ArticlePDF AvailableLiterature Review

Abstract and Figures

Bone diseases are associated with great morbidity; thus, the understanding of the mechanisms leading to their development represents a great challenge to improve bone health. Recent reports suggest that a large number of molecules produced by immune cells affect bone cell activity. However, the mechanisms are incompletely understood. This review aims to shed new lights into the mechanisms of bone diseases involving immune cells. In particular, we focused our attention on the major pathogenic mechanism underlying periodontal disease, psoriatic arthritis, postmenopausal osteoporosis, glucocorticoid-induced osteoporosis, metastatic solid tumors, and multiple myeloma.
Content may be subject to copyright.
Review Article
Bone-Immune Cell Crosstalk: Bone Diseases
Giorgio Mori,1Patrizia D’Amelio,2Roberta Faccio,3and Giacomina Brunetti4
1Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
2Department of Medical Science, Section of Gerontology and Bone Metabolism Diseases, University of Torino, 10126 Torino, Italy
3Department of Orthopedics, Washington University School of Medicine, St. Louis, MO 63110, USA
4Department of Basic Medical Sciences, Neurosciences andSenseOrgans,SectionofHumanAnatomyandHistology,
University of Bari, 70124 Bari, Italy
Correspondence should be addressed to Giacomina Brunetti; giacomina.brunetti@uniba.it
Received  October ; Revised  January ; Accepted  January 
Academic Editor: Chikao Morimoto
Copyright ©  Giorgio Mori et al. is is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Bone diseases are associated with great morbidity; thus, the understanding of the mechanisms leading to their development
represents a great challenge to improve bone health. Recent reports suggest that a large number of molecules produced by immune
cells aect bone cell activity. However, the mechanisms are incompletely understood. is review aims to shed new lights into the
mechanisms of bone diseases involving immune cells. In particular, we focused our attention on the major pathogenic mechanism
underlying periodontal disease, psoriatic arthritis, postmenopausal osteoporosis, glucocorticoid-induced osteoporosis, metastatic
solid tumors, and multiple myeloma.
1. Introduction
Bone is an active tissue that undergoes continuous remod-
elling by two distinct processes, bone formation and bone
resorption []. ese events are strongly linked and tightly
regulated to maintain skeletal homeostasis []. e bone
cells responsible for the dual processes include the bone
resorbing cells, that is, the osteoclasts (OCs), which are
dierentiated cells derived from hematopoietic cells of the
monocyte-macrophage lineage and bone forming cells, that
is, the osteoblasts (OBs), which are of mesenchymal origin.
Alteration of the dierentiation/activity of OCs as well as
OBs leads to bone diseases. e close relationship between
theboneandtheimmunesystemhasbeenincreasingly
recognized, in particular during pathological conditions in
which activation of both systems occurs []. It is known that
inammation increase leads to an augment in the immune
function, which culminates in an increased production of
tumour necrosis factor (TNF) or receptor activator of NF-
kB ligand (RANKL) by activated T cells, that has been
linked to bone loss associated diseases (inammatory and
autoimmune disease, postmenopausal osteoporosis). Dier-
ent studies have been performed to identify the T cell subset
involved in osteoclastogenesis. In general, T cells could be
classied as eector-cytotoxic T population (CD+ cells) and
helper T cells (CD+ cells). CD+ T cells, upon activation
and expansion, develop into diverse T helper () cell subsets
secreting signature cytokine proles and mediating distinct
eector functions []. Until recently, T cells were divided
into  or  cells, depending on the cytokines they
produced (with  producing IFN-gamma and IL- and
 producing primarily IL-/IL-/IL-). Regulatory T cells
(Tregs, CD+CD+Foxp+) potently inhibit the function of
eector T cells []. A third subset of IL--producing eector
T helper cells, called  cells, has been more recently dis-
covered and characterized.  cells produce IL-, IL-F,
and IL-, thereby inducing a massive tissue reaction owing
to the broad distribution of the IL- and IL- receptors.
 cells support OC formation mostly through the expres-
sion of IL-, which is recognized to induce RANK expression
on OC precursors as well as RANKL production by cells sup-
porting OC formation [,]. IL- also makes possible local
inammation through the recruitment and the activation
of immune cells, leading to the release of proinammatory
molecules, as IL- and TNF𝛼[]. ese proinammatory
molecules increase RANKL expression and synergize with
Hindawi Publishing Corporation
Journal of Immunology Research
Volume 2015, Article ID 108451, 11 pages
http://dx.doi.org/10.1155/2015/108451
Journal of Immunology Research
RANKL signalling to maximize OC formation. A relatively
high expression of RANKL on  cells may also participate
in the enhanced osteoclastogenesis. Collectively,  cells
can be considered an osteoclastogenic subset; however,
they are not the only ones. In fact, activated T cells, expressing
high RANKL levels, have the ability to directly induce OC
dierentiation by acting on OC precursor cells [].
However, because T cells/immune cells also secrete a
variety of cytokines and express membrane-bound factors
other than RANKL, which could support OC formation,
mainly in pathological condition; this issue might be further
explored, together with the mechanisms that could modulate
their expression.
We describe recent eorts highlighting the prominent
role of immune system in the alteration of bone remod-
elling, thus favouring the development of many bone dis-
eases, such as periodontal disease (PD), psoriatic arthri-
tis (PsA), postmenopausal osteoporosis, glucocorticoid-
induced osteoporosis (GIO), metastatic solid tumors, and
multiple myeloma (MM).
Periodontal Disease. PD is a common complex infection of the
oral cavity that specically aects the gingiva, the periodontal
ligament, and the alveolar bone. It is characterized by an
inammatory response to bacteria present in the gingival
pocket [] and may remain conned to the gingiva or may
progress to extreme periodontal destruction with the loss
of the alveolar bone. PD is the main cause of tooth loss
among adults and is associated with important alteration
in facial aesthetics and defeat of masticatory and phonetics
function []. It is also well recognized that the presence of
only pathogenic bacteria is insucient to PD. Progression
of this disease occurs due to a combination of factors,
including the presence of periodontopathic bacteria, high
levels of proinammatory cytokines (IL-, TNF𝛼,IL-,IL-
, IL-, and IL-), prostaglandin E (PGE2),lowlevelsof
anti-inammatory cytokines including IL-, transforming
growth factor (TGF-𝛽), and retinoic acid []. Genetic factors
increase the susceptibility of some individuals in developing
this inammatory disease. It has been supported by reports
of familial aggregation of severe forms of the disease [], and
twin studies []. Recent candidate gene studies for periodon-
tal disease have focused on genes related to host immunity
and inammatory response such as cytokines, cell-surface
receptors, chemokines, enzymes, and antigen recognition.
Histological examination of periodontitis lesions reveals that
thegranulocytesappeartoplayakeyroleinthemain-
tenance of the periodontal health. ese cells are present
in the junctional epithelium in large numbers and they
isolate tissues from the bacteria action; thus, severe forms of
periodontitis frequently aect patients with diseases such as
leukocyte adhesion deciency and neutropenia. e failure
of granulocytes to transmigrate into the endothelium results
in an increase on the inammatory response and reduces
theprotectiveresponseagainstperiodontalpathogens.Inthe
presence of active disease, the epithelial migration causes
a deep periodontal pocket resulting in bacterial invasion,
inammation, and destruction of the connective tissue, with
subsequent bone loss and possible tooth loss. Langerhans
cells and dendritic cells of bone marrow origin, that are
located within the epithelium, are a connecting link with
acquiredimmunity.eadaptiveimmuneresponseisacti-
vated when the epithelial barrier, with its innate system,
is penetrated. e dendritic cells participate to the innate
inammatory response and moreover they capture and
present antigens to B and T cells of the acquired immune
system []. Activated CD T helper cells produce subsets
of cytokines with dierent immune responses:  and
 cells, respectively, associated with cellular and humoral
immunity []. e recently described  and Treg cells
have antagonistic roles as eector and suppressive cells [].
B cells dierentiate into plasma cells producing specic
antibodies. us, tissues aected by periodontitis become
colonized with both lymphocytes subtypes with B cells being
more represented than T cells. In a not progressive lesion,
IFN-𝛾increases the phagocytic activity of both neutrophils
and macrophages and hence contains the infection. In case of
a reduced innate immune response, a consequent weak 
response may not contain infection. Moreover, activated mast
cells determine a  response, B cell activation, and antibody
production. e antibodies can control the infection or, as in
thecaseofproductionofIgGinlargeamount,thelesionwill
persist. Sustained B cell activation may lead to IL- secretion
and periodontal disease progression.  cells have been
identied in the periodontal tissues. IL- mainly produced
by  has been shown to stimulate epithelial, endothelial,
andbroblasticcellstoproduceIL-,IL-,andPGE
2,thus
sustaining the disease progression. In addition, IL- induces
RANKL production by osteoblasts stimulating bone resorp-
tion. It has been demonstrated that periodontitis bacteria
induce a signicant increase in the production of IL- [].
According to recent studies, IL- signicantly enhances
RANKL and inhibits osteoprotegerin (OPG) expression in
human periodontal ligament cells []. It has been hypoth-
esized that  cells may be involved in modulation and
enhanced inammatory mediators production by gingival
broblasts in periodontal disease. Circulating T cells express
high levels of RANKL and spontaneously promote osteoclas-
togenesis in patients [].  and  cells, as well as B cells,
increase RANKL expression []. Other studies demonstrated
that also B cells produce RANKL in response to periodontal
pathogen stimulation []. Contrarily, Treg cells decrease
RANKL secretion whereas TGF-𝛽stimulates Treg cell dier-
entiation. is process is supported by retinoic acid and coun-
teracted by IL- and IL-. In chronic inammatory disease
as PD, retinoic acid levels are suppressed and Treg activity is
inhibited in favor of  pathogenic eect []. In PD, proin-
ammatory cytokines overcome anti-inammatory ones, and
 cells surmount Treg: this inammatory state determines
the destruction of connective tissue and alveolar bone.
Psoriatic Arthritis. Psoriasis is a chronic inammatory disease
of the skin; a considerable part of patients with psori-
asis develops an inammatory arthritis characterized by
increased bone remodeling with osteolysis called PsA [].
e mechanisms responsible for the development of the
PsAshouldbebetterexplained,buttheimmunesystemplays
the main role in the pathogenesis of this disorder, that has to
Journal of Immunology Research
be considered as a chronic inammation. erefore, patients
withpsoriasishaveelevatedlevelsofcirculatingneutrophils.
e cells play an important role; in particular,  and 
are involved in the pathogenesis of the disease. PsA is char-
acterizedbyTandBcellinltratesandneoangiogenesisin
the synovial membrane and by the overexpression of inam-
matory cytokines. PsA synovitis is indicated by hyperplasia
of the synovial lining cells and mononuclear cell inltration.
Moreover, ectopic lymphoid neogenesis appears. Fibroblasts
and T cells in PsA synovial uid induce osteoclastogenesis
and bone resorption, mediated by RANKL, TNF-𝛼,andIL-
[]. Inammatory cytokine set such as TNF-𝛼,IL-𝛽,IL-,
IFN-𝛾, IL-, IL-, IL-, and IL- were highly expressed in
synovial uid of PsA patients, while broblasts isolated from
their skin and joints secreted IL- and IL-; some of these
cytokineshavealsorecognizedosteoclastogenicfeatures.
Both T cell suppression and TNF-𝛼inhibitors are eective
in humans in the treatment of psoriasis. In PsA patients, there
is a great increase in the number of peripheral blood 
cells. us, recent studies indicated that  cells []are
the cells most signicantly involved in psoriasis. Like  and
 cells,  cells appear to be evolved in inducing acquired
immune responses against microorganisms, such as bacteria.
Abnormal  responses are believed to play a signicant
role in the onset of various autoimmune diseases. Moreover,
IL- is indispensable for  eector functions in immune
disorders and maintenance of  cells.
Orphan nuclear receptor ROR𝛾t(retinoid-relatedorphan
receptor gamma t) has been identied as specic 
transcription factor []. ROR𝛾tisinvolvedintheproduction
of IL- receptor (ILR) which is expressed by monocytes,
, , , NK, and dendritic cells. ILR has an
important role in stimulating  cells. IL- receptors
promote IL- transcription and  cell dierentiation via
enforced ROR𝛾t expression. IL- acts on cells that have been
dierentiated into  cells, potentiating ROR𝛾tactivity,and
participates in maintenance and proliferation of  cells.
Clinical trials studying the eects of anti-IL- and anti-
IL- neutralizing antibodies in PsA patients are in progress
[], while the rst results seemed to be not as impressive
as those for TNF-𝛼inhibitor therapy, a recent clinical trial
indicated that Brodalumab, an IL-RA inhibitor, determined
a signicant improvement, when administered for  weeks
to PsA patients []. Moreover, new studies demonstrated
that Ustekinumab, a monoclonal antibody against both IL-
 and IL- cytokines, interfering, respectively, with  and
 activity, improved signicantly PsA symptoms, although
similar ecacy of TNF-𝛼inhibitors needs about  weeks of
treatment to be achieved []. Although immune responses
mediated by IL- and IL- are not as evident as those with
TNF-𝛼,cellsappeartoplayanimportantroleinPsA.
e activation of natural immunity in PsA stimulates
 and  cells, which sustain autoimmune pathology.
ere is an interesting report regarding the relationship of
PsA with microbial infection that is suggestive of PsA patho-
genesis [].
e observation of the lack of symptoms improvement
in PsA patients underwent to HIV infection and thus to
CD+ reduction, suggested that cells cooperate in the
pathogenesis with CD+ cells []. Probably, CD+T cells
potentiates the production of cytokines in the synovial
membrane, and the cytokines induce broblast proliferation
promoting brosis [], that probably contribute to joint
stiness and ankylosis [].
Postmenopausal Osteoporosis.Postmenopausalosteoporosis
is a systemic skeletal disorder characterized by reduced bone
mineral density and microarchitectural deterioration of bone
tissue resulting in fragility and susceptibility to fractures
[] and uncoupling of osteoblast-mediated bone formation
and osteoclast-mediated bone resorption. Postmenopausal
osteoporosis stems from the cessation of ovarian function at
menopause and from genetic and nongenetic factors which
heightenandprolongtherapidphaseofbonelosschar-
acteristic of the early postmenopausal period. OC activity
increases aer menopause; these cells may be considered as
cells at the crossroad between immune system and bone as
their precursors circulate within the mononuclear fraction
of peripheral blood [] and they interact with other
immunecellsasTcells[].
OC precursors increase during estrogen deciency []
and in condition characterized by increased bone turnover as
bone metastases [,] or inammatory diseases [].
Estrogens act on OC formation and activity both directly
and indirectly, in particular their action is mediated through
theinuenceonimmunesystem[]. In particular, estrogen
loss upregulates OC formation and activity through an
increased production of proosteoclastogenic cytokines by
bone marrow cells [], OBs [], and immune cells [,].
Proinammatory and proosteoclastogenic cytokines as
macrophage colony stimulating factor (M-CSF) and RANKL
are increased during estrogen deciency [,].
Additional inammatory cytokines are responsible for
the upregulation of OC formation observed during estrogen
deciency; some of these molecules have a well-established
role in osteoclastogenesis and bone loss, while others have
not. Among these molecules, the most involved ones in
estrogen deciency bone loss appear to be TNF-𝛼, IL-, IL-
, an d I L- [ ,]. A key role of T cell-produced TNF𝛼
has been demonstrated also in bone metastasis [,].
Estrogens are key regulators of immune function as
demonstratedbothinanimalsandinhumans[,]. Des-
pite some inverse reports [,], the main body of literature
rmlysupportstheessentialroleofactivatedTcellsin
regulating bone loss induced by estrogen deciency [,,
,].
In humans, we have demonstrated a fundamental role for
T cells in postmenopausal bone loss. In particular, we showed
that osteoclastogenesis from peripheral blood precursors
occurs only in the presence of T cells and that T cells are more
active than in healthy post- and premenopausal controls [].
T cells from osteoporotic patients produce more RANKL
and TNF-𝛼, thus inducing OC formation and activity [].
It has also been demonstrated that hormone replacement
therapy decreases osteoclastogenic cytokine production in
postmenopausal women. RANKL expression on lympho-
cytes and marrow stromal cells is signicantly elevated during
estrogen deciency in humans and correlates directly with
Journal of Immunology Research
increases in bone resorption markers and inversely with
serum estrogen levels [].
Estrogen loss promotes T cell activation by increasing
antigen presentation [,]andincreasesthymusoutput
of T cells into peripheral blood []. Estrogen loss expands
theproliferationandlifespanofbonemarrowTcells[,]
increasing expression of class II transactivator (CIITA), a
transcriptional coactivator acting on MHCII promoter [,
,].
Estrogen deciency increases the number of activated
CDL-expressing T cells that promote the expression of M-
CSF and RANKL by stromal cells and downregulates the
production of OPG. e net result is a signicant increase
intherateofosteoclastogenesis[,]. is mechanism
was also described in bone loss due to increased PTH levels
[,]. It is known that the CD/CDL system is crucial
for T cell activation and several functions of the immune sys-
tem. It promotes macrophage activation and dierentiation,
antibody isotype switching, and the adequate organization of
immunological memory in B cells.
Also, the  cells have been implicated in ovariectomy-
induced bone loss; these cells increased aer ovariectomy
and stimulate osteoclastogenesis through IL- production
[]. is eect is reversed by treatment with estradiol. IL-
 increases OB production of proosteoclastogenic cytokines
as TNF𝛼, IL-, and RANKL; these eects are antagonized by
estradiol.
ActivatedTcellshavealsobeensuggestedtoinhibit
osteoclastogenesis by diverting early OC precursors towards
dendritic cells dierentiation []. Indeed T cells have the
capacity to generate both osteoclastogenic cytokines such
as RANKL and TNF-𝛼[], as well as antiosteoclastogenic
factorssuchasIL-.Ithasalsobeensuggestedthattheeects
ofactivatedTcellsonosteoclastogenesisin vitro depend on
the manner in which they are activated []. e net eect
of T cells on OC formation may consequently represent the
prevailing balance of anti- and proosteoclastogenic T cell
cytokine secretion. However, in humans, T cells seem to be
proosteoclastogenic in dierent diseases including estrogen
deciency [,,,].
Taken together, these observations demonstrate the
causal relation among estrogen deprivation, T cell activation,
increased cytokines production, and bone demineralization.
Also another type of immune cell the B cell has recently
been studied as directly implicated in the regulation of bone
resorption and may be directly involved in the pathogenesis
of postmenopausal osteoporosis. Recent data have shown
that B cells are the dominant producers of OPG in the bone
microenvironment in vivo []. In fact, B cell KO mice
have an osteoporotic phenotype with enhanced osteoclastic
bone resorption and reconstitution with B cells by adoptive
transfer, completely rescued mice from development of osteo-
porosis, and normalizing OPG production [].
In human and animal B cells, OPG production can be sig-
nicantly upregulated by the activation of CD []. In line
with these data, both CD and CDL KO mice displayed an
osteoporotic phenotype and a signicant deciency in bone
marrow OPG concentrations [].
us, the emerging data suggest that the B lineage, rather
than the OB lineage, is likely the major source of OPG in the
bone microenvironment and that T cell signalling to B cells,
through the costimulatory molecules CDL and CD,
plays an important role in regulating basal OC formation and
in regulating bone homeostasis.
On the other hand, it has been recently demonstrated that
activated B cells overexpress RANKL, contributing to bone
resorption [,] and that ovariectomy in mice increases
thenumberofRANKL-expressingBlymphocytesinthebone
marrow [].
A recent paper shows that mice lacking RANKL in B cells
were partially protected from the ovariectomy-induced loss
of cancellous bone []. e role of B-lymphocytes has also
beenevaluatedindiseasecharacterizedbyfocalboneloss
as in periodontal inammation [,,]andrheumatoid
arthritis []. In rheumatoid arthritis, a recent paper sug-
gests that B cells depletion ameliorates the suppressed bone
turnover [].
Taken together, these data suggest that B-lymphocyte
involvement in the adaptive immune response contributes to
bone resorption by the upregulation of RANKL expression
through Toll-like receptor pathways and aligns with the
known ability of T cells to produce RANKL in the presence
of immune stimulus and to increase osteoclastogenesis. e
eect of estrogen deciency on B cell modulation may be one
of the mechanisms through which menopause aects bone
metabolism.
us, the involvement of T and B cells in the control
of bone turnover may provide a novel explanation for the
propensity to osteopenia and osteoporosis development aer
the cessation of ovarian function.
Glucocorticoid-Induced Osteoporosis.GIOisthemostfre-
quent origin of secondary osteoporosis in adults due to the
direct eects of glucocorticoids (GCs) on bone cells [].
GCs primarily aect trabecular bone, whereas the cortical
bone mass is reduced to a lower and slower extent. us,
fractures of the vertebrae are more recurrent. GC exposure
determines a rapid and early phase of bone loss, which is
the consequence of bone resorption exacerbation. is phase
is followed by a more chronic and progressive phase in
which bone mass declines because of impaired OB activity.
GCs augment RANKL expression and reduce OPG levels
in stromal and osteoblastic cells leading to the initial phase
of rapid bone loss. Further, GCs increase MCSF expression
as well as receptor subunits for osteoclastogenic cytokines
of the gp family. However, the main pathophysiological
mechanismofGIOistheimpairedboneformation,dueto
reduced OB formation and activity [,]. GCs impair on
OBs the synthesis of type I collagen, the major protein in
bone matrix. GCs may also inuence osteocyte metabolism
and function, modifying the elastic modulus adjacent to the
osteocyte lacunae leading to reduced mineral to matrix ratios
in the same areas with an enlargement of the lacunar size.
Besides the GC direct actions on bone cells, GC extraskeletal
eects on calcium metabolism have been reported. In par-
ticular, GCs decrease renal tubular calcium reabsorption and
Journal of Immunology Research
calcium absorption from the gastrointestinal tract is reduced
by mechanisms that oppose vitamin D action [].
GCs also impair bone metabolism during the growth.
In particular, in animal models, GC administration during
growth is the cause of decreased bone formation and resorp-
tion, reductions in the age-dependent increases in trabecular
bone mineral and trabecular thickness, and reductions in lin-
ear growth and accrual of cortical thickness in the femur [].
A decrease of bone mineral density (BMD) has been
reported in numerous pediatric diseases that require
GCs, both as long term replacement therapy, such as -
hydroxylase deciency (-OHD), and as treatment of acute
phase, such as asthma, systemic lupus erythematosus, juve-
nile rheumatoid arthritis, inammatory bowel disease, organ
transplantation, and steroid sensitive nephrotic syndrome
[]. In particular, in -OHD patients on chronic GC ther-
apy, the high osteoclastogenic potential of peripheral blood
mononuclear cells has been reported []. It is supported by
boththepresenceofcirculatingOCprecursorsandRANKL
released by T cells []. Further, high dickkopf- (DKK)
levels, a secreted antagonist of the Wnt/𝛽-catenin pathway,
have been demonstrated in sera and circulating monocytes,
T cells, and neutrophils from -OHD patients [].
Multiple Myeloma. MM is a haematological malignancies,
characterized by the clonal proliferation of plasma cells in
thebonemarrow[]. A major number of mechanisms have
been proposed to explain the increased formation and activ-
ity of the bone resorbing cells, the OCs in MM bone disease,
whereas few mechanisms have been identied to explain the
impairment of the bone forming cells, the OBs. In particular,
MM cells produce dierent cytokines that directly or indi-
rectly aect the bone cell activity, such as IL-, MIP-alpha,
IL-, DKK, and sclerostin []. e proposed mecha-
nism is that MM cells adhere to bone marrow stromal cells
(BMSCs) and induce the secretion of numerous proosteo-
clastogenic and antiosteoblastogenic cytokines. e adhesion
involved integrins such as CTLA- and VLA- expressed by
MM cells and VCAM- expressed on BMSCs [].
Moreover, it has previously demonstrated an important
role of T cells in supporting the formation and sur vival of OCs
from peripheral blood mononuclear cells (PBMCs) isolated
from MM patients with osteolysis, through the expression of
high levels of RANKL and decoy receptor (DcR) [,].
Interestingly, Giuliani et al. showed that malignant human
myeloma cells stimulate RANKL expression in T cells [].
Additionally, other authors demonstrated the high expression
levels of IL- in T cells from MM patients []. IL-
 plays a key function in the progression of bone disease
in MM, since the levels of IL- are higher in the more
advanced bone disease. IL- is also able to increase RANKL
expression on BMSCs, thus determining osteoclastogenesis
increase and consequently the development of bone lesions
[]. e amount of  in the bone marrow positively
correlated with the number of osteolytic lesions []as
well as the clinical tumor stage []. Very recently, the
involvement of LIGHT/TNFSF has been reported in MM-
bone disease []. LIGHT is a newly identied member of
the TNF superfamily, expressed by activated leukocytes [].
Recent literature data linked the high serum levels of LIGHT
with the bone loss associated with rheumatoid arthritis [].
Higher expression levels of LIGHT were found in CD+
T cells, monocytes, and neutrophils from osteolytic MM
patients with respect to the same cells from asymptomatic
MM patients as well as monogammopaty of undetermined
signicant (MGUS) and healthy subjects. Further, LIGHT
inhibition signicantly reduces OC formation from PBMCs
of osteolytic MM patients and stimulates OB dierentiation
in cultures derived from MM bone marrow mononuclear
cells, as demonstrated by the increase of colony forming units
of OBs and by the upregulation of osterix transcription factor,
bone sialoprotein, and osteocalcin bone matrix proteins.
Bone Metastatic Tumors. e skeleton is the predominant
metastatic site for many cancers, including breast, prostate,
and lung cancers [,]. Tumor invasion into bone is
associated with dramatic skeletal related events (SRE) such
as fractures, bone pain, hypercalcemia, and spinal cord com-
pression []. e current model for the pathophysiology of
bone metastasis centers on the interaction between tumor
cellsandOCsandisknownasthe“tumor/bonebone
vicious cycle. Tumor cells secrete a plethora of factors and
cytokines that can directly stimulate OC activation. Once
mature OCs start to resorb the bone, they release bone-
stored factors, such as TGF-𝛽, that further stimulate tumor
cell recruitment and proliferation []. Animal studies have
shown that antiresorptive therapies protect from SRE and
reduce tumor burden. us, antiresorptive agents, such as
zoledronic acid (ZOL) and the anti-RANKL monoclonal
antibody (Ab), denosumab, are widely used in the clinic in
patients with bone metastasis [,,]. Despite reducing
tumor-associated bone complications, recent meta-analysis
studies show controversial results on the antitumor eects of
OC blockade in breast cancer patients with bone metastasis
[,]. A signicant fraction of breast cancer patients with
bone metastases shows progression in their bone disease
while they are on potent antiresorptive agent treatment [
]. A recent study suggested the existence of a preosteolytic
early phase of bone metastasis that is independent of OC
activation []. Considering the complexity of the bone
microenvironment, serving as home to hematopoietic stem
cells and their progeny, which constitute the immune system,
it is logical to consider the interactions between tumor cells
and immune cells as potentially important regulators of bone
metastasis beyond the OC.
PresenceofactivatedCD+andCD+Tcellshasbeen
observed in the bone marrow of untreated patients with
breast cancer []. CD+ T cells have the capacity to specif-
ically identify and eliminate tumor cells via recognition of
tumor-specic antigens. Activated CD+ T cells can further
facilitate the development of cytotoxic CD+ T cells by
secreting numerous cytokines, including Interferon 𝛾(IFN).
Interferon 𝛾(IFN) exerts antiproliferative [], proapoptotic
[], and angiostatic [] eects resulting in the killing of a
proportion of the tumor. us, presence of CD+ and CD+
T cells at tumor site is a good prognostic indicator. However,
whetherTcellsmodulatebonemetastaticdissemination
and/or tumor growth in the bone microenvironment is not
Journal of Immunology Research
totally clear. In a recent report, Bidwell et al. demonstrated
that silencing of IFN regulatory factor (Irf), a transcription
factor controlling the induction of IFN genes, in breast
cancercellspromotesbonemetastasesthroughescapingfrom
immune control []. Importantly, an association with low
expressionofIrfsignaturesinprimarybreasttumorsand
higher number of bone metastatic events has been observed
[].isndingisastrongindicationthattheimmune
system can modulate metastatic dissemination to bone in
breast cancer patients.
Using animal models with established T cell immune
deciencies, we have also demonstrated that CD+ and
CD+ T cell populations exert antitumor eects in the
context of bone metastases []. We found that depletion of
both CD+ or CD+ T cell subsets can reduce the antitumor
eects ZOL in animals with bone metastases. Importantly,
ZOLtreatmentisstillhighlyeectiveinsuppressingtumor-
induced bone loss []. Conversely, T cell activation induced
by administration of anti-CTLA Ab can signicantly reduce
bone tumor burden []. ese observations have important
clinical implications and suggest that reduced T cell numbers
or impaired T cell activation might be the cause for the failure
of ZOL to reduce tumor burden and increase survival in
breast cancer patients.
Developing neoplasms can also acquire the ability to
escape CD+ T cell cytotoxicity by promoting expansion of
-polarized CD+ T helper and regulatory T cells, as well
as immune suppressor cells of myeloid origin reviewed in
[]. Monteiro et al. recently found that CD+ T cells
isolated from bone marrow of tumor bearing mice are potent
stimulators of osteoclastogenesis []. is subset of tumor-
specic CD+ T cells has the ability to promote OC activation
and induce osteolytic bone disease even before seeding of
tumor cells in the bone microenvironment. Importantly,
when tumor-specic CD+ T cells are adoptively transferred
into mice orthotopically injected with T tumor cells, tumor
colonization to bone, but not to other metastatic sites, is
increased. Whether this particular population of CD+ T
cells is increasing tumor bone metastases by aecting the
OCs or also by inducing an immune suppressive environment
needs to be established.
e bone microenvironment is particularly enriched in a
highly heterogeneous population of immature myeloid pro-
genitor cells that have the ability to exert immune suppressive
eects in the presence of a tumor. is immature myeloid
population, herein referred to as myeloid derived suppressor
cells (MDSCs), represents –% of the total bone marrow
cells of na¨
ıve mice and is further expanded up to –%
of total marrow cells depending on the tumor type [].
Circulating MDSCs are detected in the blood of patients with
various types of cancer []. In response to factors secreted
by a tumor, MDSCs leave the bone marrow and are found in
highnumbersincirculation,spleen,andtumorsiteswhere
they induce suppression of cytotoxic T cells []. MDSCs
exert their proneoplastic eects through the release of small
soluble oxidizers, by altering T cell/antigen recognition, and
depletion of essential amino acids from the local extracellular
environment, all ultimately leading to T cell suppression
[]. In addition, MDSCs can induce the expansion
ofregulatoryTcells,asubtypeofTcellsexertingimmune
suppressive functions. Furthermore, direct eects of MDSCs
on tumor proliferation through overproduction of cytokines
and angiogenic factors have also been proposed [].
A correlation between high MDSC numbers, advanced
stageofmalignancy,andpoorprognosishasbeenobserved.
We have recently shown that increased bone metastasis in
PLC𝛾/mice is due to suppression of antitumor T cells
responses. Although PLC𝛾 is not expressed by T cells, we
found that PLC𝛾/mice have increased MDSC numbers
with more potent immune suppressive eects than WT
[]. Downregulation of PLC𝛾activationalsooccursinthe
MDSCs of patients with advanced pancreatic cancer [].
Recent evidence also indicates that MDSCs participate
in the preparation of premetastatic niches where they create
a favorable environment for subsequent tumor colonization
[,]. Accumulation of MDSCs in bone marrow has been
observed during early stages of MM []. A role for MDSCs
in promoting tumor growth in bone through the OCs has
also been proposed. Zhuang et al. discovered that MDSCs
from mice injected with MM cells have increased osteoclasto-
genic potential. Importantly, coinjection of tumor-challenged
MDSCs together with MM cells leads to increased tumor
burden and osteolytic lesions, an eect that is inhibited
by administration of ZOL []. Similarly, Sawant et al.,
usinganimmunecompetentmodelofbreastcancerbone
metastases, showed that MDSCs isolated from the tumor
bone microenvironment dierentiate into resorbing OCs in
vitro. Remarkably, MDSCs isolated from tumor-free mice
or tumor-bearing animals without bone metastases lack the
ability to undergo OC dierentiation []. is impor-
tant observation suggests that there are intrinsic dierences
between MDSCs, depending on the tumor location. Why
MDSCs from mice bearing bone metastases have the ability
to dierentiate into OCs might depend on the proosteoclas-
togenic rich cytokine milieu that characterizes the tumor
bone microenvironment. However, it is unlikely that the
bone tumor promoting eects of this subset of MDSCs is
primarily dependent on their ability to dierentiate into
OCs. PLC𝛾/mice display increased bone metastatic
dissemination and higher MDSC numbers, but deletion of
PLC𝛾/also impairs the OC dierentiation process [,
]. us, in the context of PLC𝛾/deciency, MDSCs are
more likely to support tumor growth in bone by suppressing
T cell activity. All together, these studies indicate that MDSCs
are central players in the tumor/bone vicious cycle either
through suppression of antitumor T cell responses or through
dierentiation into resorbing OCs.
Unfortunately to date there is no curative treatment
for bone metastasis. Tumor cells that reach the bone envi-
ronment are usually resistant to the current antitumor
therapeutic approaches. e only options for these patients
are palliative treatments to reduce bone pain and prevent
additional bone destruction. More studies are needed to
exploit the importance of antitumor and tumor promoting
immune responses in patients with bone metastases and
whether manipulation of T cell-MDSC interactions could
oer therapeutic advantages to maximize the antitumor
eects of OC blockade.
Journal of Immunology Research
T : Established and possible novel therapeutic targets in the
dierent bone diseases.
(a)
Established
therapeutic
targets
Pathologies References
TNF-𝛼PsA []
RANKL
PsA, osteoporosis,
MM, and bone
metastatic tumors
[]
IL- PsA []
IL- PsA [,]
IL-RA PsA []
(b)
Possible
novel
therapeutic
targets
Pathologies References
LIGHT MM []
DKK Bone metastasis and
GIO []
IL- MM []
MDSC
targeting Bone metastasis [,]
2. Conclusions
e reviewed mechanisms underlying the bone disease
clearly highlighted the key involvement of the cells with an
immunological role. Further, it is also clear that numerous
pathways are common to the dierent diseases, whereas
others are disease-specic. us, these recent ndings repre-
sent an important issue, leading to the identication of new
therapeutic targets, mainly biological drugs, which in the last
years are in strong development (Tabl e ).
Conflict of Interests
e authors declare that there is no conict of interests
regarding the publication of this paper.
References
[] L. J. Raggatt and N. C. Partridge, “Cellular and molecular mech-
anisms of bone remodeling, e Journal of Biological Chem-
istry,vol.,no.,pp..
[] R . Tamma and A. Zallone, “Osteoblast and osteoclast crosstal ks:
from OAF to Ephrin, Inammation and Allergy—Drug Targets,
vol. , no. , pp. –, .
[] G. Mori, P. D’Amelio, R. Faccio, and G. Brunetti, “e interplay
between the bone and the immune system, Clinical and
Developmental Immunology,vol.,ArticleID,
pages, .
[] K. Sato, A. Suematsu, K. Okamoto et al., “ functions as an
osteoclastogenic helper T cell subset that links T cell activation
and bone destruction, JournalofExperimentalMedicine,vol.
, no. , pp. –, .
[] I. E. Adamopoulos, C. C. Chao, R. Geissler et al., “Interleukin-
A upregulates receptor activator of NF-kappaB on osteoclast
precursors, Arthritis Research and erapy,vol.,no.,article
R, .
[] K.SatoandH.Takayanagi,“Osteoclasts,rheumatoidarthritis,
and osteoimmunology,” Current Opinion in Rheumatology,vol.
,no.,pp.,.
[] A.B.BerezowandR.P.Darveau,“Microbialshiandperiodon-
titis, Periodontology 2000,vol.,no.,pp.,.
[] P. E. Petersen and H. Ogawa, “Strengthening the prevention of
periodontal disease: the WHO approach, Journal of Periodon-
tology,vol.,no.,pp.,.
[] E.GemmellandG.J.Seymour,“Immunoregulatorycontrolof
/ cytokine proles in periodontal disease, Periodontol-
ogy 2000,vol.,pp.,.
[] L. Sax´
en, “Heredity of juvenile periodontitis, Journal of Clinical
Periodontology,vol.,no.,pp.,.
[] B. S. Michalowicz, D. Aeppli, J. G. Virag et al., “Periodontal nd-
ings in adult twins, Journal of Periodontology,vol.,no.,pp.
–, .
[] C. W. Cutler and R. Jotwani, Antigen-presentation and the role
of dendritic cells in periodontitis, Periodontology 2000,vol.,
pp.,.
[] K. M. Murphy and S. L. Reiner, “e lineage decisions of helper
Tcells,Nature Reviews Immunology,vol.,no.,pp.,
.
[] T. Oda, H. Yoshie, and K. Yamazaki, Porphyromonas gingivalis
antigen preferentially stimulates T cells to express IL- but not
receptor activator of NF-𝜅B ligand in vitro,” Oral Microbiology
and Immunology,vol.,no.,pp.,.
[] D. Lin, L. Li, Y. Sun et al., “IL- regulates the expressions
of RANKL and OPG in human periodontal ligament cells via
TRAF/TBK-JNK/NF-𝜅Bpathways,Immunology,.
[] G. Brunetti, S. Colucci, P. Pignataro et al., “T cells support
osteoclastogenesis in an in vitro model derived from human
periodontitis patients, Journal of Periodontology,vol.,no.,
pp.,.
[] T. K a w a i , T. Mat suy a m a , Y. H o s o k awa et a l . , B a n d T lympho -
cytes are the primary sources of RANKL in the bone resorptive
lesion of periodontal disease, e American Journal of Pathol-
ogy,vol.,no.,pp.,.
[] C.T.WeaverandR.D.Hatton,“InterplaybetweentheTH
and TReg cell lineages: a co-evolutionary perspective, Nature
Reviews Immunology,vol.,no.,pp.,.
[] A. Menter, A. Gottlieb, S. R. Feldman et al., “Guidelines of care
for the management of psoriasis and psoriatic arthritis. Section
. Overview of psoriasis and guidelines of care for the treatment
of psoriasis with biologics, Journal of the American Academy of
Dermatology, vol. , no. , pp. –, .
[] M. L. Acosta Felquer, L. C. Coates, E. R. Soriano et al., “Drug
therapies for peripheral joint disease in psoriatic arthritis: a
systematic review,” e Journal of Rheumatology, vol. , no. ,
pp. –, .
[] G. Brunetti, R. Rizzi, A. Oranger et al., LIGHT/TNFSF
increasesosteoclastogenesisanddecreasesosteoblastogenesis
in multiple myeloma-bone disease, Oncotarget,vol.,no.,
pp. –, .
[] M. F. Faienza, V. Luce, A. Lonero et al., “Treatment of osteo-
porosis in children with glucocorticoid-treated diseases, Expert
Review of Endocrinology & Metabolism,vol.,no.,pp.
, .
Journal of Immunology Research
[] G. A. Clines and T. A. Guise, “Molecular mechanisms and treat-
ment of bone metastasis, Expert Reviews in Molecular Medicine,
vol. , article e, .
[] T. A. Guise, “Molecular mechanisms of osteolytic bone metas-
tases, Cancer, vol. , no. , pp. –, .
[] M. Mazzantini and O. di Munno, “Glucocorticoid-induced
osteoporosis:  update, Reumatismo,vol.,no.,pp.
, .
[] H. K. Fizazi, A. Lipton, X. Mariette et al., “Randomized phase
II trial of denosumab in patients with bone metastases from
prostate cancer, breast cancer, or other neoplasms aer intra-
venous bisphosphonates, Journal of Clinical Oncology,vol.,
no. , pp. –, .
[] S. P. Iyer, J. T. Beck, A. K. Stewart et al., “Phase IB multicentre
dose-determination study of BHQ in combination with
anti-myeloma therapy and zoledronic acid in patients with
relapsed or refractory multiple myeloma and prior skeletal-
related events,” British Journal of Haematology,vol.,no.,
pp. –, .
[] G. Brunetti, M. F. Faienza, L. Piacente et al., “High dickkopf-
levels in sera and leukocytes from children with -hydroxylase
deciency on chronic glucocorticoid treatment, e American
Journal of Physiology—Endocrinology and Metabolism,vol.,
no. , pp. E–E, .
[] A. Gottlieb, A. Menter, A. Mendelsohn et al., “Ustekinumab,
a human interleukin / monoclonal antibody, for psoriatic
arthritis: randomised, double-blind, placebo-controlled, cross-
over trial, e Lancet,vol.,no.,pp.,.
[] K. M. Dho dapkar, S. Barbuto, P. Matthews et al., “Dendritic cells
mediate the induction of polyfunctional human IL-producing
cells (- cells) enriched in the bone marrow of patients with
myeloma, Blood,vol.,no.,pp.,.
[] K. Noonan, L. Marchionni, J. Anderson, D. Pardoll, G. D.
Roodman,andI.Borrello,“AnovelroleofIL--producinglym-
phocytes in mediating lytic bone disease in multiple myeloma,
Blood,vol.,no.,pp.,.
[] R.H.Prabhala,D.Pelluru,M.Fulcinitietal.,“ElevatedIL-
produced by TH cells promotes myeloma cell growth and
inhibits immune function in multiple myeloma,” Blood,vol.,
no.,pp.,.
[] P.Davari,M.S.Leo,F.Kamangaretal.,“Ustekinumabforthe
treatment of psoriatic arthritis: an update,” Journal of Clinical,
Cosmetic and Investigational Dermatology,vol.,pp.,
.
[]E.Suzuki,V.Kapoor,A.S.Jassar,L.R.Kaiser,andS.M.
Albelda, “Gemcitabine selectively eliminates splenic Gr-+/
CDb+myeloid suppressor cells in tumor-bearing animals and
enhances antitumor immune activity, Clinical Cancer Research,
vol. , no. , pp. –, .
[] H.-J. Ko, Y.-J. Kim, Y.-S. Kim et al., A combination of chemoim-
munotherapies can eciently break self-tolerance and induce
antitumor immunity in a tolerogenic murine tumor model,
Cancer Research,vol.,no.,pp.,.
[] P. J. Mease, M. C. Genovese, M. W. Greenwald et al., “Bro-
dalumab, an anti-ILRA monoclonal antibody, in psoriatic
arthritis, e New England Journal of Medicine,vol.,no.,
pp. –, .
[] S. Colucci, G. Brunetti, F. P. Cantatore et al., “Lymphocytes and
synovial uid broblasts support osteoclastogenesis through
RANKL, TNF𝛼, and IL- in an in vitro model derived from
human psoriatic arthritis, Journal of Pathology,vol.,no.,
pp.,.
[] C. Infante-Duarte, H. F. Horton, M. C. Byrne, and T. Kamradt,
“Microbial lipopeptides induce the production of IL- in
cells, e Journal of Immunology,vol.,no.,pp.,
.
[] I. I. Ivanov, B. S. McKenzie, L. Zhou et al., “e orphan
nuclear recepto ROR𝛾t directs the dierentiation program of
pro-inammatory IL-+T helper cells, Cell,vol.,no.,pp.
–, .
[] A. Batycka-Baran, J. Maj, R. Wolf, and J. C. Szepietowski, “e
new insight into the role of antimicrobial proteins-alarmins in
the immunopathogenesis of psoriasis, Journal of Immunology
Research,vol.,ArticleID,pages,.
[] O. FitzGerald and R. Winchester, “Psoriatic arthritis: from
pathogenesis to therapy, Arthritis Research and erapy,vol.,
no. , article , .
[] L. R. Espinoza, J. L. Aguilar, C. G. Espinoza, M. L. Cuellar, E.
Scopelitis, and L. H. Silveira, “Fibroblast function in psoriatic
arthritis. I. Alteration of cell kinetics and growth factor respon
ses, e Journal of Rheumatology,vol.,no.,pp.,
.
[] L. R. Espinoza, C. G. Espinoza, M. L. Cuellar, E. Scopelitis, L. H.
Silveira, and G. R. Grotendorst, “Fibroblast function in psoriatic
arthritis. II. Increased expression of 𝛽platelet derived growth
factor receptors and increased production of growth factor and
cytokines, e Journal of Rheumatology,vol.,no.,pp.
, .
[] D. D. Gladman, “Toward unraveling the mystery of psoriatic
arthritis, Arthritis and Rheumatism, vol. , no. , pp. –,
.
[] B. Menon, N. J. Gullick, G. J. Walter et al., “Interleukin-
+CD+ T cells are enriched in the joints of patients with
psoriatic arthritis and correlate with disease activity and joint
damage progression, Arthritis and Rheumatology,vol.,no.
, pp. –, .
[] “Osteoporosis. National Institutes of Health Consensus Devel-
opment Conference Statement,” National Institutes of Health
Consensus Development Conference Statement,vol.,no.,p.
, .
[] P. DAmelio, A. Grimaldi, S. di Bella et al., “Estrogen deciency
increases osteoclastogenesis up-regulating T cells activity: a key
mechanism in osteoporosis, Bone,vol.,no.,pp.,
.
[] P. D’Amelio, M. A. Cristofaro, A. Grimaldi et al., “e role of
circulating bone cell precursors in fracture healing, Calcied
Tissue International,vol.,no.,pp.,.
[] P. D’Amelio, A. Grimaldi, M. A. Cristofaro et al., Alendronate
reduces osteoclast precursors in osteoporosis, Osteoporosis
International,vol.,no.,pp.,.
[] L. D’amico, M. A. Satolli, C. Mecca et al., “Bone metastases
in gastric cancer follow a RANKL-independent mechanism,
Oncology Reports,vol.,no.,pp.,.
[] I. Roato, P. D’Amelio, E. Gorassini et al., “Osteoclasts are active
in bone forming metastases of prostate cancer patients, PLoS
ONE,vol.,no.,ArticleIDe,.
[] A. E. Oostlander, V. Everts, T. Schoenmaker et al., “T cell-
mediated increased osteoclast formation from peripheral blood
as a mechanism for crohn’s disease-associated bone loss,
Journal of Cellular Biochemistry,vol.,no.,pp.,
.
[] M.Reed,R.J.Baker,A.B.Mehta,andD.A.Hughes,“Enhanced
dierentiation of osteoclasts from mononuclear precursors
Journal of Immunology Research
in patients with Gaucher disease, Blood Cells, Molecules, &
Diseases,vol.,no.,pp.,.
[] R. Pacici, “e immune system and bone, Archives of Bio-
chemistry and Biophysics,vol.,no.,pp.,.
[] I. Roato, E. Gorassini, G. Brunetti et al., “IL- modulates
osteoclastogenesis in patients aected by solid tumors, Annals
of the New York Academy of Sciences, vol. , pp. –, .
[] A. E. Oostlander, V. Everts, T. Schoenmaker et al., T cell-
mediated increased osteoclast formation from peripheral blood
as a mechanism for Crohn’s disease-associated bone loss,
Journal of Cellular Biochemistry, vol. , no. , pp. –,
.
[] Y. Xue, L. Jiang, Q. Cheng et al., Adipokines in psoriatic arth-
ritis patients: the correlations with osteoclast precursors and
bone erosions, PLoS ONE,vol.,no.,ArticleIDe,.
[] P.DAmelio,I.Roato,L.Damicoetal.,“Boneandbonemarrow
pro-osteoclastogenic cytokines are up-regulated in osteoporosis
fragility fractures, Osteoporosis International,vol.,no.,pp.
–, .
[] N. Udagawa, N. Takahashi, E. Jimi et al., “Osteoblasts/stromal
cells stimulate osteoclast activation through expression of
osteoclast dierentiation factor/RANKL but not macrophage
colony-stimulating factor, Bone,vol.,no.,pp.,.
[] M.Onal,J.Xiong,X.Chenetal.,“Receptoractivatorofnuclear
factor 𝜅B ligand (RANKL) protein expression by B lymphocytes
contributes to ovariectomy-induced bone loss, e Journal of
Biological Chemistry,vol.,no.,pp.,.
[] L. C. Hoauer, S. Khosla, C. R. Dunstan, D. L. Lacey, W. J.
Boyle, and B. L. Riggs, “e roles of osteoprotegerin and osteo-
protegerin ligand in the paracrine regulation of bone resorp-
tion, Journal of Bone and Mineral Research,vol.,no.,pp.
–, .
[] Y. Y. Kong, H. Yoshida, I. Sarosi et al., “OPGL is a key regulator
of osteoclastogenesis, lymphocyte development and lymph-
node organogenesis, Nature, vol. , no. , pp. –,
.
[] H. Hotokezaka, E. Sakai, N. Ohara et al., “Molecular analysis of
RANKL-independent cell fusion of osteoclast-like cells induced
by TNF-alpha, lipopolysaccharide, or peptidoglycan, Journal of
Cellular Biochemistry,vol.,no.,pp.,.
[]S.Wei,H.Kitaura,P.Zhou,F.P.Ross,andS.L.Teitelbaum,
“IL- mediates TNF-induced osteoclastogenesis, e Journal of
Clinical Investigation,vol.,no.,pp.,.
[]N.Charatcharoenwitthaya,S.Khosla,E.J.Atkinson,L.K.
McCready, and B. L. Riggs, “Eect of blockade of TNF-𝛼
and interleukin- action on bone resorption in early post-
menopausal women, Journal of Bone and Mineral Research,vol.
,no.,pp.,.
[] N.Giuliani,S.Colla,R.Salaetal.,“Humanmyelomacellsstimu-
late the receptor activator of nuclear factor-𝜅B ligand (RANKL)
in T lymphocytes: a potential role in multiple myeloma bone
disease, Blood,vol.,no.,pp.,.
[]G.Toraldo,C.Roggia,W.-P.Qian,R.Pacic,andM.N.
Weitzmann, “IL- induces bone loss in vivo by induction
of receptor activator of nuclear factor 𝜅B ligand and tumor
necrosis factor 𝛼from T cells,” Proceedings of the National
Academy of Sciences of the United States of America,vol.,no.
,pp.,.
[] M. R. Ryan, R. Shepherd, J. K. Leavey et al., An IL--dependent
rebound in thymic T cell output contributes to the bone loss
induced by estrogen deciency, Proceedings of the National
Academy of Sciences of the United States of America,vol.,no.
, pp. –, .
[] A. M. Tyagi, K. Srivastava, M. N. Mansoori, R. Trivedi, N.
Chattopadhyay, and D. Singh, “Estrogen deciency induces the
dierentiation of IL- secreting  cells: a new candidate in
the pathogenesis of osteoporosis, PloS ONE,vol.,no.,Article
ID e, .
[] R. Zhao, “Immune regulation of bone loss by  cells in
oestrogen-decient osteoporosis, European Journal of Clinical
Investigation,vol.,no.,pp.,.
[] L. Das Roy, L. B. Pathangey, T. L. Tinder, J. L. Schettini, H. E.
Gruber, and P. Mukherjee, “Breast cancer-associated metastasis
is signicantly increased in a model of autoimmune arthritis,
Breast Cancer Research, vol. , no. , article R, .
[] H. Forsblad d’Elia and H. Carlsten, e impact of hormone
replacement therapy on humoral and cell-mediated immune
responses in vivo in post-menopausal women with rheumatoid
arthritis, Scandinavian Journal of Immunology,vol.,no.,
pp.,.
[] S. Kumru, A. Godekmerdan, and B. Yilmaz, “Immune eects of
surgical menopause and estrogen replacement therapy in peri-
menopausal women, Journal of Reproductive Immunology,vol.
,no.,pp.,.
[] A. Anginot, R. Dacquin, M. Mazzorana, and P. Jurdic, “Lym-
phocytes and the Dap adaptor are key regulators of osteoclast
activation associated with gonadal failure,” PLoS ONE,vol.,no.
, Article ID e, .
[] S.-K. Lee, Y. Kadono, F. Okada et al., “T lymphocyte-decient
mice lose trabecular bone mass with ovariectomy, Journal of
Bone and Mineral Research, vol. , no. , pp. –, .
[] S. Cenci, G. Toraldo, M. N. Weitzmann et al., “Estrogen de-
ciency induces bone loss by increasing T cell proliferation and
lifespan through IFN-𝛾-induced class II transactivator, Pro-
ceedings of the National Academy of Sciences of the United States
of America, vol. , no. , pp. –, .
[] G.Eghbali-Fatourechi,S.Khosla,A.Sanyal,W.J.Boyle,D.L.
Lacey, and B. L. Riggs, “Role of RANK ligand in mediating
increased bone resorption in early postmenopausal women,
e Journal of Clinical Investigation, vol. , no. , pp. –,
.
[] Y.Gao,F.Grassi,M.R.Ryanetal.,“IFN-𝛾stimulates osteoclast
formation and bone loss in vivo via antigen-driven T cell
activation, e Journal of Clinical Investigation,vol.,no.,
pp.,.
[] Y.Li,G.Toraldo,A.Lietal.,“BcellsandTcellsarecriticalfor
the preservation of bone homeostasis and attainment of peak
bone mass in vivo, Blood,vol.,no.,pp.,.
[]J.Adamski,Z.Ma,S.Nozell,andE.N.Benveniste,“beta-
estradiol inhibits class ii major histocompatibility complex
(MHC) expression: inuence on histonemo dications and CBP
recruitment to the class II MHC promoter,” Molecular Endo-
crinology,vol.,no.,pp.,.
[] J. H. Kim, K. Kim, B. U. Youn, H. M. Jin, and N. Kim,
“MHC class II transactivator negatively regulates RANKL-
mediated osteoclast dierentiation by downregulating NFATc
and OSCAR, Cellular Signalling,vol.,no.,pp.,
.
[] R. B. Mueller, A. Skapenko, M. Grunke et al., “Regulation of
myeloid cell function and major histocompatibility complex
class II expression by tumor necrosis factor, Arthritis and
Rheumatism,vol.,no.,pp.,.
 Journal of Immunology Research
[] J.-Y. Li, H. Tawfeek, B. Bedi et al., Ovariectomy disregulates
osteoblast and osteoclast formation through the T-cell receptor
CD ligand, Proceedings of the National Academy of Sciences
of the United States of America,vol.,no.,pp.,.
[] M. Yokoyama, T. Ukai, E. R. Ayon Haro, T. Kishimoto, Y.
Yoshinaga, and Y. Hara, “Membrane-bound CD ligand on
T cells from mice injected with lipopolysaccharide accelerates
lipopolysaccharide-induced osteoclastogenesis, Journal of Peri-
odontal Research, vol. , no. , pp. –, .
[] B.Bedi,J.-Y.Li,F.Grassi,H.Tawfeek,M.N.Weitzmann,and
R. Pacici, “Inhibition of antigen presentation and T cell costi-
mulation blocks PTH-induced bone loss, Annals of the New
York Academy of Sciences,vol.,pp.,.
[]Y.Gao,X.Wu,M.Terauchietal.,“TcellspotentiatePTH-
induced cortical bone loss through CDL signaling, Cell
Metabolism,vol.,no.,pp.,.
[] D. Grˇ
cevi´
c, I. K. Luki´
c, N. Kovaˇ
ci´
c, S. Ivˇ
cevi´
c, V. Katavi´
c, and A.
Maruˇ
si´
c, Activated T lymphocytes suppress osteoclastogenesis
by diverting early monocyte/macrophage progenitor lineage
commitment towards dendritic cell dierentiation through
down-regulation of receptor activator of nuclear factor-kappaB
and c-Fos, Clinical and Experimental Immunology,vol.,no.
, pp. –, .
[] N. Wyzga, S. Varghese, S. Wikel, E. Canalis, and F. A. Sylvester,
“Eects of activated T cells on osteoclastogenesis depend on
how they are activated, Bone,vol.,no.,pp.,.
[] Y.Y.Kong,U.Feige,I.Sarosietal.,“ActivatedTcellsregulate
bone loss and joint destruction in adjuvant arthritis through
osteoprotegerin ligand,” Nature,vol.,pp.,.
[] I. Roato, D. Caldo, L. DAmico et al., “Osteoclastogenesis in
peripheral blood mononuclear cell cultures of periprosthetic
osteolysis patients and the phenotype of T cells localized in
periprosthetic tissues, Biomaterials,vol.,no.,pp.
, .
[] N. Giuliani, F. Morandi, S. Tagliaferri et al., “Interleukin- (IL-
) is overexpressed by T lymphocytes in multiple myeloma
patients, Blood,vol.,no.,pp.,.
[] X. Han, X. Lin, A. R. Seliger, J. Eastcott, T. Kawai, and M. A.
Taubman, “Expression of receptor activator of nuclear factor-
kappaB ligand by B cells in response to oral bacteria, Oral
Microbiology and Immunology,vol.,no.,pp.,.
[] L. Yeo, K.-M. Toellner, M. Salmon et al., Cytokine mRNA pro-
ling identies B cells as a major source of RANKL in rheuma-
toid arthritis, Annals of the Rheumatic Diseases,vol.,no.,
pp. –, .
[] M. Kanematsu, T. Sato, H. Takai, K. Watanabe, K. Ikeda, and
Y. Yamada, “Prostaglandin E- induces expression of receptor
activator of nuclear factor-B ligand/osteoprotegrin ligand on
pre-B cells: implications for accelerated osteoclastogenesis in
estrogen deciency, Journal of Bone and Mineral Research,vol.
,no.,pp.,.
[] X. Han, X. Lin, X. Yu et al., Porphyromonas gingivalis infection-
associated periodontal bone resorption is dependent on recep-
tor activator of NF-𝜅B ligand, Infection and Immunity,vol.,
no. , pp. –, .
[] G. Wheater, V. E. Hogan, Y. K. O. Teng et al., “Suppression of
bone turnover by B-cell depletion in patients with rheumatoid
arthritis, Osteoporosis International,vol.,no.,pp.
, .
[] G. Mazziotti, A. Angeli, J. P. Bilezikian, E. Canalis, and A.
Giustina, “Glucocorticoid-induced osteoporosis: an update,
Trends in Endocrinology and Metabolism,vol.,no.,pp.
, .
[] R. S. Weinstein, “Clinical practice. Glucocorticoid-induced
bone disease, e New England Journal of Medicine,vol.,
pp.,.
[] S. Ikeda, Y. Morishita, H. Tsutsumi et al., “Reductions in bone
turnover, mineral, and structure associated with mechanical
properties of lumbar vertebra and femur in glucocorticoid-
treated growing minipigs, Bone,vol.,no.,pp.,
.
[] M. F. Faienza, G. Brunetti, S. Colucci et al., Osteoclastogen-
esis in children with -hydroxylase deciency on long-term
glucocorticoid therapy: the role of receptor activator of nuclear
factor-𝜅B ligand/osteoprotegerin imbalance, Journal of Clinical
Endocrinology and Metabolism,vol.,no.,pp.,
.
[] G. D. Roodman, “Pathogenesis of myeloma bone disease, Leu-
kemia,vol.,no.,pp.,.
[] S.Colucci,G.Brunetti,A.Orangeretal.,“Myelomacellssup-
press osteoblasts through sclerostin secretion, Blood Cancer
Journal,vol.,no.,articlee,.
[] A.Oranger,G.Brunetti,C.Carboneetal.,“Humanmyeloma
cell lines induce osteoblast downregulation of CD which is
involved in osteoblast formation and activity, Journal of Immu-
nology Research.Inpress.
[] G. Brunetti, S. Colucci, R. Rizzi et al., “e role of OPG/TRAIL
complex in multiple myeloma,”Annals of the New York Academy
of Sciences,vol.,no.,pp.,.
[] S. Colucci, G. Brunetti, G. Mori et al., “Soluble decoy receptor
modulates the survival and formation of osteoclasts from
multiple myeloma bone disease patients,” Leukemia,vol.,no.
, pp. –, .
[] C.-J. Shen, Z.-H. Yuan, Y.-X. Liu, and G.-Y. Hu, Increased
numbers of T helper  cells and the correlation with clini-
copathological characteristics in multiple myeloma, Journal of
International Medical Research,vol.,no.,pp.,.
[] J.R.Edwards,S.G.Sun,R.Locklinetal.,“LIGHT(TNFSF),a
novel mediator of bone resorption, is elevated in rheumatoid
arthritis, Arthritis and Rheumatism,vol.,no.,pp.
, .
[]G.R.Mundy,“Metastasistobone:causes,consequencesand
therapeutic opportunities, Nature Reviews Cancer,vol.,no.
, pp. –, .
[] K. N. Weilbaecher, T. A. Guise, and L. K. McCauley, “Cancer to
bone: a fatal attraction, Nature Reviews Cancer, vol. , no. ,
pp. –, .
[]M.Clemons,K.A.Gelmon,K.I.Pritchard,andA.H.G.
Paterson, “Bone-targeted agents and skeletal-related events in
breast cancer patients with bone metastases: the state of the art,
Current Oncology,vol.,no.,pp.,.
[] S.-M. K¨
ak¨
onen, K. S. Selander, J. M. Chirgwin et al., “Trans-
forming growth factor-𝛽stimulates parathyroid hormone-
related protein and osteolytic metastases via Smad and
mitogen-activated protein kinase signaling pathways, e Jour-
nalofBiologicalChemistry,vol.,no.,pp.,
.
[] A. Valachis, N. P. Polyzos, R. E. Coleman et al., Adjuvant ther-
apy with zoledronic acid in patients with breast cancer: a sys-
tematic review and meta-analysis, e Oncologist,vol.,no.,
pp.,.
Journal of Immunology Research 
[] M. He, W. Fan, and X. Zhang, Adjuvant zoledronic acid therapy
for patients with early stage breast cancer: an updated sys-
tematic review and meta-analysis, Journal of Hematology and
Oncology, vol. , no. , article , .
[] L. S. Rosen, D. Gordon, M. Kaminski et al., “Zoledronic acid
versus pamidronate in the treatment of skeletal metastases in
patients with breast cancer or osteolytic lesions of multiple
myeloma: a phase III, double-blind, comparative trial, Cancer
Journal,vol.,no.,pp.,.
[] L. S. Rosen, D. Gordon, S. Tchekmedyian et al., “Zoledronic acid
versus placebo in the treatment of skeletal metastases in patients
with lung cancer and other solid tumors: a phase III, double-
blind, randomized trial—the zoledronic acid lung cancer and
other solid tumors study group, Journal of Clinical Oncology,
vol.,no.,pp.,.
[] A. T. Stopeck, A. Lipton, J.-J. Body et al., “Denosumab compared
with zoledronic acid for the treatment of bone metastases in
patients with advanced breast cancer: a randomized, double-
blind study, Journal of Clinical Oncology,vol.,no.,pp.
–, .
[] X. H.-F. Zhang, X. Jin, S. Malladi et al., “Selection of bone meta-
stasis seeds by mesenchymal signals in the primary tumor
stroma, Cell,vol.,no.,pp.,.
[] M. Feuerer, M. Rocha, L. Bai et al., “Enrichment of memory T
cells and other profound immunological changes in the bone
marrow from untreated breast cancer patients, International
Journal of Cancer,vol.,no.,pp.,.
[] J.F.Bromberg,C.M.Horvath,Z.Wen,R.D.Schreiber,andJ.
E. Darnell Jr., Transcriptionally active Stat is required for the
antiproliferative eects of both interferon 𝛼and interferon 𝛾,”
Proceedings of the National Academy of Sciences of the United
States of America, vol. , no. , pp. –, .
[] S. Kumar, “e Bcl- family of proteins and activation of the
ICE-CED- family of proteases: a balancing act in apoptosis?”
Cell Death and Dierentiation,vol.,no.,pp.,.
[] Z. Qin and T. Blankenstein, “CD+T cell—mediated tumor
rejection involves inhibition of angiogenesis that is dependent
on IFN gamma receptor expression by nonhematopoietic cells,
Immunity,vol.,no.,pp.,.
[] B.N.Bidwell,C.Y.Slaney,N.P.Withanaetal.,“Silencingof
Irf pathways in breast cancer cells promotes bone metastasis
through immune escape, Nature Medicine,vol.,no.,pp.
–, .
[] L. D’Amico and I. Roato, “e impact of immune system in
regulating bone metastasis formation by osteotropic tumors,
Journal of Immunology Research.Inpress.
[] M. J. Smyth, G. P. Dunn, and R. D. Schreiber, Cancer immuno-
surveillance and immunoediting: the roles of immunity in
suppressing tumor development and shaping tumor immuno-
genicity,” Advances in Immunology,vol.,pp.,.
[] D. G. DeNardo and L. M. Coussens, “Inammation and breast
cancer.B alancingimmune response: crosstalk between adaptive
and innate immune cells during breast cancer progression,
Breast Cancer Research,vol.,no.,article,.
[] A.C.Monteiro,A.C.Leal,T.Gonc¸alves-Silva et al., “T cells
induce pre-metastatic osteolytic disease and help bone meta-
stases establishment in a mouse model of metastatic breast can-
cer, PLoS ONE,vol.,no.,ArticleIDe,.
[] J. Sceneay, M. T. Chow, A. Chen et al., “Primary tumor hypoxia
recruits CDb+/LyC𝑚𝑒𝑑/LyG
+immune suppressor cells and
compromises NK cell cytotoxicity in the premetastatic niche,
Cancer Research,vol.,no.,pp.,.
[] B. Almand, J. I. Clark, E. Nikitina et al., “Increased production
of immature myeloid cells in cancer patients: a mechanism of
immunosuppression in cancer, e Journal of Immunology,vol.
, no. , pp. –, .
[] I. Marigo, L. Dolcetti, P. Serani, P. Zanovello, and V.
Bronte, “Tumor-induced tolerance and immune suppression by
myeloid derived suppressor cells, Immunological Reviews,vol.
,no.,pp.,.
[] S. Kusmartsev and D. I. Gabrilovich, “Inhibition of myeloid cell
dierentiation in cancer: the role of reactive oxygen species,
Journal of Leukocyte Biology,vol.,no.,pp.,.
[] P. Liu, J. R. Keller, M. Ortiz et al., “Bcla is essential for normal
lymphoid development, Nature Immunology,vol.,no.,pp.
–, .
[] A. Mazzoni, V. Bronte, A. Visintin et al., Myeloid suppressor
lines inhibit T cell responses by an NO-dependent mechanism,
e Journal of Immunology,vol.,no.,pp.,.
[] S. Kusmartsev, Y. Nefedova, D. Yoder, and D. I. Gabrilovich,
Antigen-specic inhibition of CD+T cell response by imma-
ture myeloid cells in cancer is mediated by reactive oxygen
species, e Journal of Immunology,vol.,no.,pp.,
.
[] S. Kusmartsev and D. I. Gabrilovich, “Eect of tumor-derived
cytokines and growth factors on dierentiation and immune
suppressive features of myeloid cells in cancer, Cancer and
Metastasis Reviews,vol.,no.,pp.,.
[] A.-H. Capietto, S. Kim, D. E. Sanford et al., “Down-regulation
of PLC𝛾-𝛽-catenin pathway promotes activation and expan-
sion of myeloid-derived suppressor cells in cancer, Journal of
Experimental Medicine,vol.,no.,pp.,.
[] D. Gao, N. Joshi, H. Choi et al., “Myeloid progenitor cells in
the premetastatic lung promote metastases by inducing mesen-
chymal to epithelial transition, Cancer Research,vol.,no.,
pp.,.
[] M. L. Ortiz, L. Lu, I. Ramachandran, and D. I. Gabrilovich,
“Myeloid-derived suppressor cells in the development of lung
cancer, Cancer Immunology Research,vol.,no.,pp.,
.
[] I.R.Ramachandran,A.Martner,A.Pisklakovaetal.,“Myeloid-
derived suppressor cells regulate growth of multiple myeloma
by inhibiting T cells in bone marrow,” e Journal of Immunol-
ogy,vol.,no.,pp.,.
[] J.Zhuang,J.Zhang,S.T.Lwinetal.,“Osteoclastsinmultiple
myeloma are derived from Gr-+CDb+myeloid-derived sup-
pressor cells, PLoS ONE,vol.,no.,ArticleIDe,.
[] A. Sawant, J. Deshane, J. Jules et al., “Myeloid-derived suppres-
sor cells function as novel osteoclast progenitors enhancing
bone loss in breast cancer, Cancer Research,vol.,no.,pp.
–, .
... These studies have provided us with novel insights and possible directions for discovery of early diagnostic markers for bone pathologies as well as targeting specific cargo for therapeutic treatment [97]. In parallel, there have been studies for better physiological understanding of the communication of bone cells with microenvironment cells such immune cells, endothelial cells, and stromal cells in influencing bone homeostasis [98,99]. In the coming sections, we will begin with describing exosomes derived from various resident bone cells followed by Biomedicines 2022, 10, 767 5 of 29 exosomes secreted by progenitor and microenvironment cells. ...
... We will also describe how tumor-derived exosomes interact with the bone cells to influence tumor progression. [98,99]. In the coming sections, we will begin with describing exosomes derived from various resident bone cells followed by exosomes secreted by progenitor and microenvironment cells. ...
... Thus, MSC-derived exosomes have been demonstrated to both promote [85][86][87][88][89][90][91][92][93][94][95][96][97] as well as inhibit osteogenic differentiation [91,[98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117] through modulation of microRNA profile as listed in Table 3. Table 3. MSC-derived exosome miRNA promoting and inhibiting osteogenic differentiation. ...
Article
Full-text available
Bone tissue remodeling is a highly regulated process balancing bone formation and resorption through complex cellular crosstalk between resident bone and microenvironment cells. This cellular communication is mediated by direct cell and cell–matrix contact, autocrine, endocrine, and paracrine receptor mediated mechanisms such as local soluble signaling molecules and extracellular vesicles including nanometer sized exosomes. An impairment in this balanced process leads to development of pathological conditions. Bone tissue engineering is an emerging interdisciplinary field with potential to address bone defects and disorders by synthesizing three-dimensional bone substitutes embedded with cells for clinical implantation. However, current cell-based therapeutic approaches have faced hurdles due to safety and ethical concerns, challenging their clinical translation. Recent studies on exosome-regulated bone homeostasis and regeneration have gained interest as prospective cell free therapy in conjugation with tissue engineered bone grafts. However, exosome research is still in its nascent stages of bone tissue engineering. In this review, we specifically describe the role of exosomes secreted by cells within bone microenvironment such as osteoblasts, osteocytes, osteoclasts, mesenchymal stem cell cells, immune cells, endothelial cells, and even tumor cells during bone homeostasis and crosstalk. We also review exosome-based osteoinductive functionalization strategies for various bone-based biomaterials such as ceramics, polymers, and metals in bone tissue engineering. We further highlight biomaterials as carrier agents for exosome delivery to bone defect sites and, finally, the influence of various biomaterials in modulation of cell exosome secretome.
... In OS, autophagy is strongly described as a tumor survival and cytoprotective mechanism to provide tumor cells energy under stress conditions and also decrease drug sensitivity [138]. Indeed, studies linked to HSP90 and its family member HSP90AA1 described autophagy in OS as a cytoprotective process and a process involved into drugs resistance [139][140] [141]. Indeed, inhibition of autophagy by 3-methyladeine (3-MA) treatment decreases proliferation rate and promotes apoptosis, reversing the autophagyinduced-cytoprotection due to low level of HSP90 (geldanamycin) or high level of HSP90AA1 in OS cells [139]. ...
... Indeed, studies linked to HSP90 and its family member HSP90AA1 described autophagy in OS as a cytoprotective process and a process involved into drugs resistance [139][140] [141]. Indeed, inhibition of autophagy by 3-methyladeine (3-MA) treatment decreases proliferation rate and promotes apoptosis, reversing the autophagyinduced-cytoprotection due to low level of HSP90 (geldanamycin) or high level of HSP90AA1 in OS cells [139]. They further show that HSP90AA1 induces autophagy by impairing PI3K/AKT/mTOR pathway [141]. ...
... In onco-hematology, GCs are used in chemotherapy regimens and as analgesics and antiemetics, and in the long-term can cause iatrogenic osteoporosis [64]. GCs damage trabecular bone, causing rapid BMD loss because of bone resorption and delayed demineralization as a result of reduced OB activity [65]. Patients with primary and secondary brain tumors require ongoing GC therapy to ameliorate neurological sequelae and peritumoral edema [66]. ...
Article
Cancer treatment-induced bone loss (CTBL) is associated with anti-tumor treatments, including endocrine therapies, chemotherapeutic treatments, radiotherapy, glucocorticoids, and tyrosine kinase inhibitors. Osteoporosis, characterized by the loss of bone mass, can increase the risk of fractures, leading to mortality and long-term disability, even after cancer remission. Cancer and osteoporosis have marked clinical and pathogenetic similarities. Both have a multifactorial etiology, affect the geriatric population, and markedly influence quality of life. Lifestyle management, including calcium and vitamin D supplementation, is recommended but the supporting evidence is limited. Oral and injectable bisphosphonates are effective for osteoporosis and malignant bone disease. Bisphosphonates increase bone mineral density (BMD) in patients with CTBL. Denosumab is also used in the management of CTBL; in clinical trials, it improved BMD and reduced the risk of fracture. Currently, there are no bone anabolic therapies for patients with cancer. Appropriate therapies are necessary to maintain optimal bone health, particularly in patients at heightened risk.
... Bone is the third most common site of metastasis for a variety of solid tumors including lung, breast, prostate, colorectal, melanoma, and others, with about 70% of patients with metastatic prostate and breast cancers harboring bone metastasis (1,2). In addition to leading to significant morbidity and mortality, bone metastases lack effective therapeutic approaches, and the most successful treatments for these patients are aimed at reducing skeletal-related events (SRE), such as fractures, bone pain, hypercalcemia, and spinal cord compression (3)(4)(5)(6). ...
Article
Full-text available
Osteoclast (OC) blockade has been successful in reducing tumor growth in bone in pre-clinical settings, but anti-resorptive drugs, such as zoledronic acid (ZA), fail to improve the overall survival rate of patients with bone metastasis, despite ameliorating skeletal complications. To address this unmet clinical need, we interrogated what other cells modulated tumor growth in bone in addition to OCs. Because myeloid-derived suppressor cells (MDSCs), heterogeneous populations expressing CD11b, Ly6C, and Ly6G markers, originate in the bone marrow and promote tumor progression, we hypothesized that their accumulation hinders ZA anti-tumor effects. By using a murine model of bone metastasis insensitive to OC blockade, we assessed the anti-tumor effect of MDSC depletion using anti-Gr1 in mice bearing skeletal lung (LLC), melanoma (B16-F10), and mammary (4T1) tumors. Differently from soft tissue tumors, anti-Gr1 did not reduce bone metastases and led to the paradoxical accumulation of bone marrow-resident CD11b+Ly6CintLy6Gint cells that differentiated into OCs when cultured in vitro. Anti-Gr1-mediated depletion of Ly6G+ granulocytic MDSCs combined with ZA-induced OC blockade reduced growth of established skeletal metastases compared to each agent alone. CD15+ granulocytic populations were increased in breast cancer patients with progressive bone disease after anti-resorptive treatment compared to those with stable bone disease. We provide evidence that anti-resorptive therapies fail to reduce bone metastases in the presence of elevated granulocytic populations and that effective treatment of established skeletal metastases requires combinatorial depletion of granulocytes and OC blockade.
Article
Full-text available
The recognition and importance of immune cells during bone regeneration, including around bone biomaterials, has led to the development of an entire field termed “osteoimmunology,” which focuses on the connection and interplay between the skeletal system and immune cells. Most studies have focused on the “osteogenic” capacity of various types of bone biomaterials, and much less focus has been placed on immune cells despite being the first cell type in contact with implantable devices. Thus, the amount of literature generated to date on this topic makes it challenging to extract needed information. This review article serves as a guide highlighting advancements made in the field of osteoimmunology emphasizing the role of the osteoimmunomodulatory properties of biomaterials and their impact on osteoinduction. First, the various immune cell types involved in bone biomaterial integration are discussed, including the prominent role of osteal macrophages (OsteoMacs) during bone regeneration. Thereafter, key biomaterial properties, including topography, wettability, surface charge, and adsorption of cytokines, growth factors, ions, and other bioactive molecules, are discussed in terms of their impact on immune responses. These findings highlight and recognize the importance of the immune system and osteoimmunology, leading to a shift in the traditional models used to understand and evaluate biomaterials for bone regeneration.
Article
Aims Glucocorticoids have adverse side effects that can trigger osteoporosis. There is evidence to show that symbiotics and curcumin can prevent bone loss. The aim of this study was to analyze the effects of curcumin and symbiotic to prevent and/or minimize bone damage in rats submitted to osteoporosis induction using dexamethasone. Methods Fifty Wistar female rats were divided into five groups: control group (CT), osteoporosis control group (CO), osteoporosis and symbiotic group (SO), osteoporosis and curcumin group (CCMO), and osteoporosis and symbiotic/curcumin group (SCO). Dexamethasone was applied three times a week, while the symbiotic and curcumin were administered daily. Alkaline phosphatase and calcium dosages, analysis of structural and material properties, and Raman analysis of femurs were performed. Key findings Alkaline phosphatase was higher in the CCMO group. Maximum load and structural stiffness were higher in the CT group. Maximum stress was lower and similar between osteoporosis groups. The CT group had a lower percentage of strain, the CO group had greater deformation compared to the CCMO group and the SO group presented more deformation than the CCMO group. The CO, SO, and SCO groups had a lower elastic modulus compared to the CT group. The 960/1660 ratios of the CO, SO, and SCO groups were different from the CT group. The 1070/1660 ratio was higher in the CCMO group. Significance It was possible to verify that curcumin showed promising effects related to the increase in bone strength and mineralization through the induction of osteoporosis by dexamethasone.
Chapter
It has long been recognized that cancer cells exhibit altered patterns of metabolism that allow them to meet the biosynthetic demands of rapid proliferation and overcome metabolic stress imposed by the microenvironment. In this chapter, we aim to recapitulate knowledge on energetic metabolism and reprogramming in osteo-oncology. Diagnosis and staging of bone metastases using ¹⁸Fluorine labeled glucose, choline, and folate and emission tomography and computerized tomography (PET/CT) reflect tumor cells energetic metabolism. Metabolites and associated gene regulators involved in bone metastases and/or osteosarcoma progression are also described, including glucose, lactate, amino acid, cholesterol, fatty acids, and derived mitochondrial activity reactive oxygen species. Moreover several master regulators of energy metabolism in normal physiology, metabolic syndromes, and cancer (PPARγ, ERRα, PGC1α, CREB, MYC, RAS, EWS-FLI, TMPRSS2-ERG, and mTOR) are identified in bone metastases and primary bone tumor. Interestingly, factors that were first detected in bone physiology (RANKL/OPG/RANK, Runx2, OPN, OCN, POSTN, and SOST) have been recently identified as regulators of energetic metabolism. Finally, in clinic, treatments targeting metabolism (everolimus, statin, etc.) have an impact on bone metastases regression, suggesting that targeting energetic metabolism may constitute a new therapeutic pathway in osteo-oncology.
Chapter
Full-text available
La pandemia por el Covid 19, ha puesto en una situación difícil a toda la humanidad. Los deportistas no son la excepción, sufriendo inconvenientes con su entrenamiento o con su vuelta a las actividades deportivas después de padecer la infección. Adicionalmente, surgen varias publicaciones que hablan de la posible asociación entre la actividad física y la prevención de infecciones virales, entre las cuales se encuentra el Coronavirus; sin embargo, la mayoría son opiniones de expertos por lo cual, se requiere hacer una revisión sensata de la literatura científica al respecto. Existe evidencia científica que demuestra que, un porcentaje importante de las personas que sufren la infección por COVID -19, quedan con alteraciones funcionales. Entre el 64% y el 69%; con fatiga crónica, el 22% con una baja capacidad para realizar ejercicio y el 72% con alteraciones en la calidad de vida. Por lo tanto, es pertinente que el reacondicionamiento físico y por ende, el regreso a las actividades de la vida cotidiana sea realizado por un grupo multidisciplinario con enfoque biopsicosocial.
Article
Full-text available
Bone metastases are frequent and debilitating consequence for many tumors, such as breast, lung, prostate, and kidney cancer. Many studies report the importance of the immune system in the pathogenesis of bone metastasis. Indeed, bone and immune system are strictly linked to each other because bone regulates the hematopoietic stem cells from which all cells of the immune system derive, and many immunoregulatory cytokines influence the fate of bone cells. Furthermore, both cytokines and factors produced by immune and bone cells promote the growth of tumor cells in bone, contributing to supporting the vicious cycle of bone metastasis. This review summarizes the current knowledge on the interactions among bone, immune, and tumor cells aiming to provide an overview of the osteoimmunology field in bone metastasis from solid tumors.
Article
Full-text available
CD99 is a transmembrane glycoprotein expressed in physiological conditions by cells of different tissues, including osteoblasts (OBs). High or low CD99 levels have been detected in various pathological conditions, and the supernatant of some carcinoma cell lines can modulate CD99 expression in OB-like cells. In the present work we demonstrate for the first time that two different human myeloma cell lines (H929 and U266) and, in a less degree, their conditioned media significantly downregulate CD99 expression in normal human OBs during the differentiation process. In the same experimental conditions the OBs display a less differentiated phenotype as demonstrated by the decreased expression of RUNX2 and Collagen I. On the contrary, when CD99 was activated by using a specific agonist antibody, the OBs become more active as demonstrated by the upregulation of Alkaline Phosphatase, Collagen I, RUNX2, and JUND expression. Furthermore, we demonstrate that the activation of CD99 is able to induce the phosphorylation of ERK 1/2 and AKT intracellular signal transduction molecules in the OBs.
Article
Full-text available
LIGHT, a TNF superfamily member, is involved in T-cell homeostasis and erosive bone disease associated with rheumatoid arthritis. Herein, we investigated whether LIGHT has a role in Multiple Myeloma (MM)-bone disease. We found that LIGHT was overproduced by CD14+ monocytes, CD8+ T-cells and neutrophils of peripheral blood and bone marrow (BM) from MM-bone disease patients. We also found that LIGHT induced osteoclastogenesis and inhibited osteoblastogenesis. In cultures from healthy-donors, LIGHT induced osteoclastogenesis in RANKL-dependent and -independent manners. In the presence of a sub-optimal RANKL concentration, LIGHT and RANKL synergically stimulated osteoclast formation, through the phosphorylation of Akt, NFκB and JNK pathways. In cultures of BM samples from patients without bone disease, LIGHT inhibited the formation of CFU-F and CFU-OB as well as the expression of osteoblastic markers including collagen-I, osteocalcin and bone sialoprotein-II. LIGHT indirectly inhibited osteoblastogenesis in part through sclerostin expressed by monocytes. In conclusion, our findings for the first time provide evidence for a role of LIGHT in MM-bone disease development.
Article
Full-text available
Psoriatic arthritis occurs in 30% of psoriasis patients, and the treatment can be challenging in some patients. Recently, the US Food and Drug Administration approved ustekinumab, a fully human monoclonal antibody, for the management of psoriatic arthritis. In this article, we review large-scale randomized clinical trials addressing the efficacy and safety profile of ustekinumab for the treatment of psoriatic arthritis.
Article
Children with 21-hydroxylase deficiency (21-OHD) need chronic glucocorticoid (cGC) therapy to replace congenital deficit of cortisol synthesis, and this therapy is the most frequent and severe form of drug-induced osteoporosis. In the study we enrolled 18 patients (9 females) and 18 sex- and age-matched controls. We found in 21-OHD patients high serum and leukocyte levels of dickkopf-1 (DKK1), a secreted antagonist of the Wnt/β-catenin signaling pathway, known to be a key regulator of bone mass. In particular, we demonstrated by flow cytometry, confocal microscopy, and real time PCR that monocytes, T lymphocytes and neutrophils from patients expressed high levels of DKK1, which may be related to the cGC therapy. In fact, we showed that dexamethasone treatment markedly induced the expression of DKK1 in a dose- and time-dependent manner in leukocytes. The serum from patients containing elevated levels of DKK1 can directly inhibit in vitro osteoblast differentiation and Receptor Activator of NF-kappaB Ligand (RANKL) expression. We also found a correlation between both DKK1 and RANKL or C-terminal telopeptides of Type I collagen serum levels in 21-OHD patients on cGC treatment. Our data indicated that DKK1, produced by leukocytes, may contribute to the alteration of bone remodeling in 21-OHD patients on cGC treatment.
Article
Interleukin-17 (IL-17 or IL-17A), a pleiotropic cytokine produced by T helper type 17 cells, is involved in the pathogenesis of various autoimmune and inflammatory disorders, including periodontitis. Although the ability of pro-inflammation in periodontitis has been widely investigated, the other biological functions of IL-17, including its role in bone remodelling and the underlying molecular mechanisms, have not been well clarified. In the present study, IL-17 could significantly enhance the expression of receptor activator for nuclear factor-κB ligand (RANKL) and inhibit the expression of osteoprotegerin (OPG) in human periodontal ligament cells, the two critical indicators for osteoclastogenesis, suggesting that IL-17 may play a destructive role in the pathogenesis of periodontal bone remodelling. Pharmaceutical signal inhibitors targeted at mitogen-activated protein kinases, Akt or nuclear factor-κB signals, inhibited IL-17-induced RANKL and OPG regulation. Notably, the enhancement of RANKL was significantly blocked by the inhibitors of c-Jun N-terminal kinase and nuclear factor-κB signals. The upstream signals were further investigated with the small interfering RNA. Both tumour necrosis factor receptor-associated factor 6 and TNF receptor associated factor (TRAF) family member-associated nuclear factor κ-light-chain enhancer of activated B cells (NF-κB) activator (TANK)- binding kinase 1 were found to be the critical signal molecules for IL-17-dependent RANKL regulation in human periodontal ligament cells. These findings may provide comprehensive understanding of the role of IL-17 in the pathogenesis of periodontitis and might also provide a reasonable route for periodontitis therapy.
Article
In 2009, GRAPPA published their first evidence-based recommendations for the treatment of psoriasis and psoriatic arthritis (PsA). Since then, new information has been published and drugs developed. We summarize evidence for the efficacy of available treatments for peripheral joint involvement in PsA. We performed a systematic review of current literature on the efficacy of different therapies, management, and therapeutic strategies for peripheral arthritis involvement in PsA, in order to provide information for the development of the new GRAPPA treatment recommendations.
Article
After encountering antigen, helper T (TH) cells undergo differentiation to effector cells, which can secrete high levels of interferon-, interleukin-4 (IL-4), IL-10 and other immunomodulators. How TH cells acquire, and remember, new patterns of gene expression is an area of intensive investigation. The process is remarkably plastic, with cytokines being key regulators. Extrinsic signals seem to be integrated into cell-intrinsic programming, in what is becoming an intriguing story of regulated development. We summarize the latest insights into mechanisms that govern the lineage choices that are made during TH-cell responses to foreign pathogens.