Accepted Manuscript Engineering of Hyaline Cartilage with a Calcified Zone Using Bone Marrow Stromal Cells Whitaik David Lee, BASc, Mark B. Hurtig, DVM, Robert M. Pilliar, PhD, William L. Stanford, PhD, Rita A. Kandel, MD PII:

S1063-4584(15)01130-9

DOI:

10.1016/j.joca.2015.04.010

Reference:

YJOCA 3459

To appear in:

Osteoarthritis and Cartilage

Received Date: 7 January 2015 Accepted Date: 8 April 2015

Please cite this article as: Lee WD, Hurtig MB, Pilliar RM, Stanford WL, Kandel RA, Engineering of Hyaline Cartilage with a Calcified Zone Using Bone Marrow Stromal Cells, Osteoarthritis and Cartilage (2015), doi: 10.1016/j.joca.2015.04.010. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

RI PT

Engineering of Hyaline Cartilage with a Calcified Zone Using Bone Marrow Stromal Cells Whitaik David Lee, BASc1; Mark B. Hurtig, DVM2; Robert M. Pilliar, PhD1,3; William L. Stanford, PhD1,4,*; Rita A. Kandel, MD1,5,*

1

SC

Institute of Biomaterials and Biomedical Engineering, University of Toronto; 164 College St., Toronto, Ontario M5S 3G9 Canada 2

M AN U

Ontario Veterinary College, University of Guelph; 50 McGulvray Lane, Guelph, Ontario N1G 2W1 Canada 3

Faculty of Dentistry, University of Toronto; 124 Edward St., Toronto, Ontario M5G 1G6 Canada 4

5

TE D

Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute & Departments of Cellular & Molecular Medicine and Biochemistry, Microbiology and Immunology, University of Ottawa 501 Smyth Road, Box 511. Ottawa, Ontario K1H 8L6 Canada Department of Pathology and Laboratory Medicine & Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto; 600 University Ave., Toronto, Ontario M5G 1X5 Canada

EP

* Co-corresponding authors

AC C

Correspondence Information:

Rita A. Kandel, MD Dept. of Pathology and Laboratory Medicine Mount Sinai Hospital 600 University Avenue Toronto, Ontario M5G 1X5 Canada Phone: +1 (416) 586-8516 Fax: +1 (416) 586-8719 Email: [email protected]

William L. Stanford, PhD Sprott Centre for Stem Cell Research Ottawa Health Research Institute 501 Smyth Road, Box 511 Ottawa, Ontario K1H 8L6 Canada Phone: +1 (613) 737-8899 x75495 Fax: +1 (613) 739-6294 Email: [email protected]

Running title (40 characters maximum): TE cartilage with a calcified interface

ACCEPTED MANUSCRIPT

Abstract

2

Objective: In healthy joints, a zone of calcified cartilage (ZCC) provides the mechanical

3

integration between articular cartilage and subchondral bone. Recapitulation of this

4

architectural feature should serve to resist the constant shear force from the movement of

5

the joint and prevent the delamination of tissue-engineered cartilage. Previous approaches

6

to create the ZCC at the cartilage-substrate interface have relied on strategic use of

7

exogenous scaffolds and adhesives, which are susceptible to failure by degradation and

8

wear. In contrast, we report a successful scaffold-free engineering of ZCC to integrate

9

tissue-engineered cartilage and a porous biodegradable bone substitute, using sheep bone

M AN U

SC

RI PT

1

marrow stromal cells (BMSCs) as the cell source for both cartilaginous zones.

11

Design: BMSCs were predifferentiated to chondrocytes, harvested and then grown on a

12

porous calcium polyphosphate substrate in the presence of triiodothyronine (T3). T3 was

13

withdrawn, and additional predifferentiated chondrocytes were placed on top of the

14

construct and grown for 21 days.

15

Results: This protocol yielded two distinct zones: hyaline cartilage that accumulated

16

proteoglycans and collagen type II, and calcified cartilage adjacent to the substrate that

17

additionally accumulated mineral and collagen type X. Constructs with the calcified

18

interface had comparable compressive strength to native sheep osteochondral tissue and

19

higher interfacial shear strength compared to control without a calcified zone.

20

Conclusion: This protocol improves on the existing scaffold-free approaches to cartilage

21

tissue engineering by incorporating a calcified zone. Since this protocol employs no

22

xenogeneic material, it will be appropriate for use in preclinical large-animal studies.

AC C

EP

TE D

10

ACCEPTED MANUSCRIPT

Introduction

2

In a synovial joint, articular cartilage bears both compressive and shear forces to facilitate

3

weight bearing and movement. Once damaged, these mechanical forces contribute to the

4

progression of cartilage damage, resulting in loss of cartilage and subchondral bone

5

remodelling. Tissue engineering is widely regarded as a promising technology to generate

6

constructs that could replace damaged cartilage and bone [1]. A wide range of

7

osteochondral tissue engineering strategies are under investigation [2], including our

8

effort to produce biphasic constructs that consist of scaffold-free cartilage tissue formed

9

by articular chondrocytes or bone marrow stromal cells (BMSCs) on porous bone

M AN U

SC

RI PT

1

substitutes (calcium polyphosphate) [3, 4]. Calcium polyphosphate is a biodegradable

11

ceramic with excellent load-bearing and osseointegrative properties [5, 6]. In our design,

12

ingrowth of bone into calcium polyphosphate holds the biphasic construct in place and

13

provides support against compressive force on tissue-engineered cartilage. However, to

14

resist shear force, a mechanically competent interface between the cartilage and its

15

substrate is needed. Preventing cartilage delamination is a common challenge shared by

16

many cartilage repair strategies [7, 8].

EP

18

Healthy cartilage tissue interfaces with bone in vivo through a zone of calcified

AC C

17

TE D

10

19

cartilage, which enhances mechanical integration by redistributing forces acting at the

20

interface [9]. BMSCs are suitable for creating calcified cartilage, because they can be

21

differentiated to produce cartilage tissues that mineralize their matrix [10]. While

22

chondrocytes from the deep zone of articular cartilage can also yield calcified cartilage in

23

vitro [11], BMSCs can be obtained without donor site morbidity and expanded to

1

ACCEPTED MANUSCRIPT

clinically relevant numbers. Furthermore, in vivo studies have shown that BMSC-derived

2

cartilage can maintain their non-calcified phenotype after being implanted in a joint

3

cavity [12, 13], demonstrating the potential of BMSCs to produce both calcified and non-

4

calcified cartilage. However, to use BMSCs as a single source of cells to create cartilage

5

tissues with a calcified cartilaginous interface, limiting mineralization of BMSC-derived

6

cartilage to the interface will be crucial for the success of these implants.

8

Using BMSCs as the cell source, we report the first successful engineering of an osteochondral-like construct that incorporates a zone of calcified cartilage at the

M AN U

9

SC

7

RI PT

1

cartilage-calcium polyphosphate interface without the use of a scaffold as the cartilage

11

tissue forms on top of a substrate. A stepwise culturing strategy enabled the selective

12

activation of BMSC-derived chondrocytes to induce mineralization only at the interface,

13

while avoiding mineralization in the contiguous cartilage tissue, thus conferring localized

14

calcified and hyaline zones to the engineered cartilage tissue. This approach is suitable

15

for clinical use as the BMSCs were expanded in media supplemented with autologous

16

serum and the construct cultured in serum-free, defined media, so this strategy can be

17

directly translated to in vivo preclinical studies.

EP

AC C

18

TE D

10

2

ACCEPTED MANUSCRIPT

Materials and methods

2

Chondrogenic predifferentiation of BMSCs in membrane cultures

3

Sheep BMSCs were isolated and expanded from bone marrow aspirates taken from ewes

4

aged between 2 and 5 years as previously described [4] using autologous serum (see

5

Supplementary Methods). To differentiate BMSCs to chondrocytes, 2.0×106 BMSCs

6

were seeded onto 12mm cell culture inserts (0.2µm pore size; Millipore, Billerica, MA)

7

that were coated with collagen type IV (50µg/insert; Sigma-Aldrich, Oakville, ON,

8

Canada). Cells were cultured in a defined chondrogenic media composed of high-glucose

9

Dulbecco’s modified Eagle medium (DMEM; Life Technologies, Burlington, ON,

10

Canada), 1× insulin-transferrin-selenium plus (ITS+) cell culture supplement (BD

11

Biosciences, Bedford, MA), 100nM dexamethasone (Sigma-Aldrich), 100µg/mL ascorbic

12

acid (Sigma-Aldrich) and 10ng/mL transforming growth factor-β3 (TGF-β3; R&D

13

Systems, Minneapolis, MN). For the first 72 hours, chondrogenic media was additionally

14

supplemented with 10µM blebbistatin (Cayman Chemical, Ann Arbor, MI) to inhibit

15

spontaneous detachment of cells. Media was changed every 2-3 days. After 3 weeks of

16

culture, cells (herein referred to as predifferentiated chondrocytes) were isolated by

17

digesting the tissue in 0.5% (w/v) collagenase (Roche Diagnostics, Indianapolis, IN) for 2

18

hours at 37ºC.

SC

M AN U

TE D

EP

AC C

19

RI PT

1

20

Preparation of porous calcium polyphosphate substrates with a hydroxyapatite coating

21

Porous calcium polyphosphate disks of 4mm diameter and 2mm height were prepared

22

from gravity-sintered 75-150µm calcium polyphosphate particles as previously described

23

[14]. Disks were incubated in phosphate-buffered saline (PBS) for 1 week. Then, a thin,

3

ACCEPTED MANUSCRIPT

submicron hydroxyapatite layer was deposited over the disks using a sol-gel processing

2

method. This hydroxyapatite layer was intended to serve as a ‘barrier’ coating inhibiting

3

the rate of release of calcium polyphosphate degradation products during in vitro

4

cartilage formation [15]. The sol was prepared with triethyl phosphite and calcium nitrate

5

tetrahydrate (Sigma-Aldrich) as previously described [16]. Disks were dipped into the sol

6

gel sideways for 8 seconds and withdrawn at a speed of 20cm/min using a stepper motor-

7

driven actuator, air-dried for 10 minutes and annealed for 15 minutes at 210ºC. To ensure

8

a continuous barrier coating, coated disks were dipped again, annealed at 500ºC for 20

9

minutes and gradually cooled. The coated disks were placed in Tygon tubing to create a

SC

M AN U

10

RI PT

1

well-like structure and subsequently sterilized by γ-irradiation (2.5 MRad).

11

Optimizing the mineralizing culture condition for predifferentiated chondrocytes

13

To find a culture condition in which predifferentiated chondrocytes would form calcified

14

cartilage tissue, predifferentiated chondrocytes were placed on the top surface of porous

15

calcium polyphosphate substrates (1.5×106 cells/substrate) and cultured in basal construct

16

media composed of DMEM, 1× ITS+, 100µg/mL ascorbic acid, 50µg/mL L-proline and

17

10mM β-glycerophosphate (Sigma-Aldrich). To determine which condition would induce

18

mineralization, 100nM dexamethasone, 3nM triiodothyronine (T3) or 100nM retinoic

19

acid (all Sigma-Aldrich) were added to cultures. The cultures were harvested at various

20

times up to 21 days for further study.

22

EP

AC C

21

TE D

12

Tissue culture of multiphasic constructs

4

ACCEPTED MANUSCRIPT

Predifferentiated chondrocytes were placed on the top surface of porous calcium

2

polyphosphate substrates (1.0×106 cells/substrate) and cultured in basal construct media,

3

with or without 3nM T3. T3 was withdrawn at day 4. At day 5, additional

4

predifferentiated chondrocytes were placed on top of the existing constructs (1.5×106

5

cells/construct). The multi-layered constructs were maintained under the same culture

6

conditions for up to 21 days. This protocol is shown in Figure 1.

RI PT

1

SC

7

Visualization and quantification of alkaline phosphate (ALP) activity

9

To visualize ALP activity, tissues were removed from their substrates and fixed in 10%

M AN U

8

neutral buffered formalin for 1 hour. Then, samples were incubated in 30% (w/v) sucrose

11

overnight at 4ºC and snap-frozen in Tissue-Tek OCT compound (Sakura Finetek,

12

Torrance, CA). 6µm cross-section cryosections were cut and mounted on silanized glass

13

slides. Sections were incubated in azo dye (Naphthol AS-MX phosphate and Fast Blue

14

BB salt, both Sigma-Aldrich) for 10 minutes, counterstained with eosin, dried and

15

mounted with coverslips. To quantify the ALP activity, cells were isolated from tissues

16

by digestion in 0.5% collagenase for 2 hours and lysed by sonication for 15 minutes in

17

0.2M Tris pH 7.4 buffer. Activity levels were quantified with p-nitrophenol phosphate

18

(Sigma-Aldrich) and measuring the absorbance at 405nm against a standard curve of p-

19

nitrophenol [11]. Values were normalized to the lysate’s DNA content (see Biochemical

20

Analysis).

22

EP

AC C

21

TE D

10

Gene expression analysis

5

ACCEPTED MANUSCRIPT

Predifferentiated chondrocytes (1.5×106 cells/substrate) were cultured on porous calcium

2

polyphosphate substrates in basal construct media with 4 days of 3nM T3. T3 was

3

removed after 4 days, and the cells were grown in basal construct media only for the

4

remainder of the 21 days of culture. Controls were cells that were untreated and grown in

5

the same media. Tissues were removed from their substrates at day 4 and 21 and

6

homogenized in 1mL TRIzol reagent (Life Technologies) by bead milling. Total RNA

7

was then extracted and reverse-transcribed using SuperScript VILO cDNA Synthesis Kit

8

(Life Technologies). Quantitative PCR was performed using a LightCycler 96 Real-Time

9

PCR System (Roche Diagnostics) and SYBR GreenER master mix (Life Technologies)

SC

M AN U

10

RI PT

1

with gene-specific primers (Supplementary Table 1).

11

Micro-computed tomography (µCT) imaging

13

Constructs were imaged using a Skyscan 1174v2 µCT scanner (Bruker, Belgium).

14

Scanning was performed at 50kV and 800µA through a 0.25mm aluminum shield with a

15

voxel size of 6.9µm. After reconstruction, cross-sectional tomographs were obtained with

16

the software provided by the manufacturer.

EP

17

TE D

12

Histological analysis of whole constructs

19

Constructs were fixed in 10% neutral buffered formalin overnight and infiltrated using

20

the Osteo-Bed bone embedding kit (Polysciences Inc., Warrington, PA). Sections were

21

cut and ground to ~50µm thickness, stained with toluidine blue and light green and

22

visualized under light microscopy.

AC C

18

23

6

ACCEPTED MANUSCRIPT

Histological analysis of cartilaginous tissues

2

Tissue mineral and proteoglycan accumulation was visualized by staining histological

3

sections (see Supplementary Methods) with von Kossa and toluidine blue. Orientation of

4

collagen fibrils was visualized by polarized light microscopy of trichrome-stained

5

sections. Visualization for collagens types I, II and X were visualized by

6

immunofluorescence (see Supplementary Methods).

SC

7

RI PT

1

Biochemical analysis of extracellular matrix and mineral accumulation

9

Tissues were digested in 40µg/mL papain (Sigma-Aldrich) and DNA, glycosaminoglycan

M AN U

8

and hydroxyproline contents were quantified as previously described [17]. To measure

11

mineral accumulation, the tissues were lyophilized, dry weights measured, and then

12

digested in 3N hydrochloric acid at 90ºC for 2 hours. The pH was adjusted to 4.0 with

13

1.5M acetate buffer. Phosphate content was determined using the heteropoly blue assay

14

and measuring the absorbance at 620nm. Calcium content was determined using the o-

15

cresolphthalein complexone assay and measuring the absorbance at 570nm.

17

Mechanical testing of multiphasic constructs

18

On day 21 of culture, the equilibrium compressive modulus of the multiphasic construct’s

19

cartilaginous tissue was determined by stress relaxation testing on the Mach-1 mechanical

20

tester (Bio-Momentum, Laval, Canada) with a 0.65mm-diameter indenter as previously

21

described [4]. Interfacial shear strength was determined by applying a force at the

22

interface region of these samples using a specially designed jig attached to an Instron

23

universal testing machine as previously described [15] (see Supplementary Methods).

AC C

EP

16

TE D

10

7

ACCEPTED MANUSCRIPT

1 Statistical testing

3

All values are expressed as mean ± 95% confidence intervals (CI). Each experiment was

4

carried out with three donor animals (N=3). Univariate analysis of variance and Tukey

5

post-hoc testing were used to analyze ALP activity levels on days 0-4, accumulation of

6

GAG, total collagen and mineral contents, as well as the compressive moduli of the

7

constructs. The interfacial shear strength data of the constructs were analyzed using t test.

8

To account for the wide animal-to-animal variability, ALP activity levels on days 7, 10

9

and 14 were each analyzed using paired t test. Similarly, gene expression levels on days 4

SC

M AN U

10

RI PT

2

and 21 were log-transformed and analyzed using paired t test.

AC C

EP

TE D

11

8

ACCEPTED MANUSCRIPT

Results

2

Generation of calcified cartilage at the calcium polyphosphate interface with

3

predifferentiated chondrocytes

4

To engineer a multiphasic construct that incorporates a zone of calcified cartilage at the

5

cartilage-calcium polyphosphate interface using predifferentiated chondrocytes, we first

6

sought to establish a culture condition in which predifferentiated chondrocytes could be

7

cultured on porous calcium polyphosphate substrates to form mineralized cartilage tissue.

8

Triiodothyronine (T3) [18], retinoic acid [19] and dexamethasone [10] have all been

9

identified as potential inducers of chondrocyte mineralization in vitro. We cultured

M AN U

SC

RI PT

1

predifferentiated chondrocytes on calcium polyphosphate substrates with either 3nM T3

11

or 100µM retinoic acid treatment in the presence or absence of dexamethasone for 1

12

week, and then quantified the alkaline phosphate activity of the cells as an indicator of

13

chondrocyte mineralization potential [20, 21]. ALP activity was the highest in

14

predifferentiated chondrocytes cultured in the absence of dexamethasone with the T3

15

treatment (Figure 2A). While ALP activity was also increased with the retinoic acid

16

treatment, continued culture did not yield cartilaginous tissues on calcium polyphosphate

17

after 3 weeks (data not shown). Therefore, treatment of predifferentiated chondrocytes

18

with T3 was selected for further study.

20

EP

AC C

19

TE D

10

When 2×106 predifferentiated chondrocytes were cultured on porous calcium

21

polyphosphate substrates for 3 weeks, the mineralized zone was found in the top of the

22

tissue (Figure 2B). Between this mineralized zone and the substrate was an intermediary

23

zone of non-mineralized cartilage, composed of cells with round morphology and

9

ACCEPTED MANUSCRIPT

extracellular matrix rich in proteoglycans. Changing the concentration of T3 treatment

2

did not change the location of the mineralized zone (data not shown). However, when the

3

number of initially seeded predifferentiated chondrocytes was decreased, the distance

4

between the cartilage-substrate interface and the zone of mineralized cartilage also

5

decreased (Figure 2C & 2D). At an initial seeding of 1×106 predifferentiated

6

chondrocytes, the zone of mineralized cartilage formed consistently adjacent to the

7

interface (Figure 2C & 2D, arrows).

SC

RI PT

1

8

Short-term treatment of predifferentiated chondrocytes with T3 was sufficient to stimulate

M AN U

9

terminal differentiation

11

ALP activity levels of predifferentiated chondrocytes were characterized over a 2-week

12

culture period. T3-treated cells had a significantly increased ALP activity by day 4

13

(Figure 3A). Even though the T3 treatment was withdrawn at day 4, the ALP activity

14

level of cells at day 7 was significantly higher than those that had been continuously

15

treated with T3, and were comparable at days 10 and 14 (Figure 3B).

EP

16

TE D

10

To confirm that the 4-day T3 treatment was sufficient to induce terminal

18

differentiation of predifferentiated chondrocytes, gene expression levels were examined.

19

As early as 4 days, collagen type X (Col10a1) expression was significantly higher in the

20

T3-treated cells compared to the untreated control (Figure 3G). By 21 days, T3-treated

21

cells expressed less collagen type II (Col2a1), aggrecan (Agc1) and Sox9, but greater

22

Col10a1 and osteopontin (Spp1) compared to the non-treated controls (Figure 3C-J). This

AC C

17

10

ACCEPTED MANUSCRIPT

1

confirmed that the 4-day T3 treatment of predifferentiated chondrocytes had altered the

2

differentiation of these cells.

3 T3-treated predifferentiated chondrocytes did not induce ALP activity in non-T3-treated

5

predifferentiated chondrocytes

6

Based on this observation, a two-stage culture protocol was developed (Figure 1). To

7

grow an interfacial zone of mineralized cartilage, 1.0×106 predifferentiated chondrocytes

8

were first seeded on porous calcium polyphosphate substrates, which was the number of

9

cells that had been previously determined to be optimal for creating the mineralized

M AN U

SC

RI PT

4

cartilage at the interface, and cultured in the presence of T3 (Figure 2C & 2D). T3 was

11

withdrawn at 4 days of culture, then 1.5×106 predifferentiated chondrocytes were seeded

12

on top of the existing tissue one day later and cultured in the presence of β-

13

glycerophosphate for up to 21 days to generate a zone of non-mineralized cartilage. To

14

confirm that the T3-treated cells would not stimulate the overlaid, non-T3-treated cells to

15

also mineralize, the tissues on calcium polyphosphate were harvested 5 days after the

16

addition of the top layer of cells (10 days of culture in total) and cryosectioned.

17

Histological analysis demonstrated that ALP activity was present only in the calcifying

18

bottom layer (Figure 4), demonstrating the lack of mineralization potential in the top

19

layer even though β-glycerophosphate was present.

EP

AC C

20

TE D

10

21

Characterization of the multiphasic constructs

22

Constructs were grown for 21 days using the two-stage culture protocol with or without

23

the 4-day T3 treatment of the interfacial layer and characterized using various methods.

11

ACCEPTED MANUSCRIPT

Histological examination of cartilage tissue on the calcium polyphosphate showed two

2

tissue zones. The upper zone of non-mineralized cartilage was hyaline cartilage, rich in

3

type II collagen and proteoglycan (Figure 5A, B, D). The interfacial zone was calcified

4

cartilage, the mineral staining with von Kossa staining and the extracellular matrix

5

staining with toluidine blue and type X collagen (Figure 5A, B, E). Ingrowth of tissue

6

into upper parts of the porous calcium polyphosphate substrate was also observed, with

7

the mineral clusters directly in contact with the calcium polyphosphate particles (Figure

8

5A). No type I collagen was detected in either zone (Figure 5C). In contrast, control

9

constructs not treated with T3 did not show a bizonal composition as there was no zone

M AN U

SC

RI PT

1

of calcified cartilage, and there was an uneven distribution of extracellular matrix (Figure

11

5F-I). Differential collagen alignment in the tissue was detected using polarized light

12

microscopy. In the superficial aspect, collagen was aligned parallel to the surface.

13

Elsewhere, collagen was observed both pericellularly and aligned orthogonal to the

14

surface. Collagen, parallel to the surface, was also observed in areas within the bottom

15

layer of tissue (Figure 5J). T3-treated constructs accumulated less proteoglycans (Figure

16

6A) and total collagens (Figure 6B) compared to those on untreated constructs, but had

17

amounts comparable to native sheep cartilage. Tissues on T3-treated constructs

18

accumulated calcium and phosphate, consistent with calcification, while untreated

19

constructs did not (Figure 6C).

21

EP

AC C

20

TE D

10

T3-treated tissues had a comparable equilibrium compressive modulus to that of

22

osteochondral tissue obtained from native sheep femoral condyles (Figure 7A, p = 0.15).

23

The untreated constructs had a significantly higher equilibrium compressive modulus

12

ACCEPTED MANUSCRIPT

compared to that of the T3-treated tissues, possibly due to their greater extracellular

2

matrix accumulation (Figure 6A, 6B). However, T3-treated constructs could withstand a

3

stronger shear force (Figure 7B) and absorb more energy (Figure 7C) than untreated

4

constructs. Histological assessment of the failed constructs confirmed that failure

5

occurred at the interface (data not shown). This demonstrated that the mineralized tissue

6

at the interface (Figure 5A) enhanced the interfacial shear strength.

RI PT

1

SC

7

AC C

EP

TE D

M AN U

8

13

ACCEPTED MANUSCRIPT

Discussion

2

A mechanically competent interface between the cartilage and its substrate is required to

3

resist the shear force generated by the movement of the joint. Previous studies have

4

sought to recreate the zone of calcified cartilage that exists between hyaline cartilage and

5

bone to reinforce this interface with limited success [22]. To overcome this problem, we

6

successfully tissue engineered osteochondral-like constructs with BMSCs as the single

7

cell source, in which cartilage tissue and a porous bone substitute substrate were formed

8

with an interfacial zone of calcified cartilage without using any exogenous scaffolds in

9

the cartilage tissue. The two-stage culture protocol enabled the selective stimulation of

M AN U

SC

RI PT

1

predifferentiated chondrocytes to form calcified cartilage only at the interface. Treatment

11

of the initially seeded cells with T3 induced the terminal differentiation of cells. This

12

resulted in a zone of calcified cartilage, which, in addition to mineral, also contained

13

collagens type II and X and proteoglycans. These features were seen only in the part of

14

the tissue that was treated with mineralization-inducing T3, which was adjacent to (and

15

integrated with) the porous calcium polyphosphate substrate. The interfacial shear

16

strength of constructs with the calcified cartilaginous interface was increased

17

significantly compared to those without it, functionally validating its intended effect. The

18

hyaline quality of non-mineralized cartilage was demonstrated by the accumulation of

19

collagen type II and the lack of mineral or collagen type I. Further study is required to

20

determine whether this osteochondral-like tissue formation will occur on substrates other

21

than hydroxyapatite-coated calcium polyphosphate substrate.

AC C

EP

TE D

10

22

14

ACCEPTED MANUSCRIPT

Thus far, approaches to induce selective mineralization of BMSC-derived

2

cartilage and create this interface include delivery of bioactive signals either locally via

3

microspheres [23] or by using gradients generated by a bioreactor [24] to undifferentiated

4

BMSCs seeded in scaffolds. In another approach, BMSCs predifferentiated to

5

chondrocytes and osteoblasts are seeded into appropriate locations within scaffolds [25,

6

26]. For both strategies, use of exogenous scaffolds is necessary, which has the potential

7

to hinder host integration and tissue regeneration [27]. Matching the rate of scaffold

8

degeneration to the rate of tissue accumulation remains a challenge when employing

9

scaffolds [22, 28]. Synthetic materials also accumulate wear over time, and may provide

M AN U

SC

RI PT

1

a focal point for shear failure if not replaced with new tissue. On the other hand, if the

11

rate of scaffold degradation exceeds the rate of cellular remodelling, the mechanical

12

integrity is compromised and the construct would fail under load. A scaffold-free

13

approach obviates this issue altogether; however, this necessitated an alternative strategy

14

to specify the location of the calcified interface.

15

TE D

10

The development of our two-stage culture protocol was first enabled by the

17

observation that BMSC-derived predifferentiated chondrocytes formed mineralized

18

cartilage at the tissue’s superficial aspect with mineral-inducing stimulus. Given that T3

19

was delivered to the tissue through culture media, and that the location of calcified

20

cartilage did not change with tissue thickness, this effect was likely due to T3 diffusion

21

into the tissue. As there was no scaffold present, selective delivery of T3 to the interface

22

was difficult to achieve with media alone. To circumvent this, we seeded a small number

23

of cells on the calcium polyphosphate in the presence of T3. This generated thin

AC C

EP

16

15

ACCEPTED MANUSCRIPT

cartilaginous tissue, forcing mineralization to take place juxtaposed to the porous calcium

2

polyphosphate substrate. This enabled the subsequent formation of a continuous layer of

3

non-mineralized cartilage formed by the same cell type in the absence of T3. The

4

presence of collagen types II, X and proteoglycans in the calcified interface, as well as

5

the absence of collagen type I, is an important finding in our study. Taken together with

6

the finding that T3-treated cells expressed hypertrophic chondrocyte makers, this

7

demonstrates that the interface is cartilaginous, not osseous, successfully recapitulating

8

characteristics of the native articular cartilage-subchondral bone interface.

SC

RI PT

1

10

M AN U

9

Histology showed that the layer of non-mineralized cartilage successfully fused to the underlying T3-treated tissue. Interestingly, fissures were observed if the intended

12

interfacial tissue (i.e., the first layer) was not treated with T3, and the total thickness of

13

cartilage tissues on these constructs was increased, with an uneven distribution of the

14

extracellular matrix. There are two possible explanations for these results. One is that the

15

manner that the interfacial layer grows may affect integration, as BMSC-derived

16

chondrocytes share many characteristics with proliferating cartilage [29] that grows

17

appositionally. In development, Indian hedgehog, FGF and BMP signaling pathways

18

regulate this appositional growth[30]; however, in an in vitro system, absence of such

19

complex regulation may allow the interfacial layer to expand laterally. The presence of

20

tubing around the constructs prevented lateral expansion by confinement, which could

21

have resulted in lateral mechanical stress and caused the tissue to buckle and shear. This

22

was not the case for T3-stimulated interfacial layer, however, whose behaviour may more

23

resemble terminal hypertrophic chondrocytes, as evidenced by their mineralized matrix.

AC C

EP

TE D

11

16

ACCEPTED MANUSCRIPT

Terminal hypertrophic chondrocytes do not proliferate and eventually undergo apoptosis

2

upon vascularization of matrix and bone formation [31]. Alternatively, the T3 treatment

3

may have affected tissue fusion. Pellets of human MSCs undergoing chondrogenesis have

4

been shown to accumulate tenascin along the outer surface that prevented them from

5

fusing with one another [32]. The fissure between the two layers of non-mineralizing

6

cartilage may stem from a similar boundary-setting phenomenon, whether driven by

7

tenascin accumulation or otherwise.

SC

RI PT

1

8

Sheep gait studies estimate the maximum anterioposterior shear stress in a stifle

M AN U

9

joint to be about 500kPa [33, 34], which is equivalent to approximately 9N of peak shear

11

force in our study. However, using our methodology the peak shear force of native

12

cartilage interface could not be measured, as it withstood the maximum load (80N)

13

without shear failure. In healthy cartilage, collagen fibrils run orthogonally through the

14

tidemark plane, which would confer a high resistance against failure in our mode of

15

study. Although our multiphasic construct possesses inferior interfacial shear strength

16

compared to articular cartilage, it still represented a four-fold improvement over the non-

17

mineralized control, and sheep gait studies suggest that this improvement is adequate for

18

preventing delamination.

20

EP

AC C

19

TE D

10

Importantly, this protocol can be expanded to generate large, anatomically shaped

21

osteochondral-like constructs as replacements for damaged joint tissues. Protocols have

22

been developed to generate ~108 BMSCs with chondrogenic potential in a good

23

manufacturing practices-complaint manner [35, 36], which can be used to generate

17

ACCEPTED MANUSCRIPT

sufficiently large numbers of chondrocytes to cover a large articular surface. We have

2

previously shown that calcium polyphosphate powders can be used in additive

3

manufacturing methods to generate substrates of required shape and size [37]. Calcium

4

polyphosphate formed in this way exhibited the same degree of osseointegration after

5

implantation as those produced conventionally [38]. As such, preclinical studies are

6

warranted to determine if these osteochondral-like constructs with a mineralized

7

cartilaginous interface could repair joint defects in vivo.

SC

8 Conclusion

M AN U

9

RI PT

1

Using BMSCs as a single cell source, an osteochondral-like construct comprised of

11

hyaline cartilage and porous bone substitute was created with a zone of calcified cartilage

12

interfacing them. The novel, two-step cell culture strategy employed no exogenous

13

scaffolds, just a substrate on which to form tissue. The presence of calcified cartilage

14

increased the shear load that the construct could withstand at the interface. As no

15

xenogeneic material was used at any point during culture, this strategy can be directly

16

translated into preclinical studies to determine whether these constructs can repair joint

17

defects in vivo.

AC C

EP

TE D

10

18

ACCEPTED MANUSCRIPT

Acknowledgments

2

We thank Eugene Hu for preparing the porous calcium polyphosphate substrates. We also

3

thank Dr. Jian Wang and Nancy Valiquette for their technical assistance. This work was

4

supported by the United States Department of Defense (to R.A.K., W81XWH-10-1-0787)

5

and the Natural Sciences and Engineering Research Council of Canada (to W.L.S.,

6

RGPIN 293170-11). W.L.S. is supported by a Canadian Research Chair.

RI PT

1

9

Author Contributions

Study conception and design: WDL, WLS, RAK.

M AN U

8

SC

7

Provision of study materials: MBH, RMP.

11

Analysis and interpretation of data: WDL, RMP, RAK.

12

Drafting of article: WDL, WLS, RAK.

13

Critical revision of the article: WDL, RMP, WLS, RAK.

14

Final approval of the article: WDL, WLS, RAK.

TE D

10

15

Role of the Funding Sources

17

None of the listed bodies played any role in this study.

AC C

18

EP

16

19

Conflict of Interest

20

No competing financial interests exist for any of the listed authors.

19

ACCEPTED MANUSCRIPT

1

References

2

1.

Swieszkowski W, Tuan BHS, Kurzydlowski KJ, Hutmacher DW. Repair and regeneration of osteochondral defects in the articular joints. Biomolecular

4

engineering 2007; 24: 489-495.

5

2.

RI PT

3

Shimomura K, Moriguchi Y, Murawski CD, Yoshikawa H, Nakamura N.

Osteochondral Tissue Engineering with Biphasic Scaffold: Current Strategies and

7

Techniques. Tissue Eng Part B Rev 2014; 20: 468-476. 3.

9

osteochondral defects with biphasic cartilage-calcium polyphosphate constructs in

10 11

Kandel RA, Grynpas M, Pilliar R, Lee J, Wang J, Waldman S, et al. Repair of

M AN U

8

SC

6

a sheep model. Biomaterials 2006; 27: 4120-4131. 4.

Lee WD, Hurtig MB, Kandel RA, Stanford WL. Membrane culture of bone marrow stromal cells yields better tissue than pellet culture for engineering

13

cartilage-bone substitute biphasic constructs in a two-step process. Tissue

14

engineering. Part C, Methods 2011; 17: 939-948.

15

5.

TE D

12

Grynpas MD, Pilliar RM, Kandel RA, Renlund R, Filiaggi M, Dumitriu M. Porous calcium polyphosphate scaffolds for bone substitute applications in vivo

17

studies. Biomaterials 2002; 23: 2063-2070. 6.

19

load-bearing bone substitutes: in vivo study. J Biomed Mater Res B Appl

20 21

Pilliar RM, Kandel RA, Grynpas MD, Hu Y. Porous calcium polyphosphate as

AC C

18

EP

16

Biomater 2013; 101: 1-8.

7.

Vahdati A, Wagner DR. Implant size and mechanical properties influence the

22

failure of the adhesive bond between cartilage implants and native tissue in a

23

finite element analysis. J Biomech 2013; 46: 1554-1560.

20

ACCEPTED MANUSCRIPT

1

8.

Harris JD, Siston RA, Brophy RH, Lattermann C, Carey JL, Flanigan DC.

2

Failures, re-operations, and complications after autologous chondrocyte

3

implantation--a systematic review. Osteoarthritis Cartilage 2011; 19: 779-791. 9.

Hoemann CD, Lafantaisie-Favreau C-H, Lascau-Coman V, Chen G, Guzmán-

RI PT

4 5

Morales J. The cartilage-bone interface. The journal of knee surgery 2012; 25: 85-

6

97. 10.

Mueller MB, Tuan RS. Functional characterization of hypertrophy in

SC

7

chondrogenesis of human mesenchymal stem cells. Arthritis and rheumatism

9

2008; 58: 1377-1388.

10

11.

M AN U

8

Allan KS, Pilliar RM, Wang J, Grynpas MD, Kandel RA. Formation of biphasic

11

constructs containing cartilage with a calcified zone interface. Tissue engineering

12

2007; 13: 167-177. 12.

Zscharnack M, Hepp P, Richter R, Aigner T, Schulz R, Somerson J, et al. Repair

TE D

13

of chronic osteochondral defects using predifferentiated mesenchymal stem cells

15

in an ovine model. The American journal of sports medicine 2010; 38: 1857-

16

1869.

18 19 20 21

13.

Shimomura K, Moriguchi Y, Ando W, Nansai R, Fujie H, Hart DA, et al. Osteochondral Repair Using a Scaffold-Free Tissue-Engineered Construct

AC C

17

EP

14

Derived from Synovial Mesenchymal Stem Cells and a Hydroxyapatite-Based

Artificial Bone. Tissue Eng Part A 2014.

14.

Waldman SD, Grynpas MD, Pilliar RM, Kandel RA. Characterization of

22

cartilagenous tissue formed on calcium polyphosphate substrates in vitro. Journal

23

of biomedical materials research 2002; 62: 323-330.

21

ACCEPTED MANUSCRIPT

1

15.

St-Pierre J-P, Gan L, Wang J, Pilliar RM, Grynpas MD, Kandel RA. The incorporation of a zone of calcified cartilage improves the interfacial shear

3

strength between in vitro-formed cartilage and the underlying substrate. Acta

4

biomaterialia 2012; 8: 1603-1615.

5

16.

RI PT

2

Gan L, Pilliar R. Calcium phosphate sol-gel-derived thin films on porous-surfaced implants for enhanced osteoconductivity. Part I: Synthesis and characterization.

7

Biomaterials 2004; 25: 5303-5312. 17.

9

accumulation by passaged chondrocytes can be enhanced through side-by-side

10 11

Taylor DW, Ahmed N, Gan L, Gross AE, Kandel RA. Proteoglycan and collagen

M AN U

8

SC

6

culture with primary chondrocytes. Tissue engineering. Part A 2010; 16: 643-651. 18.

Alini M, Kofsky Y, Wu W, Pidoux I, Poole AR. In serum-free culture thyroid hormones can induce full expression of chondrocyte hypertrophy leading to

13

matrix calcification. Journal of bone and mineral research : the official journal of

14

the American Society for Bone and Mineral Research 1996; 11: 105-113.

15

19.

TE D

12

Iwamoto M, Shapiro IM, Yagami K, Boskey AL, Leboy PS, Adams SL, et al. Retinoic acid induces rapid mineralization and expression of mineralization-

17

related genes in chondrocytes. Experimental cell research 1993; 207: 413-420.

19 20 21 22

20.

Miao D, Scutt A. Histochemical localization of alkaline phosphatase activity in

AC C

18

EP

16

decalcified bone and cartilage. J Histochem Cytochem 2002; 50: 333-340.

21.

Jiang J, Leong NL, Mung JC, Hidaka C, Lu HH. Interaction between zonal populations of articular chondrocytes suppresses chondrocyte mineralization and

this process is mediated by PTHrP. Osteoarthritis Cartilage 2008; 16: 70-82.

22

ACCEPTED MANUSCRIPT

1

22.

2 3

Huey DJ, Hu JC, Athanasiou KA. Unlike bone, cartilage regeneration remains elusive. Science 2012; 338: 917-921.

23.

Dormer NH, Singh M, Wang L, Berkland CJ, Detamore MS. Osteochondral interface tissue engineering using macroscopic gradients of bioactive signals.

5

Annals of biomedical engineering 2010; 38: 2167-2182.

6

24.

RI PT

4

Chen K, Ng KS, Ravi S, Goh JC, Toh SL. In vitro generation of whole

osteochondral constructs using rabbit bone marrow stromal cells, employing a

8

two-chambered co-culture well design. J Tissue Eng Regen Med 2013. 25.

Cheng H-W, Luk KDK, Cheung KMC, Chan BP. In vitro generation of an

M AN U

9

SC

7

10

osteochondral interface from mesenchymal stem cell-collagen microspheres.

11

Biomaterials 2011; 32: 1526-1535.

12

26.

Grayson WL, Bhumiratana S, Grace Chao PH, Hung CT, Vunjak-Novakovic G. Spatial regulation of human mesenchymal stem cell differentiation in engineered

14

osteochondral constructs: effects of pre-differentiation, soluble factors and

15

medium perfusion. Osteoarthritis and cartilage / OARS, Osteoarthritis Research

16

Society 2010; 18: 714-723.

19 20 21 22

EP

18

27.

Castro NJ, Hacking SA, Zhang LG. Recent progress in interfacial tissue engineering approaches for osteochondral defects. Annals of biomedical

AC C

17

TE D

13

engineering 2012; 40: 1628-1640.

28.

Dhote V, Vernerey FJ. Mathematical model of the role of degradation on matrix development in hydrogel scaffold. Biomech Model Mechanobiol 2014; 13: 167183.

23

ACCEPTED MANUSCRIPT

1

29.

Dickhut A, Pelttari K, Janicki P, Wagner W, Eckstein V, Egermann M, et al.

2

Calcification or dedifferentiation: requirement to lock mesenchymal stem cells in

3

a desired differentiation stage. Journal of cellular physiology 2009; 219: 219-226. 30.

5 6

Kronenberg HM. Developmental regulation of the growth plate. Nature 2003;

RI PT

4

423: 332-336. 31.

Farnum CE, Wilsman NJ. Cellular turnover at the chondro-osseous junction of

growth plate cartilage: analysis by serial sections at the light microscopical level.

8

J Orthop Res 1989; 7: 654-666. 32.

Bhumiratana S, Eton RE, Oungoulian SR, Wan LQ, Ateshian GA, Vunjak-

M AN U

9

SC

7

10

Novakovic G. Large, stratified, and mechanically functional human cartilage

11

grown in vitro by mesenchymal condensation. Proc Natl Acad Sci U S A 2014;

12

111: 6940-6945. 33.

Taylor WR, Poepplau BM, König C, Ehrig RM, Zachow S, Duda GN, et al. The

TE D

13

medial-lateral force distribution in the ovine stifle joint during walking. Journal of

15

orthopaedic research : official publication of the Orthopaedic Research Society

16

2011; 29: 567-571.

18 19 20 21

34.

Lee-Shee NK, Dickey JP, Hurtig MB. Contact mechanics of the ovine stifle during simulated early stance in gait. An in vitro study using robotics. Vet Comp

AC C

17

EP

14

Orthop Traumatol 2007; 20: 70-72.

35.

Chase LG, Yang S, Zachar V, Yang Z, Lakshmipathy U, Bradford J, et al. Development and characterization of a clinically compliant xeno-free culture

22

medium in good manufacturing practice for human multipotent mesenchymal

23

stem cells. Stem Cells Transl Med 2012; 1: 750-758.

24

ACCEPTED MANUSCRIPT

1

36.

Gottipamula S, Muttigi MS, Chaansa S, Ashwin KM, Priya N, Kolkundkar U, et

2

al. Large-scale expansion of pre-isolated bone marrow mesenchymal stromal cells

3

in serum-free conditions. J Tissue Eng Regen Med 2013. 37.

Shanjani Y, De Croos JN, Pilliar RM, Kandel RA, Toyserkani E. Solid freeform

RI PT

4

fabrication and characterization of porous calcium polyphosphate structures for

6

tissue engineering purposes. J Biomed Mater Res B Appl Biomater 2010; 93: 510-

7

519.

9

38.

Shanjani Y, Hu Y, Toyserkani E, Grynpas M, Kandel RA, Pilliar RM. Solid freeform fabrication of porous calcium polyphosphate structures for bone

M AN U

8

SC

5

10

substitute applications: in vivo studies. J Biomed Mater Res B Appl Biomater

11

2013; 101: 972-980.

12

AC C

EP

TE D

13

25

ACCEPTED MANUSCRIPT

1

Figure Legends

2 Figure 1. Line diagram of the tissue culture protocol used for forming scaffold-free

4

multiphasic osteochondral-like constructs.

RI PT

3

5

Figure 2. Predifferentiated chondrocytes formed cartilaginous tissues on porous calcium

7

polyphosphate substrates with a zone of mineralized cartilage. (A) ALP activity of

8

predifferentiated chondrocytes cultured for 7 days with 3nM triiodothyronine (T3) or

9

100nM retinoic acid (RA) with or without dexamethasone (Dex). Mean ± 95% CI, n=3,

10

nd: not detected. * p = 0.043 between +T3 –Dex and +T3 +Dex; p = 0.045 between +T3

11

–Dex and +RA –Dex. (B) Von Kossa and toluidine blue-stained sections of cartilaginous

12

tissue formed by culturing 2×106 predifferentiated chondrocytes on the substrates with

13

3nM T3 for 3 weeks, showing mineralized zone occurring at the superficial aspect of

14

cartilage. Asterisk denotes the original location of the substrate. Scale bar = 200µm. (C)

15

Cross-sectional µCT tomographs and (D) and histology of 3-week-old constructs with

16

decreasing numbers of seeded cells: 2×106 (left), 1.5×106 (middle) and 1.0×106 (right)

17

predifferentiated chondrocytes. In (D) vertical bars denote non-mineralized cartilage

18

(purple), mineralized zone (green) and the substrate (grey). Arrows indicate the lack of

19

separation between the mineralized zone and the substrate. Scale bar = 1mm (C) and

20

200µm (D).

M AN U

TE D

EP

AC C

21

SC

6

22

Figure 3. Short term T3 treatment of predifferentiated chondrocytes was sufficient to

23

stimulate terminal differentiation. Predifferentiated chondrocytes were treated with 3nM

1

ACCEPTED MANUSCRIPT

T3 for the first 4 days. In selected cultures, T3 treatment was withdrawn after 4 days

2

(WD). (A) ALP activity during the first 4 days. * p < 0.001 between days 0-4 & 2-4. (B)

3

ALP activity of WD, compared to cells in which T3 treatment was continued (+) for up to

4

14 days. * p = 0.014. (C-J) Gene expression analysis of WD on days 4 and 21 compared

5

to non-T3-treated controls (–). * p = 0.013 (Col2a1 on day 21); p = 0.031 (Agc1 on day

6

21); p = 0.009 (Sox9 on day 21); p = 0.003 (Col10a1 on days 4 & 21); p = 0.025 (Spp1 on

7

day 4) and p = 0.002 (Spp1 on day 21). Data points are color-coded for each animal

8

throughout. Results are expressed as mean ± 95% CI, n=3.

SC

RI PT

1

M AN U

9

Figure 4. Layer of predifferentiated chondrocytes treated with T3 did not induce the

11

overlaid layer of non-treated predifferentiated chondrocytes to activate ALP activity.

12

1x106 cells were cultured on porous calcium polyphosphate substrates with (A) or

13

without (B) 3nM T3 for 4 days. On day 5, 1.5x106 cells were seeded on top of these

14

cells/tissue and cultured without T3. Cryosections of tissues from day 10 multiphasic

15

constructs were stained for ALP activity. The blue staining indicative of ALP was seen

16

only in the mineralizing layer (vertical bar). Asterisk denotes the location of where the

17

substrate would have been located prior to its removal, scale bar = 200µm.

EP

AC C

18

TE D

10

19

Figure 5. A two-step culture protocol of predifferentiated chondrocytes on porous

20

calcium polyphosphate substrates produced cartilage tissue with a mineralized zone at the

21

interface. (A) Day 21 constructs with 4-day T3 treatment were processed uncalcified,

22

sectioned and stained to reveal the cartilaginous (purple), mineral (green) and substrate

23

particles (brown). (B-I) Tissues were excised from the substrate and examined

2

ACCEPTED MANUSCRIPT

histologically for the distribution of mineral and proteoglycan accumulation (B, F), by

2

immunofluorescent staining for the accumulation of different types of collagen (C-E, G-

3

I). Constructs with T3-treated interfacial layer are denoted as +T3, and untreated (no T3)

4

control constructs are denoted as –T3. Scale bars = 200µm. (J) Differential collagen fiber

5

orientations on tissues was seen by polarized light microscopy. Scale bar = 100µm.

RI PT

1

6

Figure 6. Accumulation of extracellular matrix in T3-treated (+T3) tissues was less

8

compared to those in untreated (–T3) tissues, but comparable to native articular cartilage.

9

(A) Accumulated proteoglycan and (B) total collagen were quantified and normalized by

M AN U

SC

7

their DNA content. (A) * p = 0.001, (B) * p = 0.012 between +T3 and –T3; p = 0.037

11

between +T3 and Native. (C) Accumulation of mineral in tissue was quantified by

12

measuring the calcium and phosphate contents, normalized by tissue dry weight. ** p

Engineering of hyaline cartilage with a calcified zone using bone marrow stromal cells.

In healthy joints, a zone of calcified cartilage (ZCC) provides the mechanical integration between articular cartilage and subchondral bone. Recapitul...
3MB Sizes 0 Downloads 4 Views