Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem J ; 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38785320

RESUMEN

CRISPR-Cas systems confer adaptive immunity in prokaryotes, facilitating the recognition and destruction of invasive nucleic acids. Type III CRISPR systems comprise large, multisubunit ribonucleoprotein complexes with a catalytic Cas10 subunit. When activated by the detection of foreign RNA, Cas10 generates nucleotide signalling molecules that elicit an immune response by activating ancillary effector proteins. Among these systems, the Bacteroides fragilis type III CRISPR system was recently shown to produce a novel signal molecule, SAM-AMP, by conjugating ATP and SAM. SAM-AMP regulates a membrane effector of the CorA family to provide immunity. Here, we focus on NYN, a ribonuclease encoded within this system, probing its potential involvement in crRNA maturation. Structural modelling and in vitro ribonuclease assays reveal that NYN displays robust sequence-nonspecific, Mn2+-dependent ssRNA-cleavage activity. Our findings suggest a role for NYN in trimming crRNA intermediates into mature crRNAs, which is necessary for type III CRISPR antiviral defence. This study sheds light on the functional relevance of CRISPR-associated NYN proteins and highlights the complexity of CRISPR-mediated defence strategies in bacteria.

2.
Nat Microbiol ; 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589469

RESUMEN

Prokaryotic antiviral defence systems are frequently toxic for host cells and stringent regulation is required to ensure survival and fitness. These systems must be readily available in case of infection but tightly controlled to prevent activation of an unnecessary cellular response. Here we investigate how the bacterial cyclic oligonucleotide-based antiphage signalling system (CBASS) uses its intrinsic protein modification system to regulate the nucleotide cyclase. By integrating a type II CBASS system from Bacillus cereus into the model organism Bacillus subtilis, we show that the protein-conjugating Cap2 (CBASS associated protein 2) enzyme links the cyclase exclusively to the conserved phage shock protein A (PspA) in the absence of phage. The cyclase-PspA conjugation is reversed by the deconjugating isopeptidase Cap3 (CBASS associated protein 3). We propose a model in which the cyclase is held in an inactive state by conjugation to PspA in the absence of phage, with conjugation released upon infection, priming the cyclase for activation.

3.
Nucleic Acids Res ; 52(6): 2761-2775, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38471818

RESUMEN

CRISPR-Cas provides adaptive immunity in prokaryotes. Type III CRISPR systems detect invading RNA and activate the catalytic Cas10 subunit, which generates a range of nucleotide second messengers to signal infection. These molecules bind and activate a diverse range of effector proteins that provide immunity by degrading viral components and/or by disturbing key aspects of cellular metabolism to slow down viral replication. Here, we focus on the uncharacterised effector Csx23, which is widespread in Vibrio cholerae. Csx23 provides immunity against plasmids and phage when expressed in Escherichia coli along with its cognate type III CRISPR system. The Csx23 protein localises in the membrane using an N-terminal transmembrane α-helical domain and has a cytoplasmic C-terminal domain that binds cyclic tetra-adenylate (cA4), activating its defence function. Structural studies reveal a tetrameric structure with a novel fold that binds cA4 specifically. Using pulse EPR, we demonstrate that cA4 binding to the cytoplasmic domain of Csx23 results in a major perturbation of the transmembrane domain, consistent with the opening of a pore and/or disruption of membrane integrity. This work reveals a new class of cyclic nucleotide binding protein and provides key mechanistic detail on a membrane-associated CRISPR effector.


Many anti-viral defence systems generate a cyclic nucleotide signal that activates cellular defences in response to infection. Type III CRISPR systems use a specialised polymerase to make cyclic oligoadenylate (cOA) molecules from ATP. These can bind and activate a range of effector proteins that slow down viral replication. In this study, we focussed on the Csx23 effector from the human pathogen Vibrio cholerae ­ a trans-membrane protein that binds a cOA molecule, leading to anti-viral immunity. Structural studies revealed a new class of nucleotide recognition domain, where cOA binding is transmitted to changes in the trans-membrane domain, most likely resulting in membrane depolarisation. This study highlights the diversity of mechanisms for anti-viral defence via nucleotide signalling.


