throbber
Free Radical Biology & Medicine 42 (2007) 44 – 51
`
`www.elsevier.com/locate/freeradbiomed
`
`Original Contribution
`Oxalomalate regulates ionizing radiation-induced apoptosis in mice
`Jin Hyup Lee, Jeen-Woo Park ⁎
`
`School of Life Sciences and Biotechnology, College of Natural Sciences, Kyungpook National University, Taegu 702-701, Korea
`
`Received 12 June 2006; revised 25 August 2006; accepted 15 September 2006
`Available online 23 September 2006
`
`Abstract
`
`Ionizing radiation induces the production of reactive oxygen species, which play an important causative role in apoptotic cell death. Recently,
`we demonstrated that the control of mitochondrial redox balance and the cellular defense against oxidative damage are primary functions of
`mitochondrial NADP+-dependent isocitrate dehydrogenase (IDPm) by supplying NADPH for antioxidant systems. In this paper, we demonstrate
`that modulation of IDPm activity in the kidneys of mice regulates ionizing radiation-induced apoptosis. When oxalomalate, a competitive inhibitor
`of IDPm, was administered to mice, inhibition of IDPm and enhanced susceptibility of apoptosis reflected by DNA fragmentation, the changes in
`mitochondria function, and the modulation of apoptotic marker proteins were observed upon exposure to 2 Gy of γ-irradiation. We also observed a
`significant difference in the mitochondrial redox status between the kidneys of the control and the oxalomalate-administered mice. This study
`indicates that IDPm may play an important role in regulating the apoptosis induced by ionizing radiation, presumably, through acting as an
`antioxidant enzyme.
`© 2006 Elsevier Inc. All rights reserved.
`
`Keywords: Ionizing radiation; IDPm; Oxalomalate; Apoptosis; Redox status
`
`Introduction
`
`Radiation therapy has been commonly used for the treatment
`of tumors. Ionizing radiation has been shown to generate
`reactive oxygen species (ROS) in a variety of cells [1]. When
`water, the most abundant intracellular material, is exposed to
`ionizing radiation, decomposition reactions occur and a variety
`of ROS,
`including superoxide, hydroxyl
`radicals, singlet
`oxygen, and hydrogen peroxide, are generated [2]. The
`secondary radicals formed by the interaction of hydroxyl
`radicals with organic molecules may also be of importance
`[1,2]. These ROS have the potential to damage critical cellular
`
`Abbreviations: ROS, reactive oxygen species; SOD, superoxide dismutase;
`ICDHs, isocitrate dehydrogenases; IDPm, mitochondrial ICDH; DNPH, 2,4-
`dinitrophenylhydrazine; DTNB, 5,5′-dithiobis(2-nitrobenzoic acid); TRITC,
`tetramethylrhodamine isothiocyanate; DHR, dihydrorhodamine; PARP, poly
`(ADP-ribose) polymerase; JC-1, 5,5,6,6-tetrachloro-1,1,3,3-tetraethylbenzimi-
`dazolcarbocyanine iodide; 8-OH-dG, 8-hydroxy-2′-deoxyguanosine; MDA,
`malondialdehyde; SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel
`electrophoresis.
`⁎ Corresponding author. Fax: +82 53 943 2762.
`E-mail address: parkjw@knu.ac.kr (J.-W. Park).
`
`0891-5849/$ - see front matter © 2006 Elsevier Inc. All rights reserved.
`doi:10.1016/j.freeradbiomed.2006.09.016
`
`components such as DNA, proteins, and lipids and eventually
`result in physical and chemical damage to tissues that may lead
`to cell death or neoplastic transformation [3]. In many cases,
`ionizing radiation-induced cell death has been identified as
`apoptosis [4].
`Biological systems have evolved an effective and compli-
`cated network of defense mechanisms which enable cells to
`cope with lethal oxidative environments. These defense
`mechanisms involve antioxidant enzymes, such as superoxide
`− to
`dismutase (SOD), which catalyzes the dismutation of O2
`H2O2 and O2 [5], catalase, and peroxidases which remove
`hydrogen peroxide and hydroperoxides [6]. Since ROS appear
`to be mediators of the apoptosis by ionizing radiation [7],
`factors including antioxidant enzymes that regulate the fate of
`such species may be of great importance in the protection of
`cells against ionizing radiation-induced cell death.
`The isocitrate dehydrogenases (ICDHs, EC1.1.1.41 and
`EC1.1.1.42) catalyze oxidative decarboxylation of isocitrate to
`α-ketoglutarate and require either NAD+ or NADP+, producing
`NADH and NADPH, respectively [8]. NADPH is an essential
`reducing equivalent for the regeneration of reduced glutathione
`(GSH) by glutathione reductase and for the activity of NADPH-
`
`Rigel Exhibit 1028
`Page 1 of 8
`
`

`

`J.H. Lee, J.-W. Park / Free Radical Biology & Medicine 42 (2007) 44–51
`
`45
`
`dependent thioredoxin system [9,10], both are important in the
`protection of cells from oxidative damage. Therefore, ICDH
`may play an antioxidant role during oxidative stress. We
`recently reported that mitochondrial ICDH (IDPm) is involved
`in the supply of NADPH needed for GSH production against
`mitochondrial oxidative damage [11].
`In the present report, we demonstrate that the modulation of
`IDPm activity by oxalomalate in the kidneys of mice regulates
`apoptosis induced by γ-irradiation. Oxalomalate, a tricarboxylic
`acid (α-hydroxy-β-oxalosuccinic acid) formed in vitro and in
`vivo by condensation of oxaloacetate and glyoxylate, has been
`known to be a potent inhibitor of IDPm [12]. The results suggest
`that IDPm may play an important role in regulating the
`apoptosis induced by ionizing radiation, presumably, through
`acting as an antioxidant enzyme.
`
`Materials and methods
`
`Materials
`
`β-NADP+, isocitrate, 2,4-dinitrophenylhydrazine (DNPH),
`5,5-dithiobis(2-nitrobenzoic acid) (DTNB), xylenol orange,
`propidium iodide (PI), oxalomalate, avidin-conjugated tetra-
`methylrhodamine isothiocyanate (TRITC), and anti-rabbit IgG-
`conjugated TRITC secondary antibody were obtained from
`Sigma Chemical Co. (St. Louis, MO). 5,5,6,6-Tetrachloro-
`1,1,3,3-tetraethylbenzimidazolcarbocyanine iodide (JC-1), and
`dihydrorhodamine (DHR) 123 were purchased from Molecular
`Probes (Eugene, OR). Electrophoreses reagents and the Bio-
`Rad protein assay kit were purchased from Bio-Rad (Hercules,
`CA). Antibodies against Bcl-2, Bax, cleaved caspase-3, cleaved
`poly(ADP-ribose) polymerase (PARP), cytochrome c, Noxa,
`PUMA, p21, and p53 were purchased from Santa Cruz (Santa
`Cruz, CA) or Cell Signaling (Beverly, MA).
`
`Mice
`
`Six- to 8-week-old female C57BL/6 mice were purchased
`from the National Cancer Institute (Frederick, MD) and kept in
`the oncology animal facility of the Johns Hopkins Hospital
`(Baltimore, MD). All animal procedures were performed
`according to approved protocols and in accordance with
`recommendations for the proper use and care of laboratory
`animals.
`
`Animal treatment and whole-body irradiation
`
`Two groups of 15 C57BL/6 mice each received either
`oxalomalate or 0.9% NaCl. Solutions of oxalomalate were
`freshly prepared in 0.9% NaCl and administered before
`irradiation at a dose of 25 mg/kg in volumes equivalent to 1%
`of each animal's weight once daily for 10 days. Control mice
`were given 0.9% NaCl, and all injections were administered ip.
`After irradiation with a 137Cs source at a dose rate of 1 Gy/min,
`the mice were returned to climate-controlled cages for further
`experiment. Mice were sacrificed by a cervical dislocation at
`7 days postirradiation.
`
`Tissue dissociation
`
`Dissociation of kidney tissue was performed using a petri
`dish, tweezers, and a fresh scalpel blade. Dissociated tissue was
`prevented from drying out by covering it with a few drops of
`PBS. During dissociation, the kidney epithelial cell suspension
`was aspirated repeatedly through a 20-gauge needle into a
`syringe, and consequently injected through a cell strainer into a
`50-ml tube to retain clumps. Suspensions were centrifuged and
`resuspended with a defined amount of PBS, and final cell
`number was counted from aliquots.
`
`Measurement of IDPm activity
`
`For the preparation of mitochondrial fraction from kidney
`tissue, the tissue portions were homogenized with a Dounce
`homogenizer in sucrose buffer (0.32 M sucrose, 10 mM Tris-Cl,
`pH 7.4). Tissue homogenate was centrifuged at 1000 g for 5 min
`and the supernatants were further centrifuged at 15,000 g for
`30 min. The precipitates were washed twice with sucrose buffer
`to collect mitochondria pellet. The mitochondrial pellets were
`resuspended in 1× PBS containing 0.1% Triton X-100,
`disrupted by ultrasonication (4710 Series, Cole-Palmer, Chi-
`cago, IL) twice at 40% of maximum setting for 10 s, and
`centrifuged at 15,000 g for 30 min. The supernatants were used
`to measure the activity of IDPm. The protein levels were
`determined by the method of Bradford using reagents purchased
`from Bio-Rad.
`
`Determination of apoptotic cell death in tissue
`
`tissue, unfixed, dissociated
`Following dissociation of
`epithelial cells from kidney tissue were washed again in
`PBS and then slowly injected into cold methanol (4 °C, 100%)
`to a final concentration of 70% methanol, and kept at 4 °C
`until staining. Methanol-fixed cell suspensions were stained by
`an indirect
`immunofluorescence technique. Detection of
`apoptotic cells in tissue was performed using PE-conjugated
`rabbit anti-cleaved caspase-3 IgG (BD Bioscience, San Diego,
`CA) according to the vendor's protocol after gating a cell
`population. The percentage of apoptotic cells was analyzed by
`flow cytometry.
`
`DNA fragmentation
`
`DNA fragmentation was evaluated by TdT-mediated dUTP
`nick-end labeling (TUNEL) assay. After the TUNEL reaction,
`cells or histologic paraffin-embedded sections were analyzed by
`fluorescence microscopy. The green fluorescence of fluorescein
`isothiocyanate (FITC) was recorded with excitation at 488 nm
`through a 515-nm bandpass, together with the transmission
`image.
`
`Cellular oxidative damage
`
`Intracellular hydrogen peroxide concentrations were deter-
`mined using a ferric-sensitive dye, xylenol orange, as described
`
`Rigel Exhibit 1028
`Page 2 of 8
`
`

`

`46
`
`J.H. Lee, J.-W. Park / Free Radical Biology & Medicine 42 (2007) 44–51
`
`subsequently subjected to immunoblot analysis using appro-
`priate antibodies. Immunoreactive antigen was then recog-
`nized by using horseradish peroxidase-labeled anti-rabbit IgG
`and an enhanced chemiluminescence detection kit (Amer-
`sham Pharmacia Biotech).
`
`Statistical analysis
`
`The difference between two mean values was analyzed by
`Student's t test and was considered to be statistically significant
`when p < 0.05.
`
`Results
`
`IDPm activity
`
`To study the relationship between IDPm activity and
`ionizing radiation-induced apoptotic cell death, mice were
`administered 25 mg/kg oxalomalate, a competitive inhibitor of
`IDPm, once daily for 10 days. When mice were unexposed
`and exposed to 2 Gy of γ irradiation, the enzyme activity of
`IDPm in the kidneys of oxalomalate-treated mice was
`decreased about 20% compared with that of the control mice
`(Fig. 1). Because cellular antioxidants act
`in a concerted
`manner as a team, it is important to investigate whether the
`modulation of IDPm activity caused concomitant alterations in
`the activity of other major antioxidant enzymes. The reduced
`activity of IDPm in the kidneys of the oxalomalate-treated
`mice did not significantly alter
`the activities of other
`antioxidant enzymes such as SOD, catalase, glucose 6-
`phosphate dehydrogenase, glutathione peroxidase, and glu-
`tathione reductase (data not shown), suggesting that
`the
`reduction of IDPm activity did not affect
`the activities of
`other enzymes involved in antioxidation.
`
`Apoptosis determination
`
`The effects of ionizing radiation and IDPm activity on the
`cellular markers of apoptosis were studied to determine
`whether these would correlate with changes in the apoptotic
`pathways. As shown in Fig. 2A,
`the percentage of active
`
`[13]. Thiobarbituric acid-reactive substances (TBARS) were
`determined as an independent measurement of lipid peroxida-
`tion. The tissue homogenates (500 μl) were mixed with 1 ml
`TBA solution (0.375% thiobarbituric acid in 0.25 N HCl
`containing 15% (w/w) trichloroacetic acid and heated at 100 °C
`for 15 min. Then the reaction was stopped on ice, and the
`absorbance was measured at 535 nm [14]. The protein carbonyl
`content was determined spectrophotometrically using the
`DNPH-labeling procedure as described [15]. 8-Hydroxy-2-
`deoxyguanosine (8-OH-dG)
`levels of kidney tissue were
`estimated by using a fluorescent binding assay as described
`by Struthers et al. [16]. DNA damage was visualized with
`avidin-conjugated TRITC (1:200 dilution)
`for
`fluorescent
`microscopy with 540 nm excitation and 588 nm emission.
`
`Mitochondrial redox staus and damage
`
`NADPH was measured using the enzymatic cycling
`method as described by Zerez et al. [17] and expressed as
`the ratio of NADPH to the total NADP pool. The
`concentration of total glutathione was determined by the rate
`of
`formation of 5-thio-2-nitrobenzoic acid at 412 nm
`(ε = 1.36 × 104 M−1 cm−1) as described by Akerboom and
`Sies [18], and oxidized glutathione (GSSG) was measured by
`the DTNB-GSSG reductase recycling assay after treating GSH
`with 2-vinylpyridine. The mitochondrial membrane potential
`(Δφm) was semiquantitatively determined using the mitochon-
`drial-specific lipophilic fluorescent cationic probe JC-1 as
`described [19]. Following dissociation of
`tissue, unfixed,
`dissociated cells were washed and resuspended in PBS,
`supplemented with 10 μg/ml JC-1. Then cells were incubated
`for 15 min at room temperature in the dark, washed, and
`resuspended in PBS for immediate flow cytometry using a
`FACScan cytometer. Photomultiplier settings were adjusted to
`detect JC-1 monomers and J-aggregate fluorescence on the
`FL1 (530 nm) and FL2 (585 nm) detectors, respectively. The
`fluorescence ratio at those wavelengths was used to monitor
`changes in mitochondrial membrane potential. Fragmented
`cells and debris were excluded from measurements by gating
`the remaining intact cells in a forward and side scatter
`analysis. Data were analyzed using the CellQuest software
`(Becton Dickinson)
`to calculate the percentage of JC-1-
`positive cells. To evaluate the levels of mitochondrial ROS
`dissociated cells from tissue in PBS were incubated for
`20 min at 37 °C with 5 μM DHR 123 and cells were washed
`and resuspended in complete growth media, and ionizing
`radiation was applied to the cells. The cells were then
`incubated for an additional 40 min. FACS was used for
`fluorescence intensity quantification. Intracellular ATP levels
`were determined by using luciferin-luciferase as described
`[20]. Light emission was quantitated in a Turner Designs TD
`20/20 luminometer (Stratec Biomedical Systems, Germany).
`
`Immunoblot analysis
`
`Proteins were separated on 10–12.5% SDS-polyacryla-
`mide gel,
`transferred to nitrocellulose membranes, and
`
`Fig. 1. Activity of IDPm in the kidneys of the control and the oxalomalate-
`administered mice unexposed and exposed to γ irradiation. Each value
`represents the mean ± SD of samples from five animals. *p < 0.01 vs unirradiated
`control mice. #p < 0.01 vs irradiated control mice.
`
`Rigel Exhibit 1028
`Page 3 of 8
`
`

`

`J.H. Lee, J.-W. Park / Free Radical Biology & Medicine 42 (2007) 44–51
`
`47
`
`tion were evaluated. It is well established that oxidative stress
`in various cells usually leads to accumulation of potent,
`cytotoxic lipid peroxides such as malondialdehyde (MDA) and
`4-hydroxynonenal [21]. Exposure of 2 Gy of γ-irradiation
`increased the level of MDA in the kidney of the control mice;
`however, the increase in the MDA content of the kidneys of
`the oxalomalate-administered mice was significantly higher
`than that of the control mice (Fig. 3B). To determine whether
`reduced IDPm activity enhanced the sensitivity of protein
`damage, we performed carbonyl content measurements for
`protein oxidation after exposure to ionizing radiation. When 2
`Gy of ionizing radiation was exposed,
`the kidneys of the
`oxalomalate-administered mice elicited an approximately 4.3-
`fold increase of carbonyl groups, compared to the unirradiated
`mice. Although the carbonyl content of the control mice also
`increased with irradiation, the increase was significantly lower
`than that of the oxalomalate-administered mice (Fig. 3C). The
`reaction of intracellular ROS with DNA resulted in numerous
`forms of base damage, and 8-OH-dG is one of the most
`abundant and most studied lesions generated. 8-OH-dG has
`been used as an indicator of oxidative DNA damage in vivo
`and in vitro [22]. Recently, it has been shown that 8-OH-dG
`level is specifically measured by a fluorescent binding assay
`using avidin-conjugated TRITC [16]. As shown in Fig. 3E, the
`fluorescent intensity which reflects the endogenous levels of
`8-OH-dG in DNA was significantly increased in the kidneys of
`the oxalomalate-administered mice compared to control mice
`on exposure to ionizing radiation. These results indicate that
`IDPm appears to protect mice from oxidative damage caused
`by ionizing radiation.
`
`Mitochondrial redox status and damage
`
`One important parameter of GSH metabolism is the ratio of
`GSSG/total GSH (GSHt) which may reflect the efficiency of
`GSH turnover. When the mice were exposed to 2 Gy of
`γ-irradiation, the ratio of mitochondrial [GSSG]/[GSHt] was
`significantly higher
`in the kidneys of
`the oxalomalate-
`administered mice than the control mice (Fig. 4A). These
`data indicate that GSSG in the kidney mitochondria of the
`oxalomalate-administered mice was not reduced as efficiently
`as in that of the control mice. NADPH, required for GSH
`generation by glutathione reductase, is an essential factor for
`the cellular defense against oxidative damage. The ratio for
`[NADPH]/[NADP+ + NADPH]
`(NADPt) was
`mitochondrial
`significantly decreased in mice treated with 2 Gy of
`γ-irradiation; however, the decrease in this ratio was much
`more pronounced in the kidneys of the oxalomalate-adminis-
`tered mice (Fig. 4B). Alterations in mitochondrial integrity
`and function may play an important role in the apoptotic
`cascade. MPT, associated with the opening of large pores in
`the mitochondrial membranes, is a very important event in
`apoptosis, and ROS is one of the major stimuli that change
`MPT [23]. The kidneys of
`the oxalomalate-treated mice
`showed a significant change in Δφm determined with JC-1
`(Fig. 4C). To determine if changes in MPT were accompanied
`by changes in intracellular ROS, the levels of intracellular
`
`Fig. 2. Effects of oxalomalate on ionizing radiation-induced apoptosis in
`dissociated kidney epithelial cells from mice. (A) Activation of caspase-3 by 2
`Gy of γ-irradiation can be followed by flow cytometry. Dissociated kidney cells
`from mice were stained with anti-cleaved caspase-3 IgG and analyzed by flow
`cytometry (left). Percentage of apoptotic cells calculated from data obtained by
`flow cytometry. Each value represents the mean ± SD of samples from five
`animals. (Right) *p < 0.01 vs irradiated control mice. (B) Characteristic DNA
`fragmentation in the kidney cells from mice was determined by the TUNEL
`assay and examined by fluorescence microscopy. TUNEL-positive cells were
`shown by bright FITC staining of nuclei.
`
`caspase-3-positive cells had increased and was significantly
`higher in the kidneys of the oxalomalate-treated mice than in
`the control mice on exposure to a clinically relevant dose of 2
`Gy. The DNA fragmentation evaluated by the TUNEL assay
`was significantly increased in the kidney cells of
`the
`oxalomalate-treated mice compared to the control mice when
`the mice were exposed to 2 Gy of γ-irradiation (Fig. 2B).
`
`Cellular oxidative damage
`
`To investigate whether the difference in apoptotic cell death
`of kidney cells from the oxalomalate-treated or the control
`mice on exposure to ionizing radiation is associated with ROS
`formation, the levels of intracellular hydrogen peroxide in the
`kidneys were evaluated. As shown in Fig. 3A, a significantly
`higher intracellular level of H2O2 was observed in the kidneys
`of the oxalomalate-administered mice compared to the control
`mice with the exposure of 2 Gy of γ-irradiation. These data
`strengthen the conclusion that IDPm provided protection from
`the ionizing radiation-induced apoptosis by decreasing the
`steady-state level of
`intracellular oxidants. As indicative
`markers of oxidative damage to cells,
`the occurrences of
`oxidative DNA damage, protein oxidation, and lipid peroxida-
`
`Rigel Exhibit 1028
`Page 4 of 8
`
`

`

`48
`
`J.H. Lee, J.-W. Park / Free Radical Biology & Medicine 42 (2007) 44–51
`
`Fig. 3. Effects of oxalomalate on the cellular oxidative damage of the kidneys of mice exposed to ionizing radiation. (A) Production of hydrogen peroxide in the
`kidneys of mice exposed to ionizing radiation was determined by the method described under Experimental procedures. Each value represents the mean ± SD (n = 5).
`*p < 0.01 vs irradiated control mice. (B) Lipid peroxidation of the kidneys of mice after exposure to ionizing radiation. The level of MDA accumulated in the mice
`unexposed and exposed to γ-irradiation was determined by using a TBARS assay. Each value represents the mean ± SD (n = 4). *p < 0.01 vs irradiated control mice. (C)
`Protein carbonyl content of the kidneys of mice exposed to ionizing radiation. Protein carbonyls were measured in cell-free extracts with the use of DNPH. Each value
`represents the mean ± SD (n = 5). *p < 0.01 vs irradiated control mice. (D) 8-OH-dG levels in the kidneys of irradiated mice. 8-OH-dG levels reflected by the binding of
`avidin-TRITC were visualized fluorescence microscope.
`
`peroxides in the mitochondria of mouse kidney were evaluated
`by confocal microscopy with the oxidant-sensitive probe DHR
`123. As shown in Fig. 4D, the intensity of fluorescence was
`significantly higher
`in the kidneys of
`the oxalomalate-
`administered mice compared to that in the mitochondria of
`kidneys from the control mice when mice were exposed to 2
`Gy of γ-irradiation. These results indicate that
`ionizing
`radiation most likely leads to increased mitochondrial injury
`while IDPm protects mitochondria from oxidative damage.
`Mitochondrial injury is often followed by the depletion of
`intracellular ATP level. As shown in Fig. 4E, when mice were
`exposed to 2 Gy of γ-irradiation the ATP level was
`significantly decreased in the kidneys of the oxalomalate-
`administered mice compared to the control mice, suggesting a
`protective role of IDPm against the loss of intracellular ATP
`levels.
`
`Modulation of the apoptotic marker proteins
`
`We evaluated changes in the apoptotic marker proteins as a
`result of
`ionizing radiation and the influence of
`IDPm
`expression on these proteins. The role of mitochondrial
`pathways of apoptosis in the ionizing radiation-induced cell
`death of mice kidneys was examined by immunoblot analysis
`of the abundance of Bax, Noxa, or PUMA, a proapoptotic
`protein. As shown in Fig. 5, the amount of Bax, Noxa, or
`
`PUMA was increased after exposure to ionizing radiation, and
`it was significantly increased in the kidneys of the oxaloma-
`late-administered mice compared to the control mice. The
`release of cytochrome c from mitochondria, a critical event
`provoking a cascade of caspases and eventually irreversible
`cell death [24], was increased after exposure to ionizing
`radiation. A marked increase of cytochrome c release was
`observed in the kidneys of the oxalomalate-administered mice
`compared to control mice. Caspase-3 activation in the mouse
`kidney was assessed by immunoblot analysis of kidney lysates
`from mice that had been exposed to 2 Gy of γ-irradiation.
`Ionizing radiation induced cleavage of caspase-3; however, the
`cleavage was significantly increased by the reduced activity of
`IDPm.
`Ionizing radiation also induced the formation of
`fragments which represents proteolytic cleavage of PARP,
`indicating an oncoming apoptotic process. The cleaved
`products of PARP increased markedly in the kidneys of the
`oxalomalate-administered mice compared to the control mice
`on exposure to ionizing radiation. Taken together, ionizing
`radiation-induced cleavage of procaspase-3 into the active
`form of caspase-3 and caspase-3 induces degradation of PARP.
`Ionizing radiation induces double-strand breaks in cells. One
`of the key components in damaged DNA recognition and
`signaling after γ-irradiation is the tumor suppressor protein
`p53 [25]. p53 protein levels determined by Western blotting
`were increased as a result of ionizing radiation and the reduced
`
`Rigel Exhibit 1028
`Page 5 of 8
`
`

`

`J.H. Lee, J.-W. Park / Free Radical Biology & Medicine 42 (2007) 44–51
`
`49
`
`Fig. 4. Effects of oxalomalate on the mitochondrial redox status and the mitochondrial function of the kidneys of mice exposed to ionizing radiation. Ratio of GSSG
`versus the total GSH pool (A) and NADPH versus the total NADP pool (B) in kidney mitochondria from mice. Each value represents the mean ± SD (n = 5). *p < 0.01
`vs irradiated control mice. (C) Bivariate JC-1 analysis of mitochondrial membrane potential in the dissociated kidney cells from mice by flow cytometry. (D) Effect of
`oxalomalate on mitochondrial ROS generation. DHR 123 was employed to detect mitochondrial ROS. Fluorescence was measured by flow cytometry. (E) Effect of
`oxalomalate on the levels of intracellular ATP. The kidneys of mice were assayed for intracellular ATP content. Each value represents the mean ± SD (n = 5). *p < 0.01
`vs irradiated control mice.
`
`activity of IDPm by oxalomalate enhanced the radiation-
`induced up-regulation of p53 levels. p21, as a regulator protein
`and a p53 target gene, modulates the kinase activities of a
`variety of cyclin-dependent kinases [26]. The p21 induction
`was apparent in the mouse kidney after γ-irradiation, and the
`
`Fig. 5. Immunoblot analysis of apoptotic marker proteins in the kidney cells
`from mice exposed to ionizing radiation. Cell extracts were subjected to 10-
`12.5% SDS-PAGE and immunoblotted with antibodies against p53, Bax,
`PUMA, Noxa, cytochrome c, cleaved caspase-3, cleaved PARP, or p21. β-Actin
`was run as an internal control.
`
`level of induction was much higher in the kidneys of the
`oxalomalate-administered mice.
`
`Discussion
`
`Ionizing radiation is toxic to living cells and organisms
`because it induces deleterious structural changes in essential
`biomolecules. A significant part of the initial damage done to
`U
`OH, which
`cells by ionizing radiation is due to formation of
`reacts with almost all cellular components to induce oxidative
`damage to DNA,
`lipid peroxidation, and protein oxidation
`[3,27,28]. DNA is a particularly important target, suffering
`double- and single-strand breaks, deoxyribose damage, and
`base modification. Of the total damage to DNA caused by
`ionizing radiation, as much as 80% may result from radiation-
`induced water-derived free radicals and secondary carbon-based
`radicals [1]. In addition to the generation of hydroxyl radicals,
`the hydrated electrons formed by ionizing radiation can reduce
`
`
`it to O2–. O2– can dismutate to H2O2 with the possibility of extra
`U
`OH production by a metal-catalyzed Fenton reaction [2]. Free
`radicals can also initiate a variety of cellular signal transduction
`pathways that may either aid the cell in coping with the excess
`oxidative stress resulting from radiation or set into motion
`pathways that lead to the destruction of cells damaged beyond
`the repair capabilities of the cell [29,30]. Therefore, there are
`several reports to suggest that antioxidant enzymes may prevent
`oxidative damage and cell death including apoptosis caused by
`ionizing radiation.
`
`Rigel Exhibit 1028
`Page 6 of 8
`
`

`

`50
`
`J.H. Lee, J.-W. Park / Free Radical Biology & Medicine 42 (2007) 44–51
`
`IDPm is a key enzyme in cellular defense against oxidative
`damage by supplying NADPH in the mitochondria. NADPH is
`an essential cofactor for the regeneration of GSH, the most
`abundant
`low-molecular-mass thiol
`in most organisms, by
`glutathione reductase in addition to its critical role for the
`activity of the NADPH-dependent thioredoxin system [9,10].
`Mitochondrial GSH becomes critically important against ROS-
`mediated damage because it not only functions as a potent
`antioxidant but is also required for the activities of mitochon-
`drial glutathione peroxidase and mitochondrial phospholipid
`hydroperoxide glutathione peroxidase [31], which removes
`mitochondrial peroxides. The oxidized form of thioredoxin,
`with a disulfide bridge between the half-cystines, can be
`reduced by NADPH in the presence of a flavoprotein,
`thioredoxin reductase [32]. Reduced thioredoxin may provide
`reducing equivalents to mitochondrial thioredoxin peroxidase
`family/peroxiredoxin family including peroxiredoxin III/protein
`SP-22 [33–35] and peroxiredoxin V/AOEB166 [36]. Therefore,
`any mitochondrial NADPH producer,
`if present, becomes
`critically important for cellular defense against ROS-mediated
`damage. Suppression of mitochondrial NADPH and GSH levels
`by reduction of IDPm activity by oxalomalate in turn enhanced
`the oxidative stress and concomitant ROS-mediated cell death.
`Therefore, it was plausible to assume that IDPm may play a role
`in preventing apoptosis caused by ionizing radiation in cells.
`The aim of the present work was to evaluate the role of IDPm in
`protecting mice from ionizing radiation in regard to apoptotic
`cell death, cellular redox status, mitochondrial dysfunction, and
`oxidative damage to cells. In the present study, a temporal
`pattern of events reflecting ionizing radiation-induced apoptosis
`was observed, starting from the elevation of ROS level,
`followed by MPT alteration, caspase-3 activation, and DNA
`fragmentation. The reduction of IDPm activity by oxalomalate
`significantly increased ROS level and enhanced the whole
`apoptotic pathway.
`The involvement of mitochondria in apoptosis has been
`extensively discussed [37]. All the changes caused by ionizing
`radiation are compatible with mitochondrial failure, encom-
`passing reduced production of ATP, generation of ROS, and
`modulation of mitochondrial membrane potential. A clear
`enhancement of such damages in the oxalomalate-treated mice
`compared to the control mice suggests that IDPm prevents a
`deterioration of the bioenergetic state.
`Cleavage of caspase-3 and its target protein such as PARP, a
`signature event of apoptosis, was induced by ionizing radiation.
`The reduction of IDPm activity by oxalomalate enhanced
`programmed cell death by increasing apoptotic features
`including caspase activation, and increasing proapoptotic
`molecules such as Bax, Noxa, and PUMA, presumably via
`preservation of redox status. Wild-type p53 tumor suppressor
`protein has been shown to be functionally necessary for growth
`inhibition and apoptosis following exposure to ionizing
`radiation [38]. Levels of p53 were increased in mouse kidney
`after radiation treatment. The reduction of IDPm activity
`enhanced the radiation-induced increase in p53 levels, which
`correlates with an enhancement
`in radiation-induced DNA
`damage.
`
`ionizing radiation-induced apoptosis by
`Regulation of
`oxalomalate will likely have significance in cancer treatment.
`Cancer cells increasingly become resistant
`to consecutive
`administration of chemotherapeutic drugs and radiation. There-
`fore, enhancement of tumor radioresponse may improve the
`therapeutic ratio of radiotherapy. This study indicates that
`oxalomalate enhances the susceptibility of ionizing radiation-
`induced apoptosis through inhibiting IDPm activity. The
`ovserved effects of oxalomalate in mice supported here offer
`the possibility of developing a modifier of radiation therapy.
`In conclusion, the present study demonstrates that IDPm
`abrogates the ionizing radiation-induced early production of
`ROS, leading to protection against apoptotic cell death. In this
`regard, oxalomalate, an inhibitor of IDPm, could play a role as a
`rediosensitizer in tumor radiotherapy.
`
`Acknowledgments
`
`This work was supported by grants of the National R and
`D Program for Cancer Control, Ministry of Health and
`Welfare, Republic of Korea (0420150-1), and a Nuclear
`Research Program from the Korea Science and Engineering
`Foundation (M2-0513-000102-05-B09-00-102-10).
`
`References
`
`[1] von Sonntag, C. The chemical basis of radiation biology. Taylor and
`Francis, London; 1987.
`[2] Ewing, D.; Jones, S. R. Superoxide removal and radiation protection in
`bacteria. Arch. Biochem. Biophys. 254:53–62; 1987.
`[3] Cerutti, P. A. Prooxidant states and tumor promotion. Science 227:
`375–380; 1985.
`[4] Chen, Y. R.; Wang, X.; Templeton, D.; Davis, R. J.; Tan, T. H. The role of
`c-Jun N-terminal kinase (JNK) in apoptosis induced by ultraviolet C and
`gamma radiation. duration of JNK activation may determine cell death and
`proliferation. J. Biol. Chem. 271:31929–31936; 1996.
`[5] McCord, J. M.; Fridovich, I. Superoxide dismutase. an enzymic function
`for erythrocuprein (hemocuprein). J. Biol. Chem. 244:6049–6055; 1969.
`[6] Chance, B.; Sies, H.; Boveris, A. Hydroperoxide metabolism in
`mammalian organs. Physiol. Rev. 59:527–605; 1979.
`[7] Buttke, T. M.; Sandstrom, P. A. Oxidative stress as a mediator of apoptosis.
`Immunol. Today 15:7–10; 1994.
`[8] Koshland, D. E., Jr.;

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket