Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 786
Filtrar
1.
Nat Commun ; 15(1): 3954, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729958

RESUMEN

Defense-associated sirtuin 2 (DSR2) systems are widely distributed across prokaryotic genomes, providing robust protection against phage infection. DSR2 recognizes phage tail tube proteins and induces abortive infection by depleting intracellular NAD+, a process that is counteracted by another phage-encoded protein, DSR Anti Defense 1 (DSAD1). Here, we present cryo-EM structures of Bacillus subtilis DSR2 in its apo, Tube-bound, and DSAD1-bound states. DSR2 assembles into an elongated tetramer, with four NADase catalytic modules clustered in the center and the regulatory-sensing modules distributed at four distal corners. Interestingly, monomeric Tube protein, rather than its oligomeric states, docks at each corner of the DSR2 tetramer to form a 4:4 DSR2-Tube assembly, which is essential for DSR2 NADase activity. DSAD1 competes with Tube for binding to DSR2 by occupying an overlapping region, thereby inhibiting DSR2 immunity. Thus, our results provide important insights into the assembly, activation and inhibition of the DSR2 anti-phage defense system.


Asunto(s)
Bacillus subtilis , Proteínas Bacterianas , Bacteriófagos , Microscopía por Crioelectrón , Bacillus subtilis/inmunología , Bacillus subtilis/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Bacteriófagos/genética , Bacteriófagos/inmunología , Evasión Inmune , Sirtuinas/metabolismo , Sirtuinas/genética , Proteínas Virales/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/química , Proteínas Virales/genética , Unión Proteica , Modelos Moleculares , NAD/metabolismo
2.
Nat Microbiol ; 9(5): 1312-1324, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565896

RESUMEN

Dormant prophages protect lysogenic cells by expressing diverse immune systems, which must avoid targeting their cognate prophages upon activation. Here we report that multiple Staphylococcus aureus prophages encode Tha (tail-activated, HEPN (higher eukaryotes and prokaryotes nucleotide-binding) domain-containing anti-phage system), a defence system activated by structural tail proteins of incoming phages. We demonstrate the function of two Tha systems, Tha-1 and Tha-2, activated by distinct tail proteins. Interestingly, Tha systems can also block reproduction of the induced tha-positive prophages. To prevent autoimmunity after prophage induction, these systems are inhibited by the product of a small overlapping antisense gene previously believed to encode an excisionase. This genetic organization, conserved in S. aureus prophages, allows Tha systems to protect prophages and their bacterial hosts against phage predation and to be turned off during prophage induction, balancing immunity and autoimmunity. Our results show that the fine regulation of these processes is essential for the correct development of prophages' life cycle.


Asunto(s)
Profagos , Staphylococcus aureus , Profagos/genética , Staphylococcus aureus/virología , Staphylococcus aureus/inmunología , Autoinmunidad , Lisogenia , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/inmunología , Fagos de Staphylococcus/fisiología , Proteínas de la Cola de los Virus/genética , Proteínas de la Cola de los Virus/metabolismo , Bacteriófagos/genética , Bacteriófagos/inmunología , Bacteriófagos/fisiología
3.
Nature ; 629(8011): 467-473, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38471529

RESUMEN

Prokaryotes have evolved intricate innate immune systems against phage infection1-7. Gabija is a highly widespread prokaryotic defence system that consists of two components, GajA and GajB8. GajA functions as a DNA endonuclease that is inactive in the presence of ATP9. Here, to explore how the Gabija system is activated for anti-phage defence, we report its cryo-electron microscopy structures in five states, including apo GajA, GajA in complex with DNA, GajA bound by ATP, apo GajA-GajB, and GajA-GajB in complex with ATP and Mg2+. GajA is a rhombus-shaped tetramer with its ATPase domain clustered at the centre and the topoisomerase-primase (Toprim) domain located peripherally. ATP binding at the ATPase domain stabilizes the insertion region within the ATPase domain, keeping the Toprim domain in a closed state. Upon ATP depletion by phages, the Toprim domain opens to bind and cleave the DNA substrate. GajB, which docks on GajA, is activated by the cleaved DNA, ultimately leading to prokaryotic cell death. Our study presents a mechanistic landscape of Gabija activation.


Asunto(s)
Bacillus cereus , Proteínas Bacterianas , Bacteriófagos , Microscopía por Crioelectrón , Inmunidad Innata , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/ultraestructura , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Apoproteínas/química , Apoproteínas/inmunología , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/inmunología , ADN/metabolismo , ADN/química , División del ADN , Magnesio/química , Magnesio/metabolismo , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Viabilidad Microbiana , Bacillus cereus/química , Bacillus cereus/inmunología , Bacillus cereus/metabolismo , Bacillus cereus/ultraestructura , Estructura Cuaternaria de Proteína , ADN Primasa/química , ADN Primasa/metabolismo , ADN Primasa/ultraestructura , ADN-Topoisomerasas/química , ADN-Topoisomerasas/metabolismo , ADN-Topoisomerasas/ultraestructura
4.
Nature ; 625(7996): 797-804, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38200316

RESUMEN

Prokaryotic type III CRISPR-Cas systems provide immunity against viruses and plasmids using CRISPR-associated Rossman fold (CARF) protein effectors1-5. Recognition of transcripts of these invaders with sequences that are complementary to CRISPR RNA guides leads to the production of cyclic oligoadenylate second messengers, which bind CARF domains and trigger the activity of an effector domain6,7. Whereas most effectors degrade host and invader nucleic acids, some are predicted to contain transmembrane helices without an enzymatic function. Whether and how these CARF-transmembrane helix fusion proteins facilitate the type III CRISPR-Cas immune response remains unknown. Here we investigate the role of cyclic oligoadenylate-activated membrane protein 1 (Cam1) during type III CRISPR immunity. Structural and biochemical analyses reveal that the CARF domains of a Cam1 dimer bind cyclic tetra-adenylate second messengers. In vivo, Cam1 localizes to the membrane, is predicted to form a tetrameric transmembrane pore, and provides defence against viral infection through the induction of membrane depolarization and growth arrest. These results reveal that CRISPR immunity does not always operate through the degradation of nucleic acids, but is instead mediated via a wider range of cellular responses.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , Potenciales de la Membrana , Staphylococcus aureus , Bacteriófagos/inmunología , Bacteriófagos/metabolismo , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Nucleótidos Cíclicos/metabolismo , ARN Guía de Sistemas CRISPR-Cas , Sistemas de Mensajero Secundario , Staphylococcus aureus/citología , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Staphylococcus aureus/virología
5.
Nature ; 625(7994): 360-365, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37992757

RESUMEN

Bacteria encode hundreds of diverse defence systems that protect them from viral infection and inhibit phage propagation1-5. Gabija is one of the most prevalent anti-phage defence systems, occurring in more than 15% of all sequenced bacterial and archaeal genomes1,6,7, but the molecular basis of how Gabija defends cells from viral infection remains poorly understood. Here we use X-ray crystallography and cryo-electron microscopy (cryo-EM) to define how Gabija proteins assemble into a supramolecular complex of around 500 kDa that degrades phage DNA. Gabija protein A (GajA) is a DNA endonuclease that tetramerizes to form the core of the anti-phage defence complex. Two sets of Gabija protein B (GajB) dimers dock at opposite sides of the complex and create a 4:4 GajA-GajB assembly (hereafter, GajAB) that is essential for phage resistance in vivo. We show that a phage-encoded protein, Gabija anti-defence 1 (Gad1), directly binds to the Gabija GajAB complex and inactivates defence. A cryo-EM structure of the virally inhibited state shows that Gad1 forms an octameric web that encases the GajAB complex and inhibits DNA recognition and cleavage. Our results reveal the structural basis of assembly of the Gabija anti-phage defence complex and define a unique mechanism of viral immune evasion.


Asunto(s)
Bacterias , Proteínas Bacterianas , Bacteriófagos , Evasión Inmune , Multimerización de Proteína , Bacterias/genética , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/genética , Bacteriófagos/inmunología , Bacteriófagos/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Desoxirribonucleasas/química , Desoxirribonucleasas/metabolismo , Desoxirribonucleasas/ultraestructura , ADN Viral/química , ADN Viral/metabolismo , ADN Viral/ultraestructura
6.
Nature ; 623(7987): 601-607, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37853129

RESUMEN

Many bacteria use CRISPR-Cas systems to combat mobile genetic elements, such as bacteriophages and plasmids1. In turn, these invasive elements have evolved anti-CRISPR proteins to block host immunity2,3. Here we unveil a distinct type of CRISPR-Cas Inhibition strategy that is based on small non-coding RNA anti-CRISPRs (Racrs). Racrs mimic the repeats found in CRISPR arrays and are encoded in viral genomes as solitary repeat units4. We show that a prophage-encoded Racr strongly inhibits the type I-F CRISPR-Cas system by interacting specifically with Cas6f and Cas7f, resulting in the formation of an aberrant Cas subcomplex. We identified Racr candidates for almost all CRISPR-Cas types encoded by a diverse range of viruses and plasmids, often in the genetic context of other anti-CRISPR genes5. Functional testing of nine candidates spanning the two CRISPR-Cas classes confirmed their strong immune inhibitory function. Our results demonstrate that molecular mimicry of CRISPR repeats is a widespread anti-CRISPR strategy, which opens the door to potential biotechnological applications6.


Asunto(s)
Bacterias , Bacteriófagos , Sistemas CRISPR-Cas , Imitación Molecular , ARN Viral , Bacterias/genética , Bacterias/inmunología , Bacterias/virología , Bacteriófagos/genética , Bacteriófagos/inmunología , Biotecnología/métodos , Biotecnología/tendencias , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Plásmidos/genética , Profagos/genética , Profagos/inmunología , ARN Viral/genética
8.
Nature ; 616(7956): 326-331, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36848932

RESUMEN

cGAS is an evolutionarily conserved enzyme that has a pivotal role in immune defence against infection1-3. In vertebrate animals, cGAS is activated by DNA to produce cyclic GMP-AMP (cGAMP)4,5, which leads to the expression of antimicrobial genes6,7. In bacteria, cyclic dinucleotide (CDN)-based anti-phage signalling systems (CBASS) have been discovered8-11. These systems are composed of cGAS-like enzymes and various effector proteins that kill bacteria on phage infection, thereby stopping phage spread. Of the CBASS systems reported, approximately 39% contain Cap2 and Cap3, which encode proteins with homology to ubiquitin conjugating (E1/E2) and deconjugating enzymes, respectively8,12. Although these proteins are required to prevent infection of some bacteriophages8, the mechanism by which the enzymatic activities exert an anti-phage effect is unknown. Here we show that Cap2 forms a thioester bond with the C-terminal glycine of cGAS and promotes conjugation of cGAS to target proteins in a process that resembles ubiquitin conjugation. The covalent conjugation of cGAS increases the production of cGAMP. Using a genetic screen, we found that the phage protein Vs.4 antagonized cGAS signalling by binding tightly to cGAMP (dissociation constant of approximately 30 nM) and sequestering it. A crystal structure of Vs.4 bound to cGAMP showed that Vs.4 formed a hexamer that was bound to three molecules of cGAMP. These results reveal a ubiquitin-like conjugation mechanism that regulates cGAS activity in bacteria and illustrates an arms race between bacteria and viruses through controlling CDN levels.


Asunto(s)
Bacterias , Proteínas Bacterianas , Bacteriófagos , Nucleotidiltransferasas , Ubiquitina , Animales , Bacterias/enzimología , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Bacteriófagos/inmunología , Nucleótidos Cíclicos/biosíntesis , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Ubiquitina/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Virales/metabolismo , Interacciones Microbiota-Huesped
9.
Nature ; 616(7956): 319-325, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36755092

RESUMEN

In all organisms, innate immune pathways sense infection and rapidly activate potent immune responses while avoiding inappropriate activation (autoimmunity). In humans, the innate immune receptor cyclic GMP-AMP synthase (cGAS) detects viral infection to produce the nucleotide second messenger cyclic GMP-AMP (cGAMP), which initiates stimulator of interferon genes (STING)-dependent antiviral signalling1. Bacteria encode evolutionary predecessors of cGAS called cGAS/DncV-like nucleotidyltransferases2 (CD-NTases), which detect bacteriophage infection and produce diverse nucleotide second messengers3. How bacterial CD-NTase activation is controlled remains unknown. Here we show that CD-NTase-associated protein 2 (Cap2) primes bacterial CD-NTases for activation through a ubiquitin transferase-like mechanism. A cryo-electron microscopy structure of the Cap2-CD-NTase complex reveals Cap2 as an all-in-one ubiquitin transferase-like protein, with distinct domains resembling eukaryotic E1 and E2 proteins. The structure captures a reactive-intermediate state with the CD-NTase C terminus positioned in the Cap2 E1 active site and conjugated to AMP. Cap2 conjugates the CD-NTase C terminus to a target molecule that primes the CD-NTase for increased cGAMP production. We further demonstrate that a specific endopeptidase, Cap3, balances Cap2 activity by cleaving CD-NTase-target conjugates. Our data demonstrate that bacteria control immune signalling using an ancient, minimized ubiquitin transferase-like system and provide insight into the evolution of the E1 and E2 machinery across domains of life.


Asunto(s)
Bacterias , Proteínas Bacterianas , Inmunidad Innata , Nucleotidiltransferasas , Humanos , Bacterias/enzimología , Bacterias/inmunología , Bacterias/metabolismo , Microscopía por Crioelectrón , Nucleotidiltransferasas/metabolismo , Ubiquitinas/metabolismo , Bacteriófagos/inmunología , Sistemas de Mensajero Secundario , Dominio Catalítico , Proteínas Bacterianas/metabolismo , Adenosina Monofosfato/metabolismo
10.
Nature ; 614(7946): 168-174, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36423657

RESUMEN

CRISPR defence systems such as the well-known DNA-targeting Cas9 and the RNA-targeting type III systems are widespread in prokaryotes1,2. The latter orchestrates a complex antiviral response that is initiated through the synthesis of cyclic oligoadenylates after recognition of foreign RNA3-5. Among the large set of proteins that are linked to type III systems and predicted to bind cyclic oligoadenylates6,7, a CRISPR-associated Lon protease (CalpL) stood out to us. CalpL contains a sensor domain of the SAVED family7 fused to a Lon protease effector domain. However, the mode of action of this effector is unknown. Here we report the structure and function of CalpL and show that this soluble protein forms a stable tripartite complex with two other proteins, CalpT and CalpS, that are encoded on the same operon. After activation by cyclic tetra-adenylate (cA4), CalpL oligomerizes and specifically cleaves the MazF homologue CalpT, which releases the extracytoplasmic function σ factor CalpS from the complex. Our data provide a direct connection between CRISPR-based detection of foreign nucleic acids and transcriptional regulation. Furthermore, the presence of a SAVED domain that binds cyclic tetra-adenylate in a CRISPR effector reveals a link to the cyclic-oligonucleotide-based antiphage signalling system.


Asunto(s)
Bacterias , Bacteriófagos , Proteínas Asociadas a CRISPR , Sistemas CRISPR-Cas , Nucleótidos Cíclicos , Proteasa La , Bacterias/enzimología , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Bacteriófagos/inmunología , Bacteriófagos/metabolismo , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiología , AMP Cíclico/análogos & derivados , AMP Cíclico/química , Activación Enzimática , Regulación Bacteriana de la Expresión Génica , Nucleótidos Cíclicos/inmunología , Nucleótidos Cíclicos/metabolismo , Operón , Proteasa La/química , Proteasa La/metabolismo , ARN Viral , Factor sigma , Transcripción Genética
11.
Nature ; 612(7938): 132-140, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36385533

RESUMEN

Bacteria have evolved diverse immunity mechanisms to protect themselves against the constant onslaught of bacteriophages1-3. Similar to how eukaryotic innate immune systems sense foreign invaders through pathogen-associated molecular patterns4 (PAMPs), many bacterial immune systems that respond to bacteriophage infection require phage-specific triggers to be activated. However, the identities of such triggers and the sensing mechanisms remain largely unknown. Here we identify and investigate the anti-phage function of CapRelSJ46, a fused toxin-antitoxin system that protects Escherichia coli against diverse phages. Using genetic, biochemical and structural analyses, we demonstrate that the C-terminal domain of CapRelSJ46 regulates the toxic N-terminal region, serving as both antitoxin and phage infection sensor. Following infection by certain phages, newly synthesized major capsid protein binds directly to the C-terminal domain of CapRelSJ46 to relieve autoinhibition, enabling the toxin domain to pyrophosphorylate tRNAs, which blocks translation to restrict viral infection. Collectively, our results reveal the molecular mechanism by which a bacterial immune system directly senses a conserved, essential component of phages, suggesting a PAMP-like sensing model for toxin-antitoxin-mediated innate immunity in bacteria. We provide evidence that CapRels and their phage-encoded triggers are engaged in a 'Red Queen conflict'5, revealing a new front in the intense coevolutionary battle between phages and bacteria. Given that capsid proteins of some eukaryotic viruses are known to stimulate innate immune signalling in mammalian hosts6-10, our results reveal a deeply conserved facet of immunity.


Asunto(s)
Bacteriófagos , Proteínas de la Cápside , Escherichia coli , Inmunidad Innata , Animales , Antitoxinas/inmunología , Bacteriófagos/inmunología , Proteínas de la Cápside/inmunología , Escherichia coli/inmunología , Escherichia coli/virología , Eucariontes/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología
12.
Nature ; 611(7935): 326-331, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36174646

RESUMEN

The Toll/interleukin-1 receptor (TIR) domain is a key component of immune receptors that identify pathogen invasion in bacteria, plants and animals1-3. In the bacterial antiphage system Thoeris, as well as in plants, recognition of infection stimulates TIR domains to produce an immune signalling molecule whose molecular structure remains elusive. This molecule binds and activates the Thoeris immune effector, which then executes the immune function1. We identified a large family of phage-encoded proteins, denoted here as Thoeris anti-defence 1 (Tad1), that inhibit Thoeris immunity. We found that Tad1 proteins are 'sponges' that bind and sequester the immune signalling molecule produced by TIR-domain proteins, thus decoupling phage sensing from immune effector activation and rendering Thoeris inactive. Tad1 can also efficiently sequester molecules derived from a plant TIR-domain protein, and a high-resolution crystal structure of Tad1 bound to a plant-derived molecule showed a unique chemical structure of 1 ''-2' glycocyclic ADPR (gcADPR). Our data furthermore suggest that Thoeris TIR proteins produce a closely related molecule, 1''-3' gcADPR, which activates ThsA an order of magnitude more efficiently than the plant-derived 1''-2' gcADPR. Our results define the chemical structure of a central immune signalling molecule and show a new mode of action by which pathogens can suppress host immunity.


Asunto(s)
Bacterias , Bacteriófagos , Dominios Proteicos , Receptores de Interleucina-1 , Transducción de Señal , Receptores Toll-Like , Proteínas Virales , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas de Plantas/antagonistas & inhibidores , Proteínas de Plantas/química , Proteínas de Plantas/inmunología , Proteínas de Plantas/metabolismo , Receptores de Interleucina-1/química , Transducción de Señal/inmunología , Bacteriófagos/química , Bacteriófagos/inmunología , Bacteriófagos/metabolismo , Proteínas Virales/química , Proteínas Virales/inmunología , Proteínas Virales/metabolismo , Receptores Toll-Like/química , Cristalografía por Rayos X
13.
Nature ; 608(7922): 429-435, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35922510

RESUMEN

Bacteria encode myriad defences that target the genomes of infecting bacteriophage, including restriction-modification and CRISPR-Cas systems1. In response, one family of large bacteriophages uses a nucleus-like compartment to protect its replicating genomes by excluding host defence factors2-4. However, the principal composition and structure of this compartment remain unknown. Here we find that the bacteriophage nuclear shell assembles primarily from one protein, which we name chimallin (ChmA). Combining cryo-electron tomography of nuclear shells in bacteriophage-infected cells and cryo-electron microscopy of a minimal chimallin compartment in vitro, we show that chimallin self-assembles as a flexible sheet into closed micrometre-scale compartments. The architecture and assembly dynamics of the chimallin shell suggest mechanisms for its nucleation and growth, and its role as a scaffold for phage-encoded factors mediating macromolecular transport, cytoskeletal interactions, and viral maturation.


Asunto(s)
Bacterias , Bacteriófagos , Compartimento Celular , Proteínas Virales , Ensamble de Virus , Bacterias/citología , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Bacteriófagos/química , Bacteriófagos/inmunología , Bacteriófagos/fisiología , Bacteriófagos/ultraestructura , Microscopía por Crioelectrón , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/ultraestructura
14.
Nature ; 608(7924): 803-807, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35859168

RESUMEN

Stimulator of interferon genes (STING) is an antiviral signalling protein that is broadly conserved in both innate immunity in animals and phage defence in prokaryotes1-4. Activation of STING requires its assembly into an oligomeric filament structure through binding of a cyclic dinucleotide4-13, but the molecular basis of STING filament assembly and extension remains unknown. Here we use cryogenic electron microscopy to determine the structure of the active Toll/interleukin-1 receptor (TIR)-STING filament complex from a Sphingobacterium faecium cyclic-oligonucleotide-based antiphage signalling system (CBASS) defence operon. Bacterial TIR-STING filament formation is driven by STING interfaces that become exposed on high-affinity recognition of the cognate cyclic dinucleotide signal c-di-GMP. Repeating dimeric STING units stack laterally head-to-head through surface interfaces, which are also essential for human STING tetramer formation and downstream immune signalling in mammals5. The active bacterial TIR-STING structure reveals further cross-filament contacts that brace the assembly and coordinate packing of the associated TIR NADase effector domains at the base of the filament to drive NAD+ hydrolysis. STING interface and cross-filament contacts are essential for cell growth arrest in vivo and reveal a stepwise mechanism of activation whereby STING filament assembly is required for subsequent effector activation. Our results define the structural basis of STING filament formation in prokaryotic antiviral signalling.


Asunto(s)
Proteínas Bacterianas , Microscopía por Crioelectrón , Proteínas de la Membrana , Receptores de Interleucina-1 , Sphingobacterium , Receptores Toll-Like , Animales , Antivirales/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/inmunología , Fosfatos de Dinucleósidos/metabolismo , Humanos , Inmunidad Innata , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Operón/genética , Receptores de Interleucina-1/química , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/ultraestructura , Sphingobacterium/química , Sphingobacterium/genética , Sphingobacterium/ultraestructura , Sphingobacterium/virología , Receptores Toll-Like/química , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Receptores Toll-Like/ultraestructura
15.
Nucleic Acids Res ; 50(W1): W541-W550, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35639517

RESUMEN

Most bacteria and archaea possess multiple antiviral defence systems that protect against infection by phages, archaeal viruses and mobile genetic elements. Our understanding of the diversity of defence systems has increased greatly in the last few years, and many more systems likely await discovery. To identify defence-related genes, we recently developed the Prokaryotic Antiviral Defence LOCator (PADLOC) bioinformatics tool. To increase the accessibility of PADLOC, we describe here the PADLOC web server (freely available at https://padloc.otago.ac.nz), allowing users to analyse whole genomes, metagenomic contigs, plasmids, phages and archaeal viruses. The web server includes a more than 5-fold increase in defence system types detected (since the first release) and expanded functionality enabling detection of CRISPR arrays and retron ncRNAs. Here, we provide user information such as input options, description of the multiple outputs, limitations and considerations for interpretation of the results, and guidance for subsequent analyses. The PADLOC web server also houses a precomputed database of the defence systems in > 230,000 RefSeq genomes. These data reveal two taxa, Campylobacterota and Spriochaetota, with unusual defence system diversity and abundance. Overall, the PADLOC web server provides a convenient and accessible resource for the detection of antiviral defence systems.


Asunto(s)
Archaea , Bacterias , Genoma Microbiano , Genómica , Internet , Programas Informáticos , Archaea/genética , Archaea/virología , Bacterias/genética , Bacterias/virología , Bacteriófagos/inmunología , Genoma Microbiano/genética , Plásmidos/genética , Células Procariotas/metabolismo , Células Procariotas/virología , Computadores , Genómica/métodos
16.
Nature ; 600(7887): 116-120, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34853457

RESUMEN

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.


Asunto(s)
Bacillus/inmunología , Bacillus/virología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Bacteriófagos/inmunología , Receptores de Interleucina-1/química , Transducción de Señal/inmunología , Receptores Toll-Like/química , ADP-Ribosa Cíclica/análogos & derivados , ADP-Ribosa Cíclica/metabolismo , Evolución Molecular , Modelos Moleculares , NAD/metabolismo , Dominios Proteicos , Especificidad por Sustrato/inmunología
17.
Nat Commun ; 12(1): 6817, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819498

RESUMEN

Bacterial defenses against phage, which include CRISPR-mediated immunity and other mechanisms, can carry substantial growth rate costs and can be rapidly lost when pathogens are eliminated. How bacteria preserve their molecular defenses despite their costs, in the face of variable pathogen levels and inter-strain competition, remains a major unsolved problem in evolutionary biology. Here, we present a multilevel model that incorporates biophysics of molecular binding, host-pathogen population dynamics, and ecological dynamics across a large number of independent territories. Using techniques of game theory and non-linear dynamical systems, we show that by maintaining a non-zero failure rate of defenses, hosts sustain sufficient levels of pathogen within an ecology to select against loss of the defense. This resistance switching strategy is evolutionarily stable, and provides a powerful evolutionary mechanism that maintains host-pathogen interactions, selects against cheater strains that avoid the costs of immunity, and enables co-evolutionary dynamics in a wide range of systems.


Asunto(s)
Bacterias/inmunología , Bacteriófagos/inmunología , Interacciones Huésped-Patógeno/inmunología , Memoria Inmunológica
18.
Mol Cell ; 81(24): 5039-5051.e5, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34784509

RESUMEN

Cyclic oligonucleotide-based antiphage signaling systems (CBASS) are antiviral defense operons that protect bacteria from phage replication. Here, we discover a widespread class of CBASS transmembrane (TM) effector proteins that respond to antiviral nucleotide signals and limit phage propagation through direct membrane disruption. Crystal structures of the Yersinia TM effector Cap15 reveal a compact 8-stranded ß-barrel scaffold that forms a cyclic dinucleotide receptor domain that oligomerizes upon activation. We demonstrate that activated Cap15 relocalizes throughout the cell and specifically induces rupture of the inner membrane. Screening for active effectors, we identify the function of distinct families of CBASS TM effectors and demonstrate that cell death via disruption of inner-membrane integrity is a common mechanism of defense. Our results reveal the function of the most prominent class of effector protein in CBASS immunity and define disruption of the inner membrane as a widespread strategy of abortive infection in bacterial phage defense.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteriófagos/patogenicidad , Membrana Celular/virología , Escherichia coli/virología , Yersinia/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Bacteriófagos/inmunología , Muerte Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Escherichia coli/genética , Escherichia coli/inmunología , Escherichia coli/metabolismo , Interacciones Huésped-Patógeno , Ligandos , Conformación Proteica , Multimerización de Proteína , Transporte de Proteínas , Transducción de Señal , Relación Estructura-Actividad , Yersinia/genética
19.
Front Immunol ; 12: 752898, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630431

RESUMEN

The Toll/interleukin-1 receptor (TIR) domain is the signature signalling motif of innate immunity, with essential roles in innate immune signalling in bacteria, plants, and animals. TIR domains canonically function as scaffolds, with stimulus-dependent multimerization generating binding sites for signalling molecules such as kinases and ligases that activate downstream immune mechanisms. Recent studies have dramatically expanded our understanding of the TIR domain, demonstrating that the primordial function of the TIR domain is to metabolize NAD+. Mammalian SARM1, the central executioner of pathological axon degeneration, is the founding member of the TIR-domain class of NAD+ hydrolases. This unexpected NADase activity of TIR domains is evolutionarily conserved, with archaeal, bacterial, and plant TIR domains all sharing this catalytic function. Moreover, this enzymatic activity is essential for the innate immune function of these proteins. These evolutionary relationships suggest a link between SARM1 and ancient self-defense mechanisms that has only been strengthened by the recent discovery of the SARM1 activation mechanism which, we will argue, is strikingly similar to bacterial toxin-antitoxin systems. In this brief review we will describe the regulation and function of SARM1 in programmed axon self-destruction, and highlight the parallels between the SARM1 axon degeneration pathway and bacterial innate immune mechanisms.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas del Dominio Armadillo/inmunología , Proteínas del Citoesqueleto/inmunología , Inmunidad Innata/inmunología , NAD+ Nucleosidasa/inmunología , Animales , Bacteriófagos/inmunología , Evolución Biológica , Humanos , Sistemas Toxina-Antitoxina/inmunología
20.
Mol Med Rep ; 24(5)2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34490477

RESUMEN

Phage display technology (PD) is a powerful technique for the generation of tumor­targeting antibodies. However, there are a number of different selection methods established in different laboratories around the world. Cell­based PD panning methods using primary tumor cells are particularly heterogeneous between laboratories, which can lead to inconsistent results. Therefore, the present study evaluated different cell­based PD selection methods regarding their potential to generate acute myeloid leukemia (AML) blast­binding antibodies. In addition to this evaluation, the present study improved the PD procedure by optimizing selection as well as depletion strategies. To the best of our knowledge, the current study demonstrated for the first time that antigen diversity during the depletion step is of importance for the enrichment of tumor­targeting phage antibodies. It is demonstrated that medium levels of depletion antigen diversity led to the most promising antibody candidates. In addition, it was determined that purification of blast cells from patients with AML by immunomagnetic separation ameliorated the selection of AML­binding phages during panning. Furthermore, suggesting a common design­related mechanism using a 'single­pot' PD library, such as the well­known Tomlinson single­chain fragment variable (scFv) library, the present study identified specific binding consensus phage particles in independent panning procedures. By means of these optimized strategies, four promising AML blast­binding phage particles were isolated and soluble scFv­Fc (scFv cloned to a fragment crystallizable of an IgG2a mouse antibody) fusion proteins were produced. These scFv­Fc antibodies bound the surface of AML blasts and were successfully internalized into their cytoplasm, indicating that they are potential immunoconjugate candidates for AML immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Técnicas de Visualización de Superficie Celular/métodos , Inmunoterapia/métodos , Leucemia Mieloide Aguda/terapia , Especificidad de Anticuerpos , Bacteriófagos/inmunología , Células HEK293 , Humanos , Cultivo Primario de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA