Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(4): e0302251, 2024.
Article in English | MEDLINE | ID: mdl-38635746

ABSTRACT

Sterile alpha and TIR motif-containing 1 (SARM1) is a protein involved in programmed death of injured axons. Following axon injury or a drug-induced insult, the TIR domain of SARM1 degrades the essential molecule nicotinamide adenine dinucleotide (NAD+), leading to a form of axonal death called Wallerian degeneration. Degradation of NAD+ by SARM1 is essential for the Wallerian degeneration process, but accumulating evidence suggest that other activities of SARM1, beyond the mere degradation of NAD+, may be necessary for programmed axonal death. In this study we show that the TIR domains of both human and fruit fly SARM1 produce 1''-2' and 1''-3' glycocyclic ADP-ribose (gcADPR) molecules as minor products. As previously reported, we observed that SARM1 TIR domains mostly convert NAD+ to ADPR (for human SARM1) or cADPR (in the case of SARM1 from Drosophila melanogaster). However, we now show that human and Drosophila SARM1 additionally convert ~0.1-0.5% of NAD+ into gcADPR molecules. We find that SARM1 TIR domains produce gcADPR molecules both when purified in vitro and when expressed in bacterial cells. Given that gcADPR is a second messenger involved in programmed cell death in bacteria and likely in plants, we propose that gcADPR may play a role in SARM1-induced programmed axonal death in animals.


Subject(s)
NAD , Wallerian Degeneration , Animals , Humans , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , NAD/metabolism , Drosophila melanogaster/metabolism , Axons/metabolism , Bacteria/metabolism , Adenosine Diphosphate Ribose/metabolism , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism
2.
Nature ; 627(8003): 431-436, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383786

ABSTRACT

To survive bacteriophage (phage) infections, bacteria developed numerous anti-phage defence systems1-7. Some of them (for example, type III CRISPR-Cas, CBASS, Pycsar and Thoeris) consist of two modules: a sensor responsible for infection recognition and an effector that stops viral replication by destroying key cellular components8-12. In the Thoeris system, a Toll/interleukin-1 receptor (TIR)-domain protein, ThsB, acts as a sensor that synthesizes an isomer of cyclic ADP ribose, 1''-3' glycocyclic ADP ribose (gcADPR), which is bound in the Smf/DprA-LOG (SLOG) domain of the ThsA effector and activates the silent information regulator 2 (SIR2)-domain-mediated hydrolysis of a key cell metabolite, NAD+ (refs. 12-14). Although the structure of ThsA has been solved15, the ThsA activation mechanism remained incompletely understood. Here we show that 1''-3' gcADPR, synthesized in vitro by the dimeric ThsB' protein, binds to the ThsA SLOG domain, thereby activating ThsA by triggering helical filament assembly of ThsA tetramers. The cryogenic electron microscopy (cryo-EM) structure of activated ThsA revealed that filament assembly stabilizes the active conformation of the ThsA SIR2 domain, enabling rapid NAD+ depletion. Furthermore, we demonstrate that filament formation enables a switch-like response of ThsA to the 1''-3' gcADPR signal.


Subject(s)
Bacteria , Bacterial Proteins , Bacteriophages , Adenosine Diphosphate Ribose/analogs & derivatives , Adenosine Diphosphate Ribose/biosynthesis , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Bacteria/metabolism , Bacteria/virology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Bacteriophages/chemistry , Bacteriophages/metabolism , Bacteriophages/ultrastructure , Cryoelectron Microscopy , Hydrolysis , NAD/metabolism , Protein Domains , Protein Multimerization , Protein Stability
3.
Cell Host Microbe ; 30(11): 1556-1569.e5, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36302390

ABSTRACT

Bacterial anti-phage systems are frequently clustered in microbial genomes, forming defense islands. This property enabled the recent discovery of multiple defense systems based on their genomic co-localization with known systems, but the full arsenal of anti-phage mechanisms remains unknown. We report the discovery of 21 defense systems that protect bacteria from phages, based on computational genomic analyses and phage-infection experiments. We identified multiple systems with domains involved in eukaryotic antiviral immunity, including those homologous to the ubiquitin-like ISG15 protein, dynamin-like domains, and SEFIR domains, and show their participation in bacterial defenses. Additional systems include domains predicted to manipulate DNA and RNA molecules, alongside toxin-antitoxin systems shown here to function in anti-phage defense. These systems are widely distributed in microbial genomes, and in some bacteria, they form a considerable fraction of the immune arsenal. Our data substantially expand the inventory of defense systems utilized by bacteria to counteract phage infection.


Subject(s)
Bacteriophages , Bacteriophages/genetics , Bacteria/genetics , Genome, Microbial , Genomics , Immune System
4.
Nature ; 600(7887): 116-120, 2021 12.
Article in English | MEDLINE | ID: mdl-34853457

ABSTRACT

The Toll/interleukin-1 receptor (TIR) domain is a canonical component of animal and plant immune systems1,2. In plants, intracellular pathogen sensing by immune receptors triggers their TIR domains to generate a molecule that is a variant of cyclic ADP-ribose3,4. This molecule is hypothesized to mediate plant cell death through a pathway that has yet to be resolved5. TIR domains have also been shown to be involved in a bacterial anti-phage defence system called Thoeris6, but the mechanism of Thoeris defence remained unknown. Here we show that phage infection triggers Thoeris TIR-domain proteins to produce an isomer of cyclic ADP-ribose. This molecular signal activates a second protein, ThsA, which then depletes the cell of the essential molecule nicotinamide adenine dinucleotide (NAD) and leads to abortive infection and cell death. We also show that, similar to eukaryotic innate immune systems, bacterial TIR-domain proteins determine the immunological specificity to the invading pathogen. Our results describe an antiviral signalling pathway in bacteria, and suggest that the generation of intracellular signalling molecules is an ancient immunological function of TIR domains that is conserved in both plant and bacterial immunity.


Subject(s)
Bacillus/immunology , Bacillus/virology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacteriophages/immunology , Receptors, Interleukin-1/chemistry , Signal Transduction/immunology , Toll-Like Receptors/chemistry , Cyclic ADP-Ribose/analogs & derivatives , Cyclic ADP-Ribose/metabolism , Evolution, Molecular , Models, Molecular , NAD/metabolism , Protein Domains , Substrate Specificity/immunology
5.
Nature ; 589(7840): 120-124, 2021 01.
Article in English | MEDLINE | ID: mdl-32937646

ABSTRACT

Viperin is an interferon-induced cellular protein that is conserved in animals1. It has previously been shown to inhibit the replication of multiple viruses by producing the ribonucleotide 3'-deoxy-3',4'-didehydro (ddh)-cytidine triphosphate (ddhCTP), which acts as a chain terminator for viral RNA polymerase2. Here we show that eukaryotic viperin originated from a clade of bacterial and archaeal proteins that protect against phage infection. Prokaryotic viperins produce a set of modified ribonucleotides that include ddhCTP, ddh-guanosine triphosphate (ddhGTP) and ddh-uridine triphosphate (ddhUTP). We further show that prokaryotic viperins protect against T7 phage infection by inhibiting viral polymerase-dependent transcription, suggesting that it has an antiviral mechanism of action similar to that of animal viperin. Our results reveal a class of potential natural antiviral compounds produced by bacterial immune systems.


Subject(s)
Antiviral Agents/metabolism , Archaeal Proteins/metabolism , Bacterial Proteins/metabolism , Bacteriophage T7/immunology , Evolution, Molecular , Prokaryotic Cells/metabolism , Proteins/metabolism , Antiviral Agents/immunology , Archaeal Proteins/chemistry , Bacteria/immunology , Bacteria/metabolism , Bacteria/virology , Bacterial Proteins/chemistry , Bacteriophage T7/enzymology , Bacteriophage T7/physiology , DNA-Directed DNA Polymerase/metabolism , Humans , Oxidoreductases Acting on CH-CH Group Donors , Prokaryotic Cells/immunology , Prokaryotic Cells/virology , Proteins/chemistry , Proteins/genetics , Ribonucleotides/biosynthesis , Ribonucleotides/chemistry , Ribonucleotides/metabolism , Transcription, Genetic/drug effects
6.
Nature ; 586(7829): 429-433, 2020 10.
Article in English | MEDLINE | ID: mdl-32877915

ABSTRACT

Stimulator of interferon genes (STING) is a receptor in human cells that senses foreign cyclic dinucleotides that are released during bacterial infection and in endogenous cyclic GMP-AMP signalling during viral infection and anti-tumour immunity1-5. STING shares no structural homology with other known signalling proteins6-9, which has limited attempts at functional analysis and prevented explanation of the origin of cyclic dinucleotide signalling in mammalian innate immunity. Here we reveal functional STING homologues encoded within prokaryotic defence islands, as well as a conserved mechanism of signal activation. Crystal structures of bacterial STING define a minimal homodimeric scaffold that selectively responds to cyclic di-GMP synthesized by a neighbouring cGAS/DncV-like nucleotidyltransferase (CD-NTase) enzyme. Bacterial STING domains couple the recognition of cyclic dinucleotides with the formation of protein filaments to drive oligomerization of TIR effector domains and rapid NAD+ cleavage. We reconstruct the evolutionary events that followed the acquisition of STING into metazoan innate immunity, and determine the structure of a full-length TIR-STING fusion from the Pacific oyster Crassostrea gigas. Comparative structural analysis demonstrates how metazoan-specific additions to the core STING scaffold enabled a switch from direct effector function to regulation of antiviral transcription. Together, our results explain the mechanism of STING-dependent signalling and reveal the conservation of a functional cGAS-STING pathway in prokaryotic defence against bacteriophages.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Evolution, Molecular , Membrane Proteins , Second Messenger Systems , Animals , Bacteria/chemistry , Bacteria/virology , Bacterial Proteins/chemistry , Bacteriophages , Crystallography, X-Ray , Cyclic GMP/metabolism , Membrane Proteins/chemistry , Models, Molecular , NAD/metabolism , Nucleotidyltransferases/metabolism
7.
Cell ; 172(6): 1260-1270, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29522746

ABSTRACT

Bacteriophages, discovered about a century ago, have been pivotal as models for understanding the fundamental principles of molecular biology. While interest in phage biology declined after the phage "golden era," key recent developments, including advances in phage genomics, microscopy, and the discovery of the CRISPR-Cas anti-phage defense system, have sparked a renaissance in phage research in the past decade. This review highlights recently discovered unexpected complexities in phage biology, describes a new arsenal of phage genes that help them overcome bacterial defenses, and discusses advances toward documentation of the phage biodiversity on a global scale.


Subject(s)
Bacteriophages/genetics , Biology/trends , Genome, Viral/genetics , Genomics/trends , Molecular Biology/trends , Bacteria/genetics , Bacteria/virology , Bacteriophages/physiology , CRISPR-Cas Systems , Genetic Variation , Genomics/methods , Lysogeny/genetics , Models, Genetic
8.
Science ; 359(6379)2018 03 02.
Article in English | MEDLINE | ID: mdl-29371424

ABSTRACT

The arms race between bacteria and phages led to the development of sophisticated antiphage defense systems, including CRISPR-Cas and restriction-modification systems. Evidence suggests that known and unknown defense systems are located in "defense islands" in microbial genomes. Here, we comprehensively characterized the bacterial defensive arsenal by examining gene families that are clustered next to known defense genes in prokaryotic genomes. Candidate defense systems were systematically engineered and validated in model bacteria for their antiphage activities. We report nine previously unknown antiphage systems and one antiplasmid system that are widespread in microbes and strongly protect against foreign invaders. These include systems that adopted components of the bacterial flagella and condensin complexes. Our data also suggest a common, ancient ancestry of innate immunity components shared between animals, plants, and bacteria.


Subject(s)
Bacillus subtilis/immunology , Bacillus subtilis/virology , Bacteriophages/immunology , Bacteriophages/pathogenicity , Escherichia coli/immunology , Escherichia coli/virology , Genes, Bacterial/physiology , Bacillus subtilis/genetics , Escherichia coli/genetics , Genome, Bacterial , Multigene Family
9.
Nat Microbiol ; 3(1): 90-98, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29085076

ABSTRACT

The evolutionary pressure imposed by phage predation on bacteria and archaea has resulted in the development of effective anti-phage defence mechanisms, including restriction-modification and CRISPR-Cas systems. Here, we report on a new defence system, DISARM (defence island system associated with restriction-modification), which is widespread in bacteria and archaea. DISARM is composed of five genes, including a DNA methylase and four other genes annotated as a helicase domain, a phospholipase D (PLD) domain, a DUF1998 domain and a gene of unknown function. Engineering the Bacillus paralicheniformis 9945a DISARM system into Bacillus subtilis has rendered the engineered bacteria protected against phages from all three major families of tailed double-stranded DNA phages. Using a series of gene deletions, we show that four of the five genes are essential for DISARM-mediated defence, with the fifth (PLD) being redundant for defence against some of the phages. We further show that DISARM restricts incoming phage DNA and that the B. paralicheniformis DISARM methylase modifies host CCWGG motifs as a marker of self DNA akin to restriction-modification systems. Our results suggest that DISARM is a new type of multi-gene restriction-modification module, expanding the arsenal of defence systems known to be at the disposal of prokaryotes against their viruses.


Subject(s)
Bacillus subtilis/genetics , Bacillus subtilis/virology , Bacterial Proteins/metabolism , Bacteriophages/physiology , DNA Restriction-Modification Enzymes/genetics , Multigene Family/genetics , Bacterial Proteins/genetics , Bacteriophages/classification , Bacteriophages/growth & development , Cloning, Molecular , Computational Biology , Genome, Bacterial/genetics , Genomic Islands , Methyltransferases/genetics , Models, Genetic , Sequence Deletion , Virus Replication
10.
Cell ; 168(1-2): 13-15, 2017 01 12.
Article in English | MEDLINE | ID: mdl-28086086

ABSTRACT

Extracellular membrane vesicles from bacteria are now shown to transfer phage receptors from susceptible to resistant cells, thus making them transiently sensitive to phage infection (Tzipilevich et al.).


Subject(s)
Bacteriophages
11.
EMBO J ; 34(2): 169-83, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25452498

ABSTRACT

The perpetual arms race between bacteria and phage has resulted in the evolution of efficient resistance systems that protect bacteria from phage infection. Such systems, which include the CRISPR-Cas and restriction-modification systems, have proven to be invaluable in the biotechnology and dairy industries. Here, we report on a six-gene cassette in Bacillus cereus which, when integrated into the Bacillus subtilis genome, confers resistance to a broad range of phages, including both virulent and temperate ones. This cassette includes a putative Lon-like protease, an alkaline phosphatase domain protein, a putative RNA-binding protein, a DNA methylase, an ATPase-domain protein, and a protein of unknown function. We denote this novel defense system BREX (Bacteriophage Exclusion) and show that it allows phage adsorption but blocks phage DNA replication. Furthermore, our results suggest that methylation on non-palindromic TAGGAG motifs in the bacterial genome guides self/non-self discrimination and is essential for the defensive function of the BREX system. However, unlike restriction-modification systems, phage DNA does not appear to be cleaved or degraded by BREX, suggesting a novel mechanism of defense. Pan genomic analysis revealed that BREX and BREX-like systems, including the distantly related Pgl system described in Streptomyces coelicolor, are widely distributed in ~10% of all sequenced microbial genomes and can be divided into six coherent subtypes in which the gene composition and order is conserved. Finally, we detected a phage family that evades the BREX defense, implying that anti-BREX mechanisms may have evolved in some phages as part of their arms race with bacteria.


Subject(s)
Bacillus subtilis/virology , Bacteriophages/genetics , Bacteriophages/pathogenicity , DNA Methylation , DNA Modification Methylases/genetics , Genome, Microbial , Virulence/genetics , Bacillus subtilis/genetics , Bacteriophages/growth & development , Biological Evolution , DNA Modification Methylases/metabolism , DNA, Bacterial/genetics , DNA, Viral/genetics , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...