hematology 2010 lapidot 1 6

Upload: buggie

Post on 07-Apr-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/3/2019 Hematology 2010 Lapidot 1 6

    1/6

    The Brain-Bone-Blood Triad: Traffic Lights for Stem-Cell

    Homing andMobilization

    TsveeLapidot1 and Orit Kollet1

    1Immunology Department, Weizmann Institute, Rehovot, Israel

    Navigation of transplanted stem cells to their target organs is essential for clinical bone marrow reconstitution. Recent

    studies have established that hematopoietic stem cells (HSCs) dynamically change their features and location, shifting

    from quiescent and stationary cells anchored in the bone marrow to cycling and motile cells entering the circulation.

    These changes are driven by stress signals. Bidirectional migrations to and from the bone marrow are active processes

    that form the basis for HSC transplantation protocols. However, how and why HSCs enter and exit the bone marrow as

    part of host defense and repair is not fully understood. The development of functional, preclinical, immune-deficient

    NOD/SCID (non-obese diabetic-severe combined immunodeficiency) mice transplantation models has enabled the

    characterization of normal and leukemic human HSCs and investigation of their biology. Intensive research has

    revealed multiple tasks for the chemokine SDF-1 (stromal cell-derived factor-1, also known as CXCL12) in HSC

    interactions with the microenvironment, as well as the existence of overlapping mechanisms controlling stress-

    induced mobilization and enhanced HSC homing, sequential events of major physiological relevance. These processesentail dynamically interacting, multi-system aspects that link the bone marrow vasculature and stromal cells with the

    nervous and immune systems. Neural cues act as an external pacemaker to synchronize HSC migration and

    development to balance bone remodeling via circadian rhythms in order to address blood and immune cell production

    for the physiological needs of the body. Stress situations and clinical HSC mobilization accelerate leukocyte

    proliferation and bone turnover. This review presents the concept that HSC regulation by the brain-bone-blood triad via

    stress signals controls the bone marrow reservoir of immature and maturing leukocytes.

    IntroductionIt is not the strongest of the species that survives, nor the most

    intelligent that survives. It is the one that is the most adaptable to

    change (L.C. Megginson, 1963, commenting on the studies of

    Charles Darwin). The light is what guides you home, the warmth

    is what keeps you there (attributed to Ellie Rodriguez).

    The potential of hematopoietic stem cells (HSCs) to repopulate ablated

    bone marrow is the basis for their definition as true stem cells. They are

    dynamic and versatile cells with changing phenotypes. These primitive

    progenitor cells can modulate their motility, location, and cell cycle

    status. They respond to signals from their microenvironment as well as

    from remote organs. Such modulations include intensive changes in the

    repertoire of cytokines and their cell surface receptors, adhesion

    molecules, proteolytic enzyme activation, and cytoskeletal rearrange-

    ment. The changes are aimed at facilitating hematopoietic stem and

    progenitor cell (HSPC) migration and development, which enables

    their transformation from quiescent, tissue-anchored cells to motile,

    proliferating cells. These processes are also associated with the cell

    differentiation needed for the replenishment of the blood on demand

    with new cells with a finite life span, which is required for host defense

    and repair mechanisms.

    The terms homing and mobilization are used to describe the in

    vivo migration of circulating HSPCs into their physiologic site of

    hematopoiesis, the bone marrow, and their enhanced exit from this

    organ back to the blood, respectively. HSPCs egress from the bone

    marrow during steady-state homeostasis is at a very low rate.

    However, stress conditions such as bleeding, inflammation, and

    injury greatly amplify and intensify this process. These processes

    are mimicked by clinical mobilization, in which HSPCs are

    recruited from the bone marrow to the circulation by means of

    chemotherapy induction and repeated cytokine stimulation to ex-

    pand and harvest HSPCs for clinical transplantations.

    HSPC mobilization and enhanced homing share various regulators

    and overlapping mechanisms showing that these two processes are

    sequential events with physiological relevance. Both directional

    migration processes to and from the bone marrow involve adhesion

    to the vascular wall and crossing over the endothelial blood-bone

    marrow barrier. Recently, multiple studies have established that

    murine HSPCs are preserved in their primitive phenotype within the

    bone marrow. These cells are in close contact with various stromal

    cells, including endosteal bone-lining osteoblasts, endothelial and

    peri-arterial reticular cells, as well as nestin-positive cells, all

    robustly expressing the chemokine SDF-1, which is essential for

    maintaining murine HSC quiescence.1 These nursing microenviron-

    ments, defined as stem cell niches, protect HSCs and provide

    them with signals that maintain their quiescent, slow-dividing and

    stationary state.14 Detachment of HSCs from these niches is

    believed to be associated with their entry into the cell cycle,

    proliferation, and differentiation, which is also accompanied by

    increased migration and recruitment to the circulation (as discussed

    in 57).

    The nervous system, which is the bodys master regulator, has been

    implicated in controlling the immune system (discussed in 8). Neural

    and immune cells also regulate HSPC motility and development,

    both directly via secretion of neurotransmitters and myeloid cyto-

    kines and indirectly by way of bone remodeling processes. These

    HAM-WASSERMAN LECTURE

    Hematology2010 1

  • 8/3/2019 Hematology 2010 Lapidot 1 6

    2/6

    include bone formation by osteoblasts, bone resorption by oste-

    oclasts, circadian rhythms, and the dynamic nature of the stem-cell

    niches. The current understanding of leukocyte production on

    demand, the regulation of HSPC homing and mobilization, and the

    central role of the brain-blood-bone triad in these processes will be

    discussed herein.

    Essential Tools for Human HSPC Research:

    Functional, Preclinical, Immune-Deficient Mouse

    ModelsUnderstanding the mechanisms that regulate human HSPC homing

    and mobilization in vivo required the development of animal

    models for human cell engraftment. The development of functional,

    preclinical models of transplanted, immune-deficient NOD/SCID

    (non-obese diabetic-severe combined immunodeficiency) mice pro-

    moted significant progress in normal and leukemic human HSC

    research. Particularly, these models enabled the generation of

    chimeric mice engrafted with human HSPCs, which provided

    mechanistic insights into their phenotypic characterization, as well

    as the identification of molecules participating in the regulation of

    HSPC homing, retention, proliferation, differentiation, and mobili-

    zation. The SCID, NOD/SCID, and the later-derived immune-

    deficient murine models constitute an experimentally relevant

    microenvironment for human HSPCs due to their immunodefi-

    ciency, which makes them permissive for transplanted human cells.

    In addition, high homology and partial cross-reactivity exist be-

    tween human and murine regulating factors, such as cytokines,

    chemokines, adhesion molecules, and enzymes. This enhances the

    success of transplanted human cells in these mice. NOD/SCID-

    repopulating human HSPCs, mostly defined as CD34/CD38-/low/

    CXCR4 cells, can home in the bone marrow of sub-lethally

    irradiated NOD/SCID hosts and imbue the mouse with a high

    degree of multi-lineage hematopoiesis. This process can be repeated

    in secondary transplanted recipient mice.5 However, these models

    still suffer xenogeneic limitations, because HSPCs in mice do not

    always produce the same results as in humans. For example, human

    cord blood CD34 cells engraft in these mice better than bone

    marrow or mobilized CD34 cells. Chimeric, immune-deficient

    mice have also been used for establishing both homing and

    mobilization assays that identify and characterize molecular and

    cellular components participating in human HSPC motility pro-

    cesses. The NOD/SCID mouse model was also established as a

    functional, preclinical model for leukemic HSPC cells obtained

    from some acute myeloid leukemia (AML) and precursor B-acute

    lymphocytic leukemia (ALL) patients. The severity of leukemia in

    transplanted mice correlates highly with the aggressiveness of the

    disease from the derived patient, and thus can be used to predict

    clinical outcomes. Finally, NOD/SCID mice are also used to

    monitor homing of human stromal progenitors. After certain

    manipulations, human CD44 cells could navigate to the murine

    bone marrow via interactions with selectins.9

    Non-motile, Tissue-Anchored Quiescent HSPCs:Bone Marrow ResidentsHSPCs reside primarily in the bone marrow in contact with supportive

    stromal cells, which maintain their undifferentiated phenotype and

    developmental potential. Proliferation, differentiation, and release to

    the circulation during the steady state occur at low levels, providing the

    body with short-lived hematopoietic cells as well as immature and

    maturing leukocytes as part of host immunity. Stress signals accelerate

    and intensify these processes dramatically, producing leukocytes on

    demand as part of host defense and repair10 and to address the bodys

    immediate needs during emergency situations.10

    Various molecular anchors keep HSPCs in contact with stromal

    cells in the bone marrow.4 The chemokine SDF-1 plays a central

    role in HSPC regulation, with a dose, tissue, and context depen-

    dency. SDF-1 is expressed by murine bone marrow osteoblasts,

    endothelial, reticular, nestin-positive, and other stromal cell types.

    At the homeostatic basal expression level, SDF-1 acts via its major

    receptor, CXCR4, as a survival factor for bone marrow HSPCs,

    inducing their quiescence and retention. Mouse embryos that do not

    express SDF-1 or CXCR4 have multiple lethal defects, including

    the lack of bone marrow seeding by HSPCs. Human and murineSDF-1 are cross-reactive, which explains why human HSPC can

    home in and be retained within the bone marrow of transplanted

    NOD/SCID mice (as discussed in 1,5) HSPCs residing in the bone

    marrow express a wide array of adhesion molecules for attachment

    to the stromal supportive network. For example, immature human

    CD34 cells express CD44 at high levels while retained in the bone

    marrow.11,12 This adhesion molecule anchors HSPCs to endothelial

    cells of human bone marrow blood sinusoids or endosteal osteo-

    blasts via CD44s major ligand, hyaluronan. Proteolytic enzymes,

    such as the cell-surface MT1-MMP (membrane type 1 matrix

    metalloproteinase), cleave CD44, thus negating its function. These

    proteolytic enzymes are also expressed by immature human CD34

    HSPCs and by various myeloid cells.12 During steady state, these

    interactions are tightly controlled by RECK (reversion-inducing-cysteine-rich protein with Kazal motifs), which inhibits MT1-MMP

    and MMP-2/9 activity. Human CD34 progenitors residing in the

    bone marrow do not secrete MMP-2/9, while circulating progenitors

    do produce these enzymes (discussed in5). These processes are also

    controlled by RECK, which is also expressed by human CD34

    progenitors.12 Several other molecules, such as 1-integrin and

    Rac1/Rac2,13 have also been implicated in regulating HSPC reten-

    tion. Mechanisms mediating the retention of human CD34HSPCs

    in the bone marrow are illustrated in Figure 1A.

    Stress Signals: The Impetus for HSPC Migration andDevelopmentStress conditions due to bleeding, inflammation, or DNA damage

    introduce dramatic systemic and local changes, which have a large

    impact on HSPC regulation. These changes include detachment of the

    progenitors from their bone marrow docking sites, cell cycle entry,

    increased motility, and recruitment. These cells follow the stress signals

    via the circulation toward the injured tissues. Interestingly, long-term

    repopulating HSCs extensively proliferate in response to in vivo

    bacterial infection. Both infection and interferon (IFN)- administra-

    tion also facilitate HSPC mobilization, demonstrating an important role

    of IFN- in HSPC quiescence and mobilization.14

    Inflammation is also associated with increased production of

    granulocyte colony-stimulating factor (G-CSF), which triggers

    HSPC mobilization. This physiologic effect is clinically utilized to

    expand and mobilize HSPCs from the bone marrow to the circulat-

    ing blood in order to harvest progenitors for stem-cell transplanta-

    tions. Experiments in parabiotic mice with a shared circulation

    demonstrate that HSC mobilization and homing are sequential

    events that are both triggered and enhanced by G-CSF, since HSPC

    mobilization by repeated G-CSF stimulation also leads to increased

    homing and engraftment of the partner mouses bone marrow.57

    The bone marrow is a highly vascularized organ. Sinusoidal vessels

    are believed to be the site of HSPC exchange between the bone

    marrow and the circulating blood. Thus, endothelium integrity and

    the regulation of its permeability are pivotal in the endotheliums

    role as the gatekeeper of the blood-bone marrow barrier. Bone

    2 American Society of Hematology

  • 8/3/2019 Hematology 2010 Lapidot 1 6

    3/6

    marrow endothelium permeability is increased by total body

    irradiation, chemotherapy, and inflammation (as discussed in 3,5) all

    of which enhance HSPC bidirectional transmigration.

    HSPC Mobilization: Extensive RecruitmentMobilization by repeated G-CSF stimulation requires awakening

    the quiescent HSPCs, generating guiding signals, locally repressing

    the inhibitory attachment machinery, and gaining motility. Stress

    signals induce the detachment of HSCs from their docking sites and

    inhibit bone marrow retention-inducing factors. These signaling

    molecules can be delivered via the circulating blood from a remote

    site in an endocrine-regulated fashion. However, stress signals can

    also be transmitted in a paracrine-regulated fashion to HSPC via

    stromal cells within the neighboring bone marrow vicinity. The

    chemokine SDF-1 also provides a pivotal guiding signal. The basal

    expression of SDF-1, which retains HSPC quiescence during the

    steady state, is reduced during mobilization. Studies by our group

    and others have shown that G-CSF-induced mobilization is associ-

    ated with a transient increase in SDF-1 levels, which is then

    followed by reduced production and secretion and cleavage of this

    chemokine in the bone marrow by a host of proteolytic en-

    zymes.6,11,15,16 In a complex series of events, bone marrow HSPCs

    increase their CXCR4 expression and motility. Inflammatory sig-

    nals also expand and activate mature bone-resorbing osteoclasts. In

    addition to their traditional role in bone turnover, these cells are also

    implicated in HSPC release and mobilization. Particularly, cathep-

    sin K, the enzymatic hallmark of bone degradation by osteoclasts,

    can also degrade SDF-1, osteopontin, and the membrane-bound kit

    ligand, all of which are molecules that regulate HSPCs. 17

    Thrombolytic agents are also involved in G-CSF mobilization.18

    Interestingly, G-CSF stimulation leads to a reduction in SDF-1

    production in the bone marrow due to morphological and functional

    changes of the endosteal osteoblasts, major producers of this

    chemokine that are also involved in signaling from the nervous

    system.16,19 These events are followed by the proteolytic deactiva-

    tion of SDF-1 and, concomitantly, HSPC proliferation and differen-

    tiation. While SDF-1 is reduced in the bone marrow and CXCR4 is

    increased, the level of CXCR4 in the circulation does not change

    significantly. Bone marrow HSPCs become more motile, gain

    CXCR4 expression, and the proteolytic machinery is activated (as

    discussed in 11). In parallel, human CD34 cell adhesion via CD44

    is now down-regulated due to CD44s cleavage by MT1-MMP. This

    activation of human MT1-MMP in chimeric NOD/SCID mice is

    made possible by the down-regulation of its inhibitor, RECK. 12 All

    of these adaptations act in synergy to facilitate HSPC egress from

    the bone marrow. The levels of MT1-MMP expression by myeloid

    cells in the patients blood circulation are correlated with clinical

    mobilization, and mobilized human CD34 cells have reduced

    CD44 expression.12 In recent studies, the CXCR4 antagonist

    AMD3100 (plerixafor) was shown to rapidly mobilize human

    CD34 HSPC. AMD3100 synergizes with G-CSF to increase

    HSPC motility.20,21 This process also involves innate immunity

    components such as the complement cascade.22 It is interesting that

    the mobilization of HSPCs and endothelial progenitors share

    common mechanisms.23,24

    One of the pathways that G-CSF activates in HSPC is the reactive

    oxygen species (ROS) signaling pathway. By activating the c-Met/

    HGF (hepatocyte growth factor) axis, G-CSF increases production of

    ROS, which induces HSPC mobilization. Accordingly, ROS inhibition

    reduces both G-CSF-induced mobilization and the enhanced motility

    of HSPC,25 demonstrating the importance of ROS as a regulator of

    enhanced HSPC migration, proliferation, and differentiation, leading to

    reduced long-term stem cell repopulation. However, this transient

    increase in ROS is reversible, and the long-term repopulation potential

    can be restored by in vitro ROS inhibition (as discussed in 25).

    Guided by stress-induced signals, HSPCs are recruited to injured

    sites and tissues. We have shown that local damage induced in the

    Figure 1. (A) Retention of human CD34 HSPCs via SDF-1/CXCR4 interactions, CD44/HA interactions, and inhibition of MT1-MMP and MMP2/9 by

    RECK, all leading to tissue-anchored, quiescent CD34HSPCs. (B) G-CSF-induced mobilization of human CD34HSPCs via activation of

    osteoclasts, proteloytic enzymes (including up-regulation of surface MT1-MMP), cleavage of CD44 and SDF-1, and CXCR4 up-regulation, leading toCD34HSPC proliferation, differentiation, and increased recruitment to the circulation. (C) Homing of human CD34 HSPCs in transplanted,

    immune-deficient NOD/SCID mice preconditioned with total body irradiation. Increased SDF-1 levels in the murine bonemarrow endotheliumand

    endosteum region attract human CD34/CXCR4HSPC, while CXCR4-/lowHSPCs are mostly trapped in the circulation

    Hematology2010 3

  • 8/3/2019 Hematology 2010 Lapidot 1 6

    4/6

    liver of NOD/SCID chimeric mice increased the expression of

    SDF-1 in the injured liver. This increase caused the recruitment of

    human CD34 progenitors from the engrafted murine bone mar-

    row. The human cells were observed localizing in close proximity to

    SDF-1-expressing bile duct epithelial cells. Interestingly, SDF-1

    expression in the human liver is dramatically up-regulated by

    hepatitis C virus infection and in the murine liver by irradiation,

    which suggests that orchestrated changes induced by stress signals

    can potentially participate in host defense and repair by as-yet-

    unknown mechanisms (as discussed in 26).

    The bone marrow microstructure in general, and the trabecular

    endosteum region in particular, are critical regulators of HSPC fate

    and retention. Upon osteoclast malfunction, both steady-state re-

    lease and G-CSF-induced progenitor mobilization are preferentially

    reduced.17 Osteoclast perturbations can be genetically introduced in

    young PTP KO (protein tyrosine phosphatase-epsilon knockout)

    females or can be induced by anti-osteoclast drug therapy, which

    imposes changes in bone turnover.17

    G-CSF administration activates osteoclasts, which were recently shown

    to be involved in mediating the increased proliferation of bone marrow

    stromal progenitors.27 These observations suggest that osteoclasts

    participate notonly in bone resorption and HSPC mobilization, but alsoin the regulation of the bone marrow-supporting niches. Similarly, mice

    lacking CD45 have a distorted morphology and a reduction in

    osteoclast activity, resulting in mild osteopetrosis. These mice display

    extramedullar hematopoiesis and abnormal bone microstructure, mainly

    reflected in modified distribution and density of the bone trabecules,

    implicating reduced levels of murine HSPCs in the bone marrow.

    CD45 knockout mice poorly mobilize HSPCs in response to G-CSF,

    RANKL (receptor activatorfornuclear factorB ligand), andAMD3100

    stimulation.28 Mechanisms of G-CSF-induced mobilization of human

    CD34HSPCs in NOD/SCID mice are illustrated in Figure 1B. Taken

    together, these studies demonstrate the essential function of balanced

    osteoblast/osteoclast regulation and the bone marrow microenviron-

    ment to support and allow the mobilization process. These results

    suggest that via their progeny, osteoclasts, neutrophils, and other

    myeloid cells, HSCs indirectly participate in regulating the dynamic

    bone marrow microenvironment, the immature stromal and hematopoi-

    etic pool size, and the mobilization process.

    Several other mobilizing agents, such as antibodies for VLA-4

    (very-late antigen-4) and IL-8 (interleukin-8), have been identified;

    however, in this review, we chose to focus on those acting via the

    SDF-1/CXCR4 axis, which is a pivotal regulator of HSPC function.

    Homing: The Active Crossing of the Blood-Bone

    Marrow Barrier and Retainment in the Bone MarrowMicroenvironmentClinical protocols of bone marrow transplantations are dependent

    on the homing skills of the transplanted cells. Intravenously infused

    into the peripheral blood, HSPCs find their way to the bone marrow

    and lodge there to initiate hematopoiesis and bone marrow reconsti-

    tution, which is a multi-step, coordinated process.

    The homing of human CD34 HSPCs to immune-deficient murine

    bone marrow requires preconditioning of the host mice with total body

    irradiation or other means of ablation, such as chemotherapeutic-

    induced DNA damage. This induces a dramatic elevation of SDF-1

    production by bone marrow stromal cells within 1 to 2 d leading to

    activation of proteolytic enzyme machinery. Navigating human HSPCs

    can successfully migrate to the murine bone marrow if CXCR4 is

    functionally expressed on the CD34 cell surface (as discussed in 5).

    Expression of this receptor is dynamic and can be up-regulated by short

    term, 1- to 2-d pre-stimulation of CD34 cells ex vivo with SCF (Stem

    Cell Factor), IL-6, HGF, and other cytokines. Homing of human

    CD34 HSPCs in transplanted NOD/SCID mice is regulated by the

    availability of both SDF-1 as the guiding signal and of CXCR4 as the

    detector of this signal. Importantly, the migration potential of a

    patients CD34-enriched cells toward a gradient of SDF-1 in vitro is

    also correlated with their repopulation potential in clinical autologous

    transplantations. These results suggest that the repopulation potential ofhuman CD34 cells in patients and in chimeric NOD/SCID mice is

    dependent on CXCR4 signaling (as discussed in 5). CXCR4 is also

    functionally expressed by many malignant human cells and is a poor

    prognostic factor for AML patients.

    Bone marrow endothelial cells produce and present SDF-1, as well as

    its major receptor, CXCR4. Endothelial CXCR4 binds this chemokine

    and functionally transfers it from the circulation via the vessel lumen

    into the bone marrow. Bone marrow endothelial CXCR4 transfer of

    SDF-1can also facilitate the homing of transplanted human CD34

    HSPCs to the murine bone marrow.29 Interestingly, preliminary results

    reveal that SDF-1 injected directly into the bone was transferred by

    endothelial cells and released into the circulation in a CXCR4-

    dependent manner, facilitating rapid murine HSPC mobilization.11 This

    feature of the endothelial blood-bone marrow barrier is believed to

    constitute a major vectorial regulator directing HSPCs in or out of the

    bone marrow. This vectorial regulation follows SDF-1s dynamic

    fluctuations due to stress-induced enhanced production in the bone

    marrow or in other organs. In agreement with these observations, the

    inhibition of CD26, a peptidase with SDF-1 cleavage activity, im-

    proves homing and engraftment of human CD34 progenitors in

    transplanted, immune-deficient mice.30,31

    Similarly to HSPC mobilization, bone marrow homing also requires an

    active crossing of the blood-bone marrow barrier, namely the vascular

    wall. Many molecules expressed by endothelial cells take an active part

    in this process. HSC attachment to the vascular lumen requires binding

    of selectins and integrins to their receptors, which facilitates cell

    adhesion to the vessel wall under blood flow shear stress prior to their

    crossing. Presentation of SDF-1 by endothelial cells mediates increased

    expression and activation of the HSPC adhesion machinery, which

    involves cytoskeleton rearrangement to enable cell spreading (as

    discussed in 5). Anchors required for docking in the bone marrow

    include CD44, which is also expressed by human CD34 HSPCs.

    CD44 is required for cell spreading and adhesion to hyaluronan and

    osteopontin, both of which are expressed by blood vessel walls and

    along the endosteum, and are implicated together with SDF-1 and

    membrane-bound SCF in murine HSC homing and repopulation.3234

    Indeed, blocking CD44 function prevents homing of immature human

    CD34 cell to the murine bone marrow and spleen (as discussed in 5).

    Mechanisms regulating the homing of human CD34 HSPCs to the

    NOD/SCID mouse bone marrow are illustrated in Figure 1C.

    Activation of the adhesion machinery is dynamic and requires a

    tight regulation to allow HSPC attachment and detachment, both of

    which are essential for cell migration. For example, genetic

    modulation of CD45 leads to a constant and highly adhesive HSPC

    phenotype due to Src hyperactivation, which prevents their normal

    homing to the bone marrow of wild-type hosts.28 Lodging of

    homing HSPCs to the bone marrow endosteum also requires sensing

    of the calcium levels,35 which links bone turnover with the

    regulation of murine stem cell homing. Intracellular rho-GTPases

    are also important regulators of HSC motility and adhesion. 13

    4 American Society of Hematology

  • 8/3/2019 Hematology 2010 Lapidot 1 6

    5/6

    The Brain-Blood-Bone Triad: A Circular Hierarchy

    Regulator via Dynamic InteractionsThe nervous system via its various pathways has been implicated as

    a major regulator of the immune system. However, activated

    leukocytes, including lymphocytes and lipopolysaccharide-stimu-

    lated macrophages, also secrete neurotransmitters that exert bifunc-

    tional effects on the immune cells and provide feedback to the

    nervous system. The bone marrow is highly innervated, with nerve

    fibers running along the brachiated blood vessels and into the bone

    marrow parenchyma. Thus, catecholamines produced by the sympa-

    thetic system can be delivered to the bone marrow by the blood

    circulation, but some are also secreted from nerve endings directly

    in the bone marrow to act on bone marrow resident cells in a

    paracrine fashion.8,36 Accumulated data depict a major role for

    signals coming from the sympathetic system in the regulation of

    HSPC retention, homing, and mobilization, which include direct

    and indirect effects exerted via the bone-remodeling processes.

    Human CD34 HSPCs dynamically express catecholaminergic

    receptors, with higher expression observed by the more primitive

    CD34CD38-/low subset. Repeated stimulation with the mobiliz-

    ing myeloid cytokines G-CSF and GM-CSF (granulocyte-macro-

    phage colony stimulating factor) also increases this expression.

    Therefore, mobilized human CD34 cells isolated from the periph-eral blood have higher catecholaminergic receptor expression

    compared with tissue-anchored bone marrow CD34 progenitor

    cells. These receptors play a functional role in the homing process,

    because stimulation with neurotransmitters triggers in vitro CD34

    cell proliferation and increased motility and engraftment capabili-

    ties, as measured in chimeric NOD/SCID mice. Moreover, cat-

    echolaminergic stimuli also enhance the expression of the membrane-

    bound enzyme MT1-MMP and SDF-1-directed migration.36,37

    The Wnt canonical signaling pathway and beta-catenin stabilization are

    also activated by this catecholaminergic stimulation to take part in the

    homing process. Interestingly, we found that these neural signals can

    also induce murine HSPC mobilization.37 Indeed, antagonizing the

    Wnt signaling pathway with Dickkopf-1 during steady-state homeosta-sis mobilized both hematopoietic and endothelial murine progenitor

    cells.23,38 In contrast, during alarm situations and repeated G-CSF-

    induced mobilization, inhibition of Wnt signaling inhibits mobiliza-

    tion.37 Driving forces of HSPC mobilization also include the suppres-

    sive effects that G-CSF stimulation exerts on bone-lining osteoblasts

    and their capacity to produce SDF-1. This requires peripheral beta-

    adrenergic signals.19

    Sympathetic signals also control bone-remodeling processes, which

    constitute part of the HSPC paracrine regulatory milieu, including bone

    formation by osteoblasts and degradation by osteoclasts. Nerve endings

    with norepinephrine secretion capacity are observed in close proximity

    to osteoblasts and osteoclasts near the bone-growing plates, at regions

    enriched with HSPCs. Bone mass and remodeling processes aimed atmaintaining bone integrity are known to be regulated by signals from

    the nervous system. Particularly, beta-adrenergic signals that are

    involved in HSPC migration and mobilization tilt the remodeling

    balance toward osteoclast activation and osteoblast suppression. For

    example, parathyroid hormone regulates bone turnover and in vivo

    HSPC expansion and mobilization (as discussed in 8).

    The emerging relationship among the nervous, immune, and

    bone-maintaining systems may allow us to conceive a well-

    orchestrated hierarchal network by which the body functions in the

    context of HSPC regulation. The evidence that exogenous signals

    transmitted by light/dark cycles are detected by the nervous system,

    which then translates these circadian cues to neurotransmitter and

    hormone secretion, both major regulators of HSPCs and their

    microenvironment, places the nervous system at the top of this

    regulatory hierarchy. SDF-1, the major HSPC chemokine, is

    produced and secreted in the bone marrow in association with

    circadian rhythms in a mechanism that includes beta3-adrenergic

    (AdR) activity, which is not yet fully understood.8,39 HSPC release

    from the bone marrow to the circulation in steady-state conditions is

    also synchronized with and follows bone marrow SDF-1 produc-

    tion, with anti-phase fluctuations.39 Accordingly, expression ofCXCR4 by human bone marrow CD34 HSPC cells also demon-

    strates a circadian oscillation pattern.40 The effects of immune cells

    and bone disorders on the nervous system demonstrate the circular,

    bidirectional manner by which this network exerts its regulation.

    Concluding Remarks and Future DirectionsThe nervous system is the fastest detection and message-delivery

    framework, which makes it capable of immediate response to environ-

    mental changes throughout the body. This regulatory network demon-

    strates multidirectional streaming. Thus, exogenous stimuli and other

    stimuli originating from the immune system, bone, bone marrow, and

    the stromal microenvironment all regulate HSPCs. However, the

    nervous system dominates the brain-bone-blood triad.

    Prospectively, the major question is what does the body gain by

    responding to circadian signals in controlling bone turnover and HSPC

    trafficking? This question may be answered by seeing these body

    defense tools as constantly working at low gear to maintain homeo-

    static balance. Bone maintenance requires continuous formation and

    degradation. HSC regulation requires their quiescence and retention in

    the bone marrow, but also their migration, self-renewal, differentiation,

    and recruitment to the circulation. These opposing processes may

    require an on/off switch that reliably inverts the regulatory machinery

    in opposite directions. To continuously keep this network active, there

    could be a need for repetitive, low rates of mild, stress-like signals that

    are generated in constant intervals. These intervals are provided by

    circadian peaks, serving as an external pacemaker to synchronize blood

    cell production, bone turnover, and immunity. These circadian rhythmsalso keep the body ready and well trained for dynamic and acute

    changes, including intensified stem-cell migration and development,

    which are triggered by stress signals during acute situations. This

    concept is relevant to clinical mobilization and homing during stem-

    cell transplantation. Future protocols may harness these insights to

    increase the success of HSPC transplantation by taking into consider-

    ation normal circadian rhythms, the timing of G-CSF and AMD3100

    treatments, and unique features of the patients bone, immune, and

    neural status.

    AcknowledgementsWe regret that due to the citation limit of 40 references published

    during the last 5 years, we could not cite and discuss many

    important studies on stem-cell homing and mobilization.

    DisclosureConflict-of-interest disclosure: The authors declare no competing

    financial interests.

    Off-label drug use: None disclosed.

    References1. Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of

    the hematopoietic stem cell pool by CXCL12-CXCR4 chemo-

    kine signaling in bone marrow stromal cell niches. Immunity.

    2006;25:977988.

    Hematology2010 5

  • 8/3/2019 Hematology 2010 Lapidot 1 6

    6/6

    2. Kiel MJ, Morrison SJ. Uncertainty in the niches that maintain

    haematopoietic stem cells. Nat Rev Immunol. 2008;8:290 301.

    3. Butler JM, Nolan DJ, Vertes EL, et al. Endothelial cells are

    essential for the self-renewal and repopulation of Notch-dependent

    hematopoietic stem cells. Cell Stem Cell. 2010;6:251264.

    4. Lymperi S, Ferraro F, Scadden DT. The HSC niche concept has

    turned 31. Has our knowledge matured? Ann N Y Acad Sci.

    2010;1192:1218.

    5. Lapidot T, Dar A, Kollet O. How do stem cells find their way

    home? Blood. 2005;106:19011910.6. Nervi B, Link DC, DiPersio JF. Cytokines and hematopoietic

    stem cell mobilization. J Cell Biochem. 2006;99:690 705.

    7. Papayannopoulou T, Scadden DT. Stem-cell ecology and stem

    cells in motion. Blood. 2008;111:39233930.

    8. Spiegel A, Kalinkovich A, Shivtiel S, Kollet O, Lapidot T.

    Stem cell regulation via dynamic interactions of the nervous

    and immune systems with the microenvironment. Cell Stem

    Cell. 2008;3:484 492.

    9. Sackstein R, Merzaban JS, Cain DW, et al. Ex vivo glycan

    engineering of CD44 programs human multipotent mesenchymal

    stromal cell trafficking to bone.Nat Med. 2008;14:181187.

    10. Laird DJ, von Andrian UH, Wagers AJ. Stem cell trafficking in

    tissue development, growth, and disease. Cell. 2008;132:612 630.

    11. Dar A, Kollet O, Lapidot T. Mutual, reciprocal SDF-1/CXCR4interactions between hematopoietic and bone marrow stromal

    cells regulate human stem cell migration and development in

    NOD/SCID chimeric mice. Exp Hematol. 2006;34:967975.

    12. Vagima Y, Avigdor A, Goichberg P, et al. MT1-MMP and

    RECK are involved in human CD34 progenitor cell retention,

    egress, and mobilization. J Clin Invest. 2009;119:492503.

    13. Cancelas JA, Williams DA. Rho GTPases in hematopoietic

    stem cell functions. Curr Opin Hematol. 2009;16:249 254.

    14. Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA.

    Quiescent haematopoietic stem cells are activated by IFN-gamma

    in response to chronic infection.Nature. 2010;465:793797.

    15. Kopp HG, Avecilla ST, Hooper AT, Rafii S. The bone marrow

    vascular niche: home of HSC differentiation and mobilization.

    Physiology (Bethesda). 2005;20:349 356.

    16. Christopher MJ, Liu F, Hilton MJ, Long F, Link DC. Suppres-

    sion of CXCL12 production by bone marrow osteoblasts is a

    common and critical pathway for cytokine-induced mobiliza-

    tion. Blood. 2009;114:13311339.

    17. Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal

    components and promote mobilization of hematopoietic progeni-

    tor cells. Nat Med. 2006;12:657 664.

    18. Tjwa M, Janssens S, Carmeliet P. Plasmin therapy enhances

    mobilization of HPCs after G-CSF. Blood. 2008;112:40484050.

    19. Katayama Y, Battista M, Kao WM, et al. Signals from the

    sympathetic nervous system regulate hematopoietic stem cell

    egress from bone marrow. Cell. 2006;124:407 421.

    20. Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobiliza-

    tion of murine and human hematopoietic stem and progenitor

    cells with AMD3100, a CXCR4 antagonist. J Exp Med.

    2005;201:13071318.

    21. Pusic I, Dipersio JF. Update on clinical experience with

    AMD3100, an SDF-1/CXCL12-CXCR4 inhibitor, in mobiliza-

    tion of hematopoietic stem and progenitor cells. Curr Opin

    Hematol. 2010 Jul;17(4):319 326.

    22. Lee HM, Wysoczynski M, Liu R, et al. Mobilization studies in

    complement-deficient mice reveal that optimal AMD3100

    mobilization of hematopoietic stem cells depends on comple-

    ment cascade activation by AMD3100-stimulated granulocytes.

    Leukemia. 2010;24:573582.

    23. Aicher A, Kollet O, Heeschen C, et al. The Wnt antagonist

    Dickkopf-1 mobilizes vasculogenic progenitor cells via activa-

    tion of the bone marrow endosteal stem cell niche. Circ Res.

    2008;103:796 803.

    24. Pitchford SC, Furze RC, Jones CP, Wengner AM, Rankin SM.

    Differential mobilization of subsets of progenitor cells from the

    bone marrow. Cell Stem Cell. 2009;4:6272.

    25. Tesio M, Golan K, Corso S, et al. Enhanced c-Met activity

    promotes G-CSF induced mobilization of hematopoietic progeni-

    tor cells via ROS signaling. Blood. 2010 Jun 28. [Epub ahead ofprint]

    26. Kollet O, Dar A, Lapidot T. The multiple roles of osteoclasts in

    host defense: bone remodeling and hematopoietic stem cell

    mobilization. Annu Rev Immunol. 2007;25:51 69.

    27. Brouard N, Driessen R, Short B, Simmons PJ. G-CSF increases

    mesenchymal precursor cell numbers in the bone marrow via an

    indirect mechanism involving osteoclast-mediated bone resorp-

    tion. Stem Cell Res. 2010;5:6575.

    28. Shivtiel S, Kollet O, Lapid K, et al. CD45 regulates retention,

    motility, and numbers of hematopoietic progenitors, and affects

    osteoclast remodeling of metaphyseal trabecules. J Exp Med.

    2008;205:23812395.

    29. Dar A, Goichberg P, Shinder V, et al. Chemokine receptor

    CXCR4-dependent internalization and resecretion of functionalchemokine SDF-1 by bone marrow endothelial and stromal

    cells. Nat Immunol. 2005;6:1038 1046.

    30. Campbell TB, Hangoc G, Liu Y, Pollok K, Broxmeyer HE.

    Inhibition of CD26 in human cord blood CD34 cells enhances

    their engraftment of nonobese diabetic/severe combined immu-

    nodeficiency mice. Stem Cells Dev. 2007;16:347354.

    31. Bonig H, Priestley GV, Oehler V, Papayannopoulou T. Hematopoi-

    etic progenitor cells (HPC) from mobilized peripheral blood

    display enhanced migration and marrow homing compared to

    steady-state bone marrow HPC. Exp Hematol. 2007;35:326334.

    32. Haylock DN, Nilsson SK. Stem cell regulation by the hemato-

    poietic stem cell niche. Cell Cycle. 2005;4:13531355.

    33. Lo Celso C, Fleming HE, Wu JW, et al. Live-animal tracking of

    individual haematopoietic stem/progenitor cells in their niche.

    Nature. 2009;457:9296.

    34. Xie Y, Yin T, Wiegraebe W, et al. Detection of functional

    haematopoietic stem cell niche using real-time imaging. Na-

    ture. 2009;457:97101.

    35. Adams GB, Chabner KT, Alley IR, et al. Stem cell engraftment

    at the endosteal niche is specified by the calcium-sensing

    receptor. Nature. 2006;439:599 603.

    36. Kalinkovich A, Spiegel A, Shivtiel S, et al. Blood-forming stem

    cells are nervous: direct and indirect regulation of immature

    human CD34 cells by the nervous system. Brain Behav

    Immun. 2009;23:1059 1065.

    37. Spiegel A, Shivtiel S, Kalinkovich A, et al. Catecholaminergic

    neurotransmitters regulate migration and repopulation of imma-

    ture human CD34() cells through Wnt signaling. Nat Immu-

    nol. 2007;8:11231131.

    38. Fleming HE, Janzen V, Lo Celso C, et al. Wnt signaling in the

    niche enforces hematopoietic stem cell quiescence and is

    necessary to preserve self-renewal in vivo. Cell Stem Cell.

    2008;2:274283.

    39. Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haemato-

    poietic stem cell release is regulated by circadian oscillations.

    Nature. 2008;452:442 447.

    40. 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;3:364 366.

    6 American Society of Hematology