Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Mar 1.
Published in final edited form as: Leukemia. 2011 Jul 5;25(11):1674–1686. doi: 10.1038/leu.2011.167

Bone marrow stem cell mobilization in stroke: a ‘bonehead’ may be good after all!

CV Borlongan 1
PMCID: PMC3291108  NIHMSID: NIHMS358674  PMID: 21727900

Abstract

Mobilizing bone cells to the head, astutely referred to as ‘bonehead’ therapeutic approach, represents a major discipline of regenerative medicine. The last decade has witnessed mounting evidence supporting the capacity of bone marrow (BM)-derived cells to mobilize from BM to peripheral blood (PB), eventually finding their way to the injured brain. This homing action is exemplified in BM stem cell mobilization following ischemic brain injury. Here, I review accumulating laboratory studies implicating the role of therapeutic mobilization of transplanted BM stem cells for brain plasticity and remodeling in stroke.

Keywords: stroke, cell therapy, migration, homing, neurogenesis, angiogenesis

Introduction

Bone marrow (BM) consists of a heterogeneous population stem and progenitor cells1 (Figure 1), with hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) as the two most studied BM-derived stem cells. Additionally, endothelial progenitor cells (EPCs) and very small embryonic-like stem cells (VSELs) have also been isolated from the BM. Previous reports of in vitro differentiation of BM-derived stem cells into neurons on exposure to various inducing regimens,2 and their secretion of growth factors critical for neuronal survival,35 have prompted interest in using BM as stem cell donor source for transplantation therapy in neurological disorders, such as stroke.

Figure 1.

Figure 1

Bone marrow-derived stem cells. Schematic diagram shows subsets of bone marrow-derived stem cells, including HSCs, MSCs, EPCs and VSELs, that have been examined in the laboratory and are rapidly being translated into clinical applications as efficacious stem cell sources for transplantation therapy in stroke.

Stroke remains a primary cause of death in the United States and around the world. Over the last decade, stem cell therapy has shown promise as an experimental treatment for stroke.68 Although the first clinical trial occurred in 1998,911 only recently new clinical trials on cell therapy have been again initiated in stroke patients. In the clinic, a minimally invasive transplant procedure via intravascular route is thought to be practical. However, this peripheral route of cell injection requires mobilization of the cells and their secreted products proximal to the site of injury in order to afford brain plasticity and remodeling. A better understanding of mechanisms underlying the homing of cells from the periphery to the brain is likely to aid in optimizing cell therapy for stroke. Along this theme, I discuss the unique but sometimes overlapping mobilization pathways that guide the homing of HSCs, MSCs, EPCs and VSELs from BM (and other stem cell niches) to blood and eventually into ischemic brain.

Hematopoietic stem cells

Repopulation of the ablated BM by HSCs has been arguably presented as the defining feature of HSC stemness12 (Table 1). The quiescent HSCs, although low in number during homeostasis, can quickly become motile with increased migration from their resident BM to blood circulation in response to injury.1315 A key chemokine implicated in the quiescence of HSCs within the BM is stromal derived factor-1 (SDF-1, also termed CXCL12) acting via its major receptor CXCR4 (C-X-C chemokine receptor type 4),13,46 which is also highly expressed in other stem cells niches, similarly playing the role of preserving the HSC primitive status.12,46 It is originally found to be expressed by murine and human BM endothelial and endosteal bone-lining stromal cells,47 and later also found in various tissues, including skin,48 epithelial cells in human liver bile ducts49 and brain endothelium.50 In the hematopoietic system, SDF-1 is a crucial chemoattractant for CXCR4-expressing BM-derived cells, including HSCs. This SDF-1/CXCR4 chemoattractant pathway is essential for stem cell migration and seeding. When SDF-1 is activated, these stem cells migrate out from the BM reservoir to the circulation, replenishing the blood with new cells.51

Table 1.

Characteristics of bone marrow-derived hematopoietic and nonhematopoietic stem cells

Bone marrow
subset
Phenotype Differentiation
potential
Telomerase Senescence tendency Reference
HSCs CD34+, CXCR4+ Blood cells Positive Up for debate, thought to be dysfunctional in aging HSCs, but even reduced telomere expression neither shorten telomere nor limit PP 1217
MSCs SH2+, SH3+, CD29+, CD44+, CD14−, CD34− and CD45− OCA cells Negative CPDL is ~18 PDs at ~80 days 18
Not addressed OCA cells Not addressed Mean CPDL is ~38 PDs at passage 15 19
Not addressed OCA cells Not addressed Lose MDP at 19–22 PDs. CPDL is ~22 to 23 in 80 days of culture 20
Not addressed OCA cells Not addressed Lose MDP at 19–22 PDs. CPDL is ~22 to 23 in 80 days of culture 21
Not addressed OCA cells Not addressed CPDL in serum-deprived hMSCs is ~9 PDs at passage 10. CPDL for control hMSCs is ~7 PDs at passage 9 22
Not addressed OCA cells Not addressed CPDL is 16 PDs of ~40 days of culture 23
hTERT-MSCs Not addressed OCA cells Not addressed PP remains until 80 PDs of ~400 days of culture 23
EPCs CD34+, CD31+, AC133+, VEGFR2+, Tie-2+ Endothelial cells Positive PDs until 7 to 8 days, with senescence augmented by estrogen or SDF-1 treatment 2437
VSEL cells SSEA-1+, Oct-4+, Nanog+, Rex-1+, Sca-1+, CD45− Three germ lineages Positive Expand for ~7 PDs (CV Borlongan, personal communication) 3845

Abbreviations: CPDL, cumulative population doubling level; EPC, endothelial progenitor cell; hMSC, human mesenchymal stem cell; HSC, hematopoietic stem cell; hTERT, human telomerase reverse transcriptase; MDP, multiple lineages differentiate potential; MSC, mesenchymal stem cell; OCA cell, osteogenic, chondrogenic and adipogenic cell; PD, population doubling; PP, proliferation potential; SDF-1, stromal derived factor-1; VSEL, very small embryonic-like stem cell.

Not Addressed: authors did not describe this topic in their study.

Negative/positive: the activity of telomerase is negative or positive.

The HSC exit from and re-entry to stem cell niches involve adhesion to the vascular wall and crossing over the endothelial blood–BM barrier.12 The central nervous system (CNS) contributes to this HSC mobilization via cytokine production, and under stress condition (that is, stroke) can amplify the recruitment of HSCs into the brain.1215 This cytokine-mediated recruitment of HSCs from BM to circulation is an established clinical mobilization regimen, such as treatment with granulocyte-colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF), for generation of ample supply of HSCs for transplantation.1315

Neural control of migration of BM-derived HSCs

As noted above, CNS-induced cytokine cues may serve as migratory signals to HSCs. Recently, a crosstalk via the neurotransmitter catecholaminergic signaling pathway has been proposed to link the nervous system and the immune system.12 The sympathetic system produces catecholamines, which are amplified during stress situations and secreted by activated leukocytes, including lymphocytes and lipopolysaccharide-stimulated macrophages, harnessing mobilization of BM-derived stem cells.12 In particular, catecholamines, either through the sympathetic system secreting these neurotransmitters into the blood circulation or directly from the nerve endings in the BM acting via a paracrine fashion, can modulate the migration of BM from its residence to the injured CNS.52,53 That this directed migration may entail a ligand-receptor mechanism is indicated by upregulated levels of catecholaminergic receptors in mobilized human CD34 HSCs compared with tissue-anchored BM human CD34 HSCs, with the catecholaminergic receptor expression further increased by repeated stimulation with the mobilizing agents G-CSF and GM-CSF. This homing process of BM-derived stem cells via the catecholaminergic neurotransmitter system involves multiple signaling pathways, including Wnt and β-catenin, as well as specific migratory molecules, such as membrane-bound enzyme membrane type 1 matrix metalloproteinase and SDF-15356 that altogether contributes to the cell proliferation, increased motility and engraftment capabilities of human CD34 HSCs.

HSC regulation of neural function

The neurotransmitter-mediated interaction between CNS and BM is bidirectional, with a hematopoietic signaling mechanism equally contributing to HSC regulation of CNS functions.57 Accumulating scientific evidence advances the concept that human HSCs can affect the nervous system and modulate its action.53 In the field of stroke, clinical data show that human acute stroke is followed by large and bursting mobilization of peripheral blood (PB) immature hematopoietic CD34+ cells, colony-forming cells and long-term culture-initiating cells,58 and the extent of such mobilization is directly related to recovery of function.59 The main postulated mechanism underlying HSC mobilization implicates the upregulation of SDF-1 within ischemic tissues of stroke individuals, which promotes CXCR4+ HSC recruitment from PB to the site of injury.53 A multipronged neuroprotective and/or neurorestorative set of events closely precede HSC mobilization to the ischemic site, notably neoangiogenesis that parallels the therapeutic window of G-CSF treatment for stroke therapy.60 The critical role of angiogenesis in HSC fate following ischemic injury is further supported by the observation that systemic administration of human CD34+ cells to SCID (severe combined immunodeficiency) mice exposed to stroke 48 h earlier induces neovascularization in the ischemic zone,61 thereby creating a conducive microenvironment for survival of both exogenous grafts and endogenous stem cells, which are pivotal for neuronal regeneration. In addition to stroke, HSC mobilization to the brain, which may correspond to early host endogenous repair mechanism, is also seen in other neurological diseases; for example, elevated number of human BM CD34+ cells accompany patients with chronic spinal injury.62 Similarly, not only BM-derived, but also cord blood (CB)-derived CD34+ cells have replicated the homing event and subsequent therapeutic benefits of HSCs in brain disorders. Human CB CD34+ cells injected to rats before63 or during heat stress64 significantly reduce symptoms of heatstroke and increase animal survival time by attenuating inflammatory, coagulatory and multiorgan dysfunction; transplantation of human CB CD34+ enriched cells into the injured spinal cords of rats produces a significant recovery of functional outcome and increases survival rate;65,66 and injection of human CB mononuclear cells to mice with amyothropic lateral sclerosis delays disease progression and increases lifespan.67 Recent studies have shown that systemic injections of human CB mononuclear cells in Alzheimer’s disease animal model decrease parenchymal and cerebral vascular β-amyloid deposits, and increase microglial phagocytic activity,68 whereas their transplantation in aged rats significantly enhances the hippocampal neurogenic niche characterized by rejuvenation of the aged neural stem/progenitor cells.69 Taken together, the experiments above lend support to the notion that human HSCs play key regulatory roles in the maintenance of homeostasis and the repair of the nervous system. The crosstalk between the hematopoietic and the nervous system, and the resulting HSC mobilization to the site of injury and subsequent observation of therapeutic benefits, suggest their potential application for designing treatment strategies that cater to regenerative medicine. HSCs have been proposed to be an ideal donor graft source for cell therapy, especially those derived from BM, because of their safety and efficacy profile in the clinic for other disease indications such as for treatment of cardiovascular, bone, cartilage, bladder and liver dysfunctions.7072

Mesenchymal stem cells

MSCs were initially reported by Friedenstein et al.,73 referring to a population of plastic-adherent fibroblastic cells isolated by Percoll density centrifugation (see Table 1). Human MSCs (hMSCs) express CD105 (SH2), SH3, Stro-1 and CD13, but lack the hematopoietic surface markers CD34 and CD45. MSCs have the potential to differentiate into mesodermal cell lineages such as adipocytes, chrondroblasts, fibroblasts, osteoblasts and skeletal myoblasts both in vitro and in vivo.20,7477 hMSCs lack telomerase activity, with about 18 population doublings (PDs).18 A study conducted at the single cell level by Muraglia et al.20 demonstrates MSCs, with capability of differentiation into osteogenic, chondrogenic and adipogenic phenotypes up to 19 PDs, to lose their apparent proliferation potential at 22 to 23 PDs in ~80 days in culture. Strikingly similar to this study, clonal hMSCs cultured in vitro by Banfi et al.21 are also found to gradually lose the differentiation potential when reaching 22 to 23 PDs after ~80 days of culture. The study of Kobune et al.23 reveals that MSCs exhibit a reduced mitogenic activity after ~5 PDs over the course of ~6 weeks and undergo crisis at 16 PDs. Bruder et al.19 demonstrate that the growth rate of MSCs decreases with passaging, in that the PDs for hMSCs before degenerating are around 38, even though the osteogenic differentiation potential is preserved. The cumulative population doubling level for hMSCs selected with serum-deprived medium is ~9 PDs at passage 10, whereas for hMSCs selected with regular medium, the cumulative population doubling level is ~7 PDs at passage 8.22 Although evidence on the limitation of the differentiation and proliferation potential of MSCs exists, a report from Pittenger et al.78 indicates that MSCs maintain telomerase activity in both early and late passage. The discrepancy here might be due to the fact that cell phenotypes isolated from different laboratories are distinct. Gene manipulation strategy has been utilized to overcome the senescent problem of MSCs. Hamada et al.7981 transfected MSCs with the human telomerase gene by retroviral infection to generate stable cell clones with high efficiency and low cell mortality. These cells, regarded as human telomerase reverse transcriptase (hTERT)-MSCs, can survive in culture for over 1 year and maintain their characteristic surface antigens as well as typical morphology. Cerebral infarction volume is significantly reduced and functional outcome is improved in hTERT-MSCs-treated stroke animal model.80

MSC transplantation in stroke

MSCs have been employed in experimental stroke models and shown to improve the functional recovery of neurological deficits induced by cerebral ischemia.8291 Clinical reports of MSC transplantation in stroke patients reveal that MSCs significantly improve the functional recovery of patients without adverse side effects.82 The underlying mechanism remains unclear. The subsequent sections address some potential mechanisms that might mediate the therapeutic effects of MSC implantation in stroke.

MSC-mediated trophic factor secretion

The neuronal differentiation as the underlying mechanism for grafted MSCs to produce therapeutic benefits remains controversial. Moreover, transplantation of MSCs, via intravenous, intracarotid, or even intracerebral delivery, leads to very low, or at best modest, graft survival rate.92 A far more reasonable explanation for the graft-derived beneficial effects is that MSCs secrete neurotrophic factors that may induce the host ischemic brain to activate endogenous repair mechanisms. Chopp et al.93 show that MSCs produce hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF),94 nerve growth factor,95 brain-derived neurotrophic factor (BDNF),93,95 basic fibroblast growth factor (bFGF, FGF-2)96 and insulin growth factor-1 (IGF-1).97 These neurotrophic cytokines have been implicated to play an important role in the process of neurogenesis and angiogenesis. For instance, the upregulation of VEGF and the VEGF receptor 2 (VEGFR2) at the impaired site could increase the number of enlarged and thin-walled blood vessels as well as newly formed capillaries at the ischemia border zone (IBZ).47 Although the early increase (1 h after stroke) of VEGF could increase the blood–brain barrier (BBB) leakage and hence increase ischemia hemorrhagic transformation, and exacerbate ischemic cell damage, when administered 48 h after stroke, VEGF could enhance angiogenesis in the IBZ and significantly improve neurological recovery.98 From hMSC administration to the secretion of VEGF by the graft cells, the time course highly exceeds 1 h after stroke; hence, VEGF here functions as the angiogenesis promoter and improves the functional recovery after stroke.

HGF has been demonstrated to ameliorate BBB destruction without exacerbating cerebral edema, decrease intracranial pressure and induce angiogenesis. Equally compelling evidence suggests that grafted MSCs promote neurogenesis. Over the years, laboratory studies have established the subventricular zone (SVZ) and subgranular zone as remarkable brain neurogenic sites. The presence of MSCs in the brain has been shown to promote the induction and migration of new cells from these primary sources within the SVZ and the subgranular zone into the injured brain.86,95,96 The newly differentiated cells appear as progenitor-like neurons and astrocytes. The process of neurogenesis is highly dependent upon the neurotrophic factors secreted by MSCs.96 Upregulation of BDNF levels is proven to be capable of recruiting neural progenitors from the endogenous progenitors of the forebrain.99 Recent reports show that bFGF is capable of exerting neuroprotective or vasodilating effects, as well as enhancing the proliferation of the neuronal progenitors.100,101 To enhance the efficacy of MSCs, gene-modified MSCs have been used as donor cells for transplantation in stroke models. In these experiments, specific genes, such as FGF-2/HGF/BDNF, are transfected into the cells before they are transplanted into the brain. These grafted cells subsequently are able to express the target gene in the brain. Kurozumi et al.81,102 transfected telomerized hMSCs with the BDNF gene using a fiber-mutant F/RGD adenovirus vector and transplanted these cells in stroke rats. BDNF production by MSC-BDNF cells is 23-fold greater than that seen in unmodified MSCs. Stroke rats that received MSC-BDNF displayed significantly better functional recovery than control stroke rats. This research group also conducted a parallel study in which they transfected MSCs with glial cell line-derived neurotrophic factor (GDNF), ciliary neurotrophic factor (CNTF) and neurotrophin 3 (NT3) genes.81 Stroke rats that received MSC-GDNF, but not those treated with CNTF and NT3, showed significantly greater functional improvement than control stroke rats. A study by Ikeda et al.103 demonstrated that stroke animals that received bFGF-MSCs showed significantly better attenuations of behavioral impairments and infarction volumes than those that received nontransfected MSCs. These data suggest that bFGF enhances the beneficial effects of MSCs.

A relatively new addition to the list of growth factors exerting benefits in stroke is HGF. Ischemia animals treated both with and without HGF-modified MSCs exhibit improvement of neurological deficits, yet those that received HGF-MSCs treatment display superior therapeutic effect to the nonmodified MSC-treated group.104 Although graft survival rate or neuronal differentiation appear to play minor roles in the MSC transplantation therapeutic effect, studies demonstrate that transplantation with MSCs that are committed to differentiate into a neuronal phenotype indicate better therapeutic effects. MSCs transfected with Notch intracellular domain and subsequently treated with bFGF, forskolin and CNTF have the potential to differentiate into neurons, termed by the authors as BM stromal cell-derived neuronal cells.105 Transplantation of BM stromal cell-derived neuronal cells in stroke models reveals better functional improvement than those treated with MSCs.105 Although gene-based strategies appear to improve MSC graft functional effects, the use of viral vectors poses clinical problems, especially with uncontrolled gene replications that may cause neoplasm, tumors and even death. An alternative approach to circumvent the adverse side effects associated with genetic manipulation, while still facilitating the therapeutic effects of MSCs, is to exogenously deliver neurotrophic factors. A study shows that BDNF delivered along with MSCs significantly improves the recovery of the stroke animals.106 In an effort to further improve MSC efficacy, MSC transplantation is combined with adjunctive treatment of specific reagents. A cell-permeable inhibitor of caspases, Z-Val-Ala-DL-Asp-fluoromethylketone (Z-VAD), is found to enhance graft survival and behavioral recovery107 when intracerebrally infused together with MSCs into the ischemic region. Nitric oxide has been proven to play an important role in cell proliferation and neurogenesis. Intravenous infusion of MSCs with a nitric oxide donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl) aminio] diazen-1-ium-1,2-diolate (DETA/NONOate), significantly improves functional recovery in stroke animals, with accompanying enhancement of vessel perimeter and endothelial cell proliferation. In addition, such combined DETA/NONOate and MSC grafts lead to increased neurogenesis in the SVZ, as well as VEGF and bFGF expression, in the ischemic area compared with single treatments.108

MSC-induced cell proliferation and axonal remodeling

Grafted MSCs could modulate glial cell proliferation and result in neuron remyelination as well as synaptogenesis.109 Oligogenesis and astrocytogenesis are markedly enhanced in the SVZ of the ischemia animal models treated with MSCs.92,109111 In addition to enhancing gliogenesis, reducing apoptosis is shown to be another critical effect of MSCs. Using an anaerobic chamber to duplicate the in vivo ischemia scenario in vitro, Gao et al.112 demonstrate that astrocytes, when cocultured with MSCs, suffer less cell death and apoptosis. In vivo studies indicate that intravenous administration of MCSs in stroke models could reduce apoptosis in the penumbral zone of the lesion.95,96 A recent study demonstrates that MSCs significantly increase astrocytic expression of connexin-43 (Cx43)57 and growth associated protein-43 (GAP-43).110 This Cx43 upregulation is concomitant with altered gap junction intercellular communication with the participation of phosphatidylinositol 3-kinase signaling pathway.113 In vivo data confirm that transplanted MSCs enhance bone morphogenetic protein 2/bone morphogenetic protein 4 (BMP2/4) and Cx43 expression in astrocytes.114 Cx43 is the primary component of intercellular channels in astrocytes, whereas BMP2/4 belongs to a subgroup of the transforming growth factor-β superfamily. BMP2/4 maintains extensive gap junctional communication through Cx43 and hence mediates communication between astrocytes and increases synaptogenesis in the IBZ.114 Compared with nontransplanted ischemic animals, areas of the corpus callosum and the numbers of white matter bundles in the striatum are larger in the IBZ in MSC-treated ischemic animals;109,115 at the same time, the axons and myelin thickness are increased along with scar thickness being reduced.92,110,111 In contrast, the numbers of microglia and macrophages within the scar wall are reduced.110 Whether microglia inhibit the axonal regeneration warrants further investigation.

MSC homing via SDF-1/CXCR4 chemoattractant pathway

As discussed above, SDF-1 is a crucial chemoattractant for CXCR4-expressing BM-derived cells in the hematopoietic system (that is, HSCs). In the nonhematopoietic system, CXCR4-positive cells similarly respond to SDF-1 signals secreted by injured organs, and migrate into these areas. Such SDF-1 upregulation and cell migration are found to subsequently lead to upregulation of various trophic factors either secreted by the mobilized stem cells or by endogenous cells. In stroke rats, the chemokine SDF-1 level is significantly increased in the injured hemisphere compared with the spared site.92 The upregulation of SDF-1 is mostly recognized in the IBZ and maintained for up to 30 days after the injury. Of note, CXCR4 is detected in the transplanted MSCs. The interaction between SDF-1 and CXCR4 serves as the chemoattractant guide for MSC migration toward the impaired site.92 SDF-1 gene expression is regulated by the hypoxia-responsive transcription factor, hypoxia-inducible factor 1,116 and follows a gradient pattern consistent with the hypoxia gradient in the penumbra of stroke animals. In general, the expression of SDF-1 following a brain injury serves as a significant signal for attracting the BM-derived stem cells to migrate into the damaged area, and is crucial for the functional recovery of stroke.

With MSCs being one of the most studied stem cells for therapeutic applications, although their mechanisms of action are not well understood, two recent Food and Drug Administration (FDA)-approved clinical trials have been initiated via intravenous autologous MSCs (University of Texas at Houston with Dr Sean Savitz) and stereotactic transplantation of allogeneic Notch-induced MSCs (Stanford University with Dr Gary Steinberg) in acute and chronic ischemic stroke, respectively.

Endothelial progenitor cells

EPCs, initially described by Asahara et al.,24 are immature endothelial cells that circulate in PB (see Table 1). EPCs mature as endothelial cells, and are important components of the vascular system.2529 In their pioneering study,24 transplanted EPCs, isolated from human umbilical cord blood (UCB), were found in the endothelium of newly formed vessels in ischemic regions, indicating that a discrete cell population within the human blood participates in the formation of new vessels after ischemia. Griese et al.117 also found that grafted EPCs populated the endothelium in animals with experimentally induced endothelial damage,118 further advancing the notion that EPCs contribute to the repair of damaged endotheliumx. Although EPCs are hematopoietic in origin and they can be found in PB of adults, a population of cells with similar characteristics can also be derived from human UCB.117,119 Several lines of investigations from both animal and human studies indicate that EPCs principally participate in re-endothelialization during the neovascularization of ischemic organs, suggesting that EPC modulation could be directed toward treatment of cerebrovascular diseases.25,26,29,30,117,119,120

EPC phenotypic characterization

Immunological surface markers and functional profiling via colony-formation capacity of these cells have been employed to determine their characteristics.3034 EPCs were first isolated from human PB, which expressed shared markers with HSCs, angioblasts and receptors for VEGFR2/KDR.24 Cultured EPCs could differentiate into endothelial cells and incorporate into sites of active angiogenesis in animal models of ischemia.26 Isolation of EPCs remains controversial, in that although different markers such as CD31, VE-cadherine, E-selectin, endothelial nitric oxide synthase and von Willebrand factor among others26,30,33,34,121 have been employed to harvest EPCs, equally solid evidence indicates that only CD34-positive EPCs isolated from BM or UCB possess the potency to differentiate into mature endothelial cells.29,117,122 In parallel, recent studies have used the HSC-specific AC133 as surface marker to isolate EPCs.30,31 Varying developmental stages of EPCs or the presence of residual cells derived from the mature vascular wall may influence the heterogeneity of tissue source of EPCs. The prevailing marker of choice for obtaining highly homogenous EPCs is via double labeling with CD34/VEGFR2.30,31,35

To further define the phenotypic feature of EPCs, another approach via functional profiling has been used that involves counting the number of EPC colonies formed after 7 days of culture. A colony consists of a central cluster of rounded cells with surrounding radiating thin, flat cells.26,123125 These colonies exhibit many endothelial characteristics including expression of CD31, VEGFR2 and Tie-2.26,125 Another method is to measure the uptake of Dil-labeled acetylated low-density lipoprotein or binding to specific lectins. This method has also been useful in defining endothelial cells; however, these characteristics are also displayed by most macrophages in a culture.26,32,126,127 These characteristics are mostly considered to be functional in nature and do not represent the number of EPCs present in the culture. Several sources of EPCs have been identified, including PB, BM and UCB.25,26,28,29,117 Recently, it has been reported that EPCs may even originate from the area between smooth muscle and adventitial layer of the human adult vascular wall.128 EPCs remain extremely rare in adult PB (0.01% of mononuclear cells, under-steady state conditions), which may be a contributing factor for the lack of clearly defined methods for cell isolation and definition.32 Whereas BM-derived stem/progenitor cells have been widely considered as a source of EPCs, the BM represents heterogeneous groups of cells with at least two major progenitor populations, namely MSCs and hematopoietic EPCs, with both populations capable of inducing neovascularization through vasculogenesis, making them good candidates for cell therapy in cerebrovascular diseases.117,129135 Although characterization of EPCs remains a challenging cell culture protocol, the unique properties of these cells as recommended by Finkel and co-workers122 may suffice as a starting point in delineating this novel BM cell population. In particular, EPCs are circulating, BM-derived cells distinct from mature endothelial cells due to their ability to differentiate into endothelial cells, as assessed by expression profiles and functional characteristics; and their regenerative capacity, especially for promoting vasculogenesis and/or vascular homeostasis.136

EPC, angiogenesis and vasculogenesis

The dogma that existed until recently is that neovascularization, or formation of new blood vessels, results exclusively from proliferation and migration of pre-existing endothelial cells, a process referred to as angiogenesis.137 Furthermore, vasculogenesis or vascularization, defined as in situ differentiation of vascular endothelial cells from endothelial precursor cells, was thought to occur only in the embryo during vascular development. However, recent evidence has now established that circulating BM-derived EPCs are capable of homing to neovascularization sites, proliferating and differentiating into endothelial cells.138,139 EPCs have been identified mainly in the mononuclear cell fraction of PB, leukapheresis products and in UCB,140,141 but can also be harvested from BM as presented in our preliminary data section. Over the last few years, EPCs have been studied as biomarkers to assess the risk of cardiovascular disease in human subjects. For example, a low EPC count predicts severe functional impairments in several cardiovascular pathologies such as diabetes,142 hypercholesterolemia,143 hypertension,144,145 scleroderma,146,147 aging,142,148 cigarette smoking142,149,150 and coronary artery disease.151 In addition, EPCs have been examined as potent donor graft cells for transplantation therapy. Transplantation of EPC into ischemic tissues has emerged as a promising approach in the treatment of diseases with blood vessel disorders.152154 In mouse models of ischemic injury, EPC injection led to improved neovascularization in hind limb ischemia.152154

EPCs for cell therapy

Based largely on these laboratory findings suggesting angiogenic and vasculogenic potential of EPCs, clinical studies have been initiated to reveal whether patients with lower EPC numbers are at higher risk for atherosclerotic events, and whether patients with ischemic events may benefit from EPC administration.155 Clinical studies to date suggest the therapeutic potential of EPC transplantation, although this assumption should be approached with much caution due to these studies being open-label trials, observational and/or anecdotal accounts having a limited number of patients. Ex vivo expanded EPCs, isolated from PB mononuclear cells, can incorporate into the foci of myocardial neovascularization,156,157 and intracoronary infusion of PB or BM-derived progenitors in patients with acute myocardial infarction was associated with significant benefits in post-infarction remodeling.158165 Still in observational studies in patients with myocardial infarction, higher numbers of EPCs correlate with better prognosis, more myocardial salvage,166 viability and perfusion167 and more collaterals in the ischemic zone.168 Randomized clinical trials on autologous BM-derived cells are mixed; whereas transplanted coronary artery disease patients display improved left ventricular function at least in the short term,169 transplanted patients with chronic ischemic heart failure exhibit modest to no effects on change in left ventricular function.170 Similar randomized trials of autologous BM-derived cells have been carried out in patients with peripheral artery disease and have shown improved endothelium-dependent vasodilation,171 ankle brachial index, rest pain and pain-free walking time,172 but the degree of functional recovery was not as robust as seen in animal models. Clearly, these results are obtained from autologous BM-derived cells, which are heterogeneous with scarce number of EPCs, and thus may not closely approximate EPC end points.

For clinical application of EPCs in neurovascular disease, the available studies are much more limited, with only three observational studies in patients with stroke. In 25 patients with an ischemic stroke, CD34+ cells peaked 7 days after stroke but generally reverted to baseline after 30 days.173 Interestingly, higher CD34+ cell levels at 30 days related to higher numbers of infarcts on magnetic resonance imaging and also to cerebrovascular function as measured with positron emission tomography scanning (cerebral metabolic rate of oxygen and cerebral blood flow). On the other hand, decreased numbers of clusters of rapidly adhering cells were seen after stroke and in ‘stable cerebrovascular disease,’ compared with controls free of vascular disease.174 Higher age and the presence of cerebrovascular disease in general independently related to lower EPC numbers. The discrepancies in the results of these studies may be because of mismatched controls for age of patients and/or the lack of methodological design for testing specific hypotheses on the causal role of EPCs in cerebrovascular disease.174

Although the primary mitigating mechanisms underlying stroke pathogenesis and its abrogation by cell therapy are still uncertain, there is substantial evidence implicating immunological attack upon the brain and/or its vasculature; widespread inflammatory reactions in stroke may trigger a cascade of events that alter the integrity of the BBB, resulting in migration of leukocytes into the CNS. Leukocyte transmigration across the BBB during stroke-mediated immune/inflammatory processes could influence interendothelial junctional complex function, leading to vascular endothelium damage and BBB breakdown. Equally a key component to our mechanism-based hypothesis is that disruption or dysfunction of the BBB, preceding entry of harmful substances into the brain parenchyma, could be a key initial factor in stroke pathogenesis. Thus, restoration of barrier integrity may have a critical role in preventing stroke progression, highlighting the need for EPCs as transplantable cells for stroke therapy.

Very small-like embryonic stem cells

As mentioned above, stress conditions such as ischemic injury acutely increases the number of various types of stem cells in PB in both experimental animals and patients.175178 Stem cells, including HSCs,179 MSCs180 and EPCs,181 are mobilized into PB from BM and probably other nonhematopoietic niches as well, likely contributing to an endogenous regenerative medicine.177,178,182,183 However, as brain damage after stroke still leads to irreversible brain damage, these cells circulating in PB are not sufficient to halt the stroke damage. A hostile microenvironment ensues during stroke progression that alters chemotaxis and homing of circulating PB cells to the ischemic brain. In addition, the BBB may limit the entry of circulating stem cells from periphery to the brain, thereby further blocking the therapeutic benefits of this endogenous repair mechanism. Treatment strategies designed to increase the number of circulating stem cells in PB (that is, after administration of mobilizing agents such as G-CSF and/or CXCR4 antagonists) maintain the chemoattractant pathway to provide migratory cues for circulating stem cells to home to the site of injury, and enhance permeability of the BBB to facilitate stem cell entry into the brain, or stereotactic delivery of stem cells into the peri-infarct area may foster an effective regenerative regimen against stroke.184187 Along this line, the acute endogenous stem cell mobilization following ischemic injury can be enhanced in both mice and humans after G-CSF treatment during acute cardiovascular diseases, for example, myocardial infarction and stroke.178,182 Accordingly, stem cell-based strategies aimed at regeneration of the stroke brain stands as a potent therapeutic modality. In the study of Ratajczak et al.,177 the research team recently discovered a unique population of stem cells called VSELs. Of note, the number of circulating VSELs in PB increases in mice after experimental stroke177 as well as in stroke patients,178 suggesting that VSELs residing in adult tissues or mobilized into PB are a potent source of adult tissue-derived stem cells that can be utilized for regenerative medicine specifically for neural repair after stroke.

In the pioneering studies by Ratajczak et al.,177,188 they showed that VSELs could be mobilized into PB in patients after stroke, in a similar way as in patients after acute heart infarct in humans and mice. By employing a staining strategy, they observed an increase in mRNA for both pluripotent (Oct-4 and Nanog) and neural (GFAP, Nestin, b-IIItubulin, Olig1, Olig2, Sox2, and Musashi-1) stem cell markers in PB-borne nucleated cells circulating in stroke patients, resembling the general phenotypic patterns that they previously noted in an experimental murine model of stroke,177 with the exception that maximally increased expression of neural stem cell markers in humans was delayed by 2 days (that is, 1 day for mice vs 3 days for humans). Further analyses using computer tomography scans revealed differences in VSEL mobilization in that patients with posterior circulation infarcts have the best chance of recovery, whereas partial anterior circulation infarcts are associated with the highest risk of early recurrence of stroke (that is, within 3 months), although not associated with high mortality and significant disability. Whether stroke morbidity or mortality correlates to location of the stroke area in relation to the neurogenic regions of SVZ and subgranular zone warrants further examination. Additionally, whether the number of these cells could be increased by administration of mobilization-promoting agents (for example, G-CSF or AMD3100) represents a logical extension of this therapy for clinical application. Nonetheless, these studies indicate that the mobilization of VSELs and tissue-committed progenitor cells expressing early neural markers into PB are closely associated with ischemic stroke and could be utilized as a prognostic tool.189,190

Phenotypic characterization and genomic imprinting of VSELs

VSELs express several progenitor stem cell markers38 (see Table 1), and are present in a variety of adult organs; specifically, the brain contains a relatively high number of cells that display the VSEL phenotype.38,39 Initially separated from murine BM,40 VSELs are smaller than erythrocytes (3–5 µm in diameter in mice) with a corresponding human population of small cells (5–7 µm in diameter) purified from human UCB and mobilized PB.38 In the Ratajczak laboratory, well-validated protocols for VSEL identification in human PB using flow cytometric methods have been established. Following tissue and organ injuries, human VSELs are mobilized into PB of patients and recognized as very small cells belonging to the nonhematopoietic fraction of leukocytes (Lin−/CD45− cells) expressing CD34, CD133 and CXCR4 antigens.42,177 The absolute numbers of circulating VSELs in PB are exceptionally low (1 to 2 cells in 1 ml of blood under steady-state conditions) and thus special flow cytometric protocols have to be applied for their identification. Phenotypic markers used to identify VSELs include negative expression of CD45 (mouse and human), positive expression of Sca-1 (mouse), CXCR4, CD133 and CD34+ (mouse and human), positive for progenitor stem cell markers (that is, Oct-4, Nanog and SSEA),38,4143 and express several markers characteristic of epiblast/germ line stem cells.44 Based on a multianalytical flow cytometric approach, the identity and confirmation of the presence of VSELs among blood leukocytes, in addition to a quantitative determination of the absolute numbers of these rare cells circulating in the blood of patients with various tissue/organ injuries and disorders, could be ascertained.41,177 An independent group has successfully purified VSELs from human mobilized PB as well as BM.45

In deciphering the genomic imprint of VSELs, a notable feature is that most of the homeodomain-containing developmental transcription factors in VSELs are repressed by specific epigenetic marks called bivalent domains that represent a state of the DNA structure characteristic of progenitor stem cells, where transcriptionally antagonistic histone codes physically coexist within the same promoter.190 Murine Oct-4+ VSELs do not proliferate spontaneously in vitro if cultured alone and that the quiescence of these cells is regulated by genomic imprinting through DNA methylation, which is an epigenetic program that ensures the parent-specific monoallelic transcription of some developmentally important genes such as those implicated in embryogenesis, fetal growth, maintaining the totipotential state of the zygote and maintaining the pluripotency of developmentally early stem cells.43 The expression of imprinted genes is regulated by DNA methylation with freshly isolated VSELs from murine BM deleting the paternally methylated imprints, but hypermethylating the maternally methylated imprints. Because paternally expressed imprinted genes (Igf2 and Rasgrf1) enhance embryo growth, whereas those that are maternally expressed (H19, p57KIP2, and Igf2R) inhibit cell proliferation,43 the unique genomic imprinting pattern observed in VSELs demonstrates a growth-repressive program in this developmentally early stem cell. Although the quiescent pattern of genomic imprinting is largely influenced by DNA methylation, cell culture conditions such as the formation of spheres by VSELs in cocultures with myoblastic C2C12 cells191 could prime VSELs to proliferate and differentiate. Taken together, these findings suggest that the epigenetic reprogramming of genomic imprinting is capable of maintaining quiescence of the most primitive pluripotent adult stem cells (that is, Oct-4+ VSELs) deposited in the adult body and protect them from premature aging and tumor formation, but that specific external microenvironmental signals can influence the epigenetic state of this imprinting and force VSELs to differentiate.191 Whether human VSELs, as well as for cells isolated from nonhematopoietic tissues (for example, the CNS) that display the VSEL phenotype, also recapitulate such DNA methylated genomic imprint remains to be determined.

VSELs for cell therapy

The notion that VSELs are epiblast-derived progenitor stem cells deposited early during embryonic development in immature organs, as a potential reserve pool of precursors for tissue-committed stem cells, suggests that this population has an important role in physiological tissue rejuvenation and regeneration. The observation that murine VSELs have the potential to differentiate into neurons, oligodendrocytes and macroglia further advances the utility of these cells as donor grafts for regeneration of a damaged CNS. In contemplating with clinical application of the cells, the ease of harvesting VSELs should be considered. With autologous transplantation in mind, the patient’s own BM, stored UCB and mobilized PB appear as readily accessible sources of VSELs for regenerative medicine. For allotransplantation, VSELs could be harvested from histo-compatible-related or unrelated donors as is currently done with HSC allotransplantation.191 A potentially limiting factor for clinical application is the small number of VSELs that could be harvested, requiring the need for efficient ex vivo expansion strategy, especially if the goal is to generate an ample supply of neural stem cells for stroke therapy. Another caveat to pursuing VSELs in the clinic is the observation that the number of VSELs decreases with the age, implying that enhanced regeneration potential of human patients may be correlated with higher number of these cells deposited during embryogenesis in the adult tissues.192 Of note, calorie uptake-related increase in plasma level of insulin and insulin-like growth factors may deplete VSELs in adulthood.193

Isolation of VSELs from BM, UCB and PB

In designing treatment strategies for stroke, the acute and subacute stage (time 0 to 1 week post injury) seems to offer the best opportunity to initiate the therapeutic intervention in order to immediately abrogate the rapidly deteriorating ischemic brain. Because of this limited therapeutic window, it may not be feasible and practical to purify these rare cells from BM aspirates, UCB or mobilized PB by employing multiparameter staining and regular high-speed sorting only. The Ratajczak group calculated that by employing only one cell fluorescence-activated cell sorter, isolation of all VSELs present in 100 ml of UCB would take up to 4 working days.194,195 For efficient cell isolation, they propose a relatively short and economical three-step isolation protocol that allows recovery of ~60% of the initial number of Lin−/CD45−/CD133+ UCB− VSELs. The rationale for this novel strategy takes into account lysis of erythrocytes in a hypotonic ammonium chloride solution, CD133+ cell selection by immunomagnetic beads and sorting of Lin−/CD45−/CD133+ cells by FACS with size-marker bead controls. The entire isolation procedure takes 2 to 3 h per UCB unit (but should also be applicable using BM aspirates and mobilized PB) and isolated cells are highly enriched for an Oct-4+ and SSEA-4+ population of small, highly primitive Lin−/CD45−/CD133+ cells. The envisioned clinical product will be VSELs freshly isolated from BM, PB or UCB or VSELs precommitted to the neurological lineage in ex vivo cultures.190

Conclusions

The discovery of stem cell plasticity in adult tissues has been a major driving impetus for advancing regenerative medicine in many neurological disorders, including stroke. BM-derived HSCs, MSCs, EPCs and VSELs have been proposed as potential sources of adult stem cells for regeneration of the CNS. However, the rationale for employing these non-neuronal stem cells remains controversial, and most recently MSCs have been put into the spotlight as a possible cause of tumor formation following their deposition into the brain.196 Whereas BM-, PB- or UCB-derived HSCs for regeneration of neural tissues have been explored, the postulated stem cell plasticity of HSCs, such that these cells may become neural stem cells,197 has been not confirmed in recently published studies. On the contrary, the transdifferentiation may be explained as a transient change in HSC phenotype induced by neural tissue-derived, spherical membrane fragments called microvesicles (or exosomes) that may transfer neural cell-surface receptors, mRNA and microRNA to the HSCs employed for regeneration.198 Along similar technical obstacles as HSCs, MSCs could be isolated from BM, PB or UCB by expansion of an adherent population of fibroblast-like cells,199 and it is widely accepted that MSCs contribute to the regeneration of mesenchymal tissues (that is, bone, cartilage, muscle, ligament, tendon, adipose and stromal support). MSCs could also be obtained from several other tissues (for example, PB, UCB, adipose tissue, dental pulp and menstrual blood).200,201

An unexpected discovery from a few years ago suggested that MSCs are able to give rise to neuronal cells.202 Like HSCs, this rare transdifferentiation event of MSCs was challenged, pointing to the possibility of an in vitro contamination in the cell culture media203,204 that could alter the morphology of MSCs. Under these same cell culture conditions, fibroblasts could shrink, elongate and mimic neurons, but this proved to be only an in vitro morphological artifact.203,204 Thus, the rationale behind applying MSCs, just like HSCs, in brain regeneration is not well supported. On other hand, there is no doubt that modest functional recovery occurs in animal models and patients after treatment with HSCs or MSCs.184186 The mechanism of action, however, likely does not involve differentiation of these cells into neurons, because such cells are rapidly eliminated after local delivery.184186 Instead, the most plausible explanation currently receiving much support from the literature is that HSCs or MSCs employed for regeneration could, however, propel a by-stander effect in that release of growth factors or cytokines facilitates neovascularization of damaged tissues leading to neurogenesis, as well as afford anti-inflammatory, antiapoptotic and antioxidative stress effects among other reparative responses.40,205207

More recently, my group has taken a keen interest in repairing the BBB after stroke via EPC transplantation. Our overarching hypothesis is that BBB breakdown accompanies stroke and may be exacerbated by tissue-type plasminogen activator. To date, most stroke therapies have not considered the repair of this BBB damage after stroke. If BBB restoration via EPC transplantation alone or in combination with tissue-type plasminogen activator is proven effective, we believe that direct clinical application of this cell therapy will be far reaching, as this treatment could help a large population of ischemic stroke patients who otherwise would have missed the limited 3-h tissue-type plasminogen activator window. In addition, we envision that this EPC transplantation can supplement other stroke therapeutics that require BBB manipulation in order to afford beneficial effects, and can be extended to other neurological disorders characterized by BBB breakdown.

Finally, the advent of VSELs is equally appealing for cell therapy in stroke. A major caveat in delivery of stem cells into the brain, whether via stereotactic local implantation or peripheral delivery (that is, intravascular or intraarterial), is the possibility of creating embolism with the high number of stem cells required to exert therapeutic effects. In this regard, the very small size of VSELs is one of the most unique properties of the cells that could avoid the potential adverse effects of embolism associated with cell therapy. That the VSEL isolation and expansion may require tedious steps and longer days to acquire ample supply of the cells can be circumvented by pursuing allogeneic overautologous transplants. As stroke episodes cannot be predicted, an off-the-shelf treatment intervention is likely optimal; thus, allogeneic transplant therapy may very well be more suitable than autotransplantation. Moreover, that aging or disease process entails less number of viable and healthy stem cells implies that the harvest of VSELs from young donors (that is, PB) represents an advantageous cell source.

In summary, the encouraging laboratory results of cell therapy using HSCs, MSCs, EPCs and VSELs as donor sources are rapidly being translated into clinical trials. Over the past 5 years, recommendations to better guide the successful entry of stem cell therapy into the clinic have received continuing critical assessments from unique collaborative meetings involving thought leaders from academe, industry, the National Institutes of Health (NIH) and the FDA.68 The mechanism of action underlying cell therapy remains to be fully determined, and this persisting gap in our knowledge should be seriously considered when evaluating the risk-to-benefit ratio of this novel, experimental treatment for stroke. In the end, systematically designed translational preclinical studies need to precede initiation of clinical trials in order to allow rigorous investigations of the safety and efficacy profile of these adult BM-derived stem cells for cell therapy in stroke and other neurological disorders.

Acknowledgements

I extend appreciation to Ms Loren E Glover who provided excellent technical assistance in the final preparation of the manuscript. CVB is supported by James and Esther King Foundation for Biomedical Research Program 1KG01-33966 and NIH R01 5R01NS071956-02, and received research grant support for his projects on bone marrow stem cell therapy for stroke from SanBio Inc., Celgene Cellular Therapeutics, KMPHC and Neural- Stem Inc. Some of the stem cell therapy thematic discussions originated from NINDS UO1 5U01NS055914-04.

Footnotes

Conflict of interest

The author declares no conflict of interest.

References

  • 1.Herzog EL, Chai L, Krause DS. Plasticity of marrow-derived stem cells. Blood. 2003;102:3483–3493. doi: 10.1182/blood-2003-05-1664. [DOI] [PubMed] [Google Scholar]
  • 2.Munoz-Elias G, Woodbury D, Black IB. Marrow stromal cells, mitosis, and neuronal differentiation: stem cell and precursor function. Stem Cells. 2003;21:437–448. doi: 10.1634/stemcells.21-4-437. [DOI] [PubMed] [Google Scholar]
  • 3.Hess DC, Borlongan CV. Cell-based therapy in ischemic stroke. Expert Rev Neurother. 2008;8:1193–1201. doi: 10.1586/14737175.8.8.1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hara K, Yasuhara T, Maki M, Matsukawa N, Masuda T, Yu SJ, et al. Neural progenitor NT2N cell lines from teratocarcinoma for transplantation therapy in stroke. Prog Neurobiol. 2008;85:318–334. doi: 10.1016/j.pneurobio.2008.04.005. [DOI] [PubMed] [Google Scholar]
  • 5.Hess DC, Borlongan CV. Stem cells and neurological diseases. Cell Prolif. 2008;1:94–114. doi: 10.1111/j.1365-2184.2008.00486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chopp M, Steinberg GK, Kondziolka D, Lu M, Bliss TM, Li Y, et al. Who’s in favor of translational cell therapy for stroke: STEPS forward please? Cell Transplant. 2009;18:691–693. doi: 10.3727/096368909X470883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Stem Cell Therapies as an Emerging Paradigm in Stroke Participants. Stem Cell Therapies as an Emerging Paradigm in Stroke (STEPS): bridging basic and clinical science for cellular and neurogenic factor therapy in treating stroke. Stroke. 2009;40:510–515. doi: 10.1161/STROKEAHA.108.526863. [DOI] [PubMed] [Google Scholar]
  • 8.Borlongan CV, Chopp M, Steinberg GK, Bliss TM, Li Y, Lu M, et al. Potential of stem/progenitor cells in treating stroke: the missing steps in translating cell therapy from laboratory to clinic. Regen Med. 2008;3:249–250. doi: 10.2217/17460751.3.3.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kondziolka D, Wechsler L, Goldstein S, Meltzer C, Thulborn KR, Gebel J, et al. Transplantation of cultured human neuronal cells for patients with stroke. Neurology. 2000;55:565–569. doi: 10.1212/wnl.55.4.565. [DOI] [PubMed] [Google Scholar]
  • 10.Meltzer CC, Kondziolka D, Villemangne VL, Wechsler L, Goldstein S, Thulborn KR, et al. Serial [18F] fluorodeoxyglucose positron emission tomography after human neuronal implantation for stroke. Neurosurgery. 2001;49:586–591. doi: 10.1097/00006123-200109000-00011. [DOI] [PubMed] [Google Scholar]
  • 11.Nelson PT, Kondziolka D, Wechsler L, Goldstein S, Gebel J, DeCesare S, et al. Clonal human (hNT) neuron grafts for stroke therapy: neuropathology in a patient 27 months after implantation. Am J Pathol. 2002;160:1201–1206. doi: 10.1016/S0002-9440(10)62546-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lapidot T, Kollet O. The brain-bone-blood triad: traffic lights for stem-cell homing and mobilization. Hematology Am Soc Hematol Educ Program. 2010;2010:1–6. doi: 10.1182/asheducation-2010.1.1. [DOI] [PubMed] [Google Scholar]
  • 13.Lapidot T, Dar A, Kollet O. How do stem cells find their way home? Blood. 2005;106:1901–1910. doi: 10.1182/blood-2005-04-1417. [DOI] [PubMed] [Google Scholar]
  • 14.Nervi B, Link DC, DiPersio JF. Cytokines and hematopoietic stem cell mobilization. J Cell Biochem. 2006;99:690–670. doi: 10.1002/jcb.21043. [DOI] [PubMed] [Google Scholar]
  • 15.Papayannopoulou T, Scadden DT. Stem-cell ecology and stem cells in motion. Blood. 2008;111:3923–3930. doi: 10.1182/blood-2007-08-078147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zimmermann S, Martens UM. Telomeres, senescence, and hematopoietic stem cells. Cell Tissue Res. 2008;331:79–90. doi: 10.1007/s00441-007-0469-4. [DOI] [PubMed] [Google Scholar]
  • 17.Geiger H, Rudolph KL. Aging in the lympho-hematopoietic stem cell compartment. Trends Immunol. 2009;230:360–365. doi: 10.1016/j.it.2009.03.010. [DOI] [PubMed] [Google Scholar]
  • 18.Zimmermann S, Voss M, Kaiser S, Kapp U, Waller CF, Martens UM. Lack of telomerase activity in human mesenchymal stem cells. Leukemia. 2003;17:1146–1149. doi: 10.1038/sj.leu.2402962. [DOI] [PubMed] [Google Scholar]
  • 19.Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation. J Cell Biochem. 1997;64:278–294. doi: 10.1002/(sici)1097-4644(199702)64:2<278::aid-jcb11>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 20.Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci. 2000;113:1161–1166. doi: 10.1242/jcs.113.7.1161. [DOI] [PubMed] [Google Scholar]
  • 21.Banfi A, Muraglia A, Dozin B, Mastrogiacomo M, Cancedda R, Quarto R. Proliferation kinetics and differentiation potential of ex vivo expanded human bone marrow stromal cells: implications for their use in cell therapy. Exp Hematol. 2000;28:707–715. doi: 10.1016/s0301-472x(00)00160-0. [DOI] [PubMed] [Google Scholar]
  • 22.Pochampally RR, Smith JR, Ylostalo J, Prockop DJ. Serum deprivation of human marrow stromal cells (hMSCs) selects for a subpopulation of early progenitor cells with enhanced expression of OCT-4 and other embryonic genes. Blood. 2004;103:1647–1652. doi: 10.1182/blood-2003-06-1967. [DOI] [PubMed] [Google Scholar]
  • 23.Kobune M, Kawano Y, Ito Y, Chiba H, Nakamura K, Tsuda H, et al. Telomerized human multipotent mesenchymal cells can differentiate into hematopoietic and cobble- stone area-supporting cells. Exp Hematol. 2003;31:715–722. doi: 10.1016/s0301-472x(03)00177-2. [DOI] [PubMed] [Google Scholar]
  • 24.Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of progenitor endothelial cells for angiogenesis. Science. 1997;275:964–967. doi: 10.1126/science.275.5302.964. [DOI] [PubMed] [Google Scholar]
  • 25.Asahara T, Masuda H, Takahashi T, Kalka C, Pastore C, Silver M, et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85:221–228. doi: 10.1161/01.res.85.3.221. [DOI] [PubMed] [Google Scholar]
  • 26.Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–967. doi: 10.1126/science.275.5302.964. [DOI] [PubMed] [Google Scholar]
  • 27.Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625–637. doi: 10.1016/s0092-8674(02)00754-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lin Y, Weisdorf DJ, Solovey A, Hebbel RP. Origins of circulating endothelial cells and endothelial outgrowth from blood. J Clin Invest. 2000;105:71–77. doi: 10.1172/JCI8071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.McCarty JH. Cell adhesion and signaling networks in brain neurovascular units. Curr Opin Hematol. 2009;16:209–214. doi: 10.1097/MOH.0b013e32832a07eb. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lapergue B, Mohammad A, Shuaib A. Endothelial progenitor cells and cerebroascular diseases. Pror Neurobiol. 2007;83:349–362. doi: 10.1016/j.pneurobio.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • 31.Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, et al. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med. 1999;5:434–438. doi: 10.1038/7434. [DOI] [PubMed] [Google Scholar]
  • 32.Gehling UM, Ergun S, Schumacher U, Wagener C, Pantel K, Otem M, et al. In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood. 2000;95:3106–3112. [PubMed] [Google Scholar]
  • 33.Ingram DA, Caplice NM, Yoder MC. Unresolved questions, changing definitions, and novel paradigms for defining endothelial progenitor cells. Blood. 2005;106:1525–1531. doi: 10.1182/blood-2005-04-1509. [DOI] [PubMed] [Google Scholar]
  • 34.Rustemeyer P, Wittkowski W, Jurk K, Koller A. Optimized flow cytometric analysis of endothelial progenitor cells in peripheral blood. J Immunoassay Immunochem. 2006;27:77–88. doi: 10.1080/15321810500403789. [DOI] [PubMed] [Google Scholar]
  • 35.Kocher AA, Schuster MD, Bonaros N, Lietz K, Xiang G, Martens TP, et al. Myocardial homing and neovascularization by human bone marrow angioblasts is regulated by IL-8/Gro CXC chemokines. J Mol Cell Cardiol. 2006;40:455–464. doi: 10.1016/j.yjmcc.2005.11.013. [DOI] [PubMed] [Google Scholar]
  • 36.Imanishi T, Hano T, Nishio I. Estrogen reduces endothelial progenitor cell senescence through augmentation of telomerase activity. J Hypertens. 2005;23:1699–1706. doi: 10.1097/01.hjh.0000176788.12376.20. [DOI] [PubMed] [Google Scholar]
  • 37.Zheng H, Shen CJ, Qiu FY, Zhao YB, Fu GS. Stromal cell-derived factor 1alpha reduces senescence of endothelial progenitor subpopulation in lectin-binding and DiLDL-uptaking cell through telomerase activation and telomere elongation. J Cell Physiol. 2010;223:757–763. doi: 10.1002/jcp.22086. [DOI] [PubMed] [Google Scholar]
  • 38.Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, Ratajczak J, et al. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia. 2006;20:857–869. doi: 10.1038/sj.leu.2404171. [DOI] [PubMed] [Google Scholar]
  • 39.Zuba-Surma EK, Kucia M, Wu W, Klich I, Lillard JW, Jr, Ratajczak J, et al. Very small embryonic-like stem cells are present in adult murine organs: ImageStream-based morphological analysis and distribution studies. Cytometry A. 2008;73A:1116–1127. doi: 10.1002/cyto.a.20667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hocking AM, Gibran NS. Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res. 2010;316:2213–2219. doi: 10.1016/j.yexcr.2010.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wojakowski W, Tendera M, Kucia M, Zuba-Surma E, Paczkowska E, Ciosek J, et al. Mobilization of bone marrow-derived Oct-4+ SSEA-4+ very small embryonic-like stem cells in patients with acute myocardial infarction. J Am Coll Cardiol. 2009;5:1–9. doi: 10.1016/j.jacc.2008.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kucia M, Halasa M, Wysoczynski M, Baskiewicz-Masiul M, Moldenhawer S, Zuba-Surma E, et al. Morphological and molecular characterization of novel population of CXCR4+ SSEA-4+ Oct-4+ very small embryonic-like cells purified from human cord blood: preliminary report. Leukemia. 2007;21:297–303. doi: 10.1038/sj.leu.2404470. [DOI] [PubMed] [Google Scholar]
  • 43.Shin DM, Zuba-Surma EK, Wu W, Ratajczak J, Wysoczynski M, Ratajczak MZ, et al. Novel epigenetic mechanisms that control pluripotency and quiescence of adult bone marrow-derived Oct4(+) very small embryonic like stem cells. Leukemia. 2009;23:2042–2051. doi: 10.1038/leu.2009.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shin DM, Liu R, Klich I, Wu W, Ratajczak J, Kucia M, et al. Molecular signature of adult bone marrow-purified very small embryonic-like stem cells supports their developmental epiblast/germ line origin. Leukemia. 2010;24:1450–1461. doi: 10.1038/leu.2010.121. [DOI] [PubMed] [Google Scholar]
  • 45.Sovalat H, Scrofani M, Eidenschenk A, Pasquet S, Rimelen V, Hénon P. Identification and isolation from either adult human bone marrow or G-CSF mobilized peripheral blood of CD34+/CD133+/CXCR4+/Lin-CD45- cells, featuring morphological, molecular and phenotypic characteristics of very small embryonic-like (VSEL) stem cells. Exp Hematol. 2011;39:495. doi: 10.1016/j.exphem.2011.01.003. [DOI] [PubMed] [Google Scholar]
  • 46.Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25:977–988. doi: 10.1016/j.immuni.2006.10.016. [DOI] [PubMed] [Google Scholar]
  • 47.Imai K, Kobayashi M, Wang J, Shinobu N, Yoshida H, Hamada J, et al. Selective secretion of chemoattractants for haemopoietic progenitor cells in bone marrow endothelial cells: a possible role in homing of haemopoietic progenitor cells to bone marrow. Br J Haematol. 1999;106:905–911. doi: 10.1046/j.1365-2141.1999.01644.x. [DOI] [PubMed] [Google Scholar]
  • 48.Pablos JL, Amara A, Bouloc A, Santiago B, Caruz A, Galindo M, et al. Stromal-cell derived factor is expressed by dendritic cells and endothelium in human skin. Am J Pathol. 1999;155:1577–1586. doi: 10.1016/S0002-9440(10)65474-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kollet O, Shivtiel S, Chen YQ, Suriawinata J, Thung SN, Dabeva MD, et al. HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J Clin Invest. 2003;112:160–169. doi: 10.1172/JCI17902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Stumm RK, Rummel J, Junker V, Culmsee C, Pfeiffer M, Krieglstein J, et al. A dual role for the SDF-1/CXCR4 chemokine receptor system in adult brain: isoform-selective regulation of SDF-1 expression modulates CXCR4-dependent neuronal plasticity and cerebral leukocyte recruitment after focal ischemia. J Neurosci. 2002;22:5865–5878. doi: 10.1523/JNEUROSCI.22-14-05865.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Dar A, Kollet O, Lapidot T. 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. 2006;34:967–975. doi: 10.1016/j.exphem.2006.04.002. [DOI] [PubMed] [Google Scholar]
  • 52.Spiegel A, Kalinkovich A, Shivtiel S, Kollet O, Lapidot T. Stem cell regulation via dynamic interactions of the nervous and immune systems with the microenvironment. Cell Stem Cell. 2008;3:484–492. doi: 10.1016/j.stem.2008.10.006. [DOI] [PubMed] [Google Scholar]
  • 53.Kalinkovich A, Spiegel A, Shivtiel S, Kollet O, Jordaney N, Piacibello W, et al. Blood-forming stem cells are nervous: direct and indirect regulation of immature human CD34+ cells by the nervous system. Brain Behav Immun. 2009;23:1059–1065. doi: 10.1016/j.bbi.2009.03.008. [DOI] [PubMed] [Google Scholar]
  • 54.Aicher A, Kollet O, Heeschen C, Liebner S, Urbich C, Ihling C, et al. The Wnt antagonist Dickkopf-1 mobilizes vasculogenic progenitor cells via activation of the bone marrow endosteal stem cell niche. Circ Res. 2008;103:796–803. doi: 10.1161/CIRCRESAHA.107.172718. [DOI] [PubMed] [Google Scholar]
  • 55.Spiegel A, Shivtiel S, Kalinkovich A, Lundin A, Netzer N, Goichberg P, et al. Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34(+) cells through Wnt signaling. Nat Immunol. 2007;8:1123–1131. doi: 10.1038/ni1509. [DOI] [PubMed] [Google Scholar]
  • 56.Fleming HE, Janzen V, Lo Celso C, Guo J, Leahy KM, Kronenberg HM, et al. Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell. 2008;2:274–283. doi: 10.1016/j.stem.2008.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Spiegel A, Kalinkovich A, Shivtiel S, Kollet O, Lapidot T. Stem cell regulation via dynamic interactions of the nervous and immune systems with the microenvironment. Cell Stem Cell. 2008;3:484–492. doi: 10.1016/j.stem.2008.10.006. [DOI] [PubMed] [Google Scholar]
  • 58.Hennemann B, Ickenstein G, Sauerbruch S, Luecke K, Haas S, Horn N, et al. Mobilization of CD34+ hematopoietic cells, colony-forming cells and long-term culture-initiating cells into the peripheral blood of patients with an acute cerebral ischemic insult. Cytotherapy. 2008;10:303–311. doi: 10.1080/14653240801949994. [DOI] [PubMed] [Google Scholar]
  • 59.Dunac A, Frelin C, Popolo-Blondeau M, Chatel M, Mahagne MH, Philip PJ. Neurological and functional recovery in human stroke are associated with peripheral blood CD34+ cell mobilization. J Neurol. 2007;254:327–332. doi: 10.1007/s00415-006-0362-1. [DOI] [PubMed] [Google Scholar]
  • 60.Chang YC, Shyu WC, Lin SZ, Li H. Regenerative therapy for stroke. Cell Transplant. 2007;16:171–181. [PubMed] [Google Scholar]
  • 61.Taguchi A, Soma T, Tanaka H, Kanda T, Nishimura H, Yoshikawa H, et al. Administration of CD34+ cells after stroke enhances neurogenesis via angiogenesis in a mouse model. J Clin Invest. 2004;114:330–338. doi: 10.1172/JCI20622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chernykh ER, Shevela EY, Leplina OY, Tikhonova MA, Ostanin AA, Kulagin AD, et al. Characteristics of bone marrow cells under conditions of impaired innervation in patients with spinal trauma. Bull Exp Biol Med. 2006;141:117–120. doi: 10.1007/s10517-006-0109-0. [DOI] [PubMed] [Google Scholar]
  • 63.Hwang WS, Chen SH, Lin CH, Chang HK, Chen WC, Lin MT. Human umbilical cord blood-derived CD34+ cells can be used as a prophylactic agent for experimental heatstroke. J Pharmacol Sci. 2008;106:46–55. doi: 10.1254/jphs.fp0071567. [DOI] [PubMed] [Google Scholar]
  • 64.Chen SH, Chang FM, Chang HK, Chen WC, Huang KF, Lin MT. Human umbilical cord blood-derived CD34+ cells cause attenuation of multiorgan dysfunction during experimental heatstroke. Shock. 2007;27:663–671. doi: 10.1097/01.shk.0000248593.71388.40. [DOI] [PubMed] [Google Scholar]
  • 65.Zhao ZM, Li HJ, Liu HY, Lu SH, Yang RC, Zhang QJ, et al. Intraspinal transplantation of CD34+ human umbilical cord blood cells after spinal cord hemisection injury improves functional recovery in adult rats. Cell Transplant. 2004;13:113–122. doi: 10.3727/000000004773301780. [DOI] [PubMed] [Google Scholar]
  • 66.Nishio Y, Koda M, Kamada T, Someya Y, Yoshinaga K, Okada S, et al. The use of hemopoietic stem cells derived from human umbilical cord blood to promote restoration of spinal cord tissue and recovery of hindlimb function in adult rats. J Neurosurg Spine. 2006;5:424–433. doi: 10.3171/spi.2006.5.5.424. [DOI] [PubMed] [Google Scholar]
  • 67.Garbuzova-Davis S, Willing AE, Zigova T, Saporta S, Justen EB, Lane JC, et al. Intravenous administration of human umbilical cord blood cells in a mouse model of amyotrophic lateral sclerosis: distribution, migration, and differentiation. J Hematother Stem Cell Res. 2003;12:255–270. doi: 10.1089/152581603322022990. [DOI] [PubMed] [Google Scholar]
  • 68.Nikolic WV, Hou H, Touan T, Zhu Y, Giunta B, Sanberg CD, et al. Peripherally administered human umbilical cord blood cells reduce parenchymal and vascular beta-amyloid deposits in Alzheimer mice. Stem Cells Dev. 2008;17:423–439. doi: 10.1089/scd.2008.0018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bachstetter AD, Pabon MM, Cole MJ, Hudson CE, Sanberg PR, Wiling AE, et al. Peripheral injection of human umbilical cord blood stimulates neurogenesis in the aged rat brain. BMC Neurosci. 2008;9:22. doi: 10.1186/1471-2202-9-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bajada S, Mazakova I, Richardson JB, Ashammakhi N. Updates on stem cells and their applications in regenerative medicine. J Tissue Eng Regen Med. 2008;2:169–183. doi: 10.1002/term.83. [DOI] [PubMed] [Google Scholar]
  • 71.Wojakowski W, Kucia M, Kazmierski M, Ratajczak MZ, Tendera M. Circulating progenitor cells in stable coronary heart disease and acute coronary syndromes: relevant reparatory mechanism? Heart. 2008;94:27–33. doi: 10.1136/hrt.2006.103358. [DOI] [PubMed] [Google Scholar]
  • 72.Pai M, Zacharoulis D, Milicevic MN, Helmy S, Jiao LR, Levicar N, et al. Autologous infusion of expanded mobilized adult bone marrow-derived CD34+ cells into patients with alcoholic liver cirrhosis. Am J Gastroenterol. 2008;103:1952–1958. doi: 10.1111/j.1572-0241.2008.01993.x. [DOI] [PubMed] [Google Scholar]
  • 73.Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Tissue Kinet. 1970;3:393–403. doi: 10.1111/j.1365-2184.1970.tb00347.x. [DOI] [PubMed] [Google Scholar]
  • 74.Gronthos S, Simmons PJ. The biology and application of human bone marrow stromal cell precursors. J Hematother. 1996;5:15–23. doi: 10.1089/scd.1.1996.5.15. [DOI] [PubMed] [Google Scholar]
  • 75.Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone. 1992;13:69–80. doi: 10.1016/8756-3282(92)90363-2. [DOI] [PubMed] [Google Scholar]
  • 76.Pereira RF, O’Hara MD, Laptev AV, Halford KW, Pollard MD, Class R, et al. Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta. Proc Natl Acad Sci USA. 1998;95:1142–1147. doi: 10.1073/pnas.95.3.1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71–74. doi: 10.1126/science.276.5309.71. [DOI] [PubMed] [Google Scholar]
  • 78.Pittenger MF, Mackay MA, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–147. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 79.Hamada H, Kobune M, Nakamura K, Kawano Y, Kato K, Honmou O, et al. Mesenchymal stem cells (MSC) as therapeutic cytoreagents for gene therapy. Cancer Sci. 2005;96:149–156. doi: 10.1111/j.1349-7006.2005.00032.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Honma T, Honmou O, Iihoshi S, Harada K, Houkin K, Hamada H, et al. Intravenous infusion of immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat. Exp Neurol. 2006;199:56–66. doi: 10.1016/j.expneurol.2005.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kurozumi K, Nakamura K, Tamiya T, Kawano Y, Ishii K, Kobune M, et al. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther. 2005;11:96–104. doi: 10.1016/j.ymthe.2004.09.020. [DOI] [PubMed] [Google Scholar]
  • 82.Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57:874–882. doi: 10.1002/ana.20501. [DOI] [PubMed] [Google Scholar]
  • 83.Chen J, Li Y, Wang L, Lu M, Zhang X, Chopp M. Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci. 2001;189:49–57. doi: 10.1016/s0022-510x(01)00557-3. [DOI] [PubMed] [Google Scholar]
  • 84.Chen J, Wang L, Zhang Z, Lu D, Lu M, Chopp M. Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke. 2001;32:1005–1011. doi: 10.1161/01.str.32.4.1005. [DOI] [PubMed] [Google Scholar]
  • 85.Chopp M, Li Y. Treatment of neural injury with marrow stromal cells. Lancet Neurol. 2002;1:92–100. doi: 10.1016/s1474-4422(02)00040-6. [DOI] [PubMed] [Google Scholar]
  • 86.Li Y, Chen J, Chopp M. Adult bone marrow transplantation after stroke in adult rats. Cell Transplant. 2001;10:31–40. [PubMed] [Google Scholar]
  • 87.Li Y, Chen J, Wang L, Lu M, Chopp M. Treatment of stroke in rat with intracarotid administration of marrow stromal cells. Neurology. 2002;56:1666–1672. doi: 10.1212/wnl.56.12.1666. [DOI] [PubMed] [Google Scholar]
  • 88.Li Y, Chen J, Zhang CL, Wang L, Lu D, Katakowski M, et al. Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells. Glia. 2005;49:407–417. doi: 10.1002/glia.20126. [DOI] [PubMed] [Google Scholar]
  • 89.Rempe DA, Kent TA. Using bone marrow stromal cells for treatment of stroke. Neurology. 2002;59:486–487. doi: 10.1212/wnl.59.4.486. [DOI] [PubMed] [Google Scholar]
  • 90.Song S, Kamath S, Mosquera D, Zigova T, Sanberg P, Vesely DL, et al. Expression of brain natriuretic peptide by human bone marrow stromal cells. Exp Neurol. 2004;185:191–197. doi: 10.1016/j.expneurol.2003.09.003. [DOI] [PubMed] [Google Scholar]
  • 91.Tang Y, Yasuhara T, Hara K, Matsukawa N, Maki M, Yu G, et al. Transplantation of bone marrow-derived stem cells: a promising therapy for stroke. Cell Transplant. 2007;16:159–169. [PubMed] [Google Scholar]
  • 92.Shen LH, Li Y, Chen J, Zacharek A, Gao Q, Kapke A, et al. Therapeutic benefit of bone marrow stromal cells administered 1 month after stroke. J Cereb Blood Flow Metab. 2007;27:6–13. doi: 10.1038/sj.jcbfm.9600311. [DOI] [PubMed] [Google Scholar]
  • 93.Chen X, Li Y, Katakowski M, Zhang L, Chen J, Xu Y, et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology. 2002;22:275–279. doi: 10.1046/j.1440-1789.2002.00450.x. [DOI] [PubMed] [Google Scholar]
  • 94.Chen J, Zhang ZG, Li Y, Wang L, Xu YX, Gautam SC, et al. Intravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in rats. Circ Res. 2003;92:692–699. doi: 10.1161/01.RES.0000063425.51108.8D. [DOI] [PubMed] [Google Scholar]
  • 95.Li Y, Chen J, Chen XG, Wang L, Gautam SC, Xu YX, et al. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology. 2002;59:514–523. doi: 10.1212/wnl.59.4.514. [DOI] [PubMed] [Google Scholar]
  • 96.Chen J, Li Y, Katakowski M, Chen X, Wang L, Lu D, et al. Intravenous bone marrow stromal cell therapy reduces apoptosis and promotes endogenous cell proliferation after stroke in female rat. J Neurosci Res. 2003;73:778–786. doi: 10.1002/jnr.10691. [DOI] [PubMed] [Google Scholar]
  • 97.Zhang J, Li Y, Chen J, Yang M, Katakowski M, Lu M, et al. Expression of insulin-like growth factor 1 and receptor in ischemic rats treated with human marrow stromal cells. Brain Res. 2004;1030:19–27. doi: 10.1016/j.brainres.2004.09.061. [DOI] [PubMed] [Google Scholar]
  • 98.Zhang ZG, Zhang L, Jiang Q, Zhang R, Davies K, Powers C, et al. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. J Clin Invest. 2000;106:829–838. doi: 10.1172/JCI9369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Benraiss A, Chmielnicki E, Lerner K, Roh D, Goldman SA. Adenoviral brain-derived neurotrophic factor induces both neostriatal and olfactory neuronal recruitment from endogenous progenitor cells in the adult forebrain. J Neurosci. 2001;21:6718–6731. doi: 10.1523/JNEUROSCI.21-17-06718.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ay I, Sugimori H, Finklestein SP. Intravenous basic fibroblast growth factor (bFGF) decreases DNA fragmentation and prevents downregulation of Bcl-2 expression in the ischemic brain following middle cerebral artery occlusion in rats. Brain Res Mol Brain Res. 2001;87:71–80. doi: 10.1016/s0169-328x(00)00285-0. [DOI] [PubMed] [Google Scholar]
  • 101.Rosenblatt S, Irikura K, Caday CG, Finklestein SP, Moskowitz MA. Basic fibroblast growth factor dilates rat pial arterioles. J Cereb Blood Flow Metab. 1994;14:70–74. doi: 10.1038/jcbfm.1994.11. [DOI] [PubMed] [Google Scholar]
  • 102.Kurozumi K, Nakamura K, Tamiya T, Kawano Y, Kobune M, Hirai S, et al. BDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion model. Mol Ther. 2004;9:189–197. doi: 10.1016/j.ymthe.2003.10.012. [DOI] [PubMed] [Google Scholar]
  • 103.Ikeda N, Nonoguchi N, Zhao MZ, Watanabe T, Kajimoto Y, Furutama D, et al. Bone marrow stromal cells that enhanced fibroblast growth factor-2 secretion by herpes simplex virus vector improve neurological outcome after transient focal cerebral ischemia in rats. Stroke. 2005;36:2725–2730. doi: 10.1161/01.STR.0000190006.88896.d3. [DOI] [PubMed] [Google Scholar]
  • 104.Zhao MZ, Nonoguchi N, Ikeda N, Watanabe T, Furutama D, Miyazawa D, et al. Novel therapeutic strategy for stroke in rats by bone marrow stromal cells and ex vivo HGF gene transfer with HSV-1 vector. J Cereb Blood Flow Metab. 2006;26:1176–1188. doi: 10.1038/sj.jcbfm.9600273. [DOI] [PubMed] [Google Scholar]
  • 105.Dezawa M, Hoshino M, Ide C. Treatment of neurodegenerative diseases using adult bone marrow stromal cell derived neurons. Expert Opin Biol Ther. 2005;5:427–435. doi: 10.1517/14712598.5.4.427. [DOI] [PubMed] [Google Scholar]
  • 106.Chen J, Li Y, Chopp M. Intracerebral transplantation of bone marrow with BDNF after MCAo in rat. Neuropharmacology. 2000;39:711–716. doi: 10.1016/s0028-3908(00)00006-x. [DOI] [PubMed] [Google Scholar]
  • 107.Chen J, Li Y, Wang L, Lu M, Chopp M. Caspase inhibition by Z-VAD increases the survival of grafted bone marrow cells and improves functional outcome after MCAo in rats. J Neurol Sci. 2002;199:17–24. doi: 10.1016/s0022-510x(02)00075-8. [DOI] [PubMed] [Google Scholar]
  • 108.Chen J, Li Y, Zhang R, Katakowski M, Gautam SC, Xu Y, et al. Combination therapy of stroke in rats with a nitric oxide donor and human bone marrow stromal cells enhances angiogenesis and neurogenesis. Brain Res. 2004;1005:21–28. doi: 10.1016/j.brainres.2003.11.080. [DOI] [PubMed] [Google Scholar]
  • 109.Shen LH, Li Y, Chen J, Zhang J, Vanguri P, Borneman J, et al. Intracarotid transplantation of bone marrow stromal cells increases axon-myelin remodeling after stroke. Neuroscience. 2006;137:393–399. doi: 10.1016/j.neuroscience.2005.08.092. [DOI] [PubMed] [Google Scholar]
  • 110.Li Y, Chen J, Zhang CL, Wang L, Lu D, Katakowski M, et al. Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells. Glia. 2005;49:407–417. doi: 10.1002/glia.20126. [DOI] [PubMed] [Google Scholar]
  • 111.Shen LH, Li Y, Chen J, Zacharek A, Gao Q, Kapke A, et al. Therapeutic benefit of bone marrow stromal cells administered 1 month after stroke. J Cereb Blood Flow Metab. 2007;27:6–13. doi: 10.1038/sj.jcbfm.9600311. [DOI] [PubMed] [Google Scholar]
  • 112.Gao Q, Li Y, Chopp M. Bone marrow stromal cells increase astrocyte survival via upregulation of phosphoinositide 3-kinase/threonine protein kinase and mitogen activated protein kinase kinase/extracellular signal-regulated kinase pathways and stimulate astrocyte trophic factor gene expression after anaerobic insult. Neuroscience. 2005;136:123–134. doi: 10.1016/j.neuroscience.2005.06.091. [DOI] [PubMed] [Google Scholar]
  • 113.Gao Q, Katakowski M, Chen X, Li Y, Chopp M. Human marrow stromal cells enhance connexin43 gap junction intercellular communication in cultured astrocytes. Cell Transplant. 2005;14:109–117. doi: 10.3727/000000005783983205. [DOI] [PubMed] [Google Scholar]
  • 114.Zhang C, Li Y, Chen J, Gao Q, Zacharek A, Kapke A, et al. Bone marrow stromal cells upregulate expression of bone morphogenetic proteins 2 and 4, gap junction protein connexin-43 and synaptophysin after stroke in rats. Neuroscience. 2006;141:687–695. doi: 10.1016/j.neuroscience.2006.04.054. [DOI] [PubMed] [Google Scholar]
  • 115.Li Y, McIntosh K, Chen J, Zhang C, Gao Q, Borneman J, et al. Allogeneic bone marrow stromal cells promote glial-axonal remodeling without immunologic sensitization after stroke in rats. Exp Neurol. 2006;198:313–325. doi: 10.1016/j.expneurol.2005.11.029. [DOI] [PubMed] [Google Scholar]
  • 116.Ceradini DJ, Gurtner GC. Homing to hypoxia: HIF-1 as a mediator of progenitor cell recruitment to injured tissue. Trends Cardiovasc Med. 2005;15:57–63. doi: 10.1016/j.tcm.2005.02.002. [DOI] [PubMed] [Google Scholar]
  • 117.Griese DP, Ehsan A, Melo LG, Kong D, Zhang L, Mann MJ, et al. Isolation and transplantation of autologous circulating endothelial cells into denuded vessels and prosthetic grafts: implications for cell-based vascular therapy. Circulation. 2003;108:2710–2715. doi: 10.1161/01.CIR.0000096490.16596.A6. [DOI] [PubMed] [Google Scholar]
  • 118.Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, et al. Evidence for circulating bone marrow-derived endothelial cells. Blood. 1998;92:362–367. [PubMed] [Google Scholar]
  • 119.Murohara T, Ikeda H, Duan J, Shintani S, Sasaki K, Egucji H, et al. Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. J Clin Invest. 2000;105:1527–1536. doi: 10.1172/JCI8296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood. 2000;95:952–958. [PubMed] [Google Scholar]
  • 121.Schatteman GC, Awad O. Hemangioblasts, angioblasts, and adult endothelial cell progenitors. Anat Rec A Discov Mol Cell Evol Biol. 2004;276:13–21. doi: 10.1002/ar.a.10131. [DOI] [PubMed] [Google Scholar]
  • 122.Khakoo AY, Finkel T. Endothelial progenitor cells. Annu Rev Med. 2005;56:79–101. doi: 10.1146/annurev.med.56.090203.104149. [DOI] [PubMed] [Google Scholar]
  • 123.Cines DB, Pollak ES, Buck CA, Loscalzo J, Zimmerman GA, McEver RP, et al. Endothelial cells in physiology and in the pathophysiology of vascular disorders. Blood. 1998;91:3527–3561. [PubMed] [Google Scholar]
  • 124.Ghani U, Shulaib A, Salam A, Nasir A, Shuaib U, Jeerakathil T, et al. Endothelial progenitor cells during cerebrovascular disease. Stroke. 2005;36:151–153. doi: 10.1161/01.STR.0000149944.15406.16. [DOI] [PubMed] [Google Scholar]
  • 125.Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348:593–600. doi: 10.1056/NEJMoa022287. [DOI] [PubMed] [Google Scholar]
  • 126.Ito H, Rovira II, Bloom ML, Takeda K, Ferrans VJ, Quyyumi AA, et al. Endothelial progenitor cells as putative targets for angiostatin. Cancer Res. 1999;59:5875–5877. [PubMed] [Google Scholar]
  • 127.Kreipe H, Radzun HJ, Schumacher U, Parwaresch MR. Lectin binding and surface glycoprotein pattern of human macrophage populations. Histochemistry. 1986;86:201–206. doi: 10.1007/BF00493388. [DOI] [PubMed] [Google Scholar]
  • 128.Lougheed M, Moore ED, Scriven DR, Steinbrecher UP. Uptake of oxidized LDL by macrophages differs from that of acetyl LDL and leads to expansion of an acidic endolysosomal compartment. Arterioscler Thromb Vasc Biol. 1999;19:1881–1890. doi: 10.1161/01.atv.19.8.1881. [DOI] [PubMed] [Google Scholar]
  • 129.Zengin E, Chalajour F, Gehling UM, Ito WD, Treede H, Lauke H, et al. Vascular wall resident progenitor cells: a source for postnatal vasculogenesis. Development. 2006;133:1543–1551. doi: 10.1242/dev.02315. [DOI] [PubMed] [Google Scholar]
  • 130.Borlongan CV, Ling JG, Dillon-Carter O, Yu G, Hadman M, Cheng C, et al. Bone marrow grafts restore cerebral blood flow and blood brain barrier in stroke rats. Brain Res. 2004;1010:108–116. doi: 10.1016/j.brainres.2004.02.072. [DOI] [PubMed] [Google Scholar]
  • 131.Chopp M, Li Y. Treatment of neural injury with marrow stromal cells. Lancet Neurol. 2002;1:92–100. doi: 10.1016/s1474-4422(02)00040-6. [DOI] [PubMed] [Google Scholar]
  • 132.Nan Z, Grande A, Sanberg CD, Sanberg PR, Low WC. Infusion of human umbilical cord blood ameliorates neurologic deficits in rats with hemorrhagic brain injury. Ann NY Acad Sci. 2005;1049:84–96. doi: 10.1196/annals.1334.009. [DOI] [PubMed] [Google Scholar]
  • 133.Taguchi A, Soma T, Tanaka H, Kanda T, Nishimura H, Yoshikawa H, et al. Administration of CD34+ cells after stroke enhances neurogenesis via angiogenesis in a mouse model. J Clin Invest. 2004;114:330–338. doi: 10.1172/JCI20622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Vendrame M, Cassady J, Newcomb J, Butler T, Pennypacker KR, Zigova T, et al. Infusion of human umbilical cord blood cells in a rat model of stroke dose dependently rescues behavioral deficits and reduces infarct volume. Stroke. 2004;35:2390–2395. doi: 10.1161/01.STR.0000141681.06735.9b. [DOI] [PubMed] [Google Scholar]
  • 135.Willing AE, Lixian J, Milliken M, Poulos S, Zigova T, Song S, et al. Intravenous versus intrastriatal cord blood administration in a rodent model of stroke. J Neurosci Res. 2003;73:296–307. doi: 10.1002/jnr.10659. [DOI] [PubMed] [Google Scholar]
  • 136.Hristov M, Weber C. Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance. J Cell Mol Med. 2004;8:498–508. doi: 10.1111/j.1582-4934.2004.tb00474.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005;438:932–936. doi: 10.1038/nature04478. [DOI] [PubMed] [Google Scholar]
  • 138.Masuda H, Asahara T. Post-natal endothelial progenitor cells for neovascularization in tissue regeneration. Cardiovasc Res. 2003;58:390–398. doi: 10.1016/s0008-6363(02)00785-x. [DOI] [PubMed] [Google Scholar]
  • 139.Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, et al. Evidence for circulating bone marrow-derived endothelial cells. Blood. 1998;92:362–367. [PubMed] [Google Scholar]
  • 140.Asahara T, Murohara T, Sullivan A, Silver M, van der Zeer R, Li T, et al. Isolation of progenitor endothelial cells for angiogenesis. Science. 1997;275:964. doi: 10.1126/science.275.5302.964. [DOI] [PubMed] [Google Scholar]
  • 141.Bompais H, Chagraoui J, Canron X, Crisan M, Liu XH, Anjo A, et al. Human endothelial cells derived from circulating progenitors display specific functional properties compared with mature vessel wall endothelial cells. Blood. 2004;103:2577–2584. doi: 10.1182/blood-2003-08-2770. [DOI] [PubMed] [Google Scholar]
  • 142.Fadini GP. An underlying principle for the study of circulating progenitor cells in diabetes and its complications. Diabetologia. 2008;51:1091–1094. doi: 10.1007/s00125-008-1021-0. [DOI] [PubMed] [Google Scholar]
  • 143.Chen JZ, Zhang FR, Tao QM, Wang XX, Zhu JH, Zhu JH. Number and activity of endothelial progenitor cells from peripheral blood in patients with hypercholesterolaemia. Clin Sci. 2004;107:273–280. doi: 10.1042/CS20030389. [DOI] [PubMed] [Google Scholar]
  • 144.Pirro M, Schillaci G, Menecali C, Bagalia F, Paltriccia R, Vaudo G, et al. Reduced number of circulating endothelial progenitors and HOXA9 expression in CD34+ cells of hypertensive patients. J Hypertens. 2007;25:2093–2099. doi: 10.1097/HJH.0b013e32828e506d. [DOI] [PubMed] [Google Scholar]
  • 145.Umemura T, Soga J, Hidaka T, Takemoto H, Nakamura S, Jitsuiki D, et al. Aging and hypertension are independent risk factors for reduced number of circulating endothelial progenitor cells. Am J Hypertens. 2008;21:1203–1209. doi: 10.1038/ajh.2008.278. [DOI] [PubMed] [Google Scholar]
  • 146.Del Papa N, Quirici N, Soligo D, Scavulo C, Cortiana M, Borsotti C, et al. Bone marrow endothelial progenitors are defective in systemic sclerosis. Arthritis Rheum. 2006;54:2605–2615. doi: 10.1002/art.22035. [DOI] [PubMed] [Google Scholar]
  • 147.Kuwana M, Okazaki Y, Yasuoka H, Kawakami Y, Ikeda Y. Defective vasculogenesis in systemic sclerosis. Lancet. 2004;364:603–610. doi: 10.1016/S0140-6736(04)16853-0. [DOI] [PubMed] [Google Scholar]
  • 148.Heiss C, Keymel S, Niesler U, Ziemann J, Kelm M, Kalka C. Impaired progenitor cell activity in age-related endothelial dysfunction. JACC. 2005;45:1441–1448. doi: 10.1016/j.jacc.2004.12.074. [DOI] [PubMed] [Google Scholar]
  • 149.Kondo T, Hayashi M, Takeshita K, Numaguchi Y, Kobayashi K, Iino S, et al. Smoking cessation rapidly increases circulating progenitor cells in peripheral blood in chronic smokers. Arterioscler Thromb Vasc Biol. 2004;24:1442–1447. doi: 10.1161/01.ATV.0000135655.52088.c5. [DOI] [PubMed] [Google Scholar]
  • 150.Michaud SE, Dussault S, Haddad P, Groleau J, Rivard A. Circulating endothelial progenitor cells from healthy smokers exhibit impaired functional activities. Atherosclerosis. 2006;187:423–432. doi: 10.1016/j.atherosclerosis.2005.10.009. [DOI] [PubMed] [Google Scholar]
  • 151.Kunz GA, Liang G, Cuculi F, Gregg D, Vata KC, Shaw LK, et al. Circulating endothelial progenitor cells predict coronary artery disease severity. Am Heart J. 2006;152:190–195. doi: 10.1016/j.ahj.2006.02.001. [DOI] [PubMed] [Google Scholar]
  • 152.Botta R, Gao E, Stassi G, Bonci D, Relosi E, Zwas D, et al. Heart infarct in NOD-SCID mice: therapeutic vasculogenesis by transplantation of human CD34+ cells and low dose CD34+KDR+ cells. FASEB J. 2004;18:1392–1394. doi: 10.1096/fj.03-0879fje. [DOI] [PubMed] [Google Scholar]
  • 153.Kawamoto A, Gwon HC, Iwaguro H, Yamaguchi JI, Uchida S, Masuda H, et al. Therapeutic potential of ex vivo expanded endothelial progenitor cells for myocardial ischemia. Circulation. 2001;103:634–637. doi: 10.1161/01.cir.103.5.634. [DOI] [PubMed] [Google Scholar]
  • 154.Madeddu P, Emanueli C, Pelosi E, Salis MB, Cerio AM, Bonanno G, et al. Transplantation of low dose CD34+KDR+ cells promotes vascular and muscular regeneration in ischemic limbs. FASEB J. 2004;18:1737–1739. doi: 10.1096/fj.04-2192fje. [DOI] [PubMed] [Google Scholar]
  • 155.Rouhl RP, van Oostenbrugge RJ, Damoiseaux J, Tervaert JW, Lodder J. Endothelial progenitor cell research in stroke: a potential shift in pathophysiological and therapeutical concepts. Stroke. 2008;39:2158–2165. doi: 10.1161/STROKEAHA.107.507251. [DOI] [PubMed] [Google Scholar]
  • 156.Erbs S, Linke A, Adams V, Lenk K, Thiele H, Diederich KW, et al. Transplantation of blood-derived progenitor cells after recanalization of chronic coronary artery occlusion: first randomized and placebo-controlled study. Circ Res. 2005;97:756–762. doi: 10.1161/01.RES.0000185811.71306.8b. [DOI] [PubMed] [Google Scholar]
  • 157.Li ZQ, Zhang M, Jing YZ, Zhang WW, Liu Y, Cui LJ, et al. The clinical study of autologous peripheral blood stem cell transplantation by intracoronary infusion in patients with acute myocardial infarction (AMI) Int J Cardiol. 2007;115:52–56. doi: 10.1016/j.ijcard.2006.04.005. [DOI] [PubMed] [Google Scholar]
  • 158.Fernandez-Aviles F, San Roman JA, Garcia-Frade J, Fernandez ME, Penarrubia MJ, de la Fuente L, et al. Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res. 2004;95:742–748. doi: 10.1161/01.RES.0000144798.54040.ed. [DOI] [PubMed] [Google Scholar]
  • 159.Meluzin J, Janousek S, Mayer J, Groch L, Hornacek I, Hlinomaz O, et al. Three-, 6-, and 12-month results of autologous transplantation of mononuclear bone marrow cells in patients with acute myocardial infarction. Int J Cardiol. 2008;128:185–192. doi: 10.1016/j.ijcard.2007.04.098. [DOI] [PubMed] [Google Scholar]
  • 160.Meluzin J, Mayer J, Groch L, Janousek S, Hornacek I, Hlinomaz O, et al. Autologous transplantation of mononuclear bone marrow cells in patients with acute myocardial infarction: the effect of the dose of transplanted cells on myocardial function. Am Heart J. 2006;152:975. doi: 10.1016/j.ahj.2006.08.004. [DOI] [PubMed] [Google Scholar]
  • 161.Mocini D, Staibano M, Mele L, Giannantoni P, Menichella G, Colivicchi F, et al. Autologous bone marrow mononuclear cell transplantation in patients undergoing coronary artery bypass grafting. Am Heart J. 2006;151:192–197. doi: 10.1016/j.ahj.2005.02.001. [DOI] [PubMed] [Google Scholar]
  • 162.Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita CT, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation. 2003;107:2294–2302. doi: 10.1161/01.CIR.0000070596.30552.8B. [DOI] [PubMed] [Google Scholar]
  • 163.Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Silva GV, et al. Improved exercise capacity and ischemia 6 and 12 months after transendocardial injection of autologous bone marrow mononuclear cells for ischemic cardiomyopathy. Circulation. 2004;110:213–218. doi: 10.1161/01.CIR.0000138398.77550.62. [DOI] [PubMed] [Google Scholar]
  • 164.Strauer BE, Brehm M, Zeus T, Bartsch T, Schannwell C, Antke C, et al. Regeneration of human infarcted heart muscle by intracoronary autologous bone marrow cell transplantation in chronic coronary artery disease: the IACT Study. JACC. 2005;46:1651–1658. doi: 10.1016/j.jacc.2005.01.069. [DOI] [PubMed] [Google Scholar]
  • 165.Strauer BE, Breh M, Zeus T, Kostering M, Hernandez A, Sorg RV, et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation. 2002;106:1913–1918. doi: 10.1161/01.cir.0000034046.87607.1c. [DOI] [PubMed] [Google Scholar]
  • 166.Numaguchi Y, Sone T, Okumura K, Ishii M, Morita Y, Kubota R, et al. The impact of the capability of circulating progenitor cell to differentiate on myocardial salvage in patients with primary acute myocardial infarction. Circulation. 2006;114:114–119. doi: 10.1161/CIRCULATIONAHA.105.000588. [DOI] [PubMed] [Google Scholar]
  • 167.Dobert N, Britten M, Assmus B, Berner U, Menzel C, Lehmann R, et al. Transplantation of progenitor cells after reperfused acute myocardial infarction: evaluation of perfusion and myocardial viability with FDG-PET and thallium SPECT. Eur J Nucl Med Mol Imaging. 2004;31:1146–1151. doi: 10.1007/s00259-004-1490-4. [DOI] [PubMed] [Google Scholar]
  • 168.Lev EI, Kleiman NS, Birnbaum Y, Harris D, Korbling M, Estrov Z. Circulating endothelial progenitor cells and coronary collaterals in patients with non-ST segment elevation myocardial infarction. J Vasc Res. 2005;42:408–414. doi: 10.1159/000087370. [DOI] [PubMed] [Google Scholar]
  • 169.Hristov M, Heussen N, Schober A, Weber C. Intracoronary infusion of autologous bone marrow cells and left ventricular function after acute myocardial infarction: a meta-analysis. J Cell Mol Med. 2006;10:727–733. doi: 10.1111/j.1582-4934.2006.tb00432.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Dimmeler S, Zelher AM, Schneider MD. Unchain my heart: the scientific foundations of cardiac repair. J Clin Invest. 2005;115:572–583. doi: 10.1172/JCI24283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Higashi Y, Kimura M, Hara K, Noma K, Jitsuki D, Nakagawa K, et al. Autologous bone-marrow mononuclear cell implantation improves endothelium-dependent vasodilation in patients with limb ischemia. Circulation. 2004;109:1215–1218. doi: 10.1161/01.CIR.0000121427.53291.78. [DOI] [PubMed] [Google Scholar]
  • 172.Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, et al. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet. 2002;360:427. doi: 10.1016/S0140-6736(02)09670-8. [DOI] [PubMed] [Google Scholar]
  • 173.Taguchi A, Matsuyama T, Moriwaki H, Hayashi T, Hayashida K, Nagatsuka K, et al. Circulating cd34-positive cells provide an index of cerebrovascular function. Circulation. 2004;109:2972–2975. doi: 10.1161/01.CIR.0000133311.25587.DE. [DOI] [PubMed] [Google Scholar]
  • 174.Ghani U, Shuaib A, Salam A, Nasir A, Shuaib U, Jeerakathil T, et al. Endothelial progenitor cells during cerebrovascular disease. Stroke. 2005;36:151–153. doi: 10.1161/01.STR.0000149944.15406.16. [DOI] [PubMed] [Google Scholar]
  • 175.Komitova M, Mattsson B, Johansson BB, Eriksson PS. Enriched environment increases neural stem/progenitor cell proliferation and neurogenesis in the subventricular zone of stroke-lesioned adult rats. Stroke. 2005;36:1278–1282. doi: 10.1161/01.STR.0000166197.94147.59. [DOI] [PubMed] [Google Scholar]
  • 176.Zhang RL, Zhang ZG, Zhang L, Chopp M. Proliferation and differentiation of progenitor cells in the cortex and the subventricular zone in the adult rat after focal cerebral ischemia. Neuroscience. 2001;105:33–41. doi: 10.1016/s0306-4522(01)00117-8. [DOI] [PubMed] [Google Scholar]
  • 177.Kucia M, Zhang YP, Reca R, Wysoczynski M, Machalinski B, Majka M, et al. Cells enriched in markers of neural tissue-committed stem cells reside in the bone marrow and are mobilized into the peripheral blood following stroke. Leukemia. 2006;20:18–28. doi: 10.1038/sj.leu.2404011. [DOI] [PubMed] [Google Scholar]
  • 178.Ratajczak MZ, Kim CH, Wojakowski W, Janowska-Wieczorek A, Kucia M, Ratajczak J. Innate immunity as orchestrator of stem cell mobilization. Leukemia. 2010;24:1667–1675. doi: 10.1038/leu.2010.162. [DOI] [PubMed] [Google Scholar]
  • 179.Levesque JP, Helwani FM, Winkler IG. The endosteal ‘osteo-blastic’ niche and its role in hematopoietic stem cell homing and mobilization. Leukemia. 2010;24:1979–1992. doi: 10.1038/leu.2010.214. [DOI] [PubMed] [Google Scholar]
  • 180.Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells. 2010;28:585–596. doi: 10.1002/stem.269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Mund JA, Ingram DA, Yoder MC, Case J. Endothelial progenitor cells and cardiovascular cell-based therapies. Cytotherapy. 2009;11:103–113. doi: 10.1080/14653240802714827. [DOI] [PubMed] [Google Scholar]
  • 182.Szczot M, Wojtowicz T, Mozrzymas JW. GABAergic and glutamatergic currents in hippocampal slices and neuronal cultures show profound differences: a clue to a potent homeostatic modulation. J Physiol Pharmacol. 2010;61:501–506. [PubMed] [Google Scholar]
  • 183.Gharib SA, Dayyat EA, Khalyfa A, Kim J, Clair HB, Kucia M, et al. Intermittent hypoxia mobilizes bone marrow-derived very small embryonic-like stem cells and activates developmental transcriptional programs in mice. Sleep. 2010;33:1–8. doi: 10.1093/sleep/33.11.1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Burns TC, Verfaillie CM, Low WC. Stem cells for ischemic brain injury: a critical review. J Comp Neurol. 2009;515:125–144. doi: 10.1002/cne.22038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Farin A, Liu CY, Langmoen IA, Apuzzo ML. Biological restoration of central nervous system architecture and function: part 3-stem cell- and cell-based applications and realities in the biological management of central nervous system disorders: traumatic, vascular, and epilepsy disorders. Neurosurgery. 2009;65:831–859. doi: 10.1227/01.NEU.0000351721.81175.0B. [DOI] [PubMed] [Google Scholar]
  • 186.Dwain I, Xiangpeng Y, Zeng Z, Patricia T, Joh SY. Neural stem cells–a promising potential therapy for brain tumors. Curr Stem Cell Res Ther. 2006;1:79–84. doi: 10.2174/157488806775269070. [DOI] [PubMed] [Google Scholar]
  • 187.Morancho A, Rosell A, Barcia-Bonilla L, Montaner J. Metalloproteinase and stroke size; role for anti-inflammatory treatment. Ann NY Acad Sci. 2010;1207:123–133. doi: 10.1111/j.1749-6632.2010.05734.x. [DOI] [PubMed] [Google Scholar]
  • 188.Wojakowski W, Tendera M, Kucia M, Zuba-Surma E, Paczkowska E, Ciosek J, et al. Mobilization of bone marrow-derived Oct-4+ SSEA-4+ very small embryonic-like stem cells in patients with acute myocardial infarction. J Am Coll Cardiol. 2009;53:1–9. doi: 10.1016/j.jacc.2008.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Paczkowska E, Kucia M, Koziarska D, Halasa M, Safranow K, Masiuk M, et al. Clinical evidence that very small embryonic-like stem cells are mobilized into peripheral blood in patients after stroke. Stroke. 2009;4:1237–1244. doi: 10.1161/STROKEAHA.108.535062. [DOI] [PubMed] [Google Scholar]
  • 190.Ratajczak J, Zuba-Surma E, Paczkowska W, Kucia M, Nowacki P, Ratajczak MZ. Stem cells for neural regeneration–a potential application of very small embryonic-like stem cells. J Physiol Pharmacol. 2011;62:3–12. [PubMed] [Google Scholar]
  • 191.Ratajczak MZ, Shin DM, Liu R, Marlicz W, Tarnowski M, Ratajczak J, et al. Epiblast/germ line hypothesis of cancer development revisited: lesson from the presence of Oct-4+ cells in adult tissues. Stem Cell Rev. 2010;6:307–316. doi: 10.1007/s12015-010-9143-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Ratajczak MZ, Shin DM, Ratajczak J, Kucia M, Bartke A. A novel insight into aging: are there pluripotent very small embryonic-like stem cells (VSELs) in adult tissues overtime depleted in an Igf-1-dependent manner? Aging. 2010;2:875–883. doi: 10.18632/aging.100231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Ratajczak J, Shin DM, Wan W, Liu R, Masternak MM, Piotrowska K, et al. Higher number of stem cells in bone marrow of circulating Igf-1 level low Laron dwarf mice–novel view on Igf-1, stem cells and aging. Leukemia. 2011;25:729. doi: 10.1038/leu.2010.314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zuba-Surma EK, Klich I, Greco N, Laughlin MJ, Ratajczak J, Ratajczak MZ. Optimization of isolation and further characterization of umbilical-cord-blood-derived very small embryonic/epiblast-like stem cells (VSELs) Eur J Haematol. 2010;84:34–46. doi: 10.1111/j.1600-0609.2009.01352.x. [DOI] [PubMed] [Google Scholar]
  • 195.Zuba-Surma EK, Ratajczak MZ. Overview of very small embryonic-like stem cells (VSELs) and methodology of their identification and isolation by flow cytometric methods. Chapter 9. Curr Protoc Cytom. 2010;4 doi: 10.1002/0471142956.cy0929s51. Unit 9.29. [DOI] [PubMed] [Google Scholar]
  • 196.Snyder EY. The risk of putting something where it does not belong: mesenchymal stem cells produce masses in the brain. Exp Neurol. 2011;230:75–77. doi: 10.1016/j.expneurol.2011.03.012. [DOI] [PubMed] [Google Scholar]
  • 197.Kucia M, Ratajczak J, Ratajczak MZ. Are bone marrow stem cells plastic or heterogenous–that is the question. Exp Hematol. 2005;33:613–623. doi: 10.1016/j.exphem.2005.01.016. [DOI] [PubMed] [Google Scholar]
  • 198.Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20:1487–1495. doi: 10.1038/sj.leu.2404296. [DOI] [PubMed] [Google Scholar]
  • 199.Sadan O, Melamed E, Offen D. Bone-marrow-derived mesenchymal stem cell therapy for neurodegenerative diseases. Expert Opin Biol Ther. 2009;9:1487–1497. doi: 10.1517/14712590903321439. [DOI] [PubMed] [Google Scholar]
  • 200.Kadar K, Kiraly M, Porcsalmy B, Molnar B, Racz GZ, Blazsek J, et al. Differentiation potential of stem cells from human dental origin - promise for tissue engineering. J Physiol Phramacol. 2009;60:167–175. [PubMed] [Google Scholar]
  • 201.Borlongan CV, Kaneko Y, Maki M, Yu SJ, Ali M, Allickson JG, et al. Menstrual blood cells display stem cell-like phenotypic markers and exert neuroprotection following transplantation in experimental stroke. Stem Cells Dev. 2010;19:439–452. doi: 10.1089/scd.2009.0340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Jeong JA, Gang EJ, Hong SH, Hwang SH, Kim SW, Yang IH, et al. Rapid neural differentiation of human cord blood-derived mesenchymal stem cells. NeuroReport. 2004;15:1731–1734. doi: 10.1097/01.wnr.0000134846.79002.5c. [DOI] [PubMed] [Google Scholar]
  • 203.Krabbe C, Zimmer J, Meyer M. Neural transdifferentiation of mesenchymal stem cells–a critical review. APMIS. 2005;113:831–844. doi: 10.1111/j.1600-0463.2005.apm_3061.x. [DOI] [PubMed] [Google Scholar]
  • 204.Lu P, Blesch A, Tuszynski MH. Induction of bone marrow stromal cells to neurons: differentiation, transdifferentiation, or artifact? J Neurosci Res. 2004;77:174–191. doi: 10.1002/jnr.20148. [DOI] [PubMed] [Google Scholar]
  • 205.Zoladz JA, Pilc A, Majerczak J, Grandys M, Zapart-Bukowska J, Duda K. Endurance training increases plasma brain-derived neurotrophic factor concentration in young healthy men. J Physiol Pharmacol. 2008;59:119–132. [PubMed] [Google Scholar]
  • 206.Baraniak PR, McDevitt TC. Stem cell paracrine actions and tissue regeneration. Regen Med. 2010;5:121–143. doi: 10.2217/rme.09.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Ratajczak MZ, Lee H, Wysoczynski M, Wan W, Marlicz W, Laughlin MJ, et al. Novel insight into stem cell mobilization-plasma sphingosine-1-phosphate is a major chemoattractant that directs the egress of hematopoietic stem progenitor cells from the bone marrow and its level in peripheral blood increases during mobilization due to activation of complement cascade/membrane attack complex. Leukemia. 2010;24:976–985. doi: 10.1038/leu.2010.53. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES