Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jan 1.
Published in final edited form as: Free Radic Biol Med. 2012 Oct 17;54:26–39. doi: 10.1016/j.freeradbiomed.2012.10.532

Redox Regulation of Stem/Progenitor Cells and Bone Marrow Niche

Norifumi Urao 1, Masuko Ushio-Fukai 1
PMCID: PMC3637653  NIHMSID: NIHMS415447  PMID: 23085514

Abstract

Bone marrow (BM)-derived stem and progenitor cell functions including self-renewal, differentiation, survival, migration, proliferation and mobilization are regulated by unique cell-intrinsic signals and -extrinsic signals provided by their microenvironment, also termed the ‘niche’. Reactive oxygen species (ROS), especially hydrogen peroxide (H2O2), play important roles in regulating stem and progenitor cell function in various physiologic and pathologic responses. The low level of H2O2 in quiescent hematopoietic stem cells (HSCs) contributes to maintain their stemness, whereas a higher level of H2O2 within HSCs or their niche promotes differentiation, proliferation, migration, and survival of HSCs or stem/progenitor cells. Major sources of ROS are NADPH oxidase and mitochondria. In response to ischemic injury, ROS derived from NADPH oxidase are increased in the BM microenvironment, which is required for hypoxia and HIF1α expression and expansion throughout the BM. This, in turn, promotes progenitor cell expansion and mobilization from BM, leading to reparative neovascularization and tissue repair. In pathophysiological states such as aging, atherosclerosis, heart failure, hypertension and diabetes, excess amounts of ROS create an inflammatory and oxidative microenvironment, which induces cell damage and apoptosis of stem and progenitor cells. Understanding the molecular mechanisms of how ROS regulate the functions of stem and progenitor cells and their niche in physiological and pathological conditions will lead to the development of novel therapeutic strategies.

Introduction

Adult stem cells are populations of cells that are able to regenerate the multiple differentiated cell types of the organ in which they reside and self-renew themselves. Bone marrow (BM)-derived stem and progenitor cells play an important role in neovascularization, which is involved in normal development and wound repair, as well as various pathophysiologies such as ischemic heart disease and peripheral artery disease. This process depends on angiogenesis and vasculogenesis (de novo new vessel formation through BM-derived stem and progenitor cells) [1-4]. Hematopoietic stem cells (HSCs) are the most characterized adult stem cells which produce all types of immune cells and maintain blood production for their lifetime. A subpopulation of BM-derived cells called “endothelial progenitor cells” (EPCs) has been identified by their capacity to form endothelial-like cells in vitro and in vivo [1]. However, the definition of EPCs has recently been challenged, as this concept is still lacking of formal proof in the adult and even questioned in embryonic development [5]. Moreover, hematopoietic cells are shown to be derived from endothelial cells during embryonic development in the mouse [6]. Overall, BM-derived cells appear to have a bilineage potential and interconnection between hematopoietic and endothelial cells has been introduced as a new concept [7]. This is supported by various evidence that stem and progenitor cells in the BM including HSCs, EPCs and even myeloid progenitors contribute to neovascularization and tissue repair in various injury models. Moreover, BM-derived progenitor cells isolated with hematopoietic and endothelial makers have been tested in clinical trials, while further optimization is needed regarding their feasibility, safety and benefit in patients with cardiovascular diseases.

HSC and progenitor cell function and fate are regulated by cell-intrinsic signaling and extrinsic cues provided by a distinct microenvironment called the ‘niche’ [8-13]. In the steady state of homeostatic hematopoiesis or under stress conditions such as after irradiation, growth factor stimulation and hematopoietic injury by chemotherapeutic agents, the mechanism of these regulations has been investigated regarding self-renewal, survival, differentiation, proliferation, engraftment (homing from the periphery to the niche) and mobilization (the forced migration of the cells out of the BM niche into the periphery). It is beginning to be understood about cell-intrinsic and cell-extrinsic effects on the functions of stem and progenitor cells which are involved in inflammation, neovascularization and tissue repair after injury or infection.

Reactive oxygen species (ROS) such as superoxide anion (O2•−) 2 and hydrogen peroxide (H2O2) play an important role for stem and progenitor functions. In general, there is a ‘redox window’ hypothesis; appropriate ROS production is required for physiological cellular functions, while excess ROS contribute to pathological conditions. There seems to be a clear correlation between intracellular H2O2 levels and functions in stem and progenitor cells [14-20]. A low level of endogenous H2O2 is involved in maintaining the quiescence of HSCs, whereas a higher level of H2O2 contributes to a greater proliferation, senescence or apoptosis, leading to a premature exhaustion of self-renewal in these cells [21,22]. Thus, keeping H2O2 at low level within the HSCs or stem cell niche is an important feature of stemness that is directly related to the relatively quiescent state of stem cells in vivo. H2O2 at physiological levels activate repair processes that involve recruitment and differentiation of stem/progenitor cells. ROS derived from NADPH oxidase are required for hypoxia and HIF1α expansion in the BM microenvironment in response to ischemic injury [23]. This, in turn, promotes progenitor cell expansion and mobilization from BM, leading to reparative neovascularization and tissue repair. In pathophysiological states such as aging, diabetes, hypertension, atherosclerosis and cardiac infarction, excess amounts of ROS are generated, thus creating an inflammatory and oxidant stress microenvironment, which induces cell damage and apoptosis of stem and progenitor cells.

In this review, we will summarize the recent progress regarding the role of ROS and ROS-mediated BM microenvironment in regulating stem and progenitor cell functions including self-renewal, differentiation, survival/apoptosis, proliferation, migration and mobilization. Given the significant role of BM-derived cells in physiological and pathological conditions, understanding the redox regulation of stem/progenitor cell function and BM niche will lead to the development of stem and progenitor cell- or stem cell niche-targeted therapies.

1. Role of ROS in physiological and pathological cellular function

In general, excess amounts of ROS are detrimental to cells and contribute to various pathologies such as atherosclerosis, heart failure, aging, diabetes and cancer. In contrast, ROS, especially H2O2, at physiological levels function as signaling molecules to mediate various biological responses such as cell proliferation, migration, survival, differentiation and gene expression [24-27]. Cellular ROS levels are temporally and spatially regulated by the fine-tuned balance between ROS generation system and antioxidant enzymes. ROS such as O2•− and H2O2 are generated from a number of sources including mitochondria, NADPH oxidases (NOXs), xanthine oxidase, cytochrome p450 and nitric oxide synthase (through its uncoupling). Since O2•− is produced from oxygen, oxygen concentration or hypoxic condition has a significant impact on total amount of ROS. The O2•− reacts with nitric oxide (NO) to generate peroxynitrite (OONO), thereby inhibiting endothelial function [28], while it can be quickly converted to H2O2 by superoxide dismutases (SODs) such as MnSOD, (SOD2) or Cu/ZnSODs (SOD1) or extracellular SOD (SOD3) [29,30]. H2O2 is catalyzed by catalase [31], glutathione peroxidases (GPx) [32] and Thioredoxin-peroxiredoxin (Trx-Prx) system [33] to non-reactive water (Figure 1). Since H2O2 is relatively stable and does not react with NO, it has been proposed to function as a second messenger in physiological redox signaling. Overall, the levels of ROS are determined by the balance of ROS generation and antioxidant enzyme activity. Harmful effects of ROS on the cells are DNA damage, lipid peroxidation, protein oxidation and inactivation of specific enzymes by oxidation of co-factors, linking to the pathological consequences. It is well known that growth factor signaling is mediated through H2O2 production. The biological effect of ROS in the cell is dependent on their amount and duration, their source and subcellular localization, and type of species (Figure 1). Identifying direct molecular target(s) of ROS in each cell type is important to understand the cellular mechanism of redox regulation.

Figure 1. Brief overview of reactive oxygen species (ROS) reactions and sources.

Figure 1

The biological effect of ROS in the cell is dependent on their amount and duration, their source and cellular localization, and type of species. SOD: Superoxide dismutase, GPx: Glutathione peroxidases, Trx-Prx: Thioredoxin-peroxiredoxin, O2•−: superoxide anion, H2O2: hydrogen peroxide.

NOX generates ROS by catalyzing electron transfer from NADPH to molecular oxygen, O2. Phagocyte NADPH oxidase consists of 2 membrane-bound subunits, gp91phox (NOX2) and p22phox which form the flavocytochrome b558 complex, together with the cytosolic subunits p47phox, and p67phox p40phox and the small GTPase Rac. NOXs have several homologs of NOX2 including Nox1, Nox3, Nox4, and Nox5, as well as the Dual oxidases (Duox), Duox1 and Duox2 [34,35]. Different from phagocytic NADPH oxidase that is normally quiescent but generates a large burst of O2.− (the “oxidative burst”) upon activation, the NOXs constitutively produce low levels of O2.− or H2O2 intracellularly in basal state and are further stimulated acutely by various agonists and growth factors. NOXs are now recognized to have specific subcellular localizations, which is required for localized H2O2 production and activation of specific redox signaling pathways to mediate various functions [27,36]. They are found in various cell systems including endothelial cells, hematopoietic cells, mesenchymal cells and stem cells, and regulate cell migration, proliferation, differentiation, apoptosis, senescence, inflammatory responses and oxygen sensing [34]. NOXs have been implicated in numerous physiologies such as angiogenesis, tissue repair, hematopoiesis and stem/progenitor functions [14,35,37] as well as pathophysiologies such as hypertension, atherosclerosis, cancer and immune disorders [34,38-41]. Of note, NOX2 is involved in not only host defense but also chemotaxis, immune responses, the initiation of antigen cross presentation[41], cell survival[42] as well as immunosuppressing function protecting from autoimmune development[40]. We have demonstrated that NOX2 in stem/progenitor cells including EPCs, and the BM niche play an important role in reparative neovascularization in response to ischemic injury [23,43]. This issue is discussed below.

ROS are also produced from mitochondria as a consequence of aerobic metabolism. Increasingly, mitochondrial oxidants are viewed less as byproducts of metabolism and act as signaling molecules [44]. Both complex I and III of the electron transport chain are thought to be the major sites of mitochondrial ROS production [45,46]. In addition, mutations in either mitochondrial DNA or nuclear DNA that lead to disruption in any of the components of the electron transport chain also promote ROS formation [44]. Ischemia and apoptosis are shown to trigger O2•− production by complex III [47]. Once O2•− is generated, it is immediately converted into H2O2 by MnSOD, (SOD2) or Cu/ZnSODs (SOD1) [29,30]. To avoid the potential damaging effects of H2O2, mitochondria express other antioxidant enzymes such as peroxiredoxins (Prx)3 and Prx5 and glutathione peroxidase. A number of studies suggest that hypoxic conditions increase mitochondrial ROS production, which stabilizes HIF1α protein expression. Moreover, mitochondrial oxidants also act as important signaling molecules to regulate the inflammatory response, autophagy and mitophagy as well as stem cell function [44].

The cross-talk between NOXs and mitochondrial ROS has been reported in various cell systems [48,49]. Hypoxia-induced mitochondrial H2O2 activate NOX via protein kinase C epsilon in pulmonary artery smooth muscle cells [50]. Serum withdrawal-induced mitochondria H2O2 stimulate NOX1 through PI3-kinase-Rac1 axis in human 293T cells [51]. These evidences suggest that mitochondrial H2O2 regulate NOX activity. Conversely, NOX activation induces mitochondrial H2O2 formation by opening of mitochondrial ATP-sensitive potassium channels [48]. Given that H2O2 is highly diffusible molecule, cross-talk between NOXs and mitochondrial ROS may represent positive feed-forward mechanisms that promote sustained H2O2 production and activation of redox signaling. Whether this regulatory mechanism is involved in stem and progenitor cell function is the subject of future investigation. Moreover, not only intracellular H2O2 but also extracellular H2O2 also play an important role in signal transduction, because H2O2 is relatively stable and can across the cellular membrane through aquaporin [52], and exogenously added H2O2 can activate NOXs [50]. Thus, intracellular H2O2 levels and redox signaling can be affected by extracellular H2O2 and H2O2 production from surrounding cells (cell-extrinsic effect). Cell-intrinsic and cell-extrinsic effects of ROS on stem and progenitor cell function are discussed below.

2. Cell-intrinsic effects of ROS on stem and progenitor function

Physiologic induction of ROS in stem and progenitor cells is regulated by growth factor or cytokine stimulation, changes in oxygen and/or energy metabolism, cell status and differentiation. These ROS inducing factors and situations appear to closely link to one another (Figure 2). As illustrated in Figure 3, distinguishing cell-intrinsic and -extrinsic effect of H2O2 is not always possible due to their diffusible nature. However, studies have demonstrated a clear correlation of ROS levels in stem and progenitor cells, as measured by redox-sensitive dyes which detect mainly intracellular H2O2 or O2•−, with their functions or stage of differentiation (Table 1). When the differentiation capacity is examined in the HSCs based on their H2O2 levels, ROShigh cells show higher myeloid differentiation capacity, whereas ROSlow cells retain their long-term self-renewal ability [21]. Some of the gene mutations exhibit the abnormal increase in H2O2 level, which promotes HSCs to exit from quiescence, block the self-renewal capacity and promote stem cell differentiation [18,20]. Moreover, H2O2 level is further higher in myeloid committed progenitor cells compared to HSCs and mechanism of redox regulation within both cell types is different in terms of FoxO-dependency [20]. It has been shown that a low-oxygenic niche in bone marrow limits ROS production, thus providing long-term protection for HSCs from oxidative stress [10,53,54]. This suggests that there are cell-intrinsic (by programming) and passive (by cellular adaptation) regulation for ROS levels. In Drosophila, developmentally regulated, moderately high H2O2 levels in the hematopoietic progenitors promote differentiation through JNK and FoxO activation [55]. Thus, H2O2 has cell-intrinsic effects on HSCs and hematopoietic progenitor cells during normal hematopoiesis. Cell-intrinsic effects of H2O2 on stem and progenitor cell function under stress conditions have also demonstrated. ROS levels in EPCs (which could have hematopoietic potential) is lower than those in mature ECs, which is due to higher expression of antioxidant enzymes (MnSOD, catalase and glutathione peroxidase), and is required for preserving “stemness” such as undifferentiated, self-renewing state under oxidant stress [33].

Figure 2. Situations of physiologic ROS induction in HSCs and progenitor cells.

Figure 2

Growth factor stimulation increases ROS which act as a second messenger in growth factor-mediated redox signaling. Change in oxygen concentration, which is often associated with energy metabolic alteration, actively and passively affects ROS level. Cell status change such as from quiescent to proliferative or migrating (often referred as activated status) involve an increase in ROS and is a physiologically reversible process. By contrast, differentiation, which is normally an irreversible process (such as myeloid commitment of multipotential HSCs) is also concomitant with increased ROS. Although mechanisms are not fully elucidated, these situations which increase ROS are linked to one another. Increase in ROS is achieved by increased their generation and/or decrease in antioxidant(s). Increased ROS may further promote the processes involving the redox alteration as a feed-forward mechanism (orange arrows).

Figure 3. Cell-intrinsic and cell–extrinsic effect of ROS on HSC and progenitor function.

Figure 3

Two major sources of ROS in HSCs and progenitor cells are NADPH oxidase (NOX) and mitochondria electron transport chain (ETC) (red arrows). NOX is localized at the plasma membrane and perhaps at the endosome. Mitochondria may release ROS. Each produced ROS can activate specific molecular target(s) to contribute to cell-intrinsic or cell-autonomous regulation of cellular function. As cell-extrinsic or non-cell-autonomous regulation of HSC or progenitor function, ROS released from NOX or passed through the plasma membrane increase ROS in the extracellular space (solid blue arrows) which may instruct HSC or progenitors by targeting membrane or intracellular molecules and may influence extracellular matrix or soluble factors regulating HSC or progenitor function. ROS produced from other cells in the niche may affect an important cell-cell interaction regulating HSC or progenitor function. In addition, ROS in the extracellular space may regulate the cell-cell interaction in the niche support cells (dashed blue arrow).

Table 1.

Relationship between ROS levels and cellular functions in HSCs, and hematopoietic or endothelial progenitors

Description
of cells
Isolation method
(Source of Cell)
Change in ROS level
(method used to measure)
Function(s) Reference
HSPCs Sca-1+/c-Kit+/Lin-
(BM)
Increased by S1P (DHE) motility,
mobilization from
the BM into the
circulation
[140]
HSPCs Sca-1+/c-Kit+/Lin-
(BM)
Increased by G-CSF or
HGF in vivo (DHE)
motility,
mobilization from
the BM into the
circulation
[57]
LT-HSCs CD34−/Flt3−/Sca-1+/c-
Kit+/Lin- (BM)
Increased by Paf deficiency
(DCFDA)
loss of quiescence [180]
BM-derived
EPCs
lectin+/ac-LDL+
(cultured BM-MNCs at
day 7)
Increased in cells from
STZ-mice (MitoSOX)
Impaired Matrigel
tube formation,
adhesion, and
migration
[181]
Decreased by AMPK
activation in cells from
STZ-mice (MitoSOX)
Improve diabetes-impaired
Matrigel
tube formation,
adhesion, and
migration
EPCs Early outgrowth EPCs Increased by HGF (Amplex
Red in supernatant)
mobilization from
the BM into the
circulation
[68]
HPCs CD34+ (human cord
blood)
Increased by Ras
transduction (Diogenes,
DEPMPO spin-trap, and
AmplexRed)
survival, growth
factor-independent
proliferation
[182]
BM-derived
EPCs
lectin+/ac-LDL+
(cultured rat BM-MNCs
at day 7)
Increased by AGEs
(DCFDA)
Apoptosis, reduced
migration, adhesion
and proliferation
[183]
HSPCs Drosophila lymph
gland
Increased by differentiation
under in vivo physiological
conditions
differentiation [55]
BM-derived
EPCs
lectin+/ac-LDL+
(cultured cKit+ BM-MNCs
at day 7)
Increased by high glucose
(DCFDA and DHE)
Apoptosis [157]
BM-derived
EPCs
lectin+/ac-LDL+
(cultured BM-MNCs at
day 4)
Increased by hemin
(DCFDA)
migration,
proliferation and
differentiation
[184]
BM-MNCs Density gradient
centrifugation (BM)
Increased in db/db mice
(DCFDA)
Reduced
endothelial-like
differentiation
(Flk1+/CD34+
cells)
[163]
BM-MNCs Density gradient
centrifugation (BM)
Increased by hindlimb
ischemia (DHR)
migration, adhesion [43]
HPCs Lin- (BM) Increased by adrenergic
treatment in culture
(DCFDA)
Inhibit proliferation [185]
EPCs or
BM-MNCs
CD34+/CD117(cKit)+
(Circulation)
Increased by hypertensive
rat (L-012 luminescence)
mobilization from
the BM into the
circulation
[186]
EPCs BM-derived EPCs
(BM)
Increased by erythropoietin
(Amplex Red in
supernatant)
migration,
proliferation,
mobilization from
the BM into the
circulation
[67]
HSCs Sca-1+/c-Kit+/Lin-
(BM)
Increased by conditional
deletion of tuberous
sclerosis complex 1
(DCFDA)
loss of quiescence,
rapid proliferation,
apoptosis and
leukemogenesis
[62]
HPCs CD34+/CD38- (human
cord blood)
Decreased by copper
chelator
tetraethylenepentamine
(DCFDA)
expansion in culture [187]
HSPCs CD34+ (human cord
blood)
Decreased by hypoxia in
culture (DCFDA)
expansion of
CD34+/CD38−
[129]
HSPCs Sca-1+/c-Kit+/Lin-
(BM)
Increased in Fancc−/− mice
(DCFDA)
inhibit self-renewal
or proliferation,
premature
senescence
[188]
Increased by TNFa
(DCFDA)
HSPCs CD45+/Lin- (BM) ROS high (DCFDA) myeloid skewed
ROS Low (DCFDA) quiescence, self-
renewal,lymphoid
skewed
[21]
HPCs 32Dcl3 (cell line) Increased by interleukin-3
or erythropoietin (DCFDA)
proliferation (G1 to
S transition)
[189]
HSCs Sca-1+/c-Kit+/Lin-
(BM)
Increased by conditional
deletion of FoxO1/3/4
(DCFDA)
loss of quiescence
and defective
repopulating
capacity
[20]
HSCs Sca-1+/c-Kit+/Lin-
(BM)
Increased by buthionine
sulfoximine or in Atm−/−
mice (DCFDA)
loss of quiescence
and defective
repopulating
capacity
[18]
HPCs MO7e and B1647
(megakaryocytic cell
lines)
Increased by
thrombopoietin,
granulocyte-macrophage
colony-stimulating factor,
or stem cell factor
(DCFDA)
glucose transport
activity
[42]
HPCs MO7e (megakaryocytic
cell line)
Increased by granulocyte-macrophagecolony-
stimulating
factor,
interleukin-3, steel factor
and thrombopoietin
(DCFDA)
proliferation [56]

Description of stem and/or progenitor cells is according to the original articles. HSPCs: hematopoietic stem and progenitor cells, LT-HSCs: long-term (repopulating) hematopoietic stem cells, HPCs: hematopoietic progenitor cells, EPCs: endothelial progenitor cells, BM: bone marrow, MNCs: mononuclear cells, ac-LDL: acetylated low density lipoprotein uptake, S1P:sphingosine-1-phosphate, DHE: dihydroethidium, G-CSF: granulocyte colony stimulating factor, HGF: hepatocyte colony stimulating factor, DCFDA: dichlorofluorescein diacetate, AGEs:advanced glycation end products, DHR: dihydrorhodamine, TNFa: tumor necrosis factor alpha, Fancc: Fanconi anemia proteins, particularly the complementation group C. Studies investigating leukemia or leukemic cell lines are excluded from the list.

Hematopoietic growth factors or cytokines stimulate signaling events leading to cell growth [56] or promote HSCs mobilization into the circulation [57] through the formation of H2O2. In addition, hyperbaric oxygen stimulates recruitment and differentiation of circulating stem/progenitor cells in subcutaneous Matrigel by increasing H2O2 [58]. Thus, H2O2 at appropriate levels contributes to proliferation and migration of HSCs (which links to mobilization from the BM to the circulation as well as homing to the target sites). By contrast, H2O2 at excess amount activates p38 MAPK to limit the lifespan and self-renewing capacity or expansion of HSCs, resulting in premature senescence phenotype or apoptosis [18,59]. The concept of the redox window suggests that optimal levels of ROS are required for normal responses while excess or insufficient levels of ROS are associated with cellular dysfunction and reduced growth factor signaling, respectively [60]. This notion also seems to apply to the redox regulation of stem and progenitor cell function [22]. The relationship between the change in H2O2 levels in stem and progenitor cells and their functional consequence are summarized in Figure 4. Note that in most cases, the exact source of ROS in these cells is not clear, because of the difficulty of measuring ROS, which are diffusible and short-lived, in current available detecting methods.

Figure 4. The relationship between ROS levels and stem and progenitor cell fate and function in the homeostatic state.

Figure 4

The bone marrow regulatory niches include hypoxic or normoxic (less hypoxic) niche axis. Given oxygen (O2) is required for ROS generation, ROS level or redox status of stem or progenitor cells is correlated with O2 availability. In hematopoietic stem cells (HSCs), especially ones in the quiescent state, oxidative metabolism is suppressed and NADPH oxidase (NOX) enzyme expressions are low, thereby ROS generation from mitochondria and NOX is limited (ROS low). During differentiation or migration of HSCs or in hematopoietic progenitor cells (HPCs), higher ROS (ROS high) are observed with increased mitochondrial ETC (electron transport chain) activities and/or NOX expressions and serve as signaling molecules to promote self-renewal (proliferation), differentiation, migration and survival, which in turn contribute to maintain hematopoiesis and immune function. Antioxidant enzymes play an important role in regulating basal level of ROS or in the cellular adaptation in response to altered ROS generation. These include catalase, Manganese superoxide dismutase, Cu-Zn superoxide dismutase, glutathione peroxidases and peroxiredoxins. On the other hand, further increase in ROS (ROS high) with imbalance between ROS generation and anti-oxidant activity often links to apoptosis, senescence, and oncogenesis or leukemogenesis caused by pathologic HSCs.

3. Sources of ROS regulating stem and progenitor cell function

Although there are several sources of ROS, NOX and mitochondria are major ROS generating enzymes in BM-derived stem and progenitor cells. The connection between mitochondrial ROS and stem cell function has been indicated. Liu J et al. [61] demonstrated that mice lacking the Polycomb repressor Bmi-1 mice exhibit increase in mitochondrial dysfunction, reduced ATP, increased H2O2 levels, and subsequent DNA damage in BM cells. It has been shown that the tuberous sclerosis complex (TSC)-mammalian target of rapamycin (mTOR) pathway, a key regulator of cellular metabolism, maintains the quiescence and function of HSCs by repressing mitochondrial biogenesis and the production of H2O2 [62]. This result implicates the role of mitochondrial ROS in inhibiting stemness of HSCs. Low level of H2O2 production from mitochondria may be important in metabolic adaptation under conditions of low oxygen as well as regulating biological function of stem and progenitor cells. This point should be further clarified in future studies.

Because of availability of gene silencing and mutant mice for NOX enzymes, the role of NOX-derived ROS in stem and progenitor cell functions has been extensively investigated. In murine hematopoietic progenitor cells under homeostatic conditions, all NOXs are expressed [63]. Piccoli et al. showed that human G-CSF-mobilized CD34+ HSCs express NOX1, 2 and 4, which generate low levels of ROS [64,65]. In their sequential study, the high resolution imaging of HSCs with the immunodetection of NOX indicates the presence of membrane ‘rafts’-like microcompartments where the assembly/activation of the NOX components may be functionally integrated for creating redox signaling platforms [66]. They suggested that NOXs-derived ROS play an important role in differentiation from stem and progenitor cells. Among NOXs, NOX2 is most abundantly expressed in murine BM mononuclear cells [43] and, murine and human EPCs [67]. The important role of NOX2 is also demonstrated in the BM after ischemic tissue injury. NOX2 expression and ROS production are increased in BM mononuclear cells [43] and differentiated myeloid cells after hindlimb ischemia of mice [23], which stimulates stem progenitor cells expansion and mobilization from BM [23,43]. Nox2 is also involved in HGF-[68], hypoxia- [67] induced stem/progenitor cell mobilization from BM to peripheral blood. These studies indicate that NOX2-derived ROS mediate growth factor and chemokine signaling involved in progenitor cell and EPC migration, proliferation and survival under stress conditions. Thus, NOXs play a role in maintaining adequate ROS levels in HSC and hematopoietic/endothelial progenitor cells and contribute to their physiological function.

NOX-derived ROS are involved in maintenance of stemness and cardiovascular differentiation of embryonic stem (ES) cells. Undifferentiated self-renewing ES cells generate low level of endogenous ROS with low NOX enzyme expression, and NOX is dynamically regulated during ES cell differentiation to cardiomyocytes [69]. Endothelial differentiation from mouse ES cells involves ROS from NOX2, NOX1 and NOX4 [70,71], while smooth muscle cell differentiation is mediated by NOX4-derived H2O2 [72]. ROS generation is elevated during the early stages of ES cell differentiation, and then downregulated during later stages. During the differentiation process, anti-oxidative genes are downregulated [73] while NOX1, 2 and 4 are upregulated [74], thereby increasing ROS levels. Stimulation of ES cells with mechanical strain [71] or direct current electrical field [70,75,76] or low concentration of H2O2 [69,74] or various agonists including cardiotrophin-1 [77], PDGF-BB [78] or peroxisome proliferator-activated receptor α [79] induces cardiovascular differentiation of ES cells through increase of H2O2. This in turn induces upregulation of NOX1 and 4, thus initiating a feed-forward stimulation of prolonged ROS generation [71,74]. Of note, NOX4 is involved in differentiation of ES cells to cardiomyocytes [80] and smooth muscle cells [72] while NOX2 is closely correlated to the differentiation of phagocytic cells from ES cells [81]. H2O2 derived from NOX activates ERK1/2, p38 and JNK [71], or various cardiogenic transcription factors [74], which are required for cardiomyogenesis of ES cells.

4. Regulators of ROS involved in stem and progenitor cell function

There are various regulators of ROS and their molecular targets involved in stem and progenitor cell function as described below (Figure 5).

Figure 5. Signaling pathways mediated by ROS involving stem cell fate.

Figure 5

ROS allow stem cells to shift from the quiescent state to the functional state such as differentiation and migration. ROS can promote the survival pathway, but also lead to senescence. ROS modulate the activities of various kinases and phosphatases, which in turn activate redox-sensitive signaling cascades. Of note, many of these molecules have also been shown to regulate basal ROS levels in stem cells, suggesting that feed-forward or feed-back mechanism by which stem cells respond to redox state and oxidative stress. Please see the main article for the details.

Forkhead homeobox type O (FoxO)

The forkhead homeobox type O (FOXO) transcription factors FOXO1, FOXO3a, and FOXO4 are critical mediators of the cellular responses to oxidative stress and play a pivotal role in the redox regulation of HSCs [82]. FoxO3a−/− HSCs show increased phosphorylation of p38MAPK and H2O2 as well as downregulation of antioxidant enzymes, defective maintenance of quiescence, and heightened sensitivity to cell-cycle-specific myelotoxic injury [83]. It is shown that H2O2 act through p38MAPK to limit HSC lifespan [18]. Thus, excess H2O2-p38MAPK pathway may be involved in inhibition of self-renewal function of FoxO3a−/− HSCs. Furthermore, under stress conditions, such as aging or 5-FU-induced myelosuppression, FoxO3a−/− mice develop neutrophilia associated with increased Akt and ERK and decrease of Spred2 (Sprouty-related Ena/VASP homology 1 domain-containing proteins 2). Thus, FoxO3a plays a pivotal role in maintenance, integrity, and stress resistance of HSCs through negative feedback pathways for proliferation [84]. Conditional FoxO1/3/4 knockout mice exhibit myeloid lineage expansion and a marked decrease in lineage-negative Sca1+, cKit+ (LSK) compartment and long-term repopulating activity that correlated with increased cell cycling, apoptosis and H2O2 in HSCs [20]. These FoxO-deficient HSC phenotypes are rescued by in vivo treatment with the antioxidant N-acetyl-L-cysteine (NAC). Thus, FoxOs proteins protect against oxidative stress and thereby maintain self-renewal capacity (quiescence) and enhanced survival in the HSC compartment, which is required for its long-term regenerative potential [20].

Akt, PTEN and mTOR

Akt phosphorylates FoxO to promote transition from quiescent status to myeloid-biased activated HSCs. HSCs isolated from Akt1−/−/Akt2−/− mice show defective differentiation of HSCs into multipotent progenitors by decreasing H2O2 levels [85]. Constitutively active Akt accelerates proliferation and increases H2O2 level in HSCs, resulting in depletion of HSCs, BM failure as well as myeloproliferative disease or acute myeloid leukemia [86]. Thus, H2O2 is determinant factor for myeloid commitment and its appropriate level is important for normal HSC function. In addition, H2O2-mediated enhancements in self-renewal and neurogenesis are dependent on PI3K/Akt signaling [87]. PTEN is negative regulator for PI3K/Akt pathway and contains catalytic cysteine residues that are highly susceptible to oxidation by H2O2 [88,89]. Therefore, PTEN inhibition stabilizes the active phosphorylated form of Akt. Conditional PTEN−/− mice show rapid depletion of long-term repopulating HSCs and promoting myeloproliferative diseases, which are restored by mTOR inhibition [90]. Importantly, the depletion of PTEN-deficient HSCs is not mediated through ROS [91], while the defect by FOXO deficiency is rescued by NAC [20]. G-CSF-induced mobilization of hematopoietic progenitor cells into the circulation is mediated via increase in c-Met expression and its downstream mTOR-FOXO3a-mediated accumulation of H2O2 [57]. Therefore, H2O2-dependent and -independent mechanisms may be involved in the regulation of myeloid commitment in HSCs. Tuberous sclerosis complex (TSC) has been shown to regulate ROS levels in HSCs. Tsc1 deletion in the HSCs drives them from quiescence into rapid cycling, with increased mitochondrial biogenesis and H2O2, and TSC-mediated decrease in H2O2 is mediated through mTOR inhibition[62]. Interestingly, the deletion of AMP kinase (AMPK), a downstream of TSC and a metabolic kinase, does not affect H2O2 levels [91]. Therefore, linkage between ROS and metabolic pathways appear to be complex, while both clearly regulate HSC functions.

ATM, p38MAPK and p53

The ‘ataxia telangiectasia mutated’ (ATM) gene maintains genomic stability by activating a key cell-cycle checkpoint in response to DNA damage, telomeric instability or oxidative stress. Physiological levels of H2O2 are required to maintain genomic stabilities by activating the DNA repair pathway via ATM in cardiac and ES cells [92]. HSCs from ATM−/− mice have increased levels of H2O2 [17], leading to defective reconstitutive capacity of HSCs. Thus, the reduction of intracellular ROS levels by ATM is required for maintaining the self-renewal ability of HSCs. Mechanistically, H2O2 elevation due to ATM deficiency in HSCs activates p38MAPK, which upregulates the CDK inhibitors p16Ink4a and p19Arf [17]. Treatment with NAC rescues the defects in HSC function in ATM-deficient mice [17], suggesting that elevation of H2O2 can exit HSCs from quiescence and reduces self-renewal capacity [17,20,93]. Furthermore, overexpression of polycomb RING-finger oncogene BMI1 in normal human neural stem cells directly enhances ATM recruitment to sites of DNA damage, leading to protection from ultraviolet radiation [94] presumably by preventing generation of H2O2 [61]. Prdm16, a zinc finger transcription factor, is shown to be involved in the maintenance of stem cell function by modulating the intracellular redox state. Prdm16 deficiency increases mitochondrial H2O2 through BMI1, resulting in the depletion of stem cells, cell death and altered cell-cycle distribution [95]. In addition, ATM-mediated phosphorylation of BID, a BH3-only BCL2 family member, plays an important role in maintaining the quiescence and survival of HSCs via reducing oxidative stress [96]. Thus, the ATM-BID pathway serves as a critical checkpoint for coupling HSC homeostasis and the DNA-damage stress response to enable long-term regenerative capacity. Importantly, physiological levels of intracellular H2O2 are required to activate the DNA repair pathway for maintaining stem cell genomic stability [92]. This finding suggests that the concept of “redox window” or “oxidative optimum” can be also applied for genomic stability in stem cells.

p53, a major tumor suppressor gene, has been implicated in regulation of HSC quiescence and self-renewal. Activation of p53 depletes stem cells via H2O2 accumulation, and Mdm2, an E3 ubiquitin ligase that targets p53 for degradation, is required for survival of HSCs/progenitors via dampening of H2O2 -induced p53 activity [97]. Thus, excess activation of p53 in the absence of Mdm2 induces a dysregulated p53- H2O2 positive feedback loop, indicating that an appropriate level of p53 and H2O2 is essential for the maintenance of HSCs [98].

5. Role of BM niche in regulating stem and progenitor functions

Stem cells are localized to niches formed by cells that provide a microenvironment that provides essential cues supporting their growth and fate decisions in the BM [8,10,13,54,99,100]. Interaction of stem cells with the niche is crucial for the long-term maintenance of quiescence. HSC niche consists of sinusoidal endothelial cells (ECs), sympathetic nerve fibers, cells of the osteoblastic lineage, osteoclasts [101], perivascular mesenchymal stem cells [102], macrophages [103,104], regulatory T cells [105] and other HSC progenies [9] (Figure 6). In addition to these cellular components, soluble factors such as cytokines and growth factors, extracellular matrix, oxygen concentration or hypoxia, and ROS are also the part of the niche. Extrinsic instructions provided by unique microenvironments regulate the fate and functions of HSCs and progenitors. The number of studies on the niche are reviewed by others focusing on the mechanism of HSC maintenance (quiescence and self-renewal) [10], leukemia development and chemotherapy resistance [106,107], organization of niches [11,13], HSCs mobilization (HSC egress from the niche and trafficking into the blood) and homing to the BM [108-111].

Figure 6. Cellular components of stem and progenitor niche and potential regulation through ROS.

Figure 6

Hematopoietic Stem and Progenitor cells (HSPCs) reside in a niche that consists of cellular and non-cellular components. Cellular components include stem or progenitor cells, stromal cells, neurons, immune cells, osteoblastic cells, osteoclast and endothelial cells as well as the progeny of stem or progenitor cells. These cellular niche components regulate stem and progenitor cells directly through cell-cell interactions or indirectly through modifying non-cellular components including secreted neurohormonal factors, growth factors and enzymes, and extracellular matrix and oxygen (O2) or hypoxia, as well as extracellular ROS.

6. Role of ROS, hypoxia and HIF1 α in regulating BM niche

The stem cell niche includes the low oxygen endosteal niche mainly containing quiescent hematopoietic stem cells as well as more oxygenated vascular niche containing proliferative and differentiated hematopoietic progenitors [53,99,100]. This concept is coupled with the hypothesis of passive adaptation to regulate H2O2 generation in HSCs; a low-oxygen niche in BM limits H2O2 production, thereby providing long-term protection for HSCs from oxidant stress. Thus, hypoxic microenvironment is an important determinant for the maintenance of “stemness”, and regulates stem cell self-renewal and differentiation [112]. In vitro, characteristics of undifferentiated HSCs such as colony-forming ability or reconstitution capacity are retained after hypoxic culture compared with normoxic conditions [113-115]. In vivo, Hoechst33342 dye perfusion assay reveals that long term-HSCs and a part of osteoblasts are found predominantly in poorly-perfused hypoxic niches in the BM compared to ECs and mesenchymal stem cells which show medium to high perfusion [116]. This suggests that the most quiescent HSCs localize in a region far from blood supply that delivers oxygen. The low ROS population has a higher self-renewal potential and reconstitution capacity following serial BM transplantation [21]. Thus, hypoxia and low blood perfusion seem to be correlated with low ROS level in HSCs in the BM. However, putative HSCs can also be localized at perivascular area of BM with low oxygen and high HIF1α expression [117]. Although it might be difficult to anatomically identify and define the hypoxic and relatively oxygenated microenvironment in the complex structure of BM [8], there seems to be at least theoretical hypoxic niche in the BM that leads to lower H2O2 production. Alternatively, the existence of hypoxic HSCs with perivascular localization may suggest that intracellular hypoxia can be achieved actively, not simply passively, by cell-intrinsic regulation through HIF1α[117].

In addition to the cell-intrinsic effects of H2O2 in HSCs, H2O2 produced from cells in the niche around the HSCs may act as extrinsic factors to regulate HSC functions. Cell-extrinsic effects of H2O2 are defined as HSC regulation through redox modification of niche components including cellular components, extracellular matrix and soluble factors (Figure 3). Of note, the diffusible nature of H2O2 and the difficulty of its measurement in situ often prevent investigators from identifying an action point of H2O2. Several evidence suggests that the osteoblasts, cells of mesenchymal origin positioned at the endosteal surface of bone, are essential components of the HSC niche [99]. Tie2/Angiopoietin1 (Ang1) signaling is required for the maintenance of HSCs in a quiescent state in the BM niche [17]. The role of N-cadhernin in the BM niche and regulation of HSC is a point of controversy [118-120]. Regardless, increase of H2O2 in HSCs by anti-cancer drug has been reported to suppresse N-cadherin expression in osteoblastic niche and to induce shift of side population (SP) cells to non-SP cells, allowing quiescent HSCs detached from the niche [83]. Redox-dependent expression of vascular cell adhesion protein 1 on ECs in the BM is required for the early stages of BM homing and localization of HSC after irradiation [121]. Autocrine factors from Akt-mTOR-activated ECs support the self-renewal of long-term (LT)-HSCs (which contribute to hematopoiesis for long term) and expansion of hematopoietic stem and progenitor cells (HSPC), whereas MAPK co-activation favors maintenance and lineage-specific differentiation of HSPCs [122]. EC-derived growth factors support in vitro self-renewal and in vivo repopulation of authentic LT-HSCs through Notch [122]. Although further mechanistic studies are required, these reports suggest that ROS-mediated osteoblast or EC modification may regulate HSCs-niche interaction, resulting in modulating HSC function (Figure 3). Most recently, Taniguchi et al. have shown that hematopoietic connexin-43 prevents HSC senescence by reducing ROS level in HSCs through transferring ROS to BM stromal cells [123]. This finding indicates the novel mechanism for the niche-mediated regulation of ROS levels in HSCs.

BM macrophages maintain the endosteal niche and their depletion by G-CSF and clodronate-loaded liposomes induce HSC mobilization into the blood [103]. Similarly, specific depletion of CD169+ macrophages localized in mesenchymal niche by a CXCR4 antagonist or G-CSF induces egress of HSC/progenitor cells [104]. Extensive studies are required to understand the niche regulation under stress conditions such as inflammation, in which HSC is differentiated into immune cells, and HSC and progenitor cells are mobilized from the BM to the blood circulation [124]. Thus, modulating the BM stem cell niche is important for developing novel therapeutic strategies [125,126].

Most recently, we performed in vivo injection of O2•− reactive probe and hypoxic bioprobe into mice and showed that ischemic injury increases ROS through NOX2 predominantly at the central BM in situ and at lesser extent at the endosteal regions [23]. Of note, NOX2-derived ROS are increased mainly in differentiated myeloid cells in the BM, thereby creating an oxidative BM microenvironment. We also showed that ischemic injury induces expansion of low oxygen (hypoxic) areas throughout the BM, in a NOX2-dependent manner. This in turn regulates HSPCs expansion, survival and mobilization from the BM, leading to neovascularization and tissue repair [23] (Figure 7). Consistently, granulocyte colony stimulating factor (G-CSF) or cyclophosphamide (CY), which stimulates mobilization of stem/progenitor cells, increase hypoxia in the BM [127]. Of note, G-CSF and CY-induced stem cell mobilization is blunted in NOX2−/− mice [128]. Fan et al. showed that increase in ROS is associated with a decreased oxygen percentage in CD34+ HSPCs [129]. Piccoli et al. [64] reported that the half of the oxygen consumption in HSPCs is dependent on NOX. In addition, the consumption of respiratory burst by NOX2 in differentiated myeloid cells is shown to increase local hypoxia [130]. Thus, it is likely that ischemic injury increases oxidative microenvironment mainly due to activation of NOX2 in differentiated myeloid cells, which in turn creates hypoxic niche throughout the BM by increasing oxygen consumption. These ROS-hypoxia-mediated alterations of the BM niche induced by inflammation or tissue injury may regulate stem and progenitor expansion and mobilization from BM, thereby promoting tissue repair and regeneration (Figure 3) [23]. Of note, hypoxia culture (2% O2) rather suppresses HSC proliferation under growth factor stimulation regardless of NOX2 expression (N.U. and MU-F. unpublished observation). This suggests that above mentioned NOX2-ROS-mediated increase in hypoxic microenvironment is achieved in more oxygenic condition to promote progenitor cell expansion.

Figure 7. NADPH Oxidase 2 (NOX2)-derived ROS promote hematopoietic stem/progenitor cell (HSPC) expansion and mobilization in response to ischemic injury.

Figure 7

Ischemic injury induces expansion of low oxygen (hypoxic) area, hypoxia inducible factor-1 (HIF-1) expression and Akt activation throughout the BM, in a NOX2-dependent manner. This, in turn, regulates HSPCs expansion and mobilization from BM. Hypoxia might be induced by ROS generation which consumes oxygen, especially at the sites where oxygen supply is limited, such as the bone marrow cavity. Our data also showed matrix metalloproteinases (MMPs) are regulated by NOX2-derived ROS. These ROS-hypoxia-mediated alterations of the BM microenvironment induced by inflammation or tissue injury may play an important role in regulating stem and progenitor function to promote tissue repair and neovascularization. See ref. 24 for the details.

HIF1α is a key regulator of hypoxia, metabolic and angiogenic response. HIF1αis highly expressed in LT-HSCs [117,131]. HSCs derived from conditional HIF1α knockout mice exhibit impaired reconstitution capacity [117]. HSCs utilize glycolysis instead of mitochondrial oxidative phosphorylation to meet their energy demands through HIF1α[131]. This anaerobic-biased energy metabolism promotes HSC maintenance by limiting ROS production [54]. Activation of HIF1α or treatment with the HIF stabilizer reduces HSC reconstituting ability under normoxic conditions [15], which is supported by the study using the genetic mouse model [117]. In C. elegans, mild reduction in mitochondrial respiration leads to the increase in H2O2 and HIF1α that are required for the acquisition of a long-life span [132]. These indicate that appropriate levels of HIF1α and H2O2 are responsible for the maintenance of HSCs and other stem cells. Moreover, NOX-derived ROS contribute to HIF1α stabilization in HSCs in normoxic conditions by down-regulation of the tumor suppressor von Hippel-Lindau protein (pVHL) [65]. Thus, HIF1α and ROS closely work together, along with oxygen homeostasis and energy metabolism, to maintain HSC function. In the stress response of stem and progenitor cells to ischemic injury, HIF1α in BM-derived cells promote angiogenesis [133]. Ex vivo cultured BM-derived angiogenic cells treated with the prolyl-4-hydroxylase inhibitor, which increases HIF1α and HIF2α expression, improves angiogenesis of ischemia hindlimb in old mice [134]. We have demonstrated that endosteum at the BM is hypoxic with high expression of HIF1α in basal state. In response to ischemia, NOX2-derived ROS are increased in both the endosteal and central region of BM tissue, which promotes HIF1α and VEGF expression with expansion of hypoxic areas in the BM in situ [23](Figure 7). Thus, NOX-ROS-mediated BM niche modification by ischemic injury may regulate hypoxia response in BM progenitor cells, promoting their mobilization from BM.

7. Other ROS-dependent regulators in the BM niche

ROS are involved in niche-mediated growth factor/chemokine receptor signaling through regulating its ligand expression. SDF-1α, which plays a role in stem and progenitor cell mobilization and vascular repair, regulates the trafficking of HSCs progenitors and maintaining HSC niches in BM [135]. SDF-1α is released by stromal cells and binds to its CXCR4 receptor on stem and progenitor cells. The high SDF-1α content in the BM creates a concentration gradient, which retains HSCs within the stem cell niche. Disruption of this SDF-1α gradient promotes mobilization of stem cells into the circulation, which occurs after upregulation of G-CSF levels during systemic stress or injury. In response to ischemia, myocardial infarction or hypoxia, tissue levels of SDF-1α are increased [136-139], which may attract stem cells to sites of tissue injury and ischemia. The SDF-1-CXCR4 axis induces cMet activation in the BM, which promotes G-CSF-induced mobilization of progenitor cells via increasing ROS [57]. Most recently, Golan et al. reported that sphingosine-1 phosphate (S1P) promotes hematopoietic progenitors and BM stromal cell mobilization as well as SDF-1 release via ROS [140]. Thus, dynamic cross-talk between S1P and SDF-1 via ROS signaling integrates BM stromal cells and hematopoietic progenitor cell motility.

Ischemic injury increases cytokines and VEGF in the BM and circulation, which in turn activates matrix metalloproteinase (MMP)-9 and releases soluble Kit ligand in the BM [2]. MMPs including MMP-9, which is secreted mainly by neutrophils in BM [141], and MT (membrane type)1-MMP [142], which is anchored on the cell surface, plays a significant role in stem/progenitor cell mobilization and angiogenesis. We have demonstrated that NOX2-derived ROS increased in the BM after ischemic injury regulate HSPCs function in part through regulating Akt activation, expression of MT-1-MMP, and MMP-9 activity. Therefore, ROS regulate extracellular matrix in the BM niche. Taken together, understanding mechanisms by which ischemic injury regulates BM microenvironment is essential for developing novel therapeutic strategies for various ischemic diseases.

8. Role of ROS in stem and progenitor cell function in pathological conditions

In pathological conditions such as aging, atherosclerosis and diabetes, excess amount of ROS (oxidative stress) in stem and progenitor cells as well as BM microenvironment may impair stem and progenitor function, which can inhibit HSC self-renewal and induce HSC senescence, resulting in premature exhaustion of HSCs and hematopoietic dysfunction. Recent proteomic analysis of BM stromal cells in culture reveals that older stromal cells produce more H2O2 than younger cells [143]. Thus, both intrinsic dysregulation of ROS and more oxidative environment may have deleterious effects on stem and progenitor function. Although the definitions of EPCs has been challenged [7,16], inverse correlation between circulating number of EPCs and cardiovascular risk has been shown [46,144-154]. In human EPC, angiotensin II accelerates EPC senescence through induction of oxidative stress [148]. Diabetes induces dysfunction and early senescence in stem and progenitor cells. In animal model of type I diabetes, O2•− production by eNOS uncoupling leads to reduction of EPC levels and impairment of EPC function [155]. In EPC culture, high glucose promotes EPC proliferation at early stage (3 days) and inhibits at later phase (7 days) through H2O2 accumulation [156]. The p66Shc deletion rescues the BM-derived EPCs defect induced by oxidative stress in high glucose [157]. Human EPCs from type II diabetes exhibit impaired proliferation, adhesion and incorporation into vascular structures [3]. Decreasing O2•− restores defective ischemia-induced new vessel formation induced by the glyoxalase 1 substrate methylglyoxal-mediated modification of HIF1α in EPCs [158], indicating a causal role of ROS in EPCs dysfunction in diabetes.

Circulating progenitors from healthy subjects have lower levels of H2O2 due to higher expression of the antioxidants enzymes including MnSOD, GPx, and catalase compared with human umbilical vein ECs [33]. Indeed, dysfunction of antioxidant defenses links to impaired function of EPCs; GPx-1−/− mice have no increase in circulating EPCs in response to either VEGF treatment or ischemic injury. GPx-1−/− EPCs are functionally deficient in promoting angiogenesis in vivo and in vitro, and show an increased susceptibility to oxidative stress in vitro [159]. Apoptosis signal-regulating kinase 1 (ASK1) is controlled by multiple redox-sensitive proteins including thioredoxin, glutathione-S-transferases, and glutaredoxin [16]. Ingram et al. showed that H2O2-induced increase in ASK1 activity is involved in diminished vessel-forming ability of EPCs after oxidant stress [160]. Moreover, decreased circulating progenitor cells and their dysfunction are associated with inflammation [154]. In addition, the H2O2-p38MAPK pathway accelerates senescence of EPCs by inducing pro-senescence molecule p16(INK4a) [161] in the same manner with quiescent HSCs. This indicates that HSC and EPC, or their progeny share the common pathway regarding premature senescence through excess amount of ROS.

9. Therapeutic potential of redox regulation of stem/progenitor cells and their niche

For a last decade, cytokines, chemokines and growth factors, which promote stem and progenitor egress from their niche or the mobilization from BM to the circulation, have been tested for their therapeutic potential in patients with cardiovascular diseases. The clinical trials of cell therapy using BM progenitor cells demonstrate its feasibility, safety and potential benefit in patients with ischemic disease and heart failure, but reveal that current cell-based therapy needs to be optimized to improve therapeutic efficacy [162]. In this regard, studies investigating the therapeutic potential of redox regulation of these cells have used two different strategies. The first approach is suppressing excess oxidative stress in stem and progenitor cells. Experimentally, EPC dysfunction prevents new blood vessel growth, which is restored by manipulations to decrease ROS. In vivo administration of SOD mimetic attenuates the diabetes-related impairment of BM mononuclear cells by reducing oxidative stress [163]. Thus, strategies aimed at reducing hyperglycemia-induced ROS is a useful antihyperglycemic therapies in the restoration of vasculogenesis and the prevention of diabetic complications [164]. Either transgenic expression of MnSOD or administration of SOD mimetic rescue impaired post-ischemic neovascularization and tissue survival [158]. Angiotensin II receptor and β1-adrenoceptor blockers improve the EPC dysfunction in hypertension via an antioxidant effect [165,166]. Treatment with organic nitrates increases circulating EPC levels, while increased NOX-derived ROS by isosorbide dinitrate induces their dysfunction [167,168]. Mesenchymal stem cell engraftment in the infarct heart is enhanced by anti-oxidant NAC co-injection [169]. Hypoxic preconditioning increases the survival and angiogenic potency of peripheral blood mononuclear cells through oxidative stress resistance mechanisms [170]. As a second approach, on the contrary, stimulating progenitor cells with controlled pro-oxidant has also shown to be effective on promoting their neovascular function. For example, short-term pretreatment with low-dose H2O2 enhances the efficacy of BM cells for therapeutic angiogenesis [171]. Injection of BM cells from control mice, but not NOX2-deficient mice, promotes neovascularization in response to tissue ischemia [43], suggesting that NOX2-derived ROS in BM cells is required for this response. In vitro preconditioning that stimulates mitochondrial H2O2 production increases the secretion of proangiogenic properties from adipose-derived stroma cells and the survival of these cells in ischemic tissues after in vivo injection [172]. This suggests that mitochondrial H2O2 generation in stromal cells provides essential cues for stem and progenitor cells to promote neovascularization after injury.

There is doubled-edge effect of ROS whereby physiological levels can serve as signaling molecules to promote vascular integrity [43,173], whereas excess ROS levels in pathological conditions are associated with stem/progenitor dysfunction and/or impaired post-ischemic neovascularization [172,174-176]. Thus, antioxidant therapy in pathological conditions should be carefully designed so that ROS levels are kept optimal and physiological levels in BM stem/progenitor cells and microenvironment. Alternatively, more specific approaches by targeting particular ROS generating or antioxidant systems, or a downstream of ROS-sensitive molecules in stem and progenitor cells may be more effective as a new potential therapy.

Finally, modulating the stem and progenitor niche in vivo would have therapeutic potential for inflammatory- or ischemia-related cardiovascular diseases and this may allow us to stimulate stem and progenitor cells in the longer term. It has been shown that a defective niche results in HSC disorders, further emphasizing the important function of the HSC niche in vivo [177,178]. For a last decade, cytokines, chemokines and growth factors, which promote mobilization of stem and progenitor cells from the niche, have been tested for their therapeutic potential in patients with cardiovascular diseases, while their benefits seem to be relatively limited. As described above, mice lacking essential components of the regulatory system that maintains ROS within the physiological levels, show accelerated HSCs senescence and progressive BM failure [17,20]. In type 1 diabetic mice, the elevation in mitochondrial ROS induces stem/progenitor cell depletion and dysfunction in the BM microenvironment [179]. Thus, targeting against excess levels of ROS in the BM niche or the niche components may provide new therapeutic strategies for treatment of various cardiovascular diseases.

Conclusions

The current review outlines that ROS and ROS-mediated BM niche are involved in stem and progenitor cell functions including self-renewal, differentiation, survival/apoptosis, proliferation, and mobilization. ROS levels in stem and progenitor cells have a clear correlation with cellular functions and are regulated by a fine tuning of the balance between ROS generating and anti-oxidative defense systems. Molecular targets of ROS and distinct redox signaling pathways in stem and progenitor cells have been identified with in vitro and in vivo functional consequences. ROS are also considered as niche factors which regulate stem and progenitor cells through modulating other cellular and non-cellular niche components. The role of ROS in niche modification is beginning to be investigated. Because of the complexity of the BM niche, the diffusible nature of H2O2 and the difficulty of their tracking, it could be challenging to elucidate a dynamic regulation of the BM niche, especially in the pathophysiological conditions such as aging, metabolic disorders, inflammation, response to injury or infection, and autoimmune diseases. However, with the combination of advanced in vitro, in vivo and ex vivo techniques, we will be able to extract important elements for redox regulation of stem and progenitor cells, which may develop novel cell-based and/or niche-targeted therapies.

The niche engineering will be useful to test a hypothetical model and can be directly applied to cell therapy manufacturing that produces beneficial cell populations for regenerative medicine. This interplay may discriminate between pathways resulting in oxidative stress, and induction of apoptosis versus signaling events in stem and progenitor cells. ROS promote HSCs to exit from the self-renewal capacity and function as signaling molecules to promote stem cell differentiation into multi-lineage and larger homing capacity. This may contribute to angiogenic and/or tissue repair function of BM stem and progenitor cells. These mechanisms are regulated by the intrinsic redox control in stem and progenitor cells through various redox signaling pathways as well as by the extrinsic factors generated from the BM niche such as ROS, hypoxia, and cytokines/chemokines. NOX-ROS-mediated hypoxic BM microenvironment induced by ischemic injury increases HIF1α and VEGF expression in BM as well as progenitor cell survival and expansion, thereby promoting their mobilization from BM. Understanding the redox regulation of stem and progenitor cells and BM niche as well as their underlying mechanisms in physiological and pathological conditions will lead to the development of novel therapeutic strategies.

Highlights.

  • Stem/progenitor cell function is regulated by cell-intrinsic and -extrinsic signals.

  • Reactive oxidase species regulate stem/progenitor cell function.

  • Reactive oxidase species regulate bone marrow stem cell niche.

  • Oxidative stress in pathological conditions impairs stem/progenitor cell function.

  • NADPH oxidase contributes to niche modification in response to ischemic injury.

Acknowledgments

This work was supported by funds from National Institutes of Health (NIH) R01 Heart and Lung (HL)077524, HL077524-S1 (to M.U.-F.), American Heart Association (AHA) National Center Research Program (NCRP) Innovative Research Grant 0970336N (to M.U.-F), AHA Post-doctoral Fellowship 09POST2250151 (to N.U.), and AHA Scientist Development Grant 12SDG12060100 (to N.U.).

Footnotes

Disclosure of Potential Conflicts of Interest The authors indicate no potential conflicts of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–967. doi: 10.1126/science.275.5302.964. [DOI] [PubMed] [Google Scholar]
  • [2].Rafii S, Avecilla S, Shmelkov S, Shido K, Tejada R, Moore MA, Heissig B, Hattori K. Angiogenic factors reconstitute hematopoiesis by recruiting stem cells from bone marrow microenvironment. Ann N Y Acad Sci. 2003;996:49–60. doi: 10.1111/j.1749-6632.2003.tb03232.x. [DOI] [PubMed] [Google Scholar]
  • [3].Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz GR, Levine JP, Gurtner GC. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation. 2002;106:2781–2786. doi: 10.1161/01.cir.0000039526.42991.93. [DOI] [PubMed] [Google Scholar]
  • [4].Jin DK, Shido K, Kopp HG, Petit I, Shmelkov SV, Young LM, Hooper AT, Amano H, Avecilla ST, Heissig B, Hattori K, Zhang F, Hicklin DJ, Wu Y, Zhu Z, Dunn A, Salari H, Werb Z, Hackett NR, Crystal RG, Lyden D, Rafii S. Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4+ hemangiocytes. Nat Med. 2006;12:557–567. doi: 10.1038/nm1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Goldie LC, Nix MK, Hirschi KK. Embryonic vasculogenesis and hematopoietic specification. Organogenesis. 2008;4:257–263. doi: 10.4161/org.4.4.7416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Zape JP, Zovein AC. Hemogenic endothelium: origins, regulation, and implications for vascular biology. Semin Cell Dev Biol. 2011;22:1036–1047. doi: 10.1016/j.semcdb.2011.10.003. [DOI] [PubMed] [Google Scholar]
  • [7].Chao H, Hirschi KK. Hemato-vascular origins of endothelial progenitor cells? Microvasc Res. 2010;79:169–173. doi: 10.1016/j.mvr.2010.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Kiel MJ, Morrison SJ. Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol. 2008;8:290–301. doi: 10.1038/nri2279. [DOI] [PubMed] [Google Scholar]
  • [9].Renstrom J, Kroger M, Peschel C, Oostendorp RA. How the niche regulates hematopoietic stem cells. Chem Biol Interact. 2010;184:7–15. doi: 10.1016/j.cbi.2009.11.012. [DOI] [PubMed] [Google Scholar]
  • [10].Takubo K, Ohmura M, Azuma M, Nagamatsu G, Yamada W, Arai F, Hirao A, Suda T. Stem cell defects in ATM-deficient undifferentiated spermatogonia through DNA damage-induced cell-cycle arrest. Cell Stem Cell. 2008;2:170–182. doi: 10.1016/j.stem.2007.10.023. [DOI] [PubMed] [Google Scholar]
  • [11].Levesque JP, Winkler IG. Hierarchy of immature hematopoietic cells related to blood flow and niche. Curr Opin Hematol. 2011;18:220–225. doi: 10.1097/MOH.0b013e3283475fe7. [DOI] [PubMed] [Google Scholar]
  • [12].Weber JM, Calvi LM. Notch signaling and the bone marrow hematopoietic stem cell niche. Bone. 2010;46:281–285. doi: 10.1016/j.bone.2009.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Lilly AJ, Johnson WE, Bunce CM. The haematopoietic stem cell niche: new insights into the mechanisms regulating haematopoietic stem cell behaviour. Stem Cells Int. 2011;2011:274564. doi: 10.4061/2011/274564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Sardina JL, Lopez-Ruano G, Sanchez-Sanchez B, Llanillo M, Hernandez-Hernandez A. Reactive oxygen species: Are they important for haematopoiesis? Crit Rev Oncol Hematol. 2012;81:257–274. doi: 10.1016/j.critrevonc.2011.03.005. [DOI] [PubMed] [Google Scholar]
  • [15].Eliasson P, Rehn M, Hammar P, Larsson P, Sirenko O, Flippin LA, Cammenga J, Jonsson JI. Hypoxia mediates low cell-cycle activity and increases the proportion of long-term-reconstituting hematopoietic stem cells during in vitro culture. Exp Hematol. 2010;38:301–310. e302. doi: 10.1016/j.exphem.2010.01.005. [DOI] [PubMed] [Google Scholar]
  • [16].Case J, Ingram DA, Haneline LS. Oxidative stress impairs endothelial progenitor cell function. Antioxid Redox Signal. 2008;10:1895–1907. doi: 10.1089/ars.2008.2118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Ito K, Hirao A, Arai F, Matsuoka S, Takubo K, Hamaguchi I, Nomiyama K, Hosokawa K, Sakurada K, Nakagata N, Ikeda Y, Mak TW, Suda T. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature. 2004;431:997–1002. doi: 10.1038/nature02989. [DOI] [PubMed] [Google Scholar]
  • [18].Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, Miyamoto K, Ohmura M, Naka K, Hosokawa K, Ikeda Y, Suda T. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med. 2006;12:446–451. doi: 10.1038/nm1388. [DOI] [PubMed] [Google Scholar]
  • [19].Kobayashi CI, Suda T. Regulation of reactive oxygen species in stem cells and cancer stem cells. J Cell Physiol. doi: 10.1002/jcp.22764. [DOI] [PubMed] [Google Scholar]
  • [20].Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH, Cullen DE, McDowell EP, Lazo-Kallanian S, Williams IR, Sears C, Armstrong SA, Passegue E, DePinho RA, Gilliland DG. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007;128:325–339. doi: 10.1016/j.cell.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • [21].Jang YY, Sharkis SJ. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood. 2007;110:3056–3063. doi: 10.1182/blood-2007-05-087759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Naka K, Muraguchi T, Hoshii T, Hirao A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxid Redox Signal. 2008;10:1883–1894. doi: 10.1089/ars.2008.2114. [DOI] [PubMed] [Google Scholar]
  • [23].Urao N, McKinney RD, Fukai T, Ushio-Fukai M. NADPH oxidase 2 regulates bone marrow microenvironment following hindlimb ischemia: role in reparative mobilization of progenitor cells. Stem Cells. 2012;30:923–934. doi: 10.1002/stem.1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Finkel T. Signal transduction by reactive oxygen species. J Cell Biol. 2011;194:7–15. doi: 10.1083/jcb.201102095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res. 2000;86:494–501. doi: 10.1161/01.res.86.5.494. [DOI] [PubMed] [Google Scholar]
  • [26].Rhee SG, Bae YS, Lee SR, Kwon J. Hydrogen peroxide: a key messenger that modulates protein phosphorylation through cysteine oxidation. Sci STKE. 2000;2000:PE1. doi: 10.1126/stke.2000.53.pe1. [DOI] [PubMed] [Google Scholar]
  • [27].Ushio-Fukai M. Redox signaling in angiogenesis: role of NADPH oxidase. Cardiovasc Res. 2006;71:226–235. doi: 10.1016/j.cardiores.2006.04.015. [DOI] [PubMed] [Google Scholar]
  • [28].Nathan C, Ding A. SnapShot: Reactive Oxygen Intermediates (ROI) Cell. 2010;140:951–951. e952. doi: 10.1016/j.cell.2010.03.008. [DOI] [PubMed] [Google Scholar]
  • [29].Fukai T, Ushio-Fukai M. Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid Redox Signal. 2011;15:1583–1606. doi: 10.1089/ars.2011.3999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Pervaiz S, Taneja R, Ghaffari S. Oxidative stress regulation of stem and progenitor cells. Antioxid Redox Signal. 2009;11:2777–2789. doi: 10.1089/ars.2009.2804. [DOI] [PubMed] [Google Scholar]
  • [31].Michiels C, Raes M, Toussaint O, Remacle J. Importance of Se-glutathione peroxidase, catalase, and Cu/Zn-SOD for cell survival against oxidative stress. Free Radic Biol Med. 1994;17:235–248. doi: 10.1016/0891-5849(94)90079-5. [DOI] [PubMed] [Google Scholar]
  • [32].Cohen G, Hochstein P. Glutathione Peroxidase: the Primary Agent for the Elimination of Hydrogen Peroxide in Erythrocytes. Biochemistry. 1963;2:1420–1428. doi: 10.1021/bi00906a038. [DOI] [PubMed] [Google Scholar]
  • [33].Dernbach E, Urbich C, Brandes RP, Hofmann WK, Zeiher AM, Dimmeler S. Antioxidative stress-associated genes in circulating progenitor cells: evidence for enhanced resistance against oxidative stress. Blood. 2004;104:3591–3597. doi: 10.1182/blood-2003-12-4103. [DOI] [PubMed] [Google Scholar]
  • [34].Lassegue B, SanMartin A, Griendling KK. Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system. Circ Res. 2012;110:1364–1390. doi: 10.1161/CIRCRESAHA.111.243972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Ushio-Fukai M, Urao N. Novel role of NADPH oxidase in angiogenesis and stem/progenitor cell function. Antioxid Redox Signal. 2009;11:2517–2533. doi: 10.1089/ars.2009.2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Ushio-Fukai M. Compartmentalization of redox signaling through NADPH oxidase-derived ROS. Antioxid Redox Signal. 2009;11:1289–1299. doi: 10.1089/ars.2008.2333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Jiang F, Zhang Y, Dusting GJ. NADPH oxidase-mediated redox signaling: roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol Rev. 2011;63:218–242. doi: 10.1124/pr.110.002980. [DOI] [PubMed] [Google Scholar]
  • [38].Diaz B, Courtneidge SA. Redox signaling at invasive microdomains in cancer cells. Free Radic Biol Med. 2012;52:247–256. doi: 10.1016/j.freeradbiomed.2011.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Coso S, Harrison I, Harrison CB, Vinh A, Sobey CG, Drummond GR, Williams ED, Selemidis S. NADPH oxidases as regulators of tumor angiogenesis: current and emerging concepts. Antioxid Redox Signal. 2012;16:1229–1247. doi: 10.1089/ars.2011.4489. [DOI] [PubMed] [Google Scholar]
  • [40].Sareila O, Kelkka T, Pizzolla A, Hultqvist M, Holmdahl R. NOX2 complex-derived ROS as immune regulators. Antioxid Redox Signal. 2011;15:2197–2208. doi: 10.1089/ars.2010.3635. [DOI] [PubMed] [Google Scholar]
  • [41].Lam GY, Huang J, Brumell JH. The many roles of NOX2 NADPH oxidase-derived ROS in immunity. Semin Immunopathol. 2010;32:415–430. doi: 10.1007/s00281-010-0221-0. [DOI] [PubMed] [Google Scholar]
  • [42].Fiorentini D, Prata C, Maraldi T, Zambonin L, Bonsi L, Hakim G, Landi L. Contribution of reactive oxygen species to the regulation of Glut1 in two hemopoietic cell lines differing in cytokine sensitivity. Free Radic Biol Med. 2004;37:1402–1411. doi: 10.1016/j.freeradbiomed.2004.07.022. [DOI] [PubMed] [Google Scholar]
  • [43].Urao N, Inomata H, Razvi M, Kim HW, Wary K, McKinney R, Fukai T, Ushio-Fukai M. Role of nox2-based NADPH oxidase in bone marrow and progenitor cell function involved in neovascularization induced by hindlimb ischemia. Circ Res. 2008;103:212–220. doi: 10.1161/CIRCRESAHA.108.176230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Finkel T. Signal transduction by mitochondrial oxidants. J Biol Chem. 2011;287:4434–4440. doi: 10.1074/jbc.R111.271999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Zhang M, Brewer AC, Schroder K, Santos CX, Grieve DJ, Wang M, Anilkumar N, Yu B, Dong X, Walker SJ, Brandes RP, Shah AM. NADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesis. Proc Natl Acad Sci U S A. 2010;107:18121–18126. doi: 10.1073/pnas.1009700107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Fadini GP, Sartore S, Albiero M, Baesso I, Murphy E, Menegolo M, Grego F, Vigili de Kreutzenberg S, Tiengo A, Agostini C, Avogaro A. Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy. Arterioscler Thromb Vasc Biol. 2006;26:2140–2146. doi: 10.1161/01.ATV.0000237750.44469.88. [DOI] [PubMed] [Google Scholar]
  • [47].St-Pierre J, Buckingham JA, Roebuck SJ, Brand MD. Topology of superoxide production from different sites in the mitochondrial electron transport chain. J Biol Chem. 2002;277:44784–44790. doi: 10.1074/jbc.M207217200. [DOI] [PubMed] [Google Scholar]
  • [48].Daiber A. Redox signaling (cross-talk) from and to mitochondria involves mitochondrial pores and reactive oxygen species. Biochim Biophys Acta. 2010;1797:897–906. doi: 10.1016/j.bbabio.2010.01.032. [DOI] [PubMed] [Google Scholar]
  • [49].Dikalov S. Cross talk between mitochondria and NADPH oxidases. Free Radic Biol Med. 2011;51:1289–1301. doi: 10.1016/j.freeradbiomed.2011.06.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Rathore R, Zheng YM, Niu CF, Liu QH, Korde A, Ho YS, Wang YX. Hypoxia activates NADPH oxidase to increase [ROS]i and [Ca2+]i through the mitochondrial ROS-PKCepsilon signaling axis in pulmonary artery smooth muscle cells. Free Radic Biol Med. 2008;45:1223–1231. doi: 10.1016/j.freeradbiomed.2008.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Lee SB, Bae IH, Bae YS, Um HD. Link between mitochondria and NADPH oxidase 1 isozyme for the sustained production of reactive oxygen species and cell death. J Biol Chem. 2006;281:36228–36235. doi: 10.1074/jbc.M606702200. [DOI] [PubMed] [Google Scholar]
  • [52].Miller TW, Isenberg JS, Roberts DD. Molecular regulation of tumor angiogenesis and perfusion via redox signaling. Chem Rev. 2009;109:3099–3124. doi: 10.1021/cr8005125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Parmar K, Mauch P, Vergilio JA, Sackstein R, Down JD. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci U S A. 2007;104:5431–5436. doi: 10.1073/pnas.0701152104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Suda T, Takubo K, Semenza GL. Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell. 2011;9:298–310. doi: 10.1016/j.stem.2011.09.010. [DOI] [PubMed] [Google Scholar]
  • [55].Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 2009;461:537–541. doi: 10.1038/nature08313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Sattler M, Winkler T, Verma S, Byrne CH, Shrikhande G, Salgia R, Griffin JD. Hematopoietic growth factors signal through the formation of reactive oxygen species. Blood. 1999;93:2928–2935. [PubMed] [Google Scholar]
  • [57].Tesio M, Golan K, Corso S, Giordano S, Schajnovitz A, Vagima Y, Shivtiel S, Kalinkovich A, Caione L, Gammaitoni L, Laurenti E, Buss EC, Shezen E, Itkin T, Kollet O, Petit I, Trumpp A, Christensen J, Aglietta M, Piacibello W, Lapidot T. Enhanced c-Met activity promotes G-CSF-induced mobilization of hematopoietic progenitor cells via ROS signaling. Blood. 2011;117:419–428. doi: 10.1182/blood-2009-06-230359. [DOI] [PubMed] [Google Scholar]
  • [58].Milovanova TN, Bhopale VM, Sorokina EM, Moore JS, Hunt TK, Hauer-Jensen M, Velazquez OC, Thom SR. Hyperbaric oxygen stimulates vasculogenic stem cell growth and differentiation in vivo. J Appl Physiol. 2009;106:711–728. doi: 10.1152/japplphysiol.91054.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Wang Y, Kellner J, Liu L, Zhou D. Inhibition of p38 mitogen-activated protein kinase promotes ex vivo hematopoietic stem cell expansion. Stem Cells Dev. 2011;20:1143–1152. doi: 10.1089/scd.2010.0413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Yun J, Rocic P, Pung YF, Belmadani S, Carrao AC, Ohanyan V, Chilian WM. Redox-dependent mechanisms in coronary collateral growth: the “redox window” hypothesis. Antioxid Redox Signal. 2009;11:1961–1974. doi: 10.1089/ars.2009.2476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Liu J, Cao L, Chen J, Song S, Lee IH, Quijano C, Liu H, Keyvanfar K, Chen H, Cao LY, Ahn BH, Kumar NG, Rovira II, Xu XL, van Lohuizen M, Motoyama N, Deng CX, Finkel T. Bmi1 regulates mitochondrial function and the DNA damage response pathway. Nature. 2009;459:387–392. doi: 10.1038/nature08040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Chen C, Liu Y, Liu R, Ikenoue T, Guan KL, Liu Y, Zheng P. TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J Exp Med. 2008;205:2397–2408. doi: 10.1084/jem.20081297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Terskikh AV, Miyamoto T, Chang C, Diatchenko L, Weissman IL. Gene expression analysis of purified hematopoietic stem cells and committed progenitors. Blood. 2003;102:94–101. doi: 10.1182/blood-2002-08-2509. [DOI] [PubMed] [Google Scholar]
  • [64].Piccoli C, Ria R, Scrima R, Cela O, D’Aprile A, Boffoli D, Falzetti F, Tabilio A, Capitanio N. Characterization of mitochondrial and extra-mitochondrial oxygen consuming reactions in human hematopoietic stem cells. Novel evidence of the occurrence of NAD(P)H oxidase activity. J Biol Chem. 2005;280:26467–26476. doi: 10.1074/jbc.M500047200. [DOI] [PubMed] [Google Scholar]
  • [65].Piccoli C, D’Aprile A, Ripoli M, Scrima R, Lecce L, Boffoli D, Tabilio A, Capitanio N. Bone-marrow derived hematopoietic stem/progenitor cells express multiple isoforms of NADPH oxidase and produce constitutively reactive oxygen species. Biochem Biophys Res Commun. 2007;353:965–972. doi: 10.1016/j.bbrc.2006.12.148. [DOI] [PubMed] [Google Scholar]
  • [66].Frassanito MC, Piccoli C, Capozzi V, Boffoli D, Tabilio A, Capitanio N. Topological organization of NADPH-oxidase in haematopoietic stem cell membrane: preliminary study by fluorescence near-field optical microscopy. J Microsc. 2008;229:517–524. doi: 10.1111/j.1365-2818.2008.01937.x. [DOI] [PubMed] [Google Scholar]
  • [67].Schroder K, Kohnen A, Aicher A, Liehn EA, Buchse T, Stein S, Weber C, Dimmeler S, Brandes RP. NADPH oxidase Nox2 is required for hypoxia-induced mobilization of endothelial progenitor cells. Circ Res. 2009;105:537–544. doi: 10.1161/CIRCRESAHA.109.205138. [DOI] [PubMed] [Google Scholar]
  • [68].Schroder K, Schutz S, Schloffel I, Batz S, Takac I, Weissmann N, Michaelis UR, Koyanagi M, Brandes RP. Hepatocyte growth factor induces a proangiogenic phenotype and mobilizes endothelial progenitor cells by activating Nox2. Antioxid Redox Signal. 2011;15:915–923. doi: 10.1089/ars.2010.3533. [DOI] [PubMed] [Google Scholar]
  • [69].Sauer H, Rahimi G, Hescheler J, Wartenberg M. Role of reactive oxygen species and phosphatidylinositol 3-kinase in cardiomyocyte differentiation of embryonic stem cells. FEBS Lett. 2000;476:218–223. doi: 10.1016/s0014-5793(00)01747-6. [DOI] [PubMed] [Google Scholar]
  • [70].Sauer H, Bekhite MM, Hescheler J, Wartenberg M. Redox control of angiogenic factors and CD31-positive vessel-like structures in mouse embryonic stem cells after direct current electrical field stimulation. Exp Cell Res. 2005;304:380–390. doi: 10.1016/j.yexcr.2004.11.026. [DOI] [PubMed] [Google Scholar]
  • [71].Schmelter M, Ateghang B, Helmig S, Wartenberg M, Sauer H. Embryonic stem cells utilize reactive oxygen species as transducers of mechanical strain-induced cardiovascular differentiation. Faseb J. 2006;20:1182–1184. doi: 10.1096/fj.05-4723fje. [DOI] [PubMed] [Google Scholar]
  • [72].Xiao Q, Luo Z, Pepe AE, Margariti A, Zeng L, Xu Q. Embryonic stem cell differentiation into smooth muscle cells is mediated by Nox4-produced H2O2. Am J Physiol Cell Physiol. 2009;296:C711–723. doi: 10.1152/ajpcell.00442.2008. [DOI] [PubMed] [Google Scholar]
  • [73].Saretzki G, Armstrong L, Leake A, Lako M, von Zglinicki T. Stress defense in murine embryonic stem cells is superior to that of various differentiated murine cells. Stem Cells. 2004;22:962–971. doi: 10.1634/stemcells.22-6-962. [DOI] [PubMed] [Google Scholar]
  • [74].Buggisch M, Ateghang B, Ruhe C, Strobel C, Lange S, Wartenberg M, Sauer H. Stimulation of ES-cell-derived cardiomyogenesis and neonatal cardiac cell proliferation by reactive oxygen species and NADPH oxidase. J Cell Sci. 2007;120:885–894. doi: 10.1242/jcs.03386. [DOI] [PubMed] [Google Scholar]
  • [75].Sauer H, Rahimi G, Hescheler J, Wartenberg M. Effects of electrical fields on cardiomyocyte differentiation of embryonic stem cells. J Cell Biochem. 1999;75:710–723. doi: 10.1002/(sici)1097-4644(19991215)75:4<710::aid-jcb16>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • [76].Serena E, Figallo E, Tandon N, Cannizzaro C, Gerecht S, Elvassore N, Vunjak-Novakovic G. Electrical stimulation of human embryonic stem cells: cardiac differentiation and the generation of reactive oxygen species. Exp Cell Res. 2009;315:3611–3619. doi: 10.1016/j.yexcr.2009.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Sauer H, Neukirchen W, Rahimi G, Grunheck F, Hescheler J, Wartenberg M. Involvement of reactive oxygen species in cardiotrophin-1-induced proliferation of cardiomyocytes differentiated from murine embryonic stem cells. Exp Cell Res. 2004;294:313–324. doi: 10.1016/j.yexcr.2003.10.032. [DOI] [PubMed] [Google Scholar]
  • [78].Lange S, Heger J, Euler G, Wartenberg M, Piper HM, Sauer H. Platelet-derived growth factor BB stimulates vasculogenesis of embryonic stem cell-derived endothelial cells by calcium-mediated generation of reactive oxygen species. Cardiovasc Res. 2008 doi: 10.1093/cvr/cvn258. [DOI] [PubMed] [Google Scholar]
  • [79].Sharifpanah F, Wartenberg M, Hannig M, Piper HM, Sauer H. Peroxisome proliferator-activated receptor alpha agonists enhance cardiomyogenesis of mouse ES cells by utilization of a reactive oxygen species-dependent mechanism. Stem Cells. 2008;26:64–71. doi: 10.1634/stemcells.2007-0532. [DOI] [PubMed] [Google Scholar]
  • [80].Li J, Stouffs M, Serrander L, Banfi B, Bettiol E, Charnay Y, Steger K, Krause KH, Jaconi ME. The NADPH oxidase NOX4 drives cardiac differentiation: Role in regulating cardiac transcription factors and MAP kinase activation. Mol Biol Cell. 2006;17:3978–3988. doi: 10.1091/mbc.E05-06-0532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Hannig M, Figulla HR, Sauer H, Wartenberg M. Control of leucocyte differentiation from embryonic stem cells upon vasculogenesis and confrontation with tumour tissue. J Cell Mol Med. 2010;14:303–312. doi: 10.1111/j.1582-4934.2008.00424.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Storz P. Forkhead homeobox type O transcription factors in the responses to oxidative stress. Antioxid Redox Signal. 2011;14:593–605. doi: 10.1089/ars.2010.3405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Miyamoto K, Araki KY, Naka K, Arai F, Takubo K, Yamazaki S, Matsuoka S, Miyamoto T, Ito K, Ohmura M, Chen C, Hosokawa K, Nakauchi H, Nakayama K, Nakayama KI, Harada M, Motoyama N, Suda T, Hirao A. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell. 2007;1:101–112. doi: 10.1016/j.stem.2007.02.001. [DOI] [PubMed] [Google Scholar]
  • [84].Miyamoto K, Miyamoto T, Kato R, Yoshimura A, Motoyama N, Suda T. FoxO3a regulates hematopoietic homeostasis through a negative feedback pathway in conditions of stress or aging. Blood. 2008;112:4485–4493. doi: 10.1182/blood-2008-05-159848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Juntilla MM, Patil VD, Calamito M, Joshi RP, Birnbaum MJ, Koretzky GA. AKT1 and AKT2 maintain hematopoietic stem cell function by regulating reactive oxygen species. Blood. 2010;115:4030–4038. doi: 10.1182/blood-2009-09-241000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Kharas MG, Okabe R, Ganis JJ, Gozo M, Khandan T, Paktinat M, Gilliland DG, Gritsman K. Constitutively active AKT depletes hematopoietic stem cells and induces leukemia in mice. Blood. 2010;115:1406–1415. doi: 10.1182/blood-2009-06-229443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].LeBelle JE, Orozco NM, Paucar AA, Saxe JP, Mottahedeh J, Pyle AD, Wu H, Kornblum HI. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner. Cell Stem Cell. 2011;8:59–71. doi: 10.1016/j.stem.2010.11.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Cho SH, Lee CH, Ahn Y, Kim H, Kim H, Ahn CY, Yang KS, Lee SR. Redox regulation of PTEN and protein tyrosine phosphatases in H(2)O(2) mediated cell signaling. FEBS Lett. 2004;560:7–13. doi: 10.1016/s0014-5793(04)00112-7. [DOI] [PubMed] [Google Scholar]
  • [89].Salmeen A, Barford D. Functions and mechanisms of redox regulation of cysteine-based phosphatases. Antioxid Redox Signal. 2005;7:560–577. doi: 10.1089/ars.2005.7.560. [DOI] [PubMed] [Google Scholar]
  • [90].Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, Wu H, Morrison SJ. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature. 2006;441:475–482. doi: 10.1038/nature04703. [DOI] [PubMed] [Google Scholar]
  • [91].Lee JY, Nakada D, Yilmaz OH, Tothova Z, Joseph NM, Lim MS, Gilliland DG, Morrison SJ. mTOR activation induces tumor suppressors that inhibit leukemogenesis and deplete hematopoietic stem cells after Pten deletion. Cell Stem Cell. 2010;7:593–605. doi: 10.1016/j.stem.2010.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Li TS, Marban E. Physiological levels of reactive oxygen species are required to maintain genomic stability in stem cells. Stem Cells. 2010;28:1178–1185. doi: 10.1002/stem.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Rizo A, Olthof S, Han L, Vellenga E, de Haan G, Schuringa JJ. Repression of BMI1 in normal and leukemic human CD34(+) cells impairs self-renewal and induces apoptosis. Blood. 2009;114:1498–1505. doi: 10.1182/blood-2009-03-209734. [DOI] [PubMed] [Google Scholar]
  • [94].Facchino S, Abdouh M, Chatoo W, Bernier G. BMI1 confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery. J Neurosci. 2010;30:10096–10111. doi: 10.1523/JNEUROSCI.1634-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Chuikov S, Levi BP, Smith ML, Morrison SJ. Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress. Nat Cell Biol. 2010;12:999–1006. doi: 10.1038/ncb2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Maryanovich M, Oberkovitz G, Niv H, Vorobiyov L, Zaltsman Y, Brenner O, Lapidot T, Jung S, Gross A. The ATM-BID pathway regulates quiescence and survival of haematopoietic stem cells. Nat Cell Biol. 2012;14:535–541. doi: 10.1038/ncb2468. [DOI] [PubMed] [Google Scholar]
  • [97].Abbas HA, Maccio DR, Coskun S, Jackson JG, Hazen AL, Sills TM, You MJ, Hirschi KK, Lozano G. Mdm2 is required for survival of hematopoietic stem cells/progenitors via dampening of ROS-induced p53 activity. Cell Stem Cell. 2010;7:606–617. doi: 10.1016/j.stem.2010.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Abbas HA, Pant V, Lozano G. The ups and downs of p53 regulation in hematopoietic stem cells. Cell Cycle. 2011;10:3257–3262. doi: 10.4161/cc.10.19.17721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Wilson A, Trumpp A. Bone-marrow haematopoietic-stem-cell niches. Nat Rev Immunol. 2006;6:93–106. doi: 10.1038/nri1779. [DOI] [PubMed] [Google Scholar]
  • [100].Yin T, Li L. The stem cell niches in bone. J Clin Invest. 2006;116:1195–1201. doi: 10.1172/JCI28568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Mansour A, Abou-Ezzi G, Sitnicka E, Jacobsen SE, Wakkach A, Blin-Wakkach C. Osteoclasts promote the formation of hematopoietic stem cell niches in the bone marrow. J Exp Med. 2012;209:537–549. doi: 10.1084/jem.20110994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, Scadden DT, Ma’ayan A, Enikolopov GN, Frenette PS. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829–834. doi: 10.1038/nature09262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, Poulton IJ, van Rooijen N, Alexander KA, Raggatt LJ, Levesque JP. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815–4828. doi: 10.1182/blood-2009-11-253534. [DOI] [PubMed] [Google Scholar]
  • [104].Chow A, Lucas D, Hidalgo A, Mendez-Ferrer S, Hashimoto D, Scheiermann C, Battista M, Leboeuf M, Prophete C, van Rooijen N, Tanaka M, Merad M, Frenette PS. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011;208:261–271. doi: 10.1084/jem.20101688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Fujisaki J, Wu J, Carlson AL, Silberstein L, Putheti P, Larocca R, Gao W, Saito TI, Lo Celso C, Tsuyuzaki H, Sato T, Cote D, Sykes M, Strom TB, Scadden DT, Lin CP. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature. 2011;474:216–219. doi: 10.1038/nature10160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Doan PL, Chute JP. The vascular niche: home for normal and malignant hematopoietic stem cells. Leukemia. 2012;26:54–62. doi: 10.1038/leu.2011.236. [DOI] [PubMed] [Google Scholar]
  • [107].Guerrouahen BS, Al-Hijji I, Tabrizi AR. Osteoblastic and vascular endothelial niches, their control on normal hematopoietic stem cells, and their consequences on the development of leukemia. Stem Cells Int. 2011;2011:375857. doi: 10.4061/2011/375857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Hoggatt J, Pelus LM. Many mechanisms mediating mobilization: an alliterative review. Curr Opin Hematol. 2011;18:231–238. doi: 10.1097/MOH.0b013e3283477962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Hoggatt J, Pelus LM. Mobilization of hematopoietic stem cells from the bone marrow niche to the blood compartment. Stem Cell Res Ther. 2011;2:13. doi: 10.1186/scrt54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Lam BS, Adams GB. Hematopoietic stem cell lodgment in the adult bone marrow stem cell niche. Int J Lab Hematol. 2010;32:551–558. doi: 10.1111/j.1751-553X.2010.01250.x. [DOI] [PubMed] [Google Scholar]
  • [111].Suarez-Alvarez B, Lopez-Vazquez A, Lopez-Larrea C. Mobilization and homing of hematopoietic stem cells. Adv Exp Med Biol. 2012;741:152–170. doi: 10.1007/978-1-4614-2098-9_11. [DOI] [PubMed] [Google Scholar]
  • [112].Ren B, Deng Y, Mukhopadhyay A, Lanahan AA, Zhuang ZW, Moodie KL, Mulligan-Kehoe MJ, Byzova TV, Peterson RT, Simons M. ERK1/2-Akt1 crosstalk regulates arteriogenesis in mice and zebrafish. J Clin Invest. 2010;120:1217–1228. doi: 10.1172/JCI39837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Cipolleschi MG, DelloSbarba P, Olivotto M. The role of hypoxia in the maintenance of hematopoietic stem cells. Blood. 1993;82:2031–2037. [PubMed] [Google Scholar]
  • [114].Danet GH, Pan Y, Luongo JL, Bonnet DA, Simon MC. Expansion of human SCID-repopulating cells under hypoxic conditions. J Clin Invest. 2003;112:126–135. doi: 10.1172/JCI17669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Ivanovic Z, Bartolozzi B, Bernabei PA, Cipolleschi MG, Rovida E, Milenkovic P, Praloran V, DelloSbarba P. Incubation of murine bone marrow cells in hypoxia ensures the maintenance of marrow-repopulating ability together with the expansion of committed progenitors. Br J Haematol. 2000;108:424–429. doi: 10.1046/j.1365-2141.2000.01842.x. [DOI] [PubMed] [Google Scholar]
  • [116].Winkler IG, Barbier V, Wadley R, Zannettino AC, Williams S, Levesque JP. Positioning of bone marrow hematopoietic and stromal cells relative to blood flow in vivo: serially reconstituting hematopoietic stem cells reside in distinct nonperfused niches. Blood. 2010;116:375–385. doi: 10.1182/blood-2009-07-233437. [DOI] [PubMed] [Google Scholar]
  • [117].Takubo K, Goda N, Yamada W, Iriuchishima H, Ikeda E, Kubota Y, Shima H, Johnson RS, Hirao A, Suematsu M, Suda T. Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. Cell Stem Cell. 2010;7:391–402. doi: 10.1016/j.stem.2010.06.020. [DOI] [PubMed] [Google Scholar]
  • [118].Bromberg O, Frisch BJ, Weber JM, Porter RL, Civitelli R, Calvi LM. Osteoblastic N-cadherin is not required for microenvironmental support and regulation of hematopoietic stem and progenitor cells. Blood. 2012;120:303–313. doi: 10.1182/blood-2011-09-377853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Greenbaum AM, Revollo LD, Woloszynek JR, Civitelli R, Link DC. N-cadherin in osteolineage cells is not required for maintenance of hematopoietic stem cells. Blood. 2012;120:295–302. doi: 10.1182/blood-2011-09-377457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Kiel MJ, Acar M, Radice GL, Morrison SJ. Hematopoietic stem cells do not depend on N-cadherin to regulate their maintenance. Cell Stem Cell. 2009;4:170–179. doi: 10.1016/j.stem.2008.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Lewandowski D, Barroca V, Duconge F, Bayer J, VanNhieu JT, Pestourie C, Fouchet P, Tavitian B, Romeo PH. In vivo cellular imaging pinpoints the role of reactive oxygen species in the early steps of adult hematopoietic reconstitution. Blood. 2010;115:443–452. doi: 10.1182/blood-2009-05-222711. [DOI] [PubMed] [Google Scholar]
  • [122].Kobayashi H, Butler JM, O’Donnell R, Kobayashi M, Ding BS, Bonner B, Chiu VK, Nolan DJ, Shido K, Benjamin L, Rafii S. Angiocrine factors from Akt-activated endothelial cells balance self-renewal and differentiation of haematopoietic stem cells. Nat Cell Biol. 2010;12:1046–1056. doi: 10.1038/ncb2108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].TaniguchiIshikawa E, Gonzalez-Nieto D, Ghiaur G, Dunn SK, Ficker AM, Murali B, Madhu M, Gutstein DE, Fishman GI, Barrio LC, Cancelas JA. Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells. Proc Natl Acad Sci U S A. 2012;109:9071–9076. doi: 10.1073/pnas.1120358109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Aicher A, Zeiher AM, Dimmeler S. Mobilizing endothelial progenitor cells. Hypertension. 2005;45:321–325. doi: 10.1161/01.HYP.0000154789.28695.ea. [DOI] [PubMed] [Google Scholar]
  • [125].Dimmeler S. Regulation of bone marrow-derived vascular progenitor cell mobilization and maintenance. Arterioscler Thromb Vasc Biol. 2010;30:1088–1093. doi: 10.1161/ATVBAHA.109.191668. [DOI] [PubMed] [Google Scholar]
  • [126].Krankel N, Spinetti G, Amadesi S, Madeddu P. Targeting stem cell niches and trafficking for cardiovascular therapy. Pharmacol Ther. 2011;129:62–81. doi: 10.1016/j.pharmthera.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Levesque JP, Winkler IG, Hendy J, Williams B, Helwani F, Barbier V, Nowlan B, Nilsson SK. Hematopoietic progenitor cell mobilization results in hypoxia with increased hypoxia-inducible transcription factor-1 alpha and vascular endothelial growth factor A in bone marrow. Stem Cells. 2007;25:1954–1965. doi: 10.1634/stemcells.2006-0688. [DOI] [PubMed] [Google Scholar]
  • [128].van Os R, Robinson SN, Drukteinis D, Sheridan TM, Mauch PM. Respiratory burst of neutrophils is not required for stem cell mobilization in mice. Br J Haematol. 2000;111:695–699. doi: 10.1046/j.1365-2141.2000.02374.x. [DOI] [PubMed] [Google Scholar]
  • [129].Fan J, Cai H, Tan WS. Role of the plasma membrane ROS-generating NADPH oxidase in CD34+ progenitor cells preservation by hypoxia. J Biotechnol. 2007;130:455–462. doi: 10.1016/j.jbiotec.2007.05.023. [DOI] [PubMed] [Google Scholar]
  • [130].Edlund J, Fasching A, Liss P, Hansell P, Palm F. The roles of NADPH-oxidase and nNOS for the increased oxidative stress and the oxygen consumption in the diabetic kidney. Diabetes Metab Res Rev. 2010;26:349–356. doi: 10.1002/dmrr.1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Simsek T, Kocabas F, Zheng J, Deberardinis RJ, Mahmoud AI, Olson EN, Schneider JW, Zhang CC, Sadek HA. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell. 2010;7:380–390. doi: 10.1016/j.stem.2010.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Lee SJ, Hwang AB, Kenyon C. Inhibition of respiration extends C. elegans life span via reactive oxygen species that increase HIF-1 activity. Curr Biol. 2010;20:2131–2136. doi: 10.1016/j.cub.2010.10.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Jiang M, Wang B, Wang C, He B, Fan H, Guo TB, Shao Q, Gao L, Liu Y. Angiogenesis by transplantation of HIF-1 alpha modified EPCs into ischemic limbs. J Cell Biochem. 2008;103:321–334. doi: 10.1002/jcb.21416. [DOI] [PubMed] [Google Scholar]
  • [134].Rey S, Lee K, Wang CJ, Gupta K, Chen S, McMillan A, Bhise N, Levchenko A, Semenza GL. Synergistic effect of HIF-1alpha gene therapy and HIF-1-activated bone marrow-derived angiogenic cells in a mouse model of limb ischemia. Proc Natl Acad Sci U S A. 2009;106:20399–20404. doi: 10.1073/pnas.0911921106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Kucia M, Dawn B, Hunt G, Guo Y, Wysoczynski M, Majka M, Ratajczak J, Rezzoug F, Ildstad ST, Bolli R, Ratajczak MZ. Cells expressing early cardiac markers reside in the bone marrow and are mobilized into the peripheral blood after myocardial infarction. Circ Res. 2004;95:1191–1199. doi: 10.1161/01.RES.0000150856.47324.5b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Schober A. Chemokines in vascular dysfunction and remodeling. Arterioscler Thromb Vasc Biol. 2008;28:1950–1959. doi: 10.1161/ATVBAHA.107.161224. [DOI] [PubMed] [Google Scholar]
  • [137].Pillarisetti K, Gupta SK. Cloning and relative expression analysis of rat stromal cell derived factor-1 (SDF-1)1: SDF-1 alpha mRNA is selectively induced in rat model of myocardial infarction. Inflammation. 2001;25:293–300. doi: 10.1023/a:1012808525370. [DOI] [PubMed] [Google Scholar]
  • [138].Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, Haase VH, Jaenisch R, Corr M, Nizet V, Firestein GS, Gerber HP, Ferrara N, Johnson RS. HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell. 2003;112:645–657. doi: 10.1016/s0092-8674(03)00154-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864. doi: 10.1038/nm1075. [DOI] [PubMed] [Google Scholar]
  • [140].Golan K, Vagima Y, Ludin A, Itkin T, Cohen-Gur S, Kalinkovich A, Kollet O, Kim C, Schajnovitz A, Ovadya Y, Lapid K, Shivtiel S, Morris AJ, Ratajczak MZ, Lapidot T. S1P promotes murine progenitor cell egress and mobilization via S1P1-mediated ROS signaling and SDF-1 release. Blood. 2012;119:2478–2488. doi: 10.1182/blood-2011-06-358614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol. 2007;8:221–233. doi: 10.1038/nrm2125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Masuda H, Alev C, Akimaru H, Ito R, Shizuno T, Kobori M, Horii M, Ishihara T, Isobe K, Isozaki M, Itoh J, Itoh Y, Okada Y, McIntyre BA, Kato S, Asahara T. Methodological development of a clonogenic assay to determine endothelial progenitor cell potential. Circ Res. 2011;109:20–37. doi: 10.1161/CIRCRESAHA.110.231837. [DOI] [PubMed] [Google Scholar]
  • [143].Wang W, Gou L, Xie G, Tong A, He F, Lu Z, Yao Y, Liu K, Li J, Tang M, Chen L, Yang J, Hu H, Wei YQ. Proteomic analysis of interstitial fluid in bone marrow identified that peroxiredoxin 2 regulates H(2)O(2) level of bone marrow during aging. J Proteome Res. 2010;9:3812–3819. doi: 10.1021/pr901180w. [DOI] [PubMed] [Google Scholar]
  • [144].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 (Lond) 2004;107:273–280. doi: 10.1042/CS20030389. [DOI] [PubMed] [Google Scholar]
  • [145].Fadini GP, Miorin M, Facco M, Bonamico S, Baesso I, Grego F, Menegolo M, de Kreutzenberg SV, Tiengo A, Agostini C, Avogaro A. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 2005;45:1449–1457. doi: 10.1016/j.jacc.2004.11.067. [DOI] [PubMed] [Google Scholar]
  • [146].Fadini GP, de Kreutzenberg S, Albiero M, Coracina A, Pagnin E, Baesso I, Cignarella A, Bolego C, Plebani M, Nardelli GB, Sartore S, Agostini C, Avogaro A. Gender differences in endothelial progenitor cells and cardiovascular risk profile: the role of female estrogens. Arterioscler Thromb Vasc Biol. 2008;28:997–1004. doi: 10.1161/ATVBAHA.107.159558. [DOI] [PubMed] [Google Scholar]
  • [147].Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, Finkel T. 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]
  • [148].Imanishi T, Hano T, Nishio I. Angiotensin II accelerates endothelial progenitor cell senescence through induction of oxidative stress. J Hypertens. 2005;23:97–104. doi: 10.1097/00004872-200501000-00018. [DOI] [PubMed] [Google Scholar]
  • [149].Loomans CJ, de Koning EJ, Staal FJ, Rookmaaker MB, Verseyden C, de Boer HC, Verhaar MC, Braam B, Rabelink TJ, van Zonneveld AJ. Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes. 2004;53:195–199. doi: 10.2337/diabetes.53.1.195. [DOI] [PubMed] [Google Scholar]
  • [150].Oliveras A, Soler MJ, Martinez-Estrada OM, Vazquez S, Marco-Feliu D, Vila JS, Vilaro S, Lloveras J. Endothelial progenitor cells are reduced in refractory hypertension. J Hum Hypertens. 2008;22:183–190. doi: 10.1038/sj.jhh.1002304. [DOI] [PubMed] [Google Scholar]
  • [151].Urbich C, Dimmeler S. Risk factors for coronary artery disease, circulating endothelial progenitor cells, and the role of HMG-CoA reductase inhibitors. Kidney Int. 2005;67:1672–1676. doi: 10.1111/j.1523-1755.2005.00261.x. [DOI] [PubMed] [Google Scholar]
  • [152].Valgimigli M, Rigolin GM, Fucili A, Porta MD, Soukhomovskaia O, Malagutti P, Bugli AM, Bragotti LZ, Francolini G, Mauro E, Castoldi G, Ferrari R. CD34+ and endothelial progenitor cells in patients with various degrees of congestive heart failure. Circulation. 2004;110:1209–1212. doi: 10.1161/01.CIR.0000136813.89036.21. [DOI] [PubMed] [Google Scholar]
  • [153].Vasa M, Fichtlscherer S, Aicher A, Adler K, Urbich C, Martin H, Zeiher AM, Dimmeler S. Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ Res. 2001;89:E1–7. doi: 10.1161/hh1301.093953. [DOI] [PubMed] [Google Scholar]
  • [154].Fadini GP, Maruyama S, Ozaki T, Taguchi A, Meigs J, Dimmeler S, Zeiher AM, de Kreutzenberg S, Avogaro A, Nickenig G, Schmidt-Lucke C, Werner N. Circulating progenitor cell count for cardiovascular risk stratification: a pooled analysis. PLoS One. 2010;5:e11488. doi: 10.1371/journal.pone.0011488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Thum T, Fraccarollo D, Schultheiss M, Froese S, Galuppo P, Widder JD, Tsikas D, Ertl G, Bauersachs J. Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes. 2007;56:666–674. doi: 10.2337/db06-0699. [DOI] [PubMed] [Google Scholar]
  • [156].Zhang W, Wang XH, Chen SF, Zhang GP, Lu N, Hu RM, Jin HM. Biphasic response of endothelial progenitor cell proliferation induced by high glucose and its relationship with reactive oxygen species. J Endocrinol. 2008;197:463–470. doi: 10.1677/JOE-08-0036. [DOI] [PubMed] [Google Scholar]
  • [157].DiStefano V, Cencioni C, Zaccagnini G, Magenta A, Capogrossi MC, Martelli F. p66ShcA modulates oxidative stress and survival of endothelial progenitor cells in response to high glucose. Cardiovasc Res. 2009;82:421–429. doi: 10.1093/cvr/cvp082. [DOI] [PubMed] [Google Scholar]
  • [158].Ceradini DJ, Yao D, Grogan RH, Callaghan MJ, Edelstein D, Brownlee M, Gurtner GC. Decreasing intracellular superoxide corrects defective ischemia-induced new vessel formation in diabetic mice. J Biol Chem. 2008;283:10930–10938. doi: 10.1074/jbc.M707451200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Galasso G, Schiekofer S, Sato K, Shibata R, Handy DE, Ouchi N, Leopold JA, Loscalzo J, Walsh K. Impaired angiogenesis in glutathione peroxidase-1-deficient mice is associated with endothelial progenitor cell dysfunction. Circ Res. 2006;98:254–261. doi: 10.1161/01.RES.0000200740.57764.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Ingram DA, Krier TR, Mead LE, McGuire C, Prater DN, Bhavsar J, Saadatzadeh MR, Bijangi-Vishehsaraei K, Li F, Yoder MC, Haneline LS. Clonogenic endothelial progenitor cells are sensitive to oxidative stress. Stem Cells. 2007;25:297–304. doi: 10.1634/stemcells.2006-0340. [DOI] [PubMed] [Google Scholar]
  • [161].Dai J, Zhu X, Yoder MC, Wu Y, Colman RW. Cleaved high-molecular-weight kininogen accelerates the onset of endothelial progenitor cell senescence by induction of reactive oxygen species. Arterioscler Thromb Vasc Biol. 2011;31:883–889. doi: 10.1161/ATVBAHA.110.222430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Tongers J, Losordo DW, Landmesser U. Stem and progenitor cell-based therapy in ischaemic heart disease: promise, uncertainties, and challenges. Eur Heart J. 2011;32:1197–1206. doi: 10.1093/eurheartj/ehr018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Ohshima M, Li TS, Kubo M, Qin SL, Hamano K. Antioxidant therapy attenuates diabetes-related impairment of bone marrow stem cells. Circ J. 2009;73:162–166. doi: 10.1253/circj.cj-08-0123. [DOI] [PubMed] [Google Scholar]
  • [164].Callaghan MJ, Ceradini DJ, Gurtner GC. Hyperglycemia-induced reactive oxygen species and impaired endothelial progenitor cell function. Antioxid Redox Signal. 2005;7:1476–1482. doi: 10.1089/ars.2005.7.1476. [DOI] [PubMed] [Google Scholar]
  • [165].Yao EH, Fukuda N, Matsumoto T, Kobayashi N, Katakawa M, Yamamoto C, Tsunemi A, Suzuki R, Ueno T, Matsumoto K. Losartan improves the impaired function of endothelial progenitor cells in hypertension via an antioxidant effect. Hypertens Res. 2007;30:1119–1128. doi: 10.1291/hypres.30.1119. [DOI] [PubMed] [Google Scholar]
  • [166].Yao EH, Fukuda N, Matsumoto T, Katakawa M, Yamamoto C, Han Y, Ueno T, Kobayashi N, Matsumoto K. Effects of the antioxidative beta-blocker celiprolol on endothelial progenitor cells in hypertensive rats. Am J Hypertens. 2008;21:1062–1068. doi: 10.1038/ajh.2008.233. [DOI] [PubMed] [Google Scholar]
  • [167].Thum T, Fraccarollo D, Thum S, Schultheiss M, Daiber A, Wenzel P, Munzel T, Ertl G, Bauersachs J. Differential effects of organic nitrates on endothelial progenitor cells are determined by oxidative stress. Arterioscler Thromb Vasc Biol. 2007;27:748–754. doi: 10.1161/01.ATV.0000258787.18982.73. [DOI] [PubMed] [Google Scholar]
  • [168].Thum T, Wiebking V, Ertl G, Bauersachs J. Organic nitrates differentially modulate circulating endothelial progenitor cells and endothelial function in patients with symptomatic coronary artery disease. Antioxid Redox Signal. 2011;15:925–931. doi: 10.1089/ars.2010.3503. [DOI] [PubMed] [Google Scholar]
  • [169].Song H, Cha MJ, Song BW, Kim IK, Chang W, Lim S, Choi EJ, Ham O, Lee SY, Chung N, Jang Y, Hwang KC. Reactive oxygen species inhibit adhesion of mesenchymal stem cells implanted into ischemic myocardium via interference of focal adhesion complex. Stem Cells. 2010;28:555–563. doi: 10.1002/stem.302. [DOI] [PubMed] [Google Scholar]
  • [170].Kubo M, Li TS, Suzuki R, Shirasawa B, Morikage N, Ohshima M, Qin SL, Hamano K. Hypoxic preconditioning increases survival and angiogenic potency of peripheral blood mononuclear cells via oxidative stress resistance. Am J Physiol Heart Circ Physiol. 2008;294:H590–595. doi: 10.1152/ajpheart.00856.2007. [DOI] [PubMed] [Google Scholar]
  • [171].Kubo M, Li TS, Suzuki R, Ohshima M, Qin SL, Hamano K. Short-term pretreatment with low-dose hydrogen peroxide enhances the efficacy of bone marrow cells for therapeutic angiogenesis. Am J Physiol Heart Circ Physiol. 2007;292:H2582–2588. doi: 10.1152/ajpheart.00786.2006. [DOI] [PubMed] [Google Scholar]
  • [172].Carriere A, Ebrahimian TG, Dehez S, Auge N, Joffre C, Andre M, Arnal S, Duriez M, Barreau C, Arnaud E, Fernandez Y, Planat-Benard V, Levy B, Penicaud L, Silvestre JS, Casteilla L. Preconditioning by mitochondrial reactive oxygen species improves the proangiogenic potential of adipose-derived cells-based therapy. Arterioscler Thromb Vasc Biol. 2009;29:1093–1099. doi: 10.1161/ATVBAHA.109.188318. [DOI] [PubMed] [Google Scholar]
  • [173].Tojo T, Ushio-Fukai M, Yamaoka-Tojo M, Ikeda S, Patrushev N, Alexander RW. Role of gp91phox (Nox2)-containing NAD(P)H oxidase in angiogenesis in response to hindlimb ischemia. Circulation. 2005;111:2347–2355. doi: 10.1161/01.CIR.0000164261.62586.14. [DOI] [PubMed] [Google Scholar]
  • [174].Haddad P, Dussault S, Groleau J, Turgeon J, Maingrette F, Rivard A. Nox2-derived reactive oxygen species contribute to hypercholesterolemia-induced inhibition of neovascularization: effects on endothelial progenitor cells and mature endothelial cells. Atherosclerosis. 2011;217:340–349. doi: 10.1016/j.atherosclerosis.2011.03.038. [DOI] [PubMed] [Google Scholar]
  • [175].Haddad P, Dussault S, Groleau J, Turgeon J, Michaud SE, Menard C, Perez G, Maingrette F, Rivard A. Nox2-containing NADPH oxidase deficiency confers protection from hindlimb ischemia in conditions of increased oxidative stress. Arterioscler Thromb Vasc Biol. 2009;29:1522–1528. doi: 10.1161/ATVBAHA.109.191437. [DOI] [PubMed] [Google Scholar]
  • [176].Turgeon J, Haddad P, Dussault S, Groleau J, Maingrette F, Perez G, Rivard A. Protection against vascular aging in Nox2-deficient mice: Impact on endothelial progenitor cells and reparative neovascularization. Atherosclerosis. 2012;223:122–129. doi: 10.1016/j.atherosclerosis.2012.05.003. [DOI] [PubMed] [Google Scholar]
  • [177].Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, Poznansky MC, Kos CH, Pollak MR, Brown EM, Scadden DT. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006;439:599–603. doi: 10.1038/nature04247. [DOI] [PubMed] [Google Scholar]
  • [178].Walkley CR, Olsen GH, Dworkin S, Fabb SA, Swann J, McArthur GA, Westmoreland SV, Chambon P, Scadden DT, Purton LE. A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency. Cell. 2007;129:1097–1110. doi: 10.1016/j.cell.2007.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Oikawa A, Siragusa M, Quaini F, Mangialardi G, Katare RG, Caporali A, van Buul JD, van Alphen FP, Graiani G, Spinetti G, Kraenkel N, Prezioso L, Emanueli C, Madeddu P. Diabetes mellitus induces bone marrow microangiopathy. Arterioscler Thromb Vasc Biol. 2010;30:498–508. doi: 10.1161/ATVBAHA.109.200154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Amrani YM, Gill J, Matevossian A, Alonzo ES, Yang C, Shieh JH, Moore MA, Park CY, Sant’Angelo DB, Denzin LK. The Paf oncogene is essential for hematopoietic stem cell function and development. J Exp Med. 2011;208:1757–1765. doi: 10.1084/jem.20102170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Wang XR, Zhang MW, Chen DD, Zhang Y, Chen AF. AMP-activated protein kinase rescues the angiogenic functions of endothelial progenitor cells via manganese superoxide dismutase induction in type 1 diabetes. Am J Physiol Endocrinol Metab. 2011;300:E1135–1145. doi: 10.1152/ajpendo.00001.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Hole PS, Pearn L, Tonks AJ, James PE, Burnett AK, Darley RL, Tonks A. Ras-induced reactive oxygen species promote growth factor-independent proliferation in human CD34+ hematopoietic progenitor cells. Blood. 2010;115:1238–1246. doi: 10.1182/blood-2009-06-222869. [DOI] [PubMed] [Google Scholar]
  • [183].Chen J, Song M, Yu S, Gao P, Yu Y, Wang H, Huang L. Advanced glycation endproducts alter functions and promote apoptosis in endothelial progenitor cells through receptor for advanced glycation endproducts mediate overpression of cell oxidant stress. Mol Cell Biochem. 2010;335:137–146. doi: 10.1007/s11010-009-0250-y. [DOI] [PubMed] [Google Scholar]
  • [184].Wang JY, Lee YT, Chang PF, Chau LY. Hemin promotes proliferation and differentiation of endothelial progenitor cells via activation of AKT and ERK. J Cell Physiol. 2009;219:617–625. doi: 10.1002/jcp.21711. [DOI] [PubMed] [Google Scholar]
  • [185].Schraml E, Fuchs R, Kotzbeck P, Grillari J, Schauenstein K. Acute adrenergic stress inhibits proliferation of murine hematopoietic progenitor cells via p38/MAPK signaling. Stem Cells Dev. 2009;18:215–227. doi: 10.1089/scd.2008.0072. [DOI] [PubMed] [Google Scholar]
  • [186].You D, Cochain C, Loinard C, Vilar J, Mees B, Duriez M, Levy BI, Silvestre JS. Hypertension impairs postnatal vasculogenesis: role of antihypertensive agents. Hypertension. 2008;51:1537–1544. doi: 10.1161/HYPERTENSIONAHA.107.109066. [DOI] [PubMed] [Google Scholar]
  • [187].Prus E, Fibach E. The effect of the copper chelator tetraethylenepentamine on reactive oxygen species generation by human hematopoietic progenitor cells. Stem Cells Dev. 2007;16:1053–1056. doi: 10.1089/scd.2007.0052. [DOI] [PubMed] [Google Scholar]
  • [188].Zhang X, Sejas DP, Qiu Y, Williams DA, Pang Q. Inflammatory ROS promote and cooperate with the Fanconi anemia mutation for hematopoietic senescence. J Cell Sci. 2007;120:1572–1583. doi: 10.1242/jcs.003152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Iiyama M, Kakihana K, Kurosu T, Miura O. Reactive oxygen species generated by hematopoietic cytokines play roles in activation of receptor-mediated signaling and in cell cycle progression. Cell Signal. 2006;18:174–182. doi: 10.1016/j.cellsig.2005.04.002. [DOI] [PubMed] [Google Scholar]

RESOURCES