Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Stem Cell Rev Rep ; 16(1): 198-211, 2020 02.
Article in English | MEDLINE | ID: mdl-31828580

ABSTRACT

BACKGROUND: Bone marrow-derived mononuclear cells (BM-MNC) consist of a heterogeneous mix of mesenchymal stem cells (MSC), hematopoietic progenitor cells (HPC), endothelial progenitor cells (EPC), monocytes, lymphocytes and pluripotent stem cells. Whereas the importance of MSC and EPC has been well documented in bone healing and regeneration studies, the role of pluripotent stem cells is still poorly understood. In the present study we evaluated if and how Very Small Embryonic Like cells (VSEL), isolated from rat BM-MNC, contribute to bone healing. METHODS: Large bone defects were made in the femurs of 38 Sprague Dawley female rats and treated with ß-TCP scaffold granules seeded with male VSEL; BM-MNC, VSEL-depleted BM-MNC or scaffold alone, and bone healing was evaluated at 8 weeks post-surgery. RESULTS: Bone healing was significantly increased in defects treated with VSEL and BM-MNC, compared to defects treated with VSEL-depleted BM-MNC. Donor cells were detected in new bone tissue, in all the defects treated with cells, and in fibrous tissue only in defects treated with VSEL-depleted BM-MNC. The number of CD68+ cells was the highest in the VSEL-depleted group, whereas the number of TRAP positive cells was the lowest in this group. CONCLUSIONS: Based on the results, we can conclude that VSEL play a role in BM-MNC induced bone formation. In our rat femur defect model, in defects treated with VSEL-depleted BM-MNC, osteoclastogenesis and bone formation were decreased, and foreign body reaction was increased.


Subject(s)
Adult Stem Cells/transplantation , Bone Regeneration/genetics , Mesenchymal Stem Cell Transplantation , Pluripotent Stem Cells/transplantation , Adult , Animals , Endothelial Progenitor Cells/transplantation , Humans , Monocytes/transplantation , Osteogenesis/genetics , Rats
2.
Mediators Inflamm ; 2018: 2691934, 2018.
Article in English | MEDLINE | ID: mdl-30116144

ABSTRACT

BACKGROUND: Acute myocardial infarction (AMI) and coronary artery bypass graft (CABG) surgery are associated with a pathogen-free inflammatory response (sterile inflammation). Complement cascade (CC) and bioactive sphingolipids (BS) are postulated to be involved in this process. AIM: The aim of this study was to evaluate plasma levels of CC cleavage fragments (C3a, C5a, and C5b9), sphingosine (SP), sphingosine-1-phosphate (S1P), and free hemoglobin (fHb) in AMI patients treated with primary percutaneous coronary intervention (pPCI) and stable coronary artery disease (SCAD) undergoing CABG. PATIENTS AND METHODS: The study enrolled 37 subjects (27 male) including 22 AMI patients, 7 CABG patients, and 8 healthy individuals as the control group (CTRL). In the AMI group, blood samples were collected at 5 time points (admission to hospital, 6, 12, 24, and 48 hours post pPCI) and 4 time points in the CABG group (6, 12, 24, and 48 hours post operation). SP and S1P concentrations were measured by high-performance liquid chromatography (HPLC). Analysis of C3a, C5a, and C5b9 levels was carried out using high-sensitivity ELISA and free hemoglobin by spectrophotometry. RESULTS: The plasma levels of CC cleavage fragments (C3a and C5b9) were significantly higher, while those of SP and S1P were lower in patients undergoing CABG surgery in comparison to the AMI group. In both groups, levels of CC factors showed no significant changes within 48 hours of follow-up. Conversely, SP and S1P levels gradually decreased throughout 48 hours in the AMI group but remained stable after CABG. Moreover, the fHb concentration was significantly higher after 24 and 48 hours post pPCI compared to the corresponding postoperative time points. Additionally, the fHb concentrations increased between 12 and 48 hours after PCI in patients with AMI. CONCLUSIONS: Inflammatory response after AMI and CABG differed regarding the release of sphingolipids, free hemoglobin, and complement cascade cleavage fragments.


Subject(s)
Complement System Proteins/analysis , Coronary Artery Disease/blood , Hemoglobins/analysis , Myocardial Infarction/blood , Sphingolipids/metabolism , Aged , Case-Control Studies , Coronary Artery Bypass , Female , Humans , Inflammation , Lysophospholipids/metabolism , Male , Middle Aged , Percutaneous Coronary Intervention , Sphingolipids/blood , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Treatment Outcome
3.
Leukemia ; 31(8): 1671-1677, 2017 08.
Article in English | MEDLINE | ID: mdl-28502982

ABSTRACT

Evidence has accumulated that normal human and murine hematopoietic stem cells express several functional pituitary and gonadal sex hormones, and that, in fact, some sex hormones, such as androgens, have been employed for many years to stimulate hematopoiesis in patients with bone marrow aplasia. Interestingly, sex hormone receptors are also expressed by leukemic cell lines and blasts. In this review, I will discuss the emerging question of why hematopoietic cells express these receptors. A tempting hypothetical explanation for this phenomenon is that hematopoietic stem cells are related to subpopulation of migrating primordial germ cells. To support of this notion, the anatomical sites of origin of primitive and definitive hematopoiesis during embryonic development are tightly connected with the migratory route of primordial germ cells: from the proximal epiblast to the extraembryonic endoderm at the bottom of the yolk sac and then back to the embryo proper via the primitive streak to the aorta-gonado-mesonephros (AGM) region on the way to the genital ridges. The migration of these cells overlaps with the emergence of primitive hematopoiesis in the blood islands at the bottom of the yolk sac, and definitive hematopoiesis that occurs in hemogenic endothelium in the embryonic dorsal aorta in AGM region.


Subject(s)
Embryonic Development/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Movement , Gonadal Steroid Hormones/physiology , Hematopoiesis , Humans
5.
Leukemia ; 31(2): 446-458, 2017 02.
Article in English | MEDLINE | ID: mdl-27451975

ABSTRACT

As a crucial arm of innate immunity, the complement cascade (ComC) is involved both in mobilization of normal hematopoietic stem/progenitor cells (HSPCs) from bone marrow (BM) into peripheral blood and in their homing to BM. Despite the fact that ComC cleavage fragments alone do not chemoattract normal HSPCs, we found that leukemia cell lines as well as clonogenic blasts from chronic myeloid leukemia and acute myeloid leukemia patients respond robustly to C3 and C5 cleavage fragments by chemotaxis and increased adhesion. This finding was supported by the detection of C3a and C5a receptors in cells from human malignant hematopoietic cell lines and patient blasts at the mRNA (reverse transcriptase-polymerase chain reaction) and protein level (fluorescence-activated cell sorting), and by the demonstration that these receptors respond to stimulation by C3a and C5a by phosphorylation of p42/44 and p38 mitogen-activated protein kinases (MAPK), and protein kinase B (PKB/AKT). We also found that inducible heme oxygenase 1 (HO-1) is a negative regulator of ComC-mediated trafficking of leukemic cells, and that stimulation of leukemic cells by C3 or C5 cleavage fragments activates p38 MAPK, which downregulates HO-1 expression, rendering cells more mobile. We conclude that activation of the ComC in leukemia/lymphoma patients (for example, as a result of accompanying infections) enhances the motility of malignant cells and contributes to their spread in a p38 MAPK-HO-1-dependent manner. Therefore, inhibition of p38 MAPK or upregulation of HO-1 by small-molecule modulators would have a beneficial effect on ameliorating cell migration-mediated expansion of leukemia/lymphoma cells when the ComC becomes activated.


Subject(s)
Complement Activation/immunology , Gene Expression Regulation, Leukemic , Heme Oxygenase-1/genetics , Leukemia/genetics , Leukemia/immunology , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Line, Tumor , Cell Movement/immunology , Chemotaxis/genetics , Chemotaxis/immunology , Complement C3/immunology , Complement C3/metabolism , Complement C5/immunology , Complement C5/metabolism , Down-Regulation , Flow Cytometry , Gene Knockout Techniques , Hematopoietic Stem Cells/metabolism , Heterografts , Humans , Immunophenotyping , Mice , Proteolysis , RNA, Small Interfering/genetics , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Sialic Acid Binding Ig-like Lectin 3/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Leukemia ; 30(4): 919-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26582648

ABSTRACT

Hematopoietic stem/progenitor cells (HSPCs) reside in the bone marrow (BM) microenvironment and are retained there by the interaction of membrane lipid raft-associated receptors, such as the α-chemokine receptor CXCR4 and the α4ß1-integrin (VLA-4, very late antigen 4 receptor) receptor, with their respective specific ligands, stromal-derived factor 1 and vascular cell adhesion molecule 1, expressed in BM stem cell niches. The integrity of the lipid rafts containing these receptors is maintained by the glycolipid glycosylphosphatidylinositol anchor (GPI-A). It has been reported that a cleavage fragment of the fifth component of the activated complement cascade, C5a, has an important role in mobilizing HSPCs into the peripheral blood (PB) by (i) inducing degranulation of BM-residing granulocytes and (ii) promoting their egress from the BM into the PB so that they permeabilize the endothelial barrier for subsequent egress of HSPCs. We report here that hematopoietic cell-specific phospholipase C-ß2 (PLC-ß2) has a crucial role in pharmacological mobilization of HSPCs. On the one hand, when released during degranulation of granulocytes, it digests GPI-A, thereby disrupting membrane lipid rafts and impairing retention of HSPCs in BM niches. On the other hand, it is an intracellular enzyme required for degranulation of granulocytes and their egress from BM. In support of this dual role, we demonstrate that PLC-ß2-knockout mice are poor mobilizers and provide, for the first time, evidence for the involvement of this lipolytic enzyme in the mobilization of HSPCs.


Subject(s)
Bone Marrow/enzymology , Complement C5a/metabolism , Granulocytes/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Membrane Microdomains , Phospholipase C beta/physiology , Animals , Apoptosis , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Flow Cytometry , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
8.
Am J Transplant ; 16(2): 440-53, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26550777

ABSTRACT

CD8(+)/TCR(-) facilitating cells (FCs) in mouse bone marrow (BM) significantly enhance engraftment of hematopoietic stem/progenitor cells (HSPCs). Human FC phenotype and mechanism of action remain to be defined. We report, for the first time, the phenotypic characterization of human FCs and correlation of phenotype with function. Approximately half of human FCs are CD8(+)/TCR(-)/CD56 negative (CD56(neg)); the remainder are CD8(+)/TCR(-)/CD56 bright (CD56(bright)). The CD56(neg) FC subpopulation significantly promotes homing of HSPCs to BM in nonobese diabetic/severe combined immunodeficiency/IL-2 receptor γ-chain knockout mouse recipients and enhances hematopoietic colony formation in vitro. The CD56(neg) FC subpopulation promotes rapid reconstitution of donor HSPCs without graft-versus-host disease (GVHD); recipients of CD56(bright) FCs plus HSPCs exhibit low donor chimerism early after transplantation, but the level of chimerism significantly increases with time. Recipients of HSPCs plus CD56(neg) or CD56(bright) FCs showed durable donor chimerism at significantly higher levels in BM. The majority of both FC subpopulations express CXCR4. Coculture of CD56(bright) FCs with HSPCs upregulates cathelicidin and ß-defensin 2, factors that prime responsiveness of HSPCs to stromal cell-derived factor 1. Both FC subpopulations significantly upregulated mRNA expression of the HSPC growth factors and Flt3 ligand. These results indicate that human FCs exert a direct effect on HSPCs to enhance engraftment. Human FCs offer a potential regulatory cell-based therapy for enhancement of engraftment and prevention of GVHD.


Subject(s)
CD8 Antigens/metabolism , Graft vs Host Disease/immunology , Hematopoietic Stem Cells/immunology , Interleukin Receptor Common gamma Subunit/physiology , Receptors, Antigen, T-Cell/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Graft vs Host Disease/metabolism , Hematopoietic Stem Cells/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Models, Animal , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tissue Donors , Transplantation Chimera
9.
Leukemia ; 29(7): 1452-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25748684

ABSTRACT

Cell outer membranes contain glycosphingolipids and protein receptors, which are integrated into glycoprotein microdomains, known as lipid rafts, which float freely in the membrane bilayer. These structures have an important role in assembling signaling molecules (e.g., Rac-1, RhoH and Lyn) together with surface receptors, such as the CXCR4 receptor for α-chemokine stromal-derived factor-1, the α4ß1-integrin receptor (VLA-4) for vascular cell adhesion molecule-1 and the c-kit receptor for stem cell factor, which together regulate several aspects of hematopoietic stem/progenitor cell (HSPC) biology. Here, we discuss the role of lipid raft integrity in the retention and quiescence of normal HSPCs in bone marrow niches as well as in regulating HSPC mobilization and homing. We will also discuss the pathological consequences of the defect in lipid raft integrity seen in paroxysmal nocturnal hemoglobinuria and the emerging evidence for the involvement of lipid rafts in hematological malignancies.


Subject(s)
Bone Marrow/physiology , Cell Movement/physiology , Hematopoietic Stem Cell Mobilization , Membrane Lipids , Membrane Microdomains , Animals , Humans , Signal Transduction
10.
Leukemia ; 29(4): 776-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25486871

ABSTRACT

This review presents a novel view and working hypothesis about the hierarchy within the adult bone marrow stem cell compartment and the still-intriguing question of whether adult bone marrow contains primitive stem cells from early embryonic development, such as cells derived from the epiblast, migrating primordial germ cells or yolk sac-derived hemangioblasts. It also presents a novel view of the mechanisms that govern stem cell mobilization and homing, with special emphasis on the role of the complement cascade as a trigger for egress of hematopoietic stem cells from bone marrow into blood as well as the emerging role of novel homing factors and priming mechanisms that support stromal-derived factor 1-mediated homing of hematopoietic stem/progenitor cells after transplantation.


Subject(s)
Bone Marrow Cells/cytology , Germ Cells/cytology , Hematopoietic Stem Cells/cytology , Adult , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow Cells/classification , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Lineage/genetics , Cell Lineage/immunology , Cell Movement , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Complement System Proteins/genetics , Gene Expression , Germ Cells/immunology , Germ Cells/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans
11.
Leukemia ; 28(11): 2148-54, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24667943

ABSTRACT

The role of blood proteinases in the mobilization of hematopoietic stem/progenitor cells (HSPCs) is still not well understood. As previously reported, activation of the complement cascade (ComC) and cleavage of C5 by C5 convertase are enabling events in the release of C5a that plays a crucial role in the egress of HSPCs from bone marrow (BM) into peripheral blood (PB) and explains why C5-deficient mice are poor mobilizers. Here we provide evidence that during granulocyte colony-stimulating factor- and AMD3100-induced mobilization, not only the ComC but also two other evolutionarily ancient proteolytic enzyme cascades, the coagulation cascade (CoaC) and the fibrynolytic cascade (FibC), become activated. Activation of all three cascades was measured by generation of C5a, decrease in prothrombin time and activated partial thromboplastin time as well as an increase in the concentrations of plasmin/antiplasmin and thrombin/antithrombin. More importantly, the CoaC and FibC, by generating thrombin and plasmin, respectively, provide C5 convertase activity, explaining why mobilization of HSPCs in C3-deficient mice, which do not generate ComC-generated C5a convertase, is not impaired. Our observations shed more light on how the CoaC and FibC modulate stem cell mobilization and may lead to the development of more efficient mobilization strategies in poor mobilizers. Furthermore, as it is known that all these cascades are activated in all the situations in which HSPCs are mobilized from BM into PB (for example, infections, tissue/organ damage or strenuous exercise) and show a circadian rhythm of activation, they must be involved in both stress-induced and circadian changes in HSPC trafficking in PB.


Subject(s)
Blood Coagulation/physiology , Complement C3/metabolism , Complement C5a/metabolism , Fibrinolysis/physiology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/physiology , Animals , Benzylamines , Blood Coagulation/drug effects , Complement C3/genetics , Cyclams , Female , Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Heterocyclic Compounds/pharmacology , Hirudins/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Receptors, CXCR4/antagonists & inhibitors , Recombinant Proteins/pharmacology , Tranexamic Acid/pharmacology
12.
Leukemia ; 28(3): 473-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24018851

ABSTRACT

The concept that adult tissue, including bone marrow (BM), contains early-development cells with broader differentiation potential has again been recently challenged. In response, we would like to review the accumulated evidence from several independent laboratories that adult tissues, including BM, harbor a population of very rare stem cells that may cross germ layers in their differentiation potential. Thus, the BM stem cell compartment hierarchy needs to be revisited. These dormant, early-development cells that our group described as very small embryonic-like stem cells (VSELs) most likely overlap with similar populations of stem cells that have been identified in adult tissues by other investigators as the result of various experimental strategies and have been given various names. As reported, murine VSELs have some pluripotent stem cell characteristics. Moreover, they display several epiblast/germline markers that suggest their embryonic origin and developmental deposition in adult BM. Moreover, at the molecular level, changes in expression of parentally imprinted genes (for example, Igf2-H19) and resistance to insulin/insulin-like growth factor signaling (IIS) regulates their quiescent state in adult tissues. In several emergency situations related to organ damage, VSELs can be activated and mobilized into peripheral blood, and in appropriate animal models they contribute to tissue organ/regeneration. Interestingly, their number correlates with lifespan in mice, and they may also be involved in some malignancies. VSELs have been successfully isolated in several laboratories; however, some investigators experience problems with their isolation.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Epigenesis, Genetic , Genomic Imprinting , Mice
13.
Leuk Suppl ; 3(Suppl 1): S19-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-27175266
16.
Leukemia ; 27(4): 773-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23135355

ABSTRACT

In recent years, solid evidence has accumulated that insulin-like growth factor-1 (IGF-1) and 2 (IGF-2) regulate many biological processes in normal and malignant cells. Recently, more light has been shed on the epigenetic mechanisms regulating expression of genes involved in IGF signaling (IFS) and it has become evident that these mechanisms are crucial for initiation of embryogenesis, maintaining the quiescence of pluripotent stem cells deposited in adult tissues (for example, very-small embryonic-like stem cells), the aging process, and the malignant transformation of cells. The expression of several genes involved in IFS is regulated at the epigenetic level by imprinting/methylation within differentially methylated regions (DMRs), which regulate their expression from paternal or maternal chromosomes. The most important role in the regulation of IFS gene expression is played by the Igf-2-H19 locus, which encodes the autocrine/paracrine mitogen IGF-2 and the H19 gene, which gives rise to a non-coding RNA precursor of several microRNAs that negatively affect cell proliferation. Among these, miR-675 has recently been demonstrated to downregulate expression of the IGF-1 receptor. The proper imprinting of DMRs at the Igf-2-H19 locus, with methylation of the paternal chromosome and a lack of methylation on the maternal chromosome, regulates expression of these genes so that Igf-2 is transcribed only from the paternal chromosome and H19 (including miR-675) only from the maternal chromosome. In this review, we will discuss the relevance of (i) proper somatic imprinting, (ii) erasure of imprinting and (iii) loss of imprinting within the DMRs at the Igf-2-H19 locus to the expression of genes involved in IFS, and the consequences of these alternative patterns of imprinting for stem cell biology.


Subject(s)
Aging/physiology , Cell Transformation, Neoplastic , Genomic Imprinting , Insulin-Like Growth Factor II/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction , DNA Methylation , Epigenesis, Genetic , Female , Humans , Insulin-Like Growth Factor II/genetics , Male , RNA, Long Noncoding/genetics
17.
Adv Med Sci ; 57(1): 1-17, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22515973

ABSTRACT

One of the most intriguing questions in stem cell biology is whether pluripotent stem cells exist in adult tissues. Several groups of investigators employing i) various isolation protocols, ii) detection of surface markers, and iii) experimental in vitro and in vivo models, have reported the presence of cells that possess a pluripotent character in adult tissues. Such cells were assigned various operational abbreviations and names in the literature that added confusion to the field and raised the basic question of whether these are truly distinct or overlapping populations of the same primitive stem cells. Unfortunately, these cells were never characterized side-by-side to address this important issue. Nevertheless, taking into consideration their common features described in the literature, it is very likely that various investigators have described overlapping populations of developmentally early stem cells that are closely related. These different populations of stem cells will be reviewed in this paper.


Subject(s)
Fetal Blood/cytology , Multipotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Adult , Humans , Multipotent Stem Cells/metabolism , Pluripotent Stem Cells/metabolism
19.
Leukemia ; 26(6): 1166-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22182853

ABSTRACT

Although regenerative medicine is searching for pluripotent stem cells that could be employed for therapy, various types of more differentiated adult stem and progenitor cells are in meantime being employed in clinical trials to regenerate damaged organs (for example, heart, kidney or neural tissues). It is striking that, for a variety of these cells, the currently observed final outcomes of cellular therapies are often similar. This fact and the lack of convincing documentation for donor-recipient chimerism in treated tissues in most of the studies indicates that a mechanism other than transdifferentiation of cells infused systemically into peripheral blood or injected directly into damaged organs may have an important role. In this review, we will discuss the role of (i) growth factors, cytokines, chemokines and bioactive lipids and (ii) microvesicles (MVs) released from cells employed as cellular therapeutics in regenerative medicine. In particular, stem cells are a rich source of these soluble factors and MVs released from their surface may deliver RNA and microRNA into damaged organs. Based on these phenomena, we suggest that paracrine effects make major contributions in most of the currently reported positive results in clinical trials employing adult stem cells. We will also present possibilities for how these paracrine mechanisms could be exploited in regenerative medicine to achieve better therapeutic outcomes. This approach may yield critical improvements in current cell therapies before true pluripotent stem cells isolated in sufficient quantities from adult tissues and successfully expanded ex vivo will be employed in the clinic.


Subject(s)
Cell-Derived Microparticles/physiology , Intercellular Signaling Peptides and Proteins/therapeutic use , Paracrine Communication , Pluripotent Stem Cells/cytology , Regenerative Medicine , Stem Cell Transplantation , Adult , Cell Differentiation , Humans
20.
Leukemia ; 26(1): 63-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21886175

ABSTRACT

Hematopoietic stem progenitor cells (HSPCs) respond robustly to α-chemokine stromal-derived factor-1 (SDF-1) gradients, and blockage of CXCR4, a seven-transmembrane-spanning G(αI)-protein-coupled SDF-1 receptor, mobilizes HSPCs into peripheral blood. Although the SDF-1-CXCR4 axis has an unquestionably important role in the retention of HSPCs in bone marrow (BM), new evidence shows that, in addition to SDF-1, the migration of HSPCs is directed by gradients of the bioactive lipids sphingosine-1 phosphate and ceramide-1 phosphate. Furthermore, the SDF-1 gradient may be positively primed/modulated by cationic peptides (C3a anaphylatoxin and cathelicidin) and, as previously demonstrated, HSPCs respond robustly even to very low SDF-1 gradients in the presence of priming factors. In this review, we discuss the role of bioactive lipids in stem cell trafficking and the consequences of HSPC priming by cationic peptides. Together, these phenomena support a picture in which the SDF-1-CXCR4 axis modulates homing, BM retention and mobilization of HSPCs in a more complex way than previously envisioned.


Subject(s)
Chemokine CXCL12/physiology , Hematopoietic Stem Cell Mobilization , Lipids/physiology , Peptides/physiology , Animals , Cations , Humans , Mice , Receptors, CXCR4/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...