covid
Buscar en
Inmunología
Toda la web
Inicio Inmunología Hematopoietic stem cell homing: The long, winding and adhesive road to the bone ...
Información de la revista
Vol. 27. Núm. 1.
Páginas 22-35 (enero - marzo 2008)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
9434
Vol. 27. Núm. 1.
Páginas 22-35 (enero - marzo 2008)
Acceso a texto completo
Hematopoietic stem cell homing: The long, winding and adhesive road to the bone marow
Migración de células madre hematopoiéticas: El largo, tortuoso y adhesivo camino hacia la médula ósea
Visitas
9434
A. Hidalgo
Autor para correspondencia
andres.hidalgo@mssm.edu

Correspondence to: Division of Hematology, Oncology, Department of Medicine. Mount Sinai School of Medicine, One Gustave L. Levy Place; Box # 1079, New York, 10029 NY (USA). Phone: 212-659 9695.
Division of Hematology/Oncology, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (1)
Tablas (1)
Table I. Manipulation of HSPC homing to the bone marrow
Abstract

In adult mammals, a rare population of cells that gives rise to the hematopoietic compartment dwells in niches within the bone marrow where they are nourished and protected. These blood stem cells can, however, exit these niches under physiological conditions and, in the context of bone marrow transplantation, migrate to the recipient's marrow to regenerate hematopoiesis. This review will address old and new discoveries regarding the mechanisms that allow these stem cells to find their way home through long and winding roads: From the peripheral circulation, through interactions with the vessel-lining endothelial cells in the bone marrow, and into their niches located in the intramedullary space. It will also address how these discoveries have provided us with new tools to manipulate stem cell trafficking.

Key words:
Hematopoiesis
Stem cells
Progenitor cells
Bone marrow
Resumen

En mamíferos adultos, una población celular muy restringida que da origen al tejido hematopoyético se localiza en nichos dentro de la médula ósea en los que recibe sustento y protección. Estas células madre hematopoiéticas de la sangre pueden salir de estos nichos en condiciones fisiológicas y, en el contexto del transplante de médula ósea, migrar a la médula del recipiente para regenerar su sistema hematopoyético. En esta revisión se describen las evidencias que han permitido caracterizar los mecanismos por los cuales estas células madre migran a los nichos medulares siguiendo un camino largo y tortuoso: Desde la circulación periférica, y mediante interacciones con la vasculatura medular, hasta nichos localizados en el espacio intramedular. También se discute cómo se han generado nuevas herramientas que nos permitirán manipular el tráfico de células madre.

Palabras clave:
Hematopoyesis
Células madre
Células progenitoras
Médula ósea
Texto completo
THE VOYAGE

Hematopoietic stem and progenitor cells (HSPC) are determined travelers. Beginning from their embryonic origins, HSPC move from one niche to another: The first wave of adult-type hematopoietic progenitors in the mouse appears in the aorta-gonad-mesonephros region around embryonic day 9 (E9), and by E10.5 they relocate to the fetal liver and then to the spleen and the bone marrow (BM)(1). After birth, the BM provides the definitive lodgment for HSPC. The choice of this residence is not casual: The surrounding bone provides physical protection from external aggressions, and cellular elements of the marrow nourish and maintain HSPC. The latter constitute the so-called "stem cell niche", whose components we are only now starting to identify(2). Because this niche has the basic features of what we would define as a home, the migration of HSPC to the BM is commonly referred to as homing.

Three different locations and components of this niche have been reported. Osteoblasts lining the endosteal surface of the bone were the first cellular niche to be identified though elegant genetic approaches in the mouse(3,4). Novel tools that specifically label HSPC later provided visual demonstration of their localization in close contact with endothelial cells lining the vasculature of the BM and spleen(5). More recently, a reticular-like cell type scattered in the medullary parenchyma to which HSPC associated was identified(6). These cells were identified on the basis of being the main producers of CXCL12 (also known as stromal-cell derived factor-1, or SDF-1), a chemokine essential for HSPC retention in the BM. They were thus named CXCL12-abundant reticular (CAR) cells(6). Although this review does not intend to cover details about the HSPC niche, it is important to understand the basic features of the destination of the road that HSPC follow which such remarkable determination.

Because the niche is so important for the survival and proper differentiation of HSPC, one might ask why these cells would ever leave such a protective environment. Is there a need for HSPC to migrate or home back to this niche? Why is this process worth studying?

It has been known for over 40 years that HSPC are present in the circulating blood under physiological conditions(7,8). Although dependent on several factors, such as strain or pathophysiological state, the number of HSPC in the blood of a mouse at any given moment ranges from 10 to 60 (Daniel Lucas-Alcaraz and Paul Frenette, personal communication). These circulating HSPC are functional in that they are capable of restoring hematopoiesis as demonstrated in experiments using competitive reconstituting assays and parabiotic mice(9,10). Although the significance of these observations is presently unclear, these experiments demonstrated for the first time that HSPC routinely exit and re-enter the BM.

The relevance of this process is perhaps most salient in the clinical arena. The ability of HSPC to specifically home to the BM was first demonstrated in the early 1950s in experiments showing that pluripotent cells present in the spleen or BM were capable of repopulating hematopoietic organs(11,12). These observations paved the way toward the clinical use of bone marrow transplantation (BMT) to treat hematological malignancies(13). BMT is currently a common procedure used to restore hematopoiesis in patients undergoing myeloablation, and has been also used for gene therapy in patients suffering anemia and immunodeficiencies(14,15). In this context, HSPC obtained from a donor and infused into patients must find their way through a long and winding network of veins, arteries and capillaries in order to home to the BM.

The purpose of this review is to present a description of the molecular and cellular cues that account for the exquisite specificity and efficiency of HSPC homing to their niches in the BM. Two questions that underlie both features of the homing process will be addressed: What molecular components and receptors are required for the different stretches of this voyage? What cues provide specificity for the BM? In the last part of this review some of the strategies that have been devised to improve HSPC homing and enhance the recovery of patients undergoing BMT will be discussed.

TOOLS FOR THE ROAD

Leukocyte trafficking has been extensively investigated and the major molecular components mediating their recruitment to inflammatory sites identified. In a seminal review from 1994, Timothy Springer proposed a "code" for leukocyte trafficking that was specified by the different components of a multistep process in which the combination of molecules involved in each step dictated the route followed by each leukocyte subset during physiology and disease(16).

Upon inflammation, signals emanating from the injured tissue induce the expression of receptors in the endothelium that allow leukocytes to be recruited to that specific area. Molecules that are induced include endothelial selectins, receptors of the immunoglobulin superfamily and chemokines. Recruitment is initiated by high-affinity but short-lived interactions between leukocytes and the endothelial cells lining the inflamed vessels(17). These interactions, mediated by P-and E-selectins, result in a rolling motion that allow leukocytes to become further activated. Selectins recognize highly glycosylated ligands on the surface of leukocytes(18). P-selectin glycoprotein-1 (PSGL-1) is the main, if not only, physiological ligand for P-selectin(19,20). PSGL-1 is also a physiological ligand for E-selectin(20,21), but in this context it acts in coordination with other glycoproteins, such as CD43(22,23), CD44(24) and E-selectin-ligand 1 (ESL-1)(25,26).

Although ligation of selectin ligands can result in signaling and leukocyte activation, it is the binding of small and basic cytokines present on the endothelial luminal surface, termed chemokines, that triggers potent signals which result in integrin activation(27). Chemokine signaling is mediated by G-protein coupled receptors (GPCR) that span the plasma membrane seven times. Integrins thus activated promote firm adhesion to their endothelial ligands, particularly vascular cell adhesion molecule (VCAM)-1 and intercellular cell adhesion molecule (ICAM)-1(27,28). Integrins are heterodimeric proteins comprised of α and β subunits of which two subfamilies, β1 and β2, are predominantly expressed on leukocytes and can be activated upon chemokine induced signaling(28,29). Other molecules that do not belong to either of these receptor families, such as CD44, also play a role in leukocyte and HSPC trafficking, and will be discussed below.

INITIAL INTERACTIONS WITH THE BONE MARROW MICROVASCULATURE

HSPC trafficking is thought to essentially be similar to the process by which leukocytes migrate to inflamed tissues (Fig. 1). Like leukocytes, HSPC initiate their migration to the BM through transient and weak interactions with endothelial selectins(30-34). A feature of the BM microvasculature is that it constitutively expresses endothelial selectins, the integrin counter-receptor VCAM-1 and the chemokine CXCL12 which is displayed on its luminal surface(29,35,36). Discrete areas of HSPC or leukemic cell entry in the BM have indeed been shown to display strikingly restricted expression of CXCL12 and E-selectin in vivo(37). This remarkable characteristic is critical in allowing the trafficking of HSPC as well as other leukocyte subsets to the BM.

Figure 1.

Steps of stem cell homing to the bone marrow niches. The BM microvasculature constitutively expresses endothelial selectins, integrin counter receptors and the chemokine CXCL12. This allows HSPC circulating in the blood to initiate labile interactions and a rolling motion that are mediated by endothelial selectins and their ligands. As they roll, binding of CXCL12 present on the luminal surface of endothelial cells activates adhesive receptors on HSPC that result in their firm arrest and locomotion in search of areas suitable for transendothelial migration. Once in the marrow cavity, HSPC navigate in search of niches guided by CXCL12-dependent and independent signals (big arrows), where they establish contacts (niche contacts) that allow their long-term engraftment in the vascular, endosteal and CAR niches. Egress of HSPC from the BM occurs physiologically or after injection of certain drugs (such as G CSF) by disruption of their interactions with the niche (small arrows). Controversies and alternate uses of molecules at different steps of this scheme are described in the text. Circled numbers identify specific processes that have been targeted in order to manipulate HSPC trafficking (both homing and mobilization). These are listed in Table I and discussed in the text. END, endothelial cells; OB, osteoblasts.

(0,33MB).

HSPC express high levels of ligands for both P-and E-selectins(30-32), which initiate rolling through interactions with these molecules(32,34). Inhibition or absence of both selectins results in a strong reduction in both HSPC rolling on the BM microvasculature and homing to the BM(32-34,38). It should be noted, however, that human CD34+ cells (the subset that contains most HSPC) appear to be more dependent than murine HSPC on selectin-mediated interactions to enter the BM(34). While P-selectin is the major player in initiating these interactions, E-selectin is required to allow slow rolling and activation of the adhesive machinery(39). In vivo studies showed that PSGL-1 expressed on HSPC functions as the main ligand for both selectins(38); the same study also demonstrated that additional ligands were required for the interactions with E-selectin. These additional ligands have not been characterized, but studies with human CD34+ cells and murine neutrophils suggest that CD44 may be one such ligand(24,40). Because CD44 is also a receptor for hyaluronan (HA) on activated T lymphocytes and HSPC(41,42), and HA is expressed on the surface of the BM microvasculature(43), it is conceivable that it functions as a dual receptor for both E-selectin and HA during HSPC homing.

While selectins are clearly important in mediating these initial rolling interactions, activated integrins can also mediate leukocyte rolling(44,45). In studies aimed at assessing the contribution of α4 integrins in HSPC homing, we found that simultaneous blockage or absence of these integrins and E-selectin almost completely prevented entry of HSPC into the BM(38). Expression and function of the α4β1 (or VLA-4) integrin has been extensively reported in both human and murine HSPC(46,47). We were surprised, however, to find that a second α4 integrin, α4β7, was also expressed by murine HSPC(48). The integrin α4β7 and its primary counter receptor, mucosal addressin cell adhesion molecule-1 (MadCAM-1), are involved in the entry of T lymphocytes to intestinal lymphoid organs(49). Our studies demonstrated that both α4β7 and MadCAM-1, but not α4β1, were involved in mediating rolling interactions with the BM microvasculature(48). Whether this pathway is also used by human HSPC remains undetermined. The redundancies revealed by these studies, where multiple adhesive ligandreceptor pairs appear to mediate the initial interactions of HSPC, support the notion that this step is critical for the specificity and efficiency of the homing process.

TURN SIGNALS: CXCL12

The chemokine CXCL12 is an essential component of the hematopoietic niche(50). During homing, it is thought to provide the signals that indicate to an HSPC that it has arrived at the "target" organ and that it must take a turn at that specific location. Contrary to most chemokines, CXCL12 signals exclusively through one receptor, CXCR4. Although HSPC express other chemokine receptors, CXCR4-mediated signaling triggers the most potent physiological responses in these cells(51-53). CXCL12 is one of the most extensively studied chemokines, due in part to the dramatic phenotype of both CXCL12-and CXCR4-deficient mice and the involvement of CXCR4 in HIV infection(54-56). Studies in these mutant mice demonstrated an essential role for this signaling axis during hematopoiesis, including B lymphocyte development and colonization of the BM by HSPC and myeloid progenitors during ontogeny(57). A number of experimental observations further support the critical function of CXCL12 during hematopoiesis. First, it is expressed in the very cells that form the niche in which HSPC reside in the BM and contributes to their retention in those niches(6,29,58). This contribution was further evidenced in experiments where reduction of CXCL12 levels triggered by the cytokine G-CSF resulted in egress of HSPC into the peripheral circulation(59-61). Secondly, inhibition of the CXCL12/CXCR4 axis using antibodies(52) or pertussis toxin(62), or by genetic deletion(63), prevented HSPC homing to and engraftment in the BM.

At the mechanistic level, CXCL12 is thought to control homing by enhancing the adhesiveness of HSPC. At least in T lymphocytes, this is first achieved by the induction of microvilli collapse on the cell surface to allow for a larger area of cell contact with the endothelium(64). In human CD34+ cells, CXCL12 promotes a rapid and strong increase in the affinity and avidity of the β1 integrin α4β1 for its ligands, VCAM-1 and fibronectin(29,65). The functions of other adhesion receptors on HSPC, including α5β1(28), αLβ2(29), α4β7(48) and CD44(43) can also be modulated by CXCL12. These findings support a model in which CXCL12 exposed on the luminal side of the BM microvasculature(66) is detected by CXCR4 during HSPC rolling. Signals delivered by CXCR4 in turn activate integrin and non-integrin receptors on HSPC and promote their firm attachment to the BM vasculature(28,29).

Of the adhesive receptors modulated by CXCL12, β1 integrins in particular appear to play an essential role in HSPC homing to adult hematopoietic organs. Although β1-deficient HSPC have a normal capacity to differentiate into mature leukocytes in vitro, they are unable to colonize either the fetal liver during embryonic life or the BM of adult mice(67,68). A similar phenotype was observed in chimeric mice and in mice conditionally deficient in the α4 subunit of β1 and β7 integrins(69,70). Together, these experimental data suggest that CXCL12 and the α4β1 integrin act in coordination and are critical to mediate the arrest of HSPC on the BM microvasculature(29,48,71).

CROSSING THE FENCE

While the adhesive pathways promoting the initial interactions of HSPC with the BM microvasculature have been well studied, those mediating the movement on and through the endothelium remain less well characterized. Insights into these processes have come from in vitro studies that analyzed leukocytes moving on and transmigrating through cultured endothelial cells. Recent studies have shown that leukocytes "palpate" the endothelium while they move in search of areas of transmigration(72). This movement, termed locomotion, is mediated by β2 integrins on monocytes(73) and requires the active formation of sensing protrusions on the leukocyte(72). On the endothelial side, this process is paralleled by the formation of "docking" structures that are enriched in both integrin counter-receptors (VCAM-1 and ICAM-1) and in the tetraspanins CD9 and CD151(74,75). Transendothelial migration of mature leukocytes can then occur between or through endothelial cells (referred to as the paracellular or transcellular routes, respectively), and it is mediated by homophilic as well as heterophilic interactions via receptors such as ICAM-1, platelet endothelial cell adhesion molecule (PECAM)-1, CD99, VE-cadherin, endothelial selective adhesion molecule (ESAM), and the junctional adhesion molecules (JAM)-A, B and C(76,77). Partial contributions of PECAM-1, CD99 and VE-cadherin have been described during the transmigration of human CD34+ cells through BM endothelial cells using in vitro systems(78-80). There is still a considerable gap in our understanding of the mechanisms of HSPC transmigration through the microvasculature of the BM. It is believed, however, that signals triggered by the chemokine CXCL12 are required for the diapedesis of HSPC across the BM vasculature.

What are the signals triggered by CXCL12 that are required for HSPC homing? One family of molecules activated by CXCR4 signaling has been thoroughly studied in the context of HSPC homing and engraftment. Rac1 and Rac2 belong to the Rac subfamily of Rho guanosine triphosphatases, and display a wide and hematopoietic-restricted pattern of expression, respectively(81). They play important roles in cytoskeletal reorganization, proliferation and survival of HSPC(82). In vitro migration of HSPC in response to CXCL12 was abolished in the absence of both Rac proteins(83). Although mice deficient in either protein display distinct hematopoietic defects, only the absence of Rac1 resulted in a partial deficiency in HSPC homing to the BM and an abnormal HSPC localization within the medullary space, and these deficiencies were further accentuated when both Rac proteins were absent(84). Since Rac proteins are important in the formation of lamellipodia and membrane ruffling in response to exogenous factors, these studies demonstrated redundant and important roles for Rac1 and Rac2 in integrating signals that induce HSPC motility in response to CXCL12(83,84). Associated with the function of Rac proteins, the guanine exchange factor Vav1 controls adhesive (e.g., α4 integrin activation) and chemotactic responses to CXCL12(85,86). Upstream of Rac1, cyclic AMP levels can control transendothelial migration and motility through modulation of CXCR4 levels on the cell surface. This mechanism, which also requires the (PKCζ)(87,88), may be important in all other steps of the homing process that involve CXCL12. In addition, HSPC express a number of receptors for other biological mediators such as lipids (sphingosine-1-phosphate)(89), chemoattractants (C3a)(90), nucleotides (UTP)(91) and neurotransmitters (dopamine, epinephrine or norephinephrine)(92) that can potentiate their transendothelial migration and adhesion through modulation of CXCR4-mediated signaling. Downstream of CXCR4 signaling, in vitro studies have shown that HSPC stimulated with CXCL12 or with cytokines produce matrix metalloproteases (MMP2 and MMP9) that facilitate their migration across basement membranes(93,94) and may also aid in the navigation through the marrow parenchyma(95). These observations suggest that transendothelial migration of HSPC is a tightly regulated process that results from the confluence of multiple signaling and adhesive pathways(77). Whether this process is similar to that described for mature leukocytes, and what is the contribution of the specialized BM endothelium, remain unanswered questions(96).

REACHING THE MARROW

Are HSPC that cross the BM vasculature at the end of the road? The marrow space is not a homogenous tissue. As indicated at the beginning of this review, HSPC reside in very specific niches in close contact with specific cells (Fig. 1). It has been known for more than 30 years that pluripotent HSPC reside in or near the endosteal surface(97,98), but only recently have we learned of the remarkable capacity of HSPC to migrate to and engraft in these areas of the marrow [now defined as a distance of 12 or fewer cell diameters from the bone surface(99)] within just few hours of intravenous injection(99-101). These important observations indicated not only that HSPC can specifically home to the BM from the circulation, but that they can also search for a specific and appropriate location within a solid organ. Although the "rules of the road" must change when moving from a fluid to a solid milieu, it is likely that similar cues govern these final stages of homing.

A major problem in dissecting this process has been that the BM is a difficult organ to visualize. Pioneering studies by the group of Ulrich von Andrian, however, have benefited from multiphoton microscopy technology to image the dynamic movement of T lymphocytes within the BM parenchyma(102). These innovations have not yet been applied to the study of HSPC migration inside the BM, but prove that with the help of current technologies we may be able to understand this process with greater detail in the near future.

Receptors involved in intramedullary navigation include some that are also required during previous stages of homing (CXCR4 and β1 integrins), together with receptors highly specialized in guiding HSPC within the BM. A central role for the CXCL12/CXCR4 pathway in this process was suggested from studies showing that the hypoxic conditions of the endosteal region lead to CXCL12 expression, and favor the preferential tropism of HSPC to these regions of the marrow(103). Furthermore, inhibition or absence of Rac2 results in the inability of HSPC to engraft transplanted mice without affecting their capacity to enter marrow(84), suggesting that CXCR4 modulation of Rac2 activity is specifically required for intramedullary navigation. Modulation of CXCR4 signaling through other receptors (e.g., lipids, chemoattractants, nucleotides or neurotransmitters)(89-92) likely have an impact on HSPC movement inside the marrow as well, although this has not been formally demonstrated.

Evidence that β1 integrins participate in the navigation of HSPC within the BM parenchyma has been gained by the observation that specific inhibition of the α5 and α6 subunits (VLA-5 and VLA-6, respectively) results in reduced homing(104-106). While the ligand for VLA-5, fibronectin, is widely expressed, that of VLA-6, laminin, is restricted to the subendothelial basement membrane and BM parenchyma and absent from the endothelial surface(107). This suggests that VLA-6 function is restricted to intramedullary migration. In addition, blockade of α6 integrins in mice does not result in HSPC dislodgment from the BM(105). This contrasts with the effect of α4-and α5-integrin blocking reagents(106,108,109), and indicates that VLA-6 does not mediate interactions of HSPC with their niche.

Several receptors and matrix elements not involved in other stages of homing have recently been identified as critical in guiding the movement of HSPC within the marrow space. One of these is a calcium-sensing receptor (CaR) that can detect gradients of this cation. CaR is expressed on HSPC and guides its migration to the endosteal region, where Ca2+ levels are approximately 20 times higher than those found in the blood(110). Studies in CaR-deficient mice showed that this receptor was not required for homing into the marrow space, but rather to properly position HSPC near the bone surface, in the vicinity of osteoblasts(111). A similar role has been proposed for osteopontin (OPN), a matrix component produced by osteoblasts and deposited on the endosteal surface(112,113). Studies in OPN-deficient mice suggested that engagement of OPN by VLA-4 or CD44, two of its receptors on HSPC, contributes to the intramedullary navigation of HSPC towards the niche areas(112,113). Interestingly, another ligand for CD44, hyaluronan, was found to be expressed on the surface of HSPC and also affected the distribution of homed cells(114). Altogether, these observations suggest that the initial events of homing, i.e. interactions with the BM microvasculature and transendothelial migration, and the later events within the marrow are mediated, at least in part, by a different repertoire of receptors.

CUDDLING IN THE NICHE

As mentioned earlier, several niches for HSPC exist in the BM. HSPC can associate with osteoblasts (the endosteal niche)(3,4), endothelial cells (the vascular niche)(5) or reticular cells (CAR cells)(6). Which of these niches do HSPC chose? Although it is clear that they preferentially migrate near the endosteal surface, only a small fraction of them actually contact the bone interface(5,100). This would suggest that more than one type of niche can attract HSPC. If this is true, it remains to be determined whether this positioning is a stochastic process (i.e., HSPC stay in the first niche that they find) or rather that it depends on the status of each migrating HSPC (e.g., cell cycle or differentiation profile).

HSPC that successfully arrive at their niche can then establish interactions with cellular and extracellular elements that control their survival (SCF, CXCL12, osteopontin)(112,113,115,116), proliferation/quiescence (SCF, Angiopoietin-1, Ncadherin)(3,95,117), differentiation (Notch1)(118) or retention (VCAM-1, CXCL12)(6,60,109,119). The efficiency of HSPC engraftment in this niche is illustrated by the productive establishment of long-term hematopoiesis with daily release of billions of blood cells in virtually every transplanted recipient. Interestingly, the interactions of HSPC with their niche appears to be reversible since the disruption of either the CXCL12/CXCR4 or α4β1/VCAM-1 pathways in adult mice results in a dramatic release of HSPC from the BM into the circulation(6,70,71,120).

As HSPC differentiate into different lineages, it is believed that increasingly mature cells relocate closer to the vascular areas, ready to enter the circulation(98). How this movement occurs is largely unknown, with few exceptions: Megakaryocytic progenitors are often found in contact with BM vascular sinusoids where they continuously release platelets into circulation. Two chemokines, CXCL12 and fibroblast-growth-factor 4, were shown to mediate megakaryocyte recruitment to a vascular niche thus promoting platelet production(121). This observation exemplifies that different cues can guide the migration of distinct stem and progenitor cells to their respective niches within the BM.

REPAVING THE ROAD: STRATEGIES TO IMPROVE HSPC TRAFFICKING

Detailed characterization of the "rules of traffic" followed by HSPC as they migrate to their niches will be tremendously beneficial. One very direct application for hematologists is the possibility to manipulate HSPC homing to their advantage. In the context of BMT, a major goal is to improve the process of homing to allow for faster recovery of hematological parameters in transplanted patients. It might also allow to enhance the engraftment of genetically manipulated HSPC(122). One particularly promising application is to improve the homing of cord blood (CB)-derived HSPC. CB represents an unlimited source of HSPC for BMT(123), but because the amount of blood obtained from one placenta is small (~100-200 milliliters) and the number of HSPC collected low, its use has been limited to patients of low weight and to children(124,125).

All the steps identified during the homing process can potentially be targeted in order to enhance HSPC engraftment. I will briefly review now strategies developed over the last few years that attempted to manipulate molecules at various steps of the homing process, and that have attained varying degrees of success (Figure 1 and Table I).

Table I.

Manipulation of HSPC homing to the bone marrow

Code  Targeted process  Target molecule or cell  Strategy  Outcome 
Initial interactions  Selectin ligands  Fucosylation  Enhanced (Homing unaffected) 
Firm arrest  β1 integrins  Activation  Blocked 
Arrest & transmigration  CD26  Inhibition (diprotin A)  Enhanced 
Engraftment & expansion  Niche (osteoblasts)  rPTH  Enhanced 
Mobilization  Niche (osteoblasts)  β2-AR agonist (clenbuterol)  Enhanced 

Description and outcome of the different strategies devised to enhance HSPC homing. Code numbers correspond to the targeted steps identified in Figure 1. Details of these strategies are provided in the text. rPTH, recombinant rat parathyroid hormone; β2-AR, beta 2 adrenergic receptor.

Manipulation of the initial interactions of HSPC with the BM microvasculature

Because this process is mainly mediated by endothelial selectins, we and others attempted to enhance HSPC interactions with the vascular bed of the BM by enhancing the expression of selectin ligands on HSPC(126,127). Initial experiments using CD34+ cells derived from CB samples demonstrated that, compared to those derived from mobilized peripheral blood or BM, these cells initiate rolling on the BM microvasculature of NOD/SCID mice (a common murine model to study human hematopoiesis) with low efficiency(34). This reduction was due to the lack of expression of ligands for both P-and E-selectins on about one third of CB-derived CD34+ cells. Biochemical and gene expression analyses demonstrated that one type of enzyme involved in the synthesis of selectin ligands was markedly reduced in these cells. This enzyme, a fucosyltransferase (FuT), acts in concert with other glycosyltransferases to properly modify scaffold proteins (PSGL-1 and CD44) and to generate functional selectin ligands(127). To artificially induce selectin ligands on CB-derived CD34+ cells, a recombinant soluble form of FuT6 was added to the cell media, together with the appropriate co-factors and a fucose donor (GDP-fucose). After a brief incubation at 37°C, this treatment induced a robust expression of ligands for both P-and E-selectin in all CD34+ cells. In addition, treated cells could more efficiently roll on and adhere to the BM microvasculature of NOD/SCID mice in vivo(127). Thus it was surprising and disappointing that this did not result in an increased number of cells that homed inside the BM. Conflicting results from one group suggested, however, that long term engraftment could be enhanced by fucosylated CB-CD34+ cells(126). It is currently difficult to reconciliate both observations, which might be due to minor technical variations. One potential interpretation of why increased interactions with the vasculature did not translate in enhanced homing is that a different set of molecules required for homing (e.g., chemokine receptors or signaling proteins) act as a bottleneck for the entry of CB-HSPC inside the BM. Further research to unveil these limiting molecules will be important to get a full benefit from this strategy.

Enhancing the firm arrest of HSPC on the BM vasculature

β1 integrins are essential for the firm arrest of HSPC on the BM microvasculature(29). Because cultured HSPC displayed a reduction in β1 integrin-mediated binding, a strategy was devised to increase these interactions using a monoclonal antibody that locks β1 integrins in a high affinity state. Interestingly, this treatment resulted instead in the complete inhibition of HSPC homing and engraftment(128). In retrospect, this was perhaps an expected outcome. Integrin function in HSPC interacting with the BM microvasculature is controlled by CXCL12, and is characterized by a rapid and transient increase(28,29,65). This allows for strong binding to the vasculature after rolling, followed by release of these bonds to allow further movement on and across the endothelium. Locking β1 integrins in a high-affinity state, it seems, favored the initial firm interactions but prevented the subsequent migration of HSPC. This strategy provides an example of how a biological phenomenon must be carefully characterized before it can be successfully manipulated.

Modulating the CXCL12/CXCR4 axis

As evidenced by the studies described above, CXCL12-triggered signaling is essential for HSPC homing. It controls multiple steps of this process, including firm arrest, transendothelial migration and intramedullary navigation in search of niches in the BM. Thus, a great deal of effort has been put into finding ways to enhance CXCR4-mediated signaling in HSPC. For example, upregulation of CXCR4 expression on human CD34+ cells by cytokines (SCF and interleukin-6) resulted in enhanced migration towards CXCL12 in vitro and engraftment of NOD/SCID mice(52). Alternatively, treatment of human or murine HSPC with lipids or chemoattractants (S1P and C3a, respectively) potentiated the responses mediated by CXCR4 in vitro and enhanced engraftment in vivo(89,90). Likewise, treatment of murine or human HSPC with factors present in the serum of G-CSF-mobilized blood enhanced their responses to CXCL12 and homing by promoting CXCR4 incorporation into physiological signaling platforms (lipid rafts)(129).

A markedly different and elegant strategy was described by the group of Hal Broxmeyer. Their approach was based on the specific inhibition of CD26, a serine dipeptidylpeptidase (DPP-IV) expressed on the surface of hematopoietic cells. CD26 cleaves the N-terminal dipeptide from CXCL12, thus rendering it inactive(130). In the context of homing, CD26 present on the surface of rolling HSPC is thought to inactivate CXCL12 presented by the BM endothelium, thus reducing the levels of functional chemokine over time(130,131). In vitro studies demonstrated that absence or blockage of CD26 prevented CXCL12 inactivation; in vivo this translated into a remarkable enhancement of homing and engraftment of both murine and human HSPC(131,132). This approach is probably the most promising attempt to manipulate HSPC homing to date, and serves to illustrate that this is an attainable goal that may soon benefit the clinic.

Targeting the stem cell niche

At the end of the homing process, the interactions established between HSPC and their niche favors their longterm preservation and function. The group of David Scadden hypothesized that targeting the osteoblastic cells that constitute the endosteal niche might enhance the engraftment and survival of transplanted HSPC. They took advantage of their observation that expression of a constitutive active form of the parathyroid hormone (PTH)/PTH-related peptide receptor in transgenic mice, or treatment with exogenous PTH led to an expansion of the HSPC pool(4). They thus treated mice repeatedly exposed to chemotoxic agents (cyclophosphamide) or lethally irradiated mice after BM transplantation with exogenous PTH. This treatment resulted in protection of HSPC from cytotoxic damage as well as in an expansion of the transplanted HSPC(133). The potential benefits of this strategy in the clinic are enormous, not only in the context of BMT, but also in the outcome of leukemic patients undergoing chemotherapeutic regimes.

Enhancing the mobilization of HSPC

HSPC egress from the BM can be enhanced by treatment with a number of agents, a process termed mobilization, and is generally considered to be caused by disruption of retention signals in the BM niches(134). For this reason this process is envisioned as a reversal of the last steps of homing. Mobilization is a process of considerable clinical interest since the vast majority of autologous or allogeneic BMT procedures today use HSPC obtained from mobilized donors due to the increased number and superior engrafting capabilities of these cells compared to those from BM or CB units(135).

Although many of the cellular and molecular participants of this process are still unknown, most of the signaling and adhesive molecules so far involved in HSPC mobilization are also required for their homing(134). In particular, blockage of both the VLA-4/VCAM-1 and CXCL12/CXCR4 pathways either directly [with antibodies or antagonists(60,109,136,137)] or indirectly [by cytokine-induced downregulation of gene expression or protease-mediated degradation(59,61,119)] have been extensively studied as strategies to enhance HSPC mobilization. One example of the potential utility of such strategies is the case of AMD3100. This bicyclam is a designed CXCR4 antagonist(138) that induces HSPC mobilization by itself or by synergizing with G-CSF(137), and is currently being tested in several clinical trials.

A completely different approach to enhancing HSPC mobilization was provided recently by our group. We found evidence that signaling through the sympathetic nervous system (SNS) was required for G-CSF to downregulate niche activity and to trigger mobilization. Correspondingly, simple co-injection of a β2-adrenergic receptor agonist (clenbuterol) enhanced cytokine-induced mobilization(61). These findings suggest that pharmacological manipulation of the SNS can be clinically useful to increase HSPC mobilization (using adrenergic agonists) or, conversely, to enhance their engraftment in the BM niches (using adrenergic antagonists)(61,139).

FUTURE AVENUES

The observations accumulated during the study of the mechanisms of HSPC trafficking will have an impact in several areas of stem cell biology, most importantly perhaps in tissue regeneration and cancer: HSPC accumulate in areas of damage and appear to contribute to tissue repair(140). Likewise, BM-derived HSPC migrate to pre-metastatic lesions and may contribute to tumor spread(141). As in the case of homing to the BM, both of these processes seem to require the CXCR4/CXCL12 and VLA-4/VCAM-1/fibronectin

pathways(71,103,141). Given the striking similarity between all these processes, it is tempting to speculate that the trafficking of stem and progenitor cells in search of certain niches follow the same basic rules described here. Finding strategies to potentiate HSPC recruitment to ischemic or damaged tissues, or to block it when primary tumors are detected will be major challenges for the future.

Another area of great interest will be to understand whether the homing of pathogenic stem cells to "their niches" differs from that of normal stem cells, and if so how can we take advantage of those differences. This last point is prominently illustrated by the recent finding that chronic myelogenous leukemia-initiating cells (a kind of leukemic stem cell) rely on CD44 as a BM homing receptor more heavily than normal HSPC do, and that blockage of CD44 partially protects animals from this type of leukemia(142). As more leukemia and solid tumor types that rely on founding cancer stem cells are being described(143,144), it will become a priority to characterize their migratory patterns.

Since the pioneering description in 1956 of cells from a donor that could "in special circumstances, not merely survive and multiply in another, but even replace the corresponding cells of the host and take over their functions"(145), a long and exciting road of discoveries in the biology of hematopoietic stem cells was initiated that has led to enormous medical benefits and will continue to guide us to future challenges in the biomedical research of the twenty-first century.

CONFLICT OF INTEREST

The author declares no financial conflict of interest.

ACKNOWLEDGEMENTS

I want to express my gratitude to Linnea Weiss, Daniel Lucas-Alcaraz and Weiming Kao for helpful comments and corrections during the writing of this review. The author is supported in part by a Scientist Development Grant from the American Heart Association.

BIBLIOGRAFÍA
[1.]
C. Durand, E. Dzierzak.
Embryonic beginnings of adult hematopoietic stem cells.
Haematologica, 90 (2005), pp. 100-108
[2.]
G.B. Adams, D.T. Scadden.
The hematopoietic stem cell in its place.
Nat Immunol, 7 (2006), pp. 333-337
[3.]
J. Zhang, C. Niu, L. Ye, H. Huang, X. He, W.G. Tong, et al.
Identification of the haematopoietic stem cell niche and control of the niche size.
Nature, 425 (2003), pp. 836-841
[4.]
L.M. Calvi, G.B. Adams, K.W. Weibrecht, J.M. Weber, D.P. Olson, M.C. Knight, et al.
Osteoblastic cells regulate the haematopoietic stem cell niche.
Nature, 425 (2003), pp. 841-846
[5.]
M.J. Kiel, O.H. Yilmaz, T. Iwashita, C. Terhorst, S.J. Morrison.
SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells.
Cell, 121 (2005), pp. 1109-1121
[6.]
T. Sugiyama, H. Kohara, M. Noda, T. Nagasawa.
Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches.
Immunity, 25 (2006), pp. 977-988
[7.]
D.W. Barnes, J.F. Loutit.
Haemopoietic stem cells in the peripheral blood.
Lancet, 2 (1967), pp. 1138-1141
[8.]
J.W. Goodman, G.S. Hodgson.
Evidence for stem cells in the peripheral blood of mice.
Blood, 19 (1962), pp. 702-714
[9.]
J.L. Abkowitz, A.E. Robinson, S. Kale, M.W. Long, J. Chen.
Mobilization of hematopoietic stem cells during homeostasis and after cytokine exposure.
Blood, 102 (2003), pp. 1249-1253
[10.]
D.E. Wright, A.J. Wagers, A.P. Gulati, F.L. Johnson, I.L. Weissman.
Physiological migration of hematopoietic stem and progenitor cells.
Science, 294 (2001), pp. 1933-1936
[11.]
E. Lorenz, D. Uphoff, T.R. Reid, E. Shelton.
Modification of irradiation injury in mice and guinea pigs by bone marrow injections.
J Natl Cancer Inst, 12 (1951), pp. 197-201
[12.]
L.O. Jacobson, E.K. Marks, M.J. Robson, E. Gaston, R.E. Zirkle.
The effect of spleen protection on mortality following X-radiation.
J Lab Clin Med, 34 (1949), pp. 1538-1543
[13.]
E.D. Thomas.
Bone marrow transplantation: a review.
Semin Hematol, 36 (1999), pp. 95-103
[14.]
M.A. Cabrera-Salazar, E. Novelli, J.A. Barranger.
Gene therapy for the lysosomal storage disorders.
Curr Opin Mol Ther, 4 (2002), pp. 349-358
[15.]
C. Antoine, S. Müller, A. Cant, M. Cavazzana-Calvo, P. Veys, J. Vossen, A. Fasth, et al.
Long-term survival and transplantation of haemopoietic stem cells for immunodeficiencies: Report of the European experience 1968-99.
Lancet, 361 (2003), pp. 553-560
[16.]
T.A. Springer.
Traffic signals for lymphocyte recirculation and leukocyte emigration: The multistep paradigm.
Cell, 76 (1994), pp. 301-314
[17.]
R.P. McEver.
Selectin-carbohydrate interactions during inflammation and metastasis.
Glycoconj J, 14 (1997), pp. 585-591
[18.]
J.B. Lowe.
Glycosylation in the control of selectin counter-receptor structure and function.
Immunol Rev, 186 (2002), pp. 19-36
[19.]
K.L. Moore, K.D. Patel, R.E. Bruehl, F. Li, D.A. Johnson, H.S. Lichenstein, et al.
P-selectin glycoprotein ligand-1 mediates rolling of human neutrophils on P-selectin.
J Cell Biol, 128 (1995), pp. 661-671
[20.]
L. Xia, M. Sperandio, T. Yago, J.M. McDaniel, R.D. Cummings, S. Pearson-White, et al.
P-selectin glycoprotein ligand-1-deficient mice have impaired leukocyte tethering to E-selectin under flow.
J Clin Invest, 109 (2002), pp. 939-950
[21.]
T. Hirata, G. Merrill-Skoloff, M. Aab, J. Yang, B.C. Furie, B. Furie.
P-Selectin glycoprotein ligand 1 (PSGL-1) is a physiological ligand for E-selectin in mediating T helper 1 lymphocyte migration.
J Exp Med, 192 (2000), pp. 1669-1676
[22.]
R.C. Fuhlbrigge, S.L. King, R. Sackstein, T.S. Kupper.
CD43 is a ligand for E-selectin on CLA+ human T cells.
Blood, 107 (2006), pp. 1421-1426
[23.]
M. Matsumoto, K. Atarashi, E. Umemoto, Y. Furukawa, A. Shigeta, M. Miyasaka, T. Hirata.
CD43 functions as a ligand for E-Selectin on activated T cells.
J Immunol, 175 (2005), pp. 8042-8050
[24.]
Y. Katayama, A. Hidalgo, J. Chang, A. Peired, P.S. Frenette.
CD44 is a physiological E-selectin ligand on neutrophils.
J Exp Med, 201 (2005), pp. 1183-1189
[25.]
A. Levinovitz, J. Muhlhoff, S. Isenmann, D. Vestweber.
Identification of a glycoprotein ligand for E-selectin on mouse myeloid cells.
J Cell Biol, 121 (1993), pp. 449-459
[26.]
A. Hidalgo, A.J. Peired, M.K. Wild, D. Vestweber, P.S. Frenette.
Complete identification of E-selectin ligands on neutrophils reveals distinct functions of PSGL-1 ESL-1, and CD44.
Immunity, 26 (2007), pp. 477-489
[27.]
C. Laudanna, R. Alon.
Right on the spot. Chemokine triggering of integrin-mediated arrest of rolling leukocytes.
Thromb Haemost, 95 (2006), pp. 5-11
[28.]
A. Peled, O. Kollet, T. Ponomaryov, I. Petit, S. Franitza, V. Grabovsky, 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, 95 (2000), pp. 3289-3296
[29.]
A. Peled, V. Grabovsky, L. Habler, J. Sandbank, F. Arenzana-Seisdedos, I. Petit, et al.
The chemokine SDF-1 stimulates integrin-mediated arrest of CD34+ cells on vascular endothelium under shear flow.
J Clin Invest, 104 (1999), pp. 1199-1211
[30.]
A.C. Zannettino, M.C. Berndt, C. Butcher, E.C. Butcher, M.A. Vadas, P.J. Simmons.
Primitive human hematopoietic progenitors adhere to P-selectin (CD62P).
Blood, 85 (1995), pp. 3466-3477
[31.]
J.B. Tracey, H.M. Rinder.
Characterization of the P-selectin ligand on human hematopoietic progenitors.
Exp Hematol, 24 (1996), pp. 1494-1500
[32.]
I.B. Mazo, J.C. Gutierrez-Ramos, P.S. Frenette, R.O. Hynes, D.D. Wagner, U.H. von Andrian.
Hematopoietic progenitor cell rolling in bone marrow microvessels: Parallel contributions by endothelial selectins and VCAM-1.
J Exp Med, 188 (1998), pp. 465-474
[33.]
P.S. Frenette, S. Subbarao, I.B. Mazo, U.H. von Andrian, D.D. Wagner.
Endothelial selectins and vascular cell adhesion molecule-1 promote hematopoietic progenitor homing to bone marrow.
Proc Natl Acad Sci USA, 95 (1998), pp. 14423-14428
[34.]
A. Hidalgo, L.A. Weiss, P.S. Frenette.
Functional selectin ligands mediating human CD34+ cell interactions with bone marrow endothelium are enhanced postnatally.
J Clin Invest, 110 (2002), pp. 559-569
[35.]
I.B. Mazo, E.J. Quackenbush, J.B. Lowe, U.H. von Andrian.
Total body irradiation causes profound changes in endothelial traffic molecules for hematopoietic progenitor cell recruitment to bone marrow.
Blood, 99 (2002), pp. 4182-4191
[36.]
K.M. Schweitzer, A.M. Dräger, P. van der Valk, S.F. Thijsen, A. Zevenbergen, A.P. Theijsmeijer, et al.
Constitutive expression of E-selectin and vascular cell adhesion molecule-1 on endothelial cells of hematopoietic tissues.
Am J Pathol, 148 (1996), pp. 165-175
[37.]
D.A. Sipkins, X. Wei, J.W. Wu, J.M. Runnels, D. Côté, T.K. Means, et al.
In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment.
Nature, 435 (2005), pp. 969-973
[38.]
Y. Katayama, A. Hidalgo, B.C. Furie, D. Vestweber, B. Furie, P.S. Frenette.
PSGL-1 participates in E-selectin-mediated progenitor homing to bone marrow: Evidence for cooperation between E-selectin ligands and α4 integrin.
Blood, 102 (2003), pp. 2060-2067
[39.]
U. Jung, K.E. Norman, K. Scharffetter-Kochanek, A.L. Beaudet, K. Ley.
Transit time of leukocytes rolling through venules controls cytokine-induced inflammatory cell recruitment in vivo.
J Clin Invest, 102 (1998), pp. 1526-1533
[40.]
C.J. Dimitroff, J.Y. Lee, S. Rafii, R.C. Fuhlbrigge, R. Sackstein.
CD44 is a major E-selectin ligand on human hematopoietic progenitor cells.
J Cell Biol, 153 (2001), pp. 1277-1286
[41.]
K. Miyake, C.B. Underhill, J. Lesley, P.W. Kincade.
Hyaluronate can function as a cell adhesion molecule and CD44 participates in hyaluronate recognition.
J Exp Med, 172 (1990), pp. 69-75
[42.]
H.C. DeGrendele, P. Estess, M.H. Siegelman.
Requirement for CD44 in activated T cell extravasation into an inflammatory site.
Science, 278 (1997), pp. 672-675
[43.]
A. Avigdor, P. Goichberg, S. Shivtiel, A. Dar, A. Peled, S. Samira, et al.
CD44 and hyaluronic acid cooperate with SDF-1 in the trafficking of human CD34+ stem/progenitor cells to bone marrow.
Blood, 103 (2004), pp. 2981-2989
[44.]
C.E. Green, U.Y. Schaff, M.R. Sarantos, A.F. Lum, D.E. Staunton, S.I. Simon.
Dynamic shifts in LFA-1 affinity regulate neutrophil rolling, arrest, and transmigration on inflamed endothelium.
Blood, 107 (2006), pp. 2101-2111
[45.]
B.C. Chesnutt, D.F. Smith, N.A. Raffler, M.L. Smith, E.J. White, K. Ley.
Induction of LFA-1-dependent neutrophil rolling on ICAM-1 by engagement of E-selectin.
Microcirculation, 13 (2006), pp. 99-109
[46.]
J. Teixido, M.E. Hemler, J.S. Greenberger, P. Anklesaria.
Role of beta 1 and beta 2 integrins in the adhesion of human CD34hi stem cells to bone marrow stroma.
J Clin Invest, 90 (1992), pp. 358-367
[47.]
D.A. Williams, M. Rios, C. Stephens, V.P. Patel.
Fibronectin and VLA-4 in haematopoietic stem cell-microenvironment interactions.
Nature, 352 (1991), pp. 438-441
[48.]
Y. Katayama, A. Hidalgo, A. Peired, P.S. Frenette.
Integrin α4β7 and its counterreceptor MAdCAM-1 contribute to hematopoietic progenitor recruitment into bone marrow following transplantation.
Blood, 104 (2004), pp. 2020-2026
[49.]
C. Berlin, E.L. Berg, M.J. Briskin, D.P. Andrew, P.J. Kilshaw, B. Holzmann, et al.
Alpha 4 beta 7 integrin mediates lymphocyte binding to the mucosal vascular addressin MAdCAM-1.
Cell, 74 (1993), pp. 185-195
[50.]
T. Nagasawa, A. chemokine.
SDF-1/PBSF, and its receptor, CXC chemokine receptor 4, as mediators of hematopoiesis.
Int J Hematol, 72 (2000), pp. 408-411
[51.]
A. Aiuti, I.J. Webb, C. Bleul, T. Springer, J.C. Gutierrez-Ramos.
The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood.
J Exp Med, 185 (1997), pp. 111-120
[52.]
A. Peled, I. Petit, O. Kollet, M. Magid, T. Ponomaryov, T. Byk, et al.
Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4.
Science, 283 (1999), pp. 845-848
[53.]
D.E. Wright, E.P. Bowman, A.J. Wagers, E.C. Butcher, I.L. Weissman.
Hematopoietic stem cells are uniquely selective in their migratory response to chemokines.
J Exp Med, 195 (2002), pp. 1145-1154
[54.]
T. Nagasawa, T. Nakajima, K. Tachibana, H. Iizasa, C.C. Bleul, O. Yoshie, et al.
Molecular cloning and characterization of a murine pre-B-cell growth-stimulating factor/stromal cell-derived factor 1 receptor, a murine homolog of the human immunodeficiency virus 1 entry coreceptor fusin.
Proc Natl Acad Sci USA, 93 (1996), pp. 14726-14729
[55.]
T. Nagasawa, S. Hirota, K. Tachibana, N. Takakura, S. Nishikawa, Y. Kitamura, et al.
Defects of B-cell lymphopoiesis and bonemarrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1.
Nature, 382 (1996), pp. 635-638
[56.]
Y.R. Zou, A.H. Kottmann, M. Kuroda, I. Taniuchi, D.R. Littman.
Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development.
Nature, 393 (1998), pp. 595-599
[57.]
T. Nagasawa.
The chemokine CXCL12 and regulation of HSC and B lymphocyte development in the bone marrow niche.
Adv Exp Med Biol, 602 (2007), pp. 69-75
[58.]
A. Dar, O. Kollet, T. Lapidot.
Mutual, reciprocal SDF-1/CXCR4 interactions between hematopoietic and bone marrow stromal cells regulate human stem cell migration and development in NOD/SCID chimeric mice.
Exp Hematol, 34 (2006), pp. 967-975
[59.]
C.L. Semerad, M.J. Christopher, F. Liu, B. Short, P.J. Simmons, I. Winkler, et al.
G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow.
Blood, 106 (2005), pp. 3020-3027
[60.]
I. Petit, M. Szyper-Kravitz, A. Nagler, M. Lahav, A. Peled, L. Habler, et al.
G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4.
Nat Immunol, 3 (2002), pp. 687-694
[61.]
Y. Katayama, M. Battista, W.M. Kao, A. Hidalgo, A.J. Peired, S.A. Thomas, P.S. Frenette.
Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow.
[62.]
H. Bonig, G.V. Priestley, L.M. Nilsson, Y. Jiang, T. Papayannopoulou.
PTX-sensitive signals in bone marrow homing of fetal and adult hematopoietic progenitor cells.
Blood, 104 (2004), pp. 2299-2306
[63.]
Q. Ma, D. Jones, P.R. Borghesani, R.A. Segal, T. Nagasawa, T. Kishimoto, et al.
Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice.
Proc Natl Acad Sci USA, 95 (1998), pp. 9448-9453
[64.]
M.J. Brown, R. Nijhara, J.A. Hallam, M. Gignac, K.M. Yamada, S.L. Erlandsen, et al.
Chemokine stimulation of human peripheral blood T lymphocytes induces rapid dephosphorylation of ERM proteins, which facilitates loss of microvilli and polarization.
Blood, 102 (2003), pp. 3890-3899
[65.]
A. Hidalgo, F. Sanz-Rodríguez, J.L. Rodríguez-Fernández, B. Albella, C. Blaya, N. Wright, et al.
Chemokine stromal cell-derived factor-1α modulates VLA-4 integrin-dependent adhesion to fibronectin and VCAM-1 on bone marrow hematopoietic progenitor cells.
Exp Hematol, 29 (2001), pp. 345-355
[66.]
A. Dar, P. Goichberg, V. Shinder, A. Kalinkovich, O. Kollet, N. Netzer, et al.
Chemokine receptor CXCR4-dependent internalization and resecretion of functional chemokine SDF-1 by bone marrow endothelial and stromal cells.
Nat Immunol, 6 (2005), pp. 1038-1046
[67.]
E. Hirsch, A. Iglesias, A.J. Potocnik, U. Hartmann, R. Fassler.
Impaired migration but not differentiation of haematopoietic stem cells in the absence of ‚1 integrins.
Nature, 380 (1996), pp. 171-175
[68.]
A.J. Potocnik, C. Brakebusch, R. Fassler.
Fetal and adult hematopoietic stem cells require ‚1 integrin function for colonizing fetal liver, spleen, and bone marrow.
Immunity, 12 (2000), pp. 653-663
[69.]
A.G. Arroyo, J.T. Yang, H. Rayburn, R.O. Hynes.
Differential requirements for α4 integrins during fetal and adult hematopoiesis.
Cell, 85 (1996), pp. 997-1008
[70.]
L.M. Scott, G.V. Priestley, T. Papayannopoulou.
Deletion of α4 integrins from adult hematopoietic cells reveals roles in homeostasis, regeneration, and homing.
Mol Cell Biol, 23 (2003), pp. 9349-9360
[71.]
G.V. Priestley, T. Ulyanova, T. Papayannopoulou.
Sustained alterations in biodistribution of stem/progenitor cells in Tie2Cre+ α4(f/f) mice are hematopoietic cell autonomous.
[72.]
C.V. Carman, P.T. Sage, T.E. Sciuto, M.A. de la Fuente, R.S. Geha, H.D. Ochs, et al.
Transcellular diapedesis is initiated by invasive podosomes.
Immunity, 26 (2007), pp. 784-797
[73.]
A.R. Schenkel, Z. Mamdouh, W.A. Muller.
Locomotion of monocytes on endothelium is a critical step during extravasation.
Nat Immunol, 5 (2004), pp. 393-400
[74.]
O. Barreiro, M. Yanez-Mo, J.M. Serrador, M.C. Montoya, M. Vicente-Manzanares, R. Tejedor, et al.
Dynamic interaction of VCAM-1 and ICAM-1 with moesin and ezrin in a novel endothelial docking structure for adherent leukocytes.
J Cell Biol, 157 (2002), pp. 1233-1245
[75.]
C.V. Carman, T.A. Springer.
A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them.
J Cell Biol, 167 (2004), pp. 377-388
[76.]
W.A. Muller.
Leukocyte-endothelial-cell interactions in leukocyte transmigration and the inflammatory response.
Trends Immunol, 24 (2003), pp. 327-334
[77.]
D. Vestweber.
Adhesion and signaling molecules controlling the transmigration of leukocytes through endothelium.
Immunol Rev, 218 (2007), pp. 178-196
[78.]
J.D. van Buul, C. Voermans, V. van den Berg, E.C. Anthony, F.P. Mul, S. van Wetering, et al.
Migration of human hematopoietic progenitor cells across bone marrow endothelium is regulated by vascular endothelial cadherin.
J Immunol, 168 (2002), pp. 588-596
[79.]
C. Voermans, P.M. Rood, P.L. Hordijk, W.R. Gerritsen, C.E. van der Schoot.
Adhesion molecules involved in transendothelial migration of human hematopoietic progenitor cells.
Stem Cells, 18 (2000), pp. 435-443
[80.]
A.M. Imbert, G. Belaaloui, F. Bardin, C. Tonnelle, M. Lopez, C. Chabannon.
CD99 expressed on human mobilized peripheral blood CD34+ cells is involved in transendothelial migration.
Blood, 108 (2006), pp. 2578-2586
[81.]
L. Reibel, O. Dorseuil, R. Stancou, J. Bertoglio, G. Gacon.
A hemopoietic specific gene encoding a small GTP binding protein is overexpressed during T cell activation.
Biochem Biophys Res Commun, 175 (1991), pp. 451-458
[82.]
J.A. Cancelas, M. Jansen, D.A. Williams.
The role of chemokine activation of Rac GTPases in hematopoietic stem cell marrow homing, retention, and peripheral mobilization.
Exp Hematol, 34 (2006), pp. 976-985
[83.]
Y. Gu, M.D. Filippi, J.A. Cancelas, J.E. Siefring, E.P. Williams, A.C. Jasti, et al.
Hematopoietic cell regulation by Rac1 and Rac2 guanosine triphosphatases.
Science, 302 (2003), pp. 445-449
[84.]
J.A. Cancelas, A.W. Lee, R. Prabhakar, K.F. Stringer, Y. Zheng, D.A. Williams.
Rac GTPases differentially integrate signals regulating hematopoietic stem cell localization.
Nat Med, 11 (2005), pp. 886-891
[85.]
D. García-Bernal, N. Wright, E. Sotillo-Mallo, C. Nombela-Arrieta, J.V. Stein, X.R. Bustelo, J. Teixidó.
Vav1 and Rac control chemokinepromoted T lymphocyte adhesion mediated by the integrin α4β1.
Mol Biol Cell, 16 (2005), pp. 3223-3235
[86.]
A.D. Whetton, Y. Lu, A. Pierce, L. Carney, E. Spooncer.
Lysophospholipids synergistically promote primitive hematopoietic cell chemotaxis via a mechanism involving Vav 1.
Blood, 102 (2003), pp. 2798-2802
[87.]
I. Petit, P. Goichberg, A. Spiegel, A. Peled, C. Brodie, R. Seger, et al.
Atypical PKC-z regulates SDF-1-mediated migration and development of human CD34+ progenitor cells.
J Clin Invest, 115 (2005), pp. 168-176
[88.]
P. Goichberg, A. Kalinkovich, N. Borodovsky, M. Tesio, I. Petit, A. Nagler, et al.
cAMP-induced PKC-z activation increases functional CXCR4 expression on human CD34+ hematopoietic progenitors.
Blood, 107 (2006), pp. 870-879
[89.]
G. Seitz, A.M. Boehmler, L. Kanz, R. Mohle.
The role of sphingosine 1-phosphate receptors in the trafficking of hematopoietic progenitor cells.
Ann N Y Acad Sci, 1044 (2005), pp. 84-89
[90.]
R. Reca, D. Mastellos, M. Majka, L. Marquez, J. Ratajczak, S. Franchini, et al.
Functional receptor for C3a anaphylatoxin is expressed by normal hematopoietic stem/progenitor cells, and C3a enhances their homing-related responses to SDF-1.
Blood, 101 (2003), pp. 3784-3793
[91.]
L. Rossi, R. Manfredini, F. Bertolini, D. Ferrari, M. Fogli, R. Zini, et al.
The extracellular nucleotide UTP is a potent inducer of hematopoietic stem cell migration.
[92.]
A. Spiegel, S. Shivtiel, A. Kalinkovich, A. Ludin, N. Netzer, P. Goichberg, et al.
Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling.
Nat Immunol, 8 (2007), pp. 1123-1131
[93.]
A. Janowska-Wieczorek, L.A. Marquez, A. Dobrowsky, M.Z. Ratajczak, M.L. Cabuhat.
Differential MMP and TIMP production by human marrow and peripheral blood CD34+ cells in response to chemokines.
Exp Hematol, 28 (2000), pp. 1274-1285
[94.]
A. Janowska-Wieczorek, L.A. Marquez, J.M. Nabholtz, M.L. Cabuhat, J. Montaño, et al.
Growth factors and cytokines upregulate gelatinase expression in bone marrow CD34+ cells and their transmigration through reconstituted basement membrane.
Blood, 93 (1999), pp. 3379-3390
[95.]
B. Heissig, K. Hattori, S. Dias, M. Friedrich, B. Ferris, N.R. Hackett, et al.
Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand.
Cell, 109 (2002), pp. 625-637
[96.]
A. Hidalgo, P.S. Frenette.
Leukocyte podosomes sense their way through the endothelium.
Immunity, 26 (2007), pp. 753-755
[97.]
J.K. Gong.
Endosteal marrow: a rich source of hematopoietic stem cells.
Science, 199 (1978), pp. 1443-1445
[98.]
B.I. Lord, N.G. Testa, J.H. Hendry.
The relative spatial distributions of CFUs and CFUc in the normal mouse femur.
Blood, 46 (1975), pp. 65-72
[99.]
S.K. Nilsson, M.S. Dooner, C.Y. Tiarks, U. Weier, P.J. Quesenberry.
Potential and distribution of transplanted hematopoietic stem cells in a nonablated mouse model.
Blood, 89 (1997), pp. 4013-4020
[100.]
S.K. Nilsson, H.M. Johnston, J.A. Coverdale.
Spatial localization of transplanted hemopoietic stem cells: inferences for the localization of stem cell niches.
Blood, 97 (2001), pp. 2293-2299
[101.]
O. Kollet, A. Spiegel, A. Peled, I. Petit, T. Byk, R. Hershkoviz, et al.
Rapid and efficient homing of human CD34+CD38/low CXCR4+ stem and progenitor cells to the bone marrow and spleen of NOD/SCID and NOD/SCID/B2mnull mice.
Blood, 97 (2001), pp. 3283-3291
[102.]
I.B. Mazo, M. Honczarenko, H. Leung, L.L. Cavanagh, R. Bonasio, W. Weninger, et al.
Bone marrow is a major reservoir and site of recruitment for central memory CD8+ T cells.
Immunity, 22 (2005), pp. 259-270
[103.]
D.J. Ceradini, A.R. Kulkarni, M.J. Callaghan, O.M. Tepper, N. Bastidas, M.E. Kleinman, et al.
Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1.
Nat Med, 10 (2004), pp. 858-864
[104.]
N. Asaumi, E. Omoto, N. Mahmut, Y. Katayama, K. Takeda, K. Shinagawa, M. Harada.
Very late antigen-5 and leukocyte function-associated antigen-1 are critical for early stage hematopoietic progenitor cell homing.
Ann Hematol, 80 (2001), pp. 387-392
[105.]
H. Qian, K. Tryggvason, S.E. Jacobsen, M. Ekblom.
Contribution of α6 integrins to hematopoietic stem and progenitor cell homing to bone marrow and collaboration with α4 integrins.
Blood, 107 (2006), pp. 3503-3510
[106.]
J.C. van der Loo, X. Xiao, D. McMillin, K. Hashino, I. Kato, D.A. Williams.
VLA-5 is expressed by mouse and human long-term repopulating hematopoietic cells and mediates adhesion to extracellular matrix protein fibronectin.
J Clin Invest, 102 (1998), pp. 1051-1061
[107.]
Y. Gu, L. Sorokin, M. Durbeej, T. Hjalt, J.I. Jönsson, M. Ekblom.
Characterization of bone marrow laminins and identification of α5-containing laminins as adhesive proteins for multipotent hematopoietic FDCP-Mix cells.
Blood, 93 (1999), pp. 2533-2542
[108.]
T. Papayannopoulou, B. Nakamoto.
Peripheralization of hemopoietic progenitors in primates treated with anti-VLA4 integrin.
Proc Natl Acad Sci USA, 90 (1993), pp. 9374-9378
[109.]
T. Papayannopoulou, C. Craddock, B. Nakamoto, G.V. Priestley, N.S. Wolf.
The VLA4/VCAM-1 adhesion pathway defines contrasting mechanisms of lodgement of transplanted murine hemopoietic progenitors between bone marrow and spleen.
Proc Natl Acad Sci USA, 92 (1995), pp. 9647-9651
[110.]
M.G. House, L. Kohlmeier, N. Chattopadhyay, O. Kifor, T. Yamaguchi, M.S. Leboff, et al.
Expression of an extracellular calcium-sensing receptor in human and mouse bone marrow cells.
J Bone Miner Res, 12 (1997), pp. 1959-1970
[111.]
G.B. Adams, K.T. Chabner, I.R. Alley, D.P. Olson, Z.M. Szczepiorkowski, M.C. Poznansky, et al.
Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor.
Nature, 439 (2006), pp. 599-603
[112.]
S. Stier, Y. Ko, R. Forkert, C. Lutz, T. Neuhaus, E. Grünewald, et al.
Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size.
J Exp Med, 201 (2005), pp. 1781-1791
[113.]
S.K. Nilsson, H.M. Johnston, G.A. Whitty, B. Williams, R.J. Webb, D.T. Denhardt, et al.
Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells.
Blood, 106 (2005), pp. 1232-1239
[114.]
S.K. Nilsson, D.N. Haylock, H.M. Johnston, T. Occhiodoro, T.J. Brown, P.J. Simmons.
Hyaluronan is synthesized by primitive hemopoietic cells, participates in their lodgment at the endosteum following transplantation, and is involved in the regulation of their proliferation and differentiation in vitro.
Blood, 101 (2003), pp. 856-862
[115.]
R.L. Driessen, H.M. Johnston, S.K. Nilsson.
Membrane-bound stem cell factor is a key regulator in the initial lodgment of stem cells within the endosteal marrow region.
Exp Hematol, 31 (2003), pp. 1284-1291
[116.]
J.J. Lataillade, D. Clay, P. Bourin, F. Hérodin, C. Dupuy, C. Jasmin, et al.
Stromal cell-derived factor 1 regulates primitive hematopoiesis by suppressing apoptosis and by promoting G(0)/G(1) transition in CD34+ cells: Evidence for an autocrine/paracrine mechanism.
Blood, 99 (2002), pp. 1117-1129
[117.]
F. Arai, A. Hirao, M. Ohmura, H. Sato, S. Matsuoka, K. Takubo.
Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche.
[118.]
S. Stier, T. Cheng, D. Dombkowski, N. Carlesso, D.T. Scadden.
Notch1 activation increases hematopoietic stem cell self-renewal in vivo and favors lymphoid over myeloid lineage outcome.
Blood, 99 (2002), pp. 2369-2378
[119.]
J.P. Lévesque, J. Hendy, Y. Takamatsu, B. Williams, I.G. Winkler, P.J. Simmons.
Mobilization by either cyclophosphamide or granulocyte colony-stimulating factor transforms the bone marrow into a highly proteolytic environment.
Exp Hematol, 30 (2002), pp. 440-449
[120.]
T. Ulyanova, G.V. Priestley, B. Nakamoto, Y. Jiang, T. Papayannopoulou.
VCAM-1 ablation in nonhematopoietic cells in MxCre+ VCAM-1f/f mice is variable and dictates their phenotype.
Exp Hematol, 35 (2007), pp. 565-571
[121.]
S.T. Avecilla, K. Hattori, B. Heissig, R. Tejada, F. Liao, K. Shido, et al.
Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis.
Nat Med, 10 (2004), pp. 64-71
[122.]
H.B. Gaspar, K.L. Parsley, S. Howe, D. King, K.C. Gilmour, J. Sinclair, et al.
Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector.
Lancet, 364 (2004), pp. 2181-2187
[123.]
V. Rocha, R.F. Franco, R. Porcher, H. Bittencourt, W.A. Silva Jr, A. Latouche, et al.
Comparison of outcomes of unrelated bone marrow and umbilical cord blood transplants in children with acute leukemia.
Blood, 97 (2001), pp. 2962-2971
[124.]
Y. Cohen, A. Nagler.
Umbilical cord blood transplantation--how, when and for whom?.
Blood Rev, 18 (2004), pp. 167-179
[125.]
P. Rubinstein, C. Carrier, A. Scaradavou, J. Kurtzberg, J. Adamson, A.R. Migliaccio, et al.
Outcomes among 562 recipients of placentalblood transplants from unrelated donors.
N Engl J Med, 339 (1998), pp. 1565-1577
[126.]
L. Xia, J.M. McDaniel, T. Yago, A. Doeden, R.P. McEver.
Surface fucosylation of human cord blood cells augments binding to P-selectin and E-selectin and enhances engraftment in bone marrow.
Blood, 104 (2004), pp. 3091-3096
[127.]
A. Hidalgo, P.S. Frenette.
Enforced fucosylation of neonatal CD34+ cells generates selectin ligands that enhance the initial interactions with microvessels but not homing to bone marrow.
Blood, 105 (2005), pp. 567-575
[128.]
M. Ramírez, J.C. Segovia, I. Benet, C. Arbona, G. Güenechea, C. Blaya, et al.
Ex vivo expansion of umbilical cord blood (UCB) CD34+ cells alters the expression and function of α4β‚1 and α5β1 integrins.
Br J Haematol, 115 (2001), pp. 213-221
[129.]
M. Wysoczynski, R. Reca, J. Ratajczak, M. Kucia, N. Shirvaikar, M. Honczarenko, et al.
Incorporation of CXCR4 into membrane lipid rafts primes homing-related responses of hematopoietic stem/progenitor cells to an SDF-1 gradient.
[130.]
K.W. Christopherson 2nd, G. Hangoc, H.E. Broxmeyer.
Cell surface peptidase CD26/dipeptidylpeptidase IV regulates CXCL12/stromal cell-derived factor-1 alpha-mediated chemotaxis of human cord blood CD34+ progenitor cells.
J Immunol, 169 (2002), pp. 7000-7008
[131.]
T.B. Campbell, G. Hangoc, Y. Liu, K. Pollok, H.E. Broxmeyer.
Inhibition of CD26 in human cord blood CD34+ cells enhances their engraftment of nonobese diabetic/severe combined immunodeficiency mice.
Stem Cells Dev, 16 (2007), pp. 347-354
[132.]
K.W. Christopherson 2nd, G. Hangoc, C.R. Mantel, H.E. Broxmeyer.
Modulation of hematopoietic stem cell homing and engraftment by CD26.
Science, 305 (2004), pp. 1000-1003
[133.]
G.B. Adams, R.P. Martin, I.R. Alley, K.T. Chabner, K.S. Cohen, L.M. Calvi, et al.
Therapeutic targeting of a stem cell niche.
Nat Biotechnol, 25 (2007), pp. 238-243
[134.]
I.G. Winkler, J.P. Levesque.
Mechanisms of hematopoietic stem cell mobilization: when innate immunity assails the cells that make blood and bone.
Exp Hematol, 34 (2006), pp. 996-1009
[135.]
M. Korbling, P. Anderlini.
Peripheral blood stem cell versus bone marrow allotransplantation: does the source of hematopoietic stem cells matter?.
Blood, 98 (2001), pp. 2900-2908
[136.]
W.C. Liles, H.E. Broxmeyer, E. Rodger, B. Wood, K. Hübel, S. Cooper, et al.
Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist.
Blood, 102 (2003), pp. 2728-2730
[137.]
H.E. Broxmeyer, E. Grossbard, N. Jacobsen, M.A. Moore.
Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist.
J Exp Med, 201 (2005), pp. 1307-1318
[138.]
C.W. Hendrix, C. Flexner, R.T. MacFarland, C. Giandomenico, E.J. Fuchs, E. Redpath, et al.
Pharmacokinetics and safety of AMD-3100, a novel antagonist of the CXCR-4 chemokine receptor, in human volunteers.
Antimicrob Agents Chemother, 44 (2000), pp. 1667-1673
[139.]
J. Larsson, D. Scadden.
Nervous activity in a stem cell niche.
[140.]
D.J. Ceradini, G.C. Gurtner.
Homing to hypoxia: HIF-1 as a mediator of progenitor cell recruitment to injured tissue.
Trends Cardiovasc Med, 15 (2005), pp. 57-63
[141.]
R.N. Kaplan, R.D. Riba, S. Zacharoulis, A.H. Bramley, L. Vincent, C. Costa, et al.
VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche.
Nature, 438 (2005), pp. 820-827
[142.]
D.S. Krause, K. Lazarides, U.H. von Andrian, R.A. Van Etten.
Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells.
Nat Med, 12 (2006), pp. 1175-1180
[143.]
C.A. O’Brien, A. Pollett, S. Gallinger, J.E. Dick.
A human colon cancer cell capable of initiating tumour growth in immunodeficient mice.
Nature, 445 (2007), pp. 106-110
[144.]
J.C. Wang, J.E. Dick.
Cancer stem cells: lessons from leukemia.
Trends Cell Biol, 15 (2005), pp. 494-501
[145.]
C.E. Ford, J.L. Hamerton, D.W. Barnes, J.F. Loutit.
Cytological identification of radiation-chimaeras.
Nature, 177 (1956), pp. 452-454
Copyright © 2008. Sociedad Española de Inmunología
Opciones de artículo