METABOLISM OF L-MANNOSZE IN AEROBACTER AEROGENES Thesis {or “to Dogma of m. D. MCEEGAN STATE UMVEZRSETY Joseph William Mayo 1968 0-169 Date ,4! LI 23 “AF h‘iCEl 1‘13“ Sin Us“ diversity .1“ This is to certify that the thesis entitled METABOLISM OF L-MANNOSE IN AEROBACTER AEROGENES presented by Joseph W. Mayo has been accepted towards fulfillment of the requirements for Ph.D. degree in Biochemistry fl . “a Major professor 8/15/68 i .-_.. -- -—-_ ABSTRACT METABOLISM OF L-MANNOSE IN AEROBACTER AEROGENES by Joseph W. Mayo A mutant strain of Aerobacter aerogenes PRL-Rj was isolated wnicn, unlike the parent strain, grew readily on the unnatural hexoses, L-mannose and L-fr’uc— tose, as sole carbon sources. The pathway bywhich L-mannose is degraded in this organism was elucidated. An isomerase catalyzes the conversion of L-mannose to L-fructose, which is phOSphorylated with adenosine 5'- triphosphate by a kinase to yield L-fructose l-phosphate. L—Fructose l-phosphate is cleaved by an aldolase to yield dihydroxyacetone phosphate and L-glyceraldehyde; The two intermediates in the pathway, L-fructose and L-fructose l-phosphate, were isolated and characterized. The enzymes which degraded L-mannose also degraded the naturally occurring b-deoxy hexose, L-rhamnose; Both L-mannose and L-rhamnose induced all three enzymes in both the wild-type and the Lemannose-positive cells: the ratio of the specific activities of the enzymes on L-mannose and L-rhamnose and their respeCtive metabolic intermediates were the same in cells grown on either substrate. L-Mannose Joseph W. Mayo was isomerized at a rate 25% that of L-rhamnose, L— fructose was phosphorylated at a rate 10% greater than L-rhamnulose, and L-fructose-l-P was cleaved at a rate 25-30% that of L-rhamnulose-l-P. Partial fractionation of the enzymes with ammonium sulfate and Sephadex G-100 failed to separate the enzymes of the L-mannose pathway from those of the L-rhamnose pathway. When the two sub- strates for each enzyme were mixed, the individual activities were competitive rather than additive. A mutant of A, aerogenes PRL-R3 deficient in the isomerase was unable to isomerize either L-mannose or L-rhamnose. 'The mechanism by which A, aerogenes gains the ability to metabolize many of the unnatural hexoses, pentoses, and penitols has been shown by other workers to involve the selection of derepressed mutants containing’higher levels of non-specific enzymes which have naturally occurring compounds as their normal substrates. In contrast to this, the gain in the ability of this organism to grow on L-mannose as a sole carbon source did not involve selection of derepressed mutants. L-Mannose inhibited the growth of both the wild type and the mutant at the onset of L-mannose utilization by the cells; only the mutant overcame this inhibition. When the inducer, L- mannose, was removed from actively growing mutant cells, Joseph W. Mayo a rapid decrease in the activities of all three enzymes occurred. Also, L-mannose was not utilized by the mutant cells in the presence of either chloramphenicol or pure- mycin, further indicating that growth on L-mannose required the induction of these enzymes. METABOLISM OF L-MANNOSE IN AEROBACTER AEROGENES BY Joseph William Mayo A THESIS Submitted to Michigan State university in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Biochemistry 1968 Dedicated to my parents ACKNOWLEDGMENT The author wishes to express his gratitude to Dr. Richard L. Anderson for his advice and helpful criticisms throughout the course of this work. The discussions and assistance of his fellow graduate students also are deeply appreciated. Thanks are due to Mrs, Barbara Palmer and Mrs. Shirley Randall for their help in preparing this manuscript. The financial support of the National Institutes of Health also is appreciated. ii TABLE OF CONTENTS ACKNOWLEDGMENTS............ LIST OF TABLES o g o o o o o o 9' o o 0 LIST OF FIGUES O O O O O Q 0 O O O C 0 INTRODUCTION .. . PART I. O Q 0 C THE BIODEGRADATION OF L-MANNOSE BY AEROBACTER AEROGENES. . . . . . . Experimental Procedures. . . . . . Results . . . . . . . . . . . Apparent phosphorylation of L-mannose Manometric: L-mannose-stimulated C02 evolution from bicarbonate in the presence of ATP . . . Titrametric: increased rate of acid production from ATP in the presence of L-mannose . . Colorimetric: ATP-stimulated dis- appearance of L-mannose . . . Spectrophotometric test for L- mannokinase. . . . . . . Isomerization of L-mannose to L- fructose. . . . . . . . Enzymic preparation of L- fructose . Growth of.§. aerggenes on L- fructose Phosphorylation of L-fructose to L— fructose- l- P. . . . Enzymic preparation of L- fructose 1- phosphate . . . . . . . Identification of L- fructose 1- phosphate . . . . Enzymic cleavage of L—fructose 1- phosphate . . Reduction of L-glyceraldehyde to glycerol. . . Oxidation of L-glyceraldehyde to. glyceric acid . . . .g . . . iii Page ii vi vii 11 ll 12 12 12 21 21 33 33 36 37 ‘ 41 47 47 PART II. PART III. Page Discussion . . . . . . , , , 48 COMMON IDENTITY OF THE ENZYMES OF L- MANNOSE AND L-RHAMNOSE METABOLISM. . 56 Experimental Procedures . . . . . 55 Results . . . . . . . . . . 62 Whole cell fermentation . . . . 62 Induction of enzymes by L—mannose r~ and L-rhamnose . . . 65 Properties of L-mannose (L-rhamnose) isomerase. . . . . . . 65 Activity in crude extracts . . 65 Partial fractionation. . . . 65 Substrate specificity. . . . 77 Metal*ion actiyation: effect of Mn and Co . . . . 7 Effect of mixing substrates. . 1 An isomeraseless Mutant (B- 22). 81 PH Optimum . . . . . . . 87 Temperature optimum . . 87 Properties of L-fructose (L-rhamnulose) kinase. , . . . . . . . 92 Activity in crude extracts . . 92 Partial purification of the kinase. . . 92 Km of L-fructose (L-rhamnulose) kinase. . . . . . . 92 Substrate specificity. . . . 92 Effect of mixing substrates. . 92 pH optimum . . 104 Properties of L- fructose- l- P (L- rhamnulose- l- P) aldolase. . . 104 Activity in crude extracts . . 104 Partial purification of the is aldolase . . . . , , 104 Km of the aldolase. . . . . 104 Effect of mixing substrates. . 104 Substrate specificity. . . . 116 Discussion . . . . . . . . . 116 COMPARISON OF THE UTILIZATION OF L- MANNOSE BY WILD-TYPE A. AEROGENES AND THE L-MANNOSE-POSITIVE MUTANT. . 122 Experimental Procedures . . . . . 122 iv REFERENCES Results. . . Growth of wild-type cells on D- glucose, L- -rhamnose, and L- mannose. . . . . . . . Selection of the L—mannose-positive mutant . . . . . . . . Stability of the L-mannose-positive mutant . . . Response of the wild type and mutant to Imvic tests and a specific phage. . . . . . . . Diauxie curves--effect of L-mannose on the growth of the wild type and mutant. . . . . . . Hexose utilization as a function of growth . . . . . Induction of L-mannose degradative enzymes in the wild type . . Measurement of enzymatic activity upon removal of the inducer . Effect of protein inhibitors on the utilization of L-mannose and L-rhamnose . . . . . Contribution of glyceric acid to L-glyceraldehyde metabolism . Effect of glycerol on the growth of the wild-type and mutant cells . . . . . . . . Effect of DL-glyceraldehyde on the growth of the wild-type and mutant cells . . . . . . Growth of the wild type and mutant on L-galactose . . . . . Discussion . . . . . . . . Summary . . . . . . . . . Page 123 123 123 126 126 126 141 148 148 154 159 159 160 160 166 172 174 Table I. II. III. IV. VI. VII. VIII. IX. XI. XII. XIII. XIV. LIST OF TABLES Water content of L—fructose . . . . . . Phosphate to fructose ratio of L-fructose-l -Po 0 o o o o o o o o o o 0 Induction of L-mannose and L-rhamnose enzymes in Aerobacter aerogenes . . . . . . Substrate specificity of the isomerase . . Effect of various metal ions on the isomeriza- tion of L-mannose . . . . . . . . Effect of various metal ions on the isomeriza- tion of L-rhamnose . . . . . . . . Effect of Mn++ and Co++ on the isomerization of L-mannose and L-rhamnose . . . . . Effect of mixing substrates on isomerase aCtj-Vity. o o o o o o o o o o 0 Enzyme activities in extracts of wild type (PRL-Rj) and a L-mannose/L-rhamnose- negative mutant . . . . . . . . . Partial purification of L-fructose (L- rhamnulose) kinase from wild-type cells . Substrate specificity of the kinase . . . Effect of mixing substrates on kinase activity . . . . . . . . . . . Partial purification of L-fructose-l-P (L-rhamnulose-l-P) aldolase from the L—mannose-positive mutant. . . . Effect of mixing substrates on aldolase activity. . . . . . . . . . . . Substrate specificity of the aldolase. . . vi Page .32 38 70 78 79. 80 84 85 86 95 102 105 111 ‘ 117 118 Table XVII. XVIII. XIX. Page Response of the wild type and mutant to Imvic tests . . . . . . . . 131 Induction of L-mannose enzymes in wild- type cells . . . . . . . . . 151 Determining the number of generations upon removal of the inducer. . . . 152 Measurement of enzyme activity upon removal of the inducer . . . . . 15} Induction of L-glyceraldehyde reduc- tase o o O o o o o o o o o 163 LIST OF FIGURES Figure 1. Synthesis of L-mannose from L-arabinose . . . 2. L-Mannose stimulated C02 evolution from bicar- bonate in the presence of ATP . . . . . . 3. Titrametric measurement of acid production from ATP in the presence of L-mannose. . . . . 4. ATP-stimulated disappearance of L-mannose . . 5. Lactate-dehydrogenase-pyruvate kinase-linked assay for the apparent phosphorylation of L-mannose with ATP . . . . . . ._ . . 6. Chromatography of the products of L-mannose isomerization . . . . . . . . . . . 7. Equilibrium of the L-mannose 2i L-fructose interconversion . . . . . . . . . . 8. Crystals of L-fructose. . . . . . . . . 9. Enzymatic preparation of L-fructose--genera1 outline. . . . . . . . . . . . . 10. Growth of A, aerogenes PRL-Rj on L-fructose. . ll. Enzymatic preparation of L-fructose-l-P-- general outline . . . . . . . . . . l2. Acid hydrolysis of D-fructose-l-P, D-fructose— 6-P, and the product of L-fructose phos- phorylation . . . . . . . . . . . . 13. Gas chromatography: preparation of the tri- methylsilyl derivatives-of fructose-l-P . . 14. Triose formation from L-fructose-l-P . . . . 15. Pathway of L-mannose degradation in Aerobacter aerogenes PRL-R3 . . . . . . . . . . 16. Equilibrium of the L-rhamnose :! L-rhamnulose interconversion . . . . . . . . . . vii Page 10 14 16 18 2O 23 25 29 30 35 37a 40 43 45 60 Figure Page 17. Fractionation of L-rhamnulose-l-P on Dowex-l-formate . . . . . . . . . . 64 18. ‘Whole cell fermentation. . . . . . . , 67 19. Whole cell fermentation of D-glucose, L- mannose, and L-rhamnose . . . . . . . 69 20. Activity of L-mannose (L-rhamnose) isomerase in crude extract . . . . . . . . . . 72 21. Fractionation of the isomerase on Sephadex (5-100 0 e o e o o e o e e e o e 7).} 22. Fractionation of the isomerase on DEAE- Sephadex o o e e e e e e e o o o 76 . ++ ++ 23. Effect of varying the Mn and Co concen- trations on isomerase activity . . . . . 83 24. pH optimum of the isomerase . . . . . . 89 25. Temperature optimum of the isomerase . . . 91 26. Activity of L—fructose (L-rhamnulose) kinase in crude extracts . . . . . . . . . 94 27. Fractionation of the kinase on Sephadex 6.100 a Q o o e o e e e e o o e 97 28. Lineweaver-Burk plot of L—fructose (L- rhamnulose) kinase from L-mannose-positive cells 0 o e o O o o o o e o e e 99 29. Lineweaver-Burk plot of L-fructose (L- rhamnulose) kinase from wild-type cells . . 101 30. pH optimum of the kinase in wild-type cells . 106 31. pH optimum of the kinase in the L-mannose- positive cells . . . . . . . . . . 108 32. Activity of L-fructose-l-P (L-rhamnulose-l-P) aldolase in crude extracts . . . . . . 110 33. Fractionation of the aldolase on Sephadex G-lOO e e e o o o o e e e o o o 113 viii Figure 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. an. 45. 46. 47. Lineweaver-Burk plot relating aldolase reaction velocity to substrate concen- tration O O C O O O O O O O O Biodegradative pathways of L-mannose and L- rhamnose in A, aerggenes PRL-R3 . . . Growth of wild-type cells on D-glucose, L- rhamnose, and L-mannose . . . . . . Selection of the L-mannose-positive mutant. Stability of the L-mannose-positive mutant. Phage specificity of A, aerogenes . . . Growth of wild-type A, aerogenes on D-glucose, and mixtures of D-glucose and L-rhamnose, and D-glucose and L—mannose . . . . . Growth of the L-mannose-positive strain on D-glucose and mixtures of D—glucose and L-rhamnose, and D-glucose and L-mannose . Growth of the wild-type cells on L-rhamnose, L-mannose, and a mixture of these L—hexoses . Growth of the L-mannose-positive strain on L- rhamnose, L-mannose, and a mixture of these hexoses . . . . . . . . . . . Determination of L—mannose and L-rhamnose utilization by actively growing wild-type cells C O O O O O O O O O O O Utilization of L-rhamnose, L—mannose, and a mixture of these L-hexoses by actively growing L-mannose-positive cells . . . Growth of wild-type and L—mannose-positive cells in nutrient broth supplemented with L-mannose e e e o e e e e o e 0 Effect of chloramphenicol and puromycin on enzyme induction and hexose utilization in the mutant after four generations of growth in the absence of inducer . . . ix Page 115 119 125 128 130 133 136 138 140 143 145 147 150 156 Figure Page 48. Effect of chloramphenicol and puromycin on enzyme induction and hexose utiliza- tion in the mutant after seven generations of growth in the absence of inducer . . . . 158 49. Growth of A, aerogenes on glycerol . . . . . 162 50. Growth of A, aerogenes in nutrient broth supplemented with varying amounts of DL- glyceraldehyde . . . . . . . .. . . . 165 51. Growth of A, aerogeneg on L-galactose. . . . 168 INTRODUCTION Aerobacte£_aerogenes PRL-R3 readily gains the ability to utilize rare hexoses, pentoses, and pentitols such as L—lyxose (l), L—xylose (1,2), Learabitol (3), xylitol (3), D-lyxose (4), and D-allose (5) as sole sources of carbon and energy. Investigations notably in the laboratories of Lin (6),‘Wood (7), and Mortlock (8,9) have shown that the biodegradation of these rare compounds does not involve the synthesis of new enzymes, but rather the selection of constitutive mutants containing high levels of non-specific enzymes capable of converting the uncommon sugars into readily metabolizable intermediates. The purpose of this present investigation was to elucidate the biodegradative pathway of the unnatural hex- ose Lemannose in A, aerogenes PRL-R3, to characterize the enzymes involved in the pathway, and to establish the basis for the gain in the ability to grow on this hexose. PART I The Biodegradation of LeMannose by Aerobacter Aerogenes Aerobacter aerogenes PRL-R3 readily gains the ability to utilize L-mannose as a sole carbon source. Except for a recent report (10) that liver galactose dehydrogenase oxidizes L-mannose, the metabolism of this sugar has not been previously described for any organism. This portion of the thesis elucidates the biodegradative pathway of L- mannose in A, aerogenes PRL-R3. EXPERIMENTAL PROCEDURE growth of Cells and Preparation of Extracts- A strain of A, aerogenes PRL-R3 selected for its ability to grow readily on L-mannose was used. The selection of this mutant will be described in Part III. It was grown aero- bically at 30° in a medium consisting of 0.71% Na2HPO4, 0.15% KH2P04, 0.3% (NH.)250., 0.01% M9804, 0.0005% FeSO.‘ 7H20, and 0.4% sugar (autoclaved separately). The sugar was L-mannose unless noted otherwise. The cells were harvested by centrifugation after 15-18 hours growth in Fernbach flasks on a rotary shaker. They were washed with water, suSpended in 0.02 M tris-HCl buffer (pH 7.6), and disrupted by a 4-minute eXposure in a Raytheon 10-kc sonic oscillator equipped with an icedwater cooling jacket. 2 The cellular debris was removed by centrifugation at 31,000 x g for 10 minutes. The resulting supernatant was the crude extract. Analytigalggrocedures- Reducing sugars were deter- mined by the method of Folin and Malmrose (ll). L—Fructose and L—fructose-l-P were determined by the method of Roe (12); the molar extinction of L-fructose-l-P was 80% that of fructose. Inorganic orth0phosphate was determined by the method of Fiske and SubbaRow (l3), and total phos- phate by the method of Umbreit, Burris, and Stauffer (l4). Trioses were determined by the method of Sibley and Lehn- inger (15). Reducing sugars were chromatographed on‘What- man No. 1 paper and developed with the following solvent systems: 1) water-saturated phenol; and 2) 2-butanone: acetic acid:water (75:25:10). The sugars were located with a bath of silver nitrate (l6) and Sprays of orcinol (l7) and N,N—dimethyl-p-phenylenediamine monohydrochloride (18). Sugar phosphates were chromatographed on Whatman No. 1 paper (washed with 2 N HCl and water) using Arbutyl alcdhol: water:picric acid (80:20:2) as the develOping solvent (19). The location of the compounds also was determined with a bath of silver nitrate. Glycerol and glyceric acid were chromatographed on Whatman No. 1 paper using ethyl acetate: pyridine:water (l20:50:40) as the developing solvent. The spots were located by baths of benzidine hydrochloride and of periodate (20). Protein.was measured by the method of Tombs, Souter, and Maclagan (21). Trimethylsilyl derivatives of D- and L-fructose-l-P were prepared and subjected to gas chromatography by the method of Wells g§_§A;(22). Gas chromatography was performed on a Hewlett and Packard gas chromatograph, model 402. Melting points were determined with a Kofler miro-melting point appara— tus. Optical rotations were made with a Zeiss photo- electric polarimeter or a Bendix digital pelarimeter. Light measurements were measured on a Coleman Junior Spectr0photometer (18-mm diameter round cuvettes) or a Gilford absorbance recording SpectrOphotometer (1.0-cm light path) thermostated at 25°. Manometric measurements were made with a conventiona1.Warburg reapirometer. §ngymejAssay§r The reaction mixture (0.58 ml) for Lemannose isomerase contained 16 umoles of L-mannose, 2 umoles of COClg, 8 umoles of tris-HCl buffer (pH 7.6), and enzyme. The mixture was incubated at 30°, and samples were removed at time intervals and assayed for L-fructose: the values were corrected for slight interference from L-mannose. The amounts of enzyme assayed for the time periods of incubation were selected to give linear measure- ments. .A unit of L-mannose isomerase was defined as the amount that formed 1 umole of L-fructose per minute in this assay. The L-fructose kinase activity was measured Spectro- photometrically at 340 nm and 25°. The reaction mixture (0.15 ml) consisted of 1.5 umoles of L-fructose, 0.5 umole of ATP, 1.0 pmole of MgCle, 0.5 umole of phOSphoenOl- pyruvate, 0.01 umole of NADH, 2.0 umoles of tris-HCl buffer (pH 7.6), 13 ug of lactate dehydrogenase-pyruvate kinase, and a limiting amount of L-fructose kinase preparation. A control to correct for NADH oxidase and ATTase contained all of the reaction components except L-fructose. The absence of erructose reductase activity in the extracts and fractions made a control without ATP unnecessary. The assay was linear with time and enzyme concentration. A unit of L-fructose kinase activity was defined as the amount that resulted in the oxidation of l umole of NADH per minute in this assay. LPFructose-l—P aldolase activity also was measured spectrOphotometrically at 340 nm and 25°. The reaction mixture (0.15 ml) consisted of 1.5 umole of L-fructose- l-P, 0.01 umole of NADH, 2.0 umoles of tris-HCl buffer (pH 7.6), 1 pg ofcx-glycerol phosphate dehydrogenase, and a limiting amount of L-fructose-l-P aldolase preparation.) A control to correct for NADH oxidase contained all of the reaction components except L-fructose-l-P. The assay was linear with time and enzyme concentration. A unit of L- fructose-l-P aldolase was defined as the amount that resulted in the oxidation of l umole of NADH per minute in this assay. L-Glyceraldehyde reductase activity was measured Spectrophotometrically at 340 nm and 25°. The reaction mixture (0.15 ml) consisted of 1.5 nmoles of L-glyceral- dehyde, 0.01pnmfle of NADH, 2 umoles of tris-HCl buffer (pH 7.6), and a limiting amount of reductase (45-60% (NH4)2504 fraction). Reagents- LmMannose'was prepared by the method of Sowden and Fischer (23) and L-glyceraldehyde by the method of Perlin and Brice (24). Details for the prepara— tion of these compounds are given below. D—Fructose-l—P, D-fructose-6-P, D-fructose-l,6-diP, and o-glycerol phos- phate dehydrogenase were purchased from Calbiochem, Los Angeles, California; ATP, NAD, NADH, NADP, NADPH, and phosphoenolpyruvate from P-L Biochemicals, Milwaukee, Wisconsin; lactate dehydrogenase-pyruvate kinase from Worthington Biochemical Corporation, Freehold, New Jersey: wheat genm acid phosphatase and protamine sulfate from Sigma Chemical Co., St. Louis, Missouri. L-Fructose for use as seed crystals was a generous gift from Profes—. sor M.L.‘Wolfrom, Ohio State University. Duolite resins C-25 (Na*) and A-6(ClI)‘were purchased from the Diamond Alkali 00., Redwood City, California. They were converted to the H+ and OH- forms reSpectively and mixed in a ratio of 3:5 (wet weight) cation:anion prior to preparation 0f the column. Aggparatign of L-Mannose- LeArabinose (150 g), 300 m1 of absolute methanol, and 540 ml of mitromethane were stirred rapidly in a 3-liter, 3-necked flask fitted 'with a mechanical stirrer and drying tube. To this was added a solution of sodium methoxide, prepared by dissol- ving 32 g of sodium metal in 1050 ml of absolute methanol. The mixture was stirred approximately 20 hours during which time the reaction mixture changed from a white to yellow- brown suspension. The sodium Eggfnitroalcohols were col- lected by suction filtration and washed with 450 ml each of cold absolute methanol and cold petroleum ether B. The nitroalcohols were dissolved in 1 liter of water at 0° and immediately were added dropwise to a stirred solu- tion of 210 ml of sulfuric acid in 250 m1 of water at room temperature. During the course of adding the nitroalcohol solution, the temperature of the acid solution did not rise above 43°. The resulting solution was diluted to 3000 ml with water and solid sodium carbonate added batch— wise until the solution became neutral to congo red. The mixture then was treated with a solution containing 105 ml of phenylhydrazine in 240 ml of glacial acetic acid and the resulting solution left for approximately 12 hours at 4°. The Lemannose phenylhydrazone which precipitated was collected by filtration, washed thoroughly with water, with 95% ethanol, and finally with anhydrous ether. The crude hydrazone amounted to 110 g, melting point 184-1860. The L-mannose phenylhydrazone was suspended in a solution containing 1100 ml of water, 220 m1 of ethanol, 140 ml of benzaldehyde, and 14 g of benzoic acid. The reaction mixture was refluxed for 2.5 hours, cooled to room temperature, and the aqueous phase decanted from the benzyl phenylhydrazone. The solution next was extracted three times with chloroform (500 ml of chloroform per 500 m1 of solution), decolorized with Darco G-60, and concen- trated in a vacuum to a syrup. To crystallize the L- mannose, the syrup was dissolved in a volume of warm absolute methanol equal to twice the volume of the syrup. Next, a volume of absolute methanol and iSOprOpyl alcohol (50:50) equal to the previous volume of methanol was added. The solution was seeded with L-mannose and left overnight at room.temperature. The L-mannose crystallized readily, but scratching the inside of the container with a glass rod hastened further crystallization. The Irmannose was collected by suction filtration and washed with a 75:25 (v/v) solution of absolute methanol and iSOprOpyl alcohol. The filtrate was reprocessed to Obtain additional Lemannose. The final yield of L-mannose was 45-50 9 (30-33%). The melting point was l24-l28° and the Specific rotation was Ealg$8 -l4.2° (g_l, water). A summary of the reactions for the synthesis of Lemannose is outlined in Figure l. grgparation of L-Glyceraldehyde- L—Sorbose (5.0 g) was dissolved in 10 ml of water and the resulting solution diluted to 500 ml with glacial acetic acid. After the solution was cooled with an external water-bath to.l7°, 28 g of lead tetraacetate was added over a period of 5—10 minutes with good stirring. During this time the tempera- ture did not exceed 22°. Next, a solution of oxalic acid (5.0 g of anhydrous oxalic acid in 50 m1 of glacial acetic acid) was added until a negative starch-iodide test was Obtained. The insoluble lead oxalate was removed by fil- tering the solution through filter aid. The filtrate then was concentrated in a vacuum to a syrup. Small amounts of acetic acid subsequently were removed by adding 20 ml of toluene followed by concentration to a syrup in a vacuum: this Operation was repeated several times. The syrup was' dissolved in 25 ml of 0.1 N sulfuric acid and the resulting solution stored for 24 hours at 35°. Finally the solution Figure l. SYNTHESIS OF L-MANNOSE FROM L-ARABINOSE H- =0 H- -OH CH3N02 HO- -H -—————- HO-g-H NaOCH3 H20H LnArablnose _———a- H N02 H06 - H... HO- HO- -OH -H -H H20H 1-De0xy-1-n1tr0-L-glucitol 0H N02 H-q- H H- -OH HO- -H Ho-q-H CHZOH 1) 112304 .__.2) Na2C03-——e 3) Phenyl- hydrazine 1-Deoxy-1-n1tr0-L-mann1tol -OH -H -H 03203 HO- H... HO- HO- L-Glucose phenylhydrazone (soluble) 0HeNNHC6H5 H-C-OH H- -OH 30- -H Ho-q-H CHZOH L-Mannose phenylhydrazone (insoluble) lO CH=0 H-¢-0H 1) C6HSCH=O H-q-OH ___> Ho-q-H 0003 Ho-q-H 2) 06H CHZOH 5 3) 03303203 L-Mannose 11 was passed through a column of Duolite C—25(H*) and A-6(OH_) and the neutral effluent concentrated in a vacuum to a syrup. The syrup was dissolved in water and chromatographed on paper using Arbutanolzethanol:water (52:32:16) as the develOping solvent (25). The sugar was identified with silver nitrate and showed traces of L-sorbose. Since L- sorbose did not interfere in the enzymatic assays the crude L-glyceraldehyde was used as such. RESULTS Since a variety of hexoses metabolized by A, aerogenes PRL-R3 initially are phosphorylated with ATP, my initial investigations on the metabolism of Lemannose‘were concen- trated on determining whether or not Lemannose also was phosphorylated with ATP. Preliminary investigations. involving manometric, titrametric, and colorimetric tech- niques supported this contention. These studies are summarized below. Apparent Phosphorylation of L-Mannose: .ManometrAgiqLeMannose-Stimuigted COa_Evolution ggom Agcagbonate in the Presence of ATP- 'Wild-type and Lemannose- positive cells were grown on D—glucose and.1wmannose reSpec- tively and the cell-free extracts assayed for their ability to stimulate CO; release from bicarbonate due to the phosphorylation of hexoses with.ATP. Since the enzymes 12 of D-glucose metabolism are constitutive, C02 rapidly evolved when extracts from both wild type and mutant were incubated with D-glucose and ATP (Figure 2). On the other hand, Lemannose was metabolized rapidly only by the extract of the mutant cells grown on Lemannose, suggesting that L-mannose degradation involves a phOSphorylation at some point in the pathway. Titrametric: Increased Rate of Acid groduction from Aggin the Presence of LeMannose- The proton released ' from the utilization of.ATT with L—mannose was titrated. Figure 3 shows a slightly greater increase in the activity with Lemannose compared to the endogenous ATPase. To test the validity of the phosphorylating system, D-glucose was added as a control. Colorimetgic: ATgeStimulated Disgppearance offAr Mannose- The disappearance of Lemannose in extracts of Lemannose-grown cells was stimulated by ATP, suggesting the formation of phOSphorylated intermediates (Figure 4). Again, D-glucose was employed as a positive control. SpectrOphotometringest for ArMannokinase— Attempts to demonstrate the direct phOSphorylation of Lemannose yielded negative results, but Figure 5 shows that incubating L—mannose with crude extract prior to adding ATP resulted in measurable kinase activity. This suggested that L- 13 Fig. 2. LeMannose-stimulated C02 evolution from bicar- bonate in the presence of ATP. Each Warburg veSsel con- tained in a volume of 0.5 ml: 10 umoles of hexose (side- arm), 5 umoles of ATP, 25 umoles of M9C12, 5 umoles of NaF, ll umoles of NaHC03, and crude extract (3-5 mg of protein). The reaction was carried out in an atmOSphere of 95% nitrogen: 5% carbon dioxide at a temperature of 30°. Hexose was omitted to measure the endogenous ATPase activity. Controls minus ATP gave no C02 release. Figure 2. _ A I I neocomoecM\Iv I I III. 0 . mo mmoccoZqu 1 1[/300usth o . lo; mHHoo cachetomonnmzlq mMBDZHS m.“ 0.". m meocmwoocM\\llw aromoscmztac mHHoo sackclmmoosaolm 1!:Omoosdoln lm.o lot" NIEIOHd Sm/zoo setomn 14 15 Fig. 3. Titrametric measurement of acid production from.ATP in the presence of Lemannose. The reaction mixture (1.5 ml) contained 20 umoles of hexose, 25 umoles of ATP, 50 umoles of M9C12, and crude extract (10-15 mg of protein). The reaction was carried out at room temperature with a Beckman Zeromatic pH meter. The reaction rate was recorded by adding a measured volume of 0.1 N NaOH to the reaction at a specific time to maintain the pH at 7.5. Endogenous ATPase activity was determined by leaving out the hexose from the reaction. Controls minus ATP gave no rate. Figure 3. umoles NaOH 20 15 10 -O- Endogenous -—O— L-Mannose D-Glucose added \ MINUTES 16 17 Fig. 4. ATP-stimulated disappearance of Lemannose. The reaction mixture (2.0 ml) consisted of 16 umoles of hexose, 20 umoles of ATP, 100 umoles of MgClg, 100 umoles of NaF, 100 umoles of glycylglycine buffer, pH 7.5, and crude extract (20-25 mg of protein). The reaction was incubated at 30° and 0.3 ml aliquots removed at time intervals and added immediately to 0.3 m1 of 5% ZnSO4 followed by the addition of 0.3 ml of 0.3 N Ba(OH)2. The insoluble BaSO4 was removed by centrifugation and 50 ul of the supernatant assayed for remaining reducing sugar. Figure 4. umoles HEXOSE 20 15 10 .A 7‘ N\\\_L-Mannose or D-Glucose (N0 ATP) é,,_..—------—L--Mannose + ATP 8,,a-D-G1uoose + ATP 20 4O 60 MINUTES 18 19 Fig. 5. Lactate-dehydrogenase-pyruvate kinase-linked assay for the apparent phosphorylation of L-mannose with.ATP. (Refer to Assay section on L-fructose kinase for details). Reaction mixtures (70 pl) containing 2.5 pmoles of Lemannose, l pmole of tris- HCl buffer (pH 7.6), l pmole of MgCla, and crude extract (0.024 mg of protein), were incubated for 1 hour at room temperature, after which.ATP, lactate dehydrogenase and NADH were added and activity re- corded (B). The left portion of the graph (A) depicts a control in which.ATP and NADH were added at zero time. Figure 5. mmBDZHS ma OH m 0 ma 0H m A _ _ a .1 _ .I1 m C O . msocowoecm.JL msocmwoecmllldw ll 0H omocsmth Alll.mmoccszuq 0H ma GEZIGIXO HGVN sanmu 2O 21 mannose was converted to another intermediate prior to phosphorylation. Such a conversion might conceivably involve a phosphotransferase reaction as demonstrated by Kamel and Anderson (26). They reported that when D- mannose was incubated with crude extracts, D-glucose accumulated due to phosphorylation of D-mannose with endogenous D-glucose-6-P. In the present investigation, however, Lemannose was found to be isomerized to L-fruc— tose. Isomerization of L-Mannose' to L-Fructose- L—Mannose was incubated with crude extracts and the mixture chro- matographed on paper. Figure 6 shows the accumulation of a spot which co-dhromatographed with authentic D-fructose and gave a positive orcinol test suggesting that L- mannose undergoes an isomerization to L-fructose. The preparation and isolation of the product of Lemannose isomerization is described below. Enzypic Preparation of L—Fructose- The reaction mixture (75 m1) consisted of the following: 28 mmoles of L-mannose, 5 mmoles of COC12, l2 mmoles of tris-HCl buffer (pH 7.6), and cell—free extract (300-350 mg of protein). The reaction was incubated at 300 for 2-3 hours or until equilibrium was established. At equilibrium, the ratio of LemannosezL-fructose was 38:62 (Figure 7). The reaction 22 Fig. 6. Chromatography of the products of Lemannose isomerization. The reaction mixture (1.4 ml) con- sisted of 20 pmoles of Irmannose, 100 pmoles of -glycylglycine buffer (pH 7.5), and a volume of crude extract containing 20 mg of protein. The reaction was incubated at 30° and 0.6 ml aliquots were removed at time intervals (t), heated in a boiling water-bath, and the denatured protein removed by centrifugation. The supernatant was concentrated in a vacuum to about 50 pl and a volume of this solution containing about 0.5 pmole of hexose was Spotted on paper and then deve10ped in.water-saturated phenol for 24 hours. Figure 6 . ooHHOHSoosomsosoa ocdsedo locoHASosaIQIHASpoaaouz.z \<7 o = t oASpHHE codpoeom t=60 min. D-Fructose D-Glucose L-Mannose AOZHomo \\(\()\)§(\/\/\/>Suw>wuo< .uxmu ecu cw pocHHuoo mouse umooud ou wcapuooom goose ucmHuusc no «omocamsunq aomoocmfinq segues co macaw mums mHHoo any mocowouom umuomoouo< ca moewnco mmocaenuua one omoccmanq mo cofiuooch HHH mAmfiuo<* .aflououm mo wE\cHE\uoscoum mo moHoai mm commoumxo mum mufizmmm H0.0 no.0 Hm.o O®.O HO0.0V HO0.0V wm0c¢m2nA AquuDEv No.0 no.0 OM.H OH.