Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 170(4): 787-799.e18, 2017 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-28802046

RESUMO

Replication-transcription collisions shape genomes, influence evolution, and promote genetic diseases. Although unclear why, head-on transcription (lagging strand genes) is especially disruptive to replication and promotes genomic instability. Here, we find that head-on collisions promote R-loop formation in Bacillus subtilis. We show that pervasive R-loop formation at head-on collision regions completely blocks replication, elevates mutagenesis, and inhibits gene expression. Accordingly, the activity of the R-loop processing enzyme RNase HIII at collision regions is crucial for stress survival in B. subtilis, as many stress response genes are head-on to replication. Remarkably, without RNase HIII, the ability of the intracellular pathogen Listeria monocytogenes to infect and replicate in hosts is weakened significantly, most likely because many virulence genes are head-on to replication. We conclude that the detrimental effects of head-on collisions stem primarily from excessive R-loop formation and that the resolution of these structures is critical for bacterial stress survival and pathogenesis.


Assuntos
Bacillus subtilis/fisiologia , Replicação do DNA , Listeria monocytogenes/fisiologia , Transcrição Gênica , Animais , Período de Replicação do DNA , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Camundongos , Estresse Fisiológico , Virulência
2.
Cell ; 158(6): 1389-1401, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25215494

RESUMO

Cyclic di-adenosine monophosphate (c-di-AMP) is a broadly conserved second messenger required for bacterial growth and infection. However, the molecular mechanisms of c-di-AMP signaling are still poorly understood. Using a chemical proteomics screen for c-di-AMP-interacting proteins in the pathogen Listeria monocytogenes, we identified several broadly conserved protein receptors, including the central metabolic enzyme pyruvate carboxylase (LmPC). Biochemical and crystallographic studies of the LmPC-c-di-AMP interaction revealed a previously unrecognized allosteric regulatory site 25 Å from the active site. Mutations in this site disrupted c-di-AMP binding and affected catalytic activity of LmPC as well as PC from pathogenic Enterococcus faecalis. C-di-AMP depletion resulted in altered metabolic activity in L. monocytogenes. Correction of this metabolic imbalance rescued bacterial growth, reduced bacterial lysis, and resulted in enhanced bacterial burdens during infection. These findings greatly expand the c-di-AMP signaling repertoire and reveal a central metabolic regulatory role for a cyclic dinucleotide.


Assuntos
Fosfatos de Dinucleosídeos/metabolismo , Listeria monocytogenes/metabolismo , Piruvato Carboxilase/química , Piruvato Carboxilase/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Animais , Bacteriólise , Sítios de Ligação , Cristalografia por Raios X , Interações Hospedeiro-Patógeno , Listeria monocytogenes/enzimologia , Listeria monocytogenes/crescimento & desenvolvimento , Listeriose/microbiologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular
3.
Immunity ; 46(3): 433-445, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28329705

RESUMO

Bacterial and host cyclic dinucleotides (cdNs) mediate cytosolic immune responses through the STING signaling pathway, although evidence suggests that alternative pathways exist. We used cdN-conjugated beads to biochemically isolate host receptors for bacterial cdNs, and we identified the oxidoreductase RECON. High-affinity cdN binding inhibited RECON enzyme activity by simultaneously blocking the substrate and cosubstrate sites, as revealed by structural analyses. During bacterial infection of macrophages, RECON antagonized STING activation by acting as a molecular sink for cdNs. Bacterial infection of hepatocytes, which do not express STING, revealed that RECON negatively regulates NF-κB activation. Loss of RECON activity, via genetic ablation or inhibition by cdNs, increased NF-κB activation and reduced bacterial survival, suggesting that cdN inhibition of RECON promotes a proinflammatory, antibacterial state that is distinct from the antiviral state associated with STING activation. Thus, RECON functions as a cytosolic sensor for bacterial cdNs, shaping inflammatory gene activation via its effects on STING and NF-κB.


Assuntos
Infecções Bacterianas/imunologia , Proteínas de Bactérias/imunologia , Estradiol Desidrogenases/imunologia , Inflamação/imunologia , NF-kappa B/imunologia , Animais , Ativação Enzimática/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
4.
Nature ; 579(7800): E12, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32144410

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nature ; 573(7774): 434-438, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31511694

RESUMO

The accumulation of DNA in the cytosol serves as a key immunostimulatory signal associated with infections, cancer and genomic damage1,2. Cytosolic DNA triggers immune responses by activating the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway3. The binding of DNA to cGAS activates its enzymatic activity, leading to the synthesis of a second messenger, cyclic guanosine monophosphate-adenosine monophosphate (2'3'-cGAMP)4-7. This cyclic dinucleotide (CDN) activates STING8, which in turn activates the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), promoting the transcription of genes encoding type I interferons and other cytokines and mediators that stimulate a broader immune response. Exogenous 2'3'-cGAMP produced by malignant cells9 and other CDNs, including those produced by bacteria10-12 and synthetic CDNs used in cancer immunotherapy13,14, must traverse the cell membrane to activate STING in target cells. How these charged CDNs pass through the lipid bilayer is unknown. Here we used a genome-wide CRISPR-interference screen to identify the reduced folate carrier SLC19A1, a folate-organic phosphate antiporter, as the major transporter of CDNs. Depleting SLC19A1 in human cells inhibits CDN uptake and functional responses, and overexpressing SLC19A1 increases both uptake and functional responses. In human cell lines and primary cells ex vivo, CDN uptake is inhibited by folates as well as two medications approved for treatment of inflammatory diseases, sulfasalazine and the antifolate methotrexate. The identification of SLC19A1 as the major transporter of CDNs into cells has implications for the immunotherapeutic treatment of cancer13, host responsiveness to CDN-producing pathogenic microorganisms11 and-potentially-for some inflammatory diseases.


Assuntos
DNA/metabolismo , Nucleotídeos Cíclicos/metabolismo , Proteína Carregadora de Folato Reduzido/metabolismo , Animais , Citosol , DNA/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Nucleotídeos Cíclicos/imunologia , Nucleotidiltransferases/metabolismo , Proteína Carregadora de Folato Reduzido/imunologia
6.
J Bacteriol ; 203(19): e0008021, 2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34309402

RESUMO

3'3'-Cyclic di-AMP (c-di-AMP) is an important nucleotide second messenger found throughout the bacterial domain of life. c-di-AMP is essential in many bacteria and regulates a diverse array of effector proteins controlling pathogenesis, cell wall homeostasis, osmoregulation, and central metabolism. Despite the ubiquity and importance of c-di-AMP, methods to detect this signaling molecule are limited, particularly at single-cell resolution. In this work, crystallization of the Listeria monocytogenes c-di-AMP effector protein Lmo0553 enabled structure-guided design of a Förster resonance energy transfer (FRET)-based biosensor, which we have named CDA5. CDA5 is a fully genetically encodable, specific, and reversible biosensor which allows the detection of c-di-AMP dynamics both in vitro and within live cells in a nondestructive manner. Our initial studies identified a distribution of c-di-AMP in Bacillus subtilis populations first grown in Luria broth and then resuspended in diluted Luria broth compatible with fluorescence analysis. Furthermore, we found that B. subtilis mutants lacking either a c-di-AMP phosphodiesterase and cyclase have higher and lower FRET responses, respectively. These findings provide novel insight into the c-di-AMP distribution within bacterial populations and establish CDA5 as a powerful platform for characterizing new aspects of c-di-AMP regulation. IMPORTANCE c-di-AMP is an important nucleotide second messenger for which detection methods are severely limited. In this work we engineered and implemented a c-di-AMP-specific FRET biosensor to remedy this dearth. We present this biosensor, CDA5, as a versatile tool to investigate previously intractable facets of c-di-AMP biology.


Assuntos
Técnicas Biossensoriais , Fosfatos de Dinucleosídeos/química , Transferência Ressonante de Energia de Fluorescência , Nucleotídeos/metabolismo , Bacillus subtilis/química , Bacillus subtilis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Listeria monocytogenes/metabolismo , Modelos Moleculares , Mutação , Conformação Proteica
7.
Chembiochem ; 22(6): 1030-1041, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33142009

RESUMO

Cyclic dinucleotide signaling systems, which are found ubiquitously throughout nature, allow organisms to rapidly and dynamically sense and respond to alterations in their environments. In recent years, the second messenger, cyclic di-(3',5')-adenosine monophosphate (c-di-AMP), has been identified as an essential signaling molecule in a diverse array of bacterial genera. We and others have shown that defects in c-di-AMP homeostasis result in severe physiological defects and virulence attenuation in many bacterial species. Despite significant advancements in the field, there is still a major gap in the understanding of the environmental and cellular factors that influence c-di-AMP dynamics due to a lack of tools to sensitively and rapidly monitor changes in c-di-AMP levels. To address this limitation, we describe here the development of a luciferase-based coupled enzyme assay that leverages the cyclic nucleotide phosphodiesterase, CnpB, for the sensitive and high-throughput quantification of 3'3'-c-di-AMP. We also demonstrate the utility of this approach for the quantification of the cyclic oligonucleotide-based anti-phage signaling system (CBASS) effector, 3'3'-cGAMP. These findings establish CDA-Luc as a more affordable and sensitive alternative to conventional c-di-AMP detection tools with broad utility for the study of bacterial cyclic dinucleotide physiology.


Assuntos
3',5'-GMP Cíclico Fosfodiesterases/metabolismo , Proteínas de Bactérias/metabolismo , Fosfatos de Dinucleosídeos/análise , Ensaios Enzimáticos/métodos , Monofosfato de Adenosina/metabolismo , Bactérias/metabolismo , Fosfatos de Dinucleosídeos/química , Fosfatos de Dinucleosídeos/metabolismo , Ensaios de Triagem em Larga Escala , Hidrólise , Luciferases/metabolismo , Medições Luminescentes/métodos , Mycobacterium tuberculosis/enzimologia
8.
PLoS Genet ; 14(8): e1007574, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30074984

RESUMO

The broadly conserved bacterial signalling molecule cyclic-di-adenosine monophosphate (c-di-AMP) controls osmoresistance via its regulation of potassium (K+) and compatible solute uptake. High levels of c-di-AMP resulting from inactivation of c-di-AMP phosphodiesterase activity leads to poor growth of bacteria under high osmotic conditions. To better understand how bacteria can adjust in response to excessive c-di-AMP levels and to identify signals that feed into the c-di-AMP network, we characterised genes identified in a screen for osmoresistant suppressor mutants of the high c-di-AMP Lactococcus ΔgdpP strain. Mutations were identified which increased the uptake of osmoprotectants, including gain-of-function mutations in a Kup family K+ importer (KupB) and inactivation of the glycine betaine transporter transcriptional repressor BusR. The KupB mutations increased the intracellular K+ level while BusR inactivation increased the glycine betaine level. In addition, BusR was found to directly bind c-di-AMP and repress expression of the glycine betaine transporter in response to elevated c-di-AMP. Interestingly, overactive KupB activity or loss of BusR triggered c-di-AMP accumulation, suggesting turgor pressure changes act as a signal for this second messenger. In another group of suppressors, overexpression of an operon encoding an EmrB family multidrug resistance protein allowed cells to lower their intracellular level of c-di-AMP through active export. Lastly evidence is provided that c-di-AMP levels in several bacteria are rapidly responsive to environmental osmolarity changes. Taken together, this work provides evidence for a model in which high c-di-AMP containing cells are dehydrated due to lower K+ and compatible solute levels and that this osmoregulation system is able to sense and respond to cellular water stress.


Assuntos
Proteínas de Bactérias/fisiologia , Betaína/metabolismo , AMP Cíclico/metabolismo , Lactococcus lactis/fisiologia , Osmorregulação , Potássio/metabolismo , Monofosfato de Adenosina , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Lactococcus lactis/genética , Mutação , Óperon , Concentração Osmolar , Sistemas do Segundo Mensageiro
9.
PLoS Genet ; 14(4): e1007301, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29608558

RESUMO

The broadly conserved signaling nucleotide cyclic di-adenosine monophosphate (c-di-AMP) is essential for viability in most bacteria where it has been studied. However, characterization of the cellular functions and metabolism of c-di-AMP has largely been confined to the class Bacilli, limiting our functional understanding of the molecule among diverse phyla. We identified the cyclase responsible for c-di-AMP synthesis and characterized the molecule's role in survival of darkness in the model photosynthetic cyanobacterium Synechococcus elongatus PCC 7942. In addition to the use of traditional genetic, biochemical, and proteomic approaches, we developed a high-throughput genetic interaction screen (IRB-Seq) to determine pathways where the signaling nucleotide is active. We found that in S. elongatus c-di-AMP is produced by an enzyme of the diadenylate cyclase family, CdaA, which was previously unexplored experimentally. A cdaA-null mutant experiences increased oxidative stress and death during the nighttime portion of day-night cycles, in which potassium transport is implicated. These findings suggest that c-di-AMP is biologically active in cyanobacteria and has non-canonical roles in the phylum including oxidative stress management and day-night survival. The pipeline and analysis tools for IRB-Seq developed for this study constitute a quantitative high-throughput approach for studying genetic interactions.


Assuntos
AMP Cíclico/fisiologia , Ensaios de Triagem em Larga Escala/métodos , Synechococcus/fisiologia , Proteínas de Bactérias/metabolismo , Mutação , Estresse Oxidativo , Fósforo-Oxigênio Liases/metabolismo , Proteômica , Transdução de Sinais , Synechococcus/genética , Synechococcus/metabolismo
10.
Proc Natl Acad Sci U S A ; 114(35): E7226-E7235, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28808024

RESUMO

Cyclic di-3',5'-adenosine monophosphate (c-di-AMP) is a broadly conserved bacterial second messenger that has been implicated in a wide range of cellular processes. Our earlier studies showed that c-di-AMP regulates central metabolism in Listeria monocytogenes by inhibiting its pyruvate carboxylase (LmPC), a biotin-dependent enzyme with biotin carboxylase (BC) and carboxyltransferase (CT) activities. We report here structural, biochemical, and functional studies on the inhibition of Lactococcus lactis PC (LlPC) by c-di-AMP. The compound is bound at the dimer interface of the CT domain, at a site equivalent to that in LmPC, although it has a distinct binding mode in the LlPC complex. This binding site is not well conserved among PCs, and only a subset of these bacterial enzymes are sensitive to c-di-AMP. Conformational changes in the CT dimer induced by c-di-AMP binding may be the molecular mechanism for its inhibitory activity. Mutations of residues in the binding site can abolish c-di-AMP inhibition. In L. lactis, LlPC is required for efficient milk acidification through its essential role in aspartate biosynthesis. The aspartate pool in L. lactis is negatively regulated by c-di-AMP, and high aspartate levels can be restored by expression of a c-di-AMP-insensitive LlPC. LlPC has high intrinsic catalytic activity and is not sensitive to acetyl-CoA activation, in contrast to other PC enzymes.


Assuntos
Fosfatos de Dinucleosídeos/metabolismo , Piruvato Carboxilase/metabolismo , Piruvato Carboxilase/fisiologia , Monofosfato de Adenosina/metabolismo , Ácido Aspártico/biossíntese , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cristalografia por Raios X/métodos , AMP Cíclico/metabolismo , Fosfatos de Dinucleosídeos/fisiologia , Lactobacillales/metabolismo , Lactococcus lactis/metabolismo , Conformação Proteica , Sistemas do Segundo Mensageiro/fisiologia , Relação Estrutura-Atividade
11.
Annu Rev Med ; 68: 317-330, 2017 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-27813878

RESUMO

The best known of the naturally occurring antimalarial compounds are quinine, extracted from cinchona bark, and artemisinin (qinghao), extracted from Artemisia annua in China. These and other derivatives are now chemically synthesized and remain the mainstay of therapy to treat malaria. The beneficial effects of several of the antimalarial drugs (AMDs) on clinical features of autoimmune disorders were discovered by chance during World War II. In this review, we discuss the chemistry of AMDs and their mechanisms of action, emphasizing how they may impact multiple pathways of innate immunity. These pathways include Toll-like receptors and the recently described cGAS-STING pathway. Finally, we discuss the current and future impact of AMDs on systemic lupus erythematosus, rheumatoid arthritis, and devastating monogenic disorders (interferonopathies) characterized by expression of type I interferon in the brain.


Assuntos
Acridinas/farmacologia , Antimaláricos/farmacologia , Doenças Autoimunes/tratamento farmacológico , Imunidade Inata/efeitos dos fármacos , Imunomodulação/efeitos dos fármacos , Quinolinas/farmacologia , Acridinas/química , Animais , Antimaláricos/química , Artemisininas/farmacologia , Artrite Reumatoide/tratamento farmacológico , Autofagia/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Quinolinas/química , Transdução de Sinais , Receptores Toll-Like/metabolismo
12.
Environ Microbiol ; 21(12): 4478-4487, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31251828

RESUMO

Advances in whole-genome sequencing (WGS) technologies have documented genetic diversity and epidemiology of the major foodborne pathogen Listeria monocytogenes (Lm) in Europe and North America, but data concerning South America are scarce. Here, we examined the population structure and genetic diversity of this major foodborne pathogen collected in Brazil. Based on core genome multilocus sequence typing (cgMLST), isolates from lineages I (n = 22; 63%) and II (n = 13; 37%) were distributed into 10 different sublineages (SLs) and represented 31 new cgMLST types (CTs). The most prevalent SLs were SL9 (n = 9; 26%), SL3 (n = 6; 17%) and SL2 and SL218 (n = 5; 14%). Isolates belonging to CTs L2-SL9-ST9-CT4420 and L1-SL315-ST520-CT4429 were collected 3 and 9 years apart, respectively, revealing long-term persistence of Lm in Brazil. Genetic elements associated with stress survival were present in 60% of isolates (57% SSI-1 and 3% SSI-2). Pathogenic islands were present in 100% (LIPI-1), 43% (LIPI-3) and 6% (LIPI-4) of the isolates. Mutations leading to premature stop codons were detected in the prfA and inlA virulence genes. This study is an important contribution to understanding the genomic diversity and epidemiology of Lm in South America. In addition, the results highlight the importance of using WGS to reveal Lm long-term persistence.


Assuntos
Listeria monocytogenes/genética , Listeriose/microbiologia , Brasil/epidemiologia , Microbiologia de Alimentos , Variação Genética , Genoma Bacteriano , Humanos , Listeriose/epidemiologia , Carne/microbiologia , Tipagem de Sequências Multilocus , Virulência/genética , Sequenciamento Completo do Genoma
13.
Mol Microbiol ; 104(2): 212-233, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28097715

RESUMO

Cyclic diadenosine monophosphate (c-di-AMP) is a conserved nucleotide second messenger critical for bacterial growth and resistance to cell wall-active antibiotics. In Listeria monocytogenes, the sole diadenylate cyclase, DacA, is essential in rich, but not synthetic media and ΔdacA mutants are highly sensitive to the ß-lactam antibiotic cefuroxime. In this study, loss of function mutations in the oligopeptide importer (oppABCDF) and glycine betaine importer (gbuABC) allowed ΔdacA mutants to grow in rich medium. Since oligopeptides were sufficient to inhibit growth of the ΔdacA mutant we hypothesized that oligopeptides act as osmolytes, similar to glycine betaine, to disrupt intracellular osmotic pressure. Supplementation with salt stabilized the ΔdacA mutant in rich medium and restored cefuroxime resistance. Additional suppressor mutations in the acetyl-CoA binding site of pyruvate carboxylase (PycA) rescued cefuroxime resistance and resulted in a 100-fold increase in virulence of the ΔdacA mutant. PycA is inhibited by c-di-AMP and these mutations prompted us to examine the role of TCA cycle enzymes. Inactivation of citrate synthase, but not down-stream enzymes suppressed ΔdacA phenotypes. These data suggested that c-di-AMP modulates central metabolism at the pyruvate node to moderate citrate production and indeed, the ΔdacA mutant accumulated six times the concentration of citrate present in wild-type bacteria.


Assuntos
Fosfatos de Dinucleosídeos/metabolismo , Listeria monocytogenes/metabolismo , Acetilcoenzima A/metabolismo , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Fosfatos de Dinucleosídeos/genética , Fosfatos de Dinucleosídeos/fisiologia , Resistência Microbiana a Medicamentos , Regulação Bacteriana da Expressão Gênica/genética , Listeria monocytogenes/crescimento & desenvolvimento , Osmorregulação/fisiologia , Pressão Osmótica , Fósforo-Oxigênio Liases/metabolismo , Piruvato Carboxilase/metabolismo , Sistemas do Segundo Mensageiro , Supressão Genética
14.
Proc Natl Acad Sci U S A ; 112(7): E747-56, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25583510

RESUMO

The nucleotide cyclic di-3',5'- adenosine monophosphate (c-di-AMP) was recently identified as an essential and widespread second messenger in bacterial signaling. Among c-di-AMP-producing bacteria, altered nucleotide levels result in several physiological defects and attenuated virulence. Thus, a detailed molecular understanding of c-di-AMP metabolism is of both fundamental and practical interest. Currently, c-di-AMP degradation is recognized solely among DHH-DHHA1 domain-containing phosphodiesterases. Using chemical proteomics, we identified the Listeria monocytogenes protein PgpH as a molecular target of c-di-AMP. Biochemical and structural studies revealed that the PgpH His-Asp (HD) domain bound c-di-AMP with high affinity and specifically hydrolyzed this nucleotide to 5'-pApA. PgpH hydrolysis activity was inhibited by ppGpp, indicating a cross-talk between c-di-AMP signaling and the stringent response. Genetic analyses supported coordinated regulation of c-di-AMP levels in and out of the host. Intriguingly, a L. monocytogenes mutant that lacks c-di-AMP phosphodiesterases exhibited elevated c-di-AMP levels, hyperinduced a host type-I IFN response, and was significantly attenuated for infection. Furthermore, PgpH homologs, which belong to the 7TMR-HD family, are widespread among hundreds of c-di-AMP synthesizing microorganisms. Thus, PgpH represents a broadly conserved class of c-di-AMP phosphodiesterase with possibly other physiological functions in this crucial signaling network.


Assuntos
AMP Cíclico/metabolismo , Listeria monocytogenes/enzimologia , Diester Fosfórico Hidrolases/metabolismo , Virulência , Sequência de Aminoácidos , Hidrólise , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Modelos Moleculares , Dados de Sequência Molecular , Diester Fosfórico Hidrolases/química , Ligação Proteica
15.
Mol Microbiol ; 102(2): 233-243, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27378384

RESUMO

Cellular turgor is of fundamental importance to bacterial growth and survival. Changes in external osmolarity as a consequence of fluctuating environmental conditions and colonization of diverse environments can significantly impact cytoplasmic water content, resulting in cellular lysis or plasmolysis. To ensure maintenance of appropriate cellular turgor, bacteria import ions and small organic osmolytes, deemed compatible solutes, to equilibrate cytoplasmic osmolarity with the extracellular environment. Here, we show that elevated levels of c-di-AMP, a ubiquitous second messenger among bacteria, result in significant susceptibility to elevated osmotic stress in the bacterial pathogen Listeria monocytogenes. We found that levels of import of the compatible solute carnitine show an inverse correlation with intracellular c-di-AMP content and that c-di-AMP directly binds to the CBS domain of the ATPase subunit of the carnitine importer OpuC. Biochemical and structural studies identify conserved residues required for this interaction and transport activity in bacterial cells. Overall, these studies reveal a role for c-di-AMP mediated regulation of compatible solute import and provide new insight into the molecular mechanisms by which this essential second messenger impacts bacterial physiology and adaptation to changing environmental conditions.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Bactérias/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Listeria monocytogenes/citologia , Listeria monocytogenes/metabolismo , Monofosfato de Adenosina/metabolismo , Betaína/metabolismo , Transporte Biológico Ativo , Carnitina/metabolismo , AMP Cíclico/metabolismo , Cistationina beta-Sintase/metabolismo , Concentração Osmolar , Pressão Osmótica/fisiologia
16.
J Immunol ; 195(5): 1939-43, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26223655

RESUMO

Detection of intracellular DNA triggers activation of the stimulator of IFN genes-dependent IFN-stimulatory DNA (ISD) pathway, which is essential for antiviral immune responses. However, chronic activation of this pathway is implicated in autoimmunity. Mutations in TREX1, a 3' repair exonuclease that degrades cytosolic DNA, cause Aicardi-Goutières syndrome and chilblain lupus. Trex1 (-/-) mice develop lethal, IFN-driven autoimmune disease that is dependent on activation of the ISD pathway, but the DNA sensors that detect the endogenous DNA that accumulates in Trex1 (-/-) mice have not been defined. Multiple DNA sensors have been proposed to activate the ISD pathway, including cyclic GMP-AMP synthase (cGAS). In this study, we show that Trex1 (-/-) mice lacking cGAS are completely protected from lethality, exhibit dramatically reduced tissue inflammation, and fail to develop autoantibodies. These findings implicate cGAS as a key driver of autoimmune disease and suggest that cGAS inhibitors may be useful therapeutics for Aicardi-Goutières syndrome and related autoimmune diseases.


Assuntos
Doenças Autoimunes do Sistema Nervoso/imunologia , Exodesoxirribonucleases/imunologia , Malformações do Sistema Nervoso/imunologia , Nucleotidiltransferases/imunologia , Fosfoproteínas/imunologia , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos/citologia , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Immunoblotting , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Interferons/imunologia , Interferons/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Nucleotídeos Cíclicos/imunologia , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Immunol ; 194(9): 4089-93, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25821216

RESUMO

Type I IFN is strongly implicated in the pathogenesis of systemic autoimmune diseases, such as lupus, and rare monogenic IFNopathies, including Aicardi-Goutières syndrome. Recently, a new DNA-activated pathway involving the enzyme cyclic GMP-AMP synthase (cGAS) was described and potentially linked to Aicardi-Goutières syndrome. To identify drugs that could potentially inhibit cGAS activity, we performed in silico screening of drug libraries. By computational analysis, we identified several antimalarial drugs (AMDs) that were predicted to interact with the cGAS/dsDNA complex. Our studies validated that several AMDs were effective inhibitors of IFN-ß production and that they functioned by inhibiting dsDNA stimulation of cGAS. Because AMDs have been widely used in human diseases and have an excellent safety profile, our findings suggest new therapeutic strategies for the treatment of severe debilitating diseases associated with type I IFNs due to cGAS activation.


Assuntos
Antimaláricos/farmacologia , DNA/metabolismo , Interferon beta/biossíntese , Nucleotidiltransferases/metabolismo , Linhagem Celular , DNA/química , Humanos , Modelos Moleculares , Nucleotidiltransferases/química , Ligação Proteica/efeitos dos fármacos , Relação Estrutura-Atividade
18.
mBio ; 15(8): e0133224, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38940553

RESUMO

Transposon sequencing (Tn-seq) is a powerful genome-wide technique to assess bacterial fitness under varying growth conditions. However, screening via Tn-seq in vivo is challenging. Dose limitations and host restrictions create bottlenecks that diminish the transposon mutant pool being screened. Here, we have developed a murine model with a disruption in Akr1c13 that renders the resulting RECON-/- mouse resistant to high-dose infection. We leveraged this model to perform a Tn-seq screen of the human pathogen Listeria monocytogenes in vivo. We identified 135 genes which were required for L. monocytogenes growth in mice including novel genes not previously identified for host survival. We identified organ-specific requirements for L. monocytogenes survival and investigated the role of the folate enzyme FolD in L. monocytogenes liver pathogenesis. A mutant lacking folD was impaired for growth in murine livers by 2.5-log10 compared to wild type and failed to spread cell-to-cell in fibroblasts. In contrast, a mutant in alsR, which encodes a transcription factor that represses an operon involved in D-allose catabolism, was attenuated in both livers and spleens of mice by 4-log10 and 3-log10, respectively, but showed modest phenotypes in in vitro models. We confirmed that dysregulation of the D-allose catabolism operon is responsible for the in vivo growth defect, as deletion of the operon in the ∆alsR background rescued virulence. By undertaking an unbiased, genome-wide screen in mice, we have identified novel fitness determinants for L. monocytogenes host infection, which highlights the utility of the RECON-/- mouse model for future screening efforts. IMPORTANCE: Listeria monocytogenes is the gram-positive bacterium responsible for the food-borne disease listeriosis. Although infections with L. monocytogenes are limiting in healthy hosts, vulnerable populations, including pregnant and elderly people, can experience high rates of mortality. Thus, understanding the breadth of genetic requirements for L. monocytogenes in vivo survival will present new opportunities for treatment and prevention of listeriosis. We developed a murine model of infection using a RECON-/- mouse that is restrictive to systemic L. monocytogenes infection. We utilized this model to screen for L. monocytogenes genes required in vivo via transposon sequencing. We identified the liver-specific gene folD and a repressor, alsR, that only exhibits an in vivo growth defect. AlsR controls the expression of the D-allose operon which is a marker in diagnostic techniques to identify pathogenic Listeria. A better understanding of the role of the D-allose operon in human disease may further inform diagnostic and prevention measures.


Assuntos
Listeria monocytogenes , Listeriose , Animais , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Listeria monocytogenes/crescimento & desenvolvimento , Listeriose/microbiologia , Camundongos , Modelos Animais de Doenças , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fígado/microbiologia , Elementos de DNA Transponíveis/genética , Virulência/genética , Feminino , Aptidão Genética
19.
Sci Signal ; 17(827): eade3643, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38470955

RESUMO

Activation of the endoplasmic reticulum (ER)-resident adaptor protein STING, a component of a cytosolic DNA-sensing pathway, induces the transcription of genes encoding type I interferons (IFNs) and other proinflammatory factors. Because STING is activated at the Golgi apparatus, control of the localization and activation of STING is important in stimulating antiviral and antitumor immune responses. Through a genome-wide CRISPR interference screen, we found that STING activation required the Golgi-resident protein ACBD3, which promotes the generation of phosphatidylinositol 4-phosphate (PI4P) at the trans-Golgi network, as well as other PI4P-associated proteins. Appropriate localization and activation of STING at the Golgi apparatus required ACBD3 and the PI4P-generating kinase PI4KB. In contrast, STING activation was enhanced when the lipid-shuttling protein OSBP, which removes PI4P from the Golgi apparatus, was inhibited by the US Food and Drug Administration-approved antifungal itraconazole. The increase in the abundance of STING-activating phospholipids at the trans-Golgi network resulted in the increased production of IFN-ß and other cytokines in THP-1 cells. Furthermore, a mutant STING that could not bind to PI4P failed to traffic from the ER to the Golgi apparatus in response to a STING agonist, whereas forced relocalization of STING to PI4P-enriched areas elicited STING activation in the absence of stimulation with a STING agonist. Thus, PI4P is critical for STING activation, and manipulating PI4P abundance may therapeutically modulate STING-dependent immune responses.


Assuntos
Complexo de Golgi , Fosfolipídeos , Fosfolipídeos/metabolismo , Complexo de Golgi/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
20.
Cell Rep ; 42(8): 112805, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37467105

RESUMO

Cellular stress in the form of disrupted transcription, loss of organelle integrity, or damage to nucleic acids can elicit inflammatory responses by activating signaling cascades canonically tasked with controlling pathogen infections. These stressors must be kept in check to prevent unscheduled activation of interferon, which contributes to autoinflammation. This study examines the role of the transcription factor myocyte enhancing factor 2A (MEF2A) in setting the threshold of transcriptional stress responses to prevent R-loop accumulation. Increases in R-loops lead to the induction of interferon and inflammatory responses in a DEAD-box helicase 41 (DDX41)-, cyclic GMP-AMP synthase (cGAS)-, and stimulator of interferon genes (STING)-dependent manner. The loss of MEF2A results in the activation of ATM and RAD3-related (ATR) kinase, which is also necessary for the activation of STING. This study identifies the role of MEF2A in sustaining transcriptional homeostasis and highlights the role of ATR in positively regulating R-loop-associated inflammatory responses.


Assuntos
Nucleotidiltransferases , Transdução de Sinais , Nucleotidiltransferases/metabolismo , RNA Helicases , Interferons , Imunidade Inata
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa