Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
mBio ; 15(1): e0285723, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38051116

ABSTRACT

IMPORTANCE: Bacteria are constantly exchanging DNA, which constitutes horizontal gene transfer. While some of these occurs by a non-specific process called natural transformation, some occurs by a specific mating between a donor and a recipient cell. In specific conjugation, the mating pilus is extended from the donor cell to make contact with the recipient cell, but whether DNA is actually transferred through this pilus or by another mechanism involving the type IV secretion system complex without the pilus has been an open question. Using Escherichia coli, we show that DNA can be transferred through this pilus between a donor and a recipient cell that has not established a tight mating junction, providing a new picture for the role of this pilus.


Subject(s)
Escherichia coli , Gene Transfer, Horizontal , Escherichia coli/genetics , Escherichia coli/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Conjugation, Genetic , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Plasmids
2.
Small GTPases ; 13(1): 296-306, 2022 01.
Article in English | MEDLINE | ID: mdl-35950594

ABSTRACT

The small GTPase RhoA controls many important cellular processes through its ability to activate multiple downstream effector pathways. Most RhoA effectors contain a Rho-binding domain (RBD), and interaction between active RhoA and the RBD typically induces a conformational change in effectors that stimulates their recruitment or activity. Isolated GTPase binding domains fused to GST have been widely used in so-called pulldown assays to measure the activation state of other GTPases in cell lysates. Similarly, GST fusions containing the RBD of the RhoA effector Rhotekin have been widely adopted as a standardized tool for the measurement of RhoA activation. RBDs have also been used to generate fluorescent reporter constructs to localize sites of GTPase activation in intact cells. In this report, we demonstrate that not all forms of active RhoA are capable of interacting with the Rhotekin RBD. A constitutively active RhoA-G14V mutant, which interacted with the RBDs of ROCK2 and mDIA1, was unable to bind the Rhotekin RBD as evidenced by both conventional GST pulldown assay and our newly established BRET assay. Furthermore, active RhoA induced by different stimuli in cells also displayed binding preference for its diverse effectors. Our data demonstrate that RhoA may undergo effector-specific activation for differential regulation of its downstream pathways, and that RhoA activation should not be defined solely by its interaction with Rhotekin.


Subject(s)
rhoA GTP-Binding Protein , Protein Binding , rhoA GTP-Binding Protein/metabolism
4.
Mol Biol Cell ; 33(2): ar13, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34818063

ABSTRACT

ELMODs are a family of three mammalian paralogues that display GTPase-activating protein (GAP) activity toward a uniquely broad array of ADP-ribosylation factor (ARF) family GTPases that includes ARF-like (ARL) proteins. ELMODs are ubiquitously expressed in mammalian tissues, highly conserved across eukaryotes, and ancient in origin, being present in the last eukaryotic common ancestor. We described functions of ELMOD2 in immortalized mouse embryonic fibroblasts (MEFs) in the regulation of cell division, microtubules, ciliogenesis, and mitochondrial fusion. Here, using similar strategies with the paralogues ELMOD1 and ELMOD3, we identify novel functions and locations of these cell regulators and compare them to those of ELMOD2, allowing the determination of functional redundancy among the family members. We found strong similarities in phenotypes resulting from deletion of either Elmod1 or Elmod3 and marked differences from those arising in Elmod2 deletion lines. Deletion of either Elmod1 or Elmod3 results in the decreased ability of cells to form primary cilia, loss of a subset of proteins from cilia, and accumulation of some ciliary proteins at the Golgi, predicted to result from compromised traffic from the Golgi to cilia. These phenotypes are reversed upon activating mutant expression of either ARL3 or ARL16, linking their roles to ELMOD1/3 actions.


Subject(s)
GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Cilia/metabolism , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , GTPase-Activating Proteins/physiology , Golgi Apparatus/metabolism , Mice , Microtubules/metabolism , Mitochondrial Dynamics , Signal Transduction/genetics
5.
Cell Microbiol ; 23(8): e13329, 2021 08.
Article in English | MEDLINE | ID: mdl-33742761

ABSTRACT

Upon infection of host cells, Salmonella enterica serovar Typhimurium resides in a modified-endosomal compartment referred to as the Salmonella-containing vacuole (SCV). SCV biogenesis is driven by multiple effector proteins translocated through two type III secretion systems (T3SS-1 and T3SS-2). While many host proteins targeted by these effector proteins have been characterised, the role of host lipids in SCV dynamics remains poorly understood. Previous studies have shown that S. Typhimurium infection in macrophages leads to accumulation of intracellular cholesterol, some of which concentrates in and around SCVs; however, the underlying mechanisms remain unknown. Here, we show that S. Typhimurium utilises the T3SS-2 effector SseJ to downregulate expression of the host cholesterol transporter ABCA1 in macrophages, leading to a ~45% increase in cellular cholesterol. Mechanistically, SseJ activates a signalling cascade involving the host kinases FAK and Akt to suppress Abca1 expression. Mutational inactivation of SseJ acyltransferase activity, silencing FAK, or inhibiting Akt prevents Abca1 downregulation and the corresponding accumulation of cholesterol during infection. Importantly, RNAi-mediated silencing of ABCA1 rescued bacterial survival in FAK-deficient macrophages, suggesting that Abca1 downregulation and cholesterol accumulation are important for intracellular survival.


Subject(s)
Carrier Proteins , Salmonella typhimurium , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cholesterol , Homeostasis , Macrophages/metabolism , Salmonella typhimurium/metabolism
6.
Elife ; 92020 04 01.
Article in English | MEDLINE | ID: mdl-32234213

ABSTRACT

Coordinated assembly and disassembly of integrin-mediated focal adhesions (FAs) is essential for cell migration. Many studies have shown that FA disassembly requires Ca2+ influx, however our understanding of this process remains incomplete. Here, we show that Ca2+ influx via STIM1/Orai1 calcium channels, which cluster near FAs, leads to activation of the GTPase Arf5 via the Ca2+-activated GEF IQSec1, and that both IQSec1 and Arf5 activation are essential for adhesion disassembly. We further show that IQSec1 forms a complex with the lipid transfer protein ORP3, and that Ca2+ influx triggers PKC-dependent translocation of this complex to ER/plasma membrane (PM) contact sites adjacent to FAs. In addition to allosterically activating IQSec1, ORP3 also extracts PI4P from the PM, in exchange for phosphatidylcholine. ORP3-mediated lipid exchange is also important for FA turnover. Together, these findings identify a new pathway that links calcium influx to FA turnover during cell migration.


Subject(s)
Calcium/metabolism , Fatty Acid-Binding Proteins/physiology , Focal Adhesions/physiology , Guanine Nucleotide Exchange Factors/physiology , ADP-Ribosylation Factors/physiology , Cell Membrane/metabolism , Cells, Cultured , Humans , Lipid Metabolism , Phosphatidylcholines/metabolism , Phosphatidylinositols/physiology
7.
Mol Biol Cell ; 30(11): 1249-1271, 2019 05 15.
Article in English | MEDLINE | ID: mdl-31084567

ABSTRACT

Detailed structural, biochemical, cell biological, and genetic studies of any gene/protein are required to develop models of its actions in cells. Studying a protein family in the aggregate yields additional information, as one can include analyses of their coevolution, acquisition or loss of functionalities, structural pliability, and the emergence of shared or variations in molecular mechanisms. An even richer understanding of cell biology can be achieved through evaluating functionally linked protein families. In this review, we summarize current knowledge of three protein families: the ARF GTPases, the guanine nucleotide exchange factors (ARF GEFs) that activate them, and the GTPase-activating proteins (ARF GAPs) that have the ability to both propagate and terminate signaling. However, despite decades of scrutiny, our understanding of how these essential proteins function in cells remains fragmentary. We believe that the inherent complexity of ARF signaling and its regulation by GEFs and GAPs will require the concerted effort of many laboratories working together, ideally within a consortium to optimally pool information and resources. The collaborative study of these three functionally connected families (≥70 mammalian genes) will yield transformative insights into regulation of cell signaling.


Subject(s)
GTPase-Activating Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Animals , Eukaryota/metabolism , Humans
8.
Sci Rep ; 9(1): 4497, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30872746

ABSTRACT

Adhesion signaling between epithelial cells and the extracellular matrix plays a critical role in maintaining tissue homeostasis and the response to tissue damage. Focal adhesion kinase (FAK) and its close relative Pyk2 are non-receptor tyrosine kinases that mediate adhesion signaling to promote cell proliferation, motility and survival. FAK has also been shown to act as a mechanosensor by modulating cell proliferation in response to changes in tissue compliance. We previously showed that mice lacking FAK in the intestinal epithelium are phenotypically normal under homeostatic conditions but hypersensitive to experimental colitis induced by dextran sulfate sodium (DSS). Here we report that Pyk2-deficient mice are also phenotypically normal under homeostatic conditions and are similarly hypersensitive to DSS-induced colitis. These data indicate that normal intestinal development and homeostatic maintenance can occur in the presence of either FAK or Pyk2, but that both kinases are necessary for epithelial repair following injury. In contrast, mice lacking both FAK and Pyk2 develop spontaneous colitis with 100% penetrance by 4 weeks of age. Normal colonic phenotype and function are restored upon treatment of the double knockout mice with antibiotics, implicating commensal bacteria or bacterial products in the etiology of the spontaneous colitis exhibited by these mice.


Subject(s)
Colitis/genetics , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 2/genetics , Intestinal Mucosa/cytology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cells, Cultured , Colitis/drug therapy , Colitis/metabolism , Colitis/microbiology , Disease Models, Animal , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/metabolism , Gastrointestinal Microbiome/drug effects , Gene Knockout Techniques , Homeostasis , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice
9.
J Cell Biol ; 216(10): 2995-2997, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28928133

ABSTRACT

Endosome maturation requires a coordinated change in the Rab GTPase and phosphoinositide composition of the endosomal membrane. In this issue, Liu et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201705151) identify WDR91 as a ubiquitous Rab7 effector that inhibits phosphatidylinositol 3-kinase activity on endosomes and is critical for endosome maturation, viability, and dendrite growth of neurons in vivo.


Subject(s)
Endosomes , rab GTP-Binding Proteins , Phosphatidylinositol 3-Kinases , Phosphatidylinositols
10.
Cell Mol Gastroenterol Hepatol ; 4(2): 237-243, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28660242

ABSTRACT

Autophagy is a fundamental cellular process used for the turnover and recycling of cytosolic components and damaged organelles. Originally characterized as a response to cellular stress, it now is well established that autophagy also is used as a defensive mechanism to combat the infection of host cells by intracellular pathogens. However, although this defensive strategy does limit the proliferation of most pathogens within their host cells, successful pathogens have evolved countermeasures that subvert or circumvent the autophagic response. In this review, we discuss the mechanisms used by a number of these pathogens to escape autophagy, with a particular focus on Salmonella enterica serovar Typhimurium, which has been the most extensively studied example. We also discuss the consequences of bacterial autophagy for the broader innate immune response.

11.
Small GTPases ; 7(4): 257-264, 2016 10.
Article in English | MEDLINE | ID: mdl-27739918

ABSTRACT

The IQSec/BRAG proteins are a subfamily of Arf-nucleotide exchange factors. Since their discovery almost 15 y ago, the BRAGs have been reported to be involved in diverse physiological processes from myoblast fusion, neuronal pathfinding and angiogenesis, to pathophysiological processes including X-linked intellectual disability and tumor metastasis. In this review we will address how, in each of these situations, the BRAGs are thought to regulate the surface levels of adhesive and signaling receptors. While in most cases BRAGs are thought to enhance the endocytosis of these receptors, how they achieve this remains unclear. Similarly, while all 3 BRAG proteins contain calmodulin-binding IQ motifs, little is known about how their activities might be regulated by calcium. These are some of the questions that are likely to form the basis of future research.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Myoblasts/physiology , Neovascularization, Physiologic , Neurons/metabolism , Animals , Binding Sites , Calcium/metabolism , Calmodulin/metabolism , Endocytosis , Guanine Nucleotide Exchange Factors/chemistry , Humans , Protein Binding , Signal Transduction
12.
Sci Signal ; 9(413): ra14, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26838550

