ArticlePDF Available

Abstract and Figures

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.
Content may be subject to copyright.
Oncotarget1
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Advance Publications 2014
LIGHT/TNFSF14 increases osteoclastogenesis and decreases
osteoblastogenesis in multiple myeloma-bone disease
Giacomina Brunetti1,*, Rita Rizzi2,*, Angela Oranger1, Isabella Gigante1,
Giorgio Mori3, Grazia Taurino1, Teresa Mongelli1, Graziana Colaianni1,
Adriana Di Benedetto1, Roberto Tamma1, Giuseppe Ingravallo4, Anna Napoli4,
Maria Felicia Faienza5, Anna Mestice2, Paola Curci2, Giorgina Specchia2, Silvia
Colucci1,*, Maria Grano1,*
1 Department of Basic and Medical Sciences, Neurosciences and Sense Organs, section of Human Anatomy and Histology,
University of Bari, Bari, Italy
2 Department of Emergency and Organ Transplantation, Section of Hematology with Transplantation, University of Bari,
Bari, Italy
3Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
4Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
5Department of Biomedical Sciences and Human Oncology, University of Bari, Bari, Italy
*These authors contributed equally to this work
Correspondence to:
Brunetti Giacomina, e-mail: giacomina.brunetti@uniba.it
Keywords: LIGHT/TNFSF14, multiple myeloma, bone disease, osteoclast, osteoblast
Received: August 08, 2014 Accepted: October 23, 2014 Published: November 12, 2014
ABSTRACT
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 ndings for the rst time provide evidence for a role
of LIGHT in MM-bone disease development.
INTRODUCTION
Multiple Myeloma (MM)-bone disease,
characterized by osteolytic lesions, is the most frequent
clinical manifestation of symptomatic MM, being detected
in 70 to 80% of patients at diagnosis and up to 90% at
relapse. It increases the risk of skeletal-related events
such as bone pain, pathological fractures, and spinal cord
compression [1].
Osteolytic lesions result from an imbalance
between increased osteoclast (OC) activity and reduced
osteoblast (OB) repair [2–4]. The latter has also been
related to suppressed functions of Wnt-signalling due
to MM-cells through the expression of Wnt inhibitors
such as dickkopf-1 (DKK1) and Sclerostin [5–11]. In
addition, several cytokines belonging to tumour necrosis
factor superfamily (TNFSF) have been implicated in
the increased osteoclastogenesis [2, 3, 12–13]. Among
these, decoy receptor 3 (DcR3) plays an important role
in the OC formation occurring in MM-bone disease,
as we previously described [14]. DcR3 is known to
be also a soluble receptor of LIGHT [15] (homologous
Oncotarget2
www.impactjournals.com/oncotarget
to Lymphotoxins exhibiting Inducible expression and
competing with herpes simplex virus Glycoprotein D for
herpes virus entry mediator [HVEM], a receptor expressed
by T lymphocytes), whose potential involvement in MM
is unknown.
LIGHT is a member of TNFSF (TNFSF14)
expressed on cells with an immunological role such as
activated T-cells, monocytes, granulocytes, spleen cells,
and immature dendritic cells [15, 16]. As membrane-
anchored or secreted form, LIGHT can bind two
membrane-bound TNFSF signalling receptors, HVEM
and lymphotoxin beta receptor (LTβR). HVEM is
expressed on endothelial, dendritic, natural killer,
T- and B-cells [17, 18] while LTβR is expressed on
broblasts, monocytes, endothelial, epithelial and
stromal cells [19]. Following the interaction of LIGHT
with HVEM or LTβR, the recruitment of TNF receptor
(TNFR)-associated factor-2 (TRAF2) and TRAF5
occurs, leading to gene induction through the activation
of Nuclear-Factor-kappaB (NFκB) or c-Jun N-terminal
kinase (JNK)/ activator protein 1 (AP-1) pathway, and
nally resulting in cytokine production, cell survival or
proliferation [20–23]. The LIGHT–LTβR interaction can
also lead to cell death through the recruitment of TRAF3
and subsequent activation of caspases [24, 25]. Through
the interaction with HVEM, LIGHT is described as a
potent T-cell co-stimulatory molecule [13, 17, 26, 27]; its
constitutive expression on T-cells causes activation and
expansion of these cells, favouring the development of
autoimmune diseases [28, 29]. Moreover, LIGHT has
been implicated in rheumatoid arthritis bone erosions
[30, 31]. To date, there are three literature reports on
the contribution of LIGHT to OC formation, reaching
conicting results [30–32]. In particular, LIGHT was
reported to induce in vitro differentiation of OCs from
peripheral blood (PB) CD14+ monocytes of healthy-
donors, when co-cultured with nurse-like cells isolated
from the synovium of patients with rheumatoid arthritis
[30]. Conversely, no OCs differentiated from the same
CD14+ monocytes cultured alone [30]. In addition, other
Authors reported that, in the presence or absence of the
key pro-osteoclastogenic cytokine receptor activator
of nuclear factor-kappaB ligand (RANKL), LIGHT
induced OC differentiation from human peripheral
blood mononuclear cells (PBMCs) of healthy-donors
[31, 32]. The in vitro data regarding the LIGHT pro-
osteoclastogenic role as well as the LIGHT high serum
levels [31] found in rheumatoid arthritis patients
supported a LIGHT contribution to the pathological bone
resorption.
Based on the above literature data and consistently
with our previous studies [8, 12, 14], we investigated the
expression of LIGHT in MM patients and the role that
this cytokine may play in the osteoclastogenesis and
osteoblastogenesis occurring in MM-bone disease.
RESULTS
LIGHT expression in monocytes, T-cells,
neutrophils and myeloma-cells from patients
and controls
By means of real-time PCR, western blotting,
ow cytometry and immunohistochemistry, we assessed
the expression of LIGHT in BM aspirates and biopsies
from patients as well as in PB from patients and healthy-
donors. Using these different methods, LIGHT resulted
overexpressed in 52/58 (90%) of MM-bone disease
samples, at both mRNA and protein levels; otherwise in
all the other samples, its expression resulted at the lowest
detectable levels by real-time PCR, and undetectable
by western blotting. In particular, LIGHT expression
was detected in CD14+ monocytes from all the positive
samples whereas, in 50% of them, it was detected in CD2
+
T-cells and/or neutrophils, too. The above results, referred
to PB samples analyzed by real-time PCR and western
blotting, are shown in Figures 1A and 1B, respectively.
The corresponding BM samples gave overlapping results
(data not shown). In Table 1, the mean values of the ow
cytometry results are detailed; they are referred to CD14+
monocytes, CD16+ neutrophils and CD8+ T-cells. The latter
cells were identied as the main LIGHT expressing T-cell
subset in MM-bone disease samples. Representative dot
plots of LIGHT cell expression are shown in Figure 1C.
By western blotting, we found low expression
of LIGHT in human myeloma cell lines (HMCLs -
i.e. H929, RPMI-8226, U266) as well as in CD138+
myeloma-cells, isolated from MM-bone disease
patients. In these cells, by ow cytometry, we detected
LIGHT expression at a percentage ranging from 2 to
5 (data not shown). By immunohistochemistry, we
demonstrated strong expression of LIGHT in BM biopsy
samples from MM-bone disease patients (Figure 1D).
We did not nd statistically signicant difference
in LIGHT serum levels among patients with MM-
bone disease (207.71 ± 26.53 pg/ml) or symptomatic
MM without bone disease (179.84 ± 20.48 pg/ml) as
well as in the other samples from patients with sMM
(237 ± 89 pg/ml), MGUS (183 ± 20.58 pg/ml), non-
neoplastic disease (199 ± 21.2 pg/ml) and healthy-donors
(189.84 ± 20.83 pg/ml).
Anti-LIGHT monoclonal antibody affects
osteoclast formation in cultures of PBMCs and
BMMNCs from MM-bone disease patients
In culture media of PBMCs and BM mononuclear
cells (BMMNCs) from MM-bone disease patients, we
found higher LIGHT levels than in those from controls
(1939 ± 220 pg/ml vs 74.7 ± 30 pg/ml, p < 0.001;
Oncotarget3
www.impactjournals.com/oncotarget
Figure 1: LIGHT expression in patients and controls. Monocytes, T-cells and neutrophils from peripheral blood of controls (ctr),
MGUS, smoldering MM (sMM) and symptomatic MM patients without or with bone disease were assessed for LIGHT expression by real-
time PCR (A), western blotting (B) and ow cytometry (C). LIGHT immunostaining was performed in bone marrow biopsies (D).
(Continued)
Oncotarget4
www.impactjournals.com/oncotarget
1750 ± 352 pg/ml vs 66.2 ± 44 pg/ml, p < 0.01,
respectively). The culture treatment with sequential
escalating doses, ranging from 0.005 to 500 ng/ml, of
anti-LIGHT monoclonal antibody (mAb) induced a
dose-dependent inhibition of osteoclastogenesis, quite
different in cultures derived from PBMCs and BMMNCs,
respectively. Indeed in PBMC cultures, 30% inhibition
of osteoclastogenesis was induced by the lowest dose
(0.005 ng/ml) of the anti-LIGHT mAb, increasing
up to 50% at the highest dose of the mAb (500 ng/ml)
(Figure 2A). In BMMNC cultures, the highest dose
(500 ng/ml) of the mAb was instead required to observe
Figure 1: The lowest mRNA levels of LIGHT were detected in monocytes (Aa), T-cells (Ab) and neutrophils (Ac) from
controls, MGUS, sMM and symptomatic MM patients without bone disease. As compared with all of them, LIGHT mRNA
higher levels of 11-fold (P < 0.0001) in monocytes (Aa), 9.2 fold ( p < 0.001) in T-cells (Ab) and 1.8- fold ( p < 0.01) in
neutrophils (Ac) were detected in symptomatic MM patients with bone disease. In these latter, high levels of LIGHT protein
were detected by western blotting in 90% of monocytes (Ba), 50% of T-cells (Bb) and 50% of neutrophils (Bc), whereas the
samples of symptomatic MM patients without bone disease, sMM, MGUS and controls did not display a detectable LIGHT
protein amount. Flow cytometry dot plots showed LIGHT expression on CD14+ monocytes, CD8+ T-cells and CD16+
neutrophils from a representative symptomatic MM patient without bone disease, and a symptomatic MM patient with bone-
disease; in particular, LIGHT levels were higher in the former than in the latter (C). LIGHT positive immunostaining was also
observed in the bone marrow biopsy from a representative symptomatic MM patient with bone-disease; conversely, LIGHT
resulted negative in a symptomatic MM patient without bone-disease (D). The photomicrographs were obtained using a Nikon
Eclipse E400 microscope equipped with a Nikon plan Apo 20×/0.75 DICM (Nikon, Italia), Magnication 200X.
ERK, extracellular signal-regulated kinase.
Table 1: Cytouorimetric expression of LIGHT in CD14+ Monocytes, CD8+ T-cells and CD16+
Neutrophils from all peripheral blood and bone marrow samples.
Peripheral Blood
CD14+ Monocytes CD8+ T-cells CD16+ Neutrophils
Healthy-
donors
Symptomatic
MM w/o
bone disease
Symptomatic
MM with
bone disease
Healthy-
donors
Symptomatic
MM w/o
bone disease
Symptomatic
MM with
bone disease
Healthy-
donors
Symptomatic
MM w/o
bone disease
Symptomatic
MM with
bone disease
1 ± 0.5 3.6 ± 2.8 47.1 ± 9.5* 1.1 ± 0.5 1 ± 0.5 8.0 ± 5.5§ 1 ± 0.9 2 ± 1.1 40.3 ± 17.8*
Bone Marrow
CD14+ Monocytes CD8+ T-cells CD16+ Neutrophils
Patients
with non-
neoplastic
disease
Symptomatic
MM w/o
bone disease
Symptomatic
MM with
bone disease
Patients
with non-
neoplastic
disease
Symptomatic
MM w/o
bone disease
Symptomatic
MM with
bone disease
Patients
with non-
neoplastic
disease
Symptomatic
MM w/o
bone disease
Symptomatic
MM with
bone disease
1.1 ± 0.2 3.5 ± 2.3 48.5 ± 8.5* 0.8 ± 0.3 1 ± 0.8 12.7 ± 7.8§ 0.9 ± 0.3 2.1 ± 1.1 21.2 ± 9.6*
*P < 0.0001;§P < 0.009
Oncotarget5
www.impactjournals.com/oncotarget
a 50% reduction of osteoclastogenesis rate (Figure 2B).
Since spontaneous osteoclastogenesis in vitro occurred
only in MM-bone disease patients, we did not test the
effect of anti-LIGHT mAb in PBMCs and BMMNCs of
MGUS and sMM patients.
Anti-LIGHT mAb affects osteoblast
differentiation in cultures of BMNCs
from MM-bone disease patients
To investigate whether LIGHT could be implicated
in the impaired OB differentiation occurring in MM-bone
disease, we performed long-term cultures (allowing cell
to cell contacts) of patients’ bone marrow nuclear cells
(BMNCs), in the absence or presence of the anti-LIGHT
mAb (100 or 200 ng/ml). CFU-F and CFU-OB formation
was referred to the early and the late phase of OB
differentiation, respectively. In the absence of anti-LIGHT
mAb, the formation rate of both the CFU-F and CFU-OB
was low (Figure 3 A-B). Otherwise, in the presence of anti-
LIGHT mAb at both the concentrations, a dose-dependent
increase of CFU-F and CFU-OB formation occurred
(Figure 3 A, B). The anti-LIGHT mAb seems to exert the
above effects through the induction of BM stromal fraction
proliferation, as shown with the 5’-Bromo-2’-deoxyuridine
(BrdU) test. Indeed, there was a dose-dependent increase
of BrdU incorporation by the BMNCs, cultured in
the presence of 100 or 200 ng/ml anti-LIGHT mAb
(Figure 3C). Thereafter by ow cytometry, we identied
the CD45+ cells as the source of LIGHT in CFU-F and
CFU-OB cultures from MM-bone disease patients (data
not shown). Additionally, the puried BM stromal cell
fraction (BMSCs), cultured in osteogenic medium, did
not express LIGHT mRNA in contrast with the positive
control, consisting of LIGHT expressing T-cells from
MM-bone disease patients, as shown in Figure 3D.
Figure 2: Anti-LIGHT mAb inhibits the osteoclast formation in cultures from MM-bone disease. Multinucleated and
TRAP+ cells, differentiated from peripheral blood mononuclear cell (PBMC) (A) and bone marrow mononuclear cells (BMMNC)
(B) cultures of MM-bone disease patients, were evaluated after 21 days of culture in the presence of anti-LIGHT mAb or control anti-IgG
mAb. The anti-LIGHT mAb culture treatment resulted in a dose-dependent inhibition of osteoclastogenesis, which was not affected by the
control anti-IgG mAb. The number of multinucleated and TRAP+ cells, identied as OCs, are represented in the graphs as mean ± SE of
all experiments performed in each patient’s sample.
Oncotarget6
www.impactjournals.com/oncotarget
Figure 3: Anti-LIGHT mAb effect on CFU-F and CFU-OB formation in BMNC cultures of MM-bone disease
patients. (A) CFU-F and (B) CFU-OB detected in bone marrow nuclear cell (BMNC) cultures from MM-bone disease patients, in the
absence (0) or presence of 100 or 200 ng/ml anti-LIGHT neutralizing mAb. The results were compared to those obtained from untreated
BMNC cultures. Anti-LIGHT mAb signicantly increased CFU-F (A) and CFU-OB (B) formation in a dose-dependent manner. No effect
was detected in the presence of a control anti-IgG mAb. The graphs represent the mean number of CFU-F/well or CFU-OB/well ± SE of
6 independent experiments performed in triplicate. (C) 5-bromo-2-deoxyuridine (BrdU) incorporation evaluated in BMNC cultures from
MM-bone disease patients, treated or not with 100 and 200 ng/ml anti-LIGHT mAb, which positively affected cell proliferation. The graph
represents BrdU incorporation evaluated in 6 experiments performed in quadruplicate. (D) Puried bone marrow stromal cells (BMSCs)
cultured in osteogenic medium for 0, 10 and 20 days did not express LIGHT mRNA. The positive control was represented by T-cells from
MM-bone disease patients. (E) Anti-LIGHT mAb increased the expression at mRNA level of Alkaline Phosphatase (ALP) and Collagen-I
(COLL-I). (F) Anti-LIGHT mAb increased the expression at mRNA level of osteocalcin (OCN), bone sialoprotein II (BSP II), Osterix
(OSX) and Fra-2. The graphs show 6 real-time PCR experiments performed in triplicate.
Oncotarget7
www.impactjournals.com/oncotarget
By real-time PCR in CFU-F cultures from those patients,
we found a low expression of ALP and COLL-I, strongly
increased after treatment with 100ng/ml or 200ng/ml anti-
LIGHT mAb (Figure 3E). Further in CFU-OB untreated
cultures, we detected a low expression of BSPII and OCN
(matrix glycoproteins typically expressed during the late
phase of OB differentiation) as well as of Fra-2 and OSX
(transcription factors), as represented in Figure 3F.
In the presence of 100 or 200ng/ml anti-LIGHT mAb, we
detected signicantly increased mRNA levels of the above
molecules. The control anti-IgG mAb did not affect such
mRNA expression (data not shown).
LIGHT induces osteoclast differentiation
from healthy-donor PBMCs and puried
CD14+ monocytes
We investigated the effect of LIGHT on OC
differentiation in cultures of unfractionated PBMCs
or puried CD14+ monocytes from healthy-donors. In
these cultures, the monocyte precursors differentiate
into multinucleated TRAP+ OCs within 18–21 days,
only in the presence of MCSF (25 ng/ml) and RANKL
(30 ng/ml). In PBMC cultures treated with MCSF and
LIGHT at increasing concentrations (5, 20 or 50 ng/ml), a
dose-dependent formation of multinucleated and TRAP+
cells was also seen. In the presence of LIGHT, however,
the formation of a lower number of OCs than in RANKL
(30 ng/ml) treated cultures appeared. Moreover, in PBMC
cultures treated with both LIGHT (at the concentrations
of 5, 20 or 50 ng/ml) and RANKL (at a sub-optimal
dose of 20 ng/ml), active osteoclastogenesis occurred.
Herein, the rate of OC formation was signicantly
higher than in the presence of either LIGHT or RANKL
alone (Figure 4A). The OCs formed in the presence of
LIGHT (with or without RANKL) were functional, as
demonstrated by their ability in resorbing mineralized
matrix. In particular, in the PBMC cultures, OC
resorption area increased proportionally to the escalating
concentrations of LIGHT; this aspect was more evident
in cultures treated with both LIGHT and RANKL, as
represented in Figure 4B. The same results, referred
Figure 4: LIGHT pro-osteoclastogenic effect. LIGHT treatment of cultures of PBMCs from healthy-donors resulted in dose-
dependent increase of OC formation (A) and resorption activity (B). These effects were more evident after culture treatment with both
RANKL and LIGHT. TRAP+ OC number (A), and resorption area percentage (B) were assessed, and the results represented the mean ±
SE of 12 independent experiments. (C) Western blot analysis of phosphorylated AKT, JNK, IKB in pre-osteoclasts from healthy-donors
incubated with RANKL, RANKL + LIGHT, or LIGHT over a time course (0 to 60 min).
Oncotarget8
www.impactjournals.com/oncotarget
to both the number of formed OCs and the resorption
area, were obtained in cultures from puried CD14+
monocytes (data not shown).
To further investigate LIGHT effect on OC
formation in the presence or absence of RANKL, we
analyzed both the intracellular signal and the expression of
HVEM and LTβR in pre-osteoclasts treated with RANKL
and/or LIGHT. RANKL as well as LIGHT induced the
phosphorylation of IκBα (at 5–10 min for RANKL, and
longer for LIGHT), JNK (maximum at 50–60 min) and
Akt (maximum at 40–60 min) (Figure 4C). Otherwise,
the simultaneous treatment with LIGHT and RANKL
resulted in early and/or prolonged phosphorylation of
Akt (persisting for all the investigated times), IκBα
(maximum at 5–20 min) and JNK (maximum at 40–60
min) (Figure 4C). By contrast, the expression of HVEM
and LTβR did not signicantly change after the pre-
osteoclast exposure to RANKL, LIGHT or both (data
not shown).
LIGHT effect on osteoblastogenesis
We investigated the effect of LIGHT on
osteoblastogenesis by assessment of CFU-F and CFU-OB
formation, occurring in cultures of BMNCs from patients
without MM-bone disease (including symptomatic MM,
sMM, MGUS, non-neoplastic disease), in the presence or
absence of 100 or 200 ng/ml LIGHT. In BMNC cultures,
we found that LIGHT treatment strongly inhibited both
CFU-F (Figure 5A) and CFU-OB (Figure 5B) formation
in a dose-dependent manner. Moreover, we found that
LIGHT impaired the expression of osteogenic markers
of both early and late phase of OB differentiation. In the
early phase, LIGHT particularly inhibited the expression
of Alkaline Phosphatase (ALP), and Collagen-I (COLL-I),
Fra-2 and Jun-D (Figure 5C); differently in the late phase,
it decreased mRNA levels of osteopontin (OPN), bone
sialoprotein II (BSP II), osteocalcin (OCN) and Osterix
(OSX) (Figure 5D).
Figure 5: LIGHT effect on CFU-F and CFU-OB formation. (A) CFU-F and (B) CFU-OB formation were detected in BMNC
cultures from MGUS and non-neoplastic patients in the absence (0) or presence of 20, 50 or 100 ng/ml LIGHT. The results were compared
to those obtained from parallel untreated cultures. LIGHT signicantly inhibited both CFU-F (A) and CFU-OB (B) formation in a dose-
dependent manner. The graphs represent the mean number of CFU-F/well or CFU-OB/well ± SE of 6 independent experiments performed
in triplicate. (C) LIGHT inhibited the expression at mRNA level of Alkaline Phosphatase (ALP), Collagen-I (COLL-I), Fra-2 and Jun-D.
(D) LIGHT signicantly inhibited the expression at mRNA level of osteopontin (OPN), bone sialoprotein II (BSP II), osteocalcin (OSC)
and Osterix (OSX). The graphs show 6 real-time PCR experiments performed in triplicate.
Oncotarget9
www.impactjournals.com/oncotarget
These experiments, however, did not indicate
whether LIGHT exerts a direct or indirect effect
on osteoblastogenesis, because BMNCs include
mesenchymal (i.e. stromal) cells differentiating into OBs
and hematopoietic cells. On the other hand, the latter
cells are known to express LIGHT receptors [17–19],
and to secrete soluble factors positively affecting CFU-F
and CFU-OB survival and proliferation [33]. To address
this issue, we performed cultures of either bone marrow
stromal cells (BMSCs) or mesenchymal stem cells
(MSCs) from dental follicle, which allowed to exclude
hematopoietic cell involvement in LIGHT-mediated
osteoblastogenesis; both BMSCs and MSCs are indeed
lacking in hematopoietic cell fraction. The BMSCs as
well as the MSCs were cultured in osteogenic medium
and in the presence or absence of LIGHT; MSCs were
co-cultured with monocytes and/or T-cells, too. LIGHT
resulted to affect osteoblastogenesis neither in the cultures
of BMSCs (data not shown) nor in those of MSCs alone
(Figure 6A), suggesting that it is not able to exert a
direct effect on osteoblastogenesis. Conversely, in the
co-cultures of MSCs either with monocytes plus T-cells or
with monocytes alone, we detected a signicant LIGHT
impairment of osteoblastogenesis (Figure 6A). Indeed in
Figure 6: LIGHT effect on osteoblastogenesis in co-cultures of mesenchymal stem cells and monocytes with or without
T-cells. (A) For 8 days in osteogenic medium and in the presence or absence of 100 ng/ml LIGHT, mesenchymal stem cells (MSCs) were
cultured alone or co-cultured with monocytes + T-cells, monocytes, or T-cells, respectively. In co-cultures of MSCs with either monocytes
+ T-cells or monocytes alone, LIGHT signicantly inhibited the differentiation of Alkaline Phosphatase positive (ALP+) osteoblasts. The
graphs represent the mean percentage of ALP positive area/well ± SE of 5 independent experiments performed in triplicate. (B) In the same
conditions, LIGHT inhibited the expression, at mRNA level, of Alkaline Phosphatase (ALP) and Collagen-I (COLL-I) in co-cultures of
MSCs with monocytes + T-cells, or monocytes alone, respectively.
(Continued )
Oncotarget10
www.impactjournals.com/oncotarget
LIGHT treated co-cultures, we found signicantly reduced
ALP positive surface/well (Figure 6A) as well as lower
levels of ALP and COLL-I mRNA than in the untreated
co-cultures (Figure 6B). Albeit slightly, LIGHT inhibitory
effect on osteoblastogenesis was more evident in the
co-cultures of MSCs with monocytes plus T-cells than in
those with monocytes alone. Finally, no LIGHT effect on
osteoblastogenesis was detected in the co-cultures between
MSCs and T-cells (Figure 6A).
High Sclerostin levels were associated to LIGHT
inhibitory effect on osteoblastogenesis
In order to investigate the mechanism of LIGHT
inhibitory effect on osteoblastogenesis, we treated the
co-cultures of MSCs with monocytes alone or monocytes
plus T-cells with 100 ng/ml LIGHT for 3, 6 and 12 hours,
after over-night MSC adhesion. At each time, cell
extracts from isolated monocytes, and from monocytes
Figure 6: (C) In the absence or presence of 100 ng/ml LIGHT over a time course [0 to 12 hours (h)], cultures of monocytes
+ T-cells or monocytes alone as well as co-cultures of MSCs with monocytes + T-cells, or monocytes alone were performed.
Thereby, sclerostin expression was evaluated in the cell extracts from each co-culture consisting of monocytes + T-cells or
monocytes alone. The results showed a signicant sclerostin up-regulation occurring at 6 hours in the co-coltures of MSCs
with monocytes + T-cells, and at 12 hours in those of MSCs with monocytes alone. (D) For 8 days in osteogenic medium
and in the presence or absence of 100 ng/ml LIGHT and/or 100 ng/ml anti-sclerostin mAb, mesenchymal stem cells (MSCs)
were cultured alone and co-cultured with monocytes + T-cells or monocytes, respectively. By treating the above described
co-cultures simultaneously with LIGHT and a neutralizing anti-sclerostin mAb, LIGHT inhibitory effect on the differentiation
of ALP+ osteoblasts was partially reverted.
Oncotarget11
www.impactjournals.com/oncotarget
plus T-cells were obtained after purity assessment (99%
CD45+) by ow cytometry. The above cell extracts were
then evaluated for the expression of genes involved in
osteoblastogenesis inhibition such as TNFα, DKK1 and
sclerostin. Among these, statistically signicant results
were obtained only for sclerostin, which is known to be
produced by osteocytes [34]. After 6h LIGHT treatment,
increased levels of sclerostin mRNA were detected in the
monocyte plus T-cell extracts; otherwise in the monocyte
extracts, sclerostin expression increased to a statistically
signicant level after 12h LIGHT treatment, as depicted
in Figure 6C. Therefore, the effect of LIGHT on sclerostin
expression was stronger in the RNA extracts from
monocytes than in those from monocytes plus T-cells. In
the cell extracts from the parallel cultures without MSCs,
no sclerostin mRNA expression was instead detected
(Figure 6C). Interestingly, the treatment of the above
co-cultures with both LIGHT and a neutralizing anti-
sclerostin mAb, partially reverted the sclerostin inhibitory
effect on osteoblastogenesis (Figure 6D).
DISCUSSION
The results of the present study highlighted the high
expression of LIGHT in PB and BM samples from the
large majority of MM-bone disease patients at diagnosis,
in whom LIGHT was demonstrated to be involved
in both increased osteoclastogenesis and decreased
osteoblastogenesis.
Firstly, we detected high LIGHT expression in
CD14+ monocytes, CD8+ T-cells and CD16+ neutrophils
from PB and BM of newly diagnosed patients with MM-
bone disease. Conversely, we found a low or undetectable
expression of LIGHT in the other samples from patients
affected by symptomatic MM without bone disease, sMM,
MGUS, non-neoplastic disease and in healthy-donors.
In the literature, LIGHT was reported in rheumatoid
arthritis erosive bone-disease as expressed by synovial
CD4+ T-cells [35] as well as PB and synovial uid CD14+
monocytes and CD20+ B-cells [36].
Secondly, by means of an in vitro osteoclastogenesis
model consisting of PBMC and BMMNC cultures
from MM-bone disease patients, we demonstrated that
LIGHT exerts a pro-osteoclastogenic effect. Indeed, the
neutralizing anti-LIGHT mAb induced a dose-dependent
inhibition of osteoclastogenesis spontaneously occurring
in both the above cultures derived from MM-bone disease
patients. We observed, however, that in PBMC cultures
lower doses of the mAb inhibited OC formation, compared
with those required to obtain the same effect in the parallel
BMMNC cultures. Consistently with the literature
including our previous reports [2, 12, 14, 37], these results
support the involvement of various cytokines, guring
out the occurrence of alternate or additional pathways
concerning OC formation and activity. Additionally, it
must be also considered the greater complexity of the
cultures derived from BM compared to those from PB
[37, 38].
Furthermore, we investigated LIGHT pro-
osteoclastogenic effect using cultures of PBMCs or
CD14+ monocytes from the healthy-donors, where
LIGHT dose-dependently increased the OC formation
rate. This nding was amplied in the presence of a sub-
optimal concentration of RANKL, indicating that LIGHT
pro-osteoclastogenic effect occurs both in a RANKL-
dependent and -independent way, as also reported in
the literature [31, 32]. Conversely, other Authors did
not describe a LIGHT pro-osteoclastogenic effect from
CD14+ monocyte precursors [30]; these conicting results
might be explained by different culture conditions, such
as the number of monocytes plated by them, which was
lower than ours [30]. In addition, we found that LIGHT
pro-osteoclastogenic effect is related to phosphorylation of
the intracellular signaling, typically activated by RANKL
during osteoclastogenesis and including IkB, AKT and
JNK pathways [39, 40]. However, other Authors [32]
described the phosphorylation of both IkB and AKT,
following LIGHT stimulation of RAW264 and HL60, and
a low rate of phosphorylation of JNK in HL60; both these
murine monocyte cell lines are capable of differentiating
into OCs under appropriate conditions [31, 41]. The
absence or the low rate of JNK phosphorylation might
be related to the use of cell lines rather than primary
human cells [32]. We detected early and sustained
phosphorylation of IkB, AKT and JNK in the cultures
treated with both LIGHT and RANKL, whose synergic
pro-osteoclastogenic effect resulted herein highlighted.
It seems to resemble a mechanism possibly underlying
the in vivo bone destruction occurring in MM-bone
disease, where RANKL is known to play a pivotal role
[2, 12, 13, 42].
Thirdly, the results of the present study showed for
the rst time that LIGHT also affects OB differentiation
in BMNC cultures derived from MM-bone disease
patients; in which, the osteoblastogenesis indeed resulted
signicantly improved by the addition of anti-LIGHT
mAb. Moreover in BMNC cultures from patients without
bone disease, we demonstrated that the addition of LIGHT
inhibited osteoblastogenesis. Since BMNCs include
mesenchymal cell fraction and CD45+ cells, we could
not argue whether LIGHT induces the osteoblastogenesis
impairment through a direct or indirect effect. Thus, we
carried on with our study evaluating OB differentiation
from BMSCs of patients with or without bone disease,
in the presence or absence of LIGHT. In addition,
we assessed the effects of LIGHT on MSCs isolated
from dental follicle, cultured alone or co-cultured with
monocytes and/or T-cells. This model was suitable for our
purposes since MSCs are known to be immunologically
privileged cells [43]. Since no effect of LIGHT on OB
differentiation was detected either in cultures of BMSCs
Oncotarget12
www.impactjournals.com/oncotarget
or in those of MSCs alone, the event of a LIGHT direct
effect could be excluded. On the contrary, the results
we detected in the co-cultures of MSCs with monocytes
alone and/or T-cells pointed out a LIGHT indirect effect,
to some extent occurring through the release of sclerostin
by monocytes. Sclerostin is indeed a key inhibitor of
OB differentiation, known to be produced by osteocytes
[34], whereas there are no literature data concerning
its expression by monocytes. Some Authors, however,
described sclerostin expression within the hematopoietic
islands of embryo livers and by OCs, which are known
to be derived from the fusion of monocyte precursors
[44, 45]. Therefore, we could argue that sclerostin may be
expressed by monocytes under particular conditions. There
are numerous literature reports on monocyte involvement
in osteoblastogenesis [46–48]; in particular, bone resident
macrophages have been recognized as critical regulators of
bone homeostasis and repair in a murine tibia injury model
[48]. The increase of cytokines such as IL-1β, IL-6, IL-10
and TNFα was described as following the MSC-monocyte
contact [49–53]; no role was, however, recognized to these
cytokines in the enhanced monocyte-induced osteogenesis.
In studies using MG-63 osteoblastic cell line or calvarial
OBs, IL-6 has been described as involved in the direct
promotion of osteogenic differentiation [54, 55]. More
recently, monocytes have been reported as regulating the
osteogenic differentiation of MSCs through cell-contact
mechanisms involving Oncostatin-M in monocytes, and
STAT-3 signaling in MSCs [56].
In conclusion, the results of the present study show
for the rst time high expression of LIGHT by monocytes,
T-cells and neutrophils from symptomatic MM patients
with bone disease. Further, our results show that LIGHT
seems to be implicated in the development of MM-bone
disease through a direct pro-osteoclastogenic effect and
an indirect induction of OB suppression. Based on these
ndings, LIGHT provides a potential target for novel
therapeutic strategies.
PATIENTS AND METHODS
Patients
PB, bone marrow (BM) aspirates and biopsies were
obtained from 80 patients newly diagnosed as having
symptomatic MM (58 of them with, and 22 without
related bone disease), 16 with smoldering MM (sMM),
and 35 with monoclonal gammopathy of undetermined
signicance (MGUS). The control samples included PB
and BM from 10 patients with non-neoplastic disease
without skeletal involvement, and PB from 30 healthy
volunteer blood donors (healthy-donors), age and sex
matching with the patients.
The patient characteristics are reported in table 2. All
patients underwent skeletal X-ray, and some of them also
required magnetic resonance imaging or computerized
tomography to assess symptomatic bone sites, pathological
fractures, cord compression or tumour mass. Patient
diagnoses were performed according to the International
Myeloma Working Group’s (IMWG) criteria [1, 57],
and symptomatic MM was also classied according to
the International Staging System (ISS) [58]. Patients and
controls gave their informed consent to the study performed
according to the Declaration of Helsinki, and approved by
Bari University Hospital Ethical Committee.
Cells and cell cultures
Human myeloma cell lines (HMCLs) and
CD138+ cells
HMCLs (H929, RPMI-8226, U266) were cultured in
RPMI-1640 medium supplemented with 10% fetal bovine
serum (FBS; Life Technologies, Milan, Italy). Plasma cells,
identied as CD138+ cells, were isolated from BM aspirates
[8]. RNA or proteins were extracted from HMCLs and fresh
CD138+ cells to evaluate LIGHT expression.
Bone marrow and peripheral blood cells
Buffy coat BM nuclear cells (BMNCs) and PB
mononuclear cells (PBMCs), isolated by Histopaque
1077 density gradient (Sigma, St Louis, MO), were plated.
Thereby, BM stromal cells (BMSCs) and BM mononuclear
cells (BMMNCs) were obtained from BMNC adherent and
non-adherent fractions, respectively. BMMNC and PBMC
cultures were performed to investigate osteoclastogenesis,
whereas BMNCs and BMSCs were cultured to investigate
osteoblastogenesis.
Mesenchymal stem cells (MSCs), bone marrow and
peripheral blood cells
MSCs, isolated as previously described [59], were
used for osteoblastogenesis experiments.
CD14+ monocyte, CD2+ T-cell and
neutrophil isolation
From PBMCs and BMNCs, CD14+ and CD2+ cells
were puried by immunomagnetic selection (Miltenyi
Biotec GmbH, Bergisch Gladbach, Germany), according
to the manufacturer’s instruction. Neutrophils were
isolated as previously described [60]. Only samples with a
purity >98%, checked by ow cytometry, were considered.
RNA and proteins were extracted from puried CD14+
monocytes, CD2+ T-cells and neutrophils to evaluate
LIGHT expression. CD14+ monocytes were also used in
osteoclastogenesis experiments.
Osteoclastogenesis
To investigate LIGHT-dependent OC formation,
BMMNCs and PBMCs from MM-bone disease
patients were plated at 1.5 × 106 cell/cm2 in α-Minimal
Essential Medium (α-MEM, Life Technologies),
Oncotarget13
www.impactjournals.com/oncotarget
supplemented with 10% FBS and cultured in the absence
or presence of anti-LIGHT mAb (0.005 to 500 ng/ml;
R&D Systems Inc., Minneapolis, MN) or control anti-
immunoglobulin G (IgG)-Ab. PBMCs and puried CD14+
cells from healthy-donors were cultured in the presence
or absence of 25 ng/ml recombinant human Macrophage
Colony Stimulating Factor (rh-MCSF) plus either
30 ng/ml rh-RANKL or rh-LIGHT (range 0-50 ng/ml)
(all from R&D Systems). For some experiments, PBMC and
CD14+ cell cultures were treated with MCSF (25 ng/ml),
RANKL (20 ng/ml), and/or LIGHT (range 5-50 ng/ml).
Mature OCs were identied as Tartrate-Resistant Acid
Phosphatase (Sigma) positive (TRAP+) cells containing
3 or more nuclei. OC resorbing activity was assessed
by plating the cells on Millennium multiwell slides
(Millennium Biologix, Kingston, ON, Canada).
To study the phophorylation of IkB, JNK and
AKT, healthy-donor PBMCs were cultured for 10 days
in the presence of MCSF to obtain pre-osteoclasts, that
after overnight starvation were incubated with RANKL,
RANKL + LIGHT, or LIGHT over a time course
(0 to 60 min).
Osteoblastogenesis
BMNCs were plated at 4 × 105/cm2 in osteogenic
differentiating medium [α-MEM supplemented with 10%
FBS, 50 µg/mL ascorbic acid, 10-8 M dexamethasone
and 10 mM beta-glycerophosphate (Sigma)]. BMNCs
from patients without bone disease were cultured in the
presence or absence of rh-LIGHT (range 20-100 ng/ml).
Conversely, BMNCs from MM-bone disease patients were
cultured in the absence or presence of 100 and 200 ng/
ml neutralizing anti-LIGHT mAb or control anti-IgG Ab.
After 21 days, colony forming unit-broblast (CFU-F)
formation was assessed with alkaline phosphatase (ALP,
Sigma) staining. After 30 days, in parallel cultures
CFU-OB formation was assessed with Von Kossa
staining [8]. The mRNA expression of ALP, collagen-I
(COLL-I), Fra-2 and JunD was analyzed in CFU-F
cultures, and that of osteopontin (OPN), Osterix (OSX),
bone sialoprotein II (BSP II) and osteocalcin (OCN) in
CFU-OB cultures.
BMSCs, cultured in α-MEM with 10% FBS, after
reaching conuence, were seeded in 6-well plate at
Table 2: Characteristics of patients and controls.
Parameters Symptomatic MM at diagnosis Smoldering
MM
MGUS Controls
With bone
disease
Without bone
disease
Patients with
non-neoplastic
disease
Healthy-
donors
Number of subjects 58 22 16 35 10 30
Gender (M/F) 30/28 12/10 9/7 18/17 6/4 18/12
Median age (range) 69 (60–85) 64 (54–84) 55 (31–83) 60 (29–83) 63(50–73) 62 (26–72)
Monoclonal
Component:
IgG/IgA/BJ/IgD/NS 47-8-2-1-0 18-3-1-0-0 25-10-0-0-0 - -
ISS-stage ISS-1: 20 ISS-1: 11
ISS-2: 18 ISS-2: 6
ISS-3: 20 ISS-3: 5
Hb<10 g/dL 12 5 - -
Creatinine ≥ 2 mg/dl 5 4 - -
Albumin<3.5 g/dL 15 2 - -
β2-M > 3 mg/L 8 2 - -
β2-M > 6 mg/L 7 - -
LDH > 240 U/L 4 1 - -
Calcium > 10 mg/dl - - -
Abbreviation: MM, multiple myeloma; MGUS, monoclonal gammopathy of undetermined signicance; M, male; F, female;
Ig, immunoglobulin; Hb, hemoglobin; CRP, C-reactive protein; LDH, lactate deydrogenase; ISS, International Staging
System; NS, non secretory.
Oncotarget14
www.impactjournals.com/oncotarget
100000/well and cultured in osteogenic medium for 0, 10
and 20 days. At each time, RNA was extracted to evaluate
LIGHT expression.
MSCs, seeded in 24-well-plate at 400/well, were
cultured alone for 8 days in osteogenic differentiating
medium, in the absence or presence of 100 ng/ml LIGHT
and/or anti-sclerostin mAb (R&D Systems). In the same
conditions, MSCs were also co-cultured with monocytes
alone, T-cells alone or monocytes plus T-cells at 750000
cell/well, respectively. At the end of culture period, ALP
staining or mRNA extraction were performed to evaluate
ALP and COLL-I expression.
Immunohistochemistry
BM biopsies were xed in 4% neutral buffered
formalin solution, decalcied, parafn-embedded, and
cut into 5 μm slices. After standard antigen retrieval
procedures, the sections were incubated overnight at
4°C with primary mouse anti-human LIGHT (Abcam,
Cambridge Science Park). The reaction was revealed with
Dako EnVisionTM FLEX+ detection system (Dako Italia
S.p.A. Milan, Italy).
Flow cytometry analysis
Freshly BM and PB samples were stained with
suitable conjugated antibody: PE-LIGHT (R&D Systems),
CD45-APC-Cy7, FITC-CD8, FITC-CD4, Pe-Cy-5-CD14
and FITC-CD16 (Becton Dickinson, Milan, Italy). Flow
cytometry analysis was performed on a FACSCantoTM II
ow cytometer (Becton Dickinson Immunocytometry
System, Mountain View, CA, USA). Positivity area was
determined using an isotype-matched mAb, and a total of
2000 events for each cell sub-population was acquired.
RNA isolation and real-time polymerase
chain reaction (PCR) amplication
RNA extraction and reverse-transcription as well
as Real-Time PCR amplication were performed, as
previously described [12]. The appropriate primer pairs
were listed in Table 3.
Western blot analysis
Cell protein extracts were analyzed by
western blotting, as previously described [12]. The
following primary antibodies were used: anti-LIGHT and
anti-β-actin (Santa Cruz Biotechnology, Santa Cruz, CA),
anti-pAKT, anti-pIkB, anti-pJNK, anti-total AKT, anti-
IkB, anti-total-ERK and anti-total JNK (Cell Signaling,
San Diego, CA, USA).
Cell proliferation assay
After 10 days of culture, the anti-LIGHT effect
on BMNC proliferation was evaluated with BrdU
incorporation by using a cell proliferation enzyme-linked
immunosorbent assay (ELISA) kit (Roche Diagnostics,
Mannheim, Germany) according to the manufacturer’s
instructions.
Table 3: Sense and antisense primer sequences.
Gene Sense primer Antisense primer Accession number
LIGHT 5 CAGTGTTTGTGGTGGATGG 35 GGGTTGACCTCGTGAGAC NM_003807.3
ALP 5 CGCACGGAACTCCTGACC 35 GCCACCACCACCATCTCG 3NM_000478.4
COLL 1 5 CGTGGCAGTGATGGAAGTG 35 AGCAGGACCAGCGTTACC 3NM_000089.3
OCN 5 ACACTCCTCGCCCTATTG 35 CAGCCATTGATACAGGTAGC 3NM_199173.4
BSP II 5 CTGCTACAACACTGGGCTATG 35 TTCCTTCCTCTTCCTCCTCTTC 3NM_004967.3
OSX 5 GCAAGGTGTATGGCAAGG 35 CATCCGAACGAGTGAACC 3NM_001173467.1
Fra-2 5 GAACCTCGTCTTCACCTATCC 35 CCGCTGCTACTGCTTCTG 3NM_005253.3
Jun-D 5 CTCATCATCCAGTCCAAC 35 GTTCTGCTTGTGTAAATCC 3NM_001286968.1
OPN 5 CTGATGAATCTGATGAACTGGTC 35 GTGATGTCCTCGTCTGTAGC 3NM_001251830.1
Sclerostin 5 CAGCCTTCCGTGTAGTGG 35 TTCATGGTCTTGTTGTTCTCC 3NM_025237.2
β-Actin 5 AATCGTGCGTGACATTAAG 35 GAAGGAAGGCTGGAAGAG 3NM_001101.3
Abbreviation: ALP, Alkaline Phosphatase; COLL I, Collagen Type I; OCN, Osteocalcin; BSP II, Bone Sialoprotein; OSX,
Osterix; OPN, Osteopontin.
Oncotarget15
www.impactjournals.com/oncotarget
ELISA
Patient and control sera as well as media from
BMNC, PBMC and BMMNC cultures, were evaluated
for LIGHT by ELISA (R&D Systems) according to
manufacturer’s instructions. The results were expressed
as mean ± standard error (SE).
Statistical analyses
Statistical analyses were performed by ANOVA or
Student’s t-test with the Statistical Package for the Social
Sciences (spssx/pc) software (SPSS, Chicago, IL, USA). The
results were considered statistically signicant for p < 0.05.
ACKNOWLEDGEMENTS
The authors thank Ministero dell’Istruzione
Università e Ricerca (ex 60% grant to Maria Grano) and
Associazione Italiana per la Ricerca sul Cancro (AIRC,
Grant no. IG_11957 to Maria Grano). The authors thanks
Pasqua Bellocci for technical support.
Conict of interest statement
The authors declare no conict of interest.
REFERENCES
1.
Kyle RA, Rajkumar SV. Criteria for diagnosis, staging, risk
stratication and response assessment of multiple myeloma.
Leukemia. 2009; 23:3–9.
2.
Oranger A, Carbone C, Izzo M, Grano M. Cellular
mechanisms of multiple myeloma bone disease. Clin Dev
Immunol. 2013; 2013:289458.
3.
Garcia-Gomez A, De Las Rivas J, Ocio EM,
Díaz- Rodríguez E, Montero JC, Martín M, Blanco JF,
Sanchez-Guijo FM, Pandiella A, San Miguel JF,
Garayoa M. Transcriptomic prole induced in bone marrow
mesenchymal stromal cells after interaction with multiple
myeloma cells: implications in myeloma progression and
myeloma bone disease. Oncotarget. 2014; 5:8284–8305.
4.
Demchenko YN, Kuehl WM. A critical role for the
NFkB pathway in multiple myeloma. Oncotarget. 2010;
1:59–68.
5.
Oshima T, Abe M, Asano J, Hara T, Kitazoe K, Sekimoto E,
Tanaka Y, Shibata H, Hashimoto T, Ozaki S, Kido S,
Inoue D, Matsumoto T. Myeloma cells suppressed bone
formation by secreting a soluble Wnt inhibitor, sFRP-2.
Blood. 2005; 106:3160–3165.
6.
Wang XT, He YC, Zhou SY, Jiang JZ, Huang YM,
Liang YZ, Lai YR. Bone marrow plasma macrophage
inammatory protein protein-1 alpha (MIP-1 alpha) and
sclerostin in multiple myeloma: Relationship with bone
disease and clinical characteristics. Leuk Res. 2014;
38:525–531.
7.
Brunetti G, Oranger A, Mori G, Specchia G, Rinaldi E,
Curci P, Zallone A, Rizzi R, Grano M, Colucci S. Sclerostin
is overexpressed by plasma cells from multiple myeloma
patients. Ann N Y Acad Sci. 2011; 1237:19–23.
8.
Colucci S, Brunetti G, Oranger A, Mori G, Sardone F,
Specchia G, Rinaldi E, Curci P, Liso V, Passeri G,
Zallone A, Rizzi R, Grano M. Myeloma cells suppress
osteoblasts through sclerostin secretion. Blood Cancer J.
1,e27; 2011.
9.
Terpos E, Christoulas D, Katodritou E, Bratengeier C,
Gkotzamanidou M, Michalis E, Delimpasi S, Pouli A,
Meletis J, Kastritis E, Zervas K, Dimopoulos MA. Elevated
circulating sclerostin correlates with advanced disease
features and abnormal bone remodeling in symptomatic
myeloma: reduction post-bortezomib monotherapy. Int J
Cancer. 2012; 131:1466–1471.
10.
Kristensen IB, Christensen JH, Lyng MB, Møller MB,
Pedersen L, Rasmussen LM, Ditzel HJ, Abildgaard N.
Expression of osteoblast and osteoclast regulatory genes in
the bone marrow microenvironment in multiple myeloma:
only up-regulation of Wnt inhibitors SFRP3 and DKK1 is
associated with lytic bone disease. Leuk Lymphoma. 2014;
55:911–919.
11.
Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B,
Shaughnessy JD Jr. The role of the Wnt-signaling
antagonist DKK1 in the development of osteolytic lesions
in multiple myeloma. N Engl J Med. 2003; 349:2483–2494.
12.
Colucci S, Brunetti G, Rizzi R, Zonno A, Mori G,
Colaianni G, Del Prete D, Faccio R, Liso A, Capalbo S,
Liso V, Zallone A, Grano M. T cells support
osteoclastogenesis in an in vitro model derived from human
multiple myeloma bone disease: the role of the OPG/TRAIL
interaction. Blood. 2004; 104:3722–3730.
13.
Giuliani N, Colla S, Sala R, Moroni M, Lazzaretti M,
La Monica S, Bonomini S, Hojden M, Sammarelli G,
Barillè S, Bataille R, Rizzoli V. Human myeloma
cells stimulate the receptor activator of nuclear factor-
kappa B ligand (RANKL) in T lymphocytes: a potential
role in multiple myeloma bone disease. Blood. 2002;
100:4615–4621.
14.
Colucci S, Brunetti G, Mori G, Oranger A, Centonze M,
Mori C, Cantatore FP, Tamma R, Rizzi R, Liso V,
Zallone A, Grano M. Soluble decoy receptor 3 modulates
the survival and formation of osteoclasts from multiple
myeloma bone disease patients. Leukemia. 2009;
23:2139–2146.
15.
Mauri DN, Ebner R, Montgomery RI, Kochel KD,
Cheung TC, Yu GL, Ruben S, Murphy M, Eisenberg RJ,
Cohen GH, Spear PG, Ware CF. LIGHT, a new member
of the TNF superfamily, and lymphotoxin α are ligands for
herpesvirus entry mediator. Immunity. 1998; 8:21–30.
Oncotarget16
www.impactjournals.com/oncotarget
16.
Tamada K, Shimozaki K, Chapoval AI, Zhai Y, Su J,
Chen SF, Hsieh SL, Nagata S, Ni J, Chen L. LIGHT, a
TNF-like molecule, costimulates T cell proliferation and
is required for dendritic cell-mediated allogeneic T cell
response. J Immunol. 2000; 164:4105–4110.
17.
Harrop JA, Reddy M, Dede K, Brigham-Burke M,
Lyn S, Tan KB, Silverman C, Eichman C, DiPrinzio R,
Spampanato J, Porter T, Holmes S, Young PR, Truneh A.
Antibodies to TR2 (herpesvirus entry mediator), a new
member of the TNF receptor superfamily, block T cell
proliferation, expression of activation markers, and
production of cytokines. J Immunol. 1998; 161:1786–1794.
18.
Kwon BS, Tan KB, Ni J, Oh KO, Lee ZH, Kim KK, Kim YJ,
Wang S, Gentz R, Yu GL, Harrop J, Lyn SD, Silverman C,
Porter TG, Truneh A, Young PR. A newly identied member
of the tumor necrosis factor receptor superfamily with a wide
tissue distribution and involvement in lymphocyte activation.
J Biol Chem. 1997; 272:14272–14276.
19.
Browning JL, Miatkowski K, Sizing I, Grifths D,
Zafari M, Benjamin CD, Meier W, Mackay F. Signaling
through the lymphotoxin β receptor induces the death of
some adenocarcinoma tumor lines. J Exp Med. 1996;
183:867–878.
20.
Harrop JA, McDonnell PC, Brigham-Burke M, Lyn SD,
Minton J, Tan KB, Dede K, Spampanato J, Silverman C,
Hensley P, DiPrinzio R, Emery JG, Deen K, Eichman C,
Chabot-Fletcher M, Truneh A, Young PR. Herpesvirus entry
mediator ligand (HVEM-L), a novel ligand for HVEM/TR2,
stimulates proliferation of T cells and inhibits HT29 cell
growth. J Biol Chem. 1998; 273:27548–27556.
21.
Hsu H1, Solovyev I, Colombero A, Elliott R, Kelley M,
Boyle WJ. ATAR, a novel tumor necrosis factor receptor
family member, signals through TRAF2 and TRAF5. J Biol
Chem. 1997; 272:13471–13474.
22.
Marsters SA1, Ayres TM, Skubatch M, Gray CL, Rothe M,
Ashkenazi A. Herpesvirus entry mediator, a member of the
tumor necrosis factor receptor (TNFR) family, interacts
with members of the TNFR-associated factor family and
activates the transcription factors NF-kappaB and AP-1.
J Biol Chem. 1997; 272:14029–14032.
23.
Arch RH1, Gedrich RW, Thompson CB. Tumor necrosis
factor receptor-associated factors (TRAFs)—a family of
adapter proteins that regulates life and death. Genes Dev.
1998; 12:2821–2830.
24.
Zhai Y, Guo R, Hsu TL, Yu GL, Ni J, Kwon BS, Jiang GW,
Lu J, Tan J, Ugustus M, Carter K, Rojas L, Zhu F,
Lincoln C, Endress G, Xing L, Wang S, Oh KO, Gentz R,
Ruben S, Lippman ME, Hsieh SL, Yang D. LIGHT, a novel
ligand for lymphotoxin beta receptor and TR2/HVEM
induces apoptosis and suppresses in vivo tumor formation
via gene transfer. J Clin Invest. 1998; 102:1142–1151.
25.
Rooney IA, Butrovich KD, Glass AA, Borboroglu S,
Benedict CA, Whitbeck JC, Cohen GH, Eisenberg RJ,
Ware CF. The lymphotoxin-beta receptor is necessary and
sufcient for LIGHT-mediated apoptosis of tumor cells.
J Biol Chem. 2000; 275:14307–14315.
26.
Granger SW, Rickert S. LIGHT-HVEM signaling and the
regulation of T cell-mediated immunity. Cytokine Growth
Factor Rev. 2003; 14:289–296.
27.
Tamada K, Shimozaki K, Chapoval AI, Zhu G, Sica G,
Flies D, Boone T, Hsu H, Fu YX, Nagata S, Ni J, Chen L.
Modulation of T-cell-mediated immunity in tumor and
graft-versus-host disease models through the LIGHT
co-stimulatory pathway. Nat Med. 2000; 6:283–289.
28.
Shaikh RB, Santee S, Granger SW, Butrovich K, Cheung T,
Kronenberg M, et al. Constitutive expression of LIGHT on
T cells leads to lymphocyte activation, inammation, and
tissue destruction. J Immunol. 2001; 167:6330–6337.
29.
Wang J, Lo JC, Foster A, Yu P, Chen HM, Wang Y,
Tamada K, Chen L, Fu YX. The regulation of T cell
homeostasis and autoimmunity by T cell-derived LIGHT.
J Clin Invest. 2001; 108:1771–1780.
30.
Ishida S, Yamane S, Nakano S, Yanagimoto T,
Hanamoto Y, Maeda-Tanimura M, Toyosaki-Maeda T,
Ishizaki J, Matsuo Y, Fukui N, Itoh T, Ochi T, Suzuki R.
The interaction of monocytes with rheumatoid synovial
cells is a key step in LIGHT-mediated inammatory bone
destruction. Immunology. 2009; 128:e315–e324.
31.
Edwards JR, Sun SG, Locklin R, Shipman CM,
Adamopoulos IE, Athanasou NA, Sabokbar A. LIGHT
(TNFSF14), a novel mediator of bone resorption, is
elevated in rheumatoid arthritis. Arthritis Rheum. 2006;
54:1451–1462.
32.
Hemingway F, Kashima TG, Knowles HJ, Athanasou NA.
Investigation of osteoclastogenic signalling of the
RANKL substitute LIGHT. Exp Mol Pathol. 2013; 94:
380–385.
33.
Baksh D, Davies JE, Zandstra PW. Soluble factor
cross-talk between human bone marrow-derived
hematopoietic and mesenchymal cells enhances in vitro
CFU-F and CFU-O growth and reveals heterogeneity in
the mesenchymal progenitor cell compartment. Blood.
2005; 106:3012–3019.
34.
van Bezooijen RL, ten Dijke P, Papapoulos SE, Löwik CW.
SOST/sclerostin, an osteocyte-derived negative regulator
of bone formation. Cytokine Growth Factor Rev. 2005;
16:319–327.
35. Pierer M, Brentano F, Rethage J, Wagner U, Hantzschel H,
Gay RE, Gay S, Kyburz D. The TNF superfamily member
LIGHT contributes to survival and activation of synovial
broblasts in rheumatoid arthritis. Rheumatology (Oxford).
2007; 46:1063–1070.
Oncotarget17
www.impactjournals.com/oncotarget
36.
Kang YM, Kim SY, Kang JH, Han SW, Nam EJ, Kyung HS,
Park JY, Kim IS. LIGHT up-regulated on B lymphocytes
and monocytes in rheumatoid arthritis mediates cellular
adhesion and metalloproteinase production by synoviocytes.
Arthritis Rheum. 2007; 56:1106–1117.
37.
Giuliani N, Lisignoli G, Colla S, Lazzaretti M, Storti P,
Mancini C, Bonomini S, Manferdini C, Codeluppi K,
Facchini A, Rizzoli V. CC-Chemokine ligand 20/macrophage
inammatory protein-3alpha and CC-chemokine receptor 6
are overexpressed in myeloma microenvironment related to
osteolytic bone lesions. Cancer Res. 2008; 68:6840–6850.
38.
El Hajj Dib I, Me´lanie G, Valery S, Romuald M, Michel B,
Kamel S. Multiple myeloma cells directly stimulate bone
resorption in vitro by down-regulating mature osteoclast
apoptosis. Leuk Res. 2008; 2:1279–1287.
39.
Boyle WJ, Simonet WS, Lacey DL. Osteoclast
differentiation and activation. Nature. 2003; 423:337–342.
40.
Wong BR, Besser D, Kim N, Arron JR, Vologodskaia M,
Hanafusa H, Choi Y. TRANCE, a TNF family member,
activates Akt/PKB through a signaling complex involving
TRAF6 and c-Src. Mol Cell. 1999; 4:1041–1049.
41.
Kido S, Inoue D, Hiura K, Javier W, Ito Y, Matsumoto T.
Expression of RANK is dependent upon differentiation
into the macrophage/osteoclast lineage: induction by
1alpha, 25-dihydroxyvitamin D3 and TPA in a human
myelomonocytic cell line, HL60. Bone. 2003; 32:
621–629.
42.
Pearse RN, Sordillo EM, Yaccoby S, Wong BR, Liau DF,
Colman N, Michaeli J, Epstein J, Choi Y. Multiple
myeloma disrupts the TRANCE/ osteoprotegerin
cytokine axis to trigger bone destruction and promote
tumor progression. Proc Natl Acad Sci U S A. 2001; 98:
11581–11586.
43.
Yao S, Pan F, Prpic V, Wise GE. Differentiation of stem
cells in the dental follicle. J Dent Res. 2008; 87:767–771.
44.
Ota K, Quint P, Ruan M, Pederson L, Westendorf JJ,
Khosla S, Oursler MJ. Sclerostin is expressed in
osteoclasts from aged mice and reduces osteoclast-
mediated stimulation of mineralization. J Cell Biochem.
2013; 114:1901–1907.
45.
Kusu N, Laurikkala J, Imanishi M, Usui H, Konishi M,
Miyake A, Thesleff I, Itoh N. Sclerostin is a novel secreted
osteoclast-derived bone morphogenetic protein antagonist
with unique ligand specicity. J Biol Chem. 2003;
278:24113–24117.
46.
Jung Y, Song J, Shiozawa Y, Wang J, Wang Z,
Williams B, Havens A, Schneider A, Ge C, Franceschi RT,
McCauley LK, Krebsbach PH, Taichman RS.
Hematopoietic stem cells regulate mesenchymal stromal
cell induction into osteoblasts thereby participating in
the formation of the stem cell niche. Stem Cells. 2008;
26:2042–2051.
47.
Chang MK, Raggatt LJ, Alexander KA, Kuliwaba JS,
Fazzalari NL, Schroder K, Maylin ER, Ripoll VM,
Hume DA, Pettit AR. Osteal tissue macrophages are
intercalated throughout human and mouse bone lining
tissues and regulate osteoblast function in vitro and in vivo.
J Immunol. 2008; 181:1232–1244.
48.
Alexander KA, Chang MK, Maylin ER, Kohler T,
Muller R, Wu AC, Van Rooijen N, Sweet MJ, Hume DA,
Raggatt LJ, Pettit AR. Osteal macrophages promote in vivo
intramembranous bone healing in a mouse tibial injury
model. J Bone Miner Res. 2011; 26:1517–1532.
49.
Groh ME, Maitra B, Szekely E, Koc ON. Human
mesenchymal stem cells require monocyte-mediated
activation to suppress alloreactive T cells. Exp Hematol.
2005; 33:928–934.
50.
Aggarwal S, Pittenger MF. Human mesenchymal stem cells
modulate allogeneic immune cell responses. Blood. 2005;
105:1815–1822.
51.
Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B,
Parmelee A, Doi K, Robey PG, Leelahavanichkul K,
Koller BH, Brown JM, Hu X, Jelinek I, Star RA,
Mezey E. Bone marrow stromal cells attenuate sepsis via
prostaglandin E(2)-dependent reprogramming of host
macrophages to increase their interleukin-10 production.
Nat Med. 2009; 15:42–49.
52.
Beyth S, Borovsky Z, Mevorach D, Liebergall M,
Gazit Z, Aslan H, Galun E, Rachmilewitz J. Human
mesenchymal stem cells alter antigen-presenting cell
maturation and induce T-cell unresponsiveness. Blood.
2005; 105:2214–2219.
53.
Kim J, Hematti P. Mesenchymal stem cell-educated
macrophages: a novel type of alternatively activated
macrophages. Exp Hematol. 2009; 37:1445–1453.
54.
Bellido T, Borba VZ, Roberson P, Manolagas SC.
Activation of the Janus kinase/STAT (signal transducer
and activator of transcription) signal transduction pathway
by interleukin-6-type cytokines promotes osteoblast
differentiation. Endocrinology. 1997; 138:3666–3676.
55.
Nishimura R, Moriyama K, Yasukawa K, Mundy GR,
Yoneda T. Combination of interleukin-6 and soluble
interleukin-6 receptors induces differentiation and
activation of JAK-STAT and MAP kinase pathways in
MG-63 human osteoblastic cells. J Bone Miner Res. 1998;
13:777–785.
56.
Nicolaidou V1, Wong MM, Redpath AN, Ersek A,
Baban DF, Williams LM, Cope AP, Horwood NJ.
Monocytes induce STAT3 activation in human
mesenchymal stem cells to promote osteoblast formation.
PLoS One. 2012; 7:e39871.
57.
International Myeloma Working Group Criteria
for the classication of monoclonal gammopathies,
multiple myeloma and related disorders: a report of the
Oncotarget18
www.impactjournals.com/oncotarget
International Myeloma Working Group. Br J Haematol.
2003; 121:749–757.
58.
Greipp PR, San Miguel J, Durie BG, Crowley JJ,
Barlogie B, Bladé J, Boccadoro M, Child JA,
Avet-Loiseau H, Kyle RA, Lahuerta JJ, Ludwig H,
Morgan G, Powles R, Shimizu K, Shustik C, Sonneveld P,
Tosi P, Turesson I, Westin J. International staging system
for multiple myeloma. J Clin Oncol. 2005; 23:3412–3420.
59.
Mori G, Ballini A, Carbone C, Oranger A, Brunetti G,
Di Benedetto A, Rapone B, Cantore S, Di Comite M,
Colucci S, Grano M, Grassi FR. Osteogenic
differentiation of dental follicle stem cells. Int J Med Sci.
2012; 9:480–487.
60.
Brunetti G, Faienza MF, Piacente L, Ventura A, Oranger A,
Carbone C, Di Benedetto A, Colaianni G, Gigante M,
Mori G, Gesualdo L, Colucci S, Cavallo L, Grano M. High
dickkopf-1 levels in sera and leukocytes from children
with 21-hydroxylase deciency on chronic glucocorticoid
treatment. Am J Physiol Endocrinol Metab. 2013;
304:E546–E554.
... Активированные ОК также могут увеличивать миграцию миеломных клеток [87], что приводит к расширению зоны поражения кости и прогрессированию ММ. Кроме того, ОК поддерживают иммуносупрессивную среду для миеломных клеток [8], а иммунные клетки усиливают дифференцировку ОК [20,33]. Показано, что остеокластогенез индуцируется CD14-моноцитами, CD8Т-клетками и нейтрофилами у пациентов с ММ путем повышенной продукции белка надсемейства TNF-LIGHT/ TNFSF14. ...
... Показано, что остеокластогенез индуцируется CD14-моноцитами, CD8Т-клетками и нейтрофилами у пациентов с ММ путем повышенной продукции белка надсемейства TNF-LIGHT/ TNFSF14. LIGHT/TNFSF14 (tumor necrosis factor (ligand) superfamily member14) -растворимый провоспалительный цитокин-лиганд для TNFRSF14 [20]. Установлено, что LIGHT взаимодействует с RANKL, усиливая образование ОК при ММ [20]. ...
... LIGHT/TNFSF14 (tumor necrosis factor (ligand) superfamily member14) -растворимый провоспалительный цитокин-лиганд для TNFRSF14 [20]. Установлено, что LIGHT взаимодействует с RANKL, усиливая образование ОК при ММ [20]. Уместно напомнить, что RANKL является наиболее изученным проостеокластогенным цитокином, являясь мишенью для новых методов лечения. ...
Article
Full-text available
In this review, we discuss molecular and cellular mechanisms underlying cross-talk between immune cells and bone cells, both in healthy conditions and in some diseases. We provide short description of the main cell populations of bone tissue, i.e., osteoblasts, osteoclasts, osteocytes, bone marrow macrophages, OsteoMacs, and their effects on immune cells during bone modeling and remodeling. The data are presented on regulatory molecular pathways of bone marrow cell activity, T and B cells, macrophages, and formation of “endosteal niche” by the bone cells. We describe the key system of bone tissue homeostasis: RANK/RANKL/ OPG, which regulates differentiation of osteoclasts and bone destruction. In addition, RANK/RANKL/ OPG system modulates maturation and activity of various T and B cell subsets. We present the data on pleiotropic effects of T cells, B cells, dendritic cells, macrophage subpopulations, Tregs, NK cells, neutrophils upon differentiation and function of osteoblasts and osteoclasts. These effects promote accumulation and maintenance of the bone mass. We describe mechanisms of these effects based on direct cell-to-cell contacts and various soluble mediators and intracellular signaling pathways. A brief characteristic of some diseases is provided with concomitant dysfunction of immune cells and bone cells which play a decisive pathogenetic role (fractures, rheumatoid arthritis, periodontitis, postmenopausal osteoporosis, multiple myeloma). It was shown that the destructive bone inflammation, both in RA and periodontitis, leads to loss of bone mass, being featured by similar pathophysiological mechanisms involving immune and bone cell populations. Therapy of these diseases requires newer treatment strategies aimed not only at pro-inflammatory cytokines, but for increased bone resorption. We describe involvement of activated T cells, their cytokines into the pathogenesis of postmenopausal osteoporosis, thus providing a rationale for the novel term of “immunoporosis”, coined in 2018. The relationships between multiple myeloma cells and bone marrow microenvironment are provided. This cross-talk is based on contact cell-cell interactions, as well as due to effects of soluble mediators upon osteoclasts, stromal cells, and osteoblasts. These effects result in osteolysis, loss of bone mass, and myeloma progression. In conclusion, the relationships between the immune and bone cell populations suggest that they function as an entire regulatory system. This consideration provides a framework for the development of new therapeutic targets for the treatment of bone and immune system disorders.
... It is increased in patients with obesity and has a pro-osteoclastogenic effect. It has been shown that an elevation of its levels is related to osteoporosis [121,122]. ...
Article
Full-text available
Recent scientific evidence has shown an increased risk of fractures in patients with obesity, especially in those with a higher visceral adipose tissue content. This contradicts the old paradigm that obese patients were more protected than those with normal weight. Specifically, in older subjects in whom there is a redistribution of fat from subcutaneous adipose tissue to visceral adipose tissue and an infiltration of other tissues such as muscle with the consequent sarcopenia, obesity can accentuate the changes characteristic of this age group that predisposes to a greater risk of falls and fractures. Other factors that determine a greater risk in older subjects with obesity are chronic proinflammatory status, altered adipokine secretion, vitamin D deficiency, insulin resistance and reduced mobility. On the other hand, diagnostic tests may be influenced by obesity and its comorbidities as well as by body composition, and risk scales may underestimate the risk of fractures in these patients. Weight loss with physical activity programs and cessation of high-fat diets may reduce the risk. Finally, more research is needed on the efficacy of anti-osteoporotic treatments in obese patients.
... It appears that these factors promote osteoclast differentiation through turning on adaptor proteins rather than TRAF6, activating other positive regulators of osteoclastogenesis, leading to cell fusion and maturation [50,53]. Although osteoclastogenic humoral factors are not as potent as RANKL in inducing osteoclast formation, in terms of the size of the formed cell and their resorptive activity, their potential role as osteoclastogenic factors should not be neglected, especially because they are highly expressed around osteolytic bone and joint tissues by immune cells in inflammatory diseases [50,52,57,59,61,62]. Taken together, inflammation acts as a switch that turns on pathological bone resorption through promoting osteoclastogenesis via both canonical and non-canonical pathways. ...
Article
Full-text available
Bone is a mineralized and elastic connective tissue that provides fundamental functions in the human body, including mechanical support to the muscles and joints, protection of vital organs and storage of minerals. Bone is a metabolically active organ that undergoes continuous remodeling processes to maintain its architecture, shape, and function throughout life. One of the most important medical discoveries of recent decades has been that the immune system is involved in bone remodeling. Indeed, chronic inflammation has been recognized as the most significant factor influencing bone homeostasis, causing a shift in the bone remodeling process toward pathological bone resorption. Bone osteolytic diseases typified by excessive bone resorption account for one of the greatest causes of disability worldwide, with significant economic and public health burdens. From this perspective, we discuss the recent findings and discoveries highlighting the cellular and molecular mechanisms that regulate this process in the bone microenvironment, in addition to the current therapeutic strategies for the treatment of osteolytic bone diseases.
... The LIGHT-LTβR interaction also has been reported to induce apoptosis of cancer cells (Zhai et al., 1998), it is important for macrophage activity in wound healing (Petreaca et al., 2012), and it influences lipid metabolism by regulating hepatic lipase expression in hepatocytes (Chellan et al., 2013;Lo et al., 2007). Furthermore, LIGHT participates in additional processes in which a specific receptor has not been implicated, including the resolution of inflammation in an experimental autoimmune encephalomyelitis (Mana et al., 2013), the induction of adipocyte differentiation (Tiller et al., 2011), and the induction of osteoclastogenic signals (Brunetti et al., 2014;Hemingway et al., 2013). ...
Article
Full-text available
HVEM is a TNF (tumor necrosis factor) receptor contributing to a broad range of immune functions involving diverse cell types. It interacts with a TNF ligand, LIGHT, and immunoglobulin (Ig) superfamily members BTLA and CD160. Assessing the functional impact of HVEM binding to specific ligands in different settings has been complicated by the multiple interactions of HVEM and HVEM binding partners. To dissect the molecular basis for multiple functions, we determined crystal structures that reveal the distinct HVEM surfaces that engage LIGHT or BTLA/CD160, including the human HVEM–LIGHT–CD160 ternary complex, with HVEM interacting simultaneously with both binding partners. Based on these structures, we generated mouse HVEM mutants that selectively recognized either the TNF or Ig ligands in vitro. Knockin mice expressing these muteins maintain expression of all the proteins in the HVEM network, yet they demonstrate selective functions for LIGHT in the clearance of bacteria in the intestine and for the Ig ligands in the amelioration of liver inflammation.
... TNF-α also has pro-osteoclastic synergistic effects with very small levels of RANKL, and, therefore, may well also be an OAF in the context of MBD [111,112]. Another member of the TNF superfamily, LIGHT, also appears to antagonise OB differentiation, by reducing OB precursor formation and may also lead to sclerostin expression from monocytes [113]. MIP-1α, also known as chemokine cytokine ligand 3, is expressed at higher levels in MM patients with MBD. ...
Article
Full-text available
Lytic bone disease remains a life-altering complication of multiple myeloma, with up to 90% of sufferers experiencing skeletal events at some point in their cancer journey. This tumour-induced bone disease is driven by an upregulation of bone resorption (via increased osteoclast (OC) activity) and a downregulation of bone formation (via reduced osteoblast (OB) activity), leading to phenotypic osteolysis. Treatments are limited, and currently exclusively target OCs. Despite existing bone targeting therapies, patients successfully achieving remission from their cancer can still be left with chronic pain, poor mobility, and reduced quality of life as a result of bone disease. As such, the field is desperately in need of new and improved bone-modulating therapeutic agents. One such option is the use of bone anabolics, drugs that are gaining traction in the osteoporosis field following successful clinical trials. The prospect of using these therapies in relation to myeloma is an attractive option, as they aim to stimulate OBs, as opposed to existing therapeutics that do little to orchestrate new bone formation. The preclinical application of bone anabolics in myeloma mouse models has demonstrated positive outcomes for bone repair and fracture resistance. Here, we review the role of the OB in the pathophysiology of myeloma-induced bone disease and explore whether novel OB targeted therapies could improve outcomes for patients.
... To study the effects of immune cells on bone cell differentiation coculture experiments have been realized. Furthermore, to evaluate the effect of cell-cell contacts, these cocultures can be prepared with or without a transwell insert [26][27][28][29]. Immune cells can be isolated using immunomagnetic devices, which are commercially available. ...
Article
Full-text available
Immune and bone cells cross talk has been established by different years; however the underlying mechanisms require continuous investigation. To this end both in vivo and in vitro models have been realized and some of this are described in this chapter. In particular, here we described the animal models used for the understanding of lymphocyte role in bone homeostasis, together with some in vitro models.
Article
Aim To evaluate the protein profiles in gingival crevicular fluid (GCF) in relation to clinical outcomes after periodontal surgery and examine if any selected proteins affect the mRNA expression of pro‐inflammatory cytokines in human gingival fibroblasts. Materials and Methods This exploratory study included 21 consecutive patients with periodontitis. GCF was collected, and the protein pattern ( n = 92) and clinical parameters were evaluated prior to surgery and 3, 6 and 12 months after surgery. Fibroblastic gene expression was analysed by real‐time quantitative polymerase chain reaction. Results Surgical treatment reduced periodontal pocket depth (PPD) and changed the GCF protein pattern. Twelve months after surgery, 17% of the pockets showed an increase in PPD. Levels of a number of proteins in the GCF decreased after surgical treatment but increased with early signs of tissue destruction, with LIGHT being one of the proteins that showed the strongest association. Furthermore, LIGHT up‐regulated the mRNA expression of pro‐inflammatory cytokines interleukin (IL)‐6, IL‐8 and MMP9 in human gingival fibroblasts. Conclusions LIGHT can potentially detect subjects at high risk of periodontitis recurrence after surgical treatment. Moreover, LIGHT induces the expression of inflammatory cytokines and tissue‐degrading enzymes in gingival fibroblasts.
Preprint
Full-text available
Purpose:: Multiple myeloma(MM) is a common malignant tumor in the blood system. Despite recent advances in its treatment, its symptomatic remission rate and survival rate are still not optimistic. In the future, it is necessary to continue to search for different treatment targets and new treatment methods in order to improve the quality of life and survival time of patients with MM. The study aims to explore the potential immune related pivotal genes and immune infiltration patterns in MM. Methods: The study included peripheral blood samples from patients with MM who our hospital from October 2020 to April 2022. Obtain a gene chip for research from a comprehensive gene expression database, perform differential expression analysis on the processed gene dataset, and then perform functional enrichment analysis, weighted gene co expression network analysis, GSEA immune infiltration analysis, and LASSO regression analysis on the obtained differential expression genes to obtain the hub genes. Finally, the hub gene TNFSF14 (LIGHT) was validated by qRT-PCR. Results: In the study, three immune-related hub genes (ADAM8, CR2, and TNFSF14) and three main types of peripheral immune cells (activated CD8 T cells, macrophages, and plasma cell like dendritic cells) were obtained, which are closely related to the pathogenesis of MM. Then, by collecting peripheral blood samples from some patients in our hospital and conducting real-time fluorescence quantitative polymerase chain reaction, it was confirmed that the hub gene TNFSF14 (LIGHT) mined in this study was highly expressed in peripheral blood samples from patients with MM, which may indicate that it plays a pathogenic role in MM. Conclusion: The study found that immune-related hub genes (ADAM8, CR2, and TNFSF14) are closely related to the pathogenesis of MM, and should be further researched.
Chapter
Osteoporosis is the most worldwide diffuse skeletal disease requiring new therapeutic strategies for its cure. The discovery of the pro-osteoclastogenic receptor activator of nuclear factor kappa-B ligand (RANKL) and anti-osteoblastogenic sclerostin’s role is strongly changing the therapeutic approach. In this chapter, we overview the literature and data on the use of denosumab and romosozumab, antibodies against RANKL and sclerostin respectively, for osteoporosis management. Clinical trials show that denosumab long-term treatment determines a continuous augment of the bone mineral density (BMD) with few adverse effects. Most recent trials on romosozumab treatment reports bone formation increase and BMD improvement, although there are controversial reports on its adverse effects, with particular regard to cardiovascular events.
Article
Full-text available
LIGHT, also termed TNFSF14, has been reported to play a vital role in different tumors. However, its role in glioma remains unknown. This study is aimed at unveiling the characterization of the transcriptional expression profiling of LIGHT in glioma. We selected 301 glioma patients with mRNA microarray data from the CGGA dataset and 697 glioma patients with RNAseq data from the TCGA dataset. Transcriptome data and clinical data of 998 samples were analyzed. Statistical analyses and figure generation were performed with R language. LIGHT expression showed a positive correlation with WHO grade of glioma. LIGHT was significantly increased in mesenchymal molecular subtype. Gene Ontology analysis demonstrated that LIGHT was profoundly involved in immune response. Moreover, LIGHT was found to be synergistic with various immune checkpoint members, especially HVEM, PD1/PD-L1 pathway, TIM3, and B7-H3. To get further understanding of LIGHT-related immune response, we put LIGHT together with seven immune signatures into GSVA and found that LIGHT was particularly correlated with HCK, LCK, and MHC-II in both datasets, suggesting a robust correlation between LIGHT and activities of macrophages, T-cells, and antigen-presenting cells (APCs). Finally, higher LIGHT indicated significantly shorter survival for glioma patients. Cox regression models revealed that LIGHT expression was an independent variable for predicting survival. In conclusion, LIGHT was upregulated in more malignant gliomas including glioblastoma, IDH wildtype, and mesenchymal subtype. LIGHT was mainly involved in the immune function of macrophages, T cells, and APCs and served as an independent prognosticator in glioma.
Article
Full-text available
Mesenchymal stem cells (hMSCs) are multipotent stem cells that have the capacity to differentiate into various lineages. These cells provide stromal support and can be utilized as a feeder layer for expansion of hematopoitic stem cells and embryonic stem cells. Furthermore, allo-transplanted MSCs are not rejected and have been shown to mediate immuno-modulatory functions in vitro. Also, MSCs have been found at the wound site at extended times. The mechanisms underlying MSC migration and immuno-modulation are still under investigation. Aim: To understand the factors involved in human MSC (hMSC) migration and their interaction with various immune cell types. Methods: Human MSCs were examined for the presence of cell surface receptors that may play a role in migration using quantitative RT-PCR. Next, hMSCs were co-cultured with purified immune cell types including dendritic cells (DCs), naïve T cells and NK cells. Following the co-culture, changes in the phenotype of the immune cells under activating conditions were analyzed using ELISA and functional assays. Results: Human MSCs express Toll receptors, especially TLR4, on their cell surface. The TLR4 on hMSCs is functional as seen by a several-fold increase in IL-6 and chemokine IL-8 upon incubation with TLR4 exogenous ligand lipopolysaccharide (LPS) and the endogenous ligand, soluble hyaluronic acid (sHA). When hMSCs were incubated with activated dendritic cells, there was a >50% decrease in TNF-α secretion and a >50% increase in IL-10 secretion. When hMSCs were incubated with naïve T cells, hMSCs decreased IFN-γ secretion and increased IL-4 secretion. Decreased IFN-γ was also seen when MSCs were incubated with NK cells. Conclusion: These results suggest that (i) hMSCs may respond to the signals generated by breakdown products of extracellular matrix (e.g. sHA) via TLR4 and assist in wound healing (ii) hMSCs immuno-modulatory effects are mediated by interacting with various immune cell types and altering their phenotypic response to a more tolerant and anti-inflammatory response.
Article
Full-text available
Despite evidence about the implication of the bone marrow (BM) stromal microenvironment in multiple myeloma (MM) cell growth and survival, little is known about the effects of myelomatous cells on BM stromal cells. Mesenchymal stromal cells (MSCs) from healthy donors (dMSCs) or myeloma patients (pMSCs) were co-cultured with the myeloma cell line MM.1S, and the transcriptomic profile of MSCs induced by this interaction was analyzed. Deregulated genes after co-culture common to both d/pMSCs revealed functional involvement in tumor microenvironment cross-talk, myeloma growth induction and drug resistance, angiogenesis and signals for osteoclast activation and osteoblast inhibition. Additional genes induced by co-culture were exclusively deregulated in pMSCs and predominantly associated to RNA processing, the ubiquitine-proteasome pathway, cell cycle regulation, cellular stress and non-canonical Wnt signaling. The upregulated expression of five genes after co-culture (CXCL1, CXCL5 and CXCL6 in d/pMSCs, and Neuregulin 3 and Norrie disease protein exclusively in pMSCs) was confirmed, and functional in vitro assays revealed putative roles in MM pathophysiology. The transcriptomic profile of pMSCs co-cultured with myeloma cells may better reflect that of MSCs in the BM of myeloma patients, and provides new molecular insights to the contribution of these cells to MM pathophysiology and to myeloma bone disease.
Article
Full-text available
Multiple myeloma (MM) is a hematologic malignancy of differentiated plasma cells that accumulates and proliferates in the bone marrow. MM patients often develop bone disease that results in severe bone pain, osteolytic lesions, and pathologic fractures. These skeletal complications have not only a negative impact on quality of life but also a possible effect in overall survival. MM osteolytic bone lesions arise from the altered bone remodeling due to both increased osteoclast activation and decreased osteoblast differentiation. A dysregulated production of numerous cytokines that can contribute to the uncoupling of bone cell activity is well documented in the bone marrow microenvironment of MM patients. These molecules are produced not only by malignant plasma cells, that directly contribute to MM bone disease, but also by bone, immune, and stromal cells interacting with each other in the bone microenvironment. This review focuses on the current knowledge of MM bone disease biology, with particular regard on the role of bone and immune cells in producing cytokines critical for malignant plasma cell proliferation as well as in osteolysis development. Therefore, the understanding of MM pathogenesis could be useful to the discovery of novel agents that will be able to both restore bone remodelling and reduce tumor burden.
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
Costimulatory molecules on antigen-presenting cells (APCs) play an important role in T cell activation and expansion. However, little is known about the surface molecules involved in direct T-T cell interaction required for their activation and expansion. LIGHT, a newly discovered TNF superfamily member (TNFSF14), is expressed on activated T cells and immature dendritic cells. Here we demonstrate that blockade of LIGHT activity can reduce anti-CD3–mediated proliferation of purified T cells, suggesting that T cell–T cell interaction is essential for this proliferation. To test the in vivo activity of T cell–derived LIGHT in immune homeostasis and function, transgenic (Tg) mice expressing LIGHT in the T cell lineage were generated. LIGHT Tg mice have a significantly enlarged T cell compartment and a hyperactivated peripheral T cell population. LIGHT Tg mice spontaneously develop severe autoimmune disease manifested by splenomegaly, lymphadenopathy, glomerulonephritis, elevated autoantibodies, and severe infiltration of various peripheral tissues. Furthermore, the blockade of LIGHT activity ameliorates the severity of T cell–mediated diseases. Collectively, these findings establish a crucial role for this T cell–derived costimulatory ligand in T cell activation and expansion; moreover, the dysregulation of T cell–derived LIGHT leads to altered T cell homeostasis and autoimmune disease. Elevation of autoantibodies both serves as a criterion for the clinical diagnosis of autoimmune disease and has been shown to be characteristic of MRL-lpr/lpr mice (48). We therefore tested the serum of LIGHT Tg mice and control littermates by ELISA for autoantibodies. LIGHT Tg mice demonstrated anti-DNA autoantibody levels elevated up to eightfold from those of the control littermates, whereas the level of total IgG in Tg mice was only slightly increased compared with the level in control littermates (Figure 6i). Tg mice also displayed elevated levels of rheumatoid factors, another commonly detected autoantibody in chronic inflammation and autoimmune diseases (Figure 6j). The findings of lupuslike glomerulonephritis and increased inflammatory cell infiltrate in multiple organs, along with elevations of serum autoantibodies, indicated the establishment of autoimmunity in LIGHT Tg mice. Therefore, the overproliferation and hyperactivation of T cells mediated by T cell–derived LIGHT resulted in the breakdown of B cell tolerance, supporting the notion that the dysregulation of LIGHT expression may be a critical element in the induction of both T and B cell autoimmunity and in the pathogenesis of autoimmune diseases.
Article
The monoclonal gammopathies are a group of disorders associated with monoclonal proliferation of plasma cells. The characterization of specific entities is an area of difficulty in clinical practice. The International Myeloma Working Group has reviewed the criteria for diagnosis and classification with the aim of producing simple, easily used definitions based on routinely available investigations. In monoclonal gammopathy of undetermined significance (MGUS) or monoclonal gammopathy, unattributed/unassociated (MG[u]), the monoclonal protein is < 30 g/1 and the bone marrow clonal cells < 10% with no evidence of multiple myeloma, other B-cell proliferative disorders or amyloidosis. In asymptomatic (smouldering) myeloma the M-protein is ≥ 30 g/1 and/or bone marrow clonal cells ≥ 10% but no related organ or tissue impairment (ROTI)(end-organ damage), which is typically manifested by increased calcium, renal insufficiency, anaemia, or bone lesions (CRAB) attributed to the plasma cell proliferative process. Symptomatic myeloma requires evidence of ROTL Non-secretory myeloma is characterized by the absence of an M-protein in the serum and urine, bone marrow plasmacytosis and ROTI. Solitary plasmacytoma of bone, extramedullary plasmacytoma and multiple solitary plasmacytomas (± recurrent) are also defined as distinct entities. The use of these criteria will facilitate comparison of therapeutic trial data. Evaluation of currently available prognostic factors may allow better definition of prognosis in multiple myeloma.
Article
Surface lymphotoxin (LT) is a heteromeric complex of LT-alpha and LT-beta chains that binds to the LT-beta receptor (LT-beta-R), a member of the tumor necrosis factor (TNF) family of receptors. The biological function of this receptor-ligand system is poorly characterized. Since signaling through other members of this receptor family can induce cell death, e.g., the TNF and Fas receptors, it is important to determine if similar signaling events can be communicated via the LT-beta-R. A soluble form of the surface complex was produced by coexpression of LT-alpha and a converted form of LT-beta wherein the normally type II LT-beta membrane protein was changed to a type I secreted form. Recombinant LT-alpha 1/beta 2 was cytotoxic to the human adenocarcinoma cell lines HT-29, WiDr, MDA-MB-468, and HT-3 when added with the synergizing agent interferon (IFN) gamma. When immobilized on a plastic surface, anti-LT-beta-R monoclonal antibodies (mAbs) induced the death of these cells, demonstrating direct signaling via the LT-beta-R. Anti-LT-beta-R mAbs were also identified that inhibited ligand-induced cell death, whereas others were found to potentiate the activity of the ligand when added in solution. The human WiDr adenocarcinoma line forms solid tumors in immunocompromised mice, and treatment with an anti-LT-beta-R antibody combined with human IFN-gamma arrested tumor growth. The delineation of a biological signaling event mediated by the LT-beta-R opens a window for further studies on its immunological role, and furthermore, activation of the LT-beta-R may have an application in tumor therapy.
Article
The aim of the study was to investigate the expression of MIP-1 alpha and sclerostin in bone marrow of patients with multiple myeloma (MM), the possible association of the sclerostin and MIP-1 alpha with MBD and the clinical characteristics. 53 patients (29 M, 24 F), median age 64 years were studied. MIP-1 alpha, sclerostin and bone-specific alkaline phosphatase (bALP) levels were quantified using an enzyme-linked immunosorbent assay (ELISA). Sclerostin and MIP-1 alpha mRNA expression was determined by RT-PCR. PTH and 1, 25(OH) 2D3 levels were measured with an electrochemiluminescence immunoassay. The sclerostin and MIP-1 alpha concentrations in patients with MM were higher than those in the controls. RT-PCR analysis verified that the bone marrow mononuclear cells (BMMNCs) of most patients showed sclerostin and MIP-1 alpha mRNA expression. The sclerostin and MIP-1 alpha levels in patients with ISS stage III disease were significantly higher than those in patients with ISS stage II disease(p = 0.01 and 0.06).The sclerostin and MIP-1 alpha levels in patients with BMD in group C were significantly higher than those in group A + B. There was positive correlation between sclerostin levels and MIP-1 alpha, beta2-microglobulin and aCa levels. A negative association was seen between sclerostin levels and bALP, HB and ALB levels. The MM patients with high sclerostin levels (>0.72 ng/ml) had significantly shorter median survival than those with low sclerostin levels (≤0.72 ng/ml)(χ2 = 7.574, p = 0.006).Our findings support the positive relationship between sclerostin levels and MIP-1alpha levels deserves further detailed research.a
Article
Multiple myeloma (MM) lytic bone disease (LBD) is caused by osteoclast activation and osteoblast inhibition. RANK/RANKL/OPG play central roles in osteoclast activation and Wnt inhibitor DKK1 in osteoblast inhibition. The role of other Wnt inhibitors is less clear. We evaluated gene expression of osteoclast regulators (RANK, RANKL, OPG, TRAIL, MIP1A), Wnt inhibitors (DKK1, SFRP2, SFRP3, sclerostin, WIF1) and osteoblast transcription factors (RUNX2, osterix) by quantitative reverse transcriptase polymerase chain reaction (RT-PCR) in the bone marrow (BM) microenvironment using snap-frozen BM biopsies, thereby achieving minimal post-sampling manipulation, and gene expression profiling (GEP) data, reflecting the in vivo situation. We analyzed 110 biopsies from newly diagnosed patients with MM and monoclonal gammopathy of unknown significance (MGUS) and healthy volunteers. LBD was evaluated using standard radiographs and the bone resorption marker CTX-1. Protein levels were evaluated by enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry. Among Wnt inhibitors, only SFRP3 and DKK1 were significantly overexpressed in advanced LBD, correlating with protein levels. SFRP3 correlated with CTX-1. Our findings support osteoblast inhibition as the driving force behind MM LBD.