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 46 毫秒
1.
2.
3.
4.
Lichenysins are surface-active lipopeptides with antibiotic properties produced nonribosomally by several strains of Bacillus licheniformis. Here, we report the cloning and sequencing of an entire 26.6-kb lichenysin biosynthesis operon from B. licheniformis ATCC 10716. Three large open reading frames coding for peptide synthetases, designated licA, licB (three modules each), and licC (one module), could be detected, followed by a gene, licTE, coding for a thioesterase-like protein. The domain structure of the seven identified modules, which resembles that of the surfactin synthetases SrfA-A to -C, showed two epimerization domains attached to the third and sixth modules. The substrate specificity of the first, fifth, and seventh recombinant adenylation domains of LicA to -C (cloned and expressed in Escherichia coli) was determined to be Gln, Asp, and Ile (with minor Val and Leu substitutions), respectively. Therefore, we suppose that the identified biosynthesis operon is responsible for the production of a lichenysin variant with the primary amino acid sequence l-Gln–l-Leu–d-Leu–l-Val–l-Asp–d-Leu–l-Ile, with minor Leu and Val substitutions at the seventh position.Many strains of Bacillus are known to produce lipopeptides with remarkable surface-active properties (11). The most prominent of these powerful lipopeptides is surfactin from Bacillus subtilis (1). Surfactin is an acylated cyclic heptapeptide that reduces the surface tension of water from 72 to 27 mN m−1 even in a concentration below 0.05% and shows some antibacterial and antifungal activities (1). Some B. subtilis strains are also known to produce other, structurally related lipoheptapeptides (Table (Table1),1), like iturin (32, 34) and bacillomycin (3, 27, 30), or the lipodecapeptides fengycin (50) and plipastatin (29).

TABLE 1

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

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

TABLE 1

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

8.
9.
Forty-five different point mutations in POLG, the gene encoding the catalytic subunit of the human mitochondrial DNA polymerase (pol γ), cause the early onset mitochondrial DNA depletion disorder, Alpers syndrome. Sequence analysis of the C-terminal polymerase region of pol γ revealed a cluster of four Alpers mutations at highly conserved residues in the thumb subdomain (G848S, c.2542g→a; T851A, c.2551a→g; R852C, c.2554c→t; R853Q, c.2558g→a) and two Alpers mutations at less conserved positions in the adjacent palm subdomain (Q879H, c.2637g→t and T885S, c.2653a→t). Biochemical characterization of purified, recombinant forms of pol γ revealed that Alpers mutations in the thumb subdomain reduced polymerase activity more than 99% relative to the wild-type enzyme, whereas the palm subdomain mutations retained 50–70% wild-type polymerase activity. All six mutant enzymes retained physical and functional interaction with the pol γ accessory subunit (p55), and none of the six mutants exhibited defects in misinsertion fidelity in vitro. However, differential DNA binding by these mutants suggests a possible orientation of the DNA with respect to the polymerase during catalysis. To our knowledge this study represents the first structure-function analysis of the thumb subdomain in pol γ and examines the consequences of mitochondrial disease mutations in this region.As the only DNA polymerase found in animal cell mitochondria, DNA polymerase γ (pol γ)3 bears sole responsibility for DNA synthesis in all replication and repair transactions involving mitochondrial DNA (1, 2). Mammalian cell pol γ is a heterotrimeric complex composed of one catalytic subunit of 140 kDa (p140) and two 55-kDa accessory subunits (p55) that form a dimer (3). The catalytic subunit contains an N-terminal exonuclease domain connected by a linker region to a C-terminal polymerase domain. Whereas the exonuclease domain contains essential motifs I, II, and III for its activity, the polymerase domain comprising the thumb, palm, and finger subdomains contains motifs A, B, and C that are crucial for polymerase activity. The catalytic subunit is a family A DNA polymerase that includes bacterial pol I and T7 DNA polymerase and possesses DNA polymerase, 3′ → 5′ exonuclease, and 5′-deoxyribose phosphate lyase activities (for review, see Refs. 1 and 2). The 55-kDa accessory subunit (p55) confers processive DNA synthesis and tight binding of the pol γ complex to DNA (4, 5).Depletion of mtDNA as well as the accumulation of deletions and point mutations in mtDNA have been observed in several mitochondrial disorders (for review, see Ref. 6). mtDNA depletion syndromes are caused by defects in nuclear genes responsible for replication and maintenance of the mitochondrial genome (7). Mutation of POLG, the gene encoding the catalytic subunit of pol γ, is frequently involved in disorders linked to mutagenesis of mtDNA (8, 9). Presently, more than 150 point mutations in POLG are linked with a wide variety of mitochondrial diseases, including the autosomal dominant (ad) and recessive forms of progressive external ophthalmoplegia (PEO), Alpers syndrome, parkinsonism, ataxia-neuropathy syndromes, and male infertility (tools.niehs.nih.gov/polg) (9).Alpers syndrome, a hepatocerebral mtDNA depletion disorder, and myocerebrohepatopathy are rare heritable autosomal recessive diseases primarily affecting young children (1012). These diseases generally manifest during the first few weeks to years of life, and symptoms gradually develop in a stepwise manner eventually leading to death. Alpers syndrome is characterized by refractory seizures, psychomotor regression, and hepatic failure (11, 12). Mutation of POLG was first linked to Alpers syndrome in 2004 (13), and to date 45 different point mutations in POLG (18 localized to the polymerase domain) are associated with Alpers syndrome (9, 14, 15). However, only two Alpers mutations (A467T and W748S, both in the linker region) have been biochemically characterized (16, 17).During the initial cloning and sequencing of the human, Drosophila, and chicken pol γ genes, we noted a highly conserved region N-terminal to motif A in the polymerase domain that was specific to pol γ (18). This region corresponds to part of the thumb subdomain that tracks DNA into the active site of both Escherichia coli pol I and T7 DNA polymerase (1921). A high concentration of disease mutations, many associated with Alpers syndrome, is found in the thumb subdomain.Here we investigated six mitochondrial disease mutations clustered in the N-terminal portion of the polymerase domain of the enzyme (Fig. 1A). Four mutations (G848S, c.2542g→a; T851A, c.2551a→g; R852C, c.2554c→t; R853Q, c.2558g→a) reside in the thumb subdomain and two (Q879H, c.2637g→t and T885S, c.2653a→t) are located in the palm subdomain. These mutations are associated with Alpers, PEO, mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS), ataxia-neuropathy syndrome, Leigh syndrome, and myocerebrohepatopathy (
POLG mutationDiseaseGeneticsReference
G848SAlpers syndromeIn trans with A467T, Q497H, T251I-P587L, or W748S-E1143G in Alpers syndrome15, 35, 4350
Leigh syndromeIn trans with R232H in Leigh syndrome49
MELASIn trans with R627Q in MELAS38
PEO with ataxia-neuropathyIn trans with G746S and E1143G in PEO with ataxia50
PEOIn trans with T251I and P587L in PEO51, 52
T851AAlpers syndromeIn trans with R1047W48, 53
In trans with H277C
R852CAlpers syndromeIn trans with A467T14, 48, 50
In cis with G11D and in trans with W748S-E1143G or A467T
Ataxia-neuropathyIn trans with G11D-R627Q15
R853QMyocerebrohepatopathyIn trans with T251I-P587L15
Q879HAlpers syndrome with valproate-induced hepatic failureIn cis with E1143G and in trans with A467T-T885S35, 54
T885SAlpers syndrome with valproate-induced hepatic failureIn cis with A467T and in trans with Q879H-E1143G35, 54
Open in a separate windowOpen in a separate windowFIGURE 1.POLG mutations characterized in this study. A, the location of the six mutations characterized is shown in red in the primary sequence of pol γ. Four mutations, the G848S, T851A, R852C, and R853Q, are located in the thumb domain, whereas two mutations, the Q879H and T885S, are in the palm domain of the polymerase region. B, sequence alignment of pol γ from yeast to humans. The amino acids characterized in this study are shown in red. Yellow-highlighted amino acids are highly conserved, and blue-highlighted amino acids are moderately conserved.  相似文献   

10.
Plasmid pAMS1-Encoded,Bacteriocin-Related “Siblicide” in Enterococcus faecalis     
Christine M. Sedgley  Don B. Clewell  Susan E. Flannagan 《Journal of bacteriology》2009,191(9):3183-3188
  相似文献   

11.
The Pre-mRNA Splicing Machinery of Trypanosomes: Complex or Simplified?     
Arthur Günzl 《Eukaryotic cell》2010,9(8):1159-1170
  相似文献   

12.
Single-cell-type Proteomics: Toward a Holistic Understanding of Plant Function     
Shaojun Dai  Sixue Chen 《Molecular & cellular proteomics : MCP》2012,11(12):1622-1630
  相似文献   

13.
Borna Disease Virus Nucleoprotein (p40) Is a Major Target for CD8+-T-Cell-Mediated Immune Response          下载免费PDF全文
Oliver Planz  Lothar Stitz 《Journal of virology》1999,73(2):1715-1718
  相似文献   

14.
Immunological Mechanisms Mediating Hantavirus Persistence in Rodent Reservoirs     
Judith D. Easterbrook  Sabra L. Klein 《PLoS pathogens》2008,4(11)
Hantaviruses, similar to several emerging zoonotic viruses, persistently infect their natural reservoir hosts, without causing overt signs of disease. Spillover to incidental human hosts results in morbidity and mortality mediated by excessive proinflammatory and cellular immune responses. The mechanisms mediating the persistence of hantaviruses and the absence of clinical symptoms in rodent reservoirs are only starting to be uncovered. Recent studies indicate that during hantavirus infection, proinflammatory and antiviral responses are reduced and regulatory responses are elevated at sites of increased virus replication in rodents. The recent discovery of structural and non-structural proteins that suppress type I interferon responses in humans suggests that immune responses in rodent hosts could be mediated directly by the virus. Alternatively, several host factors, including sex steroids, glucocorticoids, and genetic factors, are reported to alter host susceptibility and may contribute to persistence of hantaviruses in rodents. Humans and reservoir hosts differ in infection outcomes and in immune responses to hantavirus infection; thus, understanding the mechanisms mediating viral persistence and the absence of disease in rodents may provide insight into the prevention and treatment of disease in humans. Consideration of the coevolutionary mechanisms mediating hantaviral persistence and rodent host survival is providing insight into the mechanisms by which zoonotic viruses have remained in the environment for millions of years and continue to be transmitted to humans.Hantaviruses are negative sense, enveloped RNA viruses (family: Bunyaviridae) that are comprised of three RNA segments, designated small (S), medium (M), and large (L), which encode the viral nucleocapsid (N), envelope glycoproteins (GN and GC), and an RNA polymerase (Pol), respectively. More than 50 hantaviruses have been found worldwide [1]. Each hantavirus appears to have coevolved with a specific rodent or insectivore host as similar phylogenetic trees are produced from virus and host mitochondrial gene sequences [2]. Spillover to humans causes hemorrhagic fever with renal syndrome (HFRS) or hantavirus cardiopulmonary syndrome (HCPS), depending on the virus [3][5]. Although symptoms vary, a common feature of both HFRS and HCPS is increased permeability of the vasculature and mononuclear infiltration [4]. Pathogenesis of HRFS and HCPS in humans is hypothesized to be mediated by excessive proinflammatory and CD8+ T cell responses ().

Table 1

Summary of Immune Responses in Humans during Hantavirus Infection.
Categorical ResponseImmune MarkerEffect of InfectionVirus Speciesa In Vitro/In VivoTissue or Cell Typeb, Phase of Infectionc References
Innate RIG-IElevatedSNVIn vitroHUVEC, ≤24 h p.i. [79]
ReducedNY-1VIn vitroHUVEC, ≤24 h p.i. [37]
TLR3ElevatedSNVIn vitroHUVEC, ≤24 h p.i. [79]
IFN-βElevatedPUUV, PHV, ANDVIn vitroHSVEC, HMVEC-L, ≤24 h p.i. [36],[80]
ReducedTULV, PUUV NSsIn vitroCOS-7 and MRC5 cells, ≤24 h p.i. [32],[33]
IFN-αElevatedPUUV, HTNVIn vitroMФ, DCs, 4 days p.i. [30]
No changeHTNVIn vivoBlood, acute [81]
IRF-3, IRF-7ElevatedSNV, HTNV, PHV, ANDVIn vitroHMVEC-L, ≤24 h p.i. [33],[38]
MxAElevatedHTNV, NY-1V, PHV, PUUV, ANDV, SNV, TULVIn vitroMФ,HUVEC,HMVEC-L, 6 h–4 days p.i. [36], [39][41],[79]
MHC I and IIElevatedHTNVIn vitroDCs, 4 days p.i. [30]
CD11bElevatedPUUVIn vivoBlood, acute [82]
CD40, CD80, CD86ElevatedHTNVIn vitroDCs, 4 days p.i. [30],[83]
NK cellsElevatedPUUVIn vivoBAL, acute [84]
Proinflammatory/Adhesion IL-1βElevatedSNV, HTNVIn vivoBlood, lungs, acute [85],[86]
IL-6ElevatedSNV, PUUVIn vivoBlood, lungs, acute [85],[87],[88]
TNF-αElevatedPUUV, SNV, HTNVIn vivoBlood, lungs, kidney, acute [85],[86],[88],[89]
ElevatedHTNVIn vitroDCs, 4 days p.i. [30]
CCL5ElevatedSNV, HTNVIn vitroHMVEC-L, HUVEC, 12 h–4 days p.i. [38],[39],[90]
CXCL8ElevatedPUUVIn vivoBlood, acute [82]
ElevatedPUUVIn vivoMen, blood, acute [62]
ElevatedTULV, PHV, HTNVIn vitroHUVEC, MФ, 2–4 days p.i. [39],[91]
CXCL10ElevatedSNV, HTNV, PHVIn vitroHMVEC-L,HUVEC, 3–4 days p.i. [38],[39]
ElevatedPUUVIn vivoMen, blood, acute [62]
IL-2ElevatedSNV, HTNV, PUUVIn vivoBlood, lungs, acute [82],[86]
Nitric oxideElevatedPUUVIn vivoBlood, acute [92]
GM-CSFElevatedPUUVIn vivoWomen, blood, acute [62]
ICAM, VCAMElevatedPUUVIn vivoKidney, acute [87]
ElevatedHTNV, PHVIn vitroHUVEC, 3–4 days p.i. [30],[39]
E-selectinElevatedPUUVIn vivoBlood, acute [82]
CD8+ and CD4+ T cells IFN-γElevatedHTNV, SNVIn vivoBlood, CD4+,CD8+, lungs, acute [81],[86]
CD8+ElevatedDOBV, PUUV, HTNVIn vivoBlood, BAL, acute [52],[84],[93]
Virus-specific IFN-γ+CD8+ElevatedPUUV, SNVIn vivoPBMC, acute [45],[94]
Perforin, Granzyme BElevatedPUUVIn vivoBlood, acute [95]
CD4+CD25+ “activated”ElevatedDOBV, PUUVIn vivoPBMC, acute [89],[93]
IL-4ElevatedSNVIn vivoLungs, acute [86]
Regulatory “suppressor T cells”d ReducedHTNVIn vivoBlood, acute [52]
IL-10ElevatedPUUVIn vivoBlood, acute [86]
TGF-βElevatedPUUVIn vivoKidney, acute [89]
Humoral IgM, IgG, IgA, IgEElevatedAll hantavirusesIn vivoBlood [4]
Open in a separate windowaSNV, Sin Nombre virus; NY-1V, New York-1 virus; PUUV, Puumala virus; PHV, Prospect Hill virus; ANDV, Andes virus; TULV, Tula virus; HTNV, Hantaan virus; DOBV, Dobrava virus.bHUVEC, human umbilical vascular endothelial cells; HSVEC, human saphenous vein endothelial cells; HMVEC-L, human lung microvascular endothelial cells; COS-7, African green monkey kidney fibroblasts transformed with Simian virus 40; MRC5, human fetal lung fibroblasts; MФ, macrophages; DCs, dendritic cells; BAL, bronchoalveolar lavage, PBMC, human peripheral blood mononuclear cells.cAcute infection is during symptomatic disease in patients.dSuppressor T cells likely represent cells currently referred to as regulatory T cells.

Table 2

Summary of Immune Responses in Rodents during Hantavirus Infection.
Categorical ResponseImmune MarkerEffect of InfectionVirus Speciesa Host, Tissue or Cell Typeb Phase of Infectionc References
Innate TLR7ReducedSEOVMale Norway rats, lungsAcute, Persistent [19]
ElevatedSEOVFemale Norway rats, lungsAcute, Persistent [19]
RIG-IElevatedSEOVFemale Norway rats, lungsAcute, Persistent [19]
ElevatedSEOVNewborn rats, thalamusAcute [96]
TLR3ElevatedSEOVMale Norway rats, lungsAcute, Persistent [19]
IFN-βReducedSEOVMale Norway rats, lungsAcute, Persistent [19],[61]
ElevatedSEOVFemale Norway rat lungsAcute [19],[61]
Mx2ReducedSEOVMale Norway rats, lungsAcute, Persistent [19],[60]
ElevatedSEOVFemale Norway rats, lungsAcute, Persistent [19],[60]
ElevatedHTNV, SEOVMiced, fibroblasts transfected with Mx23–4 days p.i. [97]
JAK2ElevatedSEOVFemale Norway rats, lungsAcute [60]
MHC IIElevatedPUUVBank volesGenetic susceptibility [74]
Proinflammatory/Adhesion IL-1βReducedSEOVMale Norway rats, lungsPersistent [29]
IL-6ReducedSEOVMale and female Norway rats, lungsAcute, Persistent [29],[61]
ElevatedSEOVMale rats, spleenAcute [29]
TNF-αReducedHTNVNewborn miced, CD8+, spleenAcute [49],[50]
ReducedSEOVMale Norway rats, lungsAcute, Persistent [29],[42],[61]
ElevatedSEOVFemale Norway rats, lungsPersistent [61]
CX3CL1, CXCL10ReducedSEOVMale Norway rats, lungsAcute, Persistent [29]
ElevatedSEOVMale Norway rats, spleenAcute [29]
CCL2, CCL5ElevatedSEOVMale Norway rats, spleenAcute [29]
NOS2ReducedSEOVMale Norway rats, lungsAcute, Persistent [29],[61]
ElevatedSEOVMale Norway rats, spleenAcute [29]
ElevatedHTNVMouse MФd, in vitro6 h p.i. [98]
VCAM, VEGFElevatedSEOVMale Norway rats, spleenAcute [29]
CD8+ and CD4+ T cells CD8+ReducedHTNVNewborn miced, spleenPersistent [50]
ElevatedHTNVSCID miced, CD8+ transferred, spleenPersistence [49]
ElevatedSEOVFemale Norway rats, lungsPersistent [61]
IFN-γElevatedSEOVFemale Norway rats, lungsPersistent [61]
ElevatedSEOVMale Norway rats, spleenAcute [29]
ElevatedSEOVMale and female Norway rats, splenocytesAcute [20]
ElevatedSNVDeer mice, CD4+ T cellsAcute [48]
ElevatedHTNVNewborn miced, CD8+ T cells, spleenAcute [50]
ReducedHTNVNewborn miced, CD8+ T cells, spleenPersistent [99]
IFN-γRElevatedSEOVFemale Norway rats, lungsAcute, Persistent [60]
ReducedSEOVMale Norway rats, lungsPersistent [60]
T cellsElevatedSEOVNude ratsPersistence [47]
ElevatedHTNVNude miced Persistence [100]
IL-4ReducedSEOVMale Norway rats, lungsAcute, Persistent [61]
ElevatedSNVDeer mice, CD4+ T cellsAcute [48]
ElevatedSEOVMale and female Norway rats, splenocytesAcute [20]
Regulatory Regulatory T cellsElevatedSEOVMale Norway rats, lungsPersistent [42],[61]
FoxP3ElevatedSEOVMale Norway rats, lungsPersistent [29],[42],[61]
TGF-βElevatedSEOVMale Norway rats, lungsPersistent [29]
SNVDeer mice, CD4+ T cellsPersistent [48]
IL-10ReducedSEOVMale Norway rats, lungs and spleenAcute, Persistent [29]
ElevatedSNVDeer mice, CD4+ T cellsAcute [48]
Humoral IgGElevatedSNVDeer micePersistent [12],[57]
ElevatedSEOVNorway ratsPersistent [16],[17]
ElevatedHTNVField micePersistent [15]
ElevatedPUUVBank volesPersistent [14]
ElevatedBCCVCotton ratsPersistent [18],[58]
Open in a separate windowaSEOV, Seoul virus; HTNV, Hantaan virus, PUUV, Puumala virus; SNV, Sin Nombre virus; PUUV, Puumala virus; BCCV, Black Creek Canal virus.bMФ, macrophages.cAcute infection is <30 days p.i. and persistent infection is ≥30 days p.i.d Mus musculus, non-natural reservoir host for hantaviruses.In contrast to humans, hantaviruses persistently infect their reservoir hosts, presumably causing lifelong infections [6]. Hantaviruses are shed in saliva, urine, and feces, and transmission among rodents or from rodents to humans occurs by inhalation of aerosolized virus in excrement or by transmission of virus in saliva during wounding [7],[8]. Although widely disseminated throughout the rodent host, high amounts of hantaviral RNA and antigen are consistently identified in the lungs of their rodent hosts, suggesting that the lungs may be an important site for maintenance of hantaviruses during persistent infection [9][18]. Hantavirus infection in rodents is characterized by an acute phase of peak viremia, viral shedding, and virus replication in target tissues, followed by a persistent phase of reduced, cyclical virus replication despite the presence of high antibody titers (Figure 1) [12][16], [18][20]. The onset of persistent infection varies across hantavirus–rodent systems, but generally the acute phase occurs during the first 2–3 weeks of infection and virus persistence is established thereafter (Figure 1).Open in a separate windowFigure 1Kinetics of Hantavirus Infection in Rodents.Adapted from Lee et al. [15] and others [12][14],[16],[18],[20], the kinetics of relative hantaviral load in blood (red), saliva (green), and lung tissue (blue) and antibody responses (black) during the acute and persistent phases of infection are represented. The amount of genomic viral RNA, infectious virus titer, and/or relative amount of viral antigen have been incorporated as relative hantaviral load. The antibody response is integrated as the relative amount of anti-hantavirus IgG and/or neutralizing antibody titers.Hantavirus infection alone does not cause disease, as reservoir hosts and non-natural hosts (e.g., hamsters infected with Sin Nombre virus [SNV] or Choclo virus) may support replicating virus in the absence of overt disease [12],[14],[16],[18],[21],[22]. Our primary hypothesis is that certain immune responses that are mounted in humans during hantavirus infection are suppressed in rodent reservoirs to establish and maintain viral persistence, while preventing disease (相似文献   

15.
Protein Identification Using Top-Down Spectra     
Xiaowen Liu  Yakov Sirotkin  Yufeng Shen  Gordon Anderson  Yihsuan S. Tsai  Ying S. Ting  David R. Goodlett  Richard D. Smith  Vineet Bafna  Pavel A. Pevzner 《Molecular & cellular proteomics : MCP》2012,11(6)
In the last two years, because of advances in protein separation and mass spectrometry, top-down mass spectrometry moved from analyzing single proteins to analyzing complex samples and identifying hundreds and even thousands of proteins. However, computational tools for database search of top-down spectra against protein databases are still in their infancy. We describe MS-Align+, a fast algorithm for top-down protein identification based on spectral alignment that enables searches for unexpected post-translational modifications. We also propose a method for evaluating statistical significance of top-down protein identifications and further benchmark various software tools on two top-down data sets from Saccharomyces cerevisiae and Salmonella typhimurium. We demonstrate that MS-Align+ significantly increases the number of identified spectra as compared with MASCOT and OMSSA on both data sets. Although MS-Align+ and ProSightPC have similar performance on the Salmonella typhimurium data set, MS-Align+ outperforms ProSightPC on the (more complex) Saccharomyces cerevisiae data set.In the past two decades, proteomics was dominated by bottom-up mass spectrometry that analyzes digested peptides rather than intact proteins. Bottom-up approaches, although powerful, do have limitations in analyzing protein species, e.g. various proteolytic forms of the same protein or various protein isoforms resulting from alternative splicing. Top-down mass spectrometry focuses on analyzing intact proteins and large peptides (110) and has advantages in localizing multiple post-translational modifications (PTMs)1 in a coordinated fashion (e.g. combinatorial PTM code) and identifying multiple protein species (e.g. proteolytically processed protein species) (11). Until recently, most top-down studies were limited to single purified proteins (1215). Top-down studies of protein mixtures were restricted by difficulties in separating and fragmenting intact proteins and a shortage of robust computational tools.In the last two years, because of advances in protein separation and top-down instrumentation, top-down mass spectrometry moved from analyzing single proteins to analyzing complex samples containing hundreds and even thousands of proteins (1621). Because algorithms for interpreting top-down spectra are still in their infancy, many recent developments include computational innovations in protein identification.Because top-down spectra are complex, the first step in top-down spectral interpretation is usually spectral deconvolution, which converts a complex top-down spectrum to a list of monoisotopic masses (a deconvolved spectrum). Every protein (possibly with modifications) can be scored against a top-down deconvoluted spectrum, resulting in a Protein-Spectrum-Match (PrSM). The top-down protein identification problem is finding a protein in a database with the highest scoring PrSM for a top-down spectrum and further output the PrSM if it is statistically significant. There are several software tools for top-down protein identification (SoftwareIdentification of unexpected modificationsProteogenomics search against 6-frame translationSpeedEstimation of statistical significanceProSightPC+/−a+Fast/Slowb+PIITA+/−−Fast−UStag++Fast−MS-TopDown+−Slow−MS-Align+++Fast+Open in a separate windowa ProSightPC has various search modes that contribute to bridging the gap between blind and restrictive modes of MS/MS database search. It can identify truncated proteins by using biomarker search and identify unexpected modifications by using Δm mode and setting the error tolerance of precursor mass to a large value (e.g., 1999 Da). However, it is not designed for identifying truncated proteins with unexpected PTMs which are not represented in the “shotgun annotated” database.b In its most advances mode, ProSightPC can search the annotated top-down database that contains various protein species. However, ProSightPC searches in this mode become an order of magnitude slower.
  • ProSightPC—ProSightPC is the most commonly used tool for top-down protein identification (22, 23). ProSightPC searches spectra against a “shotgun annotated” protein database, which is generated by considering all expected PTMs. The “shotgun annotated” protein database is much larger than the original protein database. ProSightPC can identify some (but not all) proteins with unexpected PTMs using advanced search options, such as biomarker search and Δm mode, but it is not designed for identifying truncated proteins with unexpected PTMs that are not represented in the “shotgun annotated” database. ProSightPC is a fast tool that reports the statistical significance of PrSMs.
  • PIITA—Unlike ProSightPC, PIITA (19) is a precursor independent method that uses only fragment ions for protein identification. It is capable of identifying protein species with unexpected PTMs on N- or C-termini, but it cannot directly identify protein species with PTMs on both N- and C-termini. PIITA is a fast tool that provides FIT scores and Δ scores rather than statistical significance estimates.
  • USTag—Unique Sequence Tag (USTag) (17) generates long (6 amino acids or longer) peptide sequence tags to identify PrSMs. This approach, although fast, relies on long peptide sequence tags that may be difficult to obtain for some spectra. It also does not provide an estimate of the statistical significance of PrSMs.
  • MS-TopDown—MS-TopDown (24) is based on spectral alignment (25). MS-TopDown allows one to match top-down spectra to proteins with unexpected PTMs, i.e. without knowing which PTMs are present in the sample. However, MS-TopDown is rather slow when searching against large proteomes and does not provide the statistical significance of PrSMs, making it difficult to select good PrSMs.
  • In addition, MASCOT, SEQUEST, and OMSSA (16, 26, 27) have been used for top-down protein identification.
We describe MS-Align+, a fast software tool for top-down protein identification. MS-Align+ shares the spectral alignment approach with MS-TopDown, but greatly improves on speed, statistical analysis (providing E-values of PrSMs), and the number of identified PrSMs (e.g. by finding spectral alignments between spectra and truncated proteins). We benchmarked various tools for top-down protein identification on two data sets from Saccharomyces cerevisiae (SC) and Salmonella typhimurium (ST). We demonstrate that MS-Align+ significantly increase the number of identified spectra as compared with MASCOT and OMSSA on both data sets. Although MS-Align+ and ProSightPC have similar performance on the ST data set, MS-Align+ outperforms ProSightPC on the more complex SC data set.  相似文献   

16.
Loss of Par-1a/MARK3/C-TAK1 Kinase Leads to Reduced Adiposity,Resistance to Hepatic Steatosis,and Defective Gluconeogenesis     
Jochen K. Lennerz  Jonathan B. Hurov  Lynn S. White  Katherine T. Lewandowski  Julie L. Prior  G. James Planer  Robert W. Gereau  IV  David Piwnica-Worms  Robert E. Schmidt  Helen Piwnica-Worms 《Molecular and cellular biology》2010,30(21):5043-5056
Par-1 is an evolutionarily conserved protein kinase required for polarity in worms, flies, frogs, and mammals. The mammalian Par-1 family consists of four members. Knockout studies of mice implicate Par-1b/MARK2/EMK in regulating fertility, immune homeostasis, learning, and memory as well as adiposity, insulin hypersensitivity, and glucose metabolism. Here, we report phenotypes of mice null for a second family member (Par-1a/MARK3/C-TAK1) that exhibit increased energy expenditure, reduced adiposity with unaltered glucose handling, and normal insulin sensitivity. Knockout mice were protected against high-fat diet-induced obesity and displayed attenuated weight gain, complete resistance to hepatic steatosis, and improved glucose handling with decreased insulin secretion. Overnight starvation led to complete hepatic glycogen depletion, associated hypoketotic hypoglycemia, increased hepatocellular autophagy, and increased glycogen synthase levels in Par-1a−/− but not in control or Par-1b−/− mice. The intercrossing of Par-1a−/− with Par-1b−/− mice revealed that at least one of the four alleles is necessary for embryonic survival. The severity of phenotypes followed a rank order, whereby the loss of one Par-1b allele in Par-1a−/− mice conveyed milder phenotypes than the loss of one Par-1a allele in Par-1b−/− mice. Thus, although Par-1a and Par-1b can compensate for one another during embryogenesis, their individual disruption gives rise to distinct metabolic phenotypes in adult mice.Cellular polarity is a fundamental principle in biology (6, 36, 62). The prototypical protein kinase originally identified as a regulator of polarity was termed partitioning defective (Par-1) due to early embryonic defects in Caenorhabditis elegans (52). Subsequent studies revealed that Par-1 is required for cellular polarity in worms, flies, frogs, and mammals (4, 17, 58, 63, 65, 71, 89). An integral role for Par-1 kinases in multiple signaling pathways has also been established, and although not formally addressed, multifunctionality for individual Par-1 family members is implied in reviews of the list of recognized upstream regulators and downstream substrates (Table (Table1).1). Interestingly, for many Par-1 substrates the phosphorylated residues generate 14-3-3 binding sites (25, 28, 37, 50, 59, 61, 68, 69, 78, 95, 101, 103). 14-3-3 binding in turn modulates both nuclear/cytoplasmic as well as cytoplasmic/membrane shuttling of target proteins, thus allowing Par-1 activity to establish intracellular spatial organization (15, 101). The phosphorylation of Par-1 itself promotes 14-3-3 binding, thereby regulating its subcellular localization (37, 59, 101).

TABLE 1.

Multifunctionality of Par-1 polarity kinase pathwaysa
Regulator or substrateFunctionReference(s)
Regulators (upstream function)
    LKB1Wnt signaling, Peutz-Jeghers syndrome, insulin signal transduction, pattern formation2, 63, 93
    TAO1MEK3/p38 stress-responsive mitogen-activated protein kinase (MAPK) pathway46
    MARKKNerve growth factor signaling in neurite development and differentiation98
    aPKCCa2+/DAG-independent signal transduction, cell polarity, glucose metabolism14, 37, 40, 45, 59, 75, 95
    nPKC/PKDDAG-dependent, Ca2+-independent signal transduction (GPCR)101
    PAR-3/PAR-6/aPKC(−); regulates Par-1, assembly of microtubules, axon-dendrite specification19
    GSK3β(−); tau phosphorylation, Alzheimer''s dementia, energy metabolism, body patterning54, 97
    Pim-1 oncogene(−); G2/M checkpoint, effector of cytokine signaling and Jak/STAT(3/5)5
    CaMKI(−); Ca2+-dependent signal transduction, neuronal differentiation99
Substrates (downstream function)
    Cdc25CRegulation of mitotic entry by activation of the cdc2-cyclin B complex25, 72, 78, 103
    Class II HDACControl of gene expression and master regulator of subcellular trafficking28, 50
    CRTC2/TORC2Gluconeogenesis regulator via LKB1/AMPK/TORC2 signaling, PPARγ1a coactivator49
    Dlg/PSD-95Synaptogenesis and neuromuscular junction, tumor suppressor (102)104
    DisheveledWnt signaling, translocation of Dsh from cytoplasmic vesicles to cortex73, 94
    KSR1Regulation of the Ras-MAPK pathway68, 69
    MAP2/4/TAUDynamic instability (67, 83) of microtubules, Alzheimer''s dementia (30)11, 31-33, 47, 70, 96
    Mib/NotchMind bomb (Mib degradation and repression of Notch signaling results in neurogenesis)57, 74, 81
    Par3/OSKAR/LglCytoplasmic protein segregation, cell polarity, and asymmetric cell division7, 10
    Pkp2Desmosome assembly and organization; nuclear shuttling68, 69
    PTPH1Linkage between Ser/Thr and Tyr phosphorylation-dependent signaling103
    Rab11-FIPRegulation of endocytosis (23), trafficking of E-cadherin (64)34
Open in a separate windowaLKB1 also is known as Par-4; MARKK also is known as Ste20-like; (−), inhibitory/negative regulation has been shown; GPCR, G protein-coupled receptors. MARKK is highly homologous to TAO-1 (thousand-and-one amino acid kinase) (46).The mammalian Par-1 family contains four members (Table (Table2).2). Physiological functions of the Par-1b kinase have been studied using targeted gene knockout approaches in mice (9, 44). Two independently derived mouse lines null for Par-1b have implicated this protein kinase in diverse physiological processes, including fertility (9), immune system homeostasis (44), learning and memory (86), the positioning of nuclei in pancreatic beta cells (35, 38), and growth and metabolism (43).

TABLE 2.

Terminology and localization of mammalian Par-1 family members
SynonymsaSubcellular localization
Par-1a, MARK3, C-TAK1, p78/KP78, 1600015G02Rik, A430080F22Rik, Emk2, ETK-1, KIAA4230, mKIAA1860, mKIAA4230, M80359Basolateralb/apicalc
Par-1b, EMK, MARK2, AU024026, mKIAA4207Basolateral
Par1c, MARK1Basolateral
Par1d, MARK4, MARKL1Not asymmetricd
Open in a separate windowaPar should not to be confused with protease-activated receptor 1 (PAR1 [29]); C-TAK1, Cdc twenty-five C-associated kinase 1; MARK, microtubule affinity regulating kinase; MARKL, MAP/microtubule affinity-regulating kinase-like 1.bBasolateral to a lesser degree than Par-1b (37).cHuman KP78 is asymmetrically localized to the apical surface of epithelial cells (76).dVariant that does not show asymmetric localization in epithelial cells when overexpressed (95).Beyond Par-1b, most information regarding the cell biological functions of the Par-1 kinases comes from studies of Par-1a. Specifically, Par-1a has been implicated in pancreatic (76) and hepatocarcinogenesis (51), as well as colorectal tumors (77), hippocampal function (100), CagA (Helicobacter pylori)-associated epithelial cell polarity disruption (82), and Peutz-Jeghers syndrome (48), although the latter association has been excluded recently (27). As a first step toward determining unique and redundant functions of Par-1 family members, mice disrupted for a second member of the family (Par-1a/MARK3/C-TAK1) were generated. We report that Par-1a−/− mice are viable and develop normally, and adult mice are hypermetabolic, have decreased white and brown adipose tissue mass, and unaltered glucose/insulin handling. However, when challenged by a high-fat diet (HFD), Par-1a−/− mice exhibit resistance to hepatic steatosis, resistance to glucose intolerance, and the delayed onset of obesity relative to that of control littermates. Strikingly, overnight starvation results in a complete depletion of glycogen and lipid stores along with an increase in autophagic vacuoles in the liver of Par-1a−/− but not Par-1b−/− mice. Correspondingly, Par-1a−/− mice develop hypoketotic hypoglycemia. These findings reveal unique metabolic functions of two Par-1 family members.  相似文献   

17.
Different Phenotypic Classes of Sinorhizobium meliloti Mutants Defective in Synthesis of K Antigen     
Gordon R. O. Campbell  Bradley L. Reuhs  Graham C. Walker 《Journal of bacteriology》1998,180(20):5432-5436
  相似文献   

18.
Zinc-Independent Folate Biosynthesis: Genetic,Biochemical, and Structural Investigations Reveal New Metal Dependence for GTP Cyclohydrolase IB     
Banumathi Sankaran  Shilah A. Bonnett  Kinjal Shah  Scott Gabriel  Robert Reddy  Paul Schimmel  Dmitry A. Rodionov  Valérie de Crécy-Lagard  John D. Helmann  Dirk Iwata-Reuyl  Manal A. Swairjo 《Journal of bacteriology》2009,191(22):6936-6949
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   

19.
The Chlamydomonas reinhardtii ODA3 Gene Encodes a Protein of the Outer Dynein Arm Docking Complex     
Anthony Koutoulis  Gregory J. Pazour  Curtis G. Wilkerson  Kazuo Inaba  Hong Sheng  Saeko Takada  George B. Witman 《The Journal of cell biology》1997,137(5):1069-1080
  相似文献   

20.
Bar-Coded Pyrosequencing of 16S rRNA Gene Amplicons Reveals Changes in Ileal Porcine Bacterial Communities Due to High Dietary Zinc Intake     
W. Vahjen  R. Pieper  J. Zentek 《Applied and environmental microbiology》2010,76(19):6689-6691
Feeding high levels of zinc oxide to piglets significantly increased the relative abundance of ileal Weissella spp., Leuconostoc spp., and Streptococcus spp., reduced the occurrence of Sarcina spp. and Neisseria spp., and led to numerical increases of all Gram-negative facultative anaerobic genera. High dietary zinc oxide intake has a major impact on the porcine ileal bacterial composition.Zinc oxide (ZnO) is used as a feed additive for diarrhea prophylaxis in piglets (23). However, the mode of action of ZnO is not fully understood. Besides its effects on the host (10, 30, 31), high dietary zinc levels may affect the diversity of intestinal microbial communities (2, 11, 20). The prevention of postweaning diarrhea in piglets due to high dietary ZnO intake may not be directly related to a reduction of pathogenic E. coli (8) but, rather, to the diversity of the coliform community (15). Studies on the impact of high ZnO levels on the porcine ileal bacterial community are scarce but nevertheless important, as bacterial diarrhea is initiated in the small intestine (9, 17). The small intestine is a very complex habitat with many different factors shaping the bacterial community. Studies on the ecophysiology (22) and maturation of the porcine ileal microbiota (13, 27) indicate a drastic impact directly after weaning and a gradual decline of modifications during the following 2 weeks. Thus, the time point for analysis chosen in this study (14 days postweaning) does reflect a more stable period of the ileal porcine microbiota. In this study, we used bar-coded pyrosequencing of 16S rRNA genes to gain further insight into the mode of action of pharmacological levels of ZnO in the gastrointestinal tract of young pigs.Total DNA was extracted from the ileal digesta of 40- to 42-day-old piglets using a commercial kit (Qiagen stool kit; Qiagen, Hilden, Germany) and PCR amplified with unique bar-coded primer sets targeting the V1-to-V3 and the V6-to-V8 hypervariable regions (see the supplemental material for detailed methods). The rationale behind this approach was derived from the fact that no single “universal” primer pair can completely cover a complex bacterial habitat (4, 24, 32, 33). Furthermore, these studies also show that in silico information on the coverage of selected primer sets diverges from empirical results, and hence, two hypervariable regions were chosen in this study to maximize the detection of phylogenetically diverse bacterial groups.Equimolar dilutions of all samples were combined into one master sample. Pyrosequencing was performed by Agowa (Berlin, Germany) on a Roche genome sequencer FLX system using a Titanium series PicoTiterPlate. The resulting data files were uploaded to the MG-RAST server (http://metagenomics.nmpdr.org/) (19) and processed with its SEED software tool using the RDP database (5) as the reference database. After automated sequence analysis, all sequences with less than five identical reads per sample were deleted in order to increase the confidence of sequence reads and reduce bias from possible sequencing errors (12, 16). Thus, 0.43% of all sequences were not considered (1,882 of 433,302 sequences). These sequences were assigned to a total of 238 genera, of which most only occurred in a few samples (see the supplemental material). Furthermore, all unclassified sequences were removed (8.7%; 41,467 of 474,769 sequences). Due to the use of the RDP reference database, the SEED software incorrectly assigned the majority of unclassified sequences as unclassified Deferribacterales (83%; 34,393 sequences), which were actually identified as 16S soybean or wheat chloroplasts by BLAST or as cyanobacterial chloroplasts by the RDP II seqmatch tool.The pyrosequencing results for the two primer combinations were merged by taking only sequences from the primer combination that yielded the higher number of reads for a specific sequence assignment in a sample. The remaining reads were used to calculate the relative contribution of assigned sequences to total sequence reads in a sample.The Firmicutes phylum dominated the small intestinal bacterial communities in both the control group and the group with high dietary ZnO intake, with 98.3% and 97.0% of total sequence reads, respectively. No significant influence of high dietary ZnO intake was found for the main phyla Proteobacteria (0.92% versus 1.84%), Actinobacteria (0.61% versus 0.75%), Bacteroidetes (0.15% versus 0.17%), and Fusobacteria (0.09% versus 0.12%).On the order level, a total of 20 bacterial orders were detected (data not shown). Lactobacillales dominated bacterial communities in the control and high-dietary-ZnO-intake groups, with 83.37% and 93.24% of total reads. Lactic acid bacteria are well known to dominate the bacterial community in the ileum of piglets (11, 22). No significant difference between the control group and the group with high dietary ZnO intake was observed on the order level, although high dietary ZnO intake led to a strong numerical decrease for Clostridiales (14.4 ± 24.0% [mean ± standard deviation] versus 2.8 ± 1.7%), as well as to numerical increases for Pseudomonadales (0.3 ± 0.3% versus 0.6 ± 0.6%) and Enterobacteriales (0.2 ± 0.2% versus 0.5 ± 0.6%).On the genus level, a total of 103 genera were detected. Table Table11 summarizes the main 31 genera which exceeded 0.05% of total reads (see the supplemental material for a complete list). Lactobacilli clearly dominated the bacterial communities in both trial groups, but they also were numerically lower due to high dietary ZnO intake.

TABLE 1.

Bacterial genera in the ileum of piglets fed diets supplemented with 200 or 3,000 ppm ZnO
GenusProportion (% ± SD) of ileal microbiota in groupa receiving:
200 ppm ZnO3,000 ppm ZnO
Lactobacillus59.3 ± 30.640.7 ± 19.1
Weissella11.6 ± 7.8 A24.1 ± 8.3 B
Sarcina11.4 ± 20.5 A0.84 ± 1.2 B
Leuconostoc4.7 ± 3.2 A9.4 ± 3.1 B
Streptococcus1.8 ± 1.6 A5.7 ± 5.1 B
Lactococcus1.6 ± 1.52.6 ± 3.1
Veillonella0.57 ± 0.630.34 ± 0.30
Gemella0.34 ± 0.67 A0.45 ± 0.25 B
Acinetobacter0.25 ± 0.210.44 ± 0.50
Clostridium0.25 ± 0.400.22 ± 0.21
Enterococcus0.19 ± 0.150.26 ± 0.24
Acidovorax0.14 ± 0.040.16 ± 0.19
Arcobacter0.14 ± 0.150.16 ± 0.17
Neisseria0.14b0.03 ± 0.01
Enterobacter0.13 ± 0.090.29 ± 0.34
Lachnospira0.12 ± 0.130.13 ± 0.03
Peptostreptococcus0.11 ± 0.100.07 ± 0.09
Chryseobacterium0.10 ± 0.070.15 ± 0.16
Actinomyces0.09 ± 0.040.15 ± 0.16
Anaerobacter0.07 ± 0.080.02 ± 0.01
Aerococcus0.07 ± 0.040.07 ± 0.04
Dorea0.07b0.05 ± 0.05
Fusobacterium0.06 ± 0.090.08 ± 0.11
Microbacterium0.06 ± 0.010.07 ± 0.04
Carnobacterium0.06 ± 0.020.08 ± 0.13
Granulicatella0.06 ± 0.020.09 ± 0.10
Staphylococcus0.06 ± 0.040.05 ± 0.02
Facklamia0.05 ± 0.060.03 ± 0.01
Comamonas0.05 ± 0.030.04 ± 0.02
Citrobacter0.05 ± 0.020.07 ± 0.08
Erysipelothrix0.05 ± 0.010.22 ± 0.40
Open in a separate windowan = 6 piglets per trial group. A,B, results are significantly different by Kruskal-Wallis test.bSingle sample.Significant changes due to high dietary ZnO intake were observed for other lactic acid bacteria, including Weissella spp., Leuconostoc spp., and Streptococcus spp. A significant and strong decrease was observed for Sarcina spp., which is a genus of acid-tolerant strictly anaerobic species found in the intestinal tract of piglets and other mammals (6, 28, 29). This genus thus appeared to be very sensitive to modifications induced by high dietary ZnO intake.An interesting result was observed for Gram-negative Proteobacteria, (i.e., enterobacteria and relatives). Although not statistically significant, virtually all detected proteobacteria increased numerically due to high dietary ZnO intake (Enterobacter spp., Microbacterium spp., Citrobacter spp., Neisseria spp., and Acinetobacter spp.). Apparently, enterobacteria gained colonization potential by high dietary ZnO intake. This is in good agreement with the results of studies by Hojberg et al. (11), Amezcua et al. (1), and Castillo et al. (3). Therefore, the frequently observed diarrhea-reducing effect of zinc oxide may not be directly related to a reduction of pathogenic E. coli strains. Considering a possible antagonistic activity of lactobacilli against enterobacteria (25), it can be speculated that a numerical decrease of dominant lactobacilli may lead to increased colonization with Gram-negative enterobacteria. On the other hand, specific plasmid-borne genes for resistance against heavy metals have been reported for both Gram-positive and Gram-negative bacteria present in the intestine (21, 26), and an increased resistance against Zn ions may exist for Gram-negative enterobacteria. Zinc oxide is an amphoteric molecule and shows a high solubility at acid pH. The low pH in the stomach of piglets (pH 3.5 to 4.5) transforms a considerable amount of insoluble ZnO into zinc ions (54 to 84% free Zn2+ at 150 ppm and 24 ppm ZnO, respectively) (7), and thus, high concentrations of toxic zinc ions exist in the stomach. The stomach of piglets harbors large numbers of lactic acid bacteria, especially lactobacilli. Zn ions may thus lead to a modification of the lactic acid bacterial community in the stomach, and the changes observed in the ileum could have been created in the stomach. A reduction of dominant lactobacilli may thus point to an increased adaptation potential of Gram-negative facultative anaerobes and a generally increased bacterial diversity.Additionally, the direct effects of dietary ZnO on intestinal tissues include altered expression of genes responsible for glutathione metabolism and apoptosis (30), enhanced gastric ghrelin secretion, which increases feed intake (31), and increased production of digestive enzymes (10). An analysis of the intestinal morphology was beyond the scope of this study, but although ZnO concentrations are markedly increased in intestinal tissue, the influence of ZnO on morphology is apparently not always observed (10, 14, 18). Consequently, any changes in epithelial cell turnover, feed intake, or digestive capacity may influence the composition of bacterial communities in the small intestine.In conclusion, this study has shown that high dietary zinc oxide has a major impact on ileal bacterial communities in piglets. Future studies on the impact of zinc oxide in pigs should include a detailed analysis of host responses in order to identify the cause for the observed modifications of intestinal bacterial communities.  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