Sie sind auf Seite 1von 11

Biomaterials 146 (2017) 29e39

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Bioreactor culture duration of engineered constructs inuences bone


formation by mesenchymal stem cells
Debika Mitra a, Jacklyn Whitehead a, Osamu W. Yasui a, J. Kent Leach a, b, *
a
Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
b
Department of Orthopaedic Surgery, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA

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

Article history: Perfusion culture of mesenchymal stem cells (MSCs) seeded in biomaterial scaffolds provides nutrients
Received 13 April 2017 for cell survival, enhances extracellular matrix deposition, and increases osteogenic cell differentiation.
Received in revised form However, there is no consensus on the appropriate perfusion duration of cellular constructs in vitro to
24 August 2017
boost their bone forming capacity in vivo. We investigated this phenomenon by culturing human MSCs in
Accepted 25 August 2017
Available online 6 September 2017
macroporous composite scaffolds in a direct perfusion bioreactor and compared their response to
scaffolds in continuous dynamic culture conditions on an XYZ shaker. Cell seeding in continuous
perfusion bioreactors resulted in more uniform MSC distribution than static seeding. We observed
Keywords:
Perfusion
similar calcium deposition in all composite scaffolds over 21 days of bioreactor culture, regardless of pore
Bioreactor size. Compared to scaffolds in dynamic culture, perfused scaffolds exhibited increased DNA content and
Composite scaffold expression of osteogenic markers up to 14 days in culture that plateaued thereafter. We then evaluated
Mesenchymal stem cells the effect of perfusion culture duration on bone formation when MSC-seeded scaffolds were implanted
Osteogenic differentiation in a murine ectopic site. Human MSCs persisted in all scaffolds at 2 weeks in vivo, and we observed
Bone formation increased neovascularization in constructs cultured under perfusion for 7 days relative to those cultured
for 1 day within each gender. At 8 weeks post-implantation, we observed greater bone volume fraction,
bone mineral density, tissue ingrowth, collagen density, and osteoblastic markers in bioreactor con-
structs cultured for 14 days compared to those cultured for 1 or 7 days, and acellular constructs. Taken
together, these data demonstrate that culturing MSCs under perfusion culture for at least 14 days in vitro
improves the quantity and quality of bone formation in vivo. This study highlights the need for opti-
mizing in vitro bioreactor culture duration of engineered constructs to achieve the desired level of bone
formation.
2017 Elsevier Ltd. All rights reserved.

1. Introduction implantable osteogenic grafts, biomaterial scaffolds are commonly


seeded with bone-forming cells and cultured in vitro under variable
Large bone decits due to trauma, surgery, or resulting from slow conditions and durations. Compared to static or continuous dy-
and non-healing defects represent a signicant clinical problem. namic culture, scaffolds under continuous perfusion culture exhibit
More than 1 million bone repair surgeries are performed annually, improved cell growth and survival, extracellular matrix (ECM)
bringing the nancial burden to over $5 billion in the Unites States deposition, and enhanced osteogenic differentiation of mesen-
alone [1,2]. Autograft bone is the current gold standard for repairing chymal stem cells (MSCs) due to increased nutrient availability and
non-healing bone defects, yet its use has numerous drawbacks such application of mechanical stimulation [5e8]. However, the effect of
as donor site morbidity, prolonged pain, limited availability, and bioreactor culture duration in vitro on the bone forming capacity of
increased risk of infection [3,4]. Tissue engineered bone constructs MSCs upon transplantation in vivo is largely unknown.
represent a promising alternative to autografts. To generate An ideal bone scaffold for bioreactor culture of MSCs must be
osteoconductive, providing a structure and network that supports
cell attachment, growth, and osteogenic differentiation [9,10]. Com-
posite scaffolds formed of hydroxyapatite (HA) and poly(lactide-co-
* Corresponding author. University of California, Davis, Department of Biomedical glycolide) (PLG) offer tunable biodegradability, osteoconductivity,
Engineering, 451 Health Sciences Drive, Davis, CA 95616, USA.
E-mail address: jkleach@ucdavis.edu (J.K. Leach).
porosity, and mechanical properties [11e14]. Moreover, HA-PLG

http://dx.doi.org/10.1016/j.biomaterials.2017.08.044
0142-9612/ 2017 Elsevier Ltd. All rights reserved.
30 D. Mitra et al. / Biomaterials 146 (2017) 29e39

scaffolds can regulate osteogenesis and trophic factor secretion by cabinet. Prior to cell seeding, scaffolds were soaked in cell culture
MSCs in vitro and in vivo [11]. We and others reported that the pore medium for at least 30 min to facilitate cell adhesion.
size of three-dimensional macroporous scaffolds can regulate cell
differentiation under static or dynamic conditions [15e19]. Pore size 2.2. Scanning electron microscopy
also directly inuences the shear stress experienced by cells when
scaffolds are maintained under continuous perfusion, thus regulating To visualize gross morphology and pore architecture, scaffolds
osteogenic differentiation of MSCs [10,17]. Therefore, selecting the were gold coated using a sputter coater (Desk II; Denton Vacuum,
appropriate pore size for constructs cultured in perfusion bioreactors Moorestown, NJ) and imaged using scanning electron microscopy
is vital for bone tissue engineering applications. (Hitachi 3500-N, Hitachi Science Systems Ltd, Tokyo, Japan) at 5 kV.
While the exposure of osteoblasts and osteoprogenitor cells to Pore diameter was measured in ImageJ (National Institutes of
uid shear stress enhances their bone forming potential in vivo Health, Bethesda MD). Briey, pores were measured edge to edge
[6,20], the contribution of perfusion culture duration to bone for- along the maximum and minimum axis and averaged to generate a
mation has not been thoroughly investigated. Furthermore, the representative diameter. Eight pores from four separate images per
effect of culture duration on the bone-forming potential of human group were measured to determine pore diameter.
MSCs, a more clinically relevant cell source compared to osteo-
blasts, has not been reported in the literature. Bioreactor culture 2.3. Measurement of uid permeation velocity through scaffolds
durations ranging from 5 days [21] to 5 weeks [22e24] have been
used for creating implantable tissue engineered bone. Shorter Permeation velocity of uid ow through the scaffolds was
culture durations may be sufcient to prime MSCs towards osteo- measured as an indicator of pore connectivity and indirect measure
genic differentiation and boost their proangiogenic potential [25]. of uid shear stress (Fig. 1D, insert). Prior to analysis, scaffold height
Extended culture durations such as 2e5 weeks may result in a more was measured using digital calipers. Pristine, acellular scaffolds
mature construct with increased cellularity, differentiated cells and were held in place between two bioreactor adaptors housed be-
a dense ECM, which can act as a scaffold for host cell inltration and tween two rubber syringe stoppers in a syringe barrel to create a
differentiation [10,26,27]. Thus, there is a critical gap in our seal preventing uid ow around the scaffold. The syringe was
knowledge for the appropriate in vitro perfusion culture duration to lled with media, and the volume that passed through the scaffold
maximize bone formation in vivo with osteogenic grafts. was recorded. Permeation velocity was calculated using a deriva-
We hypothesized that HA-PLG scaffolds would serve as effective tive of Darcy's Law [16].
osteoconductive biomaterials to promote MSC osteogenic differ-
entiation and that longer culture durations would result in mature 2.4. Cell culture
constructs suitable for implantation. We investigated the role of
perfusion on bone formation by culturing human MSCs in porous Human bone marrow-derived MSCs (Lonza, Walkersville, MD)
HA-PLG scaffolds of varying pore sizes and durations in a contin- were expanded without further characterization in growth me-
uous perfusion bioreactor. Thus, the goals of this study were to 1) dium (GM) consisting of minimum essential alpha medium (a-
determine the effect of pore size of HA-PLG scaffolds on MSC MEM; Invitrogen, Carlsbad, CA) supplemented with 10% fetal
osteogenic differentiation in vitro; 2) characterize the osteogenic bovine serum (FBS; Atlanta Biologicals, Flowery Branch, GA) and 1%
response of MSCs in HA-PLG scaffolds when cultured in perfusion penicillin/streptomycin (Gemini Bio-Products, Sacramento, CA).
bioreactors for up to 21 days; and 3) investigate the effect of in vitro Cells were cultured under standard conditions (37  C, 21% O2, 5%
perfusion culture duration on in vivo bone forming capacity of CO2) and utilized at passages 4e5. For all experiments, cells were
tissue engineered constructs in an ectopic site. initially seeded in GM and then exposed to osteogenic medium
(OM: GM supplemented with 10 mM b-glycerophosphate, 50 mg/
2. Materials and methods mL ascorbate-2-phosphate, and 100 nM dexamethasone). Media
was changed every 2e3 or 3e4 days for dynamic and bioreactor
2.1. Scaffold preparation cultures, respectively.

Scaffolds were fabricated using a gas foaming/particulate leach- 2.5. Cell seeding and maintenance
ing method as described [11,16]. Briey, microspheres composed of
PLG (85:15, DLG 7E; Lakeshore Biomaterials, Birmingham, AL) were For characterizing the effect of pore size on cell seeding efciency,
prepared using a double-emulsion process and lyophilized to form a cells were seeded statically or in perfusion bioreactors (Fig. 2A).
free-owing powder. 9.2 mg of lyophilized microspheres were Scaffolds were soaked in medium for 30 min before seeding. For
combined with 23.1 mg of HA crystals (particle size 100 nm, Berkeley static seeding, 1.2  106 MSCs were suspended in 75 mL GM and
Advanced Biomaterials, Berkeley, CA) and 175.6 mg of NaCl to yield a applied dropwise to scaffolds. MSCs were allowed to attach for 3 h
2.5:1:19 mass ratio of ceramic:polymer:salt. Porogen diameter before transferring scaffolds into well plates with GM. For bioreactor
ranges were achieved by passing salt crystals through sieves of seeding, scaffolds were installed in U-CUP ow perfusion bioreactors
distinct size ranges (small: 125e300 mm, medium: 300e500 mm, and (Cellec Biotek, Basel, Switzerland) and 10 mL of GM was injected
large: 500e850 mm). Control PLG scaffolds were fabricated without through the bottom port into the bioreactor (Fig. 1A). 1.2  106 MSCs
HA using 250e425 mm salt crystals, as this is the range currently suspended in 2 mL GM were injected via the top port into the
employed by our group and others to support cellular ingrowth bioreactor, bringing the nal media volume in each bioreactor to a
[11,28e31]. The powdered mixture was then compressed for 1 min total of 12 mL. Up to 10 individual bioreactors were then connected
into solid disks (nal dimensions: 8 mm in diameter and 2 mm in to a syringe pump (Harvard Apparatus, Holliston, MA) to maintain
height) using a Carver Press (Carver, Inc., Wabash, IN). Compressed media at a supercial velocity of 3 mL/min for 15e18 h.
disks were exposed to high pressure CO2 gas (5.5 MPa) for at least For all in vitro studies, scaffolds were seeded using bioreactors to
16 h followed by rapid pressure release to cause polymer fusion. NaCl ensure homogenous cell distribution for all experimental groups.
particles from the scaffolds were then leached in distilled H2O for After the seeding phase (15e18 h), GM was replaced with OM, and
24 h. Scaffolds were sterilized by 70% ethanol under gentle vacuum constructs were maintained under continuous perfusion culture for
followed by two rinses in sterile PBS and dried in a sterile biosafety up to 21 days. For culture under dynamic conditions, scaffolds were
D. Mitra et al. / Biomaterials 146 (2017) 29e39 31

Fig. 1. Bioreactor setup and permeation velocity of media through scaffolds. (A) Scaffolds are held in place by two adaptors that prevent ow of media around scaffolds. Cell
suspension and media are injected in the top and bottom port of bioreactors, respectively. (B) Scaffold pore morphology as observed by scanning electron microscopy. (C) Average
pore diameter measured from SEM images. (D) Permeation velocity of media through scaffolds of varying pore size and image of experimental setup. Signicance is denoted by
alphabetical letterings; groups with signicance do not share the same letters (n 4 for pore diameter, n 3 for permeation velocity).

removed from bioreactors after the seeding phase, placed in 24 well for cDNA synthesis. 250 ng of RNA was used in reverse transcription
plates with 500 mL OM, and maintained on an XYZ shaker (The Belly reactions. qPCR was performed using a QuantiFast Probe PCR kit
Button Shaker, IBI Scientic, Peosta, IA) at 30 rpm with the platform (Qiagen) on a CFX96 Real-Time System (manufacturer). Primers and
having a maximum tilt angle of 4.5 . The shaker was kept in an probes for RPL13 (HS00204173_m1), SP7 (HS01866874_S1), COL1A1
incubator under standard conditions and was continuously in (HS00164004_M1), and IBSP (HS00173720_M1) were purchased
motion to facilitate media transport throughout the scaffolds. from Applied Biosystems (Foster City, CA). Amplication conditions
were 95  C for 3 min, followed by 40 cycles at 95  C for 3 s and 60  C
2.6. MTT staining of seeded scaffolds for 30 min. Quantitative PCR results were normalized to RPL13
transcript levels to yield DCt, and fold change in expression relative to
Scaffolds were stained with 0.5 mg/mL thiazolyl blue tetrazo- the housekeeping gene was calculated using 2eDCt. The remainder of
lium bromide (MTT) solution in PBS at 37  C. After 2 h, MTT solution the scaffold was collected in 250 mL of passive lysis buffer (Promega,
was aspirated, scaffolds were washed twice with PBS, and photo- Madison, WI), frozen at 20  C, thawed, sonicated, centrifuged at
graphs were taken using a digital camera. 5000 rpm for 10 min to pellet the cell debris, and the supernatant was
collected for DNA analysis. The supernatant was analyzed for DNA
2.7. In vitro osteogenic differentiation content using the Quant-iT PicoGreen dDNA Assay Kit (Invitrogen)
following manufacturer's instructions. The remaining pellet was
Scaffolds were retrieved from bioreactors or well plates and resuspended in 0.9 N H2SO4 and incubated overnight at 37  C. Cal-
washed twice with PBS. A 5 mm biopsy punch (Integra Miltex, York, cium deposition was quantied by reacting with o-cresolphthalein
PA) was used to harvest a disk for qPCR analysis as described [32]. complexone as previously described [33,34]. The calcium content of
Samples were collected in TRIzol reagent (Invitrogen, Carlsbad, CA) an unused composite scaffold was subtracted from that of cultured
following the manufacturer's protocol. Total RNA was isolated using composite scaffolds to determine the amount of calcium deposited by
an RNEasy micro kit (Qiagen, Valencia, CA). RNA was reverse tran- MSCs.
scribed with the QuantiTect Reverse Transcription kit (Qiagen). RNA To visualize cell distribution and markers of osteogenic differ-
purity was examined with a spectrophotometer (NanoDrop 1000, entiation within constructs, samples were washed twice in PBS and
Thermo Fisher Scientic, Waltham, MA), and only samples with high xed in 10% buffered formalin acetate (Fisher Scientic, Fair Lawn,
quality (A260/280 > 1.6 per manufacturer's instructions) were used NJ) for 24 h at 4  C. Samples were then washed twice in PBS to
32 D. Mitra et al. / Biomaterials 146 (2017) 29e39

Fig. 2. Cell distribution in HA-PLG scaffolds of varying pore sizes seeded under static or ow conditions. (A) MSCs were seeded on scaffolds under ow perfusion or statically
by pipetting cells directly on top of scaffolds. (B) MTT staining to visualize MSC distribution throughout scaffolds. Diffuse staining and bright purple staining are indicative of well-
distributed and locally concentrated cells, respectively. Scale bar represents 5 mm.

remove residual formalin acetate and preserved in 70% ethanol at (lower left), and acellular (lower right) constructs. Unlike constructs
4  C until further processing. Prior to cryosectioning, samples were cultured under perfusion, which were maintained in OM after the
soaked in Tissue-Tek OCT compound (Sakura Finetek, Torrance, CA) seeding phase, the acellular group consisted of pristine scaffolds that
overnight to allow OCT to perfuse through the entire scaffold. were soaked in GM for 30 min prior to implantation. Animals were
Constructs were then placed in fresh OCT and frozen on dry ice. euthanized at 2 or 8 weeks, and scaffolds were collected and xed in
5 mm sections were cut on a Leica CM1850 Cryostat (Leica Micro- 10% buffered formalin acetate for 2 days at room temperature.
systems, Bannockburn, IL) and mounted onto microscope slides for Samples were then washed twice in PBS to remove residual formalin
analysis. Sections were stained using hematoxylin and eosin (H&E) acetate and preserved in 70% ethanol at 4  C until further processing.
following standard protocols and immunostained using an anti- Qualitative and quantitative three-dimensional analysis of ex-
osteocalcin (OCN) antibody (1:200, AB13420, Abcam, Cambridge, plants at 8 weeks was conducted using microCT [35]. Explants were
MA). The percentage area positive for OCN was quantied by imaged (55 kVp, 145 mA, 300 ms integration time, average of 3
creating a mask for brown stain in Photoshop and then calculating images) using a high-resolution mCT specimen scanner (mCT 35,
percentage of total area (dened by a box drawn around the ex- Scanco Medical; Brttisellen, Switzerland). Contiguous slices of
tremes of the scaffold) in ImageJ. 2048  2048 pixels were imaged with 12 mm resolution and slice
thickness (voxels). Serial tomograms were reconstructed from raw
2.8. Murine subcutaneous implantation model data of 1000 projections per 180 using a cone beam ltered back
projection algorithm [36]. The tomograms were calibrated to 0.0,
Treatment of experimental animals was in accordance with UC 99.6, 200.0, 401.0 and 800.3 mg HA/cc concentrations of HA so that
Davis animal care guidelines and all National Institutes of Health grey-values of the images were converted to units of density in mg
animal handling procedures. Male and female eight-week-old non- HA/cc. The entire scaffold was analyzed by contouring to its edges
obese diabetic/severe combined immunodecient gamma (NSG, based on a threshold of 256 mg HA/cc. Material in the recon-
NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mice (Jackson Laboratories, West structed images was partitioned by a threshold of 256e3000 mg
Sacramento, CA) were anesthetized and maintained under a 2% HA/cc to discriminate between mineralized and unmineralized
isourane/O2 mixture delivered through a nose cone. Mice exhibited tissue. After thresholding, the image noise was reduced using a low
no diabetogenic effects, as streptozotocin was not administered. pass Gaussian lter (s 0.8, support 1). Bone volume fraction
Each animal received four subcutaneous implants: constructs (BV/TV) was determined by dividing the number of pixels repre-
cultured under perfusion for 1 (upper left), 7 (upper right), or 14 days senting bone tissue (BV: bone volume) by the number of pixels in
D. Mitra et al. / Biomaterials 146 (2017) 29e39 33

the cylindrical segment (TV: total volume). After microCT analysis, indicator of cell number, among composite scaffolds of different
explants were demineralized in Calci-clear (National Diagnostics, pore sizes over 21 days in continuous perfusion culture (Fig. 3A), yet
Atlanta, GA) for up to 48 h, washed twice in PBS, bisected, parafn- scaffolds with small pores had greater DNA content compared to
embedded and sectioned at 5 mm thickness. Blood vessel density at PLG controls. H&E staining revealed relatively uniform cell distri-
2 weeks was quantied using H&E stained cross-sections and bution within scaffolds of all pore sizes. (Fig. 3B). Quantitative
consisted of counting circular structures with well-dened lumens assessment of calcium deposition revealed that composite scaffolds
containing erythrocytes. 2-week tissue sections were also immu- of all sizes had signicantly higher amounts of calcium compared to
nostained using a human-specic anti-Lamin A/C antibody (1:250, PLG controls (Fig. 3C). No signicant differences in calcium depo-
Abcam, Cambridge, MA) to detect the presence of human MSCs. At sition were observed among scaffolds of different pore sizes.
8 weeks, H&E and Masson's trichrome stains of tissue sections were Qualitatively, we observed similar osteocalcin (OCN) expression
used to visualize tissue ingrowth and collagen distribution within within pores of scaffolds with medium and large pores (Fig. 3D).
scaffolds, respectively. Sections were also immunostained for bone Scaffolds with small pores were also brittle and difcult to handle
sialoprotein (BSP) using a human-specic anti-bone sialoprotein after culture, rendering them unsuitable for implantation. The thin
antibody (1:200, Abcam). pore walls of scaffolds with small pores corroborated this gross
observation (Fig. 1B). Subsequent studies were performed with
2.9. Statistical analysis scaffolds of medium pore size due to similar levels of OCN
expression and calcium content to large pore size scaffolds, ease of
All data are presented as means standard deviation of the handling, and the pore size range being closest to that used by our
mean. Statistical analysis was performed using Student's t-tests, group and others.
one-way ANOVA and two-way ANOVA with Tukey's multiple
comparison post-hoc test, where applicable. All statistical analysis 3.4. Osteogenic response of MSC-seeded constructs under perfusion
was performed in Prism 6 software (GraphPad, San Diego, CA), and culture increases with time
p values less than 0.05 were considered statistically signicant.
Signicance is denoted by alphabetical letterings; groups with no We evaluated the role of continuous dynamic or perfusion cul-
signicance are linked by the same letters, while groups with sig- ture on the osteogenic response of MSCs seeded on medium pore-
nicance do not share the same letters. sized HA-PLG scaffolds (Fig. 4A). We observed a steady increase in
DNA content from 1 to 14 days for scaffolds cultured in perfusion
3. Results bioreactors (Fig. 4B), yet there was no substantial increase in DNA
content beyond 14 days. We did not detect increases in DNA content
3.1. Porogen size increases pore diameter and permeation velocity within scaffolds under dynamic culture over time. H&Eestained
sections corroborate DNA quantication and demonstrate increases
SEM images reveal appreciable differences in pore morphology in cell number with time for bioreactor samples (Fig. 4C). Scaffolds
of scaffolds. PLG scaffolds as well as HA-PLG scaffolds with medium in dynamic culture exhibited spatial restriction of cells to the top
and large pores exhibited a uniform honeycomb pattern (Fig. 1B). surface, which did not increase appreciably over time. In contrast,
Scaffolds with small pores appeared collapsed throughout the scaffolds in perfusion culture became more cellularized over time,
scaffold. The average pore diameter was in agreement with the which was visible throughout the entire scaffold.
range of porogen particles used for each group (Fig. 1C). We also We assessed the osteogenic potential of MSCs by measuring
assessed the permeation velocity of media through scaffolds of gene expression of osteogenic markers and functional outputs such
different pore sizes as an indirect measure of the shear stress as calcium deposition (Fig. 4D) and osteocalcin expression. SP7
experienced by MSCs cultured under ow perfusion (Fig. 1D). We expression, an early to mid-transcription factor involved in MSC
measured increasing permeation velocity with increasing pore size. differentiation, was signicantly higher in bioreactor samples
However, this effect tapered between scaffolds with medium and compared to dynamic samples at 7 days, which was reduced in
large pores. Scaffolds with large pores exhibited signicantly higher both experimental groups at 14 and 21 days in culture. COL1A1
permeation velocity relative to those with small pores or PLG expression was also signicantly higher in bioreactor samples
controls. relative to dynamic samples at 7 days and followed the same trend
as SP7. Bone sialoprotein (IBSP), a late marker of osteogenic differ-
3.2. Pore size differentially affects cell seeding efciency under entiation, exhibited increased expression at 14 and 21 days in both
static and ow conditions groups relative to 7 days. Dynamic samples exhibited a signicant
increase in IBSP expression over bioreactor samples at 21 days.
Scaffolds seeded under perfusion exhibited more homogeneous MSCs in bioreactor culture deposited more calcium than those in
cell distribution throughout the scaffold than statically seeded dynamic culture at 14 and 21 days. We observed a trend for
scaffolds (Fig. 2B). Although scaffolds with small pores had fewer increasing calcium deposition for bioreactor samples over time,
cells on the underside than scaffolds with medium and large pores, although this trend was not signicant. Qualitative histological
there was no appreciable qualitative difference among scaffolds assessment and quantication of these images conrmed trends
seeded in bioreactors. For all pore sizes and the PLG control, scaf- observed in DNA content and osteogenic gene expression. OCN
folds seeded statically exhibited strong staining on only one side of expression, a late marker of osteogenic differentiation, was more
the scaffold, indicating that cells primarily adhered to the top of the uniform in bioreactor samples compared to dynamic samples
scaffold. With increasing pore size, more cells were detected near (Fig. 4E). OCN expression increased in bioreactor samples from 7 to
the center of the scaffold but the underside of the scaffold remained 14 days, but we observed minimal increase in OCN expression
largely devoid of cells. beyond 14 days. Similar to cell distribution, OCN was spatially
restricted to the top surface of the scaffold in dynamic samples.
3.3. Pore size of composite scaffolds does not alter MSC response Given that 21 days of in vitro culture did not confer signicant
under perfusion advantages to the engineered constructs over a 14 day culture
period, we selected 14 days as our longest culture duration for
We did not detect signicant differences in DNA content, an in vivo studies.
34 D. Mitra et al. / Biomaterials 146 (2017) 29e39

