You are on page 1of 7

206

Maturation increases superoxide radical


production without increasing oxidative damage in
the skeletal muscle of hooded seals
(Cystophora cristata)
J.P. Vázquez-Medina, N.O. Olguı́n-Monroy, P.D. Maldonado, A. Santamarı́a,
M. Königsberg, R. Elsner, M.O. Hammill, J.M. Burns, and T. Zenteno-Savı́n

Abstract: Diving vertebrates represent unique models for the study of the physiological responses to reactive oxygen spe-
cies (ROS) production and oxidative stress because of their adaptability to cope with dive-derived ROS production. We
hypothesized that in the skeletal muscle of a diving mammal, the hooded seal (Cystophora cristata (Erxleben, 1777)),
ROS production increases with maturation but the accumulation of oxidative damage does not. To test this, we analyzed
the tissue capacity to produce ROS, the accumulation of oxidative damage, and the activity and protein content of the
cooper, zinc, and manganese dependent superoxide dismutases (Cu,ZnSOD, MnSOD) in skeletal muscle from neonates,
weaned pups, and adult hooded seals. Our results showed higher tissue capacity to produce ROS, higher Cu,ZnSOD and
MnSOD activities, and higher MnSOD protein content in adult seals than in pups. No differences in oxidative damage to
lipids, proteins, or DNA were detected among groups. Results suggest that increased SOD activity likely counters the oxi-
dative damage commonly associated with increased ROS production. These findings highlight the unusual tolerance of
skeletal muscle of seals to increased ROS production.
Résumé : Les vertébrés plongeurs sont des modèles uniques pour l’étude des réactions physiologiques à la production
d’espèces d’oxygène réactif (ROS) et du stress oxydatif à cause de leur adaptabilité pour faire face à la production de
ROS produits durant la plongée. Nous émettons l’hypothèse selon laquelle, dans le muscle squelettique du phoque à capu-
chon (Cystophora cristata (Erxleben, 1777)), la production de ROS augmente en fonction de la maturation, mais non l’ac-
cumulation de dommages oxydatifs. Afin de tester l’hypothèse, nous analysons la capacité du tissu à produire des ROS,
l’accumulation de dommages oxydatifs, ainsi que l’activité et le contenu protéinique des superoxydes dismutases dépen-
dantes du cuivre, du zinc et du manganèse (Cu,ZnSOD, MnSOD) dans le muscle squelettique chez des phoques à capu-
chon nouveau nés, des petits sevrés et des adultes. Les tissus ont une plus forte capacité de production de ROS et
possèdent des activités plus élevées de Cu,ZnSOD et de MnSOD et des contenus protéiniques plus importants de MnSOD
chez les phoques adultes que chez les petits. Aucune différence n’a pu être décelée entre les groupes dans les dommages
oxydatifs aux lipides, aux protéines et à l’ADN. Nos résultats laissent croire que l’activité accrue des SOD bloque vrai-
semblablement les dommages oxydatifs couramment associés à la production plus élevée de ROS. Ces observations souli-
gnent la tolérance exceptionnelle du muscle squelettique des phoques à la production accrue de ROS.
[Traduit par la Rédaction]

Received 9 June 2010. Accepted 1 December 2010. Published on the NRC Research Press Web site at cjz.nrc.ca on 12 February 2011.
J.P. Vázquez-Medina,1 N.O. Olguı́n-Monroy, and T. Zenteno-Savı́n.2 Planeación Ambiental y Conservación, Centro de
Investigaciones Biológicas del Noroeste, S.C, Mar Bermejo 195, Playa Palo de Santa Rita, La Paz, B.C.S., 23090, Mexico.
P.D. Maldonado. Laboratorio de Patologı́a Vascular Cerebral, Instituto Nacional de Neurologı́a y Neurocirugı́a ‘‘Dr. Manuel Velasco
Suárez’’, Insurgentes sur 3877, Colonia La Fama, Tlalpan, Mexico City, 14269, Mexico.
A. Santamarı́a. Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurologı́a y Neurocirugı́a ‘‘Dr. Manuel Velasco
Suárez’’, Insurgentes sur 3877, Colonia La Fama, Tlalpan, Mexico City, 14269, Mexico.
M. Königsberg. Laboratorio de Bioenergética y Envejecimiento Celular, Departamento de Ciencias de la Salud, Universidad Autónoma
Metropolitana-Iztapalapa, Avenida San Rafael Atlixco No. 186, Colonia Vicentina, Delegación Iztapalapa, Mexico City, 09340, Mexico.
R. Elsner. Institute of Marine Sciences, University of Alaska Fairbanks, P.O. Box 757220, Fairbanks, AK 99775-7220, USA.
M.O. Hammill. Maurice Lamontagne Institute, Department of Fisheries and Oceans, 850, route de la Mer, Mont-Joli, QC G5H 3Z4,
Canada.
J.M. Burns. Department of Biological Sciences, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA.
1Present address: School of Natural Sciences, University of California Merced, 5200 North Lake Road, Merced, CA 95343, USA.
2Corresponding author (e-mail: tzenteno04@cibnor.mx).

Can. J. Zool. 89: 206–212 (2011) doi:10.1139/Z10-107 Published by NRC Research Press
Vázquez-Medina et al. 207

Introduction Alaska Anchorage. Samples were imported into the United


States under marine mammal permits 782-1399 and 782-
Oxygen consumption by animal cells is essential for the
1694-02, and into Mexico under the terms of permits issued
production of the energy needed to maintain cellular func-
tions and metabolic activity. Paradoxically, oxygen-mediated by Instituto Nacional de Ecologı́a and Procuradurı́a Federal
ATP production via the electron transport chain is accompa- de Protección al Ambiente.
nied by the production of potentially damaging reactive oxy- Fourteen healthy hooded seals were captured on the pack
gen species (ROS) (Halliwell and Gutteridge 2007). During ice near the Magdalen Islands, Gulf of St. Lawrence, Can-
basal metabolic conditions, 0.1% of the oxygen consumed is ada (approximately 47823’N, 61852’W). Animals were cate-
transformed into superoxide radical (O2–) (Fridovich 2004). gorized visually as adults (5 reproductive females; body
O2– is dismutated in the mitochondria by the action of the mass (mean ± SE) = 249.3 ± 25.2 kg) or pups. Pups were
enzyme manganese-dependent superoxide dismutase categorized as neonates (3 males and 1 female; body
(MnSOD), and in the cytosol, by the copper and zinc con- mass = 21.6 ± 3.3 kg; age = 1 day) or weaners (3 females
taining superoxide dismutase (Cu,ZnSOD) (Weisiger and and 2 males; body mass = 48.1 ± 4.5 kg; age = 5–14 days)
Fridovich 1973). O2– dismutation results on hydrogen per- following Bowen et al. (1987). Seals were physically re-
oxide (H2O2) generation and the potential production of strained, drugged with diazepam (0.2–0.3 mgkg–1) or
highly reactive hydroxyl (OH) radicals (Boveris and tiletamine–zolazepam (0.7–1.0 mgkg–1), and sacrificed us-
Chance 1973). ing methods approved for scientific collection by Fisheries
Excessive ROS generation and (or) inefficient ROS scav- and Oceans Canada. Tissue sample collection was per-
enging by enzymatic and nonenzymatic antioxidants lead to formed in parallel with other studies (Burns et al. 2007,
the pathophysiological condition of oxidative stress (Sies 2010; Lestyk et al. 2009) to maximize the use of the sam-
1985). During this condition, oxidative damage is promoted ples obtained from the sacrificed animals. Samples of long-
by the reaction of ROS with lipids, proteins, and DNA (Hal- isimus dorsi muscle (approximately 2 g) were obtained by
liwell and Gutteridge 2007). ROS production, oxidative dissection, placed in cryogenic vials, and stored at –80 8C
damage accumulation, and antioxidant enzymes loss of func- until analyzed.
tion increase with age progression in a variety of animal
systems (Fraga et al. 1990; Kaneko et al. 1996; Starke-Reed Biochemical assays
and Oliver 1989; Sohal et al. 1993; Barja 2002).
To our knowledge, no information on maturation-related Superoxide radical production
increases in ROS production, oxidative damage accumula- Endogenous O2– production was measured following the
tion, or antioxidant enzymes in seals or in any other marine procedures proposed by Drossos et al. (1995) in tissue slices
mammal is yet available. Although there is a considerable (approximately 50 mg) placed in test tubes containing
body of literature dealing with oxidative stress and aging in Krebs–Henseleit buffer. O2– production was calculated by
several animal species, there is very little information avail- monitoring the change of ferricytochrome c to ferrocyto-
able on oxidative stress and development. Seals represent a chrome c as previously described (Zenteno-Savı́n et al.
unique model for the study of developmental-related 2002). Results were expressed as nanomoles of O2– per mi-
changes in oxidative stress variables because seals, like nute per gram of wet tissue.
other diving vertebrates, have an enhanced antioxidant sys-
tem that protects them against the potentially damaging ef- Oxidative damage
fects of diving-induced ischemia–reperfusion, a condition Oxidative damage to lipids was measured by calculating
that increases ROS production (Elsner et al. 1998; Zenteno- the tissue content of thiobarbituric acid reactive substances
Savı́n et al. 2002; Wilhem Filho et al. 2002; Hermes-Lima (TBARS). Briefly, samples were homogenized in two vol-
and Zenteno-Savı́n 2002; Vázquez-Medina et al. 2006, umes of 0.9% NaCl as described previously (Zenteno-Savı́n
2007, 2010; Valdivia et al. 2007; Zenteno-Savı́n et al. et al. 2002; Vázquez-Medina et al. 2007). TBARS content
2010). In this study, we compared muscle ROS production, (nanomoles per gram of wet tissue) was calculated from a
oxidative damage accumulation, and SOD activities and pro- standard curve of malondialdehyde bis-(diethylacetal) run in
tein content among three age groups of hooded seals (Cysto- parallel with the samples. Oxidative damage to proteins was
phora cristata) (Erxleben, 1777). We hypothesize that determined by quantifying the tissue content of protein car-
because seals are adapted to tolerate diving-induced ROS bonyls. Samples were homogenized (1:20 m/v) in ice-cold
production, muscle of hooded seals will not accumulate oxi- 5% sulfosalicylic acid and centrifuged at 19 000 g at 4 8C
dative damage with maturation. Moreover, because diving for 5 min. Protein carbonyls content was measured by using
capacity and diving behavior of hooded seals develop with 10 mmolL–1 DNPH as previously described (Vázquez-
maturation (Burns et al. 2007, Folkow et al. 2010), we hy- Medina et al. 2007). Results were expressed as nanomoles
pothesize that the development of hooded seals is accompa- of protein carbonyls per gram of wet tissue. Oxidatively
nied by an increase in SOD activity and protein content. modified DNA content was measured by quantifying the
concentration of 8-oxo-7,8-dihydro-2’-deoxyguanosine (8-
Materials and methods oxodGuo). DNA was isolated by following the chaotropic-
NaI method (Wang et al. 1994) with modifications (Matos
Animal handling and sample collection et al. 2001). Frozen tissue (approximately 500 mg) was ho-
Animal capture and handling protocols were reviewed and mogenized in 5 mL of lysis buffer (0.32 molL–1 sucrose,
approved by Fisheries and Oceans Canada and the Institu- 5 mmolL–1 EDTA, 1% Triton X-100, 5 mmolL–1 MgCl2,
tional Animal Care and Use Committee of the University of 10 mmolL–1 Tris-HCl, 0.1 mmolL–1 desferroxamine,

Published by NRC Research Press


208 Can. J. Zool. Vol. 89, 2011

pH 7.5) and centrifuged at 1500g for 10 min. Pellets were Rad Laboratories, Hercules, California, USA). Twenty mi-
resuspended in 10 mmolL–1 Tris-HCl buffer (5 mmolL–1 crograms of total protein were resolved in 4%–12% SDS
EDTA, 10% SDS, 0.15 mmolL–1 desferroxamine, pH 8.0). gels under denaturing conditions. Proteins were electroblot-
RNAase A (10 mgmL–1) and RNAase T1 (1000 UmL–1) ted using the Bio-Rad Trans-Blot SD semi-dry cell onto
were added. Samples were incubated for 1 h at 37 8C. Sam- 0.45 mm nitrocellulose membranes. Membranes were
ples were incubated again under the same conditions after blocked for 1 h at room temperature with 3% BSA in phos-
the addition of proteinase K (20 mgmL–1). Samples were phate buffer saline containing 0.05% Tween-20 and incu-
then centrifuged at 5000g for 15 min at 4 8C. Liquid phases bated overnight with rabbit polyclonal antibodies against
were collected and a 7.6 molL–1 NaI solution (40 mmolL–1 mammalian MnSOD, Cu,ZnSOD (Assay Designs, Ann Ar-
Tris-HCl, 20 mmolL–1 EDTA-Na2, 0.3 mmolL–1 desferrox- bor, Michigan, USA; catalogue Nos. SOD-111 and SOD-
amine, pH 8.0) was added. Samples were mixed with isopro- 100) diluted 1:4000 and a goat polyclonal antibody against
panol and stored overnight at –70 8C. Precipitates were actin (Santa Cruz Biotechnology, Santa Cruz, California,
collected by centrifugation at 9000g for 15 min and washed USA; catalogue No. sc-1616) diluted 1:1000. Membranes
with 60% isopropanol and 70% ethanol. Samples were then were washed, incubated with HRP-conjugated secondary
centrifuged at 9000g for 15 min. DNA pellets were dis- antibodies (Santa Cruz Biotechnology), rewashed, and de-
solved in 0.1 mmolL–1 desferroxamine. DNA concentrations veloped using the Bio-Rad Immun-Star WesternC kit. Inten-
were measured by spectrophotometry at 260 nm. DNA pu- sities of blot bands were quantified using Bio-Rad’s
rity was assessed by calculating the A260/A280 ratio. Only Quantity One software. Values were expressed as percentage
sample extractions with ratios >1.75 were used. For each of change from neonates after band intensities were normal-
sample, 100 mg of DNA were diluted in Milli-Q water and ized against actin.
1 molL–1 sodium acetate with nuclease P1 (5 UmL–1) was
added. Samples were incubated at 37 8C for 30 min, after Statistics
which 1 molL–1 Tris-HCl buffer (pH 7.4) and acid phospha- Differences in responses among age groups were detected
tase (3 UmL–1) were added and samples were incubated at by one-way analysis of variance (ANOVA) with Bonferroni
37 8C for 1 h. Aqueous layers were obtained by centrifuga- post hoc tests. Groups were considered statistically different
tion and analyzed for 8-oxodGuo content by using high- at p < 0.05. Statistical analyses were performed using the
performance liquid chromatography (HPLC) with electro- SYSTAT# 12.0 software (Cranes Software International
chemical (EC) detection as previously described (López- Ltd., Richmond, California, USA). Results are presented as
Diazguerrero et al. 2005). Digested DNA was injected into means ± SE.
an HPLC–EC system (Waters 600 C pump, Supelco supel-
cosil LC-18 reverse-phase column, 250 mm long  4.6 mm Results
inner diameter, particle size 5 mm), using 50 mmolL–1 PBS
buffer (pH 5.5, 8% methanol) as isocratic eluent at a flow O2– production increases with maturation in the muscle
rate of 1 mLmin–1. EC detection was performed using an of hooded seals
INTRO detector (Antec Leyden, Leiden, the Netherlands) Endogenous O2– production was measured as an index of
operated at 290 mV. Elution of unmodified nucleosides was the tissue capacity to produce ROS to evaluate whether ROS
simultaneously detected at 254 nm by using a 486 Waters production increases with development in hooded seal
UV spectrometer set. The molar ratio of 8-oxodGuo to de- muscle. O2– production was higher in adults than in neo-
oxy-guanosine (dGuo) was determined. Results were ex- natal pups (F = 23.065, p = 0.037). Values in weaned pups
pressed as the ratio of 8-oxodGugo to 106 dGuo. were intermediate between those of neonates and adults
(Fig. 1). These results show that tissue capacity to produce
SOD activity and protein expression ROS is higher in the muscle of adult hooded seals than in
Total SOD (MnSOD + Cu,ZnSOD) and MnSOD activities the muscle of developing animals.
were measured following the method of Oberley and Spitz
(1984). Samples were homogenized in lysis buffer Oxidative damage does not increase with maturation in
(10 mmolL–1 Tris-HCl, 15 mmolL–1 NaCl, 0.25 mmolL–1 the muscle of hooded seals
sucrose, 1% Triton X-100, pH 7.0) containing protease in- Muscle content of TBARS, protein carbonyls, and 8-
hibitors. To measure MnSOD activity, Cu,ZnSOD was in- oxodGuo was measured in neonates, weaned pups, and adult
hibited with diethyldithiocarbamate. Assays were run as seals to evaluate whether maturation increases oxidative
described previously (Herrera-Mundo et al. 2006). damage accumulation in the skeletal muscle of hooded seals.
Cu,ZnSOD activity was calculated by subtracting MnSOD There were no significant differences in the content of oxi-
activity from total SOD activity. Results were expressed as dative damage to lipids, proteins, or DNA among the three
units of total SOD, Cu,ZnSOD, or MnSOD activity per groups (Table 1) despite the increased O2– production ob-
milligram of protein. One unit of activity is defined as the served in the muscle of adult seals. These results suggest
amount of protein that inhibits 50% of NBT reduction. that hooded seals possess mechanisms to avoid the accumu-
Cu,ZnSOD and MnSOD protein expression was evaluated lation of oxidative damage commonly associated with in-
by Western blot. Samples were homogenized 1:10 (m/v) in creased ROS production.
50 mmolL–1 potassium phosphate buffer containing
1 mmolL–1 EDTA, 1% Triton X-100, 1% PMSF, and 1% SOD activity and MnSOD protein content increase with
protease inhibitor cocktail. Total protein content was deter- maturation in the muscle of hooded seals
mined by using the Bio-Rad Bradford Protein Assay (Bio- MnSOD, Cu,ZnSOD, and total SOD activities along with

Published by NRC Research Press


Vázquez-Medina et al. 209

Table 1. Oxidative damage accumulation does not increase with maturation in the skeletal
muscle of hooded seals (Cystophora cristata).

TBARS Protein carbonyls DNA damage


Age class (nmolg wet tissue–1) (nmolg wet tissue–1) (8-oxodGuo/106 dGuo)
Neonatal pups 0.05±0.02 106.61±32 1.0±0.2
Weaned pus 0.08±0.009 103.76±31 1.5±0.3
Adults 0.05±0.01 96.87±33 0.75±0.3
Note: TBARS, thiobarbituric acid reactive substances; 8-oxodGuo, 8-oxo-7,8-dihydro-2’-
deoxyguanosine; dGuo, deoxy-guanosine.

Fig. 1. Superoxide radical (O2–) production increases with matura- Hamilton et al. 2001; Pamplona 2008). In the present study,
tion in the skeletal muscle of hooded seals (Cystophora cristata). we show that ROS production in the skeletal muscle of
O2– production in skeletal muscle from neonatal pups, weaned hooded seals increases with maturation, but lipid peroxida-
pups, and adult hooded seals. Results are expressed as means ± SE. tion, protein oxidation, and oxidatively modified DNA con-
*, p < 0.05. tent do not.
O2– production in the skeletal muscle of adult hooded
seals is within the range of values reported for the muscle
of adult ringed seals (Pusa hispida (Schreber, 1775)),
whereas O2– production in the muscle of pups was closer
to that reported for the muscle of adult pigs (Zenteno-Savı́n
et al. 2002). Protein carbonyls levels found in the muscle of
hooded seals are generally lower than the levels reported for
the muscle of adult pigs and ringed seals (Vázquez-Medina
et al. 2007), whereas TBARS levels are within the same
range as those of pigs but lower than those of ringed seals
(Zenteno-Savı́n et al. 2002). Age-related accumulation of
oxidized proteins is a complex function involving the rates
of ROS formation, the antioxidant systems, and the rates of
degradation of oxidized proteins (Starke-Reed and Oliver
MnSOD and Cu,ZnSOD protein content were measured in 1989). Tissues from adult ringed seals showed higher rates
the muscle of neonatal, weaned pups, and adult hooded seals of ROS production during in vitro experiments that simulate
to evaluate the potential role of these antioxidant enzymes in dive-induced ischemia–reperfusion than tissues from non-
counteracting the observed increases in ROS production. To- diving adult pigs (Zenteno-Savı́n et al. 2002). However, rel-
tal SOD, MnSOD, and Cu,ZnSOD activities were higher ative to terrestrial species, tissues from ringed seals also
(total SOD, F = 80.298, p < 0.001; MnSOD, F = 14.534, showed enhanced activities of key antioxidant enzymes and
p = 0.001; Cu,ZnSOD, F = 88.167, p < 0.001) in adults high glutathione content, which likely counteract the poten-
than in neonatal or weaned pups, which did not differ signif- tially negative effects of dive-derived ROS production by
icantly (Figs. 2A–2C). In the same way, MnSOD protein preventing the accumulation of oxidative damage (Vázquez-
content was higher in adults than in neonantes or weaned Medina et al. 2006, 2007). Li et al. (2005), as observed in
pups (F = 5.059, p = 0.03) (Fig. 3A), whereas Cu,ZnSOD the current study, failed to find any increases in the accumu-
protein content did not differ among groups (F = 3.943, p < lation of 8-oxodGuo when comparing mature vs. developing
0.071) (Fig. 3B). These results show that SOD activity in- individuals of several cetacean species. In contrast, Gauthier
creases with maturation in the muscle of hooded seals and et al. (1999) reported an age-related increase in micronuclei
likely helps to counteract the observed increases in ROS cells (MNC) of developing bottlenose dolphins (Tursiops
production contributing to the avoidance of oxidative dam- truncatus (Montagu, 1821)), indicating that oxidative dam-
age accumulation. age may accumulate with development among some marine
mammals.
Studies in rats have shown that the activity of the DNA-
Discussion
repairing enzyme 8-oxoguanine DNA glycosylase (OGG1),
Mitochondria play a primary role in the production of which is responsible for 8-oxodGuo repair, can be stimu-
ROS in animal cells, and it has been widely documented lated by ROS derived from exercise training protocols
that this mitochondria-derived free radical production in (swimming included), resulting in a reduction of 8-oxodGuo
skeletal muscle increases with age, even during unperturbed accumulation (Ogonovszky et al. 2005; Radák et al. 2007).
metabolic conditions (Nohl and Hegner 1978; Sohal et al. Considering that the diving lifestyle of adult hooded seals
1995; Wei 1998; Mecocci et al. 1999; Cadenas and Davies exposes them to repetitive cycles of ischemia–reperfusion
2000; Marzani et al. 2004; Halliwell and Gutteridge 2007; (Elsner 1999), which promote ROS generation (Zenteno-
Figueiredo et al. 2008). There is also evidence for the accu- Savı́n et al. 2002), it is possible that dive-derived ROS pro-
mulation of oxidized molecules with age progression in sev- duction stimulates increases in OGG1 or other DNA repair
eral terrestrial mammal species (Cutler 1985; Garland et al. mechanisms, preventing the accumulation of oxidatively
1988; Mecocci et al. 1999; Davies and Delsignore 2001; modified DNA content.

Published by NRC Research Press


210 Can. J. Zool. Vol. 89, 2011

Fig. 2. Superoxide dismutase (SOD) activity increases with matura- Fig. 3. MnSOD protein expression increases with maturation in the
tion in the muscle of hooded seals (Cystophora cristata). (A) Total muscle of hooded seals (Cystophora cristata). (A) MnSOD and
SOD, (B) MnSOD, and (C) Cu,ZnSOD activities in muscle from (B) Cu,ZnSOD relative protein content in muscle from neonatal
neonatal pups, weaned pups, and adult hooded seals. One unit of pups, weaned pups and adult hooded seals. Results are expressed as
SOD activity is defined as the amount of enzyme that inhibits 50% means ± SE of the percent change from neonatal pups. Percent
of NBT reduction. Results are expressed as means ± SE. *, p < changes were obtained after calculating the ratios of MnSOD to ac-
0.05. tin or Cu,ZnSOD to actin. *, p < 0.05.

Both diving capacity and diving behavior of hooded seals


develop with maturation (Burns et al. 2007, Folkow et al.
2010). Because growing seals possess an increased anti-
oxidant capacity (Vázquez-Medina et al. 2010), we hypothe-
size that the development of hooded seals’ diving ability is
accompanied by the development of an enhanced anti-
oxidant capacity. This would allow them to tolerate both
dive-derived and maturation-related increases in ROS pro-
duction, and to prevent the accumulation of oxidatively
modified DNA, lipids, and proteins.
Our results support the former hypothesis, as we found
higher SOD activity and MnSOD protein content in adult
seals than in pups. Even though Cu,ZnSOD protein content
is not increased in adult animals, its sustained expression
seems to be enough to promote a maturation-related increase
in Cu,ZnSOD activity. Moreover, total SOD activity values
found in the muscle from pups of hooded seals are similar
to those reported from the skeletal muscle of sea turtles
(Valdivia et al. 2007), whereas the values in muscle from
adult hooded seals are more than two times higher than
those reported for adult pigs or the short-diving ringed seal
(Vázquez-Medina et al. 2006), suggesting that the
antioxidant system may also be increased in long- and

Published by NRC Research Press


Vázquez-Medina et al. 211

deep-diving seals, such as the hooded seal, than in short and Acad. Sci. U.S.A. 82(14): 4798–4802. doi:10.1073/pnas.82.14.
shallow divers. 4798. PMID:3860823.
In summary, our results show that tissue capacity to pro- Davies, K.J.A., and Delsignore, M.E. 2001. Degradation of oxidized
duce ROS increases with maturation in the muscle of proteins by the 20S proteasome. Biochimie, 83(3–4): 301–310.
hooded seals, but these maturation-related increases in ROS doi:10.1016/S0300-9084(01)01250-0. PMID:11295490.
production are not correlated with increases in oxidative Drossos, G., Lazou, A., Panagopoulos, P., and Westaby, S. 1995.
Deferoxamine cardioplegia reduces superoxide radical produc-
damage accumulation. The observed increases in SOD activ-
tion in human myocardium. Ann. Thorac. Surg. 59(1): 169–172.
ity and MnSOD protein content seem to counteract the in-
doi:10.1016/0003-4975(94)00726-N. PMID:7818317.
creased ROS production in adult seals and to prevent, at Elsner, R. 1999. Living in water: solutions to physiological pro-
least in part, the accumulation of oxidative damage. These blems. In Biology of marine mammals. Edited by J.E. Reynolds,
observed increases in SOD activity and MnSOD protein III and S.A. Rommel. Smithsonian Institution Press, Washing-
content also seem to be correlated with the rapid physiolog- ton, D.C. pp. 73–116.
ical development of hooded seals. More work is needed to Elsner, R., Øyasaeter, S., Almaas, R., and Saugstad, O.D. 1998.
elucidate the participation of other antioxidant mechanisms Diving seals, ischemia–reperfusion and oxygen radicals. Comp.
(enzymatic and nonenzymatic) and repair systems in coun- Biochem. Physiol. A Mol. Integr. Physiol. 119(4): 975–980.
teracting ROS production in seals, and to corroborate the doi:10.1016/S1095-6433(98)00012-9.
link between the antioxidant protection against dive- and Figueiredo, P.A., Mota, M.P., Appell, H.J., and Duarte, J.A. 2008.
maturation-related ROS production. The role of mitochondria in aging of skeletal muscle.
Biogerontology, 9(2): 67–84. doi:10.1007/s10522-007-9121-7.
Acknowledgements PMID:18175203.
Folkow, L.P., Nordøy, E.S., and Blix, A.S. 2010. Remarkable de-
A. Luna, V. Pérez, O. Lugo, and D. Rojas provided in- velopment of diving performance and migrations of hooded
struction and advice on analytic techniques. The Canadian seals (Cystophora cristata) during their first year of life. Polar
Coast Guard helicopter pilots Harrison McRae and Bruce Biol. 33(4): 433–441. doi:10.1007/s00300-009-0718-y.
Kendall provided logistic support in sample collection. The Fraga, C.G., Shigenaga, M.K., Park, J.W., Degan, P., and Ames,
Château Madelinot and Roger Simon provided laboratory B.N. 1990. Oxidative damage to DNA during aging: 8-hydroxy-
space. Lena Measures, Stephan Pillet, and Sam Turgeon 2’-deoxyguanosine in rat organ DNA and urine. Proc. Natl.
assisted with sample collection. Comments and suggestions Acad. Sci. U.S.A. 87(12): 4533–4537. doi:10.1073/pnas.87.12.
from Rudy Ortiz and two anonymous reviewers helped to 4533. PMID:2352934.
improve the manuscript. J.P.V.-M. is supported by Fridovich, I. 2004. Mitochondria: are they the seat of senescence?
Programa de Estudios de Posgrado (CIBNOR), UC-Mexus, Aging Cell, 3(1): 13–16. doi:10.1046/j.1474-9728.2003.00075.x.
CONACYT, and Secretarı́a de Educación Pública PMID:14965350.
fellowships. This research was funded by grants from Garland, D., Russell, P., and Zigler, J.S., Jr. 1988. The oxidative
SEMARNAT-CONACYT and CIBNOR. modification of lens proteins. Basic Life Sci. 49(1–2): 347–352.
PMID:3250491.
References Gauthier, J.M., Dubeau, H., Rassart, E., Jarman, W.M., and Wells,
R.S. 1999. Biomarkers of DNA damage in marine mammals.
Barja, G. 2002. Rate of generation of oxidative stress-related da- Mutat. Res. 444(2): 427–439. doi:10.1016/S1383-5718(99)
mage and animal longevity. Free Radic. Biol. Med. 33(9): 1167– 00106-0. PMID:10521683.
1172. doi:10.1016/S0891-5849(02)00910-3. PMID:12398924. Halliwell, B., and Gutteridge, J.M.C. 2007. Free radicals in biology
Boveris, A., and Chance, B. 1973. The mitochondrial generation of and medicine. Oxford University Press, New York.
hydrogen peroxide: general properties and effect of hyperbaric Hamilton, M.L., Van Remmen, H., Drake, J.A., Yang, H., Guo,
oxygen. Biochem. J. 134(3): 707–716. PMID:4749271. Z.M., Kewitt, K., Walter, C.A., and Richardson, A. 2001. Does
Bowen, W.D., Boness, D.J., and Oftedal, O.T. 1987. Mass transfer oxidative damage to DNA increase with age? Proc. Natl. Acad.
from mother to pup and subsequent mass loss by the weaned Sci. U.S.A. 98(18): 10469–10474. doi:10.1073/pnas.171202698.
pup in the hooded seal, Cystophora cristata. Can. J. Zool. PMID:11517304.
65(1): 1–8. doi:10.1139/z87-001. Hermes-Lima, M., and Zenteno-Savı́n, T. 2002. Animal response to
Burns, J.M., Lestyk, K.C., Folkow, L.P., Hammill, M.O., and Blix, drastic changes in oxygen availability and physiological oxida-
A.S. 2007. Size and distribution of oxygen stores in harp and tive stress. Comp. Biochem. Physiol. C Toxic. Pharmacol.
hooded seals from birth to maturity. J. Comp. Physiol. B 133(4): 537–556. doi:10.1016/S1532-0456(02)00080-7.
Biochem. Syst. Environ. Physiol.,177(6): 687–700. doi:10.1007/ Herrera-Mundo, M.N., Silva-Adaya, D., Maldonado, P.D., Galván-
s00360-007-0167-2. PMID:17576570. Arzate, S., Andrés-Martı́nez, L., Pérez-De La Cruz, V., Pedraza-
Burns, J.M., Skomp, N., Bishop, N., Lestyk, K., and Hammill, M. Chaverrı́, J., and Santamarı́a, A. 2006. S-Allylcysteine prevents
2010. Development of aerobic and anaerobic metabolism in the rat from 3-nitropropionic acid-induced hyperactivity, early
cardiac and skeletal muscles from harp and hooded seals. J. markers of oxidative stress and mitochondrial dysfunction.
Exp. Biol. 213(5): 740–748. doi:10.1242/jeb.037929. PMID: Neurosci. Res. 56(1): 39–44. doi:10.1016/j.neures.2006.04.018.
20154189. PMID:16806549.
Cadenas, E., and Davies, K.J.A. 2000. Mitochondrial free radical Kaneko, T., Tahara, S., and Matsuo, M. 1996. Non-linear accumu-
generation, oxidative stress, and aging. Free Radic. Biol. Med. lation of 8-hydroxy-2’-deoxyguanosine, a marker of oxidized
29(3–4): 222–230. doi:10.1016/S0891-5849(00)00317-8. PMID: DNA damage, during aging. Mutat. Res. 326(5–6): 277–285.
11035250. doi:10.1016/S0921-8734(96)90010-7.
Cutler, R.G. 1985. Peroxide-producing potential of tissues: inverse Lestyk, K.C., Folkow, L.P., Blix, A.S., Hammill, M.O., and Burns,
correlation with longevity of mammalian species. Proc. Natl. J.M. 2009. Development of myoglobin concentration and acid

Published by NRC Research Press


212 Can. J. Zool. Vol. 89, 2011

buffering capacity in harp (Pagophilus groenlandicus) and Sohal, R.S., Agarwal, S., and Sohal, B.H. 1995. Oxidative stress
hooded (Cystophora cristata) seals from birth to maturity. J. and aging in the Mongolian gerbil (Meriones unguiculatus).
Comp. Physiol. B Biochem. Syst. Environ. Physiol. 179(8): Mech. Ageing Dev. 81(1): 15–25. doi:10.1016/0047-6374(94)
985–996. doi:10.1007/s00360-009-0378-9. PMID:19565249. 01578-A. PMID:7475349.
Li, C.S., Wu, K.Y., Chang-Chien, G.P., and Chou, C.C. 2005. Ana- Starke-Reed, P.E., and Oliver, C.N. 1989. Protein oxidation and
lysis of oxidative DNA damage 8-hydroxy-2’-deoxyguanosine as proteolysis during aging and oxidative stress. Arch. Biochem.
a biomarker of exposures to persistent pollutants for marine Biophys. 275(2): 559–567. doi:10.1016/0003-9861(89)90402-5.
mammals. Environ. Sci. Technol. 39(8): 2455–2460. doi:10. Valdivia, P.A., Zenteno-Savı́n, T., Gardner, S.C., and Aguirre, A.A.
1021/es0487123. PMID:15884335. 2007. Basic oxidative stress metabolites in eastern Pacific green
López-Diazguerrero, N.E., Luna-López, A., Gutiérrez-Ruiz, M.C., turtles (Chelonia mydas agassizii). Comp. Biochem. Physiol. C
Zentella, A., and Königsberg, M. 2005. Susceptibility of DNA Toxic. Pharmacol. 146(1–2): 301–308. doi:10.1016/j.cbpc.2006.
to oxidative stressors in young and aging mice. Life Sci. 06.008.
77(22): 2840–2854. doi:10.1016/j.lfs.2005.05.034. PMID: Vázquez-Medina, J.P., Zenteno-Savı́n, T., and Elsner, R. 2006.
15979101. Antioxidant enzymes in ringed seal tissues: potential protection
Marzani, B., Pansarasa, O., and Marzaticov, F. 2004. Oxidative against dive-associated ischemia/reperfusion. Comp. Biochem.
stress and muscle aging: influence of age, sex, fiber composition Physiol. C Toxic. Pharmacol. 142(3–4): 198–204. doi:10.1016/j.
and function. Basic Appl. Myol. 14(1): 37–44. cbpc.2005.09.004.
Matos, H.R., Capelozzi, V.L., Gomes, O.F., Mascio, P.D., and Me- Vázquez-Medina, J.P., Zenteno-Savı́n, T., and Elsner, R. 2007.
deiros, M.H.G. 2001. Lycopene inhibits DNA damage and liver Glutathione protection against dive-associated ischemia/
necrosis in rats treated with ferric nitrilotriacetate. Arch. reperfusion in ringed seal tissues. J. Exp. Mar. Biol. Ecol.
Biochem. Biophys. 396(2): 171–177. doi:10.1006/abbi.2001. 345(2): 110–118. doi:10.1016/j.jembe.2007.02.003.
2611. PMID:11747294. Vázquez-Medina, J.P., Crocker, D.E., Forman, H.J., and Ortiz,
Mecocci, P., Fanó, G., Fulle, S., MacGarvey, U., Shinobu, L., Poli- R.M. 2010. Prolonged fasting does not increase oxidative da-
dori, M.C., Cherubini, A., Vecchiet, J., Senin, U., and Beal, mage or inflammation in postweaned northern elephant seal
M.F. 1999. Age-dependent increases in oxidative damage to pups. J. Exp. Biol. 213(14): 2524–2530. doi:10.1242/jeb.
DNA, lipids, and proteins in human skeletal muscle. Free Radic. 041335. PMID:20581282.
Biol. Med. 26(3-4): 303–308. doi:10.1016/S0891-5849(98) Wang, L., Hirayasu, K., Ishizawa, M., and Kobayashi, Y. 1994.
00208-1. PMID:9895220. Purification of genomic DNA from human whole blood by
Nohl, H., and Hegner, D. 1978. Do mitochondria produce oxygen isopropanol-fractionation with concentrated Nal and SDS.
radicals in vivo? Eur. J. Biochem. 82(2): 563–567. doi:10.1111/ Nucleic Acids Res. 22(9): 1774–1775. doi:10.1093/nar/22.9.
j.1432-1033.1978.tb12051.x. PMID:203456. 1774. PMID:8202389.
Oberley, L.W., and Spitz, D.R. 1984. Assay of superoxide dismu- Wei, Y.H. 1998. Oxidative stress and mitochondrial DNA muta-
tase activity in tumor tissue. Methods Enzymol. 105: 457–464. tions in human aging. Proc. Soc. Exp. Biol. Med. 217(1): 53–
doi:10.1016/S0076-6879(84)05064-3. PMID:6547201. 63. PMID:9421207.
Ogonovszky, H., Berkes, I., Kumagai, S., Kaneko, T., Tahara, S., Weisiger, R.A., and Fridovich, I. 1973. Mitochondrial superoxide
Goto, S., and Radák, Z. 2005. The effects of moderate-, simutase: site of synthesis and intramitochondrial localization. J.
strenuous- and over-training on oxidative stress markers, DNA Biol. Chem. 248(13): 4793–4796. PMID:4578091.
repair, and memory, in rat brain. Neurochem. Int. 46(8): 635– Wilhem Filho, D., Sell, F., Ribeiro, L., Ghislandi, M., Carrasquedo,
640. doi:10.1016/j.neuint.2005.02.009. PMID:15863241. F., Fraga, C.G., Wallauer, J.P., Simoes-Lopes, P.C., and Uhart,
Pamplona, R. 2008. Membrane phospholipids, lipoxidative damage M.M. 2002. Comparison between the antioxidant status of ter-
and molecular integrity: a causal role in aging and longevity. restrial and diving mammals. Comp. Biochem. Physiol. A Mol.
Biochim. Biophys. Acta, 1777(10): 1249–1262. doi:10.1016/j. Integr. Physiol. 133(3): 885–892. doi:10.1016/S1095-6433(02)
bbabio.2008.07.003. PMID:18721793. 00253-2.
Radák, Z., Kumagai, S., Nakamoto, H., and Goto, S. 2007. 8-Oxo- Zenteno-Savı́n, T., Clayton-Hernandez, E., and Elsner, R. 2002.
guanosine and uracil repair of nuclear and mitochondrial DNA Diving seals: are they a model for coping with oxidative stress?
in red and white skeletal muscle of exercise-trained old rats. J. Comp. Biochem. Physiol. C Toxic. Pharmacol. 133(4): 527–536.
Appl. Physiol. 102(4): 1696–1701. doi:10.1152/japplphysiol. doi:10.1016/S1532-0456(02)00075-3.
01051.2006. PMID:17204574. Zenteno-Savı́n, T., St. Leger, J., and Ponganis, P.J. 2010. Hypoxe-
Sies, H. 1985. Oxidative stress. Academic Press, San Diego. mic and ischemic tolerance in emperor penguins. Comp.
Sohal, R.S., Agarwal, S., Dubey, A., and Orr, W.C. 1993. Protein Biochem. Physiol. C Toxic. Pharmacol. 152(1): 18–23. doi:10.
oxidative damage is associated with life expectancy of house- 1016/j.cbpc.2010.02.007.
flies. Proc. Natl. Acad. Sci. U.S.A. 90(15): 7255–7259. doi:10.
1073/pnas.90.15.7255. PMID:8346242.

Published by NRC Research Press

You might also like