ArticlePDF Available

Human Platelet Lysate Can Replace Fetal Calf Serum as a Protein Source to Promote Expansion and Osteogenic Differentiation of Human Bone-Marrow-Derived Mesenchymal Stromal Cells

Authors:

Abstract and Figures

Fetal calf serum (FCS) is frequently used as a growth factor and protein source in bone-marrow-derived mesenchymal stromal cell (BMSC) culture media, although it is a xenogenic product presenting multiple disadvantages including but not limited to ethical concerns. A promising alternative for FCS is human platelet lysate (hPL), which is produced out of human platelet concentrates and happens to be a stable and reliable protein source. In this study, we investigated the influence of hPL in an expansion medium (ESM) and an osteogenic differentiation medium (ODM) on the proliferation and osteogenic differentiation capacity of human BMSC. Therefore, we assessed population doublings during cell expansion, performed alizarin red staining to evaluate the calcium content in the extracellular matrix and determined the activity of alkaline phosphatase (ALP) as osteogenic differentiation correlates. The proliferation rate of BMSC cultured in ESM supplemented with hPL exceeded the proliferation rate of BMSC cultured in the presence of FCS. Furthermore, the calcium content and ALP activity was significantly higher in samples incubated in hPL-supplemented ODM, especially in the early phases of differentiation. Our results show that hPL can replace FCS as a protein supplier in cell culture media and does not negatively affect the osteogenic differentiation capacity of BMSC.
Content may be subject to copyright.
cells
Article
Human Platelet Lysate Can Replace Fetal Calf Serum
as a Protein Source to Promote Expansion and
Osteogenic Dierentiation of Human
Bone-Marrow-Derived Mesenchymal Stromal Cells
Maria Karadjian, Anne-Sophie Senger, Christopher Essers, Sebastian Wilkesmann,
Raban Heller , Joerg Fellenberg , Rolf Simon and Fabian Westhauser *
Center of Orthopedics, Traumatology, and Spinal Cord Injury, Heidelberg University Hospital,
Schlierbacher Landstraße 200a, 69118 Heidelberg, Germany; karadjian.maria@gmail.com (M.K.);
annesophie.senger@gmail.com (A.-S.S.); c.essers@stud.uni-heidelberg.de (C.E.);
sebastianwilkesmann@web.de (S.W.); raban.heller@outlook.com (R.H.);
Joerg.Fellenberg@med.uni-heidelberg.de (J.F.); dr.rolf.simon@web.de (R.S.)
*Correspondence: Fabian.Westhauser@med.uni-heidelberg.de; Tel.: +49-6221-56-25000
Received: 16 February 2020; Accepted: 6 April 2020; Published: 9 April 2020


Abstract:
Fetal calf serum (FCS) is frequently used as a growth factor and protein source in
bone-marrow-derived mesenchymal stromal cell (BMSC) culture media, although it is a xenogenic
product presenting multiple disadvantages including but not limited to ethical concerns. A promising
alternative for FCS is human platelet lysate (hPL), which is produced out of human platelet concentrates
and happens to be a stable and reliable protein source. In this study, we investigated the influence
of hPL in an expansion medium (ESM) and an osteogenic dierentiation medium (ODM) on
the proliferation and osteogenic dierentiation capacity of human BMSC. Therefore, we assessed
population doublings during cell expansion, performed alizarin red staining to evaluate the calcium
content in the extracellular matrix and determined the activity of alkaline phosphatase (ALP) as
osteogenic dierentiation correlates. The proliferation rate of BMSC cultured in ESM supplemented
with hPL exceeded the proliferation rate of BMSC cultured in the presence of FCS. Furthermore, the
calcium content and ALP activity was significantly higher in samples incubated in hPL-supplemented
ODM, especially in the early phases of dierentiation. Our results show that hPL can replace FCS as a
protein supplier in cell culture media and does not negatively aect the osteogenic dierentiation
capacity of BMSC.
Keywords:
osteogenic dierentiation; population doublings; human mesenchymal stromal cells; fetal
calf serum; human platelet lysate
1. Introduction
Despite some limitations, fetal calf serum (FCS) is still the most common source of proteins
and growth factors in cell culture media [
1
]. However, the composition of dierent batches of FCS
varies crucially [
2
,
3
], making it necessary to evaluate dierent batches of FCS before ordering a new
batch to cope with the batch-to-batch variability [
1
,
4
]; this evaluation takes time and resources [
1
].
The influence of FCS as a xenogenic compound of cell culture media on human cells is still not
completely understood [
1
,
5
7
] as its composition diers substantially from the very complex human
serum [
8
11
]. In addition, the conditions of FCS production regarding animal welfare have to be
viewed critically, especially since production circumstances often remain unclear and information
gathering is reported to be complicated [
12
]. This raises the question whether an alternative protein
source standard in cell culture media is required.
Cells 2020,9, 918; doi:10.3390/cells9040918 www.mdpi.com/journal/cells
Cells 2020,9, 918 2 of 12
The most common alternative to FCS is human platelet lysate (hPL) [
1
], which is produced
from human platelets and contains proteins and growth factors necessary for, inter alia, in-vitro
human bone-marrow-derived mesenchymal stromal cell (BMSC) expansion [
13
18
] and osteogenic
differentiation [
15
,
16
,
19
]. hPL products can have smaller batch-to-batch variabilities by pooling multiple
batches of platelet concentrates, hence extensive pre-testing becomes obsolete [
17
,
20
]. It has already been
shown that hPL-supplemented expansion medium (ESM) augments the proliferation rates of BMSC [
11
,
16
18
,
21
29
], and does not affect BMSC properties as part of ESM [
21
]. Several studies evaluated
BMSC differentiation potential after cell expansion in hPL-containing medium, while the differentiation
medium itself contained only FCS as a supplement [
17
,
18
,
22
,
23
,
28
30
]. There have also been multiple
studies reporting that hPL does not affect osteogenic differentiation [
16
,
17
,
21
,
22
,
24
,
27
29
,
31
] or even
increases the osteogenic dierentiation potential of BMSC [
22
,
23
,
25
,
26
]. However, it remains unclear
how hPL as a part of an osteogenic dierentiation medium (ODM) influences the kinetics of the
osteogenic dierentiation of BMSC. Furthermore, the necessary concentrations of hPL reported dier
from 2% to 10% [
18
,
20
,
25
,
26
,
29
34
], pointing out that there is no established standard protocol for hPL
supplementation in ODM for BMSC yet, although the concentration of serum supplement in media
resulting in dierent concentrations of growth factors and other components of the serum influences
cells in culture [1].
The aim of this study was to compare the performance regarding the proliferation rate and
osteogenic dierentiation of BMSC cultured in ESM and ODM containing dierent concentrations of
hPL or FCS and to evaluate the impact of the protein supplement’s concentration on the osteogenic
dierentiation of BMSC. As most available studies on hPL assess its eects on mesenchymal stromal
cells (MSC) during the expansion period, we focused on the eects of hPL as part of ODM on the
kinetics of BMSC osteogenic dierentiation by evaluating alkaline phosphatase (ALP) activity and
calcium deposition as osteogenic dierentiation markers at multiple time points.
2. Materials and Methods
The aims of this study were (i) to compare the impact of either FCS or hPL on the growth of
BMSC under expansion conditions and (ii) to evaluate the osteogenic dierentiation potential of BMSC
cultivated in either FCS or hPL containing ODM. In order to compare the proliferation potential,
population doublings of BMSC incubated in ESM with either 10% FCS or 10% hPL were calculated.
For osteogenic dierentiation experiments, the cells were incubated in ODM (containing either 1% or
10% of FCS or hPL, respectively) and measurement of ALP activity as well as alizarin red staining as
markers for osteogenic dierentiation were performed after incubation periods of 1, 7, 14 and 21 days
(D1, D7, D14, D21). Twelve identical samples were analyzed for each assay at each time point.
BMSC were harvested from the bone marrow washouts of n =10 patients undergoing surgery at
the proximal femur at Heidelberg University Hospital. Prior to bone marrow harvesting, every patient
gave written informed consent. The local ethics committee approved the study (S-443/2015) that was
conducted according to the declaration of Helsinki in its present form. Cells derived from six male
and four female patients with an average age of 50 years (range 29 to 73 years, median 46 years) were
included in the study.
BMSC were isolated from bone marrow, cultivated according to previously published protocols
and pooled in order to reduce inter-individual variability as published previously [
35
,
36
]. During
cultivation and expansion, ESM containing either 10% FCS (Thermo Fisher Scientific, Dreieich, Germany;
lot 42G2082K) according to our standard expansion protocol or 10% of commercially available hPL
(PL BioScience, Aachen, Germany) according to the manufacturer’s instructions for high proliferation
rates was used. The ESM was composed of 25 mM DMEM high-glucose (Thermo Fisher Scientific,
Dreieich, Germany), Penicillin/Streptomycin 100 mg/L (Merck, Darmstadt, Germany), L-Glutamine
200 mM, MEM non-essential amino acids solution, 2-Mercaptoethanol 50 mM (all Thermo Fisher,
Dreieich, Germany) and 4 µg/L fibroblast growth factor two (Abcam, Cambridge, United Kingdom).
Cells 2020,9, 918 3 of 12
BMSC were split after 72 hours of expansion and cultured for a further 96 hours before they were
introduced to osteogenic dierentiation conditions. Cells were then stained with trypan blue and
counted in a Neubauer’s cell chamber. Population doublings within a total incubation period of seven
days of expansion as a parameter for the proliferation rate were calculated using Formula (1).
population doublings =
log N log N0
log 2 (1)
where N =cell number at the end of the expansion period and N0 =cell number at time point zero.
Population doublings of the passages were added in order to obtain cumulative population doublings.
For osteogenic dierentiation, 35,000 BMSC were seeded in a well of a 24-well plate (Thermo Fisher
Scientific, Dreieich, Germany) and incubated in ODM. The ODM was composed of 25 mM DMEM
high-glucose with L-Glutamine (Thermo Fisher Scientific, Dreieich, Germany), Penicillin/Streptomycin
100 mg/L (Merck, Darmstadt, Germany), dexamethasone 0.1
µ
M (Sigma Aldrich, Steinheim, Germany),
ascorbic acid-2-phosphate 2.5 mg/L (Sigma Aldrich, Steinheim, Germany) and
β
-glycerophosphate
10 mM (Merck, Darmstadt, Germany). The growth factor sources in ODM were either FCS or hPL,
added in concentrations of either 1% (according to hPL’s manufacturer’s advice for dierentiation
culture) or 10% according to the well-known FCS standard concentration (Table 1). In the ODM
containing hPL, 2 IU/mL heparin (PL BioScience, Aachen, Germany) were added according to the
manufacturer’s instructions to avoid gel formation of the medium. The cells were incubated for up to
21 days at 37
C, 5% CO
2
following well-established protocols [
36
38
]. Media were changed twice
per week.
Table 1. Overview over the experimental groups.
Group Protein Source and
Concentration in ESM
Protein Source and
Concentration in ODM
F1 FCS 10% FCS 1%
F10 FCS 10% FCS 10%
H1 hPL 10% hPL 1%
H10 hPL 10% hPL 10%
ALP correlates with osteogenic dierentiation of BMSC since it is produced during dierentiation
towards osteoblasts [
37
,
38
]. The cells were washed with 1x PBS (Thermo Fisher Scientific, Dreieich,
Germany), then lysed in 1% Triton buer (Sigma Aldrich, Steinheim, Germany). The lysates were freeze
stored at
80
C until the ALP activity measurement was conducted; after thawing at room temperature
and short centrifugation, the lysates were added to a para-nitrophenylphosphat (p-NPP) (Sigma Aldrich,
Steinheim, Germany) solution for 90 minutes. ALP hydrolyzes p-NPP to para-nitrophenol (p-NP),
causing a change of color to yellow. The extinction of p-NP which corresponds to the ALP activity was
measured photometrically at 405 nm with a reference wavelength of 490 nm using a MRX Microplate
Reader (Dynatech Laboratories, Stuttgart, Germany). ALP activity was normalized to the total protein
content as correlate for the cell number of the samples, determined using the Micro BCA Protein Assay
Kit (Thermo Fisher Scientific, Dreieich, Germany) according to manufacturer’s instructions.
Alizarin red stains the calcium deposits in the extracellular matrix formed by the osteoblastic
(precursor) cells [
38
]. The cells were washed with 1x PBS and then fixed in 70% ethanol (Carl Roth,
Karlsruhe, Germany). After fixation, the ethanol was removed, the cells were washed with distilled
water (Thermo Fisher Scientific, Dreieich, Germany), then stained with alizarin red (Waldeck, Münster,
Germany) and washed again. Finally, the cells were incubated in 10% hexadecylpyridinium (Merck,
Darmstadt, Germany) to dissolve the stained calcium. The extinction of the solution corresponds to
the calcium content of the sample and was measured photometrically at 570 nm.
Cells 2020,9, 918 4 of 12
The experiments were performed on 12 experimental replicates per study group and two technical
replicates per experimental sample.
Statistical analyses were conducted with IBM SPSS Statistics (Version 25; IBM, Mannheim,
Germany) and graphs were created using GraphPad Prism (Version 7, GraphPad Software, La Jolla,
USA). Before further analysis, values were tested for normal distribution by a Shapiro-Wilk test.
Normally distributed samples were tested with the unpaired T-Test; not normally distributed samples
were tested using the Wilcoxon signed-rank test. Results were described as statistically significant for
p<0.05. Unless otherwise stated, dierences mentioned in the text are non-significant. Values are
shown as rounded means with standard deviation (SD) where applicable.
3. Results
3.1. Population Doublings
Cells cultured in 10% hPL-supplemented ESM had significantly (p<0.01) more population
doublings, with an average of 4.46 cumulative population doublings, than cells cultured in 10%
FCS-supplemented ESM with an average of 2.22 cumulative population doublings (Figure 1).
Cells 2020, 9, x FOR PEER REVIEW 4 of 12
Statistical analyses were conducted with IBM SPSS Statistics (Version 25; IBM, Mannheim,
Germany) and graphs were created using GraphPad Prism (Version 7, GraphPad Software, La Jolla,
USA). Before further analysis, values were tested for normal distribution by a Shapiro-Wilk test.
Normally distributed samples were tested with the unpaired T-Test; not normally distributed
samples were tested using the Wilcoxon signed-rank test. Results were described as statistically
significant for p < 0.05. Unless otherwise stated, differences mentioned in the text are non-significant.
Values are shown as rounded means with standard deviation (SD) where applicable.
3. Results
3.1. Population Doublings
Cells cultured in 10% hPL-supplemented ESM had significantly (p < 0.01) more population
doublings, with an average of 4.46 cumulative population doublings, than cells cultured in 10%
FCS-supplemented ESM with an average of 2.22 cumulative population doublings (Figure 1).
Figure 1. Population doublings of BMSC incubated in ESM. Values are presented as means with SD,
* marks significant differences.
3.2. Alkaline Phosphatase Activity
ALP activity kinetics differed among the four groups during the incubation period (Figure 2a).
In the F1 group, ALP activity increased significantly from D1 to its maximum on D14, then remained
on a stable level until D21, showing no significant differences between D14 and D21. The H1 group
increased almost tenfold from D1 to D7, then decreased to D14 to further decrease until D21. The F10
group showed similar kinetics to the H1 group, but presented significantly different values to all
time points. H10 showed its maximum ALP activity on D7, then significantly decreased to D14 to
re-increase until D21. Differences between the F1 and H1 group were significant in the beginning of
differentiation culture on D1 and D7, but showed no significant differences at D14 and D21.
Differences between the H1 and the F10 group were significant at any time. Differences between the
F10 and H10 group were significant on D7 and D21. In both hPL and FCS groups, cells incubated in
the 10% supplemented media showed significantly higher ALP activity than the 1% groups on D7 to
D21, but not on D1 (Figure 2b,c).
Figure 1.
Population doublings of BMSC incubated in ESM. Values are presented as means with SD,
* marks significant dierences.
3.2. Alkaline Phosphatase Activity
ALP activity kinetics diered among the four groups during the incubation period (Figure 2a).
In the F1 group, ALP activity increased significantly from D1 to its maximum on D14, then remained
on a stable level until D21, showing no significant dierences between D14 and D21. The H1 group
increased almost tenfold from D1 to D7, then decreased to D14 to further decrease until D21. The F10
group showed similar kinetics to the H1 group, but presented significantly dierent values to all time
points. H10 showed its maximum ALP activity on D7, then significantly decreased to D14 to re-increase
until D21. Dierences between the F1 and H1 group were significant in the beginning of dierentiation
culture on D1 and D7, but showed no significant dierences at D14 and D21. Dierences between the
H1 and the F10 group were significant at any time. Dierences between the F10 and H10 group were
significant on D7 and D21. In both hPL and FCS groups, cells incubated in the 10% supplemented
media showed significantly higher ALP activity than the 1% groups on D7 to D21, but not on D1
(Figure 2b,c).
Cells 2020,9, 918 5 of 12
Cells 2020, 9, x FOR PEER REVIEW 5 of 12
Figure 2. (a) Alkaline phosphatase activity during time of incubation in ODM in IU/mL of all groups.
(b) ALP activity of FCS groups. (c) ALP activity of hPL groups. Values are shown as means with SD.
* mark significant differences. D = day.
3.3. Alizarin Red Staining
Calcium content increased in all groups over time of incubation. Calcium content in the hPL
groups peaked on day 14, and the FCS groups showed maximum calcium content on day 21 (Figure
3a). During the whole differentiation period, the H1 group showed significantly higher calcium
levels compared to the F1 group. H10 showed significantly higher calcium values at D1, D7 and D14
compared to F10, but lower values at D21 however, the differences on D21 remained
non-significant. When comparing H1 and F10, H1 showed significantly higher values from D1 to
D14 and lower values on D21. When comparing the hPL groups (Figure 3c), H1 showed significantly
higher calcium deposition than H10 on D1 and D7; this relation changed on D14 and D21 where H10
presented significantly higher values than H1. F1 presented the lowest values of all four groups on
days seven to 21, significantly lower than the F10 group (Figure 3b).
Figure 2.
(
a
) Alkaline phosphatase activity during time of incubation in ODM in IU/mL of all groups.
(
b
) ALP activity of FCS groups. (
c
) ALP activity of hPL groups. Values are shown as means with SD.
* mark significant dierences. D =day.
3.3. Alizarin Red Staining
Calcium content increased in all groups over time of incubation. Calcium content in the hPL
groups peaked on day 14, and the FCS groups showed maximum calcium content on day 21 (Figure 3a).
During the whole dierentiation period, the H1 group showed significantly higher calcium levels
compared to the F1 group. H10 showed significantly higher calcium values at D1, D7 and D14
compared to F10, but lower values at D21 – however, the dierences on D21 remained non-significant.
When comparing H1 and F10, H1 showed significantly higher values from D1 to D14 and lower
values on D21. When comparing the hPL groups (Figure 3c), H1 showed significantly higher calcium
deposition than H10 on D1 and D7; this relation changed on D14 and D21 where H10 presented
significantly higher values than H1. F1 presented the lowest values of all four groups on days seven to
21, significantly lower than the F10 group (Figure 3b).
Cells 2020,9, 918 6 of 12
Figure 3.
(
a
) Calcium content after alizarin red staining during time of incubation in ODM in mg/mL of
all groups. (
b
) Calcium content of FCS groups. (
c
) Calcium content of hPL groups. (
d
g
) Cell layer in
24-well plate after alizarin red staining on D7. (
d
) F1. (
e
) F10. (
f
) H1. (
g
) H10. Values are shown as
means with SD. * mark significant dierences.
4. Discussion
The aim of the study was to investigate whether hPL is a valid substitute for FCS in cell culture
media for BMSC, especially in ODM, since FCS shows some limitations. For example, FCS composition
diers essentially from human serum: Shanskii et al. [
11
] showed that hPL has significantly higher
amounts of growth factors like IGF-1 (insulin-like growth factor 1) and PDGF (platelet-derived growth
factor) than FCS. Despite the long-term use of FCS in cell culture, the eects of this xenogenic compound
on human cells in culture remain unclear, also due to the fact that a majority of research on FCS dates
from the past century [
2
,
3
,
8
]. Apart from the mentioned scientific concerns, also an ethical component
has to be taken into account when discussing further usage of FCS: the European Food Safety Authority
(EFSA) published a scientific opinion to, amongst others, answer the central question how to treat
unborn livestock during slaughter, which is relevant for the procedure of FCS harvesting [
39
]. FCS is
produced out of the blood collected by puncturing the heart of unborn, generally non-anesthetized
cattle [
20
,
40
]. According to data obtained from Jochems et al. [
12
], cardiac puncture of the fetus begins
between five and 30 minutes after the death of the dam and the bleeding procedure takes between two
and five minutes. Another concern when it comes to ethical questions is the fact that FCS is mostly
produced outside of the European Union; specifically, the exact origin of commercially available FCS
often remains vague (e.g., origin “South America”) [
12
,
39
41
], making it almost impossible to trace
specific FCS products.
hPL is generally produced out of a varying number of pooled human platelet
concentrates [
13
,
18
,
42
45
] after platelet lysis by freezing—thawing cycles [
20
,
43
45
] or activation
by thrombin [
13
,
42
] to liberate the substances required for cell culture that are stored in platelet
granules [
14
,
15
]. However, there is no good manufacturing practice statement concerning the platelet
lysis method in hPL production [
13
]. The hPL available for scientific purposes can be produced out of
outdated platelet concentrates having no clinical use whilst being still suitable for (in-vitro research)
cell culture [
13
,
20
,
43
]. Furthermore, the hPL composition creates a milieu closer to the physiological
milieu of the human body than FCS does [
8
11
]. Therefore, hPL has favorable properties in terms
Cells 2020,9, 918 7 of 12
of ethics and scientific practice and should seriously be considered as a new standard cell culture
medium substitute.
To evaluate the impact of hPL on cell proliferation, we compared the expansion of BMSC in
ESM supplemented with either 10% FCS or 10% hPL. We could show that BMSC expanded in
hPL-supplemented ESM had more population doublings than their FCS-supplemented counterparts.
However, the approach for evaluating cell proliferation in our study is a basic approach.
For a more detailed evaluation, assays such as the 3H-thymidine incorporation assay or the
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay might provide a more
detailed and standardized quantification of cell proliferation. The promoting eects of hPL on BMSC
proliferation have been described before multiple times, in studies using the aforementioned more
detailed assays; it has been shown that an enhanced proliferation of MSC is one of the main advantages
in hPL supplementation of ESM [
11
,
17
,
21
,
23
,
25
,
26
]. Since our main focus was to assess the influence
of hPL on the osteogenic dierentiation of BMSC, we did not look into proliferation in greater detail.
However, based on the assays performed, the results presented in this study confirm the results
of previous studies that can be explained by the higher amount of growth factors in hPL, favoring
cell proliferation and leading to a higher absolute growth factor amount in cell culture media when
supplemented in the same concentration as FCS [11].
Many preliminary studies evaluated the eects of hPL-supplemented ESM on MSC in order
to assess whether hPL changes MSC characteristics during expansion. Most of these studies
evaluated the immunophenotype of the cells after expansion by analyzing cell surface markers
with fluorescence-activated cell sorting (FACS): stem cell defining surface markers [
46
] were analyzed
in almost any study; the consistent results were that expansion of MSC in hPL-supplemented ESM
does not alter the expression of stem cell defining surface markers in comparison to MSC expanded in
FCS-supplemented ESM [
16
18
,
22
,
23
,
26
,
28
,
29
,
43
,
44
,
47
]. Viau et al. [
18
] and Reis et al. [
47
] assessed the
expression of further surface markers, revealing that the expression of some surface markers diers
significantly between MSC expanded in hPL or FCS, but the majority of the markers are only mildly
influenced by the source of protein supplement in ESM. Some studies additionally performed gene
expression analyses of expanded MSC and revealed that, along with the results of protein expression
of the investigated markers, the ESM’s protein supplement only mildly influences the gene expression
pattern of MSC [
29
,
43
,
44
]. Fernandez-Rebollo et al. [
44
] performed DNA methylation analysis and
revealed no significant dierences between hPL- and FCS-expanded MSC on an epigenetic level.
Viau et al. [
18
] evaluated BMSC morphology by immunofluorescence and FACS after expansion in
hPL-containing ESM and revealed that these cells were smaller and less granular but more homogenous
than their counterparts expanded in the FCS-supplemented ESM. Given the amount of studies that
evaluated MSC characteristics after expansion in hPL-supplemented ESM and the extensiveness of
this aspect, we did not perform any experiments regarding MSC characteristics after expansion in
this study.
In order to evaluate hPL as a substitute for FCS in ODM, we compared the osteogenic dierentiation
of BMSC cultured in ODM supplemented with 1% hPL, 10% hPL, 1% FCS and 10% FCS, respectively.
Alizarin red staining and ALP activity measurement were performed at four time points that cover all
relevant steps of osteogenic dierentiation in the used setting [
48
]. Osteogenic dierentiation increased
over time in all groups. The ALP activity of the BMSC in the hPL groups was higher than or as high
as it was in the matching FCS groups. In addition, BMSC incubated in hPL-supplemented media
presented a calcium content higher than or equal to the FCS groups at any time during cell culture.
It has been shown previously that hPL can have favorable eects on the osteogenic dierentiation of
BMSC [
22
,
23
,
26
], providing every component required from serum necessary for cell culture [
13
19
]
while respecting human serum composition [
8
10
]. However, our study is the first to evaluate the
osteogenic parameters’ evolution under the influence of hPL over the dierentiation period in two
dierent concentrations, making it possible to track the influence of hPL on commonly used time
points during osteogenic dierentiation. This culture setting has been used regularly and covers
Cells 2020,9, 918 8 of 12
the major steps of osteogenic dierentiation by using the specified evaluation time points between
one and 21 days of culture [
36
38
,
49
]: as described previously, the early phases in the dierentiation
of BMSC towards osteoblasts takes place from day five to 14 in culture and is characterized by
ALP expression [
48
,
50
]. The calcification of the extracellular matrix produced during osteoblastic
dierentiation occurs from day 14 to 28 in culture [48,51,52].
The ALP activity of the hPL groups as well as the F10 group reach their maxima on day seven
and decrease afterwards, so the kinetics of cells incubated in hPL- and FCS-supplemented ESM are
similar. Only the F1 group increases until its maximum on day 14 and remains stable until day 21.
Birmingham et al. [
48
] showed that osteoblastic cells show higher ALP activity than riper stages of the
osteoblastic lineage. Considering the fact that H1, H10 and F10 groups reach their maxima earlier, it
could be stated that osteogenic dierentiation under those circumstances happens faster than under
1% FCS supplementation. There are no preliminary data concerning ALP kinetics of BMSC under
hPL supplementation, as photometrical ALP activity measurement of hPL-supplemented BMSC was
hardly performed before. Doucet et al. [
21
] described that ALP activity of BMSC in hPL-supplemented
ODM was comparable to BMSC in FCS-supplemented ODM after 21 days, but no statement regarding
ALP activity kinetics is available. Chevallier et al. [
27
] revealed that hPL can significantly up-regulate
osteogenic genes like ALP in undierentiated BMSC without exogenous osteogenic stimulation, whilst
FCS-incubated BMSC reach the same level of gene expression only after one week of culture in ODM
containing
β
-glycerophosphate and ascorbic acid. This finding could explain the superior ALP activity
of the hPL groups, especially at the beginning of cell culture.
Focusing on the kinetics of extracellular calcium content, it is remarkable that both FCS groups
increase to their maxima on day 21, whereas the hPL groups reach their maxima earlier, on day 14.
Hoemann et al. [
51
] described that in-vitro mineralization normally occurs after two or three weeks of
dierentiation culture, hence both FCS and hPL groups show adequate kinetics, whilst hPL-incubated
BMSC seem to dierentiate faster than their FCS incubated counterparts. It is remarkable that no
significant dierences can be found between the H10 and the F10 group on day 21, implying that the
final calcium content is similar in both groups while only the kinetics dier. Alizarin red staining
is probably the most investigated parameter for osteogenic dierentiation in hPL research, hence
there is a lot of data available [
16
,
22
,
23
,
25
27
,
29
]. However, most studies performed a single time
measurement and did not quantify the calcium content photometrically [
16
,
26
,
27
,
29
]. The studies that
performed quantification of calcium content revealed that hPL-incubated BMSC show higher amounts
of calcium than their FCS-incubated counterparts [22,23], which was confirmed in this study.
Comparing FCS concentrations, it is remarkable that the commonly used concentration of 10% [
1
]
seems to be necessary for adequate osteogenic dierentiation as both ALP activity and calcium content
remain higher (mostly to a significant extent) compared to 1% FCS-supplemented ODM. The hPL
manufacturer recommended 1% hPL supplementation for BMSC dierentiation, which is sucient for
a higher or comparable calcium content compared to 10% FCS (and 10% hPL). However, ALP activity
in the H1 group remains beneath F10 and H10 groups at any time but D1. Interestingly, most studies
determining hPL concentration in the context of osteogenic dierentiation only determined calcium
content [
16
,
22
,
23
,
25
27
,
29
,
33
,
53
]; only a few studies assessed ALP activity [
18
,
21
] and, therefore, did not
detect the described discrepancy in ALP activity and calcium deposition. Only 10% hPL-supplemented
ODM showed a constantly better or equal osteogenic dierentiation compared to FCS. This discrepancy
persists when comparing the absolute results of alizarin red staining and ALP activity measurement:
in alizarin red staining, the hPL groups showed calcium content higher than or as high as the FCS
groups while ALP activity varies more, especially when comparing H1 and F10 groups, where the
F10 group presents higher ALP activity throughout dierentiation from day seven onwards. For the
pairs of H1 vs. F1 and H10 vs. F10, the previously described statement of superiority or equality of the
hPL [16,17,2124,2628,31] is still valid.
According to our results and the data reported in the literature, hPL can be used as a supplement
in ESM and ODM for BMSC, providing higher proliferation rates and higher or equal osteogenic
Cells 2020,9, 918 9 of 12
dierentiation potential. Our study was the first to evaluate the dynamics of ALP activity under the
influence of two dierent concentrations of hPL in ODM, providing additional information about
the kinetics of osteogenic dierentiation under the influence of hPL. Furthermore, we reported a
discrepancy of the impact of hPL in ODM on BMSC between two osteogenic dierentiation parameters
on a cellular/protein level, as there are hardly any studies that have evaluated two quantitative
osteogenic dierentiation parameters before. As we did not perform further assays evaluating
osteogenic dierentiation of BMSC on a genetic level, future studies should also evaluate osteogenic
gene expression in order to obtain a more detailed impression of the impact of hPL in comparison to
FCS on osteogenic dierentiation in one and the same setting.
The concentration of supplementation matters; in our case, only 10% hPL-supplemented ODM
guaranteed an equal or higher osteogenic dierentiation than the common approach using 10% FCS.
However, we were the first study directly comparing two dierent concentrations of hPL in ODM,
therefore further studies should follow in order to approach an optimum hPL concentration in ODM
for BMSC. Considering the higher costs of hPL compared to FCS as well as the fact that the dierences
between the H10 and H1 groups were only detectable in the ALP activity assay, the use of hPL in
smaller concentrations seems to be reasonable in order to avoid a significant increase of the budget
necessary for cell culture experiments [
54
]. Based on the findings of this study, hPL should at least be
considered as a potential alternative to FCS when analyzing osteogenic dierentiation.
5. Conclusions
The main aim of this study was to evaluate the influence of hPL supplementation in dierent
concentrations in an osteogenic dierentiation medium on the osteogenic dierentiation of BMSC,
evaluated by alizarin red staining and ALP activity measurements. As already described in the
literature before, we could show a positive impact of hPL supplementation in an expansion medium
on BMSC population doublings, presenting a proliferation rate almost twice as fast as under FCS
supplementation. Furthermore, we showed that osteogenic dierentiation is not compromised, yet,
favored by a hPL-supplementation in ODM in a concentration-dependent manner; the positive eect is
most visible when hPL concentration is 10% and at the beginning of the dierentiation period, implying
an accelerated, but absolutely comparable osteogenic dierentiation potential of human BMSC.
Author Contributions:
Conceptualization, M.K. and F.W.; Data curation, M.K.; Formal analysis, M.K. and
R.H.; Investigation, M.K., C.E. and S.W.; Methodology, M.K. and A.-S.S.; Project administration, M.K. and
F.W.; Resources, R.S. and F.W.; Supervision, F.W.; Visualization, M.K.; Writing—original draft, M.K. and F.W.;
Writing—review & editing, M.K., A.-S.S., C.E., S.W., R.H., J.F., R.S. and F.W. All authors have read and agreed to
the published version of the manuscript.
Funding: This research received no external funding.
Acknowledgments:
The human platelet lysate used in this study was provided by PL BioScience GmbH, Aachen,
Germany. We acknowledge financial support for open access publishing by the Baden-Württemberg Ministry of
Science, Research and the Arts and by Ruprecht-Karls-Universität Heidelberg.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Mendicino, M.; Bailey, A.M.; Wonnacott, K.; Puri, R.K.; Bauer, S.R. MSC-based product characterization for
clinical trials: An FDA perspective. Cell Stem Cell 2014,14, 141–145. [CrossRef]
2.
Price, P.J.; Gregory, E.A. Relationship between
in vitro
growth promotion and biophysical and biochemical
properties of the serum supplement. In Vitro 1982,18, 576–584. [CrossRef]
3.
Boone, C.W.; Mantel, N.; Caruso, T.D., Jr.; Kazam, E.; Stevenson, R.E. Quality control studies on fetal bovine
serum used in tissue culture. In Vitro 1971,7, 174–189. [CrossRef]
4.
Zheng, X.; Baker, H.; Hancock, W.S.; Fawaz, F.; McCaman, M.; Pungor, E., Jr. Proteomic analysis for the
assessment of dierent lots of fetal bovine serum as a raw material for cell culture. Part IV. Application of
proteomics to the manufacture of biological drugs. Biotechnol. Prog. 2006,22, 1294–1300. [CrossRef]
Cells 2020,9, 918 10 of 12
5.
Sundin, M.; Ringden, O.; Sundberg, B.; Nava, S.; Gotherstrom, C.; Le Blanc, K. No alloantibodies against
mesenchymal stromal cells, but presence of anti-fetal calf serum antibodies, after transplantation in allogeneic
hematopoietic stem cell recipients. Haematologica 2007,92, 1208–1215. [CrossRef]
6.
Knepper, P.A.; Mayanil, C.S.; Goossens, W.; McLone, D.C.; Hayes, E. The presence of transcription factors in
fetal bovine sera. In Vitro Cell. Dev. Biol. Anim. 1998,34, 170–173. [CrossRef]
7.
Gstraunthaler, G. Alternatives to the use of fetal bovine serum: Serum-free cell culture. Altex
2003
,20,
275–281.
8.
Baker, H.; DeAngelis, B.; Frank, O. Vitamins and other metabolites in various sera commonly used for cell
culturing. Experientia 1988,44, 1007–1010. [CrossRef]
9.
Psychogios, N.; Hau, D.D.; Peng, J.; Guo, A.C.; Mandal, R.; Bouatra, S.; Sinelnikov, I.; Krishnamurthy, R.;
Eisner, R.; Gautam, B.; et al. The human serum metabolome. PLoS ONE 2011,6, e16957. [CrossRef]
10.
Anderson, N.L.; Anderson, N.G. The human plasma proteome: History, character, and diagnostic prospects.
Mol. Cell. Proteom. MCP 2002,1, 845–867. [CrossRef]
11.
Shanskii, Y.D.; Sergeeva, N.S.; Sviridova, I.K.; Kirakozov, M.S.; Kirsanova, V.A.; Akhmedova, S.A.;
Antokhin, A.I.; Chissov, V.I. Human platelet lysate as a promising growth-stimulating additive for culturing
of stem cells and other cell types. Bull. Exp. Biol. Med. 2013,156, 146–151. [CrossRef]
12.
Jochems, C.E.; van der Valk, J.B.; Stafleu, F.R.; Baumans, V. The use of fetal bovine serum: Ethical or scientific
problem? Altern. Lab. Anim. ATLA 2002,30, 219–227. [CrossRef]
13.
Schallmoser, K.; Henschler, R.; Gabriel, C.; Koh, M.B.C.; Burnouf, T. Production and Quality Requirements
of Human Platelet Lysate: A Position Statement from the Working Party on Cellular Therapies of the
International Society of Blood Transfusion. Trends Biotechnol. 2020,38, 13–23. [CrossRef]
14.
Rendu, F.; Brohard-Bohn, B. The platelet release reaction: Granules’ constituents, secretion and functions.
Platelets 2001,12, 261–273. [CrossRef]
15.
Nurden, A.T.; Nurden, P.; Sanchez, M.; Andia, I.; Anitua, E. Platelets and wound healing. Front. Biosci. J.
Virtual Libr. 2008,13, 3532–3548. [CrossRef]
16.
Schallmoser, K.; Bartmann, C.; Rohde, E.; Reinisch, A.; Kashofer, K.; Stadelmeyer, E.; Drexler, C.; Lanzer, G.;
Linkesch, W.; Strunk, D. Human platelet lysate can replace fetal bovine serum for clinical-scale expansion of
functional mesenchymal stromal cells. Transfusion 2007,47, 1436–1446. [CrossRef]
17.
Bieback, K.; Hecker, A.; Kocaomer, A.; Lannert, H.; Schallmoser, K.; Strunk, D.; Kluter, H. Human alternatives
to fetal bovine serum for the expansion of mesenchymal stromal cells from bone marrow. Stem Cells
2009
,27,
2331–2341. [CrossRef]
18.
Viau, S.; Lagrange, A.; Chabrand, L.; Lorant, J.; Charrier, M.; Rouger, K.; Alvarez, I.; Eap, S.; Delorme, B.
A highly standardized and characterized human platelet lysate for ecient and reproducible expansion of
human bone marrow mesenchymal stromal cells. Cytotherapy 2019,21, 738–754. [CrossRef]
19.
Kark, L.R.; Karp, J.M.; Davies, J.E. Platelet releasate increases the proliferation and migration of bone
marrow-derived cells cultured under osteogenic conditions. Clin. Oral Implant. Res.
2006
,17, 321–327.
[CrossRef]
20.
Hemeda, H.; Giebel, B.; Wagner, W. Evaluation of human platelet lysate versus fetal bovine serum for culture
of mesenchymal stromal cells. Cytotherapy 2014,16, 170–180. [CrossRef]
21.
Doucet, C.; Ernou, I.; Zhang, Y.; Llense, J.R.; Begot, L.; Holy, X.; Lataillade, J.J. Platelet lysates promote
mesenchymal stem cell expansion: A safety substitute for animal serum in cell-based therapy applications.
J. Cell. Physiol. 2005,205, 228–236. [CrossRef] [PubMed]
22.
Barro, L.; Su, Y.T.; Nebie, O.; Wu, Y.W.; Huang, Y.H.; Koh, M.B.; Knutson, F.; Burnouf, T. A double-
virally-inactivated (Intercept-solvent/detergent) human platelet lysate for
in vitro
expansion of human
mesenchymal stromal cells. Transfusion 2019,59, 2061–2073. [CrossRef] [PubMed]
23.
Xia, W.; Li, H.; Wang, Z.; Xu, R.; Fu, Y.; Zhang, X.; Ye, X.; Huang, Y.; Xiang, A.P.; Yu, W. Human platelet
lysate supports ex vivo expansion and enhances osteogenic dierentiation of human bone marrow-derived
mesenchymal stem cells. Cell Biol. Int. 2011,35, 639–643. [CrossRef]
24.
Prins, H.J.; Rozemuller, H.; Vonk-Grioen, S.; Verweij, V.G.; Dhert, W.J.; Slaper-Cortenbach, I.C.; Martens, A.C.
Bone-forming capacity of mesenchymal stromal cells when cultured in the presence of human platelet lysate
as substitute for fetal bovine serum. Tissue Eng. Part A 2009,15, 3741–3751. [CrossRef]
Cells 2020,9, 918 11 of 12
25.
Lohmann, M.; Walenda, G.; Hemeda, H.; Joussen, S.; Drescher, W.; Jockenhoevel, S.; Hutschenreuter, G.;
Zenke, M.; Wagner, W. Donor age of human platelet lysate aects proliferation and dierentiation of
mesenchymal stem cells. PLoS ONE 2012,7, e37839. [CrossRef]
26.
Horn, P.; Bokermann, G.; Cholewa, D.; Bork, S.; Walenda, T.; Koch, C.; Drescher, W.; Hutschenreuther, G.;
Zenke, M.; Ho, A.D.; et al. Impact of individual platelet lysates on isolation and growth of human
mesenchymal stromal cells. Cytotherapy 2010,12, 888–898. [CrossRef]
27.
Chevallier, N.; Anagnostou, F.; Zilber, S.; Bodivit, G.; Maurin, S.; Barrault, A.; Bierling, P.; Hernigou, P.;
Layrolle, P.; Rouard, H. Osteoblastic dierentiation of human mesenchymal stem cells with platelet lysate.
Biomaterials 2010,31, 270–278. [CrossRef]
28.
Capelli, C.; Domenghini, M.; Borleri, G.; Bellavita, P.; Poma, R.; Carobbio, A.; Mico, C.; Rambaldi, A.; Golay, J.;
Introna, M. Human platelet lysate allows expansion and clinical grade production of mesenchymal stromal
cells from small samples of bone marrow aspirates or marrow filter washouts. Bone Marrow Transplant.
2007
,
40, 785–791. [CrossRef]
29.
Ben Azouna, N.; Jenhani, F.; Regaya, Z.; Berraeis, L.; Ben Othman, T.; Ducrocq, E.; Domenech, J. Phenotypical
and functional characteristics of mesenchymal stem cells from bone marrow: Comparison of culture using
dierent media supplemented with human platelet lysate or fetal bovine serum. Stem Cell Res. Ther.
2012
,3,
6. [CrossRef]
30.
Brun, J.; Abruzzese, T.; Rolaus, B.; Aicher, W.K.; Hart, M.L. Choice of xenogenic-free expansion media
significantly influences the myogenic dierentiation potential of human bone marrow-derived mesenchymal
stromal cells. Cytotherapy 2016,18, 344–359. [CrossRef]
31.
Boraldi, F.; Burns, J.S.; Bartolomeo, A.; Dominici, M.; Quaglino, D. Mineralization by mesenchymal stromal
cells is variously modulated depending on commercial platelet lysate preparations. Cytotherapy
2018
,20,
335–342. [CrossRef]
32.
Riis, S.; Nielsen, F.M.; Pennisi, C.P.; Zachar, V.; Fink, T. Comparative Analysis of Media and Supplements on
Initiation and Expansion of Adipose-Derived Stem Cells. Stem Cells Transl. Med.
2016
,5, 314–324. [CrossRef]
33.
Pachler, K.; Lener, T.; Streif, D.; Dunai, Z.A.; Desgeorges, A.; Feichtner, M.; Oller, M.; Schallmoser, K.;
Rohde, E.; Gimona, M. A Good Manufacturing Practice-grade standard protocol for exclusively human
mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy 2017,19, 458–472. [CrossRef]
34.
Mohammadi, S.; Nikbakht, M.; Malek Mohammadi, A.; Zahed Panah, M.; Ostadali, M.R.; Nasiri, H.;
Ghavamzadeh, A. Human Platelet Lysate as a Xeno Free Alternative of Fetal Bovine Serum for the In Vitro
Expansion of Human Mesenchymal Stromal Cells. Int. J. Hematol.-Oncol. Stem Cell Res. 2016,10, 161–171.
35.
Schmitz, S.I.; Widholz, B.; Essers, C.; Becker, M.; Tulyaganov, D.U.; Moghaddam, A.; Gonzalo de Juan, I.;
Westhauser, F. Superior biocompatibility and comparable osteoinductive properties: Sodium-reduced
fluoride-containing bioactive glass belonging to the CaO-MgO-SiO2 system as a promising alternative to
45S5 bioactive glass. Bioact. Mater. 2020,5, 55–65. [CrossRef]
36.
Widholz, B.; Tsitlakidis, S.; Reible, B.; Moghaddam, A.; Westhauser, F. Pooling of Patient-Derived
Mesenchymal Stromal Cells Reduces Inter-Individual Confounder-Associated Variation without Negative
Impact on Cell Viability, Proliferation and Osteogenic Dierentiation. Cells 2019,8, 633. [CrossRef]
37.
Reible, B.; Schmidmaier, G.; Moghaddam, A.; Westhauser, F. Insulin-Like Growth Factor-1 as a Possible
Alternative to Bone Morphogenetic Protein-7 to Induce Osteogenic Dierentiation of Human Mesenchymal
Stem Cells in Vitro. Int. J. Mol. Sci. 2018,19, 1674. [CrossRef]
38.
Reible, B.; Schmidmaier, G.; Prokscha, M.; Moghaddam, A.; Westhauser, F. Continuous stimulation with
dierentiation factors is necessary to enhance osteogenic dierentiation of human mesenchymal stem cells
in-vitro. Growth Factors 2017,35, 179–188. [CrossRef]
39.
More, S.J.; Bicout, D.J.; Bøtner, A.; Butterworth, A.; Calistri, P.; Depner, K.R.; Edwards, S.A.; Garin-Bastuji, B.;
Good, M.; Schmidt, C.G.; et al. Animal welfare aspects in respect of the slaughter or killing of pregnant
livestock animals (cattle, pigs, sheep, goats, horses). EFSA J. 2017,15, e04782.
40.
Tekkatte, C.; Gunasingh, G.P.; Cherian, K.M.; Sankaranarayanan, K. "Humanized" stem cell culture techniques:
The animal serum controversy. Stem Cells Int. 2011,2011, 504723. [CrossRef]
41.
Gstraunthaler, G.; Lindl, T.; van der Valk, J. A plea to reduce or replace fetal bovine serum in cell culture
media. Cytotechnology 2013,65, 791–793. [CrossRef]
Cells 2020,9, 918 12 of 12
42.
Strunk, D.; Lozano, M.; Marks, D.C.; Loh, Y.S.; Gstraunthaler, G.; Schennach, H.; Rohde, E.;
Laner-Plamberger, S.; Oller, M.; Nystedt, J.; et al. International Forum on GMP-grade human platelet
lysate for cell propagation: Summary. Vox Sang. 2018,113, 80–87. [CrossRef]
43.
Pierce, J.; Benedetti, E.; Preslar, A.; Jacobson, P.; Jin, P.; Stroncek, D.F.; Reems, J.A. Comparative analyses of
industrial-scale human platelet lysate preparations. Transfusion 2017,57, 2858–2869. [CrossRef]
44.
Fernandez-Rebollo, E.; Mentrup, B.; Ebert, R.; Franzen, J.; Abagnale, G.; Sieben, T.; Ostrowska, A.;
Homann, P.; Roux, P.F.; Rath, B.; et al. Human Platelet Lysate versus Fetal Calf Serum: These Supplements
Do Not Select for Dierent Mesenchymal Stromal Cells. Sci. Rep. 2017,7, 5132. [CrossRef]
45.
Schallmoser, K.; Strunk, D. Preparation of pooled human platelet lysate (pHPL) as an ecient supplement
for animal serum-free human stem cell cultures. J. Vis. Exp. JoVE 2009. [CrossRef]
46.
Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.;
Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells.
The International Society for Cellular Therapy position statement. Cytotherapy 2006,8, 315–317. [CrossRef]
47.
Reis, M.; McDonald, D.; Nicholson, L.; Godthardt, K.; Knobel, S.; Dickinson, A.M.; Filby, A.; Wang, X.N.
Global phenotypic characterisation of human platelet lysate expanded MSCs by high-throughput flow
cytometry. Sci. Rep. 2018,8, 3907. [CrossRef]
48.
Birmingham, E.; Niebur, G.L.; McHugh, P.E.; Shaw, G.; Barry, F.P.; McNamara, L.M. Osteogenic dierentiation
of mesenchymal stem cells is regulated by osteocyte and osteoblast cells in a simplified bone niche.
Eur. Cells Mater. 2012,23, 13–27. [CrossRef]
49.
Westhauser, F.; Widholz, B.; Nawaz, Q.; Tsitlakidis, S.; Hagmann, S.; Moghaddam, A.; Boccaccini, A.R.
Favorable angiogenic properties of the borosilicate bioactive glass 0106-B1 result in enhanced
in vivo
osteoid
formation compared to 45S5 Bioglass. Biomater. Sci. 2019,7, 5161–5176. [CrossRef]
50.
Aubin, J.E. Regulation of osteoblast formation and function. Rev. Endocr. Metab. Disord.
2001
,2, 81–94.
[CrossRef]
51.
Hoemann, C.D.; El-Gabalawy, H.; McKee, M.D.
In vitro
osteogenesis assays: Influence of the primary cell
source on alkaline phosphatase activity and mineralization. Pathol. Biol.
2009
,57, 318–323. [CrossRef]
[PubMed]
52.
Huang, Z.; Nelson, E.R.; Smith, R.L.; Goodman, S.B. The sequential expression profiles of growth factors from
osteoprogenitors [correction of osteroprogenitors] to osteoblasts
in vitro
.Tissue Eng.
2007
,13, 2311–2320.
[CrossRef] [PubMed]
53.
Hemeda, H.; Kalz, J.; Walenda, G.; Lohmann, M.; Wagner, W. Heparin concentration is critical for cell culture
with human platelet lysate. Cytotherapy 2013,15, 1174–1181. [CrossRef] [PubMed]
54.
Pons, M.; Nagel, G.; Zeyn, Y.; Beyer, M.; Laguna, T.; Brachetti, T.; Sellmer, A.; Mahboobi, S.; Conradi, R.;
Butter, F.; et al. Human platelet lysate as validated replacement for animal serum to assess chemosensitivity.
Altex 2019,36, 277–288. [CrossRef]
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... FBS is most frequently used in a concentration of 10% for MC-MSC cocultures (5). For MSC expansion and osteogenic differentiation, 10% and 5% hPL are the most frequently used concentration (23,24,26). The 2.5% hPL concentration was added based on the hPL manufacturer's advice for MC cultures. ...
... While the use of hPL to replace FBS has been widely studied for human MSC cultures and osteogenic differentiation of these cells (19)(20)(21)(22)(23)(24)(25)(26), the influence of hPL on osteoclastic differentiation of MCs is relatively unknown. Platelet-released supernatants could stimulate osteoclast differentiation and activity of human peripheral blood mononuclear cells (PBMCs) (27). ...
... Also, for osteoclast-osteoblast co-cultures with human MCs and MSCs as progenitors, aiming at mimicking human bone remodeling and related pathologies in vitro¸FBS is still the common standard (5). hPL has been suggested as alternative for FBS and is already frequently studied for MSC propagation and osteogenic differentiation of these cells (19)(20)(21)(22)(23)(24)(25)(26). In contrast, the effect of hPL on osteoclastic differentiation of MCs is relatively unknown. ...
Article
Full-text available
Introduction To study human physiological and pathological bone remodeling while addressing the principle of replacement, reduction and refinement of animal experiments (3Rs), human in vitro bone remodeling models are being developed. Despite increasing safety-, scientific-, and ethical concerns, fetal bovine serum (FBS), a nutritional medium supplement, is still routinely used in these models. To comply with the 3Rs and to improve the reproducibility of such in vitro models, xenogeneic-free medium supplements should be investigated. Human platelet lysate (hPL) might be a good alternative as it has been shown to accelerate osteogenic differentiation of mesenchymal stromal cells (MSCs) and improve subsequent mineralization. However, for a human in vitro bone model, hPL should also be able to adequately support osteoclastic differentiation and subsequent bone resorption. In addition, optimizing co-culture medium conditions in mono-cultures might lead to unequal stimulation of co-cultured cells. Methods We compared supplementation with 10% FBS vs. 10%, 5%, and 2.5% hPL for osteoclast formation and resorption by human monocytes (MCs) in mono-culture and in co-culture with (osteogenically stimulated) human MSCs. Results and Discussion Supplementation of hPL can lead to a less donor-dependent and more homogeneous osteoclastic differentiation of MCs when compared to supplementation with 10% FBS. In co-cultures, osteoclastic differentiation and resorption in the 10% FBS group was almost completely inhibited by MSCs, while the supplementation with hPL still allowed for resorption, mostly at low concentrations. The addition of hPL to osteogenically stimulated MSC mono- and MC-MSC co-cultures resulted in osteogenic differentiation and bone-like matrix formation, mostly at high concentrations. Conclusion We conclude that hPL could support both osteoclastic differentiation of human MCs and osteogenic differentiation of human MSCs in mono- and in co-culture, and that this can be balanced by the hPL concentration. Thus, the use of hPL could limit the need for FBS, which is currently commonly accepted for in vitro bone remodeling models.
... Some studies have shown that the replacement of FCS with HPL may have a positive effect on osteogenic differentiation. In a study conducted by Karadjian and colleagues, it was shown that there was an increased ALP activity and calcification in human MSCs due to the exchange of FCS with HPL in the differentiation medium [64]. Moreover, Mollentze and coworkers discussed in their review that the utilization of xeno-free differentiation modulators, such as HPL, should be considered in in vivo studies with prospects for later clinical applications [65]. ...
Article
Full-text available
Sex-related differences are a current topic in contemporary science. In addition to hormonal regulation, cell-autonomous mechanisms are important in bone homeostasis and regeneration. In this study, human skeletal stem cells (SSCs) from female and male adults were cultured and analyzed with immunological assays and osteogenic differentiation assessments. Female SSCs exhibited a mean doubling time of 100.6 h, whereas male SSCs displayed a mean doubling time of 168.0 h. Immunophenotyping revealed the expression of the stem cell markers Nestin, CD133, and CD164, accompanied by the neural-crest marker SOX9. Furthermore, multiparameter flow cytometric analyses revealed a substantial population of multipotent SSCs, comprising up to 80% in both sexes. An analysis of the osteogenic differentiation potential demonstrated a strong mineralization in both male and female SSCs under physiological conditions. Recognizing the prevailing association of bone diseases with inflammatory processes, we also analyzed the osteogenic potential of SSCs from both sexes under pro-inflammatory conditions. Upon TNF-α and IL-1β treatment, we observed no sexual dimorphism on osteogenesis. In summary, we demonstrated the successful isolation and characterization of SSCs capable of rapid osteogenic differentiation. Taken together, in vitro cultured SSCs might be a suitable model to study sexual dimorphisms and develop drugs for degenerative bone diseases.
... In a clinical trial, autologous chondrocyte implantation was most successful under PL supplementation, showing faster regeneration and a lower inflammatory response compared to FBS supplemented groups [41]. Osteogenic differentiation of MSC was described to be enhanced in PL [42], however, in another study [43], no superior effects could be observed in osteogenic differentiation of equine bone marrow MSC under PL substitution, despite increased TGF-β1 concentration. In our study, ADSC that were cultured in PL, proliferated much faster in FBS supplemented medium, often leading to the detachment of cell layers during the differentiation period, hence not all groups could be analysed. ...
Article
Full-text available
Chronic wounds depict a silent epidemic challenging medical professionals worldwide. Regenerative medicine uses adipose-derived stem cells (ADSC) in promising new therapies. In this study, platelet lysate (PL) as a xenogen-free substitute for foetal bovine serum (FBS) in ADSC culture was used to create an ADSC secretome containing cytokines for optimal wound healing conditions. The ADSC secretome was tested on keratinocytes for migrational behaviour and viability. Therefore, human ADSC were characterized under FBS (10%) and PL (5% and 10%) substitution, regarding morphology, differentiation, viability, gene and protein expression. ADSC were then cultured in 5% PL and their secretome was used for stimulation of keratinocyte migration and viability. To enhance the effect, ADSC were treated with Epithelial Growth Factor (EGF, 100 ng/mL) and hypoxia (1% O₂). In both PL and FBS groups, ADSC expressed typical stem cell markers. PL induced a significantly higher increase in cell viability compared to FBS substitution. ADSC secretome contained various beneficial proteins which enhance the wound healing capacity of keratinocytes. This could be optimized treating ADSC with hypoxia and EGF. In conclusion, the study shows that ADSC cultivated in 5 % PL can effectively support wound healing conditions and can be considered as a promising new therapy for individual treatment of chronic wound disorders.
... hPL in culture medium accelerated the proliferation rate of hNDP-SCs at the beginning of the cultivation (2nd passage-5th passage). hPL-cultivated hNDP-SCs showed approximately two times shorter PDT ( [26,36,39,40,43,44]. At the end of cell growth, we observed the prolongation of population doubling time in the hPL-treated group. ...
Article
Full-text available
Despite several scientific or ethical issues, fetal bovine serum (FBS) remains the standard nutrient supplement in the mesenchymal stem cell cultivation medium. Cell amplification plays an important role in human stem cell therapies. Increasing interest in this field has supported attempts to find suitable human alternatives to FBS for in vitro cell propagation. Human platelet lysate (hPL) has recently been determined as one of them. Our study aimed to evaluate the influence of 2% hPL in the growth medium for in vitro expansion of human natal dental pulp stem cells (hNDP-SCs). The effect was determined on proliferation rate, viability, phenotype profile, expression of several markers, relative telomere length change, and differentiation potential of four lineages of hNDP-SCs. As a control, hNDP-SCs were simultaneously cultivated in 2% FBS. hNDP-SCs cultivated in hPL showed a statistically significantly higher proliferation rate in initial passages. We did not observe a statistically significant effect on mesenchymal stem cell marker (CD29, CD44, CD73, CD90) or stromal-associated marker (CD13, CD166) expression. The cell viability, relative telomere length, or multipotency remained unaffected in hNDP-SCs cultivated in hPL-medium. In conclusion, hPL produced under controlled and standardized conditions is an efficient serum supplement for in vitro expansion of hNDP-SCs.
... 7 In this context, human platelet lysate (hPL), composed of the platelet content released upon lysis, can be an alternative to replace FCS. [8][9][10][11] In mesenchymal stromal cells (MSC) culture, for example, it enhances proliferation, promoting the attainment of higher cell numbers in a shorter period. [12][13][14] This occurs because when activated in vivo, platelets undergo degranulation, releasing coagulation factors, adhesion molecules, growth factors, immune-related molecules, chemokines, and several other substances, which are either soluble or inside granules. ...
Article
Following health agencies warning, the use of animal origin supplements should be avoided in biological products proposed as therapy in humans. Platelet lysate and several other growth factors sources are alternatives to replace fetal calf serum, the current gold standard in clinical‐grade cell culture. However, the platelet supplement's content lacks data due to different production methods. The principle behind these products relays on the lysis of platelets that release several proteins, some of which are contained in heterogeneous granules and coordinate biological functions. This study aims to analyze the composition and reproducibility of a platelet lysate produced with a standardized method, by describing several batches' protein and particle content using proteomics and dynamic light scattering. Proteomics data revealed a diversified protein content, with some related to essential cellular processes such as proliferation, morphogenesis, differentiation, biosynthesis, adhesion, and metabolism. It also detected proteins responsible for activation and binding of transforming growth factor beta, hepatocyte growth factor, and insulin‐like growth factor. Total protein, biochemical, and growth factors quantitative data showed consistent and reproducible values across batches. Novel data on two major particle populations is presented, with high dispersion level at 231 ± 96 d.nm and at 30 ± 8 d.nm, possibly being an important way of protein trafficking through the cellular microenvironment. This experimental and descriptive analysis aims to support the content definition and quality criteria of a cell supplement for clinical applications.
Chapter
The field of regenerative medicine encompasses various treatment strategies including gene therapy, cell therapy and the use of bioengineered tissue constructs. Although the applications of regenerative medicine involving living cells have demonstrated promising results in clinical trials, challenges in the manufacturing of reproducible cell material are still present. Mesenchymal stem/stromal cells (MSCs) are involved in over a thousand of clinical trials for treating a broad spectrum of conditions and ex vivo expansion of MSCs plays a crucial role in the treatment outcome. In this chapter, we discuss key parameters of in vitro MSCs culture such as media composition, scale-up strategies, cultivation under physiological oxygen tensions and three-dimensional cultivation. We demonstrate how these parameters influence cell physiology and functioning, and underline the importance of cultivation conditions for the preservation of all possible native functions of these cells during ex vivo cultivation. Graphical abstract. (Created with Biorender.com)
Article
Background: The use of mesenchymal stem cells (MSCs) for the development of tissue-engineered constructs has advanced in recent years. However, future clinically approved products require following good manufacturing practice (GMP) guidelines. This includes using alternatives to xenogeneic-derived cell culture supplements to avoid rejection of the transplants. Consequently, human platelet lysate (PLT) has been adopted as an affordable and effective alternative to foetal bovine serum (FBS) in traditional 2D cultures. However, little is known about its effect in more advanced 3D culture systems. Methods: We evaluated bone marrow MSCs (BMSCs) proliferation and CD marker expression in cells expanded in FBS or PLT-supplemented media. Differentiation capacity of the BMSCs expanded in the presence of the different supplements was evaluated in 3D type I collagen hydrogels. Furthermore, the effects of the supplements on the process of differentiation were analyzed by using qPCR and histological staining. Results: Cell proliferation was greater in PLT-supplemented media versus FBS. BMSCs expanded in PLT showed similar osteogenic differentiation capacity in 3D compared with FBS expanded cells. In contrast, when cells were 3D differentiated in PLT they showed lower osteogenesis versus the traditional FBS protocol. This was also the case for adipogenic differentiation, in which FBS supplementation was superior to PLT. Conclusions: PLT is a superior alternative to FBS for the expansion of MSCs without compromising their subsequent differentiation capacity in 3D. However, differentiation in PLT is impaired. Thus, PLT can be used to reduce the time required to expand the necessary cell numbers for development of 3D tissue engineered MSC constructs.
Article
Full-text available
There is a demand to design microparticles holding surface topographies while presenting inherent bioactive cues for applications in the biomedical and biotechnological fields. Using the pool of proteins present in human‐derived platelet lysates (PL), it is reported the production of protein‐based microparticles via a simple and cost‐effective method, exploring the prone redox behavior of cysteine (‐SH) amino acid residues. The forced formation of new intermolecular disulfide bonds results in the precipitation of the proteins as spherical, pompon‐like microparticles with adjustable sizes (15‐50 μm in diameter) and surface topography consisting of grooves and ridges. These PL microparticles exhibit extraordinary cytocompatibility, allowing cell‐guided micro‐aggregates to form, while also working as injectable systems for cell support. Early studies also suggests that the surface topography provided by these PL microparticles can support osteogenic behavior. Consequently, these PL microparticles may find use to create live tissues via bottom‐up procedures or injectable tissue‐defect fillers, particularly for bone regeneration, with the prospect of working under xeno‐free conditions. This article is protected by copyright. All rights reserved
Chapter
This chapter is an overview of the derivation and application of stem cells in orthopedics. The identification of stem cells such as the stem cell types, characteristics, and their differentiation and tissue forming ability toward cartilage, bone, tendon, and muscle are introduced. The clinical application relevant cell types including bone marrow‐derived mesenchymal stem cells (BMSCs), adipose‐derived mesenchymal stem cells (ADSCs), tissue‐specific stem/progenitor cells, and mesenchymal stem cell‐derived extracellular vesicles (MSC‐EVs) and exosomes and their application scenarios in orthopedics are further summarized. Then, the potential application strategies and clinical translation concerns of stem cell‐based therapies are discussed. At last, several recent interesting clinical results in the treatment of bone, cartilage, tendon–ligament, and spinal diseases, such as treating bone defects, non‐union, osteochondral defect, osteoarthritis, anterior cruciate ligament reconstruction, spinal cord injuries, and spinal fusion are highlighted.
Article
Objective : Tissue engineering is promising for dental and craniofacial regeneration. The objectives of this study were to develop a novel xeno-free alginate-fibrin-platelet lysate hydrogel with human periodontal ligament stem cells (hPDLSCs) for dental regeneration, and to investigate the proliferation and osteogenic differentiation of hPDLSCs using hPL as a cell culture nutrient supplement. Methods : hPDLSCs were cultured with Dulbecco's modified eagle medium (DMEM), DMEM + 10% fetal bovine serum (FBS), and DMEM + hPL (1%, 2.5%, and 5%). hPDLSCs were encapsulated in alginate-fibrin microbeads (Alg+Fib), alginate-hPL microbeads (Alg+hPL), or alginate-fibrin-hPL microbeads (Alg+Fib+hPL). hPDLSCs encapsulated in alginate microbeads were induced with an osteogenic medium containing hPL or FBS. Quantitative real-time polymerase chain reaction (qRT-PCR), alkaline phosphatase (ALP) activity, ALP staining, and alizarin red (ARS) staining was investigated. Results : hPDLSCs were released faster from Alg+Fib+hPL than from Alg+hPL. At 14 days, ALP activity was 44.1 ± 7.61 mU/mg for Alg+Fib+hPL group, higher than 28.07 ± 5.15 mU/mg of Alg+Fib (p<0.05) and 0.95 ± 0.2 mU/mg of control (p<0.01). At 7 days, osteogenic genes (ALP, RUNX2, COL1, and OPN) in Alg+Fib+hPL and Alg+Fib were 3-10 folds those of control. At 21 days, the hPDLSC-synthesized bone mineral amount in Alg+Fib+hPL and Alg+Fib was 7.5 folds and 4.3 folds that of control group, respectively. Conclusions : The 2.5% hPL was determined to be optimal for hPDLSCs. Adding hPL into alginate hydrogel improved the viability of the hPDLSCs encapsulated in the microbeads. The hPL-based medium enhanced the osteogenic differentiation of hPDLSCs in Alg+Fib+hPL construct, showing a promising xeno-free approach for delivering hPDLSCs to enhance dental, craniofacial and orthopedic regenerations.
Article
Full-text available
Bioactive glasses (BGs) are promising bone substitute materials. However, under certain circumstances BGs such as the well-known 45S5 Bioglass® (composition in wt%: 45.0 SiO 2 , 24.5 Na 2 O, 24.5 CaO, 6.0 P 2 O 5) act cytotoxic due to a strong increase in pH caused by a burst release of sodium ions. A potential alternative is a sodium-reduced fluoride-containing BG belonging to the CaO-MgO-SiO 2 system, namely BG1d-BG (composition in wt%: 46.1 SiO 2 , 28.7 CaO, 8.8 MgO, 6.2 P 2 O 5 , 5.7 CaF 2 , 4.5 Na 2 O), that has already been evaluated in-vitro, in-vivo and in preliminary clinical trials. Before further application, however, BG1d-BG should be compared to the benchmark amongst BGs, the 45S5 Bioglass® composition, to classify its effect on cell viability, proliferation and osteogenic differentiation of human mesenchymal stem cells (MSCs). Therefore, in this study, the biocompat-ibility and osteogenic potential of both BGs were investigated in an indirect and direct culture setting to assess the effect of the ionic dissolution products and the BGs' physical presence on the cells. The results indicated an advantage of BG1d-BG over 45S5 Bioglass® regarding cell viability and proliferation. Both BGs induced an earlier onset of osteogenic differentiation and accelerated the expression of late osteoblast marker genes compared to the control group. In conclusion, BG1d-BG is an attractive candidate for further experimental investigation. The basic mechanisms behind the different impact on cell behavior should be assessed in further detail, e.g. by further alteration of the BG compositions.
Article
Full-text available
Patient-derived mesenchymal stromal cells (MSCs) play a key role in bone tissue engineering. Various donor-specific factors were identified causing significant variability in the biological properties of MSCs impairing quality of data and inter-study comparability. These limitations might be overcome by pooling cells of different donors. However, the effects of pooling on osteogenic differentiation, proliferation and vitality remain unknown and have, therefore, been evaluated in this study. MSCs of 10 donors were cultivated and differentiated into osteogenic lineage individually and in a pooled setting, containing MSCs of each donor in equal parts. Proliferation was evaluated in expansion (assessment of generation time) and differentiation (quantification of dsDNA content) conditions. Vitality was visualized by a fluorescence-microscopy-based live/dead assay. Osteogenic differentiation was assessed by quantification of alkaline phosphatase (ALP) activity and extracellular calcium deposition. Compared to the individual setting, generation time of pooled MSCs was shorter and proliferation was increased during differentiation with significantly lower variances. Calcium deposition was comparable, while variances were significantly higher in the individual setting. ALP activity showed high variance in both groups, but increased comparably during the incubation period. In conclusion, MSC pooling helps to compensate donor-dependent variability and does not negatively influence MSC vitality, proliferation and osteogenic differentiation.
Article
Full-text available
Experiments with cultured mammalian cells represent an in vitro alternative to animal experiments. Fetal calf serum (FCS) is the most commonly used medium supplement worldwide. FCS contains a variable mixture of growth factors and cyto­kines that support cell proliferation. This undefined nature of FCS is a source of experimental variation, undesired immune responses, possible contaminations and, because of the way it is obtained, an ethical concern. Thus, alternative, defined, valid, and reliable medium supplements should be characterized in a large number of experiments. Human platelet lysate (hPL) is increasingly appreciated as an alternative to FCS. Since it is unclear whether cells respond differentially to clin­ically relevant chemotherapeutics inducing replicative stress and DNA damage (hydroxyurea, irinotecan), induction of reactive oxygen species (ROS), the tyrosine kinase inhibitor (TKi) imatinib, and novel epigenetic modifiers belonging to the group of histone deacetylase inhibitors (HDACi), we investigated these issues. Here we show that cancer cells derived from leukemia and colon cancer grow very similarly in culture media supplemented with FCS or outdated hPL. Notably, cells have practically identical proteomes under both culture conditions. Moreover, cells grown with FCS or hPL responded equally to all types of drugs and stress conditions that we tested. In addition, the transfection of blood cells by electroporation can be achieved under both conditions. Furthermore, we reveal that class I HDACs, but not HDAC6, are required for the expression of the pan-leukemic marker WT1 under various culture conditions. Hence, hPL is a moderately priced substitute for FCS in various experimental settings.
Article
Full-text available
Growth factors and mesenchymal stem cells (MSC) support consolidation of bone defects. Bone Morphogenetic Protein-7 (BMP-7) has been used clinically and experimentally, but the outcomes remain controversial. Increased systemic expression of Insulin-like Growth Factor-1 (IGF-1) significantly correlates with successful regeneration of bone healing disorders, making IGF-1 a promising alternative to BMP-7. There is no experimental data comparing the osteoinductive potential of IGF-1 and BMP-7. Therefore, in this study, the influence of IGF-1 and BMP-7 in different concentrations on the osteogenic differentiation of two human MSC-subtypes, isolated from reaming debris (RMSC) and iliac crest bone marrow (BMSC) has been assessed. A more sensitive reaction of BMSC towards stimulation with IGF-1 in concentrations of 400–800 ng/mL was found, leading to a significantly higher degree of osteogenic differentiation compared to stimulation with BMP-7. RMSC react more sensitively to stimulation with BMP-7 compared to BMSC. Lower concentrations of IGF-1 were necessary to significantly increase osteogenic differentiation of RMSC and BMSC compared to BMP-7. Therefore, IGF-1 should be considered as a valuable option to improve osteogenic differentiation of MSC and merits further experimental consideration. The MSC subtype and method of differentiation factor application also have to be considered, as they affect the outcome of osteogenic differentiation.
Article
Full-text available
Mesenchymal stromal cells (MSCs) are a promising cell source to develop cell therapy for many diseases. Human platelet lysate (PLT) is increasingly used as an alternative to foetal calf serum (FCS) for clinical-scale MSC production. To date, the global surface protein expression of PLT-expended MSCs (MSC-PLT) is not known. To investigate this, paired MSC-PLT and MSC-FCS were analysed in parallel using high-throughput flow cytometry for the expression of 356 cell surface proteins. MSC-PLT showed differential surface protein expression compared to their MSC-FCS counterpart. Higher percentage of positive cells was observed in MSC-PLT for 48 surface proteins, of which 13 were significantly enriched on MSC-PLT. This finding was validated using multiparameter flow cytometry and further confirmed by quantitative staining intensity analysis. The enriched surface proteins are relevant to increased proliferation and migration capacity, as well as enhanced chondrogenic and osteogenic differentiation properties. In silico network analysis revealed that these enriched surface proteins are involved in three distinct networks that are associated with inflammatory responses, carbohydrate metabolism and cellular motility. This is the first study reporting differential cell surface protein expression between MSC-PLT and MSC-FSC. Further studies are required to uncover the impact of those enriched proteins on biological functions of MSC-PLT.
Article
The 45S5-bioactive glass (BG) composition is the most commonly investigated amongst BG-based bone substitutes. By changing BG compositions and by addition of therapeutically active ions such as boron, the biological features of BGs can be tailored towards specific needs and possible drawbacks can be overcome. The borosilicate glass 0106-B1 (composition in wt%: 37.5 SiO2, 22.6 CaO, 5.9 Na2O, 4.0 P2O5, 12.0 K2O, 5.5 MgO, 12.5 B2O3) has demonstrated pro-angiogenic properties. However, the osteogenic performance of the 0106-B1-BG and its influence on cell viability and proliferation in-vitro as well as its osteogenic and angiogenic properties in-vivo have not been investigated. Therefore, in this study, the impact of 0106-B1-BG and 45S5-BG on osteogenic differentiation, viability and proliferation on human mesenchymal stromal cells (MSCs) was assessed in-vitro. Furthermore, MSC-seeded scaffolds made from both BG types were implanted subcutaneously in immunodeficient mice for 10 weeks. Osteoid formation was quantified by histomorphometry, vascularization was visualized by immunohistological staining. Additionally, the in-vivo expression patterns of genes correlating with osteogenesis and angiogenesis were analyzed. In-vitro, the impact of 45S5-BG and 0106-B1-BG on the proliferation, viability and osteogenic differentiation of MSCs was comparable. In-vivo, scaffolds made from 0106-B1-BG significantly outperformed the 45S5-BG-based scaffolds regarding the amount and maturation of the osteoid. Furthermore, 0106-B1-BG-based scaffolds showed significantly increased angiogenic gene expression patterns. In conclusion, the beneficial angiogenic properties of 0106-B1-BG result in improved osteogenic properties in-vivo, making the 0106-B1-BG a promising candidate for further investigation, e.g. in a bone defect model.
Article
https://www.cell.com/trends/biotechnology/fulltext/S0167-7799(19)30151-9. Human platelet lysate (HPL), rich in growth factors, is an efficient alternative supplement to fetal bovine serum for ex vivo propagation of stromal cell-based medicinal products. Since 2014, HPL is a focus of the Working Party for Cellular Therapies of the International Society of Blood Transfusion (ISBT). Currently, as several GMP compliant manufacturing protocols exist, an international consensus defining the optimal modes of industrial production, product specification, pathogen safety and release criteria of this ancillary material is needed. This opinion paper by the ISBT Working Party summarizes the current knowledge on HPL production and proposes recommendations on manufacturing and quality management in line with current technological innovations and regulations of biological products and advanced therapy medicinal products.
Article
Background: Human platelet lysate (hPL) represents a powerful alternative to fetal bovine serum (FBS) for human mesenchymal stromal cell (hMSC) expansion. However, the large variability in hPL sources and production protocols gives rise to discrepancies in product quality, characterization and poor batch-to-batch standardization. Methods: hPL prepared with more than 200 donors (200+DhPL) or with five donors (5DhPL) were compared in terms of growth factor (GF) contents and biochemical analysis. A multiple protein assay and proteomic analysis were performed to further characterize 200+DhPL batches. We also compared the phenotypic and functional characteristics of bone marrow (BM)-hMSCs grown in 200+DhPL versus FBS+basic fibroblast growth factor (bFGF). Results: By contrast to 5DhPL, industrial 200+DhPL displayed a strong standardization of GF contents and biochemical characteristics. We identified specific plasmatic components and platelet-released factors as the most relevant markers for the evaluation of the standardization of hPL batches. We used a multiplex assay and proteomic analysis of 200+DhPL to establish a proteomic signature and demonstrated the robust standardization of batches. 200+DhPL was shown to improve and standardize BM-hMSC expansion compared with FBS+bFGF. The levels of expression of BM-hMSC membrane markers were found to be much more homogeneous between batches when cells were cultured in 200+DhPL. BM-hMSCs cultured in parallel under both conditions displayed similar adipogenic and osteogenic differentiation potential and immunosuppressive properties. Conclusions: We report a standardization of hPL and the importance of such standardization for the efficient amplification of more homogeneous and reproducible cell therapy products.
Article
BACKGROUND Pooled human platelet lysate (HPL) can replace fetal bovine serum (FBS) as xeno‐free supplement for ex vivo expansion of mesenchymal stromal cells (MSCs). We evaluate here whether a double‐virally‐inactivated HPL (DVI‐HPL) prepared from expired Intercept‐treated platelet concentrates (PCs) and treated by solvent/detergent (S/D) can be used for MSC expansion. STUDY DESIGN AND METHODS Expired Intercept‐treated PCs in 65% platelet (PLT) additive solution were pooled and subjected to a 1% tri‐n‐butyl phosphate/1% Triton X‐45 treatment followed by soybean oil, hydrophobic interaction chromatography purification, and sterile filtration. Bone marrow–derived MSCs (BM‐MSCs) were expanded for four passages in growth medium containing 10% DVI‐HPL, I‐HPL (from Intercept‐PC only), untreated HPL, and FBS. MSC morphology, doubling time, immunophenotype, immunosuppressive activity, and differentiation capacity were compared. RESULTS Expanded cells had typical spindle morphology and showed higher viability in all HPL conditions than in FBS. The DVI‐HPL and FBS‐expanded cells were morphologically larger than in I‐HPL and HPL supplements. The cumulative population doubling was lower using DVI‐HPL than with HPL and I‐HPL, but significantly higher than using FBS. Immunophenotype was not affected by the supplements used. Immunosuppressive activity was maintained with all supplements. Differentiation capacity into chondrocytes and osteocytes was more effective in DVI‐HPL but less toward adipocytes compared to other supplements. CONCLUSIONS Human PLT lysate made from Intercept‐PCs subjected to S/D treatment may be an alternative to untreated HPL and to I‐HPL for BM‐MSC expansion. This finding reinforces the potential of HPL as a virally safe alternative to FBS for clinical grade MSC expansion protocols.