Fig. 3. Pore diameter does not inuence DNA content or osteogenic differentiation of MSCs on HA-PLG scaffolds under perfusion culture. (A) DNA content. (B) Representative
H&E images of scaffolds with increasing pore diameter. (C) Quantication of calcium deposition in MSC-seeded constructs after 21 days. (D) Representative immunohistochemistry
for osteocalcin. Black is indicative of scaffold (S); brown is indicative of osteocalcin (OCN) and staining is highlighted by blue arrows. Signicance is denoted by alphabetical
letterings; groups with signicance do not share the same letters (n 5). Scale bar represents 500 mm. (For interpretation of the references to colour in this gure legend, the reader
is referred to the web version of this article.)

4. 14 days of perfusion culture in vitro enhances MSC density (BMD) in all cellular scaffolds, regardless of bioreactor culture
persistence and bone formation in vivo duration (Fig. 6BeC). Constructs perfused for 14 days exhibited
signicantly greater BVF and BMD relative to 1 day constructs. In
The bone forming potential of perfusion-cultured scaffolds for agreement with microCT data, H&E staining revealed that dense
increasing durations was evaluated in a murine ectopic site connective tissue was visibly greater in scaffolds cultured in vitro for
(Fig. 5A). Human MSCs persisted in all three cellular groups upon 14 days relative to other groups (Fig. 6D, top; Supplementary Fig. 2).
explantation at 2 weeks (Fig. 5B). Explanted scaffolds from all four Acellular groups were largely devoid of connective tissue on one side
groups appeared well-vascularized 2 weeks after explantation and contained fewer nuclei on the other side relative to cellular
(Fig. 5C). Quantication of blood vessel density revealed a trend for groups. Masson's trichrome staining conrmed the presence of
increased neovascularization in constructs cultured for 7 days vs. 1 denser collagen with increasing bioreactor culture duration, with 14
day constructs (Fig. 5D, top). Although signicant differences were day constructs exhibiting the most intense collagen staining
not detected within the full cohort of animals, vascular density was compared to other cellular groups (Fig. 6D, center; Supplementary
signicantly different for constructs cultured for 1 and 7 days in Fig. 2). Acellular explants contained infrequent, small areas of
animals of the same gender (Fig. 5D, bottom). Furthermore, the collagen. Immunohistochemistry for human bone sialoprotein (BSP)
magnitude of vascularization was greater in all scaffolds in male revealed positive staining in all cellular scaffolds (Fig. 6D, bottom;
mice compared to female mice. Supplementary Fig. 2). Constructs cultured for 14 days exhibited
At 8 weeks post-implantation, we observed human MSCs in all larger areas of positive BSP staining compared to 1 and 7 day
cellular constructs, with the most cells present in constructs cultured constructs.
under perfusion for 14 days (Supplementary Fig. 1). Qualitatively,
fewer human cells were present in explants 8 weeks post- 5. Discussion
implantation compared to 2 week explants (Fig. 5B, Supplementary
Fig. 1). Acellular scaffolds grossly exhibited distinct scaffold bound- Perfusion bioreactors provide a three-dimensional environment
aries and appeared the least remodeled compared to all cellular with mechanical stimulation via uid ow for engineering osteo-
scaffolds, and these differences were observable using microCT genic grafts for bone repair. In this study, we sought to determine
(Fig. 6A). Constructs cultured for 14 days appeared smaller in diam- the appropriate pore size of HA-PLG scaffolds for promoting MSC
eter than constructs cultured for 1 and 7 days, and acellular scaffolds osteogenic differentiation and the role of continuous perfusion
were the largest. Compared to acellular scaffolds, we detected sig- culture duration to generate osteogenic grafts. Larger pore sizes
nicant increases in bone volume fraction (BVF) and bone mineral resulted in more uniform cell seeding and distribution, but we did
D. Mitra et al. / Biomaterials 146 (2017) 29e39 35

Fig. 4. DNA content and osteogenic differentiation of MSCs on composite scaffolds plateau after 14 days of bioreactor culture. (A) MSCs were seeded in HA-PLG scaffolds
under perfusion and cultured under dynamic or perfusion conditions for up to 21 days. (B) DNA content. (C) Representative H&E images. Scaffold (S) is denoted by black arrow.
Dashed line demarcates the boundary of spatial restriction of cells within a scaffold. (D) Osteogenic gene expression and calcium deposition. (E) Representative immunohisto-
chemistry images for osteocalcin and percentage area positive for osteocalcin. Black is indicative of scaffold (S) and denoted by grey arrow; brown is indicative of osteocalcin (OCN)
and denoted by a green arrow. Dashed line demarcates the boundary of spatial restriction of cells within a scaffold. Signicance is denoted by alphabetical letterings; groups with
signicance do not share the same letters (n 5 for DNA and n 4 for osteogenic assays). Scale bar represents 500 mm. (For interpretation of the references to colour in this gure
legend, the reader is referred to the web version of this article.)

