Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Acta Biomaterialia 177 (2024) 132–147

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actbio

Full length article

Characterization of the molecular composition and in vitro


regenerative capacity of platelet-based bioproducts and related
subfractions
Andrea Acebes-Huerta a,b, Patricia Martínez-Botía a, Graciela Carbajo-Argüelles a,
Judit Fernández-Fuertes a,c,d, María Carmen Muñoz-Turrillas a,e, Ana María Ojea-Pérez f,
Antonio López-Vázquez g, Johannes A. Eble h, Laura Gutiérrez a,b,∗
a
Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
b
Department of Medicine, University of Oviedo, Spain
c
Department of Orthopedics and Trauma Surgery, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
d
Department of Surgery and Medical Surgical Specialties, University of Oviedo, Spain
e
Centro Regional de Transfusión de Toledo-Guadalajara, Spain
f
Centro Comunitario de Sangre y Tejidos de Asturias, Oviedo, Spain
g
Department of Immunology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
h
Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Germany

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

Article history: The use and demand of platelet-based bioproducts in regenerative medicine is steadily increasing. How-
Received 13 March 2023 ever, it is very difficult to establish the real clinical benefits of these therapies, as the lack of charac-
Revised 29 December 2023
terization and detailed production methods of platelet-based bioproducts persists in the literature and
Accepted 19 January 2024
precludes cross-study comparisons. We characterized the molecular composition and in vitro regenerative
Available online 3 February 2024
capacity of platelet-rich plasma (PRP) produced in a closed-system. Furthermore, we performed a parallel
Keywords: characterization on different PRP subfractions (plasma and plasma-free platelet lysate), identifying that
Platelets the fractions containing platelet-derived cargo exert the most potent regenerative capacity. This observa-
Platelet rich plasma (PRP) tion led us to develop a method to obtain a platelet secretome highly enriched in growth factors, free of
Plasma plasma and cellular components (PCT/IB2022/057936), with the aim of establishing a superior bioprod-
Platelet secretome uct. The molecular characterization of secretomes revealed agonist-dependent differences, which corre-
Bioactive factors
lates with beneficial grades of regenerative capacity. Importantly, secretomes showed general superiority
Growth factors
Wound healing
to PRP in vitro. We discuss the variables influencing the bioproduct quality (inter-donor variation, platelet
Regenerative medicine source and processing methods). Finally, we propose that the characteristics of secretomes circumvents
certain limitations of PRP (autologous vs allogeneic), and envision that optimizing post-processing proto-
cols (nanoencapsulation, lyophilization), would allow their clinical application even beyond regenerative
medicine.

Statement of significance

The use and demand of platelet-based bioproducts in regenerative medicine is steadily increasing. How-
ever, it is very difficult to establish the real clinical benefits of these therapies, or to improve/personalize
them, as the lack of characterization of the bioproducts and their production methods is a constant in
the literature, reason that precludes cross-study comparisons. In the present manuscript, we provide a
comprehensive molecular and functional characterization of platelet-based bioproducts and subfractions,
including platelet rich plasma, plasma fractions and platelet secretomes produced with a methodology
developed by our group. Our results show that the molecular composition of each fraction correlates


Corresponding author at: Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Edificio FINBA, Platelet Research Lab NO-F15, Avenida del Hospital Univer-
sitario S/N, 33011 Oviedo, Spain.
E-mail address: gutierrezglaura@uniovi.es (L. Gutiérrez).

https://doi.org/10.1016/j.actbio.2024.01.029
1742-7061/© 2024 The Author(s). Published by Elsevier Ltd on behalf of Acta Materialia Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

with its regenerative capacity in vitro. Thus, a rigorous characterization of platelet-derived bioproducts
will potentially allow universal use, customizing and new applications.
© 2024 The Author(s). Published by Elsevier Ltd on behalf of Acta Materialia Inc.
This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)

medicine, among others [22–24]. However, the therapeutic out-


1. Introduction come of the use of PRP remains highly controversial due to a lack
of consensus on PRP preparation, post-production processing and
Platelets are circulating anucleate blood components (2-4 μm application methods [25]. As a consequence, the quality of PRPs
in diameter) and key players in maintaining the body hemosta- varies considerably amongst studies in terms of platelet number,
sis [1]. Noteworthy, they also participate in other physiological and presence or absence of white/red blood cells, and molecular com-
pathological processes beyond hemostasis and thrombosis, such as position, which also lacks characterization [26]. This adds to the
immunomodulation, lymph and blood vessel separation during de- fact that in some studies, PRP is applied either fresh (with living
velopment, angiogenesis and tumor metastasis [2–6]. Hemostatic cellular components), or after freezing or activation which induces
platelet function and communication with the coagulation cas- platelet lysis or platelet granule cargo release. Furthermore, in PRP-
cade is regulated by receptor-mediated signaling networks leading like bioproducts, the total platelet proteome is contained within a
to platelet adhesion, secretion and aggregation [7]. As a result of mixture of plasma proteins, of which the actual bio-active compo-
vessel damage, the exposed subendothelial and secreted adhesive nents (i.e., those contained in platelet granules) represent a rather
proteins (i.e., collagen and von Willebrand factor), are recognized small sub-fraction of the product. Another important issue con-
by platelets through their receptors, which induces their activa- cerns the regulatory aspects. In most territories, the application of
tion and adhesion to the site of injury, cytoskeletal rearrangements PRP is only allowed in an autologous manner. This requires that the
(filopodia and lamellipodia formation), integrin activation (essen- patient must schedule a blood donation to prepare the PRP prior
tial for platelet aggregation), and secretion of the platelet granule application. However, it has been reported that certain pathological
cargo or secretome [8–10]. conditions might influence platelet quality and reactivity, and an
The platelet secretome consists of various sorts of molecules, autologous PRP might not always be the most suitable option for
which will assure the amplification of the platelet activation sig- therapy, since it could result in uncertain clinical outcomes, due to
nal, the recruitment of circulating blood cells such as leukocytes inter-donor as well as pathological- or subclinical-dependent vari-
and fibroblasts to the site of injury, and the repair of the injured ations [27–29].
vessel. These molecules are packed in three major types of gran- We propose that a better understanding of the nature and the
ules, which differ in content, biogenesis, trafficking, and exocyto- active components of platelet-based bioproducts is crucial to opti-
sis [11]. The alpha granules (α -granules) are the most abundant mize and standardize production methods as to improve the clini-
(approximately 50-80 per platelet) and contain the proteins that cal benefit of the therapy.
comprise the secretome core, i.e., membrane-associated receptors In the present manuscript we characterize the molecular com-
(such as α IIbβ 3 and P-selectin) and soluble cargo (growth fac- position of PRPs from different donors produced in a standardized
tors, cytokines, anti- and pro-angiogenic factors, proteases, etc.) closed-system using the infrastructure of a certified blood bank
that participate in a wide variety of biological processes, includ- and processed by snap-freezing to allow platelet lysis [30] and as-
ing hemostasis, inflammation, cancer and wound healing [10,12- sociate it to their in vitro regenerative capacity. Furthermore, we
14]. Some of these molecules are synthesized and packaged into α - delve into the contribution of different PRP subfractions (plasma
granules in the megakaryocytes prior to platelet formation, while and plasma-free platelet lysate), that have been individually char-
others, such as fibrinogen, clotting factor V and VEGF (among oth- acterized at the molecular level, to the observed regenerative ca-
ers) are (also) endocytosed and incorporated into α -granules ei- pacity. The observation that the bioproduct and subfractions that
ther by megakaryocytes or by circulating platelets [15–19]. Dense exert the most potent regenerative capacity are those contain-
granules (δ -granules) are the second most abundant granule type ing platelet-derived cargo, led us to develop a method to obtain
found in platelets, and contain small molecules such as serotonin, a platelet secretome enriched bioproduct (PLT-S), free of plasma
calcium and ADP, which serve to amplify platelet activation. Lastly, and cellular components (PCT/IB2022/057936). We deepen into the
platelets also contain lysosomes (1-3 per platelet), rich in acid molecular and functional characterization of PLT-Ss and compare
hydrolases (cathepsins, hexosaminidase, β -galactosidase, arylsulfa- them with PRP and PRP subfractions. We further discuss the vari-
tase, β -glucuronidase and acid phosphatase) [11]. Thus, the platelet ables influencing the final quality of PLT-Ss (i.e., inter-donor vari-
secretome plays a central role in the regulation of hemostasis ation, platelet biological source and processing) and the intrinsic
by driving cell-to-cell interactions and tissue regeneration during characteristics that would allow further developments of platelet-
wound healing. Additionally, it recently emerged that both the based bioproducts with clinical therapeutic purposes even beyond
qualitative and quantitative composition of the secretome, as well regenerative medicine.
as its release dynamics, may be more complex and orchestrated
than anticipated [10,14,20,21]. 2. Material and methods
In recent years, the specific healing property of platelets and
their cargo has been used as rationale for the development of The study did not require recruitment of patients or healthy
platelet-based bioproducts as adjuvant and stand-alone treatment donors, as we used interrupted blood donations or discard material
in advanced cell therapy and regenerative medicine. Platelet-rich from the Local Blood Bank (Centro Comunitario de Sangre y Tejidos
plasma (PRP) is the most popular platelet-based bioproduct and de Asturias – CCSTA). Therefore, informed consent or approval by
its therapeutic application constitutes a relatively new approach the ethics committee was not required per se in this setting. The
in regenerative medicine, with clinical benefits in a wide range line of research to which this study belongs has obtained approval
of medical fields, such as odontology and maxillofacial surgery, by the Local Medical Ethical Committee (#33/19, Hospital Universi-
ophthalmology, traumatology, sports and aesthetics, and veterinary tario Central de Asturias, Oviedo, Spain). The study was conducted

133
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

according to the Declaration of Helsinki, and all materials and sam- by incubating the platelets on ice. To collect the PLT-Ss, the differ-
ples used were anonymized. ent platelet-activation reactions were immediately centrifuged at
40 0 0 rpm for 5 minutes. The supernatant was transferred to a new
2.1. Production of PRP and PRP-derived subfractions tube, and centrifuged at 10k rpm for 2 minutes to eliminate cell
debris. Supernatants were collected through a 0.22 μm pore filter,
Platelet-rich plasma (PRP) was obtained from interrupted blood aliquoted and stored frozen at -80°C until further use.
donations (n=14). The fractionation was performed using the Alternatively, we prepared secretomes from reactions contain-
equipment of a certified blood bank, as previously described [30]. ing 6 × 108 platelets/mL, to match the average platelet concentra-
Briefly, the plasma fraction containing platelets was separated from tion of closed system PRPs and named them Supra PLT-SPRP . The bio-
the fraction containing red blood cells after centrifugation (1130 logical source used for the preparation of Supra PLT-SPRP was closed-
rpm for 5 minutes, without brake) with a press Compomat G5 sys- system PRP (Fig. 1C).
tem (Fresenius-Kabi) into a separate bag. A second centrifugation
(2500 rpm for 12 minutes, brake 4) was performed to obtain an 2.3. Quality control of secretomes: flow cytometry analysis of platelet
enriched PRP fraction, after removal of the upper phase, the so- degranulation
called platelet poor plasma (PPP), into a separate bag. The PRP
was then distributed into 3 pediatric bags (each of them contain- In order to analyze platelet degranulation capacity upon stimu-
ing approximately 10 mL), and stored at −40°C. When indicated, lation, platelets were incubated 10 minutes at room temperature
a complete blood count was performed prior freezing (Sysmex with a panel of surface markers (CD9-Alexa Fluor 647; CD62P-
XN-10/XN-20, Hematology Analyzer). For experiments and molec- PE; CD63-PECy7), before and after stimulation as described above
ular characterization, PRP samples were subject to 2 additional (section 2.2, PLT-S production). After fixation with formaldehyde
freezing-thawing cycles to assure complete platelet lysis (-20°C - to a final concentration of 0.5%, samples were analyzed by flow
room temperature) [31] and then, centrifuged at 10k rpm for 2 cytometry. Platelets were gated based on forward/side scatter and
minutes, to discard cell debris. The supernatants were collected CD9 positivity. The cut-off was set on unstimulated samples, and
through a 0.22 μm pore filter, aliquoted and stored at -80°C un- the increase of CD62P and CD63 expression were measured. All an-
til use (see Fig. 1A). tibodies were purchased from BD Biosciences. Data were acquired
To prepare PRP-derived subfractions, we used PRP produced on a FACS Aria IITM flow cytometer (BD Biosciences) and analyzed
as described above from 4 different donors. We kept samples of using FlowJo software (Tree Star, Ashland, OR, USA).
the original PRPs and respective PPPs, and further processed the
remaining PRP fraction by centrifugation (40 0 0 rpm, 5 minutes).
The supernatant was collected and tentatively named PRP Plasma, 2.4. Production of platelet-count customized PRP and PRP-derived
and the pelleted platelets were resuspended in the same origi- subfractions
nal volume of HEPES Glucose buffer (132 mM NaCl, 6 mM KCl, 1
mM MgSO4 , 1.2 mM KH2 PO4 , 20mM HEPES, pH 7.4, containing 5 Since the PLT-Ss were prepared from reactions containing
mM D-glucose) to produce Plasma-free Platelet Lysate (PFP-L) (see 2 × 108 platelets/mL, in some comparison studies we used PRP
Fig. 1B). The PPP and PRP Plasma fractions were centrifuged at 10k and PFP-L produced with the same number of platelets, namely
Custom PRP and Custom PFP-L (Fig. 1D). In brief, we counted the num-
rpm for 2 minutes, to discard cell debris, and supernatants were
collected through a 0.22 μm pore filter and kept frozen in aliquots ber of platelets in closed-system PRP samples prepared as de-
at -80°C until further use. The PRP and PFP-L fractions were sub- scribed above, and resuspended the desired number of platelets
ject to 2 freezing-thawing cycles (-20°C – room temperature), and after centrifugation at 40 0 0 rpm for 5 minutes, with either
PRP Plasma from the same sample (Custom PRP) or HEPES Glucose
then, centrifuged at 10k rpm for 2 minutes to eliminate cell debris.
Then, the supernatants were collected through a 0.22 μm pore fil- Buffer (Custom PFP-L). After 2 freezing-thawing cycles (-20°C – room
ter, aliquoted and stored frozen at -80°C until further use. temperature), samples were centrifuged at 10k rpm for 2 min-
utes to eliminate cell debris. Supernatants were collected through
2.2. Platelet secretome (PLT-S) production a 0.22 μm pore filter, aliquoted and stored frozen at -80°C until
further use.
The method to obtain PLT-S is protected by international
patent application (PCT/IB2022/057936). In brief, we prepared PLT- 2.5. In vitro scratch wound closure assays
S from two different biological sources: interrupted blood dona-
tions (whole blood -PLT-SWB -; n=4) and closed-system PRP (PLT- The regenerative potential of the different bioproducts was an-
SPRP ; n=4), (see Fig. 1C). We first collected the PRP fraction from alyzed in vitro by scratch wound closure assays. To this end, MRC-
whole blood samples by differential centrifugation (10 0 0 rpm for 5 human fetal lung fibroblast cells were seeded in IbiTreat plates
15 minutes, without brake). We counted the platelets in the PRP (μ-Plate 24-well 2-chamber insert Black, Ibidi), at a density of
fraction from whole blood and in closed-system PRP samples us- 3.5 × 105 cells/mL (70 μL volume per insert chamber) and incu-
ing an automated hematology analyzer (Sysmex XN-10/XN-20). We bated to allow cell attachment until reaching 80-90% confluence
washed the platelets by centrifuging at 40 0 0 rpm for 5 min- (approximately 24 hours) in DMEM supplemented with 1% peni-
utes, and resuspended them in HEPES Glucose Buffer to a con- cillin/streptomycin (P/S) and 10% Fetal Bovine Serum (FBS). Then,
centration of 2 × 108 platelets/mL. Subsequently, platelets were the silicone insert was carefully removed, leaving a 500 μm cell-
activated with the following agonists: TRAP6 (100 μM; BACHEM free gap (according to the manufacturerś datasheet). After wash-
H-8365.0 0 05), convulxin (0.625 ng/ml; ENZO Life Sciences ALX- ing away detached cells, culture media supplemented with 10% of
350-100-C050), phorbol 12-myristate 13-acetate (PMA; 100 ng/mL; platelet-based bioproduct (in the presence or not of 10% FBS, or
Sigma-Aldrich P8139-5MG), aggretin A (6.53 nM), ristocetin (0.5 in the presence or not of 2 U/mL of heparin to avoid the clot-
mg/mL; Sigma-Aldrich R7752-100MG) or collagen (30 μg/mL and ting, as indicated per experiment) was added. Cell-free gaps were
90 μg/mL; Sigma-Aldrich C4243-20ML), under constant stirring monitored for 48 hours using a time-lapse microscopy Zeiss Axio
during 5 minutes, at 37°C, using a thermomixer (Mixing Block MB- Observer Z1 microscope (Carl Zeiss, Germany) equipped with a
102; BIOER). We did not use platelet aggregation inhibitors such as ‘stage incubator’, keeping the temperature and the CO2 concentra-
apyrase and prostaglandin E2 (PGE2), and did not stop the reaction tion constant (37°C and 5% CO2 ). Imaging was performed with a

134
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Fig. 1. Schematic illustration of the production process of the different platelet-derived bioproducts used in the study. A. Closed-system PRP preparation using the infrastruc-
ture of a local blood bank. B-D. Schematic illustration of the processing method to obtain: (B) PRP subfractions, (C) platelet-derived secretomes (PLT-S) and (D) custom-made
PRP and PFP-L alternatives.

135
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Plan-Apochromat 10x/0.45 M27 (dt=2.1mm) objective. Mosaic im- amongst groups. Pearson’s rank correlation test was used to an-
ages (3 × 3) were taken every 12 hours (AxioCam MRm; Carl alyze the correlation between the biological parameters. Unsuper-
Zeiss). Fields were selected with micrometric precision (in a three- vised agglomerative hierarchical clustering was performed to study
dimensional manner, axes xyz) using the Tiles tool (z-position from the similarities between the different samples and conditions using
Tiles Setup) from ZEN 3.2 Blue edition software (Carl Zeiss). The the correlation distance and average linkage. The number of clus-
acquisition software creates automatically the image composite. In ters was estimated using the silhouette method and taking into ac-
some cases, when the cell confluency is not homogeneous or when count the biological information. Briefly, the silhouette coefficient
clean areas are identified, the software can make errors in overlap- estimates the average distance between clusters, and plots them
ping images; when applying automatic correction for these errors, over values of k ranging. Those with the highest coefficients rep-
a brightness shift appears in the overlap area, without affecting resent the best clustering. For the representation of the cluster-
cell localization and image analysis. The images were subsequently ing analysis, Principal Component Analysis (PCA), dendrograms and
analyzed using Fiji/ImageJ software (NIH) with the Montpellier heatmaps were plotted after scale-to-interval normalization. When
Ressources Imagerie (MRI) “Wound healing tool” macro avail- indicated, the confounding effect introduced by the inter-donor
able online at: https://github.com/MontpellierRessourcesImagerie/ variation was removed using a linear model to fit the data using
imagej_macros_and_scripts/wiki/Wound- Healing- Tool. The plugin the removeBatchEffect function from the limma package [32]. Sta-
analyzes the area of a wound in a cellular tissue. From a stack tistical analysis was performed with R version 4.2.1 (R Core Team,
of images representing a time-series, it measures the reduction of 2022) and Microsoft Excel. Figures were designed or partly cre-
the cleared space between the wound edges (cell-free gap). The ated using Servier Medical Art (provided by Servier, licensed un-
percentage of gap closure was calculated and compared between der a Creative Commons Attribution 3.0 unported license), Canva
controls and treatment groups. (https://www.canva.com/es_es/), R and Adobe Illustrator CS4.

2.6. Secretome stability and consumption in vitro 3. Results

To evaluate the time- and temperature-dependent factor stabil- 3.1. The regenerative capacity of PRPs produced using a closed system
ity and consumption of PLT-S in our culture setting, MRC-5 hu- correlates to their molecular composition
man fetal lung fibroblast cells were seeded in conventional 24-well
plates or using μ-Plate 24-well 2-chamber insert (Ibidi), at a den- Platelet-rich plasma (PRP) refers to many types of platelet-
sity of 3.5 × 105 cells/mL (3.5 × 105 or 0.49 × 105 total cells, re- based bioproducts, without making any distinction on their prepa-
spectively) and incubated to allow cell attachment in standard con- ration and processing method, or their cellular and molecular com-
ditions. After this, the medium was replaced by fresh culture me- position, variables that surely influence their clinical outcome. In a
dia without serum (DMEM+1% P/S) supplemented with 10% PLT- collaborative effort to offer a standardized production procedure,
S. Supernatants were collected after 24 and 48 hours. As controls, we previously described the implementation of a closed-system
we took supernatants of cells incubated in the absence of PLT-S, PRP preparation method using the infrastructure of our local blood
and of cultures devoid of cells but supplemented with PLT-Ss. The bank [30]. As part of the quality control of the process, we assessed
concentration of a panel of bioactive factors in the collected super- the concentration of some growth factors (GFs): EGF, HGF, PDGF-
natants from all conditions was analyzed by Luminex Technology, BB, VEGF-A, VEGF-D and FGF-23, confirming at the molecular level
as described below (Section 2.7). the acknowledged inter-donor variation of PRP-like products [30].
In order to assess the potential correlation of the molecular
2.7. Multiplex immunoassay composition of PRPs with their regenerative capacity, we first de-
cided to comprehensively characterize the PRPs (n=14) from dif-
Molecular characterization of platelet bioproducts and frac- ferent donors produced using the described method, extending the
tions was performed using high-performance multiplex immunoas- list of GFs, cytokines, chemokines and immune modulators to 37
say following the manufacturer’s instructions (Human Procartaplex different molecules, which were measured using Multiplex Tech-
customized panels; Invitrogen, Carlsbad, CA, USA). The plates were nology. Our results showed that PRP sample clustering was donor-
read in a Luminex® 200 Instrument System using the xPONENT dependent, meaning that the molecular composition varied across
software (Luminex Corporation, USA). The specific factors analyzed them, but stayed essentially identical among the aliquots from the
were: Caspase-3, CD40L, EGF, FGF-2, FGF-23, G-CSF (CSF-3), GM- same donor (Fig. 2A). These data support not only the notion of
CSF, GITRL, Granzyme B, GROα (KC/CXCL1), HGF, ICAM-1, IFN-γ , IL- inter-donor variation, but also the previously described homogene-
1α , IL-1β , IL-2, IL-6, IL-7, IL-8 (CXCL8), IL-10, MIP-1α (CCL3), MCP- ity in composition of the three aliquots prepared from the same
1 (CCL2), MCP-2 (CCL8), MCP-3 (CCL7), MIP-1β (CCL4), Osteopon- donor [30], which assures that a patient requiring a regime ther-
tin (OPN), PDGF-BB, PECAM-1, P-Selectin, RANTES (CCL5), SDF-1α , apy with serial applications is going to receive a PRP product of the
Thrombopoietin (TPO), TGF-β , TNF-α , VCAM-1, VEGF-A and VEGF- same quality. Principal Component Analysis (PCA) of the molecular
D. composition of PRPs further supports these results, as PRPs from
For factor stability and consumption studies the specific panel different individuals segregate, while aliquots from the same PRP
of factors analyzed were: EGF, G-CSF, GM-CSF, HGF, IFNg, MCP-1, donor cluster together (Fig. S1A).
PDGF-BB, SDF-1a and VEGF-A. In order to evaluate whether the platelet count of PRPs may in-
fluence the inter-donor differences identified in factor composition,
2.8. Statistical analysis we measured it on 10 PRPs from different donors (before the freez-
ing step to induce platelet lysis), and correlated it to the concentra-
When comparisons between all possible pairs of groups were tions of molecules measured with Multiplex Technology. Counter-
performed, the Tukey Honest Significant Differences (HSD) test was intuitively to what it could be anticipated, only the concentration
used; while the Dunnett procedure was chosen when several com- of 4 out of 37 molecules correlated significantly with platelet num-
parisons against a control group were involved. Both statistical ber, specifically EGF, VEGF-A, MIP-1β and Gro-α (Fig. 2B), while
methods control the family-wise error rate stemming from multi- for the other factors measured, no correlation was observed. These
ple testing. Additionally, when single pairwise comparisons were results led us to hypothesize that, on the one hand, the PRPs ob-
performed, a Studentś t-test was employed to compare means tained with this method may have an optimal platelet yield en-

136
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Fig. 2. In-depth analysis of closed-system PRP. A. Heatmap depicting the hierarchical clustering of PRP samples after scale-to-interval normalization, based on their molecular
composition as measured by Multiplex Technology. The dendrogram on the top of the heatmap shows clustering of the samples based on their differential composition.
PRPs used in wound healing assays are indicated with asterisks at the bottom. B. Scatter plots representing the Pearson’s correlation between the platelet count and the
concentration (pg/mL) of EGF, Gro-α , VEGF-A and MIP-1α . C. Principal Component Analysis (PCA) of PRP samples used for wound healing assays based on their molecular
composition as measured by Multiplex Technology. D. Line graphs depicting the % of Scratch Covered Area (SCA) of wound healing assays at 12, 24, 36 and 48 hours. Control
and PRP-treated cultures, in the presence of 2 U/mL heparin, are shown. Experiments were carried out in standard culture conditions (supplemented with 10% FBS, left
graph) or in the absence of FBS (right graph). Data are presented as the mean + standard deviation (SD) of experimental replicates (n=3) from each culture condition, with
the exception of cultures treated with PRP 2 and PRP 5 (+ FBS), and PRP 6 (- FBS), where only the mean is represented (n=2). ∗ p<0.05; ∗ ∗ p<0.01; ∗ ∗ ∗ p<0.001. E. Graphs
representing the concentration of factors in PRP samples that induce low (PRP 1, 5 and 6) or high (PRP 2, 7, 10 and 12) proliferative responses compared to control cultures,
in the presence of FBS. These factors are the significant ones separating PRP 1 and PRP 5 from the rest in the PCA (Dim 2; B). Values measured in PRP 6 and PRP 10 are
indicated.

137
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

richment range, given that the donors are eligible for the blood be equal amongst the two plasma subfractions, our results sug-
donation (i.e., there are not sensible differences in platelet count gest that PPP contains a low amount of factors in general, while
between PRPs), and therefore, platelet-count related differences PRP Plasma (which is the subfraction containing the plasma ob-

would not be significant. Substantiating this notion, previous re- tained from a third centrifugation performed on the PRP bioprod-
sults in gonarthrosis patients receiving closed-system PRP, showed uct) contains a higher amount of some factors at levels compa-
that this standardized production method ensures a PRP product rable to those measured in PRP (Fig. 3A, Fig. S2A and Table S1).
with sufficient quality to produce positive clinical responses [33]. These results suggest that these factors that are in higher con-
On the other hand, the presence of plasma proteins may limit centration in the PRP Plasma subfractions may be actively released
the detection of certain molecules making it difficult to observe a by platelets as a result of the additional processing procedure.
sensitive correlation of the concentration of factors to the platelet These factors include GFs (i.e., EGF, PDGF, VEGF-D or HGF) and
count as measured on individual PRPs. immunomodulators (interleukins and chemokines). On the other
Next, we performed in vitro wound healing/closure assays. We hand, when comparing the PRP Plasma fraction with PRP and PFP-L
tested available PRP samples with different molecular composi- samples, the molecules that remained retained within the platelets
tion as evidenced by their hierarchical clustering (Fig. 2A, C and become evident (Fig. 3A). The comparison of the composition of
Fig. S1A). We stimulated cultures of MRC-5 cells, in the presence different subfractions allowed us to identify that ICAM-1/VCAM-1
of FBS, with or without the different PRPs and the wound clo- and OPN/RANTES are mostly present in fractions containing plasma
sure (gap area) was monitored. All PRPs enhanced cell prolifer- (Table S1). In addition, the angiogenic factors VEGF-D and RANTES
ation compared to controls, however, their regenerative capacity are mainly present in PRP and PRP Plasma but not in PFP-L or PPP,
was markedly different amongst PRPs (Fig. 2D and Fig. S1B). PRPs suggesting basal release into plasma by platelets (Fig. 3A-B). Strik-
1, 5 and 6 seemed to exert a lower regenerative capacity compared ingly, PFP-L is enriched in GFs and other bioactive molecules in-
to PRPs 2, 7, 10 and 12. Furthermore, the regenerative capacity of volved in tissue regeneration (Fig. 3A-B), compared to the original
PRPs in the absence of FBS, i.e., as serum replacement (Fig. 2D), PRP and other PRP-subfractions. However, the higher concentration
maintained the same tendencies, although not so markedly, except measured in PFP-L samples (prepared with the same number of
for PRP 1, who exerted the lowest regenerative response. platelets), points toward additional reasons for the lower concen-
This supports the notion that the molecular composition of tration measured in PRP samples. Particularly, the concentration of
PRPs conditions tissue regeneration, thus partly explaining the va- EGF was 7 times higher in PFP-L samples compared to PRP (683.8
riety in the clinical outcome of patients treated with these prod- pg/mL vs. 87 pg/mL respectively, see Fig. 3B and Table S1). Con-
ucts. In an attempt to identify molecules that could be driving the sidering that PRP contains platelets in 100% plasma and PFP-L is
difference in regenerative response, we first looked into the fac- free of plasma (or may contain only traces), we propose that po-
tors of the PCA Component 2 (Dim2; Fig 2C), that separates PRP 1 tential plasma-protein sequestration of bioactive molecules might
and PRP 5 from the rest, which were MIP1-β , TGF-β and VCAM-1 explain the lower levels detected in PRP, thus interfering with the
(Fig. 2E). These PRPs showed additionally higher levels of factors detection in the Multiplex assay [35].
(i.e., inflammatory) in the larger factor cluster (see factor dendro- Next, we performed wound healing assays using the different
gram of the heatmap, Fig. 2A), and were the ones that had less PRP subfractions as supplement. In assays performed in the pres-
regenerative potential, although with differences amongst cultures ence of FBS, where we can assess the additive contribution of the
when supplemented with FBS or not (Fig. 2D and Fig. S1B). products tested to the serum-supplemented cultures, the regener-
Of note, these wound healing assays were performed in the ative potential of PRP was similar to that of the PRP Plasma frac-
presence of heparin (2 U/mL) to prevent the unavoidable clotting tion, and in both cases slightly higher than the potential shown by
of plasma factors, however, we observed that the addition of hep- PPP, which is the poorest fraction in factor concentration (Fig. 3C-
arin alone in control cultures was deleterious (Fig. S1C). In order to D). These results are in concordance with the molecular profile of
circumvent this limitation, we grew cultures with PRP in the ab- the tested fractions. Of note, this difference is more acute in the
sence of heparin, and observed that the culture medium jellified, absence of FBS, i.e., when the different fractions are tested as re-
creating a layer of a translucent fibrin jelly that still allowed imag- placement of the xenogeneic serum. As shown in Fig. 3D, the frac-
ing with good resolution (Fig. S1D). For this reason, we decided to tions or products that exert the more favoring regeneration in vitro,
continue without adding heparin to the cultures in further experi- are those that contain platelet-derived factors, while PPP supports
ments. cell-growth, however, below the growth supported by FBS alone.
At the functional level, the PFP-L achieved complete wound clo-
3.2. Regenerative capacity of PRP subfractions sure within 48 hours, showing the highest regeneration potential
and superiority compared to PRP (Fig. 3C-D) in the presence of
It has been suggested that the plasma fraction of PRP-like prod- FBS. However, under starvation conditions (i.e., without FBS) the
ucts (i.e., plasma, PPP) accounts for most of the regenerative capac- regenerative potential of PFP-L was lower than that shown by the
ity of the product, rather than the platelet-derived factors them- plasma-containing subproducts, although still rescuing the absence
selves [34]. Thus, our next goal was to compare the different sub- of FBS (Fig. 3D). We cannot assure whether this is due to the par-
fractions obtained with our standardized closed-system PRP pro- ticular composition of the lysates, or to the absence of plasma
duction method by characterizing them molecularly and by assess- in the bioproduct, or serum in the culture conditions. Plasma (or
ing their regenerative capacity in vitro. For this comparison, we serum) may provide with components that may contribute in syn-
included the original PRP and PPP. Next, an aliquot of PRP was ergy with this bioproduct, as extrapolated from the cultures with
further processed by centrifugation. The pelleted platelets were FBS, where it induces gap closure more efficiently.
used to obtain plasma-free platelet lysate (PFP-L), and the super-
natant, tentatively named PRP Plasma to distinguish it from PPP, was 3.3. Characterization of platelet secretomes (PLT-S): the platelet
also included in the comparison (Fig. 1B). In Fig. 3A, we show signaling pathway activation drives their molecular composition and
the hierarchical clustering of PRPs and derived subfractions based regenerative capacity
on the concentration of measured molecules. A first observation
from this analysis is that there are evident differences between The obtained results suggested that the fractions containing
PPP and PRP Plasma subfractions, which appears counterintuitive. platelet-derived factors, i.e., PRP, PRP Plasma and PFP-L, displayed
While it seems logical that the concentration of molecules should the highest regenerative capacity in vitro. Therefore, we next aimed

138
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Fig. 3. Molecular and functional analysis of PRP subfractions. A. Heatmap depicting the hierarchical clustering of the different PRP-subfractions based on their molecular
composition as measured by Multiplex Technology, after scale-to-interval normalization. The dendrogram on the top of the heatmap shows clustering of the samples based
on their differential composition. B. Comparison of growth factor (GF) enrichment in PRP and PFP-L bioproducts. The graphs represent the cumulative concentration (pg/mL)
of relevant groups of bioactive factors (based on molecular function) as measured in each platelet-based bioproduct. The average concentration of each factor is represented,
which is calculated from the concentrations measured in the subproducts (PRP and PFP-L) prepared from two independent donors (n=2). C. Representative images of the
wound healing assay and scratch closure monitoring over time in the presence of FBS. On the top of the panel, control culture images are shown at the beginning (T0) and
at the end (48 hours -hr-) of the assay. The scratch position is marked between dashed lines, and can be better seen on the T0 control image. Images of bioproduct-treated
cultures are shown below at the end of the assay (48 hr). D. Line graphs showing the % of Scratch Covered Area (SCA) of wound healing assays at 12, 24, 36 and 48 hours of
control and bioproduct-stimulated cultures. Experiments were carried out in standard culture conditions (supplemented with 10% FBS, left graph) or in the absence of FBS
(right graph). Data are presented as the mean + standard error of the mean (SEM) of cultures treated with PRP subfractions obtained from different donors (n=4), with the
exception of control cultures and cultures treated with PFP-L (- FBS), where n=3. ∗ p<0.05; ∗ ∗ p<0.01; ∗ ∗ ∗ p<0.001.

139
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

at producing a platelet-derived bioproduct, free of plasma and product, which in turn associated to the agonist used to activate
undesirable platelet components, made of an enriched platelet- the platelet cargo degranulation (Fig. 4C and Fig. S3E). As we ex-
granule secretome generated by activating platelets to induce their pected, based on the quantification of bioactive factors by Multi-
degranulation (PLT-S). As a quality control of appropriate cargo de- plex Technology, the secretome obtained through the stimulation
livery in our PLT-S, we evaluated platelet degranulation by flow cy- of GPVI by CVX resulted in the most mitogenic response, reaching
tometry. For this purpose, surface expression of CD62P and CD63 a 96% of wound closure compared to the 66% of the control. Sim-
was assessed upon platelet stimulation, as an indirect measure of ilar results were obtained with collagen. Likewise, the secretome
granule release (alpha granules -CD62P-, and dense granules and obtained with PMA showed a high regenerative potential, with a
lysosomes -CD63-); (Fig. S2B). PLT-S samples were selected for fur- wound closure dynamic similar to that of the PLT-S obtained with
ther molecular characterization by Multiplex Technology and func- AggA. Strikingly, the secretome obtained with TRAP6 resulted in
tional studies when unstimulated platelets did not show any signs the lowest regenerative potential (maximal scratch covered area
of pre-activation, but displayed normal degranulation upon stimu- 85%). Overall, our results highlight the fact that the regenerative
lation. capacity of PLT-Ss differs depending on the agonist used to induce
PLT-S samples were prepared in accordance with our produc- platelet degranulation, because that determines its composition. Fi-
tion method, using platelets from whole blood (WB) as start- nally, and in concordance with the poorer composition, the super-
ing material and stimulating them with several receptor agonists. natant obtained after platelet stimulation with ristocetin showed a
Specifically, we stimulated GPVI with convulxin (CVX) and high- wound closure dynamic similar to control cells (Fig. S3E-F).
dose collagen (Coll-High), integrin α 2β 1 with low-dose collagen
(Coll-Low), PAR1 with TRAP6, CLEC2 with aggretin A (AggA), in-
3.4. The platelet source used to prepare PLT-S conditions its
tegrin α IIbβ 3 with PMA, and the von Willebrand factor receptor
composition and in vitro regenerative capacity
(vWF-R; GPIB-V-IX) with ristocetin; we also included unstimulated
platelets as control (Fig. S2B-C). Subsequently, we studied the com-
We next studied whether the source of platelets used to pre-
position of the PLT-Ss obtained in response to different stimuli. The
pare PLT-Ss would condition the quality and regenerative capac-
molecular characterization of samples showed that FGF-2 and IL-
ity of PLT-Ss themselves. With this objective, we prepared PLT-
8 were either not present, or below the detection limit (see Ta-
Ss from two different sources, whole blood (PLT-SWB ), or closed-
ble S1), hence these factors were filtered out for further analysis.
system PRPs before freezing (PLT-SPRP ); and using CVX, PMA and
Of note, although we observed a clear inter-donor variability (Fig.
TRAP6 as stimuli. Molecular characterization of the bioproducts
S3A), the data also shows a clustering that depends on the platelet
by Multiplex Technology was performed. As previously observed,
response or status: active aggregation/degranulation or agglutina-
FGF-2 and IL-8 were not detected in PLT-S samples (see Table S1),
tion/resting platelets, with the exception of one sample stimulated
hence these factors were filtered out for further analysis. As shown
with ristocetin that clustered with the aggregation/degranulation
in Fig. 5A, the hierarchical clustering of samples based on their
samples (Fig. S3B). Once the inter-donor variability was removed
molecular composition shows that the platelet source is a vari-
from the data, the multiplex quantification results showed a differ-
able that significantly influences the composition of the final bio-
ent degranulation profile depending on the platelet agonist used,
product. Specifically, the PLT-SWB showed a higher concentration of
which is visualized by hierarchical clustering in the heatmap and
most GFs and bioactive molecules, which translates into a greater
PCA plots (Fig. 4A-B). It should be noted that the PCA generated
regenerative potential (in the presence of FBS) (Fig. 5B). This sug-
with the average of the raw data (pg/mL) resulted in the same
gested that the PRP manufacture procedure caused basal platelet
sample clustering, which confers reliability to the batch effect re-
degranulation that occurred either during the 24 hour storage from
moval analysis (Fig. S3C). Specifically, our results showed that the
collection to processing, or during the production itself. Since we
stimulation with CVX and collagen (high and low doses) generated
discarded the soluble fraction to prepare PLT-SPRP , most probably
a similar secretion profile, result that is consistent with the cross-
we lost the released factors in our final product. Corroborating our
talk between the signal transduction pathways activated by those
previous results, the factors were represented in the PLT-Ss in dif-
agonists. Furthermore, the stimulation with TRAP-6 and AggA gen-
ferent proportions, not only due to the influence of the platelet
erated a secretome with similar composition pattern, characterized
source, but also due to the signaling pathway activation (i.e., the
by an enrichment in the adhesion proteins ICAM-1/VCAM-1. Fi-
stimulus used to produce the secretome, Fig. 5C).
nally, the stimulation with PMA generated a unique secretion pro-
file characterized by lower levels in some growth factors (i.e., EGF,
VEGF-D and PDGF-BB, among others). In contrast, when platelets 3.5. In vitro stability and consumption of factors from PLT-S
were stimulated with ristocetin, which activates the vWF-R com- bioproducts
plex, the concentration of most of the bioactive factors drastically
decreased to levels comparable to those measured in unstimulated In an attempt to determine the stability and consumption of
platelets (Table S1 and Fig. S3D). The response to ristocetin was not factors from PLT-Ss in culture, we tested different PLT-Ss as de-
unexpected, as activation of vWF-R by this agonist does not trigger scribed in the Materials and Methods section, and we selected a
a secretion response per se, but rather it induces platelet aggluti- panel of GFs to monitor using Multiplex Technology. As shown
nation [36]. As evidenced in Fig. S2B, the stimulation with risto- in Fig. S4A, MRC-5 cells themselves produced some of the factors
cetin resulted in the creation of an agglutinate-mesh where, as we contained in our panel, and thus, present in PLT-Ss, i.e., HGF, MCP-
hypothesized, any released bioactive factors would be sequestered. 1, SDF-1α and VEGF-A, which may preclude the analysis of factors
Thus, the physical characteristics of aggregate formation might also included in our panel. As expected, the concentrations measured in
influence factor release. supernatants were increased in cultures with a higher number of
We next evaluated the in vitro regenerative potential of the PLT- cells. Next, we set out to evaluate the stability of factors in cultures
Ss and whether the differences observed in composition would devoid of cells. We observed high variability in the dynamics of the
promote associated grades of regeneration. We performed the as- concentration of factors that we were able to measure in cultured
says in the presence of FBS at first, because we intended to see dif- medium supplemented with the tested PLT-Ss after 48 hours, i.e.,
ferences in their potentiation capacity rather than evaluating their EGF, PDGF-BB and VEGF-A (the dilution factor still allowed their
serum replacement capacity. Our results show that the wound clo- detection in supernatants). From the results obtained, it appeared
sure dynamic was highly dependent on the composition of the bio- that the stability or decline of a single factor is not associated with

140
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Fig. 4. Composition and functional analysis of platelet secretomes from whole blood (PLT-SWB ). A. The heatmap depicts the hierarchical clustering of PLT-SWB samples based
on their molecular composition as measured by Multiplex Technology, after batch-effect removal and scale-to-interval normalization. The dendrogram on the top of the
heatmap shows clustering of the samples based on their differential composition. Samples from each donor (1 to 4) are indicated. The agonist used to produce secretomes
in each case, is also indicated: A, Aggretin A; L, Collagen Low concentration; H, Collagen High concentration; X, Convulxin; T, TRAP6; P, PMA. B. Principal component analysis
(PCA) of PLT-SWB samples after batch-effect-removal and scale-to-interval normalization. Component 2 (Dim 2) separates most secretomes obtained with Collagen High
and Low concentration (Coll-High and Coll-Low) and Convulxin (top quadrant), from those obtained with TRAP6, Aggretin A or PMA (bottom quadrant). C. Representative
microscopic images of control wound healing assays at the beginning (T0) and at the end (48 hr) of cultures, in the presence of FBS. The scratch position is marked between
dashed lines and can be better visualized on the T0 control image. Representative images of cultures treated with PLT-Ss obtained with different agonists are shown at the
end of the assay (48 hr). The line graphs show the % of Scratch Covered Area (SCA) of wound healing assays at 12, 24, 36 and 48 hours of control and PLT-S treated groups.
Data are presented as the mean + standard deviation (SD) of experimental replicates (n=3). ∗ p<0.05; ∗ ∗ p<0.01; ∗ ∗ ∗ p<0.001.

141
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Fig. 5. The platelet source is a key driver in the composition of PLT-Ss. A. Heatmap depicting the hierarchical clustering of PLT-SWB and PLT-SPRP samples based on their
molecular composition as measured by Multiplex Technology, after scale-to-interval normalization. Samples from each donor (1 to 4 and 15 to 18) are indicated. The agonist
used to produce secretomes in each case, is also indicated: X, Convulxin; T, TRAP6; P, PMA. B. Line graphs showing the % of Scratch Covered Area (SCA) of wound healing
assays at 12, 24, 36 and 48 hours cultured with PLT-SWB or PLT-SPRP -obtained using CVX as agonist- (in the presence of FBS). The upper graph shows the SCA from cultures
supplemented with PLT-SWB or PLT-SPRP and respective controls, and the lower graph shows the normalized SCAs after setting respective controls to 0 at each time point.
Data are presented as the mean + standard deviation (SD) of control cultures and cultures treated with bioproducts obtained from different donors (n=3). ∗ p<0.05; ∗ ∗
p<0.01. C. Lollipop plots illustrating the composition variation (Log2FC WB/PRP ratio) of PLT-Ss depending on the platelet source and conditioned by the platelet-receptor
agonist used for stimulation.

142
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

its initial concentration, which suggests that the global composi- each subproduct, in synergy with FBS, was completely in line with
tion of the bioproduct conditions the stability of single factors (Fig. the level of GFs measured in each of them (Fig. 6C and S5D).
S4B). Finally, to evaluate factor consumption we set cultures with Specifically, PLT-SWB and PFP-L outpassed the PRP-related bioprod-
a low (as in our wound healing assays) or high number of MRC- ucts. In concordance with the cumulative concentration of mea-
5 cells and supplemented them with the tested PLT-Ss. The results sured GFs, cytokines and chemokines, the regenerative capacity of
obtained show that factors were not necessarily exhausted after 48 Custom PRP and PLT-SPRP was inferior compared to PRP, PFP-L and

hours in the cultures with a lower cell number (i.e., wound heal- PLT-SWB . Surprisingly, we observed that Custom PFP-L exerted an in-
ing assay condition). When they were exhausted, it was not related hibitory effect on cell proliferation compared to the control assays.
undoubtedly to a lower initial concentration of the given factor. Of note, in this particular bioproduct, the concentration of most
In cultures with a higher cell number, the reduction in concen- factors was lower than in any other subproduct, while the concen-
tration of factors was noticeable, but it not always led to exhaus- tration of Caspase-3 remained at the levels of the ones measured
tion. Finally, in the case of VEGF-A, and not always, the addition in related PFP-L fractions (Fig. S5C).
of PLT-S might induce the production of the factor itself by MRC-5 Next, the regenerative capacity was assessed in the absence
cells, thus making it impossible to discern the remaining VEGF- of FBS, with those subproducts that showed similar or superior
A from the PLT-S supplement or the VEGF-A newly produced by regeneration potential to PRP in the previous analysis. We com-
MRC-5 cells (Fig. S4C). Overall, this preliminary data suggests that pared PRP, PFP-L, PLT-SWB and Custom PRP (Fig. 6D and Fig. S5E).
the global composition of the bioproduct may condition the stabil- Our first observation was that all bio-products were able to sup-
ity and consumption of single factors, which will also be cell-type- port cell proliferation in the absence of FBS, proving their poten-
or tissue-dependent (Fig. S4). tial use as xenogeneic serum replacement (Fig. S5E). However, the
proliferation curve showed different kinetics amongst subproducts.
3.6. Regenerative capacity and molecular composition across the Importantly, and additional to Fig. 3D, where it was shown that
different platelet-based subproducts PFP-L regenerative capacity was outpassed by PRP subproducts,
we observed in this comparative analysis that the regenerative ca-
Next, we compared PLT-Ss with our gold standard reference, pacity correlated with platelet count on customized products, i.e.
Custom PRP displayed a slightly lower regenerative capacity than PRP
closed-system PRP, as well as the other relevant PRP subfractions,
to better assess the regenerative capacity of platelet-based bio- (Fig. S5E). Interestingly, the potential to stimulate cell proliferation
products in a comprehensive and comparative manner. To this and/or migration shown by PLT-SWB was higher than that shown
end, we prepared additional formulations in which the number by the rest of PRP-based bioproducts (Fig. 6D and Fig. S5E). The su-
of platelets was even between the different bioproducts. We en- periority of PLT-S (specially the one obtained from WB) compared
gineered, from the same donor source, PRP and PFP-L contain- to not only PRP, but also to PFP-L, in terms of regenerative capacity,
ing the same concentration of platelets as we used to prepare is remarkable, as PFP-L is prepared with an initial higher number
PLT-S (Custom PRP and Custom PFP-L, respectively) and, on the other of platelets.
hand, we prepared PLT-S containing the same concentration of
platelets in PRP, which is on average 3X higher (namely Supra PLT- 4. Discussion
SPRP ; see Fig. 1C-D). The comparison of the molecular profile of
bioproducts showed that PRP samples (PRP and Custom PRP) and The use and demand of platelet-based bioproducts in regen-
PFP-L samples (PFP-L and Custom PFP-L) clustered together respec- erative medicine has increased in recent years. However, the use
tively, although the factor concentration was lower in the respec- of PRP remains highly controversial due to a lack of standard-
tive custom products, i.e., prepared with an original lower number ization of preparation and application methods, which results in
of platelets (Fig. 6A). As we observed before, the PLT-SWB showed highly variable PRP products (in terms of purity, content, quality
the most enriched composition in GFs, and if we consider the cu- and safety), which severely impact their potential clinical efficacy
mulative concentration of the pool of GFs analyzed, this variable [24,26]. Furthermore, as of today, it is very difficult to establish the
would be similar to that obtained in Supra PLT-SPRP and PFP-L, where real clinical benefits of these therapies, as the lack of characteriza-
the number of initial platelets is approximately 3X higher than in tion of the bioproducts and a detailed description of the produc-
PLT-SWB (Fig. 6B). However, it is important to acknowledge that tion methods is a constant in the literature, precluding cross-study
the proportion of the individual GFs in each sample was different. comparisons. To avoid that skepticism towards platelet-based bio-
While PLT-SWB showed a higher concentration of PDGF-BB, G-CSF products grows along with their increasingly popular use in regen-
and TGF-β , Supra PLT-SPRP displayed a higher concentration of VEGF- erative medicine, there is an urgent need for a better characteri-
A. Instead, PFP-L contained a higher concentration of TPO and EGF zation of the products used in each study and the need of stan-
(Fig. 6B and Table S1). Of note, all subfractions were prepared from dardized production methods. In this study, we characterize the
the same donors, except the PLT-SWB , so we cannot rule out donor- in vitro regenerative capacity of platelet-based bioproducts and re-
dependent variation contributing to the composition differences. lated subfractions, and associate it with their molecular composi-
Comparison between bioproducts prepared with the same initial tion profile.
platelet concentration, corroborated that cytokines and chemokines As a starting point, we first characterized PRP produced in a
were also detected in greater proportion in secretome samples (Fig. closed system with a standardized procedure, which relays on the
S5A-B). It is interesting to highlight that PLT-SWB contained a lower equipment of a certified blood bank (closed-system PRP, WB PRPFT2
proportion of the proinflammatory cytokine IL-6 and increased lev- according to the nomenclature proposed by us [24,30]). Our re-
els of GITRL and SDF-1α , key factors involved in cell migration and sults support the notion of inter-donor variation at the molecu-
proliferation [37,38]. lar and functional level, while they also highlight the fact that a
Based on this molecular characterization, we hypothesized that standardized method to produce PRP assures a certain quality of
this platelet-based bio-product, PLT-S, could have a higher regen- the product, minimizing variation due to other variables, such as
erative potential compared to PRP, as it is exclusively enriched in the platelet count in the original platelet-based bioproduct. In this
platelet granule molecules. To evaluate this hypothesis, we set out sense, some studies have suggested an impact of age or gender
to compare the functional profile of the different platelet-based on PRP GF levels. Evanson et al. reported that the concentration of
bioproducts in in vitro wound healing assays. Our results showed certain GFs was higher in women compared to men (at the same
that the wound closure dynamic in cultures supplemented with platelet ratio of PRP vs whole blood for both genders) as well as

143
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

Fig. 6. Molecular profile and regenerative capacity across different platelet-based subproducts. A. Heatmap depicting the hierarchical clustering after scale-to-interval nor-
malization of the different subproducts based on their molecular composition as measured by Multiplex Technology: PRP-based (PRP and Custom PRP), PFP-L formulations
(PFP-L and Custom PFP-L) and PLT-Ss (PLT-SWB , PLT-SPRP and Supra PLT-SPRP ). The dendrogram on the top of the heatmap shows clustering of the samples (mean values) based on
their differential composition. The agonist used to produce secretomes in each case, is indicated: A, Aggretin A; H, Collagen High concentration; L, Collagen Low concentra-
tion; X, Convulxin; T, TRAP6; P, PMA. B. The bar graph represents the cumulative concentration (pg/mL) of GFs detected by Luminex in the different platelet bioproducts.
The average concentration of each factor is represented, which is calculated from the concentrations measured in the subproducts prepared from four independent donors
(n=4), with the exception of PFP-L and Custom PFP-L (n=2). C. Line graphs shows the % of Scratch Covered Area (SCA) of wound healing assays at 12, 24, 36 and 48 hours of
control and treated groups in addition to 10% FBS, as standard culture conditions. The graph on the top compares the SCA of cultures supplemented with PLT-Ss -obtained
from whole blood (WB) or PRP sources using CVX as agonist- with those supplemented with Custom PRP or Custom PFP-L (i.e., obtained from the same number of platelets as the
PLT-Ss) and respective controls. The graph on the bottom shows the results from the same assays, after normalization to respective controls, including cultures supplemented
with regular PRP and PFP-L. Data are presented as the mean + standard error of the mean (SEM) of biological replicates (n=4, except for PLT-Ss where n=3). ∗ p<0.05; ∗ ∗
p<0.01; ∗ ∗ ∗ p<0.001. D. Representative microscopic images of wound healing assays in MRC-5 fibroblast cell cultures at the beginning and after 24 and 48 hours, and in the
absence of FBS supplementation. Untreated cells were taken as control. The line graphs show the % of SCA of wound healing assays at 12, 24, 36 and 48 hours of control
and treated groups. Data are presented as the mean + standard error of the mean (SEM) of biological replicates (n=4, except for PLT-SWB where n=3). Statistical analysis
was applied to compare PLT-SWB with Custom PRP; ∗ p<0.05; ∗ ∗ p<0.01.

144
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

in younger people (≤ 25 years old) [39]. Additionally, other stud- quality and reactivity, and the use of autologous products might
ies reported a negative correlation between age and PDGF-BB and not always be the most suitable therapeutic source [27,28,55]. Con-
IGF-1 levels [40,41]. A limitation of our study is that we could not sequently, there is an increased clinical interest in the development
possibly assess the impact of age and gender, as we used inter- of platelet-based bioproducts that could be used universally (allo-
rupted donations or discard material from the blood bank, without geneic), and would be more specific or targeted to the pathology
inclusion requirements. Another aspect to take into consideration being treated, in an effort to improve and personalize the therapy.
is that our study focused on the analysis of 37 growth factors and Interestingly, the dissection of PRP subfractions allowed us to
cytokines, which precludes data on other components (other pro- identify the fractions that actively contribute to tissue regenera-
teins, RNA, miRNA and metabolites) that may influence the regen- tion, i.e., those that contain undoubtedly platelet-derived factors.
erative capacity of each product [42–45]. Therefore, we developed a method to generate plasma-free se-
Our next objective was to assess the regenerative capacity cretomes (PLT-S; PCT/IB2022/057936). Contrarily to methods de-
of the different subfractions obtained from PRPs produced in a scribed by other authors, we did not use apyrase or prostaglandin
closed-system. With that aim, fresh-PRP samples were dissected E2 (PGE2) to minimize platelet activation during the manipula-
into their components, plasma and platelets, to identify the contri- tion steps (pipetting and centrifugation) [14]. We consider that
bution of each fraction to the wound healing process. Importantly, the use of these compounds may result in slightly anergic platelet
our experimental design allowed the comparison of different frac- responses upon stimulation. From our point of view, optimized
tions obtained from the same donor. Our study showed that PPP washing protocols with minimum steps and the use of an appro-
worked itself as a potent stimulator of cell proliferation. This ob- priate buffer (HEPES-Glucose or HEPES-Tyrode’s buffer) prevents
servation is in line with previous studies showing the effective- the pre-activation and destruction of platelets, as well as con-
ness of PPP in the healing process [46–48]. Indeed, although PPP tamination by erythrocytes and leukocytes [24,56]. Another step
by definition contains a very low concentration of platelets and, avoided in our protocol, and a matter of concern from our perspec-
therefore, smaller quantities of GFs, it still represents a reservoir tive, is the interruption of the platelet stimulation with ice [21,57].
of bioactive molecules (such as PDGF-BB, VEGF-A, TGF-β ), which It is widely known that cold temperatures promote platelet activa-
may be responsible for the observed effect [49]. In addition, the tion and aggregation [58,59]. Thus, from our point of view, placing
plasma compartment contains high levels of fibrinogen; interest- reactions on ice would introduce an additional stimulation step,
ingly, the fibrin network that it forms upon thrombin stimulation, which could affect the composition and the subsequent regener-
has been postulated as a controlled release system beneficial for ative capacity of the bioproduct. Furthermore, we would not be
correct wound healing [50,51]. able to separate the ice-activating interference and its contribution
Of relevance, our results also explain the observations described to the agonist-induced degranulation. Our analysis revealed a dif-
in other studies that claim that the plasma fraction contributes to ferent platelet secretion profile and functional response of PLT-Ss
the major regenerative potential capacity of these products. In fact, in an agonist-dependent manner. This result is consistent with the
basal degranulation of platelets due to processing, and release of observations made by other authors [21].
these factors to the so-called “plasma” fraction, might be respon- Another key aspect in the preparation of platelet-based bio-
sible for the described contribution of “plasma” to the regenera- products is the choice of the platelet source. As a consequence
tive process on those studies. Our experimental approach revealed of a spontaneous release, as we hypothesize, platelets from fresh
that the additional sample processing to separate these fractions closed-system PRP have a lower degranulation capacity in response
resulted in a PRP-like bioproduct (PRP Plasma in our experimental to agonists. Thus, PLT-Ss prepared from PRP samples have lower
approach), containing additional platelet-derived cargo molecules concentration of factors measured, as we showed, compared to
and displaying higher regenerative capacity compared to PPP, and PLT-Ss produced directly from whole blood. This observation is of
similar to that of the reference PRP. This result is of special rele- relevance, since PRP and other platelet derived products are be-
vance, given that there is not sufficient research studying the rel- ing considered to be preparedfrom platelet concentrates that are
ative contribution of both plasma and platelets to the regenera- expired for transfusion. While this option would assure a circular
tive potential of PRP, and many studies obtain plasma from platelet economy of the process, it should be acknowledged that the obten-
concentrates, downplaying the role of platelets in the observed ef- tion method should be adapted specifically to the platelet source,
fects [34,52]. The blood plasma fraction shall always contain solu- in order to obtain a product of certain quality, i.e., pooling concen-
ble platelet-derived factors, naturally, and it should be considered trates or donations for large-scale production of off-the-shelf prod-
that the processing of samples to separate platelets from plasma ucts with certified quality. This also applies to the variation in sta-
might induce additional platelet activation and degranulation into bility and consumption of factors as observed in vitro, which ap-
the plasma fraction. It is thus very difficult to separate cleanly pear to be dependent on the bioproduct composition, and hence,
a plasma fraction free of platelet-secreted molecules, and caution dependent on inter-donor variation.
should be taken when interpreting results from these studies. Lastly, we compared the composition profile and functionality
This dissection also showed that the platelet lysate fraction, of PRP, PFP-L and PLT-S subproducts. In general, GF levels were
PFP-L, induced a lower regenerative capacity, and even appeared higher (approximately 3-fold) in PLT-SWB samples compared to
to be deleterious to the cultures. Of note, we identified in this par- Custom PRP (prepared with the same number of platelets). We hy-

ticular bioproduct a higher concentration of Caspase-3, which has pothesized that the low levels of GFs and other immune media-
been recently shown to exert non-apoptotic functions related to tors detected in PRP might be due to the presence of the plasma
the proliferation and modulation of cell growth [53]. In addition, fraction. Several studies report that the association of GFs with
it has been previously described that an enhanced expression of specific plasma-binding proteins, blocks the interaction between
Caspase-3 in hypertrophic scars and keloid induces apoptosis of fi- the GF and its cell-surface receptor and modulates their functional
broblasts [54]. Taken together, and in agreement with other stud- activity [35,60]. Interestingly, we observed a superiority of PLT-S
ies, our data support a personalized approach to PRP therapy and compared to PRP in vitro, with some remarks. Thus, the depletion
underscore the need for a greater understanding of the relation- of plasma in PLT-S constitutes a great advantage compared to PRP
ships between PRP composition and clinical outcomes. in terms of bioavailability and functionality of GFs. Nevertheless, it
The autologous use of PRP bioproducts, as required by local reg- seems pertinent to highlight that, in the PRP, the growth factors
ulatory establishments, may also impede its prescription in certain sequestered by plasma proteins might be gradually released and
patients. Certain pathological conditions might influence platelet protected against degradation for a longer time, allowing longer

145
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

storage. In this sense, we envision that the encapsulation of PLT- Acknowledgements


S in nanoparticles made of different biomaterials (i.e., liposomes,
synthetic platelets, etc.) would improve their absorption, stability The authors thank all the institutions and personnel involved
and functionality, while allowing to treat non-accessible tissues in this study, with special emphasis to the Centro Comunitario de
or locations, in a targeted and personalized manner. Other post- Sangre y Tejidos de Asturias (Oviedo, Spain) and Immunology Ser-
production procedures (i.e., lyophilization), would allow the pro- vice, Hospital Universitario Central de Asturias (Oviedo, Spain), for
duction of PLT-S that could be stored at the clinical practice in their assistance with the Luminex® Analyzer.
order to be available for immediate use. Alloimmunization is an-
other important issue, which is reduced to almost zero risk when Supplementary materials
using plasma-free bioproducts, and this characteristic would allow
the allogeneic use of PLT-S. Supplementary material associated with this article can be
Overall, our findings support the idea of using PLT-S for thera- found, in the online version, at doi:10.1016/j.actbio.2024.01.029.
peutic applications as an alternative to the current autologous ap- References
plication of PRP. Given its characteristics, PLT-S represents a plau-
sive option to explore novel applications in regenerative medicine [1] P.E.J. van der Meijden, J.W.M. Heemskerk, Platelet biology and functions: new
and other clinical fields: it would allow allogeneic use and the pro- concepts and clinical perspectives, Nat. Rev. Cardiol. 16 (3) (2019) 166–179.
[2] M. Leslie, Cell biology. beyond clotting: the powers of platelets, Science 328
duction method could be optimized to minimize batch-to-batch (5978) (2010) 562–564.
variation, or to incorporate post-processing steps to allow nanoen- [3] K. Suzuki-Inoue, N. Tsukiji, T. Shirai, M. Osada, O. Inoue, Y. Ozaki, Platelet
capsulation (targeted delivery) or storage with off-the-shelf pur- CLEC-2: roles beyond hemostasis, Semin. Thromb. Hemost. 44 (2) (2018)
126–134.
poses. Future studies in more complex cellular and tissue settings [4] J.W. Semple, J.E. Italiano Jr., J. Freedman, Platelets and the immune continuum,
(3D reconstructions, organoids) and/or in vivo models are needed Nat. Rev. Immunol. 11 (4) (2011) 264–274.
to fully understand the regenerative capacity of this platelet- [5] L.J. Gay, B. Felding-Habermann, Contribution of platelets to tumour metastasis,
Nat. Rev. Cancer 11 (2) (2011) 123–134.
based bioproduct, and the specific production and post-production
[6] C.N. Jenne, R. Urrutia, P. Kubes, Platelets: bridging hemostasis, inflammation,
methodology adaptations for long-term storage and each singular and immunity, Int. J. Lab. Hematol. 35 (3) (2013) 254–261.
clinical application. [7] L.F. Brass, H. Jiang, J. Wu, T.J. Stalker, L. Zhu, Contact-dependent signaling
events that promote thrombus formation, Blood Cells Mol. Dis. 36 (2) (2006)
157–161.
[8] S.P. Jackson, W.S. Nesbitt, S. Kulkarni, Signaling events underlying thrombus
Author contributions
formation, J. Thromb. Haemost. 1 (7) (2003) 1602–1612.
[9] E.M. Battinelli, B.A. Markens, J.E. Italiano Jr., Release of angiogenesis regulatory
Conceptualization, L.G. and A.A.-H; investigation and method- proteins from platelet alpha granules: modulation of physiologic and patho-
ology, A.A.-H. and L.G.; resources and reagents, J.F.-F., A.M.O.-P., logic angiogenesis, Blood 118 (5) (2011) 1359–1369.
[10] J.A. Coppinger, P.B. Maguire, Insights into the platelet releasate, Curr. Pharm.
M.C.M.-T., A.L.-V. and J.A.E.; performance of experiments, A.A.H: Des. 13 (26) (2007) 2640–2646.
and G.C.-A.; formal analysis, A.A.-H., P.M.-B. and L.G.; writing— [11] A. Sharda, R. Flaumenhaft, The life cycle of platelet granules, F10 0 0Res. 7
original draft preparation, A.A.-H. and L.G.; writing—review and (2018) 236.
[12] D.M. Maynard, H.F. Heijnen, M.K. Horne, J.G. White, W.A. Gahl, Proteomic anal-
editing, A.A.-H., P.M.-B and L.G.; supervision, L.G. All authors have ysis of platelet alpha-granules using mass spectrometry, J. Thromb. Haemost.
read and agreed to the published version of the manuscript. 5 (9) (2007) 1945–1955.
[13] D.M. Maynard, H.F. Heijnen, W.A. Gahl, M. Gunay-Aygun, The alpha-granule
proteome: novel proteins in normal and ghost granules in gray platelet syn-
Declaration of competing interest drome, J. Thromb. Haemost. 8 (8) (2010) 1786–1796.
[14] O. Pagel, E. Walter, K. Jurk, R.P. Zahedi, Taking the stock of granule cargo:
platelet releasate proteomics, Platelets 28 (2) (2017) 119–128.
The authors declare the following financial interests/personal [15] E.M. Battinelli, J.N. Thon, R. Okazaki, C.G. Peters, P. Vijey, A.R. Wilkie, L.J. Noet-
relationships which may be considered as potential competing in- zli, R. Flaumenhaft, J.E. Italiano, Megakaryocytes package contents into sepa-
rate alpha-granules that are differentially distributed in platelets, Blood Adv. 3
terests: A.A.-H. and L.G. are co-founders of the recently constituted
(20) (2019) 3092–3098.
spin off company, Platelet Biotechnologies S.L. (PlaBiTe). The rest of [16] P. Handagama, R.M. Scarborough, M.A. Shuman, D.F. Bainton, Endocytosis of
authors have no competing interest to disclose. fibrinogen into megakaryocyte and platelet alpha-granules is mediated by al-
pha IIb beta 3 (glycoprotein IIb-IIIa), Blood 82 (1) (1993) 135–138.
[17] P. Handagama, D.F. Bainton, Y. Jacques, M.T. Conn, R.A. Lazarus, M.A. Shuman,
Data availability Kistrin, an integrin antagonist, blocks endocytosis of fibrinogen into guinea
pig megakaryocyte and platelet alpha-granules, J. Clin. Invest. 91 (1) (1993)
193–200.
The raw data required to reproduce these findings are available [18] B.A. Bouchard, N.T. Meisler, M.E. Nesheim, C.X. Liu, D.K. Strickland, P.B. Tracy,
to download from Table S1. A unique function for LRP-1: a component of a two-receptor system mediating
specific endocytosis of plasma-derived factor V by megakaryocytes, J. Thromb.
Haemost. 6 (4) (2008) 638–644.
[19] G.L. Klement, T.T. Yip, F. Cassiola, L. Kikuchi, D. Cervi, V. Podust, J.E. Ital-
Funding iano, E. Wheatley, A. Abou-Slaybi, E. Bender, N. Almog, M.W. Kieran, J. Folk-
man, Platelets actively sequester angiogenesis regulators, Blood 113 (12) (2009)
This study was partially supported by an I+D Excelencia 2020 2835–2842.
[20] H. Heijnen, P. van der Sluijs, Platelet secretory behaviour: as diverse as the
project grant (PID2020- 117265GB-I00, Ministerio de Ciencia e In- granules ... or not? J. Thromb. Haemost. 13 (12) (2015) 2141–2151.
novación -Spain-) to L.G. A.A.-H. was supported by said grant. [21] D. Jonnalagadda, L.T. Izu, S.W. Whiteheart, Platelet secretion is kinetically
P.M.-B. is supported by a Severo Ochoa Grant (Consejería de Cien- heterogeneous in an agonist-responsive manner, Blood 120 (26) (2012)
5209–5216.
cia, Innovación y Universidad del Principado de Asturias, PA-20-
[22] R. Alves, R. Grimalt, A review of platelet-rich plasma: history, biology, mecha-
PF-BP19-014). J.A.E. is financially supported by the German Re- nism of action, and classification, Skin. Appendage Disord. 4 (1) (2018) 18–24.
search Foundation (DFG – Project-ID 194468054 – SFB 1009project [23] J. Alsousou, A. Ali, K. Willett, P. Harrison, The role of platelet-rich plasma in
tissue regeneration, Platelets 24 (3) (2013) 173–182.
A09) and by the Interdisciplinary Center of Clinical Research (IZKF
[24] A. Acebes-Huerta, T. Arias-Fernandez, A. Bernardo, M.C. Munoz-Turrillas, J. Fer-
grant: Ebl-A/009/21). Patent application fees (PCT/IB2022/057936 nandez-Fuertes, J. Seghatchian, L. Gutierrez, Platelet-derived bio-products:
and P202130806) were partially covered by Technology Trans- classification update, applications, concerns and new perspectives, Transfus.
fer grants (Consejería de Ciencia, Innovación y Universidad del Apher. Sci. (2019) 102716.
[25] P. Everts, K. Onishi, P. Jayaram, J.F. Lana, K. Mautner, Platelet-rich plasma: new
Principado de Asturias AYUD/2021/57101, AYUD/2022/26253 and performance understandings and therapeutic considerations in 2020, Int. J.
AYUD/2022/33562). Mol. Sci. 21 (20) (2020).

146
A. Acebes-Huerta, P. Martínez-Botía, G. Carbajo-Argüelles et al. Acta Biomaterialia 177 (2024) 132–147

[26] P. Harrison, P. Subcommittee on Platelet, The use of platelets in regenerative patients with type 2 diabetes mellitus influences circulating MicroRNA pat-
medicine and proposal for a new classification system: guidance from the SSC terns via inflammatory cytokines, Arterioscler. Thromb. Vasc. Biol. 35 (6)
of the ISTH, J. Thromb. Haemost. 16 (9) (2018) 1895–1900. (2015) 1480–1488.
[27] P.B. Szklanna, M.E. Parsons, K. Wynne, H. O’Connor, K. Egan, S. Allen, F. Ni [43] Y. Li, Y. Liang, Y. Gao, D. Chen, X. Ran, Dynamic changes of wound-related miR-
Ainle, P.B. Maguire, The platelet releasate is altered in human pregnancy, Pro- NAs after application of autologous platelet-rich gel in diabetic wounds, Chin.
teomics. Clin. Appl. 13 (3) (2019) e1800162. Med. J. 135 (21) (2022) 2644–2646.
[28] E. Anitua, R. Prado, G. Orive, Allogeneic platelet-rich plasma: at the dawn of [44] C. Del Amo, A. Perez-Valle, L. Atilano, I. Andia, Unraveling the signaling secre-
an off-the-shelf therapy? Trends Biotechnol. 35 (2) (2017) 91–93. tome of platelet-rich plasma: towards a better understanding of its therapeutic
[29] P.B. Maguire, M.E. Parsons, P.B. Szklanna, M. Zdanyte, P. Munzer, M. Chatterjee, potential in knee osteoarthritis, J. Clin. Med. 11 (3) (2022).
K. Wynne, D. Rath, S.P. Comer, M. Hayden, F. Ni Ainle, M. Gawaz, Comparative [45] M.E.M. Parsons, P.B. Szklanna, J.A. Guerrero, K. Wynne, F. Dervin, K. O’Connell,
platelet releasate proteomic profiling of acute coronary syndrome versus stable S. Allen, K. Egan, C. Bennett, C. McGuigan, C. Gheveart, F. Ni Ainle, P.B. Maguire,
coronary artery disease, Front. Cardiovasc. Med. 7 (2020) 101. Platelet releasate proteome profiling reveals a core set of proteins with low
[30] A.M. Ojea-Perez, A. Acebes-Huerta, T. Arias-Fernandez, L. Gutierrez, variance between healthy adults, Proteomics 18 (15) (2018) e1800219.
M.C. Munoz-Turrillas, Implementation of a closed platelet-rich-plasma prepa- [46] M. Caceres, C. Martinez, J. Martinez, P.C. Smith, Effects of platelet-rich and
ration method using the local blood bank infrastructure at the Principality of -poor plasma on the reparative response of gingival fibroblasts, Clin. Oral Im-
Asturias (Spain): back to basic methodology and a demographics perspective plants Res. 23 (9) (2012) 1104–1111.
after 1 year, Transfus. Apher. Sci. (2019). [47] M. Shahidi, M. Vatanmakanian, M.K. Arami, F. Sadeghi Shirazi, N. Esmaeili,
[31] A. Caiado, G. Ferreira-Dos-Santos, S. Goncalves, L. Horta, P. Soares Branco, S. Hydarporian, S. Jafari, A comparative study between platelet-rich plasma
Proposal of a new standardized freeze-thawing technical protocol for leuco- and platelet-poor plasma effects on angiogenesis, Med. Mol. Morphol. 51 (1)
cyte-poor platelet-rich plasma preparation and cryopreservation, Cureus 12 (7) (2018) 21–31.
(2020) e8997. [48] F. Chellini, A. Tani, S. Zecchi-Orlandini, C. Sassoli, Influence of platelet-rich
[32] M.E. Ritchie, B. Phipson, D. Wu, Y. Hu, C.W. Law, W. Shi, G.K. Smyth, limma and platelet-poor plasma on endogenous mechanisms of skeletal muscle re-
powers differential expression analyses for RNA-sequencing and microarray pair/regeneration, Int. J. Mol. Sci. 20 (3) (2019).
studies, Nucleic. Acids. Res. 43 (7) (2015) e47. [49] C.E. Martinez, P.C. Smith, V.A. Palma Alvarado, The influence of platelet-derived
[33] J. Fernandez-Fuertes, T. Arias-Fernandez, A. Acebes-Huerta, M. Alvarez-Rico, products on angiogenesis and tissue repair: a concise update, Front. Physiol. 6
L. Gutierrez, Clinical response after treatment of knee osteoarthritis with (2015) 290.
a standardized, closed-system, low-cost platelet-rich plasma product: 1-year [50] A.F. Drew, H. Liu, J.M. Davidson, C.C. Daugherty, J.L. Degen, Wound-healing de-
outcomes, Orthop. J. Sports Med. 10 (3) (2022) 23259671221076496. fects in mice lacking fibrinogen, Blood 97 (12) (2001) 3691–3698.
[34] I. van der Bijl, M. Vlig, E. Middelkoop, D. de Korte, Allogeneic platelet-rich [51] C.T. Drinnan, G. Zhang, M.A. Alexander, A.S. Pulido, L.J. Suggs, Multimodal re-
plasma (PRP) is superior to platelets or plasma alone in stimulating fibrob- lease of transforming growth factor-beta1 and the BB isoform of platelet de-
last proliferation and migration, angiogenesis, and chemotaxis as relevant pro- rived growth factor from PEGylated fibrin gels, J. Control Release 147 (2) (2010)
cesses for wound healing, Transfusion 59 (11) (2019) 3492–3500. 180–186.
[35] E.W. Raines, D.F. Bowen-Pope, R. Ross, Plasma binding proteins for [52] A.T. Nurden, P. Nurden, M. Sanchez, I. Andia, E. Anitua, Platelets and wound
platelet-derived growth factor that inhibit its binding to cell-surface receptors, healing, Front. Biosci. 13 (2008) 3532–3548.
Proc. Natl. Acad. Sci. U.S.A 81 (11) (1984) 3424–3428. [53] M. Lamkanfi, N. Festjens, W. Declercq, T. Vanden Berghe, P. Vandenabeele, Cas-
[36] M. Cattaneo, M. Chiodini, P.M. Mannucci, Ristocetin and platelet aggregation, pases in cell survival, proliferation and differentiation, Cell Death Differ. 14 (1)
Br. J. Haematol. 71 (2) (1989) 301–302. (2007) 44–55.
[37] Y. He, Y. Pei, K. Liu, L. Liu, Y. Tian, H. Li, M. Cong, T. Liu, H Ma, H You, J Jia, [54] Y. Akasaka, Y. Ishikawa, I. Ono, K. Fujita, T. Masuda, N. Asuwa, K. Inuzuka,
D. Zhang, P. Wang, GITR/GITRL reverse signalling modulates the proliferation H. Kiguchi, T. Ishii, Enhanced expression of caspase-3 in hypertrophic scars and
of hepatic progenitor cells by recruiting ANXA2 to phosphorylate ERK1/2 and keloid: induction of caspase-3 and apoptosis in keloid fibroblasts in vitro, Lab.
Akt, Cell Death Dis 13 (4) (2022) 297. Invest. 80 (3) (20 0 0) 345–357.
[38] Z. Meng, G. Feng, X. Hu, L. Yang, X. Yang, Q.S.D.F. Jin, Factor-1α Promotes the [55] E. Anitua, M. de la Fuente, J. Merayo-Lloves, F. Muruzabal, G. Orive, Allogeneic
Migration, Proliferation, and Osteogenic Differentiation of Mouse Bone Marrow blood-based therapies: hype or hope? Eye 31 (4) (2017) 509–510.
Mesenchymal Stem Cells Through the Wnt/β -Catenin Pathway, Stem Cells Dev [56] J.M. Burkhart, M. Vaudel, S. Gambaryan, S. Radau, U. Walter, L. Martens,
30 (2) (2021) 106–117. J. Geiger, A. Sickmann, R.P. Zahedi, The first comprehensive and quantitative
[39] J.R. Evanson, M.K. Guyton, D.L. Oliver, J.M. Hire, R.L. Topolski, S.D. Zumbrun, analysis of human platelet protein composition allows the comparative analy-
J.C. McPherson, J.A. Bojescul, Gender and age differences in growth factor sis of structural and functional pathways, Blood 120 (15) (2012) e73–e82.
concentrations from platelet-rich plasma in adults, Mil. Med. 179 (7) (2014) [57] J. Coppinger, D.J. Fitzgerald, P.B. Maguire, Isolation of the platelet releasate,
799–805. Methods Mol. Biol. 357 (2007) 307–311.
[40] H.S. Cho, I.H. Song, S.Y. Park, M.C. Sung, M.W. Ahn, K.E. Song, Individual vari- [58] H.E. Kattlove, B. Alexander, The effect of cold on platelets. I. Cold-induced
ation in growth factor concentrations in platelet-rich plasma and its influence platelet aggregation, Blood 38 (1) (1971) 39–48.
on human mesenchymal stem cells, Korean J. Lab. Med. 31 (3) (2011) 212–218. [59] H.E. Kattlove, B. Alexander, F. White, The effect of cold on platelets. II. Platelet
[41] Y. Taniguchi, T. Yoshioka, H. Sugaya, M. Gosho, K. Aoto, A. Kanamori, M. Ya- function after short-term storage at cold temperatures, Blood 40 (5) (1972)
mazaki, Growth factor levels in leukocyte-poor platelet-rich plasma and cor- 688–696.
relations with donor age, gender, and platelets in the Japanese population, J. [60] Y.M. Kuo, T.A. Kokjohn, W. Kalback, D. Luehrs, D.R. Galasko, N. Cheval-
Exp. Orthop. 6 (1) (2019) 4. lier, E.H. Koo, M.R. Emmerling, A.E. Roher, Amyloid-beta peptides interact
[42] S. Dangwal, B. Stratmann, C. Bang, J.M. Lorenzen, R. Kumarswamy, J. Fiedler, with plasma proteins and erythrocytes: implications for their quantitation in
C.S. Falk, C.J. Scholz, T. Thum, D. Tschoepe, Impairment of wound healing in plasma, Biochem. Biophys. Res. Commun. 268 (3) (20 0 0) 750–756.

147

You might also like