Skip to main content
Antioxidants & Redox Signaling logoLink to Antioxidants & Redox Signaling
. 2009 Oct;11(10):2517–2533. doi: 10.1089/ars.2009.2582

Novel Role of NADPH Oxidase in Angiogenesis and Stem/Progenitor Cell Function

Masuko Ushio-Fukai 1,, Norifumi Urao 1
PMCID: PMC2821135  PMID: 19309262

Abstract

Neovascularization is involved in normal development and wound repair as well as ischemic heart disease and peripheral artery disease. Both angiogenesis and vasculogenesis [de novo new vessel formation through mobilization of stem/progenitor cells from bone marrow (BM) and their homing to the ischemic sites] contribute to the formation of new blood vessels after tissue ischemia. Angiogenesis is dependent on cell proliferation, migration, and capillary tube formation in endothelial cells (ECs). Stem/progenitor cells have been used for cell-based therapy to promote revascularization after peripheral or myocardial ischemia. Excess amounts of reactive oxygen species (ROS) are involved in senescence and apoptosis of ECs and stem/progenitor cells, causing defective neovascularization. ROS at low levels function as signaling molecules to mediate cell proliferation, migration, differentiation, and gene expression. NADPH oxidase is one of the major sources of ROS in ECs and stem/progenitor cells, and is activated by various growth factors, cytokines, hypoxia, and ischemia. ROS derived from NADPH oxidase play an important role in redox signaling linked to angiogenesis ECs, as well as stem/progenitor cell mobilization, homing, and differentiation, thereby promoting neovascularization. Understanding these mechanisms may provide insight into NADPH oxidase and its mediators as potential therapeutic targets for ischemic heart and limb disease. Antioxid. Redox Signal. 11, 2517–2533.

Introduction

Neovascularization is involved in normal development and wound repair, as well as various pathophysiologies such as ischemic heart disease and peripheral artery disease. Postnatal new blood vessel formation depends on angiogenesis (formation of new capillaries from preexisting vessels), arteriogenesis (formation of collateral arteries), and vasculogenesis [de novo new vessel formation through bone marrow (BM)-derived angiogenic stem/progenitor cells] (96, 109, 163, 196) (Fig. 1). Angiogenesis occurs through cell proliferation, migration, and capillary tube formation in endothelial cells (ECs) (Fig. 1). Vascular endothelial growth factor (VEGF) is one of the most potent angiogenesis growth factors, and its mitogenic and chemotactic effects in ECs are mediated mainly through VEGF receptor type 2 (VEGFR2, KDR/Flk1) (138). VEGF binding induces autophosphorylation of VEGFR2, which is followed by activation of downstream signaling pathways such as mitogen-activated protein kinases (MAPKs) and Akt, thereby stimulating angiogenesis (Fig. 2). Mobilization of endothelial progenitor cells (EPCs) and stem/progenitor cells from BM, as well as their homing to the ischemic tissues, is important for postnatal neovascularization (6, 13, 192) (Fig. 1). Understanding mechanisms by which angiogenesis signaling is regulated in ECs, or BM stem/progenitor cells are mobilized and home to the site of neovascularization will lead to development of new therapeutic strategies for ischemic cardiovascular diseases.

FIG. 1.

FIG. 1.

Role of NADPH oxidase in ischemia-induced neovascularization. Postnatal new blood vessel formation depends on angiogenesis (formation of new capillaries from preexisting vessels) and vasculogenesis (de novo new-vessel formation through bone marrow (BM)-derived angiogenic stem/progenitor cells. In response to ischemia, Nox2-based NADPH oxidase expression and reactive oxygen species (ROS) production is increased in BM, which in turn stimulates stem/progenitor cell mobilization from BM to the circulation. NADPH oxidase (NOX)-derived ROS are also involved in stem/progenitor homing and the angiogenic capacity to promote neovascularization of ischemic tissues. Angiogenesis growth factors such as VEGF, produced in response to ischemia, also stimulate endothelial cell (EC) migration, proliferation, and tube formation through an increase in NADPH oxidase–derived ROS to promote angiogenesis. Thus, NADPH oxidase plays an important role in postnatal angiogenesis and stem/progenitor cell function.

FIG. 2.

FIG. 2.

Role of NADPH oxidase in redox signaling linked to neovascularization. NADPH oxidase (Nox) is activated by angiogenesis factors such as VEGF or hypoxia, ischemia, cytokines, and angiotensin II (Ang II). ROS levels are balanced by ROS-generating enzyme, NADPH oxidase, and antioxidant enzymes such as superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPx). Nox-derived ROS activate redox-sensitive kinases/enzymes, including Akt, Src, MAP kinases and eNOS, or inactivate specific protein tyrosine phosphatases (PTPs), which negatively regulate protein tyrosine kinases (PTKs) or protein tyrosine kinases (PTKs) through oxidation of proteins. In addition, ROS activate various redox-sensitive transcription factors. The overall biologic impact of Nox activation thus involves endothelial cell and stem/progenitor cell proliferation, migration, differentiation, senescence, apoptosis, and gene expression, thereby regulating angiogenesis and vasculogenesis, which contribute to neovascularization. NF-κB, nuclear factor kappa B; AP1, activator protein 1; HIF1, hypoxia inducible factor 1; MAPK, mitogen-activated protein kinase; eNOS, endothelial nitric oxide synthase; PTP-SH, reduced form of protein tyrosine phosphatases; PTP-SOH, oxidized form of protein tyrosine phosphatases; RTK, receptor tyrosine kinases; PTK, protein tyrosine kinases.

Reactive oxygen species (ROS) such as superoxide anion (O2•−) and hydrogen peroxide (H2O2) play an important role in normal cell growth, migration, differentiation, apoptosis, and senescence (58, 70). Excess amounts of ROS are toxic and involved in EC and stem/progenitor cell senescence and apoptosis (34), thereby contributing to oxidative stress–dependent cardiovascular diseases such as hypertension, heart failure, atherosclerosis, and diabetes (233). By contrast, ROS at low levels function as signaling molecules to regulate angiogenesis and vasculogenesis (42, 135, 139, 185, 189, 194, 201, 207, 210). Signal transduction activated by ROS, “redox signaling,” has been an emerging area of investigation.

NADPH oxidase is one of major sources of ROS in ECs and stem/progenitor cells. In phagocytic cells, NADPH oxidases consist of membrane-associated cytochrome b558, comprising the catalytic gp91phox and regulatory p22phox subunits, and cytosolic components including p47phox, p67phox, p40phox, and the small GTPase Rac1 (17) (Fig. 1). In nonphagocytic cells, several homologues of gp91phox (also termed Nox2), including Nox1, Nox3, Nox4, and Nox5, as well as the dual oxidases (Duox)1/2, have been identified (64, 116, 120). Nox1, Nox2, Nox4, and Nox5 (only human) are expressed in ECs, whereas Nox2 and Nox4 are found in stem/progenitor cells. NADPH oxidase is activated by angiogenic growth factors and cytokines such as VEGF, angiopoietin-1, shear or mechanical stress, hypoxia and G protein–coupled receptor agonists, including angiotensin II (Ang II) in ECs and stem/progenitor cells (77, 208). NADPH oxidase–derived ROS are involved in redox signaling pathways, leading to angiogenesis in ECs, as well as various functions of stem/progenitor cells, including proliferation, migration, differentiation, and gene expression (27, 57, 77, 88, 158, 205, 210, 230), thereby regulating postnatal neovascularization (8, 201, 205) (Fig. 2). Because the role of excess amounts of ROS linked to stem/progenitor cell dysfunction has been reviewed by others (23, 34, 181, 203, 220, 233), this review briefly describes this aspect and mainly summarizes the recent progress and information on the role of NADPH oxidase and physiologic levels of ROS in angiogenesis and stem/progenitor cell function.

Role of NADPH Oxidase in Angiogenesis in ECs

Exogenous ROS increase VEGF or VEGFR2 expression (66) and stimulate EC proliferation and migration (44, 135, 234), as well as cytoskeletal reorganization (216) and tubular morphogenesis (183) of ECs. Furthermore, angiogenesis growth factors such as VEGF and angiopoietin-1, as well as Ang II or 20-hydroxyeicosatetraenoic acid (20-HETE), a product of cytochrome p450, or leptin, a circulating adipocytokine, hypoxia/reoxygenation, or adhesion of activated polymor-phonuclear leukocytes to ECs, stimulate angiogenesis-related responses via an increase in ROS (73, 77, 84, 88, 125, 210, 228, 230, 235). Natural antioxidants such as green tea catechins and vitamin E, as well as polyphenols from red wine, prevent angiogenic responses by decreasing ROS levels in ECs (195). These suggest that many of angiogenic-related responses are dependent on ROS in cultured ECs.

ROS are generated from a number of sources including the NADPH oxidase, mitochondrial electron-transport system, xanthine oxidase, cytochrome p450, uncoupled NOS, and myeloperoxidase (70, 128). Complex interactions may occur among different sources of ROS, as well as feed-back and feed-forward regulation of ROS accumulation (128). Mitochondria-derived ROS are involved in hypoxia inducible factor 1 (HIF1) stabilization (22) and angiogenesis (46); however, the role in receptor-activated signal transduction in ECs remains unclear. NADPH oxidase has emerged as a major source of ROS in nonphagocytic cells, including ECs, and is activated by numerous angiogenesis stimuli including VEGF, angiopoietin 1, cytokines, shear stress, hypoxia, and G protein–coupled receptor agonists, including Ang II (70) (Fig. 2). The prototype phagocyte NADPH oxidase consists of membrane-bound gp91 phox (now also termed Nox2) and p22phox, which compose the flavocytochrome b558, together with the cytosolic p47phox, p67phox, p40phox, and the small GTPase Rac (Fig. 1). Nonphagocytic NADPH oxidase (Nox) consists of several homologues of Nox2, including Nox1, Nox3, Nox4, and Nox5, as well as the dual oxidases (Duoxs) (64, 65, 117, 118). In ECs, all the phagocytic NADPH oxidase subunits including Nox2, p22phox, Rac1, p47phox, as well as Nox1, Nox4 and Nox5, are expressed. Various Nox enzymes contribute to ROS production of different degrees, locations, and EC types.

Evidence suggests that several Nox enzymes are involved in angiogenesis in ECs. Nox2 is activated by various stimulants in ECs (61, 62, 67, 126, 210). VEGF and angiopoietin-1 stimulate EC migration via activation of Nox2-based NADPH oxidase, suggesting an essential role of Nox2 in angiogenesis in ECs (4, 45, 77, 88, 130, 210, 230). Nox1 stimulates branching morphogenesis in sinusoidal ECs (106), and Nox1 overexpression increases VEGF and VEGF-receptor expression and MMP activity, thereby promoting vascularization (11). Nox4 is involved in basal ROS production and proliferation in ECs (157). Datla et al. (49) reported that overexpression of Nox4 enhances, whereas dominant negative Nox4 inhibits VEGF-induced EC migration and proliferation. The most recent study shows that overexpression of Nox5 stimulates ROS production, proliferation, and formation of capillary-like structures, whereas Nox5 siRNA prevents thrombin-induced responses in human ECs (21). Note that Nox5 is present in most mammals but is absent in rodents, in which its function may be taken over by another Nox. The role of Rac1-derived ROS in VEGF-induced VEGF expression (51) and EC migration also has been reported (77, 110, 146, 210, 214). These suggest that various Nox homologues (Nox1, Nox2, Nox4, Nox5) play an important role in angiogenesis in ECs (Fig. 1), which may depend on species and experimental conditions.

Role of NADPH Oxidase in Angiogenesis In Vivo

ROS play an important role in physiologic and pathologic angiogenesis in vivo. ROS are increased during the reperfusion of the ischemic retina, which contributes to upregulation of VEGF mRNA (113). Short periods of ischemia/reperfusion or preconditioning induce an increase in ROS, thereby stimulating myocardial angiogenesis and arteriogenesis (48, 72, 115). Thiol antioxidant, N-acetylcysteine attenuates EC invasion and angiogenesis in a tumor model in vivo (29). Pigment epithelium-derived growth factor, which has antioxidant properties, inhibits angiogenesis and melanoma growth (1). ROS inhibitors prevent neovascularization in a mouse model of angiogenesis (160, 237). Angiopoietin1-induced angiogenesis is enhanced in catalase-knockout mice, suggesting an involvement of H2O2 in angiogenesis in vivo (105). In a pathophysiologic state, a strong correlation between ROS elevation, neovascularization, and VEGF expression has been reported in eyes of humans with diabetes (30, 55, 56) and in an animal model of atherosclerosis (171). Transgenic mice overexpressing MnSOD in ECs prevent diabetic retinopathy through reducing O2 production and VEGF expression in vivo (68). Antiangiogenic therapy reduces plaque growth and intimal neovascularization in atherosclerosis (148). Antioxidant vitamins C and E reduce VEGF and VEGFR-2 expression in apolipoprotein-E–deficient mice (153), which may contribute to their inhibitory effects on atherosclerosis. These studies suggest that ROS are required for neovascularization in vivo.

The role of NADPH oxidase in pathologic and physiologic angiogenesis in vivo has been demonstrated. Neovascularization in response to VEGF is inhibited in Nox2−/− mice as well as in mice treated with antioxidant or NADPH oxidase inhibitor or gp91ds-tat (8, 89, 210). Hindlimb ischemia stimulates Nox2-dependent ROS production in ischemic tissues, and blood-flow recovery and angiogenesis in response to ischemia are markedly impaired in Nox2−/− mice (8, 201). Similarly, Nox2 expression and ROS production are increased in a mouse retinopathy model, which is attenuated by NADPH oxidase inhibitor or gp91ds-tat (8). With an in vivo arteriovenous loop–containing tissue engineering chamber, Jianq et al. (100) recently showed that Nox2 plays an important role in vascularization and tissue growth. Vallet et al. (211) reported that Nox4 expression is upregulated in new capillaries in brain ischemia–induced angiogenesis of mice. HMG-CoA reductase inhibitor, statins, which block Rac1 activity (122), prevent angiogenesis in vivo (221). Overexpression of Nox1 increases VEGF and VEGF-receptor expression and matrix metalloproteinase (MMP) activity through increases in ROS, thereby promoting tumor angiogenesis (11). Transgenic mice overexpressing p22 phox show enhanced VEGF expression and neovascularization of experimental atheromas (102). Myocardial ischemia/reperfusion of mice increases ROS in a p47phox-dependent manner, which is required for VEGF expression, sprouting, and vessel outgrowth (41). Optimal levels of NADPH oxidase–derived ROS are required for p38MAPK activation and coronary collateral growth in a rat model of repetitive ischemia (168). By contrast, overproduction of Nox2-derived ROS rather contributes to impairment of postischemic neovascularization in pathologic conditions such as diabetes (53). In line with this report, extracellular SOD–deficient mice show impaired ischemia-induced neovascularization and enhanced O2 levels, which are rescued by SOD mimetic infusion (104). These studies support a double-edged role of ROS in postischemic neovascularization; physiologic levels of ROS are required, but excess amounts of ROS are inhibitory for new blood vessel formation.

Redox Signaling in Angiogenesis

ROS can act as signaling molecules for activation of diverse signaling pathways by oxidation of reactive cysteine on the specific target molecules, including kinases, phosphatases (43, 124, 167), redox-sensitive transcription factors, AP1, Ets, NF-κB, and HIF1 (190), as well as redox-sensitive genes, VEGF, MMPs, cyclooxygenase-2 (COX-2), urokinase plasminogen activator (uPA), and plasminogen activator inhibitor-1 (PAI-1), which are involved in angiogenesis (207, 209) (Fig. 2).

NADPH oxidase is activated by numerous stimuli including VEGF, EGF, cytokines, shear stress, hypoxia, and G protein–coupled receptor agonists including Ang II in ECs (70) (Fig. 2). VEGF binds to two tyrosine kinase receptors, VEGF receptor-1 (VEGFR1, Flt-1) and VEGFR2 in ECs. The mitogenic and chemotactic effects of VEGF in ECs are mediated mainly through VEGFR2 (138), which is activated through autophosphorylation of tyrosine residues in the cytoplasmic kinase domain. This event is followed by activation of downstream signaling pathways, such as mitogen-activated protein kinases, Akt, and eNOS, which are essential for EC migration and proliferation (138) (Fig. 2). Nox2-derived ROS are involved in VEGF-induced VEGFR2 autophosphorylation in ECs (45, 88, 210, 230). Angiopoietin 1 binding to the Tie-2 receptor also stimulates Nox2-based NADPH oxidase, which is required for phosphorylation of Akt and ERK linked to EC chemotaxis (40, 77). p47phox siRNA inhibits VEGF-stimulated phosphoinositide 3-kinase (PI3K)-Akt-forkhead as well as p38MAPK, but not ERK1/2 or JNK, in ECs (3). ROS also are important for VEGF-induced cSrc activation, phosphorylation of VE-cadherin, thereby disrupting cell-to-cell contacts of established vessels to promote angiogenesis in ECs (130, 212, 229). VEGF also induces peroxynitrite formation, which is involved in phosphorylation of VEGFR2 and cSrc as well as EC migration and tube formation (54). Thus, not only ROS but also peroxynitrite mediate VEGF angiogenic signaling in ECs.

Evidence suggests that NADPH oxidases are localized within discrete subcellular compartments, which is required for localizing ROS production and activation of specific redox-signaling pathways that mediate various cell functions (198, 206). EC migration is a key event for tissue repair in response to injury, angiogenesis, and wound healing. Rac1- and Nox2-dependent NADPH oxidase plays an important role in EC migration (2, 45, 89, 210). The PI3K-Rac pathway is involved in localized production of ROS at the membrane ruffles (155), which is required for cytoskeletal reorganization and directed cell migration (89, 147). Nox2 and its regulatory subunits, p47phox and p67phox are also targeted to the focal complexes or membrane ruffles in lamellipodia (89, 225, 226). Wu et al. (226) identified targeting molecules that may specify the site of ROS production at lamellipodial focal complexes during EC migration. p47phox binds to the orphan adaptor TRAF4, which in turn binds to the focal contact scaffold Hic-5, thereby targeting p47phox to the focal complexes. The mechanism for targeting NADPH oxidase to the lamellipodial leading edge and membrane ruffles is through the interaction of p47phox with moesin and WAVE1, which are enriched within this specific structural compartment (47, 223). WAVE1 catalyzes actin nucleation responsible for lamellar structure in a Rac1-dependent manner, and thus p47phox–WAVE1 complexes contain Rac1 and Rac1 effectors PAK1, which phosphorylates p47phox. Antioxidants and inhibition of p47phox–WAVE1 interaction block ROS production and ruffle formation (225).

Another important targeting protein is IQGAP1, an actin-binding scaffold protein and Rac1 effector that is required for Rac-mediated polarized cell migration (26, 137). IQGAP1 functions as a scaffold protein to target Nox2 and Rac1 to the leading edge to localize ROS production, which may contribute to ROS-dependent directional EC migration (89). Moreover, Nox2 and Nox4 activate distinct redox-signaling pathways in HEK293 cells transfected with Nox2 or Nox4 (9), suggesting that differential expression and localization of Nox enzymes may determine the specificity of activation of redox signaling.

The signaling properties of ROS are due, in part, to reversible oxidative inactivation of redox-sensitive target proteins, including protein tyrosine phosphatases (PTPs) (43, 124, 167) (Fig. 2). Several PTPs, including SHP-1, SHP-2, and low-molecular-weight PTP (HCPTPA), inducibly associate with VEGFR2 after VEGF stimulation (74, 85, 111). HCPTPA overexpression inhibits VEGF-induced VEGFR2 autophosphorylation (85). SHP-1 mediates a TNF-α–induced inhibitory effect on VEGFR2 phosphorylation without affecting VEGF-induced responses in ECs (151). A recent report shows that SHP-1 constitutively binds to VEGFR2, and siRNA enhances VEGFR2 autophosphorylation and EC proliferation without affecting migration (24). SHP-2 negatively regulates VEGFR2 signaling in ECs that are cultured only on type I collagen (143). PTP1B siRNA or small-molecule inhibitor enhances VEGF-induced VEGFR2 autophosphorylation and proliferation of ECs (152, 186). PTP1B inhibitor promotes neovascularization in a mouse Matrigel model (186). High cell density–enhanced PTP1 (DEP-1)/CD148 attenuates phosphorylation of VEGFR2 in contact-inhibited confluent ECs (69). Angiopoietin-1 stimulates association of SHP-2 with the phosphorylated Tie-2 receptor in ECs (86), which in turn inhibits the PI3K-dependent signaling involved in EC migration. Nox-derived ROS inactivate PTP-PEST at focal contacts, thereby promoting membrane ruffling and endothelial migration (10). Thus, it is important to determine which PTPs are reversibly oxidized and inactivated by angiogenesis growth factor–induced ROS, thereby promoting tyrosine phosphorylation–dependent redox signaling linked to angiogenesis (Fig. 2).

Lipid phosphatase and tumor suppressor, PTEN, which dephosphorylates phosphatidylinositol 3,4,5-triphosphate (PIP3), a product of the PI3K activation, is also subject to the oxidative inactivation by ROS (114). ROS regulate an EGF-induced increase in VEGF and HIF-1α expression through activation of the PI3K/Akt/p706K pathway, which is involved in angiogenesis in ovarian cancer cells (131). Although underlying mechanisms remain unclear, it is possible that ROS-dependent oxidative inactivation of PTEN (114) may be involved in this response. Similarly, MnSOD overexpression increases mitochondrial H2O2, leading to PTEN oxidation and PIP3 formation at the plasma membrane, and Akt activation, which results in VEGF expression and EC tube formation (46).

In addition to phosphatases, several serine/threonine kinases, such as cAMP dependent kinase (PKA), cGMP-dependent kinase (PKG), Akt kinase, S6 kinase, and protein kinase C (PKC), all contain a Cys-SH residue within their active-site domain. These Cys-SH residues are not required for activity, but they are fully conserved among these kinases. One may speculate that interaction with regulatory proteins may be modulated by the reversible oxidation of this Cys-SH.

Role of NADPH Oxidase in Stem/Progenitor Cell Function

NADPH oxidase and stem/progenitor cell differentiation, proliferation, and senescence

Stem cells and EPCs hold great promise for tissue repair and regenerative medicine and play a significant role in neovascularization of ischemic tissue. Expression of “stemness” genes that distinguish stem cells from mature cells, as well as stress-response genes involved in redox balance, is a characteristic feature of stem cells, suggesting that one essential attribute of the stemness is high resistance to stress (98, 166). Of note, ROS levels in endothelial progenitor cells are lower than those in mature ECs, which is due to higher expression of antioxidant enzymes (MnSOD, catalase, glutathione peroxidase) and is required for preserving stemness, such as an undifferentiated, self-renewing state (50). Hypoxia effectively maintains the biologic characteristics of CD34+ cells through keeping lower intracellular ROS levels by regulating NADPH oxidase (57). Factors promoting self-renewal cause the reduction of the redox state, whereas molecules promoting differentiation lead to excess oxidation in neural progenitor cells (154). Hyperbaric oxygen stimulates vasculogenic stem/progenitor cell growth and differentiation in vivo through an increase in physiologic levels of ROS and Nox2 (141). Thus, ROS seem to be involved in the balance between self-renewal and differentiation of stem/progenitor cells (76) (Fig. 3).

FIG. 3.

FIG. 3.

Possible mechanism by which NADPH oxidase is involved in stem/progenitor cell mobilization from bone marrow, and homing and angiogenesis at the site of neovascularization. In response to ischemic stress, hematopoietic and angiogenic factors are increased in bone marrow, which in turn activates NADPH oxidase (NOX) to exit quiescent stem cells from endosteal niche to stimulate proliferation and differentiation. This mechanism may be mediated through activation of matrix metalloproteinase (MMP)-9 and subsequent release of soluble Kit ligand (sKitL), which mobilizes proangiogenic stem/progenitor cells from niches. Furthermore, NOX-derived reactive oxygen species (ROS) may activate redox signaling events involved in proliferation, migration, differentiation, and survival of stem/progenitor cells. As a consequence, stem/progenitor cells are mobilized from bone marrow to the circulation and homed to the ischemic sites through the mechanism, dependent on adhesion and migration. NOX-derived ROS may be involved in expression of adhesion molecules on endothelial surface as well as SDF-1/CXCR4– or IL-8/CXCR2–mediated stem/progenitor cell migration, which may contribute to neovascularization with endothelial cell (EC)-mediated angiogenesis. (VEGF, vascular endothelial growth factor; PlGF, placental growth factor; SDF-1, stroma-derived factor-1; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte–macrophage colony-stimulating factor; ICAM1, intercellular adhesion molecule 1; VCAM-1, vascular cell adhesion molecule 1).

Although excess amounts of ROS are toxic to EPCs and stem/progenitor cells (233), optimal levels of ROS, which are balanced by ROS-generating and antioxidant enzymes, are involved in normal stem/progenitor cell functions, such as proliferation, differentiation, and survival (50, 63, 104). NADPH oxidase components are expressed in various stem/progenitor cells including circulating human CD34+ cells (57, 158, 159), mouse embryonic stem cells (27), skeletal muscle precursor cells (145), and rat mesenchymal stem cells (218). The high-resolution imaging of hematopoietic stem cells (HSCs) with the immunodetection of NOX indicates the presence of membrane raft–like microcompartments in which the assembly/activation of the NOX components may be functionally integrated for creating redox signaling platforms (60). Thus, NADPH oxidase–derived ROS may contribute to redox signaling involved in various function of stem/progenitor cells (Figs. 2 and 3). In undifferentiated human myeloid cell lines, Nox2, p22phox, p47phox, and p67phox expression is minimal, whereas O2-producing capacity is increased to the level of normal neutrophils during differentiation, with a concomitant increase in p47phox. Thus, low expression of p22phox and Nox2 is sufficient to obtain normal O2 production, and p47phox might play an important role in the functional differentiation of myeloid cells (94).

H2O2 plays a role in native osteoclast differentiation (188). Stimulation of BM monocyte-macrophage lineage cells with receptor activator of NF-κB ligand (RANKL) promotes differentiation into osteoclasts through an increase in ROS via TNF receptor–associated factor (TRAF) 6/Rac1/Nox1 pathway (123). Nox2 and Nox4 are expressed in skeletal muscle precursor cells, which are involved in proliferation of these cells (145). By contrast, proliferation of EPCs is limited by the capacity to divide and the onset of a nondividing state known as senescence. Inhibition of telomerase activity with ROS, increased by oxidized low-density lipoprotein (oxy-LDL), accelerates the onset of EPC senescence, which leads to impaired proliferative capacity (93). This mechanism is through telomerase inactivation by ROS and subsequent inactivation of Akt (90). Thus, ROS-mediated proliferation and senescence in stem/progenitor cells may be determined by the amount, duration, and location of ROS generation, which activates specific redox-signaling pathways.

In ES cells, Nox2, p22phox, p47phox, and p67phox of NADPH oxidase are expressed early in embryonic development, and the expression occurs in a consistent sequential fashion, indicating their involvement of ES cell differentiation (18). Mechanical strain or direct-current electrical field stimulation of mouse ES cells elevates intracellular ROS through upregulation of NADPH oxidase subunits, thereby increasing expression of HIF-1α and VEGF, which contributes to cardiovascular differentiation (176, 180). Low levels of H2O2 stimulate cardiomyogenesis of ES cells and induce proliferation of cardiomyocytes derived from ES cells. This response is associated with nuclear translocation of cyclin D1, downregulation of p27Kip1, phosphorylation of retinoblastoma (Rb), and an increase of Ki-67 expression and BrdU, which are inhibited by free radical scavengers. ROS-induced cardiomyogenesis of ES cells increases expression of NADPH oxidase components, indicating feed-forward regulation of ROS generation in cardiovascular differentiation of ES cells (27). Consistent with this, mechanical-strain stimulation of embryoid bodies grown from ES cells promotes cardiovascular differentiation through ROS elevation, which is followed by induction of Nox1 and Nox4, supporting feed-forward upregulation of ROS production (178). Similarly, cytokine cardiotrophin-1 (CT-1) stimulates proliferation of ES cell–derived cardiomyocytes through an increase in ROS and activation of redox-signaling cascades (177). CT-1 expression itself is also regulated by ROS and HIF-1α, suggesting existence of a feed-forward mechanism for promoting proliferation of ES-derived cardiomyocytes by ROS (14). Peroxisome proliferator–activated receptor alpha agonists enhance cardiomyogenesis of mouse ES cells through an increase in ROS and NADPH oxidase activity (182). The role of small GTPase Rac1 in cardiac differentiation of ES cells through NADPH oxidase is also reported (162). Nox4-derived ROS promote ES cell differentiation into smooth muscle cells (227) or cardiac cells (27, 127). Stimulation of embryoid bodies by PDGF-BB increases vascular sprouting and branching of capillary-like structures via NADPH oxidase–derived ROS and its downstream activation of ERK1/2 (119). Prenylflavonoid-induced cardiomyocyte differentiation of murine ES cells is also dependent on NADPH oxidase–derived ROS and p38MAP kinase with increased expression of Nox4 (52, 224). These reports suggest that NADPH oxidase–derived ROS are involved in cardiovascular ES cell differentiation.

NADPH oxidase and mobilization of stem/progenitor cells

Stem/progenitor cells and EPCs are mobilized from BM to the circulation in response to injury or cytokines, which contributes to postnatal neovascularization (6, 13, 192) (Figs. 1 and 3). By using Nox2-deficient mice and BM transplantation, we showed that Nox2 plays an essential for increase in ROS in BM after hindlimb ischemia, which is required for stem/progenitor cell mobilization from BM (205).

In another study, a cyclophosphamide-, G-CSF–, or IL-8–induced increase in circulating stem and progenitor numbers is inhibited in Nox2−/− mice (213). Moreover, renovascular hypertension of mice that is dependent on mechanical stress stimulates EPC mobilization through p47phox-dependent ROS, which is associated with an increase in BM SDF-1 and MMP-9 (173). In humans, the number of circulating CD34+ cells is low in X-linked patients with chronic granulomatous disease (CGD) in which the gp91phox (Nox2) coding gene, CYBB, is mutated (180a). These reports suggest that ischemic or mechanic stress increases circulating cytokine levels, thereby stimulating NOX-dependent ROS production in BM, which may promote reparative mobilization of stem/progenitor cells from BM and the resulting revascularization of ischemic tissues or tissue repair (Figs. 1 and 3).

HSCs are embedded in a local BM microenvironment, the so-called stem cell “niche” (31, 103, 241), and are maintained to be quiescent with low oxygen (184) and ROS levels (99) (Fig. 3). Recent reports indicate that increased ROS in BM facilitates HSCs to exit from the quiescent state, thereby stimulating proliferation and differentiation during the early steps of commitment (99, 202). Angiogenic growth factors or cytokines increase ROS in HSCs, thus stimulating cell growth (174, 175), and exit quiescent BM stem/progenitor cells to promote cell-cycle progression (87). ROS are involved in BM stem/progenitor cells proliferation, differentiation, migration, and survival in vitro (32, 36, 75, 175) and in vivo (12, 82, 97, 144, 202), which may contribute to ROS-mediated stem/progenitor cell mobilization from BM (Fig. 3). Hematopoietic cytokine G-CSF induces both serine phosphorylation and membrane translocation of p47phox to activate NAPDH oxidase, leading to cell proliferation of BM myeloid cells (244). NADPH oxidase–derived ROS are involved in myeloid expansion (32, 244) and monocytes/macrophage survival via regulating Akt and p38MAPK (219). These suggest that Nox-dependent ROS play an important role in redox signaling involved in the mobilization of some stages of BM progenitor cells (Fig. 3).

Mobilization of stem cells from BM is a dynamic process that requires release of chemocytokines, expression of adhesion molecules, activation of proteases, and stimulation of chemotaxis of stem/progenitor cells (163, 164). Thus, ROS also may affect on some of these events, thereby promoting egress of stem/progenitor cells from the BM to the circulation. BM sinusoidal ECs are functionally and phenotypically distinct from microvascular ECs of other organs (108) and constitutively express cytokines such as SDF-1 and adhesion molecule such as E-selectin and VCAM-1 that are important for hematopoietic stem cell mobilization from the BM (15, 165). Thus, NADPH oxidase might be involved in their expression in BM sinusoidal ECs. Moreover Nox2-derived ROS are downstream mediators of CXCR4-Akt signaling, thereby stimulating migration of stem/progenitor cells (205). VEGF also is known to induce proangiogenic cell mobilization including VEGFR2+ EPCs (101) and VEGFR1+ hematopoietic cells (164) Placental growth factor also promotes recruitment of VEGFR1+ HSCs from a quiescent to a proliferative microenvironment within the BM, favoring differentiation, mobilization, and reconstitution of hematopoiesis (163). VEGF induced by acute ischemia causes MMP-9 upregulation in the BM microenvironment, resulting in the disruption of SCF-cKit bond between stem/progenitor cells and niche cells (79). In addition, activation of MMP-9 and subsequent release of soluble Kit ligand (sKitL) and mobilization of proangiogenic cells are impaired in eNOS−/− mice, suggesting a role for the eNOS-MMP9-sKitL pathway in BM EPC mobilization (5). Note that H2O2 causes endothelial NO release by phosphorylating eNOS serine 1179 (28) and that antioxidant and NADPH oxidase inhibitor attenuates eNOS activity in EC (161). In line with this, exercise training, known to increase both endothelial shear stress and H2O2, increases eNOS expression (121). Thus, NADPH oxidase–derived ROS may regulate eNOS-MMP-9 signaling, thereby promoting BM stem/progenitor cell mobilization (Fig. 3).

NAPDH oxidase and homing of stem/progenitor cells

BM-derived stem/progenitor cells homing and angiogenic capacity play in important role in postnatal neovascularization by directly incorporating into endothelial lining, structurally supporting the neovasculature, or secreting proangiogenic factor in ischemic tissues (96, 109, 134) (Figs. 1 and 3). This homing is mediated through highly concerted mechanisms, which involve adhesion, chemotaxis, and invasion, followed by integration of cells into vascular structures (38). Understanding the molecular mechanisms of homing is essential for enhancing cell-based therapy. Recent studies suggest that adhesion molecules on the endothelial surface plays an important role in the homing of progenitor/stem cells to sites of neovascularization, as demonstrated for leukocytes recruitment to the sites of inflammation. The homing capacity of EPCs and stem/progenitor cells is mediated by various adhesion molecules, including E-selectin, ICAM-1 and VCAM-1 (38), P-selectin (59), β1 integrin (Duan et al., 2006), and β2 integrin (37) (Fig. 3). NADPH oxidase–derived ROS are involved in expression of E-selectin in diabetes (80, 240) during ischemia/reperfusion (156, 172) and cytokine stimulation (35, 71, 156), as well as expression of ICAM-1, VCAM-1 (142), and P-selectin (7, 95, 193) on the endothelial surface. A recent study shows that stimulation of Rap1 through cAMP-mediated Epac1 activation increases integrin activity and integrin-dependent homing of progenitor cells, thereby enhancing their therapeutic potential in vivo (33). Given that Rap1A is involved in regulating NADPH oxidase (25, 129), NADPH oxidase might be involved in integrin-dependent homing functions of progenitor cells. β2 integrin and PI3Kγ are involved in homing capacity of EPCs and BM progenitor cells by regulating adhesion of EPCs on fibronectin and ICAM-1 (38). However, adhesion of BM cells to fibronectin and ICAM-1 is not affected in Nox2−/− cells (205), suggesting that β2 integrin– and PI3Kγ-mediated EPC adhesion capacity is not dependent on Nox2-derived ROS. Whether other Noxs are involved in the adhesion capacity of BM stem/progenitor cells remains unknown.

Intravenous infusion of Nox2−/− BM cells in a mouse hindlimb ischemia model shows that Nox2-derived ROS are required for the homing and angiogenic capacity of BM cells, thereby promoting neovascularization of ischemic tissues in vivo (205) (Fig. 1). Short-term pretreatment with low-dose H2O2 enhances the efficacy of BM cells for neovascularization in a mouse hindlimb ischemia model (112). Similarly, pretreatment with H2O2 rescues the impaired homing and migration capacity of Nox2-deficient BM cells (205), suggesting an important role of ROS in stem/progenitor cell function. Mechanistically, Nox2-derived ROS are selectively involved in actin polymerization as well as the pathways linking SDF-1/CXCR4 to Akt, thereby promoting migration of stem/progenitor cells (205) (Fig. 3). Similarly, receptor tyrosine kinase activation by hematopoietic cytokines increases generation of ROS that are involved in BM stem cells proliferation (87). IL-8 is an inflammatory chemokine that is able to stimulate angiogenesis (149) and provides a chemoattractant gradient for BM-derived endothelial progenitors or angioblasts. This chemokine-mediated homing of BM progenitors to the ischemic heart regulates their ability to induce myocardial neovascularization, protection against apoptosis, and functional cardiac recovery (107). IL-8 stimulation via CXCR2 causes NADPH oxidase activation by Rac translocation to membrane (242), and blocking CXCR2 inhibits the incorporation of human EPCs expressing CXCR2 at sites of arterial injury (83). Thus, IL-8/CXCR2-mediated activation of NADPH oxidase may be involved in EPCs and BM progenitor/stem cell homing, thereby promoting angiogenesis and repair in response to injury (Fig. 3).

NADPH oxidase and dysfunction of stem/progenitor cells in pathophysiologies

Although physiologic levels for ROS are required for normal function, excess amounts of ROS produced in various pathophysiologies contribute to dysfunction of EPC and stem/progenitor cells (20, 81, 132, 181, 215). Evidence suggests that number and functional capacity of EPCs are reduced in cardiovascular diseases, including hypertension, atherosclerosis, diabetes, coronary artery disease, and heart failure, as well as comorbid risk factors such as aging, hypercholesterolemia, and cigarette smoking (23, 34, 181, 203, 220, 233), all of which are associated with oxidative stress.

The renin–angiotensin system increases vascular oxidative stress, leading to reduction in the bioavailability of NO, thereby contributing to hypertension, diabetes, atherosclerosis, and heart failure. Ang II accelerates the senescence of EPCs by increasing oxidative stress via upregulation of Nox2, which is mediated through AT1Rs (91). Hypercholesterolemia and in vitro exposure to LDLs are associated with increased AT1R expression in CD34+ cell, and enhance AT1R-mediated HSC differentiation into proatherogenic CD11b+ monocytes (191). Impaired BM stem/progenitor cell function and postischemic revascularization in spontaneously hypertensive rats (SHRs) are rescued by cotreatment with Ang II converting enzyme (ACE) inhibitor, AT1Rs blocker, or antioxidants (238). Moreover, AT1Rs blockers or ACE inhibitors improve EPC dysfunction through antioxidative mechanisms in hypertension (239). Thus, AT1R signaling is involved in stem/progenitor cell dysfunctions leading to hypertensive vascular injury. In line with these findings, antioxidative β1-adrenoceptor blocker celiprolol decreases oxidative stress in hypertension and improves EPC numbers and function (232). Aldosterone, which increases oxidative stress, inhibits the formation of EPCs, at least in part by attenuating VEGFR2 expression and subsequent Akt signaling (136).

By contrast, Ang II at low concentrations functions as a positive regulator for EPC and stem/progenitor function. Ang II potentiates VEGF-induced human EPCs proliferation and network formation through the upregulation of VEGFR2 (92). It also promotes NO production, inhibits apoptosis and enhances adhesion potential of BM-derived EPCs via the PI3-kinase/Akt pathway (236). Ang II stimulates proliferation of hematopoietic progenitor cells as well as differentiation of dendritic cells (150). It also promotes hematopoietic recovery in multiple cellular lineages after chemotherapy through an increase in the number of early hematopoietic progenitors (169). In two-kidney, one-clip or Ang II–infused hypertensive mice, hypertension-induced mechanical stretch of the vessel wall stimulates the mobilization of EPCs from BM, in a p47phox-dependent manner (173). This response seems to be a compensatory vascular adaptation.

Increased oxidative stress plays a role in EPC dysfunction in type 1 and type 2 diabetes (133, 179). In type 1 diabetic mice, overproduction of Nox2-derived ROS contributes to impaired angiogenesis and EPC dysfunction, which is rescued in Nox2-knockout mice (53). Uncoupled eNOS in diabetic BM, glucose-treated EPCs, and EPCs from diabetes patients produce overproduction of O2•−, which results in impaired EPC mobilization and function (200). Human EPCs from type 2 diabetes, which are associated with oxidative stress, exhibit the mobilization of functionally defective EPCs with impaired proliferation, adhesion, and incorporation into existing vasculature (197). BM cells obtained from diabetic db/db mice are dysfunctional because of oxidative stress–mediated apoptosis, which is restored by the antioxidant ebselen (39). Diabetes mellitus impairs CD133+ progenitor cell function after myocardial infarction because of a lower resistance of CD133+ cells to oxidative stress (217). This explains the delayed postischemic vascular healing and myocardial recovery in diabetes patients. Diabetes patients show an increase in NADPH oxidase expression in circulating lymphomonocytes, membrane-associated PKCβ2 activity in monocytes, and a decrease in number of EPCs (16). The in vivo reendothelialization capacity of EPCs from patients with type 2 diabetes is impaired because of increased NADPH oxidase–dependent O2•− production and subsequent reduced NO bioavailability (187). AT1R antagonists increase the number of regenerative EPCs in patients with type 2 diabetes mellitus (19), presumably through inhibiting NADPH oxidase.

In myocardial infarction model of rats, ROS elevation as well as reduced ERK phosphorylation and MMP-9 activity in BM contribute to impaired EPC mobilization, which is rescued by ACE or HMG-CoA inhibitors (199). In patients with coronary artery disease, peroxisome proliferator–activated receptor-γ (PPAR gamma) agonists, which are used for the treatment of diabetes, increase EPC number and migration by inhibiting NADPH oxidase activity (222). A study using p47phox knockout mice and BM transplantation demonstrated that NADPH oxidase–derived ROS in BM contribute to lung ischemia/reperfusion injury (231). Oxidized LDL plays a key role in the pathogenesis of atherosclerosis by accelerating the onset of EPC senescence (93). In diabetes, oxidized LDL contributes to EPC dysfunction via a p53-mediated signaling pathway (170). By contrast, high-density lipoprotein, which is atheroprotective by antioxidant and antiinflammatory properties, promotes progenitor cell–mediated endothelial repair in apoE−/− mice (204). Homocysteine, which increases excess production of ROS, accelerates senescence and reduces proliferation of EPCs (243).

The availability and function of EPCs are also affected by other risk factors including cigarette smoking (140) and aging (78), which are associated with oxidative stress. ROS and the natural by-products of oxidative metabolism are known to cause DNA damage closely associated with aging. ATM (ataxia telangiectasia-mutated) is important for stem cell function in regulating ROS levels. Hematopoietic stem cells from ATM-deficient mice show enhanced ROS levels and impaired stem cell functions, rescued by the treatment with the antioxidant N-acetyl-l-cysteine (97). In addition, hematopoietic stem cells from mice lacking Foxo transcription factors show elevated ROS levels and impaired long-term repopulation activity, which is associated with increased apoptosis and reduced stem/progenitor cell reserves (202). Foxo3a deficiency alone is sufficient to elevate cellular ROS, thereby disrupting cell quiescence and reducing the size of the stem cell compartment (144). However, the role of NADPH oxidase for stem cell aging has not been demonstrated, but it is a subject for future study.

Understanding the molecular mechanisms by which oxidative stress is involved in reducing the number and functional activity of EPCs is potentially important, as it could create novel therapeutic strategies for treatment of cardiovascular diseases (203, 233). Inhibition of NADPH oxidase of progenitor cells may lead to improvement of endothelial repair and ischemia-induced neovascularization in various oxidative stress–dependent pathophysiologies (23).

Conclusion and Future Directions

NADPH oxidase–derived ROS play an important role in physiologic and pathologic postnatal angiogenesis and vasculogenesis. The effects of ROS derived from ECs and stem/progenitor cells during neovascularization are tightly regulated by the balance of prooxidant and antioxidant enzyme activity. ROS at low levels function as signaling molecules to mediate cell proliferation, migration, differentiation, survival, and angiogenic gene expression in ECs and stem/progenitor cells, which contributes to reparative neovascularization in response to injury. Moreover, NADPH oxidase expression is increased in BM after ischemic injury, which is required for BM stem/progenitor mobilization, homing, and angiogenic capacity at the site of new-vessel formation. Recent studies show the crucial role of NADPH oxidase in cardiovascular differentiation of ES cells. By contrast, excess amounts of ROS (oxidative stress) involved in various pathologies including hypertension, atherosclerosis, aging, and diabetes contribute to dysfunction of ECs and stem/progenitor cells, thereby resulting in impaired angiogenesis and vasculogenesis. Cultured EPCs express higher levels of antioxidant enzymes MnSOD and catalase than do mature ECs, thereby being resistant to oxidative stress. Significant work remains to be performed (a) to define the role of each Nox and its regulatory subunits in angiogenesis and vasculogenesis; (b) to determine the activation mechanisms of NADPH oxidase by various angiogenesis factors and cytokines in ECs and stem/progenitor cells; (c) to identify molecular targets of NADPH oxidase–derived ROS in signaling pathways involved in angiogenesis in ECs, as well as stem/progenitor cell function; and (d) to determine the contribution of ROS derived from other cell types such as inflammatory cells, platelets, and T cells in postnatal neovascularization in vivo. Understanding these mechanisms will lead to development of new therapeutic strategies for many angiogenesis- and vasculogenesis-dependent cardiovascular diseases such as ischemic heart and limb diseases and hypertension, diabetes, and atherosclerosis.

Abbreviations Used

ACE

angiotensin-converting enzyme

Ang II

angiotensin II

AP1

activator protein 1

ATM

ataxia telangiectasia–mutated

AT1R

angiotensin II type1 receptor

BM

bone marrow

cAMP

cyclic adenosine monophosphate

CT-1

cardiotrophin-1

EC

endothelial cell

EGF

epidermal growth factor

eNOS

endothelial nitric oxide synthase

EPC

endothelial progenitor cell

ERK

extracellular signal-regulated kinase

ES cell

embryonic stem cell

G-CSF

granulocyte colony-stimulating factor

HIF1

hypoxia-inducible factor 1

HMG-CoA

3-hydroxy-3-methylglutaryl-coenzyme A

H2O2

hydrogen peroxide

HPC

hematopoietic progenitor cell

HSCs

hematopoietic stem cells

ICAM-1

intercellular adhesion molecule 1

IQGAP1

IQ motif containing GTPase-activating protein 1

IL-8

interleukin 8

JNK

c-Jun N-terminal kinases

MAPKs

mitogen-activated protein kinases

MMP

matrix metalloproteinase

MnSOD

manganese superoxide dismutase

NADPH

nicotinamide adenine dinucleotide phosphate

NF-κB

nuclear factor κB

NO

nitric oxide

NOx

NADPH oxidase

O2•−

superoxide anion

PI3K

phosphoinositide 3-kinase

PKC

protein kinase C

PTEN

phosphatase and tensin homologue

PTP

protein tyrosine phosphatase

ROS

reactive oxygen species

SCF

stem cell factor

SDF-1

stromal cell–derived factor 1

SHP

Src homology phosphatase

sKitL

soluble Kit ligand

SOD

superoxide dismutase

VCAM-1

vascular cell adhesion molecule 1

VE-cadherin

vascular endothelial cadherin

VEGF

vascular endothelial growth factor

VEGFR2

VEGF receptor type 2

Acknowledgments

This work was supported by NIH R01 HL077524, AHA Grant-In-Aid 0555308B, and AHA National Innovative Research grant 0970336N.

References

  • 1.Abe R. Shimizu T. Yamagishi S. Shibaki A. Amano S. Inagaki Y. Watanabe H. Sugawara H. Nakamura H. Takeuchi M. Imaizumi T. Shimizu H. Overexpression of pigment epithelium-derived factor decreases angiogenesis and inhibits the growth of human malignant melanoma cells in vivo. Am J Pathol. 2004;164:1225–1232. doi: 10.1016/s0002-9440(10)63210-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Abid MR. Kachra Z. Spokes KC. Aird WC. NADPH oxidase activity is required for endothelial cell proliferation and migration. FEBS Lett. 2000;486:252–256. doi: 10.1016/s0014-5793(00)02305-x. [DOI] [PubMed] [Google Scholar]
  • 3.Abid MR. Spokes KC. Shih SC. Aird WC. NADPH oxidase activity selectively modulates vascular endothelial growth factor signaling pathways. J Biol Chem. 2007;282:35373–35385. doi: 10.1074/jbc.M702175200. [DOI] [PubMed] [Google Scholar]
  • 4.Abid MR. Tsai JC. Spokes KC. Deshpande SS. Irani K. Aird WC. Vascular endothelial growth factor induces manganese-superoxide dismutase expression in endothelial cells by a Rac1-regulated NADPH oxidase-dependent mechanism. FASEB J. 2001;15:2548–2550. doi: 10.1096/fj.01-0338fje. [DOI] [PubMed] [Google Scholar]
  • 5.Aicher A. Heeschen C. Mildner-Rihm C. Urbich C. Ihling C. Technau-Ihling K. Zeiher AM. Dimmeler S. Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med. 2003;9:1370–1376. doi: 10.1038/nm948. [DOI] [PubMed] [Google Scholar]
  • 6.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]
  • 7.Akgur FM. Brown MF. Zibari GB. McDonald JC. Epstein CJ. Ross CR. Granger DN. Role of superoxide in hemorrhagic shock-induced P-selectin expression. Am J Physiol. 2000;279:H791–H797. doi: 10.1152/ajpheart.2000.279.2.H791. [DOI] [PubMed] [Google Scholar]
  • 8.Al-Shabrawey M. Bartoli M. El-Remessy AB. Platt DH. Matragoon S. Behzadian MA. Caldwell RW. Caldwell RB. Inhibition of NAD(P)H oxidase activity blocks vascular endothelial growth factor overexpression and neovascularization during ischemic retinopathy. Am J Pathol. 2005;167:599–607. doi: 10.1016/S0002-9440(10)63001-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Anilkumar N. Weber R. Zhang M. Brewer A. Shah AM. Nox4 and nox2 NADPH oxidases mediate distinct cellular redox signaling responses to agonist stimulation. Arterioscler Thromb Vasc Biol. 2008;28:1347–1354. doi: 10.1161/ATVBAHA.108.164277. [DOI] [PubMed] [Google Scholar]
  • 10.Arany Z. He H. Lin J. Hoyer K. Handschin C. Toka O. Ahmad F. Matsui T. Chin S. Wu PH. Rybkin II. Shelton JM. Manieri M. Cinti S. Schoen FJ. Bassel-Duby R. Rosenzweig A. Ingwall JS. Spiegelman BM. Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab. 2005;1:259–271. doi: 10.1016/j.cmet.2005.03.002. [DOI] [PubMed] [Google Scholar]
  • 11.Arbiser JL. Petros J. Klafter R. Govindajaran B. McLaughlin ER. Brown LF. Cohen C. Moses M. Kilroy S. Arnold RS. Lambeth JD. Reactive oxygen generated by Nox1 triggers the angiogenic switch. Proc Natl Acad Sci U S A. 2002;99:715–720. doi: 10.1073/pnas.022630199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Arden KC. FoxOs in tumor suppression and stem cell maintenance. Cell. 2007;128:235–237. doi: 10.1016/j.cell.2007.01.009. [DOI] [PubMed] [Google Scholar]
  • 13.Asahara T. Takahashi T. Masuda H. Kalka C. Chen D. Iwaguro H. Inai Y. Silver M. Isner JM. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999;18:3964–3972. doi: 10.1093/emboj/18.14.3964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ateghang B. Wartenberg M. Gassmann M. Sauer H. Regulation of cardiotrophin-1 expression in mouse embryonic stem cells by HIF-1alpha and intracellular reactive oxygen species. J Cell Sci. 2006;119:1043–1052. doi: 10.1242/jcs.02798. [DOI] [PubMed] [Google Scholar]
  • 15.Avecilla ST. Hattori K. Heissig B. Tejada R. Liao F. Shido K. Jin DK. Dias S. Zhang F. Hartman TE. Hackett NR. Crystal RG. Witte L. Hicklin DJ. Bohlen P. Eaton D. Lyden D. de Sauvage F. Rafii S. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med. 2004;10:64–71. doi: 10.1038/nm973. [DOI] [PubMed] [Google Scholar]
  • 16.Avogaro A. Fadini GP. Gallo A. Pagnin E. de Kreutzenberg S. Endothelial dysfunction in type 2 diabetes mellitus. Nutr Metab Cardiovasc Dis. 2006;16(suppl 1):S39–S45. doi: 10.1016/j.numecd.2005.10.015. [DOI] [PubMed] [Google Scholar]
  • 17.Babior BM. NADPH oxidase: an update. Blood. 1999;93:1464–1476. [PubMed] [Google Scholar]
  • 18.Baehner RL. Millar-Groff S. Bringas P. Developmental expression of NADPH phagocytic oxidase components in mouse embryos. Pediatr Res. 1999;46:152–157. doi: 10.1203/00006450-199908000-00004. [DOI] [PubMed] [Google Scholar]
  • 19.Bahlmann FH. de Groot K. Mueller O. Hertel B. Haller H. Fliser D. Stimulation of endothelial progenitor cells: a new putative therapeutic effect of angiotensin II receptor antagonists. Hypertension. 2005;45:526–529. doi: 10.1161/01.HYP.0000159191.98140.89. [DOI] [PubMed] [Google Scholar]
  • 20.Ballard VL. Edelberg JM. Stem cells and the regeneration of the aging cardiovascular system. Circ Res. 2007;100:1116–1127. doi: 10.1161/01.RES.0000261964.19115.e3. [DOI] [PubMed] [Google Scholar]
  • 21.BelAiba RS. Djordjevic T. Petry A. Diemer K. Bonello S. Banfi B. Hess J. Pogrebniak A. Bickel C. Gorlach A. NOX5 variants are functionally active in endothelial cells. Free Radic Biol Med. 2007;42:446–459. doi: 10.1016/j.freeradbiomed.2006.10.054. [DOI] [PubMed] [Google Scholar]
  • 22.Bell EL. Chandel NS. Mitochondrial oxygen sensing: regulation of hypoxia-inducible factor by mitochondrial generated reactive oxygen species. Essays Biochem. 2007;43:17–27. doi: 10.1042/BSE0430017. [DOI] [PubMed] [Google Scholar]
  • 23.Besler C. Doerries C. Giannotti G. Luscher TF. Landmesser U. Pharmacological approaches to improve endothelial repair mechanisms. Expert Rev Cardiovasc Ther. 2008;6:1071–1082. doi: 10.1586/14779072.6.8.1071. [DOI] [PubMed] [Google Scholar]
  • 24.Bhattacharya R. Kwon J. Wang E. Mukherjee P. Mukhopadhyay D. Src homology 2 (SH2) domain containing protein tyrosine phosphatase-1 (SHP-1) dephosphorylates VEGF receptor-2 and attenuates endothelial DNA synthesis, but not migration. J Mol Signal. 2008;3:8. doi: 10.1186/1750-2187-3-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bokoch GM. Quilliam LA. Bohl BP. Jesaitis AJ. Quinn MT. Inhibition of Rap1A binding to cytochrome b558 of NADPH oxidase by phosphorylation of Rap1A. Science (New York) 1991;254:1794–1796. doi: 10.1126/science.1763330. [DOI] [PubMed] [Google Scholar]
  • 26.Briggs MW. Sacks DB. IQGAP proteins are integral components of cytoskeletal regulation. EMBO Rep. 2003;4:571–574. doi: 10.1038/sj.embor.embor867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.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]
  • 28.Cai H. Li Z. Davis ME. Kanner W. Harrison DG. Dudley SC., Jr. Akt-dependent phosphorylation of serine 1179 and mitogen-activated protein kinase kinase/extracellular signal-regulated kinase 1/2 cooperatively mediate activation of the endothelial nitric-oxide synthase by hydrogen peroxide. Mol Pharmacol. 2003;63:325–331. doi: 10.1124/mol.63.2.325. [DOI] [PubMed] [Google Scholar]
  • 29.Cai T. Fassina G. Morini M. Aluigi MG. Masiello L. Fontanini G. D'Agostini F. De Flora S. Noonan DM. Albini A. N-acetylcysteine inhibits endothelial cell invasion and angiogenesis. Lab Invest. 1999;79:1151–1159. [PubMed] [Google Scholar]
  • 30.Caldwell RB. Bartoli M. Behzadian MA. El-Remessy AE. Al-Shabrawey M. Platt DH. Liou GI. Caldwell RW. Vascular endothelial growth factor and diabetic retinopathy: role of oxidative stress. Curr Drug Targets. 2005;6:511–524. doi: 10.2174/1389450054021981. [DOI] [PubMed] [Google Scholar]
  • 31.Calvi LM. Adams GB. Weibrecht KW. Weber JM. Olson DP. Knight MC. Martin RP. Schipani E. Divieti P. Bringhurst FR. Milner LA. Kronenberg HM. Scadden DT. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–846. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  • 32.Carcamo JM. Borquez-Ojeda O. Golde DW. Vitamin C inhibits granulocyte macrophage-colony-stimulating factor-induced signaling pathways. Blood. 2002;99:3205–3212. doi: 10.1182/blood.v99.9.3205. [DOI] [PubMed] [Google Scholar]
  • 33.Carmona G. Chavakis E. Koehl U. Zeiher AM. Dimmeler S. Activation of Epac stimulates integrin-dependent homing of progenitor cells. Blood. 2008;111:2640–2646. doi: 10.1182/blood-2007-04-086231. [DOI] [PubMed] [Google Scholar]
  • 34.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]
  • 35.Cayatte AJ. Rupin A. Oliver-Krasinski J. Maitland K. Sansilvestri-Morel P. Boussard MF. Wierzbicki M. Verbeuren TJ. Cohen RA. S17834, a new inhibitor of cell adhesion and atherosclerosis that targets NADPH oxidase. Arterioscler Thromb Vasc Biol. 2001;21:1577–1584. doi: 10.1161/hq1001.096723. [DOI] [PubMed] [Google Scholar]
  • 36.Chang Y. Ceacareanu B. Zhuang D. Zhang C. Pu Q. Ceacareanu AC. Hassid A. Counter-regulatory function of protein tyrosine phosphatase 1B in platelet-derived growth factor- or fibroblast growth factor-induced motility and proliferation of cultured smooth muscle cells and in neointima formation. Arterioscler Thromb Vasc Biol. 2006;26:501–507. doi: 10.1161/01.ATV.0000201070.71787.b8. [DOI] [PubMed] [Google Scholar]
  • 37.Chavakis E. Aicher A. Heeschen C. Sasaki K. Kaiser R. El Makhfi N. Urbich C. Peters T. Scharffetter-Kochanek K. Zeiher AM. Chavakis T. Dimmeler S. Role of beta2-integrins for homing and neovascularization capacity of endothelial progenitor cells. J Exp Med. 2005;201:63–72. doi: 10.1084/jem.20041402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chavakis E. Urbich C. Dimmeler S. Homing and engraftment of progenitor cells: a prerequisite for cell therapy. J Mol Cell Cardiol. 2008;48:514–522. doi: 10.1016/j.yjmcc.2008.01.004. [DOI] [PubMed] [Google Scholar]
  • 39.Chen J. Park HC. Patschan S. Brodsky SV. Gealikman O. Kuo MC. Li H. Addabbo F. Zhang F. Nasjletti A. Gross SS. Goligorsky MS. Premature vascular senescence in metabolic syndrome: could it be prevented and reversed by a selenorganic antioxidant and peroxynitrite scavenger Ebselen? Drug Disc Today. 2007;4:93–99. doi: 10.1016/j.ddstr.2007.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chen JX. Zeng H. Lawrence ML. Blackwell TS. Meyrick B. Angiopoietin-1-induced angiogenesis is modulated by endothelial NADPH oxidase. Am J Physiol. 2006;291:H1563–H1572. doi: 10.1152/ajpheart.01081.2005. [DOI] [PubMed] [Google Scholar]
  • 41.Chen JX. Zeng H. Tuo QH. Yu H. Meyrick B. Aschner JL. NADPH oxidase modulates myocardial Akt, ERK1/2 activation, and angiogenesis after hypoxia-reoxygenation. Am J Physiol. 2007;292:H1664–H1674. doi: 10.1152/ajpheart.01138.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chen W. Gabel S. Steenbergen C. Murphy E. A redox-based mechanism for cardioprotection induced by ischemic preconditioning in perfused rat heart. Circ Res. 1995;77:424–429. doi: 10.1161/01.res.77.2.424. [DOI] [PubMed] [Google Scholar]
  • 43.Chiarugi P. Cirri P. Redox regulation of protein tyrosine phosphatases during receptor tyrosine kinase signal transduction. Trends Biochem Sci. 2003;28:509–514. doi: 10.1016/S0968-0004(03)00174-9. [DOI] [PubMed] [Google Scholar]
  • 44.Chua CC. Hamdy RC. Chua BH. Upregulation of vascular endothelial growth factor by H2O2 in rat heart endothelial cells. Free Radic Biol Med. 1998;25:891–897. doi: 10.1016/s0891-5849(98)00115-4. [DOI] [PubMed] [Google Scholar]
  • 45.Colavitti R. Pani G. Bedogni B. Anzevino R. Borrello S. Waltenberger J. Galeotti T. Reactive oxygen species as downstream mediators of angiogenic signaling by vascular endothelial growth factor receptor-2/KDR. J Biol Chem. 2002;277:3101–3108. doi: 10.1074/jbc.M107711200. [DOI] [PubMed] [Google Scholar]
  • 46.Connor KM. Subbaram S. Regan KJ. Nelson KK. Mazurkiewicz JE. Bartholomew PJ. Aplin AE. Tai YT. Aguirre-Ghiso J. Flores SC. Melendez JA. Mitochondrial H2O2 regulates the angiogenic phenotype via PTEN oxidation. J Biol Chem. 2005;280:16916–16924. doi: 10.1074/jbc.M410690200. [DOI] [PubMed] [Google Scholar]
  • 47.Cronin A. Mowbray S. Durk H. Homburg S. Fleming I. Fisslthaler B. Oesch F. Arand M. The N-terminal domain of mammalian soluble epoxide hydrolase is a phosphatase. Proc Natl Acad Sci U S A. 2003;100:1552–1557. doi: 10.1073/pnas.0437829100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Das DK. Maulik N. Cardiac genomic response following preconditioning stimulus. Cardiovasc Res. 2006;70:254–263. doi: 10.1016/j.cardiores.2006.02.023. [DOI] [PubMed] [Google Scholar]
  • 49.Datla SR. Peshavariya H. Dusting GJ. Jiang F. Important role of Nox4 type NADPH oxidase in angiogenic responses in human microvascular endothelial cells in vitro. Arterioscler Thromb Vasc Biol. 2007;27:2319–2324. doi: 10.1161/ATVBAHA.107.149450. [DOI] [PubMed] [Google Scholar]
  • 50.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]
  • 51.Deudero JJ. Caramelo C. Castellanos MC. Neria F. Fernandez-Sanchez R. Calabia O. Penate S. Gonzalez-Pacheco FR. Induction of hypoxia-inducible factor 1alpha gene expression by vascular endothelial growth factor. J Biol Chem. 2008;283:11435–11444. doi: 10.1074/jbc.M703875200. [DOI] [PubMed] [Google Scholar]
  • 52.Ding L. Liang XG. Hu Y. Zhu DY. Lou YJ. Involvement of p38MAPK and reactive oxygen species in icariin-induced cardiomyocyte differentiation of murine embryonic stem cells in vitro. Stem Cells Dev. 2008;17:751–760. doi: 10.1089/scd.2007.0206. [DOI] [PubMed] [Google Scholar]
  • 53.Ebrahimian TG. Heymes C. You D. Blanc-Brude O. Mees B. Waeckel L. Duriez M. Vilar J. Brandes RP. Levy BI. Shah AM. Silvestre JS. NADPH oxidase-derived overproduction of reactive oxygen species impairs postischemic neovascularization in mice with type 1 diabetes. Am J Pathol. 2006;169:719–728. doi: 10.2353/ajpath.2006.060042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.El-Remessy AB. Al-Shabrawey M. Platt DH. Bartoli M. Behzadian MA. Ghaly N. Tsai N. Motamed K. Caldwell RB. Peroxynitrite mediates VEGF's angiogenic signal and function via a nitration-independent mechanism in endothelial cells. FASEB J. 2007;21:2528–2539. doi: 10.1096/fj.06-7854com. [DOI] [PubMed] [Google Scholar]
  • 55.Ellis EA. Grant MB. Murray FT. Wachowski MB. Guberski DL. Kubilis PS. Lutty GA. Increased NADH oxidase activity in the retina of the BBZ/Wor diabetic rat. Free Radic Biol Med. 1998;24:111–120. doi: 10.1016/s0891-5849(97)00202-5. [DOI] [PubMed] [Google Scholar]
  • 56.Ellis EA. Guberski DL. Somogyi-Mann M. Grant MB. Increased H2O2, vascular endothelial growth factor and receptors in the retina of the BBZ/Wor diabetic rat. Free Radic Biol Med. 2000;28:91–101. doi: 10.1016/s0891-5849(99)00216-6. [DOI] [PubMed] [Google Scholar]
  • 57.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]
  • 58.Finkel T. Oxidant signals and oxidative stress. Curr Opin Cell Biol. 2003;15:247–254. doi: 10.1016/s0955-0674(03)00002-4. [DOI] [PubMed] [Google Scholar]
  • 59.Foubert P. Silvestre JS. Souttou B. Barateau V. Martin C. Ebrahimian TG. Lere-Dean C. Contreres JO. Sulpice E. Levy BI. Plouet J. Tobelem G. Le Ricousse-Roussanne S. PSGL-1-mediated activation of EphB4 increases the proangiogenic potential of endothelial progenitor cells. J Clin Invest. 2007;117:1527–1537. doi: 10.1172/JCI28338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.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]
  • 61.Frey RS. Rahman A. Kefer JC. Minshall RD. Malik AB. PKCzeta regulates TNF-alpha-induced activation of NADPH oxidase in endothelial cells. Circ Res. 2002;90:1012–1019. doi: 10.1161/01.res.0000017631.28815.8e. [DOI] [PubMed] [Google Scholar]
  • 62.Furst R. Brueckl C. Kuebler WM. Zahler S. Krotz F. Gorlach A. Vollmar AM. Kiemer AK. Atrial natriuretic peptide induces mitogen-activated protein kinase phosphatase-1 in human endothelial cells via Rac1 and NAD(P)H oxidase/Nox2-activation. Circ Res. 2005;96:43–53. doi: 10.1161/01.RES.0000151983.01148.06. [DOI] [PubMed] [Google Scholar]
  • 63.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]
  • 64.Geiszt M. NADPH oxidases: new kids on the block. Cardiovasc Res. 2006;71:289–299. doi: 10.1016/j.cardiores.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 65.Geiszt M. Kopp JB. Varnai P. Leto TL. Identification of renox, an NAD(P)H oxidase in kidney. Proc Natl Acad Sci U S A. 2000;97:8010–8014. doi: 10.1073/pnas.130135897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gonzalez-Pacheco FR. Deudero JJ. Castellanos MC. Castilla MA. Alvarez-Arroyo MV. Yague S. Caramelo C. Mechanisms of endothelial response to oxidative aggression: protective role of autologous VEGF and induction of VEGFR2 by H2O2. Am J Physiol. 2006;291:H1395–H1401. doi: 10.1152/ajpheart.01277.2005. [DOI] [PubMed] [Google Scholar]
  • 67.Gorlach A. Brandes RP. Nguyen K. Amidi M. Dehghani F. Busse R. A gp91phox containing NADPH oxidase selectively expressed in endothelial cells is a major source of oxygen radical generation in the arterial wall. Circ Res. 2000;87:26–32. doi: 10.1161/01.res.87.1.26. [DOI] [PubMed] [Google Scholar]
  • 68.Goto H. Nishikawa T. Sonoda K. Kondo T. Kukidome D. Fujisawa K. Yamashiro T. Motoshima H. Matsumura T. Tsuruzoe K. Araki E. Endothelial MnSOD overexpression prevents retinal VEGF expression in diabetic mice. Biochem Biophys Res Commun. 2008;366:814–820. doi: 10.1016/j.bbrc.2007.12.041. [DOI] [PubMed] [Google Scholar]
  • 69.Grazia Lampugnani M. Zanetti A. Corada M. Takahashi T. Balconi G. Breviario F. Orsenigo F. Cattelino A. Kemler R. Daniel TO. Dejana E. Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, beta-catenin, and the phosphatase DEP-1/CD148. J Cell Biol. 2003;161:793–804. doi: 10.1083/jcb.200209019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.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]
  • 71.Gu W. Pagel PS. Warltier DC. Kersten JR. Modifying cardiovascular risk in diabetes mellitus. Anesthesiology. 2003;98:774–779. doi: 10.1097/00000542-200303000-00029. [DOI] [PubMed] [Google Scholar]
  • 72.Gu W. Weihrauch D. Tanaka K. Tessmer JP. Pagel PS. Kersten JR. Chilian WM. Warltier DC. Reactive oxygen species are critical mediators of coronary collateral development in a canine model. Am J Physiol. 2003;285:H1582–H1589. doi: 10.1152/ajpheart.00318.2003. [DOI] [PubMed] [Google Scholar]
  • 73.Guo AM. Arbab AS. Falck JR. Chen P. Edwards PA. Roman RJ. Scicli AG. Activation of vascular endothelial growth factor through reactive oxygen species mediates 20-hydroxyeicosatetraenoic acid-induced endothelial cell proliferation. J Pharmacol Exp Ther. 2007;321:18–27. doi: 10.1124/jpet.106.115360. [DOI] [PubMed] [Google Scholar]
  • 74.Guo DQ. Wu LW. Dunbar JD. Ozes ON. Mayo LD. Kessler KM. Gustin JA. Baerwald MR. Jaffe EA. Warren RS. Donner DB. Tumor necrosis factor employs a protein-tyrosine phosphatase to inhibit activation of KDR and vascular endothelial cell growth factor-induced endothelial cell proliferation. J Biol Chem. 2000;275:11216–11221. doi: 10.1074/jbc.275.15.11216. [DOI] [PubMed] [Google Scholar]
  • 75.Gupta R. Karpatkin S. Basch RS. Hematopoiesis and stem cell renewal in long-term bone marrow cultures containing catalase. Blood. 2006;107:1837–1846. doi: 10.1182/blood-2005-03-1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Haneline LS. Redox regulation of stem and progenitor cells. Antioxid Redox Signal. 2008;10:1849–1852. doi: 10.1089/ars.2008.2141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Harfouche R. Malak NA. Brandes RP. Karsan A. Irani K. Hussain SN. Roles of reactive oxygen species in angiopoietin-1/tie-2 receptor signaling. FASEB J. 2005;19:1728–1730. doi: 10.1096/fj.04-3621fje. [DOI] [PubMed] [Google Scholar]
  • 78.Heiss C. Keymel S. Niesler U. Ziemann J. Kelm M. Kalka C. Impaired progenitor cell activity in age-related endothelial dysfunction. J Am Coll Cardiol. 2005;45:1441–1448. doi: 10.1016/j.jacc.2004.12.074. [DOI] [PubMed] [Google Scholar]
  • 79.Heissig B. Hattori K. Dias S. Friedrich M. Ferris B. Hackett NR. Crystal RG. Besmer P. Lyden D. Moore MA. Werb Z. Rafii S. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625–637. doi: 10.1016/s0092-8674(02)00754-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Higai K. Shimamura A. Matsumoto K. Amadori-modified glycated albumin predominantly induces E-selectin expression on human umbilical vein endothelial cells through NADPH oxidase activation. Clin Chim Acta. 2006;367:137–143. doi: 10.1016/j.cca.2005.12.008. [DOI] [PubMed] [Google Scholar]
  • 81.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]
  • 82.Hosokawa K. Arai F. Yoshihara H. Nakamura Y. Gomei Y. Iwasaki H. Miyamoto K. Shima H. Ito K. Suda T. Function of oxidative stress in the regulation of hematopoietic stem cell-niche interaction. Biochem Biophys Res Commun. 2007;363:578–583. doi: 10.1016/j.bbrc.2007.09.014. [DOI] [PubMed] [Google Scholar]
  • 83.Hristov M. Zernecke A. Bidzhekov K. Liehn EA. Shagdarsuren E. Ludwig A. Weber C. Importance of CXC chemokine receptor 2 in the homing of human peripheral blood endothelial progenitor cells to sites of arterial injury. Circ Res. 2007;100:590–597. doi: 10.1161/01.RES.0000259043.42571.68. [DOI] [PubMed] [Google Scholar]
  • 84.Hu C. Dandapat A. Mehta JL. Angiotensin II induces capillary formation from endothelial cells via the LOX-1 dependent redox-sensitive pathway. Hypertension. 2007;50:952–957. doi: 10.1161/HYPERTENSIONAHA.107.096446. [DOI] [PubMed] [Google Scholar]
  • 85.Huang L. Sankar S. Lin C. Kontos CD. Schroff AD. Cha EH. Feng SM. Li SF. Yu Z. Van Etten RL. Blanar MA. Peters KG. HCPTPA, a protein tyrosine phosphatase that regulates vascular endothelial growth factor receptor-mediated signal transduction and biological activity. J Biol Chem. 1999;274:38183–38188. doi: 10.1074/jbc.274.53.38183. [DOI] [PubMed] [Google Scholar]
  • 86.Huang L. Turck CW. Rao P. Peters KG. GRB2 and SH-PTP2: potentially important endothelial signaling molecules downstream of the TEK/TIE2 receptor tyrosine kinase. Oncogene. 1995;11:2097–2103. [PubMed] [Google Scholar]
  • 87.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]
  • 88.Ikeda S. Ushio-Fukai M. Zuo L. Tojo T. Dikalov S. Patrushev NA. Alexander RW. Novel role of ARF6 in vascular endothelial growth factor-induced signaling and angiogenesis. Circ Res. 2005;96:467–475. doi: 10.1161/01.RES.0000158286.51045.16. [DOI] [PubMed] [Google Scholar]
  • 89.Ikeda S. Yamaoka-Tojo M. Hilenski L. Patrushev NA. Anwar GM. Quinn MT. Ushio-Fukai M. IQGAP1 regulates reactive oxygen species-dependent endothelial cell migration through interacting with Nox2. Arterioscler Thromb Vasc B. 2005;25:2295–2300. doi: 10.1161/01.ATV.0000187472.55437.af. [DOI] [PubMed] [Google Scholar]
  • 90.Imanishi T. Hano T. Matsuo Y. Nishio I. Oxidized low-density lipoprotein inhibits vascular endothelial growth factor-induced endothelial progenitor cell differentiation. Clin Exp Pharmacol Physiol. 2003;30:665–670. doi: 10.1046/j.1440-1681.2003.03894.x. [DOI] [PubMed] [Google Scholar]
  • 91.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]
  • 92.Imanishi T. Hano T. Nishio I. Angiotensin II potentiates vascular endothelial growth factor-induced proliferation and network formation of endothelial progenitor cells. Hypertens Res. 2004;27:101–108. doi: 10.1291/hypres.27.101. [DOI] [PubMed] [Google Scholar]
  • 93.Imanishi T. Hano T. Sawamura T. Nishio I. Oxidized low-density lipoprotein induces endothelial progenitor cell senescence, leading to cellular dysfunction. Clin Exp Pharmacol Physiol. 2004;31:407–413. doi: 10.1111/j.1440-1681.2004.04022.x. [DOI] [PubMed] [Google Scholar]
  • 94.Inoue Y. Yagisawa M. Saeki K. Imajoh-Ohmi S. Kanegasaki S. Yuo A. Induction of phagocyte oxidase components during human myeloid differentiation: independent protein expression and discrepancy with the function. Biosci Biotechnol Biochem. 2001;65:2581–2584. doi: 10.1271/bbb.65.2581. [DOI] [PubMed] [Google Scholar]
  • 95.Ishikawa M. Stokes KY. Zhang JH. Nanda A. Granger DN. Cerebral microvascular responses to hypercholesterolemia: roles of NADPH oxidase and P-selectin. Circ Res. 2004;94:239–244. doi: 10.1161/01.RES.0000111524.05779.60. [DOI] [PubMed] [Google Scholar]
  • 96.Isner JM. Losordo DW. Therapeutic angiogenesis for heart failure. Nat Med. 1999;5:491–492. doi: 10.1038/8374. [DOI] [PubMed] [Google Scholar]
  • 97.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]
  • 98.Ivanova NB. Dimos JT. Schaniel C. Hackney JA. Moore KA. Lemischka IR. A stem cell molecular signature. Science (New York) 2002;298:601–604. doi: 10.1126/science.1073823. [DOI] [PubMed] [Google Scholar]
  • 99.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]
  • 100.Jiang F. Zhang G. Hashimoto I. Kumar BS. Bortolotto S. Morrison WA. Dusting GJ. Neovascularization in an arterio-venous loop-containing tissue engineering chamber: role of NADPH oxidase. J Cell Mol Med. 2008;12:2062–2072. doi: 10.1111/j.1582-4934.2008.00199.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kalka C. Masuda H. Takahashi T. Gordon R. Tepper O. Gravereaux E. Pieczek A. Iwaguro H. Hayashi SI. Isner JM. Asahara T. Vascular endothelial growth factor(165) gene transfer augments circulating endothelial progenitor cells in human subjects. Circ Res. 2000;86:1198–1202. doi: 10.1161/01.res.86.12.1198. [DOI] [PubMed] [Google Scholar]
  • 102.Khatri JJ. Johnson C. Magid R. Lessner SM. Laude KM. Dikalov SI. Harrison DG. Sung HJ. Rong Y. Galis ZS. Vascular oxidant stress enhances progression and angiogenesis of experimental atheroma. Circulation. 2004;109:520–525. doi: 10.1161/01.CIR.0000109698.70638.2B. [DOI] [PubMed] [Google Scholar]
  • 103.Kiel MJ. Yilmaz OH. Iwashita T. Yilmaz OH. Terhorst C. Morrison SJ. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell. 2005;121:1109–1121. doi: 10.1016/j.cell.2005.05.026. [DOI] [PubMed] [Google Scholar]
  • 104.Kim HW. Lin A. Guldberg RE. Ushio-Fukai M. Fukai T. Essential role of extracellular SOD in reparative neovascularization induced by hindlimb ischemia. Circ Res. 2007;101:409–419. doi: 10.1161/CIRCRESAHA.107.153791. [DOI] [PubMed] [Google Scholar]
  • 105.Kim YM. Kim KE. Koh GY. Ho YS. Lee KJ. Hydrogen peroxide produced by angiopoietin-1 mediates angiogenesis. Cancer Res. 2006;66:6167–6174. doi: 10.1158/0008-5472.CAN-05-3640. [DOI] [PubMed] [Google Scholar]
  • 106.Kobayashi S. Nojima Y. Shibuya M. Maru Y. Nox1 regulates apoptosis and potentially stimulates branching morphogenesis in sinusoidal endothelial cells. Exp Cell Res. 2004;300:455–462. doi: 10.1016/j.yexcr.2004.07.023. [DOI] [PubMed] [Google Scholar]
  • 107.Kocher AA. Schuster MD. Bonaros N. Lietz K. Xiang G. Martens TP. Kurlansky PA. Sondermeijer H. Witkowski P. Boyle A. Homma S. Wang SF. Itescu S. Myocardial homing and neovascularization by human bone marrow angioblasts is regulated by IL-8/Gro CXC chemokines. J Mol Cell Cardiol. 2006;40:455–464. doi: 10.1016/j.yjmcc.2005.11.013. [DOI] [PubMed] [Google Scholar]
  • 108.Kopp HG. Avecilla ST. Hooper AT. Rafii S. The bone marrow vascular niche: home of HSC differentiation and mobilization. Physiology (Bethesda) 2005;20:349–356. doi: 10.1152/physiol.00025.2005. [DOI] [PubMed] [Google Scholar]
  • 109.Kopp HG. Ramos CA. Rafii S. Contribution of endothelial progenitors and proangiogenic hematopoietic cells to vascularization of tumor and ischemic tissue. Curr Opin Hematol. 2006;13:175–181. doi: 10.1097/01.moh.0000219664.26528.da. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kraynov VS. Chamberlain C. Bokoch GM. Schwartz MA. Slabaugh S. Hahn KM. Localized Rac activation dynamics visualized in living cells. Science (New York) 2000;290:333–337. doi: 10.1126/science.290.5490.333. [DOI] [PubMed] [Google Scholar]
  • 111.Kroll J. Waltenberger J. The vascular endothelial growth factor receptor KDR activates multiple signal transduction pathways in porcine aortic endothelial cells. J Biol Chem. 1997;272:32521–32527. doi: 10.1074/jbc.272.51.32521. [DOI] [PubMed] [Google Scholar]
  • 112.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. 2007;292:H2582–H2588. doi: 10.1152/ajpheart.00786.2006. [DOI] [PubMed] [Google Scholar]
  • 113.Kuroki M. Voest EE. Amano S. Beerepoot LV. Takashima S. Tolentino M. Kim RY. Rohan RM. Colby KA. Yeo KT. Adamis AP. Reactive oxygen intermediates increase vascular endothelial growth factor expression in vitro and in vivo. J Clin Invest. 1996;98:1667–1675. doi: 10.1172/JCI118962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kwon J. Lee SR. Yang KS. Ahn Y. Kim YJ. Stadtman ER. Rhee SG. Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors. Proc Natl Acad Sci U S A. 2004;101:16419–16424. doi: 10.1073/pnas.0407396101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Lakshminarayanan V. Lewallen M. Frangogiannis NG. Evans AJ. Wedin KE. Michael LH. Entman ML. Reactive oxygen intermediates induce monocyte chemotactic protein-1 in vascular endothelium after brief ischemia. Am J Pathol. 2001;159:1301–1311. doi: 10.1016/S0002-9440(10)62517-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol. 2004;4:181–189. doi: 10.1038/nri1312. [DOI] [PubMed] [Google Scholar]
  • 117.Lambeth JD. Nox/Duox family of nicotinamide adenine dinucleotide (phosphate) oxidases. Curr Opin Hematol. 2002;9:11–17. doi: 10.1097/00062752-200201000-00003. [DOI] [PubMed] [Google Scholar]
  • 118.Lambeth JD. Cheng G. Arnold R. Edens WA. Novel homologs of gp91phox. Trends Biochem Sci. 2000;25:459–461. doi: 10.1016/s0968-0004(00)01658-3. [DOI] [PubMed] [Google Scholar]
  • 119.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;84:159–168. doi: 10.1093/cvr/cvn258. [DOI] [PubMed] [Google Scholar]
  • 120.Lassegue B. Clempus RE. Vascular NAD(P)H oxidases: specific features, expression, and regulation. Am J Physiol Regul Integr Comp Physiol. 2003;285:R277–R297. doi: 10.1152/ajpregu.00758.2002. [DOI] [PubMed] [Google Scholar]
  • 121.Lauer N. Suvorava T. Ruther U. Jacob R. Meyer W. Harrison DG. Kojda G. Critical involvement of hydrogen peroxide in exercise-induced up-regulation of endothelial NO synthase. Cardiovasc Res. 2005;65:254–262. doi: 10.1016/j.cardiores.2004.09.010. [DOI] [PubMed] [Google Scholar]
  • 122.Laufs U. Kilter H. Konkol C. Wassmann S. Bohm M. Nickenig G. Impact of HMG CoA reductase inhibition on small GTPases in the heart. Cardiovasc Res. 2002;53:911–920. doi: 10.1016/s0008-6363(01)00540-5. [DOI] [PubMed] [Google Scholar]
  • 123.Lee NK. Choi YG. Baik JY. Han SY. Jeong DW. Bae YS. Kim N. Lee SY. A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood. 2005;106:852–859. doi: 10.1182/blood-2004-09-3662. [DOI] [PubMed] [Google Scholar]
  • 124.Lee SR. Kwon KS. Kim SR. Rhee SG. Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor. J Biol Chem. 1998;273:15366–15372. doi: 10.1074/jbc.273.25.15366. [DOI] [PubMed] [Google Scholar]
  • 125.Lelkes PI. Hahn KL. Sukovich DA. Karmiol S. Schmidt DH. On the possible role of reactive oxygen species in angiogenesis. Adv Exp Med Biol. 1998;454:295–310. doi: 10.1007/978-1-4615-4863-8_35. [DOI] [PubMed] [Google Scholar]
  • 126.Li J-M. Shah AM. Intracellular localization and preassembly of the NADPH oxidase complex in cultured endothelial cells. J Biol Chem. 2002;277:19952–19960. doi: 10.1074/jbc.M110073200. [DOI] [PubMed] [Google Scholar]
  • 127.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]
  • 128.Li JM. Shah AM. Endothelial cell superoxide generation: regulation and relevance for cardiovascular pathophysiology. Am J Physiol Regul Integr Comp Physiol. 2004;287:R1014–R1030. doi: 10.1152/ajpregu.00124.2004. [DOI] [PubMed] [Google Scholar]
  • 129.Li Y. Yan J. De P. Chang HC. Yamauchi A. Christopherson KW., 2nd Paranavitana NC. Peng X. Kim C. Munugalavadla V. Kapur R. Chen H. Shou W. Stone JC. Kaplan MH. Dinauer MC. Durden DL. Quilliam LA. Rap1a null mice have altered myeloid cell functions suggesting distinct roles for the closely related Rap1a and 1b proteins. J Immunol. 2007;179:8322–8331. doi: 10.4049/jimmunol.179.12.8322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lin MT. Yen ML. Lin CY. Kuo ML. Inhibition of vascular endothelial growth factor-induced angiogenesis by resveratrol through interruption of Src-dependent vascular endothelial cadherin tyrosine phosphorylation. Mol Pharmacol. 2003;64:1029–1036. doi: 10.1124/mol.64.5.1029. [DOI] [PubMed] [Google Scholar]
  • 131.Liu LZ. Hu XW. Xia C. He J. Zhou Q. Shi X. Fang J. Jiang BH. Reactive oxygen species regulate epidermal growth factor-induced vascular endothelial growth factor and hypoxia-inducible factor-1alpha expression through activation of AKT and P70S6K1 in human ovarian cancer cells. Free Radic Biol Med. 2006;41:1521–1533. doi: 10.1016/j.freeradbiomed.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 132.Liu ZJ. Velazquez OC. Hyperoxia, endothelial progenitor cell mobilization, and diabetic wound healing. Antioxid Redox Signal. 2008;10:1869–1882. doi: 10.1089/ars.2008.2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Loomans CJ. De Koning EJ. Staal FJ. Rabelink TJ. Zonneveld AJ. Endothelial progenitor cell dysfunction in type 1 diabetes: another consequence of oxidative stress? Antioxid Redox Signal. 2005;7:1468–1475. doi: 10.1089/ars.2005.7.1468. [DOI] [PubMed] [Google Scholar]
  • 134.Losordo DW. Dimmeler S. Therapeutic angiogenesis and vasculogenesis for ischemic disease: part I: angiogenic cytokines. Circulation. 2004;109:2487–2491. doi: 10.1161/01.CIR.0000128595.79378.FA. [DOI] [PubMed] [Google Scholar]
  • 135.Luczak K. Balcerczyk A. Soszynski M. Bartosz G. Low concentration of oxidant and nitric oxide donors stimulate proliferation of human endothelial cells in vitro. Cell Biol Int. 2004;28:483–486. doi: 10.1016/j.cellbi.2004.03.004. [DOI] [PubMed] [Google Scholar]
  • 136.Marumo T. Uchimura H. Hayashi M. Hishikawa K. Fujita T. Aldosterone impairs bone marrow-derived progenitor cell formation. Hypertension. 2006;48:490–496. doi: 10.1161/01.HYP.0000235681.25685.cf. [DOI] [PubMed] [Google Scholar]
  • 137.Mateer SC. Wang N. Bloom GS. IQGAPs: integrators of the cytoskeleton, cell adhesion machinery, and signaling networks. Cell Motil Cytoskeleton. 2003;55:147–155. doi: 10.1002/cm.10118. [DOI] [PubMed] [Google Scholar]
  • 138.Matsumoto T. Claesson-Welsh L. VEGF receptor signal transduction. Sci STKE. 2001;2001:RE21. doi: 10.1126/stke.2001.112.re21. [DOI] [PubMed] [Google Scholar]
  • 139.Maulik N. Redox signaling of angiogenesis. Antioxid Redox Signal. 2002;4:805–815. doi: 10.1089/152308602760598963. [DOI] [PubMed] [Google Scholar]
  • 140.Michaud SE. Dussault S. Haddad P. Groleau J. Rivard A. Circulating endothelial progenitor cells from healthy smokers exhibit impaired functional activities. Atherosclerosis. 2006;187:423–432. doi: 10.1016/j.atherosclerosis.2005.10.009. [DOI] [PubMed] [Google Scholar]
  • 141.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]
  • 142.Min JK. Kim YM. Kim SW. Kwon MC. Kong YY. Hwang IK. Won MH. Rho J. Kwon YG. TNF-related activation-induced cytokine enhances leukocyte adhesiveness: induction of ICAM-1 and VCAM-1 via TNF receptor-associated factor and protein kinase C-dependent NF-kappaB activation in endothelial cells. J Immunol. 2005;175:531–540. doi: 10.4049/jimmunol.175.1.531. [DOI] [PubMed] [Google Scholar]
  • 143.Mitola S. Brenchio B. Piccinini M. Tertoolen L. Zammataro L. Breier G. Rinaudo MT. den Hertog J. Arese M. Bussolino F. Type I collagen limits VEGFR-2 signaling by a SHP2 protein-tyrosine phosphatase-dependent mechanism 1. Circ Res. 2006;98:45–54. doi: 10.1161/01.RES.0000199355.32422.7b. [DOI] [PubMed] [Google Scholar]
  • 144.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]
  • 145.Mofarrahi M. Brandes RP. Gorlach A. Hanze J. Terada LS. Quinn MT. Mayaki D. Petrof B. Hussain SN. Regulation of proliferation of skeletal muscle precursor cells by NADPH oxidase. Antioxid Redox Signal. 2008;10:559–574. doi: 10.1089/ars.2007.1792. [DOI] [PubMed] [Google Scholar]
  • 146.Moldovan L. Irani K. Moldovan NI. Finkel T. Goldschmidt-Clermont PJ. The actin cytoskeleton reorganization induced by Rac1 requires the production of superoxide. Antioxid Redox Signal. 1999;1:29–43. doi: 10.1089/ars.1999.1.1-29. [DOI] [PubMed] [Google Scholar]
  • 147.Moldovan L. Moldovan NI. Sohn RH. Parikh SA. Goldschmidt-Clermont PJ. Redox changes of cultured endothelial cells and actin dynamics. Circ Res. 2000;86:549–557. doi: 10.1161/01.res.86.5.549. [DOI] [PubMed] [Google Scholar]
  • 148.Moulton KS. Heller E. Konerding MA. Flynn E. Palinski W. Folkman J. Angiogenesis inhibitors endostatin or TNP-470 reduce intimal neovascularization and plaque growth in apolipoprotein E-deficient mice. Circulation. 1999;99:1726–1732. doi: 10.1161/01.cir.99.13.1726. [DOI] [PubMed] [Google Scholar]
  • 149.Muller WA. Leukocyte-endothelial cell interactions in the inflammatory response. Lab Invest. 2002;82:521–533. doi: 10.1038/labinvest.3780446. [DOI] [PubMed] [Google Scholar]
  • 150.Nahmod KA. Vermeulen ME. Raiden S. Salamone G. Gamberale R. Fernandez-Calotti P. Alvarez A. Nahmod V. Giordano M. Geffner JR. Control of dendritic cell differentiation by angiotensin II. FASEB J. 2003;17:491–493. doi: 10.1096/fj.02-0755fje. [DOI] [PubMed] [Google Scholar]
  • 151.Nakagami H. Cui TX. Iwai M. Shiuchi T. Takeda-Matsubara Y. Wu L. Horiuchi M. Tumor necrosis factor-alpha inhibits growth factor-mediated cell proliferation through SHP-1 activation in endothelial cells. Arterioscler Thromb Vasc Biol. 2002;22:238–242. doi: 10.1161/hq0202.104001. [DOI] [PubMed] [Google Scholar]
  • 152.Nakamura Y. Patrushev N. Inomata H. Mehta D. Urao N. Kim HW. Razvi M. Kini V. Mahadev K. Goldstein BJ. McKinney R. Fukai T. Ushio-Fukai M. Role of protein tyrosine phosphatase 1B in vascular endothelial growth factor signaling and cell-cell adhesions in endothelial cells. Circ Res. 2008;102:1182–1191. doi: 10.1161/CIRCRESAHA.107.167080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Nespereira B. Perez-Ilzarbe M. Fernandez P. Fuentes AM. Paramo JA. Rodriguez JA. Vitamins C and E downregulate vascular VEGF and VEGFR-2 expression in apolipoprotein-E-deficient mice. Atherosclerosis. 2003;171:67–73. doi: 10.1016/j.atherosclerosis.2003.08.009. [DOI] [PubMed] [Google Scholar]
  • 154.Noble M. Smith J. Power J. Mayer-Proschel M. Redox state as a central modulator of precursor cell function. Ann N Y Acad Sci. 2003;991:251–271. doi: 10.1111/j.1749-6632.2003.tb07481.x. [DOI] [PubMed] [Google Scholar]
  • 155.Park HS. Lee SH. Park D. Lee JS. Ryu SH. Lee WJ. Rhee SG. Bae YS. Sequential activation of phosphatidylinositol 3-kinase, beta Pix, Rac1, and Nox1 in growth factor-induced production of H2O2. Mol Cell Biol. 2004;24:4384–4394. doi: 10.1128/MCB.24.10.4384-4394.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Paysant JR. Rupin A. Verbeuren TJ. Effect of NADPH oxidase inhibition on E-selectin expression induced by concomitant anoxia/reoxygenation and TNF-alpha. Endothelium. 2002;9:263–271. doi: 10.1080/10623320214737. [DOI] [PubMed] [Google Scholar]
  • 157.Petry A. Djordjevic T. Weitnauer M. Kietzmann T. Hess J. Gorlach A. NOX2 and NOX4 mediate proliferative response in endothelial cells. Antioxid Redox Signal. 2006;8:1473–1484. doi: 10.1089/ars.2006.8.1473. [DOI] [PubMed] [Google Scholar]
  • 158.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]
  • 159.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]
  • 160.Polytarchou C. Papadimitriou E. Antioxidants inhibit angiogenesis in vivo through down-regulation of nitric oxide synthase expression and activity. Free Radic Res. 2004;38:501–508. doi: 10.1080/10715760410001684621. [DOI] [PubMed] [Google Scholar]
  • 161.Polytarchou C. Papadimitriou E. Antioxidants inhibit human endothelial cell functions through down-regulation of endothelial nitric oxide synthase activity. Eur J Pharmacol. 2005;510:31–38. doi: 10.1016/j.ejphar.2005.01.004. [DOI] [PubMed] [Google Scholar]
  • 162.Puceat M. Travo P. Quinn MT. Fort P. A dual role of the GTPase Rac in cardiac differentiation of stem cells. Mol Biol Cell. 2003;14:2781–2792. doi: 10.1091/mbc.E02-09-0562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.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]
  • 164.Rafii S. Heissig B. Hattori K. Efficient mobilization and recruitment of marrow-derived endothelial and hematopoietic stem cells by adenoviral vectors expressing angiogenic factors. Gene Ther. 2002;9:631–641. doi: 10.1038/sj.gt.3301723. [DOI] [PubMed] [Google Scholar]
  • 165.Rafii S. Mohle R. Shapiro F. Frey BM. Moore MA. Regulation of hematopoiesis by microvascular endothelium. Leuk Lymphoma. 1997;27:375–386. doi: 10.3109/10428199709058305. [DOI] [PubMed] [Google Scholar]
  • 166.Ramalho-Santos M. Yoon S. Matsuzaki Y. Mulligan RC. Melton DA. “Stemness”: transcriptional profiling of embryonic and adult stem cells. Science (New York) 2002;298:597–600. doi: 10.1126/science.1072530. [DOI] [PubMed] [Google Scholar]
  • 167.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]
  • 168.Rocic P. Kolz C. Reed R. Potter B. Chilian WM. Optimal reactive oxygen species concentration and p38 MAP kinase are required for coronary collateral growth. Am J Physiol. 2007;292:H2729–H2736. doi: 10.1152/ajpheart.01330.2006. [DOI] [PubMed] [Google Scholar]
  • 169.Rodgers K. Xiong S. DiZerega GS. Effect of angiotensin II and angiotensin(1–7) on hematopoietic recovery after intravenous chemotherapy. Cancer Chemother Pharmacol. 2003;51:97–106. doi: 10.1007/s00280-002-0509-4. [DOI] [PubMed] [Google Scholar]
  • 170.Rosso A. Balsamo A. Gambino R. Dentelli P. Falcioni R. Cassader M. Pegoraro L. Pagano G. Brizzi MF. p53 Mediates the accelerated onset of senescence of endothelial progenitor cells in diabetes. J Biol Chem. 2006;281:4339–4347. doi: 10.1074/jbc.M509293200. [DOI] [PubMed] [Google Scholar]
  • 171.Ruef J. Hu ZY. Yin LY. Wu Y. Hanson SR. Kelly AB. Harker LA. Rao GN. Runge MS. Patterson C. Induction of vascular endothelial growth factor in balloon-injured baboon arteries. Circ Res. 1997;81:24–33. doi: 10.1161/01.res.81.1.24. [DOI] [PubMed] [Google Scholar]
  • 172.Rupin A. Paysant J. Sansilvestri-Morel P. Lembrez N. Lacoste JM. Cordi A. Verbeuren TJ. Role of NADPH oxidase-mediated superoxide production in the regulation of E-selectin expression by endothelial cells subjected to anoxia/reoxygenation. Cardiovasc Res. 2004;63:323–330. doi: 10.1016/j.cardiores.2004.03.018. [DOI] [PubMed] [Google Scholar]
  • 173.Salguero G. Akin E. Templin C. Kotlarz D. Doerries C. Landmesser U. Grote K. Schieffer B. Renovascular hypertension by two-kidney one-clip enhances endothelial progenitor cell mobilization in a p47phox-dependent manner. J Hypertens. 2008;26:257–268. doi: 10.1097/HJH.0b013e3282f09f79. [DOI] [PubMed] [Google Scholar]
  • 174.Sattler M. Verma S. Shrikhande G. Byrne CH. Pride YB. Winkler T. Greenfield EA. Salgia R. Griffin JD. The BCR/ABL tyrosine kinase induces production of reactive oxygen species in hematopoietic cells. J Biol Chem. 2000;275:24273–24278. doi: 10.1074/jbc.M002094200. [DOI] [PubMed] [Google Scholar]
  • 175.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]
  • 176.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]
  • 177.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]
  • 178.Sauer H. Ruhe C. Muller JP. Schmelter M. D'Souza R. Wartenberg M. Reactive oxygen species and upregulation of NADPH oxidases in mechanotransduction of embryonic stem cells. Methods Mol Biol (Clifton, NJ) 2008;477:397–418. doi: 10.1007/978-1-60327-517-0_30. [DOI] [PubMed] [Google Scholar]
  • 179.Schatteman GC. Hanlon HD. Jiao C. Dodds SG. Christy BA. Blood-derived angioblasts accelerate blood-flow restoration in diabetic mice. J Clin Invest. 2000;106:571–578. doi: 10.1172/JCI9087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.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]
  • 180a.Sekhsaria S. Fileisher TA. Vowells S. Brown M. Miller J. Gordon I. Blaese RM. Dunbar CE. Leitman S. Malech HL. Granulocyte colony-stimulating-factor recruitment of CD34+ progenitors to peripheral blood: impaired mobilization in chronic granulomatous disease and adenosine deaminase-deficient severe combined immunodeficiency disease patients. Blood. 1996;88:1104–1112. [PubMed] [Google Scholar]
  • 181.Shantsila E. Watson T. Lip GY. Endothelial progenitor cells in cardiovascular disorders. J Am Coll Cardiol. 2007;49:741–752. doi: 10.1016/j.jacc.2006.09.050. [DOI] [PubMed] [Google Scholar]
  • 182.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]
  • 183.Shono T. Ono M. Izumi H. Jimi SI. Matsushima K. Okamoto T. Kohno K. Kuwano M. Involvement of the transcription factor NF-kappaB in tubular morphogenesis of human microvascular endothelial cells by oxidative stress. Mol Cell Biol. 1996;16:4231–4239. doi: 10.1128/mcb.16.8.4231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Simon MC. Keith B. The role of oxygen availability in embryonic development and stem cell function. Nat Rev. 2008;9:285–296. doi: 10.1038/nrm2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Skyschally A. Schulz R. Gres P. Korth HG. Heusch G. Attenuation of ischemic preconditioning in pigs by scavenging of free oxyradicals with ascorbic acid. Am J Physiol. 2003;284:H698–H703. doi: 10.1152/ajpheart.00693.2002. [DOI] [PubMed] [Google Scholar]
  • 186.Soeda S. Shimada T. Koyanagi S. Yokomatsu T. Murano T. Shibuya S. Shimeno H. An attempt to promote neo-vascularization by employing a newly synthesized inhibitor of protein tyrosine phosphatase. FEBS Lett. 2002;524:54–58. doi: 10.1016/s0014-5793(02)03002-8. [DOI] [PubMed] [Google Scholar]
  • 187.Sorrentino SA. Bahlmann FH. Besler C. Muller M. Schulz S. Kirchhoff N. Doerries C. Horvath T. Limbourg A. Limbourg F. Fliser D. Haller H. Drexler H. Landmesser U. Oxidant stress impairs in vivo reendothelialization capacity of endothelial progenitor cells from patients with type 2 diabetes mellitus: restoration by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. Circulation. 2007;116:163–173. doi: 10.1161/CIRCULATIONAHA.106.684381. [DOI] [PubMed] [Google Scholar]
  • 188.Steinbeck MJ. Kim JK. Trudeau MJ. Hauschka PV. Karnovsky MJ. Involvement of hydrogen peroxide in the differentiation of clonal HD-11EM cells into osteoclast-like cells. J Cell Physiol. 1998;176:574–587. doi: 10.1002/(SICI)1097-4652(199809)176:3<574::AID-JCP14>3.0.CO;2-#. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Stone JR. Collins T. The role of hydrogen peroxide in endothelial proliferative responses. Endothelium. 2002;9:231–238. doi: 10.1080/10623320214733. [DOI] [PubMed] [Google Scholar]
  • 190.Storz G. Polla BS. Transcriptional regulators of oxidative stress-inducible genes in prokaryotes and eukaryotes. EXS. 1996;77:239–254. doi: 10.1007/978-3-0348-9088-5_16. [DOI] [PubMed] [Google Scholar]
  • 191.Strawn WB. Ferrario CM. Angiotensin II AT1 receptor blockade normalizes CD11b+ monocyte production in bone marrow of hypercholesterolemic monkeys. Atherosclerosis. 2008;196:624–632. doi: 10.1016/j.atherosclerosis.2007.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Takahashi T. Kalka C. Masuda H. Chen D. Silver M. Kearney M. Magner M. Isner JM. Asahara T. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med. 1999;5:434–438. doi: 10.1038/7434. [DOI] [PubMed] [Google Scholar]
  • 193.Takano M. Meneshian A. Sheikh E. Yamakawa Y. Wilkins KB. Hopkins EA. Bulkley GB. Rapid upregulation of endothelial P-selectin expression via reactive oxygen species generation. Am J Physiol. 2002;283:H2054–2061. doi: 10.1152/ajpheart.01001.2001. [DOI] [PubMed] [Google Scholar]
  • 194.Tanaka K. Weihrauch D. Kehl F. Ludwig LM. LaDisa JF., Jr Kersten JR. Pagel PS. Warltier DC. Mechanism of preconditioning by isoflurane in rabbits: a direct role for reactive oxygen species. Anesthesiology. 2002;97:1485–1490. doi: 10.1097/00000542-200212000-00021. [DOI] [PubMed] [Google Scholar]
  • 195.Tang FY. Meydani M. Green tea catechins and vitamin E inhibit angiogenesis of human microvascular endothelial cells through suppression of IL-8 production. Nutr Cancer. 2001;41:119–125. doi: 10.1080/01635581.2001.9680622. [DOI] [PubMed] [Google Scholar]
  • 196.Tepper OM. Capla JM. Galiano RD. Ceradini DJ. Callaghan MJ. Kleinman ME. Gurtner GC. Adult vasculogenesis occurs through in situ recruitment, proliferation, and tubulization of circulating bone marrow-derived cells. Blood. 2005;105:1068–1077. doi: 10.1182/blood-2004-03-1051. [DOI] [PubMed] [Google Scholar]
  • 197.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]
  • 198.Terada LS. Specificity in reactive oxidant signaling: think globally, act locally. J Cell Biol. 2006;174:615–623. doi: 10.1083/jcb.200605036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Thum T. Fraccarollo D. Galuppo P. Tsikas D. Frantz S. Ertl G. Bauersachs J. Bone marrow molecular alterations after myocardial infarction: impact on endothelial progenitor cells. Cardiovasc Res. 2006;70:50–60. doi: 10.1016/j.cardiores.2006.01.002. [DOI] [PubMed] [Google Scholar]
  • 200.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]
  • 201.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]
  • 202.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. Gillil 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]
  • 203.Tousoulis D. Andreou I. Antoniades C. Tentolouris C. Stefanadis C. Role of inflammation and oxidative stress in endothelial progenitor cell function and mobilization: therapeutic implications for cardiovascular diseases. Atherosclerosis. 2008;201:236–247. doi: 10.1016/j.atherosclerosis.2008.05.034. [DOI] [PubMed] [Google Scholar]
  • 204.Tso C. Martinic G. Fan WH. Rogers C. Rye KA. Barter PJ. High-density lipoproteins enhance progenitor-mediated endothelium repair in mice. Arterioscler Thromb Vasc Biol. 2006;26:1144–1149. doi: 10.1161/01.ATV.0000216600.37436.cf. [DOI] [PubMed] [Google Scholar]
  • 205.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]
  • 206.Ushio-Fukai M. Localizing NADPH oxidase-derived ROS. Sci STKE. 2006;2006:re8. doi: 10.1126/stke.3492006re8. [DOI] [PubMed] [Google Scholar]
  • 207.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]
  • 208.Ushio-Fukai M. Alexander RW. Reactive oxygen species as mediators of angiogenesis signaling: role of NAD(P)H oxidase. Mol Cell Biochem. 2004;264:85–97. doi: 10.1023/b:mcbi.0000044378.09409.b5. [DOI] [PubMed] [Google Scholar]
  • 209.Ushio-Fukai M. Nakamura Y. Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 2008;266:37–52. doi: 10.1016/j.canlet.2008.02.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Ushio-Fukai M. Tang Y. Fukai T. Dikalov S. Ma Y. Fujimoto M. Quinn MT. Pagano PJ. Johnson C. Alexander RW. Novel role of gp91phox-containing NAD(P)H oxidase in vascular endothelial growth factor-induced signaling and angiogenesis. Circ Res. 2002;91:1160–1167. doi: 10.1161/01.res.0000046227.65158.f8. [DOI] [PubMed] [Google Scholar]
  • 211.Vallet P. Charnay Y. Steger K. Ogier-Denis E. Kovari E. Herrmann F. Michel JP. Szanto I. Neuronal expression of the NADPH oxidase NOX4, and its regulation in mouse experimental brain ischemia. Neuroscience. 2005;132:233–238. doi: 10.1016/j.neuroscience.2004.12.038. [DOI] [PubMed] [Google Scholar]
  • 212.van Buul JD. Anthony EC. Fernandez-Borja M. Burridge K. Hordijk PL. Proline-rich tyrosine kinase 2 (Pyk2) mediates vascular endothelial-cadherin-based cell-cell adhesion by regulating beta-catenin tyrosine phosphorylation. J Biol Chem. 2005;280:21129–21136. doi: 10.1074/jbc.M500898200. [DOI] [PubMed] [Google Scholar]
  • 213.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]
  • 214.van Wetering S. van Buul JD. Quik S. Mul FP. Anthony EC. ten Klooster JP. Collard JG. Hordijk PL. Reactive oxygen species mediate Rac-induced loss of cell-cell adhesion in primary human endothelial cells. J Cell Sci. 2002;115:1837–1846. doi: 10.1242/jcs.115.9.1837. [DOI] [PubMed] [Google Scholar]
  • 215.Vasa M. Fichtlscherer S. Adler K. Aicher A. Martin H. Zeiher AM. Dimmeler S. Increase in circulating endothelial progenitor cells by statin therapy in patients with stable coronary artery disease. Circulation. 2001;103:2885–2890. doi: 10.1161/hc2401.092816. [DOI] [PubMed] [Google Scholar]
  • 216.Vepa S. Scribner WM. Parinandi NL. English D. Garcia JG. Natarajan V. Hydrogen peroxide stimulates tyrosine phosphorylation of focal adhesion kinase in vascular endothelial cells. Am J Physiol. 1999;277:L150–L158. doi: 10.1152/ajplung.1999.277.1.L150. [DOI] [PubMed] [Google Scholar]
  • 217.Voo S. Dunaeva M. Eggermann J. Stadler N. Waltenberger J. Diabetes mellitus impairs CD133+ progenitor cell function after myocardial infarction. J Intern Med. 2009;265:238–249. doi: 10.1111/j.1365-2796.2008.02011.x. [DOI] [PubMed] [Google Scholar]
  • 218.Wang N. Xie K. Huo S. Zhao J. Zhang S. Miao J. Suppressing phosphatidylcholine-specific phospholipase C and elevating ROS level, NADPH oxidase activity and Rb level induced neuronal differentiation in mesenchymal stem cells. J Cell Biochem. 2007;100:1548–1557. doi: 10.1002/jcb.21139. [DOI] [PubMed] [Google Scholar]
  • 219.Wang Y. Zeigler MM. Lam GK. Hunter MG. Eubank TD. Khramtsov VV. Tridandapani S. Sen CK. Marsh CB. The role of the NADPH oxidase complex, p38 MAPK, and Akt in regulating human monocyte/macrophage survival. Am J Respir Cell Mol Biol. 2007;36:68–77. doi: 10.1165/rcmb.2006-0165OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Watson T. Goon PK. Lip GY. Endothelial progenitor cells, endothelial dysfunction, inflammation, and oxidative stress in hypertension. Antioxid Redox Signal. 2008;10:1079–1088. doi: 10.1089/ars.2007.1998. [DOI] [PubMed] [Google Scholar]
  • 221.Weis M. Heeschen C. Glassford AJ. Cooke JP. Statins have biphasic effects on angiogenesis. Circulation. 2002;105:739–745. doi: 10.1161/hc0602.103393. [DOI] [PubMed] [Google Scholar]
  • 222.Werner C. Kamani CH. Gensch C. Bohm M. Laufs U. The peroxisome proliferator-activated receptor-gamma agonist pioglitazone increases number and function of endothelial progenitor cells in patients with coronary artery disease and normal glucose tolerance. Diabetes. 2007;56:2609–2615. doi: 10.2337/db07-0069. [DOI] [PubMed] [Google Scholar]
  • 223.Wientjes FB. Reeves EP. Soskic V. Furthmayr H. Segal AW. The NADPH oxidase components p47(phox) and p40(phox) bind to moesin through their PX domain. Biochem Biophys Res Commun. 2001;289:382–388. doi: 10.1006/bbrc.2001.5982. [DOI] [PubMed] [Google Scholar]
  • 224.Wo YB. Zhu DY. Hu Y. Wang ZQ. Liu J. Lou YJ. Reactive oxygen species involved in prenylflavonoids, icariin and icaritin, initiating cardiac differentiation of mouse embryonic stem cells. J Cell Biochem. 2008;103:1536–1550. doi: 10.1002/jcb.21541. [DOI] [PubMed] [Google Scholar]
  • 225.Wu RF. Gu Y. Xu YC. Nwariaku FE. Terada LS. Vascular endothelial growth factor causes translocation of p47phox to membrane ruffles through WAVE1. J Biol Chem. 2003;278:36830–36840. doi: 10.1074/jbc.M302251200. [DOI] [PubMed] [Google Scholar]
  • 226.Wu RF. Xu YC. Ma Z. Nwariaku FE. Sarosi GA., Jr Terada LS. Subcellular targeting of oxidants during endothelial cell migration. J Cell Biol. 2005;171:893–904. doi: 10.1083/jcb.200507004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.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–C723. doi: 10.1152/ajpcell.00442.2008. [DOI] [PubMed] [Google Scholar]
  • 228.Yamagishi S. Amano S. Inagaki Y. Okamoto T. Takeuchi M. Inoue H. Pigment epithelium-derived factor inhibits leptin-induced angiogenesis by suppressing vascular endothelial growth factor gene expression through anti-oxidative properties. Microvasc Res. 2003;65:186–190. doi: 10.1016/s0026-2862(03)00005-0. [DOI] [PubMed] [Google Scholar]
  • 229.Yamaoka-Tojo M. Tojo T. Kim HW. Hilenski L. Patrushev NA. Zhang L. Fukai T. Ushio-Fukai M. IQGAP1 mediates VE-cadherin-based cell-cell contacts and VEGF signaling at adherence junctions linked to angiogenesis. Arterioscler Thromb Vasc Biol. 2006;26:1991–1997. doi: 10.1161/01.ATV.0000231524.14873.e7. [DOI] [PubMed] [Google Scholar]
  • 230.Yamaoka-Tojo M. Ushio-Fukai M. Hilenski L. Dikalov SI. Chen YE. Tojo T. Fukai T. Fujimoto M. Patrushev NA. Wang N. Kontos CD. Bloom GS. Alexander RW. IQGAP1, a novel vascular endothelial growth factor receptor binding protein, is involved in reactive oxygen species-dependent endothelial migration and proliferation. Circ Res. 2004;95:276–283. doi: 10.1161/01.RES.0000136522.58649.60. [DOI] [PubMed] [Google Scholar]
  • 231.Yang Z. Sharma AK. Marshall M. Kron IL. Laubach VE. NADPH oxidase in bone marrow-derived cells mediates pulmonary ischemia-perfusion injury. Am J Respir Cell Mol Biol. 2009;40:375–381. doi: 10.1165/rcmb.2008-0300OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.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]
  • 233.Yao EH. Yu Y. Fukuda N. Oxidative stress on progenitor and stem cells in cardiovascular diseases. Curr Pharm Biotechnol. 2006;7:101–108. doi: 10.2174/138920106776597685. [DOI] [PubMed] [Google Scholar]
  • 234.Yasuda M. Ohzeki Y. Shimizu S. Naito S. Ohtsuru A. Yamamoto T. Kuroiwa Y. Stimulation of in vitro angiogenesis by hydrogen peroxide and the relation with ETS-1 in endothelial cells. Life Sci. 1999;64:249–258. doi: 10.1016/s0024-3205(98)00560-8. [DOI] [PubMed] [Google Scholar]
  • 235.Yasuda M. Shimizu S. Tokuyama S. Watanabe T. Kiuchi Y. Yamamoto T. A novel effect of polymorphonuclear leukocytes in the facilitation of angiogenesis [In Process Citation] Life Sci. 2000;66:2113–2121. doi: 10.1016/s0024-3205(00)00537-3. [DOI] [PubMed] [Google Scholar]
  • 236.Yin T. Ma X. Zhao L. Cheng K. Wang H. Angiotensin II promotes NO production, inhibits apoptosis and enhances adhesion potential of bone marrow-derived endothelial progenitor cells. Cell Res. 2008;18:792–799. doi: 10.1038/cr.2008.69. [DOI] [PubMed] [Google Scholar]
  • 237.Yoshida A. Yoshida S. Ishibashi T. Kuwano M. Inomata H. Suppression of retinal neovascularization by the NF-kappaB inhibitor pyrrolidine dithiocarbamate in mice. Invest Ophthalmol Vis Sci. 1999;40:1624–1629. [PubMed] [Google Scholar]
  • 238.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]
  • 239.Yu Y. Fukuda N. Yao EH. Matsumoto T. Kobayashi N. Suzuki R. Tahira Y. Ueno T. Matsumoto K. Effects of an ARB on endothelial progenitor cell function and cardiovascular oxidation in hypertension. Am J Hypertens. 2008;21:72–77. doi: 10.1038/ajh.2007.5. [DOI] [PubMed] [Google Scholar]
  • 240.Yun MR. Im DS. Lee JS. Son SM. Sung SM. Bae SS. Kim CD. NAD(P)H oxidase-stimulating activity of serum from type 2 diabetic patients with retinopathy mediates enhanced endothelial expression of E-selectin. Life Sci. 2006;78:2608–2614. doi: 10.1016/j.lfs.2005.10.044. [DOI] [PubMed] [Google Scholar]
  • 241.Zhang J. Niu C. Ye L. Huang H. He X. Tong WG. Ross J. Haug J. Johnson T. Feng JQ. Harris S. Wiedemann LM. Mishina Y. Li L. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425:836–841. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
  • 242.Zhao M. Wimmer A. Trieu K. Discipio RG. Schraufstatter IU. Arrestin regulates MAPK activation and prevents NADPH oxidase-dependent death of cells expressing CXCR2. J Biol Chem. 2004;279:49259–49267. doi: 10.1074/jbc.M405118200. [DOI] [PubMed] [Google Scholar]
  • 243.Zhu JH. Chen JZ. Wang XX. Xie XD. Sun J. Zhang FR. Homocysteine accelerates senescence and reduces proliferation of endothelial progenitor cells. J Mol Cell Cardiol. 2006;40:648–652. doi: 10.1016/j.yjmcc.2006.01.011. [DOI] [PubMed] [Google Scholar]
  • 244.Zhu QS. Xia L. Mills GB. Lowell CA. Touw IP. Corey SJ. G-CSF induced reactive oxygen species involves Lyn-PI3-kinase-Akt and contributes to myeloid cell growth. Blood. 2006;107:1847–1856. doi: 10.1182/blood-2005-04-1612. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Antioxidants & Redox Signaling are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES