1
|
Qin J, Hong Y, Morona R, Totsika M. Cysteine-Dependent Conformational Heterogeneity of Shigella flexneri Autotransporter IcsA and Implications of Its Function. Microbiol Spectr 2022; 10:e0341022. [PMID: 36374106 PMCID: PMC9769942 DOI: 10.1128/spectrum.03410-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/30/2022] [Indexed: 11/16/2022] Open
Abstract
Shigella IcsA is a versatile surface virulence factor required for early and late pathogenesis stages extracellularly and intracellularly. Despite IcsA serving as a model Type V secretion system (T5SS) autotransporter to study host-pathogen interactions, its detailed molecular architecture is poorly understood. Recently, IcsA was found to switch to a different conformation for its adhesin activity upon sensing the host stimuli by Shigella Type III secretion system (T3SS). Here, we reported that the single cysteine residue (C130) near the N terminus of the IcsA passenger had a role in IcsA adhesin activity. We also showed that the IcsA passenger (IcsAp) existed in multiple conformations, and the conformation populations were influenced by a central pair of cysteine residues (C375 and C379), which was not previously reported for any Type V autotransporter passengers. Disruption of either or both central cysteine residues altered the exposure of IcsA epitopes to polyclonal anti-IcsA antibodies previously shown to block Shigella adherence, yet without loss of IcsA intracellular functions in actin-based motility (ABM). Anti-IcsA antibody reactivity was restored when the IcsA-paired cysteine substitution mutants were expressed in an ΔipaD background with a constitutively active T3SS, highlighting an interplay between T3SS and T5SS. The work here uncovered a novel molecular switch empowered by a centrally localized, short-spaced cysteine pair in the Type V autotransporter IcsA that ensured conformational heterogeneity to aid IcsA evasion of host immunity. IMPORTANCE Shigella species are the leading cause of diarrheal-related death globally by causing bacillary dysentery. The surface virulence factor IcsA, which is essential for Shigella pathogenesis, is a unique multifunctional autotransporter that is responsible for cell adhesion, and actin-based motility, yet detailed mechanistic understanding is lacking. Here, we showed that the three cysteine residues in IcsA contributed to the protein's distinct functions. The N-terminal cysteine residue within the IcsA passenger domain played a role in adhesin function, while a centrally localized cysteine pair provided conformational heterogeneity that resulted in IcsA molecules with different reactivity to adhesion-blocking anti-IcsA antibodies. In synergy with the Type III secretion system, this molecular switch preserved biological function in distinct IcsA conformations for cell adhesion, actin-based motility, and autophagy escape, providing a potential strategy by which Shigella evades host immunity and targets this essential virulence factor.
Collapse
Affiliation(s)
- Jilong Qin
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Yaoqin Hong
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Renato Morona
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
2
|
Kramer DA, Piper HK, Chen B. WASP family proteins: Molecular mechanisms and implications in human disease. Eur J Cell Biol 2022; 101:151244. [PMID: 35667337 PMCID: PMC9357188 DOI: 10.1016/j.ejcb.2022.151244] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023] Open
Abstract
Proteins of the Wiskott-Aldrich syndrome protein (WASP) family play a central role in regulating actin cytoskeletal dynamics in a wide range of cellular processes. Genetic mutations or misregulation of these proteins are tightly associated with many diseases. The WASP-family proteins act by transmitting various upstream signals to their conserved WH2-Central-Acidic (WCA) peptide sequence at the C-terminus, which in turn binds to the Arp2/3 complex to stimulate the formation of branched actin networks at membranes. Despite this common feature, the regulatory mechanisms and cellular functions of distinct WASP-family proteins are very different. Here, we summarize and clarify our current understanding of WASP-family proteins and how disruption of their functions is related to human disease.
Collapse
Affiliation(s)
- Daniel A Kramer
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Hannah K Piper
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Baoyu Chen
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA.
| |
Collapse
|
3
|
Dhanda AS, Guttman JA. Localization of host endocytic and actin-associated proteins during Shigella flexneri intracellular motility and intercellular spreading. Anat Rec (Hoboken) 2022; 306:1088-1110. [PMID: 35582740 DOI: 10.1002/ar.24955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022]
Abstract
Shigella flexneri (S. flexneri), the causative agent of bacillary dysentery, uses an effector-mediated strategy to hijack host cells and cause disease. To propagate and spread within human tissues, S. flexneri bacteria commandeer the host actin cytoskeleton to generate slender actin-rich comet tails to move intracellularly, and later, plasma membrane actin-based protrusions to move directly between adjacent host cells. To facilitate intercellular bacterial spreading, large micron-sized endocytic-like membrane invaginations form at the periphery of neighboring host cells that come into contact with S. flexneri-containing membrane protrusions. While S. flexneri comet tails and membrane protrusions consist primarily of host actin cytoskeletal proteins, S. flexneri membrane invaginations remain poorly understood with only clathrin and the clathrin adapter epsin-1 localized to the structures. Tangentially, we recently reported that Listeria monocytogenes, another actin-hijacking pathogen, exploits an assortment of caveolar and actin-bundling proteins at their micron-sized membrane invaginations formed during their cell-to-cell movement. Thus, to further characterize the S. flexneri disease process, we set out to catalog the distribution of a variety of actin-associated and caveolar proteins during S. flexneri actin-based motility and cell-to-cell spreading. Here we show that actin-associated proteins found at L. monocytogenes comet tails and membrane protrusions mimic those present at S. flexneri comet tails with the exception of α-actinins 1 and 4, which were shed from S. flexneri membrane protrusions. We also demonstrate that all known host endocytic components found at L. monocytogenes membrane invaginations are also present at those formed during S. flexneri infections.
Collapse
Affiliation(s)
- Aaron Singh Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian Andrew Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
4
|
Narayan C, Kant V, Mahajan JK, Mohan B, Taneja N. Differential dendritic immune cell responses to infection with various serotypes of Shigella. Indian J Med Microbiol 2022; 40:217-222. [DOI: 10.1016/j.ijmmb.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/28/2022]
|
5
|
Köseoğlu VK, Jones MK, Agaisse H. The type 3 secretion effector IpgD promotes S. flexneri dissemination. PLoS Pathog 2022; 18:e1010324. [PMID: 35130324 PMCID: PMC8853559 DOI: 10.1371/journal.ppat.1010324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/17/2022] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
The bacterial pathogen Shigella flexneri causes 270 million cases of bacillary dysentery worldwide every year, resulting in more than 200,000 deaths. S. flexneri pathogenic properties rely on its ability to invade epithelial cells and spread from cell to cell within the colonic epithelium. This dissemination process relies on actin-based motility in the cytosol of infected cells and formation of membrane protrusions that project into adjacent cells and resolve into double-membrane vacuoles (DMVs) from which the pathogen escapes, thereby achieving cell-to-cell spread. S. flexneri dissemination is facilitated by the type 3 secretion system (T3SS) through poorly understood mechanisms. Here, we show that the T3SS effector IpgD facilitates the resolution of membrane protrusions into DMVs during S. flexneri dissemination. The phosphatidylinositol 4-phosphatase activity of IpgD decreases PtdIns(4,5)P2 levels in membrane protrusions, thereby counteracting de novo cortical actin formation in protrusions, a process that restricts the resolution of protrusions into DMVs. Finally, using an infant rabbit model of shigellosis, we show that IpgD is required for efficient cell-to-cell spread in vivo and contributes to the severity of dysentery. The intracellular pathogen Shigella flexneri is the causative agent of bacillary dysentery (blood in stool). Invasion of epithelial cells and cell-to-cell spread are critical determinants of S. flexneri pathogenesis. Cell-to-cell spread relies on the formation of membrane protrusions that project into adjacent cells and resolve into vacuoles. The molecular mechanisms supporting this dissemination process are poorly understood. In this study, we show that S. flexneri employs the phosphatidylinositol phosphatase activity of the T3SS effector protein IpgD to manipulate phosphoinositides in the protrusion membrane. Manipulation of phosphoinositide signaling restricts the formation of actin networks underneath the protrusion membrane, which would otherwise prevent the scission of protrusions into vacuoles. We also demonstrate that IpgD is required for efficient dissemination in the colon of infant rabbits and contributes to the severity of disease. This study exemplifies how manipulation of phosphoinositide signaling by intracellular pathogens supports bacterial pathogenesis.
Collapse
Affiliation(s)
- Volkan K. Köseoğlu
- Department of Microbiology, Immunology, and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Marieke K. Jones
- Claude Moore Health Sciences Library, University of Virginia, Charlottesville, Virginia, United States of America
| | - Hervé Agaisse
- Department of Microbiology, Immunology, and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
6
|
Kutsch M, Coers J. Human guanylate binding proteins: nanomachines orchestrating host defense. FEBS J 2021; 288:5826-5849. [PMID: 33314740 PMCID: PMC8196077 DOI: 10.1111/febs.15662] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/27/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Disease-causing microorganisms not only breach anatomical barriers and invade tissues but also frequently enter host cells, nutrient-enriched environments amenable to support parasitic microbial growth. Protection from many infectious diseases is therefore reliant on the ability of individual host cells to combat intracellular infections through the execution of cell-autonomous defense programs. Central players in human cell-autonomous immunity are members of the family of dynamin-related guanylate binding proteins (GBPs). The importance of these interferon-inducible GTPases in host defense to viral, bacterial, and protozoan pathogens has been established for some time; only recently, cell biological and biochemical studies that largely focused on the prenylated paralogs GBP1, GBP2, and GBP5 have provided us with robust molecular frameworks for GBP-mediated immunity. Specifically, the recent characterization of GBP1 as a bona fide pattern recognition receptor for bacterial lipopolysaccharide (LPS) disrupting the integrity of bacterial outer membranes through LPS aggregation, the discovery of a link between hydrolysis-induced GMP production by GBP1 and inflammasome activation, and the classification of GBP2 and GBP5 as inhibitors of viral envelope glycoprotein processing via suppression of the host endoprotease furin have paved the way for a vastly improved conceptual understanding of the molecular mechanisms by which GBP nanomachines execute cell-autonomous immunity. The herein discussed models incorporate our current knowledge of the antimicrobial, proinflammatory, and biochemical properties of human GBPs and thereby provide testable hypotheses that will guide future studies into the intricacies of GBP-controlled host defense and their role in human disease.
Collapse
Affiliation(s)
- Miriam Kutsch
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina 22710, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina 22710, USA
- Department of Immunology, Duke University Medical Center, Durham, North Carolina 22710, USA
| |
Collapse
|
7
|
Kühn S, Bergqvist J, Gil M, Valenzuela C, Barrio L, Lebreton S, Zurzolo C, Enninga J. Actin Assembly around the Shigella-Containing Vacuole Promotes Successful Infection. Cell Rep 2021; 31:107638. [PMID: 32402280 PMCID: PMC7225751 DOI: 10.1016/j.celrep.2020.107638] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/10/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
The enteroinvasive bacterium Shigella flexneri forces its uptake into non-phagocytic host cells through the translocation of T3SS effectors that subvert the actin cytoskeleton. Here, we report de novo actin polymerization after cellular entry around the bacterium-containing vacuole (BCV) leading to the formation of a dynamic actin cocoon. This cocoon is thicker than any described cellular actin structure and functions as a gatekeeper for the cytosolic access of the pathogen. Host CDC42, TOCA-1, N-WASP, WIP, the Arp2/3 complex, cortactin, coronin, and cofilin are recruited to the actin cocoon. They are subverted by T3SS effectors, such as IpgD, IpgB1, and IcsB. IcsB immobilizes components of the actin polymerization machinery at the BCV dependent on its fatty acyltransferase activity. This represents a unique microbial subversion strategy through localized entrapment of host actin regulators causing massive actin assembly. We propose that the cocoon promotes subsequent invasion steps for successful Shigella infection. A thick actin cocoon forms de novo around the Shigella-containing vacuole upon entry The effector IcsB entraps host actin regulators at the vacuole by lipidation Cdc42, N-WASP, and the Arp2/3 complex are major actin cocoon regulators Cocoon formation promotes subsequent Shigella niche formation and dissemination
Collapse
Affiliation(s)
- Sonja Kühn
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - John Bergqvist
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Magdalena Gil
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Camila Valenzuela
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Laura Barrio
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Stéphanie Lebreton
- Institut Pasteur, Department of Cell Biology and Infection, Membrane Trafficking and Pathogenesis Unit, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Chiara Zurzolo
- Institut Pasteur, Department of Cell Biology and Infection, Membrane Trafficking and Pathogenesis Unit, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Jost Enninga
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
8
|
Kutsch M, González-Prieto C, Lesser CF, Coers J. The GBP1 microcapsule interferes with IcsA-dependent septin cage assembly around Shigella flexneri. Pathog Dis 2021; 79:6246431. [PMID: 33885766 DOI: 10.1093/femspd/ftab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
Many cytosolic bacterial pathogens hijack the host actin polymerization machinery to form actin tails that promote direct cell-to-cell spread, enabling these pathogens to avoid extracellular immune defenses. However, these pathogens are still susceptible to intracellular cell-autonomous immune responses that restrict bacterial actin-based motility. Two classes of cytosolic antimotility factors, septins and guanylate-binding proteins (GBPs), have recently been established to block actin tail formation by the human-adapted bacterial pathogen Shigella flexneri. Both septin cages and GBP1 microcapsules restrict S. flexneri cell-to-cell spread by blocking S. flexneri actin-based motility. While septins assemble into cage-like structures around immobile S. flexneri, GBP1 forms microcapsules around both motile and immobile bacteria. The interplay between these two defense programs remains elusive. Here, we demonstrate that GBP1 microcapsules block septin cage assembly, likely by interfering with the function of S. flexneri IcsA, the outer membrane protein that promotes actin-based motility, as this protein is required for septin cage formation. However, S. flexneri that escape from GBP1 microcapsules via the activity of IpaH9.8, a type III secreted effector that promotes the degradation of GBPs, are often captured within septin cages. Thus, our studies reveal how septin cages and GBP1 microcapsules represent complementary host cell antimotility strategies.
Collapse
Affiliation(s)
- Miriam Kutsch
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Coral González-Prieto
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02115, USA.,Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Cammie F Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02115, USA.,Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.,Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
9
|
Dhanda AS, Yang D, Guttman JA. Localization of alpha-actinin-4 during infections by actin remodeling bacteria. Anat Rec (Hoboken) 2020; 304:1400-1419. [PMID: 33099893 DOI: 10.1002/ar.24548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/13/2020] [Accepted: 09/12/2020] [Indexed: 11/12/2022]
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.
Collapse
Affiliation(s)
- Aaron S Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Diana Yang
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
10
|
Whelan R, McVicker G, Leo JC. Staying out or Going in? The Interplay between Type 3 and Type 5 Secretion Systems in Adhesion and Invasion of Enterobacterial Pathogens. Int J Mol Sci 2020; 21:E4102. [PMID: 32521829 PMCID: PMC7312957 DOI: 10.3390/ijms21114102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Enteric pathogens rely on a variety of toxins, adhesins and other virulence factors to cause infections. Some of the best studied pathogens belong to the Enterobacterales order; these include enteropathogenic and enterohemorrhagic Escherichia coli, Shigella spp., and the enteropathogenic Yersiniae. The pathogenesis of these organisms involves two different secretion systems, a type 3 secretion system (T3SS) and type 5 secretion systems (T5SSs). The T3SS forms a syringe-like structure spanning both bacterial membranes and the host cell plasma membrane that translocates toxic effector proteins into the cytoplasm of the host cell. T5SSs are also known as autotransporters, and they export part of their own polypeptide to the bacterial cell surface where it exerts its function, such as adhesion to host cell receptors. During infection with these enteropathogens, the T3SS and T5SS act in concert to bring about rearrangements of the host cell cytoskeleton, either to invade the cell, confer intracellular motility, evade phagocytosis or produce novel structures to shelter the bacteria. Thus, in these bacteria, not only the T3SS effectors but also T5SS proteins could be considered "cytoskeletoxins" that bring about profound alterations in host cell cytoskeletal dynamics and lead to pathogenic outcomes.
Collapse
Affiliation(s)
| | | | - Jack C. Leo
- Antimicrobial Resistance, Omics and Microbiota Group, Department of Biosciences, Nottingham Trent University, Nottingham NG1 4FQ, UK; (R.W.); (G.M.)
| |
Collapse
|
11
|
Kühn S, Enninga J. The actin comet guides the way: How
Listeria
actin subversion has impacted cell biology, infection biology and structural biology. Cell Microbiol 2020; 22:e13190. [DOI: 10.1111/cmi.13190] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Sonja Kühn
- Unit of Dynamics of Host‐Pathogen InteractionsInstitut Pasteur Paris France
- Centre National de la Recherche Scientifique (CNRS‐UMR3691) Paris France
| | - Jost Enninga
- Unit of Dynamics of Host‐Pathogen InteractionsInstitut Pasteur Paris France
- Centre National de la Recherche Scientifique (CNRS‐UMR3691) Paris France
| |
Collapse
|
12
|
The virulence domain of Shigella IcsA contains a subregion with specific host cell adhesion function. PLoS One 2020; 15:e0227425. [PMID: 31910229 PMCID: PMC6946128 DOI: 10.1371/journal.pone.0227425] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/18/2019] [Indexed: 02/08/2023] Open
Abstract
Shigella species cause bacillary dysentery, especially among young individuals. Shigellae target the human colon for invasion; however, the initial adhesion mechanism is poorly understood. The Shigella surface protein IcsA, in addition to its role in actin-based motility, acts as a host cell adhesin through unknown mechanism(s). Here we confirmed the role of IcsA in cell adhesion and defined the region required for IcsA adhesin activity. Purified IcsA passenger domain was able block S. flexneri adherence and was also used as a molecular probe that recognised multiple components from host cells. The region within IcsA's functional passenger domain (aa 138-148) was identified by mutagenesis. Upon the deletion of this region, the purified IcsAΔ138-148 was found to no longer block S. flexneri adherence and had reduced ability to interact with host molecules. Furthermore, S. flexneri expressing IcsAΔ138-148 was found to be significantly defective in both cell adherence and invasion. Taken together, our data identify an adherence region within the IcsA functional domain and provides useful information for designing therapeutics for Shigella infection.
Collapse
|
13
|
Wang QC, Wan X, Jia RX, Xu Y, Liu X, Zhang Y, Sun SC. Inhibition of N-WASP affects actin-mediated cytokinesis during porcine oocyte maturation. Theriogenology 2020; 144:132-138. [PMID: 31940504 DOI: 10.1016/j.theriogenology.2020.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/02/2019] [Accepted: 01/05/2020] [Indexed: 02/07/2023]
Abstract
N-WASP is the mammalian ortholog of WASP which is an actin nucleation promoting factor and has been reported to regulate actin nucleation and polymerization for multiple cell activities. However, the expression and functions of N-WASP in porcine oocytes are still unclear. In this study, we showed that N-WASP expressed at all stages during porcine oocyte maturation, and immunofluorescence staining indicated that N-WASP mainly accumulated at the cortex in different stages of meiosis. Inhibition of N-WASP activity by Wiskostatin significantly decreased the rate of first polar body extrusion and disturbed the cell cycle progression of porcine oocytes. Further analysis indicated that cortical actin distribution was interfered by N-WASP inhibition, and this might be through its regulatory roles on the expression and localization of ARP2, a key component of actin nucleator Arp2/3 complex. Moreover, the expression of N-WASP decreased after ROCK activity inhibition, indicating a ROCK-N-WASP-ARP2/3 pathway for actin assembly in porcine oocytes. Taken together, these results suggest that N-WASP is critical for the regulation of actin filaments for cytokinesis during porcine oocyte maturation.
Collapse
Affiliation(s)
- Qiao-Chu Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiang Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ru-Xia Jia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yao Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiuhong Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
14
|
Velle KB, Fritz-Laylin LK. Diversity and evolution of actin-dependent phenotypes. Curr Opin Genet Dev 2019; 58-59:40-48. [DOI: 10.1016/j.gde.2019.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/19/2019] [Accepted: 07/20/2019] [Indexed: 12/20/2022]
|
15
|
Evolutionary Perspectives on the Moonlighting Functions of Bacterial Factors That Support Actin-Based Motility. mBio 2019; 10:mBio.01520-19. [PMID: 31455648 PMCID: PMC6712393 DOI: 10.1128/mbio.01520-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Various bacterial pathogens display an intracellular lifestyle and spread from cell to cell through actin-based motility (ABM). ABM requires actin polymerization at the bacterial pole and is mediated by the expression of bacterial factors that hijack the host cell actin nucleation machinery or exhibit intrinsic actin nucleation properties. Various bacterial pathogens display an intracellular lifestyle and spread from cell to cell through actin-based motility (ABM). ABM requires actin polymerization at the bacterial pole and is mediated by the expression of bacterial factors that hijack the host cell actin nucleation machinery or exhibit intrinsic actin nucleation properties. It is increasingly recognized that bacterial ABM factors, in addition to having a crucial task during the intracellular phase of infection, display “moonlighting” adhesin functions, such as bacterial aggregation, biofilm formation, and host cell adhesion/invasion. Here, we review our current knowledge of ABM factors and their additional functions, and we propose that intracellular ABM functions have evolved from ancestral, extracellular adhesin functions.
Collapse
|
16
|
Michard C, Yum LK, Agaisse H. WIPF2 promotesShigella flexneriactin‐based motility and cell‐to‐cell spread. Cell Microbiol 2019; 21:e13098. [DOI: 10.1111/cmi.13098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Céline Michard
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of Virginia Charlottesville Virginia
| | - Lauren K. Yum
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of Virginia Charlottesville Virginia
| | - Hervé Agaisse
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of Virginia Charlottesville Virginia
| |
Collapse
|
17
|
Krokowski S, Atwal S, Lobato-Márquez D, Chastanet A, Carballido-López R, Salje J, Mostowy S. Shigella MreB promotes polar IcsA positioning for actin tail formation. J Cell Sci 2019; 132:jcs.226217. [PMID: 30992346 PMCID: PMC6526709 DOI: 10.1242/jcs.226217] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/01/2019] [Indexed: 01/17/2023] Open
Abstract
Pathogenic Shigella bacteria are a paradigm to address key issues of cell and infection biology. Polar localisation of the Shigella autotransporter protein IcsA is essential for actin tail formation, which is necessary for the bacterium to travel from cell-to-cell; yet how proteins are targeted to the bacterial cell pole is poorly understood. The bacterial actin homologue MreB has been extensively studied in broth culture using model organisms including Escherichia coli, Bacillus subtilis and Caulobacter crescentus, but has never been visualised in rod-shaped pathogenic bacteria during infection of host cells. Here, using single-cell analysis of intracellular Shigella, we discover that MreB accumulates at the cell pole of bacteria forming actin tails, where it colocalises with IcsA. Pharmacological inhibition of host cell actin polymerisation and genetic deletion of IcsA is used to show, respectively, that localisation of MreB to the cell poles precedes actin tail formation and polar localisation of IcsA. Finally, by exploiting the MreB inhibitors A22 and MP265, we demonstrate that MreB polymerisation can support actin tail formation. We conclude that Shigella MreB promotes polar IcsA positioning for actin tail formation, and suggest that understanding the bacterial cytoskeleton during host–pathogen interactions can inspire development of new therapeutic regimes for infection control. This article has an associated First Person interview with the first author of the paper. Summary: The pathogen Shigella forms actin tails to move through the cytosol of infected cells. We show that the bacterial actin homologue MreB can help to position the autotransporter protein IcsA for such actin tail formation.
Collapse
Affiliation(s)
- Sina Krokowski
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK.,Department of Immunology & Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Sharanjeet Atwal
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7JT, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400 PHRI 07103, Thailand.,Public Health Research Institute, Rutgers Biomedical and Health Science, Newark, New Jersey NJ 07103, USA
| | - Damián Lobato-Márquez
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK.,Department of Immunology & Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Arnaud Chastanet
- MICALIS Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Rut Carballido-López
- MICALIS Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Jeanne Salje
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7JT, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400 PHRI 07103, Thailand.,Public Health Research Institute, Rutgers Biomedical and Health Science, Newark, New Jersey NJ 07103, USA
| | - Serge Mostowy
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, London SW7 2AZ, UK .,Department of Immunology & Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|
18
|
Velle KB, Campellone KG. Enteropathogenic E. coli relies on collaboration between the formin mDia1 and the Arp2/3 complex for actin pedestal biogenesis and maintenance. PLoS Pathog 2018; 14:e1007485. [PMID: 30550556 PMCID: PMC6310289 DOI: 10.1371/journal.ppat.1007485] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/28/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
Enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) are closely related extracellular pathogens that reorganize host cell actin into “pedestals” beneath the tightly adherent bacteria. This pedestal-forming activity is both a critical step in pathogenesis, and it makes EPEC and EHEC useful models for studying the actin rearrangements that underlie membrane protrusions. To generate pedestals, EPEC relies on the tyrosine phosphorylated bacterial effector protein Tir to bind host adaptor proteins that recruit N-WASP, a nucleation-promoting factor that activates the Arp2/3 complex to drive actin polymerization. In contrast, EHEC depends on the effector EspFU to multimerize N-WASP and promote Arp2/3 activation. Although these core pathways of pedestal assembly are well-characterized, the contributions of additional actin nucleation factors are unknown. We investigated potential cooperation between the Arp2/3 complex and other classes of nucleators using chemical inhibitors, siRNAs, and knockout cell lines. We found that inhibition of formins impairs actin pedestal assembly, motility, and cellular colonization for bacteria using the EPEC, but not the EHEC, pathway of actin polymerization. We also identified mDia1 as the formin contributing to EPEC pedestal assembly, as its expression level positively correlates with the efficiency of pedestal formation, and it localizes to the base of pedestals both during their initiation and once they have reached steady state. Collectively, our data suggest that mDia1 enhances EPEC pedestal biogenesis and maintenance by generating seed filaments to be used by the N-WASP-Arp2/3-dependent actin nucleation machinery and by sustaining Src-mediated phosphorylation of Tir. Microbial pathogens that rearrange the host actin cytoskeleton have made valuable contributions to our understanding of cell signaling and movement. The assembly and organization of the actin cytoskeleton is driven by proteins called nucleators, which can be manipulated by bacteria including enteropathogenic Escherichia coli (EPEC), a frequent cause of pediatric diarrhea in developing countries. After ingestion, EPEC adhere tightly to cells of the intestine and hijack the underlying cytoskeleton to create protrusions called actin pedestals. While mechanisms of pedestal assembly involving a nucleator called the Arp2/3 complex have been defined for EPEC, the contribution of additional host nucleators has not been determined. We assessed the roles of several actin nucleators in EPEC pedestals and found that in addition to Arp2/3 complex-mediated nucleation, the formin mDia1 is a key contributor to actin assembly. These findings highlight the importance of nucleator collaboration in pathogenesis, and also advance our understanding of the molecular and cellular basis of EPEC infection, which is ultimately important for the discovery of new drug targets.
Collapse
Affiliation(s)
- Katrina B. Velle
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Kenneth G. Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
19
|
Higgs HN. A fruitful tree: developing the dendritic nucleation model of actin-based cell motility. Mol Biol Cell 2018. [PMCID: PMC6333179 DOI: 10.1091/mbc.e18-07-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
A fundamental question in cell biology concerns how cells move, and this has been the subject of intense research for decades. In the 1990s, a major leap forward was made in our understanding of cell motility, with the proposal of the dendritic nucleation model. This essay describes the events leading to the development of the model, including findings from many laboratories and scientific disciplines. The story is an excellent example of the scientific process in action, with the combination of multiple perspectives leading to robust conclusions.
Collapse
Affiliation(s)
- Henry N. Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
20
|
Dorman MJ, Dorman CJ. Regulatory Hierarchies Controlling Virulence Gene Expression in Shigella flexneri and Vibrio cholerae. Front Microbiol 2018; 9:2686. [PMID: 30473684 PMCID: PMC6237886 DOI: 10.3389/fmicb.2018.02686] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Gram-negative enteropathogenic bacteria use a variety of strategies to cause disease in the human host and gene regulation in some form is typically a part of the strategy. This article will compare the toxin-based infection strategy used by the non-invasive pathogen Vibrio cholerae, the etiological agent in human cholera, with the invasive approach used by Shigella flexneri, the cause of bacillary dysentery. Despite the differences in the mechanisms by which the two pathogens cause disease, they use environmentally-responsive regulatory hierarchies to control the expression of genes that have some features, and even some components, in common. The involvement of AraC-like transcription factors, the integration host factor, the Factor for inversion stimulation, small regulatory RNAs, the RNA chaperone Hfq, horizontal gene transfer, variable DNA topology and the need to overcome the pervasive silencing of transcription by H-NS of horizontally acquired genes are all shared features. A comparison of the regulatory hierarchies in these two pathogens illustrates some striking cross-species similarities and differences among mechanisms coordinating virulence gene expression. S. flexneri, with its low infectious dose, appears to use a strategy that is centered on the individual bacterial cell, whereas V. cholerae, with a community-based, quorum-dependent approach and an infectious dose that is several orders of magnitude higher, seems to rely more on the actions of a bacterial collective.
Collapse
Affiliation(s)
- Matthew J Dorman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Charles J Dorman
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Siqueira MDS, Ribeiro RDM, Travassos LH. Autophagy and Its Interaction With Intracellular Bacterial Pathogens. Front Immunol 2018; 9:935. [PMID: 29875765 PMCID: PMC5974045 DOI: 10.3389/fimmu.2018.00935] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
Abstract
Cellular responses to stress can be defined by the overwhelming number of changes that cells go through upon contact with and stressful conditions such as infection and modifications in nutritional status. One of the main cellular responses to stress is autophagy. Much progress has been made in the understanding of the mechanisms involved in the induction of autophagy during infection by intracellular bacteria. This review aims to discuss recent findings on the role of autophagy as a cellular response to intracellular bacterial pathogens such as, Streptococcus pyogenes, Mycobacterium tuberculosis, Shigella flexneri, Salmonella typhimurium, Listeria monocytogenes, and Legionella pneumophila, how the autophagic machinery senses these bacteria directly or indirectly (through the detection of bacteria-induced nutritional stress), and how some of these bacterial pathogens manage to escape from autophagy.
Collapse
Affiliation(s)
- Mariana da Silva Siqueira
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato de Moraes Ribeiro
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo H Travassos
- Laboratory of Immunoreceptors and Signaling, Immunobiology Program, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Aktories K, Schwan C, Lang AE. ADP-Ribosylation and Cross-Linking of Actin by Bacterial Protein Toxins. Handb Exp Pharmacol 2017; 235:179-206. [PMID: 27316913 DOI: 10.1007/164_2016_26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Actin and the actin cytoskeleton play fundamental roles in host-pathogen interactions. Proper function of the actin cytoskeleton is crucial for innate and acquired immune defense. Bacterial toxins attack the actin cytoskeleton by targeting regulators of actin. Moreover, actin is directly modified by various bacterial protein toxins and effectors, which cause ADP-ribosylation or cross-linking of actin. Modification of actin can result in inhibition or stimulation of actin polymerization. Toxins, acting directly on actin, are reviewed.
Collapse
Affiliation(s)
- Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany. .,Freiburg Institute of Advanced Studies (FRIAS), Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany.
| | - Carsten Schwan
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany
| | - Alexander E Lang
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany
| |
Collapse
|
23
|
Leupold S, Büsing P, Mas PJ, Hart DJ, Scrima A. Structural insights into the architecture of the Shigella flexneri virulence factor IcsA/VirG and motifs involved in polar distribution and secretion. J Struct Biol 2017; 198:19-27. [PMID: 28268178 DOI: 10.1016/j.jsb.2017.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/23/2017] [Accepted: 03/03/2017] [Indexed: 12/12/2022]
Abstract
IcsA/VirG is a key virulence factor of the human pathogen Shigella flexneri, acting as both an adhesin and actin-polymerizing factor during infection. We identified a soluble expression construct of the IcsA/VirG α-domain using the ESPRIT library screening system and determined its structure to 1.9Å resolution. In addition to the previously characterized autochaperone domain, our structure reveals a new domain, which shares a common fold with the autochaperone domains of various autotransporters. We further provide insight into the previously structurally uncharacterized β-helix domain that harbors the polar targeting motif and passenger-associated transport repeat. This structure is the first of any member of the recently identified passenger-associated transport repeat-containing autotransporters. Thus, it provides new insights into the overall architecture of this class of autotransporters, the function of the identified additional autochaperone domain and the structural properties of motifs involved in polar targeting and secretion of the Shigella flexneri virulence factor IcsA/VirG.
Collapse
Affiliation(s)
- Stefan Leupold
- Structural Biology of Autophagy, Helmholtz-Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Petra Büsing
- Structural Biology of Autophagy, Helmholtz-Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Philippe J Mas
- European Molecular Biology Laboratory Grenoble Outstation and Unit of Virus Host-Cell Interactions, University Grenoble Alpes-CNRS-EMBL, 71 Avenue des Martyrs, CS 90181, 38042 Grenoble Cedex 9, France
| | - Darren J Hart
- European Molecular Biology Laboratory Grenoble Outstation and Unit of Virus Host-Cell Interactions, University Grenoble Alpes-CNRS-EMBL, 71 Avenue des Martyrs, CS 90181, 38042 Grenoble Cedex 9, France
| | - Andrea Scrima
- Structural Biology of Autophagy, Helmholtz-Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany.
| |
Collapse
|
24
|
Pizarro-Cerdá J, Chorev DS, Geiger B, Cossart P. The Diverse Family of Arp2/3 Complexes. Trends Cell Biol 2016; 27:93-100. [PMID: 27595492 DOI: 10.1016/j.tcb.2016.08.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 11/17/2022]
Abstract
The Arp2/3 complex has so far been considered to be a single seven-subunit protein complex required for actin nucleation and actin filament polymerization in diverse critical cellular functions including phagocytosis, vesicular trafficking and lamellipodia extension. The Arp2/3 complex is also exploited by bacterial pathogens and viruses during cellular infectious processes. Recent studies suggest that some subunits of the complex are dispensable in specific cellular contexts, pointing to the existence of alternative 'hybrid Arp2/3 complexes' containing other components such as vinculin or α-actinin, as well as different isoforms or phosphorylation variants of canonical Arp2/3 subunits. Therefore, this diversity should be now considered when assigning specific Arp2/3 assemblies to different actin-dependent cellular processes.
Collapse
Affiliation(s)
- Javier Pizarro-Cerdá
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France; INSERM, U604, Paris F-75015, France; INRA, USC2020, Paris F-75015, France.
| | - Dror Shlomo Chorev
- The Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot 761001, Israel; Department of Chemistry, University of Oxford, Physical and Theoretical Chemistry Laboratory, Oxford OX1 3QZ, UK
| | - Benjamin Geiger
- The Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot 761001, Israel
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris F-75015, France; INSERM, U604, Paris F-75015, France; INRA, USC2020, Paris F-75015, France.
| |
Collapse
|
25
|
Krokowski S, Mostowy S. Investigation of septins using infection by bacterial pathogens. Methods Cell Biol 2016; 136:117-34. [PMID: 27473906 DOI: 10.1016/bs.mcb.2016.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Investigation of the host cytoskeleton during infection by bacterial pathogens has significantly contributed to our understanding of cell biology and host defense. Work has shown that septins are recruited to the phagocytic cup as collarlike structures and enable bacterial entry into host cells. In the cytosol, septins can entrap actin-polymerizing bacteria in cage-like structures for targeting to autophagy, a highly conserved intracellular degradation process. In this chapter, we describe methods to investigate septin assembly and function during infection by bacterial pathogens. Use of these methods can lead to in-depth understanding of septin biology and suggest therapeutic approaches to combat infectious disease.
Collapse
Affiliation(s)
- S Krokowski
- Imperial College London, London, United Kingdom
| | - S Mostowy
- Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Agaisse H. Molecular and Cellular Mechanisms of Shigella flexneri Dissemination. Front Cell Infect Microbiol 2016; 6:29. [PMID: 27014639 PMCID: PMC4786538 DOI: 10.3389/fcimb.2016.00029] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/26/2016] [Indexed: 11/13/2022] Open
Abstract
The intracellular pathogen Shigella flexneri is the causative agent of bacillary dysentery in humans. The disease is characterized by bacterial invasion of intestinal cells, dissemination within the colonic epithelium through direct spread from cell to cell, and massive inflammation of the intestinal mucosa. Here, we review the mechanisms supporting S. flexneri dissemination. The dissemination process primarily relies on actin assembly at the bacterial pole, which propels the pathogen throughout the cytosol of primary infected cells. Polar actin assembly is supported by polar expression of the bacterial autotransporter family member IcsA, which recruits the N-WASP/ARP2/3 actin assembly machinery. As motile bacteria encounter cell-cell contacts, they form plasma membrane protrusions that project into adjacent cells. In addition to the ARP2/3-dependent actin assembly machinery, protrusion formation relies on formins and myosins. The resolution of protrusions into vacuoles occurs through the collapse of the protrusion neck, leading to the formation of an intermediate membrane-bound compartment termed vacuole-like protrusions (VLPs). VLP formation requires tyrosine kinase and phosphoinositide signaling in protrusions, which relies on the integrity of the bacterial type 3 secretion system (T3SS). The T3SS is also required for escaping double membrane vacuoles through the activity of the T3SS translocases IpaB and IpaC, and the effector proteins VirA and IcsB. Numerous factors supporting envelope biogenesis contribute to IcsA exposure and maintenance at the bacterial pole, including LPS synthesis, membrane proteases, and periplasmic chaperones. Although less characterized, the assembly and function of the T3SS in the context of bacterial dissemination also relies on factors supporting envelope biogenesis. Finally, the dissemination process requires the adaptation of the pathogen to various cellular compartments through transcriptional and post-transcriptional mechanisms.
Collapse
Affiliation(s)
- Hervé Agaisse
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine Charlottesville, VA, USA
| |
Collapse
|
27
|
Ashida H, Sasakawa C. Shigella IpaH Family Effectors as a Versatile Model for Studying Pathogenic Bacteria. Front Cell Infect Microbiol 2016; 5:100. [PMID: 26779450 PMCID: PMC4701945 DOI: 10.3389/fcimb.2015.00100] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/14/2015] [Indexed: 02/03/2023] Open
Abstract
Shigella spp. are highly adapted human pathogens that cause bacillary dysentery (shigellosis). Via the type III secretion system (T3SS), Shigella deliver a subset of virulence proteins (effectors) that are responsible for pathogenesis, with functions including pyroptosis, invasion of the epithelial cells, intracellular survival, and evasion of host immune responses. Intriguingly, T3SS effector activity and strategies are not unique to Shigella, but are shared by many other bacterial pathogens, including Salmonella, Yersinia, and enteropathogenic Escherichia coli (EPEC). Therefore, studying Shigella T3SS effectors will not only improve our understanding of bacterial infection systems, but also provide a molecular basis for developing live bacterial vaccines and antibacterial drugs. One of Shigella T3SS effectors, IpaH family proteins, which have E3 ubiquitin ligase activity and are widely conserved among other bacterial pathogens, are very relevant because they promote bacterial survival by triggering cell death and modulating the host immune responses. Here, we describe selected examples of Shigella pathogenesis, with particular emphasis on the roles of IpaH family effectors, which shed new light on bacterial survival strategies and provide clues about how to overcome bacterial infections.
Collapse
Affiliation(s)
- Hiroshi Ashida
- Division of Bacterial Infection Biology, Institute of Medical Science, University of Tokyo Tokyo, Japan
| | - Chihiro Sasakawa
- Division of Bacterial Infection Biology, Institute of Medical Science, University of TokyoTokyo, Japan; Nippon Institute for Biological ScienceTokyo, Japan; Medical Mycology Research Center, Chiba UniversityChiba, Japan
| |
Collapse
|
28
|
Van Nhieu GT, Romero S. Common Themes in Cytoskeletal Remodeling by Intracellular Bacterial Effectors. Handb Exp Pharmacol 2016; 235:207-235. [PMID: 27807696 DOI: 10.1007/164_2016_42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bacterial pathogens interact with various types of tissues to promote infection. Because it controls the formation of membrane extensions, adhesive processes, or the junction integrity, the actin cytoskeleton is a key target of pathogens during infection. We will highlight common and specific functions of the actin cytoskeleton during bacterial infections, by first reviewing the mechanisms of intracellular motility of invasive Shigella, Listeria, and Rickettsia. Through the models of EPEC/EHEC, Shigella, Salmonella, and Chlamydia spp., we will illustrate various strategies of diversion of actin cytoskeletal processes used by these bacteria to colonize or breach epithelial/endothelial barriers.
Collapse
Affiliation(s)
- Guy Tran Van Nhieu
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, 75005, Paris, France. .,Institut National de la Santé et de la Recherche Médicale U1050, 75005, Paris, France. .,Centre National de la Recherche Scientifique UMR7241, 75005, Paris, France. .,MEMOLIFE Laboratory of Excellence and Paris Science Lettre, 75005, Paris, France.
| | - Stéphane Romero
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, 75005, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1050, 75005, Paris, France.,Centre National de la Recherche Scientifique UMR7241, 75005, Paris, France.,MEMOLIFE Laboratory of Excellence and Paris Science Lettre, 75005, Paris, France
| |
Collapse
|
29
|
Fan J, Zhai Z, Yan C, Xu C. Arabidopsis TRIGALACTOSYLDIACYLGLYCEROL5 Interacts with TGD1, TGD2, and TGD4 to Facilitate Lipid Transfer from the Endoplasmic Reticulum to Plastids. THE PLANT CELL 2015; 27:2941-55. [PMID: 26410300 PMCID: PMC4682317 DOI: 10.1105/tpc.15.00394] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/24/2015] [Accepted: 09/04/2015] [Indexed: 05/20/2023]
Abstract
The biogenesis of photosynthetic membranes in the plastids of higher plants requires an extensive supply of lipid precursors from the endoplasmic reticulum (ER). Four TRIGALACTOSYLDIACYLGLYCEROL (TGD) proteins (TGD1,2,3,4) have thus far been implicated in this lipid transfer process. While TGD1, TGD2, and TGD3 constitute an ATP binding cassette transporter complex residing in the plastid inner envelope, TGD4 is a transmembrane lipid transfer protein present in the outer envelope. These observations raise questions regarding how lipids transit across the aqueous intermembrane space. Here, we describe the isolation and characterization of a novel Arabidopsis thaliana gene, TGD5. Disruption of TGD5 results in similar phenotypic effects as previously described in tgd1,2,3,4 mutants, including deficiency of ER-derived thylakoid lipids, accumulation of oligogalactolipids, and triacylglycerol. Genetic analysis indicates that TGD4 is epistatic to TGD5 in ER-to-plastid lipid trafficking, whereas double mutants of a null tgd5 allele with tgd1-1 or tgd2-1 show a synergistic embryo-lethal phenotype. TGD5 encodes a small glycine-rich protein that is localized in the envelope membranes of chloroplasts. Coimmunoprecipitation assays show that TGD5 physically interacts with TGD1, TGD2, TGD3, and TGD4. Collectively, these results suggest that TGD5 facilitates lipid transfer from the outer to the inner plastid envelope by bridging TGD4 with the TGD1,2,3 transporter complex.
Collapse
Affiliation(s)
- Jilian Fan
- Biological, Environmental, and Climate Sciences Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Zhiyang Zhai
- Biological, Environmental, and Climate Sciences Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Chengshi Yan
- Biological, Environmental, and Climate Sciences Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Changcheng Xu
- Biological, Environmental, and Climate Sciences Department, Brookhaven National Laboratory, Upton, New York 11973
| |
Collapse
|
30
|
Abstract
Benanti et al. report that Burkholderia pseudomallei and Burkholderia mallei bacteria express proteins that mimic Ena/Vasp family proteins to polymerize actin, thereby inducing actin-based motility. Thus, bacteria can use the various cellular actin polymerization mechanisms for intra- and inter-cellular dissemination.
Collapse
Affiliation(s)
- Edith Gouin
- Unité des Interactions bactéries-Cellules, Institut Pasteur, 75015 Paris, France; Inserm U604, 75015 Paris, France; INRA USC2020, 75015 Paris, France
| | | | - Juan-Jose Quereda Torres
- Unité des Interactions bactéries-Cellules, Institut Pasteur, 75015 Paris, France; Inserm U604, 75015 Paris, France; INRA USC2020, 75015 Paris, France
| | - Pascale Cossart
- Unité des Interactions bactéries-Cellules, Institut Pasteur, 75015 Paris, France; Inserm U604, 75015 Paris, France; INRA USC2020, 75015 Paris, France.
| |
Collapse
|
31
|
Kuehl CJ, Dragoi AM, Talman A, Agaisse H. Bacterial spread from cell to cell: beyond actin-based motility. Trends Microbiol 2015; 23:558-66. [PMID: 26021574 DOI: 10.1016/j.tim.2015.04.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/14/2015] [Accepted: 04/28/2015] [Indexed: 01/01/2023]
Abstract
Several intracellular pathogens display the ability to propagate within host tissues by displaying actin-based motility in the cytosol of infected cells. As motile bacteria reach cell-cell contacts they form plasma membrane protrusions that project into adjacent cells and resolve into vacuoles from which the pathogen escapes, thereby achieving spread from cell to cell. Seminal studies have defined the bacterial and cellular factors that support actin-based motility. By contrast, the mechanisms supporting the formation of protrusions and their resolution into vacuoles have remained elusive. Here, we review recent advances in the field showing that Listeria monocytogenes and Shigella flexneri have evolved pathogen-specific mechanisms of bacterial spread from cell to cell.
Collapse
Affiliation(s)
- Carole J Kuehl
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, CT, USA
| | - Ana-Maria Dragoi
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, CT, USA
| | - Arthur Talman
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, CT, USA
| | - Hervé Agaisse
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, CT, USA.
| |
Collapse
|
32
|
Ashida H, Mimuro H, Sasakawa C. Shigella manipulates host immune responses by delivering effector proteins with specific roles. Front Immunol 2015; 6:219. [PMID: 25999954 PMCID: PMC4423471 DOI: 10.3389/fimmu.2015.00219] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 04/22/2015] [Indexed: 11/30/2022] Open
Abstract
The intestinal epithelium deploys multiple defense systems against microbial infection to sense bacterial components and danger alarms, as well as to induce intracellular signal transduction cascades that trigger both the innate and the adaptive immune systems, which are pivotal for bacterial elimination. However, many enteric bacterial pathogens, including Shigella, deliver a subset of virulence proteins (effectors) via the type III secretion system (T3SS) that enable bacterial evasion from host immune systems; consequently, these pathogens are able to efficiently colonize the intestinal epithelium. In this review, we present and select recently discovered examples of interactions between Shigella and host immune responses, with particular emphasis on strategies that bacteria use to manipulate inflammatory outputs of host-cell responses such as cell death, membrane trafficking, and innate and adaptive immune responses.
Collapse
Affiliation(s)
- Hiroshi Ashida
- Division of Bacterial Infection Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Hitomi Mimuro
- Division of Bacteriology, Department of Infectious Diseases Control, International Research Center for Infectious Diseases, The Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Chihiro Sasakawa
- Division of Bacterial Infection Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan ; Nippon Institute for Biological Science , Tokyo , Japan ; Medical Mycology Research Center, Chiba University , Chiba , Japan
| |
Collapse
|
33
|
Ud-Din A, Wahid S. Relationship among Shigella spp. and enteroinvasive Escherichia coli (EIEC) and their differentiation. Braz J Microbiol 2015; 45:1131-8. [PMID: 25763015 PMCID: PMC4323284 DOI: 10.1590/s1517-83822014000400002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 12/13/2013] [Indexed: 11/30/2022] Open
Abstract
Shigellosis produces inflammatory reactions and ulceration on the intestinal epithelium followed by bloody or mucoid diarrhea. It is caused by enteroinvasive E. coli (EIEC) as well as any species of the genus Shigella, namely, S. dysenteriae, S. flexneri, S. boydii, and S. sonnei. This current species designation of Shigella does not specify genetic similarity. Shigella spp. could be easily differentiated from E. coli, but difficulties observed for the EIEC-Shigella differentiation as both show similar biochemical traits and can cause dysentery using the same mode of invasion. Sequencing of multiple housekeeping genes indicates that Shigella has derived on several different occasions via acquisition of the transferable forms of ancestral virulence plasmids within commensal E. coli and form a Shigella-EIEC pathovar. EIEC showed lower expression of virulence genes compared to Shigella, hence EIEC produce less severe disease than Shigella spp. Conventional microbiological techniques often lead to confusing results concerning the discrimination between EIEC and Shigella spp. The lactose permease gene (lacY) is present in all E. coli strains but absent in Shigella spp., whereas β-glucuronidase gene (uidA) is present in both E. coli and Shigella spp. Thus uidA gene and lacY gene based duplex real-time PCR assay could be used for easy identification and differentiation of Shigella spp. from E. coli and in particular EIEC.
Collapse
Affiliation(s)
- Abu Ud-Din
- Identification and Subtyping of Enteric Pathogens International Centre for Diarrhoeal Disease Research in Bangladesh Dhaka Bangladesh Identification and Subtyping of Enteric Pathogens, International Centre for Diarrhoeal Disease Research in Bangladesh, Dhaka, Bangladesh
| | - Syeda Wahid
- Center for Food and Water Borne Disease International Centre for Diarrhoeal Disease Research in Bangladesh Dhaka Bangladesh Center for Food and Water Borne Disease, International Centre for Diarrhoeal Disease Research in Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
34
|
Brotcke Zumsteg A, Goosmann C, Brinkmann V, Morona R, Zychlinsky A. IcsA is a Shigella flexneri adhesin regulated by the type III secretion system and required for pathogenesis. Cell Host Microbe 2015; 15:435-45. [PMID: 24721572 DOI: 10.1016/j.chom.2014.03.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/20/2014] [Accepted: 02/24/2014] [Indexed: 01/05/2023]
Abstract
Following contact with the epithelium, the enteric intracellular bacterial pathogen Shigella flexneri invades epithelial cells and escapes intracellular phagosomal destruction using its type III secretion system (T3SS). The bacterium replicates within the host cell cytosol and spreads between cells using actin-based motility, which is mediated by the virulence factor IcsA (VirG). Whereas S. flexneri invasion is well characterized, adhesion mechanisms of the bacterium remain elusive. We found that IcsA also functions as an adhesin that is both necessary and sufficient to promote contact with host cells. As adhesion can be beneficial or deleterious depending on the host cell type, S. flexneri regulates IcsA-dependent adhesion. Activation of the T3SS in response to the bile salt deoxycholate triggers IcsA-dependent adhesion and enhances pathogen invasion. IcsA-dependent adhesion contributes to virulence in a mouse model of shigellosis, underscoring the importance of this adhesin to S. flexneri pathogenesis.
Collapse
Affiliation(s)
- Anna Brotcke Zumsteg
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin 13353, Germany
| | - Christian Goosmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Berlin 13353, Germany
| | - Volker Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Berlin 13353, Germany
| | - Renato Morona
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide 5000, South Australia, Australia
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin 13353, Germany.
| |
Collapse
|
35
|
Abstract
The invasion process of S. flexneri is well characterized, but mechanisms underlying this bacterium's adhesion to host cells have remained obscure. In this issue of Cell Host & Microbe, Brotcke Zumsteg et al. (2014) report a surprising role for the Shigella virulence factor IcsA (VirG) as an adhesin.
Collapse
|
36
|
Analysis on the interaction domain of VirG and apyrase by pull-down assay. Molecules 2014; 19:18090-101. [PMID: 25379645 PMCID: PMC6271496 DOI: 10.3390/molecules191118090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/19/2014] [Accepted: 10/21/2014] [Indexed: 01/19/2023] Open
Abstract
VirG is outer membrane protein of Shigella and affects the spread of Shigella. Recently it has been reported that apyrase influences the location of VirG, although the underlying mechanism remains poorly understood. The site of interaction between apyrase and VirG is the focus of our research. First we constructed recombinant plasmid pHIS-phoN2 and pS-(v1-1102, v53-758, v759-1102, v53-319, v320-507, v507-758) by denaturation-renaturation, the phoN2:kan mutant of Shigella flexneri 5a M90T by a modified version of the lambda red recombination protocol originally described by Datsenko and Wanner and the complemented strain M90TΔphoN2/pET24a(PhisphoN2). Second, the recombinant plasmid pHIS-phoN2 and the pS-(v1-1102, v53-758, v759-1102, v53-319, v320-507, v507-758) were transformed into E. coli BL21 (DE3) and induced to express the fusion proteins. Third, the fusion proteins were purified and the interaction of VirG and apyrase was identified by pull-down. Fourth, VirG was divided and the interaction site of apyrase and VirG was determined. Finally, how apyrase affects the function of VirG was analyzed by immunofluorescence. Accordingly, the results provided the data supporting the fact that apyrase combines with the α-domain of VirG to influence the function of VirG.
Collapse
|
37
|
Khaitlina SY. Intracellular transport based on actin polymerization. BIOCHEMISTRY (MOSCOW) 2014; 79:917-27. [DOI: 10.1134/s0006297914090089] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
38
|
The serine/threonine kinase STK11 promotes Shigella flexneri dissemination through establishment of cell-cell contacts competent for tyrosine kinase signaling. Infect Immun 2014; 82:4447-57. [PMID: 25114112 DOI: 10.1128/iai.02078-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Shigella flexneri is an intracellular pathogen that disseminates in the intestinal epithelium by displaying actin-based motility. We found that although S. flexneri displayed comparable actin-based motilities in the cytosols of HeLa229 and HT-29 epithelial cell lines, the overall dissemination process was much more efficient in HT-29 cells. Time-lapse microscopy demonstrated that as motile bacteria reached the cell cortex in HT-29 cells, they formed membrane protrusions that resolved into vacuoles, from which the bacteria escaped and gained access to the cytosol of adjacent cells. In HeLa229 cells, S. flexneri also formed membrane protrusions that extended into adjacent cells, but the protrusions rarely resolved into vacuoles. Instead, the formed protrusions collapsed and retracted, bringing the bacteria back to the cytosol of the primary infected cells. Silencing the serine/threonine kinase STK11 (also known as LKB1) in HT-29 cells decreased the efficiency of protrusion resolution into vacuoles. Conversely, expressing STK11 in HeLa229 cells, which lack the STK11 locus, dramatically increased the efficiency of protrusion resolution into vacuoles. S. flexneri dissemination in HT-29 cells led to the local phosphorylation of tyrosine residues in protrusions, a signaling event that was not observed in HeLa229 cells but was restored in STK11-expressing HeLa229 cells. Treatment of HT-29 cells with the tyrosine kinase inhibitor imatinib abrogated tyrosine kinase signaling in protrusions, which correlated with a severe decrease in the efficiency of protrusion resolution into vacuoles. We suggest that the formation of STK11-dependent lateral cell-cell contacts competent for tyrosine kinase signaling promotes S. flexneri dissemination in epithelial cells.
Collapse
|
39
|
Lum M, Morona R. Myosin IIA is essential for Shigella flexneri cell-to-cell spread. Pathog Dis 2014; 72:174-87. [PMID: 24989342 DOI: 10.1111/2049-632x.12202] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/29/2014] [Accepted: 06/24/2014] [Indexed: 11/26/2022] Open
Abstract
A key feature of Shigella pathogenesis is the ability to spread from cell-to-cell post-invasion. This is dependent on the bacteria's ability to initiate de novo F-actin tail polymerisation, followed by protrusion formation, uptake of bacteria-containing protrusion and finally, lysis of the double membrane vacuole in the adjacent cell. In epithelial cells, cytoskeletal tension is maintained by the actin-myosin II networks. In this study, the role of myosin II and its specific kinase, myosin light chain kinase (MLCK), during Shigella intercellular spreading was investigated in HeLa cells. Inhibition of MLCK and myosin II, as well as myosin IIA knockdown, significantly reduced Shigella plaque and infectious focus formation. Protrusion formation and intracellular bacterial growth was not affected. Low levels of myosin II were localised to the Shigella F-actin tail. HeLa cells were also infected with Shigella strains defective in cell-to-cell spreading. Unexpectedly loss of myosin IIA labelling was observed in HeLa cells infected with these mutant strains. This phenomenon was not observed with WT Shigella or with the less abundant myosin IIB isoform, suggesting a critical role for myosin IIA.
Collapse
Affiliation(s)
- Mabel Lum
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | | |
Collapse
|
40
|
Baxt LA, Goldberg MB. Host and bacterial proteins that repress recruitment of LC3 to Shigella early during infection. PLoS One 2014; 9:e94653. [PMID: 24722587 PMCID: PMC3983221 DOI: 10.1371/journal.pone.0094653] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/17/2014] [Indexed: 11/19/2022] Open
Abstract
Shigella spp. are intracytosolic gram-negative pathogens that cause disease by invasion and spread through the colonic mucosa, utilizing host cytoskeletal components to form propulsive actin tails. We have previously identified the host factor Toca-1 as being recruited to intracellular S. flexneri and being required for efficient bacterial actin tail formation. We show that at early times during infection (40 min.), the type three-secreted effector protein IcsB recruits Toca-1 to intracellular bacteria and that recruitment of Toca-1 is associated with repression of recruitment of LC3, as well as with repression of recruitment of the autophagy marker NDP52, around these intracellular bacteria. LC3 is best characterized as a marker of autophagosomes, but also marks phagosomal membranes in the process LC3-associated phagocytosis. IcsB has previously been demonstrated to be required for S. flexneri evasion of autophagy at late times during infection (4-6 hr) by inhibiting binding of the autophagy protein Atg5 to the Shigella surface protein IcsA (VirG). Our results suggest that IcsB and Toca-1 modulation of LC3 recruitment restricts LC3-associated phagocytosis and/or LC3 recruitment to vacuolar membrane remnants. Together with published results, our findings suggest that IcsB inhibits innate immune responses in two distinct ways, first, by inhibiting LC3-associated phagocytosis and/or LC3 recruitment to vacuolar membrane remnants early during infection, and second, by inhibiting autophagy late during infection.
Collapse
Affiliation(s)
- Leigh A. Baxt
- Department of Medicine, Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marcia B. Goldberg
- Department of Medicine, Massachusetts General Hospital, Cambridge, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
41
|
Scribano D, Petrucca A, Pompili M, Ambrosi C, Bruni E, Zagaglia C, Prosseda G, Nencioni L, Casalino M, Polticelli F, Nicoletti M. Polar localization of PhoN2, a periplasmic virulence-associated factor of Shigella flexneri, is required for proper IcsA exposition at the old bacterial pole. PLoS One 2014; 9:e90230. [PMID: 24587292 PMCID: PMC3937361 DOI: 10.1371/journal.pone.0090230] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/27/2014] [Indexed: 12/18/2022] Open
Abstract
Proper protein localization is critical for bacterial virulence. PhoN2 is a virulence-associated ATP-diphosphohydrolase (apyrase) involved in IcsA-mediated actin-based motility of S. flexneri. Herein, by analyzing a ΔphoN2 mutant of the S. flexneri strain M90T and by generating phoN2::HA fusions, we show that PhoN2, is a periplasmic protein that strictly localizes at the bacterial poles, with a strong preference for the old pole, the pole where IcsA is exposed, and that it is required for proper IcsA exposition. PhoN2-HA was found to be polarly localized both when phoN2::HA was ectopically expressed in a Escherichia coli K-12 strain and in a S. flexneri virulence plasmid-cured mutant, indicating a conserved mechanism of PhoN2 polar delivery across species and that neither IcsA nor the expression of other virulence-plasmid encoded genes are involved in this process. To assess whether PhoN2 and IcsA may interact, two-hybrid and cross-linking experiments were performed. While no evidence was found of a PhoN2-IcsA interaction, unexpectedly the outer membrane protein A (OmpA) was shown to bind PhoN2-HA through its periplasmic-exposed C-terminal domain. Therefore, to identify PhoN2 domains involved in its periplasmic polar delivery as well as in the interaction with OmpA, a deletion and a set of specific amino acid substitutions were generated. Analysis of these mutants indicated that neither the (183)PAPAP(187) motif of OmpA, nor the N-terminal polyproline (43)PPPP(46) motif and the Y155 residue of PhoN2 are involved in this interaction while P45, P46 and Y155 residues were found to be critical for the correct folding and stability of the protein. The relative rapid degradation of these amino acid-substituted recombinant proteins was found to be due to unknown S. flexneri-specific protease(s). A model depicting how the PhoN2-OmpA interaction may contribute to proper polar IcsA exposition in S. flexneri is presented.
Collapse
Affiliation(s)
- Daniela Scribano
- Dipartimento di Scienze Sperimentali e Cliniche, Università “G. D'Annunzio”, Chieti, Italy
| | - Andrea Petrucca
- Dipartimento di Scienze Sperimentali e Cliniche, Università “G. D'Annunzio”, Chieti, Italy
| | - Monica Pompili
- Dipartimento di Sanità Pubblica e Malattie Infettive Università “Sapienza” di Roma, Rome, Italy
| | - Cecilia Ambrosi
- Dipartimento di Sanità Pubblica e Malattie Infettive Università “Sapienza” di Roma, Rome, Italy
| | - Elena Bruni
- Dipartimento di Sanità Pubblica e Malattie Infettive Università “Sapienza” di Roma, Rome, Italy
| | - Carlo Zagaglia
- Dipartimento di Sanità Pubblica e Malattie Infettive Università “Sapienza” di Roma, Rome, Italy
| | - Gianni Prosseda
- Dipartimento di Biologia e Biotecnologie “C. Darwin”, Università Sapienza di Roma, Rome, Italy
| | - Lucia Nencioni
- Dipartimento di Sanità Pubblica e Malattie Infettive Università “Sapienza” di Roma, Rome, Italy
| | | | - Fabio Polticelli
- Dipartimento di Scienze, Università di “Roma Tre”, Rome, Italy
- Istituto Nazionale di Fisica Nucleare, Sezione di “Roma Tre”, Rome, Italy
| | - Mauro Nicoletti
- Dipartimento di Scienze Sperimentali e Cliniche, Università “G. D'Annunzio”, Chieti, Italy
| |
Collapse
|
42
|
Abstract
Shigella flexneri is an enteropathogenic bacterium responsible for approximately 100 million cases of severe dysentery each year. S. flexneri colonization of the human colonic epithelium is supported by direct spread from cell to cell, which relies on actin-based motility. We have recently uncovered that, in intestinal epithelial cells, S. flexneri actin-based motility is regulated by the Bruton's tyrosine kinase (Btk). Consequently, treatment with Ibrutinib, a specific Btk inhibitor currently used in the treatment of B-cell malignancies, effectively impaired S. flexneri spread from cell to cell. Thus, therapeutic intervention capitalizing on drugs interfering with host factors supporting the infection process may represent an effective alternative to treatments with antimicrobial compounds.
Collapse
|
43
|
Lum M, Attridge SR, Morona R. Impact of dynasore an inhibitor of dynamin II on Shigella flexneri infection. PLoS One 2013; 8:e84975. [PMID: 24367704 PMCID: PMC3868620 DOI: 10.1371/journal.pone.0084975] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 11/27/2013] [Indexed: 11/19/2022] Open
Abstract
Shigella flexneri remains a significant human pathogen due to high morbidity among children < 5 years in developing countries. One of the key features of Shigella infection is the ability of the bacterium to initiate actin tail polymerisation to disseminate into neighbouring cells. Dynamin II is associated with the old pole of the bacteria that is associated with F-actin tail formation. Dynamin II inhibition with dynasore as well as siRNA knockdown significantly reduced Shigella cell to cell spreading in vitro. The ocular mouse Sereny model was used to determine if dynasore could delay the progression of Shigella infection in vivo. While dynasore did not reduce ocular inflammation, it did provide significant protection against weight loss. Therefore dynasore's effects in vivo are unlikely to be related to the inhibition of cell spreading observed in vitro. We found that dynasore decreased S. flexneri-induced HeLa cell death in vitro which may explain the protective effect observed in vivo. These results suggest the administration of dynasore or a similar compound during Shigella infection could be a potential intervention strategy to alleviate disease symptoms.
Collapse
Affiliation(s)
- Mabel Lum
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Stephen R. Attridge
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Renato Morona
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
44
|
Kolonko M, Geffken AC, Blumer T, Hagens K, Schaible UE, Hagedorn M. WASH-driven actin polymerization is required for efficient mycobacterial phagosome maturation arrest. Cell Microbiol 2013; 16:232-46. [PMID: 24119059 DOI: 10.1111/cmi.12217] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/21/2013] [Accepted: 09/19/2013] [Indexed: 12/24/2022]
Abstract
Pathogenic mycobacteria survive in phagocytic host cells primarily as a result of their ability to prevent fusion of their vacuole with lysosomes, thereby avoiding a bactericidal environment. The molecular mechanisms to establish and maintain this replication compartment are not well understood. By combining molecular and microscopical approaches we show here that after phagocytosis the actin nucleation-promoting factor WASH associates and generates F-actin on the mycobacterial vacuole. Disruption of WASH or depolymerization of F-actin leads to the accumulation of the proton-pumping V-ATPase around the mycobacterial vacuole, its acidification and reduces the viability of intracellular mycobacteria. This effect is observed for M. marinum in the model phagocyte Dictyostelium but also for M. marinum and M. tuberculosis in mammalian phagocytes. This demonstrates an evolutionarily conserved mechanism by which pathogenic mycobacteria subvert the actin-polymerization activity of WASH to prevent phagosome acidification and maturation, as a prerequisite to generate and maintain a replicative niche.
Collapse
Affiliation(s)
- Margot Kolonko
- Section Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Besingi RN, Chaney JL, Clark PL. An alternative outer membrane secretion mechanism for an autotransporter protein lacking a C-terminal stable core. Mol Microbiol 2013; 90:1028-45. [PMID: 24118465 DOI: 10.1111/mmi.12414] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2013] [Indexed: 01/24/2023]
Abstract
Autotransporter (AT) proteins are a broad class of virulence factors from Gram-negative pathogens. AT outer membrane (OM) secretion appears simple in many regards, yet the mechanism that enables transport of the central AT 'passenger' across the OM remains unclear. OM secretion efficiency for two AT passengers is enhanced by approximately 20 kDa stable core at the C-terminus of the passenger, but studies on a broader range of AT proteins are needed in order to determine whether a stability difference between the passenger N- and C-terminus represents a truly common mechanistic feature. Yersinia pestis YapV is homologous to Shigella flexneri IcsA, and like IcsA, YapV recruits mammalian neural Wiskott-Aldrich syndrome protein (N-WASP). In vitro, the purified YapV passenger is functional and rich in β-sheet structure, but lacks a approximately 20 kDa C-terminal stable core. However, the N-terminal 49 residues of the YapV passenger globally destabilize the entire YapV passenger, enhancing its OM secretion efficiency. These results indicate that the contributions of AT passenger sequences to OM secretion efficiency extend beyond a C-terminal stable core, and highlight a role of the passenger N-terminus in reducing passenger stability in order to facilitate OM secretion of some AT proteins.
Collapse
Affiliation(s)
- Richard N Besingi
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | | | | |
Collapse
|
46
|
Abstract
Intracellular pathogens have developed elaborate mechanisms to exploit the different cellular systems of their unwilling hosts to facilitate their entry, replication, and survival. In particular, a diverse range of bacteria and viruses have evolved unique strategies to harness the power of Arp2/3-mediated actin polymerization to enhance their cell-to-cell spread. In this review, we discuss how studying these pathogens has revolutionized our molecular understanding of Arp2/3-dependent actin assembly and revealed key signaling pathways regulating actin assembly in cells. Future analyses of microbe-host interactions are likely to continue uncovering new mechanisms regulating actin assembly and dynamics, as well as unexpected cellular functions for actin. Further, studies with known and newly emerging pathogens will also undoubtedly continue to enhance our understanding of the role of the actin cytoskeleton during pathogenesis and potentially highlight future therapeutic approaches.
Collapse
Affiliation(s)
- Matthew D Welch
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | | |
Collapse
|
47
|
Small-molecule inhibitor of the Shigella flexneri master virulence regulator VirF. Infect Immun 2013; 81:4220-31. [PMID: 24002059 DOI: 10.1128/iai.00919-13] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
VirF is an AraC family transcriptional activator that is required for the expression of virulence genes associated with invasion and cell-to-cell spread by Shigella flexneri, including multiple components of the type three secretion system (T3SS) machinery and effectors. We tested a small-molecule compound, SE-1 (formerly designated OSSL_051168), which we had identified as an effective inhibitor of the AraC family proteins RhaS and RhaR, for its ability to inhibit VirF. Cell-based reporter gene assays with Escherichia coli and Shigella, as well as in vitro DNA binding assays with purified VirF, demonstrated that SE-1 inhibited DNA binding and transcription activation (likely by blocking DNA binding) by VirF. Analysis of mRNA levels using real-time quantitative reverse transcription-PCR (qRT-PCR) further demonstrated that SE-1 reduced the expression of the VirF-dependent virulence genes icsA, virB, icsB, and ipaB in Shigella. We also performed eukaryotic cell invasion assays and found that SE-1 reduced invasion by Shigella. The effect of SE-1 on invasion required preincubation of Shigella with SE-1, in agreement with the hypothesis that SE-1 inhibited the expression of VirF-activated genes required for the formation of the T3SS apparatus and invasion. We found that the same concentrations of SE-1 had no detectable effects on the growth or metabolism of the bacterial cells or the eukaryotic host cells, respectively, indicating that the inhibition of invasion was not due to general toxicity. Overall, SE-1 appears to inhibit transcription activation by VirF, exhibits selectivity toward AraC family proteins, and has the potential to be developed into a novel antibacterial agent.
Collapse
|
48
|
Abstract
Several bacterial pathogens, including Listeria monocytogenes, Shigella flexneri and Rickettsia spp., have evolved mechanisms to actively spread within human tissues. Spreading is initiated by the pathogen-induced recruitment of host filamentous (F)-actin. F-actin forms a tail behind the microbe, propelling it through the cytoplasm. The motile pathogen then encounters the host plasma membrane, forming a bacterium-containing protrusion that is engulfed by an adjacent cell. Over the past two decades, much progress has been made in elucidating mechanisms of F-actin tail formation. Listeria and Shigella produce tails of branched actin filaments by subverting the host Arp2/3 complex. By contrast, Rickettsia forms tails with linear actin filaments through a bacterial mimic of eukaryotic formins. Compared with F-actin tail formation, mechanisms controlling bacterial protrusions are less well understood. However, recent findings have highlighted the importance of pathogen manipulation of host cell–cell junctions in spread. Listeria produces a soluble protein that enhances bacterial protrusions by perturbing tight junctions. Shigella protrusions are engulfed through a clathrin-mediated pathway at ‘tricellular junctions’—specialized membrane regions at the intersection of three epithelial cells. This review summarizes key past findings in pathogen spread, and focuses on recent developments in actin-based motility and the formation and internalization of bacterial protrusions.
Collapse
Affiliation(s)
- Keith Ireton
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
49
|
Bettencourt P, Marion S, Pires D, Santos LF, Lastrucci C, Carmo N, Blake J, Benes V, Griffiths G, Neyrolles O, Lugo-Villarino G, Anes E. Actin-binding protein regulation by microRNAs as a novel microbial strategy to modulate phagocytosis by host cells: the case of N-Wasp and miR-142-3p. Front Cell Infect Microbiol 2013; 3:19. [PMID: 23760605 PMCID: PMC3672780 DOI: 10.3389/fcimb.2013.00019] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 05/17/2013] [Indexed: 12/26/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a successful intracellular pathogen that thrives in macrophages (Mφs). There is a need to better understand how Mtb alters cellular processes like phagolysosome biogenesis, a classical determinant of its pathogenesis. A central feature of this bacteria's strategy is the manipulation of Mφ actin. Here, we examined the role of microRNAs (miRNAs) as a potential mechanism in the regulation of actin-mediated events leading to phagocytosis in the context of mycobacteria infection. Given that non-virulent Mycobacterium smegmatis also controls actin filament assembly to prolong its intracellular survival inside host cells, we performed a global transcriptomic analysis to assess the modulation of miRNAs upon M. smegmatis infection of the murine Mφ cell line, J774A.1. This approach identified miR-142-3p as a key candidate to be involved in the regulation of actin dynamics required in phagocytosis. We unequivocally demonstrate that miR-142-3p targets N-Wasp, an actin-binding protein required during microbial challenge. A gain-of-function approach for miR-142-3p revealed a down-regulation of N-Wasp expression accompanied by a decrease of mycobacteria intake, while a loss-of-function approach yielded the reciprocal increase of the phagocytosis process. Equally important, we show Mtb induces the early expression of miR-142-3p and partially down-regulates N-Wasp protein levels in both the murine J774A.1 cell line and primary human Mφs. As proof of principle, the partial siRNA-mediated knock down of N-Wasp resulted in a decrease of Mtb intake by human Mφs, reflected in lower levels of colony-forming units (CFU) counts over time. We therefore propose the modulation of miRNAs as a novel strategy in mycobacterial infection to control factors involved in actin filament assembly and other early events of phagolysosome biogenesis.
Collapse
Affiliation(s)
- Paulo Bettencourt
- Centro de Patogénese Molecular, Faculdade de Farmácia, Unidade dos Retrovírus e Infecções Associadas e Instituto de Medicina Molecular, Universidade de Lisboa Lisboa, Portugal
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Human endothelial cells internalize Candida parapsilosis via N-WASP-mediated endocytosis. Infect Immun 2013; 81:2777-87. [PMID: 23690407 DOI: 10.1128/iai.00535-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Candida parapsilosis is a frequent cause of disseminated candidiasis and is associated with significant morbidity and mortality. Although important in pathogenesis, interactions of this organism with endothelial cells have received less attention than those of Candida albicans. Internalization of C. parapsilosis by monolayers of human endothelial cells was examined in an in vitro assay and compared to that of C. albicans. Both live and heat-killed yeast were efficiently internalized, with heat-killed yeast subsequently being detected in an acidic subcompartment. Internalization was marked by a process of engulfment by thin membrane extensions from the endothelium. Efficiency of internalization differed among different clinical isolates and species of yeast. Opsonization of C. parapsilosis by serum factors was not sufficient to cause endocytosis; instead, serum appeared to directly stimulate endothelial uptake. Colocalization of endothelial actin and N-WASP at sites of C. parapsilosis internalization was observed. A Förster-resonance energy transfer (FRET) probe for N-WASP activity showed active N-WASP at sites of internalization for both live and heat-killed C. parapsilosis and C. albicans. An actin nucleation inhibitor (cytochalasin D) and an N-WASP inhibitor (wiskostatin) both inhibited uptake of heat-killed C. parapsilosis, as did short interfering RNA-mediated ablation of N-WASP. Thus, endocytosis by endothelial cells may represent a means of traversal of the blood vessel wall by yeast during disseminated candidiasis, and N-WASP may play a key role in the process.
Collapse
|