Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(25): e2316143121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38861595

RESUMEN

Vibrio vulnificus causes life-threatening wound and gastrointestinal infections, mediated primarily by the production of a Multifunctional-Autoprocessing Repeats-In-Toxin (MARTX) toxin. The most commonly present MARTX effector domain, the Makes Caterpillars Floppy-like (MCF) toxin, is a cysteine protease stimulated by host adenosine diphosphate (ADP) ribosylation factors (ARFs) to autoprocess. Here, we show processed MCF then binds and cleaves host Ras-related proteins in brain (Rab) guanosine triphosphatases within their C-terminal tails resulting in Rab degradation. We demonstrate MCF binds Rabs at the same interface occupied by ARFs. Moreover, we show MCF preferentially binds to ARF1 prior to autoprocessing and is active to cleave Rabs only subsequent to autoprocessing. We then use structure prediction algorithms to demonstrate that structural composition, rather than sequence, determines Rab target specificity. We further determine a crystal structure of aMCF as a swapped dimer, revealing an alternative conformation we suggest represents the open, activated state of MCF with reorganized active site residues. The cleavage of Rabs results in Rab1B dispersal within cells and loss of Rab1B density in the intestinal tissue of infected mice. Collectively, our work describes an extracellular bacterial mechanism whereby MCF is activated by ARFs and subsequently induces the degradation of another small host guanosine triphosphatase (GTPase), Rabs, to drive organelle damage, cell death, and promote pathogenesis of these rapidly fatal infections.


Asunto(s)
Toxinas Bacterianas , Vibrio vulnificus , Proteínas de Unión al GTP rab , Animales , Femenino , Humanos , Ratones , Factores de Ribosilacion-ADP/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/química , Células HEK293 , Ratones Endogámicos ICR , Proteolisis , Proteínas de Unión al GTP rab/metabolismo , Vibriosis/microbiología , Vibriosis/metabolismo , Vibrio vulnificus/metabolismo , Vibrio vulnificus/patogenicidad
2.
Proc Natl Acad Sci U S A ; 121(25): e2319903121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38870058

RESUMEN

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


Asunto(s)
Agrobacterium tumefaciens , Proteínas Bacterianas , Biopelículas , GMP Cíclico , Pterinas , Biopelículas/crecimiento & desarrollo , Agrobacterium tumefaciens/metabolismo , Agrobacterium tumefaciens/genética , Pterinas/metabolismo , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteobacteria/metabolismo , Proteobacteria/genética , Cofactores de Molibdeno , Periplasma/metabolismo , Proteínas Periplasmáticas/metabolismo , Proteínas Periplasmáticas/genética , Proteínas de Unión Periplasmáticas/metabolismo , Proteínas de Unión Periplasmáticas/genética , Regulación Bacteriana de la Expresión Génica
3.
Mol Ther ; 31(7): 1904-1919, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-36945775

RESUMEN

The function and significance of RAS proteins in cancer have been widely studied for decades. In 2013, the National Cancer Institute established the RAS Initiative to explore innovative approaches for attacking the proteins encoded by mutant forms of RAS genes and to create effective therapies for RAS-driven cancers. This initiative spurred researchers to develop novel approaches and to discover small molecules targeting this protein that was at one time termed "undruggable." More recently, advanced efforts in RAS degraders including PROTACs, linker-based degraders, and direct proteolysis degraders have been explored as novel strategies to target RAS for cancer treatment. These RAS degraders present new opportunities for RAS therapies and may prove fruitful in understanding basic cell biology. Novel delivery strategies will further enhance the efficacy of these therapeutics. In this review, we summarize recent efforts to develop RAS degraders, including PROTACs and E3 adaptor and ligase fusions as cancer therapies. This review also details the direct RAS protease degrader, RAS/RAP1-specific endopeptidase that directly and specifically cleaves RAS.


Asunto(s)
Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Proteolisis , Proteínas/metabolismo , Endopeptidasas/genética , Proteínas ras/genética , Ubiquitina-Proteína Ligasas
4.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-33972410

RESUMEN

The genome of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronavirus has a capping modification at the 5'-untranslated region (UTR) to prevent its degradation by host nucleases. These modifications are performed by the Nsp10/14 and Nsp10/16 heterodimers using S-adenosylmethionine as the methyl donor. Nsp10/16 heterodimer is responsible for the methylation at the ribose 2'-O position of the first nucleotide. To investigate the conformational changes of the complex during 2'-O methyltransferase activity, we used a fixed-target serial synchrotron crystallography method at room temperature. We determined crystal structures of Nsp10/16 with substrates and products that revealed the states before and after methylation, occurring within the crystals during the experiments. Here we report the crystal structure of Nsp10/16 in complex with Cap-1 analog (m7GpppAm2'-O). Inhibition of Nsp16 activity may reduce viral proliferation, making this protein an attractive drug target.


Asunto(s)
Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , SARS-CoV-2/química , Cristalografía , Metilación , Metiltransferasas/química , Metiltransferasas/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Análogos de Caperuza de ARN/química , Análogos de Caperuza de ARN/metabolismo , Caperuzas de ARN/química , ARN Mensajero/química , ARN Viral/química , S-Adenosilhomocisteína/química , S-Adenosilhomocisteína/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Sincrotrones , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(12): 6811-6821, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32156726

RESUMEN

Emerging evidence suggests the Pseudomonas aeruginosa accessory genome is enriched with uncharacterized virulence genes. Identification and characterization of such genes may reveal novel pathogenic mechanisms used by particularly virulent isolates. Here, we utilized a mouse bacteremia model to quantify the virulence of 100 individual P. aeruginosa bloodstream isolates and performed whole-genome sequencing to identify accessory genomic elements correlated with increased bacterial virulence. From this work, we identified a specific contact-dependent growth inhibition (CDI) system enriched among highly virulent P. aeruginosa isolates. CDI systems contain a large exoprotein (CdiA) with a C-terminal toxin (CT) domain that can vary between different isolates within a species. Prior work has revealed that delivery of a CdiA-CT domain upon direct cell-to-cell contact can inhibit replication of a susceptible target bacterium. Aside from mediating interbacterial competition, we observed our virulence-associated CdiA-CT domain to promote toxicity against mammalian cells in culture and lethality during mouse bacteremia. Structural and functional studies revealed this CdiA-CT domain to have in vitro tRNase activity, and mutations that abrogated this tRNAse activity in vitro also attenuated virulence. Furthermore, CdiA contributed to virulence in mice even in the absence of contact-dependent signaling. Overall, our findings indicate that this P. aeruginosa CDI system functions as both an interbacterial inhibition system and a bacterial virulence factor against a mammalian host. These findings provide an impetus for continued studies into the complex role of CDI systems in P. aeruginosa pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Inhibición de Contacto/genética , Escherichia coli/crecimiento & desarrollo , Genómica/métodos , Pseudomonas aeruginosa/crecimiento & desarrollo , Factores de Virulencia/metabolismo , Virulencia , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Femenino , Genoma Bacteriano , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación , Pseudomonas aeruginosa/metabolismo , Transducción de Señal , Factores de Virulencia/genética
6.
Proc Natl Acad Sci U S A ; 117(29): 16938-16948, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32616570

RESUMEN

Despite nearly four decades of effort, broad inhibition of oncogenic RAS using small-molecule approaches has proven to be a major challenge. Here we describe the development of a pan-RAS biologic inhibitor composed of the RAS-RAP1-specific endopeptidase fused to the protein delivery machinery of diphtheria toxin. We show that this engineered chimeric toxin irreversibly cleaves and inactivates intracellular RAS at low picomolar concentrations terminating downstream signaling in receptor-bearing cells. Furthermore, we demonstrate in vivo target engagement and reduction of tumor burden in three mouse xenograft models driven by either wild-type or mutant RAS Intracellular delivery of a potent anti-RAS biologic through a receptor-mediated mechanism represents a promising approach to developing RAS therapeutics against a broad array of cancers.


Asunto(s)
Toxina Diftérica/metabolismo , Endopeptidasas/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Proteolisis , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo , Animales , Antineoplásicos/uso terapéutico , Células Cultivadas , Toxina Diftérica/química , Toxina Diftérica/genética , Endopeptidasas/química , Endopeptidasas/genética , Femenino , Células HCT116 , Humanos , Masculino , Ratones , Ratones Desnudos , Mutación , Señales de Clasificación de Proteína , Proteínas Recombinantes/uso terapéutico , Proteínas ras/genética
7.
J Bacteriol ; 204(5): e0055521, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35435721

RESUMEN

Alpha-pore-forming toxins (α-PFTs) are secreted by many species of bacteria, including Escherichia coli, Aeromonas hydrophila, and Bacillus thuringiensis, as part of their arsenal of virulence factors, and are often cytotoxic. In particular, for α-PFTs, the membrane-spanning channel they form is composed of hydrophobic α-helices. These toxins oligomerize at the surface of target cells and transition from a soluble to a protomer state in which they expose their hydrophobic regions and insert into the membrane to form a pore. The pores may be composed of homooligomers of one component or heterooligomers with two or three components, resulting in bi- or tripartite toxins. The multicomponent α-PFTs are often expressed from a single operon. Recently, motility-associated killing factor A (MakA), an α-PFT, was discovered in Vibrio cholerae. We report that makA is found on the V. cholerae GI-10 genomic island within an operon containing genes for two other potential α-PFTs, MakB and MakE. We determined the X-ray crystal structures for MakA, MakB, and MakE and demonstrated that all three are structurally related to the α-PFT family in the soluble state, and we modeled their protomer state based on the α-PFT AhlB from A. hydrophila. We found that MakA alone is cytotoxic at micromolar concentrations. However, combining MakA with MakB and MakE is cytotoxic at nanomolar concentrations, with specificity for J774 macrophage cells. Our data suggest that MakA, -B, and -E are α-PFTs that potentially act as a tripartite pore-forming toxin with specificity for phagocytic cells. IMPORTANCE The bacterium Vibrio cholerae causes gastrointestinal, wound, and skin infections. The motility-associated killing factor A (MakA) was recently shown to be cytotoxic against colon, prostate, and other cancer cells. However, at the outset of this study, the capacity of MakA to damage cells in combination with other Mak proteins encoded in the same operon had not been elucidated. We determined the structures of three Mak proteins and established that they are structurally related to the α-PFTs. Compared to MakA alone, the combination of all three toxins was more potent specifically in mouse macrophages. This study highlights the idea that the Mak toxins are selectively cytotoxic and thus may function as a tripartite toxin with cell type specificity.


Asunto(s)
Vibrio cholerae , Animales , Citotoxinas/genética , Citotoxinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Islas Genómicas , Ratones , Proteínas Citotóxicas Formadoras de Poros , Subunidades de Proteína/metabolismo , Vibrio cholerae/metabolismo , Factores de Virulencia/metabolismo
8.
Infect Immun ; 90(4): e0062721, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35254094

RESUMEN

Vibrio vulnificus is an opportunistic pathogen that causes gastroenteritis and septicemia in humans. The V. vulnificus multifunctional-autoprocessing repeats-in-toxin (MARTX) toxin is a pore-forming toxin that translocates multiple functionally independent effector domains into target cells and an essential virulence factor for fatal disease. The effector repertoire delivered and thus the mechanism of action of the toxin can differ dramatically across V. vulnificus isolates. Here, we utilize a strain of V. vulnificus that carries an F-type MARTX toxin that delivers an actin cross-linking domain (ACD) and four other effector domains. We demonstrate that ACD is the primary driver of virulence following intragastric infection and of bacterial dissemination to distal organs. We additionally show that ACD activates the transcription of intermediate early response genes in cultured intestinal epithelial cells (IECs). However, the genes activated by ACD are suppressed, at least in part, by the codelivered Ras/Rap1-specific endopeptidase (RRSP). The transcriptional response induced by strains translocating only RRSP results in a unique transcriptional profile, demonstrating that the transcriptional response to V. vulnificus is remodeled rather than simply suppressed by the MARTX toxin effector repertoire. Regardless, the transcriptional response in the intestinal tissue of infected mice is dominated by ACD-mediated induction of genes associated with response to tissue damage and is not impacted by RRSP or the three other effectors codelivered with ACD and RRSP. These data demonstrate that while other effectors do remodel early intestinal innate immune responses, ACD is the dominant driver of disease progression by ACD+ V. vulnificus during intestinal infection.


Asunto(s)
Toxinas Bacterianas , Vibriosis , Vibrio vulnificus , Actinas , Animales , Toxinas Bacterianas/genética , Progresión de la Enfermedad , Ratones , Vibriosis/microbiología , Vibrio vulnificus/genética , Factores de Virulencia/genética
9.
Antimicrob Agents Chemother ; 66(10): e0098522, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36129295

RESUMEN

Resistance to antipseudomonal penicillins and cephalosporins is often driven by the overproduction of the intrinsic ß-lactamase AmpC. However, OXA-10-family ß-lactamases are a rich source of resistance in Pseudomonas aeruginosa. OXA ß-lactamases have a propensity for mutation that leads to extended spectrum cephalosporinase and carbapenemase activity. In this study, we identified isolates from a subclade of the multidrug-resistant (MDR) high risk P. aeruginosa clonal complex CC446 with a resistance to ceftazidime. A genomic analysis revealed that these isolates harbored a plasmid containing a novel allele of blaOXA-10, named blaOXA-935, which was predicted to produce an OXA-10 variant with two amino acid substitutions: an aspartic acid instead of a glycine at position 157 and a serine instead of a phenylalanine at position 153. The G157D mutation, present in OXA-14, is associated with the resistance of P. aeruginosa to ceftazidime. Compared to OXA-14, OXA-935 showed increased catalytic efficiency for ceftazidime. The deletion of blaOXA-935 restored the sensitivity to ceftazidime, and susceptibility profiling of P. aeruginosa laboratory strains expressing blaOXA-935 revealed that OXA-935 conferred ceftazidime resistance. To better understand the impacts of the variant amino acids, we determined the crystal structures of OXA-14 and OXA-935. Compared to OXA-14, the F153S mutation in OXA-935 conferred increased flexibility in the omega (Ω) loop. Amino acid changes that confer extended spectrum cephalosporinase activity to OXA-10-family ß-lactamases are concerning, given the rising reliance on novel ß-lactam/ß-lactamase inhibitor combinations, such as ceftolozane-tazobactam and ceftazidime-avibactam, to treat MDR P. aeruginosa infections.


Asunto(s)
Ceftazidima , Infecciones por Pseudomonas , Humanos , Ceftazidima/farmacología , Pseudomonas aeruginosa , Inhibidores de beta-Lactamasas/farmacología , Cefalosporinasa/genética , Ácido Aspártico , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Tazobactam/farmacología , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Cefalosporinas/farmacología , Compuestos de Azabiciclo/farmacología , Serina , Fenilalanina , Glicina , Infecciones por Pseudomonas/tratamiento farmacológico
10.
Regul Toxicol Pharmacol ; 131: 105146, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35219763

RESUMEN

On October 21-22, 2020 the HESI (Health and Environmental Sciences Institute) Protein Allergens, Toxins, and Bioinformatics Committee, and the Society of Toxicology Food Safety Specialty Section co-hosted a virtual workshop titled "From Protein Toxins to Applied Toxicological Testing". The workshop focused on the safety assessment of novel proteins contained in foods and feeds, was globally represented by over 200 stakeholder attendees, and featured contributions from experts in academia, government and non-government organizations, and agricultural biotechnology developers from the private sector. A range of topics relevant to novel protein safety were discussed, including: the state of protein toxin biology, modes and mechanisms of action, structures and activity, use of bioinformatic analyses to assess the safety of a protein, and ways to leverage computational biology with in silico approaches for protein toxin identification/characterization. Key outcomes of the workshop included the appreciation of the complexity of developing a definition for a protein toxin when viewed from the perspective of food and feed safety, confirming the need for a case-by-case hypothesis-driven interpretation of bioinformatic results that leverages additional metadata rather than an alignment threshold-driven interpretation, and agreement that a "toxin protein database" is not necessary, as the bioinformatic needs for toxin detection may be accomplished by existing databases such as Pfam and UniProtKB/Swiss-Prot. In this paper, a path forward is proposed.


Asunto(s)
Biología Computacional , Inocuidad de los Alimentos , Alérgenos/química , Alérgenos/toxicidad , Biotecnología/métodos , Bases de Datos de Proteínas
11.
J Bacteriol ; 203(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33753465

RESUMEN

VxrA and VxrB are cognate histidine kinase (HK) - response regulator (RR) pairs of a two-component signaling system (TCS) found in Vibrio cholerae, a bacterial pathogen that causes cholera. The VxrAB TCS positively regulates virulence, the Type VI Secretion System, biofilm formation, and cell wall homeostasis in V. cholerae, providing protection from environmental stresses and contributing to the transmission and virulence of the pathogen. The VxrA HK has a unique periplasmic sensor domain (SD) and, remarkably, lacks a cytoplasmic linker domain between the second transmembrane helix and the dimerization and histidine phosphotransfer (DHp) domain, indicating that this system may utilize a potentially unique signal sensing and transmission TCS mechanism. In this study, we have determined several crystal structures of VxrA-SD and its mutants. These structures reveal a novel structural fold forming an unusual ß hairpin-swapped dimer. A conformational change caused by relative rotation of the two monomers in a VxrA-SD dimer could potentially change the association of transmembrane helices and, subsequently, the pairing of cytoplasmic DHp domains. Based on the structural observation, we propose a putative scissor-like closing regulation mechanism for the VxrA HK.IMPORTANCE V. cholerae has a dynamic life cycle, which requires rapid adaptation to changing external conditions. Two-component signal transduction (TCS) systems allow V. cholerae to sense and respond to these environmental changes. The VxrAB TCS positively regulates a number of important V. cholerae phenotypes, including virulence, the Type Six Secretion System, biofilm formation, and cell wall homeostasis. Here, we provide the crystal structure of the VxrA sensor histidine kinase sensing domain and propose a mechanism for signal transduction. The cognate signal for VxrAB remains unknown, however, in this work we couple our structural analysis with functional assessments of key residues to further our understanding of this important TCS.

12.
PLoS Pathog ; 15(6): e1007895, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31216353

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1002581.].

13.
Cell Microbiol ; 22(4): e13178, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32185903

RESUMEN

Intimate interactions between the armament of pathogens and their host dictate tissue and host susceptibility to infection also forging specific pathophysiological outcomes. Studying these interactions at the molecular level has provided an invaluable source of knowledge on cellular processes, as ambitioned by the Cellular Microbiology discipline when it emerged in early 90s. Bacterial toxins act on key cell regulators or membranes to produce major diseases and therefore constitute a remarkable toolbox for dissecting basic biological processes. Here, we review selected examples of recent studies on bacterial toxins illustrating how fruitful the discipline of cellular microbiology is in shaping our understanding of eukaryote processes. This ever-renewing discipline unveils new virulence factor biochemical activities shared by eukaryotic enzymes and hidden rules of cell proteome homeostasis, a particularly promising field to interrogate the impact of proteostasis breaching in late onset human diseases. It is integrating new concepts from the physics of soft matter to capture biomechanical determinants forging cells and tissues architecture. The success of this discipline is also grounded by the development of therapeutic tools and new strategies to treat both infectious and noncommunicable human diseases.


Asunto(s)
Toxinas Bacterianas/metabolismo , Células Eucariotas/microbiología , Células Eucariotas/fisiología , Interacciones Huésped-Patógeno , Microambiente Celular , Humanos , Proteostasis , Factores de Virulencia
14.
Cell Microbiol ; 22(2): e13133, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31658406

RESUMEN

Studies have successfully elucidated the mechanism of action of several effector domains that comprise the multifunctional-autoprocessing repeats-in-toxins (MARTX) toxins of Vibrio vulnificus. However, the biochemical linkage between the cysteine proteolytic activity of Makes Caterpillars Floppy (MCF)-like effector and its cellular effects remains unknown. In this study, we identify the host cell factors that activate in vivo and in vitro MCF autoprocessing as adenosine diphosphate (ADP)-Ribosylation Factor 1 (ARF1) and ADP-Ribosylation Factor 3 (ARF3). Autoprocessing activity is enhanced when ARF1 is in its active [guanosine triphosphate (GTP)-bound] form compared to the inactive [guanosine diphosphate (GDP)-bound] form. Subsequent to auto-cleavage, MCF is acetylated on its exposed N-terminal glycine residue. Acetylation apparently does not dictate subcellular localization as MCF is found localized throughout the cell. However, the cleaved form of MCF gains the ability to bind to the specialized lipid phosphatidylinositol 5-phosphate enriched in Golgi and other membranes necessary for endocytic trafficking, suggesting that a fraction of MCF may be subcellularly localized. Traditional thin-section electron microscopy, high-resolution cryoAPEX localization, and fluorescent microscopy show that MCF causes Golgi dispersal resulting in extensive vesiculation. In addition, host mitochondria are disrupted and fragmented. Mass spectrometry analysis found no reproducible modifications of ARF1 suggesting that ARF1 is not post-translationally modified by MCF. Further, catalytically active MCF does not stably associate with ARF1. Our data indicate not only that ARF1 is a cross-kingdom activator of MCF, but also that MCF may mediate cytotoxicity by directly targeting another yet to be identified protein. This study begins to elucidate the biochemical activity of this important domain and gives insight into how it may promote disease progression.


Asunto(s)
Factor 1 de Ribosilacion-ADP/metabolismo , Toxinas Bacterianas/metabolismo , Aparato de Golgi/metabolismo , Vibrio vulnificus/metabolismo , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Procesamiento Proteico-Postraduccional , Transporte de Proteínas
15.
J Bacteriol ; 202(24)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32900828

RESUMEN

Pathogenic Vibrio species use many different approaches to subvert, attack, and undermine the host response. The toxins they produce are often responsible for the devastating effects associated with their diseases. These toxins target a variety of host proteins, which leads to deleterious effects, including dissolution of cell organelle integrity and inhibition of protein secretion. Becoming increasingly prevalent as cofactors for Vibrio toxins are proteins of the small GTPase families. ADP-ribosylation factor small GTPases (ARFs) in particular are emerging as a common host cofactor necessary for full activation of Vibrio toxins. While ARFs are not the direct target of Vibrio cholerae cholera toxin (CT), ARF binding is required for its optimal activity as an ADP-ribosyltransferase. The makes caterpillars floppy (MCF)-like and the domain X (DmX) effectors of the Vibrio vulnificus multifunctional autoprocessing repeats-in-toxin (MARTX) toxin also both require ARFs to initiate autoprocessing and activation as independent effectors. ARFs are ubiquitously expressed in eukaryotes and are key regulators of many cellular processes, and as such they are ideal cofactors for Vibrio pathogens that infect many host species. In this review, we cover in detail the known Vibrio toxins that use ARFs as cross-kingdom activators to both stimulate and optimize their activity. We further discuss how these contrast to toxins and effectors from other bacterial species that coactivate, stimulate, or directly modify host ARFs as their mechanisms of action.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Toxinas Bacterianas/metabolismo , Vibriosis/enzimología , Vibrio vulnificus/metabolismo , Factores de Ribosilacion-ADP/genética , Animales , Toxinas Bacterianas/genética , Interacciones Huésped-Patógeno , Humanos , Familia de Multigenes , Vibriosis/genética , Vibriosis/microbiología , Vibrio vulnificus/clasificación , Vibrio vulnificus/genética
16.
Chembiochem ; 21(19): 2772-2776, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32369652

RESUMEN

The nontoxic, anthrax protective antigen/lethal factor N-terminal domain (PA/LFN ) complex is an effective platform for translocating proteins into the cytosol of cells. Mutant PA (mPA) was recently fused to epidermal growth factor (EGF) to retarget delivery of LFN to cells bearing EGF receptors (EGFR), but the requirement for a known cognate ligand limits the applicability of this approach. Here, we render practical protective antigen retargeting to a variety of receptors with mPA single-chain variable fragment (scFv) fusion constructs. Our design enables the targeting of two pancreatic cancer-relevant receptors, EGFR and carcinoembryonic antigen. We demonstrate that fusion to scFvs does not disturb the basic functions of mPA. Moreover, mPA-scFv fusions enable cell-specific delivery of diphtheria toxin catalytic domain and Ras/Rap1-specific endopeptidase to pancreatic cancer cells. Importantly, mPA-scFv fusion-based treatments display potent cell-specific toxicity in vitro, opening fundamentally new routes toward engineered immunotoxins and providing a potential solution to the challenge of targeted protein delivery to the cytosol of cancer cells.


Asunto(s)
Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Antígeno Carcinoembrionario/metabolismo , Endopeptidasas/metabolismo , Neoplasias Pancreáticas/metabolismo , Antígenos Bacterianos/genética , Toxinas Bacterianas/genética , Citosol/metabolismo , Receptores ErbB/metabolismo , Humanos , Modelos Moleculares , Neoplasias Pancreáticas/patología
17.
J Infect Dis ; 219(6): 889-897, 2019 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-30289477

RESUMEN

BACKGROUND: The bacterial pathogen Vibrio vulnificus causes severe septic foodborne infections. The multifunctional autoprocessing repeats-in-toxins (MARTX) toxin is an important secreted virulence factor. The effector domain region is essential for lethal intestinal infection in mice, but the contribution of each of the 5 effector domains to infection has not been investigated. METHODS: V. vulnificus mutants with varying effector domain content were inoculated intragastrically to mice, and the time to death was monitored to establish the contribution of each effector domain to overall virulence. Each strain was also tested for bacterial dissemination from the intestine to internal organs and for inhibition of phagocytosis. RESULTS: The effector domain region was required for V. vulnificus to inhibit phagocytosis by J774 macrophages, but no single effector domain was required. No single MARTX effector domain was necessary for bacterial dissemination. Nonetheless, overall survival of infected mice differed with respect to the infecting V. vulnificus strain. Removal of rid or rrsp significantly reduced the virulence potential of V. vulnificus, while deletion of duf1 or abh accelerated the time to death. CONCLUSION: Rho GTPases inactivation domain and Ras/Rap1-specific endopeptidase each exert greater effects on virulence than other MARTX domains, suggesting that modulation of the Rho/Ras family of GTPases is a critical function of the toxin during intestinal infection.


Asunto(s)
Toxinas Bacterianas/metabolismo , Endopeptidasas/metabolismo , Vibriosis/microbiología , Vibrio vulnificus/patogenicidad , Factores de Virulencia/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Toxinas Bacterianas/genética , Femenino , Ratones Endogámicos ICR , Fagocitosis , Dominios Proteicos , Especificidad por Sustrato , Vibrio vulnificus/genética , Virulencia , Factores de Virulencia/genética , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo , Proteínas de Unión al GTP rho/genética
18.
Mol Microbiol ; 107(2): 133-141, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29114985

RESUMEN

Bacteria often coordinate virulence factors to fine-tune the host response during infection. These coordinated events can include toxins counteracting or amplifying effects of another toxin or though regulating the stability of virulence factors to remove their function once it is no longer needed. Multifunctional autoprocessing repeats-in toxin (MARTX) toxins are effector delivery toxins that form a pore into the plasma membrane of a eukaryotic cell to deliver multiple effector proteins into the cytosol of the target cell. The function of these proteins includes manipulating actin cytoskeletal dynamics, regulating signal transduction pathways and inhibiting host secretory pathways. Investigations into the molecular mechanisms of these effector domains are providing insight into how the function of some effectors overlap and regulate one another during infection. Coordinated crosstalk of effector function suggests that MARTX toxins are not simply a sum of all their parts. Instead, modulation of cell function by effector domains may depend on which other effector domain are co-delivered. Future studies will elucidate how these effectors interact with each other to modulate the bacterial host interaction.


Asunto(s)
Bacterias/patogenicidad , Toxinas Bacterianas/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Citoesqueleto de Actina/metabolismo , Autofagia , Bacterias/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Membrana Celular/microbiología , Células Eucariotas/microbiología , Interacciones Microbiota-Huesped , Humanos , Vías Secretoras , Virulencia
19.
PLoS Pathog ; 13(1): e1006119, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28060924

RESUMEN

Vibrio vulnificus causes highly lethal bacterial infections in which the Multifunctional Autoprocessing Repeats-in-Toxins (MARTX) toxin product of the rtxA1 gene is a key virulence factor. MARTX toxins are secreted proteins up to 5208 amino acids in size. Conserved MARTX N- and C-terminal repeat regions work in concert to form pores in eukaryotic cell membranes, through which the toxin's central region of modular effector domains is translocated. Upon inositol hexakisphosphate-induced activation of the of the MARTX cysteine protease domain (CPD) in the eukaryotic cytosol, effector domains are released from the holotoxin by autoproteolytic activity. We previously reported that the native MARTX toxin effector domain repertoire is dispensable for epithelial cellular necrosis in vitro, but essential for cell rounding and apoptosis prior to necrotic cell death. Here we use an intragastric mouse model to demonstrate that the effector domain region is required for bacterial virulence during intragastric infection. The MARTX effector domain region is essential for bacterial dissemination from the intestine, but dissemination occurs in the absence of overt intestinal tissue pathology. We employ an in vitro model of V. vulnificus interaction with polarized colonic epithelial cells to show that the MARTX effector domain region induces rapid intestinal barrier dysfunction and increased paracellular permeability prior to onset of cell lysis. Together, these results negate the inherent assumption that observations of necrosis in vitro directly predict bacterial virulence, and indicate a paradigm shift in our conceptual understanding of MARTX toxin function during intestinal infection. Results implicate the MARTX effector domain region in mediating early bacterial dissemination from the intestine to distal organs-a key step in V. vulnificus foodborne pathogenesis-even before onset of overt intestinal pathology.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Uniones Estrechas/patología , Vibriosis/transmisión , Vibrio vulnificus/patogenicidad , Animales , Apoptosis/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/ultraestructura , Toxinas Bacterianas/genética , Membrana Celular/patología , Modelos Animales de Enfermedad , Epitelio/microbiología , Epitelio/patología , Femenino , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/ultraestructura , Ratones , Ratones Endogámicos ICR , Ácido Fítico/farmacología , Estructura Terciaria de Proteína , Vibriosis/microbiología , Vibrio vulnificus/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
20.
J Bacteriol ; 200(15): e00304-18, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29866809

RESUMEN

Vibrio2017: The ASM Conference on the Biology of Vibrios, was held in November 2017. The conference focused on all aspects of biology related to the bacterial genus Vibrio. The meeting highlighted that the Vibrios have a tremendous impact on humans, both directly by Vibrio-related diseases, as well as indirectly through their interactions with other animal species, e.g. fish and shellfish, and with our environment, including influencing the health of our coastal waters and coral reefs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA