Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
1.
Arch Biochem Biophys ; 745: 109704, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37527700

RESUMEN

Sodium dodecyl sulfate (SDS) is a well-known protein denaturing agent. A less known property of this detergent is that it can activate or inactivate some enzymes at sub-denaturing concentrations. In this work we explore the effect of SDS on the ATPase activity of a hyper-thermophilic and a mesophilic Cu(I) ATPases reconstituted in mixed micelles of phospholipids and a non-denaturing detergent. An iterative procedure was used to evaluate the partition of SDS between the aqueous and the micellar phases, allowing to determine the composition of micelles prepared from phospholipid/detergent mixtures. The incubation of enzymes with SDS in the presence of different amounts of phospholipids reveals that higher SDS concentrations are required to obtain the same degree of inactivation when the initial concentration of phospholipids is increased. Remarkably, we found that, if represented as a function of the mole fraction of SDS in the micelle, the degree of inactivation obtained at different amounts of amphiphiles converges to a single inactivation curve. To interpret this result, we propose a simple model involving active and inactive enzyme molecules in equilibrium. This model allowed us to estimate the Gibbs free energy change for the inactivation process and its derivative with respect to the mole fraction of SDS in the micellar phase, the latter being a measure of the susceptibility of the enzyme to SDS. Our results showed that the inactivation free energy changes are similar for both proteins. Conversely, susceptibility to SDS is significantly lower for the hyperthermophilic ATPase, suggesting an inverse relation between thermophilicity and susceptibility to SDS.


Asunto(s)
Adenosina Trifosfatasas , Biocatálisis , Cobre , Detergentes , Micelas , Dodecil Sulfato de Sodio , Adenosina Trifosfatasas/metabolismo , Archaeoglobus fulgidus/enzimología , Biocatálisis/efectos de los fármacos , Calorimetría , Cobre/metabolismo , Detergentes/farmacología , Hidrólisis/efectos de los fármacos , Legionella pneumophila/enzimología , Dodecil Sulfato de Sodio/farmacología , Temperatura , Termodinámica
2.
J Biol Chem ; 298(10): 102414, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36007613

RESUMEN

Legionella pneumophila, a bacterial pathogen that causes a severe pneumonia known as Legionnaires' disease, extensively exploits the ubiquitin (Ub) pathway in the infected host cells through certain virulence effectors excreted by the Dot/Icm system. To date, several Dot/Icm effectors have been found to act as Ub ligases, and four effectors, including LotA, LotB, LotC, and Ceg7, have been identified as deubiquitinases (DUBs) from the ovarian tumor (OTU) domain family. LotA is unique among other OTU DUBs because it possesses two distinct DUB domains and exclusively exhibits catalytic activity against K6-linked diUb and polyUb chains. However, the structure of LotA and the molecular mechanism for the dual DUB activity remains elusive. In this study, we solved the structure of LotA in complex with proximally bound Ub and distal covalently bound Ub. Both Ub molecules are bound to the DUB1 domain and mimic a K6-linked diUb. Structural analysis reveals that the DUB1 domain utilizes a distinct mechanism for recognition of the K6-linked diUb within a large S1' binding site that is uncommon to OTU DUBs. Structural fold of the LotA DUB2 domain closely resembles LotB and LotC, similarly containing an extra α-helix lobe that has been demonstrated to play an important role in Ub binding. Collectively, our study uncovers the structural basis for the dual catalytic activity of the unique OTU family DUB LotA.


Asunto(s)
Proteínas Bacterianas , Enzimas Desubicuitinizantes , Legionella pneumophila , Proteínas Bacterianas/química , Enzimas Desubicuitinizantes/química , Legionella pneumophila/enzimología , Ubiquitina/metabolismo , Catálisis , Dominios Proteicos , Conformación Proteica en Hélice alfa
3.
J Bacteriol ; 204(1): e0037621, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34633867

RESUMEN

Pathogenic bacteria have acquired a vast array of eukaryotic-protein-like proteins via intimate interaction with host cells. Bacterial effector proteins that function as ubiquitin ligases and deubiquitinases (DUBs) are remarkable examples of such molecular mimicry. LotA, a Legionella pneumophila effector, belongs to the ovarian tumor (OTU) superfamily, which regulates diverse ubiquitin signals by their DUB activities. LotA harbors two OTU domains that have distinct reactivities; the first one is responsible for the cleavage of the K6-linked ubiquitin chain, and the second one shows an uncommon preference for long chains of ubiquitin. Here, we report the crystal structure of a middle domain of LotA (LotAM), which contains the second OTU domain. LotAM consists of two distinct subdomains, a catalytic domain having high structural similarity with human OTU DUBs and an extended helical lobe (EHL) domain, which is characteristically conserved only in Legionella OTU DUBs. The docking simulation of LotAM with ubiquitin suggested that hydrophobic and electrostatic interactions between the EHL of LotAM and the C-terminal region of ubiquitin are crucial for the binding of ubiquitin to LotAM. The structure-based mutagenesis demonstrated that the acidic residue in the characteristic short helical segment termed the "helical arm" is essential for the enzymatic activity of LotAM. The EHL domain of the three Legionella OTU DUBs, LotA, LotB, and LotC, share the "helical arm" structure, suggesting that the EHL domain defines the Lot-OTUs as a unique class of DUBs. IMPORTANCE To successfully colonize, some pathogenic bacteria hijack the host ubiquitin system. Legionella OTU-like-DUBs (Lot-DUBs) are novel bacterial deubiquitinases found in effector proteins of L. pneumophila. LotA is a member of Lot-DUBs and has two OTU domains (OTU1 and OTU2). We determined the structure of a middle fragment of LotA (LotAM), which includes OTU2. LotAM consists of the conserved catalytic domain and the Legionella OTUs-specific EHL domain. The docking simulation with ubiquitin and the mutational analysis suggested that the acidic surface in the EHL is essential for enzymatic activity. The structure of the EHL differs from those of other Lot-DUBs, suggesting that the variation of the EHL is related to the variable cleaving specificity of each DUB.


Asunto(s)
Proteínas Bacterianas/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Legionella pneumophila/enzimología , Ubiquitina/metabolismo , Proteínas Bacterianas/genética , Cristalización , Enzimas Desubicuitinizantes/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
4.
Biochim Biophys Acta Biomembr ; 1864(2): 183822, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34826402

RESUMEN

Cu+-ATPases are integral membrane proteins belonging to the IB subfamily of the P-type ATPases that couple Cu+ transport to the hydrolysis of ATP. As some structural and functional particularities arise for Cu+-ATPases, several authors suggest that some of the reaction steps of the Albers-Post model postulated for other P-ATPases may be different. In this work we describe a functional characterization of Legionella pneumophila Cu+-ATPase (LpCopA), the first PIB-ATPase whose structure was determined by X-ray crystallography. Cu+-ATPase activity of the enzyme presents a maximum at ∼37 °C and pH 6.6-6.8. Phospholipids enhance LpCopA Cu+-ATPase activity in a non-essential mode where optimal activity is achieved at an asolectin molar fraction of 0.15 and an amphiphile-protein ratio of ~30,000. As described for other P-ATPases, Mg2+ acts as an essential activator. Furthermore, Cu+-ATPase activity dependence on [Cu+] and [ATP] can both be described by a sum of two hyperbolic functions. Based on that, and the [Cu+] and [ATP] dependencies of the best fitting parameters of the hyperbolae pointed above, we propose a minimal reaction scheme for the catalytic mechanism that shares the basic reaction steps of the Albers-Post model for P-type ATPases. The reaction scheme postulated contemplates two different binding affinities for a single ATP (apparent affinities of 0.66 and 550 µM at [Cu+] â†’ ∞) and binding of at least 2 Cu+ with different affinities as well (apparent affinities of 1.4 and 102.5 µM at [ATP] â†’ ∞).


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Legionella pneumophila/enzimología , Transporte Iónico , Cinética , Modelos Moleculares , Unión Proteica
5.
Biomolecules ; 11(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34944446

RESUMEN

Legionella pneumophila is a Gram-negative intracellular pathogen that causes Legionnaires' disease in elderly or immunocompromised individuals. This bacterium relies on the Dot/Icm (Defective in organelle trafficking/Intracellular multiplication) Type IV Secretion System (T4SS) and a large (>330) set of effector proteins to colonize the host cell. The structural variability of these effectors allows them to disrupt many host processes. Herein, we report the crystal structure of MavL to 2.65 Å resolution. MavL adopts an ADP-ribosyltransferase (ART) fold and contains the distinctive ligand-binding cleft of ART proteins. Indeed, MavL binds ADP-ribose with Kd of 13 µM. Structural overlay of MavL with poly-(ADP-ribose) glycohydrolases (PARGs) revealed a pair of aspartate residues in MavL that align with the catalytic glutamates in PARGs. MavL also aligns with ADP-ribose "reader" proteins (proteins that recognize ADP-ribose). Since no glycohydrolase activity was observed when incubated in the presence of ADP-ribosylated PARP1, MavL may play a role as a signaling protein that binds ADP-ribose. An interaction between MavL and the mammalian ubiquitin-conjugating enzyme UBE2Q1 was revealed by yeast two-hybrid and co-immunoprecipitation experiments. This work provides structural and molecular insights to guide biochemical studies aimed at elucidating the function of MavL. Our findings support the notion that ubiquitination and ADP-ribosylation are global modifications exploited by L. pneumophila.


Asunto(s)
Legionella pneumophila/crecimiento & desarrollo , Sistemas de Secreción Tipo IV/química , Sistemas de Secreción Tipo IV/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/enzimología , Modelos Moleculares , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Células THP-1 , Ubiquitinación
6.
mBio ; 12(5): e0231621, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34488448

RESUMEN

Legionella pneumophila promotes its survival and replication in phagocytes by actively modulating cellular processes using effectors injected into host cells by its Dot/Icm type IV secretion system. Many of these effectors function to manipulate the ubiquitin network of infected cells, thus contributing to the biogenesis of the Legionella-containing vacuole (LCV), which is permissive for bacterial replication. Among these, members of the SidE effector family (SidEs) catalyze ubiquitination of functionally diverse host proteins by a mechanism that is chemically distinct from the canonical three-enzyme cascade. The activity of SidEs is regulated by two mechanisms: reversal of the phosphoribosyl ubiquitination by DupA and DupB and direct inactivation by SidJ, which is a calmodulin-dependent glutamylase. In many L. pneumophila strains, SidJ belongs to a two-member protein family. Its homolog SdjA appears to function differently from SidJ despite the high-level similarity in their primary sequences. Here, we found that SdjA is a bifunctional enzyme that exhibits distinct activities toward members of the SidE family. It inhibits the activity of SdeB and SdeC by glutamylation. Unexpectedly, it also functions as a deglutamylase that reverses SidJ-induced glutamylation on SdeA. Our results reveal that an enzyme can catalyze two completely opposite biochemical reactions, which highlights the distinct regulation of phosphoribosyl ubiquitination by the SidJ effector family. IMPORTANCE One unique feature of L. pneumophila Dot/Icm effectors is the existence of protein families with members of high-level similarity. Whereas members of some families are functionally redundant, as suggested by their primary sequences, the relationship between SidJ and SdjA, the two members of the SidJ family, has remained mysterious. Despite their sharing 57% identity, sdjA cannot complement the defects in virulence displayed by a mutant lacking sidJ. SidJ inhibits the activity of the SidE family by a calmodulin (CaM)-dependent glutamylase activity. Here, we found that SdjA is a dual function protein: it is a CaM-dependent glutamylase against SdeB and SdeC but exhibits deglutamylase activity toward SdeA that has been modified by SidJ, indicating that SdjA functions to fine-tune the activity of SidEs. These findings have paved the way for future structural and functional analysis of SdjA, which may reveal novel mechanism for isopeptide bond cleavage and provide insights into the study of protein evolution.


Asunto(s)
Proteínas Bacterianas/metabolismo , Legionella pneumophila/enzimología , Enfermedad de los Legionarios/microbiología , Proteínas Bacterianas/genética , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ácidos Fosfóricos/metabolismo , Ubiquitinación , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074754

RESUMEN

The virulence factor PlaB promotes lung colonization, tissue destruction, and intracellular replication of Legionella pneumophila, the causative agent of Legionnaires' disease. It is a highly active phospholipase exposed at the bacterial surface and shows an extraordinary activation mechanism by tetramer deoligomerization. To unravel the molecular basis for enzyme activation and localization, we determined the crystal structure of PlaB in its tetrameric form. We found that the tetramer is a dimer of identical dimers, and a monomer consists of an N-terminal α/ß-hydrolase domain expanded by two noncanonical two-stranded ß-sheets, ß-6/ß-7 and ß-9/ß-10. The C-terminal domain reveals a fold displaying a bilobed ß-sandwich with a hook structure required for dimer formation and structural complementation of the enzymatic domain in the neighboring monomer. This highlights the dimer as the active form. Δß-9/ß-10 mutants showed a decrease in the tetrameric fraction and altered activity profiles. The variant also revealed restricted binding to membranes resulting in mislocalization and bacterial lysis. Unexpectedly, we observed eight NAD(H) molecules at the dimer/dimer interface, suggesting that these molecules stabilize the tetramer and hence lead to enzyme inactivation. Indeed, addition of NAD(H) increased the fraction of the tetramer and concomitantly reduced activity. Together, these data reveal structural elements and an unprecedented NAD(H)-mediated tetramerization mechanism required for spatial and enzymatic control of a phospholipase virulence factor. The allosteric regulatory process identified here is suited to fine tune PlaB in a way that protects Legionella pneumophila from self-inflicted lysis while ensuring its activity at the pathogen-host interface.


Asunto(s)
Proteínas Bacterianas/química , Legionella pneumophila/enzimología , NAD/química , Fosfolipasas/química , Multimerización de Proteína , Proteínas Bacterianas/genética , Cristalografía por Rayos X , Legionella pneumophila/genética , NAD/genética , Fosfolipasas/genética , Conformación Proteica en Lámina beta , Estructura Cuaternaria de Proteína
8.
Science ; 372(6545): 935-941, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33927055

RESUMEN

During infection, intracellular bacterial pathogens translocate a variety of effectors into host cells that modify host membrane trafficking for their benefit. We found a self-organizing system consisting of a bacterial phosphoinositide kinase and its opposing phosphatase that formed spatiotemporal patterns, including traveling waves, to remodel host cellular membranes. The Legionella effector MavQ, a phosphatidylinositol (PI) 3-kinase, was targeted to the endoplasmic reticulum (ER). MavQ and the Legionella PI 3-phosphatase SidP, even in the absence of other bacterial components, drove rapid PI 3-phosphate turnover on the ER and spontaneously formed traveling waves that spread along ER subdomains inducing vesicle and tubule budding. Thus, bacteria can exploit a self-organizing membrane-targeting mechanism to hijack host cellular structures for survival.


Asunto(s)
Proteínas Bacterianas/metabolismo , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Legionella pneumophila/fisiología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Proteínas Bacterianas/química , Células COS , Chlorocebus aethiops , Retículo Endoplásmico/ultraestructura , Retroalimentación Fisiológica , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Membranas Intracelulares/ultraestructura , Legionella pneumophila/enzimología , Legionella pneumophila/genética , Legionella pneumophila/crecimiento & desarrollo , Ratones , Mutación , Fosfatidilinositol 3-Quinasa/química , Fosfatos de Fosfatidilinositol/química , Monoéster Fosfórico Hidrolasas/metabolismo , Dominios Proteicos , Células RAW 264.7
9.
Protein Sci ; 30(5): 940-955, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33660322

RESUMEN

Legionella pneumophila is an intracellular pathogen that causes Legionnaire's disease in humans. This bacterium can be found in freshwater environments as a free-living organism, but it is also an intracellular parasite of protozoa. Human infection occurs when inhaled aerosolized pathogen comes into contact with the alveolar mucosa and replicates in alveolar macrophages. Legionella enters the host cell by phagocytosis and redirects the Legionella-containing phagosomes from the phagocytic maturation pathway. These nascent phagosomes fuse with ER-derived secretory vesicles and membranes forming the Legionella-containing vacuole. Legionella subverts many host cellular processes by secreting over 300 effector proteins into the host cell via the Dot/Icm type IV secretion system. The cellular function for many Dot/Icm effectors is still unknown. Here, we present a structural and functional study of L. pneumophila effector RavA (Lpg0008). Structural analysis revealed that the RavA consists of four ~85 residue long α-helical domains with similar folds, which show only a low level of structural similarity to other protein domains. The ~90 residues long C-terminal segment is predicted to be natively unfolded. We show that during L. pneumophila infection of human cells, RavA localizes to the Golgi apparatus and to the plasma membrane. The same localization is observed when RavA is expressed in human cells. The localization signal resides within the C-terminal sequence C409 WTSFCGLF417 . Yeast-two-hybrid screen using RavA as bait identified RAB11A as a potential binding partner. RavA is present in L. pneumophila strains but only distant homologs are found in other Legionella species, where the number of repeats varies.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Legionella pneumophila/enzimología , Adenosina Trifosfatasas/genética , Proteínas Bacterianas/genética , Células HEK293 , Humanos , Legionella pneumophila/genética , Conformación Proteica en Hélice alfa , Dominios Proteicos , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
10.
Biochem Biophys Res Commun ; 540: 90-94, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33450485

RESUMEN

MapA is a histidine acid phosphatase (HAP) from Legionella pneumophila that catalyzes the hydroxylation of a phosphoryl group from phosphomonoesters by an active-site histidine. Several structures of HAPs, including MapA, in complex with the inhibitor tartrate have been solved and the substrate binding tunnel identified; however, the substrate recognition mechanism remains unknown. To gain insight into the mechanism of substrate recognition, the crystal structures of apo-MapA and the MapAD281A mutant in complex with 5'-AMP were solved at 2.2 and 2.6 Å resolution, respectively. The structure of the MapAD281A/5'-AMP complex reveals that the 5'-AMP fits fully into the substrate binding tunnel, with the 2'-hydroxyl group of the ribose moiety stabilized by Glu201 and the adenine moiety sandwiched between His205 and Phe237. This is the second structure of a HAP/AMP complex solved with 5'-AMP binding in a unique manner in the active site. The structure presents a new substrate recognition mechanism of HAPs.


Asunto(s)
Fosfatasa Ácida/química , Fosfatasa Ácida/metabolismo , Histidina/metabolismo , Legionella pneumophila/enzimología , Fosfatasa Ácida/genética , Adenina/metabolismo , Secuencia de Aminoácidos , Apoenzimas/metabolismo , Dominio Catalítico , Legionella pneumophila/genética , Modelos Moleculares , Mutación , Fenilalanina/metabolismo , Unión Proteica , Ribosa/metabolismo , Alineación de Secuencia , Especificidad por Sustrato , Tartratos/metabolismo
11.
Cell Microbiol ; 23(5): e13313, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33491325

RESUMEN

ProA is a secreted zinc metalloprotease of Legionella pneumophila causing lung damage in animal models of Legionnaires' disease. Here we demonstrate that ProA promotes infection of human lung tissue explants (HLTEs) and dissect the contribution to cell type specific replication and extracellular virulence mechanisms. For the first time, we reveal that co-incubation of HLTEs with purified ProA causes a significant increase of the alveolar septal thickness. This destruction of connective tissue fibres was further substantiated by collagen IV degradation assays. The moderate attenuation of a proA-negative mutant in A549 epithelial cells and THP-1 macrophages suggests that effects of ProA in tissue mainly result from extracellular activity. Correspondingly, ProA contributes to dissemination and serum resistance of the pathogen, which further expands the versatile substrate spectrum of this thermolysin-like protease. The crystal structure of ProA at 1.48 Å resolution showed high congruence to pseudolysin of Pseudomonas aeruginosa, but revealed deviations in flexible loops, the substrate binding pocket S1 ' and the repertoire of cofactors, by which ProA can be distinguished from respective homologues. In sum, this work specified virulence features of ProA at different organisational levels by zooming in from histopathological effects in human lung tissue to atomic details of the protease substrate determination.


Asunto(s)
Proteínas Bacterianas/metabolismo , Colágeno Tipo IV/metabolismo , Legionella pneumophila/enzimología , Legionella pneumophila/patogenicidad , Pulmón/microbiología , Metaloendopeptidasas/metabolismo , Alveolos Pulmonares/patología , Factores de Virulencia/metabolismo , Células A549 , Proteínas Bacterianas/química , Actividad Bactericida de la Sangre , Humanos , Legionella pneumophila/crecimiento & desarrollo , Pulmón/patología , Metaloendopeptidasas/química , Proteolisis , Alveolos Pulmonares/metabolismo , Células THP-1 , Virulencia , Factores de Virulencia/química
12.
EMBO Rep ; 22(3): e51163, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33492731

RESUMEN

The phagosome harboring the bacterial pathogen Legionella pneumophila is known to be enriched with phosphatidylinositol 4-phosphate (PtdIns4P), which is important for anchoring a subset of its virulence factors and potentially for signaling events implicated in the biogenesis of the Legionella-containing vacuole (LCV) that supports intracellular bacterial growth. Here we demonstrate that the effector MavQ is a phosphoinositide 3-kinase that specifically catalyzes the conversion of phosphatidylinositol (PtdIns) into PtdIns3P. The product of MavQ is subsequently phosphorylated by the effector LepB to yield PtdIns(3,4)P2, whose 3-phosphate is then removed by another effector SidF to generate PtdIns4P. We also show that MavQ is associated with the LCV and the ∆mavQ mutant displays phenotypes in the anchoring of a PtdIns4P-binding effector similar to those of ∆lepB or ∆sidF mutants. Our results establish a mechanism of de novo PtdIns4P biosynthesis by L. pneumophila via a catalysis axis comprised of MavQ, LepB, and SidF on the surface of its phagosome.


Asunto(s)
Proteínas Bacterianas , Legionella pneumophila , Fosfatidilinositol 3-Quinasas , Fosfatidilinositoles , Proteínas Bacterianas/genética , Legionella pneumophila/enzimología , Legionella pneumophila/genética , Fagosomas
13.
J Biol Chem ; 296: 100301, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33476647

RESUMEN

ADP-ribosyltransferases (ARTs) are a widespread superfamily of enzymes frequently employed in pathogenic strategies of bacteria. Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaire's disease, has acquired over 330 translocated effectors that showcase remarkable biochemical and structural diversity. However, the ART effectors that influence L. pneumophila have not been well defined. Here, we took a bioinformatic approach to search the Legionella effector repertoire for additional divergent members of the ART superfamily and identified an ART domain in Legionella pneumophila gene0181, which we hereafter refer to as Legionella ADP-Ribosyltransferase 1 (Lart1) (Legionella ART 1). We show that L. pneumophila Lart1 targets a specific class of 120-kDa NAD+-dependent glutamate dehydrogenase (GDH) enzymes found in fungi and protists, including many natural hosts of Legionella. Lart1 targets a conserved arginine residue in the NAD+-binding pocket of GDH, thereby blocking oxidative deamination of glutamate. Therefore, Lart1 could be the first example of a Legionella effector which directly targets a host metabolic enzyme during infection.


Asunto(s)
ADP Ribosa Transferasas/química , Proteínas Bacterianas/química , Glutamato Deshidrogenasa/química , Legionella pneumophila/genética , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/metabolismo , ADP-Ribosilación , Secuencia de Aminoácidos , Amoeba/microbiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Clonación Molecular , Desaminación , Escherichia coli/genética , Escherichia coli/metabolismo , Hongos , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Interacciones Huésped-Patógeno , Cinética , Legionella pneumophila/enzimología , Legionella pneumophila/patogenicidad , Modelos Moleculares , Oxidación-Reducción , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Especificidad por Sustrato
14.
J Mol Biol ; 433(3): 166746, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33309852

RESUMEN

Legionella pneumophila infects alveolar macrophages and can cause life-threatening pneumonia in humans. Upon internalization into the host cell, L. pneumophila injects numerous effector proteins into the host cytoplasm as a part of its pathogenesis. LegK7 is an effector kinase of L. pneumophila that functionally mimics the eukaryotic Mst kinase and phosphorylates the host MOB1 protein to exploit the Hippo pathway. To elucidate the LegK7 activation mechanism, we determined the apo structure of LegK7 in an inactive form and performed a comparative analysis of LegK7 structures. LegK7 is a non-RD kinase that contains an activation segment that is ordered, irrespective of stimulation, through a unique ß-hairpin-containing segment, and it does not require phosphorylation of the activation segment for activation. Instead, bacterial LegK7 becomes an active kinase via its heterologous molecular interaction with the host MOB1 protein. MOB1 binding triggers reorientation of the two lobes of the kinase domain, as well as a structural change in the interlobe hinge region in LegK7, consequently reshaping the LegK7 structure into an ATP binding-compatible closed conformation. Furthermore, we reveal that LegK7 is an atypical kinase that contains an N-terminal capping domain and a hydrophilic interlobe linker motif, which play key roles in the MOB1-induced activation of LegK7.


Asunto(s)
Quimiocina CXCL10/metabolismo , Interacciones Huésped-Patógeno , Legionella pneumophila/enzimología , Enfermedad de los Legionarios/metabolismo , Enfermedad de los Legionarios/microbiología , Proteínas Quinasas/metabolismo , Quimiocina CXCL10/química , Quimiocina CXCL10/genética , Activación Enzimática , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Fosforilación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Quinasas/química , Análisis Espectral , Relación Estructura-Actividad
15.
J Biol Chem ; 296: 100146, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33277357

RESUMEN

The siderophore rhizoferrin (N1,N4-dicitrylputrescine) is produced in fungi and bacteria to scavenge iron. Putrescine-producing bacterium Ralstonia pickettii synthesizes rhizoferrin and encodes a single nonribosomal peptide synthetase-independent siderophore (NIS) synthetase. From biosynthetic logic, we hypothesized that this single enzyme is sufficient for rhizoferrin biosynthesis. We confirmed this by expression of R. pickettii NIS synthetase in Escherichia coli, resulting in rhizoferrin production. This was further confirmed in vitro using the recombinant NIS synthetase, synthesizing rhizoferrin from putrescine and citrate. Heterologous expression of homologous lbtA from Legionella pneumophila, required for rhizoferrin biosynthesis in that species, produced siderophore activity in E. coli. Rhizoferrin is also synthesized by Francisella tularensis and Francisella novicida, but unlike R. pickettii or L. pneumophila, Francisella species lack putrescine biosynthetic pathways because of genomic decay. Francisella encodes a NIS synthetase FslA/FigA and an ornithine decarboxylase homolog FslC/FigC, required for rhizoferrin biosynthesis. Ornithine decarboxylase produces putrescine from ornithine, but we show here in vitro that FigA synthesizes N-citrylornithine, and FigC is an N-citrylornithine decarboxylase that together synthesize rhizoferrin without using putrescine. We co-expressed F. novicida figA and figC in E. coli and produced rhizoferrin. A 2.1 Å X-ray crystal structure of the FigC N-citrylornithine decarboxylase reveals how the larger substrate is accommodated and how active site residues have changed to recognize N-citrylornithine. FigC belongs to a new subfamily of alanine racemase-fold PLP-dependent decarboxylases that are not involved in polyamine biosynthesis. These data reveal a natural product biosynthetic workaround that evolved to bypass a missing precursor and re-establish it in the final structure.


Asunto(s)
Proteínas Bacterianas/metabolismo , Compuestos Férricos/metabolismo , Hierro/metabolismo , Péptido Sintasas/metabolismo , Putrescina/metabolismo , Ralstonia pickettii/enzimología , Sideróforos/metabolismo , Citratos/metabolismo , Francisella/enzimología , Legionella pneumophila/enzimología
16.
Chemistry ; 27(7): 2506-2512, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33075184

RESUMEN

Legionnaires' disease is caused by infection with the intracellularly replicating Gram-negative bacterium Legionella pneumophila. This pathogen uses an unconventional way of ubiquitinating host proteins by generating a phosphoribosyl linkage between substrate proteins and ubiquitin by making use of an ADPribosylated ubiquitin (UbADPr ) intermediate. The family of SidE effector enzymes that catalyze this reaction is counteracted by Legionella hydrolases, which are called Dups. This unusual ubiquitination process is important for Legionella proliferation and understanding these processes on a molecular level might prove invaluable in finding new treatments. Herein, a modular approach is used for the synthesis of triazole-linked UbADPr , and analogues thereof, and their affinity towards the hydrolase DupA is determined and hydrolysis rates are compared to natively linked UbADPr . The inhibitory effects of modified Ub on the canonical eukaryotic E1-enzyme Uba1 are investigated and rationalized in the context of a high-resolution crystal structure reported herein. Finally, it is shown that synthetic UbADPr analogues can be used to effectively pull-down overexpressed DupA from cell lysate.


Asunto(s)
ADP-Ribosilación , Legionella pneumophila/enzimología , Enfermedad de los Legionarios/microbiología , Ubiquitina/química , Ubiquitina/metabolismo , Proteínas Bacterianas/metabolismo , Células HEK293 , Humanos , Hidrolasas/metabolismo , Legionella pneumophila/crecimiento & desarrollo , Enzimas Activadoras de Ubiquitina/metabolismo , Ubiquitinación
17.
Mol Microbiol ; 115(4): 539-553, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33034117

RESUMEN

The production of the pyrimidine moiety in thiamine synthesis, 2-methyl-4-amino-5-hydroxymethylpyrimidine phosphate (HMP-P), has been described to proceed through the Thi5-dependent pathway in Saccharomyces cerevisiae and other yeast. Previous work found that ScThi5 functioned poorly in a heterologous context. Here we report a bacterial ortholog to the yeast HMP-P synthase (Thi5) was necessary for HMP synthesis in Legionella pneumophila. Unlike ScThi5, LpThi5 functioned in vivo in Salmonella enterica under multiple growth conditions. The protein LpThi5 is a dimer that binds pyridoxal-5'-phosphate (PLP), apparently without a solvent-exposed Schiff base. A small percentage of LpThi5 protein co-purifies with a bound molecule that can be converted to HMP. Analysis of variant proteins both in vivo and in vitro confirmed that residues in sequence motifs conserved across bacterial and eukaryotic orthologs modulate the function of LpThi5. IMPORTANCE: Thiamine is an essential vitamin for the vast majority of organisms. There are multiple strategies to synthesize and salvage this vitamin. The predominant pathway for synthesis of the pyrimidine moiety of thiamine involves the Fe-S cluster protein ThiC. An alternative pathway utilizes Thi5, a novel enzyme that uses PLP as a substrate. The Thi5-dependent pathway is poorly characterized in yeast and has not been characterized in Bacteria. Here we demonstrate that a Thi5-dependent pathway is necessary for thiamine biosynthesis in Legionella pneumophila and provide biochemical data to extend knowledge of the Thi5 enzyme, the corresponding biosynthetic pathway, and the role of metabolic network architecture in optimizing its function.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Legionella pneumophila/química , Legionella pneumophila/enzimología , Legionella pneumophila/metabolismo , Fosfato de Piridoxal/metabolismo , Pirimidinas/biosíntesis , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Vías Biosintéticas , Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Legionella pneumophila/genética , Filogenia , Unión Proteica , Conformación Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Salmonella enterica/metabolismo , Tiamina/biosíntesis
18.
Elife ; 92020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33136002

RESUMEN

Legionella pneumophila extensively modulates the host ubiquitin network to create the Legionella-containing vacuole (LCV) for its replication. Many of its virulence factors function as ubiquitin ligases or deubiquitinases (DUBs). Here, we identify Lem27 as a DUB that displays a preference for diubiquitin formed by K6, K11, or K48. Lem27 is associated with the LCV where it regulates Rab10 ubiquitination in concert with SidC and SdcA, two bacterial E3 ubiquitin ligases. Structural analysis of the complex formed by an active fragment of Lem27 and the substrate-based suicide inhibitor ubiquitin-propargylamide (PA) reveals that it harbors a fold resembling those in the OTU1 DUB subfamily with a Cys-His catalytic dyad and that it recognizes ubiquitin via extensive hydrogen bonding at six contact sites. Our results establish Lem27 as a DUB that functions to regulate protein ubiquitination on L. pneumophila phagosomes by counteracting the activity of bacterial ubiquitin E3 ligases.


Asunto(s)
Proteínas Bacterianas/metabolismo , Enzimas Desubicuitinizantes/metabolismo , Legionella pneumophila/enzimología , Fagosomas/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Enzimas Desubicuitinizantes/genética , Legionella pneumophila/química , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Pargilina/análogos & derivados , Pargilina/metabolismo , Fagosomas/metabolismo , Propilaminas/metabolismo , Ubiquitina/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Vacuolas/enzimología , Vacuolas/genética , Vacuolas/metabolismo
19.
PLoS Pathog ; 16(8): e1008734, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32853279

RESUMEN

AMPylation, the post-translational modification with adenosine monophosphate (AMP), is catalyzed by effector proteins from a variety of pathogens. Legionella pneumophila is thus far the only known pathogen that, in addition to encoding an AMPylase (SidM/DrrA), also encodes a deAMPylase, called SidD, that reverses SidM-mediated AMPylation of the vesicle transport GTPase Rab1. DeAMPylation is catalyzed by the N-terminal phosphatase-like domain of SidD. Here, we determined the crystal structure of full length SidD including the uncharacterized C-terminal domain (CTD). A flexible loop rich in aromatic residues within the CTD was required to target SidD to model membranes in vitro and to the Golgi apparatus within mammalian cells. Deletion of the loop (Δloop) or substitution of its aromatic phenylalanine residues rendered SidD cytosolic, showing that the hydrophobic loop is the primary membrane-targeting determinant of SidD. Notably, deletion of the two terminal alpha helices resulted in a CTD variant incapable of discriminating between membranes of different composition. Moreover, a L. pneumophila strain producing SidDΔloop phenocopied a L. pneumophila ΔsidD strain during growth in mouse macrophages and displayed prolonged co-localization of AMPylated Rab1 with LCVs, thus revealing that membrane targeting of SidD via its CTD is a critical prerequisite for its ability to catalyze Rab1 deAMPylation during L. pneumophila infection.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Membrana Celular/microbiología , Legionella pneumophila/enzimología , Enfermedad de los Legionarios/microbiología , Adenosina Monofosfato/metabolismo , Animales , Proteínas Bacterianas/genética , Femenino , Aparato de Golgi/metabolismo , Humanos , Legionella pneumophila/química , Legionella pneumophila/genética , Ratones , Dominios Proteicos
20.
Chembiochem ; 21(20): 2903-2907, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32421893

RESUMEN

Stable NAD+ analogues carrying single atom substitutions in either the furanose ring or the nicotinamide part have proven their value as inhibitors for NAD+ -consuming enzymes. To investigate the potential of such compounds to inhibit the adenosine diphosphate ribosyl (ADPr) transferase activity of the Legionella SdeC enzyme, we prepared three NAD+ analogues, namely carbanicotinamide adenosine dinucleotide (c-NAD+ ), thionicotinamide adenosine dinucleotide (S-NAD+ ) and benzamide adenosine dinucleotide (BAD). We optimized the chemical synthesis of thionicotinamide riboside and for the first time used an enzymatic approach to convert all three ribosides into the corresponding NAD+ mimics. We thus expanded the known scope of substrates for the NRK1/NMNAT1 enzyme combination by turning all three modified ribosides into NAD+ analogues in a scalable manner. We then compared the three NAD+ mimics side-by-side in a single assay for enzyme inhibition on Legionella effector enzyme SdeC. The class of SidE enzymes to which SdeC belongs was recently identified to be important in bacterial virulence, and we found SdeC to be inhibited by S-NAD+ and BAD with IC50 values of 28 and 39 µM, respectively.


Asunto(s)
Legionella pneumophila/enzimología , NAD/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Modelos Moleculares , Conformación Molecular , NAD/síntesis química , NAD/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/síntesis química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...