Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
mBio ; 12(6): e0293921, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34781738

ABSTRACT

Direct cell-to-cell spreading of Listeria monocytogenes requires the bacteria to induce actin-based finger-like membrane protrusions in donor host cells that are endocytosed through caveolin-rich membrane invaginations by adjacent receiving cells. An actin shell surrounds these endocytic sites; however, its structure, composition, and functional significance remain elusive. Here, we show that the formin mDia1, but surprisingly not the Arp2/3 complex, is enriched at the membrane invaginations generated by L. monocytogenes during HeLa and Jeg-3 cell infections. Electron microscopy reveals a band of linear actin filaments that run along the longitudinal axis of the invagination membrane. Mechanistically, mDia1 expression is vital for the assembly of this F-actin shell. mDia1 is also required for the recruitment of Filamin A, a caveola-associated F-actin cross-linking protein, and caveolin-1 to the invaginations. Importantly, mixed-cell infection assays show that optimal caveolin-based L. monocytogenes cell-to-cell spreading correlates with the formation of the linear actin filament-containing shell by mDia1. IMPORTANCE Listeria monocytogenes spreads from one cell to another to colonize tissues. This cell-to-cell movement requires the propulsive force of an actin-rich comet tail behind the advancing bacterium, which ultimately distends the host plasma membrane into a slender bacterium-containing membrane protrusion. These membrane protrusions induce a corresponding invagination in the membrane of the adjacent host cell. The host cell that receives the protrusion utilizes caveolin-based endocytosis to internalize the structures, and filamentous actin lines these membrane invaginations. Here, we set out to determine the structure and function of this filamentous actin "shell." We demonstrate that the formin mDia1, but not the Arp2/3 complex, localizes to the invaginations. Morphologically, we show that this actin is organized into linear arrays and not branched dendritic networks. Mechanistically, we show that the actin shell is assembled by mDia1 and that mDia1 is required for efficient cell-to-cell transfer of L. monocytogenes.


Subject(s)
Actins/metabolism , Cell Membrane/microbiology , Formins/metabolism , Listeria monocytogenes/physiology , Listeriosis/metabolism , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/microbiology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Filamins/genetics , Filamins/metabolism , Formins/genetics , HeLa Cells , Humans , Listeria monocytogenes/genetics , Listeriosis/genetics , Listeriosis/microbiology
2.
Anat Rec (Hoboken) ; 304(5): 919-938, 2021 05.
Article in English | MEDLINE | ID: mdl-33022122

ABSTRACT

The enteric bacterial pathogens Listeria monocytogenes (Listeria) and enteropathogenic Escherichia coli (EPEC) remodel the eukaryotic actin cytoskeleton during their disease processes. Listeria generate slender actin-rich comet/rocket tails to move intracellularly, and later, finger-like membrane protrusions to spread amongst host cells. EPEC remain extracellular, but generate similar actin-rich membranous protrusions (termed pedestals) to move atop the host epithelia. These structures are crucial for disease as diarrheal (and systemic) infections are significantly abrogated during infections with mutant strains that are unable to generate the structures. The current repertoire of host components enriched within these structures is vast and diverse. In this protein catalog, we and others have found that host actin crosslinkers, such as palladin and α-actinin-1, are routinely exploited. To expand on this list, we set out to investigate the distribution of PDLIM1, a scaffolding protein and binding partner of palladin and α-actinin-1, during bacterial infections. We show that PDLIM1 localizes to the site of initial Listeria entry into cells. Following this, PDLIM1 localizes to actin filament clouds surrounding immotile bacteria, and then colocalizes with actin once the comet/rocket tails are generated. Unlike palladin or α-actinin-1, PDLIM1 is maintained within the actin-rich core of membrane protrusions. Conversely, α-actinin-1, but not PDLIM1 (or palladin), is enriched at the membrane invagination that internalizes the Listeria-containing membrane protrusion. We also show that PDLIM1 is a component of the EPEC pedestal core and that its recruitment is dependent on the bacterial effector Tir. Our findings highlight PDLIM1 as another protein present within pathogen-induced actin-rich structures.


Subject(s)
Actin Cytoskeleton/metabolism , Escherichia coli Infections/metabolism , LIM Domain Proteins/metabolism , Transcription Factors/metabolism , Enteropathogenic Escherichia coli , HeLa Cells , Humans , Listeria monocytogenes
3.
Anat Rec (Hoboken) ; 304(7): 1400-1419, 2021 07.
Article in English | MEDLINE | ID: mdl-33099893

ABSTRACT

Bacterial pathogens cause disease by subverting the structure and function of their target host cells. Several foodborne agents such as Listeria monocytogenes (L. monocytogenes), Shigella flexneri (S. flexneri), Salmonella enterica serovar Typhimurium (S. Typhimurium) and enteropathogenic Escherichia coli (EPEC) manipulate the host actin cytoskeleton to cause diarrheal (and systemic) infections. During infections, these invasive and adherent pathogens hijack the actin filaments of their host cells and rearrange them into discrete actin-rich structures that promote bacterial adhesion (via pedestals), invasion (via membrane ruffles and endocytic cups), intracellular motility (via comet/rocket tails) and/or intercellular dissemination (via membrane protrusions and invaginations). We have previously shown that actin-rich structures generated by L. monocytogenes contain the host actin cross-linker α-actinin-4. Here we set out to examine α-actinin-4 during other key steps of the L. monocytogenes infectious cycle as well as characterize the subcellular distribution of α-actinin-4 during infections with other model actin-hijacking bacterial pathogens (S. flexneri, S. Typhimurium and EPEC). Although α-actinin-4 is absent at sites of initial L. monocytogenes invasion, we show that it is a new component of the membrane invaginations formed during secondary infections of neighboring host cells. Importantly, we reveal that α-actinin-4 also localizes to the major actin-rich structures generated during cell culture infections with S. flexneri (comet/rocket tails and membrane protrusions), S. Typhimurium (membrane ruffles) and EPEC (pedestals). Taken together, these findings suggest that α-actinin-4 is a host factor that is exploited by an assortment of actin-hijacking bacterial pathogens.


Subject(s)
Actin Cytoskeleton/metabolism , Actinin/metabolism , Cell Membrane/metabolism , Epithelial Cells/metabolism , Caco-2 Cells , Enteropathogenic Escherichia coli , HeLa Cells , Humans , Listeria monocytogenes
4.
mBio ; 11(1)2020 01 21.
Article in English | MEDLINE | ID: mdl-31964732

ABSTRACT

Listeria monocytogenes moves from one cell to another using actin-rich membrane protrusions that propel the bacterium toward neighboring cells. Despite cholesterol being required for this transfer process, the precise host internalization mechanism remains elusive. Here, we show that caveolin endocytosis is key to this event as bacterial cell-to-cell transfer is severely impaired when cells are depleted of caveolin-1. Only a subset of additional caveolar components (cavin-2 and EHD2) are present at sites of bacterial transfer, and although clathrin and the clathrin-associated proteins Eps15 and AP2 are absent from the bacterial invaginations, efficient L. monocytogenes spreading requires the clathrin-interacting protein epsin-1. We also directly demonstrated that isolated L. monocytogenes membrane protrusions can trigger the recruitment of caveolar proteins in a neighboring cell. The engulfment of these bacterial and cytoskeletal structures through a caveolin-based mechanism demonstrates that the classical nanometer-scale theoretical size limit for this internalization pathway is exceeded by these bacterial pathogens.IMPORTANCEListeria monocytogenes moves from one cell to another as it disseminates within tissues. This bacterial transfer process depends on the host actin cytoskeleton as the bacterium forms motile actin-rich membranous protrusions that propel the bacteria into neighboring cells, thus forming corresponding membrane invaginations. Here, we examine these membrane invaginations and demonstrate that caveolin-1-based endocytosis is crucial for efficient bacterial cell-to-cell spreading. We show that only a subset of caveolin-associated proteins (cavin-2 and EHD2) are involved in this process. Despite the absence of clathrin at the invaginations, the classical clathrin-associated protein epsin-1 is also required for efficient bacterial spreading. Using isolated L. monocytogenes protrusions added onto naive host cells, we demonstrate that actin-based propulsion is dispensable for caveolin-1 endocytosis as the presence of the protrusion/invagination interaction alone triggers caveolin-1 recruitment in the recipient cells. Finally, we provide a model of how this caveolin-1-based internalization event can exceed the theoretical size limit for this endocytic pathway.


Subject(s)
Caveolin 1/metabolism , Host-Pathogen Interactions , Listeria monocytogenes/physiology , Listeriosis/metabolism , Listeriosis/microbiology , Animals , Biomarkers , Cell Line , Fluorescent Antibody Technique , Humans
5.
Anat Rec (Hoboken) ; 303(7): 1859-1864, 2020 07.
Article in English | MEDLINE | ID: mdl-31595676

ABSTRACT

Klebsiella pneumoniae has become a growing concern within hospitals due to multidrug resistant strains and increasing mortality rates. Recently, we showed that at the subcellular level, K. pneumoniae compromises the integrity of the epithelia by disassembling the microtubule networks of cells through the actions of katanin microtubule severing proteins. In this study, we report on the observation that mitotic cells are targeted by K. pneumoniae and that during infections, the katanin proteins are excluded from the microtubule organizing centers of dividing cells, resulting in the alteration of the microtubule cytoskeleton. Anat Rec, 2019. © 2019 American Association for Anatomy Anat Rec, 303:1859-1864, 2020. © 2019 American Association for Anatomy.


Subject(s)
Katanin/metabolism , Klebsiella pneumoniae/metabolism , Lung/microbiology , Microtubules/metabolism , Mitosis/physiology , Cell Line , Humans , Lung/cytology , Lung/metabolism
6.
Microbiol Res ; 229: 126325, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31563838

ABSTRACT

Edwardsiella bacteria cause economic losses to a variety of commercially important fish globally. Human infections are rare and result in a gastroenteritis-like illness. Because these bacteria are evolutionarily related to other Enterobacteriaceae and the host cytoskeleton is a common target of enterics, we hypothesized that Edwardsiella may cause similar phenotypes. Here we use HeLa and Caco-2 infection models to show that microtubules are severed during the late infections. This microtubule alteration phenotype was not dependant on the type III or type VI secretion system (T3SS and T6SS) of the bacteria as ΔT3SS and ΔT6SS mutants of E. piscicida EIB202 and E. tarda ATCC15947 that lacks both also caused microtubule disassembly. Immunolocalization experiments showed the host katanin catalytic subunits A1 and A like 1 proteins at regions of microtubule severing, suggesting their involvement in the microtubule disassembly events. To identify bacterial components involved in this phenotype, we screened a 2,758 transposon library of E. piscicida EIB202 and found that 4 single mutations in the atpFHAGDC operon disrupted microtubule disassembly in HeLa cells. We then constructed three atp deletion mutants; they all could not disassemble host microtubules. This work provides the first clear evidence of host cytoskeletal alterations during Edwardsiella infections.


Subject(s)
Edwardsiella/physiology , Enterobacteriaceae Infections/veterinary , Epithelial Cells/metabolism , Fish Diseases/metabolism , Microtubules/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Caco-2 Cells , Edwardsiella/genetics , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Epithelial Cells/microbiology , Fish Diseases/microbiology , Gene Expression Regulation, Bacterial , HeLa Cells , Host-Pathogen Interactions , Humans , Operon , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism
7.
Anat Rec (Hoboken) ; 302(12): 2224-2232, 2019 12.
Article in English | MEDLINE | ID: mdl-31443124

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) and Salmonella enterica serovar Typhimurium (S. Typhimurium) are highly infectious gastrointestinal human pathogens. These microbes inject bacterial-derived effector proteins directly into the host cell cytosol as part of their disease processes. A common host subcellular target of these pathogens is the actin cytoskeleton, which is commandeered by the bacteria and is used during their attachment onto (EPEC) or invasion into (S. Typhimurium) the host cells. We previously demonstrated that the host enzyme cyclophilin A (CypA) is recruited to the actin-rich regions of EPEC pedestals and S. Typhimurium membrane ruffles. To further expand the growing catalogue of host proteins usurped by actin-hijacking bacteria, we examined the host plasma membrane protein and cognate receptor of CypA, CD147, during EPEC and S. Typhimurium infections. Here, we show that CD147 is enriched at the basolateral regions of pedestals but, unlike CypA, it is absent from their actin-rich core. We show that the CD147 recruitment to these areas requires EPEC pedestal formation and not solely bacteria-host cell contact. Additionally, we demonstrate that the depletion of CD147 by siRNA does not alter the formation of pedestals. Finally, we show that CD147 is also a component of actin-rich membrane ruffles generated during S. Typhimurium invasion of host cells. Collectively, our findings establish CD147 as another host component present at dynamic actin-rich structures formed during bacterial infections. Anat Rec, 302:2224-2232, 2019. © 2019 American Association for Anatomy.


Subject(s)
Actin Cytoskeleton/metabolism , Basigin/metabolism , Cell Membrane/metabolism , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Salmonella Infections/metabolism , Salmonella enterica/metabolism , Escherichia coli Infections/microbiology , HeLa Cells , Humans , Salmonella Infections/microbiology
8.
Cell Mol Life Sci ; 76(20): 4165-4178, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31076805

ABSTRACT

Efficient cell-to-cell transfer of Listeria monocytogenes (L. monocytogenes) requires the proper formation of actin-rich membrane protrusions. To date, only the host proteins ezrin, the binding partner of ezrin, CD44, as well as cyclophilin A (CypA) have been identified as crucial components for L. monocytogenes membrane protrusion stabilization and, thus, efficient cell-to-cell movement of the microbes. Here, we examine the classical binding partner of CypA, CD147, and find that this membrane protein is also hijacked by the bacteria for their cellular dissemination. CD147 is enriched at the plasma membrane surrounding the membrane protrusions as well as the resulting invaginations generated in neighboring cells. In cells depleted of CD147, these actin-rich structures appear similar to those generated in CypA depleted cells as they are significantly shorter and more contorted as compared to their straighter counterparts formed in wild-type control cells. The presence of malformed membrane protrusions hampers the ability of L. monocytogenes to efficiently disseminate from CD147-depleted cells. Our findings uncover another important host protein needed for L. monocytogenes membrane protrusion formation and efficient microbial dissemination.


Subject(s)
Basigin/genetics , Cell Membrane/microbiology , Host-Pathogen Interactions/genetics , Listeria monocytogenes/physiology , Shigella flexneri/physiology , A549 Cells , Actins/genetics , Actins/metabolism , Animals , Basigin/antagonists & inhibitors , Basigin/metabolism , Caco-2 Cells , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cyclophilin A/deficiency , Cyclophilin A/genetics , Endocytosis , Fibroblasts/microbiology , Fibroblasts/ultrastructure , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/ultrastructure , Mice , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shigella flexneri/pathogenicity , Shigella flexneri/ultrastructure , Signal Transduction
9.
J Infect Dis ; 219(1): 145-153, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29733369

ABSTRACT

Background: Listeria generate actin-rich tubular protrusions at the plasma membrane that propel the bacteria into neighboring cells. The precise molecular mechanisms governing the formation of these protrusions remain poorly defined. Methods: In this study, we demonstrate that the prolyl cis-trans isomerase (PPIase) cyclophilin A (CypA) is hijacked by Listeria at membrane protrusions used for cell-to-cell spreading. Results: Cyclophilin A localizes within the F-actin of these structures and is crucial for their proper formation, as cells depleted of CypA have extended actin-rich structures that are misshaped and are collapsed due to changes within the F-actin network. The lack of structural integrity within the Listeria membrane protrusions hampers the microbes from spreading from CypA null cells. Conclusions: Our results demonstrate a crucial role for CypA during Listeria infections.


Subject(s)
Cell Surface Extensions/metabolism , Cell Surface Extensions/microbiology , Cyclophilin A/metabolism , Listeria/metabolism , Listeriosis/metabolism , A549 Cells , Actin Cytoskeleton/metabolism , Actins/metabolism , Actins/ultrastructure , Cell Membrane/metabolism , Cell Membrane/microbiology , Cell Surface Extensions/ultrastructure , Epithelial Cells/metabolism , Epithelial Cells/microbiology , HeLa Cells , Host-Pathogen Interactions/physiology , Humans , Listeria/pathogenicity , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Peptidylprolyl Isomerase/metabolism
10.
Anat Rec (Hoboken) ; 301(12): 2086-2094, 2018 12.
Article in English | MEDLINE | ID: mdl-30312007

ABSTRACT

Salmonella enterica serovar Typhimurium (S. Typhimurium), enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) commandeer the actin cytoskeleton of their host cells as a crucial step in their infectious processes. These pathogens depend on the injection of their own effectors directly into target host cells in order to usurp cellular signaling pathways that lead to morphological actin rearrangements in those cells. Here we show that the PPIase Cyclophilin A (CypA) is a novel component of S. Typhimurium-induced membrane ruffles and functions to restrict bacterial invasion levels, as in cells depleted of CypA, bacterial loads increase. We also demonstrate that CypA requires the EPEC effector Tir as well as pedestal formation for its recruitment to bacterial attachment sites and that its presence at pedestals also holds during EHEC infections. Finally, we demonstrate that CypA is found at lamellipodia; actin-rich structures at the leading edge of motile cells. Our findings further establish CypA as a component of dynamic actin-rich structures formed during bacterial infections and within cells in general. Anat Rec, 301:2086-2094, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cyclophilin A/metabolism , Escherichia coli/metabolism , Salmonella/metabolism , Actin Cytoskeleton/chemistry , Actins/analysis , Animals , Cyclophilin A/analysis , HeLa Cells , Humans , Mice , Potoroidae
11.
Anat Rec (Hoboken) ; 301(12): 1986-1990, 2018 12.
Article in English | MEDLINE | ID: mdl-30312025

ABSTRACT

The actin cytoskeleton has long been recognized as a crucial sub-cellular filament system that is responsible for governing fundamental events ranging from cell division and muscle contraction to whole cell motility and the maintenance of tissue integrity. Consequently, it is not surprising that this network is the focus of over 100,000 different manuscripts. Alterations in the actin cytoskeleton lead to an assortment of diseases and serve as a target for a variety of pathogens. Here we have brought together a collection of primary research articles and reviews that underscore the broad influence this filament system has on organisms. Anat Rec, 301:1986-1990, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Actin Cytoskeleton/physiology , Actin Cytoskeleton/ultrastructure , Actins/physiology , Actins/ultrastructure , Cell Movement/physiology , Actin Cytoskeleton/chemistry , Actins/analysis , Animals , Humans , Microfilament Proteins/analysis , Microfilament Proteins/ultrastructure
12.
Anat Rec (Hoboken) ; 301(12): 2095-2102, 2018 12.
Article in English | MEDLINE | ID: mdl-30312532

ABSTRACT

Enteropathogenic Escherichia coli (EPEC), Salmonella typhimurium, and Listeria monocytogenes usurp the actin cytoskeleton for their attachment, internalization and transport within and amongst infected cells. To try to gain a greater understanding of the molecular components utilized by these microbes during their infections we previously concentrated actin-rich structures generated during EPEC infections (called pedestals) and identified the heat shock cognate 70 protein (Hsc70) as a potential candidate. This multifunctional protein classically acts as a chaperone for the proper folding of a variety of proteins and is involved in uncoating clathrin from coated pits. Here we demonstrated that Hsc70 is recruited to actin structures generated during EPEC, Listeria and Salmonella infections, but not to the same location as clathrin. Anat Rec, 301:2095-2102, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Actins/metabolism , HSC70 Heat-Shock Proteins/metabolism , Listeria monocytogenes/metabolism , Actins/analysis , Animals , HSC70 Heat-Shock Proteins/analysis , HeLa Cells , Humans , Listeria monocytogenes/chemistry
13.
Anat Rec (Hoboken) ; 301(12): 2103-2111, 2018 12.
Article in English | MEDLINE | ID: mdl-30312538

ABSTRACT

The ingestion of enteropathogenic Escherichia coli (EPEC), Listeria monocytogenes, or Salmonella enterica serovar Typhimurium leads to their colonization of the intestinal lumen, which ultimately causes an array of ailments ranging from diarrhea to bacteremia. Once in the intestines, these microbes generate various actin-rich structures to attach, invade, or move within the host intestinal epithelial cells. Although an assortment of actin-associated proteins has been identified to varying degrees at these structures, the localization of many actin stabilizing proteins have yet to be analyzed. Here, we examined the recruitment of the actin-associated proteins, calponin 1 and 2 at EPEC pedestals, L. monocytogenes actin clouds, comet tails and listeriopods, and S. Typhimurium membrane ruffles. In other systems, calponins are known to bind to and stabilize actin filaments. In EPEC pedestals, calponin 1 was recruited uniformly throughout the structures while calponin 2 was enriched at the apical tip. During L. monocytogenes infections, calponin 1 was found through all the actin-rich structures generated by the bacteria, while calponin 2 was only present within actin-rich structures formed by L. monocytogenes near the host cell membrane. Finally, both calponins were found within S. Typhimurium-generated membrane ruffles. Taken together, we have shown that although calponin 1 is recruited to actin-rich structures formed by the three bacteria, calponin 2 is specifically recruited to only membrane-bound actin-rich structures formed by the bacteria. Thus, our findings suggest that calponin 2 is a novel marker for membrane-bound actin structures formed by pathogenic bacteria. Anat Rec, 301:2103-2111, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Actins/metabolism , Calcium-Binding Proteins/metabolism , Enteropathogenic Escherichia coli/metabolism , Listeria monocytogenes/metabolism , Microfilament Proteins/metabolism , Salmonella enterica/metabolism , Actins/analysis , Caco-2 Cells , Calcium-Binding Proteins/analysis , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Enteropathogenic Escherichia coli/chemistry , Humans , Listeria monocytogenes/chemistry , Microfilament Proteins/analysis , Salmonella enterica/chemistry , Calponins
14.
mBio ; 9(2)2018 04 10.
Article in English | MEDLINE | ID: mdl-29636431

ABSTRACT

Palladin is an important component of motile actin-rich structures and nucleates branched actin filament arrays in vitro Here we examine the role of palladin during Listeria monocytogenes infections in order to tease out novel functions of palladin. We show that palladin is co-opted by L. monocytogenes during its cellular entry and intracellular motility. Depletion of palladin resulted in shorter and misshapen comet tails, and when actin- or VASP-binding mutants of palladin were overexpressed in cells, comet tails disintegrated or became thinner. Comet tail thinning resulted in parallel actin bundles within the structures. To determine whether palladin could compensate for the Arp2/3 complex, we overexpressed palladin in cells treated with the Arp2/3 inhibitor CK-666. In treated cells, bacterial motility could be initiated and maintained when levels of palladin were increased. To confirm these findings, we utilized a cell line depleted of multiple Arp2/3 complex subunits. Within these cells, L. monocytogenes failed to generate comet tails. When palladin was overexpressed in this Arp2/3 functionally null cell line, the ability of L. monocytogenes to generate comet tails was restored. Using purified protein components, we demonstrate that L. monocytogenes actin clouds and comet tails can be generated (in a cell-free system) by palladin in the absence of the Arp2/3 complex. Collectively, our results demonstrate that palladin can functionally replace the Arp2/3 complex during bacterial actin-based motility.IMPORTANCE Structures containing branched actin filaments require the Arp2/3 complex. One of the most commonly used systems to study intracellular movement generated by Arp2/3-based actin motility exploits actin-rich comet tails made by Listeria Using these infections together with live imaging and cell-free protein reconstitution experiments, we show that another protein, palladin, can be used in place of Arp2/3 to form actin-rich structures. Additionally, we show that palladin is needed for the structural integrity of comet tails as its depletion or mutation of critical regions causes dramatic changes to comet tail organization. These findings are the first to identify a protein that can functionally replace the Arp2/3 complex and have implications for all actin-based structures thought to exclusively use that complex.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Cytoskeletal Proteins/metabolism , Endocytosis , Host-Pathogen Interactions , Listeria monocytogenes/physiology , Locomotion , Phosphoproteins/metabolism , Actin-Related Protein 2-3 Complex/antagonists & inhibitors , Animals , Cell Line , Humans , Indoles/metabolism
15.
Acta Crystallogr D Struct Biol ; 73(Pt 10): 814-821, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28994410

ABSTRACT

Francisella tularensis is a potent human pathogen that invades and survives in macrophage and epithelial cells. Two identical proteins, FTT_0924 from F. tularensis subsp. tularensis and FTL_1286 from F. tularensis subsp. holarctica LVS, have previously been identified as playing a role in protection of the bacteria from osmotic shock and its survival in macrophages. FTT_0924 has been shown to localize to the inner membrane, with its C-terminus exposed to the periplasm. Here, crystal structures of the F. novicida homologue FTN_0802, which we call FvfA, in two crystal forms are reported at 1.8 Šresolution. FvfA differs from FTT_0924 and FTL_1286 by a single amino acid. FvfA has a DUF1471 fold that closely resembles the Escherichia coli outer membrane lipoprotein RscF, a component of a phosphorelay pathway involved in protecting bacteria from outer membrane perturbation. The structural and functional similarities and differences between these proteins and their implications for F. tularensis pathogenesis are discussed.


Subject(s)
Bacterial Proteins/chemistry , Francisella tularensis/chemistry , Virulence Factors/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Sequence Alignment , Tularemia/microbiology
16.
J Proteome Res ; 15(5): 1613-22, 2016 05 06.
Article in English | MEDLINE | ID: mdl-27018634

ABSTRACT

Enteropathogenic and enterohemorrhagic Escherichia coli cause enteric diseases resulting in significant morbidity and mortality worldwide. These pathogens remain extracellular and translocate a set of type III secreted effector proteins into host cells to promote bacterial virulence. Effectors manipulate host cell pathways to facilitate infection by interacting with a variety of host targets, yet the binding partners and mechanism of action of many effectors remain elusive. We performed a mass spectrometry screen to identify host targets for a library of effectors. We found five known effector targets and discovered four novel interactions. Interestingly, we identified multiple effectors that interacted with the microtubule associated protein, ensconsin. Using co-immunoprecipitations, we confirmed that NleB1 and EspL interacted with ensconsin in a region that corresponded to its microtubule binding domain. Ensconsin is an essential cofactor of kinesin-1 that is required for intracellular trafficking, and we demonstrated that intracellular trafficking was severely disrupted during wild type EPEC infections but not during infections with ΔnleB1 or ΔespL mutants. Our findings demonstrate the efficacy of quantitative proteomics for identifying effector-host protein interactions and suggest that vesicular trafficking is a crucial cellular process that may be targeted by NleB1 and EspL through their interaction with ensconsin.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/pathogenicity , Host-Pathogen Interactions , Type III Secretion Systems/metabolism , Virulence Factors/metabolism , Cell Line , Humans , Immunoprecipitation , Mass Spectrometry , Microtubule-Associated Proteins/metabolism , Protein Binding , Type III Secretion Systems/chemistry
17.
Cell Tissue Res ; 363(2): 449-59, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26239909

ABSTRACT

Francisella novicida is a surrogate pathogen commonly used to study infections by the potential bioterrorism agent, Francisella tularensis. One of the primary sites of Francisella infections is the liver where >90% of infected cells are hepatocytes. It is known that once Francisella enter cells it occupies a membrane-bound compartment, the Francisella-containing vacuole (FCV), from which it rapidly escapes to replicate in the cytosol. Recent work examining the Francisella disulfide bond formation (Dsb) proteins, FipA and FipB, have demonstrated that these proteins are important during the Francisella infection process; however, details as to how the infections are altered in epithelial cells have remained elusive. To identify the stage of the infections where these Dsbs might act during epithelial infections, we exploited a hepatocyte F. novicida infection model that we recently developed. We found that F. novicida ΔfipA-infected hepatocytes contained bacteria clustered within lysosome-associated membrane protein 1-positive FCVs, suggesting that FipA is involved in the escape of F. novicida from its vacuole. Our morphological evidence provides a tangible link as to how Dsb FipA can influence Francisella infections.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Francisella/physiology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Animals , Bacterial Proteins/genetics , Cell Line , Epithelial Cells/ultrastructure , Francisella/ultrastructure , Hepatocytes/microbiology , Hepatocytes/pathology , Lysosomal Membrane Proteins/metabolism , Mice, Inbred BALB C , Mutation/genetics , Vacuoles/metabolism , Vacuoles/ultrastructure
18.
J Proteome Res ; 14(6): 2520-7, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25907766

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) co-opt host signaling pathways and recruit numerous host proteins to motile morphological structures, called pedestals, at sites of bacterial attachment. These pedestals are hallmarks of EPEC-based disease, and the identification and characterization of the functions of pedestal proteins continue to steadily increase. To identify additional constituents in an unbiased manner, we developed a strategy where EPEC pedestals were elongated artificially, severed, and then concentrated prior to their analysis by mass spectrometry (MS)-based proteomics. We identified >90 unique mammalian proteins over multiple experimental trials from our preparations. Seventeen predicted molecules were significantly higher in abundance (p < 0.05) when compared to both the negative controls and sample means. Validation of two identified proteins (cyclophilin A [nonactin-associated] and transgelin [actin-associated]) by immunolocalization was used to confirm our analysis, and both showed enrichment at EPEC pedestals. The EPEC pedestal concentration technique developed here together with the identification of novel pedestal proteins not only provides a resource for the further characterization of molecular components within these structures but also demonstrates that EPEC pedestals can be used as a model system for the identification of novel functions of proteins not normally thought to be at actin-based structures.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Mass Spectrometry/methods , Proteomics , HeLa Cells , Humans
19.
Int J Biochem Cell Biol ; 44(12): 2223-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22999844

ABSTRACT

Ubiquitylation is a widespread post-translational global regulatory system that is essential for the proper functioning of various cellular events. Recent studies have shown that certain types of Escherichia coli can exploit specific aspects of the ubiquitylation system to influence downstream targets. Despite these findings, examination of the effects pathogenic E. coli have on the overall host ubiquitylation system remain unexplored. To study the impact that pathogenic E. coli have on the ubiquitylation levels of host proteins during infections, we analyzed the entire ubiquitylation system during enteropathogenic E. coli infections of cultured cells. We found that these microbes caused a dramatic decrease in ubiquitylated host proteins during these infections. This occurred with a concomitant reduction in the expression of essential E1 activating enzymes in the host, which are integral for the initiation of the ubiquitylation cascade. Control of host E1 enzyme levels was dependent on the E. coli adherence factor plasmid which acted on host aspartyl proteases within enteropathogenic E. coli. Hijacking of the ubiquitylation system did not require the plasmid-encoded regulator or bundle forming pilus expression, as enteropathogenic E. coli mutated in those factors did not revert the ubiquitylation of host proteins or the abundance of E1 enzyme proteins to uninfected levels. Our work shows that E. coli have developed strategies to usurp post-translational systems by targeting crucial enzymes. The ability of enteropathogenic E. coli to inactivate host protein ubiquitylation could enable more efficient effector protein functionality, providing increased bacterial control of host cells during enteropathogenic E. coli pathogenesis.


Subject(s)
Aspartic Acid Proteases/metabolism , Enteropathogenic Escherichia coli/enzymology , Gene Expression Regulation, Enzymologic , Plasmids/physiology , Ubiquitin-Activating Enzymes/metabolism , Bacterial Secretion Systems , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/physiology , Epithelial Cells/enzymology , Epithelial Cells/microbiology , HeLa Cells , Host-Pathogen Interactions , Humans , Polyubiquitin/metabolism , Thiolester Hydrolases/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitinated Proteins/metabolism , Ubiquitination
20.
Anat Rec (Hoboken) ; 295(8): 1230-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22648930

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) is an extracellular pathogen that alters many host subcellular components during its infectious processes. We have previously shown that EPEC hijacks a large assortment of host cell endocytic components and uses these proteins to form protruding structures called "pedestals" rather than triggering internalization of the bacteria. Other invasive pathogens that also recruit similar endocytic components have been shown to enter their host cells on the ubiquitylation of their host cell receptors. Therefore, we hypothesize that EPEC remains extracellular by maintaining its receptor, translocated intimin receptor (Tir), in an unubiquitylated state. Using immunoprecipitation-Western blots, we demonstrate no association of ubiquitin with Tir. To further elucidate the effect Tir ubiquitylation would have on EPEC during their infections, we engineered Tir-ubiquitin fusion constructs, expressed them in host epithelial cells, and infected them with Δtir EPEC. We found these cells induced a significant increase in EPEC invasion as compared with cells that expressed the Tir construct that lacked ubiquitin conjugation. Our results indicate that the lack of EPEC receptor ubiquitylation is a contributing factor that these microbes use to prevent their internalization into epithelial cells.


Subject(s)
Cell Membrane/metabolism , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Enteropathogenic Escherichia coli/growth & development , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoprecipitation , Protein Binding , Protein Transport , Receptors, Cell Surface/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...