Tesis Igor Irastorza Epelde
Tesis Igor Irastorza Epelde
Tesis Igor Irastorza Epelde
Thesis directors:
Dr. Fernando Unda Rodriguez
Dr. Gaskon Ibarrete Bilbao
Abstract …………………………………………………………………………………………………..…….…………… 1
Abbreviations ………………………………………………………………………………………..…………………… 5
Introduction ……………………………………………………………………………………………………………… 11
Introduction to tissue engineering ………………………………………………………………………. 13
Stem cells ……………………………………………………………………………………………..…………….. 14
Adult stem cells …………………………………………………………………………………….…………. 15
Mesenchymal stem cells ……………………………………………………………….…………….. 17
Dental pulp stem cells (DPSC) ………………………………………………………………….. 18
General introduction ……………………………………………………………..…………… 18
Surface markers ………………………………………………………………..……………….. 20
Cell differentiation of DPSCs ……………………………………………………..………… 21
Bone marrow stem cells (BMSC) …………………………………………………..…………. 24
General introduction ……………………………………………………………….…………. 24
Surface markers …………………………………………………………………………………. 25
Cell differentiation of BMSCs ……………………………………….……………………… 26
Scaffolds for bone tissue engineering ………………………………………………………………….. 28
Titanium ………………………………………………………………………………………..………………… 30
Decellularized adipose tissue ……………………………………………………………..……………. 33
Growth factors ………………………………………………………………………………….………………… 34
Plasma derived products ………………………………………………………….…………………….. 35
Material and Methods ……………………………………………………………………………….…………….. 37
Hypothesis ……………………………………………………………………………………………….………………. 49
General Objectives …………………………………………………………………………………….…………….. 53
Results ……………………………………………………………………………………………………………………… 57
Discussion ……………………………………………………………………………………………..…………………. 95
Conclussions …………………………………………………………………………………………….……………. 109
Annex I (patents) ……………………………………………………………………………………..…………….. 113
Annex II (articles) …………………………………………………………………………………..……………….. 117
Bibliography ………………………………………………………………………………………….……………….. 133
Abstract
Abstract
In the present work, we studied the human Dental Pulp Stem Cells (hDPSCs)
adherence, proliferation, viability and osteo-differentiation potential when cultured on
widely used Ti6AL4V titanium surface and a new biomimetic porous surface (BAS TM), in
the presence or absence of osteoblastic differentiation media supplemented with
plasma rich in growth factors (PRGF) and platelet rich fibrin (PRF).
The results showed that hDPSCs showed a good adherence ability and non
affected viability and proliferation when cultured on Ti6AL4V and BAS titanium surfaces.
Moreover, both titanium surfaces demonstrated having osteoblastic differentiation
induction effect on hDPSCs without using osteoblastic differentiation media. Besides,
the two plasma derived products showed interesting results. On one hand, the PRGF
induced higher cell proliferation on in vitro hDPSC cultures becoming a good FBS
substitute for the use on autologous cell therapies. On the other hand, PRF enhanced
osteoblastic cell differentiation of hDPSCs with higher calcified bone matrix production.
Finally, the combination of PRF with BAS titanium surface maximized hDPSCs
osteoblastic differentiation to bone-producing cells. The results obtained on this work
provides experimental support to the commonly used fibrin clots on clinical practice to
enhance bone production around dental implants.
3
Igor Irastorza Epelde
Keywords: dental pulp stem cells, DPSC, bone marrow stem cells, BMSC, tissue
engineering, scaffold, cell differentiation, plasma-derived products, PRGF, PRF, titanium,
dezellularized adipose tissue, regenerative medicine.
4
Abbreviations
Abbreviations
7
Igor Irastorza Epelde
8
Abbreviations
9
Introduction
Introduction
The term “Tissue engineering” was created in 1988 by the National Science
Foundation workshop as “the application of principles and methods of engineering and
life sciences toward the fundamental understanding of structure-function relationships
in normal and pathological mammalian tissues and the development of biological
substitutes to restore, maintain or improve tissue function”. Tissue engineering is a
multidisciplinary and relatively new field, which interconnects different disciplines as
clinical medicine, material science, mechanical engineering and genetics (Berthiaume et
al., 2011). Tissue engineering strives for functional restoration of lost or damaged tissues
by seeding cells and growth factors on three-dimensional scaffolds (Chaudhari et al.,
2016), conforming what is known as “tissue-engineering triad”, which can be set up in a
bioreactor in a controlled environment (Dlaska et al., 2015; O’Brien, 2011).
Taking into account bone tissue engineering criteria, craniofacial tissue engineering goes
forward by trying to develop biomaterials for the regeneration of dental and oral tissues,
like bone, salivary glands, periodontal ligaments, mucosa, cementum and dentin
(Rahman et al., 2018).
13
Igor Irastorza Epelde
regeneration”. This technique is based on the use of barrier membranes in alveolar ridge
defects to enhance the bone growth on the surrounding affected area of dental
implants. The alveolar bone growth had to be enhanced first to give local mechanical
stability for the correct bone-implant interface creation (Pilipchuk et al., 2015).
Summarizing, the three pillars on which tissue engineering is sustained are stem
cells, scaffolds and growth factors (figure 1).
Cells
Biomaterials
Autologous
Natural
Heterologous
Synthetic
Differentiated
Hydrogels Tissue Stem cell
Meshes
engineering
Signals
Growth factors
Small molecules
Mechanical forces
Figure 1. The tissue engineering triad. A combination of cells cultured on a biomaterial scaffold with
appropriate biophysical and chemical signals coordinate to regenerate the desired tissue.
1. Stem cells
The first described stem cells were obtained by the isolation of the embryonal
carcinoma cells (ECCs) from teratocarcinomas in 1954 (Stevens and Little, 1954). Ten
years later, the two main characteristics of these cells were described: their self-renewal
ability and their capacity to differentiate to the three different germ layers in vitro,
describing them as pluripotent stem cells (Kleinsmith and Pierce, 1964). In 1970, they
14
Introduction
were also described on in vivo models (Kahan and Ephrussi, 1970). Few years later, in
1981, they isolated the mouse embryonic stem cells (mESCs) from the inner cell mass of
the blastocyst (ICM) and in 1998, the human ESCs (Martin, 1980; Thomson et al., 1998).
In recent years, a new concept of stem cells was born, induced pluripotent stem cells
(iPSCs). In 2006, it was firstly described on mouse cells and in 2007 on human cells
(Takahashi et al., 2007; Takahashi and Yamanaka, 2006).
Totipotent: These types of stem cells have the potential to create an entire individual.
They are able to form an entire embryo and the temporary support tissues (umbilical
cord and placenta). This potential lasts until the blastomeric stage (Posfai et al., 2021).
Pluripotent: After the blastocyst is formed, the pluripotent stem cells forming the IMC
are able to differentiate into the three germ layers. These stem cells are capable to
generate all the adult tissues and organs but not the temporary support tissues (Smith,
2001).
Oligopotent: These term refers to the stem cells which can only differentiate to very few
and restricted types of cells like the myeloid and lymphoid cells (Majo et al., 2008).
Unipotent: The unipotent stem cells are able only to produce one type of cell. They are
considered stem cells because of their self-renewal capacity, like muscular stem cells
(Seale et al., 2001).
When the blastocyst is developed, the ESCs from the ICM create the three germ
layers: endoderm, mesoderm and ectoderm. After the tissue and organ formation,
some stem cells are retained without a terminal differentiation as in the bone marrow,
blood, liver, skin, brain, teeth, bone or muscle (Denham et al., 2005; Vats et al., 2005).
15
Igor Irastorza Epelde
The plasticity of these stem cells can vary from the potential to differentiate to many
different cell types (multipotent) to being able only to differentiate to a single type of
cell (unipotent) (Almeida-Porada et al., 2001; Wagers and Weissman, 2004). Besides, the
proliferation rate of these cells varies from tissue to tissue. The highest proliferative
stem cells can be found in high cell turnover tissues as bone marrow, skin or intestine
(Baulies et al., 2020; Lenkiewicz, 2019; Yadav et al., 2020). On the other hand, other
stem cells will only proliferate to replace damaged cells or in a response to an injury such
as in the liver, heart or the nervous system (Angelini et al., 2004; Duncan et al., 2009;
Mansergh et al., 2000) (figure 2).
16
Introduction
It was 1990 when the term “mesenchymal stem cell “ started to be popular
thanks to its use by Caplan but it took until 2000 to be accepted by the scientific
community (Caplan, 1991; Horwitz and Keating, 2000). They are named MSCs the non-
hematopoietic stem cells with the capacity to differentiate to different mesenchymal
tissue cells. They can be isolated from various tissues and organs such as muscles,
cartilage, bone, tendons, adipose tissue and perivascular area (Chamberlain et al., 2007;
Crisan et al., 2008), and from umbilical cord, menstrual blood, placenta, large intestine
and dental pulp (Du et al., 2016; Ma et al., 2014; Portmann-Lanz et al., 2006; Tirino et
al., 2011).
Different MSC populations have been described in the adult body. The first MSC
population to be described were the bone-marrow mesenchymal stem cells (BMSCs)
(Anjos-Afonso and Bonnet, 2007; Friedenstein et al., 1976). Then they followed different
MSC populations such as umbilical cord mesenchymal stem cells, discovered in 1991 and
confirmed as MSCs in 2004 (McElreavey et al., 1991; Wang et al., 2004), dental pulp
mesenchymal stem cells (Gronthos et al., 2000), cardiac mesenchymal stem cells
(Beltrami et al., 2003), pulmonary mesenchymal stem cells (Griffiths et al., 2005),
peripheral blood mesenchymal stem cells (Cao et al., 2005) or adipose tissue
mesenchymal stem cells (AT-MSCs) (Fraser et al., 2006). Despite most of the cell types
mentioned before come from the mesenchymal germ layer, the origin of oral cavity
mesenchymal stem cells, such as DPSCs, is in the neural crest, as DPSCs. Even been
widely classified as mesenchymal stem cells, the ectodermal germ layer origin gives
these cells have a higher ability to differentiate to neuron-like cells compared to MSCs,
being possible to call them ectomesenchymal stem cells (Ibarretxe et al., 2012a).
17
Igor Irastorza Epelde
enamel
crown dentin
pulp
gum
(gingiva)
cementum
root blood
vassels
periodontal
ligament
lateral
canals
nerve
DPSCs were described for the first time in 2000 as mesenchymal stem cells from
the dental pulp similar to BMSC (Gronthos et al., 2000). It was two years later when they
were named as dental pulp stem cells and they were characterized as highly clonogenic
18
Introduction
and proliferative cells (Gronthos et al., 2002). These cells are located in the “pulp
chamber” inside the dental crown (Figure 3). Dental pulp is a connective tissue with a
heterogeneous composition, including different cell types such as
odonto/osteoprogenitor cell populations, neural cells, vascular cells, fibroblasts (known
as pulpoblasts) and immune cells as granulocyte and macrophages (Goldberg and Smith,
2004). It is also important to mention that some studies located the DPSCs in the
vascular pericyte compartment (Shi and Gronthos, 2003).
Hard tissues as cementum, dentin and enamel compose the adult tooth as well
as the soft tissues, like the dental pulp. Dental formation starts with the interaction
between the MSCs and the oral ectodermal epithelial cells. Dentin is the first tissue being
formed, followed by enamel and the dental follicle. The epithelial cells form ameloblasts,
which create the enamel. On the other hand, MSCs are the responsible for generating
the dentin, cementum, periodontal ligament and pulp (Sedgley and Botero, 2012;
Thesleff and Aberg, 1999) (Figure 3).
One of the main functions of the dental pulp in the adult body is the repair and
maintenance of the dentin. In cases with severe damage as in deep caries, the DPSCs
migrate to the injury zone and they differentiate to odontoblasts to create reparative
dentin (Dimitrova-Nakov et al., 2014; Tziafas et al., 2000).
Even if the DPSCs are the most known and more commonly used dental stem cells
in research, we can find different stem cell population with different properties. The
dental follicle stem cells (DFSCs) can be isolated during tooth development from the
ectomesenchymal embryonic tissue where they are covering the tooth germ (Zhang et
al., 2019). They can differentiate in vitro to other cell linages apart from periodontal cells
(Honda et al., 2010). Moreover, stem cells from apical papilla (SCAP) are localized in the
apices of the root of developing tooth as in pre-erupting wisdom teeth with high
proliferative ability (Bakopoulou et al., 2011). As the other stem cell subpopulation of
this group, because of the ectomesenchymal origin, they are also able to differentiate
to angiogenic and neurogenic cells (Dagnino et al., 2020; Nada and El Backly, 2018).
Furthermore, periodontal ligament stem cells (PDLSCs) were found to have very similar
characteristics to DPSCs as pluripotency markers and differentiation potential (Liu et al.,
2018; Seo et al., 2004). Finally, primary exfoliated deciduous teeth stem cells (SHED) also
19
Igor Irastorza Epelde
show pluripotency markers, neural markers and high proliferation rate (Kerkis et al.,
2006; Miura et al., 2003). Like the rest of the dental stem cells, SHED have demonstrated
the ability to differentiate into mesenchymal linage cells at the same time as to neural
cells (Karbalaie et al., 2021; Sordi et al., 2021; Zhang et al., 2016) (Figure 4).
Figure 4. Timeline about the highlights in the history of the isolation of dental-related stem cells.
(Karamzadeh and Eslaminejad, 2013).
20
Introduction
commonly reported marker expression pattern for DPSCs, some studies showed
expression of CD34 in certain DPSC subpopulations (Carnevale et al., 2018) (table 1).
21
Igor Irastorza Epelde
vary, dexamethasone, β-glycerol phosphate and L-ascorbic acid are constantly repeated,
as they are the most important components. These molecules are essential to activate
the expression of collagen type I, alkaline phosphatase, Run-related transcription factor
2 (RUNX2), osterix, osteopontin, osteocalcin and osteonectin, necessary for the
osteodifferentiation of the DPSCs (Ajlan et al., 2015; Atari et al., 2012a; Bhuptani and
Patravale, 2016; Goto et al., 2016; Riccio et al., 2010). Among all these genes, RUNX2 is
one of the first being expressed. RUNX2 is an early stage osteodifferentiation gene
whose activation provokes the expression of both dentin sialoprotein (DSP) and dentin
sialophosphoprotein (DSPP) (Han et al., 2014; Vimalraj et al., 2015; Xu et al., 2015).
DPSCs are also capable to differentiate to other cells types such as endothelial
cells (Luzuriaga et al., 2019a; Luzuriaga et al., 2020; Marchionni et al., 2009),
cardiomyocytes (Ferro et al., 2012) and smooth muscle (Song et al., 2016; Zhao et al.,
2012). In addition, recent studies obtained endothelial cell differentiation using serum-
free media, an interesting approach for autologous cell therapies (Luzuriaga et al.,
2019a; Luzuriaga et al., 2020).
22
Introduction
Apart from the differentiation to mesenchymal germ layer cells one of the most
interesting characteristic of DPSCs is their neural crest origin. Due to this origin, DPSCs
are able to differentiate to neuron-like cells. Many neuron differentiation inductive
media used in the last decade are based on neurotrophins and other small molecules
like NGF, GDNF, BDNF, epidermal growth factor (EGF), fibroblast growth factor (FGF),
sonic hedgehog, forskolin, heparin, NT-3 and retinoic acid (RA). It is also remarkable the
use of ITSx, N2, B27 and non-essential amino acids (Arthur et al., 2008; Chang et al.,
2014; Gervois et al., 2015; Kanafi et al., 2014; Király et al., 2011; Luzuriaga et al., 2019b;
Osathanon et al., 2014; Xiao and Tsutsui, 2013; Zhang et al., 2017) (figure 5).
Adipogenic Myogenic
Chondrogenic Neurogenic
Osteo/dentinogenic
23
Igor Irastorza Epelde
DPSCs showed the potential to generate endodermal lineage cells (Atari et al.,
2012b). The ability to differentiate to endoderm germ-line cells as pancreatic cells was
demonstrated by obtaining cells capable to produce glucagon, somatostatin, insulin and
pancreatic polypeptide being the CD117+ cells the subpopulation with the best
differentiation potential. The WNT and PI3K/AKT pathways showed to have a key role
on this differentiation (Ishkitiev et al., 2013; Yagi Mendoza et al., 2018). Moreover,
hepatic-like differentiation had been carried with the use of dexamethasone, ITSx, HGF
and oncostatin M cultured in serum-free or low serum (1 %-2 %) media (Chen et al.,
2016; Han et al., 2017; Ishkitiev et al., 2012).
The CFU-Fs showed to have a low frequency, ranging from 1/10.000 to 1/100.000
of bone marrow mononuclear cells (MNCs) (Castro-Malaspina et al., 1980). The
frequency of BMSCs is much lower than hematopoietic stem cells (HSCs) also clustered
in the bone marrow, being 1% of the MNC fraction (Civin et al., 1996).
Moreover, the BMSCs cell cycle investigations showed that a small fraction of
these cells are proliferative (10 % at S + G2 + M), and all the rest at G0/G1 phase (Conget
and Minguell, 1999). Depending on patients variability, the proliferation potential of
BMSCs can vary from four doublings to going over 15 doublings (Digirolamo et al., 1999;
Phinney et al., 1999). Even if the proliferative cells are a small fraction, they have high
proliferation rate ex vivo. This proliferation potential can vary depending on the
procedure to harvest the cells, the frequency of BMSCs and the age of the donor (Blazsek
et al., 1999; Koç et al., 1999). Recent studies have improved in vitro proliferation of
24
Introduction
BMSCs using bioreactors (Bhat et al., 2021; Santos et al., 2011). Despite telomerase
activity being lost in somatic cell proliferation and differentiation causing cell death by
senescence, BMSCs maintain the telomerase activity (Pittenger et al., 1999). However,
after extensive passages, they show apoptosis and senescence signs (Dhanasekaran et
al., 2013).
Some investigations suggested that BMSCs, as DPSCs, are located in the vascular
pericyte, the cell lining of the outer surface of blood vessels (Shi and Gronthos, 2003;
Short et al., 2003; da Silva Meirelles et al., 2006). Besides, other studies indicates that
the BMSCs could be identical to bone marrow stromal supportive cells termed
adventitial reticular cells (ARCs). As a result, BMSCs are also referred as multipotent
mesenchymal stromal cells or marrow stromal stem cells (Gronthos et al., 2003; Horwitz
et al., 2005).
25
Igor Irastorza Epelde
(Endoglin) and SSEA-4 (stage-specific embryonic antigen) (Boiret et al., 2005; Delorme
and Charbord, 2007; Gang et al., 2007; Jones et al., 2006). Finally, it is important to point
CD271 (low-affinity nerve growth factor receptor), as one of the best bone marrow
subpopulation markers for BMSCs. This expression difference between subpopulations
is based on the high expression on these cells and low expression in all other marrow
cell populations (Bühring et al., 2007; Cuthbert et al., 2015) (Table 1).
c. Differentiation
The MSCs from the bone marrow are non-hematopoietic cells that exhibit
multilineage differentiation capacity. Many studies demonstrated the ability of these
cells to give rise to diverse mesenchymal derivatives like cartilage, cardiac muscle,
skeletal muscle, bone and adipose tissue (Arthur et al., 2009; Bianco et al., 2001; Conget
and Minguell, 1999; Matsuda et al., 2005; Prockop, 1997; Zheng et al., 2013). Contrary
to their original germ line, they have also the potential to differentiate into
neuroectodermal cells as neuron-like cells (Krampera et al., 2007; Sanchez-Ramos et al.,
2000; Zhang et al., 2012). Even so, this neuron-like cell differentiation potential is lower
than DPSCs differentiation ability (Isobe et al., 2016; Pagella et al., 2020).
Otherwise, for the adipogenic differentiation, the most used media cocktails are
based on dexamethasone, indomethacin and IBMX (Zheng et al., 2013).
26
Introduction
27
Igor Irastorza Epelde
Figure 6. Differentiation potential of bone marrow mesenchymal stem cells (BMSCs). (Muruganandan
et al., 2009).
28
Introduction
polymers (hyaluronic acid and chitosan) and synthetic ones (polyglicolide and
polylactide) are common examples (Navarro et al., 2008).
Figure 7. Bone tissue engineering. Bone scaffold with autologous stem cells put in a bioreactor with
growth factors enable the growth of autologous bone grafts in vitro for bone repair applications in vivo.
(Ng et al., 2017).
The most commonly used synthetic polymers are poly-glycolic acid (PGA), poly-
lactic acid (PLA) and their copolymers. Their degradation rate control and excellent
mechanical properties make them a good choice for cell transplantation for tissue
engineering (Dorati et al., 2017).
29
Igor Irastorza Epelde
Moreover, natural biological components are proteins (collagen, fibrin gels, soy
and silk) and polysaccharides (alginate, chitin/chitosan, starch, and hyaluronic acid
derivates). They are a good option for enhancing cell adhesion, however, they allow less
control on the biodegradability and mechanical characteristics. Besides, they could
produce some immunogenicity (Dorati et al., 2017).
Finally, we can find inorganic materials such as, metals, tricalcium phosphate
(TCP), bioactive glasses, hydroxyapatite (HA), wollastonite and several combination of
them as HA-TCP biphasic ceramics. The best feature they have is the similarity to the
bone mineral phase (Kokubo and Takadama, 2006; Lakshmi and Sasikumar, 2015;
Stevens, 2008). The main disadvantage of these materials is the low or lack of
degradation so they cannot be reabsorbed, thus hindering the creation of new tissue.
One of the features that seems to be crucial for cell proliferation and
differentiation in recent investigations is the porosity of the scaffolds. Some studies
showed that pore sizes between 100 and 400 µm were the perfect size for the formation
of mature bone by enhancing vascularization and cell infiltration (Boyan et al., 2016a;
Murphy et al., 2010), while other studies suggested that 800 µm pore size was more
appropriate for the cell growth (Roosa et al., 2010). In contrast, small pore sizes (<100
µm) are related with fibrous tissue and non-mineralized osteoid formation (Iviglia et al.,
2019; Liu et al., 2018). At the same time, micropores have more surface area, promoting
ion exchange and cell and bone protein adhesion (Abbasi et al., 2019; Diaz-Rodriguez et
al., 2018; Morejón et al., 2019).
2.1. Titanium
In the last half a century, titanium implants have been established as the best
option for teeth replacement due to their resistance, durability and their high
30
Introduction
Despite titanium proved to be the best material for dental implants, in the last
decades it has been demonstrated the importance of the design and manufacturing
different implant surfaces in order to enhance their osteointegraton (Gasik et al., 2015;
Rani et al., 2012; Rupp et al., 2018; Salou et al., 2015). Apart from the composition of
the titanium, it is important to consider the topography of its surface for a durable
anchorage of the titanium implant (Annunziata and Guida, 2015; Jemat et al., 2015; Le
Guéhennec et al., 2007; Naves et al., 2015).
Rough titanium surfaces, maximizing the contact surface with the surrounding
alveolar bone, has proven to have a better osteointegration compared to smooth
titanium ones (Coelho et al., 2009). The better integration in the bone could be
attributed to the microporosity of the titanium surface due to the promotion of
osteoblastic differentiation of surrounding mesenchymal cells. Lastly, these
mesenchymal cells are responsible for consolidation and bone healing after the
placement of the implant (Boyan et al., 2016a; Graziano et al., 2008).
31
Igor Irastorza Epelde
1- Α phase stabilizers: carbon (C), oxygen (O), aluminium (Al) and nitrogen (N).
2- Β phase stabilizers:
a. Isomorphous: tantalum (Ta), molybdenum (Mo), vanadium (V) and
niobum (Nb).
b. Eutectoid: silicon (Si), chromium (Cr), copper (Cu), hydrogen (H),
manganese (Mn), nickel (Ni) and iron (Fe).
3- Neutral additions: tin (Sn) and zirconium (Zr).
32
Introduction
Most of the actual dental implant roots are inert surface-micropatterned metal
alloys. The use of these implants alone or in combination with decellularized bone
matrix and plasma derived growth factors promotes the osteodifferentiation of
mesenchymal stem cells, generating bone directly in the implant surface (Anitua et al.,
2016; Boyan et al., 2016a; Mayer et al., 2018; Zanicotti et al., 2018). The main problem
of actual dental implants is developing periimplantitis. This is caused by the continuous
mechanical pressure transferred from the implant to the bone tissue during natural
mastication. In the worst scenario, periimplantitis leads to the loss of the dental implant
(Bertolini et al., 2019). In natural teeth, this problem is solved thanks to the periodontal
ligament (PDL), a highly innervated and vascularized connective tissue composed by
strong collagen bundles. These collagen fiber bundles are anchored to the dental root
cement and the alveolar bone by lightly calcified edges, also named as Sharpey´s
perforating fibers (Aaron, 2012). The principal PDL function is acting as a cushion to
absorb the masticatory forces to protect alveolar bone.
33
Igor Irastorza Epelde
formulation is the solid foam due to the porosity and the high volume per weight ratio,
allowing nutrients, signals and cell mobility through the scaffold matrix (figure 9).
Figure 9. Macroscopic (A, B) and scanning electron microscopic (SEM) (C, D), images of native porcine
adipose tissue (A, C) and decellularized extracellular matrix (ECM) (B, D). Scale bar: 50 µm. (Choi et al.,
2012).
3. Growth factors
With the purpose of enhancing local healing in the damaged zone, the concept
of concentrated platelets and autologous growth factors was born by collecting plasma
solutions (Anfossi et al., 1989; Fijnheer et al., 1990). The name given in the late 1990s
was platelet rich plasma (PRP) (Jameson, 2007; Marx et al., 1998; Whitman et al., 1997).
34
Introduction
PRP is composed of platelets that when activated releases growth factors enhancing cell
proliferation, adhesion and migration of the cells in the damaged zone (Jameson, 2007;
Marx, 2004). After PRP, a second plasma derived product was formulated using
anticoagulants, which was termed platelet-rich growth factor (PRGF) (Anitua, 1999).
Moreover, to avoid the biggest limitation of these two products, being difficult to
handle, a second generation of plasma derived product was created by Choukoun et al.
in 2001 to enhance hard and soft tissue regeneration in maxillofacial and oral surgery,
platelet rich fibrin (PRF) (Kumar and Shubhashini, 2013).
Among the growth factors and cytokines being released by activated platelets
the following can be included: transforming growth factor β1 (TGF-β1), platelet derived
growth factor (PDGF), vascular endothelial growth factor (VEGF), fibroblast growth
factor (FGF), epidermal growth factor (EGF) and interleukins (IL) as IL-1β, IL-4 and IL-6
(Dohan et al., 2006).
Despite the preparation of PRP and PRGF is almost the same, PRGF contains more
plasma proteins and coagulation factors, enhancing the effect comparing to PRP (Anitua,
1999).
The mixture of factors of both plasma-derived products, PRGF and PRF, induces
the proliferation and differentiation of the surrounding mesenchymal cells to
osteoblastic cells. One of the best characteristics of this product is the possibility to be
used as autologous personalized therapy (Anitua et al., 2013). The benefits of the use of
this product are widely exploited in different experimental and clinical tissue
regeneration therapies and especially in implant dentistry therapies (Anitua et al., 2016;
Masuki et al., 2016) (figure 10).
35
Igor Irastorza Epelde
To obtain PRF it is necessary a fast centrifugation after the blood extraction since
it is extracted in absence of anticoagulants. After the separation of blood fractions, the
clot containing platelet and leukocytes, can be easily manipulated and flattened into a
membrane. The most interesting feature of it is the progressive release of the growth
factors due to the degranulation of the platelet over time. After surgery, these growth
factors stimulate tissue healing upon enhancing cell proliferation and chemotaxis. The
fibrin clot is easy to manipulate, resistant, strong and flexible, giving them great
adaptability characteristics to be used in varied anatomical surfaces (Khurana et al.,
2017; Kumar et al., 2016).
Nowadays, the use of plasma rich in growth factors (PRGF) and platelet rich fibrin (PRF)
in the dental clinic to promote bone healing for the placement of the titanium implant
after the tooth extraction is widely extended. It is known as plasma-derived advanced
medical therapy products (AMTP) (Giannini et al., 2015; Kobayashi et al., 2016;
Nishiyama et al., 2016).
36
Materials and Methods
Material and Methods
The hBMSCs were obtained in the Laboratory for Cell and Tissue Engineering
(Ehrbarlab) of the University of Zurich (Zurich, Switzerland) with the following
procedure: the hBMSCs were obtained from aspirates of the bone marrow during
orthopedic surgical procedures of human healthy donors (average age 45 years old). The
aspirate was obtained after informed consent established by the local ethical committee
(University Hospital Basel; Prof. Kummer; approval date 26/03/2007 Ref. Number
78/07). For the isolation of nucleated cells, the red blood cells from the aspirate were
lysed using a buffer containing 0.15 M NH4Cl (Sigma, Switzerland), 1 mM KHCO3 (Sigma,
Switzerland) and 0.1 mM Na2EDTA (Fluka, Switzerland). hBMSC cells were cultured in
minimal essential medium (αMEM, Gibco, NY, USA) supplemented with 10 % FBS (Gibco,
NY, USA), 1 % penicillin-streptomycin (Gibco, NY, USA) and 5 ng/ml fibroblast growth
factor 2 (FGF2) (PeproTech, NJ, USA) at 37 °C in 5 % CO2. Passages between 3 and 8
maximum culture age were used on experimentation.
Human dental pulp stem cells (hDPSCs) were isolated from the third molar teeth
of young healthy patients (age 18-30) in dental clinics. All the patients signed an
informed consent. The competent authority (Ethics committee of the University of the
Basque Country) under the M10_2016_088 protocol had previously approved the
procedures officially.
Dental pulp extraction was performed by fracturing the molar. The obtained pulp
was digested with an enzymatic solution containing 3 mg/mL collagenase type I (Gibco,
NY, USA) and 4 mg/mL dispase (Sigma, MO, USA) in Hank´s balanced salt solution (HBSS,
Gibco, NY, USA), and then incubated at 37 °C for 1 hour. The enzymatic digestion
solution was neutralized with Dulbecco´s modified eagle medium (DMEM, Lonza,
Switzerland) supplemented with 10 % fetal bovine serum (FBS, HyClone, UT, USA). Then,
the cells were centrifuged at 1500 rpm for 5 minutes. The pellet was mechanically
disrupted by 18G needles (BD Microlance, Fisher Scientific, UK). After the disassociation,
39
Igor Irastorza Epelde
the cells were seeded in 25 cm2 flask (Sarstedt, Nümbrecht, Germany) with DMEM
supplemented with 10 % FBS, 1 % penicillin-streptomycin (Gibco, NY, USA) and 1 % L-
glutamine (Sigma, MO, USA) at 37 °C in 5 % CO2. After reaching 80 % confluence, the
cells were subcultured to 75 cm2 flasks (Sarstedt, Nümbrecht, Germany). Passages
between 4 and 10 and maximum culture age were used on experimentation.
For hDPSC cultured on titanium surfaces and pDAT solid foams experimentation
the osteogenic media was composed by DMEM (10 % FBS, 1 % penicillin/streptomycin
and 1 % L-glutamine) supplemented with 50 µM ascorbic acid (Sigma, MO, USA), 20 mM
β-glycerolphosphate (Sigma, MO, USA) and 10 nM dexamethasone (Sigma, MO, USA).
For the comparative study of hDPSCs and hBMSCs the osteogenic media were composed
by DMEM (10 % FBS, 1 % penicillin/streptomycin and 1 % L-glutamine) supplemented
with 50 µM ascorbic acid, 10 mM β-glycerolphosphate and 100 nM dexamethasone. For
these comparative experiments, both control and osteogenic media were
supplemented with 5 ng/ml EGF (AF-100-15-500UG, PeproTech, NJ, USA) .
Ti6Al4V 2 mm thick discs were obtained cutting a 5.5 mm diameter bar, provided
by Avinent Implant System SLU (Barcelona, Spain). For the manufacture of BAS™
(Biomimetic Advanced Surface) titanium surface, one face of the titanium disc was
treated by shot blasting using white corundum (Al2O3) F60 with projection particle sizes
of 212-300 μm. Next, the discs were washed in an ultrasonic bath for 10 min and rinsed
with deionized water. Afterwards, they were anodized by connecting them to a DC
power source anode and using calcium and phosphorus as an electrolyte in an aqueous
solution. A current was applied to this solution with a density of 0.75 mA/mm2 and the
potential was freely allowed to increase until reaching 130 V. Subsequently, the titanium
discs were rinsed by deionized water for 10 minutes in an ultrasonic bath. Ti6Al4V
titanium discs did not undergo any surface treatment or polishing.
40
Material and Methods
Porcine adipose tissue was harvested in a local food company (Jaucha SL,
Navarra, Spain). The tissue was defrosted at room temperature, cleaned and creamed
by a beater. Then, tissue was homogenized on ice by Polyton (PT3100) with two different
rods at 1200 rpm for 5 minutes. To produce phase separation of lipids, homogenized
tissue was centrifuged at 900 g with ultrapure water for 5 minutes. Lipids were discarded
manually and proteins conserved. The protein pellets were treated overnight under
orbital shaking with isopropanol (Merk Life Science, Spain) at room temperature.
Afterwards, it was cleaned with PBS and treated using 1 % Triton X-100 and 0.1 %
ammonium hydroxide (Merk Life Science, Spain) in an orbital shaker for 36 hours at
room temperature. After cleaning the material again with PBS it was lyophilized to
completely dry up. Then, the tissue was milled by a mixer mill (Retsch MM400) to obtain
fine-grained powder suitable for processing. The resulting powder was frozen in liquid
nitrogen and storage at 4°C in a vacuum desiccator.
Solid foams were prepared by pDAT freeze-drying method. 0.5 M acetic acid was
added to 0.5 % pDAT, the solution was homogenized by magnetic stirring for 48 hours
at room temperature. Teflon moulds of 20 mm diameter and 3 mm thickness were used
for Scanning Electron Microscopy (SEM) with 1 ml solution. In the case of in vitro cell
culture assays, solid foams were formed in Millicell EZ-slide 8 well glass slides (Merk
Millipore) using 120 µl solution per well. The resulting slides were compatible for optic
and fluorescence microscopy. To obtain solid foams the samples were frozen at -20 °C
and freeze-dried (0.63 mbar and -10 °C). Solid foams for cell culture were sterilized by
ethylene oxide (Esterilizacion SL, Barcelona, Spain).
For the preparation of PRGF, blood from human healthy young donors was
collected in 5 ml blood collection tubes with 3.8 % sodium citrate anticoagulant (BD
Vacutainer, Plymouth, UK). After centrifugation at 580 g for 8 minutes, platelet-rich
fraction was transferred to 15 ml falcon tubes. Then, the platelets were activated with
41
Igor Irastorza Epelde
For obtaining PRF, blood was collected in tubes without anticoagulant (BD
Vacutainer, Plymouth, UK). The samples were immediately centrifuged at 580 g for 8
minutes to avoid the blood coagulation. After the coagulation and separation of blood
fraction and fibrin clot, the platelet-containing fibrin clot was separated from the
hematocrit fraction by scalpel and forceps. Then it was compressed between two glasses
by weight for 1 hour at room temperature. The fibrin clots were cut to obtain smaller
sections of around 1 cm2 (figure 11).
Anticoagulant
Platelet
activation
Centrifuge blood with CaCl2
PRGF
sample
Plasma PRF
Blood Buffy coat
withdraw Erytrocytes Fibrin
coagule
No anticoagulant formation Compression
device
Figure 11. Illustration of plasma-derived PRGF and PRF preparation. (Own creation).
When the cells reached 80 % confluence, they were detached from the bottom
of the flask with 0.05 % trypsin-EDTA, centrifuged and counted. Different densities of
cells were seeded on Ti6Al4V and BAS™ titanium discs, depending on the assay. To
42
Material and Methods
improve attachment, 15.000 cells were seeded in a 70 µl drop on the discs, and
incubated for 3 hours at 37 °C. Afterwards, 700 µl of DMEM medium was added for 24
well plates and 4 ml for 6 well plates.
When the cells reached 80 % confluence, they were detached from the bottom
of the flask with 0.05 % trypsin-EDTA, centrifuged and counted. pDAT (Tecnalia,
Donostia, Spain) was placed in 24 well plates (Sarstedt, Nümbrecht, Germany) and 8 well
Ibidi plates (Ibidi, Munich, Germany). After hydrating the pDAT with DMEM medium, the
hDPSCs were seeded in a concentration of 10.000-20.000 cells/well (depending on the
assay). The medium was changed every 2-3 days.
After 4 days of culture, the cells were washed with PBS. This step was followed
by an incubation of the cells with 3 µM calcein-AM (Molecular probes, OR, USA) and 2.5
μM propidium iodide (Sigma, MO, USA) for 30 minutes at 37 °C. Afterwards, the cells
were washed with PBS three times and photographed by using an Axioskop fluorescence
microscope (Zeiss, Overkochen, Germany) and a Nikon DS-Qi1 camera (Nikon, Tokyo,
Japan).
Immunocytochemistry
After culture, the cells were washed with PBS and fixed in 4 % paraformaldehyde
(PFA) at room temperature for 10 minutes. Then, the cells were incubated for 10
minutes with 10 % goat serum (Invitrogen, CA, USA) at room temperature. The blocking
step was followed by overnight incubation at 4 °C in 0.1 % Triton X-100/ 1 % BSA/ PBS
with the following antibodies: BGLAP (ab93876, Rabbit polyclonal, Abcam, Cambridge,
UK), Caspase-3 (ab32351, Rabbit monoclonal, Abcam, Cambridge, UK), Ki67 (ab15580,
Rabbit polyclonal, Abcam, Cambridge, UK), Osterix (ab22552, Rabbit polyclonal, Abcam,
Cambridge, UK) and SPARC (ab14174, Rabbit polyclonal, Abcam, Cambridge, UK). Alexa
43
Igor Irastorza Epelde
488 conjugated rabbit IgG (A11008, Abcam, Cambridge, UK) was used as secondary
antibody for primary antibody localization in 1 % BSA/ PBS for 1 hour at room
temperature. The nuclei were stained with 4’,6-diamino-2-phenilindol (DAPI, Invitrogen,
CA, USA). The images were taken using an Axioskop fluorescence microscope (Zeiss,
Overkochen, Germany) and Nikon DS-Qi1 camera (Nikon, Tokyo, Japan) and by Leica
DM6000 B microscope (Leica, Wetzlar, Germany), Leica DFC420 C camera with
3.3.3.16958 Leica application suite X (Leica, Wetzlar, Germany).
Flow cytometry
The cells cultured on both titanium surfaces were fixed with 2 % glutaraldehyde
(Sigma, MO, USA) in 0.1 M Sorensen phosphate buffer (SPB, Thermo Fisher Scientific,
MA, USA) for 1 hour at 4 °C. The samples were washed 3 times with 4-8 % sucrose in 0.1
M SPB and followed by other three washes in 0.1 M SPB. Then, the cells were dehydrated
with 15 minutes series of ethanol (30 %, 50 %, 70 %, 90 % and 100 %) and a final wash
by hexamethyldisilazane (Sigma, MO, USA). After dehydration, the samples were air-
dried and sputten coated by 15 nm gold. The images were taken using a scanning
electron microscope (SEM) (S4800, Hitachi High Technologies, Tokyo, Japan). Moreover,
images of titanium discs without cells were also obtained by SEM.
44
Material and Methods
The samples were fixed with 2 % glutaraldehyde for 15 minutes and embedded
in Epon Polarbed resin (Electron Microscopy Science, Hatfield, PA, USA). 70 nm ultrathin
sections were deposited onto 150 mesh copper grids (150 Square Mesh xopper 3,05
mm, AGAR Scientific, UK). Post-staining was performed with 2 % uranyl acetate in
distilled water (AGAR Scientific, UK) and 0.2 % lead in citrate in distilled water (AGR1210,
AGAR Scientific, UK). The images were taken in a Philips EM208S transmission electron
microscope with an integrated Jeol JEM 1400 Plus camera.
Cell pellets were stored frozen at -80 °C until use. The RNA extraction was
performed using RNeasy mini kit (Qiagen, Hilden, Germany). RNA concentration and
purity was measured by 260/280 nm absorbance in Nanodrop Synergy HT device
(Biotek, VT, USA). cDNA synthesis was made by retrotranscription was made starting
from 1000 ng of RNA using iScript cDNA Kit (Biorad, CA, USA). The retrotranscription
cycle started with 5 minutes at 25 °C, followed by 20 minutes at 46 °C and finished by 1
minute at 95 °C. Once the retrotranscription finished the lid stayed at 4 °C until the
samples were frozen at -20 °C.
RNA sequencing
hDPSCs and hBMSCs were cultured on plastic, Ti6AL4V and BAS titanium surfaces
in presence or absence of osteoblastic differentiation media for 14 days. 24 seeded discs
were used for each condition, after tripsinization the cells were put together and the
RNA was extracted using Qiagen RNA extraction kit. The RNA extracted from hDPSCs
and hBMSCs were sequenced by the Genomic center of the University of Zurich using
Illumina (Illunmina, CA, USA) obtaining 25.000 RNA lectures per sample. The gene
expression were analyzed by presence/absence (ON/OFF) term of different condition
45
Igor Irastorza Epelde
comparison and the threshold was set at limit of 10 raw reads to categorize a gene as
effectively expressed (ON). Statistical significantly activated pathways were analyzed by
“Pathway Enrichment Analysis” using The Connectivity Map (CMAP) and Gene Ontology
enrichment Biological Process (GOBP) by the Genome Analysis Platform of the CIC
bioGUNE (CIC bioGUNE, Derio, Spain).
Power SYBR Green PCR Master Mix (Applied biosystems, CA, USA) was used to
perform the Q-PCR. The initial step for the amplification program was at 95 °C for 10
minutes, followed by 40 cycles of 95°C for 20 seconds, 59 °C for 1 minute. The
housekeeping genes used were β-actin 5´-GTTGTCGACGACGAGCG-3´ and 5´-
GCACAGAGCCTCGCCTT-3´ and Gapdh 5´ CTTTTGCGTCGCCAG -3´ and 5´-
TTGATGGCAACAATATCCAC -3´. The target genes forward and reverse sequences were
as follows: collagen I (COL I) 5´ -GGCCCCCTGGTATGACTGGCT-3´ and 5´-
CGCCACGGGGACCACGAATC-3´, osteonectin (SPARC) 5´-GAAAGAAGATCCAGGCCCTC-3´
and 5´-CTTCAGACTGCCCGGAGA-3´, osteocalcin (BGLAP) 5´-CGCCTGGGTCTCTTCACTAC-
3´ and 5´-CTCACACTCCTCGCCCTATT-3´, dentin sialophosphoprotein (DSPP) 5´-
TGCCCAAATGCAAAAATATG-3´ and 5´-GTGGGCCACTTTCAGTCTTC-3´, osterix (OSX) 5´-
TGAGGAGGAAGTTCACTATG-3´ and 5´-CATTAGTGCTTGTAAAGGGG-3´and runx2
(RUNX2) 5´-CACTCACTACCACACCTACC-3´ and 5´-TTCCATCAGCGTCAACAC-3´. The target
gene expression was normalized against the housekeeping genes.
Briefly, the cells were fixed with 4 % PFA for 1 minute and washed 3 times by
0.05 % Tween 20-PBS. For the ALP staining, 5-bromo-4-chloro-3-indolyl phosphate/nitro
blue tetrazolium (NBT/BCIP; Sigma, MO, USA) was used as a substrate and the staining
was checked every 3 minutes. After the staining was concluded, cells were washed three
times for 5 minutes with PBS. The images were taken using a Zeiss Stemi 2000-C
stereoscopic microscope (Zeiss, Germany) and Canon PowerShot A80 camera (Canon,
Tokyo, Japan). For ALP enzymatic activity quantification, cells were detached by 0.05 %
46
Material and Methods
To prove the osteogenic potential of hDPSCs and hBMSCs, Alizarin Red was used
to stain extracellular calcium deposits. After 21 days of culture, the cells were fixed for
10 minutes with 4% paraformaldehyde. The samples were washed with distilled water
and then stained in dark using 2 % ARS (Acros organics, Sigma, MO, USA), pH 4.2 at room
temperature for 45 minutes. The staining was then washed with distilled water,
followed by 3 rinses of 5 minutes each until no ARS remaining was observed. The images
were taken by a Zeiss Stemi 2000-C stereoscopic microscope (Zeiss, Germany) and a
Canon PowerShot A80 camera (Canon, Tokyo, Japan). Afterwards, for quantification, the
Alizarin Red stained calcium-containing deposits were dissolved in acetic acid (Sigma,
MO, USA) and the absorbance of solubilized ARS was measured at 405 nm using a
Synergy HT microplate reader (BioTek, VT, USA).
Statistical analysis
Statistical analyses were carried out using the IBM SPSS Statistical software (v.
26.0), calculating the mean and standard error for each condition. The data from the
different experimental conditions were compared using a Mann Whitney test or ANOVA
followed by the Bonferroni or Games-Howell test depending on the homogeneity of the
variances. The confidence intervals were fixed at 95 % (p < 0.05), 99 % (p < 0.01) and
99.9 % (p < 0.001).
47
Hypothesis
Hypothesis
In the last decades, dental implants and scaffolds for bone tissue engineering and
dental implantology have improved a lot. New materials, surface topography and
roughness changes showed to have a direct impact on mesenchymal stem cell
differentiation to osteoblasts. With all these new scaffolds that had shown a good
induction of osteoblastic differentiation potential due to upgrades in porosity,
roughness or shape, the best option for clinical use is still unclear. Thanks to their high
proliferation and differentiation potential, MSCs seemed to be the future of bone tissue
engineering; however, the debate about the best MSCs for bone regeneration therapies
is still open. Finally, tissue engineering is facing the challenge of a widespread use of
animal-derived sera like fetal bovine serum (FBS), which must be replaced in order to
inactivate the immune response of the cell grafted patients.
With the aim to create a substitute for crucial tissue as periodontal ligament to
avoid implant loose due to periimplantitis, we hypothesized that biomimetic BAS TM
titanium surfaces and Decellularized Adipose Tissue (DAT) would provide a more
effective implant material and scaffold for stem cell osteogenic differentiation for bone
tissue engineering. Furthermore, the easily extracted, highly proliferative and
osteodifferentiable human dental pulp stem cells (hDPSCs) would be a very good option
for autologous regenerative cell therapies. Lastly, plasma-derived products like Plasma
rich in growth factors (PRGF) and Platelet rich fibrin (PRF) could provide an excellent FBS
substitute for in vitro cell cultures. Since it is possible to obtain these plasma products
from the patients themselves, this would avoid non-desirable immune responses on
regenerative cell therapies.
51
General Objectives
General Objectives
The main objective of this work was the study of hDPSCs osteoblastic
differentiation potential and interactions with biomaterials for bone tissue engineering.
To this end, we assessed the effect of Ti6AL4V and BASTM titanium surfaces and porcine
Decellularized Adipose Tissue (pDAT), in combination with plasma-derived products and
osteoblastic differentiation media.
Specific objectives
55
Results
Results
Human dental pulp stem cells (hDPSCs) were isolated from third molars and
cultured in DMEM supplemented with fetal bovine serum (10 %), penicillin/
streptomycin (1 %) and L-Glutamine (1 %). After 2-3 weeks, the cells formed multiple
adherent colonies reaching subconfluence. On culture, the hDPSCs showed spindle-like
fibroblast shape morphology; the cells maintained this feature as well as being highly
proliferative for several passages (Figure 12).
A B
C D
Figure 12. Culture of hDPSCs in vitro. Phase-contrast microscopy images for adherent hDPSCs in culture
under subconfluence (A, C) and confluence (B, D) at low and high magnification. Adapted from (Irastorza
et al., 2019).
For the evaluation of the cellular markers of the hDPSCs, we analyzed the cell
markers by flow cytometry using antibodies against the following CD (clusters of
differentiation) antigens: CD45, CD73, CD90 and CD105. The hDPSCs were negative for
CD45, the hematopoietic marker. On the other hand, they were positive for
mesenchymal stem cell markers CD73 (100 %), CD90 (100 %) and CD105 (100 %). With
these results, we can conclude the mesenchymal stem cell phenotype of the dental pulp
stem cells on culture (Gronthos et al., 2003) (Figure 13).
59
Igor Irastorza Epelde
CD 45 A B CD 73
3.0% 100.0%
CD 90 C D CD 105
100.0% 99.9%
Figure 13. Stem cell markers of the hDPSCs by flow cytometry. Flow cytometry revealed that hDPSCs
were all negative for the hematopoietic marker CD45 (A) and all positive for the mesenchymal stem cell
markers CD73, CD90 and CD105 (B, C, D, respectively). Adapted from (Irastorza et al., 2019).
60
Results
A B
Ti6AL4V
C D
BAS®
Figure 14. SEM images of Ti6AL4V and BAS titanium surfaces. Scanning electron microscopy images of
titanium surfaces, Ti6AL4V (A, B) and BAS (C, D) with small and high magnifications. Adapted from
(Irastorza et al., 2019).
Cell adhesion, migration and viability of hDPSCs on Ti6AL4V and BAS™ titanium
surfaces
hDPSCs were seeded on Ti6A14V and BAS™ titanium surfaces, 20,000 cells/disc,
for 4 days. The hDPSCs strongly adhered and spread all over the smooth titanium surface
Ti6AL4V. The cells grew following the orientation of the disc´s groove pattern and
showed several cell elongations similar to lamellipodia (Figure 15 A). On the other hand,
the hDPSCs seeded on BAS titanium surface adhered by adapting their morphology to
the porous surface, penetrating deeper into the porous structure (Figure 15 B). In both
titanium surfaces the cell adhesion showed to be consistent and maintained throughout
the entire culture time.
We performed a cell viability and death assay of the hDPSCs seeded on both
titanium surfaces by culturing them in the presence of Calcein-AM (green fluorescence)
and propidium iodide (red fluorescence). The fluorescence images showed that the
hDPSCs were 100 % calcein positive on both titanium discs, demonstrating the high
61
Igor Irastorza Epelde
viability of the hDPSCs grown on the smooth and rough titanium surfaces (Figure 15C,
D). Moreover, none of the hDPSCs showed PI red fluorescence, demonstrating that cell
death was negligible in these cultures.
In addition, where the nuclei of the hDPSCs had been stained in blue by DAPI,
revealed that the cells followed the groove directions on the Ti6A14V surface orientating
themselves in a spiral form (Figure 15 E), whereas the hDPSCs had no preferential
orientation while been seeded in the BAS titanium (Figure 15 F).
A C E
Ti6AL4V
B D F
BAS®
Figure 15. SEM images, viability, orientation and mobility of hDPSCs on titanium surfaces. Scanning
electron microscopy images of both titanium surfaces with hDPSC cells seeded on them (A, B,
respectively). Fluorescent microscopy images of the hDPSCs cultured for 4 days on both titanium discs to
test viability by calcein (green) and cell death by propidium iodide (red) (C, D). Scale bar: 100 µm.
Fluorescent microscopy image mosaic showing the entire titanium discs with hDPSCs nuclei stained in
blue with DAPI on Ti6A14V (E) and on BAS (F). Scale bar: 2 mm. Adapted from (Irastorza et al., 2019).
Cell proliferation assay of hDPSCs combined with PRGF grown on Ti6AL4V and BAS TM
titanium surfaces
62
Results
Control PRGF ®
A B
Ti6AL4V
Ki67/ DAPI
C D
BAS®
1,5 1,5
1 1
0,5 0,5
0 0
Control Ti/PRGF Ti Control BAS/PRGF BAS Control Ti/PRGF Ti Control BAS/PRGF BAS
63
Igor Irastorza Epelde
Figure 16. Proliferation assay of the hDPSCs cultured on presence of PRGF on titanium surfaces.
Immunofluorescence images of hDPSCs for Ki67 proliferation marker (green) and the nuclei stained with
DAPI (blue) in control conditions on Ti6AL4V (A) and BAS (C) titanium discs, and cultured in presence of
20 % PRGF on Ti6AL4V (B) and BAS (D) discs for 4 days. PRGF treatment increased the number of Ki67+
cells. Scale bar: 50 µm. Graphs illustrating comparison of total amount of cycling cells (KI67+) between
control (normalized number of cells) and PRGF conditions (E) and total number of nuclei (DAPI; F) of
hDPSCs grown on both titanium surfaces. Statistical significance was set at p ≤ 0.05. Adapted from
(Irastorza et al., 2019).
64
Results
10
B 9
8
7
Normalized ALP
absorbance
6
5
4
*
3
2
1
0
CL GL PL CDL GDL PDL CR GR PR CDR GDR PDR
Ti6AL4V BAS
65
Igor Irastorza Epelde
Figure 17. Alkaline phosphatase assay of hDPSCs cultured on both titanium surfaces, in
presence/absence of osteoblastic differentiation media and plasma derived products (PRGF and PRF).
Images of alkaline phosphatase staining of the hDPSCs cultured on Ti6AL4V and BAS titanium discs for 14
days with osteoblastic differentiation media, PRF and PRGF (A). Graph illustrating the normalized
quantification of the ALP activity (B). Graph acronym meaning: C (control), G (PRGF), P (PRF), D
(differentiation medium), L (Ti6AL4V titanium) and R (BAS titanium). Statistical significance was set at (*)
p ≤ 0.05. Scale bar: 1 mm. Adapted from (Irastorza et al., 2019).
Alizarin red staining of hDPSCs grown on Ti6AL4V and BASTM titanium surfaces on
terminal osteogenic differentiation
The main evidence of terminal osteogenic differentiation of the stem cells is the
formation of calcified bone matrix nodules. This staining was performed by Alizarin red,
which stains the calcified extracellular bone matrix nodules in red. The hDPSCs were
seeded on plastic, Ti6AL4V and BAS titanium surfaces, grown with the osteoblastic
differentiation media for 14 days before alizarin red staining. Images taken by the
stereoscopic microscope demonstrated that there was no alizarin red staining on the
hDPSCs cultured on plastic in control condition. On the contrary, the hDPSCs grown on
both titanium surfaces, showed alizarin stained red spots corresponding to calcified
bone matrix depositions. These bone matrix depositions could be identified at the
macroscopic level and even in control conditions, being more consistent on the BAS
titanium surface (Figure 18C-J). Therefore, as assessed by alizarin red staining, the effect
of titanium surfaces demonstrated by itself to be enough to induce the hDPSCs
osteoblastic differentiation. Comparing to controls, the effect of the pharmacological
osteoblastic induction media seemed not to produce an additional increase of the total
number of stained matrix depositions spots when the hDPSCs were grown on the
titanium discs. The calcium depositions of the HDPSCs treated with the osteoblastic
induction cocktail give the impression of being more intensely stained with alizarin red,
particularly on the cells seeded on BAS titanium (Figure 18. I, J). The biggest evidence of
the effect of osteoblastic differentiation induction media could be found on the hDPSCs
cultured on plastic. On this condition, the differentiation cocktail composed by β-
glycerophosphate, dexamethasone and ascorbic acid, lead to a high number of
66
Results
mineralized bone matrix depositions with respect to the control conditions (Figure 18A,
B).
Control Differentiation
A B
C D
Ti6AL4V
E F
G H
BAS®
I J
67
Igor Irastorza Epelde
Figure 18. Alizarin red staining assay of hDPSCs grown on Ti6AL4V and BAS titanium surfaces. Alizarin
red staining of deposited bone matrix spots in control conditions on plastic (A), Ti6AL4V (C, E) and BAS (H,
E) in comparison with the cells treated with osteoblastic differentiation media for 14 days on plastic (B),
Ti6AL4V (D, F) and BAS (I, J). Scale bar (A, B): 100 µm; scale bar (C, D, G, and H): 2mm; scale bar (E, F, I and
J): 0.5 mm. Adapted from (Irastorza et al., 2019).
68
Results
2,5 B
Normalized ARS absorbance *
2
1,5
*
0,5
0
CR GR PR CDR GDR PDR
Figure 19. Alizarin red staining and photometric quantification of hDPSCs grown in presence of PRGF
and PRF on BAS titanium surface. Alizarin red staining of hDPSCs cultures on BAS titanium surface in
control and osteoblastic differentiation conditions and in presence or absence of PRGF and PRF for 21
days (A). Normalized photometric quantification of Alizarin red staining (B). Graph acronym meaning: C
(control), G (PRGF), P (PRF) and D (differentiation medium). Statistical significance was set at (*) p ≤ 0.05.
Scale bar: 250 μm. Adapted from (Irastorza et al., 2019).
The staining of the mineralized bone matrix deposits by alizarin red showed the
osteoblastic differentiation of the hDPSCs cultured in the presence of PRGF and PRF.
However, how the osteoblastic differentiation molecular signaling pathways were
affected by the plasma derived products remained unclear. To shed light to this
question, we chose representative gene markers involved on the different stages of the
osteoblastic differentiation. This battery of gene markers was composed by Collagen I
(main organic compound of the bone extracellular matrix), RUNX2 (immature osteoblast
marker), SPARC (osteonectin, intermediate secretory osteoblast marker) and
OSTERIX/SP7 (fully mature secretory osteoblast marker).
The RT-QPCR results showed that even the hDPSCs cultured without osteoblastic
differentiation media expressed Col-I, RUNX2, SPARC and OSTERIX when they were
69
Igor Irastorza Epelde
seeded on both titanium surfaces (Figure 20). Besides, most of the conditions where
the hDPSCs were grown in presence of PRGF or PRF, revealed an increase of the
expression of these markers. The expression of Collagen 1 between control and plasma
derived products containing conditions showed certain variability. Nevertheless, the
conditions where the hDPSCs were cultured in presence of PRGF and PRF demonstrated
a remarkable increase of the expression of pre-osteoblastic markers RUNX2 and SPARC.
Between these two markers, we have to mention especially SPARC marker, whose
expression levels in the conditions containing PRGF and PRF raised 6-11 fold with respect
to the controls, either in presence or absence of the osteoblastic differentiation
treatment (Figure 20 C). Even when the PRGF and PRF demonstrated having an
enhancing effect in the osteoblastic differentiation process of the hDPSCs as
demonstrated by the raise of expression of the pre-osteoblastic markers RUNX2 and
SPARC, we found other interesting differences between these two plasma-derived
products. On contrary to RUNX2 and SPARC expression increase can make us think, the
expression levels of fully mature osteoblast marker OSTERIX was only increased on the
conditions containing PRF. It is important to mention that the hDPSCs in control
conditions expresses Collagen 1, RUNX2 and SPARC, being naturally on an osteoblastic
pre-differentiated stage. It is noticeable the significant augment (10-30 fold) of OSTERIX
gene expression in all conditions containing PRF, either in presence or absence of the
osteoblastic differentiation media (Figure 20 D). Nevertheless, it is also remarkable that
the conditions where the hDPSCs were cultured in presence of PRGF did not have an
increase of the expression of this marker but the expression of OSTERIX was even lower
than in control conditions.
70
Results
Collagen I RunX2
A B
* *
3,5
mRNA Fold Change
SPARC Osterix
C D
** *
16 ** 35
mRNA Fold Change
Figure 20. QPCR of osteoblastic differentiation gene expression of hDPSCs cultured on Ti6AL4V and
BASTM titanium surfaces in presence or absence of PRGF and PRF. Normalized mRNA expression of
hDPSCs after 14 days of culture for Collagen I, RUNX2, SPARC and OSTERIX. Statistical significance was set
at (*p ≤ 0.05) and (**p ≤ 0.01). CL: control Ti6aI4V; CR: control BAS; CDL: control/ Ti6aI4V/differentiation
treatment; CDR: control/BAS/differentiation treatment; GL: Ti6aI4V/PRGF; GR: BAS/PRGF; GDL: Ti6aI4V/
PRGF/differentiation treatment; GDR: BAS/PRGF/differentiation treatment; PL: TI6AI4V/PRF; PR:
BAS/PRF; PDL: TI6AI4V/PRF/differentiation treatment; PDR: BAS/PRF/differentiation treatment. Adapted
from (Irastorza et al., 2019).
71
Igor Irastorza Epelde
a widely used marker of cell death by apoptosis. To assess the differences in cellular
death between hDPSCs and hBMSCs, influenced by titanium surfaces, both types of cells
were seeded on Ti6AL4V and BAS titanium discs for 24 and 48 hours. After that period
we performed an immunofluorescence assay to detect apoptotic cells (caspase 3 +). The
results demonstrated that none of the two titanium surfaces had any cytotoxic effect on
hDPSCs and hBMSCs at 24 and 48 hours. Both types of cells showed having only one or
two apoptotic cell in the whole disc, being dying cell proportion less than 0.1 %. The cell
nuclei were stained in blue with DAPI (Figure 21).
48H
BMSC
DPSC
Figure 21. Immunofluorescence images of cell death marker Caspase 3 in hDPSCs and hBMSCs cultured
for 24 and 48 hours on coverslips, Ti6AL4V and BAS titanium surfaces. Immunofluorescence images of
hDPSCs and hBMSCs stained with apoptotic cell marker Caspase 3 (green) and nuclei stained with DAPI
72
Results
(blue) seeded on coverslips, Ti6AL4V and BAS titanium discs for 24 and 48 hours. The images
demonstrated that both titanium surfaces had no cytotoxic effect on hDPSCs and hBMSCs finding only
one or two positive cells for Caspase 3 in the whole disc. Scale bar: 100 µm.
73
Igor Irastorza Epelde
A
BMSC Coverslips Ti6AL4V BAS
24H
DPSC
BMSC
48H
DPSC
B Ki67/DAPI Ki67/DAPI
* 24H 48H *
2,5 1,4
Normalized Ki67/DAPI
1,2
Normalized Ki67/DAPI
2 *
1
1,5 0,8
1 0,6
0,4
0,5
0,2
0 0
Coverslips Ti6AL4V BAS Coverslips Ti6AL4V BAS
BMSC DPSC
74
Results
Figure 22. Proliferation assay between hDPSCs and hBMSCs in coverslips, Ti6AL4V and BAS titanium
surfaces at 24 and 48 hour of culture. Images taken by immunofluorescence microscope of hDPSCs and
hBMSCs stained with proliferation marker Ki67 (green) and the nuclei counterstained with DAPI (blue) at
24 and 48 hours of culture (A). Graphs showing the proliferation rate difference between both types of
cells while being cultured on different surfaces (B). Scale bar: 100 µm. Statistical significance was set at
(*p ≤ 0.05).
75
Igor Irastorza Epelde
Control Differentiation
Ti6AL4V
BMSC
BAS
Ti6AL4V
DPSC
BAS
Figure 23. Immunofluorescence images of osteoblastic differentiation marker SPARC in hDPSCs and
hBMSCs seeded on Ti6AL4V and BAS titanium discs and grown in presence or absence of osteoblastic
differentiation media. hDPSCs and hBMSCs were cultured on both titanium discs and grown with control
and osteoblastic induction media for 14 days. After fixation, they were stained for osteoblastic
differentiation marker SPARC (green) and counterstained with DAPI (nuclei). Scale bar: 100 µm.
76
Results
Control Differentiation
Ti6AL4V
BMSC
BAS
Ti6AL4V
DPSC
BAS
Figure 24. Immunofluorescence images of osteoblastic differentiation marker Osterix in hDPSCs and
hBMSCs seeded on Ti6AL4V and BAS titanium discs and grown in presence or absence of osteoblastic
differentiation media. hDPSCs and hBMSCs were cultured on both titanium discs and grown with control
and osteoblastic induction media for 14 days. After fixation, they were stained for osteoblastic
differentiation marker Osterix (green) and counterstained with DAPI (nuclei). Scale bar: 100 µm.
77
Igor Irastorza Epelde
3,5
2,5 BMSC
2 DPSC
1,5
0,5
78
Results
Figure 25. Graph illustrating Alizarin red absorbance of hDPSCs and hBMSCs cultured on plastic, Ti6AL4V
and BAS titanium surfaces grown in presence or absence of osteoblastic differentiation media. After
culturing the cells for 21 days and dying the extracellular bone matrix deposits, the Alizarin red was
dissolved in Acetic acid. A microplate reader measured the absorbance.
RNA sequencing of hDPSCs and hBMSCs cultured on plastic, Ti6AL4V and BAS TM
titanium surfaces in presence or absence of osteoblastic differentiation media
79
Igor Irastorza Epelde
Table 2. Table illustrating the quantity of genes expressed in one condition and not in the other.
Different quantity of genes expression patterns were found when comparing different conditions.
Abbreviations: DPSC: dental pulp stem cells; BMSC: bone marrow stem cells; C: control media; T:
osteoblastic differentiation media; Flask: plastic culture surface; TI: Ti6AL4V titanium and BAS: Biomimetic
Advanced Surface titanium.
80
Results
81
Igor Irastorza Epelde
82
Results
transcription at 6
hours after
transduction UP
TAKEDA_NUP8_HO 118 4 0.00178462 HOXA3, HOXA5, effects of NUP98-
XA9_8D_UP HOXA7, HOXB3 HOXA9 on gene
transcription at 8
days after
transduction UP
TAKEDA_NUP8_HO 147 4 0.00369089 HOXA3, HOXA5, effects of NUP98-
XA9_10D_UP HOXA7, HOXB3 HOXA9 on gene
transcription at 10
days after
transduction UP
DPSC-C- BASSO_GERMINAL 93 3 0.0191607 BATF, BCL2A1, Gene up-regulated
FLASK _CENTER_CD40_UP HLA-DQB1 by CD40 signaling
VS in Ramos cells
DPSC-T-
FLASK
CMV_HCMV_TIME 25 2 0.0377068 RSAD2, TNFSF10 Genes up-regulated
COURSE_12HRS_U after infection with
P HCMV at 12 h
VERHAAK_AML_NP 173 3 0.040305 BCL2A1, Genes up-regulated
M1_MUT_VS_WT_ SERPINA1, in acute myeloid
UP TNFSF10 leukemia (AML)
CARIES_PULP_UP 200 3 0.0462674 BCL2A1, HLA- Immune/cytokine
DQB1, SERPINA1 response
Table 4. Pathway enrichment analysis performed with The Connectivity map (CMAP). Pathway
enrichment comparative analysis between hDPSCs and hBMSCs grown on plastic and titanium surfaces,
in the presence or absence of osteoblastic differentiation media. The absent comparisons are not in the
table because they did not meet the established n= 2 and p< 0.05 requirements. Abbreviations: DPSC:
dental pulp stem cells; BMSC: bone marrow stem cells; C: control media; T: osteoblastic differentiation
media; Flask: plastic culture surface; TI: Ti6AL4V titanium and BAS: Biomimetic Advanced Surface
titanium.
83
Igor Irastorza Epelde
84
Results
SEM images of decellularized porcine adipose tissue (pDAT) processed as solid foam
The porcine adipose tissue was extracted and the decellularization was
performed using isopropanol and Triton X-100, a non-ionic detergent. Once the
decellularization process concluded, in order to be used as a biological scaffold for cell
cultures in vitro, the solid foam was formed by freeze-drying method (Figure 26A, B).
SEM images demonstrated the highly interconnected porous structure. The pore size
varied between 50 and 100 µm (Figure 26C, D).
85
Igor Irastorza Epelde
A B
C D
Figure 26. Images of decellularized porcine adipose tissue for in vitro use and porous structure details
by SEM. Macroscopic images of decellularized porcine adipose tissue for in vitro use in combination with
cells cultures (A, B). SEM images of the porous structure of pDAT showing 50-100 µm prous size (C, D).
To exclude the cytotoxic effect of pDAT, 15.000 hDPSCs cells were seeded on
both empty well (plastic) and pDAT solid foams for 4 days. Cell viability assay was
performed culturing the hDPSCs in presence of live cell dyer Calcein-AM (green
fluorescence) and death cell dyer propidium iodide (red fluorescence). Fluorescence
microscope images demonstrated that the hDPSCs grown on both surfaces had almost
100% live cells, with all the cells dyed with green fluorescence. Only sporadically were
cells found dyed in red with propidium iodide showing the lack of cytotoxicity of pDAT
(Figure 27). The main difference we found between this two surfaces was the higher
amount of cells found in control plastic (Figure 27A, B). Higher amount of hDPSCs that
apparently we had in plastic could be due to monolayer growing they have in plastic in
comparison to the 3 dimensional pDAT surface.
86
Results
Plastic pDAT
A C
B D
Figure 27. Calcein-AM/ propidium iodire images of hDPSCs cultured on plastic and pDAT solid foams.
HDPSCs were seeded at 15.000 cell/well for 4 days and stained with live cell marker Calcein-AM (green)
and death cell marker propidium iodide (red). The cells grown on plastic (A, B) showed higher amount of
hDPSCs than pDAT (C, D) due to monolayer growing. Scale bar (A, C): 100 µm; (B, D): 50 µm.
hDPSCs culture on solid foam pDAT and bone-producing osteoblastic cell markers
immunofluorescence.
pDAT solid foam was integrated to 8-well Minicell EZ slides (Merck Millipore
PEZGS0816) for in vitro use in cell culture. The hDPSCs were seeded al 15.000 cell/ well
density and grown for 14 days in presence of control media (DMEM) and osteoblastic
differentiation media. Cell nuclei stained with DAPI showed less number of cells on pDAT
porous structure than in plastic wells were the hDPSCs had grown as an adherent
monolayer (Figure 28A). After the 14 days of culture, immunofluorescence assay was
performed for the detection of osteoblastic differentiation markers Osteocalcin (BGLAP)
and Osteonectin (SPARC). hDPSCs seeded in plastic and pDAT expressed this two
87
Igor Irastorza Epelde
500
B
% normalized IF intensity
BGLAP ***
400
300
A Control Differentiation
200
Plastic
100
0
DMEM DMEM
control pDAT
C
% normalized IF intensity
500
***
400
pDAT
300
200
100
0
Osteo Osteo
Control pDAT
Figure 28. Osteocalcin (BGLAP) immunofluorescence images of hDPSCs cultured in plastic and pDAT in
presence or absence of osteoblastic differentiation media and intensity quantification.
Immunofluorescence images of hDPSCs seeded on plastic or pDAT-containing EZ-slide wells and cultured
with control and osteoblastic induction media for 2 weeks (A). Quantification of relative BGALP IF labeling
respect to the cell number in different conditions by ImageJ (B, C). Scale bar: 50 µm. Statistical significance
was set at (***p ≤ 0.001).
88
Results
% normalized IF intensity
300
SPARC 250
200
A Control Differentiation
150
100
Plastic
50
0
DMEM DMEM
C Control pDAT
% normalized IF intensity
350
***
300
pDAT
250
200
150
100
50
0
Osteo Osteo
Control pDAT
Figure 29. Osteonectin (SPARC) immunofluorescence images of hDPSCs cultured in plastic and pDAT in
presence or absence of osteoblastic differentiation media and intensity quantification.
Immunofluorescence images of hDPSCs seeded on plastic or pDAT-containing EZ-slide wells and cultured
with control and osteoblastic induction media for 2 weeks (A). Quantification of relative SPARC IF labeling
respect to the cell number in different conditions by ImageJ (B, C). Scale bar: 50 µm. Statistical significance
was set at (***p ≤ 0.001).
ALP staining and quantification of hDPSCs cultured on plastic and pDAT in presence or
absence of osteoblastic differentiation media.
hDPSCs were seeded on plastic and pDAT for 14 days with control and
osteoblastic differentiation media. After that period, the cells were dyed for alkaline
phosphatase enzyme, mineralizing bone cells characteristic enzyme. The images showed
89
Igor Irastorza Epelde
the hDPSCs had ALP enzymatic activity in both surfaces (Figure 30A). The quantification
of ALP demonstrated being significantly higher in the cells grown in presence of
osteoblastic differentiation media both in plastic and pDAT (Figure 30B, C).
Control Differentiation B **
A 300
50
0
Plastic
C 1000 **
400
200
0
Osteo
DMEM
pDAT
Figure 30. ALP activity quantification of hDPSCs cultured on plastic and pDAT and grown on presence or
absence of osteoblastic differentiation media. The hDPSCs were cultured for 14 days on empty well
(plastic) or pDAT-containing EZ-slide wells with control or osteoblastic differentiation media. ALP activity
was detected by the purple/black precipitate detection (A). Quantification of ALP activity was performed
measuring the relative ALP absorbance with ImageJ. Statistical significance was set at (**p ≤ 0.01).
Alizarin red staining and quantification of hDPSCs seeded on plastic and pDAT solid
foam grown in presence or absence of osteoblastic differentiation media.
90
Results
(Figure 31A). Nevertheless, the highest Alizarin red precipitate density was found in the
hDPSCs grown on pDAT (Figure 31A). Semi-quantitative measurement indicated that in
both surfaces the osteoblastic differentiation media caused more intense staining
(Figure 31B). Moreover, we used bright field images combined with
immunofluorescence staining for DAPI in order to observe the relative cellular location
respect to bone matrix deposits. The hDPSCs appeared to be more densely located
within the borders of mineralized areas. Moreover, the few hDPSC cells found in close
contact with calcified nodules had healthy-looking cell nuclei (Figure 31A).
B 300 200 *
250
150
200
150 100
100
50
50
0 0
DMEM Osteo DMEM Osteo
Plastic pDAT
91
Igor Irastorza Epelde
Figure 31. Alizarin red-DAPI double staining of hDPSCs cultured on plastic and pDAT solid foams in
presence or absence of osteoblastic differentiation media. hDPSCs were seeded on empty wells (plastic)
or pDAT solid foams with control media and osteoblastic induction media for 4 weeks before performing
Alizarin red staining. The cells were counterstained with DAPI. The bright field images and
immunofluorescence images were merged to study the hDPSCs location in mineralized bone matrix
nodules areas (A). Quantification of relative Alizarin red absorbance after background subtraction (B).
Scale bar: 50 µm. Statistical significance was set at (*p ≤ 0.05).
TEM images of hDPSCs cultured in pDAT scaffolds crating large Sharpey-like collagen
fiber bundles
With the aim of studying in detail the bone-like ECM ultra-structure the hDPSCs
cultured in pDAT solid foams created, TEM images were taken. Small collagen fibers
spread in large areas in the pDAT matrix scaffold were observed in control conditions
beside some interspersed lipid droplets coming from the remains of decellularized
adipose tissue. Interestingly, when hDPSCs were cultured on pDAT in presence of
osteoblastic differentiation media, sharp transitions between thin collagen areas and
thick and mineralized collagen areas were also identified (Figure 32; yellow dashed
lines). This thick collagen fiber bundles had higher electron density and they were
connected to the rest of the pDAT solid foam, which was composed by thinner and less
calcified collagen fibers. In the electrodense collagen areas within the solid foam,
intramembranous ossification sites were also found (arrowheads in figure 33A, B).
Mineralized bone matrix was also observed anchored to the rest of the collagen fibers
in electrodense areas by Sharpey-like fibers (asterisk in Figure 33B). This electrodense
collagen areas and intramembranous ossification sites were not observed in solid foams
without cell seeding (data not shown), indicating the de novo generation of these
structures by hDPSCs.
92
Results
C D
E F
Figure 32. TEM images of the ultrastructure features of hDPSC cultured in pDAT solid foams. hDPSCs
were cultured for 4 weeks with control media (left column) and osteoblastic differentiation media (right
column). It coul be observed the transition area between hDPSCs produced bone ECM with thicker and
more calcified collagen fibers and thinner collagen fibers of pDAT solid foam matrix (yellow dashed line).
Higher amount of electrodense collagen fibers were found in osteoblastic differentiation conditions
(bottom high-magnification images). Scale bars: 1 µm (A, C and D); 500 nm (B); 200 nm (E and F).
93
Igor Irastorza Epelde
B
* *
*
*
* *
Figure 33. TEM images of intramembranous ossification sites and Sharpey-like fibers generated by
hDPSCs cultured in the pDAT solid foams. Transition between non-calcified collagen areas and calcified
thick collagen fiber bundle areas of hDPSCs seeded in pDAT solid foams (dashed line). Intramembranous
ossification sites were found on this thick collagen containing electrodense structures. Magnification of
arrowheads marked areas in the right panels (A). Presence of perforating Sharpey-like fibers on the edges
of intramembranous ossification sites (asterisk in right panels), showing anchoring to the rest of the
collagen matrix. Magnification of arrowheads marked areas in the right panels (B). Scale bars: 2 µm (top
left image); 1 µm (bottom left image); 500 nm (top right images); 200 nm (bottom right images).
94
Discussion
Discussion
One of the aims of this study was the evaluation of the hDPSCs combined with
autologous plasma components on biomimetic titanium dental implant materials for
bone regeneration in vitro. Within this framework, this study was focused on testing the
possible enhancements in the osteoblastic differentiation process by assessing the
deposition of secreted bone-matrix of hDPSCs cultured on standard Ti6AL4V and
biomimetic BASTM (Avinent implant system) titanium surfaces in combination of plasma-
derived products PRGF and PRF. After Gronthos et al. isolated hDPSCs for the first time
(Gronthos et al., 2000), a lot of different laboratories have confirmed the capability of
those neural crest derived stem cells to differentiate into different cell lineages of
different germ layers (Nuti et al., 2016). These differentiations include bone-producing
osteoblastic cell differentiation being one of the most interesting in the field of
implantology (Giuliani et al., 2013; Tatullo, 2017).
97
Igor Irastorza Epelde
98
Discussion
hDPSCs in vitro expansion and maintenance were also tested for both Ti6AL4V
(smooth) and BAS (rough) titanium surfaces. Both surfaces preserved cell viability,
allowing a good cell proliferation. This was assessed by the detection of Ki67 positive
cells in basal conditions. The hDPSCs cultured on Ti6AL4V titanium surface showed
higher cell mobility compared to BAS titanium due to the flat surface. Despite both
plasma derived products allowed a good hDPSC growth, cell proliferation was
significantly enhanced by the addition of 20 % PRGF to the culture media, but not by
insoluble PRF. This difference may be caused by the different application method of both
plasma products. PRF membranes were put into the culture wells and maintained over
the duration of the experiment, releasing molecules gradually. On the contrary, the
soluble plasma fraction PRGF was added to the culture media at 20 % concentration and
renewed with each media change every 2-3 days. It is possible to hypothesize that high
concentration of growth factors induced hDPSCs the high concentration of growth
factors, caused by the addition of soluble PRGF to the culture medium, would induce
the proliferation of hDPSCs. This would have a negative effect over mature osteoblastic
differentiation, as both processes (cell proliferation vs differentiation) are antagonist to
each other. In hDPSCs cultured with PRGF, osteoblastic pre-commitment marker
expression RUNX2 and SPARC mRNA levels were actually increased. But somehow these
hDPSCs failed to differentiate into mature osteoblast cells, as seen by a lower expression
of OSTERIX and a decreased amount of secreted bone matrix detected by Alizarin red.
In contrast, hDPSC cultures supplemented with PRF membranes would arguably
generate conditions where by a slow gradual growth factor release from the fibrin clot
would favor the mature osteoblast differentiation of hDPSCs. It is noteworthy that the
combination of plasma rich fibrin with biomimetic BAS titanium surface was the most
effective condition to induce in vitro osteoblastic differentiation of hDPSCs grown on
titanium surfaces. This is where BAS rough titanium surface demonstrated being a better
option with respect to the smooth Ti6AL4V titanium, as OSTERIX expression levels could
be substantially enhanced when we combined BAS titanium with PRF.
99
Igor Irastorza Epelde
To achieve a full translation of this methodology to the dental clinic, more in vivo
studies will be necessary. In spite of not being the most abundant source of
mesenchymal stem cells in the human adult body, for the fields of implantology and
craniomaxilofacial surgery, hDPSCs are particularly interesting cells. For oral and
maxillofacial trauma patient who lose several teeth in an accidental episode, this holds
especially true. They might very well benefit from an approach whereby clinicians took
advantage of the lost dental pieces to extract autologous hDPSCs to aid in the
reconstruction of the completely affected bone area. Whatever the case, when cultured
on microporous biomimetic titanium surfaces such as BASTM, hDPSCs respond with
efficacy to osteoblastic differentiation protocols, especially in combination with platelet
rich fibrin clot.
100
Discussion
Figure 34. Summary of plasma derived product effects on hDPSCs osteoblastic differentiation processes
on culture. Summary of the theoretical model of the different effects caused by the supplementation with
pharmacological differentiation reagents, growth substrates (Ti6AL4V and BAS) and plasma derived
products (PRGF and PRF) over the osteoblastic differentiation process. Mesenchymal stem cells grows in
vitro in an undifferentiated state. They can be induced to commit osteoblastic differentiation where in
the first stages cells will enhance the expression of the transcription factor RUNX2, as they will express
progressively higher amounts of Collagen I and calcium-binding proteins like SPARC. In the final stage of
osteoblastic differentiation, the cells acquire the ability to mineralize the extracellular matrix, forming the
mature bone tissue. At the cellular/molecular level, this is characterized by the rise of the expression of
OSTERIX, the mature osteoblast/osteocyte gene marker. Pharmacological reagents, plasma-derived PRGF
and PRF as well as titanium surfaces appear all to influence the preosteoblast transition of hDPSCs. This is
confirmed by the detection of higher levels of RUNX2 and SPARC expression and/or phosphatase alkaline
staining found in all these conditions. However, hDPSCs cultured with soluble PRGF did not achieve
mature osteoblast stage, possibly due to proliferation/differentiation signaling conflict. On contrary,
hDPSCs seeded in the presence of PRF clots showed the highest expression levels of osteoblastic
differentiation, especially on BAS biomimetic titanium surface combined with osteoblastic induction
reagents.
The previous study showed the good cell viability, proliferation and osteoblastic
differentiation features of hDPSCs grown over titanium surfaces. Mesenchymal stem
cells had demonstrated being a great option for their use in bone tissue engineering due
to their capacity to adhere, proliferate and differentiate to different cell types when
seeded on different biomaterials. However, with all the different mesenchymal stem cell
sources found in the human adult body it is still unclear which one is the best option for
the use on tissue engineering. For the effective development of bone and dental
regeneration therapies, different types of post-natal stem cells need to be compared to
determine the best MSC option. To bring light to this question, we next performed a
comparative study between hDSPCs and hBMSCs, a promising MSC type due to its
natural bone differentiation ability.
The aim of this comparative study was to compare the viability, proliferation and
osteoblastic differentiation between hDPSCs and hBMSCs cultured on Ti6AL4V and
BASTM titanium surfaces in the presence or absence of osteoblastic differentiation
media. For the evaluation of these results, we have to take into account the patients age
101
Igor Irastorza Epelde
difference between hDPSC and hBMSC donors for the evaluation of the results, which
was 16-30 years for hDPSCs and 45 years for BMSCs.
Both hDPSCs and hBMSCs seeded on Ti6AL4V and BAS titanium discs showed no
cytotoxic effect from the titanium surfaces. These two types of cells showed almost 100
% viability while being seeded on titanium surfaces where only isolated single cells were
found stained by propidium iodide. On the contrary, the cell proliferation assay did
exhibit significant differences between hDPSCs and hBMSCs. The comparative cell
proliferation experiment showed a greater growth potential of hDPSCs compared to
hBMSCs when seeded on both titanium surfaces after 24 and 48 hours. When the cells
were cultured on coverslips this difference was only observed after the initial 24 hours
of culture while at 48 hours both cells had similar proliferative activity. These results
confirmed what was shown in other articles where the cells had been seeded in different
biomaterials (Amid et al., 2021; Ponnaiyan and Jegadeesan, 2014).
Apart from this, the RNA sequencing was analyzed following expression/ lack of
expression criteria. To achieve this aim, we performed a comparative RNA expression
analysis between pairs of different conditions to find out the genes that were expressed
in one condition and not in the other. We chose a presence/absence (ON/OFF)
comparison to detect specific sets of genes, whose expression would be specifically
triggered in osteogenic differentiation enhancement conditions, to potentially identify
new gene targets and signaling pathways involved in the osteoblastic differentiation of
hDPSCs and hBMSCs.
102
Discussion
The main difference in terms of genes expression between hDPSCs and hBMSCs
grown in plastic with control media was the HOX gene family expressed by hBMSCs. HOX
genes are responsible for embryonic MSC proliferation and for the regional
specifications and related tissue-specificity (Ackema and Charité, 2008). Previous studies
had demonstrated the differences in the HOX expression pattern in mandibular neural
crest cells and adult mandibles, in contrast to distal mesoderm-derived mesenchyme
and bones (Dong et al., 2014; Lee et al., 2015b; Wehrhan et al., 2011). It have been
suggested that this gene family could be related to the site-specificity of MSCs,
depending on the anatomical position (Wang et al., 2009). These HOX genes expression
difference could explain the different cellular behavior associated to autogenous cell
grafts of different embryonic origins (Leucht et al., 2008). The results obtained on this
study are consistent with previous studies which demonstrated the HOX gene
expression decrease in mandibles compared to that of long bones (Lee et al., 2015b;
Leucht et al., 2008).
103
Igor Irastorza Epelde
Osx
Sp1
ZBTB16
Sp1
Figure 35. Osterix binds to the Sp1 sequence of the ZBTB16 promoter region.
Among other interesting differentially expressed genes, NTRK3 and GFRA2 genes
were expressed in hDPSCs seeded on plastic with control media but this expression
disappeared by the effect of the differentiation media and/or titanium surface.
Neurotrophin receptor genes, and particularly NTRK3, had been suggested to be hDPSCs
stemness markers, hence these results are consistent with previous studies (Luzuriaga
et al., 2019b).
104
Discussion
in our different culture conditions. We used the connectivity map (CMAP) and gene
ontology biological process (GOBP) to identify the most likely affected signaling
pathways in each experimental comparison. We selected those pathways that
presented at least two differentially expressed genes and a significant p value of <0.05.
These databases showed the different biological processes and diseases where these
genes are involved. Most of the outputs of the CMAP comparative analysis between
hBMSCs and hDPSCs were related to HOX genes, being related to diverse processes
ranging from acute myeloid leukemia to hematopoietic disorders. On the other hand,
GOBP related the expression of different clusters of genes to anterior/posterior pattern
specification, blood vessel development, cardiovascular system development,
regulation of blood vessel size, hormone transport and hematopoietic stem cell
differentiation. The significant differences detected on pathways related to
anterior/posterior pattern specification and hematopoietic stem cell differentiation can
be clearly related to the different embryonic origin and physiological function of hDPSCs
and hBMSCs.
As already mentioned, the dental implants have upgraded considerably over the
last years, improving implant survival rate. This survival achieved around 95 % after 10
years post-loading (Moraschini et al., 2015) and almost 88 % after 20 years post-loading
(Chrcanovic et al., 2018). However, with the increase of life expectancy and population
105
Igor Irastorza Epelde
ageing in developed countries, the demand for very long-term durable dental implants
seems likely to be maintained in the future.
Over the last decades, dental implant osseointegration has obtained big
improving results; however, some significant challenges remain. One of the biggest
problems in oral implantology is the Marginal Bone Loss (MLS), which is directly
correlated to implant loss due to the mechanical stress that the alveolar bone suffers
because of masticatory function in the absence of a functional PDL. The entire
masticatory load is transferred to alveolar bone tissue because dental implants are
directly anchored on it, which can lead to its resorption. There are many studies
associating a high MLB with a high risk of periimplantitis and implant failure (Chrcanovic
et al., 2018; Coli and Jemt, 2021; Galindo-Moreno et al., 2015). The most effective
strategy to reduce periimplantitis and MBL has been proposed to be the reconstruction
of PDL, which naturally acts like a cushion between alveolar bone and the dental piece
by absorbing mechanical forces. However, PDL regeneration and engineering
constitutes an extraordinary challenge (Lee et al., 2020). Any material used to substitute
PDL should firmly anchor to both alveolar bone and the dental implant while conserving
a highly vascularized tissue strip in between. In practice, this problem could only be
solved by seeding osteogenic and vasculogenic stem cells on biomaterials.
In the context of PDL regeneration by stem cell therapy, the best scenario would
be that the stem cells cultured on the biomaterial came from the patients themselves
(autologous graft), with which under adequate manipulation we could avoid immune
rejection issues. As previously mentioned, some of the most promising stem cell types
for dental implantology are hDPSCs. These cells are particularly well suited for
cryopreservation and autologous transplant (Ibarretxe et al., 2012b; Raik et al., 2020).
Another important characteristic of hDPSCs is the high capacity of differentiation to a
very large variety on both mesenchymal and non-mesenchymal cell types they have in
the absence of xenogenic cell culture compounds like animal serum. Interestingly, the
capacity of hDPSCs to differentiate to both vasculogenic and osteogenic cells has been
shown in serum free media. We recently published and patented an animal serum free
methodology to obtain vasculogenic cells (endothelia and pericytes) (Luzuriaga et al.,
106
Discussion
2020; Pineda Martí et al., 2020). This method could be potentially applied to hDPSCs
seeded on biomaterials to vascularize scaffolds as pDAT for their utilization in cell
therapies.
The results obtained in this investigation confirms that pDAT solid foams could
establish a very important biomaterial for PDL and dental bone regeneration. This
formulation of pDAT significantly enhanced secreted bone matrix by hDPSCs, as
evaluated by phosphatase alkaline and Alizarin red staining. The hDPSCs cultured on the
scaffold showed no loss of viability under experimental conditions. hDPSCs were seeded
at relatively low initial desities (15.000 cells/well; 21.428 cells/cm2) and also low volume
of pDAT scaffold (120 µl/well) to couple it for long-term cell culture in vivo in EZ-slides.
Maintaining higher cell densities over longer periods would possibly require better
nutrient and oxygen supply through a vascular network (Nakamura et al., 2019). Further
in vivo experiments will be necessary to test the potential of this material on the healing
of bone tissue and PDL lesions.
Finally, we also observed the production of thick calcified collagen fiber bundles
and intramembranous ossification sites of hDPSCs cultured on pDAT. This calcification
sties were attached to the pDAT scaffold by Sharpey´s fibres. All these results support
107
Igor Irastorza Epelde
that the combination of hDPSCs, or other mesenchymal stem cells, with pDAT is not only
a good osteoblastic differentiation induction ECM environment but could also help on
the graft and surrounding hard tissue attachment. In the case of bone healing, this
attachment has a particular interest, where physical anchoring of the scaffold to the
bone tissue is necessary. Transplantation experiments in vivo will be needed to
corroborate these findings, where apart of being a vehicle of stem cells, the scaffold
would act as a bioinductive ECM which in the best scenario would enhance the calcified
attachment to hard bone tissue and also bone mesenchymal stem cells recruitment and
activation of the host bone tissue.
108
Conclusions
Conclusions
In the last decades, different mesenchymal stem cells had proven their potential
to be used in bone tissue engineering. Their high proliferation and multi-linage
differentiation ability make these cells ideal candidates for regeneration therapies.
Moreover, the improvements on implants and scaffolds manufacturing give rise to
different scaffold upgrades in porosity, roughness or durability. These modifications are
focused on enhancing biocompatibility for stem cell growth and differentiation. Finally,
the use of plasma derived growth factors showed to be a perfect solution for in vitro
autologous stem cell proliferation and differentiation in bone tissue engineering
therapies, avoiding non-desirable immune responses because of the use of animal
derived serums in cell cultures.
The following conclusions have been drawn from the results obtained in this work:
1. hDPSCs showed non affected cell viability and proliferation when cultured on
Ti6AL4V and BAS titanium surfaces.
2. Both titanium surfaces had osteoinductive effect on hDPSCs without the need of
osteoblastic differentiation media.
3. The plasma derived product PRGF induced cell proliferation on in vitro hDPSC
cultures while PRF maximized osteoblastic cell differentiation of hDPSCs and
calcified bone matrix production.
4. The combination of BAS titanium surface with plasma derived PRF enhanced
differentiation of hDPSCs to bone-producing cells. These results provide strong
experimental support to the widespread use of plasma derived fibrin clots in
common clinical practice to enhance bone production around microporous
titanium implant surfaces.
5. The comparative study of hDPSCs and hBMSCs cultured on Ti6AL4V and BAS
titanium surfaces suggested a higher cell proliferation and mineralization of
hDPSCs compared to hBMSCs, but, more data are needed to make a firm
conclusion. Due to the easier and less invasive isolation, higher proliferation and
bone mineral depositions hDPSCs could be a better option than hBMSCs for bone
regeneration therapies.
111
Igor Irastorza Epelde
Taking into account the relatively abundant and available source of human-derived raw
adipose tissue material and that both DAT and hDPSCs can be isolated from human
donors; this seems a great opportunity for their combined use in personalized clinical
therapies for the healing of bone lesions and dental implantology.
112
Annex I
Patents
114
Annex I
(12) INTERNATIONAL APPLICATION PUBLISHED UNDER TUE PATENT COOPERATION TREATY (PCT)
DAZIOA, Science Park of the UPV/EHU, Sede HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KM, RN,
Building, 3rd floor, Barrio Sarriena, sin, 48940 KP, KR, KW, KZ, LA, LC, LK, LR, LS, W, LY, MA,
LEIOA (ES). LUZURIAGA GONZÁLEZ, Jon; Dep. MD, ME, MG, MK, MN, MW, NIX, MY, MZ, NA,
Bio. cel. Facultad de Medicina y Enfermería, NG, M, NO, NZ, 0M, PA, PE, PG, PH, PL, PT, QA,
UPV/EHU, 48940 LEIOA (ES). UNDA RO, RS, RU, RW, SA, SC, SD, SE, SG, SK, SL, SM,
RODRÍGUEZ, Fernando; Dep. Bio. cel. Facultad ST, SV, sy, TH, TJ, TM, TN, TR, TT, TZ, UA, UG,
de Medicina y Enfermeffa, UPV/EHU, 48940 US, UZ, VC, VN, ZA, ZM, ZW.
LEIOA (ES). PASTOR ALONSO, Oier•, (84) Designated States (unless otherwise indica/ed,
ACHUCARRO BASQUE CENTER FOR for evuy kind of regional pro/ec/Žon available):
NEUROSCIENCE FUN- ARIPO (B W, GM,
DAZIOA, Science Park of the UPV/EHU, Sede
Building, 3rd floor, San-iena, sin, 48940 LEIOA UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU,
TJ, TM), European (AL, AT, BE, BG, CM, cy, CZ,
115
Igor Irastorza Epelde
DE, DR, EE, ES, Fl, FR, GB, GR, HR, HU, IE, IS, IT, as to applicant's
LT, W, LV, MC, MK, MT, NL, NO, PL, PT, RO, RS, entitlement to apply for and he granted a
SE, Sl, SK, SM, TR), OAPI (BF, BJ, CF, CG, Cl, CM, patent (Rule 4.1 7(ii))
GA, GN, GQ, GW, KM, ML, MR, NE, SN, TD, TG). Published:
with international search
Declarations under Rule 4.17:
report (Art. 21 (3)) with sequence listing
part of description (Rule 5.2(a))
(54) Title: CELLULAR AGGREGATES FOR USE IN
VASCULARISATION THERAPY
(57) Abstract: Thc prcscnl invcnlion provides a
scrum-frcc cndothclial cell diffcrcnlialion cullurc
medium comprising (a) a basal cullurc medium and
(b) an cndothclial cell diffcrcnlialion combination of
EGF-FGFand VEGF prolcin, whcrcin lhc amount of
EGF is highcr than lhc amount of FGF prolcin. Thc
prcscnl invcnlion furlhcr provides a process for lhc
preparation of cellular aggrcgalc suspensions
comprising differentiated endothelial cells from
dental stem cells using the serum-free medium, as
well as the use of the rcsulling suspension in
therapy.
116
Annex II
Articles
Annex II
REVIEW
published:16 October 2015
doi:10.3389/fphys.2015.0028 9
Specialty section: Dental pulp stem cells, or DPSC, are neural crest-derived cells
This article was submitted to with an outstanding capacity to differentiate along multiple cell
Craniofacial
Biology, a section of lineages of interest for cell therapy. In particular, highly
the journal Frontiers in efficient osteo/dentinogenic differentiation of DPSC can be
Physiology
achieved using simple in vitro protocols, making these cells a
Received: 31 July 2015
Accepted: 01 October 2015 very attractive and promising tool for the future treatment of
Published: 16 October 2015 dental and periodontal diseases. Among craniomaxillofacial
Citation: organs, the tooth and salivary gland are two such cases in
Aurrekoetxea M, Garcia-Gallastegui
P, which complete regeneration by tissue engineering using
Irastorza I, Luzuriaga J, DPSC appears to be possible, as research over the last
Uribe-Etxebarria V, Unda F and
decade has made substantial progress in experimental
Ibarretxe G (2015) Dental pulp stem
cells as a multifaceted tool for models of partial or total regeneration of both organs, by cell
bioengineering and the regeneration
of craniomaxillofacial tissues. Front. recombination technology. Moreover, DPSC seem to be a
Physiol. 6:289. doi: particularly good choice for the regeneration of nerve tissues,
10.3389/fphys.2015.00289
including injured or transected cranial nerves. In this context,
the oral cavity appears to be an excellent testing ground for
new regenerative therapies using DPSC. However, many
issues and challenges need yet to be addressed before these
cells can be employed in clinical therapy. In this review, we
point out some important aspects on the biology of DPSC with
regard to their use for the reconstruction of different
craniomaxillofacial tissues and organs, with special emphasis
on cranial bones, nerves, teeth, and salivary glands. We
suggest new ideas and strategies to fully exploit the capacities
of DPSC for bioengineering of the aforementioned tissues.
Keywords: DPSC, differentiation, tooth, bone, salivary gland, nerve, cell therapy
119
Annex II
ORIGINALRESEARC H
published:30 March 2016
doi:10.3389/fcell.2016.0002 5
121
Annex II
Gaskon Ibarretxe Cell Biology & Histology Department, Faculty of Medicine and Nursing, University of the Basque
Country, UPV/ and Jose R. Pineda EHU; Achucarro Basque Center for Neuroscience Fundazioa, Barrio Sarriena s/n; Sede
Building 3rd floor, Leioa,
Bizkaia, 48940 (Spain) Tel. (+34) 946013218, E-Mail gaskon.ibarretxe@ehu.eus; jr.pineda@achucarro.org
123
Annex II
European Cells and Materials Vol. 38 2019 (pages 201-214) I Irastorza et al.
hDPSCs with PRF and PRGF on biomimetic titaniumDOI: 10.22203/eCM.v038a14 ISSN 1473-2262
Dental implants are the usual therapy of choice in the dental clinic to replace a loss of natural teeth.
Over recent decades there has been an important progress in the design and manufacturing of
titanium implant surfaces with the goal of improving their osteointegration. In the present work, the
aim was to evaluate the usefulness of hDPSCs (human dental pulp stem cells), in combination with
autologous plasma components, for in vitro bone generation on biomimetic titanium dental implant
materials. In this context, the combination of hDPSCs stimulated by PRGF or PRF and cultured on
standard Ti6A14V and biomimetic BAS™ (Avinent Implant System) titanium surfaces were studied
in order to evaluate possible enhancements in the osteoblastic differentiation process out of human
mesenchymal cells, as well as bone matrix secretion on the implant surface. The results obtained in
this in vitro model of osteogenesis suggested a combination of biomimetic rough titanium surfaces,
such as BAS™, with autologous plasma-derived fibrin-clot membranes such as PRF and/or insoluble
PRGF formulations, but not with an addition of water-soluble supplements of plasma-derived growth
factors, to maximise osteoblastic cell differentiation, bone generation, anchorage and osteointegration
of titanium-made dental implants.
Keywords: Dental pulp stem cells, titanium implants, osteoblast differentiation, platelet rich in
growth factors, platelet rich fibrin, biomimetic advanced surface.
*Address for correspondence: Fernando Unda, Cell Biology and Histology Department. Faculty of
Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, 48940, Bizkaia, Spain.
Telephone number: +34 946012857 Email: fernandoundarodriguez@gmail.com
Copyright policy: This article is distributed in accordance with Creative Commons Attribution
Licence (http://creativecommons.org/licenses/by-sa/4.0/).
125
Annex II
biomedicines
Article
Abstract: The generation of vasculature is one of the most important challenges in tissue
engineering and regeneration. Human dental pulp stem cells (hDPSCs) are some of the most
promising stem cell types to induce vasculogenesis and angiogenesis as they not only secrete
vascular endothelial growth factor (VEGF) but can also differentiate in vitro into both
endotheliocytes and pericytes in serum-free culture media. Moreover, hDPSCs can generate
complete blood vessels containing both endothelial and mural layers in vivo, upon transplantation
into the adult brain. However, many of the serum free media employed for the growth of hDPSCs
contain supplements of an undisclosed composition. This generates uncertainty as to which of its
precise components are necessary and which are dispensable for the vascular differentiation of
hDPSCs, and also hinders the transfer of basic research findings to clinical cell therapy. In this work,
we designed and tested new endothelial differentiation media with a fully defined composition
using standard basal culture media supplemented with a mixture of B27, heparin and growth
factors, including VEGF-A165 at different concentrations. We also optimized an in vitro Matrigel
assay to characterize both the ability of hDPSCs to differentiate to vascular cells and their capacity
to generate vascular tubules in 3D cultures. The description of a fully defined serum-free culture
medium for the induction of vasculogenesis using human adult stem cells highlights its potential as
a relevant innovation for tissue engineering applications. In conclusion, we achieved efficient
vasculogenesis starting from hDPSCs using serum-free culture media with a fully defined
composition, which is applicable for human cell therapy purposes.
127
Annex II
cells
Article
Abstract: Dental pulp stem cells (DPSCs) from adult teeth show the expression of a very
complete repertoire of stem pluripotency core factors and a high plasticity for cell
reprogramming. Canonical
WntandNotchsignalingpathwaysregulatestemnessandtheexpressionofpluripotencycorefactor
sin DPSCs, and even very short-term (48 h) activations of the Wnt pathway induce a profound
remodeling of DPSCs at the physiologic and metabolic levels. In this work, DPSC cultures were
exposed to treatments modulating Notch and Wnt signaling, and also induced to
differentiate to osteo/adipocytes. DNA methylation, histone acetylation, histone
methylation, and core factor expression levels where assessed by mass spectroscopy,
Western blot, and qPCR. A short-term activation of Wnt signaling by WNT-3A induced a
genomic DNA demethylation, and increased histone acetylation and histone methylation in
DPSCs. The efficiency of cell reprogramming methods relies on the ability to surpass the
epigenetic barrier, which determines cell lineage specificity. This study brings important
information about the regulation of the epigenetic barrier by Wnt signaling in DPSCs, which
could contribute to the development of safer and less aggressive reprogramming
methodologies with a view to cell therapy.
Keywords: dental pulp stem cells; chromatin remodeling; cell cycle; pluripotency; DNA
methylation; histone acetylation; histone methylation; Notch pathway; Wnt pathway
129
Annex II
BASIC SCIENCE
Nanomedicine: Nanotechnology, Biology, and Medicine
31 (2021) 102314
Abstract
Withinthe field of neural tissue engineering,there is a huge need for the development of materials that promote the
adhesion,aligned migration and differentiation of stem cells into neuronal and supportive glial cells. In this study, we
have fabricated bioresorbable elastomeric scaffolds combining an ordered nanopatterned topography together with a
surface functionalization with graphene oxide (GO) in mild conditions. These scaffolds allowed the attachment of
murine neural stem cells (NSCs) without the need of any further coating of its surface with extracellular matrix adhesion
proteins. The NSCs were able to give rise to both immature neurons and supporting glial cells over the nanostructured
scaffolds in vitro, promoting their aligned migration in cell clusters following the nanostructured grooves. This system
has the potential to reestablish spatially oriented neural precursor cell connectivity, constituting a promising tool for
future cellular therapy including nerve tissue regeneration. © 2020 Elsevier Inc. All rights reserved.
Key words: Micro- and nanopatterning; Neural stem cells; Migration; Cell differentiation; Graphene oxide; Biodegradable polymer
Funding sources: Basque Government (GV/EJ) Regeneration of the nervous system still remains very challenging
Department of Education, Linguistic Politics and due to its limited plasticity and poor ability to heal ments for this
Culture (GIC 15/52, IT-927-16), MINECO «Ramón y
specific biomedical application play a pivotal role. Much progress
Cajal» program RYC-2013-13450 (JRP), MINECO
PID2019104766RB-C21, The University of The Basque
has been made in determining the ideal features a biomaterial
Country (UPV/EHU) by GIU16/66, UFI 11/44, should have for its use as a neural replacement graft, and in
COLAB19/03 and IKERTU-2020.0155. GV/EJ IT831- understanding the interactions of growing axons within
13, Hazitek ZE-2019/00012-IMABI and ELKARTEK thesebiomaterials;however,theregenerationlevelsinducedbythe
KK-2019/ 00093. Polimerbio and Y. P. have a Bikaintek biomaterial usually do not match those obtained by nerve tissue
PhD grant (20-AF-W2-201800001) and J.L. has a autografts and the development of new and effective nerve
UPV/EHU grant DOKBERRI 2019 (DOCREC19/49). regeneration therapies is still an urgent clinical need.2,3
Conflict of interest: The authors declare that there is no
conflict of interest. The biomaterials for nerve tissue regeneration should be
Correspondence to: J.R. Pineda, Cell Signaling lab,biocompatible and biodegradable, while providing structural cues
University of the Basque Country (UPV/EHU), Leioa, Spain. that promote oriented axon regeneration and guidance signals
Correspondence to: A. Larrañaga, Group of Science from extracellular matrix (ECM)-like components. Additionally,
and Engineering of Polymeric Biomaterials (ZIBIO they shouldalsopresentlong-
Group), University of the Basque Country (UPV/EHU). termstoragecapabilityandeaseofhandling/ suturing.4–6 One
E-mail addresses: joseramon.pinedam@ehu.eus, (J.R. important aspect to take in consideration is that the
Pineda), aitor.larranagae@ehu.eus. (A. Larrañaga).
1
These authors contributed equally to this work.
https://doi.org/10.1016/j.nano.2020.102314
1549-9634/© 2020 Elsevier Inc. All rights reserved.
131
Bibliography
Bibliography
Aaron JE (2012) Periosteal Sharpey’s fibers: a novel bone matrix regulatory system?
Front Endocrinol (Lausanne) 3: 98. doi:10.3389/fendo.2012.00098.
Abe K, Saito H (2000) Neurotrophic effect of basic fibroblast growth factor is mediated
by the p42/p44 mitogen-activated protein kinase cascade in cultured rat cortical neurons. Brain
Res Dev Brain Res 122: 81–85. doi:10.1016/s0165-3806(00)00054-7.
Ackema KB, Charité J (2008) Mesenchymal stem cells from different organs are
characterized by distinct topographic Hox codes. Stem Cells Dev 17: 979–991.
doi:10.1089/scd.2007.0220.
Ajlan SA, Ashri NY, Aldahmash AM, Alnbaheen MS (2015) Osteogenic differentiation of
dental pulp stem cells under the influence of three different materials. BMC Oral Health 15: 132.
doi:10.1186/s12903-015-0113-8.
Almeida-Porada G, Porada C, Zanjani ED (2001) Adult stem cell plasticity and methods
of detection. Rev Clin Exp Hematol 5: 26–41. doi:10.1046/j.1468-0734.2001.00027.x.
Alraies A, Waddington RJ, Sloan AJ, Moseley R (2020) Evaluation of Dental Pulp Stem
Cell Heterogeneity and Behaviour in 3D Type I Collagen Gels. Biomed Res Int 2020: 3034727.
doi:10.1155/2020/3034727.
Amini AR, Laurencin CT, Nukavarapu SP (2012) Bone tissue engineering: recent advances
and challenges. Crit Rev Biomed Eng 40: 363–408. doi:10.1615/critrevbiomedeng.v40.i5.10.
Anitua E (1999) Plasma rich in growth factors: preliminary results of use in the
preparation of future sites for implants. Int J Oral Maxillofac Implants 14: 529–535.
135
Igor Irastorza Epelde
Anitua E, Orive G, Pla R, Roman P, Serrano V, Andía I (2009) The effects of PRGF on bone
regeneration and on titanium implant osseointegration in goats: a histologic and
histomorphometric study. J Biomed Mater Res A 91: 158–165. doi:10.1002/jbm.a.32217.
Anitua E, Tejero R, Zalduendo MM, Orive G (2013) Plasma rich in growth factors
promotes bone tissue regeneration by stimulating proliferation, migration, and autocrine
secretion in primary human osteoblasts. J. Periodontol. 84: 1180–1190.
doi:10.1902/jop.2012.120292.
Arthur A, Rychkov G, Shi S, Koblar SA, Gronthos S (2008) Adult human dental pulp stem
cells differentiate toward functionally active neurons under appropriate environmental cues.
Stem Cells 26: 1787–1795. doi:10.1634/stemcells.2007-0979.
Bae S, Kang B, Lee H, Luu H, Mullins E, Kingsley K (2021) Characterization of Dental Pulp
Stem Cell Responses to Functional Biomaterials Including Mineralized Trioxide Aggregates. J
Funct Biomater 12. doi:10.3390/jfb12010015.
136
Bibliography
Baulies A, Angelis N, Li VSW (2020) Hallmarks of intestinal stem cells. Development 147.
doi:10.1242/dev.182675.
Bertolini MM, Del Bel Cury AA, Pizzoloto L, Acapa IRH, Shibli JA, Bordin D (2019) Does
traumatic occlusal forces lead to peri-implant bone loss? A systematic review. Braz Oral Res 33:
e069. doi:10.1590/1807-3107bor-2019.vol33.0069.
Bhat S, Chiew GGY, Ng JX, Lin X, Seetharam RN (2021) Optimization of culture conditions
for human bone marrow-derived mesenchymal stromal cell expansion in macrocarrier-based
tide motion system. Biotechnol J: e2000540. doi:10.1002/biot.202000540.
Bhuptani RS, Patravale VB (2016) Porous microscaffolds for 3D culture of dental pulp
mesenchymal stem cells. Int J Pharm 515: 555–564. doi:10.1016/j.ijpharm.2016.10.040.
Bianchi M, Urquia Edreira ER, Wolke JGC, Birgani ZT, Habibovic P, Jansen JA, Tampieri A,
Marcacci M, Leeuwenburgh SCG, van den Beucken JJJP (2014) Substrate geometry directs the in
vitro mineralization of calcium phosphate ceramics. Acta Biomater 10: 661–669.
doi:10.1016/j.actbio.2013.10.026.
Bianco P, Riminucci M, Gronthos S, Robey PG (2001) Bone marrow stromal stem cells:
nature, biology, and potential applications. Stem Cells 19: 180–192. doi:10.1634/stemcells.19-
3-180.
137
Igor Irastorza Epelde
Bühring H-J, Battula VL, Treml S, Schewe B, Kanz L, Vogel W (2007) Novel markers for
the prospective isolation of human MSC. Ann N Y Acad Sci 1106: 262–271.
doi:10.1196/annals.1392.000.
Cao C, Dong Y, Dong Y (2005) [Study on culture and in vitro osteogenesis of blood-
derived human mesenchymal stem cells]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 19: 642–
647.
Chang C-C, Chang K-C, Tsai S-J, Chang H-H, Lin C-P (2014) Neurogenic differentiation of
dental pulp stem cells to neuron-like cells in dopaminergic and motor neuronal inductive media.
J Formos Med Assoc 113: 956–965. doi:10.1016/j.jfma.2014.09.003.
Chaudhari AA, Vig K, Baganizi DR, Sahu R, Dixit S, Dennis V, Singh SR, Pillai SR (2016)
Future Prospects for Scaffolding Methods and Biomaterials in Skin Tissue Engineering: A Review.
Int J Mol Sci 17. doi:10.3390/ijms17121974.
Chen YK, Huang AHC, Chan AWS, Lin LM (2016) Human dental pulp stem cells derived
from cryopreserved dental pulp tissues of vital extracted teeth with disease demonstrate
hepatic-like differentiation. J Tissue Eng Regen Med 10: 475–485. doi:10.1002/term.1763.
Cheng L, Qasba P, Vanguri P, Thiede MA (2000) Human mesenchymal stem cells support
megakaryocyte and pro-platelet formation from CD34(+) hematopoietic progenitor cells. J Cell
Physiol 184: 58–69. doi:10.1002/(SICI)1097-4652(200007)184:1<58::AID-JCP6>3.0.CO;2-B.
138
Bibliography
Choi YC, Choi JS, Kim BS, Kim JD, Yoon HI, Cho YW (2012) Decellularized extracellular
matrix derived from porcine adipose tissue as a xenogeneic biomaterial for tissue engineering.
Tissue Eng Part C Methods 18: 866–876. doi:10.1089/ten.TEC.2012.0009.
Civin CI, Trischmann T, Kadan NS, Davis J, Noga S, Cohen K, Duffy B, Groenewegen I,
Wiley J, Law P, Hardwick A, Oldham F, Gee A (1996) Highly purified CD34-positive cells
reconstitute hematopoiesis. J Clin Oncol 14: 2224–2233. doi:10.1200/JCO.1996.14.8.2224.
Coelho PG, Granjeiro JM, Romanos GE, Suzuki M, Silva NRF, Cardaropoli G, Thompson
VP, Lemons JE (2009) Basic research methods and current trends of dental implant surfaces. J.
Biomed. Mater. Res. Part B Appl. Biomater. 88: 579–596. doi:10.1002/jbm.b.31264.
Coli P, Jemt T (2021) Are marginal bone level changes around dental implants due to
infection? Clin Implant Dent Relat Res. doi:10.1111/cid.12971.
Conget PA, Minguell JJ (1999) Phenotypical and functional properties of human bone
marrow mesenchymal progenitor cells. J Cell Physiol 181: 67–73. doi:10.1002/(SICI)1097-
4652(199910)181:1<67::AID-JCP7>3.0.CO;2-C.
Crisan M, Yap S, Casteilla L, Chen C-W, Corselli M, Park TS, Andriolo G, Sun B, Zheng B,
Zhang L, Norotte C, Teng P-N, Traas J, Schugar R, Deasy BM, Badylak S, Buhring H-J, Giacobino J-
P, Lazzari L, Huard J, Péault B (2008) A perivascular origin for mesenchymal stem cells in multiple
human organs. Cell Stem Cell 3: 301–313. doi:10.1016/j.stem.2008.07.003.
Cuthbert RJ, Giannoudis PV, Wang XN, Nicholson L, Pawson D, Lubenko A, Tan HB,
Dickinson A, McGonagle D, Jones E (2015) Examining the Feasibility of Clinical Grade CD271+
Enrichment of Mesenchymal Stromal Cells for Bone Regeneration. PLoS One 10.
doi:10.1371/journal.pone.0117855.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4356586/.
Dagnino APA, Chagastelles PC, Medeiros RP, Estrázulas M, Kist LW, Bogo MR, Weber
JBB, Campos MM, Silva JB (2020) Neural Regenerative Potential of Stem Cells Derived from the
Tooth Apical Papilla. Stem Cells Dev 29: 1479–1496. doi:10.1089/scd.2020.0121.
Davies OG, Cooper PR, Shelton RM, Smith AJ, Scheven BA (2015) A comparison of the in
vitro mineralisation and dentinogenic potential of mesenchymal stem cells derived from adipose
139
Igor Irastorza Epelde
tissue, bone marrow and dental pulp. J Bone Miner Metab 33: 371–382. doi:10.1007/s00774-
014-0601-y.
Denham M, Conley B, Olsson F, Cole TJ, Mollard R (2005) Stem cells: an overview. Curr
Protoc Cell Biol Chapter 23: Unit 23.1. doi:10.1002/0471143030.cb2301s28.
Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, Prockop DJ (1999) Propagation
and senescence of human marrow stromal cells in culture: a simple colony-forming assay
identifies samples with the greatest potential to propagate and differentiate. Br J Haematol 107:
275–281. doi:10.1046/j.1365-2141.1999.01715.x.
Dlaska CE, Andersson G, Brittberg M, Suedkamp NP, Raschke MJ, Schuetz MA (2015)
Clinical Translation in Tissue Engineering—The Surgeon’s View. Curr Mol Bio Rep 1: 61–70.
doi:10.1007/s40610-015-0013-3.
Dohan DM, Choukroun J, Diss A, Dohan SL, Dohan AJJ, Mouhyi J, Gogly B (2006) Platelet-
rich fibrin (PRF): a second-generation platelet concentrate. Part II: platelet-related biologic
features. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 101: e45-50.
doi:10.1016/j.tripleo.2005.07.009.
140
Bibliography
Duncan AW, Dorrell C, Grompe M (2009) Stem Cells and Liver Regeneration.
Gastroenterology 137: 466–481. doi:10.1053/j.gastro.2009.05.044.
Fijnheer R, Pietersz RN, de Korte D, Gouwerok CW, Dekker WJ, Reesink HW, Roos D
(1990) Platelet activation during preparation of platelet concentrates: a comparison of the
platelet-rich plasma and the buffy coat methods. Transfusion 30: 634–638. doi:10.1046/j.1537-
2995.1990.30790385523.x.
Fraser JK, Wulur I, Alfonso Z, Hedrick MH (2006) Fat tissue: an underappreciated source
of stem cells for biotechnology. Trends Biotechnol 24: 150–154.
doi:10.1016/j.tibtech.2006.01.010.
Friedenstein AJ, Gorskaja JF, Kulagina NN (1976) Fibroblast precursors in normal and
irradiated mouse hematopoietic organs. Exp Hematol 4: 267–274.
141
Igor Irastorza Epelde
Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RCR (2007) SSEA-4
identifies mesenchymal stem cells from bone marrow. Blood 109: 1743–1751.
doi:10.1182/blood-2005-11-010504.
Gothard D, Dawson JI, Oreffo ROC (2013) Assessing the potential of colony morphology
for dissecting the CFU-F population from human bone marrow stromal cells. Cell Tissue Res 352:
237–247. doi:10.1007/s00441-013-1564-3.
Griffiths MJD, Bonnet D, Janes SM (2005) Stem cells of the alveolar epithelium. Lancet
366: 249–260. doi:10.1016/S0140-6736(05)66916-4.
Gronthos S, Brahim J, Li W, Fisher LW, Cherman N, Boyde A, DenBesten P, Robey PG, Shi
S (2002) Stem cell properties of human dental pulp stem cells. J Dent Res 81: 531–535.
doi:10.1177/154405910208100806.
Gronthos S, Mankani M, Brahim J, Robey PG, Shi S (2000) Postnatal human dental pulp
stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. U.S.A. 97: 13625–13630.
doi:10.1073/pnas.240309797.
Gronthos S, Zannettino ACW, Hay SJ, Shi S, Graves SE, Kortesidis A, Simmons PJ (2003)
Molecular and cellular characterisation of highly purified stromal stem cells derived from human
bone marrow. J Cell Sci 116: 1827–1835. doi:10.1242/jcs.00369.
142
Bibliography
Grottkau BE, Purudappa PP, Lin Y (2010) Multilineage differentiation of dental pulp stem
cells from green fluorescent protein transgenic mice. Int J Oral Sci 2: 21–27.
doi:10.4248/IJOS10015.
Han Y-J, Kang Y-H, Shivakumar SB, Bharti D, Son Y-B, Choi Y-H, Park W-U, Byun J-H, Rho
G-J, Park B-W (2017) Stem Cells from Cryopreserved Human Dental Pulp Tissues Sequentially
Differentiate into Definitive Endoderm and Hepatocyte-Like Cells in vitro. Int J Med Sci 14: 1418–
1429. doi:10.7150/ijms.22152.
Haynesworth SE, Baber MA, Caplan AI (1992) Cell surface antigens on human marrow-
derived mesenchymal cells are detected by monoclonal antibodies. Bone 13: 69–80.
doi:10.1016/8756-3282(92)90363-2.
Hench LL, Polak JM (2002) Third-generation biomedical materials. Science 295: 1014–
1017. doi:10.1126/science.1067404.
Hodgkinson T, Yuan X-F, Bayat A (2009) Adult stem cells in tissue engineering. Expert
Rev Med Devices 6: 621–640. doi:10.1586/erd.09.48.
Honda MJ, Imaizumi M, Tsuchiya S, Morsczeck C (2010) Dental follicle stem cells and
tissue engineering. J Oral Sci 52: 541–552. doi:10.2334/josnusd.52.541.
Horwitz EM, Keating A (2000) Nonhematopoietic mesenchymal stem cells: what are
they? Cytotherapy 2: 387–388.
Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, Deans RJ,
Krause DS, Keating A, International Society for Cellular Therapy (2005) Clarification of the
nomenclature for MSC: The International Society for Cellular Therapy position statement.
Cytotherapy 7: 393–395. doi:10.1080/14653240500319234.
143
Igor Irastorza Epelde
combined with plasma derived products. Eur Cell Mater 38: 201–214.
doi:10.22203/eCM.v038a14.
Iviglia G, Kargozar S, Baino F (2019) Biomaterials, Current Strategies, and Novel Nano-
Technological Approaches for Periodontal Regeneration. J Funct Biomater 10.
doi:10.3390/jfb10010003.
Jameson CA (2007) Autologous Platelet Concentrate for the Production of Platelet Gel.
Laboratory Medicine 38: 39–42. doi:10.1309/3UA5HWYVKNCE01AR.
Jang J-H, Lee H-W, Cho KM, Shin H-W, Kang MK, Park SH, Kim E (2016) In vitro
characterization of human dental pulp stem cells isolated by three different methods. Restor
Dent Endod 41: 283–295. doi:10.5395/rde.2016.41.4.283.
Jemat A, Ghazali MJ, Razali M, Otsuka Y (2015) Surface Modifications and Their Effects
on Titanium Dental Implants. Biomed Res Int 2015: 791725. doi:10.1155/2015/791725.
Jones EA, English A, Kinsey SE, Straszynski L, Emery P, Ponchel F, McGonagle D (2006)
Optimization of a flow cytometry-based protocol for detection and phenotypic characterization
of multipotent mesenchymal stromal cells from human bone marrow. Cytometry B Clin Cytom
70: 391–399. doi:10.1002/cyto.b.20118.
Jones EA, Kinsey SE, English A, Jones RA, Straszynski L, Meredith DM, Markham AF, Jack
A, Emery P, McGonagle D (2002) Isolation and characterization of bone marrow multipotential
mesenchymal progenitor cells. Arthritis Rheum 46: 3349–3360. doi:10.1002/art.10696.
144
Bibliography
doi:10.5772/55927. https://www.intechopen.com/books/regenerative-medicine-and-tissue-
engineering/dental-related-stem-cells-and-their-potential-in-regenerative-medicine.
Kawashima N (2012) Characterisation of dental pulp stem cells: a new horizon for tissue
regeneration? Arch Oral Biol 57: 1439–1458. doi:10.1016/j.archoralbio.2012.08.010.
Kerkis I, Kerkis A, Dozortsev D, Stukart-Parsons GC, Gomes Massironi SM, Pereira LV,
Caplan AI, Cerruti HF (2006) Isolation and characterization of a population of immature dental
pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs
184: 105–116. doi:10.1159/000099617.
Khan SN, Cammisa FP, Sandhu HS, Diwan AD, Girardi FP, Lane JM (2005) The biology of
bone grafting. J Am Acad Orthop Surg 13: 77–86.
Khanabdali R, Saadat A, Fazilah M, Bazli KFK, Qazi R-M, Khalid RS, Hasan Adli DS,
Moghadamtousi SZ, Naeem N, Khan I, Salim A, Shamsuddin SA, Mohan G (2016) Promoting
effect of small molecules in cardiomyogenic and neurogenic differentiation of rat bone marrow-
derived mesenchymal stem cells. Drug Des Devel Ther 10: 81–91. doi:10.2147/DDDT.S89658.
Khurana R, Kudva PB, Husain SY (2017) Comparative evaluation of the isolation and
quantification of stem cells derived from dental pulp and periodontal ligament of a permanent
tooth and to assess their viability and proliferation on a platelet-rich fibrin scaffold. J Indian Soc
Periodontol 21: 16–20. doi:10.4103/jisp.jisp_182_16.
Király M, Kádár K, Horváthy DB, Nardai P, Rácz GZ, Lacza Z, Varga G, Gerber G (2011)
Integration of neuronally predifferentiated human dental pulp stem cells into rat brain in vivo.
Neurochem Int 59: 371–381. doi:10.1016/j.neuint.2011.01.006.
Knychala J, Bouropoulos N, Catt CJ, Katsamenis OL, Please CP, Sengers BG (2013) Pore
geometry regulates early stage human bone marrow cell tissue formation and organisation. Ann
Biomed Eng 41: 917–930. doi:10.1007/s10439-013-0748-z.
145
Igor Irastorza Epelde
Koç ON, Peters C, Aubourg P, Raghavan S, Dyhouse S, DeGasperi R, Kolodny EH, Yoseph
YB, Gerson SL, Lazarus HM, Caplan AI, Watkins PA, Krivit W (1999) Bone marrow-derived
mesenchymal stem cells remain host-derived despite successful hematopoietic engraftment
after allogeneic transplantation in patients with lysosomal and peroxisomal storage diseases.
Exp Hematol 27: 1675–1681. doi:10.1016/s0301-472x(99)00101-0.
Kokubo T, Takadama H (2006) How useful is SBF in predicting in vivo bone bioactivity?
Biomaterials 27: 2907–2915. doi:10.1016/j.biomaterials.2006.01.017.
Kumar KR, Genmorgan K, Abdul Rahman SM, Rajan MA, Kumar TA, Prasad VS (2016)
Role of plasma-rich fibrin in oral surgery. J Pharm Bioallied Sci 8: S36–S38. doi:10.4103/0975-
7406.191963.
Kumar RV, Shubhashini N (2013) Platelet rich fibrin: a new paradigm in periodontal
regeneration. Cell Tissue Bank 14: 453–463. doi:10.1007/s10561-012-9349-6.
Lee CP, Colombo JS, Ayre WN, Sloan AJ, Waddington RJ (2015a) Elucidating the cellular
actions of demineralised dentine matrix extract on a clonal dental pulp stem cell population in
orchestrating dental tissue repair. J Tissue Eng 6: 2041731415586318.
doi:10.1177/2041731415586318.
Lee J-S, Kim S-K, Gruber R, Kim C-S (2020) Periodontal healing by periodontal ligament
fiber with or without cells: A preclinical study of the decellularized periodontal ligament in a
tooth replantation model. J Periodontol 91: 110–119. doi:10.1002/JPER.19-0126.
Lee J-T, Choi S-Y, Kim H-L, Kim J-Y, Lee H-J, Kwon T-G (2015b) Comparison of gene
expression between mandibular and iliac bone-derived cells. Clin Oral Invest 19: 1223–1233.
doi:10.1007/s00784-014-1353-8.
Lenkiewicz AM (2019) Epidermal Stem Cells. Adv Exp Med Biol 1201: 239–259.
doi:10.1007/978-3-030-31206-0_12.
Leucht P, Kim J-B, Amasha R, James AW, Girod S, Helms JA (2008) Embryonic origin and
Hox status determine progenitor cell fate during adult bone regeneration. Development 135:
2845–2854. doi:10.1242/dev.023788.
146
Bibliography
Liu N, Zhou M, Zhang Q, Yong L, Zhang T, Tian T, Ma Q, Lin S, Zhu B, Cai X (2018) Effect
of substrate stiffness on proliferation and differentiation of periodontal ligament stem cells. Cell
Prolif 51: e12478. doi:10.1111/cpr.12478.
Madarieta Pardo I, García Urquia N, Fernandez García R (2017) Method for Producing a
Decellularized Tissue Matrix. July 6.
https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017114902.
Majo F, Rochat A, Nicolas M, Jaoudé GA, Barrandon Y (2008) Oligopotent stem cells are
distributed throughout the mammalian ocular surface. Nature 456: 250–254.
doi:10.1038/nature07406.
Mansergh FC, Wride MA, Rancourt DE (2000) Neurons from stem cells: implications for
understanding nervous system development and repair. Biochem Cell Biol 78: 613–628.
Marx RE, Carlson ER, Eichstaedt RM, Schimmele SR, Strauss JE, Georgeff KR (1998)
Platelet-rich plasma: Growth factor enhancement for bone grafts. Oral Surg Oral Med Oral
Pathol Oral Radiol Endod 85: 638–646. doi:10.1016/s1079-2104(98)90029-4.
147
Igor Irastorza Epelde
Marx RE (2004) Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg
62: 489–496. doi:10.1016/j.joms.2003.12.003.
Mayer Y, Ginesin O, Khutaba A, Machtei EE, Zigdon Giladi H (2018) Biocompatibility and
osteoconductivity of PLCL coated and noncoated xenografts: An in vitro and preclinical trial. Clin
Implant Dent Relat Res 20: 294–299. doi:10.1111/cid.12596.
McElreavey KD, Irvine AI, Ennis KT, McLean WH (1991) Isolation, culture and
characterisation of fibroblast-like cells derived from the Wharton’s jelly portion of human
umbilical cord. Biochem Soc Trans 19: 29S. doi:10.1042/bst019029s.
Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S (2003) SHED: stem cells
from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A 100: 5807–5812.
doi:10.1073/pnas.0937635100.
Murphy CM, Haugh MG, O’Brien FJ (2010) The effect of mean pore size on cell
attachment, proliferation and migration in collagen-glycosaminoglycan scaffolds for bone tissue
engineering. Biomaterials 31: 461–466. doi:10.1016/j.biomaterials.2009.09.063.
148
Bibliography
Nada OA, El Backly RM (2018) Stem Cells From the Apical Papilla (SCAP) as a Tool for
Endogenous Tissue Regeneration. Front Bioeng Biotechnol 6: 103.
doi:10.3389/fbioe.2018.00103.
Naves MM, Menezes HHM, Magalhães D, Ferreira JA, Ribeiro SF, de Mello JDB, Costa HL
(2015) Effect of Macrogeometry on the Surface Topography of Dental Implants. Int J Oral
Maxillofac Implants 30: 789–799.
Nemeth CL, Janebodin K, Yuan AE, Dennis JE, Reyes M, Kim D-H (2014) Enhanced
chondrogenic differentiation of dental pulp stem cells using nanopatterned PEG-GelMA-HA
hydrogels. Tissue Eng Part A 20: 2817–2829. doi:10.1089/ten.TEA.2013.0614.
O’Brien FJ (2011) Biomaterials & scaffolds for tissue engineering. Materials Today 14:
88–95. doi:10.1016/S1369-7021(11)70058-X.
Olivares-Navarrete R, Hyzy SL, Hutton DL, Erdman CP, Wieland M, Boyan BD, Schwartz Z
(2010) Direct and indirect effects of microstructured titanium substrates on the induction of
mesenchymal stem cell differentiation towards the osteoblast lineage. Biomaterials 31: 2728–
2735. doi:10.1016/j.biomaterials.2009.12.029.
Onizuka S, Iwata T, Park S-J, Nakai K, Yamato M, Okano T, Izumi Y (2016) ZBTB16 as a
Downstream Target Gene of Osterix Regulates Osteoblastogenesis of Human Multipotent
Mesenchymal Stromal Cells. J Cell Biochem 117: 2423–2434. doi:10.1002/jcb.25634.
Orimo H (2010) The mechanism of mineralization and the role of alkaline phosphatase
in health and disease. J Nippon Med Sch 77: 4–12. doi:10.1272/jnms.77.4.
Pagella P, Miran S, Neto E, Martin I, Lamghari M, Mitsiadis TA (2020) Human dental pulp
stem cells exhibit enhanced properties in comparison to human bone marrow stem cells on
149
Igor Irastorza Epelde
Pilipchuk SP, Plonka AB, Monje A, Taut AD, Lanis A, Kang B, Giannobile WV (2015) Tissue
engineering for bone regeneration and osseointegration in the oral cavity. Dent Mater 31: 317–
338. doi:10.1016/j.dental.2015.01.006.
Pineda Martí JR, Luzuriaga González J, Unda Rodríguez F, Pastor Alonso O, Encinas Pérez
JM, Ibarretxe Bilbao G, Irastorza Epelde I (2020) Cellular Aggregates for Use in Vascularisation
Therapy. January 9. https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2020007878.
Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA,
Simonetti DW, Craig S, Marshak DR (1999) Multilineage potential of adult human mesenchymal
stem cells. Science 284: 143–147. doi:10.1126/science.284.5411.143.
Potten CS, Loeffler M (1990) Stem cells: attributes, cycles, spirals, pitfalls and
uncertainties. Lessons for and from the crypt. Development 110: 1001–1020.
150
Bibliography
Powers CJ, McLeskey SW, Wellstein A (2000) Fibroblast growth factors, their receptors
and signaling. Endocr Relat Cancer 7: 165–197. doi:10.1677/erc.0.0070165.
Prockop DJ (1997) Marrow stromal cells as stem cells for nonhematopoietic tissues.
Science 276: 71–74. doi:10.1126/science.276.5309.71.
Raik S, Kumar A, Rattan V, Seth S, Kaur A, Bhatta Charyya S (2020) Assessment of Post-
thaw Quality of Dental Mesenchymal Stromal Cells After Long-Term Cryopreservation by
Uncontrolled Freezing. Appl Biochem Biotechnol 191: 728–743. doi:10.1007/s12010-019-
03216-6.
Rao SM, Ugale GM, Warad SB (2013) Bone Morphogenetic Proteins: Periodontal
Regeneration. N Am J Med Sci 5: 161–168. doi:10.4103/1947-2714.109175.
Reuss B, von Bohlen und Halbach O (2003) Fibroblast growth factors and their receptors
in the central nervous system. Cell Tissue Res 313: 139–157. doi:10.1007/s00441-003-0756-7.
Roberts TT, Rosenbaum AJ (2012) Bone grafts, bone substitutes and orthobiologics: the
bridge between basic science and clinical advancements in fracture healing. Organogenesis 8:
114–124. doi:10.4161/org.23306.
Roosa SMM, Kemppainen JM, Moffitt EN, Krebsbach PH, Hollister SJ (2010) The pore size
of polycaprolactone scaffolds has limited influence on bone regeneration in an in vivo model. J
Biomed Mater Res A 92: 359–368. doi:10.1002/jbm.a.32381.
Santos F dos, Andrade PZ, Abecasis MM, Gimble JM, Chase LG, Campbell AM, Boucher
S, Vemuri MC, Silva CL da, Cabral JMS (2011) Toward a clinical-grade expansion of mesenchymal
stem cells from human sources: a microcarrier-based culture system under xeno-free
conditions. Tissue Eng Part C Methods 17: 1201–1210. doi:10.1089/ten.tec.2011.0255.
151
Igor Irastorza Epelde
Scintu F, Reali C, Pillai R, Badiali M, Sanna MA, Argiolu F, Ristaldi MS, Sogos V (2006)
Differentiation of human bone marrow stem cells into cells with a neural phenotype: diverse
effects of two specific treatments. BMC Neurosci 7: 14. doi:10.1186/1471-2202-7-14.
Sedgley CM, Botero TM (2012) Dental stem cells and their sources. Dent Clin North Am
56: 549–561. doi:10.1016/j.cden.2012.05.004.
Seo B-M, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, Young M, Robey PG,
Wang C-Y, Shi S (2004) Investigation of multipotent postnatal stem cells from human periodontal
ligament. Lancet 364: 149–155. doi:10.1016/S0140-6736(04)16627-0.
Shyamala K, Yanduri S, Girish HC, Murgod S (2015) Neural crest: The fourth germ layer.
J Oral Maxillofac Pathol 19: 221–229. doi:10.4103/0973-029X.164536.
da Silva Meirelles L, Chagastelles PC, Nardi NB (2006) Mesenchymal stem cells reside in
virtually all post-natal organs and tissues. J Cell Sci 119: 2204–2213. doi:10.1242/jcs.02932.
Smith AG (2001) Embryo-derived stem cells: of mice and men. Annu. Rev. Cell Dev. Biol.
17: 435–462. doi:10.1146/annurev.cellbio.17.1.435.
Solchaga LA, Penick KJ, Welter JF (2011) Chondrogenic Differentiation of Bone Marrow-
Derived Mesenchymal Stem Cells: Tips and Tricks. Methods Mol Biol 698: 253–278.
doi:10.1007/978-1-60761-999-4_20.
Song B, Jiang W, Alraies A, Liu Q, Gudla V, Oni J, Wei X, Sloan A, Ni L, Agarwal M (2016)
Bladder Smooth Muscle Cells Differentiation from Dental Pulp Stem Cells: Future Potential for
Bladder Tissue Engineering. Stem Cells Int 2016: 6979368. doi:10.1155/2016/6979368.
Sordi MB, Curtarelli RB, da Silva IT, Fongaro G, Benfatti CAM, de Souza Magini R, Cabral
da Cruz AC (2021) Effect of dexamethasone as osteogenic supplementation in in vitro osteogenic
differentiation of stem cells from human exfoliated deciduous teeth. J Mater Sci Mater Med 32:
1. doi:10.1007/s10856-020-06475-6.
152
Bibliography
Stevens MM (2008) Biomaterials for bone tissue engineering. Materials Today 11: 18–
25. doi:10.1016/S1369-7021(08)70086-5.
Tatullo M ed. (2017) MSCs and Innovative Biomaterials in Dentistry. Humana Press. Stem
Cell Biology and Regenerative Medicine. //www.springer.com/us/book/9783319556444.
Thesleff I, Aberg T (1999) Molecular regulation of tooth development. Bone 25: 123–
125. doi:10.1016/s8756-3282(99)00119-2.
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones
JM (1998) Embryonic stem cell lines derived from human blastocysts. Science 282: 1145–1147.
Tirino V, Paino F, d’Aquino R, Desiderio V, De Rosa A, Papaccio G (2011) Methods for the
identification, characterization and banking of human DPSCs: current strategies and
perspectives. Stem Cell Rev 7: 608–615. doi:10.1007/s12015-011-9235-9.
Tziafas D, Smith AJ, Lesot H (2000) Designing new treatment strategies in vital pulp
therapy. J Dent 28: 77–92. doi:10.1016/s0300-5712(99)00047-0.
Vats A, Bielby RC, Tolley NS, Nerem R, Polak JM (2005) Stem cells. Lancet 366: 592–602.
doi:10.1016/S0140-6736(05)66879-1.
Vega-Lopez GA, Cerrizuela S, Aybar MJ (2017) Trunk neural crest cells: formation,
migration and beyond. Int J Dev Biol 61: 5–15. doi:10.1387/ijdb.160408gv.
Wagers AJ, Weissman IL (2004) Plasticity of adult stem cells. Cell 116: 639–648.
doi:10.1016/s0092-8674(04)00208-9.
Wang D-R, Wang Y-H, Pan J, Tian W-D (2020) Neurotrophic effects of dental pulp stem
cells in repair of peripheral nerve after crush injury. World J Stem Cells 12: 1196–1213.
doi:10.4252/wjsc.v12.i10.1196.
153
Igor Irastorza Epelde
Wang H-S, Hung S-C, Peng S-T, Huang C-C, Wei H-M, Guo Y-J, Fu Y-S, Lai M-C, Chen C-C
(2004) Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells
22: 1330–1337. doi:10.1634/stemcells.2004-0013.
Wang KC, Helms JA, Chang HY (2009) Regeneration, repair and remembering identity:
the three Rs of Hox gene expression. Trends Cell Biol 19: 268–275.
doi:10.1016/j.tcb.2009.03.007.
Whitman DH, Berry RL, Green DM (1997) Platelet gel: an autologous alternative to fibrin
glue with applications in oral and maxillofacial surgery. J Oral Maxillofac Surg 55: 1294–1299.
doi:10.1016/s0278-2391(97)90187-7.
Wu J, Zhang W, Ran Q, Xiang Y, Zhong JF, Li SC, Li Z (2018) The Differentiation Balance of
Bone Marrow Mesenchymal Stem Cells Is Crucial to Hematopoiesis. Stem Cells Int 2018:
1540148. doi:10.1155/2018/1540148.
Yadav P, Vats R, Bano A, Bhardwaj R (2020) Hematopoietic Stem Cells Culture, Expansion
and Differentiation: An Insight into Variable and Available Media. Int J Stem Cells 13: 326–334.
doi:10.15283/ijsc19157.
Yang J-Z, Qiu L-H, Xiong S-H, Dang J-L, Rong X-K, Hou M-M, Wang K, Yu Z, Yi C-G (2020)
Decellularized adipose matrix provides an inductive microenvironment for stem cells in tissue
regeneration. World J Stem Cells 12: 585–603. doi:10.4252/wjsc.v12.i7.585.
Yildirim S (2013) Dental Pulp Stem Cells. New York: Springer-Verlag. SpringerBriefs in
Stem Cells. doi:10.1007/978-1-4614-5687-2.
https://www.springer.com/gp/book/9781461456865.
154
Bibliography
Zanicotti DG, Duncan WJ, Seymour GJ, Coates DE (2018) Effect of Titanium Surfaces on
the Osteogenic Differentiation of Human Adipose-Derived Stem Cells. Int J Oral Maxillofac
Implants 33: e77–e87. doi:10.11607/jomi.5810.
Zhang H-T, Liu Z-L, Yao X-Q, Yang Z-J, Xu R-X (2012) Neural differentiation ability of
mesenchymal stromal cells from bone marrow and adipose tissue: a comparative study.
Cytotherapy 14: 1203–1214. doi:10.3109/14653249.2012.711470.
Zhang J, Ding H, Liu X, Sheng Y, Liu X, Jiang C (2019) Dental Follicle Stem Cells: Tissue
Engineering and Immunomodulation. Stem Cells Dev 28: 986–994. doi:10.1089/scd.2019.0012.
Zhang J, Lian M, Cao P, Bao G, Xu G, Sun Y, Wang L, Chen J, Wang Y, Feng G, Cui Z (2017)
Effects of Nerve Growth Factor and Basic Fibroblast Growth Factor Promote Human Dental Pulp
Stem Cells to Neural Differentiation. Neurochem Res 42: 1015–1025. doi:10.1007/s11064-016-
2134-3.
Zhang N, Chen B, Wang W, Chen C, Kang J, Deng SQ, Zhang B, Liu S, Han F (2016)
Isolation, characterization and multi-lineage differentiation of stem cells from human exfoliated
deciduous teeth. Mol Med Rep 14: 95–102. doi:10.3892/mmr.2016.5214.
Zhao X, Gong P, Lin Y, Wang J, Yang X, Cai X (2012) Characterization of α-smooth muscle
actin positive cells during multilineage differentiation of dental pulp stem cells. Cell Prolif 45:
259–265. doi:10.1111/j.1365-2184.2012.00818.x.
Zheng Y-H, Xiong W, Su K, Kuang S-J, Zhang Z-G (2013) Multilineage differentiation of
human bone marrow mesenchymal stem cells in vitro and in vivo. Exp Ther Med 5: 1576–1580.
doi:10.3892/etm.2013.1042.
155
ZELULEN BIOLOGIA ETA HISTOLOGIA SAILA
MEDIKUNTZA ETA ERIZAINTZA FAKULTATEA
EUSKAL HERRIKO UNIBERTSITATEA
Laburpena …………………………………………………………………….………………………..…….…………… 1
Laburdurak ………………………………………………………………………………….………..…………………… 5
Sarrera ..…………………………………………………………………………………………………………………… 11
Ehun ingeniaritzari buruzko sarrera ….………………………………………..………………………. 13
Zelulak amak ………………………………………………………………………………………..…………….. 14
Zelula ama helduak ……..……………………………………………………………………….…………. 16
Zelula ama mesenkimalak ….……………………………………………………..….……..…….. 17
Hortz mamiko zelula amak (DPSC) …………………………………………………..…….. 18
Ezaugarri orokorrak ……………………………………………………………..……….…… 18
Azalera markatzaileak ….…………………………………………………..……………….. 20
DPSC-en desberdintzapena ……………………………………………………..………… 22
Hezur muineko zelula amak (BMSC) ….………………………………………..…………. 25
Ezaugarri orokorrak ..…………………………………………………………….………..…. 25
Azalera markatzaileak ….……………………………………………………………………. 26
BMSC-en desberdintzapena ..…………………………………….…………………….… 27
Hezur ehun ingeniaritzarako aldamioak (Scaffolds) ……………………………………………. 29
Titanioa ..……………………………………………………………………………………..……………….… 31
Dezelularizatutako ehun adiposoa ….……………………………………………..……………….. 34
HAzkuntza faktoreak ….…………………………………………………………………….………………… 35
Plasmatik eratorritako produktuak ….………………………………….…………………………. 36
Metodologia ..…………………………………………………………………………….……………………………. 39
Hipotesia ..…………………………………………………………………………………………….…………………. 51
Helburu nagusiak ..………………………………………………………………………………….……………….. 55
Emaitzak …..……………………………………………………………………………………………………………… 59
Eztabaida ……………………………………………………………………………………………..……………….…. 97
Ondorioak …………………………………………………………………………………………….……………….. 111
I Eranskina (patenteak) ..………………………………………………………………………..………………. 115
II Eranskina (artikuluak) ..……………………………………………………………………..………………... 119
Bibliografia ..……………………………………………………………………………………….…………………. 135
Laburpena
Laburpena
Hezur ehun ingeniaritza, arlo erlatiboki berri eta multi-disziplinarra da eta mindu
edo galdutako ehunen funtzioa mantendu, hobetu eta birsortzeko ingeniaritza eta bizi
zientzien aplikazioan oinarritzen da ordezko biologikoak garatzeko. Ehun ingeniaritzaren
oinarrizko hiru zutabeak aldamioak (scaffold), ama zelulak eta hazkuntza faktoreak dira.
Azken hamarkadetako teknologia aurrerapenekin eta material berri eta azaleren
aldaketekin, hezur ehun ingeniaritzan eta hortz inplantologian erabiltzeko scaffoldek
hobekuntza handiak jasan dituzte. Aldaketa hauen, MSC-en desberdintzapen
osteoblastiko indukzio ezaugarri onak erakutsi dituzte. Gainera, MSC-ek ugalketa handia
eta desberdintzapen gaitasun onak dituztela erakutsi dituzte hezur ehun ingeniaritzan
erabiliak izateko. Hala ere, zein MSC zelula mota den hezur birsorkuntza terapietarako
aukerarik onena oraindik ez dago garbi. Ama zelula autologoen erabileraren arazorik
handiena, zelulen in vitro hazkuntzetan ohikoak diren animali jatorriko serumen
erabilera da, behi suero fetala (BSF) bezala. Ama zelulak txertaturiko pazienteen
erantzun immunea ekiditeko, suero hauen ordezkapena beharrezkoa da.
Ikerketa honetan, giza hortz mamiko ama zelulen (gDPSC) itsaspen, ugalketa,
bideragarritasuna eta desberdintzapen osteoblastiko gaitasuna aztertu dira zabalki
erabilitako Ti6AL4V titanio eta berria den azalera porotsu biomimetikodun (BAS TM)
titanio gainean haztean, desberdintzapen osteoblastiko medioaren presentzian edo
gabezian, plasmatik eratorritako hazkuntza faktoreetan aberatsa den plasma (PRGF) eta
plaketetan aberatsa den fibrina-z (PRF) osaturik.
3
Igor Irastorza Epelde
egun klinika praktikan hortz inplanteen inguruko hezur sorrera bultzatzeko ohikoak
diren fibrina koaguluen erabilera bermatzen du.
Hitz gakoak: hortz mamiko ama zelulak, DPSC, hezur muineko ama zelulak, BMSC, ehun
ingeniaritza, scaffold, zelula desberdintzapena, plasmatik eratorritako produktuak,
PRGF, PRF, titanioa, dezelularizatutako ehun adiposoa, birsortze medikuntza.
4
Laburdurak
Laburdurak
7
Igor Irastorza Epelde
8
Laburdurak
9
Sarrera
Sarrera
13
Igor Irastorza Epelde
Zelulak
Biomaterialak
Autologoak
Naturalak
Heterologoak
Sintetikoak
Desberdinduak
Hidrogelak Ehun Ama zelulak
Meshes
ingeniaritza
Seinaleak
Hazkuntza faktoreak
Molekula txikiak
Indar mekanikoak
1. Irudia. Ehun ingeniaritza hirukotea. Scaffoldetan ereindako zelula amak seinale biofisiko eta kimiko
aproposekin koordinatua ehunen birsortzea bultzatzeko konbinazioa.
1. Zelula amak
Zelula amek, zelula ama bezala deituak izateko, hiru ezaugarri nagusi bete
behar dituzte. Klonalitate, auto-berritze gaitasuna eta ehun eta organo ezberdinetako
zelula helduetara desberdintzeko abilezia (Potten and Loeffler, 1990).
14
Sarrera
Totipotenteak: Mota honetako zelula amek gizabanako osoa sortzeko gaitasuna dute.
Enbrioi oso zein denboraldiko sostengu ehunak (plazenta eta zilbor-hestea) sortzeko
gaitasuna dute. Abilezia hau aldi blastomeriko arte mantentzen da (Posfai et al., 2021).
Oligopotenteak: zelula ama hauek desberdintzapen abilezia oso mugatua dute zelula
mota oso gutxietara desberdindu ahal izanik, adibidez, zelula mieloide eta linfoideak
(Majo et al., 2008).
Unipotenteak: zelula ama hauek zelula mota bakarra sortuko dute, gihar zelula amak
adibidez. Zelula ama gisa ezagutzen zaie auto-berritze gaitasuna dela eta (Seale et al.,
2001).
15
Igor Irastorza Epelde
16
Sarrera
Gorputz helduan MSC populazio ezberdinak deskribatuak izan dira. Hezur muineko
zelula ama mesenkimalak (ingelesez, bone marrow stem cells, BMSCs) izan ziren lehenik
deskribaturiko MSC-ak (Anjos-Afonso and Bonnet, 2007; Friedenstein et al., 1976).
Hauei jarraituz, zilbor hesteko zelula ama mesenkimalak (ingelesez, umbilical cord
mesenchymal stem cells) 1991-n aurkituak eta 2004 urtean baieztatuak MSC bezala
(McElreavey et al., 1991; Wang et al., 2014), hortz mamiko zelula ama mesenkimalak
(ingelesez, dental pulp stem cells, DPSCs) (Gronthos et al., 2000), zelula ama mesenkimal
kardiakoak (Beltrami et al., 2003), biriketako zelula ama mesenkimalak (Griffiths et al.,
2005), odol periferiko-ko zelula ama mesenkimalak (Cao et al., 2005) edo gantz-ehuneko
zelula ama mesenkimalak (ingelesez, Adipose tissue mesenchymal stem cells, AT-MSCs)
(Fraser et al., 2006) deskribatu ziren. Naiz eta lehen deskribaturiko zelula moten jatorria
hozi-geruza mesenkimala izan, aho barrunbeko zelula ama askoren jatorria gandor
neuralean kokatzen da, DPSC-en kasuan bezala. Zelula hauek ama zelula mesenkimal
17
Igor Irastorza Epelde
esmaltea
koroia dentina
pulpa
hortzoia
zementua
sustraia odol
hodiak
lotailu
periodontala
Alboko
kanalak
nerbioa
18
Sarrera
DPSC-ak 2000. urtean deskribatu ziren lehen aldiz hortz mamiko zelula ama
mesenkimal moduan, BMSC-en antzera (Gronthos et al., 2000). Bi urte pasa ziren hortz
mamiko zelula ama gisa deituak izan ziren arte, ezaugarritzat klonogenizitate eta
ugalketa maila altuko zelulak izanik (Gronthos et al., 2002). Zelula hauek “mami
ganbera”-n kokaturik daude, hortz koroiaren barnean (3. irudia). Hortz mamia
konposizio heterogeneoko ehun konektiboa da eta odonto/osteoprogenitore zelula,
zelula neural, baskulatura zelula, fibroblasto eta granulozito eta makrofagoak bezalako
zelula inmuneak bezalako zelula mota ezberdinez osaturik dago (Goldberg and Smith,
2004). Onez gain, garrantzitsua da ikerketa batzuk DPSC-ak perizito baskularraren
konpartimentuan kokatu dituztela esatea (Shi and Gronthos, 2003).
Hortz mami moduko ehun biguinez gain, hortz helduak zementu, dentina eta
esmaltea bezalako ehun gogorrez osaturik daude. Hortzen sorreraren hasiera MSC-en
eta aho zelula ektodermal epitelialen arteko elkarrekintzekin hasten da. Dentina da
sortzen lehena eta honi jarraituz, esmaltea eta hortz folikulua. Zelula epitelialetatik
eratorritako ameloblastoek sortzen dute esmaltea. Beste alde batetik, MSC-ak dira
dentina, zementu, lotailu periodontal eta hortz mamiaren sortzaile (Sedgley and Botero,
2012; Thesleff and Aberg, 1999).
Nahiz eta DPSC-ak izan ikerketan ondoen ezaguturiko eta normalki erabilitako
hortz zelula amak, ezaugarri ezberdinetako zelula ama populazio ezberdinak aurki
ditzakegu. Hortz folikuluko zelula amak (ingelesez, dental follicle stem cells, DFSCs) hortz
garapenean ehun enbrioniko ektomesenkimaletik isolatu daitezke non hortz-hozia
inguratzen hari diren (Zhang et al., 2019). Zelula periodontaletaz gain, beste zelula leinu
batzuetara desberdinduak izan daitezke in vitro (Honda et al., 2010). Hauez gain, papilla
puntako zelula amak (ingelesez, stem cells from apical papilla, SCAP), garapenean
dauden hortzen sustrai erpinetan kokatzen diren hazkuntza ratio handiko zelulak dira
(Bakopoulou et al., 2011). Talde honetako beste zelula amen antzera, beraien jatorri
19
Igor Irastorza Epelde
20
Sarrera
21
Igor Irastorza Epelde
c. DPSC-en desberdintzapena
Lehen aipatu bezala, DPSC-ek jatorri ektomesenkimala dute. Ezaugarri hau dela
eta, osteo/odontoblasto, adipozito, kondrozito eta muskulu zeluletara desberdintzeko
gaitasuna dute (Gronthos et al., 2002; Kawashima, 2012). Honez gain, gandor neural
jatorriari esker, MSC zelulek baina neurona-antzerako zeluletara desberdintzeko
ahalmena handiagoa dute (Luzuriaga et al., 2021). Gainera, ikerketa batzuk
endodermoko zeluletara desberdintzeko abilezia dutela iradoki dute, DPSC-ek
pluripotentzi-antzeko ezaugarria dutela esanez (Atari et al., 2011). Zoritxarrez, naiz eta
endodermo-antzeko zelulak lortu, DPSC-en pluripotentzi gaitasuna ez da eztabaidaezin-
ki frogatua izan.
22
Sarrera
23
Igor Irastorza Epelde
Adipogenikoa Miogenikoa
Kondrogenikoa Neurogenikoa
Osteo/dentinogenikoa
24
Sarrera
Friedenstein eta bere lankideek aurkitu zituzten hezur muineko zelula ama
mesenkimalak 1970-ean, plastikoan itsasten ziren hezur muinetik eratorritako zelulen
isolamendu metodoari esker (Friedenstein et al., 1970). Zelula hauek plastikoa hasten
direnean, ugaritze ratio handiko kolonia konpaktu gisa fibroblasto itxurarekin. Hauek
kolonia osatzaile unitate fibroblastoak gisa izendatu ziren (ingelesez, colony forming unit
fibroblast, CFU-F) (Castro-Malaspina et al., 1980; Gothard et al., 2013).
25
Igor Irastorza Epelde
26
Sarrera
c. BMSC-en desberdintzapena
Hauez gain, BMSC-ak ere zelula kardiako, gihar eskeletiko, lotailu eta estroma
hematopoietiko-laguntzailea bezalako zelula mesenkimaletara ere desberdin daitezke
(Guo et al., 2018; Matsuda et al., 2005; Tian et al., 2010; Wu et al., 2018) (6. irudia).
27
Igor Irastorza Epelde
6. irudia. Hezur muineko zelula amen desberdintzapen gaitasuna. (Muruganandan et al., 2009).
28
Sarrera
Hezur ehun ingeniaritzan, scaffoldek hiru ezaugarri bete behar dituzte hezur
injerto bezala erabiliak izateko: osteo-konduktibitatea, osteo-induktibitatea eta osteo-
integrazioa (Albrektsson and Johansson, 2001). Injertoan hezurra hazi baldin badaiteke
osteo-konduktibitate deitzen zaio, inplantearen azalera eta hezurraren arteko kontaktu
zuzenari berriz osteo-integrazioa. Ezaugarri hauek ezberdinak izan daitezke
materialaren arabera, inplante lekuaren arabera, etab. (Khan et al., 2005). Azkenik,
osteo-induktibitate gisa ezagutzen da hezur sorrera eragin dezaketen zeluletan
desberdintzeko ama zelula multipotenteei bultzatzeko gaitasunari (Roberts and
Rosenbaum, 2012).
29
Igor Irastorza Epelde
7. irudia. Hezur ehun ingeniaritza. Hezur scaffold-a eta zelula ama autologoak bioerreaktorean hazkuntza
faktoreekin haziak hezur autologoaren sorrera bultzatzen dute in vitro, hezur konponketa
aplikazioetarako in vivo.
30
Sarrera
2.1. Titanioa
31
Igor Irastorza Epelde
Nahiz eta hortz inplanteetarako aukerarik hoberena titanioa dela ikusi den arren,
azken hamarkadetan, inplante azalera ezberdinen diseinu eta fabrikazioak osteo-
integrazioa hobetzean duen garrantzia frogatu da (Gasik et al., 2015; Rani et al., 2012;
Rupp et al., 2018; Salou et al., 2015). Titanioaren konposizioaz gain, garrantzitsua da
azalera hauen topografia kontuan hartzea inplantearen ainguraketa iraunkorra lortzeko
(Annunziata and Guida, 2015; Jemat et al., 2015; Le Guéhennec et al., 2007; Naves et
al., 2015).
Lehen aipatu bezala, ikerketa ezberdinek frogatu duten bezala, poro tamaina
txikiek (<180 µm) zelulen desberdintzapena bultzatzen dute inplantearen sendaketaren
hasieran, aldi berean, tamaina handiko poroek (>300 µm) zelulen ugalketa eta hezur
sorrera bultzatzen dute (Coelho et al., 2015). Hala ere, porositateak materialen ezaugarri
mekanikoetan eragin zuzena du, beraz, ezinbestekoa da oreka egoki bat mantentzea
porositate eta gogortasunaren artean inplante motaren arabera. Hare gehiago, beste
ikerketa batzuk erakutsi dutenez, titanio porotsuek korrosioaren aurkako erresistentzia
txikiagoa dute gorputzeko fluidoen fluxu dinamikoaren ondorioz. Porositateak
gorputzeko fluidoen fluxua errazten du, degradazioa azkartuz (Chen et al., 2017).
32
Sarrera
1- A fase egonkortzaileak: karbono (C), oxigeno (O), aluminio (Al) eta nitrogenoa
(N).
2- B fase egonkortzaileak:
a. Isomorfoak: tantalo (Ta), molibdeno (Mo), banadio (V) eta niobioa (Nb).
b. Euktoideak: silikona (Si), kromo (Cr), kobre (Cu), hidrogeno (H),
manganeso (Mn), nikel (Ni) eta burdina (Fe).
3- Gehikuntza neutroak: estainu (Sn) eta zirkonioa (Zr).
33
Igor Irastorza Epelde
Gaur egun erabiltzen diren hortz inplante gehienak metal aleazio inerteak dira.
Inplante hauek bakarrik erabiliz edota dezelularizatutako hezur matrize eta plasmatik
eratorritako hazkuntza faktoreekin konbinatuz, ama zelula mesenkimalenosteo-
desberdintzapena bultzatzen dute, inplante azaleran hezurra sortuz (Anitua et al.,
2016b; Boyan et al., 2016a; Mayer et al., 2018; Zanicotti et al., 2018). Gaur egun, hortz
inplanteen arazorik nagusiena periinplantitisaren garapena da. Periinplantitisa,
murtxikatze naturalean inplanteek hezur ehunari transferitzen dion presio mekaniko
jarraiaren ondorioz sortzen da. Kasu okerrenetan, periinplantitisak inplantearen galera
eragiten du (Bertolini et al., 2019). Hortz naturaletan, arazo hau lotailu periodontalari
(PDL) esker konpontzen da, inerbazio eta baskularizazio altuko ehun konektiboa da,
kolageno sorta indartsuez osaturik dagoena. Kolageno fibra sorta hauek hortz sustrai
zementu eta hezur albeolarrari ainguraturik daude kaltzifikazio arineko ertzei esker.
Fibra hauei Sharpey fibra zulatzaileak bezala ezagutzen zaie (Aaron, 2012). PDL-aren
funtzio nagusia mastekatzean sortzen den indarra xurgatzea da kuxin bezala jokatuz,
honela hezur albeolarra babestuz
34
Sarrera
9. irudia. Irudi makroskopiko (A, B) eta ekortzeko mikroskopio elektronikoko irudiak (SEM) (C, D),
txerrien ehun adiposo ez manipulatua (A, C) eta dezelularizaturiko matrize extrazelularra (ECM) (B, D).
Eskala barra: 50 µm. (Choi et al., 2012).
3. Hazkuntza faktoreak
35
Igor Irastorza Epelde
Nahiz eta PRP eta PRGF-aren prestaketa oso antzekoa den arren, PRGF-ak plasma
proteina eta koagulazio faktore gehiago ditu, efektu handiagoa eraginez PRP-arekin
alderatuz (Anitua, 1999).
36
Sarrera
37
Igor Irastorza Epelde
Gaur egun, hazkuntza faktoreetan aberatsa den plasma (PRGF) eta plaketetan
aberatsa den fibrinaren (PRF) erabilera klinikoa zabaldua izan da hortz erauzketa osteko
titanio inplanteen inguruko hezurra sendatzeko. Produktu hauei plasmatik eratorritako
terapia medikorako produktu aurreratu (ingelesez, plasma-derived advanced medical
therapy products, AMTP) bezala ezagutzen zaie (Giannini et al., 2015; Kobayashi et al.,
2016; Nishiyama et al., 2016).
38
Metodologia
Metodologia
Giza hortz mamiko zelula amak (gDPSC), hortz klinikan paziente gazte
osasuntsuetatik (18-tik 30 urte bitartekoak) erauzitako hirugarren molarretatik isolatu
ziren. Pazienteek baimen informatua sinatu zuten aldez aurretik onartutako
M10_2016_088 protokoloa jarraituz (Euskal Herriko Unibertsitateko etika batzordea).
41
Igor Irastorza Epelde
eta % 1 L-Glutamina-z (Sigma, MO, AEB) osaturiko DMEM medioan erein ziren 37 °C-tan
% 5-eko CO2-arekin. 5 80-ko konfluentzia lortu ostean zelulak 75 zm2-ko flasketara
(Sarstedt, Nümbrecht, Alemania) pasa ziren. Esperimentuetarako 4 eta 10 pase
bitarteko zelulak erabili ziren.
2 mm-ko lodiera duten Ti6AL4V diskoak, 5.5 mm-ko diametroa duen barra bat
moztuz lortu ziren Avinent Implant System SLU (Bartzelona, Espainia) enpresatik. BASTM
titanio azaleraren fabrikaziorako, titanio diskoaren aurpegi bat korindoi zuriz (Al2O3)
F60 eginiko “shot blastin” teknika erabili zen, 212-300 µm-ko tamainako partikulak
proiektatuz. Ondoren, diskoak bainu ultrasoniko batean garbitu ziren 10 minutuz eta ur
distilatuz garbitu ziren. Garbiketen ostean, anodizatuak izan ziren DC energia iturriko
anodoari lotuz eta kaltzio eta fosforoa erabiliz ur soluzioko elektrolito gisa. 0.75
mA/mm2-eko korronte dentsitatea aplikatu zen eta potentzialari libreki handitzen utzi
zitzaion 130 V-raino heldu arte. Azkenik, bainu ultrasonikoan ur destilatuzko 10
minutuko garbiketekin bukatu zen fabrikazio prozesua. Ti6Al4V titaniozko diskoek ez
zuten inolako leuntze edo tratamendurik jasan.
42
Metodologia
Txerrietatik eratorritako ehun adiposoa tokiko janari enpresa batetik (Jaucha SL,
Nafarroa, Espainia) lortu zen. Ehuna gela tenperaturan desizoztu, garbitu eta irabiagailu
batez krematua izan zen. Ondoren, ehuna izotzetan homogeneizatu zen bi hagaxka
ezberdinezko Plyton-a (PT3100) erabiliz 1200 rpm-etan 5 minutuz. Lipidoen fase
separazioa lortzeko, homogeneizaturiko ehuna 900 g-etan zentrifugatu zen ur ultra
purua erabiliz 5 minutuz. Lipidoak eskuz baztertuak izan ziren eta proteinak
kontserbatuak. Proteina pelletak gauan zehar isopropanolez (Merk Life Science,
Espainia) tratatuak izan ziren astintzaile orbital batez gela tenperaturan. PBS-ez garbitu
ondoren, % 1 Triton X-100 eta % 0.1 amonio hidroxido-z (Merk Life Science, Espainia)
tratatu zen 36 orduz gela tenperaturan astintzaile orbitalean. PBS-ezko garbiketen
ostean, liofilizatua izan zen erabat lehortu arte. Lorturiko ehuna fresatu egin zen
nahasgailu errota (Retsch MM400) erabiliz prozesatuak izateko grano fineko hautsa
lortu arte. Lorturiko hautsa nitrogeno likidoz izoztu zen eta 4 °C-tan gorde zen hutseko
lehorgailuan.
PRGF-a prestatzeko, giza emaile gazte osasuntsuetatik lortu zen odola % 3.8
sodio zitrato antikoagulatzailea zuten 5 ml-ko odola biltzeko hodiak (BD Vacutainer,
Plymouth, UK) erabiliz. 8 minutuz 580 g-etara zentrifugatu ondoren, plaketetan aberatsa
den frakzioa 15 ml-lo falkon hodietara pasa zen. Plaketak, % 10 kaltzio kloruroz (Braun
43
Igor Irastorza Epelde
medical, Melsungen, Alemania) aktibatuak izan ziren ordu batez 37 °C-etan. Inkubazio
honen ostean, plasma 3000 g-etan zentrifugatu zen 15 minutuz 4 °C-tan eta filtratua izan
zen 0.2 µm-ko poro tamainako iragazkiak erabiliz (Sarstedt, Nümbrecht, Alemania).
Azkenik, plasma alikuotatu eta izoztuta gorde zen -20 °C-tan erabili arte. PRGF-a medio
aldaketa bakoitzarekin gehitu zen (10. irudia).
Antikoagulatzailea CaCl2
bidezko
Odol plaketa
zentrifugazioa aktibazioa
PRGF
PRF
Plasma
Odol Kapa leukozitarioa
erauzketa Eritrozitoak Fibrina
koaguluaren
Antikoagulatzailerik formazioa Konpresio
gabe tresna
10. irudia. PRGF eta PRF-aren prestakuntza ilustrazioa. (Autoreak sortua).
44
Metodologia
areagotzeko, 15.000 zelula 70 µl-ko tantetan erein ziren diskoen gainean eta 3 orduz 37
°C-etan inkubatu ziren. Inkubazio honen ostean 700 µl DMEM medio gehitu ziren 24
putzuko plakentzako eta 4 ml 6 putzuko plakentzako.
4 eguneko hazkundearen ostean, zelulak PBS-z garbitu ziren. Garbiketak egin eta
gero, zelulak 3 µM kaltzeina-AM (Molecular probes, OR, AEB) eta 2.5 µM propidio
ioduro-arekin (Sigma, MO, AEB) inkubatu ziren 30 minutuz 37 °C-tan. Azkenik, zelulei 3
PBS garbiketa egin zitzaizkien eta argazkiak Axioskop mikroskopio fluoreszentea (Zeiss,
Overkochen, Alemania) eta Nikon DS-Qi1 kamera erabiliz (Nikon, Tokio, Japonia) atera
ziren.
Immunozitokimika
45
Igor Irastorza Epelde
Cambridge, UK). Alexa 488 conjugated rabbit IgG (A11008, Abcam, Cambridge, UK)
erabili zen antigorputz sekundario gisa antigorputz primarioak lokalizatzeko %1 BSA/
PBS erabiliz ordubetez gela tenperaturan. Zelulen nukleoak 4’,6-diamino-2-fenilindol-ez
(DAPI, Invitrogen, CA, AEB) tindatu ziren. Irudiak, Axioskop mikroskopio fluoreszentea
(Zeiss, Overkochen, Alemania) eta Nikon DS-Qi1 kamera erabiliz (Nikon, Tokio, Japonia)
eta Leica DM6000 B mikorskopioa (Leica, Wetzlar, Alemania), Leica DFC420 C kamera
eta 3.3.3.16958 Leica application suite X (Leica, Wetzlar, Alemania) programaren bidez
atera ziren.
Fluxu zitometria
46
Metodologia
Zelula peletak -80 °C-tan gorde ziren erabiliak izan arte. RNA, RNeasy mini kit-a
(Qiagen, Hilden Alemania) erabiliz erauzi zen. RNA-ren kontzentrazio eta purutasuna
260/ 280 nm-ko absorbantziaz neurtu zen Nanodrop Synergy HT-ren bidez (Biotek, VT,
AEB). cDNA-ren sintesia erretrotranskripzioz lortu zen 1000 ng-ko hasierako RNA-tik
iScript cDNA kit-a erabiliz (Biorad, CA, AEB). Retrotranskripzio zikloak 25 °C-tako 5
minutuko pausoarekin hasi zen, honi jarraituz 46 °C-ko 20 minutu zikloa eta amaitzeko
minutu bateko 95 °C-ko zikloarekin. Behin erretrotranskripzioa amaiturik 4 °C-tan
mantendu ziren -20 °C-tan izoztu arte.
RNA sekuentziazioa
gDPSC eta gBMSC-ak plastiko, Ti6Al4V eta BAS titanio azaleretan medio kontrol
edo osteoblastikoarekin hazi ziren 14 egunez. 24 disko erabili ziren kondizio bakoitzeko
eta tripsinarekin zelulak desitsatsi ostean, RNA Quiagen RNA erauzketa kitarekin lortu
zen. gDPSC eta gBMSC-etatik erauzitako RNA Zuritxeko Genomika zentroan sekuentziatu
zen Illumina sekuentziagailuaz (Illumina, CA, AEB), 25.000 RNA irakurketa lortuz
baldintza bakoitzeko. Gene hauen adierazpena presentzia/gabezia (ON/OFF) kriterioa
jarraituz analizatu zen baldintza ezberdinen konparaketetan eta 10 irakurketa
gordinetan jarri zen muga gene baten adierazpena positibotzat (ON) hartzeko.
Aktibaturiko gene bideak “Pathway Enrichment Analysis” erabiliz analizatu zen, “The
47
Igor Irastorza Epelde
Connectivity Map” (CMAP) eta “Gene Ontology enrichment Biological Process” (GOBP)
datu baseak erabiliz CIC bioGUNE-n (CIC bioGUNE, Derio, Espainia).
Power SYBR Green PCR Master Mix-a (Applied biosystems, CA, AEB) erabili zen
qPCR-a egiteko. Anplifikatzeko programaren zikloak 10 minutu 95 °C-tan eta 95 °C 20
segundo eta 59 °C-ko minutu bateko 40 ziklorekin egin zen. Erabili ziren “etxezaintza”
geneak β-aktina 5´-GTTGTCGACGACGAGCG-3´ eta 5´-GCACAGAGCCTCGCCTT-3´ eta
Gapdh 5´ CTTTTGCGTCGCCAG -3´ eta 5´- TTGATGGCAACAATATCCAC -3´ izan ziren.
Azterturiko geneak hauek izan ziren: kolageno I (COL I) 5´ -GGCCCCCTGGTATGACTGGCT-
3´ eta 5´-CGCCACGGGGACCACGAATC-3´, osteonektina (SPARC) 5´-
GAAAGAAGATCCAGGCCCTC-3´ eta 5´-CTTCAGACTGCCCGGAGA-3´, osteokaltzina
(BGLAP) 5´-CGCCTGGGTCTCTTCACTAC-3´ eta 5´-CTCACACTCCTCGCCCTATT-3´, dentin
sialofosfoproteina (DSPP) 5´-TGCCCAAATGCAAAAATATG-3´ eta 5´-
GTGGGCCACTTTCAGTCTTC-3´, osterix (OSX) 5´-TGAGGAGGAAGTTCACTATG-3´ eta 5´-
CATTAGTGCTTGTAAAGGGG-3´eta runx2 (RUNX2) 5´-CACTCACTACCACACCTACC-3´ eta
5´-TTCCATCAGCGTCAACAC-3´. Ikerturiko geneen adierazpena etxezaintza geneen
adierazpenarekin normalizatuak izan ziren.
Zelulak minutu batez % 4 PFA-az fijatu ondoren 3 aldiz garbitu ziren % 0.05 Tween
20-PBS-z. ALP tindaketarako, 5-bromo-4-kloro-3-indolil fosfato/ tetrazolio nitro urdina
(NBT/BCIP, Sigma, MO, AEB) erabili zen eta tindaketa 3 minuturo begiratu zen. Tindaketa
amaitu ondoren, PBS-zko 5 minutuko 3 garbiketa egin ziren. Irudiak, Zeiss Stemi 2000-C
mikroskopio estereoskopikoarekin (Zeiss, Alemania) atera ziren Canon PowerShot A80
kamera (Canon, Tokio, Japonia) erabiliz. ALP aktibitate entzimatikoa kuantifikatzeko,
zelulak tripsina-EDTA-rekin desitsatsi ziren. Absorbantzia 420 nm-tan neurtu zen
Synergy HT-ren bidez (BioTek, VT, AEB) Gen5 1.11 programa erabilita.
48
Metodologia
Analisi estatistikoa
49
Hipotesia
Hipotesia
53
Helburu nagusiak
Helburu nagusiak
Helburu zehatzak
57
Emaitzak
Emaitzak
Giza hortz muineko zelula amak (gDPSC) hirugarren haginetatik isolatu ziren eta
behi serum fetal (% 10), penizilina/estreptomizina (% 1) eta L-Glutamina-z (% 1)
osaturiko DMEM medioan hazi ziren. 2-3 aste eta gero, zelulek kolonia atxikitu anitzak
eratu zituzten, sub-konfluentzia lortuz. gDPSC-ek fibroblasto itxurako morfologia
erakusten dute hazkuntza kultiboetan; zelulek ezaugarri mantentzen dute ugaltze maila
altuarekin batera pase askotan (12. irudia).
A B
D E
12. irudia. gDPSC-en in vitro hazkuntza. gDPSC hazkuntzen fase kontraste mikroskopia irudiak egoera
sub-konfluente (A, D) eta konfluentean (B, E) handipen txiki eta handiarekon. (Irastorza et al., 2019)-tik
moldatua.
61
Igor Irastorza Epelde
CD 45 A B CD 73
3.0% 100.0%
CD 90 D E CD 105
100.0% 99.9%
13. irudia. Fluxu zitometria bidezko gDPSCen ama zelula markatzaileak. Fluxu zitometriak erakutsi
duenez gDPSC-ak negatiboak ziren CD45 markatzaile hematopoietikoarentzat (A) eta positiboak CD73,
CD90 eta CD105 (B, D eta E) zelula ama mesenkimal markatzaileentzat. (Irastorza et al., 2019)-etik
moldatua.
Ekortze mikroskopio elektronikozko (SEM) Ti6AL4V eta BASTM titanio azaleren mikro-
topografia analisia
Zelulak gabeko titanio azaleren SEM irudiak (Ti6AL4V) zirrikitu paralelo kontzentrikoak
erakutsiz (14. irudia A, B), bestalde, titanio azalera latza (BASTM) hobi eta uniformeki
sakabanaturiko mikro-poro txikiak (< 10 µm) erakutsiz azalera guztian zehar (14. irudia
D, E).
62
Emaitzak
A B
Ti6AL4V
D E
BAS®
14. irudia. Ti6AL4V eta BAS titanio azaleren SEM irudiak. Ti6AL4V (A, B) eta BAS (D, E) titanio azaleren
ekortze mikroskopio elektronikoaren irudiak handipen txiki eta handiarekin. (Irastorza et al., 2019)-tik
moldatua.
gDPSC-ak Ti6AL4V eta BASTM titanio azaleretan erein ziren 20,00 zelula/diskoko
dentsitatean 4 egunez. Zelulak sendoki atxikitu eta zabaldu ziren Ti6AL4V titanio
leunaren azaleran zehar. Zelulak, diskoaren zirrikitu kontzentrikoak jarraituz orientaturik
hazi ziren eta lamelipodio gisako zelula luzaketak erakutsi zituzten (15. irudia A).
Bestalde, BAS titanio azaleran hazitako gDPSC-ak beraien morfologia moldatu behar izan
zuten azalera porotsuan barneratzeko (15. irudia B). bi titanio azaleretan, zelulen
atxikimendua hazkuntza denbora guztian zehar mantendu zen.
63
Igor Irastorza Epelde
A D F
Ti6AL4V
B E G
BAS®
15. irudia. Titanio azaleretan hazitako gDPSC-en bideragarritasun, orientazio, mobilitate eta SEM
irudiak. Ekortze mikroskopio elektronikoaren gDPSC-ak ereindako bi titanio azaleren irudiak (A, B). 4
egunez bi titanio azaleretan hazitako gDPSC-en fluoreszentzia irudiak bizirik dauden zelula (berde) eta
hildakoak (gorri) erakutsiz (D, E). Eskala barra: 100 µm. Nukleoak DAPI-z urdinez tindaturiko gDPSC-ak,
titanio disko osoko mosaiko fluoreszentzia irudietan. Ti6AL4V (F) eta BAS (G). Eskala barra: 2 mm.
(Irastorza et al., 2019)-tik moldatua.
Ti6AL4V eta BASTM titanio azaleretan hazitako gDPSC-en ugaritzearen azterketa PRGF-
arekin konbinatzean.
64
Emaitzak
zeuden, gehienak metasafe eta anafasean. Emaitza hauek, gDPSC-en ugaltze normala
erakusti zuten. Hala eta guztiz ere, gDPSC-ak PRGF solublearekin haztean, ugalketa
esanguratsuki areagotua zuten (% 20-tik % 50-raino; p < 0.05) 4 egunek hazkundearen
ostean kontrolekin alderatuta (16. Irudia G). Emaitza hauek erakusten dutenez, gDPSC-
ak plasmatik eratorritako osagarriekin titanio azaleretan haztean, zelula ugalketa
areagotuta dute. Emaitzan berresteko, Ki67 immunofluoreszentzi entsegua egin zen.
Ki67-arekin tindaturiko gDPSC-ak, bi titanio azaleretan PRGF-arekin haziak, kontatuak
izan ziren, berriz ere, ugaltzen hari diren zelula kopurua areagotua dagoela erakutsiz (16.
irudia A-F).
Kontrola PRGF ®
A B
Ti6AL4V
Ki67/ DAPI
D E
BAS®
1,5 1,5
1 1
0,5 0,5
0 0
Control Ti/PRGF Ti Control BAS/PRGF BAS Control Ti/PRGF Ti Control BAS/PRGF BAS
65
Igor Irastorza Epelde
16. irudia. Titanio azaleren gainean PRGF-arekon hazitako gDPSC-en ugalketa entsegua. Ki67 ugalketa
markatzaile (berde) eta DAPI-z nukleoak markaturiko (urdin) gDPSC-en immunofluoreszentzi irudiak
baldintza kontroletan Ti6AL4V (A) eta BAS (D) titanio diskoetan, eta % 20 PRGF-rekin haziak Ti6AL4V (B)
eta BAS (E) diskoetan 4 egunez. PRGF tratamenduak Ki67+ zelulen kopurua handitu zuen. Barra eskala: 50
µm. Titanio azaleren gainean kontrol eta PRGF baldintzetan mitosian dauden zelulen (Ki67+) arteko
konparaketa (F) eta nukleo kopuru totalen konparaketa (DAPI; G). estatistikoki esanguratsua p < 0.05
denean. (Irastorza et al., 2019)-tik moldatua.
66
Emaitzak
10
B 9
8
7
ALP absorbantzia
normalizatua
6
5
4
*
3
2
1
0
CL GL PL CDL GDL PDL CR GR PR CDR GDR PDR
Ti6AL4V BAS
67
Igor Irastorza Epelde
17. irudia. Bi titanio azaleretan, desberdintzapen osteoblastiko medioaren presentzia edo absentzian
eta plasmatik eratorritako produktuekin (PRGF eta PRF) hazitako gDPSC-en fosfatasa alkalino entsegua.
14 egunez Ti6AL4V eta BAS titanio diskoetan, desberdintzapen osteoblastikoaren presentzian edo
gabezian hazitako gDPSC-en fosfatasa alkalino tindaketa irudiak (A). ALP aktibitatearen kuantifikazio
normalizatua erakusten duen grafiak (B). akronimoen esanahia: C (kontrola), G (PRGF), P (PRF), D
(desberdintzapen medioa), L (Ti6AL4V titanioa) era R (BAS titanioa). Estatistikoki esanguratsua (*) p< 0.05
denean. Eskala barra: 1 mm. (Irastorza et al., 2019)-tik moldatua.
68
Emaitzak
Kontrola Desberdintzapena
A B
D E
Ti6AL4V
F G
H I
BAS®
J K
69
Igor Irastorza Epelde
18. irudia. Ti6AL4V eta BAS titanio azaleretan hazitako gDPSC-en alizarin gorri tindaketa. 14 egunez,
baldintza kontroletan alizarin gorriz tindatutako hezur matrize gordailuak plastikoan (A), Ti6AL4V (D, F)
eta BAS-ean (I, F) haziak, desberdintzapen osteoblastikoarekin trataturiko plastiko (B), Ti6AL4V (E, G) eta
BAS-etan (J, K) hazitako gDPSC-ekin konparatuta. Eskala barra (A, B): 100 µm; (D, E, H eta I): 2 mm; (F, G,
J eta K): 0.5 mm. (Irastorza et al., 2019)-tik moldatua.
70
Emaitzak
2,5
ARS absorbantzia normalizatua
*
2
1,5
*
0,5
0
CR GR PR CDR GDR PDR
19. irudia. PRGF eta PRF-arekin BAS titanio azalera gainean hazitako gDPSC-en alizarin gorri tindaketa
eta kuantifikazio fotometrikoa. Desberdintzapen eta control medioak eta PRGF eta PRF-rekin BAS titanio
azalera gainean hazitako gDPSC-en alizarin gorri tindaketa 21 egunen ostean (A). Alizarin gorriaren
kuantifikazio fotometriko normalizatua (B). Akronimoen esanahia: C (kontrola), G (PRGF), P (PRF) eta D
(desberdintzapen medioa). Estatistikoki esanguratsua (*) p< 0.05 denean. Eskala barra: 250 µm. (Irastorza
et al., 2019)-tik moldatua.
Ti6AL4V eta BASTM titanio azaleretan PRGF eta PRF-rekin edo hauen gabezian hazitako
gDPSC-en desberdintzapen markatzaileen azterketa RT-QPCR-bidez
71
Igor Irastorza Epelde
72
Emaitzak
Kolageno I RunX2
A B
* *
3,5
mRNA Fold Change
D SPARC Osterix
** E
16 ** 35
*
mRNA Fold Change
20. irudia. Ti6AL4V eta BASTM titanio azaleretan, PRGF eta PRF-arekin edo haue gabezian hazitako
gDPSC-en desberdintzapen osteoblastikoaren gene adierazpenen QPCR-a. 14 eguneko hazkuntza eta
gero eginiko Kolageno I, RUNX2, SPARC eta OSTERIX-en mRNA adierazpen normalizatua. Estatistikoki
esanguratsua (*p< 0.05) eta (**p< 0.01) denean. Akronimoen esanahia: C (kontrola), D (desberdintzapen
medioa), L (Ti6AL4V), R (BAS), G (PRGF) eta P (PRF). (Irastorza et al., 2019)-tik moldatua.
73
Igor Irastorza Epelde
48H
BMSC
DPSC
21. irudia. 24 eta 48 orduz kristal, Ti6AL4V eta BAS titanio azaleren gainean hazitako gDPSC eta gBMSC-
en Kaspasa 3 heriotza zelular markatzailearen immunofluoreszentzia irudiak. Kristal, Ti6AL4V eta BAS
titanio azaleretan hazitako gDPSC eta gBMSC-en Kaspasa 3 heriotza zelular markatzaile (berde) eta DAPI
tindatzaile nuklearraren (urdin) immunofluoreszentzi irudiak 24 eta 48 orduren ostean. Bi titanio azalerek
eragin zitotoxikorik ez zutela erakutsi zuten irudiek, bi zelula motetan Kaspasa 3 positibo diren zelula
bakarrak aurkituz titanio disko osoan. Eskala barra: 100 µm.
74
Emaitzak
Ti6AL4C eta BASTM titanio azaleretan 24 eta 48 orduz hazitako gDPSC eta gBMSC-en
arteko ugalketa konparazio ikerketa
Kristal, Ti6AL4V eta BAS titanio diskoen gainean 24 eta 48 orduz hazitako gDPSC
eta gBMSC-en arteko ugaltze ezberdintasunak ikertzeko, immunofluoreszentzi ikerketa
egin zen zelula ugalketaren markatzaile den Ki67 (berde) eta DAPI nukleo markatzailea
(urdin) antzemateko. Titanio diskoen opakotasunaren ondorioz, zelula kopuru totala
(DAPI+) eta zelula proliferatzaile kopuru totala (Ki67+), eskuz kontatuz lortu ziren.
Espero bezala, bi zelula moten gehiengoak interfasean ageri ziren, hala ere,
ezberdintasun interesgarriak aurkitu ziren kristal gainean hazitako gDPSC eta gBMSC-en
artean, eta baita titanio azaleren gainean hazitakoen artean ere (22. irudia A). Ki67/DAPI
normalizatuaren grafiketan ikus zitzkenez, 24 orduren ondoren ezberdintasun
esanguratsuak daude gDPSC eta gBMSC-en ugalketa proportzioen artean kristal
gainean, gDPSC-ek izanik ugaltze mailarik handiena. Proportzio hau ere handiagoa zen
gDPSC-etan Ti6AL4V eta BAS titanio diskoetan haztean ere, zertxobait handiagoa izanik
Ti6AL4V titanioan bi zelula motetan BAS titanioan baino. 48 orduko hazkundearen
ostean, gDPSC eta gBMSC-en ugalketa maila berdindu egin zela ikusi zen kristalean eta
ez zen ezberdintasun estatistikorik aurkitu. Dena dela, Ti6AL4V titanioan, gDPSC-ek
gBMSC-ak baina ugalketa maila handipen esanguratsua zutela frogatu zuten. Azkenik,
BAS titanio azalerak ere emaitza interesgarriak erakutsi zituen, gDPSC-en ugalketa maila
gBMSC-ena baino handiagoa izanik estatistikoki. Gainera, hDPSC-en ugalketa maila
kontrol baldintzarenarekin parekatzea lortu zuten. Bestalde, gBMSC-ek BAS titanioaren
gainean, 24 ordutan baino ugaltze maila txikiagoa erakutsi zuten, esperimentu osoko
mailarik txikiena erakutsiz (22. irudia B).
75
Igor Irastorza Epelde
24H
DPSC
BMSC
48H
DPSC
B Ki67/DAPI Ki67/DAPI
* 24H 48H *
2,5 1,4
Ki67/DAPI normalizatua
Ki67/DAPI normalizatua
1,2
2 *
1
1,5 0,8
1 0,6
0,4
0,5
0,2
0 0
Coverslips Ti6AL4V BAS Coverslips Ti6AL4V BAS
BMSC DPSC
76
Emaitzak
22. irudia. Kristal, Ti6AL4V eta BAS titanio azaleretan 24 eta 48 orduz hazitako gDPSC eta gBMSC-en
arteko ugaritze ikerketa. Immunofluoreszentzia mikroskopio bidez ateratako gDPSC eta gBMSC-en Ki67
ugalketa markatzaile (berde) eta DAPI nukleo markatzaileen (urdin) irudiak 24 eta 48 orduren ostean (A).
azalera ezberdinetan hazitako bi zelula moten ugalketa ratioaren grafika (B). Eskala barra: 100 µm.
Estatistikoki esanguratsua (*p< 0.05) denean.
77
Igor Irastorza Epelde
Kontrola Desberdintzapena
Ti6AL4V
BMSC
BAS
Ti6AL4V
DPSC
BAS
23. irudia. Ti6AL4V eta BASTM titanio azaleren gainean desberdintzapen osteoblastiko medioaren
presentzian edo gabezian hazitako gDPSC eta gBMSC-en SPARC desberdintzapen osteoblastiko
markatzailearen immunofluorezentzia irudiak. Bi titanio azaleretan hazi ziren gDPSC eta gBMSC-ak
desberdintzapen osteoblastiko medioaren presentzian eta gabezian 14 egunez. Fijaketaren ondoren,
SAPRC desberdintzapen osteoblastikoaren markatzailea (berde) eta DAPI-z zelulen nukleoak (urdin)
tindatu ziren. Eskala barra: 100 µm.
78
Emaitzak
Kontrola Desberdintzapena
Ti6AL4V
BMSC
BAS
Ti6AL4V
DPSC
BAS
24. irudia. Ti6AL4V eta BASTM titanio azaleren gainean desberdintzapen osteoblastiko medioaren
presentzian edo gabezian hazitako gDPSC eta gBMSC-en Osterix desberdintzapen osteoblastiko
markatzailearen immunofluorezentzia irudiak. Bi titanio azaleretan hazi ziren gDPSC eta gBMSC-ak
desberdintzapen osteoblastiko medioaren presentzian eta gabezian 14 egunez. Fijaketaren ondoren,
Osterix desberdintzapen osteoblastikoaren markatzailea (berde) eta DAPI-z zelulen nukleoak (urdin)
tindatu ziren. Eskala barra: 100 µm.
79
Igor Irastorza Epelde
Ti6AL4V eta BASTM titanio azaleretan hazitako gDPSC eta gBMSC-en mineralizaturiko
hezur matrize gordailuen konparaketa Alizarina gorriaren bidez
3,5
2,5 BMSC
2 DPSC
1,5
0,5
80
Emaitzak
25. irudia. Ti6AL4V eta BASTM titanio azaleretan hazitako gDPSC eta gBMSC-en mineralizaturiko hezur
matrize gordailuen Alizarina gorriaren absorbantzia grafikoa. 21 egunez hazi ondoren, hezur matrize
extrazelular gordailuak Alizarin gorriz tindatu ziren, honen ostean, Alizarina azido azetikoan disolbatu zen
eta plaka irakurle batez neurtu zen absorbantzia.
81
Igor Irastorza Epelde
2. taula. Baldintza batean adierazita eta bestean adierazi gabe dauden geneen kantitateak erakusten
dituen taula. Adierazitako geneen kantitate patroi ezberdinak aurkitu ziren baldintza ezberdinak
alderatzean. Laburdurak: DPSC: hortz mamiko zelula amak; BMSC: hezur muineko zelula amak; C: kontrol
medioa; T: desberdintzapen osteoblastiko medioa; Flask: plastiko azalera; TI: Ti6AL4V titanioa eta BAS:
“Biomimetic Advanced Surface” titanioa.
82
Emaitzak
83
Igor Irastorza Epelde
84
Emaitzak
hours after
transduction UP
TAKEDA_NUP8_HO 118 4 0.00178462 HOXA3, HOXA5, effects of NUP98-
XA9_8D_UP HOXA7, HOXB3 HOXA9 on gene
transcription at 8
days after
transduction UP
TAKEDA_NUP8_HO 147 4 0.00369089 HOXA3, HOXA5, effects of NUP98-
XA9_10D_UP HOXA7, HOXB3 HOXA9 on gene
transcription at 10
days after
transduction UP
DPSC-C- BASSO_GERMINAL 93 3 0.0191607 BATF, BCL2A1, Gene up-regulated
FLASK _CENTER_CD40_UP HLA-DQB1 by CD40 signaling
VS in Ramos cells
DPSC-T-
FLASK
CMV_HCMV_TIME 25 2 0.0377068 RSAD2, TNFSF10 Genes up-regulated
COURSE_12HRS_U after infection with
P HCMV at 12 h
VERHAAK_AML_NP 173 3 0.040305 BCL2A1, Genes up-regulated
M1_MUT_VS_WT_ SERPINA1, in acute myeloid
UP TNFSF10 leukemia (AML)
CARIES_PULP_UP 200 3 0.0462674 BCL2A1, HLA- Immune/cytokine
DQB1, SERPINA1 response
4. taula. Gene bideen aberastasun analisia “The Connectivity Map”-ez (CMAP) egina. Plastiko eta titanio
azaleren gainean desberdintzapen osteoblastiko medioaren presentzian edo gabezia hazitako gDPSC eta
gBMSC-en gene bideen aberaspen analisi konparatiboa. Taulan agertzen ez diren konparaketak ez zuten
ezarritako n= 2 eta p< 0.05 eskakizuna bete. Laburdurak: DPSC: hortz mamiko zelula amak; BMSC: hezur
muineko zelula amak; C: kontrol medioa; T: desberdintzapen osteoblastiko medioa; Flask: plastiko azalera;
TI: Ti6AL4V titanioa eta BAS: “Biomimetic Advanced Surface” titanioa.
85
Igor Irastorza Epelde
86
Emaitzak
BMSC-C epithelial to
TI mesenchymal
transition
BMSC-C- GO:0035912 5 2 0,04224312 HEY2, DLL4 dorsal aorta
FLASK VS 7 morphgenesis
BMSC-T-
FLASK
DPSC-C- GO:0060218 13 2 0,02773949 BATF, SP7 hematopoietic
FLASK VS 1 stem cell
DPSC-T- differentiation
TI
5. taula. Gene bideen aberastasun analisia “Gene Ontology Biological Process”-ez (GOBP) egina. Plastiko
eta titanio azaleren gainean desberdintzapen osteoblastiko medioaren presentzian edo gabezia hazitako
gDPSC eta gBMSC-en gene bideen aberaspen analisi konparatiboa. Taulan agertzen ez diren konparaketak
ez zuten ezarritako n= 2 eta p< 0.05 eskakizuna bete. Laburdurak: DPSC: hortz mamiko zelula amak; BMSC:
hezur muineko zelula amak; C: kontrol medioa; T: desberdintzapen osteoblastiko medioa; Flask: plastiko
azalera; TI: Ti6AL4V titanioa eta BAS: “Biomimetic Advanced Surface” titanioa.
Dezelulaturiko txerri ehun adiposoa (pDAT) apar solido bezala prozesatuaren SEM
irudiak
A B
C D
87
Igor Irastorza Epelde
26. irudia. In vitro erabilerarako dezelularizaturiko txerri ehun adiposoaren egitura porotsua SEM
irudietan. In vitro zelulekin konbinatuta erabiltzeko dezelularizaturiko txerri ehun adiposoaren irudi
makroskopikoa (A, B). pDAT-ren SEM irudian 50-100 µm arteko poro tamainak erakutsiz (C, D).
pDAT-ren eragin zitotoxikoa baztertzeko, 15.000 gDPSC zelula erein ziren bai
hobitxo hutsetan (plastikoa) eta baita pDAT apar solidoetan 4 egunez. Zelulen
bideragarritasun entsegua gDPSC-ak, Kaltzeina-AM (fuoreszentzia berdea) zelula bizien
tindatzailearekin eta propidio ioduro (fluoreszentzi gorria) hildako zelulen
tindatzailearekin haziz burutu zen. Fluoreszentzi mikroskopioarekin ateratako irudiek,
gDPSC-ak pDAT-an haztean ia % 100-eko bideragarritasuna zutela frogatu zuten, zelula
guztiak fluoreszentzi berdez tindaturik aurkituz. Zelula isolatuak bakarrik aurkitu ziren
gorriz tindaturik, pDAT-ren zitotoxizitate eza erakutsiz (28. irudia). Bi azalera hauen
arteko alderik handiena, plastikoan aurkituriko zelula kopuru handia izan zen (27. irudia
A, B). Aurkitutako gDPSC zelula kopuru desberdintasun hau, plastikoan duten mono-
geruza hazkuntzarengatik izan daiteke, pDAT-an duten 3 dimentsiotako hazkuntzarekin
alderatuz.
Plastiko pDAT
A a D
B E
88
Emaitzak
27. irudia. Plastiko eta pDAT apar solidoetan hazitako gDPSC-en Kaltzeina_AM/ propidio ioduro irudiak.
gDPSC-ak 15.000 zelula/ hobitxo dentsitatean hazi ziren 4 egunez eta Kaltzeina-AM (berde) zelula bizi
markatzaile eta propidio ioduro (gorri) zelula hilen markatzaileekin tindatu ziren. Plastiko gainean hazitako
zelulek (A, B), pDAT-an hazitakoek (D, E) baina kopuru handiagoa erakutsi zuten, mono-geruza hazkundea
dela eta. Eskala barra (A, D): 100 µm; (B, E): 50 µm.
BGLAP 400
***
300
A Kontrola Desberdintzapena
200
Plastiko
100
0
a
DMEM DMEM
control pDAT
D
% normalized IF intensity
500
***
400
pDAT
300
200
100
0
Osteo Osteo
Control pDAT
89
Igor Irastorza Epelde
28. irudia. Plastiko eta pDAT-an desberdintzapen osteoblastikoaren presentzian edo gabezian hazitako
gDPSC-en Osteokaltzina (BGLAP) immunofluoreszentzia irudiak eta intentsitate kuantifikazioa. Plastiko
eta pDAT-a duten EZ-portetan 2 astez kontrol eta desberdintzapen medioekin hazitako gDPSC-en
immunofluoreszentzia irudiak (A). ImageJ-rekin eginiko BGLAP-aren IF markaketaren kuantifikazio
erlatiboa baldintza ezberdinetako zelula kopuruekiko (B, D). Eskala barra: 50 µm. Estatistikoki
esanguratsua (***p ≤ 0.001) denean.
% normalized IF intensity
300
SPARC 250
200
A Kontrola Derberdintzapena
150
Plastikoa
100
50
0
DMEM DMEM
D Control pDAT
% normalized IF intensity
350
***
300
pDAT
250
200
150
100
50
0
Osteo Osteo
Control pDAT
29. irudia. Plastiko eta pDAT-an desberdintzapen osteoblastikoaren presentzian edo gabezian hazitako
gDPSC-en Osteonektina (SPARC) immunofluoreszentzia irudiak eta intentsitate kuantifikazioa. Plastiko
eta pDAT-a duten EZ-portetan 2 astez kontrol eta desberdintzapen medioekin hazitako gDPSC-en
immunofluoreszentzia irudiak (A). ImageJ-rekin eginiko SPARC-en IF markaketaren kuantifikazio erlatiboa
baldintza ezberdinetako zelula kopuruekiko (B, D). Eskala barra: 50 µm. Estatistikoki esanguratsua (***p
≤ 0.001) denean.
90
Emaitzak
gDPSC-ak plastiko eta pDAT-an hazi ziren 14 egunez kontrol eta desberdintzapen
osteoblastiko medioekin. Ondoren, mineralizatzen duten hezur zelulen ezaugarri den
fosfatasa alkalino entzima tindatu zen. Irudiek erakutsi zutenez, gDPSC-ek ALP
aktibitatea zuten bi azaleretan (30. irudia A). ALP kuantifikazioak, bi azaleretan
desberdintzapen medioarekin hazitako zelulen markaketa handipen esanguratsua
frogatu zuten (30. irudia B, D).
Kontrola Desberdintzapena B **
A 300
100
50
0
Plastiko
D 1000 **
% relative ALP absorbance
800
600
pDAT
400
200
0
Osteo
DMEM
pDAT
30. irudia. Plastiko eta pDAT-an desberdintzapen osteoblastikoaren presentzian edo gabezian hazitako
gDPSC-en ALP aktibitate kuantifikazioa. gDPSC-ak 14 egunez hazi ziren plastiko eta pDAT-a zuten EZ-
portetan kontrol eta desberdintzapen osteoblastiko medioarekin. ALP aktibitatea prezipitatu
more/beltzen detekzioz gauzatu zen (A). ALP-ren kuantifikazioa, absorbantzia erlatiboaren ImageJ-rekin
neurtuz gauzatu zen. Estatistikoki esanguratsua (**p ≤ 0.01) demean.
91
Igor Irastorza Epelde
92
Emaitzak
Plastikoa
Desberdintzapena
Plastikoa
Desberdintzapena
% relative ARS absorbance
B 300 200 *
250
150
200
150 100
100
50
50
0 0
DMEM Osteo DMEM Osteo
Plastikoa pDAT
31. irudia. Plastiko eta pDAT-an desberdintzapen osteoblastikoaren presentzian edo gabezian hazitako
gDPSC-en Alizarin gorri-DAPI tindaketa bikoitza eta kuantifikazioa. gDPSC-ak hobitxo hutsetan
(plastikoa) eta pDAT apar solidoetan hazi ziren kontrol eta desberdintzapen osteoblastiko medioarekin 4
astez Alizarin gorri tindaketa egin baino lehen. Zelulen nukleoak DAPI-z tindatu ziren. Eremu argiko eta
immunofluoreszentzia irudiak batu ziren gDPSC-en lokalizazioa aztertzeko mineralizaturiko hezur matrize
noduluen inguruan (A). Alizarin gorriaren kuantifikazio erlatiboa neurtu zen (B). Eskala barra: 50 µm.
Estatistikoki esanguratsua (*p ≤ 0.05) denean.
93
Igor Irastorza Epelde
94
Emaitzak
D E
F G
32. irudia. pDAT- hazitako gDPSC-en ultra-egiturara ezaugarriak TEM irudien bidez. gDPSC-ak 4 astez
hazi ziren kontrol (ezker zutabea) eta desberdintzapen osteoblastiko medioekin (eskuin zutabea). gDPSC-
ek sorturiko kolageno fibra lodiago eta kaltzifikatuen hezur ECM-a eta pDAT apar solido matrizeko
kolageno fibra meheen arteko trantsizio area aztertu zen (marratxo horia). Kolageno fibra elektro-dentso
kantitate gehiago ikusi zen desberdintzapen osteoblastiko baldintzetan (beheko handipen altuko irudiak).
Eskala barra: 1 µm (A, D eta E); 500 nm (B); 200 nm (F eta G).
95
Igor Irastorza Epelde
B
* *
*
*
* *
33. irudia. pDAT apar solidoetan hazitako gDPSC-ek sortutako mintz-barneko osifikazio gune eta
Sharpey-gisako fibrak TEM irudien bidez. pDAT apar solidoetan hazitako gDPSC-en kaltzifikaturiko
kolageno sorta lodi arearen eta kolageno area ez kaltzifikatuaren arteko trantsizioa (marratxo horia).
Mintz-barneko osifikazio guneak aurkitu ziren kolageno lodia zuten egitura elektro-dentsoetan. Gezi
puntek seinalaturiko guneen handipenak eskuin panelean (A). Sharpey-moduko fibra zulatzaileen
presentzia mintz barneko osifikazio guneen ertzetan (eskuin paneleko izartxoak), kolageno matrizeari
ainguratzea erakutsiz. Gezi puntek seinalaturiko guneen handipenak eskuin panelean (B). Eskala barra: 2
µm (goiko ezkerreko irudia); 1 µm (beheko ezkerreko irudia); 500 nm (goiko eskuineko irudiak); 200 nm
(beheko eskuineko irudiak).
96
Eztabaida
Eztabaida
99
Igor Irastorza Epelde
100
Eztabaida
gDPSC-en in vitro hedapen eta mantentzea ere ikertua izan zen Ti6AL4V (lehun)
eta BAS (gogor) titanio azaleretan. Bi azalerek, zelulen bideragarritasuna mantendu
zuten, zelulen ugaltze ona baimenduz. Hau, Ki67-rako positibo ziren zelulak detektatuz
egiaztatu zen baldintza basaletan. Ti6AL4V titanio azaleran hazitako gDPSC-ek
mobilitate handiagoa erakutsi zuten BAS titanio azaleraren gainean hazitakoekin
alderatuta, azalera lauaren ondorioz. Nahiz eta plasmatik eratorritako bi produktuek
gDPSC-en ugalketa ona baimendu zuten, PRF-ak ez bezala, % 20-ko PRGF-z osaturiko
medioak esanguratsuki handitu zuen zelulen hazkuntza. Emaitzen ezberdintasun hau, bi
produktu hauen erabilera metodo ezberdinengatik eragina izan zitekeen. PRF mintzak
plaken hobitxoetan mantendu ziren esperimentuak iraun zuen denbora osoan zehar,
molekulak pixkanaka askatuz. Honen aurka, disolbagarria den PRGF-a, medio aldaketa
bakoitzarekin % 20-ko kontzentrazioan berritu zen 2-3 egunero. Posiblea da, hazkunde
medioari PRGF-a gehitzean eragindako hazkuntza faktoreen kontzentrazio handiak
gDPSC-en ugalketa areagotu zuela esatea, eta aldi berean honek, osteoblasto
helduetara desberdintzeko prozesuan eragin negatiboa zuela esatea. Bi prozesu hauek
(zelula ugalketa vs desberdintzapena) prozesu antagonikoak direlako. PRGF-arekin
hazitako gDPSC-ek, RUNX2 eta SPARC bezalako osteoblasto konpromezu markatzaileen
adierazpen mailak handituak zituzten. Baina nolabait, GDPSC-ek huts egin zuten
osteoblastu zelula helduetara desberdintzean, OSTERIX transkripzio faktorearen
adierazpen maila baxuak eta Alizarina gorriz tindatutako jariaturiko hezur matrize
mineralizatuaren detekzioak erakutsi zuen bezala. Bestalde, PRF-z osaturiko
medioarekin hazitako gDPSC-ak, fibrina mintz honek pixkanaka askaturiko hazkuntza
faktore hauei esker, osteoblasto helduen desberdintzapena areagotu zuten.
Garrantzitsua da aipatzea, gDPSC-al BAS titanio azaleraren gainean PRF-rekin haztean
izan zela in vitro desberdintzapen osteoblastikorako baldintzarik eraginkorrena. Hau
izan zen BAS titanio gogor azalerak Ti6AL4V titanioarekin alderatuta aukera hobea zela
erakutsi zuen unea.
101
Igor Irastorza Epelde
Metodologia hau hortz klinikara itzuli ahal izateko, in vivo ikerketa gehiago
beharrezkoak dira. Nahiz eta ez izan giza gorputz helduan zelula ama mesenkimal iturri
ugariena, inplantologia eta ebakuntza kraniomaxilofazial arloetarako, gDPSC-ak oso
zelula interesgarriak dira. Istripuren batean hortzak galdu dituzten pazienteen kasuan,
hau bereziki interesgarria da. Galdutako hortz piezatik lorturiko gDPSC autologoen
erabilerak onura handiak ekar ditzake kalteturiko hezur gunearen birsorpena
laguntzean. Kasua edozein izanik ere, BAS bezalako mikro-porodun titanio azalera
biomimetikoan haztean, gDPSC-ek eraginkortasunez erantzuten dute desberdintzapen
osteoblastiko protokoloei, bereziki plaketetan aberatsa den fibrina mintzarekin
konbinatzean.
102
Eztabaida
pausoan zelulek RUNX2 transkripzio faktorearen adierazpena areagotzen duten, eta poliki-poliki geroz eta
Kolageno I eta SPARC gehiago. Desberdintzapen osteoblastikoaren azken etapan, zelulek matrize extra-
zelularra mineralizatzeko gaitasuna lortzen dute, hezur ehun heldua sortuz. Zelula/molekula mailan,
OSTERIX transkripzio faktorearen adierazpena du ezaugarritzat, osteoblasto/osteozito helduen gene
markatzailea. Erreaktibo farmakologiko, plasmatik eratorritako PRGF eta PRF, eta titanio azalerek gDPSC-
en osteoblasto desberdintzapen konpromezuan eragina dutela ikusi da. Hau, RUNX2 eta SPARC eta
fosfatasa alkalinoaren adierazpenaren detekzioz egiaztatu da baldintza guzti hauetan. Hala ere, PRGF
disolbagarriarekin hazitako gDPSC-ekez zuten osteoblasto heldu etapara eltzea lortu, seguraski
ugaltze/desberdintze prozesu antagonikoen eraginez. Honen aurka, PRF mintzekin hazitako gDPSC-ek
desberdintzapen osteoblastiko markatzaileen adierazpen handienak izan zituzten, bereziki BAS titanio
azalera biomimetikoa eta desberdintzapen osteoblastiko medioarekin konbinatzean.
Ti6AL4V eta BAS titanio azaleren gainean hazitako gDPSC eta gBMSC-ek ez zuten
zito-toxizitaterik erakutsi. Bi zelula mota hauek, ia % 100-eko bideragarritasuna adierazi
zuten titanio azaleren gainean haztean, non isolaturiko zelularen bat bakarrik aurkitu
103
Igor Irastorza Epelde
zen ioduro propidioarekin tindatua. Hala eta guztiz ere, zelula ugalketa esperimentuak
gDPSC eta gBMSC-en arteko ezberdintasunak erakutsi zituen. 24 eta 48 orduz titanio
gainean hazi ostean gDPSC-ek, gBMC-ek baino ugalketa maila handiagoa erakutsi zuten.
Kristal gainean haztean, aldaketa hau 24 orduren ostean bakarrik ikusi zen, bi zelulen
artean 48 orduren ondoren ugalketa maila antzekoa ikusiz. Emaitza hauek lehenago
eginiko artikuluekin bat egiten dute, non emaitza berberak izan zituzten zelulak bio-
material ezberdinen gainean haztean (Amid et al., 2021; Ponnaiyan and Jegadeesan,
2014).
gDPSC eta gBMSC-ek jariaturiko mineral gordailuen sorrera ere ezberdina izan
zen kristal, Ti6AL4V eta BAS-ean. Alizarin gorri tindaketaren kuantifikazioak erakutsi
zuenez, gDPSC-ek hezur mineral produkzio handiagoa eduki zuten gBMSC-ekin
alderatuz- gainera, Ti6AL4V eta BAS titanio azaleren gainean desberdintzapen
osteoblastikoarekin hazitako gDPSC-ek erakutsi zuten baldintza guztien artean hezur
mineral sorrera handiena. Erlazionaturiko ikerketetan ikusi zuten bezala, gDPSC-ek
gBMSC-ek baino mineralizazio handiagoa eduki zuten bi titanio azaleretan haztean
(Davies et al., 2015; Mohanram et al., 2020).
104
Eztabaida
105
Igor Irastorza Epelde
al., 2014a; Felthaus et al., 2014b; Onizuka et al., 2016). Orokorrean, gDPSC eta gBMSC-
ak kultibo baldintza ezberdinetan haztean egin den RNA sekuentziazio honek ZBTB16-ek
osteogenesiaren erregulazioan rol garrantzitsua izan dezakeela iradoki dute.
Osx
Sp1
ZBTB16
Sp1
106
Eztabaida
Lehen aipatu bezala, hortz inplanteak asko hobetu dira azken urteetan
inplanteen biziraupen maila areagotuz. Biziraupen hau % 95 inguruan egongo litzateke
inplantea jarri eta 10 urte igaro ondoren (Moraschini et al., 2015) eta % 88-an 20 urte
107
Igor Irastorza Epelde
igaro ondoren (Chrcanovic et al., 2018). Hala ere, bizi-itxaropenaren handipenarekin eta
herrialde garatuen biztanleriaren zahartzearengatik, denbora luzeko iraupena duten
hortz inplanteen eskaria mantendu egingo dela ematen du etorkizunean.
108
Eztabaida
baskulogeniko eta osteogenikoak lortuz. Gure taldeak orain dela gutxi argitaratu eta
patentatu zuen zelula baskulogenikoak lortzeko (endotelia eta perizitoak) animali
serumik gabeko metodologia (Luzuriaga et al., 2020; Pineda Martí et al., 2020). Metodo
hau bio-materialetan ereindako gDPSC-ei aplika daiteke pDAT-a bezalako scaffold-ak
baskularizatzeko eta honela zelula terapietan erabili ahal izateko.
Ikerketa honetan lorturiko emaitzek, PDL eta hortz hezur birsorkuntzarako pDAT
apar solidoak bio-material garrantzitsua izan daitekeela frogatu du. pDAT-aren
formulazio honek, gDPSC-ek jariaturiko hezur matrizea esanguratsuki handitu zuen,
fosfatasa alkalino eta Alizarin gorriaren ebaluaketak erakusti zuen bezala. Material
honetan ereindako gDPSC-ek ez zuten bideragarritasun galerarik erakutsi baldintza
esperimentaletan. gDPSC-ak dentsitate baxuetan erein ziren (15.000 zelula/hobitxo;
21.428 zelula/ zm2) eta baita pDAT scaffoldaren bolumen baxuak (120 µl/ hobitxo)
denbora luzeko ikerketetarako. Zelula dentsitate handiagoen erabilerak seguraski,
baskulatura bidezko mantenugai eta oxigeno horniketa beharko luke (Nakamura et al.,
2019). In vivo esperimentu gehiago beharrezkoak izango dira material honen potentziala
ikertzeko hezur ehun eta PDL lesioen sendakuntzan.
Azkenik, pDAT-an ereindako gDPSC-ek kaltzifikaturiko kolageno fibra sorta lodiak eta
mintz barneko osifikazio lekuak sortu zituzten. Kaltzifikaturiko gune hauek pDAT-ri
109
Igor Irastorza Epelde
itsatsirik daude Sharpey fibren bidez. Emaitza guzti hauek erakutsi dutenez gDPSC-ak,
edo beste ama zelula mesenkimalen bat, pDAT-rekin konbinatzean desberdintzapen
osteoblastikoa induzitzeaz gain, injertoa inguruko ehun gogorrari itsasten laguntzen du.
Hezur sendaketaren kasuan itsaspen honek interes berezia du, non scaffold-a eta hezur
ehunaren arteko itsaspen fisikoa beharrezkoa den. In vivo transplantazio esperimentuak
beharko diraaurkiketa hauek baieztatzeko, non zelula amen garraiobide izateaz aparte,
scaffold-ak ECM bio-induktore gisa jokatuko luke, hezur ehun gogorrarekin itsaspen
kaltzifikatua areagotuz eta baita hezur ehun ostalariko ama zelula mesenkimalen
errekrutatze eta aktibazioa eraginez.
110
Ondorioak
Ondorioak
1. gDPSC-en bideragarritasuna eta ugalketa ez dira kaltetu Ti6AL4V eta BAS titanio
azaleretan haztean.
2. Bi titanio azalerek eragin osteo-induktiboa erakutsi dute gDPSC-engan
desberdintzapen osteoblastiko medioaren beharrik gabe.
3. Plasmatik eratorritako PRGF-ak gDPSC-en in vitro ugalketa areagotu du, aldi
berean PRF-ak desberdintzapen osteoblastikoa eta kaltzifikaturiko hezur matrize
produkzioa maximizatu du.
4. BAS titanio azalera plasmatik eratorritako PRF-arekin konbinaturik, gDPSC-en
zelula hezur-sortzaileetarako desberdintzapen osteoblastikoa bultzatu du.
Emaitza hauek, klinika praktika ohikoetan zabalduriko plasmatik eratorritako
fibrina koaguluaren erabilerari laguntza esperimentala eman diote, mikro-
porodun titanio inplante azaleren aldameneko hezur produkzioa bultzatzeko.
5. Ti6AL4V eta BAS titanio azaleretan hazitako gDPSC eta gBMSC-en ikerketa
konparatiboak, gDPSC-ek ugalketa eta mineralizazio handiagoa dutela erakutsi
dute gBMSC-ekin alderatuta, baina esanahi estatistikoaren faltaren ondorioz,
datu esperimental gehiagoren beharra erakutsi dute ondorio sendoak
ateratzeko. Isolamendu errazago eta inbasibotasun txikiagoa, ugaltze ratio altu
113
Igor Irastorza Epelde
114
I Eranskina
patenteak
I Eranskina (patenteak)
(12) INTERNATIONAL APPLICATION PUBLISHED UNDER TUE PATENT COOPERATION TREATY (PCT)
UPV/EHU, Sede Building, 3rd floor, Barrio Sarriena, s/n, 48940 LEIOA (ES).
(72) Inventors: PINEDA MARTÍ, José Ramón; ACHUCAR-
RO BASQUE CENTER FOR NEUROSCIENCE FUN-
DAZIOA, Science Park of the UPV/EHU, Sede Building, 3rd floor, Barrio Sarriena, sin, 48940 LEIOA (ES).
LUZURIAGA GONZÁLEZ, Jon; Dep. Bio. cel. Facultad de Medicina y Enfermería, UPV/EHU, 48940 LEIOA
(ES). UNDA RODRÍGUEZ, Fernando; Dep. Bio. cel. Facultad de Medicina y Enfermeffa, UPV/EHU, 48940
LEIOA (ES). PASTOR ALONSO, Oier•, ACHUCARRO BASQUE CENTER FOR NEUROSCIENCE FUN-
DAZIOA, Science Park of the UPV/EHU, Sede Building, 3rd floor, San-iena, sin, 48940 LEIOA (ES).
ENCINAS PÉREZ, Juan Manuel; ACHUCARRO BASQUE
CENTER FOR NEUROSCIENCE FUNDAZIOA, Science Park ofthe UPV/EHU, Scdc Building, 3rd floor, Bam
Sarfiena, 48940 LEIOA (ES). 'BARRETXE BILBAO, Gaskon; Dcp. Bio. Ccl. Facultad dc Medicina y
Ellfcrmcría,
UPV/EHU, 48940 LEIOA (ES). IRASTORZA EPELDE, Igor; Dcp. Bio. Ccl. Facultad dc Medicina y
Enfcrmcría, UPV/EHU, 48940 LEIOA CES).
(74) Agent: ZBM PATENTS - ZEA, BARLOCCI & MARKVARDSEN; Rambla Catalunya 123, 08008 Barcelona
CES).
(81) Designated States (unless otherwise indica/ed, ./ôr evey:v kind ofna/Ìona/ pro/ec/Ìon available): AE,
AG, AL, AM AO, AT, AU, AZ, BA, BB, BG, Bil, BN, BR, BW, BY, BZ,
CA, Cll, CL, CN, CO, CR, CU, CZ, DE, D], DR, DM, DO,
117
Igor Irastorza Epelde
HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KM, RN, KP, KR, KW, KZ, LA, LC, LK, LR, LS, W, LY, MA, MD, ME,
MG, MK, MN, MW, NIX, MY, MZ, NA, NG, M, NO, NZ, 0M, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW,
SA, SC, SD, SE, SG, SK, SL, SM, ST, SV, sy, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM,
ZW.
(84) Designated States (unless otherwise indica/ed, for evuy kind of regional pro/ec/Žon available): ARIPO
(B W, GM,
UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, TM), European (AL, AT, BE, BG, CM, cy, CZ, DE, DR,
EE, ES, Fl, FR, GB, GR, HR, HU, IE, IS, IT, LT, W, LV, MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, Sl, SK, SM,
TR), OAPI (BF, BJ, CF, CG, Cl, CM, GA, GN, GQ, GW, KM, ML, MR, NE, SN, TD, TG).
REVIEW
published:16 October 2015
doi:10.3389/fphys.2015.0028 9
121
II Ernaskina (artikuluak)
ORIGINALRESEARC H
published:30 March 2016
doi:10.3389/fcell.2016.0002 5
123
II Ernaskina (artikuluak)
Key Words: Serum-free culture media • Calcium imaging • Cell differentiation • Dental Pulp Stem
Cells • Brain Derived Neurotrophic Factor
Abstract
Background/Aims: Human Dental Pulp Stem Cells (hDPSCs) are one of the most promising
types of cells to regenerate nerve tissues. Standard DMEM+10% fetal bovine serum (FBS) culture
medium allows a fast expansion of hDPSC as a surface-adherent cell monolayer. However, the
use of FBS also compromises the clinical use of these protocols, and its longterm presence favors
hDPSCs differentiation toward mesenchymal cell-derived lineages, at the expense of a reduced
capability to generate neural cells. The objective of this work was to characterize the role of
neurotrophin signaling on hDPSCs using a serum-free culture protocol, and to assess the
neurogenic and gliogenic capacity of hDPSCs for future nerve tissue bioengineering and
regeneration. Methods: We compared the different expression of neurotrophin receptors by RT-
PCR, Q-PCR, and IF of hDPSCs cultured with different growth media in the presence or absence
of serum. Moreover, we assessed the response of hDPSCs to stimulation of neurotransmitter
receptors by live cell calcium imaging under these different media. Finally, we compared the
osteogenic potential of hDPSCs by Alizarin red staining, and the differentiation to
gliogenic/neurogenic fates by immunostaining for Schwann lineage
Gaskon Ibarretxe Cell Biology & Histology Department, Faculty of Medicine and Nursing, University of the Basque Country, UPV/ and
Jose R. Pineda EHU; Achucarro Basque Center for Neuroscience Fundazioa, Barrio Sarriena s/n; Sede Building 3rd floor, Leioa,
Bizkaia, 48940 (Spain) Tel. (+34) 946013218, E-Mail gaskon.ibarretxe@ehu.eus; jr.pineda@achucarro.org
125
II Ernaskina (artikuluak)
European Cells and Materials Vol. 38 2019 (pages 201-214) I Irastorza et al.
hDPSCs with PRF and PRGF on biomimetic titanium DOI: 10.22203/eCM.v038a14 ISSN 1473-2262
Department of Cell Biology and Histology. Faculty of Medicine and Nursing, University
1
Dental implants are the usual therapy of choice in the dental clinic to replace a loss of natural teeth. Over
recent decades there has been an important progress in the design and manufacturing of titanium implant
surfaces with the goal of improving their osteointegration. In the present work, the aim was to evaluate the
usefulness of hDPSCs (human dental pulp stem cells), in combination with autologous plasma components,
for in vitro bone generation on biomimetic titanium dental implant materials. In this context, the combination
of hDPSCs stimulated by PRGF or PRF and cultured on standard Ti6A14V and biomimetic BAS™ (Avinent
Implant System) titanium surfaces were studied in order to evaluate possible enhancements in the
osteoblastic differentiation process out of human mesenchymal cells, as well as bone matrix secretion on the
implant surface. The results obtained in this in vitro model of osteogenesis suggested a combination of
biomimetic rough titanium surfaces, such as BAS™, with autologous plasma-derived fibrin-clot membranes
such as PRF and/or insoluble PRGF formulations, but not with an addition of water-soluble supplements of
plasma-derived growth factors, to maximise osteoblastic cell differentiation, bone generation, anchorage and
osteointegration of titanium-made dental implants.
Keywords: Dental pulp stem cells, titanium implants, osteoblast differentiation, platelet rich in growth
factors, platelet rich fibrin, biomimetic advanced surface.
*Address for correspondence: Fernando Unda, Cell Biology and Histology Department. Faculty of Medicine
and Nursing, University of the Basque Country, UPV/EHU, Leioa, 48940, Bizkaia, Spain. Telephone number:
+34 946012857 Email: fernandoundarodriguez@gmail.com
Copyright policy: This article is distributed in accordance with Creative Commons Attribution Licence
(http://creativecommons.org/licenses/by-sa/4.0/).
127
II Ernaskina (artikuluak)
biomedicines
Article
Abstract: The generation of vasculature is one of the most important challenges in tissue
engineering and regeneration. Human dental pulp stem cells (hDPSCs) are some of the most
promising stem cell types to induce vasculogenesis and angiogenesis as they not only secrete
vascular endothelial growth factor (VEGF) but can also differentiate in vitro into both
endotheliocytes and pericytes in serum-free culture media. Moreover, hDPSCs can generate
complete blood vessels containing both endothelial and mural layers in vivo, upon transplantation
into the adult brain. However, many of the serum free media employed for the growth of hDPSCs
contain supplements of an undisclosed composition. This generates uncertainty as to which of its
precise components are necessary and which are dispensable for the vascular differentiation of
hDPSCs, and also hinders the transfer of basic research findings to clinical cell therapy. In this work,
we designed and tested new endothelial differentiation media with a fully defined composition
using standard basal culture media supplemented with a mixture of B27, heparin and growth
factors, including VEGF-A165 at different concentrations. We also optimized an in vitro Matrigel
assay to characterize both the ability of hDPSCs to differentiate to vascular cells and their capacity
to generate vascular tubules in 3D cultures. The description of a fully defined serum-free culture
medium for the induction of vasculogenesis using human adult stem cells highlights its potential as
a relevant innovation for tissue engineering applications. In conclusion, we achieved efficient
vasculogenesis starting from hDPSCs using serum-free culture media with a fully defined
composition, which is applicable for human cell therapy purposes.
129
II Ernaskina (artikuluak)
cells
Article
Abstract: Dental pulp stem cells (DPSCs) from adult teeth show the expression of a very complete
repertoire of stem pluripotency core factors and a high plasticity for cell reprogramming. Canonical
WntandNotchsignalingpathwaysregulatestemnessandtheexpressionofpluripotencycorefactorsin
DPSCs, and even very short-term (48 h) activations of the Wnt pathway induce a profound
remodeling of DPSCs at the physiologic and metabolic levels. In this work, DPSC cultures were
exposed to treatments modulating Notch and Wnt signaling, and also induced to differentiate to
osteo/adipocytes. DNA methylation, histone acetylation, histone methylation, and core factor
expression levels where assessed by mass spectroscopy, Western blot, and qPCR. A short-term
activation of Wnt signaling by WNT-3A induced a genomic DNA demethylation, and increased
histone acetylation and histone methylation in DPSCs. The efficiency of cell reprogramming
methods relies on the ability to surpass the epigenetic barrier, which determines cell lineage
specificity. This study brings important information about the regulation of the epigenetic barrier
by Wnt signaling in DPSCs, which could contribute to the development of safer and less
aggressive reprogramming methodologies with a view to cell therapy.
Keywords: dental pulp stem cells; chromatin remodeling; cell cycle; pluripotency; DNA methylation;
histone acetylation; histone methylation; Notch pathway; Wnt pathway
131
II Ernaskina (artikuluak)
BASIC SCIENCE
Nanomedicine: Nanotechnology, Biology, and Medicine
31 (2021) 102314
Abstract
Withinthe field of neural tissue engineering,there is a huge need for the development of materials that promote the
adhesion,aligned migration and differentiation of stem cells into neuronal and supportive glial cells. In this study, we have
fabricated bioresorbable elastomeric scaffolds combining an ordered nanopatterned topography together with a surface
functionalization with graphene oxide (GO) in mild conditions. These scaffolds allowed the attachment of murine neural
stem cells (NSCs) without the need of any further coating of its surface with extracellular matrix adhesion proteins. The
NSCs were able to give rise to both immature neurons and supporting glial cells over the nanostructured scaffolds in vitro,
promoting their aligned migration in cell clusters following the nanostructured grooves. This system has the potential to
reestablish spatially oriented neural precursor cell connectivity, constituting a promising tool for future cellular therapy
including nerve tissue regeneration. © 2020 Elsevier Inc. All rights reserved.
Key words: Micro- and nanopatterning; Neural stem cells; Migration; Cell differentiation; Graphene oxide; Biodegradable polymer
Funding sources: Basque Government (GV/EJ) Regeneration of the nervous system still remains very challenging
Department of Education, Linguistic Politics and due to its limited plasticity and poor ability to heal ments for this
Culture (GIC 15/52, IT-927-16), MINECO «Ramón y specific biomedical application play a pivotal role. Much progress
Cajal» program RYC-2013-13450 (JRP), MINECO
has been made in determining the ideal features a biomaterial
PID2019104766RB-C21, The University of The Basque
Country (UPV/EHU) by GIU16/66, UFI 11/44, should have for its use as a neural replacement graft, and in
COLAB19/03 and IKERTU-2020.0155. GV/EJ IT831- understanding the interactions of growing axons within
13, Hazitek ZE-2019/00012-IMABI and ELKARTEK thesebiomaterials;however,theregenerationlevelsinducedbythe
KK-2019/ 00093. Polimerbio and Y. P. have a Bikaintek biomaterial usually do not match those obtained by nerve tissue
PhD grant (20-AF-W2-201800001) and J.L. has a autografts and the development of new and effective nerve
UPV/EHU grant DOKBERRI 2019 (DOCREC19/49). regeneration therapies is still an urgent clinical need.2,3
Conflict of interest: The authors declare that there is no
conflict of interest. The biomaterials for nerve tissue regeneration should be
Correspondence to: J.R. Pineda, Cell Signaling lab,biocompatible and biodegradable, while providing structural cues
University of the Basque Country (UPV/EHU), Leioa, Spain. that promote oriented axon regeneration and guidance signals
Correspondence to: A. Larrañaga, Group of Science from extracellular matrix (ECM)-like components. Additionally,
and Engineering of Polymeric Biomaterials (ZIBIO they shouldalsopresentlong-
Group), University of the Basque Country (UPV/EHU). termstoragecapabilityandeaseofhandling/ suturing.4–6 One
E-mail addresses: joseramon.pinedam@ehu.eus, (J.R. important aspect to take in consideration is that the
Pineda), aitor.larranagae@ehu.eus. (A. Larrañaga).
1
These authors contributed equally to this work.
https://doi.org/10.1016/j.nano.2020.102314
1549-9634/© 2020 Elsevier Inc. All rights reserved.
133
Bibliografia
Bibliografia
Ackema KB, Charité J (2008) Mesenchymal stem cells from different organs are
characterized by distinct topographic Hox codes. Stem Cells Dev 17: 979–991.
doi:10.1089/scd.2007.0220.
137
Igor Irastorza Epelde
Alraies A, Waddington RJ, Sloan AJ, Moseley R (2020) Evaluation of Dental Pulp
Stem Cell Heterogeneity and Behaviour in 3D Type I Collagen Gels. Biomed Res Int 2020:
3034727. doi:10.1155/2020/3034727.
Amini AR, Laurencin CT, Nukavarapu SP (2012) Bone tissue engineering: recent
advances and challenges. Crit Rev Biomed Eng 40: 363–408.
doi:10.1615/critrevbiomedeng.v40.i5.10.
Anitua E (1999) Plasma rich in growth factors: preliminary results of use in the
preparation of future sites for implants. Int J Oral Maxillofac Implants 14: 529–535.
138
Bibliografia
Anitua E, Orive G, Pla R, Roman P, Serrano V, Andía I (2009) The effects of PRGF
on bone regeneration and on titanium implant osseointegration in goats: a histologic
and histomorphometric study. J Biomed Mater Res A 91: 158–165.
doi:10.1002/jbm.a.32217.
Anitua E, Tejero R, Zalduendo MM, Orive G (2013) Plasma rich in growth factors
promotes bone tissue regeneration by stimulating proliferation, migration, and
autocrine secretion in primary human osteoblasts. J. Periodontol. 84: 1180–1190.
doi:10.1902/jop.2012.120292.
Arthur A, Rychkov G, Shi S, Koblar SA, Gronthos S (2008) Adult human dental pulp
stem cells differentiate toward functionally active neurons under appropriate
environmental cues. Stem Cells 26: 1787–1795. doi:10.1634/stemcells.2007-0979.
139
Igor Irastorza Epelde
140
Bibliografia
Bertolini MM, Del Bel Cury AA, Pizzoloto L, Acapa IRH, Shibli JA, Bordin D (2019)
Does traumatic occlusal forces lead to peri-implant bone loss? A systematic review. Braz
Oral Res 33: e069. doi:10.1590/1807-3107bor-2019.vol33.0069.
Bianchi M, Urquia Edreira ER, Wolke JGC, Birgani ZT, Habibovic P, Jansen JA,
Tampieri A, Marcacci M, Leeuwenburgh SCG, van den Beucken JJJP (2014) Substrate
geometry directs the in vitro mineralization of calcium phosphate ceramics. Acta
Biomater 10: 661–669. doi:10.1016/j.actbio.2013.10.026.
141
Igor Irastorza Epelde
Bühring H-J, Battula VL, Treml S, Schewe B, Kanz L, Vogel W (2007) Novel markers
for the prospective isolation of human MSC. Ann N Y Acad Sci 1106: 262–271.
doi:10.1196/annals.1392.000.
142
Bibliografia
Cao C, Dong Y, Dong Y (2005) [Study on culture and in vitro osteogenesis of blood-
derived human mesenchymal stem cells]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 19:
642–647.
Chang C-C, Chang K-C, Tsai S-J, Chang H-H, Lin C-P (2014) Neurogenic
differentiation of dental pulp stem cells to neuron-like cells in dopaminergic and motor
neuronal inductive media. J Formos Med Assoc 113: 956–965.
doi:10.1016/j.jfma.2014.09.003.
Chaudhari AA, Vig K, Baganizi DR, Sahu R, Dixit S, Dennis V, Singh SR, Pillai SR
(2016) Future Prospects for Scaffolding Methods and Biomaterials in Skin Tissue
Engineering: A Review. Int J Mol Sci 17. doi:10.3390/ijms17121974.
143
Igor Irastorza Epelde
Chen YK, Huang AHC, Chan AWS, Lin LM (2016) Human dental pulp stem cells
derived from cryopreserved dental pulp tissues of vital extracted teeth with disease
demonstrate hepatic-like differentiation. J Tissue Eng Regen Med 10: 475–485.
doi:10.1002/term.1763.
Choi YC, Choi JS, Kim BS, Kim JD, Yoon HI, Cho YW (2012) Decellularized
extracellular matrix derived from porcine adipose tissue as a xenogeneic biomaterial for
tissue engineering. Tissue Eng Part C Methods 18: 866–876.
doi:10.1089/ten.TEC.2012.0009.
Civin CI, Trischmann T, Kadan NS, Davis J, Noga S, Cohen K, Duffy B, Groenewegen
I, Wiley J, Law P, Hardwick A, Oldham F, Gee A (1996) Highly purified CD34-positive cells
reconstitute hematopoiesis. J Clin Oncol 14: 2224–2233.
doi:10.1200/JCO.1996.14.8.2224.
144
Bibliografia
Coelho PG, Granjeiro JM, Romanos GE, Suzuki M, Silva NRF, Cardaropoli G,
Thompson VP, Lemons JE (2009) Basic research methods and current trends of dental
implant surfaces. J. Biomed. Mater. Res. Part B Appl. Biomater. 88: 579–596.
doi:10.1002/jbm.b.31264.
Coli P, Jemt T (2021) Are marginal bone level changes around dental implants
due to infection? Clin Implant Dent Relat Res. doi:10.1111/cid.12971.
Crisan M, Yap S, Casteilla L, Chen C-W, Corselli M, Park TS, Andriolo G, Sun B,
Zheng B, Zhang L, Norotte C, Teng P-N, Traas J, Schugar R, Deasy BM, Badylak S, Buhring
H-J, Giacobino J-P, Lazzari L, Huard J, Péault B (2008) A perivascular origin for
mesenchymal stem cells in multiple human organs. Cell Stem Cell 3: 301–313.
doi:10.1016/j.stem.2008.07.003.
Cuthbert RJ, Giannoudis PV, Wang XN, Nicholson L, Pawson D, Lubenko A, Tan
HB, Dickinson A, McGonagle D, Jones E (2015) Examining the Feasibility of Clinical Grade
CD271+ Enrichment of Mesenchymal Stromal Cells for Bone Regeneration. PLoS One 10.
doi:10.1371/journal.pone.0117855.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4356586/.
145
Igor Irastorza Epelde
Dagnino APA, Chagastelles PC, Medeiros RP, Estrázulas M, Kist LW, Bogo MR,
Weber JBB, Campos MM, Silva JB (2020) Neural Regenerative Potential of Stem Cells
Derived from the Tooth Apical Papilla. Stem Cells Dev 29: 1479–1496.
doi:10.1089/scd.2020.0121.
Davies OG, Cooper PR, Shelton RM, Smith AJ, Scheven BA (2015) A comparison
of the in vitro mineralisation and dentinogenic potential of mesenchymal stem cells
derived from adipose tissue, bone marrow and dental pulp. J Bone Miner Metab 33:
371–382. doi:10.1007/s00774-014-0601-y.
Denham M, Conley B, Olsson F, Cole TJ, Mollard R (2005) Stem cells: an overview.
Curr Protoc Cell Biol Chapter 23: Unit 23.1. doi:10.1002/0471143030.cb2301s28.
146
Bibliografia
Dohan DM, Choukroun J, Diss A, Dohan SL, Dohan AJJ, Mouhyi J, Gogly B (2006)
Platelet-rich fibrin (PRF): a second-generation platelet concentrate. Part II: platelet-
related biologic features. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 101: e45-50.
doi:10.1016/j.tripleo.2005.07.009.
147
Igor Irastorza Epelde
Duncan AW, Dorrell C, Grompe M (2009) Stem Cells and Liver Regeneration.
Gastroenterology 137: 466–481. doi:10.1053/j.gastro.2009.05.044.
148
Bibliografia
Fijnheer R, Pietersz RN, de Korte D, Gouwerok CW, Dekker WJ, Reesink HW, Roos
D (1990) Platelet activation during preparation of platelet concentrates: a comparison
of the platelet-rich plasma and the buffy coat methods. Transfusion 30: 634–638.
doi:10.1046/j.1537-2995.1990.30790385523.x.
Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RCR (2007) SSEA-4
identifies mesenchymal stem cells from bone marrow. Blood 109: 1743–1751.
doi:10.1182/blood-2005-11-010504.
149
Igor Irastorza Epelde
Gothard D, Dawson JI, Oreffo ROC (2013) Assessing the potential of colony
morphology for dissecting the CFU-F population from human bone marrow stromal
cells. Cell Tissue Res 352: 237–247. doi:10.1007/s00441-013-1564-3.
150
Bibliografia
significantly affects human stem cell bone tissue engineering. J. Cell. Physiol. 214: 166–
172. doi:10.1002/jcp.21175.
Griffiths MJD, Bonnet D, Janes SM (2005) Stem cells of the alveolar epithelium.
Lancet 366: 249–260. doi:10.1016/S0140-6736(05)66916-4.
Gronthos S, Mankani M, Brahim J, Robey PG, Shi S (2000) Postnatal human dental
pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. U.S.A. 97: 13625–13630.
doi:10.1073/pnas.240309797.
Gronthos S, Zannettino ACW, Hay SJ, Shi S, Graves SE, Kortesidis A, Simmons PJ
(2003) Molecular and cellular characterisation of highly purified stromal stem cells
derived from human bone marrow. J Cell Sci 116: 1827–1835. doi:10.1242/jcs.00369.
151
Igor Irastorza Epelde
Han Y-J, Kang Y-H, Shivakumar SB, Bharti D, Son Y-B, Choi Y-H, Park W-U, Byun J-
H, Rho G-J, Park B-W (2017) Stem Cells from Cryopreserved Human Dental Pulp Tissues
Sequentially Differentiate into Definitive Endoderm and Hepatocyte-Like Cells in vitro.
Int J Med Sci 14: 1418–1429. doi:10.7150/ijms.22152.
Haynesworth SE, Baber MA, Caplan AI (1992) Cell surface antigens on human
marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone 13:
69–80. doi:10.1016/8756-3282(92)90363-2.
Hodgkinson T, Yuan X-F, Bayat A (2009) Adult stem cells in tissue engineering.
Expert Rev Med Devices 6: 621–640. doi:10.1586/erd.09.48.
Honda MJ, Imaizumi M, Tsuchiya S, Morsczeck C (2010) Dental follicle stem cells
and tissue engineering. J Oral Sci 52: 541–552. doi:10.2334/josnusd.52.541.
152
Bibliografia
153
Igor Irastorza Epelde
Jang J-H, Lee H-W, Cho KM, Shin H-W, Kang MK, Park SH, Kim E (2016) In vitro
characterization of human dental pulp stem cells isolated by three different methods.
Restor Dent Endod 41: 283–295. doi:10.5395/rde.2016.41.4.283.
Jemat A, Ghazali MJ, Razali M, Otsuka Y (2015) Surface Modifications and Their
Effects on Titanium Dental Implants. Biomed Res Int 2015: 791725.
doi:10.1155/2015/791725.
Jones EA, Kinsey SE, English A, Jones RA, Straszynski L, Meredith DM, Markham
AF, Jack A, Emery P, McGonagle D (2002) Isolation and characterization of bone marrow
multipotential mesenchymal progenitor cells. Arthritis Rheum 46: 3349–3360.
doi:10.1002/art.10696.
154
Bibliografia
Khan SN, Cammisa FP, Sandhu HS, Diwan AD, Girardi FP, Lane JM (2005) The
biology of bone grafting. J Am Acad Orthop Surg 13: 77–86.
Khanabdali R, Saadat A, Fazilah M, Bazli KFK, Qazi R-M, Khalid RS, Hasan Adli DS,
Moghadamtousi SZ, Naeem N, Khan I, Salim A, Shamsuddin SA, Mohan G (2016)
Promoting effect of small molecules in cardiomyogenic and neurogenic differentiation
of rat bone marrow-derived mesenchymal stem cells. Drug Des Devel Ther 10: 81–91.
doi:10.2147/DDDT.S89658.
155
Igor Irastorza Epelde
Király M, Kádár K, Horváthy DB, Nardai P, Rácz GZ, Lacza Z, Varga G, Gerber G
(2011) Integration of neuronally predifferentiated human dental pulp stem cells into rat
brain in vivo. Neurochem Int 59: 371–381. doi:10.1016/j.neuint.2011.01.006.
Knychala J, Bouropoulos N, Catt CJ, Katsamenis OL, Please CP, Sengers BG (2013)
Pore geometry regulates early stage human bone marrow cell tissue formation and
organisation. Ann Biomed Eng 41: 917–930. doi:10.1007/s10439-013-0748-z.
156
Bibliografia
Kumar KR, Genmorgan K, Abdul Rahman SM, Rajan MA, Kumar TA, Prasad VS
(2016) Role of plasma-rich fibrin in oral surgery. J Pharm Bioallied Sci 8: S36–S38.
doi:10.4103/0975-7406.191963.
Lee CP, Colombo JS, Ayre WN, Sloan AJ, Waddington RJ (2015a) Elucidating the
cellular actions of demineralised dentine matrix extract on a clonal dental pulp stem cell
population in orchestrating dental tissue repair. J Tissue Eng 6: 2041731415586318.
doi:10.1177/2041731415586318.
Lee J-S, Kim S-K, Gruber R, Kim C-S (2020) Periodontal healing by periodontal
ligament fiber with or without cells: A preclinical study of the decellularized periodontal
ligament in a tooth replantation model. J Periodontol 91: 110–119.
doi:10.1002/JPER.19-0126.
157
Igor Irastorza Epelde
Lee J-T, Choi S-Y, Kim H-L, Kim J-Y, Lee H-J, Kwon T-G (2015b) Comparison of gene
expression between mandibular and iliac bone-derived cells. Clin Oral Invest 19: 1223–
1233. doi:10.1007/s00784-014-1353-8.
Lenkiewicz AM (2019) Epidermal Stem Cells. Adv Exp Med Biol 1201: 239–259.
doi:10.1007/978-3-030-31206-0_12.
Leucht P, Kim J-B, Amasha R, James AW, Girod S, Helms JA (2008) Embryonic
origin and Hox status determine progenitor cell fate during adult bone regeneration.
Development 135: 2845–2854. doi:10.1242/dev.023788.
Liu N, Zhou M, Zhang Q, Yong L, Zhang T, Tian T, Ma Q, Lin S, Zhu B, Cai X (2018)
Effect of substrate stiffness on proliferation and differentiation of periodontal ligament
stem cells. Cell Prolif 51: e12478. doi:10.1111/cpr.12478.
158
Bibliografia
Mansergh FC, Wride MA, Rancourt DE (2000) Neurons from stem cells:
implications for understanding nervous system development and repair. Biochem Cell
Biol 78: 613–628.
159
Igor Irastorza Epelde
Marx RE, Carlson ER, Eichstaedt RM, Schimmele SR, Strauss JE, Georgeff KR
(1998) Platelet-rich plasma: Growth factor enhancement for bone grafts. Oral Surg Oral
Med Oral Pathol Oral Radiol Endod 85: 638–646. doi:10.1016/s1079-2104(98)90029-4.
Marx RE (2004) Platelet-rich plasma: evidence to support its use. J Oral Maxillofac
Surg 62: 489–496. doi:10.1016/j.joms.2003.12.003.
McElreavey KD, Irvine AI, Ennis KT, McLean WH (1991) Isolation, culture and
characterisation of fibroblast-like cells derived from the Wharton’s jelly portion of
human umbilical cord. Biochem Soc Trans 19: 29S. doi:10.1042/bst019029s.
160
Bibliografia
Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S (2003) SHED: stem
cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A 100: 5807–5812.
doi:10.1073/pnas.0937635100.
Murphy CM, Haugh MG, O’Brien FJ (2010) The effect of mean pore size on cell
attachment, proliferation and migration in collagen-glycosaminoglycan scaffolds for
bone tissue engineering. Biomaterials 31: 461–466.
doi:10.1016/j.biomaterials.2009.09.063.
Nada OA, El Backly RM (2018) Stem Cells From the Apical Papilla (SCAP) as a Tool
for Endogenous Tissue Regeneration. Front Bioeng Biotechnol 6: 103.
doi:10.3389/fbioe.2018.00103.
161
Igor Irastorza Epelde
Naves MM, Menezes HHM, Magalhães D, Ferreira JA, Ribeiro SF, de Mello JDB,
Costa HL (2015) Effect of Macrogeometry on the Surface Topography of Dental Implants.
Int J Oral Maxillofac Implants 30: 789–799.
Nemeth CL, Janebodin K, Yuan AE, Dennis JE, Reyes M, Kim D-H (2014) Enhanced
chondrogenic differentiation of dental pulp stem cells using nanopatterned PEG-GelMA-
HA hydrogels. Tissue Eng Part A 20: 2817–2829. doi:10.1089/ten.TEA.2013.0614.
O’Brien FJ (2011) Biomaterials & scaffolds for tissue engineering. Materials Today
14: 88–95. doi:10.1016/S1369-7021(11)70058-X.
Olivares-Navarrete R, Hyzy SL, Hutton DL, Erdman CP, Wieland M, Boyan BD,
Schwartz Z (2010) Direct and indirect effects of microstructured titanium substrates on
the induction of mesenchymal stem cell differentiation towards the osteoblast lineage.
Biomaterials 31: 2728–2735. doi:10.1016/j.biomaterials.2009.12.029.
162
Bibliografia
Onizuka S, Iwata T, Park S-J, Nakai K, Yamato M, Okano T, Izumi Y (2016) ZBTB16
as a Downstream Target Gene of Osterix Regulates Osteoblastogenesis of Human
Multipotent Mesenchymal Stromal Cells. J Cell Biochem 117: 2423–2434.
doi:10.1002/jcb.25634.
163
Igor Irastorza Epelde
Pilipchuk SP, Plonka AB, Monje A, Taut AD, Lanis A, Kang B, Giannobile WV (2015)
Tissue engineering for bone regeneration and osseointegration in the oral cavity. Dent
Mater 31: 317–338. doi:10.1016/j.dental.2015.01.006.
Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman
MA, Simonetti DW, Craig S, Marshak DR (1999) Multilineage potential of adult human
mesenchymal stem cells. Science 284: 143–147. doi:10.1126/science.284.5411.143.
164
Bibliografia
Potten CS, Loeffler M (1990) Stem cells: attributes, cycles, spirals, pitfalls and
uncertainties. Lessons for and from the crypt. Development 110: 1001–1020.
Powers CJ, McLeskey SW, Wellstein A (2000) Fibroblast growth factors, their
receptors and signaling. Endocr Relat Cancer 7: 165–197. doi:10.1677/erc.0.0070165.
Rao SM, Ugale GM, Warad SB (2013) Bone Morphogenetic Proteins: Periodontal
Regeneration. N Am J Med Sci 5: 161–168. doi:10.4103/1947-2714.109175.
Reuss B, von Bohlen und Halbach O (2003) Fibroblast growth factors and their
receptors in the central nervous system. Cell Tissue Res 313: 139–157.
doi:10.1007/s00441-003-0756-7.
165
Igor Irastorza Epelde
Roosa SMM, Kemppainen JM, Moffitt EN, Krebsbach PH, Hollister SJ (2010) The
pore size of polycaprolactone scaffolds has limited influence on bone regeneration in an
in vivo model. J Biomed Mater Res A 92: 359–368. doi:10.1002/jbm.a.32381.
Santos F dos, Andrade PZ, Abecasis MM, Gimble JM, Chase LG, Campbell AM,
Boucher S, Vemuri MC, Silva CL da, Cabral JMS (2011) Toward a clinical-grade expansion
of mesenchymal stem cells from human sources: a microcarrier-based culture system
under xeno-free conditions. Tissue Eng Part C Methods 17: 1201–1210.
doi:10.1089/ten.tec.2011.0255.
Scintu F, Reali C, Pillai R, Badiali M, Sanna MA, Argiolu F, Ristaldi MS, Sogos V
(2006) Differentiation of human bone marrow stem cells into cells with a neural
phenotype: diverse effects of two specific treatments. BMC Neurosci 7: 14.
doi:10.1186/1471-2202-7-14.
166
Bibliografia
Sedgley CM, Botero TM (2012) Dental stem cells and their sources. Dent Clin
North Am 56: 549–561. doi:10.1016/j.cden.2012.05.004.
Seo B-M, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, Young M, Robey
PG, Wang C-Y, Shi S (2004) Investigation of multipotent postnatal stem cells from human
periodontal ligament. Lancet 364: 149–155. doi:10.1016/S0140-6736(04)16627-0.
Shyamala K, Yanduri S, Girish HC, Murgod S (2015) Neural crest: The fourth germ
layer. J Oral Maxillofac Pathol 19: 221–229. doi:10.4103/0973-029X.164536.
167
Igor Irastorza Epelde
Smith AG (2001) Embryo-derived stem cells: of mice and men. Annu. Rev. Cell
Dev. Biol. 17: 435–462. doi:10.1146/annurev.cellbio.17.1.435.
Sordi MB, Curtarelli RB, da Silva IT, Fongaro G, Benfatti CAM, de Souza Magini R,
Cabral da Cruz AC (2021) Effect of dexamethasone as osteogenic supplementation in in
vitro osteogenic differentiation of stem cells from human exfoliated deciduous teeth. J
Mater Sci Mater Med 32: 1. doi:10.1007/s10856-020-06475-6.
168
Bibliografia
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS,
Jones JM (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:
1145–1147.
Tziafas D, Smith AJ, Lesot H (2000) Designing new treatment strategies in vital
pulp therapy. J Dent 28: 77–92. doi:10.1016/s0300-5712(99)00047-0.
Vats A, Bielby RC, Tolley NS, Nerem R, Polak JM (2005) Stem cells. Lancet 366:
592–602. doi:10.1016/S0140-6736(05)66879-1.
Vega-Lopez GA, Cerrizuela S, Aybar MJ (2017) Trunk neural crest cells: formation,
migration and beyond. Int J Dev Biol 61: 5–15. doi:10.1387/ijdb.160408gv.
169
Igor Irastorza Epelde
Wagers AJ, Weissman IL (2004) Plasticity of adult stem cells. Cell 116: 639–648.
doi:10.1016/s0092-8674(04)00208-9.
Wang D-R, Wang Y-H, Pan J, Tian W-D (2020) Neurotrophic effects of dental pulp
stem cells in repair of peripheral nerve after crush injury. World J Stem Cells 12: 1196–
1213. doi:10.4252/wjsc.v12.i10.1196.
Wang KC, Helms JA, Chang HY (2009) Regeneration, repair and remembering
identity: the three Rs of Hox gene expression. Trends Cell Biol 19: 268–275.
doi:10.1016/j.tcb.2009.03.007.
Whitman DH, Berry RL, Green DM (1997) Platelet gel: an autologous alternative
to fibrin glue with applications in oral and maxillofacial surgery. J Oral Maxillofac Surg
55: 1294–1299. doi:10.1016/s0278-2391(97)90187-7.
170
Bibliografia
Yang J-Z, Qiu L-H, Xiong S-H, Dang J-L, Rong X-K, Hou M-M, Wang K, Yu Z, Yi C-G
(2020) Decellularized adipose matrix provides an inductive microenvironment for stem
cells in tissue regeneration. World J Stem Cells 12: 585–603.
doi:10.4252/wjsc.v12.i7.585.
171
Igor Irastorza Epelde
Zanicotti DG, Duncan WJ, Seymour GJ, Coates DE (2018) Effect of Titanium
Surfaces on the Osteogenic Differentiation of Human Adipose-Derived Stem Cells. Int J
Oral Maxillofac Implants 33: e77–e87. doi:10.11607/jomi.5810.
Zhang H-T, Liu Z-L, Yao X-Q, Yang Z-J, Xu R-X (2012) Neural differentiation ability
of mesenchymal stromal cells from bone marrow and adipose tissue: a comparative
study. Cytotherapy 14: 1203–1214. doi:10.3109/14653249.2012.711470.
Zhang J, Ding H, Liu X, Sheng Y, Liu X, Jiang C (2019) Dental Follicle Stem Cells:
Tissue Engineering and Immunomodulation. Stem Cells Dev 28: 986–994.
doi:10.1089/scd.2019.0012.
Zhang J, Lian M, Cao P, Bao G, Xu G, Sun Y, Wang L, Chen J, Wang Y, Feng G, Cui
Z (2017) Effects of Nerve Growth Factor and Basic Fibroblast Growth Factor Promote
Human Dental Pulp Stem Cells to Neural Differentiation. Neurochem Res 42: 1015–1025.
doi:10.1007/s11064-016-2134-3.
Zhang N, Chen B, Wang W, Chen C, Kang J, Deng SQ, Zhang B, Liu S, Han F (2016)
Isolation, characterization and multi-lineage differentiation of stem cells from human
exfoliated deciduous teeth. Mol Med Rep 14: 95–102. doi:10.3892/mmr.2016.5214.
172
Bibliografia
173