not detect differences in calcium accumulation under perfusion as a stress on cells attached to the pores of scaffolds. Fluid shear stress
function of pore diameter. Compared to PLG controls, HA-PLG magnitudes between 0.1 and 0.3 dyn/cm2 are considered stimula-
scaffolds enhanced the osteogenic differentiation of MSCs. Con- tory for osteoblastic cells in ow perfusion bioreactors [38e40]. The
structs cultured under continuous perfusion exhibited increased ow rate used in this study (3 mL/min) was optimized to reect a
DNA content, cell distribution, and osteogenic markers compared to range of uid shear stress at the time of cell seeding (small pores:
their dynamic counterparts, yet this favorable cell response to 0.35 dyn/cm2, medium pores: 0.2 dyn/cm2, large pores: 0.15 dyn/
perfusion plateaued after 14 days. Upon implantation into an ectopic cm2). Shear stress was calculated from the ow rate, pore diameter,
site, constructs cultured in perfusion for up to 14 days resulted in and viscosity of media using previously established methods and
signicantly greater bone formation. These data demonstrate that assuming uniform pore interconnectivity [22]. These values reect
optimizing the bioreactor culture duration in vitro can enhance the only a rst approximation of shear stress due to limited pore
capacity of MSC-seeded HA-PLG constructs to contribute to bone interconnectivity [41] and cell secretion of ECM during prolonged
formation in vivo. culture [38], thereby reducing conduit diameters for uid ow and
Perfusion culture promotes homogenous cell distribution potentially increasing shear stress.
throughout the construct [37] and efcient transport of critical Common synthetic biomaterials under investigation for biore-
nutrients by forcing media through the pores of the scaffold. actor culture include titanium ber mesh [20], polyurethane [42], b-
Scaffold pore size was directly associated with seeding efciency of tricalcium phosphate [43], PLG scaffolds [44], and HA scaffolds
statically seeded samples, where cells inltrated farther into scaf- [21,45]. We previously reported that HA-PLG composite scaffolds
folds with larger pore sizes. We observed a similar trend in scaffolds (pore size 250e425 mm) enhance osteogenesis and trophic factor
seeded under perfusion, though this effect was less pronounced. secretion of human MSCs in static culture compared to PLG scaffolds
Direct perfusion of media through scaffolds also exerts uid shear [11]. In the present study, we demonstrate MSC differentiation is
36 D. Mitra et al. / Biomaterials 146 (2017) 29e39

Fig. 5. MSC persistence and scaffold neovascularization is inuenced by duration of perfusion culture. (A) Subcutaneous implantation of constructs maintained in perfusion
culture for 1, 7, and 14 days. (B) Human cells are observed in representative immunohistochemistry images for Lamin A/C in constructs 2 weeks post-implantation. Black arrows
denote positive staining. (C) H&E staining of explants reveals scaffold neovascularization. White arrows denote blood vessels. (D) Quantication of blood vessel density in scaffolds
retrieved from all animals (top) and as a function of gender (bottom) (n 5e6 or n 2e3 for males and females, respectively). *p < 0.05 and **p < 0.01. Scale bar represents 100 mm.

enhanced in HA-PLG scaffolds relative to PLG scaffolds cultured in shear stress. Since ow rate was constant among all experimental
perfusion bioreactors. While composite scaffolds using HA and groups, one might postulate that mass transport had a stronger ef-
polycaprolactone have been employed in perfusion bioreactors [46], fect on cell function than shear stress in this study. This phenome-
this is the rst demonstration of the synergistic effect of osteo- non merits further investigation using composite scaffolds with
conductive HA-PLG scaffolds and direct perfusion stimulation of consistent pore size and geometry. Others who modulate pore size,
MSCs. Contrary to our hypothesis, we did not detect increases in distribution, and interconnectivity using additive manufacturing to
DNA content or MSC differentiation with increases in pore diameter regulate geometry reported the favorable effect of a porosity
when cultured under continuous perfusion. Previous ndings from gradient within scaffolds on MSC osteogenic differentiation
our group and others reported that larger pore sizes improve MSC [50e53].
proliferation and differentiation towards the osteogenic and chon- Upon establishing an appropriate pore size, we cultured MSCs in
drogenic lineages under static or dynamic culture [15,16,47,48]. bioreactors or dynamic conditions for up to 21 days. While some
However, mass transport limitations are mitigated under ow groups consider short bioreactor culture durations (5 days) suf-
conditions, enabling comparable cell attachment and proliferation cient prior to implantation [21], others use moderate durations
in scaffolds of varying sizes. Numerous studies demonstrate that (10e21 days) [5,26,45,54e57] that are pulsatile or intermittent
MSC osteogenic differentiation is enhanced in scaffolds with smaller [58,59], or up to 5 weeks to generate more mature constructs
pores, likely due to increased shear stress [10,17,38,49]. However, [22e24]. To minimize delays between harvesting autologous cells
Holtorf et al. reported that larger pore size promoted early differ- and generating an osteogenic graft, we characterized MSC response
entiation, while smaller pore size was associated with later differ- under perfusion in vitro to select the shortest culture duration
entiation and ECM deposition, conrming the temporal effect of necessary for construct maturity. In agreement with other reports
perfusion culture on osteogenic differentiation [17]. Scaffolds [6], DNA content and osteogenic differentiation markers increased
formed by the gas foaming method possess macropores with up to 14 days and plateaued, motivating our exclusion of constructs
limited interconnectivity, which are supported by numerous mi- cultured for longer periods in vitro. When comparing constructs
cropores that facilitate nutrient transport and may alter permeation cultured under dynamic and perfusion conditions, we observed
velocity of media through scaffolds. Furthermore, scaffolds contain a higher IBSP mRNA expression in dynamic samples. This could be
distribution of pore sizes, which may mask subtle differences in attributed to accelerated differentiation in bioreactor samples.
D. Mitra et al. / Biomaterials 146 (2017) 29e39 37

Fig. 6. MSC-seeded HA-PLG constructs maintained under ow perfusion for 14 days in vitro result in the greatest bone formation in vivo. (A) Representative microCT images
of explants at 8 weeks post-implantation. Scale bar represents 1 mm. (B) Bone volume fraction and (C) bone mineral density are greatest in 14-day cultured constructs compared to
1 day constructs or acellular controls. (D) Representative H&E images (top) reveal increased tissue ingrowth and density. Masson's trichrome images (center) exhibit greater
collagen content and density with increases in culture duration. Dashed line demarcates areas with distinct levels of connective tissue and cellularization. Representative
immunohistochemistry images for bone sialoprotein (BSP) (bottom). Blue arrows indicate pore walls while green arrows mark positive staining for BSP. Scale bar represents 250 mm.
Signicance is denoted by alphabetical letterings; groups with signicance do not share the same letters (n 6). (For interpretation of the references to colour in this gure legend,
the reader is referred to the web version of this article.)

Dynamic constructs may be lagging behind in the osteogenic dif- function of perfusion culture duration in vitro. Constructs cultured
ferentiation timeline, and peak IBSP expression in bioreactor sam- for 1 day contain primarily undifferentiated MSCs. While constructs
ples could have been missed by only examining days 7, 14, and 21. in perfusion culture for longer durations contain more MSCs, cells
Although two-week culture durations in bioreactors are common are further differentiated toward the osteoblastic lineage. We pre-
for generating osteogenic grafts [6,45,55,57,60], these data, specif- viously demonstrated in monolayer culture that MSCs secrete more
ically changes in IBSP gene expression and calcium secretion, proangiogenic factors when less committed to the osteoblastic
highlight the role of temporal changes in MSC differentiation under phenotype [25,61]. Thus, it is not surprising that constructs
perfusion culture. cultured for 7 days exhibited a trend for greater vessel density than
Next, we evaluated the in vivo bone-forming potential of 14 day constructs. These data are in accordance with our previous
perfusion-cultured osteogenic grafts. To our knowledge, this is the nding of lower vessel density in scaffolds containing osteogeni-
rst evaluation of human MSCs osteoinduced on macroporous cally induced MSCs versus undifferentiated cells [62]. Acellular
scaffolds for varying culture durations in a continuous perfusion scaffolds were soaked in growth media for a brief period, much less
bioreactor. Sikavitsas et al. demonstrated that the stage of osteo- than those in perfusion culture exposed to osteogenic media. Thus,
blastic differentiation of rat bone marrow stromal osteoblasts in their osteogenic potential may be due in part to different treatment
perfusion culture inuences the performance of engineered con- regimens. The ndings in this study suggest a combined effect of
structs in vivo [20]. Yeatts et al. reported that human MSCs cultured more cells due to prolonged culture duration possessing increased
in tubular perfusion bioreactors for 10 days demonstrated signi- osteogenic potential but with blunted proangiogenic potential. We
cantly increased bone formation compared to statically cultured or also observed signicant increases in vascular density for con-
acellular scaffolds [56]. Although tubular perfusion bioreactors structs cultured for 7 days versus 1 day within each gender,
enable culture of numerous scaffolds simultaneously, scaffolds emphasizing the need to consider gender-related effects on the
were not cultured under direct perfusion to force media through proangiogenic nature of MSCs. At 8 weeks post-implantation, we
the full scaffold volume, and no other time points were examined. detected a small number of MSCs in all cellular scaffolds, suggesting
In this study, we observed changes in vascular density in vivo as a that human MSCs persist in this model for extended durations. In
38 D. Mitra et al. / Biomaterials 146 (2017) 29e39

agreement with our in vitro ndings, constructs cultured for 14 References


days, but not 7 days, exhibited signicantly greater bone formation
in an ectopic site. Thus, constructs with higher DNA content and [1] A.J. Salgado, O.P. Coutinho, R.L. Reis, Bone tissue engineering: state of the art
and future trends, Macromol. Biosci. 4 (8) (2004) 743e765.
human MSCs at a later stage of differentiation at the time of im- [2] J.D. Kretlow, A.G. Mikos, Review: mineralization of synthetic polymer scaffolds
plantation contribute to more robust bone formation in vivo. Chen for bone tissue engineering, Tissue Eng. 13 (5) (2007) 927e938.
et al. also reported that perfusion culture of macrotissues for 14 [3] E.D. Arrington, W.J. Smith, H.G. Chambers, A.L. Bucknell, N.A. Davino, Com-
plications of iliac crest bone graft harvesting, Clin. Orthop. Relat. Res. (329)
days led to more homogeneously distributed bone matrix than 7 (1996) 300e309.
days [63]. However, our data are contradictory to the ndings of [4] J.G. Seiler 3rd, J. Johnson, Iliac crest autogenous bone grafting: donor site
Sikavitsas et al. in which bone formation in an orthotopic defect complications, J. South Orthop. Assoc. 9 (2) (2000) 91e97.
[5] G.N. Bancroft, V.I. Sikavitsas, J. van den Dolder, T.L. Shefeld, C.G. Ambrose,
was enhanced in constructs containing stromal osteoblasts at an J.A. Jansen, A.G. Mikos, Fluid ow increases mineralized matrix deposition in
early stage of differentiation [20]. These differences may relate to 3D perfusion culture of marrow stromal osteoblasts in a dose-dependent
the in vivo models used, the biomaterials employed, or species manner, Proc. Natl. Acad. Sci. U. S. A. 99 (20) (2002) 12600e12605.
[6] Y. Wang, T. Uemera, J. Dong, H. Kojima, J. Tanaka, T. Tateishi, Application of
differences in the cell source used to generate osteogenic grafts. The
perfusion culture system improves in vitro and in vivo osteogenesis of bone
interplay between cell number, differentiation state, trophic factor marrow-derived osteoblastic cells in porous ceramic materials, Tissue Eng.
secretion, and the collective effect on the in vivo bone forming Part A 9 (6) (2003) 1205e1213.
[7] A.B. Yeatts, J.P. Fisher, Bone tissue engineering bioreactors: dynamic culture
potential of MSCs merits further study.
and the inuence of shear stress, Bone 48 (2) (2011) 171e181.
[8] A.B. Yeatts, E.M. Geibel, F.F. Fears, J.P. Fisher, Human mesenchymal stem cell
position within scaffolds inuences cell fate during dynamic culture, Bio-
6. Conclusion technol. Bioeng. 109 (9) (2012) 2381e2391.
[9] A.R. Shrivats, M.C. McDermott, J.O. Hollinger, Bone tissue engineering: state of
the union, Drug Discov. Today 19 (6) (2014) 781e786.
The results of this study demonstrate that HA-PLG scaffolds can
[10] N. Datta, Q.P. Pham, U. Sharma, V.I. Sikavitsas, J.A. Jansen, A.G. Mikos, In vitro
augment osteogenic differentiation of MSCs under perfusion. generated extracellular matrix and uid shear stress synergistically enhance
Moreover, this study provides detailed insight into construct 3D osteoblastic differentiation, Proc. Natl. Acad. Sci. U. S. A. 103 (8) (2006)
cellularity and osteogenic potential of MSCs as a function of 2488e2493.
[11] J. He, D.C. Genetos, J.K. Leach, Osteogenesis and trophic factor secretion are
bioreactor culture duration. While perfusion culture for only 7 days inuenced by the composition of hydroxyapatite/poly(lactide-co-glycolide)
leads to improved neovascularization, 14 days of bioreactor culture composite scaffolds, Tissue Eng. Part A 16 (1) (2010) 127e137.
is necessary for robust bone formation. As such, the duration of [12] D.G. Morales-Hernandez, D.C. Genetos, D.M. Working, K.C. Murphy, J.K. Leach,
Ceramic identity contributes to mechanical properties and osteoblast
mechanical stimulation of MSCs seeded in composite scaffolds is an behavior on macroporous composite scaffolds, J. Funct. Biomater. 3 (2) (2012)
important consideration for future studies aimed at generating 382e397.
osteogenic grafts. [13] S.S. Kim, M. Sun Park, O. Jeon, C. Yong Choi, B.S. Kim, Poly(lactide-co-glyco-
lide)/hydroxyapatite composite scaffolds for bone tissue engineering, Bio-
materials 27 (8) (2006) 1399e1409.
[14] S. Tajbakhsh, F. Hajiali, A comprehensive study on the fabrication and prop-
Author's contributions erties of biocomposites of poly(lactic acid)/ceramics for bone tissue engi-
neering, Mater Sci. Eng. C Mater Biol. Appl. 70 (Pt 1) (2017) 897e912.
D. Mitra: Conception and design, nancial support, collection [15] A. Matsiko, J.P. Gleeson, F.J. O'Brien, Scaffold mean pore size inuences
mesenchymal stem cell chondrogenic differentiation and matrix deposition,
and/or assembly of data, data analysis and interpretation, manu- Tissue Eng. Part A 21 (3e4) (2015) 486e497.
script writing. J. Whitehead: Collection and/or assembly of data. [16] C.A.B. Vissers, J.N. Harvestine, J.K. Leach, Pore size regulates mesenchymal
O.W. Yasui: Collection and/or assembly of data. J.K. Leach: stem cell response to Bioglass-loaded composite scaffolds, J. Mater. Chem. B 3
(44) (2015) 8650e8658.
Conception and design, nancial support, data analysis and inter- [17] H.L. Holtorf, N. Datta, J.A. Jansen, A.G. Mikos, Scaffold mesh size affects the
pretation, manuscript writing, nal approval of manuscript. osteoblastic differentiation of seeded marrow stromal cells cultured in a ow
perfusion bioreactor, J. Biomed. Mater Res. A 74 (2) (2005) 171e180.
[18] H.J. Kim, I.K. Park, J.H. Kim, C.S. Cho, M.S. Kim, Gas foaming fabrication of
Disclosure statement porous biphasic calcium phosphate for bone regeneration, Tissue Eng. Regen.
Med. 9 (2) (2012) 63e68.
[19] L.-C. Gerhardt, A.R. Boccaccini, Bioactive glass and glass-ceramic scaffolds for
The authors indicate no potential conicts of interest. bone tissue engineering, Materials 3 (7) (2010) 3867e3910.
[20] V.I. Sikavitsas, J. van den Dolder, G.N. Bancroft, J.A. Jansen, A.G. Mikos, Inu-
ence of the in vitro culture period on the in vivo performance of cell/titanium
Acknowledgements bone tissue-engineered constructs using a rat cranial critical size defect
model, J. Biomed. Mater Res. A 67 (3) (2003) 944e951.
[21] A. Scherberich, R. Galli, C. Jaquiery, J. Farhadi, I. Martin, Three-dimensional
This work was supported by the California Institute of Regen- perfusion culture of human adipose tissue-derived endothelial and osteo-
erative Medicine (RT3-07981) and the National Institutes of Health blastic progenitors generates osteogenic constructs with intrinsic vasculari-
zation capacity, Stem Cells 25 (7) (2007) 1823e1829.
(NIDCR DE025475) to JKL. DM is grateful for nancial support [22] W.L. Grayson, S. Bhumiratana, C. Cannizzaro, P.H. Chao, D.P. Lennon,
through an industry/campus supported fellowship under the A.I. Caplan, G. Vunjak-Novakovic, Effects of initial seeding density and uid
Training Program in Biomolecular Technology (T32-GM008799) at perfusion rate on formation of tissue-engineered bone, Tissue Eng. Part A 14
(11) (2008) 1809e1820.
the University of California, Davis. JW was supported by a National [23] W.L. Grayson, D. Marolt, S. Bhumiratana, M. Frohlich, X.E. Guo, G. Vunjak-
Science Foundation Graduate Research Fellowship (1650042). The Novakovic, Optimizing the medium perfusion rate in bone tissue engineering
content is solely the responsibility of the authors and does not bioreactors, Biotechnol. Bioeng. 108 (5) (2011) 1159e1170.
[24] G.M.d. Peppo, I.N. Marcos-Campos, D.J. Kahler, D. Alsalman, L. Shang,
necessarily represent the ofcial views of the National Institutes of
G. Vunjak-Novakovic, D. Marolt, Engineering bone tissue substitutes from
Health, CIRM or any other agency of the State of California. The human induced pluripotent stem cells, Proc. Natl. Acad. Sci. U. S. A. 110 (1)
funders had no role in the decision to publish, or preparation of the (2013) 8680e8685.
manuscript. [25] A.I. Hoch, B.Y. Binder, D.C. Genetos, J.K. Leach, Differentiation-dependent
secretion of proangiogenic factors by mesenchymal stem cells, PLoS One 7 (4)
(2012), e35579.
[26] S. Bhumiratana, J.C. Bernhard, D.M. Al, K. Yeager, R.E. Eton, J. Bova, F. Shah,
Appendix A. Supplementary data J.M. Gimble, M.J. Lopez, S.B. Eisig, G. Vunjak-Novakovic, Tissue-engineered
autologous grafts for facial bone reconstruction, Sci. Transl. Med. 8 (343)
(2016), 343ra83.
Supplementary data related to this article can be found at lu,
[27] I.G. Beskardes, R.S. Hayden, D.L. Glettig, D.L. Kaplan, M. Gmsdereliog
https://doi.org/10.1016/j.biomaterials.2017.08.044.
D. Mitra et al. / Biomaterials 146 (2017) 29e39 39

Bone tissue engineering with scaffold-supported perfusion co-cultures of D.W. Hutmacher, I. Martin, A collagen network phase improves cell seeding of
human stem cell-derived osteoblasts and cell line-derived osteoclasts, Process open-pore structure scaffolds under perfusion, J. Tissue Eng. Regen. Med. 7 (3)
Biochem. (2016), http://dx.doi.org/10.1016/j.procbio.2016.05.008 (in press). (2013) 183e191.
[28] L. Thorrez, J. Shansky, L. Wang, L. Fast, T. VandenDriessche, M. Chuah, [47] M. Schumacher, F. Uhl, R. Detsch, U. Deisinger, G. Ziegler, Static and dynamic
D. Mooney, H. Vandenburgh, Growth, differentiation, transplantation and cultivation of bone marrow stromal cells on biphasic calcium phosphate
survival of human skeletal myobers on biodegradable scaffolds, Biomater 29 scaffolds derived from an indirect rapid prototyping technique, J. Mater Sci.
(1) (2008) 75e84. Mater Med. 21 (11) (2010) 3039e3048.
[29] S.P. Pathi, D.D. Lin, J.R. Dorvee, L.A. Estroff, C. Fischbach, Hydroxyapatite [48] A.B. Yeatts, D.T. Choquette, J.P. Fisher, Bioreactors to inuence stem cell fate:
nanoparticle-containing scaffolds for the study of breast cancer bone metas- augmentation of mesenchymal stem cell signaling pathways via dynamic
tasis, Biomaterials 32 (22) (2011) 5112e5122. culture systems, Biochim. Biophys. Acta 1830 (2) (2013) 2470e2480.
[30] J. He, M.L. Decaris, J.K. Leach, Bioceramic-mediated trophic factor secretion by [49] T. Mygind, M. Stiehler, A. Baatrup, H. Li, X. Zou, A. Flyvbjerg, M. Kassem,
mesenchymal stem cells enhances in vitro endothelial cell persistence and C. Bunger, Mesenchymal stem cell ingrowth and differentiation on coralline
in vivo angiogenesis, Tissue Eng. Part A 18 (13e14) (2012) 1520e1528. hydroxyapatite scaffolds, Biomaterials 28 (6) (2007) 1036e1047.
[31] R.M. Boehler, S. Shin, A.G. Fast, R.M. Gower, L.D. Shea, A PLG/HAp composite [50] A. Di Luca, A. Longoni, G. Criscenti, C. Mota, C. van Blitterswijk, L. Moroni,
scaffold for lentivirus delivery, Biomaterials 34 (21) (2013) 5431e5438. Toward mimicking the bone structure: design of novel hierarchical scaffolds
[32] D. Mitra, H. Fatakdawala, M. Nguyen-Truong, A. Creecy, J. Nyman, L. Marcu, with a tailored radial porosity gradient, Biofabrication 8 (4) (2016) 045007.
J.K. Leach, Detection of pentosidine cross-links in cell-secreted decellularized [51] A. Di Luca, B. Ostrowska, I. Lorenzo-Moldero, A. Lepedda, W. Swieszkowski,
matrices using Time Resolved Fluorescence Spectroscopy, ACS Biomater. Sci. C. Van Blitterswijk, L. Moroni, Gradients in pore size enhance the osteogenic
Eng. (2016), http://dx.doi.org/10.1021/acsbiomaterials.6b00029 (in press). differentiation of human mesenchymal stromal cells in three-dimensional
[33] J.N. Harvestine, N. Vollmer, S. Ho, C. Zikry, M. Lee, K. Leach, Extracellular scaffolds, Sci. Rep. 6 (2016) 22898.
matrix-coated composite scaffolds promote mesenchymal stem cell persis- [52] A.M. Leferink, Y.C. Chng, C.A. van Blitterswijk, L. Moroni, Distribution and
tence and bone formation, Biomacromolecules 22 (2016) 3524e3531. viability of fetal and adult human bone marrow stromal cells in a biaxial
[34] K.C. Murphy, R.S. Stilhano, D. Mitra, D. Zhou, S. Batarni, E.A. Silva, J.K. Leach, rotating vessel bioreactor after seeding on polymeric 3D additive manufac-
Hydrogel biophysical properties instruct coculture-mediated osteogenic po- tured scaffolds, Front. Bioeng. Biotechnol. 3 (2015) 169.
tential, FASEB J. 30 (1) (2016) 477e486. [53] H.S. Yu, J.E. Won, G.Z. Jin, H.W. Kim, Construction of mesenchymal stem cell-
[35] A.I. Hoch, V. Mittal, D. Mitra, N. Vollmer, C.A. Zikry, J.K. Leach, Cell-secreted containing collagen gel with a macrochanneled polycaprolactone scaffold and
matrices perpetuate the bone-forming phenotype of differentiated mesen- the ow perfusion culturing for bone tissue engineering, Biores Open Access 1
chymal stem cells, Biomaterials 74 (2016) 178e187. (3) (2012) 124e136.
[36] L.A. Feldkamp, L.C. Davis, J.W. Kress, Practical cone-beam algorithm, J. Opt. [54] H.L. Holtorf, J.A. Jansen, A.G. Mikos, Flow perfusion culture induces the oste-
Soc. Am. A Opt. Image Sci. Vis. 1 (6) (1984) 612e619. oblastic differentiation of marrow stromal cell-scaffold constructs in the
[37] M.B. Keogh, S. Partap, J.S. Daly, F.J. O'Brien, Three hours of perfusion culture absence of dexamethasone, J. Biomed. Mater Res. A 72 (3) (2005) 326e334.
prior to 28 days of static culture, enhances osteogenesis by human cells in a [55] S. Seitz, K. Ern, G. Lamper, D. Docheva, I. Drosse, S. Milz, W. Mutschler,
collagen GAG scaffold, Biotechnol. Bioeng. 108 (5) (2011) 1203e1210. M. Schieker, Inuence of in vitro cultivation on the integration of cell-matrix
[38] V.I. Sikavitsas, G.N. Bancroft, H.L. Holtorf, J.A. Jansen, A.G. Mikos, Mineralized constructs after subcutaneous implantation, Tissue Eng. 13 (5) (2007)
matrix deposition by marrow stromal osteoblasts in 3D perfusion culture 1059e1067.
increases with increasing uid shear forces, Proc. Natl. Acad. Sci. U. S. A. 100 [56] A.B. Yeatts, S.K. Both, W. Yang, H.S. Alghamdi, F. Yang, J.P. Fisher, J.A. Jansen,
(25) (2003) 14683e14688. In vivo bone regeneration using tubular perfusion system bioreactor cultured
[39] V.I. Sikavitsas, G.N. Bancroft, J.J. Lemoine, M.A. Liebschner, M. Dauner, nanobrous scaffolds, Tissue Eng. Part A 20 (1e2) (2014) 139e146.
A.G. Mikos, Flow perfusion enhances the calcied matrix deposition of [57] J. Kim, T. Ma, Bioreactor strategy in bone tissue engineering: pre-culture and
marrow stromal cells in biodegradable nonwoven ber mesh scaffolds, Ann. osteogenic differentiation under two ow congurations, Tissue Eng. Part A
Biomed. Eng. 33 (1) (2005) 63e70. 18 (21e22) (2012) 2354e2364.
[40] D. Egger, S. Spitz, M. Fischer, S. Handschuh, M. Glosmann, B. Friemert, [58] K.D. Kavlock, A.S. Goldstein, Effect of pulse frequency on the osteogenic dif-
M. Egerbacher, C. Kasper, Application of a parallelizable perfusion bioreactor ferentiation of mesenchymal stem cells in a pulsatile perfusion bioreactor,
for physiologic 3D cell culture, Cells Tissues Organs 203 (5) (2017) 316e326. J. Biomech. Eng. 133 (9) (2011) 091005.
[41] W.L. Murphy, R.G. Dennis, J.L. Kileny, D.J. Mooney, Salt fusion: an approach to [59] L. Liu, B. Yu, J. Chen, Z. Tang, C. Zong, D. Shen, Q. Zheng, X. Tong, C. Gao,
improve pore interconnectivity within tissue engineering scaffolds, Tissue J. Wang, Different effects of intermittent and continuous uid shear stresses
Eng. 8 (1) (2002) 43e52. on osteogenic differentiation of human mesenchymal stem cells, Biomech.
[42] C. Liu, R. Abedian, R. Meister, C. Haasper, C. Hurschler, C. Krettek, G. von Model Mechanobiol. 11 (3e4) (2012) 391e401.
Lewinski, M. Jagodzinski, Inuence of perfusion and compression on the [60] J.V. Araujo, C. Cunha-Reis, T. Rada, M. Alves da Silva, M.E. Gomes, Y. Yang,
proliferation and differentiation of bone mesenchymal stromal cells seeded N. Ashammakhi, R.L. Reis, A.J. El-Haj, N.M. Neves, Dynamic culture of osteo-
on polyurethane scaffolds, Biomaterials 33 (4) (2012) 1052e1064. genic cells in biomimetically coated poly(caprolactone) nanobre mesh con-
[43] D. Wang, H. Jiang, S. Wang, H. Li, H. Zhang, L. Zhao, T. Peng, Z. Cao, S. Sun, structs, Tissue Eng. Part A 16 (2) (2010) 557e563.
Construction of tissue-engineered bone using a bioreactor and platelet-rich [61] B.Y. Binder, D.C. Genetos, J.K. Leach, Lysophosphatidic acid protects human
plasma, Exp. Ther. Med. 8 (2) (2014) 413e418. mesenchymal stromal cells from differentiation-dependent vulnerability to
[44] A.S. Goldstein, G. Zhu, G.E. Morris, R.K. Meszlenyi, A.G. Mikos, Effect of oste- apoptosis, Tissue Eng. Part A 20 (7e8) (2014) 1156e1164.
oblastic culture conditions on the structure of poly(DL-lactic-co-glycolic acid) [62] M.L. Decaris, B.Y. Binder, M.A. Soicher, A. Bhat, J.K. Leach, Cell-derived matrix
foam scaffolds, Tissue Eng. 5 (5) (1999) 421e434. coatings for polymeric scaffolds, Tissue Eng. Part A 18 (19e20) (2012)
[45] A. Braccini, D. Wendt, C. Jaquiery, M. Jakob, M. Heberer, L. Kenins, A. Wodnar- 2148e2157.
Filipowicz, R. Quarto, I. Martin, Three-dimensional perfusion culture of human [63] M. Chen, M. Zhou, Z. Ye, Y. Zhou, W.S. Tan, Ectopic osteogenesis of macro-
bone marrow cells and generation of osteoinductive grafts, Stem Cells 23 (8) scopic tissue constructs assembled from human mesenchymal stem cell-laden
(2005) 1066e1072. microcarriers through in vitro perfusion culture, PLoS One 9 (10) (2014),
[46] A. Papadimitropoulos, S.A. Riboldi, B. Tonnarelli, E. Piccinini, M.A. Woodruff, e109214.

Das könnte Ihnen auch gefallen