac- with foreign body reaction is not significant; a significant
tion was present in the cell-seeded group. These results response would include a large amount of neutrophil and
suggest the feasibility of this scaffold material as well as eosinophil infiltration and even biomaterial encapsulation
the potential use of MSC augmentation in tissue engineer- and failure. In addition, the degradation profiles of PCL
ing. MSCs have the ability to differentiate into chondro- extended to 12 months in another study25; thus, the scaf-
cytes, adipocytes, and osteoblasts, and they have immune fold can sustain loading until adequate tissue ingrowth
regulatory capacity. Although residual materials consist- occurs with time.
ing mainly of polymeric components caused foreign body The characterization of meniscus cells appears inconsistent
reaction, most of the implants showed new vessel ingrowth in the literature, and various terms are used (ie, fibrochondro-
and excellent integration with surrounding tissues. In the cytes, meniscus cells, fibrocytes, fibroblasts, chondrocytes).39
present study, the response of the host tissue against more Regardless of the term used, it is apparent that outer zone
1508 Zhang et al The American Journal of Sports Medicine

Figure 10. Biomechanical properties. (A) Tensile modulus and (C) ultimate tensile strength of the scaffold (time 0) and implants
compared with the native medial meniscus. (B) Compressive aggregate modulus and (D) permeability of the scaffold (time 0) and
implants compared with the native posterior portion of medial meniscus (*P \ .05, ***P \ .001; n 5 6 per group).

cells have a fusiform shape and are similar to fibroblasts. Several researchers have reported the development of
These fibroblast-like cells are surrounded with collagen I tissue-engineered meniscus using a biodegradable scaffold
(80% composition by dry weight).4 In contrast, round seeded with native meniscus cells or chondrocytes.5,11,21
chondrocyte-like cells in the inner region are embedded in We believe that autologous cells will be safe for clinical
an ECM composed largely of collagen II and glycosamino- treatment. However, the protocol of using autologous cells
glycans. In previous research regarding tissue-engineered exhibited several limitations. Two surgical interventions
meniscus, Kon et al25 reported that a polycaprolactone/hya- would be required. Moreover, given the scarcity and dedif-
luronic acid scaffold seeded with autologous chondrocytes ferentiation of autologous cells after culture and expansion,
was implanted into a sheep model. After 12 months, the his- as well as the possibility that autologous cells are in either
tological image of the implant showed cartilage metaplasia an age-related disease state or a degenerated state, their
with spherical and chondroid cells, while large amounts of application in tissue engineering is limited.9 The present
fibroblast-like cells and fibrous tissue deposition were pre- study is a primary exploration of the potential of allogenic
sented in the cell-free scaffold group.25 Other investigators MSCs for meniscal tissue engineering. No reports are avail-
also reported the presence of collagen I and II in implanted able describing measurable immune response when allo-
meniscal scaffolds.21,22,36,41 In our study, the various cell genic or xenogeneic cell sources are used, so for further
phenotypes within implants at different zones were clinical translation, a 1-step strategy using a bioactive factor
observed in the cell-seeded group at 12 weeks. Meanwhile, for endogenous cell (ie, stem cell, progenitor cell) homing
ECM deposition, with histological evidence of collagen I will be investigated to overcome the limitations of allogenic
and II, occurred at a higher level in the cell-seeded group cell transplantation.30 As previously reported, MSCs can
compared with the cell-free group. In particular, the escape immune recognition and exhibit an immune-
fibroblast-like cells around the scaffold in the outer two- tolerance capacity.12,29 Moreover, the allogenic MSC aug-
thirds appeared elongated, suggesting that tensile strength mentation approach for meniscal defect or cartilage repair
oriented circumferentially and radially might stimulate has proven successful.13,16 In the present study, although
MSCs to differentiate into fibroblasts and produce collagen we noted no allogenic response of New Zealand White rab-
I.7 The rounded shape of cells in the inner margin implied bits, we observed no obvious immunological rejection or sig-
a response to compression, and research has demonstrated nificant inflammatory reactions in histological and synovial
that compressive loading induces MSC chondrogenesis.40 fluid assessment. Therefore, the present study using allo-
Although Patel et al41 also showed various distributions of genic MSCs is a preliminary examination of a model that
cellular phenotypes and collagens in a 1-year in vivo exper- requires further study. In regard to clinical application,
iment using an cell-free scaffold, a similar result was the presence and outcome of immune reaction should be
observed in the current study at 24 weeks in the cell-seeded investigated. In the present study, we observed not only
group. These data confirm the benefit of seeding cells in increased quantity but also improved quality of the regener-
order to produce meniscal regeneration. ated fibrocartilaginous tissue in knees implanted with MSC-
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1509

seeded scaffolds. Although these cell-free scaffolds showed meniscal tissue could not completely simulate the biochem-
an IOD per area value of collagen I similar to that of the ical and biomechanical properties of the native meniscus or
cell-seeded group at 12 weeks (P 5 .063, Figure 6A), the fully prevent cartilage degeneration. We acknowledge that
inferior cartilage metaplasia features of implants seen in sham and total meniscectomy in the same rabbit might
Table 2 illustrated that these scaffolds ultimately filled have little effect on the results due to weight distribution
with fibrous tissue as opposed to meniscus fibrocartilage. after surgery. However, the sham operation simply
In contrast, scaffolds treated with MSCs regenerated involved exposure of the medial joint and closure in layers,
with well-bonded tissue resembling the native meniscus in which had no effect on the menisci and articular cartilage.
matrix composition and cellularity, suggesting the potential As shown by the histological and SEM images of cartilage
of MSCs in constructing a functional tissue-engineered and meniscus, the degeneration of cartilage in the Meni
meniscus. group increased with time and few signs of joint degrada-
As tissue-engineered meniscus plays a key role in joint tion were observed in the sham control. Therefore, we con-
load distribution, similar to the native meniscus, the sidered that the bilateral surgery had little effect on the
implant should become resistive to tensile and compressive results.
loads in vivo. Unfortunately, few reported scaffolds have The major limitation of the present study is that we did
strength comparable to that of the native meniscus.30 As not follow the outcome of the implanted MSCs within the
anticipated in the current study, ultimate tensile strength knee joint. Although the distribution, migration, and out-
and tensile modulus were reduced after implantation, due come of these transplanted cells are still unclear due to
to polymer fiber degradation. However, from 12 to 24 the limited tracking methods, green fluorescent protein
weeks, tensile properties did not change significantly, or other markers might be needed to label MSCs for track-
which suggested that regenerated tissue took over the ing progeny after implantation.16 However, the labeling
load that the scaffold could no longer bear in its degrading time was limited and the seeded MSCs would eventually
state. Confined compression testing revealed a significant proliferate and differentiate into chondrocytes, fibrochon-
decrease in permeability of the scaffolds in both the cell- drocytes, or other phenotypes, which might increase the
seeded and the cell-free groups from time zero to 24 weeks difficulty of cell labeling and tracing. Other limitations of
after implantation. This decreased permeability may have translational assumptions in this study include those
resulted from tissue infiltration into the scaffold. In addi- that are inherent to the animal model. Rabbit knees may
tion, the value of aggregate modulus in implants was lower have a weightbearing pattern that differs from that of
than that of native meniscus, although the implants in the human knees, for the rabbit is a mostly sedentary animal
cell-seeded group were significantly stiffer than unseeded whose knees are flexed more than 90°. On the basis of
implants at week 24. In the cell-seeded group, the compres- these results, a longer implantation time will be observed
sive value at week 24 was double that of time-zero scaf- in another model (ie, sheep, goat, or primate). Finally, after
folds, which might be due to the tissue deposition and the operation, animals were allowed unlimited movement,
organization. To our knowledge, no tissue-engineered which was not representative of the restrictions on move-
meniscus has achieved greater compressive moduli than ment and loading after procedures in humans. In light of
native meniscus, and few in vivo studies have reported this, caution should be used when extrapolating these
these values in a confined creep model.25 Further research results to clinical treatment.
is needed to increase the initial tensile and compressive
stiffness of the scaffold and deposition of ECM.
Although the chondroprotective effect of the cell-seeded CONCLUSION
scaffold was demonstrated based on ICRS and Mankin
scores, cartilage degeneration was not completely pre- In this study, we demonstrated that a novel 3D-printed PCL
vented. Significant differences were found between the scaffold augmented with MSCs acted as a functional menis-
cell-seeded group and the sham group at each time point cal replacement and had a chondroprotective effect after 24
in each scoring system. This could be attributed to several weeks of implantation. The cell-seeded scaffold showed
reasons. One concern was the biochemical content of the superiority in tissue regeneration, biomechanical strength,
implant. Native meniscus is an anisotropic fibrocartilagi- and chondroprotection. The present study is also a primary
nous tissue, with various regions composed of different exploration of regenerating menisci with MSCs, which is
types of collagen resistant to diverse loading.33 The trans- rarely reported but might be promising for meniscal tissue
planted tissue in our study mainly contained collagen I and engineering. In future, a 3D-printed, human meniscus–
II, but the distribution and deposition of these collagens shaped biomaterial scaffold will be investigated if tissue
did not provide the tissue with normal biological capacity. organization and mechanics are comparable with the full
One possible reason is that the distribution of new tissue size of the human meniscus. Moreover, further research
could have been affected by extrusion and/or thinning. will focus on a delivery approach for a human clinical trial
Further study is needed to create meniscal constructs in which bioactive polypeptides or drugs are packaged in
with appropriate fixation to replicate the biochemical prop- a good laboratory practice (GLP)/good manufacturing prac-
erties of native tissue. As well, the scaffold degradation tice (GMP) facility. Thus, rather than transplant an allo-
and tissue ingrowth played key roles in protection of carti- graft meniscus, surgeons could implant a ready-made
lage, as previously described. Therefore, the cell-seeded meniscal product using the existing arthroscopic procedure.
1510 Zhang et al The American Journal of Sports Medicine

REFERENCES a preliminary study on the role of pore sizes in ingrowth and differen-
tiation of fibrocartilage. Clin Mater. 1993;14(1):1-11.
1. Aufderheide AC, Athanasiou KA. Comparison of scaffolds and culture 23. Kohn D, Wirth CJ, Reiss G, et al. Medial meniscus replacement by
conditions for tissue engineering of the knee meniscus. Tissue Eng. a tendon autograft: experiments in sheep. J Bone Joint Surg Br.
2005;11(7-8):1095-1104. 1992;74(6):910-917.
2. Baker BM, Mauck RL. The effect of nanofiber alignment on the matura- 24. Kon E, Chiari C, Marcacci M, et al. Tissue engineering for total menis-
tion of engineered meniscus constructs. Biomaterials. 2007;28(11):1967- cal substitution: animal study in sheep model. Tissue Eng Part A.
1977. 2008;14(6):1067-1080.
3. Brittberg M, Winalski CS. Evaluation of cartilage injuries and repair. 25. Kon E, Filardo G, Tschon M, et al. Tissue engineering for total menis-
J Bone Joint Surg Am. 2003;85(suppl 2):58-69. cal substitution: animal study in sheep model—results at 12 months.
4. Cheung HS. Distribution of type I, II, III and V in the pepsin solubilized Tissue Eng Part A. 2012;18(15-16):1573-1582.
collagens in bovine menisci. Connect Tissue Res. 1987;16(4):343- 26. Krause W, Pope M, Johnson R, Wilder D. Mechanical changes in the
356. knee after meniscectomy. J Bone Joint Surg. 1976;58(5):599-604.
5. Chiari C, Koller U, Dorotka R, et al. A tissue engineering approach to 27. Kweon H, Yoo MK, Park IK, et al. A novel degradable polycaprolactone
meniscus regeneration in a sheep model. Osteoarthritis Cartilage. networks for tissue engineering. Biomaterials. 2003;24(5):801-808.
2006;14(10):1056-1065. 28. Lantada AD, Morgado PL. Rapid prototyping for biomedical engi-
6. Civitarese D, Donahue TL, LaPrade CM, et al. Qualitative and quan- neering: current capabilities and challenges. Annu Rev Biomed
titative measurement of the anterior and posterior meniscal root Eng. 2012;14:73-96.
attachments of the New Zealand white rabbit. J Exp Orthop. 29. Le Blanc K, Rasmusson I, Sundberg B, et al. Treatment of severe
2016;3(1):10. acute graft-versus-host disease with third party haploidentical mes-
7. Connelly JT, Vanderploeg EJ, Mouw JK, Wilson CG, Levenston ME. enchymal stem cells. Lancet. 2004;363(9419):1439-1441.
Tensile loading modulates bone marrow stromal cell differentiation 30. Lee CH, Rodeo SA, Fortier LA, Lu C, Erisken C, Mao JJ. Protein-
and the development of engineered fibrocartilage constructs. Tissue releasing polymeric scaffolds induce fibrochondrocytic differentiation
Eng Part A. 2010;16(6):1913-1923. of endogenous cells for knee meniscus regeneration in sheep. Sci
8. Cook JL, Tomlinson JL, Kreeger JM, Cook CR. Induction of meniscal Transl Med. 2014;6(266):266ra171.
regeneration in dogs using a novel biomaterial. Am J Sports Med. 31. Li P, Feng X, Jia X, Fan Y. Influences of tensile load on in vitro deg-
1999;27(5):658-665. radation of an electrospun poly(L-lactide-co-glycolide) scaffold. Acta
9. Darling EM, Athanasiou KA. Rapid phenotypic changes in passaged Biomater. 2010;6(8):2991-2996.
articular chondrocyte subpopulations. J Orthop Res. 2005;23(2):425-432. 32. Liu C, Abedian R, Meister R, et al. Influence of perfusion and com-
10. Drury JL, Dennis RG, Mooney DJ. The tensile properties of alginate pression on the proliferation and differentiation of bone mesenchymal
hydrogels. Biomaterials. 2004;25(16):3187-3199. stromal cells seeded on polyurethane scaffolds. Biomaterials.
11. Esposito AR, Moda M, Cattani SM et al. PLDLA/PCL-T scaffold for 2012;33(4):1052-1064.
meniscus tissue engineering. Biores Open Access. 2013;2(2):138-147. 33. Makris EA, Hadidi P, Athanasiou KA. The knee meniscus: structure-
12. Frank MH, Sayegh MH. Immunomodulatory functions of mesenchy- function, pathophysiology, current repair techniques, and prospects
mal stem cells. Lancet. 2004;363(9419):1411-1412. for regeneration. Biomaterials. 2011;32(30):7411-7431.
13. Fu WL, Zhou CY, Yu JK. A new source of mesenchymal stem cells for 34. Mankin HJ, Dorfman H, Lippiello L, Zarins A. Biochemical and meta-
articular cartilage repair: MSCs derived from mobilized peripheral bolic abnormalities in articular cartilage from osteo-arthritic human
blood share similar biological characteristics in vitro and chondro- hips, II: correlation of morphology with biochemical and metabolic
genesis in vivo as MSCs from bone marrow in a rabbit model. Am data. J Bone Joint Surg Am. 1971;53(3):523-537.
J Sports Med. 2014;42(3):592-601. 35. Martin Seitz A, Galbusera F, Krais C, Ignatius A, Durselen L. Stress-
14. Garrett WE Jr, Swiontkowski MF, Weinstein JN, et al. American relaxation response of human menisci under confined compression
Board of Orthopaedic Surgery Practice of the Orthopaedic Surgeon: conditions. J Mech Behav Biomed Mater. 2013;26:68-80.
Part II, certification examination case mix. J Bone Joint Surg Am. 36. Merriam AR, Patel JM, Culp BM, Gatt CJ Jr, Dunn MG. Successful
2006;88(3):660-667. total meniscus reconstruction using a novel fiber-reinforced scaffold:
15. Holzer LA, Leithner A, Holzer G. Surgery versus physical therapy for a 16- and 32-week study in an ovine model. Am J Sports Med.
meniscal tear and osteoarthritis. N Engl J Med. 2013;369(7):677. 2015;43(10):2528-2537.
16. Horie M, Driscoll MD, Sampson HW, et al. Implantation of allogenic 37. Moran CJ, Busilacchi A, Lee CA, Athanasiou KA, Verdonk PC. Bio-
synovial stem cells promotes meniscal regeneration in a rabbit logical augmentation and tissue engineering approaches in meniscus
meniscal defect model. J Bone Joint Surg Am. 2012;94(8):701-712. surgery. Arthroscopy. 2015;31(5):944-955.
17. Huang H, Zhang X, Hu, X et al. A functional biphasic biomaterial hom- 38. Mow VC, Kuei SC, Lai WM, Armstrong CG. Biphasic creep and
ing mesenchymal stem cells for in vivo cartilage regeneration. Bioma- stress relaxation of articular cartilage in compression? Theory and
terials. 2014;35(36):9608-9619. experiments. J Biomech Eng. 1980;102(1):73-84.
18. Jiang D, Ao YF, Gong X, Wang YJ, Zheng ZZ, Yu JK. Comparative 39. Nakata K, Shino K, Hamada M, et al. Human meniscus cell: charac-
study on immediate versus delayed meniscus allograft transplanta- terization of the primary culture and use for tissue engineering. Clin
tion: 4- to 6-year follow-up. Am J Sports Med. 2014;42(10):2329- Orthop Relat Res. 2001;391(suppl):S208-218.
2337. 40. O’Conor CJ, Case N, Guilak F. Mechanical regulation of chondrogen-
19. Jiang D, Yu JK. Immediate versus delayed meniscus allograft trans- esis. Stem Cell Res Ther. 2013;4(4):61.
plantation: response. Am J Sports Med. 2015;43(5):NP9-10; discus- 41. Patel JM, Merriam AR, Culp BM, Gatt CJ Jr, Dunn MG. One-year out-
sion NP11-12. comes of total meniscus reconstruction using a novel fiber-reinforced
20. Jiang D, Zhao LH, Tian M, Zhang JY, Yu JK. Meniscus transplanta- scaffold in an ovine model. Am J Sports Med. 2016;44(4):898-907.
tion using treated xenogeneic meniscal tissue: viability and 42. Sastre S, Suso S, Segur JM, et al. Hyaline cartilage surface study
chondroprotection study in rabbits. Arthroscopy. 2012;28(8):1147- with an environmental scanning electron microscope: an experimen-
1159. tal study. J Mater Sci Mater Med. 2009;20(11):2181-2187.
21. Kang SW, Son SM, Lee JS, et al. Regeneration of whole meniscus 43. Steadman JR, Rodkey WG. Tissue-engineered collagen meniscus
using meniscal cells and polymer scaffolds in a rabbit total meniscec- implants: 5- to 6-year feasibility study results. Arthroscopy. 2005;
tomy model. J Biomed Mater Res A. 2006;77(4):659-671. 21(5):515-525.
22. Klompmaker J, Jansen HW, Veth RP, Nielsen HK, de Groot JH, Pen- 44. Sweigart MA, Athanasiou KA. Tensile and compressive properties of the
nings AJ. Porous implants for knee joint meniscus reconstruction: medial rabbit meniscus. Proc Inst Mech Eng H. 2005;219(5):337-347.
AJSM Vol. 45, No. 7, 2017 Scaffold Augmented With MSCs for Total Meniscal Substitution 1511

45. Szycher M. Biostability of polyurethane elastomers: a critical review. 50. Zhang Z-Z, Jiang D, Wang S-J, et al. Scaffolds drive meniscus tissue
J Biomater Appl. 1988;3(2):297-402. engineering. RSC Advances. 2015;5(95):77851-77859.
46. Tienen TG, Heijkants RG, de Groot JH, et al. Replacement of the 51. Zhang ZZ, Jiang D, Ding JX, et al. Role of scaffold mean pore size in
knee meniscus by a porous polymer implant: a study in dogs. Am meniscus regeneration. Acta Biomater. 2016;43:314-326.
J Sports Med. 2006;34(1):64-71. 52. Zhang ZZ, Jiang D, Wang SJ, Qi YS, Zhang JY, Yu JK. Potential of
47. Wang CJ, Zhou ZG, Holmqvist A, et al. Survivin expression quantified centrifugal seeding method in improving cells distribution and
by Image Pro-Plus compared with visual assessment. Appl Immuno- proliferation on demineralized cancellous bone scaffolds for tissue-
histochem Mol Morphol. 2009;17(6):530-535. engineered meniscus. ACS Appl Mater Interfaces. 2015;7(28):
48. Wang S-J, Zhang Z-Z, Jiang D, et al. Thermogel-coated poly(e-cap- 15294-15302.
rolactone) composite scaffold for enhanced cartilage tissue engi- 53. Zouein FA, Kurdi M, Booz GW, Fuseler JW. Applying fractal dimen-
neering. Polymers. 2016;8(5):200. sion and image analysis to quantify fibrotic collagen deposition and
49. Zein I, Hutmacher DW, Tan KC, Teoh SH. Fused deposition modeling organization in the normal and hypertensive heart. Microsc Micro-
of novel scaffold architectures for tissue engineering applications. anal. 2014;20(4):1134-1144.
Biomaterials. 2002;23(4):1169-1185.

For reprints and permission queries, please visit SAGE’s Web site at http://www.sagepub.com/journalsPermissions.nav.

You might also like