~ HO0.0V HO0.0V omocfimnmuu Nmum mo.o mo.o om.H OH.H mo.o HH.O mmocnm21H Ausmummv no.0 HH.o o:.H mm.H no.0 sm.o mmeaemam-a masque m-H- m-H- omouusumaa omoHJcEmAmuA mmouonumuu mmoHsnamnm-A omcssz-A mmonEm£MuA HmosncH cflmnum immmaocfi< *mmmnflm mmmnofiomH .uoumm>umn ohms mHHmo on“ mouzcaE on we oswu toquuoccfi cm swumm com coaumfisoczfl w¢umm meme; 0 cocoa mm3 omo:Emnm IA .nzmop cm; nonummflfiaun mmonCmfi-A cons woumw>umn wum3 mflfloo ago wow scmumanuocfl mum3 mHHwo «no mafia 08mm msu um Bahama ausoum can we cwocm mm: mmoncmzAH .Anomw ma Ommv mmochEuA no wmonEwnH IA uwfiuflm Sues couswEmemsm Luopn unwfluuoa mo HE 00m 5H nachm onwz mHHmo mmum cam mmhu cHHS unmasa o>Hunwoszomommmnkug\omonomaaq m com “mm-ammv damn cHfi3 mo muomnumw ow mowufi>fiuom maNNmm xH mqm<fi 87 was not indiced by either Lamannose or Iwrhamnose, and no isomerase activity was d--ected on either of these two substrates. (9) pH Optimum— The pH Optimum of the isomerase was determined with both L-mannose and Lwrhamnose as substrates and in the presence and absence of their activating metals (Figure 24). In the presence of cd*+. Lemannose was isomerized maximally over a pH range of 4. 7.5-7.7, and Without Co‘ over a pH range of 7.4-8.5. The decrease in the pH Optimum in the presence of Cc>++ probably was due to cobalt hydroxide formation. The shift in the pH Optimum.was more clearly defined when L-rhamnose was the substrate. In the presence of ++ Mn L-rhamnose was isomerized at a maximum rate when 3 , ++ . the pH was 7.3-707 and in the absence of Mn a pH shift to 8.7-9.0 occurred. The rapid decrease in L-rhamnose isomerization after pH 7.? probably was due to manganese dioxide formation. (h) Temperattre Ootimsm- With Iwrhamnose as the substrate and Mnfl+ as the activating metal, the isomerase exhibited a temperature Optimum of 60-650 compared to . +4." . . 55—600 in the absence of.Mn (Figure 25). With L—mannose as substrate the isomerase in the presence or absence of Coh+ had a temperature Optimum of 50-550. Fig. 24. The pH Optimum of the isomerase. The stan- dard assay for measuring isomerase activity was employed. The crude extract was fractionated on Sephadex G~lOO and the fraction having the highest isomerase activity was used. The amount of protein per assay was 0.52 mg. Each of the buffers employed was 0.02 M and had the following pH ranges: sodium phoSphate (pH 6.7-7.5), tris-HCl (pH 7.4-8.8), gly- cine-NaOH (pH 8.9-9.4)9 and carbonate-bicarbonate (pH 9.8-lO.l). The pH of the reaction was record-d at the end of the incubation period. umoles LuFBUCTOSE/BO MIN umoles L-RHAMNULOSE/lo MIN Figure 1.5 0.5 1.5 0.5 24. LmMANNOSE ._ CO++ o— No Metal e/o’o—O—L a 1 ’.L~RHAMNOSE Q_.Mn++ O-No Metal 7.0 8.0 9.0 10.0 pH 89 90 Fig. 25. Temperature Optimum of the isomerase. The enzyme was fractionated on Sephadex GelOO before use. To measure isomerase a-tivity the assay mixture con- taining all the components except the substrate was heated for five inates at a Specific temperature, after which the substrate was added to initiate the reaction. When Lwrhamnose was the snbstrate, the assay contained 0.17 mg of protein; when meannose was the substrate, the reaction mixture contained 0.52 mg of protein. Figure 25. Aoov mmseammmsme om on om om 0: on _ fl _ Hmpmz oz «0 ++oo :0 _ Mmozz<21A o.m 0.: MIN OC/HSOLOflHfl‘T setomn Aoov mmseammmzmea om on om on 0:. on _ _ _ Haves 02 IO ++sz I o mmozz¢mmlq 0H NH ad 0H NIH OT/HSOTHNNVHH”T setomfi 91 92 Properties of LoFructose (L-Rhamnulose) Kinase: (a) Activity of the Kinase in Crude Extracts- The rates of phOSphorylation of L-fructose and L-rhamnulose were compared as a ftnction of kinase concentration in crude extracts of both wild-type and mutant cells (Figure 26). Although the kinase activity varied in each extract. the ratio of the activities of L-fructose to L-rhamnulose remained constant. (b) Partial Purification of the Kinase- The kinase was purified about 7—fold with a 20% recovery of activity (Table X). Fractionation with ammonium sulfate and Sepha- dex G-100 failed to separate the L-fructose from the L—rhamnulose kinase activity (Figure 27). (c) Km of LmFructose (L-RhamnuloselKinase- The Km's for both L-fructose and Lmrhamnulose were the same for partially purified kinase obtained from both wild type and mutant cells (Figures 28 and 29). The kinase bound Lmrhamnulose more strongly than L~fructose. The Km value for L-rhamnulose was 0.05 mM compared to 1.7 mM for L- fructose. (d) Substrate Specificitye The kinase was Specific for L-fructose and Lmrhamnulose (Table XI). Activity on. D-fructose was attributed to D-fructose kinase. (e) Effect of Mixing Substrates- Kinase activity on L-fructose and Lmrhamnose was not additive (Table XII) - uulfii“. ll. . m: “Mb | .v' III“~ r‘m‘Irb‘ 93 Fig. 26. Activity of L—fructose (L-rhamnulose) kinase in crude extracts. The standard kinase assay was employed except for the change in protein concentration. Figure 26 . szBomm ma 3.0 m.o N.o Hoo omoassamnmaq Z 7 mmopogmnq BZdHDS om o.Q:.§.m£maaH\v 4f! omopofihmaq mmHB QHHz on or om NIH/deVEmS setomu 94 S O/ .muzawa nod pmumamuosamoad macasdswsuuq no mmouosumng mo mmaoaouowz.* Hm.m mm.m ww.o Hw.o hm.o Om.o macaadamsuua omou03HMuA afimuoum‘ a\muwcb Hm ovumw OOH :H ma m: mm mm as macassamnuua mmouosumnq Ll. *muHGD ooH-o xmemsamm mummasm aswaoaa€ uumuuxm dunno sua>auu< oawdumam zum>oomm mufi>wuu< Hmuoe coauomum mHHmu omhunpaws scum ommaax AeneasdEmnuulq‘mmouusuwua mo dowumowmausm Hmfiuumm N mamProtein .— L-Fructose 9- L-Bhamulose WILD TYPE __ 60 FRACTION NO. 97 nmoles ADP/5 MIN “'1 l 1rillrt.fv\ vne, ' F. a. v 98 Fig. 28. Lineweaver-Burk plot of L-fructose (L- rhamnulose) kinase from L-mannose-positive cells. The standard kinase assay was used except that the substrate concentration was varied as indicated with the kinase (Sephadex G—100 fraction) concentration constant. Figure 28. Km for L-Fructose = 1.60 x 10-3M L-Fructose (mm) for L-Bhamnulose = 4.3 x 10-5M .l x 10”3 v 0.5 ' I l l l l o 0.1 0.2 0.3 0.4 0.5 1 L-Bhamnulose (mm) 99 .eui I.‘ L‘w‘ . 100 Fig. 29. Lineweaver—Burk plot of L—fructose (L- rhamnulose) kinase from wild-type cells. Figure 29 . Km for L-Fructose = 1.67 x 10‘3M 2.0... 3]? x 10'"3 1.0__ / I I I I I o 1 2 3 4 5 ___A 1 L-Fructose (mMT Km for L-Rhamnulose = 5.2 x 10-5M 1.0— % x 10"3 0.05-—— l J l l I 0 001 002 003 001* 0.5 1 L-Bhamnulose (mMT 101 102 TABLE XI Substrate specificity of the kinase The routine kinase assay was employed. With the exception of L-rhamnulose the assay contained 1.5 pmoles of each substrate. The enzyme was fractionated on Sephadex G-100 before use. L-Rhamnose-Grown Cells LdMannose—Grown Cells Substrate Comparative Rate (%) Comparative Rate (fi) L-Rhamulose 100 100 L-Fructose 110 110 D-Fructose 1.8 2.5 L-Sorbose 0 0 L4Mannose 0 0 105 TABLE XII Effect of mixing substrates on kinase activity The routine kinase assay was used. 'When both substrates were mixed the concentration of each substrate was the same as that used in the routine assay. The kinase was fractionated on Sephadex G-100 before use. Growth Substrate Substrate L-Rhamnose L4Mannose Specific Activity (pmoleslmin/mg) L-Rhamnulose 1.76 1.h3 L-Fructose 1.95 1.60 L-Rhamnulose-+ L-Fructose 1.85 1.55 104 when assayed from cells grown on either L—mannose or L-rhamnose. (f) pH Optimum- The pH Optimum of the kinase ranged from pH 7.0-8.0 in both wild—type and mutant cells when either L-fructose or L-rhamnulose was the substrate (Fig- urea 30 and 31). PrOpertie; of L-Fructose-l:g_(L-Rhamnulose-l-P) Aldolase: (a) Activity in Crude Extracts— The rates of cleavage of L-fructose-l-P and Lprhamnulose-l-P were compared as a function of aldolase concentration in crude extracts from both wild-type and mutant cells (Figure 32). (b) Partial Purification of the Aldolase- The aldo- lase was purified about 6—fold with a 20% recovery of total units (Table XIII). Fractionation with ammonium sulfate and Sephadex G-100 failed to separate the L- fructose-l-P from the L-rhamnulose-l-P aldolase activity (Figure 33). (c) Km of the Aldolase- The aldolase bound Lprhamnu- lose-l-P 10 times more strongly than Lpfructose-l-P. The Km value for L-rhamnulose-l-P was 0.5 mM compared to 5 mM for L-fructose-l-P (Figure 34). (d) Effect of Mixing Substrates- ‘When L-fructose-l—P and Lprhamnulose-l-P were mixed together, their combined l‘ ne'er." 105 Fig. 30. pH Optimum of the kinase in wild-type cells. The routine kinase assay was used. The kinase was fractionated on Sephadex G-100 before use. Details of the buffers and pH ranges used are given in Figure 24. The pH of the reaction was recorded at the end of the incubation period. The amount of protein per assay was 0.08 mg. Figure 30. L-RHAMNULOSE 20... -0- Tris-H01 10 _ —O — Ph08phate a -I - Glycine-NaOH ”Q a I - . l I I 3 L-FHUCTOSE H E 30.__ 20.__ (M 10,—— I I I I 6 7 8 9 pH 106 107 Fig. 31. pH Optimum of the kinase in the Lemannose- positive cells. Refer to Figure 30 for details. The amount of protein per assay was 0.04 mg. Figure 31. 30..— 20... 10 ___ -O-—Tris.HCl -9 -PhOSPhate a - I - Glyc ins-NaOH 2 Q _- a l A f I l I m L-FBUCTOSE‘ .9. E3 30 __. 20 — l 0 _ F— — pH 108 109 “E tans”— " Fig. 32. Activity of L-fructose—l-P (L-rhamnulose- l-P) in crude extracts. The routine aldolase assay was used except that the protein was varied. Figure 32. szeomm ms 30.0 «0.0 o mIHIomoposHmlq .\\IJW mIHImmOHsssdsmlq NAME QAHz 30.0 No.0 mIHIomopoohmIA mnflummoasnamsmuq BZdBD: NIH/JVHG satomu 110 111 .zmmmm pumuamum msu cw 0mm um munowa Hon pouflpaxo mmwuu< oamaommm zum>oomm zua>wuo< kuOH dowuomnm udmusfi w>fiuamo Immoodwaua map Eoum wmmaovam NmIH-mmoHscsmnuuav muaummouoonwng mo dofiumoamwus Hmauumm HHHN mam¢a 112 Fig. 33. Fractionation of the aldolase on Sephadex G-100. The routine aldolase assay was employed. Details of the procedure are outlined in the text. Each fraction was assayed for activity on L-fructose- l-P and L-rhamnulose-l—P. Figure 33 . mg PROTEIN/FRACT ION MUTANT 16" o-Prote1n "" 4 o- L—Fructose-l-P 111__ o- L-Rhamnulose-l -P 12 —- —. 3 O 10.— 8 L_ _ 2 6_ ' .\ 1+... ’ \ ° ._. 1 \\ O O 2__ \\ . \ O ‘O\ o I I I \ 0 LI 5 6 7 FRACTION NO . 113 pmoles DHAP PRODUCED/MIN/FRACTION 114 Fig. 34. Lineweaver-Burk plot relating aldolase reaction velocity to substrate concentration. The routine assay was used except that the substrate was varied as indicated with the aldolase (Sephadex G-100 fraction) concentration constant. The enzyme was obtained from cells previously grown on L-mannose. Figure 34. Km for L-Bhamnulose-l-P = 5.0 x 10' / I I I 0 0.1 .0.2 0.3 .‘ . 1 L-Rhamnulose-l-P (mm) Km for Fructose-l-P = 5.0 x 10‘3M 8.0.—— 6.0.... l x 1.0"3 4.0—— 2.0 —" I I I 0 2 4 6 L-Fructose Imfi) 115 116 aldolase activities were not additive (Table XIV). (e) Substrate Specifigityr The aldolase when purified from both.wild-type and mutant cells had the same substrate Specificity (Table XV). It cleaved only L- rhamnulose-l-P and L—fructose-l-P, and did not cleave D-fructose-l-P or D-glucose-l-P. Activity on the other substrates was due to contaminating enzymes in the preparation and are listed in the table. DISCUSSION The evidence Obtained in Part II of this thesis indicates that the same enzymes degrade L-mannose and L—rhamnose (Figure 35). They were induced by Lemannose and Lnrhamnose in both the‘wildatype and mutant cells and the ratios of the Specific activities of the individual enzymes for Lemannose, L-rhamnose, and their metabolic intermediates were the same in each strain. In addition, whether the enzymes were isolated from the wild-type or mutant cells, the activities on Lemannose or L-rhamnose could not be separated when subjected to partial purifi- cation. The enzymes showed the same substrate specificity when induced by either Lphexose and their individual Specific activities were not additive when measured in - the presence of both substrates. Finally, neither L- mannose nor Lprhamnose was able to induce the isomerase 117 TABLE XIV Effect of mixing substrates on aldolase activity The routine aldolase assay was employed. When both sub- strates were mixed, the concentration of each substrate was the same as that used in the routine assay. The aldolase was frac- tionated previously on Sephadex G-100 before use. Substrate Specific Activity (pmoles DHAP produced/ min/mg protein) L-Fructose-l-PO4 0.20 L-Rhamnulose-l-P04 0.33 L-Fructose-l-P04 + L-Rhamnulose-l-P04 0.24 118 TABLE XV Substrate specificity of the aldolase The routine aldolase assay was used. With the exception of L- rhamnulose-l-P, the assay contained 1.5 nmoles of each substrate when tested. Contaminating activities are listed in cases where activity was expressed but was not due to cleavage by the aldolase. Substrate Comparative Rate (%) L-Rhamnulose-l-P 100 L-Fructose-l-P h0-60 D-Fructose-l-P 0 D-Glucose-l-P 0 D-Fructose-6-P mannitol-l-P dehydrogenase activity D-Glucose-6-P D-Fructose-6-P isomerase activity D-Fructose-lr, 6-diP glycolytic aldolase (trace) Figure 35. mmwmmmfldfioddlq madmmmomm mzoamodeommfimHm mmwmmqqdmmquclg madmmwcmm MZOBHU¢HXOmQHmHQ mIHImmOADzzdmmIA Alllll. mmOADzzwuo< .oummaom soaaoaam nuHB woaumGOwuomum Houwo poawsuouop whoa moaua>wuom ommfiopam 0am mmmcax 0:9 * 00.0 00.0 0m.s 0H.H 00.0 HH.0 mmoaamzwa no.0 HH.0 0:.H mm.a s0.0 sm.0 mmoasmam-a m-~- m-H- 44 mmouosumuu omoHssamAMuA omouosumua anodsssmamug omonsszA omOnEM£MuA *ommaopa< *mmmaaM mmmumEOmH HoosocH . .umo>um£ muomon massage Oh Hmcoauwpvm cm 0x030 Ou posoflam paw omosamnuug mo we 0mm :uHB pmudmswamdsm mums moan aoasa umumm «mason 0-0 you :uoun usoauus: as mHHonnouom saouw whoa mHHoo 0:9 ummossonMuA kn sowuospsH .n .0oosoomu was omoaomsiu an nuaouw mo cowufinwncw cos? woumo>umn one: mHHmo 0:9 .mmosamsrA mo wa_0mn nuwz pouaoEwHQQSm suoun uswwuusc cw zaamownouom aaouw mums mHHwo may nomoaaszq mp coauusoaH .m mHHoo unauupaga aw moshuso omossmaiu mo aoauospoH HH>x manum£ mums magma as» msoaumuosmw so>mm 0cm soon you wdasouw Houm¢ umo:0¢« o£u mo Hm>osuu comm mua>auom umwuso mo uaosousmmmz NHN mgm<8 154 decrease in the level of all three enzymes indicated that the mutant was not constitutive for them. Effect of Protein Inhibitors on the Utilization of §;Mannose and L-Rhamnose- To show further that the first three enzymes were not constitutive, the inducers L-mannose and L-rhamnose were added back to the mutant cells prev- iously grown in the absence of inducers and the rate of their utilization measured manometrically. To distin- guish between constitutive and newly induced enzymes, either chloramphenicol or puromycin was added to the reaction. As Figures 47 and 48 indicate, in the absence of these inhibitors L-rhamnose was readily utilized and reached a maximal rate comparable to D-glucose. L- Mannose also was oxidized but at a slower rate than L- rhamnose. In the presence of either chloramphenicol or puromycin neither Irmannose nor L-rhamnose was utilized. The results thus showed that the oxidation of L-mannose and L-rhamnose by the mutant cells was due to the bio- synthesis of new enzymes and not to a constitutive muta- tion. The results indicated that another mechanism was Operating within the cell to regulate growth on Lsmannose. Since dihydroxyacetone phosphate was readily metabolized, L-glyceraldehyde was restudied to determine if it exerted 155 Fig. 47. Effect of chloramphenicol and puromycin on enzyme induction and hexose utilization in the mutant after four generations of growth in the absence of inducer. Each.Warburg vessel contained in a volume of 0.55 ml, 10 pmoles of either L-rhamnose or L-mannose, 180 pmoles of KOH (center well), 1000 pg of chloramphenicol or 500 pg of puromycin, and about 2 mg dry weight of cells. As a control water was substituted for the inhibiter of protein biosyn- thesis and for endogenous activity water was sub- stituted for the hexose. Figure 47 . 0 "7'" \ 5 D-Glucose 3 (.3 & ° . L-Bhamnose <3 o—l <——/ E! m g o A 2 . - 3'; 5 0— ~ ‘ \K L Mannose . 2 >4 a) E . f! V N— Endogenous H . 1 g) ' / . I ‘ I 0 4o 80 120 MINUTES 50"“ CHLOBAMPHENICOL \ “:3 g o A L-Rhamnose SS 25 . a, N L-Manno se is a: 3 a \ Endogenous H "=13 I I 0 40 80 120 MINUTES 50 .— \m . PUBOMYCIN 8E}: E—c 0 Egg N D-Glucose 2 ~—— -Rh 0 5 L amnose \ >4 '0 m s c: 3V ‘ L M - annose : g) \ Errdogenous :1 , I 0 “0 .«'80 120 MINUTES '156’ 157 Fig. 48. Effect of chloramphenicol and puromycin on enzyme induction and hexose utilization in the mutant after seven generations of growth in the absence of the inducer. Refer to Figure 47 for details. Figure 48. uliters 02 UPTAKE/ mg (DRY WT) CELLS UPTAKE/ mg (DRY WT) CELLS uliters O uliters O UPTAKE/ mg (DRY Hf),CELLs 50 25 50 25 __ °D-Glucose ' L-Bhamnose .‘K/ L-Mannose \u - . v~—Endogenous 0 40 80 120 MINUTES 7. CHLOBAMPHENICOL f’,,D-Gluoose T— L-Rhamnose..:§ L-Mannose “~—Endfigenous 0 40 80 120 50 25 MINUTES PUROMYCIN D-Gluoose <"’/ L-Rhamnose \ L-Mannose K- Endogenoui 1 O 40 80 120 MINUTES 158 159 any effect on the cells. Part I of this thesis showed that L-glyceraldehyde underwent oxidation to glyceric acid and reduction to glycerol. Thus, investigations were conducted to establish if these reactions were instrumental in regulating Limannose metabolism. Since the wild-type cells completely utilized L-mannose with— out growth, the possibility existed that a degradative product of L—mannose which inhibited growth accumulated in the growth medium. Attempts to find such a product failed. Contribution of Glyceric Acid to L-Glyceraldehyde Metabolism- The enzyme which converts L-glyceraldehyde to glyceric acid was detected only by identification of the reaction product (see Part 1). Paper Chromatography of the growth media obtained.from both wild-type and mutant cells actively utilizing L-mannose showed the accumulation of a compound which co-chromatographed with DL-glyceric acid. NWhether or not some of the glyceric acid was metabolized by these cells is not known.. How— ever, since it did accumulate as an excretory product of Irmannose degradation by both cell types, it was eliminated as a possible growth inhibitor. Effect of Glycerol on the Growth of the‘Wild—Type and Mutant Cells- Both the mutant and the wild-type cells 160 grew equally well on glycerol (Figure 49). Although the reduction of L-glyceraldehyde to glycerol could not be assayed in crude extracts because of interfering reac- tions, partial fractionation with ammonium sulfate yielded a 45-60% fraction.which showed reduction Of L-glyceralde- hyde. This enzyme was detected in fractionated extracts Obtained from both wild-type and mutant cells previously grown on L-rhamnose or Lsmannose respectively (Table XX). In addition, L-mannose also induced the enzyme in wild- type cells after they were previously grown on D-glucose. Since both L—hexoses induced the reductase in both cell types, the reduction of Lpglyceraldehyde to glycerol was not considered to be an influential reaction in inhibiting the growth Of the parent strain on Lemannose. Effect of DL-Glyceraldehyde on the Growth of the Wild-Eype and Mutant Cellg: DL-Glyceraldehyde inhibited the growth of both wild-type and mutant cells above a concentration of 0.007 mM. Figure 50 compares the effect of DL-glyceraldehyde on the growth of the cells. In neither case was the inhibitory effect overcome at the higher concentration. No conclusion4was drawn as to which enantiomorph caused the growth inhibition. grgwth of the Wild gype and Mutant on L—Galactose- E, aerogenes re5ponded to growth on L-galactose, a rare 161 Fig. 49. Growth Of E, aerogenes on glycerol. Figure 49. 0.8 __ 0. Wild Type O-Mutant 0.6.— O l-r E“ 0. O 0.4_. 0.2_ l_ I l l l l O 2 4 6 8 10 12 HOURS 162 163 TABLE XX lnduction of L-giyperaldehyde reductase Reductase activity was measured using the routine assay. The enzyme was from a 45-60% ammonium sulfate fraction. Cell Type Growth Substrate Reductase Activity* Wild L-Rhamnose 0.088 D-Glucose and IrMannose 0.020 D-Glucose 0.000 Mutant IwMannose 0.021 D-Glucose 0.000 * Activity is expressed as pmoles NADH oxidized/min/mg of protein. 164 Fig. 50. Growth Of E, aerogenes in nutrient broth supplemented with varying amounts of DL—glyceralde- hyde. Figure 50. 0H 0H 0 2s 4H0.0 28 500.0:l/K 820902 mmDOm N.o :.0 0.0 as 0H0.0_\\\» mmwa QAHz N.0 0.0 owS.q.O 165 166 hexose, in the same way as it did to L-mannose. Although L-galactose and L-mannose differ structurally, they both undergo similar biodegradative patterns and produce L- glyceraldehyde as a major intermediate. L-Galactose metabolism has been established in.E, aerogenes (Mayo and Anderson, unpublished results) and found to be isomerized to L-tagatose, phOSphorylated with ATP to L-tagatose-l-P, and cleaved to dihydroxyacetone phosphate and L-glyceral- dehyde. If the basis for growth on Limannose involved L—glyceraldehyde or a subsequent metabolic intermediate, then the L-mannose positive mutant should also grow on L-galactose more readily than the wild type. As Figure 51 indicates, a 754hour lag preceded growth of the mutant on L-galactose whereas the wild type showed no appreciable growth even after 120 hours. There was some initial growth by both cell types but this was attributed to an unknown contaminent rather than to growth on L-galactose. The mutant cells after growing on L-galactose also grew on L-mannose. DISCUSS ION The growth of E, aerogenes on Lamannose involves a mechanism other than a constitutive mutation for the, first three enzymes in the pathway. The evidence in this investigation suggests that the basis for the gain in the [Eu 4- 167 Fig. 51. Growth of E. aerogenes on L-galactose. Figure 51. 00000 04H 40 0: 0 00H IILNoO 110.0 110.0 1 924902 _ L moo mama and: 30° 0.0 .moO 0175.0.0 ' 168 169 ability of the mutant to grow on Lsmannose lies in its ability to metabolize L-glyceraldehyde or a subsequent metabolic intermediate. The growth of both the wild type and the mutant was inhibited when the cells began to uti- lize Lemannose, but only the mutant overcame the inhibi- tion. This suggested that a compound accumulated within the cells which inhibited their growth rather than the selection of a mutant strain which resisted the inhibi- tory effect Of this product. Since Lemannose induced the isomerase, kinase, and aldolase in both types Of cells, the first three enzymes in the biodegradative pathway were not influential in regulating growth of the cells on Lemannose. In addition, none Of the enzymes was constitutive when the inducer was removed. L-Glyceraldehyde inhibits a variety of cellular processes but the exact site(s) Of inhibition are still questionable. .Mendel (74) was the first to report that L-glyceraldehyde inhibited the formation of lactic acid from glucose in tumor cells. Rudney (75) had shown that hexokinase in rat skeletal muscle, rat sarcoma, beef brain, and yeast also was inhibited by L-glyceraldehyde. In a later report Wenzel, Joel, and Oelkers (76) demonstrated that Lpglyceraldehyde inhibited glycolysis in Ehrlich ascite tumor cells. On the other hand, D-glyceraldehyde 170 has been implicated by Rapkine g£_gl_(77) to inhibit D- glyceraldehyde-3-P dehydrogenase. Possibly DL-glyceral- dehyde is working in a dual capacity to inhibit growth of E, aerogenes. Lardy and coworkers (78) have shown that rabbit muscle aldolase condenses L-glyceraldehyde and dihydroxyacetone phosphate to yield L—sorbose-l-P which strongly inhibits hexokinase. Whether or not a similar reaction occurs in bacteria is not known. Possibly the bacterial glycolytic aldolase is capable of condensing the products Of the L—fructose-l-P cleavage producing L- sorbose-l-P which may be toxic to the cell. If this is the case, the mutant may overcome this toxicity by synthe- sizing an enzyme which converts storbose-l-P to a common metabolite. Although L-glyceraldehyde is simultaneously reduced to glycerol and oxidized to glyceric acid, the enzymes were present in both wild type and mutant. Both cell types grew readily on glycerol and when grown on L- mannose, glyceric acid accumulated in the growth medium. Thus, these reactions were not considered to be significant in regulating Lemannose metabolism.) The mutant cells, however, were capable of growing on L-galactose more readily than the wild type and Lnglyceraldehyde is a 171 common product Of both L-hexoses. Previous investigators have shown that the meta- bolism of unnatural sugars involved a constitutive mutation not for the synthesis Of new enzymes but rather for the production of a non-specific enzyme capable of converting the unnatural substrate to a common metabolic intermediate. Mortlock §£_§E_(7) have shown that growth of E, aerogenes on xylitol and L-arabitol was explainable by the selection Of a mutant constitutive for ribitol dehydrogenase, which converted xylitol and Learabitol to D- and L-xylulose, resPectively. Similarly, growth on L-xylose was due to selection Of a mutant constitutive for L-fucose isomerase (2). In contrast, D-lyxose appeared to be an exception to this generalized phenomenon. Allison and Anderson (4) reported that D-lyxose was isomerized to D-xylulose but they were unable to isolate constitutive mutants for D- lyxose isomerase. The enzyme was induced only by D-lyxose, and although D-mannose also was isomerized by this same enzyme, it was unable to induce it. Lin gt §E_(6) reported that two genetic events regulate the metabolism of mannitol in E, aerogenes. The suppression of the phosphotransferase pathway by which mannitol was normally metabolized gave rise to a constitutive mutation for the production of D-arabitol dehydrogenase which converted 172 mannitol to D-fructose. Other systems involving the utilization of an unnatural substrate by derepression of an enzyme include: altrose-galactoside via a-galactosi- dase (79), B-glycerolphosphate via alkaline phOSphatase (80), and putrescine via diamine41-keoglutarate transami- nase (81). The enzymes of L-mannose metabolism also are non—Specific in that they degrade the naturally occurring hexose, L-rhamnose. However, unlike the previously men— tioned cases, the first three enzymes involved in L- mannose metabolism are not constitutive. To conclude, both mutant and wild-type cells utilized L-mannose and their growth was inhibited by a product of L-fructose-l-P. This inhibitor may be L-glyceraldehyde or some product thereof. However, only the mutant was able to overcome the inhibition resulting in active growth on L-mannose. SUMMARY The biodegradative pathway of L-mannose by E, aerogenes PRL-R3 has been elucidated. LeMannose is isomerized to L-fructose which is phosphorylated with.ATT and a kinase to L-fructose-l-P. LPFructose-l-P is cleaved by an aldolase to dihydroxyacetone phosphate and L- . glyceraldehyde. L-Fructose and a new sugar phOSphate, 173 L-fructose-l-P, were isolated and identified, and an alternative procedure was develOped for the preparation of L-fructose. The enzymes in the pathway have been characterized. They were induced by either Lemannose or L-rhamnose and the biodegradation of these two L-hexoses has been shown to involve the same enzymes. A mutant strain of E, aerogenes has been isolated, which, unlike the wild type, grows readily on Lemannose as the sole carbon source. The mutant is not constitutive for the firstthree enzymes in the biodegradative pathway. Thus, the basis for growth of the mutant on Lemannose possibly lies in its ability to metabolize L-glyceralde- hyde or a product thereof. 10. 11. l2. 13. 14. 15. REFERENCES ANDERSON R. L., and WOOD, w .A., J. Biol. Chem., 2 6( 1962) CAMYRE, K.P. and MORTLOCK, R. P., J. Bacteriol. 2_. 115 57 (1965) FOSSITT, D., MORTLOCK, R_.P., ANDERSON, R. L., and WOOD, W.A., g. Biol. Chem., 259, 2110 (1964). ANDERSON, R. L., and ALLISON, D. P., J. Biol. Chem. , 240, 2567 (1965) GIBBONS, L.N. and SIMPSON, F. J., Can. J. Eicrobiol., 9. 769 (19655 10, 829 (1964) LERNER, s .A., WU _i_. 1515 (19645 MORTLOCK, R. R, FOSSITT, D. D., and woon Natl. Acad. Sci. U. s. 154, 572 (1965). T. T., and LIN, E. C. C., Science, W. A., Proc. BISSON, T. M.,6 and MORTLOCK, R. P., J. Bacteriol., 9.5..» 925 (196 8). BISSON, T. 14., OLIVER, E. J., and MORTLOCK, R. 2., _J_. Bacteriol., 95, 952 (1968). CUATRECASAS P., and SEGAL, S., J. Biol. Chem., 241 5910 (1966). FOLIN 0., and MALMROSE, H., J. sio___l_. Chem, 85, 115 (1925) ROE, J.H., g. BiO1.Chem., 101, 15 (1954). FISKE, C. H., and SUBBAROW, Y., J. Biol. Chem., 66 575 (1925) UMBREIT, w.w.,BURRIs, 11.11., and STAUFFER, J.F., Manometric Egchnigues, Burgess Publishing Co., Minneapolis, 1957, p. 273. , SIBLEY, J.A., and LEHNINGER, A. L., J. B___io1. C__h__em., .11. 859 (1949) 174 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31.- 175 TRAVELYAN, W.E., PROCTOR, D.P., and HARRISON, J.A., Nature, 166, 444 (1950). BEVENUE, A., and WILLIAMS, K.T., Arch. Biochem. Bioghys., 4, 225 (1951). KOCH, R.B., GEDDES‘, W.F., and SMITH, F., Cereal Chem., 28, 424 (1951). BENSON, A.A.', Methods Enzmol., 5, 116 (1957) . BEAN, R.C., and PORTER, G.G., Anal. Chem., Ll, 1929 (1959) . ‘ TOMBS, M.P., SOUTER, P., and MACLAGAN, N.F., Biochem. J., 12, 167 (1950). WELLS, W.W., KATACI, T., BENTLEY, R., and SWEELEY, C.C., Biochem. Bithxs. Aota.,8_2_, 408 (1964) . SCHDEN, J.C., and FISCHER, H.O.L., J. Amer. Chem. __Soc-. 69, 791 (1946). PERLIN, A.S., and BRICE, C., Egg. J. gem” 23, 85 (1956). PUTMAN, E.W., Methods Enzmol., 1, 67 (1957) . MMEL, M.Y., and ANDERSON, R.L., J. Bacteriol., g, 1689 (1966). VOSBURGH, W.C'., J. Amer. Chem. Soc., $2,, 1696 (1920). GARARD, D.D., and SHERMAN, H.C., J. Amer. Chem. Soc., 45;, 955 (1918). HANN, R.M., and HUDSON, C.s., J. Amer. Chem. Soc., 66. 755 (1944). WOLFROM, M.L., and THOMPSON, A., J. Amer. Chem. _6_8 c-. 66.. 791 (1946)- YOUNG, F.E., JONES, F.T., and LEWIS, H.J., J. .th . __Chem-. L6. 758 (192). 32. 33- 3h. 35- 36. 37- 38. 39- 40. 41. 1+2. 1+3. 44. 45. 45a. 176 JONES, F.T., YOUNG, F.E., and BLACK, D.R., Anal. _Chem-. 25. 649 (1953)- SCHLENK, H., and GELLERMAN, J.L., Anal. Chem., 22, 1412 (1960). LEUTHARDT, F. 1. 320 (19555. MAYO, J.W., and ANDERSON, R.L., Carbohydrate Res., in press. and WOLF, H.P., Methods Enz 01., ENGLESBERG, E., Arch. Biochem. Bio h 3., Z_]_., 179 1957 - MGK, GoFo, and ZECH, R., m- E’) 222, 145 (1963). DCMAGK, G.F., and HEINRICH, R., Biochem an §41, 420 (1965). TAKAGI Y., and SWADA, H., Biochim. Biophys. Acta. 92, 16 (196M). TAKAGI, Y., and SWADA, H., Biochim. Bioghzs. Acta., 22, 18 (1964). TAKAGI, Y., and SWADA, H., Biochdm. Bioghxs. Acta., 92, 26 (1964). HEATH, E.C. and GHAIAMBOR, M.A., _q_. Biol. Chem., 221, 2423 (1962). GHALAMBOR, M.A., and HEATH, E.C., ,1. Biol. Chem., 221, 2427 (1962). FISCHER, E., §g£,, 22, 389 (1890). ASHMORE, J., and RENOLD,.A.E., g,.Amer. Chem. Soc., 16. 6189 (1954)- HANSON, T.E., and ANDERSON R.L., Proc. Natl. Acad. Sci. U.S., in press (19685. 46. 47. 48. 49. 50. 51. 52. 53- 54. 55- 56. 57- 58. 59. 60. 177 HANSON, T.E., and ANDERSON, R.L., g. Biol. Chem., 241, 16u4 (1966). HOLLDORF, A., HOLLDORF, C., SCHNEIDER, S., and HOLZER, H., a; Naturf, 1gp, 229 (1959). DAWKINS, P.D., and DICKENS, P., m. _q_., 95,, 353 (1965). DICKENS, F., and WILLIAMSON, D.H., Biochem g” E, 496 (1959). HADORN, 13“., LEUTHARDT, P., MENARD E., and VISCHER, D., Helv. Chim..Acta., £6, 2003 (1963). BURTON, R.M., Methods Enzmol., 1, 397 (1955). LIN, E.C.C. and MAGASANIK, B., Q, Biol. Chem., 225, 1820 (1960). LIN, E.C.C., LEVIN, A.P. and MAGASANIK, 3., g, Biol. Chem., 2i5, 1825 (1960). KLECZKOWSKI, A., and WIERZCHOWSKI, P., Soil Sci., 12. 193 (1940)- SMITH, N.R., GORDON, R.E., and CLARK,E.E., Aerobic .MeSOEhiliC Spgre Forming Bacteria, U.S. Dept. Agric. Misc. Publications, no. 559. FORSYTH, W.G.C., and WEBLEY, D.M., Biochem in 35, 455 (1949)- FISCHER, E., g;;,, 22, 370 (1890); 23, 2683 (1891). VAN EKERSTEIN,‘W.A. and ELANKSMA, J.J., Chem. Weekblad, 11, 902 (1914); Chem. Zentr., 85, II, 1265 (191477’ AHMED, Z.F., RIDE, A.M. and HAMMOUDA, F.M., g, Pharm. Sci., 53, 1060 (1965). SEED, J.R., BAQUERO, M.A., and DUDA, J.E., ExEtl. Parasitol., _]._§, 363 (1965). 61. 62. 53: 64. 65. 66. 67. 68. 69. 70. 71. 72. 73- 74. 75- 76. 178 SEED, J.R. 427 (1965). DISCHE, z. and BORENFREUND, E., g, Biol. Chem., 192, 583 (1951). and BAQUERO, M.A., g, Protozool., 12, ‘WARBURG, 0., and CHRISTIAN, W., Biochem. §,, 310, 384 (1942). HUDSON, C.s., g,.Amer. Chem. Sgg,, 13, 4498 (1951). FRUSH, H.L., and ISBELL, H.S., U.S. Atomic Energy Commission, NBS-5845 (1954). CHIU, T.H., and FEINGOLD, D.S., Biochim. Bioghxs. Acta., 92, 489 (1964). CHIU, T.H., and FEINGOLD, D.S., Biochem.and Biophys. Res. Comm., 19, 511 (1965). FOLK, J.E., and GIADMER, J:A., Biochim. Biophys. Acta., 48, 139 (1961). LEHMAN, I.R., BESSMAN, N.J., SIMMS, E.S. and KORNBERG, A., g, Biol. Chem., 233, 163 (1958). BALLERONI, N.J. 219; 535 (19555. DIFCO.MANUAL, 9th ed., p. 54 (1953). and DOUDOROFF, H., g, Biol. Chem., DISCHE, z. 595 (19485. FRUSH, H.L., and ISBELL, H.S., Methods in Carbohy— ‘drate Chemistr ,7}; 127 (1962 . SHETTLES, L.B., g, Biol. Chem., 115, MENDEL, B., Klina Wschr., §, 169 (1929). RUDNEY, H., Arch. Biochem. Bioghxs. g3, 67 (1949). WENZEL, H., JOEL, I., OELKERS,.W., Naturwissen- schaften, 53, 82 (1966). 77. 78. 79. 80. 81. 179 NEEDHAM, D.M., smINOVITCH, L., and RAPKINE, S.M., Biochem. 51., 49, 113 (1951). IARDY, H.A.,WIEBELHAUS, V.D., MANN, K.M., g. Biol. Chem., 187, 325 (1950). LEDERBERG, J., Genetics 9; the 20th Centu , ed. by Macmillan, New York, p. 263 (1951).. TORRIANI, A., and ROTHMAN, F., _J_. Bacteriol., §_];, 835 (1961). Km, K., and TCHEN, T.T., Biochem. Bithxs. Res. Comm., 9, 99 (1962). RES 4441)) 444444)”