Sei sulla pagina 1di 9

Nephrol Dial Transplant (2018) 1–9

doi: 10.1093/ndt/gfy115

Bone, inflammation and the bone marrow niche in chronic


kidney disease: what do we know?

REVIEW
Sandro Mazzaferro1,6, Giuseppe Cianciolo2, Antonio De Pascalis3, Chiara Guglielmo2,
Pablo A. Urena Torres4, Jordi Bover5, Lida Tartaglione1, Marzia Pasquali6 and Gaetano La Manna2
1
Department of Cardiovascular Respiratory Nephrologic Geriatric and Anesthetic Sciences, Sapienza University of Rome, Rome, Italy,
2
Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital,
University of Bologna, Bologna, Italy, 3Nephrology, Dialysis and Renal Transplant Unit, Vito Fazzi Hospital, Lecce, Italy, 4Ramsay-Générale de
Santé, Clinique du Landy, Department of Nephrology and Dialysis and Department of Renal Physiology, Necker Hospital, University of Paris
Descartes, Paris, France, 5Fundació Puigvert, Department of Nephrology IIB Sant Pau, RedinRen, Barcelona, Catalonia, Spain and 6Nephrology
Unit, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Rome, Italy

Correspondence and offprint requests to: Sandro Mazzaferro; E-mail: sandro.mazzaferro@uniroma1.it

ABSTRACT clinical entity of chronic kidney disease–mineral and bone dis-


Recent improvements in our understanding of physiology have orders and its cardiovascular burden. Bone is thus becoming a
altered the way in which bone is perceived: no longer is it con- recurrently considered paradigm for different inter-organ com-
sidered as simply the repository of divalent ions, but rather as a munications that needs to be considered in patients with com-
sophisticated endocrine organ with potential extraskeletal plex diseases.
effects. Indeed, a number of pathologic conditions involving Keywords: atherosclerosis, bone marrow niche, chronic kid-
bone in different ways can now be reconsidered from a bone- ney disease, CKD-MBD, FGF23, inflammation, PTH, renal
centred perspective. For example, in metabolic bone diseases osteodystrophy, vitamin D
like osteoporosis (OP) and renal osteodystrophy (ROD), the as-
sociation with a worse cardiovascular outcome can be tenta-
tively explained by the possible derangements of three recently INTRODUCTION
discovered bone hormones (osteocalcin, fibroblast growth fac- The recognition in recent years of the embryonic origin of bone
tor 23 and sclerostin) and a bone-specific enzyme (alkaline cells (osteoclasts from haematopoietic stem/progenitor cells or
phosphatase). Further, in recent years the close link between HSPCs, and osteoblasts from mesenchymal stem cells or MSCs)
bone and inflammation has been better appreciated and a wide and of the hormone-like properties of a number of ‘non-collag-
range of chronic inflammatory states (from rheumatoid arthri- enous bone proteins’ confers on bone the potential for systemic
tis to ageing) are being explored to discover the biochemical extraskeletal effects [1]. In addition, the clinical effects of
changes that ultimately lead to bone loss and OP. Also, it has chronic inflammation in various systemic diseases has increas-
been acknowledged that the concept of the bone–vascular axis ingly been recognized. Specifically, the mild chronic increase in
may explain, for example, the relationship between bone metab- blood and tissues of an ever-growing number of cytokines is as-
olism and vessel wall diseases like atherosclerosis and arterio- sociated with worse clinical outcomes [2]. Another recent dis-
sclerosis, with potential involvement of a number of cytokines covery is the osteoblastic niche, the functional unit where stem
and metabolic pathways. A very important discovery in bone cells, precursors of both haematopoietic and bone cell lineages
physiology is the bone marrow (BM) niche, the functional unit (respectively HSPCs and MSCs), share a common environ-
where stem cells interact, exchanging signals that impact on ment. In this niche, these cells exchange signals that modify
their fate as bone-forming cells or immune-competent haema- their fate in terms of differentiation towards immune-
topoietic elements. This new element of bone physiology has competent haematopoietic elements (among which osteoclasts
been recognized to be dysfunctional in diabetes (so-called dia- are now included) and bone-forming cells (i.e. osteoblasts and
betic mobilopathy), with possible clinical implications. In our osteocytes). There is evidence that HSPCs and MSCs share
opinion, ROD, the metabolic bone disease of renal patients, will functional integrations of potential clinical relevance [3]. The
in the future probably be identified as a cause of BM niche dys- aim of this review is to highlight the emerging potential links
function. An integrated view of bone, which includes the BM between bone, chronic inflammation and the bone marrow
niche, now seems necessary in order to understand the complex (BM) niche, in particular in chronic kidney disease (CKD)

C The
Downloaded from
V Author(s) 2018. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved.
https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858 1
by University College London user
on 05 May 2018
patients. These patients are special because they suffer a specific Sost, produced by osteocytes, is a powerful inhibitor of the
type of metabolic bone disease and low-grade chronic inflam- canonical Wnt (wingless-type mouse mammary tumour virus
mation that can affect, in still underappreciated ways, the func- integration site) pathway and, as such, is both an inhibitor of
tion of the osteoblastic niche; dysfunction of the latter has osteoblasts and osteocytes and a promoter of osteoclast activity
recently been recognized for the first time in diabetes [4]. [19]. Indeed, human inactivating mutations of the Sost gene are
associated with sclerosteosis [20], while absence of mechanical
BONE, INTER-ORGAN COMMUNICATIONS load (e.g. due to inactivity or absence of gravity) increases Sost
AND CKD expression in bone, promoting bone resorption and osteoporo-
sis (OP) [21]. Also, Sost stimulates FGF23 synthesis, thus exert-
Until recently, bone was perceived as the repository of ions— ing indirect effects on mineral metabolism [22]. Importantly,
mainly calcium (Ca) and phosphate (P)—whose movement extraskeletal roles are envisaged, since Sost mRNA expression
into and out of bone was regulated by parathyroid hormone has been described [23], although not consistently [24], in calci-
(PTH) and active vitamin D through actions on osteoblasts and fied aortic valves of haemodialysis patients, while higher circu-
osteoclasts. This view changed with the discovery that osteo- lating levels are invariably associated with aortic valve
cytes, previously thought to be inactive, in fact orchestrate oste- calcification [23, 24]. Vessel wall media layer calcification, com-
oblast and osteoclast activity, including the synthesis of proteins monly identified as arteriosclerosis, is a typical finding of age-
with hormonal or hormone-like properties. Accordingly, bone ing, diabetes and CKD whose pathogenesis is now regarded as a
is now recognized as an endocrine organ [5], and at least three cell-mediated active process involving osteoblast-like cells de-
bone proteins have been claimed to have potential systemic rived from vascular smooth muscle cells. Therefore, a modula-
effects: osteocalcin (OC), fibroblast growth factor 23 (FGF23), tor of osteoblast activity like Sost could well be involved.
and sclerostin (Sost). Hypothetically, increased tissue levels of Sost might reflect local
OC, the most abundant non-collagenous bone protein, is inhibition of the osteoblast-like cell-mediated process of calcifi-
produced by osteoblasts during bone synthesis; however, during cation or high circulating levels of Sost might be a marker of
osteoclastic resorption OC is transformed into undercarboxy- low bone turnover (which in turn would favour vascular calcifi-
lated OC (UcOC), which is freed into blood. By binding to a G cations). Thus, the extraskeletal effects of Sost have potential
protein-coupled receptor in beta pancreatic cells, circulating clinical implications for cardiovascular diseases and represent a
UcOC affects insulin sensitivity and muscle energy metabolism. new player in the bone–vascular axis.
Accordingly, OC is regarded as a biochemical mediator of inter- Besides UcOC, FGF23 and Sost, which can be regarded as
organ communication between bone and muscle energy [6]. the principal mediators of the systemic ‘CKD-MBD syndrome’
FGF23, mainly produced by osteocytes, is a completely new [25], other non-collagenous bone proteins like bone alkaline
player in the field of so-called CKD–mineral and bone disorders phosphatase [26], the Small Integrin-Binding Ligand, N-linked
(CKD-MBD) [7]. By binding to FGF receptors, and in the pres- Glycoprotein (SIBLING) proteins [18], DKK-1 and activin A
ence of a co-receptor, alpha-Klotho, which is mainly produced [27] are under investigation to elucidate links between bone dis-
in the kidney [8], circulating FGF23 regulates P handling and ease, derangements of divalent ion metabolism, and the burden
vitamin D synthesis in renal tubular cells. The FGF23/Klotho of mortality and morbidity (in particular cardiovascular) car-
system is essential for P and vitamin D metabolism and repre- ried by CKD-MBD [25].
sents the biochemical substrate of the inter-organ communica- In summary, bone is no longer to be viewed as a lifeless
tion between bone and kidney, useful for better classification of framework for muscle action; rather, it is a sophisticated organ
some rare diseases [9]. Notably, in end-stage renal disease functionally connected with muscle energy metabolism, with
(ESRD) circulating FGF23 levels increase dramatically as an ex- the kidney and with the cardiovascular system (Figure 1). CKD
treme bone response to the burden of P load and altered catabo- deeply disturbs bone physiology (as reflected by changes in cir-
lism. This strong interaction between P and FGF23 has culating biomarkers), impairs its mechanical competence (as
widened the spectrum of P toxicity to include, besides second- reflected by an increased fracture rate) and contributes to the
ary hyperparathyroidism, cardiovascular disease (similarly to complex endocrinopathy now associated with worse cardiovas-
cholesterol) [10, 11]. In fact, higher quartiles of FGF23 have cular outcomes. For this reason, there is growing awareness that
been associated with poor cardiovascular outcomes in renal in renal patients it is important to recognize the specific types of
[12–14] and non-renal patients [15], probably due to a direct, renal osteodystrophy (ROD) through a bone biopsy, which is
receptor-mediated effect of FGF23 on myocardiocytes that is not routinely performed although it is minimally invasive and
capable of inducing left ventricular hypertrophy and myocar- is considered the diagnostic gold standard [28].
dial fibrosis independently of the FGF co-receptor Klotho [16].
Further, Ca deficiency reduces circulating levels of FGF23, thus BONE AND INFLAMMATION
decreasing the FGF23-mediated inhibition of 1, 25(OH)2D3, The link between bone and inflammation is evidenced in the pro-
which would exacerbate hypocalcaemia [17]. FGF23 synthesis cess of bone fracture repair, which is, in fact, a true acute inflam-
in bone is regulated by a number of bone proteins with either matory response of the innate immunity type. At the site of a
inhibitory or stimulatory effects [18]. Therefore, FGF23 can fracture, bone cells and inflammatory cells are recruited, with re-
also be regarded as an inter-organ communication factor be- sultant intense crosstalk between HSPC (monocyte–macrophage–
tween bone and the heart. osteoclast) and MSC (pre-osteoblast–osteoblast) derived cells [29].

Downloaded from2 https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858 S. Mazzaferro et al.


by University College London user
on 05 May 2018
FIGURE 1: UcOC, FGF23 and Sost are the principal mediators of the systemic metabolic effects of bone. UcOC increases insulin response of
target organs and is involved with energy metabolism. Klotho-dependent receptor-mediated effects of FGF23 mainly affect mineral metabo-
lism, while Klotho-independent effects on heart and liver produce systemic effects. Sost, a major regulator of bone cells activity, may have a
role in vascular calcifications. Mineral and bone disorders resulting from CKD are expected to influence these physiologic links.

T lymphocytes (which can stimulate osteoclastogenesis) and B animals. This hypothesis suggests that acute inflammation induces
cells [which can regulate the receptor activator of nuclear factor a ‘sickness behaviour’ (malaise, fatigue, anorexia, etc.) that, in the
jB (RANK)/receptor activator of nuclear factor jB ligand affected animals, is necessary to spare the energy required by the
(RANKL)/osteoprotegerin (OPG) axis] are also involved, with immune response. This adaptive behaviour relies on a complex in-
eventual increases in circulating cytokines [30]. Obviously, this tegrated energetic-neuroendocrine-immune response that
healing process is the same even in the case of asymptomatic includes increased bone resorption to guarantee sufficient
microfractures, so that pathologic increases in microfractures can amounts of two vital ions like Ca and P, which a resting animal
turn into systemic inflammation. Upon reflection, any chronic in- would not be able to gather. This scenario offers support for the
flammatory state can be expected to affect bone cell activity, as is importance of the above-mentioned link between bone and en-
illustrated by rheumatoid arthritis (RA), OP and atherosclerosis. ergy. Further, one can envisage that, inasmuch as the healing pro-
In RA, increases in circulating inflammatory cytokines [tumour cess is incomplete and becomes chronic, it will become
necrosis factor-alpha (TNF-a), interleukin (IL)-17, RANKL] stim- maladaptive and responsible for inflammation-related osteopae-
ulate osteoclast maturation and activity, thereby increasing bone nia (so-called smoldering inflammation) [35] (Figure 2).
resorption, while increases in DKK-1 and Sost inhibit bone forma- The link between atherosclerosis, inflammation and bone
tion [31], inducing OP. Ageing, recently regarded as ‘inflammag- deserves special consideration. Atherosclerosis is a chronic in-
ing’ [32], i.e. a chronic inflammatory condition, is considered to flammatory process in all of its stages. The effect of atheroscle-
induce senile OP through similar immunologic mechanisms. rosis, as an inflammatory disease, on bone metabolism and the
Therefore, the link between bone cells and inflammatory cells is development of OP is suggested by observations confirming
well established, as is encapsulated in the new term ‘osteoimmu- that decreased bone mineral density is a good predictor of car-
nology’ [33]. As for the link with CKD, a recent paper has diovascular events and coronary disease in postmenopausal
highlighted how CKD could be regarded as a model of accelerated women and men >50 years [36]. Moreover, growing evidence
ageing, with resultant bone and cardiovascular disease [34]. indicates the existence of a correlation between OP and athero-
Interestingly, according to a recent hypothesis, the link between sclerosis regardless of age, body mass index and cardiovascular
bone and inflammation may be of evolutionary value in terrestrial risk factors [37]. Furthermore, chronic inflammatory processes

Bone,https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858
Downloaded from inflammation and bone marrow niche in CKD 3
by University College London user
on 05 May 2018
FIGURE 2: Bone fracture elicits an acute inflammatory response that is energy demanding and associated with a ‘sickness behaviour’ usually
ending with complete recovery. At variance, multiple diffuse microfractures may lead to chronic subtle inflammation and to osteopaenia.

contribute to vascular calcification, and the common finding of NO inhibits platelet adherence and aggregation, suppresses
simultaneous vascular calcification and OP in individual vasoconstriction, reduces the adherence of leucocytes to the en-
patients suggests that local tissue factors govern the regulation dothelium, and suppresses the proliferation of vascular smooth
of biomineralization [38]. New terms like ‘calcification paradox’ muscle cells. Therefore, a reduction in NO activity contributes
[39] and ‘osteocardiology’ [40] are being coined to illustrate this to a proinflammatory and prothrombotic milieu.
clinical link. Vascular calcification in atherosclerosis is triggered In CKD, an increase in the inflammatory biomarkers TNF-
by the response to injury caused by oxidized low-density lipo- a, IL-6, IL-1, CRP and fibrinogen has been reported in the
protein (LDLox). LDLox initiates the inflammatory process, blood [44]. This increase is caused by several mechanisms, in-
which is amplified by the exposure of adhesion molecules and cluding reduced clearance of proinflammatory cytokines, in-
by the secretion of interleukins, C-reactive protein (CRP) and creased local production (e.g. due to the blood–membrane
bone morphogenetic proteins (BMPs) by endothelial cells contact in dialysis patients), pathologic permeability of gut to
and smooth muscle cells. All these processes promote increased toxins (so-called leaky gut syndrome) and induction of macro-
oxidative stress and decreased calcification inhibitors, such as phage activation by metabolic acidosis [45–47]. Monocytes and
matrix Glutamic acid (Gla)-protein and osteopontin. macrophages are increased in the peripheral blood of uraemic
Experimental evidence implies that atherosclerotic inflamma- patients even when there is no clinical evidence of an active in-
tory activity has an interrelationship with osteogenic modula- flammatory process or an increase in the peripheral blood of
tion. When exposed to LDLox, endothelial cells express BMPs. other inflammatory markers, such as CRP or proinflammatory
Additionally, TNF-a and interferon-gamma stimulate the en- cytokines [48]. Further, in CKD the mitochondrial respiratory
dothelium to express OPG, which is also produced in osteo- system is impaired, which may be both a consequence and a
blasts and in smooth muscle cells when stimulated with cause of enhanced oxidative stress. Through elevated produc-
proinflammatory interleukins [41]. Hyperproduction of inflam- tion of reactive oxygen species (ROS), the damaged mitochon-
matory markers such as CRP, IL-1, IL-6 and TNF-a is directly dria of uraemic patients may be able to activate the NLRP3
related to the severity of atherosclerosis and the stimulation of inflammasome, a deregulated biological system newly identified
osteoclastogenesis [42]. in CKD-5D patients [49–52]. Increased generation of ROS in
Monocytes and macrophages (after HSPC recruitment from chronic renal failure can damage proteins, lipids and nucleic
BM) are the dominant type of atherosclerotic inflammatory cell acids and consequently influence cell function, inhibit enzy-
infiltrates and represent more than half of all cells at the imme- matic activities of the cellular respiratory chain and accelerate
diate site of plaque rupture. Furthermore, leakage of cytokines progression of CKD [53]. Changes in oxidative and antioxidant
and leukotrienes from activated macrophages in the atheroscle- status, which occur from the early stages of CKD, may be exac-
rotic plaque enriches the systemic proinflammatory milieu [43]. erbated by haemodialysis [54, 55]. Therefore, oxidative stress
Another implicated factor is endothelium-derived nitric oxide and chronic inflammation are both important players in the
(NO), which is reduced at the site of vascular injury. Indeed, mechanisms underlying CKD-related accelerated atherogenesis

Downloaded from4 https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858 S. Mazzaferro et al.


by University College London user
on 05 May 2018
FIGURE 3: CKD is a chronic inflammatory state with increased circulating pro-inflammatory cytokines (IL-17, TNF-a, RANKL, BMPs, etc.)
and decreased calcification inhibitors (MGP, OPN, etc.). Circulating monocytes and enhanced oxidative stress (via the NLRP3 inflammasome)
accelerate atherogenesis. Local response to the injury caused by LDLox is amplified by exposure of adhesion molecules and secretion of inter-
leukins and BMP by endothelial cells and smooth muscle cells. All these processes further increase systemic oxidative stress, decrease calcifying
inhibitors and promote vessel wall calcification. ROD, by interfering with BM niche function, is expected to contribute to this systemic micro-
inflammatory burden that accelerates atherosclerosis, vessel calcification and osteopaenia.

and ageing [34]. In turn, accelerated atherogenesis will nega- are key modulators of HSPCs, keeping them quiescent for the
tively affect bone metabolism (Figure 3). purposes of maintenance and self-renewal. The second element
is the vascular niche, composed of vascular sinuses, lining endo-
THE BM NICHE: WHERE BONE AND MARROW thelial cells, CXCL12-abundant reticular (CAR) cells, sympa-
MEET thetic neurons and HSPCs. Each HSPC evolves inside a
Bone is the chest for BM cells and the place where the egress of specialized and specific niche [57]. Egress of HSPCs out of the
stem cells, including those already committed towards some spe- BM and into the bloodstream is known as ‘mobilization’ and is
cific lineages, is orchestrated [56]. The transfer of HSPCs out of the coupled with HSPC ‘homing’. Homing is a set of complex path-
BM to the circulation requires the integrity of bone microarchitec- ways that modulate the mobilization of HSPCs towards both
ture, within which is contained the BM functional unit: the niche. peripheral BM niches and peripheral tissues [59].
A niche is defined as a specialized microenvironment of the The niche composition and function ultimately depends on
BM, specific for each cell lineage, that hosts and modulates the activity of the bone cells since most HSPCs are found in the
HSPC renewal and egress into the bloodstream. Inside the trabecular bone, suggesting that the function of the niche (mo-
niche, a complicated network of hormones, soluble mediators bilization and homing) may also be regulated by factors in-
and surface cell receptors regulates the HSPC number, fate and volved in bone remodelling [57, 58, 60].
location [57]. The niche is perivascular and located within the A high number of osteoblasts raises the stem cell pool size
trabecular bone and is settled by osteoblastic cells, endothelial and adherence in the niche, whereas an increase in osteoclasts
cells and perivascular MSCs that interact closely with each other degrades the niche and promotes the egress of HSPCs [61].
[57, 58]. The BM niche consists of two major elements. The first These processes are physiologically carried by the joint effect
is the osteoblastic niche, where cells of the osteoblastic lineage of PTH and inflammatory cytokines. PTH plays the role of

Bone,https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858
Downloaded from inflammation and bone marrow niche in CKD 5
by University College London user
on 05 May 2018
pivotal director of the niche through activation of PTH/PTHrP proinflammatory osteal macrophages [4, 77, 78]. This novel type
receptors (PPRs), leading to HSPC expansion. Following PPR of diabetic complication, termed ‘stem cell mobilopathy’, is the
activation, osteoblastic cells produce high levels of the Notch pathway via which diabetes accelerates atherosclerosis; it does this
ligand, jagged-1, which elicits an increase in the number of by inducing a shortage of vascular regenerative cells and by shift-
HSPCs [58, 62, 63]. Furthermore, in osteoblasts PTH upregu- ing the differentiation of BM progenitor cells to pro-calcific [77].
lates both granulocyte colony-stimulating factor (GCSF), which In CKD patients the uraemic inflammatory burden is en-
in turn regulates the expression of inflammatory cytokines hanced by the frequent coexistence of diabetes, atherosclerosis
(IL-6 and IL-11) and CXCL12. CXCL12 is the hinge chemokine and ageing [34]. In addition, in CKD patients, inflammation is
involved in the mobilization and homing processes as its inter- triggered by specific pathways related to the CKD-MBD syn-
action with the homing receptor CXCR-4, expressed on many drome. Besides PTH, FGF23 can directly bind and activate
progenitors, is the most important pathway for retention of FGFR4 and calcineurin/NFAT signaling in hepatocytes in the ab-
HSPCs within the BM as well as for their mobilization. GCSF sence of its classic co-receptor alpha-Klotho, leading to increased
depletes osteoblasts and reduces CXCL12 expression in both expression and secretion of inflammatory cytokines (Figure 1).
osteoblasts and CAR cells, so promoting mobilization of HSPCs The relationship between FGF23 and inflammation seems to
into the vascular sinuses [57, 58, 64]. be bidirectional since inflammation increases FGF23 transcription
No less relevant, within the BM niche, is the role of proin- in osteocytes. However, whether FGF23 stimulates inflammatory
flammatory cytokines, which maintain the HSPC pool by cytokine expression by other target cells such as adipocytes and
tuning size, cell lineage, distribution and phenotype [65]. osteoblasts is a matter of discussion [79]. FGF23 may influence
Inflammatory cytokines also affect the phenotype of BM mac- the bone microarchitecture by directly tuning bone remodelling.
rophages (also called osteal macrophages): this process is FGF23, through a soluble Klotho/MAPK-mediated process in-
known as macrophage polarization. Osteal macrophages are in- volving Dkk1 expression, inhibits the osteoblastic Wnt pathway,
volved in bone repair and remodelling by regulating the cross- so contributing to bone loss in CKD [69]. Moreover, both human
talk between osteoclasts and osteoblasts [66, 67]. pre-osteoclasts and mature osteoclasts express FGFR1 at all stages
Besides PTH, other factors involved in bone remodelling, of differentiation, and FGF23 displays biphasic effects on human
such as FGF23/klotho, Wnt inhibitors, vitamin D, vitamin D re- osteoclasts, with inhibition of osteoclast differentiation at the early
ceptor and Ca sensing receptor (CaSR), are able to influence the stages of maturation and stimulation of activity at later stages [80].
activity of the BM niche and the HSPC fate [9, 22, 68–70]. CKD-related inflammation and bone mineral disorders
Moreover, MSCs give rise to osteoblasts, and their differentia- could affect the complex balance within the BM niche, thus de-
tion is stimulated by 1, 25(OH)2D. In addition to being targets ranging the mobilization and homing of HSPCs and fostering
of 1, 25(OH)2D, MSCs can synthesize it [71, 72]. Normal CaSR the development of cell subsets expressing an osteogenic pheno-
expression on HSPCs is an absolute requirement for their lodg- type. This latter finding may represent a common thread that
ing in the endosteal niche of the BM [73]. links bone remodelling, the BM niche and vascular calcification
Given these tight morphological and functional links, any in chronic renal failure.
pathological condition able to induce an imbalance in bone Cells with an osteogenic phenotype may originate from vas-
remodelling and a derangement in cell signaling may disrupt cular wall-resident MSCs, transdifferentiated mature or circulat-
both the bone microarchitecture and the BM niche function ing vascular smooth muscle cells, or circulating calcifying cells
and consequently the HSPC traffic [74]. (CCCs) [75, 81]. CCCs comprise several osteogenic cell subsets
All these findings are new features of the complex scenario that express different but interrelated phenotypes, share a com-
of the bone–vascular axis. In fact, HSPCs and the precursors of mon origin from BM progenitor cells, and are able to promote
cardiovascular cells may also be resident in the vascular or val- intimal calcification. Regardless of the type of BM progenitor
vular wall or be part of the uninterrupted flow of HSPCs ensur- cell, CCCs are defined by the expression of OC and bone alkaline
ing adequate cell renewal and contributing physiologically to phosphatase. Their pool includes circulating (mesenchymal)
vascular health [75, 76] (Figure 4). osteoprogenitor cells, circulating calcifying endothelial progeni-
Several chronic diseases such as obesity, atherosclerosis, diabe- tor cells (EPCs) and myeloid calcifying cells (MCCs) [82]. EPCs
tes and CKD display a unique proinflammatory milieu that, along have been associated with coronary artery disease, calcific aortic
with a cluster of metabolic derangements and oxidative stress fac- stenosis, OP, diabetes and ESRD [72, 83–85]. MCCs belong to
tors, may impair mobilization and homing, thereby inducing the myeloid lineage (monocytes–macrophages) and have been
shortage and functional impairment of HSPCs, shifting the pro- found to be significantly increased in the presence of either car-
genitor cell phenotype and ultimately influencing pathological diovascular disease or diabetes. In addition, MCC numbers are
processes such as atherosclerosis and vascular calcification. higher in diabetic versus non-diabetic patients regardless of the
Diabetes is characterized by a broad derangement of the BM coexistence of cardiovascular disease, and they are also increased
niche, with an expanded pool of quiescent HSPCs as well as a re- in the BM and atherosclerotic plaques [86].
duced number of osteoblasts slightly expressing CXCL12 and
unchanged CXCL12 expression in CAR cells, resulting in de- BONE, INFLAMMATION AND THE BM NICHE
creased mobilization of haematopoietic stem cells [4]. These IN CKD: FINAL CONSIDERATIONS
changes are acknowledged to be driven by chronic inflammation, The role of BM niche impairment in vascular disease in the set-
stimulation of innate immunity receptors, and an increase in ting of diabetes and atherosclerosis has been assessed and such

Downloaded from6 https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858 S. Mazzaferro et al.


by University College London user
on 05 May 2018
FIGURE 4: The fate of HSPCs out of the BM physiologically reflects the joined action of bone remodelling and inflammatory cytokines. PTH,
by acting on osteoblasts both directly and indirectly (through JAG 1, IL-6 and GCSF), is pivotal director of HSPCs ‘mobilization’ and ‘homing’
into the bloodstream. Other factors involved with bone metabolism like FGF23, calcitriol, their receptors and the Ca-sensing receptor are
relevant regulators of BM niche function. Their derangements in CKD, together with the resulting damage in terms of bone turnover and
microarchitecture, are most likely responsible for BM niche dysfunction.

impairment is also considered to be the first step in the process affect the function of the BM niche. Similarly, the different types
leading to the appearance of CCCs. This is undoubtedly the of ROD (e.g. high- or low-turnover bone disease with resulting
most intriguing, though still relatively uncharted area in the differences in bone cell numbers and activity) most probably
multifaceted scenario of the bone–vascular axis. The discovery impact the BM niche. Therefore, assessment of BM niche func-
of BM niche impairment in vascular disease is particularly im- tion in CKD patients could be important for the discovery of
portant considering that the derangement of the bone–vascular new pathways in the complex metabolic disturbance of uraemia
axis is amplified by ageing and by CKD, diabetes and athero- and its heavy burden of morbidity and mortality.
sclerosis, the incidence of which is constantly rising in the gen-
eral population. It is possible to speculate that inflammation is
CONFLICT OF INTEREST STATEMENT
the shared pathogenetic link, also bearing in mind the possible
coexistence of diabetes, atherosclerosis and CKD, and the po- None declared. The results presented in this article have not been
tential effects on bone remodelling and thus on BM niche func- published previously in whole or part, except in abstract format.
tion. Studies on impairment of the BM niche in CKD are still at
an early stage. This is all the more surprising if we consider the REFERENCES
crucial role that PTH, CaSR, FGF23, vitamin D, inflammatory 1. Vervloet M, Massy Z, Brandenburg V et al. Bone: a new endocrine organ at
cytokines and bone cells are acknowledged to play in regulating the heart of chronic kidney disease and mineral and bone disorders. Lancet
the expansion, mobilization and homing of HSPCs. The inflam- Diabetes Endocrinol 2014; 2: 427–436
matory burden and the impairment of bone remodelling inher- 2. Tousoulis D, Economou E, Oikonomou E et al. The role and predictive
value of cytokines in atherosclerosis and coronary artery disease. Curr Med
ent to the CKD-MBD syndrome most likely compromise the
Chem 2015; 22: 2636–2650
renewal of HSPCs and the provision of cell progenitors to vas- 3. Reagan M, Rosen C. Navigating the bone marrow niche: translational
cular tissues, as well as promoting the development of cell sub- insights and cancer-driven dysfunction. Nat Rev Rheumatol 2016; 12:
sets that express an osteogenic phenotype and thus probably 154–168
affect the process of vascular calcification. Overall, like diabetes, 4. Di Persio J. Diabetic stem-cell ‘mobilopathy’. N Engl J Med 2011; 365:
2536–2538
CKD is a potential cause of BM niche dysfunction or mobilop-
5. Fukumoto S, Martin T. Bone as an endocrine organ. Trends Endocrinol
athy and this urgently needs to be appreciated. Indeed, the met- Metab 2009; 164: 230–236
abolic derangements of mineral metabolism and the chronic 6. Karsenty G, Olson E. Bone and muscle endocrine functions: unexpected
inflammatory burden of renal insufficiency can predictably paradigms of inter-organ communication. Cell 2016; 164: 1248–1256

Bone,https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858
Downloaded from inflammation and bone marrow niche in CKD 7
by University College London user
on 05 May 2018
7. Cozzolino M, Mazzaferro S. The fibroblast growth factor 23: a new player in 34. Covic A, Vervloet M, Massy ZA et al. Bone and mineral disorders in chronic
the field of cardiovascular, bone and renal disease. Curr Vasc Pharmacol kidney disease: implications for cardiovascular health and ageing in the gen-
2010; 8: 404–411 eral population. Lancet Diabetes Endocrinol 2018; 6: 319–331
8. Kuro-O M, Moe O. FGF23-aklotho as a paradigm for a kidney-bone net- 35. Straub R, Cutolo M, Pacifici R. Evolutionary medicine and bone loss in
work. Bone 2017; 100: 4–18 chronic inflammatory diseases - a theory of inflammation-related osteope-
9. Mazzaferro S, Pasquali M, Pirrò G et al. The bone and the kidney. Arch nia. Semin Arthritis Rheum 2015; 45: 220–228
Biochem Biophys 2010; 503: 95–102 36. Von der Recke P, Hansen M, Hassager C. The association between low bone
10. Ellam T, Chico T. ‘Phosphate: the new cholesterol? The role of the phos- mass at the menopause and cardiovascular mortality. Am J Med 1999; 106:
phate axis in non-uremic vascular disease. Atherosclerosis 2012; 220: 273–278
310–318 37. Schulz E, Arfai K, Liu X et al. Aortic calcification and the risk of osteoporo-
11. Cozzolino M, Gentile G, Mazzaferro S et al. Blood pressure, proteinuria, sis and fractures. J Clin Endocrinol Metab 2004; 89: 4246–4253
and phosphate as risk factors for progressive kidney disease: a hypothesis. 38. Parhami F, Morrow A, Balucan J et al. Lipid oxidationproducts have oppo-
Am J Kidney Dis 2013; 62: 984–992 site effects on calcifying vascular celland bone cell differentiation. A possible
12. Gutiérrez O, Mannstadt M, Isakova T et al. Fibroblast growth factor 23 and explanation for the paradox of arterial calcification in osteoporoticpatients.
mortality among patients undergoing hemodialysis. N Engl J Med 2008; 1997; 17: 680–687
359: 584–592 39. Persy V, D’Haese P. Vascular calcification and bone disease: the calcification
13. Scialla J, Xie H, Rahman M et al. Fibroblast growth factor-23 and cardiovas- paradox. Trends Mol Med 2009; 15: 405–416
cular events in CKD. J Am Soc Nephrol 2014; 25: 349–360 40. Rajamannan N. Osteocardiology: The Atherosclerotic Bone Paradox.
14. Gutiérrez O, Januzzi J, Isakova T et al. Fibroblast growth factor-23 and left Springer, Cham, 2018
ventricular hypertrophy in chronic kidney disease. Circulation 2009; 119: 41. Liberman M, Bassi E, Martinatti M et al. Oxidant generation predominates
2545–2552 around calcifying foci and enhances progression of aortic valve calcification.
15. Mirza M, Larsson A, Melhus H et al. Serum intact FGF23 associate with left Arterioscler Thromb Vasc Biol 2008; 28: 463–470
ventricular mass, hypertrophy and geometry in an elderly population. 42. Danilevicius C, Lopes J, Pereira R. Bone metabolism and vascular calcifica-
Atherosclerosis 2009; 207: 546–551 tion. 2007; 40: 435–442
16. Grabner A, Amaral A, Schramm K et al. Activation of cardiac fibroblast 43. Bäck M, Weber C, Lutgens E. Regulation of atherosclerotic plaque inflam-
growth factor receptor 4 causes left ventricular hypertrophy. Cell Metab mation. J Intern Med 2015; 278: 462–482
2015; 22: 1020–1032 44. Gupta J, Mitra N, Kanetsky PA et al. Association between albuminuria, kid-
17. Rodriguez-Ortiz M, Lopez I, Mu~ noz-Casta~ neda J et al. Calcium deficiency
ney function, and inflammatory biomarker profile in CKD in CRIC. Clin J
reduces circulating levels of FGF23. J Am Soc Nephrol 2012; 23: 1190–1197
Am Soc Nephrol 2012; 7: 1938–1946
18. Rowe P. A unified model for bone-renal mineral and energy metabolism.
45. Fallahzadeh M, Roozbeh J, Geramizadeh B et al. Interleukin-2 serum levels
Curr Opin Pharmacol 2015; 22: 64–71
are elevated in patients with uremic pruritus: a novel finding with practical
19. Kobayashi Y, Uehara S, Udagawa N et al. Regulation of bone metabolism by
implications. Nephrol Dial Transplant 2011; 26: 3338–3344
Wnt signals. J Biochem 2016; 159: 387–392
46. Sitter T, Bergner A, Schiffl H. Dialysate related cytokine induction and re-
20. Hamersma H, Gardner J, Beighton P. The natural history of sclerosteosis.
sponse to recombinant human erythropoietin in haemodialysis patients.
Clin Genet 2003; 63: 192–197
Nephrol Dial Transplant 2000; 15: 1207–1211
21. Robling A, Niziolek P, Baldridge L et al. Mechanical stimulation of bone
47. Bellocq A, Suberville S, Philippe C et al. Low environmental pH is responsi-
in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem 2008;
ble for the induction of nitric-oxide synthase in macrophages. Evidence for
283: 5866–5875
involvement of nuclear factor-kappaB activation. J Biol Chem 1998; 273:
22. Ryan Z, Ketha H, McNulty M et al. Sclerostin alters serum vitamin D me-
tabolite and fibroblast growth factor 23 concentrations and the urinary ex- 5086–5092
48. RamirezCarracedo R, Berdud JI et al. Microinflammation in hemodialysis
cretion of calcium. Proc Natl Acad Sci USA 2013; 110: 6199–6204
23. Brandenburg V, Kramann R, Koos R et al. Relationship between sclerostin is related to a preactivated subset of monocytes. Hemodial Int 2006; 10
and cardiovascular calcification in hemodialysis patients: a cross-sectional (Suppl 1): S24–S27
study. BMC Nephrol 2013; 14: 219 49. Anders H, Muruve D. The inflammasomes in kidney disease. J Am Soc
24. Qureshi A, Olauson H, Witasp A et al. Increased circulating sclerostin levels Nephrol 2011; 22: 1007–1018
in end-stage renal disease predict biopsy-verified vascular medial calcifica- 50. Anders H, Banas B, Schlöndorff D. Signaling danger: toll-like receptors
tion and coronary artery calcification. Kidney Int 2015; 88: 1356–1364 and their potential roles in kidney disease. J Am Soc Nephrol 2004; 15:
25. Cozzolino M, Ure~ na-Torres P, Vervloet M et al. Is chronic kidney disease- 854–867
mineral bone disorder (CKD-MBD) really a syndrome?. Nephrol Dial 51. Granata S, Zaza G, Simone S et al. Mitochondrial dysregulation and oxidative
Transplant 2014; 29: 1815–1820 stress in patients with chronic kidney disease. BMC Genomics 2009; 10: 388
26. Haarhaus M, Brandenburg V, Kalantar-Zadeh K et al. Alkaline phospha- 52. Granata S, Masola V, Zoratti E et al. NLRP3 inflammasome activation in di-
tase: a novel treatment target for cardiovascular disease in CKD. Nat Rev alyzed chronic kidney disease patients. PLoS One 2015; 10: e0122272
Nephrol 2017; 13: 429–442 53. Modaresi A, Nafar M, Sahraei Z. Oxidative stress in chronic kidney disease.
27. Hruska K, Sugatani T, Agapova O et al. The chronic kidney disease - min- Iran J Kidney Dis 2015; 9: 165–179
eral bone disorder (CKD-MBD): advances in pathophysiology. Bone 2017; 54. Canaud B, Cristol J, Morena M et al. Imbalance of oxidants and antioxi-
100: 80–86 dants in haemodialysis patients. Blood Purif 1999; 17: 99–106
28. Torres PU, Bover J, Mazzaferro S et al. When, how and why a bone biopsy 55. Morena M, Delbosc S, Dupuy A et al. Overproduction of reactive oxygen
should be performed in patients with chronic kidney disease. Sem Nephrol species in end-stage renal disease patients: a potential component of
2014; 34: 612–625 hemodialysis-associated inflammation. Hemodial Int 2005; 9: 37–46
29. Loi F, Córdova L, Pajarinen J et al. Inflammation, fracture and bone repair. 56. Thompson B, Towler D. Arterial calcification and bone physiology: role of
Bone 2016; 66: 119–130 the bone-vascular axis. Nat Rev Endocrinol 2012; 8: 529–543
30. Pietschmann P, Mechtcheriakova D, Meshcheryakova A et al. Immunology 57. Morrison S, Scadden D. The bone marrow niche for haematopoietic stem
of osteoporosis: a mini-review. Gerontology 2016; 62: 128–137 cells. Nature 2014; 505: 327–334
31. D’Amelio P, Sassi F. Osteoimmunology: from mice to humans. Bonekey Rep 58. Hoffman C, Calvi L. Minireview: complexity of hematopoietic stem cell reg-
2016; 5: 802 ulation in the bone marrow microenvironment. Mol Endocrinol 2014; 28:
32. Franceschi C, Bonafè M, Valensin S et al. Inflamm-aging. An evolutionary 1592–1601
perspective on immunosenescence. Ann N Y Acad Sci 2000; 908: 244–254 59. Brunner S, Huber B, Fischer R et al. G-CSF treatment after myocardial in-
33. Arron J, Choi Y. Osteoimmunology: bone versus immune system. Nature farction: impact on bone marrow-derived vs cardiac progenitor cells. Exp
2000; 408: 535–536 Hematol 2008; 36: 695–702

Downloaded from8 https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858 S. Mazzaferro et al.


by University College London user
on 05 May 2018
60. Fadini G, Ferraro F, Quaini F et al. Concise review: diabetes, the bone mar- 75. Fadini G. Task for today: complete the puzzle of circulating stem cells and
row niche, and impaired vascular regeneration. Stem Cells Transl Med 2014; the atherosclerotic burden. Circ Res 2016; 119: 502–504
3: 949–957 76. Zhang L, Xu Q. Stem/Progenitor cells in vascular regeneration. Arterioscler
61. Hoggatt J, Pelus L. Mobilization of hematopoietic stem cells from the bone Thromb Vasc Biol 2014; 34: 1114–1119
marrow niche to the blood compartment. Stem Cell Res Ther 2011; 2: 13 77. Fadini G, Ciciliot S, Albiero M. Concise review: perspectives and clinical
62. Calvi L, Adams G, Weibrecht K et al. Osteoblastic cells regulate the haema- implications of bone marrow and circulating stem cell defects in diabetes.
topoietic stem cell niche. Nature 2003; 425: 841–846 Stem Cells 2017; 35: 106–116
63. Porter R, Calvi L. Communications between bone cells and hematopoietic 78. Albiero M, Poncina N, Ciciliot S et al. Bone marrow macrophages contrib-
stem cells. Arch Biochem Biophys 2008; 483: 193–200 ute to diabetic stem cell mobilopathy by producing oncostatin M. Diabetes
64. Huber B, Grabmaier U, Brunner S. Impact of parathyroid hormone on 2015; 64: 2957–2968
bone marrow-derived stem cell mobilization and migration. World J Stem 79. Singh S, Grabner A, Yanucil C et al. Fibroblast growth factor 23 directly tar-
Cells 2014; 6: 637–643 gets hepatocytes to promote inflammation in chronic kidney disease.
65. Mirantes C, Passegué E, Pietras E. Pro-inflammatory cytokines: emerging Kidney Int 2016; 90: 985–996
players regulating HSC function in normal and diseased hematopoiesis. Exp 80. Allard L, Demoncheaux N, Machuca-Gayet I et al. Biphasic effects of vita-
Cell Res 2014; 329: 248–254 min D and FGF23 on human osteoclast biology. Calcif Tissue Int 2015; 97:
66. Gibon E, Lu L, Goodman S. Aging, inflammation, stem cells, and bone heal- 69–79
ing. Stem Cell Res Ther 2016; 7: 44 81. Bobryshev Y. Transdifferentiation of smooth muscle cells into chondrocytes
67. Davies L, Jenkins S, Allen J et al. Tissue-resident macrophages. Nat in atherosclerotic arteries in situ: implications for diffuse intimal calcifica-
Immunol 2013; 14: 986–995 tion. J Pathol 2005; 205: 641–650
68. Komaba H, Kaludjerovic J, Hu D et al. Klotho expression in osteocytes regu- 82. Fadini G, Rattazzi M, Matsumoto T et al. Emerging role of circulating calci-
lates bone metabolism and controls bone formation. Kidney Int 2017; 92: fying cells in the bone-vascular axis. Circulation 2012; 125: 2772–2781
599–611 83. Rose J, Erzurum S, Asosingh K. Biology and flow cytometry of proangio-
69. Carrillo-López N, Panizo S, Alonso-Montes C et al. Direct inhibition of os- genic hematopoietic progenitors cells. Cytometry A 2015; 87: 5–19
teoblastic Wnt pathway by fibroblast growth factor 23 contributes to bone 84. Fadini G, Albiero M, Menegazzo L et al. Procalcific phenotypic drift of cir-
loss in chronic kidney disease. Kidney Int 2016; 90: 77–89 culating progenitor cells in type 2 diabetes with coronary artery disease. Exp
70. Goltzman D, Hendy G. The calcium-sensing receptor in bone–mechanistic Diabetes Res 2012; 2012: 1
and therapeutic insights. Nat Rev Endocrinol 2015; 11: 298–307 85. Gössl M, Mödder U, Atkinson E et al. Osteocalcin expression by circulating
71. Zhou S, Glowacki J. Chronic kidney disease and vitamin D metabolism in endothelial progenitor cells in patients with coronary atherosclerosis. J Am
human bone marrow-derived MSCs. Ann N Y Acad Sci 2017; 1402: 43–55 Coll Cardiol 2008; 33: 1314–1325
72. Cianciolo G, Capelli I, Cappuccilli M et al. Is chronic kidney disease- 86. Fadini G, Albiero M, Menegazzo L et al. Widespread increase in myeloid
mineral and bone disorder associated with the presence of endothelial pro- calcifying cells contributes to ectopic vascular calcification in type 2 diabetes.
genitor cells with a calcifying phenotype? Clin Kidney J 2017; 10: 389–396 Circ Res 2011; 108: 1112–1121
73. Drüeke T. Haematopoietic stem cells–role of calcium-sensing receptor in
bone marrow homing. Nephrol Dial Transplant 2006; 21: 2072–2074 Received: 10.1.2018; Editorial decision: 2.4.2018
74. Viaene L, Behets G, Heye S et al. Inflammation and the bone-vascular axis
in end-stage renal disease. Osteoporos Int 2016; 27: 489–497

Bone,https://academic.oup.com/ndt/advance-article-abstract/doi/10.1093/ndt/gfy115/4992858
Downloaded from inflammation and bone marrow niche in CKD 9
by University College London user
on 05 May 2018

Potrebbero piacerti anche