Many studies have provided evidence for the crucial role of the reactive oxygen species (ROS) and reactive nitrogen species (RNS) in the regulation of differentiation and/or self-renewal, and the balance between quiescence and proliferation of hematopoietic stem cells (HSCs). Several metabolic regulators have been implicated in the maintenance of HSC redox homeostasis; however, the mechanisms that are regulated by ROS and RNS, as well as their downstream signaling are still elusive. This is partially owing to a lack of suitable methods that allow unequivocal and specific detection of ROS and RNS. In this Opinion, we first discuss the limitations of the commonly used techniques for detection of ROS and RNS, and the problem of heterogeneity of the cell population used in redox studies, which, together, can result in inaccurate conclusions regarding the redox biology of HSCs. We then propose approaches that are based on single-cell analysis followed by a functional test to examine ROS and RNS levels specifically in HSCs, as well as methods that might be used in vivo to overcome these drawbacks, and provide a better understanding of ROS and RNS function in stem cells.

Recent studies have revealed that a population of hematopoietic stem cells (HSCs) that resides in the hypoxic niche and mostly relies on anaerobic glycolysis is highly sensitive to oxidative stress (Simsek et al., 2010; Suda et al., 2011; Unwin et al., 2006). HSCs, and stem cells in general (Han et al., 2008), are highly vulnerable to increased levels of reactive oxygen species (ROS), whereas committed hematopoietic progenitor cells (HPCs) are not (Chen et al., 2008b; Ito et al., 2004; Jang and Sharkis, 2007). Nevertheless, an appropriate production of ROS is required for HSC functions, such as mobilization, survival, differentiation and proliferation (Guzy and Schumacker, 2006; Hole et al., 2010; Hosokawa et al., 2007; Jang and Sharkis, 2007; Sattler et al., 1999; Tesio et al., 2011; Wang et al., 2013). Furthermore, ROS modify gene expression and the activity of key metabolic regulators, thereby determining specific metabolic patterns and cell-fate decision (Bigarella et al., 2014). However, an excess of ROS is associated with DNA damage, lipid peroxidation, senescence and apoptosis (Naka et al., 2008; Navarro-Yepes et al., 2014; Shao et al., 2011). Furthermore, elevated ROS levels and accumulation of damaged DNA in human cord blood LinCD34+38 cells (enriched in HSCs) result in a reduction of their capacity to reconstitute hematopoiesis in immunosuppressed mice (Yahata et al., 2011). In addition to ROS, reactive nitrogen species (RNS) are integral to the redox status of the cell; ROS and RNS have overlapping functions and also affect each other's functions (Nathan and Cunningham-Bussel, 2013). Considering the role of RNS, nitric oxide (NO•) inhibits proliferation of HSCs and HPCs, and blocks their progression through the cell cycle, as well as induces apoptosis of CD34+ bone marrow (BM) cells in vitro (Reykdal et al., 1999). Furthermore, inhibition of nitric oxide synthase (NOS) increases the number of stem cells in the BM and the longevity of hematopoiesis in continuous BM cultures (Epperly et al., 2007; Michurina et al., 2004). However, compared with the work on ROS, there are considerably fewer studies that address RNS-mediated effects on HSCs and the signaling networks they participate in (Aleksinskaya et al., 2013; Bonafè et al., 2015; Nogueira-Pedro et al., 2014).

It has proven difficult to identify the specific ROS and RNS species that affect the functions of HSCs. This is partly due to a lack of suitable methods and chemical probes that allow the unequivocal distinction between different ROS and RNS, and to detect the complex intracellular chemistry of these diffusible and short-lived species. Furthermore, with the exception of a few studies that have been performed by using cell populations that were highly enriched in functional HSCs (Chuikov et al., 2010; Jung et al., 2013; Liu et al., 2009; Taniguchi Ishikawa et al., 2012), most studies that describe the role or consequence of ROS levels in redox measurements of HSCs were performed in heterogeneous cell populations that consist mainly of committed progenitor cells and only a small portion of functional HSCs (Table 1). In this Opinion article, we discuss how these limitations can lead to incorrect interpretation of data. Moreover, on the basis of recent advances in research, we present strategies that might enable to accurately detect ROS and RNS in HSCs and, thus, yield a better interpretation of the obtained results. We believe that these methods might result in important progress in our understanding of the redox regulation of stem cell physiology.

Table 1.

Overview of studies that have addressed the roles of ROS and RNS in HSC fate

Overview of studies that have addressed the roles of ROS and RNS in HSC fate
Overview of studies that have addressed the roles of ROS and RNS in HSC fate

The biochemical pathways in aerobic organisms that utilize O2 continuously generate the metabolic by-products ROS and RNS. Thus, aerobes have developed physiological mechanisms that enable ROS and RNS to be either detoxified or used as intracellular or intercellular signaling messengers to maintain cellular homeostasis. However, ROS and RNS are not single entities but a broad range of chemically distinct reactive species with diverse biological reactivities (Nathan and Cunningham-Bussel, 2013). There are several intracellular sources of ROS, including the mitochondrial electron transport chain (ETC), the membrane-bound NADPH oxidase (NOX) complex, the endoplasmic reticulum, oxidoreductase enzymes and metal-catalyzed oxidation reactions (Nathan and Cunningham-Bussel, 2013; Wang et al., 2013) (Fig. 1). Leaked electrons from the ETC can be captured by O2 to form the anion superoxide (O2), which – together with (NO•) – initiates the production of ROS and RNS. O2 can be rapidly converted into hydrogen peroxide (H2O2) by superoxide dismutases (SODs) or can react with NO• to form peroxynitrite (ONOO). H2O2 can be converted into the hydroxyl radical (•HO) in the presence of metal ions (Fe2+ or Cu 2+) in the Fenton reaction. In the presence of CO2, which is formed during multiple intracellular enzymatic reactions, ONOO gives rise to the RNS nitrosoperoxocarboxylate (ONO2CO2), which decomposes rapidly into the nitrogen dioxide (NO2•) and carbonate (CO3) radicals. The vast majority of biological reactions of ONO2CO2, including those with oxidant-sensitive probes, are mediated by NO2• and CO3 (Ferrer-Sueta and Radi, 2009). In the presence of chloride or thiocyanate, myeloperoxidase catalyzes the conversion of H2O2 into the oxidants hypochlorous acid (HOCl) or hypothiocyanous acid (HOSCN), respectively (Wardman, 2007; Winterbourn, 2014). The cellular levels of these oxidants are determined by the balance between their generation and capture by a number of antioxidant enzymes (e.g. catalases, peroxiredoxins, thioredoxins, thioredoxin reductases, and methionine sulphoxide reductases) (Nathan and Cunningham-Bussel, 2013). In addition, important redox cellular regulators are enzymes of the glutathione redox cycle, that detoxify H2O2 by using the reduced glutathione (GSH) (i.e. glutathione peroxidase) and regenerate oxidized glutathione (GSSG) to reduced glutathione (glutathione reductase) (Nathan and Cunningham-Bussel, 2013).

Fig. 1.

The cellular ROS and RNS networks. Leaked electrons from the mitochondria electro-transport chain can be captured by O2, forming anion superoxide (O2) that, together with nitric oxide (NO•), initiates production of ROS and RNS. O2 can be generated by the membrane-bound NADPH oxidase (NOX) complex, the endoplasmic reticulum or the mitochondrial electron transport chain (ETC). Once formed, O2 is rapidly converted into hydrogen peroxide (H2O2) by superoxide dismutase 1 (SOD1) in the cytoplasm and SOD2 in mitochondria or reacts with NO• to form peroxynitrite (ONOO). H2O2 can be converted into the hydroxyl radical (•HO) in the presence of metal ions (Fe2+ or Cu2+) in the Fenton reaction. In the presence of CO2, ONOO forms the RNS nitrosoperoxocarboxylate (ONO2CO2) that degrades rapidly into a nitrogen dioxide radical (NO2•) and a carbonate radical (CO3). In the presence of chloride or thiocyanate (Cl or SCN), myeloperoxidase catalyzes the conversion of H2O2 to the oxidants hypochlorous (HOCl) or hypothiocyanous acid (HOSCN), respectively. Catalase, glutathione peroxidase (Gpx) or peroxiredoxin (Prx) enzymes decompose H2O2 to H2O. C1–C5, respiratory chain complexes (C) 1–5. ROS and RNS suggested to be involved in HSC fate are shown in red.

Fig. 1.

The cellular ROS and RNS networks. Leaked electrons from the mitochondria electro-transport chain can be captured by O2, forming anion superoxide (O2) that, together with nitric oxide (NO•), initiates production of ROS and RNS. O2 can be generated by the membrane-bound NADPH oxidase (NOX) complex, the endoplasmic reticulum or the mitochondrial electron transport chain (ETC). Once formed, O2 is rapidly converted into hydrogen peroxide (H2O2) by superoxide dismutase 1 (SOD1) in the cytoplasm and SOD2 in mitochondria or reacts with NO• to form peroxynitrite (ONOO). H2O2 can be converted into the hydroxyl radical (•HO) in the presence of metal ions (Fe2+ or Cu2+) in the Fenton reaction. In the presence of CO2, ONOO forms the RNS nitrosoperoxocarboxylate (ONO2CO2) that degrades rapidly into a nitrogen dioxide radical (NO2•) and a carbonate radical (CO3). In the presence of chloride or thiocyanate (Cl or SCN), myeloperoxidase catalyzes the conversion of H2O2 to the oxidants hypochlorous (HOCl) or hypothiocyanous acid (HOSCN), respectively. Catalase, glutathione peroxidase (Gpx) or peroxiredoxin (Prx) enzymes decompose H2O2 to H2O. C1–C5, respiratory chain complexes (C) 1–5. ROS and RNS suggested to be involved in HSC fate are shown in red.

ROS and RNS have distinct chemical properties that result from differences in their reactivity, half-live and interaction with lipid moieties (D'Autréaux and Toledano, 2007). A selective reactivity with biological targets is the basis for the specific signaling pathways that are induced by individual ROS and RNS (D'Autréaux and Toledano, 2007). For instance, O2 preferentially reacts with proteins that contain iron–sulphur [Fe–S] clusters, whereas the uncharged H2O2 does not react with these proteins. Instead, H2O2 preferentially reacts with cysteine thiol groups that are present in the catalytic centers of enzymes. Moreover, not all cysteine residues are equally able to be oxidized by H2O2; this provides specificity of signaling induced by this ROS. NO• interacts with proteins that contain Fe–S clusters, heme (Cooper, 1999) or specific cysteine thiol groups (Marozkina and Gaston, 2012). The reactions mentioned above induce allosteric or conformational changes in target proteins (Bigarella et al., 2014; Finkel, 2011). These changes affect the function of proteins and their interaction with other proteins, and so can trigger distinct signaling events (D'Autréaux and Toledano, 2007; Stamler et al., 1992).

Several metabolic regulators and signaling pathways have been implicated in the maintenance of the redox homeostasis of HSCs. Activities of these regulators predispose HSCs to certain cell fates as summarized in Table 1.

It must be noted that the definition of an HSC is functional, based on the self-renewal capacity and differentiation potential of the cell. Unlike committed progenitors, HSCs are rare (1 in 10,000 within adult bone marrow cells), multipotent, self-renewing cells with a remarkable regenerative capacity that enables these cells to sustain hematopoiesis over a lifetime. As stated by John Dick: “The only conclusive method to assay stem cells is to follow their ability to repopulate conditioned recipients…” (Dick et al., 1997). HSCs can be identified either by limiting dilution or by single-cell transplantation assays that are based on the capacity of HSCs to hematopoietically reconstitute. For detection of human HSCs, immunosuppressed mice are typically used as recipients and their fate is followed up over 12 to 16 weeks (Ivanovic et al., 2011; Szilvassy et al., 1990; Till et al., 1964). A single HSC is sufficient for successful long-term hematopoietic reconstitution of an immunosuppressed mouse (Osawa et al., 1996).

The first indication that ROS are important in stem cells came from the work of Ito and co-workers (Ito et al., 2004). They demonstrated increased ROS levels in HSC-enriched populations after deletion of the ataxia telangiectasia mutated (ATM) gene in mice (Atm−/− mice) (Ito et al., 2004). These results were obtained in a phenotypically defined cell population that was Lineage (Lin, ‘L’) negative, stem cell antigen (Sca-1, ‘S’) positive, and c-kit (CD117, ‘K’) positive. This so-called LSK cell population contains at best ∼30% of ‘true’ HSCs or multipotent progenitors (MPPs) (Oguro et al., 2013). However, often <10% of LSK cells have real HSC potential based on in vivo long term-reconstitution capacity (Osawa et al., 1996); instead, the vast majority of these cells are multipotent and committed progenitors (Bryder et al., 2006). Furthermore, the LSK population can be subdivided into fractions based on the expression of SLAM family markers, CD150 (Slamf1) and CD48 (Slamf2): HSCs are particularly enriched in the CD150+CD48LSK population, MPPs in the CD150CD48LSK population, whereas CD150CD48+LSK and CD150+CD48+LSK cells contain heterogeneous committed progenitors (Kiel et al., 2005, 2008). The HSCs in the CD150+48 population can be further divided into cells that do or do not express the CD34 marker. The most quiescent and potent HSCs were found in the CD34CD150+CD48 subpopulation (Wilson et al., 2008). Despite the availability of these markers to determine a high enrichment in HSCs and MPPs, the resulting populations remain functionally heterogeneous (Oguro et al., 2013).

In Atm−/− mice, an increase in the levels of ROS coincides with the loss of HSC activity despite the maintenance of a phenotypically-defined LSK population (Ito et al., 2004). The underlying mechanisms were shown to involve ROS-mediated activation of p38 mitogen-activated protein kinases (MAPK11 and MAPK14), which upregulates the cyclin-dependent kinase inhibitors p16Ink4a and p19Arf. Upregulation of these inhibitors results in a consecutive block of cell-cycle-induced senescence and premature exhaustion of HSCs (Ito et al., 2006). Furthermore, Forkhead box class O (FoxO) transcription factors are crucial regulators of oxidative stress defense and ROS production (Storz, 2011). FoxO-deficient mice showed elevated levels of ROS associated with increased cycling and impaired long-term repopulating potential of LSK cells (Miyamoto et al., 2007; Tothova et al., 2007). FoxO-mediated redox regulation in HSCs is achieved partly through the regulation of ATM expression (Yalcin et al., 2008). Treatment of Atm−/− or FoxO-deficient mice with N-acetyl cysteine (NAC), a scavenger of HOCl, •HO, and H2O2 (Aruoma et al., 1989), reverses the ROS-induced defects, suggesting these oxidants have roles in HSCs and HPCs. Recently, it has been shown that ATM is activated directly by oxidative stress, which – in turn – stimulates an anti-oxidative cell response (Ditch and Paull, 2012). One such anti-oxidative protective mechanism is ATM-mediated suppression of the pro-apoptotic protein BID, which is known to act as an inducer of mitochondrial ROS (Maryanovich et al., 2012). The tuberous sclerosis complex (TSC)-mammalian target of rapamycin (mTOR) pathway is a crucial regulator of cell metabolism (Wullschleger et al., 2006). In response to oxidative stress, ATM-dependent inhibition of mTOR maintains quiescence of HSCs by repressing ROS production and mitogenesis (Chen et al., 2008a; Ditch and Paull, 2012). In addition, ROS induce activation of p53, which triggers apoptosis or the expression of the anti-oxidative defense enzymes in LSK cells (Abbas et al., 2010). Hypoxia-inducible factor 1-alpha (HIF1α) has also been implicated in a negative feedback loop of ROS regulation (Honma et al., 2013; Simsek et al., 2010; Takubo et al., 2010); ROS-stabilized HIFα (Piccoli et al., 2007a) induces adaptive metabolic responses that ensure redox homeostasis (Cam et al., 2010), such as inhibition of mTOR signaling and mitochondrial activity (Cam et al., 2010), as well as activation of glycolytic metabolism (Takubo et al., 2013).

Several factors are known to coordinate the balance between stem cell self-renewal and commitment with mitochondrial function. These include members of the polycomb family of proteins, such as B lymphoma Mo-MLV insertion region 1 homolog (Bmi1) and PR-domain-containing 16 (Prdm-16), as well as regulators of the DNA damage response, for example apurinic/apyrimidinic (AP) endonuclease1/redox factor-1 and redox regulator nuclear factor erythroid-2-related factor 2 (Nrf2) (Alfadda and Sallam, 2012; Chuikov et al., 2010; Liu et al., 2009; Wang et al., 2013).

An increase in ROS levels in an LSK population is paralleled by a decline in HSC activity (as measured by reduction of short- and long-term BM-reconstitution capacity) (Ito et al., 2004; Maryanovich et al., 2012; Miyamoto et al., 2007). However, in view of the fact that the functional HSCs represent often <10% of the LSK population, it is not clear whether ROS levels increase in HSCs themselves. In fact, the exhaustion of HSCs that have been detected in these experiments might originate from a release of ROS by non-HSCs, which represent the vast majority of LSK cells. These released ROS could induce HSC differentiation, senescence and apoptosis and/or necrosis.

Despite progress in understanding their effects on regulation of HSC and HPC functions, the exact ROS- and RNS-mediated mechanisms and their downstream signaling components have not been elucidated. For instance, it is unknown how ROS and RNS mediate cell fate at the molecular level. To fully understand how ROS and RNS contribute to the HSC fate, it is essential to detect these species accurately with regard to their identity, localization, kinetics and quantity because different reactive oxygen and nitrogen species might have very different roles in cellular signaling. For instance, compared with other ROS species, H2O2 is relatively stable (with a cellular half-life of ∼1 ms and steady-state levels of 10−7 M), diffusible and has selective reactivity (D'Autréaux and Toledano, 2007). These characteristics make it a likely candidate for a signaling molecule. Therefore, it is important to accurately detect this specific molecule without any erroneous contributions of other ROS and RNS species. However, most currently utilized techniques for the detection of ROS and RNS in HSCs and HPCs are not specific for a particular species, and any obtained measurements can be affected by the presence of other ROS or RNS species. Below, we will discuss the limitations and caveats of the commonly used techniques for the analysis of the redox status of stem cells.

The probes most commonly used to detect ROS and RNS are oxidant-sensitive compounds; these probes are often cell-permeable lipophilic esters that undergo intracellular hydrolysis and are retained in the cell, where – upon oxidation – they become fluorescent. Dihydrodichlorofluorescein (DCFH2), hydroethidine (HE), diaminofluorescein (DFA-2), Amplex Red as well as the chemiluminescence detectors, belong in this category (Wardman, 2007; Winterbourn, 2014) (Table 2).

Table 2.

Probes and sensors for ROS and RNS detection

Probes and sensors for ROS and RNS detection
Probes and sensors for ROS and RNS detection

These probes can be oxidized directly by various cellular radical species or by metal-dependent processes, which result in a radical-probe intermediate that is subsequently oxidized into the fluorescent product (Wardman, 2007). The radical-probe intermediate can react with various oxidants or can be captured and scavenged by cellular antioxidants. This means that oxidant-sensitive probes are generally non-specific for a particular oxidant, and their complex chemical behavior makes them unsuitable for mechanistic studies of cellular events that are induced by specific ROS and RNS species (Wardman, 2007; Winterbourn, 2014). Owing to the fact that a positive signal can be generated from non-specific reactions, the reactivity of oxidant-sensitive probes with particular ROS or RNS species should be confirmed by other techniques or by use of appropriate inhibitors (Zielonka et al., 2012b). However, their main advantage is that they are easy to use.

Non-specific detection of ROS and RNS: DCFH2

A common ROS probe is DCFH2 or its widely used esterified cell-permeable diacetate (DCFH2-DA); neither react directly with H2O2 to form the fluorescent product dichlorofluorescein (DCF) but require a peroxidase or redox-active metal catalyst (i.e. Fe2+). Indeed, it has been shown that the exposure to H2O2 alone is unable to oxidize DCFH2 (Karlsson et al., 2010). This requirement might lead to misinterpretations of data obtained with this probe and this issue has been discussed in detail in a number of reviews (Kalyanaraman et al., 2012; Karlsson et al., 2010; Wardman, 2007; Winterbourn, 2014). For example, a negative signal could be obtained if a metal catalyst is absent, whereas increased oxidation of DCFH2 in the presence of cytochrom c – which is released during apoptosis or under conditions of increased iron availability from the lysozyme – could be mistakenly associated with an increased oxidant production, as critically analyzed elsewhere (Burkitt and Wardman, 2001; Karlsson et al., 2010). Furthermore, oxidation of DCFH2 is entirely non-specific and can be achieved by a number of different free radicals – such as derivates of ONOO, tyrosine, thiols and HOCl as well as by light (Marchesi et al., 1999) – independently of H2O2 (Wardman, 2007; Wrona et al., 2005).

Thus, in contrast to what has been concluded in numerous reports with regard to a redox analysis of HSCs and HPCs (Ito et al., 2004, 2006; Miyamoto et al., 2007; Piccoli et al., 2007b, 2005) (Table 1), DCF fluorescence cannot be used as a direct measure of cellular H2O2 levels, nor as direct evidence that H2O2 is involved in a signaling event (Kalyanaraman et al., 2012; Karlsson et al., 2010; Wardman, 2007; Winterbourn, 2014).

DCFH2 has also been used as a general indicator of the redox status of HSCs and HPCs (Abbas et al., 2010; Chen et al., 2008a; Chuikov et al., 2010; Fan et al., 2007; Hosokawa et al., 2007; Jang and Sharkis, 2007; Jung et al., 2013; Juntilla et al., 2010; Liu et al., 2009; Pazhanisamy et al., 2011; Piccoli et al., 2007a; Shao et al., 2014; Yahata et al., 2011) (Table 1). However, using DCFH2 as a probe for the overall cellular redox status is highly unreliable owing to three limiting factors. First, oxidation of DCFH2 to DCF involves the intermediate radical DCFH• or DCF•, which – in the presence of O2 – can generate O2 (Wrona and Wardman, 2006). The subsequent dismutase-mediated oxidation of O2 produces additional H2O2, resulting in an artificial amplification of the initial DCF-induced fluorescence signal (Folkes et al., 2009). Second, the efficiency of oxidation of the intermediate DCFH2 probe radical into the final product depends on the environmental oxygen concentration (Wrona and Wardman, 2006). This issue is of particular importance when DCFH2 is used as a probe to study the effect of the O2 concentration on ROS generation in HSCs and HPCs (Fan et al., 2007; Hao et al., 2011). Third, cellular conditions may also alter the oxidation efficiency of DCFH2. For example, a higher detection sensitivity of DCFH2 in HSCs was reported in the presence of an inhibitor of the P-glycoprotein multidrug-resistant 1 (MDR1) pump (Piccoli et al., 2005); here, an apparent higher sensitivity of DCFH2 might be attributed to reduced expulsion of the probes by MDR1. In this context, it is worth noting that MDR1 is highly expressed in primitive HSCs (defined as cells that can in vivo provide long-term bone-marrow hematopoiesis or that can in vitro act as long-term culture-initiating cells) (Scharenberg et al., 2002).

Detection of O2: HE, MitoSOX

Hydroethidine (HE) is commonly used for the detection of intracellular O2 in studies of HSCs and HPCs (Jung et al., 2013; Nogueira-Pedro et al., 2014; Tesio et al., 2011; Wang et al., 2010). Although O2 can directly oxidize HE to its radical, the reaction is relatively slow and is more likely to be performed by other cellular oxidants present (Winterbourn, 2014). Once formed, the HE radical combines rapidly with O2 to generate the specific product 2-hydroxyethidium (2-OH-E+). However, HE can also react with other oxidants (e.g. ONOO, H2O2, •HO) to generate the non-specific product ethidium (E+) (Kalyanaraman et al., 2012; Wardman, 2007; Winterbourn, 2014). Although 2-OH-E+ and E+ have slightly different fluorescence spectra, they cannot be distinguished easily (Robinson et al., 2006). Therefore, a simple fluorescence assay performed with HE cannot provide sufficient specific information regarding intra- and extracellular O2 formation. Because the fluorescence signal can be generated from the non-specific reaction, any specificity of the fluorescent signal for O2 needs to be confirmed with ROS-scavenging enzymes and NOS inhibitors (Zielonka et al., 2012b).

In stem cell redox studies, mitochondrial O2 has been measured by mitochondrial-targeted HE (Mito-HE or MitoSOX) (Cho et al., 2014; Hole et al., 2010; Taniguchi Ishikawa et al., 2012; The et al., 2013). MitoSOX reacts with O2 in a similar manner as HE and, therefore, has the same limitations (Winterbourn, 2014). But as a positively charged molecule, MitoSOX has an increased affinity to react with O2, making the reaction more specific than the HE reaction with O2. Oxidation of MitoSOX by O2 generates the specific product red-fluorescent 2-hydroxymitoethidium (2-OH-Mito-E+), whereas its reaction with other oxidants results in the non-specific Mito-E (Zielonka and Kalyanaraman, 2010). However, because of the overlap of their fluorescence spectra, MitoSOX cannot be utilized on its own as a reliable indicator of mitochondrial O2 formation (Zielonka and Kalyanaraman, 2010).

Detection of H2O2: Amplex Red

The peroxidase substrate Amplex Red is used to detect H2O2 that has been extracellularly generated, as well as any that is produced inside the cell and has diffused out (Hole et al., 2010; Skinner et al., 2008; Thompson et al., 2008; Vlaski et al., 2014). Amplex Red can be oxidized to the fluorescent product resorufin by H2O2 or other electron oxidants (e.g. O2 and NO•). However, the specificity of Amplex Red for H2O2 depends on the presence of sufficient amounts of horseradish peroxidase (HRP). In this case, HRP catalyzes the oxidation of its substrate Amplex Red by H2O2 to resorufin in two one-electron oxidation steps (Gorris and Walt, 2009). However, there are some limitations, in particular the light-induced reduction of resorufin in the presence of biological reducing agents, such as NAD(P)H or glutathione, which results in an artificial amplification of the fluorescent signal (Zhao et al., 2011). Thus, in light-protected condition, Amplex Red can be used to specifically measure the extracellular formation of H2O2 in the presence of HRP (Kalyanaraman et al., 2012).

Detection of NO•

The probe diaminofluorecscein (DAF-2) and its cell-permeable diacetate (DFA-2DA) are used to monitor the formation of cellular NO• based on the generation of the resulting green fluorescent product DFA-2T (Zielonka et al., 2012b). DAF-2 does not react directly with NO•, but with N2O3, which is generated upon NO• oxidation and can be attenuated in the presence of the ascorbic acid (Ye et al., 2004). This method for NO• detection has been used in hematological studies (Čokić et al., 2009; Gorbunov et al., 2000). However, under conditions that generate both NO• and O2, it is not a suitable method to measure NO• selectively, because O2 contributes to the oxidation of DAF-2 into fluorescent DFA-2T. Therefore, NOS inhibitors or ROS-scavengers should be included in the assay in order to discriminate oxidation that is mediated by O2 from that mediated by NO• (Zielonka et al., 2012b).

Chemiluminescent probes

Chemiluminescent assays have been used to detect ROS in HSCs and HPCs (Hole et al., 2010) due to their high sensitivity and the fact they only require a relatively small number of cells (Winterbourn, 2014). However, these assays have the same limitations as oxidant-sensitive fluorescent probes. For instance, an assay based on luminol is unable to discriminate among individual ROS species present in biological systems or provide any mechanistic information (Zielonka et al., 2013). The luminol radical that is generated in the initial, non-specific oxidation can react in numerous interactions that can influence the resulting overall chemiluminescence, as described in detail elsewhere (Wardman, 2007). Luciferin analogues are more specific for the chemiluminescent detection of extracellular O2 (Teranishi, 2007), and this approach could, thus, be used to confirm the production of O2 that has been detected by using an alternative procedure.

Cell-permeable boronate probes contain a blocking group that is attached to the fluorophore. First, an oxidant reacts with and oxidizes the blocking group without changing the oxidation state of the fluorophore. The oxidized form of the blocking group is then released, resulting in fluorescence. These probes are thus referred to as ‘non-redox’ probes. This mechanism avoids several of the problematic steps that affect the probes discussed above, such as oxidation of the probe, generation of probe-radical intermediates and complex chemical reactions (Winterbourn, 2014). The first application of a non-redox probe was the detection of H2O2 in living cells (Dickinson et al., 2013; Lin et al., 2013; Lippert et al., 2011; Woolley et al., 2012). It was later shown that these probes (particularly in the case of aromatic boronate probes) react with ONOO much faster than with H2O2 (about a million-fold) (Sikora et al., 2009). Thus, the use of the ROS-scavenging enzymes [catalase, polyethylene glycol (PEG) catalase, SOD or PEG-SOD] and a NOS inhibitor (Nω-nitro-L-arginine methyl ester), is necessary to distinguish between ONOO-mediated and H2O2-mediated-fluorescence signals (Zielonka et al., 2012a).

Altogether, the studies presented above suggest that currently used techniques for ROS and RNS detection are not sufficiently specific. In addition, with exception of the boronate probe, they cannot be used in vivo. In light of these limitations, which are all related to the complexity of cellular redox chemistry, a better approach to examine ROS and RNS in vitro and in vivo – and specifically in HSCs as opposed to HPC systems – is needed.

A possible assay that could be applied to stem cell biology came from a recent study suggesting that fluorescence-based techniques using HE, boronate probes, DAF-DA, can be used for initial screening before – in a second step – specificity of these probes for certain reactive species is confirmed with NOS inhibitors and ROS-detoxifying enzymes (Zielonka et al., 2012b). It should be noted that there is always uncertainty as to whether the ROS scavengers reach the site of oxidation. Because all these probes allow analysis at single-cell level by flow cytometry, we propose the following procedure to measure ROS and RNS in LSK cell populations that contain HSCs and HPCs (Fig. 2A). A cell population is assayed by using any of these probes (step 1) and specificity of fluorescence signal confirmed before cells are sorted into subpopulations on the basis whether or not they respond to a particular ROS or RNS probe (‘ROS/RNS low’ or ‘ROS/RNS high’) (step 2). The following step 3 could then include the sorting of individual cells by their respective phenotypic properties. In the examples shown in Fig. 2A, individual cells within the resulting subpopulations (i.e. ROS/RNS low and ROS/RNS high) are sorted according to whether they express the SLAM family phenotypic markers, CD150 and CD48. The CD150+CD48LSK population that is enriched in HSCs can so be distinguished from a population that contains mainly MPPs (CD150CD48LSK) or HPCs (CD150+CD48+LSKs) (Oguro et al., 2013). This step could then be followed by the functional analysis of the resulting single cells in order to determine whether they are true HSCs because this cannot be determined with certainty on the basis of surface markers analysis alone (step 4). Such a functional analysis could involve the testing of single cells for their capacity for hematopoietic reconstitution or their proliferative potential (Fig. 2A). This type of approach would not only help to overcome the problem of functional heterogeneity in phenotypically homogeneous HSCs/HPCs populations (such as LSK cells), but also allow to determine the ROS and/or RNS profiles of functional HSC subpopulations.

Fig. 2.

Possible single-cell approach for ROS and RNS detection in HSCs. (A) Possible experimental single-cell approach to analyse the ROS and RNS content in functional HSCs. A cell population enriched in HSCs (LSK cells in this case) is treated with a fluorescent probe for ROS and RNS (step 1) and examined by flow cytometry to sort the LSK cells into subpopulations on the basis of whether or not they respond to the ROS or RNS probe (i.e. ROS/RNS low or ROS/RNS high) (step 2). Step 3 involves the sorting of individual cells on the basis of their phenotypic properties, such as expression of the SLAM family phenotypic markers CD150 and CD48. The CD150+CD48LSK subpopulation (enriched in HSCs) can be distinguished from CD150CD48LSK cells (enriched in MPPs), CD150+CD48+LSK cells and CD150CD48+LSK cells that mainly contain HPCs. Subsequently, single cells from the HSC-enriched CD150+CD48LSK subpopulation are analyzed to identify true HSCs on the basis of their capacity for hematopoietic reconstitution or proliferative potential (step 4). Consecutively, results from the functional assay are correlated with the ROS/RNS status determined in step 2. (B) Possible approach to detect ROS in HSC-enriched cell populations in situ. To determine the redox state of a HSC compared with that of HPCs in situ, a first step could involve the creation of transgenic mice that express the genetically encoded fluorescent-protein-based redox sensors roGFP or Hyper. The redox state or H2O2 levels could then be followed in a sorted, HSC-enriched subpopulation (CD150+CD48LSK cells). The same strategy could also include the use of boronate-based fluorescent probes that allow to measure changes in H2O2 levels in real-time.

Fig. 2.

Possible single-cell approach for ROS and RNS detection in HSCs. (A) Possible experimental single-cell approach to analyse the ROS and RNS content in functional HSCs. A cell population enriched in HSCs (LSK cells in this case) is treated with a fluorescent probe for ROS and RNS (step 1) and examined by flow cytometry to sort the LSK cells into subpopulations on the basis of whether or not they respond to the ROS or RNS probe (i.e. ROS/RNS low or ROS/RNS high) (step 2). Step 3 involves the sorting of individual cells on the basis of their phenotypic properties, such as expression of the SLAM family phenotypic markers CD150 and CD48. The CD150+CD48LSK subpopulation (enriched in HSCs) can be distinguished from CD150CD48LSK cells (enriched in MPPs), CD150+CD48+LSK cells and CD150CD48+LSK cells that mainly contain HPCs. Subsequently, single cells from the HSC-enriched CD150+CD48LSK subpopulation are analyzed to identify true HSCs on the basis of their capacity for hematopoietic reconstitution or proliferative potential (step 4). Consecutively, results from the functional assay are correlated with the ROS/RNS status determined in step 2. (B) Possible approach to detect ROS in HSC-enriched cell populations in situ. To determine the redox state of a HSC compared with that of HPCs in situ, a first step could involve the creation of transgenic mice that express the genetically encoded fluorescent-protein-based redox sensors roGFP or Hyper. The redox state or H2O2 levels could then be followed in a sorted, HSC-enriched subpopulation (CD150+CD48LSK cells). The same strategy could also include the use of boronate-based fluorescent probes that allow to measure changes in H2O2 levels in real-time.

Furthermore, there are other approaches to determine the redox state of HSCs compared with that of HPCs in their physiological environment. A major breakthrough of ROS detection in vivo within cellular and subcellular compartments, as well as in vitro, was the development of genetically encoded fluorescent-protein-based redox sensors that are based on the introduction of cysteines into yellow or green fluorescent protein (rxYFP or roGFP) (Dooley et al., 2004; Østergaard et al., 2001). Signal detection is based on the change in the chromophore spectrum upon oxidation or reduction of these cysteines (Lukyanov and Belousov, 2014). The extent of the redox change can be estimated from the relative fluorescence intensity of the two fluorescence excitation maxima of the sensor, making the measurement ratiometric (Lukyanov and Belousov, 2014). These sensors enable specific, real-time, reversible, ratiometric detection of the changes in the redox state (Lukyanov and Belousov, 2014). In addition, these probes enable the specific detection and quantification of H2O2 or of the glutathione redox potential (GSH:GSSG ratio) at the single-cell level in vitro (Shimi et al., 2011) and in vivo (Albrecht et al., 2011; Kojer et al., 2012). For instance, using roGTP transgenic flies, this approach has enabled the in vivo mapping of H2O2 and of oxidized glutathione during development and aging (Albrecht et al., 2011). Another H2O2-sensitive sensor, Hyper, was developed by inserting the circularly permuted YFP (cpYFP) into the regulatory domain of the E. coli H2O2-sensing protein OxyR (Choi et al., 2001). This probe enabled in vivo imaging of H2O2 gradients at tissue-scale as well as their quantification (Bilan et al., 2013; Love et al., 2013). Furthermore, this method was used to follow redox changes specifically in Hyper-expressing neutrophils in vivo (Pase et al., 2012).

By using these genetically modified approaches, it should be possible to measure the redox state of a HSC in situ. For instance, a mouse expressing roGFP or Hyper could be generated, from which HSC-enriched subpopulations are obtained according to the flow cytometry-based sorting procedure outlined above in order to determine and analyze their ROS levels (Fig. 2B). Alternatively, boronate-based fluorescent probes that enable measurement of real-time changes in H2O2 in tissue, cells and subcellular compartments could be used to assess the HSC redox status in situ (Cochemé et al., 2011; Dickinson et al., 2011; Van de Bittner et al., 2010). Here, boronate probes could be administered to mice via a tail-vein injection before their bone marrow cells are extracted, analyzed and sorted by flow cytometry with respect to their redox status and surface markers (Fig. 2B).

Other promising tools for the ROS and RNS detection in HSCs are fluorescence resonance energy transfer (FRET)-based redox sensors. In FRET, energy is transferred from a light-excited fluorophore (donor) to a longer-wavelength fluorophore (acceptor), whose absorption spectrum overlaps with the emission spectrum of the donor (described in detail by Lukyanov and Belousov, 2014). The main advantage of FRET-based probes lies in the fact that the ratio between the two wavelengths is measured. Unfortunately, these sensors exhibit a low dynamic range and are pH sensitive. Nevertheless, FRET probes have proven very useful and can also be used to probe subcellular compartments or to measure NADPH-oxidase-generated H2O2 (Enyedi et al., 2013; Kolossov et al., 2008). However, to date, FRET-based redox sensors have not been used in vivo.

It is of particular interest to measure ROS levels (e.g. H2O2) under physiological or stress conditions in a hematopoietic niche, as this would help to shed light on the relationship between the state of stem cells and the microenvironment in which they reside. This has been recently achieved for the measurement of the absolute pO2 concentration in the bone marrow hematopoietic niche by using a metallo-porphyrin-based two-photon-enhanced platinum-porphyrin phosphorescent oxygen-sensitive probe (PtP-C343) (Spencer et al., 2014).

Genetically encoded fluorescent probes or boronate-based fluorescent probes have been used to detect H2O2 in various tissues (Bilan et al., 2013; Logan et al., 2014; Love et al., 2013) but not in bone marrow. We believe that their use for H2O2 detection in bone marrow should be considered. Another possibility is the use of nanoparticle-based sensors, which have shown to be useful for ROS detection in whole-animal studies (Uusitalo and Hempel, 2012). Nanoparticles can be conjugated with different moieties for their targeting to certain cells or subcellular compartments (Uusitalo and Hempel, 2012). Recently, intravenous injection of specially designed, liver-targeting semiconducting polymer nanoparticles enabled the simultaneous detection of fluorescence and chemiluminescence in real-time to discriminate between ONOO and H2O2 generation (Shuhendler et al., 2014), making this a particular promising approach for the detection of specific ROS in HSCs. However, further studies are needed to determine whether these probes can be used in HSCs.

In view of the data demonstrating that an anaerobic metabolic character is related to the maintenance of stemness, methods to accurately measure the levels of ROS and RNS in stem cells are of paramount importance. These highly reactive molecules have regulatory roles in energy metabolism that might impact on the HSC fate. Current evidence indicates that variations in the levels of ROS and RNS affect the balance between HSC self-renewal and commitment. As discussed above for commonly used techniques for the measurement of reactive species, there are clear imprecisions in the currently used approaches that must be overcome in order to help us to better understand how ROS and RNS impact on HSC biology. We suggest here that, by combining new advances in the detection of specific ROS and RNS with protocols that allow to achieve a higher enrichment of HSCs, it will be possible to confirm and elucidate any of functional roles of ROS and RNS in stem cell maintenance.

We are thankful to Mrs Elisabeth Doutreloux-Volkmann (EFS-Aqli, Bordeaux), Dr Jacqueline R. Wyatt (Wyatt Scientific Editing Service) and Mr Jerold Morgan (Biorad Laboratories, Seattle) for the language corrections.

Author contributions

M.V.L. and Z.I. designed and wrote the manuscript.

Funding

This work was funded by Etablissement Français du Sang, Aquitaine-Limousin, Bordeaux, France.

Abbas
,
H. A.
,
Maccio
,
D. R.
,
Coskun
,
S.
,
Jackson
,
J. G.
,
Hazen
,
A. L.
,
Sills
,
T. M.
,
You
,
M. J.
,
Hirschi
,
K. K.
and
Lozano
,
G.
(
2010
).
Mdm2 is required for survival of hematopoietic stem cells/progenitors via dampening of ROS-induced p53 activity
.
Cell Stem Cell
7
,
606
-
617
.
Albrecht
,
S. C.
,
Barata
,
A. G.
,
Grosshans
,
J.
,
Teleman
,
A. A.
and
Dick
,
T. P.
(
2011
).
In vivo mapping of hydrogen peroxide and oxidized glutathione reveals chemical and regional specificity of redox homeostasis
.
Cell Metab.
14
,
819
-
829
.
Aleksinskaya
,
M. A.
,
van Faassen
,
E. E. H.
,
Nelissen
,
J.
,
Janssen
,
B. J. A.
,
De Mey
,
J. G. R.
,
Hanemaaijer
,
R.
,
Rabelink
,
T.
and
van Zonneveld
,
A. J.
(
2013
).
Identification of free nitric oxide radicals in rat bone marrow: implications for progenitor cell mobilization in hypertension
.
PLoS ONE
8
,
e57761
.
Alfadda
,
A. A.
and
Sallam
,
R. M.
(
2012
).
Reactive oxygen species in health and disease
.
J. Biomed. Biotechnol.
2012
,
936486
.
Aruoma
,
O. I.
,
Halliwell
,
B.
,
Hoey
,
B. M.
and
Butler
,
J.
(
1989
).
The antioxidant action of N-acetylcysteine: its reaction with hydrogen peroxide, hydroxyl radical, superoxide, and hypochlorous acid
.
Free Radic. Biol. Med.
6
,
593
-
597
.
Bigarella
,
C. L.
,
Liang
,
R.
and
Ghaffari
,
S.
(
2014
).
Stem cells and the impact of ROS signaling
.
Development
141
,
4206
-
4218
.
Bilan
,
D. S.
,
Pase
,
L.
,
Joosen
,
L.
,
Gorokhovatsky
,
A. Y.
,
Ermakova
,
Y. G.
,
Gadella
,
T. W. J.
,
Grabher
,
C.
,
Schultz
,
C.
,
Lukyanov
,
S.
and
Belousov
,
V. V.
(
2013
).
HyPer-3: a genetically encoded H(2)O(2) probe with improved performance for ratiometric and fluorescence lifetime imaging
.
ACS Chem. Biol.
8
,
535
-
542
.
Bonafè
,
F.
,
Guarnieri
,
C.
and
Muscari
,
C.
(
2015
).
Nitric oxide regulates multiple functions and fate of adult progenitor and stem cells
.
J. Physiol. Biochem.
71
,
141
-
153
.
Bryder
,
D.
,
Rossi
,
D. J.
and
Weissman
,
I. L.
(
2006
).
Hematopoietic stem cells: the paradigmatic tissue-specific stem cell
.
Am. J. Pathol.
169
,
338
-
346
.
Burkitt
,
M. J.
and
Wardman
,
P.
(
2001
).
Cytochrome C is a potent catalyst of dichlorofluorescin oxidation: implications for the role of reactive oxygen species in apoptosis
.
Biochem. Biophys. Res. Commun.
282
,
329
-
333
.
Cam
,
H.
,
Easton
,
J. B.
,
High
,
A.
and
Houghton
,
P. J.
(
2010
).
mTORC1 signaling under hypoxic conditions is controlled by ATM-dependent phosphorylation of HIF-1alpha
.
Mol. Cell
40
,
509
-
520
.
Chen
,
C.
,
Liu
,
Y.
,
Liu
,
R.
,
Ikenoue
,
T.
,
Guan
,
K.-L.
,
Liu
,
Y.
and
Zheng
,
P.
(
2008a
).
TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species
.
J. Exp. Med.
205
,
2397
-
2408
.
Chen
,
J.
,
Ellison
,
F. M.
,
Keyvanfar
,
K.
,
Omokaro
,
S. O.
,
Desierto
,
M. J.
,
Eckhaus
,
M. A.
and
Young
,
N. S.
(
2008b
).
Enrichment of hematopoietic stem cells with SLAM and LSK markers for the detection of hematopoietic stem cell function in normal and Trp53 null mice
.
Exp. Hematol.
36
,
1236
-
1243
.
Cho
,
J.
,
Yusuf
,
R.
,
Kook
,
S.
,
Attar
,
E.
,
Lee
,
D.
,
Park
,
B.
,
Cheng
,
T.
,
Scadden
,
D. T.
and
Lee
,
B. C.
(
2014
).
Purinergic P2Y(1)(4) receptor modulates stress-induced hematopoietic stem/progenitor cell senescence
.
J. Clin. Invest.
124
,
3159
-
3171
.
Choi
,
H.-J.
,
Kim
,
S.-J.
,
Mukhopadhyay
,
P.
,
Cho
,
S.
,
Woo
,
J.-R.
,
Storz
,
G.
and
Ryu
,
S.-E.
(
2001
).
Structural basis of the redox switch in the OxyR transcription factor
.
Cell
105
,
103
-
113
.
Chuikov
,
S.
,
Levi
,
B. P.
,
Smith
,
M. L.
and
Morrison
,
S. J.
(
2010
).
Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress
.
Nat. Cell Biol.
12
,
999
-
1006
.
Cochemé
,
H. M.
,
Quin
,
C.
,
McQuaker
,
S. J.
,
Cabreiro
,
F.
,
Logan
,
A.
,
Prime
,
T. A.
,
Abakumova
,
I.
,
Patel
,
J. V.
,
Fearnley
,
I. M.
,
James
,
A. M.
, et al. 
(
2011
).
Measurement of H2O2 within living Drosophila during aging using a ratiometric mass spectrometry probe targeted to the mitochondrial matrix
.
Cell Metab.
13
,
340
-
350
.
Čokić
,
V. P.
,
Beleslin-Čokić
,
B. B.
,
Smith
,
R. D.
,
Economou
,
A. P.
,
Wahl
,
L. M.
,
Noguchi
,
C. T.
and
Schechter
,
A. N.
(
2009
).
Stimulated stromal cells induce gamma-globin gene expression in erythroid cells via nitric oxide production
.
Exp. Hematol.
37
,
1230
-
1237
.
Cooper
,
C. E.
(
1999
).
Nitric oxide and iron proteins
.
Biochim. Biophys. Acta
1411
,
290
-
309
.
D'Autréaux
,
B.
and
Toledano
,
M. B.
(
2007
).
ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis
.
Nat. Rev. Mol. Cell Biol.
8
,
813
-
824
.
Dick
,
J. E.
,
Bhatia
,
M.
,
Gan
,
O.
,
Kapp
,
U.
and
Wang
,
J. C. Y.
(
1997
).
Assay of human stem cells by repopulation of NOD/SCID mice
.
Stem Cells
15
Suppl. 1
,
199
-
207
;
discussion 204-7
.
Dickinson
,
B. C.
,
Tang
,
Y.
,
Chang
,
Z.
and
Chang
,
C. J.
(
2011
).
A nuclear-localized fluorescent hydrogen peroxide probe for monitoring sirtuin-mediated oxidative stress responses in vivo
.
Chem. Biol.
18
,
943
-
948
.
Dickinson
,
B. C.
,
Lin
,
V. S.
and
Chang
,
C. J.
(
2013
).
Preparation and use of MitoPY1 for imaging hydrogen peroxide in mitochondria of live cells
.
Nat. Protoc.
8
,
1249
-
1259
.
Ditch
,
S.
and
Paull
,
T. T.
(
2012
).
The ATM protein kinase and cellular redox signaling: beyond the DNA damage response
.
Trends Biochem. Sci.
37
,
15
-
22
.
Dooley
,
C. T.
,
Dore
,
T. M.
,
Hanson
,
G. T.
,
Jackson
,
W. C.
,
Remington
,
S. J.
and
Tsien
,
R. Y.
(
2004
).
Imaging dynamic redox changes in mammalian cells with green fluorescent protein indicators
.
J. Biol. Chem.
279
,
22284
-
22293
.
Enyedi
,
B.
,
Zana
,
M.
,
Donkó
,
A.
and
Geiszt
,
M.
(
2013
).
Spatial and temporal analysis of NADPH oxidase-generated hydrogen peroxide signals by novel fluorescent reporter proteins
.
Antioxid. Redox Signal.
19
,
523
-
534
.
Epperly
,
M. W.
,
Cao
,
S.
,
Zhang
,
X.
,
Franicola
,
D.
,
Shen
,
H.
,
Greenberger
,
E. E.
,
Epperly
,
L. D.
and
Greenberger
,
J. S.
(
2007
).
Increased longevity of hematopoiesis in continuous bone marrow cultures derived from NOS1 (nNOS, mtNOS) homozygous recombinant negative mice correlates with radioresistance of hematopoietic and marrow stromal cells
.
Exp. Hematol.
35
,
137
-
145
.
Fan
,
J.
,
Cai
,
H.
and
Tan
,
W.-S.
(
2007
).
Role of the plasma membrane ROS-generating NADPH oxidase in CD34+ progenitor cells preservation by hypoxia
.
J. Biotechnol.
130
,
455
-
462
.
Ferrer-Sueta
,
G.
and
Radi
,
R.
(
2009
).
Chemical biology of peroxynitrite: kinetics, diffusion, and radicals
.
ACS Chem. Biol.
4
,
161
-
177
.
Finkel
,
T.
(
2011
).
Signal transduction by reactive oxygen species
.
J. Cell Biol.
194
,
7
-
15
.
Folkes
,
L. K.
,
Patel
,
K. B.
,
Wardman
,
P.
and
Wrona
,
M.
(
2009
).
Kinetics of reaction of nitrogen dioxide with dihydrorhodamine and the reaction of the dihydrorhodamine radical with oxygen: implications for quantifying peroxynitrite formation in cells
.
Arch. Biochem. Biophys.
484
,
122
-
126
.
Gorbunov
,
N. V.
,
Pogue-Geile
,
K. L.
,
Epperly
,
M. W.
,
Bigbee
,
W. L.
,
Draviam
,
R.
,
Day
,
B. W.
,
Wald
,
N.
,
Watkins
,
S. C.
and
Greenberger
,
J. S.
(
2000
).
Activation of the nitric oxide synthase 2 pathway in the response of bone marrow stromal cells to high doses of ionizing radiation
.
Radiat. Res.
154
,
73
-
86
.
Gorris
,
H. H.
and
Walt
,
D. R.
(
2009
).
Mechanistic aspects of horseradish peroxidase elucidated through single-molecule studies
.
J. Am. Chem. Soc.
131
,
6277
-
6282
.
Guzy
,
R. D.
and
Schumacker
,
P. T.
(
2006
).
Oxygen sensing by mitochondria at complex III: the paradox of increased reactive oxygen species during hypoxia
.
Exp. Physiol.
91
,
807
-
819
.
Han
,
M.-K.
,
Song
,
E.-K.
,
Guo
,
Y.
,
Ou
,
X.
,
Mantel
,
C.
and
Broxmeyer
,
H. E.
(
2008
).
SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization
.
Cell Stem Cell
2
,
241
-
251
.
Hao
,
Y.
,
Cheng
,
D.
,
Ma
,
Y.
,
Zhou
,
W.
and
Wang
,
Y.
(
2011
).
The relationship between oxygen concentration, reactive oxygen species and the biological characteristics of human bone marrow hematopoietic stem cells
.
Transplant. Proc.
43
,
2755
-
2761
.
Hole
,
P. S.
,
Pearn
,
L.
,
Tonks
,
A. J.
,
James
,
P. E.
,
Burnett
,
A. K.
,
Darley
,
R. L.
and
Tonks
,
A.
(
2010
).
Ras-induced reactive oxygen species promote growth factor-independent proliferation in human CD34+ hematopoietic progenitor cells
.
Blood
115
,
1238
-
1246
.
Honma
,
N.
,
Mori
,
S.
,
Zhou
,
H.
,
Ikeda
,
S.
,
Mieno
,
M. N.
,
Tanaka
,
N.
,
Takubo
,
K.
,
Arai
,
T.
,
Sawabe
,
M.
,
Muramatsu
,
M.
, et al. 
(
2013
).
Association between estrogen receptor-beta dinucleotide repeat polymorphism and incidence of femoral fracture
.
J. Bone Miner. Metab.
31
,
96
-
101
.
Hosokawa
,
K.
,
Arai
,
F.
,
Yoshihara
,
H.
,
Nakamura
,
Y.
,
Gomei
,
Y.
,
Iwasaki
,
H.
,
Miyamoto
,
K.
,
Shima
,
H.
,
Ito
,
K.
and
Suda
,
T.
(
2007
).
Function of oxidative stress in the regulation of hematopoietic stem cell-niche interaction
.
Biochem. Biophys. Res. Commun.
363
,
578
-
583
.
Ito
,
K.
,
Hirao
,
A.
,
Arai
,
F.
,
Matsuoka
,
S.
,
Takubo
,
K.
,
Hamaguchi
,
I.
,
Nomiyama
,
K.
,
Hosokawa
,
K.
,
Sakurada
,
K.
,
Nakagata
,
N.
, et al. 
(
2004
).
Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells
.
Nature
431
,
997
-
1002
.
Ito
,
K.
,
Hirao
,
A.
,
Arai
,
F.
,
Takubo
,
K.
,
Matsuoka
,
S.
,
Miyamoto
,
K.
,
Ohmura
,
M.
,
Naka
,
K.
,
Hosokawa
,
K.
,
Ikeda
,
Y.
, et al. 
(
2006
).
Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells
.
Nat. Med.
12
,
446
-
451
.
Ivanovic
,
Z.
,
Duchez
,
P.
,
Chevaleyre
,
J.
,
Vlaski
,
M.
,
Lafarge
,
X.
,
Dazey
,
B.
,
Robert-Richard
,
E.
,
Mazurier
,
F.
and
Boiron
,
J.-M.
(
2011
).
Clinical-scale cultures of cord blood CD34(+) cells to amplify committed progenitors and maintain stem cell activity
.
Cell Transplant.
20
,
1453
-
1463
.
Jang
,
Y.-Y.
and
Sharkis
,
S. J.
(
2007
).
A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche
.
Blood
110
,
3056
-
3063
.
Jung
,
H.
,
Kim
,
M. J.
,
Kim
,
D. O.
,
Kim
,
W. S.
,
Yoon
,
S.-J.
,
Park
,
Y.-J.
,
Yoon
,
S. R.
,
Kim
,
T.-D.
,
Suh
,
H.-W.
,
Yun
,
S.
, et al. 
(
2013
).
TXNIP maintains the hematopoietic cell pool by switching the function of p53 under oxidative stress
.
Cell Metab.
18
,
75
-
85
.
Juntilla
,
M. M.
,
Patil
,
V. D.
,
Calamito
,
M.
,
Joshi
,
R. P.
,
Birnbaum
,
M. J.
and
Koretzky
,
G. A.
(
2010
).
AKT1 and AKT2 maintain hematopoietic stem cell function by regulating reactive oxygen species
.
Blood
115
,
4030
-
4038
.
Kalyanaraman
,
B.
,
Darley-Usmar
,
V.
,
Davies
,
K. J. A.
,
Dennery
,
P. A.
,
Forman
,
H. J.
,
Grisham
,
M. B.
,
Mann
,
G. E.
,
Moore
,
K.
,
Roberts
,
L. J.
, II
and
Ischiropoulos
,
H.
(
2012
).
Measuring reactive oxygen and nitrogen species with fluorescent probes: challenges and limitations
.
Free Radic. Biol. Med.
52
,
1
-
6
.
Karlsson
,
M.
,
Kurz
,
T.
,
Brunk
,
U. T.
,
Nilsson
,
S. E.
and
Frennesson
,
C. I.
(
2010
).
What does the commonly used DCF test for oxidative stress really show?
Biochem. J.
428
,
183
-
190
.
Kiel
,
M. J.
,
Yilmaz
,
Ö. H.
,
Iwashita
,
T.
,
Yilmaz
,
O. H.
,
Terhorst
,
C.
and
Morrison
,
S. J.
(
2005
).
SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells
.
Cell
121
,
1109
-
1121
.
Kiel
,
M. J.
,
Yilmaz
,
O. H.
and
Morrison
,
S. J.
(
2008
).
CD150- cells are transiently reconstituting multipotent progenitors with little or no stem cell activity
.
Blood
111
,
4413
-
4414
;
author reply 4414-5
.
Kojer
,
K.
,
Bien
,
M.
,
Gangel
,
H.
,
Morgan
,
B.
,
Dick
,
T. P.
and
Riemer
,
J.
(
2012
).
Glutathione redox potential in the mitochondrial intermembrane space is linked to the cytosol and impacts the Mia40 redox state
.
EMBO J.
31
,
3169
-
3182
.
Kolossov
,
V. L.
,
Spring
,
B. Q.
,
Sokolowski
,
A.
,
Conour
,
J. E.
,
Clegg
,
R. M.
,
Kenis
,
P. J.
and
Gaskins
,
H. R.
(
2008
).
Engineering redox-sensitive linkers for genetically encoded FRET-based biosensors
.
Exp. Biol. Med. (Maywood)
233
,
238
-
248
.
Lin
,
V. S.
,
Dickinson
,
B. C.
and
Chang
,
C. J.
(
2013
).
Boronate-based fluorescent probes: imaging hydrogen peroxide in living systems
.
Methods Enzymol.
526
,
19
-
43
.
Lippert
,
A. R.
,
Van de Bittner
,
G. C.
and
Chang
,
C. J.
(
2011
).
Boronate oxidation as a bioorthogonal reaction approach for studying the chemistry of hydrogen peroxide in living systems
.
Acc. Chem. Res.
44
,
793
-
804
.
Liu
,
J.
,
Cao
,
L.
,
Chen
,
J.
,
Song
,
S.
,
Lee
,
I. H.
,
Quijano
,
C.
,
Liu
,
H.
,
Keyvanfar
,
K.
,
Chen
,
H.
,
Cao
,
L.-Y.
, et al. 
(
2009
).
Bmi1 regulates mitochondrial function and the DNA damage response pathway
.
Nature
459
,
387
-
392
.
Logan
,
A.
,
Shabalina
,
I. G.
,
Prime
,
T. A.
,
Rogatti
,
S.
,
Kalinovich
,
A. V.
,
Hartley
,
R. C.
,
Budd
,
R. C.
,
Cannon
,
B.
and
Murphy
,
M. P.
(
2014
).
In vivo levels of mitochondrial hydrogen peroxide increase with age in mtDNA mutator mice
.
Aging Cell
13
,
765
-
768
.
Love
,
N. R.
,
Chen
,
Y.
,
Ishibashi
,
S.
,
Kritsiligkou
,
P.
,
Lea
,
R.
,
Koh
,
Y.
,
Gallop
,
J. L.
,
Dorey
,
K.
and
Amaya
,
E.
(
2013
).
Amputation-induced reactive oxygen species are required for successful Xenopus tadpole tail regeneration
.
Nat. Cell Biol.
15
,
222
-
228
.
Lukyanov
,
K. A.
and
Belousov
,
V. V.
(
2014
).
Genetically encoded fluorescent redox sensors
.
Biochim. Biophys. Acta
1840
,
745
-
756
.
Marchesi
,
E.
,
Rota
,
C.
,
Fann
,
Y. C.
,
Chignell
,
C. F.
and
Mason
,
R. P.
(
1999
).
Photoreduction of the fluorescent dye 2′-7′-dichlorofluorescein: a spin trapping and direct electron spin resonance study with implications for oxidative stress measurements
.
Free Radic. Biol. Med.
26
,
148
-
161
.
Marozkina
,
N. V.
and
Gaston
,
B.
(
2012
).
S-Nitrosylation signaling regulates cellular protein interactions
.
Biochim. Biophys. Acta
1820
,
722
-
729
.
Maryanovich
,
M.
,
Oberkovitz
,
G.
,
Niv
,
H.
,
Vorobiyov
,
L.
,
Zaltsman
,
Y.
,
Brenner
,
O.
,
Lapidot
,
T.
,
Jung
,
S.
and
Gross
,
A.
(
2012
).
The ATM–BID pathway regulates quiescence and survival of haematopoietic stem cells
.
Nat. Cell Biol.
14
,
535
-
541
.
Michurina
,
T.
,
Krasnov
,
P.
,
Balazs
,
A.
,
Nakaya
,
N.
,
Vasilieva
,
T.
,
Kuzin
,
B.
,
Khrushchov
,
N.
,
Mulligan
,
R. C.
and
Enikolopov
,
G.
(
2004
).
Nitric oxide is a regulator of hematopoietic stem cell activity
.
Mol. Ther.
10
,
241
-
248
.
Miyamoto
,
K.
,
Araki
,
K. Y.
,
Naka
,
K.
,
Arai
,
F.
,
Takubo
,
K.
,
Yamazaki
,
S.
,
Matsuoka
,
S.
,
Miyamoto
,
T.
,
Ito
,
K.
,
Ohmura
,
M.
, et al. 
(
2007
).
Foxo3a is essential for maintenance of the hematopoietic stem cell pool
.
Cell Stem Cell
1
,
101
-
112
.
Naka
,
K.
,
Muraguchi
,
T.
,
Hoshii
,
T.
and
Hirao
,
A.
(
2008
).
Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells
.
Antioxid. Redox Signal.
10
,
1883
-
1894
.
Nathan
,
C.
and
Cunningham-Bussel
,
A.
(
2013
).
Beyond oxidative stress: an immunologist's guide to reactive oxygen species
.
Nat. Rev. Immunol.
13
,
349
-
361
.
Navarro-Yepes
,
J.
,
Burns
,
M.
,
Anandhan
,
A.
,
Khalimonchuk
,
O.
,
del Razo
,
L. M.
,
Quintanilla-Vega
,
B.
,
Pappa
,
A.
,
Panayiotidis
,
M. I.
and
Franco
,
R.
(
2014
).
Oxidative stress, redox signaling, and autophagy: cell death versus survival
.
Antioxid. Redox Signal.
21
,
66
-
85
.
Nogueira-Pedro
,
A.
,
Dias
,
C. C.
,
Segreto
,
H. R.
,
Addios
,
P. C.
,
Lungato
,
L.
,
D'Almeida
,
V.
,
Barros
,
C. C.
,
Higa
,
E. M.
,
Buri
,
M. V.
,
Paredes-Gamero
,
E. J.
, et al. 
(
2014
).
Nitric oxide-induced murine hematopoietic stem cell fate involves multiple signaling proteins, gene expression, and redox modulation
.
Stem Cells
32
,
2949
-
2960
.
Oguro
,
H.
,
Ding
,
L.
and
Morrison
,
S. J.
(
2013
).
SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors
.
Cell Stem Cell
13
,
102
-
116
.
Osawa
,
M.
,
Hanada
,
K.
,
Hamada
,
H.
and
Nakauchi
,
H.
(
1996
).
Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell
.
Science
273
,
242
-
245
.
Østergaard
,
H.
,
Henriksen
,
A.
,
Hansen
,
F. G.
and
Winther
,
J. R.
(
2001
).
Shedding light on disulfide bond formation: engineering a redox switch in green fluorescent protein
.
EMBO J.
20
,
5853
-
5862
.
Pase
,
L.
,
Layton
,
J. E.
,
Wittmann
,
C.
,
Ellett
,
F.
,
Nowell
,
C. J.
,
Reyes-Aldasoro
,
C. C.
,
Varma
,
S.
,
Rogers
,
K. L.
,
Hall
,
C. J.
,
Keightley
,
M. C.
, et al. 
(
2012
).
Neutrophil-delivered myeloperoxidase dampens the hydrogen peroxide burst after tissue wounding in zebrafish
.
Curr. Biol.
22
,
1818
-
1824
.
Pazhanisamy
,
S. K.
,
Li
,
H.
,
Wang
,
Y.
,
Batinic-Haberle
,
I.
and
Zhou
,
D.
(
2011
).
NADPH oxidase inhibition attenuates total body irradiation-induced haematopoietic genomic instability
.
Mutagenesis
26
,
431
-
435
.
Piccoli
,
C.
,
Ria
,
R.
,
Scrima
,
R.
,
Cela
,
O.
,
D'Aprile
,
A.
,
Boffoli
,
D.
,
Falzetti
,
F.
,
Tabilio
,
A.
and
Capitanio
,
N.
(
2005
).
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.
280
,
26467
-
26476
.
Piccoli
,
C.
,
D'Aprile
,
A.
,
Ripoli
,
M.
,
Scrima
,
R.
,
Boffoli
,
D.
,
Tabilio
,
A.
and
Capitanio
,
N.
(
2007a
).
The hypoxia-inducible factor is stabilized in circulating hematopoietic stem cells under normoxic conditions
.
FEBS Lett.
581
,
3111
-
3119
.
Piccoli
,
C.
,
D'Aprile
,
A.
,
Ripoli
,
M.
,
Scrima
,
R.
,
Lecce
,
L.
,
Boffoli
,
D.
,
Tabilio
,
A.
and
Capitanio
,
N.
(
2007b
).
Bone-marrow derived hematopoietic stem/progenitor cells express multiple isoforms of NADPH oxidase and produce constitutively reactive oxygen species
.
Biochem. Biophys. Res. Commun.
353
,
965
-
972
.
Reykdal
,
S.
,
Abboud
,
C.
and
Liesveld
,
J.
(
1999
).
Effect of nitric oxide production and oxygen tension on progenitor preservation in ex vivo culture
.
Exp. Hematol.
27
,
441
-
450
.
Robinson
,
K. M.
,
Janes
,
M. S.
,
Pehar
,
M.
,
Monette
,
J. S.
,
Ross
,
M. F.
,
Hagen
,
T. M.
,
Murphy
,
M. P.
and
Beckman
,
J. S.
(
2006
).
Selective fluorescent imaging of superoxide in vivo using ethidium-based probes
.
Proc. Natl. Acad. Sci. USA
103
,
15038
-
15043
.
Rouault-Pierre
,
K.
,
Lopez-Onieva
,
L.
,
Foster
,
K.
,
Anjos-Afonso
,
F.
,
Lamrissi-Garcia
,
I.
,
Serrano-Sanchez
,
M.
,
Mitter
,
R.
,
Ivanovic
,
Z.
,
de Verneuil
,
H.
,
Gribben
,
J.
, et al. 
(
2013
).
HIF-2alpha protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress
.
Cell Stem Cell
13
,
549
-
563
.
Sattler
,
M.
,
Winkler
,
T.
,
Verma
,
S.
,
Byrne
,
C. H.
,
Shrikhande
,
G.
,
Salgia
,
R.
and
Griffin
,
J. D.
(
1999
).
Hematopoietic growth factors signal through the formation of reactive oxygen species
.
Blood
93
,
2928
-
2935
.
Scharenberg
,
C. W.
,
Harkey
,
M. A.
and
Torok-Storb
,
B.
(
2002
).
The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors
.
Blood
99
,
507
-
512
.
Shao
,
L.
,
Li
,
H.
,
Pazhanisamy
,
S. K.
,
Meng
,
A.
,
Wang
,
Y.
and
Zhou
,
D.
(
2011
).
Reactive oxygen species and hematopoietic stem cell senescence
.
Int. J. Hematol.
94
,
24
-
32
.
Shao
,
L.
,
Feng
,
W.
,
Li
,
H.
,
Gardner
,
D.
,
Luo
,
Y.
,
Wang
,
Y.
,
Liu
,
L.
,
Meng
,
A.
,
Sharpless
,
N. E.
and
Zhou
,
D.
(
2014
).
Total body irradiation causes long-term mouse BM injury via induction of HSC premature senescence in an Ink4a- and Arf-independent manner
.
Blood
123
,
3105
-
3115
.
Shimi
,
T.
,
Butin-Israeli
,
V.
,
Adam
,
S. A.
,
Hamanaka
,
R. B.
,
Goldman
,
A. E.
,
Lucas
,
C. A.
,
Shumaker
,
D. K.
,
Kosak
,
S. T.
,
Chandel
,
N. S.
and
Goldman
,
R. D.
(
2011
).
The role of nuclear lamin B1 in cell proliferation and senescence
.
Genes Dev.
25
,
2579
-
2593
.
Shuhendler
,
A. J.
,
Pu
,
K.
,
Cui
,
L.
,
Uetrecht
,
J. P.
and
Rao
,
J.
(
2014
).
Real-time imaging of oxidative and nitrosative stress in the liver of live animals for drug-toxicity testing
.
Nat. Biotechnol.
32
,
373
-
380
.
Sikora
,
A.
,
Zielonka
,
J.
,
Lopez
,
M.
,
Joseph
,
J.
and
Kalyanaraman
,
B.
(
2009
).
Direct oxidation of boronates by peroxynitrite: mechanism and implications in fluorescence imaging of peroxynitrite
.
Free Radic. Biol. Med.
47
,
1401
-
1407
.
Simsek
,
T.
,
Kocabas
,
F.
,
Zheng
,
J.
,
Deberardinis
,
R. J.
,
Mahmoud
,
A. I.
,
Olson
,
E. N.
,
Schneider
,
J. W.
,
Zhang
,
C. C.
and
Sadek
,
H. A.
(
2010
).
The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche
.
Cell Stem Cell
7
,
380
-
390
.
Skinner
,
A. M.
,
O'Neill
,
S. L.
,
Grompe
,
M.
and
Kurre
,
P.
(
2008
).
CXCR4 induction in hematopoietic progenitor cells from Fanca(−/−), -c(−/−), and -d2(−/−) mice
.
Exp. Hematol.
36
,
273
-
282
.
Spencer
,
J. A.
,
Ferraro
,
F.
,
Roussakis
,
E.
,
Klein
,
A.
,
Wu
,
J.
,
Runnels
,
J. M.
,
Zaher
,
W.
,
Mortensen
,
L. J.
,
Alt
,
C.
,
Turcotte
,
R.
, et al. 
(
2014
).
Direct measurement of local oxygen concentration in the bone marrow of live animals
.
Nature
508
,
269
-
273
.
Stamler
,
J. S.
,
Singel
,
D. J.
and
Loscalzo
,
J.
(
1992
).
Biochemistry of nitric oxide and its redox-activated forms
.
Science
258
,
1898
-
1902
.
Storz
,
P.
(
2011
).
Forkhead homeobox type O transcription factors in the responses to oxidative stress
.
Antioxid. Redox Signal.
14
,
593
-
605
.
Suda
,
T.
,
Takubo
,
K.
and
Semenza
,
G. L.
(
2011
).
Metabolic regulation of hematopoietic stem cells in the hypoxic niche
.
Cell Stem Cell
9
,
298
-
310
.
Szilvassy
,
S. J.
,
Humphries
,
R. K.
,
Lansdorp
,
P. M.
,
Eaves
,
A. C.
and
Eaves
,
C. J.
(
1990
).
Quantitative assay for totipotent reconstituting hematopoietic stem cells by a competitive repopulation strategy
.
Proc. Natl. Acad. Sci. USA
87
,
8736
-
8740
.
Takubo
,
K.
,
Goda
,
N.
,
Yamada
,
W.
,
Iriuchishima
,
H.
,
Ikeda
,
E.
,
Kubota
,
Y.
,
Shima
,
H.
,
Johnson
,
R. S.
,
Hirao
,
A.
,
Suematsu
,
M.
, et al. 
(
2010
).
Regulation of the HIF-1alpha level is essential for hematopoietic stem cells
.
Cell Stem Cell
7
,
391
-
402
.
Takubo
,
K.
,
Nagamatsu
,
G.
,
Kobayashi
,
C. I.
,
Nakamura-Ishizu
,
A.
,
Kobayashi
,
H.
,
Ikeda
,
E.
,
Goda
,
N.
,
Rahimi
,
Y.
,
Johnson
,
R. S.
,
Soga
,
T.
, et al. 
(
2013
).
Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells
.
Cell Stem Cell
12
,
49
-
61
.
Taniguchi Ishikawa
,
E.
,
Gonzalez-Nieto
,
D.
,
Ghiaur
,
G.
,
Dunn
,
S. K.
,
Ficker
,
A. M.
,
Murali
,
B.
,
Madhu
,
M.
,
Gutstein
,
D. E.
,
Fishman
,
G. I.
,
Barrio
,
L. C.
, et al. 
(
2012
).
Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells
.
Proc. Natl. Acad. Sci. USA
109
,
9071
-
9076
.
Teranishi
,
K.
(
2007
).
Luminescence of imidazo[1,2-a]pyrazin-3(7H)-one compounds
.
Bioorg. Chem.
35
,
82
-
111
.
Tesio
,
M.
,
Golan
,
K.
,
Corso
,
S.
,
Giordano
,
S.
,
Schajnovitz
,
A.
,
Vagima
,
Y.
,
Shivtiel
,
S.
,
Kalinkovich
,
A.
,
Caione
,
L.
,
Gammaitoni
,
L.
, et al. 
(
2011
).
Enhanced c-Met activity promotes G-CSF-induced mobilization of hematopoietic progenitor cells via ROS signaling
.
Blood
117
,
419
-
428
.
The
,
A. C.
,
Aad
,
G.
,
Abajyan
,
T.
,
Abbott
,
B.
,
Abdallah
,
J.
,
Abdel Khalek
,
S.
,
Abdelalim
,
A. A.
,
Abdinov
,
O.
,
Aben
,
R.
,
Abi
,
B.
, et al. 
(
2013
).
Search for a light charged Higgs boson in the decay channel [Formula: see text] in [Formula: see text] events using collisions at [Formula: see text] with the ATLAS detector
.
Eur. Phys. J. C Part Fields
73
,
2465
.
Thompson
,
J. S.
,
Asmis
,
R.
,
Chu
,
Y.
,
Glass
,
J.
,
Nelson
,
B.
and
Brown
,
S. A.
(
2008
).
Amifostine prior to lethal irradiation prevents allogeneic bone marrow engraftment in mice
.
Bone Marrow Transplant.
41
,
927
-
934
.
Till
,
J. E.
,
McCulloch
,
E. A.
and
Siminovitch
,
L.
(
1964
).
A stochastic model of stem cell proliferation, based on the growth of spleen colony-forming cells
.
Proc. Natl. Acad. Sci. USA
51
,
29
-
36
.
Tothova
,
Z.
,
Kollipara
,
R.
,
Huntly
,
B. J.
,
Lee
,
B. H.
,
Castrillon
,
D. H.
,
Cullen
,
D. E.
,
McDowell
,
E. P.
,
Lazo-Kallanian
,
S.
,
Williams
,
I. R.
,
Sears
,
C.
, et al. 
(
2007
).
FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress
.
Cell
128
,
325
-
339
.
Unwin
,
R. D.
,
Smith
,
D. L.
,
Blinco
,
D.
,
Wilson
,
C. L.
,
Miller
,
C. J.
,
Evans
,
C. A.
,
Jaworska
,
E.
,
Baldwin
,
S. A.
,
Barnes
,
K.
,
Pierce
,
A.
, et al. 
(
2006
).
Quantitative proteomics reveals posttranslational control as a regulatory factor in primary hematopoietic stem cells
.
Blood
107
,
4687
-
4694
.
Uusitalo
,
L. M.
and
Hempel
,
N.
(
2012
).
Recent advances in intracellular and in vivo ROS sensing: focus on nanoparticle and nanotube applications
.
Int. J. Mol. Sci.
13
,
10660
-
10679
.
Van de Bittner
,
G. C.
,
Dubikovskaya
,
E. A.
,
Bertozzi
,
C. R.
and
Chang
,
C. J.
(
2010
).
In vivo imaging of hydrogen peroxide production in a murine tumor model with a chemoselective bioluminescent reporter
.
Proc. Natl. Acad. Sci. USA
107
,
21316
-
21321
.
Vlaski
,
M.
,
Negroni
,
L.
,
Kovacevic-Filipovic
,
M.
,
Guibert
,
C.
,
de la Grange
,
P. B.
,
Rossignol
,
R.
,
Chevaleyre
,
J.
,
Duchez
,
P.
,
Lafarge
,
X.
,
Praloran
,
V.
, et al. 
(
2014
).
Hypoxia/hypercapnia-induced adaptation maintains functional capacity of cord blood stem and progenitor cells at 4°C
.
J. Cell Physiol.
229
,
2153
-
2165
.
Wang
,
Y.
,
Liu
,
L.
,
Pazhanisamy
,
S. K.
,
Li
,
H.
,
Meng
,
A.
and
Zhou
,
D.
(
2010
).
Total body irradiation causes residual bone marrow injury by induction of persistent oxidative stress in murine hematopoietic stem cells
.
Free Radic. Biol. Med.
48
,
348
-
356
.
Wang
,
K.
,
Zhang
,
T.
,
Dong
,
Q.
,
Nice
,
E. C.
,
Huang
,
C.
and
Wei
,
Y.
(
2013
).
Redox homeostasis: the linchpin in stem cell self-renewal and differentiation
.
Cell Death Dis.
4
,
e537
.
Wardman
,
P.
(
2007
).
Fluorescent and luminescent probes for measurement of oxidative and nitrosative species in cells and tissues: progress, pitfalls, and prospects
.
Free Radic. Biol. Med.
43
,
995
-
1022
.
Wilson
,
A.
,
Laurenti
,
E.
,
Oser
,
G.
,
van der Wath
,
R. C.
,
Blanco-Bose
,
W.
,
Jaworski
,
M.
,
Offner
,
S.
,
Dunant
,
C. F.
,
Eshkind
,
L.
,
Bockamp
,
E.
, et al. 
(
2008
).
Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair
.
Cell
135
,
1118
-
1129
.
Winterbourn
,
C. C.
(
2014
).
The challenges of using fluorescent probes to detect and quantify specific reactive oxygen species in living cells
.
Biochim. Biophys. Acta
1840
,
730
-
738
.
Woolley
,
J. F.
,
Naughton
,
R.
,
Stanicka
,
J.
,
Gough
,
D. R.
,
Bhatt
,
L.
,
Dickinson
,
B. C.
,
Chang
,
C. J.
and
Cotter
,
T. G.
(
2012
).
H2O2 production downstream of FLT3 is mediated by p22phox in the endoplasmic reticulum and is required for STAT5 signalling
.
PLoS ONE
7
,
e34050
.
Wrona
,
M.
and
Wardman
,
P.
(
2006
).
Properties of the radical intermediate obtained on oxidation of 2′,7′-dichlorodihydrofluorescein, a probe for oxidative stress
.
Free Radic. Biol. Med.
41
,
657
-
667
.
Wrona
,
M.
,
Patel
,
K.
and
Wardman
,
P.
(
2005
).
Reactivity of 2′,7′-dichlorodihydrofluorescein and dihydrorhodamine 123 and their oxidized forms toward carbonate, nitrogen dioxide, and hydroxyl radicals
.
Free Radic. Biol. Med.
38
,
262
-
270
.
Wullschleger
,
S.
,
Loewith
,
R.
and
Hall
,
M. N.
(
2006
).
TOR signaling in growth and metabolism
.
Cell
124
,
471
-
484
.
Yahata
,
T.
,
Takanashi
,
T.
,
Muguruma
,
Y.
,
Ibrahim
,
A. A.
,
Matsuzawa
,
H.
,
Uno
,
T.
,
Sheng
,
Y.
,
Onizuka
,
M.
,
Ito
,
M.
,
Kato
,
S.
, et al. 
(
2011
).
Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells
.
Blood
118
,
2941
-
2950
.
Yalcin
,
S.
,
Zhang
,
X.
,
Luciano
,
J. P.
,
Mungamuri
,
S. K.
,
Marinkovic
,
D.
,
Vercherat
,
C.
,
Sarkar
,
A.
,
Grisotto
,
M.
,
Taneja
,
R.
and
Ghaffari
,
S.
(
2008
).
Foxo3 is essential for the regulation of ataxia telangiectasia mutated and oxidative stress-mediated homeostasis of hematopoietic stem cells
.
J. Biol. Chem.
283
,
25692
-
25705
.
Yamaguchi
,
M.
and
Kashiwakura
,
I.
(
2013
).
Role of reactive oxygen species in the radiation response of human hematopoietic stem/progenitor cells
.
PLoS One
8
,
e70503
.
Ye
,
X.
,
Kim
,
W.-S.
,
Rubakhin
,
S. S.
and
Sweedler
,
J. V.
(
2004
).
Measurement of nitric oxide by 4,5-diaminofluorescein without interferences
.
Analyst
129
,
1200
-
1205
.
Zhao
,
B.
,
Ranguelova
,
K.
,
Jiang
,
J.
and
Mason
,
R. P.
(
2011
).
Studies on the photosensitized reduction of resorufin and implications for the detection of oxidative stress with Amplex Red
.
Free Radic. Biol. Med.
51
,
153
-
159
.
Zielonka
,
J.
and
Kalyanaraman
,
B.
(
2010
).
Hydroethidine- and MitoSOX-derived red fluorescence is not a reliable indicator of intracellular superoxide formation: another inconvenient truth
.
Free Radic. Biol. Med.
48
,
983
-
1001
.
Zielonka
,
J.
,
Sikora
,
A.
,
Hardy
,
M.
,
Joseph
,
J.
,
Dranka
,
B. P.
and
Kalyanaraman
,
B.
(
2012a
).
Boronate probes as diagnostic tools for real time monitoring of peroxynitrite and hydroperoxides
.
Chem. Res. Toxicol.
25
,
1793
-
1799
.
Zielonka
,
J.
,
Zielonka
,
M.
,
Sikora
,
A.
,
Adamus
,
J.
,
Joseph
,
J.
,
Hardy
,
M.
,
Ouari
,
O.
,
Dranka
,
B. P.
and
Kalyanaraman
,
B.
(
2012b
).
Global profiling of reactive oxygen and nitrogen species in biological systems: high-throughput real-time analyses
.
J. Biol. Chem.
287
,
2984
-
2995
.
Zielonka
,
J.
,
Lambeth
,
J. D.
and
Kalyanaraman
,
B.
(
2013
).
On the use of L-012, a luminol-based chemiluminescent probe, for detecting superoxide and identifying inhibitors of NADPH oxidase: a reevaluation
.
Free Radic. Biol. Med.
65
,
1310
-
1314
.

Competing interests

The authors declare no competing or financial interests.