Similar to their healthy counterpart, malignant hematopoietic stem cells in myeloid malignancies, such as myeloproliferative neoplasms, myelodysplastic syndromes, and acute myeloid leukemia, reside in a highly complex and dynamic cellular microenvironment in the bone marrow. This environment provides key regulatory signals for and tightly controls cardinal features of hematopoietic stem cells (HSCs), including self-renewal, quiescence, differentiation, and migration. These features are essential to maintaining cellular homeostasis and blood regeneration throughout life. A large number of studies have extensively addressed the composition of the bone marrow niche in mouse models, as well as the cellular and molecular communication modalities at play under both normal and pathogenic situations. Although instrumental to interrogating the complex composition of the HSC niche and dissecting the niche remodeling processes that appear to actively contribute to leukemogenesis, these models may not fully recapitulate the human system due to immunophenotypic, architectural, and functional inter-species variability. This review summarizes several aspects related to the human hematopoietic niche: (1) its anatomical structure, composition, and function in normal hematopoiesis; (2) its alteration and functional relevance in the context of chronic and acute myeloid malignancies; (3) age-related niche changes and their suspected impact on hematopoiesis; (4) ongoing efforts to develop new models to study niche-leukemic cell interaction in human myeloid malignancies; and finally, (5) how the knowledge gained into leukemic stem cell (LSC) niche dependencies might be exploited to devise novel therapeutic strategies that aim at disrupting essential niche-LSC interactions or improve the regenerative ability of the disease-associated hematopoietic niche.

Hematopoietic stem cells (HSCs) are primitive multipotent tissue stem cells that reside in the bone marrow (BM) and ensure life-long regeneration of the blood system. This vital process is controlled by both cell-intrinsic features of HSCs and extrinsic signals, emanating from the BM microenvironment.

Perturbations of the BM niche inevitably affect normal hematopoiesis and are thought to contribute to the emergence of hematopoietic malignancies in both mice and humans. Therefore, the cellular components of the BM niche and the means by which they modulate normal and malignant hematopoiesis have been extensively interrogated in the past decade. Taking advantage of new technologies, such as intravital imaging and genetic mouse models, that allow for marking or deleting specific cell types, the field has recognized that the BM niche is a complex structure that contains a number of different cell types: multipotent mesenchymal stem cells (MSCs) and their progeny, a complex vascular network, nerve fibers, mature blood cells, and others. These cell types have all been shown to modulate the function of HSCs in mice, and be perturbed in the context of malignancies. A new picture has emerged in which normal and malignant cells engage in bidirectional cross talk with their surrounding niche cells and establish specific interdependencies. These cellular interactions are thought to contribute to the emergence of hematopoietic malignancies by modulating important failsafe mechanisms related to self-renewal, proliferation, survival, and immune evasion. These mouse studies have recently been reviewed elsewhere1,2  and will not be discussed here.

This review puts emphasis on our current knowledge of the human HSC niche and its active involvement in the pathogenesis of human myeloid malignancies, namely myeloproliferative neoplasms (MPN, including chronic myeloid leukemia [CML]), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML). Moreover, this review provides an overview of emerging strategies being used to improve current models to study human myeloid malignancies, including integrating the human niche component. Importantly, we discuss how this new area of research is paving the way to the development of novel therapeutic strategies.

Compared with the mouse system, much less detailed knowledge exists regarding the architecture of the human BM and the function of its different cellular constituents. Using a comprehensive set of descriptive (flow cytometry, immunohistochemistry of BM trephine) and functional analysis in xenografts, Guezguez and colleagues3  demonstrated that human HSCs (linCD34+CD38CD45RACD49f+)4  preferentially locate to trabecular areas of the BM.3  In both humans and mice, these areas contain various types of stromal cells, endothelial cells (ECs), and nerve fibers; the latter are arranged mainly around medium-sized arteries and capillaries,5,6  also pointing to a conserved BM innervation by the sympathetic nervous system (Figure 1).7,8 

Figure 1.

Schematic view of the anatomical structure of the human hematopoietic niche. This schema provides a simplified view of the cellular architecture of the human BM niche. It reflects the idea that hHSCs are mainly found in close proximity to the vasculature in direct contact with CD271+ MSCs.3,9  Similar to the mouse BM niche, arterioles and capillaries near the endosteum are ensheated by sympathetic nerve fibers.10  Surface markers distinguishing different cellular subsets are also indicated. CD146+ MSCs are close to the vasculature.11  Nestin and endoglin (CD105) have been proposed to mark arteriolar and sinusoidal ECs, respectively.5,20 

Figure 1.

Schematic view of the anatomical structure of the human hematopoietic niche. This schema provides a simplified view of the cellular architecture of the human BM niche. It reflects the idea that hHSCs are mainly found in close proximity to the vasculature in direct contact with CD271+ MSCs.3,9  Similar to the mouse BM niche, arterioles and capillaries near the endosteum are ensheated by sympathetic nerve fibers.10  Surface markers distinguishing different cellular subsets are also indicated. CD146+ MSCs are close to the vasculature.11  Nestin and endoglin (CD105) have been proposed to mark arteriolar and sinusoidal ECs, respectively.5,20 

Close modal

Sympathetic nerve fibers

In mice, HSCs’ egress from their quiescence-enforcing niches follows a circadian rhythm imposed by the cyclic expression of Cxcl12 on MSCs and its receptor, Cxcr4, on HSCs, which are under the control of β3-adrenergic signals and the clock gene (Bmal1), respectively.8,12  Although inverted, timing-wise, human HSCs’ (hHSCs) egress also fluctuates in a circadian manner.12  Notably, β2-adrenergic receptor (β2-AR) signaling directly modulates the motility and proliferation of immature CD34+ human hematopoietic stem/progenitor cells (hHSPCs), and the expression of ARs is induced by granulocyte colony-stimulating factor (G-CSF), a potent HSPC mobilizing agent.13  These data point to an essential role of the sympathetic nervous system in the human HSC niche. Whether the molecular mechanisms at play in humans and mice are similar remains to be tested.

Endothelial cells

Comparable to mice,14-18  hHSCs mainly reside within a few cell diameters of the BM vasculature in direct contact with CD271+ MSCs.3,5,12  In mice, the vascular network is composed of fenestrated sinusoidal vessels and endosteal arterioles, which are proposed to differentially harbor active versus dormant HSCs, a topic that is still a matter of debate.16-19  Nevertheless, a similar architecture exists in human BM, where endoglin (CD105) and nestin expression has been proposed to preferentially identify sinusoidal vessels and arterioles/capillaries, respectively.5,20  Although enriched in the arterioles containing trabecular areas of the human BM, CD34+ hHSPCs have been reported to also localize close to sinusoidal vascular cells in contact with CD271+ MSCs.3  Functionally, human BM-derived ECs transformed with an AKT-activating adenoviral gene21  support the in vitro expansion of linCD45+CD34+CD45RACD49f+ hHSCs capable of multilineage reconstitution in immune-deficient mice,22  thereby arguing in favor of a direct effect on hHSC maintenance.

Multipotent MSCs

Human MSCs (hMSCs) are functionally defined by their clonogenic potential (colony-forming unit-fibroblasts) and ability to assemble a functional BM niche in vivo. These cells are proposed to reside within the linCD45CD271+CD140alow/– fraction.23-25  CD271+ hMSCs can further be subdivided by CD146 expression, which marks MSCs that locate close to the vasculature14  and are reported to express high levels of well-characterized HSC-supportive genes (IGF2, WNT3A, JAG1, CXCL12, KITLG, ANGPTL1).26,27  Similar to mice,28  these cells also express LEPR,25  CD105, and CD90.29,30  However, mouse and fetal MSCs are rather enriched in the PDGFRa+ (CD140a+) fraction,25,31  which appears to be a developmentally regulated marker in humans. Nestin expression, however, is restricted to a subset of hMSCs, as suggested by analysis of liquid biopsies32,33  versus in situ core biopsies (BM trephine), where high nestin expression is rather proposed to mark ECs.5  Functionally, however, both fetal and adult BM nestin+ MSCs were shown to enhance the long-term multilineage reconstitution activity of HSPCs.25,29,31  Recently, more restricted osteolineage progenitors were shown to promote, ex vivo, the expansion of hHSCs capable of hematopoietic reconstitution through the secretion of microRNA-containing extracellular vesicles.34 

Although further studies are required, these data provide compelling evidence that the anatomical structure of the human and mouse BM niche are highly conserved, which hints toward potentially conserved non–cell autonomous mechanisms of HSC maintenance.

Genetic events are well established as main intrinsic drivers of leukemogenesis. These transformation events are thought to primarily occur in self-renewing HSCs that gain a competitive advantage over normal HSCs and give rise to the disease-propagating leukemic cells, also referred to as leukemic stem cells (LSCs).35-39  Nevertheless, evidence also exists to support transformation of early progenitors, such as lymphoid-primed multipotent progenitors and granulocyte macrophage progenitors in AML,40,41  a granulocyte macrophage progenitor-like subset in blast crisis CML,42  and multipotent progenitors in sporadic MDS cases.43,44  Whether the cell of origin defines the specific interdependencies with niche cells is largely unknown. More broadly, the issue of spatial distribution of LSCs in the human BM and their niche dependency remains highly underexplored. Studies using in situ localization and functional repopulation assays showed that, in xenografts, AML LSC-enriched populations exhibit a spatial and functional distribution similar to that of normal HSCs.45,46  This localization to physiological HSC niches appears to be different in T-cell acute lymphoblastic leukemia.47  Nevertheless, these findings add to the emerging importance of the BM niche, which can either instigate hematopoietic changes and myeloproliferation or, more frequently, act as a driver of disease maintenance/progression.

Further aspects regarding the genetic and functional definition of LSCs in myeloid malignancies as well as emerging markers that distinguish normal HSCs from malignant LSCs, are extensively discussed in the accompanying reviews on leukemic stem cells in MDS,48  MPN49 , and AML.50 

The concept of niche-induced disease initiation in vivo has been demonstrated using animal models of MNP,51-55  MDS,56,57  and AML,58,59  a topic that was extensively reviewed elsewhere.1,60  Evidence to support this in humans is mainly based on the occurrence of rare but notable cases of donor-derived leukemia, observed in patients receiving BM transplant, where preexisting niche changes in the host are thought to initiate leukemogenesis.61  This idea of microenvironment-driven disease postulates that in the context of a healthy donor microenvironment, a genetically abnormal “primed” HSC clone would be kept in check, while possibly gaining competitive advantage and undergoing further clonal evolution in the disease-primed microenvironment of the recipient.

Along this line, analysis of ex vivo expanded mesenchymal stromal niche cells from patients with MDS and AML are reported to exhibit a wide range of functional and molecular alterations, including chromosomal aberrations,62-66  transcriptional,67  and epigenetic changes68  as well as functional alterations in their differentiation potential and HSC-supportive activities69,70  (reviewed in Pleyer et al71 ). Genetic and transcriptomic alterations were also reported in unmanipulated CD271+ MSCs directly isolated from MDS patients by array comparative hybridization and transcriptomic profiling.26,72,73  Most notably, some of the events shown to initiate malignant transformation in mice are also reported in patient-derived mesenchymal cells, including downregulation of DICER and SBDS57,70,73  and activation of β-catenin in osteoblastic cells.58  More recently, Dong et al55  reported that the expression of a PTPN11 mutant found in juvenile myelomonocytic leukemia (JMML) cases (an aggressive form of childhood MPN) associated with Noonan syndrome recapitulates JMML in nestin+ cells in mice, strongly arguing for its likely involvement as an initiating event in the pathogenesis of congenital JMML.

In addition to these studies addressing the contribution of mesenchymal niche cells, emerging evidence also points to a potential role for ECs in disease initiation. JAK2V617F+ ECs have been reported in a subpopulation of MPN patients74  and recently demonstrated to actively contribute to the emergence of MPN in mice by favoring the expansion of Jak2V617F HSC over the Jak2wt counterpart.75  These effects appear to be mediated through upregulation of Cxcl12 and Scf by the vascular niche cells. Likewise, another study presented at the 58th annual meeting of the American Society of Hematology reported that EC-specific KRasG12D expression causes an MPD phenotype and had a detrimental effect on HSC function.76 

In mice, evidence indicates that malignant cells actively shape their microenvironment to reinforce disease progression at the expense of normal hematopoiesis (reviewed in Hoggat et al1  and Schepers et al60 ). Remodeling of the mesenchymal, endothelial, and nerve components appear to play a central role in this process. Evidence pointing to the existence of conserved mechanisms in humans is progressively becoming available. However, experimental data remain scarce.

Deregulated CXCL12/CXCR4 signaling

Deregulated expression of CXCL12 by niche cells appears to be a unifying feature in myeloid malignancies. CXCL12 binding to its receptor, CXCR4, enhances the affinity of integrins that mediate HSC adhesion to their niches.77-79  Integrin-mediated cellular adhesion and lodging of normal human HSCs into supportive niches was recently demonstrated and visualized using human HSCs xenograft models and intravital imaging.80  The critical role of this axis in normal HSC function is further underscored by the use of CXCR4 receptor antagonist, such as AMD3100, a potent HSC mobilizing compound.81,82  Therefore, altered expression/distribution of CXCL12 or integrins by disease-associated stromal cells is likely to impinge on the ability of normal HSCs to find and reside in their quiescence maintaining niches, thereby possibly promoting their exhaustion and the progressive clonal dominance of the malignant HSCs observed in myeloid malignancies. In a recent study, Agarwal and colleagues83  showed increased expression of Cxcl12 by ECs and decreased expression by MSCs in a mouse model of CML. The study reported that loss of MSC-derived CXCL12 favors the loss of normal HSCs, whereas its increased expression by ECs favors the expansion of malignant cells. These findings suggest that the niche function of CXCL12 appears to be dependent on its cellular origin.83  Similar to mice, CXCL12 is expressed by both mesenchymal cells and ECs in the human BM.9  This data adds yet another layer of complexity to the niche; however, its relevance to the human system remains unexplored.

Sympathetic neuropathy

Neuropathy, which is characterized by decreased sympathetic nerve fibers and ensheathing Schwann cells, is readily observed in the BM of newly diagnosed MPN and AML patients.10,84  Moreover, denervated mice that were transplanted with primary human AML exhibited higher levels of BM infiltration compared with controls,85  suggesting that neuropathy exacerbates the AML phenotype and/or improves homing/engraftment of the AML cells. In a mouse model of MPN, the overproduction of interleukin-1β (IL-1β) by mutant progenitors was shown to damage the Schwann cells, which ensheath and protect the ending of the sympathetic nerve, leading to neuropathy, increased MSC apoptosis, and decreased expression of HSC retention factors/adhesion molecules by MSCs (Vcam1, Cxcl12, Kitl). This ultimately enforces mobilization of normal HSCs to the periphery.84,85  Importantly, high levels of IL1-β are readily detected in human MPN, possibly pointing to a conserved mechanism of MPN-induced neuropathy in mice and humans.84  Of note, a chronic increase in IL-1β is also anticipated to deplete normal HSCs through cell-intrinsic effects86  while concomitantly expanding malignant cells through binding to IL1RAP, a newly identified putative LSC marker and therapeutic target in MDS, AML, and CML.87-91 

Although sympathetic nerve degeneration is observed both in MPN and AML, experimental modulation in mice demonstrated that defective β3-adrenergic signals are responsible for the phenotypes observed in MPN, whereas β2-adrenergic signals seem to be the main mediators in AML. Although β3-AR agonist or IL-1β antagonist appear to be promising therapeutic strategies, the use of β2-AR agonists is mitigated by the risk of promoting the growth of the AML LSCs, which also express β2-AR.

Notably, the relevance of sympathetic neuropathy in the emergence or progression of other myeloid neoplasms, such as MDS or CML, remains to be tested.

Mesenchymal niche remodeling

BM fibrosis, a consequence of extensive mesenchymal niche remodeling, is observed, to various degrees, across the full spectrum of myeloid malignancies, where it often correlates with poor outcome.92-94 

The concept of leukemia-induced fibrosis was elegantly exemplified by the landmark study from the Passegué laboratory,95  demonstrating that mouse CML cells are able, through a combination of direct cell-cell contact and soluble factors (TPO and CCL3), to stimulate the overproduction of osteoblastic cells by MSCs, thereby leading to fibrosis. Likewise, our group recently demonstrated that, in a coculture model, human primary MDS cells were sufficient to instruct stromal cells from healthy volunteers to adopt MDS-like features, such as high expression of proangiogenic factors (VEGFA, IGFs, and EGFs) and mediators of fibrosis (LOXL, TGF-β, and LIF) (Medyouf et al67  and unpublished data). The functional relevance of these changes is supported by experimental data demonstrating that MDS diseased stem cells specifically rely on their disease-associated MSCs for disease propagation in vivo.67  Lastly, preliminary data from a phase II clinical trial indicate that treatment of JAK2V617F-positive MPN patients with a β3-AR agonist (mirabergon) slightly improves the fibrotic manifestations associated with MPN, possibly pointing to a conserved mechanism of MPN-induced fibrosis in humans and mice.84,96 

Reduced expression of essential HSC niche factors (CXCL12, ANGPT1, and KITL) and increased expression of proinflammatory factors (TNF, IL-6, IL-8, CCL3, and S100A8/A9) has also been documented in unmanipulated CD271+ MSCs.26,73  For instance, in mice, damage-associated molecular pattern (DAMP) molecules S100A8/A9 DAMPs, are shown to activate TLR4 signaling in HSPCs and induce genotoxic stress, a mechanism by which niche cells may cooperate with aberrant HSPCs toward leukemogenic transformation. Notably, S100A8/A9 signaling in mesenchymal niche cells appears to be an independent predictor of disease outcome in human MDS.73  Moreover, S100A9 binding to CD33 is also reported to expand myeloid-derived suppressor cells, which contribute to the development of myelodysplastic phenotypes in mice.97  Importantly, human myeloid-derived suppressor cells also express CD3397  and their number correlates with the number of regulatory T cells and disease progression in MDS patients.98  Taken together, these data argue in favor of a critical role of niche-derived S100A8/A9 in promoting disease evolution through both direct induction of genotoxic stress on hematopoietic cells and indirect mechanisms linked to increased immune suppression.

Vascular remodeling

Vascular remodeling is yet another unifying feature in myeloid malignancies, which likely contributes to disease progression (reviewed in Testa et al99 ). As such, increased microvessel density is predictive of a poor outcome in most myeloid malignancies.100-105  The activation of a leukemia-associated neoangiogenic program is instructed by proangiogenic factors released by the neoplastic cells, including VEGF, HGF, bFGF, and TNFα.106-109  Cytokine contribution to angiogenesis in non–BCR-ABL MPN has recently been reviewed elsewhere.110  VEGF, for instance, is highly expressed by megakaryocytes, myeloid progenitors, and granulocytes in chronic and blast crisis CML,111  with increased levels also observed in human66,107  and mouse models of MDS,112  where it appears to be equally secreted by malignant and stromal niche cells. EC activation by proinflammatory cytokines secreted by malignant cells (eg, TNFα, IL-6, and IL-1β) increases EC proliferation, EC expression of myeloid-promoting cytokines (G-CSF, granulocyte-macrophage–CSF [GM-CSF]) and might compromise vascular integrity. In mice, experimental alterations of endothelial barrier integrity trigger profound negative effects both on HSCs and other nonhematopoietic niche components,16  pointing to a potential mechanism by which vascular niche remodeling may contribute to malignant progression. Along these lines, recent evidence from the Bonnet laboratory113  show that human AML cells induce increased vascular permeability in PDX models, a phenomenon that appears to be mediated by increased nitric oxide (NO) production. Most notably, the study shows that inhibition of NO production reverts vascular permeability, potentiates normal HSC function, and improves treatment response in PDX.113  Evaluation of this niche-therapeutic approach in other myeloid malignancies may have tremendous therapeutic implications. Lastly, IL-33–expressing ECs are reported to be increased in BM biopsies of BCR-ABL MPN patients114  and might also contribute to malignant progression. Indeed, IL-33 stimulates the expression of myeloid cytokines (IL-6 and GM-CSF) by myeloid and nonhematopoietic niche cells. It specifically promotes the proliferation and colony formation of human primary CD34+ MPN, but not of their normal counterpart.114  Of note, IL-33 is also a ligand for IL1RAP, which has emerged as a putative selective LSC marker and therapeutic target in higher-risk MDS, AML, and CML.87-89 

In summary, pharmacological approaches aiming at reverting niche changes are increasingly recognized to be of therapeutic interest. These include, but are not limited to, reverting vascular leakiness (eg, NO synthase inhibitors), sympathetic neuropathy (eg, neuroprotective agents), and stromal remodeling (eg, β3-AR agonists and CCL3 antagonists). Future work, aiming at identifying the instructive signals that alter the function of the HSC niche, will likely identify new potential therapeutic targets that could be used to prevent the acquisition of disease-promoting niche features and contribute to improving patient outcomes (Figure 2).

Figure 2.

BM alterations and emerging niche-targeted therapeutic strategies in myeloid malignancies. This schema summarizes the most recurrent niche changes observed in human myeloid malignancies and the pharmacological approaches being explored to target niche-leukemia interdependencies. Depicted niche changes reflect mesenchymal, neurological, and endothelial niche remodeling. Pharmacological approaches include strategies to revert vascular leakiness (eg, NO synthase inhibitors)113  and prevent nerve damage (eg, IL-1 antagonists, and neuroprotective agents such as 4-methylcatechol [4-MC]) or its impact on MSCs (β3-AR agonists, such as mirabegron in phase II clinical trial in MPN [identifier NCT02311569]).84,96  Approaches to prevent or overcome therapeutic resistance include the inhibition of the FAO by blocking the function of the rate-limiting enzyme CPT-1 (eg, etomoxir, ranolazine),115  or interfering with mitochondrial function using tigecycline.

Figure 2.

BM alterations and emerging niche-targeted therapeutic strategies in myeloid malignancies. This schema summarizes the most recurrent niche changes observed in human myeloid malignancies and the pharmacological approaches being explored to target niche-leukemia interdependencies. Depicted niche changes reflect mesenchymal, neurological, and endothelial niche remodeling. Pharmacological approaches include strategies to revert vascular leakiness (eg, NO synthase inhibitors)113  and prevent nerve damage (eg, IL-1 antagonists, and neuroprotective agents such as 4-methylcatechol [4-MC]) or its impact on MSCs (β3-AR agonists, such as mirabegron in phase II clinical trial in MPN [identifier NCT02311569]).84,96  Approaches to prevent or overcome therapeutic resistance include the inhibition of the FAO by blocking the function of the rate-limiting enzyme CPT-1 (eg, etomoxir, ranolazine),115  or interfering with mitochondrial function using tigecycline.

Close modal

The role of the niche in therapeutic resistance has recently been discussed.2  However, novel emerging means of niche-mediated therapeutic resistance will be discussed in this section. In particular, new studies pointing to heterocellular transfer of material as well as metabolic adaptation allowing LSCs to strive in novel or harsh environments will be discussed.

Heterocellular transfer of material

Moschoi and colleagues116  demonstrated the existence of a contact-dependent transfer of functional mitochondria from MSCs to AML cells in vivo, leading to a 1.5-fold increase in energy production and significantly better survival rate upon chemotherapy. Mitochondrial transfer was increased under chemotherapy conditions, suggesting the existence of specific, but yet to be identified, “feed me signals” from the leukemic cells that instructed the transfer.116  A similar study reported transfer of mitochondria from primary MSCs to AML blasts ex vivo and in vivo (in PDX) and provided additional evidence that this unidirectional transfer was sensitive to cytochalasin treatment, highlighting the possible involvement of tunneling nanotubes.117  A study from the Lapidot laboratory118  also supports this mechanism. However, in this case, the mitochondria transfer was primarily directed from the AML blasts toward the stromal cells, suggesting the existence of bidirectional mitochondrial transfer between leukemic HSCs and their BM stromal cells. In line with these AML data, tyrosine kinase inhibitor–resistant CML stem cells appear to be dependent on mitochondrial oxidative metabolism for their survival.119  Consequently, CML LSCs could be efficiently targeted in vivo by a Food and Drug Administration–approved pharmacological inhibitor of mitochondrial protein synthesis, namely tigecycline used in combination with imatinib (Figure 2).

In summary, although the functional significance of this transfer is in its infancy, targeting mitochondrial transfer or function could possibly emerge as a new means to sensitize leukemic cells or overcome therapeutic resistance.

Metabolic adaptation

Another means of resistance has recently been reported by the Jordan laboratory,115  which identified a specific subset of LSCs, in blast crisis CML and AML, that were able to highjack the lipolytic process in gonadal adipose tissue to fuel their metabolic needs and evade chemotherapy-induced apoptosis. These cells expressed the fatty acid transporter, CD36. Interestingly, high expression of CD36 in AML was previously shown to be associated with a poor prognosis.120  Moreover, the fatty acid oxidation (FAO) pathway has been suggested as an alternative carbon-producing anabolic process used by cancer cells, but its most notable effect in leukemia could rely on its ability to interfere with the oligomerization of BAX and BAK in response to apoptotic stimuli.121,122  Consequently, carnitine O-palmitoyltransferase 1 (CPT-1), a protein involved in FAO, has been shown to have antiapoptotic functions in myeloid malignancies.123  A CPT-1 inhibitor, etomoxir,124  inhibits FAO and sensitizes leukemic cells to therapeutic challenge (Figure 2). These data provide evidence for yet another mechanism by which specific niches might shelter defined LSC subsets and promote their therapeutic resistance, thereby providing new opportunities for therapeutic intervention.

The cellular and molecular mechanisms that contribute to the age-associated increase in myeloid malignancies remain poorly defined. Beyond the well-recognized cell-intrinsic changes affecting aged HSCs,125-130  emerging studies in mice point to the importance of a complex remodeling and intrinsic modifications in HSC-supportive niche cells that are detrimental to HSC function. These include nerve damage and dramatic reduction of HSC niche-forming vessels, which lead to the acquisition of niche features that are reminiscent of the patient’s situation, such as increased vascular density and expanded MSCs that exhibit decreased stem cell potential and reduced expression of HSC maintenance factors (CXCL12, SCF, and ANGPT1).18,131  In 1 study, these complex phenotypes could be reverted by the EC-specific activation of Notch signaling,18  whereas, in the second study, nerve damage–mediated niche aging and HSC functional decline could be rescued by β3-AR agonists.131  Functionally, transplantation of young HSCs to aged recipients results in reduced homing and engraftment, increased myeloid output, and a shift toward reduced clonality.132,133  Conversely, exposure of aged HSCs to a young environment reduces the myeloid skewing.134  These experimental data support the hypothesis that an aged environment could contribute to the emergence of a myeloid skewing phenotype and possibly promote myeloid malignancies. In support of this, a mouse study showed that aged animals transplanted with AML1-ETO–positive HSCs exhibited more severe myeloproliferative phenotypes and increased immature myeloid load than young recipients.133  Likewise, aged MSCs exhibit decreased colony-forming unit fibroblast potential, increased adipogeneic output, and increased senescence both in mice and human.135,136  These changes are associated with reduced hematopoietic support.137,138 

In summary, these studies argue in favor of a prominent role of niche-related extrinsic changes in the pathogenesis of myeloid malignancies, in particular as potential mediators of normal HSC loss, which consequently would provide a selective advantage to mutant HSCs. It would thus be of great interest to reevaluate the impact of hematopoietic-specific mutations when introduced in aged HSCs and/or exposed to an aged environment. This could lead to the development of therapeutic approaches to improve the regenerative potential of the niche, which is critical in the context of a BM stem cell transplant setting, while possibly also interfering with detrimental phenotypes that actively promote disease progression (eg, β3-AR agonists MPN).

In addition to genetic mouse models, the development of highly immune-compromised model systems, such as the NOD-SCIDPrkdcIL2rγ−/− (NSG) mouse strain,139  which allow for the in vivo propagation of human cancers in a relevant environment, is a matter of intense investigation. Notably, propagation of human myeloid leukemia in vivo remains challenging, even for a subset of AML, a fully penetrant and deadly disease in patients. Besides the notoriously poor engraftment of MDS37,140-142  and MPNs,143,144  ∼50% of AMLs fail to engraft immune-compromised hosts.145  This fraction of “engrafter” cases could be further increased in AML by using new mouse lines that express a number of human cytokines that are poorly or not at all cross-reactive between mice and humans.146  Some of these mouse lines have been discussed in an excellent review by Goyama and colleagues,147  whereas new models are detailed in Table 1 and are briefly discussed below. The NSGS model has been reported to improve engraftment of AML148  as well as a very limited number of chronic-phase CML cases.88  However, in the context of MDS PDX, excessive myeloid stimulatory signals in this mouse line was reported to lead to the rapid exhaustion of malignant stem cells (CD34+)67  or engraftment of healthy HSPCs.43  New, improved models expressing human cytokines that are knocked-in to the endogenous mouse loci, namely MITRG or MISTRG (Table 1), have demonstrated engraftment of “favorable risk “AML cases149  that are notoriously difficult to propagate in vivo and were used to demonstrate the specific dependency of inv16 AMLs on exogenous niche-derived macrophage-CSF signals.149  The ability of such models to support other myeloid malignancies or AML subtypes remains to be tested. Last but not least, NSG models expressing a hypomorphic Kit receptor variant (NBSGW[Kitw41],150  NSGKitWv, or NSGKitw41151 ) have recently been generated. These kit mutants weaken the interaction of endogenous HSCs with kit ligand (SCF)–expressing niche cells, thereby providing a competitive advantage for exogenous HSCs expressing wild-type Kit variant. These mice are of particular interest when investigating niche-HSC or niche-LSC interactions because they do not require preconditioning and therefore would allow modeling in an intact (nondamaged) niche. In addition to improving the mouse models per se, experimental models in which malignant HSC engraftment was improved by coinjection of MSC cell lines or patient-derived MSCs64,152  strongly emphasize the importance of a humanized niche. Importantly, initial studies show that PDX models can, at least in part, preserve intraindividual heterogeneity, thereby partially recapitulating the disease complexity seen in patients.39,44,67 

Table 1.

Advances in immune-deficient mice for xenograft studies

Genetic backgroundRagIL-2rγSirpαc-KitHuman cytokinesWT HSCLeukemia models
NSG NOD WT Null NOD Sirpα WT None 139  Widely used 
NSGS NOD WT Null NOD Sirpα WT Transgenic hIL3/hGM-CSF/hSCF — AML148 ; MDS43,67 ; CML88  
NSGW41(Wv) NOD WT Null NOD Sirpα W41/W41 None 151  — 
NBSGW NOD WT Null NOD Sirpα W41/W41 None 150  — 
C57BL/6 
MITRG BALB/c Null Null BALB/c Sirpα WT Knock-in:hIL3/ hMCSF/hTPO/hGM-CSF 158  Inv16/NPM1 AMLs149  
MISTRG BALB/c Null Null Transgenic hSIRPa WT Knock-in:hIL3/ hMCSF/hTPO/hGM-CSF 158  Inv16/NPM1 AMLs149  
Genetic backgroundRagIL-2rγSirpαc-KitHuman cytokinesWT HSCLeukemia models
NSG NOD WT Null NOD Sirpα WT None 139  Widely used 
NSGS NOD WT Null NOD Sirpα WT Transgenic hIL3/hGM-CSF/hSCF — AML148 ; MDS43,67 ; CML88  
NSGW41(Wv) NOD WT Null NOD Sirpα W41/W41 None 151  — 
NBSGW NOD WT Null NOD Sirpα W41/W41 None 150  — 
C57BL/6 
MITRG BALB/c Null Null BALB/c Sirpα WT Knock-in:hIL3/ hMCSF/hTPO/hGM-CSF 158  Inv16/NPM1 AMLs149  
MISTRG BALB/c Null Null Transgenic hSIRPa WT Knock-in:hIL3/ hMCSF/hTPO/hGM-CSF 158  Inv16/NPM1 AMLs149  

The table provides a brief overview of the recent mouse models that have significant potential to improve the engraftment of human myeloid malignancies. Previous models are detailed in a recent review by Goyama and colleagues.147  Of note, due to high levels of cytokines, the NSGS model does not support long-term engraftment of normal hHSCs (H.M. and A. Trumpp, unpublished data). Note that both NBSGW and NSGW (W41 or Wv) lines do not require preconditioning prior to human HSC/leukemia transplantation. The NBSGW line has a mixed genetic background. MISTRG/MITRG have the advantage of expressing endogenous levels of human cytokines and are reported to engraft favorable-risk AMLs.

— , Not determined; NSG, NOD-SCIDPrkdcIL2rγ−/−; WT, wild-type.

Finally, to overcome the limitation imposed by the limited availability of patient material, several laboratories have embarked on experimental approaches that allow for the genetic engineering of human stem/progenitor cells to interrogate the effects of individual or combinatorial occurrence of specific somatic events (reviewed in Goyama et al147 ), which are being further developed thanks to the multiplexing power offered by CRISPR-Cas technology.153  Likewise, several laboratories are developing 3-dimensional (3D) niche models that mimic human bone structure and composition to propagate primary specimens both in vitro and in vivo upon implantation to mice. Such models were successfully used to engraft AMLs, blast crisis CML,154-156 and a limited number of MPNs (myelofibrosis).157 

These emerging models represent invaluable and cost-effective platforms for preclinical studies. However, propagation of chronic diseases, such as chronic phase CML, MDS, and a majority of MPNs, remains a challenge and may reflect their dependency on a more complex environment that has coevolved with the diseased cells, as was recently demonstrated in our niche-dependent PDX model of MDS.67  Of note, current 3D scaffolds rely on MSCs isolated from healthy volunteers, which are molecularly and functionally distinct from disease-“educated” ones. Moreover, the vasculature is exclusively of mouse origin or supported by transformed human ECs. The use of patient-derived niche components and biologically relevant scaffold materials may further improve these models and help interrogate the role of intercellular communication in disease-relevant accessible systems that are amenable to experimental manipulation.

Our understanding of niche contributions to hematological malignancies has tremendously increased over the past decade; however, many open questions remain, in particular, as to whether the observed phenotypes and molecular mechanisms identified in mouse models are conserved and therapeutically relevant in the human situation. This inevitably relies on a better understanding of the architecture and the cellular interactions at play in the human BM and how these are modulated during aging. Such studies will without doubt benefit from new technologies, such as high performance in vivo imaging and in situ analysis of core biopsies. Prospective studies linking age-associated bone phenotypes and hematological malignancies may also help address some of these pending questions.

More work is also required to interrogate the “functional” niche dependency of human LSCs as well as their in situ localization in the human BM. Are these features influenced by disease subtype, disease stage, and/or even genetic make-up? Can we devise niche-targeting strategies that hamper the growth of diseased cells, while concomitantly improving the fitness of residual normal HSCs (or at the very least preleukemic subclones)? Importantly, such niche-directed treatment strategies could possibly be applicable to a wide range of patients, across different disease subtypes, if conserved mechanisms are at play. In addition, resistance is less likely to emerge because these therapies will target normal cells with more limited cellular plasticity.

Functional assessment of niche contribution requires the establishment of disease-relevant model systems. Beyond PDX models, bone tissue engineering approaches are being used to build 3D model systems that aim to closely recapitulate the human BM niche complexity. Whether such systems maintain the complex genetic diversity seen in individual patients remains to be seen. Can they be used to interrogate the role of age-associated niche changes on disease initiation? Can they test whether age- or leukemia-induced niche changes are reversible? Can we use them to assess the (clone-specific) response to therapeutic challenge or to explore mechanisms of niche-mediated resistance and/or mimic minimal residual disease or relapse?

Finally, the applicability of easy-to-use genome editing technologies to human cells, such as CRISPR/Cas, opens new possibilities for modeling genetic complexity and moving forward with functional investigations using primary human cells instead of ill-defined cell lines that often do not recapitulate the outcomes observed with primary patient material.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The author thanks Henner Farin for critical reading of the manuscript. The author apologizes to the authors of the many excellent studies that are not included in this concise review.

H.M. is supported by the European Research Council (ERC grant no. 639795) and the German José Carreras Leukemia Foundation (award no. DJCLS A 14/01).

Contribution: H.M. wrote the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Hind Medyouf, Georg-Speyer-Haus Institute for Tumor Biology and Experimental Therapy, Paul-Ehrlich-Str, 42-44, 60596 Frankfurt, Germany; e-mail: hind.medyouf@medyouflab.com and medyouf@gsh.uni-frankfurt.de.

1.
Hoggatt
J
,
Kfoury
Y
,
Scadden
DT
.
Hematopoietic stem cell niche in health and disease
.
Annu Rev Pathol
.
2016
;
11
:
555
-
581
.
2.
Korn
C
,
Méndez-Ferrer
S
.
Myeloid malignancies and the microenvironment.
Blood
.
2017
;
129
(
7
):
811
-
822
.
3.
Guezguez
B
,
Campbell
CJ
,
Boyd
AL
, et al
.
Regional localization within the bone marrow influences the functional capacity of human HSCs
.
Cell Stem Cell
.
2013
;
13
(
2
):
175
-
189
.
4.
Notta
F
,
Doulatov
S
,
Laurenti
E
,
Poeppl
A
,
Jurisica
I
,
Dick
JE
.
Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment
.
Science
.
2011
;
333
(
6039
):
218
-
221
.
5.
Flores-Figueroa
E
,
Varma
S
,
Montgomery
K
,
Greenberg
PL
,
Gratzinger
D
.
Distinctive contact between CD34+ hematopoietic progenitors and CXCL12+ CD271+ mesenchymal stromal cells in benign and myelodysplastic bone marrow
.
Lab Invest
.
2012
;
92
(
9
):
1330
-
1341
.
6.
Chen
B
,
Pei
GX
,
Jin
D
,
Wei
KH
,
Qin
Y
,
Liu
QS
.
Distribution and property of nerve fibers in human long bone tissue
.
Chin J Traumatol
.
2007
;
10
(
1
):
3
-
9
.
7.
Katayama
Y
,
Battista
M
,
Kao
WM
, et al
.
Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow
.
Cell
.
2006
;
124
(
2
):
407
-
421
.
8.
Méndez-Ferrer
S
,
Lucas
D
,
Battista
M
,
Frenette
PS
.
Haematopoietic stem cell release is regulated by circadian oscillations
.
Nature
.
2008
;
452
(
7186
):
442
-
447
.
9.
Flores-Figueroa
E
,
Gratzinger
D
.
Beyond the niche: myelodysplastic syndrome topobiology in the laboratory and in the clinic
.
Int J Mol Sci
.
2016
;
17
(
4
):
553
.
10.
Chen
C
,
Liu
Y
,
Hua
M
,
Li
X
,
Ji
C
,
Ma
D
.
Neuropathy correlated with imbalanced Foxp3/IL-17 in bone marrow microenvironment of patients with acute myeloid leukemia
.
Oncotarget
.
2016
;
7
(
17
):
24455
-
24465
.
11.
Tormin
A
,
Li
O
,
Brune
JC
, et al
.
CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization
.
Blood
.
2011
;
117
(
19
):
5067
-
5077
.
12.
Lucas
D
,
Battista
M
,
Shi
PA
,
Isola
L
,
Frenette
PS
.
Mobilized hematopoietic stem cell yield depends on species-specific circadian timing
.
Cell Stem Cell
.
2008
;
3
(
4
):
364
-
366
.
13.
Spiegel
A
,
Shivtiel
S
,
Kalinkovich
A
, et al
.
Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling
.
Nat Immunol
.
2007
;
8
(
10
):
1123
-
1131
.
14.
Kiel
MJ
,
Yilmaz
OH
,
Iwashita
T
,
Yilmaz
OH
,
Terhorst
C
,
Morrison
SJ
.
SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells
.
Cell
.
2005
;
121
(
7
):
1109
-
1121
.
15.
Sipkins
DA
,
Wei
X
,
Wu
JW
, et al
.
In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment
.
Nature
.
2005
;
435
(
7044
):
969
-
973
.
16.
Acar
M
,
Kocherlakota
KS
,
Murphy
MM
, et al
.
Deep imaging of bone marrow shows non-dividing stem cells are mainly perisinusoidal
.
Nature
.
2015
;
526
(
7571
):
126
-
130
.
17.
Itkin
T
,
Gur-Cohen
S
,
Spencer
JA
, et al
.
Distinct bone marrow blood vessels differentially regulate haematopoiesis
.
Nature
.
2016
;
532
(
7599
):
323
-
328
.
18.
Kusumbe
AP
,
Ramasamy
SK
,
Itkin
T
, et al
.
Age-dependent modulation of vascular niches for haematopoietic stem cells
.
Nature
.
2016
;
532
(
7599
):
380
-
384
.
19.
Kunisaki
Y
,
Bruns
I
,
Scheiermann
C
, et al
.
Arteriolar niches maintain haematopoietic stem cell quiescence
.
Nature
.
2013
;
502
(
7473
):
637
-
643
.
20.
Ponzoni
M
,
Savage
DG
,
Ferreri
AJ
, et al
.
Chronic idiopathic myelofibrosis: independent prognostic importance of bone marrow microvascular density evaluated by CD105 (endoglin) immunostaining
.
Mod Pathol
.
2004
;
17
(
12
):
1513
-
1520
.
21.
Seandel
M
,
Butler
JM
,
Kobayashi
H
, et al
.
Generation of a functional and durable vascular niche by the adenoviral E4ORF1 gene
.
Proc Natl Acad Sci USA
.
2008
;
105
(
49
):
19288
-
19293
.
22.
Butler
JM
,
Gars
EJ
,
James
DJ
,
Nolan
DJ
,
Scandura
JM
,
Rafii
S
.
Development of a vascular niche platform for expansion of repopulating human cord blood stem and progenitor cells
.
Blood
.
2012
;
120
(
6
):
1344
-
1347
.
23.
Churchman
SM
,
Ponchel
F
,
Boxall
SA
, et al
.
Transcriptional profile of native CD271+ multipotential stromal cells: evidence for multiple fates, with prominent osteogenic and Wnt pathway signaling activity
.
Arthritis Rheum
.
2012
;
64
(
8
):
2632
-
2643
.
24.
Quirici
N
,
Soligo
D
,
Bossolasco
P
,
Servida
F
,
Lumini
C
,
Deliliers
GL
.
Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies
.
Exp Hematol
.
2002
;
30
(
7
):
783
-
791
.
25.
Li
H
,
Ghazanfari
R
,
Zacharaki
D
, et al
.
Low/negative expression of PDGFR-α identifies the candidate primary mesenchymal stromal cells in adult human bone marrow
.
Stem Cell Rep
.
2014
;
3
(
6
):
965
-
974
.
26.
Chen
S
,
Zambetti
NA
,
Bindels
EM
, et al
.
Massive parallel RNA sequencing of highly purified mesenchymal elements in low-risk MDS reveals tissue-context-dependent activation of inflammatory programs
.
Leukemia
.
2016
;
30
(
9
):
1938
-
1942
.
27.
Matsuoka
Y
,
Nakatsuka
R
,
Sumide
K
, et al
.
Prospectively isolated human bone marrow cell-derived MSCs support primitive human CD34-negative hematopoietic stem cells
.
Stem Cells
.
2015
;
33
(
5
):
1554
-
1565
.
28.
Zhou
BO
,
Yue
R
,
Murphy
MM
,
Peyer
JG
,
Morrison
SJ
.
Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow
.
Cell Stem Cell
.
2014
;
15
(
2
):
154
-
168
.
29.
Isern
J
,
Martín-Antonio
B
,
Ghazanfari
R
, et al
.
Self-renewing human bone marrow mesenspheres promote hematopoietic stem cell expansion
.
Cell Reports
.
2013
;
3
(
5
):
1714
-
1724
.
30.
Mabuchi
Y
,
Morikawa
S
,
Harada
S
, et al
.
LNGFR(+)THY-1(+)VCAM-1(hi+) cells reveal functionally distinct subpopulations in mesenchymal stem cells
.
Stem Cell Rep
.
2013
;
1
(
2
):
152
-
165
.
31.
Pinho
S
,
Lacombe
J
,
Hanoun
M
, et al
.
PDGFRα and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion
.
J Exp Med
.
2013
;
210
(
7
):
1351
-
1367
.
32.
Crisan
M
,
Yap
S
,
Casteilla
L
, et al
.
A perivascular origin for mesenchymal stem cells in multiple human organs
.
Cell Stem Cell
.
2008
;
3
(
3
):
301
-
313
.
33.
Méndez-Ferrer
S
,
Michurina
TV
,
Ferraro
F
, et al
.
Mesenchymal and haematopoietic stem cells form a unique bone marrow niche
.
Nature
.
2010
;
466
(
7308
):
829
-
834
.
34.
Morhayim
J
,
van de Peppel
J
,
Braakman
E
, et al
.
Osteoblasts secrete miRNA-containing extracellular vesicles that enhance expansion of human umbilical cord blood cells
.
Sci Rep
.
2016
;
6
:
32034
.
35.
Huntly
BJ
,
Gilliland
DG
.
Leukaemia stem cells and the evolution of cancer-stem-cell research
.
Nat Rev Cancer
.
2005
;
5
(
4
):
311
-
321
.
36.
Nilsson
L
,
Astrand-Grundström
I
,
Anderson
K
, et al
.
Involvement and functional impairment of the CD34(+)CD38(-)Thy-1(+) hematopoietic stem cell pool in myelodysplastic syndromes with trisomy 8
.
Blood
.
2002
;
100
(
1
):
259
-
267
.
37.
Nilsson
L
,
Astrand-Grundström
I
,
Arvidsson
I
, et al
.
Isolation and characterization of hematopoietic progenitor/stem cells in 5q-deleted myelodysplastic syndromes: evidence for involvement at the hematopoietic stem cell level
.
Blood
.
2000
;
96
(
6
):
2012
-
2021
.
38.
Rowley
JD
.
Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining
.
Nature
.
1973
;
243
(
5405
):
290
-
293
.
39.
Mossner
M
,
Jann
JC
,
Wittig
J
, et al
.
Mutational hierarchies in myelodysplastic syndromes dynamically adapt and evolve upon therapy response and failure
.
Blood
.
2016
;
128
(
9
):
1246
-
1259
.
40.
Goardon
N
,
Marchi
E
,
Atzberger
A
, et al
.
Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia
.
Cancer Cell
.
2011
;
19
(
1
):
138
-
152
.
41.
Levis
M
,
Murphy
KM
,
Pham
R
, et al
.
Internal tandem duplications of the FLT3 gene are present in leukemia stem cells
.
Blood
.
2005
;
106
(
2
):
673
-
680
.
42.
Jamieson
CH
,
Ailles
LE
,
Dylla
SJ
, et al
.
Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML
.
N Engl J Med
.
2004
;
351
(
7
):
657
-
667
.
43.
Rouault-Pierre
K
,
Smith
AE
,
Mian
SA
, et al
.
Myelodysplastic syndrome can propagate from the multipotent progenitor compartment
.
Haematologica
.
2017
;
102
(
1
):
e7
-
e10
.
44.
Woll
PS
,
Kjällquist
U
,
Chowdhury
O
, et al
.
Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo [published correction appears in Cancer Cell. 2015;22(9):1490]
.
Cancer Cell
.
2014
;
25
(
6
):
794
-
808
.
45.
Boyd
AL
,
Campbell
CJ
,
Hopkins
CI
, et al
.
Niche displacement of human leukemic stem cells uniquely allows their competitive replacement with healthy HSPCs
.
J Exp Med
.
2014
;
211
(
10
):
1925
-
1935
.
46.
Ishikawa
F
,
Yoshida
S
,
Saito
Y
, et al
.
Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region
.
Nat Biotechnol
.
2007
;
25
(
11
):
1315
-
1321
.
47.
Hawkins
ED
,
Duarte
D
,
Akinduro
O
, et al
.
T-cell acute leukaemia exhibits dynamic interactions with bone marrow microenvironments
.
Nature
.
2016
;
538
(
7626
):
518
-
522
.
48.
Shastri
A
,
Will
B
,
Ulrich
S
,
Verma
A
.
Stem and progenitor cell alterations in myelodysplastic syndromes
.
Blood
.
2017
;
129
(
12
):
1586
-
1594
.
49.
Mead
AJ
,
Mullally
A
.
Myeloproliferative neoplasm stem cells
.
Blood
.
2017
;
129
(
12
):
1607
-
1616
.
50.
Thomas
D
,
Majeti
R
.
Biology and relevance of human acute myeloid leukemia stem cells
.
Blood
.
2017
;
129
(
12
):
1577
-
1585
.
51.
Walkley
CR
,
Shea
JM
,
Sims
NA
,
Purton
LE
,
Orkin
SH
.
Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment
.
Cell
.
2007
;
129
(
6
):
1081
-
1095
.
52.
Walkley
CR
,
Olsen
GH
,
Dworkin
S
, et al
.
A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency
.
Cell
.
2007
;
129
(
6
):
1097
-
1110
.
53.
Kim
YW
,
Koo
BK
,
Jeong
HW
, et al
.
Defective Notch activation in microenvironment leads to myeloproliferative disease
.
Blood
.
2008
;
112
(
12
):
4628
-
4638
.
54.
Wang
L
,
Zhang
H
,
Rodriguez
S
, et al
.
Notch-dependent repression of miR-155 in the bone marrow niche regulates hematopoiesis in an NF-κB-dependent manner
.
Cell Stem Cell
.
2014
;
15
(
1
):
51
-
65
.
55.
Dong
L
,
Yu
WM
,
Zheng
H
, et al
.
Leukaemogenic effects of Ptpn11 activating mutations in the stem cell microenvironment
.
Nature
.
2016
;
539
(
7628
):
304
-
308
.
56.
Rupec
RA
,
Jundt
F
,
Rebholz
B
, et al
.
Stroma-mediated dysregulation of myelopoiesis in mice lacking I kappa B alpha
.
Immunity
.
2005
;
22
(
4
):
479
-
491
.
57.
Raaijmakers
MH
,
Mukherjee
S
,
Guo
S
, et al
.
Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia
.
Nature
.
2010
;
464
(
7290
):
852
-
857
.
58.
Kode
A
,
Manavalan
JS
,
Mosialou
I
, et al
.
Leukaemogenesis induced by an activating β-catenin mutation in osteoblasts
.
Nature
.
2014
;
506
(
7487
):
240
-
244
.
59.
Kode
A
,
Mosialou
I
,
Manavalan
SJ
, et al
.
FoxO1-dependent induction of acute myeloid leukemia by osteoblasts in mice
.
Leukemia
.
2016
;
30
(
1
):
1
-
13
.
60.
Schepers
K
,
Campbell
TB
,
Passegué
E
.
Normal and leukemic stem cell niches: insights and therapeutic opportunities
.
Cell Stem Cell
.
2015
;
16
(
3
):
254
-
267
.
61.
Wiseman
DH
.
Donor cell leukemia: a review
.
Biol Blood Marrow Transplant
.
2011
;
17
(
6
):
771
-
789
.
62.
Blau
O
,
Baldus
CD
,
Hofmann
WK
, et al
.
Mesenchymal stromal cells of myelodysplastic syndrome and acute myeloid leukemia patients have distinct genetic abnormalities compared with leukemic blasts
.
Blood
.
2011
;
118
(
20
):
5583
-
5592
.
63.
Blau
O
,
Hofmann
WK
,
Baldus
CD
, et al
.
Chromosomal aberrations in bone marrow mesenchymal stroma cells from patients with myelodysplastic syndrome and acute myeloblastic leukemia
.
Exp Hematol
.
2007
;
35
(
2
):
221
-
229
.
64.
Flores-Figueroa
E
,
Arana-Trejo
RM
,
Gutiérrez-Espíndola
G
,
Pérez-Cabrera
A
,
Mayani
H
.
Mesenchymal stem cells in myelodysplastic syndromes: phenotypic and cytogenetic characterization
.
Leuk Res
.
2005
;
29
(
2
):
215
-
224
.
65.
Huang
JC
,
Basu
SK
,
Zhao
X
, et al
.
Mesenchymal stromal cells derived from acute myeloid leukemia bone marrow exhibit aberrant cytogenetics and cytokine elaboration
.
Blood Cancer J
.
2015
;
5
:
e302
.
66.
von der Heide
EK
,
Neumann
M
,
Vosberg
S
, et al
.
Molecular alterations in bone marrow mesenchymal stromal cells derived from acute myeloid leukemia patients [published online ahead of print 11 November 2016]
.
Leukemia
.
doi:10.1038/leu.2016.324
.
67.
Medyouf
H
,
Mossner
M
,
Jann
JC
, et al
.
Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit
.
Cell Stem Cell
.
2014
;
14
(
6
):
824
-
837
.
68.
Kim
Y
,
Jekarl
DW
,
Kim
J
, et al
.
Genetic and epigenetic alterations of bone marrow stromal cells in myelodysplastic syndrome and acute myeloid leukemia patients
.
Stem Cell Res (Amst)
.
2015
;
14
(
2
):
177
-
184
.
69.
Geyh
S
,
Oz
S
,
Cadeddu
RP
, et al
.
Insufficient stromal support in MDS results from molecular and functional deficits of mesenchymal stromal cells
.
Leukemia
.
2013
;
27
(
9
):
1841
-
1851
.
70.
Santamaría
C
,
Muntión
S
,
Rosón
B
, et al
.
Impaired expression of DICER, DROSHA, SBDS and some microRNAs in mesenchymal stromal cells from myelodysplastic syndrome patients
.
Haematologica
.
2012
;
97
(
8
):
1218
-
1224
.
71.
Pleyer
L
,
Valent
P
,
Greil
R
.
Mesenchymal stem and progenitor cells in normal and dysplastic hematopoiesis-masters of survival and clonality?
Int J Mol Sci
.
2016
;
17
(
7
):
E1009
.
72.
Lopez-Villar
O
,
Garcia
JL
,
Sanchez-Guijo
FM
, et al
.
Both expanded and uncultured mesenchymal stem cells from MDS patients are genomically abnormal, showing a specific genetic profile for the 5q- syndrome
.
Leukemia
.
2009
;
23
(
4
):
664
-
672
.
73.
Zambetti
NA
,
Ping
Z
,
Chen
S
, et al
.
Mesenchymal inflammation drives genotoxic stress in hematopoietic stem cells and predicts disease evolution in human pre-leukemia
.
Cell Stem Cell
.
2016
;
19
(
5
):
613
-
627
.
74.
Teofili
L
,
Martini
M
,
Iachininoto
MG
, et al
.
Endothelial progenitor cells are clonal and exhibit the JAK2(V617F) mutation in a subset of thrombotic patients with Ph-negative myeloproliferative neoplasms
.
Blood
.
2011
;
117
(
9
):
2700
-
2707
.
75.
Zhan
H
,
Lin
CHS
,
Segal
Y
,
Kaushansky
K
.
JAK2V617F-mutant vascular niche promotes JAK2V617F clonal expansion in myeloproliferative neoplasms [abstract]
.
Blood
.
2016
;
128
(
22
).
Abstract 952
.
76.
Hochstetler
CL
,
Feng
Y
,
Zheng
Y
.
An altered bone marrow vascular niche impacts normal hematopoiesis and tilts the balance between myelopoiesis and lymphopoiesis [abstract]. Blood. 2016;128(22). Abstract 174
.
77.
Peled
A
,
Grabovsky
V
,
Habler
L
, et al
.
The chemokine SDF-1 stimulates integrin-mediated arrest of CD34(+) cells on vascular endothelium under shear flow
.
J Clin Invest
.
1999
;
104
(
9
):
1199
-
1211
.
78.
Peled
A
,
Kollet
O
,
Ponomaryov
T
, et al
.
The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice
.
Blood
.
2000
;
95
(
11
):
3289
-
3296
.
79.
Peled
A
,
Petit
I
,
Kollet
O
, et al
.
Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4
.
Science
.
1999
;
283
(
5403
):
845
-
848
.
80.
Foster
K
,
Lassailly
F
,
Anjos-Afonso
F
,
Currie
E
,
Rouault-Pierre
K
,
Bonnet
D
.
Different motile behaviors of human hematopoietic stem versus progenitor cells at the osteoblastic niche
.
Stem Cell Rep
.
2015
;
5
(
5
):
690
-
701
.
81.
Broxmeyer
HE
,
Orschell
CM
,
Clapp
DW
, et al
.
Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist
.
J Exp Med
.
2005
;
201
(
8
):
1307
-
1318
.
82.
Liles
WC
,
Broxmeyer
HE
,
Rodger
E
, et al
.
Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist
.
Blood
.
2003
;
102
(
8
):
2728
-
2730
.
83.
Agarwal
P
,
Li
H
,
Paterson
AJ
,
He
J
,
Nagasawa
T
,
Bhatia
R
.
Role of CXCL12-expressing bone marrow populations in leukemic stem cell regulation [abstract]. Blood. 2016;128(22). Abstract 26
.
84.
Arranz
L
,
Sánchez-Aguilera
A
,
Martín-Pérez
D
, et al
.
Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms
.
Nature
.
2014
;
512
(
7512
):
78
-
81
.
85.
Hanoun
M
,
Zhang
D
,
Mizoguchi
T
, et al
.
Acute myelogenous leukemia-induced sympathetic neuropathy promotes malignancy in an altered hematopoietic stem cell niche
.
Cell Stem Cell
.
2014
;
15
(
3
):
365
-
375
.
86.
Pietras
EM
,
Mirantes-Barbeito
C
,
Fong
S
, et al
.
Chronic interleukin-1 exposure drives haematopoietic stem cells towards precocious myeloid differentiation at the expense of self-renewal
.
Nat Cell Biol
.
2016
;
18
(
6
):
607
-
618
.
87.
Ågerstam
H
,
Karlsson
C
,
Hansen
N
, et al
.
Antibodies targeting human IL1RAP (IL1R3) show therapeutic effects in xenograft models of acute myeloid leukemia
.
Proc Natl Acad Sci USA
.
2015
;
112
(
34
):
10786
-
10791
.
88.
Ågerstam
H
,
Hansen
N
,
von Palffy
S
, et al
.
IL1RAP antibodies block IL-1-induced expansion of candidate CML stem cells and mediate cell killing in xenograft models
.
Blood
.
2016
;
128
(
23
):
2683
-
2693
.
89.
Barreyro
L
,
Will
B
,
Bartholdy
B
, et al
.
Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS
.
Blood
.
2012
;
120
(
6
):
1290
-
1298
.
90.
Askmyr
M
,
Ågerstam
H
,
Hansen
N
, et al
.
Selective killing of candidate AML stem cells by antibody targeting of IL1RAP
.
Blood
.
2013
;
121
(
18
):
3709
-
3713
.
91.
Järås
M
,
Johnels
P
,
Hansen
N
, et al
.
Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein
.
Proc Natl Acad Sci USA
.
2010
;
107
(
37
):
16280
-
16285
.
92.
Vener
C
,
Fracchiolla
NS
,
Gianelli
U
, et al
.
Prognostic implications of the European consensus for grading of bone marrow fibrosis in chronic idiopathic myelofibrosis
.
Blood
.
2008
;
111
(
4
):
1862
-
1865
.
93.
Buesche
G
,
Teoman
H
,
Wilczak
W
, et al
.
Marrow fibrosis predicts early fatal marrow failure in patients with myelodysplastic syndromes
.
Leukemia
.
2008
;
22
(
2
):
313
-
322
.
94.
Della Porta
MG
,
Malcovati
L
,
Boveri
E
, et al
.
Clinical relevance of bone marrow fibrosis and CD34-positive cell clusters in primary myelodysplastic syndromes
.
J Clin Oncol
.
2009
;
27
(
5
):
754
-
762
.
95.
Schepers
K
,
Pietras
EM
,
Reynaud
D
, et al
.
Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche
.
Cell Stem Cell
.
2013
;
13
(
3
):
285
-
299
.
96
Drexler
B
,
Passweg
J
,
Bigler
M
, et al
.
Effects of the sympathicomimetic agonist mirabegron on disease course, mutant allele burden, marrow fibrosis, and nestin positive stem cell niche in patients with JAK2-mutated myeloproliferative neoplasms. a prospective multicenter phase II trial SAKK 33/14
.
2016
;
128
(
22
):
3108
.
97.
Chen
X
,
Eksioglu
EA
,
Zhou
J
, et al
.
Induction of myelodysplasia by myeloid-derived suppressor cells
.
J Clin Invest
.
2013
;
123
(
11
):
4595
-
4611
.
98.
Kittang
AO
,
Kordasti
S
,
Sand
KE
, et al
.
Expansion of myeloid derived suppressor cells correlates with number of T regulatory cells and disease progression in myelodysplastic syndrome
.
OncoImmunology
.
2015
;
5
(
2
):
e1062208
.
99.
Testa
U
,
Saulle
E
,
Castelli
G
,
Pelosi
E
.
Endothelial progenitor cells in hematologic malignancies
.
Stem Cell Investig
.
2016
;
3
:
26
.
100.
Hussong
JW
,
Rodgers
GM
,
Shami
PJ
.
Evidence of increased angiogenesis in patients with acute myeloid leukemia
.
Blood
.
2000
;
95
(
1
):
309
-
313
.
101.
Korkolopoulou
P
,
Viniou
N
,
Kavantzas
N
, et al
.
Clinicopathologic correlations of bone marrow angiogenesis in chronic myeloid leukemia: a morphometric study
.
Leukemia
.
2003
;
17
(
1
):
89
-
97
.
102.
Lundberg
LG
,
Lerner
R
,
Sundelin
P
,
Rogers
R
,
Folkman
J
,
Palmblad
J
.
Bone marrow in polycythemia vera, chronic myelocytic leukemia, and myelofibrosis has an increased vascularity
.
Am J Pathol
.
2000
;
157
(
1
):
15
-
19
.
103.
Padró
T
,
Ruiz
S
,
Bieker
R
, et al
.
Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia
.
Blood
.
2000
;
95
(
8
):
2637
-
2644
.
104.
Savic
A
,
Cemerikic-Martinovic
V
,
Dovat
S
, et al
.
Angiogenesis and survival in patients with myelodysplastic syndrome
.
Pathol Oncol Res
.
2012
;
18
(
3
):
681
-
690
.
105.
Shih
TT
,
Hou
HA
,
Liu
CY
, et al
.
Bone marrow angiogenesis magnetic resonance imaging in patients with acute myeloid leukemia: peak enhancement ratio is an independent predictor for overall survival
.
Blood
.
2009
;
113
(
14
):
3161
-
3167
.
106.
Aguayo
A
,
Kantarjian
H
,
Manshouri
T
, et al
.
Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes
.
Blood
.
2000
;
96
(
6
):
2240
-
2245
.
107.
Bellamy
WT
,
Richter
L
,
Sirjani
D
, et al
.
Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes
.
Blood
.
2001
;
97
(
5
):
1427
-
1434
.
108.
Janowska-Wieczorek
A
,
Majka
M
,
Marquez-Curtis
L
,
Wertheim
JA
,
Turner
AR
,
Ratajczak
MZ
.
Bcr-abl-positive cells secrete angiogenic factors including matrix metalloproteinases and stimulate angiogenesis in vivo in Matrigel implants
.
Leukemia
.
2002
;
16
(
6
):
1160
-
1166
.
109.
Sainson
RC
,
Johnston
DA
,
Chu
HC
, et al
.
TNF primes endothelial cells for angiogenic sprouting by inducing a tip cell phenotype
.
Blood
.
2008
;
111
(
10
):
4997
-
5007
.
110.
Hoermann
G
,
Greiner
G
,
Valent
P
.
Cytokine regulation of microenvironmental cells in myeloproliferative neoplasms
.
Mediators Inflamm
.
2015
;
2015
:
869242
.
111.
Krauth
MT
,
Simonitsch
I
,
Aichberger
KJ
, et al
.
Immunohistochemical detection of VEGF in the bone marrow of patients with chronic myeloid leukemia and correlation with the phase of disease
.
Am J Clin Pathol
.
2004
;
121
(
4
):
473
-
481
.
112.
Balderman
SR
,
Li
AJ
,
Hoffman
CM
, et al
.
Targeting of the bone marrow microenvironment improves outcome in a murine model of myelodysplastic syndrome
.
Blood
.
2016
;
127
(
5
):
616
-
625
.
113.
Passaro
D
,
Di Tullio
A
,
Abarrategi
A
, et al
.
Increased vascular permeability in the bone marrow microenvironment contributes to acute myeloid leukemia progression and drug response [abstract]
.
Blood
.
2016
;
128
:(
22
). Abstract
2662
.
114.
Mager
LF
,
Riether
C
,
Schürch
CM
, et al
.
IL-33 signaling contributes to the pathogenesis of myeloproliferative neoplasms
.
J Clin Invest
.
2015
;
125
(
7
):
2579
-
2591
.
115.
Ye
H
,
Adane
B
,
Khan
N
, et al
.
Leukemic stem cells evade chemotherapy by metabolic adaptation to an adipose tissue niche
.
Cell Stem Cell
.
2016
;
19
(
1
):
23
-
37
.
116.
Moschoi
R
,
Imbert
V
,
Nebout
M
, et al
.
Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy
.
Blood
.
2016
;
128
(
2
):
253
-
264
.
117
Marlein
C
,
Zaitseva
L
,
Piddock
R
, et al
.
Bone marrow mesenchymal stromal cells transfer their mitochondria to acute myeloid leukaemia blasts to support their proliferation and survival
[abstract]. Blood.
2016
;
128
(
22
). Abstract
772
.
118.
Golan
G
,
Wellendorf
A
,
Takihara
Y
, et al
.
Mitochondria transfer from hematopoietic stem and progenitor cells to Pdgfrα+/Sca-1−/CD48dim BM stromal cells via CX43 gap junctions and AMPK signaling inversely regulate ROS generation in both cell populations
[abstract]. Blood.
2016
;
128
(
22
). Abstract 5.
119.
Kuntz
EM
,
Baquero
P
,
Holyoake
TL
,
Gottlieb
E
,
Helgason
GV
.
Therapy resistant CML stem cells are dependent on mitochondrial oxidative metabolism for their survival [abstract]
.
Blood
.
2016
;
128
(
22
). Abstract
932
.
120.
Perea
G
,
Domingo
A
,
Villamor
N
, et al
;
CETLAM GroupSpain
.
Adverse prognostic impact of CD36 and CD2 expression in adult de novo acute myeloid leukemia patients
.
Leuk Res
.
2005
;
29
(
10
):
1109
-
1116
.
121.
Carracedo
A
,
Cantley
LC
,
Pandolfi
PP
.
Cancer metabolism: fatty acid oxidation in the limelight
.
Nat Rev Cancer
.
2013
;
13
(
4
):
227
-
232
.
122.
Samudio
I
,
Harmancey
R
,
Fiegl
M
, et al
.
Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction
.
J Clin Invest
.
2010
;
120
(
1
):
142
-
156
.
123.
Ricciardi
MR
,
Mirabilii
S
,
Allegretti
M
, et al
.
Targeting the leukemia cell metabolism by the CPT1a inhibition: functional preclinical effects in leukemias
.
Blood
.
2015
;
126
(
16
):
1925
-
1929
.
124.
Pike
LS
,
Smift
AL
,
Croteau
NJ
,
Ferrick
DA
,
Wu
M
.
Inhibition of fatty acid oxidation by etomoxir impairs NADPH production and increases reactive oxygen species resulting in ATP depletion and cell death in human glioblastoma cells
.
Biochim Biophys Acta
.
2011
;
1807
(
6
):
726
-
734
.
125.
Busque
L
,
Patel
JP
,
Figueroa
ME
, et al
.
Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis
.
Nat Genet
.
2012
;
44
(
11
):
1179
-
1181
.
126.
Genovese
G
,
Kähler
AK
,
Handsaker
RE
, et al
.
Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence
.
N Engl J Med
.
2014
;
371
(
26
):
2477
-
2487
.
127.
Xie
M
,
Lu
C
,
Wang
J
, et al
.
Age-related mutations associated with clonal hematopoietic expansion and malignancies
.
Nat Med
.
2014
;
20
(
12
):
1472
-
1478
.
128.
Pang
WW
,
Price
EA
,
Sahoo
D
, et al
.
Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age
.
Proc Natl Acad Sci USA
.
2011
;
108
(
50
):
20012
-
20017
.
129.
Rossi
DJ
,
Bryder
D
,
Zahn
JM
, et al
.
Cell intrinsic alterations underlie hematopoietic stem cell aging
.
Proc Natl Acad Sci USA
.
2005
;
102
(
26
):
9194
-
9199
.
130.
Chandel
NS
,
Jasper
H
,
Ho
TT
,
Passegué
E
.
Metabolic regulation of stem cell function in tissue homeostasis and organismal ageing
.
Nat Cell Biol
.
2016
;
18
(
8
):
823
-
832
.
131.
Maryanovich
M
,
Zahalka
AH
,
Pierce
H
, et al
.
Loss of adrenergic nerves in the bone marrow microenvironment drives an aging HSC niche phenotype [abstract]. Blood. 2016;128(22). Abstract 169
.
132.
Liang
Y
,
Van Zant
G
,
Szilvassy
SJ
.
Effects of aging on the homing and engraftment of murine hematopoietic stem and progenitor cells
.
Blood
.
2005
;
106
(
4
):
1479
-
1487
.
133.
Vas
V
,
Wandhoff
C
,
Dörr
K
,
Niebel
A
,
Geiger
H
.
Contribution of an aged microenvironment to aging-associated myeloproliferative disease
.
PLoS One
.
2012
;
7
(
2
):
e31523
.
134.
Ergen
AV
,
Boles
NC
,
Goodell
MA
.
Rantes/Ccl5 influences hematopoietic stem cell subtypes and causes myeloid skewing
.
Blood
.
2012
;
119
(
11
):
2500
-
2509
.
135.
Baxter
MA
,
Wynn
RF
,
Jowitt
SN
,
Wraith
JE
,
Fairbairn
LJ
,
Bellantuono
I
.
Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion
.
Stem Cells
.
2004
;
22
(
5
):
675
-
682
.
136.
Stenderup
K
,
Justesen
J
,
Clausen
C
,
Kassem
M
.
Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells
.
Bone
.
2003
;
33
(
6
):
919
-
926
.
137.
Naveiras
O
,
Nardi
V
,
Wenzel
PL
,
Hauschka
PV
,
Fahey
F
,
Daley
GQ
.
Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment
.
Nature
.
2009
;
460
(
7252
):
259
-
263
.
138.
Omatsu
Y
,
Seike
M
,
Sugiyama
T
,
Kume
T
,
Nagasawa
T
.
Foxc1 is a critical regulator of haematopoietic stem/progenitor cell niche formation
.
Nature
.
2014
;
508
(
7497
):
536
-
540
.
139.
Shultz
LD
,
Lyons
BL
,
Burzenski
LM
, et al
.
Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells
.
J Immunol
.
2005
;
174
(
10
):
6477
-
6489
.
140.
Benito
AI
,
Bryant
E
,
Loken
MR
, et al
.
NOD/SCID mice transplanted with marrow from patients with myelodysplastic syndrome (MDS) show long-term propagation of normal but not clonal human precursors
.
Leuk Res
.
2003
;
27
(
5
):
425
-
436
.
141.
Martin
MG
,
Welch
JS
,
Uy
GL
, et al
.
Limited engraftment of low-risk myelodysplastic syndrome cells in NOD/SCID gamma-C chain knockout mice
.
Leukemia
.
2010
;
24
(
9
):
1662
-
1664
.
142.
Thanopoulou
E
,
Cashman
J
,
Kakagianne
T
,
Eaves
A
,
Zoumbos
N
,
Eaves
C
.
Engraftment of NOD/SCID-beta2 microglobulin null mice with multilineage neoplastic cells from patients with myelodysplastic syndrome
.
Blood
.
2004
;
103
(
11
):
4285
-
4293
.
143.
Ishii
T
,
Zhao
Y
,
Sozer
S
, et al
.
Behavior of CD34+ cells isolated from patients with polycythemia vera in NOD/SCID mice
.
Exp Hematol
.
2007
;
35
(
11
):
1633
-
1640
.
144.
Wang
X
,
Prakash
S
,
Lu
M
, et al
.
Spleens of myelofibrosis patients contain malignant hematopoietic stem cells
.
J Clin Invest
.
2012
;
122
(
11
):
3888
-
3899
.
145.
Pearce
DJ
,
Taussig
D
,
Zibara
K
, et al
.
AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML
.
Blood
.
2006
;
107
(
3
):
1166
-
1173
.
146.
Manz
MG
.
Human-hemato-lymphoid-system mice: opportunities and challenges
.
Immunity
.
2007
;
26
(
5
):
537
-
541
.
147.
Goyama
S
,
Wunderlich
M
,
Mulloy
JC
.
Xenograft models for normal and malignant stem cells
.
Blood
.
2015
;
125
(
17
):
2630
-
2640
.
148.
Wunderlich
M
,
Chou
FS
,
Link
KA
, et al
.
AML xenograft efficiency is significantly improved in NOD/SCID-IL2RG mice constitutively expressing human SCF, GM-CSF and IL-3
.
Leukemia
.
2010
;
24
(
10
):
1785
-
1788
.
149.
Ellegast
JM
,
Rauch
PJ
,
Kovtonyuk
LV
, et al
.
inv(16) and NPM1mut AMLs engraft human cytokine knock-in mice
.
Blood
.
2016
;
128
(
17
):
2130
-
2134
.
150.
McIntosh
BE
,
Brown
ME
,
Duffin
BM
, et al
.
Nonirradiated NOD,B6.SCID Il2rγ-/- Kit(W41/W41) (NBSGW) mice support multilineage engraftment of human hematopoietic cells
.
Stem Cell Rep
.
2015
;
4
(
2
):
171
-
180
.
151.
Cosgun
KN
,
Rahmig
S
,
Mende
N
, et al
.
Kit regulates HSC engraftment across the human-mouse species barrier
.
Cell Stem Cell
.
2014
;
15
(
2
):
227
-
238
.
152.
Li
X
,
Deeg
HJ
.
Murine xenogeneic models of myelodysplastic syndrome: an essential role for stroma cells
.
Exp Hematol
.
2014
;
42
(
1
):
4
-
10
.
153.
Tothova
Z
,
Krill-Burger
J
,
Popova
K
, et al
.
Generation of models of human hematologic malignancies using CRISPR genome engineering
.
Blood
.
2016
;
128
(
22
):
741
.
154.
Antonelli
A
,
Noort
WA
,
Jaques
J
, et al
.
Establishing human leukemia xenograft mouse models by implanting human bone marrow-like scaffold-based niches
.
Blood
.
2016
;
128
(
25
):
2949
-
2959
.
155.
Sontakke
P
,
Carretta
M
,
Jaques
J
, et al
.
Modeling BCR-ABL and MLL-AF9 leukemia in a human bone marrow-like scaffold-based xenograft model
.
Leukemia
.
2016
;
30
(
10
):
2064
-
2073
.
156.
Abarrategi
A
,
Foster
K
,
Hamilton
A
, et al
.
Versatile humanized niche model enables study of normal and malignant human hematopoiesis
.
J Clin Invest
.
2017
;
127
(
2
):
543
-
548
.
157.
Reinisch
A
,
Thomas
D
,
Corces
MR
, et al
.
A humanized bone marrow ossicle xenotransplantation model enables improved engraftment of healthy and leukemic human hematopoietic cells
.
Nat Med
.
2016
;
22
(
7
):
812
-
821
.
158.
Rongvaux
A
,
Willinger
T
,
Martinek
J
, et al
.
Development and function of human innate immune cells in a humanized mouse model
.
Nat Biotechnol
.
2014
;
32
(
4
):
364
-
372
.
Sign in via your Institution