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.
2.
3.
Specific therapy is not available for hantavirus cardiopulmonary syndrome caused by Andes virus (ANDV). Peptides capable of blocking ANDV infection in vitro were identified using antibodies against ANDV surface glycoproteins Gn and Gc to competitively elute a cyclic nonapeptide-bearing phage display library from purified ANDV particles. Phage was examined for ANDV infection inhibition in vitro, and nonapeptides were synthesized based on the most-potent phage sequences. Three peptides showed levels of viral inhibition which were significantly increased by combination treatment with anti-Gn- and anti-Gc-targeting peptides. These peptides will be valuable tools for further development of both peptide and nonpeptide therapeutic agents.Andes virus (ANDV), an NIAID category A agent linked to hantavirus cardiopulmonary syndrome (HCPS), belongs to the family Bunyaviridae and the genus Hantavirus and is carried by Oligoryzomys longicaudatus rodents (11). HCPS is characterized by pulmonary edema caused by capillary leak, with death often resulting from cardiogenic shock (9, 16). ANDV HCPS has a case fatality rate approaching 40%, and ANDV is the only hantavirus demonstrated to be capable of direct person-to-person transmission (15, 21). There is currently no specific therapy available for treatment of ANDV infection and HCPS.Peptide ligands that target a specific protein surface can have broad applications as therapeutics by blocking specific protein-protein interactions, such as preventing viral engagement of host cell receptors and thus preventing infection. Phage display libraries provide a powerful and inexpensive tool to identify such peptides. Here, we used selection of a cyclic nonapeptide-bearing phage library to identify peptides capable of binding the transmembrane surface glycoproteins of ANDV, Gn and Gc, and blocking infection in vitro.To identify peptide sequences capable of recognizing ANDV, we panned a cysteine-constrained cyclic nonapeptide-bearing phage display library (New England Biolabs) against density gradient-purified, UV-treated ANDV strain CHI-7913 (a gift from Hector Galeno, Santiago, Chile) (17, 18). To increase the specificity of the peptides identified, we eluted phage by using monoclonal antibodies (Austral Biologicals) prepared against recombinant fragments of ANDV Gn (residues 1 to 353) or Gc (residues 182 to 491) glycoproteins (antibodies 6B9/F5 and 6C5/D12, respectively). Peptide sequences were determined for phage from iterative rounds of panning, and the ability of phage to inhibit ANDV infection of Vero E6 cells was determined by immunofluorescent assay (IFA) (7). Primary IFA detection antibodies were rabbit polyclonal anti-Sin Nombre hantavirus (SNV) nucleoprotein (N) antibodies which exhibit potent cross-reactivity against other hantavirus N antigens (3). ReoPro, a commercially available Fab fragment which partially blocks infection of hantaviruses in vitro by binding the entry receptor integrin β3 (5), was used as a positive control (80 μg/ml) along with the original antibody used for phage elution (5 μg/ml). As the maximum effectiveness of ReoPro in inhibiting hantavirus entry approaches 80%, we set this as a threshold for maximal expected efficacy for normalization. The most-potent phage identified by elution with the anti-Gn antibody 6B9/F5 bore the peptide CPSNVNNIC and inhibited hantavirus entry by greater than 60% (61%) (Table (Table1).1). From phage eluted with the anti-Gc antibody 6C5/D12, those bearing peptides CPMSQNPTC and CPKLHPGGC also inhibited entry by greater than 60% (66% and 72%, respectively).

TABLE 1.

Peptide-bearing phage eluted from ANDV
Phage% Inhibition (SD)aP valueb
Phage bearing the following peptides eluted with anti-Gn antibody 6B9/F5
    Group 1 (<30% inhibition)
        CDQRTTRLC8.45 (15.34)0.0002
        CPHDPNHPC9.94 (7.72)0.333
        CQSQTRNHC11.76 (13.25)0.0001
        CLQDMRQFC13.26 (9.92)0.0014
        CLPTDPIQC15.70 (14.05)0.0005
        CPDHPFLRC16.65 (15.22)0.8523
        CSTRAENQC17.56 (16.50)0.0004
        CPSHLDAFC18.98 (20.06)0.0017
        CKTGHMRIC20.84 (7.47)0.0563
        CVRTPTHHC20.89 (27.07)0.1483
        CSGVINTTC21.57 (19.61)0.0643
        CPLASTRTC21.65 (5.98)0.004
        CSQFPPRLC22.19 (8.26)0.0004
        CLLNKQNAC22.34 (7.78)0.001
        CKFPLNAAC22.89 (6.15)0.0001
        CSLTPHRSC23.63 (16.74)0.0563
        CKPWPMYSC23.71 (6.68)0.0643
        CLQHDALNC24.01 (7.60)1
        CNANKPKMC24.67 (11.67)0.0004
        CPKHVLKVC25.30 (28.36)0.0003
        CTPDKKSFC26.91 (11.15)0.399
        CHGKAALAC27.22 (32.53)0.005
        CNLMGNPHC28.08 (21.35)0.0011
        CLKNWFQPC28.64 (18.49)0.0016
        CKEYGRQMC28.76 (29.33)0.0362
        CQPSDPHLC29.44 (31.22)0.0183
        CSHLPPNRC29.70 (17.37)0.0061
    Group 2 (30-59% inhibition)
        CSPLLRTVC33.05 (20.26)0.0023
        CHKGHTWNC34.17 (12.50)0.0795
        CINASHAHC35.62 (13.03)0.3193
        CWPPSSRTC36.75 (26.95)0.0006
        CPSSPFNHC37.78 (7.11)0.0001
        CEHLSHAAC38.47 (7.60)0.0115
        CQDRKTSQC38.74 (9.12)0.1802
        CTDVYRPTC38.90 (25.03)0.006
        CGEKSAQLC39.11 (27.52)0.0013
        CSAAERLNC40.13 (6.33)0.0033
        CFRTLEHLC42.07 (5.01)0.0608
        CEKLHTASC43.60 (27.92)0.1684
        CSLHSHKGC45.11 (49.81)0.0864
        CNSHSPVHC45.40 (28.80)0.0115
        CMQSAAAHC48.88 (44.40)0.5794
        CPAASHPRC51.84 (17.09)0.1935
        CKSLGSSQC53.90 (13.34)0.0145
    Group 3 (60-79% inhibition)
        CPSNVNNIC61.11 (25.41)0.1245
Negative control0 (6.15)
6B9/F5 (5 μg/ml)26.77 (5.33)
ReoPro (80 μg/ml)79.86 (4.88)
Phage bearing the following peptides eluted with anti-Gc antibody 6C5/D12
    Group 1 (<30% inhibition)
        CHPGSSSRC1.01 (7.03)0.0557
        CSLSPLGRC10.56 (13.62)0.7895
        CTARYTQHC12.86 (3.83)0.3193
        CHGVYALHC12.91 (7.32)0.0003
        CLQHNEREC16.79 (13.72)0.0958
        CHPSTHRYC17.23 (14.53)0.0011
        CPGNWWSTC19.34(9.91)0.1483
        CGMLNWNRC19.48 (19.42)0.0777
        CPHTQFWQC20.44 (13.65)0.0008
        CTPTMHNHC20.92 (11.68)0.0001
        CDQVAGYSC21.79 (23.60)0.0063
        CIPMMTEFC24.33 (9.28)0.2999
        CERPYSRLC24.38 (9.09)0.0041
        CPSLHTREC25.06 (22.78)0.1202
        CSPLQIPYC26.30 (34.29)0.4673
        CTTMTRMTC (×2)29.27 (8.65)0.0001
    Group 2 (30-59% inhibition)
        CNKPFSLPC30.09 (5.59)0.4384
        CHNLESGTC31.63 (26.67)0.751
        CNSVPPYQC31.96 (6.51)0.0903
        CSDSWLPRC32.95 (28.54)0.259
        CSAPFTKSC33.40 (10.64)0.0052
        CEGLPNIDC35.63 (19.90)0.0853
        CTSTHTKTC36.28 (13.42)0.132
        CLSIHSSVC36.40 (16.44)0.8981
        CPWSTQYAC36.81 (32.81)0.5725
        CTGSNLPIC36.83 (31.64)0.0307
        CSLAPANTC39.73 (4.03)0.1664
        CGLKTNPAC39.75 (16.98)0.2084
        CRDTTPWWC40.08 (18.52)0.0004
        CHTNASPHC40.26 (4.77)0.5904
        CTSMAYHHC41.89 (8.61)0.259
        CSLSSPRIC42.13 (29.75)0.2463
        CVSLEHQNC45.54 (6.55)0.5065
        CRVTQTHTC46.55 (8.45)0.3676
        CPTTKSNVC49.28 (14.00)0.3898
        CSPGPHRVC49.50 (42.60)0.0115
        CKSTSNVYC51.20 (4.60)0.0611
        CTVGPTRSC57.30 (11.31)0.0176
    Group 3 (60-79% inhibition)
        CPMSQNPTC65.60 (13.49)0.014
        CPKLHPGGC71.88 (27.11)0.0059
Negative control0.26 (4.53)
6C5/D12 (5 μg/ml)22.62 (8.40)
ReoPro (80 μg/ml)80.02 (76.64)
Open in a separate windowaStandard deviations of four experiments are shown in parentheses. Peptide-bearing phage were added at 109 phage/μl.bP values for the pairwise amino acid alignment score of each peptide versus that of integrin β3 were determined using an unpaired Student''s t test. P values considered statistically significant are shown in bold.To determine whether the peptide sequences of any of the identified inhibitory phage showed homology to integrin β3, a known entry receptor for pathogenic hantaviruses (6, 7), we used the Gap program to perform a pairwise amino acid alignment of each peptide versus the extracellular portion of integrin β3 and determined P values for the alignments. Of 45 phage eluted with the anti-Gn antibody, 6B9/F5, 27 of the peptide sequences showed homology to integrin β3 (P < 0.05), and 9 were highly significant (P ≤ 0.0005) (Fig. (Fig.1A).1A). Of the latter, CKFPLNAAC and CSQFPPRLC map to the hybrid domain (Fig. (Fig.1B),1B), which is proximal to the plexin-semaphorin-integrin domain (PSI) containing residue D39, shown to be critical for viral entry in vitro (19). Five sequences (CPSSPFNH, CPKHVLKVC, CNANKPKMC, CQSQTRNHC, and CDQRTTRLC) map to the I-like (or βA) domain near the binding site of ReoPro (2). Finally, CLPTDPIQC maps to the epidermal growth factor 4 (EGF-4) domain, and CSTRAENQC aligns to a portion of β3 untraceable in the crystal structure, specifically the linker region between the hybrid domain and EGF-1. Although this represents a disordered portion of the protein (22), the location of this loop proximal to the PSI domain is worth noting, due to the role of the PSI domain in facilitating viral entry (19). Therefore, 60% of phage eluted with the anti-Gn antibody showed some homology to integrin β3, and those with highly significant P values predominantly mapped to or proximal to regions of known interest in viral entry.Open in a separate windowFIG. 1.Inhibitory peptides identified through phage panning against ANDV show homology to integrin β3. (A) Alignment of phage peptide sequences with P values for integrin β3 pairwise alignment of less than 0.05. Residues comprising the signal peptide, transmembrane, and cytoplasmic domains, which were not included during pairwise alignment, are underlined. Residues 461 to 548, which are missing in the crystal structure, are italicized. Residues involved in the ReoPro binding site are highlighted in green (2). Residue D39 of the PSI domain is highlighted in yellow (19). Peptides are shown above the sequence of integrin β3, with antibody 6C5/D12-eluted sequences shown in blue text and sequences eluted with antibody 6B9/F5 shown in red. Peptide sequences with alignment P values of ≤0.0005 are highlighted in yellow. Percent inhibition of the peptide-bearing phage is shown in parentheses. (B) View of integrin αvβ3 (PDB ID 1U8C [23]). αv is shown in blue ribbon diagram, and β3 is shown in salmon-colored surface representation, with specific domains circled. Residues corresponding to the ReoPro binding site are shown in green, as in panel A, and D39 is shown in yellow. Regions corresponding to 6C5/D12-eluted peptides with P values of ≤0.0005 for alignment with integrin β3 (highlighted in panel A) are shown in blue, and those corresponding to 6B9/F5-eluted peptides with P values of ≤0.0005 for alignment with integrin β3 are shown in red. Alignment of peptide PLASTRT (P value of 0.0040) adjacent to D39 of the PSI domain is shown in magenta. Graphics were prepared using Pymol (DeLano Scientific LLC, San Carlos, CA).Of the 41 peptide-bearing phage eluted with the anti-Gc antibody 6C5/D12, 14 showed sequence homology to integrin β3 (P < 0.05), 4 of which had P values of ≤0.0005 (Fig. (Fig.1A).1A). Of the latter, sequence CTTMTRMTC mapped to the base of the I-like domain (Fig. (Fig.1B),1B), while CHGVYALHC and CRDTTPWWC mapped to the EGF-3 domain. Finally, sequence CTPTMHNHC mapped to the linker region untraceable in the crystal structure. Therefore, in contrast to peptide sequences identified by competition with the anti-Gn antibody, sequences identified by competition with the anti-Gc antibody 6C5/D12 appear to be mostly unrelated to integrin β3.As a low level of pathogenic hantavirus infection can be seen in cells lacking integrin β3, such as CHO cells (19), we asked if any of the identified peptide sequences could represent a previously unidentified receptor. We used the Basic Local Alignment Search Tool to search a current database of human protein sequences for potential alternate receptors represented by these peptides. However, none of the alignments identified proteins that are expressed at the cell surface, eliminating them as potential candidates for alternate viral entry receptors. This suggests that the majority of the peptides identified here likely represent novel sequences for binding ANDV surface glycoproteins.To determine whether synthetic peptides would also block infection, we synthesized cyclic peptides based on the 10 most-potent peptide-bearing phage. These peptides, in the context of phage presentation, showed levels of inhibition ranging from 44 to 72% (Table (Table2).2). When tested by IFA at 1 mM, four of the synthetic peptides showed inhibition levels significantly lower than those of the same peptide presented in the context of phage. This is not surprising, as steric factors due to the size of the phage and the multivalent presentation of peptide in the context of phage may both contribute to infection inhibition (8). However, there was no significant difference in inhibition by synthetic peptide versus peptide-bearing phage for six of the sequences, implying that inhibition in the context of phage was due solely to the nature of the peptide itself and not to steric factors or valency considerations contributed by the phage, which contrasts with our previous results, determined by using phage directed against αvβ3 integrin (10).

TABLE 2.

Synthetic cyclic peptides inhibit ANDV infection
TargetSample% Inhibition bya:
Peptide-bearing phageSynthetic peptide
GnCMQSAAAHC48.88 (44.40)59.66 (11.17)
GcCTVGPTRSC57.30 (11.31)46.47 (7.61)
GnCPSNVNNIC61.11 (25.41)44.14 (10.74)
GnCEKLHTASC43.60 (27.92)34.87 (9.26)
GcCPKLHPGGC71.88 (27.11)30.95 (7.73)b
GnCSLHSHKGC45.11 (49.81)29.79 (9.34)
GcCPMSQNPTC65.60 (13.49)18.19 (8.55)b
GnCKSLGSSQC53.90 (13.34)18.10 (7.55)b
GnCNSHSPVHC45.40 (28.80)15.52 (10.48)
GnCPAASHPRC51.84 (17.09)0 (10.72)b
Integrin β3ReoPro80.10 (7.72)
Gn6B9/F5 antibody42.72 (6.75)
Gc6C5/D12 antibody31.04 (7.81)
Open in a separate windowaStandard deviations of the results of at least four experiments are shown in parentheses.bMean percent inhibition between phage and synthetic peptide differs significantly (P < 0.05).The three most-potent synthetic peptides were examined for their ability to inhibit ANDV entry in a dose-dependent manner. The concentration of each peptide that produces 50% of its maximum potential inhibitory effect was determined. As shown in Fig. Fig.2A,2A, the 50% inhibitory concentration for each of the peptides was in the range of 10 μM, which from our experience is a reasonable potency for a lead compound to take forward for optimization.Open in a separate windowFIG. 2.Activities of synthetic peptides in inhibition of ANDV infection in vitro. (A) Peptides were examined for their ability to block ANDV infection of Vero E6 cells in a dose-dependent manner by IFA. (B) Peptides were tested in parallel for the ability to block infection of Vero E6 cells by ANDV, SNV, HTNV, and PHV. (C) Peptides were tested, singly or in combination, for the ability to block ANDV infection of Vero E6 cells. For all experiments, controls included media, ReoPro at 80 μg/ml, and monoclonal antibodies 6C5/D12 and 6B9/F5 at 5 μg/ml. All peptides were used at 1 mM. Data points represent n = 2 to 6, with error bars showing the standard errors of the means. Statistical analyses were performed on replicate samples using an unpaired Student''s t test.In order to determine the specificity of the three most-potent synthetic cyclic peptides in blocking ANDV, we examined them for inhibition of ANDV infection versus two other pathogenic hantaviruses, SNV and Hantaan virus (HTNV), or the nonpathogenic hantavirus Prospect Hill virus (PHV). As shown in Fig. Fig.2B,2B, ReoPro, which binds integrin β3, showed inhibition of infection by each of the pathogenic hantavirus strains, known to enter cells via β3, but not the nonpathogenic PHV, which enters via integrin β1 (6, 7). In contrast, peptides selected for the ability to bind ANDV were highly specific inhibitors of ANDV versus SNV, HTNV, or PHV. The specificities of peptides eluted by the anti-Gn monoclonal antibody are not surprising, as they are likely due to global differences in the Gn amino acid sequence. Specifically, sequence homologies between ANDV and SNV, HTNV, and PHV are 61%, 36%, and 51%, respectively, for the region corresponding to the immunogen for antibody 6B9/F5. Although homology between the immunogen for antibody 6C5/D12 and the corresponding Gc region of these viruses is somewhat higher (82% with SNV, 63% with HTNV, and 71% with PHV), the possibility that the monoclonal antibody used here recognizes a three-dimensional epitope lends itself to the high specificity of the peptides.The current model for cellular infection by hantaviruses (14) is as follows. Viral binding of the host cell surface target integrin is followed by receptor-mediated endocytosis and endosome acidification. Lowered pH induces conformational changes in Gn and/or Gc, which facilitate membrane fusion and viral release into the cytosol. As there is currently little information available about whether one glycoprotein is dominant in mediating infection, and as neutralizing epitopes have been found on both Gn and Gc glycoproteins (1, 4, 12, 13, 20), we examined whether combining anti-Gn- and anti-Gc-targeted synthetic peptides would lead to an increased infection blockade compared to those for single treatments. As shown in Fig. Fig.2C,2C, the combination of anti-Gn and anti-Gc peptides CMQSAAAHC and CTVGPTRSC resulted in a significant increase in infection inhibition (P = 0.0207 for CMQSAAAHC, and P = 0.0308 for CTVGPTRSC) compared to that resulting from single treatments. Although the high specificity of the peptides for ANDV makes it unlikely that this combination treatment will lead to more cross-reactivity with other pathogenic hantaviruses, this can be determined only by additional testing. Regardless, these data suggest a unique role for each of these viral proteins in the infection process as well as the benefits of targeting multiple viral epitopes for preventing infection.To our knowledge, the peptides reported here are the first identified that directly target ANDV, and this work further illustrates the power of coupling phage display and selective elution techniques in the identification of novel peptide sequences capable of specific protein-protein interactions from a large, random pool of peptide sequences. These novel peptide inhibitors (R. S. Larson, P. R. Hall, H. Njus, and B. Hjelle, U.S. patent application 61/205,211) provide leads for the development of more-potent peptide or nonpeptide organics for therapeutic use against HCPS.  相似文献   

4.
5.
GTP cyclohydrolase I (GCYH-I) is an essential Zn2+-dependent enzyme that catalyzes the first step of the de novo folate biosynthetic pathway in bacteria and plants, the 7-deazapurine biosynthetic pathway in Bacteria and Archaea, and the biopterin pathway in mammals. We recently reported the discovery of a new prokaryotic-specific GCYH-I (GCYH-IB) that displays no sequence identity to the canonical enzyme and is present in ∼25% of bacteria, the majority of which lack the canonical GCYH-I (renamed GCYH-IA). Genomic and genetic analyses indicate that in those organisms possessing both enzymes, e.g., Bacillus subtilis, GCYH-IA and -IB are functionally redundant, but differentially expressed. Whereas GCYH-IA is constitutively expressed, GCYH-IB is expressed only under Zn2+-limiting conditions. These observations are consistent with the hypothesis that GCYH-IB functions to allow folate biosynthesis during Zn2+ starvation. Here, we present biochemical and structural data showing that bacterial GCYH-IB, like GCYH-IA, belongs to the tunneling-fold (T-fold) superfamily. However, the GCYH-IA and -IB enzymes exhibit significant differences in global structure and active-site architecture. While GCYH-IA is a unimodular, homodecameric, Zn2+-dependent enzyme, GCYH-IB is a bimodular, homotetrameric enzyme activated by a variety of divalent cations. The structure of GCYH-IB and the broad metal dependence exhibited by this enzyme further underscore the mechanistic plasticity that is emerging for the T-fold superfamily. Notably, while humans possess the canonical GCYH-IA enzyme, many clinically important human pathogens possess only the GCYH-IB enzyme, suggesting that this enzyme is a potential new molecular target for antibacterial development.The Zn2+-dependent enzyme GTP cyclohydrolase I (GCYH-I; EC 3.5.4.16) is the first enzyme of the de novo tetrahydrofolate (THF) biosynthesis pathway (Fig. (Fig.1)1) (38). THF is an essential cofactor in one-carbon transfer reactions in the synthesis of purines, thymidylate, pantothenate, glycine, serine, and methionine in all kingdoms of life (38), and formylmethionyl-tRNA in bacteria (7). Recently, it has also been shown that GCYH-I is required for the biosynthesis of the 7-deazaguanosine-modified tRNA nucleosides queuosine and archaeosine produced in Bacteria and Archaea (44), respectively, as well as the 7-deazaadenosine metabolites produced in some Streptomyces species (33). GCYH-I is encoded in Escherichia coli by the folE gene (28) and catalyzes the conversion of GTP to 7,8-dihydroneopterin triphosphate (55), a complex reaction that begins with hydrolytic opening of the purine ring at C-8 of GTP to generate an N-formyl intermediate, followed by deformylation and subsequent rearrangement and cyclization of the ribosyl moiety to generate the pterin ring in THF (Fig. (Fig.1).1). Notably, the enzyme is dependent on an essential active-site Zn2+ that serves to activate a water molecule for nucleophilic attack at C-8 in the first step of the reaction (2).Open in a separate windowFIG. 1.Reaction catalyzed by GCYH-I, and metabolic fate of 7,8-dihydroneopterin triphosphate.A homologous GCYH-I is found in mammals and other higher eukaryotes, where it catalyzes the first step of the biopterin (BH4) pathway (Fig. (Fig.1),1), an essential cofactor in the biosynthesis of tyrosine and neurotransmitters, such as serotonin and l-3,4-dihydroxyphenylalanine (3, 52). Recently, a distinct class of GCYH-I enzymes, GCYH-IB (encoded by the folE2 gene), was discovered in microbes (26% of sequenced Bacteria and most Archaea) (12), including several clinically important human pathogens, e.g., Neisseria and Staphylococcus species. Notably, GCYH-IB is absent in eukaryotes.The distribution of folE (gene product renamed GCYH-IA) and folE2 (GCYH-IB) in bacteria is diverse (12). The majority of organisms possess either a folE (65%; e.g., Escherichia coli) or a folE2 (14%; e.g., Neisseria gonorrhoeae) gene. A significant number (12%; e.g., B. subtilis) possess both genes (a subset of 50 bacterial species is shown in Table Table1),1), and 9% lack both genes, although members of the latter group are mainly intracellular or symbiotic bacteria that rely on external sources of folate. The majority of Archaea possess only a folE2 gene, and the encoded GCYH-IB appears to be necessary only for the biosynthesis of the modified tRNA nucleoside archaeosine (44) except in the few halophilic Archaea that are known to synthesize folates, such as Haloferax volcanii, where GCYH-IB is involved in both archaeosine and folate formation (13, 44).

TABLE 1.

Distribution and candidate Zur-dependent regulation of alternative GCYH-I genes in bacteriaa
OrganismcPresence of:
folEfolE2
Enterobacteria
    Escherichia coli+
    Salmonella typhimurium+
    Yersinia pestis+
    Klebsiella pneumoniaeb++a
    Serratia marcescens++a
    Erwinia carotovora+
    Photorhabdus luminescens+
    Proteus mirabilis+
Gammaproteobacteria
    Vibrio cholerae+
    Acinetobacter sp. strain ADP1++a
    Pseudomonas aeruginosa++a
    Pseudomonas entomophila L48++a
    Pseudomonas fluorescens Pf-5++a
    Pseudomonas syringae++a
    Pseudomonas putida++a
    Hahella chejuensis KCTC 2396++a
    Chromohalobacter salexigens DSM 3043++a
    Methylococcus capsulatus++a
    Xanthomonas axonopodis++a
    Xanthomonas campestris++a
    Xylella fastidiosa++a
    Idiomarina loihiensis+
    Colwellia psychrerythraea++
    Pseudoalteromonas atlantica T6c++a
    Pseudoalteromonas haloplanktis TAC125++
    Alteromonas macleodi+
    Nitrosococcus oceani++
    Legionella pneumophila+
    Francisella tularensis+
Betaproteobacteria
    Chromobacterium violaceum+
    Neisseria gonorrhoeae+
    Burkholderia cepacia R18194++
    Burkholderia cenocepacia AU 1054++
    Burkholderia xenovorans+
    Burkholderia mallei+
    Bordetella pertussis+
    Ralstonia eutropha JMP134+
    Ralstonia metallidurans++
    Ralstonia solanacearum+
    Methylobacillus flagellatus+
    Nitrosomonas europaea+
    Azoarcus sp.++
Bacilli/Clostridia
    Bacillus subtilisd++
    Bacillus licheniformis++
    Bacillus cereus+
    Bacillus halodurans++
    Bacillus clausii+
    Geobacillus kaustophilus+
    Oceanobacillus iheyensis+
    Staphylococcus aureus+
Open in a separate windowaGenes that are preceded by candidate Zur binding sites.bZur-regulated cluster is on the virulence plasmid pLVPK.cExamples of organisms with no folE genes are in boldface type.dZn-dependent regulation of B. subtilis folE2 by Zur was experimentally verified (17).Expression of the Bacillus subtilis folE2 gene, yciA, is controlled by the Zn2+-dependent Zur repressor and is upregulated under Zn2+-limiting conditions (17). This led us to propose that the GCYH-IB family utilizes a metal other than Zn2+ to allow growth in Zn2+-limiting environments, a hypothesis strengthened by the observation that an archaeal ortholog from Methanocaldococcus jannaschii has recently been shown to be Fe2+ dependent (22). To test this hypothesis, we investigated the physiological role of GCYH-IB in B. subtilis, an organism that contains both isozymes, as well as the metal dependence of B. subtilis GCYH-IB in vitro. To gain a structural understanding of the metal dependence of GCYH-IB, we determined high-resolution crystal structures of Zn2+- and Mn2+-bound forms of the N. gonorrhoeae ortholog. Notably, although the GCYH-IA and -IB enzymes belong to the tunneling-fold (T-fold) superfamily, there are significant differences in their global and active-site architecture. These studies shed light on the physiological significance of the alternative folate biosynthesis isozymes in bacteria exposed to various metal environments, and offer a structural understanding of the differential metal dependence of GCYH-IA and -IB.  相似文献   

6.
Hereditary haemochromatosis (HH) is a common genetic disorder of iron metabolism in individuals of Northern European ancestry which leads to inappropriate iron absorption from the intestine and iron overload in susceptible individuals. Iron overload is suggested by elevations in serum ferritin and transferrin saturation. The majority of patients with clinically significant iron overload are homozygous for the C282Y mutation of the HFE gene, however only a minority of C282Y homozygotes fully express the disease clinically. Those with a high serum ferritin (>1000 μg/L) and additional hepatic insults from cofactors are more likely to develop cirrhosis and its complications. The mainstay of treatment is venesection. Those without cirrhosis who undergo appropriate venesection have a normal life expectancy. Family screening is recommended for all first degree relatives of an individual with the disease.HH refers to a group of inherited disorders that result in progressive iron overload. Mutations of the HFE gene are responsible for the majority of cases of HH,1 although disease expression is highly variable.2 The ready availability of testing for the two clinically relevant mutations: C282Y and H63D, has substantially altered the approach to suspected iron overload in clinical practice. A number of rare but important forms of non-HFE related HH have also been described.3 The other main causes of iron overload are outlined in the
HH
 HFE related HH
 (C282Y/C282Y, C282Y/H63D)
 Non-HFE related HH
  Juvenile Haemochromatosis
   Hemojuvelin related
   Hepcidin related
  Transferrin receptor-2 related HH
 Ferroportin related HH
Secondary Iron Overload
 Iron loading anaemia
  Thalassaemia major
  Sideroblastic anaemia
  Chronic haemolytic anaemia
 Parenteral iron overload (multiple transfusions)
Others
 Metabolic syndrome
 Chronic liver disease
  Hepatitis C
  Alcoholic liver disease
  Non-alcoholic steatohepatitis
  Porphyria cutanea tarda
 African Iron Overload
 Acaeruloplasminaemia
 Atransferrinaemia
 Neonatal iron overload
Open in a separate window  相似文献   

7.
Expanding Small-Molecule Functional Metagenomics through Parallel Screening of Broad-Host-Range Cosmid Environmental DNA Libraries in Diverse Proteobacteria     
Jeffrey W. Craig  Fang-Yuan Chang  Jeffrey H. Kim  Steven C. Obiajulu  Sean F. Brady 《Applied and environmental microbiology》2010,76(5):1633-1641
  相似文献   

8.
Rethinking Guard Cell Metabolism     
Diana Santelia  Tracy Lawson 《Plant physiology》2016,172(3):1371-1392
Stomata control gaseous fluxes between the internal leaf air spaces and the external atmosphere and, therefore, play a pivotal role in regulating CO2 uptake for photosynthesis as well as water loss through transpiration. Guard cells, which flank the stomata, undergo adjustments in volume, resulting in changes in pore aperture. Stomatal opening is mediated by the complex regulation of ion transport and solute biosynthesis. Ion transport is exceptionally well understood, whereas our knowledge of guard cell metabolism remains limited, despite several decades of research. In this review, we evaluate the current literature on metabolism in guard cells, particularly the roles of starch, sucrose, and malate. We explore the possible origins of sucrose, including guard cell photosynthesis, and discuss new evidence that points to multiple processes and plasticity in guard cell metabolism that enable these cells to function effectively to maintain optimal stomatal aperture. We also discuss the new tools, techniques, and approaches available for further exploring and potentially manipulating guard cell metabolism to improve plant water use and productivity.Stomata are microscopic, adjustable pores on the leaf surface. The evolution of stomata more than 400 million years ago (Edwards et al., 1986, 1992, 1998) helped facilitate the adaptation of plants to a terrestrial environment, where water is typically a limiting resource. Each stoma is composed of two kidney- or dumbbell-shaped guard cells, whose volume changes to adjust pore aperture, allowing plants to simultaneously regulate CO2 uptake and water loss. This facilitation of gas exchange by stomatal opening is one of the most essential processes in plant photosynthesis and transpiration, affecting plant water use efficiency and agricultural crop yields (Lawson and Blatt, 2014).Plant physiologists have a long history of investigating the behavior of these fascinating structures, reaching back more than a century to the pioneering work of Sir Francis Darwin (Darwin, 1916) and the American botanist Francis Ernest Lloyd (Lloyd, 1908). Major contributions to stomatal research arose from inventing and improving equipment and methods for quantitatively measuring the effects of environmental factors on stomatal pore aperture. After Darwin’s work, it became clear that the stomatal aperture actively responds to changes in the environment and regulates leaf transpiration rates (Meidner, 1987). Over the past century, much has been learned about their structure, development, and physiology.Despite the anatomical simplicity of the stomatal valve, the surrounding guard cells are highly specialized. Guard cells are morphologically distinct from general epidermal cells and possess complex signal transduction networks, elevated membrane ion transport capacity, and modified metabolic pathways. These features allow rapid modulations in guard cell turgor in response to endogenous and environmental signals, promoting the opening and closure of the stomatal pore in time scales of seconds to hours (Assmann and Wang, 2001). A variety of osmotically active solutes contribute to the buildup of stomatal turgor. Potassium (K+) and chloride (Cl) act as the main inorganic ions, and malate2− and sucrose (Suc) function as the main organic solutes. Whereas K+ and Cl are taken up from the apoplast, Suc and malate2− can be imported or synthesized internally using carbon skeletons deriving from starch degradation and/or CO2 fixation in the guard cell chloroplast (Roelfsema and Hedrich, 2005; Vavasseur and Raghavendra, 2005; Lawson, 2009; Kollist et al., 2014). The accumulation of these osmotica lowers the water potential, promoting the inflow of water, the swelling of guard cells, and the opening of the stomatal pore. Most of the ions taken up, or synthesized by guard cells, are sequestered into the vacuole (Barbier-Brygoo et al., 2011). As a result, the guard cell vacuoles undergo dynamic changes in volume and structure, which are crucial for achieving the full amplitude of stomatal movements (Gao et al., 2005; Tanaka et al., 2007; Andrés et al., 2014). During stomatal closure, guard cells reduce their volume through the release of ions into the cell wall and the consequent efflux of water.The transport of osmolytes across the plasma and tonoplast guard cell membranes is energized by H+-ATPase activity, which generates a proton motive force by translocating H+ ions against their concentration gradient (Blatt, 1987a, 1987b; Thiel et al., 1992; Roelfsema and Hedrich, 2005; Gaxiola et al., 2007). After the pioneering work of Fischer demonstrated the importance of K+ uptake in stomatal opening (Fischer, 1968; Fischer and Hsiao, 1968), K+ transport became of central interest and has long been considered the essence of stomatal movement regulation. The development of the voltage clamp technique, along with the relative easy acquisition of knockout mutants and transgenics in the model plant Arabidopsis (Arabidopsis thaliana), helped to uncover the precise mechanism and function of K+ fluxes in guard cells. It is well established that changes in membrane potential in response to several stimuli (e.g. light/darkness, CO2, and abscisic acid [ABA]) alter the direction of K+ transport (Thiel et al., 1992; Blatt, 2000; Roelfsema et al., 2001, 2002, 2004). During stomatal opening, the activation of the proton pump generates a sufficiently negative electric potential to cause the uptake of K+ through the inward-rectifying K+ channels (K+in; Fig. 1). During stomatal closure, K+ outflow from outward-rectifying K+ channels (K+out) results from membrane depolarization (Fig. 2; Blatt, 1988; Schroeder, 1988; Anderson et al., 1992; Sentenac et al., 1992). Besides being gated by opposing changes in voltage, the activation of (K+out) channels is dependent on the extracellular K+ concentration, while that of K+in is not (Blatt, 1988, 1992; Roelfsema and Prins, 1997; Dreyer and Blatt, 2009). There is also strong evidence for H+-coupled K+ symport in guard cells, which could account for up to 50% of total K+ uptake during stomatal opening (Blatt and Clint, 1989; Clint and Blatt, 1989; Hills et al., 2012). At least for K+in, the loss of a single-channel gene in Arabidopsis has little or no impact on stomatal movement (Szyroki et al., 2001), showing the redundancy among the different K+in isoforms and of K+ transport in general.Open in a separate windowFigure 1.Integration of guard cell carbohydrate metabolism with membrane ion transport during stomatal opening. Sugars in guard cells can be imported from the apoplast, derive from starch breakdown, or be synthesized in the Calvin cycle. These sugars then can be stored as osmotically active solutes in the vacuole or metabolized in the cytosol to yield energy, reducing equivalents, and phosphoenolpyruvate (PEP). PEP can be further metabolized to pyruvate in the mitochondrial tricarboxylic acid (CAC) cycle or used as carbon skeletons for the biosynthesis of malate via PEP carboxylase (PEPC) and NAD-dependent malate dehydrogenase (NAD-MDH). Malate (which also can be imported from the apoplast) and the inorganic ions K+ and Cl accumulate in the vacuole, lowering the guard cell osmotic potential, thereby promoting stomatal opening. ABCB14, ATP-binding cassette transporter B14; AcetylCoA, acetyl-CoA; ALMT, aluminum-activated malate transporter; ATP-PFK, ATP-dependent phosphofructokinase; AttDT, dicarboxylate transporter; cINV, cytosolic invertase; cwINV, cell wall invertase; Fru6P, Fru-6-P; Fru1,6P2, fructose 1,6-bisphosphate; Gl6P, Glc-6-P; G3P, glyceraldehyde 3-phosphate; iPGAM, phosphoglycerate mutase isoforms; NRGA1, negative regulator of guard cell ABA signaling1; OAA, oxaloacetate; 2-PGA, 2-phosphoglycerate; 3-PGA, 3-phosphoglycerate; PPi-PFK, PPi-dependent Fru-6-P phosphotransferase; STP, monosaccharide/H+ cotransporter; SUC, Suc/H+ cotransporter; SuSy, Suc synthase; TPT, triose phosphate/phosphate translocator. Compartments are not to scale. The dotted line indicates multiple metabolic steps.Open in a separate windowFigure 2.Proposed pathways of osmolyte dissipation during stomatal closure. While the removal of Cl and K+ is well described in the literature, the fate of Suc and malate during stomatal closure is unclear. Suc can be cleaved by cytosolic invertase (cINV), and the resulting hexoses can be imported into the chloroplast in the form of Glc-6-P (Glc6P). Glc6P is used subsequently for starch biosynthesis. Malate can be removed from the cell via decarboxylation to pyruvate by malic enzyme (ME) and the subsequent complete oxidation in the mitochondrial tricarboxylic acid (CAC) cycle. Alternatively, malate can be converted to PEP via NAD+-dependent malate dehydrogenase (NAD-MDH) and PEP carboxykinase (PEPCK). Gluconeogenic conversion of PEP to Glc6P establishes a possible link between malate removal and starch synthesis. Compartments are not to scale. PEP, Phosphoenolpyruvate; OAA; oxaloacetate; STP, monosaccharide/H+ cotransporter; SUC, Suc/H+ cotransporter; SuSy, Suc synthase; cINV, cytosolic invertase; NRGA1, negative regulator of guard cell ABA signaling1; ALMT, aluminum-activated malate transporter; GPT, Glc-6-P/Pi translocator; cwINV, cell wall invertase; HK, hexokinase; QUAC1, quickly activating anion channel1.Despite the undisputed importance of K+ uptake in stomatal opening, the accumulation of K+ ions alone cannot account for the increase in osmotic pressure necessary to explain stomatal aperture. Studies from the 1980s by MacRobbie and Fischer demonstrated that Vicia faba guard cells take up approximately 2 pmol of K+ during stomatal opening. Assuming that K+ uptake is balanced by the accumulation of similar amounts of counter ions (Cl and/or malate2−), the expected increase in stomatal turgor to approximately 3 MPa is less than the 4.5 MPa expected for fully open stomata (Fischer, 1972; MacRobbie and Lettau, 1980a, 1980b; Chen et al., 2012). The realization that other solutes must accumulate in addition to K+ salts was one of the major paradigm shifts in stomatal physiology research in the last decades, equal to the discovery of ion channels. Suc was put forward as the most likely candidate for the additional osmoticum to support stomatal opening (MacRobbie, 1987; Tallman and Zeiger, 1988; Talbott and Zeiger, 1993, 1998). Nonetheless, this research area subsequently failed to attract notice commensurate with its importance.In the last few years, the metabolism of starch, sugars and, organic acids in guard cells has seen a rebirth, making this the perfect time to review the developments in this field. In this review, we focus on photosynthetic carbon assimilation and respiratory metabolism in guard cells and provide a historical overview of the subject that highlights the most up-to-date and novel discoveries in guard cell research. We describe the various metabolic pathways separately, but as metabolism is an integrated network, we also discuss their reciprocal and beneficial interactions. Finally, we highlight their connection with the metabolism in the subjacent mesophyll cells and how they integrate with guard cell signal transduction networks and membrane ion transport to regulate stomatal movements. The enzymes and transporters discussed in this review are listed in Arabidopsis Genome Initiative CodeGeneProteinFunctionMalate transport AT1G28010ABCB14ATP-binding cassette transporter B14Import of apoplastic malate AT5G47560tDTDicarboxylate transporterTransport of carboxylates into the vacuole AT3G18440ALMT9Aluminum-activated malate transporter9Transport of Cl/malate2− into the vacuole AT2G17470ALMT6Aluminum-activated malate transporter6Transport of malate2− into the vacuole AT4G17970ALMT12/QUAC1Aluminum-activated malate transporter12Export of cytosolic Cl/malate2− to the apoplastMalate metabolism –PEPCPhosphoenolpyruvate carboxylaseβ-Carboxylation of PEP to OAA –NAD-MDHNAD+-dependent malate dehydrogenaseReduction of OAA to malate –MEMalic enzymeOxidative decarboxylation of malate to pyruvate AT4G37870PEPCK1PEP carboxykinase1Conversion of OAA to PEP –PPDKPyruvate, orthophosphate dikinaseConversion of pyruvate to PEPOther carboxylates –TPTTriose phosphate/phosphate translocatorExport of triose phosphate from the chloroplast to the cytosol AT4G05590NRGA1Negative regulator of guard cell ABA signaling1Putative mitochondrial pyruvate carrier –SDH2Succinate dehydrogenase2Oxidation of succinate to fumarate AT2G47510FUM1Fumarase1Hydration of fumarate to malate –iPGAMPhosphoglycerate mutaseInterconversion of 3-PGA to 2-PGA –PPi-PFKPPi-dependent Fru-6-P phosphotransferasePhosphorylation of Fru-6-P to Fru-1,6-bisphosphate –ATP-PFKATP-dependent phosphofructokinasePhosphorylation of Fru-6-P to Fru-1,6-bisphosphateCalvin cycle –RubiscoRubiscoCarboxylation of ribulose 1,5-bisphosphate AT3G55800SBPaseSedoheptulose-bisphosphataseDephosphorylation of sedoheptulose-1,7-bisphosphate to sedoheptulose-7-phosphateSugar metabolism AT4G29130HK1Hexokinase1Phosphorylation of Glc to Glc-6-P AT4G02280SuSySuc synthase3Interconversion of Suc to Fru and UDP-Glc –cINVCytosolic invertaseHydrolysis of Suc to Fru and Glc –cwINVCell wall invertaseHydrolysis of Suc to Fru and GlcSugar transport AT1G11260STP1Monosaccharide/H+ cotransporter1Import of apoplastic hexose sugars AT3G19930STP4Monosaccharide/H+ cotransporter4Import of apoplastic hexose sugars AT1G71880SUC1Suc/H+ cotransporter1Import of apoplastic Suc AT2G02860SUC3Suc/H+ cotransporter3Import of apoplastic SucStarch degradation AT3G23920BAM1β-Amylase1Hydrolysis of α-1,4 external glucoside linkages in starch AT1G69830AMY3α-Amylase3Hydrolysis of α-1,4 internal glucoside linkages in starchStarch synthesis –GPTGlc-6-P/Pi translocatorUptake of cytosolic Glc-6-P into the chloroplast AT4G24620PGIPhosphoglucose isomeraseConversion of Fru-6-P to Glc-6-P AT5G51820PGM1Phosphoglucomutase1Conversion of Glc-6-P to Glc-1-P AT5G48300APS1ADPGlc pyrophosphorylase small subunitConversion of Glc-1-P to ADPGlc, catalytic subunit –APLADPGlc pyrophosphorylase large subunitConversion of Glc-1-P to ADPGlc, regulatory subunitVarious AT3G45780PHOT1Phototropin1Blue light photoreceptor AT5G58140PHOT2Phototropin2Blue light photoreceptor AT4G14480BLUS1Blue light signaling1Protein kinase, regulator of blue light-induced stomatal opening –PP1Protein phosphatase1Regulator of blue light-induced stomatal opening AT3G01500CA1Carbonic anhydrase1Interconversion of CO2 and water into H2CO3 AT1G70410CA4Carbonic anhydrase4Interconversion of CO2 and water into H2CO3 AT1G62400HT1High leaf temperature1Protein kinase, regulator of CO2-induced stomatal closureOpen in a separate window  相似文献   

9.
Genetic Transformation and Mutagenesis via Single-Stranded DNA in the Unicellular,Diazotrophic Cyanobacteria of the Genus Cyanothece     
Hongtao Min  Louis A. Sherman 《Applied and environmental microbiology》2010,76(22):7641-7645
  相似文献   

10.
Alleviation of Proteolytic Sensitivity To Enhance Recombinant Lipase Production in Escherichia coli     
Niju Narayanan  C. Perry Chou 《Applied and environmental microbiology》2009,75(16):5424-5427
Two amino acids, Leu149 and Val223, were identified as proteolytically sensitive when Pseudozyma antarctica lipase (PalB) was heterologously expressed in Escherichia coli. The functional expression was enhanced using the double mutant for cultivation. However, the recombinant protein production was still limited by PalB misfolding, which was resolved by DsbA coexpression.Though Escherichia coli retains popularity as a host for recombinant protein production, it has technical limitations for expressing eukaryotic proteins. Host/protein incompatibility is critical, since eukaryotic proteins heterologously expressed in E. coli are recognized as a foreign object that can induce heat shock responses and drive protease-mediated degradation. E. coli contains several proteases situated in various intracellular compartments, including the cytoplasm, periplasm, inner membrane, and outer membrane (7, 9). While these proteases play a pivotal role in maintaining cell physiology by clearing up abnormal proteins (4), they can potentially degrade recombinant proteins (6). Nevertheless, the mechanisms associated with protein degradation and protein substrate specificity are incompletely understood.Since the proteolysis of recombinant proteins is a common issue, approaches to overcome this limitation have been extensively explored. These include using protease-deficient mutants as an expression host (10, 18), lowering the cultivation temperature (3), coexpressing folding factors (19, 20), applying protein fusion technology (2, 8), eliminating protease cleavage sites (5, 15), and modifying target protein hydrophobicity (11). While genetic elimination of proteolytically sensitive sequences without impacting the target protein''s bioactivity appears to be technically attractive, there is no effective prediction or identification of potential protease cleavage sites. Proteolytically sensitive sequences might not be specific to certain proteases. Various factors affecting the folding, solubility, steric hindrance, and binding of the substrate protein, e.g., neighboring domains or amino acids, can contribute to the sensitivity of proteolytic sites, making sequence manipulation for proteolysis alleviation more difficult.Herein, the recombinant protein''s sensitivity toward intracellular proteolysis was genetically alleviated to enhance the functional expression of PalB, an industrial enzyme with justified applications (1), in E. coli and to understand the relationship between proteolytic specificity and substrate protein sequence. Since there are three intramolecular disulfide bonds whose formation can potentially affect PalB structure and bioactivity, PalB expression in the oxidative periplasm of E. coli was explored (20, 22). However, the expressed PalB protein was extremely sensitive to intracellular proteolysis, resulting in complete degradation. To overcome this problem, PalB mutant variants that were stable against proteolysis were derived (Table (Table11).

TABLE 1.

Strains, plasmids, and oligonucleotides
Strain, plasmid, or oligonucleotideRelevant genotype or DNA sequencea
Strain
    BL21(DE3)FompT dcm lon hsdSB (rB mB) gal λ(DE3[lacIind1sam7 nin5 lacUV5-T7 gene 1])
Plasmids
    pARDsbA (20)ParaB::dsbA, Ori (pACYC184), Cmr
    pARDsbC (20)ParaB::dsbA, Ori (pACYC184), Cmr
    pETG (20)PT7-pelB::palB, Ori (pBR322), Apr
    pETGM-2pETG derivative containing two mutations of Leu149Val and Val223Ile in palB (by ep-PCR with pETG and LB-P10/LB-P11)
    pETGM-3pETG derivative containing mutation of Leu149Gly in palB (by SDMb with pETG and P104/P114)
    pETGM-4pETG derivative containing mutation of Leu149Val in palB (by SDM with pETG and P103/P113)
    pETGM-5pETG derivative containing mutation of Leu149Ala in palB (by SDM with pETG and P105/P115)
    pETGM-6pETG derivative containing mutation of Leu149Met in palB (by SDM with pETG and P106/P116)
    pETGM-7pETG derivative containing mutation of Leu149Ile in palB (by SDM with pETG and P107/P117)
    pETGM-8pETG derivative containing mutation of Val223Phe in palB (by SDM with pETG and P112/P122)
    pETGM-9pETG derivative containing mutation of Val223Met in palB (by SDM with pETG and P111/P121)
    pETGM-10pETG derivative containing mutation of Val223Ala in palB (by SDM with pETG and P110/P120)
    pETGM-11pETG derivative containing mutation of Val223Gly in palB (by SDM with pETG and P109/P119)
    pETGM-12pETG derivative containing mutation of Val223Ile in palB (by SDM with pETGM-2 and P136/P137, i.e., by reverting the mutation of Val149 to Leu in pETGM-2)
Oligonucleotides
    LB-P105′-GGCCATGGGTCTACCTTCCGGTTCGG-3′
    LB-P115′-CTGAATTCTCAGGGGGTGACGATGCCGGAGCAGG-3′
    P1035′-CCTCTCGATGCAGTCGCGGTTAGTGCACCC-3′
    P1135′-GGGTGCACTAACCGCGACTGCATCGAGAGG-3′
    P1045′-CCTCTCGATGCAGGCGCCGTTAGTGCACCC-3′
    P1145′-GGGTGCACTAACGGCGCCTGCATCGAGAGG-3′
    P1055′-CCTCTCGATGCAGCCGCGGTTAGTGCACCC-3′
    P1155′-GGGTGCACTAACCGCGGCTGCATCGAGAGG-3′
    P1065′-CCTCTCGATGCCATGGCGGTTAGTGCACCC-3′
    P1165′-GGGTGCACTAACCGCCATGGCATCGAGAGG-3′
    P1075′-CCTCTCGATGCGATCGCGGTTAGTGCACCC-3′
    P1175′-GGGTGCACTAACCGCGATCGCATCGAGAGG-3′
    P1095′-GGGCCGCTGTTCGGGATCGACCATGCAGGC-3′
    P1195′-GCCTGCATGGTCGATCCCGAACAGCGGCCC-3′
    P1105′-GGGCCGCTGTTCGCGATCGACCATGCAGGC-3′
    P1205′-GCCTGCATGGTCGATCGCGAACAGCGGCCC-3′
    P1115′-GGGCCGCTGTTCATGATCGACCATGCAGGC-3′
    P1215′-GCCTGCATGGTCGATCATGAACAGCGGCCC-3′
    P1125′-GGGCCGCTGTTCTTTATCGACCATGCAGGC-3′
    P1225′-GCCTGCATGGTCGATAAAGAACAGCGGCCC-3′
    P1365′-CCTCTCGATGCACTGGCGGTTAGTGCACCC-3′
    P1375′-GGGTGCACTAACCGCCAGTGCATCGAGAGG-3′
Open in a separate windowaFor oligonucleotides, italic indicates mutated nucleotides, underlining indicates restriction site (silent mutation), and bold indicates a mutation codon.bSDM, site-directed mutagenesis.The results for functional expression of various PalB variants are summarized in Table Table22 and Fig. Fig.1.1. Neither PalB activity nor PalB-related polypeptide was detectable for the BL21(DE3)(pETG) culture sample, suggesting that the expressed gene product was degraded. Error-prone PCR (ep-PCR) was conducted using pETG as the template and LB-P10/LB-P11 as the primers. Potential mutants were screened on tributyrin agar plates. A colony developed a large halo, and the harboring plasmid (i.e., pETGM-2) was purified for sequencing. Two mutations, Leu149Val and Val223Ile, were identified in the expressed PalB mutant (i.e., M-2). High PalB activity was obtained for the BL21(DE3) (pETGM-2) culture sample. However, a PalB-related polypeptide was detected in both the soluble and insoluble fractions, implying potential protein misfolding. It was previously reported that the functional expression of PalB could be limited by disulfide bond formation associated with folding (21). It was intriguing to observe the structural effect of these two simple mutations on improving PalB stabilization and functional expression.Open in a separate windowFIG. 1.Western blotting analysis of the culture samples for the expression of various PalB mutants. Both soluble and insoluble fractions are shown. The number (n = 2 to 12) represents the PalB mutant (M-n) with the use of BL21(DE3) harboring the expression plasmid pETGM-n, summarized in Tables Tables11 and and2.2. “G” represents the control experiment using BL21(DE3) harboring pETG. “C” and “I” represent the cultures without and with IPTG induction, respectively.

TABLE 2.

Cultivation performance for production of PalB mutant variantsa
PlasmidCell density (OD600)b
Activity (U/liter/OD600)cMutation in PalB
w/o IPTGw/IPTG
pETG3.1 ± 0.03.3 ± 0.1NDNone
pETGM-23.4 ± 0.13.3 ± 0.0155Leu149Val, Val223Ile
pETGM-33.2 ± 0.13.3 ± 0.0NDLeu149Gly
pETGM-43.4 ± 0.13.6 ± 0.160Leu149Val
pETGM-53.3 ± 0.13.5 ± 0.1NDLeu149Ala
pETGM-62.7 ± 0.13.0 ± 0.1NDLeu149Met
pETGM-73.5 ± 0.03.2 ± 0.038Leu149Ile
pETGM-83.0 ± 0.03.6 ± 0.050Val223Phe
pETGM-92.9 ± 0.12.8 ± 0.0NDVal223Met
pETGM-103.4 ± 0.13.3 ± 0.1NDVal223Ala
pETGM-113.5 ± 0.03.5 ± 0.130Val223Gly
pETGM-123.6 ± 0.13.5 ± 0.157Val223Ile
Open in a separate windowaBL21(DE3) was used as an expression host to harbor various plasmids. PalB enzyme assay was conducted at 37°C and pH 8.0 using a pH stat with olive oil as a substrate. One unit of enzyme activity is defined as the amount of enzyme required to liberate one μmole of fatty acid per min.bOD600, optical density at 600 nm; w/o and w/IPTG, without and with isopropyl-β-d-thiogalactopyranoside, respectively.cThese activities represent IPTG-induced cultures. No activity was detected for all control cultures without IPTG induction. ND, not detected.Site-directed mutagenesis was conducted to introduce various single mutations of Leu149 and Val223 for a further understanding of the structural effect. Note that only hydrophobic amino acid residues were selected for the replacement to avoid any major structural disturbances. Among the five single mutations on Leu149, only Leu149Val and Leu149Ile were associated with a slight improvement in functional expression, and Leu149Val was more effective than Leu149Ile, whereas the others behaved similarly to the wild type. Among the five single mutations at Val223, only Val223Phe, Val223Gly, and Val223Ile showed a slight improvement in functional expression, with Val223Ile as the most effective one, whereas the others showed no improvement. The results suggest that Leu149 and Val223 are two amino acids critically affecting the proteolytic susceptibility of PalB in E. coli, with a synergistic effect on PalB stabilization from Leu149Val and Val223Ile mutations.Several amino acids were identified as being involved in the catalytic mechanism (17), such as Ser107-Asp189-His226, forming the catalytic triad, and Thr42 and Gly108, acting as the hydrogen bond donors in the oxyanion hole of the active site. However, Leu149 is located at the entrance of the partially formed lid of the protein structure, whereas Val223 is situated near one of the catalytic triad residues, His226, in the active site. The substitutions at Leu149 and/or Val223 with a PalB stabilization effect potentially convert the overall protein conformation into a less proteolytically sensitive form without changing the enzyme kinetics or activity. It was interesting to observe a similarly positive effect upon replacing Val223 with either a bulky Phe or a small Gly, implying that the molecular size of this amino acid residue was not critical for alleviation of the proteolytic sensitivity. Note that PalB-related polypeptide was present in the insoluble fraction for all the PalB mutants with improved functional expression, suggesting another expression hurdle of protein misfolding.To overcome the protein misfolding, coexpression of two periplasmic folding factors, DsbA and DsbC, was explored, and the cultivation results are summarized in Fig. Fig.2.2. DsbA coexpression significantly boosted functional expression, with specific PalB activity of more than twofold that of the control culture and a slight reduction of insoluble PalB. However, the improving effect associated with DsbC coexpression was minimal. The results suggest that the initiation but not isomerization for disulfide bond formation in the periplasm became limiting. The chaperone activity associated with DsbA could assist the folding of the expressed PalB double mutant.Open in a separate windowFIG. 2.Effect of DsbA or DsbC coexpression on the functional expression of the PalB double mutant. Cultivations were performed in a bioreactor containing 1 liter LB medium and operated at pH 7.0, 28°C, and 650 rpm. The cultures were induced with 0.1 mM isopropyl-β-d-thiogalactopyranoside and/or 0.2 g/liter arabinose at a ∼0.5 optical density at 600 nm (OD600) cell density. (A) Time profiles of cell density; (B) time profiles of the specific PalB activity [note that there was no detectable activity for BL21(DE3) (pETG) culture samples]; (C) Western blotting analysis of the final samples of the four cultures. Both soluble and insoluble fractions are shown. Lanes 1 and 5, BL21(DE3) (pETG); lanes 2 and 6, BL21(DE3) (pETGM-2); lane 3 and 7, BL21(DE3) (pETGM-2, pARDsbC); lanes 4 and 8, BL21(DE3) (pETGM-2, pARDsbA).While many studies based on rational mutagenesis, directed evolution, and gene shuffling are conducted to derive PalB mutants with improved enzyme properties, such as thermostability, bioactivity, or enantioselectivity (12-14, 16), this is an approach to improve protein manufacturing by deriving the variants with less proteolytic susceptibility. Though it has been perceived that the proteolytic specificity can be intrinsically determined by the protein substrate sequence, experimental demonstration of this is rarely reported. This study complements the lack of such experimental demonstration by showing the proteolytic specificity and sensitivity of PalB heterologously expressed in E. coli can be drastically altered by simple amino acid substitutions. It also demonstrates the application of molecular manipulation to enhance recombinant protein production in E. coli.  相似文献   

11.
Repression of recA Induction by RecX Is Independent of the RecA Protein in Deinococcus radiodurans     
Duohong Sheng  Mingfeng Li  Jiandong Jiao  Xiehuang Sheng  Wenqiang Deng  Yuejin Hua 《Journal of bacteriology》2010,192(13):3540-3544
  相似文献   

12.
Variation in Adult Plant Phenotypes and Partitioning among Seed and Stem-Borne Roots across Brachypodium distachyon Accessions to Exploit in Breeding Cereals for Well-Watered and Drought Environments     
Vincent Chochois  John P. Vogel  Gregory J. Rebetzke  Michelle Watt 《Plant physiology》2015,168(3):953-967
Seedling roots enable plant establishment. Their small phenotypes are measured routinely. Adult root systems are relevant to yield and efficiency, but phenotyping is challenging. Root length exceeds the volume of most pots. Field studies measure partial adult root systems through coring or use seedling roots as adult surrogates. Here, we phenotyped 79 diverse lines of the small grass model Brachypodium distachyon to adults in 50-cm-long tubes of soil with irrigation; a subset of 16 lines was droughted. Variation was large (total biomass, ×8; total root length [TRL], ×10; and root mass ratio, ×6), repeatable, and attributable to genetic factors (heritabilities ranged from approximately 50% for root growth to 82% for partitioning phenotypes). Lines were dissected into seed-borne tissues (stem and primary seminal axile roots) and stem-borne tissues (tillers and coleoptile and leaf node axile roots) plus branch roots. All lines developed one seminal root that varied, with branch roots, from 31% to 90% of TRL in the well-watered condition. With drought, 100% of TRL was seminal, regardless of line because nodal roots were almost always inhibited in drying topsoil. Irrigation stimulated nodal roots depending on genotype. Shoot size and tillers correlated positively with roots with irrigation, but partitioning depended on genotype and was plastic with drought. Adult root systems of B. distachyon have genetic variation to exploit to increase cereal yields through genes associated with partitioning among roots and their responsiveness to irrigation. Whole-plant phenotypes could enhance gain for droughted environments because root and shoot traits are coselected.Adult plant root systems are relevant to the size and efficiency of seed yield. They supply water and nutrients for the plant to acquire biomass, which is positively correlated to the harvest index (allocation to seed grain), and the stages of flowering and grain development. Modeling in wheat (Triticum aestivum) suggested that an extra 10 mm of water absorbed by such adult root systems during grain filling resulted in an increase of approximately 500 kg grain ha−1 (Manschadi et al., 2006). This was 25% above the average annual yield of wheat in rain-fed environments of Australia. This number was remarkably close to experimental data obtained in the field in Australia (Kirkegaard et al., 2007). Together, these modeling and field experiments have shown that adult root systems are critical for water absorption and grain yield in cereals, such as wheat, emphasizing the importance of characterizing adult root systems to identify phenotypes for productivity improvements.Most root phenotypes, however, have been described for seedling roots. Seedling roots are essential for plant establishment, and hence, the plant’s potential to set seed. For technical reasons, seedlings are more often screened than adult plants because of the ease of handling smaller plants and the high throughput. Seedling-stage phenotyping may also improve overall reproducibility of results because often, growth media are soil free. Seedling soil-free root phenotyping conditions are well suited to dissecting fine and sensitive mechanisms, such as lateral root initiation (Casimiro et al., 2003; Péret et al., 2009a, 2009b). A number of genes underlying root processes have been identified or characterized using seedlings, notably with the dicotyledonous models Arabidopsis (Arabidopsis thaliana; Mouchel et al., 2004; Fitz Gerald et al., 2006; Yokawa et al., 2013) and Medicago truncatula (Laffont et al., 2010) and the cereals maize (Zea mays; Hochholdinger et al., 2001) and rice (Oryza sativa; Inukai et al., 2005; Kitomi et al., 2008).Extrapolation from seedling to adult root systems presents major questions (Hochholdinger and Zimmermann, 2008; Chochois et al., 2012; Rich and Watt, 2013). Are phenotypes in seedling roots present in adult roots given developmental events associated with aging? Is expression of phenotypes correlated in seedling and adult roots if time compounds effects of growth rates and growth conditions on roots? Watt et al. (2013) showed in wheat seedlings that root traits in the laboratory and field correlated positively but that neither correlated with adult root traits in the field. Factors between seedling and adult roots seemed to be differences in developmental stage and the time that growing roots experience the environment.Seedling and adult root differences may be larger in grasses than dicotyledons. Grass root systems have two developmental components: seed-borne (seminal) roots, of which a number emerge at germination and continue to grow and branch throughout the plant life, and stem-borne (nodal or adventitious) roots, which emerge from around the three-leaf stage and continue to emerge, grow, and branch throughout the plant life. Phenotypes and traits of adult root systems of grasses, which include the major cereal crops wheat, rice, and maize, are difficult to predict in seedling screens and ideally identified from adult root systems first (Gamuyao et al., 2012).Phenotyping of adult roots is possible in the field using trenches (Maeght et al., 2013) or coring (Wasson et al., 2014). A portion of the root system is captured with these methods. Alternatively, entire adult root systems can be contained within pots dug into the ground before sowing. These need to be large; field wheat roots, for example, can reach depths greater than 1.5 m depending on genotype and environment. This method prevents root-root interactions that occur under normal field sowing of a plant canopy and is also a compromise.A solution to the problem of phenotyping adult cereal root systems is a model for monocotyledon grasses: Brachypodium distachyon. B. distachyon is a small-stature grass with a small genome that is fully sequenced (Vogel et al., 2010). It has molecular tools equivalent to those available in Arabidopsis (Draper et al., 2001; Brkljacic et al., 2011; Mur et al., 2011). The root system of B. distachyon reference line Bd21 is more similar to wheat than other model and crop grasses (Watt et al., 2009). It has a seed-borne primary seminal root (PSR) that emerges from the embryo at seed germination and multiple stem-borne coleoptile node axile roots (CNRs) and leaf node axile roots (LNRs), also known as crown roots or adventitious roots, that emerge at about three leaves through to grain development. Branch roots emerge from all root types. There are no known anatomical differences between root types of wheat and B. distachyon (Watt et al., 2009). In a recent study, we report postflowering root growth in B. distachyon line Bd21-3, showing that this model can be used to answer questions relevant to the adult root systems of grasses (Chochois et al., 2012).In this study, we used B. distachyon to identify adult plant phenotypes related to the partitioning among seed-borne and stem-borne shoots and roots for the genetic improvement of well-watered and droughted cereals (Fig. 1; Krassovsky, 1926; Navara et al., 1994), nitrogen, phosphorus (Tennant, 1976; Brady et al., 1995), oxygen (Wiengweera and Greenway, 2004), soil hardness (Acuna et al., 2007), and microorganisms (Sivasithamparam et al., 1978). Of note is the study by Krassovsky (1926), which was the first, to our knowledge, to show differences in function related to water. Krassovsky (1926) showed that seminal roots of wheat absorbed almost 2 times the water as nodal roots per unit dry weight but that nodal roots absorbed a more diluted nutrient solution than seminal roots. Krassovsky (1926) also showed by removing seminal or nodal roots as they emerged that “seminal roots serve the main stem, while nodal roots serve the tillers” (Krassovsky, 1926). Volkmar (1997) showed, more recently, in wheat that nodal and seminal roots may sense and respond to drought differently. In millet (Pennisetum glaucum) and sorghum (Sorghum bicolor), Rostamza et al. (2013) found that millet was able to grow nodal roots in a dryer soil than sorghum, possibly because of shoot and root vigor.Open in a separate windowFigure 1.B. distachyon plant scanned at the fourth leaf stage, with the root and shoot phenotypes studied indicated. Supplemental Table S1.
PhenotypeAbbreviationUnitRange of Variation
All Experiments (79 Lines and 582 Plants)Experiment 6 (36 Lines)
Whole plant
TDWTDWMilligrams88.6–773.8 (×8.7)285.6–438 (×1.5)
Shoot
SDWSDWMilligrams56.4–442.5 (×7.8)78.2–442.5 (×5.7)
 No. of tillersTillerNCount2.8–20.3 (×7.4)10–20.3 (×2)
Total root system
TRLTRLCentimeters1,050–10,770 (×10.3)2,090–5,140 (×2.5)
RDWRDWMilligrams28.9–312.17 (×10.8)62.2–179.1 (×2.9)
RootpcRootpcPercentage (of TDW)20.5–60.6 (×3)20.5–44.3 (×2.2)
R/SR/SUnitless ratio0.26–1.54 (×6)0.26–0.80 (×3.1)
PSRs
 Length (including branch roots)PSRLCentimeters549.1–4,024.6 (×7.3)716–2,984 (×4.2)
PSRpcPSRpcPercentage (of TRL)14.9–94.1 (×6.3)31.3–72.3 (×2.3)
 No. of axile rootsPSRcountCount11
 Length of axile rootPSRsumCentimeters17.45–52 (×3)17.45–30.3 (×1.7)
 Branch rootsPSRbranchCentimeters · (centimeters of axile root)−119.9–109.3 (×5.5)29.3–104.3 (×3.6)
CNRs
 Length (including branch roots)CNRLCentimeters0–3,856.70–2,266.5
CNRpcCNRpcPercentage (of TRL)0–57.10–49.8
 No. of axile rootsCNRcountCount0–20–2
 Cumulated length of axile rootsCNRsumCentimeters0–113.90–47.87
 Branch rootsCNRbranchCentimeters · (centimeters of axile root)−10–77.80–77.8
LNRs
 Length (including branch roots)LNRLCentimeters99.5–5,806.5 (×58.5)216.1–2,532.4 (×11.7)
LNRpcLNRpcPercentage (of TRL)4.2–72.7 (×17.5)6–64.8 (×10.9)
LNRcountLNRcountCount2–22.2 (×11.1)3.3–15.3 (×4.6)
LNRsumLNRsumCentimeters25.9–485.548–232 (×4.8)
 Branch rootsLNRbranchCentimeters · (centimeters of axile root)−12.1–25.4 (×12.1)3.2–15.9 (×5)
Open in a separate windowThe third reason for dissecting the different root types in this study was that they seem to have independent genetic regulation through major genes. Genes affecting specifically nodal root growth have been identified in maize (Hetz et al., 1996; Hochholdinger and Feix, 1998) and rice (Inukai et al., 2001, 2005; Liu et al., 2005, 2009; Zhao et al., 2009; Coudert et al., 2010; Gamuyao et al., 2012). Here, we also dissect branch (lateral) development on the seminal or nodal roots. Genes specific to branch roots have been identified in Arabidopsis (Casimiro et al., 2003; Péret et al., 2009a), rice (Hao and Ichii, 1999; Wang et al., 2006; Zheng et al., 2013), and maize (Hochholdinger and Feix, 1998; Hochholdinger et al., 2001; Woll et al., 2005).This study explored the hypothesis that adult root systems of B. distachyon contain genotypic variation that can be exploited through phenotyping and genotyping to increase cereal yields. A selection of 79 wild lines of B. distachyon from various parts of the Middle East (Fig. 2 shows the geographic origins of the lines) was phenotyped. They were selected for maximum genotypic diversity from 187 diploid lines analyzed with 43 simple sequence repeat markers (Vogel et al., 2009). We phenotyped shoots and mature root systems concurrently because B. distachyon is small enough to complete its life cycle in relatively small pots of soil with minimal influence of pot size compared with crops, such as wheat. We further phenotyped a subset of this population under irrigation (well watered) and drought to assess genotype response to water supply. By conducting whole-plant studies, we aimed to identify phenotypes that described partitioning among shoot and root components and within seed-borne and stem-borne roots. Phenotypes that have the potential to be beneficial to shoot and root components may speed up genetic gain in future.Open in a separate windowFigure 2.B. distachyon lines phenotyped in this study and their geographical origin. Capital letters in parentheses indicate the country of origin: Turkey (T), Spain (S), and Iraq (I; Vogel et al., 2009). a, Adi3, Adi7, Adi10, Adi12, Adi13, and Adi15; b, Bd21 and Bd21-3 are the reference lines of this study. Bd21 was the first sequenced line (Vogel et al., 2010) and root system (described in detail in Watt et al., 2009), and Bd21-3 is the most easily transformed line (Vogel and Hill, 2008) and parent of a T-DNA mutant population (Bragg et al., 2012); c, Gaz1, Gaz4, and Gaz7; d, Kah1, Kah2, and Kah3. e, Koz1, Koz3, and Koz5; f, Tek1 and Tek6; g, exact GPS coordinates are unknown for lines Men2 (S), Mur2 (S), Bd2.3 (I), Bd3-1 (I), and Abr1 (T).  相似文献   

13.
Characterization of a Thermostable Short-Chain Alcohol Dehydrogenase from the Hyperthermophilic Archaeon Thermococcus sibiricus     
Tatiana N. Stekhanova  Andrey V. Mardanov  Ekaterina Y. Bezsudnova  Vadim M. Gumerov  Nikolai V. Ravin  Konstantin G. Skryabin  Vladimir O. Popov 《Applied and environmental microbiology》2010,76(12):4096-4098
Short-chain alcohol dehydrogenase, encoded by the gene Tsib_0319 from the hyperthermophilic archaeon Thermococcus sibiricus, was expressed in Escherichia coli, purified and characterized as an NADPH-dependent enantioselective oxidoreductase with broad substrate specificity. The enzyme exhibits extremely high thermophilicity, thermostability, and tolerance to organic solvents and salts.Alcohol dehydrogenases (ADHs; EC 1.1.1.1.) catalyze the interconversion of alcohols to their corresponding aldehydes or ketones by using different redox-mediating cofactors. NAD(P)-dependent ADHs, due to their broad substrate specificity and enantioselectivity, have attracted particular attention as catalysts in industrial processes (5). However, mesophilic ADHs are unstable at high temperatures, sensitive to organic solvents, and often lose activity during immobilization. In this relation, there is a considerable interest in ADHs from extremophilic microorganisms; among them, Archaea are of great interest. The representatives of all groups of NAD(P)-dependent ADHs have been detected in genomes of Archaea (11, 12); however, only a few enzymes have been characterized, and the great majority of them belong to medium-chain (3, 4, 14, 16, 19) or long-chain iron-activated ADHs (1, 8, 9). Up to now, a single short-chain archaeal ADH from Pyrococcus furiosus (10, 18) and only one archaeal aldo-keto reductase also from P. furiosus (11) have been characterized.Thermococcus sibiricus is a hyperthermophilic anaerobic archaeon isolated from a high-temperature oil reservoir capable of growth on complex organic substrates (15). The complete genome sequence of T. sibiricus has been recently determined and annotated (13). Several ADHs are encoded by the T. sibiricus genome, including three short-chain ADHs (Tsib_0319, Tsib_0703, and Tsib_1998) (13). In this report, we describe the cloning and expression of the Tsib_0319 gene from T. sibiricus and the purification and the biochemical characterization of its product, the thermostable short-chain ADH (TsAdh319).The Tsib_0319 gene encodes a protein with a size of 234 amino acids and the calculated molecular mass of 26.2 kDa. TsAdh319 has an 85% degree of sequence identity with short-chain ADH from P. furiosus (AdhA; PF_0074) (18). Besides AdhA, close homologs of TsAdh319 were found among different bacterial ADHs, but not archaeal ADHs. The gene flanked by the XhoI and BamHI sites was PCR amplified using two primers (sense primer, 5′-GTTCTCGAGATGAAGGTTGCTGTGATAACAGGG-3′, and antisense primer, 5′-GCTGGATCCTCAGTATTCTGGTCTCTGGTAGACGG-3′) and cloned into the pET-15b vector. TsAdh319 was overexpressed, with an N-terminal His6 tag in Escherichia coli Rosetta-gami (DE3) and purified to homogeneity by metallochelating chromatography (Hi-Trap chelating HP column; GE Healthcare) followed by gel filtration on Superdex 200 10/300 GL column (GE Healthcare) equilibrated in 50 mM Tris-HCl (pH 7.5) with 200 mM NaCl. The homogeneity and the correspondence to the calculated molecular mass of 28.7 kDa were verified by SDS-PAGE (7). The molecular mass of native TsAdh319 was 56 to 60 kDa, which confirmed the dimeric structure in solution.The standard ADH activity measurement was made spectrophotometrically at the optimal pH by following either the reduction of NADP (in 50 mM Gly-NaOH buffer; pH 10.5) or the oxidation of NADPH (in 0.1 M sodium phosphate buffer; pH 7.5) at 340 nm at 60°C. The enzyme exhibited a strong preference for NADP(H) and broad substrate specificity (Table (Table1).1). The highest oxidation rates were found with pentoses d-arabinose (2.0 U mg−1) and d-xylose (2.46 U mg−1), and the highest reduction rates were found with dimethylglyoxal (5.9 U mg−1) and pyruvaldehyde (2.2 U mg−1). The enzyme did not reduce sugars which were good substrates for the oxidation reaction. The kinetic parameters of TsAdh319 determined for the preferred substrates are shown in Table Table2.2. The enantioselectivity of the enzyme was estimated by measuring the conversion rates of 2-butanol enantiomers. TsAdh319 showed an evident preference, >2-fold, for (S)-2-butanol over (RS)-2-butanol. The enzyme stereoselectivity is confirmed by the preferred oxidation of d-arabinose over l-arabinose (Table (Table1).1). The fact that TsAdh319 is metal independent was supported by the absence of a significant effect of TsAdh319 preincubation with 10 mM Me2+ for 30 min before measuring the activity in the presence of 1 mM Me2+ or EDTA (Table (Table3).3). TsAdh319 also exhibited a halophilic property, so the enzyme activity increased in the presence of NaCl and KCl and the activation was maintained even at concentration of 4 M and 3 M, respectively (Table (Table33).

TABLE 1.

Substrate specificity of TsAdh319
SubstrateaRelative activity (%)
Oxidation reactionb
    Methanol0
    2-Methoxyethanol0
    Ethanol36
    1-Butanol80
    2-Propanol100
    (RS)-(±)-2-Butanol86
    (S)-(+)-2-Butanol196
    2-Pentanol67
    1-Phenylmethanol180
    1.3-Butanediol91
    Ethyleneglycol0
    Glycerol16
    d-Arabinose*200
    l-Arabinose*17
    d-Xylose*246
    d-Ribose*35
    d-Glucose*146
    d-Mannose*48
    d-Galactose*0
    Cellobiose*71
Reduction reactionc
    Pyruvaldehyde100
    Dimethylglyoxal270
    Glyoxylic acid36
    Acetone0
    Cyclopentanone0
    Cyclohexanone4
    3-Methyl-2-pentanone*13
    d-Arabinose*0
    d-Xylose*0
    d-Glucose*0
    Cellobiose*0
Open in a separate windowaSubstrates were present in 250 mM or 50 mM (*) concentrations.bRelative rates, measured under standard conditions, were calculated by defining the activity for 2-propanol as 100%, which corresponds to 1.0 U mg−1. Data are averages from triplicate experiments.cRelative rates, measured under standard conditions, were calculated by defining the activity for pyruvaldehyde as 100%, which corresponds to 2.2 U mg−1. Data are averages from triplicate experiments.

TABLE 2.

Apparent Km and Vmax values for TsAdh319
Coenzyme or substrateApparent Km (mM)Vmax (U mg−1)kcat (s−1)
NADPa0.022 ± 0.0020.94 ± 0.020.45 ± 0.01
NADPHb0.020 ± 0.0033.16 ± 0.111.51 ± 0.05
2-Propanol168 ± 291.10 ± 0.090.53 ± 0.04
d-Xylose54.4 ± 7.41.47 ± 0.090.70 ± 0.04
Pyruvaldehyde17.75 ± 3.384.26 ± 0.402.04 ± 0.19
Open in a separate windowaActivity was measured under standard conditions with 2-propanol. Data are averages from triplicate experiments.bActivity was measured under standard conditions with pyruvaldehyde. Data are averages from triplicate experiments.

TABLE 3.

Effect of various ions and EDTA on TsAdh319a
CompoundConcn (mM)Relative activity (%)
None0100
NaCl400206
600227
4,000230
KCl600147
2,000200
3,000194
MgCl21078
CoCl210105
NiSO410100
ZnSO41079
FeSO41074
EDTA1100
580
Open in a separate windowaThe activity was measured under standard conditions with 2-propanol; relative rates were calculated by defining the activity without salts as 100%, which corresponds to 0.9 U mg−1. Data are averages from duplicate experiments.The most essential distinctions of TsAdh319 are the thermophilicity and high thermostability of the enzyme. The optimum temperature for the 2-propanol oxidation catalyzed by TsAdh319 was not achieved. The initial reaction rate of oxidation increased up to 100°C (Fig. (Fig.1).1). The Arrhenius plot is a straight line, typical of a single rate-limited thermally activated process, but there is no obvious transition point due to the temperature-dependent conformational changes of the protein molecule. The activation energy for the oxidation of 2-propanol was estimated at 84.0 ± 5.8 kJ·mol−1. The thermostability of TsAdh319 was calculated from residual TsAdh319 activity after preincubation of 0.4 mg/ml enzyme solution in 50 mM Tris-HCl buffer (pH 7.5) containing 200 mM NaCl at 70, 80, 90, or 100°C. The preincubation at 70°C or 80°C for 1.5 h did not cause a decrease in the TsAdh319 activity, but provoked slight activation. The residual TsAdh319 activities began to decrease after 2 h of preincubation at 70°C or 80°C and were 10% and 15% down from the control, respectively. The determined half-life values of TsAdh319 were 2 h at 90°C and 1 h at 100°C.Open in a separate windowFIG. 1.Temperature dependence of the initial rate of the 2-propanol reduction by TsAdh319. The reaction was initiated by enzyme addition to a prewarmed 2-propanol-NADP mixture. The inset shows the Arrhenius plot of the same data.Protein thermostability often correlates with such important biotechnological properties as increased solvent tolerance (2). We tested the influence of organic solvents at a high concentration (50% [vol/vol]) on TsAdh319 by using either preincubation of the enzyme at a concentration of 0.2 mg/ml with solvents for 4 h at 55°C or solvent addition into the reaction mixture to distinguish the effect of solvent on the protein stability and on the enzyme activity. TsAdh319 showed significant solvent tolerance in both cases (Table (Table4),4), and the effects of solvents could be modulated by salts, acting apparently as molecular lyoprotectants (17). Furthermore, TsAdh319 maintained 57% of its activity in 25% (vol/vol) 2-propanol, which could be used as the cosubstrate in cofactor regeneration (6).

TABLE 4.

Influence of various solvents on TsAdh319 activitya
SolventRelative activity (%)bRelative activity (%)c
Buffer without NaClBuffer with 600 mM NaCl
None100100100
DMSOd98040
DMFAe1011341
Methanol98259
Acetonitrile9500
Ethyl acetate470*33*
Chloroform10579*81*
n-Hexane10560*118*
n-Decane3691*107*
Open in a separate windowaThe activity measured at the standard condition with 2-propanol as a substrate. Data are averages from triplicate experiments.bPreincubation for 4 h at 55°C in the presence of 50% (vol/vol) of solvent prior the activity assay.cWithout preincubation, solvent addition to the reaction mixture up to 50% (vol/vol) or using the buffer saturated by a solvent (*).dDMSO, dimethyl sulfoxide.eDMFA, dimethylformamide.From all the aforesaid we may suppose TsAdh319 or its improved variant to be interesting both for the investigation of structural features of protein tolerance and for biotechnological applications.  相似文献   

14.
Extracytoplasmic Function σ Factors Regulate Expression of the Bacillus subtilis yabE Gene via a cis-Acting Antisense RNA     
Warawan Eiamphungporn  John D. Helmann 《Journal of bacteriology》2009,191(3):1101-1105
  相似文献   

15.
A Systematic Proteomic Analysis of Listeria monocytogenes House-keeping Protein Secretion Systems     
Sven Halbedel  Swantje Reiss  Birgit Hahn  Dirk Albrecht  Gopala Krishna Mannala  Trinad Chakraborty  Torsten Hain  Susanne Engelmann  Antje Flieger 《Molecular & cellular proteomics : MCP》2014,13(11):3063-3081
  相似文献   

16.
Analysis of the Clonal Relationship of Serotype O26:H11 Enterohemorrhagic Escherichia coli Isolates from Cattle     
Lutz Geue  Sabrina Klare  Christina Schnick  Birgit Mintel  Katharina Meyer  Franz J. Conraths 《Applied and environmental microbiology》2009,75(21):6947-6953
Twelve cluster groups of Escherichia coli O26 isolates found in three cattle farms were monitored in space and time. Cluster analysis suggests that only some O26:H11 strains had the potential for long-term persistence in hosts and farms. As judged by their virulence markers, bovine enterohemorrhagic O26:H11 isolates may represent a considerable risk for human infection.Shiga toxin (Stx)-producing Escherichia coli (STEC) strains comprise a group of zoonotic enteric pathogens (42). In humans, infections with some STEC serotypes result in hemorrhagic or nonhemorrhagic diarrhea, which can be complicated by hemolytic-uremic syndrome (HUS) (49). These STEC strains are also designated “enterohemorrhagic E. coli” (EHEC). Consequently, EHEC strains represent a subgroup of STEC with a high pathogenic potential for humans. Strains of the E. coli serogroup O26 were originally classified as enteropathogenic E. coli due to their association with outbreaks of infantile diarrhea in the 1940s. In 1977, Konowalchuk et al. (37) recognized that these bacteria produced Stx, and 10 years later, the Stx-producing E. coli O26:H11/H− strains were classified as EHEC. EHEC O26 strains constitute the most common non-O157 EHEC group associated with diarrhea and HUS in Europe (12, 21, 23, 24, 26, 27, 55, 60). Reports on an association between EHEC O26 and HUS or diarrhea from North America including the United States (15, 30, 33), South America (51, 57), Australia (22), and Asia (31, 32) provide further evidence for the worldwide spread of these organisms. Studies in Germany and Austria (26, 27) on sporadic HUS cases between 1996 and 2003 found that EHEC O26 accounted for 14% of all EHEC strains and for ∼40% of non-O157 EHEC strains obtained from these patients. A proportion of 11% EHEC O26 strains was detected in a case-control study in Germany (59) between 2001 and 2003. In the age group <3 years, the number of EHEC O26 cases was nearly equal to that of EHEC O157 cases, although the incidence of EHEC O26-associated disease is probably underestimated because of diagnostic limitations in comparison to the diagnosis of O157:H7/H− (18, 34). Moreover, EHEC O26 has spread globally (35). Beutin (6) described EHEC O26:H11/H−, among O103:H2, O111:H, O145:H28/H−, and O157:H7/H−, as the well-known pathogenic “gang of five,” and Bettelheim (5) warned that we ignore the non-O157 STEC strains at our peril.EHEC O26 strains produce Stx1, Stx2, or both (15, 63). Moreover, these strains contain the intimin-encoding eae gene (11, 63), a characteristic feature of EHEC (44). In addition, EHEC strains possess other markers associated with virulence, such as a large plasmid that carries further potential virulence genes, e.g., genes coding for EHEC hemolysin (EHEC-hlyA), a catalase-peroxidase (katP), and an extracellular serine protease (espP) (17, 52). The efa1 (E. coli factor for adherence 1) gene was identified as an intestinal colonization factor in EHEC (43). EHEC O26 represents a highly dynamic group of organisms that rapidly generate new pathogenic clones (7, 8, 63).Ruminants, especially cattle, are considered the primary reservoir for human infections with EHEC. Therefore, the aim of this study was the molecular characterization of bovine E. coli field isolates of serogroup O26 using a panel of typical virulence markers. The epidemiological situation in the beef herds from which the isolates were obtained and the spatial and temporal behavior of the clonal distribution of E. coli serogroup O26 were analyzed during the observation period. The potential risk of the isolates inducing disease in humans was assessed.In our study, 56 bovine E. coli O26:H11 isolates and one bovine O26:H32 isolate were analyzed for EHEC virulence-associated factors. The isolates had been obtained from three different beef farms during a long-term study. They were detected in eight different cattle in farm A over a period of 15 months (detected on 10 sampling days), in 3 different animals in farm C over a period of 8 months (detected on 3 sampling days), and in one cow on one sampling day in farm D (Table (Table1)1) (28).

TABLE 1.

Typing of E. coli O26 isolates
Sampling day, source, and isolateSerotypeVirulence profile by:
fliC PCR-RFLPstx1 genestx2 geneStx1 (toxin)Stx2 (toxin)Subtype(s)
efa1 genebEHEC-hlyA genekatP geneespP genePlasmid size(s) in kbCluster
stx1/stx2eaetirespAespB
Day 15
    Animal 6 (farm A)
        WH-01/06/002-1O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/06/002-2O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/06/002-3O26:H11H11++stx1ββββ+/++++110, 127
    Animal 8 (farm A)
        WH-01/08/002-2O26:H11H11++stx1ββββ+/++++110, 127
    Animal 26 (farm A)
        WH-01/26/001-2O26:H11H11++stx1ββββ+/++++130, 127
        WH-01/26/001-5O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/26/001-6O26:H11H11++stx1ββββ+/++++110, 127
        WH-01/26/001-7O26:H11H11++stx1ββββ+/−+++110, 127
Day 29
    Animal 2 (farm A)
        WH-01/02/003-1O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-2O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-5O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-6O26:H11H11++stx1ββββ+/+++110, 126
        WH-01/02/003-7O26:H11H11++stx1ββββ+/++++110, 126
        WH-01/02/003-8O26:H11H11++stx1ββββ−/++++110, 126
        WH-01/02/003-9O26:H11H11++stx1ββββ+/++++1106
        WH-01/02/003-10O26:H11H11++stx1ββββ+/++++1106
    Animal 26 (farm A)
        WH-01/26/002-2O26:H11H11++stx1ββββ+/++++130, 125
        WH-01/26/002-5O26:H11H11++stx1ββββ+/++++130, 125
        WH-01/26/002-8O26:H11H11++stx1ββββ+/++++130, 125
        WH-01/26/002-9O26:H11H11++stx1ββββ+/++110, 125
        WH-01/26/002-10O26:H11H11++stx1ββββ+/++++130, 125
Day 64
    Animal 20 (farm A)
        WH-01/20/005-3O26:H11H11++stx1ββββ+/+130, 2.52
Day 78
    Animal 29 (farm A)
        WH-01/29/002-1O26:H11H11++stx1ββββ+/−+130, 12, 2.54
        WH-01/29/002-2O26:H11H11++stx1ββββ+/++++130, 12, 2.54
        WH-01/29/002-3O26:H11H11++stx1ββββ+/++++130, 12, 2.54
        WH-01/29/002-4O26:H11H11++stx1ββββ+/++++130, 12, 2.54
        WH-01/29/002-5O26:H11H11++stx1ββββ+/++130, 12, 2.54
Day 106
    Animal 27 (farm A)
        WH-01/27/005-2O26:H11H11++stx1ββββ+/−+++145, 110, 123
        WH-01/27/005-5O26:H11H11++stx1ββββ+/++++130, 12, 2.55
        WH-01/27/005-6O26:H11H11++stx1ββββ+/+130, 12, 2.55
Day 113
    Animal 7 (farm C)
        WH-04/07/001-2O26:H11H11++++stx1/stx2ββββ+/+++55, 35, 2.511
        WH-04/07/001-4O26:H11H11++++stx1/stx2ββββ+/++++5512
        WH-04/07/001-6O26:H11H11++++stx1/stx2ββββ+/++++5512
Day 170
    Animal 22 (farm C)
        WH-04/22/001-1O26:H11H11++stx1ββββ+/++++110, 12, 6.312
        WH-04/22/001-4O26:H11H11++stx1ββββ+/++++110, 12, 6.312
        WH-04/22/001-5O26:H11H11++stx1ββββ+/++++110, 12, 6.312
Day 176
    Animal 14 (farm D)
        WH-03/14/004-8O26:H11H11++stx1ββββ+/+++11010
Day 218
    Animal 27 (farm A)
        WH-01/27/009-1O26:H11H11++++stx1/stx2ββββ+/++++110, 129
        WH-01/27/009-2O26:H11H11++++stx1/stx2ββββ+/++++110, 129
        WH-01/27/009-3O26:H11H11++++stx1/stx2ββββ+/++++110, 128
        WH-01/27/009-8O26:H11H11++++stx1/stx2ββββ+/++110, 128
        WH-01/27/009-9O26:H11H11++++stx1/stx2ββββ+/++++110, 129
Day 309
    Animal 29 (farm A)
        WH-01/29/010-1O26:H11H11++stx1ββββ+/++++110, 35, 124
        WH-01/29/010-2O26:H11H11++stx1ββββ+/++130, 55, 358
        WH-01/29/010-3O26:H11H11++stx1ββββ+/++++130, 35, 128
Day 365
    Animal 8 (farm C)
        WH-04/08/008-6O26:H11H11++stx1ββββ+/++++110, 5512
Day 379
    Animal 9 (farm A)
        WH-01/09/016-2O26:H32H32++stx1/stx2−/−145, 130, 1.81
    Animal 27 (farm A)
        WH-01/27/014-3O26:H11H11++stx1ββββ+/++++110, 129
        WH-01/27/014-4O26:H11H11++stx1ββββ+/++++110, 129
        WH-01/27/014-5O26:H11H11++stx1ββββ+/++++110, 128
Day 407
    Animal 29 (farm A)
        WH-01/29/013-4O26:H11H11++stx1ββββ+/++++110, 12, 2.58
        WH-01/29/013-7O26:H11H11++stx1ββββ+/++++110, 12, 2.58
Day 478
    Animal 27 (farm A)
        WH-01/27/017-1O26:H11H11++++stx1/stx2ββββ+/++++110, 128
        WH-01/27/017-5O26:H11H11++++stx1/stx2ββββ+/++++110, 128
        WH-01/27/017-6O26:H11H11++++stx1/stx2ββββ+/++++1108
        WH-01/27/017-7O26:H11H11++++stx1/stx2ββββ+/++++1108
        WH-01/27/017-10O26:H11H11+++stx1ββββ+/++++130, 12, 2.58
Open in a separate windowastx1/stx2, gene stx1 or stx2.befa1 was detected by two hybridizations (with lifA1-lifA2 and lifA3-lifA4 probes). +/+, complete gene; +/− or −/+, incomplete gene; −/−, efa1 negative.The serotyping of the O26 isolates was confirmed by the results of the fliC PCR-restriction fragment length polymorphism (RFLP) analysis performed according to Fields et al. (25), with slight modifications described by Zhang et al. (62). All O26:H11 isolates showed the H11 pattern described by Zhang et al. (62). In contrast, the O26:H32 isolate demonstrated a different fliC RFLP pattern that was identical to the H32 pattern described by the same authors. It has been demonstrated that EHEC O26:H11 strains belong to at least four different sequence types (STs) in the common clone complex 29 (39). In the multilocus sequence typing analysis for E. coli (61), the tested five EHEC O26:H11 isolates (WH-01/02/003-1, WH-01/20/005-3, WH-01/27/009-9, WH-03/14/004-8, and WH-04/22/001-1) of different farms and clusters were characterized as two sequence types (ST 21 and ST 396). The isolates from farms A and C belong to ST 21, the most frequent ST of EHEC O26:H11 isolates found in humans and animals (39), but the single isolate from farm D was characterized as ST 396.Typing and subtyping of genes (stx1 and/or stx2, eae, tir, espA, espB, EHEC-hlyA, katP, and espP) associated with EHEC were performed with LightCycler fluorescence PCR (48) and different block-cycler PCRs. To identify the subtypes of the stx2 genes and of the locus of enterocyte effacement-encoding genes eae, tir, espA, and espB, the PCR products were digested by different restriction endonucleases (19, 26, 46). The complete pattern of virulence markers was detected in most bovine isolates examined in our study. An stx1 gene was present in all O26 isolates. In addition, an stx2 gene was found in nine O26:H11 isolates in farm A and in three isolates of the same type in farm C, as well as in the O26:H32 isolate. Both Stx1 and Stx2 were closely related to families of Stx1 and Stx2 variants or alleles. EHEC isolates with stx2 genes are significantly more often associated with HUS and other severe disease manifestations than isolates with an stx1 gene, which are more frequently associated with uncomplicated diarrhea and healthy individuals (13). In contrast to STEC strains harboring stx2 gene variants, however, STEC strains of the stx2 genotype were statistically significantly associated with HUS (26). The stx2 genotype was found in all O26 isolates with an stx2 gene, while the GK3/GK4 amplification products after digestion with HaeIII and FokI restriction enzymes showed the typical pattern for this genotype described by Friedrich et al. (26). The nucleotide sequences of the A and B subunits of the stx2 gene of the selected bovine O26:H11 isolate WH-01/27/017-1 (GenBank accession no. EU700491) were identical to the stx2 genes of different sorbitol-fermenting EHEC O157:H− strains associated with human HUS cases and other EHEC infections in Germany (10) and 99.3% identical in their DNA sequences to the stx2 gene of the EHEC type strain EDL933, a typical O157:H7 isolate from an HUS patient. A characteristic stx1 genotype was present in all O26 isolates. The nucleotide sequences of the A and B subunits of the stx1 gene of the tested bovine O26:H11 isolate WH-01/27/017-1 (GenBank accession no. EU700490) were nearly identical to those of the stx1 genes of the EHEC O26:H11 reference type strains H19 and DEC10B, which had been associated with human disease outbreaks in Canada and Australia. Nucleotide exchanges typical for stx1c and stx1d subtypes as described by Kuczius et al. (38) were not found. All bovine O26:H11 strains produced an Stx1 with high cytotoxicity for Vero cells tested by Stx enzyme-linked immunosorbent assay and Vero cell neutralization assay (53). The Stx2 cytotoxicity for Vero cells was also very high in the O26:H11 isolates.Not only factors influencing the basic and inducible Stx production are important in STEC pathogenesis. It has been suggested that the eae and EHEC-hlyA genes are likely contributors to STEC pathogenicity (2, 3, 13, 50). Ritchie et al. (50) found both genes in all analyzed HUS-associated STEC isolates. In all O26:H11 isolates we obtained, stx genes were present in combination with eae genes. Only the O26:H32 isolate lacked an eae gene. To date, 10 distinct variants of eae have been described (1, 19, 36, 45, 47). Some serotypes were closely associated with a particular intimin variant: the O157 serogroup was linked to γ-eae, the O26 serogroup to β-eae, and the O103 serogroup to ɛ-eae (4, 19, 20, 58). Our study confirms these associations. All bovine O26:H11 isolates were also typed as members of the β-eae subgroup. A translocated intimin receptor gene (tir gene) and the type III secreted proteins encoded by the espA and espB genes were found in all 56 O26:H11 isolates but not in the O26:H32 isolate. These other tested locus of enterocyte effacement-associated genes belonged to the β-subgroups. These results are in accord with the results of China et al. (19), who detected the pathotypes β-eae, β-tir, β-espA, and β-espB in all investigated human O26 strains. Like the eae gene, the EHEC-hlyA gene was found in association with severe clinical disease in humans (52). Aldick et al. (2) showed that EHEC hemolysin is toxic (cytolytic) to human microvascular endothelial cells and may thus contribute to the pathogenesis of HUS. In our study, the EHEC-hlyA gene was detected in 50 of the 56 bovine E. coli O26:H11 isolates which harbored virulence-associated plasmids of different sizes (Table (Table1).1). The presence of virulence-associated plasmids corresponded to the occurrence of additional virulence markers such as the espP and katP genes (17). The katP gene and the espP gene were detected in 49 and 50 of the 56 O26:H11 isolates, respectively. The espP gene was missing in six of the seven bovine O26:H11 isolates in which the katP genes were also absent. Both genes were not found in the O26:H32 isolate (Table (Table1).1). Although we found large plasmids of the same size in O26:H11 isolates, they lacked one or more of the plasmid-associated virulence factors (Table (Table1).1). Two DNA probes were used to detect the efa1 genes by colony hybridization. (DNA probes were labeled with digoxigenin [DIG] with lifA1-lifA2 and lifA3-lifA4 primers [14] using the PCR DIG probe synthesis kit [Roche Diagnostics, Mannheim, Germany]; DIG Easy Hyb solution [Roche] was used for prehybridization and hybridization.) Positive results with both DNA probes were obtained for 52 of 56 E. coli O26:H11 isolates. A positive signal was only found in three isolates with the lifA1-lifA2 DNA probe and in one isolate with the lifA3-lifA4 probe. An efa1 gene was not detected in the O26:H32 isolate (Table (Table11).We also analyzed the spatial and temporal behavior of the O26:H11/H32 isolates in the beef herds by cluster analysis (conducted in PAUP* for Windows version 4.0, 2008 [http://paup.csit.fsu.edu/about.html]). This was performed with distance matrices using the neighbor-joining algorithm, an agglomerative cluster method which generates a phylogenetic tree. The distance matrices were calculated by pairwise comparisons of the fragmentation patterns produced by genomic typing through pulsed-field gel electrophoresis analysis with four restriction endonucleases (XbaI, NotI, BlnI, and SpeI) and the presence or absence of potential virulence markers (Fig. (Fig.11 and Table Table1).1). To this end, the total character difference was used, which counts the pairwise differences between two given patterns. During a monitoring program of 3 years in four cattle farms (29), different O26:H11 cluster groups and one O26:H32 isolate were detected in three different farms. The genetic distance of the O26:H32 isolate was very high relative to the O26:H11 isolates. Therefore, the O26:H32 isolate was outgrouped. The O26:H11 isolates of each farm represented independent cluster groups. The single isolate from farm D fitted better to the isolates from farm C than to those from farm A. This finding is in accord with the geographical distance between the farms. The fact that the farms were located in neighboring villages may suggest that direct or indirect connections between the farms were possible (e.g., by person contacts or animal trade). However, the isolates from farm C and farm D belonged to different sequence types (ST 21 and ST 396), which may argue against a direct connection. Interestingly, O26:H11 isolates with and without stx2 genes were detected in the same clusters. This phenomenon was observed in both farm A and farm C. In farm A, the isolates with additional stx2 genes were found in animal 27 and were grouped in clusters 8 and 9 (day 218). An stx2 gene was repeatedly found (four isolates) in the same animal (animal 27). The isolates grouped in cluster 8 on a later day of sampling (day 478). All other O26:H11 isolates grouped in the same clusters and obtained from the same animals (27 and 29) on different sampling days lacked an stx2 gene. Also, the isolates obtained from animal 27 on previous sampling days, which grouped in clusters 3 and 5, exhibited no stx2 genes. In farm C, the three isolates with additional stx2 genes obtained from animal 7 grouped in clusters 11 and 12. An stx2 gene was absent from all other O26:H11 isolates grouped in the same cluster 12 on later sampling days, and no other isolates of cluster 11 were found later on. However, we detected members of many clusters over relatively long periods (clusters 5, 8, and 9 in farm A and cluster 12 in farm C), but members of other clusters were only found on single occasions. This patchy temporal pattern is apparently not a unique property of O26:H11, as we found similar results for cluster groups of other EHEC serotypes of bovine origin (28). The isolates grouped in the dominant cluster 8 were found on 5 of 9 sampling days over a period of 10 months. In contrast, we found the members of clusters 4, 5, 9, and 12 only on two nonconsecutive sampling days. The period during which isolates of these groups were not detected was particularly long for cluster 4 (231 days). We also observed the coexistence of different clusters over long periods in the same farm and in the same cattle (clusters 8 and 9), while one of the clusters dominated. Transmission of clusters between cattle was also observed. These results suggest that some of the EHEC O26:H11 strains had the potential for a longer persistence in the host population, while others had not. The reasons for this difference are not yet clear. Perhaps the incomplete efa1 gene found in isolates of clusters which were only detected once might explain why some strains disappeared rapidly. Efa1 has been discussed as a potential E. coli colonization factor for the bovine intestine used by non-O157 STEC, including O26 (54, 56). The O165:H25 cluster detected during a longer period in farm B may have disappeared after it had lost its efa1 gene (28). The precise biological activity of Efa1 in EHEC O26 is not yet known, but it has been demonstrated that the molecule is a non-Stx virulence determinant which can increase the virulence of EHEC O26 in humans (8).Open in a separate windowFIG. 1.Neighbor-joining tree of bovine E. coli O26:H11/H32 strains based on the restriction pattern obtained after digestion with XbaI, NotI, BlnI, and SpeI.We distinguished 12 different clusters, but complete genetic identity was only found in two isolates. The variations in the O26:H11 clusters may be due to increasing competition between the bacterial populations of the various subtypes in the bovine intestine or to potential interactions between EHEC O26:H11 and the host.The ephemeral occurrence of additional stx2 genes in different clusters and farms may be the result of recombination events due to horizontal gene transfer (16). The loss of stx genes may occur rapidly in the course of an infection, but the reincorporation by induction of an stx-carrying bacteriophage into the O26:H11 strains is possible at any time (9, 40). Nevertheless, an additional stx2 gene may increase the dangerousness of the respective EHEC O26:H11 strains. While all patients involved in an outbreak caused by an EHEC O26:H11 strain harboring the gene encoding Stx2 developed HUS (41), the persons affected by another outbreak caused by an EHEC O26:H11 strain that produced exclusively Stx1 had only uncomplicated diarrhea (60).In conclusion, our results showed that bovine O26:H11 isolates can carry virulence factors of EHEC that are strongly associated with EHEC-related disease in humans, particularly with severe clinical manifestations such as hemorrhagic colitis and HUS. Therefore, strains of bovine origin may represent a considerable risk for human infection. Moreover, some clusters of EHEC O26:H11 persisted in cattle and farms over longer periods, which may increase the risk of transmission to other animals and humans even further.  相似文献   

17.
Identification of Enhancer Binding Proteins Important for Myxococcus xanthus Development     
Krista M. Giglio  Jessica Eisenstatt  Anthony G. Garza 《Journal of bacteriology》2010,192(1):360-364
  相似文献   

18.
Isolation and Characterization of Xenorhabdus nematophila Transposon Insertion Mutants Defective in Lipase Activity against Tween     
Gregory R. Richards  Eugenio I. Vivas  Aaron W. Andersen  Delmarie Rivera-Santos  Sara Gilmore  Garret Suen  Heidi Goodrich-Blair 《Journal of bacteriology》2009,191(16):5325-5331
  相似文献   

19.
Escherichia coli O157:H7 and Other E. coli Strains Share Physiological Properties Associated with Intestinal Colonization     
Lisa Jacobsen  Lisa Durso  Tyrell Conway  Kenneth W. Nickerson 《Applied and environmental microbiology》2009,75(13):4633-4635
Escherichia coli isolates (72 commensal and 10 O157:H7 isolates) were compared with regard to physiological and growth parameters related to their ability to survive and persist in the gastrointestinal tract and found to be similar. We propose that nonhuman hosts in E. coli O157:H7 strains function similarly to other E. coli strains in regard to attributes relevant to gastrointestinal colonization.Escherichia coli is well known for its ecological versatility (15). A life cycle which includes both gastrointestinal and environmental stages has been stressed by both Savageau (15) and Adamowicz et al. (1). The gastrointestinal stage would be subjected to acid and detergent stress. The environmental stage is implicit in E. coli having transport systems for fungal siderophores (4) as well as pyrroloquinoline quinone-dependent periplasmic glucose utilization (1) because their presence indicates evolution in a location containing fungal siderophores and pyrroloquinoline quinone (1).Since its recognition as a food-borne pathogen, there have been numerous outbreaks of food-borne infection due to E. coli O157:H7, in both ground beef and vegetable crops (6, 13). Cattle are widely considered to be the primary reservoir of E. coli O157:H7 (14), but E. coli O157:H7 does not appear to cause disease in cattle. To what extent is E. coli O157:H7 physiologically unique compared to the other naturally occurring E. coli strains? We feel that the uniqueness of E. coli O157:H7 should be evaluated against a backdrop of other wild-type E. coli strains, and in this regard, we chose the 72-strain ECOR reference collection originally described by Ochman and Selander (10). These strains were chosen from a collection of 2,600 E. coli isolates to provide diversity with regard to host species, geographical distribution, and electromorph profiles at 11 enzyme loci (10).In our study we compared the 72 strains of the ECOR collection against 10 strains of E. coli O157:H7 and six strains of E. coli which had been in laboratory use for many years (Table (Table1).1). The in vitro comparisons were made with regard to factors potentially relevant to the bacteria''s ability to colonize animal guts, i.e., acid tolerance, detergent tolerance, and the presence of the Entner-Doudoroff (ED) pathway (Table (Table2).2). Our longstanding interest in the ED pathway (11) derives in part from work by Paul Cohen''s group (16, 17) showing that the ED pathway is important for E. coli colonization of the mouse large intestine. Growth was assessed by replica plating 88 strains of E. coli under 40 conditions (Table (Table2).2). These included two LB controls (aerobic and anaerobic), 14 for detergent stress (sodium dodecyl sulfate [SDS], hexadecyltrimethylammonium bromide [CTAB], and benzalkonium chloride, both aerobic and anaerobic), 16 for acid stress (pH 6.5, 6.0, 5.0, 4.6, 4.3, 4.2, 4.1, and 4.0), four for the ability to grow in a defined minimal medium (M63 glucose salts with and without thiamine), and four for the presence or absence of a functional ED pathway (M63 with gluconate or glucuronate). All tests were done with duplicate plates in two or three separate trials. The data are available in Tables S1 to S14 in the supplemental material, and they are summarized in Table Table22.

TABLE 1.

E. coli strains used in this study
E. coli strain (n)Source
ECOR strains (72)Thomas Whittman
Laboratory adapted (6)
    K-12 DavisPaul Blum
    CG5C 4401Paul Blum
    K-12 StanfordPaul Blum
    W3110Paul Blum
    BTyler Kokjohn
    AB 1157Tyler Kokjohn
O157:H7 (10)
    FRIK 528Andrew Benson
    ATCC 43895Andrew Benson
    MC 1061Andrew Benson
    C536Tim Cebula
    C503Tim Cebula
    C535Tim Cebula
    ATCC 43889William Cray, Jr.
    ATCC 43890William Cray, Jr.
    ATCC 43888Willaim Cray, Jr.
    ATCC 43894William Cray, Jr.
Open in a separate window

TABLE 2.

Physiological comparison of 88 strains of Escherichia coli
Growth medium or conditionOxygencNo. of strains with type of growthb
ECOR strains (n = 72)
Laboratory strains (n = 6)
O157:H7 strains (n = 10)
GoodPoorNoneVariableGoodPoorNoneVariableGoodPoorNoneVariable
LB controlaBoth72000600010000
1% SDSAerobic6930060008002
5% SDSAerobic6840060008200
1% SDSAnaerobic53154023101702
5% SDSAnaerobic0684004200704
CTABd (all)Both00720006000100
0.05% BACAerobic31158202220091
0.2% BACAerobic01710105000100
0.05% BACAnaerobic2367001500091
0.2% BACAnaerobic00720006000100
pH 6.5Both72000600010000
pH 6Both72000600010000
pH 5Both7020060009001
pH 4.6Both70200600010000
pH 4.3Aerobic14015731203205
pH 4.3Anaerobic6930031201100
pH 4.1 or 4.2Aerobic00720NDgND
pH 4.0Both0072000600091
M63 with supplemente
    GlucoseAerobicf6912050109010
    GlucoseAnaerobicf7002050109010
    GluconateBoth6912050109010
    GlucuronateAerobic6822050109010
    GlucuronateAnaerobic6912050109010
Open in a separate windowaEight LB controls were run, two for each set of LB experiments: SDS, CTAB, benzalkonium chloride (BAC), and pH stress.bGrowth was measured as either +++, +, or 0 (good, poor, and none, respectively), with +++ being the growth achieved on the LB control plates. “Variable” means that two or three replicates did not agree. All experiments were done at 37°C.c“Anaerobic” refers to use of an Oxoid anaerobic chamber. Aerobic and anaerobic growth data are presented together when the results were identical and separately when the results were not the same or the anaerobic set had not been done. LB plates were measured after 1 (aerobic) or 2 (anaerobic) days, and the M63 plates were measured after 2 or 3 days.dCTAB used at 0.05, 0.2%, and 0.4%.eM63 defined medium (3) was supplemented with glucose, gluconate, or glucuronate, all at 0.2%.fIdentical results were obtained with and without 0.0001% thiamine.gND, not determined.  相似文献   

20.
Focus on Metabolism: Posttranslational Protein Modifications in Plant Metabolism     
Giulia Friso  Klaas J. van Wijk 《Plant physiology》2015,169(3):1469-1487
Posttranslational modifications (PTMs) of proteins greatly expand proteome diversity, increase functionality, and allow for rapid responses, all at relatively low costs for the cell. PTMs play key roles in plants through their impact on signaling, gene expression, protein stability and interactions, and enzyme kinetics. Following a brief discussion of the experimental and bioinformatics challenges of PTM identification, localization, and quantification (occupancy), a concise overview is provided of the major PTMs and their (potential) functional consequences in plants, with emphasis on plant metabolism. Classic examples that illustrate the regulation of plant metabolic enzymes and pathways by PTMs and their cross talk are summarized. Recent large-scale proteomics studies mapped many PTMs to a wide range of metabolic functions. Unraveling of the PTM code, i.e. a predictive understanding of the (combinatorial) consequences of PTMs, is needed to convert this growing wealth of data into an understanding of plant metabolic regulation.The primary amino acid sequence of proteins is defined by the translated mRNA, often followed by N- or C-terminal cleavages for preprocessing, maturation, and/or activation. Proteins can undergo further reversible or irreversible posttranslational modifications (PTMs) of specific amino acid residues. Proteins are directly responsible for the production of plant metabolites because they act as enzymes or as regulators of enzymes. Ultimately, most proteins in a plant cell can affect plant metabolism (e.g. through effects on plant gene expression, cell fate and development, structural support, transport, etc.). Many metabolic enzymes and their regulators undergo a variety of PTMs, possibly resulting in changes in oligomeric state, stabilization/degradation, and (de)activation (Huber and Hardin, 2004), and PTMs can facilitate the optimization of metabolic flux. However, the direct in vivo consequence of a PTM on a metabolic enzyme or pathway is frequently not very clear, in part because it requires measurements of input and output of the reactions, including flux through the enzyme or pathway. This Update will start out with a short overview on the major PTMs observed for each amino acid residue (PTMs, including determination of the localization within proteins (i.e. the specific residues) and occupancy. Challenges in dealing with multiple PTMs per protein and cross talk between PTMs will be briefly outlined. We then describe the major physiological PTMs observed in plants as well as PTMs that are nonenzymatically induced during sample preparation (PTMs, in particular for enzymes in primary metabolism (Calvin cycle, glycolysis, and respiration) and the C4 shuttle accommodating photosynthesis in C4 plants (PTMs observed in plants
Amino Acid ResidueObserved Physiological PTM in PlantsPTMs Caused by Sample Preparation
Ala (A)Not known
Arg (R)Methylation, carbonylation
Asn (N)Deamidation, N-linked gycosylationDeamidation
Asp (D)Phosphorylation (in two-component system)
Cys (C)Glutathionylation (SSG), disulfide bonded (S-S), sulfenylation (-SOH), sulfonylation (-SO3H), acylation, lipidation, acetylation, nitrosylation (SNO), methylation, palmitoylation, phosphorylation (rare)Propionamide
Glu (E)Carboxylation, methylationPyro-Glu
Gln (Q)DeamidationDeamidation, pyro-Glu
Gly (G)N-Myristoylation (N-terminal Gly residue)
His (H)Phosphorylation (infrequent)Oxidation
Ile (I)Not known
Leu (L)Not known
Lys (K)N-ε-Acetylation, methylation, hydroxylation, ubiquitination, sumoylation, deamination, O-glycosylation, carbamylation, carbonylation, formylation
Met (M)(De)formylation, excision (NME), (reversible) oxidation, sulfonation (-SO2), sulfoxation (-SO)Oxidation, 2-oxidation, formylation, carbamylation
Phe (F)Not known
Pro (P)CarbonylationOxidation
Ser (S)Phosphorylation, O-linked glycosylation, O-linked GlcNAc (O-GlcNAc)Formylation
Thr (T)Phosphorylation, O-linked glycosylation, O-linked GlcNAc (O-GlcNAc), carbonylationFormylation
Trp (W)Glycosylation (C-mannosylation)Oxidation
Tyr (Y)Phosphorylation, nitration
Val (V)Not known
Free NH2 of protein N terminiPreprotein processing, Met excision, formylation, pyro-Glu, N-myristoylation, N-acylation (i.e. palmitoylation), N-terminal α-amine acetylation, ubiquitinationFormylation (Met), pyro-Glu (Gln)
Open in a separate window

Table II.

Most significant and/or frequent PTMs observed in plants
Type of PTM (Reversible, Except if Marked with an Asterisk)Spontaneous (S; Nonenzymatic) or Enzymatic (E)Comment on Subcellular Location and Frequency
Phosphorylation (Ser, Thr, Tyr, His, Asp)EHis and Asp phosphorylation have low frequency
S-Nitrosylation (Cys) and nitration* (Tyr)S (RNS), but reversal is enzymatic for Cys by thioredoxinsThroughout the cell
Acetylation (N-terminal α-amine, Lys ε-amine)EIn mitochondria, very little N-terminal acetylation, but high Lys acetylation; Lys acetylation correlates to [acetyl-CoA]
Deamidation (Gln, Asn)S, but reversal of isoAsp is enzymatic by isoAsp methyltransferaseThroughout the cell
Lipidation (S-acetylation, N-meristoylation*, prenylation*; Cys, Gly, Lys, Trp, N terminal)ENot (or rarely) within plastids, mitochondria, peroxisomes
N-Linked glycosylation (Asp); O linked (Lys, Ser, Thr, Trp)EOnly proteins passing through the secretory system; O linked in the cell wall
Ubiquination (Lys, N terminal)ENot within plastids, mitochondria, peroxisomes
Sumoylation (Lys)ENot within plastids, mitochondria, peroxisomes
Carbonylation* (Pro, Lys, Arg, Thr)S (ROS)High levels in mitochondria and chloroplast
Methylation (Arg, Lys, N terminal)EHistones (nucleus) and chloroplasts; still underexplored
Glutathionylation (Cys)EHigh levels in chloroplasts
Oxidation (Met, Cys)S (ROS) and E (by PCOs; see Fig. 1B), but reversal is enzymatic by Met sulfoxide reductases, glutaredoxins, and thioredoxins, except if double oxidizedHigh levels in mitochondria and chloroplast
Peptidase* (cleavage peptidyl bond)EThroughout the cell
S-Guanylation (Cys)S (RNS)Rare; 8-nitro-cGMP is signaling molecule in guard cells
Formylation (Met)S, but deformylation is enzymatic by peptide deformylaseAll chloroplasts and mitochondria-encoded proteins are synthesized with initiating formylated Met
Open in a separate window

Table III.

Regulation by PTMs in plant metabolism and classic examples of well-studied enzymes and pathwaysMany of these enzymes also undergo allosteric regulation through cellular metabolites. GAPDH, Glyceraldehyde-3-phosphate dehydrogenase; PRK, phosphoribulokinase.
ProcessEnzymesPTMs, Protein Modifiers, LocalizationReferences
Calvin-Benson cycle (chloroplasts)Many enzymesOxidoreduction of S-S bonds, reversible nitrosylation, glutathionylation; through ferredoxin/ferredoxin-thioredoxin reductase/thioredoxins (mostly f and m) and glutaredoxins; proteomics studies in Arabidopsis and C. reinhardtiiMichelet et al. (2013)
RubiscoMethylation, carbamylation, acetylation, N-terminal processing, oligomerization; classical studies in pea (Pisum sativum), spinach (Spinacia oleracea), and ArabidopsisHoutz and Portis (2003); Houtz et al. (2008)
GAPDH/CP12/PRK supercomplexDynamic heterooligomerization through reversible S-S bond formation controlled by thioredoxinsGraciet et al. (2004); Michelet et al. (2013); López-Calcagno et al. (2014)
GlycolysisCytosolic PEPCPhosphorylation (S, T), monoubiquitinationO’Leary et al. (2011)
PhotorespirationSeven enzymes are phosphorylatedPhosphorylation from meta-analysis of public phosphoproteomics data for Arabidopsis; located in chloroplasts, peroxisomes, mitochondriaHodges et al. (2013)
Maize glycerate kinaseRedox-regulated S-S bond; thioredoxin f; studied extensively in chloroplasts of C4 maizeBartsch et al. (2010)
Respiration (mitochondria)Potentially many enzymes, but functional/biochemical consequences are relatively unexploredRecent studies suggested PTMs for many tricarboxylic acid cycle enzymes, including Lys acetylation and thioredoxin-driven S-S formation; in particular, succinate dehydrogenase and fumarase are inactivated by thioredoxinsLázaro et al. (2013); Schmidtmann et al. (2014); Daloso et al. (2015)
PDHSer (de)phosphorylation by intrinsic kinase and phosphatase; ammonia and pyruvate control PDH kinase activity; see Figure 1BThelen et al. (2000); Tovar-Méndez et al. (2003)
C4 cycle (C3 and C4 homologs also involved in glycolysis and/or gluconeogenesis)Pyruvate orthophosphate dikinasePhosphorylation by pyruvate orthophosphate dikinase-RP, an S/T bifunctional kinase-phosphatase; in chloroplastsChastain et al. (2011); Chen et al. (2014)
PEPCPhosphorylation; allosteric regulation by malate and Glc-6-P; in cytosol in mesophyll cells in C4 species (e.g. Panicum maximum); see Figure 1AIzui et al. (2004); Bailey et al. (2007)
PEPC kinaseUbiquitination resulting in degradation (note also diurnal mRNA levels and linkage to activity level; very low protein level); in cytosol in mesophyll cells in C4 species (e.g. Flaveria spp. and maize)Agetsuma et al. (2005)
PEPC kinasePhosphorylation in cytosol in bundle sheath cellsBailey et al. (2007)
Starch metabolism (chloroplasts)ADP-Glc pyrophosphorylaseRedox-regulated disulfide bonds and dynamic oligomerization; thioredoxins; see Figure 1CGeigenberger et al. (2005); Geigenberger (2011)
Starch-branching enzyme IIPhosphorylation by Ca2+-dependent protein kinase; P-driven heterooligomerizationGrimaud et al. (2008); Tetlow and Emes (2014)
Suc metabolism (cytosol)SPS (synthesis of Suc)(De)phosphorylation; SPS kinase and SPS phosphatase; 14-3-3 proteins; cytosol (maize and others)Huber (2007)
Suc synthase (breakdown of Suc)Phosphorylation; Ca2+-dependent protein kinase; correlations to activity, localization, and turnoverDuncan and Huber (2007); Fedosejevs et al. (2014)
Photosynthetic electron transport (chloroplast thylakoid membranes)PSII core and light-harvesting complex proteins(De)phosphorylation by state-transition kinases (STN7/8) and PP2C phosphatases (PBCP and PPH1/TAP38)Pesaresi et al. (2011); Tikkanen et al. (2012); Rochaix (2014)
Nitrogen assimilationNitrate reductase(De)phosphorylation; 14-3-3 proteinsLillo et al. (2004); Huber (2007)
Open in a separate windowThere are many recent reviews focusing on specific PTMs in plant biology, many of which are cited in this Update. However, the last general review on plant PTMs is from 2010 (Ytterberg and Jensen, 2010); given the enormous progress in PTM research in plants over the last 5 years, a comprehensive overview is overdue. Finally, this Update does not review allosteric regulation by metabolites or other types of metabolic feedback and flux control, even if this is extremely important in the regulation of metabolism and (de)activation of enzymes. Recent reviews for specific pathways, such as isoprenoid metabolism (Kötting et al., 2010; Banerjee and Sharkey, 2014; Rodríguez-Concepción and Boronat, 2015), tetrapyrrole metabolism (Brzezowski et al., 2015), the Calvin-Benson cycle (Michelet et al., 2013), starch metabolism (Kötting et al., 2010; Geigenberger, 2011; Tetlow and Emes, 2014), and photorespiration (Hodges et al., 2013) provide more in-depth discussions of metabolic regulation through various posttranslational mechanisms. Many of the PTMs that have been discovered in the last decade through large-scale proteomics approaches have not yet been integrated in such pathway-specific reviews, because these data are not always easily accessible and because the biological significance of many PTMs is simply not yet understood. We hope that this Update will increase the general awareness of the existence of these PTM data sets, such that their biological significance can be tested and incorporated in metabolic pathways.  相似文献   

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