Vous êtes sur la page 1sur 10

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/20299986

Biosynthesis and regulation of superoxide


dismutase

Article in Free Radical Biology and Medicine · February 1988


DOI: 10.1016/0891-5849(88)90111-6 · Source: PubMed

CITATIONS READS

167 106

1 author:

Hosni M Hassan
North Carolina State University
133 PUBLICATIONS 5,016 CITATIONS

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Development of novel Salmonella control practices and integrated education program to reduce
Salmonellosis View project

Engineering Lactobacillus for biotherapy of IBD, colon cancer View project

All content following this page was uploaded by Hosni M Hassan on 01 December 2017.

The user has requested enhancement of the downloaded file.


Free Radical Biology & Medicine. Vol. 5. pp. 377-385, 1988 0891-5849/88 $3.00+ .00
Printedin the USA. All rightsreserved. © 1988PergamonPressplc

Review Article
BIOSYNTHESIS AND REGULATION OF SUPEROXIDE DISMUTASES

HOSNI M. HASSAN
Departments of Food Science and Microbiology, North Carolina State University, Raleigh, NC 27695-7624, U.S.A.

Abstract--The past two decades have witnessed an explosion in our understanding of oxygen toxicity. The
discovery of superoxide dismutases (SODs) (EC. 1.15.1.1), which specifically catalyze the dismutation of super-
oxide radicals (02-) to hydrogen peroxide (H202) and oxygen, has indicated that O2- is a normal and common
byproduct of oxygen metabolism. There is an increasing evidence to support the conclusion that superoxide radicals
play a major role in cellular injury, mutagenesis, and many diseases. In all cases SODs have been shown to protect
the cells against these deleterious effects. Recent advances in molecular biology and the isolation of different SOD
genes and SOD c-DNAs have been useful in. proving beyond doubt the physiological function of the enzyme. The
biosynthesis of SODs, in most biological systems, is under rigorous controls. In general, exposure to increased
pO2, increased intracellular fluxes of O2-, metal ions perturbation, and exposures to several environmental oxidants
have been shown to influence the rate of SOD synthesis in both prokaryotic and eukaryotic organisms. Recent
developments in the mechanism of regulation of the manganese-containing superoxide dismutase of Escherichia
coli will certainly open new research avenues to better understand the regulation of SODs in other organisms.

Keywords--pO2, O2-, Growth rate, Metal depletion/repletion, Anaerobic synthesis of MnSOD, Redox-sensitive
repressor, Autogenous regulation, Environmental stresses, Free radical

INTRODUCTION and .OH) generated during the normal biological re-


duction of dioxygen. SODs are unique in that their
Superoxide dismutases (SODs) (EC. 1.15.1.1) are me-
substrate is a free radical and that they catalyze the
talloenzymes that are widely distributed among oxy-
dismutation of O2- to H20: and 02 at a very fast rate,
gen-consuming organisms, aerotolerant anaerobes, and 2 x 109 M-Is -1.
some obligate anaerobes. They are essential for the
SODs, isolated from a wide range of organisms, fall
defense against oxygen toxicity, which is mediated by
into three types depending on the metal found in their
the partially reduced oxygen intermediates (O2-, HzO2,
active center. McCord and Fridovich ~were the first to
describe the superoxide-dismuting ability of a green
Paper number 11454 of the Journal Series of the North Carolina
Agricultural Research Service, Raleigh, NC 27695. The use of trade copper-containing protein that was isolated some 30
names in this publication does not imply endorsement by the North years earlier,: and thought to be a copper storage pro-
Carolina Agricultural Research Service of the products named, nor tein. This enzyme is now known to contain both copper
criticism of similar ones not mentioned.
Hosni M. Hassan was born in 1937 in Alexandria, Egypt. He and zinc (CuZnSOD) where the zinc plays a structural
received his B. Sc (Agriculture) in 1959 from the University of Ain- role. The other two types of SODs were soon discov-
Shams, Cairo; and his Ph.D. (Microbiology)in 1967, from the Uni- ered: one contains manganese (MnSOD)) and the sec-
versity of California at Davis (UCD). He was an assistant professor
of microbiology and biochemistry at Cairo High Polytechnical In- ond contains iron (FeSOD)? The distribution of these
stitute, then at the University of Alexandria. In 1972, he moved three forms of SODs is distinctly different, and may
back to North America. He was a visiting professor at the Macdonald provide an interesting evolutionary scheme. The
College of McGill University, Assistant Microbiologist at the Uni-
versity of Maine, and a Research Associate in Fridovich's laboratory CuZnSODs are typically found in the cytosol of eu-
at Duke University. Currently he is a Professor of Food Science, karyotes, while FeSODs are found in prokaryotes. On
Microbiologyand Toxicologyat N.C. State University. Dr. Hassan's
research interests are in the areas of Microbial Physiology and En-
the other hand, MnSODs are found in prokaryotes and
zymology. During the past 13 years, he has been interested in oxygen in mitochondria. There are, however, some exceptions
toxicity, biological roles of antioxidant enzymes, and the biosyn- to this simplified evolutionary scheme (For a recent
thesis and regulation of superoxide dismutases in microorganisms. complete account, see Bannister et al)). Amino acid
Dr. Hassan is an American Fulbright Senior Research-Scholar in
France. 1987-1988. He enjoys travel, photography, walking, and sequence data show that the three types of SODs fall
reading. into two distinct phylogenetic families, the CuZnSOD
377
378 H.M. HASSAN

and the Fe/MnSOD. There is no sequence homology regard to aerobic growth on minimal medium and tol-
or secondary structure similarity between the two fam- erance to paraquat. 35 The oxygen-dependent enhance-
ilies. 6 The FeSODs and MnSODs show a high degree ment of mutagenesis in the sod-negative mutants is
of amino acid sequence and structural homologies, while independent of RecA but dependent on the presence of
they are completely unrelated to CuZnSODs. 7.s Thus, exonuclease III.16 Oxygen is more mutagenic in sodA-
it is clear that these two families of SODs have evolved sodB and sodA mutants than in sodB mutant or in the
independently in response to a common selective pres- wild-type strain. ~6Conversely, the presence of a plas-
sure, that is, the evolution of oxygen and the threat of mid overproducing either FeSOD or MnSOD reduces
its toxicity. In this review I will concentrate on the the mutagenic frequency seen in the double mutant
biological function and the biosynthesis of SODs in (sodA-sodB) to that of the wild-type.
response to various environmental stimuli or stresses The recent advances in molecular biology and the
in an attempt to understand the regulatory mecha- isolation of SOD genes and SOD c-DNAs have facil-
nism(s) that govern their biosynthesis in different liv- itated more direct studies into the physiological func-
ing cells. tion of superoxide dismutases. 36 Thus, the human
copper-zinc superoxide dismutase (h-CuZnSOD) com-
plements SOD-deficiency in E. c0li.37 The double mu-
BIOLOGICAL FUNCTION
tant (sodA-sodB) is unable to grow aerobically in min-
All living cells are prone to oxygen toxicity, 9-~1 and imal medium and growth in rich medium is inhibited
mutagenicity. 12-16Several lines of evidence are avail- by the presence of paraquat or hydrogen peroxide,
able which support the superoxide theory of oxygen however, the expression of the human enzyme in this
toxicity and the protective role of superoxide dismu- mutant converts it back to the wild-type phenotype.
tases. 17.LsMcCord et al. 19 reported that aerobic organ- When the human-CuZnSOD gene is transfected into
isms contain more SOD than aerotolerant organisms, human Hela cells or mouse L-cells, transformants ex-
while anaerobes have no detectable SOD activity. Later pressing elevated levels of h-CuZnSOD are more re-
studies demonstrated the presence of SOD in some sistant to paraquat than normal cells/8 Similar results
obligate anaerobes, 2°-22 and a positive correlation is are obtained with cultured N IH/3T3 mammalian cells
found between the degree of oxygen tolerance and the transfected with a cDNA expressing high levels of the
level of SOD in anaerobic organisms. 23 The presence human-CuZnSOD. 39Furthermore, SOD is essential for
of low concentrations of SOD in many anaerobes may the activity of E. coli ribonucleotide reductase. ~°4~
represent an evolutionary response to ensure their sur- Recent studies have demonstrated the direct toxicity
vival during transient exposures to oxygen which.they of 02- without invoking the metal-catalyzed Haber-
encounter in route from one anaerobic niche to another. Weiss reaction. For example: (a) in E. coli, a,13-di-
The finding that some anaerobes are able to induce hydroxyisovalerate dehydrogenase, an enzyme essen-
SOD when exposed to very low concentrations of ox- tial for the biosynthesis of branch-chain amino acids,
ygen, 24 lends further support to the biological role of is extremely sensitive to increasing intracellular fluxes
SOD as an antioxidant enzyme. of 02- caused by the presence of redox-active com-
Increased levels of MnSOD in E. coli, induced by pounds in the growth medium; 42 (b) bovine liver cat-
a variety of physiological conditions, impart resistance alase is inactivated by O2- and SOD protects; 43 and
against oxygen toxicity, 25-33 and against the mutagen- (c) exposure of a purified glutathione peroxidase to an
icity of oxyradicals. 15.34The physiological role of SODs enzymatic source of O2- plus H202 causes inactivation
in protecting E. coli against the toxicity and mutagen- of the enzyme which is preventable by SOD, but not
icity of oxyradicals has been further explored in mu- by catalase. .4
tants lacking MnSOD (sodA), FeSOD (sodB), or These findings clearly support the conclusion that
both.16'35 The double mutant (sodA-sodB) is unable to superoxide radicals are toxic or may yield toxic prod-
grow aerobically in minimal medium, but grows nor- ucts, and that superoxide dismutases provide, in vivo,
mally in absence of oxygen. In rich medium, the sodB cellular protection by virtue of their ability to catalyt-
mutant exhibits the same sensitivity towards paraquat, ically dismute O2-. 17,18,45
an oxyradical generator, as the wild-type strain, whereas
the soda mutant is more sensitive to paraquat than the
BIOSYNTHESIS AND REGULATION
sodB mutant or the wild-type. In minimal medium,
however, both soda and sodB mutants are more sen- Exposure of living cells to physiological extremes
sitive to paraquat than the wild-type.3~ The introduction is known to evoke several global cellular responses
of a plasmid overproducing Mn- or FeSOD into the designed to provide cellular protection and to insure
double mutant restores its wild type phenotype with the survival of the species. The finding that superoxide
Control of SOD biosynthesis 379

dismutases are important in normal aerobic survival teriaceae family and all possess a constitutive FeSOD
aroused the interest of many biologists, biochemists, and an inducible MnSOD. 52 It is clear that the mech-
and geneticists, to attempt to understand the regulation anisms for biosynthesis and regulation of SODs in pro-
of SODs in different organisms. The findings clearly karyotes are not universal. Therefore, in the following
demonstrate that the biosynthesis of superoxide dis- sections I will concentrate on the biosynthesis and reg-
mutases, in most biological systems, are under rigorous ulation of MnSOD with special reference to E. coli.
controls.
Synthesis of SOD in response to changes in specific
growth rate and metabolism. Growth of E. coli in a
Prokaryotes
glucose-limited chemostat culture maintained under
Prokaryotes exhibit the broadest range of diversity constant and abundant aeration demonstrated that the
with regard to their oxygen requirement and tolerance, rate of MnSOD biosynthesis is proportional to the spe-
which makes them an ideal model system for studying cific rate of growth and the rate of respiration. 2s These
the regulation of SODs biosynthesis. Prokaryotes are results indicate that the inducer for MnSOD cannot be
also unique in their ability to grow fast and to readjust molecular oxygen, per se, but rather a unique product
their cellular composition to meet the fast and sudden of its metabolism. These results also advanced the hy-
changes in their environment. Cellular modulation, ac- pothesis that the concentration of MnSOD in the cells
complished via induction, repression, derepression, is modulated to correlate with the intracellular flux of
a c t i v a t i o n . . , etc., is the key for achieving fast ad- O2- generated during aerobic growth. It was also noted
aptation and the best cellular economy. Indeed, mod- that when E. coli is grown in rich medium containing
ulation of SOD biosynthesis in a given organism is limited amounts of glucose (i.e., trypticase-soy/yeast
finely-tuned to protect the cells against oxidative extract, TSY) the cells' content of MnSOD remains
damages. low while utilizing glucose but increases after glucose
is exhausted. 26 In general, if E. coli is utilizing a fer-
Synthesis of SOD in response to changes in p02. Aer- mentable carbon source, such as glucose, the MnSOD
obically grown E. coli contains three distinct isozymes level is much lower than when an oxidizable carbon
of superoxide dismutase,~ one of which contains man- source, such as succinate, lactate, or trypticase-pep-
ganese, 3 the second contains iron, 4 and the third is a tone, is utilized. 2~The lower MnSOD level seen when
hybrid consisting of one subunit from each of the other the cells are utilizing glucose is not due to catabolite
two isozymes and originally thought to contain only repression. ~ These results, led to the conclusion that
iron. 47 More recent biochemical48 and radiolabeling49 the rate of intracellular flux of 02- in E. coli is low
studies have clearly demonstrated that the hybrid en- during glucose fermentation, but increases during ox-
zyme contains both iron and manganese. Anaerobically idative metabolism and that the cells are capable of
grown E. coli, in contrast, contains only FeSOD. ~ modulating their content of MnSOD to meet their
Thus, the iron-enzyme is present under both anaerobic changing needs for the scavenging of 02-. This sup-
and aerobic conditions and is, therefore, constitutive. position led us to study the effects of redox-cycling
In contrast the Mn- and the hybrid-forms are absent compounds, capable of artificially increasing the in-
under anaerobic conditions but are rapidly synthesized tracellular flux of O2-, on the rate of MnSOD biosyn-
upon exposure to air. 46 It has been proposed that the thesis in E. coli. 26"27"56
presence of a constitutive FeSOD in E. coli provides
a back-up defense mechanism against sudden aerobic Synthesis of SOD in response to increased intracel-
exposures to oxygen) ° The anaerobe, Bacteroidesfra- lular/ extracellular flaxes of 02-. Several redox-cy-
gilis contains low levels of FeSOD when grown ana- cling compounds can enter E. coli, 27'56 and other
erobically, and exposure to an atmosphere containing bacterias2 and cause increase in the intracellular pro-
2% oxygen induces the enzyme. 24 Exposure to high duction of O2-. In general, these compounds are first
concentrations of oxygen have been shown to induce reduced by nonspecific diaphorases present in the cells;
MnSOD in E. coli, 5~ Streptococcus faecalis,51's2 Pro- the reduced forms are rapidly reoxidized by oxygen to
teus vulgaris, s2 Enterobacter cloacae,~2 Staphylococ- generate 02-.s6 The generation of 02- by these redox-
cus aureus, 52 Staphylococcus epidermidis, 52 Listeria active compounds is dependent upon their ability to
monocytogenes,S2 and Streptococcus sanguis,Sa as well enter the cells, the presence of oxygen, and on the
as FeSOD in Rhizobium japonicum, ~ Bdellovibrio presence of a metabolizable substrate that can provide
stolpii, 55 Alcaligenes faecalis, 52 and Pseudomonas a reducing equivalent. 28 Paraquat, pyocyanine, strep-
aeruginosa) 2 P. vulgaris, E. cloacae, Salmonella ty- tonigrin, juglone, plumbagin, menadione, methylene
phimurium, and E. coli all belong to the Enterobac- blue, and azure C are among the compounds which
380 H.M. I-IASSAN

function in this way. 56 They all act catalytically to 2,2'-dipyridyl, 1,10-phenanthroline, 8-OH-quinoline.
increase the intracellular flux of O2- and in doing so and ferrocyphen, cause a 3- to 7-fold increase in
they increase the rate of cyanide-insensitive respira- M n S O D ) 8-6° The iron chelator, 2,2'-dipyridyl, also
tion, 27"56since they provide a cytochrome-independent induces MnSOD in P. vulgaris, E. cloacae, S. aureus.
pathway to oxygen. These compounds exhibit an ox- and S. typhimurium: 2 Most fascinating, however, is
ygen-dependent toxicity, and in a real sense they ex- the finding that ferrous iron chelators can also induce
acerbate the toxicity of o x y g e n : 7 The cells respond to MnSOD in E. coli in the absence of dioxygen. 6° A
such threat by inducing MnSOD. Kinetic analysis of model has been proposed which accommodates all of
MnSOD induction by paraquat, showed that the ad- the above findings. 6° In this model, the synthesis of
dition of 0.5 mM paraquat to aerobically growing cul- MnSOD is envisioned as being regulated by a negative-
ture results, after a brief lag, in a rapid linear increase control operon, where the repressor protein (RP) con-
in MnSOD. 27 Removal of paraquat from the growth tains iron (Fig. 1). It is proposed that the active re-
medium causes a sharp decline in the rate of MnSOD pressor contains ferrous iron (RP-Fe-'+), while the
biosynthesis. The induction of MnSOD by paraquat is inactive form contains either ferric iron (RP-Fe 3+) or
prevented by inhibitors of transcription or translation, no iron (RP). Accordingly, in the absence of oxygen
but not by inhibitors of replication. 56 Paraquat also the active repressor will be predominant and no MnSOD
induces MnSOD in P. vulgaris, 52 E. cloacae: 2 S. au- is made. On the other hand, conditions known to ox-
reus, 52 and S. typhimurium. ~5Similar inductive effects idize Fe 2+ to Fe 3+ (i.e., oxygen or oxyradicals) or to
are seen with all the redox-active compounds listed remove Fe 2+ from the cells (i.e., iron chelators) will
above, s6 On the other hand, extracellularly generated generate the inactive forms of the repressor and will
0 2 - , via the action of xanthine/xanthine oxidase or result in the synthesis of the enzyme, even in the ab-
riboflavin and light, does not induce MnSOD in E. sence of oxygen. This model also suggests that con-
coli, due to the fact the O2- does not cross the E. coli ditions known to change the redox status of the cell
cell envelope. 3t These results indicate that the induc- may also affect the synthesis of MnSOD in absence of
tion of MnSOD in E. coli is under transcriptional con- oxygen. Indeed, this is the case. 49'6t Thus, under an-
trol and that intracellular 02- or a product thereof is aerobic conditions, active MnSOD is induced in the
the effector. presence of potassium ferricyanide, copper-cyanide
complex, ammonium persulfate, and hydrogen per-
Synthesis of SOD in response to metal depletion~re- oxide, 6~ as well as in the presence of nitrate plus para-
pletion. The role of iron in the regulation of MnSOD quat. 49 Western blot analysis revealed that the induc-
biosynthesis in E. coli was accidentally discovered dur- tion of MnSOD by these oxidants is associated with
ing a study on the effects of inhibitors of DNA bio- de novo protein biosynthesis. 61 Furthermore, the pres-
synthesis on the induction of MnSOD by paraquat. A ence of increasing concentrations of ozone, a powerful
2-fold increase in MnSOD is noted when nalidixic acid oxidant, induces MnSOD in E. coli. 62 These results
is used, instead of hydroxyurea, as an inhibitor of DNA suggest that the redox state of the cell influences the
biosynthesis) 8 Nalidixic acid has a synergistic effect biosynthesis and/or the activity of the proposed iron-
on the induction of MnSOD by paraquat (Hassan, un-
published data). It has been shown that the induction
I to I I PI~ o I
of MnSOD by nalidixic acid is not due to its known SG
ability to inhibit DNA gyrase nor to increase the in-
tracellular flux of O2-, but rather due to its ability to jl
chelate ferrous i r o n ) 8 Further studies have also shown / MnSOD
that the addition of excess iron to the growth media of RP" RP-Fe 3+ I
's
E. coli has a repressive effect on the biosynthesis of
MnSOD under normal (aerobic) or inducing conditions Fe2+ O~ ,,/
(i.e., in presence of 0.2 mM paraquat)) 9'6° Addition -o~,,'
of manganese to cultures already derepressed for MnSOD
biosynthesis causes a further increase in the amount of ll~.Fe 2'"
active enzyme, 6° probably due to a post-translational
Fig. 1. Schematicmodel for the regulation of MnSOD by oxygen,
activation of the Mn-apoenzyme. 49 Removal of metals superoxideradicals, and ferrousiron chelators. RG, regulatorygene;
from the growth medium by Chelex-lO0 also de- P, promotor; O, operator; SG, structural gene for MnSOD; RP.
represses the synthesis of MnSOD, but repletion of aporepressorprotein (inactive);RP-Fe3*, ferric repressor (inactive);
RP-Fe2*, ferrous repressor (active). (From Moody, C. S.; Hassan,
the medium with iron abolishes this e f f e c t : ° Further- H. M., J. Biol. Chem 259:12821-12825; 1984. Reprinted by per-
more, chelators specific for ferrous iron, that is, mission.)
Control of SOD biosynthesis 381

containing repressor that serves to negatively regulate Fe2+ for the active sites in the apo-proteins. Thus,
MnSOD biosynthesis. The utilization of reduction po- under anaerobic conditions the apo-MnSOD is sup-
tential in the regulation of MnSOD biosynthesis in E. posed to be made but the active site is occupied by
coli seems logical when viewed in the following light. iron and is therefore catalytically inactive. Aerobically
Facultative anaerobes take advantage of both aero- and in presence of O2--generators, Mn2+ is supposed
biosis and anaerobiosis. Exposure to oxygen, however, to be oxidized to Mn 3+ which in-turn is a strong com-
poses problems arising from oxygen toxicity. E. coil petitor for the active site of apo-MnSOD and therefore
overcomes oxygen toxicity by inducing antioxidant en- generates catalytically active MnSOD. 65 This model
zymes, MnSOD in particular. The reduction potential also proposes autogenous repression by the apo-pro-
is a very sensitive but complex indicator of the inte- teins and by the inactive enzymes that contain the in-
grated physiological state of the cell. Thus, very small correct metals.~'65 Fortunately, both regulatory models
changes in oxygen concentration can result in large are testable.
changes in reduction potential. If E. coil is pro- Recent studies, 49 have demonstrated that 59Fe binds
grammed to respond to changes in reduction potential to all the dismutase isozymes of E. coli, whereas 54Mn
instead of oxygen concentrations, then the cells could is more specific for the manganese and the hybrid iso-
induce the antioxidant enzymes at very low pO2 and zymes. In the presence of paraquat, an Oe- generator,
thus be poised and ready to combat the toxic effects more 59Fe is incorporated into MnSOD, whereas no
of oxygen when confronted with a higher pO:. 6~ 54Mn is incorporated into FeSOD. 49These data are the
Another model has been proposed to account for the opposite to what is predicted by the autoregulatory
effects of iron, manganese and chelators on the bio- model.65 Furthermore, the inactive form of the MnSOD
synthesis of both Mn- and Fe-SODs in E. c o l l . 63-65 In (i.e., the one containing 59Fe) appears to have no au-
this model (Fig. 2), both FeSOD and MnSOD are viewed toregulatory function, since its abundant presence does
as autogenously regulated catalysts whose conversion not prevent the induction of MnSOD by paraquat. 49
from inactive apo-proteins to catalytically active holo- Preliminary evidence was also presented for the pres-
enzymes depends on the availability of the appropriate ence of two unique iron-proteins49that may qualify for
metal cofactor and upon a prerequisit unknown con- the repressor function proposed in the transcriptional
centration of O:- capable of converting sufficient amount repression model. 6° Furthermore, the presence of a
of Mn2÷ to Mn 3+ in order to compete effectively with multicopy plasmid carrying the MnSOD gene (SodA)
was shown to cause the anaerobic expression of the
Aulooenous gene, presumably by neutralizing the limited number
of repressor molecules found in the cells. 49'67The most
convincing arguments in support of the negatively con-
trolled operon, 6° come from the following findings: (a)
western blot analysis of cell-free extracts prepared from
Fe-m-opo (inactive) anaerobically grown cells shows the absence of MnSOD-
antigen;35"6~ (b) the DNA sequence of sodA shows a
AMINO ACIDS region of dyad symmetry at the - 35 region which may
serve as a binding site for a repressor molecule;~ (c)
~~. ~.~FMn-f-OlDO(Inactive) 13-galactosidase is induced by paraquat in a strain of
E. coil carrying sodA-lacZ fusion;67 (d) MnSOD is
induced anaerobically by IPTG in ptac-sodA operon
fusion strains;68 and (e) MnSOD is induced by oxygen
Ao;.,,.72," F. lA,,,,.i strains overproducing FeSOD. 69
These results clearly indicate: (a) the absence of
active or inactive MnSOD in anaerobically grown cells;
Mn(I~),O~ . 2 H ° " M*llllloH20- / (b) the gene for MnSOD possesses a potential binding
Fe(lll).O~ . • Fe{ll)*O 2 site for a regulatory protein; (c) the induction of the
soda gene by paraquat is controlled at the transcrip-
Fig. 2. Effects of metals on the biosynthesis of SODs--a proposal. tional level and manganese ions play no regulatory
Abbreviations: m-apo, apoprotein of MnSOD; f-apo, apoprotein of
FeSOD. The valences of the metals are not shown. It is assumed role; (d) under anaerobic conditions, manganese ions
that Fe(lI) binds to both m-apo mad f-apo more avidly than Mn(lI), of the proper valence are present in high concentration
but not more avidly than Mn(III). The reactions illustrated at the to allow the synthesis of about 60 U of active MnSOD
bottom indicate why O:- increases the availability of Mn(III). (From
Pugh, S. Y.; Fridovich, I. J. Bacteriol. 162:196-202; 1985. Re- per mg of protein, as seen in the ptac-sodA operon
printed by permission.) fusion strains; and (e) superoxide radicals are not di-
382 H.M. HASSAN

rectly involved in the regulation of MnSOD. Further- to compare their ligand fields with the more fastidious
more, MnSOD regulatory mutants have recently been Fe/MnSODs.
isolated (Hassan and Touati, manuscript in prepara-
tion) and found to express the soda gene anaerobically Synthesis of SOD in response to other environmental
and to be uninducible by paraquat or chelators. In toto, stresses. Global cellular responses designed to provide
the data support the proposed modeP ° for a negatively protection against several environmental stress factors
controlled operon where the repressor molecule is now have been described. 77-79For example, the S O S , 77 heat
envisioned as an allosteric redox sensing protein. 61 shock, 78 and oxidative s t r e s s 79 responses are induced
However, the recent work by TouatP s suggests the by DNA damaging agents, exposure to high temper-
presence of a positive transcriptional control by O2-, atures, and exposure to hydrogen peroxide; and are
and an autogenous control that does not require the controlled by the recA, htpR, and oxyR gene products,
whole apoprotein, besides the originally proposed 6° respectively, Oxyradicals have been shown to cause
negative transcriptional control by an iron-repressor. DNA damage; s° and to induce the SOS response, 81
Indeed, the availability of regulatory mutants will al- endonuclease IV, s2 and a new type of DNA repair
low a better understanding of MnSOD regulation. pathway. 83 The possibility that the synthesis of
MnSOD is under the control of these global stress
Effect of metal substitution on the activity and synthesis regulons has been tested by several investigators. Thus,
of SODs. Metal substitution studies are valuable tools the regulation of the MnSOD in E. coli has been shown
in understanding the structure/function of SODs and to be independent of the inducible DNA repair (SOS)
the catalytic role of the native metal. Removal of the system. 84On the other hand, oxyR mutants were shown
metal from SODs results in loss of activity. Attempts to contain more MnSOD, 79 and exposure of E. coli
at metal-replacements with FeSODs and MnSODs from culture to 48°C induced MnSOD upon return of the
different bacterial species have demonstrated strict metal culture to 37°C. 8~ However, recent studies on the ef-
cofactor specificity. 7° Earlier studies with Fe/Mn- fects of paraquat on the synthesis of MnSOD (Lee and
SODs, from E. coli, have demonstrated that substi- Hassan, and Bowen and Hassan, manuscripts in prep-
tution of iron for manganese, or vice versa, results in aration) and on the synthesis of 13-galactosidase using
inactive enzymes. 71,7z Thus, it is clear that the two an sodA-lacZ fusion, 69 in mutants deficient in heat
enzymes, in spite of their relatedness, 6'8 must have shock response (htpR) or in oxidative stress response
diverged in a way that only the native metals can restore (oxyR), have clearly demonstrated that the regulation
their catalytic activity. of MnSOD is independent of these two regulons. The
The story is different, however, for the FeSODs possibility of a superoxide inducible (soi) regulon is
from anaerobic organisms, where the protein can be being examined by Kogema et al. 86
reconstituted or modulated during biosynthesis to yield
an active enzyme containing manganese in place of Genetic studies. The genes for MnSOD (sodA) and
iron. 73-76 Thus, the FeSOD from B. fragilis can be FeSOD (sodB) of E. coli have been cloned, 87'88 and
reconstituted as a Mn-containing enzyme o73 Further- sequenced. 66"89The DNA sequence for sodA indicates
more, anaerobically grown B. fragilis contains FeSOD, that the gene constitutes a single operon having two
but oxygen-stressed cells apparently use the same apo- possible promoters, but only one of them appears active
protein to make an active MnSOD. 74 Also, Propioni- under normal aerobic conditions. 66 The function of the
bacterium shermanii, 75 and Streptococcus mutans, 76 second promoter is not known at the present time, but
are able to synthesize either FeSOD or MnSOD, using its involvement under highly inducing conditions is a
the same apoprotein, depending on the metal supplied viable possibility. As discussed above, the sequence
in their growth media. The evolutionary significance also shows the presence of an almost perfect 19-base
of this, cambialistic, 76 class of enzymes is not clear. palindrome at the - 3 5 region which represents a po-
However, it is interesting to note that they are from tential binding site for a regulatory molecule. 66 The
cells that do not use oxygen as an electron-acceptor, linearized sodA is a better substrate for in vitro tran-
and only possess the FeSOD gene. Accordingly, one scription than the supercoiled f o r m , 66 this may be re-
may propose that organisms capable of using oxygen lated, in part, to our earlier observation on the effect
as a terminal electron-acceptor or possess MnSOD gene of nalidixic acid on MnSOD biosynthesis. 58 On the
or both Fe/MnSOD genes may not be able to exchange other hand, the DNA sequence of the sodB gene shows
their metal-cofactors and remain active. This possi- the lack of a regulatory element, 89 which is in agree-
bility needs to be tested. Also, complete amino acid ment with the fact that FeSOD, in E. coli, is a con-
sequences and X-ray crystallographic data for these stitutive enzyme. 46 For a complete review the reader
"cambialistic" enzymes are needed in order to be able is referred to the chapter by Touati. 36
Control of SOD biosynthesis 383

Eukaryotes REFERENCES

The biosynthesis of SODs in eukaryotes in response 1. McCord, J. M.; Fridovich, I. Superoxide dismutase: an en-
zymatic function for erythrocuprein (bemocuprein). J. Biol.
to increased oxygen concentrations, the presence of Chem. 244:6049-6055; 1969.
superoxide radical generators, and the availability of 2. Mann, T.; Keilin, D. Hvmocuprein and Hvpatocuprein, cop-
per-protein compounds of blood and liver in mammals. Proc.
metals is not much different from that reported in
R. Soc. London B. Biol. Sci. 126:303-315; 1938.
prokaryotes. 3. Keele, B. B.; McCord, J. M.; Fridovich, I. Superoxide dis-
S a c c h a r o m y c e s cerevisiae has been used as a model mutase from Escherichia coli B: A new manganese-containing
enzyme. J. Biol. Chem. 245:6176-6181; 1970.
system to study the biosynthesis of SODs in eukar-
4. Yost, F. J.; Fridovich, I. An iron-containing superoxide dis-
yotes. Under aerobic conditions, a cytochrome-c de- mutase from Escherichia call B. J. Biol. Chem. 248:4905-
ficient mutant is found to make twice as much SOD 4908; 1973.
as its isogenic wild-type strain. 9° The increased syn- 5. Bannister, J. V.; Bannister, W. H.; Rotilio, G. Aspects of the
structure, function, and applications of superoxide dismutase.
thesis of SOD in the mutant is due to increased intra- C R C CriticalRev. Biochem. 22:111-180; 1987.
cellular flux of O2- caused by the lack of a functional 6. Harris, J. I.; Auffret, A. D.; Northrop, F. D.; Walker, J. E.
electron transport chain, as depicted from the increased Structural comparisons of superoxide dismutase. Eur. J.
Biochem. 106:297-303; 1980.
rate of cyanide-insensitive respiration. 9° Addition of 7. Steinman, H. M.; Hill, R. L. Sequence homologies among
paraquat also causes an increase in the rate of cyanide- bacterialand mitochondrial superoxide dismutases. Proc. Natl.
insensitive respiration and induces the synthesis of Acad. Sci. USA 70:3725-3729; 1973.
8. Stallings,W. C.; Pauridge, K. A.; Strong, R. K.; Ludwig, M.
MnSOD and CuZnSOD; the addition of copper further L. Manganese and iron superoxide dismutase are structural
stimulates the synthesis of CuZnSOD. 9~ Similar find- homologs. J. Biol. Chem. 259:10695-10699; 1984.
ings were reported for S. cerevisiae growing in che- 9. Haugaard, N. Cellular mechanisms of oxygen toxicity.Phys-
iol. Rev. 48:311-373; 1968.
mostat cultures. 92 Feeding chicks a diet low in copper I0. Gottleib, S. E Effects of hyperbaric oxygen on microorga-
(<1 m g / k g dry matter) results in a significant decrease nisms. Annu. Rev. Microbiol. 25:I 11-152; 1971.
in the activity of erythrocytes' SOD compared to that I I. Fridovich. I. The biology of oxygen radicals.Science 201:875-
880; 1978.
of copper-supplemented (10 m g / k g dry matter) con- 12. Fvnn, W. O.; Gerschman, R.~ Gilbert, D. L.; Terwilliger, D.
trois.93 Subcutaneous injection of copper sulfate (5 rag/ E.; Gothran, F. V. Mutagvnic effects of high oxygen tensions
kg body weight) in rats causes an increase in the ac- on Escherichia coil Proc. Natl. Acad. 5ci. USA 43:1027-
1032; 1958.
tivity of CuZnSODs found in the cytosol and the mi- 13. Grifford, G. D. Mutation of an auxo~ophic strain of Esche~
tochondrial inter-membrane spaces. 94 The effect of richia coli by high pressure oxygen. Biochem. Biophys. Res.
copper on SOD biosynthesis was also observed in the Commun. 33:294-298; 1968.
14. Bruyninckx, W. J.; Mason, H. S.; Morse, S. A. Are physio-
fungus D a c t y l i u m dendroides, which contains both logical oxygen concentrations mutagenic? Nature (London)
CuZnSOD and MnSOD. 95 The fungus makes less 274:606-607; 1978.
CuZnSOD but more MnSOD, when grown in a copper- 15. Moody, C. S.; Hassan, H. M. Mutagenicity of oxygen free
radicals. Proc. Natl. Acad. Sci. USA 79:2855-2859; 1982.
deficient medium, so that the total SOD in the cell 16. Farr, S. B.; D'Ari, R.; Touati, D. Oxygen-dependent muta-
remains unaffected by the copper deficiency. genesis in Escherichia coil lacking superoxidedismutase. Proc.
The synthesis of SOD has been shown to increase Natl. Acad. Sci. USA 83:8268-8272; 1986.
17. Fridovich, I. Superoxide dismutases. Annu. Rev. Biochem.
in response to increased pO2 in yeast, 96 rat lung, 97,9s 44:147-159; 1975.
endothelial cells, 99 and in maize leaves. ~00The air po- 18. Fridovich. I. Superoxide dismutases. Adv. Enzymol. 58:62-
lutant SO2 was shown to induce SOD in the leaves of 97; 1986.
19. McCord, J. M.; Keele, B. B.; Fridovich, I. An enzyme-based
poplar tree, ~°~ but not in maize leaves. ~°° Exposure of theory of obligate anaerobiosis: the physiological function of
ten-day-old maize leaves to 10 -5 M paraquat for twelve superoxide dismutase. Proc. Natl. Acad. Sci. USA 68:1024-
hours results in 40% increase in total SOD activity. ~02 1027; 1971.
20. Hewitt, J.; Morris, J. G. Superoxide dismutase in some
More significant increases are seen in chloroplast and obligately anaerobic bacteria. FEBS Left. $0:315-318;
cytosolic SODs (CuZnSODs) than in the mitochondrial 1975.
MnSOD. The increases in cytosolic and mitochondrial 21. Carlsson, J.; Wrethen, J.; Beckman, G. Superoxide dismutase
in Bacteriodes fragilis and relatedBacteroides species.J. Clin.
SODs are the result of increased synthesis of these Microbiol. 6:280-284; 1977.
proteins. As in prokaryotes, heat-shock treatment does 22. Kirby, T. W.; Lancaster, J. R.; Fridovich, I. Isolation and
not increase the biosynthesis of SOD in maize.~°3 For characterization of the iron-containing superoxide dismutase
of Methanobacterium bryantii. Arch. Biochem. Biophys.
a complete review on the molecular biology of eukar- 210:140-148; 1981.
yotic superoxide dismutases see Touati. 36 23. Tally, F. P.; Goldin, B. R.; Jacobus, N. B.; Gorbach, S. L.
Superoxide dismutase in anaerobic bacteria of clinical signif-
icance. Infect. lmmun. 16:20-25; 1977.
Acknowledgements--This work was supported by DMB-8609239 24. Privalle, C. T.; Gregory, E. M. Superoxide dismutase and O:
from the National Science Foundation. H.M.H. is an American Ful- lethality in Bacteroides fragilis. J. Bacteriol. 138:139-145;
bright Senior Research-Scholar at the Institute of Jacques Monod, 1979.
University of Paris 7, Paris, France. 25. Hassan, H. M.; Fridovich, I. Physiological function of super-
384 H.M. HASSAN

oxide dismutase in glucose-limited chemostat cultures of Esch- containing superoxide dismutase from Escherichia coli. J. Biol.
erichia coli. J. Bacteriol. 130:805-811; 1977. Chem. 253:5220-5223: 1978.
26. Hassan, H. M.; Fridovich, I. Regulation of superoxide dis- 48. Clare, D. A.; Blum, J.; Fridovich, I. A hybrid superoxide
mutase synthesis in Escherichia coli: Glucose effect. J. Bac- dismutase containing both functional iron and manganese. J.
teriol. 132:505-510; 1977. Biol. Chem. 259:5932-5936; 1984.
27. Hassan, H. M.; Fridovich. I. Regulation of the synthesis of 49. Hassan, H. M.; Moody, C. S. Regulation of manganese-con-
superoxide dismutase in Escherichia coli: induction by methyl taining superoxide dismutase in Escherichia coli: anaerobic
viologen. J. Biol. Chem. 252:7667-7672; 1977. induction by nitrate. J. Biol. Chem. 262:17173-17177; 1987.
28. Hassan, H. M.; Fridovich. I. Superoxide radical and the ox- 50. Hassan, H. M. Superoxide and superoxide dismutases in Esch-
ygen enhancement of the toxicity of paraquat in Escherichia erichia coli: threat and defense. In: Bloch, K.; Bolis, L.; Toste-
coli. J. Biol. Chem. 253:8143-8148; 1978. son, D. C., eds. Membranes, molecules, toxins and cells.
29. Hassan, H. M.; Fridovich, I. Superoxide, hydrogen peroxide, Boston: John Wright, PSG Inc.; 1981:123-133.
and oxygen tolerance of oxygen-sensitive mutants of Esche- 51. Gregory, E. M.; Fridovich. I. Induction of superoxide dis-
richia coli. Rev. Infect. Dis. 1:357-369; 1979. mutase by molecular oxygen. J. Bacteriol. 114:543-548; 1973.
30. Hassan, H. M.; Fridovich, I. Superoxide dismutase andits role 52. Schiavone, J. R.; Hassan, H. M. Biosynthesis of superoxide
for survival in the presence of oxygen. In: Shilo, M., ed. dismutase in eight prokaryotes: effects of oxygen, paraquat
Strategy of microbial life in extreme environments. New York: and an iron chelator. FEMS Microbiol. Lett. 42:33-38; 1987.
Verlag Chemie; 1979:179-193. 53. DiGuiseppi, J.; Fridovich, I. Oxygen toxicity in Streptococcus
31. Hassan, H. M.; Fridovich, I. Paraquat and Escherichia coli: sanguis: the relative importance of superoxide and hydroxyl
mechanism of production of extracellular superoxide radical. radicals. J. Biol. Chem. 257:4046-4051; 1982.
J. Biol. Chem. 254:10846-10852; 1979. 54. Stowers, M. D.; Elkan, G. H. An inducible iron-containing
32. Hassan, H. M.; Fridovich, I. The mechanism of the antibiotic superoxide dismutase in Rhizobium japonicum. Can. J. Mi-
action of pyocyanine. J. Bacteriol. 141:156-163; 1980. crobiol. 27:1202-1208; 1981.
33. Hassan, H. M.; Fridovich, I. Exacerbation of oxygen toxicity 55. Von Stein, R. S.; Barber, L. E.; Hassan, H. M. Biosynthesis
by redox active compounds. In: King, T. E.; Mason, H. S.; of oxygen-detoxifying enzymes in Bdellovibrio stolpii. J. Bac-
Morrison, M., eds. Oxidases and related redox systems. Bal- teriol. 152:792-796; 1982.
timore, MD: University Park Press; 1982:151-167. 56. Hassan, H. M.; Fridovich, I. Intracellular production of su-
34. Hassan, H. M.; Moody, C. S. Superoxide dismutase protects peroxide radical and of hydrogen peroxide by redox active
against the toxicity and mutagenicity of oxygen free radicals compounds. Arch. Biochem. 196:385-395; 1979.
in Salmonella typhimurium. Can. J. Physiol. Pharmacol. 60:82- 57. Fridovich, I.; Hassan, H. M. Paraquat and the exacerbation
92; 1982. of oxygen toxicity. Trends in Biochem. Sci. 4:113-115; 1979.
35. Carlioz, A.; Touati, D. Isolation of superoxide dismutase mu- 58. Hassan, H. M.; Moody, C. S. Induction of the manganese-
tants in Escherichia coil: is superoxide dismutase necessary containing superoxide dismutase in Escherichia coli by nali-
for aerobic life? EMBO J. 5:623-630; 1986. dixic acid and by iron chelators. FEMS Microbiol. Lett. 25:233-
36. Touati, D. Molecular genetics of superoxide dismutases. Free 236; 1984.
Radic. Biol. & Med. 5:393-402. 59~ Moody, C. S.; Hassan, H. M. Induction of manganese super-
37. Natvig, D. O.; Imlay, K.; Touati, D.; Hallewell, R. Human oxide dismutase in Escherichia coli by iron chelators. Fed.
copper-zinc superoxide dismutase complements superoxide °Proc. 43:1759 abstr. (1984).
dismutase deficient E. coli mutants. J. Biol. Chem. 262:14697- 60. Moody, C. S.; Hassan, H. M. Anaerobic biosynthesis of the
14701; t987. manganese-containing superoxide dismutase in Escherichia coll.
38. Elroy-Stein, O.; Bernstein, Y.; Groner, Y. Overproduction of J. Biol. Chem. 259:12821-12825; 1984.
human Cu/Zn-superoxide dismutase in transfected cells: ex- 61. Schiavone, J. R.; Hassan, H. M. The role of redox in the
tenuation of paraquat-mediated cytotoxicity and enhancement regulation of manganese-containing superoxide dismutase bio-
of lipid peroxidation. EMBO J. 5:615-622; 1986. synthesis in Escherichia coli. J. Biol. Chem. 263 in press:
39. Krall, J.; Bagley, A. C.; Mullenbach, G. T.; Hallewell, R. 1988.
A.; Lynch, R. E. Superoxide mediates the toxicity of paraquat 62. Whiteside, C.; Hassan, H. M. Induction and inactivation of
for cultured mammalian cells. J. Biol. Chem. in press; 1988. catalase and superoxide dismutase of Escherichia coil by ozone.
40. Eliasson, R.; Jornvall, H.; Reichard, P. Superoxide dismutase Arch. Biochem. Biophys. 257:464-471; 1987.
participates in the enzymatic formation of the tyrosine radical 63. laugh, S. Y.; Fridovich, I. Induction of superoxide dismutases
of ribonucleotide reductase from Escherichia coli. Proc. Natl. and hydroperoxidases in Escherichia coli B BI2 (ATCC #29682)
Acad. Sci. USA 83:2373-2377; 1986. by manganese, iron and chelating agents. Fed. Proc. 43 3721
41. Fontecave, M.; Griislund, A.; Reichard, P. The function of abstr. (1984).
superoxide dismutase during the enzymatic formation of the 64. Pugh, S. Y.; DiGuiseppi, J. L.; Fridovich, I. Induction of
free radical of ribonucleotide reductase. J. Biol. Chem. superoxide dismutases in Escherichia coli by manganese and
262:12332-12336; 1987. iron. J. Bacteriol. 160:137-142; 1984.
42. Kuo, C. F.; Mashino, T.; Fridovich, I. et,13-Dihydroxyisoval- 65. laugh, S. Y.; Fridovich, I. Induction of superoxide dismutases
crate dehydrogenase: a superoxide-sensitive enzyme. J. BioL in Escherichia coli by metal chelators. J. Bacteriol. 162:196-
Chem. 262:4724-4727; 1987. 202; 1985.
43. Koao, Y.; Fridovich. I. Superoxide radical inhibits catalase. 66. Takeda, Y.; Avila, H. Structure and gene expression of the
J. Biol. Chem. 257:5751-5754; 1982. Escherichia coli Mn-superoxide dismutase gene. Nucleic Acid
44. Blum, J.; Fridovich, I. Inactivation of glutathione peroxidase Res. 14:4577-4589; 1986.
by superoxide radical. Arch. Biochem. Biophys. 240:500-508; 67. Touati, D.; Carlioz, A. Superoxide dismutase mutants of E.
1985. coli. In: Rotilio, G., ed. Superoxide and superoxide dismutase
45. Hassan, H. M.; Fridovich, I. Superoxide dismutases: detoxi- in chemistry, biology and medicine. North Holland: Elsevier
cation of a free radical. In: Jakoby, W. B., ed. Enzymatic basis Science Publisher; 1986:287-292.
ofdetoxication. Vol. 1. New York: Academic Press; 1980:311- 68. Touati, D. Transcriptional and post-transcriptional regulation
332. of MnSOD biosynthesis in E. coli: a study using operon and
46. Hassan, H. M.; Fridovich, I. Enzymatic defenses against the protein fusions. J. Bacteriol. in press; 1988.
toxicity of oxygen and of streptonigrin in Escherichia coll. J. 69. Nettleton, C. J.; Bull, C.; Baldwin, T. O.; Fee, J. A. Isolation
Bacteriol. 129:1574-1583; 1977. of the Escherichia coli iron superoxide dismutase gene: evi-
47. Dougherty, H. W.; Sandowski, S. J.; Baker, E. E. A new iron- dence that intracellular superoxide concentration does not reg-
Control of SOD biosynthesis 385

ulate oxygen-dependent synthesis of the manganese superox- of operon fusions (soi-lac Z) inducible by oxidative stress in
ide dismutase. Proc. Natl. Acad. Sci. USA 81:4970-4973; E. coll. ln: UCLA symposium mechanisms and consequences
! 984. of DNA damage processings. Abstr; 1988.
70. Kirby, T.; Blum, J.; Kahane, I.; Fridovich, 1. Distinguishing 87. Touati, D. Cloning and mapping of the manganese superoxide
between Mn-containing and Fe-containing superoxide dismu- dismutase gene (sad A) of Escherichia coil K-12. J. Bacteriol.
tase in crude extracts of cells. Arch. Biochem. Biophys. 201:55 i - 155:1078-1087; 1983.
555; 1980. 88. Sakamoto, H.; Touati, D. Cloning of the iron superoxide dis-
71. Ose, D. E.; Fridovich, I. Superoxide dismutase: reversible mutase gene (sod B) in Escherichia coil K-12. J. Bacteriol.
removal of manganese and its substitution by cobalt, nickel, 159:418-420; 1984.
or zinc. J. Biol. Chem. 2,51:1217-1218; 1976. 89. Carlioz, A.; Ludwig, M. L.; Stallings, W. C.; Fee, J. A.;
72. Ose, D. E.; Fridovich, I. Manganese-containing superoxide Steinman, H. M.; Touati, D. Iron superoxide dismutases: nu-
dismutase from Escherichia coil: reversible resolution and metal cleotide sequence of the gene from E. coli K- 12 and correlation
replacement. Arch. Biochem. Biophys. 194:360-364; 1979. with crystal structures. J. Biol. Chem. in press; 1988.
73. Gregory, E. M.; Dapper, C. H. Isolation of iron-containing 90. Lee, F. J.; Hassan, H. M. Biosynthesis of superoxide dismutase
superoxide dismutase from Bacteroidesfragilis: reconstitution and catalase in Saccharomyces cerevisiae: effects of oxygen
as a Mn-containing enzyme. Arch. Biochem. Biophys. 220:293- and cytochrome-e deficiency. J. Industr. Microbiol. 1:187-
300; 1983. 193; 1986.
74. Gregory, E. M. Characterization of the O2-induced man- 91. Lee, F. J.; Hassan, H. M. Biosynthesis of superoxide dismutase
ganese-containing superoxide dismutase from Bacteroidesfra- in Saccharomyces cerevisiae: effects of paraquat and copper.
gills. Arch. Biochem. Biophys. 23&83-89; 1985. J. Free Radicals Biol. Med. 1:319-325; 1986.
75. Meier, B.; Barra, D.; Bossa, E; Calabrese, L.; Rotilio, G. 92. Lee, F. J.; Hassan, H. M. Biosynthesis of superoxide dismutase
Synthesis of either Fe- or Mn-superoxide dismutase with an and catalase in chemostat culture of Saccharomyces cerevisiae.
apparently identical protein moiety by an anaerobic bacterium Appl. Microbiol. Biotech. 26:531-536; 1987.
dependent on the metal supplied. J. Biol. Chem. 257:13977- 93. Bettger, W. J.; Savage, J. E.; O'Dell, B. L. Effects of dietary
13980; 1982. copper and zinc on erythrocyte superoxide dismutase activity
76. Martin, M. E.; Byers, B. R.; Olson, M. O.J.; Salin, M. L.; in the chick. Nutr. Rep. int. 19:893-900; 1979.
Arceneaux, J. E.L.; Tolbert, C. A Streptococcus mutans su- 94. Ljutakova, S. G.; Russanov, E. M.; Liochev, S. I. Copper
peroxide dismutase that is active with either manganese or iron increases superoxide dismutase activity in rat liver. Arch.
as a cofactor. J. Biol. Chem. /,61:9361-9367; 1986. Biochem. Biophys. 235:636-643; 1984.
77. Little, J. W.; Mount, D. W. The SOS regulatory system of 95. Statzman, A. R.; Kosman, D. J. The utilization of copper and
Escherichia coll. Cell 29:11-22; 1982. its role in the biosynthesis of copper-containing proteins in the
78. Neidhardt, E C.; Varbogelen, R. A.; Vaughn, V. The genetics fungus, Dactylium dendroides. Biochim. Biophys. Acta 544:163-
and regulation of heat shock proteins. Ann. Rev. Genet. 18:295- 179; 1978.
329; 1984. 96. Gregory, E. M.; Goscin, S. A.; Fridovich, I. Superoxide and
79. Christman, M. F.; Morgan, R. W.; Jacobson, F. S.; Ames, B. oxygen toxicity in a eukaryote. J. Bacteriol. 117:456-460;
N. Positive control of a regulon for defenses against oxidative 1974.
stress and some heat shock proteins in Salmonella typhimu- 97. Crape, J. D.; Tierney, D. L. Superoxide dismutase and pul-
rium. Cell 41:753-762; 1985. monary oxygen toxicity. Am. J. Physiol. 2,2,6:1401-1407; 1974.
80. Brawn, K.; Fridovich, I. Strand scission by enzymically gen- 98. Stevens, J. B.; Autor, A. P. Induction of superoxide dismutase
erated oxygen radicals. Arch. Biochem. Biophys. 206:414- in neonatal rat lung. J. Biol. Chem. 2,52:3509-3514; 1977.
419; 1981. 99. Housset, B.; Junod, A. E Enzyme response of cultured en-
81. Brawn, M. K.; Fridovich, I. Increased superoxide radical pro- dothelial cells to hyperoxia. Bull. Eur. Physiopathol. Respir.
duction evokes inducible DNA repair in Escherichia coll. J. 17(suppl.):107-110; 1981.
Biol. Chem. 260:922-925; 1985. 100. Scandalios, J. G. The antioxidant enzyme genes Cat and Sod
82. Chan, E.; Weiss, B. Endonuclease IV of Escherichia coil is of maize: regulation, functional significance, and molecular
induced by paraquat. Proc. Natl. Acad. Sci. USA 84:3189- biology. In: Rattazzi, M. C.; Scandalais, J. G.; Whitt, G. S.,
3193; 1987. eds. isozymes: current topics in biological and medical re-
83. Farr, S. B.; Natvig, D. O.; Kogoma, T. Toxicity and muta- search. Vol. 14. New York: Alan R. Liss; 1987:19-44.
genicity of plumbagin and the induction of a possible new DNA 101. Tanaka, K.; Sugahara, K. Role of superoxide dismutase in
repair pathway in Escherichia coll. J. Bacteriol. 164:1309- defense against sulfur dioxide toxicity and an increase in su-
1316; 1985. peroxide dismutase activity with sulfur dioxide fumigation.
84. Hancock, L.; Hassan, H. M. Regulation of the manganese- Plant Cell Physiol. 21:601-612; 1980.
containing superoxide dismutase is independent of the indu- 102. Matters, G. L.; Scandalios, J. G. Effect of the free radical-
cible DNA repair system in Escherichia coll. J. Biol. Chem. generating herbicide paraquat on the expression of the super-
260:12954.-12956; 1985. oxide dismutase (Sod) genes in maize. Biochim. Biophys. Acta
85. Privalle, C. T.; Fridovich, I. Induction of superoxide dismutase 882:29-38; 1986.
in Escherichia coli by heat shock. Proc. Natl. Acad. Sci. USA 103. Matters, G. L.; Scandalios, J. G. Effect of elevated temper-
84:2723-2726; 1987. ature on catalase and superoxide dismutase during maize de-
86. Kogoma, T.; Farr, S. B.; Joyce, K.; Natvig, D. O. Isolation velopment. Differentiation 30:190-196; 1986.

View publication stats

Vous aimerez peut-être aussi