ArticlePDF AvailableLiterature Review

OsteoMacs: Key players around bone biomaterials

Authors:

Figures

Framework for Describing Activated Macrophages (reprinted with permission from Ref. [125]), (A) Examples of widely used macrophage preparations. CSF-1-grown mouse adherent macrophages from bone marrow (BM) or CD14 þ monocytes are used as the exemplars for marker evaluation and standardized activation conditions. Macrophages can also be generated with GM-CSF, where a CD11c þ dendritic cell (DC) population is also present depending on the culture conditions. In mice, thioglycollate injection followed by peritoneal lavages is used for generating macrophage populations with differing yields and properties, whereas many organ systems in mice and humans are sources of tissue- in fi ltrating macrophages., (B) Marker systems for activated macrophages. Shown are functional subdivisions according to stimulation of mouse CSF-1 macrophages or human monocyte-derived CSF-1 macrophages with the existing M1-M2 spectrum concept [1,99,228]. Stimulation conditions are IL-4, immune complexes (Ic), IL-10, glucocorticoids (GC) with transforming growth factor b (TGF- b ), glucocorticoids alone, LPS, LPS and IFN- g , and IFN- g alone. Marker data were drawn from a wide range of published and unpublished data from the authors' laboratories and represent a starting consensus [229 e 238]. An asterisk indicates corroboration of human IL-4 genes by deep sequencing., (C) Using genetics to aid in macrophage-activation studies. Mutations in Akt1 and Klf4 cause a “ switch ” to M(LPS)- and M(IFN- g )-associated gene expression, whereas mutations in Akt2 and Klf6 show the reverse phenotype. Mutations in Stat6 , Ppard , Pparg , and Irf4 and IRF5 depletion are involved in the maintenance and/or amplitude of activation.
… 
Content may be subject to copyright.
Review
OsteoMacs: Key players around bone biomaterials
Richard J. Miron
*
, Dieter D. Bosshardt
**
Department of Oral Surgery and Stomatology, Department of Periodontology, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
article info
Article history:
Received 1 October 2015
Received in revised form
12 December 2015
Accepted 15 December 2015
Available online 20 December 2015
Keywords:
Macrophage
Bone regeneration
Osteoimmunology
Biomaterial integration
Tissue response
Multi-nucleated giant cells
OsteoMacs
Foreign body cells
abstract
Osteal macrophages (OsteoMacs) are a special subtype of macrophage residing in bony tissues. Inter-
esting ndings from basic research have pointed to their vast and substantial roles in bone biology by
demonstrating their key function in bone formation and remodeling. Despite these essential ndings,
much less information is available concerning their response to a variety of biomaterials used for bone
regeneration with the majority of investigation primarily focused on their role during the foreign body
reaction. With respect to biomaterials, it is well known that cells derived from the monocyte/macro-
phage lineage are one of the rst cell types in contact with implanted biomaterials. Here they demon-
strate extremely plastic phenotypes with the ability to differentiate towards classical M1 or M2
macrophages, or subsequently fuse into osteoclasts or multinucleated giant cells (MNGCs). These MNGCs
have previously been characterized as foreign body giant cells and associated with biomaterial rejection,
however more recently their phenotypes have been implicated with wound healing and tissue regen-
eration by studies demonstrating their expression of key M2 markers around biomaterials. With such
contrasting hypotheses, it becomes essential to better understand their roles to improve the develop-
ment of osteo-compatible and osteo-promotive biomaterials. This review article expresses the necessity
to further study OsteoMacs and MNGCs to understand their function in bone biomaterial tissue inte-
gration including dental/orthopedic implants and bone grafting materials.
©2015 Elsevier Ltd. All rights reserved.
1. Introduction
Monocytes and macrophages are some of the most abundant
cell type found in the bone marrow. Furthermore, they represent
the rst cell types that interact with foreign pathogens and
implanted medical devices. Classical studies have demonstrated
that macrophages are rapidly recruited to infectious and injury
sites where they play critical roles in innate immunity. Here they
demonstrate broad roles and are responsible for regulating tissue
homeostasis including innate and adaptive immunity, wound
healing, hematopoiesis and malignancy [1].
Based on their crucial and distinct roles in tissue homeostasis
and immunity, they are attractive therapeutic targets for a broad
range of pathologies. Furthermore, they are key players in tissue
integration of various biomaterials across a wide range of tissues.
Yet the eld of bone-biomaterial biology has largely omitted their
importance over the years. For instance, a recent systematic review
of dental and orthopedic implants found that over 90% of research
in this area focused primarily on in vitro behavior of osteoblasts on
implant surfaces while only a small percentage (roughly 10%) was
dedicated to immune cells including monocytes, macrophages,
osteoclasts, leukocytes and multinucleated giant cells (MNGCs) [2].
With the advancements made in the eld of osteoimmunology, it
becomes vital to better understand the response of these cell types
to various bone biomaterials. Immune cells play a pivotal role in
determining the in vivo fate of bone biomaterials by either facili-
tating new bone formation around bone-implanted devices but
have also been associated with creating an inammatory brous
tissue encapsulation. It is now understood that macrophages are
the major effector cell in immune reactions to biomaterials where
they are indispensable for osteogenesis. Knockout models have
demonstrated that a loss of macrophages around bone grafting
materials may entirely abolish their osteoinductive potential, thus
conrming their primary role in the immune system modulation
later responsible for guiding osteogenesis [3].
Over the years, complex studies from basic research have
revealed the dynamic interactions between the skeletal system and
immune system [4e6]. It has been shown that a population of
*Corresponding author. University of Bern, Head of Oral Cell Biology,
Switzerland.
** Corresponding author. University of Bern, Head of Oral Histology, Switzerland.
E-mail addresses: richard.miron@zmk.unibe.ch (R.J. Miron), dieter.bosshardt@
zmk.unibe.ch (D.D. Bosshardt).
Contents lists available at ScienceDirect
Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials
http://dx.doi.org/10.1016/j.biomaterials.2015.12.017
0142-9612/©2015 Elsevier Ltd. All rights reserved.
Biomaterials 82 (2016) 1e19
tissue macrophages named OsteoMacsresides within bone as a
distinctive canopy structure overlying mature osteoblasts [6].
Although initial bone fracture healing experiments have been
characterized by inltration of inammatory cells, most of these
initial studies focused primarily on the secretion of various cyto-
kines and growth factors important for the inammatory process
including cell recruitment [7e9] and neovascularization [10].
Although macrophages in general have been implicated as key
contributors to inammation, a series of experiments have also
revealed their essential roles in bone repair with recent ndings
demonstrating that even MNGCs may be categorized with a tissue
repair phenotype by demonstrating release of M2-related cyto-
kines and growth factors [11]. Thus, the differentiation of mono-
cytes towards M1 or M2 macrophages, as well as their fusion to
osteoclasts or MNGCs in response to various biomaterials remains
extremely poorly understood. Furthermore, the main factors
responsible for directing their phenotypes towards more special-
ized cell-types in response to biomaterials also remains poorly
characterized.
Human histological samples from our dental clinic using a variety
of bone grafting materials for bone augmentation procedures have
consistently shown a substantially high number of MNGCs around
bone substitute materials grafts in stable situations harvested years
after original surgeries were performed [12]. Furthermore, a select
class of bone substitutes grafts consistently associated with higher
than average maintenance of bone mass in grafted sites, are
routinely found with signicantly higher numbers of MNGCs (Fig.1).
This has led our research team to further question the role of MNGCs
on biomaterials as these cells were once thought to only contribute
to the foreign body reaction [11]. Interestingly, studies investigating
atherosclerotic plaque have provided evidence that macrophages
very commonly fuse into MNGCs (also termed foam cells) that
enhance calcied tissues surrounding arterial walls; an area that
otherwise should not produce any mineralized tissues [13e17].
While the production of mineralized tissues from MNGCs in
atherosclerosis leads to a pathological state, recently our group has
questioned whether this situation might be advantageous around
bone biomaterials. Thus, it is clear that a substantial amount of
additional work is needed with respect to understanding macro-
phage and MNGCs function especially as it relates to bone bio-
materials. It may be possible that MNGCs in certain situations
leading to a pathological state (e.g. calcied tissues around arteries)
might be therapeutic in others (bone biomaterials).
As part of an overview on the current knowledge regarding
immune cells and bone biomaterials, this review article aims to: 1)
Characterize and review the key basic science studies involving
OsteoMacs that demonstrate their pivotal role in bone biology. 2)
Provide background knowledge on monocytes and the great po-
tential for these cells to differentiate into a variety of cell-types
including M1 and M2 macrophages, MNGCs, FBGCs and osteo-
clasts. 3) Review the current literature on monocyte/macrophage
studies with respect to bone biomaterials including bone grafts and
dental/orthopedic implants. 4) Provide additional evidence from
calcied atherosclerotic plaque that macrophages/MNGCs are
potent inducers of mineralization by demonstrating that even in a
pathological state, macrophages/MNGCs are the responsible cell-
type contributing to calcication in arteries. 5) Demonstrate evi-
dence from animal and human histological samples from our
research center that MNGCs are routinely found around bone bio-
materials in high numbers and commonly associated with the
maintenance of high bone volume leading to the hypothesis that
these cells may very well be one of the key players responsible for
the maintenance of bone homeostasis.
2. OsteoMacs: biological basis and key roles in bone
formation
The term OsteoMacswas originally given by a group of basic
researchers in Australia led by Allison Pettit. Original observations
described in the mid 1980s sought to characterize the role of osteal
macrophages in bone biology [18]. Hume et al. were one of the rst
to clearly demonstrate that periosteal and endosteal tissues con-
tained a discrete population of resident tissue macrophages in line
with traditional bone cell nomenclature [5,6]. OsteoMacs constitute
approximately one sixth of all cells residing in bone marrow and
display a stellate morphology allowing them to achieve extension
coverage of bone surfaces suggesting that they may form a
comprehensive communication network [6]. It is clear now that
this subset of CD68þcell-type is derived from a resident population
of macrophages like macrophages found in other tissues [19e21].
More recent research has clearly conrmed that macrophages may
subdivide and proliferate from resident tissues contrasting original
theories expressing that these cells are derived from monocyte
precursors from the blood-stream [22e25].
The general role of OsteoMacs has been described as immune
surveillance cells in the bone microenvironment. A number of
previous studies have demonstrated that this subset of macro-
phages are able to function as phagocytes [26,27], are capable of
detecting bacterial products [28,29], and respond to antigens
[27,30]. In vitro cell culture systems have further provided evidence
by demonstrating how primary murine osteoblast cultures are able
to respond to pathophysiological levels of lipopolysaccharide (LPS),
characteristics of the M1 macrophage later discussed in this article
[6]. These observations as well as others report the potential cross-
Fig. 1. MNGCs on bone substitute materials including (A, D) Bio-Oss
®
(HA), (B, E) Straumann
®
BoneCeramic (BCP), and (C, F) NanoBone
®
(HA-silica gel). LM (AeC) and TRAP
histochemistry (DeF).
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e192
lineage plasticity and crosstalk between osteoblasts and hemato-
poietic cells in vitro [31e33], which adds difculty to clearly assign
the cells responsible for specic functions reported in primary
osteoblastcultures. This has since best been exemplied in a study
by Chang et al. who showed that by removing macrophages from
primary osteoblast cultures, a 23-fold reduction in mineral depo-
sition was observed [6]. It was concluded that it was the OsteoMacs,
and not the osteoblasts as originally hypothesized, within these
in vitro culture systems that responded to pathophysiological
concentrations of cytokines and their removal from calvarial cul-
tures signicantly decreased in vitro mineralization by osteoblasts
[6].
2.1. Are OsteoMacs precursors to osteoclasts and multinucleated
giant cells (MNGCs)?
Due to common precursors [34] and anatomical location, an
obvious hypothesis would be to assume that OsteoMacs in osteal
tissues serve as precursors for osteoclasts and MNGCs. In bone
tissues, osteoclasts have historically been characterized by their
multinucleated phenotype and ability to resorb bone [35]. More
recently however, MNGCs have been found in bone tissues with
slightly different histological appearance largely characterized by
their inability to quickly resorb bone grafts [36]. Differences in
surface markers and gene expression between osteoclasts and
MNGCs are discussed later in this article. To date, the differentiation
and transcription factors responsible for the formation of osteo-
clasts versus MNGCs from precursor cells in bone tissues has
remained poorly characterized.
Although OsteoMacs are clearly not osteoclasts (both conrmed
by the lack of multinucleation and lack of expression of common
osteoclast markers such as F4/80 antigen) [34,37], their plasticity in
situ suggests that these cells can further respond to stimuli by
locally differentiating towards various multinucleated cell pheno-
types. In vitro research has demonstrated that bone marrow-
derived macrophages [38] and rheumatoid synovium-derived
macrophages [39] can be differentiated into osteoclasts conrm-
ing that mature macrophages can at least in vitro, serve as multi-
nucleated cell precursors. Alternatively, it is well known that
more undifferentiated cells from the myeloid lineage also available
within bone can more efciently differentiate into multinucleated
cells [40]. These ndings have certainly argued against OsteoMacs
being the preferredmultinucleated precursor cell in basic bone
biology research [41]. Very recent research has demonstrated that
MNGCs express many M2 macrophage-like markers implicated in
wound healing and tissue regeneration and it was suggested that
this specialized cell type resembled predominantly an M2 wound
healing OsteoMac [11]. Yet it has also been argued that OsteoMacs
may contribute to the osteoclast precursor pool and MNGCs under
pathological conditions. Thus, it remains somewhat unclear how
each terminal cell type from the monocyte lineage is capable of
fully differentiating under various conditions/micro-environments.
It is evident that macrophages demonstrate a great phenotypic
plasticity in vivo and seem to respond to varying stimuli by
differentiating towards various cell types at least in vitro. Never-
theless, there is clearly a lack of knowledge for which factors guide
these cells to differentiate and/or fuse into MNGCs under various
conditions. Ongoing research in this area is urgently needed.
Currently, it is known that activated OsteoMacs are able to
produce a wide variety of either pro- or anti-inammatory cyto-
kines leading to multinucleated cell formation. OsteoMacs have
been shown to secrete TNF-alpha [42,43], interleukin IL-6 [44], IL-1
[42,45] and interferon-
b
[46,47] in response to various in vitro
conditions. The number of factors released by OsteoMacs demon-
strates their ability to regulate cell fusion of monocytic cells and
their subsequent differentiation. How OsteoMacs specically
respond to various biomaterials will be discussed later in this
article.
2.2. OsteoMacs function in osteoblast mineralization
The preliminary nding from primary osteal tissues clearly
demonstrated that OsteoMacs play a pronounced role in osteoblast
function and differentiation by demonstrating a 23-fold decrease in
mineralization potential when OsteoMacs were removed from
in vitro culture systems [6]. Interestingly, depletion of OsteoMacs
in vivo by various knockout systems has also been shown to
markedly reduce bone formation [6,48]. These previous studies are
similar to a number of investigations over the years describing
osteoclasteosteoblast coupling mechanisms in vivo [49e51].
Interestingly in the mid-2000s, it was shown that macrophage-
specic genes (csf1r, CD14) are induced in primary mouse chon-
drocyte differentiation cultures [52] raising the possibility that
macrophages may also be heavily implicated in chondrogenesis, a
key step in endochondral ossication. This hypothesis was later
conrmed in a recent macrophage knockout system [48]. Previous
reports have also shown that macrophages may produce a number
of potent bioactive growth factors for osteoblasts including trans-
forming growth factor
b
(TGF-
b
)[53], osteopontin [54], 1,25-
dihydroxy-vitamin D3 [55] and BMP-2 [56]. These factors are
known inducers of extracellular matrix deposition and new bone
formation and are classical characteristics of the M2 macrophage.
The plasticity of macrophages suggests that their trophic role in
bone tissues are highly regulated by changes to the microenvi-
ronment. OsteoMacs are capable of promoting anabolic function in
certain conditions whereas in others they are responsible for
creating and directing an inammatory environment.
2.3. OsteoMacs and bone modeling
Bone modeling is an anabolic process involving new bone
deposition and is unlike bone remodeling, which involves the
careful and coordinated balance between osteoclasts and osteo-
blasts [51,57]. It has previously been reported that macrophages are
found localized at the bone modeling site on cortical diaphyseal
endosteal bone surfaces without the presence of osteoclasts in the
vicinity [6]. This process has been described as forming a canopy-
like cell structurewhere OsteoMacs were seen encapsulating the
functionally mature osteoblasts suggesting that they are heavily
implicated in the bone modeling process [6,58]. Once again, the
functional importance of OsteoMacs was demonstrated by
knockout systems where macrophages were depleted using a Fas-
induced apoptosis (Maa) transgenic mouse model, which can
induce macrophage depletion via synthetic ligand treatment [59].
In this system, OsteoMac canopy architecture was disrupted lead-
ing to a complete loss of mature osteoblasts and bone modeling at
the bone interface [59].
It was originally proposed that during bone remodeling, osteo-
clasts provide a coupling signalto promote and coordinate oste-
oblast activity [51]. Interestingly with the numerous advancements
made in the eld of osteocyte biology, it has recently been proposed
that osteocytes are also implicated in the bone remodeling process
by dictating both osteoblast and osteoclast activity [60]. Given that
the bone modeling animal models lack osteocytes and osteoclasts
during the developmental stages of bone modeling, it was pro-
posed that OsteoMacs may be the cells responsible for coupling-
like signals dictating osteoblast function. While evidence from
the literature has previously suggested that TGF-
b
and ephrin B2
are implicated as possible coupling factors between osteoclasts and
osteoblasts [51,61e63], macrophages have also been shown to
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 3
produce TGF-
b
[53] and ephrin B2 [64,65] opening the possibility
that OsteoMacs are also capable of fullling such roles. Neverthe-
less, a great deal of research is still necessary to further understand
the role of OsteoMacs during bone modeling where there is an
absence of osteoclasts and osteocytes.
It is interesting to note that depletion of OsteoMacs in vivo using
the macrophage Fas-induced apoptosis (Maa) mouse caused a
complete loss of osteoblast bone formation at the bone surface
demonstrating that OsteoMacs are an integral component of bone
tissues and play a pivotal role in bone homeostasis [6]. These pro-
posed models show that OsteoMacs have as function to both survey
for alteration in the local environment as well as guide bone for-
mation in vivo. In response to anabolic stimuli, they function to
recruit mesenchymal progenitor cells and induce their proliferation
and differentiation towards bone forming osteoblasts. They sub-
sequently provide ongoing anabolic signals to the underlying os-
teoblasts [6]. It has been further proposed that once the stimulus is
removed, OsteoMacs within the canopy withdraw and either
migrate to a new area, or undergo apoptosis whereas the under-
lying mature osteoblasts revert to a bone lining cell phenotype,
become embedded within bone as osteocytes or undergo apoptosis
[6].
2.4. OsteoMacs and bone remodeling
As described earlier, bone remodeling implicates the ne bal-
ance between bone-resorbing osteoclasts and bone-forming oste-
oblasts [51,57]. Resorption signals including RANKL and CSF-1 are
expressed by bone lining cells and osteocytes, and are necessary for
directing osteoclastogenesis and bone resorption. It was previously
proposed that osteoclasts subsequently provide the coupling signal
coordinating osteoblast activity to facilitate bone deposition and
mineralization [51]. It has been reported that during this process,
osteoclasts are only located at the leading edge of the formation
phase and have moved or undergone apoptosis before new bone
formation is completed [66]. Therefore, certain investigators have
posed the question: what cellular/molecular mechanism drives
osteoblasts to initiate mineralization and complete the remodelling
cycle following osteoclast apoptosis?[67].
As previously mentioned, OsteoMacs have been shown to form a
cellular canopy structure around osteoblasts during bone modeling.
This process was postulated to create an enclosed compartment for
local communication and coordination during the complex
remodelling process [68]. Although it is proposed that osteoclasts
likely have the dominant role in orchestrating the recruitment,
proliferation and initial differentiation of pre-osteoblasts during
bone remodeling based on the release of cytokines from resorbed
bone, the various roles of OsteoMacs in combination with their
anatomical location and canopy architecture have recently postu-
lated the idea that they may also be a necessary requirement for
optimal mineralization by osteoblasts [6]. Furthermore, due to their
close proximity to bone surfaces and well-known ability to detect
dying cells [69], OsteoMacs are an obvious candidate to detect and
respond to bone damage, a critical event for osteoclast recruitment,
thus initiation the bone remodeling phase [70].
More recently, it has been demonstrated that bone marrow
macrophages are responsible for maintaining hematopoietic stem
cell (HSC) niches and that subsequent depletion facilitates the
mobilization of HSCs [23]. Winkler et al. investigated the regulation
of endosteal niches, by studying the mobilization of HSCs into the
blood-stream in response to granulocyte colony-stimulating factor
(G-CSF) [23]. They found that G-CSF rapidly depletes endosteal
osteoblasts leading to the suppression of endosteal bone formation
and decreases the expression of factors required for HSC retention
and self-renewal. G-CSF administration also depleted a population
of OsteoMacs found in endosteal tissues, which subsequently dis-
rupted osteoblast function. Furthermore, using 2 separate animal
models to deplete OsteoMacs (Maa transgenic mice or adminis-
tration of clodronate-loaded liposomes) it was found that 1) there
was a loss of endosteal osteoblasts, 2) there were marked re-
ductions of HSC trophic cytokines at the endosteum and 3) HSC
mobilization into the blood occurred. Furthermore, RT-PCR ana-
lyses on the endosteal RNA from treated Maa mice revealed a 35-
fold decrease in osteocalcin expression when compared to controls.
Taken together these results demonstrate the pivotal role of
maintaining endosteal HSC niches that subsequently leads to
downstream OsteoMac function and osteoblast bone formation
[23].
In a similar study, Alexander et al. demonstrated also that osteal
macrophages promote in vivo intramembranous bone healing in a
mouse tibial model [71]. The authors used a very similar approach
to the previous study by knocking out macrophage populations
using Maa mice and clodronate liposome delivery. Following tibial
injury, they demonstrated that the depletion of OsteoMacs led to
signicantly reduced intramembranous ossication bone healing
whereas administration of CSF-1 in the animal models led to an
increase in OsteoMac number at the injury site, which concurrently
increased new matrix deposition and mineralization [71]. A study
with a similar animal model also demonstrated that fracture
healing via periosteal callus formation also requires OsteoMacs for
both the initiation and progression of early endochondral ossi-
cation [48]. Furthermore, a separate group found that depletion of
macrophages using Maa mice led to early skeletal growth retar-
dation and progressive osteoporosis (25% reduction in bone min-
eral density, 60% reduction in number of mesenchymal progenitor
cells) by 3 months [72]. Of particular interest, animals that were
treated with anabolic factors such as PTH showed a signicantly
higher level of OsteoMacs further suggesting their important role in
bone remodeling [73].
It is difcult to assess technically whether osteoclasts or mac-
rophages are more important for bone remodeling and regulating
osteoblast activity. The main reason is that most of the mutations to
date that affect macrophages also have a large impact on osteo-
clasts, since they are derived from the same precursor cells [74].Itis
therefore extremely difcult to knock down just macrophages
without compromising osteoclast activity [74]. In contrast, it is
possible to abolish osteoclasts by specically targeting OPG by
blocking the actions of RANKL [75]. Since there is a close lineage
relationship between macrophages and osteoclasts [34], the cur-
rent in vivo models could benet from future rening as consid-
erable cellular plasticity is found between these 2 cell types [38].
Further specic investigation on the role of OsteoMacs versus os-
teoclasts and their contribution to bone remodeling is needed to
clearly delineate all the cellular participants and molecular factors
in osteoblast coupling.
3. Monocyte/macrophage characterization and
differentiation
The goal of the present review article is not to fully detail
monocyte differentiation towards their numerous downstream cell
types. For an excellent summary on these events/topic, the reader is
kindly directed to the following extensive review articles [1,76e78].
Instead we focus on presenting a broad overview on the current
differentiation parameters seen in monocytes and specically look
at key markers and gene expression patterns of macrophage pop-
ulations, MNGCs and osteoclasts.
While the role of monocyte-derived macrophages during
pathological tissue inammation has recently been comprehen-
sively reviewed [79], recent research has demonstrated that most
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e194
monocytes do not substantially contribute to macrophage pop-
ulations in the steady state. Until recently, a fundamental dogma in
immunology was reversed whereby it was originally hypothesized
that all tissue-resident macrophages were derived from a constant
recruitment of blood monocytes, as originally described by van
Furth and Cohn [80]. More recently however, data from a number of
studies have challenged these original ndings and shown that the
majority of tissue resident macrophages including OsteoMacs are
self-renewable within resident tissues [22e25]. Since macrophages
have many functions in development, tissue homeostasis and the
resolution of inammation, accordingly there is much interest in
potentially manipulating these cells for therapeutic benet[76].For
this to occur, better understanding over their broad plasticity and
key parameters inducing their differentiation towards certain
subsets is absolutely necessary.
Colony-stimulating factor 1 (CSF1, also known as M-CSF) is the
growth factor by which monocyte development is completely
dependent. Mice that are decient in CSF1 or its receptor CSF1R
(CD115) exhibit severe malformations and monocytopenia [81e83].
Therefore, perhaps the single most important gene to the mono-
cytic lineage is CSF1, as macrophages, osteoclasts and MNGCs are all
dependent on its expression for survival.
The existence of monocyte subsets in human blood in the late
1980s described 2 main subpopulations dened as CD14
þ
mono-
cytes (which can be further subdivided into distinct populations of
CD14
þ
CD16
þ
and CD14
þ
CD16
monocytes) and CD14
low
CD16
þ
monocytes [84]. Cell differences in the monocyte lineages have
been observed between species and most notably between humans
and mice. In mice, LY6C
low
cells are the equivalent of human
CD14
low
CD16
þ
and may therefore be terminally differentiated to-
wards resident macrophages [85,86]. By contrast, mice LY6C
hi
monocytes are the equivalent of CD14
þ
monocytes in humans
representing the classical monocytes recruited to sites of inam-
mation [87]. Both of these subsets of monocytes may give rise to
macrophages.
Early studies have long considered macrophages as long-lived,
terminally differentiated cells with little capacity to proliferate
[88,89], however, recent ndings that macrophages can self-renew
in resident tissues such as bone contradicted these original nd-
ings. In certain settings, IL-4 has been identied as the major factor
inducing macrophage proliferation [90] and CSF-1 has also been
shown to promote proliferation as well as monocyte recruitment
[91].
Since the survival, proliferation and differentiation of macro-
phages is dependent on CSF-1 [92], it is also known to be more
highly expressed during infection, inammation and tissue injury
leading to the rapid recruitment of monocytes to induce their dif-
ferentiation towards macrophages that drive innate and adaptive
immune responses [19]. Macrophages and bone resorbing osteo-
clasts are closely related however can be distinguished by the
presence of additional nuclei in osteoclasts as well as morpholog-
ical features and expression of marker proteins later discussed
[19,93].
Macrophages have a large capacity to secrete a wide range of
cytokines and regulatory molecules in response to their micro-
environment [81,94,95]. The most widely used antibody to iden-
tify macrophages in murine tissues has been the F4/80 monoclonal
antibody [37]. This marker is also used to distinguish them from
osteoclasts as F4/80 is rapidly down-regulated during
osteoclastogenesis.
Initial macrophage experiments identied macrophages into 2
specic cell types, classical M1 pro-inammatory macrophages and
M2 tissue resolution/wound healing macrophages (Fig. 2). Classical
pro-inammatory stimuli in response to LPS include TNF-alpha
[42,43], IL-6 [96,97] and IL-1
b
[42,45] all contributing to tissue
inammation and osteoclastogenesis. M2 macrophages typically
produce TGF-
b
and arginase, both factors implicated in tissue-
repair processes [98e101].Table 1 presents a general overview of
differences observed between M1 and M2 macrophages.
Fig. 2 presents an overview of general cell-types derived from
the monocyte lineage. In vitro differentiation of macrophage to-
wards the M1 phenotype can best be produced with IFN-
g
þþLPS
and TNF-
a
, whereas M2 macrophages are typically produced with
either IL-4 or IL-13 [78]. The in vitro culture with IL-4 causes up-
regulation of 2 key M2 markers including TGF-
b
and arginase
largely assumed to participate in tissue regeneration [78,98e101 ] .
It has also been shown that IL-4 increases the expression of the
mannose receptor CD206. Whereby once M2 macrophages
included a wide variety of characteristics as originally dened,
more recent research has subdivided their classication in M2a/b/c
to further express the differences found between certain M2
macrophages [102] (Fig. 2). Briey, the M2a phenotype is produced
by exposure to IL-4 þIl-13 acting through IL-4R
a
to increase the
expression of CD206, arginase, and TGF-
b
[99,103e106] . The M2b
phenotype has been described following exposure to a combination
of IgG-immune complexes and IL-1R ligands in turn increasing IL-
10 production and decreasing IL-12 largely contributing to anti-
inammatory properties [107,108]. Cell culture with IL-10 or glu-
cocorticoids produces the M2c phenotype characterized by high IL-
10 and low IL-12 production [108], as well as increased surface
receptor CD163 [109,110]. While the aim of this review article is not
to give a background on the specicity of the various M2 macro-
phage subgroups, it remains important to note that various cell
culture models have distinct M2 macrophage characteristics. For an
excellent overview on this topic, the reader is kindly directed to a
recent review article on M1 and M2 paradigm of macrophage
activation [1].
Phenotyping macrophages has typically been carried out with
cell surface markers including CD11b, CD68, macrophage antigen-
2, and F4/80. Interestingly, intensive research over the last decade
has provided new and improved ways for further phenotyping
macrophages based on their gene-expression proles in response
to various stimuli. As such, early functional studies of macrophage
phenotyping noted that gene-expression plasticity existed when
macrophages were polarizedto either the M1 or M2 phenotypes
[108,111e113 ] . Gene expression makes sense in these scenarios as
macrophages are likely more responsive to various stimuli such as
infection, cell death, implanted biomaterials yet in general seem to
maintain the plasticity to subsequently return to more polarized
states based on signals from their micro-environment. Despite the
research that has been performed on this topic over the past
decade, one common reported difculty in the literature with
respect to macrophage phenotyping is the fact that major differ-
ences between human and mice macrophages exist complicating
the overall progression of the eld of osteoimmunology as dis-
cussed below.
3.1. Macrophage phenotyping - mouse versus human macrophages;
how different are they?
One of the most controversial aspects of macrophage biology
that has certainly limited research progress is the concern over
perceived differences between mouse and human macrophages.
Some researchers have gone to such lengths as to suggest that
human macrophages are fundamentallydifferent from their
mouse counterparts and thus should be studied as entirely separate
entities [114,115].
An example of this is the fact that mouse M2a macrophages
have been phenotypically characterized by their expression of
arginase, an important component of tissue regeneration [99,116].
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 5
Interestingly however is the fact that M2a macrophages in humans
do not express arginase at all [117,118]. In general, arginase is highly
expressed during injury [119e122] and is thought to contribute to
collagen production during wound healing [119,123,124]. The dif-
ferences between human and mouse macrophages extends beyond
the scope of this review article, however a brief list of reported
differences is compiled in Fig. 3 [125].
In summary, it is evident that there exists a lack of information
that has thus far prevented progress in macrophage biology.
Furthermore, evidence that demonstrates quite signicant differ-
ences between human and mouse cell surface markers adds to the
complexity encountered studying their phenotypes [108,126e129].
Ideally knockout animals are utilized to determine the role of
specic genes in biology. However, based on the perceived differ-
ences between species, some have questioned this line of research
is even valid or necessary at all with respect to macrophage biology.
Although in general many studies demonstrate an M1 macrophages
phenotype by demonstrating the expression of IL-1B, IL-6, IL-8,
TNF-alpha, and IFN-gamma to promote inammation, or M2
macrophages by producing IL1Ra, IL-4, IL-10 and arginase to pro-
mote inammatory resolution, future research is necessary to
determine the differences between human and mouse phenotypes
as there are perceived differences from the literature
[99,102,130e133].
Fig. 2. Monocyte differentiation including expression markers into Osteoclasts, M1, M2a, M2b, M2c macrophages and MNGCs.
Table 1
Summary of in vitro culture conditions of M1 and M2 macrophages.
M1 macrophage M2 macrophage
Activator IFN-gamma, TNF-alpha, LPS IL-4, IL-13
Proinammatory cytokines IL-1B, TNF-alpha, IL6, IL12 Low
iNOS High (in rodents only) Low
Anti-inammatory cytokines Low TGF-B high, IL-10 Low
CD206 Low High
Dectin-1 Low High
Ym1 Low High
Phagocytosis/Endocytosis High Decreased phagocytosis of implanted particles
Matrix proteins MMP9 FN, TGFB1, MMP1, MMP12, TG, F13A1
Markers
Human: CD64, IDO, SOCS1, CXCL10 MRC1, TGM2, CD23, CCL22
Mouse: CXCL9, CXCL10, CXCL11, NOS2 Mrc1, tgm2, FizzI, Ym1/2, Arg1
Transcription factors
Human: pSTAT1, IRF5 IRF4, SOCS1*, GAT3* SOCS3
Mouse: pSTAT1, pSTAT6-ve, Socs1 pSTAT6, pSTAT1-, Soc2
Cytokines
Human: TNF, IL6, IL1b, IL12A, IL12b, IL23A IL-10
Mouse: TNF, IL-6, IL-27, Tnf23a IL-10, IL-6
Chemokines
Human: CXCL10, IL8, CCL5, CXCL9, CCL4, CCL13, CCL17, CCL18,
Mouse: CXCL11, CCL18-ve CCL17 CCL24, CXCL13, CCL1, CCL22, CCL20
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e196
3.2. Giant-cell formation and function
It is well recognized that multinucleated giant-cell (MNGC)
formation is the result of cells derived from the monocyte/macro-
phage lineage. While various names have been given to these cell
types over the years (including foreign body cell (FBC), foreign body
giant cell (FBGC), multinucleated cell (MNC), multinucleated giant
cell (MNGC), giant-body foreign cell (GBFC), or foamcell), it is
important to note that very early experiments dealt primarily with
their characterization in response to pathogens. In vitro models
demonstrated that large MNGCs with more than 15 nuclei were
found in response to high-virulence mycobacterium whereas low-
virulence mycobacterium consistently produced low numbers of
nuclei per cell (less than 7) [134]. These studies point to the fact that
monocyte/macrophage stimulation in response to pathogens
formed MNGCs, which were at the time described as FBGCs. These
Fig. 3. Framework for Describing Activated Macrophages (reprinted with permission from Ref. [125]), (A) Examples of widely used macrophage preparations. CSF-1-grown mouse
adherent macrophages from bone marrow (BM) or CD14
þ
monocytes are used as the exemplars for marker evaluation and standardized activation conditions. Macrophagescan also
be generated with GM-CSF, where a CD11c
þ
dendritic cell (DC) population is also present depending on the culture conditions. In mice, thioglycollate injection followed by
peritoneal lavages is used for generating macrophage populations with differing yields and properties, whereas many organ systems in mice and humans are sources of tissue-
inltrating macrophages., (B) Marker systems for activated macrophages. Shown are functional subdivisions according to stimulation of mouse CSF-1 macrophages or human
monocyte-derived CSF-1 macrophages with the existing M1-M2 spectrum concept [1,99,228]. Stimulation conditions are IL-4, immune complexes (Ic), IL-10, glucocorticoids (GC)
with transforming growth factor
b
(TGF-
b
), glucocorticoids alone, LPS, LPS and IFN-
g
, and IFN-
g
alone. Marker data were drawn from a wide range of published and unpublished
data from the authors' laboratories and represent a starting consensus [229e238]. An asterisk indicates corroboration of human IL-4 genes by deep sequencing., (C) Using genetics
to aid in macrophage-activation studies. Mutations in Akt1 and Klf4 cause a switchto M(LPS)- and M(IFN-
g
)-associated gene expression, whereas mutations in Akt2 and Klf6 show
the reverse phenotype. Mutations in Stat6,Ppard,Pparg, and Irf4 and IRF5 depletion are involved in the maintenance and/or amplitude of activation.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 7
unhappymacrophages seemed to fuse in response to foreign
pathogens and this has been the basis over the last several decades
for terming these cells as FBGCs. Today, much has been learned
with respect to multinucleated giant cells. Many aspects of their
behavior have been discovered including their recognition motifs,
adhesion, fusion and activation molecules as well as specic
intercellular and intracellular signaling pathways [135,136]. Yet a
great deal of information is still lacking regarding their cellular
control which is vital to further utilize macrophages as therapeutic
targets for the treatment of various pathologies associated with
their misbalance. Below we describe osteoclast differentiation, and
later contrast their differences with MNGCs (Table 2).
3.3. Osteoclast formation and function
Simply put, osteoclasts are the multinucleated counterpart to
osteoblasts responsible for bone resorption. Along with osteoblasts,
they play a pivotal role in bone homeostasis and bone remodeling
by continuously maintaining a steady state between bone forma-
tion and bone resorption. Disruption in their activity has the po-
tential to cause major systemic conditions such as osteoporosis
characterized by low bone mass typically found in post-
menopausal women [137]. Interestingly, osteoclast precursors are
also derived from the bone marrow like early monocytes and
macrophages that circulate through blood and bind to the surface
of bone. Although the exact molecular mechanism involved in the
recruitment and targeting of osteoclasts to bone surfaces remains
largely unknown, it has been shown that integrin [alpha]
v
[beta]
3
is
the dominant osteoclast binding domain and one of the typical
markers used to identify the osteoclast phenotype but is absent on
macrophage precursors however progressively induced by RANKL
[138]. These integrins recognize a number of ECM molecules found
in bone including osteopontin, bronectin, vitronectin and brin-
ogen which typically bind through an RGD peptide domain
[139,140].
Following adhesion to bone, monocyte precursors and macro-
phage precursors absolutely require 2 main cytokines for their
differentiation to osteoclasts, CSF1 and RANKL. Animals lacking
CSF1 are osteopetrotic and have severe growth retardation
[67,141,142]. Furthermore, mutation in CSF1 receptor has a similar
phenotype [82]. While CSF1 is also required for macrophage dif-
ferentiation, RANKL is typically considered the master gene for
osteoclast differentiation and regulation [143]. OPG is another
peptide synthesized by osteoblasts and osteocytes known to
recognize RANKL and thus acts as a decoy receptor competing with
RANK [75]. It has been shown that overproduction of RANKL can
lead to osteoporosis, whereas osteopetrosis is caused by increased
OPG production.
One question that remained following these key studies was
how/what factors were responsible for the fusion of multinucleated
cells. Yagi et al. showed in a knockout animal model that dendritic
cell-specic trans-membrane protein (DC-STAMP) was required for
the fusion of both osteoclasts and MNGCs [144]. It was observed
that osteoclasts found in DC-STAMP knockout animals were
mononuclear, maintained their bone-resorbing activity, expressed
osteoclast markers and cytoskeletal structure but were not capable
of cell fusion [144]. Further experiments using retroviral intro-
duction of DC-STAMP in mononuclear osteoclast precursors
showed the re-establishment of the multinucleation found in cells
derived from DC-STAMP knockout animals further conrming the
Table 2
Contributing factors and differences between osteoclasts and multi-nucleated giant cell activation, adhesion and fusion (adapted from Brodbeck and Anderson 2009).
Contributing factor Osteoclasts Multinucleated giant cell
Adhesion substrate Bone
Dentin
Implanted biomaterial
Adsorbed surface proteins Osteopontin
Vitronectin
Fibrin(ogen)
Bone sialoprotein
Complement component (iC3b)
Vitronectin
Fibrin(ogen)
Adhesion receptors
a
V
b
3
a
M
b
2
,
a
X
b
2
CD47
a
5
b
1,
a
3
b
1
a
5
b
1
,
a
V
b
1
CD44
ICAM-1
Soluble fusion mediators CCL-2
RANKL
M-CSF
TNF-alpha
IL-1
CCL-2
IL-4
IL-13
INF-
g
IL-3
Con A
PHA
MMP9
Cell surface fusion mediator DC-STAMP DC-STAMP
Cell fusion receptors CD44, CD47, CD200, signal regulatory protein 1a, IL-4r, E-cadherin, mannore receptor
MFR MFR
CD48 Mannose receptor (CD26)
AvB3
RANKL
E-Cadherin
CD44, CD81, CD9
Connexin 43
CD13 (aminopeptidase N)
Galectin-3
E-Cadherin
CD44, CD81, CD9
Connexin 43
P2X7 receptor
Presenilin 2
Phenotypic expression Cathepsin K
Acid
CD13þ, CD14þ, CD68þ, CD56-
GrB-, Ki67-
Phagocytosis (frustrated)
Acid
Reactive oxygen intermediates
Lymphocyte co-stimulators
(HLA-DR, B7-2, B7eH1, CD98) CD44 (HCAM)
CD13þ, CD14þ, CD68þ, CD56-
GrBþ, Ki67þ
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e198
essential role of DC-STAMP on cell fusion. Since then, forced oste-
oclast differentiation using CSF1, RANKL and IL-4 (a known inducer
of cellecell fusion) were incapable of initiating cell fusion in DC-
STAMP knockout cells in vitro [145]. Since these ndings, DC-
STAMP has been investigated as a therapeutic option to minimize
MNGC formation both as a surface protein expressed by macro-
phages or as a released soluble factor. Research in this area is
ongoing.
3.4. Multinucleated giant cell (MNGC) formation and function
To date, most of the literature available on MNGCs involves their
role around tissue/biomaterial interfaces of implanted medical
devices [136]. From this point of view, numerous attempts have
been made to characterize these cells as FBGCs, as part of a foreign
body reaction. While these cells are not found abundantly in
normal physiological tissues, they are found in great number
around bone biomaterials with their roles highly left unstudied.
Fig. 4 presents a summary of the events taking place between the
interactions of MNGC, biomaterials, and exudate/tissue inamma-
tion. Interestingly, MNGCs have also been seen in several tissues
where the size of the foreign particle is greater than permitted for
macrophage phagocytosis to occur [146]. Thereafter, macrophages
have been suggested to fuse in response to this larger than average
particle size [146];afrustratedmacrophage. In the context of
biomaterial integration, it has been accepted that FBGCs are
generated by macrophage fusion and serve the same purpose as
osteoclasts, degradation/resorption/removal of the underlying
substrate[147]. While we do not oppose this view, it is interesting
to point out that MNGCs/FBGCs are also found in response to
implanted bone grafting materials and bone-implant titanium
biomaterials. Therefore, if these cells serve the same purpose as
osteoclasts in bone, why are these cells found in bone altogether?
A great deal of information on MNGCs originates from implan-
ted medical devices in soft tissues. It has been shown that beta1 and
beta2 integrin receptors are the predominant binding domains
during monocyte to macrophage development and that IL-4 in-
duces cellecell fusion during FBGC formation [148]. Surface re-
ceptors during fusion include CD44, CD47, CD200, signal regulatory
protein 1a, IL-4r, E-cadherin, and mannose receptor [136]. Other
alpha integrin-binding partners have also been identied in
expression prolife studies including [alpha]
M
[beta]
2
, [alpha]
X
[-
beta]
2
, [alpha]
5
[beta]
1
more than [alpha]
V
[beta]
1
more than
[alpha]
3
[beta]
1
, and [alpha]
2
[beta]
1
[140]. MNGCs have been shown
to adhere to complement components and brinogen and at later
time points vitronectin [149]. Other molecules/proteins have been
found implicated in MNGC fusion, function and survival including
STAT6, P2X7 receptor, and Connexin 43 [150e153]. IL-4 and IL-13
are two important cytokines for MNGC fusion and formation and
are thought to be produced mainly by T lymphocytes [154].
Furthermore, macrophage fusion to form giant cells has been
shown to also require MMP9 [155]. It has been shown that mac-
rophages/FBGC strongly express HLA-DR, CD98, B7-2 (CD86), and
B7eH1 (PD-L1), but not B7-1 (CD80) or B7eH2 (B7RP-1). Molecules
expressed on osteoclasts including calcitonin receptor, tartrate-
resistant acid phosphatase and RANK or dendritic cells including
CD1a, CD40, CD83, CD95/fas are found undetectable in MNGCs
[156]. In contrast, it has been shown that fusing macrophages/
FBGCs strongly express
a
X integrin (CD11c), CD68, and dendritic
cell-specic intercellular adhesion molecule-3-grabbing non-
integrin (DC-SIGN), whereas CD14 is completely down-regulated
with IL-4-induced macrophage fusion [156].Table 2 summarizes
previous reports demonstrating differences in factors important to
osteoclast and FBGCs formation and function.
4. Monocytes, macrophages, MNGCs and biomaterials
To date, most of our understanding of macrophages/MNGCs
around biomaterials has been driven by key studies performed
outside the bone biology eld [157,158]. Current strategies adapted
from soft tissue integration of biomaterials have since been
demonstrated as effective strategies in bone biology by mainly
focusing on reducing the possibility of a foreign body reaction by
reducing either protein adsorption, initial cell adhesion, inam-
matory cytokine secretion and/or FBGC fusion around biomaterials
[159]. Pioneering research conducted in large part by James
Anderson's group revealed that hydrophilic and anionic substrate
surfaces caused less macrophage adherence and increased their
apoptosis and reduced foreign body giant cell formation [160].
In vitro studies have demonstrated that MNGCs produce a large
array of pro-inammatory cytokines including IL-1
b
, TNF-alpha, IL-
6, IL-8, and macrophage inammatory protein (MIP)-1[beta]
[161,162]. Interestingly, polymer biomaterial surface topography
and chemistry has been shown to affect the release of these cyto-
kines with preference for hydrophilic/neutral and hydrophilic/
anionic surfaces [161e163]. Furthermore, it has been shown that
hydrophilic surfaces markedly reduced the number of adherent
macrophages and MNGCs on the surface when compared to hy-
drophobic surfaces. These studies clearly showed that material
surface/chemistry is a strong regulator of induced MNGCs on
polymer surfaces for soft tissue integration [162,164]. It has been
suggested that unlike osteoclasts, MNGCs adhere to markedly
different synthetic surfaces [147], however while the eld of
MNGCs adherence and expression of integrin protein domains has
been largely studied on polymers, very little information is avail-
able on a wide array of very frequently used bone biomaterials such
Fig. 4. Proposed foreign body giant cell formation: Inammatory and wound healing
responses to implanted medical devices, prostheses and biomaterials (adapted from
Brodbeck and Anderson (2009).
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 9
as dental/orthopedic implants and bone grafting materials.
In addition, it was also shown that topographical effects had an
inuence on TNF-alpha and VEGF secretion; characteristics of the
M2 macrophage [159]. More recently, phenotypic expression of
human monocyte-derived IL-4 induced FBGCs and macrophages
in vitro has revealed the strong dependence on material surface
properties with certain substrates being more prone to form FBGCs
[165]. These key ndings generated over the past 2 decades have
largely been adopted to bone biomaterials without much further
study. Below we summarize the current literature involving
searches for macrophages, monocytes, FBGCs and MNGCs around
bone grafting materials and dental/orthopedic implants.
4.1. Macrophages, MNGCs and dental/orthopedic implants
The role of macrophages around bone implants has been a topic
of study both in vitro and in vivo over several decades. While it
became clear several years ago that cells from the monocytic
lineage were one of the rst to come in contact with titanium
surfaces, their exact role in implant dentistry was largely left
unanswered In fact, a recent in vitro systematic review relating to
the topic of cells and dental implant surfaces revealed that
approximately 90% of all published papers on the topic focused
mainly on osteoblast behavior on a variety of implant surfaces with
either varying surface topography, surface material compositions or
surface hydrophilicity [2]. This nding grossly demonstrates the
lack of study with immune-modulation of implant surface; i.e. the
entire eld of osteoimmunology.
It remains interesting to note that to this day, a small percentage
of implants are lost every year for completely unknown causes
likely dealing with humoral immunity [166]. Recently, a prominent
group in Sweden working in the eld of dental implants has been
largely implicated and interested in the foreign body reaction
around dental implants [167,168]. While their key research topic
relates primarily to how cells derived from the monocytic lineage
are able to fuse and form MNGCs (also termed GFBCs by these
authors), their main research focus is on how these cells are
implicated in longeterm equilibrium to avoid material rejection
citing their role in biomaterial failure [167e169]. However, inter-
esting recent ndings have demonstrated clearly through a variety
of experimental designs that MNGCs express a large variety of
markers associated with M2 macrophages in vivo [11]. These sur-
prising ndings have led to the hypothesis that these giant cells
found around implanted biomaterials may in fact contribute more
so to tissue integration as opposed to material rejection. Interested
by these ndings, our group recently established new parameters
used to quantify these cell types around implant surfaces termed
MNGC-to-implant contact (MIC) and quantied the data in relation
to bone-to-implant contact (BIC) and peri-implant bone density
(BD) (Fig. 5)[170e172 ] . Despite MNGCs being present on all tested
implant surfaces, MNGCs were not associated with an inamma-
tory cell inltrate or with brous encapsulation in osseointegration
of this defect model in miniature pigs. In the paper by Chappuis
et al. (2015), MNGCs were less numerous on the Ti implants (range:
3.9e5.2%) compared to the two types of ceramic implants (range:
17.6e30.3%, p <0.0001) (Fig. 6). However, no correlation could be
found between the newly formed peri-implant bone density,
dened as the percentage of new bone area inside the screw
threads (nBD), and the presence of MNGCs [172]. Therefore, it be-
comes very difcult to assess the role of these cells as almost no
information has characterized these cells in vivo or investigated the
release of factors implicated in the differentiation of cells leading to
hard tissue formation around bone biomaterials. Below we sum-
marize studies that have focused on the roles of cells derived from
the monocytic lineage on bone implant surfaces (Table 3).
Over 10 years ago, Takabe et al. investigated the effect of
commercially pure titanium substrate topography on adherent
macrophage osteogenic and osteoinductive cytokine expression
[173]. The J774A.1 murine macrophage was investigated for cell
adhesion and TGF-
b
1 and BMP-2 gene expression onto polished,
machined, and grit-blasted cpTi surfaces. Macrophage adhesion
increased with time on all surfaces and spreading increased with
increasing surface roughness (polished <machined <grit-blasted).
BMP-2 expression was also further enhanced on roughened sur-
faces [173]. Refai et al. further showed that surface roughness
increased secretion of pro-inammatory markers including IL-1B,
IL-6, and TNF-alpha, and chemokines including monocyte chemo-
attractant protein 1 and macrophage inammatory protein
1alpha with and without culture containing LPS [174]. In a second
study investigating the role of titanium surface topography on
J774A.1 macrophage inammatory cytokines and nitric oxide pro-
duction, it was once again conrmed via real-time PCR that pro-
inammatory mediators were more highly expressed on grit-
blasted/acid etched rough surfaces than on smoother ones [175].
Ghrebi et al. later showed that macrophage shape was signicantly
altered on roughened surfaces activating early ERK1/2 signaling as
well as vinculin and pFAK expression in macrophages [176].
In 2007, Makihira et al. was one of the rst to show that titanium
surface roughness accelerated RANKL-dependent differentiation of
RAW264.7 macrophages into osteoclasts [177]. They found that
surface roughness promoted the expression of TRAP and Cathepsin
K[177]. In 2008, one of the rst studies investigated the cross-talk
between macrophages and osteoblasts on titanium-based particles
[178]. Within their experimental design it was found that exposure
of co-cultured macrophages to sub-cytotoxic doses of titanium
particles did not change the osteoblastic expression of RANKL or
OPG into media, however both IL6 and PGE2 levels increased to a
similar extent after treatment with Ti particles [178]. The results
from that study indicate that interactions of osteoblasts and mac-
rophages respond to micro-particles of titanium and their cross-
talk seems to play an important role in guiding new bone forma-
tion [178]. In 2010, an animal study conrmed that new bone for-
mation in vivo is preceded by macrophage accumulation [179].It
was found that although roughened SLA surfaces displayed higher
levels of mineralization, the analysis of immunohistochemistry
demonstrated that the predominant cell type at 1 week prior to
mineralization was the macrophage, whereas polished surfaces
demonstrated less adhered macrophages found on the surfaces
with little to no signs of mineralization suggesting for one of the
rst times an important role of macrophages on guiding new bone
formation [179]. These results were later conrmed in a clinical
study assessing the early molecular changes of osseointegration of
dental implants in humans via Affymetrix gene analysis [180].It
was shown that an abundant upregulation of several differential
markers of alternative activated macrophages was observed
including MRC1, MSR1, MS4A4A, SLC38A6 and CCL18 [180].
In 2010, Hefti et al. compared osteoclast resorption pits on bone
with titanium and zirconia surfaces [181]. Results from this study
showed that osteoclasts resorb bone in similar roughness and pit
sizes as titanium surfaces which may additional provide some
insight into structural requirements for bone remodeling on
implanted biomaterials [181].
Miller et al. investigated for the rst time the in vitro effects of
hydrophilic roughened titanium surfaces on blood clot formation,
platelet activation and activation of the complement system [182].
They found that untreated Ti surfaces displayed thin blood clots
whereas alkali treatment of roughened surfaces enhanced the
ability for nucleated cells to adhere from whole blood [182]. Hamlet
et al. then showed that surface hydrophilicity down-regulated key
pro-inammatory cytokines including TNF-alpha, IL-1alpha and IL-
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e1910
1B and chemokine CCL2, whereas hydrophobic rough surfaces
tended to increase them [183]. In 2014, Alfari et al. further showed
that hydrophilic SLA surfaces down-regulated the expression of 10
key pro-inammatory genes namely TNF, IL-1a, IL-1B, CCL-1, -3, -19
and 20, CXCL-1 and 8 and IL-1 receptor type 1 when compared to
SLA [184].
In 2012, ceramic modications of porous titanium surfaces were
investigated for their effect on macrophage activation [185]. Tita-
nium surfaces were covered with bioactive hydroxyapatite (HA),
bioglass and calcium silicate. RAW264.7 macrophage adhesion,
morphology and activation were assessed. It was found that cal-
cium silicate decreased the macrophage adherence and upregu-
lated pro-inammatory mediators such as TNF-alpha, IL-6 and IL-
12. HA decreased cell adherence with little change in mediators
[185]. This study proposed for the rst time that bioactive materials
such as HA and BG may improve osseointegration via macrophage
activation [185]. Brinkmann et al. investigated the effects of surface
roughness of titanium implants on osteoclast behavior in vitro
Fig. 5. MNGCs on dental implants made of Ti, TiZr, TAV, ZrO2, and ZrO2/Al2O3. Undecalcied ground sections (reprinted with permission from Ref. [172]).
Fig. 6. Inuence of ER stress pathways in macrophage differentiation (reprinted with
permission from Ref. [132])., ER stress pathways include the following: c-Jun N-ter-
minal kinase (JNK), PPAR
g
, scavenger receptor CD36, and SR-A1. Macrophage M1 re-
ceptors include CCR7 and CD86; M2 receptors include MR and CD163.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 11
[186]. It was found that osteoclasts show similar characteristics on
rough titanium surfaces when compared to bone, whereas reduced
activity was observed on smooth titanium surfaces [186].
As the eld of osteoimmunology was further advanced, Barth
et al. showed that surface roughness promoted an M2 macrophage
phenotype suggesting that these cells may additionally contribute
to enhanced wound healing [187]. Interestingly, Ma et al. demon-
strated in an in vivo study that anodization at 5 and 20 V as well as
UV irradiation used to generate hydrophilic titanium surfaces could
also be utilized as strategies to increase M2 macrophage polariza-
tion [188]. Furthermore, Lu et al. further showed that a reduction in
immune response was observed on nano- and submicron rough
titanium demonstrating for the rst time that nanofeatures on
implant surfaces may also be controlled to reduce pro-
inammatory mediators and induce an M2 macrophage
phenotype onto bone biomaterials [189].
4.2. Macrophages, MNGCs and bone grafting materials
The role of macrophages around bone grafting materials has
been studied much earlier than on titanium implants and in recent
years has gained tremendous momentum with the advancements
made in the eld of osteoimmunology (Table 4). Despite recent
trends studying the role of monocyte-derived cells in vitro, the
difculty in culturing cells on 3-dimensional bone grafts with
varying particle sizes signicantly increases the degree of difculty
to perform such in vitro research. Therefore, large gaps can be
found in the literature with little investigation in the early 2000s
studying the role of macrophages and osteoclasts on these bone
grafting materials. In recent years, the paradigm has shifted from
Table 3
Studies investigating monocytes, macrophages, osteoclasts or MNGCs on dental/orthopedic implant surfaces.
Author Year Main nding
Takebe et al. 2003 Surface roughness increased J774A.1 macrophage adhesion and release of BMP2.
Refai et al. 2004 Surface roughness increases RAW 264.7 macrophage secretion of pro-inammatory cytokines. LPS further increases this nding.
Tan et al. 2006 Surface roughness increased pro-inammatory cytokine production in J774A.1 macrophages
Makihira et al. 2007 Surface roughness increased osteoclast differentiation of RAW 264.7 macrophages cultured with RANKL.
Valles et al. 2008 Titanium particles inuence macrophage cross-talk with osteoblasts and modulate IL6 and PGE2 expression.
Chehroudi et al. 2009 Bone formation on rough surfaces is preceded by macrophage accumulation on rough titanium surfaces in vivo
Hefti et al. 2010 Osteoclasts resorb bone in similar shapes and sizes to titanium implant surfaces
Milleret et al. 2011 Alkaline treatment and hyperhydrophilic surfaces improve nucleated cell adhesion and blood clot thickness
Hamlet 2011 Hydrophilic titanium surfaces reduces pro-inammatory cytokine gene expression in RAW 264.7 macrophages
Scislowska-Czarnecka
et al.
2012 Ceramic modications to porous titanium can modulate gene expression of RAW 264.7 macrophages.
Brinkmann et al. 2012 Surface roughness increases osteoclast differentiation and actin rings on titanium surfaces
Barth et al. 2012 Surface roughness promotes macrophage differentiation towards the M2 phenotype responsible for enhanced wound repair
Ghrebi et al. 2013 Surface topography modulates cell spreading and increases vinculin distribution and ERK1/2 signaling in macrophages.
Alfarsi et al. 2014 Hydrophilic SLA signicantly downregulated 10 pro-inammatory cytokines in macrophages.
Thalji et al. 2014 Clinical study demonstrating that early osseointegration of human dental implants involve an abundance of upregulated activated
macrophage genes
Ma et al. 2014 Anodization and UV irradiation can be used to generate hydrophilic titanium surfaces able to modulate pro M2 polarization of macrophages
both in vitro and in vivo
Lu et al. 2015 Nano-micron rough titanium surfaces may also further reduce a pro-inammatory macrophage phenotype
Table 4
Studies investigating monocytes, macrophages, osteoclasts or MNGCs on bone grafting materials.
Author Year Main nding
Yamada et al. 1996 Osteoclasts capable of resorbing BCP bone grafts
Benahmed
et al.
1996 First in vitro model testing monocyte/macrophage degradation of bone grafts including BCP and HA. It was found that macrophages could degrade
BCP faster than HA.
Benahmed
et al.
1997 LPS increases biomaterial degradation by human monocytes/macrophages in vitro
Silva et al. 2003 Macrophages seeded on BCP release higher levels of calcium when compared to neighboring cells.
Rice et al. 2003 The percentage of TCP within the BCP granules was a governing factor in macrophage cellular response.
Curran et al. 2005 The percentage content of TCP was a more signicant factor than granule size on macrophage pro-inammatory release.
Xia et al. 2006 Established a model to investigate cell-mediated cement degradation in vitro
Fellah et al. 2007 The BCP micro particles <20 micro m initiated an inammatory response which might play an important role in osteogenesis.
Fellah et al. 2010 The smallest microparticles decreased the viability of macrophages and enhanced the secretion of pro-inammatory cytokines (IL-6 and TNF-alpha)
Egli et al. 2011 Thermal treatment of calcium phosphates affect osteoclast activity in vitro
Gamblin et al. 2014 BCP bone grafts with MSCs increased osteoblast gene expression and macrophages at 2 and 4 weeks, osteoclastogenesis at 4 and 8 weeks.
Davison et al. 2014a Liposomal clodronate inhibition of macrophage/osteoclastogenesis inhibits osteoinduction in beta-TCP scaffolds
Davison et al. 2014b Osteoclast activation occurs more favorably on submicron topographies
Davison et al. 2014c On submicrostructured TCPs, osteoclasts survived, fused, differentiated, and extensively resorbed TCP whereas FBGC had no ability to resorb TCP.
Kweon et al. 2014 Inhibition of foreign body giant cell formation on silk broin scaffolds can be achieved with 4-hexylresorcinol through suppression of diacylglycerol
kinase delta gene expression
Chen et al. 2014 B-TCP extracts were able to switch macrophage phenotype towards the M2 extreme and increase the expression of BMP2
Chen et al. 2014b Magnesium is able to improve the osteoimmunomodulatory properties of macrophages which enhances osteogenic differentiation and inhibits
osteoclastogenesis
Chen et al. 2015 Cobalt switched mcrophage phenotype towards the M1 extreme, releasing pro-inammatory cytokines and bone catabolic factors
Davison et al. 2015 Material parameters - namely, surface microstructure, macrostructure, and surface chemistry eare critical in promoting osteoclastogenesis and
triggering ectopic bone formation
Shiwaku et al. 2015 Osteoclast differentiation is partially impaired by increased HA content, but not by the presence of micropores within BCP scaffolds, thus favouring
osteoblast crosstalk
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e1912
the osteopromotive potential of bone substitute materials towards
their immunomodulatory behavior thus emphasizing the impor-
tance of initial immune cell responses to biomaterial interactions
and subsequent effects of factors released by immune cells on
osteoblastic cells. It has previously been reported that macrophages
are the major effector cells in immune reaction to implants andthat
they are indispensable for osteogenesis and that their heteroge-
neity and plasticity render macrophages a primer target for im-
mune system modulation [190].
In one of the rst studies, Yamada et al. studied the effects of
osteoclast resorption on biphasic calcium phosphate (BCP) ceramic
bone grafts and observed after 4 days by SEM that degraded cal-
cium phosphate crystals inside the resorption lacunae appeared to
have been dissolved by acids [191]. Furthermore, Behnamed et al.
investigated the inuence of monocyte and macrophage phagocy-
tosis of calcium phosphate particles and their possible involvement
in their degradation due to their sensitivity to secreted cytokines
[192]. They tested the behavior of human monocytes placed on the
surface of HA and BCP tablets in the presence of vitamin D3 and
INF-gamma. After short-term culture (6 days), morphological
events were observed by histological and SEM analysis character-
izing resorption lacunae demonstrating for the rst time the ability
for cells to degrade bone grafts of various compositions (HA versus
BCP) [192]. In 1997, these same authors found that addition of LPS
into their culture system increased biomaterial degradation [193].
In 2003, Silva et al. investigated the effects of BCP on macro-
phage function in vitro [194]. They described an in vitro phenom-
enon regarding the effect of surface reactivity of BCP granules on
human macrophage locomotion, secretion and allowed further
macrophage adhesion [194]. Furthermore in 2003, Rice et al.
investigated various bone grafts ranging in tricalcium phosphate
(TCP) and HA composition from 20, 50, 80, and 100% TCP [195].
Grafts were also classied into two distinct size ranges, small
2e4 mm in diameter and large 4e6 mm in diameter, and their
potential as bone grafting materials was assessed using biocom-
patibility cell culture systems of primary-derived peripheral human
blood monocytes and human osteoblasts isolated from bone. It was
found that the higher content TCP materials, 80% and 100% TCP, had
a detrimental effect on viable cell adhesion after day 1 whereas low
% of TCP granules decreased a pro-inammatory response [195].
Furthermore in 2005, Curran et al. investigated the inammatory
potential of BCP granules in an osteoblast/macrophage co-culture
system with different HA/TCP ratios [196]. Again it was found
that higher content of beta-TCP materials, (80% and 100% TCP) did
not support viable cell adhesion after 1 day, whereas lower content
TCP materials, (20% and 50% TCP) granules supported viable cell
adhesion throughout the observation period [196].
In 2006, Xia et al. investigated the in vitro biodegradation of 3
brushite calcium phosphate cements by RAW264.7 macrophages
[197]. The results demonstrated that RAW264.7 cells formed
multinucleated TRAP-positive osteoclast-like cells capable of
rufed border formation and lacunar resorption. This rst study of
its kind set up a useful model to investigate the cell-mediated
brushite calcium phosphate cement degradation in vitro by mac-
rophages and osteoclasts [197].
Fellah et al. investigated the inammatory reaction in rat muscle
after implantation of BCP micro particles [198]. Three fractions of
BCP micro particles <20, 40e80, and 80e200
m
m were sieved and
the micro particles were carefully characterized by using X-ray
diffraction (XRD), SEM and laser scattering. A brous tissue
encapsulation of the BCP micro particles was observed for all 3
groups of micro particles. The comparison of the cellular response
indicated that the total number of cells was signicantly higher for
BCP <20
m
m than for 40e80 and 80e200
m
m. The number of
macrophages was relatively higher for the smallest than for the
intermediate and largest fractions, whereas the relative percentage
of giant cells was higher for the intermediate and largest size of
particles [198]. These authors later performed a second study
investigating the effect of micro particles on macrophage viability
and release of pro-inammatory factors [199]. It was found that the
smallest micro particles decreased the viability of macrophages and
enhanced the secretion of pro-inammatory cytokines (IL-6 and
TNF-alpha) by macrophages [199]. Based on these observations, it
may be concluded that particle size seems to greatly inuence both
macrophage activation as well as their fusion to MNGCs.
Egli et al. demonstrated the effect of thermal treatments of
calcium phosphate biomaterials to ne tune the physico-chemical
properties on osteoclast resorption [200]. In that study, investiga-
tion of alpha-TCP, beta-TCP and HA demonstrated that a simple
thermal treatment at temperatures of 450e600
Celcius favored
osteoclast behavior [200]. In 2014, an interesting study sought to
investigate the role of macrophages and osteoclasts in an ectopic
bone model [201]. It was found that bone grafts fabricated from BCP
implanted with human mesenchymal stem cells (hMSCs) demon-
strated more signs of ectopic bone formation by increasing
macrophage recruitment at 2 and 4 weeks, osteoclastogenesis and
osteogenesis at 4 and 8 weeks [201]. Implantation of bone grafts
and hMSCs with an anti-RANKL treatment signicantly impaired
bone formation thus conrming the necessity of monocyte-derived
cells to induce new bone formation in this model [201]. In a similar
study, Davison et al. used liposomal clodronate inhibition of
macrophage/osteoclast progenitors and demonstrated that without
this cell source, osteoinductive beta-TCP bone grafts were no longer
capable of forming ectopic bone formation [3]. Further work by this
group showed that osteoclast activation occurs more favorably on
submicron topographies which thereafter affects ectopic bone
formation. These authors proposed that ectopic bone formation in
intramuscular sites is controlled by monocyte-derived cells [202].
Later in 2014, Davison et al. differentiated monocyte-derived cells
into either osteoclasts or FBGCs and thereafter seeded the cells on
various bone grafting materials [203]. It was found that by changing
the scale of surface architecture of TCP, cellular resorption could be
inuenced [203]. On submicrostructured TCPs, osteoclasts sur-
vived, fused, differentiated, and extensively resorbed the substrate;
however, on microstructured TCP, osteoclast survival, TRAP acti-
vation, and fusion were signicantly decreased. Interestingly, it was
found that FBGC could not resorb either TCP material, suggesting
that osteoclast-specic machinery is necessary to resorb TCP [203].
In 2015, Davison et al. hypothesized that surface structural di-
mensions of 1
m
m may be responsible for triggering osteoinduc-
tion and osteoclast formation irrespective of macrostructure (e.g.,
concavities, interconnected macropores, interparticle space) or
surface chemistry [204]. Their results indicate that of the material
parameters tested - namely, surface microstructure, macrostruc-
ture, and surface chemistry emicrostructural dimensions are
critical in promoting osteoclastogenesis and triggering ectopic bone
formation [204]. Shiwaku et al. showed that osteoclast differenti-
ation is partially impaired by increased HA content, but not by the
presence of micropores within BCP scaffolds, thus favoring osteo-
blast crosstalk [205]. The combination of the above mentioned
studies have provided rational that 1) surface micro and nano-
topography of bone grafting materials are highly responsible for
dictating osteoclast-osteoblast crosstalk affecting osteoinduction;
2) Surface material composition also affects osteoinduction and 3)
FBGCs are not responsible for material resorption as only osteo-
clasts are capable of resorbing bone grafting particles at least
in vitro. Future research investigating the role of macrophages in
the above-mentioned scenarios is ongoing.
Kweon et al. performed an interesting study investigating the
role of FBGC formation through a knock down system [206].
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 13
Inhibition of foreign body giant cell formation could be achieved by
4-hexylresorcinol (4HR) through suppression of diacylglycerol ki-
nase delta gene expression [206]. Thereafter, silk broin scaffolds
were grafted into bone defects with 4HR and displayed a signicant
reduction of granuloma formation and increases in the extent of
new bone formation in a rabbit calvarial defect model [206]. Also in
2014, Chen et al. investigated the osteogenic differentiation of bone
marrow MSCs by beta-TCP stimulating macrophages via BMP2
signaling pathway [190]. In this study, they used beta-TCP as a
model biomaterial to investigate the role of macrophages on
biomaterial-stimulated osteogenesis. The extracts from beta-TCP
were able to direct macrophage phenotype towards the M2
extreme, which was related to activation of calcium-sensing re-
ceptor (CaSR) pathway. It was also found that these macrophages
signicantly upregulated BMP2 following stimulation with beta-
TCP, indicating that macrophages may participate in osteogenesis
[190]. Thereafter, this same group performed 2 studies demon-
strating that the osteoimmunomodulatory properties of magne-
sium scaffolds signicantly enhanced osteogenic differentiation of
BMSCs in vitro, whereas osteoclastogenesis was inhibited [207].In
a very recent article, Chen et al. demonstrated that cobalt incor-
poration into beta-TCP bone grafts switched the macrophage
phenotype towards the M1 extreme, releasing pro-inammatory
cytokines and bone catabolic factors in vitro [208]. Therefore,
these recent studies also point to the ability for incorporation of
various trace elements such as cobalt or magnesium to affect the
osteoimmunological behavior of bone grafts.
5. Macrophages and atherosclerosis
One of the main features in the development of atherosclerosis
is the critical role and involvement of macrophages. It has been
reported that atherosclerotic plaque contains high levels of IFN-
gamma, a T-helper1 cytokine that is a known inducer of the clas-
sically activated M1 macrophage (1e3). Interestingly, tissue mac-
rophages found in arteries are known to induce ectopic bone
formation in and around vascular tissues, an area where bone
should otherwise not form [209]. Two key players known to
regulate calcication in vascular tissues are TNF-alpha [209,210]
and oncostatin M [210]. In combination with these ndings, it has
also been shown that macrophage depletion reduces osteophyte
formation in osteoarthritic models [211e213] and macrophages
have been key players in various other bone loss disorders
[214,215]. The combination of these ndings have strongly sug-
gested that macrophages play critical roles in bone formation even
long before key basic research experiments pointed to their vast
roles using transgenic knockout mouse models.
In atherosclerosis, an accumulation of LY6C
hi
monocytes has
been the characteristic cell type known to affect both human and
experimental animal models [216,217]. Monocytes have been
shown to differentiate into macrophages in the artery intima (the
most internal layer) and ingest modied lipoproteins via scavenger
receptors and secrete inammatory mediators. Over time, these
macrophages give rise to lipid-rich macrophages, which have been
given the name foam cells, and thereby become the key contrib-
utors of lipid core buildup followed by subsequent ectopic bone
formation [218].
Two key ndings recently changed our understanding of tissue
macrophages in atherosclerosis. First, it was originally thought that
foam-cells were continuously formed from the contributions from
blood-bound monocytes requiring continual recruitment. Howev-
er, it was recently shown that these foam-cellsare also able to self-
renew by local proliferation [219]. Secondly, it was originally
thought that all macrophages involved in atherosclerotic plaque
were classical M1 phenotype macrophages. In 2012, Oh et al.
demonstrated that alternatively it was stimulated M2 macrophages
that lead to an increase in foam cell formation inducing scavenger
receptor CD36 and SR-A1 expression [132]. The formation of M2
macrophages was primarily activated by endoplasmic reticulum
(ER) stress (Fig. 5). Interestingly, these authors proposed that sup-
pression of ER stress can shift M2 macrophages towards an M1
phenotype and subsequently suppress foam cell formation, thus a
potential therapeutic option for resolution of atherosclerosis [132].
Thus, it demonstrates the extreme plasticity of these cell types and
also how therapeutic options are greatly altered depending on the
region. In atherosclerotic plaque, it would be of therapeutic benet
to reduce M2 (ectopic bone forming multinucleated giant foam-
cells) into more of an M1 phenotype. In contrast, implanted bone
biomaterials would greatly benet from M2 macrophage activation
and their potential to form ectopic bone formation. The research
outlined by the eld of atherosclerosis suggests that the potential
activation of macrophages via ER stress might be a potential ther-
apeutic option for bone biomaterials. This hypothesis, however,
requires a great deal of further investigation.
6. Future research outlooks
In summary, this review article summarized the currently
available literature on OsteoMacs, their fusion to MNGCs and their
role in bone biomaterial integration. It has been previously shown
that depletion of macrophages from primary calvarial osteoblast
cultures led to a 23-fold decrease in osteogenic differentiation and
mineralization [6,41]. Furthermore, depletion of OsteoMacs from
knockout animals led to markedly reduced endochondral and
intramembranous bone formation [6,41,72,73], and knockout sys-
tems around bone grafting materials completely abolished their
osteoinductive activity [3]. Taken together, these ndings strongly
conrm the essential roles of OsteoMacs in normal bone develop-
ment, bone remodeling and bone formation.
Despite this, there exists a great lack of information surrounding
their important roles around bone biomaterials. To date, most of
the literature has either focused on osteoblast behavior on various
bone biomaterials or on FBGCs around biomaterials implanted in
soft tissues. Therefore, there remains a great deal of missing
knowledge required to further improve bone biomaterials in the
future. Furthermore, additional studies characterizing the differ-
ences between mouse and human monocytes/macrophages is
pivotal in order to implement therapeutic strategies aimed at
manipulating these cells during biomaterial tissue integration.
Another area of research that might be valuable to investigators
is to determine the differences between MNGCs from varying tis-
sues. How different are MNGCs observed around biomaterial im-
plants fabricated from PLA scaffolds from MNGCs observed around
bone grafting materials and titanium implant surfaces? Do they
differ substantially from foam-cellsderived in calcied tissues?
Future investigation spanning several elds of research would be
greatly benecial to further improve our understanding of these
cell types.
Some have dened foreign body reactions as the presence of
macrophages and foreign body giant cells on the surface of the
biomaterial, which is an end-stage event in the tissue response
continuum but remains for the duration of the implant[220].
Surprisingly, very little is known about the turnover of macro-
phages and MNGCs at the implant/biomaterial interface. This
question is even more intriguing around bone biomaterials where
recruitment of cells to bone-biomaterial surfaces is greatly limited
due to bone encapsulation. Data from our laboratory thus far has
indicated that these MNGCs remain on the surfaces of bone grafting
materials even 10 years after implantation in a completely stable
and healthy environment.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e1914
Another aspect that remains largely untouched is the effect of
multiple biomaterials in small dimensional bone areas. For
example, tooth loss in dentistry is commonly replaced by dental
implants fabricated from titanium with the great majority of these
procedures requiring contour augmentation procedures utilizing
bone grafts [221]. In such cases, the facial wall thickness of bone in
normal tissues has been estimated less than 1 mm in the majority
of patients [222]. During routine augmentation procedures
following tooth extraction, a titanium implant is placed and
routinely augmented with a sandwich approach combining 2 bone
grafts including an autograft followed by a second bone grafting
material. Thereafter, a barrier membrane commonly fabricated
from porcine derived collagen membrane is utilized to prevent soft
tissue inltration into the slowly growing bony tissue [221].
Therefore, within this 1 mm region, typical cells are in contact with
3 biomaterials including 1) a titanium dental implant, 2) a bone
grafting material from both autogenous source as well as either an
allograft or xenograft, and 3) a collagen barrier membrane [221].It
is unknown at present what the inuence of having so many
different biomaterials might have on such a conned limited space.
It may be that certain of these biomaterials favor macrophage ho-
meostasis and tissue integration whereas other biomaterials might
cause an inammatory reaction. If this is the case, it is completely
unknown how macrophages situated in such a conned space
interact with one another in response to the various signaling
molecules secreted in response to the various biomaterials. This
area of research is one that is specically highly relevant to the
dental eld where a combination of biomaterials, barrier mem-
branes, bone grafts, growth factors and titanium implants are
routinely utilized.
Furthermore, although the effects of macrophage polarization
has thus far been studied individually on various biomaterials such
as metals, bioceramics and biopolymers, rarely have these mate-
rials been compared directly in well-conducted studies. Instead
most studies to date have focused on individual classes of bio-
materials making it difcult to assess which class of biomaterial is
better suited as an osteo-compatible material versus another.
Therefore, the effects of various classes of biomaterials on Osteo-
Mac polarization in relation to their different physical and chemical
properties is necessary to further pursue osteo-immunological
materials capable of minimizing a foreign body reaction and ma-
terial rejection and at the same time enhancing bone regeneration.
While a recent article has compared MNGC formation on zirconia,
alumina-toughened zirconia and titanium implant surfaces
(Chappuis et al., 2015), very few studies have compared this highly
relevant topic in bone grafting material research where a much
greater number of biomaterials exists requiring much further
investigation.
The effect of various cell types such as MNGCs in bone ho-
meostasis in response to biomaterials is another aspect that is
highly relevant and should therefore be studied in the future.
Perhaps the best evidence that MNGCs contribute to bone ho-
meostasis came from a recent study conducted by Katsuyama et al.,
in 2015 [223]. They show specically that MNGCs do not resorb
bone but rather express M2 macrophage-like wound healing and
inammation-terminating molecules [223]. They report critical
ndings that strongly suggest that implant failure due to bone loss
likely results from the activity of osteoclasts and not MNGCs and
that MNGCs are unable to resorb bone but rather express wound
healing and inammation terminating molecules such as Ym1 and
Alox15 [223,224]. Nevertheless, it must also be taken into consid-
eration that other studies demonstrated that revisited joint re-
placements extracted following failure consistently found FBGCs
predominantly expressing inammatory M1 factors
[146,225e227]. Therefore, it remains a valuable therapeutic option
to better understand the patho-physiological regulation of these
cell types for therapeutic benet.
It also remains interesting to note that strategies are now
implemented in atherosclerosis research to reduce M2 macrophage
MNGCs (described as foam-cells) in order to reduce ectopic bone
formation. In light of these ndings, it remains of great interest to
collaborate with researchers in these elds to better understand
how pathology in certain elds is of potential benet in others. In
conclusion, it is clear that our incomplete understanding of tissue
macrophages and fusion to MNGCs has thus far hindered thera-
peutic advances and possibilities of new technologies but should
with time improve clinical settings. It thus becomes vital that more
research be performed on this relatively understudied cell-type
called OsteoMac to further improve the development of osteo-
compatible and osteo-promotive bone biomaterials.
References
[1] F.O. Martinez, S. Gordon, F1000prime reports, The M1 and M2 Paradigm of
Macrophage Activation: Time for Reassessment, vol. 6, 2014, p. 13.
[2] G. Thalji, L.F. Cooper, Molecular assessment of osseointegration in vitro: a
review of current literature, Int. J. Oral Maxillofac. Implants 29 (2014)
e171e99.
[3] N.L. Davison, A.L. Gamblin, P. Layrolle, H. Yuan, J.D. de Bruijn, F. Barrere-de
Groot, Liposomal clodronate inhibition of osteoclastogenesis and osteoin-
duction by submicrostructured beta-tricalcium phosphate, Biomaterials 35
(2014) 5088e5097.
[4] J.R. Arron, Y. Choi, Bone versus immune system, Nature 408 (2000) 535e536.
[5] D.A. Hume, J.F. Loutit, S. Gordon, The mononuclear phagocyte system of the
mouse dened by immunohistochemical localization of antigen F4/80:
macrophages of bone and associated connective tissue, J. Cell Sci. 66 (1984)
189e194.
[6] M.K. Chang, L.J. Raggatt, K.A. Alexander, J.S. Kuliwaba, N.L. Fazzalari,
K. Schroder, et al., Osteal tissue macrophages are intercalated throughout
human and mouse bone lining tissues and regulate osteoblast function
in vitro and in vivo, J. Immunol. Baltim. Md 1950) 181 (2008) 1232e1244.
[7] W.T. Bourque, M. Gross, B.K. Hall, Expression of four growth factors during
fracture repair, Int. J. Dev. Biol. 37 (1993) 573e579.
[8] T.A. Einhorn, The cell and molecular biology of fracture healing, Clin. Orthop.
Relat. Res. (1998) S7eS21.
[9] L.C. Gerstenfeld, D.M. Cullinane, G.L. Barnes, D.T. Graves, T.A. Einhorn, Frac-
ture healing as a post-natal developmental process: molecular, spatial, and
temporal aspects of its regulation, J. Cell. Biochem. 88 (2003) 873e884.
[10] J. Street, M. Bao, L. deGuzman, S. Bunting, F.V. Peale Jr., N. Ferrara, et al.,
Vascular endothelial growth factor stimulates bone repair by promoting
angiogenesis and bone turnover, Proc. Natl. Acad. Sci. U. S. A. 99 (2002)
9656e9661.
[11] D.P. Vasconcelos, M. Costa, I.F. Amaral, M.A. Barbosa, A.P. Aguas, J.N. Barbosa,
Modulation of the inammatory response to chitosan through M2 macro-
phage polarization using pro-resolution mediators, Biomaterials 37 (2015)
116e123.
[12] S.S. Jensen, D.D. Bosshardt, R. Gruber, D. Buser, Long-term stability of contour
augmentation in the esthetic zone: histologic and histomorphometric eval-
uation of 12 human biopsies 14 to 80 months after augmentation,
J. Periodontol. 85 (2014) 1549e1556.
[13] D.A. Chistiakov, Y.V. Bobryshev, N.G. Nikiforov, N.V. Elizova, I.A. Sobenin,
A.N. Orekhov, Macrophage phenotypic plasticity in atherosclerosis: the
associated features and the peculiarities of the expression of inammatory
genes, Int. J. Cardiol. 184 (2015) 436e445.
[14] D.A. Chistiakov, Y.V. Bobryshev, A.N. Orekhov, Changes in transcriptome of
macrophages in atherosclerosis, J. Cell. Mol. Med. 19 (2015) 1163e1173.
[15] C.D. Mills, L.L. Lenz, K. Ley, Macrophages at the fork in the road to health or
disease, Front. Immunol. 6 (2015) 59.
[16] C. Roma-Lavisse, M. Tagzirt, C. Zawadzki, R. Lorenzi, A. Vincentelli, S. Haulon,
et al., M1 and M2 macrophage proteolytic and angiogenic prole analysis in
atherosclerotic patients reveals a distinctive prole in type 2 diabetes, Dia-
bet. Vasc. Dis. Res. 12 (2015) 279e289.
[17] V.J. Swier, L. Tang, M.M. Radwan, W.J. Hunter 3rd, D.K. Agrawal, The role of
high cholesterol-high fructose diet on coronary arteriosclerosis, Histol. His-
topathol. (2015) 11652.
[18] M.J. Favus, Primer on the metabolic bone disease and disorders of mineral
metabolism, Revue Fr. d Endocrinol. Clin. Nutr. Metabol. 37 (1996) 553e554.
[19] S. Gordon, P.R. Taylor, Monocyte and macrophage heterogeneity, Nat. Rev.
Immunol. 5 (2005) 953e964.
[20] D.A. Hume, The mononuclear phagocyte system, Curr. Opin. Immunol. 18
(2006) 49e53.
[21] P.R. Taylor, L. Martinez-Pomares, M. Stacey, H.H. Lin, G.D. Brown, S. Gordon,
Macrophage receptors and immune recognition, Annu. Rev. Immunol. 23
(2005) 901e944.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 15
[22] D. Hashimoto, A. Chow, C. Noizat, P. Teo, M.B. Beasley, M. Leboeuf, et al.,
Tissue-resident macrophages self-maintain locally throughout adult life with
minimal contribution from circulating monocytes, Immunity 38 (2013)
792e804.
[23] I.G. Winkler, N.A. Sims, A.R. Pettit, V. Barbier, B. Nowlan, F. Helwani, et al.,
Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches
and their depletion mobilizes HSCs, Blood 116 (2010) 4815e4828.
[24] L.C. Davies, S.J. Jenkins, J.E. Allen, P.R. Taylor, Tissue-resident macrophages,
Nat. Immunol. 14 (2013) 986e995.
[25] L.C. Davies, M. Rosas, S.J. Jenkins, C.T. Liao, M.J. Scurr, F. Brombacher, et al.,
Distinct bone marrow-derived and tissue-resident macrophage lineages
proliferate at key stages during inammation, Nat. Commun. 4 (2013) 1886.
[26] D.E. Heinemann, C. Lohmann, H. Siggelkow, F. Alves, I. Engel, G. Koster,
Human osteoblast-like cells phagocytose metal particles and express the
macrophage marker CD68 in vitro, J. Bone Jt. Surg. Br. Vol. 82 (2000)
283e289.
[27] C. Ruiz, E. Perez, M. Vallecillo-Capilla, C. Reyes-Botella, Phagocytosis and
allogeneic T cell stimulation by cultured human osteoblast-like cells. Cellular
physiology and biochemistry, Int. J. Exp. Cell. Physiol., Biochem. Pharmacol.
13 (2003) 309e314.
[28] T. Kikuchi, T. Matsuguchi, N. Tsuboi, A. Mitani, S. Tanaka, M. Matsuoka, et al.,
Gene expression of osteoclast differentiation factor is induced by lipopoly-
saccharide in mouse osteoblasts via Toll-like receptors, J. Immunol. Baltim.
Md 1950) 166 (2001) 3574e3579.
[29] K. Maruyama, G. Sano, K. Matsuo, Murine osteoblasts respond to LPS and
IFN-gamma similarly to macrophages, J. Bone Miner. Metab. 24 (2006)
454e460.
[30] C. Reyes-Botella, M.J. Montes, M.F. Vallecillo-Capilla, E.G. Olivares, C. Ruiz,
Expression of molecules involved in antigen presentation and T cell activa-
tion (HLA-DR, CD80, CD86, CD44 and CD54) by cultured human osteoblasts,
J. Periodontol. 71 (2000) 614e617.
[31] D.E. Heinemann, H. Siggelkow, L.M. Ponce, V. Viereck, K.G. Wiese, J.H. Peters,
Alkaline phosphatase expression during monocyte differentiation. Over-
lapping markers as a link between monocytic cells, dendritic cells, osteo-
clasts and osteoblasts, Immunobiology 202 (2000) 68e81.
[32] M. Kuwana, Y. Okazaki, H. Kodama, K. Izumi, H. Yasuoka, Y. Ogawa, et al.,
Human circulating CD14þmonocytes as a source of progenitors that exhibit
mesenchymal cell differentiation, J. Leukoc. Biol. 74 (2003) 833e845.
[33] E.A. Olmsted-Davis, Z. Gugala, F. Camargo, F.H. Gannon, K. Jackson,
K.A. Kienstra, et al., Primitive adult hematopoietic stem cells can function as
osteoblast precursors, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 15877e15882.
[34] J.M. Lean, K. Matsuo, S.W. Fox, K. Fuller, F.M. Gibson, G. Draycott, et al.,
Osteoclast lineage commitment of bone marrow precursors through
expression of membrane-bound TRANCE, Bone 27 (2000) 29e40.
[35] W.J. Boyle, W.S. Simonet, D.L. Lacey, Osteoclast differentiation and activation,
Nature 423 (2003) 337e342.
[36] S.S. Jensen, A. Yeo, M. Dard, E. Hunziker, R. Schenk, D. Buser, Evaluation of a
novel biphasic calcium phosphate in standardized bone defects. A histologic
and histomorphometric study in the mandibles of minipigs, Clin. Oral Im-
plants Res. 18 (2007) 752e760.
[37] J.M. Austyn, S. Gordon, F4/80, a monoclonal antibody directed specically
against the mouse macrophage, Eur. J. Immunol. 11 (1981) 805e815.
[38] S. Takeshita, K. Kaji, A. Kudo, Identication and characterization of the new
osteoclast progenitor with macrophage phenotypes being able to differen-
tiate into mature osteoclasts, J. Bone Miner. Res.: Off. J. Am. Soc. Bone Miner.
Res. 15 (2000) 1477e1488.
[39] D.R. Haynes, T.N. Crotti, M. Loric, G.I. Bain, G.J. Atkins, D.M. Findlay, Osteo-
protegerin and receptor activator of nuclear factor kappaB ligand (RANKL)
regulate osteoclast formation by cells in the human rheumatoid arthritic
joint, Rheumatol. Oxf. Engl. 40 (2001) 623e630.
[40] J.M. Hodge, M.A. Kirkland, C.J. Aitken, C.M. Waugh, D.E. Myers, C.M. Lopez, et
al., Osteoclastic potential of human CFU-GM: biphasic effect of GM-CSF,
J. Bone Miner. Res.: ofcial J. Am. Soc. Bone Miner.Res. 19 (2004) 190e199.
[41] A.R. Pettit, M.K. Chang, D.A. Hume, L.J. Raggatt, Osteal macrophages: a new
twist on coupling during bone dynamics, Bone 43 (2008) 976e982.
[42] K.J. Stacey, M.J. Sweet, D.A. Hume, Macrophages ingest and are activated by
bacterial DNA, J. Immunol. Baltim. Md 1950) 157 (1996) 2116e2122.
[43] K. Kobayashi, N. Takahashi, E. Jimi, N. Udagawa, M. Takami, S. Kotake, et al.,
Tumor necrosis factor alpha stimulates osteoclast differentiation by a
mechanism independent of the ODF/RANKL-RANK interaction, J. Exp. Med.
191 (2000) 275e286.
[44] K. Itoh, N. Udagawa, K. Kobayashi, K. Suda, X. Li, M. Takami, et al., Lipo-
polysaccharide promotes the survival of osteoclasts via Toll-like receptor 4,
but cytokine production of osteoclasts in response to lipopolysaccharide is
different from that of macrophages, J. Immunol. Baltim. Md 1950) 170 (2003)
3688e3695.
[45] E. Jimi, I. Nakamura, L.T. Duong, T. Ikebe, N. Takahashi, G.A. Rodan, et al.,
Interleukin 1 induces multinucleation and bone-resorbing activity of oste-
oclasts in the absence of osteoblasts/stromal cells, Exp. Cell Res. 247 (1999)
84e93.
[46] J. van Holten, T.J. Smeets, P. Blankert, P.P. Tak, Expression of interferon beta
in synovial tissue from patients with rheumatoid arthritis: comparison with
patients with osteoarthritis and reactive arthritis, Ann. Rheum. Dis. 64
(2005) 1780e1782.
[47] H. Takayanagi, S. Kim, K. Matsuo, H. Suzuki, T. Suzuki, K. Sato, et al., RANKL
maintains bone homeostasis through c-Fos-dependent induction of inter-
feron-beta, Nature 416 (2002) 744e749.
[48] L.J. Raggatt, M.E. Wullschleger, K.A. Alexander, A.C. Wu, S.M. Millard, S. Kaur,
et al., Fracture healing via periosteal callus formation requires macrophages
for both initiation and progression of early endochondral ossication, Am. J.
Pathol. 184 (2014) 3192e3204.
[49] G.A. Rodan, T.J. Martin, Role of osteoblasts in hormonal control of bone
resorptionea hypothesis, Calcif. Tissue Int. 33 (1981) 349e351.
[50] T. Suda, N. Takahashi, T.J. Martin, Modulation of osteoclast differentiation,
Endocr. Rev. 13 (1992) 66e80.
[51] T.J. Martin, N.A. Sims, Osteoclast-derived activity in the coupling of bone
formation to resorption, Trends Mol. Med. 11 (2005) 76e81.
[52] C.G. James, C.T. Appleton, V. Ulici, T.M. Underhill, F. Beier, Microarray ana-
lyses of gene expression during chondrocyte differentiation identies novel
regulators of hypertrophy, Mol. Biol. Cell 16 (2005) 5316e5333.
[53] R.K. Assoian, B.E. Fleurdelys, H.C. Stevenson, P.J. Miller, D.K. Madtes,
E.W. Raines, et al., Expression and secretion of type beta transforming
growth factor by activated human macrophages, Proc. Natl. Acad. Sci. U. S. A.
84 (1987) 6020e6024.
[54] F. Takahashi, K. Takahashi, K. Shimizu, R. Cui, N. Tada, H. Takahashi, et al.,
Osteopontin is strongly expressed by alveolar macrophages in the lungs of
acute respiratory distress syndrome, Lung 182 (2004) 173e185.
[55] M. Kreutz, R. Andreesen, S.W. Krause, A. Szabo, E. Ritz, H. Reichel, 1,25-
dihydroxyvitamin D3 production and vitamin D3 receptor expression are
developmentally regulated during differentiation of human monocytes into
macrophages, Blood 82 (1993) 1300e1307.
[56] C.M. Champagne, J. Takebe, S. Offenbacher, L.F. Cooper, Macrophage cell lines
produce osteoinductive signals that include bone morphogenetic protein-2,
Bone 30 (2002) 26e31.
[57] R. Hattner, B.N. Epker, H.M. Frost, Suggested sequential mode of control of
changes in cell behaviour in adult bone remodelling, Nature 206 (1965)
489e490.
[58] A.M. Partt, The bone remodeling compartment: a circulatory function for
bone lining cells, J. Bone Miner. Res. 16 (2001) 1583e1585.
[59] S.H. Burnett, E.J. Kershen, J. Zhang, L. Zeng, S.C. Straley, A.M. Kaplan, et al.,
Conditional macrophage ablation in transgenic mice expressing a Fas-based
suicide gene, J. Leukoc. Biol. 75 (2004) 612e623.
[60] L.F. Bonewald, The amazing osteocyte, J. Bone Miner. Res.: Off. J. Am. Soc.
Bone Miner. Res. 26 (2011) 229e238.
[61] G.R. Mundy, F. Elefteriou, Boning up on ephrin signaling, Cell 126 (2006)
441e443.
[62] M. Sandberg, T. Vuorio, H. Hirvonen, K. Alitalo, E. Vuorio, Enhanced
expression of TGF-beta and c-fos mRNAs in the growth plates of developing
human long bones, Dev. Camb. Engl. 102 (1988) 461e470.
[63] C. Zhao, N. Irie, Y. Takada, K. Shimoda, T. Miyamoto, T. Nishiwaki, et al.,
Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis, Cell
Metab. 4 (2006) 111e121.
[64] G. Yu, H. Luo, Y. Wu, J. Wu, Mouse ephrinB3 augments T-cell signaling and
responses to T-cell receptor ligation, J. Biol. Chem. 278 (2003) 47209e47216.
[65] G. Yu, J. Mao, Y. Wu, H. Luo, J. Wu, Ephrin-B1 is critical in T-cell development,
J. Biol. Chem. 281 (2006) 10222e10229.
[66] K. Matsuo, N. Irie, Osteoclast-osteoblast communication, Arch. Biochem.
Biophys. 473 (2008) 201e209.
[67] L.J. Raggatt, N.C. Partridge, Cellular and molecular mechanisms of bone
remodeling, J. Biol. Chem. 285 (2010) 25103e25108.
[68] A.M. Partt, The bone remodeling compartment: a circulatory function for
bone lining cells, J. Bone Miner. Res.: Off. J. Am. Soc. Bone Miner. Res. 16
(2001) 1583e1585.
[69] P.M. Henson, D.A. Hume, Apoptotic cell removal in development and tissue
homeostasis, Trends Immunol. 27 (2006) 244e250.
[70] R.L. Jilka, R.S. Weinstein, A.M. Partt, S.C. Manolagas, Quantifying osteoblast
and osteocyte apoptosis: challenges and rewards, J. bone Miner. Res. Off. J.
Am. Soc. Bone Miner. Res. 22 (2007) 1492e1501.
[71] K.A. Alexander, M.K. Chang, E.R. Maylin, T. Kohler, R. Muller, A.C. Wu, et al.,
Osteal macrophages promote in vivo intramembranous bone healing in a
mouse tibial injury model, J. Bone Miner. Res.: Off. J. Am. Soc. Bone Miner.
Res. 26 (2011) 1517e1532.
[72] L. Vi, G.S. Baht, H. Whetstone, A. Ng, Q. Wei, R. Poon, et al., Macrophages
promote osteoblastic differentiation in-vivo: implications in fracture repair
and bone homeostasis, J. Bone Miner. Res.: Off. J. Am. Soc. Bone Miner. Res.
30 (2015) 1090e1102.
[73] S.W. Cho, F.N. Soki, A.J. Koh, M.R. Eber, P. Entezami, S.I. Park, et al., Osteal
macrophages support physiologic skeletal remodeling and anabolic actions
of parathyroid hormone in bone, Proc. Natl. Acad. Sci. U. S. A. 111 (2014)
1545e1550.
[74] K.S. Selander, J. Monkkonen, E.K. Karhukorpi, P. Harkonen, R. Hannuniemi,
H.K. Vaananen, Characteristics of clodronate-induced apoptosis in osteo-
clasts and macrophages, Mol. Pharmacol. 50 (1996) 1127e1138.
[75] W.S. Simonet, D.L. Lacey, C.R. Dunstan, M. Kelley, M.S. Chang, R. Luthy, et al.,
Osteoprotegerin: a novel secreted protein involved in the regulation of bone
density, Cell 89 (1997) 309e319.
[76] F. Ginhoux, S. Jung, Monocytes and macrophages: developmental pathways
and tissue homeostasis, Nat. Rev. Immunol. 14 (2014) 392e404.
[77] P.J. Murray, T.A. Wynn, Obstacles and opportunities for understanding
macrophage polarization, J. Leukoc. Biol. 89 (2011) 557e563.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e1916
[78] M.L. Novak, T.J. Koh, Macrophage phenotypes during tissue repair, J. Leukoc.
Biol. 93 (2013) 875e881.
[79] T.A. Wynn, A. Chawla, J.W. Pollard, Macrophage biology in development,
homeostasis and disease, Nature 496 (2013) 445e455.
[80] R. van Furth, Z.A. Cohn, The origin and kinetics of mononuclear phagocytes,
J. Exp. Med. 128 (1968) 415e435.
[81] M.G. Cecchini, M.G. Dominguez, S. Mocci, A. Wetterwald, R. Felix, H. Fleisch,
et al., Role of colony stimulating factor-1 in the establishment and regulation
of tissue macrophages during postnatal development of the mouse, Dev.
Camb. Engl. 120 (1994) 1357e1372.
[82] X.M. Dai, G.R. Ryan, A.J. Hapel, M.G. Dominguez, R.G. Russell, S. Kapp, et al.,
Targeted disruption of the mouse colony-stimulating factor 1 receptor gene
results in osteopetrosis, mononuclear phagocyte deciency, increased
primitive progenitor cell frequencies, and reproductive defects, Blood 99
(2002) 111e120.
[83] W. Wiktor-Jedrzejczak, S. Gordon, Cytokine regulation of the macrophage (M
phi) system studied using the colony stimulating factor-1-decient op/op
mouse, Physiol. Rev. 76 (1996) 927e947.
[84] B. Passlick, D. Flieger, H.W. Ziegler-Heitbrock, Identication and character-
ization of a novel monocyte subpopulation in human peripheral blood, Blood
74 (1989) 2527e2534.
[85] C. Auffray, D. Fogg, M. Garfa, G. Elain, O. Join-Lambert, S. Kayal, et al.,
Monitoring of blood vessels and tissues by a population of monocytes with
patrolling behavior, Science 317 (2007) 666e670.
[86] L.M. Carlin, E.G. Stamatiades, C. Auffray, R.N. Hanna, L. Glover, G. Vizcay-
Barrena, et al., Nr4a1-dependent Ly6C(low) monocytes monitor endothelial
cells and orchestrate their disposal, Cell 153 (2013) 362e375.
[87] L. Ziegler-Heitbrock, T.P. Hofer, Toward a rened denition of monocyte
subsets, Front. Immunol. 4 (2013) 23.
[88] K. Liu, G.D. Victora, T.A. Schwickert, P. Guermonprez, M.M. Meredith, K. Yao,
et al., In vivo analysis of dendritic cell development and homeostasis, Science
324 (2009) 392e397.
[89] F. Ginhoux, K. Liu, J. Helft, M. Bogunovic, M. Greter, D. Hashimoto, et al., The
origin and development of nonlymphoid tissue CD103þDCs, J. Exp. Med.
206 (2009) 3115e3130.
[90] S.J. Jenkins, D. Ruckerl, P.C. Cook, L.H. Jones, F.D. Finkelman, N. van Rooijen, et
al., Local macrophage proliferation, rather than recruitment from the blood,
is a signature of TH2 inammation, Science 332 (2011) 1284e1288.
[91] S.J. Jenkins, D. Ruckerl, G.D. Thomas, J.P. Hewitson, S. Duncan, F. Brombacher,
et al., IL-4 directly signals tissue-resident macrophages to proliferate beyond
homeostatic levels controlled by CSF-1, J. Exp. Med. 210 (2013) 2477e2491.
[92] C. Bonifer, D.A. Hume, The transcriptional regulation of the Colony-
Stimulating Factor 1 Receptor (csf1r) gene during hematopoiesis, Front.
Biosci.: J. Virtual Libr. 13 (2008) 549e560.
[93] W.J. Boyle, W.S. Simonet, D.L. Lacey, Osteoclast differentiation and activation,
Nature 423 (2003) 337e342.
[94] I. Theurl, G. Fritsche, S. Ludwiczek, K. Garimorth, R. Bellmann-Weiler,
G. Weiss, The macrophage: a cellular factory at the interphase between iron
and immunity for the control of infections, Biometals: Int. J. Role Metal Ions
Biol. Biochem. Med. 18 (2005) 359e367.
[95] D.A. Hume, I.L. Ross, S.R. Himes, R.T. Sasmono, C.A. Wells, T. Ravasi, The
mononuclear phagocyte system revisited, J. Leukoc. Biol. 72 (2002) 621e627.
[96] G.B. Lipford, T. Sparwasser, M. Bauer, S. Zimmermann, E.S. Koch, K. Heeg, et
al., Immunostimulatory DNA: sequence-dependent production of potentially
harmful or useful cytokines, Eur. J. Immunol. 27 (1997) 3420e3426.
[97] T. Sparwasser, T. Miethke, G. Lipford, A. Erdmann, H. Hacker, K. Heeg, et al.,
Macrophages sense pathogens via DNA motifs: induction of tumor necrosis
factor-alpha-mediated shock, Eur. J. Immunol. 27 (1997) 1671e1679.
[98] A.P. Moreira, C.M. Hogaboam, Macrophages in allergic asthma: ne-tuning
their pro- and anti-inammatory actions for disease resolution,
J. Interferon Cytokine Res.: Off. J. Int. Soc. Interferon Cytokine Res. 31 (2011)
485e491.
[99] D.M. Mosser, J.P. Edwards, Exploring the full spectrum of macrophage acti-
vation, Nat. Rev. Immunol. 8 (2008) 958e969.
[100] F. Heymann, C. Trautwein, F. Tacke, Monocytes and macrophages as cellular
targets in liver brosis, Inamm. Allergy Drug Targ. 8 (2009) 307e318.
[101] S.D. Ricardo, H. van Goor, A.A. Eddy, Macrophage diversity in renal injury and
repair, J. Clin. Invest. 118 (2008) 3522e3530.
[102] A. Mantovani, A. Sica, S. Sozzani, P. Allavena, A. Vecchi, M. Locati, The che-
mokine system in diverse forms of macrophage activation and polarization,
Trends Immunol. 25 (2004) 677e686.
[103] S. Gordon, Alternative activation of macrophages, Nat. Rev. Immunol. 3
(2003) 23e35.
[104] C.A. Louis, V. Mody, W.L. Henry Jr., J.S. Reichner, J.E. Albina, Regulation of
arginase isoforms I and II by IL-4 in cultured murine peritoneal macro-
phages, Am. J. Physiol. 276 (1999) R237eR242.
[105] E. Song, N. Ouyang, M. Horbelt, B. Antus, M. Wang, M.S. Exton, Inuence of
alternatively and classically activated macrophages on brogenic activities of
human broblasts, Cell. Immunol. 204 (2000) 19e28.
[106] D.M. Mosser, The many faces of macrophage activation, J. Leukoc. Biol. 73
(2003) 209e212.
[107] C.F. Anderson, J.S. Gerber, D.M. Mosser, Modulating macrophage function
with IgG immune complexes, J. Endotoxin Res. 8 (2002) 477e481.
[108] R.D. Stout, C. Jiang, B. Matta, I. Tietzel, S.K. Watkins, J. Suttles, Macrophages
sequentially change their functional phenotype in response to changes in
microenvironmental inuences, J. Immunol. (Baltim. Md: 1950) 175 (2005)
342e349.
[109] C. Buechler, M. Ritter, E. Orso, T. Langmann, J. Klucken, G. Schmitz, Regula-
tion of scavenger receptor CD163 expression in human monocytes and
macrophages by pro- and antiinammatory stimuli, J. Leukoc. Biol. 67 (2000)
97e103.
[110] C.A. Ambarus, S. Krausz, M. van Eijk, J. Hamann, T.R. Radstake, K.A. Reedquist,
et al., Systematic validation of specic phenotypic markers for in vitro
polarized human macrophages, J. Immunol. Methods 375 (2012) 196e206.
[111] M. Modolell, I.M. Corraliza, F. Link, G. Soler, K. Eichmann, Reciprocal regu-
lation of the nitric oxide synthase/arginase balance in mouse bone marrow-
derived macrophages by TH1 and TH2 cytokines, Eur. J. Immunol. 25 (1995)
1101e1104.
[112] K.J. Mylonas, M.G. Nair, L. Prieto-Lafuente, D. Paape, J.E. Allen, Alternatively
activated macrophages elicited by helminth infection can be reprogrammed
to enable microbial killing, J. Immunol. (Baltim. Md: 1950) 182 (2009)
3084e3094.
[113] R. Rutschman, R. Lang, M. Hesse, J.N. Ihle, T.A. Wynn, P.J. Murray, Cutting
edge: Stat6-dependent substrate depletion regulates nitric oxide production,
J. Immunol. (Baltim. Md: 1950) 166 (2001) 2173e2177.
[114] M. Schneemann, G. Schoeden, Macrophage biology and immunology: man is
not a mouse, J. Leukoc. Biol. 81 (2007) 579 discussion 80.
[115] M. Schneemann, G. Schoedon, Species differences in macrophage NO pro-
duction are important, Nat. Immunol. 3 (2002) 102.
[116] J.D. Shearer, J.R. Richards, C.D. Mills, M.D. Caldwell, Differential regulation of
macrophage arginine metabolism: a proposed role in wound healing, Am. J.
Physiol. 272 (1997) E181eE190.
[117] M. Munder, F. Mollinedo, J. Calafat, J. Canchado, C. Gil-Lamaignere,
J.M. Fuentes, et al., Arginase I is constitutively expressed in human gran-
ulocytes and participates in fungicidal activity, Blood 105 (2005)
2549e2556.
[118] G. Raes, L. Brys, B.K. Dahal, J. Brandt, J. Grooten, F. Brombacher, et al.,
Macrophage galactose-type C-type lectins as novel markers for alternatively
activated macrophages elicited by parasitic infections and allergic airway
inammation, J. Leukoc. Biol. 77 (2005) 321e327.
[119] M. Wehling-Henricks, M.C. Jordan, T. Gotoh, W.W. Grody, K.P. Roos,
J.G. Tidball, Arginine metabolism by macrophages promotes cardiac and
muscle brosis in mdx muscular dystrophy, PLoS One 5 (2010) e10763.
[120] M. Hesse, M. Modolell, A.C. La Flamme, M. Schito, J.M. Fuentes, A.W. Cheever,
et al., Differential regulation of nitric oxide synthase-2 and arginase-1 by
type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the
pattern of L-arginine metabolism, J. Immunol. Baltim. Md 1950) 167 (2001)
6533e6544.
[121] A. Jansen, S. Lewis, V. Cattell, H.T. Cook, Arginase is a major pathway of L-
arginine metabolism in nephritic glomeruli, Kidney Int. 42 (1992)
1107e1112.
[122] E.B. Jude, A.J. Boulton, M.W. Ferguson, I. Appleton, The role of nitric oxide
synthase isoforms and arginase in the pathogenesis of diabetic foot ulcers:
possible modulatory effects by transforming growth factor beta 1, Dia-
betologia 42 (1999) 748e757.
[123] T.A. Wynn, L. Barron, Macrophages: master regulators of inammation and
brosis, Seminars Liver Dis. 30 (2010) 245e257.
[124] S.A. Villalta, H.X. Nguyen, B. Deng, T. Gotoh, J.G. Tidball, Shifts in macrophage
phenotypes and macrophage competition for arginine metabolism affect the
severity of muscle pathology in muscular dystrophy, Hum. Mol. Genet. 18
(2009) 482e496.
[125] P.J. Murray, J.E. Allen, S.K. Biswas, E.A. Fisher, D.W. Gilroy, S. Goerdt, et al.,
Macrophage activation and polarization: nomenclature and experimental
guidelines, Immunity 41 (2014) 14e20.
[126] F.M. Menzies, F.L. Henriquez, J. Alexander, C.W. Roberts, Sequential expres-
sion of macrophage anti-microbial/inammatory and wound healing
markers following innate, alternative and classical activation, Clin. Exp.
Immunol. 160 (2010) 369e379.
[127] J.M. Daley, S.K. Brancato, A.A. Thomay, J.S. Reichner, J.E. Albina, The pheno-
type of murine wound macrophages, J. Leukoc. Biol. 87 (2010) 59e67.
[128] R. Mirza, T.J. Koh, Dysregulation of monocyte/macrophage phenotype in
wounds of diabetic mice, Cytokine 56 (2011) 256e264.
[129] M. Spencer, A. Yao-Borengasser, R. Unal, N. Rasouli, C.M. Gurley, B. Zhu, et al.,
Adipose tissue macrophages in insulin-resistant subjects are associated with
collagen VI and brosis and demonstrate alternative activation, Am. J.
Physiol. Endocrinol. Metab. 299 (2010) E1016eE1027.
[130] D.R. Balce, B. Li, E.R. Allan, J.M. Rybicka, R.M. Krohn, R.M. Yates, Alternative
activation of macrophages by IL-4 enhances the proteolytic capacity of their
phagosomes through synergistic mechanisms, Blood 118 (2011) 4199e4208.
[131] R.D. Stout, S.K. Watkins, J. Suttles, Functional plasticity of macrophages: in
situ reprogramming of tumor-associated macrophages, J. Leukoc. Biol. 86
(2009) 1105e1109.
[132] J. Oh, A.E. Riek, S. Weng, M. Petty, D. Kim, M. Colonna, et al., Endoplasmic
reticulum stress controls M2 macrophage differentiation and foam cell for-
mation, J. Biol. Chem. 287 (2012) 11629e11641.
[133] W. Zhang, W. Xu, S. Xiong, Macrophage differentiation and polarization via
phosphatidylinositol 3-kinase/Akt-ERK signaling pathway conferred by
serum amyloid P component, J. Immunol. (Baltim. Md: 1950) 187 (2011)
1764e1777.
[134] G. Lay, Y. Poquet, P. Salek-Peyron, M.P. Puissegur, C. Botanch, H. Bon, et al.,
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 17
Langhans giant cells from M. tuberculosis-induced human granulomas
cannot mediate mycobacterial uptake, J. Pathol. 211 (2007) 76e85.
[135] L. Helming, S. Gordon, The molecular basis of macrophage fusion, Immu-
nobiology 212 (2007) 785e793.
[136] J.M. Anderson, A. Rodriguez, D.T. Chang, Foreign body reaction to bio-
materials, Seminars Immunol. 20 (2008) 86e100.
[137] M.T. Drake, B.L. Clarke, E.M. Lewiecki, The pathophysiology and treatment of
osteoporosis, Clin. Ther. 37 (8) (2015 Aug) 1837e1850.
[138] S.L. Teitelbaum, Osteoporosis and integrins, J. Clin. Endocrinol. Metab. 90
(2005) 2466e2468.
[139] A.K. McNally, J.M. Anderson, Beta1 and beta2 integrins mediate adhesion
during macrophage fusion and multinucleated foreign body giant cell for-
mation, Am. J. Pathol. 160 (2002) 621e630.
[140] A.K. McNally, S.R. Macewan, J.M. Anderson, alpha subunit partners to beta1
and beta2 integrins during IL-4-induced foreign body giant cell formation,
J. Biomed. Mater. Res. Part A 82 (2007) 568e574.
[141] W. Wiktor-Jedrzejczak, A. Bartocci, A.W. Ferrante Jr., A. Ahmed-Ansari,
K.W. Sell, J.W. Pollard, et al., Total absence of colony-stimulating factor 1 in
the macrophage-decient osteopetrotic (op/op) mouse, Proc. Natl. Acad. Sci.
U. S. A. 87 (1990) 4828e4832.
[142] L. Van Wesenbeeck, P.R. Odgren, C.A. MacKay, M. D'Angelo, F.F. Safadi,
S.N. Popoff, et al., The osteopetrotic mutation toothless (tl) is a loss-of-
function frameshift mutation in the rat Csf1 gene: evidence of a crucial
role for CSF-1 in osteoclastogenesis and endochondral ossication, Proc.
Natl. Acad. Sci. U. S. A. 99 (2002) 14303e14308.
[143] Y.Y. Kong, H. Yoshida, I. Sarosi, H.L. Tan, E. Timms, C. Capparelli, et al., OPGL is
a key regulator of osteoclastogenesis, lymphocyte development and lymph-
node organogenesis, Nature 397 (1999) 315e323.
[144] M. Yagi, T. Miyamoto, Y. Sawatani, K. Iwamoto, N. Hosogane, N. Fujita, et al.,
DC-STAMP is essential for cell-cell fusion in osteoclasts and foreign body
giant cells, J. Exp. Med. 202 (2005) 345e351.
[145] A. Vignery, Macrophage fusion: the making of osteoclasts and giant cells,
J. Exp. Med. 202 (2005) 337e340.
[146] A.J. Rao, E. Gibon, T. Ma, Z. Yao, R.L. Smith, S.B. Goodman, Revision joint
replacement, wear particles, and macrophage polarization, Acta Biomater. 8
(2012) 2815e2823.
[147] W.G. Brodbeck, J.M. Anderson, Giant cell formation and function, Curr. Opin.
Hematol. 16 (2009) 53e57.
[148] A.K. McNally, J.M. Anderson, Interleukin-4 induces foreign body giant cells
from human monocytes/macrophages. Differential lymphokine regulation of
macrophage fusion leads to morphological variants of multinucleated giant
cells, Am. J. Pathol. 147 (1995) 1487e1499.
[149] A.K. McNally, J.A. Jones, S.R. Macewan, E. Colton, J.M. Anderson, Vitronectin is
a critical protein adhesion substrate for IL-4-induced foreign body giant cell
formation, J. Biomed. Mater. Res. Part A 86 (2008) 535e543.
[150] L. Helming, S. Gordon, Macrophage fusion induced by IL-4 alternative acti-
vation is a multistage process involving multiple target molecules, Eur. J.
Immunol. 37 (2007) 33e42.
[151] J.L. Moreno, I. Mikhailenko, M.M. Tondravi, A.D. Keegan, IL-4 promotes the
formation of multinucleated giant cells from macrophage precursors by a
STAT6-dependent, homotypic mechanism: contribution of E-cadherin,
J. Leukoc. Biol. 82 (2007) 1542e1553.
[152] A.K. McNally, J.M. Anderson, Multinucleated giant cell formation exhibits
features of phagocytosis with participation of the endoplasmic reticulum,
Exp. Mol. Pathol. 79 (2005) 126e135.
[153] I. Lemaire, S. Falzoni, N. Leduc, B. Zhang, P. Pellegatti, E. Adinol, et al.,
Involvement of the purinergic P2X7 receptor in the formation of multinu-
cleated giant cells, J. Immunol. (Baltim. Md: 1950) 177 (2006) 7257e7265.
[154] A. Rodriguez, S.R. Macewan, H. Meyerson, J.T. Kirk, J.M. Anderson, The
foreign body reaction in T-cell-decient mice, J. Biomed. Mater. Res. Part A
90 (2009) 106e113.
[155] S. MacLauchlan, E.A. Skokos, N. Meznarich, D.H. Zhu, S. Raoof, J.M. Shipley, et
al., Macrophage fusion, giant cell formation, and the foreign body response
require matrix metalloproteinase 9, J. Leukoc. Biol. 85 (2009) 617e626.
[156] A.K. McNally, J.M. Anderson, Foreign body-type multinucleated giant cells
induced by interleukin-4 express select lymphocyte co-stimulatory mole-
cules and are phenotypically distinct from osteoclasts and dendritic cells,
Exp. Mol. Pathol. 91 (2011) 673e681.
[157] K.M. DeFife, C.R. Jenney, E. Colton, J.M. Anderson, Cytoskeletal and adhesive
structural polarizations accompany IL-13-induced human macrophage
fusion, J. Histochem. Cytochem.: Off. J. Histochem. Soc. 47 (1999) 65e74.
[158] C.R. Jenney, K.M. DeFife, E. Colton, J.M. Anderson, Human monocyte/
macrophage adhesion, macrophage motility, and IL-4-induced foreign body
giant cell formation on silane-modied surfaces in vitro. Student Research
Award in the Master's Degree Candidate Category, in: 24th Annual Meeting
of the Society for Biomaterials, San Diego, CA, April 22-26, 1998. J Biomed
Mater Res, 41, 1998, pp. 171e184.
[159] S. Chen, J.A. Jones, Y. Xu, H.Y. Low, J.M. Anderson, K.W. Leong, Character-
ization of topographical effects on macrophage behavior in a foreign body
response model, Biomaterials 31 (2010) 3479e3491.
[160] W.G. Brodbeck, J. Patel, G. Voskerician, E. Christenson, M.S. Shive,
Y. Nakayama, et al., Biomaterial adherent macrophage apoptosis is increased
by hydrophilic and anionic substrates in vivo, Proc. Natl. Acad. Sci. U. S. A. 99
(2002) 10287e10292.
[161] D.T. Chang, J.A. Jones, H. Meyerson, E. Colton, I.K. Kwon, T. Matsuda, et al.,
Lymphocyte/macrophage interactions: biomaterial surface-dependent
cytokine, chemokine, and matrix protein production, J. Biomed. Mater. Res.
Part A 87 (2008) 676e687.
[162] J.A. Jones, D.T. Chang, H. Meyerson, E. Colton, I.K. Kwon, T. Matsuda, et al.,
Proteomic analysis and quantication of cytokines and chemokines from
biomaterial surface-adherent macrophages and foreign body giant cells,
J. Biomed. Mater. Res. Part A 83 (2007) 585e596.
[163] D.T. Chang, E. Colton, J.M. Anderson, Paracrine and juxtacrine lymphocyte
enhancement of adherent macrophage and foreign body giant cell activation,
J. Biomed. Mater. Res. Part A 89 (2009) 490e498.
[164] J.M. Anderson, J.A. Jones, Phenotypic dichotomies in the foreign body reac-
tion, Biomaterials 28 (2007) 5114e5120.
[165] A.K. McNally, J.M. Anderson, Phenotypic expression in human monocyte-
derived interleukin-4-induced foreign body giant cells and macrophages
in vitro: dependence on material surface properties, J. Biomed. Mater. Res.
Part A 103 (2015) 1380e1390.
[166] M. Kronstr
om, B. Svensson, E. Erickson, L. Houston, P. Braham, G.R. Persson,
Humoral immunity host factors in subjects with failing or successful tita-
nium dental implants, J. Clin. Periodontol. 27 (2000) 875e882.
[167] T. Albrektsson, C. Dahlin, T. Jemt, L. Sennerby, A. Turri, A. Wennerberg, Is
marginal bone loss around oral implants the result of a provoked foreign
body reaction? Clin. Implant Dent. Relat. Res. 16 (2014) 155e165.
[168] R. Trindade, T. Albrektsson, P. Tengvall, A. Wennerberg, Foreign body reac-
tion to biomaterials: on mechanisms for buildup and breakdown of
Osseointegration, Clin. Implant Dent. Relat. Res. (2014 Sep 25) http://dx.doi.
org/10.1111/cid.12274 (Epub ahead of print).
[169] R. Trindade, T. Albrektsson, A. Wennerberg, Current concepts for the bio-
logical basis of dental implants: foreign body equilibrium and osseointe-
gration dynamics, Oral Maxillofac. Surg. Clin. N. Am. 27 (2015) 175e183.
[170] N. Saulacic, D.D. Bosshardt, M.M. Bornstein, S. Berner, D. Buser, Bone appo-
sition to a titanium-zirconium alloy implant, as compared to two other
titanium-containing implants, Eur. Cells Mater. 23 (2012) 273e286 discus-
sion 86e8.
[171] N. Saulacic, R. Erdosi, D.D. Bosshardt, R. Gruber, D. Buser, Acid and alkaline
etching of sandblasted zirconia implants: a histomorphometric study in
miniature pigs, Clin. Implant Dent. Relat. Res. 16 (2014) 313e322.
[172] V. Chappuis, Y. Cavusoglu, R. Gruber, U. Kuchler, D. Buser, D.D. Bosshardt,
Osseointegration of zirconia in the presence of multinucleated giant cells,
Clin. Implant Dent. Relat. Res. (2015 Sep 17) http://dx.doi.org/10.1111/cid.
12375 (Epub ahead of print).
[173] J. Takebe, C.M. Champagne, S. Offenbacher, K. Ishibashi, L.F. Cooper, Titanium
surface topography alters cell shape and modulates bone morphogenetic
protein 2 expression in the J774A.1 macrophage cell line, J. Biomed. Mater.
Res. Part A 64 (2003) 207e216.
[174] A.K. Refai, M. Textor, D.M. Brunette, J.D. Watereld, Effect of titanium surface
topography on macrophage activation and secretion of proinammatory
cytokines and chemokines, J. Biomed. Mater. Res. Part A 70 (2004) 194e205.
[175] K.S. Tan, L. Qian, R. Rosado, P.M. Flood, L.F. Cooper, The role of titanium
surface topography on J774A.1 macrophage inammatory cytokines and
nitric oxide production, Biomaterials 27 (2006) 5170e5177.
[176] S. Ghrebi, D.W. Hamilton, J. Douglas Watereld, D.M. Brunette, The effect of
surface topography on cell shape and early ERK1/2 signaling in macro-
phages; linkage with FAK and Src, J. Biomed. Mater. Res. Part A 101 (2013)
2118e2128.
[177] S. Makihira, Y. Mine, E. Kosaka, H. Nikawa, Titanium surface roughness ac-
celerates RANKL-dependent differentiation in the osteoclast precursor cell
line, RAW264.7, Dent. Mater. J. 26 (2007) 739e745.
[178] G. Valles, E. Gil-Garay, L. Munuera, N. Vilaboa, Modulation of the cross-talk
between macrophages and osteoblasts by titanium-based particles, Bio-
materials 29 (2008) 2326e2335.
[179] B. Chehroudi, S. Ghrebi, H. Murakami, J.D. Watereld, G. Owen,
D.M. Brunette, Bone formation on rough, but not polished, subcutaneously
implanted Ti surfaces is preceded by macrophage accumulation, J. Biomed.
Mater. Res. Part A 93 (2010) 724e737.
[180] G.N. Thalji, S. Nares, L.F. Cooper, Early molecular assessment of osseointe-
gration in humans, Clin. Oral Implants Res. 25 (2014) 1273e1285.
[181] T. Hefti, M. Frischherz, N.D. Spencer, H. Hall, F. Schlottig, A comparison of
osteoclast resorption pits on bone with titanium and zirconia surfaces,
Biomaterials 31 (2010) 7321e7331.
[182] V. Milleret, S. Tugulu, F. Schlottig, H. Hall, Alkali treatment of microrough
titanium surfaces affects macrophage/monocyte adhesion, platelet activation
and architecture of blood clot formation, Eur. Cells Mater. 21 (2011)
430e444 discussion 44.
[183] S. Hamlet, M. Alfarsi, R. George, S. Ivanovski, The effect of hydrophilic tita-
nium surface modication on macrophage inammatory cytokine gene
expression, Clin. Oral Implants Res. 23 (2012) 584e590.
[184] M.A. Alfarsi, S.M. Hamlet, S. Ivanovski, Titanium surface hydrophilicity
modulates the human macrophage inammatory cytokine response,
J. Biomed. Mater. Res. Part A 102 (2014) 60e67.
[185] A. Scislowska-Czarnecka, E. Menaszek, B. Szaraniec, E. Kolaczkowska,
Ceramic modications of porous titanium: effects on macrophage activation,
Tissue Cell 44 (2012) 391e400.
[186] J. Brinkmann, T. Hefti, F. Schlottig, N.D. Spencer, H. Hall, Response of oste-
oclasts to titanium surfaces with increasing surface roughness: an in vitro
study, Biointerphases 7 (2012) 34.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e1918
[187] K.A. Barth, J.D. Watereld, D.M. Brunette, The effect of surface roughness on
RAW264.7 macrophage phenotype, J. Biomed. Mater. Res. Part A 101 (2013)
2679e2688.
[188] Q.L. Ma, L.Z. Zhao, R.R. Liu, B.Q. Jin, W. Song, Y. Wang, et al., Improved
implant osseointegration of a nanostructured titanium surface via mediation
of macrophage polarization, Biomaterials 35 (2014) 9853e9867.
[189] J. Lu, T.J. Webster, Reduced immune cell responses on nano and submicron
rough titanium, Acta Biomater. 16 (2015) 223e231.
[190] Z. Chen, C. Wu, W. Gu, T. Klein, R. Crawford, Y. Xiao, Osteogenic differenti-
ation of bone marrow MSCs by beta-tricalcium phosphate stimulating
macrophages via BMP2 signalling pathway, Biomaterials 35 (2014)
1507e1518.
[191] S. Yamada, D. Heymann, J.M. Bouler, G. Daculsi, Osteoclastic resorption of
biphasic calcium phosphate ceramic in vitro, J. Biomed. Mater Res. 37 (1997)
346e352.
[192] M. Benahmed, J.M. Bouler, D. Heymann, O. Gan, G. Daculsi, Biodegradation of
synthetic biphasic calcium phosphate by human monocytes in vitro: a
morphological study, Biomaterials 17 (1996) 2173e2178.
[193] M. Benahmed, D. Heymann, P. Pilet, J. Bienvenu, G. Daculsi, LPS increases
biomaterial degradation by human monocytes in vitro, J. Biomed. Mater. Res.
34 (1997) 115e119.
[194] S.N. Silva, M.M. Pereira, A.M. Goes, M.F. Leite, Effect of biphasic calcium
phosphate on human macrophage functions in vitro, J. Biomed. Mater. Res.
Part A 65 (2003) 475e481.
[195] J.M. Rice, J.A. Hunt, J.A. Gallagher, Quantitative evaluation of the biocom-
patible and osteogenic properties of a range of biphasic calcium phosphate
(BCP) granules using primary cultures of human osteoblasts and monocytes,
Calcif. Tissue Int. 72 (2003) 726e736.
[196] J.M. Curran, J.A. Gallagher, J.A. Hunt, The inammatory potential of biphasic
calcium phosphate granules in osteoblast/macrophage co-culture, Bio-
materials 26 (2005) 5313e5320.
[197] Z. Xia, L.M. Grover, Y. Huang, I.E. Adamopoulos, U. Gbureck, J.T. Triftt, et al.,
In vitro biodegradation of three brushite calcium phosphate cements by a
macrophage cell-line, Biomaterials 27 (2006) 4557e4565.
[198] B.H. Fellah, N. Josselin, D. Chappard, P. Weiss, P. Layrolle, Inammatory re-
action in rats muscle after implantation of biphasic calcium phosphate micro
particles, J. Mater. Sci. Mater. Med. 18 (2007) 287e294.
[199] B.H. Fellah, B. Delorme, J. Sohier, D. Magne, P. Hardouin, P. Layrolle,
Macrophage and osteoblast responses to biphasic calcium phosphate micro
particles, J. Biomed. Mater. Res. Part A 93 (2010) 1588e1595.
[200] R.J. Egli, S. Gruenenfelder, N. Doebelin, W. Hofstetter, R. Luginbuehl,
M. Bohner, Thermal Treatments of Calcium Phosphate Biomaterials to Tune
the Physico-Chemical Properties and Modify the In Vitro Osteoclast
Response, Adv. Eng. Mater. 13 (2011) B102eB107.
[201] A.L. Gamblin, M.A. Brennan, A. Renaud, H. Yagita, F. Lezot, D. Heymann, et al.,
Bone tissue formation with human mesenchymal stem cells and biphasic
calcium phosphate ceramics: the local implication of osteoclasts and mac-
rophages, Biomaterials 35 (2014) 9660e9667.
[202] N.L. Davison, X. Luo, T. Schoenmaker, V. Everts, H. Yuan, F. Barrere-de Groot,
et al., Submicron-scale surface architecture of tricalcium phosphate directs
osteogenesis in vitro and in vivo, Eur. Cells Mater. 27 (2014) 281e297 dis-
cussion 96e7.
[203] N.L. Davison, B. ten Harkel, T. Schoenmaker, X. Luo, H. Yuan, V. Everts, et al.,
Osteoclast resorption of beta-tricalcium phosphate controlled by surface
architecture, Biomaterials 35 (2014) 7441e7451.
[204] N.L. Davison, J. Su, H. Yuan, J.J. van den Beucken, J.D. de Bruijn, F. Barrere-de
Groot, Inuence of surface microstructure and chemistry on osteoinduction
and osteoclastogenesis by biphasic calcium phosphate discs, Eur. Cells Mater.
29 (2015) 314e329.
[205] Y. Shiwaku, L. Neff, K. Nagano, K. Takeyama, J. de Bruijn, M. Dard, et al., The
crosstalk between osteoclasts and osteoblasts is dependent upon the
composition and structure of biphasic calcium phosphates, PLoS One 10
(2015) e0132903.
[206] H. Kweon, S.G. Kim, J.Y. Choi, Inhibition of foreign body giant cell formation
by 4- hexylresorcinol through suppression of diacylglycerol kinase delta
gene expression, Biomaterials 35 (2014) 8576e8584.
[207] Z. Chen, X. Mao, L. Tan, T. Friis, C. Wu, R. Crawford, et al., Osteoimmuno-
modulatory properties of magnesium scaffolds coated with beta-tricalcium
phosphate, Biomaterials 35 (2014) 8553e8565.
[208] Z. Chen, J. Yuen, R. Crawford, J. Chang, C. Wu, Y. Xiao, The effect of
osteoimmunomodulation on the osteogenic effects of cobalt incorporated
beta-tricalcium phosphate, Biomaterials 61 (2015) 126e138.
[209] Y. Tintut, J. Patel, M. Territo, T. Saini, F. Parhami, L.L. Demer, Monocyte/
macrophage regulation of vascular calcication in vitro, Circulation 105
(2002) 650e655.
[210] A. Shioi, M. Katagi, Y. Okuno, K. Mori, S. Jono, H. Koyama, et al., Induction of
bone-type alkaline phosphatase in human vascular smooth muscle cells:
roles of tumor necrosis factor-alpha and oncostatin M derived from mac-
rophages, Circ. Res. 91 (2002) 9e16.
[211] A.B. Blom, P.L. van Lent, A.E. Holthuysen, P.M. van der Kraan, J. Roth, N. van
Rooijen, et al., Synovial lining macrophages mediate osteophyte formation
during experimental osteoarthritis. Osteoarthritis and cartilage/OARS,
Osteoarthr. Res. Soc. 12 (2004) 627e635.
[212] P.L. van Lent, A.B. Blom, P. van der Kraan, A.E. Holthuysen, E. Vitters, N. van
Rooijen, et al., Crucial role of synovial lining macrophages in the promotion
of transforming growth factor beta-mediated osteophyte formation, Arthritis
Rheum. 50 (2004) 103e111.
[213] S. Kamekura, K. Hoshi, T. Shimoaka, U. Chung, H. Chikuda, T. Yamada, et al.,
Osteoarthritis development in novel experimental mouse models induced by
knee joint instability. Osteoarthritis and cartilage/OARS, Osteoarthr. Res. Soc.
13 (2005) 632e641.
[214] M. Kaneko, T. Tomita, T. Nakase, Y. Ohsawa, H. Seki, E. Takeuchi, et al.,
Expression of proteinases and inammatory cytokines in subchondral bone
regions in the destructive joint of rheumatoid arthritis, Rheumatol. Oxf. Engl.
40 (2001) 247e255.
[215] D.R. Haynes, S.J. Hay, S.D. Rogers, S. Ohta, D.W. Howie, S.E. Graves, Regulation
of bone cells by particle-activated mononuclear phagocytes, J. bone Jt. Surg.
Br. Vol. 79 (1997) 988e994.
[216] S.M. Lessner, H.L. Prado, E.K. Waller, Z.S. Galis, Atherosclerotic lesions grow
through recruitment and proliferation of circulating monocytes in a murine
model, Am. J. Pathol. 160 (2002) 2145e2155.
[217] F.K. Swirski, M.J. Pittet, M.F. Kircher, E. Aikawa, F.A. Jaffer, P. Libby, et al.,
Monocyte accumulation in mouse atherogenesis is progressive and pro-
portional to extent of disease, Proc. Natl. Acad. Sci. U. S. A. 103 (2006)
10340e10345.
[218] L. Landsman, L. Bar-On, A. Zernecke, K.W. Kim, R. Krauthgamer,
E. Shagdarsuren, et al., CX3CR1 is required for monocyte homeostasis and
atherogenesis by promoting cell survival, Blood 113 (2009) 963e972.
[219] C.S. Robbins, I. Hilgendorf, G.F. Weber, I. Theurl, Y. Iwamoto, J.L. Figueiredo,
et al., Local proliferation dominates lesional macrophage accumulation in
atherosclerosis, Nat. Med. 19 (2013) 1166e1172.
[220] J.M. Anderson, Exploiting the inammatory response on biomaterials
research and development, J. Mater. Sci. Mater. Med. 26 (2015) 121.
[221] D. Buser, V. Chappuis, U. Kuchler, M.M. Bornstein, J.G. Wittneben, R. Buser, et
al., Long-term stability of early implant placement with contour augmen-
tation, J. Dent. Res. 92 (2013) 176se182s.
[222] V. Chappuis, O. Engel, M. Reyes, K. Shahim, L.P. Nolte, D. Buser, Ridge al-
terations post-extraction in the esthetic zone: a 3D analysis with CBCT,
J. Dent. Res. 92 (2013) 195se201s.
[223] E. Katsuyama, H. Miyamoto, T. Kobayashi, Y. Sato, W. Hao, H. Kanagawa, et
al., Interleukin-1 receptor-associated kinase-4 (IRAK4) promotes inamma-
tory osteolysis by activating osteoclasts and inhibiting formation of foreign
body giant cells, J. Biol. Chem. 290 (2015) 716e726.
[224] S.K. Biswas, M. Chittezhath, I.N. Shalova, J.Y. Lim, Macrophage polarization
and plasticity in health and disease, Immunol. Res. 53 (2012) 11e24.
[225] E. Ingham, J. Fisher, The role of macrophages in osteolysis of total joint
replacement, Biomaterials 26 (2005) 1271e1286.
[226] S.Y. Yang, H. Yu, W. Gong, B. Wu, L. Mayton, R. Costello, et al., Murine model
of prosthesis failure for the long-term study of aseptic loosening, J. Orthop.
Res.: Off. Publ. Orthop. Res. Soc. 25 (2007) 603e611.
[227] C. Nich, Y. Takakubo, J. Pajarinen, M. Ainola, A. Salem, T. Sillat, et al., Mac-
rophages-Key cells in the response to wear debris from joint replacements,
J. Biomed. Mater. Res. Part A 101 (2013) 3033e3045.
[228] R.D. Stout, J. Suttles, Functional plasticity of macrophages: reversible adap-
tation to changing microenvironments, J. Leukoc. Biol. 76 (2004) 509e513.
[229] J.P. Edwards, X. Zhang, K.A. Frauwirth, D.M. Mosser, Biochemical and func-
tional characterization of three activated macrophage populations, J. Leukoc.
Biol. 80 (2006) 1298e1307.
[230] A.J. Fleetwood, H. Dinh, A.D. Cook, P.J. Hertzog, J.A. Hamilton, GM-CSF- and
M-CSF-dependent macrophage phenotypes display differential dependence
on type I interferon signaling, J. Leukoc. Biol. 86 (2009) 411e421.
[231] A. Gratchev, J. Kzhyshkowska, S. Kannookadan, M. Ochsenreiter, A. Popova,
X. Yu, et al., Activation of a TGF-beta-specic multistep gene expression
program in mature macrophages requires glucocorticoid-mediated surface
expression of TGF-beta receptor II, J. Immunol. Baltim. Md 1950) 180 (2008)
6553e6565.
[232] U.M. Gundra, N.M. Girgis, D. Ruckerl, S. Jenkins, L.N. Ward, Z.D. Kurtz, et al.,
Alternatively Activated Macrophages Derived from Monocytes and Tissue
Macrophages are Phenotypically and Functionally Distinct, vol. 123, 2014,
pp. e110e22.
[233] T. Krausgruber, K. Blazek, T. Smallie, S. Alzabin, H. Lockstone, N. Sahgal, et al.,
IRF5 promotes inammatory macrophage polarization and TH1-TH17 re-
sponses, Nat. Immunol. 12 (2011) 231e238.
[234] R. Lang, D. Patel, J.J. Morris, R.L. Rutschman, P.J. Murray, Shaping gene
expression in activated and resting primary macrophages by IL-10,
J. Immunol. Baltim. Md 1950) 169 (2002) 2253e2263.
[235] K.A. Shirey, L.E. Cole, A.D. Keegan, S.N. Vogel, Francisella tularensis live
vaccine strain induces macrophage alternative activation as a survival
mechanism, J. Immunol. Baltim. Md 1950) 181 (2008) 4159e4167.
[236] K.A. Shirey, L.M. Pletneva, A.C. Puche, A.D. Keegan, G.A. Prince, J.C. Blanco, et
al., Control of RSV-induced lung injury by alternatively activated macro-
phages is IL-4R alpha-, TLR4-, and IFN-beta-dependent, Mucosal Immunol. 3
(2010) 291e300.
[237] K.A. Shirey, W. Lai, L.M. Pletneva, C.L. Karp, S. Divanovic, J.C. Blanco, et al.,
Role of the lipoxygenase pathway in RSV-induced alternatively activated
macrophages leading to resolution of lung pathology, Mucosal Immunol. 7
(2014) 549e557.
[238] J. Xue, S.V. Schmidt, J. Sander, A. Draffehn, W. Krebs, I. Quester, et al.,
Transcriptome-based network analysis reveals a spectrum model of human
macrophage activation, Immunity 40 (2014) 274e288.
R.J. Miron, D.D. Bosshardt / Biomaterials 82 (2016) 1e19 19
... Macrophages can be activated in response to pathophysiological stimuli and polarized into two distinct phenotypes: pro-inflammatory classically activated macrophages (M1 phenotype) and anti-inflammatory alternatively activated macrophages (M2 phenotype). Following bone injury, M1 macrophages play a role in the early stages of inflammation, while M2 macrophages are involved in later bone regeneration processes [19,20]. M1 macrophages secrete various inflammatory factors such as tumor necrosis factor (TNF ) and interleukin-1 (IL1 ), which contribute to the inflammatory response. ...
Article
Full-text available
Background Biomaterials used in bone tissue engineering must fulfill the requirements of osteoconduction, osteoinduction, and osseointegration. However, biomaterials with good osteoconductive properties face several challenges, including inadequate vascularization, limited osteoinduction and barrier ability, as well as the potential to trigger immune and inflammatory responses. Therefore, there is an urgent need to develop guided bone regeneration membranes as a crucial component of tissue engineering strategies for repairing bone defects. Methods The mZIF-8/PLA membrane was prepared using electrospinning technology and simulated body fluid external mineralization method. Its ability to induce biomimetic mineralization was evaluated through TEM, EDS, XRD, FT-IR, zeta potential, and wettability techniques. The biocompatibility, osteoinduction properties, and osteo-immunomodulatory effects of the mZIF-8/PLA membrane were comprehensively evaluated by examining cell behaviors of surface-seeded BMSCs and macrophages, as well as the regulation of cellular genes and protein levels using PCR and WB. In vivo, the mZIF-8/PLA membrane’s potential to promote bone regeneration and angiogenesis was assessed through Micro-CT and immunohistochemical staining. Results The mineralized deposition enhances hydrophilicity and cell compatibility of mZIF-8/PLA membrane. mZIF-8/PLA membrane promotes up-regulation of osteogenesis and angiogenesis related factors in BMSCs. Moreover, it induces the polarization of macrophages towards the M2 phenotype and modulates the local immune microenvironment. After 4-weeks of implantation, the mZIF-8/PLA membrane successfully bridges critical bone defects and almost completely repairs the defect area after 12-weeks, while significantly improving the strength and vascularization of new bone. Conclusions The mZIF-8/PLA membrane with dual osteoconductive and immunomodulatory abilities could pave new research paths for bone tissue engineering.
... It has now been established that monocytes (macrophages), which appear in the infiltrate in the implantation zone, participate not only in the formation of connective tissue, but are also the main effector cells in immune reactions to biomaterials that modulate osteogenesis (Miron and Bosshardt, 2016). Studying envelope models demonstrated that the loss of macrophages around bone grafting materials can completely abolish their osteoinductive capacity, confirming their key role in modulating the immune sys-CELL AND TISSUE BIOLOGY Vol. ...
... Osteal macrophages (OsteoMacs) are a specific subset of macrophages present in bone tissues. Recently, it has been hypothesized that osteomacs play an essential role in implant osseointegration [40,53]. Studies accompanied with removal of osteomacs were associated with reduction in bone remodeling and repair that is also a major function of osteoclasts [40,54]. ...
Article
Full-text available
Introduction Cell signaling pathways are the biological reactions that control cell functions and fate. They also directly affect the body reactions to implanted biomaterials. It is well-known that dental implants success depends on a successful integration with the alveolar bone: “osseointegration” which events comprise early and later responses to the implanted biomaterials. The early events are mainly immune-inflammatory responses to the implant considered by its microenvironment as a foreign body. Later reactions are osteogenic aiming to regulate bone formation and remodeling. All these events are controlled by the cell signaling pathways in an incredible harmonious coordination. Aim The number of pathways having a role in osseointegration is so big to be reviewed in a single article. So the aim of this review was to study only two of the most relevant ones: the inflammatory Nuclear Factor Kappa B (NF-κB) pathway regulating the early osseointegration events and the osteogenic Wnt pathway regulating later events. Methods We conducted a literature review using key databases to provide an overview about the NF-κB and Wnt cell signaling pathways and their mutual relationship with dental implants. A simplified narrative approach was conducted to explain these cell signaling pathways, their mode of activation and how they are related to the cellular events of osseointegration. Results and conclusion NF-κB and Wnt cell signaling pathways are important cross-talking pathways that are affected by the implant’s material and surface characteristics. The presence of the implant itself in the bone alters the intracellular events of both pathways in the adjacent implant’s cellular microenvironment. Both pathways have a great role in the success or failure of osseointegration. Such knowledge can offer a new hope to treat failed implants and enhance osseointegration in difficult cases. This is consistent with advances in Omics technologies that can change the paradigm of dental implant therapy.
... Thus, F Loi et al. [60] suggested that consistent changes in macrophage phenotypes make a significant contribution to the process of osteogenesis. Macrophages control the physiological process of bone repair by secreting various factors that are osteoinductive and, conversely, inhibiting bone regeneration [61]. Intercellular interactions are mediated by the release into the local environment of exosomes (Exos) that are extracellular vesicles ranging in size from 40 to 150 nm, containing proteins, lipids and nucleic acids, including miRs [62,63]. ...
Article
Full-text available
Introduction Bone defect management is a critical stage of treatment and rehabilitation that still remains a challenging problem for traumatologists and orthopaedists. The need for tissue engineering techniques is due to limited abilities of the human body to correct bone tissue autoregeneration, especially in comorbid and elderly patients with osteoporosis. Bone autografts is a gold standard in those cases but is associated with certain restrictions. Regenerative medicine and stem cell biology development opened up capabilities to employ new methods for enhancement of bone tissue repair. A special interest of researchers is focused on mesenchymal stem cells and extracellular vesicles for bone tissue regeneration optimization. Purpose of this review was to show mesenchymal stem cells and exosomes effeciency in bone defect treatment. Materials and methods Open electronic databases of scientific literature, PubMed and e-Library, were used. The literature data search was carried out using the keywords: regenerative medicine, bone defects, exosomes, mesenchymal stem cells. Results and discussion The review presents current ideas about mesenchymal stem cells, their microenvironment and exosomes influence on bone tissue repair. Clinical need in effective bone regeneration is still high. Mesenchymal stem cells and acellular regenerative treatments have shown good results in bone defects repair and are perspective directions. Productive use of mesenchymal stem cells and exosomes in bone defects treatment requires further study of their mechanisms of action, the regenerative techniques efficacy and safety evaluation in preclinical and clinical studies. Conclusion The use of mesenchymal stem cells and cell-free regenerative approaches has demonstrated good results in the restoration of bone tissue defects and is a promising direction.
... Macrophages are immune cells that have a prominent role in bone homeostasis and the osseointegration of dental implants [147]. They have been found to rapidly accumulate on implant surfaces prior to bone formation, and their presence positively mediates new bone formation and mineralisation of bone [148,149]. ...
Article
Full-text available
This mapping review highlights the need for a new paradigm in the understanding of peri-implantitis pathogenesis. The biofilm-mediated inflammation and bone dysregulation (BIND) hypothesis is proposed, focusing on the relationship between biofilm, inflammation, and bone biology. The close interactions between immune and bone cells are discussed, with multiple stable states likely existing between clinically observable definitions of peri-implant health and peri-implantitis. The framework presented aims to explain the transition from health to disease as a staged and incremental process, where multiple factors contribute to distinct steps towards a tipping point where disease is manifested clinically. These steps might be reached in different ways in different patients and may constitute highly individualised paths. Notably, factors affecting the underlying biology are identified in the pathogenesis of peri-implantitis, highlighting that disruptions to the host-microbe homeostasis at the implant-mucosa interface may not be the sole factor. An improved understanding of disease pathogenesis will allow for intervention on multiple levels and a personalised treatment approach. Further research areas are identified, such as the use of novel biomarkers to detect changes in macrophage polarisation and activation status, and bone turnover.
Article
The process of bone healing is a complex orchestration involving various bone cells such as osteoblasts, osteoclasts, and mesenchymal stem cells (MSCs), along with immune cells like macrophages. Essential cytokines such as transforming growth factor-β (TGF-β), bone morphogenetic protein-2 (BMP-2), and vascular endothelial derived growth factor (VEGF) play pivotal roles in this intricate mechanism. Despite advancements in treating bone defects, the healing of significant bone injuries remains suboptimal, posing a critical challenge in clinical orthopedics. The bone healing process comprises three phases–inflammation, regeneration, and remodeling–each overlapping with the others. The early-stage, involving hematoma formation and immune cell activation, plays a crucial role in determining the subsequent bone healing process. Recent evidence suggests that biomaterials can influence hematoma and immune responses in bone healing, offering potential for improved osteoinductive and osteoconductive effects. This summary explores the impact of biomaterials on the initial-stage of bone healing, examining how their physiochemical properties can affect hematoma formation and interactions with immune cells. The findings propose strategies for bone tissue regeneration by targeting early-stage bone healing.
Article
One of the leading causes of hospitalization, disability and mortality of 50% of women and 20% of men over the age of 50 are bone fractures and their complications caused by diseases of the musculoskeletal system. In this regard, an active search for a solution to the problem associated with the limitations of the use of auto-, allo-, and xenografts in the clinic to replace bone defects initiated the development of a regenerative approach based on the gradual replacement of artificial material with growing bone tissue. Promising in this regard are materials based on calcium phosphates, which act as an active source of chemical elements (calcium, phosphorus, etc.), which can optimize the process of bone defect fusion and ensure the replacement of the implant with new bone tissue. The review summarizes literature data on local biological activity, target cells, and molecular effects of calcium phosphates. It has been shown that calcium phosphate materials are biocompatible, capable of adsorbing regulatory proteins and cells, influencing their genetic and secretory apparatus and triggering the process of MSC differentiation in the osteogenic direction. At the same time, the successful implementation of local mechanisms of osseointegration at the “bone/implant” interface reduces the risk of periprosthetic infection (PJI) and rejection of artificial devices. Further study and use of calcium phosphate materials will make it possible to make a significant breakthrough in solving modern problems of bone tissue regeneration associated with an accurate (digital) bioengineering approach based on additive technologies and artificial intelligence.
Article
Full-text available
Bioceramics are vital for treating bone defects, and bioactive glasses (exemplified by 45S5) and calcium phosphate ceramics (CaPs, exemplified by tricalcium phosphate [β‐TCP]) are extensively explored. β‐TCP exhibits superior biocompatibility, degradability, and osteoconductive properties than 45S5; however, it lacks bioactivity, such as mineralization capability. To harness the synergies of both, four 3D printing bioceramic scaffolds: 45S5, 70% 45S5 + 30% TCP, 30% 45S5 + 70% TCP, and TCP, are manufactured. Furthermore, the investigation elucidates the correlation between their in situ mineralization capabilities and the intracellular calcium oscillations within macrophages and determines how they impact macrophage phenotypic transitions. Notably, during bioceramic degradation, there is an initial rise followed by a decline in calcium ion concentration, which results in intracellular calcium ion oscillations within macrophages. In the 70% 45S5 + 30% TCP group, early release of calcium ions promotes M1 macrophage polarization. Subsequently, rapid in situ mineralization causes a decrease in extracellular calcium ions, thus accelerating the transition of M1 to M2 macrophages and facilitating bone repair. The present study reveals a novel mechanism through which bioceramics modulate macrophage polarization, offers new insights into the initial foreign body response to bioceramics and presents a perspective on expeditious progression toward tissue repair.
Article
Full-text available
While bone tissue is known for its inherent regenerative abilities, various pathological conditions and trauma can disrupt its meticulously regulated processes of bone formation and resorption. Bone tissue engineering aims to replicate the extracellular matrix of bone tissue as well as the sophisticated biochemical mechanisms crucial for effective regeneration. Traditionally, the field has relied on external agents like growth factors and pharmaceuticals to modulate these processes. Although efficacious in certain scenarios, this strategy is compromised by limitations such as safety issues and the transient nature of the compound release and half-life. Conversely, bioactive elements such as zinc (Zn), magnesium (Mg) and silicon (Si), have garnered increasing interest for their therapeutic benefits, superior stability, and reduced biotic risks. Moreover, these elements are often incorporated into biomaterials that function as multifaceted bioactive components, facilitating bone regeneration via release on-demand. By elucidating the mechanistic roles and therapeutic efficacy of the bioactive elements, this review aims to establish bioactive elements as a robust and clinically viable strategy for advanced bone regeneration.
Article
Full-text available
Bone loss around replacement prostheses may be related to the activation of mononuclear phagocytes (MNP) by prosthetic wear particles. We investigated how osteoblast-like cells were regulated by human MNP stimulated by particles of prosthetic material. Particles of titanium-6-aluminium-4-vanadium (TiAlV) stimulated MNP to release interleukin (IL)-1β, tumour necrosis factor (TNF)α, IL-6 and prostaglandin E 2 (PGE 2 ). All these mediators are implicated in regulating bone metabolism. Particle-activated MNP inhibited bone cell proliferation and stimulated release of IL-6 and PGE 2 . The number of cells expressing alkaline phosphatase, a marker associated with mature osteo-blastic cells, was reduced. Experiments with blocking antibodies showed that TNFα was responsible for the reduction in proliferation and the numbers of cells expressing alkaline phosphatase. By contrast, IL-1β stimulated cell proliferation and differentiation. Both IL-1β and TNFα stimulated IL-6 and PGE 2 release from the osteoblast-like cells. Our results suggest that particle-activated mono-nuclear phagocytes can induce a change in the balance between bone formation and resorption by a number of mechanisms.
Article
Full-text available
Periprosthetic osteolysis is a major cause of aseptic loosening in artificial joint replacement. It is assumed to occur in conjunction with the activation of macrophages. We have shown in vitro that human osteoblast-like cells, isolated from bone specimens obtained from patients undergoing hip replacement, phagocytose fine particles of titanium alloy (TiAlV). The human osteoblast-like cells were identified immunocytochemically by the presence of bone-specific alkaline phosphatase (BAP). With increasing duration of culture, a variable number of the osteoblastic cells became positive for the macrophage marker CD68, independent of the phagocytosis of particles, with a fine granular cytoplasmic staining which was coexpressed with BAP as revealed by immunodoublestaining. The metal particles were not toxic to the osteoblastic cells since even in culture for up to four weeks massively laden cells were vital and had a characteristic morphology. Cells of the human osteosarcoma cell line (HOS 58) were also able to phagocytose metal particles but had only a low expression of the CD68 antigen. Fluorescence-activated cell scanning confirmed our immunocytochemical results. Additionally, the cells were found to be negative for the major histocompatibility complex-II (MHC-II) which is a marker for macrophages and other antigen-presenting cells. Negative results of histochemical tests for tartrate-resistant acid phosphatase excluded the contamination by osteoclasts or macrophages in culture. Our observations suggest that the osteoblast can either change to a phagocytosing cell or that the phagocytosis is an underestimated property of the osteoblast. The detection of the CD68 antigen is insufficient to prove the monocytic lineage. In order to discriminate between macrophages and osteoblasts additional markers should be used. To our knowledge, this is the first demonstration of cells of an osteoblastic origin which have acquired a mixed phenotype of both osteoblasts and macrophages.
Article
The macrophage-specific antigen F4/80 has been localized in adult mouse bone and connective tissue. F4/80 positive cells form the centre of haemopoietic islands, line the periosteal and subendosteal bone surfaces and are a major component of connective tissue and the synovial membrane (presumptive type A cells). F4/80 is absent from fibroblasts, chondrocytes, osteoblasts, osteocytes, osteoclasts and a subpopulation of synovial lining cells (presumptive type B cells).
Article
It has been well established that human mononuclear phagocytes have the capacity to produce 1,25-dihydroxy-vitamin D3 [1,25(OH)3D3] and express the vitamin D receptor (VDR). However, 1 alpha-hydroxylase activity and VDR receptor expression during differentiation of monocytes (MO) into mature macrophages (MAC) have not been previously examined. The in vitro maturation of blood MO can serve as a model for the in vivo transformation of immature blood MO into MAC. Here, when cultured in the presence of serum, MO undergo characteristic changes in morphology, antigenic phenotype, and functional activity consistent with their differentiation into MAC. We serially measured 1,25(OH)2D3 and 24,25- dihydroxyvitamin D3 [24,25(OH)2D3] synthesis, specific [3H]-1,25(OH)2D3 binding, and VDR mRNA levels during in vitro maturation of MO into MAC and correlated these functions with maturation-associated changes in the phenotype (MAX.1 and CD71) and secretory repertoire (interleukin-1 beta [IL-1 beta], neopterin) of the cells. MO showed only little conversion of 25-(OH)D3 into 1,25(OH)2D3 (1.4 +/- 0.4 pmol/10(6) cells/6 h, n = 5) that increased gradually during maturation into MAC at day 8 of culture (5.3 +/- 4.3 pmol/10(6) cells/6 h, n = 5). Interferon-gamma (IFN-gamma) increased baseline 1,25(OH)2D3-synthesis approximately twofold during all phases of differentiation. The time course of increased 1,25(OH)2D3-synthesis correlated with enhanced secretion of neopterin and expression of MAX.1 and CD71. The addition of exogenous 1,25(OH)2D3 did not influence constitutive 1,25(OH)2D3 synthesis, but IFN-gamma-stimulated production was suppressed to baseline levels. Exogenous 1,25(OH)2D3 also stimulated 24,25(OH)2D3 synthesis in freshly isolated MO (from 1.0 +/- 0.8 pmol/6 h to 5.6 +/- 0.9 pmol), whereas matured MAC showed no 24,25(OH)2D3 synthesis. Furthermore, we examined the expression of the VDR during the differentiation process. VDR mRNA and protein were constitutively expressed in MO, whereas VDR was downregulated in mature MAC on both the mRNA and protein levels. Homologous upregulation of VDR protein by 1,25(OH)2D3 occurred in MO and, to a lesser degree, in MAC. In contrast, VDR mRNA concentrations were not influenced by 1,25(OH)2D3. Taken together, our results show that MO into MAC differentiation in vitro is associated with (1) an enhanced capacity to synthesize 1,25(OH)2D3, (2) a loss of 24,25(OH)2D3-synthesizing activity, and (3) a decrease in the expression of VDR mRNA and protein. Because 1,25(OH)2D3 was shown to induce differentiation of MO into MAC, our data sugest an autoregulatory mechanism of MO/MAC generation by 1,25(OH)2D3.
Article
With the aid of two-color immunofluorescence and flow cytometry, a new subset of cells coexpressing CD14 and CD16 antigens can be identified in human peripheral blood. Using the monoclonal antibody My4, these CD14+/CD16+ cells account for 2.2% of the mononuclear cells and form about 13% of all cells identified by the monocyte-specific CD14 monoclonal antibody. The CD14+/CD16+ cells can be assigned to the monocyte lineage based on typical morphology, on expression of additional monocyte-associated molecules, on the ability to form reactive oxygen intermediates and on the expression of monocyte- specific NaF-sensitive esterase. Light scatter analysis revealed lower forward angle and right angle light scatter for the CD14+/CD16+ cells compared with the regular monocytes, and the average cell size was determined to be 13.8 and 18.4 microns, respectively. Expression of class II antigens on these “small monocytes” was twofold higher compared with the regular monocytes. By contrast, the capacity to perform adherence to plastic surfaces, as well as the ability to phagocytize antibody-coated erythrocytes was clearly reduced in the CD14+/CD16+ monocyte subset as compared with the regular monocytes. Hence the CD14+/CD16+ cells appear to represent a new monocyte subset with a distinct functional repertoire. A survey of various tissues revealed that a large proportion of the alveolar macrophages, but not of the peritoneal macrophages, express the CD14+/CD16+ phenotype.
Article
The cytokines IL-4 and IL-13 inhibit the production of NO from activated macrophages through an unresolved molecular mechanism. We show here that IL-4 and IL-13 regulate NO production through depletion of arginine, the substrate of inducible NO synthase (iNOS). Inhibition of NO production from murine macrophages stimulated with LPS and IFN-gamma by IL-4 or IL-13 was dependent on Stat6, cell density in the cultures, and pretreatment for at least 6 h. IL-4/IL-13 did not interfere with the expression or activity of iNOS but up-regulated arginase I (the liver isoform of arginase) in a Stat6-dependent manner. Addition of exogenous arginine completely restored NO production in IL-4-treated macrophages. Furthermore, impaired killing of the intracellular pathogen Toxoplasma gondii in IL-4-treated macrophages was overcome by supplementing L-arginine. The simple system of regulated substrate competition between arginase and iNOS has implications for understanding the physiological regulation of NO production.