<<

Pediat. Res. 8: 724-734 (1 974)

A Review: Biological and Clinical Aspects of

RODNEY L. LEVINE, NICHOLAS J. HOOGENRAAD, AND NORMAN KRETCHMER(' ) Division of Developmental , Department of Pediatrics, Stanford University Medial Center, Stanford, California, USA

Interest in pyrimidine has stemmed or the of ), ATP, aspartate. and largely from the importance of these as components in the form of phosphoribosyl pyr~phos~hate.A of nucleic . Regulation of pyrimidine biosynthesis has reutilization ("salvage")-. mechanism is also available to cells. been most extensively investigated in micro- in whereby preformed pyrimidine bases or can be which cell division is effectively controlled by regulation of converted to the nucleotide by the addition of ribose the activity of the of the de novo pathway (77, 11 0). or phosphate. The relation between pyrimidine biosynthesis and cellular proliferation in has become clearer with the DE NOVO MECHANISM accumulation of a considerable body of evidence which indicates that there is a close correlation between the activity De novo biosynthesis of uridylic required six enzymatic of enzymes involved in pyrimidine biosynthesis and the rate of steps (Fig. 1). In procaryotes one supplies the cellular proliferation. carbamyl phosphate required for both pyrimidine nucleotide The de novo pyrimidine biosynthetic pathway and the and biosynthesis. Consequently, aspartate trans- cycle both utilize carbamyl phosphate, and important nitrogen carbamylase is the first enzyme unique to the de novo carrier. The biosynthesis of pyrimidine and pyrimidine pathway and an important locus for control of the are connected by a common intermediate, phosphoribosyl pathway (77). (PRPP). In addition, pyrimidine nucleotides In contrast, in ureotelic vertebrates, there are two carbamyl act as and phospholipid carriers and are important in phosphate synthetases. The first, carbamyl phosphate synthe- lipopolysaccharide biosynthesis. The importance of these tase I, is located in the inner membrane of the interrelations is most forcibly emphasized by certain clinical of the and also appears to be present in small quantities in phenomena which will be discussed later. and small intestine (82). The second, carbamyl This paper will attempt to review the broad biologic aspects phosphate syntlietase 11, has a more ubiquitous distribution of pyrimidine nucleotide biosynthesis. A detailed considera- and was first demonstrated to be present in Ehrlich ascites tion of de novo pyrimidine biosynthesis in eucaryotic cells is cells (58, 59) and in hematopoietic mouse (75, 143). available in the recent review by Jones (81). Pyrimidine This enzyme was originally difficult to detect because of its biosynthesis in procaryotes has been reviewed in depth by relatively low activity and high lability. However, the O'Donovan and Neuhard (1 10). discovery that the enzyme could be stabilized by its , -ATP (59), and by dimethyl sulfoxide and PATHWAYS OF PYRIMIDINE NUCLEOTIDE BIOSYNTHESIS (144, 146), made it possible to demonstrate that the enzyme was widely distributed in mammalian tissues (74, 128, 144, In cells from multicellular organisms, pyrimidine nucleotide 163). biosynthesis can proceed by two mechanisms. De novo The two synthetases have different properties which lends biosynthesis of uridylic acid proceeds from simple components support to the concept that they have distinct roles in such as dioxide, a source of nitrogen (either supplying carbamyl phosphate for either arginine biosynthesis

CO, + Mg-ATP + GLUTAMINE c0i$2+-:e+ CARBAMYL- P

ASPARTATE

tronscorbamylose

DIHYDRpOROTATE. dihydroytoseCARBAMYLASPARTATE

orotate phosphoribosyl tronsferose r MONOPHOSPHATE I / \* PRPP 2 Pi orotidylate decorboxy lose 4 UMP Fig. 1. Pathway for de novo biosynthesis of pyrimidine nucleotides in eucaryotic cells. PRPP: phosphoribosyl pyrophosphate. 7 24 PYRIMIDINE METABOLISM 725

or for uridylic acid biosynthesis (80). The mitochondria1 be effectively channeled into de novo pyrimidine biosynthesis carbamyl phosphate synthetase I can utilize ammonia as a (36, 80). source of nitrogen and is activated by N-acetyl glutamate, It has become evident recently that the conversion of which also stimulates urea biosynthesis in liver slices. orotate to uridylate may be an important site of regulation in Carbamyl phosphate synthetase 11, found in the , de novo pyrimidine biosynthesis. substantially inhibits utilizes glutamine as its normal nitrogen source as indicated by the incorporation of (' c)aspartate (1 17) and ' ~0~ (70) the K, values for glutamine and ammonia (144). In addition, into uridine nucleotides in hepatoma cells. Hoogenraad and glutamine analogs inhibit this enzyme but do not affect Lee (70) showed that the effects of uridine were caused by an carbamyl phosphate synthetase I (59, 60, 163). The role of effect on the level of activity of orotate phosphoribosyl carbamyl phosphate synthetase I1 in de novo pyrimidine , even though uridine or its nucleotides did not biosynthesis is indicated, not only by its general distribution in directly inhibit this enzyme. In addition, it appears that the most tissues, but also by the factors that control its activity. availability of PRPP may be crucial in the regulation of de UTP, an end of the de novo pyrimidine pathway, is a novo pyrimidine biosynthesis, inasmuch as orotate accumu- feedback inhibitor of carbamyl phosphate synthetase I1 (74, lates in cultures of mammalian cells to which small amounts of 94, 143), apparently acting in an allosteric fashion (94). have been added (73), and the toxic effects of adenine Phosphoribosyl pyrophosphate, a substrate required for both (73, 102) may be removed by supplementation of cells with purine and pyrimidine biosynthesis, enhances the activity of uridine (73), showing that the toxicity is caused by pyrimidine carbamyl phosphate synthetase I1 when added at low starvation. concentrations (132, 145, 147). The activity of orotidylate decarboxylase also can be The second enzyme of the do novo pathway is aspartate regulated by the pyrimidine nucleotides, UMP and CMP (3, 15, transcarbamylase (EC. 2.1.3.2). In fungi this enzyme is part of 16, 33). However, it is doubtful that this is an important a multifunctional complex which also contains carbamyl regulatory mechanism, because the Ki for inhibition by these phosphate synthetase I1 (8 1). These two enzymes are also in nucleotides is high and the levels of UMP and CMP are usually close physical association in mammalian tissues (71, 80). In extremely low (3, 1 17). addition, Shoaf and Jones (130-132) found that the third enzyme of the de novo pyrimidine pathway, REUTILIZATION OF PYRIMIDINE NUCLEOSIDES (EC. 3.5.2.3), is also present in the complex. Pyrimidine nucleosides from exogenous sources and from The aspartate transcarbamylase activity of the mammalian endogenous breakdown of nucleic acids are available for complex, unlike the enzyme from many procaryotic species pyrimidine nucleotide biosynthesis through the mediation of a and , is not sensitive to feedback regulation by number of enzymes which have collectively been called the nucleotides (35, 7 1, 120). "salvage pathway" or "reutilization pathway." The enzymes Dihydro-orotase activity is only marginally affected by of this pathway (Fig. 2) found in mammalian tissues are pyrimidine compounds, with the exception of , uridine (EC. 2.4.2.3), uridine (EC. which substantially inhibits the enzyme (19, 58). Dihydro- 2.7.1.48), and kinase (EC. 2.4.2.4). orotic acid (EC. 1.3.3.1), the next enzyme in catalyzes reversibly the reaction the pathway, is possibly the least studied of the de novo between and ribose 1-phosphate to form uridine and pyrimidine enzymes. The electron acceptor for the conversion inorganic phosphate. The enzyme is widely distributed in ani- of dihydro-orotic acid to orotic acid has not been character- mal tissues (23, 86). catalyzes the conversion ized for the mammalian enzyme. This enzyme is apparently of uridine to UMP. The name is somewhat limiting because the associated with the cell membranes since Shoaf and Jones enzyme will also convert to CMP (134). Uridine (132) showed that, in contrast to the other enzymes required kinase is required for the conversion of the antineoplastic for de novo pyrimidine nucleotide biosynthesis, the majority agent 5- to the active nucleotide, and resistance to of the dihydro-orotate dehydrogenase activity from Ehrlich this drug occurs when the enzyme becomes modified (135). In ascites cells sediments with the nuclei and the sequence of reactions leading from uridine to UTP, the fraction. The terminal two enzymes of the de novo pyrimidine conversion of uridine to UMP is rate limiting and sensitive to inhibition by UTP and CTP (1). The of pathway also exist as a complex in mammals. Appel (2) found that orotate phosphoribosyl transferase and orotidylate de- cytidine is also inhibited by UTP and CTP. carboxylase copurified when these enzymes were isolated from is also widely distributed in tissues as evidenced by the widespread use of radioactively labeled bovine brain. The two enzymes have also been copurified from gland and erythrocytes (63, 83) and they cosediment thymidine to measure DNA biosynthesis. As is the case with uridine kinase (90), thymidine kinase appears to be present in in a gradient when extracted from Ehrlich ascites cells fetal and adult liver in different forms (22, 87), and its activity (13 1, 132). The complex from thymus gland can be separated is subject to product inhibition by dTTP and dCTP (22). on gel, but when separated, the transferase activity may be completely lost (83). INTERCONVERSIONS OF PYRIMIDINE NUCLEOTIDES Genetic evidence from with congenital is consistent with the concept that orotate phos- Uridylic acid is converted into the range of pyrimidine phoribosyl transferase and orotidylate decarboxylase form as nucleotides required for mammalian metabolism by a series of enzyme complex. In all but one of the cases reported, only enzymes, many of which appear to be under complex very low levels of both of the enzymes were detected (12,136). The one exception was the case described by Fox et al. (47), uridine URlDlNE Pi URACIL + RIBOSE-I-P + in which a very low orotidylate decarboxylase level was phosphorylase ' accompanied by an elevated orotate phosphoribosyl trans- ferase. URlDlNE uridine UMP OR + Mg- ATP > OR + Mg-ADP The presumed advantage to the cell of enzyme complexes in CYTlDlNE kinase CMP the de novo pathway lies in the physical separation of thymidine pyrimidine and arginine-urea biosynthesis. It has been shown DEOXYTHYMIDINE + Mg -ATP >dTMP + Mg-ADP that, in Neurospora, the complex existing between carbamyl phosphate synthetase I1 and aspartate transcarbamylase en- Fig. 2. Reutilization pathway for the conversion of preformed ables the carbamyl phosphate synthesized in the to pyrimidine nucleosides into nucleotides. 726 LEVINE, HOOGENRAAD, AND KRETCHMER

d~hydrouracil metabolic control. These enzymes have been extensively studied in micro-organisms (23, 1 lo), and research with the dehydrogenase mammalian enzymes indicates that many of the controls are similar to those found in micro-organisms (86, 137). Figure 3 dihydropyrirnidinase UREIDOPROPIONATE summarizes the reactions involved in the interconversions of DlHY DROURACIL pyrimidine nucleotides. ureidopropionase UREIDOPROPIONATE .p-+ C02 + NH3 PYRIMIDINE NUCLEOTIDES IN Fig. 4. Pathway for the degradation of uracil into p-alanine and AND BIOSYNTHESIS . The same enzymes also catalyze the degradation of . The importance of pyrimidine nucleotides in is exemplified by the role of UDP- as a requires NADP for the reduction of uracil or thymine (56, substrate for synthetase (68) and of UDP- as 153). This enzyme is rate limiting in the degradative pathway a substrate for galactose transferase (4). The rapidity with of (27), and its activity is inversely proportional which glucogen pools are turned over and the extent of to the growth rate of liver cells and hepatomas (155). biosynthesis in lactating mammary glands support the quanti- The (EC. 3.5.2.2) of liver is capable of tative importance of uridine nucleotides in the cellular hydrolyzation of both dihydrouracil and metabolism of . Synthesis of cell membranes also requires pyrimidine nucle- (1 53). The carbamyl 0-amino acids formed in this reaction are otides as carriers of lipopolysaccharide precursors. For exam- further degraded to carbon dioxide and ammonia by ureido- propionase (EC. 3.5.1.6), which is specific for the carbamyl ple, cytidine nucleotides are required in the de novo biosynthe- sis of , ethanolamine, and glycerolphosphates. 0-amino acids derived from uracil and thymine (28). The rate of pyrimidine breakdown appears to vary inversely The biosynthesis of phosphatides apparently takes place in the with the rate of growth in mammalian cells. For example, the endoplasmic reticulum of mammalian cells, whereas the activities of enzymes in this pathway are low in liver of cydidine nucleotides are produced in the cytosol (103), the newborn (42) and increase with age, so that the ability site of the de novo pyrimidine pathway. Pyrimidine nucleo- of adult liver to degrade pyrimidine bases far exceeds its tides are also required in the biosynthesis of glycolipids, ability to utilize them for biosynthesis (1 18). As a , and free which are found on result of this degradative capacity, label from tritiated mammalian cell surfaces (1 14, 16 1). thymidine or is incorporated into lipid of adult Although it is clear that pyrimidine nucleotides have liver more rapidly than into DNA (1 19, 127). widespread functions in cellular metabolism, it is not known whether there are separate controls that regulate the supply of pyrimidine nucleotides for carbohydrate and . INTERRELATION OF UREA AND PYRIMIDINE PATHWAYS: It would seem unlikely that the biosynthesis of pyrimidine AND OROTIC ACIDURIA nucleotides is regulated solely by cell division, as metabolic processes such as glycogen biosynthesis require that there be a Carbamyl phosphate is an initial reactant in both urea and considerable supply of pyrimidine nucleotides even in the de novo pyrimidine biosynthesis. As discussed above, there are absence of cellular division. Interference with this supply of two distinct carbamyl phosphate synthetases. The mitochon- pyrimidine nucleotides can lead to pathologic changes in drial synthetase subserves arginine and urea biosynthesis tissues (see below). whereas the nonmitochondrial synthetase provides carbamyl phosphate for pyrimidine biosynthesis (79, 80). With the DEGRADATION OF PYRIMIDINES detection of two synthetases there was no basis to assume that urea and pyrimidine biosynthesis were closely linked. How- Pyrimidines are degraded in the liver primarily to carbon ever, recent evidence suggests that the two carbamyl phos- dioxide and 0-alanine. The degradative route for pyrimidine phate pools are not always completely compartmentalized and nucleotides proceeds via uracil or thymine. These two that carbamyl phosphate produced in liver mitochondria may pyrimidine bases share a common degradative pathway which become available for utilization in de novo pyrimidine utilizes the same enzymes. The degradation of biosynthesis. Natale and Tremblay (107) showed that intact therefore requires prior to uracil which is isolated mitochondria from rat liver may release carbamyl accomplished by cytidine (EC. 3.5.4.5). This phosphate which is presumably available for pyrimidine enzyme has been found in a number of animal tissues (26, 34, biosynthesis or other reactions (109). 148). Hyperammonemia occurs whenever there is a genetic defect In animals degradation of pyrimidine bases proceeds by in one of the enzymes (32). If the enzymatic defect three enzymatic steps as shown in Figure 4. Dihydrouracil also causes an accumulation of carbamyl phosphate, such as dehydrogenase (EC. 1.3.1.2) has been isolated from liver and with a deficiencv of transcarbamvlase (EC. 2.1.3.3).-2 the patient may excrete orotic acid, uridine, and other pyrimidine-containing-. compounds (129). Thus, orotic aciduria can occur both in hereditary orotic aciduria (1 36) because of a block in de novo pyrimidine biosynthesis at the orotate phosphoribosyl transferase and orotidylate decarboxylase steps, and in urea cycle defects with h~peramrnonemi~(5Sj. m- UDP- U;P The of orotic acid in urea cycle defects presumably occurs because mitochondria1 carbamyl phosphate which I cannot be utilized for urea biosynthesis becomes available for extramitochondrial pyrimidine biosynthesis. Thus, Goldstein et al. (55) found that excretion of orotic acid in parallels the blood ammonia concentration in a patient with congenital hyperammonemia due to ornithine transcarbamylase defi- Fig. 3. Reactions involved in the interconversions of pyrimidine ciency (Fig. 5). nucleotides. In the last few years total parenteral nutrition ("hyperali- METABOLISM 727

utilization by other organs via the reutilization pathway. COMMENCE PERITONEAL DIALYSIS Evidence for this key role of the liver as an exporter of J. is accumulating, but investigations on pyrimidine export are limited. Nevertheless, available evidence indicates -OROTIC ACID that the liver synthesizes pyrimidine nucleosides de novo for export to peripheral tissues. Nonhepatic tissues can incor- porate blood-borne nucleosides via the reutilization pathway, although they may also synthesize pyrimidine nucleotides de novo. As mentioned previously, de novo pyrimidine biosyn- thesis occurs in dividing cells derived from a number of different tissues. The major techniques employed by investigators to infer whether a given tissue utilized the de novo or reutilization pathway have been the determination of tissue enzyme activities and measurement of the incorporation of labeled precursors into nucleic acids. The difficulty of drawing conclusions from these studies is demonstrated by measure- ments made with bone marrow. Studies on nucleic acid turnover in rat marrow indicated that 80% of thymine was

123 4 5678 91011 1213141516 incorporated via the reutilization pathway (40). Bone marrow DAY OF SAMPLING makes a considerable amount of nucleic acid available for reutilization through the extrusion of reticulocyte nuclei (14), Fig. 5. Correlation of orotic acid in urine and ammonia in plasma in a and it has been shown that at least 35% of the thymine male infant with a deficiency of ornithine transcarbamylase. This figure liberated by bone marrow is reutilized by the marrow itself is reproduced from Goldstein et al. (55) with permission of the authors (41). In spite of the apparent importance of the reutilization and publisher. pathway, marrow has relatively high levels of activity of enzymes from the de novo pyrimidine biosynthetic pathway, mentation") has been introduced as a means of supplying all which has led to the conclusion that this pathway could be the nutritional requirements of patients. This method of quantitatively more important than the reutilization pathway. feeding is widely used with infants, but it was shown that Based on their incorporation studies, Hogans et al. (69) hyperammonemia may be a complication of total parenteral concluded that the brain normally utilizes preformed pyrimi- nutrition (78). Amino acids are usually supplied by a solution dine precursors. They isolated surgically all organs below the of casein or fibrin hydrolysate which may contain 36,000 pg diaphragm from the circulation, eliminating hepatic participa- ammonia in each 100 ml solution. This concentration of tion. Uridine was then found to be far more effective than ammonia would give rise to only 100 mg/kg/24 hr of urea, an orotic acid in labeling brain RNA of the rat. In contrast, amount which even a premature infant should be able to Michaelis and Mandel (104) followed the incorporation of synthesize (78, 108). Furthermore, Heird and associates (66) labeled precursors when injected directly into the cerebro- have reported hyperammonemia in infants receiving total spinal fluid. In this case there was effective labeling by orotic parenteral nutrition from a synthetic solution of amino acids acid as well as uridine. Developmental patterns for enzymes in which the ammonia content was negligible. It may be, required for pyrimidine nucleotide biosynthesis in rat brain therefore, that hyperammonemia associated with total par- suggest that de novo pyrimidine biosynthesis is important in enteral nutrition is secondary to a metabolic imbalance which immature animals, whereas adult brains rely primarily on the inhibits urea biosynthesis. reutilization pathway (156). The site of the potential metabolic block in the urea cycle is The clearest indication of the role of the liver in providing not known, but we have found the orotic acid content of urine pyrimidine nucleosides for the brain comes from a beautiful from infants on total parenteral nutrition was usually elevated series of experiments by Geiger (53) and Geiger and Yamasaki during periods of hyperamrnonemia. The presence of orotic (54). Using isolated, perfused cat brain they demonstrated that aciduria suggests that there is a block in urea biosynthesis at the presence of cytidine and uridine in the blood is essential the ornithine transcarbamylase step. It seems likely that the for maintaining normal brain functions. Isolated, perfused cat enzyme is intact in such patients, so the block is presumed to brains show an increasing degree of anaerobic , be caused either by the presence of an inhibitor of the enzyme develop lactic acidosis and hypoglycemia, and die within 1 hr or by a deficiency of ornithine, a substrate of the enzyme. of isolation. of the liver into the circulation corrects In support of the latter suggestion, it has recently been the changes in carbohydrate metabolism and allows survival reported that growing rats fed a diet low in arginine developed for 2-3 hr. Geiger and Yamasaki (54) showed that inclusion hyperammonemia and had orotic aciduria because of a of the liver was unnecessary if small amounts of cytidine and lowered availability of ornithine for urea biosynthesis (105). uridine were included in the perfusate. Addition of these In addition, Heird et al. (66) have reported that the infusion of nucleosides apparently facilitated the transfer of glucose into additional arginine relieves rapidly the hyperammonemia the brain, which restored the brain and glucose associated with total parenteral nutrition. concentrations to normal and allowed survival for 5 hr. The following pyrimidine nucleosides appear to be required for lipid metabolism as well. Cytidine prevented or restored the DE NOVO VERSUS REUTILIZATION PATHWAYS: POSSIBLE loss of phosphatides, presumably by providing CDP carriers for ROLE OF LIVER lipid biosynthesis. Uridine prevented or restored the loss of The nucleotide requirements of different tissues may be galactolipids, presumably by providing UDP carriers for sugar supplied by de novo pyrimidine and purine biosynthesis or by biosynthesis. The ability of cytidine and uridine to replace the utilization of preformed nucleosides carried in the blood liver provides evidence implicating the liver in export of stream. Murray (1 06) has recently reviewed evidence support- pyrimidine nucleosides to the brain. ing the hypothesis that the liver is the major source *of The growing fetus, with its rapid rate of cellular division, has preformed purine nucleosides. These nucleosides, synthesized a great demand for pyrimidine nucleotides. The origin of these de novo in the liver, are transported by the circulation for nucleotides has not been established, but there is good 728 LEVINE, HOOGENRAAD, AND KRETCHMER evidence for maternal-fetal transfer of pyrimidine nucleosides. that incorporation of radioactive carbon dioxide into the Cytidine and uridine readily traverse the and label uridine nucleotide pool of lymphocytes was stimulated before fetal nucleic acids (57, 64, 65, 91). Rapid transfer is observed RNA biosynthesis began, suggesting an increased activity of in the rat after the 17th day of gestation, a time which the de novo pathway. Further, there was an increase in the corresponds to a period of rapid absolute growth when activity of the glutamine-requiring carbamyl phosphate synthe- demand for pyrimidines would be high (65). These studies also tase preceding the increased incorporation of labeled carbon indicate that uridine may be the species exported dioxide. The increased synthetase activity could be blocked by by the maternal liver. At 19 days of gestation, orotic acid actinomycin D. This finding is an important one in view of the labels maternal liver RNA, but uridine does not; the placenta fact that the activity of carbamyl phosphate synthetase is so and fetus are labeled by both precursors (65). There is also a low as to limit the rate of de novo pyrimidine biosynthesis in very high incorporation of orotic acid and cytidine into RNA normal lymphocytes. Thus the two- to fourfold stimulation of maternal liver as compared with uridine (Table 1). observed in the activity of this enzyme has an important effect Gurpide et al. (57) have shown that about 40-60% of fetal on the availability of pyrimidine nucleotides for nucleic acid blood uridine is of maternal origin. The demonstration of high biosynthesis (76). rates of uridine production coupled with the large maternal- Measurement of the incorporation of radioactive precursors fetal contribution suggest an important role for blood-borne into nucleic acids has often been used to study the relation of uridine as a precursor of nucleic acids. Tseng et al. (1 50) also pyrimidine metabolism to cellular proliferation. The careful found a very high rate of irreversible removal of pyrimidine studies of Forsdyke (43,44), using phytohemagglutinin-stimu- nucleosides from the circulation of dogs and humans, which lated lymphocytes, have emphasized the difficulties associated again implicated the blood supply as an important carrier of with such measurements. Forsdyke found that there were nucleosides. fluctuations in the size of endogenous pyrimidine pools after Dietary intake also provides a source of preformed stimulation with phytohemagglutinin. The extent of dilution pyrimidines, although they are rapidly catabolized and are not of labeled precurosr before incorporation into nucleic acids significant sources of nucleosides (13, 1 18). Consequently, depended on culture conditions and the time which had humans can be maintained readily on -free diets. elapsed since stimulation. Furthermore, the incorporation of Furthermore, isotope studies in man have demonstrated that orotic acid into RNA was shown to be dependent upon the there is significant reutilization of the nucleoside uridine, but size of the endogenous pools of preformed nucleosides present not of the free base, uracil (157), which suggests that dietary in the lymphocytes and in exogenously added serum. In tbess nucleic acids probably account for the postprandial rise in experiments there was no stimulation by phytohemagglutinin blood uridine noted by Tseng et al. (150). of incorporation of tritiated uridine and tritiated orotic acid into DNA. Uridine was incorporated readily into RNA, but PYRIMIDINE METABOLISM IN MODELS OF orotic acid was not, particularly when uridine was also added. PROLIFERATION AND DIFFERENTIATION The incorporation of orotic acid into RNA may have been inhibited because of the presence of a phosphorylated Pyrimidine nucleotides are involved in a variety of impor- derivative of uridine or cytidine (43) which could inhibit the tant pathways within the matrix of cellular . conversion of orotate to uridylate (see earlier discussion). However, their key role in nucleic acid biosynthesis has led to many studies of the pathway in systems which are character- ISOPROTERENOL STIMULATED SALIVARY GLANDS ized by rapid cellular division. A single injection of the sympathomimetic , isopro- PHYTOHEMAGGLUTININ-STIMULATED LYMPHOCYTES terenol, results in a marked stimulation of DNA biosynthesis in the salivary glands of mice and rats. This is followed by Lymphocytes can be stimulated to transform into cells increased mitotic activity and cell division (5, 7, 8). Twenty to in the presence of phytohemagglutinin. The stimulated cells 30 hr elapse between the time of injection and DNA synthesize DNA and then undergo mitosis and cell division. In biosynthesis, making this experimental model a suitable one the lag period preceding DNA biosynthesis there is a for the study of the events that lead to DNA biosynthesis and stimulated RNA biosynthesis, but there is no increase in cellular division. Many of the phenomena that occur during activities of many of the enzymes associated with nucleic acid this lag period have been reviewed by Baserga (9). metabolism (97, 122). The reutilization pathway enzyme, uri- Important events occur in the 1st hr after stimulation which dine kinase, is one of the few enzymes with stimulated level are crucial for subsequent DNA biosynthesis. For example, the of activity reaching a peak 6-10 hr after stimulation with stimulated DNA biosynthesis is inhibited by actinomycin D phytohemagglutinin (97). Thymidine kinase activity also is given either 30 min before or 60 min after administration of stimulated, reaching a peak at a time when DNA biosynthesis isoproterenol (10). Administration of cycloheximide in the is at a maximum (122). The activities of the de novo first hours after stimulation likewise blocks the increased DNA pyrimidine pathway enzymes aspartate transcarbamylase, biosynthesis (126). Events that occur before DNA biosynthesis dihydro-orotase, orotate phosphoribosyl transferase, and oroti- include an increased incorporation of radioactively labeled dylate decarboxylase are not affected by the phytohemagglu- orotic acid and uridine into RNA (6, 10, 99, 125), tinin (98). More recently however, Ito and Ichino (76) found biosynthesis of acidic nuclear (14 I), and biosynthesis of glycolipid (38, 51). In these stimulated salivary glands, as Table 1. Incorporation of labeled nucleosides into RNA of well as in phytohemagglutinin-stimulated lymphocytes (43), maternal liver compared with concentration in maternal blood there is little or no stimulation of the incorporation of uridine or orotic acid into DNA. Ratio of concentration, maternal Some of the enzymes involved in pyrimidine nucleotide Nucleoside liver/maternal blood metabolism have been investigated in isoproterenol-stimulated salivary glands. Within the first 4 hr after injection of Thymidine isoproterenol there is an elevation of activity of the first two Uridine enzymes of the de novo pyrimidine pathway, carbamyl Cytidine phosphate synthetase and aspartate transcarbamylase (123). Orotic Acid This increased enzyme activity is found at a time when there is a maximum incorporation of orotic acid into RNA. The ' Data from Hayashi and Garvey (64). activity of uridine kinase is elevated at the same time that PYRIMIDINE METABOLISM 729 there is maximal incorporation of uridine into RNA. The REGENERATING LIVER importance of the elevated de novo pathway activity for the DNA biosynthesis was demonstrated by inhibiting orotidylate After partial hepatectomy, there is a 24-hr lag before cell decarboxylase with 6-azauridine. It was shown that thymidine division occurs. A number of studies have attempted to incorporation into DNA was almost completely blocked with elucidate the events that occur during this lag period. DNA an amount of inhibitor adequate to completely block orotic biosynthesis occurs after some 12-39 hr, followed 6-8 hr acid incorporation into RNA, but insufficient to block uridine later by mitosis. The activity of a number of enzymes rises incorporation into RNA (1 23). These experiments indicate the with the increase in DNA biosynthesis. For example, importance of the de novo pyrimidine pathway as a source of thymidine kinase activity increases with DNA biosynthesis precursors for RNA biosynthesis. This newly synthesized RNA (17) and so does the activity of a species of DNA polymerase is then required for subsequent biosynthesis of DNA. (1 13). After partial hepatectomy there is a rapid change in In addition to its effect on de novo pyrimidine activity, RNA metabolism. For example, incorporation of radioactive isoproterenol also results in increased activities of thymidine orotic acid into RNA reaches a peak at 5 hr (50). This RNA kinase, thymidylate kinase, and DNA polymerase, although biosynthesis is required for later cell division; inasmuch as these enzymes attain maximum activities at approximately the DNA biosynthesis is prevented by p-fluorophenylalanine and same time that DNA biosynthesis takes place (9). actinomycin D (50). A feature of the RNA biosynthesis is that most of the newly formed RNA is found in the nucleus (50), INTESTINE as is the RNA made by kidney cortex cells after explantation (95), and by salivary glands after stimulation with isopro- Intestinal crypt cells show one of the highest rates of terenol(6, 123). cellular proliferation found in mammalian tissues (67). After Despite the rapid changes in RNA synthesis which result division in the crypt, cells migrate up the villus, during which from partial hepatectomy, aspartate transcarbamylase and time they develop the anatomic and biochemical characteris- dihydroorotase do not increase until 12-24 hr after partial tics of mature intestinal cells. It is possible to prepare serial hepatectomy. Although orotate phosphoribosyl transferase slices of intestine, from crypt to villus, and subject those slices and orotidylate decarboxylase activities increased within 2 hr to histologic and biochemical study. The transformation of (18), Fausto (39) found that these enzymes are not proliferating cells into differentiated cells can then be rate-limiting for RNA biosynthesis in regenerating liver. characterized by studying enzymes involved in pyrimidine Enzymes of the reutilization pathway of pyrimidine biosyn- biosynthesis and those involved in intestinal . In the thesis, such as uridine kinase, uridine phosphorylase, and crypt, one finds high activity of enzymes from both de novo kinase also increased in activity after partial and reutilization pathways. Activity decreases along the villus hepatectomy, but again, the stimulation was not observed to a minimum at the tip (3 1, 44, 67, 72, 149). Conversely, until at least 24 hr after surgery (133). enzymes typically associated with the differentiated intestinal Thus, although many of the events preceding cell division in cell increase from the crypt to the villus (44). A typical regenerating rat liver are known, the mechanism by which the pattern is shown in Figure 6. pyrimidine nucleotides required for early RNA biosynthesis in A compensatory increase in villus length occurs in patients this tissue are synthesized is still unclear. The time course of who have undergone resection of a large part of the small stimulation of enzymes studied does not correlate with the bowel. Consequently, there is increased activity of functional biosynthesis of these nucleotides. However, it should be noted enzymes such as lactase, sucrase, and maltase and an that the enzyme likely to be rate-limiting for de novo amelioration of the "short gut syndrome" (162). There is a pyrimidine biosynthesis, the glutamine-requiring carbamyl definite increase in cellular proliferation and of cellular phosphate synthetase, has not been measured in regenerating migration. Patients with celiac disease also seem to have liver. increased intestinal proliferative activity, with an increase in activity of enzymes of the de novo pyrimidine pathway (3 1). NEOPLASIA Neoplastic lesions of the colon demonstrate an enzyme pattern limited normally to the proliferating cells of the crypts (149). The transformation of cells into the neoplastic state is usually characterized by slow displacement in the molecular constitution. In general, rate-limiting enzymes show the greatest change, but all enzymes of nucleic acid biosynthesis tend to increase in activity while those involved in nucleic acid degradation tend to decrease (154). The ratio of opposing pathways characteristically changes as the rate of tissue growth changes. For example, the ratio of thymidine incorporation to thymidine degradation increases with the growth rate of a series of hepatomas (42). Also, the decreased activity of the rate-limiting enzyme in thymidine degradation, dihydrouracil dehydrogenase, significantly correlates with growth rate in a series of hepatomas (1 12, 121). Many enzymes involved in pyrimidine biosynthesis and degradation show the expected changes with different growth rates (154, 155). The activity of aspartate transcarbamylase correlates positively with the growth rate of normal tissues (52, 96, 164), and with a series of neoplasms with varying growth rates (21, 25, 112, 142). The glutamine-requiring carbamyl phosphate synthetase is correlated with the mitotic Fig. 6. Topographic distribution of enzymic activities in different index in normal tissue, and also with the growth rates of a regions of the rat jejunum. Enzyme assays were carried out on series of tumors (74, 163). Similar relations have been hpmogenates of tissue obtained by serial sectioning of the jejunum reported for other enzymes in the synthetic pathway (20, 93, using a cryostat. This figure is reproduced from Fortin-Magana et al. 1 16, 142). Conversely, enzymes in the degradative pathway (45) with permission of the authors and publisher. Copyright 1970 by show a negative correlation with growth rate (21, 42, 121, the American Association for the Advancement of Science. 138, 155). 730 LEVINE, HOOGENRAAD, AND KRETCHMER

There does not appear to be a single, unique enzyme which drug decreases production by inhibiting establishes the virulence of neoplastic growth. Instead, the oxidase (EC. 1.2.3.2), but it has been discovered recently that gradual replacement of normal enzyme patterns by those metabolites of the drug also inhibit de novo pyrimidine characteristic of neoplasia correlates with the virulence of the biosynthesis. causes an excretion of orotic acid tumor (88, 155). Knox (88) has emphasized that one must and orotidine in a pattern similar to that found in a variant of examine the pattern formed by a variety of cellular in hereditary orotic aciduria in which only orotidylate decar- order to make meaningful comparisons between biologically boxylase activity is low (46, 84, 139). This observation distinct cells. Using a given reference tissue (e.g., adult liver for suggests that allopurinol acts by inhibiting the decarboxylase hepatomas) a number can be derived statistically which (46, 48), probably through the conversion of allopurinol to provides a quantitative measure of the difference between oxypurinol (1 1). tissues. A gradual progression in pattern is observed: less Orotic acid may have a variety of clinical effects. The differentiated tumors resembling each other more than the purported pharmacologic uses of orotic acid are numerous parent tissue; differentiated tumors resemble the parent tissue (30). Orotic acid has been claimed to increase the rate of more than themselves (Fig. 7) (88). These models of regeneration after myocardial infarction; to counteract galac- proliferation provide examples of the two paradigms often tose-induced cataract and aid in the management of galac- utilized to define the biologic significance of pyrimidine tosemia; to nullify the diabetogenic action of ; and to biosynthesis. The first, or correlative paradigm, seeks to have an anti-inflammatory action in gouty similar to correlate biologic behavior with the molecular constitution of . The usefulness of allopurinol in could depend a cell. The possibility of identifying a key controlling enzyme in part upon this last action of orotic acid. in cells led to the measurement of many enzyme Pyrimidine nucleotides are required as carriers of and activities in neoplasms of varying growth rates. However, . This may account for other pharmacologic effects of virtually all enzymes studied have varied in a manner orotic acid. For example, orotic acid has significant protective consistent with rate of tissue growth. Biologically it seems as effects on the liver. A low dose mixture of hepatotoxic valuable to emphasize the characteristic pattern of changes in substances (carbon tetrachloride, chlorophenothane, and pyrimidine metabolism seen in neoplasia, as to search for a 9,lO-dimethyl-l,2-benzanthracene)causes pathologic liver unique factor which would explain the behavior of tumors. changes when fed to rats for 4 months. The changes are The second, or developmental paradigm, emphasizes the prevented by the addition of orotic acid to the diet (1 15). temporal relation of genetic events which control the Small doses of d-galactosamine can induce a viral-like molecular machinery of dividing cells. Careful studies of the . The rate-limiting step in the disposal of galac- sequence of events which occur before cell division have begun tosamine involves the formation of UDP-galactosamine. The to delineate the role of pyrimidine biosynthesis in cellular step appears to be limited not by the activity of the enzyme proliferation. In model mammalian systems, brief interference involved but by the availability of uridine nucleotides: The with protein synthesis or enzyme activity may prevent later uridylate pool drops after the administration of galactosamine, cellular division. and this decrease may be the cause of the hepatitis (37). The fall in the concentration of uridylate can be prevented by PHARMACOLOGIC PROPERTIES OF OROTIC ACID administration of orotic acid; hepatitis then does not develop. The limited ability of neonates to conjugate bilirubin has Studies of hereditary orotic aciduria have yielded important been attributed generally to the limited activity of glucuronyl information concerning de novo pyrimidine biosynthesis (136) transferase (EC. 2.4.1.17). However, as in the case of and control mechanisms of this pathway in higher organisms. galactosamine hepatitis, it is possible that UTP required for the Heriditary orotic aciduria is rare, although orotic aciduria is formation of the substrate, UDP-, is limiting, found commonly in patients receiving the anti-gout drug, but the available experimental evidence is conflicting. allopurinol. Allopurinol is useful in treating gout because the The involvement of orotic acid in hepatic metabolism is further corroborated by evidence that oral administration of ADULT orotic acid has a marked effect on lipid metabolism and at LIVER certain doses produces a severe fatty liver in rats. Standerfer ,-• Dl FFERENTIATED __==5-' and Handler (140) originally described the marked fatty infiltration caused by the addition of orotic acid to the diet of rats. The usual lipotropic agents such as folic acid, UNDIFFERENTIATED - Biz, , and choline could not prevent the infiltra- tion. Weanling rats fed a defined diet plus 1% orotic acid , rapidly showed marked changes in hepatic nucleotides and 1' 0.523 1.010 1.533 fat content (62, 152). After only 10 days on the diet, uridine nucleotides were increased fourfold and fat content was increased over threefold. The liver content of adenine nucleotides was decreased by 5076, and supplementation of the CARCINOMA ----_-%.- - _0.613 diet with 0.25% adenine sulfate completely prevented develop- 5A ment of the fatty liver. The decrease in hepatic adenine P

33. Creasey, W. A., and Handschumacher, R. E.: Purification and for the synthesis of carbamyl phosphate for pyrimidine properties of orotidylate decarboxylases from yeast and rat biosynthesis in fetal rat liver. J. Biol. Chem., 242: 5674 (1967). liver. J. Biol. Chem., 236: 2058 (1961). 61. Hallanger, L. E., Laakso, J. W., and Schultze, M. 0.: Orotic acid 34. Creasey, W. A,: Studies on the metabolism of 5-iodo-2'-deoxy- in milk. J. Biol. Chem., 202: 83 (1953). cytidine in vitro. Purification of nucleoside deaminase from 62. Handschumacber, R. E., Creasy, W. A., Jaffe, J. J., Pasternak, C. mouse kidney. J. Biol. Chem., 238: 1772 (1963). A,, and Hankin, L.: Biochemical and nutritional studies on the 35. Curci, M. R., and Donachie, W. D.: An attempt to find pyrimidine induction of fatty by dietary orotic acid. Proc. Nat. Acad. inhibitors of a mammalian aspartate carbamoyltransferase. Sci. U.S.A., 46: 178 (1960). Biochim. Biophys. Acta, 85: 338 (1964). 63. Hatfield, D., and Wyngaarden, J. B.: 3-Ribosylpurines. I. 36. Davis, R. H.: Metabolite distribution in cells. Science, 178: 835 Synthesis of (3-ribosyluric acid) 5'-phosphate and (3-ribosylxan- (1972). thine) 5'-phosphate by a pyrimidine pyrophos- 37. Decker, K., Keppler, D., Rudigier, J., and Domschke, W.: Cell phorylase of beef erythrocytes. J. Biol. Chem., 239: 2580 damage by trapping of biosynthetic intermediates. The role of (1964). uracil nucleotides in experimental hepatitis. Hoppe Seyler's Z. 64. Hayashi, T. T., and Garvery, B. I.: Transplacental passage of Physiol. Chem., 352: 412 (197 1). nucleotides, nucleosides, and bases. Amer. J. Obstet. Gynecol., 38. Fallon, H. J., Frei, E., 111, Block, J., and Seegmiller, J. E.: The 102: 1154 (1968). uricosuria and orotic aciduria induced by 6-azauridine. J. Clin. 65. Hayashi, T. T., Shin, D. H., and Wiand, S.: Placental transfer of Invest., 40: 1906 (1961). orotic acid, uridine, and UMP. I. Comparison of acid-soluble 39. Fausto, N.: The control of RNA synthesis during liver regenera- and acid-insoluble counts. Amer. J. Obstet. Gynecol. 102: 1144 tion. Orotidine 5'-phosphate pyrophosphorylase and decarbox- (1968). ylase activities in normal and regenerating liver. Biochim. 66. Heird, W. C., Nicholson, J. F., Driscoll, J. M., Schullinger, J. N., Biophys. Acta, 182: 66 (1969). and Winters, R. W.: Hyperammonemia resulting from intrave- 40. Feinendegen, L. E., Bond, V. P., Cronkite, E. P., and Hughes, W. nous alimentation using a maxture of synthetic L-amino acids: L.: RNA turnover in normal rat bone marrow. Ann. N. Y. Acad. A preliminary report. J. Pediat., 81: 162 (1972). Sci., 113: 727 (1964). 67. Herbst, J. J., Fortin-Magana, R., and Sunshine, P.: Relationship of 41. Feinendegen, L. E., Bond, V. P., and Hughes, W. L.: Physiological pyrimidine biosynthetic enzymes to cellular proliferation in rat thymidine reutilization in rat bone marrow. Proc. Soc. Exp. intestine during development. Gastroenterology, 59: 240 Biol. Med., 122: 448 (1966). (1970). 42. Ferdinandus, J. A,, Morris, H. P., and Weber, G.: Behavior of 68. Hers, H. G., De Wulf, H., Stahlmans, W., and Van Den Berghe, G.: opposing pathways of thymidine utilization in differentiating, The Control of glycogen synthesis in the liver. Advan. Enzyme regenerating, and neoplastic liver. Cancer Res., 31: 550 (1971). Regul., 8: 171 (1969). 43. Forsdyke, D. R.: Studies of the incorporation of (5-H-3) uridine 69. Hogans, A. F., Guroff, G., and Udenfriend, S.: Studies on the during activation and transformation of lymphocytes induced origin of pyrimidines for biosynthesis of neural RNA in the rat. by phytohaemagglutinin. Dependence of the incorporation rate J. Neurochem., 18: 1699 (197 1). on uridine concentration. Biochem. J., 107: 197 (1968). 70. Hoogenraad, N. J., and Lee, D. C.: Effect of uridine on de novo 44. Forsdyke, D. R.: Application of the isotope-dilution principle to pyrimidine biosynthesis in rat hepatoma cells in culture. J. Biol. the analysis of factors affecting the incorporation of H-3 uridine Chem., in press. and H-3 cytidine into cultured lymphocytes: Evaluation of 71. Hoogenraad, N. J., Levine, R. L., and Kretchmer, N.: Copurifica- pools. Biochem. J., 125: 721 (1971). tion of synthetase and aspartate trans- 45. Fortin-Magana, R., Hunvitz, R., Herbst, J. J., and Kretchmer, N.: carbarnolase from mouse spleen. Biochem. Biophys. Res. Intestinal enzymes: Indicators of proliferation and differentia- Commun., 44: 981 (1971). tion in the jejunum. Science, 167: 1627 (1970). 72. Imondi, A. R., Lipkin, M., and Balis, M. E.: Enzyme and template 46. Fox, R. M., and O'Sullivan, W. J.: Purine and pyrimidine stability as regulatory mechanisms in differentiating intestinal biosynthesis. Allopurinol and inborn errors. Minn. Med., 54: epithelial cells. J. Biol. Chem., 245: 2194 (1970). 397 (1971). 73. Ishii, K., and Green, H.: Lethality of for cultured 47. Fox, R. M., O'Sullivan, W. J., and Firkin, B. G.: Orotic aciduria. mammalian cells by interference with pyrimidine biosynthesis. Differing enzyme patterns. Amer. J. Med., 47: 332 (1969). J. Cell Sci., 13: 429 (1973). 48. Fox, R. M., Wood, M. H., and O'Sullivan, W. J.: Studies on the 74. Ito, K., Nakanishi, S., Terada, M., and Tatibana, M.: Control of coordinate activity and lability of orotidylate phosphoribosyl- pyrimidine biosynthesis in mammalian tissues. 11. Glutamine transferase and decarboxylase in human erythrocytes, and the utilizing carbamoyl phosphate synthetase of various experimen- effects of allopurinol. J. Clin, Invest. 50: 1050 (1971). tal tumors: distribution, purification, and characterization. 49. Fredrickson, D. S., Gotto, A. M., and Levy, R. I.: Familial Biochim. Biophys. Acta, 220: 477 (1970). lipoprotein deficiency. In: J. B. Stanbury, J. B. Wyngaarden, 75. Ito, K., and Tatibana, M.: Operation of orotic acid pathways as and D. S. Fredrickson: The Metabolic Basis of Inherited the major source of pyrimidines in the bematopoietic mouse Disease, Ed. 3, p. 493 (McGraw-Hill, New York, 1972). spleen. Biochem. Biophys. Res. Commun., 23: 672 (1966). 50. Fujioka, M., Koga, M., and Lieberman, I.: Metabolism of 76. Ito, K., and Uchino, H.: Control of pyrimidine biosynthesis in ribonucleic acid after partial hepatectomy. J. Biol. Chem., 238: human lymphocytes: Induction of glutamine utilizing carbamyl 3401 (1963). phosphate synthetase and operation of orotic acid pathway 51. Galanti, N., and Baserga, R.: Glycolipid synthesis in the early during blastogenesis. J. Biol. Chem., 246: 4060 (1971). prereplicative phase of isoproterenol stimulated salivary glands 77. Jacobson, G. R., and Stark, G. R.: Aspartate transcarbamylases. of mice. J. Biol. Chem., 246: 6814 (1971). In: P. D. Boyer: The Enzymes, Ed. 3, Vol. 9, p. 225 (Academic 52. Galofr;, A,, and Kretcbmer, N.: Biosynthesis of pyrimidines by Press, New York, 1973). various organs of the chick during embryogenesis. Pediat. Res., 78. Johnson, J. D., Albritton, W. L., and Sunshine, P.: Hyperammo- 4: 55 (1970). nemia accompanying parenteral nutrition in newborn infants. J. 53. Geiger, A.: Correlation of brain metabolism and function by the Pediat., 81: 154 (1972). use of a brain perfusion method in situ. Physiol. Rev., 38: 1 79. Jones, M. E.: Vertebrate carbamyl phosphate synthetase I and 11. (1958). In: B. Schmidt-Nielson: Urea and the Kidney, International 54. Geiger, A., and Yamasaki, S.: Cytidine and uridine requirement of Congress Series, No. 195, p. 35 (Excerpta Medica Foundation, the brain. J. Neurochem., 1: 93 (1956). Amsterdam, 1970). 55. Goldstein, A. S., Hoogenraad, N. J., Johnson, J. D., Fukanaga, K., 80. Jones, M. E.: Regulation of pyrimidine and arginine biosynthesis Swierczewski, E., Cann, H. M., and Sunshine, P.: Metabolic and in mammals. Advan. Enzyme Regul., 9: 19 (1970). genetic studies of a family with ornithine transcarbamylase 81. Jones, M. E.: Regulation of uridylic acid biosynthesis in deficiency. Pediat. Res., 8: 5 (1974). eucaryotic cells. Curr. Top. Cell Regul., 6: 227 (1972). 56. Grisolia, S., and Cardoso, S. S.: The purification and properties of 82. Jones, M. E., Anderson, A. D., Anderson, C., and Hodes, S.: hydropyrimidine dehydrogenase. Biochim. Biophys. Acta, 25: synthesis in rat tissues. Arch. Biochem., 95: 499 430 (1957). (1961). 57. Gurpide, E., Tseng, J., Escarcena, L., Fahning, M., Gibson, C., and 83. Kasbekar, D. K., Nagabhushanam, A., and Greenberg, D. M.: Fehr, P.: Fetomaternal production and transfer of Purification and properties of orotic acid decarboxylating and uridine in sheep. Amer. J. Obstet. Gynec., 113: 21 (1972). enzymes from calf thymus. J. Biol. Chem., 239: 4245 (1964). 58. Hager, S. E., and Jones, M. E.: Initial steps in pyrimidine 84. Kelley, W. N., and Beardmore, T. D.: Allopurinol: Alteration in synthesis in Ehrlich ascites carcinoma in vitro. I. Factors pyrimidine metabolism in man. Science, 169: 388 (1970). affecting the incorporation of ' C- into carbon 2 of 85. Kelley, W. N., Greene, M. L., Fox, I. H., Rosenbloom, F. M., the uracil ring of the acid soluble nucleotides. J. Biol. Chem., Levy, R. I., and Seegmiller, J. E.: Effects of orotic acid on 240: 4556 (1965). purine and lipoprotein metabolism in man. Metabolism, 19: 59. Hager, S. E., and Jones, M. E.: Initial steps in pyrimidine 1025 (1970). synthesis in Ehrlich ascites carcinoma in vitro. 11. The 86. Kit, S.: Nucleotides and nucleic acids In: D. M. Greenberg: glutamine-dependent carbamyl phosphate synthetase. J. Biol. Metabolic Pathways, Ed. 3, Vol. 4, p. 69 (Academic Press, New Chem., 242: 5667 (1967). York, 1970). 60. Hager, S. E., and Jones, M. E.: A glutamine-dependent enzyme 87. Klemperer, H. G., and Haynes, G. R.: Thymidine kinase in rat PYRIMIDINE METABOLISM 733

liver during development. Biochem. J., 108: 541 (1968). of nucleosides into nucleotide pools and pool sizes during 88. Knox, W. E.: The protoplasmic patterns of tissues and tumors. growth cycle. J. Cell Physiol., 77: 213 (1971). Amer. Sci., 60: 480 (1972). 118. Plentl, A. A,, and Schoenheimer, R.: Studies in the metabolism of 89. Kobata, A., Furuno, F., and Ziro, S.: The chemical components purines and pyrimidines by means of isotopic nitrogen. J. Biol. of milk which prevent the fatty liver induced by orotic acid. J. Chem., 153: 203 (1944). Biochem., 55: 92 (1964). 119. Potter, V. R.: Metabolic products formed from thymidine. In: F. 90. Krystal, G., and Webb, T. E.: Multiple forms of uridine kinase in Stohlman, Jr.: The Kinetics of Cellular Proliferation, p. 104 normal and neoplastic rat liver. Biochem J., 124: 1943 (1971). (Grune and Stratton, New York, 1959). 91. Larkin, L. H., and Stevens, A. R.: Foetal and maternal RNA 120. Prager, M.D., Young J. E., and Atkins, I. C.: A study of the labelled with H-3 cytidine. Nature New Biol., 235: 107 (1972). possible role of feedback inhibition of aspartate transcar- 92. Larson, B. L., and Hageman, E. C.: Ejection of orotic acid from bamylase in regulation of pyrimidine synthesis in human the bovine mammary secretory cell. Fed. Proc., 29: 445 (1970). leukocytes. J. Lab. Clin. Med., 70: 768 (1967). 93. Lea, M. A., Morris, H. P., and Weber, G: Comparative 121. Queener, S. F., Morris, H. P., and Weber, G.: Dihydrouracil biochemistry of hepatomas. VI. Thymidine incorporation into dehydrogenase activity in normal, differentiating, and regenerat- DNA as a measure of hepatoma growth rate. Cancer Res., 26: ing liver and in hepatomas. Cancer Res., 31: 1004 (1971). 465 (1966). 122. Rabinowitz, Y., Wong, P., and Wilhite, B.: Thymidine metaboliz- 94. Levine, R. L., Hoogenraad, N. J., and Kretchmer, N.: Regulation ing enzymes of normal lymphocytes cultured with phytohemag- of activity of carbamoyl phosphate synthetase from mouse glutinin. In: 0. R. McIntyre: Proceedings of the Fourth Annual spleen. Biochemistry, 10: 3694 (1 97 1). Leucocyte Culture Conference, p. 81 (Appleton-Century-Crofts, 95. Lieberman, I. Abrams, R., and Ove, P.: Changes in the New York, 1969). metabolism of ribonucleic acid preceding the synthesis of 123. Roux, J. M., Hoogenraad, N. J., and Kretchmer, N.: Biosynthesis deoxyribonucleic acid in mammalian cells cultured from the of pyrimidine 'ucleotides in mouse salivary glands stimulated animal. J. Biol. Chem., 238: 2141 (1963). with isoproterenol. J. Biol. Chem., 248: 1196 (1973). 96. Lowenstein, J. M., and Cohen, P. P.: Studies on the biosynthesis 124. Sarma, D. S., and Sidransky, H.: Studies on orotic acid fatty liver of carbamylaspartic acid. J. Biol. Chem., 220: 57 (1956). in rats: Factors influencing the induction of fatty liver. J. Nutr., 97. Lucas, 2. J.: Pyrimidine nucleotide synthesis: Regulatory control 98: 33 (1969). during transformation of lymphocytes in vitro. Science, 156: 125. Sasaki, T., and Baserga, R.: Cytoplasmic ribonucleic acid 1237 (1967). synthesis in the prereplicative phase of isoproterenol induced 98. Lucas, Z. J.: Recent studies on the regulatory control of cell proliferation. Exp. Mol. Pathol., 13: 25 (1970). pyrimidine nucleotide synthesis in lymphocytes. In: W. 0. 126. Sasaki, T., Litwack, G., and Baserga, R.: Protein synthesis in the Rieke: Proceedings of the Third Annual Leucocyte Culture early prereplicative phase of isoproterenol stimulated synthesis Conference, p. 655 (Appleton-Century-Crofts, New York, of deoxyribonucleic acid. J. Biol. Chem., 244: 4831 (1969). 1969). 127. Schneider, W. C., and Greco, A. E.: Incorporation of pyrimidine 99. Malamud, D., and Baserga, R.: Pool size and specific activity of into liver lipids and other components. UTP in isoproterenol stimulated salivary glands. Biochim. Biochim. Biophys. Acta, 228: 610 (1971). Biophys. Acta, 195: 258 (1969). 128. Shambaugh, G. E., 111, Metzger, B. E., and Freinkel, N.: 100. Marchetti, M., Puddu, P., and Caldarera, C. M.: Liver acid soluble Glutamine dependent carbamyl phosphate synthetase in pla- nucleotides in orotic acid fed rats. Biochim. Biophys. Acta, 61: centa and fetal structures of the rat. Biochem. Biophys. Res. 826 (1 962). Commun., 42: 155 (197 1). 101. Marchetti, M., Puddu, P., and Caldarera, C. M.: Metabolic aspects 129. Shih, V. E., and Efron, M. L.: Urea cycle disorders. In: J. B. of "orotic acid fatty liver." Nucleotide control mechanisms of Stanbury, J. B. Wyngaarden, and D. S. Fredrickson: The lipid metabolism. Biochem. J., 92: (1964). Metabolic Basis of Inherited Disease, Ed. 3, p. 370 (McGraw- , 102. Martin, D. W., and Owen, N. T.: Repression and derepression of Hill, New York, 1972). purine biosynthesis in mammalian hepatoma cells in culture. J. 130. Shoaf, W. T., and Jones, M. E.: Initial steps in pyrimidine Biol. Chem., 247: 5477 (1972). synthesis in Ehrlich ascites carcinoma. Biochem. Biophys. Res. 103. McMurray, W. C., and Magee, W. L.: Phospholipid metabolism. Commun., 45: 796 (1971). Annu. Rev. Biochem., 41: 129 0972). 131. Shoaf, W. T., and Jones, M. E.: Enzyme complexes of the 104. Michaelis, P., and Mandel, P: Etude du passage de quelques pyrimidine pathway in Ehrlich ascites tumor cells. Fed. Proc., precurseurs de acides nucleiques du liquide cephaloarachidien 31: 473 (1972). dans le systeme nerveux central. C. R. Soc. Biol. (Paris), 158: 132. Shoaf, W. T., and Jones, M. E.: Uridylic acid synthesis in Ehrlich 1588 (1964). ascites carcinoma. Properties, subcellular distribution, and 105. Milner, J. A., and Visek, W. J.: Orotic aciduria and arginine nature of enzyme complexes of the six biosynthetic enzymes. deficiency. Nature New Biol., 245: 21 1 (1973). Biochemistry, 12: 4039 (1973). 106. Murray, A. W.: The biological significance of purine salvage. 133. Skold, 0.: Enzymes of uracil metabolism in tissues with different Annu. Rev. Biochem., 40: 811 (1971). growth characteristics. Biochim. Biophys. Acta, 44: 1 (1960). 107. Natale, P. J., and Tremblay, G. C.: On the availability of 134. Skold, 0.: Uridine kinase from Ehrlich ascites tumor: Purification intramitochondrial carbamoylphosphate for the extramito- and properties. J. Biol. Chem., 235: 3273 (1960). chondrial biosynthesis of pyrimidines. Biochem. Biophys. Res. 135. Skold, 0.: Studies on resistance against 5-fluorouracil. IV, Commun., 37: 5 12 (1969). Evidence for an altered uridine kinase in resistant cells. 108. Nicholson, J. F.: Metabolism of ingested -N-15 by Biochim. Biophys. Acta, 76: 160.(1963). premature infants. Pediat. Res., 4: 398 (1970). 136. Smith, L. H., Jr., Huguley, C. M., Jr., and Bain, J. A.: Hereditary 109. Nordlie, R. C., Lueck, J. D., Hanson, T. L., and Johns, P. J.: The orotic aciduria. In: J. 9. Stanbury, J. B. Wyngaarden, and D. S. nature of pH-discriminant differences in behavior of various Frederickson: The Metabolic Basis of Inherited Disease, Ed. 3, phosphoanhydrides, mixed phosphate anhydrides, and phos- p. 1003 (McGraw-Hill, New York, 1972). phate esters as substrates and inhibitors with microsomal 137. Sneider, T. W., and Potter, V. R.: Deoxycytidylate deaminase and glucose 6-. J. Biol. Chem., 246: 4807 (1971). related enzymes of metabolism in 110. O'Donovan, G. A., and, Neuhard, J.: Pyrimidine metabolism in hepatomas and precancerous rat liver. Advan. Enzyme Regul., . Bacteriol. Rev., 34: 278 (1970). 7: 375 (1968). 111. Okonkwo, P. O., and Kinsella, J. E.: Orotic acid in milk 138. Sneider, T. W., Potter, V. R., and Morris, H. P.: Enzymes of powders. Amer. J. Clin. Nutr., 22: 532 (1969). thymidine triphosphate synthesis in selected Morris hepatomas. 112. Ono, T., Blair, D. G. R., Potter, V. R., and Morris, H. P.: The Cancer Res., 29: 40 (1969). comparative enzymology and cell origin of rat hepatomas. IV. 139. Soutter, G. B., Yu, J. S., Lovric, A., and Stapleton, T.: Hereditary Pyrimidine metabolism in minimal deviation tumors. Cancer orotic aciduria. Aust. Paediat. J., 6: 47 (1970). Res., 23: 240 (1963). 140. Standerfer, S. B., and Handler, P.: Fatty liver induced by orotic 113. Ove, P., Laszlo, J., Jenkins, M.D., and Morris, H. P.: Increased acid feeding. Proc. Soc. Exp. Biol. Med., 90: 270 (1955). DNA polymerase activity in a series of rat hepatomas. Cancer 141. Stein, G., and Baserga, R.: The synthesis of acidic nuclear Res., 29: 1557 (1969). proteins in the prereplicative phase of the isoproterenol 114. Parsons, D. F., and Subjeck, J. R.: The morphology of the stimulated salivary gland. J. Biol. Chem., 245: 6097 (1970). coat of mammalian cells. Biochim. Biophys. 142. Sweeney, M. J., Hoffman, D. H., and Poore, G. A.: Enzymes in Acta, 265: 85 (1972). pyrimidine biosynthesis. Advan. Enzyme Regul., 9: 51 (1970). 115. Pates, M. M., Tseitina, A. Y., Pomerantseva, I. I., Tunitskaya, T. 143. Tatibana, M., and Ito, K.: Carbamyl phosphate synthetase of the A., Kurkina, V. S., and Turetskaya, I. M.: Protective action of hematopoietic mouse spleen and the control of pyrimidine orotic acid on the liver following exposure to low doses of toxic biosynthesis. Biochem. Biophys. Res. Commun., 26: 221 substances. Chem. Abstr., 70: 561 16e (1 969). (1967). 116. Pausch, J., Keppler, D., and Decker, K.: Activity and distribution 144. Tatibana, M., and Ito, K.: Control of pyrimidine biosynthesis in of the enzymes of uridylate synthesis from orotate in animal mammalian tissues. I. Partial purification and characterization tissues. Biochim. Biophys. Acta, 258: 395 (1972). of glutamine-utilizing carbamyl phosphate synthetase of mouse 117. Plageman, P. G. W.: Nucleotide pools of Novikoff rat hepatoma spleen and its tissue distribution. J. Biol. Chem, 244: 5403 cells growing in suspension culture. I. Kinetics of incorporation (1969). 734 LEVINE, HOOGENRAAD, AND KRETCHMER

145. Tatibana, M., and Shigesada, K.: Activation by 5-phosphoribosyl- isotopically labeled uracil and uridine in man. Metabolism, 12: pyrophosphate of glutamine dependent carbamyl phosphate 60 (1963). synthetase from mouse spleen. Biochem. Biophys. Res. Com- 158. Windmueller, H. G., Levy, R. I., and Spaeth, A. E.: Selective mun., 46: 491 (1972). inhibition of hepatic but not intestinal beta-lipoprotein produc- 146. Tatibana, M., and Shigesada, K.: Control of pyrimidine biosynthe- tion and triglyceride transport in rats given orotic acid. Advan. sis in mammalian tissues. IV. Requirements of a quantitative Exp. Med. Biol., 4: 365 (19693. assay of glutamine dependent carbamyl phosphate synthetase 159. Windmueller, H. G., and Spaeth, A. E.: Stimulation of hepatic and effect of magnesium as essential activator. J. Biochem., 72: purine biosynthesis by orotic acid. J. Biol. Chem., 240: 4398 537 (1972). (1965). 147. Tatibana, M., and Shigesada, K.: Control of pryimidine biosynthe- 160. Windmueller, H. G., and Von Euler, L. H.: Prevention of orotic sis in mammalian tissues. V. Regulation of glutamine dependent acid induced fatty liver with allopurinol. Proc. Soc. Exp. Biol. carbamyl phosphate synthetase: Activation by 5-phosphori- Med., 136: 98 (1971). bosyl I-pyrophosphate and inhibition by UTP. J. Biochem., 72: 161. Winzler, R. J.: Carbohydrate in cell surfaces. Int. Rev. Cytol., 29: 549 (1972). 77 (1970). 148. Tomchick, R., Saslaw, L. D., and Waravdekar, V. S.: Mouse 162. Wright, H. K., and Tilson, M.D.: The short gut syndrome: kidney : Purification and properties. J. Biol. Pathophysiology and treatment. Curr. Probl. Surg., June: 1 Chem., 243: 2534 (1968). (1971). 149. Troncale, F., Hertz, R., and Lipkin, M.: Nucleic acid metabolism 163. Yip, M. C., and Knox, W. E.: Glutamine-dependent carbamyl in proliferating and differentiating colonic cells of man and in phosphate synthetase: Properties and distribution in normal and neoplastic lesions of the colon. Cancer Res., 31: 463 (1971). neoplastic rat tissues. J. Biol. Chem., 245: 2199 (1970). 150. Tseng, J., Barelkovski, J., and Gurpide, E.: Rates of formation of 164. Young, J. E., Prager, M.D., and Atkins, I. C.: Comparative blood-borne uridine and cytidine in dogs. Amer. J. Physiol., activities of aspartate transcarbamylase in various tissues of the 221: 869 (1971). rat. Proc. Soc. Exp. Biol. Med., 125: 860 (1967). 15 1. Valli, E. A., Sarma, D. S. R., and Sarma, P. S.: Species specificity 165. Work from this laboratory was supported in part by grants from in orotic acid induced fatty liver. Indian J. Biochem., 5: 120 the National Foundation-March of Dimes, the Lockheed (1968). Employee's Bucks-of-the-Month Club, and the National Insti- 152. Von Euler, L. H., Rubin, R. J., and Handschumacher, R. E.: tutes of Health HD-00391, HD-02147, and Training Grant No. Fatty livers induced by orotic acid. 11. Changes in nucleotide HD-00049, from the United States Public Health Service. metabolism. J. Biol. Chem., 238: 2464 (1963). Computer support was provided by the Advanced Computer for \C 153. Wallach, D. P.: Grisolia, S.: The purification and properties of Medical Research (ACME) Facility of Stanford University hydropyrimidine hydrase. J. Biol. Chem., 226: 277 (1957). Medical Center, with support from the Biotechnology Re- 154. Weber, G., and Lea, M. A.: The molecular correlation concept. An sources Branch of the National Institutes of Health under Grant experimental and conceptual method in cancer research. In: H. No. RR-003 11. Busch: Methods in Cancer Research, Vol. 2, p. 523 (Academic 166. Dr. R. L. Levine was a Predoctoral Fellow of the Medical Scientist Press, New York, 1967). Training Program, under National Institutes of Health Grant 155. Weber, G., Queener, S. F., and Ferdinandus, J. A.: Control of No. GM-1922. expression in carbohydrate, pyrimidine, and DNA metabo- 167. Requests for reprints should be addressed to: N. Kretchmer, lism. Advan. Enzyme Regul., 9: 63 (1970). M.D., Ph.D., Division of Developmental Biology, Department of 156. Weichsel, M. E., Jr., Hoogenraad, N. J., Levine, R. L., and Pediatrics, Stanford University Medical Center, Stanford, Calif. Kretchmer, N.: Pyrimidine biosynthesis during development of 94305 (USA). rat cerebellum. Pediat. Res., 6: 682 (1972). 168. Accepted for publication March 18, 1974. 157. Weissman, S. M., Lewis, M., and Karon, M.: The metabolism of

Copyright O 1974 International Pediatric Research Foundation, Inc. Prin red in U.S.A.