Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 177(6): 1649-1661.e9, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31080069

ABSTRACT

Current machine learning techniques enable robust association of biological signals with measured phenotypes, but these approaches are incapable of identifying causal relationships. Here, we develop an integrated "white-box" biochemical screening, network modeling, and machine learning approach for revealing causal mechanisms and apply this approach to understanding antibiotic efficacy. We counter-screen diverse metabolites against bactericidal antibiotics in Escherichia coli and simulate their corresponding metabolic states using a genome-scale metabolic network model. Regression of the measured screening data on model simulations reveals that purine biosynthesis participates in antibiotic lethality, which we validate experimentally. We show that antibiotic-induced adenine limitation increases ATP demand, which elevates central carbon metabolism activity and oxygen consumption, enhancing the killing effects of antibiotics. This work demonstrates how prospective network modeling can couple with machine learning to identify complex causal mechanisms underlying drug efficacy.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Metabolic Networks and Pathways/drug effects , Adenine/metabolism , Computational Biology/methods , Drug Evaluation, Preclinical/methods , Escherichia coli/metabolism , Machine Learning , Metabolic Networks and Pathways/immunology , Models, Theoretical , Purines/metabolism
2.
Cell ; 178(1): 152-159.e11, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31178121

ABSTRACT

Intrinsic and acquired drug resistance and induction of secondary malignancies limit successful chemotherapy. Because mutagenic translesion synthesis (TLS) contributes to chemoresistance as well as treatment-induced mutations, targeting TLS is an attractive avenue for improving chemotherapeutics. However, development of small molecules with high specificity and in vivo efficacy for mutagenic TLS has been challenging. Here, we report the discovery of a small-molecule inhibitor, JH-RE-06, that disrupts mutagenic TLS by preventing recruitment of mutagenic POL ζ. Remarkably, JH-RE-06 targets a nearly featureless surface of REV1 that interacts with the REV7 subunit of POL ζ. Binding of JH-RE-06 induces REV1 dimerization, which blocks the REV1-REV7 interaction and POL ζ recruitment. JH-RE-06 inhibits mutagenic TLS and enhances cisplatin-induced toxicity in cultured human and mouse cell lines. Co-administration of JH-RE-06 with cisplatin suppresses the growth of xenograft human melanomas in mice, establishing a framework for developing TLS inhibitors as a novel class of chemotherapy adjuvants.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Mutagenesis/drug effects , Neoplasms/drug therapy , Quinolines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/adverse effects , Cisplatin/pharmacology , DNA Damage/drug effects , DNA-Directed DNA Polymerase , Female , Gene Knockdown Techniques , Humans , Mad2 Proteins/metabolism , Mice , Mice, Nude , Mice, Transgenic , Neoplasms/metabolism , Neoplasms/pathology , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Quinolines/chemistry , Quinolines/pharmacology , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Mol Cell ; 83(20): 3575-3577, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37863023
4.
J Biol Chem ; 299(2): 102859, 2023 02.
Article in English | MEDLINE | ID: mdl-36592930

ABSTRACT

Translesion synthesis (TLS) DNA polymerase Polζ is crucial for the bypass replication over sites of DNA damage. The Rev7 subunit of Polζ is a HORMA (Hop1, Rev7, Mad2) protein that facilitates recruitment of Polζ to the replication fork via interactions with the catalytic subunit Rev3 and the translesion synthesis scaffold protein Rev1. Human Rev7 (hRev7) interacts with two Rev7-binding motifs (RBMs) of hRev3 by a mechanism conserved among HORMA proteins whereby the safety-belt loop of hRev7 closes on the top of the ligand. The two copies of hRev7 tethered by the two hRev3-RBMs form a symmetric head-to-head dimer through the canonical HORMA dimerization interface. Recent cryo-EM structures reveal that Saccharomyces cerevisiae Polζ (scPolζ) also includes two copies of scRev7 bound to distinct regions of scRev3. Surprisingly, the HORMA dimerization interface is not conserved in scRev7, with the two scRev7 protomers forming an asymmetric head-to-tail dimer with a much smaller interface than the hRev7 dimer. Here, we validated the two adjacent RBM motifs in scRev3, which bind scRev7 with affinities that differ by two orders of magnitude and confirmed the 2:1 stoichiometry of the scRev7:Rev3 complex in solution. However, our biophysical studies reveal that scRev7 does not form dimers in solution either on its own accord or when tethered by the two RBMs in scRev3. These findings imply that the scRev7 dimer observed in the cryo-EM structures is induced by scRev7 interactions with other Polζ subunits and that Rev7 homodimerization via the HORMA interface is a mechanism that emerged later in evolution.


Subject(s)
DNA Replication , DNA-Directed DNA Polymerase , Humans , DNA Damage , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Mad2 Proteins/chemistry , Mad2 Proteins/metabolism , Nucleotidyltransferases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
5.
Microb Pathog ; 185: 106442, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37944675

ABSTRACT

Alphaproteobacteria include organisms living in close association with plants or animals. This interaction relies partly on orthologous two-component regulatory systems (TCS), with sensor and regulator proteins modulating the expression of conserved genes related to symbiosis/virulence. We assessed the ability of the exoS+Sm gene, encoding a sensor protein from the plant endosymbiont Sinorhizobium meliloti to substitute its orthologous bvrS in the related animal/human pathogen Brucella abortus. ExoS phosphorylated the B. abortus regulator BvrR in vitro and in cultured bacteria, showing conserved biological function. Production of ExoS in a B. abortus bvrS mutant reestablished replication in host cells and the capacity to infect mice. Bacterial outer membrane properties, the production of the type IV secretion system VirB, and its transcriptional regulators VjbR and BvrR were restored as compared to parental B. abortus. These results indicate that conserved traits of orthologous TCS from bacteria living in and sensing different environments are sufficient to achieve phenotypic plasticity and support bacterial survival. The knowledge of bacterial genetic networks regulating host interactions allows for an understanding of the subtle differences between symbiosis and parasitism. Rewiring these networks could provide new alternatives to control and prevent bacterial infection.


Subject(s)
Brucella abortus , Genes, Bacterial , Animals , Mice , Humans , Virulence/genetics , Histidine Kinase/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Mammals/genetics , Mammals/metabolism
6.
Proc Natl Acad Sci U S A ; 117(46): 28922-28924, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33144509

ABSTRACT

Cisplatin is a standard of care for lung cancer, yet platinum therapy rarely results in substantial tumor regression or a dramatic extension in patient survival. Here, we examined whether targeting Rev7 (also referred to as Mad2B, Mad2L2, and FANCV), a component of the translesion synthesis (TLS) machinery, could potentiate the action of cisplatin in non-small cell lung cancer (NSCLC) treatment. Rev7 loss led to an enhanced tumor cell sensitivity to cisplatin and dramatically improved chemotherapeutic response in a highly drug-resistant mouse model of NSCLC. While cisplatin monotherapy resulted in tumor cell apoptosis, Rev7 deletion promoted a cisplatin-induced senescence phenotype. Moreover, Rev7 deficiency promoted greater cisplatin sensitivity than that previously shown following targeting of other Pol ζ-proteins, suggesting that Pol ζ-dependent and -independent roles of Rev7 are relevant to cisplatin response. Thus, targeting Rev7 may represent a unique strategy for altering and enhancing chemotherapeutic response.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , Lung Neoplasms/drug therapy , Mad2 Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mad2 Proteins/metabolism , Mice , Mutagenesis , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 117(46): 28918-28921, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33168727

ABSTRACT

REV1/POLζ-dependent mutagenic translesion synthesis (TLS) promotes cell survival after DNA damage but is responsible for most of the resulting mutations. A novel inhibitor of this pathway, JH-RE-06, promotes cisplatin efficacy in cancer cells and mouse xenograft models, but the mechanism underlying this combinatorial effect is not known. We report that, unexpectedly, in two different mouse xenograft models and four human and mouse cell lines we examined in vitro cisplatin/JH-RE-06 treatment does not increase apoptosis. Rather, it increases hallmarks of senescence such as senescence-associated ß-galactosidase, increased p21 expression, micronuclei formation, reduced Lamin B1, and increased expression of the immune regulators IL6 and IL8 followed by cell death. Moreover, although p-γ-H2AX foci formation was elevated and ATR expression was low in single agent cisplatin-treated cells, the opposite was true in cells treated with cisplatin/JH-RE-06. These observations suggest that targeting REV1 with JH-RE-06 profoundly affects the nature of the persistent genomic damage after cisplatin treatment and also the resulting physiological responses. These data highlight the potential of REV1/POLζ inhibitors to alter the biological response to DNA-damaging chemotherapy and enhance the efficacy of chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Nitroquinolines/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Aging/drug effects , Aging/pathology , Aging/physiology , Animals , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/pharmacology , DNA/biosynthesis , DNA Damage/physiology , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Inhibitors/administration & dosage , Humans , Mad2 Proteins/metabolism , Mice , Mutagenesis , Neoplasms/enzymology , Neoplasms/pathology , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
8.
Int J Mol Sci ; 24(21)2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37958783

ABSTRACT

Rev7 is a regulatory protein with roles in translesion synthesis (TLS), double strand break (DSB) repair, replication fork protection, and cell cycle regulation. Rev7 forms a homodimer in vitro using its HORMA (Hop, Rev7, Mad2) domain; however, the functional importance of Rev7 dimerization has been incompletely understood. We analyzed the functional properties of cells expressing either wild-type mouse Rev7 or Rev7K44A/R124A/A135D, a mutant that cannot dimerize. The expression of wild-type Rev7, but not the mutant, rescued the sensitivity of Rev7-/- cells to X-rays and several alkylating agents and reversed the olaparib resistance phenotype of Rev7-/- cells. Using a novel fluorescent host-cell reactivation assay, we found that Rev7K44A/R124A/A135D is unable to promote gap-filling TLS opposite an abasic site analog. The Rev7 dimerization interface is also required for shieldin function, as both Rev7-/- cells and Rev7-/- cells expressing Rev7K44A/R124A/A135D exhibit decreased proficiency in rejoining some types of double strand breaks, as well as increased homologous recombination. Interestingly, Rev7K44A/R124A/A135D retains some function in cell cycle regulation, as it maintains an interaction with Ras-related nuclear protein (Ran) and partially rescues the formation of micronuclei. The mutant Rev7 also rescues the G2/M accumulation observed in Rev7-/- cells but does not affect progression through mitosis following nocodazole release. We conclude that while Rev7 dimerization is required for its roles in TLS, DSB repair, and regulation of the anaphase promoting complex, dimerization is at least partially dispensable for promoting mitotic spindle assembly through its interaction with Ran.


Subject(s)
DNA Repair , DNA Replication , Animals , Mice , Anaphase-Promoting Complex-Cyclosome/metabolism , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Mitosis/genetics
9.
Nucleic Acids Res ; 48(1): 332-348, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31777930

ABSTRACT

Single-strand specific endoribonuclease YbeY has been shown to play an important role in the processing of the 3' end of the 16S rRNA in Escherichia coli. Lack of YbeY results in the accumulation of the 17S rRNA precursor. In contrast to a previous report, we show that Sinorhizobium meliloti YbeY exhibits endoribonuclease activity on single-stranded RNA substrate but not on the double-stranded substrate. This study also identifies the previously unknown metal ion involved in YbeY function to be Zn2+ and shows that the activity of YbeY is enhanced when the occupancy of zinc is increased. We have identified a pre-16S rRNA precursor that accumulates in the S. meliloti ΔybeY strain. We also show that ΔybeY mutant of Brucella abortus, a mammalian pathogen, also accumulates a similar pre-16S rRNA. The pre-16S species is longer in alpha-proteobacteria than in gamma-proteobacteria. We demonstrate that the YbeY from E. coli and S. meliloti can reciprocally complement the rRNA processing defect in a ΔybeY mutant of the other organism. These results establish YbeY as a zinc-dependent single-strand specific endoribonuclease that functions in 16S rRNA processing in both alpha- and gamma-proteobacteria.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Metalloproteins/genetics , RNA, Ribosomal, 16S/genetics , Sinorhizobium meliloti/genetics , Zinc/metabolism , Base Pairing , Cations, Divalent , Enzyme Assays , Escherichia coli/metabolism , Gene Expression , Genetic Complementation Test , Isoenzymes/deficiency , Isoenzymes/genetics , Metalloproteins/deficiency , Mutation , Nucleic Acid Conformation , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Ribosomal, 16S/metabolism , Sinorhizobium meliloti/metabolism
10.
PLoS Genet ; 14(4): e1007357, 2018 04.
Article in English | MEDLINE | ID: mdl-29672509

ABSTRACT

Bacterial genome evolution is characterized by gains, losses, and rearrangements of functional genetic segments. The extent to which large-scale genomic alterations influence genotype-phenotype relationships has not been investigated in a high-throughput manner. In the symbiotic soil bacterium Sinorhizobium meliloti, the genome is composed of a chromosome and two large extrachromosomal replicons (pSymA and pSymB, which together constitute 45% of the genome). Massively parallel transposon insertion sequencing (Tn-seq) was employed to evaluate the contributions of chromosomal genes to growth fitness in both the presence and absence of these extrachromosomal replicons. Ten percent of chromosomal genes from diverse functional categories are shown to genetically interact with pSymA and pSymB. These results demonstrate the pervasive robustness provided by the extrachromosomal replicons, which is further supported by constraint-based metabolic modeling. A comprehensive picture of core S. meliloti metabolism was generated through a Tn-seq-guided in silico metabolic network reconstruction, producing a core network encompassing 726 genes. This integrated approach facilitated functional assignments for previously uncharacterized genes, while also revealing that Tn-seq alone missed over a quarter of wild-type metabolism. This work highlights the many functional dependencies and epistatic relationships that may arise between bacterial replicons and across a genome, while also demonstrating how Tn-seq and metabolic modeling can be used together to yield insights not obtainable by either method alone.


Subject(s)
Genome, Bacterial , Replicon , Sinorhizobium meliloti/genetics , Computer Simulation , Conserved Sequence , DNA Transposable Elements , DNA, Bacterial/genetics , Ecosystem , Epistasis, Genetic , Evolution, Molecular , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Metabolic Networks and Pathways/genetics , Models, Genetic , Molecular Sequence Annotation , Mutation , Sequence Analysis, DNA , Sinorhizobium meliloti/growth & development , Sinorhizobium meliloti/metabolism , Symbiosis/genetics
11.
Proc Natl Acad Sci U S A ; 115(35): E8191-E8200, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30111544

ABSTRACT

The translesion synthesis (TLS) polymerases Polζ and Rev1 form a complex that enables replication of damaged DNA. The Rev7 subunit of Polζ, which is a multifaceted HORMA (Hop1, Rev7, Mad2) protein with roles in TLS, DNA repair, and cell-cycle control, facilitates assembly of this complex by binding Rev1 and the catalytic subunit of Polζ, Rev3. Rev7 interacts with Rev3 by a mechanism conserved among HORMA proteins, whereby an open-to-closed transition locks the ligand underneath the "safety belt" loop. Dimerization of HORMA proteins promotes binding and release of this ligand, as exemplified by the Rev7 homolog, Mad2. Here, we investigate the dimerization of Rev7 when bound to the two Rev7-binding motifs (RBMs) in Rev3 by combining in vitro analyses of Rev7 structure and interactions with a functional assay in a Rev7-/- cell line. We demonstrate that Rev7 uses the conventional HORMA dimerization interface both to form a homodimer when tethered by the two RBMs in Rev3 and to heterodimerize with other HORMA domains, Mad2 and p31comet Structurally, the Rev7 dimer can bind only one copy of Rev1, revealing an unexpected Rev1/Polζ architecture. In cells, mutation of the Rev7 dimer interface increases sensitivity to DNA damage. These results provide insights into the structure of the Rev1/Polζ TLS assembly and highlight the function of Rev7 homo- and heterodimerization.


Subject(s)
Mad2 Proteins , Nuclear Proteins , Nucleotidyltransferases , Protein Multimerization , Cell Line , DNA Damage , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Mad2 Proteins/chemistry , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Domains
14.
Proc Natl Acad Sci U S A ; 114(34): 9164-9169, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28794281

ABSTRACT

Downstream metabolic events can contribute to the lethality of drugs or agents that interact with a primary cellular target. In bacteria, the production of reactive oxygen species (ROS) has been associated with the lethal effects of a variety of stresses including bactericidal antibiotics, but the relative contribution of this oxidative component to cell death depends on a variety of factors. Experimental evidence has suggested that unresolvable DNA problems caused by incorporation of oxidized nucleotides into nascent DNA followed by incomplete base excision repair contribute to the ROS-dependent component of antibiotic lethality. Expression of the chimeric periplasmic-cytoplasmic MalE-LacZ72-47 protein is an historically important lethal stress originally identified during seminal genetic experiments that defined the SecY-dependent protein translocation system. Multiple, independent lines of evidence presented here indicate that the predominant mechanism for MalE-LacZ lethality shares attributes with the ROS-dependent component of antibiotic lethality. MalE-LacZ lethality requires molecular oxygen, and its expression induces ROS production. The increased susceptibility of mutants sensitive to oxidative stress to MalE-LacZ lethality indicates that ROS contribute causally to cell death rather than simply being produced by dying cells. Observations that support the proposed mechanism of cell death include MalE-LacZ expression being bacteriostatic rather than bactericidal in cells that overexpress MutT, a nucleotide sanitizer that hydrolyzes 8-oxo-dGTP to the monophosphate, or that lack MutM and MutY, DNA glycosylases that process base pairs involving 8-oxo-dGTP. Our studies suggest stress-induced physiological changes that favor this mode of ROS-dependent death.

15.
Proc Natl Acad Sci U S A ; 113(36): 10157-62, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27551097

ABSTRACT

Interactions of rhizobia with legumes establish the chronic intracellular infection that underlies symbiosis. Within nodules of inverted repeat-lacking clade (IRLC) legumes, rhizobia differentiate into nitrogen-fixing bacteroids. This terminal differentiation is driven by host nodule-specific cysteine-rich (NCR) peptides that orchestrate the adaptation of free-living bacteria into intracellular residents. Medicago truncatula encodes a family of >700 NCR peptides that have conserved cysteine motifs. NCR247 is a cationic peptide with four cysteines that can form two intramolecular disulfide bonds in the oxidized forms. This peptide affects Sinorhizobium meliloti transcription, translation, and cell division at low concentrations and is antimicrobial at higher concentrations. By preparing the three possible disulfide-cross-linked NCR247 regioisomers, the reduced peptide, and a variant lacking cysteines, we performed a systematic study of the effects of intramolecular disulfide cross-linking and cysteines on the activities of an NCR peptide. The relative activities of the five NCR247 variants differed strikingly among the various bioassays, suggesting that the NCR peptide-based language used by plants to control the development of their bacterial partners during symbiosis is even greater than previously recognized. These patterns indicate that certain NCR bioactivities require cysteines whereas others do not. The results also suggest that NCR247 may exert some of its effects within the cell envelope whereas other activities occur in the cytoplasm. BacA, a membrane protein that is critical for symbiosis, provides protection against all bactericidal forms of NCR247. Oxidative folding protects NCR247 from degradation by the symbiotically relevant metalloprotease HrrP (host range restriction peptidase), suggesting that disulfide bond formation may additionally stabilize NCR peptides during symbiosis.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Plant , Medicago truncatula/microbiology , Membrane Transport Proteins/genetics , Peptides/genetics , Plant Proteins/genetics , Sinorhizobium meliloti/drug effects , Symbiosis/genetics , Amino Acid Motifs , Bacterial Proteins/metabolism , Cysteine/chemistry , Disulfides/chemistry , Host Specificity , Medicago truncatula/genetics , Medicago truncatula/metabolism , Membrane Transport Proteins/metabolism , Nitrogen Fixation , Peptides/metabolism , Peptides/pharmacology , Plant Proteins/biosynthesis , Plant Proteins/pharmacology , Root Nodules, Plant/genetics , Root Nodules, Plant/metabolism , Root Nodules, Plant/microbiology , Signal Transduction , Sinorhizobium meliloti/genetics , Sinorhizobium meliloti/growth & development , Sinorhizobium meliloti/metabolism , Structure-Activity Relationship
16.
J Bacteriol ; 200(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29914987

ABSTRACT

YbeY is a highly conserved, multifunctional endoribonuclease that plays a significant role in ribosome biogenesis and has several additional roles. Here we show that overexpression of the conserved GTPase Era in Escherichia coli partially suppresses the growth defect of a ΔybeY strain while improving 16S rRNA processing and 70S ribosome assembly. This suppression requires both the ability of Era to hydrolyze GTP and the function of three exoribonucleases, RNase II, RNase R, and RNase PH, suggesting a model for the action of Era. Overexpression of Vibrio cholerae Era similarly partially suppresses the defects of an E. coli ΔybeY strain, indicating that this property of Era is conserved in bacteria other than E. coliIMPORTANCE This work provides insight into the critical, but still incompletely understood, mechanism of processing of the E. coli 16S rRNA 3' terminus. The highly conserved GTPase Era is known to bind to the precursor of the 16S rRNA near its 3' end. Both the endoribonuclease YbeY, which binds to Era, and four exoribonucleases have been implicated in this 3'-end processing. The results reported here offer additional insights into the role of Era in 16S rRNA 3'-end maturation and into the relationship between the action of the endoribonuclease YbeY and that of the four exoribonucleases. This study also hints at why YbeY is essential only in some bacteria and suggests that YbeY could be a target for a new class of antibiotics in these bacteria.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , GTP-Binding Proteins/metabolism , Metalloproteins/genetics , RNA, Ribosomal, 16S/genetics , RNA-Binding Proteins/metabolism , Ribosomes/metabolism , Endoribonucleases/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Exoribonucleases/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , Gene Expression Regulation, Bacterial , Guanosine Triphosphate/metabolism , Hydrolysis , RNA-Binding Proteins/genetics , Vibrio cholerae/genetics
17.
J Bacteriol ; 200(13)2018 07 01.
Article in English | MEDLINE | ID: mdl-29632097

ABSTRACT

Sinorhizobium meliloti enters into beneficial symbiotic interactions with Medicago species of legumes. Bacterial exopolysaccharides play critical signaling roles in infection thread initiation and growth during the early stages of root nodule formation. After endocytosis of S. meliloti by plant cells in the developing nodule, plant-derived nodule-specific cysteine-rich (NCR) peptides mediate terminal differentiation of the bacteria into nitrogen-fixing bacteroids. Previous transcriptional studies showed that the intensively studied cationic peptide NCR247 induces expression of the exo genes that encode the proteins required for succinoglycan biosynthesis. In addition, genetic studies have shown that some exo mutants exhibit increased sensitivity to the antimicrobial action of NCR247. Therefore, we investigated whether the symbiotically active S. meliloti exopolysaccharide succinoglycan can protect S. meliloti against the antimicrobial activity of NCR247. We discovered that high-molecular-weight forms of succinoglycan have the ability to protect S. meliloti from the antimicrobial action of the NCR247 peptide but low-molecular-weight forms of wild-type succinoglycan do not. The protective function of high-molecular-weight succinoglycan occurs via direct molecular interactions between anionic succinoglycan and the cationic NCR247 peptide, but this interaction is not chiral. Taken together, our observations suggest that S. meliloti exopolysaccharides not only may be critical during early stages of nodule invasion but also are upregulated at a late stage of symbiosis to protect bacteria against the bactericidal action of cationic NCR peptides. Our findings represent an important step forward in fully understanding the complete set of exopolysaccharide functions during legume symbiosis.IMPORTANCE Symbiotic interactions between rhizobia and legumes are economically important for global food production. The legume symbiosis also is a major part of the global nitrogen cycle and is an ideal model system to study host-microbe interactions. Signaling between legumes and rhizobia is essential to establish symbiosis, and understanding these signals is a major goal in the field. Exopolysaccharides are important in the symbiotic context because they are essential signaling molecules during early-stage symbiosis. In this study, we provide evidence suggesting that the Sinorhizobium meliloti exopolysaccharide succinoglycan also protects the bacteria against the antimicrobial action of essential late-stage symbiosis plant peptides.


Subject(s)
Medicago truncatula/microbiology , Polysaccharides, Bacterial/metabolism , Sinorhizobium meliloti/physiology , Symbiosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Medicago truncatula/physiology , Nitrogen Fixation , Plant Roots/microbiology , Plant Roots/physiology , Sinorhizobium meliloti/genetics
18.
J Bacteriol ; 200(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29632093

ABSTRACT

The YbeY endoribonuclease is one of the best-conserved proteins across the kingdoms of life. In the present study, we demonstrated that YbeY in Brucella abortus is linked to a variety of important activities, including proper cellular morphology, mRNA transcript levels, and virulence. Deletion of ybeY in B. abortus led to a small-colony phenotype when the bacteria were grown on agar medium, as well as to significant aberrations in the morphology of the bacterial cell as evidenced by electron microscopy. Additionally, compared to the parental strain, the ΔybeY strain was significantly attenuated in both macrophage and mouse models of infection. The ΔybeY strain also showed increased sensitivities to several in vitro-applied stressors, including bile acid, hydrogen peroxide, SDS, and paraquat. Transcriptomic analysis revealed that a multitude of mRNA transcripts are dysregulated in the ΔybeY strain, and many of the identified mRNAs encode proteins involved in metabolism, nutrient transport, transcriptional regulation, and flagellum synthesis. We subsequently constructed gene deletion strains of the most highly dysregulated systems, and several of the YbeY-linked gene deletion strains exhibited defects in the ability of the bacteria to survive and replicate in primary murine macrophages. Taken together, these data establish a clear role for YbeY in the biology and virulence of Brucella; moreover, this work further illuminates the highly varied roles of this widely conserved endoribonuclease in bacteria.IMPORTANCEBrucella spp. are highly efficient bacterial pathogens of animals and humans, causing significant morbidity and economic loss worldwide, and relapse of disease often occurs following antibiotic treatment of human brucellosis. As such, novel therapeutic strategies to combat Brucella infections are needed. Ribonucleases in the brucellae are understudied, and these enzymes represent elements that may be potential targets for future treatment approaches. The present work demonstrates the importance of the YbeY endoribonuclease for cellular morphology, efficient control of mRNA levels, and virulence in B. abortus Overall, the results of this study advance our understanding of the critical roles of YbeY in the pathogenesis of the intracellular brucellae and expand our understanding of this highly conserved RNase.


Subject(s)
Bacterial Proteins/metabolism , Brucella abortus/enzymology , Brucella abortus/pathogenicity , Brucellosis/microbiology , Endoribonucleases/metabolism , Animals , Bacterial Proteins/genetics , Brucella abortus/genetics , Brucella abortus/growth & development , Endoribonucleases/genetics , Female , Gene Expression Regulation, Bacterial , Humans , Macrophages/microbiology , Male , Mice , Mice, Inbred BALB C , Virulence
19.
Annu Rev Pharmacol Toxicol ; 55: 313-32, 2015.
Article in English | MEDLINE | ID: mdl-25251995

ABSTRACT

We face an impending crisis in our ability to treat infectious disease brought about by the emergence of antibiotic-resistant pathogens and a decline in the development of new antibiotics. Urgent action is needed. This review focuses on a less well-understood aspect of antibiotic action: the complex metabolic events that occur subsequent to the interaction of antibiotics with their molecular targets and play roles in antibiotic lethality. Independent lines of evidence from studies of the action of bactericidal antibiotics on diverse bacteria collectively suggest that the initial interactions of drugs with their targets cannot fully account for the antibiotic lethality and that these interactions elicit the production of reactive oxidants including reactive oxygen species that contribute to bacterial cell death. Recent challenges to this concept are considered in the context of the broader literature of this emerging area of research. Possible ways that this new knowledge might be exploited to improve antibiotic therapy are also considered.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Microbial Viability/drug effects , Oxidants/pharmacology , Oxidative Stress/drug effects , Animals , Bacteria/genetics , Bacteria/metabolism , Drug Resistance, Bacterial/genetics , Humans , Hydrogen Peroxide/metabolism , Hydrogen Sulfide/metabolism , Mutagenesis/drug effects , Nitric Oxide/metabolism , Nucleotides/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Systems Biology
20.
Mol Cell ; 37(3): 408-17, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20159559

ABSTRACT

Translesion synthesis is an essential cell survival strategy to promote replication after DNA damage. The accumulation of Y family polymerases (pol) iota and Rev1 at the stalled replication machinery is mediated by the ubiquitin-binding motifs (UBMs) of the polymerases and enhanced by PCNA monoubiquitination. We report the solution structures of the C-terminal UBM of human pol iota and its complex with ubiquitin. Distinct from other ubiquitin-binding domains, the UBM binds to the hydrophobic surface of ubiquitin centered at L8. Accordingly, mutation of L8A, but not I44A, of ubiquitin abolishes UBM binding. Human pol iota contains two functional UBMs, both contributing to replication foci formation. In contrast, only the second UBM of Saccharomyces cerevisiae Rev1 binds to ubiquitin and is essential for Rev1-dependent cell survival and mutagenesis. Point mutations disrupting the UBM-ubiquitin interaction also impair the accumulation of pol iota in replication foci and Rev1-mediated DNA damage tolerance in vivo.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , Nucleotidyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/chemistry , Amino Acid Sequence , DNA Damage , DNA-Directed DNA Polymerase/metabolism , Humans , Molecular Sequence Data , Nucleotidyltransferases/chemistry , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Sequence Alignment , Ubiquitin/metabolism , Ubiquitination , DNA Polymerase iota
SELECTION OF CITATIONS
SEARCH DETAIL