Pediatr Blood Cancer

REVIEW Fanconi Anemia (FA) and Crosslinker Sensitivity: Re-Appraising the Origins of FA Definition Giovanni Pagano,

MSc,

1

* Marco d’Ischia,

The commonly accepted definition of Fanconi anemia (FA) relying on DNA repair deficiency is submitted to a critical review starting from the early reports pointing to mitomycin C bioactivation and to the toxicity mechanisms of diepoxybutane and a group of nitrogen mustards causing DNA crosslinks in FA cells. A critical analysis of the literature prompts revisiting the FA phenotype and

Key words:

PhD,

2

and Federico V. Pallardo ,

MD, PhD

3

crosslinker sensitivity in terms of an oxidative stress (OS) background, redox-related anomalies of FA (FANC) proteins, and mitochondrial dysfunction. This re-appraisal of FA basic defect might lead to innovative approaches both in elucidating FA phenotypes and in clinical management. Pediatr Blood Cancer # 2015 Wiley Periodicals, Inc.

bioactivation; crosslinkers; diepoxybutane; FANC proteins; Fanconi anemia; glutathione; melphalan; mitomycin C; oxidative stress

INTRODUCTION Fanconi anemia (FA) is generally defined as a genetic disease characterized by bone marrow failure, excess risk of malignancies, and by excess sensitivity to crosslinkers, that is, agents causing DNA interstrand crosslinks (ICL). FA’s crosslinker sensitivity is commonly attributed to deficiency in DNA repair pathways since early reports dating back to mid-1970s [1–8], in analogy with other genetic diseases categorized for DNA repair defects [9,10]. A mechanistic alternative to this theory is provided by the body of literature pointing to toxicity mechanisms of crosslinkers, such as mitomycin C (MMC), diepoxybutane (DEB) used in FA diagnosis, and of a nitrogen mustard, cyclophosphamide (CP) used in pretransplant conditioning. Some of these xenobiotics either require biotransformation leading to formation of alkylating and toxic derivatives (MMC, CP), or their toxicities involve redox-related effectors such as glutathione (GSH) for DEB. The present review critically discusses the work that found metabolic errors in FA phenotype and the redox functions of FA (FANC) proteins. This re-appraisal may lead to an unprecedented impetus in elucidating FA’s cellular and clinical phenotype, along with long-awaited efforts in improving FA’s clinical management.

CROSSLINKER-ASSOCIATED TOXICITY MECHANISMS Early research on new drugs for antineoplastic chemotherapy prompted investigations on toxicity mechanisms of candidate antineoplastic drugs, including biotransformation leading to directly acting derivatives/metabolites [11]. This was especially the case for various xenobiotics resulting in DNA alkylation and in particular, some agents causing ICL, which were termed crosslinkers because they produce disarrangement of the DNA chains. A different focus originally arose for the other major xenobiotic used for FA diagnostic purposes, DEB, which was discovered as a directly acting carcinogenic and mutagenic metabolite of butadiene, utilized in the production of synthetic rubber [12].

of cell extracts activating MMC to its directly acting derivatives. Subsequent studies found that MMC bioactivation is associated with redox mechanisms involving NADPH cytochrome P-450 reductase producing reactive oxygen species (ROS) with formation of semiquinone radicals [15]; moreover, MMC toxicity was found to strictly depend on hypoxia [16]. MMC bioactivation was found to result in lipid peroxidation that was inhibited by antioxidant activities (superoxide dismutase [SOD] and catalase [CAT]), by glutathione (GSH), and ascorbic acid (AA) [17–22].

Diepoxybutane Diepoxybutane has long been known as a directly acting mutagen following 1,3-butadiene biotransformation [23]. Subsequent investigations elucidated the role of GSH and GSH-related activities (GSH S-transferase, GST) in coping with DEB-induced toxicity. As shown in Table II, Pelin et al. [24] reported that GST activity was involved in a major detoxification pathway for DEB, and Landi et al. [25] found that DEB-induced sister chromatid exchange (SCE) was dependent on the expression of GST, while GSH depletion was found to enhance DEB toxicity [26,27]. Other studies reported on redox modulation of DEB toxicity, such as expression of two important antioxidant enzymes, thioredoxin (Trx), and GSH peroxidase (GSH-Px) in FA cells, along with DEBinduced ROS formation, and inhibition of CYP2E1 activity [28–32]. Altogether, the available database on DEB-induced toxicity points to combined redox-dependent mechanisms, including ROS formation and dependence on GSH-related detoxification, 1

Istituto Nazionale Tumori Fondazione G. Pascale—Cancer Research Center at Mercogliano (CROM), Mercogliano (AV), Italy; 2Department of Chemical Sciences, University of Naples “Federico II,” Naples, Italy; 3University of Valencia—INCLIVA, CIBERER (Centro de Investigaci on Biome´dica en Red de Enfermedades Raras), Valencia, Spain Conflict of interest: Nothing to declare.



Mitomycin C As shown in Table I, early reports by Iyer and Szybalski since 1963 [13,14] demonstrated that MMC toxicity requires the addition

2015 Wiley Periodicals, Inc. DOI 10.1002/pbc.25452 Published online in Wiley Online Library (wileyonlinelibrary.com).

C

Correspondence to: Giovanni Pagano, Istituto Nazionale Tumori Fondazione G. Pascale—Cancer Research Center at Mercogliano (CROM)—IRCCS, Mercogliano (AV), 83013, Italy. E-mail: [email protected] Received 3 November 2014; Accepted 12 January 2015

2

Pagano et al.

TABLE I. Historical Summary of Early Reports of MMC Bio-Reductive Activation (1960’s–1980’s) Authors (years) Iyer & Szybalski (1963–1964) Handa & Sato (1975) Bachur et al. (1979)

Kennedy et al. (1980) Teicher et al. (1981) Tomasz & Lipman (1981) Rockwell et al. (1982) Trush et al. (1982) Gutteridge et al. (1984)

Main findings

References

MMC in vitro effect on DNA requires a cell extract addition; formation of links between complementary DNA strands by metabolically activated MMC Quinone-containing anticancer chemicals form semiquinone radicals NADPH cytochrome P-450 reductase catalyzes the single-electron reduction of quinone antibiotics (including MMC); MMC is activated to free radical intermediates which transfer their single electron to O2 to form O2 MMC toxicity is enhanced in hypoxic cells and requires NADPH-generating system ROS formation during MMC bioactivation

[13,14] [15] [16]

[17] [18–20]

MMC biotransformation generates lipid peroxidation-associated chemiluminescence; lipid peroxidation was inhibited by SOD, GSH, ascorbic acid, and CAT MMC stimulates deoxyribose degradation with peroxidation products under hypoxia. This damage is inhibited by scavengers of the OH radical, Fe chelators, CAT and SOD.

