Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.680
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 178(2): 290-301.e10, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31230712

RESUMEN

How the central innate immune protein, STING, is activated by its ligands remains unknown. Here, using structural biology and biochemistry, we report that the metazoan second messenger 2'3'-cGAMP induces closing of the human STING homodimer and release of the STING C-terminal tail, which exposes a polymerization interface on the STING dimer and leads to the formation of disulfide-linked polymers via cysteine residue 148. Disease-causing hyperactive STING mutations either flank C148 and depend on disulfide formation or reside in the C-terminal tail binding site and cause constitutive C-terminal tail release and polymerization. Finally, bacterial cyclic-di-GMP induces an alternative active STING conformation, activates STING in a cooperative manner, and acts as a partial antagonist of 2'3'-cGAMP signaling. Our insights explain the tight control of STING signaling given varying background activation signals and provide a therapeutic hypothesis for autoimmune syndrome treatment.


Asunto(s)
Proteínas de la Membrana/metabolismo , Sitios de Unión , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Dimerización , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Ligandos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Nucleótidos Cíclicos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Transducción de Señal
2.
Nat Rev Mol Cell Biol ; 21(9): 501-521, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32424334

RESUMEN

The cGAS-STING signalling axis, comprising the synthase for the second messenger cyclic GMP-AMP (cGAS) and the cyclic GMP-AMP receptor stimulator of interferon genes (STING), detects pathogenic DNA to trigger an innate immune reaction involving a strong type I interferon response against microbial infections. Notably however, besides sensing microbial DNA, the DNA sensor cGAS can also be activated by endogenous DNA, including extranuclear chromatin resulting from genotoxic stress and DNA released from mitochondria, placing cGAS-STING as an important axis in autoimmunity, sterile inflammatory responses and cellular senescence. Initial models assumed that co-localization of cGAS and DNA in the cytosol defines the specificity of the pathway for non-self, but recent work revealed that cGAS is also present in the nucleus and at the plasma membrane, and such subcellular compartmentalization was linked to signalling specificity of cGAS. Further confounding the simple view of cGAS-STING signalling as a response mechanism to infectious agents, both cGAS and STING were shown to have additional functions, independent of interferon response. These involve non-catalytic roles of cGAS in regulating DNA repair and signalling via STING to NF-κB and MAPK as well as STING-mediated induction of autophagy and lysosome-dependent cell death. We have also learnt that cGAS dimers can multimerize and undergo liquid-liquid phase separation to form biomolecular condensates that could importantly regulate cGAS activation. Here, we review the molecular mechanisms and cellular functions underlying cGAS-STING activation and signalling, particularly highlighting the newly emerging diversity of this signalling pathway and discussing how the specificity towards normal, damage-induced and infection-associated DNA could be achieved.


Asunto(s)
Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Animales , Autofagia , AMP Cíclico/metabolismo , AMP Cíclico/fisiología , GMP Cíclico/metabolismo , GMP Cíclico/fisiología , Citosol/metabolismo , ADN/metabolismo , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Proteínas de la Membrana/fisiología , Nucleótidos Cíclicos , Nucleotidiltransferasas/genética , Transducción de Señal
3.
Cell ; 165(4): 936-48, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27062922

RESUMEN

Neurons receive input from the outside world or from other neurons through neuronal receptive endings (NREs). Glia envelop NREs to create specialized microenvironments; however, glial functions at these sites are poorly understood. Here, we report a molecular mechanism by which glia control NRE shape and associated animal behavior. The C. elegans AMsh glial cell ensheathes the NREs of 12 neurons, including the thermosensory neuron AFD. KCC-3, a K/Cl transporter, localizes specifically to a glial microdomain surrounding AFD receptive ending microvilli, where it regulates K(+) and Cl(-) levels. We find that Cl(-) ions function as direct inhibitors of an NRE-localized receptor-guanylyl-cyclase, GCY-8, which synthesizes cyclic guanosine monophosphate (cGMP). High cGMP mediates the effects of glial KCC-3 on AFD shape by antagonizing the actin regulator WSP-1/NWASP. Components of this pathway are broadly expressed throughout the nervous system, suggesting that ionic regulation of the NRE microenvironment may be a conserved mechanism by which glia control neuron shape and function.


Asunto(s)
Caenorhabditis elegans/metabolismo , Neuroglía/metabolismo , Células Receptoras Sensoriales/metabolismo , Simportadores/metabolismo , Animales , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , GMP Cíclico/metabolismo , Guanilato Ciclasa/química , Guanilato Ciclasa/metabolismo , Potasio/metabolismo , Dominios Proteicos , Simportadores/química , Simportadores/genética , Sensación Térmica , Cotransportadores de K Cl
4.
Cell ; 158(5): 1136-1147, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171413

RESUMEN

The cyclic dinucleotide c-di-GMP is a signaling molecule with diverse functions in cellular physiology. Here, we report that c-di-GMP can assemble into a tetramer that mediates the effective dimerization of a transcription factor, BldD, which controls the progression of multicellular differentiation in sporulating actinomycete bacteria. BldD represses expression of sporulation genes during vegetative growth in a manner that depends on c-di-GMP-mediated dimerization. Structural and biochemical analyses show that tetrameric c-di-GMP links two subunits of BldD through their C-terminal domains, which are otherwise separated by ~10 Å and thus cannot effect dimerization directly. Binding of the c-di-GMP tetramer by BldD is selective and requires a bipartite RXD-X8-RXXD signature. The findings indicate a unique mechanism of protein dimerization and the ability of nucleotide signaling molecules to assume alternative oligomeric states to effect different functions.


Asunto(s)
Proteínas Bacterianas/metabolismo , GMP Cíclico/análogos & derivados , Streptomyces/crecimiento & desarrollo , Streptomyces/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Dimerización , Modelos Moleculares , Datos de Secuencia Molecular , Alineación de Secuencia , Esporas Bacterianas/metabolismo , Streptomyces/citología , Factores de Transcripción/química
5.
Nature ; 624(7991): 425-432, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38057665

RESUMEN

Maintenance of renal function and fluid transport are essential for vertebrates and invertebrates to adapt to physiological and pathological challenges. Human patients with malignant tumours frequently develop detrimental renal dysfunction and oliguria, and previous studies suggest the involvement of chemotherapeutic toxicity and tumour-associated inflammation1,2. However, how tumours might directly modulate renal functions remains largely unclear. Here, using conserved tumour models in Drosophila melanogaster3, we characterized isoform F of ion transport peptide (ITPF) as a fly antidiuretic hormone that is secreted by a subset of yki3SA gut tumour cells, impairs renal function and causes severe abdomen bloating and fluid accumulation. Mechanistically, tumour-derived ITPF targets the G-protein-coupled receptor TkR99D in stellate cells of Malpighian tubules-an excretory organ that is equivalent to renal tubules4-to activate nitric oxide synthase-cGMP signalling and inhibit fluid excretion. We further uncovered antidiuretic functions of mammalian neurokinin 3 receptor (NK3R), the homologue of fly TkR99D, as pharmaceutical blockade of NK3R efficiently alleviates renal tubular dysfunction in mice bearing different malignant tumours. Together, our results demonstrate a novel antidiuretic pathway mediating tumour-renal crosstalk across species and offer therapeutic opportunities for the treatment of cancer-associated renal dysfunction.


Asunto(s)
Fármacos Antidiuréticos , Enfermedades Renales , Neoplasias , Neuropéptidos , Receptores de Neuroquinina-3 , Animales , Humanos , Ratones , Fármacos Antidiuréticos/metabolismo , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Enfermedades Renales/complicaciones , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Túbulos de Malpighi/citología , Túbulos de Malpighi/metabolismo , Neoplasias/complicaciones , Neoplasias/metabolismo , Óxido Nítrico Sintasa/metabolismo , Receptores de Neuroquinina-3/antagonistas & inhibidores , Receptores de Neuroquinina-3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Arginina Vasopresina/metabolismo , Proteínas de Drosophila/metabolismo , Neuropéptidos/metabolismo
6.
Cell ; 154(4): 748-62, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23910378

RESUMEN

Binding of dsDNA by cyclic GMP-AMP (cGAMP) synthase (cGAS) triggers formation of the metazoan second messenger c[G(2',5')pA(3',5')p], which binds the signaling protein STING with subsequent activation of the interferon (IFN) pathway. We show that human hSTING(H232) adopts a "closed" conformation upon binding c[G(2',5')pA(3',5')p] and its linkage isomer c[G(2',5')pA(2',5')p], as does mouse mSting(R231) on binding c[G(2',5')pA(3',5')p], c[G(3',5')pA(3',5')p] and the antiviral agent DMXAA, leading to similar "closed" conformations. Comparing hSTING to mSting, 2',5'-linkage-containing cGAMP isomers were more specific triggers of the IFN pathway compared to the all-3',5'-linkage isomer. Guided by structural information, we identified a unique point mutation (S162A) placed within the cyclic-dinucleotide-binding site of hSTING that rendered it sensitive to the otherwise mouse-specific drug DMXAA, a conclusion validated by binding studies. Our structural and functional analysis highlights the unexpected versatility of STING in the recognition of natural and synthetic ligands within a small-molecule pocket created by the dimerization of STING.


Asunto(s)
Antivirales/farmacología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Nucleótidos Cíclicos/metabolismo , Xantonas/farmacología , Animales , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Modelos Moleculares , Mutagénesis , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad
7.
Mol Cell ; 77(3): 586-599.e6, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31810759

RESUMEN

Streptomyces are our primary source of antibiotics, produced concomitantly with the transition from vegetative growth to sporulation in a complex developmental life cycle. We previously showed that the signaling molecule c-di-GMP binds BldD, a master repressor, to control initiation of development. Here we demonstrate that c-di-GMP also intervenes later in development to control differentiation of the reproductive hyphae into spores by arming a novel anti-σ (RsiG) to bind and sequester a sporulation-specific σ factor (σWhiG). We present the structure of the RsiG-(c-di-GMP)2-σWhiG complex, revealing an unusual, partially intercalated c-di-GMP dimer bound at the RsiG-σWhiG interface. RsiG binds c-di-GMP in the absence of σWhiG, employing a novel E(X)3S(X)2R(X)3Q(X)3D motif repeated on each helix of a coiled coil. Further studies demonstrate that c-di-GMP is essential for RsiG to inhibit σWhiG. These findings reveal a newly described control mechanism for σ-anti-σ complex formation and establish c-di-GMP as the central integrator of Streptomyces development.


Asunto(s)
GMP Cíclico/análogos & derivados , Factor sigma/metabolismo , Streptomyces/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , GMP Cíclico/metabolismo , GMP Cíclico/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Dominios Proteicos , ARN Bacteriano/metabolismo , Esporas Bacterianas/metabolismo , Streptomyces/genética
8.
Annu Rev Biochem ; 81: 533-59, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22404633

RESUMEN

Nitric oxide (NO) is an essential signaling molecule in biological systems. In mammals, the diatomic gas is critical to the cyclic guanosine monophosphate (cGMP) pathway as it functions as the primary activator of soluble guanylate cyclase (sGC). NO is synthesized from l-arginine and oxygen (O(2)) by the enzyme nitric oxide synthase (NOS). Once produced, NO rapidly diffuses across cell membranes and binds to the heme cofactor of sGC. sGC forms a stable complex with NO and carbon monoxide (CO), but not with O(2). The binding of NO to sGC leads to significant increases in cGMP levels. The second messenger then directly modulates phosphodiesterases (PDEs), ion-gated channels, or cGMP-dependent protein kinases to regulate physiological functions, including vasodilation, platelet aggregation, and neurotransmission. Many studies are focused on elucidating the molecular mechanism of sGC activation and deactivation with a goal of therapeutic intervention in diseases involving the NO/cGMP-signaling pathway. This review summarizes the current understanding of sGC structure and regulation as well as recent developments in NO signaling.


Asunto(s)
Guanilato Ciclasa/química , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Animales , GMP Cíclico/metabolismo , Guanilato Ciclasa/aislamiento & purificación , Guanilato Ciclasa/metabolismo , Humanos , Isoenzimas/metabolismo , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Guanilil Ciclasa Soluble
9.
EMBO J ; 42(12): e114091, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37051729

RESUMEN

Cyclic di-GMP signaling regulates sessile-to-motile lifestyle transition and associated physiological and metabolic features in bacteria. The presence of potential cyclic di-GMP turnover proteins in deepest branching bacteria indicates that cyclic di-GMP is an ancient signaling molecule. In this issue of The EMBO Journal, Cai et al (2023) describe light-induced activation of a thiosulfate oxidation pathway in the deep-sea cold-seep bacterium Qipengyuania flava, thus coupling cyclic di-GMP with the regulation of the global abiotic sulfur cycle.


Asunto(s)
Proteínas Bacterianas , Proteínas de Escherichia coli , Proteínas Bacterianas/metabolismo , GMP Cíclico/metabolismo , Transducción de Señal , Proteínas de Escherichia coli/metabolismo , Bacterias/metabolismo , Comunicación Celular , Regulación Bacteriana de la Expresión Génica , Biopelículas
10.
Annu Rev Microbiol ; 76: 503-532, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35671532

RESUMEN

Biofilms are a widely observed growth mode in which microbial communities are spatially structured and embedded in a polymeric extracellular matrix. Here, we focus on the model bacterium Vibrio cholerae and summarize the current understanding of biofilm formation, including initial attachment, matrix components, community dynamics, social interactions, molecular regulation, and dispersal. The regulatory network that orchestrates the decision to form and disperse from biofilms coordinates various environmental inputs. These cues are integrated by several transcription factors, regulatory RNAs, and second-messenger molecules, including bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP). Through complex mechanisms, V. cholerae weighs the energetic cost of forming biofilms against the benefits of protection and social interaction that biofilms provide.


Asunto(s)
Biopelículas , Vibrio cholerae , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , GMP Cíclico/metabolismo , Regulación Bacteriana de la Expresión Génica , Factores de Transcripción/metabolismo , Vibrio cholerae/genética , Vibrio cholerae/fisiología
11.
Proc Natl Acad Sci U S A ; 121(18): e2313107121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652742

RESUMEN

Full understanding of proteostasis and energy utilization in cells will require knowledge of the fraction of cell proteins being degraded with different half-lives and their rates of synthesis. We therefore developed a method to determine such information that combines mathematical analysis of protein degradation kinetics obtained in pulse-chase experiments with Bayesian data fitting using the maximum entropy principle. This approach will enable rapid analyses of whole-cell protein dynamics in different cell types, physiological states, and neurodegenerative disease. Using it, we obtained surprising insights about protein stabilities in cultured cells normally and upon activation of proteolysis by mTOR inhibition and increasing cAMP or cGMP. It revealed that >90% of protein content in dividing mammalian cell lines is long-lived, with half-lives of 24 to 200 h, and therefore comprises much of the proteins in daughter cells. The well-studied short-lived proteins (half-lives < 10 h) together comprise <2% of cell protein mass, but surprisingly account for 10 to 20% of measurable newly synthesized protein mass. Evolution thus appears to have minimized intracellular proteolysis except to rapidly eliminate misfolded and regulatory proteins.


Asunto(s)
Entropía , Proteolisis , Proteoma , Proteoma/metabolismo , Humanos , Animales , Teorema de Bayes , Proteostasis , Cinética , AMP Cíclico/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , GMP Cíclico/metabolismo
12.
Proc Natl Acad Sci U S A ; 121(13): e2320410121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38498718

RESUMEN

Biofilms of sulfate-reducing bacterium (SRB) like Desulfovibrio vulgaris Hildenborough (DvH) can facilitate metal corrosion in various industrial and environmental settings leading to substantial economic losses. Although the mechanisms of biofilm formation by DvH are not yet well understood, recent studies indicate the large adhesin, DvhA, is a key determinant of biofilm formation. The dvhA gene neighborhood resembles the biofilm-regulating Lap system of Pseudomonas fluorescens but is curiously missing the c-di-GMP-binding regulator LapD. Instead, DvH encodes an evolutionarily unrelated c-di-GMP-binding protein (DVU1020) that we hypothesized is functionally analogous to LapD. To study this unusual Lap system and overcome experimental limitations with the slow-growing anaerobe DvH, we reconstituted its predicted SRB Lap system in a P. fluorescens strain lacking its native Lap regulatory components (ΔlapGΔlapD). Our data support the model that DvhA is a cell surface-associated LapA-like adhesin with a N-terminal "retention module" and that DvhA is released from the cell surface upon cleavage by the LapG-like protease DvhG. Further, we demonstrate DVU1020 (named here DvhD) represents a distinct class of c-di-GMP-binding, biofilm-regulating proteins that regulates DvhG activity in response to intracellular levels of this second messenger. This study provides insight into the key players responsible for biofilm formation by DvH, thereby expanding our understanding of Lap-like systems.


Asunto(s)
Pseudomonas fluorescens , Pseudomonas fluorescens/genética , Pseudomonas fluorescens/metabolismo , Sulfatos/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Biopelículas , Proteínas Portadoras/metabolismo , GMP Cíclico/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica
13.
Proc Natl Acad Sci U S A ; 121(39): e2411981121, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39284057

RESUMEN

Bacterial biofilms have been implicated in several chronic infections. After initial attachment, a critical first step in biofilm formation is a cell inducing a surface-sensing response. In the Gram-negative opportunistic pathogen Pseudomonas aeruginosa, two second messengers, cyclic diguanylate monophosphate (c-di-GMP) and cyclic adenosine monophosphate (cAMP), are produced by different surface-sensing mechanisms. However, given the disparate cellular behaviors regulated by these second messengers, how newly attached cells coordinate these pathways remains unclear. Some of the uncertainty relates to studies using different strains, experimental systems, and usually focusing on a single second messenger. In this study, we developed a tricolor reporter system to simultaneously gauge c-di-GMP and cAMP levels in single cells. Using PAO1, we show that c-di-GMP and cAMP are selectively activated in two commonly used experimental systems to study surface sensing. By further examining the conditions that differentiate a c-di-GMP or cAMP response, we demonstrate that an agarose-air interface activates cAMP signaling through type IV pili and the Pil-Chp system. However, a liquid-agarose interface favors the activation of c-di-GMP signaling. This response is dependent on flagellar motility and correlated with higher swimming speed. Collectively, this work indicates that c-di-GMP and cAMP signaling responses are dependent on the surface context.


Asunto(s)
Biopelículas , AMP Cíclico , GMP Cíclico , Pseudomonas aeruginosa , Pseudomonas aeruginosa/fisiología , Pseudomonas aeruginosa/metabolismo , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Biopelículas/crecimiento & desarrollo , Transducción de Señal , Sistemas de Mensajero Secundario/fisiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
14.
Proc Natl Acad Sci U S A ; 121(25): e2319903121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38870058

RESUMEN

Biofilm formation and surface attachment in multiple Alphaproteobacteria is driven by unipolar polysaccharide (UPP) adhesins. The pathogen Agrobacterium tumefaciens produces a UPP adhesin, which is regulated by the intracellular second messenger cyclic diguanylate monophosphate (c-di-GMP). Prior studies revealed that DcpA, a diguanylate cyclase-phosphodiesterase, is crucial in control of UPP production and surface attachment. DcpA is regulated by PruR, a protein with distant similarity to enzymatic domains known to coordinate the molybdopterin cofactor (MoCo). Pterins are bicyclic nitrogen-rich compounds, several of which are produced via a nonessential branch of the folate biosynthesis pathway, distinct from MoCo. The pterin-binding protein PruR controls DcpA activity, fostering c-di-GMP breakdown and dampening its synthesis. Pterins are excreted, and we report here that PruR associates with these metabolites in the periplasm, promoting interaction with the DcpA periplasmic domain. The pteridine reductase PruA, which reduces specific dihydro-pterin molecules to their tetrahydro forms, imparts control over DcpA activity through PruR. Tetrahydromonapterin preferentially associates with PruR relative to other related pterins, and the PruR-DcpA interaction is decreased in a pruA mutant. PruR and DcpA are encoded in an operon with wide conservation among diverse Proteobacteria including mammalian pathogens. Crystal structures reveal that PruR and several orthologs adopt a conserved fold, with a pterin-specific binding cleft that coordinates the bicyclic pterin ring. These findings define a pterin-responsive regulatory mechanism that controls biofilm formation and related c-di-GMP-dependent phenotypes in A. tumefaciens and potentially acts more widely in multiple proteobacterial lineages.


Asunto(s)
Agrobacterium tumefaciens , Proteínas Bacterianas , Biopelículas , GMP Cíclico , Pterinas , Biopelículas/crecimiento & desarrollo , Agrobacterium tumefaciens/metabolismo , Agrobacterium tumefaciens/genética , Pterinas/metabolismo , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteobacteria/metabolismo , Proteobacteria/genética , Cofactores de Molibdeno , Periplasma/metabolismo , Proteínas Periplasmáticas/metabolismo , Proteínas Periplasmáticas/genética , Proteínas de Unión Periplasmáticas/metabolismo , Proteínas de Unión Periplasmáticas/genética , Regulación Bacteriana de la Expresión Génica
15.
PLoS Pathog ; 20(9): e1012533, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39231185

RESUMEN

The body temperature of Warm-blooded hosts impedes and informs responses of bacteria accustomed to cooler environments. The second messenger c-di-GMP modulates bacterial behavior in response to diverse, yet largely undiscovered, stimuli. A long-standing debate persists regarding whether a local or a global c-di-GMP pool plays a critical role. Our research on a Stenotrophomonas maltophilia strain thriving at around 28°C, showcases BtsD as a thermosensor, diguanylate cyclase, and effector. It detects 37°C and diminishes c-di-GMP synthesis, resulting in a responsive sequence: the periplasmic c-di-GMP level is decreased, the N-terminal region of BtsD disengages from c-di-GMP, activates the two-component signal transduction system BtsKR, and amplifies sod1-3 transcription, thereby strengthening the bacterium's pathogenicity and adaptation during infections in 37°C warm Galleria mellonella larvae. This revelation of a single-protein c-di-GMP module introduces unrecognized dimensions to the functional and structural paradigms of c-di-GMP modules and reshapes our understanding of bacterial adaptation and pathogenicity in hosts with a body temperature around 37°C. Furthermore, the discovery of a periplasmic c-di-GMP pool governing BtsD-BtsK interactions supports the critical role of a local c-di-GMP pool.


Asunto(s)
Proteínas Bacterianas , GMP Cíclico , Infecciones por Bacterias Gramnegativas , Stenotrophomonas maltophilia , Stenotrophomonas maltophilia/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Animales , Infecciones por Bacterias Gramnegativas/microbiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Transducción de Señal , Temperatura Corporal/fisiología , Regulación Bacteriana de la Expresión Génica , Liasas de Fósforo-Oxígeno/metabolismo , Liasas de Fósforo-Oxígeno/genética
16.
PLoS Pathog ; 20(6): e1012360, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38935780

RESUMEN

The cGMP-dependent protein kinase (PKG) is the sole cGMP sensor in malaria parasites, acting as an essential signalling hub to govern key developmental processes throughout the parasite life cycle. Despite the importance of PKG in the clinically relevant asexual blood stages, many aspects of malarial PKG regulation, including the importance of phosphorylation, remain poorly understood. Here we use genetic and biochemical approaches to show that reduced cGMP binding to cyclic nucleotide binding domain B does not affect in vitro kinase activity but prevents parasite egress. Similarly, we show that phosphorylation of a key threonine residue (T695) in the activation loop is dispensable for kinase activity in vitro but is essential for in vivo PKG function, with loss of T695 phosphorylation leading to aberrant phosphorylation events across the parasite proteome and changes to the substrate specificity of PKG. Our findings indicate that Plasmodium PKG is uniquely regulated to transduce signals crucial for malaria parasite development.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico , GMP Cíclico , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Fosforilación , GMP Cíclico/metabolismo , Malaria/parasitología , Malaria/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Animales , Plasmodium falciparum/metabolismo , Plasmodium falciparum/genética , Humanos , Transducción de Señal , Eritrocitos/parasitología , Eritrocitos/metabolismo
17.
PLoS Pathog ; 20(9): e1012529, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39241032

RESUMEN

Acinetobacter baumannii is an opportunistic nosocomial pathogen with high morbidity and mortality rates. Current treatment options for this pathogen are limited due to its increasing resistance to last-resort antibiotics. Despite A. baumannii's leading position in the World Health Organisations priority pathogens list, little is known about its virulence regulation. Through a high-throughput screening approach to identify novel biofilm regulators, we identified a previously uncharacterised predicted adenylate cyclase (AC), CavA, as a central regulator of this phenotype. cAMP is a crucial mediator of various aspects of bacterial physiology in other species but information about its role in A. baumannii is limited. We confirm that CavA AC is functional and synthesizes cAMP in A. baumannii. Using dRNA-seq, we verify that CavA is a negative biofilm formation regulator affecting Csu pili and exopolysaccharide production. We demonstrate for the first time that in A. baumannii, cAMP is atop of a hierarchical signalling cascade controlling inter- and intrabacterial signalling by modulating quorum sensing and cyclic di-GMP systems, ultimately governing virulence in vivo and adaptive antibiotic resistance. In contrast to the well-established paradigm in other bacteria where cAMP and cyclic di-GMP levels are inversely regulated, we uncover that the levels of these second messengers are directly proportional in A. baumannii. Overall, this study uncovers the central role of CavA and cAMP in the pathogenic success of A. baumannii and highlights this signalling cascade as a high potential target for novel therapeutic development.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Biopelículas , AMP Cíclico , Transducción de Señal , Acinetobacter baumannii/patogenicidad , Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/genética , AMP Cíclico/metabolismo , Virulencia , Biopelículas/crecimiento & desarrollo , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Ratones , Regulación Bacteriana de la Expresión Génica , Percepción de Quorum/fisiología , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Adenilil Ciclasas/metabolismo , Antibacterianos/farmacología
18.
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
19.
Nucleic Acids Res ; 52(1): 186-203, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38000372

RESUMEN

The ubiquitous bacterial second messenger cyclic diguanylate (c-di-GMP) coordinates diverse cellular processes through its downstream receptors. However, whether c-di-GMP participates in regulating nitrate assimilation is unclear. Here, we found that NasT, an antiterminator involved in nitrate assimilation in Pseudomonas putida, specifically bound c-di-GMP. NasT was essential for expressing the nirBD operon encoding nitrite reductase during nitrate assimilation. High-level c-di-GMP inhibited the binding of NasT to the leading RNA of nirBD operon (NalA), thus attenuating the antitermination function of NasT, resulting in decreased nirBD expression and nitrite reductase activity, which in turn led to increased nitrite accumulation in cells and its export. Molecular docking and point mutation assays revealed five residues in NasT (R70, Q72, D123, K127 and R140) involved in c-di-GMP-binding, of which R140 was essential for both c-di-GMP-binding and NalA-binding. Three diguanylate cyclases (c-di-GMP synthetases) were found to interact with NasT and inhibited nirBD expression, including WspR, PP_2557, and PP_4405. Besides, the c-di-GMP-binding ability of NasT was conserved in the other three representative Pseudomonas species, including P. aeruginosa, P. fluorescens and P. syringae. Our findings provide new insights into nitrate assimilation regulation by revealing the mechanism by which c-di-GMP inhibits nitrate assimilation via NasT.


Asunto(s)
Proteínas Bacterianas , GMP Cíclico , Nitratos , Pseudomonas putida , Proteínas Bacterianas/metabolismo , GMP Cíclico/metabolismo , Regulación Bacteriana de la Expresión Génica , Simulación del Acoplamiento Molecular , Nitratos/metabolismo , Nitrito Reductasas/genética , Nitrito Reductasas/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas putida/genética , Pseudomonas putida/metabolismo
20.
PLoS Genet ; 19(6): e1010819, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37339150

RESUMEN

C-di-GMP is a bacterial second messenger that regulates diverse processes in response to environmental or cellular cues. The nucleoid-associated protein (NAP) CdbA in Myxococcus xanthus binds c-di-GMP and DNA in a mutually exclusive manner in vitro. CdbA is essential for viability, and CdbA depletion causes defects in chromosome organization, leading to a block in cell division and, ultimately, cell death. Most NAPs are not essential; therefore, to explore the paradoxical cdbA essentiality, we isolated suppressor mutations that restored cell viability without CdbA. Most mutations mapped to cdbS, which encodes a stand-alone c-di-GMP binding PilZ domain protein, and caused loss-of-function of cdbS. Cells lacking CdbA and CdbS or only CdbS were fully viable and had no defects in chromosome organization. CdbA depletion caused post-transcriptional upregulation of CdbS accumulation, and this CdbS over-accumulation was sufficient to disrupt chromosome organization and cause cell death. CdbA depletion also caused increased accumulation of CsdK1 and CsdK2, two unusual PilZ-DnaK chaperones. During CdbA depletion, CsdK1 and CsdK2, in turn, enabled the increased accumulation and toxicity of CdbS, likely by stabilizing CdbS. Moreover, we demonstrate that heat stress, possibly involving an increased cellular c-di-GMP concentration, induced the CdbA/CsdK1/CsdK2/CdbS system, causing a CsdK1- and CsdK2-dependent increase in CdbS accumulation. Thereby this system accelerates heat stress-induced chromosome mis-organization and cell death. Collectively, this work describes a unique system that contributes to regulated cell death in M. xanthus and suggests a link between c-di-GMP signaling and regulated cell death in bacteria.


Asunto(s)
Proteínas Bacterianas , Myxococcus xanthus , Proteínas Bacterianas/metabolismo , Myxococcus xanthus/genética , Proteínas Portadoras/genética , Chaperonas Moleculares/genética , Muerte Celular , Cromosomas/metabolismo , GMP Cíclico/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA