Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 724
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 172(4): 771-783.e18, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29358050

RESUMO

As in eukaryotes, bacterial genomes are not randomly folded. Bacterial genetic information is generally carried on a circular chromosome with a single origin of replication from which two replication forks proceed bidirectionally toward the opposite terminus region. Here, we investigate the higher-order architecture of the Escherichia coli genome, showing its partition into two structurally distinct entities by a complex and intertwined network of contacts: the replication terminus (ter) region and the rest of the chromosome. Outside of ter, the condensin MukBEF and the ubiquitous nucleoid-associated protein (NAP) HU promote DNA contacts in the megabase range. Within ter, the MatP protein prevents MukBEF activity, and contacts are restricted to ∼280 kb, creating a domain with distinct structural properties. We also show how other NAPs contribute to nucleoid organization, such as H-NS, which restricts short-range interactions. Combined, these results reveal the contributions of major evolutionarily conserved proteins in a bacterial chromosome organization.


Assuntos
Adenosina Trifosfatases , Cromossomos Bacterianos , Proteínas de Ligação a DNA , Escherichia coli K12 , Complexos Multiproteicos , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/ultraestrutura , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos Bacterianos/genética , Cromossomos Bacterianos/metabolismo , Cromossomos Bacterianos/ultraestrutura , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/ultraestrutura , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Escherichia coli K12/ultraestrutura , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Estrutura Quaternária de Proteína , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
2.
Mol Cell ; 81(9): 2000-2012.e3, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33705710

RESUMO

The ß-barrel assembly machine (BAM) integrates ß-barrel proteins into the outer membrane (OM) of Gram-negative bacteria. An essential BAM subunit (BamA) catalyzes integration by promoting the formation of a hybrid-barrel intermediate state between its own ß-barrel domain and that of its client proteins. Here we show that in addition to catalyzing the integration of ß-barrel proteins, BamA functions as a polypeptide export channel. In vivo structural mapping via intermolecular disulfide crosslinking showed that the extracellular "passenger" domain of a member of the "autotransporter" superfamily of virulence factors traverses the OM through the BamA ß-barrel lumen. Furthermore, we demonstrate that a highly conserved residue within autotransporter ß-barrels is required to position the passenger inside BamA to initiate translocation and that during translocation, the passenger stabilizes the hybrid-barrel state. Our results not only establish a new function for BamA but also unify the divergent functions of BamA and other "Omp85" superfamily transporters.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Transporte Biológico , Sequência Conservada , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Conformação Proteica , Dobramento de Proteína , Relação Estrutura-Atividade , Triptofano
3.
Mol Cell ; 81(9): 1988-1999.e4, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33705712

RESUMO

Bacterial small RNAs (sRNAs) regulate the expression of hundreds of transcripts via base pairing mediated by the Hfq chaperone protein. sRNAs and the mRNA sites they target are heterogeneous in sequence, length, and secondary structure. To understand how Hfq can flexibly match diverse sRNA and mRNA pairs, we developed a single-molecule Förster resonance energy transfer (smFRET) platform that visualizes the target search on timescales relevant in cells. Here we show that unfolding of target secondary structure on Hfq creates a kinetic energy barrier that determines whether target recognition succeeds or aborts before a stable anti-sense complex is achieved. Premature dissociation of the sRNA can be alleviated by strong RNA-Hfq interactions, explaining why sRNAs have different target recognition profiles. We propose that the diverse sequences and structures of Hfq substrates create an additional layer of information that tunes the efficiency and selectivity of non-coding RNA regulation in bacteria.


Assuntos
Escherichia coli K12/metabolismo , Regulação Bacteriana da Expressão Gênica , RNA Bacteriano/metabolismo , RNA Mensageiro/metabolismo , Pequeno RNA não Traduzido/metabolismo , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Transferência Ressonante de Energia de Fluorescência , Fator Proteico 1 do Hospedeiro/genética , Fator Proteico 1 do Hospedeiro/metabolismo , Cinética , Microscopia de Fluorescência , Conformação de Ácido Nucleico , Estabilidade Proteica , Estrutura Secundária de Proteína , Desdobramento de Proteína , Estabilidade de RNA , RNA Bacteriano/genética , RNA Mensageiro/genética , Pequeno RNA não Traduzido/genética , Análise de Célula Única , Relação Estrutura-Atividade
4.
Mol Cell ; 75(5): 957-966.e8, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31178354

RESUMO

Present in all realms of life, dinucleoside tetraphosphates (Np4Ns) are generally considered signaling molecules. However, only a single pathway for Np4N signaling has been delineated in eukaryotes, and no receptor that mediates the influence of Np4Ns has ever been identified in bacteria. Here we show that, under disulfide stress conditions that elevate cellular Np4N concentrations, diverse Escherichia coli mRNAs and sRNAs acquire a cognate Np4 cap. Purified E. coli RNA polymerase and lysyl-tRNA synthetase are both capable of adding such 5' caps. Cap removal by either of two pyrophosphatases, ApaH or RppH, triggers rapid RNA degradation in E. coli. ApaH, the predominant decapping enzyme, functions as both a sensor and an effector of disulfide stress, which inactivates it. These findings suggest that the physiological changes attributed to elevated Np4N concentrations in bacteria may result from widespread Np4 capping, leading to altered RNA stability and consequent changes in gene expression.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Estabilidade de RNA , RNA Bacteriano/metabolismo , Hidrolases Anidrido Ácido/genética , Fosfatos de Dinucleosídeos/genética , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , RNA Bacteriano/genética
5.
PLoS Genet ; 20(6): e1011335, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38913742

RESUMO

The outer membrane of gram-negative bacteria is a barrier to chemical and physical stress. Phospholipid transport between the inner and outer membranes has been an area of intense investigation and, in E. coli K-12, it has recently been shown to be mediated by YhdP, TamB, and YdbH, which are suggested to provide hydrophobic channels for phospholipid diffusion, with YhdP and TamB playing the major roles. However, YhdP and TamB have different phenotypes suggesting distinct functions. It remains unclear whether these functions are related to phospholipid metabolism. We investigated a synthetic cold sensitivity caused by deletion of fadR, a transcriptional regulator controlling fatty acid degradation and unsaturated fatty acid production, and yhdP, but not by ΔtamB ΔfadR or ΔydbH ΔfadR. Deletion of tamB recuses the ΔyhdP ΔfadR cold sensitivity further demonstrating the phenotype is related to functional diversification between these genes. The ΔyhdP ΔfadR strain shows a greater increase in cardiolipin upon transfer to the non-permissive temperature and genetically lowering cardiolipin levels can suppress cold sensitivity. These data also reveal a qualitative difference between cardiolipin synthases in E. coli, as deletion of clsA and clsC suppresses cold sensitivity but deletion of clsB does not. Moreover, increased fatty acid saturation is necessary for cold sensitivity and lowering this level genetically or through supplementation of oleic acid suppresses the cold sensitivity of the ΔyhdP ΔfadR strain. Together, our data clearly demonstrate that the diversification of function between YhdP and TamB is related to phospholipid metabolism. Although indirect regulatory effects are possible, we favor the parsimonious hypothesis that YhdP and TamB have differential phospholipid-substrate transport preferences. Thus, our data provide a potential mechanism for independent control of the phospholipid composition of the inner and outer membranes in response to changing conditions based on regulation of abundance or activity of YhdP and TamB.


Assuntos
Proteínas de Escherichia coli , Fosfolipídeos , Fosfolipídeos/metabolismo , Fosfolipídeos/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Transporte Biológico/genética , Cardiolipinas/metabolismo , Cardiolipinas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Temperatura Baixa , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Regulação Bacteriana da Expressão Gênica , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Ácidos Graxos/metabolismo , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas de Transferência de Fosfolipídeos/metabolismo
6.
Brief Bioinform ; 25(4)2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38975892

RESUMO

Understanding the biological functions and processes of genes, particularly those not yet characterized, is crucial for advancing molecular biology and identifying therapeutic targets. The hypothesis guiding this study is that the 3D proximity of genes correlates with their functional interactions and relevance in prokaryotes. We introduced 3D-GeneNet, an innovative software tool that utilizes high-throughput sequencing data from chromosome conformation capture techniques and integrates topological metrics to construct gene association networks. Through a series of comparative analyses focused on spatial versus linear distances, we explored various dimensions such as topological structure, functional enrichment levels, distribution patterns of linear distances among gene pairs, and the area under the receiver operating characteristic curve by utilizing model organism Escherichia coli K-12. Furthermore, 3D-GeneNet was shown to maintain good accuracy compared to multiple algorithms (neighbourhood, co-occurrence, coexpression, and fusion) across multiple bacteria, including E. coli, Brucella abortus, and Vibrio cholerae. In addition, the accuracy of 3D-GeneNet's prediction of long-distance gene interactions was identified by bacterial two-hybrid assays on E. coli K-12 MG1655, where 3D-GeneNet not only increased the accuracy of linear genomic distance tripled but also achieved 60% accuracy by running alone. Finally, it can be concluded that the applicability of 3D-GeneNet will extend to various bacterial forms, including Gram-negative, Gram-positive, single-, and multi-chromosomal bacteria through Hi-C sequencing and analysis. Such findings highlight the broad applicability and significant promise of this method in the realm of gene association network. 3D-GeneNet is freely accessible at https://github.com/gaoyuanccc/3D-GeneNet.


Assuntos
Redes Reguladoras de Genes , Software , Algoritmos , Biologia Computacional/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo
7.
Mol Cell ; 68(4): 686-697.e3, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149596

RESUMO

The evolutionary stability of synthetic genetic circuits is key to both the understanding and application of genetic control elements. One useful but challenging situation is a switch between life and death depending on environment. Here are presented "essentializer" and "cryodeath" circuits, which act as kill switches in Escherichia coli. The essentializer element induces cell death upon the loss of a bi-stable cI/Cro memory switch. Cryodeath makes use of a cold-inducible promoter to express a toxin. We employ rational design and a toxin/antitoxin titering approach to produce and screen a small library of potential constructs, in order to select for constructs that are evolutionarily stable. Both kill switches were shown to maintain functionality in vitro for at least 140 generations. Additionally, cryodeath was shown to control the growth environment of a population, with an escape frequency of less than 1 in 105 after 10 days of growth in the mammalian gut.


Assuntos
DNA Bacteriano , Evolução Molecular Direcionada , Escherichia coli K12 , Proteínas de Escherichia coli , Animais , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
8.
J Biol Chem ; 299(2): 102846, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36586436

RESUMO

Escherichia coli K-12 possesses two versions of Trk/Ktr/HKT-type potassium ion (K+) transporters, TrkG and TrkH. The current paradigm is that TrkG and TrkH have largely identical characteristics, and little information is available regarding their functional differences. Here, we show using cation uptake experiments with K+ transporter knockout mutants that TrkG and TrkH have distinct ion transport activities and physiological roles. K+-transport by TrkG required Na+, whereas TrkH-mediated K+ uptake was not affected by Na+. An aspartic acid located five residues away from a critical glycine in the third pore-forming region might be involved in regulation of Na+-dependent activation of TrkG. In addition, we found that TrkG but not TrkH had Na+ uptake activity. Our analysis of K+ transport mutants revealed that TrkH supported cell growth more than TrkG; however, TrkG was able to complement loss of TrkH-mediated K+ uptake in E. coli. Furthermore, we determined that transcription of trkG in E. coli was downregulated but not completely silenced by the xenogeneic silencing factor H-NS (histone-like nucleoid structuring protein or heat-stable nucleoid-structuring protein). Taken together, the transport function of TrkG is clearly distinct from that of TrkH, and TrkG seems to have been accepted by E. coli during evolution as a K+ uptake system that coexists with TrkH.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Escherichia coli K12 , Proteínas de Escherichia coli , Canais de Potássio , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transporte Biológico , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Potássio/metabolismo , Canais de Potássio/metabolismo
9.
Mol Cell ; 64(4): 826-834, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27871367

RESUMO

During type I-E CRISPR-Cas immunity, the Cascade surveillance complex utilizes CRISPR-derived RNAs to target complementary invasive DNA for destruction. When invader mutation blocks this interference activity, Cascade instead triggers rapid primed adaptation against the invader. The molecular basis for this dual Cascade activity is poorly understood. Here we show that the conformation of the Cse1 subunit controls Cascade activity. Using FRET, we find that Cse1 exists in a dynamic equilibrium between "open" and "closed" conformations, and the extent to which the open conformation is favored directly correlates with the attenuation of interference and relative increase in priming activity upon target mutation. Additionally, the Cse1 L1 motif modulates Cascade activity by stabilizing the closed conformation. L1 mutations promote the open conformation and switch immune response from interference to priming. Our results demonstrate that Cascade conformation controls the functional outcome of target recognition, enabling tunable CRISPR immune response to combat invader evolution.


Assuntos
Proteínas Associadas a CRISPR/imunologia , Sistemas CRISPR-Cas/imunologia , Escherichia coli K12/imunologia , Proteínas de Escherichia coli/imunologia , Regulação Bacteriana da Expressão Gênica , Plasmídeos/metabolismo , Sítios de Ligação , Proteínas Associadas a CRISPR/química , Proteínas Associadas a CRISPR/genética , Carbocianinas/química , DNA Helicases/química , DNA Helicases/genética , DNA Helicases/imunologia , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Mutação , Plasmídeos/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Coloração e Rotulagem/métodos
10.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39000123

RESUMO

Gemcitabine (2',2'-difluoro-2'-deoxycytidine), a widely used anticancer drug, is considered a gold standard in treating aggressive pancreatic cancers. Gamma-proteobacteria that colonize the pancreatic tumors contribute to chemoresistance against gemcitabine by metabolizing the drug to a less active and deaminated form. The gemcitabine transporters of these bacteria are unknown to date. Furthermore, there is no complete knowledge of the gemcitabine transporters in Escherichia coli or any other related proteobacteria. In this study, we investigate the complement of gemcitabine transporters in E. coli K-12 and two common chemoresistance-related bacteria (Klebsiella pneumoniae and Citrobacter freundii). We found that E. coli K-12 has two high-affinity gemcitabine transporters with distinct specificity properties, namely, NupC and NupG, whereas the gemcitabine transporters of C. freundii and K. pneumoniae include the NupC and NupG orthologs, functionally indistinguishable from their counterparts, and, in K. pneumoniae, one additional NupC variant, designated KpNupC2. All these bacterial transporters have a higher affinity for gemcitabine than their human counterparts. The highest affinity (KM 2.5-3.0 µΜ) is exhibited by NupGs of the bacteria-specific nucleoside-H+ symporter (NHS) family followed by NupCs (KM 10-13 µΜ) of the concentrative nucleoside transporter (CNT) family, 15-100 times higher than the affinities reported for the human gemcitabine transporter hENT1/SLC29A1, which is primarily associated with gemcitabine uptake in the pancreatic adenocarcinoma cells. Our results offer a basis for further insight into the role of specific bacteria in drug availability within tumors and for understanding the structure-function differences of bacterial and human drug transporters.


Assuntos
Desoxicitidina , Gencitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Humanos , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/genética , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Escherichia coli K12/efeitos dos fármacos , Gammaproteobacteria/genética , Gammaproteobacteria/metabolismo , Gammaproteobacteria/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Farmacorresistência Bacteriana/genética , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/metabolismo
11.
J Bacteriol ; 205(1): e0040322, 2023 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-36622229

RESUMO

The Gram-negative outer membrane (OM) is an asymmetric bilayer with phospholipids in its inner leaflet and mainly lipopolysaccharide (LPS) in its outer leaflet and is largely impermeable to many antibiotics. In Enterobacterales (e.g., Escherichia, Salmonella, Klebsiella, and Yersinia), the outer leaflet of the OM also contains phosphoglyceride-linked enterobacterial common antigen (ECAPG). This molecule consists of the conserved ECA carbohydrate linked to diacylglycerol-phosphate (DAG-P) through a phosphodiester bond. ECAPG contributes to the OM permeability barrier and modeling suggests that it may alter the packing of LPS molecules in the OM. Here, we investigate, in Escherichia coli K-12, the reaction synthesizing ECAPG from ECA precursor linked to an isoprenoid carrier to identify the lipid donor that provides the DAG-P moiety to ECAPG. Through overexpression of phospholipid biosynthesis genes, we observed alterations expected to increase levels of phosphatidylglycerol (PG) increased the synthesis of ECAPG, whereas alterations expected to decrease levels of PG decreased the synthesis of ECAPG. We discovered depletion of PG levels in strains that could synthesize ECAPG, but not other forms of ECA, causes additional growth defects, likely due to the buildup of ECA precursor on the isoprenoid carrier inhibiting peptidoglycan biosynthesis. Our results demonstrate ECAPG can be synthesized in the absence of the other major phospholipids (phosphatidylethanolamine and cardiolipin). Overall, these results conclusively demonstrate PG is the lipid donor for the synthesis of ECAPG and provide a key insight into the reaction producing ECAPG. In addition, these results provide an interesting parallel to lipoprotein acylation, which also uses PG as its DAG donor. IMPORTANCE The Gram-negative outer membrane is a permeability barrier preventing cellular entry of antibiotics. However, outer membrane biogenesis pathways are targets for small molecule development. Here, we investigate the synthesis of a form of enterobacterial common antigen (ECA), ECAPG, found in the outer membrane of Enterobacterales (e.g., Escherichia, Salmonella, and Klebsiella). ECAPG consists of the conserved ECA carbohydrate unit linked to diacylglycerol-phosphate-ECA is a phospholipid headgroup. The details of the reaction forming this molecule from polymerized ECA precursor are unknown. We determined the lipid donor providing the phospholipid moiety is phosphatidylglycerol. Understanding the synthesis of outer membrane constituents such as ECAPG provides the opportunity for development of molecules to increase outer membrane permeability, expanding the antibiotics available to treat Gram-negative infections.


Assuntos
Escherichia coli K12 , Lipopolissacarídeos , Lipopolissacarídeos/metabolismo , Diglicerídeos/metabolismo , Fosfolipídeos/metabolismo , Fosfatidilgliceróis , Escherichia coli K12/metabolismo , Escherichia coli/genética , Antígenos de Bactérias/metabolismo , Antibacterianos/metabolismo , Terpenos/metabolismo
12.
Mol Microbiol ; 118(4): 417-425, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36054785

RESUMO

Glucose is taken up by Escherichia coli through the phosphotransferase system (PTS) as the preferred carbon source. PTS mutants grow with glucose as a carbon source only in the presence of pyrroloquinoline quinone (PQQ), which is needed as a redox cofactor for the glucose dehydrogenase Gcd. The membrane-anchored Gcd enzyme oxidises glucose to gluconolactone in the periplasm. For this reaction to occur, external supply of PQQ is required as E. coli is unable to produce PQQ de novo. Growth experiments show that PqqU (previously YncD) is the TonB-ExbBD-dependent transporter for PQQ through the outer membrane. PQQ protected the cells from the PqqU-dependent phage IsaakIselin (Bas10) by competition for the receptor protein. As a high affinity uptake system, PqqU allows E. coli to activate Gcd even at surrounding PQQ concentrations of about 1 nmoL/L. At about 30-fold higher PQQ concentrations, the activation of Gcd gets PqqU independent. Due to its small size, Pqq may also pass the outer membrane through porins. The PQQ-dependent production of gluconate has been demonstrated in many plant growth-promoting bacteria that solubilise phosphate minerals in the soil by secreting this acid. Under phosphate limiting conditions also E. coli induces the glucose dehydrogenase and secretes gluconate, even in absence of PTS, that is, even when the bacterium is unable to grow on glucose without PQQ.


Assuntos
Escherichia coli K12 , Cofator PQQ , Carbono/metabolismo , Escherichia coli/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Gluconatos/metabolismo , Glucose/metabolismo , Glucose 1-Desidrogenase/genética , Glucose 1-Desidrogenase/metabolismo , Fosfatos/metabolismo , Fosfotransferases/metabolismo , Porinas/metabolismo , Cofator PQQ/metabolismo , Quinonas/metabolismo , Solo
13.
Microbiology (Reading) ; 169(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37040165

RESUMO

Pyridoxal 5'-phosphate (PLP) is the active form of vitamin B6 and a cofactor for many essential metabolic processes such as amino acid biosynthesis and one carbon metabolism. 4'-deoxypyridoxine (4dPN) is a long known B6 antimetabolite but its mechanism of action was not totally clear. By exploring different conditions in which PLP metabolism is affected in the model organism Escherichia coli K12, we showed that 4dPN cannot be used as a source of vitamin B6 as previously claimed and that it is toxic in several conditions where vitamin B6 homeostasis is affected, such as in a B6 auxotroph or in a mutant lacking the recently discovered PLP homeostasis gene, yggS. In addition, we found that 4dPN sensitivity is likely the result of multiple modes of toxicity, including inhibition of PLP-dependent enzyme activity by 4'-deoxypyridoxine phosphate (4dPNP) and inhibition of cumulative pyridoxine (PN) uptake. These toxicities are largely dependent on the phosphorylation of 4dPN by pyridoxal kinase (PdxK).


Assuntos
Escherichia coli K12 , Proteínas de Escherichia coli , Piridoxina/metabolismo , Vitamina B 6/metabolismo , Escherichia coli K12/metabolismo , Fosfato de Piridoxal/metabolismo , Homeostase , Vitaminas , Proteínas de Transporte , Proteínas de Escherichia coli/metabolismo
14.
Nucleic Acids Res ; 49(17): 9696-9710, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34428301

RESUMO

Bacteria regulate gene expression to adapt to changing environments through transcriptional regulatory networks (TRNs). Although extensively studied, no TRN is fully characterized since the identity and activity of all the transcriptional regulators comprising a TRN are not known. Here, we experimentally evaluate 40 uncharacterized proteins in Escherichia coli K-12 MG1655, which were computationally predicted to be transcription factors (TFs). First, we used a multiplexed chromatin immunoprecipitation method combined with lambda exonuclease digestion (multiplexed ChIP-exo) assay to characterize binding sites for these candidate TFs; 34 of them were found to be DNA-binding proteins. We then compared the relative location between binding sites and RNA polymerase (RNAP). We found 48% (283/588) overlap between the TFs and RNAP. Finally, we used these data to infer potential functions for 10 of the 34 TFs with validated DNA binding sites and consensus binding motifs. Taken together, this study: (i) significantly expands the number of confirmed TFs to 276, close to the estimated total of about 280 TFs; (ii) provides putative functions for the newly discovered TFs and (iii) confirms the functions of four representative TFs through mutant phenotypes.


Assuntos
Escherichia coli K12/genética , Proteínas de Escherichia coli/metabolismo , Fatores de Transcrição/metabolismo , Sítios de Ligação , Sequenciamento de Cromatina por Imunoprecipitação , Escherichia coli K12/metabolismo , Fatores de Transcrição/fisiologia
15.
Int J Mol Sci ; 24(5)2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36902204

RESUMO

The World Health Organization has cautioned that antimicrobial resistance (AMR) will be responsible for an estimated 10 million deaths annually by 2050. To facilitate prompt and accurate diagnosis and treatment of infectious disease, we investigated the potential of amino acids for use as indicators of bacterial growth activity by clarifying which amino acids are taken up by bacteria during the various growth phases. In addition, we examined the amino acid transport mechanisms that are employed by bacteria based on the accumulation of labeled amino acids, Na+ dependence, and inhibitory effects using a specific inhibitor of system A. We found that 3H-L-Ala accurately reflects the proliferative activity of Escherichia coli K-12 and pathogenic EC-14 in vitro. This accumulation in E. coli could be attributed to the amino acid transport systems being different from those found in human tumor cells. Moreover, biological distribution assessed in infection model mice with EC-14 using 3H-L-Ala showed that the ratio of 3H-L-Ala accumulated in infected muscle to that in control muscle was 1.20. By detecting the growth activity of bacteria in the body that occurs during the early stages of infection by nuclear imaging, such detection methods may result in expeditious diagnostic treatments for infectious diseases.


Assuntos
Infecções Bacterianas , Escherichia coli K12 , Animais , Camundongos , Humanos , Escherichia coli/metabolismo , Escherichia coli K12/metabolismo , Bactérias , Aminoácidos/metabolismo , Alanina/metabolismo
16.
J Bacteriol ; 204(1): e0042021, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34694904

RESUMO

Escherichia coli survives under acid stress conditions by the glutamic acid-dependent acid resistance (GAD) system, which enzymatically decreases intracellular protons. We found a linkage between GAD and flagellar systems in E. coli. The hdeD gene, one of the GAD cluster genes, encodes an uncharacterized membrane protein. A reporter assay showed that the hdeD promoter was induced in a GadE-dependent manner when grown in the M9 glycerol medium. Transcriptome analysis revealed that most of the transcripts were from genes involved in flagellum synthesis, and cell motility increased not only in the hdeD-deficient mutant but also in the gadE-deficient mutant. Defects in both the hdeD and gadE increased the intracellular level of FliA, an alternative sigma factor for flagellum synthesis, activated by the master regulator FlhDC. The promoter activity of the lrhA gene, which encodes repressor for the flhDC operon, was found to decrease in both the hdeD- and gadE-deficient mutants. Transmission electron microscopy showed that the number of flagellar filaments on the hdeD-, gadE-, and lrhA-deficient cells increased, and all three mutants showed higher motility than the parent strain. Thus, HdeD in the GAD system activates the lrhA promoter, resulting in a decrease in flagellar filaments in E. coli cells. We speculated that the synthesis of HdeD, stimulated in E. coli exposed to acid stress, could control the flagellum biosynthesis by sensing slight changes in pH at the cytoplasmic membrane. This could help in saving energy through termination of flagellum biosynthesis and improve bacterial survival efficiency within the animal digestive system. IMPORTANCE E. coli cells encounter various environments from the mouth down to the intestines within the host animals. The pH of gastric juice is lower than 2.0, and the bacterial must quickly respond and adapt to the following environmental changes before reaching the intestines. The quick response plays a role in cellular survival in the population, whereas adaptation may contribute to species survival. The GAD and flagellar systems are important for response to low pH in E. coli. Here, we identified the novel inner membrane regulator HdeD, encoding in the GAD cluster, to repress the synthesis of flagella. These insights provide a deeper understanding of how the bacteria enter the animal digestive system, survive, and form colonies in the intestines.


Assuntos
Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Flagelos/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Fatores de Transcrição/metabolismo , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Genoma Bacteriano , Proteínas de Membrana/genética , Mutação , Fatores de Transcrição/genética , Transcriptoma
17.
J Bacteriol ; 204(5): e0008122, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35442066

RESUMO

In Escherichia coli K-12, RecA binds to single-strand DNA (ssDNA) created by DNA damage to form a protein-DNA helical filament that serves to catalyze LexA autoproteolysis, which induces the SOS response. The SOS constitutive (SOSC) mutations recA730(E38K) and recA1202(Q184K) are both on the outside of the RecA filament, opposite to the face that binds DNA. recA730(E38K) is also able to suppress the UV sensitivity caused by recF mutations. Both SOSC expression and recF suppression are thought to be due to RecA730's ability to compete better for ssDNA coated with ssDNA-binding protein than the wild type. We tested whether other positively charged residues at these two positions would lead to SOSC expression and recF suppression. We found that 5/6 positively charged residues were SOSC and 4/5 of these were also recF suppressors. While other mutations at these two positions (and others) were recF suppressors, none were SOSC. Three recF suppressors could be made moderately SOSC by adding a recA operator mutation. We hypothesize two mechanisms for SOSC expression: the first suggests that the positive charge at positions 38 and 184 attract negatively charged molecules that block interactions that would destabilize the RecA-DNA filament, and the second involves more stable filaments caused by increases in mutant RecA concentration. IMPORTANCE In Escherichia coli K-12, SOS constitutive (SOSC) mutants of recA turn on the SOS response in the absence of DNA damage. Some SOSC mutants are also able to indirectly suppress the UV sensitivity of recF mutations. Two SOSC mutations, recA730(E38K) and recA1202(Q184K), define a surface on the RecA-DNA filament opposite the surface that binds DNA. Both introduce positive charges, and recA730 is a recF suppressor. We tested whether the positive charge at these two positions was required for SOSC expression and recF suppression. We found a high correlation between the positive charge, SOSC expression and recF suppression. We also found several other mutations (different types) that provide recF suppression but no SOSC expression.


Assuntos
Escherichia coli K12 , Proteínas de Escherichia coli , Proteínas de Bactérias/metabolismo , DNA de Cadeia Simples/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Mutação , Fenótipo , Recombinases Rec A/genética , Recombinases Rec A/metabolismo , Resposta SOS em Genética
18.
J Biol Chem ; 297(4): 101168, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34487760

RESUMO

LysO, a prototypical member of the LysO family, mediates export of L-lysine (Lys) and resistance to the toxic Lys antimetabolite, L-thialysine (Thl) in Escherichia coli. Here, we have addressed unknown aspects of LysO function pertaining to its membrane topology and the mechanism by which it mediates Lys/Thl export. Using substituted cysteine (Cys) accessibility, here we delineated the membrane topology of LysO. Our studies support a model in which both the N- and C-termini of LysO are present at the periplasmic face of the membrane with a transmembrane (TM) domain comprising eight TM segments (TMSs) between them. In addition, a feature of intramembrane solvent exposure in LysO is inferred with the identification of membrane-located solvent-exposed Cys residues. Isosteric substitutions of a pair of conserved acidic residues, one E233, located in the solvent-exposed TMS7 and the other D261, in a solvent-exposed intramembrane segment located between TMS7 and TMS8, abolished LysO function in vivo. Thl, but not Lys, elicited proton release in inside-out membrane vesicles, a process requiring the presence of both E233 and D261. We postulate that Thl may be exported in antiport with H+ and that Lys may be a low-affinity export substrate. Our findings are compatible with a physiological scenario wherein in vivo LysO exports the naturally occurring antimetabolite Thl with higher affinity over the essential cellular metabolite Lys, thus affording protection from Thl toxicity and limiting wasteful export of Lys.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/química , Membrana Celular/química , Escherichia coli K12/química , Proteínas de Escherichia coli/química , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Transporte Biológico Ativo , Membrana Celular/genética , Membrana Celular/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Domínios Proteicos , Relação Estrutura-Atividade
19.
J Biol Chem ; 297(6): 101419, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34801550

RESUMO

A profound understanding of the molecular interactions between receptors and ligands is important throughout diverse research, such as protein design, drug discovery, or neuroscience. What determines specificity and how do proteins discriminate against similar ligands? In this study, we analyzed factors that determine binding in two homologs belonging to the well-known superfamily of periplasmic binding proteins, PotF and PotD. Building on a previously designed construct, modes of polyamine binding were swapped. This change of specificity was approached by analyzing local differences in the binding pocket as well as overall conformational changes in the protein. Throughout the study, protein variants were generated and characterized structurally and thermodynamically, leading to a specificity swap and improvement in affinity. This dataset not only enriches our knowledge applicable to rational protein design but also our results can further lay groundwork for engineering of specific biosensors as well as help to explain the adaptability of pathogenic bacteria.


Assuntos
Escherichia coli K12/química , Proteínas de Escherichia coli/química , Proteínas Periplásmicas de Ligação/química , Receptores de Amina Biogênica/química , Espermidina/química , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Periplásmicas de Ligação/genética , Proteínas Periplásmicas de Ligação/metabolismo , Ligação Proteica , Receptores de Amina Biogênica/genética , Receptores de Amina Biogênica/metabolismo , Espermidina/metabolismo
20.
Mol Microbiol ; 116(4): 1140-1150, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34423481

RESUMO

In Escherichia coli, PriA, PriB, PriC, and DnaT proteins mediate three pathways for Replication Restart called PriA-PriB, PriA-PriC, and PriC. PriA is crucial for two of the three pathways. Its absence leads to slow growth, high basal levels of SOS expression, poorly partitioning nucleoids, UV sensitivity, and recombination deficiency. PriA has ATPase and helicase activities and interacts with PriB, DnaT, and single-stranded DNA-binding protein (SSB). priA300 (K230R) and priA301 (C479Y) have no phenotype as single mutants, but each phenocopy a priA-null mutant combined with ∆priB. This suggested that the two priA mutations affected the helicase activity that is required for the PriA-PriC pathway. To further test this, the biochemical activities of purified PriA300 and PriA301 were examined. As expected, PriA300 lacks ATPase and helicase activities but retains the ability to interact with PriB. PriA301, however, retains significant PriB-stimulated helicase activity even though PriA301 interactions with PriB and DNA are weakened. A PriA300,301 variant retains only the ability to interact with DNA in vitro and phenocopies the priA-null phenotype in vivo. This suggests that there are two biochemically and genetically distinct PriA-PriB pathways. One uses PriB-stimulated helicase activity to free a region of ssDNA and the other uses helicase-independent remodeling activity.


Assuntos
DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , DNA Helicases/metabolismo , Replicação do DNA , DNA Bacteriano , Proteínas de Ligação a DNA/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA