Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 71(2): 331-48, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23760205

ABSTRACT

Programmed necrosis is important in many (patho)physiological settings. For specific therapeutic intervention, however, a better knowledge is required whether necrosis occurs through one single "core program" or through several independent pathways. Previously, the poly(ADP-ribose) polymerase (PARP) pathway has been suggested as a crucial element of tumor necrosis factor (TNF)-mediated necroptosis. Here, we show that TNF-induced necroptosis and the PARP pathway represent distinct and independent routes to programmed necrosis. First, DNA-alkylating agents such as 1-methyl-3-nitro-1-nitrosoguanidine (MNNG) or methyl methanesulfonate rapidly activate the PARP pathway, whereas this is a late and secondary event in TNF-induced necroptosis. Second, inhibition of the PARP pathway does not protect against TNF-induced necroptosis, e.g., the PARP-1 inhibitor 3-AB prevented MNNG- but not TNF-induced adenosine-5'-triposphate depletion, translocation of apoptosis-inducing factor, and necrosis. Likewise, olaparib, a more potent and selective PARP-1 inhibitor failed to block TNF-induced necroptosis, identical to knockdown/knockout of PARP-1, pharmacologic and genetic interference with c-Jun N-terminal kinases and calpain/cathepsin proteases as further components of the PARP pathway. Third, interruption of TNF-induced necroptosis by interference with ceramide generation, RIP1 or RIP3 function or by the radical scavenger butylated hydroxyanisole did not prevent programmed necrosis through the PARP pathway. In summary, our results suggest that the currently established role of the PARP pathway in TNF-induced necroptosis needs to be revised, with consequences for the design of future therapeutic strategies.


Subject(s)
Apoptosis/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antineoplastic Agents, Alkylating/pharmacology , Benzamides/pharmacology , Calpain/metabolism , Cathepsins/metabolism , Cell Line , Ceramides/metabolism , Free Radical Scavengers/pharmacology , Guanidines/pharmacology , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , MCF-7 Cells , Methyl Methanesulfonate/pharmacology , Mice , Necrosis , Nuclear Pore Complex Proteins/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
2.
Cell Commun Signal ; 11: 76, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24090154

ABSTRACT

BACKGROUND: In apoptosis, proteolysis by caspases is the primary mechanism for both initiation and execution of programmed cell death (PCD). In contrast, the impact of proteolysis on the regulation and execution of caspase-independent forms of PCD (programmed necrosis, necroptosis) is only marginally understood. Likewise, the identity of the involved proteases has remained largely obscure. Here, we have investigated the impact of proteases in TNF-induced necroptosis. RESULTS: The serine protease inhibitor TPKC protected from TNF-induced necroptosis in multiple murine and human cells systems whereas inhibitors of metalloproteinases or calpain/cysteine and cathepsin proteases had no effect. A screen for proteins labeled by a fluorescent TPCK derivative in necroptotic cells identified HtrA2/Omi (a serine protease previously implicated in PCD) as a promising candidate. Demonstrating its functional impact, pharmacological inhibition or genetic deletion of HtrA2/Omi protected from TNF-induced necroptosis. Unlike in apoptosis, HtrA2/Omi did not cleave another protease, ubiquitin C-terminal hydrolase (UCH-L1) during TNF-induced necroptosis, but rather induced monoubiquitination indicative for UCH-L1 activation. Correspondingly, pharmacologic or RNA interference-mediated inhibition of UCH-L1 protected from TNF-induced necroptosis. We found that UCH-L1 is a mediator of caspase-independent, non-apoptotic cell death also in diseased kidney podocytes by measuring cleavage of the protein PARP-1, caspase activity, cell death and cell morphology. Indicating a role of TNF in this process, podocytes with stably downregulated UCH-L1 proved resistant to TNF-induced necroptosis. CONCLUSIONS: The proteases HtrA2/Omi and UCH-L1 represent two key components of TNF-induced necroptosis, validating the relevance of proteolysis not only for apoptosis, but also for caspase-independent PCD. Since UCH-L1 clearly contributes to the non-apoptotic death of podocytes, interference with the necroptotic properties of HtrA2/Omi and UCH-L1 may prove beneficial for the treatment of patients, e.g. in kidney failure.


Subject(s)
Apoptosis/physiology , Mitochondrial Proteins/metabolism , Serine Endopeptidases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Cells, Cultured , HT29 Cells , High-Temperature Requirement A Serine Peptidase 2 , Humans , Jurkat Cells , Mice , NIH 3T3 Cells , Podocytes/metabolism , Rats , Rats, Wistar
3.
Blood ; 118(3): 598-608, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21613257

ABSTRACT

The regulation of cellular survival and apoptosis is of critical importance for the immune system to maintain immune homeostasis and to establish tolerance. Here, we demonstrate that the immune specific cell surface molecule Toso exhibits antiapoptotic effects on death receptor signaling by a novel regulatory mechanism involving the adaptor kinase RIP1. The antiapoptotic function of Toso depends on RIP1 ubiquitination and involves the recruitment of the death adaptor FADD to a Toso/RIP1 protein complex. In response to CD95L and TNFα, Toso promotes the activation of MAPK and NF-κB signaling pathways. Because of this relative augmentation of survival versus apoptotic signals, Toso raises the threshold for death receptor-mediated apoptosis. Our analysis of Toso-deficient mice revealed that Toso is essential for TNFα-mediated liver damage. Furthermore, the antiapoptotic function of Toso could be blocked by a Toso-specific monoclonal antibody, opening up new therapeutic prospects for the treatment of immune disorders and hematologic malignancies.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/immunology , Membrane Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/immunology , Ubiquitination/physiology , Animals , Antibodies, Monoclonal/immunology , Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Survival/immunology , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Gene Knockdown Techniques , Humans , Immune Tolerance/immunology , Jurkat Cells , Liver Diseases/immunology , Liver Diseases/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nuclear Pore Complex Proteins/genetics , RNA-Binding Proteins/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
4.
Proc Natl Acad Sci U S A ; 107(3): 1112-7, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20080539

ABSTRACT

The phospholipase neutral sphingomyelinase (N-SMase) has been recognized as a major mediator of processes such as inflammation, development and growth, differentiation and death of cells, as well as in diseases such as Alzheimer's, atherosclerosis, heart failure, ischemia/reperfusion damage, or combined pituitary hormone deficiency. Although activation of N-SMase by the proinflammatory cytokine TNF was described almost two decades ago, the underlying signaling pathway is unresolved. Here, we identify the Polycomb group protein EED (embryonic ectodermal development) as an interaction partner of nSMase2. In yeast, the N terminus of EED binds to the catalytic domain of nSMase2 as well as to RACK1, a protein that modulates the activation of nSMase2 by TNF in concert with the TNF receptor 1 (TNF-R1)-associated protein FAN. In mammalian cells, TNF causes endogenous EED to translocate from the nucleus and to colocalize and physically interact with both endogenous nSMase2 and RACK1. As a consequence, EED and nSMase2 are recruited to the TNF-R1.FAN.RACK1-complex in a timeframe concurrent with activation of nSMase2. After knockdown of EED by RNA interference, the TNF-dependent activation of nSMase2 is completely abrogated, identifying EED as a protein that both physically and functionally couples TNF-R1 to nSMase2, and which therefore represents the "missing link" that completes one of the last unresolved signaling pathways of TNF-R1.


Subject(s)
Receptors, Tumor Necrosis Factor/metabolism , Repressor Proteins/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Enzyme Activation , HeLa Cells , Humans , Polycomb Repressive Complex 2
5.
Exp Cell Res ; 313(12): 2703-18, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17512928

ABSTRACT

FAN (factor associated with neutral sphingomyelinase [N-SMase] activation) exhibits striking structural homologies to Lyst (lysosomal trafficking regulator), a BEACH protein whose inactivation causes formation of giant lysosomes/Chediak-Higashi syndrome. Here, we show that cells lacking FAN show a statistically significant increase in lysosome size (although less pronounced as Lyst), pointing to previously unrecognized functions of FAN in regulation of the lysosomal compartment. Since FAN regulates activation of N-SMase in complex with receptor for activated C-kinase (RACK)1, a scaffolding protein that recruits and stabilizes activated protein kinase C (PKC) isotypes at cellular membranes, and since an abnormal (calpain-mediated) downregulation/membrane recruitment of PKC has been linked to the defects observed in Lyst-deficient cells, we assessed whether PKC is also of relevance in FAN signaling. Our results demonstrate that activation of PKC is not required for regulation of N-SMase by FAN/RACK1. Conversely, activation of PKC and recruitment/stabilization by RACK1 occurs uniformly in the presence or absence of FAN (and equally, Lyst). Furthermore, regulation of lysosome size by FAN is not coupled to an abnormal downregulation/membrane recruitment of PKC by calpain. Identical results were obtained for Lyst, questioning the previously reported relevance of PKC for formation of giant lysosomes and in Chediak-Higashi syndrome. In summary, FAN mediates activation of N-SMase as well as regulation of lysosome size by signaling pathways that operate independent from activation/membrane recruitment of PKC.


Subject(s)
Cell Membrane/enzymology , Down-Regulation/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/enzymology , Protein Kinase C/metabolism , Repetitive Sequences, Amino Acid , Animals , Cell Membrane/drug effects , Down-Regulation/drug effects , Enzyme Activation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Isoenzymes/metabolism , Lysosomes/drug effects , Mice , Neuropeptides/deficiency , Neuropeptides/metabolism , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proteins/metabolism , Receptors for Activated C Kinase , Tumor Necrosis Factor-alpha/pharmacology , Vesicular Transport Proteins
6.
Exp Cell Res ; 312(19): 3808-21, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17026999

ABSTRACT

Death receptors such as the 55 kDa tumor necrosis factor (TNF) receptor (TNF-R55) or Fas can initiate both apoptotic (caspase-dependent) and caspase-independent routes to programmed cell death (PCD). Here, we demonstrate for the first time that the single murine receptor for (TNF)-related apoptosis-inducing ligand (mTRAIL-R2) can induce a caspase-independent form of PCD with necrosis-like features in addition to apoptosis. Analysis of morphological and cellular features of caspase-independent PCD in response to TRAIL and TNF suggests that mTRAIL-R2 and TNF-R55 elicit caspase-independent PCD through similar pathways, although without participation of cathepsins. Cells overexpressing acid ceramidase (AC), an enzyme that metabolizes the sphingolipid ceramide, show enhanced survival from TRAIL-induced caspase-independent PCD but not from apoptosis, implicating a function of ceramide as a key mediator in caspase-independent PCD (but not apoptosis) induced by mTRAIL-R2. In concert with the enhanced resistance of AC-overexpressing cells against caspase-independent PCD induced by TNF, our results suggest that ceramide acts as a common mediator of caspase-independent PCD caused by death receptors such as mTRAIL-R2 and TNF-R55.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Ceramides/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Cell Line , Galactosylgalactosylglucosylceramidase/metabolism , Humans , Mice , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor Decoy Receptors/metabolism
7.
FASEB J ; 19(14): 1945-56, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16319138

ABSTRACT

Although numerous studies have implicated the sphingolipid ceramide in the induction of cell death, a causative function of ceramide in caspase-dependent apoptosis remains a highly debated issue. Here, we show that ceramide is a key mediator of a distinct route to programmed cell death (PCD), i.e., caspase-independent PCD. Under conditions where apoptosis is either not initiated or actively inhibited, TNF induces caspase-independent PCD in L929 fibrosarcoma cells, NIH3T3 fibroblasts, human leukemic Jurkat T cells, and lung fibroblasts by increasing intracellular ceramide levels prior to the onset of cell death. Survival is significantly enhanced when ceramide accumulation is prevented, as demonstrated in fibroblasts genetically deficient for acid sphingomyelinase, in L929 cells overexpressing acid ceramidase, by pharmacological intervention, or by RNA interference. Jurkat cells deficient for receptor-interacting protein 1 (RIP1) do not accumulate ceramide and therefore are fully resistant to caspase-independent PCD whereas Jurkat cells overexpressing the mitochondrial protein Bcl-2 are partially protected, implicating RIP1 and mitochondria as components of the ceramide death pathway. Our data point to a role of caspases (but not cathepsins) in suppressing the ceramide death pathway under physiological conditions. Moreover, clonogenic survival of tumor cells is clearly reduced by induction of the ceramide death pathway, promising additional options for the development of novel tumor therapies.


Subject(s)
Apoptosis , Caspases/metabolism , Ceramides/pharmacology , Animals , Benzoquinones , Cell Line , Ceramides/metabolism , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Flow Cytometry , Humans , Immunoblotting , Jurkat Cells , Lactams, Macrocyclic , Lung/metabolism , Membrane Potentials , Mice , Mitochondria/metabolism , NIH 3T3 Cells , Protein Serine-Threonine Kinases/metabolism , Quinones/pharmacology , RNA Interference , Reactive Oxygen Species , Receptor-Interacting Protein Serine-Threonine Kinases , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Biochem Biophys Res Commun ; 335(2): 575-83, 2005 Sep 23.
Article in English | MEDLINE | ID: mdl-16083851

ABSTRACT

Tumor necrosis factor (TNF) can induce caspase-dependent (apoptotic) and caspase-independent pathways to programmed cell death (PCD). Here, we demonstrate that stable transfection of a cDNA encompassing the C-terminal apoptosis inhibitory domain (AID) of FE65-like protein 1 into mouse L929 fibrosarcoma cells protects from caspase-independent as well as from apoptotic PCD induced by TNF. We show that the AID does not protect from caspase-independent PCD elicited by 1-methyl-3-nitro-1-nitrosoguanidine, suggesting that the AID might prevent cell death by affecting assembly of the death inducing signaling complex of the 55 kDa TNF receptor or clustering of the receptor itself. Interference with caspase-independent PCD mediated by the sphingolipid ceramide further increases protection conferred by the AID, as does the antioxidant butylated hydroxyanisole, implicating ceramide and reactive oxygen species as potential factors interacting with caspase-independent PCD regulated by the AID.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Apoptosis , Caspases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Antioxidants/pharmacology , Base Sequence , Cell Death , Cell Line , Cell Line, Tumor , Cell Survival , Ceramides/metabolism , Cloning, Molecular , Dose-Response Relationship, Drug , Genetic Vectors , Humans , Immunoblotting , Mice , Molecular Sequence Data , Nitrosoguanidines/pharmacology , Polymerase Chain Reaction , Protein Structure, Tertiary , Reactive Oxygen Species , Sequence Homology, Amino Acid , Sphingolipids/chemistry , Time Factors , Transfection
9.
Biochem Biophys Res Commun ; 329(1): 397-405, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15721320

ABSTRACT

Tumor necrosis factor (TNF) contributes to insulin resistance by binding to the 55kDa TNF receptor (TNF-R55), resulting in serine phosphorylation of proteins such as insulin receptor (IR) substrate (IRS)-1, followed by reduced tyrosine phosphorylation of IRS-1 through the IR and, thereby, diminished IR signal transduction. Through independent receptor domains, TNF-R55 activates a neutral (N-SMase) and an acid sphingomyelinase (A-SMase), that both generate the sphingolipid ceramide. Multiple candidate kinases have been identified that serine-phosphorylate IRS-1 in response to TNF or ceramide. However, due to the fact that the receptor domain of TNF-R55 mediating inhibition of the IR has not been mapped, it is currently unknown whether TNF exerts these effects with participation of N-SMase or A-SMase. Here, we identify the death domain of TNF-R55 as responsible for the inhibitory effects of TNF on tyrosine phosphorylation of IRS-1, implicating ceramide generated by A-SMase as a downstream mediator of inhibition of IR signaling.


Subject(s)
Insulin/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cells, Cultured , Ceramides/chemistry , Ceramides/metabolism , Cryopreservation , Fibroblasts/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Sphingomyelin Phosphodiesterase/chemistry , Sphingomyelin Phosphodiesterase/metabolism , Transfection , Tyrosine/chemistry
10.
J Immunol ; 169(9): 5161-70, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391233

ABSTRACT

Factor associated with neutral sphingomyelinase activation (FAN) represents a p55 TNFR (TNF-R55)-associated protein essential for the activation of neutral sphingomyelinase. By means of the yeast interaction trap system, we have identified the scaffolding protein receptor for activated C-kinase (RACK)1 as an interaction partner of FAN. Mapping studies in yeast revealed that RACK1 is recruited to the C-terminal WD-repeat region of FAN and binds to FAN through a domain located within WD repeats V to VII of RACK1. Our data indicate that binding of both proteins is not mediated by linear motifs but requires folding into a secondary structure, such as the multibladed propeller characteristic of WD-repeat proteins. The interaction of FAN and RACK1 was verified in vitro by glutathione S-transferase-based coprecipitation assays as well as in eukaryotic cells by coimmunoprecipitation experiments. Colocalization studies in transfected cells suggest that TNF-R55 forms a complex with FAN and that this complex recruits RACK1 to the plasma membrane. Furthermore, activation of N-SMase by TNF was strongly enhanced when RACK1, FAN, and a noncytotoxic TNF-R55 mutant were expressed concurrently, suggesting RACK1 as a modulator of N-SMase activation. Together, these findings implicate RACK1 as a novel component of the signaling pathways of TNF-R55.


Subject(s)
Antigens, CD/physiology , Protein Kinase C/metabolism , Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/immunology , Sphingomyelin Phosphodiesterase/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , COS Cells , Cell Line , Enzyme Activation/genetics , Enzyme Activation/immunology , HeLa Cells , Humans , Intracellular Fluid/metabolism , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Precipitin Tests , Protein Binding/genetics , Protein Binding/immunology , Protein Interaction Mapping/methods , Protein Kinase C/chemistry , Protein Kinase C/genetics , Proteins/chemistry , Proteins/genetics , Receptors for Activated C Kinase , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Repetitive Sequences, Amino Acid , Signal Transduction/genetics , Sphingomyelin Phosphodiesterase/chemistry , Sphingomyelin Phosphodiesterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...