Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
mBio ; 6(3): e00354-15, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25944859

ABSTRACT

UNLABELLED: Legionella pneumophila, the etiological agent of legionellosis, replicates within phagocytic cells. Crucial to biogenesis of the replicative vacuole is the Dot/Icm type 4 secretion system, which translocates a large number of effectors into the host cell cytosol. Among them is LegK2, a protein kinase that plays a key role in Legionella infection. Here, we identified the actin nucleator ARP2/3 complex as a target of LegK2. LegK2 phosphorylates the ARPC1B and ARP3 subunits of the ARP2/3 complex. LegK2-dependent ARP2/3 phosphorylation triggers global actin cytoskeleton remodeling in cells, and it impairs actin tail formation by Listeria monocytogenes, a well-known ARP2/3-dependent process. During infection, LegK2 is addressed to the Legionella-containing vacuole surface and inhibits actin polymerization on the phagosome, as revealed by legK2 gene inactivation. Consequently, LegK2 prevents late endosome/lysosome association with the phagosome and finally contributes to remodeling of the bacterium-containing phagosome into a replicative niche. The inhibition of actin polymerization by LegK2 and its effect on endosome trafficking are ARP2/3 dependent since it can be phenocopied by a specific chemical inhibitor of the ARP2/3 complex. Thus, LegK2-ARP2/3 interplay highlights an original mechanism of bacterial virulence with an unexpected role in local actin remodeling that allows bacteria to control vesicle trafficking in order to escape host defenses. IMPORTANCE: Deciphering the individual contribution of each Dot/Icm type 4 secretion system substrate to the intracellular life-style of L. pneumophila remains the principal challenge in understanding the molecular basis of Legionella virulence. Our finding that LegK2 is a Dot/Icm effector that inhibits actin polymerization on the Legionella-containing vacuole importantly contributes to the deciphering of the molecular mechanisms evolved by Legionella to counteract the endocytic pathway. Indeed, our results highlight the essential role of LegK2 in preventing late endosomes from fusing with the phagosome. More generally, this work is the first demonstration of local actin remodeling as a mechanism used by bacteria to control organelle trafficking. Further, by characterizing the role of the bacterial protein kinase LegK2, we reinforce the concept that posttranslational modifications are key strategies used by pathogens to evade host cell defenses.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Legionella pneumophila/enzymology , Legionella pneumophila/pathogenicity , Phagosomes/metabolism , Phagosomes/microbiology , Protein Kinases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endosomes/metabolism , Legionella pneumophila/genetics , Lysosomes/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Transport , Vacuoles/microbiology
2.
Cell Host Microbe ; 17(4): 515-25, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25771791

ABSTRACT

Xenophagy, an essential anti-microbial cell-autonomous mechanism, relies on the ability of the autophagic process to selectively entrap intracellular pathogens within autophagosomes to degrade them in autolysosomes. This selective targeting is carried out by specialized autophagy receptors, such as NDP52, but it is unknown whether the fusion of pathogen-containing autophagosomes with lysosomes is also regulated by pathogen-specific cellular factors. Here, we show that NDP52 also promotes the maturation of autophagosomes via its interaction with LC3A, LC3B, and/or GABARAPL2 through a distinct LC3-interacting region, and with MYOSIN VI. During Salmonella Typhimurium infection, the regulatory function of NDP52 in autophagosome maturation is complementary but independent of its function in pathogen targeting to autophagosomes, which relies on the interaction with LC3C. Thus, complete xenophagy is selectively regulated by a single autophagy receptor, which initially orchestrates bacteria targeting to autophagosomes and subsequently ensures pathogen degradation by regulating pathogen-containing autophagosome maturation.


Subject(s)
Autophagy , Epithelial Cells/immunology , Epithelial Cells/microbiology , Nuclear Proteins/metabolism , Phagosomes/metabolism , Salmonella typhimurium/immunology , HeLa Cells , Humans , Lysosomes/metabolism
3.
PLoS Pathog ; 9(9): e1003599, 2013.
Article in English | MEDLINE | ID: mdl-24086130

ABSTRACT

The interplay between autophagy and intracellular pathogens is intricate as autophagy is an essential cellular response to fight against infections, whereas numerous microbes have developed strategies to escape this process or even exploit it to their own benefit. The fine tuned timing and/or selective molecular pathways involved in the induction of autophagy upon infections could be the cornerstone allowing cells to either control intracellular pathogens, or be invaded by them. We report here that measles virus infection induces successive autophagy signallings in permissive cells, via distinct and uncoupled molecular pathways. Immediately upon infection, attenuated measles virus induces a first transient wave of autophagy, via a pathway involving its cellular receptor CD46 and the scaffold protein GOPC. Soon after infection, a new autophagy signalling is initiated which requires viral replication and the expression of the non-structural measles virus protein C. Strikingly, this second autophagy signalling can be sustained overtime within infected cells, independently of the expression of C, but via a third autophagy input resulting from cell-cell fusion and the formation of syncytia. Whereas this sustained autophagy signalling leads to the autophagy degradation of cellular contents, viral proteins escape from degradation. Furthermore, this autophagy flux is ultimately exploited by measles virus to limit the death of infected cells and to improve viral particle formation. Whereas CD150 dependent virulent strains of measles virus are unable to induce the early CD46/GOPC dependent autophagy wave, they induce and exploit the late and sustained autophagy. Overall, our work describes distinct molecular pathways for an induction of self-beneficial sustained autophagy by measles virus.


Subject(s)
Measles virus/metabolism , Measles virus/pathogenicity , Measles/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Antigens, CD/genetics , Antigens, CD/metabolism , Autophagy , Carrier Proteins/genetics , Carrier Proteins/metabolism , Giant Cells/metabolism , Giant Cells/pathology , Giant Cells/virology , Golgi Matrix Proteins , HeLa Cells , Humans , Measles/genetics , Measles/pathology , Measles virus/genetics , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
4.
Autophagy ; 8(7): 1136-7, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22722598

ABSTRACT

Several intracellular pathogens have the ability to avoid or exploit the otherwise destructive process of autophagy. RNA viruses are constantly confronted with cellular autophagy, and several of them hijack autophagy during the infectious cycle to improve their own replication. Nevertheless, our knowledge of viral molecular strategies used to manipulate autophagy remains limited. Our study allowed the identification of molecular interactions between 44 autophagy-associated proteins and 83 viral proteins belonging to five different RNA virus families. This interactome revealed that the autophagy network machinery is highly targeted by RNA viruses. Interestingly, whereas some autophagy-associated proteins are targeted by only one RNA virus family, others are recurrent targets of several families. Among them, we found IRGM as the most targeted autophagy-associated protein. Downregulation of IRGM expression prevents autophagy induction by measles virus, HCV and HIV-1, and compromises viral replication. Our work combined interactomic and analytical approaches to identify potential pathogen virulence factors targeting autophagy.

5.
Autophagy ; 8(5): 767-79, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22622177

ABSTRACT

Initially described as a nonspecific degradation process induced upon starvation, autophagy is now known also to be involved in the degradation of specific ubiquitinated substrates such as mitochondria, bacteria and aggregated proteins, ensuring crucial functions in cell physiology and immunity. We report here that the deubiquitinating enzyme USP36 controls selective autophagy activation in Drosophila and in human cells. We show that dUsp36 loss of function autonomously inhibits cell growth while activating autophagy. Despite the phenotypic similarity, dUSP36 is not part of the TOR signaling pathway. Autophagy induced by dUsp36 loss of function depends on p62/SQSTM1, an adaptor for delivering cargo marked by polyubiquitin to autophagosomes. Consistent with p62 requirement, dUsp36 mutant cells display nuclear aggregates of ubiquitinated proteins, including Histone H2B, and cytoplasmic ubiquitinated proteins; the latter are eliminated by autophagy. Importantly, USP36 function in p62-dependent selective autophagy is conserved in human cells. Our work identifies a novel, crucial role for a deubiquitinating enzyme in selective autophagy.


Subject(s)
Autophagy , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Endopeptidases/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitinated Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Nucleus/metabolism , Cell Proliferation , DNA-Binding Proteins , Enzyme Activation , Fat Body/cytology , Fat Body/metabolism , Gene Silencing , HeLa Cells , Humans , Larva/cytology , Larva/enzymology , Larva/growth & development , Mutation/genetics , Nuclear Proteins/metabolism , Sequestosome-1 Protein , Signal Transduction
6.
PLoS Pathog ; 7(12): e1002422, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174682

ABSTRACT

Autophagy is a conserved degradative pathway used as a host defense mechanism against intracellular pathogens. However, several viruses can evade or subvert autophagy to insure their own replication. Nevertheless, the molecular details of viral interaction with autophagy remain largely unknown. We have determined the ability of 83 proteins of several families of RNA viruses (Paramyxoviridae, Flaviviridae, Orthomyxoviridae, Retroviridae and Togaviridae), to interact with 44 human autophagy-associated proteins using yeast two-hybrid and bioinformatic analysis. We found that the autophagy network is highly targeted by RNA viruses. Although central to autophagy, targeted proteins have also a high number of connections with proteins of other cellular functions. Interestingly, immunity-associated GTPase family M (IRGM), the most targeted protein, was found to interact with the autophagy-associated proteins ATG5, ATG10, MAP1CL3C and SH3GLB1. Strikingly, reduction of IRGM expression using small interfering RNA impairs both Measles virus (MeV), Hepatitis C virus (HCV) and human immunodeficiency virus-1 (HIV-1)-induced autophagy and viral particle production. Moreover we found that the expression of IRGM-interacting MeV-C, HCV-NS3 or HIV-NEF proteins per se is sufficient to induce autophagy, through an IRGM dependent pathway. Our work reveals an unexpected role of IRGM in virus-induced autophagy and suggests that several different families of RNA viruses may use common strategies to manipulate autophagy to improve viral infectivity.


Subject(s)
Autophagy/physiology , GTP-Binding Proteins/metabolism , RNA Virus Infections/metabolism , RNA Virus Infections/transmission , RNA Viruses/metabolism , Base Sequence , Blotting, Western , Computational Biology , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Microscopy, Confocal , Molecular Sequence Data , Open Reading Frames/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics , RNA, Small Interfering , Transfection , Two-Hybrid System Techniques , Viral Proteins/metabolism
7.
Med Sci (Paris) ; 27(1): 41-7, 2011 Jan.
Article in French | MEDLINE | ID: mdl-21299961

ABSTRACT

Autophagy is a highly conserved, self-degradative pathway for clearance and recycling of cytoplasmic contents. This ubiquitous cell intrinsic process can be used as a defence mechanism against intracellular pathogens. Indeed autophagy is increased upon pathogen detection, and experimental extinction in vitro and in vivo of this cellular process has been demonstrated as a crucial role to control intracellular pathogens. Co-evolution between host-cells and pathogens has selected numerous micoorganisms able to avoid or usurp autophagy to their own benefit. Understanding mechanisms underlying the anti-microbial properties of autophagy as well as those used by certain pathogens to escape this cellular process might be crucial to manipulate this cellular function in order to prevent or treat infectious diseases.


Subject(s)
Autophagy , Host-Pathogen Interactions/physiology , Animals , Bacterial Physiological Phenomena , Cells/microbiology , Cells/parasitology , Cells/virology , Eukaryotic Cells/physiology , HIV/physiology , Humans , Interferon Type I/physiology , Membrane Fusion , Models, Biological , Phagosomes/physiology , Proteasome Endopeptidase Complex/physiology , Receptors, Pattern Recognition/physiology , Selection, Genetic , Small Ubiquitin-Related Modifier Proteins/physiology , Unfolded Protein Response/physiology
8.
Autophagy ; 6(2): 299-300, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20087059

ABSTRACT

Autophagy is a degradative mechanism involved in cell protection against invading pathogens. Although the autophagic process is well characterized, the molecular pathways leading to its activation upon pathogen binding remain poorly understood. Our recent work demonstrates that the cell surface pathogen receptor CD46 induces autophagy upon pathogen recognition. The molecular pathway linking CD46 to the autophagosome machinery relies on the scaffold protein GOPC and on the autophagosome formation complex Beclin 1/VPS34. The CD46-dependent autophagy is critical to an early control of infection.


Subject(s)
Autophagy/immunology , Membrane Cofactor Protein/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Host-Pathogen Interactions , Mice , Phagosomes/metabolism
9.
Cell Host Microbe ; 6(4): 354-66, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19837375

ABSTRACT

Autophagy is a highly regulated self-degradative mechanism required at a basal level for intracellular clearance and recycling of cytoplasmic contents. Upon intracellular pathogen invasion, autophagy can be induced as an innate immune mechanism to control infection. Nevertheless, pathogens have developed strategies to avoid or hijack autophagy for their own benefit. The molecular pathways inducing autophagy in response to infection remain poorly documented. We report here that the engagement of CD46, a ubiquitous human surface receptor able to bind several different pathogens, is sufficient to induce autophagy. CD46-Cyt-1, one of the two C-terminal splice variants of CD46, is linked to the autophagosome formation complex VPS34/Beclin1 via its interaction with the scaffold protein GOPC. Measles virus and group A Streptococcus, two CD46-binding pathogens, induce autophagy through a CD46-Cyt-1/GOPC pathway. Thus, upon microorganism recognition, a cell surface pathogen receptor can directly trigger autophagy, a critical step to control infection.


Subject(s)
Autophagy , Measles virus/immunology , Membrane Cofactor Protein/immunology , Streptococcus pyogenes/immunology , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Carrier Proteins/metabolism , Golgi Matrix Proteins , Humans , Membrane Proteins/metabolism , Membrane Transport Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Interaction Mapping
10.
Blood ; 113(7): 1598-607, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19036700

ABSTRACT

Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). These end-products are responsible for much of the biologic activity of HO-1, including anti-inflammatory, antiapo-ptotic, antiproliferative, and antioxidant effects. We have identified an additional cytoprotective action, the regulation of complement activation, mediated via induction of decay-accelerating factor (DAF). Pharmacologic inhibition or short-interfering RNA (siRNA) depletion of HO-1 prevented induction of DAF expression in human endothelial cells. In contrast, HO-1 agonists hemin and cobalt protoporphyrin IX significantly increased DAF protein expression, reflecting an increase in transcription and steady-state mRNA. Adenoviral-mediated overexpression of HO-1 increased DAF expression, enhancing protection against C3 deposition and complement-mediated lysis, and this was reversed by DAF inhibitory monoclonal antibody (mAb) 1H4. Likewise, bilirubin, Fe chelation, and overexpression of heavy-chain ferritin all induced DAF expression in endothelial cells (EC). Analysis of cardiac endothelial cells isolated from Hmox1(-/-) mice revealed a 60% reduction in DAF expression compared with Hmox1(+/+) EC, and Hmox1(-/-) cells showed enhanced sensitivity to complement. We propose that modulation of complement activation through induction of DAF represents an important component of the cytoprotective effects of HO-1 against vascular injury, such as that associated with posttransplant vasculopathy, allograft rejection, and ischemia reperfusion.


Subject(s)
Bilirubin/metabolism , CD55 Antigens/metabolism , Complement System Proteins/immunology , Endothelial Cells/immunology , Ferritins/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Animals , CD55 Antigens/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation/physiology , Heme Oxygenase-1/genetics , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Protoporphyrins/pharmacology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Umbilical Veins/cytology , Vascular Diseases/immunology , Vascular Diseases/metabolism
11.
J Immunol ; 179(11): 7840-51, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18025230

ABSTRACT

Heme oxygenase-1 (HO-1; encoded by the Hmox1 gene) catalyzes the degradation of free heme into biliverdin, via a reaction that releases iron (Fe) and carbon monoxide. We report that HO-1 down-regulates the proinflammatory phenotype associated with endothelial cell (EC) activation by reducing intracellular nonprotein-bound Fe (labile Fe). EC isolated from Hmox1(-/-) mice have higher levels of intracellular labile Fe and reactive oxygen species (ROS) as compared with EC isolated from Hmox1(+/+) mice. Basal and TNF-induced expression of VCAM-1, ICAM-1, and E-selectin were increased in Hmox1(-/-) vs Hmox1(+/+) EC, an effect reversed by Fe chelation using deferoxamine mesylate (DFO). Fe chelation inhibits TNF-driven transcription of Vcam-1, Icam-1, and E-selectin, as assessed using luciferase reporter assays. This effect is associated with inhibition of the transcription factor NF-kappaB via a mechanism that is not associated with the inhibition of IkappaBalpha phosphorylation/degradation or NF-kappaB (i.e., RelA) nuclear translocation, although it affects very modestly NF-kappaB binding to DNA kappaB consensus sequences in the Vcam-1 and E-selectin promoters. HO-1 inhibits NF-kappaB (i.e., RelA) phosphorylation at Ser(276), a phosphoacceptor that is critical to sustain TNF-driven NF-kappaB activity in EC. This effect was mimicked by Fe chelation as well as by antioxidants (N-acetylcysteine). In conclusion, we demonstrate a novel mechanism via which HO-1 down-modulates the proinflammatory phenotype of activated EC, i.e., the inhibition of RelA phosphorylation at Ser(276).


Subject(s)
Cell Adhesion Molecules/immunology , Endothelial Cells/immunology , Gene Expression Regulation/immunology , Heme Oxygenase-1/immunology , Serine/immunology , Transcription Factor RelA/metabolism , Animals , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/genetics , Cells, Cultured , Down-Regulation/immunology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Ferric Compounds/immunology , Gene Expression Regulation/drug effects , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/pharmacology , Inflammation , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Phosphorylation/drug effects , Serine/drug effects , Transcription Factor RelA/drug effects , Transcription, Genetic/drug effects , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/drug effects
12.
Nat Med ; 13(6): 703-10, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496899

ABSTRACT

Cerebral malaria claims more than 1 million lives per year. We report that heme oxygenase-1 (HO-1, encoded by Hmox1) prevents the development of experimental cerebral malaria (ECM). BALB/c mice infected with Plasmodium berghei ANKA upregulated HO-1 expression and activity and did not develop ECM. Deletion of Hmox1 and inhibition of HO activity increased ECM incidence to 83% and 78%, respectively. HO-1 upregulation was lower in infected C57BL/6 compared to BALB/c mice, and all infected C57BL/6 mice developed ECM (100% incidence). Pharmacological induction of HO-1 and exposure to the end-product of HO-1 activity, carbon monoxide (CO), reduced ECM incidence in C57BL/6 mice to 10% and 0%, respectively. Whereas neither HO-1 nor CO affected parasitemia, both prevented blood-brain barrier (BBB) disruption, brain microvasculature congestion and neuroinflammation, including CD8(+) T-cell brain sequestration. These effects were mediated by the binding of CO to hemoglobin, preventing hemoglobin oxidation and the generation of free heme, a molecule that triggers ECM pathogenesis.


Subject(s)
Carbon Monoxide/physiology , Heme Oxygenase-1/physiology , Heme/metabolism , Malaria, Cerebral/enzymology , Animals , Disease Models, Animal , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Plasmodium berghei
13.
J Immunol ; 177(3): 1894-903, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16849502

ABSTRACT

Heme oxygenase-1 (HO-1) protects endothelial cells (EC) from undergoing apoptosis. This effect is mimicked by CO, generated via the catabolism of heme by HO-1. The antiapoptotic effect of CO in EC was abrogated when activation of the p38alpha and p38beta MAPKs was inhibited by the pyridinyl imidazole SB202190. Using small interfering RNA, p38beta was found to be cytoprotective in EC, whereas p38alpha was not. When overexpressed in EC, HO-1 targeted specifically the p38alpha but not the p38beta MAPK isoform for degradation by the 26S proteasome, an effect reversed by the 26S proteasome inhibitors MG-132 or lactacystin. Inhibition of p38alpha expression was also observed when HO-1 was induced physiologically by iron protoporphyrin IX (hemin). Inhibition of p38alpha no longer occurred when HO activity was inhibited by tin protoporphyrin IX, suggesting that p38alpha degradation was mediated by an end product of heme catabolism. Exogenous CO inhibited p38alpha expression in EC, suggesting that CO is the end product that mediates this effect. The antiapoptotic effect of HO-1 was impaired when p38alpha expression was restored ectopically or when its degradation by the 26S proteasome was inhibited by MG-132. Furthermore, the antiapoptotic effect of HO-1 was lost when p38beta expression was targeted by a specific p38beta small interfering RNA. In conclusion, the antiapoptotic effect of HO-1 in EC is dependent on the degradation of p38alpha by the 26S proteasome and on the expression of p38beta.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Endothelial Cells/enzymology , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Heme Oxygenase-1/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Carbon Monoxide/physiology , Cattle , Cell Line , Cytoprotection/physiology , Endothelial Cells/cytology , Enzyme Activation/physiology , HeLa Cells , Heme Oxygenase-1/antagonists & inhibitors , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Mitogen-Activated Protein Kinase 11/antagonists & inhibitors , Mitogen-Activated Protein Kinase 11/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/biosynthesis , Mitogen-Activated Protein Kinase 14/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
14.
J Immunol ; 172(6): 3553-63, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15004156

ABSTRACT

Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Heme-derived Fe2+ induces the expression of the iron-sequestering protein ferritin and activates the ATPase Fe2+-secreting pump, which decrease intracellular free Fe2+ content. Based on the antioxidant effect of bilirubin and that of decreased free cellular Fe2+, we questioned whether HO-1 would modulate the expression of proinflammatory genes associated with endothelial cell (EC) activation. We tested this hypothesis specifically for the genes E-selectin (CD62), ICAM-1 (CD54), and VCAM-1 (CD106). We found that HO-1 overexpression in EC inhibited TNF-alpha-mediated E-selectin and VCAM-1, but not ICAM-1 expression, as tested at the RNA and protein level. Heme-driven HO-1 expression had similar effects to those of overexpressed HO-1. In addition, HO-1 inhibited the activation of NF-kappaB, a transcription factor required for TNF-alpha-mediated up-regulation of these genes in EC. Bilirubin and/or Fe2+ chelation mimicked the effects of HO-1, whereas biliverdin or carbon monoxide did not. In conclusion, HO-1 inhibits the expression of proinflammatory genes associated with EC activation via a mechanism that is associated with the inhibition of NF-kappaB activation. This effect of HO-1 is mediated by bilirubin and/or by a decrease of free intracellular Fe2+ but probably not by biliverdin or carbon monoxide.


Subject(s)
Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Heme Oxygenase (Decyclizing)/physiology , Adenoviridae/genetics , Animals , Bilirubin/pharmacology , Carbon Monoxide/pharmacology , Cattle , Cell Adhesion Molecules , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/immunology , E-Selectin/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Heme/pharmacology , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Iron Chelating Agents/pharmacology , MAP Kinase Signaling System/immunology , Membrane Proteins , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Swine , Transduction, Genetic , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/biosynthesis , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...