Asunto(s)
Proteínas Bacterianas , Proteínas Asociadas a CRISPR , Vibrio cholerae , Nucleótidos de Adenina/metabolismo , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Nucleótidos Cíclicos , Sistemas de Mensajero Secundario , Proteínas Bacterianas/metabolismo , Vibrio cholerae/metabolismo
4.
Curr Opin Microbiol ; 78: 102436, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38368839

RESUMEN

Bacteria have evolved a variety of defence mechanisms to protect against mobile genetic elements, including restriction-modification systems and CRISPR-Cas. In recent years, dozens of previously unknown defence systems (DSs) have been discovered. Notably, diverse DSs often coexist within the same genome, and some co-occur at frequencies significantly higher than would be expected by chance, implying potential synergistic interactions. Recent studies have provided evidence of defence mechanisms that enhance or complement one another. Here, we review the interactions between DSs at the mechanistic, regulatory, ecological and evolutionary levels.


Asunto(s)
Bacteriófagos , Sistemas CRISPR-Cas , Bacterias/genética , Evolución Biológica , Bacteriófagos/genética
5.
Nature ; 622(7984): 826-833, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37853119

RESUMEN

CRISPR systems are widespread in the prokaryotic world, providing adaptive immunity against mobile genetic elements1,2. Type III CRISPR systems, with the signature gene cas10, use CRISPR RNA to detect non-self RNA, activating the enzymatic Cas10 subunit to defend the cell against mobile genetic elements either directly, via the integral histidine-aspartate (HD) nuclease domain3-5 or indirectly, via synthesis of cyclic oligoadenylate second messengers to activate diverse ancillary effectors6-9. A subset of type III CRISPR systems encode an uncharacterized CorA-family membrane protein and an associated NrN family phosphodiesterase that are predicted to function in antiviral defence. Here we demonstrate that the CorA-associated type III-B (Cmr) CRISPR system from Bacteroides fragilis provides immunity against mobile genetic elements when expressed in Escherichia coli. However, B. fragilis Cmr does not synthesize cyclic oligoadenylate species on activation, instead generating S-adenosyl methionine (SAM)-AMP (SAM is also known as AdoMet) by conjugating ATP to SAM via a phosphodiester bond. Once synthesized, SAM-AMP binds to the CorA effector, presumably leading to cell dormancy or death by disruption of the membrane integrity. SAM-AMP is degraded by CRISPR-associated phosphodiesterases or a SAM-AMP lyase, potentially providing an 'off switch' analogous to cyclic oligoadenylate-specific ring nucleases10. SAM-AMP thus represents a new class of second messenger for antiviral signalling, which may function in different roles in diverse cellular contexts.


Asunto(s)
Adenosina Trifosfato , Bacteroides fragilis , Sistemas CRISPR-Cas , Escherichia coli , S-Adenosilmetionina , Sistemas de Mensajero Secundario , Adenosina Trifosfato/metabolismo , Bacteroides fragilis/enzimología , Bacteroides fragilis/genética , Bacteroides fragilis/inmunología , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Sistemas CRISPR-Cas/fisiología , Endonucleasas/química , Endonucleasas/metabolismo , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , ARN/inmunología , ARN/metabolismo , S-Adenosilmetionina/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
6.
Nucleic Acids Res ; 51(19): 10590-10605, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37747760

RESUMEN

Type III CRISPR systems synthesize cyclic oligoadenylate (cOA) second messengers as part of a multi-faceted immune response against invading mobile genetic elements (MGEs). cOA activates non-specific CRISPR ancillary defence nucleases to create a hostile environment for MGE replication. Csm6 ribonucleases bind cOA using a CARF (CRISPR-associated Rossmann Fold) domain, resulting in activation of a fused HEPN (Higher Eukaryotes and Prokaryotes Nucleotide binding) ribonuclease domain. Csm6 enzymes are widely used in a new generation of diagnostic assays for the detection of specific nucleic acid species. However, the activation mechanism is not fully understood. Here we characterised the cyclic hexa-adenylate (cA6) activated Csm6' ribonuclease from the industrially important bacterium Streptococcus thermophilus. Crystal structures of Csm6' in the inactive and cA6 bound active states illuminate the conformational changes which trigger mRNA destruction. Upon binding of cA6, there is a close to 60° rotation between the CARF and HEPN domains, which causes the 'jaws' of the HEPN domain to open and reposition active site residues. Key to this transition is the 6H domain, a right-handed solenoid domain connecting the CARF and HEPN domains, which transmits the conformational changes for activation.


Asunto(s)
Ribonucleasas , Streptococcus thermophilus , Dominio Catalítico , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Sistemas CRISPR-Cas , Nucleótidos Cíclicos , Ribonucleasas/química , Ribonucleasas/metabolismo , Sistemas de Mensajero Secundario , Streptococcus thermophilus/química
7.
Microbiology (Reading) ; 169(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37526970

RESUMEN

The CRISPR-Cas system functions as a prokaryotic immune system and is highly diverse, with six major types and numerous sub-types. The most abundant are type I CRISPR systems, which utilize a multi-subunit effector, Cascade, and a CRISPR RNA (crRNA) to detect invading DNA species. Detection leads to DNA loading of the Cas3 helicase-nuclease, leading to long-range deletions in the targeted DNA, thus providing immunity against mobile genetic elements (MGE). Here, we focus on the type I-G system, a streamlined, 4-subunit complex with an atypical Cas3 enzyme. We demonstrate that Cas3 helicase activity is not essential for immunity against MGE in vivo and explore applications of the Thioalkalivibrio sulfidiphilus Cascade effector for genome engineering in Escherichia coli. Long-range, bidirectional deletions were observed when the lacZ gene was targeted. Deactivation of the Cas3 helicase activity dramatically altered the types of deletions observed, with small deletions flanked by direct repeats that are suggestive of microhomology mediated end joining. When donor DNA templates were present, both the wild-type and helicase-deficient systems promoted homology-directed repair (HDR), with the latter system providing improvements in editing efficiency, suggesting that a single nick in the target site may promote HDR in E. coli using the type I-G system. These findings open the way for further application of the type I-G CRISPR systems in genome engineering.


Asunto(s)
Proteínas Asociadas a CRISPR , Escherichia coli , Escherichia coli/genética , Escherichia coli/metabolismo , Replicación del ADN , ADN , Sistemas CRISPR-Cas , ADN Helicasas/genética , ADN Helicasas/metabolismo , Genoma Bacteriano/genética , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo
8.
Curr Res Struct Biol ; 5: 100098, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36843655

RESUMEN

CRISPR-Cas is a prokaryotic adaptive immune system, classified into six different types, each characterised by a signature protein. Type III systems, classified based on the presence of a Cas10 subunit, are rather diverse multi-subunit assemblies with a range of enzymatic activities and downstream ancillary effectors. The broad array of current biotechnological CRISPR applications is mainly based on proteins classified as Type II, however recent developments established the feasibility and efficacy of multi-protein Type III CRISPR-Cas effector complexes as RNA-targeting tools in eukaryotes. The crenarchaeon Saccharolobus solfataricus has two type III system subtypes (III-B and III-D). Here, we report the cryo-EM structure of the Csm Type III-D complex from S. solfataricus (SsoCsm), which uses CRISPR RNA to bind target RNA molecules, activating the Cas10 subunit for antiviral defence. The structure reveals the complex organisation, subunit/subunit connectivity and protein/guide RNA interactions of the SsoCsm complex, one of the largest CRISPR effectors known.

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.
Nucleic Acids Res ; 50(19): 11214-11228, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36305833

RESUMEN

Type I CRISPR systems are the most common CRISPR type found in bacteria. They use a multisubunit effector, guided by crRNA, to detect and bind dsDNA targets, forming an R-loop and recruiting the Cas3 enzyme to facilitate target DNA destruction, thus providing immunity against mobile genetic elements. Subtypes have been classified into families A-G, with type I-G being the least well understood. Here, we report the composition, structure and function of the type I-G Cascade CRISPR effector from Thioalkalivibrio sulfidiphilus, revealing key new molecular details. The unique Csb2 subunit processes pre-crRNA, remaining bound to the 3' end of the mature crRNA, and seven Cas7 subunits form the backbone of the effector. Cas3 associates stably with the effector complex via the Cas8g subunit and is important for target DNA recognition. Structural analysis by cryo-Electron Microscopy reveals a strikingly curved backbone conformation with Cas8g spanning the belly of the structure. These biochemical and structural insights shed new light on the diversity of type I systems and open the way to applications in genome engineering.


Asunto(s)
Proteínas Bacterianas , Proteínas Asociadas a CRISPR , Ectothiorhodospiraceae , Proteínas Asociadas a CRISPR/química , Sistemas CRISPR-Cas , Microscopía por Crioelectrón , ADN/química , Proteínas Bacterianas/química
12.
Nature ; 608(7924): 808-812, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948638

RESUMEN

Cyclic nucleotide signalling is a key component of antiviral defence in all domains of life. Viral detection activates a nucleotide cyclase to generate a second messenger, resulting in activation of effector proteins. This is exemplified by the metazoan cGAS-STING innate immunity pathway1, which originated in bacteria2. These defence systems require a sensor domain to bind the cyclic nucleotide and are often coupled with an effector domain that, when activated, causes cell death by destroying essential biomolecules3. One example is the Toll/interleukin-1 receptor (TIR) domain, which degrades the essential cofactor NAD+ when activated in response to infection in plants and bacteria2,4,5 or during programmed nerve cell death6. Here we show that a bacterial antiviral defence system generates a cyclic tri-adenylate that binds to a TIR-SAVED effector, acting as the 'glue' to allow assembly of an extended superhelical solenoid structure. Adjacent TIR subunits interact to organize and complete a composite active site, allowing NAD+ degradation. Activation requires extended filament formation, both in vitro and in vivo. Our study highlights an example of large-scale molecular assembly controlled by cyclic nucleotides and reveals key details of the mechanism of TIR enzyme activation.


Asunto(s)
Bacterias , Nucleótidos Cíclicos , Receptores de Interleucina-1 , Receptores Toll-Like , Animales , Antivirales/inmunología , Antivirales/metabolismo , Bacterias/inmunología , Bacterias/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , NAD/metabolismo , Nucleótidos Cíclicos/química , Nucleótidos Cíclicos/inmunología , Nucleótidos Cíclicos/metabolismo , Receptores de Interleucina-1/química , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Sistemas de Mensajero Secundario , Receptores Toll-Like/química , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
13.
Annu Rev Virol ; 9(1): 451-468, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-35567297

RESUMEN

Advances in our understanding of prokaryotic antiphage defense mechanisms in the past few years have revealed a multitude of new cyclic nucleotide signaling molecules that play a crucial role in switching infected cells into an antiviral state. Defense pathways including type III CRISPR (clustered regularly interspaced palindromic repeats), CBASS (cyclic nucleotide-based antiphage signaling system), PYCSAR (pyrimidine cyclase system for antiphage resistance), and Thoeris all use cyclic nucleotides as second messengers to activate a diverse range of effector proteins. These effectors typically degrade or disrupt key cellular components such as nucleic acids, membranes, or metabolites, slowing down viral replication kinetics at great cost to the infected cell. Mechanisms to manipulate the levels of cyclic nucleotides are employed by cells to regulate defense pathways and by viruses to subvert them. Here we review the discovery and mechanism of the key pathways, signaling molecules and effectors, parallels and differences between the systems, open questions, and prospects for future research in this area.


Asunto(s)
Bacteriófagos , Ácidos Nucleicos , Antivirales , Bacteriófagos/genética , Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Nucleótidos Cíclicos , Pirimidinas
14.
R Soc Open Sci ; 9(1): 211981, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35116170
15.
Nucleic Acids Res ; 49(22): 13122-13134, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34871408

RESUMEN

Type III CRISPR systems detect invading RNA, resulting in the activation of the enzymatic Cas10 subunit. The Cas10 cyclase domain generates cyclic oligoadenylate (cOA) second messenger molecules, activating a variety of effector nucleases that degrade nucleic acids to provide immunity. The prophage-encoded Vibrio metoecus type III-B (VmeCmr) locus is uncharacterised, lacks the HD nuclease domain in Cas10 and encodes a NucC DNA nuclease effector that is also found associated with Cyclic-oligonucleotide-based anti-phage signalling systems (CBASS). Here we demonstrate that VmeCmr is activated by target RNA binding, generating cyclic-triadenylate (cA3) to stimulate a robust NucC-mediated DNase activity. The specificity of VmeCmr is probed, revealing the importance of specific nucleotide positions in segment 1 of the RNA duplex and the protospacer flanking sequence (PFS). We harness this programmable system to demonstrate the potential for a highly specific and sensitive assay for detection of the SARS-CoV-2 virus RNA with a limit of detection (LoD) of 2 fM using a commercial plate reader without any extrinsic amplification step. The sensitivity is highly dependent on the guide RNA used, suggesting that target RNA secondary structure plays an important role that may also be relevant in vivo.


Asunto(s)
Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas/genética , Endodesoxirribonucleasas/metabolismo , Endorribonucleasas/metabolismo , ARN Viral/genética , SARS-CoV-2/genética , Animales , COVID-19 , Línea Celular , Chlorocebus aethiops , Humanos , Profagos/genética , Células Vero , Vibrio/virología
16.
RNA ; 2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986148

RESUMEN

In prokaryotes, CRISPR-Cas immune systems recognise and cleave foreign nucleic acids to defend against Mobile Genetic Elements (MGEs). Type III CRISPR-Cas complexes also synthesise cyclic oligoadenylate (cOA) second messengers, which activate CRISPR ancillary proteins involved in antiviral defence. In particular, cOA-stimulated nucleases degrade RNA and DNA non-specifically, which slows MGE replication but also impedes cell growth, necessitating mechanisms to eliminate cOA in order to mitigate collateral damage. Extant cOA is degraded by a new class of enzyme termed a 'ring nuclease', which cleaves cOA specifically and switches off CRISPR ancillary enzymes. Several ring nuclease families have been characterised to date, including a family used by MGEs to circumvent CRISPR immunity, and encompass diverse protein folds and distinct cOA cleavage mechanisms. In this review we outline cOA signalling, discuss how different ring nucleases regulate the cOA signalling pathway, and reflect on parallels between cyclic nucleotide-based immune systems to reveal new areas for exploration.

17.
Nucleic Acids Res ; 49(5): 2777-2789, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33590098

RESUMEN

Cells and organisms have a wide range of mechanisms to defend against infection by viruses and other mobile genetic elements (MGE). Type III CRISPR systems detect foreign RNA and typically generate cyclic oligoadenylate (cOA) second messengers that bind to ancillary proteins with CARF (CRISPR associated Rossman fold) domains. This results in the activation of fused effector domains for antiviral defence. The best characterised CARF family effectors are the Csm6/Csx1 ribonucleases and DNA nickase Can1. Here we investigate a widely distributed CARF family effector with a nuclease domain, which we name Can2 (CRISPR ancillary nuclease 2). Can2 is activated by cyclic tetra-adenylate (cA4) and displays both DNase and RNase activity, providing effective immunity against plasmid transformation and bacteriophage infection in Escherichia coli. The structure of Can2 in complex with cA4 suggests a mechanism for the cA4-mediated activation of the enzyme, whereby an active site cleft is exposed on binding the activator. These findings extend our understanding of type III CRISPR cOA signalling and effector function.


Asunto(s)
Proteínas Asociadas a CRISPR/química , Sistemas CRISPR-Cas , Desoxirribonucleasa I/química , Ribonucleasas/química , Clostridiales/enzimología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN/química , Desoxirribonucleasa I/metabolismo , Activación Enzimática , Escherichia coli/virología , Secuencias Repetitivas Esparcidas , Metales/química , Modelos Moleculares , Dominios Proteicos , Ribonucleasas/metabolismo
18.
Cell ; 182(1): 5-6, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32649879

RESUMEN

Increasingly, cyclic nucleotide second messengers are implicated in antiviral defense systems in bacteria and archaea as well as in eukaryotes. In this issue of Cell, Lowey et al. describe SAVED-a widespread, uncharacterized cyclic nucleotide sensor protein domain that activates cell defense systems. The structure of SAVED reveals links to the CRISPR system, which also generates cyclic nucleotides in response to viral infection.


Asunto(s)
Bacteriófagos , Virus , Archaea/genética , Bacterias/genética , Bacteriófagos/genética , Oligonucleótidos , Virus/genética
19.
Elife ; 92020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32597755

RESUMEN

Type III CRISPR systems detect foreign RNA and activate the cyclase domain of the Cas10 subunit, generating cyclic oligoadenylate (cOA) molecules that act as a second messenger to signal infection, activating nucleases that degrade the nucleic acid of both invader and host. This can lead to dormancy or cell death; to avoid this, cells need a way to remove cOA from the cell once a viral infection has been defeated. Enzymes specialised for this task are known as ring nucleases, but are limited in their distribution. Here, we demonstrate that the widespread CRISPR associated protein Csx3, previously described as an RNA deadenylase, is a ring nuclease that rapidly degrades cyclic tetra-adenylate (cA4). The enzyme has an unusual cooperative reaction mechanism involving an active site that spans the interface between two dimers, sandwiching the cA4 substrate. We propose the name Crn3 (CRISPR associated ring nuclease 3) for the Csx3 family.


Bacteria protect themselves from infections using a system called CRISPR-Cas, which helps the cells to detect and destroy invading threats. The type III CRISPR-Cas system, in particular, is one of the most widespread and efficient at killing viruses. When a bacterium is infected, the CRISPR-Cas system takes a fragment of the genetic material of the virus, and copies it into a molecule. These molecular 'police mugshots' are then loaded into a complex of Cas proteins that patrol the cell, looking for a match and destroying any virus that can be identified. Some Cas proteins also produce alarm signals, called cyclic oligoadenylates (cOAs), which can trigger additional defences. However, this process can damage the genetic material of the bacterium, harming or even killing the cell. Enzymes known as ring nucleases can promptly degrade cOAs and turn off this defence system before it causes harm. However, ring nucleases have only been found in a few species to date; how most bacteria deal with cOA toxicity has remained unknown. Here, Athukoralage et al. set out to determine whether a widespread enzyme known as Csx3, which is often associated with type III CRISPR-Cas systems, could be an alternative off switch for cOA triggered defences. Initial 'test tube' experiments with purified Csx3 proteins confirmed that the enzyme could indeed break down cOAs. A careful dissection of Csx3's molecular structure, using biochemical and biophysical techniques, revealed that it worked by 'sandwiching' a cOA molecule between two co-operating portions of the enzyme. As a final test, Csx3 was introduced into strains of bacteria genetically engineered to have a fully functional Type III CRISPR-Cas system. In these cells, Csx3 successfully turned off the Type III immune response. These results reveal a new way that bacteria avoid the toxic side effects of their own immune defences. Ultimately, this could pave the way for the development of anti-bacterial drugs that work by blocking Csx3 or similar proteins.


Asunto(s)
Archaeoglobus fulgidus/enzimología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ribonucleasas/metabolismo , Archaeoglobus fulgidus/genética , Proteínas Asociadas a CRISPR/metabolismo , Catálisis , Dominio Catalítico , Endonucleasas/metabolismo , Escherichia coli/metabolismo , Cinética , Methanosarcina , Modelos Moleculares , Oligonucleótidos/química , Multimerización de Proteína , ARN/metabolismo , Ribonucleasas/genética , Sistemas de Mensajero Secundario , Transducción de Señal
20.
Protein Expr Purif ; 174: 105660, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32473323

RESUMEN

Transcription factor IIH (TFIIH) plays essential roles in both the initiation of RNA Polymerase II-mediated transcription and the Nucleotide Excision Repair (NER) pathway in eukaryotes. In NER, the 7-subunit TFIIH Core sub-complex is responsible for the opening and extension of the DNA bubble created at the lesion site, utilizing the molecular motors XPB and XPD. Mutations in Core subunits are associated with a series of severe autosomal recessive disorders characterised by symptoms such as mild-to-extreme photosensitivity, premature ageing, physical and neurological anomalies, and in some cases an increased susceptibility to cancer. Although TFIIH Core has been successfully obtained in the past, the process has always remained challenging and laborious, involving many steps that severely hindered the amount of pure, active complex obtained. This has limited biochemical and functional studies of the NER process. Here we describe improved and simplified processes for the cloning, expression and purification of the 7-subunit TFIIH Core sub-complex. The combined use of auto-cleavable 2A-like sequences derived from the Foot-and-Mouth Disease Virus (FMDV) and the MultiBac™ cloning system, a powerful baculoviral expression vector specifically conceived for the obtaining of multi-subunit eukaryotic complexes, allowed us to obtain a single, 7-gene plasmid in a short time using regular restriction cloning strategies. Additionally, expression of the construct in High Five™ insect cells paired with a simple 5-step purification protocol allowed the extraction of a pure, active TFIIH Core sub-complex in milligram quantities.


Asunto(s)
Expresión Génica , Factor de Transcripción TFIIH , Animales , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Células Sf9 , Spodoptera , Factor de Transcripción TFIIH/biosíntesis , Factor de Transcripción TFIIH/genética , Factor de Transcripción TFIIH/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...