Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 866
Filtrar
1.
Nat Commun ; 15(1): 3825, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714645

RESUMO

c-di-AMP is an essential and widespread nucleotide second messenger in bacterial signaling. For most c-di-AMP synthesizing organisms, c-di-AMP homeostasis and the molecular mechanisms pertaining to its signal transduction are of great concern. Here we show that c-di-AMP binds the N-acetylglucosamine (GlcNAc)-sensing regulator DasR, indicating a direct link between c-di-AMP and GlcNAc signaling. Beyond its foundational role in cell-surface structure, GlcNAc is attractive as a major nutrient and messenger molecule regulating multiple cellular processes from bacteria to humans. We show that increased c-di-AMP levels allosterically activate DasR as a master repressor of GlcNAc utilization, causing the shutdown of the DasR-mediated GlcNAc signaling cascade and leading to a consistent enhancement in the developmental transition and antibiotic production in Saccharopolyspora erythraea. The expression of disA, encoding diadenylate cyclase, is directly repressed by the regulator DasR in response to GlcNAc signaling, thus forming a self-sustaining transcriptional feedback loop for c-di-AMP synthesis. These findings shed light on the allosteric regulation by c-di-AMP, which appears to play a prominent role in global signal integration and c-di-AMP homeostasis in bacteria and is likely widespread in streptomycetes that produce c-di-AMP.


Assuntos
Acetilglucosamina , Proteínas de Bactérias , Fosfatos de Dinucleosídeos , Regulação Bacteriana da Expressão Gênica , Saccharopolyspora , Transdução de Sinais , Acetilglucosamina/metabolismo , Regulação Alostérica , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Fosfatos de Dinucleosídeos/metabolismo , Saccharopolyspora/metabolismo , Saccharopolyspora/genética
2.
Proc Natl Acad Sci U S A ; 121(18): e2318666121, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38652747

RESUMO

In bacteria, intracellular K+ is involved in the regulation of membrane potential, cytosolic pH, and cell turgor as well as in spore germination, environmental adaptation, cell-to-cell communication in biofilms, antibiotic sensitivity, and infectivity. The second messenger cyclic-di-AMP (c-di-AMP) has a central role in modulating the intracellular K+ concentration in many bacterial species, controlling transcription and function of K+ channels and transporters. However, our understanding of how this regulatory network responds to c-di-AMP remains poor. We used the RCK (Regulator of Conductance of K+) proteins that control the activity of Ktr channels in Bacillus subtilis as a model system to analyze the regulatory function of c-di-AMP with a combination of in vivo and in vitro functional and structural characterization. We determined that the two RCK proteins (KtrA and KtrC) are neither physiologically redundant or functionally equivalent. KtrC is the physiologically dominant RCK protein in the regulation of Ktr channel activity. In explaining this hierarchical organization, we found that, unlike KtrA, KtrC is very sensitive to c-di-AMP inactivation and lack of c-di-AMP regulation results in RCK protein toxicity, most likely due to unregulated K+ flux. We also found that KtrC can assemble with KtrA, conferring c-di-AMP regulation to the functional KtrA/KtrC heteromers and potentially compensating KtrA toxicity. Altogether, we propose that the central role of c-di-AMP in the control of the K+ machinery, by modulating protein levels through gene transcription and by regulating protein activity, has determined the evolutionary selection of KtrC as the dominant RCK protein, shaping the hierarchical organization of regulatory components of the K+ machinery.


Assuntos
Bacillus subtilis , Proteínas de Bactérias , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Bacillus subtilis/metabolismo , Bacillus subtilis/genética , Potássio/metabolismo , Regulação Bacteriana da Expressão Gênica , Fosfatos de Dinucleosídeos/metabolismo , Canais de Potássio/metabolismo , Canais de Potássio/genética
3.
Acta Crystallogr D Struct Biol ; 80(Pt 5): 350-361, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38682668

RESUMO

CdaA is the most widespread diadenylate cyclase in many bacterial species, including several multidrug-resistant human pathogens. The enzymatic product of CdaA, cyclic di-AMP, is a secondary messenger that is essential for the viability of many bacteria. Its absence in humans makes CdaA a very promising and attractive target for the development of new antibiotics. Here, the structural results are presented of a crystallographic fragment screen against CdaA from Listeria monocytogenes, a saprophytic Gram-positive bacterium and an opportunistic food-borne pathogen that can cause listeriosis in humans and animals. Two of the eight fragment molecules reported here were localized in the highly conserved ATP-binding site. These fragments could serve as potential starting points for the development of antibiotics against several CdaA-dependent bacterial species.


Assuntos
Listeria monocytogenes , Listeria monocytogenes/enzimologia , Cristalografia por Raios X/métodos , Sítios de Ligação , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Fosfatos de Dinucleosídeos/metabolismo , Fosfatos de Dinucleosídeos/química , Antibacterianos/farmacologia , Humanos , Fósforo-Oxigênio Liases/química , Fósforo-Oxigênio Liases/metabolismo , Conformação Proteica
4.
Angew Chem Int Ed Engl ; 63(6): e202314951, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-37934413

RESUMO

The recent expansion of the field of RNA chemical modifications has changed our understanding of post-transcriptional gene regulation. Apart from internal nucleobase modifications, 7-methylguanosine was long thought to be the only eukaryotic RNA cap. However, the discovery of non-canonical RNA caps in eukaryotes revealed a new niche of previously undetected RNA chemical modifications. We are the first to report the existence of a new non-canonical RNA cap, diadenosine tetraphosphate (Ap4 A), in human and rat cell lines. Ap4 A is the most abundant dinucleoside polyphosphate in eukaryotic cells and can be incorporated into RNA by RNA polymerases as a non-canonical initiating nucleotide (NCIN). Using liquid chromatography-mass spectrometry (LC-MS), we show that the amount of capped Ap4 A-RNA is independent of the cellular concentration of Ap4 A. A decapping enzyme screen identifies two enzymes cleaving Ap4 A-RNA,NUDT2 and DXO, both of which also cleave other substrate RNAs in vitro. We further assess the translatability and immunogenicity of Ap4 A-RNA and show that although it is not translated, Ap4 A-RNA is recognized as self by the cell and does not elicit an immune response, making it a natural component of the transcriptome. Our findings open a previously unexplored area of eukaryotic RNA regulation.


Assuntos
Fosfatos de Dinucleosídeos , Capuzes de RNA , Ratos , Animais , Humanos , Fosfatos de Dinucleosídeos/metabolismo , Mamíferos/metabolismo , Nudix Hidrolases , Monoéster Fosfórico Hidrolases
5.
Mol Microbiol ; 120(6): 791-804, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37898560

RESUMO

Cyclic dimeric adenosine monophosphate (c-di-AMP) has been well studied in bacteria, including those of the genus Streptococcus, since the first recognition of this dinucleotide in 2008. Streptococci possess a sole diadenylate cyclase, CdaA, and distinct c-di-AMP phosphodiesterases. Interestingly, cdaA is required for viability of some streptococcal species but not all when streptococci are grown in standard laboratory media. Bacteria of this genus also have distinct c-di-AMP effector proteins, diverse c-di-AMP-signaling pathways, and subsequent biological outcomes. In streptococci, c-di-AMP may influence bacterial growth, morphology, biofilm formation, competence program, drug resistance, and bacterial pathogenesis. c-di-AMP secreted by streptococci has also been shown to interact with the mammalian host and induces immune responses including type I interferon production. In this review, we summarize the reported c-di-AMP networks in seven species of the genus Streptococcus, which cause diverse clinical manifestations, and propose future perspectives to investigate the signaling molecule in these streptococcal pathogens.


Assuntos
Proteínas de Bactérias , Sistemas do Segundo Mensageiro , Animais , Proteínas de Bactérias/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , AMP Cíclico/metabolismo , Bactérias/metabolismo , Streptococcus/metabolismo , Mamíferos/metabolismo
6.
Biochemistry ; 62(15): 2238-2243, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37418448

RESUMO

Adenylate kinases play a crucial role in cellular energy homeostasis through the interconversion of ATP, AMP, and ADP in all living organisms. Here, we explore how adenylate kinase (AdK) from Escherichia coli interacts with diadenosine tetraphosphate (AP4A), a putative alarmone associated with transcriptional regulation, stress, and DNA damage response. From a combination of EPR and NMR spectroscopy together with X-ray crystallography, we found that AdK interacts with AP4A with two distinct modes that occur on disparate time scales. First, AdK dynamically interconverts between open and closed states with equal weights in the presence of AP4A. On a much slower time scale, AdK hydrolyses AP4A, and we suggest that the dynamically accessed substrate-bound open AdK conformation enables this hydrolytic activity. The partitioning of the enzyme into open and closed states is discussed in relation to a recently proposed linkage between active site dynamics and collective conformational dynamics.


Assuntos
Adenilato Quinase , Escherichia coli , Escherichia coli/metabolismo , Adenilato Quinase/química , Hidrólise , Fosfatos de Dinucleosídeos/metabolismo , Catálise , Domínio Catalítico
7.
Nat Commun ; 14(1): 3683, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37344476

RESUMO

Cyclic di-AMP is the only known essential second messenger in bacteria and archaea, regulating different proteins indispensable for numerous physiological processes. In particular, it controls various potassium and osmolyte transporters involved in osmoregulation. In Bacillus subtilis, the K+/H+ symporter KimA of the KUP family is inactivated by c-di-AMP. KimA sustains survival at potassium limitation at low external pH by mediating potassium ion uptake. However, at elevated intracellular K+ concentrations, further K+ accumulation would be toxic. In this study, we reveal the molecular basis of how c-di-AMP binding inhibits KimA. We report cryo-EM structures of KimA with bound c-di-AMP in detergent solution and reconstituted in amphipols. By combining structural data with functional assays and molecular dynamics simulations we reveal how c-di-AMP modulates transport. We show that an intracellular loop in the transmembrane domain interacts with c-di-AMP bound to the adjacent cytosolic domain. This reduces the mobility of transmembrane helices at the cytosolic side of the K+ binding site and therefore traps KimA in an inward-occluded conformation.


Assuntos
AMP Cíclico , Prótons , Proteínas de Bactérias/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Potássio/metabolismo , Fosfatos de Dinucleosídeos/metabolismo
8.
J Bacteriol ; 205(4): e0002323, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37022175

RESUMO

Cyclic dimeric AMP (c-di-AMP) is a widespread second messenger that controls such key functions as osmotic homeostasis, peptidoglycan biosynthesis, and response to various stresses. C-di-AMP is synthesized by diadenylate cyclases that contain the DAC (DisA_N) domain, which was originally characterized as the N-terminal domain in the DNA integrity scanning protein DisA. In other experimentally studied diadenylate cyclases, DAC domain is typically located at the protein C termini and its enzymatic activity is controlled by one or more N-terminal domains. As in other bacterial signal transduction proteins, these N-terminal modules appear to sense environmental or intracellular signals through ligand binding and/or protein-protein interactions. Studies of bacterial and archaeal diadenylate cyclases also revealed numerous sequences with uncharacterized N-terminal regions. This work provides a comprehensive review of the N-terminal domains of bacterial and archaeal diadenylate cyclases, including the description of five previously undefined domains and three PK_C-related domains of the DacZ_N superfamily. These data are used to classify diadenylate cyclases into 22 families, based on their conserved domain architectures and the phylogeny of their DAC domains. Although the nature of the regulatory signals remains obscure, the association of certain dac genes with anti-phage defense CBASS systems and other phage-resistance genes suggests that c-di-AMP might also be involved in the signaling of phage infection.


Assuntos
Archaea , Fósforo-Oxigênio Liases , Humanos , Archaea/genética , Archaea/metabolismo , Fósforo-Oxigênio Liases/metabolismo , Proteínas de Bactérias/metabolismo , Bactérias/genética , Bactérias/metabolismo , Sistemas do Segundo Mensageiro , AMP Cíclico/metabolismo , Fosfatos de Dinucleosídeos/metabolismo
9.
Cell Rep ; 42(4): 112305, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36952342

RESUMO

Programmed cell suicide of infected bacteria, known as abortive infection (Abi), serves as an immune defense strategy to prevent the propagation of bacteriophage viruses. Many Abi systems utilize bespoke cyclic nucleotide immune messengers generated upon infection to mobilize cognate death effectors. Here, we identify a family of bacteriophage nucleotidyltransferases (NTases) that synthesize competitor cyclic dinucleotide (CDN) ligands and inhibit TIR NADase effectors activated via a linked STING CDN sensor domain (TIR-STING). Through a functional screen of NTase-adjacent phage genes, we uncover candidate inhibitors of cell suicide induced by heterologous expression of tonically active TIR-STING. Among these, we demonstrate that a virus MazG-like nucleotide pyrophosphohydrolase, Atd1, depletes the starvation alarmone (p)ppGpp, revealing a potential role for the alarmone-activated host toxin MazF as an executioner of TIR-driven Abi. Phage NTases and counterdefenses like Atd1 preserve host viability to ensure virus propagation and represent tools to modulate TIR and STING immune responses.


Assuntos
Bacteriófagos , Guanosina Pentafosfato , Bactérias/metabolismo , Bactérias/virologia , Bacteriófagos/fisiologia , Fosfatos de Dinucleosídeos/metabolismo , Imunidade , Nucleotídeos , Nucleotidiltransferases/metabolismo
10.
Protein Sci ; 32(3): e4568, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36660887

RESUMO

Cyclic-di-nucleotide-based secondary messengers regulate various physiological functions, including stress responses in bacteria. Cyclic diadenosine monophosphate (c-di-AMP) has recently emerged as a crucial second messenger with implications in processes including osmoregulation, antibiotic resistance, biofilm formation, virulence, DNA repair, ion homeostasis, and sporulation, and has potential therapeutic applications. The contrasting activities of the enzymes diadenylate cyclase (DAC) and phosphodiesterase (PDE) determine the equilibrium levels of c-di-AMP. Although c-di-AMP is suspected of playing an essential role in the pathophysiology of bacterial infections and in regulating host-pathogen interactions, the mechanisms of its regulation remain relatively unexplored in mycobacteria. In this report, we biochemically and structurally characterize the c-di-AMP synthase (MsDisA) from Mycobacterium smegmatis. The enzyme activity is regulated by pH and substrate concentration; conditions of significance in the homoeostasis of c-di-AMP levels. Substrate binding stimulates conformational changes in the protein, and pApA and ppApA are synthetic intermediates detectable when enzyme efficiency is low. Unlike the orthologous Bacillus subtilis enzyme, MsDisA does not bind to, and its activity is not influenced in the presence of DNA. Furthermore, we have determined the cryo-EM structure of MsDisA, revealing asymmetry in its structure in contrast to the symmetric crystal structure of Thermotoga maritima DisA. We also demonstrate that the N-terminal minimal region alone is sufficient and essential for oligomerization and catalytic activity. Our data shed light on the regulation of mycobacterial DisA and possible future directions to pursue.


Assuntos
Proteínas de Bactérias , Mycobacterium smegmatis , Mycobacterium smegmatis/genética , Proteínas de Bactérias/química , Fosfatos de Dinucleosídeos/química , Fosfatos de Dinucleosídeos/metabolismo , Bacillus subtilis/genética
11.
FEBS J ; 290(11): 2968-2992, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36629470

RESUMO

Cyclic di-AMP is an essential signalling molecule in Gram-positive bacteria. This second messenger regulates the osmotic pressure of the cell by interacting directly with the regulatory domains, either RCK_C or CBS domains, of several potassium and osmolyte uptake membrane protein systems. Cyclic di-AMP also targets stand-alone CBS domain proteins such as DarB in Bacillus subtilis and CbpB in Listeria monocytogenes. We show here that the CbpB protein of Group B Streptococcus binds c-di-AMP with a very high affinity. Crystal structures of CbpB reveal the determinants of binding specificity and significant conformational changes occurring upon c-di-AMP binding. Deletion of the cbpB gene alters bacterial growth in low potassium conditions most likely due to a decrease in the amount of ppGpp caused by a loss of interaction between CbpB and Rel, the GTP/GDP pyrophosphokinase.


Assuntos
Proteínas de Transporte , Streptococcus agalactiae , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo , Guanosina Pentafosfato , Guanosina Tetrafosfato , Proteínas de Bactérias/metabolismo , AMP Cíclico , Fosfatos de Dinucleosídeos/metabolismo , Potássio/metabolismo
12.
Angew Chem Int Ed Engl ; 62(8): e202213279, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36524454

RESUMO

Diadenosine polyphosphates (Apn As) are non-canonical nucleotides whose cellular concentrations increase during stress and are therefore termed alarmones, signaling homeostatic imbalance. Their cellular role is poorly understood. In this work, we assessed Apn As for their usage as cosubstrates for protein AMPylation, a post-translational modification in which adenosine monophosphate (AMP) is transferred to proteins. In humans, AMPylation mediated by the AMPylator FICD with ATP as a cosubstrate is a response to ER stress. Herein, we demonstrate that Ap4 A is proficiently consumed for AMPylation by FICD. By chemical proteomics using a new chemical probe, we identified new potential AMPylation targets. Interestingly, we found that AMPylation targets of FICD may differ depending on the nucleotide cosubstrate. These results may suggest that signaling at elevated Ap4 A levels during cellular stress differs from when Ap4 A is present at low concentrations, allowing response to extracellular cues.


Assuntos
Guanosina Pentafosfato , Proteínas , Humanos , Guanosina Pentafosfato/metabolismo , Proteínas/metabolismo , Monofosfato de Adenosina/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Processamento de Proteína Pós-Traducional
13.
J Pept Sci ; 29(3): e3458, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36264037

RESUMO

Intracellular dinucleoside polyphosphates (Npn Ns) have been known for decades but the functional role remains enigmatic. Diadenosine triphosphate (Ap3 A) is one of the most prominent examples, and its intercellular concentration was shown to increase upon cellular stress. By employment of previously reported Ap3 A-based photoaffinity-labeling probes (PALPs) in chemical proteomics, we investigated the Ap3 A interactome in the human lung carcinoma cell line H1299. The cell line is deficient of the fragile histidine triade (Fhit) protein, a hydrolase of Ap3 A and tumor suppressor. Overall, the number of identified potential interaction partners was significantly lower than in the previously investigated HEK293T cell line. Gene ontology term analysis revealed that the identified proteins participate in similar pathways as for HEK293T, but the percentage of proteins involved in RNA-related processes is higher for H1299. The obtained results highlight similarities and differences of the Ap3 A interaction network in different cell lines and give further indications regarding the importance of the presence of Fhit.


Assuntos
Fosfatos de Dinucleosídeos , Neoplasias , Humanos , Fosfatos de Dinucleosídeos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Guanosina Pentafosfato , Hidrolases Anidrido Ácido/genética , Hidrolases Anidrido Ácido/metabolismo , Células HEK293 , Proteômica
14.
Nature ; 612(7938): 170-176, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36265513

RESUMO

Cyclic dinucleotides (CDNs) are ubiquitous signalling molecules in all domains of life1,2. Mammalian cells produce one CDN, 2'3'-cGAMP, through cyclic GMP-AMP synthase after detecting cytosolic DNA signals3-7. 2'3'-cGAMP, as well as bacterial and synthetic CDN analogues, can act as second messengers to activate stimulator of interferon genes (STING) and elicit broad downstream responses8-21. Extracellular CDNs must traverse the cell membrane to activate STING, a process that is dependent on the solute carrier SLC19A122,23. Moreover, SLC19A1 represents the major transporter for folate nutrients and antifolate therapeutics24,25, thereby placing SLC19A1 as a key factor in multiple physiological and pathological processes. How SLC19A1 recognizes and transports CDNs, folate and antifolate is unclear. Here we report cryo-electron microscopy structures of human SLC19A1 (hSLC19A1) in a substrate-free state and in complexes with multiple CDNs from different sources, a predominant natural folate and a new-generation antifolate drug. The structural and mutagenesis results demonstrate that hSLC19A1 uses unique yet divergent mechanisms to recognize CDN- and folate-type substrates. Two CDN molecules bind within the hSLC19A1 cavity as a compact dual-molecule unit, whereas folate and antifolate bind as a monomer and occupy a distinct pocket of the cavity. Moreover, the structures enable accurate mapping and potential mechanistic interpretation of hSLC19A1 with loss-of-activity and disease-related mutations. Our research provides a framework for understanding the mechanism of SLC19-family transporters and is a foundation for the development of potential therapeutics.


Assuntos
Microscopia Crioeletrônica , Fosfatos de Dinucleosídeos , Antagonistas do Ácido Fólico , Ácido Fólico , Nucleotídeos Cíclicos , Animais , Humanos , Fosfatos de Dinucleosídeos/metabolismo , Ácido Fólico/metabolismo , Antagonistas do Ácido Fólico/farmacologia , Mamíferos/metabolismo , Nucleotídeos Cíclicos/metabolismo , Proteína Carregadora de Folato Reduzido/química , Proteína Carregadora de Folato Reduzido/genética , Proteína Carregadora de Folato Reduzido/metabolismo , Proteína Carregadora de Folato Reduzido/ultraestrutura
15.
ACS Sens ; 7(9): 2732-2742, 2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-35981239

RESUMO

Fluorescence-guided cancer surgery can dramatically improve recurrence rates and postoperative quality of life of patients by accurately distinguishing the boundary between normal and cancer tissues during surgery, thereby minimizing excision of normal tissue. One promising target in early stage cancer is fragile histidine triad (FHIT), a cancer suppressor protein with dinucleoside triphosphate hydrolase activity. In this study, we have developed fluorescence probes containing a nucleoside diphosphate moiety, which dramatically improves the reactivity and specificity for FHIT, and a moderately lipophilic ester moiety to increase the membrane permeability. The ester moiety is cleaved by ubiquitous intracellular esterases, and then, FHIT in the cells specifically cleaves nucleoside monophosphate. The remaining phosphate moiety is rapidly cleaved by ubiquitous intracellular phosphatases to release the fluorescent dye. We confirmed that this probe can detect FHIT activity in living cells. A comprehensive evaluation of the effects of various ester moieties revealed that probes with CLogP = 5-7 showed good membrane permeability and were good substrates of the target enzyme; these findings may be helpful in the rational design of other multiple phosphate-containing probes targeting intracellular enzymes.


Assuntos
Hidrolases Anidrido Ácido , Histidina , Hidrolases Anidrido Ácido/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Difosfatos , Esterases , Ésteres , Fluorescência , Corantes Fluorescentes , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Neoplasias/metabolismo , Nucleosídeos , Monoéster Fosfórico Hidrolases , Qualidade de Vida
16.
Nature ; 608(7924): 803-807, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35859168

RESUMO

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.


Assuntos
Proteínas de Bactérias , Microscopia Crioeletrônica , Proteínas de Membrana , Receptores de Interleucina-1 , Sphingobacterium , Receptores Toll-Like , Animais , Antivirais/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Bacteriófagos/imunologia , Fosfatos de Dinucleosídeos/metabolismo , Humanos , Imunidade Inata , Proteínas de Membrana/química , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Óperon/genética , Receptores de Interleucina-1/química , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/ultraestrutura , Sphingobacterium/química , Sphingobacterium/genética , Sphingobacterium/ultraestrutura , Sphingobacterium/virologia , Receptores Toll-Like/química , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Receptores Toll-Like/ultraestrutura
17.
NPJ Biofilms Microbiomes ; 8(1): 53, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794154

RESUMO

Microbial pathogens employ signaling systems through cyclic (di-) nucleotide monophosphates serving as second messengers to increase fitness during pathogenesis. However, signaling schemes via second messengers in Porphyromonas gingivalis, a key Gram-negative anaerobic oral pathogen, remain unknown. Here, we report that among various ubiquitous second messengers, P. gingivalis strains predominantly synthesize bis-(3',5')-cyclic di-adenosine monophosphate (c-di-AMP), which is essential for their growth and survival. Our findings demonstrate an unusual regulation of c-di-AMP synthesis in P. gingivalis. P. gingivalis c-di-AMP phosphodiesterase (PDE) gene (pdepg) positively regulates c-di-AMP synthesis and impedes a decrease in c-di-AMP concentration despite encoding conserved amino acid motifs for phosphodiesterase activity. Instead, the predicted regulator gene cdaR, unrelated to the c-di-AMP PDE genes, serves as a potent negative regulator of c-di-AMP synthesis in this anaerobe. Further, our findings reveal that pdepg and cdaR are required to regulate the incorporation of ATP into c-di-AMP upon pyruvate utilization, leading to enhanced biofilm formation. We show that shifts in c-di-AMP signaling change the integrity and homeostasis of cell envelope, importantly, the structure and immunoreactivity of the lipopolysaccharide layer. Additionally, microbe-microbe interactions and the virulence potential of P. gingivalis were modulated by c-di-AMP. These studies provide the first glimpse into the scheme of second messenger signaling in P. gingivalis and perhaps other Bacteroidetes. Further, our findings indicate that c-di-AMP signaling promotes the fitness of the residents of the oral cavity and the development of a pathogenic community.


Assuntos
AMP Cíclico , Porphyromonas gingivalis , Monofosfato de Adenosina , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , AMP Cíclico/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Homeostase , Diester Fosfórico Hidrolases/genética , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/metabolismo , Virulência
18.
Environ Microbiol ; 24(9): 4466-4488, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35688634

RESUMO

The human pathogen Listeria monocytogenes synthesizes and degrades c-di-AMP using the diadenylate cyclase CdaA and the phosphodiesterases PdeA and PgpH respectively. c-di-AMP is essential because it prevents the uncontrolled uptake of osmolytes. Here, we studied the phenotypes of cdaA, pdeA, pgpH and pdeA pgpH mutants with defects in c-di-AMP metabolism and characterized suppressor mutants restoring their growth defects. The characterization of the pdeA pgpH mutant revealed that the bacteria show growth defects in defined medium, a phenotype that is invariably suppressed by mutations in cdaA. The previously reported growth defect of the cdaA mutant in rich medium is suppressed by mutations that osmotically stabilize the c-di-AMP-free strain. We also found that the cdaA mutant has an increased sensitivity against isoleucine. The isoleucine-dependent growth inhibition of the cdaA mutant is suppressed by codY mutations that likely reduce the DNA-binding activity of encoded CodY variants. Moreover, the characterization of the cdaA suppressor mutants revealed that the Opp oligopeptide transport system is involved in the uptake of the antibiotic fosfomycin. In conclusion, the suppressor analysis corroborates a key function of c-di-AMP in controlling osmolyte homeostasis in L. monocytogenes.


Assuntos
Fosfomicina , Listeria monocytogenes , Acetamidas , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , DNA/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Fosfomicina/metabolismo , Fosfomicina/farmacologia , Humanos , Isoleucina/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Oligopeptídeos/metabolismo , Diester Fosfórico Hidrolases/genética , Fósforo-Oxigênio Liases/genética
19.
J Biol Chem ; 298(7): 102144, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35714772

RESUMO

The bacterial second messenger c-di-AMP controls essential cellular processes, including potassium and osmolyte homeostasis. This makes synthesizing enzymes and components involved in c-di-AMP signal transduction intriguing as potential targets for drug development. The c-di-AMP receptor protein DarB of Bacillus subtilis binds the Rel protein and triggers the Rel-dependent stringent response to stress conditions; however, the structural basis for this trigger is unclear. Here, we report crystal structures of DarB in the ligand-free state and of DarB complexed with c-di-AMP, 3'3'-cGAMP, and AMP. We show that DarB forms a homodimer with a parallel, head-to-head assembly of the monomers. We also confirm the DarB dimer binds two cyclic dinucleotide molecules or two AMP molecules; only one adenine of bound c-di-AMP is specifically recognized by DarB, while the second protrudes out of the donut-shaped protein. This enables DarB to bind also 3'3'-cGAMP, as only the adenine fits in the active site. In absence of c-di-AMP, DarB binds to Rel and stimulates (p)ppGpp synthesis, whereas the presence of c-di-AMP abolishes this interaction. Furthermore, the DarB crystal structures reveal no conformational changes upon c-di-AMP binding, leading us to conclude the regulatory function of DarB on Rel must be controlled directly by the bound c-di-AMP. We thus derived a structural model of the DarB-Rel complex via in silico docking, which was validated with mass spectrometric analysis of the chemically crosslinked DarB-Rel complex and mutagenesis studies. We suggest, based on the predicted complex structure, a mechanism of stringent response regulation by c-di-AMP.


Assuntos
Proteínas de Bactérias , Fosfatos de Dinucleosídeos , Adenina/metabolismo , Monofosfato de Adenosina/metabolismo , Bacillus subtilis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Fosfatos de Dinucleosídeos/química , Fosfatos de Dinucleosídeos/metabolismo
20.
Nature ; 604(7906): 557-562, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35388221

RESUMO

Stimulator of interferon genes (STING) is an adaptor protein in innate immunity against DNA viruses or bacteria1-5. STING-mediated immunity could be exploited in the development of vaccines or cancer immunotherapies. STING is a transmembrane dimeric protein that is located in the endoplasmic reticulum or in the Golgi apparatus. STING is activated by the binding of its cytoplasmic ligand-binding domain to cyclic dinucleotides that are produced by the DNA sensor cyclic GMP-AMP (cGAMP) synthase or by invading bacteria1,6,7. Cyclic dinucleotides induce a conformational change in the STING ligand-binding domain, which leads to a high-order oligomerization of STING that is essential for triggering the downstream signalling pathways8,9. However, the cGAMP-induced STING oligomers tend to dissociate in solution and have not been resolved to high resolution, which limits our understanding of the activation mechanism. Here we show that a small-molecule agonist, compound 53 (C53)10, promotes the oligomerization and activation of human STING through a mechanism orthogonal to that of cGAMP. We determined a cryo-electron microscopy structure of STING bound to both C53 and cGAMP, revealing a stable oligomer that is formed by side-by-side packing and has a curled overall shape. Notably, C53 binds to a cryptic pocket in the STING transmembrane domain, between the two subunits of the STING dimer. This binding triggers outward shifts of transmembrane helices in the dimer, and induces inter-dimer interactions between these helices to mediate the formation of the high-order oligomer. Our functional analyses show that cGAMP and C53 together induce stronger activation of STING than either ligand alone.


Assuntos
Proteínas de Membrana , Nucleotídeos Cíclicos , Proteínas de Ciclo Celular , Microscopia Crioeletrônica , Fosfatos de Dinucleosídeos/metabolismo , Humanos , Imunidade Inata , Ligantes , Proteínas de Membrana/metabolismo , Nucleotídeos Cíclicos/metabolismo , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...