Articulo Histología

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Cell Tissue Bank (2017) 18:369–381

DOI 10.1007/s10561-017-9635-4

Co-culture of dedifferentiated and primary human


chondrocytes obtained from cadaveric donor enhance
the histological quality of repair tissue: an in-vivo animal
study
Anell Olivos-Meza . Cristina Velasquillo Martı́nez . Brenda Olivos Dı́az .
Carlos Landa-Solı́s . Mats Brittberg . Raul Pichardo Bahena . Carmina Ortega Sanchez .
Valentin Martı́nez . Enrique Alvarez Lara . José Clemente Ibarra-Ponce de León

Received: 21 December 2016 / Accepted: 26 May 2017 / Published online: 5 June 2017
Ó Springer Science+Business Media Dordrecht 2017

Abstract To compare the quality of the repair tissue subcutaneous tissue of athymic mice and left for
in three-dimensional co-culture of human chondro- 3 months growth. Safranin-O and Alcian blue staining
cytes implanted in an in vivo model. Six cadaveric and were used to glycosaminoglycan content assessment.
five live human donors were included. Osteochondral Aggrecan and type-II collagen were evaluated by
biopsies from the donor knees were harvested for immunohistochemistry. New-formed tissue quality
chondrocyte isolation. Fifty percent of cadaveric was evaluated with an adaptation of the modified
chondrocytes were expanded until passage-2 (P2) O’Driscoll score. Histological quality of non-co-
while the remaining cells were cryopreserved in cultured group was 4.37 (SD ±4.71), while co-
passage-0 (P0). Fresh primary chondrocytes (P0f) cultured groups had a mean score of 8.71 (SD
obtained from live human donors were co-cultured. ±3.98) for the fresh primary chondrocytes and 9.57
Three-dimensional constructs were prepared with a (SD ±1.27) in the cryopreserved chondrocytes. In
monolayer of passage-2 chondrocytes, collagen mem- immunohistochemistry, Co-culture groups were
brane (Geistlich Bio-GideÒ), and pellet of non-co- strongly stained for type-II and aggrecan not seen in
cultured (P2) or co-cultured chondrocytes (P2 ? P0c, the non-co-cultured group. It is possible to isolate
P2 ? P0f). Constructs were implanted in the viable chondrocytes from cadaveric human donors in

A. Olivos-Meza M. Brittberg
Orthopedic Sports Medicine and Arthroscopy Service, Region Halland Orthopaedics, Kungsbacka Hospital,
Instituto Nacional de Rehabilitación, Mexico City, Kungsbacka, Sweden
Mexico
R. Pichardo Bahena  E. Alvarez Lara
C. Landa-Solı́s  C. Ortega Sanchez  V. Martı́nez Pathology Service, Instituto Nacional de Rehabilitación,
Tissue Engineering, Cell Therapy and Regenerative Mexico City, Mexico
Medicine Unit, Instituto Nacional de Rehabilitación,
Mexico City, Mexico J. C. Ibarra-Ponce de León (&)
Instituto Nacional de Rehabilitación, Av México-
B. Olivos Dı́az Xochimilco No. 289, Arenal de Guadalupe, ZC,
Universidad Nacional Autónoma de México, 14389 Mexico City, Mexico
Mexico City, Mexico e-mail: cibarra.corresponding@hotmail.com

B. Olivos Dı́az C. Velasquillo Martı́nez


Instituto Nacional de Rehabilitación, Mexico City, Subdirection of technological research, Instituto Nacional
Mexico de Rehabilitación, Mexico City, Mexico

123
370 Cell Tissue Bank (2017) 18:369–381

samples processed in the first 48-h of dead. There is only a small number of cells can be retrieved from a
non-significant difference between the numbers of patient. This issue is one major problem in the field of
chondrocytes isolated from live or cadaveric donors. cartilage repair (Giovannini et al. 2010). Since cell-
Cryopreservation of cadaveric primary chondrocytes based therapy requires a large quantity of cells, it seems
does not alter the capability to form cartilage like that any attempt to ‘‘regenerate’’ hyaline like cartilage
tissue. Co-culture of primary and passaged chondro- requires in vitro chondrocyte expansion to increase cell
cytes enhances the histological quality of new-formed number by passaging and to be able to fill a cartilage
tissue compared to non-co-cultured cells. defect with chondrogenic cells in high density (Bagha-
ban Eslaminejad et al. 2009; Bian et al. 2011; Casper
Keywords Chondrocyte dedifferentiation  Co- et al. 2010; Oseni et al. 2013).
culture  Cartilage repair  Tissue engineering  Chondrocytes possess a high proliferative capacity
Cadaveric chondrocytes when being cultured in monolayers; however, the
propensity of chondrocytes to dedifferentiate into
fibroblasts over repeated passage is a major problem
(Baghaban Eslaminejad et al. 2009; Gartland et al.
Introduction 2005; Oseni et al. 2013; Qing et al. 2011).
Dedifferentiated chondrocytes changed their
Articular cartilage lesions have been reported in more morphology from rounded to fibroblastic and
than 60% of knee arthroscopic procedures in young steadily lose the expression of the cartilage-specific
patients (Curl et al. 1997; Farr et al. 2011; Hjelle et al. genes such as those encoding collagen type II and
2002; Johnstone et al. 2013; McNickle et al. 2008; proteoglycans (Ahmed et al. 2009; Baghaban
Widuchowski et al. 2007). Chondral lesions generally Eslaminejad et al. 2009; Hamada et al. 2013;
do not heal or heal partially under biological conditions Kolettas et al. 2001). It has been strongly suggested
if the defects penetrate subchondrally into bone that dedifferentiation of chondrocytes affects the
marrow area. The physiological process of sponta- quality of the cartilage regenerated by Autologous
neous repair plays a key role in a number of surgically Chondrocyte Implantation (ACI) (Kolettas et al.
based techniques (Hunziker 2002). The treatment of 2001). Reports have shown that co-culture in
cartilage defects is a major challenge in the orthopedic in vitro systems enhance the quality of expanded
field because articular cartilage has a low potential of dedifferentiated chondrocytes (Juhasz et al. 2014).
intrinsic repair (Ahmed et al. 2009; Giovannini et al. Two types of co-cultures have been described:
2010; Hamada et al. 2013; Kolettas et al. 2001; Negri direct (cell–cell contact) and indirect (soluble
et al. 2007; Olivos-Meza et al. 2010; Wei et al. 2012). mediators). Some studies suggest that soluble
Surgical strategies available to treat cartilage defects mediators such as TGF-B1, IGF-I, and BM-P2
are in general two. The first approach is to enhance the may play a role in cell–cell interaction in co-
intrinsic healing capacity of subchondral bone and cultures both in vivo and in vitro (Juhasz et al. 2014;
cartilage through a migration of mesenchymal stem Tew et al. 2008). However other studies have shown
cells (bone marrow stimulation techniques) into the that direct cell–cell contacts are preferentially
defect area leading to the formation of fibrocartilage responsible for improved chondrogenesis in co-
which does not have the resistance of hyaline cartilage culture systems (Bian et al. 2011; Juhasz et al.
due to its biochemical composition (Ibarra et al. 2014; 2014). In a rabbit study, Chang et al. reported that
Kolettas et al. 2001; Negri et al. 2007). The second mature cartilage cells secrete growth factors such as
alternative is to elicit a biological repair by tissue transforming growth factor-B (TGF-B) and insulin-
engineering methods that provide a formation of like growth factor-1 (IGF-1), which induce chon-
hyaline like cartilage. The last approach has resulted drocyte differentiation of Bone Marrow Stem Cells
in long-term durability and functionality compared (BMSCs) in co-cultures. Although BMSCs could be
with the bone marrow stimulation techniques. How- induced into cartilage cell-like cell, they did not
ever, with chondrocytes contributing to less than 5% of determine whether the co-culture of chondrocytes
the total tissue volume, and the limited biopsy size that and BMSCs was more effective than single culture
can be collected from a minor weight-bearing area, of each cell type. The same effect could promote

123
Cell Tissue Bank (2017) 18:369–381 371

reversible morphological and physiological changes patients who were scheduled to trans-tibial ACL
in dedifferentiated chondrocytes co-cultured with reconstruction due to this procedure requires taking a
primary chondrocytes (Ahmed et al. 2009). cartilage fragment of the lateral wall of the notch
Limited donor tissue could be solved with the use of (notchplasty) to see perfectly the background of the
allogenic cartilage as a source for the acquisition of notch. All patients underwent regional anesthesia, after
sufficient number of chondrocytes of the appropriate diagnostic arthroscopy and confirmation of the ACL
phenotype for cryopreservation, expansion, or co- rupture, cleaning of the footprint and notchplasty of the
culture models. However, the use of cadaveric carti- lateral femoral condyle were performed. Instead of
lage for chondrocyte isolation, its chondrogenic traditional notchplasty with bone burr, two cylinders of
capability, and cell viability through the time has not 4 mm diameter were obtained from a non-weight-
been described in the literature. bearing area adjacent to the intercondylar notch (COR;
In this study, we compared the quality of the DePuy Mitek, Raynham, MA) (Fig. 1). The osteochon-
repaired tissue in three-dimensional co-culture (por- dral cylinders were placed in a sterile container with
cine collagen type III and type I scaffold) of human transport media (Dulbecco’s Modified Eagle Medium
chondrocytes implanted in an in vivo model. F12 GIBCO, Grand Island, NY) containing 10%
antibiotics/antimycotic agents and sent out to the tissue
engineering laboratory for chondrocyte isolation, co-
Materials and methods culturing, and cell-collagen scaffold formation (con-
struct). After osteochondral cylinder harvesting,
Cadaveric donor cartilage harvesting remodeling of lateral wall of the notch was finished
with an arthroscopic 5 mm bone burr; then ACL
Cadaveric human donors from Musculoskeletal Tissue reconstruction was performed in a regular manner.
and Skin Bank (Biograft of Mexico) with age from 15
to 50 years were enrolled in this study. Osteochondral Chondrocyte isolation
cylinders of 8–10 mm (COR; DePuy Mitek, Rayn-
ham, MA) diameter were harvested from any healthy, The cadaveric and live donor’s osteochondral cylin-
smooth and white articular cartilage area from both ders were processed in the Tissue Engineering and
knees. Biopsies were harvested in sterile conditions by Regenerative Medicine Laboratory at the National
the procuration staff of Biograft tissue banking after Institute of Rehabilitation. Under sterile conditions in
the donor arrangements were solved. Osteochondral a laminar flow hood (100-class), transported medium
samples were placed in a sterile container with culture was drained, samples were washed three times with
medium (Dulbecco’s Modified Eagle Medium F12 PBS 10% antibiotic/antimycotic, and cartilage was
GIBCO, Grand Island, NY) containing 10% antibi- separated from bone by sharp dissection. Chondral
otics/antimycotic. Samples were transported at 4 °C fragments were chopped in small pieces with a scalpel
from the hospital were the patient died to the National and then fragments were digested with type-I-colla-
Institute of Rehabilitation where they were processed. genase at 0.3% in DMEM-F12 medium for around 5-h.
One osteochondral plug in every donor was taken as a Isolated cells were counted in Neubauer chamber and
control for histology and immunohistochemistry. viability was assessed by trypan-blue stain by two
blind observers. Eighty percent of primary isolated
Live donor cartilage harvesting chondrocytes from cadaveric donors were seeded in
T25 culture flask at a density of 250,000 cells and were
After institutional review board evaluation and expanded until Passage-2 (P2). The remaining twenty
approval of this study from local ethics committee, percent of cadaveric donor chondrocytes were stored
live donors were invited to participate in this study. If and cryopreserved in cryo-vials with 10% DMSO and
subjects accepted to participate informed consent was Bovine Fetal Serum (BFS) at a density of 5 9 105
signed. Patients scheduled for arthroscopic anterior cells per vial. Temperature was reduced gradually and
cruciate ligament (ACL) reconstruction between 18 and cells were stored in liquid nitrogen at -280 °C.
50 years of age were included as donors for recently Primary fresh chondrocytes (P0f) were obtained from
isolated chondrocytes biopsies. We enrolled only live donors until the cadaveric chondrocytes reached

123
372 Cell Tissue Bank (2017) 18:369–381

Fig. 1 a Two osteochondral biopsies with a diameter of 4 m care do not touch and damage the articular cartilage. c Ten
were harvested in the lateral notch, then the notchplasty was millimeters of bone were preserved to manipulate and process
finished with a burr. b Samples were handled carefully, taking the cartilage without causing damage of the tissue

passage-2. Isolation was performed as described DMSO and Bovine Fetal Serum (BFS) at a density
above and used immediately for co-culturing and of 5 9 105 cells per vial. Temperature was reduced
construct formation (P2 ? P0f group). gradually. First received samples were stored in a
refrigerator at minus 20 °C for 20 min, then vials were
Expansion and chondrocyte dedifferentiation moved to -80 °C for 24-h. Finally cells were stored in
liquid nitrogen at -280 °C. Primary cryopreserved
Primary isolated chondrocytes from cadaveric donors chondrocytes (P0c) were stored until the cultured cells
were seeded into T-25 culture flasks at a density of reached the Passage-2 for co-culture and cell-collagen
250,000 cells with 5 ml of culture medium (Dul- scaffold formation (P2 ? P0c group).
becco’s Modified Eagle Medium F12 GIBCO, Grand
Island, NY), 1% antibiotic–antimycotic agents, and 3D cell-collagen scaffold preparation
supplemented with 10% human serum. Cells were
expanded in monolayer culture until 90–100% con- Once the cadaveric chondrocytes expanded until
fluence was reached. Cultures were trypsinized and re- passage-2 reached a confluency of 90–100% the
seeded for cell expansion until passage-2. At the constructs were prepared from a monolayer of chon-
beginning of the second passage, fifty percent of cells drocytes in passage-2 together with a collagen type I
were seeded in T-25 flasks for pellet formation. To and III membrane (Geistlich Bio-GideÒ) as scaffold,
obtain a handle monolayer, the remain chondrocytes and a pellet of either passaged-2 chondrocytes (non-
were seeded in 60 9 15 mm Petri dishes. Culture cocultured) or co-cultured cells (P2 ? P0c, P2 ? P0f)
medium for these cells was supplemented with was added (Figs. 2, 3).
ascorbic acid (150 mcg/50 ml medium). Medium First, the peripheral borders of the monolayer were
changes were performed twice a week. peeled-off from the bottom of the petri dish (Fig. 2a)
followed by a circle of 8 mm of collagen membrane
Cryopreservation of primary chondrocytes that was placed over the center of monolayer with the
porous-absorbable side facing up (Fig. 2b). Finally, a
The remaining cadaveric donor primary chondrocytes pellet of co-cultured or non-co-cultured cells was
were cryopreserved in 2 ml-cryovials with 10% added with a 200 ll micropipette over the scaffold

123
Cell Tissue Bank (2017) 18:369–381 373

(Fig. 2c). The pellet in every construct contained a Modified Eagle Medium F12 GIBCO, Grand Island,
total of 2 9 106 chondrocytes as follow: (a) No- NY) containing 1% antibiotics/antimycotic supple-
cocultured group: 2 9 106 Passage-2 cadaveric chon- mented with 10% human serum, and ascorbic acid was
drocytes (P2); (b) Co-Cultured Cadaveric cells: added (Fig. 2f). The construct was left in the incubator
1.6 9 106 Passage-2 chondrocytes ? 4 9 105 Pas- for 3 days to enhance the consistency.
sage-0 chondrocytes (P2 ? P0c); and (c) Co-cultured
Cadaveric Chondrocytes/Fresh Chondrocytes from In-vivo animal model implantation
living donors: 1.6 9 106 Passage-2 cadaveric chon-
drocytes ? 4 9 105 Fresh Passage-0 Chondrocytes All work in this study was conducted with the approval
(P2 ? P0f) (Fig. 4). Co-cultured cells were mixed of local Institutional Animal care and use committee.
with a ratio of 4:1 (4 dedifferentiated chondrocytes, Five days after the construct was formed, it was
P2: 1 differentiated chondrocytes, P0) in every implanted in male athymic mouse (Nu/Nu) with age of
construct. After the pellet was absorbed the borders 4-weeks. Surgeries were done in a laminar flow hood
of the monolayer were taken up to cover the unit in the animal facility of the National Institute of
scaffold-pellet (Fig. 2d). In this way, a construct like Rehabilitation. Animals underwent general anesthesia
crepe was formed as described by Masri et al. (2007) with Isoflurane. Incision length of 6–8 mm was
(Fig. 2e). To gain consistency, the construct was performed in the dorsal area (Fig. 5a). Subcutaneous
placed in the incubator without medium during tissue was released (Fig. 5b) and the cell construct was
30 min. After that, 5 ml of medium (Dulbecco’s implanted into this area (Fig. 5c). One or two 3-0

Fig. 2 Construct formation. a Peripheral borders of the borders of the monolayer were taken up to cover the unit
monolayer were peel from the bottom of the petri dish. b The scaffold-pellet. e, f The crepe was formed and placed in the
collagen scaffold was placed over the center of monolayer with incubator without medium during 5 min, then medium and
the porous-absorbable side facing up. c A pellet of co-cultured ascorbic acid was added. Implantation is done after 5-days
or no co-cultured cells was added over the scaffold. d The

123
374 Cell Tissue Bank (2017) 18:369–381

Fig. 3 Schematic
representation of ‘‘the crepe
technique’’. (1) A
monolayer of Passage-2
cadaveric chondrocytes was
formed adding ascorbic acid
to cultures. (2) the
monolayer was peel-off
from the bottom of the
culture plate. (3) An 8-mm
diameter collagen type III
and I scaffold was placed
over the monolayer. (4) A
pellet of 2 9 106 cells was
added to the scaffold (co-
cultured or no co-cultured
chondrocytes). (5) The
monolayer was folding to
cover the scaffold-pellet unit
in a crepe manner. (6) The
3D construct is formed and
supplemented with culture
medium and leave in the
incubator during 3 days
before implantation

nylon simple stitches were used to close the skin Carl Zeiss microscope and AxioVision 4.8.2. Imaging
(Fig. 5d). After 3-months post-implantation, animals system. Histological parameters were assessed and
were sacrificed (based in the animal regulation NOM- scored.
062-ZOO-1999). New-formed tissue was harvested
and used for histology (Fig. 6). Immunohistochemical evaluation

Histological evaluation Tissue sections were fixed on a slide if de-waxed,


followed by treatment with hyaluronidase (Sigma;
The cartilage and new-formed tissue were fixed in 4% 4800 U/ml in 0.025 M NaCl 0.05 M sodium acetate
phosphate-buffered with formaldehyde, after being pH 5.0, 2 h at RT for frozen sections) to unmask the
dehydrated and embedded in paraffin: Three-micron collagen antigens. Sections were then incubated for
thickness sections were taken from the center and 1 h at room temperature with primary antibodies
stained with Hematoxylin–Eosin (H&E) to assess against type II collagen. Endogenous peroxidase was
morphology, Safranin-O (SO), and Alcian Blue (AB) blocked with 0.3% hydrogen peroxide in methanol
to localize the amount of glycosaminoglycan content. before sections were incubated with the biotinylated
We performed visualization and photography using a secondary antibody anti-mouse. The signal was

123
Cell Tissue Bank (2017) 18:369–381 375

Fig. 4 Cell groups


distribution. Two groups of
donors were used to isolate
chondrocytes: cadaveric and
living donors. Fifty percent
of chondrocytes isolated
form cadaveric donors were
expanded until passage-2 to
form a monolayer culture
while the remain 50% was
cryopreserved in passage-0
for further use in
cryopreserved chondrocytes
co-culture. The
chondrocytes isolated from
living donors were not
expanded, they were use in
passage-0 for co-culture of
fresh chondrocytes

Fig. 5 After mouse was anesthetized, an incision of 8–10 mm was performed in the dorsal area (a), subcutaneous tissue under the skin
incision was released (b), and the construct was implanted (c). One 3-0 nylon simple stitches were used to close the skin (d)

amplified using an avidin–biotin–peroxidase reagent normal mouse IgG, normal rabbit serum or phosphate
(Vectastain Elite ABC kit; Vector Laboratories, buffered saline alone, in place of the primary antibod-
Peterborough, UK) and labelling was visualized with ies and then treatment continued in the same way as all
diaminobenzidine as substrate. Sections were then the other slides.
washed, dehydrated, and mounted in pertex. ‘Control’ The degree of immunohistochemical labelling was
sections of biopsy samples were treated either with assessed semi-quantitatively by measuring the areas of

123
376 Cell Tissue Bank (2017) 18:369–381

Fig. 6 a New-formed tissue (black arrow) in the subcutaneous and measured; macroscopic characteristics were recorded and
area of atomic mouse. b After 3 months of construct implan- tissue was processed for histology and immunohistochemistry
tation mice were sacrificed and new formed tissue was harvested

the repair cartilage, on adjacent sections where 3,044,908 (±1,161,090) viable isolated cells. Mean
possible, which were immunostained for collagen number of chondrocytes per milligram of cartilage
type II. Images were captured using an Optivision was 1951 (±70.3). Mean time from death to cartilage
Image Capture system. The area of positive staining processing was 21.75 h (±6.13). Causes of death were
was delineated and this was calculated as a percentage liver failure, lung thromboembolism, brain death and
of the total area. cranioencephalic trauma. All osteochondral biopsies
were transparent, pearly bluish in color with firm
Statistical analysis consistency. No crystal deposition or chondral damage
was identified in any sample.
Data are reported as means (±) SD and were analyzed
statistically using the SPSS version 20.0 software Live donor osteochondral biopsies
(LEAD Technologies, Inc., USA). The statistical
significance of the differences between groups was Five live donors were included in this study. Three
determined by one-way analyses of variance subjects were men (60%) and two were women (40%).
(ANOVA). p \ 0.05 was considered to be statistically All patients had diagnosis of ACL rupture. Mean
significant. donor age was 32 (±11.33), we obtained an average of
1.4 (±0.54) biopsies from every patient with a
diameter of 4 mm. Mean cartilage weight per donor
Results was 97.54 mg (±69.28) with a mean of 605,600
(±514,451) viable isolated cells. The mean number of
Cadaveric osteochondral biopsies chondrocytes per milligram of cartilage was 4396
(±2506). Mean time from osteochondral biopsy
We received a total of thirty osteochondral samples harvesting to cartilage processing was 1.9 h (±0.54).
from six cadaveric donors. However, two of those As in the cadaveric donor all biopsies were transpar-
were excluded due to positive serological testing to ent, pearly bluish in color with firm consistency. No
Human T-Lymphotropic Virus (HTLV) and Syphilis; crystal deposition or chondral damage was identified
eight biopsies from these positive donors were in any sample.
excluded.
Mean donor’s age was 30.5 years (15–41). All Live donor versus cadaveric donor isolated
donors were men; the mean number of biopsies chondrocytes
obtained per donor was 5.5 (±1.91) with a mean
diameter of 8.5 mm (±1.0). Mean cartilage weight per Significant statistical difference was found in process-
donor was 1560 mg (±587.88) with a mean of ing time (biopsy to chondrocyte isolation) between

123
Cell Tissue Bank (2017) 18:369–381 377

cadaveric donor (mean 21.7-h) and live donor samples Non-co-culture chondrocytes seeded in 3D-cell-
(mean 1.9-h) (p = 0.007). However, we did not found collagen-constructs had fibroblastic-like cells, while
significant difference between the number of primary co-cultured chondrocytes either cryopreserved or
chondrocytes per milligram of tissue isolated from freshly isolated retained the rounded shape character-
cadaveric versus live donor cartilage (p = 0.12). istic from chondrocytes.

Chondrocyte viability Tissue morphology and glycosaminoglycan


content evaluation
After isolation, chondrocyte viability was assessed in
the 18-cadaveric and five-live donor processed sam- Hyaline-like cartilage observed in the co-cultured
ples. Total surface area of cartilage from cadaveric constructs differed clearly from the fibrous-like tissue
samples was 184 mm with a weight of 6240 mg, and formed by non-co-cultured passaged cells because of
12,179,632 viable chondrocytes while in live donors its homogenous appearance of the matrix and the
total cartilage surface was 28 mm with a total weight round or oval shape of cells, often surrounded by
of 487.7 mg, and 3,028,000 viable chondrocytes. lacunae (Fig. 7b, c). Fibrous-like cartilage in contrast,
had obvious bundles of collagen fibers lying in a
Chondrocytes expansion random, irregular manner and often elliptical shaped
cells (Fig. 7d).
Mean number of seeded chondrocytes at Passage-1 Glycosaminoglycan (GAGs) content was assessed
was 1,250,160 (±353,402) cells obtaining a mean of by Safranin-O and Alcian blue staining. There was
12,138,000 (±3,744,224) chondrocytes at the end of more intense matrix around lacunae in new-formed
Passage-2 with a mean expansion rate of 10.03 tissue from co-cultured cryopreserved chondrocytes
(±3.82). (Fig. 7b) than in the tissue obtained from co-cultured
In this study, we observed a close relation between fresh chondrocytes (Fig. 7c) but in both cell types
age and expansion rate. The younger the patient the there were extensive stained areas.
greater the number of chondrocytes at the final culture.
The youngest donor was 15 years-old and had the Quality of the repair tissue
greatest expansion rate (15.2 fold) while the oldest
donor was 41 years-old and showed the lowest Although in different amount, as illustrated in histo-
number of expanded chondrocytes (6.67 fold). logical images, neocartilage was formed in either non-
co-cultured and co-cultured groups. However, the
Gross observations of new-formed tissue structural characteristics in the repair tissue appeared
to be better in the cryopreserved and fresh primary
To minimize manipulation of the implanted samples chondrocytes group compared to Passage-2 con-
during harvesting, precise measurements of the size of structs. These observations are reflected in the histo-
new-formed tissue were not taken. However, we logical scores. Three items of the modified O’Driscoll
observed a reduction of the original size. After 3 months score (Table 1) were used to quantify the structural
of implantation, the consistency of the constructs characteristics of the new-formed tissue, which
changed from soft and viscous tissue to firm consistency includes cellular morphology, Safranin-O staining,
like a cartilage tissue. Surface was regular and smooth, and hypo cellularity. Additional to those three scor-
but the color kept yellow as in the beginning. ings, we enclosed how easy it was to find the new-
formed tissue with different microscope objectives
Cell morphology (49, 109, and 209). The maximum score of 14
corresponded to normal articular cartilage. In this
Chondrocyte morphology changed through the sub- study, we found the best mean score (14, SD ±0.0) in
cultures from round or polyedrich in recently isolated the control samples (hyaline cartilage), while the worst
cells to fibroblastic-like when cells were passaged in histologic results were observed in the non-co-cul-
monolayer cultures. Dedifferentiation process tured group (4.37, SD ±4.71). Although co-cultured
increased with passage numbers. chondrocytes did not get as good score as control

123
378 Cell Tissue Bank (2017) 18:369–381

Fig. 7 a Alcian blue stained histological sections of human formed tissue obtained from co-cultured cryopreserved chon-
knee articular cartilage obtained from cadaveric donor used as a drocytes than in the tissue from co-cultured fresh chondrocytes.
control to compare the presence of GAGs content with the d Small zones of light staining were observed in tissue formed
repaired tissue in experimental groups. b, c Alcian blue staining by no–co-cultured cells (P2)
was more intense and stained more extensive areas in the new-

Table 1 Modified O’Driscoll histological score (modified)


O’Driscoll score with
detailed breakdown of the Cellular morphology
histological items to
Hyaline articular cartilage 4
consider in the evaluation of
the neo-formed cartilage Incompletely differentiated mesenchyme 2
Fibrous tissue or bone 0
Safranin-O staining
Normal or nearly normal 3
Moderate 2
Slight 1
None 0
Hypocellularity
Considered characteristics
to evaluate in histology are: Normal cellularity 3
cell morphology, intensity Slight hypocellularity 2
of Safranin-O staining, Moderate hypocellularity 1
hypocellularity, and
feasibility to identify Severe hypocellularity 0
cartilage-like tissue by Microscopic identification
microscope. Highest score Easy identification at 49 4
is fourteen, those results
Easy identification at 109 3
means that repaired tissue is
more like cartilage. New- Easy identification at 209 2
formed tissue with more Difficult identification at 209 1
fibrous characteristics has No identification of cartilage-like structure 0
lowest scores closed to zero

123
Cell Tissue Bank (2017) 18:369–381 379

Fig. 8 Safranin-O stained


histological sections of
human knee articular
cartilage (control) and
repaired tissue from co-
cultured and no co-cultured
chondrocytes. a Cadaveric
biopsy of articular cartilage
was taken as a control (14,
SD ±0.0) in the histological
evaluation with the
O’Driscoll score system;
good quality cartilage is
observed. b, c Moderate
quality cartilage is observed
in repair tissue obtained
from co-culture of
cryopreserved chondrocytes
(9.57, ±1.27) and co-culture
of fresh chondrocytes (8.71,
±3.98). d Low quality
cartilage is observed in no
co-cultured cells (4.37, SD
±4.71) with presence of
fibrous tissue characteristics
that demonstrates few
matrix formation

samples, those groups showed better results than tissue cartilage from co-cultured chondrocytes (P2 ? P0c
originated from de-differentiated chondrocytes with and P2 ? P0f) had strong immunopositivity for type II
mean score of 8.71 (±3.98) for the fresh primary collagen and aggrecan to varying extents in all
chondrocytes co-cultured group and 9.57 (±1.27) in samples of those groups. However, only light if any
the cryopreserved primary co-cultured chondrocytes. immunopositivity for type-II collagen and aggrecan
No statistical difference was found between non-co- were present in no-co-cultured group (P2). Unspecific
cultured chondrocytes and fresh primary co-cultured immunopositive was observed in collagen type I–III
group (p = 0.623); cryopreserved co-cultured chon- membrane used as scaffold (Geistlich Bio-GideÒ) in
drocytes and fresh co-cultured cells neither showed all experimental groups.
significant difference. However, the histological score
was significantly higher (p = 0.053) in the cryopre-
served co-cultured group compared to non-co-cultured Discussion
group (Fig. 8).
Among all the cell sources considered for cartilage
Immunohistochemistry analysis repair, chondrocytes remain the cells of choice. Most
of the available studies reports the use of human
Staining for type-II collagen and aggrecan were more articular chondrocytes recently isolated in live donor,
intense on chondrocytes and the matrix around specialty for ACI technique. The use of cadaveric
lacunae in normal cartilage (control). Regenerated donor articular chondrocytes either fresh or

123
380 Cell Tissue Bank (2017) 18:369–381

cryopreserved has been not reported in the literature. Acknowledgements This work is supported by the Mexican
This strategy would help to cover one of the major Council of Science (CONACyT) Grant SALUD-PDCPN-2013-
01-215138; Technology, Science and Innovation Secretary
limitations for cell-based therapeutic approaches for Grants: SECITI 079 BIS/2013 and SECITI/INR/GOB-25/
articular cartilage regeneration, the lack of sufficient 2013. The authors wish specialty to thank to Biograft of
number of chondrocytes because of the donor site México Musculoskeletal Tissue and Skin Bank; Blood Bank &
morbidity and dedifferentiation process because of Pathology Service at the National Institute of Rehabilitation;
and Autonomous National University of Mexico (UNAM). We
expansion methods. also appreciate the support and collaboration of Veterinary
In this study, we demonstrate the feasibility to Facility at the National Institute of Rehabilitation: Hugo-Lecona
obtain viable cadaveric chondrocytes despite the time DMV and Javier-Perez DMV.
between donor dead and chondrocyte isolation in the
Compliance with ethical standards
laboratory. Our findings suggest the possibility to
eliminate the necessity of autologous osteochondral Conflict of interest The authors declare that they have no
biopsy in ACI technique. This strategy could reduce conflict of interest.
patient morbidities, time, costs, and removing one
surgery for cartilage biopsy.
By exploiting the paracrine signals from primary References
chondrocytes (P0) over the passaged cells (P2), we
demonstrated by histology that cartilage like-tissue Ahmed N, Gan L, Nagy A, Zheng J, Wang C, Kandel RA (2009)
Cartilage tissue formation using redifferentiated passaged
formation can be achieved by in a 3D cell-collagen chondrocytes in vitro. Tissue Eng Part A 15:665–673
scaffold co-cultured chondrocytes. In contrast, non- Baghaban Eslaminejad M, Taghiyar L, Falahi F (2009) Quan-
co-cultured cells showed predominately fibrous repair titative analysis of the proliferation and differentiation of
tissue despite their culture in three dimensional rat articular chondrocytes in alginate 3D culture. Iran
Biomed J 13:153–160
scaffolds. Bian L, Zhai DY, Mauck RL, Burdick JA (2011) Coculture of
This study has limitations in that we have used human mesenchymal stem cells and articular chondrocytes
immunohistochemistry to assess the number of char- reduces hypertrophy and enhances functional properties of
acteristic components of hyaline cartilage as type-II engineered cartilage. Tissue Eng Part A 17:1137–1145
Casper ME, Fitzsimmons JS, Stone JJ, Meza AO, Huang Y,
collagen and aggrecan. This method does not permit a Ruesink TJ, O’Driscoll SW, Reinholz GG (2010) Tissue
truly quantitative assessment to compare the results engineering of cartilage using poly-epsilon-caprolactone
between groups. To have more specific results, DNA nanofiber scaffolds seeded in vivo with periosteal cells.
quantification to determine cartilage-specific matrix Osteoarthritis Cartilage 18:981–991
Curl WW, Krome J, Gordon ES, Rushing J, Smith BP, Poehling
content between groups is needed. GG (1997) Cartilage injuries: a review of 31,516 knee
In order to translate our results to the clinic, further arthroscopies. Arthrosc J Arthrosc Relat Surg 13:456–460
investigations should be performed regarding the Farr J, Cole B, Dhawan A, Kercher J, Sherman S (2011) Clinical
chondrocyte immune responses and the potential cartilage restoration: evolution and overview. Clin Orthop
Relat Res 469:2696–2705
disease transmission to mitigate such concerns. Gartland A, Mechler J, Mason-Savas A, MacKay CA, Mailhot
G, Marks SC Jr, Odgren PR (2005) In vitro chondrocyte
differentiation using costochondral chondrocytes as a
Conclusions source of primary rat chondrocyte cultures: an improved
isolation and cryopreservation method. Bone 37:530–544
Giovannini S, Diaz-Romero J, Aigner T, Heini P, Mainil-Varlet
It is possible to isolate viable chondrocytes from P, Nesic D (2010) Micromass co-culture of human articular
cadaveric human donors articular cartilage in samples chondrocytes and human bone marrow mesenchymal stem
processed within the first 48-h. There is not significant cells to investigate stable neocartilage tissue formation
in vitro. Eur Cells Mater 20:245–259
difference between the numbers of chondrocytes Hamada T, Sakai T, Hiraiwa H, Nakashima M, Ono Y, Mit-
isolated from live or cadaveric human donors. Cryop- suyama H, Ishiguro N (2013) Surface markers and gene
reservation of cadaveric primary chondrocytes does expression to characterize the differentiation of monolayer
not alter the capability to form cartilage like tissue. expanded human articular chondrocytes. Nagoya J Med Sci
75:101–111
Co-culture of primary and passaged chondrocytes Hjelle K, Solheim E, Strand T, Muri R, Brittberg M (2002)
enhances the histological quality of new-formed tissue Articular cartilage defects in 1,000 knee arthroscopies.
compared to non-co-cultured cells. Arthrosc J Arthrosc Relat Surg 18:730–734

123
Cell Tissue Bank (2017) 18:369–381 381

Hunziker EB (2002) Articular cartilage repair: basic science and Negri S, Fila C, Farinato S, Bellomi A, Pagliaro PP (2007)
clinical progress. A review of the current status and pro- Tissue engineering: chondrocyte culture on type 1 collagen
spects. Osteoarthritis Cartilage 10:432–463 support. Cytohistological and immunohistochemical study.
Ibarra C, Izaguirre A, Villalobos E, Masri M, Lombardero G, J Tissue Eng Regen Med 1:158–159
Martinez V, Velasquillo C, Meza AO, Guevara V, Ibarra Olivos-Meza A, Fitzsimmons JS, Casper ME, Chen Q, An KN,
LG (2014) Follow-up of a new arthroscopic technique for Ruesink TJ, O’Driscoll SW, Reinholz GG (2010) Pre-
implantation of matrix-encapsulated autologous chondro- treatment of periosteum with TGF-beta1 in situ enhances
cytes in the knee. Arthrosc J Arthrosc Relat Surg the quality of osteochondral tissue regenerated from
30:715–723 transplanted periosteal grafts in adult rabbits. Osteoarthritis
Johnstone B, Alini M, Cucchiarini M, Dodge GR, Eglin D, Cartilage 18:1183–1191
Guilak F, Madry H, Mata A, Mauck RL, Semino CE, Oseni AO, Butler PE, Seifalian AM (2013) Optimization of
Stoddart MJ (2013) Tissue engineering for articular carti- chondrocyte isolation and characterization for large-scale
lage repair—the state of the art. Eur Cells Mater cartilage tissue engineering. J Surg Res 181:41–48
25:248–267 Qing C, Wei-ding C, Wei-min F (2011) Co-culture of chon-
Juhasz T, Matta C, Somogyi C, Katona E, Takacs R, Soha RF, drocytes and bone marrow mesenchymal stem cells in vitro
Szabo IA, Cserhati C, Szody R, Karacsonyi Z, Bako E, enhances the expression of cartilaginous extracellular
Gergely P, Zakany R (2014) Mechanical loading stimulates matrix components. Braz J Med Biol Res 44:303–310
chondrogenesis via the PKA/CREB-Sox9 and PP2A Tew SR, Murdoch AD, Rauchenberg RP, Hardingham TE
pathways in chicken micromass cultures. Cell Signal (2008) Cellular methods in cartilage research: primary
26:468–482 human chondrocytes in culture and chondrogenesis in
Kolettas E, Muir HI, Barrett JC, Hardingham TE (2001) human bone marrow stem cells. Methods 45:2–9
Chondrocyte phenotype and cell survival are regulated by Wei Y, Zeng W, Wan R, Wang J, Zhou Q, Qiu S, Singh SR
culture conditions and by specific cytokines through the (2012) Chondrogenic differentiation of induced pluripotent
expression of Sox-9 transcription factor. Rheumatology stem cells from osteoarthritic chondrocytes in alginate
40:1146–1156 matrix. Eur Cells Mater 23:1–12
Masri M, Lombardero G, Velasquillo C, Martinez V, Neri R, Widuchowski W, Widuchowski J, Trzaska T (2007) Articular
Villegas H, Ibarra C (2007) Matrix-encapsulation cell- cartilage defects: study of 25,124 knee arthroscopies. Knee
seeding technique to prevent cell detachment during 14:177–182
arthroscopic implantation of matrix-induced autologous
chondrocytes. Arthrosc J Arthrosc Relat Surg 23:877–883
McNickle AG, Provencher MT, Cole BJ (2008) Overview of
existing cartilage repair technology. Sports Med Arthrosc
Rev 16:196–201

123

You might also like