Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pharmacol Ther ; 258: 108653, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679204

RESUMEN

Cyclic nucleotides are important signaling molecules that play many critical physiological roles including controlling cell fate and development, regulation of metabolic processes, and responding to changes in the environment. Cyclic nucleotides are also pivotal regulators in immune signaling, orchestrating intricate processes that maintain homeostasis and defend against pathogenic threats. This review provides a comprehensive examination of the pharmacological potential of cyclic nucleotide signaling pathways within the realm of immunity. Beginning with an overview of the fundamental roles of cAMP and cGMP as ubiquitous second messengers, this review delves into the complexities of their involvement in immune responses. Special attention is given to the challenges associated with modulating these signaling pathways for therapeutic purposes, emphasizing the necessity for achieving cell-type specificity to avert unintended consequences. A major focus of the review is on the recent paradigm-shifting discoveries regarding specialized cyclic nucleotide signals in the innate immune system, notably the cGAS-STING pathway. The significance of cyclic dinucleotides, exemplified by 2'3'-cGAMP, in controlling immune responses against pathogens and cancer, is explored. The evolutionarily conserved nature of cyclic dinucleotides as antiviral agents, spanning across diverse organisms, underscores their potential as targets for innovative immunotherapies. Findings from the last several years have revealed a striking diversity of novel bacterial cyclic nucleotide second messengers which are involved in antiviral responses. Knowledge of the existence and precise identity of these molecules coupled with accurate descriptions of their associated immune defense pathways will be essential to the future development of novel antibacterial therapeutic strategies. The insights presented herein may help researchers navigate the evolving landscape of immunopharmacology as it pertains to cyclic nucleotides and point toward new avenues or lines of thinking about development of therapeutics against the pathways they regulate.


Asunto(s)
Nucleótidos Cíclicos , Transducción de Señal , Humanos , Animales , Nucleótidos Cíclicos/metabolismo , Inmunidad Innata , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Nucleotidiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo
3.
Cell Host Microbe ; 31(9): 1420-1422, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37708847

RESUMEN

Viral strategies to lyse host microbes can sometimes involve just a single gene, although the mechanism of action can be hard to discern. In Science, Orta et al. present structures of a protein complex in which protein E of bacteriophage φX174 functions to inhibit host peptidoglycan synthesis, thereby inducing lysis.

4.
Cell ; 186(15): 3261-3276.e20, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37379839

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is an enzyme in human cells that controls an immune response to cytosolic DNA. Upon binding DNA, cGAS synthesizes a nucleotide signal 2'3'-cGAMP that activates STING-dependent downstream immunity. Here, we discover that cGAS-like receptors (cGLRs) constitute a major family of pattern recognition receptors in innate immunity. Building on recent analysis in Drosophila, we identify >3,000 cGLRs present in nearly all metazoan phyla. A forward biochemical screening of 150 animal cGLRs reveals a conserved mechanism of signaling including response to dsDNA and dsRNA ligands and synthesis of isomers of the nucleotide signals cGAMP, c-UMP-AMP, and c-di-AMP. Combining structural biology and in vivo analysis in coral and oyster animals, we explain how synthesis of distinct nucleotide signals enables cells to control discrete cGLR-STING signaling pathways. Our results reveal cGLRs as a widespread family of pattern recognition receptors and establish molecular rules that govern nucleotide signaling in animal immunity.


Asunto(s)
Inmunidad Innata , Nucleotidiltransferasas , Humanos , Animales , Nucleotidiltransferasas/metabolismo , Inmunidad Innata/genética , Transducción de Señal/genética , ADN/metabolismo , Receptores de Reconocimiento de Patrones
5.
Curr Opin Microbiol ; 73: 102295, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37011504

RESUMEN

The innate immune system is the first line of defense against microbial pathogens. Many of the features of eukaryotic innate immunity have long been viewed as lineage-specific innovations, evolved to deal with the challenges and peculiarities of multicellular life. However, it has become increasingly apparent that in addition to evolving their own unique antiviral immune strategies, all lifeforms have some shared defense strategies in common. Indeed, critical fixtures of animal innate immunity bear striking resemblance in both structure and function to the multitude of diverse bacteriophage (phage) defense pathways discovered hidden in plain sight within the genomes of bacteria and archaea. This review will highlight many surprising examples of the recently revealed connections between prokaryotic and eukaryotic antiviral immune systems.


Asunto(s)
Bacterias , Bacteriófagos , Animales , Bacterias/genética , Células Procariotas , Archaea/genética , Inmunidad Innata , Bacteriófagos/genética
6.
bioRxiv ; 2023 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-36865129

RESUMEN

cGAS (cyclic GMP-AMP synthase) is an enzyme in human cells that controls an immune response to cytosolic DNA. Upon binding DNA, cGAS synthesizes a nucleotide signal 2'3'-cGAMP that activates the protein STING and downstream immunity. Here we discover cGAS-like receptors (cGLRs) constitute a major family of pattern recognition receptors in animal innate immunity. Building on recent analysis in Drosophila , we use a bioinformatic approach to identify >3,000 cGLRs present in nearly all metazoan phyla. A forward biochemical screen of 140 animal cGLRs reveals a conserved mechanism of signaling including response to dsDNA and dsRNA ligands and synthesis of alternative nucleotide signals including isomers of cGAMP and cUMP-AMP. Using structural biology, we explain how synthesis of distinct nucleotide signals enables cells to control discrete cGLR-STING signaling pathways. Together our results reveal cGLRs as a widespread family of pattern recognition receptors and establish molecular rules that govern nucleotide signaling in animal immunity.

7.
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
8.
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
9.
Nature ; 605(7910): 522-526, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35395152

RESUMEN

The cyclic oligonucleotide-based antiphage signalling system (CBASS) and the pyrimidine cyclase system for antiphage resistance (Pycsar) are antiphage defence systems in diverse bacteria that use cyclic nucleotide signals to induce cell death and prevent viral propagation1,2. Phages use several strategies to defeat host CRISPR and restriction-modification systems3-10, but no mechanisms are known to evade CBASS and Pycsar immunity. Here we show that phages encode anti-CBASS (Acb) and anti-Pycsar (Apyc) proteins that counteract defence by specifically degrading cyclic nucleotide signals that activate host immunity. Using a biochemical screen of 57 phages in Escherichia coli and Bacillus subtilis, we discover Acb1 from phage T4 and Apyc1 from phage SBSphiJ as founding members of distinct families of immune evasion proteins. Crystal structures of Acb1 in complex with 3'3'-cyclic GMP-AMP define a mechanism of metal-independent hydrolysis 3' of adenosine bases, enabling broad recognition and degradation of cyclic dinucleotide and trinucleotide CBASS signals. Structures of Apyc1 reveal a metal-dependent cyclic NMP phosphodiesterase that uses relaxed specificity to target Pycsar cyclic pyrimidine mononucleotide signals. We show that Acb1 and Apyc1 block downstream effector activation and protect from CBASS and Pycsar defence in vivo. Active Acb1 and Apyc1 enzymes are conserved in phylogenetically diverse phages, demonstrating that cleavage of host cyclic nucleotide signals is a key strategy of immune evasion in phage biology.


Asunto(s)
Bacteriófagos , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Bacteriófago T4/metabolismo , Bacteriófagos/fisiología , Sistemas CRISPR-Cas/genética , Endonucleasas/metabolismo , Escherichia coli/metabolismo , Nucleótidos Cíclicos/metabolismo , Oligonucleótidos , Pirimidinas/metabolismo
10.
Cell ; 184(23): 5728-5739.e16, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34644530

RESUMEN

The cyclic pyrimidines 3',5'-cyclic cytidine monophosphate (cCMP) and 3',5'-cyclic uridine monophosphate (cUMP) have been reported in multiple organisms and cell types. As opposed to the cyclic nucleotides 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP), which are second messenger molecules with well-established regulatory roles across all domains of life, the biological role of cyclic pyrimidines has remained unclear. Here we report that cCMP and cUMP are second messengers functioning in bacterial immunity against viruses. We discovered a family of bacterial pyrimidine cyclase enzymes that specifically synthesize cCMP and cUMP following phage infection and demonstrate that these molecules activate immune effectors that execute an antiviral response. A crystal structure of a uridylate cyclase enzyme from this family explains the molecular mechanism of selectivity for pyrimidines as cyclization substrates. Defense systems encoding pyrimidine cyclases, denoted here Pycsar (pyrimidine cyclase system for antiphage resistance), are widespread in prokaryotes. Our results assign clear biological function to cCMP and cUMP as immunity signaling molecules in bacteria.


Asunto(s)
Bacterias/inmunología , Bacterias/virología , Bacteriófagos/fisiología , CMP Cíclico/metabolismo , Nucleótidos Cíclicos/metabolismo , Uridina Monofosfato/metabolismo , Secuencia de Aminoácidos , Bacterias/genética , Burkholderia/enzimología , CMP Cíclico/química , Ciclización , Escherichia coli/enzimología , Modelos Moleculares , Mutación/genética , Nucleótidos Cíclicos/química , Liasas de Fósforo-Oxígeno/química , Liasas de Fósforo-Oxígeno/metabolismo , Pirimidinas/metabolismo , Uridina Monofosfato/química
11.
Nature ; 597(7874): 109-113, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34261127

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that produces the second messenger cG[2'-5']pA[3'-5']p (2'3'-cGAMP) and controls activation of innate immunity in mammalian cells1-5. Animal genomes typically encode multiple proteins with predicted homology to cGAS6-10, but the function of these uncharacterized enzymes is unknown. Here we show that cGAS-like receptors (cGLRs) are innate immune sensors that are capable of recognizing divergent molecular patterns and catalysing synthesis of distinct nucleotide second messenger signals. Crystal structures of human and insect cGLRs reveal a nucleotidyltransferase signalling core shared with cGAS and a diversified primary ligand-binding surface modified with notable insertions and deletions. We demonstrate that surface remodelling of cGLRs enables altered ligand specificity and used a forward biochemical screen to identify cGLR1 as a double-stranded RNA sensor in the model organism Drosophila melanogaster. We show that RNA recognition activates Drosophila cGLR1 to synthesize the novel product cG[3'-5']pA[2'-5']p (3'2'-cGAMP). A crystal structure of Drosophila stimulator of interferon genes (dSTING) in complex with 3'2'-cGAMP explains selective isomer recognition, and 3'2'-cGAMP induces an enhanced antiviral state in vivo that protects from viral infection. Similar to radiation of Toll-like receptors in pathogen immunity, our results establish cGLRs as a diverse family of metazoan pattern recognition receptors.


Asunto(s)
Drosophila melanogaster/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , ARN Bicatenario/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Sistemas de Mensajero Secundario , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/virología , Femenino , Humanos , Inmunidad Innata , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Nucleotidiltransferasas/química , Nucleotidiltransferasas/inmunología , ARN Bicatenario/análisis , ARN Bicatenario/inmunología , Receptores de Reconocimiento de Patrones/química , Receptores de Reconocimiento de Patrones/inmunología , Virus/inmunología
12.
Nature ; 586(7829): 429-433, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32877915

RESUMEN

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.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , GMP Cíclico/análogos & derivados , Evolución Molecular , Proteínas de la Membrana , Sistemas de Mensajero Secundario , Animales , Bacterias/química , Bacterias/virología , Proteínas Bacterianas/química , Bacteriófagos , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Proteínas de la Membrana/química , Modelos Moleculares , NAD/metabolismo , Nucleotidiltransferasas/metabolismo
13.
Cell ; 182(1): 38-49.e17, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32544385

RESUMEN

cGAS/DncV-like nucleotidyltransferase (CD-NTase) enzymes are immune sensors that synthesize nucleotide second messengers and initiate antiviral responses in bacterial and animal cells. Here, we discover Enterobacter cloacae CD-NTase-associated protein 4 (Cap4) as a founding member of a diverse family of >2,000 bacterial receptors that respond to CD-NTase signals. Structures of Cap4 reveal a promiscuous DNA endonuclease domain activated through ligand-induced oligomerization. Oligonucleotide recognition occurs through an appended SAVED domain that is an unexpected fusion of two CRISPR-associated Rossman fold (CARF) subunits co-opted from type III CRISPR immunity. Like a lock and key, SAVED effectors exquisitely discriminate 2'-5'- and 3'-5'-linked bacterial cyclic oligonucleotide signals and enable specific recognition of at least 180 potential nucleotide second messenger species. Our results reveal SAVED CARF family proteins as major nucleotide second messenger receptors in CBASS and CRISPR immune defense and extend the importance of linkage specificity beyond mammalian cGAS-STING signaling.


Asunto(s)
Bacterias/virología , Bacteriófagos/metabolismo , Sistemas CRISPR-Cas , Inmunidad , Oligonucleótidos/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Desoxirribonucleasa I/metabolismo , Ligandos , Mutagénesis/genética , Nucleotidiltransferasas/metabolismo , Unión Proteica , Sistemas de Mensajero Secundario
14.
ACS Chem Biol ; 14(9): 2035-2043, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31433159

RESUMEN

Linalyl diphosphate (LPP) is the postulated intermediate in the enzymatic cyclization of monoterpenes catalyzed by terpene synthases. LPP is considered an obligate intermediate due to the conformationally restrictive trans-C2-C3 double bond of the substrate, geranyl diphosphate (GPP), which precludes the proper positioning of carbons C1 and C6 to enable cyclization. However, because of the complexity of potential carbocation-mediated rearrangements in these enzymatic reactions, it has proven difficult to directly demonstrate the formation of LPP despite significant efforts. Here we synthesized a fluorinated substrate analog, 8,9-difluorogeranyl diphosphate (DFGPP), which is designed to allow initial ionization/isomerization and form the fluorinated equivalent of LPP (DFLPP) while preventing the subsequent ionization/cyclization to produce the α-terpinyl cation. Steady-state kinetic studies with the model enzyme (+)-limonene synthase (LS) under catalytic conditions show that the cyclization of DFGPP is completely blocked and a single linear product, difluoromyrcene, is produced. When crystals of apo-LS are soaked with DFGPP under conditions limiting turnover of the enzyme, we show, using X-ray crystallography, that DFLPP is produced in the enzyme active site and trapped in the crystals. Clear electron density is observed in the active site of the enzyme, but it cannot be appropriately fit with a model for the DFGPP substrate analog, whereas it can accommodate an extended conformation of DFLPP. This result supports the current model for monoterpene cyclization by providing direct evidence of LPP as an intermediate.


Asunto(s)
Monoterpenos Acíclicos/química , Difosfatos/química , Diterpenos/química , Inhibidores Enzimáticos/química , Liasas Intramoleculares/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/química , Dominio Catalítico , Citrus sinensis/enzimología , Cristalografía por Rayos X , Difosfatos/síntesis química , Diterpenos/síntesis química , Pruebas de Enzimas , Inhibidores Enzimáticos/síntesis química , Liasas Intramoleculares/química
15.
Nature ; 567(7747): 194-199, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30787435

RESUMEN

Cyclic dinucleotides (CDNs) have central roles in bacterial homeostasis and virulence by acting as nucleotide second messengers. Bacterial CDNs also elicit immune responses during infection when they are detected by pattern-recognition receptors in animal cells. Here we perform a systematic biochemical screen for bacterial signalling nucleotides and discover a large family of cGAS/DncV-like nucleotidyltransferases (CD-NTases) that use both purine and pyrimidine nucleotides to synthesize a diverse range of CDNs. A series of crystal structures establish CD-NTases as a structurally conserved family and reveal key contacts in the enzyme active-site lid that direct purine or pyrimidine selection. CD-NTase products are not restricted to CDNs and also include an unexpected class of cyclic trinucleotide compounds. Biochemical and cellular analyses of CD-NTase signalling nucleotides demonstrate that these cyclic di- and trinucleotides activate distinct host receptors and thus may modulate the interaction of both pathogens and commensal microbiota with their animal and plant hosts.


Asunto(s)
Proteínas Bacterianas/metabolismo , Nucleótidos/biosíntesis , Nucleótidos/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Animales , Cristalografía por Rayos X , Fosfatos de Dinucleósidos/biosíntesis , Fosfatos de Dinucleósidos/metabolismo , Células HEK293 , Humanos , Ratones , Nucleótidos/química , Nucleotidiltransferasas/genética , Operón/genética , Simbiosis
16.
Cell ; 174(2): 300-311.e11, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-30007416

RESUMEN

Cyclic GMP-AMP synthase (cGAS) recognition of cytosolic DNA is critical for immune responses to pathogen replication, cellular stress, and cancer. Existing structures of the mouse cGAS-DNA complex provide a model for enzyme activation but do not explain why human cGAS exhibits severely reduced levels of cyclic GMP-AMP (cGAMP) synthesis compared to other mammals. Here, we discover that enhanced DNA-length specificity restrains human cGAS activation. Using reconstitution of cGAMP signaling in bacteria, we mapped the determinant of human cGAS regulation to two amino acid substitutions in the DNA-binding surface. Human-specific substitutions are necessary and sufficient to direct preferential detection of long DNA. Crystal structures reveal why removal of human substitutions relaxes DNA-length specificity and explain how human-specific DNA interactions favor cGAS oligomerization. These results define how DNA-sensing in humans adapted for enhanced specificity and provide a model of the active human cGAS-DNA complex to enable structure-guided design of cGAS therapeutics.


Asunto(s)
ADN/metabolismo , Vigilancia Inmunológica/fisiología , Nucleotidiltransferasas/metabolismo , Animales , Benzofuranos/química , Benzofuranos/metabolismo , Sitios de Unión , Dominio Catalítico , Quimiotaxis/efectos de los fármacos , ADN/química , Humanos , Ratones , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Nucleótidos Cíclicos/metabolismo , Nucleótidos Cíclicos/farmacología , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/genética , Multimerización de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Especificidad de la Especie , Vibrio cholerae/metabolismo , Vibrio cholerae/fisiología
17.
J Biol Chem ; 292(52): 21578-21589, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29118188

RESUMEN

RhoGC is a fusion protein from the aquatic fungus Blastocladiella emersonii, combining a type I rhodopsin domain with a guanylyl cyclase domain. It has generated excitement as an optogenetics tool for the manipulation of cyclic nucleotide signaling pathways. To investigate the regulation of the cyclase activity, we isolated the guanylyl cyclase domain from Escherichia coli with (GCwCCRho) and without (GCRho) the coiled-coil linker. Both constructs were constitutively active but were monomeric as determined by size-exclusion chromatography and analytical ultracentrifugation, whereas other class III nucleotidyl cyclases are functional dimers. We also observed that crystals of GCRho have only a monomer in an asymmetric unit. Dimers formed when crystals were grown in the presence of the non-cyclizable substrate analog 2',3'-dideoxyguanosine-5'-triphosphate, MnCl2, and tartrate, but their quaternary structure did not conform to the canonical pairing expected for class III enzymes. Moreover, the structure contained a disulfide bond formed with an active-site Cys residue required for activity. We consider it unlikely that the disulfide would form under intracellular reducing conditions, raising the possibility that this unusual dimer might have a biologically relevant role in the regulation of full-length RhoGC. Although we did not observe it with direct methods, a functional dimer was identified as the active state by following the dependence of activity on total enzyme concentration. The low affinity observed for GCRho monomers is unusual for this enzyme class and suggests that dimer formation may contribute to light activation of the full-length protein.


Asunto(s)
Guanilato Ciclasa/metabolismo , Optogenética/métodos , Rodopsina/metabolismo , Secuencia de Aminoácidos , Blastocladiella/metabolismo , Dominio Catalítico , GMP Cíclico/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Nucleótidos Cíclicos/metabolismo , Dominios Proteicos , Transducción de Señal/fisiología
18.
Biochemistry ; 56(12): 1706-1715, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28272875

RESUMEN

Terpenes make up the largest and most diverse class of natural compounds and have important commercial and medical applications. Limonene is a cyclic monoterpene (C10) present in nature as two enantiomers, (+) and (-), which are produced by different enzymes. The mechanism of production of the (-)-enantiomer has been studied in great detail, but to understand how enantiomeric selectivity is achieved in this class of enzymes, it is important to develop a thorough biochemical description of enzymes that generate (+)-limonene, as well. Here we report the first cloning and biochemical characterization of a (+)-limonene synthase from navel orange (Citrus sinensis). The enzyme obeys classical Michaelis-Menten kinetics and produces exclusively the (+)-enantiomer. We have determined the crystal structure of the apoprotein in an "open" conformation at 2.3 Å resolution. Comparison with the structure of (-)-limonene synthase (Mentha spicata), which is representative of a fully closed conformation (Protein Data Bank entry 2ONG ), reveals that the short H-α1 helix moves nearly 5 Å inward upon substrate binding, and a conserved Tyr flips to point its hydroxyl group into the active site.


Asunto(s)
Apoproteínas/química , Citrus sinensis/química , Ciclohexenos/química , Liasas Intramoleculares/química , Proteínas de Plantas/química , Proteínas Recombinantes de Fusión/química , Terpenos/química , Apoproteínas/genética , Apoproteínas/metabolismo , Dominio Catalítico , Citrus sinensis/enzimología , Clonación Molecular , Cristalografía por Rayos X , Ciclohexenos/metabolismo , Difosfatos/química , Difosfatos/metabolismo , Diterpenos/química , Diterpenos/metabolismo , Pruebas de Enzimas , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Liasas Intramoleculares/genética , Liasas Intramoleculares/metabolismo , Cinética , Limoneno , Mentha spicata/química , Mentha spicata/enzimología , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Estereoisomerismo , Terpenos/metabolismo
19.
Biochemistry ; 56(12): 1716-1725, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28272876

RESUMEN

The stereochemical course of monoterpene synthase reactions is thought to be determined early in the reaction sequence by selective binding of distinct conformations of the geranyl diphosphate (GPP) substrate. We explore here formation of early Michaelis complexes of the (+)-limonene synthase [(+)-LS] from Citrus sinensis using monofluorinated substrate analogues 2-fluoro-GPP (FGPP) and 2-fluoroneryl diphosphate (FNPP). Both are competitive inhibitors for (+)-LS with KI values of 2.4 ± 0.5 and 39.5 ± 5.2 µM, respectively. The KI values are similar to the KM for the respective nonfluorinated substrates, indicating that fluorine does not significantly perturb binding of the ligand to the enzyme. FGPP and FNPP are also substrates, but with dramatically reduced rates (kcat values of 0.00054 ± 0.00005 and 0.00024 ± 0.00002 s-1, respectively). These data are consistent with a stepwise mechanism for (+)-LS involving ionization of the allylic GPP substrate to generate a resonance-stabilized carbenium ion in the rate-limiting step. Crystals of apo-(+)-LS were soaked with FGPP and FNPP to obtain X-ray structures at 2.4 and 2.2 Å resolution, respectively. The fluorinated analogues are found anchored in the active site through extensive interactions involving the diphosphate, three metal ions, and three active-site Asp residues. Electron density for the carbon chains extends deep into a hydrophobic pocket, while the enzyme remains mostly in the open conformation observed for the apoprotein. While FNPP was found in multiple conformations, FGPP, importantly, was in a single, relatively well-defined, left-handed screw conformation, consistent with predictions for the mechanism of stereoselectivity in the monoterpene synthases.


Asunto(s)
Apoproteínas/química , Citrus sinensis/química , Ciclohexenos/química , Diterpenos/química , Inhibidores Enzimáticos/química , Liasas Intramoleculares/química , Organofosfatos/química , Proteínas de Plantas/química , Terpenos/química , Apoproteínas/antagonistas & inhibidores , Apoproteínas/genética , Apoproteínas/metabolismo , Dominio Catalítico , Citrus sinensis/enzimología , Clonación Molecular , Cristalografía por Rayos X , Ciclohexenos/metabolismo , Diterpenos/metabolismo , Pruebas de Enzimas , Inhibidores Enzimáticos/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Liasas Intramoleculares/antagonistas & inhibidores , Liasas Intramoleculares/genética , Liasas Intramoleculares/metabolismo , Cinética , Ligandos , Limoneno , Modelos Moleculares , Organofosfatos/metabolismo , Proteínas de Plantas/antagonistas & inhibidores , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Fosfatos de Poliisoprenilo/química , Fosfatos de Poliisoprenilo/metabolismo , Dominios Proteicos , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Estereoisomerismo , Terpenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...