You are on page 1of 67

BAcmrzowoGIcAL Rzvows, June 1976, p. 403-468 Vol. 40, No.

2
Copyright C 1976 American Society for Microbiology Printed in U.S.A.

Degradation of Purines and Pyrimidines by Microorganisms


G. D. VOGELS* AND C. VAN DER DRIFT
Department of Microbiology, Faculty of Science, University of Nijmegen, Nijmegen, The Netherlands
INTRODUCTION ............................................................ 404
Purine Degradation in Animals .............................................. 404
Purine Metabolism in Plants ............................................... 405
Degradation of Purines and Pyrimidines by Microorganisms ........ ........... 405
AEROBIC DEGRADATION OF PURINES: ENZYMATIC STEPS .............. 406
Methylpurines ............................................................. 406

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Adenine .............................................................. 406
Xanthine Dehydrogenase .................................................... 407
Cofactors ............................................................. 408
Specificity .............................................................. 408
Other enzymes oxidizing purines ................... ........................ 409
Uricase ............................................................. 409
Properties .............................................................. 409
Specificity ............................................................. 411
Mechanism of action ...................................................... 411
Uricase-like processes ...................................................... 412
Allantoin ............................................................. 413
Allantoin racemase ........................................................ 413
Allantoinase ............................................................. 413
Allantoate Amidohydrolase and Allantoicase .................................. 414
Ureidoglycolase ............................................................. 415
Urea Degradation ........................................................... 416
Glyoxylate Degradation ..................................................... 416
AEROBIC DEGRADATION OF PURINES BY VARIOUS MICROORGANISMS . 416
Protozoa ............................................................ 416
Algae....................................................................... 417
Fungi ............................ ................................ 417
Basidiomycetes ........................................................... 417
Phycomycetes ............................................................ 417
Ascomycetes ............................................................ 417
Fungi imperfecti .......................................................... 419
Yeasts.................................................................... 419
Bacteria . ............................................................ 425
Cyanobacteria .......................................................... 420
Pseudomonas ............................................................. 420
Alcaligenes ............................................................ 421
Arthrobacter and Brevibacterium ............. ............................. 421
Bacius ............................................................. 421
Mycobacteria ............................................................ 422
Actinomycetales .......................................................... 422
Various bacteria .......................................................... 422
DEGRADATION BY ENTEROBACTERIACEAE AND STREPTOCOCCI ........ 423
Enterobacteriaceae .......................................................... 423
Streptococcus allantoicus . .................................................... 425
Catabolic Pathway of S. allantoicus and Enterobacteriaceae ....... ............ 425
Conversion of allantoin to ureidoglycolate ......... ......................... 426
Reductive degradation of glyoxylate ............ ............................ 426
Tartronate-semialdehyde reductase ............ ............................. 426
Oxidative production of oxalurate ............. ............................. 426
Oxamate formation ....................................................... 427
Concluding remarks ....................................................... 428
Purine Degradation by Other Streptococci ................ .................... 429
ANAEROBIC DEGRADATION OF PURINES ................................. 429
Clostridium acidiurici and C. cylindrosporum .............. ................... 429
Degradation
Degradation~
of purines......430
prn ..................................................43
~ ~to formiminoglycine ........
Conversion of xanthine ........................ 430
Utilization of formiminoglycine and glycine by whole cells ...... ............ 433
Conversion of formiminoglycine .............. ............................. 433
403
404 VOGELS AND VAN DER DRIFTBBACTERIOL. RHv.
Specificity of the THFA derivatives ........................................
435
Degradation of serine .....................................................436
Glycine synthase system ...................................................437
Formate dehydrogenase ................................................... 438
Reduction of C02. 438
Ferredoxin. 439
Quantitative aspects .......................................................
439
Regulatory aspects of the coenzymes .......................................440
Peptococcus aerogenes ...................................................... 440
Veillonella alcalescens ....................................................... 441
DEGRADATION OF PYRIMIDINES .......................................... 441
Reductive Pathway .......................................................... 442
Degradation of Orotic Acid .................................................. 444

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Pyrimidine Biosynthetic Pathway ............................................ 445
Oxidative Pathway ................................................ 446
Pyrimidine Degradation by Various Microorganisms .......................... 447
ECOLOGICAL ASPECTS ..................................................... 447
Organisms .................................................................. 447
Ecosystems ................................................................. 448
Mammal intestines ........................................................ 448
Avian caecum and insect intestines ......................................... 448
Skin ...................................................................... 449
Rumen ................................................................ 449
Symbiotic uric acid-degrading bacteria ..................................... 449
LITERATURE CITED ........................................................ 449
INTRODUCTION well as adult life. The waste product of an
Dedicated to H. A. Barker animal is ammonia, if ample water is available
for the rapid removal of this highly toxic sub-
In spite of the long history of studies on the stance. When water is not readily available,
biological conversions of purines and pyrimi- urea may serve as a water-conserving device
dines, only a few reviews on the microbial deg- and is temporarily retained as a less toxic ex-
radation of these compounds have been pub- cretory product. Such a situation occurs during
lished (147, 167, 207). Some of the degradation aestivation of the African lungfish, in which
products, i.e., uric acid, allantoin, and urea, urea accumulates to values as high as 0.5 g/100
were described in the eighteenth century (47, g of body weight in 1 year (160).
83, 457); in 1853 Wohler (598) first studied the Needham (362) generalizes that animals
degradation of allantoin by yeast, and a few whose eggs are protected against water loss
years later Stokvis (489) demonstrated the con- (cleidoic) tend to excrete relatively nontoxic
version of uric acid to allantoin in biological and insoluble compounds such as uric acid. A
materials. number of animals store uric acid, i.e., ascidi-
Purines and pyrimidines, the building stones ans, insects, terrestial gastropods, and land
of nucleic acids, are degraded in animals to crabs. Insects accumulate uric acid in special
waste nitrogenous substances; some of these organs, i.e., a fat body or rectal sac, which in
products appear to perform special functions in certain lepidopterans consists of 55% uric acid
plants. The compounds occur in large quanti- (414). The amount of uric acid formed depends
ties in nature and are subject to further degra- on the quantities of endogenous or exogenous
dation by various microorganisms. This review proteins metabolized.
will deal mainly with the degradation of pu- Variable amounts of white particulate mat-
rines which, as a result of their complexity, ter occur in the homocoel of the land crab Car-
offer a larger versatility of degradative routes disoma guanhumi. This material, mainly uric
and have been the subject of a larger number of acid, accounts for 0.2 to 15.9% of the total
investigations than have the pyrimidines. weight of a number of crabs and appears to
increase during intermolt. In one instance 55%
Purine Degradation in Animals of the nonshell solids was uric acid (187).
The products formed from purines by various Uricase is absent in humans, anthropoid
animals are given in Table 1. The phylogenetic apes, and several South American monkeys
significance of the predominant nitrogenous (109, 358). The enzyme of Old World Monkeys
waste products of animals, namely, uric acid, has been found to be highly unstable (109).
urea, and ammonia, was first discussed by However, small amounts of uric acid produced
Needham (361). It is related to the water econ- in humans may be degraded in three possible
omy of the animal during both embryonic as ways: by peroxidases, which have been re-
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 405
TABLE 1. Products ofpurine degradation in animals
Product excreted or stored Animals Reference
Guanine and/or xanthine Molluscs (e.g., Octopus, gastropods) 145, 238, 386, 387
Uric acid Primates 109, 358
Dalmation dog 212, 275, 293
Birds
Reptiles . 128, 342, 426
Molluscs 145, 387
Ascidians 191
Some terrestial crustaceans 187
Various insects 268, 389, 414

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Allantoin Other mammals
Various insects 51, 268, 389, 414
Allantoic acid Various insects 414
Urea Some aquatic reptiles 426
Terrestial amphibians 23
Teleost fishes 190
Dipnoans (during aestivation) 75, 190
Elasmobranchii 75, 510
Ammonia Aquatic amphibians 23
Crustaceans 474
Dipnoans 190
Echinoids 309
Some insects 268

(203), or by the action of intestinal flora (184, ence of allantoicase could be established only in
319, 320, 501). very few higher plants (479, 514), whereas He-
The feeding of yeast or other single cells to paticae showed a highly efficient hydrolysis of
humans can increase urinary uric acid excre- allantoic acid (418).
tion, so that in individuals with a genetic tend- In a number of plants, allantoin and allantoic
ency to primary overproduction of uric acid acid play an important role in the storage and
there may be precipitation of uric acid crystals translocation of nitrogen. In the bleeding sap of
in joints (gout), in soft tissues (tophi), or in the maple, allantoin and allantoic acid account for
formation of stones in the urinary tract. The as much as 70 to 100% of the total soluble
effect of feeding yeast (Torulopsis utilis) or nitrogen. In spring these compounds ascend
yeast ribonucleic acid on the serum levels of chiefly in the xylem, providing nitrogen for
uric acid and the excretion of uric acid was protein systhesis, and in the fall the reciprocal
tested by Edozien et al. (149) and Waslien et al. process takes place (418).
(581). The results indicate that an amount of 2 g The amount of allantoin present in various
of single-cell nucleic acid (equivalent to about Leguminosae is reported to be as high as 3.3 g/
30 g of food yeast) is probably a safe limit for kg of plant material (514).
most normal subjects, whereas 3 g of single-cell Tracey (514) concluded in 1955 that the im-
nucleic acid per day doubles the daily uric acid portance of allantoin and allantoate seems to
excretion (0.4 to 0.6 g) to an undesirably high have been insufficiently appreciated so far.
level (149). The excretion of nitrogenous waste Twenty years later we want to stress this state-
includes several billion kilograms of uric acid ment.
and allantoin per year which are recycled by
microorganisms. Degradation of Purines and Pyrimidines by
Microorganisms
Purine Metabolism in Plants Purines are degraded to the level of xanthine
Uric acid, allantoin, and allantoic acid are and uric acid along pathways that are not
present in a large number of plants (61, 418, strongly influenced by the presence of oxygen.
514). Allantoin and allantoinase, in particular, Microorganisms which use purines under aero-
are common components of plants, but the pres- bic conditions convert uric acid to allantoin,
406 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
ported by Canellakis et al. (97) to attack uric 0
acid in a manner qualitatively similar to that of 11 CH3
uricase, by the cytochrome oxidase system C
whereas under anaerobic conditions xanthine
(and perhaps other purines) is converted along O C2
I
C
7 CH
9
pathways, avoiding the involvement of uricase. N N
Allantoin can be degraded both under aerobic
and anaerobic conditions. Pyrimidines are de- CH3
graded along pathways that involve either a Caffeine
oxidative or a reductive step.
1,3,7- Trimethylxanthine)
AEROBIC DEGRADATION OF PURINES: H20

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


ENZYMATIC STEPS
Methylpurines Theophylline and Theobromine
Caffeine and related methylated xanthines (1,3 -dimethylxanthine) (3.7-dimethylxanthine)
are widely distributed in nature and are espe- H20
cially produced in high concentration in the
tissues of a number of well-known beverage 1 -Methylxanthine - l-Methyluric acid
plants. Caffeine has been reported to bring 3- Methylxanthine
about an immediate, although reversible, inhi- * 3-Methyluric acid
bition of both the synthesis of ribonucleic acid 7- Methylxanthine
and of protein in sensitive bacteria (392), and it H20
acts as a potent mutagen by inhibition of repair
processes (204, 563). Xanthine * Uric acid
Recent studies of Woolfolk (603) have demon-
strated that some strains of Pseudomonas pu- CH30H - bHCHO - HCOOH CO2
tida and Pseudomonas fluorescens are able to Methanol 2H Formaldehyde 2H Formic acid 2H
grow in media containing caffeine (1,3,7-tri-
methylxanthine) as the sole source of carbon FIG. 1. Degradation of caffeine by Pseudomonas
and nitrogen. P. putida will grow on any N- putida (603).
methyl derivative of xanthine containing one
or more methyl groups at the 1, 3, or 7 positions; inhibited under conditions of methanol accu-
the methyl groups are hydrolytically removed, mulation (603).
and methanol is formed (Fig. 1). It will not Caffeine, theobromine, and xanthine can be
grow on methanol as a sole source of carbon, used as the sole source of either carbon or nitro-
but glyoxylate that is formed by degradation of gen for growth by Penicillium roqueforti and a
uric acid is used in this way. Urea is not hy- species of Stemphylium that was isolated by
drolyzed. enrichment on caffeine-containing agar. A
Cells grown on any one of the N-methylpu- strain of Bacillus coagulans isolated from soil
rines given in Fig. 1 displayed activity toward utilizes caffeine in this way, too (292). A com-
all these compounds, but cells grown on xan- plete degradation of the substrates was ob-
thine were active only toward xanthine or uric served, but no intermediates were detected
acid. Cells grown on succinate and ammonia (292). Theophylline formation from caffeine
were not active with any of the compounds. was observed in studies with P. roqueforti
1-Methyluric acid is not degraded by caffeine- (468).
grown cells, but xanthine dehydrogenase from
the cells is active against xanthine (100%), 1- Adenine
methylxanthine (19%), and 3-methylxanthine Adenine deaminase (adenine aminohydro-
(41%) when tested in a ferricyanide-linked reac- lase, EC 3.5.4.2), which converts adenine to
tion (603). The specificity of this enzyme hypoxanthine and ammonia (Fig. 2), is com-
against methylxanthines will be discussed in a monly found among microorganisms but ap-
later section. pears to be absent in animals in which the
Methanol is converted to formaldehyde by aminohydrolase reaction involves mostly the
methanol dehydrogenase. This enzyme is inac- respective nucleoside and nucleotide.
tivated by its substrate. The demethylation re- Adenine deaminase exhibits no activity to-
actions, except for 7-methylxanthine, seem to ward adenosine (434, 436) and catalyzes a reac-
depend on the activity of methanol dehydrogen- tion that is virtually irreversible (229). The
ase and, as a consequence, degradation of caf- enzyme of Azotobacter vinelandii, Candida
feine and the other methylated compounds is utilis (211), and Schizosaccharomyces pombe
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 407
NH2 pass appears to be an alternative route to S.
N
pombe (1). Adenosine 5'-monophosphate
N C HN z
1I CH
.C

It CH (AMP), inosinic acid, and xanthylic acid are


HC "oC N/ H2N- C ,C N involved in the conversion of adenine to xan-
N H
Adenine Guanine thine by Candida guilliermondii (462, 463,
Adenine ,, H20 H20 Guanine
475).
deaminase NH3
NH3 deaminase Adenine, adenosine, and 2'-deoxyadenosine
exert an inhibiting effect on the growth of a
HN C H2,0 2H
CH
N
11 CH number of organisms. The inhibiting effect, ob-
HC .C N/ Xanthine =
C~ C served in studies with Aerobacter aerogenes,
N
H dehydrogenase N
H
Hypoxanthine
H
Xanthine
was reversed by thiamine or its pyrimidine
moiety 4-amino-5-hydroxymethyl 2-methylpy-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


a n Xanthene
2Nd dehydrogenase rimidine and also by histidine and succinate
Is H (72, 354). The inhibitory effect was explained as
HN C
11 C=O
feedback inhibition of purine nucleotide syn-
O=C NC oC--N thesis (354). Growth inhibition by adenine and
N
H H adenosine was reported for Escherichia coli
Uric acid
(228, 363, 422) and is not overcome by thiamine
02+ 2H20 '11 (228, 350, 422). Consequently, this inhibition
Uricase
CO2 + H202, must be caused in a manner different from that
H2N OC
-
NH Allanotoi NH,CO NH2
proposed for A. aerogenes. The bacteriostatic
I CO 40 OC I effect of adenine on E. coli appeared to be due to
OC C racemmase 0N0..C N"OCO inhibition of de novo synthesis of pyrimidine
HN
R 1-1-ALtantoin S l -Allantoin nucleotides. However, the mechanism of the
H20 j S()-Allantoinase inhibition remains obscure and was attributed
either to an inhibition of the reaction catalyzed
NH2 +NH3 eCO2 NH2 NHO by orotidine-5'-phosphate pyrophosphorylase
H20
CO COOH . CO COOH CO
(EC 2.4.2.10) or orotidine-5'-phosphate decar-
NH-CH-NH, 2° NH-CH - NH
boxylase (EC 4.1.1.23) or to an effect on the
Ureidoglycine H2( Allantoic acid
level of 5-phosphoryl-ribose-l-pyrophosphate
A -Allantoate H \A to,
amidohydrolase
A 0Z04
Allantoicase (228).
NH3 Inhibition was also reported for Agmenellum
COON COOH + NH2-CO-NH2 quadruplicatum (234), Corynebacterium sepe-
C
donicum (64), Staphylococcus aureus (134),
NHN-CO-NN -.OH HO I. NH-CO-NH, Pseudomonas acidovorans (263), and Myxococ-
H H
R- Ureidoglycolic acid S-Ureidoglycolic acid cus virescens (155). Adenine inhibits growth of
R-Ureidoglycolase\ S -Ureidoglycolase
Brevibacterium vitarumen var. uricum when
orAllantoicase \ urate served as the sole nitrogen source but not
COOH NH2-CO-NH Urease H20 when NH4+ salts were used as such (265).
OCH Urea Allophanate pathway
Glyoxyllc acid 2 NH,+CO2 Xanthine Dehydrogenase
FIG. 2. Pathways of purine degradation under Enzymes that catalyze the oxidation of hypo-
aerobic conditions. The various enzymatic steps are xanthine and xanthine have been obtained
discussed in the text. Organisms possessing allantoin from a wide variety of biological sources (166,
racemase are given in Table 3. The presence of RS-
allantoinase in bakers' yeast (307) is not accounted 291). Some are readily autoxidizable in oxygen,
for in the figure. The occurrence of allantoicase and whereas others autoxidize relatively slowly
allantoate amidohydrolase in various microorga- compared with their dehydrogenation by elec-
nisms is given in Table 4. R-ureidoglycolase is dem- tron acceptors, such as nicotinamide adenine
onstrated only in Pseudomonas acidovorans. dinucleotide (NAD), ferredoxin, or various
dyes.
(1) acts also upon 6-chloro-, 6-iodo-, and 6-hy- Oxygen is readily used as hydrogen acceptor
drazinopurines and replaces these groups by a by the mammalian species, among which is the
hydroxyl group. intensively studied milk xanthine oxidase
Some microorganisms which lack adenine de- (xanthine:oxygen oxidoreductase [EC 1.2.3.2]).
aminase are able to convert adenine into hypo- This enzyme was first described by Spitzer (484)
xanthine or xanthine by the use of bypasses. and Wiener (597) in 1899 and later by Schitten-
Adenosine and inosine are intermediates in the helm (461). The Schardinger (456) enzyme,
conversion of adenine to hypoxanthine by Sal- which was found in 1902 in milk and catalyzes
monella typhimurium (225), and the same by- the oxidation of formaldehyde in the presence of
408 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
methylene blue, is identical to it (347). The Purine
,,-' 427
xanthine dehydrogenases of mammalian liver
and milk can use also NAD as hydrogen accep- 2-OH 8-OH 6-OH (Hypoxanthine)
tor (37, 130). ;30,,,-' ~7 z~t
2,8-(OH)2 6,8 -(OH)2 2,6- (OH)2 (Xanthine)
Cofactors. Xanthine dehydrogenase of Veil- 30 1
lonella alcalescens (Micrococcus lactilyticus) 2,6,8-(OH)3 (Uric acid)
was purified 550-fold by Smith et al. (482) and
exhibits a molecular weight of about 250,000. It 2- OH, 7-Me 6-OH, 7-Me 6-OH, 1-Me
contains nonheme iron, sulfide, flavin, and mo- H6 j 17
lybdenum in a molar ratio of 8:8:2:>1.5. These 2,8- ( OH12 7- Mt 2,6- (08)2, 7-Me 6,B-(OH)2, 1-Me
values are quite similar to those found for milk
xanthine oxidase and chicken liver xanthine FIG. 3. Oxidation (downwards arrows) and re-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


dehydrogenase. The same cofactors are present duction (upwards arrows) reactions catalyzed by
in the 26-fold purified enzyme from Clostrid- xanthine dehydrogenase from Clostridium cylindros-
ium cylindrosporum. Molybdenum is also re- porum (66). The various purine derivatives are indi-
cated by use of symbols for the location of the hy-
quired in the activity of xanthine dehydrogen- droxyl (OH) and methyl (Me) groups at the purine
ase of P. aeruginosa (3, 344), and it has been skeleton. Solid arrows: knoWn reactions. Broken ar-
suggested that a molybdenum-containing cofac- rows: possible reactions. Numbers adjacent to the
tor is common to dissimilatory and assimilatory arrows give relative Vmax for the oxidation reactions.
nitrate reductase and xanthine dehydrogenase
for Aspergillus nidulans (19), Neurospora
crassa (359), and P. aeruginosa (554). xanthine, 6,8-dihydroxypurine, benzaldehyde,
The reaction of milk xanthine oxidase can be and acetaldehyde are oxidized at values less
described by the following formulation: than 13% (482). The numbers within parenthe-
ses refer to the relative velocities of the oxida-
E,), + X i EXXEred + U tions. The enzyme of P. aeruginosa is most
Er,,d + 02 4:± Ered 02 *°2Eox + H202 active with xanthine, 3-methylxanthine, hypo-
xanthine, 6-mercaptopurine, 2-thioxanthine,
where EOX and Ered represent oxidized and re- and 3-methyl-2-thioxanthine (50, 139). 2-Ami-
duced enzyme and X and U represent xanthine nopurine, its mono- and dimethylamino deriva-
and uric acid, respectively (373). The two half- tives, and 6-mercaptopurine are attacked at po-
reactions are physically separate steps. If flav- sition C2, in contrast to the milk enzyme which
ine adenine dinucleotide (FAD) is removed oxidizes at position C8 (48, 50). The enzyme of
from the enzyme, the enzyme exhibits no oxi- P. putida oxidizes both 1- and 3-methylxanthine
dase activity but is still reduced by xanthine to (603), but P. aeruginosa attacks 3-methylxan-
the same extent and at an even higher rate thine only (50). The enzyme of P. acidovorans
than that of the native xanthine oxidase. In oxidizes xanthine (100%), hypoxanthine (65%),
contrast to the native enzyme which exhibits and purine (4%) (478).
NADH2 oxidase activity, the deflavoenzyme Hypoxanthine can be converted to uric acid
can no longer react with NADH2, which do- via xanthine or 6,8-dihydroxypurine. The inter-
nates electrons via oxidized FAD. Xanthine mediary state of the latter compound was dem-
reacts directly with molybdenum, and electrons onstrated in the metabolism ofProteus rettgeri
are donated through this group to Fe/S centers and Serratia marcescens (150, 341) and of var-
and FAD (373). Xanthine is probably bound to a ious Streptomyces species (584). Adenine and
persulfide of the enzyme during the reaction guanine react only very slowly or not at all
(148), and the major rate-limiting step in the with the xanthine dehydrogenases.
oxidation of the substrate is the breakage of the Some of the compounds given in Fig. 3 have
persulfide bond (374). been shown to serve also as electron acceptors
Specificity. Various electron donors and ac- in the enzymic reaction. Reduction of uric acid
ceptors may be used by these enzymes. A num- by xanthine dehydrogenase constitutes the first
ber of the substrates that are oxidized by xan- step in the degradation of this compound under
thine dehydrogenase of C. cylindrosporum and anaerobic conditions, as will be discussed be-
the relative velocities of the oxidations are rep- low. The enzymes of C. cylindrosporum and V.
resented in Fig. 3. It is evident that the substit- alcalescens catalyze some reversible dismuta-
uents present in the molecule exert a directing tion reactions in which the same compound,
effect upon the point of oxidative attack. The e.g., xanthine or 6,8-dihydroxypurine, serves
enzyme of V. alcalescens oxidizes xanthine as electron donor and acceptor (66, 482). Cata-
(100%), hypoxanthine (94%), purine (87%), and lytic amounts of a viologen dye enhance this
salicylaldehyde (13%), whereas 9-methylhypo- dismutation reaction in C. cylindrosporum
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 409
(66). Smith et al. (482) tested the dismutation dehydrogenase; the other is very specific with
reaction with the purified enzyme of V. alcales- regard to the substrates oxidized and will oxi-
cens and observed that an equimolar mixture of dize only 2-oxypurine and closely related com-
hypoxanthine and uric acid is converted under pounds. It isnamed 2-oxypurine dehydrogenase
anaerobic conditions to an equilibrium mixture and was purified some 1,000-fold. 2-Oxypurine,
consisting of 64.0% xanthine, 17.3% uric acid, 2-oxy-8-azaxanthine, 2,8-dioxypurine, 2-oxy-
10.5% hypoxanthine, and 8.2% 6,8-dihydroxy- pyrimidine, and 2-oxypteridine are oxidized ex-
purine. The reduction of uric acid occurs also in clusively at position 6. A wide variety of artifi-
the presence of sodium hydrosulfite, 2-hydroxy- cial electron acceptors is used, but molecular
purine (66), or salicylaldehyde (482). oxygen is very poorly utilized. The enzyme con-
The xanthine-oxidizing enzymes utilize var- tains nonheme iron but not flavin or molybde-
ious nonphysiological compounds as electron num (605).

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


acceptors; ferricyanide, 2,6-dichlorophenolindo- A separate enzyme catalyzing the oxidation
phenol, methylene blue, and tetrazolium de- of 6,8-dihydroxypurine was proposed (584) to
rivatives are used by the enzymes of C. cylindro- occur in Streptomyces species on the basis of the
sporum and V. alcalescens (66, 482), but the sensitivity of this reaction to chloramphenicol
enzyme of P. acidovorans did not use the latter inhibition, but further studies are desirable to
dye (478). warrant this conclusion.
NAD is used by the avian enzymes (6) and
the enzyme of P. acidovorans (478). The latter Uricase
enzyme exposes a 10-fold lower activity with
NADP. NAD and NADP are not used by the In contrast to the foregoing enzyme, uricase
enzymes of C. cylindrosporum and V. alcales- (urate:oxygen oxidoreductase [EC 1.7.3.3]) ex-
cens (6, 66, 482). As a consequence, the enzyme hibits a stringent specificity regarding both the
of C. cylindrosporum exhibits little or no electron donor and acceptor. Only oxygen is
NADH2 oxidase activity (66). The natural ac- used in the latter way and, therefore, no orga-
ceptor in these anaerobic bacteria is ferredoxin nism is known to pass the uric acid barrier
(6, 67, 534). under anaerobic conditions.
The various enzymes react also with cyto- Uric acid is one of the oldest organic com-
chrome c and oxygen but generally at much pounds known. It was isolated from human
lower but apparently significant rates (66, 139, urine in 1776 by Scheele (457) and Bergman
478, 482). This rate is greatly enhanced by the (47). The structure of uric acid was elucidated
addition of ferredoxin to the enzyme of V. alca- in 1889 by Behrend and Roosen (43), and the
lescens (482). first studies on uricase were performed by
The enzymes differ considerably as to the Schittenhelm (461), Wiechowski (596), and Bat-
relative specificity to substrates as well as to telli and Stern (38) in the first decade of this
electron donors and electron acceptors. More- century.
over, the sensitivity to various inhibitors varies Microbial uricases have been recently ap-
strongly; e.g., the enzyme of V. alcalescens is plied in the treatment of children suffering
rather insensitive to cyanide, arsenite, and bo- from hyperuricemia (146) and patients with pri-
rate and very sensitive to methanol, in contrast mary gout (71, 269). Intramuscularly or intra-
to milk xanthine oxidase (482). However, the venously administered uricase of Aspergillus
similarities in the catalyzed reactions and en- flavus caused a rapid reduction in the serum
zyme composition warrant consideration of and urinary levels of uric acid; allantoin was
them all as a multifunctional xanthine dehy- excreted.
drogenase. Properties. Table 2 summarizes the proper-
Other enzymes oxidizing purines. Scazzoc- ties of purified uricases of various species.
chio et al. (125, 454, 455) presented immunologi- The uricases of animal origin (129, 218), the
cal and genetic evidence indicating the pres- protozoan uricases of Acanthamoeba terricola
ence of two distinct xanthine dehydrogenases in (357), Hartmanella culbertsoni (105), Polyto-
A. nidulans. The wild-type enzyme (XDH I) is mella caeca (185), Ochromonas malhamensis
induced by uric acid, and a constitutive enzyme (322), and presumably also of Chaos chaos and
(XDH II) is present in allopurinol-resistant mu- Amoeba proteus (356), the algal uricase of
tants. Both enzymes share one or more common Chlorogonium elongatum (485), and some
components. yeast uricases (378) were found to be associated
In Peptococcus aerogenes (Micrococcus aero- with the peroxisomes. Uricase of Alcaligenes
genes) two enzymes are present that catalyze eutrophus (Hydrogenomonas H16), Micrococ-
the oxidation of 2-oxypurine (605). One has the cus denitrificans, and P. aeruginosa appear to
broad substrate specificity typical of xanthine be firmly bound to structural components, since
410 VOGELS AND VAN DER DRIFT BACTERIOL. REzV.
TABLE 2. Properties of purified uricases of various sources
Sp act of Homoge- Metal content (at-
Source Times puri- purified ma-
neity of Molecular
terial (U/ purified mass
Isoelectric oms/mol of enzyme): Flavopro- pH opti-
fied mg of pro-
/-Tffi
tein mum
tein)a
Cu2+ | Fe3+

Swine liver 5,000 10.2 1


Rat liver 300 9.3 8.5-9.3
Ox kidney 30
Acanthamoeba 2.5
terricola
Neurospora 400
crassa

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Alternaria ten- 7
uis
Aspergillus fla- 8.5
vus
Candida utilis 130 10.5 <0.12 0.7-1.0 8.5
Arthrobacter 4,100 20.5 <0.2 <0.2 9.2
pascens
Micrococcus 5 20 8.5-8.8
varians
Alcaligenes eu- 8.5-9.0
trophus
Bacillus fasti- 3e 37e 9.0
diosus IIn b
-I- -i
Inhibition by Inhibition by:
Km (10-6
CN at: SH en
Source zyme
References
M) Cu2+ (1 Cu2+ (1 Fe3+
1 mM 1 zM PCMBb EDTA Hg2+ mm) AM)

Swine liver 20 + + 230,328,381,


382
Rat liver 513
Ox kidney 17.5 237, 515
Acanthamoeba + 357
terricola
Neurospora 201
crassa
Alternaria ten- 3,000 172
uis
Aspergillus fla- 60 (-) 295-297
vus +

Candida utilis 6 + 235, 438


Arthrobacter 200 +
d
18, 369
pascens
Micrococcus 267
varians
Alcaligenes eu- . 1,100 + 248, 250
trophus
Bacillus fasti- 250e 329
diosus
a
Micromoles of uric acid oxidized per minute per milligram of protein.
b
PCMB, p-chloromercuribenzoate.
c
Composed of four subunits with a molecular weight of 32,000.
d Stimulation by reducing substances.
eG. Bongaerts, unpublished data.

they sediment from ultrasonic preparations at throbacter (18, 369), and Fusarium
pascens
100,000 x g (250). Also, the enzyme of Histo- oxysporum (330) are
soluble.
plasma capsulatum is particle bound (316), but Uricase is commonly reported to be a metal-
the uricases of Bacillus fastidiosus (253), Ar- loenzyme containing either Cu2+ (swine liver)
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 411
or Fe3+ (C. utilis). However, the data are not seems to be the only known electron acceptor
conclusive. The enzyme of C. utilis was re- (172, 357, 381). Due to this stringent specificity,
ported to be a copper enzyme by Roush and uricase plays a role in the degradation of pu-
Shieh (438), but Itaya et al. (235) demonstrated rines by aerobic organisms only; anaerobic mi-
the presence of only trace amounts of copper croorganisms have evolved degradative routes
and 0.7 to 1.0 atoms of Fe3+ per mol of enzyme. of purines evading this reaction. H202 is formed
The presence of functional Cu2+ in the enzyme in stoichiometric amounts during the reaction
of swine liver is questioned by Truscoe and (357), and on this basis a quantitative determi-
Williams (515). The assumed presence of copper nation of uric acid can be performed (140, 161).
is partly based on the following observations: (i) Smaller amounts of H202 (70%) (172) or no H202
Urate oxidation to allantoin can be mediated (250) is formed in a number of bacteria or fungi
solely by Cu2+ or by Cu2+ plus H202 at pH 6.0 to due to the presence of catalase (250). On the

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


8.2 (459) or at higher pH values in the presence other hand, the accumulation of H202 may re-
of polyvalent anions or cyanide (40). Fe3+ was duce the amount of allantoin formed; catalase,
without effect (459). (ii) Cyanide exerts an in- when added to incubation mixtures lacking this
hibiting effect. The presence of functional enzyme, enhanced substantially the amount of
metal ions needs further investigation for all allantoin formed with the uricase of A. flavus
uricases. (297).
The effects exerted by p-chloromercuriben- The stoichiometric amount of allantoin is
zoate and heavy metal ions indicate that most formed only when uricase is tested in buffers
uricases contain an essential SH group and, as unlike borate (172, 583). In the presence of bo-
far as tested (180, 235), uricase appears to be rate, a stoichiometric amount of oxygen is con-
no flavoprotein. The pH optima of the enzymes sumed, but a large part (70%) of urate is not
are all located aroung pH 9, except for the converted to allantoin and CO2 but to equimo-
enzyme of Alternaria tenuis which has a pH lar amounts of alloxanic acid and urea (Fig. 4).
optimum at pH 7 (172) and that of a Strepto- In all studies performed with various uricases
myces for which a pH optimum in the range pH in borate buffer (pH, about 9), a fairly constant
6.0 to 8.5 was reported (583). The Km values of amount of allantoin (30%) was observed among
the enzymes differ strongly. However, Km val- the products of the reaction (96, 230, 297).
ues measured for uricase are only reliable when Mechanism of action. The mechanism of the
measured with freshly prepared solutions, uricase reaction has been studied extensively
since uric acid is readily oxidized in alkaline during the past 50 years, but no clear-cut pic-
solution to oxonate, a very potent inhibitor of ture is yet available. Since the uricases of A.
the enzyme. Fridovich (175) obtained for hog flavus (296), C. utilis (235), and A. pascens (18)
liver uricase a Ki value for oxonate and a Km are obtained in a homogeneous form, the uri-
value for urate of 1.1 x 10-7 M and 0.5 x 10-5 M, case action must be due to one enzyme. The
respectively. A very low Km value (3 x 10-8 to 6 physiological product formed by this enzyme is
x 10-8 M) was reported for liver uricase from allantoin, which occurs in nature in the opti-
freshwater teleosts (121). In various instances cally active S(+) form. Moreover, a number of
uricase exhibits substrate inhibition when natural allantoinases are specific for this opti-
tested at higher substrate concentrations; this cal isomer. Therefore, it is possible that uricase
inhibition may be due to the accumulation of an forms S(+)-allantoin (173).
intermediate (380). The uricase activity of A. The reaction mechanism must explain a one-
pascens is strongly influenced by the ionic enzyme reaction yielding S(+)-allantoin via a
strength of the test medium (18) in contrast to number of intermediates, as well as an oxida-
the enzyme of swine liver (39). Fitzpatrick et al. tive and a decarboxylative step. Figure 4 sum-
(158) compared the immunological properties of marizes the results obtained. Bentley and Neu-
uricase of B. fastidiosus, C. utilis, and Asper- berger (45) demonstrated in experiments with
gillus niger and observed a complete antigenic 1802 and H2180 that the oxygen of H202 origi-
independence. nated exclusively from gaseous oxygen. There-
Specificity. The physiological reaction cata- fore, the oxidation of uric acid consists in the
lyzed by uricase is shown in Fig. 2. Uricase transfer of two electrons or two H atoms from
exhibits a stringent specificity. A large number each uric acid molecule to molecular oxygen.
of purine bases and related compounds have No superoxide radical is produced during this
been examined; a number of them inhibit the reaction (381). The electron transfer would
reaction, but none appears to be a substrate, probably result in the formation of a carbonium
except perhaps 8-aminoxanthine (39, 327, 329), ion, the rearrangement of which leads to char-
which is oxidized at about 10-3-fold the rate of acteristic products. The primary intermediate
uric acid (327). On the other hand, oxygen formed (96, 327, 380, 391) is given as compound
412 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
0 active allantoin is most probably the product.
11 H Therefore, the reactions involved in the decay
HN1 NN
6 C 7N\5 of compound I to allantoin must proceed in an
2
11
I C=O
asymmetrical way, either due to the action of
O=C1%.3 ,,C , 9 /
N N uricase or due to chemical reactions in which
H H
Uric acid the asymmetry is conserved.
H+2e -P -H H202
Hydroxyacetylenediureine carboxylic acid
H 02'H H
(HDC) was isolated as a silver salt from a uri-
-,
N\ C
N... C N case system and as a product of the chemical
C=O - I C=O oxidation of uric acid by Schuler and Reindel
N
O C "C N/ (464). The participation of HDC in the degrada-
tion of uric acid needs further investigation,

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


H H
since the chemical conversion of HDC to allan-
toin appears to require relatively drastic condi-
H COOH H tions (464). If HDC is an intermediate in the
OC/N ,
N\C 0OH-or Ho N NH2 uricase system, then the cis form is the likely
O=C C=O -~ O=:
N .Cs / C=I candidate to conserve the asymmetry in the
\ NOC---"N/ T1/2 =18BO sec molecule (96).
H pHA8.! .5H H Canellakis and Cohen (96) studied the degra-
Compound I S -Atlantoin dation of uric acid-2-'4C and uric acid-8-'4C and
Xma,, 305nm Possible intermediates: proved that uric acid and a proposed intermedi-
pK, = 4.5
H COOHH
ate UIDC (5-ureido-2-imidazolidone-4,5-diol-4-
H2BO T1/2 =lO sec carboxylic acid), a hydrolytic product of HDC,
pH =8.5
O=C\ I C=O decompose asymmetrically to urea and allox-
H N-. -N
anic acid at pH 7.2; urea is derived from the C8
COOH
NH2 H HO H atom of uric acid and alloxanic acid is derived
/N-COH from the C2 atom of uric acid. This asymmetry
O=C 2 + IC0= HOC (cis)

\NH \C NH2 Hydroxyacetylenediureine is not maintained in incubation mixtures at pH


0 carboxylic acid 9.0, indicating that one of the intermediates
Atloxanic acid Urea H COOH
undergoes a secondary reaction.
N
-C/OH Moreover, Canellakis and Cohen demon-
O=C [O strated the formation of an unidentified inter-
N-\OH mediate in allantoin production under these
H NH-CO-NH2 conditions (0.1 M borate, pH 9.0). According to
UIOC these authors, the C2 atom of uric acid is
5-Ureido - 2-imidazolidone - equally distributed among the ureido group
4,5-diol-4-carboxylic acid and the hydantoin moiety of allantoin, and
Brown et al. (74) observed that the degradation
FIG. 4. Mechanism of uricase action (45, 96, 327, of 1,3-15N-labeled uric acid yielded in vivo al-
380, 391). The numbers in the molecules refer to the lantoin which is equally labeled in both the
origin of the atoms with respect to the uric acid ureido group and the hydantoin moiety. These
molecule (96, 124). T,12 refers to the half-life time of
the molecules (327). results hardly tally with the production of S(+)-
allantoin by uricase. However, the exchange of
the label may be due to the conditions used in
I in Fig. 4. The rate of further degradation of the hydroiodic acid degradation of allantoin.
this intermediate is independent of the amount Further studies are needed to reveal the ex-
of KCN-inhibited enzyme present in the incu- act mechanism ofthe uricase reaction, in which
bation mixture (380). Pitts and Priest (380) as- the asymmetry of the intermediates and the
sumed that the cyanide ion interacts with the products, allantoin and alloxanic acid, is ac-
oxygen site on the enzyme, thus preventing counted for.
only the binding of oxygen, and concluded that Uricase-like processes. Uric acid can be oxi-
the decay of compound I is independent of uri- dized to allantoin by a large number of oxidiz-
case. The final products of this decay can vary, ing reagents. In their study, Wohler and Liebig
depending upon the absence or presence of bo- (599) synthesized allantoin from uric acid by the
rate. Hydroxyl and borate ions act catalytically use of lead dioxide, whereas Schieper (460), Von
in these decays. Pitts and Priest assumed that Gorup-Besanez (574), Wheeler (586), and Claus
an unstable and symmetrical intermediate is (112) used potassium ferricyanide, ozone, man-
traversed prior to the formation of allantoin, ganese dioxide, and potassium permanganate,
but this view is hardly tenable since optically respectively.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 413
It is not unusual that besides uricase other to the reviews of Vitkz (561), Lubowe and Mecca
biological systems are able to oxidize uric acid. (318), and Greenbaum (200). The therapeuthic
Such a system was described by Canellakis et effects of plants on wounds and the maggot
al. (97) to explain some degradation of uric acid therapy have been attributed to the presence of
in humans in the absence of uricase. Uric acid allantoin in these plants and the excretory
is degraded to allantoin by lactoperoxidase- products of maggots.
H202, verdoperoxidase-H202, or horseradish Allantoin racemase. The optical antipodes of
peroxidase-H202 at a neutral pH in phosphate allantoin are subject to a rapid racemization,
buffer. In borate alloxanic acid, allantoin and especially at higher pH values. At pH 8.5 and
other products are formed. 30 C, about 3% of an isomer will racemize per
Other uricase-like processes are catalyzed by minute. The optical forms are rather stable at

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


the cytochrome-oxidase system (203) and by the pH values between 1 and 4, where the racemi-
pigments involved in the photosynthesis of cy- zation rate at 30 C amounts to about 0.1%/h
anobacteria and algae. The photosynthetic pig- (569).
ment system is highly bleached by uric acid, In a number of Pseudomonas species (Table
and allantoin or other oxidized products are 3), the presence of allantoinase is accompanied
formed (11, 53, 180, 541, 542). These reactions by allantoin racemase (EC 5.1.99.3), thus ena-
will be discussed in the section dealing with bling the organisms possessing an S(+)-allan-
cyanobacteria and algae. toinase to use both forms of allantoin.
The enzyme from P. testosteroni was purified
133-fold and shows optimal activity at pH 8.
Allantoin The reaction follows pseudo first-order kinetics
In the 8th year of the French Revolution, the at different initial substrate concentrations
citizens Buniva and Vauquelin (83) reported on since S- and R-allantoin are equally effective as
the isolation of a compound that they named the substrate. A number of other hydantoins
"acide amniotique." It was obtained as needle- are not racemized by the enzyme. In contrast to
shaped crystals by evaporation of the amniotic other racemases, allantoin racemase does not
fluid of cattle and extraction of the residue with require metal ions, reducing compounds, fla-
boiling alcohol. Buniva and Vauquelin as- vins, or pyridoxal-5'-phosphate for full activity
sumed it to be chemically related to uric acid, (553).
which was previously (in 1776), isolated from Allantoinase. Allantoinase (allantoin ami-
human urine by Scheele (457) and Bergman dohydrolase [EC 3.5.2.5]) is a very common en-
(47). zyme in nature. It occurs in many animals (168,
Gmelin (188) questioned in 1820 the acidic 303), higher plants (514), and microorganisms.
nature of this compound. In 1821 Lassaigne The first studies on the enzyme were made in
(304) distinguished between the allantoic and 1920 by Nemec (365), but Fosse and Brunel
amniotic fluids and, since the compound could (162) proved that allantoate is formed as the
be isolated from the allantoic fluid, Lassaigne product of the reaction.
changed the name to allantoic acid. Wohler and The enzymes from Streptococcus allantoicus,
Liebig (599) changed the name to allantoin and P. rettgeri, E. coli (570) and bakers' yeast (307)
described the synthesis of allantoin from uric convert both R(-)- and S(+)-allantoin to allan-
acid. In 1876 Grimaux (205, 206) established the toate. The aspecificity was not caused by the
chemical structure of allantoin and synthesized presence of allantoin racemase in the enzyme
it from simpler organic compounds, i.e., urea
and glyoxylic acid. Fosse et al. (163-165) iso-
lated the two optical isomers from allantoin. TABLE 3. Presence and absence of allantoin
Dextrorotatory allantoin was isolated from racemase in Pseudomonas (440, 453) a
leaves ofPlatanus orientalis and from the urine Enzyme present Enzyme absent
of cattle; levorotatory allantoin was obtained P. fluorescens biotypes P. fluorescens biotype F
after incubation of the racemic mixture with A through E
ground beans of Soja hispida. The absolute P. putida P. aeruginosa
configuration of the two optical antipodes of P. testosteroni P. stutzeri
allantoin was established by 's-Gravenmade et P. multivorans P. acidovorans
al. (196). The dextrorotatory and levorotatory P. mildenbergii P. alcaligenes
forms are S. and R-allantoin, according to the P. tolaasii P. pseudoalcaligenes
nomenclature of Cahn et al. (86, 87). The bene- P. maltophilia
ficial effect of allantoin in wound healing and a The
allantoinases of P. fluorescens, P. aerugi-
the maintenance of normal skin properties is nosa, and P. acidovorans are proven to be specific for
reported in a large number of papers. We refer S(+)-allantoin.
414 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
preparations (553, 568). The enzymes of animal using the name glyoxylate also for the hydrated
and plant sources and from P. acidovorans, P. form.
fluorescens (517, 568, 570), P. aeruginosa (196), Two enzymes are known to catalyze the deg-
and B. fastidiosus (62) are specific for S-allan- radation of allantoate, i.e., allantoicase (allan-
toin. toate amidinohydrolase [EC 3.5.3.4]) and allan-
Allantoinases differ strongly as to the influ- toate amidohydrolase (allantoate amidinohy-
ence exerted on them by Mn2+ ions and reduc- drolase [decarboxylating] [EC 3.5.3.9]). Allan-
ing agents. The enzymes from plants, S. allan- toicase was found independently by Krebs and
toicus, P. rettgeri, and E. coli are activated, but Weil (290) in frog liver and by Brunel (77) in the
the enzymes from the Pseudomonas species are mycelium ofA. niger (Sterigmatocystis nigra).
inhibited by Mn2+ ions. Cysteine and other re- Originally allantoicase was assumed to cata-
lyze the conversion of allantoate into 2 mol of

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


ducing agents stimulate the activity of the en-
zymes from S. allantoicus, P. rettgeri, and E. urea and 1 mol of glyoxylate, but Valentine et
coli but inhibit the plant and animal enzymes al. (530, 537) have shown that a separate en-
(571) as well as the enzyme from bakers' yeast zyme is involved in the degradation of ureido-
(307). Besides allantoin, methylolallantoin and glycolate. Probably, a two-step reaction is in-
5-aminohydantoin can serve as substrates. volved in all biological systems which convert
However, the latter compound is only degraded allantoate into 2 mol of urea and 1 mol of glyox-
appreciably by the enzymes of S. allantoicus ylate.
and P. rettgeri (565, 570). Allantoicases of P. aeruginosa, Penicillum
Franke and Taha (171) reported on an oxy- citreo-viride (518), and frog liver (520) form
gen-dependent degradation of allantoin by ex- S(-)-ureidoglycolate, which is probably the
tracts of Alternaria tenuis. They postulated the product of all allantoicase reactions. In contrast
presence of allantoin oxidase, which converts to the indication S, which refers to the absolute
allantoin into oxonic acid. However, these re- configuration, the notation (-) is almost mean-
sults were not substantiated by further evi- ingless in this instance. It refers to the sign of
dence. the optical rotation of a dilute, neutral, or alka-
line solution of S-ureidoglycolate measured at
wavelengths above 310 nm. If one of these con-
Allantoate Amidohydrolase and Allantoicase ditions is not fulfilled, the solution is dextroro-
Allantoate was first synthesized by Schieper tatory (195).
(460) in 1848. It is readily formed from allantoin Moreover, a second reaction appears to be
under strongly alkaline conditions (569). Allan- catalyzed by allantoicase. The 190-fold purified
toate, in turn, is subject to a series of reactions enzyme of P. aeruginosa and the enzyme of frog
which are given in Fig. 5. Equilibria are formed liver (520) are able to degrade R-ureidoglyco-
in each of the given reactions, which are exam- late, the optical antipode of the product formed
ples of known processes in which nucleophilic from allantoate, into glyoxylate and urea (519,
reagents, i.e., urea and water, act on the car- 562). Apparently, R-ureidoglycolate is bound to
bon atom of a C=O group or a C=N bond. the enzyme; its ureido group is cleaved off in a
Water and urea are bound to glyoxylate with similar way as the ureido group that is split
almost equal velocities, whereas urea is bound from allantoate. Both hydrolytic processes are
to allanturate about five times more rapidly reversible and may be represented by the reac-
than water (195). In aqueous solutions ureido- tion sequence (567):
glycolate (195) and the hydrated form of glyox- E +S*-ES ES + H2O*-EP + urea
ylate (294) are formed from allanturate and EP < E + P
glyoxylate, respectively. However, in this re-
view we will follow the common practice of in which E is allantoicase, S is allantoate or R-
NH2
NH2 NH2 CO NH2
CO ICOO CO NH2 NH2 CO
* ~* 1 0)
NH-CH-NH + COO CO * NH2 COO COO
+ I
CH=N + * HO- O=CH HO-CH-OH
+ * -,
H20 H20
Allantoate Allanturate Ureidoglycolate Glyoxylate Glyoxylate
hydrate
FIG. 5. Equilibria formed in the hydrolytic degradation of allantoate (195, 294, 572, 573).
VoL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 415
ureidoglycolate, and P is S-ureidoglycolate or icus at pH values between 5 and 8. The mecha-
the hydrated form of glyoxylate. The dissocia- nism of the activation and inactivation proc-
tion constants of ES and EP are about equal (25 esses can be explained by assuming an incor-
mM) for allantoate, ureidoglycolate, and the rect binding of Mn2+ ions to the enzyme at pH
hydrated form of glyoxylate, but urea is not values below 8. The activation processes re-
appreciably bound to the enzyme. The similar- move Mn2+ ions and, when the pH of the solu-
ity between the chemical (Fig. 5) and enzy- tion is brought to values above 8, the apoen-
matic hydrolysis is obvious, and ES and EP zyme binds Mn2+ ions in the correct way (551).
must be regarded as activated forms of allan- In our opinion the activation and inactivation
turate and glyoxylate. The equilibrium con- must reflect some physiological function of the
stant of the enzymatic hydrolysis of allantoate enzyme since it is conserved or has originated

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


(519) amounts to: independently in the evolution of bacteria as
different as E. coli, P. rettgeri, B. fastidiosus,
K [allantoate] S. allantoicus, and P. acidovorans.
[urea] [S-ureidoglycolate] In many instances it is not yet known
= 4.8 M-1 (pH 7.5, 30 0) whether either allantoate amidohydrolase or
Allantoicase ofP. aeruginosa is a metalloen- allantoicase is involved in the degradation of
zyme containing strongly bound Mn2+ ions. The allantoate. In urease-negative organisms, a dif-
apoenzyme is inactive, but the activity can be ferentiation can be made, since ammonia is
restored by various cations, although with dif- produced by the former enzyme and 1 mol of
fering effectiveness (552). The binding subsites urea per mol of allantoate is produced by the
of the enzyme (567) and its subunit structure latter enzyme. In urease-positive organisms,
(194) were studied. the enzyme must be separated from urease by
Allantoate amidohydrolase catalyzes the con- purification procedures, or activation by acid or
version of allantoate into 2 mol of ammonia, 1 complexing agents may be used as an indica-
mol of C02, and 1 mol of ureidoglycolate. A tion for the presence of allantoate amidohydro-
similar chemical hydrolysis of allantoate occurs lase. The known data on the distribution of
slowly in alkaline media (572). Ureidoglycine is both enzymes in biological materials are pre-
an intermediate in the enzymatic reaction: sented in Table 4.
The presence of allantoicase is not yet proven
allantoate -) NH3 + C02 + ureidoglycine unequivocally for Desmidiales (559), marine al-
ureidoglycine -* NH3 + ureidoglycolate gae (560), basidiomycetes (76, 77, 79), A. niger
(77), Aspergillus fumigatus (77), Aspergillus
The existence of a separate enzyme, ureidogly- oryzae (77), Penicillium chrysogenum (7), Geo-
cine aminohydrolase, was proposed (565, 566), trichum candidum (26), Fusarium monilo-
and Wu et al. (609) obtained evidence for it forme (9), Neurospora crassa (419), Saccharo-
from studies with mutants of P. acidovorans. myces cerevisiae (115, 307), and Histoplasma
However, no firm proof has been obtained, capsulatum (316). In spite of the lack of definite
partly due to the fact that ureidoglycine is not evidence for the presence of allantoicase, all
available as a substrate (545). Its accumulation allantoate-degrading enzymes will be called al-
during the enzymatic reaction is evident since lantoicases in this review, unless the presence
it reacts with glyoxylate in a nonenzymatic of allantoate amidohydrolase is proven.
transamination reaction in which glycine is Ureidoglycolase
formed (545, 566). Allantoate amidohydrolase
from S. allantoicus and B. fastidiosus (62) pro- The intermediary role of ureidoglycolate in
duces S(-)-ureidoglycolate from allantoate the catabolism of purines was first established
(545, 549), whereas R(+)-ureidoglycolate is by Valentine et al. (530, 537, 539). The com-
formed by the enzyme from P. acidovorans pound can be synthesized by the incubation of a
(517). concentrated solution of urea and glyoxylate
All allantoate amidohydrolases tested so far (517, 537) and hydrolyzes to these compounds in
have one unique property in common: the en- diluted solutions. These chemical reactions are
zyme is present in a partly inactive form in cell- catalyzed by bivalent cations and phosphate
free extracts. The activity can be increased con- ions (183, 195, 573).
siderably by pretreatment at pH values below 5 Besides allantoicase, which catalyzes the hy-
(517, 548, 550, 565, 566) or at pH values around drolysis of R(+)-ureidoglycolate, two other en-
6 in the presence of complexing anions (548, zymes are known to convert ureidoglycolate to
550). The enzyme exhibits optimal activity at glyoxylate and urea (Fig. 2). These ureidoglyco-
pH 8.5 and requires Mn2+ ions (544). However, lases (ureidoglycollate urea-lyase [EC 4.3.2.3])
Mn2+ ions inactivate the enzyme of S. allanto- differ as to the optical specifity. R-ureidoglyco-
416 VOGELS AND VAN DER DRIFT BACTERIOL. Rzv.
TABLE 4. Distribution of allantoicase and allantoate amidohydrolase in biological materials
Allantoate amidohydrolase present Reference Allantoicase present Reference
Bacillus fastidiosusa 62 Frog livera 520
Streptococcus allantoicusa 549, 566 Ochromonas malhamensis 321
Alcaligenes eutrophus 251, 252 Penicillium notatum 518
Escherichia coli 566 P. citreo-viridea 518
E. coli var. acidilactici 566 Aspergillus nidulans 454
Citrobacter freundii 566 Candida utilis 106
Proteus rettgeri 566 Brevibacterium linens 16
Pseudomonas acidovoransb 517, 609 Arthrobacter globiformis 289
Pseudomonas aeruginosaa 518
P. fluorescens 518

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


a S-ureidoglycolate is formed.
b R-ureidoglycolate is formed.

lase is present in P. acidovorans (517) and S- coo -


ureidoglycolase is present in P. aeruginosa, P. NH2 Mg 2+,K+lo
NH
fluorescens, P. citreo-viride, S. allantoicus, C=O + ATP +HCO- 4 C=O + ADP + P
B. fastidiosus, and frog liver (62, 518-520, 530, NH2 NH2
549, 568). The enzymes are slightly stimulated Urea Allophanate
by Mn2+ ions (183, 517, 565), and the enzyme of
S. allantoicus is strongly inhibited by p-chlo- U20 + H +
romercuribenzoate, Hg2+ ions, and Zn2+ ions 2HCO-+ 2NH4+
(183).
The enzymatic and nonenzymatic reactions tion. FIG. 6. Allophanate pathway of urea degrada-
led to the establishment of an equilibrium with
a constant (183, 519):
(urea:CO2 ligase [ADP] [EC 6.3.4.6]) and allo-
K [ureidoglycolate] phanate hydrolase (allophanate amidohydro-
=

[urea] [glyoxylate] lase [EC 3.5.1.13]). ATP is used by the first


= 7.4 M-1 (pH 7.5, 30 C) enzyme, to which biotin serves as a prosthetic
group. Biotin is not necessary for allophanate
The enzyme of S. allantoicus was purified 77- hydrolase (593). In contrast to the enzymes
fold by Gaudy et al. (182). from Chlorella vulgaris (508), the enzymes
from S. cerevisiae are components of a multien-
Urea Degradation zyme complex (592).
Urea formed in the degradation of purines is Glyoxylate Degradation
degraded in various organisms by urease (urea In most, if not all, instances glyoxylate is
amidohydrolase [EC 3.5.1.5]). Urease synthesis degraded in a reaction sequence involving tar-
is strongly repressed by ammonia or NH4+ ions tronate-semialdehyde synthase (glyoxylate car-
in various organisms, which in this way pre- boxy-lase [dimerizing] [EC 4.1.1.47]) and tar-
vent an unlimited rise of the alkalinity in the tronate-semial dehyde reductase [D-glycer-
medium (252, 287, 488, 556). Omura et al. re- ate:NAD (P)+ oxidoreductase (EC 1.1.1.60)].
ported on the presence of urea dehydrogenase The former enzyme was first described by Kra-
in Scenedesmus species and higher plants (375, kow and Barkulis (284) for E. coli and is a
376). The enzyme prefers NADP to NAD and flavoprotein (208), using thiamine pyrophos-
acts also with a-ketoglutaric acid oxime and y- phate and Mg2+ ions as cofactors. It was shown
glutamylhydroxamic acid as substrates. to be present in various other microorganisms
In some reports an adenosine 5'-triphosphate (236, 253, 281, 530, 565).
(ATP)-yielding hydrolysis of urea was consid-
ered (241, 538), but the only known hydrolytic AEROBIC DEGRADATION OF PURINES
alternative of urease is an ATP-requiring reac- BY VARIOUS MICROORGANISMS
tion, which was first described by Roon and Protozoa
Levenberg (430) and is operative in yeast (429,
431, 592, 593), algae (224, 308, 472, 508), and The scant information on the purine degrada-
some fungi (477). Two enzymes are involved in tion by protozoa is summarized in Table 5. Te-
the reaction sequence (Fig. 6): urea carboxylase trahymena pyriformis requires preformed pu-
VOL. 40, 19 76 DEGRADATION OF PURINES AND PYRIMIDINES 417
TABLz 5. Enzymes involved in purine degradation by protozoa
Enzyme" (reference)
Organism
Ade Gua XDH Uncaseb
Tetrahymena +(470) +(470) +(470, 557) +(470)
pyriformis -(151, 306) -(151, 221) -(221, 306) -(221, 306, 355, 557)
Chaos chaos + (356) -(356) + (356)
Amoeba proteus ± (356) -(356) + (356)
Paramecium aurelia -(483) -(483)
Acanthamoeba terricola +(357)

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Polytomella caeca +(185)
Hartmanella culbertsoni +(105)
Ochromonas malhamensis +(321, 322)
a Ade, Gua, and XDH refer to adenine deaminase, guanine deaminase, and xanthine dehydrogenase or
xanthine oxidase, respectively; + and - refer to the presence or absence of the enzyme, and ± refers to the
presence of weak enzymatic activity.
b Presumably in all instances the enzyme is localized in peroxisomes, when present.

rines for growth since it is unable to synthesize which were allantoin, urea, cyanuric acid, and
the purine nucleus (219, 220). In Ochromonas parabanic acid (11). Allantoin is not utilized as
malhamensis both allantoinase and allanto- a nitrogen source for growth, presumably due
icase are present, but the intermediary position to the inability to transport this compound,
of ureidoglycolate has still to be established since allantoin formed from purines is degraded
(321). Epidinium ecaudatum caudatum con- in the cell (10). Urea carboxylase of this orga-
verts guanine and adenine to xanthine and hy- nism is induced by urea and repressed by NH4+
poxanthine, respectively, and xanthine is de- ions, whereas allophanate hydrolase is a consti-
graded further along a pathway in which uric tutive enzyme (224). The latter enzymes are
acid, allantoin, and allantoate may participate also present in other Chlorophyceae (308).
(113). Villeret (559) was the first author who dem-
onstrated allantoinase and allantoicase activity
Algae in algae grown on mineral medium or peptone.
Reports on the degradation of purines by al- All 21 freshwater algae investigated contained
gae are restricted to studies on the use of these allantoinase, and only five species, all belong-
compounds as a nitrogen source for growth ing to the Desmidiales, degraded allantoate.
and on the occurrence of some of the enzymes Allantoinase is also present in 90% of 51 species
involved in degradation (360). Most of the 38 of marine algae (Chlorophyta, Phaeophyta,
chlamydomonad species tested by Cain (88) and Rhodophyta), but allantoicase activity was
could use adenine, whereas about half of them found in only about 20% of the strains (560).
used uric acid. Chlamydomonas reinhardi uses
both xanthine and uric acid (53) and contains Fungi
the urea carboxylase-allophanate hydrolase Basidiomycetes. Uricase, allantoinase, and
system, which is induced by urea and repressed allantoicase activities were demonstrated in a
by NH4+ ions (224, 472). large number of Basidiomycetes by Brunel (76,
Xanthine and uric acid serve as a nitrogen 77) and Brunel and Capelle (79).
source to Chlorella pyrenoidosa, Chlorella vul- Phycomycetes The results for the Phycomy-
garis (53), and Monodus subterraneus (337) but cetes are summarized in Table 6.
not to Porphyridium cruentum and Euglena Ascomycetes. Part of the data for the Asco-
gracilis (53). C. pyrenoidosa is able to use ade- mycetes is summarized in Table 7. Penicillium
nine as well as hypoxanthine and contains uri- chrysogenum utilizes guanosine, adenosine
case (10). Uric acid is degraded in a photochem- (153), adenine, hypoxanthine, and xanthine
ical process, when a solution ofthis substance is (332) as sole sources of nitrogen for growth. The
illuminated in the presence of chlorophyll a or b presence of adenine deaminase (8), a constitu-
extracted from this organism. At least seven tive xanthine dehydrogenase (495), uricase (7,
degradation products were observed, among 172) and allantoinase (7), and an inducible al-
418 VOGELS AND VAN DER DRIFTBBACTZRIOL. REV.
TABLE 6. Degradation of purines by Phycomycetes
Substrate used as N Enzymea
and C source
Organism Reference
Uric acid Man-
toin Ade XDH Uricase
Phytophtora infestans + - 283, 377
Mucor boidin 283
Mucor spinosus (M. plumbeus) + + 172
Mucor racemonsw + 495, 565
Rhizopuw nigricans + + 172, 495, 565
Cunninghamella elegans + 172

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


a Ade and XDH refer to adenine deaminase and xanthine dehydrogenase, respectively.
b Uric acid tested as sole nitrogen source.

TABLE 7. Growth of Ascomycetes on uric acid or allantoin and the formation of xanthine dehydrogenase
(XDH), uricase (Uri), allantoinase (An), allantoicase (Ac), and ureidoglycolase (Ug)
Substrate-b Enzyme present"
Organism
Uric acid Allantoin XDH Uri An Ac Ug
Penicillium species
P. brevicaule (Sco- CN (283)
pulariopsis brev-
icaulis)
P. chrysogenum N (7) (495) (7, 172) (7) (7)
P. citreo-viride CN (565) (565) (518) (518)
P. frequentans (P. N (172) (172)
globrum)
P. glaucum CN (172, 283) (495) (172)
P. notatum CN (565) (172) (565) (518) (518)
Aspergillus species
A. flavus (296)
A. fumigatus N (172) (495) (172)
A. nidulans N (126) N (126) (454) (126) (454) (454) (454)
A. niger CN (77, 172, 283) N (77) (495) (77, 172) (77) (77)
A. niveo-glaucus CN (283)
(A. glaucus)
A. oryzae N (77) (495) (172) (77) (77)
A. phoenicis N (77) N (77) (77) (77) (77)
Beavaria bassiana N (349)
(Botrytis bas-
siana)
Geotrichum candi- N (26) N (26) (26) (26) (26) (26)
dum
Gliocladium sp. (172)
Neurospora crassa N (419) N (367, 419) (201, 419) (367, 419)(419) (419)
N. sitophila (172)
Sporotrichum goug- N (349)
eroti
Trichophyta viola- N (349)
ceum (Achorion
violaceum)
a References are given within parentheses.
b N and CN refer to use of the substrates as sole nitrogen source or sole nitrogen and carbon source,
respectively.

lantoicase (7) were demonstrated. Penicillium for Aspergillus nidulans, which contains uri-
roqueforti can utilize methylpurines and xan- case (126), xanthine dehydrogenase, allanto-
thine as sole sources of either carbon or nitro- inase, allantoicase, and ureidoglycolase (454).
gen for growth (292). Two different xanthine dehydrogenases are re-
Hypoxanthine, xanthine, uric acid, allan- ported to be present in this organism (125, 454,
toin, and allantoate serve as nitrogen sources 455). One enzyme is constitutive; the other one
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 419
(as well as uricase) is induced by uric acid (454). onstrated in the cell-free extracts (9). Xanthine
Optimal induction ofallantoinase requires both dehydrogenase and uricase of Fusarium sam-
uric acid and allantoin. This phenomenon may bucinum are constitutive enzymes (172). Uri-
be significant in view of the reported toxicity of case is also present in Fusarium niveum, Fu-
allantoin to this organism (454). In the presence sarium semitietum (172), and Fusarium oxy-
of histidine, A. nidulans can no longer use sporum (330) but absent in Fusarium equiseti
hypoxanthine, uric acid, allantoin, and urea as (172).
nitrogen sources. This compound represses the The tobacco-wilt organism Fusarium oxyspo-
synthesis of urease and affects the activity of rum var. nicotianae can use uric acid and al-
both xanthine dehydrogenase and uricase. The lantoin as nitrogen sources (600). Uric acid
effects are not due to ammonia, which may be serves as both a nitrogen and carbon source for

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


produced from histidine, but reflect a preferen- Cladosporium herbanum and Paecilomyces
tial use of the latter substance (385). Aspergil- farinosus (283) and as a nitrogen source for
lus fumigatus contains adenine deaminase Microsporum lanosum, Trichophyton ferrugi-
(495). neum (490), Mastigocladium blochi, and Epi-
Geotrichum candidum uses xanthine, uric dermophyton inquinale (349). Uricase is pres-
acid, and allantoin as nitrogen sources (26). ent in Cladosporium, Trichoderma koningi,
Uricase was present in spores of this organism and Botrytis cinerea (172). The fungal pathogen
germinated in the presence of glutamine or Histoplasma capsulatum is capable of utilizing
urea, but its level increased ninefold when uric uric acid as well as its degradation products as
acid was present during germination (26). Urea nitrogen sources for growth. Uricase, allanto-
is degraded along the allophanate pathway, inase, allantoicase, and urease are inducible
which is not induced by urea (477). Allanto- (316).
inase and ureidoglycolase are constitutive en- Yeasts. The first experiment on the degrada-
zymes in this organism, whereas allantoicase is tion of purines and its catabolites by microorga-
induced by allantoin (26). nisms was performed by Wohler (598) who in
Neurospora crassa uses adenine, hypoxan- 1853 inoculated a solution of allantoin with an
thine (127), uric acid (419), allantoin (367, 419), unidentified yeast. Among the end products
and allantoate (420) as nitrogen sources. A com- formed after 4 days at 30 C were ammonia,
mon transport system is present for adenine, urea, oxalate, and carbonate.
guanine, and hypoxanthine and also a specific Growth of yeasts on purines is a widespread
one is present for adenine (326). Although Mc- property. Adenine, hypoxanthine, xanthine,
Elroy and Mitchell (332) could not observe ade- and guanine support the growth of 108, 82, 108,
nine deaminase in Neurospora, the presence of and 116 strains, respectively, out of a total of
this enzyme could be deduced from the growth 123 yeast strains belonging to a wide variety of
pattern of wild-type and mutant strains (127). genera (302). Candida utilis readily uses the
Uricase of N. crassa is induced by uric acid and purine bases as a nitrogen source, whereas Sac-
allantoinase is induced by either uric acid or charomyces cerevisiae utilizes adenine and
allantoin. Both enzymes are repressed by am- guanine moderately well but is not able to grow
monia. Ureidoglycolase and urease are consti- on hypoxanthine and xanthine (137, 302).
tutive enzymes (419). S. cerevisiae, C. utilis, and Schizosaccharo-
Fungi imperfecti. Adenine deaminase, xan- myces pombe degrade the purines along the
thine dehydrogenase, and uricase are demon- common pathways in which uric acid is an in-
strated in Alternaria tenuis (167, 172). Growth termediate and adenine deaminase, guanine
of this organism on xanthine as the nitrogen deaminase, xanthine dehydrogenase, and uri-
source is almost negligible (495). Uricase is re- case are involved as inducible enzymes (299,
ported to be constitutive in A. tenuis and Al- 388, 434-437). In S. pombe another route may
ternaria solani; in Alternaria tenuissime and be involved in the conversion of adenine to
Alternaria porri the basal level could be en- hypoxanthine where AMP, adenosine, and ino-
hanced by growth on uric acid (172). Uricase is sine are intermediates (1).
present in Alternaria gossypina and Stemphy- Xanthine dehydrogenase could not be demon-
lium (172). A species of Stemphylium is re- strated in cell-free extracts of C. utilis, presum-
ported to utilize caffeine, theobromine, and ably due to the lability of the enzyme (436). The
xanthine as sources of either carbon or nitrogen enzyme of Candida guilliermondii was re-
for growth (292). ported to be unable to oxidize hypoxanthine,
Fusarium moniliforme can grow on hypo- which is converted to xanthine via the interme-
xanthine, xanthine, uric acid, and allantoin as diates inosinic acid and xanthylic acid (462,
nitrogen sources. Xanthine dehydrogenase, ur- 463, 475).
icase, allantoinase, and allantoicase were dem- Uric acid, allantoin, and allantoate sup-
420 VOGELS AN4D VAN DER DRIFT BACTERIOL. REV.
ported the growth of 109, 113, and 118 species trogen source. The growth rate was much re-
out of 123 tested yeasts from various genera. duced and the photosynthetic pigment system
The ability of yeasts to use purines and their was highly bleached (542). It was concluded
degradation products is so widely distributed that besides uricase, also a nonenzymatic oxida-
that it is of no obvious value as a generic char- tive attack on uric acid occurs in cyanobacteria.
acteristic (302). Allantoin is degraded in a path- In A. nidulans, which could not grow on uric
way involving allantoinase, allantoicase, urei- acid (53), this substance was degraded almost
doglycolase, urea carboxylase, and allophanate completely to allantoin in the light. This pho-
hydrolase. Biotin is required for the function of tooxidation was mediated by a pigment not
urea carboxylase and, thus, its stimulating role identical to chlorophyll a or phycocyanin.
in the degradation of allantoin, allantoate, and Pseudomonas. Various members of the ge-
urea is obvious (136, 141, 348). The urea carbox- nus Pseudomonas are able to grow on purines

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


ylase-allophanate hydrolase system offers either as a nitrogen source or as a nitrogen and
ureaseless yeasts a way of using nitrogen from carbon source. Adenine, guanine, hypoxan-
urea and allantoin; its discovery resolved pre- thine, and xanthine serve as nitrogen sources
vious problems (114, 260). An inducible allanto- for P. aeruginosa (170, 433), P. acidovorans
inase is present in Saccharomyces carlsbergen- (263), and an unidentified pseudomonad (139),
sis (543). Allantoinase, allantoicase, and urei- and these purines are used also as sole carbon
doglycolase of S. cerevisiae were reported to be and nitrogen sources by P. acidovorans (478)
constitutive (106, 108, 307) or inducible (141). and other pseudomonads (20, 90). Adenine in-
Allophanate induces the allantoin degradative hibits the growth of some strains ofP. acidovor-
enzymes together with the multienzyme com- ans on hypoxanthine (263, 478), probably due to
plex involved in the allophanate pathway (115) inhibitory effects of adenine which were dis-
and the urea uptake system (116). The synthe- cussed in a previous section. Adenine deami-
sis of the complex is furthermore repressed by nase and guanine deaminase were demon-
NH4+ ions in S. cerevisiae (594), C. utilis (432), strated in P. aeruginosa (170), P. acidovorans
and C. guilliermondii (512). (478), and P. oleovorans (448). Xanthine dehy-
Allantoinase, allantoicase, and ureidoglyco- drogenase is present in P. aeruginosa (49, 111,
lase of C. utilis are inducible enzymes (106-108, 139, 170, 344) and P. acidovorans (478); the
307). C. guilliermondii contains allantoinase enzyme can use oxygen as an electron acceptor.
(462, 475), allantoicase (475), and the enzymes Uric acid is used as carbon and nitrogen
of the allophanate pathway. The latter path- sources by P. aeruginosa (250, 314, 433), P.
way is induced by the presence of both urea and putida (P. eisenbergii) (132, 314), and P. cal-
biotin (512). coacetica (314). P. fluorescens utilizes this com-
Studies on the transport mechanism of pu- pound as a nitrogen source (170, 433) and proba-
rines and pyrimidines in yeast revealed that S. bly also as carbon and nitrogen sources (132,
cerevisie possesses a common active transport 314). The presence of uricase was demonstrated
system for adenine, guanine, hypoxanthine, for P. aeruginosa (49, 111, 139, 169, 250, 433), P.
and cytosine (384, 416) and a specific one for fluorescens (433), and P. acidovorans (478). The
uracil (202) and urea (116). Purine uptake is activity of this enzyme was lost after sonifica-
activated by protons and inhibited by K+ ions, tion of the cells (49), probably due to the fact
and it was concluded that the purine transport that it is membrane bound (478).
system in S. cerevisiae acts as a proton sym- The enzymes involved in the degradation of
porter and a K+ antiporter (415). NH4+ ions purines are inducible enzymes (20, 89, 169, 170,
exert an inhibitory effect on uric acid transport 250, 433, 440, 478).
in C. utilis (436) and Pichia guilliermondii Allantoin appears to be an attractive sub-
(476), but the mechanism of the inhibition is strate to a large number of species. It is used as
unknown. carbon and nitrogen sources, by P. aeruginosa,
P. fluorescens (314, 570), P. putida (P. eisenber-
gii) (133, 314, 553), P. calcoacetica (314), P.
Bacteria acidovorans (570), P. mildenbergii, P. tolaasii,
Cyanobacteria. Anacystis nidulans and Sy- P. testosteroni, P. multivorans (440), and sev-
nechococcus cedrorum can not use xanthine eral unclassified strains (89, 132). It serves as a
and uric acid as a sole source of nitrogen (53), nitrogen source of P. maltophilia, P. alcali-
but Agmenellum quadruplicatum is able to uti- genes, P. pseudoalcaligenes, P. stutzeri (553),
lize adenine, hypoxanthine, xanthine, and uric P. iodinum, and P. oleovorans (440).
acid as a nitrogen source for growth (256). The various pathways involved in the degra-
Several species of marine cyanobacteria show dation of purines are given in the enzymatic
atypical growth when uric acid serves as a ni- section on the aerobic degradation. Preliminary
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 421
studies on the genetics of allantoin metabolism / NHCO C02 H c H20 /NH2
in P. aeruginosa were made by Gaudy and OC\ OH OC oCCO COOH
Bruce (181). NH H' \COO \NHN " CHOH
Part of the pathway (Fig. 2) may be involved COON NH-CHOH
in alloxanate degradation by a soil pseudo- Altoxanic acid 5-Hydroxyhydantoin Ureidoglycolic acid
monad and was studied by Gerhart et al. (186)
and Gray et al. (198, 199). This microbe grows FIG. 7. Degradation of alloxanic acid by a Pseu-
well aerobically in media containing alloxanic domonas species (186,198,199).
acid, allantoin, or uric acid as sole sources of
carbon, nitrogen, and energy. Alloxanate is skii and Popova (232, 233) who isolated the
converted to 5-hydroxyhydantoin, and the lat- organism (A. ureafaciens and A. pascens) from
ter yields glyoxylate and urea (Fig. 7). The peat soil, but Rouf and Lomprey (433) found

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


authors mentioned allanturic acid as a possible contrasting results. Uricase of A. globiformis
intermediate. However, it seems more likely (A. pascens) was studied by Arima and Nose
that 5-hydroxyhydantoin is hydrolyzed by hy- (18).
dantoinase into ureidoglycolate, which in turn Arthrobacter strains AC1 and AC207 are able
is degraded by ureidoglycolase. to utilize uric acid and allantoin as sole sources
Alcaligenes. Autotrophically grown A. eu- of carbon, nitrogen, and energy, but in contrast
trophus can use uric acid and allantoin either to A. globiformis 4 mol of ammonia is formed
as a sole nitrogen or sole carbon source for instead of urea (16). A. tumescens and A. sim-
growth when energy is available from the oxi- plex are uricase negative (16).
dation of H2 gas, which is required for the incor- Brevibacterium vitarumen var. uricum con-
poration of ammonia into cell material (12). tains uricase, whose induction was stimulated
Kaltwasser (248-252) demonstrated that this by FeSO4 (266), but Imshenetskii and'Popova
organism can use uric acid as a sole nitrogen (232, 233) reported on a strain that appears to
and carbon source. A particle-bound and induc- contain a constitutive uricase. B. linens de-
ible uricase is involved. The presence of allan- grades both xanthine and uric acid (481). The
toate amidohydrolase is obvious since 1 mol of latter substance is used as a nitrogen source
urea and 2 mol of ammonia are formed from 1 (481) but not as a nitrogen and carbon source
mol of uric acid (251, 252) in spite of the ability (16). Washed cells convert 1 mol of uric acid or
of the organism to produce urease. This enzyme allantoin to 1 mol of urea (16).
is fully repressed by ammonia during growth Bacillus. Various reactions of the purine
with uric acid or allantoin (252). Glyoxylate is degradative pathway can be performed by a
degraded along the tartronic semialdehyde number of Bacillus species, but only a few spe-
pathway to glyceric acid and phosphoenolpyru- cies degrade these substances fully. Adenine is
vate (251). The enzymes of this pathway are converted to hypoxanthine by Bacillus anthra-
induced when cells grow on uric acid, allantoin, cis (345) and B. subtilis (131). The latter orga-
or glyoxylate (251, 252). nism degrades also xanthine and uric acid
Various heterotrophic strains of Hydrogeno- (139), which is used as the sole source of nitro-
monas utilize adenine, guanine, xanthine, hy- gen, carbon, and energy (433). This property is
poxanthine, uric acid, allantoin, and urea for an. adaptive one (433).
growth (249). Alcaiigenes faecalis can use uric B. polymyxa utilizes xanthine and guanine
acid as a source of nitrogen (169, 170, 516), but as nitrogen sources (132). Moreover, uric acid
Rouf and Lomprey' (433) reported the contrary. and allantoin are used in such a way by this
This organism contains uricase (612). Other organism and by B. megaterium, B. guano, B.
species of Alcaligenes can use uric acid as a hollandicus, and B. subtilis var. niger (B. vul-
secondary carbon and energy source (458). gatus) (132, 486). However, the results are not
Arthrobacter and Brevibacterium. Krebs unequivocally confirmed (139, 169, 170, 516,
and Eggleston (289) demonstrated that Arthro- 613, 614). Guanine, xanthine, uric acid, and
bacter globiformis (A. ureafaciens) (110) can allantoin are not used as nitrogen sources by B.
grow in media containing hypoxanthine, uric cereus var. mycoides (132), and B. stearother-
acid, or allantoin as the sole organic substrate. mophilus cannot use allantoin in this way
From uric acid 2 mol of urea is formed, and (433). An unidentified species ofBacillus grows
allantoin and allantoate are intermediates in at the expense of adenine, guanine, hypoxan-
the degradation. The results suggest the in- thine, xanthine, uric acid, or allantoin as nitro-
volvement of allantoinase and allantoicase, gen sources. Growth on these compounds as
which are both induced by growth on urate. sole organic substrates is slight. The growth on
The growth of A. globiformis on uric acid as uric acid is an adaptive property (433).
the sole substrate was confirmed by Imshenet- B. fastidiosus was first isolated in 1929 by
422 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
Den Dooren de Jong (133). The organism is grinum, M. fortuitum, and M. thamnopheos
widely distributed in soil and was named for its (82). In M. chelonei (M. borstelense) ammonia
fastidious demand for uric acid or allantoin as is formed from xanthine but not from uric acid
carbon and energy sources. No growth occurred (428). The data concerning purine degradation
when B. fastidiosus was inoculated in various by M. phlei are very inconsistent and contradic-
rich media (133). Recently the bacterium was tory (63, 132, 138, 169, 274, 433, 494) and do not
isolated and studied by Leadbetter and Holt allow any conclusion. As far as studied, the
(226, 305), Mahler (329), Kaltwasser (253), and mycobacteria can use purines and their degra-
Bongaerts and Vogels (62). The organism grows dation products only as a source of nitrogen, but
well in a synthetic medium containing uric M. phlei is claimed to be capable of a limited
acid, allantoin, or allantoate as the sole organic growth on uric acid alone (433). Most, if not all,
substrate but does not utilize common organic of the above-mentioned species contain urease

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


substrates other than these compounds (62, 133, as detected by a direct test (138, 274, 494) or by
226, 305). Even adenine, guanine, xanthine, an obvious deduction: the production of 4 mol of
and hypoxanthine are not used for growth ammonia from uric acid or allantoin (63, 274).
(253). A soluble uricase is induced by growth on Therefore, no conclusion can be made on the
urate and is almost absent in most strains involvement of allantoicase or allantoate ami-
when grown on allantoin. This enzyme is ap- dohydrolase in the pathways. M. butyricum
plied in clinical measurements of uric acid (213, was reported to be urease negative, but Klem-
329). The degradative pathway of allantoin is perer et al. (274) reported the presence of
catalyzed by S-allantoinase, allantoate amido- urease.
hydrolase, and S-ureidoglycolase (62). Glyoxyl- Actinomycetales. The results obtained with
ate is degraded along the tartronic semialde- Nocardia, Saccharopolyspora, and Actinoma-
hyde pathway (253). dura species are compiled in Table 8.
Mycobacteria. A large number of mycobacte- An unidentified streptomycete produces
ria were investigated for their ability to de- large amounts of uricase when cells grown in a
grade purines or the products of purine degra- peptone-glucose medium were incubated with
dation, either for taxonomic reasons or for test- uric acid or other purines (179, 582, 583). Uric
ing tuberculostatic compounds (63, 132, 138, acid degradation in these cells requires the
169, 274, 428, 433, 494). In most instances the presence of K+ ions. Allantoin accumulates
degradation was tested in suspensions of the quantitatively because of the absence of allan-
bacteria in phosphate buffers containing the toinase (583). Hypoxanthine is converted to
substrate (63, 138, 274, 428), and the production xanthine and 6,8-dihydroxypurine in a ratio of
of ammonia, glyoxylate derivatives, or '4CO2 3 to 1 by various species of Streptomyces. Both
was tested. products are converted to uric acid and are
Most of the species investigated were unable further degraded (584).
to produce ammonia from adenine, hypoxan- Various bacteria. Aeromonas hydrophila
thine, xanthine, uric acid (428), or allantoin grows poorly in media containing uric acid as a
(63, 494). The degradative pathway is incom- sole nitrogen source (433). Adenine is utilized
pletely present in the investigated species, but as a source of nitrogen by Azotobacter chroococ-
the part which is present leads to formation of cum and A. vinelandii, which contain adenine
ammonia in most cases. It starts with adenihe deaminase (227, 229). Guanine and allantoin
and guanine and is, therefore, most complete in are doubtful sources of nitrogen to these orga-
the case of Mycobacterium tuberculosis BCG nisms and uric acid is not used (227, 433). Cory-
(138); the data of Bonicke (63) are inconsistent nebacterium xerosis, C. minutissimum, C.
with these results, since he could not demon- striatum, C. diphtheriae, and C. bovis are able
strate the degradation of allantoin. to degrade uric acid (481), but C. pseudodiph-
M. thamnopheos (63, 428) and M. smegmatis theriticum is unable to grow in a medium con-
(138, 274, 428, 494) can degrade the compounds taining uric acid as the sole nitrogen source
from hypoxanthine on. However, some of the (433). Lactobacillus casei does not degrade pu-
data are contradictory for the latter organism rines (24). A study was made on the xanthine
as to hypoxanthine (138) and allantoin degrada- dehydrogenase of this organism (558). L. bul-
tion (63, 494). M. marinum (428) starts with garicus cannot degrade allantoin when this
xanthine, but Bonicke (63) did not observe al- compound is added to nutrient broth (613).
lantoinase activity in two strains tested. M. Micrococcus roseus (M. agilis) is unable to
butyricum (138, 274) and M. stercoris start with grow in a medium containing uric acid as a sole
uric acid, and the allantoin-degrading system is nitrogen source (433); however, this compound
present in M. fortuitum (63). Allantoinase is is used in this way by M. varians, in which the
reported to be present in M. vaccae, M. pere- presence of uricase was demonstrated (266). M.
VoL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 423
TABLE 8. Hydrolysis of purines by Actinomycetales
Substrate"
Organism Reference
Gua Ade Hypo Xan UA All
Nocardia otitidis-caviarum (N. - (+) + + +b 82, 192, 298
caviae)
N. brasiliensis + -c + -c _C +b 82, 192, 298
N. asteroides - -c _ -c +b 82, 192, 298
N. coeliaeca + + + + + 82
N. polychromogenes + + + + 132
N. blackwellii + 82
N. rubropertineta + 82

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


N. rubra + 428
N. opaca + + 132
N. erythropolis + 82
N. pellegrino + 428
N. corallina - +d +d _ 428
Saccharopolyspora hirsuta + + + 298
Actinomadura dassonvillei + + + 298
A. madurae - (+) - 298
A. pelletieri - + - 298
a
Abbreviations: Gua, guanine; Ade, adenine; Hypo, hypoxanthine; Xan, xanthine; UA, uric acid; All,
allantoin.
b Allantoinase present (192).
c Reported to be positive for one strain (428).
d
Reported to be negative for one strain (428).

luteus (Sarcina aurantiaca, S. lutea) utilizes DEGRADATION BY


guanine, xanthine, uric acid, and allantoin as ENTEROBACTERIACEAE AND
nitrogen sources (139). Allantoin can replace STREPTOCOCCI
urea as a requirement for T-strain mycoplas-
mas (331). Myxococcus virescens, M. fulvus, Enterobacteriaceae
and M. coralloides (Chondrococcus coral- Various authors have studied the ability of
loides) utilize adenosine and guanosine as Enterobacteriaceae to degrade uric acid and al-
sources of carbon, nitrogen, and energy (368). lantoin. The reported results are summarized
The nitrification process in Nitrosomonas euro- in Table 9. Although the results are contradic-
paea can occur at the expense of amino groups tory in some instances, one may conclude that
of guanine, uric acid, and allantoin, which sug- the ability to degrade uric acid and allantoin is
gests the presence of a degradation pathway for widely distributed among the Enterobacteria-
purines (439). Paracoccus denitrificans (M. ceae.
denitrificans) can use uric acid as a sole source Growth tests on uric acid and allantoin were
of nitrogen and carbon. Uricase is strongly in- proposed to differentiate the coli-aerogenes
duced by growth on uric acid (250). Spirillum group (282, 343). Escherichia coli was assumed
tenue utilizes xanthine, guanine, uric acid, and to be unable to use hypoxanthine (282), uric
allantoin as a source of nitrogen (132). acid (27, 282, 343, 433, 481), or allantoin (343),
Both hypoxanthine and guanine can be con- whereas Aerobacter aerogenes and A. cloacae
verted to adenine by Staphylococcus aureus, but could use these compounds as a sole source of
the reverse transition is not possible due to lack nitrogen (Table 9). However, a number of E.
of the ability to deaminate adenine (601). Xan- coli strains obtained from soil can use uric acid
thine is not degraded by S. aureus, but uric as a nitrogen source (104) and, when judged
acid and allantoin are used (481). S. aureus (S. under anaerobic conditions, a majority of E.
albus) does not utilize xanthine, guanine, and coli strains tested can use allantoin as a sole
uric acid as a nitrogen source, but allantoin is source of nitrogen, carbon, and energy (565).
used (132). Purines are deaminated only slowly Moreover, the nitrogen of xanthine (132, 343),
or not at all by Vibrio cholerae according to adenine (343), and guanine (132) was used byE.
Agarwala et al. (4), but Dikstein et al. (139) coli; cell suspensions of E. coli B convert iso-
demonstrated degradation of xanthine and uric guanine (6-amino-2-hydroxypurine) to xanthine
acid by this organism. (176). The capability of E. coli to use purines
424 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
TABLE 9. Ability of Enterobacteriaceae to use uric acid or allantoina
Organism Uric acid Allantoin Reference
Escherichia coli -N 282,.433
-N, +Nb 104
-N -N 343
(+)N, -CN (+)N, -CN 132
+0, -N 27, 517
+0 613
+N +N 132
+ CNcd 565
Citrobacter freundii -N -N 343

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


+CNd 565
Aerobacter aerogenes -CN 565
+0 613
+N 104, 282, 349, 517
+N, -CN +N, (-)CN 132
+N, +CN +N, +CN 343, 433
Klebsiella pneumoniae +CN +CN 433
Enterobacter cloacae (A. cloacae) -CN 565
+N, -CN +N, -CN 343
Serratia marcescens (also S. kiliensis) -CN 565
+N, -CN +N, (-)CN 132
+CN 433
Proteus rettgeri +CNd 565
P. vulgaris +CN, (-)N (-)CN, (-)N 132
+0 517
+0 613
516
P. mirabilis (-)N 433
Erwinia herbicola (Bacterium herbicola) (+)N (+)N 132
Paracolobactrum aerogenoides +CN +CN 509
a
Tests in which the compounds are tested as sole carbon, nitrogen, and energy source (CN), as sole
nitrogen source (N), or as a secondary carbon and energy source (0) for growth are represented as: +, good
growth; (+), weak growth; (-), uncertain growth; and -, no growth.
bStrains from feces -N; 50% of the strains from soils +N.
c Ten out of 16 strains were positive.
d
Under anaerobic conditions.

has been underestimated in the past since var- cescens (S. kiliensis) (433). Proteus vulgaris
ious enzymes of the catabolic pathway are pres- can use xanthine but not guanine as a nitrogen
ent in the cells, i.e., adenosine deaminase (68), source (132). In contrast to the above results,
adenine deaminase (68), uricase (496), and a set Dikstein et al. (139) found no degradation of
of enzymes similar to those found in Streptococ- xanthine by cell suspensions of E. coli, A. aero-
cus allantoicus, as will be discussed below. genes, and P. vulgaris.
Guanine and xanthine are used as a nitro- Salmonella typhimurium does not contain
gen source by Serratia marcescens (132, 433), physiologically significant amounts of adenine
A. aerogenes, and Erwinia herbicola (Bacte- deaminase. The conversion of adenine to hypo-
rium herbicola) (132). Adenine and hypoxan- xanthine takes place via adenosine and inosine
thine are used as a nitrogen source and to a less (225). The presence of uricase was demon-
extent also as sole organic substrates by A. strated in E. coli, Proteus morganii (Morga-
aerogenes, Klebsiella pneumoniae, and S. mar- nella), P. inconstans (Providencia), P. mirabi-
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 425
lis, and Serratia species (496). Growth on uric sorbitol, and salicin. Little or no acid was
acid is an adaptive property inA. aerogenes, K. formed from arabinose, xylose, fructose, galac-
pneumoniae, and S. marcescens (433). tose, or rhamnose. No acid was formed from
The degradative route of allantoin used by E. inulin, starch, or glycerol. Gelatin and sodium
coli, Citrobacter freundii (E. freundii), and P. hippurate are not hydrolyzed, but esculin is
rettgeri are similar to those described below for split. No ammonia is formed from arginine and
S. allantoicus and are perhaps common to all urea is -not split. No diacetyl is formed in sugar
Enterobacteriaceae. The contradictory results media. It can be isolated from black shore mud
represented in Table 9 are partly due to the (28), ditch mud (564, 565), and duck ponds (536).
conditions used during incubation. .The cata- The following products are formed under anaer-
bolic routes involve uricase, which is operative obic conditions per 100 mol of glucose: (+)-lac-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


only under aerobic conditions, whereas utiliza- tate, 106 mol; acetate, 36 mol; formate, 23.9
ble carbon compounds are produced under an- mol; CO2, 9.7 mol; and ethanol, 27.6 mol (28).
aerobic conditions only. The isolate is able to grow very well in media
containing no carbohydrate or polyhydroxyal-
STREPTOCOCCUS ALLANTOICUS cohol and is not identical to known Streptococ-
S. allantoicus was isolated in 1943 by Barker cus species. Therefore, Barker (28) created the
(28) from black mud from the shore of San new species Streptococcus allantoicus.
Francisco Bay. Later, this organism was iso- The guanine plus cytosine content of deoxy-
lated from various sources according to the en- ribonucleic acid, determined by chemical analy-
richment procedure of Barker, which involves sis (73) of three different strains (V4012, V4031,
anaerobic incubation in a medium containing and V4023), revealed values of 46.0, 47.5, and
allantoin and a small amount of yeast extract 49.7 (+ 0.3) mol% (unpublished data). These
(536, 565), followed by isolation of the strepto- values are rather high for streptococci.
cocci on agar plates containing glucose.
The same enrichment procedure resulted in Catabolic Pathway of S. allantoicus and
the isolation of another bacterium, which was Enterobacteriaceae
called Arthrobacter allantoicus (564, 565) but Barker (28) observed that growth ofS. allan-
was identified later on as P. rettgeri. toicus was much better under anaerobic than
S. allantoicus is a gram-positive, catalase- under aerobic conditions. The presence of a
negative, nonmotile streptococcus, with spheri- small amount of yeast extract in the growth
cal or ovoid cells that are 1.1 to 1.5 ,um in medium is required, perhaps because biotin is a
diameter, mostly occurring in pairs or short cofactor for growth. Under anaerobic conditions
chains; the chains are formed by pairs oflanceo- 100 mol of allantoin are converted to 226 mol of
late cells. No reaction was observed with group ammonia, 62.3 mol of urea, 14.8 mol of acetate,
A-T sera according to Lancefield (565). Barker 1.5 mol of lactate, 168 mol of CO2, 9.4 mol of
(28) reported the production of dextran as an formate, 44.8 mol of oxamate, and 13.8 mol of a
extracellular polysaccharide for his isolate, but compound which was thought to be glycolate
a negative cross-reaction with type II pneumo- (28). The identification and quantitative deter-
coccus antiserum proves the absence of dextran mination of oxamate as a new microbial prod-
in the isolate of Vogels (565). The action on uct was a stimulus for further studies. More-
blood is indifferent (gamma hemolysis) (28, over, Barker pointed to the fact that the ratio of
565). The colonies on agar media are partly ammonia to urea is constant (3.64:1). He con-
transparent, circular (1 to 2 mm), and convex cluded that ammonia is formed directly from
with undulate margin. The bacteria grow at 10 allantoin or some intermediate, rather than by
and 40 C but not at 45 C (565). Barker (28) did hydrolysis of urea, since cell suspensions of S.
not observe growth above 36.5 C. Treatment for allantoicus are unable to decompose urea (28,
15 min at 50 C kills the cells (565). They grow in 565). Barker (29) proved also that allantoate is
yeast extract-glucose media containing 6.5% readily decomposed by whole cells of S. allanto-
NaCl (28, 565). No growth was observed at pH iCus.
9.6 on blood agar containing 40% bile or in skim Later studies (565) revealed that P. rettgeri,
milk containing 0.005% methylene blue. E. coli, and C. freundii produce oxamate from
Growth is strongly inhibited by bacitracin (5 U/ allantoin; the amounts formed per 100 mol of
ml). Litmus milk is weakly acidified with some allantoin are 59, 51, and 53 mol of oxamate,
reduction but not curdled. The final pH in yeast respectively. Growth of the bacteria is strongly
extract-glucose was 5.0 to 5.3. Acid, but no gas, inhibited in the presence of oxygen, and under
was slowly formed from glucose, maltose, lac- this condition S. allantoicus formed 80 mol of
tose, sucrose, trehalose, raffinose, mannitol, oxamate per 100 mol of allantoin degraded,
426 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
whereas the amount of urea dropped to 20 mol which is present in S. allantoicus (530, 533,
(565). These results suggest that urea and oxa- 565), P. rettgeri (565), and E. coli (284). The
mate are formed along different pathways, i.e., reaction is stimulated by the presence of thia-
an oxidative one resulting in the formation of mine pyrophosphate (284, 530), Mg2+ ions (284,
oxamate and a reductive one in which urea is 533, 565), and anaerobic conditions (284, 565).
produced. In the presence of oxygen the interre- Tartronate-semialdehyde reductase. Tar-
lation between the pathways is disturbed, and tronate-semialdehyde reductase of S. allanto-
the oxidative route prevails. The degradative icus (193, 530) catalyzes the reversible (533)
routes were resolved by studies of Valentine et conversion of tartronate-semialdehyde to glyc-
al. (59, 530, 531, 533, 535-539), Vogels (564, erate in the presence of NAD. In tests in which
565), and Van der Drift et al. (547). both tartronate-semialdehyde synthetase and
Conversion of allantoin to ureidoglycolate. tartronate-semialdehyde reductase are allowed

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


The catabolic pathway followed by S. allanto- to act on glyoxylate, glycolate is formed (533).
icus and Enterobacteriaceae (E. coli, E. coli Glycerate is converted to 3-phosphoglycerate,
var. acidilactici, C. freundii, and P. rettgeri) is which can yield pyruvate via the Embden-Mey-
described in Fig. 8. The enzymes involved in erhof pathway and acetate and formate by py-
the degradation of allantoin to glyoxylate are ruvate degradation (530). Acetate and formate
discussed extensively in a previous section. were demonstrated as products of allantoin deg-
The allantoinases of this group of organisms radation by Barker (28).
exhibit a number of common properties which Oxidative production of oxalurate. Ureido-
distinguish them from the allantoinases of glycolate is dehydrogenated to oxalurate by urei-
other microorganisms, plants, and animals. doglycolate dehydrogenase in S. allantoicus
They are activated by Mn2+ ions and reducing (530, 538, 565), P. rettgeri (547, 565), and E. coli
substances and are the only known aspecific (565). The purified enzyme from P. rettgeri con-
allantoinases; i.e., R- and S-allantoin are de- verts specifically S(-)-ureidoglycolate and uses
graded with about equal velocities (570, 571). both NAD and NADP equally well. A number
This may reflect the scavenging character of of other hydroxy acids cannot replace ureido-
the degradation of allantoin by these orga- glycolate as a substrate. The pH optimum is 8.0
nisms, since the natural dextrorotatory S-al- to 8.4, and the enzymatic activity depends
lantoin (196) is subject to a rapid racemization strongly on the ionic strength of the solution,
in neutral and alkaline media (569). The prod- being optimal atl = 0.07 to 0.12. The reversibil-
uct of the allantoinase reaction is allantoate, ity of the reaction could not be demonstrated,
which was reported as an intermediate in the perhaps due to the fact that the equilibrium
degradation by Barker (29). position of the reaction lies strongly in the di-
Allantoate is not degraded by allantoicase, rection of oxalurate formation (547).
since less than 2 mol of urea and a substantial The chemical relationship between uric acid
amount of ammonia are formed from allantoate and allantoin on one hand and oxalurate on the
by the urease-negative S. allantoicus, E. coli, other hand has been known for a long time.
and C. freundii. A new enzyme was found, Wohler and Liebig (599) prepared oxalurate in
allantoate amidohydrolase (allantoate amidi- 1838 by treatment of uric acid with moderately
nohydrolase [decarboxylating] [EC 3.5.3.9]), concentrated nitric acid. Biltz and Schauder
which catalyzes the conversion of allantoate (52) prepared oxalurate from allantoin by oxi-
into S(-)-ureidoglycolate, ammonia, and CO2 dation of the latter with H202 and from uric
(545, 550, 551, 565, 566). S(-)-ureidoglycolase is acid with KMnO4 in alkaline solution. Oxalur-
present in S. allantoicus (182, 183, 537, 565), in ate is formed among other products from 14C-
P. rettgeri, and probably also in E. coli and C. labeled urate by a catalase-ethyl hydrogen per-
freundii (565). The intermediary function of oxide system (97).
glyoxylate in the degradation of allantoin by S. The occurrence of oxalurate in human urine
allantoicus was established by Valentine et al. was claimed in 1867 by Schunck (467), in 1868
(531). by Neubauer (366), and in 1938 by Flas-
Reductive degradation of glyoxylate. Glyox- chentrager and Muller (159). Oxalurate was
ylate is converted to glycerate according to a supposed to be involved in the production of
reaction sequence similar to that described by renal calculi composed wholly or partly of cal-
Kornberg and Gotto (193, 281) and Dagley et al. cium oxalate (101). It was thought (101, 487)
(122) in the aerobic glycine metabolism by a that uracil is converted to oxalurate in dogs; N-
pseudomonad. Tartronate-semialdehyde, which formyloxalurate was supposed to be an inter-
was identified in various ways (530, 533, 565), mediate in this conversion. Parabanate was
and CO2 are formed in a reaction catalyzed also reported as a source of oxalurate, which is
by tartronate-semialdehyde synthase (Fig. 8), formed by parabanase.
VOL. 40, 19f76 DEGRADATION OF PURINES AND PYRIMIDINES 427

CO-NH NH2
COOH COOH
CO - +H20 +H20 4 + CO
Parabanase CO-NH-CO-NH2 Oxaluricase COOH
CO-NH NH2
(Parabanic acid) (Oxaluric acid) (Oxalic (Urea)
acid)
This enzyme was considered to be present in The purified enzyme exhibits an absolute re-
liver from frogs (290) and in microbes (78, 136, quirement for bivalent ions. Mg2+ and Mn2+
198). Both reaction sequences leading to oxalur- ions yield the highest activity, but Ca2+ and
ate are not well established and are questiona- C02+ ions can partly substitute (59, 511, 536,
ble. The salts of parabanate are hydrolyzed 565). The enzyme differs from ornithine car-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


readily to salts of oxalurate in aqueous solution bamoyltransferase (carbamoylphosphate:L-or-
(15), whereas the latter are relatively stable at nithine carbamoyltransferase [EC 2.1.3.3]) (59)
pH values below 11 and are hydrolyzed to oxa- and is inactive with carbamoyl derivates of a
late and urea in strongly alkaline solutions number of amino acid tested and with N-
(143). formyloxalurate, acetylurea, formylurea, and
Oxaluricase was reported to be present in biuret. Moreover, acetyl phosphate cannot
dogs (101, 487), in the livers of frogs (290), in replace carbamoyl phosphate (511, 565).
Aspergillus niger (78), in Saccharomyces cere- If tested in the presence of phosphate, equi-
visiae (136), and in an unidentified bacterium molar amounts of oxalurate and phosphate are
(198). New studies are required to prove the needed (536). Replacement of phosphate by ar-
presence of this enzyme since at least a number senate increases the reaction rate, and ammo-
of the results can be explained by assuming the nia and C02 are formed instead of carbamoyl
presence of oxamate transcarbamoylase, which phosphate (530, 535, 538). As a consequence,
is described below. arsenate is not used in stoichiometric amounts
Oxamate formation. The presence of oxamate (538).
among the end products of allantoin degrada- The formation of carbamoyl phosphate was
tion by S. allantoicus was established by Bar- demonstrated by coupling the oxamate trans-
ker (28). carbamoylase reaction to two transferases. (i)
Oxamate has been reported to occur in na- Other carbamoyltransferases, such as orni-
ture in only one other source. Kminek (277) thine carbamoyltransferase (present in the cell-
demonstrated its occurrence in beets. Since free extracts of S. allantoicus [530, 535, 538,
these plants also contain allantoin (276), a simi- 5641 and P. rettgeri [564, 5651), can be used in
lar relationship between allantoin and oxamate this way, but the carbamoyl group can also be
may exist in plants as is described here for S. transferred to glycine or citrulline by enzymes
allantoicus. Oxamate is used as an inhibitor of present in both organisms (565). (ii) ATP:car-
lactate dehydrogenase (370). bamate phosphotransferase (EC 2.7.2.2), pres-
The enzyme catalyzing oxamate formation in ent in S. allantoicus (531, 535, 539) and P.
S. allantoicus was first studied by Valentine et rettgeri (564, 565) can also be used.
al. (60, 531, 535, 539). A phosphorolytic or ar- The latter enzyme catalyzes the following re-
senolytic cleavage of oxalurate (carbamoyloxa- action: NH2 -- CO P + ADP w± NH3 + C02 +
-

mate) is catalyzed by oxamate transcarbamoy- ATP. The enzyme is stimulated by Mg2+ and
lase (carbamoylphosphate:oxamate carbamoyl- Mn2+ ions (242, 565) and furnishes energy for
transferase [EC 2.1.3.-I). Oxalurate accumu- cell growth during allantoin degradation (536).
lates during the degradation of allantoin or Cell-free extracts of P. rettgeri contain also a
allantoate in the absence of phosphate and ar- hydrolytic enzyme which degrades carbamoyl
senate (530, 531, 539) and during the degrada- phosphate in the absence of adenosine 5'-di-
tion of allantoin by cell suspensions in the pres- phosphate (ADP) (565). A similar enzyme was
ence of ethylenediaminetetraacetate (564). found in Clostridium uracilicum by Campbell
The enzyme is also present in P. rettgeri, (92). The reaction sequence involved in ATP
group D streptococci, and E. coli (511). How- formation from oxalurate resembles the phos-
ever, Valentine et al. (530) reported its absence phorolytic cleavage of citrulline described by
in E. coli K-12. Jones et al. (243) for Streptococcus faecalis.
The enzyme catalyzes a reversible reaction Valentine and Wolfe (538) discussed a phos-
(59, 511) (Fig. 8) with an apparent equilibrium phorylytic cleavage of urea by S. allantoicus.
constant (59): Such cleavage could be brought about by the
KaPP = combined action of ureidoglycolase, ureidogly-
[oxaluratel [phosphate] 0.623 colate dehydrogenase, oxamate transcarbamoy-
[oxamate] [carbamoyl phosphate] lase, and enzymes hydrolyzing oxamate to oxa-
428 VOGELS AND VAN DER DRIFTBBACTECRIOL. REV..
,NH co NH2 NH2 OC / N
oc\ CO I Co
NH / NH NH /NH
H H
S ( - Allantoin R -) -Allantoin
+ H20 RS-Allantoinase
COOH

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


NH2- CO- NH hfNH -CO -NH,
H NH3+ CO2 +ATP
Allantoic acid JA TP: carbamate
+ H20 Allantoate amidohydrolasie 8 phosphotrans -
ADP ferase
NH3+ CO2 COOH COOH
+ I
NH2- CH -NH -CO -NH2 OC-NH2 + NH2-CO-O-PO3(
Oxamate Carbamoyl phosphate
Ureidoglycine
+ H20 Allantoate amidohydrolasi Oxamate
transcarbamoylase
COOH S /-J- IIreidoglycolate COOH
I dehy
vdrogenase
HO-J-NH-CO-NH2 OC -NH-CO-NH2
H
S(-)-Ureidoglycolic acid Oxaluric acid
i NAD (P) NAD (P) H2
S(-)-Ureidoglycolase
NH2_ CO- NH2 COOH
+ I
Urea OCH
Glyoxylic acid
tTartronate -semi
aldehyde NADH2 NAB
synthase
COOH COOH
1/2 CHOH 1/2 CHOH
I Tartronate -semialdehyde I
COH reductase CH20H
Tartronate -semialdehyde glyceric acid

Further degradation
FIG. 8. Degradation of allantoin by Streptococcus allantoicus and Enterobacteriaceae.
late and converting the latter to glyoxylate. icus, P. rettgeri, andE. coli can be explained by
However, the two latter enzymes are not yet the presence of allantoate amidohydrolase in-
found in S. allantoicus. Moreover, urea ap- stead of allantoicase.
pears to be a final product, and its apparent Concluding remarks. The reactions involved
degradation in the urease-negative S. allanto- in the anaerobic degradation of allantoin by S.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 429
allantoicus and Enterobacteriaceae are given banate, it is highly probable that the enzyme
schematically in Fig. 9. The expected fermenta- was induced by oxalurate.
tion balance, accounting for the reduction
equivalents but not for the products of further Purine Degradation by Other Streptococci
degradation of pyruvate, is represented: 100 al- Among the streptococci only strains of S.
lantoin + 33 Pi + 33 ADP + 266 H20 -* 33 faecalis exhibit activity against degradation
oxamate (45) + 33 ATP + 66 urea (62) + 166 products of purines. S. agalactiae, S. lactis,
C02 (168) + 233 NH3 (226) + 33 pyruvate (15 and a ,B-hemolytic streptococcus did not grow in
acetate, 15 lactate, 9 formate, 14 unknown). a medium with uric acid as a sole nitrogen
The values found by Barker (28) are given source (433). S. hemolyticus cannot decompose
within the parentheses; they are very close to allantoin present in nutrient broth (613).

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


the expected values. Mead (334) tested a number of strains of S.
In the presence of oxygen, NADH2 is oxidized faecalis, S. faecium, and S. durans for their
by NADH2 oxidase present in the cells of S. ability to degrade uric acid present in solid
allantoicus and P. rettgeri (565). As a conse- media. Positive results were only obtained with
quence, the amount of oxamate rises and the S. faecalis. When incubated in air, a few
amount of urea drops, which results in a strong strains of S. faecalis were positive, although
inhibition of the growth of these organisms Young and Hawkins (613) found no degrada-
since no carbon becomes available to the cells. tion, but under anaerobic conditions a large
Thus, the anaerobic character of the degrada- majority of the strains decomposed uric acid.
tion of allantoin by these organisms can be Highly active strains were found among the
explained on the basis of the reactions involved isolates from chicken cecal samples and sam-
in the catabolism. ples of human feces. None of the strains was
Substantial amounts of glycine are formed able to produce ammonia from adenine, gua-
during the degradation of allantoate by prolif- nine, or xanthine (334), but Dikstein et al.
erating cells, resting cell suspensions, and cell- (139) presented evidence for xanthine degrada-
free extracts of S. allantoicus but not of P. tion by S. faecalis.
rettgeri. Glycine may result from an enzymatic These results taken in combination with the
or nonenzymatic transamination involving ur- studies of Tigier and Grisolia (511) on the pres-
eidoglycine and glyoxylate (545, 565). ence of oxamate transcarbamoylase in group D
The enzymatic system responsible for the ca- streptococci indicate that further studies are
tabolism of allantoin is inducible and only pres- needed on uric acid and allantoin degradation
ent in cells of S. allantoicus grown on allantoin by S. faecalis. The unique and similar routes
(31). Cells grown on glucose contain no oxa- followed by S. allantoicus, which may be eco-
mate transcarbamoylase (59), no ureidoglyco- logically related to S. faecalis and Enterobacte-
lase, and only trace amounts of tartronate-sem- riaceae, should be considered against the back-
ialdehyde synthase (183). Tigier and Grisolia ground of possible gene transfer among these
(511) demonstrated that oxamate transcarba- enteric organisms.
moylase is induced in group D streptococci by
parabanate but only slightly by allantoin. ANAEROBIC DEGRADATION OF
Since these authors used autoclaved para-
PURINES
Clostridium acidiurici and C. cylindrosporum
RS-Allantoin Liebert, working in Beijerinck's institute at
3H2O
.2NH3 + CO? pi
Delft, isolated in 1909 a sporeforming, obliga-
S-Ureidogtycolate Ozalurate * Oxamate
tively anaerobic bacterium that decomposed
NAO(P) NAO(PIH2 uric acid only under anaerobic conditions. This
organism, C. acidiurici (Bacillus acidi-urici),
Urea + Glyoxylate Carbamoyl phosphate breaks down uric acid to CO2, acetic acid, and
'/2 NADH2 ADP ammonia. Glycine could not be detected as a
N' ATP
/2NAO product, but Liebert suggested that it is con-
/2C02 + 1/2 Glycerate NH3 + CO2 verted to acetate (314). Uricase is not involved
1/UH20 in this degradation, since allantoin and allan-
toate are not converted to ammonia or CO2 by
1/z Pyruvate Acetate, lactate, formate, cell-C cell suspensions (32) or cell-free extracts of C.
hG. 9. Schematic outline of allantoin degrada- acidiurici (409), nor are allantoin or urea at-
tion by Streptococcus allantoicus and Enterobacteri- tacked in the presence or absence of urate (32).
aceae. In 1941, Barker and Beck (32, 33) observed
430 VOGELS AND VAN DER DRIFT BACTICIOL. REV.
that bacteria capable of decomposing uric acid ine, and uric acid riboside. These results sug-
under anaerobic conditions are widely distrib- gest the presence of a rather specific purine-
uted in soils, and they isolated two organisms nucleoside phosphorylase (purine-nucleo-
from soil. Both organisms, C. acidiurici and C. side:orthophosphate ribosyl-transferase [EC
cylindrosporum, grow in media containing cer- 2.4.2.1]) in this organism (398). Washed cells of
tain purines (as the principal source of carbon C. cylindrosporum grown on uric acid form CO2
and nitrogen) and small amounts of yeast au- from guanosine but not from inosine and xan-
tolysate. The organisms were initially differen- thosine (398). The nucleosides are poor growth
tiated on a morphological basis, but further substrates (if used at all) for both clostridia (32,
studies also revealed differences in the fermen- 33, 398).
tation products. C. cylindrosporum forms gly- Guanine is converted to xanthine by guanine
cine as a product of purine fermentation, deaminase (guanine aminohydrolase [EC

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


whereas this amino acid could not be detected 3.5.4.3]) present in C. cylindrosporum (398) and
in fermentation liquors of C. acidiurici (32). is decomposed by growing cultures of both clos-
Considerable evidence was obtained, which im- tridia (32, 33, 398). Adenine (32, 33, 398) and
plicated glycine as an intermediate in the fer- various guanine isomers (398) are not used.
mentation of purines by C. acidiurici (32). Fur- Hypoxanthine is attacked by growing cul-
thermore, C. cylindrosporum forms 1.0 mol of tures and cell suspensions of C. acidiurici after
formic acid and 0.4 mol of acetic acid per mol of adaptation (32) but less readily by C. cylindros-
guanine fermented, whereas C. acidiurici porum (398). This could be explained on the
forms 1.0 mol of acetic acid and only 0.2 mol of basis that hypoxanthine is less readily attacked
formic acid (397). A typical fermentation by xanthine dehydrogenase (66, 398), but the
balance for C. cylindrosporum is given in Table lack of oxidizing equivalents formed in the fer-
10. Growth of this organism is more rapid on mentation may, as well, render hypoxanthine
umic acid than on guanine, but similar products (398) and purine (32) as poor substrates for the
are formed. However, the yields of formic acid organism. Hypoxanthine is converted to 6,8-
and glycine from uric acid are only half those dihydroxypurine by xanthine dehydrogenase of
from guanine (397). C. acidiurici grows vigor- C. cylindrosporum (66). 6,8-Dihydroxypurine is
ously in media containing uric acid but de- oxidized by xanthine dehydrogenase to uric
velops poorly or not at all in media containing acid, which in turn is reduced to xanthine (66,
complex nitrogenous material such as tryptone 398). The coupling of these reactions does not
or yeast extract in the absence of added pu- result in a net requirement for reducing or
rines. The ability of this organism to attack oxidizing equivalents, and this may explain
nitrogenous compounds is very restricted (32). why 6,8-dihydroxypurine is decomposed by
Degradation of purines. Cell-free extracts of washed cell suspensions at a higher rate than
C. cylindrosporum convert guanosine and ino- either uric acid or hypoxanthine (398).
sine into guanine and hypoxanthine but only in Uric acid and xanthine are readily decom-
the presence of phosphate (Fig. 10). Simultane- posed by growing cultures of both clostridia (32,
ously, ribose-1-phosphate is formed, but no re- 33, 398). Uric acid is utilized much more rapidly
action was detected with adenosine, xanthos- by C. acidiurici (32), whereas cells of C. cylin-
drosporum use xanthine better than uric acid
TABLE 10. Typical fermentation balances of C. (398). These results indicate that the former
cylindrosporum and C. acidiurici organism is able to produce the reducing equiv-
C. cylindrosporum C. acidiurici
alents needed for uric acid conversion more
readily and may even produce these equiva-
Cells Cell-free lents superfluously.
growing extract The conversion of uric acid to xanthine is
Product on gua- and xan- Cell suspensions reversible (66, 398). In cell-free extracts urate
nine thine and formiminogly- decomposition is quickly and totally inhibited
(mol/100 (mol/100 cine (mol/100 mol
mol of mol of of substrate) by oxygen (409), but cell-free extracts and dried
sub- sub- cell preparations decompose xanthine both in
strate) strate) the presence and absence of oxygen. Methylene
NH3 480 240 200 blue is used as an electron acceptor and reduced
CO2 280 160 92 benzyl viologen is used as an electron donor in
Glycine 33 71 the xanthine dehydrogenase reaction (398). The
Formic acid 99 97 natural electron donor in uric acid degradation
Acetic acid 38 112 is reduced ferredoxin. Its role will be discussed
Reference (397) (398) (400, 404, 408)
below.
Conversion of xanthine to formiminogly-
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 431
H H H
N1 6 5 C7 \ - < CNv NC
I? jj eCH
H C,<3 4C -- /
0 H H
11 Purine 8 -Hydroxypurine
HN CC N
O |A
11 H
Inosine C II CH
_~ HN c N
Pi '~~CN H A I I c=a

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Purine nucleoside Hypoxanthine N H
phosphorylase Ribose -1 -phosphate 6,8 - Dihydroxypurine
Pi t O
0 0 IA
~ AH
1
HN C \\ HN C N\
I.+H20,-NH3 I 1 ~,CH I II /C=o
Guanine N H N H
H
Guanine deaminase Xanthine Uric acid

Further degradations
FIG. 10. Conversion of purines to xanthine by Clostridium cylindrosporum. (A) refers to reactions cata-
lyzed by xanthine dehydrogenase (66).

cine. The degradative pathway of xanthine was CO?


resolved by two methods, i.e., studies with la-
beled xanthine, uric acid, formiminoglycine,
t
glycine, and CO2 and studies on the successive 0
reactions. 11 I'

The results of the experiments with labeled HN1 6 C 7


N 6C~
compounds were obtained with growing cells as I 1I 9CH'
C N , C 9N
2
well as with washed cell suspensions and are O
~NH N
summarized in Fig. 11. Besides the main routes /
indicated in this figure, an extensive conver- C02 Xanthine
sion of CO2 to formic acid, acetic acid, and
glycine was observed in studies with C. cylin- N ,'
drosporum (397). The methyl group of acetic H2C
acid contained 5.2 times the activity of the car-
boxyl group (397, 466). In contrast, Karlsson Formiminogtycine UCOH,' CH
and Barker (259) and recently Schulman et al. NH HCOOH 4-. CO2
'-

(466) reported only 1.5 times more labeling in Formic acid


the methyl group in studies with C. acidiurici.
These different results will be discussed later in
this section. CH3N,-- CO2
Radin and Barker (409) observed that during 1/NH2 "I

the degradation of purines by C. acidiurici sub- H2C -C COOH--- CO2


stances were formed that reacted positively in I Acetic acid
the Pauly test. Therefore, the initial split in the C02 ------0 COOH *CO02
purines occurs in the pyrimidine ring to yield
imidazole derivatives. Cell-free extracts of both Glycine
clostridia degrade xanthine to the same prod- FIG. 11. Degradation of purines by Clostridium
ucts as growing cultures, but acetate is not cylindrosporum (34, 397) and C. acidiurici (259,
formed (397, 409). Results of a typical experi- 397, 408, 446). Main (solid lines) and secondary
ment with cell-free extracts of C. cylindrospo- (dashed lines) routes of labeled carbon atoms.
432 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
rum are presented in Table 10. allantoin, barbituric acid, and dihydroorotic
The successive reactions of the degradation of acid to allantoic acid, ureidomalonic acid, and
xanthine to formiminoglycine were studied carbamoyl aspartic acid, respectively. These re-
with cell-free extracts of C. cylindrosporum actions will be discussed elsewhere in this re-
(Fig. 12). However, the quantitative and quali- view.
tative similarity of the products formed and the The subsequent intermediate, 4-amino-5-imi-
studies with labeled compounds revealed strong dazolecarboxylic acid, accumulates when xan-
evidence that the degradative routes are quite thine (393) or 4-ureido-5-imidazolecarboxylic
similar for both clostridia. Crude extracts con- acid (403) are degraded by extracts of C. cylin-
vert xanthine quantitatively to 4-ureido-5-imi- drosporum above pH 8.5. 4-Amino-5-imidazole-
dazolecarboxylic acid in the presence of seques- carboxamide is not degraded by the extracts
tering agents, i.e., ethylenediaminetetraace- (259, 403), nor does it accumulate under condi-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


tate, a,a'-dipyridyl, or potassium cyanide, tions favorable for xanthine decomposition (30).
since Mn2+, Fe2+, or, to a less extent, Mg2+ ions The enzymatic conversion of 4-ureido-5-imi-
are required in the subsequent reaction (403). 4- dazolecarboxylic acid resembles the degrada-
Ureido-5-imidazolecarboxylic acid was isolated tion of allantoate by allantoate amidohydrolase
from the incubation mixture and was found to and the oxalurate degradation by oxamate
be identical to the authentic substance synthe- transcarbamoylase. These enzymes are opera-
sized by the authors (396, 403). Rabinowitz and tive in Streptococcus allantoicus and Entero-
Pricer (403) pointed out the similarity of this bacteriaceae and deliver ammonia and CO2,
reaction to those involved in the conversion of and carbamoyl phosphate, respectively. No
studies are available to establish whether car-
bamoyl phosphate is also produced from 4-
0
ureido-5-imidazolecarboxylic acid, which would
11 COOH render the clostridia an extra source of energy
HN C N iH20 H2N C N as supposed by Barker (30).
I II CH II CH The decarboxylation of 4-amino-5-imidazole-
=C
N N N H
carboxylic acid to 4-aminoimidazole was dem-
H H H onstrated by Rabinowitz (393) who incubated
Xanthine 4 -Ureido -5 -imidazole -
the substrate with crude extracts in the pres-
carboxylic acid
ence of ethylenediaminetetraacetate. Both 4-
amino-5-imidazolecarboxylic acid and 4-ami-
(2)
Mn2+or Fe 2+ noimidazole formed in the latter two reactions
+ H20
were isolated and found identical to the com-
NH3t CO2 COOH pounds produced by catalytic reduction of the
CO2
+ HC-N -N corresponding nitroderivatives (393).
Crude extracts convert 4-aminoimidazole to
11 CH COH CH
ammonia and formiminoglycine (404). The re-
- 3) 1l /C
H2N _ C /
H2N N action is a two-step one, and 4-imidazolone was
H
4 -Aminoimidazote 4-Amino-5-imidazole
identified as an intermediate (174). The first
-
enzyme involved in the degradation of 4-ami-
Fe2' |H20 carboxylic acid noimidazole, 4-aminoimidazole aminohydro-
(4) + lase (EC 3.5.4.8), was purified by Rabinowitz
H2C N~
+H20 H --
\ and Pricer (404) and requires Fe2+ ions for full
NH3+ CH - activity. C02+, Mn2+, or Ni2+ ions can replace
,0 CH/ COOH yn Fe2+ ions to some extent, but the former two
H
reduce the activity observed in the presence of
4 - Imidazolone Formiminogtycine Fe2+ ions. The enzyme was activated and stabi-
lized by the presence of cysteine and various
FIG. 12. Conversion of xanthine to formiminogly- other reducing agents (404). 4-Imidazolone is
cine by C. cylindrosporum and C. acidiurici. En- easily transformed nonenzymatically into for-
zymes involved: (1) xanthinase (xanthine amidohy-
drolase [EC 3.52.-); (2) 4-ureido-5-imidazolecar- miminoglycine (174) and into glycine under
boxylate amidohydrolase (EC 3.5.1.-); (3) 4-amino- acid and alkaline conditions (404). An enzyme
imidazolecarboxylate decarboxylase (4-amino-5-imi- catalyzing the conversion of 4-imidazolone to
dazolecarboxylate carboxy-lyase [EC 411.-]; (4) 4- formiminoglycine was found to be present in C.
aminoimidazole deaminase (4-aminoimidazole ami- cylindrosporum (174).
nohydrolase [EC 3.5.4.8]); (5) 4-imidazolonase (4- Similar splits of imidazole rings to formimino
imidazolone amidohydrolase [EC 3.52.-D. derivatives were observed in the conversion of
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 433
imidazole via imidazolone to formiminoglycine Conversion of formiminoglycine. Formimi-
by a pseudomonad, ATCC 23438, which is capa- noglycine is converted to glycine and (-)-5-
ble of growing on imidazole as a sole source of formimino-tetrahydrofolate (THFA) (Fig. 13)
carbon and nitrogen (372), in the conversion of by glycine formiminotransferase, which was
urocanic acid to L-4-imidazolone-5-propionate first demonstrated in extracts of C. cylindro-
by urocanase (4-imidazolone-5-propionate hy- sporum and was partially purified from that
drolyase [EC 4.2.1.49]), and in the conversion of source (402, 405, 523). 5-Formimino-THFA is
the latter to N-formiminoglutamate by imida- labile at 37 C in aqueous solutions with a half-
zolonepropionase (4-imidazolone-5-propionate life time of 60 min and can be determined by
amidohydrolase [EC 3.5.2.7]) (255, 413, 423).
Formiminoglutamate may be converted in ani- H 7 H

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


mal liver to glutamate by the transfer of the N N
formimino group to tetrahydrofolic acid (493) or H2C" NCH HC- CH2-N-
may be hydrolyzed to ammonia and N-formyl- 1I +
COOH NH CH2
L-glutamate, which yields glutamate and
formate in a pseudomonad (595) and in Formiminoglycine (-H-Tetrahydrotolic acid
Pseudomonas fluorescens (325, 492), or may be (1 (THFA) CH=NH
(I)\ I. H
split to glutamate and formamide inAerobacter ..ONH2 + N.- I
aerogenes (323, 325, 423), Salmonella typhimu- H2C HC- CH2-N--
rium (335), and Bacillus subtilis (255). Form- COOHU CH2
amide cannot be metabolized further and accu- Glycine (-5 Formimino -THFA
-

mulates in histidine-containing cultures of A. +H+


aerogenes (364); it also accumulates as a prod- (2)
uct of histidine fermentation by Clostridium THFA+NAD ssNH3 i
+o CH
tetanomorphum (576). THFA 1t)
Utilization of formiminoglycine and gly- Gtycine synthase HC-CH2-N-
+ 4 system NAH2 /C2
cine by whole cells. Formiminoglycine is not CH20H*-
used for growth by either C. cylindrosporum or NADPH2
C. acidiurici (404, 408). It is converted to gly- HC -NH2 CO2+ CH,+ (NAD) 1+)-5,10-Methenyl-THFA
cine, when incubated with washed cell suspen- COOH NADH2 NAD>P /
sions of C. cylindrosporum, or to acetic acid, L- Serine ,+ H20 ,>3) (51
CO2 and ammonia (Table 10) when incubated N CH2 H H
with washed cell suspensions of C. acidiurici HC-CH2-N-
N CH O
(400, 404, 408). Cell suspensions of C. acidiurici CH2 ~~HC-CH2-N-
decompose formiminoglycine, but glycine is not
decomposed under comparable conditions, nor 1+1 -5,10-Methyleine-THFA oCH2
is the rate of formiminoglycine conversion af- AOP+ Pi 10-Formyl-THFA
fected by the addition of glycine. However, gly-
cine is completely utilized if formiminoglycine ATP 6
is also present in the incubation mixture. Up to
3.7 mol of glycine could be utilized per mol of CO2 4-*Formate +F- THFA
(7)
formiminoglycine degraded. A number of other FIG. 13. Role of tetrahydrofolic acid (THFA) de-
formimino derivatives or one-carbon com- rivatives in the degradation of formiminoglycine by
pounds, including formate and formaldehyde, C. acidiurici and C. cylindrosporum. Enzymes in-
were not able to replace formiminoglycine in its volved: (1) glycine formiminotransferase (5-formi-
stimulating role in the utilization of glycine mino-THFA:glycine N-formiminotransferase [EC
(400, 408). These results indicate that the formi- 21.2.4]); (2) Formimino-THFA cyclodeaminase (5-
mino group derived from formiminoglycine formimino-THFA ammonia-lyase [cyclizing] [EC
plays an essential part in the degradation of 4.31.41); (3) methylene-THFA dehydrogenase (5,10-
glycine. methylene-THFA: NADP+ oxidoreductase [EC
Glycine stimulates urate degradation by cell 1.5.1.51); (4) serine hydroxymethyltransferase (5,10-
suspensions of C. acidiurici, probably because methylene-THFA: glycine hydroxymethyltransferase
[EC 21.21]); (5) methenyl-THFA cyclohydrolase
it serves as a reducing agent (409). Glycine is (5,10-methenyl-THFA-5-hydrolase [decyclizing] [EC
degraded by growing cultures in the presence of 3.5.4.9]); (6) formyl-THFA synthetase (for-
the fermentable substrates, uric acid, xan- mate:THFA ligase [ADP-forming] [EC 6.3.4.3]); (7)
thine, and guanine. Approximately 1.5 mol of formate dehydrogenase; (8) glycine synthase (5,10-
glycine was degraded per mol of uric acid uti- methylene-THFA:ammonia hydroxymethyltransfer-
lized (32). ase [carboxylating, reducing] [EC 21.210]).
434 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
heating the solution for a short period at 100 C sporum, respectively. The equilibrium of the
in acidic medium. 5,10-Methenyl-THFA is reaction favors ATP utilization, but the activity
formed and can be determined by measure- of the enzyme in the direction of ATP synthesis
ment of the absorbancy at 350 nm (523). can be demonstrated by trapping ATP in the
The reaction catalyzed by the enzyme is read- presence of hexokinase (401). Under appropri-
'ily reversible with an apparent equilibrium ate conditions, the rate of ATP formation by
constant (523): the enzyme of C. cylindrosporum is only eight-
fold lower than the rate of 10-formyl-THFA
K [formimino-THFA] [glycine] 0 32 synthesis (118). The enzymes of both clostridia
[THFA] [formiminoglycine] were purified, and the enzyme of C. cylindro-
Formiminoaspartate, formiminoglutamate, and sporum was obtained in crystalline form (407).
The enzymes of both bacteria are composed of

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


N-formylglycine are inactive as substrates,
but formiminoalanine and the methyl ester of four identical subunits (324, 469, 585) which
formiminoglycine were active in one of the as- reversibly dissociate at pH values below 7 (84,
says of the enzyme. Zn2+ and Fe2+ ions inhibit 585) or in the absence of monovalent cations
glycine formiminotransferase strongly (523). (324, 585) and irreversibly at pH values above
Formimino-THFA cyclodeaminase catalyzes 11 (585). The monomeric units are catalytically
a reversible (524) reaction in which formimino- inactive (324). The reassociation of the mono-
THFA is converted to (+)-5,10-methenyl-THFA mers is promoted by ATP, ADP, and to a less
and ammonia. The enzyme is present in both extent by AMP (324). One nucleotide binding
clostridia and was first demonstrated in these site is present per monomer. This binding site
organisms by Rabinowitz and Pricer (405). is not altered by dissociation of the tetramer.
Uyeda and Rabinowitz (524) purified the en- The intact folate binding sites are created by
zyme from C. cylindrosporum 40-fold, but it the association of the monomers and render the
still contained 5,10-methenyl-THFA cyclohy- enzyme catalytically active (120).
drolase activity. The two enzyme activities The catalytic mechanism of the enzyme was
could not be separated, which suggests that extensively studied by Buttlaire et al. (85),
they are associated with one protein molecule. Joyce and Himes (244, 245), and Rabinowitz
However, the formimino-THFA cyclodeami- and Himes (394, 399). The enzyme requires
nase in cell-free extracts of C. acidiurici is ac- Mg2+ ions or other bivalent cations for full ac-
companied by only small amounts of 5,10-meth- tivity, NH4+ ions or other monovalent ions for
enyl-THFA cyclohydrolase activity (524). tetramer formation and possibly for catalytic
(+)-5,10-Methenyl-THFA can be used in two activity, and a reducing agent, e.g., mercapto-
ways. It can be reduced to 5,10-methylene- ethanol (84, 120, 223, 406, 585). It can be used
THFA and, thus, provides the condensing sin- for quantitative determination of formic acid
gle-carbon moiety for the synthesis of serine (406).
from glycine, or it can be hydrolyzed to 10- (+)-5,10-Methylene-THFA dehydrogenase
formyl-THFA, subsequently yielding ATP for occurs in the extracts of both clostridia. The
cell growth. enzyme from C. cylindrosporum was purified
5,10-Methenyl-THFA cyclohydrolase cata- 80-fold and is completely specific for NADPH2
lyzes a reversible reaction that also proceeds (525) as most microbial 5,10-methylene-THFA
rather rapidly in the absence of the enzyme dehydrogenases, except that of C. formicoaceti-
under neutral and alkaline conditions (119, 394, cum, which is completely specific for NADH2.
412, 525). The equilibrium constant The enzyme of C. acidiurici is active with both
NADH2 and NADPH2 (346). Uyeda and Rabino-
K [10-formyl-THFA] [H+] witz (525) studied the reaction in the reverse di-
[5,10-methenyl-THFA] [H20] rection and demonstrated that the product
= 2.4 x 10-8 formed was 5,10-methenyl-THFA and not 10-
formyl-THFA. The enzymatic dehydrogenation
indicates that the extents of hydrolysis at equi- results in the formation of an equilibrium with
librium are 40, 93, and 97% at pH 5.7, 7.0, and an apparent equilibrium constant (525):
7.4, respectively (493).
10-Formyl-THFA is degraded to formate, K=
THFA, and ATP by formyl-THFA synthetase. [(+)-5,10-methenyl-THFA] [NADPH2]
This reaction constitutes one of the main [(+)-5,10-methylene-THFA] [NADP]
sources of ATP in the clostridia (118). Rabino- = 0.14
witz and Pricer (407) calculated that this en-
zyme constitutes approximately 2 and 3% of the ATP inhibits the enzyme of both clostridia (525)
dry weight of C. acidiurici and C. cylindro- and may play an important role in the regula-
VOL. 40, 19 76 DEGRADATION OF PURINES AND PYRIMIDINES 435

tory mechanism as suggested for the enzyme other by y-linkages of 1-glutamate (399, 525).
from a strain of Salmonella (123). Curthoys et al. (119) described a procedure to
Serine hydroxymethyltransferase catalyzes prepare (+) - 5,10 - methenyltetrahydropteroyl -
the conversion of glycine and (+)-5,10-methyl- triglutamate (,y-linkage) from C. acidiurici. A
ene-THFA to L-serine and (-)-THFA. The en- total of 25 mg of this stable derivative was
zyme is present in both clostridia, and its activ- obtained per 100 g of wet cells. Out of this
ity depends on the presence of catalytic compound they prepared the natural isomer of
amounts of pyridoxal phosphate (526). tetrahydropteroyltriglutamate and its 10-for-
Specificity of the THFA derivatives. Three myl and 5,10-methylene derivatives. Besides
aspects of the specificity of the THFA deriva- the natural tetrahydropteroyltriglutamate de-
tives (Fig. 14) will be discussed below: (i) the rivatives, a number of other tetrahydropter-
amount of glutamate residues present, (ii) the oylderivatives can be used by the enzymes of

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


amino acid present, and (iii) the optical speci- clostridia (Table 11). However, the Km values
ficity of the coenzyme. measured for the analogues differ substan-
The purine-fermenting clostridia contain un- tially. TheK. values of the naturally occurring
usually high concentrations of folate coen- tetrahydropteroyltriglutamate coenzymes are
zymes. The in vivo concentration of folate coen- considerably lower than those of tetrahydrop-
zymes in C. acidiurici was estimated to be 1 to 2 teroate coenzymes for glycine formiminotrans-
mM (119). Although Wright (606, 607) sepa- ferase (523) and methylene-THFA dehydrogen-
rated the folate derivatives from extracts of C. ase (525) and approximately 80-fold lower than
cylindrosporum into five groups containing dif- that of the THFA coenzyme for formyl-THFA
ferent amounts of glutamate and phosphate, synthetase (32, 118, 223). The Km value of the
more recent studies indicate that the coenzyme natural coenzyme is about half of that of THFA
in C. cylindrosporum and C. acidiurici occurs for serine hydroxymethyltransferase (526). In
exclusively in the form of triglutamate deriva- spite of these differences, the observed Vma,
tives of pteroic acid, presumably linked to each values of these enzymes were equal or only
twofold higher for the naturally occurring coen-
zyme as compared with the above-mentioned
OH H 0
analogues (223, 523, 525). These results suggest
N
C-N-
4" C -,NH-CH-COOH
1-1 _019 CH~~~~~~~~~~~~~~~~~~~~~~
1NC
that the additional glutamyl residues of the
N3 C 6 C.. natural coenzyme are not involved in the enzy-
matic reaction mechanism. The folate binding
H CIC NACH2
N COOH sites of the enzymes appear to contain noncata-
H2N N
H lytic subsites that bind the additional glutamyl
Tetrahydropteroate residues and increase the affinity for the folate
coenzymes significantly (118).
Tetrahydrofolate (THFAI
All folate derivatives contain L-glutamate,
FIG. 14. Structure of tetrahydropteroate and but two optical antipodes can be distinguished
THFA. on the basis of the configuration at the C6 atom

TABLE 11. Coenzyme specificity of the enzymes involved in the single carbon conversions in C.
cylindrosporum
Tetrahydro(TH)-folate analogue tested

Enzyme-ptteer- TH-folate
Enzyme TH-ptero- ~~~TH-Ptr oyl-tglu-
ydi-glutaoL-H THte-H-ter-
oylT-ae- Refer-
ence
oate
EnzymeaTHpter- THolae matet
l mate
~~~mateP
glua tamate partate

Glycine formiminotransfer- + + + + + + 523


ase
Formimino-THFA cyclo- + + + + + 524
deaminase
Methylene-THFA dehydro- + + + NDb ND 525
genase
Serine hydroxymethyltrans- + + + ND ND 526
ferase
Formyl-THFA synthetase + + +C + + 118, 223
a
y-linkage; the naturally occurring coenzyme of C. acidiurici and C. cylindrosporum.
b ND, Not determined.
c
TH-pteroyltri-a-glutamate is almost completely devoid of activity.
436 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
of the tetrahydrofolate derivatives. The opticalpyrophosphate, 6 mol of nonheme iron, and 3
rotation of the naturally occurring family of mol of acid-labile sulfur (527) and probably no
antipodes is given in Table 12. Only this familylipoic acid (65) per 240,000 daltons, the molecu-
of isomers is active in the enzyme systems lar mass of the enzyme (527). It does not require
(119). coenzymes of the B,2 group, as previously sup-
Degradation of serine. The studies described posed (395). CoA, 2-mercaptoethanol, and cer-
in this section were performed with C. acidi- tain transition state metal ions are required in
urici. As given in Table 10, acetate is also the C02-exchange activity of the enzyme from
produced, albeit in lower amounts, by C. cylin- C. acidiurici (410). Similar results were ob-
drosporum, but studies on the mechanism of
the acetate production in this organism are CH2OH
I CH3 + NH4

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


scarce.
The activities of the enzymes involved in the H-C-NH2 Fez+
*C=O
degradation of serine are considerably higher COOH L-Serine dehydratase Ie0
in crude extracts ofC. acidiurici than the over- COO
L -Serine
L iv
all rate of uric acid fermentation by whole cells, Pyruvate
which was estimated at 14 ,umol/h per mg of CO2 - + Enz-TPP
cells (443). Pyruvate is formed from L-serine by i~~~~~~~~~~~~
L-serine dehydratase (L-serine hydro-lyase Enz-TPP -2e1
A

[deaminating][EC 4.2.1.13]) (42, 46, 409, 445) I


(Fig. 15). The enzyme from C. acidiurici was -CH,-CO
purified 370-fold by Sagers and Carter (445). Oxidized Pyruvate
Fe2+ ions and sulfhydryl reagents are essential Ferredoxin synthase
for its activity (46, 445). The spectral properties Reduced

of the enzyme strongly indicate the presence of Ferredoxin Enz -TPP


pyridoxal phosphate in the enzyme (445). D- I
Serine is attacked only slowly, whereas L-threo-
nine and L-cysteine are not degraded (46, 409,
445).
Pyruvate synthase (pyruvate: ferredoxin oxi-
doreductase [EC 1.2.7.1]) catalyzes the revers-
ible production of acetyl-coenzyme A (CoA) and CH,- C -SCoA
CO2 from pyruvate. Part of this reaction is Acetyl -CoA
formed by an enzymatic exchange of the car- Phosphate tFe' Pi
boxylate group of pyruvate with CO2, which acetyltransferase 0
can be measured separately by use of labeled
compounds (395, 441). CH3-C O-PO) Acetate krnase CH3COO0
The enzyme from C. acidiurici was purified Acetyiphosphate Acetate
50-fold by Raeburn and Rabinowitz (410). It AOP ATP
appears to contain 0.5 to 0.8 mol of thiamine FIG. 15. Degradation of L-serine by C. acidiurici.
TABLE 12. Specific optical rotations ([aID) of tetrahydrofolic acid (THFA) derivatives
Natural isomer Racemic mixture"
Compounda Reference Reference
[a]D Temp (C) [aID Temp (C)
THFAc -16.9 27 57 +14.9 27 57
5,10-Methenyl-THFA +46 + 3 23 57 -36 23 57
+68 23 57
5,10-Methylene-THFA +163 + 11 23.5 412
+142.5 611
+159 9 27 56
10-Formyl-THFA -32 ± 3 23 57 -9 23 57
-42 23 57
a The natural isomer of formimino-THFA is levorotatory.
b With respect to the C6 atom.
e Measured in 0.1 N NaOH.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 437
taimed for the enzyme from C. formicoaceticum COOHNH2
(13). H2C
The reaction mechanism has been studied by CONH + Pi - PyP -CHO
Sagers et al. (443) and Uyeda and Rabinowitz
(528). Thauer et al. (507) demonstrated that
CO2 rather than HCO3- (or H2CO3) is the active
species of "C02" utilized by the enzyme from
Clostridium pasteurianum. The natural elec-
tron acceptor is ferredoxin (529, 532), but FAD,
2,3,5-triphenyltetrazolium, and methyl violo-
gen may also be used by the enzyme of C.
acidiurici (410). P3 (red)

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Phosphate acetyltransferase (acetyl-CoA:
orthophosphate acetyltransferase [EC 2.3.1.8])
transfers reversibly acetyl units between CoA PN (oxIa)
and phosphate. The enzyme from C. acidiurici
requires a divalent metal, e.g., Fe2+ (444). NAD NADH2
Arsenate failed to uncouple the high-energy
acyl compounds, as was observed for a number
of other phosphotransacetylase systems. This
may indicate a high degree of specificity giving
preference to phosphate over arsenate. Rela- OSH
tively high concentrations of arsenate inhibit P2S
both the degradation of pyruvate and the for- SH
mation of acetyl phosphate (444). N: CH2
Acetate kinase (ATP:acetate phosphotrans- HC CH2N"N
ferase [EC 2.7.2.1]) of C. acidiurici was purified NH3+ I
eightfold by Sagers et al. (443). L7H2
Glycine synthase system. Results of tracer (+)- 5,10- Methytene -THFA.
studies with C. acidiurici indicate that a sub- FIG. 16. Reactions catalyzed by the glycine syn-
stantial amount of the methyl group of acetate thase system from Peptococcus glycinophilus (70),
is derived from the C5 urate atom and from the rat liver mitochondria (353), Arthrobacter globifor-
methylene group of glycine (Fig. 11) (259, 397). mis (279), and probably also C. acidiurici (446). P1,
Sagers and Gunsalus (446) demonstrated that P2, P3, and P4 are four proteins involved in the reac-
the mixing of carbons during acetate genera- tion (see text). P2S2 and P2(SH)2 represent the oxi-
tion results from a cleavage reaction of glycine dized and reduced form ofP2. P1-PyP-CHO represents
to one-carbon units in this organism. This protein P1 with its pyridoxal phosphate prosthetic
cleavage was shown to be dependent on the group.
presence of THFA and benzyl viologen. The was later found in rat liver mitochondria (279,
methylene group of glycine is incorporated into 351-353, 452, 453), in Arthrobacter globiformis
5,10-methylene-THFA. This cleavage allows (240, 279), and in Escherichia coli (383).
the conversion of glycine to acetate, C02, and The reversible (272, 452) cleavage of glycine
ammonia in the presence of an added electron (Fig. 16) involves four proteins among which
acceptor (446). are a pyridoxal phosphate-containing enzyme,
The reaction is catalyzed by the glycine syn- P1 (270, 271, 351), and a colorless heat-stable
thase system (5,10-methylene-THFA:ammonia protein, P2 (270, 271), which is also called H
hydroxymethyltransferase [carboxylating, re- (hydrogen carrier)-protein (279). Both proteins
ducing] [EC 2.1.2.10]). The glycine synthase are involved in glycine decarboxylation and in
system is also present in Peptococcus glycino- the exchange of bicarbonate with the carboxyl
philus, which converts glycine to acetate (35, group of glycine. P2 is of low molecular weight
98) according to the equation: 4 NH2CH2COOH and contains one functional disulfide group per
+ 2H20 -* 3 CH3COOH + 4 NH3+ 2 C02. The molecule (279), which is part of a lipoic acid
overall process involves, besides the glycine molecule (279, 425). As a result of these reac-
synthase system, also a conversion of C02 into tions, a complex is formed between P2 and the
acetate (35), but Schulman et al. (466) found product of glycine decarboxylation, which
only a small labeling of acetate by C02. should be in the -CH2NH2 form. This complex
Extensive studies have been made on the was isolated by Kochi and Kikuchi using
glycine synthase system of P. glycinophilus purified preparations of proteins P1 and P2
(22, 270-273, 447). A similar enzyme system from A. globiformis (279).
438 VOGELS AND VAN DER DRIFT BACTERPIOL. REzV.
Transfer of the glycine a-carbon to THFA to Fe2+ ions, tungstate, and selenite (13). Formate
form 5,10-methylene-THFA and production of dehydrogenase is probably a molybdoenzyme
ammonia are catalyzed by protein P4, which is (504, 505).
also called T (catalyzing THFA-dependent step) Reduction of CO2. Tracer studies performed
protein (279, 353). Electron transfer to NAD with C. acidiurici and C. cylindrosporum re-
(but not to NADP) or to a dye with a low vealed that C02 is reduced by these organisms
oxidation-reduction potential is catalyzed by an and incorporated in the final products (Fig. 11).
FAD-containing enzyme P3 (22, 272), which is This is also evident from the fact that more
also called L protein (279, 353) because it ex- than 1 mol of acetate is formed per mol of
hibits lipoamide dehydrogenase activity (22, hypoxanthine by C. acidiurici (259). In fermen-
272, 279, 353). The above-mentioned complex tation experiments performed by Schulman
between P2 and the -CH2NH2 group is also and co-workers (466), C. acidiurici yielded 183

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


formed on incubation of 5,10-methylene-THFA, mol of acetate, 49 mol of butyrate, and 55 mol of
NH4Cl, P3-protein, P4-protein in the presence of formate from 100 mol of hypoxanthine and C.
some reducing system such as lipoamide dehy- cylindrosporum yielded 168 mol of acetate, 29
drogenase plus NADH2, or dithiothreitol (279). mol of butyrate, and 21 mol of formate. Thir-
Throughout the whole glycine cleavage proc- teen and 11% of acetate and 23 and 65% of
ess, the hydrogen atoms at the a-position are formate produced by these organisms, respec-
not labilized (279). tively, were derived from C02 that was present
Formate dehydrogenase. Formate dehydro- in large excess during the fermentation. Stud-
genase from C. acidiurici was purified 30-fold ies with 13C02 and 14C02 performed with C.
by Kearny and Sagers (261). The purified en- acidiurici revealed that 18.7% of the methyl
zyme catalyzes the following reaction: groups and 9.3% of the carboxyl groups of ace-
tate were derived from C02 and that 9.1% of the
HCOO- + H20 + X a HC03- + XH2 molecules were labeled in both the methyl and
(formate) (bicarbonate) the carboxyl group and 9.5% only in one group.
Schulman et al. (466) assumed that C02 is
in whichX represents a suitable electron accep- reduced to formate, which in turn is reduced to
tor. The purified enzyme may use benzyl violo- 5,10-methylene-THFA by a reaction sequence
gen in the forward reaction but not other elec- depicted in Fig. 13. They proposed that this
tron acceptors such as tetrazolium dyes, ferricy- compound is further reduced to 5-methyl-THFA
anide, dichloroindophenol, and methylene blue and methyl corrinoid in C. thermoaceticum
(261). (315). In this reaction sequence 5,10-methenyl-
In crude extracts of C. acidiurici, ferredoxin THFA and 5,10-methylene-THFA are common
is involved in the transfer of electrons from to the pathway of purine degradation and the
formate to NAD (69, 261, 529) and perhaps also pathway of C02 reduction. Thus, any methyl
to NADP (261). NAD is not reduced by the group of acetate that was derived from 13C02
purified enzyme in the presence of ferredoxin and that passed through the common interme-
and formate (261), probably due to the absence diates would have its '3C concentration diluted
of NADH2:ferredoxin oxidoreductase. by unlabeled carbon from the purine, and any
The enzymes of C. acidiurici (261) and C. acetate totally synthesized from C02 would in-
pasteurianum (506) also catalyze an isotopic volve dilution of the 13C in the methyl group of
exchange between C02 and formate in the ab- acetate. However, the methyl group of acetate
sence of ferredoxin. This exchange was also is labeled almost twofold more than the car-
observed with whole cells of C. acidiurici (442). boxyl group. Schulman et al. (466) assumed
The reverse formate dehydrogenation reac- that this result may be explained by compart-
tion, which expresses the C02-reductase activ- mentalization of the two pools of 5,10-methyl-
ity of the enzyme, does not proceed when re- ene-THFA and/or 5,10-methenyl-THFA either
duced benzyl viologen is used as the electron by physical separation of the pathways or by
donor (261). NADH2 and reduced ferredoxin are use of different forms of THFA coenzymes.
required for formate synthesis from C02 in C. Later studies (465) demonstrated that in C.
acidiurici (503). In Clostridium thermoaceti- thermoaceticum the carboxyl of acetate was
cum the reduction can be achieved by NADPH2 mainly derived from the carboxyl of pyruvate.
(502) or reduced methyl viologen (14). The for- If this reaction is also valid for C. acidiurici,
mate dehydrogenase of C. acidiurici (261) and this could explain the extra dilution of this
C. pasteurianum (506) is rapidly inactivated by group during the degradation of purines in the
molecular oxygen, and light inhibits its activity presence of labeled C02. However, such an ex-
(261). The production of the enzyme in growing planation is hardly tenable if one takes into
cells of C. formicoaceticum is stimulated by consideration the exchange of the carboxylate
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 439
group of pyruvate with CO2, which is catalyzed Electron donating Coenzyme Electron requiring
by pyruvate synthase. reactions reactions
Schulman and co-workers overlooked an- Xanthine dehydrogenase Xanthine dehydrogenase
other explanation of their results which does (purine, 8 -hydroxypurine uric acid I
not involve the proposed (but not yet demon- hypoxanthine )
strated) participation of methyl-THFA and Formate dehydrogenase Formate dehydrogenase
methyl corrinoid intermediates. A total synthe- IHCOOH- CO2) * Ferredoxin 1C02- reduction)
sis of acetate from C02 can be explained on the
basis of a combination of the following proc- Pyruvate synthase / t
esses: (i) synthesis of 5,10-methylene-THFA Glycine synthase \A' Glycine synthase
from C02, (ii) synthesis of glycine by a glycine (cleaving glycine)I (CO2 reduction to acetate)
synthase system, (iii) synthesis of serine from

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


NAOH2: terredoxin
glycine and 5,10-methylene-THFA, (iv) conver- oxidoreductase |,
sion of serine to acetate. The occurrence of
these reactions, which have been demonstrated VC. acidirc
in C. acidiurici, may also explain why the car- NAD
boxyl group of acetate that originates via a
longer bypass from C02 is less labeled than the
methyl group. Glycine which is produced from |Transhydrogenase j Methylene -THFA
t dehydrogenase
the purines will introduce an additional dilu-
tion of the labeled C atom which finally delivers NADP
the carboxyl group of acetate. Cl. acidiurici
Ferredoxin. The role of ferredoxin in the CI. cylindrosporum
metabolism of C. acidiurici, C. cylindrospo- FIG. 17. Role of ferredoxin, NAD, and NADP in
rum, and other organisms has been reviewed the degradation of purines by C. acidiurici and C.
by Valentine (529). The ferredoxins ofC. acidi- cylindrosporum. The coenzyme involved in the gly-
urici and C. cylindrosporum were crystallized cine synthase reaction is not unequivocally estab-
by Lovenberg et al. (317). They differ as to the lished for the clostridia.
crystal form, the absorption at 280 to 300 nm,
and the amino acid composition (317). More- doxin oxidoreductase, which was first found in
over, the ferredoxin of C. cylindrosporum was C. acidiurici. In C. pasteurianum ferredoxin
unstable after diethylaminoethyl-cellulose pu- interacted specifically with NADP, no NAD
rification (67, 317) and therefore is used less in being reduced under any of the experimental
metabolic studies. Both ferredoxins contain 5 conditions (529).
mol of iron and 4 mol of inorganic sulfide per Quantitative aspects. C. cylindrosporum
mol of protein (317). forms relatively high amounts of formate;
Ferredoxin is generally found in hydrogen- about 1 mol of this compound is formed per mol
evolving species, but C. acidiurici is an inter- of xanthine or guanine degraded (Table 10).
esting exception to this rule. It possesses a high These results indicate that this organism con-
content of ferredoxin (81) but does not produce verts the C8 urate atom preferentially to for-
H2 as a fermentation product (29) because of the mate, which is not further degraded to C02.
low level or absence of hydrogenase in this Under these conditions glycine degradation to
organism (529, 532). acetate is hampered and the former compound
Ferredoxin is the primary electron acceptor accumulates (Fig. 18). Cell suspensions of this
and donor in the main redox reactions of the organism cannot convert formiminoglycine be-
purine degradation (Fig. 17). The coupling of yond the glycine level.
some of the reactions involving ferredoxin was In contrast to C. cylindrosporum, C. acidi-
shown by Valentine et al. (532), who demon- urici converts formiminoglycine readily to ace-
strated the ferredoxin-coupled pyruvate oxida- tate (408). Probably due to the low activity of
tion and urate reduction for C. acidiurici. The methenyl-THFA cyclohydrolase (524), meth-
same could be demonstrated for C. cylindrospo- enyl-THFA can be used as an electron acceptor
rum by Bradshaw and Reeder (67), who used for the pyruvate synthase reaction, and about 1
the ferredoxin of C. acidiurici instead of the mol of acetate is formed per mol of formimino-
less stable ferredoxin from C. cylindrosporum. glycine degraded (Table 10). Accordingly, for-
The primary electron acceptor of the glycine mate is only a minor product of uric acid fer-
cleavage system has still to be determined. mentation in C. acidiurici, and tracer studies
Except for methylene-THFA, NAD and (259, 400) have shown that the C8 atom of the
NADP are formed in secondary reactions. A purine molecule is incorporated predominantly
key enzyme in these reactions is NADH2:ferre- into the methyl carbon of acetate in this orga-
440 VOGELS AND VAN DER DRIFr BACTERIOL. REV.
Xanthine C Inhibition reduction of half of the amount of methylene-
Cl. acidiurici THFA to glycine (Fig. 16), synthesis of serine
---- C cylindrosporum and conversion of serine to acetate (Fig. 15). As
Formiminoglycine a result of these processes the amount of ATP
formed during the degradation is reduced by
//"i 25%. The shift in the balance of urate fermenta-
/'THFA ?
Formimino-THFA tion as compared with xanthine fermentation
Zf 1@ sX THFA can be explained by a process in which one-
quarter of the amount of glycine formed is con-
Gtycine * - Methylene-THFA n. Methenyt-THFA .- Formate verted to methylene-THFA. As a consequence,
half of the amount of methenyl-THFA formed is
superfluous to serine synthesis and can be con-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


verted to CO2. Thus, the reduction equivalents
Serine -------------
NAO/NAOP needed to reduce uric acid are produced and
1.25 mol of ATP are formed instead of 1 mol in
Pyruvate the case of xanthine. The higher yield of energy
-. - ;9 Ferredoxin------------------------------ may explain why uric acid is a better substrate
ATP
to C. acidiurici than xanthine and hypoxan-
thine (32).
Acetate Urate Hypoxanthine Regulatory aspects of the coenzymes. Formi-
FIG. 18. Main routes involved in purine degrada- mino-THFA cyclodeaminase is strongly in-
tion by C. acidiurici (solid lines) and C. cylindrospo- hibited by THFA and some other folate deriva-
rum (dashed lines). The reduction equivalents tives (524). The low Ki value (4.10-6 M) of
needed in urate utilization or produced in the degra- THFA and the high in vivo concentration of
dation of hypoxanthine are compensated by the inter- folate coenzymes in C. acidiurici, which was
conversions indicated by dotted lines. estimated to be 1 to 2 mM (119), suggest that
formimino-THFA cyclodeaminase is under
nism. Therefore, it is unlikely that the 10-for- stringent control in vivo (Fig. 18). Perhaps
myl-THFA synthetase reaction is a major en- THFA also controls the glycine synthase sys-
ergy-yielding step in this organism, even tem in the clostridia, since the exchange reac-
though the enzyme is present in very high tion catalyzed by purified preparations of the P1
amounts in the cells (407, 443). ATP is produced and P2 proteins of A. globiformis are markedly
mainly by the acetate reaction in C. acidiurici inhibited by THFA (279). The regulatory as-
and by the formyl-THFA synthetase pathway pects of the coenzymes involved in the reduc-
in C. cylindrosporum (Fig. 18). tion and oxidation processes, i.e., NAD, NADP,
If the cells grow on substrates that are more and ferredoxin, should be examined further.
reduced (hypoxanthine, 8-hydroxypurine, and Ferredoxin plays a major role, but one crucial
purine) or more oxidized (uric acid) than xan- reaction catalyzed by methylene-THFA dehy-
thine, the fermentation balance shifts by the drogenase uses NAD or NADP (Fig. 17).
involvement of other electron-requiring or -do-
nating reactions, respectively (Fig. 17). C. aci- Peptococcus aerogenes
diurici forms 125 mol of acetate and 250 mol of Whiteley (587) demonstrated that cell sus-
CO2 per 100 mol of hypoxanthine and 75 mol of pensions of Peptococcus aerogenes (Micrococcus
acetate and 350 mol of CO2 per 100 mol of urate aerogenes) which had been grown in a rich
(259). The shift in the balance in the case of medium without added purines or pyrimidines
hypoxanthine fermentation as compared with convert guanine, adenine, xanthine, hypoxan-
xanthine fermentation may be explained by the thine, thymine, uracil, and cytosine to ammo-
following reactions: nia, H2, CO2, acetic acid, and lactic acid. Thy-
hypoxanthine + X mine and uracil accumulate during the degra-
6,8-dihydroxypurine + XH2 (1) dation of purines. Uric acid and allantoin are
not degraded. Two pathways are perhaps in-
6,8-dihydroxypurine xanthine (2) volved, the greater portion being decomposed to
--

0,5 C02 + 0,25 ATP + XH2 yield an imidazole-containing compound and a


0,25 acetate + 0,25 ADP + 0,25 Pi (3) small portion of the substrate undergoing
cleavage to yield pyrimidines.
in which X is NAD, NADP, and/or ferredoxin. Later studies revealed the presence of xan-
Reaction (3) represents the net result of the thine dehydrogenase and 2-oxypurine dehydro-
following processes: reduction of C02 to methyl- genase in this organism (605). In analogy to
ene-THFA (reactions 3, 5, 6, and 7, Fig. 13), C. cylindrosporum and Veillonella alcalescens,
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 441
the specific activity of '0N-formyl-THFA syn- dehydrogenase-hydrogenase system, the con-
thetase is high in P. aerogenes (590). The path- version of hypoxanthine to xanthine and the
ways ofpurine degradation remain to be solved. decomposition of hypoxanthine are inhibited in
an atmosphere of H2. Uric acid is reduced under
VeilloneUa alcalescens such conditions (588, 589). The hydrogenase-
V. alcalescens (Micrococcus lactilyticus), an ferredoxin system is involved in H2 production
anaerobic bacterium indigenous to saliva, de- from pyruvate and a-ketoglutarate (529, 540,
composes certain purines. Adenine and gua- 591) and the reduction of pyruvate, fumarate,
nine are slowly deaminated. Xanthine and hy- and NADP, but not of NAD, by molecular H2
poxanthine are poor substrates for growth of (591).
certain strains of V. alcalescens, and other pu- Extracts of V. alcalescens incubated in an
rines are not substrates (588). Growth of V. atmosphere of H2 reduce the following com-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


parvula is stimulated by hypoxanthine (427). pounds (listed in order of decreasing activity):
The products formed by cell suspensions of V. 2,4-dinitrophenol (2,4-diaminophenol), selenite
alcalescens from hypoxanthine, xanthine, and (Se), metabisulfite (S2032-), hydrosulfite, hy-
uric acid are given in Table 13. The possible droxylamine (NH3), teflurite (Te), arsenate (ar-
intermediary state of glycine and uracil was senite), nitrate (NH3), and nitrite (NH3). The
considered by Whiteley and Douglas (588), but reduced products are placed in parentheses.
no ammonia is formed from uracil; other pyrim- The presence of carrier amounts of benzyl violo-
idines or amino acids are not attacked. Up to gen stimulates most of these reductions (604).
this moment the reaction pathway remains un- The reduction of arsenate by H2 is catalyzed by
known. A point of interest is formed by the the enzymes hydrogenase and arsenate reduc-
capability of one of the tested strains to hydro- tase. The overall reaction is markedly stimu-
lyze allantoin completely to urea and an uni- lated by K+ or NH4+ but not by Na+ and is
dentified compound, which may be glyoxylic inhibited by phosphate and arsenite. Arsenate
acid (588). reduction can be coupled to hypoxanthine oxi-
Xanthine dehydrogenase of V. alcalescens dation (604).
was extensively studied by Smith et al. (482)
and is discussed in a previous section. A novel DEGRADATION OF PYRIMIDINES
electron carrier was postulated to be involved Until the early fifties, the only reported deg-
in this reaction by Whiteley and Ordal (589) radation route of pyrimidines was that pro-
and was identified as ferredoxin by Valentine posed by Cerecedo for mammals (102). This
et al. (534). Ferredoxin is also involved in the pathway involves isobarbituric acid, formylox-
pyruvate synthase reaction (529, 540, 591), the aluric acid, and oxaluric acid as intermediates
conversion of a-ketoglutarate to propionyl but has not been substantiated by later studies.
phosphate (540), and a large number of other The main routes involved in pyrimidine deg-
reductions (540, 591). V. alcalescens contains radation by microorganisms were established
hydrogenase which catalyzes also the produc- in 1951 to 1954 by studies of Lara (300, 301) with
tion of hydrogen from hypoxanthine (Table 13). Nocardia corallina, by Lieberman and Korn-
As a result of the reversibility of the xanthine berg (310-312) with Clostridium oroticum, by
Hayaishi and Kornberg (214, 216) with strains
TABLE 13. Fermentation products of Veillonella of Corynebacterium and Mycobacterium, by
alcalescens strain 416 (588) Wang and Lampen (578-580) with a gram-posi-
tive, motile nonsporulating rod, and by Di
Substrate (10 Amol/reaction vessel) Carlo et al. (135, 137) with yeasts. These studies
incubated in N! atmosphere with
cell suspensions: provided the basis for elucidation of the degra-
Product dative route of pyrimidines in animals and the
Hypoxan- synthetic route of pyrimidines by bacteria.
thine Xanthine Uric acid
(Amoll (JAMOI/ (,umol/ Fink et al. (156, 157) described a reductive
vessel) vessel) vessel) degradation of pyrimidines in rat liver. In es-
CO2 19.81 15.4 6.00 sence, this route resembles that used by certain
H2 8.01 0.72 1.03 bacteria (Fig. 19). Uracil and thymine are re-
Acetic acid 7.20 3.67 0 duced to the dihydroderivatives, which in turn
Propionic acid 2.00 1.14 0 are hydrolyzed to carbamoyl-,/-alanine and car-
Ammonia 20.83 18.80 2.61 bamoyl-,¢aminoisobutyric acid, respectively.
Urea 9.81 1.08 Dihydropyrimidinase (5,6-dihydropyrimidine
Uric acid 4.75 9.0 amido-hydrolase [EC. 3.5.2.2]) of calf liver cata-
Xanthine 0 0 0 lyzes not only the hydrolysis of dihydrouracil
Hypoxanthine 0 0 0 and dihydrothymine but also that of hydantoin
442 VOGELS AND VAN DER DRIFT BACTECRIOL. REV.
NH2 to N-carbamoyl glycine (577), of R(-)-5-phenyl-
hydantoin to R( - )-2-phenylhydantoic acid
N NCoC CH
(144), and of various R-amino acid hydantoins
11 (100). The reductive degradation route of py-
U CL ~ CH
rimidines is also operative in plants (521).
N The pyrimidine synthetic pathway was estab-
H lished by Yates and Pardee (610) and is pre-
Cytosine sented in Fig. 20. A number of reactions resem-
kH20 ble the route (Fig. 21) of orotate degradation by
NH34 1) C. oroticum (310-313).
0 REDUCTIVE PATHWAY

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


0

A relatively small number of microorga-


HN CH HN C-CH3 nisms (Table 14) is known to be able to de-
11 I 11 grade pyrimidines along a pathway involving
U=CL CH 0=C C the reduction of either uracil or thymine. This
N N pathway is depicted in Fig. 19 and will be
H H called the reductive pathway.
Uracil Thymine Neurospora crassa utilizes uracil, and to a
2H 2H less extent, dihydrouracil and N-carbamoyl-,B-
( 2) (2 b) alanine as a source of nitrogen. Studies with
mutant strains and identification of the degra-
0 0
dation products revealed that the reductive
pathway is operative in this organism (602).
HN CH2 Thymine is oxidatively demethylated to uracil
I
NHC I
CH in N. crassa in a pathway involving 5-hydroxy-
methyluracil, 5-formyluracil, and uracil-5-car-
N N boxylic acid as intermediates (2, 473).
H H Di Carlo et al. (137) found that Candida
Dihydrouracil Oihydrothymine utilis grows well on cytosine and uracil as a
H20 13) H20 (3) nitrogen source but not on thymine, whereas
Saccharomyces cerevisiae grows moderately
H2N COOH H2N COOH well on cytosine, but other pyrimidines are not
I used.
O=C CH2 O=C CH-CH3 The presence of cytosine deaminase (cytosine
HN CH2
- HN -
CH2 aminohydrolase [EC 3.5.4.1]) in S. cerevisiae
N- Carbamoyl-3-aminoiso- was shown 50 years ago (209), and the enzyme
N-Carbamoyl--alanine
butyric acid
also degrades 5-methylcytosine to thymine
(288). Di Carlo et al. (135) suggested, on the
(3-Ureidopropionic acid) (13-Ureidoisobutyric acid)
Aspartate (1) (2)
H20 (4) H20 (4?) +

Carbamoyl phosphate
~ N-Carbamoyl aspartate .
L-Oihydroorotate

NH3 COOH NH3 COOH Uridine-5- (5) Orotidine-5-


+ +
phophate *- phosphate Orotate
CO2 CH2 CO2 CH-CH3 FIG. 20. Pyrimidine biosynthetic pathway (610).
+
H2N-CH2
+

H2N-CH2 (1) Aspartate carbamoyltransferase (carbamoylphos-


phate:L-aspartate carbamoyltransferase [EC
13-Alanine r3-Aminoisobutyric acid 2.1.3.2]); (2) dihydro-orotase (L-5,6-dihydro-orotate
FIG. 19. Reductive pathway ofpyrimidine degra- amidohydrolase [EC 3.5.2.3D); (3) dihydro-orotate
dation. (1) Cytosine deaminase (cytosine aminohy- oxidase (L-5,6-dihydro-orotate:oxygen oxidoreduc-
drolase [EC 3.5.4 1]. Acts also on 5-methylcytosine. tase [EC 1.3.3.1]); (4) orotate phosphoribosyltrans-
(2a) Dihydrouracil dehydrogenase (5,6-dihydroura- [erase (orotidine-5'-phosphate: pyrophosphate phos-
cil:NAD+ oxidoreductase [EC 1.3.11]). Active in C. phoribosyltransferase [EC 2.4.210]); (5) orotidine-
uracilicum (93). (2b) Dihydrouracil dehydrogena4e 5'-phosphate decarboxylase (orotidine-5'-phosphate
(NADP+) (5,6-dihydrouracil: NADP+ oxidoreduc- carboxy-lyase [EC 41.1 231).
tase [EC 1.3.121). Acts also on dihydrothymine. (3)
Dihydropyrimidinase (5,6-dihydropyrimidine ami- Ureidopropionase (N-carbamoyl-/3-alanine amido-
dohydrolase [EC 3.5221). Acts also on 5,6-dihydro- hydrolase [EC 3.5.1.6D). The animal enzyme acts
uracil, 5,6-dihydrothymine, and hydantoin. (4) A- also on N-carbamoyl-3-aminoisobutyric acid.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 443
TABLE 14. Microorganisms known to degrade pyrimidines along a reductive pathway
Pyrimidine tested Microorganism Reference
Uracil Chlorella fusca 278
Uracil Neurospora crassa 602
Uracil, cytosine Candida utilis 135, 379
Uracil, cytosine, thymine Various yeast species 302
Uracil Pseudomonas aeruginosa 156
Uracil, cytosine, thymine P. facilis (Hydrogenomonas facilis) 285, 286
Uracil, cytosine Mycobacterium speciesa 428
Thymine Nocardia rubraa 428
Uracil Clostridium uracilicum 91-95
Uracil, cytosine, thymine C. sporogenesb 222

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Uracil C. botulinum speciesb 222
Orotic acid C. oroticum 310-313
Orotic acid Corynebacterium species 424
a Evidence for the reductive pathway based only on the inability of the organisms to degrade barbiturate.
b The pyrimidines are reduced to the dihydroderivatives, but no evidence is available for further
degradation of the latter.

basis ofgrowth experiments, that dihydrouracil smegmatis and M. chelonei (M. borstelense)
and dihydroorotic acid are intermediates in the but in no case from barbituric acid (428). Proba-
degradation of uracil to urea by C. utilis, but bly, the reductive pathway is operative in these
later studies demonstrated that the reductive organisms. Ammonia is formed from thymine
pathway is followed (379). Also the product of by Nocardia rubra and by one strain of N.
this route, (8-alanine, is used as a nitrogen brasiliensis. The latter strain attacks uracil,
source by this organism (379). too. Other Mycobacterium and Nocardia spe-
The ability to use cytosine and uracil as ni- cies tested were inactive in the production of
trogen sources is widely distributed among ammonia from pyrimidines (428).
yeast strains, but only a few yeasts degrade Clostridium uracilicum was isolated by
thymine (302). The degradations proceed ac- Campbell (91) from an enrichment medium
cording to the reactions given in Fig. 19 (302). containing uracil and yeast extract. Uracil is
Pseudomonas aeruginosa accumulates dihy- readily degraded, but it does not stimulate
drouracil and N-carbamoyl-,&alanine when growth in a chemically defined medium, proba-
grown on uracil (156). bly due to the inability of the organism to de-
P. facilis (Hydrogenomonas facilis) grows at grade ,B-alanine (91). The enzymes involved in
the expense of cytosine, uracil, thymine, 5- the reductive pathway are induced by the re-
methylcytosine, orotic acid, and j3alanine as spective substrates (92). Hilton et al. (222)
nitrogen sources, but barbituric acid is not usedtested a large number of Clostridium species
(286). P. facilis can use the carbon skeleton offor the ability to metabolize uracil. Only C.
pyrimidines since it is able to degrade ,B-ala- sporogenes and the proteolytic strains of C.
nine (286). Besides C. utilis andP. facilis, nonebotulinum types A and B convert uracil to dihy-
of the microorganisms that degrade pyrimi- drouracil by an inducible dihydrouracil dehy-
dines along the reductive pathway is known to drogenase, but growth of the organisms was not
use the carbon skeleton, probably due to the stimulated by uracil. Washed cells incubated in
inability to degrade (3-alanine. This compound an H2 atmosphere reduce uracil, 5-aminouracil,
can be degraded and used as sole organic sub- thymine, and isobarbituric acid to the corre-
strate by P. aeruginosa (546). (3Alanine trans- sponding dihydropyrimidines and cytosine to
aninase of P. fluorescens (217) and Clostrid- dihydrouracil (222). Thus, these cells contain
ium propionicum (189) converts ,&alanine into several dihydropyrimidine dehydrogenases or a
malonaldehydic acid, which may yield acetyl rather aspecific dihydrouracil dehydrogenase.
CoA. The enzymes involved in the pyrimidine The dehydrogenases from P. facilis (285) and
degradation byP. facilis are induced by growth from animal liver reduce thymine, too. The
on uracil, but cytosine deaminase and the enzy- enzymes ofC. sporogenes (222), P. facilis (285),
matic system that degrades (3alanine are also and plasma membranes of animal liver cells
present in cells grown in the presence of ammo- (480) are specific for NADP, but an NAD-de-
nium chloride (286). pendent enzyme is present predominantly in
Ammonia is formed from cytosine by Myco- the mitochondria of liver cells (480). In contrast
bacterium smegmatis, M. vaccae, M. fortui- to these results, dihydrouracil dehydrogenase
tum, and M. diernhoferi and from uracil by M. of C. uracilicum is specific for NAD (92), and
444 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
the 27-fold purified enzyme does not react with 0
1,
other pyrimidines (93).
Dihydropyrimidinase ofP. facilis hydrolyzes HN CH
dihydrouracil and dihydrothymine (285). The 11
°C .C - COH 02 Methylene blue
enzyme from C. uracilicum needs Mg2+ or Mn2+ N 4 , Cytochrome c
ions for activity and does not act on dihydrothy- Orotic acid
mine (94). FHz NAO
,8-Ureidopropionase of C. uracilicum was Orotate / /
purified 100-fold and catalyzes a reaction that is reductase
essentially irreversible. Carbamoyl phosphate
was not found as an intermediate (95).
Conclusively, it may be stated that the reduc- HN
HN CH2
FHZ NADH

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


tive pathway appears to be unattractive to most O=C
I I
C
microorganisms. Some of them use the availa- N H COOH
ble nitrogen at the expense of one equivalent of HH
NADPH2 and leave the carbon skeleton as 38- S(Ls)-5,6-Dihydroorotic acid
alanine for other microorganisms. Clostridium H20 4 Dihydroorotase
species are provided with an additional electron
acceptor. Only P. facilis and perhaps C. utilis,
ffiCOOH s~~~~~~~~CH2-COOH
too, also utilize the carbon atoms, but probably H2N CH2 H20 HN C
no energy is furnished by the degradation to I I - I i
0=C .C Carboxymethyl- ° C .NC- O
the level of 8-alanine. N COOH hydantoinase H
H H
Degradation of Orotic Acid N-Carbamoyl-S(Ls)-aspartic acid S(Ls)-5-Carboxymethyl-
Orotic acid was isolated in 1905 by Biscaro H20 Ureidosuccinase hydantoin
and Belloni (54), and its structure was deter-
mined by Bachstez (21) in 1930. Two strains of NH3
+
COOH
U
the genus Mycobacterium isolated from soil C02 CH2
grow on synthetic media containing orotic acid + I
as the sole source of nitrogen (562). Orotic acid H2N bCOOH
is converted to uracil, which in turn is degraded
along the oxidative pathway that will be dis- SfLs)-Aspartic acid
cussed below. In all other known instances or- FIG. 21. Degradation of orotic acid by Clostrid-
otic acid is degraded along a reductive path- ium oroticum (177,310-313). F and FH2 represent
way, either by conversion to uracil and dihy- the oxidized and reduced flavin prosthetic group,
drouracil, as suggested for P. facilis (285), or by respectively.
a separate route, as reported for C. oroticum
(Zymobacterium oroticum) (310-313) and two flavin. The enzyme catalyzes: (i) the oxidation
unidentified corynebacteria (424). of NADH2 by orotate or oxygen (177); (ii) the
Kornberg isolated from bay mud a bacterium oxidation of dihydro-orotate by NAD or oxygen
capable of growing anaerobically in media con- (58, 177); (iii) a diaphorase reaction catalyzing
taining orotic acid as the main organic sub- the anaerobic reduction of methylene blue
strate. It was characterized as Z. oroticum by (177), cytochrome c (411), or other electron ac-
Wachsman and Barker (575) and renamed C. ceptors (5) by NADH2. The enzyme is rather
oroticum (99) since it produces heat-resistant specific for NAD, since the reaction rate with
spores. NADP was less than 2% of.that observed with
The first enzyme involved in the degradative NAD (311); 5-fluoro-orotate (177) and 5-bromo-
pathway (Fig. 21), orotate reductase (L- orotate (5) can substitute orotate in the reaction
5,6-dihydro-orotate:NAD+ oxidoreductase [EC and are even more active substrates. The inter-
1.3.1.14]), was recognized as a flavoprotein by action of the enzyme with orotate or dihydro-
Graves and Vennesland (197). It was purified orotate is dependent on added cysteine,
and studied by Lieberman and Kornbery (311), whereas the interaction with NADH2 is not (6,
Aleman et al. (5, 6, 210), and Miller and Massey 177, 178). On addition of high amounts of
(339, 340); Friedmann and Vennesland (177, NADH2, not only flavins present in the enzyme
178) crystallized the enzyme. It contains equal are reduced but also other chromophoric
amounts of FAD and riboflavine 5'-phosphate groups, such as iron possibly in concert with the
(FMN) (5, 178) and about 1 mol each of iron (5, labile sulfide (339).
178, 339) and labile sulfide (5, 339) per mol of C. oroticum is reported to contain also a bio-
VOL. 40, 19#76 DEGRADATION OF PURINES AND PYRIMDINES 445
synthetic-type constitutive enzyme, dihydro-or- stead of NAD in the orotate reductase (L-5,6-
otate oxidase (L-5,6-dihydro-orotate:oxygen oxi- dihydro-orotate:NADP+ oxidoreductase [EC
doreductase [EC 1.3.3.11), which is induced by 1.3.1.15]) and the absence of the enzyme in-
growth in media containing orotate (177, 499). volved in the reversible conversion of N-carba-
Moreover, such conditions cause a threefold in- moyl aspartate to 5-carboxymethylhydantoin
crease of both FMN and FAD in the cells, and (424).
Kondo et al. (280) presented evidence that this Orotate reductase (NADP) is also present in
enhancement is related to the formation of this an unidentified aerobic bacterium and pos-
flavoprotein. sesses many characteristics similar to the en-
As a result of the orotate reductase action, zyme from C. oroticum (522).
S()-5,6-dihydro-orotate is formed. The free
acid of this compound exhibits a specific optical Pyrimidine Biosynthetic Pathway

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


rotation of [a](water) = +66.0° (312). The same The pyrimidine biosynthetic pathway (Fig.
optical isomer of dihydro-orotate is degraded by 20) has been reviewed by O'Donovan and Neu-
dihydroorotase (L-5,6-dihydro-orotate amidohy- hard (371). The biosynthetic dihydro-orotate ox-
drolase [EC 3.5.2.3]) of C. oroticum (312). The idase system differs markedly from orotate re-
enzyme requires a cation, probably Zn2, in its ductase described in the previous section. The
catalytic function (451) and is noncompetitively biosynthetic system in Escherichia coli is con-
inhibited by substituted sulfonamide. A sepa- stitutive, is composed of particle-bound en-
rate enzyme, carboxymethylhydantoinase (L- zymes, and is linked with the respiratory chain
5-carboxymethylhydantoin amidohydrolase (257, 258, 264, 497, 498, 610). Similar results
[EC 3.5.2.4]), enables the bacterium to convert were obtained in studies on the enzyme of a
N-carbamoyl-S(i,)-aspartate reversibly into pseudomonad (338, 499). However, when oro-
S(iL)-5-carboxymethylhydantoin (312). The tate was added to the growth medium, a new
free acid of this compound exhibits a specific soluble NADP-linked orotate reductase was
optical rotation of [a](water) = -98.9° (312). formed in this organism (499). Soluble and
This reaction is a spur off the main pathway of bound forms of enzymes catalyzing the conver-
metabolism (313). sion of dihydro-orotate to orotate are found in
The equilibria formed during both enzy- Staphylococcus aureus, which synthesizes py-
matic conversions of N-carbamoyl-L-aspartate rimidines according to the reaction given in
are described (312) by the equilibrium con- Fig. 20 (333). Both forms are active in 2,6-
stants: dichlorophenol- indophenol reduction assays;
the particulate enzyme normally links to oxy-
K - [N-carbamoyl-S(i,)-aspartate]= 19 gen via a cytochrome system. Orotate is also in-
[S(iW)-5,6-dihydroorotate] volved in the synthesis of pyrimidines in Lacto-
[N-carbamoyl-S(L)-aspartate] bacillus bulgaricus (608). Dihydro-orotate oxi-
K= 1.9 dase of this organism is a soluble flavoprotein,
[S(i,)-5-carboxymethylhydantoinI s containing only FMN as the prosthetic group.
N-carbamoyl-Lraspartate is degraded to i-as- Redox dyes, oxygen, or cytochrome c are used
partate, ammonia, and CO2 by ureidosuccinase as electron acceptors, but the purified enzyme
(N-carbamoyl-Iraswprtate amidohydrolase [EC is not active with pyridine nucleotides. The
3.5.1.7]). The 10-fold purified enzyme from C. synthesis of the enzyme is repressed in cells
oroticum is not active with the R(D.)-isomer, growing in the presence of orotate and uracil
with S(i.)-5-carboxymethylhydantoin, or with (500).
a number of other compounds (313). It showed The enzymic activity of pyrimidine synthe-
an absolute requirement for metal ions; Mn2+ sis in S. aureus is 6- to 20-fold higher in anaero-
and Fe2+ ions were found to give the greatest bically or semianaerobically grown bacteria
effect. Cysteine stimulates the reaction several than in those grown in air, unless uracil is
fold. Since phosphate is not required in the present. Aerobic cultures rapidly accumulate
reaction, carbamoyl phosphate is most probably dihydro-orotate in the medium after transfer to
not an intermediate (313). anaerobic conditions. These results show that
Reynolds et al. (424) described the metabolic the requirement for uracil displayed by S. au-
pathway of orotic acid degradation by two uni- reus, when growing anaerobically, is due to its
dentified corynebacteria isolated from soil. One inability to dehydrogenate dihydro-orotate in
of the organisms grows in a medium containing the absence of oxygen. The enzyme synthesis is
orotic acid as the sole organic substrate. The derepressed in response to pyrimidine starva-
pathway is similar to that found in C. oroti- tion (331). Dihydro-orotase is present in E. coli
cum, except for the involvement of NADP in- (450, 497, 610) and P. fluorescens (336). The
446 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
enzyme from E. coli was purified 145-fold and cells are induced by growth on either thymine
does not react with dihydrouracil and dihydro- or uracil. These cells oxidize barbiturate as
thymine (450). well, but a number of other pyrimidines, in-
cluding cytosine and dihydrouracil, were not
Oxidative Pathway oxidized (216). Wang and Lampen (578-580) iso-
In 1952 three groups of authors found an lated from soil a gram-positive, motile, nonspo-
oxidative pathway of pyrimidine degradation rulating rod, which utilized uracil, cytosine,
(Fig. 22) in bacteria which are able to grow thymine, or barbituric acid as the sole source of
aerobically in media containing uracil or thy- C and N for growth, but orotic acid was not
mine as the sole source of nitrogen and carbon. used. Lara (300, 301) observed that members of
Hayaishi and Kornberg (216) isolated two the genera Corynebacterium and Nocardia are

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


bacteria from etirichment cultures which con- able to grow aerobically in media containing
tained thymine and uracil as the sole organic either thymine or uracil as the only carbon,
substrates and which were inoculated with soil. nitrogen, and energy source. Cells of N. coral-
One was tentatively assigned to the genus Cor- lina adapted to thymine were simultaneously
ynebacterium, and the other was a Mycobacte- adapted to uracil and barbituric acid (300).
rium. The thymine-oxidizing enzymes of these Later, Vitols et al. (562) reported on two
strains of the genus Mycobacterium that grow
NH2
on synthetic media containing orotic acid as the
NH2
sole source of nitrogen. Cells of these bacteria
Nl- I,
CH N C-CH3
convert orotic acid to uracil; barbituric acid and
11CH urea were identified in the culture liquid,
O=C
N
. whereas no dihydroorotic acid, carbamoyl as-
H H partate, or aspartate was formed. Cytosine
Cytosine
H20
5- Methylcytosine (216, 580) and 5-methylcytosine (216) are deami-
tH20 nated by an inducible (216) cytosine deaminase.
NH3 (1) NH34 (1)
Uracil dehydrogenase is able to oxidize ura-
0 0 0 cil, thymine (216, 580), and probably also 5-
I'l- K.. CO2 -C Ill l'
aminothymine (580). The electron acceptors of
HN CH HN CH HN C-CH3
1 11CH 11 this enzyme are not NAD or NADP, but meth-
O=C CH O=C N O=C C ylene blue may be used stoichiometrically un-
N COOH N N
H H H der anaerobic conditions or in lesser amounts
Orotic acid Uracil Thymine under aerobic conditions where it is autooxidiz-
H20 (2) (2) H20 able (216, 580). Barbituric acid is formed, which
ZH 2H is transformed to urea and malonic acid (214,
~~~~~~~~~0
11 MB 11 216, 301). Ureidomalonic acid may be an inter-
mediate in this reaction that is catalyzed by
HN CH2 HN CH-CH3 barbiturase. Hayaishi and Kornberg (215, 216)
02
=CL C=O O=C H~C=O purified barbiturase eightfold and showed that
N
H
NH it does not react with a number of other com-
Barbituric acid 5-Methylbarbituric acid pounds, including 5-methylbarbiturate. The
1
(3) fate of 5-methylbarbiturate is still unknown
H2N COOH
and, thus, the question is left open as to how
O=C tH2
the organisms gain carbon and nitrogen from
HN-C=0
thymine. N. corallina splits urea to CO2 and
Ureidomalonic acid ammonia (301), but the nonsporulating rod,
studied by Wang and Lampen (580), does not
1 H3) contain urease. Nevertheless, 2 mol of ammonia
H2N COOH
O=C + CH2
is formed from uracil and thymine, which
H2N COOH suggests that the ureido group is not released
Urea Malonic acid as a unit during the oxidative degradation.
The role of barbiturate in the metabolism of
FIG. 22. Oxidative pathway of pyrimidine degra- Bacillus popilliae is unclear. Barbiturate is re-
dation. (1) Cytosine deaminase (cytosine aminohy-
drolase [EC 3.5.4.1]. Acts also on 5-methylcytosine. quired for constant growth in a synthetic me-
(2) Uracil dehydrogenase (uracil:[acceptor] oxidore- dium, and this requirement was not replaced
ductase [EC 12.99.1]. Also oxidizes thymine. (3) by the common pyrimidines and purines. It
Barbiturase (barbiturate amidohydrolase [EC stimulates the synthesis of both nucleic acid
3.52.1D. MB, Methylene blue. and protein (117, 491).
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 447
Pyrimidine Degradation by Various dines in the degradation of purines by Veillo-
Microorganisms nella alcalescens was discussed in a previous
A number of microorganisms have been section.
tested for their ability to use pyrimidines as a ECOLOGICAL ASPECTS
source of nitrogen or for the presence ofcytosine
deaminase (Table 15). Tetrahymena pyriformis Large amounts of purines, pyrimidines, uric
requires preformed pyrimidines for growth acid, and allantoin are produced in a number of
since it is unable to synthesize the pyrimidine ecosystems. Various bacteria appear to be par-
nucleus (220). Uracil is degraded by fruiting ticularly adapted to growth in such ecosystems.
bodies of Agaricus bisporus and Lycoperdon This adaptation and some examples of the eco-
pyriforme to urea. It was postulated that a systems will be dealt with in this section.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


reductive pattern implying ribotide derivatives
might be operative in the degradation (417). Organisms
The cyanobacterium Agmenellum quadrupli- A special kind of adaptation to substrates
catum shows scant growth on thymine and ura- like methylpurines, uric acid, and allantoin is
cil but no growth on orotic acid, dihydroorotic observed in studies with Clostridium and Ba-
acid, and cytosine (256). S. cerevisiae possesses cillus but may be more widely distributed in
a common active transport system for adenine, nature. Three examples are given here.
guanine, hypoxanthine, and cytosine (384, 416) Clostridium acidiurici and C. cylindrospo-
and a specific one for uracil (202). Pyrimidines rum grow in media containing 0.2% uric acid
are deaminated only slowly or not at all by Vi- and 0.5% (volume) yeast autolysate as sole or-
brio cholerae (4) and not at all by strains of ganic substrates (33). No growth was observed
Streptococcus faecalis, S. faecium, and S. dur- in media containing peptone, tryptone, yeast
ans (334). In E. coli the uptake of uracil is autolysate, or other rich compounds instead of
controlled by cyclic AMP, which may explain uric acid. Only very small amounts of ammonia
part of the inhibiting effect ofcyclic AMP on the were produced from these media when offered
growth of E. coli on glucose (246). E. coli con- together with uric acid. Glucose was not de-
verts the C2 atom of thymine and uracil to CO. graded by cells growing on uric acid (33).
The induction of the enzymes involved in the Kurtzman and Schwimmer (292) isolated a
degradation is prevented by the presence of strain ofBacillus coagulans from soil on plates
NH4+ in the medium (25). The role of pyrimi- containing 0.01 M caffeine in Czapek medium.
TABLE 15. Utilization of pyrimidines by various microorganisms and presence of cytosine deaminasea
Substrate Presence of
Organism cytosine de- Reference
Cytosine Uracil Thymine aminase
Tetrahymena pyriformis - 152
38 Chlamydomonad algae -N -N -N 88
Myxococcus virescens b +CN 368
M. fulvus b +CN 368
M. coralloides (Chondrococcus b +CN 368
coralloides)
Pseudomonas acidovorans +N +N +N, -C + 263
P. oleovorans + 448
Alcaligenes eutrophus H16 (Hy- +N -N -N +C 254
drogenomonas H16)
Azotobacter chroococcum (-)N -N 227
A. vinelandii (-)N -N 227
Aerobacter cloacae +N 343
A. aerogenes +N 343
Escherichia coli +Nd +N + 25, 103, 343
Salmonella typhimurium +N +N 41
Veillonella alcalescens (Micro- +N +N +N + 587
coccus lactilyticus)
a The substrates were tested as carbon (C), nitrogen (N), or carbon and nitrogen (CN) sources for growth.
Symbols: +, positive results; (-), doubtful results; and -, negative results.
b Cytidine is utilized as the source of carbon, nitrogen, and energy.
c
Inducible.
d
A majority of E. coli strains use uracil as a sole source of nitrogen (343).
448 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
Subsequent cultures of the isolate degraded caf- Lond Island, contains a high amount of organic
feine readily, but growth requirements on other nitrogen, particularly uric acid, due to the pres-
media appeared to be rather fastidious. An au- ence of duck farms located around the Bay.
thentic B. coagulans did not grow in the afore- Estuarine waters contain about 106 urea-
mentioned medium. The authors suggest a de- decomposing bacteria per liter, whereas the
gree of selective adaptation in this organism. number in marine waters is around 3 x 103 to
B. fastidiosus can be readily isolated from 4 x 103 cells/liter (421). Uric acid may be de-
various sources, including soil. The fastidious graded also in the absence of microorganisms,
requirement for uric acid and for its degrada- since Antia and Landymore (17) reported uric
tion products was described by Den Dooren de acid degradation in a seawater medium mainly
Jong (133) and confirmed by four independent as a result of the presence of traces of metal
groups (62, 253, 305, 329). No rich medium is ions.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


known that supports growth of this organism to Mammal intestines. The effect of feeding
a level comparable to synthetic media contain- yeast or other single cells on the serum level of
ing uric acid, allantoin, or allantoate as the uric acid and the excretion of uric acid is dis-
sole organic substrates (62). Besides these or- cussed in the Introduction. Upon feeding higher
ganisms, perhaps other ones are adaptively amounts of yeast, the amount of uric acid ex-
specialized in the degradation of purines and creted does not increase linearly with the
pyrimidines, but no studies have been made in amount of yeast ingested (149). Perhaps intes-
this field. Moreover, it should be worthwhile to tinal organisms are involved in the degradation
investigate the possible plasmid-born genetic of uric acid. Such degradation was found by
information enabling some bacteria, especially many other authors (184, 319, 320, 501) for uric
those of the Streptococcus allantoicus-Entero- acid administered orally to humans and allan-
bacteriaceae group, to degrade uric acid and toin or uric acid fed to other mammals (231);
allantoin. This might explain why Escherichia intravenously administered uric acid was re-
coli and S. allantoicus subcultured in media covered almost completely as urinary uric acid
without uric acid or allantoin gradually lose (184), but perhaps part of a person's daily uric
the capacity to degrade these substances acid excretion is normally recycled to the intes-
(Vogels and Van der Drift, unpublished data). tine from the blood and degraded by the urico-
lytic bacteria. This was found to be true for
Ecosystems urea (555). A number of the bacteria described
Den Dooren de Jong noticed (133) that about above in the section on the anaerobic, degrada-
10% of the bacterial strains isolated from the tive pathways of purines and allantoin are
Maas River and about half of the strains iso- likely candidates for intestinal uricolysis, but
lated from tap water were able to degrade uric in analogy to the avian caecum many other
acid. Antheunisse (16) tested the ability of soil bacterial species may be involved (36).
microorganisms to degrade uric acid. About Urea formed in these reactions can be con-
half of the microorganisms present in clay soil verted to ammonia by Selenomonas ruminan-
(2 x 107 to 4 x 107/g) were able to decompose tium and Peptostreptococcus productus, which
uric acid in the presence of yeast extract and contain a urease that is strongly repressed by
glucose. A large number of coryneform strains NH4+ ions (556). Ammonia is the major nitro-
isolated from sandy soils, peaty soil, cheese, gen source for growth of intestinal bacteria and
fish, seawater, and sewage decompose uric acid is essential for growth of Ruminococcus bromii
(16). (80).
No quantitative data are known concerning Avian caecum and insect intestines. Uric
the concentration of purines, pyrimidines, or acid is the main excretory nitrogenous sub-
their degradation products in marine environ- stance of birds (about 6% of dried poultry waste
ments, but the studies of Remsen et al. (421) consists of uric acid [55]) and insects. Anaerobic
demonstrated that urea is a major nitrogen uric acid-degrading bacteria were found by Bar-
source in the upper marine water layers as ker and Beck (33) in fecal material of the yel-
compared with inorganic nitrogen sources as low-shafted flicker and were demonstrated to
ammonia, nitrate, and nitrite. The urea con- occur in large numbers in intestines of termites
centration in the open North Atlantic is about and cockroaches (471) and poultry caeca (36,
0.28 uM. A relatively high concentration (1.7 449). In the latter case the number of uric acid-
,MM) is found off the coast of Peru due to the decomposing anaerobic bacteria amounts to be-
huge quantities of bony fish and birds, both of tween 5.4 x 108 and 1.8 x 1010/g (wet weight) of
which excrete large amounts of uric acid. By caecal material (36). Among them were Bacte-
the same token, the water of Great South Bay, roides, Sphaerophorus, Fusobacterium, Eu-
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 449
bacterium, an anaerobic Streptococcus, P. pro- groups appear to be unable to utilize uric acid
ductus, (as well as other Peptostreptococcus spe- as a sole nitrogen source (481).
cies), and Clostridium malenominatum (36). Symbiotic uric acid-degrading bacteria. In
No studies were made on the degradation path- the fat body and ovaries of cockroaches, special
way of uric acid in these organisms. cells, mycetocytes, are found in which sym-
The decomposition of uric acid in built-up biotic bacteria are present. The bacteria have
poultry litter has been studied (458). The num- been isolated by Keller (262) and have been
ber of uric acid decomposers varied between 2.2 shown to grow in vitro under aerobic conditions
x 109 and 64 x 109/g (fresh weight) of the mate- on a medium containing uric acid as the sole
rial. Their proportion of the total number of source of carbon and nitrogen. The ability of the
bacteria present was about 25% and included bacteria to utilize uric acid is of particular in-
Corynebacterium, Nocardia, Streptomyces, Pseu- terest, since this compound is the main end

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


domonas, Alcaligenes, Achromobacter, and Cyto- product of nitrogen metabolism in the cock-
phaga, which were able to transform uric acid roach and is deposited within fat body cells as
into ammonia or urea. Only small amounts of well as excreted. The symbiotic bacteria enable
anaerobic uric acid decomposers were found, but the host to utilize the waste material and per-
the presence of organisms resembling C. acidi- haps offer a route for the remobilization of ni-
urici was suggested (458). trogen stored as uric acid should this subse-
Rumen. Besides the purines and pyrimidines quently be required by the host.
as constituents of nucleic acids, plants also con- Donnellan and Kilby (142) isolated a motile,
tain free purines, uric acid, and allantoin. gram-negative curved rod from the fat body of
About halfofthe nonprotein nitrogen content of adult Periplaneta americana. The bacterium
grassland herbage consists of purines and py- grows aerobically in a medium containing uric
rimidines, either free or bound (154). Allantoin acid as the organic substrate in the presence of
and uric acid are present in various Gramineae small amounts of Lab-lemco. Uric acid is de-
and Leguminosae (514) and are nitrogen graded by uricase, which can be extracted from
sources for the rumen microflora (44). The con- insoluble cell debris at pH 9. Allantoin is
centration of uric acid in bovine rumen content formed in this reaction and is subsequently
is reported to be 5 mg/100 ml, but no data are degraded to allantoate, -areidoglycolate, and
available on the amount of allantoin, purines, glyoxylate. Ammonia is formed from urea by
or pyrimidines present (509). The bacteria may urease, and glyoxylate is degraded along the
benefit the host by converting these compounds tartronate semialdehyde pathway.
into a usable form. Cell suspensions of bovine LITERATURE CITED
rumen bacteria degrade xanthine, guanine,
and uric acid to ammonia, fatty acids, and CO2, 1. Abbondandolo, A., A. Weyer, H. Heslot, and
but hypoxanthine and adenine are less readily M. Lambert. 1971. Study of adenine amino-
hydrolase in the yeast Schizosaccharomyces
attacked (247). Reports on the degradation of pombe. J. Bacteriol. 108:959-963.
purines or pyrimidines by pure cultures of ru- 2. Abbott, M. T., E. K. Schandl, R. F. Lee, T. S.
men bacteria are scant. Forty-two percent of Parker, and R. J. Midgett. 1967. Cofactor
the rumen coliform isolates are capable of grow- requirements of thymine 7-hydroxylase.
ing on a medium that contained uric acid as the Biochim. Biophys. Acta 132:525-528.
primary source of carbon and nitrogen. The 3. Affonso, 0. R., and E. Mitidieri. 1965. Molyb-
predominant uricolytic organism in this group denum requirement and tungstate inhibition
is Paracolobactrum aerogenoides. This bacte- of xanthine dehydrogenase activity of Pseu-
rium degrades uric acid in a pathway involving domonas aeruginosa. An. Acad. Bras. Cienc.
37:289-294.
allantoin and glyoxylate, and urea is an end 4. Agarwala, S. C., C. R. Krishna Murti, D. L.
product (509). Shrivastava, and A. Sen Gupta. 1954. Metab-
S. ruminantium can use adenine and uric olism of purine and pyrimidine compounds
acid as nitrogen source, but allantoin, xan- by vibrios. Enzymologia 16:322-328.
thine, and uracil are not used (239). 5. Aleman, V., and P. Handler. 1967. Dihydrooro-
Skin. Another ecosystem in which uric acid tate dehydrogenase. I. General properties. J.
together with amino acids, ammonia, urea, and Biol. Chem. 242:40874096.
creatinine act as the most common nitrogen 6. Aleman, V., S. T. Smith, K. V. Rajagopalan,
source is formed by the human integument and and P. Handler. 1966. The mechanism of
action of some soluble metallo-flavoproteins,
its autochthonous skin organisms. A considera- p. 99-114. In E. C. Slater and C. Veeger
ble part of the staphylococci, the diphtheroids, (ed.), Symposium on flavins and flavopro-
and gram-negative bacteria present in the skin teins. American Elsevier Publishing Co.
is able to degrade uric acid, but the first two Inc., New York.
450 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
7. Allam, A. M., and T. A. Elzainy. 1969. Degra- 22. Baginsky, M. L., and F. M. Huennekens. 1966.
dation of xanthine by Penicillium chryso- Electron transport function of a heat-stable
genum. J. Gen. Microbiol. 56:293-300. protein and a flavoprotein in the oxidative
8. Allam, A. M., and T. A. Elzainy. 1970. Utiliza- decarboxylation of glycine by Peptococcus
tion and deamination of adenine by Penicil- glycinophilus. Biochem. Biophys. Res. Com-
lium chrysogenum. U.A.R. J. Chem. 13:253- mun. 23:600-605.
255. 23. Balinsky, J. B. 1970. Nitrogen metabolism in
9. Allam, A. M., and T. A. Elzainy. 1970. Purine amphibia in relation to environment, p. 15-
catabolism in Fusarium moniliforme. J. 22. In B. Schmidt-Nielsen and D.W.S. Kerr
Gen. Microbiol. 63:183-187. (ed.), Urea and the kidney. Excerpta Medica
10. Ammann, E. C. B., and V. H. Lynch. 1964. Foundation, Amsterdam.
Purine metabolism by unicellular algae. II. 24. Balis, M. E., D. H. Levin, G. B. Brown, G. B.
Adenine, hypoxanthine, and xanthine deg- Elion, H. VanderWerff, and G. H. Hitch-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


radation by Chlorella pyrenoidosa. Biochim. ings. 1952. The incorporation of exogenous
Biophys. Acta 87:370-379. purines into pentose nucleic acid by Lactoba-
11. Ammann, E. C. B., and V. H. Lynch. 1966. cillus casei. J. Biol. Chem. 196:729-747.
Purine metabolism by unicellular algae. III. 25. Ban, J., L. Vitale, and E. Kos. 1972. Thymine
The photochemical degradation of uric acid and uracil catabolism in Escherichia coli. J.
by chlorophyll. Biochim. Biophys. Acta Gen. Microbiol. 73:267-272.
120:181-182. 26. Barash, I. 1972. Accumulation of urea and al-
12. Ammann, E. C. B., and L. L. Reed. 1967. lantoin during purine utilization by germi-
Metabolism of nitrogen compounds by Hy- nating spores of Geotrichum candidum. J.
drogenomonas eutropha. I. Utilization of uric Gen. Microbiol. 72:539-542.
acid, allantoin, hippuric acid, and creati- 27. Bare, L. N., R. F. Wiseman, and I. Ruchman.
nine. Biochim. Biophys. Acta 141:135-143. 1966. Uricolysis by Escherichia spp. Appl.
13. Andreesen, J. R., E. El Ghazzawi, and G. Microbiol. 14:474.
Gottschalk. 1974. The effect of ferrous ions, 28. Barker, H. A. 1943. Streptococcus allantoicus
tungstate and selenite on the level of formate and the fermentation of allantoin. J. Bacte-
dehydrogenase in Clostridium formicoaceti- riol. 46:251-259.
cum and formate synthesis from C02 during 29. Barker, H. A. 1956. Bacterial fermentations.
pyruvate fermentation. Arch. Microbiol. John Wiley and Sons Inc., New York.
96:103-118. 30. Barker, H. A. 1956. Bacterial fermentations, p.
14. Andreesen, J. R., and L. Ljungdahl. 1971. Con- 70-86. In D. E. Green (ed.), Currents in bio-
version of C02 (HCO3-) to formyltetrahydro- chemical research. Interscience Publishers,
folate by formate dehydrogenase and formyl- New York.
tetrahydrofolate synthetase from Clostrid- 31. Barker, H. A. 1961. Fermentation of nitroge-
ium thermoaceticum. Bacteriol. Proc., p. 166. nous organic compounds, p. 193-194. In I. C.
15. Andrews, J. C., and I. T. Sell. 1955. The prop- Gunsalus and R. Y. Stanier (ed.), The bacte-
erties and interrelationship of oxaluric and ria, vol. 2. Academic Press Inc., New York.
parabanic acids. Arch. Biochem. Biophys. 32. Barker, H. A., and J. V. Beck. 1941. The fer-
56:405-411. mentative decomposition of purines by Clos-
16. Antheunisse, J. 1972. Decomposition of nucleic tridium acidi-urici and Clostridium cylin-
acids and some of their degradation products drosporum. J. Biol. Chem. 141:3-27.
by microorganisms. Antonie van Leeuwen- 33. Barker, H. A., and J. V. Beck. 1942. Clostrid-
hoek J. Microbiol. Serol. 38:311-327. ium acidi-urici and Clostridium cylindrospo-
17. Antia, N. J., and A. F. Landymore. 1974. Phys- rum, organisms fermenting uric acid and
iological and ecological significance of the some other purines. J. Bacteriol. 43:291-304.
chemical instability of uric acid and related 34. Barker, H. A., and S. R. Elsden. 1947. Carbon
purines in sea water and marine algal cul- dioxide utilization in the formation of gly-
ture medium. J. Fish. Res. Board Can. cine and acetic acid. J. Biol. Chem. 167:619-
31:1327-1335. 620.
18. Arima, K., and K. Nose. 1968. Studies on bacte- 35. Barker, H. A., B. E. Volcani, and B. P. Car-
rial urate: oxygen oxidoreductase. I. Purifi- don. 1948. Tracer experiments on the mecha-
cation and properties of the enzyme. nism of glycine fermentation by Diplococcus
Biochim. Biophys. Acta 151:54-62. glycinophilus. J. Biol. Chem. 173:803-804.
19. Arst, H. N., Jr., D. W. MacDonald, and D. J. 36. Barnes, E. M., and C. S. Impey. 1974. The
Cove. 1970. Molybdate metabolism in Asper- occurrence and properties of uric acid decom-
gillus nidulans. I. Mutations affecting ni- posing anaerobic bacteria in the avian cae-
trate reductase and/or xanthine dehydrogen- cum. J. Appl. Bacteriol. 37:393-409.
ase. Mol. Gen. Genet. 108:129-145. 37. Battelli, M. G., E. Lorenzoni, and F. Stirpe.
20. Bachrach, U. 1957. The aerobic breakdown of 1973. Milk xanthine oxidase Type D (dehy-
uric acid by certain pseudomonads. J. Gen. drogenase) and Type 0 (oxidase). Purifica-
Microbiol. 17:1-11. tion, interconversion and some properties.
21. Bachstez, M. 1930. Constitution of orotic acid. Biochem. J. 131:191-198.
Ber. Dtsch. Chem. Ges. B 63:1000-1007. 38. Battelli, F., and L. Stern. 1909. Untersuchun-
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 451
gen uber die Urikase in den Tiergeweben. 57. Blakley, R. L. 1969. The biochemistry of folic
Biochem. Z. 19:219-253. acid and related pteridines. North Holland
39. Baum, H., G. Hubscher, and H. R. Mahler. Publishing Co., Amsterdam.
1956. Studies on uricase. II. The enzyme- 58. Blattmann, P., and J. Rktey. 1972. Stereospeci-
substrate complex. Biochim. Biophys. Acta ficity of the dihydroorotate-dehydrogenase
22:514-527. reaction. Eur. J. Biochem. 30:130-137.
40. Baum, H., H. R. Mahler, and G. Hubscher. 59. Bojanowski, R., E. Gaudy, R. C. Valentine,
1956. Studies on uricase. III. The oxidation of and R. S. Wolfe. 1964. Oxamic transcarbam-
uric acid by model copper complexes. Bio- ylase of Streptococcus allantoicus. J. Bacte-
chim. Biophys. Acta 22:528-536. riol. 87:75-80.
41. Beck, C. F., J. L. Ingraham, J. Neuhard, and 60. Bojanowski, R., R. C. Valentine, E. T. Gaudy,
E. Thomassen. 1972. Metabolism of pyrimi- and R. S. Wolfe. 1962. Oxamic transcarbam-
dines and pyrimidine nucleosides by Salmo- ylase. Bacteriol. Proc., p. 103.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


nella typhimurium. J. Bacteriol. 110:219- 61. Bollard, E. G. 1959. Urease, urea and ureides
228. inoplants. Symp. Soc. Exp. Biol. 13:304-329.
42. Beck, J. V., R. D. Sagers, and L. R. Morris. 62. Bongaerts, G. P. A., and G. D. Vogels. 1976.
1957. Intermediary metabolism of Clostrid- Uric acid degradation by Bacillus fastidiosus
ium acidi-urici. I. Formation of pyruvate strains. J. Bacteriol. 125:689-697.
from glycine. J. Bacteriol. 73:465-469. 63. Bonicke, R. 1970. Ober das Vorkommen von
43. Behrend, R., and 0. Roosen. 1889. Synthese Acylamidasen in Mycobakterien. Ill. Der en-
der Harnsaure. Ann. Chem. 251:235-256. zymatische Abbau von Allantoin. Zentralbl.
44. Belasco, I. J. 1954. New nitrogen feed com- Bakteriol. Parasitenkd. Infektionkr. Hyg.
pounds for ruminants-a laboratory evalua- Abt. 1 Orig. 178:186-194.
tion. J. Anim. Sci. 13:601-610. 64. Bordeleau, L. M., and R.-A. Lachanche. 1968.
45. Bentley, R., and A. Neuberger. 1952. The Effets des purines et des pyrimidines sur la
mechanism of the action of uricase. Biochem. croissance de Corynebacterium sepedonicum
J. 52:694-699. (Spiek. & Kott.) Skapt. & Burkh. Can J.
46. Benziman, M., R. D. Sagers, and I. C. Gunsa- Microbiol. 14:475-478.
lus. 1960. L-Serine-specific dehydrase from 65. Bothe, H., and U. Nolteernsting. 1975. Pyru-
Clostridium acidurici. J. Bacteriol. 79:474- vate dehydrogenase complex, pyruvate: fer-
479. redoxin oxidoreductase and lipoic acid con-
47. Bergman, T. 1776. K. Vet. Acad. Handlingar tent in microorganisms. Arch. Microbiol.
(Stockholm) 37:333. 102:53-57.
48. Bergmann, F., H. Kwietny, G. Levin, and H. 66. Bradshaw, W. H., and H. A. Barker. 1960.
Engelberg. 1960. Studies on the enzymic oxi- Purification and properties of xanthine dehy-
dation of aminopurines. Biochim. Biophys. drogenase from Clostridium cylindrosporum.
Acta 37:433-441. J. Biol. Chem. 235:3620-3629.
49. Bergmann, F., H. Ungar-Waron, and H. 67. Bradshaw, W. H., and D. J. Reeder. 1964. Fer-
Kwietny-Govrin. 1964. Action of 8-azagua- redoxin coupling of formate oxidation to ur-
nine and 8-azaxanthine on Pseudomonas ate reduction in extracts of Clostridium cy-
aeruginosa. Biochem. J. 91:270-276. lindrosporum. Bacteriol. Proc., p. 110.
50. Bergmann, F., H. Ungar-Waron, H. Kwietny- 68. Brana, H. 1967. Degradation of 5'-adenosine-
Govrin, H. Goldberg, and S. Leon. 1962. monophosphate in a cell-free system ofEsch-
Some specific reactions of the purine oxidi- erichia coli. Folia Microbiol. (Prague) 12:1-5.
zing system of Pseudomonas aeruginosa. 69. Brill, W. J., E. A. Wolin, and R. S. Wolfe.
Biochim. Biophys. Acta 55:512-522. 1964. Anaerobic formate oxidation: a ferre-
51. Berridge, M. J. 1965. The physiology of excre- doxin-dependent reaction. Science 144:297-
tion in the cotton stainer, Dysdercus fascia- 298.
tus Signoret. III. Nitrogen excretion and ex- 70. Brodie, J. D. 1967. Origin of photolabile methyl
cretory mechanism. J. Exp. Biol. 43:535-552. groups in methionine biosynthesis. Biochem.
52. Biltz, H., and H. Schauder. 1923. Ober die Biophys. Res. Commun. 26:261-264.
Oxydation der Harnsaure. J. Prakt. Chem. 71. Brogard, J. M., D. Coumaros, J. Franckhau-
106:108-172. ser, A. Stahl, and J. Stahl. 1972. Enzymatic
53. Birdsey, E. C., and V. H. Lynch. 1962. Utiliza- uricolysis: a study of the effect of a fungal
tion of nitrogen compounds by unicellular urate-oxidase. Rev. Eur. Etud. Clin. Biol.
algae. Science 137:763-764. 17:890-895.
54. Biscaro, G., and E. Belloni. 1905. Sur un nou- 72. Brooke, M. S., and B. Magasanik. 1954. The
veau constituant du lait. Monit. Sci. (Paris) metabolism of purines in Aerobacter aero-
19:384. genes: a study of purineless mutants. J. Bac-
55. Blair, R. 1974. Evaluation of dehydrated poul- teriol. 68:727-733.
try waste as a feed ingredient for poultry. 73. Broughton, W. J., M. J. Dilworth, and I. K.
Fed. Proc. 33:1934-1936. Passmore. 1972. Base ratio determination
56. Blakley, R. L. 1963. The biosynthesis of thymi- using unpurified DNA. Anal. Biochem.
dylic acid. IV. Further studies on thymidyl- 46:164-172.
ate synthetase. J. Biol. Chem. 238:2113- 74. Brown, G. B., P. M. Roll, and L. F. Cavalieri.
2118. 1947. The in vivo oxidation of uric acid. J.
452 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
Biol. Chem. 171:835. allantoin degradation by a Pseudomonass. J.
75. Brown, G. W., Jr., J. James, R. J. Henderson, Bacteriol. 68:598-603.
W. N. Thomas, R. 0. Robinson, A. L. 90. Campbell, L. L., Jr. 1955. Oxidative degrada-
Thompson, E. Brown, and S. G. Brown. tion of uric acid by cell extracts of a Pseu-
1966. Uricolytic enzymes in liver of the dip- domonas. Biochim. Biophys. Acta 18:160-
noan Protopterus aethiopicus. Science 161.
153:1653-1654. 91. Campbell, L. L., Jr. 1957. Reductive degrada-
76. Brunel, A. 1931. Pr6sence de l'allantoinase tion of pyrimidines. I. The isolation and
dans de nomibreux champignons. C. R. Acad. characterization of a uracil fermenting bac-
Sci. 192:442-444. terium, Clostridium uracilicum nov. spec. J.
77. Brunel, A. 1936. Le m6tabolisme de l'azote Bacteriol. 73:220-224.
d'origine purique chez les champignons. 92. Campbell, L. L., Jr. 1957. Reductive degrada-
Ph.D. thesis, Univ. of Paris, Pierre Andre, tion of pyrimidines. II. Mechanism of uracil

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Paris. degradation by Clostridium uracilicum. J.
78. Brunel, A. 1939. Sur l'utilisation par. le Sterig- Bacteriol. 73:225-229.
matocystis nigra des substances azotees prov- 93. Campbell, L. L., Jr. 1957. Reductive degrada-
enant de la degradation de l'acide urique. tion of pyrimidines. III. Purification and
Mesoxaldscarboxylase, oxaluricase et para- properties of dihydrouracil dehydrogenase.
banase. Bull. Soc. Chim. Biol. 21:388-406. J. Biol. Chem. 227:693-700.
79. Brunel, A., and G. Capelle. 1947. Sur 94. Campbell, L. L., Jr. 1958. Reductive degrada-
l'importance biologique des ureides glyoxy- tion of pyrimidines. IV. Purification and
liques chez les etres vivants. I. L'allantoine properties of dihydrouracil hydrase. J. Biol.
et l'acide allantoique chez les vegetaux. Bull. Chem. 233:1236-1240.
Soc. Chim. Biol. 29:427-444. 95. Campbell, L. L., Jr. 1960. Reductive degrada-
80. Bryant, M. P. 1974. Nutritional features and tion of pyrimidines. Enzymatic conversion of
ecology of predominant anaerobic bacteria of N-carbamyl-,f-alanine to ,8-alanine, carbon
the intestinal tract. Am. J. Clin. Nutr. dioxide, and ammonia. J. Biol. Chem.
27:1313-1319. 235:2375-2378.
81. Buchanan, B. B., W. Lovenberg, and J. C. 96. Canellakis, E. S., and P. P. Cohen. 1955. The
Rabinowitz. 1963. A comparison of clostri- end-products and intermediates of uric acid
dial ferredoxins. Proc. Natl. Acad. Sci. oxidation by uricase. J. Biol. Chem. 213:385-
U.S.A. 49:345-353. 395.
82. Buchanan, R. E., and N. E. Gibbons (ed.). 97. Canellakis, E. S., A. L. Tuttle, and P. P.
1974. Bergey's manual of determinative bac- Cohen. 1955. A comparative study of the end-
teriology, 8th ed. The Williams & Wilkins products of uric acid oxidation by peroxi-
Co., Baltimore. dases. J. Biol. Chem. 213:397-404.
83. Buniva and Vauquelin. 1800. Extrait d'un 98. Cardon, B. P., and H. A. Barker. 1947. Amino
m6moire des cit. BUNIVA et VAUQUELIN, acid fermentations by Clostridium propioni-
sur l'eau de l'amnios de femme et de vache. cum and Diplococcus glycinophilus. Arch.
Ann. Chim. (Paris) (1) 33:269-282. Biochem. 12:165-180.
84. Buttlaire, D. H., R. T. Hersh, and R. H. 99. Cato, E., W. E. C. Moore, and L. V. Holde-
Himes. 1972. Hydrogen ion-induced reversi- man. 1968. Clostridium oroticum comb. nov.
ble inactivation and dissociation of formylte- Amended description. Int. J. Syst. Bacteriol.
trahydrofolate synthetase. J. Biol. Chem. 18:9-13.
247:2059-2068. 100. Cecere, F., G. Galli, and F. Morisi. 1975. Sub-
85. Buttlaire, D. H., G. H. Reed, and R. Himes. strate and steric specificity of hydropyrimi-
1975. Electron paramagnetic resonance and dine hydrase. FEBS Lett. 57:192-194.
water proton relaxation rate studies of for- 101. Cerecedo, L. R. 1931. Studies on the physiology
myltetrahydrofolate synthetase-manganous of pyrimidines. IV. Further experiments on
ion complexes. Evidence for involvement of the intermediary metabolism of uracil. J.
substrates in the promotion of a catalytically Biol. Chem. 93:269-274.
competent active site. J. Biol. Chem. 102. Cerecedo, L. R. 1940. The chemistry and me-
250:261-270. tabolism of the nucleic acids, purines and
86. Cahn, R. S., and C. K. Ingold. 1951. Specifica- pyrimidines. Annu. Rev. Biochem. 2:183-
tion of configuration about quadricovalent 198.
asymmetric atoms. J. Chem. Soc. 1951:612- 103. Chargaff, E., and J. Kream. 1948. Procedure
623. for the study of certain enzymes in minute
87. Cahn, R. S., C. K. Ingold, and V. Prelog. 1956. amounts and its application to the investiga-
The specification of asymmetric configura- tion of cytosine deaminase. J. Biol. Chem.
tion in organic chemistry. Experientia 12:81- 175:993-994.
94. 104. Chen, C. C., and L. F. Retger. 1920. A correla-
88. Cain, J. 1965. Nitrogen utilization in 38 fresh- tion study of the colon-aerogenes group of
water chlamydomonad algae. Can. J. Bot. bacteria with special reference to organisms
43:1367-1378. occurring in the soil. J. Bacteriol. 5:253-298.
89. Campbell, L. L., Jr. 1954. The mechanism of 105. Childs, G. E. 1973. Hartmannella culbertsoni:
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 453
enzymatic, ultrastructural, and cytochemi- 120. Curthoys, N. P., L.D'A. Straus, and J. C. Ra-
cal characteristics of peroxisomes in a den- binowitz. 1972. Formyltetrahydrofolate syn-
sity gradient. Exp. Parasitol. 34:44-55. thetase. Substrate binding to monomeric
106. Choi, K. S., K. W. Lee, S. C. Y. Hico, and A. subunits. Biochemistry 11:345-349.
H. Roush. 1968. Assay, purification and 121. Cvancara, V. A. 1969. Comparative study of
properties of allantoicase from Candida liver uricase activity in fresh-water teleosts.
utilis. Arch. Biochem. Biophys. 126:261-268. Comp. Biochem. Biophys. 28:725-732.
107. Choi, K. S., K. W. Lee, and A. H. Roush. 1966. 122. Dagley, S., P. W. Trudgill, and A. G. Callely.
The assay of yeast ureidoglycolatase. Anal. 1961. Synthesis of cell constituents from gly-
Biochem. 17:413-422. cine by a Pseudomonas. Biochem. J. 81:623-
108. Choi, K. S., and A. H. Roush. 1965. Assay, 631.
purification, and properties of glyoxylurease 123. Dalal, R., and J. S. Gots. 1966. Inhibition of
and allantoicase from the yeast Candida 5,10-methylenetetrahydrofolate dehydrogen-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


utilis. Fed. Proc. 24:594. ase by purine nucleotides. Biochem. Bio-
109. Christen, P., W. C. Peacock, A. E. Christen, phys. Res. Commun. 22:340-345.
and W. E. C. Wacker. 1970. Urate oxidase in 124. Dalgliesh, C. E., and A. Neuberger. 1954. The
primate phylogenesis. Eur. J. Biochem. mechanism for the conversions of uric acid
12:3-5. into allantoin and glycine. J. Chem. Soc.
110. Clark, F. E. 1955. The designation of Coryne- 1954:3407-3414.
bacterium ureafaciens Krebs and Eggleston 125. Darlington, A. J., and C. Scazzocchio. 1968.
as Arthrobacter ureafaciens (Krebs and Eg- Evidence for an alternative pathway of xan-
gleston) comb. nov. Int. Bull. Bacteriol. No- thine oxidation in Aspergillus nidulans.
mencl. Taxon. 5:111-113. Biochim. Biophys. Acta 166:569-571.
111. Clarke, P. H., and P. M. Meadow. 1966. The 126. Darlington, A. J., C. Scazzocchio, and J. A.
effect of 8-azaguanine on the inducible oxida- Pateman. 1965. Biochemical and genetical
tion of guanine by Pseudomonas aeruginosa. studies of purine breakdown in Aspergillus.
J. Gen. Microbiol. 44:195-208. Nature (London) 206:599-600.
112. Claus, A. 1874. Mittheilungen aus dem Uni- 127. Davis, R. H. 1970. Sources of urea in Neuro-
versit&tslaboratorium zu Freiburg i.B. Ber. spora. Biochim. Biophys. Acta 215:412-414.
Dtsch. Chem. Des. 7:226-237. 128. Dawbin, W. H., and L. Hill. 1969. Uric acid
113. Coleman, G. S., and J. I. Laurie. 1974. The and urea excretion in the tuatara Sphenodon
metabolism of starch, glucose, amino acids, punctatus. Nature (London) 224:1325-1326.
purines, pyrimidines and bacteria by three 129. De Duve, C., and P. Baudhuin. 1966. Peroxi-
Epidinium spp. isolated from the rumen. J. somes (microbodies and related particles).
Gen. Microbiol. 85:244-256. Physiol. Rev. 46:323-357.
114. Cook, A. R., and D. Boulter. 1964. Utilization 130. Della Corte, E., and F. Stirpe. 1972. The regu-
of urea by Candida flareri. Phytochemistry lation of rat liver xanthine oxidase. Involve-
3:313-319. ment of thiol groups in the conversion of the
115. Cooper, T. G., and R. Lawther. 1973. Induction enzyme activity from dehydrogenase (type
of the allantoin degradative enzymes by allo- D) into oxidase (type 0) and purification of
phanic acid, the last intermediate of the the enzyme. Biochem. J. 126:739-745.
pathway. Biochem. Biophys. Res. Commun. 131. Demain, A. L., and D. Hendlin. 196.7. Phos-
52:137-142. phohydrolases of a Bacillus subtilis mutant
116. Cooper, T. G., and R. Sumrada. 1975. Urea accumulating inosine and hypoxanthine. J.
transport in Saccharomyces cerevisiae. J. Bacteriol. 94:66-74.
Bacteriol. 121:571-576. 132. Den Dooren de Jong, L. E. 1926. Bijdrage tot
117. Coulter, W. H., and R. N. Costilow. 1970. The de kennis van het mineralisatieproces.
role of barbituric acid in the nutrition of Ph.D. thesis, Institute of Technology, Delft.
Bacillus popilliae. Can. J. Microbiol. 16:801- 133. Den Dooren de Jong, L. E. 1929. Uber Bacil-
807. lus fastidiosus. Zentralbl. Bakteriol. Para-
118. Curthoys, N. P., and J. C. Rabinowitz. 1972. sitenkd. Infektionskr. Hyg. Abt. 2. Orig. 79:
Formyltetrahydrofolate synthetase. Binding 344-353.
of folate substrates and kinetics of the re- 134. De Repentigny, J., L. G. Mathieu, S. Turgeon,
verse reaction. J. Biol. Chem. 247:1965-1971. and S. Sonea. 1968. Effects of exogenous pu-
119. Curthoys, N. P., J. M. Scott, and J. C. Rabi- rines on growth rates and other properties of
nowitz. 1972. Folate coenzymes of Clostrid- Staphylococcus aureus strains including a
ium acidi-urici. The isolation of (1-)-5,10- pyrimidineless mutant of the Wood 46 strain.
methenyltetrahydropteroyltriglutamate, its Can. J. Microbiol. 14:39-44.
conversion to (l)-tetrahydropteroyltrigluta- 135. Di Carlo, F. J., A. S. Schultz, and A. M. Kent.
mate and (j)-10-['4C4formyltetrahydro- 1952. On the mechanism of pyrimidine me-
pteroyltriglutamate, and the synthesis of tabolism by yeasts. J. Biol. Chem. 199:333-
(1) - 10 - formyl - [6,7 - 3H2]tetrahydropteroyltri- 343.
glutamate and (l)-[6,7-3H2]tetrahydropter- 136. Di Carlo, F. J., A. S. Schultz, and A. M. Kent.
oyltriglutamate. J. Biol. Chem. 247:1959- 1953. The mechanism of allantoin catabolism
1964. by yeast. Arch. Biochem. Biophys. 44:468-
454 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
474. don) 172:346-347.
137. Di Carlo, F. J., A. S. Schultz, and D. K. Mc- 155. Finck, G. 1950. Biologische und stoffwechsel-
Manus. 1951. The assimilation of nucleic physiologische Studien an Myxococcaceen.
acid derivatives and related compounds by Arch. Mikrobiol. 15:358-388.
yeasts. J. Biol. Chem. 189:151-157. 156. Fink, R. M., R. E. Cline, and H. M. G. Koch.
138. Di Fonzo, M. 1952. Purine enzymes in myco- 1954. Chromatographic detection of pyrimi-
bacteria. Am. Rev. Tuberc. 66:240-243. dine reduction products: microbiological ap-
139. Dikstein, S., F. Bergmann, and Y. Henis. plication. Fed. Proc. 13:207-208.
1957. Studies on uric acid and related com- 157. Fink, R. M., K. Fink, and R. B. Henderson.
pounds. IV. The specificity of bacterial xan- 1953. p-Amino acid formation by tissue slices
thine oxidases. J. Biol. Chem. 224:67-77. incubated with pyrimidines. J. Biol. Chem.
140. Domagk, G. F., and H. H. Schlicke. 1968. A 201:349-355.
colorimetric method using uricase and perox- 158. Fitzpatrick, D. A., 0. FitzGerald, and K. F.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


idase for the determination of uric acid. McGeeney. 1971. An immunological compar-
Anal. Biochem. 22:219-224. ison of uricase from microbial sources. J.
141. Domnas, A. 1962. Amide metabolism in yeasts. Gen. Microbiol. 68:XVI.
II. The uptake of amide and amide like com- 159. Flaschentrager, B., and P. B. Muller. 1938.
pounds by yeast. J. Biochem. (Tokyo) 52:149- Zur Biologie der Oxalsaure. II. Versuche
154. uber oxalogene Verbindungen im Harn. Z.
142. Donnellan, J. F., and B. A. Kilby. 1967. Uric Physiol. Chem. 251:61-69.
acid metabolism by symbiotic bacteria from 160. Forster, R. P., and L. Goldstein. 1966. Urea
the fat body of Periplaneta americana. Comp. synthesis in the lungfish: relative impor-
Biochem. Physiol. 22:235-252. tance of purine and ornithine cycle path-
143. Dryhurst, G., and P. J. Elving. 1968. Polaro- ways. Science 153:1650-1652.
graphic reduction of oxaluric acid. Analyti- 161. Forstmeyer, H. 1974. Enzymic color test for the
cal application. Anal. Chem. 40:492-495. determination of uric acid. Aerztl. Lab.
144. Dudley, K. H., T. C. Butler, and D. L. Bius. 20:125-132.
1974. Role of dihydropyrimidinase in the me- 162. Fosse, R., and A. Brunel. 1929. Un nouveau
tabolism of some hydantoin and succinimide ferment. C.R. Acad. Sci. 188:426-428.
drugs. Drug Metab. Dispos. 2:103-112. 163. Fosse, R., P.-E. Thomas, and P. de Graeve.
145. Duerr, F. G. 1967. The uric acid content of 1934. Allantoine douee du pouvoir rotatoire.
several species of prosobranch and pulmon- C.R. Acad. Sci. 198:689-693.
ate snails as related to nitrogen excretion. 164. Fosse, R., P.-E. Thomas, and P. de Graeve.
Comp. Biochem. Physiol. 22:333-340. 1934. Allantoine l6vogyre. C.R. Acad. Sci.
146. Dumas, R., J. Castel, and R. Jean. 1973. Urate 198:1374-1376.
oxydase en pediatrie. Pathol. Biol. 21:425- 165. Fosse, R., P.-E. Thomas, and P. de Graeve.
429. 1934. Allantoine dextrogyre. Sa presence
147. Durand, G., Y. Guitton, and A. Brunel. 1965. dans le regne vegetal (Platanus orientalis).
Le metabolisme de l'azote chez les microor- C.R. Acad. Sci. 198:1953-1955.
ganismes et les Vegetaux superieurs. II. Les 166. Francois, C. J. 1973. Oxidase activity of xan-
ur6ides. Bull. Soc. Fr. Physiol. Veg. 11:15- thine oxidoreductase, class-specific to mam-
36. mals. Biochem. Syst. 1:231-236.
148. Edmondson, D., V. Massey, G. Palmer, L. M. 167. Franke, W. 1953. Zum Stoffwechsel der Purine
Beacham III, and G. B. Elion. 1972. The und Pyrimidine. Z. Vitam. Horm. Ferment-
resolution of active and inactive xanthine forsch. 5:279-314.
oxidase by affinity chromatography. J. Biol. 168. Franke, W. 1966. Allantoinase und allanto-
Chem. 247:1597-1604. icase, p. 368-383. In F. Hoppe-Seyler and H.
149. Edozien, J. C., U. U. Udo, V. R. Young, and N. Thierfelder (ed.), Handbuch der physiolo-
S. Scrimshaw. 1970. Effects of high levels of gisch- und pathologiseh-chemischen Ana-
yeast feeding on uric acid metabolism of lyse, vol. VT/c. Springer-Verlag, Berlin.
young men. Nature (London) 228:180. 169. Franke, W., and G. E. Hahn. 1955. Untersu-
150. Eguchi, S., S. Okumura, and N. Kasuya. 1967. chungen zum bakteriellen Purinabbau. I.
6,8-Dihydroxypurine. Jpn. Patent 6250. Ab- Ober den Harnsaureabbau durch Pseudomo-
stract no. 48307. Chem. Abstr. 68:4663. nas aeruginosa (Bact. pyocyaneum). Z. Phys-
151. Eichel, H. J. 1956. Purine-metabolizing en- iol. Chem. 299:15-38.
zymes of Tetrahymena pyriformis. J. Biol. 170. Franke, W., and G. E. Hahn. 1955. Untersu-
Chem. 220:209-220. chungen zum bakteriellen Purin-abbau. II.
152. Eichel, H. J. 1957. Some pyrimidine-metaboliz- lber den Abbau von Amino-, Oxy-, und
ing enzymes of Tetrahymena pyriformis. J. Methylpurinen durch Pseudomonas aerugi-
Protozool. 4(Suppl.):16. nosa. (B. pyocyaneum). Z. Physiol. Chem.
153. Elzainy, T. A., and A. M. Allam. 1973. Nucleo- 301:90-106.
side catabolism in Penicillium chrysogenum. 171. Franke, W., and E.-E.M. Taha. 1952. Purinox-
Egypt. J. Chem. 16:329-335. ydierende Fermente aus Schimmelpilzen.
154. Ferguson, W. S., and R. A. Terry. 1953. Pu- III. Zur Kenntnis der Alternaria-Uricasen.
rines in grassland herbage. Nature (Lon- Chem. Ber. 85:913-923.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 455
172. Franke, W., E.-E. M. Taha, and L. Krieg. pionic acid metabolism. V. The conversion of
1952. Purinoxydierende Fermente aus ,-alanine to propionic acid by cell-free ex-
Schimmelpilzen. I. Ober die Uricase der tracts of Clostridium propionicum. J. Biol.
Schimmelpilze. Arch. Mikrobiol. 17:255- Chem. 235:2238-2245.
291. 190. Goldstein, L., and R. P. Forster. 1965. The role
173. Franke, W., A. Thientann, C. Remily, L. of uricolysis in the production of urea by
Mochel, and K. Heye. 1965. Zur Kenntnis fishes and other aquatic vertebrates. Comp.
ureidspaltender Enzyme. I. Soja-Al- Biochem. Physiol. 14:567-576.
lantoinase. Enzymologia 29:251-271. 191. Goodbody, J. 1965. Nitrogen excretion in Asci-
174. Freter, K., J. C. Rabinowitz, and B. Witkop. diacea. II. Storage excretion and the uricoly-
1957. Labile Stoffwechselprodukte. V. Zur tic enzyme system. J. Exp. Biol. 42:299-305.
Biogenese des Formiminoglycins aus 4(5H)- 192. Goodfellow, M., A. Lind, H. Mordarska, S.
imidazolon. Ann. Chem. 607:174-187. Pattyn, and M. Tsukamura. 1974. A co-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


175. Fridovich, I. 1965. The competitive inhibition operative numerical analysis of cultures
of uricase by oxonate and by related deriva- considered to belong to the "rhodochrous"
tives of s-triazines. J. Biol. Chem. 240:2491- taxon. J. Gen. Microbiol. 85:291-302.
2494. 193. Gotto, A. M., and H. L. Kornberg. 1961. The
176. Friedman, S., and J. S. Gots. 1951. Deamina- metabolism of C2 compounds in micro-orga-
tion of isoguanine by Escherichia coli. Arch. nisms. 7. Preparation and properties of crys-
Biochem. Biophys. 32:227-229. talline tartronic semialdehyde reductase.
177. Friedmann, H. C., and B. Vennesland. 1958. Biochem. J. 81:273-284.
Purification and properties of dihydroorotic 194. 's-Gravenmade, E. J., C. van der Drift, and G.
dehydrogenase. J. Biol. Chem. 233:1398- D. Vogels. 1971. Conformation of allanto-
1406. icase in aqueous solution. Biochim. Biophys.
178. Friedmann, H. C., and B. Vennesland. 1960. Acta 251:393406.
Crystalline dihydroorotic dehydrogenase. J. 195. 's-Gravenmade, E. J., G. D. Vogels, and C. van
Biol. Chem. 235:1526-1532. der Drift. 1970. Hydrolysis, racemization,
179. Fukumoto, J., Y. Watanabe, and M. Yano. and absolute configuration of ureidoglyco-
1967. Studies on uricase. I. Induced produc- late, a substrate of allantoicase. Biochim.
tion of uricase by Streptomyces sp. J. Agric. Biophys. Acta 198:569-582.
Chem. Soc. Jpn. 41:540-545. 196. 's-Gravenmade, E. J., G. D. Vogels, and C.
180. Gassner, E. B. 1962. On the pigment absorbing van Pelt. 1969. Preparation, properties and
at 750 mp occurring in some blue-green al- absolute configuration of (-)-alantoin. Rec.
gae. Plant Physiol. 37:637-639. Trav. Chim. Pays-Bas 88:929-939.
181. Gaudy, E. T., and B. Bruce. 1965. Transduc- 197. Graves, J. L., and B. Vennesland. 1957. The
tion with allantoin mutants ofPseudomonas stereospecific hydrogen exchange in the di-
aeruginosa. Bacteriol. Proc., p. 95. hydroorotic dehydrogenase reaction. J.
182. Gaudy, E. T., R. Bojanowski, R. C. Valentine, Biol. Chem. 226:307-316.
and R. S. Wolfe. 1965. Ureidoglycolate syn- 198. Gray, C. T., M. S. Brooke, and J. C. Gerhart.
thetase of Streptococcus allantoicus. I. Meas- 1961. Metabolism of alloxanic acid in a soil
urement of glyoxylate and enzyme purifica- microorganism. J. Bacteriol. 81:755-761.
tion. J. Bacteriol. 90:1525-1530. 199. Gray, C. T., J. C. Gerhart, and M. S. Brooke.
183. Gaudy, E. T., and R. S. Wolfe. 1965. Ureido- 1959. Intermediary metabolism of glyoxylate
glycolate synthetase of Streptococcus allan- and hydroxypyruvate in a soil bacterium.
toicus. II. Properties of the enzyme and Fed. Proc. 18:571.
reaction equilibrium. J. Bacteriol. 90:1531- 200. Greenbaum, F. R. 1940. The story of allantoin.
1536. Am. J. Pharm. 112:205-216.
184. Geren, W., A. Bendich, 0. Bodansky, and G. 201. Greene, R. C., and H. K. Mitchell. 1957. Uri-
B. Brown. 1950. The fate of uric acid in man. case in Neurospora crassa. Arch. Biochem.
J. Biol. Chem. 183:21-31. Biophys. 70:603-613.
185. Gerhardt, B. 1971. Zur Lokalisation von Enzy- 202. Grenson, M. 1969. The utilization of exogenous
men der Microbodies in Polytomella caeca. pyrimidines and the recycling of uridine-5'-
Arch. Mikrobiol. 80:205-218. phosphate derivatives in Saccharomyces cer-
186. Gerhart, J. C., C. T. Gray, and M. S. Brooke. evisiae, as studied by means of mutants af-
1959. The metabolism of alloxanic acid by a fected in pyrimidine uptake and metabolism.
soil bacterium. Bacteriol. Proc., p. 106. Eur. J. Biochem. 11:249-260.
187. Gifford, C. A. 1968. Accumulation of uric acid 203. Gniffiths, M. 1952. Oxidation of uric acid cata-
in the land crab, Cardisoma guanhumi. Am. lyzed by copper and by the cytochrome-cyto-
Zool. 8:521-528. chrome oxidase system. J. Biol. Chem.
188. Gmelin, C. G. 1820. Ueber die Coagulation des 197:399-407.
Eiweiszstoffs durch galvanische Elektricitat, 204. Grigg, G. W. 1972. Effects of coumarin, py-
ein Anfrage, und Bemerkungen Qber die ronin Y, 6,9-dimethyl-2-methylthiopurine
Amniossdure. Ann. Phys. (Gilbert's Ann.) and caffeine on excision repair and recombi-
64:347-353. nation repair in Escherichia coli. J. Gen.
189. Goldfine, H., and E. R. Stadtman. 1960. Pro- Microbiol. 70:221-230.
456 VOGELS AND VAN DER DRIFT BACTERIOL. REv.
205. Grimaux, E. 1876. Report session at Paris. Ber. acteristics of the enzyme and the enzymic
Dtsh. Chem. Ges. 9:1131. reaction. J. Biol. Chem. 237:2903-2914.
206. Grimaux, E. 1877. Recherches synth6tiques 224. Hodson, R. C., S. K. Williams II, and W. R.
sur la serie urique. Ann. Chim. Phys. (5) Davidson, Jr. 1975. Metabolic control of
11:356-432. urea catabolism in Chlamydomonas rein-
207. Guitton, Y., G. Durand, and A. Brunel. 1965. hardi and Chlorella pyrenoidosa. J. Bacte-
Le metabolisme de l'azote chez les microor- riol. 121:1022-1035.
ganismes et les vegetaux superieurs. I. 225. Hoffmeyer, J., and J. Neuhard. 1971. Metabo-
L'urde. Bull. Soc. Fr. Physiol. Veg. 11:3-15. lism of exogenous purine bases and nucleo-
208. Gupta, N. K., and B. Vennesland. 1964. Glyox- sides by Salmonella typhimurium. J. Bacte-
ylate carboligase of Escherichia coli: a flavo- riol. 106:14-24.
protein. J. Biol. Chem. 239:3787-3789. 226. Holt, S. C., and E. R. Leadbetter. 1969. Com-
209. Hahn, A., and W. Lentzel. 1923. Ober das Ver- parative ultrastructure of selected aerobic

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


halten von Pyrimidinderivaten in den Or- spore-forming bacteria: a freeze-etching
ganismen. I. Einfluss von Hefe and Pyrimi- study. Bacteriol. Rev. 33:346-378.
dinderivate. Z. Biol. 79:179-190. 227. Homer, C. K., and F. E. Allison. 1944. Utiliza-
210. Handler, P., K. V. Rajagopalan, and V. Ale- tion of fixed nitrogen by Azotobacter and in-
man. 1964. Structure and function of iron- fluence of nitrogen fixation. J. Bacteriol.
flavoproteins. Fed. Proc. 23:30-38. 47:1-14.
211. Hartenstein, R. C., and I. Fridovich. 1967. Ad- 228. Hosono, R., and S. Kuno. 1974. Mechanism of
enine aminohydrolase. An investigation of inhibition of bacterial growth by adenine. J.
specificity. J. Biol. Chem. 242:740-746. Biochem. (Tokyo) 75:215-220.
212. Harvey, A. M., and H. N. Christensen. 1964. 229. Howell, L. G., and I. Fridovich. 1967. Adenine
Uric acid transport system: apparent ab- aminohydrolase. An investigation of mecha-
sence in erythrocytes of the dalmatian coach nism. J. Biol. Chem. 242:4930-4932.
hound. Science 145:826-827. 230. Hubscher, G., H. Baum, and H. R. Mahler.
213. Haury, H., K. Kronister, and C. Schiener. 1957. Studies on uricase. IV. The nature and
1973. Enzymic measurement of uric acid by composition of some stable reaction products.
UV-test. Med. Klin. (Munich) 68:1161-1163. Biochim. Biophys. Acta 23:43-53.
214. Hayaishi, 0. 1952. Enzymatic conversion of 231. Hunter, R. A., and M. H. Givens. 1914. The
barbituric acid to urea and malonic acid. metabolism of endogenous and exogenous
Fed. Proc. 11:227-228. purines in the monkey. J. Biol. Chem. 17:37-
215. Hayaishi, O., and A. Kornberg. 1951. Enzy- 53.
matic formation of barbituric acid from ura- 232. Imshenetskii, A. A., and L. S. Popova. 1970.
cil and 5-methylbarbituric acid from thy- Isolation and characterization of microorga-
mine. J. Am. Chem. Soc. 73:2975-2976. nisms that decompose uric acid. Mikrobiolo-
216. Hayaishi, O., and A. Kornberg. 1952. Metabo- giya 39:805-811.
lism of cytosine, thymine, uracil, and barbi- 233. Imshenetskii, A. A., and L. S. Popova. 1971.
turic acid by bacterial enzymes. J. Biol. Decomposition of uric acid by aerobic bacte-
Chem. 197:717-732. ria isolated from soil. Mikrobiologiya 40:269-
217. Hayaishi, O., Y. Nishizuka, M. Tatibana, M. 274.
Takeshita, and S. Kuno. 1961. Enzymatic 234. Ingram, L. O., and W. D. Fisher. 1972. Selec-
studies on the metabolism of f3-alanine. J. tive inhibition of deoxyribonucleic acid syn-
Biol. Chem. 236:781-790. thesis by 2-deoxyadenosine in the blue-green
218. Hayashi, H., T. Suga, and S. Niinobe. 1973. bacterium Agmenellum quadruplicatum. J.
Studies on peroxisomes. III. Further studies Bacteriol. 112:170-175.
on the intraparticulate localization of peroxi- 235. Itaya, K., J. Fukumoto, and T. Yamamoto.
somal components in the liver of the rat. 1971. Studies on urate oxidase of Candida
Biochim. Biophys. Acta 297:110-119. utilis. II. Some physical and chemical prop-
219. Heinrich, M. R., V. C. Dewey, and G. W. Kid- erties of the purified enzyme. Agric. Biol.
der. 1953. Utilization of guanine by Tetrahy- Chem. 35:813-821.
menageleii. J. Am. Chem. Soc. 75:1741-1742. 236. Jaenicke, L., and J. Koch. 1962. Zum Mechan-
220. Heinrich, M. R., V. C. Dewey, and G. W. Kid- ismus der Carboligasereaktion. Hydroxy-
der. 1957. The origin of thymine and cytosine methyl-thiaminpyrophosphat, ein neues
in Tetrahymena. Biochim. Biophys. Acta aktives Kohlenstoff-fragment. Biochem. Z.
25:199-200. 336:432-443.
221. Hill, D. L., and P. Chambers. 1967. The purine 237. James, K. A. C., W. P. Tate, and R. Truscoe.
and pyrimidine metabolism of Tetrahymena 1969. Effects of treatment with dithiothreitol
pyriformis. J. Cell. Physiol. 69:321-330. on the extraction, activity and purification of
222. Hilton, M. G., G. C. Mead, and S. R. Elsden. ox-kidney urate oxidase. Enzymologia
1975. The metabolism of pyrimidines by pro- 37:131-152.
teolytic clostridia. Arch. Microbiol. 102:145- 238. Jezewska, M. M. 1972. Purinotelism in slugs
149. Limacidae and Arionidae. Bull. Acad. Pol.
223. Himes, R. H., and J. C. Rabinowitz. 1962. For- Sci. 20:365-368.
matetetrahydrofolate synthetase. II. Char- 239. John, A., H. R. Isaacson, and M. P. Bryant.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 457
1974. Isolation and characteristics of a ureo- tung von Cytosin und Uracil durch Hydro-
lytic strain of Selenomonas ruminantium. J. genomonas facilis und Hydrogenomonas
Dairy Sci. 57:1003-1014. H16. Arch. Mikrobiol. 60-.172-181.
240. Jones, K. M., and E. S. Bridgeland. 1966. The 255. Kaminskas, E., Y. Kimhi, and B. Magasanik.
metabolism of glycine by Arthrobacter globi- 1970. Urocanase and N-formimino-L-gluta-
formis. Biochem. J. 99:25P. mate formiminohydrolase of Bacillus sub-
241. Jones, M. E., and F. Lipmann. 1960. Chemical tilis, two enzymes of the histidine degrada-
and enzymatic synthesis of carbamyl phos- tion pathway. J. Biol. Chem. 245:3536-3544.
phate. Proc. Natl. Acad. Sci. U.S.A. 46:1194- 256. Kapp, R., S. E. Stevens, Jr., and J. L. Fox.
1205. 1975. A survey of available nitrogen sources
242. Jones, M. E., L. Spector, and F. Lipmann. for the growth of the blue-green alga, Agme-
1955. Carbamyl phosphate, the carbamyl do- neUum quadruplicatum. Arch. Microbiol.
nor in enzymatic citrulline synthesis. J. Am. 104:135-138.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Chem. Soc. 77:819-820. 257. Karibian, D. 1973. Dihydro-orotate dehydro-
243. Jones, M. E., L. Spector, and F. Lipmann. genase ofEscherichia coli K12: effects of Tri-
1956. Carbamyl phosphate, p. 278-281. In ton X-100 and phospholipids. Biochim. Bio-
Proceedings of the Third International Con- phys. Acta 302:205-215.
gress on Biochemistry, Brussels, 1955, Per- 258. Karibian, D., and P. Couchoud. 1974. Dihydro-
gamon Press Ltd.. London. orotate oxidase ofEscherichia coli K12: puri-
244. Joyce, B. K., and R. H. Himes. 1966. Formylte- fication, properties, and relation to the cyto-
trahydrofolate synthetase. A study of equi- plasmic membrane. Biochim. Biophys. Acta
librium reaction rates. J. Biol. Chem. 364:218-232.
241:5716-5724. 259. Karlsson, J. L., and H. A. Barker. 1949. Tracer
245. Joyce, B. K., and R. H. Himes. 1966. Formylte- experiments on the mechanism of uric acid
trahydrofolate synthetase. Initial velocity decomposition and acetic acid synthesis by
and product inhibition studies. J. Biol. Clostridium acidi-urici. J. Biol. Chem.
Chem. 241:5725-5731. 178:891-902.
246. Judewicz, N. D., E. M. De Robertis, Jr., and H. 260. Kating, H. 1962. Untersuchungen uber den
N. Torres. 1974. Control of uracil transport Stoffwechsel des Harnstoffs bei Mikroorgan-
by cyclic AMP in E. coli. FEBS Lett. 45:155- ismen. II. Die Verarbeitung des Harnstoffs
158. bei Endomycopsis vernalis und Torulopsis
247. Jurtshuk, P., Jr., R. N. Doetsch, and J. C. utilis. Biochem. Z. 335:366-381.
Shaw. 1958. Anaerobic purine dissimilation 261. Kearny, J. J., and R. D. Sagers. 1972. Formate
by washed suspensions of bovine rumen bac- dehydrogenase from Clostridium acidiurici.
teria. J. Dairy Sci. 41:190-202. J. Bacteriol. 109:152-161.
248. Kaltwasser, H. 1967. Bildung partikelgebunde- 262. Keller, H. 1950. Die Kultur der intrazellularen
ner Uricase bei Hydrogenomonas H16 und Symbionten von Periplaneta orientalis. Z.
anderen aeroben Bakterien. Zentrabl. Bak- Naturforsch. Teil B 5:269-273.
teriol. Parasitenkd. Infektionskr. Hyg. Abt. 263. Kelln, R. A., and R. A. J. Warren. 1974. Pyrim-
1 Orig. 205:87-88. idine metabolism in Pseudomonas acidovor-
249. Kaltwasser, H. 1967. Verwertung organischer ans. Can. J. Microbiol. 20:427-433.
Stickstoffverbitidungen durch Hydrogeno- 264. Kerr, C. T., and R. W. Miller..968. Dihydroor-
monas-Arten, p. 153-160. 2. Arbeitstagung otate-ubiquinone reductase complex of Esch-
Extraterrestrische Biophysik und Biologie erichia coli B. J. Biol. Chem. 243:2963-2968.
und Raumfahrtmedizin, Marburg/Lahn. 265. Kida, J. 1969. Studies on bacterial uricase. III.
250. Kaltwasser, H. 1968. Induktive Bildung parti- Influence of adenine on growth and uricase
kelgebundener Uricase bei Hydrogenomonas production ofBrevibacterium vitarumen var.
H16 und anderen aeroben Bakterien. Arch. uricum. J. Ferment. Technol. 47:617-622.
Mikrobiol. 60:160-171. 266. Kida, J., and M. Kunihisa. 1966. Studies on
251. Kaltwasser, H. 1968. Harnsaureabbau und bacterial uricase. I. Isolation of uricase pro-
Biosynthese der Enzyme Uricase, Glyoxylat- ducing bacteria and some culture conditions
carboligase und Urease bei Hydrogeno- for the production. J. Ferment. Technol.
monas H16. I. Bildung von Glyoxylatcarboli- 44:789-796.
gase und D-Glycerat-3-Dehydrogenase. 267. Kida, J., M. Kunihisa, and S. Shiraishi. 1966.
Arch. Mikrobiol. 64:71-84. Studies on bacterial uricase. II. Extraction of
252. Kaltwasser, H. 1969. Harnsaureabbau und uricase from bacterial cells and some proper-
Biosynthese der Enzyme Uricase, Glyoxylat- ties of crude enzymes. J. Ferment. Technol.
carboligase und Urease bei Hydrogeno- 44:797-804.
monas H16. II. Einfluss von Harnsaure, 268. Kirschner, L. B. 1967. Comparative physiol-
Fructose und Stickstoffmangel. Arch. Mikro- ogy: invertebrate excretory organs. Annu.
biol. 65:288-302. Rev. Physiol. 29:169-196.
253. Kaltwasser, H. 1971. Studies on the physiology 269. Kissel, P., M. Lamarche, and R. Royer. 1968.
of Bacillus fastidiosus. J. Bacteriol. 107:780- Modification of uricaemia and the excretion
786. of uric acid nitrogen by an enzyme of fungal
254. Kaltwasser, H., and J. Krimer. 1968. Verwer- origin. Nature (London) 217:72-74.
458 VOGELS AND VAN DER DRIYT BACTERIOL. RiV.
270. Klein, S. M., and R. D. Sagers. 1966. Glycine Acta 21:593-594.
metabolism. I. Properties of the system cata- 285. Kramer, J., and H. Kaltwasser. 1969. Verwer-
lyzing the exchange of bicarbonate with the tung von Pyrimidinderivaten durch Hydro-
carboxyl group of glycine in Peptococcus gly- genomonas facilis. II. Abbau von Thymin
cinophilus. J. Biol. Chem. 241:197-205. und Uracil durch Wildstamm und Mutanten.
271. Klein, S. M., and R. D. Sagers. 1966. Glycine Arch. Mikrobiol. 69:138-148.
metabolism. II. Kinetic and optical studies 286. Kramer, J., and H. Kaltwasser. 1969. Verwer-
on the glycine decarboxylase system from tung von Pyrimidinderivaten durch Hydro-
Peptococcus glycinophilus. J. Biol. Chem. genomonas facilis. I. Intermediarprodukte
241:206-209. und Enzyme des Cytosinabbaues. Arch. Mik-
272. Klein, S. M., and R. D. Sagers. 1967. Glycine robiol. 68:227-235.
metabolism. III. A flavin-linked dehydrogen- 287. Kramer, J., H. Kaltwasser, and H. G. Schlegel.
ase associated with the glycine cleavage sys- 1967. Die Bedeutung der Urease Repression

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


tem in Peptococcus glycinophilus. J. Biol. fur die taxonomische Klassifizierung von
Chem. 242:297-300. Bakterien. Zentralbl. Bakteriol. Parasi-
273. Klein, S. M., and R. D. Sagers. 1967. Glycine tenkd. Infektionskr. Hyg. Abt. 2. Orig.
metabolism. IV. Effect of borohydride reduc- 121:414-423.
tion on the pyridoxal phosphate-containing 288. Kream, J., and E. Chargaff. 1952. On the cyto-
glycine decarboxylase from Peptococcus gly- sine deaminase of yeast. J. Am. Chem. Soc.
cinophilus. J. Biol. Chem. 242:301-305. 74:5157-5160.
274. Klemperer, F., C. Scott, and S. Bagchi. 1967. 289. Krebs, H. A., and L. V. Eggleston. 1939. Bacte-
Uric acid oxidation by mycobacteria. Am. rial urea formation (Metabolism of Coryne-
Rev. Respir. Dis. 95:833-837. bacterium ureafaciens). Enzymologia 7:310-
275. Klemperer, F. W., H. C. Trimble, and A. Baird 320.
Hastings. 1938. The uricase of dogs, includ- 290. Krebs, H. A., and H. Weil. 1935. Investigation
ing the Dalmatian. J. Biol. Chem. 125:445- of the uricolytic enzymes (uricase, allanto-
449. inase, allantoicase), p. 497-509. Probl. Biol.
276. Kminek, M. 1936. Studies of the oxalogenic Med., Moscow. Taken from Chem. Zentralbl.
substances in sugar beets. I. Allantoin stud- 1938 (II):1428.
ies and determinations. Listy Cukrov. 291. Krenitsky, T. A., J. V. Tuttle, E. L. Cattau,
54:461-465. (Taken from Chem. Abstr. Jr., and P. Wang. 1974. Comparison of the
30:7899-7900, 1936.) distribution and electron acceptor specifici-
277. Kminek, M. 1936. Studies of the oxalogenic ties of xanthine oxidase and aldehyde oxi-
substances in sugar beets. II. A new nonsu- dase. Comp. Biochem. Physiol. B 49:687-
gar, oxamic acid isolated from sugar beets. 703.
Listy Cukrov. 54:469-479. (Taken from 292. Kurtzman, Jr., R. H., and S. Schwimmer.
Chem. Abstr. 30:7900, 1936.) 1971. Caffeine removal from growth media
278. Knutsen, G. 1972. Degradation of uracil by by microorganisms. Experientia 27:481-482.
synchronous cultures of Chlorella fusca. 293. Kuster, G., R. G. Shorter, B. Dawson, and G.
Biochim. Biophys. Acta 269:333-343. A. Hallenbeck. 1972. Uric acid metabolism
279. Kochi, H., and G. Kikuchi. 1974. Mechanism of in dalmatians and other dogs. Arch. Intern.
the reversible glycine cleavage reaction in Med. 129:492-496.
Arthrobacter globiformis. I. Purification and 294. Kikta, J., and P. Valenta. 1963. Ermittlung der
function of protein components required for Hydratationsgleichgewichtskonstanten der
the reaction. J. Biochem. (Tokyo) 75:1113- Glyoxalsaure und ihres Anions durch Oszil-
1127. lographie mit vorgegebenem Spannungsver-
280. Kondo, H., H. C. Friedmann, and B. Vennes- lauf. Collect. Czech. Chem. Commun.
land. 1960. Flavin changes accompanying 28:1593-1597.
adaptation of Zymobacterium oroticum to or- 295. Laboureur, P., and C. Langlois. 1967. Proper-
otate. J. Biol. Chem. 235:1533-1535. ties of a highly active fungal urate oxidase.
281. Kornberg, H. L., and A. M. Gotto. 1961. The C.R. Acad. Sci. 264:2244-2246.
metabolism of C2 compounds in micro-orga- 296. Laboureur, P., and C. Langlois. 1968. Urate
nisms. 6. Synthesis of cell constituents from oxydase d'Aspergillus flavus. I. Obtention,
glycollate by Pseudomonas sp. Biochem. J. purification, proprietes. Bull. Soc. Chim.
78:69-82. Biol. 50:811-825.
282. Koser, S. A. 1918. The employment of uric acid 297. Laboureur, P., and C. Langlois. 1968. Urate
synthetic medium for the differentiation of oxydase d'Aspergillus flavus. II.
B. coli and B. aerogenes. J. Infect. Dis. M6tabolisme, inhibitions, specificite. Bull.
23:377-379. Soc. Chim. Biol. 50:827-841.
283. Kossowicz, A. 1914. Zur Kenntnis der Assimi- 298. Lacey, J., and M. Goodfellow. 1975. A novel
lation von Kohlenstoff- und Stickstoffverbin- actinomycete from sugar-cane bagasse: Sac-
dungen durch Schimmelpilze. Biochem. Z. charopolyspora hirsuta gen. et sp. nov. J.
67:391-399. Gen. Microbiol. 88:75-85.
284. Krakow, G., and S. S. Barkulis. 1956. Conver- 299. Lahou, J. 1958. Metabolisme de la guanine-8-
sion of glyoxylate to hydroxypruvate by ex- 14C chez la levure. Biochim. Biophys. Acta
tracts of Escherichia coli. Biochim. Biophys. 27:371-377.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 459
300. Lara, F. J. S. 1952. On the decomposition of Rabinowitz. 1963. Studies on the chemical
pyrimidines by bacteria. I. Studies by means nature of clostridial ferredoxin. J. Biol.
of the technique of simultaneous adaptation. Chem. 238:3899-3913.
J. Bacteriol. 64:271-277. 318. Lubowe, I. I., and S. B. Mecca. 1964. A new
301. Lara, F. J. S. 1952. On the decomposition of series of novel allantoin complexes for topi-
pyrimidines by bacteria. II. Studies with cal use. Proc. Sci. Sect. Toilet Goods Assoc.
cell-free enzyme preparations. J. Bacteriol. 42:6-8.
64:279-285. 319. Lucke, H. 1931. The physiology and pathology
302. LaRue, T. A., and J. F. T. Spencer. 1968. The of human uric acid metabolism. VIII. The
utilization of purines and pyrimidines by uric acid content of the intestines and feces.
yeasts. Can. J. Microbiol. 14:79-86. Z. Gesamte Exp. Med. 76:180-187.
303. Laskowski, M. 1951. Allantoinase and allanto- 320. Lucke, H. 1931. The physiology and pathology
icase, p. 946-950. In J. B. Sumner and M. of human uric acid metabolism. IX. The im-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Myrback (ed.), The enzymes. Chemistry and portance of intestinal uric acid in the regula-
mechanism of action, vol. I, part 2. Academic tion of human endogenous uric acid metabo-
Press Inc., New York. lism. Z. Gesamte Exp. Med. 76:188-192.
304. Lassaigne, J. L. 1821. Nouvelles recherches sur 321. Lui, N. S. T., and 0. A. Roels. 1970. Nitrogen
la composition des eaux de l'allantoide et de metabolism of aquatic organisms. I. The as-
l'amnios de la vache. Ann. Chim. Phys. (2) similation and formation of urea in Ochro-
17:295-305. monas maihamensis. Arch. Biochem. Bio-
305. Leadbetter, E. R., and S. C. Holt. 1968. The phys. 139:269-277.
fine structure of Bacillus fastidiosus. J. Gen. 322. Lui, N. S. T., 0. A. Roels, M. E. Trout, and 0.
Microbiol. 52:299-307. R. Anderson. 1968. Subcellular distribution
306. Leboy, P. S., S. G. Cline, and R. L. Conner. of enzymes in Ochromonas malhamensis. J.
1964. Phosphate, purines, and pyrimidines Protozool. 15:536-542.
as excretory products of Tetrahymena. J. 323. Lund, P., and B. Magasanik. 1965. N-formi-
Protozool. 11:217-222. mino-L-glutamate formiminohydrolase of
307. Lee, K. W., and A. H. Roush. 1964. Allanto- Aerobacter aerogenes. J. Biol. Chem.
inase assays and their application to yeast 240:4316-4319.
and soybean allantoinases. Arch. Biochem. 324. MacKenzie, R. E., and J. C. Rabinowitz. 1971.
Biophys. 108:460-467. Cation-dependent reassociation of subunits
308. Leftley, J. W., and P. J. Syrett. 1973. Urease of N10-formyltetrahydrofolate synthetase
and ATP: urea amidolyase activity in unicel- from Clostridium acidi-urici and Clostrid-
lular algae. J. Gen. Microbiol. 77:109-115. ium cylindrosporum. J. Biol. Chem.
309. Lewis, J. B. 1967. Nitrogenous excretion in the 246:3731-3736.
tropical sea urchin Diadema antillarum phi- 325. Magasanik, B., and H. R. Bowser. 1955. The
lippi. Biol. Bull. 132:34-37. degradation of histidine by Aerobacter aero-
310. Lieberman, I., and A. Kornberg. 1953. Enzy- genes. J. Biol. Chem. 213:571-580.
matic synthesis and breakdown of orotic 326. Magill, J. M., and C. W. Magill. 1975. Purine
acid. Fed. Proc. 12:239-240. base transport in Neurospora crassa. J. Bac-
311. Lieberman, I., and A. Kornberg. 1953. En- teriol. 124:149-154.
zymic synthesis and breakdown of a pyrimi- 327. Mahler, H. R., H. M. Baum, and G. Hubscher.
dine, orotic acid. I. Dihydro-orotic dehydro- 1956. Enzymatic oxidation of urate. Science
genase. Biochim. Biophys. Acta 12:223-234. 124:705-708.
312. Lieberman, I., and A. Kornberg. 1954. Enzy- 328. Mahler, H. R., G. Habscher, and H. Baum.
matic synthesis and breakdown of a pyrimi- 1955. Studies on uricase. I. Preparation, pu-
dine, orotic acid. II. Dihydro-orotic acid, ur- rification, and properties of a cuproprotein.
eidosuccinic acid, and 5-carboxymethylhy- J. Biol. Chem. 216:625-641.
dantoin. J. Biol. Chem. 207:911-924. 329. Mahler, J. L. 1970. A new bacterial uricase for
313. Lieberman, I., and A. Kornberg. 1955. Enzy- uric acid determination. Anal. Biochem.
matic synthesis and breakdown of a pyrimi- 38:65-84.
dine, orotic acid. mI. Ureidosuccinase. J. 330. Maruyama, Y., and M. Alexander. 1962. Local-
Biol. Chem. 212:909-920. ization of enzymes in the mycelium and mi-
314. Liebert, F. 1909. The decomposition of uric acid croconidia of Fusarium oxysporum. J. Bacte-
by bacteria. Proc. K. Acad. Ned. Wet. 12: riol. 84:307-312.
54-64. 331. Masover, G. K., J. R. Benson, and L. Hayflick.
315. Ljungdahl, L. G., and H. G. Wood. 1969. Total 1974. Growth of T-strain mycoplasmas in me-
synthesis of acetate from CO2 by hetero- dium without added urea: effect of trace
trophic bacteria. Annu. Rev. Microbiol. amounts of urea and of a urease inhibitor. J.
23:515-538. Bacteriol. 117:765-774.
316. Lockwood, G. F., and R. G. Garrison. 1968. 332. McElroy, W. D., and H. K. Mitchell. 1946.
The possible role of uric acid in the ecology of Enzyme studies on a temperature sensitive
Histoplasma capsulatum. Mycopathol. My- mutant of Neurospora. Fed. Proc. 5:376-379.
col. Appl. 35:377-388. 333. McIllmurray, M. B., and J. Lascelles. 1970.
317. Lovenberg, W., B. B. Buchanan, and J. C. Anaerobiosis and the activity of enzymes of
460 VOGELS AND VAN DER DRIFT BACTERIOL. Rzv.
pyrimidine biosynthesis in Staphylococcus 349. Morris, J. L., and E. E. Ecker. 1924. Destruc-
aureus. J. Gen. Microbiol. 64:269-277. tion of uric acid by bacteria and molds. J.
334. Mead, G. C. 1974. Anaerobic utilization of uric Infect. Dis. 34:592-598.
acid by some group D streptococci. J. Gen. 350. Mosteller, R. D., and R. V. Goldstein. 1975.
Microbiol. 82:421-423. Unusual sensitivity ofEscherichia coli to ad-
335. Meiss, H. K., W. J. Brill, and B. Magasanik. enine or adenine plus histidine. J. Bacteriol.
1969. Genetic control of histidine degrada- 123:750-751.
tion in Salmonella typhimurium, strain LT- 351. Motokawa, Y., and G. Kikuchi. 1969. Glycine
2. J. Biol. Chem. 244:5382-5391. metabolism in rat liver mitochondria. II.
336. Meyer, E., and B. Wurtz. 1967. Recherches sur Methylene tetrahydrofolate as the direct one
le mecanisme des oxydations chez Pseudomo- carbon donor in the reaction of glycine syn-
nas fluorescens. III. Mise en evidence d'une thesis. J. Biochem. (Tokyo) 65:71-75.
activite dihydroorotate-dsshydrogenasique. 352. Motokawa, Y., and G. Kikuchi. 1969. Glycine

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


C. R. Soc. Biol. 161:2056-2059. metabolism by liver mitochondria. IV. Isola-
337. Miller, J. D. A., and G. E. Fogg. 1958. Studies tion and characterization of hydrogen carrier
on the growth of Xanthophyceae in pure cul- protein, an essential factor for glycine me-
ture. II. The relations of Monodus subterra- tabolism. Arch. Biochem. Biophys. 135:402-
neus to organic substances. Arch. Mikrobiol. 409.
30:1-16. 353. Motokawa, Y., and G. Kikuchi. 1974. Glycine
338. Miller, R. W., and C. T. Kerr. 1967. Particulate metabolism by rat liver mitochondria. VI.
dihydroorotate oxidase system from a pseu- Reconstitution of the reversible glycine
domonad. Linkage with the respiratory cleavage system with partially purified pro-
chain. Can. J. Biochem. 45:1283-1294. tein components. Arch. Biochem. Biophys.
339. Miller, R. W., and V. Massey. 1965. Dihydroor- 164:624-633.
otic dehydrogenase. I. Some properties of the 354. Moyed, H. S. 1964. Inhibition of the biosyn-
enzyme. J. Biol. Chem. 240:1453-1465. thesis of the pyrimidine portion of thiamine
340. Miller, R. W., and V. Massey. 1965. Dihydroor- by adenosine. J. Bacteriol. 88:1024-1029.
otic dehydrogenase. II. Oxidation and reduc- 355. Mtkller, M., J. F. Hogg, and C. de Duve. 1968.
tion of cytochrome c. J. Biol. Chem. 240: Distribution of tricarboxylic acid cycle en-
1466-1472. zymes and glyoxylate cycle enzymes between
341. Mimura, A., T. Akimoto, and R. Kodaira. mitochondria and peroxisomes in Tetrahy-
1969. Transformation of an organic com- mena pyriformis. J. Biol. Chem. 243:5385-
pound by microorganisms. Formation of 6,8- 5395.
dihydroxypurine. J. Ferment. Technol. 356. Muller, M., and K. M. Moller. 1969. Studies on
47:268-273. some enzymes of purine metabolism in the
342. Minnich, J. E. 1972. Excretion of urate salts by amoebae Chaos chaos and Amoeba proteus.
reptiles. Comp. Biochem. Physiol. A 41:535- C. R. Trav. Lab. Carlsberg 36:463-497.
549. 357. Muller, M., and K. M. Moller. 1969. Urate
343. Mitchell, N. B., and M. Levine. 1938. Nitrogen oxidase and its association with peroxisomes
availability as an aid in the differentiation of inAcanthamoeba sp. Eur. J. Biochem. 9:424-
bacteria in the coli-aerogenes group. J. Bac- 430.
teriol. 36:587-597. 358. Nakajima, Y., and G. H. Bourne. 1970. Histo-
344. Mitidieri, E., and 0. R. Affonso. 1965. Molyb- chemical studies on urate oxidase in several
denum requirement for bacterial xanthine mammals with special reference to uricolytic
dehydrogenase activity. Biochem. Biophys. ability of primates. Histochemie 22:20-24.
Acta 105:371-373. 359. Nason, A., K.-Y. Lee, S.-S. Pan, and R. H.
345. Molnar, J., and B. Prigai. 1973. Possible role Erickson. 1973. Evidence for a molybdenum
of adenosine deaminase in suboptimal cofactor common to all molybdenum enzymes
growth of the Bacillus anthracis adenine based on the in vitro assembly of assimila-
auxotroph mutant. Acta Microbiol. Acad. tory NADPH-nitrate reductase using the
Sci. Hung. 20:255-265. Neurospora mutant nit-1, p. 233-239. In
346. Moore, M. R., W. E. O'Brien, and L. G. Ljung- P.C.H. Mitchell (ed.), Proceedings of the
dahl. 1974. Purification and characterization First Conference on the Chemical Uses of
of nicotinamide adenine dinucleotide-de- Molybdenum. Climax Molybdenum Co. Ltd.,
pendent methylenetetrahydrofolate dehy- London.
drogenase from Clostridium formicoaceti- 360. Naylor, A.. W. 1970. Phylogenetic aspects of
cum. J. Biol. Chem. 249:5250-5253. nitrogen metabolism in the algae. Ann. N.Y.
347. Morgan, E. J., C. P. Stewart, and F. G. Hop- Acad. Sci. 175:511-523.
kins. 1922. On the anaerobic and aerobic oxi- 361. Needham, J. 1931. Chemical embryology, vol.
dation of xanthine and hypoxanthine by tis- 2, p. 615-1253. Cambridge University Press,
sues and by milk. Proc. R. Soc. London Ser. London.
B 94:109-131. 362. Needham, J. 1938. Contributions of chemical
348. Morlion, M., and A. Domnas. 1962. Over het physiology to the problem of reversibility in
assimilatiemechanisme en het gebruik van evolution. Biol. Rev. 13:224-251.
allantoine en allantoinezuur bij gist; Natu- 363. Neidhardt, F. C. 1963. Properties of a bacterial
urwet. Tijdschr. (Ghent) 44:100-121. mutant lacking amino acid control of RNA
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 461
synthesis. Biochim. Biophys. Acta 68:365- state kinetic investigation of the uricase re-
379. action mechanism. Arch. Biochem. Biophys.
364. Neidhardt, F. C., and B. Magasanik. 1957. Re- 163:359-366.
versal of the glucose inhibition of histidase 382. Pitts, 0. M., D. G. Priest, and W. W. Fish.
biosynthesis inAerobacter aerogenes. J. Bac- 1974. Uricase. Subunit composition and re-
teriol. 73:253-259. sistance to denaturants. Biochemistry
365. Ntmec, A. 1920. tJber Uricase in Samen Or- 13:888-892.
ganismus. Biochem. Z. 112:286-290. 383. Pizer, L. I. 1965. Glycine synthesis and metab-
366. Neubauer, C. 1868. Beitrage zur Harnanalyse. olism in Escherichia coli. J. Bacteriol. 89:
Z. Anal. Chem. 7:225-235. 1145-1150.
367. Nirmala, J., and K. S. Sastry. 1973. The effect 384. Polak, A., and M. Grenson. 1973. Evidence for
of thiourea on ureide metabolism in Neuro- a common transport system for cytosine, ade-
spora crassa. Biochem. J. 136:749-755. nine and hypoxanthine in Saccharomyces

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


368. Noren, B. 1955. Studies on myxobacteria. Ill. cerevisiae and Candida albicans. Eur. J. Bio-
Organic factors in nutrition. Bot. Not. chem. 32:276-282.
108:81-134. 385. Polkinghorne, M., and M. J. Hynes. 1975. Ef-
369. Nose, K., and K. Arima. 1968. Studies on bacte- fect of L-histidine on the catabolism of nitrog-
rial urate: oxygen oxidoreductase. II. Obser- enous compounds in Aspergillus nidulans. J.
vations concerning the properties and compo- Gen. Microbiol. 87:185-187.
nents of the active site. Biochim. Biophys. 386. Potts, W. T. W. 1965. Ammonia excretion in
Acta 151:63-69. Octopus dofleini. Comp. Biochem. Physiol.
370. Novoa, W. B., A. D. Winer, A. J. Glaid, and G. 14:339-355.
W. Schwert. 1959. Lactic dehydrogenase. V. 387. Potts, W. T. W. 1967. Excretion in the mol-
Inhibition by oxamate and by oxalate. J. luscs. Biol. Rev. 42:1-41.
Biol. Chem. 234:1143-1148. 388. Pourquie, J., and H. Heslot. 1971. Utilization
371. O'Donovan, G. A., and J. Neuhard. 1970. Py- and interconversions of purine derivatives in
rimidine metabolism in microorganisms. the fission yeast Schizosaccharomyces
Bacteriol. Rev. 34:278-343. pombe. Genet. Res. 18:33-44.
372. Oien, H. G., and L. D. Wright. 1971. Metabo- 389. Powles, M. A., P. A. Janssens, and D. Gil-
lism of imidazole by a pseudomonad. J. Bac- mour. 1972. Urea formation in the green veg-
teriol. 105:1229-1231. etable bug Nezara viridula. J. Insect Phys-
373. Olson, J. S., D. P. Ballou, G. Palmer, and V. iol. 18:2343-2358.
Massey. 1974. The reaction of xanthine oxi- 390. Pradhan, T. K., and E. G. Sander. 1973. Non-
dase with molecular oxygen. J. Biol. Chem. competitive inhibition by substituted sulfon-
249:4350-4362. amides of dihydroorotase from Zymobacter-
374. Olson, J. S., D. P. Ballou, G. Palmer, and V. ium oroticum. Life Sci. 13:1747-1752.
Massey. 1974. The mechanism of action of 391. Praetorius, E. 1948. The enzymatic conversion
xanthine oxidase. J. Biol. Chem. 249:4363- of uric acid. Spectrophotometric analy-
4382. sis. Biochim. Biophys. Acta 2:602-613.
375. Omura, H., Y. Osajima, and T. Tsukamoto. 392. Putrament, A., H. Baranowska, T. Bilifiski,
1966. Properties of urea dehydrogenase in and W. Prazmo. 1972. On the specificity of
tissues of higher plants. Enzymologia caffeine effects. Inhibition by caffeine of
31:129-154. RNA and protein synthesis in yeast and
376. Omura, H., Y. Osajima, R. Uchio, and Y. Nak- Escherichia coli. Mol. Gen. Genet. 118:373-
amura. 1969. Urea dehydrogenase of green 379.
algae. V. Enzymic activity requiring p-ami- 393. Rabinowitz, J. C. 1956. Purine fermentation by
nosalicylic acid, isonicotinic acid hydrazide, Clostridium cylindrosporum. III. 4-Amino-5-
a-ketoglutaric acid oxime, or y-glutamylhy- imidazolecarboxylic acid and 4-aminoimida-
droxamic acid as electron donor. Eiyo To zole. J. Biol. Chem. 218:175-187.
Shokuryo 22:468-472. 394. Rabinowitz, J. C. 1960. Folic acid, p. 185-252.
377. Page, 0. T., and M. C. Clark. 1964. Chromato- In P.D. Boyer, H. Lardy and K. Myrback
graphic analyses of purine breakdown by (ed.), The enzymes, vol. 2. Academic Press
Phytophthora infestans. Phytopathology Inc., New York.
54:1181-1186. 395. Rabinowitz, J. C. 1960. Factor B and other
378. Parish, R. W. 1972. Peroxisomes from the compounds related to vitamin B12 in pyruvic
Arum italicum appendix. Z. Pflanzenphysiol. acid-CO2 exchange. J. Biol. Chem. 235:PC50-
67:430-442. 51.
379. Piret, M. Cl., R. Crokaert, and J. Christophe. 396. Rabinowitz, J. C. 1963. Intermediates in pu-
1964. Le catabolisme r6ductif de l'uracile rine breakdown, p. 703-713. In S.P. Colowick
chez Torulopsis utilis. Arch. Int. Physiol. and N.O. Kaplan (ed.), Methods in enzymol-
Biochim. 72:256-266. ogy, vol. VI. Academic Press Inc., New York.
380. Pitts, 0. M., and D. G. Priest. 1973. Uricase 397. Rabinowitz, J. C., and H. A. Barker. 1956.
reaction intermediate. Mechanism of borate Purine fermentation by Clostridium cylin-
and hydroxide ion catalysis. Biochemistry drosporum. I. Tracer experiments on the
12:1358-1363. fermentation of guanine. J. Biol. Chem.
381. Pitts, 0. M., and D. G. Priest. 1974. A steady- 218:147-160.
462 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
398. Rabinowitz, J. C., and H. A. Barker. 1956. Biol. Chem. 236:1758-1763.
Purine fermentation by Clostridium cylin- 414. Razet, P. 1966. Les elements terminaux du
drosporum. II. Purine transformations. J. catabolisme azote chez les insectes. Ann~e
Biol. Chem. 218:161-173. Biol. 5:43-73.
399. Rabinowitz, J. C., and R. H. Himes. 1960. Folic 415. Reichert, U., R. Schmidt, and M. Foret. 1975.
acid coenzymes. Fed. Proc. 19:963-970. A possible mechanism of energy coupling in
400. Rabinowitz, J. C., and W. E. Pricer, Jr. 1956. purine transport of Saccharomyces cerevi-
Metabolism of formiminoglycine and glycine siae. FEBS Lett. 52:100-102.
by Clostridium acidi-urici. Fed. Proc. 15: 416. Reichert, U., and M. Winter. 1974. Uptake and
332-333. accumulation of purine bases by stationary
401. Rabinowitz, J. C., and W. E. Pricer, Jr. 1956. yeast cells pretreated with glucose. Biochim.
The enzymatic synthesis of N'0-formylte- Biophys. Acta 356:108-116.
trahydrofolic acid and its role in ATP forma- 417. Reinbothe, H. 1964. Urea formation from py-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


tion during formiminoglycine degradation. rimidines in fruit-bodies of higher basidi-
J. Am. Chem. Soc. 78:41764178. omycetes. Tetrahedron Lett. 37:2651-2657.
402. Rabinowitz, J. C., and W. E. Pricer, Jr. 1956. 418. Reinbothe, H., and K. Mothes. 1962. Urea,
Formimino-tetrahydrofolic acid and methen- ureides, and guanidines in plants. Annu.
yltetrahydrofolic acid as intermediates in Rev. Plant Physiol. 13:129-150.
the formation of N'0-formyltetrahydrofolic 419. Reinert, W. R., and G. A. Marzluf. 1975. Regu-
acid. J. Am. Chem. Soc. 78:5702-5704. lation of the catabolic enzymes in Neuro-
403. Rabinowitz, J. C., and W. E. Pricer, Jr. 1956. spora crassa. Arch. Biochem. Biophys.
Purine fermentation by Clostridium cylin- 166:565-574.
drosporum. IV. 4-Ureido-5-imidazolecarbox- 420. Reinert, W. R., and G. A. Marzluf. 1975. Ge-
ylic acid. J. Biol. Chem. 218:189-199. netic and metabolic control of the purine
404. Rabinowitz, J. C., and W. E. Pricer, Jr. 1956. catabolic enzymes of Neurospora crassa.
Purine fermentation by Clostridium cylin- Mol. Gen. Genet. 139:39-55.
drosporum. V. Formiminoglycine. J. Biol. 421. Remsen, C. C., E. J. Carpenter, and B. W.
Chem. 222:537-554. Schroeder. 1974. The role of urea in marine
405. Rabinowitz, J. C., and W. E. Pricer, Jr. 1957. microbial ecology, p. 286-304. In R. R. Col-
Formation, isolation and properties of 5-for- well and R. Y. Morita (ed.), Effect of the
miminotetrahydrofolic acid. Fed. Proc. ocean environment on microbial activity.
16:236. University Park Press, Baltimore.
406. Rabinowitz, J. C., and W. E. Pricer, Jr. 1957. 422. Remy, C. N., and S. H. Love. 1968. Induction of
An enzymatic method for the determination adenosine deaminase in Escherichia coli. J.
of formic acid. J. Biol. Chem. 229:321-328. Bacteriol. 96:76-85.
407. Rabinowitz, J. C., and W. E. Pricer, Jr. 1962. 423. Revel, H. R. B., and B. Magasanik. 1958. The
Formyltetrahydrofolate synthetase. I. Isola- enzymatic degradation of urocanic acid. J.
tion and crystallization of the enzyme. J. Biol. Chem. 233:930-935.
Biol. Chem. 237:2898-2902. 424. Reynolds, E. S., I. Lieberman, and A. Korn-
408. Rabinowitz, J. C., and W. E. Pricer, Jr. 1965. berg. 1955. The metabolism of orotic acid in
Metabolism of formiminoglycine. Degrada- aerobic bacteria. J. Bacteriol. 69:250-255.
tion by whole cells. J. Biol. Chem. 240:1696- 425. Robinson, J. R., S. M. Klein, and R. D. Sagers.
1700. 1973. Glycine metabolism. Lipoic acid as the
409. Radin, N. S., and H. A. Barker. 1953. Enzymic prosthetic group in the electron transfer pro-
reactions in purine decomposition by prepa- tein P2 from Peptococcus glycinophilus. J.
rations of Clostridium acidi-urici. Proc. Biol. Chem. 248:5319-5323.
Natl. Acad. Sci. U.S.A. 39:1196-1204. 426. Rogers, L. J. 1966. The nitrogen excretion of
410. Raeburn, S., and J. C. Rabinowitz. 1965. Pyru- Chelodina longicollis under conditions of hy-
vate synthesis by a partically purified en- dration and dehydration. Comp. Biochem.
zyme from Clostridium acidi-urici. Bio- Physiol. 18:249-260.
chem. Biophys. Res. Commun. 18:303-307. 427. Rogosa, M., and F. S. Bishop. 1964. The genus
411. Rajagopalan, K. V., V. Aleman, P. Handler, Veillonella. II. Nutritional studies. J. Bacte-
W. Heinen, G. Palmer, and H. Beinert. 1962. riol. 87:574-580.
Electron paramagnetic resonance studies of 428. Rohrscheidt, E., Zs. Tarnok, and I. Tarnok.
iron reduction and semiquinone formation in 1970. Purin- und pyrimidinabbauende En-
metalloflavoproteins. Biochem. Biophys. zyme in Mycobakterien und Nocardien. Zen-
Res. Commun. 8:220-226. tralbl. Bakteriol. Parasitenkd. Infektionskr.
412. Ramasastri, B. V., and R. L. Blakley. 1963. Hyg. Abt. 1 Orig. 215:550-554.
Optical rotations of the diastereoisomers of 429. Roon, R. J., J. Hampshire, and B. Levensberg.
dl, L-methylenetetrahydrofolate. Biochem. 1972. Urea amidolyase. The involvement of
Biophys. Res. Commun. 12:478-482. biotin in urea cleavage. J. Biol. Chem.
413. Rao, D. R., and D. M. Greenberg. 1961. Studies 247:7539-7545.
on the enzymic decomposition of urocanic 430. Roon, R. J., and B. Levenberg. 1968. An aden-
acid. IV. Purification and properties of 4(5)- osine triphosphate-dependent avidin-sensi-
imidazolone-5(4)-propionic acid hydrolase. J. tive enzymatic cleavage of urea in yeast and
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 463
green algae. J. Biol. Chem. 243:5213-5215. teria. II. Studies on the regulation system of
431. Roon, R. J., and B. Levenberg. 1970. C02 fixa- the degradation of nucleotides in Pseudomo-
tion and the involvement of allophanate in nas oleovorans. J. Ferment. Technol. 49:488-
the biotin-enzyme-catalyzed cleavage of 498.
urea. J. Biol. Chem. 245:4593-4595. 449. Salanitro, J. P., I. G. Blake, and P. A. Muir-
432. Roon, R. J., and B. Levenberg. 1972. Urea head. 1974. Studies on the cecal microflora of
amidolyase. I. Properties of the enzyme from commercial broiler chickens. Appl. Micro-
Candida utilis. J. Biol. Chem. 247:4107- biol. 28:439-447.
4113. 450. Sander, E. G., and M. J. Heeb. 1971. Purifica-
433. Rouf, M. A., and R. F. Lomprey, Jr. 1968. tion and properties of dihydroorotase from
Degradation of uric acid by certain aerobic Escherichia coli B. Biochim. Biophys. Acta
bacteria. J. Bacteriol. 96:617-622. 227:442-452.
451. Sander, E. G., L. D. Wright, and D. B. Mc-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


434. Roush, A. H. 1954. Yeast adenase. Arch. Bio-
chem. Biophys. 50:510-512. Cormick. 1965. Evidence for function of a
435. Roush, A. H., and A. J. Domnas. 1956. Induced metal ion in the activity of dihydroorotase
biosynthesis of uricase in yeast. Science from Zymobacterium oroticum. J. Biol.
124:125-126. Chem. 240:3628-3630.
436. Roush, A. H., L. M. Questiaux, and A. J. Dom- 452. Sato, T., H. Kochi, Y. Motokawa, H. Kawa-
nas. 1959. The active transport and metabo- saki, and G. Kikuchi. 1969. Glycine metabo-
lism of purines in the yeast, Candida utilis. lism by rat liver mitochondria. I. Synthesis
J. Cell. Comp. Physiol. 54:275-286. of two molecules of glycine from one mole-
437. Roush, A. H., and M. Saeed. 1960. Adenine cule each of serine, bicarbonate and ammo-
metabolism in Saccharomyces cerevisiae. Ad- nia. J. Biochem. (Tokyo) 65:63-70.
enase from bakers' yeast. Biochem. Biophys. 453. Sato, T., H. Kochi, N. Sato, and G. Kikuchi.
Res. Commun. 2:43-47. 1969. Glycine metabolism by rat liver mito-
438. Roush, A. H., and T. R. Shieh. 1963. Purifica- chondria. III. The glycine cleavage and the
tion and properties of induced uricase from exchange of carboxyl carbon of glycine with
the yeast Candida utilis. Fed. Proc. 22:292. bicarbonate. J. Biochem. (Tokyo) 65:77-83.
439. Ruban, E. L. 1958. Nitrogen metabolism in 454. Scazzocchio, C., and A. J. Darlington. 1968.
Nitrosomonas europaea. Mikrobiologiya The induction repression of the enzymes of
27:536-640. purine breakdown in Aspergillus nidulans.
440. Rinjerse, V. F. M. 1973. Regulation of allan- Biochim. Biophys. Acta 166:557-568.
toin metabolism in the genus Pseudomonas. 455. Scazzocchio, C., F. B. Holl, and A. I. Foguel-
Ph.D. thesis, Univ. of Nijmegen, Nijmegen. man. 1973. The genetic control of molybdo-
441. Sagers, R. D., and J. V. Beck. 1955. Exchange flavoproteins in Aspergillus nidulans. Allo-
of radioactive bicarbonate with the carboxyl purinol-resistant mutants constitutive for
group of pyruvate by Clostridium acidiurici. xanthine-dehydrogenase. Eur. J. Biochem.
Arch. Biochem. Biophys. 54:249-250. 36:428-445.
442. Sagers, R. D., and J. V. Beck. 1956. Studies on 456. Schardinger, F. 1902. Ueber das Verhalten der
the formation of formate, glycine, serine, py- Kuhmilch gegen Methylenblau und seine
ruvate, and acetate from purines by Clostrid- Verwendung zur Unterscheidung von unge-
ium acidi-urici. J. Bacteriol. 72:199-208. kochter und gekochter Milch. Z. Unters.
443. Sagers, R. D., M. Benziman, and I. C. Gunsa- Nahr. Genussm. Gebrauchsgegenstaende 5:
lus. 1961. Acetate formation in Clostridium 1113-1121.
acidi-urici: acetokinase. J. Bacteriol. 82:233- 457. Scheele, C. W. 1776. K. Vet. Acad. Handlingar
238. (Stockholm) 37:327-331.
444. Sagers, R. D., M. Benziman, and S. M. Klein. 458. Schefferle, H. E. 1965. The decomposition of
1963. Failure of arsenate to uncouple the uric acid in built up poultry litter. J. Appl.
phosphotransacetylase system in Clostrid- Bacteriol. 28:412-420.
ium acidiurici. J. Bacteriol. 86:978-984. 459. Schein, A. H., and A. S. Kunin. 1969. Urate
445. Sagers, R. D., and J. E. Carter. 1971. L-serine oxidation by cupric ion (Cu++). FEBS Lett.
dehydratase (Clostridium acidi-urici), p. 2:339-341.
351-356. In S.P. Colowick and N.O. Kaplan 460. Schieper, A. 1848. Ueber die Oxydation der
(ed.), Methods in enzymology, vol. XVII, Harnsaure vermittelst Kaliumeisencyanid.
part B. Academic Press Inc., New York. Ann. Chem. Pharm. 67:214-235.
446. Sagers, R. D., and I. C. Gunsalus. 1958. Gly- 461. Schittenhelm, A. 1905. Uber die Harnsaure-
cine cleavage and one-carbon transfer reac- bildung und die Harnsaurezersetzung in den
tions in Clostridium acidi-urici and Diplococ- Auszugen der Rinderorgane. Z. Physiol.
cus glycinophilus. Bacteriol. Proc., p. 119. Chem. 46:121-151.
447. Sagers, R. D., and I. C. Gunsalus. 1961. Inter- 462. Schlee, D., W. Fritsche, and H. Reinbothe.
mediary metabolism of Diplococcus glycino- 1966. Purinstoffwechsel in Candida guillier-
philus. I. Glycine cleavage and one-carbon mondii (Cast) Lang G. Flora Abt. A 156:
interconversions. J. Bacteriol. 81:541-549. 390-403.
448. Sakai, T., T. Watanabe, and I. Chibata. 1971. 463. Schlee, D., H. Reinbothe, and W. Fritsche.
Metabolism of pyrimidine nucleotides in bac- 1968. Der Einfluss von Eisen auf den Purin-
464 VOGELS AND VAN DER DRIFT BACTEIZIOL. REV.
stoffwechsel und die Riboflavinbildung von 478. Sin, I. L. 1975. Purification and properties of
Candida guilliermondii (Cast) Lang G. Z. xanthine dehydrogenase from Pseudomonas
Allg. Mikrobiol. 8:127-138. acidovorans. Biochim. Biophys. Acta 410:12-
464. Schuler, W., and W. Reindel. 1933. Die Uriko- 20.
lyse. III. Z. Physiol. Chem. 215:258-266. 479. Singh, R. 1968. Evidence for the presence of
465. Schulman, M., R. K. Ghambeer, L. G. Ljung- allantoicase in germinating peanuts. Phyto-
dahl, and H. G. Wood. 1973. Total synthesis chemistry 7:1503-1508.
of acetate from CO2. VII. Evidence with Clos- 480. Smith, A. E., and E. W. Yamada. 1971. Dihy-
tridium thermoaceticum that the carboxyl of drouracil dehydrogenase of rat liver. Separa-
acetate is derived from the carboxyl of pyru- tion of hydrogenase and dehydrogenase ac-
vate by transcarboxylation and not by fixa- tivities. J. Biol. Chem. 246:3610-3617.
tion of CO2. J. Biol. Chem. 248:6255-6261. 481. Smith, R. F. 1970. Nitrogen requirements and
466. Schulman, M., D. Parker, L. G. Ljungdahl, uricolytic activity of cutaneous bacteria.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


and H. G. Wood. 1972. Total syntheis of ace- Appl. Microbiol. 19:643-648.
tate from CO2. V. Determination by mass 482. Smith, S. T., K. V. Rajagopalan, and P. Han-
analysis of the different types of acetate dler. 1967. Purification and properties of
formed from 13CO2 by heterotrophic bacteria. xanthine dehydrogenase from Micrococcus
J. Bacteriol. 109:633-644. lactilyticus. J. Biol. Chem. 242:4108-4117.
467. Schunck, E. 1867. On oxalurate of ammonia as 483. Soldo, A. T., and W. J. van Wagtendonk. 1961.
a constituent of human urine. Proc. R. Soc. Nitrogen metabolism in Paramecium aure-
London 16:140-143. lia. J. Protozool. 8:41-55.
468. Schwimmer, S., R. H. Kurtzman, Jr., and E. 484. Spitzer. 1899. Zur Physiologie der Harnsaure.
Heftmann. 1971. Caffeine metabolism by Jahresber. Sches. Ges. Vaterl. Cult. Abt. I
Penicillium roqueforti. Arch. Biochem. Bio- 77:55-57.
phys. 147:109-113. 485. Stabenau, H., and H. Beevers. 1974. Isolation
469. Scott, J. M., and J. C. Rabinowitz. 1967. The and characterization of microbodies from the
association-dissociation of formyltetrahydro- alga Chlorogonium elongatum. Plant Phys-
folate synthetase and its relation to monova- iol. 53:866-869.
lent cation activation of catalytic activity. 486. Stapp, C. 1920. Botanische Untersuchungen ei-
Biochem. Biophys. Res. Commun. 29:418- niger neuer Bakterienspezies, welche mit
423. reiner Harnsaure oder Hippursaure als al-
470. Seaman, G. R. 1963. Metabolism of purines by leinigen organischem Nahrstoffauskommen.
extracts of Tetrahymena. J. Protozool. 10:87- Zentralbl. Bakteriol. Parasitenkd. Infek-
91. tionskr. Abt. 2 Orig. 51:1-71.
471. Sebald, M. 1962. etude sur les bact6ries 487. Stekol, J., and L. R. Cerecedo. 1933. Studies on
anaerobies Gram-negatives asporulees. the physiology of pyrimidines. VII. The me-
Ph.D. thesis, Univ. of Paris, Paris. tabolism of isobarbituric acid in man. J.
472. Semler, B. L., R. C. Hodson, S. K. Williams II, Biol. Chem. 100:653-656.
and S. H. Howell. 1975. The induction of 488. Stewart, D. J. 1965. The urease activity of flu-
allophanate lyase during the vegetative cell orescent pseudomonads. J. Gen. Microbiol.
cycle in the light-synchronized cultures of 41:169-174.
Chlamydomonas reinhardi. Biochem. Bio- 489. Stokvis, B. J. 1859. Bijdragen tot de physiolo-
phys. Acta 399:71-78. gie van het acidum uricum. Ned. Tijdschr.
473. Shaffer, P. M., C.-A. Hsu, and M. T. Abbott. Geneeskd. 3:587-607.
1975. Metabolism of pyrimidine deoxyribo- 490. Sukhenko, F. T., and E. S. Podgainaia. 1958.
nucleosides in Neurospora crassa. J. Bacte- Transformations of uric acid by some fungi.
riol. 121:648-655. Biochemistry (USSR) 23:175-182.
474. Sharma, M. L., and M. C. Neveu. 1971. Studies 491. Sylvester, C. J., and R. N. Costilow. 1964. Nu-
on the role of ornithine cycle and purine tritional requirements of Bacillus popilliae.
catabolism in urea biosynthesis in fresh-wa- J. Bacteriol. 87:114-119.
ter crayfish, Orconectus rusticus. Comp. Bio- 492. Tabor, H., and A. H. Mehler. 1954. Isolation of
chem. Physiol. B 40:863-870. N-formyl-L-glutamic acid as an intermediate
475. Shavlovsky, G. M., G. P. Ksheminskaya, and in the enzymatic degradation of L-histidine.
R. A. Kuznetsova. 1971. Candida guillier- J. Biol. Chem. 210:559-568.
mondii mutants with blocked purine catabo- 493. Tabor, H., and L. Wyngarden. 1959. The enzy-
lism and their flavinogenic activity. Mikro- matic formation of formiminotetrahydrofolic
biologiya 40:1070-1076. acid, 5, 10-methenyltetrahydrofolic acid, and
476. Shavlovsky, G. M., and A. A. Sibirny. 1973. 10-formyltetrahydrofolic acid in the metabo-
Regulation of uric acid uptake in the yeast lism of formiminoglutamic acid. J. Biol.
Pichia guilliernondii. FEBS Lett. 31:313- Chem. 234:1830-1846.
316. 494. Tacquet, A., F. Tison, P. Roos, and B. De-
477. Shorer, J., I. Zelmanowicz, and I. Barash. vulder. 1967. Activite amidasique des myco-
1972. Utilization and metabolism of urea bacteries. Technique qualitative nouvelle
during spore germination by Geotrichum d'etude en milieu de culture solide. Ann.
candidum. Phytochemistry 11:595-605. Inst. Pasteur 112:378-383.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 465
495. Taha, E.-E. M., L. Storck-Krieg, and W. adults of the euryhaline bull shark Carcha-
Franke. 1955. Purinoxydierende Fermente rhinus leucas from freshwater and saline en-
aus Schimmelpilzen. IV. Cber die Xanthin- vironments. Physiol. Zool. 46:29-42.
dehydrase der Schimmelpilze. Arch. Mikro- 511. Tigier, H., and S. Grisolia. 1965. Induction of
biol. 23:67-68. carbamyl-P specific oxamate transcarbamyl-
496. Taupitz-Stahn, E. L. 1974. Bacterial uricase ase by parabanic acid in a Streptococcus. Bio-
effects in vitro. Aerztl. Lab. 20:8-12. chem. Biophys. Res. Commun. 19:209-214.
497. Taylor, W. H., and G. D. Novelli. 1964. En- 512. Tintemann, and H. Reinbothe. 1973. Harn-
zymes of the pyrimidine pathway in Esche- stoffassimilation in der Hefe Candida guil-
richia coli. I. Synthesis by cells and sphero- liermondii durch ATP: urea-amidolyase.
plasts. J. Bacteriol. 88:99-104. Arch. Biol. Med. Germ. 30:317-322.
498. Taylor, W. H., and M. L. Taylor. 1964. En- 513. Townsend, D., and G. F. Lata. 1969. Purifica-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


zymes of the pyrimidine pathway in Esche- tion of urate oxidase; a sex dependent en-
richia coli. II. Intracellular localization and zyme from rat liver. Arch. Biochem. Bio-
properties of dihydroorotic dehydrogenase. phys. 135:166-172.
J. Bacteriol. 88:105-110. 514. Tracey, M. V. 1955. Urea and ureides, p. 119-
499. Taylor, W. H., M. L. Taylor, and D. F. Eames. 141. In K. Paech and M. V. Tracey (ed.),
1966. Two fimctionally different dihydroor- Moderne Methoden der Pflanzenanalyse,
otic dehydrogenases in bacteria. J. Bacteriol. vol. IV. Springer-Verlag, Berlin.
91:2251-2256. 515. Truscoe, R., and V. Williams. 1965. Effect of
500. Taylor, M. L., W. H. Taylor, D. F. Eames, and inhibitors on activity of ox-kidney urate oxi-
C. D. Taylor. 1971. Biosynthetic dihydrooro- dase. Biochim. Biophys. Acta 105:292-300.
tate dehydrogenase from Lactobacillus bul- 516. Truszkowski, R. 1930. Uricase and its action.
garicus. J. Bacteriol. 105:1015-1027. II. Bacterial nature of the action of uricolytic
501. Thannhauser, S. J., and G. Dorfminller. 1918. extracts and dialysates. Biochem. J. 24:1340-
Experimentelle Studien uber den Nuclein- 1348.
stoffwechsel. V. Ober die Aufspaltung des 517. Trijbels, F., and G. D. Vogels. 1966. Degrada-
Purinringes durch Bacterien der men- tion of allantoin by Pseudomonas acidovor-
schlichen Darmflora. Z. Physiol. Chem. ans. Biochim. Biophys. Acta 113:292-301.
102:148-159. 518. Trijbels, F., and G. D. Vogels. 1966. Allanto-
502. Thauer, R. K. 1972. C02-reduction to formate icase and ureidoglycolase in Pseudomonas
by NADPH. The initial step in the total syn- and Penicillium species. Biochim. Biophys.
thesis of acetate from CO2 in Clostridium Acta 118:387-395.
thermoaceticum. FEBS Lett. 27:111-115. 519. Trijbels, F., and G. D. Vogels. 1967. Allantoate
503. Thauer, R. K. 1973. C02 reduction to formate and ureidoglycolate degradation by Pseu-
in Clostridium acidi-urici. J. Bacteriol. domonas aeruginosa. Biochim. Biophys.
114:443 444. Acta 132:115-126.
504. Thauer, R. K., G. Fuchs, and K. Jungermann. 520. Trijbels, F., and G. D. Vogels. 1969. Catabo-
1974. Reduced ferredoxin: C02 oxidoreduc- lism of allantoate and ureidoglycolate in
tase from Clostridium pasteurianum: its role Rana esculenta. Comp. Biochem. Physiol.
in formate metabolism. J. Bacteriol. 30:359-365.
118:758-760. 521. Tsai, C. S., and B. Axelrod. 1965. Catabolism
505. Thauer, R. K., G. Fuchs, U. Schnitker, and K. of pyrimidines in rape seedlings. Plant Phys-
Jungermann. 1973. C02 reductase from Clos- iol. 40:39-44.
tridium pasteurianum: molybdenum depend- 522. Ukada, S., and B. Vennesland. 1962. Proper-
ence of synthesis and inactivation by cyanide. ties of triphosphopyridine nucleotide-linked
FEBS Lett. 38:45-48. dihydroorotic dehydrogenase. J. Biol. Chem.
506. Thauer, R. K., B. Kaufer, and G. Fuchs. 1975. 237:2018-2024.
The active species of 'CO2' utilized by re- 523. Uyeda, K., and J. C. Rabinowitz. 1965. Metab-
duced ferredoxin: C02 oxidoreductase from olism of formiminoglycine. Glycine formimi-
Clostridiumpasteurianum. Eur. J. Biochem. notransferase. J. Biol. Chem. 240:1701-1710.
55:111-117. 524. Uyeda, K., and J. C. Rabinowitz. 1967. Metab-
507. Thauer, R. K., B. Kaufer, and P. Scherer. olism of formiminoglycine. Formiminote-
1975. The active species of "CO2" utilized in trahydrofolate cyclodeaminase. J. Biol.
ferredoxin-linked carboxylation reactions. Chem. 242:24-31.
Arch. Microbiol. 104:237-240. 525. Uyeda, K., and J. C. Rabinowitz. 1967. En-
508. Thompson, J. F., and A.-M. E. Muenster. 1971. zymes of clostridial purine fermentation.
Separation of the Chlorella ATP: urea Methylenetetrahydrofolate dehydrogenase.
amido-lyase into two components. Biochem. J. Biol. Chem. 242:4378-4385.
Biophys. Res. Commun. 43:1049-1055. 526. Uyeda, K., and J. C. Rabinowitz. 1968. En-
509. Thornsberry, C., and R. F. Wiseman. 1960. zymes of the clostridial purine fermentation.
Uric acid utilization by a rumen coliform Serine hydroxymethyltransferase. Arch.
bacterium. Bacteriol. Proc., p. 32. Biochem. Biophys. 123:271-278.
510. Thorson, T. B., C. M. Cowan, and D. E. Wat- 527. Uyeda, K., and J. C. Rabinowitz. 1971. Pyru-
son. 1973. Body fluid solutes of juveniles and vate-ferredoxin oxidoreductase. III. Purifica-
466 VOGELS AND VAN DER DRIFT BACTRIUOL. REV.
tion and properties of the enzyme. J. Biol. 545. Van der Drift, C., F. E. de Windt, and G. D.
Chem. 246:3111-3119. Vogels. 1970. Allantoate hydrolysis by allan-
528. Uyeda, K., and J. C. Rabinowitz. 1971. Pyru- toate amidohydrolase. Arch. Biochem. Bio-
vate-ferredoxin oxidoreductase. IV. Studies phys. 136:273-279.
on the reaction mechanism. J. Biol. Chem. 546. Van der Drift, C., and H. C. J. Ketelaars. 1974.
246:3120-3125. Carnosinase: its presence in Pseudomonas
529. Valentine, R. C. 1964. Bacterial ferredoxin. aeruginosa. Antonie van Leeuwenhoek J.
Bacteriol. Rev. 28:497-517. Microbiol. Serol. 40:377-384.
530. Valentine, R. C., R. Bojanowski, E. Gaudy, 547. Van der Drift, C., P. E. M. van Helvoort, and
and R. S. Wolfe. 1962. Mechanism of allan- G. D. Vogels. 1971. S-Ureidoglycolate dehy-
toin fermentation. J. Biol. Chem. 237:2271- drogenase: purification and properties. Arch.
2277. Biochem. Biophys. 145:465-469.

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


531. Valentine, R. C., R. Bojanowski, and R. S. 548. Van der Drift, C., and G. D. Vogels. 1967.
Wolfe. 1961. The allantoin fermentation. Activation and inactivation of allantoate
Bacteriol. Proc., p. 184. amidohydrolase. Biochim. Biophys. Acta
532. Valentine, R. C., W. J. Brill, and R. D. Sagers. 139:162-168.
1963. Ferredoxin linked DPN reduction by 549. Van der Drift, C., and G. D. Vogels. 1969.
pyruvate in extracts of Clostridium acidi- Stereospecificity of allantoin degradation in
urici. Biochem. Biophys. Res. Commun. Streptococcus allantoicus. Experientia 25:
12:315-319. 477.
533. Valentine, R. C., H. Drucker, and R. S. Wolfe. 550. Van der Drift, C., and G. D. Vogels. 1969.
1964. Glyoxylate fermentation by Streptococ- Allantoate amidohydrolase. I. pH- and an-
cus allantoicus. J. Bacteriol. 87:241-246. ion-dependent activation. Enzymologia
534. Valentine, R. C., R. L. Jackson, and R. S. 36:269-277.
Wolfe. 1962. Role of ferredoxin in hydrogen 551. Van der Drift, C., and G. D. Vogels. 1969.
metabolism of Micrococcus lactilyticus. Bio- Allantoate amidohydrolase. II. Inactivation
chem. Biophys. Res. Commun. 7:453-456. and instability. Enzymologia 36:278-286.
535. Valentine, R. C., and R. S. Wolfe. 1960. Phos- 552. Van der Drift, C., and G. D. Vogels. 1970.
phorolysis of carbamyl oxamic acid. Effect of metal and hydrogen ions on the
Biochim. Biophys. Acta 45:389-391. activity and stability of allantoicase.
536. Valentine, R. C., and R. S. Wolfe. 1960. Bio- Biochim. Biophys. Acta 198:339-352.
synthesis of carbamyl oxamic acid. Biochem. 553. Van der Drift, L., G. D. Vogels, and C. van der
Biophys. Res. Commun. 2:384-387. Drift. 1975. Allantoin racemase: a new en-
537. Valentine, R. C., and R. S. Wolfe. 1961. Glyox- zyme from Pseudomonas species. Biochim.
ylurea. Biochem. Biophys. Res. Commun. Biophys. Acta 391:240-248.
5:305-308. 554. Van Hartingsveldt, J., and A. H. Stouthamer.
538. Valentine, R. C., and R. S. Wolfe. 1961. Phos- 1973. Mapping and characterization of mu-
phate-dependent degradation of urea. Na- tants ofPseudomonas aeruginosa affected in
ture (London) 191:925-926. nitrate respiration in aerobic and anaerobic
539. Valentine, R. C., and R. S. Wolfe. 1961. The growth. J. Gen. Microbiol. 74:97-106.
allantoin fermentation,. p. 334. In N. M. 555. Varel, V. H., and M. P. Bryant. 1974. Nutri-
Sissakian (ed.), Proceedings of the 5th Inter- tional features of Bacteroides fragilis subsp.
national Congress on Biochemistry, vol. IX, fragilis. Appl. Microbiol. 28:251-257.
Moscow 1960. Pergamon Press Ltd., London. 556. Varel, V. H., M. P. Bryant, L. V. Holdeman,
540. Valentine, R. C., and R. S. Wolfe. 1963. Role of and W. E. C. Moore. 1974. Isolation of ureo-
ferredoxin in the metabolism of molecular lytic Peptostreptococcus productus from feces
hydrogen. J. Bacteriol. 85:1114-1120. using defined medium; failure of common
541. Van Baalen, C. 1965. The photooxidation of urease tests. Appl. Microbiol. 28:594-599.
uric acid by Anacystis nidulans. Plant'Phys- 557. Villela, G. G. 1965. Dehydrogenases of purine
iol. 40:368-371. metabolism in Tetrahymena pyriformis.
542. Van Baalen, C., and J. E. Marler. 1963. Char- Proc. Soc. Exp. Biol. Med. 118:834-838.
acteristics of marine blue-green algae with 558. Villela, G. G., 0. R. Affonso, and E. Mitidieri.
uric acid as nitrogen source. J. Gen. Micro- 1955. Xanthine oxidase irn Lactobacillus
biol. 32:457-463. casei. Arch. Biochem. Biophys. 59:532-533.
543. Van de Poll, K. W., A. A. G. Verwey, and V. V. 559. Villeret, S. 1955. Sur la presence des enzymes
Koningsberger. 1968. Repression and de- des ureides glyoxyliques chez les Algues
repression of allantoinase synthesis in proto- d'eau douce. C.R. Acad. Sci. 241:90-92.
plasts of Saccharomyces carlsbergensis. I. 560. Villeret, S. 1958. Recherches sur la presence
Characteristics of the enzyme and introduc- des enzymes ureides glyoxyliques chez les
tory experiments on the derepression of its Algues marines. C.R. Acad. Sci. 246:1452-
synthesis. Proc. K. Ned. Akad. Wet. Ser. B. 1454.
71:344-358. 561. Vitez, V. 1962. Das Allantoin und dessen Deri-
544. Van der Drift, C. 1968. The effect of bivalent vate vom Standpunkt der dermatologischen
cations on some enzymes of the allantoin und kosmetologischen Anwendung. Riechst.
metabolism. Ph.D. thesis, Univ. of Nijme- Aromen 1962:1-6.
gen, Nijmegen. 562. Vitols, M. J., V. N. Shaposhnikov, and Yu. P.
VOL. 40, 1976 DEGRADATION OF PURINES AND PYRIMIDINES 467
Shvachkin. 1967. A new path of orotic acid from uracil oxidation. J. Biol. Chem.
catabolism in microorganisms. Doki. Akad. 194:785-791.
Nauk. SSR 174:1202-1204. (Taken from ab- 581. Waslien, C. I., D. H. Calloway, and S. Margen.
stract no. 79858, Chem. Abstr. 67:7515-7516 1968. Uric acid production of men fed graded
[1967].) amounts of egg protein and yeast nucleic
563. Vizdalova, M., E. Janovska, and V. D. Zhes- acid. Am. J. Clin. Nutr. 21:892-897.
tyanikov. 1971. Effect of dark-repair inhibi- 582. Watanabe, Y. 1971. Formation of uricase by
tors on the survival of Escherichia coli B Streptomyces. III. Effect of incubation with
under different postirradiation conditions. or without purines. Agric. Biol. Chem.
Int. J. Radiat. Biol. 20:49-59. 35:2008-2014.
564. Vogels, G. D. 1961. Intermediates in anaerobic 583. Watanabe, Y., and J. Fukumoto. 1970. Studies
allantoin degradation by bacteria. Biochem. on the formation of uricase by Streptomyces.
Z. 334:457-461. II. The induced formation of uricase by the

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


565. Vogels, G. D. 1963. On the microbial metabo- resting cells. Agric. Biol. Chem. 34:1625-
lism of allantoin. Ph.D. thesis, Institute of 1632.
Technology, Delft. 584. Watanabe, Y., and T. Ohe. 1972. Oxidation of
566. Vogels, G. D. 1966. Reversible activation of hypoxanthine to uric acid by Streptomyces.
allantoate amidohydrolase by acid-pretreat- Agric. Biol. Chem. 36:785-792.
ment and other properties of the enzyme. 585. Welch, W. H., D. H. Buttlaire, R. T. Hersh,
Biochim. Biophys. Acta 113:277-291. and R. H. Himes. 1971. The subunit struc-
567. Vogels, G. D. 1969. Specificity of binding sub- ture of formyltetrahydrofolate synthetase.
sites of allantoicase. Biochim. Biophys. Acta Biochim. Biophys. Acta 236:599-611.
185:186-197. 586. Wheeler, C. G. 1866. Notiz uber die Einwir-
568. Vogels, G. D. 1969. Stereospecificity in the al- kung von Mangansuperoxyd auf Harnsaure.
lantoin metabolism. Antonie van Leeuwen- Z. Chem. 9:746-747.
hoek J. Microbiol. Serol. 35:236-238. 587. Whiteley, H. R. 1952. The fermentation of pu-
569. Vogels, G. D., F. E. de Windt, and W. Bassie. rines by Micrococcus aerogenes. J. Bacteriol.
1969. Hydrolysis and racemization of allan- 63:163-175.
toin. Rec. Trav. Chim. Pays-Bas 88:940-950. 588. Whiteley, H. R., and H. C. Douglas. 1951. The
570. Vogels, G. D., F. Trijbels, and A. Uffink. 1966. fermentation of purines by Micrococcus lac-
Allantoinases from bacterial, plant, and ani- tilyticus. J. Bacteriol. 61:605-616.
mal sources. I. Purification and enzymic 589. Whiteley, H. R., and E. J. Ordal. 1956. The
properties. Biochim. Biophys. Acta 122:482- production of hydrogen by hypoxanthine oxi-
496. dation, p. 521-538. In W. D. McElroy and
571. Vogels, G. D., and C. van der Drift. 1966. Al- B. H. Glass (ed.), Symposion on inorganic
lantoinases from bacterial, plant, and ani- nitrogen metabolism. The John Hopkins
mal sources. II. Effect of bivalent cations and Press Inc., Baltimore.
reducing substances on the enzymic activity. 590. Whiteley, H. R., M. J. Osborn, and F. M.
Biochim. Biophys. Acta 122:497-509. Huennekens. 1959. Purification and proper-
572. Vogels, G. D., and C. van der Drift. 1969. Hy- ties of the formate-activating enzyme from
drolysis of allantoate. Rec. Trav. Chim. Micrococcus aerogenes. J. Biol. Chem. 234:
Pays-Bas 88:951-957. 1538-1543.
573. Vogels, G. D., and C. van der Drift. 1970. Dif- 591. Whiteley, H. R., and C. A. Woolfolk. 1962.
ferential analyses of glyoxylate derivatives. Ferredoxin-dependent reactions in Micrococ-
Anal. Biochem. 33:143-157. cus lactilyticus. Biochem. Biophys. Res.
574. Von Gorup-Besanez, E. 1859. Ueber die Ein- Commun. 9:517-522.
wirkung des Ozons auf organische Verbin- 592. Whitney, P. A., and T. G. Cooper. 1972. Urea
dungen. Ann. Chem. Pharm. 110:86-107. carboxylase and allophanate hydrolase: two
575. Wachsman, J. T., and H. A. Barker. 1954. components of a multienzyme complex in
Characterization of an orotic acid fermenting Saccharomyces cerevisiae. Biochem. Bio-
bacterium, Zymobacterium oroticum, nov. phys. Res. Commun. 49:45-51.
gen., nov. spec. J. Bacteriol. 68:400-404. 593. Whitney, P. A., and T. G. Cooper. 1972. Urea
576. Wachsman, J. T., and H. A. Barker. 1955. The carboxylase and allophanate hydrolase. Two
accumulation of formamide during the fer- components of adenosine triphosphate: urea
mentation of histidine by Clostridium tetano- amido-lyase in Saccharomyces cerevisiae. J.
morphum. J. Bacteriol. 69:83-89. Biol. Chem. 247:1349-1353.
577. Wallach, D. P., and S. Grisolia. 1957. The pu- 594. Whitney, P. A., T. G. Cooper, and B. Magasa-
rification and properties of hydropyrimidine nik. 1973. The induction of urea carboxylase
hydrase. J. Biol. Chem. 226:277-288. and allophanate hydrolase in Saccharomyces
578. Wang, T. P., and J. 0. Lampen. 1951. Bacterial cerevisiae. J. Biol. Chem. 248:6203-6209.
oxidation of pyrimidines. Fed. Proc. 10:267. 595. Wickner, R. B., and H. Tabor. 1972. N-formi-
579. Wang, T. P., and J. 0. Lampen. 1952. Metabo- mino-L-glutamate iminohydrolase from his-
lism of pyrimidines by a soil bacterium. J. tidine-adapted Pseudomonas. Purification
Biol. Chem. 194:775-783. and properties. J. Biol. Chem. 247:1605-
580. Wang, T. P., and J. 0. Lampen. 1952. Uracil 1609.
oxidase and the isolation of barbituric acid 596. Wiechowski, W. 1907. Die Produkte der fer-
468 VOGELS AND VAN DER DRIFT BACTERIOL. REV.
mentativen Harnsaurezersetzung durch properties. J. Biol. Chem. 245:3167-3178.
Tierische Organe. Beitr. Chem. Physiol. Pa- 606. Wright, B. E. 1955. Poly-glutamyl pteridine
thol. 9:295-310. coenzymes. J. Am. Chem. Soc. 77:3930-3932.
597. Wiener, H. 1899. Ueber Zersetzung und Bil- 607. Wright, B. E. 1956. The r6le of polyglutamyl
dung der Harnsaure im Thierkorper. Arch. pteridine coenzymes in serine metabolism.
Exp. Pathol. Pharmakol. 42:375-398. II. A comparison of various pteridine deriva-
598. Wohler, F. 1853. Gahrung des Allantoins. tives. J. Biol. Chem. 219:873-883.
Ann. Chem. Pharm. 88:100-101. 608. Wright, L. D., C. S. Miller, H. R. Skeggs, J. W.
599. Wohler, F., and J. Liebig. 1838. Untersuchun- Huff, L. L. Weed, and D. W. Wilson. 1951.
gen uber die Natur der Harnsaure. Ann. Biological precursors of the pyrimidines. J.
Pharm. 26:241-340. Am. Chem. Soc. 73:1898-1899.
600. Wolf, F. T. 1955. Nutrition and metabolism of 609. Wu, C.-H., E. J. Eisenbaum, and E. T. Gaudy.
the tobacco-wilt Fusarium. Bull. Torrey Bot. 1970. Enzymatic degradation of ureidogly-

Downloaded from http://mmbr.asm.org/ on December 4, 2019 by guest


Club 82:343-354. cine by Pseudomonas acidovorans. Biochem.
601. Wood, R. C., and E. Steers. 1959. The purine Biophys. Res. Comun. 39:976-982.
metabolism of Staphylococcus aureus. J. 610. Yates, R. A., and A. B. Pardee. 1956. Pyrimi-
Bacteriol. 77:760-765. dine biosynthesis in Escherichia coli. J. Biol.
602. Woodward, V. W., K. D. Munkres, and Y. Chem. 221:743-756.
Suyama. 1957. Uracil metabolism in Neuro- 611. Yeh, Y.-C., and D. M. Greenberg. 1965. Purifi-
spora crassa. Experientia 13:484-486. cation and properties of N5,N10-methylenete-
603. Woolfolk, C. A. 1975. Metabolism of N-methyl- trahydrofolate dehydrogenase of calf thy-
purines by a Pseudomonas putida strain iso- mus. Biochim. Biophys. Acta 105:279-291.
lated by enrichment on caffeine as the sole 612. Yoshimura, Y., T. Yokokawa, and N. Suna-
source of carbon and nitrogen. J. Bacteriol. hara. 1973. Microbial production of uricase.
123:1088-1106. Jpn. patent 73.18,473. Abstract no. 90519.
604. Woolfolk, C. A., E. J. Ordal, and H. R. White- Chem. Abstr. 79(15):303 (1973).
ley. 1959. The reduction of arsenate and 613. Young, E. G., and W. W. Hawkins. 1944. The
other inorganic anions by Micrococcus lacti- decomposition of allantoin by intestinal bac-
lyticus. Bacteriol. Proc., p. 116-117. teria. J. Bacteriol. 47:351-353.
605. Woolfolk, C. A., B. S. Woolfolk, and H. R. 614. Young, E. G., C. C. MacPherson, H. P. Went-
Whiteley. 1970. 2-Oxypurine dehydrogenase worth, and W. W. Hawkins. 1944. The esti-
from Micrococcus aerogenes. I. Isolation, mation of allantoin in blood. J. Biol. Chem.
specificity and some chemical and physical 152:245-253.
ERRATUM
Degradation of Purines and Pyrimidines by Microorganisms
G. D. VOGELS AND C. VAN DER DRIFf
Department of Microbiology, Faculty of Science, University of Nijmegen, Nijmegen, The Netherlands

Volume 40, no. 2, p. 406, column 1, first 3 lines: move to p. 405, top of column 1.
Page 418, Table 6, opposite "Mucor boidin" under column heading "Uric acid": insert "+".
Page 447, Table 15, under column heading "Presence of cytosine deaminase" opposite "Salmo-
nella typhimurium": insert "+". Under column heading "Presence of cytosine deaminase," oppo-
site "Veillonella alcalescens (Micrococcus lactilyticus)": remove "+".

963

You might also like