Skip to main content
Microbiology and Molecular Biology Reviews : MMBR logoLink to Microbiology and Molecular Biology Reviews : MMBR
. 1999 Sep;63(3):675–707. doi: 10.1128/mmbr.63.3.675-707.1999

Helicobacter pylori Physiology Predicted from Genomic Comparison of Two Strains

Peter Doig 1,*, Boudewijn L de Jonge 1, Richard A Alm 1, Eric D Brown 1,, Maria Uria-Nickelsen 1, Brian Noonan 1, Scott D Mills 1, Peter Tummino 1, Gilles Carmel 2, Braydon C Guild 2, Donald T Moir 2, Gerald F Vovis 2,, Trevor J Trust 1
PMCID: PMC103750  PMID: 10477312

Abstract

Helicobacter pylori is a gram-negative bacteria which colonizes the gastric mucosa of humans and is implicated in a wide range of gastroduodenal diseases. This paper reviews the physiology of this bacterium as predicted from the sequenced genomes of two unrelated strains and reconciles these predictions with the literature. In general, the predicted capabilities are in good agreement with reported experimental observations. H. pylori is limited in carbohydrate utilization and will use amino acids, for which it has transporter systems, as sources of carbon. Energy can be generated by fermentation, and the bacterium possesses components necessary for both aerobic and anaerobic respiration. Sulfur metabolism is limited, whereas nitrogen metabolism is extensive. There is active uptake of DNA via transformation and ample restriction-modification activities. The cell contains numerous outer membrane proteins, some of which are porins or involved in iron uptake. Some of these outer membrane proteins and the lipopolysaccharide may be regulated by a slipped-strand repair mechanism which probably results in phase variation and plays a role in colonization. In contrast to a commonly held belief that H. pylori is a very diverse species, few differences were predicted in the physiology of these two unrelated strains, indicating that host and environmental factors probably play a significant role in the outocme of H. pylori-related disease.


Helicobacter pylori is a gram-negative bacterium which colonizes the gastric mucosa of humans, causes gastritis and peptic ulcer disease, and is associated with certain types of gastric cancer (27, 65, 87). Once colonized, the host can be chronically infected for life unless antimicrobial therapy is administered. The ability to colonize and persist in the human stomach for many years indicates that H. pylori is specifically adapted to occupy only this niche, and such adaptation should be reflected in a unique complement of physiological capabilities. Furthermore, physiological differences resulting from the apparent genomic variation among strains have been suggested to be responsible for the diversity of diseases associated with H. pylori infection (16, 95).

Bacterial genomics, the identification and annotation of the entire coding potential of a bacterium, allows a more complete understanding of bacterial physiology and pathogenesis. The recent analysis of the complete genomic sequence of two unrelated, pathogenic H. pylori strains (J99 and 26695) demonstrated that even though the chromosomes are organized differently in a limited number of discrete regions, the genome size, genetic content, and gene order of these two strains are remarkably similar (6). We have used the data resulting from this comparative sequence analysis as the starting point to examine, from a functional perspective, the genes that are common and unique to the two strains. The presence or absence of orthologous genes or metabolic pathways in both unrelated H. pylori strains implies that these genes or pathways are present or absent, respectively, in this species. Our comparison has defined the set of common H. pylori metabolic capabilities as well as a small number that are strain specific.

ANALYSES OF GENETIC AND FUNCTIONAL CONSERVATION

The predicted genes from both H. pylori genomes (6) were assigned a likely function, if their predicted amino acid sequence exhibited similarity to a protein of known function, and categorized as shown in Table 1. Function was annotated conservatively; e.g., proteins which showed sequence similarity to transporters, the majority of which did not have the same substrate specificity, were assigned to the general category of transporters pending experimental evidence for their specificity. The two H. pylori genomes are highly conserved with respect to gene content (1,495 and 1,552 open reading frames [ORFs] in J99 and 26695, respectively [6]), functional categorization (Table 2), and gene order (Fig. 1). In both strains, approximately 58% of the gene products were assigned a putative function based upon their having significant sequence similarity to a protein of known function; nearly 18% were conserved in other species but had no known function; and about 23% were specific to H. pylori (Table 2). Eighty-nine genes were specific to strain J99, and 117 were specific to strain 26695; 26 of these genes in each of the strains J99 and 26695 had an assigned function. Preliminary analysis of the Campylobacter jejuni genome (based on analysis of the recently completed genome by the Sanger Centre) indicated that approximately 90 of the H. pylori specific genes will have an orthologue in this closely related species. This will reduce the proportion of H. pylori-specific genes to approximately 17%.

TABLE 1.

List of H. pylori J99 genes and corresponding 26695 orthologs with putative functional assignmentsa

Gene no. in:
Gene name Function
J99 26695
Amino acid biosynthesis
 General
  206 0220 Aminotransferase
  568 0624 Aminotransferase
  632 0696 Hydantoin utilization
  633 0695 Hydantoin utilization
  673 0736 Aminotransferase
  976 0405 Aminotransferase
 Aromatic amino acid family
  122 0134 aroF Phospho-2-dehydro-3-deoxyheptonate aldolase
  145 0157 aroK Shikamate kinase I
  268 0283 aroB 3-Dehydroquinate synthase
  386 1038 aroD 3-Dehydroquinate dehydratase
  608 0663 aroC Chorismate synthase
  980 0401 aroA 3-Phosphoshikimate 1-carboxyvinyl transferase
  1170 1249 aroE Shikimate 5-dehydrogenase
  1198 1277 trpA Tryptophan synthase alpha chain
  1199 1278 trpB Tryptophan synthase beta chain
  1200 1279 trpC Indole-3-glycerol phosphate synthase
  1201 1280 trpD Anthranilate phosphoribosyltransferase
  1202 1281 trpG Anthranilate synthase component II
  1203 1282 trpE Anthranilate synthase component I
  1294 1380 tyrA Prephenate dehydrogenase
 Aspartate family
  90 0098 thrC Threonine synthase
  98 0106 metB Cystathionine gamma-synthase
  198 0212 dapE Succinyl-diaminopimelate desuccinylase
  275 0290 lysA Diaminopimelate decarboxylase
  375 1050 thrB Homoserine kinase
  410 1013 dapA Dihydrodipicolinate synthase
  460 0510 dapB Dihydrodipicolinate reductase
  513 0566 dapF Diaminopimelate epimerase
  570 0626 dapD 2,3,4,5-Tetrahydropyridine-2-carboxylate-N-succinyltransferase
  594 0649 aspA Aspartate ammonia-lyase
  615 0672 aspB Aspartate aminotransferase
  761 0822 hom Homoserine dehydrogenase
  1114 1189 asd Aspartate-semialdehyde dehydrogenase
  1150 1229 lysC Aspartokinase 2 alpha and beta subunits
 Glutamate family
  461 0512 glnA Glutamine synthetase
  1001 0380 gdhA Glutamate dehydrogenase
  1085 1158 proC Pyrroline-5-carboxylate reductase
 Pyruvate family
  313 0330 ilvC Ketol-acid reductoisomerase
  1361 1468 ilvE Branched-chain amino acid aminotransferase
 Serine family
  99 0107 cysK Cysteine synthase
  171 0183 glyA Serine hydroxymethyltransferase
  597 0652 serB Phosphoserine phosphatase
  984 0397 serA Phosphoglycerate dehydrogenase
  1133 1210 cysE Serine acetyltransferase
Biosynthesis of cofactors, prosthetic groups, and carriers
 Biotin
  25 0029 bioD Dethiobiotin synthetase
  545 0598 bioF 8-Amino-7-oxononanoate synthase
  910 0976 bioA Adenosylmethionine-8-amino-7-oxononanoate aminotransferase
  1068 1140 Biotin activation protein
  1298 1406 bioB Biotin synthetase
 Folic acid
  278 0293 pabB p-Aminobenzoate synthetase
  388 1036 folK 7,8-Dihydro-6-hydroxymethylpterin-pyrophosphokinase
  524 0577 folD Methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase
  862 0928 folE GTP cyclohydrolase I
  863 0928 folE GTP cyclohydrolase I
  1153 1232 folP Dihydropteroate synthase
  1403 1510 folB Dihydroneopterin aldolase
  1454 1545 folC Folylpolyglutamate synthase
 Heme and porphyrin
  150 0163 hemB δ-Aminolevulinic acid dehydratase
  222 0237 hemC Porphobilinogen deaminase
  224 0239 hemA Glutamyl-tRNA reductase
  291 0306 hemL Glutamate-1-semialdehyde-2,1-aminomutase
  551 0604 hemE Uroporphyrinogen decarboxylase
  610 0665 hemN Oxygen-independent coproporphyrinogen III oxidase
  1000 0381 hemG Protoporphyrinogen oxidase
  1005 0376 hemH Ferrochelatase
  1145 1224 hemD Uroporphyrinogen III synthase
  1147 1226 hemN Oxygen-independent coproporphyrinogen III oxidase
 Menaquinone and ubiquinone
  225 0240 ispB Octaprenyl-diphosphate synthase
  864 0929 ispA Geranyltransferase
  1278 1360 ubiA 4-Hydroxybenzoate octaprenyltransferase
  1369 1476 ubiD Octaprenyl-4-hydroxybenzoate carboxy-lyase
  1376 1483 ubiE Ubiquinone/menaquinone biosynthesis methyltransferase
 Molybdopterin
  158 0172 moeA Molybdopterin biosynthesis protein
  705 0768 moaA Molybdopterin cofactor biosynthetic protein
  706 0769 mobA Molybdopterin-guanine dinucleotide biosynthesis protein A
  734 0798 moaC Molybdenum cofactor biosynthesis protein C
  735 0799 mog Molybdopterin biosynthesis protein
  736 0800 moaE Molybdopterin-converting factor, subunit 2
  737 0801 moaD Molybdopterin-converting factor, subunit 1
  750 0814 moeB Molybdopterin-synthase sulfurylase
 Pantothenate
  6 0006 panC Pantoate-β-alanine ligase
  30 0034 panD Aspartate-1-decarboxylase
  367 1058 panB 3-Methyl-2-oxobutanoate hydroxymethyltransferase
  779 0841 dfp Pantothenate metabolism flavoprotein
 Pyridoxine
  328 0354 dxs 1-Deoxyxylulose-5-phosphate synthase
  1489 1582 pdxJ Pyridoxal phosphate synthetase
  1490 1583 pdxA Pyridoxal phosphate biosynthetic protein A
 Riboflavin
  2 0002 ribE Riboflavin synthase beta chain
  338 1087 ribF Riboflavin kinase
  738 0802 ribA GTP cyclohydrolase II
  740 0804 ribBA GTP cyclohydrolase II/3,4-dihydroxy-2-butanone 4-phosphate synthase
  1398 1505 ribD Riboflavin-specific deaminase
  1482 1574 ribC Riboflavin synthase alpha chain
 Thioredoxin, glutaredoxin, and glutathione
  763 0824 trxA Thioredoxin
  764 0825 trxB Thioredoxin reductase
  1046 1118 ggt Gamma-glutamyl transpeptidase
  1091 1164 trxB Thioredoxin reductase
  1351 1458 Thioredoxin
 Thiamine
  781 0843 thiE Thiamine phosphate pyrophosphorylase
  782 0844 thiD Phosphomethylpyrimidine kinase
  783 0845 thiM Hydroxyethylthiazole kinase
 Pyridine nucleotides
  312 0329 nadE NH3-dependent NAD+ synthetase
  1273 1355 nadC Nicotinate-nucleotide pyrophosphorylase
  1274 1356 nadA Quinolinate synthetase
Cell envelope
 Membranes and porins
  7 0009 Outer membrane protein
  21 0025 Outer membrane protein
  73 0078 Outer membrane protein
0079
  117 0127 Outer membrane protein
  195 0209 Outer membrane protein
  212 0227 Outer membrane protein
  214 0229 hopA Outer membrane protein—porin
  237 0252 Outer membrane protein
  238 0253, 0254 Outer membrane protein
  307 0324 Outer membrane protein
  342 1083 Outer membrane protein
  359 1066 Outer membrane protein
  424 0472 Outer membrane protein
  429 0477 Outer membrane protein
  438 0486 Outer membrane protein
  439 0487 Outer membrane protein
  456 0506 Outer membrane protein
  514 0567 Inner membrane protein
  581 0638 Outer membrane protein
  600 0655 Outer membrane protein
  614 0671 Outer membrane protein
  634 0694 Outer membrane protein
  645 0706 hopE Outer membrane protein—porin
  649 0710 Outer membrane protein
  659 0722 Outer membrane protein
  662 0725 Outer membrane protein
  663 0726 Outer membrane protein
  719 0782 Outer membrane protein
  725 0788 Outer membrane protein
  732 0796 Outer membrane protein
  777 0839 Outer membrane protein
  810 0876 frpB Iron-regulated outer membrane protein
  833 1243 babB Outer membrane protein—adhesin
  848 0912 hopC Outer membrane protein—porin
  849 0913 hopB Outer membrane protein—porin
  850 0914 Outer membrane protein
  851 0915, 0916 frpB Iron-regulated outer membrane protein
  857 0923 Outer membrane protein
  870 Outer membrane protein
  1008 0373 Outer membrane protein
  1022 0358 Outer membrane protein
  1034 1107 Outer membrane protein
  1040 1113 Outer membrane protein
  1054 1125 Outer membrane protein
  1083 1156 Outer membrane protein
  1084 1157 Outer membrane protein
  1094 1167 Outer membrane protein
  1103 1177 Outer membrane function
  1138 1215, 1216 imp Role in outer membrane permeability
  1164 0896 babA Outer membrane protein—adhesin
  1261 1342 Outer membrane protein
  1343 1450 Inner membrane protein
  1346 1453 Outer membrane protein
  1349 1456 lpp20 Conserved lipoprotein
  1360 1467 Outer membrane protein
  1362 1469 Outer membrane protein
  1394 1501 Outer membrane protein
  1405 1512 frpB Iron-regulated outer membrane protein
  1432 1395 Outer membrane protein
  1472 1564 Outer membrane protein
  1479 1571 Outer membrane protein
 Murein sacculus and peptidoglycan
  445 0493 mraY Phospho-N-acetylmuramoyl-pentapeptide-transferase
  446 0494 murD UDP-N-acetylmuramoylalanine-d-glutamate ligase
  496 0549 murI Glutamate racemase
  544 0597 Penicillin-binding protein
  567 0623 murC UDP-N-acetylmuramate-alanine ligase
  590 0645 Lytic murein transglycosylase
  593 0648 murA UDP-N-acetylglucosamine enolpyruvyltransferase
  675 0738 ddl d-Alanine–d-alanine ligase
  677 0740 murF d-Alanyl–d-alanine-adding enzyme
  709 0772 amiA Probable N-acetylmuramoyl-l-alanine amidase
  876 0941 alr Alanine racemase, biosynthetic
  1082 1155 murG UDP-N-acetylglucosamine-N-acetylmuramyl-(pentapeptide) pyrophosphoryl-undecaprenoln-acetylglucosaminetransferase
  1313 1418 murB UDP-N-acetylenolpyruvoyl glucosamine reductase
  1387 1494 murE UDP-N-acetylmuramyl-tripeptide synthetase
  1464 1556 Penicillin-binding protein
  1473 1565 Penicillin-binding protein
 Surface polysaccharides, lipopolysaccharides, and antigens
  3 0003 kdsA 3-Deoxy-d-manno-octulosonic acid 8-phosphate synthase
  37 0043 manA/manC Phosphomannose isomerase/GDP-mannose pyrophosphorylase
  38 0044 Gmd GDP-d-mannose dehydratase
  39 0045 Sugar nucleotide biosynthesis
  70 0075 glmM Phosphoglucosamine mutase
  86 0093, 0094 α-(1,2)-Fucosyltransferase
  147 0159 Lipopolysaccharide biosynthesis protein
  166 0178 neuB Sialic acid synthase
  182 0196 lpxD UDP-3-O-[3-hydroxymyristoyl] glucosamine N-acetyltransferase
  194 0208 Lipopolysaccharide biosynthesis protein
  215 0230 kdsB 3-Deoxy-manno-octulosonate cytidylyltransferase
  264 0279 waaC Lipopolysaccharide heptosyltransferase-1
  265 0280 waaM Lipid A biosynthesis acyltransferase
  309 0326 neuA Acylneuraminate cytidylyltransferase
  311 0328 lpxK Tetraacyldisaccharide-1-p 4′-kinase
  373 1052 lpxC UDP-3-O-[3-hydroxymyristoyl]-N-acetylglucosamine deacetylase
  562 Lipopolysaccharide biosynthesis protein
  563 0619 Lipopolysaccharide biosynthesis protein
  596 0651 fucT α-(1,3)-Fucosyltransferase
  620 0679 Lipopolysaccharide biosynthesis protein
  741 0805 Lipopolysaccharide biosynthesis protein
  765 0826 Lipopolysaccharide biosynthesis protein
  778 0840 Sugar nucleotide biosynthesis protein
  791 0857 gmhA Phosphoheptose isomerase
  792 0858 waaE ADP-d-glycero-d-mannoheptose synthase
  793 0859 gmhD ADP-l-glycero-d-mannoheptose-6-epimerase
  801 0867 lpxB Lipid-A-disaccharide synthase
  820 Lipopolysaccharide biosynthesis protein
  891 0957 waaA 3-Deoxy-d-manno-octulosonic-acid transferase
  963 0421 Polysaccharide biosynthesis protein
  1002 0379 fucU α-(1,3)-Fucosyltransferase
  1015 0366 Sugar nucleotide biosynthesis
  1020 0360 galE UDP-glucose 4-epimerase
  1031 1105 Lipopolysaccharide biosynthesis protein
  1032 Lipopolysaccharide biosynthesis protein
  1116 1191 waaF ADP-heptose-lipopolysaccharide heptosyltransferase II
  1196 1275 manB Phosphomannomutase
  1289 1375 lpxA UDP-N-acetylglucosamine acyltransferase
  1311 1416 Lipopolysaccharide biosynthesis protein
  1368 1475 kdtB Lipopolysaccharide core biosynthesis protein
  1488 1581 wecA Undecaprenyl-phosphate-α-N-acetylglucosaminyltransferase
 Surface structures
  107 0115 flaB Flagellin B
  159 0173 fliR Flagellar biosynthesis protein
  217 0232 Motility protein
  231 0246 flgI Flagellar P-ring protein
  280 0295 flgL Flagellar hook-associated protein 3 (hap3)
  308 0325 flgH Flagellar L-ring protein precursor (basal body L-ring protein)
  310 0327 flaG Flagellar biosynthesis protein
  325 0351 fliF Flagellar M-ring protein
  326 0352 fliG Flagellar motor switch protein
  327 0353 fliH Flagellar export apparatus
  333 1092 flgG Flagellar basal-body rod protein
  383 1041 flhA Flagellar biosynthesis protein
  389 1035 flhF Flagellar biosynthesis protein
  393 1031 fliM Flagellar motor switch protein
  394 1030 Flagellar motor switch protein
  444 0492 Paralogue of hpaA
  531 0584 fliN Flagellar motor switch protein
  548 0601 flaA Flagellin A
  625 0684, 0685 fliP Flagellar biosynthesis protein
  688 0751 Flagellin protein
  689 0752 fliD Flagellar hook-associated protein 2 (hap2)
  690 0753 fliS Flagellar protein
  707 0770 flhB Flagellar biosynthesis protein
  733 0797 hpaA Neuraminyllactose-binding hemagglutinin precursor
  745 0809 fliL Flagellar biosynthesis protein
  751 0815 motA Chemotaxis protein (motility protein a)
  752 0816 motB Flagellar motor protein
  804 0870 flgE Flagellar hook protein
  843 0907 Flagellar biosynthesis protein
  844 0908 Flagellar basal-body/rod/hook protein
  971 0410 Paralogue of hpaA
  1047 1119 flgK Flagellar hook-associated protein 1 (Hap1)
  1117 1192 Motility protein
  1195 1274 pflA Flagellar functional protein
  1314 1419 fliQ Flagellar biosynthesis protein
  1315 1420 fliI Flagellum-specific ATP synthase
  1355 1462 Motility protein
  1465 1557 fliE Flagellar hook–basal-body complex protein
  1466 1558 flgC Flagellar basal-body rod protein
  1467 1559 flgB Flagellar basal-body rod protein
  1483 1575 flhB Flagellar biosynthesis protein
  1492 1585 flgG Flagellar basal-body rod protein (distal rod protein)
Cellular processes
 General
  4 0004 icfA Carbonic anhydrase
  161 0175 Peptidyl-prolyl cis-trans isomerase
  183 0197 metK S-Adenosylmethionine synthetase
  228 0243 napA Neutrophil-activating protein A
  466 0517 era GTP-binding protein
  678 0741 HIT family protein
  865 0930 surE Stationary-phase protein
  977 0404 HIT family protein
  1112 1186 Carbonic anhydrase
 Cell division
  314 0331 minD Cell division inhibitor
  315 0332 minE Cell division topological specificity factor
  335 1090 ftsK Septum formation protein
  680 0743 rodA Rod shape-determining protein
  912 0978 ftsA Septum formation protein
  913 0979 ftsZ GTPase in circumferential ring formation
  1086 1159 fic cAMP-induced cell filamentation protein
  1287 1373 mreB Rod shape-determining protein
  1468 1560 rodA Rod shape-determining protein
 Cell killing
  274 0289 Vacuolating cytotoxin (VacA) paralogue
  339 1086 hlyA Hemolysin
  556 0609, 0610 Vacuolating cytotoxin (VacA) paralogue
  819 0887 vacA Vacuolating cytotoxin
  856 0922 Vacuolating cytotoxin (VacA) paralog
 Cag island proteins and transposable elements
  15 0017 virB4a DNA transfer protein
  36 0041, 0042 virB10 DNA transfer protein
  469 0520 orf6 Cag island protein
  470 0521 orf7 Cag island protein
  471 0522 orf8 Cag island protein
  472 0523 orf9 Cag island protein
  473 0524 virD4 Cag island protein, DNA transfer protein
  474 0525 virB11a Cag island protein, DNA transfer protein
  475 0526 orf12 Cag island protein
  476 0527 orf13/14 Cag island protein
  477 0528 orf15 Cag island protein
  478 0529 orf16 Cag island protein
  479 0530 orf17 Cag island protein
  480 0531 orf18 Cag island protein
  481 0532 cagT Cag island protein
  482 0534 cagS Cag island protein
  483 0535 cagQ Cag island protein
  484 0536 cagP Cag island protein
  485 0537 cagM Cag island protein
  486 0538 cagN Cag island protein
  487 0539 cagL Cag island protein
  488 0540 cagI Cag island protein
  489 0541 cagH Cag island protein
  490 0542 cagG Cag island protein
  491 0543 cagF Cag island protein
  492 0544 cagE DNA transfer protein (Agrobacterium VirB4 homologue)
  493 0545 cagD Cag island protein
  494 0546 cagC Cag island protein
  495 0547 cagA Cag island protein, cytotoxicity-associated immunodominant antigen
  826 tnpB IS606 transposase
  827 tnpA IS606 transposase
  917 virB4b DNA transfer protein
  918 virB4c DNA transfer protein
  1279 1361 comEC DNA transfer protein
  1316 1421 virB11b DNA transfer protein
 Chaperones
  8 0010 groEL 60-kDa chaperone
  9 0011 groES 10-kDa chaperone
  101 0109 dnaK 70-kDa chaperone
  102 0110 grpE 24-kDa chaperone
  196 0210 htpG 90-kDa chaperone
  400 1024 dnaJ2 Cochaperone with DnaK
  861 0927 htpX Stress protein
  1252 1332 dnaJ1 Cochaperone with DnaK
 Detoxification
  809 0875 katA Catalase
  991 0390 tpx Thiol peroxidase
  992 0389 sodF Iron-dependent superoxide dismutase
  1471 1563 tsaA Peroxidase
 Protein and peptide secretion
  69 0074 lspA Lipoprotein signal peptidase
  168 0180 lnt Apolipoprotein N-acyltransferase
  329 0355 lepA GTP-binding protein
  523 0576 lepB Signal peptidase I
  700 0763 ftsY Functional homolog of srp receptor
  723 0786 secA Preprotein translocase subunit
  731 0795 tig Trigger factor
  889 0955 lgt Prolipoprotein diacylglyceryl transferase
  1079 1152 ffh Signal recognition particle protein
  1126A 1203A secE Preprotein translocase subunit
  1176 1255 secG Protein export membrane protein
  1220 1300 secY Preprotein translocase subunit
  1449 1550 secD Protein export membrane protein
  1450 1549 secF Protein export membrane protein
 Phosphorus compounds
  413 1010 ppk Polyphosphate kinase
  564 0620 ppa Inorganic pyrophosphatase
 Polyamine biosynthesis
  18 0020 nspC Carboxynorspermidine decarboxylase
  771 0832 speE Spermidine synthase
  962 0422 speA Arginine decarboxylase
 Urea
  62 0067 ureH Urease accessory protein
  63 0068 ureG Urease accessory protein
  64 0069 ureF Urease accessory protein
  65 0070 ureE Urease accessory protein
  67 0072 ureB Urease beta subunit
  68 0073 ureA Urease alpha subunit
DNA replication
  10 0012 dnaG DNA primase
  108 0116 topA DNA topoisomerase I
  199 0213 gidA Glucose-inhibited division protein A
  362 1063 gidB Glucose-inhibited division protein B
  452 0500 dnaN DNA polymerase III, beta chain
  453 0501 gyrB DNA gyrase subunit B
  558 0615 lig DNA ligase
  641 0701 gyrA DNA gyrase subunit A
  655 0717 dnaX DNA polymerase III subunits gamma and tau
  847 0911 ATP-dependent helicase
  919 topA Topoisomerase I
  931 topA Topoisomerase I
  994 0387 priA Primosomal protein n′ (replication factor y)
  1152 1231 holB DNA polymerase III subunit delta′
  1166 1245 ssb Single-strand binding protein
  1280 1362 dnaB Replicative DNA helicase
  1353 1460 dnaE DNA polymerase III, alpha chain
  1363 1470 polA DNA polymerase I
  1371 1478 rep ATP-dependent DNA helicase
  1412 1523 recG ATP-dependent DNA helicase
  1417 1529 dnaA Chromosomal replication initiator protein
  1438 1387 DNA polymerase III
  1446 1553 pcrA ATP-dependent helicase
DNA restriction, modification, recombination, and repair
  43 0050 Type II DNA modification enzyme (methyltransferase)
  44 Type II DNA modification enzyme (methyltransferase)
  45 Type II DNA modification enzyme (methyltransferase)
  46 Type II restriction enzyme
  84 0091 Type II restriction enzyme
  85 0092 Type II DNA modification enzyme (methyltransferase)
  130 0142 mutY A/G-specific adenine glycosylase
  141 0153 recA Recombination protein
  164 Restriction enzyme
  209 0223 radA DNA repair protein
  243 0259 xseA Exodeoxyribonuclease large subunit
  244 0260 Type II DNA modification enzyme (methyltransferase)
  248 0263 Type II DNA modification enzyme (methyltransferase)
  306 0323 Endonuclease
  322 0348 recJ Single-stranded-DNA-specific exonuclease
  366 1059 ruvB Holliday junction DNA helicase
  414 hsdS1 Type I restriction enzyme (specificity subunit)
  415 0463 hsdM1 Type I restriction enzyme (modification subunit)
  416 0464 hsdR1 Type I restriction enzyme (restriction subunit)
  430 0478 Type II DNA modification enzyme (methyltransferase)
  433 0481 Type II DNA modification enzyme (methyltransferase)
  435 0483 Type II DNA modification enzyme (methyltransferase)
  532 0585 nth Endonuclease III
  549 0602 Endonuclease III
  565 0621 mutS DNA mismatch repair protein
  606 0661 rnhA RNase HI
  617 0675 Integrase-recombinase protein (xerCD family)
  618 0676 ogt Methylated-DNA–protein-cysteine methyltransferase
  629 Type II DNA modification enzyme (methyltransferase)
  630 Type II restriction enzyme
  644 0705 uvrA Excinuclease ABC subunit A
  726 hsdS Type I restriction enzyme (specificity subunit)
  756 Type II DNA modification enzyme (methyltransferase)
  760 0821 uvrC Excinuclease ABC subunit C
  784 0846 hsdR2 Type I restriction enzyme (restriction subunit)
  785 0848, 0849 hsdS2 Type I restriction enzyme (specificity subunit)
  786 0850 hsdM2 Type I restriction enzyme (modification subunit)
  811 0877 ruvC Crossover junction endodeoxyribonuclease
  815 0883 ruvA Holliday junction DNA helicase
  846 0910 Type II DNA modification enzyme (methyltransferase)
  859 0925 recR Recombination protein
  941 0995 Integrase/recombinase (xerCD family)
  951 Integrase/recombinase (xerCD family)
  1012 0369 Type II DNA modification enzyme (methyltransferase)
  1041 1114 uvrB Excinuclease ABC subunit B
  1050 1121 Type II DNA modification enzyme (methyltransferase)
  1131 1208 M.HpyI Type II DNA modification enzyme (methyltransferase)
  1149 1228 mutT dGTP pyrophosphohydrolase
  1243 1323 rnhB RNase HII
  1266 1347 ung Uracil-DNA glycosylase
  1271 1352 Type II DNA modification enzyme (methyltransferase)
  1284 Type II DNA modification enzyme (methyltransferase)
  1295 1382 Endonuclease
  1296 mod1 Type III DNA modification enzyme (methyltransferase)
  1297 res1 Type III restriction enzyme
  1364 1471 Type II restriction enzyme
  1365 1472 Type II DNA modification enzyme (methyltransferase)
  1409 Type II DNA modification enzyme (methyltransferase)
  1410 1521 res2 Type III restriction enzyme
  1411 1522 mod2 Type III DNA modification enzyme (methyltransferase)
  1415 1526 exoA Exodeoxyribonuclease
  1422 hsdS3 Type I restriction enzyme (specificity subunit)
  1423 1403 hsdM3 Type I restriction enzyme (modification subunit)
  1424 1402 hsdR3 Type I restriction enzyme (restriction subunit)
  1434 1393 recN DNA repair protein
  1442 1366 Type II restriction enzyme
Energy metabolism
 Amino acids and amines
  120 0132 sdaB l-Serine/l-Threonine deaminase
  279 0294 aimE Aliphatic amidase
  585 3-Hydroxyacid dehydrogenase
  661 0723 ansB l-Asparaginase II
  1159 1238 Aliphatic amidase
  1427 1399 rocF Arginase
  1428 1398 ald l-Alanine dehydrogenase
 ATP-proton motive force interconversion
  767 0828 atpB ATP synthase F0, subunit a
  1059 1131 atpC ATP synthase F1, subunit epsilon
  1060 1132 atpD ATP synthase F1, subunit beta
  1061 1133 atpG ATP synthase F1, subunit gamma
  1062 1134 atpA ATP synthase F1, subunit alpha
  1063 1135 atpH ATP synthase F1, subunit delta
  1064 1136 atpF ATP synthase F0, subunit b
  1065 1137 atpX ATP synthase B′
  1135 1212 atpE ATP synthase F0, subunit c
 Electron transport
  40 0047 hypE Hydrogenase expression/formation protein
  48 0056 putA Proline/pyrroline-5-carboxylate dehydrogenase
  132 0144 fixN Cytochrome oxidase (CBB3-TYPE)
  133 0145 fixO Cytochrome oxidase (CBB3-TYPE)
  134 0146 fixQ Cytochrome oxidase (CBB3-TYPE)
  135 0147 fixP Cytochrome oxidase (CBB3-TYPE)
  177 0191 frdB Fumarate reductase
  178 0192 frdA Fumarate reductase
  179 0193 frdC Fumarate reductase
  250 0265 ccdA Cytochrome c biogenesis protein
  262 0277 Ferredoxin
  459 0509 glcD Glycolate oxidase
  574 0631 hyaA Hydrogenase, small subunit
  575 0632 hyaB Hydrogenase, large subunit
  576 0633 hyaC Hydrogenase, cytochrome subunit
  577 0634 hyaD Hydrogenase expression/formation protein
  611 0666 glpC Glycerol-3-phosphate dehydrogenase
  803 0869 hypA Hydrogenase expression/formation protein
  835 0898 hypD Hydrogenase expression/formation protein
  836 0899 hypC Hydrogenase expression/formation protein
  837 0900 hypB Hydrogenase expression/formation protein
  878 0943 dadA d-Amino acid dehydrogenase
  895 0961 gpsA Glycerol-3-phosphate dehydrogenase (NAD+)
  974 0407 S/N-oxide reductase
  1003 0378 Cytochrome c biogenesis protein
  1035 1108 porG Pyruvate ferrodoxin oxidoreductase
  1036 1109 porD Pyruvate ferrodoxin oxidoreductase
  1037 1110 porA Pyruvate ferrodoxin oxidoreductase
  1038 1111 porB Pyruvate ferrodoxin oxidoreductase
  1088 1161 fldA Flavodoxin
  1090 1163 fixS Component of cation transport for cbb3-type oxidase
  1143 1222 dld d-Lactate dehydrogenase
  1148 1227 Periplasmic cytochrome c-553
  1181 1260 nuoA NADH oxidoreductase I
  1182 1261 nuoB NADH oxidoreductase I
  1183 1262 nuoC NADH oxidoreductase I
  1184 1263 nuoD NADH oxidoreductase I
  1185 1264 nuoE NADH oxidoreductase I
  1186 1265 nuoF NADH oxidoreductase I
  1187 1266 nuoG NADH oxidoreductase I
  1188 1267 nuoH NADH oxidoreductase I
  1189 1268 nuoI NADH oxidoreductase I
  1190 1269 nuoJ NADH oxidoreductase I
  1191 1270 nuoK NADH oxidoreductase I
  1192 1271 nuoL NADH oxidoreductase I
  1193 1272 nuoM NADH oxidoreductase I
  1194 1273 nuoN NADH oxidoreductase I
  1354 1461 Cytochrome c peroxidase
  1401 1508 fixG Component of cation transport for cbb3-type oxidase
  1459 1540 petA Ubiquinol cytochrome c oxidoreductase, 2Fe-2S subunit
  1460 1539 petB Ubiquinol cytochrome c oxidoreductase, cytochrome b subunit
  1461 1538 petC Ubiquinol cytochrome c oxidoreductase, cytochrome c1 subunit
 Entner-Doudoroff pathway
  1025 1099 eda 2-Keto-3-deoxy-6-phosphogluconate aldolase
  1026 1100 edd Phosphogluconate dehydratase
 Fermentation
  840 0903 ackA Acetate kinase
  841 0904, 0905 pta Phosphotransacetylase
  1030 1104 Zinc-dependent alcohol dehydrogenase
  1429 Zinc-dependent alcohol dehydrogenase
 Gluconeogenesis
  111 0121 ppsA Phosphoenolpyruvate synthase
  142 0154 eno Enolase
  162 0176 fba Fructose-bisphosphate aldolase
  180 0194 tpi Triose-phosphate isomerase
  855 0921 gap Glyceraldehyde-3-phosphate dehydrogenase
  908 0974 pgm 2,3-Bisphosphoglycerate-independent phosphoglycerate mutase
  1093 1166 pgi Glucose-6-phosphate isomerase
  1264 1345 pgk Phosphoglycerate kinase
  1265 1346 gap Glyceraldehyde 3-phosphate dehydrogenase
  1440 1385 fbp Fructose-1,6-bisphosphatase
 Phosphopentose pathway
  337 1088 tktA Transketolase
  521 0574 rpi Ribose 5-phosphate isomerase
  1027 1101 zwf Glucose-6-phosphate-1-dehydrogenase
  1388 1495 tal Transaldolase
  1439 1386 rpe Ribulose-phosphate-3-epimerase
 Sugars
  1029 1103 glk Glucokinase
 Tricarboxylic acid cycle
  22 0026 gltA Citrate synthase
  23 0027 icd Isocitrate dehydrogenase
  536 0588 oorD Subunit of 2-oxoglutarate oxidoreductase
  537 0589 oorA Subunit of 2-oxoglutarate oxidoreductase
  538 0590 oorB Subunit of 2-oxoglutarate oxidoreductase
  539 0591 oorC Subunit of 2-oxoglutarate oxidoreductase
  716 0779 acnB Aconitate hydratase
  1245 1325 fumC Fumarase
 Other
  88 0096 Keto-acid dehydrogenase
  586 0642 Oxidoreductase
  888 0954 Aldehyde dehydrogenase
  1023 0357 Oxidoreductase
  1028 1102 Dehydrogenase
  1345 1452 thdF Thiophene/furan oxidation protein
Fatty acid and phospholipid metabolism
  83 0090 fabD Malonyl-CoA-ACP transacylase
  176 0190 Cardiolipin synthase
  181 0195 fabI Enoyl-ACP reductase
  187 0201 plsX Fatty acid/phospholipid synthesis protein
  188 0202 fabH β-Ketoacyl-ACP synthase III
  201 0215 cdsA CDP-diacylglycerol synthase
  354 1071 pssA Phosphatidylserine synthase
  407 1016 pgsA Phosphatidylglycerophosphate synthase
  409 1014 Short-chain dehydrogenase
  451 0499 pldA Phospholipase A1
  504 0557 accA Acetyl-CoAa carboxylase subunit A
  505 0558 fabB β-Ketoacyl-ACP synthase I
  506 0559 acpP ACP
  508 0561 fabG Acetyl-CoA carboxylase subunit A
  636 0692 scoB 3-Oxoacid CoA-transferase, subunit B
  637 0691 scoA 3-Oxoacid CoA-transferase, subunit A
  638 0690 thl Acetyl-CoA acetyltransferase
  640 0700 dgkA Diacylglycerol kinase
  674 0737 pgpA Phosphatidylglycerophosphatase A
  744 0808 acpS Holo-ACP synthase
  805 0871 cdh CDP-diacylglycerol pyrophosphatase
  884 0950 accD Acetyl-CoA carboxylase subunit B
  968 0416 cfa Cyclopocyclopropane fatty acid synthase
  1010 0371 accB Biotin carboxyl carrier protein
  1011 0370 accC Biotin carboxylase
  1267 1348 plsC 1-Acyl-SN-glycerol-3-phosphate acyltransferase
  1275 1357 psd Phosphatidylserine decarboxylase
  1290 1376 fabZ Hydroxymyristoyl-ACP dehydratase
Purines, pyrimidines, nucleosides and nucleotides
 2′-Deoxyribonucleotide metabolism
  621 0680 nrdA Ribonucleoside-diphosphate reductase 1 alpha chain
  799 0865 dut Deoxyuridine 5′-triphosphate nucleotidohydrolase
  1009 0372 dcd Deoxycytidine triphosphate deaminase
  1016 0364 nrdB Ribonucleoside-diphosphate reductase 1 beta chain
 Purine ribonucleotide biosynthesis
  1039 1112 purB Adenylosuccinate lyase
  1140 1218 purD Glycinamide ribonucleotide synthetase
  1327 1434 purU Formyltetrahydrofolate hydrolase
 Pyrimidine ribonucleotide biosynthesis
  5 0005 pyrF Orotidine 5′-phosphate decarboxylase
  184 0198 ndk Nucleoside diphosphate kinase
  251 0266 pyrC1 Dihydroorotase
  323 0349 pyrG CTP synthase
  341 1084 pyrB Aspartate carbamoyltransferase catalytic chain
  412 1011 pyrD Dihydroorotate dehydrogenase
  528 0581 pyrC2 Dihydroorotase
  714 0777 pyrH Uridylate kinase
  853 0919 pyrA1 Carbamoyl-phosphate synthase large chain
  1158 1237 pyrA2 Carbamoyl-phosphate synthase small chain
  1178 1257 pyrE Orotate phosphoribosyltransferase
 Salvage and interconversion of nucleosides and nucleotides
  96 0104 cpdB 2′,3′-Cyclic nucleotide 2′-phosphodiesterase
  239 0255 purA Adenylosuccinate synthetase
  304 0321 gmk Guanylate kinase
  519 0572 apt Adenine phosphoribosyltransferase
  561 0618 adk Adenylate kinase
  672 0735 gpt Xanthine-guanine phosphoribosyltransferase
  679 0742 prsA Phosphoribosyl pyrophosphate synthetase
  768 0829 guaB Inosine-5′-monophosphate dehydrogenase
  790 0854 guaC GMP reductase
  972 0409 guaA GMP synthetase
  1104 1178 deoD Purine nucleoside phosphorylase
  1105 1179 deoB Phosphopentomutase
  1367 1474 tmk Thymidylate kinase
 Sugar-nucleotide biosynthesis and conversions
  591 0646 galU UTP-glucose-1-phosphate uridylyltransferase
  624 0683 glmU UDP-N-acetylglucosamine pyrophosphorylase
  1420 1532 glmS Glutamine fructose-6-phosphate minotransferase
Regulatory functions
 General
  41 0048 Transcriptional regulator
  81 0088 rpoD RNA polymerase sigma 70 factor
  151 0164, 0165 Histidine kinase sensor protein
  152 0166 Transcriptional regulator
  229 0244 Histidine kinase sensor protein
  263 0278 gppA Guanosine-5′-triphosphate,3′-diphosphate pyrophosphatase
  381 1043 Transcriptional regulator
  392 1032 fliA RNA polymerase sigma 28 factor
  397 1027 fur Ferric uptake regulation protein
  399 1025 Transcriptional regulator
  403 1021 Transcriptional regulator
  643 0703 Transcriptional regulator
  652 0714 rpoN RNA polymerase sigma-54 factor
  664 0727 Transcriptional regulator
  712 0775 spoT Guanosine-3′,5′-bis(diphosphate)-3′-pyrophosphohydrolase
  981 0400 lytB Lysis tolerance protein
  1207 1287 Transcriptional regulator
  1282 1364 Histidine kinase sensor protein
  1283 1365 Transcriptional regulator
  1335 1442 csrA Carbon storage regulator
  1443 1365 Transcriptional regulator
  1480 1572 dniR Regulatory protein
 Chemotaxis and motility
  17 0019 cheV1 Chemotaxis protein
  75 0082 MCP
  91 0099 MCP
  95 0103 MCP
  358 1067 cheY Response regulator
  546 0599 MCP
  559 0616 cheV2 Chemotaxis protein
  988 0393 cheV3 Chemotaxis protein
  989 0392 cheA Histidine kinase
  990 0391 cheW Histidine kinase-MCP coupling protein
Transcription
 Degradation of RNA
  1136 1213 pnp Polyribonucleotide nucleotidyltransferase
  1169 1248 vacB RNase II family protein
  1299 1407 rbn RNase N
 DNA-dependent RNA polymerase
  1121 1198 rpoB DNA-directed RNA polymerase, beta subunit
  1213 1293 rpoA DNA-directed RNA polymerase, alpha subunit
  1458 1541 mfd Transcription-repair coupling factor
 Transcription factors
  1 0001 nusB Transcription termination
  497 0550 rho Transcription termination factor
  800 0866 greA Transcription elongation factor (transcript cleavage factor)
  1126 1203 nusG Transcription antitermination protein
  1407 1514 nusA N utilization substance protein A
 RNA processing
  583 0640 pcnB Polynucleotide adenylyltransferase
  607 0662 rnc RNase III
Translation
 Aminoacyl-tRNA synthetases
  113 0123 thrS Threonyl-tRNA synthetase
  170 0182 lysS Lysyl-tRNA synthetase
  223 0238 proS Prolyl-tRNA synthetase
  302 0319 argS Arginyl-tRNA synthetase
  428 0476 gltX Glutamyl-tRNA synthetase
  560 0617 aspS Aspartyl-tRNA synthetase
  588 0643 gltX Glutamyl-tRNA synthetase
  711 0774 tyrS Tyrosyl-tRNA synthetase
  818 0886 cysS Cysteinyl-tRNA synthetase
  894 0960 glyQ Glycyl-tRNA synthetase alpha chain
  906 0972 glyS Glycyl-tRNA synthetase beta chain
  967 0417 metG Methionyl-tRNA synthetase
  978 0403 pheS Phenylalanyl-tRNA synthetase alpha chain
  979 0402 pheT Phenylalanyl-tRNA synthetase beta chain
  1080 1153 valS Valyl-tRNA synthetase
  1115 1190 hisS Histidyl-tRNA synthetase
  1162 1241 alaS Alanyl-tRNA synthetase
  1174 1253 trpS Tryptophanyl-tRNA synthetase
  1317 1422 ileS Isoleucyl-tRNA synthetase
  1373 1480 serS Seryl-tRNA synthetase
  1452 1547 leuS Leucyl-tRNA synthetase
 Degradation of proteins, peptides and glycopeptides
  29 0033 clpA ATP-dependent protease, ATP-binding subunit
  155 0169 Protease
  249 0264 clpB Heat shock protein
  271 0286 ftsH ATP-dependent Zn metallopeptidase
  356 1069 ftsH ATP-dependent Zn metallopeptidase
  387 1037 pepQ Proline peptidase
  405 1019 htrA Protease DO
  411 1012 Zn protease
  422 0470 pepF Oligopeptidase
  464 0515 hslV Heat shock protein
  465 0516 hslU Heat shock protein
  517 0570 pepA Aminopeptidase
  602 0657 Processing protease
  603 0658 gatB Glu-tRNA amidotransferase, subunit B
  730 0794 clpP ATP-dependent protease, proteolytic subunit
  769 0830 gatA Glu-tRNA amidotransferase, subunit A
  909 0975 gatC Glu-tRNA amidotransferase, subunit C
  999 0382 Zn-metallo protease
  1269 1350 prc Carboxyl-terminal protease
  1288 1374 clpX ATP-dependent protease, ATP-binding subunit
  1293 1379 lon ATP-dependent protease 1a
  1328 1435 sppA Protease
  1491 1584 ydiE O-Sialoglycoprotein endopeptidase
 Nucleoproteins
  774 0835 DNA-binding protein HU
  1052 1123 slyD FKBP-type peptidyl-prolyl cis-trans isomerase
 Protein modification
  210 0224 Peptide methionine sulfoxide reductase
  729 0793 def Polypeptide deformylase
  1017 0363 pcm Protein-l-isoaspartate O-methyltransferase
  1219 1299 map Methionine aminopeptidase
  1334 1441 ppiA Peptidyl-prolyl cis-trans isomerase
 Ribosomal proteins, synthesis and modification
  71 0076 rpsT 30S ribosomal protein S20
  76 0083 rpsI 30S ribosomal protein S9
  77 0084 rplM 50S ribosomal protein L13
  115 0125 rpmI 50S ribosomal protein L35
  116 0126 rplT 50S ribosomal protein L20
  186 0200 rpmF 50S ribosomal protein L32
  281 0296 rplU 50S ribosomal protein L21
  282 0297 rpmA 50S ribosomal protein L27
  357 1068 prmA Ribosomal protein L11 methyltransferase
  378 1047 rbfA Ribosome-binding factor A
  384 1040 rpsO 30S ribosomal protein S15
  443 0491 rpmB 50S ribosomal protein L28
  463 0514 rplI 50S ribosomal protein L9
  498 0551 rpmE 50S ribosomal protein L31
  509 0562 rpsU 30S ribosomal protein S21
  982 0399 rpsA 30S ribosomal protein S1
  1074 1147 rplS 50S ribosomal protein L19
  1078 1151 rpsP 30S ribosomal protein S16
  1119 1196 rpsG 30S ribosomal protein S7
  1120 1197 rpsL 30S ribosomal protein S12
  1122 1199 rplL 50S ribosomal protein L7/L12
  1123 1200 rplJ 50S ribosomal protein L10
  1124 1201 rplA 50S ribosomal protein L1
  1125 1202 rplK 50S ribosomal protein L11
  1127 1204 rpmG 50S ribosomal protein L33
  1165 1244 rpsR 30S ribosomal protein S18
  1167 1246 rpsF 30S ribosomal protein S6
  1212 1292 rplQ 50S ribosomal protein L17
  1214 1294 rpsD 30S ribosomal protein S4
  1215 1295 rpsK 30S ribosomal protein S11
  1217 1296 rpsM 30S ribosomal protein S13
  1217 1297 rpmJ 50S ribosomal protein L36
  1221 1301 rplO 50S ribosomal protein L15
  1222 1302 rpsE 30S ribosomal protein S5
  1223 1303 rplR 50S ribosomal protein L18
  1224 1304 rplF 50S ribosomal protein L6
  1225 1305 rpsH 30S ribosomal protein S8
  1226 1306 rpsN 30S ribosomal protein S14
  1227 1307 rplE 50S ribosomal protein L5
  1228 1308 rplX 50S ribosomal protein L24
  1229 1309 rplN 50S ribosomal protein L14
  1230 1310 rpsQ 30S ribosomal protein S17
  1231 1311 rpmC 50S ribosomal protein L29
  1232 1312 rplP 50S ribosomal protein L16
  1233 1313 rpsC 30S ribosomal protein S3
  1234 1314 rplV 50S ribosomal protein L22
  1235 1315 rpsS 30S ribosomal protein S19
  1236 1316 rplB 50S ribosomal protein L2
  1237 1317 rplW 50S ribosomal protein L23
  1238 1318 rplD 50S ribosomal protein L4
  1239 1319 rplC 50S ribosomal protein L3
  1240 1320 rpsJ 30S ribosomal protein S10
  1340 1447 rpmH 50S ribosomal protein L34
  1389 1496 rplY 50S ribosomal protein L25
  1445 1554 rpsB 30S ribosomal protein S2
 tRNA modification
  266 0281 tgt Queuine-tRNA-ribosyltransferase
  363 1062 queA S-Adenosylmethionine tRNA ribosyltransferase-isomerase
  1019 0361 truA Pseudouridylate synthase I
  1069 1141 fmt Methionyl-tRNA formyltransferase
  1075 1148 trmD tRNA (guanine-n1)-methyltransferase
  1254 1335 trmU tRNA(5-methylaminomethyl-2-thiouridylate)-methyltransferase
  1310 1415 miaA tRNA delta(2)-isopentenylpyrophosphate transferase
  1341 1448 rnpA RNase protein component
  1390 1497 pth Peptidyl-tRNA hydrolase
  1406 1513 selA l-Seryl-tRNA selenium transferase
 Translation factors
  72 0077 prfA Peptide chain release factor 1
  114 0124 infC Translation initiation factor IF-3
  157 0171 prfB Peptide chain release factor 2 (RF-2)
  163 0177 efp Elongation factor P (EF-P)
  232 0247 deaD ATP-dependent RNA helicase dead
  377 1048 infB Translation initiation factor IF-2
  1118 1195 fusA Elongation factor G (EF-G)
  1128 1205 tufA Elongation factor TU (EF-TU)
  1177 1256 frr Ribosome recycling factor (ribosome-releasing factor [RRF])
  1218 1298 infA Translation initiation factor IF-1
  1322 1431 ksgA Dimethyladenosine transferase
  1444 1555 tfs Elongation factor TS (EF-TS)
Transport and binding proteins
 General
  66 0071 ureI Urea transporter
  167 0179 ABC transporter, ATP-binding protein
  200 0214 Transporter
  235 0250 ABC transporter, ATP-binding protein
  236 0251 ABC transporter, permease
  300 0613 ABC transporter, ATP-binding protein
  343 1082 msbA Multidrug resistance protein
  449 0497 Transporter
  450 0498 Transporter
  547 0600 Secretion/efflux ABC transporter, ATP-binding protein
  553 0606 Efflux transporter
  554 0607 Efflux transporter
  653 0715 ABC transporter, ATP-binding protein
  685 0748 ABC transporter, ATP-binding protein
  754 0818 Osmoprotection binding protein
  757 0818 Osmoprotection binding protein
  758 0819 Osmoprotection ATP-binding protein
  789 0853 ABC transporter, ATP-binding protein
  806 0872 phnA Alkylphosphonate uptake protein
  871 0936 proP Proline/betaine transporter
  1055 1126 tolB tonB-independent protein-uptake protein
  1057 1129 exbD1 Biopolymer transport protein
  1058 1130 exbB1 Biopolymer transport protein
  1107 1181 Transporter
  1129 1206 ABC transporter, ATP-binding protein
  1141 1220 ABC transporter, ATP-binding protein
  1320 1427 Histidine-rich metal-binding protein
  1321 1432 Histidine- and glutamine-rich metal-binding protein
  1338 1445 exbB3 Biopolymer transport protein
  1339 1446 exbD3 Biopolymer transport protein
  1484 1576 ABC transporter, ATP-binding protein
  1485 1577 ABC transporter, permease
 Amino acids, peptides, and amines
  47 0055 putP Sodium/proline symporter
  121 0133 sdaC l-Serine transporter
  283 0298 dppA Periplasmic dipeptide transport substrate-binding protein
  284 0299 dppB Dipeptide transport system permease protein
  285 0300 dppC Dipeptide transport system permease protein
  286 0301 dppD Dipeptide transport system ATP-binding protein
  287 0302 dppF Dipeptide transport system ATP-binding protein
  406 1017 Amino acid permease
  874 0939 Amino acid ABC transporter, permease protein
  875 0940 Amino acid ABC transporter, binding protein precursor
  877 0942 Sodium/alanine symporter
  1096 1169 Amino acid ABC transporter, permease protein
  1097 1170 Amino acid ABC transporter, permease protein
  1098 1171 Amino acid ABC transporter, ATP-binding protein
  1099 1172 Amino acid ABC transporter, binding protein precursor
  1172 1251 Peptide ABC transporter, ATP-binding protein
  1358 1465 Amino acid ABC transporter, ATP-binding protein
  1399 1506 gltS Sodium/glutamate symporter
 Anions
  425 0473 modA Molybdenum ABC transporter, periplasmic binding protein
  426 0474 modB Molybdenum ABC transporter, permease
  427 0475 modC Molybdenum ABC transporter, ATP-binding protein
  1384 1491 Phosphate permease
 Carbohydrates, organic alcohols, and acids
  128 0140 lldP l-Lactate permease
  129 0141 lldP l-Lactate permease
  334 1091 kgtP α-Ketoglutarate permease
  635 0693 atoE Short-chain fatty acids transporter
  660 0724 dcuA Anaerobic C4-dicarboxylate membrane transporter
  1101 1174 gluP Glucose/galactose transporter
 Cations
  124 0136 bcp Bacterioferritin comigratory protein
  348 1077 nixA High-affinity nickel transport protein
  352 1073 copP Copper-associated protein
  353 1072 copA Copper-transporting P-type ATPase
  423 0471 kefB Glutathione-regulated potassium efflux system protein
  442 0490 Putative potassium channel protein
  529 0582 tonB1 Siderophore-mediated iron transport protein
  598 0653 pfr Nonheme iron-containing ferritin
  626 0686 fecA1 Iron(III) dicitrate transport protein
  627 0687 feoB Ferrous iron transport protein B
  727 0791 hmcT Heavy-metal cation-transporting P-type ATPase
  743 0807 fecA2 Iron(III) dicitrate transport protein
  821 0888 fecE Iron(III) dicitrate transport system ATP-binding protein
  822 0889 fecD Iron(III) dicitrate transport system permease protein
  903 0969 czcA1 Cation efflux system protein
  904 0970 czcB1 Cation efflux system protein
  1109 1183 Na+/H+ antiporter
  1248 1328 czcB2 Cation efflux system protein
  1249 1329 czcA2 Cation efflux system protein
  1258 1339 exbB2 Biopolymer transport protein
  1259 1340 exbD2 Biopolymer transport protein
  1260 1341 tonB2 Siderophore-mediated iron transport protein
  1263 1344 corA Magnesium and cobalt transport protein
  1396 1503 fixI Component of cation transport for cbb3-type oxidase
  1426 1400 fecA3 Iron(III) dicitrate transport protein
  1447 1552 nhaA Na+/H+ antiporter I
 Nucleosides, purines, and pyrimidines
  1106 1180 Nucleoside transporter
  1210 1290 pnuC Nicotinamide mononucleotide transporter
a

Gene numbers correspond to those in Fig. 1

TABLE 2.

Annotation and classification of genes from H. pylori J99 and 26695

Annotation category No. of genes in:
H. pylori J99 H. pylori 26695 Both strainsa
Functionally classified 877 898
Conserved with no known function 275 290
H. pylori specific 343 364
Total 1,495 1,552
Amino acid biosynthesis 44 44 44
Biosynthesis of cofactors, etc. 60b 59 59
Cell envelope 160 164 156cde
Cellular processes 96 113 92cde
DNA replication 23 23 21ce
DNA restriction-modification, etc. 66 68 51cde
Energy metabolism 104 104 102cde
Fatty acid and phospholipid metabolism 28 29 28e
Purine and pyrimidine biosynthesis 34 34 34
Regulatory functions 32 32 31df
Transcription 13 13 13
Translation 128 128 128
Transport and binding proteins 88g 87 87
Conserved with no known function 275 290 267cde
H. pylori specific 343 364 288cde
Total 1,495 1,552 1,401h
a

Using J99 genes as the basis for counting. 

b

Includes the partial duplication of folE (JHP862). 

c

Categories which include H. pylori J99-specific genes (see the text for details). 

d

These numbers include the “split” genes based on the H. pylori J99 definition. There are 6 J99 genes which constitute 12 26695 genes in the cell envelope; 2 J99 genes which constitute 4 26695 genes in cellular processes; 1 J99 gene which constitutes 2 26695 genes in each of DNA restriction-modification, energy metabolism, and Regulatory functions; 5 J99 genes which constitute 7 26695 genes in conserved with no known function; and 24 J99 genes which constitute 33 26695 genes in H. pylori specific. 

e

Categories which include H. pylori 26695-specific genes. 

f

Does not include the duplication of the response regulator (JHP1283 and JHP1443). 

g

Includes the partial duplication of proX (JHP754). 

h

The remaining 94 genes represent the 89 J99-specific genes, tnpA/B from IS606, and the partial or complete duplications of three genes. 

FIG. 1.

Linear representation of the H. pylori J99 chromosome, illustrating the location of each predicted protein-coding region, rRNA gene, tRNA gene, IS605 or IS606 element and related fragment, and NotI endonuclease site. The predicted protein-coding regions are color coded based on functional classification (see the bottom of the figure for the code), with the direction of transcription indicated by an arrowhead. H. pylori J99 ORFs are numbered sequentially in red, and the corresponding homologous gene, if it exists in strain 26695, is numbered in black. The positions of the NotI endonuclease sites in J99 are indicated with the number of conserved nucleotides in the recognition sequence (x/8) at the corresponding position in strain 26695. The numbers associated with the tRNA symbols (inverted triangles) represent the number of tRNA genes at a specific locus. Vertical hash marks, below the linear chromosome, are located every 20 kb.

Comparison of orthologous genes and their encoded products showed a high degree of conservation. Sequence variation between the two strains was significantly greater at the nucleotide level than at the amino acid level. Because the nucleotide variation occurred most commonly in the third position of a coding triplet, the primary sequence of the encoded protein was highly conserved (Table 3). The fact that many of the nucleotide differences are silent with respect to the protein sequence suggests that there is a strong selective pressure for functional conservation at the protein level.

TABLE 3.

Nucleotide and amino acid identity between genes common to H. pylori J99 and 26695

% Identity No. (%) of predicted ORFsa
Nucleotide Amino acid
100 0 (0) 41 (2.9)
98.0–99.9 8 (0.6) 269 (19.3)
96.0–97.9 249 (17.8) 359 (25.7)
94.0–95.9 566 (40.5) 279 (20.0)
92.0–93.9 306 (21.9) 169 (12.1)
90.0–91.9 89 (6.4) 86 (6.2)
85.0–89.9 81 (5.8) 77 (5.5)
<85 97 (7.0) 116 (8.3)
Total 1,396 (100) 1,396 (100)
a

Genes that appear “split” by putative frameshifts in either strain have been classed as the larger ORF in the above analysis. 

The nucleotide drift in the third position of a coding triplet is probably responsible for the majority of the DNA-based “diversity” reported for H. pylori (3, 4, 12, 54, 75, 77, 148). For example, pulsed-field gel electrophoresis mapping data have been interpreted to mean that the gene order and physical arrangement of the chromosome are highly variable from strain to strain (75, 148). By using this technique, strain J99 and strain 26695 would appear to be highly divergent in both the number of NotI fragments and gene location (6). This apparent genetic diversity is easily explained by two inversions in combination with the silent nucleotide drift, which is responsible for six of the seven additional NotI sites found in strain J99 compared to strain 26695 (6). Although the genomic content and the resulting physiological capabilities of the two strains are almost identical, these few differences in gene arrangement would have classified these strains as diverse. This example reveals the limitations of DNA-based methods when used to examine strain diversity.

NUTRITIONAL REQUIREMENTS

Amino Acids and Polyamines

Both sequenced strains of H. pylori have homologues to all the genes that would be needed to synthesize eight amino acids from central intermediary metabolites (Table 4). Studies of the growth requirements for several strains of H. pylori have shown an absolute need for arginine, histidine, leucine, isoleucine, valine, methionine, and phenylalanine (118, 132), a finding consistent with the genomic sequence information.

TABLE 4.

Predicted biosynthetic abilities and auxotrophies of H. pylori

Category and Compound No. of genes presenta Predicted synthetic abilityb
Amino acids
 Aspartic acid 2 (2) Y
 Cysteine 2 (3)c Y
 Glutamic acid 1 (1) Y
 Glutamine 1 (1) Y
 Glycine 1 (1) Y
 Lysine 6 (7)d Y
 Threonine 5 (5) Y
 Tryptophan 5 (5) Y
 Alanine 1 (2) N
 Arginine 0 (9) N
 Histidine 0 (8) N
 Isoleucine 2 (9) N
 Leucine 1 (6) N
 Methionine 1 (7) N
 Phenylalanine 1 (3) N
 Proline 1 (3) N
 Valine 2 (9) N
 Asparagine 0 (2)e ?
 Serine 2 (3) ?
 Tyrosine 1 (3) ?
Cofactors and vitamins
 Biotin 4 (4) Y
 CoA 0 (5)d Y
 Folate 7 (10)d Y
 Molybdopterin 8 (9)c Y
 Panothenate 4 (4) Y
 Protoheme 10 (10) Y
 Pyridine nucleotides 3 (5)d Y
 Pyridoxial phosphate 3 (?)d Y
 Riboflavin 6 (7)c Y
 Thiamine 3 (9)cd Y
 Thioredoxin 2 (2) Y
 B12
 Glutathione 0 (2) N
 Siroheme 0 (1) N
 Ubiquinone 3 (8) ?
 Menaquinone 0 (6) ?f
Polyamines
 Agmatine 1 (1) Y
 Putrescine 1 (3) N
 Spermidine 3 (5) ?g
Pyrimidines 11 (10)h Y
Purines 3 (10) Ni
a

Number of genes assigned in E. coli shown in parentheses. Data from reference 119

b

Y, yes; N, no; ?, not clear. 

c

H. pylori has a gene for each step in the biosynthetic pathway. The number of genes in E. coli is larger due to redundancy. 

d

Not all genes in this pathway have been identified in E. coli

e

Synthesis may occur via tRNA (see the text). 

f

The presence of menaquinones in H. pylori has been demonstrated (see the text). 

g

Spermidine synthesis may be mediated by NapC. 

h

Two copies of pyrC were found in H. pylori

i

Salvage pathway is present (see the text). 

Although no homologues to the genes involved in the amidation of aspartate were detected in H. pylori, several strains have been reported to grow in the absence of asparagine (132). It is possible that asparagine is synthesized by an aspartyl-tRNA-asparagine amidotransferase, similar to what has been observed with glutaminyl-tRNA biosynthesis in Bacillus subtilis (see “Transcription and translation” below) (29).

Both the serine and tyrosine biosynthetic pathways were complete except for a homologue to their respective specific transaminase. However, each of these reactions may be catalyzed by one of the several identified transaminases with undetermined substrate specificity (JHP206/HP220, JHP568/HP624, JHP673/HP736, and JHP976/HP405). Such an enzymatic activity would allow the de novo synthesis of these amino acids, as observed in some strains of H. pylori (132). Regardless of whether H. pylori can synthesize serine, it possesses a specific transporter which allows the acquisition of this amino acid from the environment. sdaC, which encodes the serine transporter, is contiguous with sdaB, whose protein product in Escherichia coli converts l-serine to pyruvate. Similarly, putP, which encodes a proline transporter, is adjacent to putA, which encodes a bifunctional enzyme that oxidizes proline to l-glutamate in E. coli. The alanine transporter gene (JHP877/HP0942) is clustered with two other genes (alr and dadA) which are involved in alanine metabolism (Fig. 2). A positive regulator of the dad operon is thought to be upstream of this gene cluster in E. coli. In H. pylori, a putative regulatory gene (JHP879/HP0944) has also been identified upstream of the dad gene cluster. This putative regulatory gene does not have homology to the putative E. coli regulator, a finding which may indicate that the regulation of alanine catabolism is different in these two species. The H. pylori gene clusters described above would allow for the uptake and utilization of serine, proline, and alanine as carbon and nitrogen sources. In addition, H. pylori has a transporter for the uptake of glutamate (JHP1399/HP1506), an amino acid abundant in gastric juice (82).

FIG. 2.

FIG. 2

Central metabolic pathways of H. pylori. Boxed compounds are key central intermediates. Black lines are reactions predicted to occur from the genomic analysis. Blue lines represent reactions that have been reported in the literature and are consistent with genomic analysis. Green lines represent a predicted reaction occurring only in strain 26695. Yellow lines represent a predicted reaction occurring only in strain J99. Broken magenta lines represent reactions reported in the literature but for which no homologue to the enzyme has been identified in either genome. Red lines represent key steps regulating gluconeogenesis/glycolysis.

H. pylori also encodes homologues for four other amino acid uptake systems with unknown specificity. One of these systems consists of a gene cluster encoding a multisubunit periplasmic permease (JHP1096–1099/HP1169–1172). The putative operon encodes two permeases, an ATP-binding protein and a periplasmic binding protein. Based on sequence similarity, the ligand for this high-affinity transporter may be either glutamine, histidine, or arginine. H. pylori is unable to synthesize the last two amino acids and therefore requires transport systems for them. Furthermore, the apparent inability of H. pylori to synthesize phenylalanine, methionine, and the branched-chain amino acids necessitates specific transport of these amino acids. No such specific transport systems were identified, but they may be encoded by any of the transport systems with unassigned ligand specificity.

In addition to transporting amino acids, H. pylori may have the ability to transport the abundant peptides which are found in the stomach. Homologues to a dipeptide transport system are present and its five genes (JHP283–287/HP0298–0302) are arranged contiguously, similar to the organization of the dpp operon in E. coli (1). There is also a single gene (JHP1172/HP1251) which displays significant sequence similarity to an oligopeptide transporter.

The genomic sequence provides little information on the composition of polyamines in H. pylori, which are needed for optimal growth in most cells. The homologue of SpeA allows the conversion of arginine to agmatine in H. pylori. Although H. pylori has a homologue to speE, which encodes spermidine synthetase, it is unlikely that this enzyme can catalyze spermidine biosynthesis since no homologues for the enzymes that provide precursors for SpeE (SpeB to SpeD) were detected. However, H. pylori may be able to synthesize spermidine through nspC. The product of this gene can synthesize spermidine by decarboxylating carboxyspermidine (117). It is also possible that H. pylori uses nspC for the synthesis of norspermidine, a polyamine found in Vibrio alginolyticus.

Cofactors and Vitamins

Both sequenced strains of H. pylori have all the identified genes needed for the biosynthesis of biotin, folate, heme, molybdopterin, pantothenate, pyridoxial phosphate, riboflavin, and thioredoxin (Table 4). H. pylori has all the genes necessary for the synthesis of NAD with the exception of nadB, which encodes the aspartate oxidase subunit of quinolate synthetase in E. coli (138). This polypeptide is the oxygen-utilizing subunit of an enzyme which converts l-aspartate to iminoaspartate. The absence of NadB is not unexpected since the mechanism by which anaerobic or microaerophilic bacteria, such as H. pylori, synthesize iminoaspartate is unknown and is not likely to be oxygen dependent. In addition, nicotinamide mononucleotide transporter (JHP1210/HP1290) was identified. No homologues to enzymes involved in vitamin B12 and coenzyme A biosynthesis were identified. Vitamin B12 is an important cofactor for certain enzymes involved in anaerobic metabolism such as methionine synthase. H. pylori may not need to synthesize this vitamin, since homologues to B12-requiring enzymes were not identified. Bacteria synthesize coenzyme A de novo from pantothenate. However, no homologues to known enzymes involved in its biosynthesis were identified in H. pylori, making this pathway unique with respect to those previously reported. The pathway for thiamine biosynthesis has not been completely defined. Some of the genes believed to be involved in thiamine synthesis were found, suggesting that H. pylori can make this vitamin. However, it has been reported that H. pylori requires thiamine for growth (118).

H. pylori has homologues to all of the genes necessary to produce riboflavin. A single gene in H. pylori, homologous to both ribB and ribA (JHP740/HP0804), encodes a bifunctional enzyme with both GTP cyclohydrolase II and 3,4-dihydroxy-2-butanone 4-phosphate synthase activities (158). In addition, H. pylori has a separate GTP cyclohydrolase II (RibA) homologue (JHP738/HP802) downstream from the bifunctional RibAB. Worst et al. (158) have shown that expression of RibAB but not RibA is regulated by iron limitation in H. pylori. The significance of this enzymatic duplication and differential gene regulation is unknown.

Purine and Pyrimidine Biosynthesis, Salvage, and Interconversion

Enzymes for the de novo biosynthesis of purines are largely absent in both sequenced strains of H. pylori, which implies that this bacterium cannot synthesize purine nucleotides from formate, glycine, or serine. Genes that encode homologues for all of the purine salvage and interconversion enzymes are present (Fig. 3A). No homologues to a purine transporter were identified, although there is biochemical evidence for the transport of purine bases in H. pylori (107, 118). Based on similarity, it is likely that the putative transporter (JHP1106/HP1180) is specific for nucleosides, which would allow H. pylori to obtain purines via the salvage and interconversion pathways.

FIG. 3.

FIG. 3

Pyrimidine salvage and interconversion (A) and purine salvage pathways (B). A, adenosine; G, guanine; HX, hypoxanthine; X, xanthine; R, ribonucleoside; dR, deoxyribonucleoside. Adapted from reference 119.

H. pylori possesses homologues to all of the genes necessary for the de novo synthesis of UTP and CTP, consistent with experimental results that show that radiolabeled pyrimidine nucleotide precursors are incorporated into DNA (108). Unlike E. coli, H. pylori possesses a second pyrC homologue encoding dihydroorotase, raising the possibility that the H. pylori enzyme exists as a heterodimer rather than as a homodimer as reported in E. coli. One of the two homologues (JHP528/HP0581) is more closely related to a PyrC in gram-negative organisms, whereas the other homologue (JHP251/HP0266) is more closely related to a PyrC in gram-positive organisms. Furthermore, H. pylori lacks a homologue for the regulatory chain of the aspartate transcarbamoylase (PyrI), suggesting that either a paralogous gene serves this function or PyrB functions in the absence of a regulatory subunit.

H. pylori has homologues for all of the enzymes used for the interconversion of pyrimidine deoxyribonucleotides (Fig. 3B) with the exception of thymidylate synthase (ThyA), which is required for the interconversion of dUMP to dTMP. In addition, homologues for all of the enzymes associated with the pyrimidine salvage pathway are absent except for DeoB. This absence is consistent with poor utilization of uracil and uridine and with the failure to detect incorporation of added thymine, cytosine, or deoxycytidine into DNA by H. pylori (108). Although thymidine kinase activity has been found in crude extracts of H. pylori (108), no gene encoding a homologue of thymidine kinase (tdk) was identified in H. pylori.

Inorganic Elements and Heavy Metals

Phosphorus is an essential element in bacteria. H. pylori possesses homologues to both polyphosphate kinase and inorganic pyrophosphatase. These enzymes confer the ability to synthesize and hydrolyze polyphosphate, in agreement with experimental evidence (17). H. pylori also possesses a phosphate transporter (JHP1384/HP1491).

Sulfur assimilation is restricted in H. pylori compared to E. coli. Homologues to the genes necessary for the assimilation of sulfide and cysteine (cysE and cysK) are present in both sequenced strains, whereas those for the assimilation of sulfate (cysA, cysC, cysD, cysH, and cysN), an energy-consuming process, are not. The absence of an identifiable sulfate permease supports the apparent inability of H. pylori to use sulfate. Whereas H. pylori can utilize only sulfide as a source of inorganic sulfur, the closely related bacterium Campylobacter jejuni has homologues to the genes necessary to assimilate sulfate, sulfite, and sulfide (based on analysis of the recently completed genome by the Sanger Centre). This difference in sulfur assimilation between H. pylori and Campylobacter spp. is also seen in sulfur dissimilation. Unlike many Campylobacter spp., H. pylori does not have the homologues necessary for the respiration of many sulfur compounds. The absence of these sulfur assimilatory and dissimilatory genes in H. pylori probably reflects the evolved physiology resulting from its unique gastric niche.

In E. coli and Salmonella typhimurium, nitrogen is derived mainly from the primary products of ammonia assimilation, i.e., glutamate and glutamine. Glutamine synthetase (GlnA) catalyzes the formation of glutamine, while either glutamate dehydrogenase (GdhA) or glutamate synthase (GltBD) catalyzes the formation of glutamate (131). Homologues to glnA and gdhA were identified, and the gene product of glnA has been characterized in H. pylori (45). Glutamate dehydrogenase and glutamine synthetase allow H. pylori to incorporate nitrogen from urea into amino acids, presumably via ammonia, as demonstrated previously (157). No homologue of GltBD was identified. In other bacteria, the absence of this enzyme results in the inability to grow in a medium with low levels of free ammonia (18, 32, 126). Presumably, H. pylori does not need GltBD because sufficient levels of free ammonia are generated by enzymes such as urease.

Homologues of genes belonging to several iron uptake systems were identified in both sequenced strains, indicating the importance of iron metabolism in H. pylori is similar to that in other pathogenic bacteria. H. pylori possesses homologues to some of the genes involved in the ferric citrate (Fec) transport system, but no identifiable homologues to FecB, a periplasmic protein, or FecC, a component of the cytoplasmic membrane channel, were found. Three homologues to genes encoding the outer membrane receptor FecA are present in H. pylori, one of which may be involved in iron uptake via the ferric citrate system. One of the three fecA homologues is adjacent to the feoB homologue, a gene encoding a cytoplasmic ferrous iron permease. These two genes may be involved in ferrous, rather than ferric, uptake. Another fecA homologue may be part of the TonB-dependent iron uptake system (55, 80). The tonB homologue is adjacent to homologues of the remaining two genes (exbB and exbD) comprising the TonB-dependent iron uptake system. In addition, two other sets of exbB and exbD genes were identified but are probably involved in other transport processes (13). H. pylori may also acquire iron through FrpB, for which four homologues were identified. In Neisseria spp., FrpB paralogs are outer membrane proteins that are induced at low iron concentrations, have homology to lactoferrin- and heme-binding proteins, and are postulated to be involved in iron acquisition. Under iron limitation, many bacteria use siderophores to acquire iron. Whether H. pylori produces siderophores is controversial (67, 70). No homologues to genes involved in siderophore biosynthesis were found. H. pylori may not need siderophores, because the amount of free inorganic iron present in the stomach should be sufficient to support bacterial growth.

In addition to iron transport, H. pylori possesses homologues to several other heavy-metal transporters, including NixA, which is responsible for Ni2+ uptake, a function necessary for urease activity (114), and CopAP, which is responsible for Cu2+ uptake (47). H. pylori also possesses homologues to a high-affinity, multisubunit molybdate uptake system, which is needed for the biosynthesis of molybdopterin.

Carbohydrates

Genomic analyses indicate that H. pylori has a limited capability to acquire sugars from the environment, which is in agreement with experimental findings (102). Only a homologue for a phosphoenolpyruvate-independent glucose and galactose transporter was identified, consistent with previous metabolic studies (98, 102). The apparent absence of other sugar uptake systems suggests a limited ability for sugar catabolism in H. pylori (see below). Several homologues to organic acid transporters were identified: two l-lactate permeases, a ketoglutarate permease, and a C4-dicarboxylate transporter that is used under anaerobic conditions in other bacteria. The presence of these transporters suggests that organic acids serve as important sources of carbon for H. pylori (see below).

In summary, based on the analysis of the sequence from both strains, H. pylori is capable of synthesizing the cofactors necessary for growth and acquiring important inorganic elements, although it is limited in its ability to use sulfur. H. pylori would be auxotrophic for at least nine amino acids and purines. Interestingly, no complex sugar transport or degradation homologues were found, suggesting that the bacterium does not acquire sugars from the environment and uses them as sources of energy or as sugar precursors.

CENTRAL INTERMEDIARY AND ENERGY METABOLISM

Central Intermediary Metabolism

Glycolysis and gluconeogenesis.

Genes involved in the metabolism of saccharides to simple sugars were not identified. This finding is consistent with metabolic studies which suggest that complex sugars are not a major energy source for H. pylori (102). Homologues for all enzymes, which carry out reversible steps in the pathway, are present in both sequenced strains of H. pylori. Since homologues of the two nonreversible gluconeogenic enzymes (fructose-1,6-bisphosphatase and pyruvate dikinase) are present and since homologues of the two nonreversible glycolytic enzymes (phosphofructose kinase and pyruvate kinase) were not identified, it appears that H. pylori uses the enzymes of the glycolytic/gluconeogenic pathway for anabolic biosynthesis rather than for catabolic energy production. The experimental evidence supports this hypothesis (59).

Entner-Doudoroff and phosphopentose pathways.

The genomic sequences show that H. pylori possesses homologues of all the genes involved in the Entner-Doudoroff pathway, suggesting that glucose can be used as a source of energy, which is consistent with published data (20, 103, 104). H. pylori has homologues of genes that encode all the enzymes in the phosphopentose shunt except gluconate-6-phosphate dehydrogenase. The presence of this enzymatic activity in crude extracts of H. pylori has been suggested (59, 100). However, the conversion of gluconate-6-phosphate to ribulose-5-phosphate could occur indirectly via the Entner-Doudoroff and the phosphopentose pathways (Fig. 2). The phosphopentose pathway enzymes that were identified in H. pylori allow for the generation of all the intermediates normally produced by this pathway.

Pyruvate metabolism.

The genomic analyses suggest that glucose or malate is not the primary source for production of pyruvate in H. pylori but, rather, that lactate, l-alanine, l-serine, and d-amino acids are the primary sources, which is consistent with the literature (106, 143). H. pylori appears only to convert pyruvate to acetyl coenzyme Δ (acetyl-6A) by pyruvate oxidoreductase (63, 64), lacking homologues for pyruvate dehydrogenase, pyruvate formate-lyase, and pyruvate oxidase. H. pylori dissimilates pyruvate to produce acetate, formate, succinate, and lactate (99, 106), an experimental observation consistent with the genomic analyses.

Fermentation.

H. pylori ferments pyruvate to acetate. Genetic analysis indicates that strain J99 can carry out this fermentation but that strain 26695 cannot do so due to a frameshift in its phosphotransacetylase gene. This mutation also implies that strain 26695 cannot convert acetate to acetyl-CoA by running the fermentative pathway in the reverse direction. However, in H. pylori 26695 the single, identifiable strain-specific gene involved in energy metabolism is an acetyl-CoA synthetase (HP1045) (149), which allows the direct conversion of acetate to acetyl-CoA.

The presence of alcohol dehydrogenase activity in H. pylori suggests that this bacterium can ferment pyruvate to ethanol (135, 136). Indeed, a homologue of this enzyme was identified in both strains. In addition, the single, identifiable strain-specific gene involved in energy metabolism in strain J99 (JHP1429) is a second alcohol dehydrogenase homologue. The role of alcohol dehydrogenase in H. pylori is unclear, because this organism apparently cannot ferment pyruvate to ethanol due to the absence of an identifiable acetaldehyde dehydrogenase homologue. The absence of such a homologue is consistent with biochemical studies (135, 136). The ability of H. pylori to produce acetaldehyde from ethanol (133) suggests that this bacterium is very sensitive to alcohols since it cannot detoxify the resulting acetaldehyde.

Tricarboxylic acid cycle.

The tricarboxylic acid (TCA) cycle of H. pylori in both sequenced strains is similar to the branched anaerobic TCA pathway used by E. coli (Fig. 2) with the following exceptions. Succinyl-CoA is generated from 2-oxoglutarate rather than from succinate (64). Furthermore, in H. pylori, fumarate is generated in the TCA pathway from aspartate rather than from malate. In contrast, genomic analysis suggests that in the closely related bacterium C. jejuni, succinyl-CoA is synthesized from succinate and that fumarate is synthesized from malate (based on analysis of the recently completed genome by the Sanger Centre). H. pylori possesses a homologue of fumarase, explaining the conversion of malate to fumarate observed in crude extracts (101). Unlike C. jejuni, no other malate-utilizing enzymes, such as malate dehydrogenase, malate synthase, or malate oxidoreductase, were identified in H. pylori.

Fatty acid degradation.

Both sequenced strains of H. pylori contain the genes necessary for C2 or short-chain fatty acid atabolism (25) and for a short-chain fatty acid transporter. No identifiable homologues were found to the genes involved in long-chain fatty acid β-oxidation. Together, these observations indicate that H. pylori may utilize acetoacetate and not acetobutyrate as a source for short chain fatty acid catabolites.

Electron Transport Chain

Electron donors.

The initial transfer of electrons during the oxidation of d-lactate and NADH may be performed by homologues to d-lactate dehydrogenase (dld), NADH dehydrogenase I (nuoA-nouN), and hydrogenase (hyaA to hyaDD). The presence of these genes in both sequenced strains is consistent with measured activities in H. pylori cell membranes (22, 93, 101, 105). Although homologues to NuoE and NuoF were not identified in H. pylori, the nuo cluster contained two ORFs in an identical location and of similar size to the two E. coli genes. Thus, it is likely that these two H. pylori ORFs encode proteins with orthologous functions to NuoE and NuoF. In addition, homologues for the following electron-transferring systems were found: pyruvate ferredoxin oxidoreductase (porA, porB, porG, and porD), glycerol-3-phosphate dehydrogenase (glpC), and proline dehydrogenase (putA).

No genes encoding a succinate dehydrogenase homologue were identified, although such an activity has been observed in extracts from several H. pylori strains, including J99 (34, 59, 120). This apparent discrepancy can be explained by the observation that fumarate reductase can convert succinate to fumarate in vitro. The importance of fumarate reductase in respiration depends on environmental conditions. Under microaerophilic conditions, fumarate reductase (46) is not essential in H. pylori, which explains why high concentrations of fumarate reductase-specific antimicrobials are required to inhibit the growth of and kill H. pylori in vitro (105). Under these conditions, oxygenic respiration may be used by the bacterium and hence reduces the importance of fumarate reductase in metabolism. In the absence of oxygen, this enzyme may be essential. In the presence of oxygen and fumarate, H. pylori, like members of the related genus Wolinella, may prefer fumarate as a terminal electron acceptor over performing oxygenic respiration (61). The role fumarate reductase plays in respiration cannot be assessed until the microenvironment of H. pylori is better defined.

Quinones and cytochromes.

Analysis of the genomic sequence provides no clear indication to the composition of the H. pylori quinone pool. No homologues to genes involved in the biosynthesis of menaquinones, elements of anaerobic respiration, were identified despite the reported presence of menaquinone-6 and menaquinone-1 in the cell membranes of H. pylori (52, 94). The absence of identifiable homologues implies that H. pylori obtains menaquinones either by synthesis with genes that have yet to be identified or by uptake from its environment. H. pylori does contain homologues for three ubiquinone-biosynthetic enzymes (UbiA, UbiD, and UbiE), but no significant homologues for UbiB, UbiC, UbiF, UbiG, or UbiH were identified. UbiB, UbiF, and UbiH are oxygen-utilizing enzymes, and their apparent absence in H. pylori might be the result of the microaerophilic metabolism of the bacterium.

The biosynthesis of cytochromes by H. pylori has been reviewed in detail recently (51, 125). H. pylori uses a type II system for such biosynthesis, which is similar to that in many gram-positive bacteria and some members of the β-proteobacteria. Both strains possess homologues to all of the components needed for this biosynthetic system.

Terminal electron acceptors.

There appear to be three putative terminal electron acceptor systems in both sequenced strains of H. pylori, i.e., fumarate reductase, N-oxide reductase, and cytochrome c oxidase, suggesting that H. pylori may be able to use fumarate, N-oxides (i.e., dimethyl sulfoxide and trimethylamine-N-oxide), or oxygen as electron sinks. Whereas the presence of a cytochrome c oxidase in H. pylori would allow aerobic respiration, the presence of both fumarate reductase and an N-oxide reductase suggests that the bacterium may respire anaerobically as well. The terminal oxidase complex is similar to cbb3-type oxidase complexes and is encoded by a gene cluster composed of homologues to genes encoding the Rhizobium FixN, FixO, FixP, and FixQ subunits. The arrangement of these genes is identical to that found in Rhizobium spp. (130), and the presence of such a terminal oxidase in H. pylori is consistent with previous findings (116).

ATP-Proton Motive Force Conversion

The bacterial ATP synthase, a multisubunit enzyme, is composed of the F0 complex, which consists of three subunits that form a proton channel, and the F1 complex, which consists of five subunits that constitute the catalytic site for ATP synthesis. In E. coli, all eight subunits are encoded within the atp operon (76). All five subunits of the F1 complex and the F0 b subunit are contiguous on the H. pylori chromosome. The remaining two subunits of the F0 complex are encoded by genes present in other chromosomal regions. H. pylori has an additional subunit (JHP1065/HP1137), which is homologous to the ATPase b′, a diverged and duplicated form of the b subunit found among plants and photosynthetic bacteria. The gene encoding this homologue is located at one end of the ATP synthase gene cluster. Functionally, the H. pylori ATPase is similar to other bacterial ATPases in that it uses the proton motive force generated by the electron transport chain to synthesize ATP (97).

Detoxification

Organisms that come in contact with oxygen, like the microaerophilic H. pylori, must be able to protect themselves from the toxic products of oxygen metabolism, such as superoxide and hydrogen peroxide. Both sequenced strains of H. pylori possess a superoxide dismutase and a catalase, consistent with previous biochemical findings (57, 122, 140). Thus, H. pylori is a microaerophile not because of an inability to neutralize the toxic products of oxygen metabolism but, more probably, as a consequence of other metabolic limitations. H. pylori has two genes encoding peroxidases (JHP991/HP0390 and JHP1471/HP1563), one of which is located adjacent to the superoxide dismutase gene. The ability to isolate catalase-negative mutants of H. pylori (122, 156) suggests that at least one of the peroxidases can function as a catalase.

In summary, it would appear that H. pylori can use amino acids or simple carbohydrates as a major sources of energy. The bacterium is restricted with respect to pyruvate metabolism. Further, H. pylori possesses an electron transport chain that can use oxygen as a terminal electron acceptor, but homologues to fumarate reductase and N-oxide reductase suggest that the bacterium is capable of at least limited anaerobic metabolism.

MACROMOLECULE BIOSYNTHESIS AND MODIFICATION

DNA Replication, Recombination, and Restriction-Modification

H. pylori contains genes encoding homologues to the DnaE, DnaN, DnaX, DnaQ, and HolB subunits of the DNA polymerase III holoenzyme, which is responsible for DNA replication. While the H. pylori holoenzyme contains fewer than the 10 subunits that comprise DNA polymerase III in E. coli, the total number of subunits is consistent with that found in other bacterial genomes (24, 31, 43, 81, 84, 139). Indeed, the only common subunits among these different species are DnaE, DnaN, and DnaX. H. pylori contains homologues to all genes encoding enzymes involved in initiation and DNA chain elongation, except dnaC, which is also absent in several other bacterial genomes (24, 31, 43, 81, 84, 139).

H. pylori possesses homologues to several nucleases, including UvrABC endonuclease, UvrD, ExoA, and RecJ. Even though H. pylori contains homologues to xseA and mutS, it lacks recognizable homologues encoding the other subunits of exonuclease VII (XseB) and the MutHLS endonuclease repair complex, respectively. Recombinational repair in H. pylori appears to occur in a RecBC-independent manner (92), since the RecBCD exonuclease V is absent and a RecR homologue was identified. Other homologues identified in the recombination system include RecA and RecN, while no homologue to recE encoded exonuclease VIII was found. In addition, homologues of ruvABC and recG, whose products are involved in the branch migration and resolution of Holliday structures, were identified in H. pylori. Both H. pylori genomes also possess several homologues to the DNA topoisomerase I gene (topA), some of which are strain specific (6). There are genes encoding several other ATP-dependent helicases in H. pylori, including a homologue for pcrA and rep. Both sequenced H. pylori strains have numerous genes with similarity to DNA restriction and modification enzymes, many of which are strain specific. This finding suggests that H. pylori strains have their own unique complement of these genes (2, 6). While many of the products of these genes can be classified as type I, II, or III restriction or modification enzymes, their exact specificity remains to be determined.

Both H. pylori genomes appear to have three type I systems. In all three systems, the modification (HsdM) and restriction (HsdR) subunits are highly homologous between the strains but the specificity subunits (HsdS) have limited identity. Domains of HsdS proteins can be shuffled to produce new specificity, and it is this modular nature which allows the type I systems to evolve rapidly to a new DNA specificity.

Some of the type II systems in H. pylori 26695 and J99 appear to be functionally equivalent and may possess the same DNA specificity, while others have identity within the methyltransferase enzyme but differences in the restriction enzyme. There are nine type II modification methylases common to both genomes (6). In addition, H. pylori J99 possesses two unique type II restriction-modification systems. In H. pylori 26695 and several other strains, the M.HpyI gene, encoding a type II modification enzyme (159), flanks iceA, which encodes a putative type II restriction endonuclease (149). The absence of iceA in H. pylori J99, a recent clinical isolate from a patient diagnosed with a duodenal ulcer, suggests that iceA is not required for gastrointestinal disease and may not represent an informative epidemiological marker of pathogenicity and virulence, as previously hypothesized (15, 152).

Both H. pylori 26695 and J99 contain two type III restriction-modification systems, one of which is strain specific. Whereas in the other system (JHP1410/1411, HP1521/1522) the restriction gene product is 93% identical in the two strains, the modification gene product is conserved only at the N and C termini. This difference may result in unique specificity for each modification enzyme. The modification gene appears to be regulated by a slipped-strand repair mechanism and is frameshifted in both J99 and 26695 (6).

Transcription and Translation

Analysis of the H. pylori genomes identified only three sigma factors (RpoD, RpoN, and FliA). The presence of these three sigma factors had been suggested by putative promotors found upstream of H. pylori genes (83, 86, 89, 122, 141, 147). No homologue to the stationary-phase sigma factor (RpoS) or the heat shock-specific sigma factor (RpoH) was identified, implying that H. pylori responds to stress in different fashion from that described in other bacteria. Both sequenced H. pylori strains contain a fusion of rpoB and rpoC, which encode the β and β′ subunits of RNA polymerase (149, 160). H. pylori contains homologues to three termination factors (nusA, nusB, and rho) and lacks identifiable homologues to the tRNA maturation genes rnd, rph, and rnpB. The lack of identifiable transcriptional termination stem-loop structures suggests that in H. pylori termination is largely Rho dependent (6).

All the aminoacyl-tRNA synthetases are present in H. pylori, except glutaminyl- and asparaginyl-tRNA synthetases. Two copies of the gene encoding glutamyl-tRNA synthetase (gltX) are present in both H. pylori strains. It had been suggested that one of these copies may function as a glutaminyl-tRNA synthetase (149). However, the presence of homologues to the gatA, gatB, and gatC genes, which have been demonstrated to replace glutaminyl-tRNA synthetase activity in Bacillus subtilis (29, 146), makes it more likely that glutaminyl-tRNA synthetase activity in H. pylori is encoded by the gatABC homologues rather than gltX (6). Thus, the role of the second glutamyl-tRNA synthetase in H. pylori is unclear. A similar transamidation reaction, encoded by these three homologues or other genes, may function in H. pylori to synthesize asparaginyl-tRNA from aspartyl-tRNA, explaining the ability of H. pylori to grow without added asparagine despite its apparent inability to synthesize this amino acid.

Fatty Acid and Phospholipid

Identifiable homologues were found to many of the genes required for initiation and elongation of fatty acid biosynthesis. Both H. pylori strains contain the characteristically small acyl carrier protein (ACP) (78 amino acids) (JHP744/HP0808). H. pylori 26695 but not J99 possesses a second, significantly larger (153-amino-acid) homologue with an extended N-terminal domain (HP962). Genomic analysis indicates that H. pylori has a homologue to cyclopropane fatty acid synthase (JHP969/HP0416), consistent with the experimental evidence that H. pylori has a preponderance of C19:0 cyclopropane chains (73). No homologue to β-hydroxydecanoyl-ACP dehydrase, which catalyzes the formation of cis-3-decenoyl-ACP, an important intermediate in the biosynthesis of unsaturated fatty acids in E. coli, was found. The absence of this homologue is surprising, since unsaturated fatty acids are present in H. pylori (73).

The phospholipid composition of H. pylori consists mainly of phosphatidylethanolamine, cardiolipin, and phosphotidylglycerol, with smaller quantities of phosphatidylserine and phosphatidylcholine (73). The genome appears to encode all of the proteins necessary for the synthesis of these phospholipids except for cardiolipin synthase (cls), which catalyzes the final step of cardiolipin synthesis in E. coli. The H. pylori genome encodes at least two of the three enzymes necessary for phosphatidic acid synthesis (JHP895/HP0961 and JHP1267/HP1348). A homologue to the glycerol-3-phosphate acyltransferase was not identified in the H. pylori genome.

A characteristic feature of the lipid profile of H. pylori is the presence of cholesterol glucosides, which account for about 25% of the total lipid of the bacterium (56, 58). H. pylori does not appear to encode known enzymes for the synthesis of cholesterol and presumably scavenges this molecule from the environment. Little is known about glucoside-modified cholesterol synthesis, but the enzymes responsible are expected to be found among the species-specific genes with unknown function.

Peptidoglycan

The cytoplasmic synthesis of UDP-activated precursors of bacterial peptidoglycan assembly is well understood. The H. pylori genome encodes homologues to all of the enzymes in this pathway, beginning with the synthesis of UDP-N-acetylmuramic acid and ending with UDP-disaccharide pentapeptide linked to an undecaprenol lipid carrier (Fig. 4).

FIG. 4.

FIG. 4

Peptidoglycan synthesis and recycling. GlcNAc, N-acetylglucosamine; MurNAc, N-acetylmuramic acid; Ala, alanine; Glu, glutamate, Dap, diaminopimelic acid. Adapted from reference 62.

After transport through the cytoplasmic membrane, peptidoglycan precursors are incorporated into the existing peptidoglycan layer by the penicillin-binding proteins (PBPs) (50). The genome of H. pylori encodes three proteins which show homology to PBPs, in agreement with a published gel electrophoresis study (68). The precise metabolic function of each PBP is uncertain. However, one of the PBPs (JHP544/HP597) has a transglycosylase motif. Since no additional genes with a similar motif were detected, this PBP may be the only enzyme in H. pylori that is involved in lengthening the glycan chain.

From the analysis of the genomes, it is uncertain if peptidoglycan fragments can be recycled by H. pylori. Although this bacterium has homologues to genes encoding a lytic amidase (JHP709/HP0772) and an N-acetylmuranoyl-l-alanine transglycosylase (JHP590/HP0645), it does not appear to have the other genes required for this recycling system (ampG, ampD, mpl) (127).

Outer Membrane

Approximately 4% of the coding capacity of both strains is devoted to outer membrane proteins. This amount is significantly larger than that of any other bacterial genome sequenced to date. The majority of these proteins belong to three paralogous families, the largest having 20 and 21 members in J99 and 26695, respectively (6). Several members of the largest paralogous family are porins (33, 41) or adhesins specific for the Lewis B carbohydrate moiety found on host cells (72). The sequence identity of orthologous members of this large family is high (greater than 95%), suggesting that strain-specific sequence differences play only a limited role in antigenic variation. Five orthologous pairs of this outer membrane protein family, including BabB (Lewis B adhesin), contain dinucleotide (CT) repeats in their signal sequences. Slipped-strand repair has been proposed to regulate the expression of these proteins (6, 149). Significantly, the predicted expression status (in frame or out of frame) of these five pairs is identical despite the presence of a different number of dinucleotide repeats in the two strains in each case (6).

Lipopolysaccharide

H. pylori has homologues to all enzymes required for 2-keto-3-deoxyoctulosonic acid (KDO)-lipid A biosynthesis. Compared to lipid A in E. coli, this moiety in H. pylori is underacylated, has longer fatty acid chains (C16 and C18), and lacks a phosphate group at the 4′-hydroxyl position on the nonreducing glucosamine of the disaccharide (115). Taken together, these findings suggest that in H. pylori lipid A is assembled as an acylated and diphosphorylated disaccharide, which is then modified by an unidentified phosphatase and esterase enzyme(s).

The chemical structures of the lipopolysaccharide cores from two H. pylori strains were shown to be identical heptasaccharides (9, 10). Synthesis of such a structure requires several glycosyltransferases. There are seven ORFs encoding these putative glycosyltransferases (JHP147/HP0159, JHP194/HP0208, JHP563/HP0619, JHP620/HP0679, JHP741/HP0805, JHP765/HP0826, and JHP1031/HP1105), which are common to both strains, three strain-specific ORFs in J99 (JHP562, JHP820, and JHP1032), and one strain-specific ORF in 26695 (HP1578). However, none of these ORFs can be assigned a substrate specificity. Whether the presence of these strain-specific glycosyltransferases in J99 and 26695 results in a different lipopolysaccharide core structure remains to be determined.

The O chain from lipopolysaccharides of H. pylori is composed of Lewis acids [Lex and Ley; mono- and difucosylated repeating disaccharides of β-(1,4)-linked galactose and N-acetylglucosamine, respectively] (9, 10). These carbohydrate moieties, which are identical to those found on host tissues, have been implicated in colonization and persistence and may also play a role in autoimmunity (7, 8). The biosynthetic pathway of these O chains has not been determined. One enzyme, α-(1,3)-fucosyltransferase (JHP596/HP0651), is to be involved in this pathway (21, 96). Each genome has two α-(1,3)-fucosyltransferases which differ in the number of a 7-amino-acid sequence repeat (YDDLRVN). Regulation of these genes appears to occur through slipped-strand repair at two distinct polynucleotide repeats (6).

Flagella

Flagellar biosynthesis in gram-negative bacteria has been extensively studied. The assembly of a functional flagellum requires numerous proteins and is a highly regulated process. Homologues to all of the required genes involved in flagellar biosynthesis were identified in both sequenced strains. In other gram-negative bacteria, including C. jejuni, inactivation of the biosynthetic pathway disrupts the expression of the system as a whole. However, in H. pylori, inactivation of the hook protein does not result in suppression of flagellin expression (124), indicating that flagellar biosynthesis is not as highly regulated as in other bacteria. This difference can be explained by the absence of FlgM in H. pylori, a protein that in other bacteria controls feedback regulation of the flagellar biosynthetic cascade.

The flagellar filament of H. pylori is composed of two flagellin subunits, FlaA and FlaB, and genes encoding both are present. Despite reports suggesting that the genes for these flagellin subunits exhibit strain variation (66, 123), in strains J99 and 26695 the FlaA protein sequences are identical and the FlaB protein sequences differ by only a single amino acid. In addition, the protein sequences reported for FlaA and FlaB from another strain are nearly identical to those from strains J99 and 26695 (89).

The flagellar filament of H. pylori is encased within a sheath that is continuous with the outer membrane (90, 91, 128). The sheath, whose composition has yet to be defined, probably protects the polymeric filament from dissociation in the low pH of the stomach. Likewise, other polymeric structures, such as fimbriae, would also be subject to dissociation at low pH, thus explaining why adhesion to the host epithelium by H. pylori appears to be mediated by integral outer membrane proteins (71, 72).

In summary, H. pylori has the necessary homologues for DNA, RNA, and protein synthesis.

CELLULAR PROCESSES

Protein Secretion

H. pylori has homologues to two leader peptidases, enzymes required for protein secretion. The type I leader peptidase, LepB, is responsible for cleavage of the signal sequence from most periplasmic and outer membrane proteins. The lspA (previously called ureD) gene product is the leader peptidase responsible for processing prelipoproteins.

The secretion of proteins through the cytoplasmic membrane utilizes specific machinery which consists of several sec gene products. H. pylori contains all the known Sec proteins except SecB and SecG. Mutation of secB, which encodes a chaperone, affects the secretion of only a limited number of proteins in E. coli, and other preproteins may utilize alternative chaperones. Indeed, it has been demonstrated that groESL mutants in E. coli have significant effects on the secretion of some proteins (85). Thus, it is likely that in H. pylori, other cytoplasmic chaperones are used to usher proteins, in conjunction with SecA, to the cytoplasmic membrane in preparation for secretion. SecE is proposed to be integral to the translocation machinery (121). Initial analysis of both genomes did not identify a SecE orthology. However, a region between JHP1126/HP1203 and JHP1127/HP1204 contains a 59-amino-acid ORF that has similarity to the functionally important region of E. coli SecE.

Cag Pathogenicity Island

H. pylori strains associated with clinically severe gastric disease (peptic ulcers) more commonly possess an approximately 40-kb pathogenicity island containing cytotoxin-associated genes (Cag pathogenicity island [cagPAI]) than do strains isolated from patients with uncomplicated gastritis (26). Type I strains have been defined as H. pylori isolates which have the entire cagPAI, express the cytotoxicity-associated immunodominant antigen (CagA) and an active vacuolating cytotoxin (VacA), and induce interleukin-8 (IL-8) secretion by gastric epithelial cells. In addition to cagA, it has been demonstrated by mutational analysis that several of the genes in the cagPAI are required for wild-type induction of IL-8 secretion by gastric epithelial cells. In contrast, Type II strains do not express CagA, have no vacuolating activity even though a truncated VacA may be produced, and do not induce IL-8 secretion at levels comparable to that induced by type I strains.

The initial cagPAI sequence was produced in two parts by two different laboratories, each of which sequenced clones from the same ordered cosmid library that had been constructed from H. pylori NCTC 11638 (2, 19). These groups showed that the cagPAI was separated into the cagI and cagII segments by an intervening stretch of DNA that was itself bordered on each side by a newly identified insertion sequence (IS605) element. In contrast, the cagPAI in H. pylori J99 and 26695, both type I strains, consists of the cagI and cagII segments fused as a single unit without the stretch of intervening DNA flanked on each side by IS605. In all three strains, the cagI and cagII segments are essentially the same and the cagPAI is found in the same relative location.

Many of the genes in the cagPAI are required for specific host cell responses to infection by H. pylori, including induction of (i) IL-8 secretion by gastric epithelial cells (2, 19, 137), (ii) tyrosine phosphorylation in host proteins (137), and (iii) cytoskeletal rearrangements during actin pedestal formation at the host cell surface (137). The manner in which cagPAI genes cause these host cell responses is unknown. However, five ORFs show significant sequence similarity to genes encoding the VirB protein family, which is responsible for DNA transfer in Agrobacterium tumefaciens (type IV secretion system) (23). These genes also have sequence similarity to genes involved in conjugative transfer of plasmids in E. coli and protein export in Bordetella pertussis (155). It is unknown how these components of conjugative and protein transport systems function in H. pylori. It has been suggested that the cagPAI encodes a type of contact-mediated secretion apparatus analogous to the type III secretion systems identified and characterized in several enteric pathogens such as Yersinia, Salmonella, Shigella, and pathogenic E. coli (88). It is therefore likely that the virB homologues of the cagPAI region encode components of the secretion apparatus which may deliver effector molecules (DNA or protein) directly to the host cell to elicit the responses mentioned above.

Insertion Elements

The IS element IS605 contains genes encoding two previously identified transposases flanked by a short nucleotide sequence with dyadic symmetry and a common central core sequence (19). IS605 transposes as a single unit in E. coli, suggesting that it could also be functional in H. pylori (79). One of the transposase genes (tnpA) is related to IS200 found in gram-negative bacteria, and the other (tnpB) is related to IS1341 found in a gram-positive thermophylic bacterium. It is unusual to find an insertion element with transposases from apparently two different origins. The IS605 element was first described within the cagPAI of NCTC 11638. Strain 26695 has five full copies of IS605 and one copy of a related insertion element, IS606, none of which is located within the cagPAI. Strain J99 has no complete copies of IS605 but has one copy of IS606 on its chromosome. The short flanking sequence of IS605, without the transposases (is605), is present on both ends of the cagPAI in strain NCTC 11638. These sequences flanking cagPAI are thought to be remnants of a recent transposition or of another type of recombinational event. The dyadic repeats of IS605 and IS606 are also found within the J99 and 26695 genomes, at both common and distinct locations. IS605 dyadic repeats are coincident, in one or the other genomes, with several of the major organizational differences between the two sequenced strains. H. pylori plasmid pHPM186 (GenBank accession no. AF077006) contains an IS605 element which is adjacent to three genes that flank an IS605 element in the plasticity zone of strain 26695 (6). This finding suggests that plasmid integration plays a role in generating the limited genomic diversity in H. pylori. Thus, insertion elements, such as IS605, may have been involved in the acquisition of cagPAI and the plasticity zone by H. pylori via horizontal transfer, as has also been postulated for other pathogenicity islands found in a wide range of pathogenic bacteria.

Transformation

Many H. pylori strains are naturally competent for DNA transformation, and the efficiency of this process varies from strain to strain. Besides the conjugative DNA transfer/protein export homologues (VirB proteins) encoded by the cagPAI, both strains contain additional members of this family, some of which are strain specific. Recently, some members of the VirB family have been shown to play a role in DNA transformation in H. pylori (60). The existence of strain-specific VirB family homologues may explain the variation in DNA transformation efficiency seen between strains. One additional transformation-associated gene (comEC), which is required for uptake of DNA into B. subtilis, was also identified.

Chemotaxis

Chemotaxis, the sensory adaptation mechanism by which motile bacteria recognize and react to environmental conditions, has been found in H. pylori (110). Three homologues of the chemotaxis pathway in E. coli (CheW, CheA, and CheY), as well as four methyl-accepting chemotaxis proteins (MCPs), which mediate specificity for ligands, were identified in both sequenced strains of H. pylori. Proteins with similarity to MCPs are not necessarily involved in flagellar chemotaxis (5, 30). Neither strain contains identifiable homologues to CheR or CheB, enzymes which, respectively, add methyl groups to or remove methyl groups from the MCPs, precisely modulating the chemotactic response. By contrast, C. jejuni does possess homologues to both CheR and CheB. Therefore, the chemotaxis observed in H. pylori may occur by a CheB- and CheR-independent mechanism, similar to that seen in CheB CheR mutants of E. coli (145). The apparent inability of H. pylori to precisely modulate chemotaxis may reflect its limited but unique gastric niche.

Both H. pylori strains have three homologues to CheV, another chemotaxis protein, which has an N-terminal domain similar to CheW and a C-terminal response regulator domain similar to CheY (44). The CheW protein and the N-terminal domain of CheV are both capable of modulating the CheA-MCP interaction (134). The three CheV orthologous pairs have greater than 97% identity, whereas none of the paralogues have more than 40% identity at the amino acid level, suggesting that each orthologous pair has functional similarity and that each paralogue has a different specific function.

Cell Division and Morphology

Several genes implicated in bacterial cell division were found in both sequenced strains of H. pylori. Among these are ftsZ and ftsA, which are adjacent to each other, as is generally observed (154). H. pylori does not possess an unambiguous homologue to FtsW, but it does possess two homologues to RodA, a protein with significant sequence similarity to FtsW (69). It is possible that one of the RodA homologues actually functions as FtsW. In E. coli, at least one of the peptidoglycan-synthesizing enzymes is specifically involved in cell division (142). H. pylori has three homologues to peptidoglycan-synthesizing enzymes, one of which could act specifically during cell division. Two homologues of the metalloprotease FtsH, phenotypically associated with cell division, were identified. Interestingly, mutagenesis studies have shown that one of the FtsH homologues (JHP356/HP1069) is essential for growth in vitro (48). This suggests that the second homologue (JHP271/HP0286) is unable to functionally replace the first homologue and probably has a different function. In addition, homologues of ftsK and fic were identified.

No homologues to ZipA, which is believed to initiate formation of the FtsZ ring (53), SulA, which is thought to inhibit the formation of the FtsZ ring (14), FtsQ, FtsN, or FtsL were identified. Also, no homologue to MinC, a cell division inhibitor, was found, whereas homologues to the activator (MinD) and cofactor (MinE) of MinC were identified.

Two homologues of cell morphology-determining proteins, RodA and MreB, were found. RodA is required for the catalytic activity of a PBP during elongation in E. coli (74), whereas the function of MreB is unknown.

Virulence Factors

A number of H. pylori proteins have been implicated in pathogenesis (for reviews, see references 36 and 95). All of the reported genes encoding potential virulence factors in H. pylori were identified in both strains, with the exception of iceA, which is missing from strain J99 (see “DNA replication, recombination, and restriction-modification” above).

Bacterial motility has been suggested to be required for colonization of the gastric mucosa, since H. pylori mutants unable to synthesize either of the flagellar subunits (FlaA or FlaB) cannot colonize gnotobiotic piglets (38, 40). Whether motility is needed for the persistence of an infection is not known, but owing to the rapid gastric epithelium and mucous turnover, it would probably be required.

The cytotoxin VacA induces vacuole formation in cultured epithelial cells and may be an important component in the induction of gastric cell lesions by H. pylori (27, 49). Different alleles of the vacA gene have been reported (11, 28). Strains J99 and 26695 possess different vacA alleles (s1b/ml and s1a/ml, respectively) and were isolated from patients with H. pylori-related disease of different severity. Epidemiological studies with humans have correlated the level of expression of the vacA gene with disease outcome (42), and particular vacA genotypes are associated with more severe disease symptoms (109, 144, 151). Interestingly, in gnotobiotic piglets, a mutant with a knockout mutation of vacA had no discernible effect on colonization, epithelial vacuolation, or gastritis, suggesting that VacA is not a virulence factor in this animal model (37).

The urease enzyme of H. pylori has been extensively studied (for reviews, see references 111 to 113) and has been shown to be a colonization factor (39, 78, 150). Urease is found in the cytoplasm as well as on the surface of H. pylori. The mechanism by which this enzyme is translocated is controversial (129, 153).

Other components which have been implicated in virulence, include the cagPAI, lipopolysaccharide, outer membrane proteins, and a number of enzymes, such as phospholipases, catalase, superoxide dismutase, and a mucinase homologue (36). The involvement of these components in pathogenesis remains to be elucidated. Homologues to the genes encoding all these products were present in both strains.

In summary, H. pylori possess homologues to the genes necessary for DNA replication, transcription, translation, and cell division, as well for as the synthesis of other important cellular macromolecules such as lipids and peptidoglycan. Each strain appears to possess a number of unique restriction enzymes that would result in selectivity to transformation with foreign DNA. Further features of this bacterium are the large number of related outer membrane proteins and the presence of a putative pathogenicity island.

CONCLUSION

The genomic analysis suggests that both strains have essentially identical metabolic potential. The strain-specific genes that encode proteins with an assigned function are predicted to have little impact on the physiology of H. pylori. For the most part, the reported experimental data for biochemical activities present or absent in H. pylori are in agreement with the predicted metabolic capabilities. Our genomic analysis reveals nutritional requirements which would restrict the environments in which H. pylori can survive and, in part, explains its limited niche. H. pylori has broad catabolic capabilities. While it can perform oxygenic respiration, anaerobic respiration, and fermentation, H. pylori is limited in what it can use as a carbon source. Carbohydrate utilization is restricted to glucose, via the Entner-Doudoroff pathway, and to sugars with shorter carbon backbones than C6, for which transport systems are present. In addition, H. pylori possesses numerous transporter systems for the uptake of amino acids, which must be available in the gastric environment. Other than carbohydrates, H. pylori apparently uses amino acids as an important source of carbon which would yield less energy than using hexoses and could result in slow growth as occurs in vitro. Further, the deamination of amino acids, together with the action of urease, requires a mechanism to deal with increased levels of ammonia.

Colonization by H. pylori involves an interaction between the outer membrane of the bacterium and the gastric epithelium of the host. The outer membrane composition of H. pylori is unique in its protein content and lipopolysaccharide structure, which are consistent with the persistence of H. pylori in a restricted niche. Compared to other bacteria, H. pylori devotes a significantly higher percentage of its coding capacity to that of outer membrane proteins, further emphasizing the importance of these proteins. A number of genes involved in determining the composition of the outer membrane are differentially regulated by slipped-strand repair (6). This differential regulation and strain-specific outer membrane-related genes may play a role in the severity of H. pylori-related disease and the ability of H. pylori to persist chronically in its host. The other strain-specific genes may also play a role in these aspects of pathogenesis. None of these strain-specific genes with an assigned function are predicted to have a significant impact on the pathophysiological capabilities of H. pylori, although those with an unassigned function may be important. However, there is evidence that host factors are involved in H. pylori-related diseases (35), and our genomic analyses suggest that these host factors may play a more significant role than was previously appreciated.

The analysis presented here, like previous analyses of other sequenced bacterial genomes, has found several biochemical pathways for which we were unable to identify all of the genes which should be present for that pathway to be functional. Some of these genes may not be present in the genome. In other cases, the biochemical activity may be present within the organism but the gene responsible for this activity may be unidentifiable by current in silico techniques. The apparent incompleteness of the same metabolic pathways in two unrelated strains may suggest that these pathways are functional in H. pylori and that the unidentified genes are different from previously described orthologues. These genes would be among the 40% of the genome to which no function has been assigned.

For the first time, the genomes of two strains from the same bacterial species have been compared (6). This publication provided an opportunity to begin defining the physiology of H. pylori, a globally important pathogen. Future genomic comparisons of multiple strains, carefully correlated with epidemiological data, will identify the minimal genomic complement of this species and the genes required for virulence. Such an approach will be applied to other pathogenic bacteria, and the genes identified from these studies will become candidates for therapeutic intervention.

REFERENCES

  • 1.Abouhamad W N, Manson M, Gibson M M, Higgins C F. Peptide transport and chemotaxis in Escherichia coli and Salmonella typhimurium: characterization of the dipeptide permease (Dpp) and the dipeptide-binding protein. Mol Microbiol. 1991;5:1035–1047. doi: 10.1111/j.1365-2958.1991.tb01876.x. [DOI] [PubMed] [Google Scholar]
  • 2.Akopyants N, Clifton S, Kersulyte D, Crabtree J, Youree B, Reece C, Bukanov N, Drazek E, Roe B, Berg D. Analyses of the cag pathogenicity island of Helicobacter pylori. Mol Microbiol. 1998;28:37–53. doi: 10.1046/j.1365-2958.1998.00770.x. [DOI] [PubMed] [Google Scholar]
  • 3.Akopyanz N, Bukanov N, Westblom T, Berg D. PCR-based RFLP analysis of DNA sequence diversity in the gastric pathogen Helicobacter pylori. Nucleic Acids Res. 1992;20:6221–6225. doi: 10.1093/nar/20.23.6221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Akopyanz N, Bukanov N, Westblom T, Kresovich S, Berg D. DNA diversity among clinical isolates of Helicobacter pylori detected by PCR-based RAPD fingerprinting. Nucleic Acids Res. 1992;20:5137–5142. doi: 10.1093/nar/20.19.5137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Alm R, Manning P. Characterization of the hlyB gene and its role in the production of the El Tor haemolysin of Vibrio cholerae O1. Mol Microbiol. 1990;4:413–425. doi: 10.1111/j.1365-2958.1990.tb00608.x. [DOI] [PubMed] [Google Scholar]
  • 6.Alm R A, Lee L-S, Moir D T, King B L, Brown E D, Doig P C, Smith D R, Noonan B, Guild B C, deJonge B L, Carmel G, Tummino P J, Caruso A, Uria-Nickelsen M, Mills D M, Ives C, Gibson R, Merberg D, Mills S D, Jiang Q, Taylor D E, Vovis G F, Trust T J. Genomic sequence comparison of two unrelated isolates of the human gastric pathogen Helicobacter pylori. Nature. 1999;397:176–180. doi: 10.1038/16495. [DOI] [PubMed] [Google Scholar]
  • 7.Appelmelk B, Negrini R, Moran A, Kuipers E. Molecular mimicry between Helicobacter pylori and the host. Trends Microbiol. 1997;5:70–73. doi: 10.1016/S0966-842X(96)10084-6. [DOI] [PubMed] [Google Scholar]
  • 8.Appelmelk B, Simoons-Smit I, Negrini R, Moran A, Aspinall G, Forte J, deVries T, Quan H, Verboom T, Maaskant J, Ghiara P, Kuipers E, Bloemena E, Tadema T, Townsend R, Tyagarajan K, Jr J C, Monteiro M, Savio A, deGraaff J. Potential role of molecular mimicry between Helicobacter pylori lipopolysaccharide and host Lewis blood group antigens in autoimmunity. Infect Immun. 1996;64:2031–2040. doi: 10.1128/iai.64.6.2031-2040.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Aspinall G O, Monteiro M A. Lipopolysaccharides of Helicobacter pylori strains P466 and Mo19—structures of the O antigen and core oligosaccharide regions. Biochemistry. 1996;35:2498–2504. doi: 10.1021/bi951853k. [DOI] [PubMed] [Google Scholar]
  • 10.Aspinall G O, Monteiro M A, Pang H, Walsh E J, Moran A P. Lipopolysaccharide of the Helicobacter pylori type strain NCTC 11637 (Atcc 43504)—structure of the O antigen chain and core oligosaccharide regions. Biochemistry. 1996;35:2489–2497. doi: 10.1021/bi951852s. [DOI] [PubMed] [Google Scholar]
  • 11.Atherton J, Cao P, Jr R P, Tummuru M, Blaser M, Cover T. Mosaicism in vacuolating cytotoxin alleles of Helicobacter pylori. J Biol Chem. 1995;270:17771–17777. doi: 10.1074/jbc.270.30.17771. [DOI] [PubMed] [Google Scholar]
  • 12.Berg D, Gilman R, Lelwala-Guruge J, Srivastava K, Valdez Y, Watanabe J, Miyagi J, Akopyants N, Ramirez-Ramos A, Yoshiwara T, Recavarren S, Leon-Barua R. Helicobacter pylori populations in Peruvian patients. Clin Infect Dis. 1997;25:996–1002. doi: 10.1086/516081. [DOI] [PubMed] [Google Scholar]
  • 13.Berg D E, Hoffman P S, Appelmelk B J, Kusters J G. The Helicobacter pylori genome sequence: genetic factors for long life in the gastric mucosa. Trends Microbiol. 1997;5:468–474. doi: 10.1016/s0966-842x(97)01164-5. [DOI] [PubMed] [Google Scholar]
  • 14.Bi E, Lutkenhaus J. Cell division inhibitors SulA and MinCD prevent formation of the FtsZ ring. J Bacteriol. 1993;175:1118–1125. doi: 10.1128/jb.175.4.1118-1125.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Blaser M. Heterogeneity of Helicobacter pylori. Eur J Gastroenterol Hepatol. 1997;9:S3–S6. doi: 10.1007/978-94-009-1792-7_3. [DOI] [PubMed] [Google Scholar]
  • 16.Blaser M. Not all Helicobacter pylori strains are created equal: should all be eliminated? Lancet. 1997;349:1020–1022. doi: 10.1016/S0140-6736(96)09133-7. [DOI] [PubMed] [Google Scholar]
  • 17.Bode G, Mauch F, Ditschuneit H, Malfertheiner P. Identification of structures containing polyphosphate in Helicobacter pylori. J Gen Microbiol. 1993;139:3029–3033. doi: 10.1099/00221287-139-12-3029. [DOI] [PubMed] [Google Scholar]
  • 18.Brenchley J E, Magasanik B. Mutants of Klebsiella aerogenes lacking glutamate dehydrogenase. J Bacteriol. 1974;117:544–550. doi: 10.1128/jb.117.2.544-550.1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Censini S, Lange C, Xiang Z, Crabtree J, Ghiara P, Borodovsky M, Rappuoli R, Covacci A. cag, a pathogenicity island of Helicobacter pylori, encodes type I-specific and disease-associated virulence factors. Proc Natl Acad Sci USA. 1996;93:14648–14653. doi: 10.1073/pnas.93.25.14648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chalk P A, Roberts A D, Blows W M. Metabolism of pyruvate and glucose by intact cells of Helicobacter pylori studied by 13C NMR spectroscopy. Microbiology. 1994;140:2085–2092. doi: 10.1099/13500872-140-8-2085. [DOI] [PubMed] [Google Scholar]
  • 21.Chan G Z, Palcic M M, Taylor D E. Cloning and heterologous expression of an α(1,3)-fucosyltransferase from the gastric pathogen Helicobacter pylori. J Biol Chem. 1997;272:21357–21363. doi: 10.1074/jbc.272.34.21357. [DOI] [PubMed] [Google Scholar]
  • 22.Chang H T, Marcelli S W, Davison A A, Chalk P A, Poole R K, Miles R J. Kinetics of substrate oxidation by whole cells and cell membranes of Helicobacter pylori. FEMS Microbiol Lett. 1995;129:33–38. doi: 10.1016/0378-1097(95)00130-W. [DOI] [PubMed] [Google Scholar]
  • 23.Christie P J. Agrobacterium tumefaciens T-complex transport apparatus: a paradigm for a new family of multifunctional transporters in eubacteria. J Bacteriol. 1997;179:3085–3094. doi: 10.1128/jb.179.10.3085-3094.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cole S, Brosch R, Parkhill J, Garnier T, Churcher C, Harris D, Gordon S, Eiglmeier K, Gas S, III C B, Tekaia F, Badcock K, Basham D, Brown D, Chillingworth T, Connor R, Davies R, Devlin K, Feltwell T, Gentles S, Hamlin N, Holroyd S, Hornsby T, Jagels K, Krogh A, McLean J, Moule S, Murphy L, Oliver K, Osborne J, Quail M, Rajandream M-A, Rogers J, Rutter S, Seeger K, Skelton J, Squares R, Squares S, Sulston J, Taylor K, Whitehead S, Barrell B. Deciphering the biology of Mycobacterium tuberculosis from the complete genome sequence. Nature. 1998;393:537–544. doi: 10.1038/31159. [DOI] [PubMed] [Google Scholar]
  • 25.Corthesy-Theulaz I E, Bergonzelli G E, Henry H, Bachmann D, Schorderet D F, Blum A L, Ornston L N. Cloning and characterization of Helicobacter pylori succinyl CoA:acetoacetate CoA-transferase, a novel prokaryotic member of the CoA-transferase family. J Biol Chem. 1997;272:25659–25667. doi: 10.1074/jbc.272.41.25659. [DOI] [PubMed] [Google Scholar]
  • 26.Covacci A, Falkow S, Berg D, Rappuoli R. Did the inheritance of a pathogenicity island modify the virulence of Helicobacter pylori? Trends Microbiol. 1997;5:205–208. doi: 10.1016/S0966-842X(97)01035-4. [DOI] [PubMed] [Google Scholar]
  • 27.Cover T, Blaser M. Helicobacter pylori and gastroduodenal disease. Annu Rev Med. 1992;42:135–145. doi: 10.1146/annurev.me.43.020192.001031. [DOI] [PubMed] [Google Scholar]
  • 28.Cover T L, Tummuru M K, Cao P, Thompson S A, Blaser M J. Divergence of genetic sequences for the vacuolating cytotoxin among Helicobacter pylori strains. J Biol Chem. 1994;269:10566–10573. [PubMed] [Google Scholar]
  • 29.Curnow A, Hong K, Yuan R, Kim S, Martins O, Winkler W, Henkin T, Soll D. Glu-tRNAGln amidotransferase: a novel heterotrimeric enzyme required for correct decoding of glutamine codons during translation. Proc Natl Acad Sci USA. 1997;94:11819–11826. doi: 10.1073/pnas.94.22.11819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Darzins A. Characterization of a Pseudomonas aeruginosa gene cluster involved in pilus biosynthesis and twitching motility: sequence similarity to the chemotaxis proteins of enterics and the gliding bacterium Myxococcus xanthus. Mol Microbiol. 1994;11:137–153. doi: 10.1111/j.1365-2958.1994.tb00296.x. [DOI] [PubMed] [Google Scholar]
  • 31.Deckert G, Warren P V, Gaasterland T, Young W G, Lenox A L, Graham D E, Overbeek R, Snead M A, Keller M, Aujay M, Huber R, Feldman R A, Short J M, Olsen G J, Swanson R V. The complete genome of the hyperthermophilic bacterium Aquifex aeolicus. Nature. 1998;392:353–358. doi: 10.1038/32831. [DOI] [PubMed] [Google Scholar]
  • 32.Dendinger S M, Patil L G, Brenchley J E. Salmonella thyphimurium mutants with altered glutamate dehydrogenase and glutamate synthase activities. J Bacteriol. 1980;141:140–148. doi: 10.1128/jb.141.1.190-198.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Doig P, Exner M, Hancock R, Trust T. Isolation and characterization of a conserved porin protein from Helicobacter pylori. J Bacteriol. 1995;177:5447–5452. doi: 10.1128/jb.177.19.5447-5452.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Doig P, Trust T. Identification of surface-exposed outer membrane antigens of Helicobacter pylori. Infect Immun. 1994;62:4526–4533. doi: 10.1128/iai.62.10.4526-4533.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Dubois A, Berg D, Incecik E, Fiala N, Heman-Ackah L, Perez-Perez G, Blaser M. Transient and persistent infection of nonhuman primates with Helicobacter pylori: implications for human disease. Infect Immun. 1996;64:2885–2891. doi: 10.1128/iai.64.8.2885-2891.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Dunn B, Cohen H, Blaser M. Helicobacter pylori. Clin Microbiol Rev. 1997;10:720–741. doi: 10.1128/cmr.10.4.720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Eaton K, Cover T, Tummuru M, Blaser M, Krakowka S. Role of vacuolating cytotoxin in gastritis due to Helicobacter pylori in gnotobiotic piglets. Infect Immun. 1997;65:3462–3464. doi: 10.1128/iai.65.8.3462-3464.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Eaton K, Morgan D, Krakowka S. Motility as a factor in the colonisation of gnotobiotic piglets by Helicobacter pylori. J Med Microbiol. 1992;37:123–127. doi: 10.1099/00222615-37-2-123. [DOI] [PubMed] [Google Scholar]
  • 39.Eaton K A, Brooks C L, Morgan D R, Krakowka S. Essential role of urease in pathogenesis of gastritis induced by Helicobacter pylori in gnotobiotic piglets. Infect Immun. 1991;59:2470–2475. doi: 10.1128/iai.59.7.2470-2475.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Eaton K A, Suerbaum S, Josenhans C, Krakowka S. Colonization of gnotobiotic piglets by Helicobacter pylori deficient in two flagellin genes. Infect Immun. 1996;64:2445–2448. doi: 10.1128/iai.64.7.2445-2448.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Exner M, Doig P, Trust T, Hancock R. Isolation and characterization of a family of porin proteins from Helicobacter pylori. Infect Immun. 1995;63:1567–1572. doi: 10.1128/iai.63.4.1567-1572.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Forsyth M H, Atherton J C, Blaser M J, Cover T L. Heterogeneity in levels of vacuolating cytotoxin gene (vacA) transcription among Helicobacter pylori strains. Infect Immun. 1998;66:3088–3094. doi: 10.1128/iai.66.7.3088-3094.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Fraser C M, Norris S J, Weinstock G M, White O, Sutton G G, Dodson R, Gwinn M, Hickey E K, Clayton R, Ketchum K A, Sodergren E, Hardham J M, McLeod M P, Salzberg S, Peterson J, Khalak H, Richardson D, Howell J K, Chidambaram M, Utterback T, McDonald L, Artiach P, Bowman C, Cotton M D, Venter J C, et al. Complete genome sequence of Treponema pallidum, the syphilis spirochete. Science. 1998;281:375–388. doi: 10.1126/science.281.5375.375. [DOI] [PubMed] [Google Scholar]
  • 44.Fredrick K, Helmann J. Dual chemotaxis signaling pathways in Bacillus subtilis: a sigma D-dependent gene encodes a novel protein with both CheW and CheY homologous domains. J Bacteriol. 1994;176:2727–2735. doi: 10.1128/jb.176.9.2727-2735.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Garner R M, Fulkerson J J, Mobley H L. Helicobacter pylori glutamine synthetase lacks features associated with transcriptional and posttranslational regulation. Infect Immun. 1998;66:1839–1847. doi: 10.1128/iai.66.5.1839-1847.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ge Z, Jiang Q, Kalisiak M S, Taylor D E. Cloning and functional characterization of Helicobacter pylori fumarate reductase operon comprising three structural genes coding for subunits C, A and B. Gene. 1997;204:227–234. doi: 10.1016/s0378-1119(97)00550-7. [DOI] [PubMed] [Google Scholar]
  • 47.Ge Z, Taylor D E. Helicobacter pylori genes hpcopA and hpcopP constitute a cop operon involved in copper export. FEMS Microbiol Lett. 1996;145:181–188. doi: 10.1111/j.1574-6968.1996.tb08575.x. [DOI] [PubMed] [Google Scholar]
  • 48.Ge Z, Taylor D E. Sequencing, expression, and genetic characterization of the Helicobacter pylori ftsH gene encoding a protein homologous to members of a novel putative ATPase family. J Bacteriol. 1996;178:6151–6157. doi: 10.1128/jb.178.21.6151-6157.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ghiara P, Marchetti M, Blaser M J, Tummuru M K, Cover T L, Segal E D, Tompkins L S, Rappuoli R. Role of the Helicobacter pylori virulence factors vacuolating cytotoxin, CagA, and urease in a mouse model of disease. Infect Immun. 1995;63:4154–4160. doi: 10.1128/iai.63.10.4154-4160.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ghuyssen J-M. Serine β-lactamases and penicillin-binding proteins. Annu Rev Microbiol. 1994;45:37–67. doi: 10.1146/annurev.mi.45.100191.000345. [DOI] [PubMed] [Google Scholar]
  • 51.Goldman B S, Kranz R G. Evolution and horizontal transfer of an entire biosynthetic pathway for cytochrome C biogenesis—Helicobacter, Deinococcus, Archaea and more. Mol Microbiol. 1998;27:871–873. doi: 10.1046/j.1365-2958.1998.00708.x. [DOI] [PubMed] [Google Scholar]
  • 52.Goodwin C S, Collins M D, Blincow E. The absence of thermoplasmaquinones in Campylobacter pyloridis, and its temperature and pH growth range. Microbiol Lett. 1986;32:137–140. [Google Scholar]
  • 53.Hale C A, deBoer P A J. Direct binding of FtsZ to ZipA, an essential component of the septal ring structure that mediates cell division in Escherichia coli. Cell. 1997;96:1–20. doi: 10.1016/s0092-8674(00)81838-3. [DOI] [PubMed] [Google Scholar]
  • 54.Han J, Yu E, Lee I, Lee Y. Diversity among clinical isolates of Helicobacter pylori in Korea. Mol Cells. 1997;7:544–547. [PubMed] [Google Scholar]
  • 55.Hancock R E, Hantke K, Braun V. Iron transport in Escherichia coli K-12. 2,3-Dihydroxybenzoate-promoted iron uptake. Arch Microbiol. 1997;114:231–239. doi: 10.1007/BF00446867. [DOI] [PubMed] [Google Scholar]
  • 56.Haque M, Hirai Y, Yokota K, Mori N, Jahan I, Ito H, Hotta H, Yano I, Kanemasa Y, Oguma K. Lipid profile of Helicobacter spp.: presence of cholesterol glucoside as a characteristic feature. J Bacteriol. 1996;178:2065–2070. doi: 10.1128/jb.178.7.2065-2070.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hazell S L, Evans D J, Jr, Graham D Y. Helicobacter pylori catalase. J Gen Microbiol. 1991;137:57–61. doi: 10.1099/00221287-137-1-57. [DOI] [PubMed] [Google Scholar]
  • 58.Hirai Y, Haque M, Yoshida T, Yokota K, Yasuda T, Oguma K. Unique cholesteryl glucosides in Helicobacter pylori: composition and structural analysis. J Bacteriol. 1995;177:5327–5333. doi: 10.1128/jb.177.18.5327-5333.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hoffman P S, Goodwin A, Johnsen J, Magee K, Veldhuyzen van Zanten S J. Metabolic activities of metronidazole-sensitive and -resistant strains of Helicobacter pylori: repression of pyruvate oxidoreductase and expression of isocitrate lyase activity correlate with resistance. J Bacteriol. 1996;178:4822–4829. doi: 10.1128/jb.178.16.4822-4829.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hofreuter D, Odenbreit S, Henke G, Haas R. Natural competence for DNA transformation in Helicobacter pylori: identification and genetic characterization of the comB locus. Mol Microbiol. 1998;28:1027–1038. doi: 10.1046/j.1365-2958.1998.00879.x. [DOI] [PubMed] [Google Scholar]
  • 61.Holt J G, Krieg N R, Sneath P H A, Staley J T, Williams S T. Bergey’s manual of determinative bacteriology. 9th ed. Baltimore, Md: The Williams & Wilkins Co.; 1994. [Google Scholar]
  • 62.Holtje J V. Growth of the stress-bearing and shape-maintaining murine sacculus of Escherichia coli. Microbiol Mol Biol Rev. 1998;62:181–203. doi: 10.1128/mmbr.62.1.181-203.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hughes N J, Chalk P A, Clayton C L, Kelly D J. Identification of carboxylation enzymes and characterization of a novel four-subunit pyruvate:flavodoxin oxidoreductase from Helicobacter pylori. J Bacteriol. 1995;177:3953–3959. doi: 10.1128/jb.177.14.3953-3959.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hughes N J, Clayton C L, Chalk P A, Kelly D J. Helicobacter pylori porCDAB and oorDABC genes encode distinct pyruvate:flavodoxin and 2-oxoglutarate:acceptor oxidoreductases which mediate electron transport to NADP. J Bacteriol. 1998;180:1119–1128. doi: 10.1128/jb.180.5.1119-1128.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hunt R. The role of Helicobacter pylori in pathogenesis: the spectrum of clinical outcomes. Scand J Gastroenterol Suppl. 1996;220:3–9. [PubMed] [Google Scholar]
  • 66.Hurtado A, Owen R J, Desai M. Flagellin gene profiling of Helicobacter pylori infecting symptomatic and asymptomatic individuals. Res Microbiol. 1994;145:585–594. doi: 10.1016/0923-2508(94)90075-2. [DOI] [PubMed] [Google Scholar]
  • 67.Husson M O, Legrand D, Spik G, Leclerc H. Iron acquisition by Helicobacter pylori: importance of human lactoferrin. Infect Immun. 1993;61:2694–2697. doi: 10.1128/iai.61.6.2694-2697.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ikeda F, Yokota Y, Mine Y, Tatsuta M. Activity of cefixime against Helicobacter pylori and affinities for the penicillin-binding proteins. Antimicrob Agents Chemother. 1990;34:2426–2428. doi: 10.1128/aac.34.12.2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ikeda M, Sato T, Wachi M, Jung H K, Ishino F, Kobayashi Y, Matsuhashi M. Structural similarity among Escherichia coli FtsW and RodA proteins and Bacillus subtilis SpoVE protein, which function in cell division, cell elongation, and spore formation, respectively. J Bacteriol. 1989;171:6375–6378. doi: 10.1128/jb.171.11.6375-6378.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Illingworth D S, Walter K S, Griffiths P L, Barclay R. Siderophore production and iron-regulated envelope proteins of Helicobacter pylori. Int J Med Microbiol Virol Parasitol Infect Dis. 1993;280:113–119. [PubMed] [Google Scholar]
  • 71.Ilver D, Arnquist A, Frick I-M, Engstrand L, Covacci A, Magnusson K-E, Boren T. The Helicobacter pylori blood group antigen binding adhesin. Gut. 1996;39:A55. . (Abstract.) [Google Scholar]
  • 72.Ilver D, Arnqvist A, Ogren J, Frick I, Kersulyte D, Incecik E, Berg D, Covacci A, Engstrand L, Boren T. Helicobacter pylori adhesin binding fucosylated histo-blood group antigens revealed by retagging. Science. 1998;279:373–377. doi: 10.1126/science.279.5349.373. [DOI] [PubMed] [Google Scholar]
  • 73.Inamoto Y, Ariyama S, Hamanaka Y, Okita K, Kaneta Y, Nagate T, Kondou I, Takemoto T. Lipid analysis of Helicobacter pylori. J Clin Gastroenterol. 1993;17:S136–S139. doi: 10.1097/00004836-199312001-00024. [DOI] [PubMed] [Google Scholar]
  • 74.Ishino F, Park W, Tomioka S, Tamaki S, Takase I. Peptidoglycan synthetic activities in membranes of Escherichia coli caused by overproduction of penicillin-binding protein 2 and RodA protein. J Biol Chem. 1986;261:7024–7031. [PubMed] [Google Scholar]
  • 75.Jiang Q, Hiratsuka K, Taylor D. Variability of gene order in different Helicobacter pylori strains contributes to genome diversity. Mol Microbiol. 1996;20:833–842. doi: 10.1111/j.1365-2958.1996.tb02521.x. [DOI] [PubMed] [Google Scholar]
  • 76.Kadner R. Cytoplasmic membrane. In: Neidhardt F C, Curtiss III R, Ingraham J L, Lin E C C, Low K B, Magasanik B, Reznikoff W S, Riley M, Schaechter M, Umbarger H E, editors. Escherichia coli and Salmonella: cellular and molecular biology. Vol. 1. Washington, D.C: ASM Press; 1996. pp. 58–87. [Google Scholar]
  • 77.Kansau I, Raymond J, Bingen E, Courcoux P, Kalach N, Bergeret M, Braimi N, Dupont C, Labigne A. Genotyping of Helicobacter pylori isolates by sequencing of PCR products and comparison with the RAPD technique. Res Microbiol. 1996;147:661–669. doi: 10.1016/0923-2508(96)84023-x. [DOI] [PubMed] [Google Scholar]
  • 78.Karita M, Tsuda M, Nakazawa T. Essential role of urease in vitro and in vivo Helicobacter pylori colonization study using a wild-type and isogenic urease mutant strain. J Clin Gastroenterol. 1995;21:S160–163. [PubMed] [Google Scholar]
  • 79.Kersulyte D, Akopyants N S, Clifton S, Roe B S, Berg D E. Novel sequence organization and insertion specificity of IS605 and IS606: chimeric transposable elements of Helicobacter pylori. Gene. 1998;223:175–186. doi: 10.1016/s0378-1119(98)00164-4. [DOI] [PubMed] [Google Scholar]
  • 80.Klebba P E, Rutz J M, Liu J, Murphy C K. Mechanisms of TonB-catalyzed iron transport through the enteric bacterial cell envelope. J Bioenerg Biomembr. 1993;25:603–611. doi: 10.1007/BF00770247. [DOI] [PubMed] [Google Scholar]
  • 81.Klenk H P, Clayton R A, Tomb J F, White O, Nelson K E, Ketchum K A, Dodson R J, Gwinn M, Hickey E K, Peterson J D, Richardson D L, Kerlavage A R, Graham D E, Kyrpides N C, Fleischmann R D, Quackenbush J, Lee N H, Sutton G G, Gill S, Kirkness E F, Dougherty B A, McKenney K, Adams M D, Loftus B, Venter J C, et al. The complete genome sequence of the hyperthermophilic, sulphate-reducing archaeon Archaeoglobus fulgidus. Nature. 1997;390:364–370. doi: 10.1038/37052. [DOI] [PubMed] [Google Scholar]
  • 82.Komorowska M, Szafran H, Popiela T, Szafran Z. Free amino acids in gastric juice. Acta Physiol Pol. 1981;32:559–567. [PubMed] [Google Scholar]
  • 83.Kostrzynska M, O’Toole P W, Taylor D E, Trust T J. Molecular characterization of a conserved 20-kilodalton membrane-associated lipoprotein antigen of Helicobacter pylori. J Bacteriol. 1994;176:5938–5948. doi: 10.1128/jb.176.19.5938-5948.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kunst F, Ogasawara N, Moszer I, Albertini A, Alloni G, Azevedo V, Bertero M, Bessieres P, Bolotin A, Borchert S, Borriss R, Boursier L, Brans A, Braun M, Brignell S, Bron S, Brouillet S, Bruschi C, Caldwell B, Capuano V, Carter N, Choi S-K, Codani J-J, Connerton I, Cummings N, Daniel R, Denizot F, Devine K, Dusterhoft A, Ehrlich S, Emmerson P, Entian K, Errington J, Fabret C, Ferrari E, Foulger D, Fritz C, Fujita M, Fujita Y, Fuma S, Galizzi A, Galleron N, Ghim S-Y, Glaser P, Goffeau A, Golightly E, Grandi G, Guiseppi G, Guy B, Haga K, Haiech J, Harwood C, Henaut A, Hilbert H, Holsappel S, Hosono S, Hullo M-F, Itaya M, Jones L, Joris B, Karamata D, Kasahara Y, Klaerr-Blanchard M, Klein C, Kobayashi Y, Loetter P, Koningstein G, Krogh S, Kumano M, Kurita K, Lapidus A, Lardinois S, Lauber J, Lazarevic V, Lee S-M, Levine A, Liu H, Masuda S, Mauel C, Medigue C, Median N, Mellado R, Mizuno M, Moestl D, Nakai S, Noback M, Noone D, O’Reilly M, Ogawa K, Ogiwara A, Oudega B, Park S-H, Parro V, Pohl T, Portetelle D, Porwollik S, Prescott A, Presecan E, Pujic P, Purnelle B, et al. The complete genome sequence of the Gram-positive bacterium Bacillus subtilis. Nature. 1997;390:249–256. doi: 10.1038/36786. [DOI] [PubMed] [Google Scholar]
  • 85.Kusukawa N, Yura T, Ueguchi C, Akiyama Y, Ito K. Effects of mutations in heat-shock genes groES and groEL on protein export in Escherichia coli. EMBO J. 1989;8:3517–3521. doi: 10.1002/j.1460-2075.1989.tb08517.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Labigne A, Cussac V, Courcoux P. Shuttle cloning and nucleotide sequences of Helicobacter pylori genes responsible for urease activity. J Bacteriol. 1991;173:1920–1931. doi: 10.1128/jb.173.6.1920-1931.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Labigne A, deReuse H. Determinants of Helicobacter pylori pathogenicity. Infect Agents Dis. 1996;5:191–202. [PubMed] [Google Scholar]
  • 88.Lee C A. Type III secretion systems: machines to deliver bacterial proteins into eukaryotic cells? Trends Microbiol. 1997;5:148–156. doi: 10.1016/S0966-842X(97)01029-9. [DOI] [PubMed] [Google Scholar]
  • 89.Leying H, Suerbaum S, Geis G, Haas R. Cloning and genetic characterization of a Helicobacter pylori flagellin gene. Mol Microbiol. 1992;6:2863–2874. doi: 10.1111/j.1365-2958.1992.tb01466.x. [DOI] [PubMed] [Google Scholar]
  • 90.Luke C, Penn C. Identification of a 29kDa flagellar sheath protein in Helicobacter pylori using a murine monoclonal antibody. Microbiology. 1995;141:597–604. doi: 10.1099/13500872-141-3-597. [DOI] [PubMed] [Google Scholar]
  • 91.Luke C J, Kubiak E, Cockayne A, Elliott T S, Penn C W. Identification of flagellar and associated polypeptides of Helicobacter (formerly Campylobacter) pylori. FEMS Microbiol Lett. 1990;59:225–230. doi: 10.1016/0378-1097(90)90061-t. [DOI] [PubMed] [Google Scholar]
  • 92.Mahdi A A, Lloyd R G. Identification of the recR locus of Escherichia coli K-12 and analysis of its role in recombination and DNA repair. Mol Gen Genet. 1989;216:503–510. doi: 10.1007/BF00334397. [DOI] [PubMed] [Google Scholar]
  • 93.Maier R J, Fu C, Gilbert J, Moshiri F, Olson J, Plaut A G. Hydrogen uptake hydrogenase in Helicobacter pylori. FEMS Microbiol Lett. 1996;141:71–76. doi: 10.1111/j.1574-6968.1996.tb08365.x. [DOI] [PubMed] [Google Scholar]
  • 94.Marcelli S W, Chang H T, Chapman T, Chalk P A, Miles R J, Poole R K. The respiratory chain of Helicobacter pylori: identification of cytochromes and the effects of oxygen on cytochrome and menaquinone levels. FEMS Microbiol Lett. 1996;138:59–64. doi: 10.1111/j.1574-6968.1996.tb08135.x. [DOI] [PubMed] [Google Scholar]
  • 95.Marshall D G, Dundon W G, Beesley S M, Smyth C J. Helicobacter pylori—a conundrum of genetic diversity. Microbiology. 1998;144:2925–2939. doi: 10.1099/00221287-144-11-2925. [DOI] [PubMed] [Google Scholar]
  • 96.Martin S, Edbrooke M, Hodgman T, van der Eijnden D, Bird M. Lewis X biosynthesis in Helicobacter pylori. Molecular cloning of an α(1,3)-fucosyltransferase gene. J Biol Chem. 1997;272:21349–21356. doi: 10.1074/jbc.272.34.21349. [DOI] [PubMed] [Google Scholar]
  • 97.Matin A, Zychlinsky E, Keyhan M, Sachs G. Capacity of Helicobacter pylori to generate ionic gradients at low pH is similar to that of bacteria which grow under strongly acidic conditions. Infect Immun. 1996;64:1434–1436. doi: 10.1128/iai.64.4.1434-1436.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mendz G L, Burns B P, Hazell S L. Characterisation of glucose transport in Helicobacter pylori. Biochim Biophys Acta. 1995;1244:269–276. doi: 10.1016/0304-4165(95)00018-7. [DOI] [PubMed] [Google Scholar]
  • 99.Mendz G L, Hazell S L. Amino acid utilization by Helicobacter pylori. Int J Biochem Cell Biol. 1995;27:1085–1093. doi: 10.1016/1357-2725(95)00069-2. [DOI] [PubMed] [Google Scholar]
  • 100.Mendz G L, Hazell S L. Evidence for a pentose phosphate pathway in Helicobacter pylori. FEMS Lett. 1991;84:331–336. [Google Scholar]
  • 101.Mendz G L, Hazell S L. Fumarate catabolism in Helicobacter pylori. Biochem Mol Biol Int. 1993;31:325–332. [PubMed] [Google Scholar]
  • 102.Mendz G L, Hazell S L. Glucose phosphorylation in Helicobacter pylori. Arch Biochem Biophys. 1993;300:522–525. doi: 10.1006/abbi.1993.1071. [DOI] [PubMed] [Google Scholar]
  • 103.Mendz G L, Hazell S L, Burns B P. The Entner-Doudoroff pathway in Helicobacter pylori. Arch Biochem Biophys. 1994;312:349–356. doi: 10.1006/abbi.1994.1319. [DOI] [PubMed] [Google Scholar]
  • 104.Mendz G L, Hazell S L, Burns B P. Glucose utilization and lactate production by Helicobacter pylori. J Gen Microbiol. 1993;139:3023–3028. doi: 10.1099/00221287-139-12-3023. [DOI] [PubMed] [Google Scholar]
  • 105.Mendz G L, Hazell S L, Srinivasan S. Fumarate reductase: a target for therapeutic intervention against Helicobacter pylori. Arch Biochem Biophys. 1995;321:153–159. doi: 10.1006/abbi.1995.1380. [DOI] [PubMed] [Google Scholar]
  • 106.Mendz G L, Hazell S L, van Gorkom L. Pyruvate metabolism in Helicobacter pylori. Arch Microbiol. 1994;162:187–192. doi: 10.1007/BF00314473. [DOI] [PubMed] [Google Scholar]
  • 107.Mendz G L, Jimenez B M, Hazell S L, Gero A M, O’Sullivan W J. Salvage synthesis of purine nucleotides by Helicobacter pylori. J Appl Bacteriol. 1994;77:674–681. doi: 10.1111/j.1365-2672.1994.tb02818.x. [DOI] [PubMed] [Google Scholar]
  • 108.Mendz G L, Jiminez B M, Hazell S L, Gero A M, O’Sullivan W J. De novo synthesis of pyrimidine nucleotides by Helicobacter pylori. J Appl Bacteriol. 1994;77:1–8. doi: 10.1111/j.1365-2672.1994.tb03036.x. [DOI] [PubMed] [Google Scholar]
  • 109.Miehlke S, Meining A, Morgner A, Bayerdorffer E, Lehn N, Stolte M, Graham D Y, Go M F. Frequency of vacA genotypes and cytotoxin activity in Helicobacter pylori associated with low-grade gastric mucosa-associated lymphoid tissue lymphoma. J Clin Microbiol. 1998;36:2369–2370. doi: 10.1128/jcm.36.8.2369-2370.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mizote T, Yoshiyama H, Nakazawa T. Urease-independent chemotactic responses of Helicobacter pylori to urea, urease inhibitors, and sodium bicarbonate. Infect Immun. 1997;65:1519–1521. doi: 10.1128/iai.65.4.1519-1521.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mobley H L. Defining Helicobacter pylori as a pathogen: strain heterogeneity and virulence. Am J Med. 1996;100:2S–9S. doi: 10.1016/s0002-9343(96)80223-3. [DOI] [PubMed] [Google Scholar]
  • 112.Mobley H L. Helicobacter pylori factors associated with disease development. Gastroenterology. 1997;113:S21–S28. doi: 10.1016/s0016-5085(97)80006-6. [DOI] [PubMed] [Google Scholar]
  • 113.Mobley H L, Island M D, Hausinger R P. Molecular biology of microbial ureases. Microbiol Rev. 1995;59:451–480. doi: 10.1128/mr.59.3.451-480.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mobley H L T, Garner R M, Bauerfeind P. Helicobacter pylori nickel-transport gene nixA: synthesis of catalytically active urease in Escherichia coli independent of growth conditions. Mol Microbiol. 1995;16:97–109. doi: 10.1111/j.1365-2958.1995.tb02395.x. [DOI] [PubMed] [Google Scholar]
  • 115.Moran A P, Lindner B, Walsh E. Structural characterization of the lipid A component of Helicobacter pylori rough- and smooth-form lipopolysaccharides. J Bacteriol. 1997;179:6453–6463. doi: 10.1128/jb.179.20.6453-6463.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nagata K, Tsukita S, Tamura T, Sone N. A cb-type cytochrome-c oxidase terminates the respiratory chain in Helicobacter pylori. Microbiology. 1996;142:1757–1763. doi: 10.1099/13500872-142-7-1757. [DOI] [PubMed] [Google Scholar]
  • 117.Nakao H, Shioda S, Yamamoto S. Purification and properties of carboxynorspermidine decarboxylase, a novel enzyme involved in norspermidine biosynthesis from Vibrio alginolyticus. J Gen Microbiol. 1990;136:1699–1704. [Google Scholar]
  • 118.Nedeskov P. Nutritional requirements for growth of Helicobacter pylori. Appl Environ Microbiol. 1994;60:3450–3453. doi: 10.1128/aem.60.9.3450-3453.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Neidhardt F C, Curtiss III R, Ingraham J L, Lin E C C, Low K B, Magasanik B, Reznikoff W S, Riley M, Schaechter M, Umbarger H E, editors. Escherichia coli and Salmonella: cellular and molecular biology. 2nd ed. Washington, D.C: ASM Press; 1996. [Google Scholar]
  • 120.Newell D. Identification of the outer membrane proteins of Campylobacter pyloridis and antigenic cross-reactivity between C. pyloridis and C. jejuni. J Gen Microbiol. 1987;133:163–170. doi: 10.1099/00221287-133-1-163. [DOI] [PubMed] [Google Scholar]
  • 121.Nishiyama K, Mizushima S, Tokuda H. A novel membrane protein involved in protein translocation across the cytoplasmic membrane of Escherichia coli. EMBO J. 1993;12:3409–3415. doi: 10.1002/j.1460-2075.1993.tb06015.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Odenbreit S, Wieland B, Haas R. Cloning and genetic characterization of Helicobacter pylori catalase and construction of a catalase-deficient mutant strain. J Bacteriol. 1996;178:6960–6967. doi: 10.1128/jb.178.23.6960-6967.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Ohta-Tada U, Takagi A, Koga Y, Kamiya S, Miwa T. Flagellin gene diversity among Helicobacter pylori strains and IL-8 secretion from gastric epithelial cells. Scand J Gastroenterol. 1997;32:455–459. doi: 10.3109/00365529709025080. [DOI] [PubMed] [Google Scholar]
  • 124.O’Toole P W, Kostrzynska M, Trust T J. Non-motile mutants of Helicobacter pylori and Helicobacter mustelae defective in flagellar hook production. Mol Microbiol. 1994;14:691–703. doi: 10.1111/j.1365-2958.1994.tb01307.x. [DOI] [PubMed] [Google Scholar]
  • 125.Page M D, Tomlinson E, Ferguson S J. Unexpected implications from the Helicobacter pylori genome for understanding periplasmic c-type cytochrome assembly in gram-negative bacteria in coexistence with disulphide bond formation. Mol Microbiol. 1997;26:413–415. doi: 10.1046/j.1365-2958.1997.6061962.x. [DOI] [PubMed] [Google Scholar]
  • 126.Pahel G, Zelenetz A D, Tyler B M. gltB gene and regulation of nitrogen metabolism by glutamine synthetase in Escherichia coli. J Bacteriol. 1978;133:139–148. doi: 10.1128/jb.133.1.139-148.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Park J T. Why does Escherichia coli recycle its cell wall peptides? Mol Microbiol. 1995;17:421–426. doi: 10.1111/j.1365-2958.1995.mmi_17030421.x. [DOI] [PubMed] [Google Scholar]
  • 128.Penn C W, Luke C J. Bacterial flagellar diversity and significance in pathogenesis. FEMS Microbiol Lett. 1992;79:331–336. doi: 10.1111/j.1574-6968.1992.tb14060.x. [DOI] [PubMed] [Google Scholar]
  • 129.Phadnis S H, Parlow M H, Levy M, Ilver D, Caulkins C M, Connors J B, Dunn B E. Surface localization of Helicobacter pylori urease and a heat shock protein homolog requires bacterial autolysis. Infect Immun. 1996;64:905–912. doi: 10.1128/iai.64.3.905-912.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Preisig O, Anthamatten D, Hennecke H. Genes for a microaerobically induced oxidase complex in Bradyrhizobium japonicum are essential for a nitrogen-fixing endosymbiosis. Proc Nat Acad Sci USA. 1993;90:3309–3013. doi: 10.1073/pnas.90.8.3309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Reitzer L. Ammonia assimilation and the biosynthesis of glutamine, glutamate, aspartate, asparagine, l-alanine, and d-alanine. In: Neidhardt F C, Curtiss III R, Ingraham J L, Lin E C C, Low K B, Magasanik B, Reznikoff W S, Riley M, Schaechter M, Umbarger H E, editors. Escherichia coli and Salmonella: cellular and molecular biology. Vol. 1. Washington, D.C: ASM Press; 1996. pp. 391–407. [Google Scholar]
  • 132.Reynolds D J, Penn C W. Characteristics of Helicobacter pylori growth in a defined medium and determination of its amino acid requirements. Microbiology. 1994;140:2649–2656. doi: 10.1099/00221287-140-10-2649. [DOI] [PubMed] [Google Scholar]
  • 133.Roine R P, Salmela K S, Salaspuro M. Alcohol metabolism in Helicobacter pylori-infected stomach. Ann Med. 1995;27:583–588. doi: 10.3109/07853899509002473. [DOI] [PubMed] [Google Scholar]
  • 134.Rosario M M, Fredrick K L, Ordal G W, Helmann J D. Chemotaxis in Bacillus subtilis requires either of two functionally redundant CheW homologs. J Bacteriol. 1994;176:2736–2739. doi: 10.1128/jb.176.9.2736-2739.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Salmela K S, Roine R P, Hook-Nikanne J, Kosunen T U, Salaspuro M. Acetaldehyde and ethanol production by Helicobacter pylori. Scand J Gastroenterol. 1994;29:309–312. doi: 10.3109/00365529409094841. [DOI] [PubMed] [Google Scholar]
  • 136.Salmela K S, Roine R P, Koivisto T, Hook-Nikanne J, Kosunen T U, Salaspuro M. Characteristics of Helicobacter pylori alcohol dehydrogenase. Gastroenterology. 1993;105:325–330. doi: 10.1016/0016-5085(93)90704-g. [DOI] [PubMed] [Google Scholar]
  • 137.Segal E D, Lange C, Covacci A, Tompkins L S, Falkow S. Induction of host signal transduction pathways by Helicobacter pylori. Proc Natl Acad Sci USA. 1997;94:7595–7599. doi: 10.1073/pnas.94.14.7595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Seifert J, Kunz N, Flachmann R, Laufer A, Jany K D, Gassen H G. Expression of the E. coli nadB gene and characterization of the gene product l-aspartate oxidase. Biol Chem Hoppe-Seyler. 1990;371:239–248. [PubMed] [Google Scholar]
  • 139.Smith D, Douchette-Stamm L, Deloughery C, Lee H, Dubois J, Aldredge T, Bashirzadeh R, Blakely D, Cook R, Gilbert K, Harrison D, Hoang L, Keagle P, Lumm W, Pothier B, Qiu D, Spadafors R, Vicaire R, Wang Y, Wierzbowski J, Gibson R, Jiwani N, Caruso A, Bush D, Safer H, Patwell D, Prabhakar S, McDougall S, Shimer G, Goyal A, Pietrokovski S, Church G, Daniels C, Mao J-I, Rice P, Nolling J, Reeve J. Complete genome sequence of Methanobacterium thermoautotrophicum deltaH: functional analysis and comparative genomics. J Bacteriol. 1997;179:7135–7155. doi: 10.1128/jb.179.22.7135-7155.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Spiegelhalder C, Gerstenecker B, Kersten A, Schiltz E, Kist M. Purification of Helicobacter pylori superoxide dismutase and cloning and sequencing of the gene. Infect Immun. 1993;61:5315–5325. doi: 10.1128/iai.61.12.5315-5325.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Spohn G, Beier D, Rappuoli R, Scarlato V. Transcriptional analysis of the divergent cagAB genes encoded by the pathogenecity island of Helicobacter pylori. Mol Microbiol. 1997;26:361–372. doi: 10.1046/j.1365-2958.1997.5831949.x. [DOI] [PubMed] [Google Scholar]
  • 142.Spratt B G. Properties of the penicillin-binding proteins of Escherichia coli K12. Eur J Biochem. 1977;72:341–352. doi: 10.1111/j.1432-1033.1977.tb11258.x. [DOI] [PubMed] [Google Scholar]
  • 143.Stark R M, Suleiman M S, Hassan I J, Greenman J, Millar M R. Amino acid utilisation and deamination of glutamine and asparagine by Helicobacter pylori. J Med Microbiol. 1997;46:793–800. doi: 10.1099/00222615-46-9-793. [DOI] [PubMed] [Google Scholar]
  • 144.Stephens J C, Stewart J A, Folwell A M, Rathbone B J. Helicobacter pylori cagA status, vacA genotypes and ulcer disease. Eur J Gastroenterol Hepatol. 1998;10:381–384. doi: 10.1097/00042737-199805000-00005. [DOI] [PubMed] [Google Scholar]
  • 145.Stock A, Schaeffer E, Koshland D E, Stock J. A second type of protein methylation reaction in bacterial chemotaxis. J Biol Chem. 1987;262:8011–8014. [PubMed] [Google Scholar]
  • 146.Strauch M A, Zalkin H, Aronson A I. Characterization of the glutamyl-tRNA(Gln)-to-glutaminyl-tRNA(Gln) amidotransferase reaction of Bacillus subtilis. J Bacteriol. 1988;170:916–920. doi: 10.1128/jb.170.2.916-920.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Suerbaum S, Josenhans C, Labigne A. Cloning and genetic characterization of the Helicobacter pylori and Helicobacter mustellae flaB genes and construction of H. pylori flaA- and flaB-negative mutants by allelic exchange. J Bacteriol. 1993;175:3278–3288. doi: 10.1128/jb.175.11.3278-3288.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Taylor D, Eaton M, Chang N, Salama S. Construction of a Helicobacter pylori genome map and demonstration of diversity at the genome level. J Bacteriol. 1992;174:6800–6806. doi: 10.1128/jb.174.21.6800-6806.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Tomb J-F, White O, Kerlavage A, Clayton R, Sutton G, Fleischmann R, Ketchum K, Klenk H, Gill S, Dougherty B, Nelson K, Quackenbush J, Zhou L, Kirkness E, Peterson S, Loftus B, Richardson D, Dodson R, Khalak H, Glodek A, McKenney K, Fitzegerald L, Lee N, Adams M, Hickey E, Berg D, Gocayne J, Utterback T, Peterson J, Kelley J, Cotton M, Weidman J, Fujii C, Bowman C, Watthey L, Wallin E, Hayes W, Borodovsky M, Karp P, Smith H, Fraser C, Venter J. The complete genome sequence of the gastric pathogen Helicobacter pylori. Nature. 1997;388:539–547. doi: 10.1038/41483. [DOI] [PubMed] [Google Scholar]
  • 150.Tsuda M, Karita M, Morshed M, Okita K, Nakazawa T. A urease-negative mutant of Helicobacter pylori constructed by allelic exchange mutagenesis lacks the ability to colonize the nude mouse stomach. Infect Immun. 1994;62:3586–3589. doi: 10.1128/iai.62.8.3586-3589.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.van Doorn L J, Figueiredo C, Sanna R, Pena S, Midolo P, Ng E K, Atherton J C, Blaser M J, Quint W G. Expanding allelic diversity of Helicobacter pylori vacA. J Clin Microbiol. 1998;36:2597–2603. doi: 10.1128/jcm.36.9.2597-2603.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.van Doorn L J, Figueiredo C, Sanna R, Plaisier A, Schneeberger P, de Boer W, Quint W. Clinical relevance of the cagA, vacA, and iceA status of Helicobacter pylori. Gastroenterology. 1998;115:58–66. doi: 10.1016/s0016-5085(98)70365-8. [DOI] [PubMed] [Google Scholar]
  • 153.Vanet A, Labigne A. Evidence for specific secretion rather than autolysis in the release of some Helicobacter pylori proteins. Infect Immun. 1998;66:1023–1027. doi: 10.1128/iai.66.3.1023-1027.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Vincente M, Gomez M J, Agata J A. Regulation of transcription of cell division genes in the Escherichia coli dcw cluster. Cell Mol Life Sci. 1990;59:317–324. doi: 10.1007/s000180050158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Weiss A A, Johnson F D, Burns D L. Molecular characterization of an operon required for pertussis toxin secretion. Proc Natl Acad Sci USA. 1993;90:2970–2974. doi: 10.1073/pnas.90.7.2970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Westblom T U, Phadnis S, Langenberg W, Yoneda K, Madan E, Midkiff B R. Catalase negative mutants of Helicobacter pylori. Eur J Clin Microbiol Infect Dis. 1992;11:522–526. doi: 10.1007/BF01960807. [DOI] [PubMed] [Google Scholar]
  • 157.Williams C L, Preston T, Hossack M, Slater C, McColl K E. Helicobacter pylori utilises urea for amino acid synthesis. FEMS Immunol Med Microbiol. 1996;13:87–94. doi: 10.1111/j.1574-695X.1996.tb00220.x. [DOI] [PubMed] [Google Scholar]
  • 158.Worst D J, Gerrits M M, Vandenbroucke-Grauls C M, Kusters J G. Helicobacter pylori ribBA-mediated riboflavin production is involved in iron acquisition. J Bacteriol. 1998;180:1473–1479. doi: 10.1128/jb.180.6.1473-1479.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Xu Q, Peek R M J, Miller G G, Blaser M J. The Helicobacter pylori genome is modified at CATG by the product of hpyIM. J Bacteriol. 1997;179:6807–6815. doi: 10.1128/jb.179.21.6807-6815.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Zakharova N, Hoffman P S, Berg D E, Severinov K. The largest subunits of RNA polymerase from gastric helicobacters are tethered. J Biol Chem. 1998;273:19371–19374. doi: 10.1074/jbc.273.31.19371. [DOI] [PubMed] [Google Scholar]

Articles from Microbiology and Molecular Biology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES