首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Transplantation with bone marrow (BM) hematopoietic stem cells (HSC) has been used for curative therapy of hematologic diseases and inborn errors of metabolism for decades. More recently, alternative sources of HSC, particularly those induced to exit marrow and traffic to peripheral blood in response to external stimuli, have become the most widely used hematopoietic graft and show significant superiority to marrow HSC. Although a variety of agents can mobilize stem cells with different kinetics and efficiencies and these agents can be additive or synergistic when used in combination, currently G-CSF is the predominant stem cell mobilizer used clinically based upon potency, predictability and safety. Recent studies have demonstrated that the interaction between the chemokine stromal-derived factor 1 (SDF-1/CXCL12) and its receptor CXCR4 serves as a key regulator of HSC trafficking. AMD3100, a novel bicyclam CXCR4 antagonist, induces the rapid mobilization of HSC with both short- and long-term repopulation capacity. Mobilization with G-CSF and AMD3100 in clinical trials resulted in more patients achieving sufficient PBSC for transplantation than with G-CSF alone. Thus, chemokine axis-mobilization could allow rapid PBSC harvests with increased cell yields in difficult-to mobilize patients. Studies of autologous and allogeneic transplantation of AMD3100 mobilized grafts demonstrated prompt and stable engraftment.Enhanced homing properties of chemokine axis-mobilized PBSC suggest that these cells may have greater therapeutic utility in other areas including tissue repair and regeneration.  相似文献   

2.
Granulocyte colony-stimulating factor (G-CSF), the prototypical mobilizing cytokine, induces hematopoietic stem and progenitor cell (HSPC) mobilization from the bone marrow in a cell-nonautonomous fashion. This process is mediated, in part, through suppression of osteoblasts and disruption of CXCR4/CXCL12 signaling. The cellular targets of G-CSF that initiate the mobilization cascade have not been identified. We use mixed G-CSF receptor (G-CSFR)-deficient bone marrow chimeras to show that G-CSF-induced mobilization of HSPCs correlates poorly with the number of wild-type neutrophils. We generated transgenic mice in which expression of the G-CSFR is restricted to cells of the monocytic lineage. G-CSF-induced HSPC mobilization, osteoblast suppression, and inhibition of CXCL12 expression in the bone marrow of these transgenic mice are intact, demonstrating that G-CSFR signals in monocytic cells are sufficient to induce HSPC mobilization. Moreover, G-CSF treatment of wild-type mice is associated with marked loss of monocytic cells in the bone marrow. Finally, we show that bone marrow macrophages produce factors that support the growth and/or survival of osteoblasts in vitro. Together, these data suggest a model in which G-CSFR signals in bone marrow monocytic cells inhibit the production of trophic factors required for osteoblast lineage cell maintenance, ultimately leading to HSPC mobilization.  相似文献   

3.
Improving approaches for hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) mobilization is clinically important because increased numbers of these cells are needed for enhanced transplantation. Chemokine stromal cell derived factor-1 (also known as CXCL12) is believed to be involved in retention of HSCs and HPCs in bone marrow. AMD3100, a selective antagonist of CXCL12 that binds to its receptor, CXCR4, was evaluated in murine and human systems for mobilizing capacity, alone and in combination with granulocyte colony-stimulating factor (G-CSF). AMD3100 induced rapid mobilization of mouse and human HPCs and synergistically augmented G-CSF-induced mobilization of HPCs. AMD3100 also mobilized murine long-term repopulating (LTR) cells that engrafted primary and secondary lethally-irradiated mice, and human CD34(+) cells that can repopulate nonobese diabetic-severe combined immunodeficiency (SCID) mice. AMD3100 synergized with G-CSF to mobilize murine LTR cells and human SCID repopulating cells (SRCs). Human CD34(+) cells isolated after treatment with G-CSF plus AMD3100 expressed a phenotype that was characteristic of highly engrafting mouse HSCs. Synergy of AMD3100 and G-CSF in mobilization was due to enhanced numbers and perhaps other characteristics of the mobilized cells. These results support the hypothesis that the CXCL12-CXCR4 axis is involved in marrow retention of HSCs and HPCs, and demonstrate the clinical potential of AMD3100 for HSC mobilization.  相似文献   

4.
目的探讨基质细胞衍生因子-1(SDF-1)及其特异性受体CXCR4在G-CSF诱导的造血干/祖细胞(HSPC)动员中的作用。方法应用酶联免疫吸附实验(ELISA)、免疫组织化学、流式细胞术等方法检测健康供者稳态及G-CSF动员过程中骨髓、外周血SDF-1/CXCR4的变化,并应用SDF-1中和性抗体阻断BALB/c小鼠SDF-1信号通路,进一步验证SDF-1/CXCR4在动员中的作用。结果G-CSF动员前骨髓和外周血的SDF-1浓度分别为(7.23±0.66)μg/L和(5.43±0.35)μg/L,动员后分别为(5.88±1.03)μg/L和(5.42±0.52)μg/L。动员后骨髓SDF-1蛋白水平下降(P<0.05),骨髓和外周血之间的SDF-1浓度梯度消失(P>0.05);稳态骨髓、动员后骨髓和动员后外周血的CD34^+ CXCR4^+细胞在CD34^+细胞群中的比例分别为(40.98±21.56)%、(65.80±24.68)%和(27.54±26.03)%。动员后CXCR4在骨髓CD34^+胞上表达增加(P<0.05),而外周血CD34^+细胞CXCR4表达降低(P<0.05)。SDF-1中和性抗体可降低G-CSF动员的BALB/c小鼠外周血成熟白细胞和祖细胞集落数量(P<0.05)。结论骨髓中SDF-1水平的降低以及CXCR4在HSPC上表达的下降促进了G-CSF介导的动员的发生。  相似文献   

5.
造血干细胞移植(HSCT)是治疗白血病、淋巴瘤、再生障碍性贫血等恶性血液病的一种有效治疗手段.但是,HSCT后造血功能的恢复与归巢至骨髓造血微环境中的造血干细胞(HSC)数目有关.HSCT输注入受者体内的HSC不会立刻发挥作用,而是先经历一系列复杂的过程归巢至骨髓造血微环境后,才能继续增殖并分化为相应的效应细胞发挥作用.其中,供者HSC与众多细胞及细胞因子间的相互作用是决定HSC归巢、增殖及分化的关键因素.基质细胞衍生因子(SDF)-1及其唯一的受体CXC趋化因子受体(CXCR)4所构成的SDF-1/CXCR4信号通路,可能在HSC归巢中发挥重要作用.为了更深入地研究HSC归巢的具体过程和SDF-1/CXCR4信号通路在该过程中发挥的作用,现对SDF-1、CXCR4的结构、作用机制,以及SDF1/CXCR4信号通路在HSC归巢中作用的研究进展进行综述.  相似文献   

6.
Regulation of hematopoietic stem and progenitor cell (HSPC) steady-state egress from the bone marrow (BM) to the circulation is poorly understood. While glycogen synthase kinase-3β (GSK3β) is known to participate in HSPC proliferation, we revealed an unexpected role in the preferential regulation of CXCL12-induced migration and steady-state egress of murine HSPCs, including long-term repopulating HSCs, over mature leukocytes. HSPC egress, regulated by circadian rhythms of CXCL12 and CXCR4 levels, correlated with dynamic expression of GSK3β in the BM. Nevertheless, GSK3β signaling was CXCL12/CXCR4 independent, suggesting that synchronization of both pathways is required for HSPC motility. Chemotaxis of HSPCs expressing higher levels of GSK3β compared with mature cells was selectively enhanced by stem cell factor–induced activation of GSK3β. Moreover, HSPC motility was regulated by norepinephrine and insulin-like growth factor-1 (IGF-1), which increased or reduced, respectively, GSK3β expression in BM HSPCs and their subsequent egress. Mechanistically, GSK3β signaling promoted preferential HSPC migration by regulating actin rearrangement and microtubuli turnover, including CXCL12-induced actin polarization and polymerization. Our study identifies a previously unknown role for GSK3β in physiological HSPC motility, dictating an active, rather than a passive, nature for homeostatic egress from the BM reservoir to the blood circulation.  相似文献   

7.
The quiescence of hematopoietic stem cells (HSCs) is critical for preserving a lifelong steady pool of HSCs to sustain the highly regenerative hematopoietic system. It is thought that specialized niches in which HSCs reside control the balance between HSC quiescence and self-renewal, yet little is known about the extrinsic signals provided by the niche and how these niche signals regulate such a balance. We report that CXCL12 produced by bone marrow (BM) stromal cells is not only the major chemoattractant for HSCs but also a regulatory factor that controls the quiescence of primitive hematopoietic cells. Addition of CXCL12 into the culture inhibits entry of primitive hematopoietic cells into the cell cycle, and inactivation of its receptor CXCR4 in HSCs causes excessive HSC proliferation. Notably, the hyperproliferative Cxcr4(-/-) HSCs are able to maintain a stable stem cell compartment and sustain hematopoiesis. Thus, we propose that CXCR4/CXCL12 signaling is essential to confine HSCs in the proper niche and controls their proliferation.  相似文献   

8.
Hematopoietic progenitor cells (HPCs) normally reside in the bone marrow (BM) but can be mobilized into the peripheral blood (PB) after treatment with GCSF or chemotherapy. In previous studies, we showed that granulocyte precursors accumulate in the BM during mobilization induced by either GCSF or cyclophosphamide (CY), leading to the accumulation of active neutrophil proteases in this tissue. We now report that mobilization of HPCs by GCSF coincides in vivo with the cleavage of the N-terminus of the chemokine receptor CXCR4 on HPCs resident in the BM and mobilized into the PB. This cleavage of CXCR4 on mobilized HPCs results in the loss of chemotaxis in response to the CXCR4 ligand, the chemokine stromal cell-derived factor-1 (SDF-1/CXCL12). Furthermore, the concentration of SDF-1 decreased in vivo in the BM of mobilized mice, and this decrease coincided with the accumulation of serine proteases able to directly cleave and inactivate SDF-1. Since both SDF-1 and its receptor, CXCR4, are essential for the homing and retention of HPCs in the BM, the proteolytic degradation of SDF-1, together with that of CXCR4, could represent a critical step leading to the mobilization of HPCs into the PB in response to GCSF or CY.  相似文献   

9.
HSCs differ during ontogeny in some important parameters, including anatomic site of residence and cell cycling characteristics. In this issue of the JCI, Bowie et al. show that postnatal HSCs as well as fetal liver HSCs in mice are active in the cell cycle at much higher rates than that of adult HSCs; however, this increased frequency of cycling abruptly ceases 4 weeks after birth (see the related article beginning on page 2808). The cycling postnatal HSCs expressed high levels of CXC chemokine ligand 12 (CXCL12, also known as stromal cell-derived factor 1 [SDF-1]), a chemokine previously implicated in stem cell trafficking to the marrow cavity and shown to be expressed by cells within the hematopoietic microenvironment. These cells also possessed an engraftment defect impeding reconstitution in irradiated recipient mice, which was reversible by pretransplant administration of antagonists of the CXCL12 receptor, CXCR4. Such agents are currently clinically available, suggesting that this approach could be used to improve stem cell transplantation and engraftment.  相似文献   

10.
基质细胞衍生因子(SDF)属CXC型趋化因子,主要在骨髓基质细胞和骨髓内皮等细胞表达,特异性地引起表达CXCR4的造血干细胞的趋化反应,因此在造血干细胞的迁移和归巢中起着重要作用。本文对SDF及其受体CXCR4在造血干细胞动员以及归巢过程中的机理进行简要综述。  相似文献   

11.
Peripheral blood stem cells (PBSC) have nearly replaced bone marrow (BM) as the preferred source of hematopoietic rescue for patients undergoing high-dose chemotherapy. However, some patients fail to mobilize sufficient numbers of PBSC into the peripheral blood thereby putting high-dose chemotherapy at risk. The present article reviews mobilization of PBSC with a special focus on poor mobilizers. Under steady-state conditions less than 0.05% of the white blood cells (WBC) are CD34+ cells. Chemotherapy results in a 5-15-fold increase of PBSC. Combining chemotherapy and growth factors increases CD34+ cells up to 6% of WBC. Several factors affect the mobilization of PBSC: age, gender, type of growth factor, dose of the growth factor and in the autologous setting patient's diagnosis, chemotherapy regimen and number of previous chemotherapy cycles or radiation. Poor mobilizers are defined as patients with less than 10 CD34+ cells/mul in the peripheral blood during mobilization. Promising approaches for those patients rely on remobilization, use of high doses of granulocyte-colony stimulating factor (G-CSF), or the combination of G-CSF and granulocyte macrophage (GM)-CSF, which successfully mobilized the majority of poor mobilizing patients. New agents such as long lasting variants of G-CSF and CXCR4 antagonists are at the horizon and studied in clinical trials as mobilizing agents. Muscle and bone pain are frequent adverse events in stem cell mobilization but are usually tolerated under the use of analgesics. Large volume apheresis (LVL) with a processed volume of more than 4-fold patient's blood volume is an approach to increase the CD34+ yield in patients with low CD34+ pre-counts resulting in higher yields of CD34+ cells for transplantation. Processing of more blood in LVL is achieved by an increase of the blood flow rate and an altered anticoagulation regimen with the occurrence of more citrate reactions.  相似文献   

12.
13.
Stem cells harvested from peripheral blood are the most commonly used graft source in hematopoietic stem cell transplantation. While G-CSF is the most frequently used agent for stem cell mobilization, the use of G-CSF alone results in suboptimal stem cell yields in a significant proportion of patients undergoing autologous transplantation. Plerixafor (AMD3100, Genzyme Corporation) is a bicyclam molecule that antagonizes the binding of the chemokine stromal cell-derived factor-1 (SDF-1) to its cognate receptor CXCR4. Plerixafor results in the rapid and reversible mobilization of hematopoietic stem cells into the peripheral circulation and is synergistic when combined with G-CSF. In clinical studies of autologous stem cell transplantation, the combination of plerixafor and G-CSF allows the collection of large numbers of stem cells in fewer apheresis sessions and can salvage those who fail G-CSF mobilization alone.  相似文献   

14.
Stem cells harvested from peripheral blood are the most commonly used graft source in hematopoietic stem cell transplantation. While G-CSF is the most frequently used agent for stem cell mobilization, the use of G-CSF alone results in suboptimal stem cell yields in a significant proportion of patients undergoing autologous transplantation. Plerixafor (AMD3100, Genzyme Corporation) is a bicyclam molecule that antagonizes the binding of the chemokine stromal cell-derived factor-1 (SDF-1) to its cognate receptor CXCR4. Plerixafor results in the rapid and reversible mobilization of hematopoietic stem cells into the peripheral circulation and is synergistic when combined with G-CSF. In clinical studies of autologous stem cell transplantation, the combination of plerixafor and G-CSF allows the collection of large numbers of stem cells in fewer apheresis sessions and can salvage those who fail G-CSF mobilization alone.  相似文献   

15.
The chemokine stromal cell-derived factor (SDF-1; also known as chemokine ligand 12 [CXCL12]) regulates many essential biological processes, including cardiac and neuronal development, stem cell motility, neovascularization, angiogenesis, apoptosis, and tumorigenesis. It is generally believed that SDF-1 mediates these many disparate processes via a single cell surface receptor known as chemokine receptor 4 (CXCR4). This paper characterizes an alternate receptor, CXCR7, which binds with high affinity to SDF-1 and to a second chemokine, interferon-inducible T cell alpha chemoattractant (I-TAC; also known as CXCL11). Membrane-associated CXCR7 is expressed on many tumor cell lines, on activated endothelial cells, and on fetal liver cells, but on few other cell types. Unlike many other chemokine receptors, ligand activation of CXCR7 does not cause Ca2+ mobilization or cell migration. However, expression of CXCR7 provides cells with a growth and survival advantage and increased adhesion properties. Consistent with a role for CXCR7 in cell survival and adhesion, a specific, high affinity small molecule antagonist to CXCR7 impedes in vivo tumor growth in animal models, validating this new receptor as a target for development of novel cancer therapeutics.  相似文献   

16.
Neutrophils are a major component of the innate immune response. Their homeostasis is maintained, in part, by the regulated release of neutrophils from the bone marrow. Constitutive expression of the chemokine CXCL12 by bone marrow stromal cells provides a key retention signal for neutrophils in the bone marrow through activation of its receptor, CXCR4. Attenuation of CXCR4 signaling leads to entry of neutrophils into the circulation through unknown mechanisms. We investigated the role of CXCR2-binding ELR+ chemokines in neutrophil trafficking using mouse mixed bone marrow chimeras reconstituted with Cxcr2–/– and WT cells. In this context, neutrophils lacking CXCR2 were preferentially retained in the bone marrow, a phenotype resembling the congenital disorder myelokathexis, which is characterized by chronic neutropenia. Additionally, transient disruption of CXCR4 failed to mobilize Cxcr2–/– neutrophils. However, neutrophils lacking both CXCR2 and CXCR4 displayed constitutive mobilization, showing that CXCR4 plays a dominant role in neutrophil trafficking. With regard to CXCR2 ligands, bone marrow endothelial cells and osteoblasts constitutively expressed the ELR+ chemokines CXCL1 and CXCL2, and CXCL2 expression was induced in endothelial cells during G-CSF–induced neutrophil mobilization. Collectively, these data suggest that CXCR2 signaling is a second chemokine axis that interacts antagonistically with CXCR4 to regulate neutrophil release from the bone marrow.  相似文献   

17.
Hematopoietic stem cells may be obtained by collection of bone marrow, mobilization and collection of peripheral blood stem cells or umbilical cord blood. Transplantation of peripheral blood hematopoietic cells has increased due to faster engraftment and practicability in both the related, unrelated or haploidentical setting. We reviewed the question of which stem cell source - bone marrow (BM) or peripheral blood (PBSC) - is the most suitable for individuals undergoing haploidentical stem cell transplantation. BM or PBSC could be safely used as allograft sources for haploidentical transplantation with good outcomes and acceptable rates of GVHD and graft failure. Prospective randomized studies are needed to evaluate the effect of PB vs BM in haploidentical settings.  相似文献   

18.
The mechanisms of BM hematopoietic stem/progenitor cell (HSPC) adhesion, engraftment, and mobilization remain incompletely identified. Here, using WT and transgenic mice, we have shown that membrane-anchored plasminogen activator, urokinase receptor (MuPAR) marks a subset of HSPCs and promotes the preservation of the size of this pool of cells in the BM. Loss or inhibition of MuPAR increased HSPC proliferation and impaired their homing, engraftment, and adhesion to the BM microenvironment. During mobilization, MuPAR was inactivated by plasmin via proteolytic cleavage. Cell-autonomous loss of the gene encoding MuPAR also impaired long-term engraftment and multilineage repopulation in primary and secondary recipient mice. These findings identify MuPAR and plasmin as regulators of the proliferation, marrow pool size, homing, engraftment, and mobilization of HSPCs and possibly also of HSCs.  相似文献   

19.
Since the demonstration that hematopoietic cells are present in circulating blood, peripheral blood stem cell transplantation (PBSCT) has become an area of interest. The invention of growth factors such as the granulocyte colony-stimulating factor (G-CSF) and the availability of apheresis techniques allowed the wide application of peripheral blood stem cells (PBSC) in both autologous and allogeneic hematopoietic stem cell transplantation settings. It has been since 1986 that clinically introduced, peripheral blood stem cells replaced bone marrow as a stem-cell source to nearly 100% in the autologous and to approximately 75% in the allogeneic transplantation setting. During this period of time, remarkable development occurred in both stem cell mobilizing agents (i.e. CXCR4 antagonists) and apheresis techniques. Currently, apheresis technology is being increasingly used in not only for collection of PBSC or blood product support, but also for treatment and/or prevention of several transplantations related complications. Apheresis technology also allows to manipulate stem cells and thus provides opportunity to curative treatment of certain diseases.  相似文献   

20.
BackgroundThe stem cell niche in human bone marrow provides scaffolds, cellular frameworks and essential soluble cues to support the stemness of hematopoietic stem and progenitor cells (HSPCs). To decipher this complex structure and the corresponding cellular interactions, a number of in vitro model systems have been developed. The cellular microenvironment is of key importance, and mesenchymal stromal cells (MSCs) represent one of the major cellular determinants of the niche. Regulation of the self-renewal and differentiation of HSPCs requires not only direct cellular contact and adhesion molecules, but also various cytokines and chemokines. The C-X-C chemokine receptor type 4/stromal cell-derived factor 1 axis plays a pivotal role in stem cell mobilization and homing. As we have learned in recent years, to realistically simulate the physiological in vivo situation, advanced model systems should be based on niche cells arranged in a three-dimensional (3D) structure. By providing a dynamic rather than static setup, microbioreactor systems offer a number of advantages. In addition, the role of low oxygen tension in the niche microenvironment and its impact on hematopoietic stem cells need to be taken into account and are discussed in this review.SummaryThis review focuses on the role of MSCs as a part of the bone marrow niche, the interplay between MSCs and HSPCs and the most important regulatory factors that need to be considered when engineering artificial hematopoietic stem cell niche systems.ConclusionAdvanced 3D model systems using MSCs as niche cells and applying microbioreactor-based technology are capable of simulating the natural properties of the bone marrow niche more closely than ever before.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号