Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nature ; 592(7855): 611-615, 2021 04.
Article in English | MEDLINE | ID: mdl-33828299

ABSTRACT

Horizontal gene transfer and mutation are the two major drivers of microbial evolution that enable bacteria to adapt to fluctuating environmental stressors1. Clustered, regularly interspaced, short palindromic repeats (CRISPR) systems use RNA-guided nucleases to direct sequence-specific destruction of the genomes of mobile genetic elements that mediate horizontal gene transfer, such as conjugative plasmids2 and bacteriophages3, thus limiting the extent to which bacteria can evolve by this mechanism. A subset of CRISPR systems also exhibit non-specific degradation of DNA4,5; however, whether and how this feature affects the host has not yet been examined. Here we show that the non-specific DNase activity of the staphylococcal type III-A CRISPR-Cas system increases mutations in the host and accelerates the generation of antibiotic resistance in Staphylococcus aureus and Staphylococcus epidermidis. These mutations require the induction of the SOS response to DNA damage and display a distinct pattern. Our results demonstrate that by differentially affecting both mechanisms that generate genetic diversity, type III-A CRISPR systems can modulate the evolution of the bacterial host.


Subject(s)
CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , Mutagenesis , Mutation , Staphylococcus/genetics , Anti-Bacterial Agents/pharmacology , Bacteriophages/classification , Bacteriophages/physiology , CRISPR-Associated Proteins/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Deoxyribonucleases/metabolism , Drug Resistance, Microbial/drug effects , SOS Response, Genetics/drug effects , Staphylococcus/drug effects , Staphylococcus/immunology , Staphylococcus/virology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/virology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/virology , Time Factors
2.
Mol Cell ; 73(2): 264-277.e5, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30503773

ABSTRACT

Type ΙΙΙ CRISPR-Cas systems provide robust immunity against foreign RNA and DNA by sequence-specific RNase and target RNA-activated sequence-nonspecific DNase and RNase activities. We report on cryo-EM structures of Thermococcus onnurineus CsmcrRNA binary, CsmcrRNA-target RNA and CsmcrRNA-target RNAanti-tag ternary complexes in the 3.1 Å range. The topological features of the crRNA 5'-repeat tag explains the 5'-ruler mechanism for defining target cleavage sites, with accessibility of positions -2 to -5 within the 5'-repeat serving as sensors for avoidance of autoimmunity. The Csm3 thumb elements introduce periodic kinks in the crRNA-target RNA duplex, facilitating cleavage of the target RNA with 6-nt periodicity. Key Glu residues within a Csm1 loop segment of CsmcrRNA adopt a proposed autoinhibitory conformation suggestive of DNase activity regulation. These structural findings, complemented by mutational studies of key intermolecular contacts, provide insights into CsmcrRNA complex assembly, mechanisms underlying RNA targeting and site-specific periodic cleavage, regulation of DNase cleavage activity, and autoimmunity suppression.


Subject(s)
Autoimmunity , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Deoxyribonucleases/metabolism , RNA Stability , RNA, Bacterial/metabolism , RNA-Binding Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/ultrastructure , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/immunology , CRISPR-Associated Proteins/ultrastructure , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Cryoelectron Microscopy , Deoxyribonucleases/genetics , Deoxyribonucleases/immunology , Deoxyribonucleases/ultrastructure , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/immunology , Gene Expression Regulation, Bacterial , Models, Molecular , Multiprotein Complexes , Mutation , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/genetics , RNA, Bacterial/immunology , RNA, Bacterial/ultrastructure , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , RNA-Binding Proteins/ultrastructure , Structure-Activity Relationship , Thermococcus/enzymology , Thermococcus/genetics , Thermococcus/immunology
3.
Mol Cell ; 73(2): 278-290.e4, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30503774

ABSTRACT

Adaptive immune systems must accurately distinguish between self and non-self in order to defend against invading pathogens while avoiding autoimmunity. Type III CRISPR-Cas systems employ guide RNA to recognize complementary RNA targets, which triggers the degradation of both the invader's transcripts and their template DNA. These systems can broadly eliminate foreign targets with multiple mutations but circumvent damage to the host genome. To explore the molecular basis for these features, we use single-molecule fluorescence microscopy to study the interaction between a type III-A ribonucleoprotein complex and various RNA substrates. We find that Cas10-the DNase effector of the complex-displays rapid conformational fluctuations on foreign RNA targets, but is locked in a static configuration on self RNA. Target mutations differentially modulate Cas10 dynamics and tune the CRISPR interference activity in vivo. These findings highlight the central role of the internal dynamics of CRISPR-Cas complexes in self versus non-self discrimination and target specificity.


Subject(s)
Autoimmunity , Bacterial Proteins/immunology , CRISPR-Associated Proteins/immunology , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , RNA, Bacterial/immunology , Self Tolerance , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/immunology , Kinetics , Microscopy, Fluorescence , Mutation , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Signal Transduction , Single Molecule Imaging/methods , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/immunology , Structure-Activity Relationship
4.
Mol Cell ; 72(4): 608-609, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30444997

ABSTRACT

Athukoralage et al. (2018) identify a new class of nuclease that degrades cyclic oligoadenylate (cOA), a second messenger that activates non-specific RNA degradation by the type III CRISPR-Cas accessory RNase Csm6/Csx1. This discovery provides a mechanism for regulating the degradation of foreign transcripts during the type III CRISPR immune response.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Ribonucleases , Adenine Nucleotides , CRISPR-Cas Systems , Oligoribonucleotides
5.
PLoS Genet ; 14(6): e1007405, 2018 06.
Article in English | MEDLINE | ID: mdl-29856734

ABSTRACT

Biochemical pathways are often genetically encoded as simple transcription regulation networks, where one transcription factor regulates the expression of multiple genes in a pathway. The relative timing of each promoter's activation and shut-off within the network can impact physiology. In the DNA damage repair pathway (known as the SOS response) of Escherichia coli, approximately 40 genes are regulated by the LexA repressor. After a DNA damaging event, LexA degradation triggers SOS gene transcription, which is temporally separated into subsets of 'early', 'middle', and 'late' genes. Although this feature plays an important role in regulating the SOS response, both the range of this separation and its underlying mechanism are not experimentally defined. Here we show that, at low doses of DNA damage, the timing of promoter activities is not separated. Instead, timing differences only emerge at higher levels of DNA damage and increase as a function of DNA damage dose. To understand mechanism, we derived a series of synthetic SOS gene promoters which vary in LexA-operator binding kinetics, but are otherwise identical, and then studied their activity over a large dose-range of DNA damage. In distinction to established models based on rapid equilibrium assumptions, the data best fit a kinetic model of repressor occupancy at promoters, where the drop in cellular LexA levels associated with higher doses of DNA damage leads to non-equilibrium binding kinetics of LexA at operators. Operators with slow LexA binding kinetics achieve their minimal occupancy state at later times than operators with fast binding kinetics, resulting in a time separation of peak promoter activity between genes. These data provide insight into this remarkable feature of the SOS pathway by demonstrating how a single transcription factor can be employed to control the relative timing of each gene's transcription as a function of stimulus dose.


Subject(s)
Bacterial Proteins/metabolism , DNA Damage/genetics , Escherichia coli/genetics , Repressor Proteins/metabolism , SOS Response, Genetics/genetics , Serine Endopeptidases/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Regulatory Networks/genetics , Genes, Bacterial , Kinetics , Models, Genetic , Promoter Regions, Genetic , Repressor Proteins/genetics , Serine Endopeptidases/genetics , Time Factors
6.
Nucleic Acids Res ; 42(15): 9964-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25064858

ABSTRACT

Antibody maturation is a critical immune process governed by the enzyme activation-induced deaminase (AID), a member of the AID/APOBEC DNA deaminase family. AID/APOBEC deaminases preferentially target cytosine within distinct preferred sequence motifs in DNA, with specificity largely conferred by a small 9-11 residue protein loop that differs among family members. Here, we aimed to determine the key functional characteristics of this protein loop in AID and to thereby inform our understanding of the mode of DNA engagement. To this end, we developed a methodology (Sat-Sel-Seq) that couples saturation mutagenesis at each position across the targeting loop, with iterative functional selection and next-generation sequencing. This high-throughput mutational analysis revealed dominant characteristics for residues within the loop and additionally yielded enzymatic variants that enhance deaminase activity. To rationalize these functional requirements, we performed molecular dynamics simulations that suggest that AID and its hyperactive variants can engage DNA in multiple specific modes. These findings align with AID's competing requirements for specificity and flexibility to efficiently drive antibody maturation. Beyond insights into the AID-DNA interface, our Sat-Sel-Seq approach also serves to further expand the repertoire of techniques for deep positional scanning and may find general utility for high-throughput analysis of protein function.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , DNA/metabolism , Alanine/genetics , Cytidine Deaminase/genetics , High-Throughput Nucleotide Sequencing , Humans , Molecular Dynamics Simulation , Mutagenesis , Sequence Analysis, DNA , Structure-Activity Relationship
7.
Biochemistry ; 54(23): 3573-82, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26016604

ABSTRACT

Bacteria possess a remarkable ability to rapidly adapt and evolve in response to antibiotics. Acquired antibiotic resistance can arise by multiple mechanisms but commonly involves altering the target site of the drug, enzymatically inactivating the drug, or preventing the drug from accessing its target. These mechanisms involve new genetic changes in the pathogen leading to heritable resistance. This recognition underscores the importance of understanding how such genetic changes can arise. Here, we review recent advances in our understanding of the processes that contribute to the evolution of antibiotic resistance, with a particular focus on hypermutation mediated by the SOS pathway and horizontal gene transfer. We explore the molecular mechanisms involved in acquired resistance and discuss their viability as potential targets. We propose that additional studies into these adaptive mechanisms not only can provide insights into evolution but also can offer a strategy for potentiating our current antibiotic arsenal.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Evolution, Molecular , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Models, Genetic , SOS Response, Genetics/drug effects , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Bacterial/drug effects , Gene Transfer, Horizontal/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/growth & development , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/growth & development , Gram-Positive Bacteria/metabolism , Humans , Mutagenesis/drug effects
8.
Biochemistry ; 53(19): 3158-68, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24779472

ABSTRACT

Bacteria utilize the tightly regulated stress response (SOS) pathway to respond to a variety of genotoxic agents, including antimicrobials. Activation of the SOS response is regulated by a key repressor-protease, LexA, which undergoes autoproteolysis in the setting of stress, resulting in derepression of SOS genes. Remarkably, genetic inactivation of LexA's self-cleavage activity significantly decreases acquired antibiotic resistance in infection models and renders bacteria hypersensitive to traditional antibiotics, suggesting that a mechanistic study of LexA could help inform its viability as a novel target for combating acquired drug resistance. Despite structural insights into LexA, a detailed knowledge of the enzyme's protease specificity is lacking. Here, we employ saturation and positional scanning mutagenesis on LexA's internal cleavage region to analyze >140 mutants and generate a comprehensive specificity profile of LexA from the human pathogen Pseudomonas aeruginosa (LexAPa). We find that the LexAPa active site possesses a unique mode of substrate recognition. Positions P1-P3 prefer small hydrophobic residues that suggest specific contacts with the active site, while positions P5 and P1' show a preference for flexible glycine residues that may facilitate the conformational change that permits autoproteolysis. We further show that stabilizing the ß-turn within the cleavage region enhances LexA autoproteolytic activity. Finally, we identify permissive positions flanking the scissile bond (P4 and P2') that are tolerant to extensive mutagenesis. Our studies shed light on the active site architecture of the LexA autoprotease and provide insights that may inform the design of probes of the SOS pathway.


Subject(s)
Bacterial Proteins/chemistry , Mutagenesis , Proteolysis , Pseudomonas aeruginosa/enzymology , SOS Response, Genetics , Serine Endopeptidases/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Humans , Hydrophobic and Hydrophilic Interactions , Pseudomonas aeruginosa/genetics , Serine Endopeptidases/genetics , Substrate Specificity/physiology
9.
Front Microbiol ; 9: 2961, 2018.
Article in English | MEDLINE | ID: mdl-30619111

ABSTRACT

Many antibiotics, either directly or indirectly, cause DNA damage thereby activating the bacterial DNA damage (SOS) response. SOS activation results in expression of genes involved in DNA repair and mutagenesis, and the regulation of the SOS response relies on two key proteins, LexA and RecA. Genetic studies have indicated that inactivating the regulatory proteins of this response sensitizes bacteria to antibiotics and slows the appearance of resistance. However, advancement of small molecule inhibitors of the SOS response has lagged, despite their clear promise in addressing the threat of antibiotic resistance. Previously, we had addressed this deficit by performing a high throughput screen of ∼1.8 million compounds that monitored for inhibition of RecA-mediated auto-proteolysis of Escherichia coli LexA, the reaction that initiates the SOS response. In this report, the refinement of the 5-amino-1-(carbamoylmethyl)-1H-1,2,3-triazole-4-carboxamide scaffold identified in the screen is detailed. After development of a modular synthesis, a survey of key activity determinants led to the identification of an analog with improved potency and increased breadth, targeting auto-proteolysis of LexA from both E. coli and Pseudomonas aeruginosa. Comparison of the structure of this compound to those of others in the series suggests structural features that may be required for activity and cross-species breadth. In addition, the feasibility of small molecule modulation of the SOS response was demonstrated in vivo by the suppression of the appearance of resistance. These structure activity relationships thus represent an important step toward producing Drugs that Inhibit SOS Activation to Repress Mechanisms Enabling Resistance (DISARMERs).

10.
ACS Infect Dis ; 4(3): 349-359, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29275629

ABSTRACT

The RecA/LexA axis of the bacterial DNA damage (SOS) response is a promising, yet nontraditional, drug target. The SOS response is initiated upon genotoxic stress, when RecA, a DNA damage sensor, induces LexA, the SOS repressor, to undergo autoproteolysis, thereby derepressing downstream genes that can mediate DNA repair and accelerate mutagenesis. As genetic inhibition of the SOS response sensitizes bacteria to DNA damaging antibiotics and decreases acquired resistance, inhibitors of the RecA/LexA axis could potentiate our current antibiotic arsenal. Compounds targeting RecA, which has many mammalian homologues, have been reported; however, small-molecules targeting LexA autoproteolysis, a reaction unique to the prokaryotic SOS response, have remained elusive. Here, we describe the logistics and accomplishments of an academic-industry partnership formed to pursue inhibitors against the RecA/LexA axis. A novel fluorescence polarization assay reporting on RecA-induced self-cleavage of LexA enabled the screening of 1.8 million compounds. Follow-up studies on select leads show distinct activity patterns in orthogonal assays, including several with activity in cell-based assays reporting on SOS activation. Mechanistic assays demonstrate that we have identified first-in-class small molecules that specifically target the LexA autoproteolysis step in SOS activation. Our efforts establish a realistic example for navigating academic-industry partnerships in pursuit of anti-infective drugs and offer starting points for dedicated lead optimization of SOS inhibitors that could act as adjuvants for current antibiotics.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacterial Proteins/metabolism , Intersectoral Collaboration , Proteolysis , SOS Response, Genetics/drug effects , Serine Endopeptidases/metabolism , Biomedical Research/organization & administration , Drug Discovery/organization & administration , High-Throughput Screening Assays , Protease Inhibitors/isolation & purification , Protease Inhibitors/pharmacology
11.
ACS Synth Biol ; 6(11): 2067-2076, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28826208

ABSTRACT

The bacterial SOS stress-response pathway is a pro-mutagenic DNA repair system that mediates bacterial survival and adaptation to genotoxic stressors, including antibiotics and UV light. The SOS pathway is composed of a network of genes under the control of the transcriptional repressor, LexA. Activation of the pathway involves linked but distinct events: an initial DNA damage event leads to activation of RecA, which promotes autoproteolysis of LexA, abrogating its repressor function and leading to induction of the SOS gene network. These linked events can each independently contribute to DNA repair and mutagenesis, making it difficult to separate the contributions of the different events to observed phenotypes. We therefore devised a novel synthetic circuit to unlink these events and permit induction of the SOS gene network in the absence of DNA damage or RecA activation via orthogonal cleavage of LexA. Strains engineered with the synthetic SOS circuit demonstrate small-molecule inducible expression of SOS genes as well as the associated resistance to UV light. Exploiting our ability to activate SOS genes independently of upstream events, we further demonstrate that the majority of SOS-mediated mutagenesis on the chromosome does not readily occur with orthogonal pathway induction alone, but instead requires DNA damage. More generally, our approach provides an exemplar for using synthetic circuit design to separate an environmental stressor from its associated stress-response pathway.


Subject(s)
Bacterial Proteins , DNA Damage , Escherichia coli , Gene Regulatory Networks , Rec A Recombinases , SOS Response, Genetics , Serine Endopeptidases , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Rec A Recombinases/genetics , Rec A Recombinases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
12.
mSphere ; 1(4)2016.
Article in English | MEDLINE | ID: mdl-27536734

ABSTRACT

The bacterial SOS response is a DNA damage repair network that is strongly implicated in both survival and acquired drug resistance under antimicrobial stress. The two SOS regulators, LexA and RecA, have therefore emerged as potential targets for adjuvant therapies aimed at combating resistance, although many open questions remain. For example, it is not well understood whether SOS hyperactivation is a viable therapeutic approach or whether LexA or RecA is a better target. Furthermore, it is important to determine which antimicrobials could serve as the best treatment partners with SOS-targeting adjuvants. Here we derived Escherichia coli strains that have mutations in either lexA or recA genes in order to cover the full spectrum of possible SOS activity levels. We then systematically analyzed a wide range of antimicrobials by comparing the mean inhibitory concentrations (MICs) and induced mutation rates for each drug-strain combination. We first show that significant changes in MICs are largely confined to DNA-damaging antibiotics, with strains containing a constitutively repressed SOS response impacted to a greater extent than hyperactivated strains. Second, antibiotic-induced mutation rates were suppressed when SOS activity was reduced, and this trend was observed across a wider spectrum of antibiotics. Finally, perturbing either LexA or RecA proved to be equally viable strategies for targeting the SOS response. Our work provides support for multiple adjuvant strategies, while also suggesting that the combination of an SOS inhibitor with a DNA-damaging antibiotic could offer the best potential for lowering MICs and decreasing acquired drug resistance. IMPORTANCE Our antibiotic arsenal is becoming depleted, in part, because bacteria have the ability to rapidly adapt and acquire resistance to our best agents. The SOS pathway, a widely conserved DNA damage stress response in bacteria, is activated by many antibiotics and has been shown to play central role in promoting survival and the evolution of resistance under antibiotic stress. As a result, targeting the SOS response has been proposed as an adjuvant strategy to revitalize our current antibiotic arsenal. However, the optimal molecular targets and partner antibiotics for such an approach remain unclear. In this study, focusing on the two key regulators of the SOS response, LexA and RecA, we provide the first comprehensive assessment of how to target the SOS response in order to increase bacterial susceptibility and reduce mutagenesis under antibiotic treatment.

SELECTION OF CITATIONS
SEARCH DETAIL