Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.545
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34115982

RESUMEN

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Asunto(s)
Citomegalovirus/fisiología , Macrófagos Alveolares/virología , Animales , Presentación de Antígeno , Efecto Espectador , Ciclo Celular , Línea Celular Transformada , Reprogramación Celular , Citomegalovirus/patogenicidad , Citomegalovirus/ultraestructura , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Proteínas Fluorescentes Verdes/metabolismo , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Células Madre/patología , Replicación Viral/fisiología , Vía de Señalización Wnt
2.
Cell ; 173(5): 1231-1243.e16, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29731171

RESUMEN

Ubiquitination constitutes one of the most important signaling mechanisms in eukaryotes. Conventional ubiquitination is catalyzed by the universally conserved E1-E2-E3 three-enzyme cascade in an ATP-dependent manner. The newly identified SidE family effectors of the pathogen Legionella pneumophila ubiquitinate several human proteins by a different mechanism without engaging any of the conventional ubiquitination machinery. We now report the crystal structures of SidE alone and in complex with ubiquitin, NAD, and ADP-ribose, thereby capturing different conformations of SidE before and after ubiquitin and ligand binding. The structures of ubiquitin bound to both mART and PDE domains reveal several unique features of the two reaction steps catalyzed by SidE. Further, the structural and biochemical results demonstrate that SidE family members do not recognize specific structural folds of the substrate proteins. Our studies provide both structural explanations for the functional observations and new insights into the molecular mechanisms of this non-canonical ubiquitination machinery.


Asunto(s)
Proteínas Bacterianas/química , Legionella pneumophila/metabolismo , Hidrolasas Diéster Fosfóricas/química , Ubiquitina/química , Proteínas Bacterianas/metabolismo , Biocatálisis , Cristalografía por Rayos X , Dimerización , Hidrolasas Diéster Fosfóricas/metabolismo , Unión Proteica , Dominios Proteicos , Estructura Cuaternaria de Proteína , Ubiquitina/metabolismo , Ubiquitinación
3.
Mol Cell ; 83(1): 105-120.e5, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36538933

RESUMEN

The versatility of ubiquitination to control vast domains of eukaryotic biology is due, in part, to diversification through differently linked poly-ubiquitin chains. Deciphering signaling roles for some chain types, including those linked via K6, has been stymied by a lack of specificity among the implicated regulatory proteins. Forged through strong evolutionary pressures, pathogenic bacteria have evolved intricate mechanisms to regulate host ubiquitin during infection. Herein, we identify and characterize a deubiquitinase domain of the secreted effector LotA from Legionella pneumophila that specifically regulates K6-linked poly-ubiquitin. We demonstrate the utility of LotA for studying K6 poly-ubiquitin signals. We identify the structural basis of LotA activation and poly-ubiquitin specificity and describe an essential "adaptive" ubiquitin-binding domain. Without LotA activity during infection, the Legionella-containing vacuole becomes decorated with K6 poly-ubiquitin as well as the AAA ATPase VCP/p97/Cdc48. We propose that LotA's deubiquitinase activity guards Legionella-containing vacuole components from ubiquitin-dependent extraction.


Asunto(s)
Legionella pneumophila , Ubiquitina , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinación , Poliubiquitina/genética , Poliubiquitina/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
4.
Mol Cell ; 81(21): 4527-4539.e8, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34407442

RESUMEN

The kinase domain transfers phosphate from ATP to substrates. However, the Legionella effector SidJ adopts a kinase fold, yet catalyzes calmodulin (CaM)-dependent glutamylation to inactivate the SidE ubiquitin ligases. The structural and mechanistic basis in which the kinase domain catalyzes protein glutamylation is unknown. Here we present cryo-EM reconstructions of SidJ:CaM:SidE reaction intermediate complexes. We show that the kinase-like active site of SidJ adenylates an active-site Glu in SidE, resulting in the formation of a stable reaction intermediate complex. An insertion in the catalytic loop of the kinase domain positions the donor Glu near the acyl-adenylate for peptide bond formation. Our structural analysis led us to discover that the SidJ paralog SdjA is a glutamylase that differentially regulates the SidE ligases during Legionella infection. Our results uncover the structural and mechanistic basis in which the kinase fold catalyzes non-ribosomal amino acid ligations and reveal an unappreciated level of SidE-family regulation.


Asunto(s)
Proteínas Bacterianas/química , Pliegue de Proteína , Proteínas/química , Factores de Virulencia/química , Proteínas Bacterianas/metabolismo , Calmodulina/química , Catálisis , Dominio Catalítico , Microscopía por Crioelectrón , Legionella/enzimología , Mutagénesis , Péptidos/química , Unión Proteica , Conformación Proteica , Dominios Proteicos , Espectrometría de Fluorescencia , Ubiquitina-Proteína Ligasas/química , Factores de Virulencia/metabolismo
5.
Annu Rev Cell Dev Biol ; 30: 79-109, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25103867

RESUMEN

Pathogens use a vast number of strategies to alter host membrane dynamics. Targeting the host membrane machinery is important for the survival and pathogenesis of several extracellular, vacuolar, and cytosolic bacteria. Membrane manipulation promotes bacterial replication while suppressing host responses, allowing the bacterium to thrive in a hostile environment. This review provides a comprehensive summary of various strategies used by both extracellular and intracellular bacteria to hijack host membrane trafficking machinery. We start with mechanisms used by bacteria to alter the plasma membrane, delve into the hijacking of various vesicle trafficking pathways, and conclude by summarizing bacterial adaptation to host immune responses. Understanding bacterial manipulation of host membrane trafficking provides insights into bacterial pathogenesis and uncovers the molecular mechanisms behind various processes within a eukaryotic cell.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Membrana Celular/metabolismo , Células/microbiología , Interacciones Huésped-Patógeno/fisiología , Animales , Autofagia/fisiología , Proteínas Bacterianas/fisiología , Toxinas Bacterianas/farmacología , Transporte Biológico , Permeabilidad de la Membrana Celular , Células/ultraestructura , Citosol/microbiología , Endocitosis/fisiología , Humanos , Lisosomas/fisiología , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Fagosomas/fisiología , Transporte de Proteínas , Vacuolas/microbiología , Vacuolas/fisiología
6.
Mol Cell ; 77(1): 164-179.e6, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31732457

RESUMEN

The family of bacterial SidE enzymes catalyzes non-canonical phosphoribosyl-linked (PR) serine ubiquitination and promotes infectivity of Legionella pneumophila. Here, we describe identification of two bacterial effectors that reverse PR ubiquitination and are thus named deubiquitinases for PR ubiquitination (DUPs; DupA and DupB). Structural analyses revealed that DupA and SidE ubiquitin ligases harbor a highly homologous catalytic phosphodiesterase (PDE) domain. However, unlike SidE ubiquitin ligases, DupA displays increased affinity to PR-ubiquitinated substrates, which allows DupA to cleave PR ubiquitin from substrates. Interfering with DupA-ubiquitin binding switches its activity toward SidE-type ligase. Given the high affinity of DupA to PR-ubiquitinated substrates, we exploited a catalytically inactive DupA mutant to trap and identify more than 180 PR-ubiquitinated host proteins in Legionella-infected cells. Proteins involved in endoplasmic reticulum (ER) fragmentation and membrane recruitment to Legionella-containing vacuoles (LCV) emerged as major SidE targets. The global map of PR-ubiquitinated substrates provides critical insights into host-pathogen interactions during Legionella infection.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Serina/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Células A549 , Proteínas Bacterianas/metabolismo , Dominio Catalítico/fisiología , Línea Celular , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Humanos , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/metabolismo , Vacuolas/metabolismo
7.
Proc Natl Acad Sci U S A ; 120(33): e2303942120, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37549300

RESUMEN

Legionella pneumophila grows intracellularly within the membrane-bound Legionella-containing vacuole (LCV) established by proteins translocated via the bacterial type IV secretion system (T4SS). The Sde family, one such group of translocated proteins, catalyzes phosphoribosyl-ubiquitin (pR-Ub) modification of target substrates. Mutational loss of the entire Sde family results in small defects in intracellular growth, making it difficult to identify a clear role for this posttranslational modification in supporting the intracellular lifestyle. Therefore, mutations that aggravate the loss of sde genes and caused intracellular growth defects were identified, providing a mechanistic connection between Sde function and vacuole biogenesis. These double mutants drove the formation of LCVs that showed vacuole disintegration within 2 h of bacterial contact. Sde proteins appeared critical for blocking access of membrane-disruptive early endosomal membrane material to the vacuole, as RNAi depletion of endosomal pathway components partially restored LCV integrity. The role of Sde proteins in preventing host degradation of the LCV was limited to the earliest stages of infection. The time that Sde proteins could prevent vacuole disruption, however, was extended by deletion of sidJ, which encodes a translocated protein that inactivates Sde protein active sites. These results indicate that Sde proteins act as temporally regulated vacuole guards during the establishment of the replication niche, possibly by constructing a physical barrier that blocks access of disruptive host compartments during the earliest steps of LCV biogenesis.


Asunto(s)
Legionella pneumophila , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Vacuolas/metabolismo , Ubiquitina/metabolismo , Endosomas/metabolismo , Membranas/metabolismo , Proteínas Bacterianas/metabolismo
8.
Proc Natl Acad Sci U S A ; 120(23): e2119658120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252954

RESUMEN

Peptidoglycan is a critical component of the bacteria cell envelope. Remodeling of the peptidoglycan is required for numerous essential cellular processes and has been linked to bacterial pathogenesis. Peptidoglycan deacetylases that remove the acetyl group of the N-acetylglucosamine (NAG) subunit protect bacterial pathogens from immune recognition and digestive enzymes secreted at the site of infection. However, the full extent of this modification on bacterial physiology and pathogenesis is not known. Here, we identify a polysaccharide deacetylase of the intracellular bacterial pathogen Legionella pneumophila and define a two-tiered role for this enzyme in Legionella pathogenesis. First, NAG deacetylation is important for the proper localization and function of the Type IVb secretion system, linking peptidoglycan editing to the modulation of host cellular processes through the action of secreted virulence factors. As a consequence, the Legionella vacuole mis-traffics along the endocytic pathway to the lysosome, preventing the formation of a replication permissive compartment. Second, within the lysosome, the inability to deacetylate the peptidoglycan renders the bacteria more sensitive to lysozyme-mediated degradation, resulting in increased bacterial death. Thus, the ability to deacetylate NAG is important for bacteria to persist within host cells and in turn, Legionella virulence. Collectively, these results expand the function of peptidoglycan deacetylases in bacteria, linking peptidoglycan editing, Type IV secretion, and the intracellular fate of a bacterial pathogen.


Asunto(s)
Legionella pneumophila , Legionella , Enfermedad de los Legionarios , Humanos , Legionella pneumophila/metabolismo , Peptidoglicano/metabolismo , Vacuolas/metabolismo , Legionella/metabolismo , Lisosomas/metabolismo , Proteínas Bacterianas/metabolismo , Enfermedad de los Legionarios/microbiología
9.
Proc Natl Acad Sci U S A ; 120(36): e2308752120, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37639588

RESUMEN

The causative agent of human Q fever, Coxiella burnetii, is highly adapted to infect alveolar macrophages by inhibiting a range of host responses to infection. Despite the clinical and biological importance of this pathogen, the challenges related to genetic manipulation of both C. burnetii and macrophages have limited our knowledge of the mechanisms by which C. burnetii subverts macrophages functions. Here, we used the related bacterium Legionella pneumophila to perform a comprehensive screen of C. burnetii effectors that interfere with innate immune responses and host death using the greater wax moth Galleria mellonella and mouse bone marrow-derived macrophages. We identified MceF (Mitochondrial Coxiella effector protein F), a C. burnetii effector protein that localizes to mitochondria and contributes to host cell survival. MceF was shown to enhance mitochondrial function, delay membrane damage, and decrease mitochondrial ROS production induced by rotenone. Mechanistically, MceF recruits the host antioxidant protein Glutathione Peroxidase 4 (GPX4) to the mitochondria. The protective functions of MceF were absent in primary macrophages lacking GPX4, while overexpression of MceF in human cells protected against oxidative stress-induced cell death. C. burnetii lacking MceF was replication competent in mammalian cells but induced higher mortality in G. mellonella, indicating that MceF modulates the host response to infection. This study reveals an important C. burnetii strategy to subvert macrophage cell death and host immunity and demonstrates that modulation of the host antioxidant system is a viable strategy to promote the success of intracellular bacteria.


Asunto(s)
Antioxidantes , Coxiella , Humanos , Animales , Ratones , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Estrés Oxidativo , Muerte Celular , Mamíferos
10.
Mol Microbiol ; 121(2): 243-259, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38153189

RESUMEN

The intracellular pathogen Legionella pneumophila translocates more than 300 effector proteins into its host cells. The expression levels of the genes encoding these effectors are orchestrated by an intricate regulatory network. Here, we introduce LelA, the first L. pneumophila LysR-type transcriptional regulator of effectors. Through bioinformatic and experimental analyses, we identified the LelA target regulatory element and demonstrated that it directly activates the expression of three L. pneumophila effectors (legL7, legL6, and legU1). We further found that the gene encoding LelA is positively regulated by the RpoS sigma factor, thus linking it to the known effector regulatory network. Examination of other species throughout the Legionella genus revealed that this regulatory element is found upstream of 34 genes encoding validated effectors, putative effectors, and hypothetical proteins. Moreover, ten of these genes were examined and found to be activated by the L. pneumophila LelA as well as by their orthologs in the corresponding species. LelA represents a novel type of Legionella effector regulator, which coordinates the expression of both adjacently and distantly located effector-encoding genes, thus forming small groups of co-regulated effectors.


Asunto(s)
Legionella pneumophila , Legionella , Legionella/genética , Legionella/metabolismo , Proteínas Bacterianas/metabolismo , Legionella pneumophila/metabolismo , Factor sigma/genética , Factor sigma/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos
11.
Mol Microbiol ; 121(3): 368-384, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37891705

RESUMEN

The phenomenon of host cell escape exhibited by intracellular pathogens is a remarkably versatile occurrence, capable of unfolding through lytic or non-lytic pathways. Among these pathogens, the bacterium Legionella pneumophila stands out, having adopted a diverse spectrum of strategies to disengage from their host cells. A pivotal juncture that predates most of these host cell escape modalities is the initial escape from the intracellular compartment. This critical step is increasingly supported by evidence suggesting the involvement of several secreted pathogen effectors, including lytic proteins. In this intricate landscape, L. pneumophila emerges as a focal point for research, particularly concerning secreted phospholipases. While nestled within its replicative vacuole, the bacterium deftly employs both its type II (Lsp) and type IVB (Dot/Icm) secretion systems to convey phospholipases into either the phagosomal lumen or the host cell cytoplasm. Its repertoire encompasses numerous phospholipases A (PLA), including three enzymes-PlaA, PlaC, and PlaD-bearing the GDSL motif. Additionally, there are 11 patatin-like phospholipases A as well as PlaB. Furthermore, the bacterium harbors three extracellular phospholipases C (PLCs) and one phospholipase D. Within this comprehensive review, we undertake an exploration of the pivotal role played by phospholipases in the broader context of phagosomal and host cell egress. Moreover, we embark on a detailed journey to unravel the established and potential functions of the secreted phospholipases of L. pneumophila in orchestrating this indispensable process.


Asunto(s)
Legionella pneumophila , Enfermedad de los Legionarios , Humanos , Fosfolipasas/metabolismo , Enfermedad de los Legionarios/microbiología , Vacuolas/metabolismo , Proteínas Bacterianas/metabolismo , Legionella pneumophila/metabolismo , Fosfolipasas A/metabolismo
12.
Mol Microbiol ; 121(3): 497-512, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38130174

RESUMEN

Legionella pneumophila, the causative agent of a life-threatening pneumonia, intracellularly replicates in a specialized compartment in lung macrophages, the Legionella-containing vacuole (LCV). Secreted proteins of the pathogen govern important steps in the intracellular life cycle including bacterial egress. Among these is the type II secreted PlaA which, together with PlaC and PlaD, belongs to the GDSL phospholipase family found in L. pneumophila. PlaA shows lysophospholipase A (LPLA) activity which increases after secretion and subsequent processing by the zinc metalloproteinase ProA within a disulfide loop. Activity of PlaA contributes to the destabilization of the LCV in the absence of the type IVB-secreted effector SdhA. We here present the 3D structure of PlaA which shows a typical α/ß-hydrolase fold and reveals that the uncleaved disulfide loop forms a lid structure covering the catalytic triad S30/D278/H282. This leads to reduction of substrate access before activation; however, the catalytic site gets more accessible when the disulfide loop is processed. After structural modeling, a similar activation process is suggested for the GDSL hydrolase PlaC, but not for PlaD. Furthermore, the size of the PlaA substrate-binding site indicated preference toward phospholipids comprising ~16 carbon fatty acid residues which was verified by lipid hydrolysis, suggesting a molecular ruler mechanism. Indeed, mutational analysis changed the substrate profile with respect to fatty acid chain length. In conclusion, our analysis revealed the structural basis for the regulated activation and substrate preference of PlaA.


Asunto(s)
Legionella pneumophila , Lisofosfolipasa , Lisofosfolipasa/genética , Lisofosfolipasa/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Proteínas Bacterianas/metabolismo , Disulfuros/metabolismo , Vacuolas/metabolismo , Ácidos Grasos/metabolismo , Relación Estructura-Actividad
13.
Mol Microbiol ; 121(2): 167-195, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37908155

RESUMEN

Legionella pneumophila is a gram-negative bacteria found in natural and anthropogenic aquatic environments such as evaporative cooling towers, where it reproduces as an intracellular parasite of cohabiting protozoa. If L. pneumophila is aerosolized and inhaled by a susceptible person, bacteria may colonize their alveolar macrophages causing the opportunistic pneumonia Legionnaires' disease. L. pneumophila utilizes an elaborate regulatory network to control virulence processes such as the Dot/Icm Type IV secretion system and effector repertoire, responding to changing nutritional cues as their host becomes depleted. The bacteria subsequently differentiate to a transmissive state that can survive in the environment until a replacement host is encountered and colonized. In this review, we discuss the lifecycle of L. pneumophila and the molecular regulatory network that senses nutritional depletion via the stringent response, a link to stationary phase-like metabolic changes via alternative sigma factors, and two-component systems that are homologous to stress sensors in other pathogens, to regulate differentiation between the intracellular replicative phase and more transmissible states. Together, we highlight how this prototypic intracellular pathogen offers enormous potential in understanding how molecular mechanisms enable intracellular parasitism and pathogenicity.


Asunto(s)
Legionella pneumophila , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Virulencia , Factor sigma/metabolismo , Proteínas Bacterianas/metabolismo
14.
Cell Mol Life Sci ; 81(1): 249, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38836877

RESUMEN

Protein ubiquitination is one of the most important posttranslational modifications (PTMs) in eukaryotes and is involved in the regulation of almost all cellular signaling pathways. The intracellular bacterial pathogen Legionella pneumophila translocates at least 26 effectors to hijack host ubiquitination signaling via distinct mechanisms. Among these effectors, SidC/SdcA are novel E3 ubiquitin ligases with the adoption of a Cys-His-Asp catalytic triad. SidC/SdcA are critical for the recruitment of endoplasmic reticulum (ER)-derived vesicles to the Legionella-containing vacuole (LCV). However, the ubiquitination targets of SidC/SdcA are largely unknown, which restricts our understanding of the mechanisms used by these effectors to hijack the vesicle trafficking pathway. Here, we demonstrated that multiple Rab small GTPases and target soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) proteins are bona fide ubiquitination substrates of SidC/SdcA. SidC/SdcA-mediated ubiquitination of syntaxin 3 and syntaxin 4 promotes their unconventional pairing with the vesicle-SNARE protein Sec22b, thereby contributing to the membrane fusion of ER-derived vesicles with the phagosome. In addition, our data reveal that ubiquitination of Rab7 by SidC/SdcA is critical for its association with the LCV membrane. Rab7 ubiquitination could impair its binding with the downstream effector Rab-interacting lysosomal protein (RILP), which partially explains why LCVs avoid fusion with lysosomes despite the acquisition of Rab7. Taken together, our study reveals the biological mechanisms employed by SidC/SdcA to promote the maturation of the LCVs.


Asunto(s)
Legionella pneumophila , Fagosomas , Proteínas SNARE , Ubiquitinación , Proteínas de Unión al GTP rab , Legionella pneumophila/metabolismo , Humanos , Fagosomas/metabolismo , Fagosomas/microbiología , Proteínas SNARE/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Animales , Proteínas Qa-SNARE/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Vacuolas/metabolismo , Vacuolas/microbiología , Células HEK293 , Ratones , Proteínas de Unión a GTP rab7/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Retículo Endoplásmico/metabolismo
15.
Proc Natl Acad Sci U S A ; 119(23): e2122872119, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35653564

RESUMEN

Adenosine diphosphate (ADP) ribosylation is a reversible posttranslational modification involved in the regulation of numerous cellular processes. Prototype ADP ribosyltransferases (ARTs) from many pathogenic bacteria are known to function as toxins, while other bacterial ARTs have just recently emerged. Recent studies have shown that bacteria also possess enzymes that function as poly-ADP ribose (ADPr) glycohydrolases (PARGs), which reverse poly-ADP ribosylation. However, how bacteria manipulate host target proteins by coordinated reactions of ARTs and ADPr hydrolases (ARHs) remains elusive. The intracellular bacterial pathogen Legionella pneumophila, the causative agent of Legionnaires' disease, transports a large array of effector proteins via the Dot/Icm type IV secretion system to host cells. The effector proteins, which mostly function as enzymes, modulate host cellular processes for the bacteria's benefit. In this study, we identified a pair of L. pneumophila effector proteins, Lpg0080 and Lpg0081, which function as an ART and an ARH, respectively. The two proteins were shown to coordinately modulate mitochondrial ADP/adenosine triphosphate (ATP) translocases (ANTs) by their enzymatic activities to conjugate ADPr to, and remove it from, a key arginine residue. The crystal structures of Lpg0081 and the Lpg0081:ADPr complex indicated that Lpg0081 is a macroD-type ARH with a noncanonical macrodomain, whose folding topology is strikingly distinct from that of the canonical macrodomain that is ubiquitously found in eukaryotic PARGs and ARHs. Our results illustrate that L. pneumophila has acquired an effector pair that coordinately manipulate mitochondrial activity via reversible chemical modification of ANTs.


Asunto(s)
Legionella pneumophila , Legionella , Adenosina Difosfato , Adenosina Trifosfato , Proteínas Bacterianas , Mitocondrias/fisiología , Translocasas Mitocondriales de ADP y ATP
16.
Proc Natl Acad Sci U S A ; 119(48): e2209149119, 2022 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-36413498

RESUMEN

Intravacuolar pathogens need to gradually expand their surrounding vacuole to accommodate the growing number of bacterial offspring during intracellular replication. Here we found that Legionella pneumophila controls vacuole expansion by fine-tuning the generation of lysophospholipids within the vacuolar membrane. Upon allosteric activation by binding to host ubiquitin, the type IVB (Dot/Icm) effector VpdC converts phospholipids into lysophospholipids which, at moderate concentrations, are known to promote membrane fusion but block it at elevated levels by generating excessive positive membrane curvature. Consequently, L. pneumophila overproducing VpdC were prevented from adequately expanding their surrounding membrane, trapping the replicating bacteria within spatially confined vacuoles and reducing their capability to proliferate intracellularly. Quantitative lipidomics confirmed a VpdC-dependent increase in several types of lysophospholipids during infection, and VpdC production in transiently transfected cells caused tubulation of organelle membranes as well as mitochondria fragmentation, processes that can be phenocopied by supplying cells with exogenous lysophospholipids. Together, these results demonstrate an important role for bacterial phospholipases in vacuolar expansion.


Asunto(s)
Legionella , Enfermedad de los Legionarios , Humanos , Legionella/metabolismo , Vacuolas/metabolismo , Enfermedad de los Legionarios/microbiología , Fosfolipasas/metabolismo , Ubiquitina/metabolismo , Proteínas Bacterianas/metabolismo , Lisofosfolípidos/metabolismo
17.
J Bacteriol ; 206(4): e0043323, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38493438

RESUMEN

In bacteria, disulfide bonds contribute to the folding and stability of proteins important for processes in the cellular envelope. In Escherichia coli, disulfide bond formation is catalyzed by DsbA and DsbB enzymes. DsbA is a periplasmic protein that catalyzes disulfide bond formation in substrate proteins, while DsbB is an inner membrane protein that transfers electrons from DsbA to quinones, thereby regenerating the DsbA active state. Actinobacteria including mycobacteria use an alternative enzyme named VKOR, which performs the same function as DsbB. Disulfide bond formation enzymes, DsbA and DsbB/VKOR, represent novel drug targets because their inhibition could simultaneously affect the folding of several cell envelope proteins including virulence factors, proteins involved in outer membrane biogenesis, cell division, and antibiotic resistance. We have previously developed a cell-based and target-based assay to identify molecules that inhibit the DsbB and VKOR in pathogenic bacteria, using E. coli cells expressing a periplasmic ß-Galactosidase sensor (ß-Galdbs), which is only active when disulfide bond formation is inhibited. Here, we report the construction of plasmids that allows fine-tuning of the expression of the ß-Galdbs sensor and can be mobilized into other gram-negative organisms. As an example, when expressed in Pseudomonas aeruginosa UCBPP-PA14, which harbors two DsbB homologs, ß-Galdbs behaves similarly as in E. coli, and the biosensor responds to the inhibition of the two DsbB proteins. Thus, these ß-Galdbs reporter plasmids provide a basis to identify novel inhibitors of DsbA and DsbB/VKOR in multidrug-resistant gram-negative pathogens and to further study oxidative protein folding in diverse gram-negative bacteria. IMPORTANCE: Disulfide bonds contribute to the folding and stability of proteins in the bacterial cell envelope. Disulfide bond-forming enzymes represent new drug targets against multidrug-resistant bacteria because inactivation of this process would simultaneously affect several proteins in the cell envelope, including virulence factors, toxins, proteins involved in outer membrane biogenesis, cell division, and antibiotic resistance. Identifying the enzymes involved in disulfide bond formation in gram-negative pathogens as well as their inhibitors can contribute to the much-needed antibacterial innovation. In this work, we developed sensors of disulfide bond formation for gram-negative bacteria. These tools will enable the study of disulfide bond formation and the identification of inhibitors for this crucial process in diverse gram-negative pathogens.


Asunto(s)
Proteínas Bacterianas , Escherichia coli , Proteínas Bacterianas/metabolismo , Escherichia coli/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Factores de Virulencia/metabolismo , Disulfuros/química , Oxidación-Reducción
18.
Traffic ; 23(8): 414-425, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35701729

RESUMEN

Many intracellular pathogens, such as bacteria and large viruses, enter eukaryotic cells via phagocytosis, then replicate and proliferate inside the host. To avoid degradation in the phagosomes, they have developed strategies to modify vesicle trafficking. Although several strategies of bacteria have been characterized, it is not clear whether viruses also interfere with the vesicle trafficking of the host. Recently, we came across SNARE proteins encoded in the genomes of several bacteria of the order Legionellales. These pathogenic bacteria may use SNAREs to interfere with vesicle trafficking, since SNARE proteins are the core machinery for vesicle fusion during transport. They assemble into membrane-bridging SNARE complexes that bring membranes together. We now have also discovered SNARE proteins in the genomes of diverse giant viruses. Our biochemical experiments showed that these proteins are able to form SNARE complexes. We also found other key trafficking factors that work together with SNAREs such as NSF, SM, and Rab proteins encoded in the genomes of giant viruses, suggesting that viruses can make use of a large genetic repertoire of trafficking factors. Most giant viruses possess different collections, suggesting that these factors entered the viral genome multiple times. In the future, the molecular role of these factors during viral infection need to be studied.


Asunto(s)
Eucariontes , Células Eucariotas , Eucariontes/metabolismo , Células Eucariotas/metabolismo , Fusión de Membrana , Fagosomas/metabolismo , Proteínas SNARE/metabolismo
19.
Semin Cell Dev Biol ; 132: 230-241, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35177348

RESUMEN

Legionella pneumophila, a Gram-negative intracellular bacterium, is one of the major causes of Legionnaires' disease, a specific type of atypical pneumonia. Despite intensive research efforts that elucidated many relevant structural, molecular and medical insights into Legionella's pathogenicity, Legionnaires' disease continues to present an ongoing public health concern. Legionella's virulence is based on its ability to simultaneously hijack multiple molecular pathways of the host cell to ensure its fast replication and dissemination. Legionella usurps the host ubiquitin system through multiple effector proteins, using the advantage of both conventional and unconventional (phosphoribosyl-linked) ubiquitination, thus providing optimal conditions for its replication. In this review, we summarize the current understanding of L. pneumophila from medical, biochemical and molecular perspectives. We describe the clinical disease presentation, its diagnostics and treatment, as well as host-pathogen interactions, with the emphasis on the ability of Legionella to target the host ubiquitin system upon infection. Furthermore, the interdisciplinary use of innovative technologies enables better insights into the pathogenesis of Legionnaires' disease and provides new opportunities for its treatment and prevention.


Asunto(s)
Legionella pneumophila , Enfermedad de los Legionarios , Ubiquitina , Humanos , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Ubiquitina/metabolismo
20.
J Biol Chem ; 299(12): 105376, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37866633

RESUMEN

Legionella pneumophila is an environmental bacterium, which replicates in amoeba but also in macrophages, and causes a life-threatening pneumonia called Legionnaires' disease. The opportunistic pathogen employs the α-hydroxy-ketone compound Legionella autoinducer-1 (LAI-1) for intraspecies and interkingdom signaling. LAI-1 is produced by the autoinducer synthase Legionella quorum sensing A (LqsA), but it is not known, how LAI-1 is released by the pathogen. Here, we use a Vibrio cholerae luminescence reporter strain and liquid chromatography-tandem mass spectrometry to detect bacteria-produced and synthetic LAI-1. Ectopic production of LqsA in Escherichia coli generated LAI-1, which partitions to outer membrane vesicles (OMVs) and increases OMV size. These E. coli OMVs trigger luminescence of the V. cholerae reporter strain and inhibit the migration of Dictyostelium discoideum amoeba. Overexpression of lqsA in L.pneumophila under the control of strong stationary phase promoters (PflaA or P6SRNA), but not under control of its endogenous promoter (PlqsA), produces LAI-1, which is detected in purified OMVs. These L. pneumophila OMVs trigger luminescence of the Vibrio reporter strain and inhibit D. discoideum migration. L. pneumophila OMVs are smaller upon overexpression of lqsA or upon addition of LAI-1 to growing bacteria, and therefore, LqsA affects OMV production. The overexpression of lqsA but not a catalytically inactive mutant promotes intracellular replication of L. pneumophila in macrophages, indicating that intracellularly produced LA1-1 modulates the interaction in favor of the pathogen. Taken together, we provide evidence that L. pneumophila LAI-1 is secreted through OMVs and promotes interbacterial communication and interactions with eukaryotic host cells.


Asunto(s)
Legionella pneumophila , Percepción de Quorum , Humanos , Proteínas Bacterianas/genética , Dictyostelium , Escherichia coli , Legionella , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA