You are on page 1of 14

Original Article

Toxicologic Pathology
1-14

Bone and the Immune System ª The Author(s) 2017


Reprints and permission:
sagepub.com/journalsPermissions.nav
DOI: 10.1177/0192623317735316
journals.sagepub.com/home/tpx

M. Neale Weitzmann1,2

Abstract
Osteoporosis increases fracture risk, a cause of crippling morbidity and mortality. The immunoskeletal interface (ISI) is a cen-
tralization of cell and cytokine effectors shared between skeletal and immune systems. Consequently, the immune system mediates
powerful effects on bone turnover. Physiologically, B cells secrete osteoprotegerin (OPG), a potent anti-osteoclastogenic factor
that preserves bone mass. However, activated T cells and B cells secrete pro-osteoclastogenic factors including receptor activator
of Nuclear factor-kappaB (NF-kB) ligand (RANKL), Interleukin (IL)-17A, and tumor necrosis factor (TNF)-a promoting bone loss in
inflammatory states such as rheumatoid arthritis. Recently, ISI disruption has been linked to osteoporosis in human immunode-
ficiency virus (HIV) infection/acquired immunodeficiency syndrome (AIDS), where elevated B cell RANKL and diminished OPG
drive bone resorption. HIV-antiretroviral therapy paradoxically intensifies bone loss during disease reversal, as immune recon-
stitution produces osteoclastogenic cytokines. Interestingly, in estrogen deficiency, activated T cells secrete RANKL, TNF, and IL-
17A that amplify bone resorption and contribute to postmenopausal osteoporosis. T cell–produced TNF and IL-17A further
contribute to bone loss in hyperparathyroidism, while T cell production of the anabolic Wingless integration site (Wnt) ligand,
Wnt10b, promotes bone formation in response to anabolic parathyroid hormone and the immunomodulatory costimulation
inhibitor cytotoxic T lymphocyte–associated protein-4-IgG (abatacept). These findings provide a window into the workings of the
ISI and suggest novel targets for future therapeutic interventions to reduce fracture risk.

Keywords
bone, immunoskeletal interface, T cell, B cell, RANKL, osteoprotegerin, osteoimmunology

Osteoporosis (Latin for porous bone) is a common malady of 2012), and the worldwide incidence of fracture is expected to
the skeleton and ensues when the rate of osteoclastic bone rise to 6 million by 2050 (Cummings and Melton 2002) with
resorption outpaces the rate of osteoblastic bone formation, costs projected to reach US$25 billion in the United States
conditions that predispose to bone loss. From a clinical perspec- alone by 2025 (Burge et al. 2007).
tive, osteoporosis is most commonly defined as a bone mineral An interesting and unexpected finding that has emerged in
density (BMD) T score of less than or equal to 2.5 standard the field of bone biology is the repurposing by the skeleton of
deviations from a young adult reference range, following a bone key immune elements including adaptive and innate immune
densitometry scan using dual-energy X-ray absorptiometry cells and key immune-derived cytokines. As a result, physio-
(DXA; Unnanuntana et al. 2010). From a biological standpoint logical functions of the adaptive immune system beneficially
and at the molecular level, osteoporotic bone is characterized by regulate the skeleton. However, under states of pathologic
a loss of BMD and structural integrity, leading to a conforma- immune dysfunction such as immunodeficiency or inflamma-
tion with diminished load-bearing capacity and with an tory responses to infection/disease, the skeleton is at the mercy
increased propensity to fracture. of the immune response and serious collateral damage to bone
The consequences of fragility fractures can be devastating may result leading to osteoporosis and an elevated risk for bone
and are commonly associated with crippling disability that
necessitates intensive rehabilitation in as many as 75% of
cases. In the case of hip fractures, mortality rates of 24% to 1
Department of Veterans Affairs, Atlanta VA Medical Center, Decatur,
33% are common in the first year following a fracture (Bass Georgia, USA
et al. 2007; Johnell and Kanis 2006; Lewis et al. 2006). The 2
Department of Medicine, Division of Endocrinology and Metabolism and
associated loss of independence during and often beyond, the Lipids, Emory University School of Medicine, Atlanta, Georgia, USA
fracture rehabilitation period, can lead to a marked decline in
quality of life especially in aged fracture victims (Kates, Kates, Corresponding Author:
M. Neale Weitzmann, Department of Medicine, Division of Endocrinology and
and Mendelson 2007; Johnell and Kanis 2006). It is estimated Metabolism and Lipids, Emory University School of Medicine, 101 Woodruff
that almost 50% of women and 30% of men over the age of 50 Circle, 1305 WMB, Atlanta, GA 30322, USA.
will suffer a fragility fracture due to osteoporosis (Eisman et al. Email: mweitzm@emory.edu
2 Toxicologic Pathology XX(X)

fracture (Weitzmann and Ofotokun 2016; Weitzmann and is the receptor for the key osteoclastogenic cytokine RANK
Pacifici 2006). Although bone loss is a natural consequence ligand (RANKL; Simonet et al. 1997; Anderson et al. 1997;
of aging, it is exacerbated by many common pathological Wong et al. 1997), which upon binding to RANK in the presence
conditions that afflict many people. This includes inflamma- of the trophic factor, macrophage colony-stimulating factor,
tory diseases such as rheumatoid arthritis (RA), periodontal causes the osteoclast precursor to differentiate into a preosteo-
infection, and inflammatory bowel diseases including Crones clast (Lacey et al. 1998). These preosteoclasts further fuse
disease. The immunodeficiency state resulting from HIV infec- together to form giant multinucleated bone resorbing mature
tion further promotes bone resorption. Another key protagonist osteoclasts. As with many biological pathways, there are checks
of exacerbated skeletal decline is the result of estrogen decline and balances, and in this pathway, this takes the form of a soluble
following the menopause (postmenopausal osteoporosis). RANKL decoy receptor called osteoprotegerin (OPG; Lacey
These bone loss states are all characterized by an increase in et al. 1998; Tsuda et al. 1997). OPG binds to RANKL and pre-
osteoclastic bone resorption relative to osteoblastic bone for- vents it from associating with RANK, thus reducing the rate of
mation, leading to net bone loss (Weitzmann and Ofotokun osteoclast differentiation and bone resorption. As RANKL and
2016; Weitzmann and Pacifici 2006). OPG are the final downstream effectors of osteoclastogenesis,
Although it has long been recognized that osteoclast precur- the ratio of RANKL to that of OPG in the bone marrow micro-
sors derive from the monocyte/macrophage lineage (Walker environment is a key determinant of the rate of osteoclastic bone
1972, 1975; Buring 1975), cells of immune origin, in the past resorption occurring in humans and animals (Teitelbaum 2000).
2 decades, the new field of “osteoimmunology” has blossomed, This pathway is summarized diagrammatically in Figure 1.
revealing a deeply integrated immunoskeletal interface (ISI) in It is further recognized that the role of inflammatory cytokines
which immune cells direct physiological and pathological bone in promoting osteoclastogenesis is indirect and mediated through
resorption and bone formation (Weitzmann and Ofotokun the regulation of the RANK/RANKL/OPG axis. For example,
2016; Weitzmann and Pacifici 2006). tumor necrosis factor alpha (TNF), a potent inflammatory and
Although many of the concepts described below have been indeed osteoclastogenic cytokine, drives up bone resorption by
uncovered using animal models, predominantly mice and rats, promoting RANK expression on monocytes thus converting them
tantalizing evidence is beginning to emerge to support a role into osteoclast precursors, upregulates RANKL expression by
for the immune response in human bone diseases. It should be osteoblast lineage cells (Hofbauer et al. 1999), and down mod-
pointed out, however, that osteoimmunology does not replace ulates osteoblastic production of OPG. Although other inflamma-
traditional bone biology. In most cases, the ISI simply adds a tory cytokines such as IL-1, IL-6, IL-7, and Interleukin (IL)-17A
new layer to the underlying principles already elucidated. also upregulate osteoclastogenesis by regulating RANK,
The following review outlines some of the past and ongoing RANKL, and/or OPG production, TNF mediates 1 other unique
investigations by our group and others into the role of the ISI in function making it an extremely potent osteoclastogenic factor.
the regulation of the skeleton. TNF synergizes with RANKL at the signal transduction level to
further intensify osteoclastogenesis and bone resorption (Cenci
et al. 2000; Lam et al. 2000; Zhang et al. 2001; Fuller et al. 2002).
Osteoclasts and the Receptor Activator of
Nuclear factor-kappaB (NF-kB) (RANK)/ The ISI
Receptor Activator of NF-kB Ligand
Although long recognized that osteoclasts derive from cells of the
(RANKL)/Osteoprotegerin (OPG) System monocyte lineage (immune cells), an unexpected finding was the
Although osteoclasts are the sole bone resorbing cells of the existence of a deeply rooted nexus between the immune and ske-
body and increased osteoclastic bone resorption underlies letal systems, the ISI. This is the result of a centralization of mul-
bone loss and osteoporosis development, it is only compara- tiple cells of the adaptive immune response and their cytokine
tively recently that the process underlying osteoclastogenesis effectors, which serve dual functions in the regulation of the ske-
has been unveiled. It has long been recognized that osteoclast leton. One key aspect of this involves activated lymphocytes, both
precursors are significantly more frequent in inflammatory T- and B cells, which secrete RANKL and TNF under inflamma-
conditions in both humans and animals, particularly when tory conditions, driving up osteoclast formation and bone resorp-
inflammatory cytokines are elevated (Pacifici et al. 1991; tion in inflammatory states including the autoimmune disease RA,
Kitazawa et al. 1994; Kimble et al. 1996). However, adding inflammatory bowel diseases including Crohn’s disease, and per-
inflammatory cytokines to osteoclast precursors is under most iodontal infection. Consequently, these conditions are all associ-
circumstances ineffective in promoting osteoclast formation ated with bone loss (Weitzmann and Ofotokun 2016).
suggesting an alternative mechanism. In contrast to these inflammatory states, under physiological
In 1997 and 1998, a series of key discoveries unveiled a conditions, B cells are a significant source of the osteoclast
previously unknown pathway for stimulating osteoclast forma- inhibitor OPG. Early studies recognized that human tonsil B
tion (Simonet et al. 1997). It is now evident that what differenti- cells secrete OPG and that in vitro activation of cluster of
ates an osteoclast precursor from a regular monocyte is differentiation (CD) 40 costimulatory pathway on B cells fur-
expression on its surface of a specific molecule, RANK. RANK ther upregulates OPG production (Yun et al. 1998). We later
Weitzmann 3

Figure 1. Osteoclastogenesis and the receptor activator of NF-kB (RANK)/receptor activator of NF-kB ligand (RANKL)/osteoprotegerin (OPG)
axis. Osteoclast precursors derive from the monocyte lineage and express the RANK that is a receptor for the key osteoclastogenic cytokine
RANKL. RANKL, in the presence of permissive concentrations of the tropic factor, macrophage colony-stimulating factor (M-CSF), causes
differentiation of osteoclast precursors into preosteoclasts that fuse together to form mature multinucleated bone resorbing osteoclasts. RANKL
activity is moderated by a soluble decoy receptor OPG. Inflammatory cytokines promote osteoclastogenesis and bone resorption by stimulating
RANK and/or increasing production of RANKL and/or suppressing expression of OPG.

ratified these data in the murine system where we demonstrated conditions that disrupt adaptive immunity would lead to an
that in mouse bone marrow, the B cell lineage contributed 64% imbalance in bone turnover promoting osteoclast formation,
of the total OPG production (Y. Li, Toraldo, et al. 2007). Con- increased bone resorption and bone loss. A test of this predic-
sistent with these data, B cell knockout (KO) mice were found tion involves infection by the HIV-1 virus that devastates adap-
to have a bone marrow deficit in OPG, an enhanced rate of tive immunity leading to AIDS. HIV dramatically depletes
osteoclastic bone resorption, and significantly diminished CD4þ T cells, the lynchpin of adaptive immunity, leading to
BMD and bone mass. Reconstitution of B cells into young B a decline in number and function of other adaptive immune
cell null mice rescued these alterations to the ISI and prevented components including B cells and CD8þ T cells.
bone loss (Y. Li, Toraldo, et al. 2007). In fact, bone loss is widespread in human subjects infected
As CD40 on B cells functions as a receptor for CD40 ligand by HIV and a meta-analysis has concluded that overall there is
(CD40L) expressed predominantly by activated T cells, we fur- a 50% to 70% incidence of osteopenia and 15% incidence of
ther investigated a role for T cell CD40/CD40L costimulation osteoporosis in this population (Brown and Qaqish 2006).
and found that deletion of CD40 or CD40L or of T cells also led Importantly, this loss of BMD has been demonstrated in several
to diminished B cell OPG production and bone loss (Y. Li, large population-based studies to translate into a significantly
Toraldo, et al. 2007). elevated prevalence of bone fracture (Prior et al. 2007; Triant
Taken together, these data demonstrate that under inflam- et al. 2008; Young et al. 2011; Womack et al. 2011; Guerri-
matory conditions B cells and T cells damage the skeleton Fernandez et al. 2013; Prieto-Alhambra et al. 2014; Sharma
though copious secretion of RANKL and inflammatory cyto- et al. 2015). In fact, both men and women are affected and with
kines such as TNF. By contrast, under basal conditions, B cells, fracture prevalence affected over a wide age range, such that in
regulated by costimulatory actions through T cells, protect the the context of HIV, even relatively younger men are at
skeleton by promoting production of OPG. This pathway is increased risk of a fragility fracture (Triant et al. 2008).
summarized diagrammatically in Figure 2. The mechanisms underlying bone loss and increased fracture
incidence have been difficult to assess in humans, given a sig-
nificant number of coexisting osteoporosis risk factors in this
The Bone Loss of HIV-1 Infection demographic. These include metabolic complications of AIDS
Given the existence of the ISI and the protective role of such as hypogonadism and renal disease and lifestyle factors
immune cells on basal bone turnover, one might predict that including high rates of smoking and alcohol and recreational
4 Toxicologic Pathology XX(X)

Figure 2. The immunoskeletal interface (ISI). The ISI is a nexus between immune and skeletal systems with key immune components repurposed
for skeletal functions. Under basal physiological conditions, B cell, under the regulation of T cells, secretes copious concentrations of the receptor
activator of NF-kB ligand (RANKL) decoy receptor osteoprotegerin (OPG), moderating osteoclastogenesis and maintaining homeostatic bone
turnover. Under pathological conditions such as inflammation, activated B cells and T cells secrete large concentrations of RANKL, driving up
osteoclastogenesis, disrupting basal bone homeostasis, and leading to bone loss.

drug use, which can all impact the skeleton. Finally, complex HIV Tg rats. As many cell types have the capacity to secrete
invasive mechanistic studies in humans are severely limited in both OPG and RANKL, we next investigated production of these
scope, ultimately necessitating the use of an animal model. The factors by specific cell populations. Immunomagnetic isolation
animal model we chose for these studies was the HIV transgenic of B cells followed by RT-PCR confirmed that the changes in
(Tg) rat, a small animal model of HIV infection that develops RANKL and OPG observed were exclusively a consequence of
multiple clinical manifestations of human AIDS (Reid et al. altered B cell function (Vikulina et al. 2010).
2001). To interrogate changes in the skeleton, we performed To determine whether these changes in B cell production of
bone densitometry to quantify BMD using DXA. Tg rats dis- RANKL and OPG are representative of human HIV, we per-
played significantly diminished BMD at lumbar vertebrae and formed a translational clinical study in which we recruited 58
femurs and in both trabecular and cortical compartments as HIV-negative control subjects and 62 HIV-positive patients
assessed by microcomputed tomography (μCT; Vikulina et al. and collected peripheral blood mononuclear cells and
2010). Biochemical markers of in vivo bone resorption further plasma/serum (Titanji et al. 2014). As expected, we identified
identified a significant increase in the bone resorption marker C- a significant elevation in bone resorption marker (CTx) in HIV-
terminal telopeptide of type I collagen (CTx), a sensitive and infected subjects compared to uninfected controls. Flow cyto-
specific marker of bone resorption, while osteocalcin, a marker metric analysis of peripheral blood B cells further ratified a
of bone formation, was unchanged. Histological and histomor- significant 20% decline in the percentage of OPG producing
phometric analysis of tartrate resistant acid phosphatase positive B cells and a significant 60% increase in the percentage of B
multinucleated cells (osteoclasts) confirmed a significant cells expressing RANKL in conditions of HIV infection.
increase in osteoclast number and the area of bone surface cov- Although cells in the peripheral circulation may not accurately
ered by osteoclasts in HIV Tg rats. To understand the mechan- represent conditions in the bone microenvironment and may
istic basis for increased osteoclasts, we next examined the only provide muted evidence of changes at other sites, overall
expression of RANKL and OPG in the bone marrow by real- the data strongly support an altered B cell RANKL/OPG ratio
time reverse transcriptase (RT)-polymerase chain reaction in HIV-infected patients. Importantly, we further identified a
(PCR) analysis of whole bone marrow and observed a significant significant inverse correlation between BMD at the femoral
elevation in expression of RANKL and a simultaneous signifi- neck and total hip with the B cell RANKL/OPG ratio, suggest-
cant decline in OPG. These data suggest an imbalance in the ing a pathophysiological effect of B cells on BMD (Titanji
RANKL/OPG ratio may underlie the osteoclastic bone loss in et al. 2014).
Weitzmann 5

Figure 3. The bone loss of human immunodeficiency virus (HIV) infection. HIV infection leads to a disruption of the immunoskeletal interface.
Damage to adaptive immune components, especially CD4þ T cells, leads to a loss of costimualtion and changes in cytokine production, causing a
significant decline in B cell osteoprotegerin (OPG) production and a significant upswing in production of receptor activator of NF-kB ligand
(RANKL). This RANKL/OPG imbalance drives up osteoclastogenesis and bone resorption, leading to bone loss.

Although bone loss in HIV-infected subjects is likely multi- than others (particularly tenofovir disoproxil fumarate contain-
factorial and exacerbated by AIDS sequelae and lifestyle fac- ing regimens; Wohl et al. 2016), it is now recognized that
tors, we propose that 1 major underlying cause is an virtually all ART mediate some degree of bone loss, and recent
immunoskeletal defect in B cell RANKL and OPG production. evidence supports the contention that much, if not most, of the
HIV infection likely results in significant alterations in CD4þ T bone loss is independent of the specific antiviral agents used
cell costimulation, a consequence of severe CD4 depletion as (Brown et al. 2009; Piso et al. 2011; Bruera et al. 2003).
well as functional changes. Irrespective of the underlying dis- How ART affects the skeleton is unclear and has traditionally
turbance, B cell function changes from that of OPG production been considered to be a direct toxic effect on bone cells such as
to that of RANKL production (an activated condition). The osteoblasts increasing RANKL production. However, studies of
increase in free bioactive RANKL thus increases osteoclast antiretroviral drugs in culture systems (Gibellini et al. 2010;
differentiation, driving up bone resorption and leading to bone Grigsby, Pham, Gopalakrishnan, et al. 2010; Grigsby, Pham,
loss. This model is presented diagrammatically in Figure 3. Mansky, et al. 2010) or in vivo in mice (Wang et al. 2004)
generally fail to recapitulate the effects on bone turnover that
are observed when administered to humans with HIV infection.
The Bone Loss of HIV Antiretroviral
Furthermore, recent studies in which ART is used to treat human
Therapy (ART) hepatitis B virus infection cause little bone loss compared to
Years (often decades) of undiagnosed HIV infection drive a HIV-infected subjects (Chan et al. 2016). Taken together, the
significant deterioration of the skeleton. Once diagnosed, how- data do not support a direct toxic action of ART on bone cells
ever, the majority of patients begin ART also known as highly and suggest a mechanism unique to HIV infection.
active antiretroviral therapy and combinatorial antiretroviral What all HIV agents have in common is that they suppress
therapy. In most cases, ART comprises a cocktail of multiple viral replication and reduce viral load. As a consequence,
agents from different drug classes. Modern ART drug combi- adaptive immune function undergoes, in part, reconstitution
nations are extremely effective at reducing HIV viral load and and reactivation. The recovery of CD4þ T cells involves
reversing many of the classic manifestations of AIDS. homeostatic reconstitution, a process involving T cell prolif-
Paradoxically, the skeleton does not improve with ART and eration and expansion to fill available immunological space
in fact, in most cases, actually undergoes a further deterioration (Ernst et al. 1999; Surh and Sprent 2000). Similar processes
with an additional average loss of up to 6% in BMD within the are involved in CD8þ T cell and B cell recovery and are
first 1 to 2 years of therapy initiation (McComsey et al. 2010). driven in part through cytokine-mediated processes. Through
Although specific ART classes and indeed specific drugs costimulatory interactions and cytokine production, CD4þ T
within those classes appear to mediate more dramatic bone loss cell subsets further regulate other adaptive immune
6 Toxicologic Pathology XX(X)

Figure 4. The bone loss of human immunodeficiency virus (HIV) antiretroviral therapy (ART). HIV ART significantly diminishes viral load allowing
a partial CD4þ T cell recovery. CD4þ T cells undergo homeostatic expansion and repopulation of immunological niches. This process is
inflammatory and leads to the release of receptor activator of NF-kB ligand (RANKL) and tumor necrosis factor (TNF) driving up osteoclasto-
genesis. In addition, reactivation of adaptive immune function causes additional RANKL and/or TNF production by APC, CD8þ T cells, and B cells.
These high levels of osteoclastogenic cytokines promote elevated osteoclastogenesis and stimulate bone resorption, leading to additional bone
loss over and above that causes by HIV infection itself.

components including humoral immunity (B cells) and anti- only partly diminished bone loss, consistent with TNF produc-
gen presenting cells including macrophages, dendritic cells, tion by not only T cells but also other adaptive immune cells
and B cells. The regeneration and rekindling of adaptive (Ofotokun et al. 2015).
immunity thus has the potential to produce inflammatory Taken together, the data confirm that homeostatic repopula-
events that may have the capacity to drive osteoclastogenesis tion of T cells leads to an inflammatory environment capable of
and bone loss, as is characteristic of other inflammatory states driving significant bone resorption and loss of BMD and mass.
(Kotake et al. 2001; P. Li and Schwarz 2003). This model is presented diagrammatically in Figure 4.
To test this hypothesis, we mimicked the process of T cell Whether this mechanism is a significant contributor to bone
repopulation following ART in humans by reconstituting T loss in humans initiating ART remains to be determined,
cells by syngeneic adoptive transfer into T cell deficient TCRb although in support of this mechanism, recent clinical data
KO mice and quantified bone turnover and structure over by us (Ofotokun et al. 2016) and others (Grant et al. 2013)
3 months (Ofotokun et al. 2015). Using prospective DXA to reveal that subjects with low starting CD4þ T cell number and
quantify BMD, we found a dramatic loss of bone mass in those undergoing the most robust T cell reconstitution (Ofoto-
reconstituted mice, as T cells homeostatically expanded and kun et al. 2016) undergo to most aggressive bone loss.
engaged adaptive immunity. μCT further revealed significant A translational clinical study to validate homeostatic recon-
deterioration of both cortical and trabecular bone compart- stitution and immune activation as a key mechanism underlying
ments. A marker of bone resorption (CTx) revealed signifi- ART bone loss is presently in progress.
cantly increased bone resorption and serum levels of RANKL
and TNF were found to be significantly elevated, similarly to
Estrogen Deficiency Bone Loss and the Role
human HIV patients initiating ART (Ofotokun et al. 2016).
Using flow cytometry of whole bone marrow and/or spleen, of the ISI
we further demonstrated that both CD4þ and CD8þ T cells Postmenopausal osteoporosis is the archetypal bone disease of
produced significantly increased levels of RANKL, while mul- women and is the result of an imbalance in bone turnover as a
tiple adaptive immune components including CD4þ and CD8þ consequence of estrogen deficiency following the menopause
T cells, B cells, and macrophages all produced significant lev- (Weitzmann and Pacifici 2006). As in other osteoporotic con-
els of TNF. Reconstitution of T cells from RANKL KO mice ditions, estrogen deficiency leads to an enhanced rate of bone
led to significant protection from bone loss, suggesting a key resorption relative to bone formation, leading to net bone loss.
role of T cell RANKL while transplantation of TNF KO T cells Ovariectomy in mice or rats is a traditional animal model of
Weitzmann 7

postmenopausal osteoporosis, as removal of the ovaries causes immunosuppressive effects in part, through induction of regu-
a rapid drop in estrogen levels that leads to increased bone latory T cells (Tregs) that down modulate T cell activation.
resorption and significant cortical and trabecular bone loss This model is presented diagrammatically in Figure 5.
within a period of just 2 to 4 weeks (Weitzmann et al. 2002). A perplexing aspect of ovariectomy-induced bone loss is the
Estrogen is established to mediate potent anti-inflammatory apparent antigen-dependent nature of T cell activation in this
effects in the body, and loss of estrogen has been shown to response. To validate a need for antigens, we performed ovar-
cause significant expansion of lymphocytes, both T cells iectomy in mice with silenced antigen presentation due to a
(Cenci et al. 2003) and B cells (Miyaura et al. 1997). To test transgenic T cell receptor in all T cells, responsive only to
whether T cell expansion following estrogen decline promotes ovalbumin, a protein not endogenously present in mice (Cenci
bone loss as in other inflammatory conditions, we performed et al. 2003). In the absence of antigen presentation, these mice
ovariectomy in T cell deficient nude mice (Cenci et al. 2000). were fully protected from ovariectomy-induced bone loss.
While control Wild Type (WT) mice underwent significant loss However, after exogenous administration of antigen (ovalbu-
of BMD due to increased bone resorption, nude mice were min), robust bone loss was again elicited by estrogen defi-
protected from osteoclastic bone loss. Although activated T ciency. These data confirmed the requirement for antigen
cells are a significant source of RANKL, this cytokine is not presentation in this response but also suggested that no specific
typically observed to be increased in T cells under conditions of type of antigen was needed and that any antigen capable of
estrogen deficiency in mice. By contrast, TNF is significantly activating a T cell might suffice. Although a low baseline of
increased in the bone marrow, a consequence of T cells (Cenci antigen presentation is always occurring even in healthy
et al. 2000). Although TNF production per cell was not humans and animals, recently, the likely nature of the antigens
increased, the expansion of T cells in bone marrow and other that are permissive for ovariectomy bone loss have been uncov-
peripheral sites leads to an overall increase in TNF. TNF has ered. Evidence now suggests that these antigens are gut derived
indeed been shown to be significantly elevated in both human (J. Y. Li et al. 2016). The gut of humans and animals is a vast
peripheral blood cells in surgically ovariectomized women receptor for a myriad of microorganisms collectively referred
(Pacifici et al. 1991) and in mice (Cenci et al. 2000). Attesting to as the gut microbiota, which have the capacity to control
to the importance of TNF in ovariectomy bone loss, TNF and bone mass, as germ-free mice have increased BMD (Sjogren
TNF receptor I (p55) KO mice fail to undergo bone loss in et al. 2012). The efflux of bacterial antigens into the circulation
response to ovariectomy (Cenci et al. 2000). These studies led is controlled by tight junctions in the cells of the gut wall.
to a model whereby estrogen decline leads to an expansion in T Interestingly, these adhesion molecules are regulated by estro-
cells secreting TNF, and this TNF amplifies RANKL-induced gen, and their expression is down modulated by estrogen defi-
osteoclastic bone resorption causing bone loss (Weitzmann and ciency (J. Y. Li et al. 2016). The net effect is a leaky gut lining
Pacifici 2006). Although this general model still appears to that allows bacterial and other antigens to enter the circulation.
hold true, the regulation of the signaling cascades that lead to Attesting to the importance of the gut microbiota in estrogen
T cell expansion has turned out to be exceedingly complex. deficiency bone loss, germ-free mice do not loss bone mass
Among the key upstream events, driving T cell expansion is an after estrogen deprivation but again undergo bone resorption
increase in IL-7 production by multiple tissues within the body and loss following recolonization of the gut microbiota (J. Y. Li
(Ryan et al. 2005). IL-7 is a potent lymphopoietic cytokine that et al. 2016). Other studies have shown that treatment with
controls multiple steps in T cell biology. IL-7 increases the probiotics can prevent ovariectomy-induced bone loss (Britton
sensitivity of T cells to otherwise tolerogenic antigens decreas- et al. 2014; Ohlsson et al. 2014).
ing the threshold for antigen-dependent T cell activation Addition of other factors to this complex network driving
(Weitzmann and Pacifici 2006). Differentiation of T cells into estrogen deficiency bone loss continues relentlessly with identi-
different T-helper subsets including Th1 cells leads to produc- fication of additional participants including insulin-like growth
tion of TNF but also of Interferon (IFN)g, a cytokine that factor (IGF)-1 (Lindberg et al. 2006) and reactive oxygen spe-
upregulates the transcription factor transactivator (CIITA) in cies to the scheme (Grassi et al. 2007; Almeida et al. 2007).
macrophages causing upregulation of major histocompatibility Although early studies using B cell KO mice did not identify a
complex II (MHCII) and increased antigen presentation to role for B cells in ovariectomy-induced bone loss in mice (Y. Li,
T cells that further amplifies T cell activation (Cenci et al. Li, et al. 2007), recent studies using sophisticated RANKL condi-
2003). Another helper subset induced by this process is tional KO in the B and T lineage have revealed a partial contribu-
Th17, characterized by production of IL-17A, a potent osteo- tion of B cells to trabecular (but not cortical) bone loss in
clastogenic cytokine that induces production by osteoblast ovariectomy (Onal et al. 2012). As in our previous studies of
lineage cells of RANKL, the final osteoclastogenic effector murine ovariectomy, this study found no role for RANKL produc-
cytokine. IL-17 production is elevated in ovariectomy condi- tion by T cells in ovariectomy-induced bone loss (Onal et al. 2012).
tions (Tyagi et al. 2012) and anti-IL-17 antibodies (Tyagi et al. Although studies validating these responses in the human
2014), and IL-17 genetic deletion (DeSelm et al. 2012) context remain few and far between, at least one examination
ameliorates bone loss in ovariectomized mice. This process is of human bone marrow cells has revealed that both T cells
further sustained by downregulation of Transforming growth and B cells produce significantly elevated concentrations
factor (TGF)b, an estrogen-regulated cytokine, which mediates of RANKL, in postmenopausal women compared to
8 Toxicologic Pathology XX(X)

Figure 5. The bone loss of estrogen deficiency. Estrogen (E2) deficiency leads to bone loss though a complex cascade of interacting pathways
involving the immunoskeletal interface. Among the upstream events involved is an upswing in IL-7 production driving CD4þ T cell proliferation
and activation. These events are amplified by production of IFNg that upregulates antigen presentation further stimulating T cell activation. Th1
effector T cells secrete tumor necrosis factor (TNF) which is key to the inflammatory response in estrogen deficiency leading to bone loss. TNF
amplifies receptor activator of NF-kB ligand (RANKL)-induced osteoclastogenesis and stimulates RANKL production through formation of
Th17T cells which secrete IL-17A, a cytokine that promotes RANKL production by osteoblasts. Finally, estrogen deficiency leads to down-
regulation of the anti-inflammatory cytokine TGFb, leading to a decline in formation of regulatory T cells (Treg). Decline in Tregs further leads to
increased immune activation sustaining the inflammatory cascade.

premenopausal controls (Eghbali-Fatourechi et al. 2003). agents have found therapeutic application in RA (Kuek,
Treatment of postmenopausal women with estrogen replace- Hazleman, and Ostor 2007), an autoimmune inflammatory
ment therapy significantly diminished T cell and B cell disease characterized by focal destruction of bone and cartilage
RANKL production. Additional evidence in humans has also in joints and by systemic osteoporosis resulting from release
been provided by a clinical study examining peripheral blood of inflammatory cytokines into the circulation (Feldmann,
T cells which found that RANKL and TNF production was Brennan, and Maini 1996). Much of these cytokines emanate
significantly increased in postmenopausal women with osteo- from activated immune cells, and animal studies have demon-
porosis but not postmenopausal women without osteoporosis strated that conditional loss of RANKL production in the
or in premenopausal women (D’Amelio et al. 2008). adjuvant-induced osteoporosis animal model of RA protects
The role of the ISI in postmenopausal bone loss remains mice from bone loss (Kong et al. 1999).
controversial, as not all studies have validated significant Given the role of inflammation in RA etiology, a T cell costi-
effects of T cells in the response and some mouse and rat mulation inhibitor cytotoxic T lymphocyte-associated protein-4
models of T and/or B cell deficiency have produced variable (CTLA-4) immunoglobulin (Ig), also referred to as abatacept, is
outcomes (Lee, Kadono, et al. 2006; Lee, Kalinowski, et al. now Food and Drug Administration (FDA) approved for intract-
2006; Sass et al. 1997). able cases of RA. CTLA4-Ig blunts T cell activation and dimin-
Studies in animal and human systems continue, however, ished inflammation and bone loss downstream of the
and new findings are constantly being added to the literature. If inflammatory cascades (McCoy and Le Gros 1999).
validated, an interesting conclusion may be that estrogen defi- Because of the role of lymphocytes in the regulation of
ciency bone loss has significant hallmarks of an inflammatory basal osteoclastogenesis, an interesting question is whether
state (Weitzmann and Pacifici 2006). long-term therapy with CTLA4-Ig would be detrimental to
the skeleton by causing a RANKL/OPG imbalance and hence
causing collateral damage to the skeleton as was observed in
Bone Formation and the ISI HIV-induced immunodeficiency. To test this theory, we
An interesting finding is that the ISI not only plays key roles in injected abatacept into normal C57BL6 mice to examine its
regulation of bone resorption but also mediates potent effects long-term effects (6 month) on basal bone turnover (Roser-
on bone formation. In recent year’s potent, immunomodulatory Page et al. 2014). After 6 months of therapy, BMD was
Weitzmann 9

Figure 6. Bone formation by cytotoxic T lymphocyte-associated protein-4 immunoglobulin (CTLA4-Ig) and parathyroid hormone (PTH).
CTLA4-Ig targets and disrupts the CD28 costimulatory interaction with CD80/86. This signal is necessary to activate phosphodiesterase 4b
(PDE4b), which prevents accumulation of Cyclic adenosine monophosphate (cAMP) initiated by T cell receptor (TCR) binding to an antigen
bearing major histocompatibility complex (MHC) I complex on an antigen presenting cell. Sustained cAMP signaling renders the T cell anergic
(dormant) through Protein kinase A (PKA)-induced activation of cAMP responsive element-binding protein (CREB), anergic T cells also secrete
the bone anabolic ligand Wnt10b, following binding of CREB to CREB responsive elements (CRE) in its promoter. Wnt10b binds to its receptors
(LDL receptor related protein (LRP)5/6) on the osteoblasts leading to bone formation. PTH, when administered in an intermittent fashion, is also
able to stimulate Wnt10b production by CD8þ T cells following binding to its receptor (PTH-R), although the intracellular signaling pathways have
yet to be defined. PTH action is also dependent on direct effects mediated on osteoblasts that promote proliferation, differentiation, and increase
longevity of the bone-forming cells.

significantly elevated in CTLA4-Ig-treated mice compared to necessary requirement to pacify inflammation and prevent a
mice receiving Ig control. Surprisingly, metabolic turnover runaway immune response. CTLA4-Ig is a pharmacological
markers did not reveal any changes in baseline bone resorp- derivative of the natural CTLA4 molecule and functions simi-
tion; however, markers of bone formation were significantly larly to resolve inflammation by rendering T cells anergic
increased. These data were confirmed by quantitative bone (McCoy and Le Gros 1999).
histomorphometry, the gold standard for assessment of Interestingly, it has been reported that anergic T cells secrete
dynamic changes in bone formation in vivo (Roser-Page high levels of a Wingless integration site (Wnt) pathway agonist
et al. 2014). called Wnt10b (Zha et al. 2006). The Wnt pathway is potently
The mechanism of CTLA4-Ig action involves suppression bone anabolic and canonical Wnt signaling modulates many
of CD28 signaling in the T cell. T cell activation occurs aspects of osteoblast physiology including proliferation, differ-
through a process involving 2 separate steps (the dual signal entiation, bone matrix formation/mineralization, and apoptosis
hypothesis; Sayegh 1999). The first signal involves the presen- (Bodine and Komm 2006). Consequently, a logical hypothesis is
tation of an antigen as part of an major histocompatibility that anergic T cells promote bone anabolic responses by secre-
complex (MHC) complex by an Antigen presenting cell (APC) tion of Wnt10b. To test this model, we quantified Wnt10b pro-
to the T cell receptor complex. This is not an activation signal duction by real-time RT-PCR in an in vitro APC assay in which
but in fact renders the T cell anergic (dormant) unless a second transgenic T cells (bearing a T cell receptor specific for ovalbu-
costimulatory signal is received. The second signal is trans- min) are cocultured with ovalbumin (antigen)-loaded APC (den-
mitted by the APCs CD80 or CD86 costimulatory ligands to dritic cells). While T cells alone or APC alone did not express
the CD28 receptor on the T cell. Activation of CD28 alleviates Wnt10b, T cell engagement of antigen on dendritic cells led to a
the anergic block allowing T cell activation, differentiation into small induction of Wnt10b expression. Addition of CTLA4-Ig,
T helper cells, and effector function (Sayegh 1999). CTLA4 is breaking costimulation and rendering the activating T cells aner-
a physiological molecule produced by activated T cells and gic, potently upregulated Wnt10b expression by more than an
Tregs that acts as a decoy receptor for CD28, blocking CD80 order of magnitude (Roser-Page et al. 2014).
and CD86, and preventing CD28 activation. CTLA4 thus ter- To further validate this hypothesis in vivo, we treated mice
minates immune responses following resolution of infection, a with CTLA4-Ig and then immuno-magnetically purified T
10 Toxicologic Pathology XX(X)

Figure 7. Parathyroid hormone (PTH) and bone loss. The bone catabolic effects of chronic PTH result from production of tumor necrosis factor
(TNF) by T cells that feeds back though cell autonomous effects to promote Th17 effector cell formation. Th17T cells secrete IL-17A, a cytokine
that binds to receptors (IL-17R) on osteoblasts and osteocytes stimulating receptor activator of NF-kB ligand (RANKL) production that promotes
osteoclastogenesis and bone resorption leading to bone loss. Finally, TNF upregulates CD40 receptor on bone marrow stromal cells. Binding of
CD40 with its ligand CD40L on T cells imparts proliferative and survival cues to osteoblasts.

cells. T cells were cultured overnight, and secretion of Wnt10b resorptive response that is overshadowed by a robust increase
into the medium was quantified by ELISA. Secretion of in bone formation leading to a significant net bone gain. In
Wnt10b protein was significantly elevated by in vivo treatment fact, teriparatide, a fragment of human PTH injected daily, is
of mice with CTLA4-Ig (Roser-Page et al. 2014). an FDA approved bone anabolic modality for treatment of
Taken together, the data show a surprising T cell mediated osteoporosis through stimulation of bone anabolism in
capacity to not only promote inflammatory bone loss but also a humans (Pacifici 2013).
capacity to potentially repair collateral damage to bone follow-
ing resolution of inflammation through bone anabolic actions
involving T cell secretion of Wnt10b. This model is presented Anabolic Actions of PTH and the ISI
diagrammatically in Figure 6. Interestingly, anabolic actions of PTH involve the ISI. A tra-
ditional view of anabolic PTH is that PTH induces direct osteo-
Role of T Cells in Parathyroid Hormone blast proliferation and differentiation, activation of quiescent
lining osteoblasts, increasing osteoblast life span by suppres-
(PTH)-Induced Bone Formation and
sing apoptosis, and down modulation of the Wnt receptor
Bone Loss antagonist sclerostin in osteocytes (Pacifici 2013). Although
PTH is a complex and paradoxical factor. As an important the Wnt pathway has been recognized as a key target of PTH
regulator of calcium metabolism in the body, it defends in bone anabolic effects, the source of the Wnt ligands driving
against hypocalcemia by stimulating the release of calcium bone formation in this context is poorly characterized and
from the skeleton by promoting bone resorption (Weitzmann largely considered to be secreted in a cell autonomous fashion
and Pacifici 2017). However, primary or secondary hyperpar- by osteoblast-lineage cells themselves. However, recent data
athyroidism causes sustained overproduction of PTH driving suggest that a critical source of Wnt ligand is in fact the T cell.
severe osteoclastic bone resorption and skeletal deterioration As with CTLA4-Ig, PTH induces significant production of
(Pacifici 2010). PTH-induced bone loss may be modeled in Wnt10b from T cells. Attesting to the importance of T cell–
mice by the chronic administration of PTH through mini- produced Wnt10b, reconstitution of WT T cells into TCRb KO
osmotic pumps leading to a balance in bone turnover in which mice restores the anabolic activity of PTH, while adoptive
bone resorption outpaces that of bone formation leading to transfer of T cells from Wnt10b KO mice fails to rescue the
bone loss. By contrast, when administered in an intermittent anabolic activity of PTH (Terauchi et al. 2009). This model is
(pulsatile) manner paradoxically, PTH leads to a weak bone presented diagrammatically in Figure 6.
Weitzmann 11

Catabolic Actions of PTH and the ISI Authors’ Note


The traditional view of PTH-induced bone resorption is that The contents of this manuscript do not represent the views of the
Department of Veterans Affairs, the National Institutes of Health, or
PTH targets cells of the osteoblast lineage to increase the pro-
the United States Government.
duction of RANKL and decrease the production of OPG. This
rebalances the RANKL/OPG ratio in favor of increased osteo- Acknowledgments
clastogenesis and bone resorption (Pacifici 2013).
The author gratefully acknowledges research support from the
Evidence for a role of T cells in PTH-induced bone
Biomedical Laboratory Research and Development Service of the
resorption came from studies almost 2 decades ago report- VA Office of Research and Development (5I01BX000105) and from
ing that transplantation of human parathyroid glands from the National Institutes of Health (the National Institute of Arthritis and
patients with hyperparathyroidism into T cell deficient nude Musculoskeletal and Skin Diseases [grant numbers AR056090,
mice failed to induce bone loss, suggesting a role for T cells AR059364, AR068157, and AR070091] and the National Institute
in the catabolic activity of PTH (Hory et al. 2000). Follow- on Aging [grant number AG040013]).
up studies by our group almost a decade later confirmed that
administration of PTH in TCRb KO T cell deficient mice Author Contribution
does not induce a catabolic effect on the skeleton. However, All authors (MW) contributed to conception or design; data acquisi-
adoptive transfer of WT T cells into TCRb KO mice res- tion, analysis, or interpretation; drafting the manuscript; and critically
cued the catabolic action (Gao et al. 2008). Evidence that revising the manuscript. All authors gave final approval and agreed to
PTH acts directly on T cells came from additional studies in be accountable for all aspects of work in ensuring that questions
which the PTH receptor was conditionally deleted on T cells relating to the accuracy or integrity of any part of the work are appro-
leading to a failure of PTH to induce bone loss (Bedi et al. priately investigated and resolved.
2012). Mechanistically, PTH promotes secretion of TNF
from T cells, which in addition to its amplificatory role in Declaration of Conflicting Interests
RANKL signaling and suppression of OPG feeds back on T The author(s) declared no potential conflicts of interest with respect to
cells to simulate their differentiation to Th17 subsets that the research, authorship, and/or publication of this article.
secrete IL-17A leading to additional RANKL production by
osteoblast lineage cells. Finally, TNF upregulates CD40, a Funding
costimulatory receptor, on bone marrow stromal cells. Bind- The author(s) disclosed receipt of the following financial support for
ing of CD40 with its ligand CD40L on T cells imparts the research, authorship, and/or publication of this article: Biomedical
proliferative and survival cues to osteoblast precursors fur- Laboratory Research and Development Service of the VA Office of
Research and Development (5I01BX000105) and from the National
ther contributing to the catabolic action of PTH (Gao et al.
Institutes of Health (the National Institute of Arthritis and Musculos-
2008; Bedi et al. 2010; Tawfeek et al. 2010). This model is
keletal and Skin Diseases [grant numbers AR056090, AR059364,
presented diagrammatically in Figure 7. AR068157, and AR070091] and the National Institute on Aging [grant
number AG040013]).

Conclusions References
Our understanding of the ISI and the field of osteoimmunology Almeida, M., Han, L., Martin-Millan, M., Plotkin, L. I., Stewart, S. A., Rober-
continues to evolve rapidly with new discoveries attesting to son, P. K., Kousteni, S., et al. (2007). Skeletal involution by age-associated
oxidative stress and its acceleration by loss of sex steroids. J Biol Chem
the considerable depth of the ISI and the influence of the 282, 27285–97.
immune system on bone. The immune system mediates protec- Anderson, D. M., Maraskovsky, E., Billingsley, W. L., Dougall, W. C.,
tive actions on physiological bone turnover under basal condi- Tometsko, M. E., Roux, E. R., Teepe, M. C., et al. (1997). A homologue
tions but causes pathological destruction when activated (or of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell
function. Nature 390, 175–79.
compromised as in immunosuppressive states) and causes
Bass, E., French, D. D., Bradham, D. D., and Rubenstein, L. Z. (2007). Risk-
increased fracture incidence in a number of disease states. The adjusted mortality rates of elderly veterans with hip fractures. Ann Epide-
immune response further regulates bone resorption and bone miol 17, 514–19.
formation depending on context. The latter discovery may have Bedi, B., Li, J. Y., Grassi, F., Tawfeek, H., Weitzmann, M. N., and Pacifici, R.
important implications for the future development of bone ana- (2010). Inhibition of antigen presentation and T cell costimulation blocks
bolic agents to combat osteoporosis. Teriparatide (anabolic PTH-induced bone loss. Ann N Y Acad Sci 1192, 215–21.
Bedi, B., Li, J. Y., Tawfeek, H., Baek, K. H., Adams, J., Vangara, S. S., Chang,
PTH) is already an FDA approved modality for increasing bone M. K., et al. (2012). Silencing of parathyroid hormone (PTH) receptor 1 in
formation. Our recent work is further focusing on the use of T cells blunts the bone anabolic activity of PTH. Proc Natl Acad Sci U S A
anergic T cells, as bone anabolic agents and immunosuppres- 109, E725–33.
sive drugs such as abatacept to further promote bone anabolism Bodine, P. V., and Komm, B. S. (2006). Wnt signaling and osteoblastogenesis.
Rev Endocr Metab Disord 7, 33–39.
alone or to amplify the effects of teriparatide. With new under-
Britton, R. A., Irwin, R., Quach, D., Schaefer, L., Zhang, J., Lee, T., Para-
standing of the integration between immune and skeletal func- meswaran, N., and McCabe, L. R. (2014). Probiotic L. reuteri treatment
tions, yet additional immune targets are likely to emerge for prevents bone loss in a menopausal ovariectomized mouse model. J Cell
amelioration of bone loss in the future. Physiol 229, 1822–30.
12 Toxicologic Pathology XX(X)

Brown, T. T., McComsey, G. A., King, M. S., Qaqish, R. B., Bernstein, B. M., deficient mice through enhanced bone marrow dendritic cell activation.
and da Silva, B. A. (2009). Loss of bone mineral density after antiretroviral Proc Natl Acad Sci U S A 104, 15087–92.
therapy initiation, independent of antiretroviral regimen. J Acquir Immune Grigsby, I. F., Pham, L., Gopalakrishnan, R., Mansky, L. M., and Mansky, K.
Defic Syndr 51, 554–61. C. (2010). Downregulation of Gnas, Got2 and Snord32a following tenofo-
Brown, T. T., and Qaqish, R. B. (2006). Antiretroviral therapy and the pre- vir exposure of primary osteoclasts. Biochem Biophys Res Commun 391,
valence of osteopenia and osteoporosis: A meta-analytic review. AIDS 20, 1324–29.
2165–74. Grigsby, I. F., Pham, L., Mansky, L. M., Gopalakrishnan, R., Carlson, A. E.,
Bruera, D., Luna, N., David, D. O., Bergoglio, L. M., and Zamudio, J. (2003). and Mansky, K. C. (2010). Tenofovir treatment of primary osteoblasts
Decreased bone mineral density in HIV-infected patients is independent of alters gene expression profiles: Implications for bone mineral density loss.
antiretroviral therapy. AIDS 17, 1917–23. Biochem Biophys Res Commun 394, 48–53.
Burge, R., Dawson-Hughes, B., Solomon, D. H., Wong, J. B., King, A., and Guerri-Fernandez, R., Vestergaard, P., Carbonell, C., Knobel, H., Aviles, F. F.,
Tosteson, A. (2007). Incidence and economic burden of osteoporosis- Castro, A. S., Nogues, X., Prieto-Alhambra, D., and Diez-Perez, A. (2013).
related fractures in the United States, 2005-2025. J Bone Miner Res 22, HIV infection is strongly associated with hip fracture risk, independently of
465–75. age, gender, and comorbidities: A population-based cohort study. J Bone
Buring, K. (1975). On the origin of cells in heterotopic bone formation. Clin Miner Res 28, 1259–63.
Orthop Relat Res 110, 293–301. Hofbauer, L. C., Lacey, D. L., Dunstan, C. R., Spelsberg, T. C., Riggs, B. L.,
Cenci, S., Toraldo, G., Weitzmann, M. N., Roggia, C., Gao, Y., Qian, W. P., and Khosla, S. (1999). Interleukin-1beta and tumor necrosis factor-alpha,
Sierra, O., and Pacifici, R. (2003). Estrogen deficiency induces bone loss by but not interleukin-6, stimulate osteoprotegerin ligand gene expression in
increasing T cell proliferation and lifespan through IFN-gamma-induced human osteoblastic cells. Bone 25, 255–59.
class II transactivator. Proc Natl Acad Sci U S A 100, 10405–10. Hory, B. G., Roussanne, M. C., Rostand, S., Bourdeau, A., Drueke, T. B., and
Cenci, S., Weitzmann, M. N., Roggia, C., Namba, N., Novack, D., Woodring, Gogusev, J. (2000). Absence of response to human parathyroid hormone in
J., and Pacifici, R. (2000). Estrogen deficiency induces bone loss by enhan- athymic mice grafted with human parathyroid adenoma, hyperplasia or
cing T-cell production of TNF-a. J Clin Invest 106, 1229–37. parathyroid cells maintained in culture. J Endocrinol Invest 23, 273–79.
Chan, H. L., Fung, S., Seto, W. K., Chuang, W. L., Chen, C. Y., Kim, H. J., Hui, Johnell, O., and Kanis, J. A. (2006). An estimate of the worldwide prevalence
A. J., et al. (2016). Tenofovir alafenamide versus tenofovir disoproxil and disability associated with osteoporotic fractures. Osteoporos Int 17,
fumarate for the treatment of HBeAg-positive chronic hepatitis B virus 1726–33.
infection: A randomised, double-blind, phase 3, non-inferiority trial. Lan- Kates, S. L., Kates, O. S., and Mendelson, D. A. (2007). Advances in the
cet Gastroenterol Hepatol 1, 185–95. medical management of osteoporosis. Injury 38, S17–23.
Cummings, S. R., and Melton, L. J. (2002). Epidemiology and outcomes of Kimble, R. B., Srivastava, S., Ross, F. P., Matayoshi, A., and Pacifici, R.
osteoporotic fractures. Lancet 359, 1761–1767. (1996). Estrogen deficiency increases the ability of stromal cells to support
D’Amelio, P., Grimaldi, A., Di Bella, S., Brianza, S. Z., Cristofaro, M. A., murine osteoclastogenesis via an interleukin-1and tumor necrosis factor-
Tamone, C., Giribaldi, G., et al. (2008). Estrogen deficiency increases mediated stimulation of macrophage colony-stimulating factor production.
osteoclastogenesis up-regulating T cells activity: A key mechanism in J Biol Chem 271, 28890–97.
osteoporosis. Bone 43, 92–100. Kitazawa, R., Kimble, R. B., Vannice, J. L., Kung, V. T., and Pacifici, R.
DeSelm, C. J., Takahata, Y., Warren, J., Chappel, J. C., Khan, T., Li, X., Liu, (1994). Interleukin-1 receptor antagonist and tumor necrosis factor binding
C., et al. (2012). IL-17 mediates estrogen-deficient osteoporosis in an Act1- protein decrease osteoclast formation and bone resorption in ovariecto-
dependent manner. J Cell Biochem 113, 2895–902. mized mice. J Clin Invest 94, 2397–406.
Eghbali-Fatourechi, G., Khosla, S., Sanyal, A., Boyle, W. J., Lacey, D. L., and Kong, Y. Y., Feige, U., Sarosi, I., Bolon, B., Tafuri, A., Morony, S., Capparelli,
Riggs, B. L. (2003). Role of RANK ligand in mediating increased bone C., et al. (1999). Activated T cells regulate bone loss and joint destruction
resorption in early postmenopausal women. J Clin Invest 111, 1221–30. in adjuvant arthritis through osteoprotegerin ligand. Nature 402, 304–9.
Eisman, J. A., Bogoch, E. R., Dell, R., Harrington, J. T., McKinney, R. E. Jr., Kotake, S., Udagawa, N., Hakoda, M., Mogi, M., Yano, K., Tsuda, E., Taka-
McLellan, A., Mitchell, P. J., et al. (2012). Making the first fracture the last hashi, K., et al. (2001). Activated human T cells directly induce osteoclas-
fracture: ASBMR task force report on secondary fracture prevention. J togenesis from human monocytes: Possible role of T cells in bone
Bone Miner Res 27, 2039–46. destruction in rheumatoid arthritis patients. Arthritis Rheum 44, 1003–12.
Ernst, B., Lee, D. S., Chang, J. M., Sprent, J., and Surh, C. D. (1999). The Kuek, A., Hazleman, B. L., and Ostor, A. J. (2007). Immune-mediated inflam-
peptide ligands mediating positive selection in the thymus control T cell matory diseases (IMIDs) and biologic therapy: A medical revolution. Post-
survival and homeostatic proliferation in the periphery. Immunity 11, grad Med J 83, 251–60.
173–81. Lacey, D. L., Timms, E., Tan, H. L., Kelley, M. J., Dunstan, C. R., Burgess, T.,
Feldmann, M., Brennan, F. M., and Maini, R. (1996). Rheumatoid arthritis. Elliott, R., et al. (1998). Osteoprotegerin ligand is a cytokine that regulates
Cell 85, 307–10. osteoclast differentiation and activation. Cell 93, 165–76.
Fuller, K., Murphy, C., Kirstein, B., Fox, S. W., and Chambers, T. J. (2002). Lam, J., Takeshita, S., Barker, J. E., Kanagawa, O., Ross, F. P., and Teitel-
TNFa potently activates osteoclasts, through a direct action independent of baum, S. L. (2000). TNF-a induces osteoclastogenesis by direct stimulation
and strongly synergistic with RANKL. Endocrinology 143, 1108–18. of macrophages exposed to permissive levels of RANK ligand. J Clin
Gao, Y., Wu, X., Terauchi, M., Li, J. Y., Grassi, F., Galley, S., Yang, X., Invest 106, 1481–88.
Weitzmann, M. N., and Pacifici, R. (2008). T cells potentiate PTH- Lee, S. K., Kadono, Y., Okada, F., Jacquin, C., Koczon-Jaremko, B., Grono-
induced cortical bone loss through CD40 L signaling. Cell Metab 8, wicz, G., Adams, D. J., et al. (2006). T lymphocyte-deficient mice lose
132–45. trabecular bone mass with ovariectomy. J Bone Miner Res 21, 1704–12.
Gibellini, D., Borderi, M., de Crignis, E., Clo, A., Miserocchi, A., Viale, P., and Lee, S. K., Kalinowski, J. F., Jacquin, C., Adams, D. J., Gronowicz, G., and
Re, M. C. (2010). Analysis of the effects of specific protease inhibitors on Lorenzo, J. A. (2006). Interleukin-7 influences osteoclast function in vivo
OPG/RANKL regulation in an osteoblast-like cell line. New Microbiol 33, but is not a critical factor in ovariectomy-induced bone loss. J Bone Miner
109–15. Res 21, 695–702.
Grant, P. M., Kitch, D., McComsey, G. A., Dube, M. P., Haubrich, R., Huang, Lewis, J. R., Hassan, S. K., Wenn, R. T., and Moran, C. G. (2006). Mortality
J., Riddler, S., et al. (2013). Low baseline CD4þ count is associated with and serum urea and electrolytes on admission for hip fracture patients.
greater bone mineral density loss after antiretroviral therapy initiation. Clin Injury 37, 698–704.
Infect Dis 57, 1483–88. Li, J. Y., Chassaing, B., Tyagi, A. M., Vaccaro, C., Luo, T., Adams, J., Darby,
Grassi, F., Tell, G., Robbie-Ryan, M., Gao, Y., Terauchi, M., Yang, X., Roma- T. M., et al. (2016). Sex steroid deficiency-associated bone loss is micro-
nello, M., et al. (2007). Oxidative stress causes bone loss in estrogen- biota dependent and prevented by probiotics. J Clin Invest 126, 2049–63.
Weitzmann 13

Li, P., and Schwarz, E. M. (2003). The TNF-a transgenic mouse model of output contributes to the bone loss induced by estrogen deficiency. Proc
inflammatory arthritis. Springer Semin Immunopathol 25, 19–33. Natl Acad Sci U S A 102, 16735–40.
Li, Y., Li, A., Yang, X., and Weitzmann, M. N. (2007). Ovariectomy-induced Sass, D. A., Liss, T., Bowman, A. R., Rucinski, B., Popoff, S. N., Pan, Z., Ma,
bone loss occurs independently of B cells. J Cell Biochem 100, 1370–75. Y. F., and Epstein, S. (1997). The role of the T-lymphocyte in estrogen
Li, Y., Toraldo, G., Li, A., Yang, X., Zhang, H., Qian, W. P., and Weitzmann, deficiency osteopenia. J Bone Miner Res 12, 479–86.
M. N. (2007). B cells and T cells are critical for the preservation of bone Sayegh, M. H. (1999). Finally, CTLA4Ig graduates to the clinic. J Clin Invest
homeostasis and attainment of peak bone mass in vivo. Blood 109, 103, 1223–25.
3839–48. Sharma, A., Shi, Q., Hoover, D. R., Anastos, K., Tien, P. C., Young, M. A.,
Lindberg, M. K., Svensson, J., Venken, K., Chavoshi, T., Andersson, N., Cohen, M. H., et al. (2015). Increased fracture incidence in middle-aged
Moverare Skrtic, S., Isaksson, O., et al. (2006). Liver-derived IGF-I is HIV-infected and HIV-uninfected women: Updated results from the
permissive for ovariectomy-induced trabecular bone loss. Bone 38, 85–92. women’s interagency HIV study. J Acquir Immune Defic Syndr 70, 54–61.
McComsey, G. A., Tebas, P., Shane, E., Yin, M. T., Overton, E. T., Huang, J. Simonet, W. S., Lacey, D. L., Dunstan, C. R., Kelley, M., Chang, M. S., Luthy,
S., Aldrovandi, G. M., et al. (2010). Bone disease in HIV infection: A R., Nguyen, H. Q., et al. (1997). Osteoprotegerin: A novel secreted protein
practical review and recommendations for HIV care providers. Clin Infect involved in the regulation of bone density. Cell 89, 309–19.
Dis 51, 937–46. Sjogren, K., Engdahl, C., Henning, P., Lerner, U. H., Tremaroli, V., Lagerquist,
McCoy, K. D., and Le Gros, G. (1999). The role of CTLA-4 in the regulation of M. K., Backhed, F., and Ohlsson, C. (2012). The gut microbiota regulates
T cell immune responses. Immunol Cell Biol 77, 1–10. bone mass in mice. J Bone Miner Res 27, 1357–67.
Miyaura, C., Onoe, Y., Inada, M., Maki, K., Ikuta, K., Ito, M., and Suda, T. Surh, C. D., and Sprent, J. (2000). Homeostatic T cell proliferation: How far
(1997). Increased B-lymphopoiesis by interleukin 7 induces bone loss in can T cells be activated to self-ligands? J Exp Med 192, F9–14.
mice with intact ovarian function: Similarity to estrogen deficiency. Proc Tawfeek, H., Bedi, B., Li, J. Y., Adams, J., Kobayashi, T., Weitzmann, M. N.,
Natl Acad Sci U S A 94, 9360–65. Kronenberg, H. M., and Pacifici, R. (2010). Disruption of PTH receptor 1 in
Ofotokun, I., Titanji, K., Vikulina, T., Roser-Page, S., Yamaguchi, M., T cells protects against PTH-induced bone loss. PLoS One 5, e12290.
Zayzafoon, M., Williams, I. R., and Weitzmann, M. N. (2015). Role Teitelbaum, S. L. (2000). Bone resorption by osteoclasts. Science 289, 1504–8.
of T-cell reconstitution in HIV-1 antiretroviral therapy-induced bone Terauchi, M., Li, J. Y., Bedi, B., Baek, K. H., Tawfeek, H., Galley, S., Gilbert,
loss. Nat Commun 6, 8282. L., et al. (2009). T lymphocytes amplify the anabolic activity of parathyroid
Ofotokun, I., Titanji, K., Vunnava, A., Roser-Page, S., Vikulina, T., Villinger, hormone through Wnt10b signaling. Cell Metab 10, 229–40.
F., Rogers, K., et al. (2016). Antiretroviral therapy induces a rapid increase Titanji, K., Vunnava, A., Sheth, A. N., Delille, C., Lennox, J. L., Sanford, S. E.,
in bone resorption that is positively associated with the magnitude of Foster, A., et al. (2014). Dysregulated B cell expression of RANKL and
immune reconstitution in HIV infection. AIDS 30, 405–14. OPG correlates with loss of bone mineral density in HIV infection. PLoS
Ohlsson, C., Engdahl, C., Fak, F., Andersson, A., Windahl, S. H., Farman, H. Pathog 10, e1004497.
H., Moverare-Skrtic, S., Islander, U., and Sjogren, K. (2014). Probiotics Triant, V. A., Brown, T. T., Lee, H., and Grinspoon, S. K. (2008). Fracture
protect mice from ovariectomy-induced cortical bone loss. PLoS One 9, prevalence among human immunodeficiency virus (HIV)-infected versus
e92368. non-HIV-infected patients in a large U.S. healthcare system. J Clin Endo-
Onal, M., Xiong, J., Chen, X., Thostenson, J. D., Almeida, M., Manolagas, S. crinol Metab 93, 3499–504.
C., and O’Brien, C. A. (2012). Receptor activator of nuclear factor kappaB Tsuda, E., Goto, M., Mochizuki, S., Yano, K., Kobayashi, F., Morinaga, T.,
ligand (RANKL) protein expression by B lymphocytes contributes to and Higashio, K. (1997). Isolation of a novel cytokine from human fibro-
ovariectomy-induced bone loss. J Biol Chem 287, 29851–60. blasts that specifically inhibits osteoclastogenesis. Biochem Biophys Res
Pacifici, R. (2010). T cells: Critical bone regulators in health and disease. Bone Commun 234, 137–42.
47, 461–71. Tyagi, A. M., Mansoori, M. N., Srivastava, K., Khan, M. P., Kureel, J., Dixit,
Pacifici, R. (2013). Role of T cells in the modulation of PTH action: Physio- M., Shukla, P., et al. (2014). Enhanced immunoprotective effects by anti-
logical and clinical significance. Endocrine 44, 576–82. IL-17 antibody translates to improved skeletal parameters under estrogen
Pacifici, R., Brown, C., Puscheck, E., Friedrich, E., Slatopolsky, E., Maggio, deficiency compared with anti-RANKL and anti-TNF-a antibodies.
D., McCracken, R., and Avioli, L. V. (1991). Effect of surgical menopause J Bone Miner Res 29, 1981–92.
and estrogen replacement on cytokine release from human blood mono- Tyagi, A. M., Srivastava, K., Mansoori, M. N., Trivedi, R., Chattopadhyay,
nuclear cells. Proc Natl Acad Sci U S A 88, 5134–38. N., and Singh, D. (2012). Estrogen deficiency induces the differentiation
Piso, R. J., Rothen, M., Rothen, J. P., and Stahl, M. (2011). Markers of bone of IL-17 secreting Th17 cells: A new candidate in the pathogenesis of
turnover are elevated in patients with antiretroviral treatment independent osteoporosis. PLoS One 7, e44552.
of the substance used. J Acquir Immune Defic Syndr 56, 320–24. Unnanuntana, A., Gladnick, B. P., Donnelly, E., and Lane, J. M. (2010). The
Prieto-Alhambra, D., Guerri-Fernandez, R., De Vries, F., Lalmohamed, A., assessment of fracture risk. J Bone Joint Surg Am 92, 743–53.
Bazelier, M., Starup-Linde, J., Diez-Perez, A., Cooper, C., and Vester- Vikulina, T., Fan, X., Yamaguchi, M., Roser-Page, S., Zayzafoon, M., Guidot,
gaard, P. (2014). HIV infection and its association with an excess risk of D. M., Ofotokun, I., and Weitzmann, M. N. (2010). Alterations in the
clinical fractures: A nationwide case-control study. J Acquir Immune Defic immuno-skeletal interface drive bone destruction in HIV-1 transgenic rats.
Syndr 66, 90–95. Proc Natl Acad Sci U S A 107, 13848–53.
Prior, J., Burdge, D., Maan, E., Milner, R., Hankins, C., Klein, M., and Walms- Walker, D. G. (1972). Congenital osteopetrosis in mice cured by parabiotic
ley, S. (2007). Fragility fractures and bone mineral density in HIV positive union with normal siblings. Endocrinology 91, 916–20.
women: A case-control population-based study. Osteoporos Int 18, Walker, D. G. (1975). Bone resorption restored in osteopetrotic mice by trans-
1345–53. plants of normal bone marrow and spleen cells. Science 190, 784–85.
Reid, W., Sadowska, M., Denaro, F., Rao, S., Foulke, J. Jr., Hayes, N., Wang, M. W., Wei, S., Faccio, R., Takeshita, S., Tebas, P., Powderly, W. G.,
Jones, O., et al. (2001). An HIV-1 transgenic rat that develops Teitelbaum, S. L., and Ross, F. P. (2004). The HIV protease inhibitor rito-
HIV-related pathology and immunologic dysfunction. Proc Natl Acad navir blocks osteoclastogenesis and function by impairing RANKL-induced
Sci U S A 98, 9271–76. signaling. J Clin Invest 114, 206–13.
Roser-Page, S., Vikulina, T., Zayzafoon, M., and Weitzmann, M. N. (2014). Weitzmann, M. N., and Ofotokun, I. (2016). Physiological and pathophysio-
CTLA-4Ig-induced T cell anergy promotes Wnt-10b production and bone logical bone turnover—Role of the immune system. Nat Rev Endocrinol
formation in a mouse model. Arthritis Rheumatol 66, 990–99. 12, 518–32.
Ryan, M. R., Shepherd, R., Leavey, J. K., Gao, Y., Grassi, F., Schnell, F. J., Weitzmann, M. N., and Pacifici, R. (2006). Estrogen deficiency and bone loss:
Qian, W. P., et al. (2005). An IL-7-dependent rebound in thymic T cell An inflammatory tale. J Clin Invest 116, 1186–94.
14 Toxicologic Pathology XX(X)

Weitzmann, M. N., and Pacifici, R. (2017). Parathyroid diseases and T cells. expressed in T cells, is a dendritic cell-specific survival factor. J Exp Med
Curr Osteoporos Rep 15, 135–41. 186, 2075–80.
Weitzmann, M. N., Roggia, C., Toraldo, G., Weitzmann, L., and Pacifici, R. Young, B., Dao, C. N., Buchacz, K., Baker, R., and Brooks, J. T. (2011).
(2002). Increased production of IL-7 uncouples bone formation from bone Increased rates of bone fracture among HIV-infected persons in the HIV
resorption during estrogen deficiency. J Clin Invest 110, 1643–50. outpatient study (HOPS) compared with the US general population, 2000-
Wohl, D., Oka, S., Clumeck, N., Clarke, A., Brinson, C., Stephens, J., Tashima, 2006. Clin Infect Dis 52, 1061–68.
K., et al. (2016). A randomized, double-blind comparison of tenofovir Yun, T. J., Chaudhary, P. M., Shu, G. L., Frazer, J. K., Ewings, M. K.,
alafenamide (TAF) vs. tenofovir disoproxil fumarate (TDF), each cofor- Schwartz, S. M., Pascual, V., Hood, L. E., and Clark, E. A. (1998).
mulated with elvitegravir, cobicistat, and emtricitabine (E/C/F) for initial OPG/FDCR-1, a TNF receptor family member, is expressed in lymphoid
HIV-1 treatment: Week 96 results. J Acquir Immune Defic Syndr 72, cells and is up-regulated by ligating CD40. J Immunol 161, 6113–21.
58–64. Zha, Y., Marks, R., Ho, A. W., Peterson, A. C., Janardhan, S., Brown, I.,
Womack, J. A., Goulet, J. L., Gibert, C., Brandt, C., Chang, C. C., Gulanski, B., Praveen, K., et al. (2006). T cell anergy is reversed by active Ras and is
Fraenkel, L., et al. (2011). Increased risk of fragility fractures among HIV regulated by diacylglycerol kinase-alpha. Nat Immunol 7, 1166–73.
infected compared to uninfected male veterans. PLoS One 6, e17217. Zhang, Y. H., Heulsmann, A., Tondravi, M. M., Mukherjee, A., and Abu-
Wong, B. R., Josien, R., Lee, S. Y., Sauter, B., Li, H. L., Steinman, R. M., and Amer, Y. (2001). Tumor necrosis factor-a (TNF) stimulates RANKL-
Choi, Y. (1997). TRANCE (tumor necrosis factor [TNF]-related induced osteoclastogenesis via coupling of TNF type 1 receptor and RANK
activation-induced cytokine), a new TNF family member predominantly signaling pathways. J Biol Chem 276, 563–68.

You might also like