Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Commun Signal ; 9: 16, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21635783

ABSTRACT

BACKGROUND: Cytoplasmic viral double-stranded RNA (dsRNA) is detected by a class of ubiquitous cytoplasmic RNA helicases, retinoic acid inducible gene-I (RIG-I) and melanoma differentiation antigen-5 (MDA5), which initiate a signaling cascade via their common adaptor called interferon-ß (IFN-ß) promoter stimulator-1 (IPS-1). This leads to the production of proinflammatory and antiviral cytokines, the type I Interferons, via mainly nuclear factor kappa B (NF-κB) and interferon response factor-3 (IRF3) transcription factors. Fas-associated death domain (FADD) protein, receptor-interacting protein (RIP1), caspase-8 and tumor necrosis factor receptor (TNFR)-associated death domain (TRADD) protein, all traditionally associated with death receptor signaling, are also involved in RIG-I/MDA5 signaling pathway. We previously showed that FLIP (Flice-like inhibitory protein), also designated as cflar (CASP8 and FADD-like apoptosis regulator), negatively regulates lipopolysaccharide (LPS)-induced toll-like receptor 4 (TLR4) signaling in endothelial cells and mouse embryonic fibroblasts (MEFs) and protected against TLR4-mediated apoptosis. RESULTS: In this study, we investigated the role of FLIP in cellular response to cytoplasmic polyinosinic:polycytidylic acid, poly(I:C), a synthetic analog of dsRNA. Consistent with the previously described role of FADD in RIG-I/MDA5-mediated apoptosis, we found that FLIP-/- MEFs were more susceptible to killing by cytoplasmic poly(I:C). However, FLIP-/- MEFs also exhibited markedly increased expression of NF-κB-and IRF3- dependent genes in response to cytoplasmic poly(I:C). Importantly, reconstitution of FLIP in FLIP-/-MEFs reversed the hyper-activation of IRF3- and NF-κB-mediated gene expression. Further, we found that caspase-8 catalytic activity was not required for cytoplasmic poly(I:C)-mediated NF-κB and IRF3 signaling. CONCLUSIONS: These results provide evidence for a crucial dual role for FLIP in antiviral responses to cytoplasmic dsRNA: it protects from cytoplasmic dsRNA-mediated cell death while down-regulating IRF3-and NF-κB-mediated gene expression. Since the pathogenesis of several viral infections involves a heightened and dysregulated cytokine response, a possible therapy could involve modulating FLIP levels.

2.
PLoS One ; 6(2): e14729, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21390214

ABSTRACT

BACKGROUND: Severe sepsis and septic shock are major causes of morbidity and mortality worldwide. In experimental sepsis there is prominent apoptosis of various cell types, and genetic manipulation of death and survival pathways has been shown to modulate organ injury and survival. METHODOLOGY/PRINCIPAL FINDINGS: We investigated the effect of extracellular administration of two anti-apoptotic members of the BCL2 (B-cell lymphoma 2) family of intracellular regulators of cell death in a murine model of sepsis induced by cecal ligation and puncture (CLP). We show that intraperitoneal injection of picomole range doses of recombinant human (rh) BCL2 or rhBCL2A1 protein markedly improved survival as assessed by surrogate markers of death. Treatment with rhBCL2 or rhBCL2A1 protein significantly reduced the number of apoptotic cells in the intestine and heart following CLP, and this was accompanied by increased expression of endogenous mouse BCL2 protein. Further, mice treated with rhBCL2A1 protein showed an increase in the total number of neutrophils in the peritoneum following CLP with reduced neutrophil apoptosis. Finally, although neither BCL2 nor BCL2A1 are a direct TLR2 ligand, TLR2-null mice were not protected by rhBCL2A1 protein, indicating that TLR2 signaling was required for the protective activity of extracellularly adminsitered BCL2A1 protein in vivo. CONCLUSIONS/SIGNIFICANCE: Treatment with rhBCL2A1 or rhBCL2 protein protects mice from sepsis by reducing apoptosis in multiple target tissues, demonstrating an unexpected, potent activity of extracellularly administered BCL2 BH4-domain proteins.