ABSTRACT

The detection of microbes and initiation of an innate immune response occur through pattern recognition receptors (PRRs), which are critical for the production of inflammatory cytokines and activation of the cellular microbicidal machinery. In particular, the production of reactive oxygen species (ROS) by the NADPH oxidase complex is a critical component of the macrophage bactericidal machinery. We previously characterized brain-specific angiogenesis inhibitor 1 (BAI1), a member of the adhesion family of G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors (GPCRs), as a PRR that mediates the selective phagocytic uptake of Gram-negative bacteria by macrophages. We showed that BAI1 promoted phagosomal ROS production through activation of the Rho family guanosine triphosphatase (GTPase) Rac1, thereby stimulating NADPH oxidase activity. Primary BAI1-deficient macrophages exhibited attenuated Rac GTPase activity and reduced ROS production in response to several Gram-negative bacteria, resulting in impaired microbicidal activity. Furthermore, in a peritoneal infection model, BAI1-deficient mice exhibited increased susceptibility to death by bacterial challenge because of impaired bacterial clearance. Together, these findings suggest that BAI1 mediates the clearance of Gram-negative bacteria by stimulating both phagocytosis and NADPH oxidase activation, thereby coupling bacterial detection to the cellular microbicidal machinery.


Subject(s)
Angiogenic Proteins/metabolism , Gram-Negative Bacteria , Macrophages, Peritoneal/metabolism , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Enzyme Activation , Mice , NADPH Oxidases , Neuropeptides/metabolism , rac1 GTP-Binding Protein/metabolism
13.
mBio ; 7(1): e02051-15, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26884434

ABSTRACT

UNLABELLED: Salmonella enterica is an intracellular pathogen that causes diseases ranging from gastroenteritis to typhoid fever. Salmonella bacteria trigger an autophagic response in host cells upon infection but have evolved mechanisms for suppressing this response, thereby enhancing intracellular survival. We recently reported that S. enterica serovar Typhimurium actively recruits the host tyrosine kinase focal adhesion kinase (FAK) to the surface of the Salmonella-containing vacuole (SCV) (K. A. Owen et al., PLoS Pathog 10:e1004159, 2014). FAK then suppresses autophagy through activation of the Akt/mTORC1 signaling pathway. In FAK(-/-) macrophages, bacteria are captured in autophagosomes and intracellular survival is attenuated. Here we show that the cell-autonomous bacterial suppression of autophagy also suppresses the broader innate immune response by inhibiting production of beta interferon (IFN-ß). Induction of bacterial autophagy (xenophagy), but not autophagy alone, triggers IFN-ß production through a pathway involving the adapter TRIF and endosomal Toll-like receptor 3 (TLR3) and TLR4. Selective FAK knockout in macrophages resulted in rapid bacterial clearance from mucosal tissues after oral infection. Clearance correlated with increased IFN-ß production by intestinal macrophages and with IFN-ß-dependent induction of IFN-γ by intestinal NK cells. Blockade of either IFN-ß or IFN-γ increased host susceptibility to infection, whereas experimental induction of IFN-ß was protective. Thus, bacterial suppression of autophagy not only enhances cell-autonomous survival but also suppresses more-systemic innate immune responses by limiting type I and type II interferons. IMPORTANCE: Salmonella enterica serovar Typhimurium represents one of the most commonly identified bacterial causes of foodborne illness worldwide. S. Typhimurium has developed numerous strategies to evade detection by the host immune system. Autophagy is a cellular process that involves the recognition and degradation of defective proteins and organelles. More recently, autophagy has been described as an important means by which host cells recognize and eliminate invading intracellular pathogens and plays a key role in the production of cytokines. Previously, we determined that Salmonella bacteria are able to suppress their own autophagic capture and elimination by macrophages. Building on that study, we show here that the inhibition of autophagy by Salmonella also prevents the induction of a protective cytokine response mediated by beta interferon (IFN-ß) and IFN-γ. Together, these findings identify a novel virulence strategy whereby Salmonella bacteria prevent cell autonomous elimination via autophagy and suppress the activation of innate immune responses.


Subject(s)
Autophagy , Interferon-beta/biosynthesis , Interferon-beta/immunology , Macrophages/immunology , Macrophages/microbiology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/pathogenicity , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Immunity, Innate , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Intestines/cytology , Killer Cells, Natural/immunology , Mice , Mice, Knockout , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/immunology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
14.
J Virol ; 90(1): 605-10, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26468524

ABSTRACT

Ebolavirus, a deadly hemorrhagic fever virus, was thought to enter cells through endolysosomes harboring its glycoprotein receptor, Niemann-Pick C1. However, an alternate model was recently proposed in which ebolavirus enters through a later NPC1-negative endosome that contains two-pore Ca(2+) channel 2 (TPC2), a newly identified ebolavirus entry factor. Here, using live cell imaging, we obtained evidence that in contrast to the new model, ebolavirus enters cells through endolysosomes that contain both NPC1 and TPC2.


Subject(s)
Calcium Channels/metabolism , Carrier Proteins/metabolism , Ebolavirus/physiology , Endosomes/virology , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Animals , Cell Line , Chlorocebus aethiops , Host-Pathogen Interactions , Humans , Intracellular Signaling Peptides and Proteins , Microscopy , Models, Biological , Niemann-Pick C1 Protein
15.
Cell Host Microbe ; 18(5): 517-9, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26567504

ABSTRACT

Many intracellular pathogens, including Salmonella typhimurium, trigger autophagy in host cells, which is widely thought to restrict intracellular growth and survival. In this issue of Cell Host & Microbe, Kreibich et al. (2015) demonstrate a role for the autophagic machinery in the repair of damaged Salmonella-containing vacuoles (SCVs).


Subject(s)
Autophagy , Salmonella typhimurium , Cytoplasm , Humans , Salmonella Infections , Vacuoles
16.
J Virol ; 89(5): 2931-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552710

ABSTRACT

UNLABELLED: Ebola virus (EBOV) causes hemorrhagic fevers with high mortality rates. During cellular entry, the virus is internalized by macropinocytosis and trafficked through endosomes until fusion between the viral and an endosomal membrane is triggered, releasing the RNA genome into the cytoplasm. We found that while macropinocytotic uptake of filamentous EBOV viruslike particles (VLPs) expressing the EBOV glycoprotein (GP) occurs relatively quickly, VLPs only begin to enter the cytoplasm after a 30-min lag, considerably later than particles bearing the influenza hemagglutinin or GP from lymphocytic choriomeningitis virus, which enter through late endosomes (LE). For EBOV, the long lag is not due to the large size or unusual shape of EBOV filaments, the need to prime EBOV GP to the 19-kDa receptor-binding species, or a need for unusually low endosomal pH. In contrast, since we observed that EBOV entry occurs upon arrival in Niemann-Pick C1 (NPC1)-positive endolysosomes (LE/Lys), we propose that trafficking to LE/Lys is a key rate-defining step. Additional experiments revealed, unexpectedly, that severe acute respiratory syndrome (SARS) S-mediated entry also begins only after a 30-min lag. Furthermore, although SARS does not require NPC1 for entry, SARS entry also begins after colocalization with NPC1. Since the only endosomal requirement for SARS entry is cathepsin L activity, we tested and provide evidence that NPC1(+) LE/Lys have higher cathepsin L activity than LE, with no detectable activity in earlier endosomes. Our findings suggest that both EBOV and SARS traffic deep into the endocytic pathway for entry and that they do so to access higher cathepsin activity. IMPORTANCE: Ebola virus is a hemorrhagic fever virus that causes high fatality rates when it spreads from zoonotic vectors into the human population. Infection by severe acute respiratory syndrome coronavirus (SARS-CoV) causes severe respiratory distress in infected patients. A devastating outbreak of EBOV occurred in West Africa in 2014, and there was a significant outbreak of SARS in 2003. No effective vaccine or treatment has yet been approved for either virus. We present evidence that both viruses traffic late into the endocytic pathway, to NPC1(+) LE/Lys, in order to enter host cells, and that they do so to access high levels of cathepsin activity, which both viruses use in their fusion-triggering mechanisms. This unexpected similarity suggests an unexplored vulnerability, trafficking to NPC1(+) LE/Lys, as a therapeutic target for SARS and EBOV.


Subject(s)
Biological Transport , Ebolavirus/physiology , Endosomes/virology , Lysosomes/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Internalization , Carrier Proteins/analysis , Cell Line , Endosomes/chemistry , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/chemistry , Membrane Glycoproteins/analysis , Niemann-Pick C1 Protein , Time Factors , Virosomes/metabolism
17.
Cell Mol Gastroenterol Hepatol ; 1(3): 311-324, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-26878033

ABSTRACT

BACKGROUNDS AND AIMS: 4-6 million people die of enteric infections each year. After invading intestinal epithelial cells, enteric bacteria encounter phagocytes. However, little is known about how phagocytes internalize the bacteria to generate host responses. Previously, we have shown that BAI1 (Brain Angiogenesis Inhibitor 1) binds and internalizes Gram-negative bacteria through an ELMO1 (Engulfment and cell Motility protein 1)/Rac1-dependent mechanism. Here we delineate the role of ELMO1 in host inflammatory responses following enteric infection. METHODS: ELMO1-depleted murine macrophage cell lines, intestinal macrophages and ELMO1 deficient mice (total or myeloid-cell specific) was infected with Salmonella enterica serovar Typhimurium. The bacterial load, inflammatory cytokines and histopathology was evaluated in the ileum, cecum and spleen. The ELMO1 dependent host cytokines were detected by a cytokine array. ELMO1 mediated Rac1 activity was measured by pulldown assay. RESULTS: The cytokine array showed reduced release of pro-inflammatory cytokines, including TNF-α and MCP-1, by ELMO1-depleted macrophages. Inhibition of ELMO1 expression in macrophages decreased Rac1 activation (~6 fold) and reduced internalization of Salmonella. ELMO1-dependent internalization was indispensable for TNF-α and MCP-1. Simultaneous inhibition of ELMO1 and Rac function virtually abrogated TNF-α responses to infection. Further, activation of NF-κB, ERK1/2 and p38 MAP kinases were impaired in ELMO1-depleted cells. Strikingly, bacterial internalization by intestinal macrophages was completely dependent on ELMO1. Salmonella infection of ELMO1-deficient mice resulted in a 90% reduction in bacterial burden and attenuated inflammatory responses in the ileum, spleen and cecum. CONCLUSION: These findings suggest a novel role for ELMO1 in facilitating intracellular bacterial sensing and the induction of inflammatory responses following infection with Salmonella.

18.
Cell Microbiol ; 17(6): 843-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25486861

ABSTRACT

Salmonella enterica Typhimurium induces intestinal inflammation through the activity of type III secreted effector (T3SE) proteins. Our prior results indicate that the secretion of the T3SE SipA and the ability of SipA to induce epithelial cell responses that lead to induction of polymorphonuclear transepithelial migration are not coupled to its direct delivery into epithelial cells from Salmonella. We therefore tested the hypothesis that SipA interacts with a membrane protein located at the apical surface of intestinal epithelial cells. Employing a split ubiquitin yeast-two-hybrid screen, we identified the tetraspanning membrane protein, p53 effector related to PMP-22 (PERP), as a SipA binding partner. SipA and PERP appear to have intersecting activities as we found PERP to be involved in proinflammatory pathways shown to be regulated by SipA. In sum, our studies reveal a critical role for PERP in the pathogenesis of S. Typhimurium, and for the first time demonstrate that SipA, a T3SE protein, can engage a host protein at the epithelial surface.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions , Inflammation/microbiology , Inflammation/pathology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Salmonella typhimurium/immunology , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Genes, Tumor Suppressor , Humans , Protein Binding , Protein Interaction Mapping , Transendothelial and Transepithelial Migration , Two-Hybrid System Techniques
19.
PLoS Pathog ; 10(6): e1004159, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24901456

ABSTRACT

Autophagy has emerged as an important antimicrobial host defense mechanism that not only orchestrates the systemic immune response, but also functions in a cell autonomous manner to directly eliminate invading pathogens. Pathogenic bacteria such as Salmonella have evolved adaptations to protect themselves from autophagic elimination. Here we show that signaling through the non-receptor tyrosine kinase focal adhesion kinase (FAK) is actively manipulated by the Salmonella SPI-2 system in macrophages to promote intracellular survival. In wild-type macrophages, FAK is recruited to the surface of the Salmonella-containing vacuole (SCV), leading to amplified signaling through the Akt-mTOR axis and inhibition of the autophagic response. In FAK-deficient macrophages, Akt/mTOR signaling is attenuated and autophagic capture of intracellular bacteria is enhanced, resulting in reduced bacterial survival. We further demonstrate that enhanced autophagy in FAK(-/-) macrophages requires the activity of Atg5 and ULK1 in a process that is distinct from LC3-assisted phagocytosis (LAP). In vivo, selective knockout of FAK in macrophages resulted in more rapid clearance of bacteria from tissues after oral infection with S. typhimurium. Clearance was correlated with reduced infiltration of inflammatory cell types into infected tissues and reduced tissue damage. Together, these data demonstrate that FAK is specifically targeted by S. typhimurium as a novel means of suppressing autophagy in macrophages, thereby enhancing their intracellular survival.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Macrophages, Peritoneal/immunology , Phagocytosis , Proto-Oncogene Proteins c-akt/metabolism , Salmonella typhimurium/immunology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy , Autophagy-Related Protein 5 , Autophagy-Related Protein-1 Homolog , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Enzyme Activation , Escherichia coli/immunology , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Focal Adhesion Kinase 1/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Microbial Viability , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Specific Pathogen-Free Organisms , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
20.
Curr Biol ; 24(11): 1187-98, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24835460

ABSTRACT

BACKGROUND: Early, sorting endosomes are a major crossroad of membrane traffic, at the intersection of the endocytic and exocytic pathways. The sorting of endosomal cargo for delivery to different subcellular destinations is mediated by a number of distinct coat protein complexes, including adaptor protein 1 (AP-1), AP-3, and Golgi-localized, gamma adaptin ear-containing, Arf-binding (GGAs) protein. Ultrastructural studies suggest that these coats assemble onto tubular subdomains of the endosomal membrane, but the mechanisms of coat recruitment and assembly at this site remain poorly understood. RESULTS: Here we report that the endosomal Rab protein Rab4 orchestrates a GTPase cascade that results in the sequential recruitment of the ADP-ribosylation factor (Arf)-like protein Arl1; the Arf-specific guanine nucleotide exchange factors BIG1 and BIG2; and the class I Arfs, Arf1 and Arf3. Knockdown of Arf1, or inhibition of BIG1 and BIG2 activity with brefeldin A results in the loss of AP-1, AP-3, and GGA-3, but not Arl1, from endosomal membranes and the formation of elongated tubules. In contrast, depletion of Arl1 randomizes the distribution of Rab4 on endosomal membranes, inhibits the formation of tubular subdomains, and blocks recruitment of BIG1 and BIG2, Arfs, and adaptor protein complexes to the endosome. CONCLUSIONS: Together these findings indicate that Arl1 links Rab4-dependent formation of endosomal sorting domains with downstream assembly of adaptor protein complexes that constitute the endosomal sorting machinery.


Subject(s)
ADP-Ribosylation Factors/genetics , Endosomes/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/genetics , Signal Transduction , rab4 GTP-Binding Proteins/genetics , ADP-Ribosylation Factors/metabolism , Animals , COS Cells , Chlorocebus aethiops , GTP Phosphohydrolases/metabolism , Humans , Membrane Proteins/metabolism , rab4 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...