[21] [22]

MMC, mitomycin C; ROS, reactive oxygen species; SOD, superoxide dismutase; CAT, catalase.

by testifying a role for cell redox systems in modulating DEB toxicity. Thus, one may consider the excess DEB sensitivity of FA cells as related to abnormal redox status, and specifically, to anomalies in GST expression.

Melphalan and Cyclophosphamide The alkylating action of several nitrogen mustards has long been related to GSH depletion that is involved in detoxification and repair of cellular injury, along with induction of oxidative damage and with increased hypoxia sensitivity [33–37]. Melphalan (MEL) toxicity was tested in early reports as a crosslinker in FA cells [8] (see below). As shown in Table III, subsequent studies found that MEL toxicity is enhanced by GSH depletion and hypoxia [38–39] by increasing DNA fragmentation, peroxide accumulation, and endogenous formation of H2O2 [40–43]. Altogether, the direct MEL-associated crosslinking activity can be related to the multifold relationships of MEL toxicity with GSH levels, GSH-related activities, and with MEL-induced prooxidant state. Analogous to MEL, as shown in Table III, CP toxicity was found to relate with GSH depletion, and CP biotransformation resulted in ROS-forming active metabolites, along with oxidative DNA

damage and showing protection by antioxidants [44–51]. A specific relationship of CP toxicity with FA phenotype was reported by Joenje and Oostra [52], who found a clastogenic effect of nonactivated CP in FA lymphocyte cultures, suggesting a spontaneous—and excess—propensity of FA cells in CP bioactivation. This finding provided direct evidence for a metabolic anomaly of FA cells in CP bioactivation, while this subject was not further investigated and still deserves ad hoc studies.

OXIDATIVE STRESS AND MITOCHONDRIAL DYSFUNCTION IN FA PHENOTYPE As shown in Table IV, a series of independent investigations related FA phenotype with OS since the pioneering report by Ingrid Emerit in 1977 [53], who found SOD-induced decrease of FA cell chromosomal instability, which was confirmed in MMC-treated FA cells. Excess oxygen and iron sensitivity was reported in FA cells, both involving cell viability and G2 cell growth phase arrest [54–58]. Excess oxidative DNA damage was observed in FA lymphoblasts, which was enhanced in MMC-exposed cells, and was partly attributed by Takeuchi and Morimoto to decreased CAT

TABLE II. Historical Summary of Published Evidence for DEB Toxicity Mechanisms Authors (years) Pelin et al. (1996) Landi et al. (1996) Vlachodimitropoulos et al. (1997) Spano` et al. (1998) Ruppitsch et al. (1998) Erexson & Tindall (2000) Davies et al. (2005) Yadavilli et al. (2007) Hartman et al. (2014)

Main findings

References

GSTT1 activity as a major detoxification pathway for DEB SCE induction by DEB is enhanced in GSTT1-null homozygotes Increased DEB sensitivity in lymphocytes from GSTT-deficient donors Glutathione depletion enhances DEB toxicity Overexpression of thioredoxin in FA cells abolished MMC- and DEB-induced DNA damage GSH-Px is involved in the detoxification of DEB-induced DNA damage; DEB causes ROS formation DEB sensitivity was significantly increased in GSTT1-null FA patients; GSTM1 influenced time to BMF in FA-C patients DEB induced apoptosis and ROS formation; trans-membrane potential in DEBinduced apoptosis DEB inhibits CYP2E1 activity

[24] [25] [26] [27] [28]

DEB, diepoxybutane; GST, GSH S-transferase; SCE, sister chromatid exchange; CYP, cytochrome P 450. Pediatr Blood Cancer DOI 10.1002/pbc

[29] [30] [31] [32]

Crosslinking Agents and Fanconi Anemia

3

TABLE III. Historical Summary of Published Evidence for Toxicity Mechanisms of Melphalan and Cyclophosphamide Agents Melphalan (MEL)

Authors (years) Roizin-Towle (1985) Lunel-Orsini et al. (1995) Vahrmeijer et al. (1999)

de Wilt et al. (1999) Troyano et al. (2001) Matsura et al. (2004) Cyclophosphamide (CP)

Crook et al. (1986) LeBlanc & Waxman (1990) Stankiewicz & Skrzydlewska (2003)

Murata et al. (2004) Selvakumar et al. (2006) Sadir et al. (2007) Kasapovic´ et al. (2010) Liu et al. (2012)

Main findings

References

Increased toxicity by GSH depletion and hypoxia GSH depletion increased MEL toxicity GSH depletion caused a fourfold increase in DNA fragmentation and a sevenfold increase in the fraction of apoptotic cells Hypoxia enhanced anti-tumor activity of MEL GSH depletion enhanced peroxide accumulation and apoptosis H2O2 formation, phosphatidylserine oxidation, cyt C release in cytosol CP metabolite acrolein was effective in GSH depletion CP and acrolein modulate hepatic P-450 expression CP is biotransformed into ROS-forming active metabolites; decreased SOD, GPx, GR, and CAT; decreased GSH, Vit C and E 4-hydroperoxyCP caused oxidative DNA damage (8-oxodG) Protection by a-lipoic acid of CYP-induced DNA damage Protection by antioxidants (melatonin) of CYP-induced OS CP causes oxidative DNA damage and decreases antioxidant activities (Cu,ZnSOD, GPx, GR) and GSH levels CP metabolite acrolein impairs the cytoskeleton and induces OS

[38] [39] [40]

[41] [42] [43] [44] [45] [46]

[47] [48] [49] [50] [51]

GPx, glutathione peroxidase; GR, glutathione reductase; 8-oxodG, 8-Oxo-20 -deoxyguanosine.

activity [59], while Trx overexpression counteracted MMC- and DEB-induced toxicity in FA fibroblasts [28]. Unconfined to in vitro testing, a series of independent studies conducted on blood cells and biological fluids from patients with FA found an in vivo prooxidant state assessed on a set of redox endpoints, including oxidative DNA damage, GSH imbalance (expressed as GSH:GSSG ratio), WBC chemiluminescence, methylglyoxal levels, and SOD expression [60–63]. The formation and toxicity of aldehydes have been focused in recent years as a hallmark in FA phenotype. Aldehydes are established biochemical correlates of OS in a broad variety of physiological and pathological conditions (reviewed in [64,65]). Polyunsaturated lipids and sugars are typical sources of OS-related aldehydes, such as 4-hydroxynonenal, acrolein, MDA, and glyoxal/ methylglyoxal, whose formation is an index of, and responsible for,

extensive cell damage. Besides being a footprint of ROS generation, aldehydes can mediate a number of toxicity pathways which may ultimately lead to DNA cross-linking [65]. Several aldehydes have been implicated in formation of covalent adducts and crosslinks targeted to dG [66]. A series of reports have focused on a deficiency of aldehyde dehydrogenase 2 (ALDH2) in FANCD2 cells and fancd2 ( / ) mice [67–69], relating ALDH2 deficiency to inability to counteract acetaldehyde toxicity. The relevance of these data has found support by the report of Hira et al. [70], who found that a ALDH2 variant is associated with accelerated progression of bone marrow failure in Japanese patients with FA. However, whether an ALDH2 defect does actually play a significant and specific role in acetaldehyde detoxification awaits verification, in view of the mentioned excess in vivo levels of other aldehydes in patients with FA [62,63]. Moreover, ALDH2 displays low specificity in aldehyde

TABLE IV. Early Studies Assessing OS-Related Mechanisms in FA Cells Authors (years) Nordenson (1977) Joenje et al. (1981) Joenje & Oostra (1983) Nagasawa & Little (1983)

Saito et al. (1993) Takeuchi & Morimoto (1993) Poot et al. (1996) Ruppitsch et al. (1998)

Main findings

References

Chromosomal instability of FA cells is decreased by SOD The rate of chromosomal breaks in FA cells is decreased by low-level oxygen The influence of low-level oxygen in enhanced in MMC-treated FA cells MMC cytotoxicity is suppressed by SOD in FA cells; “Hypersensitivity to MMC cytoxicity in FA cells may involve among other factors a deficiency in the inactivation of certain free radical intermediates” “The sensitivity to MMC in different FA lines varied from normal level to extreme hypersensitivity, whereas all the FA lines showed similar hypersensitivity to oxygen” FA-A cells have increased susceptibility to oxidative DNA damage; this increased susceptibility is possibly due to decreased CAT activity A G2 phase defect is dependent upon the oxygen concentration; exposure to excess iron was very toxic to FA cells at 20%, but less toxic at 5% oxygen Overexpression of thioredoxin cDNA in FA fibroblasts abolished the DNA damaging effects of MMC and DEB

[53] [54] [55] [56]

Pediatr Blood Cancer DOI 10.1002/pbc

[57] [58] [59] [28]

4

Pagano et al.

clearance by counteracting 4-hydroxynonenal toxicity [71]. The relative impact of aldehyde mediated toxicity versus other OSrelated pathways of cell damage remains little understood whereby excess aldehyde formation (or a detoxification defect) stands at present as only one of the many possible contributory factors produced in a pro-oxidant state. This is the case, for example, for oxidative DNA damage (8-OHdG) that occurs in FA phenotype [58,61–63], and displays a cascade of adverse effects including mutagenesis [72,73]. Thus, a possible attempt to improve FA’s clinical course by means of aldehyde detoxifying agents might not compensate the basic—and multi-faceted—pro-oxidant state featured in FA phenotype. A few studies tested some different antioxidants in FA knock-out mice reporting protective effects of some antioxidants (tempol and resveratrol, but not N-acetylcysteine) [74–76]. These findings might suggest an appropriate design of further animal studies, and possibly, of clinical trials. Closely related to redox impairment, mitochondrial abnormalities have been reported in FA cells of four genetic subtypes (A, C, D2, and G), pointing to multi-faceted links between mitochondrial dysfunction in FA cellular phenotype and the available database on the roles for OS in FA cells [77–79]. Alterations in mitochondrial morphology were reported by Kumari et al. [78], including damaged mitochondrial membranes, abnormal shapes, and possible events of mitophagy consistent with decreased mitochondrial transmembrane potential (DCm) and overproduction of ROS in FA cells. We have recently evaluated transcriptomal analyses on RNA obtained from bone marrow cells from patients with FA versus healthy volunteers, and identified significant changes in the expression of genes involved in bioenergetic pathways and in antioxidant activities [80]. Altogether, an in vivo, as yet unexplored, involvement of MDF in patients with FA may be viewed as a realistic working hypothesis based on the current database and prompting further investigations in this subject.

REDOX-RELATED ROLES OF FANC PROTEINS Some FANC proteins have been associated with several redoxrelated activities and/or interactors, as shown in Table V. Independent studies by Kruyt et al. and by Futaki et al. found that FANCC and FANCG proteins bind to and regulate the activity of cytochrome-P450 [81,82], microsomal membrane proteins involved in electron transfer, and long recognized as effectors in MMC bioactivation [83]. A direct involvement of BRCA1 and BRCA2 (FANCD1) proteins was reported in the repair of 8oxoguanine damage, thus, implying their direct role in redox pathways [84], as oxidation of guanine is a well-known parameter of OS and key factor in cancer proneness. Other investigations found functional interactions of FANCC and FANCG proteins with NADPH cytochrome P450 reductase and by redox regulation of GSTP1 [85]. Park et al. found that FANCA and FANCG proteins were sensitive to redox conditions since their monomers became multimers following H2O2 treatment [86]. The links of the FANCD2 protein with OS were reported in independent studies that found interactions with ATM (Ataxia–Telangiectasia mutated protein) and FOXO3a (forkhead box O3) exerting coordinated actions with FANCD2 consistent with the established functions of both ATM and FOXO3a in response to oxidative DNA damage [87– 89]. An extensive database points to the multiple links of the FANCJ (FANCJ/BACH1/BRIP1) protein with OS. This protein displays functional interaction with BRCA1 in breast and ovary cancer suppression [90,91] and in turn, BRCA1 is known to be involved in OS- related mechanisms [92]. Moreover, FANCJ/BACH1/BRIP1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants [93]. FANCJ/BACH1/BRIP1 features a helicase activity and an ATPase activity, a twofold activity that is recognized for other RECQ helicases that are associated with ATPase activity [94]. FANCJ/BACH1/BRIP1 regulates heme

TABLE V. Redox-Related Functions of FANC Proteins Consistent With a Prooxidant State in FA Cells Authors (years) Kruyt et al. (1998) Futaki et al. (2002) Le Page et al. (2000) Cumming et al. (2001) Park et al. (2004) Li et al. (2010) Castillo et al. (2011) Du et al. (2012) Cantor and Andreassen (2006) Levran et al. (2005) Dhakshinamoorthy et al. (2005) Mukhopadhyay et al. (2006) Kitamuro et al. (2003) Okada et al. (2010) Suhasini et al. (2009)

Main findings

References

FANCC protein binds to NADPH CYP-P450 reductase, involved in electron transfer FANCG interacts with cytochrome P450 2E1 (CYP2E1), which is associated with the production of ROS and the bioactivation of carcinogens BRCA1 and BRCA2 proteins are involved in the repair of 8-oxoguanine damage FANCC protein interacts glutathione S-transferase P1-1, which catalyzes the detoxification of xenobiotics and by-products of oxidative stress FANCA and FANCG respond to oxidative damage by forming complexes Functional interaction of the FANCD2 protein and the forkhead box O 3a (FOXO3a) FANCD2 colocalized with FOXO3a in response to oxidative stress; FANCD2 as interactor with Ataxia telangiectasia mutated protein (ATM) OS-induced FANCD2 ubiquitination interacts with FA-BRG1–promoter complex RECQ helicase associated with ATPase activity FANCJ/BACH1/BRIP1 interacts with heme oxygenase-1; BRCA 1 ARE-NAD(P)H:quinone oxidoreductase-1; sensing thymin glycol damage FANCG protein is found in mitochondria; wild-type but not mutant FANCG physically interacts with the mitochondrial peroxidase PRDX3 Bach1 as hypoxia-inducible repressor for the heme oxygenase-1 gene

[81] [82]

FANCJ/BACH1/BRIP1 senses oxidative DNA damage

[84] [85] [86] [87] [88] [89] [90] [91] [93] [77] [95] [96] [97]

FANC, proteins encoded by FA defective genes; BRCA, breast cancer-related genes and proteins; RECQ helicase, family of helicase enzymes; ARE, antioxidant response element; PRDX3, peroxiredoxin-3. Pediatr Blood Cancer DOI 10.1002/pbc

Crosslinking Agents and Fanconi Anemia oxygenase-1 [95,96], senses oxidative DNA damage, and is stimulated by replication protein A to unwind the damaged DNA substrate in a strand-specific manner [97]. Altogether, the database on interactions of several FANC proteins points to multiple molecular interactions of at least five FANC (A, C, D2, G, and J) proteins with redox-related mechanisms.

DEFECTIVE DNA REPAIR: TOWARD THE ROOTS OF A SHARED AXIOM By far, the most shared opinion, defective DNA repair with excess sensitivity to crosslinkers is associated with FA definition and provides the basis for most of the FA literature up to recent reviews [9,10]. Albeit currently assumed as an undisputed axiom, this theory dates back to a few early studies that may be analyzed under a historical viewpoint. The pioneering studies by Sasaki [1,2] tested the susceptibility to chromosome breakage of peripheral blood lymphocytes from patients with FA exposed to a number of clastogens (including MMC). The increased susceptibility to compounds resulting in DNA ICL was interpreted as an indication that the FA cells are defective in DNA repair mechanisms. No mention was provided to the toxicity mechanisms of the tested clastogens, namely related to MMC requirement of bioactivation, as available in the previous and contemporary literature [15–19]. Excision repair ability was investigated in FA cells in terms of MMC cell killing and by DNA sucrose sedimentation compared to HeLa cells and Xeroderma pigmentosum (XP) cells [3,4]. The results showed that FA DNA sedimented much faster after MMC treatment suggesting a possible impairment in FA cell’s ability to unhook crosslinks [4]. However, no reference was provided to the available literature, whether the excess MMC sensitivity of FA cells might be due to an abnormal propensity to activate MMC to its directly acting metabolites [3,4]. Latt et al. [5] found a decreased SCE induction in FA cells versus normal cells, while MMC treatment resulted in a partial suppression of mitosis and a marked increase in chromatid breaks and rearrangements. The authors suggested that chromosomal breaks and rearrangements in FA cells might result from a defect in repair of DNA damage. Unlike the above studies, Auerbach and Wolman [6] in 1976 investigated the effects of DEB and methyl methane sulfonate (MMS) as directly acting mutagens by prolonged exposure of FA fibroblasts versus XP and versus control cells, and by evaluating the frequency of chromosomal aberrations after DEB or MMS removal. This study was designed by assuming the direct mutagenic action for DEB, as still currently recognized, while the GSH-related modulation of DEB toxicity (Table II) would be reported some 20 years later [24–26]. A quite singular conclusion was offered in the report by Raj and Heddle [7], who tested the induction of chromosomal breakage in FA and normal cells with or without MMC treatment and cultured with SOD, or CAT, or l-cysteine. These authors found that antioxidant treatments resulted in decreased chromosomal breaks both in FA and in normal cells, thus, they stated that the effect was not specific, by hence concluding for the occurrence of DNA repair deficiency. Opposite conclusions were provided by Nagasawa and Little [56], who found that survival of MMC-treated FA cells was increased by adding SOD, unlike MMC-treated normal cells. These authors suggested that the hypersensitivity to “MMC cytoxicity in Pediatr Blood Cancer DOI 10.1002/pbc

5

FA cells may involve among other factors a deficiency in the inactivation of certain free radical intermediates [56].” A major support to the theory of DNA repair deficiency was provided by Ishida and Buchwald [8]. This report evaluated growth inhibition of FA lymphoblasts exposed to a number of mono- and bifunctional crosslinking agents including, among others, MMC and MEL, and found that FA cells are more sensitive than normal cell lines to all DNA crosslinking agents examined. Interestingly, these authors stated that “MMC requires activation by a NADPHgenerating system [14] before it will cross-link DNA while melphalan does not [98], yet FA cells have similar sensitivities to the two drugs. [...] It is probable, therefore, that the defect(s) in FA is at the level of repair of the damage in DNA [8].” About MMC biotransformation, one may comment on the quite outdated citation from Iyer and Szybalski [14], published almost 20 years before this report by disregarding the recent and contemporary literature providing evidence for redox-related MMC biotransformation [15– 19]. On the other hand, one should recognize that studies of redoxrelated toxicity mechanisms of MEL were not yet available. Thus, this basic report on FA-associated DNA repair deficiency reached a conclusion partly unaware of the up-to-date literature (MMC bioactivation), and partly surpassed by following investigations (MEL-associated redox toxicity mechanisms). Altogether, the founding publications supporting the theory of DNA repair deficiency in FA either failed to consider the available literature pointing to MMC bioactivation requirements or could not foresee the subsequent evidence for redox toxicity (GSH-related) mechanisms of DEB and MEL. That theory, though presently shared almost universally in the scientific community, cannot be regarded as an undisputable truth, as far as its origins can be critically reconstructed in a historical framework, displaying some inborn pitfalls as discussed here.

RESEARCH PROSPECTS The prevailing opinion attributing FA’s basic defect to impaired DNA repair has led to the current state of art in clinical practice, as yet confined to androgen therapy and/or to optimized transplantation protocols. A long-lasting, so far unsuccessful prospect treatment has been targeted to gene therapy. We can hope that new developments make feasible this therapy. On the other hand, the hitherto overlooked evidence for OS and MDF in FA relies on a body of independent reports encompassing redox toxicity mechanisms of FA-related crosslinkers, multifaceted evidence for OS in FA cells and patients, redox-related functions of several FANC proteins, and impaired mitochondrial function in FA cells from four genetic subtypes. Further studies designed to test this theory might contribute to basic elucidation of FA phenotype and might provide prospects for innovative clinical interventions in patients with FA.

REFERENCES 1. Sasaki MS, Tonomura A. A high susceptibility of Fanconi’s anemia to chromosome breakage by DNA cross-linking agents. Cancer Res 1973;33:1829–1836. 2. Sasaki MS. Is Fanconi’s anaemia defective in a process essential to the repair of DNA cross links. Nature 1975;257:501–503. 3. Fujiwara Y, Tatzumi M. Repair of mitomycin C damage to DNA in mammalian cells and its impairment in Fanconi’s anemia cells. Biochem Biophys Res Commun 1975;66:592–598. 4. Fujiwara Y. Defective repair of mitomycin C crosslinks in Fanconi’s anemia and loss in confluent normal human and xeroderma pigmentosum cells. Biochim Biophys Acta 1982;699:217–225. 5. Latt SA, Stetten G, Juergens LA, Buchanan GR, Gerald PS. Induction by alkylating agents of sister chromatid exchanges and chromatid breaks in Fanconi’s anemia. Proc Natl Acad Sci USA 1975;72: 4066–4070.

6

Pagano et al.

6. Auerbach AD, Wolman SR. Susceptibility of Fanconi’s anaemia fibroblasts to chromosome damage by carcinogens. Nature 1976;261:494–496. 7. Raj AS, Heddle JA. The effect of superoxide dismutase, catalase and L-cysteine on spontaneous and on mitomycin C induced chromosomal breakage in Fanconi’s anemia and normal fibroblasts as measured by the micronucleus method. Mutat Res 1980;78:59–66. 8. Ishida R, Buchwald M. Susceptibility of Fanconi’s anemia lymphoblasts to DNA-cross-linking and alkylating agents. Cancer Res 1982;42:4000–4006. 9. Kupfer GM. Fanconi anemia: A signal transduction and DNA repair pathway. Yale J Biol Med 2013;86:491–497. 10. Kim H, D’Andrea AD. Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway. Genes Dev 2012;26:1393–1408. 11. Fell HB, Allsopp CB. The action of mustard gas on living cells in vitro. Cancer Res 1948;8:145–161. 12. Norppa H, Sorsa M. Genetic toxicity of 1,3-butadiene and styrene. IARC Sci Publ 1993;127:185–193. 13. Iyer VN, Szybalski W. A molecular mechanism of mitomycin action: Linking of complementary DNA strands. Proc Natl Acad Sci USA 1963;50:355–362. 14. Iyer VN, Szybalski W. Mitomycins and porfiromycin: Chemical mechanism of activation and crosslinking of DNA. Science 1964;145:55–58. 15. Handa K, Sato S. Generation of free radicals of quinone group-containing anti-cancer chemicals in NADPH-microsome system as evidenced by initiation of sulfite oxidation. Gann 1975;66:43–47. 16. Bachur NR, Gordon SL, Gee MV, Kon H. NADPH cytochrome P-450 reductase activation of quinone anticancer agents to free radicals. Proc Natl Acad Sci USA 1979;76:954–957. 17. Kennedy KA, Rockwell S, Sartorelli AC. Preferential activation of mitomycin C to cytotoxic metabolites by hypoxic tumor cells. Cancer Res 1980;40:2356–2360. 18. Teicher BA, Lazo JS, Sartorelli AC. Classification of antineoplastic agents by their selective toxicities toward oxygenated and hypoxic tumor cells. Cancer Res 1981;41:73–81. 19. Tomasz M, Lipman R. Reductive metabolism and alkylating activity of mitomycin C induced by rat liver microsomes. Biochemistry 1981;20:5056–5061. 20. Rockwell S, Kennedy KA, Sartorelli AC. Mitomycin-C as a prototype bioreductive alkylating agent: In vitro studies of metabolism and cytotoxicity. Int J Radiat Oncol Biol Phys 1982;8:753–755. 21. Trush MA, Mimnaugh EG, Ginsburg E, Gram TE. Studies on the in vitro interaction of mitomycin C, nitrofurantoin and paraquat with pulmonary microsomes. Stimulation of reactive oxygen-dependent lipid peroxidation. Biochem Pharmacol 1982;31:805–814. 22. Gutteridge JM, Quinlan GJ, Wilkins S. Mitomycin C-induced deoxyribose degradation inhibited by superoxide dismutase. A reaction involving iron, hydroxyl and semiquinone radicals. FEBS Lett 1984;167:37–41. 23. Olszewska E, Kilbe BJ. The mutagenic activity of diepoxybutane in yeast. Mutat Res 1975;33:383–390. 24. Pelin K, Hirvonen A, Norppa H. Influence of erythrocyte glutathione S-transferase T1 on sister chromatid exchanges induced by diepoxybutane in cultured human lymphocytes. Mutagenesis 1996;11:213–215. 25. Landi S, Ponzanelli I, Hirvonen A, Norppa H, Barale R. Repeated analysis of sister chromatid exchange induction by diepoxybutane in cultured human lymphocytes: Effect of glutathione S-transferase T1 and M1 genotype. Mutat Res 1996;351:79–85. 26. Vlachodimitropoulos D, Norppa H, Autio K, Catala´n J, Hirvonen A, Tasa G, Uusku¨la M, Demopoulos NA, Sorsa M. GSTT1-dependent induction of centromere-negative and -positive micronuclei by 1,2:3,4diepoxybutane in cultured human lymphocytes. Mutagenesis 1997;12:397–403. 27. Spano` M, Cordelli E, Leter G, Pacchierotti F. Diepoxybutane cytotoxicity on mouse germ cells is enhanced by in vivo glutathione depletion: A flow cytometric approach. Mutat Res 1998;397:37–43. 28. Ruppitsch W, Meisslitzer C, Hirsch-Kauffmann M, Schweiger M. Overexpression of thioredoxin in Fanconi anemia fibroblasts prevents the cytotoxic and DNA damaging effect of mitomycin C and diepoxybutane. FEBS Lett 1998;422:99–102. 29. Erexson GL, Tindall KR. Reduction of diepoxybutane-induced sister chromatid exchanges by glutathione peroxidase and erythrocytes in transgenic Big Blue mouse and rat fibroblasts. Mutat Res 2000;447:267– 274. 30. Davies SM, Radloff GA, DeFor TE, Levran O, Batish SD, Hanenberg H, Auerbach AD. GST genotype may modify clinical phenotype in patients with Fanconi anaemia. Br J Haematol 2005;131:118–122. 31. Yadavilli S, Martinez-Ceballos E, Snowden-Aikens J, Hurst A, Joseph T, Albrecht T, Muganda PM. Diepoxybutane activates the mitochondrial apoptotic pathway and mediates apoptosis in human lymphoblasts through oxidative stress. Toxicol In Vitro 2007;21:1429–1441. 32. Hartman JH, Miller GP, Boysen G. Inhibitory potency of 4-carbon alkanes and alkenes toward CYP2E1 activity. Toxicology 2014;318:51–58. 33. Robson CN, Lewis AD, Wolf CR, Hayes JD, Hall A, Proctor SJ, Harris AL, Hickson ID. Reduced levels of drug-induced DNA cross-linking in nitrogen mustard-resistant Chinese hamster ovary cells expressing elevated glutathione S-transferase activity. Cancer Res 1987;47:6022–6027. 34. Rappeneau S, Baeza-Squiban A, Braut-Boucher F, Aubery M, Gendron MC, Marano F. Use of fluorescent probes to assess the early sulfhydryl depletion and oxidative stress induced by mechlorethamine in human bronchial epithelial cells. Toxicol In Vitro 1999;13:765–771. 35. Ucar M, Korkmaz A, Reiter RJ, Yaren H, Oter S, Kurt B, Topal T. Melatonin alleviates lung damage induced by the chemical warfare agent nitrogen mustard. Toxicol Lett 2007;173:124–131. 36. Zheng R, Po I, Mishin V, Black AT, Heck DE, Laskin DL, Sinko PJ, Gerecke DR, Gordon MK, Laskin JD. The generation of 4-hydroxynonenal, an electrophilic lipid peroxidation end product, in rabbit cornea organ cultures treated with UVB light and nitrogen mustard. Toxicol Appl Pharmacol 2013;272: 345–355. 37. Arrick BA, Nathan CF. Glutathione metabolism as a determinant of therapeutic efficacy: A review. Cancer Res 1984;44:4224–4232. 38. Roizin-Towle L. Selective enhancement of hypoxic cell killing by melphalan via thiol depletion: In vitro studies with hypoxic cell sensitizers and buthionine sulfoximine. J Natl Cancer Inst 1985;74:151–157. 39. Lunel-Orsini C, Buttin G, De Saint Vincent. A glutathione depletion selectively imposed on mu glutathione S-transferase overproducing cells increases nitrogen mustard toxicity. Biochem Pharmacol 1995;49:329–338. 40. Vahrmeijer AL, van Dierendonck JH, Schutrups J, van de Velde CJ, Mulder GJ. Effect of glutathione depletion on inhibition of cell cycle progression and induction of apoptosis by melphalan (L-phenylalanine mustard) in human colorectal cancer cells. Biochem Pharmacol 1999;58:655–664. 41. de Wilt JHW, Manusama ER, van Tiel ST, van Ijken MG, ten Hagen, Eggermont TL. Prerequisites for effective isolated limb perfusion using tumour necrosis factor alpha and melphalan in rats. Brit J Cancer 1999;80:161–166. 42. Troyano A, Ferna´ndez C, Sancho P, de Blas E, Aller P. Effect of glutathione depletion on antitumor drug toxicity (apoptosis and necrosis) in U-937 human promonocytic cells. The role of intracellular oxidation. J Biol Chem 2001;276:47107–47115. 43. Matsura T, Kai M, Jiang J, Babu H, Kini V, Kusumoto C, Yamada K, Kagan VE. Endogenously generated hydrogen peroxide is required for execution of melphalan-induced apoptosis as well as oxidation and externalization of phosphatidylserine. Chem Res Toxicol 2004;17:685–696. 44. Crook TR, Souhami RL, Whyman GD, McLean AE. Glutathione depletion as a determinant of sensitivity of human leukemia cells to cyclophosphamide. Cancer Res 1986;46:5035–5038. 45. LeBlanc GA, Waxman DJ. Mechanisms of cyclophosphamide action on hepatic P-450 expression. Cancer Res 1990;50:5720–5726.

Pediatr Blood Cancer DOI 10.1002/pbc

46. Stankiewicz A, Skrzydlewska E. Protection against cyclophosphamide-induced renal oxidative stress by amifostine: The role of antioxidative mechanisms. Toxicol Mech Methods 2003;13:301–308. 47. Murata M, Suzuki T, Midorikawa K, Oikawa S, Kawanishi S. Oxidative DNA damage induced by a hydroperoxide derivative of cyclophosphamide. Free Radic Biol Med 2004;37:793–802. 48. Selvakumar E, Prahalathan C, Sudharsan PT, Varalakshmi P. Chemoprotective effect of lipoic acid against cyclophosphamide-induced changes in the rat sperm. Toxicology 2006;217:71–78. 49. Sadir S, Deveci S, Korkmaz A, Oter S. Alpha-tocopherol, beta-carotene and melatonin administration protects cyclophosphamide-induced oxidative damage to bladder tissue in rats. Cell Biochem Funct 2007;25:521–526. 50. Kasapovic´ J, Pejic´ S, Stojiljkovic´ V, Todorovic´ A, Radosˇevic´-Jelic´ L, Saicˇic´ ZS, Pajovic´ SB. Antioxidant status and lipid peroxidation in the blood of breast cancer patients of different ages after chemotherapy with 5-fluorouracil, doxorubicin and cyclophosphamide. Clin Biochem 2010;43:1287–1293. 51. Liu F, Li XL, Lin T, He DW, Wei GH, Liu JH, Li LS. The cyclophosphamide metabolite, acrolein, induces cytoskeletal changes and oxidative stress in Sertoli cells. Mol Biol Rep 2012;39:493–500. 52. Joenje H, Oostra AB. Clastogenicity of cyclophosphamide in Fanconi’s anemia lymphocytes without exogenous metabolic activation. Cancer Genet Cytogenet 1986;22:339–345. 53. Nordenson I. Effect of superoxide dismutase and catalase on spontaneously occurring chromosome breaks in patients with Fanconi’s anemia. Hereditas 1977;86:147–150. 54. Joenje H, Arwert F, Eriksson AW, de Koning H, Oostra AB. Oxygen-dependence of chromosomal aberrations in Fanconi’s anaemia. Nature 1981;290:142–143. 55. Joenje H, Oostra AB. Effect of oxygen tension on chromosomal aberrations in Fanconi anaemia. Hum Genet 1983;65:99–101. 56. Nagasawa H, Little JB. Suppression of cytotoxic effect of mitomycin-C by superoxide dismutase in Fanconi’s anemia and dyskeratosis congenita fibroblasts. Carcinogenesis 1983;4:795–799. 57. Saito H, Hammond AT, Moses RE. Hypersensitivity to oxygen is a uniform and secondary defect in Fanconi anemia cells. Mutat Res 1993;294:255–262. 58. Takeuchi T, Morimoto K. Increased formation of 8-hydroxydeoxyguanosine, an oxidative DNA damage, in lymphoblasts from Fanconi’s anemia patients due to possible catalase deficiency. Carcinogenesis 1993;14:1115–1120. 59. Poot M, Gross O, Epe B, Pflaum M, Hoehn H. Cell cycle defect in connection with oxygen and iron sensitivity in Fanconi anemia lymphoblastoid cells. Exp Cell Res 1996;222:262–268. 60. Rumiantsev AG, Samochatova EV, Afanas’ev IB, Korkina LG, Suslova TB. The role of free oxygen radicals in the pathogenesis of Fanconi’s anemia. Ter Arkh 1989;61:32–36. 61. Degan P, Bonassi S, De Caterina M, Korkina LG, Pinto L, Scopacasa F, Zatterale A, Calzone R, Pagano G. In vivo accumulation of 8-hydroxy-2’-deoxyguanosine in DNA correlates with release of reactive oxygen species in Fanconi’s anaemia families. Carcinogenesis 1995;16:735–742. 62. Pagano G, Degan P, d’Ischia M, Kelly FJ, Pallardo FV, Zatterale A, Anak SS, Akisik EE, Beneduce G, Calzone R, De Nicola E, Dunster C, Lloret A, Manini P, Nobili B, Saviano A, Vuttariello E, Warnau M. Gender- and age-related distinctions for the in vivo prooxidant state in Fanconi anaemia patients. Carcinogenesis 2004;25:1899–1909. 63. Petrovic´ S, Leskovac A, Kotur-Stevuljevic´ J, Joksic J, Guc-Scekic M, Vujic D, Joksic G. Gender-related differences in the oxidant state of cells in Fanconi anemia heterozygotes. Biol Chem 2011;392:625–632. 64. Pagano G, Talamanca AA, Castello G, Cordero MD, d’Ischia M, Gadaleta MN, Pallardo FV, Petrovic´ S, Tiano L, Zatterale A. Oxidative stress and mitochondrial dysfunction across broad-ranging pathologies: Toward a rational design of chemoprevention strategies by means of mitochondrial nutrients. Oxid Med Cell Longev 2014;541230. 65. Villamena FA. Molecular basis of oxidative stress: Chemistry, mechanisms, and disease pathogenesis. John Wiley & Sons, 2013. 66. Seitz HK, Stickel F. Molecular mechanisms of alcohol-mediated carcinogenesis. Nature Rev Cancer 2007;7:599–612. 67. Marietta C, Thompson LH, Lamerdin JE, Brooks PJ. Acetaldehyde stimulates FANCD2 monoubiquitination, H2AX phosphorylation, and BRCA1 phosphorylation in human cells in vitro: Implications for alcohol-related carcinogenesis. Mutat Res 2009;664:77–83. 68. Langevin F, Crossan GP, Rosado IV, Arends MJ, Patel KJ. Fancd2 counteracts the toxic effects of naturally produced aldehydes in mice. Nature 2011;475:53–58. 69. Garaycoechea JI, Crossan GP, Langevin F, Daly M, Arends MJ, Patel KJ. Genotoxic consequences of endogenous aldehydes on mouse haematopoietic stem cell function. Nature 2012;489:571–575. 70. Hira A, Yabe H, Yoshida K, Okuno Y, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Nakamura J, Kojima S, Ogawa S, Matsuo K, Takata M, Yabe M. Variant ALDH2 is associated with accelerated progression of bone marrow failure in Japanese Fanconi anemia patients. Blood 2013;122:3206–3209. 71. Guo JM, Liu AJ, Zang P, Dong WZ, Ying L, Wang W, Xu P, Song XR, Cai J, Zhang SQ, Duan JL, Mehta JL, Su DF. ALDH2 protects against stroke by clearing 4-HNE. Cell Res 2013;23:915–930. 72. Takeuchi T, Matsugo S, Morimoto K. Mutagenicity of oxidative DNA damage in Chinese hamster V79 cells. Carcinogenesis 1997;18:2051–2055. 73. Halliwell B. Oxygen nitrogen, pro-carcinogens. Damage to DNA by reactive oxygen, chlorine and nitrogen species: Measurement, mechanism and the effects of nutrition. Mutat Res 1999;443:37–52. 74. Zhang QS, Eaton L, Snyder ER, Houghtaling S, Mitchell JB, Finegold M, Van Waes C, Grompe M. Tempol protects against oxidative damage and delays epithelial tumor onset in Fanconi anemia mice. Cancer Res 2008;68:1601–1608. 75. Zhang QS, Marquez-Loza L, Eaton L, Duncan AW, Goldman DC, Anur P, Watanabe-Smith K, Rathbun RK, Fleming WH, Bagby GC, Grompe M. Fancd2-/- mice have hematopoietic defects that can be partially corrected by resveratrol. Blood 2010;116:5140–5148. 76. Zhang QS, Marquez-Loza L, Sheehan AM, Watanabe-Smith K, Eaton L, Benedetti E, Major A, Schubert K, Deater M, Joseph E, Grompe M. Evaluation of resveratrol and N-acetylcysteine for cancer chemoprevention in a Fanconi anemia murine model. Pediatr Blood Cancer 2014;61:740–742. 77. Mukhopadhyay SS, Leung KS, Hicks MJ, Hastings PJ, Youssoufian H, Plon SE. Defective mitochondrial peroxiredoxin-3 results in sensitivity to oxidative stress in Fanconi anemia. J Cell Biol 2006;175:225–235. 78. Kumari U, Jun WY, Bay BH, Lyakhovich A. Evidence of mitochondrial dysfunction and impaired ROS detoxifying machinery in Fanconi anemia cells. Oncogene 2014;33:165–172. 79. Ravera S, Vaccaro D, Cuccarolo P, Columbaro M, Capanni C, Bartolucci M, Panfoli I, Morelli A, Dufour C, Cappelli E, Degan P. Mitochondrial respiratory chain Complex I defects in Fanconi anemia complementation group A. Biochimie 2013;95:1828–1837. 80. Pagano G, Talamanca AA, Castello G, d’Ischia M, Pallardo FV, Petrovic´ S, Porto B, Tiano L, Zatterale A. Bone marrow cell transcripts from Fanconi anaemia patients reveal in vivo alterations in mitochondrial, redox and DNA repair pathways. Eur J Haematol 2013;91:141–151. 81. Kruyt FA, Hoshino T, Liu JM, Joseph P, Jaiswal AK, Youssoufian H. Abnormal microsomal detoxification implicated in Fanconi anemia group C by interaction of the FAC protein with NADPH cytochrome P450 reductase. Blood 1998;92:3050–3056. 82. Futaki M, Igarashi T, Watanabe S, Kajigaya S, Tatsuguchi A, Wang J, Liu JM. The FANCG Fanconi anemia protein interacts with CYP2E1: Possible role in protection against oxidative DNA damage. Carcinogenesis 2002;23:67–72. 83. Keyes SR, Fracasso PM, Heimbrook DC, Rockwell S, Sligar SG, Sartorelli AC. Role of NADPH: cytochrome c reductase and DT-diaphorase in the biotransformation of mitomycin C1. Cancer Res 1984;44:5638–5643.

Crosslinking Agents and Fanconi Anemia 84. Le Page F, Randrianarison V, Marot D, Cabannes J, Perricaudet M, Feunteun J, Sarasin A. BRCA1 and BRCA2 are necessary for the transcription-coupled repair of the oxidative 8-oxoguanine lesion in human cells. Cancer Res 2000;60:5548–5552. 85. Cumming RC, Lightfoot J, Beard K, Youssoufian H, O’Brien PJ, Buchwald M. Fanconi anemia group C protein prevents apoptosis in hematopoietic cells through redox regulation of GSTP1. Nature Med 2001;7:814–820. 86. Park SJ, Ciccone SL, Beck BD, Hwang B, Freie B, Clapp DW, Lee SH. Oxidative stress/damage induces multimerization and interaction of Fanconi anemia proteins. J Biol Chem 2004;279:30053–30059. 87. Li J, Du W, Maynard S, Andreassen PR, Pang Q. Oxidative stress-specific interaction between FANCD2 and FOXO3a. Blood 2010;115:1545–1548. 88. Castillo P, Bogliolo M, Surralles J. Coordinated action of the Fanconi anemia and ataxia telangiectasia pathways in response to oxidative damage. DNA Repair 2011;10:518–525. 89. Du W, Rani R, Sipple J, Schick J, Myers KC, Mehta P, Andreassen PR, Davies SM, Pang Q. The FA pathway counteracts oxidative stress through selective protection of antioxidant defense gene promoters. Blood 2012;119:4142–4151. 90. Cantor SB, Andreassen PR. Assessing the link between BACH1 and BRCA1 in the FA pathway. Cell Cycle 2006;5:164–167. 91. Levran O, Attwooll C, Henry RT, Milton KL, Neveling K, Rio P, Batish SD, Kalb R, Velleuer E, Barral S, Ott J, Petrini J, Schindler D, Hanenberg H, Auerbach AD. The BRCA1-interacting helicase BRIP1 is deficient in Fanconi anemia. Nat Genet 2005;37:931–933.

Pediatr Blood Cancer DOI 10.1002/pbc

7

92. Vurusaner B, Poli G, Basaga H. Tumor suppressor genes and ROS: Complex networks of interactions. Free Radic Biol Med 2012;52:7–18. 93. Dhakshinamoorthy S, Jain AK, Bloom DA, Jaiswal AK. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants. J Biol Chem 2005;280:16891–16900. 94. Monnat RJ Jr. Human RECQ helicases: Roles in DNA metabolism, mutagenesis and cancer biology. Semin Cancer Biol 2010;20:329–339. 95. Kitamuro T, Takahashi K, Ogawa K, Udono-Fujimori R, Takeda K, Furuyama K, Nakayama M, Sun J, Fujita H, Hida W, Hattori T, Shirato K, Igarashi K, Shibahara S. Bach1 functions as a hypoxiainducible repressor for the heme oxygenase-1 gene in human cells. J Biol Chem 2003;278: 9125–9133. 96. Okada S, Muto A, Ogawa E, Nakanome A, Katoh Y, Ikawa S, Aiba S, Igarashi K, Okuyama R. Bach1dependent and -independent regulation of heme oxygenase-1 in keratinocytes. J Biol Chem 2010;285:23581–23589. 97. Suhasini AN, Sommers JA, Mason AC, Voloshin ON, Camerini-Otero RD, Wold MS. Brosh RM Jr. FANCJ helicase uniquely senses oxidative base damage in either strand of duplex DNA and is stimulated by replication protein A to unwind the damaged DNA substrate in a strand-specific manner. J Biol Chem 2009;284:18458–18470. 98. Parsons PG, Carter FB, Morrison L, Regius MS. Mechanism of melphalan resistance developed in vitro in human melanoma cells. Cancer Res 1981;41:1525–1534.

Fanconi anemia (FA) and crosslinker sensitivity: Re-appraising the origins of FA definition.

The commonly accepted definition of Fanconi anemia (FA) relying on DNA repair deficiency is submitted to a critical review starting from the early rep...
352KB Sizes 0 Downloads 4 Views