Subject(s)
Apoptosis/drug effects , Proto-Oncogene Proteins c-bcl-2/pharmacology , Sepsis/mortality , Animals , Cecum/pathology , Cecum/surgery , Disease Models, Animal , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Extracellular Space/drug effects , Humans , Ligation , Mice , Minor Histocompatibility Antigens , Proto-Oncogene Proteins c-bcl-2/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Sepsis/drug therapy , Sepsis/pathology , Wounds, Penetrating/pathology
3.
Biochem Pharmacol ; 74(7): 1039-49, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17681284

ABSTRACT

Apoptosis may be regulated by oxidants such as peroxynitrite (ONOO(-)). The tumour suppressor, p53, has been reported to play a crucial role in apoptosis induced by oxidants, therefore we assessed the ability of a ONOO(-) donor, GEA 3162, to activate caspases and induce mitochondrial permeability in a p53-deficient murine bone marrow cell line, Jaws II. Furthermore, these cells were stably transfected with Bcl-2, in order to investigate the impact of this survival protein on ONOO(-)-induced apoptosis. GEA 3162 activated caspases and induced loss of mitochondrial membrane potential in Jaws II cells. In particular, caspases 3 and 2 were activated, alongside minor activation of caspases 8 and 9, and apoptosis was partially dependent upon p38 MAP kinase activation, with little or no role for JNK. Overexpression of Bcl-2 abolished activation of all caspases and reduced the change in mitochondrial membrane potential. Thus, we have demonstrated that the ONOO(-) donor, GEA 3162, induces apoptosis in Jaws II murine myeloid cells despite lacking functional p53, via a pathway that principally involves caspases 2 and 3 and mitochondrial changes. This is blocked by overexpression of Bcl-2 via a mechanism that does not appear to merely reflect stabilisation of the mitochondrial membrane.


Subject(s)
Apoptosis/drug effects , Bone Marrow Cells/drug effects , Nitric Oxide Donors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Triazoles/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Caspases/metabolism , Cell Line , Cell Survival , Dose-Response Relationship, Drug , Enzyme Activation , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Mol Cancer Res ; 2(11): 620-31, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15561778

ABSTRACT

The t(14;18)(q32;q21), resulting in deregulated expression of B-cell-leukemia/lymphoma-2 (Bcl-2), represents the genetic hallmark in human follicular lymphomas. Substantial evidence supports the hypothesis that the t(14;18) and Bcl-2 overexpression are necessary but not solely responsible for neoplastic transformation and require cooperating genetic derangements for neoplastic transformation to occur. To investigate genes that cooperate with Bcl-2 to influence cellular signaling pathways important for neoplastic transformation, we used oligonucleotide microarrays to determine differential gene expression patterns in CD19+ B cells isolated from Emu-Bcl-2 transgenic mice and wild-type littermate control mice. Fifty-seven genes were induced and 94 genes were repressed by > or =2-fold in Emu-Bcl-2 transgenic mice (P < 0.05). The suppressor of cytokine signaling-3 (SOCS3) gene was found to be overexpressed 5-fold in B cells from Emu-Bcl-2 transgenic mice. Overexpression of Bcl-2 in both mouse embryo fibroblast-1 and hematopoietic cell lines resulted in induction of SOCS3 protein, suggesting a Bcl-2-associated mechanism underlying SOCS3 induction. Immunohistochemistry with SOCS3 antisera on tissue from a cohort of patients with de novo follicular lymphoma revealed marked overexpression of SOCS3 protein that, within the follicular center cell region, was limited to neoplastic follicular lymphoma cells and colocalized with Bcl-2 expression in 9 of 12 de novo follicular lymphoma cases examined. In contrast, SOCS3 protein expression was not detected in the follicular center cell region of benign hyperplastic tonsil tissue. These data suggest that Bcl-2 overexpression leads to the induction of activated signal transducer and activator of transcription 3 (STAT3) and to the induction of SOCS3, which may contribute to the pathogenesis of follicular lymphoma.


Subject(s)
B-Lymphocytes/metabolism , Biomarkers, Tumor/metabolism , Lymphoma, Follicular/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Up-Regulation/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Humans , Lymphoma, Follicular/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Palatine Tonsil/metabolism , Palatine Tonsil/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcriptional Activation/genetics
5.
Microb Pathog ; 35(2): 87-93, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12901848

ABSTRACT

Shiga toxin (Stx) has been implicated in the pathogenesis of several human and animal disease states. A key host target of Stx is the endothelial cell. Stx induces endothelial cell apoptosis through a mechanism that remains unknown. In the present report, we demonstrate that Stx-1 and Stx-2 inhibit endothelial cell expression of the anti-apoptotic Bcl-2 family member, Mcl-1. Decreased expression of Mcl-1 preceded the onset of Stx-induced apoptosis. Further, Stx-1-induced decrements in Mcl-1 expression correlated in a dose-dependent manner with sensitization to Stx-1-induced apoptosis. Finally, inhibition of Mcl-1 degradation with the proteasome inhibitor, lactacystin, protected against Stx-1-induced apoptosis. These combined data suggest a role for Mcl-1 in protecting endothelial cells against Stx-1-induced apoptosis.


Subject(s)
Apoptosis , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2 , Shiga Toxin 1/toxicity , Shiga Toxin 2/toxicity , Caspase 3 , Caspases/metabolism , Cell Line , Histones/metabolism , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/physiology
6.
Infect Immun ; 71(8): 4414-20, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12874320

ABSTRACT

Many of the proinflammatory effects of gram-negative bacteria are elicited by the interaction of bacterial lipopolysaccharide (LPS) with Toll-like receptor 4 (TLR4) expressed on host cells. TLR4 signaling leads to activation of NF-kappa B and transcription of many genes involved in the inflammatory response. In this study, we examined the signaling pathways involved in NF-kappa B activation by TLR4 signaling in human microvascular endothelial cells. Akt is a major downstream target of phosphoinositide 3 kinase (PI3-kinase), and PI3-kinase activation is necessary and sufficient for Akt phosphorylation. Consequently, Akt kinase activation was used as a measure of PI3-kinase activity. In a stable transfection system, dominant-negative mutants of myeloid differentiation factor 88 (MyD88) and interleukin-1 (IL-1) receptor-associated kinase 1 (IRAK-1) (MyD88-TIR and IRAK-DD, respectively) blocked Akt kinase activity in response to LPS and IL-1 beta. A dominant-negative mutant (Mal-P/H) of MyD88 adapter-like protein (Mal), a protein with homology to MyD88, failed to inhibit LPS- or IL-1 beta-induced Akt activity. Moreover, a dominant-negative mutant of p85 (p85-DN) inhibited the NF-kappa B luciferase activity, IL-6 production, and I kappa B alpha degradation elicited by LPS and IL-1 beta but not that stimulated by tumor necrosis factor alpha. The dominant-negative mutant of Akt partially inhibited the NF-kappa B luciferase activity evoked by LPS and IL-1 beta. However, expression of a constitutively activated Akt failed to induce NF-kappa B luciferase activity. These findings indicate that TLR4- and IL-1R-induced PI3-kinase activity is mediated by the adapter proteins MyD88 and IRAK-1 but not Mal. Further, these studies suggest that PI3-kinase is an important mediator of LPS and IL-1 beta signaling leading to NF-kappa B activation in endothelial cells and that Akt is necessary but not sufficient for NF-kappa B activation by TLR4.


Subject(s)
Endothelium, Vascular/metabolism , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Receptors, Cell Surface/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Gram-Negative Bacteria/pathogenicity , Humans , Interleukin-1/pharmacology , Interleukin-1 Receptor-Associated Kinases , Interleukin-6/biosynthesis , Lipopolysaccharides/toxicity , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/pharmacology
7.
J Biol Chem ; 277(18): 15309-16, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11839765

ABSTRACT

Geranylgeranylation of RhoA small G-protein is essential for its localization to cell membranes and for its biological functions. Many RhoA effects are mediated by its downstream effector RhoA kinase. The role of protein geranylgeranylation and the RhoA pathway in the regulation of endothelial cell survival has not been elucidated. The hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitor lovastatin depletes cellular pools of geranylgeranyl pyrophosphate and farnesol pyrophosphate and thereby inhibits both geranylgeranylation and farnesylation. Human umbilical vein endothelial cells (HUVECs) were exposed to lovastatin (3 microm-30 microm) for 48 h, and cell death was quantitatively determined by cytoplasmic histone-associated DNA fragments as well as caspase-3 activity. The assays showed that lovastatin caused a dose-dependent endothelial cell death. The addition of geranylgeraniol, which restores geranylgeranylation, rescued HUVEC from apoptosis. The geranylgeranyltransferase inhibitor GGTI-298, but not the farnesyltransferase inhibitor FTI-277, induced apoptosis in HUVEC. Cell death was also induced by a blockade of RhoA function by exoenzyme C3. In addition, treatment of HUVEC with the RhoA kinase inhibitors Y-27632 and HA-1077 caused dose-dependent cell death. Y-27632 did not inhibit other well known survival pathways, such as NF-kappa B, ERK, and phosphatidylinositol 3-kinase/Akt. However, there was an increase in p53 protein level concomitant with Y-27632-induced cell death. Unlike the apoptosis induced by TNF-alpha, which occurs only with inhibition of new protein synthesis, apoptosis induced by inhibitors of HMG-CoA reductase, geranylgeranyltransferase, or RhoA kinase was blocked by cycloheximide. Our data indicate that inhibition of protein geranylgeranylation and RhoA pathways induce apoptosis in HUVEC and that induction of p53 or other proapoptotic proteins is required for this process.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Diterpenes/pharmacology , Endothelium, Vascular/cytology , Lovastatin/pharmacology , Methionine/analogs & derivatives , Protein Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Apoptosis/drug effects , Benzamides/pharmacology , Caspase 3 , Cell Death/drug effects , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins , Methionine/pharmacology , Polyisoprenyl Phosphates , Protein Prenylation , Umbilical Veins , rho-Associated Kinases
8.
J Clin Invest ; 109(3): 419-25, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11828002

ABSTRACT

Activation of NF-kappa B by bacterial LPS promotes the upregulation of proinflammatory cytokines that contribute to the pathogenesis of Gram-negative septic shock. LPS activation of NF-kappa B is dependent upon the interaction of two death domain-containing (DD-containing) proteins, MyD88 and IL-1 receptor-associated kinase IRAK. Another DD-containing protein, Fas-associated death domain (FADD), also binds MyD88 through respective DD-DD interactions. Although FADD has been classically described as a proapoptotic signaling molecule, several reports have implicated a role for FADD in mediating NF-kappa B activation. In the present report, we investigated whether FADD could mediate LPS activation of NF-kappa B. Overexpression of FADD blocked LPS-induced NF-kappa B activation, whereas absence of FADD enhanced activation of NF-kappa B by LPS. Further, LPS-induced expression of two NF-kappa B-dependent gene products, IL-6 and KC, was enhanced in FADD(-/-) mouse embryo fibroblasts (MEFs) compared with wild-type. This increase in NF-kappa B activity correlated with enhanced I kappa B degradation. FADD(-/-) MEFs were also resistant to NF-kappa B activation induced by IL-1 beta. Finally, reconstitution of full-length FADD in the FADD(-/-) MEFs completely reversed the enhanced activation of NF-kappa B elicited by either LPS or IL-1 beta. Together, these data indicate that FADD negatively regulates LPS- and IL-1 beta-induced NF-kappa B activation and that this regulation occurs upstream of I kappa B degradation.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Chemokines, CXC , I-kappa B Proteins , Intercellular Signaling Peptides and Proteins , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Chemokine CXCL1 , Chemotactic Factors/biosynthesis , DNA-Binding Proteins/metabolism , Fas-Associated Death Domain Protein , Gene Expression , Growth Substances/biosynthesis , Humans , Interleukin-6/biosynthesis , Mice , Mice, Knockout , NF-KappaB Inhibitor alpha , Transfection
9.
J Biol Chem ; 277(10): 8048-53, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11777917

ABSTRACT

The vascular endothelium is a key target of circulating bacterial lipopolysaccharide (LPS). LPS elicits a wide array of endothelial responses, including the up-regulation of cytokines, adhesion molecules, and tissue factor, many of which are dependent on NF-kappa B activation. In addition, LPS has been demonstrated to induce endothelial apoptosis both in vitro and in vivo. Although the mechanism by which LPS activates NF-kappa B has been well elucidated, the signaling pathway(s) involved in LPS-induced apoptosis remains unknown. Using a variety of dominant negative constructs, we have identified a role for MyD88 and interleukin-1 receptor-associated kinase-1 (IRAK-1) in mediating LPS pro-apoptotic signaling in human endothelial cells. We also demonstrate that LPS-induced endothelial NF-kappa B activation and apoptosis occur independent of one another. Together, these data suggest that the proximal signaling molecules involved in LPS-induced NF-kappa B activation have a requisite involvement in LPS-induced apoptosis and that the pathways leading to NF-kappa B activation and apoptosis diverge downstream of IRAK-1.


Subject(s)
Apoptosis , Lipopolysaccharides/metabolism , Protein Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Adenoviridae/metabolism , Antigens, Differentiation/metabolism , Caspases/metabolism , Cell Adhesion , Cloning, Molecular , DNA, Complementary/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Escherichia coli/metabolism , Humans , Immunoblotting , Interleukin-1/metabolism , Interleukin-1 Receptor-Associated Kinases , Luciferases/metabolism , Myeloid Differentiation Factor 88 , NF-kappa B/metabolism , Polymyxin B/pharmacology , Protein Binding , Receptors, Immunologic/metabolism , Time Factors , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL