首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 78 毫秒
1.
Peptidoglycan from Deinococcus radiodurans was analyzed by high-performance liquid chromatography and mass spectrometry. The monomeric subunit was: N-acetylglucosamine–N-acetylmuramic acid–l-Ala–d-Glu-(γ)–l-Orn-[(δ)Gly-Gly]–d-Ala–d-Ala. Cross-linkage was mediated by (Gly)2 bridges, and glycan strands were terminated in (1→6)anhydro-muramic acid residues. Structural relations with the phylogenetically close Thermus thermophilus are discussed.The gram-positive bacterium Deinococcus radiodurans is remarkable because of its extreme resistance to ionizing radiation (14). Phylogenetically the closest relatives of Deinococcus are the extreme thermophiles of the genus Thermus (4, 11). In 16S rRNA phylogenetic trees, the genera Thermus and Deinococcus group together as one of the older branches in bacterial evolution (11). Both microorganisms have complex cell envelopes with outer membranes, S-layers, and ornithine-Gly-containing mureins (7, 12, 19, 20, 22, 23). However, Deinococcus and Thermus differ in their response to the Gram reaction, having positive and negative reactions, respectively (4, 14). The murein structure for Thermus thermophilus HB8 has been recently elucidated (19). Here we report the murein structure of Deinococcus radiodurans with similar detail.D. radiodurans Sark (23) was used in the present study. Cultures were grown in Luria-Bertani medium (13) at 30°C with aeration. Murein was purified and subjected to amino acid and high-performance liquid chromatography (HPLC) analyses as previously described (6, 9, 10, 19). For further analysis muropeptides were purified, lyophilized, and desalted as reported elsewhere (6, 19). Purified muropeptides were subjected to plasma desorption linear time-of-flight mass spectrometry (PDMS) as described previously (1, 5, 16, 19). Positive and negative ion mass spectra were obtained on a short linear 252californium time-of-flight instrument (BioIon AB, Uppsala, Sweden). The acceleration voltage was between 17 and 19 kV, and spectra were accumulated for 1 to 10 million fission events. Calibration of the mass spectra was done in the positive ion mode with H+ and Na+ ions and in the negative ion mode with H and CN ions. Calculated m/z values are based on average masses.Amino acid analysis of muramidase (Cellosyl; Hoechst, Frankfurt am Main, Germany)-digested sacculi (50 μg) revealed Glu, Orn, Ala, and Gly as the only amino acids in the muramidase-solubilized material. Less than 3% of the total Orn remained in the muramidase-insoluble fraction, indicating an essentially complete solubilization of murein.Muramidase-digested murein samples (200 μg) were analyzed by HPLC as described in reference 19. The muropeptide pattern (Fig. (Fig.1)1) was relatively simple, with five dominating components (DR5 and DR10 to DR13 [Fig. 1]). The muropeptides resolved by HPLC were collected, desalted, and subjected to PDMS. The results are presented in Table Table11 compared with the m/z values calculated for best-matching muropeptides made up of N-acetylglucosamine (GlucNAc), N-acetylmuramic acid (MurNAc), and the amino acids detected in the murein. The more likely structures are shown in Fig. Fig.1.1. According to the m/z values, muropeptides DR1 to DR7 and DR9 were monomers; DR8, DR10, and DR11 were dimers; and DR12 and DR13 were trimers. The best-fitting structures for DR3 to DR8, DR11, and DR13 coincided with muropeptides previously characterized in T. thermophilus HB8 (19) and had identical retention times in comparative HPLC runs. The minor muropeptide DR7 (Fig. (Fig.1)1) was the only one detected with a d-Ala–d-Ala dipeptide and most likely represents the basic monomeric subunit. The composition of the major cross-linked species DR11 and DR13 confirmed that cross-linking is mediated by (Gly)2 bridges, as proposed previously (20). Open in a separate windowFIG. 1HPLC muropeptide elution patterns of murein purified from D. radiodurans. Muramidase-digested murein samples were subjected to HPLC analysis, and the A204 of the eluate was recorded. The most likely structures for each muroeptide as deduced by PDMS are shown. The position of residues in brackets is the most likely one as deduced from the structures of other muropeptides but could not be formally demonstrated. R = GlucNac–MurNac–l-Ala–d-Glu-(γ)→.

TABLE 1

Calculated and measured m/z values for the molecular ions of the major muropeptides from D. radiodurans
MuropeptideaIonm/z
ΔmbError (%)cMuropeptide composition
Muropeptide abundance (mol%)
CalculatedMeasuredNAGdNAMeGluOrnAlaGly
DR1[M+H]+699.69700.10.410.0611101012.0
DR2[M+H]+927.94928.30.360.041111125.7
DR3[M+Na]+1,006.971,007.50.530.051111133.0
DR4[M+Na]+963.95964.60.650.071111212.5
DR5[M+H]+999.02999.80.780.0811112227.7
[M−H]997.00997.30.300.03
DR6[M+Na]+1,078.51,078.80.750.071111232.4
DR7[M+H]+1,070.091,071.00.900.081111322.2
DR8[M+Na]+1,520.531,521.61.080.071122442.2
DR9[M+Na]+701.64702.10.460.0311f10105.0
DR10[M+H]+1,907.941,907.80.140.0122223410.1
[M−H]1,905.921,906.60.680.04
DR11[M+H]+1,979.011,979.10.090.0122224419.1
[M−H]1,977.001,977.30.300.02
DR12[M+H]+2,887.932,886.5−1.43−0.053333564.4
[M−H]2,885.912,885.8−0.11−0.01
DR13[M+H]+2,959.002,957.8−1.20−0.043333663.6
[M−H]2,956.992,955.9−1.09−0.04
Open in a separate windowaDR5 and DR10 to DR13 were analyzed in both the positive and negative ion modes. Muropeptides DR1 to DR4 and DR6 to DR9 were analyzed in the positive mode only due to the small amounts of sample available. bMass difference between measured and calculated quasimolecular ion values. c[(Measured mass−calculated mass)/calculated mass] × 100. dN-Acetylglucosamine. eN-Acetylmuramitol. f(1→6)Anhydro-N-acetylmuramic acid. Structural assignments of muropeptides DR1, DR2, DR8 to DR10, and DR12 deserve special comments. The low m/z value measured for DR1 (700.1) fitted very well with the value calculated for GlucNAc–MurNAc–l-Ala–d-Glu (699.69). Even smaller was the mass deduced for DR9 from the m/z value of the molecular ion of the sodium adduct (702.1) (Fig. (Fig.2).2). The mass difference between DR1 and DR9 (19.9 mass units) was very close indeed to the calculated difference between N-acetylmuramitol and the (1→6)anhydro form of MurNAc (20.04 mass units). Therefore, DR9 was identified as GlucNAc–(1→6)anhydro-MurNAc–l-Ala–d-Glu (Fig. (Fig.1).1). Muropeptides with (1→6)anhydro muramic acid have been identified in mureins from diverse origins (10, 15, 17, 19), indicating that it might be a common feature among peptidoglycan-containing microorganisms. Open in a separate windowFIG. 2Positive-ion linear PDMS of muropeptide DR9. Muropeptide DR9 was purified, desalted by HPLC, and subjected to PDMS to determine the molecular mass. The masses for the dominant molecular ions are indicated.The measured m/z value for the [M+Na]+ ion of DR8 was 1,521.6, very close to the mass calculated for a cross-linked dimer without one disaccharide moiety (1,520.53) (Fig. (Fig.1;1; Table Table1).1). Such muropeptides, also identified in T. thermophilus HB8 and other bacteria (18, 19), are most likely generated by the enzymatic clevage of MurNAc–l-Ala amide bonds in murein by an N-acetylmuramyl–l-alanine amidase (21). In particular, DR8 could derive from DR11. The difference between measured m/z values for DR8 and DR11 was 478.7, which fits with the mass contribution of a disaccharide moiety (480.5) within the mass accuracy of the instrument.The m/z values for muropeptides DR2, DR10, and DR12 supported the argument for structures in which the two d-Ala residues from the d-Ala–d-Ala C-terminal dipeptide were lost, leaving Orn as the C-terminal amino acid.The position of one Gly residue in muropeptides DR2, DR8, and DR10 to DR13 could not be formally demonstrated. One of the Gly residues could be at either the N- or the C-terminal positions. However, the N-terminal position seems more likely. The structure of the basic muropeptide (DR7), with a (Gly)2 acylating the δ-NH2 group of Orn, suggests that major muropeptides should present a (Gly)2 dipeptide. The scarcity of DR3 and DR6, which unambiguously have Gly as the C-terminal amino acid (Fig. (Fig.1),1), supports our assumption.Molar proportions for each muropeptide were calculated as proposed by Glauner et al. (10) and are shown in Table Table1.1. For calculations the structures of DR10 to DR13 were assumed to be those shown in Fig. Fig.1.1. The degree of cross-linkage calculated was 47.2%. Trimeric muropeptides were rather abundant (8 mol%) and made a substantial contribution to total cross-linkage. However, higher-order oligomers were not detected, in contrast with other gram-positive bacteria, such as Staphylococcus aureus, which is rich in such oligomers (8). The proportion of muropeptides with (1→6)anhydro-muramic acid (5 mol%) corresponded to a mean glycan strand length of 20 disaccharide units, which is in the range of values published for other bacteria (10, 17).The results of our study indicate that mureins from D. radiodurans and T. thermophilus HB8 (19) are certainly related in their basic structures but have distinct muropeptide compositions. In accordance with the phylogenetic proximity of Thermus and Deinococcus (11), both mureins are built up from the same basic monomeric subunit (DR7 in Fig. Fig.1),1), are cross-linked by (Gly)2 bridges, and have (1→6)anhydro-muramic acid at the termini of glycan strands. Most interestingly, Deinococcus and Thermus are the only microorganisms identified at present with the murein chemotype A3β as defined by Schleifer and Kandler (20). Nevertheless, the differences in muropeptide composition were substantial. Murein from D. radiodurans was poor in d-Ala–d-Ala- and d-Ala–Gly-terminated muropeptides (2.2 and 2.4 mol%, respectively) but abundant in Orn-terminated muropeptides (23.8 mol%) and in muropeptides with a peptide chain reduced to the dipeptide l-Ala–d-Glu (18 mol%). In contrast, neither Orn- nor Glu-terminated muropeptides have been detected in T. thermophilus HB8 murein, which is highly enriched in muropeptides with d-Ala–d-Ala and d-Ala–Gly (19). Furthermore, no traces of phenyl acetate-containing muropeptides, a landmark for T. thermophilus HB8 murein (19), were found in D. radiodurans. Cross-linkage was definitely higher in D. radiodurans than in T. thermophilus HB8 (47.4 and 27%, respectively), largely due to the higher proportion of trimers in the former.The similarity in murein basic structure suggests that the difference between D. radiodurans and T. thermophilus HB8 with respect to the Gram reaction may simply be a consequence of the difference in the thickness of cell walls (2, 3, 23). Interestingly, D. radiodurans murein turned out to be relatively simple for a gram-positive organism, possibly reflecting the primitive nature of this genus as deduced from phylogenetic trees (11). Our results illustrate the phylogenetic proximity between Deinococcus and Thermus at the cell wall level but also point out the structural divergences originated by the evolutionary history of each genus.  相似文献   

2.
Vancomycin-resistant enterococci acquire high-level resistance to glycopeptide antibiotics through the synthesis of peptidoglycan terminating in d-alanyl-d-lactate. A key enzyme in this process is a d-alanyl-d-alanine ligase homologue, VanA or VanB, which preferentially catalyzes the synthesis of the depsipeptide d-alanyl-d-lactate. We report the overexpression, purification, and enzymatic characterization of DdlN, a VanA and VanB homologue encoded by a gene of the vancomycin-producing organism Amycolatopsis orientalis C329.2. Evaluation of kinetic parameters for the synthesis of peptides and depsipeptides revealed a close relationship between VanA and DdlN in that depsipeptide formation was kinetically preferred at physiologic pH; however, the DdlN enzyme demonstrated a narrower substrate specificity and commensurately increased affinity for d-lactate in the C-terminal position over VanA. The results of these functional experiments also reinforce the results of previous studies that demonstrated that glycopeptide resistance enzymes from glycopeptide-producing bacteria are potential sources of resistance enzymes in clinically relevant bacteria.The origin of antibiotic resistance determinants is of significant interest for several reasons, including the prediction of the emergence and spread of resistance patterns, the design of new antimicrobial agents, and the identification of potential reservoirs for resistance elements. Antibiotic resistance can occur either through spontaneous mutation in the target or by the acquisition of external genetic elements such as plasmids or transposons which carry resistance genes (7). The origins of these acquired genes are varied, but it has long been recognized that potential reservoirs are antibiotic-producing organisms which naturally harbor antibiotic resistance genes to protect themselves from the actions of toxic compounds (6).High-level resistance to glycopeptide antibiotics such as vancomycin and teicoplanin in vancomycin-resistant enterococci (VRE) is conferred by the presence of three genes, vanH, vanA (or vanB), and vanX, which, along with auxiliary genes necessary for inducible gene expression, are found on transposons integrated into plasmids or the bacterial genome (1, 20). These three genes are essential to resistance and serve to change the C-terminal peptide portion of the peptidoglycan layer from d-alanyl-d-alanine (d-Ala-d-Ala) to d-alanyl-d-lactate (d-Ala-d-Lac). This change results in the loss of a critical hydrogen bond between vancomycin and the d-Ala-d-Ala terminus and in a 1,000-fold decrease in binding affinity between the antibiotic and the peptidoglycan layer, which is the basis for the bactericidal action of this class of compounds (5). The vanH gene encodes a d-lactate dehydrogenase which provides the requisite d-Lac (3, 5), while the vanX gene encodes a highly specific dd-peptidase which cleaves only d-Ala-d-Ala produced endogenously while leaving d-Ala-d-Lac intact (19, 21). The final gene, vanA or vanB, encodes an ATP-dependent d-Ala-d-Lac ligase (4, 8, 10). This enzyme has sequence homology with the chromosomal d-Ala-d-Ala ligases, which are essential for peptidoglycan synthesis but which generally lack the ability to synthesize d-Ala-d-Lac (9).We have recently cloned vanH, vanA, and vanX homologues from two glycopeptide antibiotic-synthesizing organisms: Amycolatopsis orientalis C329.2, which produces vancomycin, and Streptomyces toyocaensis NRRL 15009, which produces A47934 (14). In addition, the vanH-vanA-vanX gene cluster was identified in several other glycopeptide producers. We have also demonstrated that the VanA homologue from S. toyocaensis NRRL 15009 can synthesize d-Ala-d-Lac in vitro and in the glycopeptide-sensitive host Streptomyces lividans (15, 16). We now report the expression of the A. orientalis C329.2 VanA homologue DdlN in Escherichia coli, its purification, and its enzymatic characterization. These data reinforce the striking similarity between vancomycin resistance elements in VRE and glycopeptide-producing organisms and support the possibility of a common origin for these enzymes.

Expression, purification, and specificity of DdlN.

DdlN was overexpressed in E. coli under the control of the bacteriophage T7 promoter. The construct gave good yields of highly purified enzyme following a four-step purification procedure (Table (Table1;1; Fig. Fig.1).1). Like other dd-ligases, DdlN behaved like a dimer in solution (not shown).

TABLE 1

Purification of DdlN from E. coli BL21 (DE3)/pETDdlN
SampleProtein (mg)Activity (nmol/min)Sp act (nmol/ min/mg)Recovery (%)Purification (fold)
Lysate1248436.82100
Ammonium sulfate (20–50% saturation)67.678011.5921.7
Sephacryl S20011.682571.49811
Q Sepharose2.87422658839
Phenyl Superose0.429974835110
Open in a separate windowOpen in a separate windowFIG. 1Purification of DdlN from E. coli BL21 (DE3)/pETDdlN. Proteins were separated on an SDS–11% polyacrylamide gel and stained with Coomassie blue. Lane 1, molecular mass markers (masses are noted at the left in kilodaltons); lane 2, whole-cell lysate; lane 3, ammonium sulfate fraction (20 to 50% saturation); lane 4, Sephacryl S200; lane 5, Q Sepharose; lane 6, phenyl Superose.The amino acid substrate specificity of DdlN was assessed by incubation of 14C-d-Ala with all 20 common amino acids in the d configuration. Purified DdlN catalyzed the synthesis of d-Ala-d-Ala in addition to that of several other mixed dipeptides, including d-Ala-d-Met and d-Ala-d-Phe (Fig. (Fig.2).2). Thus, DdlN exhibits a substrate specificity which is similar to that of VanA (4), with the capacity to synthesize not only d-Ala-d-Ala but also mixed dipeptides with bulky side chains in the C-terminal position.Open in a separate windowFIG. 2Substrate specificity of DdlN. Autoradiogram from thin-layer chromatography analysis of DdlN substrate specificity. All reaction mixtures contained 2.5 mM d-Ala and 1 mM ATP, and the radiolabel was 14C-d-Ala, except where noted. Lane 1, d-Ala; lane 2, d-Lac with 14C-d-Lac label; lane 3, d,l-methionine; lane 4, dl-phenylalanine; lane 5, d-Hbut; lane 6, d-hydroxyvalerate. Letters indicate the following: A, d-Ala-d-Lac; B, d-Lac; C, d-Ala-d-Met; D, d-Ala-d-Phe; E, d-Ala-d-Hbut; F, d-Ala-d-hydroxyvalerate.Importantly, DdlN is a depsipeptide synthase with the ability to synthesize d-Ala-d-Lac, d-Ala-d-hydroxybutyrate (Hbut), and d-Ala-d-hydroxyvalerate (Fig. (Fig.2).2). However, unlike VanA (5), d-hydroxycaproate and d-phenyllactate are not substrates (not shown). Thus, DdlN is a broad-spectrum d-Ala-d-X ligase with depsipeptide synthase activity.

Characterization of d-Ala-d-X ligase activity.

Following the initial assessment of the specificity of the enzyme, several substrates were selected for quantitative analysis by evaluation of their steady-state kinetic parameters (Table (Table2).2). DdlN has two amino acid (or hydroxy acid) Km values. Steady-state kinetic plots indicated that, like other dd-ligases, the N-terminal Km (Km1) was significantly lower (higher specificity) than the C-terminal Km (Km2). Since the former value is expected to be independent of the C-terminal substrate, only Km2 values were determined and are reported here.

TABLE 2

Characterization of steady-state parameters of DdlN and VanA
LigaseSubstrateKm2 (mM)kcat (min−1)kcat/Km2 (M−1 s−1)
DdlNd-Ala21 ± 2229 ± 71.8 × 102
d-Lac0.4 ± 0.0555 ± 12.3 × 103
d-Hbut2.5 ± 0.332 ± 22.1 × 102
ATPa1.2 ± 0.271 ± 50.98 × 102
DdlMbd-Ala166 ± 27
d-Lac1.08 ± 0.10
VanAcd-Ala382951.3 × 102
d-Lac7.1942.2 × 102
d-Hbut0.601083.0 × 103
Open in a separate windowa Determined in the presence of 10 mM d-Lac. b Data from reference 16c Data from reference 5. DdlN showed good d-Ala-d-Ala ligase activity but with a very high and physiologically questionable Km2 (21 mM). On the other hand, d-Ala-d-Lac synthesis was excellent, with a 4-fold decrease in kcat, compared to d-Ala-d-Ala synthesis, which was offset by a 52-fold drop in Km that resulted in a >12-fold increase in specificity (kcat/Km2). d-Hbut was also a good substrate, with a kcat/Km2 comparable to that of d-Ala.Steady-state kinetic parameters for d-Ala-d-X formation showed trends similar to those found with both VanA and DdlN. For example, the kcat values between VanA and DdlN were virtually the same for most substrates. There were significant differences, however. For instance, while the Km2 values for d-Ala were very high for all three enzymes, DdlN does have greater affinity for d-Ala, with a 1.8- and 7.9-fold lower Km2 than those of VanA and DdlM, respectively. Additionally, the Km2 for d-Lac was 17.8- and 2.7-fold lower than those for VanA and DdlM. Thus, DdlN has a more restrictive specificity for the C-terminal residue than VanA, which is compensated for by a higher affinity for the critical substrate d-Lac.

pH dependence of peptide versus that of depsipeptide synthesis activity.

The partitioning of the syntheses of d-Ala-d-Ala and d-Ala-d-Hbut in VanA and other depsipeptide-competent dd-ligases has been shown to be pH dependent (17). Determination of the pH dependence of DdlN in synthesizing peptide versus depsipeptide (Fig. (Fig.3)3) directly paralleled the results obtained with VanA in similar experiments. At lower pHs (<7), d-Ala-d-Hbut synthesis predominates and is exclusive at a pH of <6 (Fig. (Fig.3).3). At pH 7.5, levels of synthesis of d-Ala-d-Hbut and d-Ala-d-Ala are relatively equal, while at a pH greater than 8, the capacity to synthesize peptide overtakes the capacity to synthesize depsipeptide, although the latter is never abolished. Open in a separate windowFIG. 3pH dependence of partitioning of the syntheses of peptide and depsipeptide by DdlN. (A) Autoradiogram of a thin-layer chromatography separation of the products of reaction mixtures containing 14C-D-Ala, unlabeled D-Ala, and d-Hbut. (B) Quantification of reaction products following phosphorimage analysis. Filled circles, D-Ala-d-Hbut; open circles, D-Ala-D-Ala.The partitioning of the formation of peptide versus depsipeptide as a function of pH by DdlM is comparable to that by VanA and depsipeptide-competent mutants of DdlB (17), which show essentially exclusively depsipeptide formation at lower pHs and increasing peptide formation as the pH increases. This implies a potential role for the protonated ammonium group of d-Ala2 in second-substrate recognition and suggests a mechanism for the discrimination between d-Ala and d-Lac at physiologic pH. The structural basis for this distinction remains obscure for DdlB and VanA or DdlN.

Concluding remarks.

Resistance to vancomycin and other glycopeptides is mediated through the synthesis of a peptidoglycan which does not terminate with the canonical d-Ala-d-Ala dipeptide. Thus, enterococci which exhibit the VanC phenotype, which consists of low-level, noninducible resistance to vancomycin only, have peptidoglycan terminating in d-Ala-d-Ser (19). On the other hand, bacteria which are constitutively resistant to high concentrations of glycopeptides, such as lactic acid bacteria and VRE exhibiting the VanA or VanB phenotype (high-level inducible resistance to vancomycin), incorporate the depsipeptide d-Ala-d-Lac into their cell walls (2, 12, 13). The enzymes responsible for the intracellular synthesis of d-Ala-d-Lac not surprisingly have significant amino acid sequence similarity with d-Ala-d-Ala ligases, which are responsible for d-Ala-d-Ala synthesis in all bacteria with a cell wall (9).The d-Ala-d-Lac synthases can be subdivided into two groups based on sequence homology: those found in the constitutively resistant lactic acid bacteria and those found in glycopeptide-producing organisms and VanA or VanB VRE (9, 14). The former have more similarity with exclusive d-Ala-d-Ala ligases. Indeed, single point mutations in d-Ala-d-Ala ligases which yield sequences more similar to those of lactic acid bacterium d-Ala-d-Lac ligases are sufficient to induce significant depsipeptide synthase activity in these enzymes (17). Similarly, mutational studies of the d-Ala-d-Lac ligase from Leuconostoc mesenteroides have demonstrated that the converse also holds (18). On the other hand, the molecular basis for depsipeptide synthesis by the VanA or VanB ligases is unknown, in large part due to the lack of protein structural information on which to base mutational studies, unlike the situation with d-Ala-d-Ala ligases, where the E. coli DdlB structure serves as a template for mechanistic research (11).Significantly, a major difference in the VanA or VanB ligases and other dd-ligases lies in the amino acid sequence of the ω-loop region, which closes off the active site of DdlB (11) and has been shown to contribute amino acid residues with the capacity to control the syntheses of d-Ala-d-Ala and d-Ala-d-Lac, notably, Tyr216 (17, 18). Until recently, the VanA and VanB ligases were exceptional in amino acid structure and had no known homologues. The sequencing of resistance genes from glycopeptide-producing bacteria has uncovered enzymes with >60% homology to VanA or VanB and which are virtually superimposable in the critical ω-loop region (14, 15). One of these, DdlM from S. toyocaensis NRRL 15009, has been shown to have d-Ala-d-Lac ligase ability (15, 16), although no rigorous analysis of this activity has been performed. The results presented here demonstrate that DdlN from the vancomycin producer A. orientalis C329.2 not only is a d-Ala-d-Lac ligase but also has significant functional homology with VanA. It is not known at present if, like S. toyocaensis NRRL 15009 (16), A. orientalis C329.2 also possess a d-Ala-d-Ala-exclusive ligase, though the presence of a vanX gene (14) suggests that it may.These studies demonstrate that DdlN cloned from a vancomycin-producing bacterium is a d-Ala-d-Lac ligase which has not only amino acid sequence homology with the dd-ligases from VRE but also functional homology. Thus, VanA, VanB, DdlN, and DdlM have likely evolved from similar origins. The fact that a vanH-vanA-vanX gene cluster can be found in other glycopeptide producers as well (14) suggests that the genes now found in VRE may have originated in glycopeptide-producing bacteria. Our finding that overexpressed, purified, DdlN shows many enzymatic characteristics similar (though not identical) to those of VanA suggests that the genes from glycopeptide-producing bacteria can be important in elucidating biochemical and protein structural aspects of the VRE proteins.  相似文献   

3.
4.
Lichenysins are surface-active lipopeptides with antibiotic properties produced nonribosomally by several strains of Bacillus licheniformis. Here, we report the cloning and sequencing of an entire 26.6-kb lichenysin biosynthesis operon from B. licheniformis ATCC 10716. Three large open reading frames coding for peptide synthetases, designated licA, licB (three modules each), and licC (one module), could be detected, followed by a gene, licTE, coding for a thioesterase-like protein. The domain structure of the seven identified modules, which resembles that of the surfactin synthetases SrfA-A to -C, showed two epimerization domains attached to the third and sixth modules. The substrate specificity of the first, fifth, and seventh recombinant adenylation domains of LicA to -C (cloned and expressed in Escherichia coli) was determined to be Gln, Asp, and Ile (with minor Val and Leu substitutions), respectively. Therefore, we suppose that the identified biosynthesis operon is responsible for the production of a lichenysin variant with the primary amino acid sequence l-Gln–l-Leu–d-Leu–l-Val–l-Asp–d-Leu–l-Ile, with minor Leu and Val substitutions at the seventh position.Many strains of Bacillus are known to produce lipopeptides with remarkable surface-active properties (11). The most prominent of these powerful lipopeptides is surfactin from Bacillus subtilis (1). Surfactin is an acylated cyclic heptapeptide that reduces the surface tension of water from 72 to 27 mN m−1 even in a concentration below 0.05% and shows some antibacterial and antifungal activities (1). Some B. subtilis strains are also known to produce other, structurally related lipoheptapeptides (Table (Table1),1), like iturin (32, 34) and bacillomycin (3, 27, 30), or the lipodecapeptides fengycin (50) and plipastatin (29).

TABLE 1

Lipoheptapeptide antibiotics of Bacillus spp.
LipopeptideOrganismStructureReference
Lichenysin AB. licheniformisFAa-L-Glu-L-Leu-D-Leu-L-Val-L-Asn-D-Leu-L-Ile51, 52
Lichenysin BFAa-L-Glu-L-Leu-D-Leu-L-Val-L-Asp-D-Leu-L-Leu23, 26
Lichenysin CFAa-L-Glu-L-Leu-D-Leu-L-Val-L-Asp-D-Leu-L-Ile17
Lichenysin DFAa-L-Gln-L-Leu-D-Leu-L-Val-L-Asp-D-Leu-L-IleThis work
Surfactant 86B. licheniformisFAa-L-Glxd-L-Leu-D-Leu-L-Val-L-Asxd-D-Leu-L-Ilee14, 15
L-Val
SurfactinB. subtilisFAa-L-Glu-L-Leu-D-Leu-L-Val-L-Asp-D-Leu-L-Leu1, 7, 49
EsperinB. subtilisFAb-L-Glu-L-Leu-D-Leu-L-Val-L-Asp-D-Leu-L-Leue45
L-Val 
Iturin AB. subtilisFAc-L-Asn-D-Tyr-D-Asn-L-Gln-L-Pro-D-Asn-L-Ser32
Iturin CFAc-L-Asn-D-Tyr-D-Asn-L-Gln-L-Pro-D-Asne-L-Asne34
D-Ser-L-Thr 
Bacillomycin LB. subtilisFAc-L-Asp-D-Tyr-D-Asn-L-Ser-L-Gln-D-Proe-L-Thr3
D-Ser- 
Bacillomycin DFAc-L-Asp-D-Tyr-D-Asn-L-Pro-L-Glu-D-Ser-L-Thr30, 31
Bacillomycin FFAc-L-Asn-D-Tyr-D-Asn-L-Gln-L-Pro-D-Asn-L-Thr27
Open in a separate windowaFA, β-hydroxy fatty acid. The β-hydroxy group forms an ester bond with the carboxy group of the C-terminal amino acid. bFA, β-hydroxy fatty acid. The β-hydroxy group forms an ester bond with the carboxy group of Asp5. cFA, β-amino fatty acid. The β-amino group forms a peptide bond with the carboxy group of the C-terminal amino acid. dOnly the following combinations of amino acid 1 and 5 are allowed: Gln-Asp or Glu-Asn. eWhere an alternative amino acid may be present in a structure, the alternative is also presented. In addition to B. subtilis, several strains of Bacillus licheniformis have been described as producing the lipopeptide lichenysin (14, 17, 23, 26, 51). Lichenysins can be grouped under the general sequence l-Glx–l-Leu–d-Leu–l-Val–l-Asx–d-Leu–l-Ile/Leu/Val (Table (Table1).1). The first amino acid is connected to a β-hydroxyl fatty acid, and the carboxy-terminal amino acid forms a lactone ring to the β-OH group of the lipophilic part of the molecule. In contrast to the lipopeptide surfactin, lichenysins seem to be synthesized during growth under aerobic and anaerobic conditions (16, 51). The isolation of lichenysins from cells growing on liquid mineral salt medium on glucose or sucrose basic has been studied intensively. Antimicrobial properties and the ability to reduce the surface tension of water have also been described (14, 17, 26, 51). The structural elucidation of the compounds revealed slight differences, depending on the producer strain. Various distributions of branched and linear fatty acid moieties of diverse lengths and amino acid variations in three defined positions have been identified (Table (Table11).In contrast to the well-defined methods for isolation and structural characterization of lichenysins, little is known about the biosynthetic mechanisms of lichenysin production. The structural similarity of lichenysins and surfactin suggests that the peptide moiety is produced nonribosomally by multifunctional peptide synthetases (7, 13, 25, 49, 53). Peptide synthetases from bacterial and fungal sources describe an alternative route in peptide bond formation in addition to the ubiquitous ribosomal pathway. Here, large multienzyme complexes affect the ordered recognition, activation, and linking of amino acids by utilizing the thiotemplate mechanism (19, 24, 25). According to this model, peptide synthetases activate their substrate amino acids as aminoacyl adenylates by ATP hydrolysis. These unstable intermediates are subsequently transferred to a covalently enzyme-bound 4′-phosphopantetheinyl cofactor as thioesters. The thioesterified amino acids are then integrated into the peptide product through a stepwise elongation by a series of transpeptidations directed from the amino terminals to the carboxy terminals. Peptide synthetases have not only awakened interest because of their mechanistic features; many of the nonribosomally processed peptide products also possess important biological and medical properties.In this report we describe the identification and characterization of a putative lichenysin biosynthesis operon from B. licheniformis ATCC 10716. Cloning and sequencing of the entire lic operon (26.6 kb) revealed three genes, licA, licB, and licC, with structural patterns common to peptide synthetases and a gene designated licTE, which codes for a putative thioesterase. The modular organization of the sequenced genes resembles the requirements for the biosynthesis of the heptapeptide lichenysin. Based on the arrangement of the seven identified modules and the tested substrate specificities, we propose that the identified genes are involved in the nonribosomal synthesis of the portion of the lichenysin peptide with the primary sequence l-Gln–l-Leu–d-Leu–l-Val–l-Asp–d-Leu–l-Ile (with minor Val and Leu substitutions).  相似文献   

5.
The intraflagellar transport machinery is required for the assembly of cilia. It has been investigated by biochemical, genetic, and computational methods that have identified at least 21 proteins that assemble into two subcomplexes. It has been hypothesized that complex A is required for retrograde transport. Temperature-sensitive mutations in FLA15 and FLA17 show defects in retrograde intraflagellar transport (IFT) in Chlamydomonas. We show that IFT144 and IFT139, two complex A proteins, are encoded by FLA15 and FLA17, respectively. The fla15 allele is a missense mutation in a conserved cysteine and the fla17 allele is an in-frame deletion of three exons. The flagellar assembly defect of each mutant is rescued by the respective transgenes. In fla15 and fla17 mutants, bulges form in the distal one-third of the flagella at the permissive temperature and this phenotype is also rescued by the transgenes. These bulges contain the complex B component IFT74/72, but not α-tubulin or p28, a component of an inner dynein arm, which suggests specificity with respect to the proteins that accumulate in these bulges. IFT144 and IFT139 are likely to interact with each other and other proteins on the basis of three distinct genetic tests: (1) Double mutants display synthetic flagellar assembly defects at the permissive temperature, (2) heterozygous diploid strains exhibit second-site noncomplemention, and (3) transgenes confer two-copy suppression. Since these tests show different levels of phenotypic sensitivity, we propose they illustrate different gradations of gene interaction between complex A proteins themselves and with a complex B protein (IFT172).CILIA and flagella are microtubule-based organelles that are found on most mammalian cells. They provide motility to cells and participate in many sensory processes. Defects in or loss of cilia/flagella cause a variety of human diseases that include polycystic kidney disease, retinal degeneration, infertility, obesity, respiratory defects, left–right axis determination, and polydactyly (Fliegauf et al. 2007). Mouse mutants demonstrate that cilia are essential for viability, neural tube closure, and bone development (Eggenschwiler and Anderson 2007; Fliegauf et al. 2007). Cilia and flagella are also present in protists, algae, moss, and some fungi.The assembly and maintenance of cilia and flagella require intraflagellar transport (IFT) (Kozminski et al. 1995). IFT involves the movement of 100- to 200-nm-long protein particles from the basal body located in the cell body to the tip of the flagella using the heterotrimeric kinesin-2 (anterograde movement) (Kozminski et al. 1995) and movement back to the cell body (retrograde movement) using the cytoplasmic dynein complex (Pazour et al. 1999; Porter et al. 1999). IFT particles change their direction of movement as well as their size, speed, and frequency at the ends of the flagella as they switch from anterograde to retrograde movement (Iomini et al. 2001). Biochemical isolation of IFT particles reveals that they are composed of at least 16 proteins and that these particles can be dissociated into two complexes in vitro by changing the salt concentration (Cole et al. 1998; Piperno et al. 1998). Recent genetic and bioinformatics analysis adds at least 7 more proteins to the IFT particle (Follit et al. 2009) (Eggenschwiler and Anderson 2007).

TABLE 1

Proteins and gene names for the intraflagellar transport particles in Chlamydomonas, C. elegans, and mouse
ProteinMotifChlamydomonas geneC. elegans geneMouse geneReferences to worm and mouse genes
Complex A
IFT144WDFLA15
IFT140WDche-11Qin et al. (2001)
IFT139TRPFLA17dyf-2THM1Efimenko et al. (2006); Tran et al. (2008)
IFT122WDIFTA-1Blacque et al. (2006)
IFT121WDdaf-10Bell et al. (2006)
IFT43
Complex B
IFT172WDFLA11osm-1WimpleHuangfu et al. (2003); Pedersen et al. (2005); Bell et al. (2006)
IFT88TRPIFT88osm-5Tg737/PolarisPazour et al. (2000); Qin et al. (2001)
IFT81Coilift-81CDV1Kobayashi et al. (2007)
IFT80WDche-2Wdr56Fujiwara et al. (1999)
IFT74/72Coilift-74Cmg1Kobayashi et al. (2007)
IFT57/55Coilche-13HippiHaycraft et al. (2003)
IFT54Microtubule binding domain MIP-T3dyf-11Traf3IP1Kunitomo and Iino (2008); Li et al. (2008); Omori et al. (2008); Follit et al. (2009)
IFT52ABC typeBLD1osm-6Ngd2Brazelton et al. (2001); Bell et al. (2006)
IFT46IFT46dyf-6Bell et al. (2006); Hou et al. (2007)
IFT27G proteinNot presentRabl4
IFT25Hsp20Not presentHSP16.1Follit et al. (2009)
IFT22G proteinIFTA-2Rabl5Schafer et al. (2006)
IFT20CoilFollit et al. (2006)
FAP22Cluamp related proteindyf-3Cluamp1Murayama et al. (2005); Follit et al. (2009)
DYF13


dyf-13
Ttc26
Blacque et al. (2005)
Open in a separate window—, no mutant found to date in Chlamydomonas.A collection of temperature-sensitive mutant strains that fail to assemble flagella at the restrictive temperature of 32° was isolated in Chlamydomonas (Huang et al. 1977; Adams et al. 1982; Piperno et al. 1998; Iomini et al. 2001). Analysis of the flagella at 21° permits the measurement of the velocity and frequency of IFT particles in the mutant strains. This analysis suggested that assembly has four phases: recruitment to the basal body, anterograde movement (phases I and II), retrograde movement, and return to the cytoplasm (phases III and IV) (Iomini et al. 2001). Different mutants were classified as defective in these four phases. However, because different alleles of FLA8 were classified as defective in different phases (Iomini et al. 2001; Miller et al. 2005), we combined mutants with IFT defects into just two classes. The first group (phases I and II) includes mutant strains that show decreased anterograde velocities, a decreased ratio of anterograde to retrograde particles, and an accumulation of complex A proteins at the basal body. This group includes mutations in the FLA8 and FLA10 genes, which encode the two motor subunits of kinesin-2 (Walther et al. 1994; Miller et al. 2005), as well as mutations in three unknown genes (FLA18, FLA27, and FLA28). The second group includes mutant strains that show the reciprocal phenotype (phases III and IV); these phenotypes include decreased retrograde velocities, an increased ratio of anterograde to retrograde particles, and an accumulation of complex B proteins in the flagella. With the exception of the FLA11 gene, which encodes IFT172, a component of complex B (Pedersen et al. 2005), the gene products in this class are unknown (FLA2, FLA15, FLA16, FLA17, and FLA24). One might predict that mutations in this group would map to genes that encode complex A or retrograde motor subunits. Interestingly, IFT particles isolated from fla11, fla15, fla16, and fla17-1 flagella show depletion of complex A polypeptides (Piperno et al. 1998; Iomini et al. 2001). The inclusion of IFT172 in this class is explained by the observations that IFT172 plays a role in remodeling the IFT particles at the flagellar tip to transition from anterograde to retrograde movement (Pedersen et al. 2005). The remaining mutant strains do not show obvious defects in velocities, ratios, or accumulation at 21° and may reflect a less severe phenotype at the permissive temperature or a non-IFT role for these genes.Direct interactions occur between components of complex B. IFT81 and IFT74/72 interact to form a scaffold required for IFT complex B assembly (Lucker et al. 2005). IFT57 and IFT20 also interact with each other and kinesin-2 (Baker et al. 2003). While physical interactions are being used to define IFT particle architecture, genetic interactions among loci encoding IFT components should be instructive regarding their function as well. To probe retrograde movement and its function, we have identified the gene products encoded by two retrograde defective mutant strains. They are FLA15 and FLA17 and encode IFT144 and IFT139, respectively. The genetic interactions of these loci provide interesting clues about the assembly of the IFT particles and possible physical interactions in the IFT particles.  相似文献   

6.
The presence of two systems in Escherichia coli for gluconate transport and phosphorylation is puzzling. The main system, GntI, is well characterized, while the subsidiary system, GntII, is poorly understood. Genomic sequence analysis of the region known to contain genes of the GntII system led to a hypothesis which was tested biochemically and confirmed: the GntII system encodes a pathway for catabolism of l-idonic acid in which d-gluconate is an intermediate. The genes have been named accordingly: the idnK gene, encoding a thermosensitive gluconate kinase, is monocistronic and transcribed divergently from the idnD-idnO-idnT-idnR operon, which encodes l-idonate 5-dehydrogenase, 5-keto-d-gluconate 5-reductase, an l-idonate transporter, and an l-idonate regulatory protein, respectively. The metabolic sequence is as follows: IdnT allows uptake of l-idonate; IdnD catalyzes a reversible oxidation of l-idonate to form 5-ketogluconate; IdnO catalyzes a reversible reduction of 5-ketogluconate to form d-gluconate; IdnK catalyzes an ATP-dependent phosphorylation of d-gluconate to form 6-phosphogluconate, which is metabolized further via the Entner-Doudoroff pathway; and IdnR appears to act as a positive regulator of the IdnR regulon, with l-idonate or 5-ketogluconate serving as the true inducer of the pathway. The l-idonate 5-dehydrogenase and 5-keto-d-gluconate 5-reductase reactions were characterized both chemically and biochemically by using crude cell extracts, and it was firmly established that these two enzymes allow for the redox-coupled interconversion of l-idonate and d-gluconate via the intermediate 5-ketogluconate. E. coli K-12 strains are able to utilize l-idonate as the sole carbon and energy source, and as predicted, the ability of idnD, idnK, idnR, and edd mutants to grow on l-idonate is altered.In Escherichia coli, the Entner-Doudoroff (ED) pathway serves as a metabolic “funnel” receiving intermediates formed by catabolism of several sugar acids (17). Hexuronic acids undergo rearrangement in the inducible Ashwell pathways (1) to form 2-keto-3-deoxygluconate, which is then phosphorylated to produce 2-keto-3-deoxy-6-phosphogluconate (KDPG). KDPG is cleaved by KDPG aldolase, encoded by eda, providing for entry of carbon into glycolysis. The other enzyme of the ED pathway is 6-phosphogluconate dehydratase, encoded by edd, which is induced only for catabolism of gluconate and also forms KDPG, the key intermediate of the ED pathway (7). Long considered to be of more significance than is readily obvious (9), the finding that eda and edd eda double mutants are unable to colonize the mouse large intestine underscores the possible ecological importance of ED metabolism (32). The implication from these colonization studies is that colonic mucus, which contains several sugar acids, may serve as an important source of nutrients for E. coli in the gut.Also participating in gluconate catabolism are several gluconate transporters and two gluconate kinases which appear, based upon their regulation, to comprise two distinct systems (2, 13). The GntI (main) system consists of gntT, gntU, and gntK, which code for high- and low-affinity gluconate transporters and a thermoresistant gluconate kinase, respectively (2325, 33). Expression of the GntR regulon, that is, GntI together with the edd-eda operon, is negatively controlled by the gntR gene product. The GntII (subsidiary) system is comprised of a thermosensitive gluconate kinase and a gluconate transporter which function for gluconate catabolism in the absence of the GntI system (2, 11, 13, 22). It appears that the subsidiary gluconate transporter, which has an apparent Km for gluconate of 60 μM (23), is encoded by a gene (idnT) which is adjacent to the gene encoding the thermosensitive gluconokinase (idnK) at 96.8 min.The DNA sequence of the GntII system genes, located at 4492 kb on the genome, was revealed by the E. coli Genome Project (5, 6). If the GntII system had evolved as a subsidiary pathway for gluconate catabolism, one would expect it to contain only a gluconate transporter and gluconate kinase. However, in addition to the divergent idnK and idnT genes, this region also encodes two “dehydrogenase-like” enzymes. The similarity of idnO to gno of Gluconobacter oxydans, which encodes d-gluconate:NADP 5-oxidoreductase (GNO) (15), led to the testing of ketogluconates as enzyme substrates for the two newly identified dehydrogenases. A process of deductive reasoning and biochemical experiments led to the conclusion that the GntII system in fact comprises a novel metabolic pathway for catabolism of l-idonic acid, in which gluconate is a key intermediate. Accordingly, the genes involved in l-idonate metabolism have been given the designation idn (see Table Table11 for gene nomenclature).

TABLE 1

Gene and enzyme nomenclaturea
Gene designation
Gene product% Identity of proteinb
PreviousNew (acces- sion no.)
gntVidnK (P39208)d-Gluconate kinase45 (GntKc)
yjgVidnD (P39346)l-Idonate 5-dehydrogenase30.6 (sheep DHSOd)
yjgUidnO (P39345)5-Keto-d-gluconate 5-reductase56 (GNOe)
gntWidnT (P39344)l-Idonate transporter61 (GntTf)
yjgSidnR (P39343)l-Idonate regulator46 (GntRg)
Open in a separate windowaAll accession numbers are Swiss-Prot database accession numbers. bPercent identity of the amino acid sequence of the Idn protein to that of the protein shown in parentheses. cE. coli gluconate kinase encoded by gntK (P46859). dSheep sorbitol dehydrogenase encoded by sorD (P07846). eG. oxydans gluconate:NADP 5-oxidoreductase encoded by gno (P50199). fE. coli gluconate transporter encoded by gntT (P39835). gE. coli gluconate regulator encoded by gntR (P46860). (Part of this work has been presented previously [3].)  相似文献   

7.
The capacity for phenotypic evolution is dependent upon complex webs of functional interactions that connect genotype and phenotype. Wrinkly spreader (WS) genotypes arise repeatedly during the course of a model Pseudomonas adaptive radiation. Previous work showed that the evolution of WS variation was explained in part by spontaneous mutations in wspF, a component of the Wsp-signaling module, but also drew attention to the existence of unknown mutational causes. Here, we identify two new mutational pathways (Aws and Mws) that allow realization of the WS phenotype: in common with the Wsp module these pathways contain a di-guanylate cyclase-encoding gene subject to negative regulation. Together, mutations in the Wsp, Aws, and Mws regulatory modules account for the spectrum of WS phenotype-generating mutations found among a collection of 26 spontaneously arising WS genotypes obtained from independent adaptive radiations. Despite a large number of potential mutational pathways, the repeated discovery of mutations in a small number of loci (parallel evolution) prompted the construction of an ancestral genotype devoid of known (Wsp, Aws, and Mws) regulatory modules to see whether the types derived from this genotype could converge upon the WS phenotype via a novel route. Such types—with equivalent fitness effects—did emerge, although they took significantly longer to do so. Together our data provide an explanation for why WS evolution follows a limited number of mutational pathways and show how genetic architecture can bias the molecular variation presented to selection.UNDERSTANDING—and importantly, predicting—phenotypic evolution requires knowledge of the factors that affect the translation of mutation into phenotypic variation—the raw material of adaptive evolution. While much is known about mutation rate (e.g., Drake et al. 1998; Hudson et al. 2002), knowledge of the processes affecting the translation of DNA sequence variation into phenotypic variation is minimal.Advances in knowledge on at least two fronts suggest that progress in understanding the rules governing the generation of phenotypic variation is possible (Stern and Orgogozo 2009). The first stems from increased awareness of the genetic architecture underlying specific adaptive phenotypes and recognition of the fact that the capacity for evolutionary change is likely to be constrained by this architecture (Schlichting and Murren 2004; Hansen 2006). The second is the growing number of reports of parallel evolution (e.g., Pigeon et al. 1997; ffrench-Constant et al. 1998; Allender et al. 2003; Colosimo et al. 2004; Zhong et al. 2004; Boughman et al. 2005; Shindo et al. 2005; Kronforst et al. 2006; Woods et al. 2006; Zhang 2006; Bantinaki et al. 2007; McGregor et al. 2007; Ostrowski et al. 2008)—that is, the independent evolution of similar or identical features in two or more lineages—which suggests the possibility that evolution may follow a limited number of pathways (Schluter 1996). Indeed, giving substance to this idea are studies that show that mutations underlying parallel phenotypic evolution are nonrandomly distributed and typically clustered in homologous genes (Stern and Orgogozo 2008).While the nonrandom distribution of mutations during parallel genetic evolution may reflect constraints due to genetic architecture, some have argued that the primary cause is strong selection (e.g., Wichman et al. 1999; Woods et al. 2006). A means of disentangling the roles of population processes (selection) from genetic architecture is necessary for progress (Maynard Smith et al. 1985; Brakefield 2006); also necessary is insight into precisely how genetic architecture might bias the production of mutations presented to selection.Despite their relative simplicity, microbial populations offer opportunities to advance knowledge. The wrinkly spreader (WS) morphotype is one of many different niche specialist genotypes that emerge when experimental populations of Pseudomonas fluorescens are propagated in spatially structured microcosms (Rainey and Travisano 1998). Previous studies defined, via gene inactivation, the essential phenotypic and genetic traits that define a single WS genotype known as LSWS (Spiers et al. 2002, 2003) (Figure 1). LSWS differs from the ancestral SM genotype by a single nonsynonymous nucleotide change in wspF. Functionally (see Figure 2), WspF is a methyl esterase and negative regulator of the WspR di-guanylate cyclase (DGC) (Goymer et al. 2006) that is responsible for the biosynthesis of c-di-GMP (Malone et al. 2007), the allosteric activator of cellulose synthesis enzymes (Ross et al. 1987). The net effect of the wspF mutation is to promote physiological changes that lead to the formation of a microbial mat at the air–liquid interface of static broth microcosms (Rainey and Rainey 2003).Open in a separate windowFigure 1.—Outline of experimental strategy for elucidation of WS-generating mutations and their subsequent identity and distribution among a collection of independently evolved, spontaneously arising WS genotypes. The strategy involves, first, the genetic analysis of a specific WS genotype (e.g., LSWS) to identify the causal mutation, and second, a survey of DNA sequence variation at specific loci known to harbor causal mutations among a collection of spontaneously arising WS genotypes. For example, suppressor analysis of LSWS using a transposon to inactivate genes necessary for expression of the wrinkly morphology delivered a large number of candidate genes (top left) (Spiers et al. 2002). Genetic and functional analysis of these candidate genes (e.g., Goymer et al. 2006) led eventually to the identity of the spontaneous mutation (in wspF) responsible for the evolution of LSWS from the ancestral SM genotype (Bantinaki et al. 2007). Subsequent analysis of the wspF sequence among 26 independent WS genotypes (bottom) showed that 50% harbored spontaneous mutations (of different kinds; see Open in a separate windowFigure 2.—Network diagram of DGC-encoding pathways underpinning the evolution of the WS phenotype and their regulation. Overproduction of c-di-GMP results in overproduction of cellulose and other adhesive factors that determine the WS phenotype. The ancestral SBW25 genome contains 39 putative DGCs, each in principle capable of synthesizing the production of c-di-GMP, and yet WS genotypes arise most commonly as a consequence of mutations in just three DGC-containing pathways: Wsp, Aws, and Mws. In each instance, the causal mutations are most commonly in the negative regulatory component: wspF, awsX, and the phosphodiesterase domain of mwsR (see text).To determine whether spontaneous mutations in wspF are a common cause of the WS phenotype, the nucleotide sequence of this gene was obtained from a collection of 26 spontaneously arising WS genotypes (WSA-Z) taken from 26 independent adaptive radiations, each founded by the same ancestral SM genotype (Figure 1): 13 contained mutations in wspF (Bantinaki et al. 2007). The existence of additional mutational pathways to WS provided the initial motivation for this study.

TABLE 1

Mutational causes of WS
WS genotypeGeneNucleotide changeAmino acid changeSource/reference
LSWSwspFA901CS301RBantinaki et al. (2007)
AWSawsXΔ100-138ΔPDPADLADQRAQAThis study
MWSmwsRG3247AE1083KThis study
WSAwspFT14GI5SBantinaki et al. (2007)
WSBwspFΔ620-674P206Δ (8)aBantinaki et al. (2007)
WSCwspFG823TG275CBantinaki et al. (2007)
WSDwspEA1916GD638GThis study
WSEwspFG658TV220LBantinaki et al. (2007)
WSFwspFC821TT274IBantinaki et al. (2007)
WSGwspFC556TH186YBantinaki et al. (2007)
WSHwspEA2202CK734NThis study
WSIwspEG1915TD638YThis study
WSJwspFΔ865-868R288Δ (3)aBantinaki et al. (2007)
WSKawsOG125TG41VThis study
WSLwspFG482AG161DBantinaki et al. (2007)
WSMawsRC164TS54FThis study
WSNwspFA901CS301RBantinaki et al. (2007)
WSOwspFΔ235-249V79Δ (6)aBantinaki et al. (2007)
WSPawsR222insGCCACCGAA74insATEThis study
WSQmwsR3270insGACGTG1089insDVThis study
WSRmwsRT2183CV272AThis study
WSSawsXC472TQ158STOPThis study
WSTawsXΔ229-261ΔYTDDLIKGTTQThis study
WSUwspFΔ823-824T274Δ (13)aBantinaki et al. (2007)
WSVawsXT74GL24RThis study
WSWwspFΔ149L49Δ (1)aBantinaki et al. (2007)
WSXb???This study
WSYwspFΔ166-180Δ(L51-I55)Bantinaki et al. (2007)
WSZ
mwsR
G3055A
A1018T
This study
Open in a separate windowaP206Δ(8) indicates a frameshift; the number of new residues before a stop codon is reached is in parentheses.bSuppressor analysis implicates the wsp locus (17 transposon insertions were found in this locus). However, repeated sequencing failed to identify a mutation.Here we define and characterize two new mutational routes (Aws and Mws) that together with the Wsp pathway account for the evolution of 26 spontaneously arising WS genotypes. Each pathway offers approximately equal opportunity for WS evolution; nonetheless, additional, less readily realized genetic routes producing WS genotypes with equivalent fitness effects exist. Together our data show that regulatory pathways with specific functionalities and interactions bias the molecular variation presented to selection.  相似文献   

8.
9.
Alexey Yanchukov 《Genetics》2009,182(4):1117-1127
A model of genomic imprinting with complete inactivation of the imprinted allele is shown to be formally equivalent to the haploid model of parental selection. When single-locus dynamics are considered, an internal equilibrium is possible only if selection acts in the opposite directions in males and females. I study a two-locus version of the latter model, in which maternal and paternal effects are attributed to the single alleles at two different loci. A necessary condition for the allele frequency equilibria to remain on the linkage equilibrium surface is the multiplicative interaction between maternal and paternal fitness parameters. In this case the equilibrium dynamics are independent at both loci and results from the single-locus model apply. When fitness parameters are additive, analytic treatment was not possible but numerical simulations revealed that stable polymorphism characterized by association between loci is possible only in several special cases in which maternal and paternal fitness contributions are precisely balanced. As in the single-locus case, antagonistic selection in males and females is a necessary condition for the maintenance of polymorphism. I also show that the above two-locus results of the parental selection model are very sensitive to the inclusion of weak directional selection on the individual''s own genotypes.PARENTAL genetic effects refer to the influence of the mother''s and father''s genotypes on the phenotypes of their offspring, not attributable just to the transfer of genes. Examples have been documented across a wide range of areas of organism biology; see, for example, Wade (1998) and and22 in Rasanen and Kruuk (2007). Parental selection is a more formal concept used in theoretical modeling and concerns situations where the fitness of the offspring depends, besides other factors, on the genotypes of its parent(s) (generalizing from Kirkpatrick and Lande 1989).

TABLE 1

Frequencies of genotypes and fitness parameterizations in model 1
Gametes/haploidsFrequency before selectionFitness
ZygoteMaleFemale
(A)AApfpm1 − α1 − δ
(A)a1/2 A 1/2 apf(1 − pm)11
(a)A1/2 a 1/2 A(1 − pf)pm1 − α1 − δ
(a)aA(1 − pf)(1 − pm)11
Open in a separate windowParentheses in the first column indicate maternal genotype (parental selection model) or inactivation of the maternally derived allele (imprinting model). Whether selection occurs at the diploid (first column) or subsequent haploid (second column) stage does not change the resulting allele frequencies.

TABLE 2

Offspring genotypic proportions from different mating types, sorted among four phenotypic groups/combinations of maternal and paternal effects: model 2
Offspring genotypes/phenotypes
Parental genotypes
Paternal (φ = 1)
Joint (φ = 4)
MaleFemaleABAbaBAbABAbaBab
ABAB1
Ab
aB
ab(1−r)/2r/2r/2(1−r)/2
AbAB
Ab1
aBr/2(1−r)/2(1−r)/2r/2
ab
Offspring genotypes/phenotypes
Parental genotypes
Maternal (φ = 2)
None (φ = 3)
MaleFemaleABAbaBAbABAbaBab
aBAB
Abr/2(1 − r)/2(1 − r)/2r/2
aB1
ab
abAB(1 − r)/2(1 − r)/2
Ab
aB
ab1
Open in a separate windowAnother well-known parent-of-origin phenomenon is genomic imprinting. Here, the level of expression of one of the alleles depends on which parent it is inherited from. Often it is difficult to tell apart the phenotypic patterns due to parental effects and genomic imprinting, and thus a problem arises in the process of identifying the candidate genes for such effects (Hager et al. 2008). Analytic methods (Weinberg et al. 1998; Santure and Spencer 2006; Hager et al. 2008) have been developed to quantify subtle differences between the two. In this article, I point out that a simple mathematical model, first suggested for genomic imprinting at a diploid locus, can be interpreted, without any formal changes, to describe parental selection on haploids.While there has been much progress in understanding the evolution of genomic imprinting (Hunter 2007), including advances in modeling (Spencer 2000, 2008), the population genetics theory of parental effects received less attention. Existing major-locus effect models of parental selection are single-locus, two-allele, and mostly concern uniparental (maternal) selection (Wright 1969; Spencer 2003; Gavrilets and Rice 2006; Santure and Spencer 2006), with only one specific case where the fitness effects of both parents interact studied by Gavrilets and Rice (2006). No attempt to extend this theory into multilocus systems has yet been made. Considering a two-locus model with both parents playing a role in selection on the offspring is called for by the observation that many maternal and paternal effects aim at the different traits or different life stages of their progeny. Among birds, for example, body condition soon after hatching is largely determined by the mother, while paternally transmitted sexual display traits develop much later in life (Price 1998). Such effects are therefore unlikely to be regulated within a single locus. Sometimes the effects are on the same trait, but still attributed to different loci: expression of gene Avy that causes the “agouti” phenotype (yellow fur coat and obesity) in mice is enhanced by maternal epigenetic modification (Rakyan et al. 2003), while paternal mutations at the other locus, MommeD4, contribute to a reverse phenotypic pattern in the offspring (Rakyan et al. 2003). The epigenetic state of the murine AxinFu allele is both maternally and paternally inherited (Rakyan et al. 2003).Focusing selection on haploids reduces the number of genotypes that need to be taken into account, while preserving the main properties of the multilocus system. Genes with haploid expression and a potential of parental effects can be found in two major taxonomic kingdoms. A notable candidate is Spam1 in mice, which is expressed during spermogenesis and encodes a factor that enables sperm to penetrate the egg cumulus (Zheng et al. 2001). This gene remains a target for effectively haploid selection, because its product is not shared via cytoplasm bridges between developing spermatides. Mutations at Spam1 alter performance of the male gametes that carry it and might indirectly, perhaps by altering the timing of fertilization, affect the fitness of the zygote. The highest estimated number of mouse genes expressed in the male gametes is currently 2375 (Joseph and Kirkpatrick 2004), and one might expect some of them to have similar paternal effects. Plants go through a profound haploid stage in their life cycles, and genes involved at this stage have an inevitable effect on the fitness of the future generations. In angiosperms, seed development is known to be controlled by both maternal (Chaudhury and Berger 2001; Yadegari and Drews 2004) and paternal (Nowack et al. 2006) effect genes, expressed, respectively, in female and male gametophytes.Under haploid selection, there can be no overdominance, and thus polymorphism is much more difficult to maintain than in diploid selection models (summarized in Feldman 1971). Nevertheless, differential or antagonistic selection between sexes can lead to a new class of stable internal equilibria in the diploid systems (Owen 1953; Bodmer 1965; Mandel 1971; Kidwell et al. 1977; Reed 2007), and I make use of this property in the haploid models developed below. In the experiment by Chippindale and colleagues (Chippindale et al. 2001), ∼75% of the total fitness variation in the adult stage of Drosophila melanogaster was negatively correlated between males and females, which suggests that a substantial portion of the fruit fly expressed genome is under sexually antagonistic selection. I assume that the effect of either parent on the fitness of the individual depends on the sex of the latter, which in respect to modeling is equivalent to the assumption of differential viability between the sexes in the progeny of the same parent(s). Biological systems that satisfy the latter assumptions can be found among colonial green algae: many members of the order Volvocales are haploid except for the short zygotic stage, and during sexual reproduction, they are also dioecious and anisogametic. I return to this example in the discussion. The possibility that genes expressed in animal gametes may be under antagonistic selection between sexes has been discussed (Bernasconi et al. 2004). For example, a (hypothetical) mutation increasing the ATP production in mitochondria would be beneficial in sperm, because of the increased mobility of the latter, but neutral or detrimental in the egg, due to a higher level of oxidative damage to DNA (Zeh and Zeh 2007).My main purpose was to derive conditions for existence and stability of the internal equilibria of the model(s). I begin with a simple one-locus case, which can be analyzed explicitly, and show how these one-locus results can be extended to the case of two recombining loci with multiplicative fitness. Then, I assume an additive relation between the maternal and paternal effect parameters and study the special cases where parental effects are symmetric.  相似文献   

10.
The Gram-negative bacteria Campylobactor jejuni is the primary bacteria responsible for food poisoning in industrialized countries, and acute diarrheal illness is a leading cause of mortality among children in developing countries. C. jejuni are commensal in chickens. They are particularly abundant in the caecal crypts, and poultry products are commonly infected as a result of cross-contamination during processing. The interactions between C. jejuni and chicken intestinal tissues as well as the pathogenic molecular mechanisms of colonization in humans are unknown, but identifying these factors could provide potential targets to reduce the incidence of campylobacteriosis. Recently, purified chicken intestinal mucin was shown to attenuate adherence and invasion of C. jejuni in the human colorectal adenocarcinoma cell line HCT-8 in vitro, and this effect was attributed to mucin O-glycosylation. Mucins from different regions of the chicken intestine inhibited C. jejuni binding and internalization differentially, with large intestine>small intestine>caecum. Here, we use LC-MS to perform a detailed structural analysis of O-glycans released from mucins purified from chicken large intestine, small intestine, and caecum. The O-glycans identified were abundantly sulfated compared with the human intestines, and sulfate moieties were present throughout the chicken intestinal tract. Interestingly, alpha 1–2 linked fucose residues, which have a high binding affinity to C. jejuni, were identified in the small and large intestines. Additionally, N-glycolylneuraminic/N-acetylneuraminic acid containing structures present as Sda-like epitopes were identified in large intestine samples but not small intestine or caecum. O-glycan structural characterization of chicken intestinal mucins provides insights into adherence and invasion properties of C. jejuni, and may offer prospective candidate molecules aimed at reducing the incidence of infection.Campylobactor jejuni infection is widespread in industrialized countries and is the greatest source of food poisoning worldwide. Infection and the resulting diarrhea are the most common cause of death among young children in developing countries, with an estimated 2.1–2.5 million cases per year in the USA. C. jejuni leads to severe gastroenteritis and is also linked to Guillain-Barré and irritable bowel disease (1, 2). Contaminated poultry meat is the most common source of infection, with up to 88% of poultry products being infected (3). The bacteria are commensal in chickens and exist throughout their intestinal tract. However, they do not invade the epithelium of the gut. Purified mucin glycoproteins from the caecum and small and large intestine have been shown to exhibit inhibitory properties against the bacteria in vitro (4). The commensal relationship between C. jejuni occurrence in chickens and the inability to invade the intestinal tissues is poorly understood. Enhanced understanding of how the properties of the chicken mucosal barrier differ from humans; as well as changes in mucin composition along the chicken intestinal tract, have the potential to explain the commensal behavior of C. jejuni in this species. Such information could also provide leads for the development of agents to limit infection.Intestinal mucus in vertebrates is a hydrated gel comprised mainly of very high molecular weight and polymeric secreted mucin glycoproteins that are heavily O-glycosylated. O-linked glycans constitute up to 80% of the mucin by weight and represent an abundant potential carbon source for the resident microflora. They also present targets for bacterial adhesion and chemotaxis that may be exploited by both commensal and pathogenic organisms. Intestinal mucus is a dynamic barrier layer that is mainly secreted by goblet cells. It forms a supramucosal layer to protect the gastrointestinal epithelial cells against infection. A density gradient of viscoelastic and highly hydrated mucins polymers exists from the inner to the outer mucus layer. This stratification is most clearly demonstrable in the colon, where a relatively tightly attached mucus layer proximal to the epithelium is normally devoid of bacteria, but a less cross-linked more superficial layer is heavily colonized (5). In humans, it has been shown that the abundance of bacteria is highest in the large intestine, where colonization of the mucosal epithelium is contested by a thick mucus layer (∼700 μm) and more rapid mucus turnover (6). In the small intestine, however, where bacteria are much less abundant, the mucus layer is between 100–400 μm thick. O-glycosylation confers the mucus component of the mucosal barrier with much of the protective properties required to separate the abundant gut microflora from the immune system of its host. Defects in mucin glycosylation lead to severe inflammation and susceptibility to infection, and the glycans themselves have been shown to be ligands that can block the binding of microorganisms (714). Thus, secreted gel-forming mucins are key components of intestinal defense, beyond which bacterial pathogens must normally penetrate to cause pathology through interaction with epithelial cells.Several C. jejuni adhesion proteins required for colonization of chickens have been identified (15), but the mechanisms by which the bacteria initiate interactions with carbohydrates on mucosal surfaces remain unclear. However, many hypotheses can be made from previous studies where bacterial-carbohydrate interactions have been investigated (7). Additionally, bovine mucins and l-fucose are known chemoattractants for C. jejuni (8), and l-fucose is a substrate for C. jejuni growth (13). Glycans from human breast milk bearing the H(O) blood group antigen (which presents α1–2-linked fucose) also inhibits infection (12). d-glucose, d-mannose, and d-fucose, but not the l-sugar equivalents, inhibited the binding of C. jejuni to human colonic Caco-2 cells (16). Muc1, a membrane-bound mucin, is up-regulated during infection in mice, and knockouts are highly prone to C. jejuni infection with transepithelial translocation (11). These studies have provided considerable insight into C. jejuni behavior, but the specific relationship between the bacteria and mucin glycans in vivo remains unidentified.

Table I

Oligosaccharide-specific C. jejuni interactions
SourceTypeObservationsRef.
Glycan arraymannose Lewisa Lewisb Lewisx Lewisy α2–3Neu5Ac α2–6Neu5Ac terminal- galactoseBacterial binding was affected by temperature, strain and oxygen concentration7
Human milkLewisb α1–2 fucoseInhibition of bacterial growth12
Monosaccharided-mannose d-glucose d-fucoseDecrease in association and invasion with Caco-2 human colorectal cells15
Monosaccharidel-fucoseChemotaxis towards l-fucose and mucins8
Bovine mucinO-glycans
Murine MUC- 1O-glycansUp-regulated MUC1 during infection and KO mice are more susceptible to infection11
Monosaccharidel-fucosel-fucose improved bacterial growth as energy source13
Open in a separate windowLewisa, Galβ1–3(Fucα1–4)GlcNac; Lewisb, Fucα1–2Galβ1–3(Fucα1–4)GlcNac; Lewisx, Galβ1–4(Fucα1–3)GlcNac; Lewisy, Fucα1–2Galβ1–4(Fucα1–3)GlcNac; Neu5Ac, N-acetylneuraminic acid.Recently, Alemka et al. demonstrated that adherence and internalization of C. jejuni of the human intestinal cell line HCT-8 was inhibited by mucins purified from chicken caecum and small and large intestines in vitro (4). In this study, the strongest inhibition of binding and invasion was found with O-glycosylated mucins from the large intestine followed by the small intestine and caecum, respectively. Following incubation with small and large intestinal mucins, C. jejuni showed a 6.5-fold decrease in binding to HCT-8 cells and a >1,000-fold and ∼150-fold decrease in internalization, respectively. Mucin from the chicken caecum only exhibited a fivefold reduction in internalization. The presence of chicken mucin had no effect in bacterial viability. When mucin O-glycans from each chicken intestinal section were oxidized with sodium metaperiodate, binding and internalization by HCT-8 cells returned to levels observed in the untreated controls. These results illustrate that the O-glycans expressed on the secreted intestinal mucins of chickens can inhibit colonization regiospecifically. A detailed structural characterization of chicken intestinal mucin glycans from these different sites may provide insights into what makes this species resistant to the pathology caused by this organism in humans. Therefore, we now report the mucin O-glycan structures from healthy chicken caecum, small intestine, and large intestine. O-glycan analysis by LC-MS identified structures that were not only unique to the different intestinal sections but also others that have never been identified in the human intestinal tract.  相似文献   

11.
Small-subunit ribosomal DNA (SSU rDNA) from 20 phenotypically distinct strains of 2,4-dichlorophenoxyacetic acid (2,4-D)-degrading bacteria was partially sequenced, yielding 18 unique strains belonging to members of the alpha, beta, and gamma subgroups of the class Proteobacteria. To understand the origin of 2,4-D degradation in this diverse collection, the first gene in the 2,4-D pathway, tfdA, was sequenced. The sequences fell into three unique classes found in various members of the beta and gamma subgroups of Proteobacteria. None of the α-Proteobacteria yielded tfdA PCR products. A comparison of the dendrogram of the tfdA genes with that of the SSU rDNA genes demonstrated incongruency in phylogenies, and hence 2,4-D degradation must have originated from gene transfer between species. Only those strains with tfdA sequences highly similar to the tfdA sequence of strain JMP134 (tfdA class I) transferred all the 2,4-D genes and conferred the 2,4-D degradation phenotype to a Burkholderia cepacia recipient.Bacteria capable of mineralizing 2,4-dichlorophenoxyacetic acid (2,4-D), a commonly used herbicide, are found in many different phylogenetic groups (2, 3, 7, 11, 22, 23). Evidence suggests that numerous variants of 2,4-D catabolic genes exist and that catabolic operons consist of a near-random mixing of these variants (7). Interspecies gene transfer is a well-documented phenomenon (13), and horizontal gene transfer of the 2,4-D-degrading plasmid pJP4 has been shown (3, 5). However, not all 2,4-D catabolic operons are found on plasmids (10, 11, 16, 20). The extent to which other 2,4-D genes have been exchanged in nature is unknown. The aim of this research was to assess the role of horizontal gene transfer in the evolution of 2,4-D-degrading strains. This article summarizes the results of two aspects of this work—the study of the transfer of the entire 2,4-D pathway by using standard mating experiments and a phylogenetic study of the tfdA gene. The tfdA gene codes for an α-ketoglutarate-dependent 2,4-D dioxygenase which converts 2,4-D into 2,4-dichlorophenol and glyoxylate (6). This 861-bp gene was first sequenced from Ralstonia eutropha JMP134 (19). Two more tfdA genes were cloned from chromosomal locations in Burkholderia strain RASC and Burkholderia strain TFD6 (16, 20). These proved to be identical to each other and 78.5% similar to the original. An alignment of the two variants allowed conserved areas to be identified and primers to be designed for the amplification of tfdA-like genes from other sources (24). Sequence analysis of putative tfdA fragments and the small-subunit ribosomal DNA (SSU rDNA) of the strains carrying them allowed us to construct phylogenies of the genes and their hosts and to look for congruency between them.

Mating experiments.

A collection of 2,4-D degraders containing 15 unique strains as determined by genomic fingerprinting (7) was used as a source of donors in a series of mating experiments (Table (Table1).1). Burkholderia cepacia D5, lacking the ability to grow on 2,4-D and not hybridizing to any tfd genes, was used as a recipient in mating experiments. Strain D5 contains neomycin phosphotransferase genes (nptII) carried on transposon Tn5 and is resistant to 50 μg each of kanamycin, carbenicillin, and bacitracin per ml. All of the 2,4-D strains used were sensitive to these antibiotics. Filter matings were performed with a donor-to-recipient ratio of 1:10. Colonies which grew on selective medium (500 ppm of 2,4-D in mineral salts agar [MMO] [23] including 50 μg of kanamycin, carbenicillin, and bacitracin per ml) were subjected to further tests. Their ability to catabolize 2,4-D was tested in liquid medium (same composition as that described above).

TABLE 1

2,4-D-degrading strains, geographic origins, and GenBank accession numbers
StrainGenBank accession no. (SSU rDNA)OriginMost similar to genus and/or speciesaTransferbtfdA typecGenBank accession no. (tfdA gene)Reference or source
JMP134AF049542AustraliaRalstonia eutropha+IM167303
EML1549AF049546OregonBurkholderia sp.+I2
TFD39AF049539SaskatchewanBurkholderia sp.+IU4319723
K712AF049543MichiganBurkholderia sp.+IU4327611
TFD9AF049537SaskatchewanAlcaligenes xylosoxidans+IU4327623
TFD41AF049541MichiganRalstonia eutropha+I23
TFD38AF049540MichiganRalstonia eutropha+NDc23
TFD23AF049536MichiganRhodoferax fermentans+IU4327623
RASCAF049544OregonBurkholderia sp.(+)IIU257172
TFD6AF049546MichiganBurkholderia sp.II23
TFD2AF049545MichiganBurkholderia sp.II23
TFD31AF049536SaskatchewanRhodoferax fermentansIII23
B6-9AF049538OntarioRhodoferax fermentansNDIIIU431969
I-18U22836OregonHalomonas sp.NDIIIU2249915
K1443AF049531MichiganSphingomonas sp.d11
2,4-D1AF049535MontanaSphingomonas sp.R. Sanford
B6-5AF049533OntarioSphingomonas sp.ND9
B6-10AF049534OntarioSphingomonas sp.ND9
EML146AF049532OregonSphingomonas sp.2
M1AF049530French PolynesiaRhodospeudomonas sp.NDR. Fulthorpe
Open in a separate windowaThe generus and/or species most similar to the strain is given based on similarities of SSU rDNA sequences. bSymbols: +, able to transfer 2,4-D degradation to B. cepacia D5; (+), able to transfer at very low frequency; −, no transfer detected. cND, not determined. d—, no amplificate was obtained. The disappearance of 2,4-D from the culture medium was monitored by high-performance liquid chromatography. Cells were removed by centrifugation, and the supernatant was filtered through 0.2-μm-pore-size filters. These samples were then analyzed on a Lichrosorb Rp-18 column (Anspec Co., Ann Arbor, Mich.) with 60% methanol–40% 0.1% H3PO4 as the eluant. 2,4-D was detected by measuring light absorption at 230 nm. The presence of tfd genes was detected by hybridizing colony blots with a DNA probe derived from the entire pJP4 plasmid. The identity of the colonies was confirmed by probing with the nptII gene of Tn5 (found in B. cepacia D5). Probes were labeled with random hexanucleotides incorporating [32P]dCTP (3,000 Ci/mmol; New England Nuclear, Boston, Mass.). Hybridizations were done under high-stringency conditions by using 50% formamide and Denhardt’s solution (18) at 42°C. Of the 15 unique strains tested, 9 transferred 2,4-D degradation abilities to D5. This transfer was confirmed by hybridization with pJP4 for eight of these strains. B. cepacia RASC could transfer degradative abilities, but neither it nor the transconjugant hybridized to the pJP4 probe. Work subsequent to this study has confirmed that the genes carried by RASC do not hybridize to those found on pJP4 under high-stringency conditions (7).

Phylogenetic analyses.

Total genomic DNA was isolated from 20 unique 2,4-D-degrading strains (including all 15 used for mating experiments) grown on 500 ppm of 2,4-D mineral salts medium amended with 50 ppm of yeast extract. SSU rDNA was amplified by using fD1 and rD1 as primers (25). Putative tfdA fragments were amplified by using primers TVU and TVL as previously described (24). PCR products were purified with a Gene Clean kit (Bio 101, La Jolla, Calif.). Sequencing was done with an Applied Biosystems model 373A automatic sequencer (Perkin-Elmer Cetus) by using fluorescently labeled dye termination at the Michigan State University Sequencing Facility. The sequencing primer used for SSU rDNA fragments was 519R (5′ GTA TTA CCG CGG CTG CTG G-3′). For tfdA fragments, the sequencing primers were the same as the amplification primers. GenBank accession numbers for these sequences are given in Table Table11.The SSU rDNA sequences were compared to sequences in GenBank by using the Basic Local Alignment Search Tool (BLAST) (1), and those strains with the highest maximal segment pair scores were retrieved from GenBank and included in the phylogenetic analysis. Sequences were aligned manually with the software SeqEd (Applied Biosystems) and with MacClade (14). Sites where nucleotides were not resolved for all sequences were deleted from the alignment, as were those nucleotides corresponding to the small loop in this region that is absent in the alpha subgroup of the class Proteobacteria. These deletions left 283 unambiguous sites for the construction of the SSU rDNA phylogenies. Phylogenetic trees were constructed by using the neighbor-joining analysis of pairwise Jukes-Cantor distances (4), and the topology was confirmed by using the maximum parsimony method PAUP (21). Desulfomonile tiedjei of the δ-Proteobacteria was used as an outgroup. Bootstrap analysis based on 100 replicates was used to place confidence estimates on the tree. Only bootstrap values of greater than 50 were used.

2,4-D degrader diversity.

The 2,4-D degraders in this study were distributed throughout the alpha, beta, and gamma subgroups of the Proteobacteria (Fig. (Fig.1).1). The lack of representation of gram-positive bacteria is likely a reflection of isolation methods, not of the lack of gram-positive 2,4-D degraders. The majority of these strains were members of the beta subgroup of Proteobacteria, five of which were most closely related to the genus Burkholderia, having at least 92% sequence similarity with each other. Three were closely related to Rhodoferax fermentans (close to the class Comamonadaceae), three were related to Ralstonia eutropha, and one was related to Alcaligenes xylosoxidans. TFD39 falls outside any clear cluster. One member of the γ-Proteobacteria, strain I-18, a haloalkaliphile, was found to be closely related to the salt-loving genus Halomonas (15). The remaining six strains all clustered in the alpha branch of Proteobacteria (Fig. (Fig.1).1). Of this subgroup, five were most closely related to the genus Sphingomonas. One member of the α-Proteobacteria, strain M1, which is the most oligotrophic and slow growing of all the strains used in this study, is 97% similar to Rhodopseudomonas palustris. The character of strain M1 correlates well with its phylogenetic placement near the slow-growing genus Bradyrhizobium. Open in a separate windowFIG. 1Neighbor-joining dendrogram (Jukes-Cantor distances) of SSU rDNA from 2,4-D-degrading bacteria (indicated in boldface type) and reference strains (indicated in italic type). Class I (•), class II (▴), and class III (■) types of tfdA genes are indicated. Bootstrap confidence limits (percentages) are indicated above each branch. Scale bar represents a Jukes-Cantor distance of 0.01.

tfdA gene fragments.

tfdA gene fragments were successfully amplified and sequenced from 10 strains of β-Proteobacteria and 1 strain of γ-Protobacteria. None of the strains from the α-Proteobacteria gave any amplificates with these primers. These 313 contiguous nucleotides were aligned with additional tfdA sequences from JMP134 and from strain RASC (Fig. (Fig.2).2). Three distinct classes of tfdA gene sequences with slight variations in each class were found. Class I included fragments from JMP134, TFD39, TFD23, K712, and TFD9 that differed from each other by 2 bp at the most. Class I tfdA genes are probably plasmid encoded. All strains with a class I tfdA gene examined so far contained broad-host-range, self-transmissible plasmids containing 2,4-D genes (2, 3, 11, 17). All of the strains with a class I tfdA gene were able to transfer the 2,4-D phenotype in the mating studies reported above. The class II tfdA sequences included identical fragments amplified from RASC, TFD6, and TFD2 which were 76% similar to those in class I. Class III included identical fragments from strains TFD31, B6-9, and I-18 which were 77% similar to class I genes and 80% similar to class II genes. Both class II and III tfdA genes differed from each other and from class I genes in the same nine sites corresponding to the third base pair of the codons. The tfdA phylogenetic tree is a simple one, with three distinct branches that are incongruent with the SSU rDNA-derived phylogeny (Fig. (Fig.3).3). Class I tfdA sequences were found in Burkholderia-like strains, in strains related to the Comamonas-Rhodoferax group, and in the Ralstonia-Acaligenes group, all in the β-Proteobacteria. Class II sequences are less widely distributed, found only in Burkholderia-like branches. However, even in this subgroup, this tfdA variant is found in strains that differ by 7% at the SSU rDNA level (RASC and TFD2). However, the class III sequences were most interesting, being found both in the Comamonas-Rhodoferax group and in a strain of the γ-Proteobacteria, I-18, strains that differ by 24% at the SSU rDNA level. Class III genes have since been found in a collection of randomly isolated non-2,4-D degraders, including gram-positive bacilli, as well as in various gram-negative bacteria, even though the gene is not expressed (10). Open in a separate windowFIG. 2Alignment of 313 nucleotides of internal fragments of tfdA genes from representative strains. Nucleotides identical to tfdA from pJP4 are represented by periods.Open in a separate windowFIG. 3Phylogenetic incongruency of tfdA genes and SSU rDNA from diverse 2,4-D-degrading bacteria. Dendrograms for tfdA and SSU rDNA are indicated. Shading indicates the type of tfdA sequence, either class I, II, or III. Note that branch lengths are not drawn to scale.An interesting result was the detection of two different tfdA gene variants in sibling strains. TFD23 and TFD31 are identical at the ribosomal gene level, but one harbors a class I gene and the other harbors a class III gene. Similarly, TFD6 and EML159 are rRNA siblings that carry a class II and class I gene, respectively.None of the α-Proteobacteria yielded a PCR product when amplified with the conserved tfdA primers. This finding complements our observation that none of these bacteria hybridized to the tfdA gene, even under conditions of low stringency, indicating that any tfdA-like genes in the α-Proteobacteria are likely to be more divergent from the ones sequenced here (7, 11). In addition, none of the Sphingomonas strains in the study hybridized with a whole pJP4 probe, and similarly, no Sphingomonas strains scored positive for transfer of 2,4-D-degrading ability to recipient B. cepacia D5. Together these results suggest a reduced gene flow between members of the α- and β- or γ-Proteobacteria or poor gene expression of β- or γ-derived genes by α-Proteobacteria. Although plasmid pJP4 is a broad-host-range plasmid and has been known to transfer to α-Proteobacteria such as Rhizobium and Agrobacterium species and to γ-Proteobacteria such as Pseudomonas putida, Pseudomonas fluorescens, and Pseudomonas aeruginosa, the 2,4-D pathway is not expressed in these strains of the α- or γ-Proteobacteria (3). Phylogenetically limited expression of plasmid-borne 3-chlorobenzoate-degradative genes has also been noted for the pseudomonads (8). Subsequent studies have found divergent but related sequences for the tfdB and tfdC genes in 2,4-D-degrading Sphingomonas strains (7, 12, 24).With the exceptions of the minor differences within the class I pJP4-like tfdA sequences, there were no intermediate tfdA sequences. The most likely explanation of this is that the rate of horizontal transfer of the tfd genes is high relative to the rate at which mutations can accumulate. Examination of sequences of tfdA genes from a greater variety of organisms may turn up more intermediate variation.  相似文献   

12.
13.
14.
15.
16.
The site-specific recombinase IntI1 found in class 1 integrons catalyzes the excision and integration of mobile gene cassettes, especially antibiotic resistance gene cassettes, with a site-specific recombination system. The integron integrase belongs to the tyrosine recombinase (phage integrase) family. The members of this family, exemplified by the lambda integrase, do not share extensive amino acid identities, but three invariant residues are found within two regions, designated box I and box II. Two conserved residues are arginines, one located in box I and one in box II, while the other conserved residue is a tyrosine located at the C terminus of box II. We have analyzed the properties of IntI1 variants carrying point mutations at the three conserved residues of the family in in vivo recombination and in vitro substrate binding. We have made four proteins with mutations of the conserved box I arginine (R146) and three mutants with changes of the box II arginine (R280); of these, MBP-IntI1(R146K) and MBP-IntI1(R280K) bind to the attI1 site in vitro, but only MBP-IntI1(R280K) is able to excise cassettes in vivo. However, the efficiency of recombination and DNA binding for MBP-IntI1(R280K) is lower than that obtained with the wild-type MBP-IntI1. We have also made two proteins with mutations of the tyrosine residue (Y312), and both mutant proteins are similar to the wild-type fusion protein in their DNA-binding capacity but are unable to catalyze in vivo recombination.Integrons are DNA elements that capture genes, especially antibiotic resistance genes, by a site-specific recombination system (32). The recombination system consists of a DNA integrase (Int) and two types of recombination sites, attI and attC (59-base element). The integrase gene (int) is located in the 5′ conserved segment of the integron structure (Fig. (Fig.1)1) and is a member of the tyrosine recombinase family (1, 4, 13, 23, 24). Three types of integrases, sharing around 50% identity among themselves, have been identified; they define integron classes 1, 2, and 3 (30). The 5′ conserved segment found in class 1 integrons also contains a promoter region responsible for the expression of inserted cassettes (11, 21) and the recombination site attI1 (31). The 3′ conserved segment of the class 1 integrons includes an ethidium bromide resistance determinant (qacEΔ1), a sulfonamide resistance gene (sulI), an open reading frame (ORF5) of unknown function, and further sequences that differ from one integron to another (5, 6, 28). The 3′ conserved segment of class 2 integrons includes transposition genes (20) while that of class 3 integrons has not yet been studied (2). The variable region, located between the two conserved segments, usually contains antibiotic resistance genes; In0 contains no inserted genes while In21 possesses eight cassettes with ten genes (or ORFs) in this region (5, 16). These genes are part of mobile cassettes which include a recombination site, attC, that differs from one gene to another (18, 33). Incoming genes must be associated with an attC to be recognized by the integron integrase and are preferentially inserted at the recombination site attI1 (11). Cassettes are excised as circular intermediates and integrated at core sites by the action of the integrase (810). The core site, defined as GTTRRRY, makes up the 3′ end of attI1 and attC, with the crossover taking place between the G and the first T (19). Antibiotic selection pressure can reveal cassette rearrangements in which a given resistance is nearest the promoter and thus most strongly expressed (10). Open in a separate windowFIG. 1General structure of class 1 integrons. Cassettes are inserted in the integron variable region by a site-specific recombination mechanism. The attI1 site is shown by a black circle, core sites are represented by ovals, the attC site is indicated by a black rectangle, and promoters are denoted by P. intIl, integrase gene; qacEΔ1, antiseptic resistance gene; sulI, sulfonamide resistance gene; orf5, gene of unknown function.Site-specific recombination, unlike homologous recombination, is characterized by relatively short, specific DNA sequences and requires only limited homology of the recombining partners (12). Site-specific recombination is an entirely conservative process since all DNA strands that are broken (two per exchange site) are rejoined in a process that involves neither ATP nor DNA synthesis. Homology alignments of site-specific recombinases assign them to two families: the resolvase family, named after the TnpR proteins encoded by the transposons γδ and Tn3, and the integrase family. The integrase family includes over 140 members to date, but they are highly diversified proteins (13, 23). Members of this family, which include the well-studied λ integrase, recombine DNA duplexes by executing two consecutive strand breakage and rejoining steps and a topoisomerization of the reactants. The first pair of exchanges form a four-way Holliday junction and the second pair resolve the junction to complete the recombination. The integrase nucleophile is a conserved tyrosine that becomes associated with a phosphate group on DNA. The cleavage sites on each DNA duplex are separated by 6 to 8 bp with a 5′ stagger, and the tyrosine joins to the 3′ phosphate (17).The initial definition of the integrase family was based on comparisons of seven sequences, and three invariant residues were identified: an HXXR cluster and a Y residue (4). Alignment of 28 sequences identified a fourth invariant position, occupied by an arginine residue (1). These four conserved residues are found in two boxes located in the second half of the protein. A recent analysis has shown that the conserved histidine is present in 136 of the 147 members (93%); this residue is then not conserved in all members of the family (13). Another recent analysis has identified three patches of residues located around box I, which seem to be important in the secondary structure of these proteins (23). In this study, we analyzed the properties of several mutants of the conserved residues R146, R280, and Y312 of the integron integrase IntI1 in in vivo recombination and in vitro substrate binding.

Construction of plasmids overexpressing mutant MBP-IntI1 fusion proteins.

The plasmids encoding various mutants of MBP-IntI1 were constructed by PCR using pLQ369 (50 ng) as a template (15). Two primer pairs, designed with the OLIGO software package (version 4.1; National Biosciences, Plymouth, Minn.), were used to construct each set of mutants. The R146 mutants were constructed with an XcmI-BamHI primer pair [IntI1(R146)-XcmI, 5′-TTCACCAGCTTCTGTATGGAACGGGCATG(A/G)(A/T)AATCAG-3′; IntI1(R146)-BamHI, 5′-CCGGATCCCTACCTCTCACT-3′], the R280 mutants were constructed with an NruI-XmnI primer pair [IntI1(R280)-NruI, 5′-AGCCGTCGCGAACGAGTGC(C/T)(C/T)GAGGG-3′; IntI1(R280)-XmnI, 5′-ACCCCTAATGAAGTGGTTCGTATCC-3′], and the Y312 mutants were constructed with a AatII-ScaI primer pair [IntI1(Y312)-AatII, 5′-ATTCCGACGTCTCTACTACGATGATTT(C/T)CACGC-3′; pLQ369-ScaI, 5′-ATGCTTTTCTGTGACTGGTG-3′] (restriction sites within primer sequences are underlined). PCR conditions were 10 min at 94°C, three cycles consisting of 45 s at 94°C, 45 s at 47°C, and 90 s at 72°C, 30 cycles consisting of 45 s at 94°C, 45 s at 60°C (50°C for Y312 mutants), and 90 s at 72°C, and a final elongation step of 10 min at 72°C. The XcmI, NruI, and AatII primers were degenerate in one or two positions, so that a single primer could give all mutants. Mutant PCR fragments were digested and cloned directly into pLQ369 digested with the same enzymes, except for the R146 mutant fragments that were subcloned into pLQ364 at first. New mutant PCR fragments were then amplified on these subclones, using IntI1(R146)-BamHI and IntI1(R280)-XmnI primers. These mutant PCR fragments were cleaved with BamHI and XmnI, and the resulting fragments were cloned into pLQ369. This avoids the necessity of partial digestion of pLQ369 with XcmI. Mutant clones were digested with restriction endonucleases and sequenced to determine the mutation.

In vivo recombination.

Mutant MBP-IntI1 clones were introduced into Escherichia coli TB1 {F′ araΔ(lac-proAB) rpsL (Strr) [φ80dlacΔ(lacZ)M15] hsdR(rKmK)} containing pLQ428 by transformation (Fig. (Fig.22 and Table Table1).1). E. coli TB1 cells containing pLQ428 and one of the MBP-IntI1 mutants were grown at 37°C for 3 h in Luria-Bertani medium. Excision of the aacA1-ORFG and/or ORFH cassettes was induced by the overexpression of the malE-intI1 gene by using 0.3 mM isopropyl-β-d-thiogalactopyranoside (IPTG; Sigma Chemical Co.) and by incubation at 37°C for another 3 h. Cell culture was done in the presence of 50 μg of ampicillin per ml, 15 μg of amikacin per ml, and 50 μg of chloramphenicol per ml. Plasmid DNA was then prepared from 5-ml cultures with the Perfect Prep DNA extraction kit (Mandel Corporation). In order to determine the capacity of mutant MBP-IntI1 proteins to excise aacA1-ORFG and/or ORFH cassettes of In21, we used PCR primers pACYC184-5′ (5′-TGTAGCACCTGAAGTCAGCC-3′) and pACYC184-3′ (5′-ATACCCACGCCGAAACAAG-3′) (Fig. (Fig.2,2, primers 1 and 2) to detect the reduction of pLQ428 length. PCR conditions were 10 min at 94°C, 30 cycles consisting of 1 min at 94°C, 1 min at 60°C, and 5 min at 72°C, and a final elongation step of 10 min at 72°C. A major PCR fragment can be seen in each lane containing a DNA preparation from a mutant clone (Fig. (Fig.3,3, lanes 2 to 9). This band is 2,499 bp long and, as determined by restriction enzyme digestions, represents the pLQ428 clone without any cassette excision (data not shown). This band is also observed in the negative control, which is the pMAL-c2 vector without any gene fused to malE (Fig. (Fig.3,3, lane 12). Open in a separate windowFIG. 2Representation of plasmids used in this study. The positions of the three invariant residues of the integrase family are indicated, along with restriction sites used to construct mutant proteins. Core sites are represented by black circles, and attCs are shown by white boxes. The numbered arrows represent the PCR primers used to detect excision events, pACYC184-5′ (1) and pACYC184-3′ (2). bla, gene encoding β-lactamase; cat, gene encoding chloramphenicol acetyltransferase; intIl, gene encoding the integron integrase (IntI1); malE, gene encoding the maltose binding protein (MBP); ori, origin of replication; Ptac, tac promoter; Ptet, tetracycline promoter. Only relevant restriction sites are indicated.

TABLE 1

Plasmids used in this study
PlasmidCharacteristic(s)aReference or source
pLQ3632,190-bp EcoRI-HincII fragment of pLQ161 cloned in pLQ402 (Apr)16
pLQ3641,027-bp NcoI-BamHI PCR fragment amplified on pLQ860 and cloned in pET-3d (Apr)This study
pLQ3691,019-bp NdeI-BamHI PCR fragment modified to create a blunt-end 5′-ATG and cloned in pMAL-c2 cut with XmnI-BamHI (Apr)15
pLQ376pLQ369 MBP-IntI1(R146K) (Apr)This study
pLQ377pLQ369 MBP-IntI1(R146E) (Apr)This study
pLQ378pLQ369 MBP-IntI1(R146I) (Apr)This study
pLQ379pLQ369 MBP-IntI1(R146V) (Apr)This study
pLQ388pLQ369 MBP-IntI1(R280G) (Apr)This study
pLQ390pLQ369 MBP-IntI1(R280E) (Apr)This study
pLQ391pLQ369 MBP-IntI1(R280K) (Apr)This study
pLQ393pLQ369 MBP-IntI1(Y312S) (Apr)This study
pLQ394pLQ369 MBP-IntI1(Y312F) (Apr)This study
pLQ4282,133-bp EcoRI (filled in)-BglII fragment of pLQ363 cloned in pACYC184 cut with EcoRV-BamHI (Akr Cmr)This study
pLQ8602,900-bp BamHI fragment of pVS1 cloned in pTZ19R (Apr Sulr)5
Open in a separate windowaAkr, Apr, and Cmr, resistance to amikacin, ampicillin, and chloramphenicol. Open in a separate windowFIG. 3Electrophoresis of PCR products obtained with the pACYC184 primer pair and 100 ng of DNA preparations from overexpressed cultures on a 1% agarose gel. Lane 1, 1-kb DNA ladder (Gibco BRL); lane 2, DNA preparation of pLQ428-pLQ377 (R146E); lane 3, pLQ428-pLQ378 (R146I); lane 4, pLQ428-pLQ376 (R146K); lane 5, pLQ428-pLQ379 (R146V); lane 6, pLQ428-pLQ390 (R280E); lane 7, pLQ428-pLQ388 (R280G); lane 8, pLQ428-pLQ391 (R280K); lane 9, pLQ428-pLQ394 (Y312F); lane 10, pLQ428-pLQ393 (Y312S); lane 11, pLQ428-pLQ369 (wild type); lane 12, pLQ428-pMAL-c2 (MBP).The 2,499-bp PCR product was not obtained in the reaction containing the wild-type MBP-IntI1-expressing clone pLQ369 (Fig. (Fig.3,3, lane 11), indicating that there were no remaining full-length pLQ428 molecules. This shows that the wild-type fusion protein is very efficient in site-specific recombination and that all pLQ428 clones have undergone an excision of one or both cassettes. In this PCR, we observed two major bands of 1,341 and 889 bp. The 1,341-bp PCR product was digested with restriction enzymes to show that it represents a pLQ428 clone which has lost the aacA1-ORFG cassette (data not shown). The 889-bp band was also digested with restriction enzymes to show that it represents a pLQ428 clone which has lost both aacA1-ORFG and ORFH cassettes (data not shown). These two PCR products are also observed in the reaction containing the mutant clone pLQ391, which expresses the MBP-IntI1(R280K) fusion protein. This mutant protein is, however, less efficient than the wild-type protein, as seen by the intensity of the PCR products (Fig. (Fig.3,3, lane 8). We were not able to detect a PCR product of 2,047 bp, corresponding to the excision of the ORFH cassette alone; this is not surprising since this event has been shown in another study to be rare (16). It is possible to observe another band in pLQ428-pLQ391 (R280K) and pLQ428-pLQ369 (wild type) PCRs (Fig. (Fig.3,3, lanes 8 and 11); this PCR product is 1,100 bp long and probably represents a recombination event at a secondary site. Restriction enzyme digestions were done on this product, but we were unable to identify its origin. This product results from an event mediated by the integron integrase since it is seen only in reactions containing active proteins. An 1,800-bp PCR band is also present in the negative control and in all PCRs containing a mutant clone. This product appears to be nonspecific, and the fact that it is not seen in the PCR containing the pLQ428-pLQ369 (wild-type) clones probably results from the PCR being more favorable to smaller PCR products.

In vitro substrate binding.

The experiments described above demonstrate that only one of our mutants of IntI1 protein is able to catalyze in vivo recombination. Can all mutant proteins recognize and bind to the IntI1 recombination site in a manner similar to the wild-type protein? To investigate this question, we used purified fusion proteins and a gel retardation assay with the complete attI1 site (5′ site) of the integron. MBP-IntI1 fusion proteins were purified as suggested by New England Biolabs. The concentration of the purified fusion protein was determined by using the Bradford protein assay (Bio-Rad). The protein solution was then made 20% in glycerol and stored at −80°C. The purity of MBP-IntI1 was evaluated as >90% by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (data not shown). Binding reactions were done with labeled 5′-site DNA fragments (20,000 cpm, 0.25 pmol), incubated with different concentrations of MBP-IntI1 in a 10-μl volume containing 10 mM HEPES (K+, pH 8.0), 60 mM KCl, 4 mM MgCl2, 100 μM EDTA (pH 8.0), 100 μg of bovine serum albumin per ml, 250 μM dithiothreitol, 100 ng of poly(dI-dC), and 10% glycerol. Reaction mixtures were incubated at room temperature for 15 min prior to electrophoresis through 4 or 5% prerun, nondenaturing polyacrylamide gels buffered with 0.5× Tris-borate-EDTA. Dried gels were subjected to autoradiography. The wild-type fusion protein and native IntI1 were shown to lead to the same four distinct complexes (I, II, III, and IV) with this DNA substrate (Fig. (Fig.4)4) (15). These complexes represent the binding of four IntI1 molecules to four different sites in the attI1 site (15). Figure Figure44 shows results obtained with nine mutants of the MBP-IntI1 fusion protein. We observed that MBP-IntI1(R146E), MBP-IntI1(R146I), and MBP-IntI1(R146V) lost their ability to bind to the attI1 site, as no complexes are seen in the gel retardation experiment (Fig. (Fig.4A).4A). However, MBP-IntI1(R146K) formed four IntI1-DNA complexes with the 5′ site DNA fragment. The band pattern and the intensity observed with this mutant protein are similar to those observed with the wild-type protein, suggesting that MBP-IntI1(R146K) and MBP-IntI1 bind DNA with similar affinities. Open in a separate windowFIG. 4Binding of mutant MBP-IntI1 fusion proteins purified from E. coli TB1 to the 5′-site DNA fragment containing the complete attI1 site of the In2 integron (from nucleotide −96 to nucleotide +71, relative to the G residue of the core site as position 0). (A) MBP-IntI1(R146) mutants; (B) MBP-IntI1(R280) mutants; (C) MBP-IntI1(Y312) mutants. A purified labeled fragment was incubated with different concentrations of mutant fusion proteins. Free DNA (F) and protein-DNA complexes (I, II, III, and IV) were separated on 4 or 5% polyacrylamide gels and are indicated by arrows. Lanes 1, free DNA; lanes 2 through 7, purified fusion protein at 250, 375, 500, 12.5, 37.5, and 62.5 nM, respectively. The wild-type (WT) lanes in panel C were from a separate gel.Competition with a specific fragment with a 30-fold excess of unlabeled DNA competed away all four complexes, while a 100-fold excess of a nonspecific unlabeled DNA fragment did not compete away any complexes, indicating their specificity (data not shown) (15). We observed that MBP-IntI1(R280G) and MBP-IntI1(R280E) lost their ability to bind the 5′-site DNA fragment, while the MBP-IntI1(R280K) could still bind the attI1 site (Fig. (Fig.4B).4B). However, the band pattern obtained with this mutant protein is weaker than that obtained with the wild-type integrase. For example, at a protein concentration of 250 nM MBP-IntI1(R280K) (lane 2), we observed the formation of complexes I, II, and III, with a stronger intensity for the fastest-migrating complexes, while the intensity of the fourth complex was very weak. At the same concentration of the wild-type protein, we observed the formation of all four complexes, with a stronger intensity for the slowest-migrating complexes and no unbound DNA. These results show that MBP-IntI1(R280K) binds the attI1 site with a lower affinity than the wild-type fusion protein. As shown in Fig. Fig.4C,4C, both MBP-IntI1(Y312F) and MBP-IntI1(Y312S) lead to the formation of four complexes that migrate similarity to those obtained with wild-type MBP-IntI1, as judged by the gel migration of these complexes. The band pattern observed shows that the binding affinity of these mutant proteins is the same as or even better than that of the wild-type protein.

Relationships with other members of the family.

We found that MBP-IntI1 recombinase in which Arg-146 has been changed to lysine [MBP-IntI1(R146K)] by PCR mutagenesis cannot excise cassettes but can bind to the attI1 site with the same efficiency as the wild-type fusion protein. However, MBP-IntI1(R146I), MBP-IntI1(R146E), and MBP-IntI1(R146V) mutant proteins have completely lost both phenotypes. These findings are different from those for other members of the family. The only mutant protein of the lambda integrase at this residue [λ(R212Q)] binds the core site partially and is not able to catalyze in vivo or in vitro recombination (22). Mutants of the Cre recombinase with a change at this residue [Cre(R173K)] bind DNA as well as the wild-type protein but cannot catalyze in vivo or in vitro recombination (1). Mutants of Flp [Flp(R191K) and Flp(R191E)] bind FRT recombination sites as well as the wild-type protein but cannot carry out in vivo or in vitro recombination, except for the Flp(R191K) protein, which has shown slight activity in in vivo recombination (Table (Table2)2) (7, 14, 25). Therefore, the Cre(R173K) and Flp(R146K) mutants have the same phenotype as the MBP-IntI1(R146K) protein. However, the Flp(R191E) mutant protein shows efficient DNA binding while MBP-IntI1(R146E) does not bind to the attI1 site. We interpret these results according to the charge of the Arg-146 residue. The positively charged side chain of this residue makes contact with the DNA, which is negatively charged. This contact is probably important for the good conformation of the protein molecule in positioning the tyrosine residue to perform recombination. When this residue is exchanged for a lysine, DNA contacts are still able to take place because of the charge of the residue, but the side chain is smaller and the lysine is probably not able to position the tyrosine to catalyze recombination. We think that the charge of this residue is very important in the formation of DNA-protein complexes in the integron system, since all other MBP-IntI1 mutants tested are unable to bind DNA. This observation differs from those for Flp, because even when the wild-type residue was replaced by a negatively charged one, it could still bind DNA as well as the wild-type protein (Table (Table2).2).

TABLE 2

Mutational analysis of IntI1 and corresponding residues of other recombinases from the Int family
RecombinaseMutationDNA bindingRecombinationReference(s)
λIntR212QYesaNo22
λIntY342FYesNo22, 26
FlpR191EYesNo7
FlpR191KYesYes7, 14
FlpR308GYesNo27
FlpR308KYesYesa27
FlpY343FYesNo29
FlpY343SYesNo29
CreR173KYesNo1
P2R272KNDbNo23
XerCY275FYesNo3
XerDY279FYesNo3
IntI1R146ENoNoThis study
IntI1R146INoNoThis study
IntI1R146KYesNoThis study
IntI1R146VNoNoThis study
IntI1R280ENoNoThis study
IntI1R280GNoNoThis study
IntI1R280KYesYesaThis study
IntI1Y312FYesNoThis study
IntI1Y312SYesNoThis study
Open in a separate windowaLess efficient than the wild-type protein. bND, not determined. We have also made proteins with mutations at position 280; these were MBP-IntI1(R280E), MBP-IntI1(R280G), and MBP-IntI1(R280K). We found that the MBP-IntI1(R280K) mutant protein binds the attI1 site and excises integron cassettes with a lower efficiency than the wild-type MBP-IntI1, while MBP-IntI1(R280E) and MBP-IntI1(R280G) have completely lost both phenotypes. The Flp(R308K) mutant protein has been shown to bind DNA as well as the wild-type protein, but it recombines DNA with a lower efficiency than wild-type Flp (27). Another mutant protein of Flp [Flp(R308G)] has also been shown to bind DNA as well as the wild-type protein, but it was unable to catalyze in vivo or in vitro recombination (27). These results show that Flp(R308K) and MBP-IntI1(R280K) act similarly but that the other Flp mutant [Flp(R308G)] can bind DNA while the MBP-IntI1 mutant [MBP-IntI1(R280G)] cannot (Table (Table2).2). We also think that the positive charge of this residue is important for the binding of the recombinase to DNA, but Arg-280 does not seem to be implicated in the positioning of the tyrosine residue, since the MBP-IntI1(R280K) mutant protein can perform recombination.We found that MBP-IntI1(Y312S) and MBP-IntI1(Y312F) mutant proteins bind the attI1 site with the same efficiency as the wild-type protein but are not able to catalyze in vivo recombination. As expected, these results are the same as those obtained with the lambda integrase [λ(Y342F)], the XerC and XerD recombinases [XerC(Y275F) and XerD(Y279F)], and the Flp recombinases [Flp(Y343S) and Flp(Y343F)] (Table (Table2)2) (3, 22, 26, 29). The loss of the catalytic activity of the MBP-IntI1(Y312F) mutant protein is not surprising, since the hydroxyl group of the tyrosine, which is responsible for the nucleophilic attack of the DNA at the recombination site, is not present on the phenylalanine residue. The phenotype of MBP-IntI1(Y312S) indicates that the conformation of the tyrosine residue is important for the good activity of the recombinase, because even if the serine residue has a hydroxyl group, it is not able to catalyze recombination. These results indicate that the integron integrase IntI1 uses the hydroxyl group of the conserved tyrosine (Y312) to catalyze site-specific recombination, like other members of the family. However, in vitro recombination using this mutant protein needs to be done to confirm this.These results of point mutations show that mutations of the conserved arginines by nonpositively charged residues abolish substrate recognition, unlike the corresponding mutants of other members of the family. However, further mutational analysis, such as of residues around and in patch III, would be interesting, since only integron integrases contain more residues in this region than other members of the family (23). In vitro recombination assays with purified mutant proteins also need to be done in order to study thoroughly the mechanism of site-specific recombination in integrons.  相似文献   

17.
18.
Heterosexual transmission of a murine leukemia virus mixture named LP-BM5 MuLV, which is known as the murine AIDS virus, was investigated. Our results indicated that the heterosexual transmission of LP-BM5 MuLV occurs in both directions with high frequency and that the frequencies of virus transmission in the cervix and penis are higher than those in other genital organs. The results suggested that infection by LP-BM5 MuLV via heterosexual transmission may initially take place at particular retrovirus-sensitive sites (cells) in the genital organs.Human immunodeficiency virus (HIV) infection is now pandemic. In many countries, HIV has been spread mainly by heterosexual transmission (3, 5). For the prevention of HIV infection, as well as for the development of vaccines against HIV, it is of a great importance to understand the mechanisms of the heterosexual transmission of retroviruses. Since it is difficult to investigate the mechanisms of heterosexual transmission of HIV in humans experimentally, an animal model with a retrovirus which induces an acquired immunodeficiency syndrome like human AIDS would be useful. A murine leukemia virus mixture called LP-BM5 MuLV induces a severe acquired immunodeficiency syndrome termed murine AIDS (MAIDS) in susceptible strains of mice (10). The mixture includes a replication-competent ecotropic virus, mink cell focus-inducing virus, and a replication-defective virus which has been considered to be involved in the pathogenesis of MAIDS (4). With many similarities to human AIDS patients, mice infected with the LP-BM5 MuLV mixture develop splenomegaly, systemic lymphadenopathy, and severe immunodeficiency (4, 11). We previously reported that maternal transmission of LP-BM5 MuLV occurs via mother’s milk with high frequency (12). In the present study, we demonstrate that the heterosexual transmission of LP-BM5 MuLV also occurs with high frequency via genital organs.C57BL/10 (B10) mice were purchased from Japan SLC Inc., Shizuoka, Japan. All mice were specific-pathogen free and were housed in an air-conditioned room. They were given autoclaved water and sterilized pelleted feed. An SC-1 clone chronically infected with LP-BM5 MuLV, the G6 cell line, was kindly supplied by H. C. Morse III, National Institutes of Health, Bethesda, Md. Virus was prepared from the supernatant of G6 cells as previously described (12). The virus preparation was stored at −70°C until use. B10 mice were inoculated by the intraperitoneal route with 0.3 ml of the LP-BM5 MuLV preparation. To increase the frequency of sexual contacts and to avoid pregnancy in the female mice, all male mice were sterilized by vasectomy under anesthesia with pentobarbital (Nembutal). The vasectomized male mice were mated with female mice at least 4 weeks postoperation, since sperm are usually kept alive for 2 to 3 weeks in spermiducts. Excised genital organs were crushed with plastic sticks in 1 ml of lysis buffer containing 10 mM Tris-HCl (pH 8.0), 100 mM NaCl, 1 mM EDTA, 0.5% sodium dodecyl sulfate, and proteinase K (0.5 mg/ml). Spleen cells were lysed after hemolysis with 0.83% NH4Cl. Lysed samples were incubated at 50°C for 3 h. DNA was extracted three times with phenol-chloroform, precipitated with cold ethanol, treated with RNase and proteinase K, and dissolved in 0.1 ml of H2O. LP-BM5 MuLV defective virus genome was detected by Southern blot hybridization combined with PCR as described previously (12). In brief, template DNAs (1 μg per tube) were added to a cocktail adjusted to final concentrations of 10 mM Tris-HCl (pH 8.3), 50 mM KCl, 1.5 mM MgCl2, 0.01% gelatin, 200 μM deoxynucleoside triphosphate, 100 pmol of each primer (5′-CCTCTTCCTTTATCGACACT-3′ [sense] and 5′-ATTAGGGGGGGAATAGCTCG-3′ [antisense]), and 2 U of Taq DNA polymerase (Boehringer Mannheim) in a total volume of 100 μl and were subjected to 32 cycles of amplification. In each cycle of PCR, the mixture was denatured at 95°C for 1 min (5 min for the first cycle), annealed at 55°C for 3 min, and extended at 72°C for 1 min. The PCR-amplified products were subjected to gel electrophoresis (1.5% agarose) and transferred to a Hybond N+ membrane (Amersham) by the alkaline blotting method. Hybridization was achieved with a 5′ 32P-labeled probe (5′-TGTCAAAGGGACCAGTTAAG-3′) at 45°C overnight in 6× SSC (1× SSC is 0.15 M NaCl plus 0.015 M sodium citrate)–0.5% sodium dodecyl sulfate–100 μg of salmon sperm DNA per ml. Hybridized membranes were washed twice in 2× SSC at 37°C for 10 min and then in 0.5× SSC at 45°C for 30 min. DNA derived from uterine cervices of uninfected mice was used as a negative control. The limit of sensitivity was approximately 10 copies per tube, as assessed by Southern blot analysis with plasmid DNAs (1/10 of the PCR product).Concanavalin A (ConA) was obtained from Pharmacia Fine Chemicals, Uppsala, Sweden. Responder spleen cells (2 × 105) were cultured with ConA (5 μg/ml) in 96-well flat-bottomed microculture plates in 0.2 ml of culture medium at 37°C in 7.5% CO2. The culture medium consisted of RPMI 1640 supplemented with 10% fetal calf serum, penicillin (5,000 IU/100 ml), streptomycin (5,000 μg/100 ml), nonessential amino acids, sodium pyruvate (11.0 mg/100 ml), 2-mercaptoethanol (5 × 10−5 M), and l-glutamine (29.2 mg/100 ml). On day 2, cultures were pulsed with 1 μCi of [3H]thymidine and incubated for an additional 12 to 18 h. Incorporation of [3H]thymidine into responder spleen cells was quantitated by liquid scintillation counting. Determinations were performed in triplicate; standard errors of the means were generally <5% and therefore have not been indicated.As illustrated in Fig. Fig.1,1, in order to investigate the heterosexual transmission of LP-BM5 MuLV from male to female mice, normal male mice were inoculated with LP-BM5 MuLV and vasectomized 1 week later. At 5 weeks after virus inoculation, they were mated with uninfected female mice. After 8 weeks of breeding, female mice were sacrificed and their vaginae, cervices uteri, corpora uteri, inguinal lymph nodes, and spleens were removed and stored at −70°C until use. In the opposite direction, to investigate virus transmission from female to male, normal female mice were inoculated with LP-BM5 MuLV and then mated with uninfected, vasectomized male mice as described above. After 8 weeks of breeding, male mice were sacrificed and their penes, prepuces, inguinal lymph nodes and spleens were removed and stored at −70°C until use. Figure Figure22 shows the detection by PCR of the LP-BM5 defective virus genome in genital organs and spleens that were taken from mice mated with their virus-infected counterparts. It was demonstrated that although the defective virus genome was detected in both spleens and genital organs in some male mice (2 of 17 [see Table Table1]),1]), as shown in Fig. Fig.2,2, lanes 3 and 4, the defective virus genome was detected only in the genital organs, not the spleens (Fig. (Fig.2,2, lanes 5 and 6), from most of the male mice. In contrast, all of the female mice were positive for defective virus genome only in the genital organs (Fig. (Fig.2,2, lanes 1 and 2). None of the mice examined were positive for the virus genome only in the spleens (this issue is discussed below). It should be noted here that the efficacy of PCR amplification, which was measured by experiments using the mixture of genomic DNA and plasmid DNA containing the defective virus, did not differ among the genital organs and spleens. By using the above strategy, the heterosexual transmission of LP-BM5 MuLV was investigated according to the protocol shown in Fig. Fig.1.1. Open in a separate windowFIG. 1Experimental design for examination of heterosexual transmission of the MAIDS virus in B10 mice. i.p., intraperitoneal.Open in a separate windowFIG. 2Detection of the LP-BM5 MuLV defective virus genome by PCR in genital organs and spleens. The template DNAs (1 μg) derived from female or male mice which were bred with LP-BM5 MuLV-infected mice were amplified by PCR. Samples were prepared from either female (lanes 1 and 2) or male (lanes 3 to 6) mice. Lanes 1, 3, and 5, spleen; lane 2, uterine cervix; lanes 4 and 6, penis (from two representative male mice). The PCR products (5 μl) were applied to a 1.5% agarose gel and analyzed by Southern blotting with a probe for the defective virus (12).

TABLE 1

Heterosexual transmission of LP-BM5 MuLV
ExptClinical condition
Detection of LP-BM5 MuLV (no. positive/total [%])
MaleFemaleSpleenInguinal lymph nodeCervixCorpusVaginaPenisPrepuce
1MAIDSNormal0/25 (0)0/16 (0)9/25 (36)NDaND
2MAIDSNormal0/11 (0)ND3/11 (27)0/11 (0)1/11 (9)
3NormalMAIDS1/8 (12)3/8 (38)6/8 (75)0/8 (0)
4NormalMAIDS1/9 (11)ND5/9 (56)1/9 (11)
Open in a separate windowaND, not done. Twenty-five female mice that were mated with the virus-infected male mice were analyzed for the presence of LP-BM5 defective genome in their genital organs, lymph nodes, and spleens. As summarized in Table Table1,1, the defective virus genome was detected with high frequency in cervices (9 of 25). However, the defective virus genome was not detected in spleens at all (0 of 25). The female genital organs are divided into three parts, namely, the vagina, cervix of uterus, and corpus of uterus. As also shown in Table Table1,1, the cervix appears to be more sensitive to virus infection than the other organs. Since MAIDS virus was not detected in castrated female mice, which were kept with virus-infected male mice in the same cage, the virus infection occurred via heterosexual transmission rather than by nonheterosexual horizontal transmission (data not shown). In 17 male mice mated with the virus-infected female mice (Table (Table1),1), the defective virus genome was detected in penes with high frequency (11 of 17). The defective virus genome was detected in DNA prepared from spleens with much lower frequency (2 of 17). In male mice, the penis seems to be much more sensitive to virus infection than are the prepuce and spleen (Table (Table1).1). In experiments 1 and 3, we also examined the inguinal lymph nodes from 16 female mice and 8 male mice. The defective virus genome was detected in some of the male mice (3 of 8) but not at all in the female mice examined. These results suggest that the LP-BM5 MuLV mixture initially infects the cervix or penis and then spreads over the whole body, including the lymph nodes and spleen.To determine whether mice infected with LP-BM5 MuLV by heterosexual transmission in fact develop MAIDS, we examined both spleen weights and mitogen (ConA) responses of female mice at 10 months after mating. As shown in Table Table2,2, female mice which were infected with LP-BM5 MuLV by heterosexual transmission (i.e., the defective virus genome was detected in the cervix) developed MAIDS as assessed by splenomegaly and decreased mitogen response, although the symptoms were less severe than of mice directly infected with LP-BM5 MuLV via the intraperitoneal route. Therefore, the cells in the genital organs were not only infected by the MAIDS virus but also able to replicate and spread the virus.

TABLE 2

Development of MAIDS in heterosexually infected B10 mice
Clinical condition
Spleen wt (mg)Mitogen response (cpm)Detection of LP-BM5 MuLV
MaleFemaleSpleenCervix
NormalNormal10539,981
9219,317
MAIDSNormal13610,346++
1867,799++
2454,911++
Open in a separate windowThe main route of HIV infection is heterosexual transmission (3, 5). However, the mechanisms of heterosexual transmission of retroviruses have been ill defined. HIV infection has been thought to occur during sexual contacts through slight injuries in the genital organs and to subsequently spread over the whole body. Among the genital organs of females, the parts of direct contact with male genital organs and semen are the vagina and cervix of uterus. The vagina is covered by a thick stratified squamous epithelium, while the cervix is covered by a monolayer columnar epithelium in addition to a squamous epithelium (2, 7). Histological examination (13) showed the presence of HIV-infected cells in the cervices derived from HIV carrier females (those infected with HIV by drug injections rather than by heterosexual transmission). Furthermore, a previous study utilizing female chimpanzees demonstrated that transmission of HIV could occur by insertion of cotton containing HIV into the vagina (8). These results suggested the presence of retrovirus-sensitive cells in genital organs. In our study, the cervix and penis are shown to be sensitive sites for virus infection (Table (Table1).1). Our assumption that there might be retrovirus-sensitive cells in a particular genital organ is currently under investigation by using in situ hybridization and immunohistochemical analyses.The heterosexual LP-BM5 MuLV infection rate for females to males appeared to be higher than that for males to females (Table (Table1).1). The mating frequency of normal male mice with infected female mice is supposed to be higher than that of normal female mice with infected male mice, since normal female mice fall into false pregnancy after mating and therefore reject male mice for a few weeks. This difference may also be attributed to the longer retention of genital secretions containing LP-BM5 MuLV in the male genital organs because of their phimoses (9). In fact, the defective virus genome was detected in vaginal secretions (both in secreted fluid and cells) by PCR (data not shown). Alternatively, the penis might be a highly sensitive site for retrovirus infection. In this regard, it is interesting that the defective virus genome was detected with very low frequency (1 of 17 male mice) in the prepuce even though it is constantly in contact with the penis. It is worth mentioning that contamination by retroviruses in the seminal fluid may happen at the prostate, seminal vesicle, vas deferens, Cowper’s glands, or penile urethra, since the sterilized (vasectomized) mice were still capable of transmitting the viruses to female mice (1, 6).The animal model for heterosexual transmission of retroviruses presented here has practical advantages, including (i) the high frequency of virus transmission and (ii) the possibility of rapid and cost-effective screening for antiretroviral agents (drugs and vaccines, etc.). This model may provide valuable information relating to heterosexual transmission of retroviruses including HIV and may further contribute to the prevention of HIV infection and the development of a remedy for AIDS.  相似文献   

19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号