You are on page 1of 7

Immunology Letters 168 (2015) 129–135

Contents lists available at ScienceDirect

Immunology Letters
journal homepage: www.elsevier.com/locate/immlet

BMSCs and hematopoiesis


Andrés García-García a,b , Carlos L.F. de Castillejo a,b , Simón Méndez-Ferrer a,b,∗
a
Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
b
Stem Cell Institute and Department of Haematology, University of Cambridge, and National Health Service Blood and Transplant, Cambridge Biomedical
Campus, CB20PT Cambridge, United Kingdom

a r t i c l e i n f o a b s t r a c t

Article history: Recent discoveries have significantly expanded our previous knowledge about the role of bone marrow
Received 23 June 2015 mesenchymal stem cells (BMSCs) in hematopoiesis. BMSCs and their derivatives modulate blood pro-
Accepted 23 June 2015 duction and immunity at different levels but a prominent role has emerged for BMSCs in the regulation
Available online 17 July 2015
of hematopoietic stem and progenitor cells (HSPCs). Additionally, BMSC-like cells regulate B and T cell
lymphopoiesis and also probably myelopoiesis. Furthermore, BMSCs might also exhibit key regulatory
properties in non-physiological conditions. BMSCs in extramedullary sites might provide a permissive
microenviroment to allow for transient hematopoiesis. BMSCs might be also involved in the manifesta-
tion and/or the development of hematopoietic diseases, as stemming from their emerging roles in the
progression of hematological malignancies. Here we review some key molecular pathways, adhesion
molecules and ligand/receptor interactions that mediate the crosstalk between BMSCs and hematopoi-
etic stem cells (HSCs) in health and disease. The development of novel markers to visualize and isolate
individual cells will help to dissect the stromal–hematopoietic interplay.
Crown Copyright © 2015 Published by Elsevier B.V. All rights reserved.

1. BMSCs & HSPCs signaling [8]. Simultaneously, Zhang et al. also identified bone-
lining osteoblasts as key regulators of bone marrow niche size.
BMSCs and HSCs were described as the initiating cells responsi- In this case, the molecular pathway involved was BMP signaling
ble for bone marrow reconstitution after mechanical depletion of [9]. Angiopoietin-1 produced by endosteal cells was also found
marrow cavity (see Ref. [1] for a review). BMSCs lie at the top of the to promote HSC quiescence by binding to Tie2 receptor in HSCs
hierarchy of bone marrow stromal elements giving rise to cartilage, [10]. Angiopoietin-1 has been recently found to be mainly pro-
bone and adipose tissue. Since their discovery in the bone marrow duced by leptin receptor+ mesenchymal stromal cells and also
by Friedenstein et al. [2], it has been proposed that BMSCs support by HSPCs, which contribute in this way to regenerate their own
hematopoiesis through direct and indirect mechanisms. Dexter niche after irradiation [11]. Osteoblastic cells also regulate prim-
et al. devised murine long-term cultures in which bone marrow itive HSCs through thrombopoietin/MPL signaling [12], although
mesenchymal stromal cells could maintain hematopoiesis in vitro the megakaryocytes themselves have been recently proposed by
[3]. By preserving HSC properties in the bone marrow, BMSCs are the same group as the main source of thrombopoietin the bone
therefore key components of the so-called “stem cell niche” [4]. marrow [13]. However, more primitive mesenchymal cells pro-
When the first evidences of the existence of a stem cell gressively acquired a more prominent role in the maintenance and
niche in the bone marrow emerged, several studies suggested regulation of HSCs, compared with mature osteoblasts.
that osteoblastic cells could have an active role in the regula- In the mouse, BMSCs identified based on the expression of the
tion of hematopoiesis given the proximity between HSPCs and intermediate filament protein nestin were found highly enriched
the inner bone layer (“endosteum”) of the bone marrow [5–7]. In in fibroblastic colony-forming-unit activity and self-renewal capac-
2003 a turning point took place as Calvi et al. provided for the ity in serial transplantations. Nestin+ BMSCs express high levels of
first time in vivo data suggesting that osteoblastic lineage cells HSC maintenance genes, like Cxcl12 [14] and c-kit ligand, and their
are regulatory components of the HSC niche via Notch pathway depletion reduced HSC activity and bone marrow homing [15].
Mesenchymal progenitor activity was soon after found also
enriched in a bone marrow reticular stromal cell population charac-
∗ Corresponding author at: NHS Blood and Transplant. Cambridge Biomedical terized by high expression of the key HSC maintenance chemokine
Campus. Cambridge, CB2 0PT, UK. Cxcl12, therefore named Cxcl12-abundant reticular (CAR) cells
E-mail address: sm2116@cam.ac.uk (S. Méndez-Ferrer).

http://dx.doi.org/10.1016/j.imlet.2015.06.020
0165-2478/Crown Copyright © 2015 Published by Elsevier B.V. All rights reserved.
130 A. García-García et al. / Immunology Letters 168 (2015) 129–135

Fig. 1. Interactions between BMSCs identified using different markers and HSPCs. Diagram showing several pathways mediating the interaction of BMSCs identified using
markers like osterix (Osx), nestin, Leptin receptor, neural/glial antigen 2(NG2), Cxccl12::GFP knock-in (CAR), derivative of Paired related homeobox 1 (Prx1)+ lateral plate
mesoderm. CLP, common lymphoid progenitor; CMP, common myeloid progenitor; MPP, multipotent progenitor; BMPs, bone morphogenetic proteins; OPN, osteopontin;
HA, hyaluronic acid.

[16,17]. Later, several studies confirmed the importance of Cxcl12 seem to improve engraftment when co-transplanted with HSCs
expressing cells in HSPC regulation but suggested different roles [28,29]. Several works showed that ex vivo expanded BMSCs could
depending on their source and location. Cxcl12 expression by engraft in recipient marrow after intramedullary transplantation.
osteoblastic cells would be important in B-cell lymphoiesis and in These BMSCs could give rise to most stromal component and
HPC retention in the bone marrow; however, perivascular Cxcl12 enhance engraftment and HSC function in the recipient marrow
expression would be key to directly support HSCs [18,19]. [30]. Le Blanc et al. co-transplanted BMSC with HSC and achieved
The precise identity of the mesenchymal stromal populations faster engraftment of neutrophils and platelets with 100% donor
that contain BMSC and HSC niche functions remains debated. In chimerism [31].
2012 Ding et al. identified perivascular HSC-supporting stromal Multiple studies have shown that BMSCs contribute to HSC
cells using the expression of the HSC maintenance factor stem long-term maintenance, proliferation and differentiation through
cell factor (Scf; also known as kitl). This cell population over- different signalling pathways (Fig. 1). As indicated before,
lapped only with Nestin–GFPlo cells, and not with Nestin–GFPhi Cxcl12–Cxcr4 axis is a key player in the interaction between BMSCs
cells [20]. However, Kunisaki et al. proposed NG2+ Nestin–GFPhi and HSCs, but many other signaling pathways participate in this
cells as components of an arteriolar HSC–BMSC niche. Depletion of crosstalk. For instance, Rac GTPases in nestin+ perivascular BMSCs
NG2+ cells promoted HSC proliferation and reduced its long-term have been recently involved in HSC regulation [32,33]. Other stud-
repopulating capacity in vivo [21]. Finally, Zhou et al. suggested that ies suggest that long-term activation of Gs signaling in osteoblastic
leptin receptor positive cells contained all BMSC activity and rep- cells negatively affect HSC maintenance [34,35]. Also, Wnt signal-
resented the main source of adult bone formation [22], although ing regulates the survival, self-renewal and differentiation of HSPCs
leptin receptor+/Lepr-cre-traced cells appeared more abundant [36,37]. Agrin, a proteoglycan involved in neuromuscular junction,
than previously described and could contain different cell types also participate in the crosstalk between BMSCs and HSCs [38].
with distinct functions. In addition to soluble secreted factors or extracellular proteins,
In the human system, CD146+ perivascular BMSCs, reported cummulative evidence suggests that direct cell-to-cell contact is
initially by Sacchetti et al. as the in vivo correlate of human CFU- important in HSC regulation by BMSCs. As mentioned before, Notch
F-initiating cells [23], can support long-term persistence of human pathway is one example. Gottschling et al. reported that ␤1-
myelolymphoid HSPCs through cell to cell contact and via Notch integrin expression in human BMSCs could promote self-renewing
pathway [24]. Other studies also used the CD105 antigen to identify divisions of HSCs [39]. There are discrepancies whether BMSCs
murine mesenchymal stem cells. In this sense, non-adherent mes- and/or osteobalstic cells can modulate HSPCs through the adhe-
enchymal spheres derived from human CD146+ CD105+ nestin+ sion molecule N-cadherin [40,41]. Other molecules that have been
cells could promote human umbilical blood HSC expansion [25]. involved in BMSC–HSC interactions will not be discussed here due
Pinho et al. also showed HSC supporting activity using murine or to space constrictions.
adult fetal PDGFR␣ + CD51+ nestin+ BMSCs [26]. However, adult
human BMSCs are highly enriched in the PDGFR␣-CD271+ cell frac-
2. BMSCs, lymphopoiesis and myelopoiesis
tion [27].
There is now broad evidence suggesting that BMSCs have posi-
As part of the study of the bone marrow stem cell niches, many
tive effects in HSC transplantation. Both human and murine BMSCs
efforts have been directed towards understanding the interactions
A. García-García et al. / Immunology Letters 168 (2015) 129–135 131

originate a suitable microenvironment for HSPCs [51]. In addition,


mesenchymal stromal cells implanted subcutaneously together
with endothelial colony-forming cells can establish a functional
hematopoietic microenvironment. Human normal and leukemic
cells engrafted and remained functional in this extramedullary site
[52].

4. BMSCs in hematological malignancies

Research in the past ten years has definitely proved that


the microenvironment, and specially BMSCs, play more than a
bystander role in hematopoietic diseases. Alterations of stromal
cells have been implicated in the pathogenesis of myeloprolif-
erative neoplasms (MPNs), including chronic myeloid leukemias
(CMLs), and also in acute myeloid leukemias (AMLs). MPNs are
hematological malignancies arising from mutations in the HSCs,
and present with an overproduction and accumulation of myeloid
cells. Several mutations have been described in human patients,
Fig. 2. BMSCs and/or their derivatives directly regulate HSPCs and mature blood
including mutations in the Janus Kinase 2 gene (the most frequent
cells. Molecules involved in the crosstalk between mesenchymal stromal cells, lym-
phopoiesis and myelopoiesis. Mesenchymal stromal cells modulate T cell early one being JAK2V617F) [53–56], the thrombopoietin receptor MPL
lymphopoiesis by cell-to-cell contact through CD49d and CD90. They also stimu- [57], and calreticulin [58,59]. Furthermore, the list of additional sec-
late B cell lymphopiesis through VCAM-1, CXC12 and IL-7. However, Activin A and ondary mutations, mostly in epigenetic regulators, has increased as
G-CSF inhibit B cell formation, prompting myeloid differentiation. KITL, IL-6 and IL-3 of late. Mutations in ASXL1, CBL, IDH, IKZF1, and TET2 have been
directly contribute to myelopoiesis.
linked to an increased frequency of leukemic transformation in
patients (see Ref. [60] for a review). In the case of leukemias, cytoge-
between BMSCs and HSCs. However, cummulative evidence sug- netic abnormalities and chromosomal translocation originate the
gests that the BMSC derivatives could modulate hematopoiesis disease and promote the expansion of the leukemic stem cell (LSC)
by regulating committed hematopoietic progenitors. An in vitro clone (reviewed in Ref. [61]).
study suggested a role of mesenchymal stroma in early T-cell In 2007, Walkley et al. published two groundbreaking studies
lymphopoiesis through adhesion molecules [42]. Another study providing (i) direct verification that niche alterations were required
proposed a negative effect of mesenchymal stroma in B-cell for MPN-like disease inception and (ii) validation of the hypothesis
lymphopoiesis mediated by activin A [43]. However, Ichii demon- that cells from lineages other than the hematopoietic had leuke-
strated that human BMSCs in coculture with human CD34+ cord mogenic potential. Genetic deletion of the retinoblastoma gene
blood cells can support B lymphopoiesis [44]. Zhu et al. showed [62] or the retinoic acid receptor gamma (RAR␥) [63] provoked a
in vitro that osteoblasts are necessary and sufficient for B-cell com- non-transplantable MPN-like phenotype. More strikingly, in both
mitment and maturation [45]. Recently, it has been proposed that models the disease was recapitulated only when cells from disease-
granulocyte colony-stimulating factor (G-CSF) could modify BMSCs carrying mice were transplanted into recipients bearing the genetic
properties resulting in suppression of B lymphopoiesis in mice [46]. lesions, hence providing categorical confirmation of the necessity
As occurs with lymphopoiesis, some studies indicate that BMSCs of an altered niche for disease manifestation. Thereafter, several
and/or their derivatives could affect myelopoiesis. Previous evi- reports have surfaced to substantiate the idea that modifications
dence suggested the possibility that osteoblasts could stimulate in stromal cells are essential to obtain a transplantable pheno-
myelopoiesis by sustained G-CSF release [47]. Later, Angelopoulou type. Specific deletion of Notch pathway component Mind bomb
et al. showed that myelopoiesis and megakaryiopoiesis were 1 in stromal cells [64], deletion of miRNA processing gene Dicer1
improved when CD34+ HSCs and human BMSCs were cotrans- in BMSCs [65], and the presence of novel mutations in stromal
planted in immunodeficient mice [48]. However, it was described cells from human patients different from the HSC driver mutation
later that the myelopoietic supporting capacity of BMSCs is inde- in patients [66] altogether suggest a prominent role of BMSCs in
pendent of their multipotency and it is more related to their myeloid neoplasias.
ostoegenic/adipogenic differentiation and their glycosylation sta- Some questions have arisen in the field: are the lesions of the
tus [49]. microenvironment secondary events? To what extent do stromal
Interestingly, mature hematopoietic cells can also modulate cells switch from their normal physiology to a malignant one?
hematopoietic progenitors through BMSCs. For instance, CD8+ T- Could aberrant BMSCs actually drive and initiate disease? Many
cells can activate early multipotent progenitors during acute viral advances have been made using human samples from patients
infection to promote temporarily myelopoiesis [50]. suffering from MPNs as well as an ample variety of murine mod-
In summary, it seems that BMSCs and/or their derivatives els. Data from both of these sources portray consistent results:
(particularly osteoblasts) naturally support B-cell lymphopoiesis; mesenchymal stromal cells display aberrant secretory phenotypes
however, inhibiting this capacity could favor myelopoiesis (Fig. 2). [67–69], disrupted signaling pathways [70–72] or atypical gene
expression patterns [73]. For instance, in myelodysplastic syn-
3. BMSCs in extramedullary hematopoiesis drome (MDS), BMSCs express anomalous levels of CXCL12 [74],
leukemic inhibitory factor (LIF), VEGF, and N-Cadherin or DICER
Hematopoiesis outside the bone marrow is often associated [75], among others. The altered expression profile of these BMSCs
with pathological processes. Some studies indicate that BMSCs has been proposed to be crucial for the remodeling of the neopla-
could also favor hematopoiesis in these abnormal situations. sic niche by creating a malignant microenvironment which favors
Khaldovanidi et al. reported that interleukin 5 induce spleen col- the development and maintenance of mutated MPN cells. Convinc-
onization with mesenchymal progenitor cells, which in turn favor ing data have been presented using xenograft models in which
the establishment of hematopoiesis providing necessary signals to CD34+ hematopoietic cells plus mesenchymal stromal cells from
132 A. García-García et al. / Immunology Letters 168 (2015) 129–135

Fig. 3. Some regulatory mechanisms in place within the malignant bone marrow niche. (A) Osteoblastic lineage bias. (B) Disruption of signal transduction pathways in BMSCs.
(C) Defective sympathetic innervation on nestin+ cells. (D) Abnormal production or inhibition of healthy HSC maintenance factors. (E) Activation of niche remodeling led by
the bone-lining osteoblasts acting on the neoplasic clone. (F) Expansion of the mutated blood cell. (G) Reinforcement of the malignant microenvironment through paracrine
loops between the BMSCs and the mutated HSCs.

disease-carrying donors were transplanted into immunodeficient mutant HSCs and their healthy counterparts that are transformed
NSG mice. Strikingly, mice transplanted with mesenchymal stromal into “leukemic-like” pro-inflammatory cells [82,83]. Nevertheless,
cells and CD34+ cells displayed a significantly higher reconstitu- several key studies have situated stromal elements in the midst of
tion rate than mice transplanted only with CD34+ cells or together fundamental niche transformations. One example of the stromal
with mesenchymal stromal cells from healthy donors [73]. These alterations in myeloid diseases is the loss of bone mass occurring
cotransplation setups have aided to illustrate the remodeling prin- in the blast phase of CML or in AML. Although partially explained
ciple: mutant cells hijack the normal function of the stromal cells by an increase of osteaclastic bone resorption, the decrease in bony
and therefore a modified, disease-prone, and even drug-resistant matrix was the result of an increase in the levels of cytokine CCL3
niche [76–80] is formed. produced by leukemic cells [84], which effectively decreased the
Drawing parallels to the homeostasis situation, mutant HSCs number of both osteoprogenitors and the more mature osteoblasts
reside within their particular microenvironment that works to [85]. Similar endocrine signals can regulate both osteoprogenitor
maintain the neoplasm and to suit the regulatory pathways cells and HSCs. For instance, work from the Morrison lab and our
involved in the expansion and progression of disease. Multiple group has shown recently that the female sex hormones estro-
reports have shed light on niche remodeling mechanisms. One is gens, known to be bone anabolic agents, also differentially regulate
based on the transcriptional reprogramming of BMSCs to (i) con- HSC self-renewal, proliferation and survival [86,87]. Endogenous
fer an advantage to the mutant hematopoietic cell, or (ii) impair estrogens might partially explain the higher incidence and sever-
healthy hematopoiesis; or possibly both concomitantly. Neopla- ity of hematological malignancies in men, compared with women,
sic cells rely most heavily on signaling cascades prompted by the and selective estrogen receptor modulators might be useful in the
paracrine action of secreted factors or cytokines. In CML, mutant treatment of MPN [86]. Linked to the notion of a dysfunctional
cells have been reported to induce the upregulation and secretion mesenchyme compartment, alterations in gene expression in mes-
of placental growth factor (PIGF) in stromal cells, which in turn enchymal cells have been deemed to have oncogenic potential and
stimulates the proliferation of CML cells in a positive loop fashion to increase the likelihood of leukemic transformation in human
[80]. In an additive manner, G-CSF secreted by CML cells leads to a patients. Specific deletion of the Schwachman–Bodian–Diamond
decrease in BMSC-produced CXCL12, leading to complications with (sbds) gene in cells from mesenchymal origin prompted myelodys-
normal HSC maintenance and retention [81]. plastic symptoms in mice [65]. Furthermore, it has been proposed
In an attempt to establish unified patterns for the whole spectra that, in MPNs, BMSCs might develop a bias towards generation
of myeloid malignancies, one needs to underscore and highlight of malfunctioning osteoblastic progenitors, ultimately leading to
the pivotal role of BMSCs and their progeny as major sources of faulty hematopoiesis and creating fertile grounds for marrow fibro-
cytokine production. Paracrine loops are known to occur between sis and osteosclerosis [88]. Altogether, available data seem to agree
A. García-García et al. / Immunology Letters 168 (2015) 129–135 133

with the hypothesis that mutated HSPCs cause damage to BMSCs F., who is also supported in part by an International Early Career
that is partially responsible for disease manifestation and progres- Scientist grant of the Howard Hughes Medical Institute. We apolo-
sion (Fig. 3). gize for omission of relevant literature due to space contraints.
Recent reports have added the peripheral nervous system as
yet another layer of regulation in normal hematopoiesis [89–91]
but also in the development of MPNs, as neuropathy-triggered References
inflammatory damage to BMSCs seems to be required for MPN
manifestation [92]. In a murine model of JAK2V617F polycythemia [1] H.M. Patt, M.A. Maloney, Bone marrow regeneration after local injury: a
vera, we have shown that the overproduction of interleukin-1␤ review, Exp. Hematol. 3 (1975) 135–148.
[2] A.J. Friedenstein, R.K. Chailakhjan, K.S. Lalykina, The development of
by mutant HSPCs triggers the death of Schwann cells and causes fibroblast colonies in monolayer cultures of guinea-pig bone marrow and
neuropathy in sympathetic nerve terminals in the bone marrow, spleen cells, Cell Tissue Kinet. 3 (1970) 393–403.
sensitizing nestin+ BMSCs to undergo apoptosis. Acceleration of [3] T.M. Dexter, E.G. Wright, F. Krizsa, L.G. Lajtha, Regulation of haemopoietic
stem cell proliferation in long term bone marrow cultures,
the polycythemic phenotype can be directly attributed to the loss Biomedicine/[publiee pour l’A.A.I.C.I.G.] 27 (1977) 344–349.
of BMSCs in the marrow, since in vivo depletion of nestin+ BMSCs [4] R. Schofield, The relationship between the spleen colony-forming cell and
or their Cxcl12 production fueled disease progression, whereas the the haemopoietic stem cell, Blood Cells 4 (1978) 7–25.
[5] B.I. Lord, N.G. Testa, J.H. Hendry, The relative spatial distributions of CFUs
pharmacological rescue of the sympathetic innervation of BMSCs and CFUc in the normal mouse femur, Blood 46 (1975) 65–72.
through administration of sympathomimetic agents was able to [6] J.K. Gong, Endosteal marrow: a rich source of hematopoietic stem cells,
prevent nestin+ BMSC loss and thus block mutant HSC expan- Science 199 (1978) 1443–1445.
[7] S.K. Nilsson, H.M. Johnston, J.A. Coverdale, Spatial localization of
sion [92]. Similar findings were soon after reported in a MLL-AF9
transplanted hemopoietic stem cells: inferences for the localization of stem
acute myeloid leukemia (AML) murine model [93], although the cell niches, Blood 97 (2001) 2293–2299.
relevance of bone marrow neuropathy in this setting is unclear [8] L.M. Calvi, et al., Osteoblastic cells regulate the haematopoietic stem cell
niche, Nature 425 (2003) 841–846.
since, unlike in MPN [92], adrenergic signals did not seem to affect
[9] J. Zhang, et al., Identification of the haematopoietic stem cell niche and
significantly AML progression [93]. Due to its clinical relevance control of the niche size, Nature 425 (2003) 836–841.
and impact on public health, AML has been extensively studied. [10] F. Arai, et al., Tie2/angiopoietin-1 signaling regulates hematopoietic stem
Many relevant insights have been made on the crosstalk between cell quiescence in the bone marrow niche, Cell 118 (2004) 149–161.
[11] B.O. Zhou, L. Ding, S.J. Morrison, Hematopoietic stem and progenitor cells
leukemic stem cells (LSCs) and their niche, both in xenograft and regulate the regeneration of their niche by secreting Angiopoietin-1, eLife 4
murine models. Whereas the niche in acute leukemias can assist in (2015) e05521, http://dx.doi.org/10.7554/eLife.05521
LSC engraftment and homing [94,95] and promote chemotherapy- [12] H. Yoshihara, et al., Thrombopoietin/MPL signaling regulates hematopoietic
stem cell quiescence and interaction with the osteoblastic niche, Cell Stem
resistance [96], it remains to be determined whether it can also Cell 1 (2007) 685–697, http://dx.doi.org/10.1016/j.stem.2007.10.020
regulate other aspects of LSC biology in AML. Dissecting the involve- [13] A. Nakamura-Ishizu, K. Takubo, M. Fujioka, T. Suda, Megakaryocytes are
ment of stromal cells in leukemic maintenance and development essential for HSC quiescence through the production of thrombopoietin,
Biochem. Biophys. Res. Commun. 454 (2014) 353–357, http://dx.doi.org/10.
will be vital to design meaningful and powerful therapeutic strate- 1016/j.bbrc.2014.10.095
gies. [14] T. Nagasawa, et al., Defects of B-cell lymphopoiesis and bone-marrow
In summary, multiple studies using genetically-modified mouse myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1, Nature 382
(1996) 635–638.
strain converge in the notion that perivascular BMSCs cooper-
[15] S. Mendez-Ferrer, et al., Mesenchymal and haematopoietic stem cells form a
ate with endothelial cells in regulating HSCs. Similar conclusions unique bone marrow niche, Nature 466 (2010) 829–834, http://dx.doi.org/
have also been obtained in phylogenetically distant species, such 10.1038/nature09262
[16] T. Sugiyama, H. Kohara, M. Noda, T. Nagasawa, Maintenance of the
as zebrafish [97]. Parallels are emerging in the skeletal and the
hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in
hematopoietic systems, since the physiological turnover of both bone marrow stromal cell niches, Immunity 25 (2006) 977–988.
bone [98,99] and blood [100] seem to rely mostly in committed [17] Y. Omatsu, et al., The essential functions of adipo-osteogenic progenitors as
progenitors or short-term stem cells, but not in the most primitive the hematopoietic stem and progenitor cell niche, Immunity 33 (2010)
387–399, http://dx.doi.org/10.1016/j.immuni.2010.08.017,
stem cells. However, during stress situations MSCs and HSCs can be S1074-7613(10)00322-5 [pii].
activated and contribute to organ turnover, such as bone fracture [18] A. Greenbaum, et al., CXCL12 in early mesenchymal progenitors is required
or myeloablation, respectively. for haematopoietic stem-cell maintenance, Nature 495 (2013) 227–230,
http://dx.doi.org/10.1038/nature11926
Also, BMSCs have emerged as key contributors and partici- [19] L. Ding, S.J. Morrison, Haematopoietic stem cells and early lymphoid
pants in hematopoietic malignancies. Despite the ever-increasing progenitors occupy distinct bone marrow niches, Nature 495 (2013)
amount of sound reports, we are in need of a thorough characteri- 231–235, http://dx.doi.org/10.1038/nature11885
[20] L. Ding, T.L. Saunders, G. Enikolopov, S.J. Morrison, Endothelial and
zation of the chronological events that end in niche remodeling in perivascular cells maintain haematopoietic stem cells, Nature 481 (2012)
order to obtain a clear picture of the disease-initiating or disease- 457–462, http://dx.doi.org/10.1038/nature10783
propagating mechanisms. With the advent of improved models and [21] Y. Kunisaki, et al., Arteriolar niches maintain haematopoietic stem cell
quiescence, Nature 502 (2013) 637–643, http://dx.doi.org/10.1038/
technological advances to study individual cells, the field of BMSCs
nature12612
and hematopoiesis will surely witness its landscape modified in [22] B.O. Zhou, R. Yue, M.M. Murphy, G. Peyer, S. Morrison,
coming years. Leptin-receptor-expressing mesenchymal stromal cells represent the main
source of bone formed by adult bone marrow, Cell Stem Cell (2014), http://
dx.doi.org/10.1016/j.stem.2014.06.008
[23] B. Sacchetti, et al., Self-renewing osteoprogenitors in bone marrow sinusoids
Acknowledgements can organize a hematopoietic microenvironment, Cell 131 (2007) 324–336.
[24] M. Corselli, et al., Perivascular support of human hematopoietic
stem/progenitor cells, Blood 121 (2013) 2891–2901, http://dx.doi.org/10.
A.G.-G. is the recipient of La Caixa-Severo Ochoa PhD fellow- 1182/blood-2012-08-451864
ship. C.L.F. de C. has been awarded a PhD fellowship by Boehringer [25] J. Isern, et al., Self-renewing human bone marrow mesenspheres promote
Ingelheim Fonds. This work was supported by the Fundación Cen- hematopoietic stem cell expansion, Cell Rep. 3 (2013) 1714–1724, http://dx.
doi.org/10.1016/j.celrep.2013.03.041
tro Nacional de Investigaciones Cardiovasculares Carlos III, the [26] S. Pinho, et al., PDGFRalpha and CD51 mark human Nestin+ sphere-forming
Spanish Ministry of Economy and Competitiveness (TerCel, Span- mesenchymal stem cells capable of hematopoietic progenitor cell
ish Cell Therapy Network; Plan Nacional grant SAF-2011-30308; expansion, J. Exp. Med. 210 (2013) 1351–1367, http://dx.doi.org/10.1084/
jem.20122252
Ramón y Cajal Program grant RYC-2009-04703), Marie Curie Career [27] H. Li, et al., Low/negative expression of PDGFR-alpha identifies the candidate
Integration Programme grant (FP7-PEOPLE-2011-RG-294096) and primary mesenchymal stromal cells in adult human bone marrow, Stem Cell
ConSEPOC-Comunidad de Madrid S2010/BMD-2542 grant to S.M.- Rep. 3 (2014) 965–974, http://dx.doi.org/10.1016/j.stemcr.2014.09.018
134 A. García-García et al. / Immunology Letters 168 (2015) 129–135

[28] O.N. Koc, et al., Rapid hematopoietic recovery after coinfusion of [53] E.J. Baxter, et al., Acquired mutation of the tyrosine kinase JAK2 in human
autologous-blood stem cells and culture-expanded marrow mesenchymal myeloproliferative disorders, Lancet 365 (2005) 1054–1061, http://dx.doi.
stem cells in advanced breast cancer patients receiving high-dose org/10.1016/S0140-6736(05)71142-9
chemotherapy, J. Clin. Oncol. 18 (2000) 307–316. [54] C. James, et al., A unique clonal JAK2 mutation leading to constitutive
[29] M. Bensidhoum, et al., Homing of in vitro expanded Stro-1- or Stro-1+ signalling causes polycythaemia vera, Nature 434 (2005) 1144–1148, http://
human mesenchymal stem cells into the NOD/SCID mouse and their role in dx.doi.org/10.1038/nature03546
supporting human CD34 cell engraftment, Blood 103 (2004) 3313–3319, [55] R.L. Levine, et al., Activating mutation in the tyrosine kinase JAK2 in
http://dx.doi.org/10.1182/blood-2003-04-1121 polycythemia vera, essential thrombocythemia, and myeloid metaplasia
[30] Y. Muguruma, et al., Reconstitution of the functional human hematopoietic with myelofibrosis, Cancer Cell 7 (2005) 387–397, http://dx.doi.org/10.
microenvironment derived from human mesenchymal stem cells in the 1016/j.ccr.2005.03.023
murine bone marrow compartment, Blood 107 (2006) 1878–1887, http:// [56] R. Kralovics, et al., A gain-of-function mutation of JAK2 in myeloproliferative
dx.doi.org/10.1182/blood-2005-06-2211 disorders, N. Engl. J. Med. 352 (2005) 1779–1790, http://dx.doi.org/10.1056/
[31] K. Le Blanc, et al., Transplantation of mesenchymal stem cells to enhance NEJMoa051113
engraftment of hematopoietic stem cells, Leukemia 21 (2007) 1733–1738, [57] A.D. Pardanani, et al., MPL515 mutations in myeloproliferative and other
http://dx.doi.org/10.1038/sj.leu.2404777 myeloid disorders: a study of 1182 patients, Blood 108 (2006) 3472–3476,
[32] A. Sanchez-Aguilera, et al., Guanine nucleotide exchange factor Vav1 http://dx.doi.org/10.1182/blood-2006-04-018879
regulates perivascular homing and bone marrow retention of hematopoietic [58] T. Klampfl, et al., Somatic mutations of calreticulin in myeloproliferative
stem and progenitor cells, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) neoplasms, N. Engl. J. Med. 369 (2013) 2379–2390, http://dx.doi.org/10.
9607–9612, http://dx.doi.org/10.1073/pnas.1102018108, 1102018108 [pii]. 1056/NEJMoa1311347
[33] M.F. Ciuculescu, et al., Perivascular deletion of murine Rac reverses the ratio [59] J. Nangalia, et al., Somatic CALR mutations in myeloproliferative neoplasms
of marrow arterioles and sinusoid vessels and alters hematopoiesis in vivo, with nonmutated JAK2, N. Engl. J. Med. 369 (2013) 2391–2405, http://dx.
Blood 125 (2015) 3105–3113, http://dx.doi.org/10.1182/blood-2014-10- doi.org/10.1056/NEJMoa1312542
604892 [60] A. Tefferi, W. Vainchenker, Myeloproliferative neoplasms: molecular
[34] G.B. Adams, et al., Haematopoietic stem cells depend on Galpha(s)-mediated pathophysiology, essential clinical understanding, and treatment strategies,
signalling to engraft bone marrow, Nature (2009). J. Clin. Oncol. 29 (2011) 573–582, http://dx.doi.org/10.1200/JCO.2010.29.
[35] K. Schepers, E.C. Hsiao, T. Garg, M.J. Scott, E. Passegue, Activated Gs signaling 8711
in osteoblastic cells alters the hematopoietic stem cell niche in mice, Blood [61] A.V. Krivtsov, S.A. Armstrong, MLL translocations, histone modifications and
120 (2012) 3425–3435, http://dx.doi.org/10.1182/blood-2011-11-395418 leukaemia stem-cell development, Nat. Rev. Cancer 7 (2007) 823–833,
[36] T. Reya, et al., A role for Wnt signalling in self-renewal of haematopoietic http://dx.doi.org/10.1038/nrc2253
stem cells, Nature 423 (2003) 409–414. [62] C.R. Walkley, J.M. Shea, N.A. Sims, L.E. Purton, S.H. Orkin, Rb regulates
[37] M.C. Florian, et al., A canonical to non-canonical Wnt signalling switch in interactions between hematopoietic stem cells and their bone marrow
haematopoietic stem-cell ageing, Nature 503 (2013) 392–396, http://dx.doi. microenvironment, Cell 129 (2007) 1081–1095, http://dx.doi.org/10.1016/j.
org/10.1038/nature12631 cell.2007.03.055
[38] C. Mazzon, et al., The critical role of agrin in the hematopoietic stem cell [63] C.R. Walkley, et al., A microenvironment-induced myeloproliferative
niche, Blood 118 (2011) 2733–2742, http://dx.doi.org/10.1182/blood-2011- syndrome caused by retinoic acid receptor gamma deficiency, Cell 129
01-331272 (2007) 1097–1110, http://dx.doi.org/10.1016/j.cell.2007.05.014
[39] S. Gottschling, et al., Human mesenchymal stromal cells regulate initial [64] Y.W. Kim, et al., Defective Notch activation in microenvironment leads to
self-renewing divisions of hematopoietic progenitor cells by a myeloproliferative disease, Blood 112 (2008) 4628–4638, http://dx.doi.org/
beta1-integrin-dependent mechanism, Stem Cells 25 (2007) 798–806, 10.1182/blood-2008-03-148999
http://dx.doi.org/10.1634/stemcells 2006-0513 [65] M.H. Raaijmakers, et al., Bone progenitor dysfunction induces
[40] Y. Xie, et al., Detection of functional haematopoietic stem cell niche using myelodysplasia and secondary leukaemia, Nature 464 (2010) 852–857,
real-time imaging, Nature 457 (2009) 97–101, http://dx.doi.org/10.1038/ http://dx.doi.org/10.1038/nature08851, nature08851[pii].
nature07639, nature07639[pii]. [66] O. Blau, et al., Mesenchymal stromal cells of myelodysplastic syndrome and
[41] M.J. Kiel, M. Acar, G.L. Radice, S.J. Morrison, Hematopoietic stem cells do not acute myeloid leukemia patients have distinct genetic abnormalities
depend on N-cadherin to regulate their maintenance, Cell Stem Cell 4 compared with leukemic blasts, Blood 118 (2011) 5583–5592, http://dx.doi.
(2009) 170–179, http://dx.doi.org/10.1016/j.stem.2008.10.005, org/10.1182/blood-2011-03-343467
S1934-5909(08)528-6[pii]. [67] R.K. Schneider, et al., Activated fibronectin-secretory phenotype of
[42] M. Barda-Saad, et al., Adhesion molecules involved in the interactions mesenchymal stromal cells in pre-fibrotic myeloproliferative neoplasms, J.
between early T cells and mesenchymal bone marrow stromal cells, Exp. Hematol. Oncol. 7 (2014) 92, http://dx.doi.org/10.1186/s13045-014-0092-2
Hematol. 27 (1999) 834–844. [68] L. Varricchio, A. Mancini, A.R. Migliaccio, Pathological interactions between
[43] T. Shoham, R. Parameswaran, Y. Shav-Tal, M. Barda-Saad, D. Zipori, The hematopoietic stem cells and their niche revealed by mouse models of
mesenchymal stroma negatively regulates B cell lymphopoiesis through the primary myelofibrosis, Expert Rev. Hematol. 2 (2009) 315–334, http://dx.
expression of activin A, Ann. N. Y. Acad. Sci. 996 (2003) 245–260. doi.org/10.1586/ehm.09.17
[44] M. Ichii, et al., Regulation of human B lymphopoiesis by the transforming [69] R.A. Ferrer, et al., Mesenchymal stromal cells from patients with
growth factor-beta superfamily in a newly established coculture system myelodyplastic syndrome display distinct functional alterations that are
using human mesenchymal stem cells as a supportive microenvironment, modulated by lenalidomide, Haematologica 98 (2013) 1677–1685, http://
Exp. Hematol. 36 (2008) 587–597, http://dx.doi.org/10.1016/j.exphem.2007. dx.doi.org/10.3324/haematol.2013.083972
12.013 [70] G. Varga, et al., Inappropriate Notch activity and limited mesenchymal stem
[45] J. Zhu, et al., Osteoblasts support B-lymphocyte commitment and cell plasticity in the bone marrow of patients with myelodysplastic
differentiation from hematopoietic stem cells, Blood 109 (2007) 3706–3712. syndromes, Pathol. Oncol. Res. 13 (2007) 311–319, PAOR.2007.13.4.0311.
[46] R.B. Day, D. Bhattacharya, T. Nagasawa, D.C. Link, Granulocyte [71] J.C. Huang, et al., Mesenchymal stromal cells derived from acute myeloid
colony-stimulating factor reprograms bone marrow stromal cells to actively leukemia bone marrow exhibit aberrant cytogenetics and cytokine
suppress B lymphopoiesis in mice, Blood 125 (2015) 3114–3117, http://dx. elaboration, Blood Cancer J. 5 (2015) e302, http://dx.doi.org/10.1038/bcj.
doi.org/10.1182/blood-2015-02-629444 2015.17
[47] R.S. Taichman, S.G. Emerson, Human osteoblasts support hematopoiesis [72] A. Kode, et al., Leukaemogenesis induced by an activating beta-catenin
through the production of granulocyte colony-stimulating factor, J. Exp. mutation in osteoblasts, Nature 506 (2014) 240–244, http://dx.doi.org/10.
Med. 179 (1994) 1677–1682. 1038/nature12883
[48] M. Angelopoulou, et al., Cotransplantation of human mesenchymal stem [73] H. Medyouf, et al., Myelodysplastic cells in patients reprogram mesenchymal
cells enhances human myelopoiesis and megakaryocytopoiesis in NOD/SCID stromal cells to establish a transplantable stem cell niche disease unit, Cell
mice, Exp. Hematol. 31 (2003) 413–420. Stem Cell 14 (2014) 824–837, http://dx.doi.org/10.1016/j.stem.2014.02.014
[49] V. Morad, et al., The myelopoietic supportive capacity of mesenchymal [74] E. Flores-Figueroa, S. Varma, K. Montgomery, P.L. Greenberg, D. Gratzinger,
stromal cells is uncoupled from multipotency and is influenced by lineage Distinctive contact between CD34+ hematopoietic progenitors and CXCL12+
determination and interference with glycosylation, Stem Cells 26 (2008) CD271+ mesenchymal stromal cells in benign and myelodysplastic bone
2275–2286, http://dx.doi.org/10.1634/stemcells.2007-0518 marrow, Lab. Invest. 92 (2012) 1330–1341, http://dx.doi.org/10.1038/
[50] C.M. Schurch, C. Riether, A.F. Ochsenbein, Cytotoxic CD8+ T cells stimulate labinvest.2012.93
hematopoietic progenitors by promoting cytokine release from bone [75] C. Santamaria, et al., Impaired expression of DICER, DROSHA, SBDS and some
marrow mesenchymal stromal cells, Cell Stem Cell 14 (2014) 460–472, microRNAs in mesenchymal stromal cells from myelodysplastic syndrome
http://dx.doi.org/10.1016/j.stem.2014.01.002 patients, Haematologica 97 (2012) 1218–1224, http://dx.doi.org/10.3324/
[51] S. Khaldoyanidi, L. Sikora, D.H. Broide, M.E. Rothenberg, P. Sriramarao, haematol.2011.054437
Constitutive overexpression of IL-5 induces extramedullary hematopoiesis [76] T. Manshouri, et al., Bone marrow stroma-secreted cytokines protect
in the spleen, Blood 101 (2003) 863–868, http://dx.doi.org/10.1182/blood- JAK2(V617F)-mutated cells from the effects of a JAK2 inhibitor, Cancer Res.
2002-03-0735 71 (2011) 3831–3840, http://dx.doi.org/10.1158/0008-5472CAN-10-4002
[52] Y. Chen, et al., Human extramedullary bone marrow in mice: a novel in vivo [77] F. Vianello, et al., Bone marrow mesenchymal stromal cells non-selectively
model of genetically controlled hematopoietic microenvironment, Blood protect chronic myeloid leukemia cells from imatinib-induced apoptosis via
119 (2012) 4971–4980, http://dx.doi.org/10.1182/blood-2011-11-389957 the CXCR4/CXCL12 axis, Haematologica 95 (2010) 1081–1089, http://dx.doi.
org/10.3324/haematol.2009.017178
A. García-García et al. / Immunology Letters 168 (2015) 129–135 135

[78] L. Jin, et al., CXCR4 up-regulation by imatinib induces chronic myelogenous [89] Y. Katayama, et al., Signals from the sympathetic nervous system regulate
leukemia (CML) cell migration to bone marrow stroma and promotes hematopoietic stem cell egress from bone marrow, Cell 124 (2006) 407–421.
survival of quiescent CML cells, Mol. Cancer Ther. 7 (2008) 48–58, http://dx. [90] A. Spiegel, et al., Catecholaminergic neurotransmitters regulate migration
doi.org/10.1158/1535-7163 MCT-07-0042 and repopulation of immature human CD34+ cells through Wnt signaling,
[79] B. Zhang, et al., Microenvironmental protection of CML stem and progenitor Nat. Immunol. 8 (2007) 1123–1131, http://dx.doi.org/10.1038/ni1509,
cells from tyrosine kinase inhibitors through N-cadherin and ni1509[pii].
Wnt-beta-catenin signaling, Blood 121 (2013) 1824–1838, http://dx.doi.org/ [91] S. Mendez-Ferrer, D. Lucas, M. Battista, P.S. Frenette, Haematopoietic stem
10.1182/blood-2012-02-412890 cell release is regulated by circadian oscillations, Nature 452 (2008)
[80] T. Schmidt, et al., Loss or inhibition of stromal-derived PlGF prolongs 442–447, http://dx.doi.org/10.1038/nature06685
survival of mice with imatinib-resistant Bcr-Abl1(+) leukemia, Cancer Cell [92] L. Arranz, et al., Neuropathy of haematopoietic stem cell niche is essential
19 (2011) 740–753, http://dx.doi.org/10.1016/j.ccr.2011.05.007 for myeloproliferative neoplasms, Nature 512 (2014) 78–81, http://dx.doi.
[81] B. Zhang, et al., Altered microenvironmental regulation of leukemic and org/10.1038/nature13383
normal stem cells in chronic myelogenous leukemia, Cancer Cell 21 (2012) [93] M. Hanoun, et al., Acute myelogenous leukemia-induced sympathetic
577–592, http://dx.doi.org/10.1016/j.ccr.2012.02.018 neuropathy promotes malignancy in an altered hematopoietic stem cell
[82] M. Kleppe, et al., JAK-STAT pathway activation in malignant and niche, Cell Stem Cell (2014), http://dx.doi.org/10.1016/j.stem.2014.06.020
nonmalignant cells contributes to MPN pathogenesis and therapeutic [94] S.W. Lane, et al., Differential niche and Wnt requirements during acute
response, Cancer Discov. 5 (2015) 316–331, http://dx.doi.org/10.1158/2159- myeloid leukemia progression, Blood 118 (2011) 2849–2856, http://dx.doi.
8290 CD-14-0736 org/10.1182/blood-2011-03-345165
[83] R.S. Welner, et al., Treatment of chronic myelogenous leukemia by blocking [95] F. Ishikawa, et al., Chemotherapy-resistant human AML stem cells home to
cytokine alterations found in normal stem and progenitor cells, Cancer Cell and engraft within the bone-marrow endosteal region, Nat. Biotechnol. 25
27 (2015) 671–681, http://dx.doi.org/10.1016/j.ccell.2015.04.004 (2007) 1315–1321, http://dx.doi.org/10.1038/nbt1350
[84] T. Baba, et al., MIP-1alpha/CCL3-mediated maintenance of [96] S. Iwamoto, K. Mihara, J.R. Downing, C.H. Pui, D. Campana, Mesenchymal
leukemia-initiating cells in the initiation process of chronic myeloid cells regulate the response of acute lymphoblastic leukemia cells to
leukemia, J. Exp. Med. 210 (2013) 2661–2673, http://dx.doi.org/10.1084/ asparaginase, J. Clin. Invest. 117 (2007) 1049–1057, http://dx.doi.org/10.
jem.20130112 1172/JCI30235
[85] B.J. Frisch, et al., Functional inhibition of osteoblastic cells in an in vivo [97] O.J. Tamplin, et al., Hematopoietic stem cell arrival triggers dynamic
mouse model of myeloid leukemia, Blood 119 (2012) 540–550, http://dx. remodeling of the perivascular niche, Cell 160 (2015) 241–252, http://dx.
doi.org/10.1182/blood-2011-04-348151 doi.org/10.1016/j.cell.2014.12.032
[86] A. Sanchez-Aguilera, et al., Estrogen signaling selectively induces apoptosis [98] D. Park, et al., Endogenous bone marrow MSCs are dynamic, fate-restricted
of hematopoietic progenitors and myeloid neoplasms without harming participants in bone maintenance and regeneration, Cell Stem Cell 10 (2012)
steady-state hematopoiesis, Cell Stem Cell 15 (2014) 791–804, http://dx.doi. 259–272, http://dx.doi.org/10.1016/j.stem.2012.02.003
org/10.1016/j.stem.2014.11.002 [99] C.K. Chan, et al., Identification and specification of the mouse skeletal stem
[87] D. Nakada, et al., Oestrogen increases haematopoietic stem-cell self-renewal cell, Cell 160 (2015) 285–298, http://dx.doi.org/10.1016/j.cell.2014.12.002
in females and during pregnancy, Nature 505 (2014) 555–558, http://dx.doi. [100] K. Busch, et al., Fundamental properties of unperturbed haematopoiesis
org/10.1038/nature12932 from stem cells in vivo, Nature 518 (2015) 542–546, http://dx.doi.org/10.
[88] K. Schepers, et al., Myeloproliferative neoplasia remodels the endosteal 1038/nature14242.
bone marrow niche into a self-reinforcing leukemic niche, Cell Stem Cell 13
(2013) 285–299, http://dx.doi.org/10.1016/j.stem.2013.06.009

You might also like