Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(6): 1012-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25430793

ABSTRACT

Receptor-interacting protein kinase 4 (RIPK4)-deficient mice have epidermal defects and fusion of all external orifices. These are similar to Bartsocas-Papas syndrome and popliteal pterygium syndrome (PPS) in humans, for which causative mutations have been documented in the RIPK4 and IRF6 (interferon regulatory factor 6) gene, respectively. Although genetically distinct, these syndromes share the anomalies of marked pterygia, syndactyly, clefting and hypoplastic genitalia. Despite the strong resemblance of these two syndromes, no molecular connection between the transcription factor IRF6 and the kinase RIPK4 was known and the mechanism underlying the phenotype was unclear. Here we describe that RIPK4 deficiency in mice causes epithelial fusions associated with abnormal periderm development and aberrant ectopic localization of E-cadherin on the apical membrane of the outer peridermal cell layers. In Xenopus, RIPK4 depletion causes the absence of ectodermal epiboly and concomitant gastrulation defects that phenocopy ectopic expression of dominant-negative IRF6. We found that IRF6 controls RIPK4 expression and that wild-type, but not kinase-dead, RIPK4 can complement the gastrulation defect in Xenopus caused by IRF6 malfunctioning. In contrast to the mouse, we observed only minor effects on cadherin membrane expression in Xenopus RIPK4 morphants. However, gastrulation defects were associated with a virtual absence of cortical actin in the ectodermal cells that face the blastocoel cavity and this was phenocopied in embryos expressing dominant-negative IRF6. A role for RIPK4 in actin cytoskeleton organization was also revealed in mouse epidermis and in human epithelial HaCaT cells. In conclusion, we showed that in mice RIPK4 is implicated in cortical actin organization and in E-cadherin localization or function, which can explain the characteristic epithelial fusions observed in PPSs. In addition, we provide a novel molecular link between IRF6 and RIPK4 that unifies the different PPSs to a common molecular pathway.


Subject(s)
Cleft Lip/metabolism , Cleft Palate/metabolism , Eye Abnormalities/metabolism , Fingers/abnormalities , Interferon Regulatory Factors/metabolism , Knee Joint/abnormalities , Lower Extremity Deformities, Congenital/metabolism , Protein Serine-Threonine Kinases/metabolism , Syndactyly/metabolism , Urogenital Abnormalities/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cleft Lip/genetics , Cleft Palate/genetics , Eye Abnormalities/genetics , Humans , Immunohistochemistry , Interferon Regulatory Factors/genetics , Keratinocytes/cytology , Keratinocytes/metabolism , Knee Joint/metabolism , Lentivirus , Lower Extremity Deformities, Congenital/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Protein Serine-Threonine Kinases/genetics , Syndactyly/genetics , Urogenital Abnormalities/genetics
2.
Cell Death Dis ; 3: e437, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23190609

ABSTRACT

Necrostatin-1 (Nec-1) is widely used in disease models to examine the contribution of receptor-interacting protein kinase (RIPK) 1 in cell death and inflammation. We studied three Nec-1 analogs: Nec-1, the active inhibitor of RIPK1, Nec-1 inactive (Nec-1i), its inactive variant, and Nec-1 stable (Nec-1s), its more stable variant. We report that Nec-1 is identical to methyl-thiohydantoin-tryptophan, an inhibitor of the potent immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO). Both Nec-1 and Nec-1i inhibited human IDO, but Nec-1s did not, as predicted by molecular modeling. Therefore, Nec-1s is a more specific RIPK1 inhibitor lacking the IDO-targeting effect. Next, although Nec-1i was ∼100 × less effective than Nec-1 in inhibiting human RIPK1 kinase activity in vitro, it was only 10 times less potent than Nec-1 and Nec-1s in a mouse necroptosis assay and became even equipotent at high concentrations. Along the same line, in vivo, high doses of Nec-1, Nec-1i and Nec-1s prevented tumor necrosis factor (TNF)-induced mortality equally well, excluding the use of Nec-1i as an inactive control. Paradoxically, low doses of Nec-1 or Nec-1i, but not Nec -1s, even sensitized mice to TNF-induced mortality. Importantly, Nec-1s did not exhibit this low dose toxicity, stressing again the preferred use of Nec-1s in vivo. Our findings have important implications for the interpretation of Nec-1-based data in experimental disease models.


Subject(s)
Imidazoles/administration & dosage , Imidazoles/chemistry , Indoles/administration & dosage , Indoles/chemistry , Systemic Inflammatory Response Syndrome/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Disease Models, Animal , Drug Therapy , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Species Specificity
3.
Cell Death Dis ; 2: e220, 2011 Oct 20.
Article in English | MEDLINE | ID: mdl-22012254

ABSTRACT

Caspases are aspartate-specific cysteine proteases that have an essential role in apoptosis and inflammation, and contribute to the maintenance of homeostasis in the intestine. These facts, together with the knowledge that caspases are implicated in host-microbe crosstalk, prompted us to investigate the effect of caspase (Casp)1, -3 and -7 deficiency on the composition of the murine gut microbiota. We observed significant changes in the abundance of the Firmicutes and Bacteroidetes phyla, in particular the Lachnospiraceae, Porphyromonodaceae and Prevotellacea families, when comparing Casp-1, -7 and -3 knockout mice with wild-type mice. Our data point toward an intricate relationship between these caspases and the composition of the murine gut microflora.


Subject(s)
Caspases/deficiency , Gastrointestinal Tract/enzymology , Gastrointestinal Tract/microbiology , Animals , Apoptosis/physiology , Caspases/biosynthesis , Caspases/genetics , Metagenome , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Cell Death Differ ; 18(12): 1845-53, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21566665

ABSTRACT

The ubiquitin-editing enzyme A20 (tumor necrosis factor-α-induced protein 3) serves as a critical brake on nuclear factor κB (NF-κB) signaling. In humans, polymorphisms in or near the A20 gene are associated with several inflammatory disorders, including psoriasis. We show here that epidermis-specific A20-knockout mice (A20(EKO)) develop keratinocyte hyperproliferation, but no signs of skin inflammation, such as immune cell infiltration. However, A20(EKO) mice clearly developed ectodermal organ abnormalities, including disheveled hair, longer nails and sebocyte hyperplasia. This phenotype resembles that of mice overexpressing ectodysplasin-A1 (EDA-A1) or the ectodysplasin receptor (EDAR), suggesting that A20 negatively controls EDAR signaling. We found that A20 inhibited EDAR-induced NF-κB signaling independent from its de-ubiquitinating activity. In addition, A20 expression was induced by EDA-A1 in embryonic skin explants, in which its expression was confined to the hair placodes, known to be the site of EDAR expression. In summary, our data indicate that EDAR-induced NF-κB levels are controlled by A20, which functions as a negative feedback regulator, to assure proper skin homeostasis and epidermal appendage development.


Subject(s)
Cysteine Endopeptidases/genetics , Epidermis/physiology , Homeostasis , Intracellular Signaling Peptides and Proteins/genetics , Keratinocytes/metabolism , NF-kappa B/metabolism , Animals , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/physiology , Ectodysplasins/pharmacology , Ectodysplasins/physiology , Edar Receptor/agonists , Edar Receptor/antagonists & inhibitors , Edar Receptor/metabolism , Epidermis/pathology , Feedback, Physiological , Genes, Reporter , HEK293 Cells , Hair/abnormalities , Hair/embryology , Humans , Hyperplasia , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Keratinocytes/physiology , Ki-67 Antigen/metabolism , Luciferases/biosynthesis , Luciferases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Tissue Culture Techniques , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology
6.
Cell Death Dis ; 1: e18, 2010.
Article in English | MEDLINE | ID: mdl-21364619

ABSTRACT

Autophagy and apoptosis are two important and interconnected stress-response mechanisms. However, the molecular interplay between these two pathways is not fully understood. To study the fate and function of autophagic proteins at the onset of apoptosis, we used a cellular model system in which autophagy precedes apoptosis. IL-3 depletion of Ba/F3 cells caused caspase (casp)-mediated cleavage of Beclin-1 and PI3KC3, two crucial components of the autophagy-inducing complex. We identified two casp cleavage sites in Beclin-1, TDVD(133) and DQLD(149), cleavage at which yields fragments lacking the autophagy-inducing capacity. Noteworthy, the C-terminal fragment, Beclin-1-C, localized predominantly at the mitochondria and sensitized the cells to apoptosis. Moreover, on isolated mitochondria, recombinant Beclin-1-C was able to induce the release of proapoptotic factors. These findings point to a mechanism by which casp-dependent generation of Beclin-1-C creates an amplifying loop enhancing apoptosis upon growth factor withdrawal.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Autophagy , Caspases/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/genetics , Beclin-1 , Cell Line , Humans , Interleukin-3/genetics , Interleukin-3/metabolism , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Cell Death Differ ; 17(6): 922-30, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20010783

ABSTRACT

Necroptosis, necrosis and secondary necrosis following apoptosis represent different modes of cell death that eventually result in similar cellular morphology including rounding of the cell, cytoplasmic swelling, rupture of the plasma membrane and spilling of the intracellular content. Subcellular events during tumor necrosis factor (TNF)-induced necroptosis, H(2)O(2)-induced necrosis and anti-Fas-induced secondary necrosis were studied using high-resolution time-lapse microscopy. The cellular disintegration phase of the three types of necrosis is characterized by an identical sequence of subcellular events, including oxidative burst, mitochondrial membrane hyperpolarization, lysosomal membrane permeabilization and plasma membrane permeabilization, although with different kinetics. H(2)O(2)-induced necrosis starts immediately by lysosomal permeabilization. In contrast, during TNF-mediated necroptosis and anti-Fas-induced secondary necrosis, this is a late event preceded by a defined signaling phase. TNF-induced necroptosis depends on receptor-interacting protein-1 kinase, mitochondrial complex I and cytosolic phospholipase A(2) activities, whereas H(2)O(2)-induced necrosis requires iron-dependent Fenton reactions.


Subject(s)
Necrosis/metabolism , Animals , Cell Line, Tumor , Cell Membrane Permeability , Electron Transport Complex I/metabolism , Hydrogen Peroxide/toxicity , Iron/metabolism , Lysosomes/metabolism , Membrane Potential, Mitochondrial , Mice , Necrosis/chemically induced , Necrosis/enzymology , Phospholipases A2, Cytosolic/metabolism , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/toxicity
8.
Cell Death Differ ; 14(1): 44-55, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17053807

ABSTRACT

Caspases, a family of evolutionarily, conserved cysteinyl proteases, mediate both apoptosis and inflammation through aspartate-specific cleavage of a wide number of cellular substrates. Most substrates of apoptotic caspases have been conotated with cellular dismantling, while inflammatory caspases mediate the proteolytic activation of inflammatory cytokines. Through detailed functional analysis of conditional caspase-deficient mice or derived cells, caspase biology has been extended to cellular responses such as cell differentiation, proliferation and NF-kappaB activation. Here, we discuss recent data indicating that non-apoptotic functions of caspases involve proteolysis exerted by their catalytic domains as well as non-proteolytic functions exerted by their prodomains. Homotypic oligomerization motifs in the latter mediate the recruitment of adaptors and effectors that modulate NF-kappaB activation. The non-apoptotic functions of caspases suggest that they may become activated independently of--or without--inducing an apoptotic cascade. Moreover, the existence of non-catalytic caspase-like molecules such as human caspase-12, c-FLIP and CARD-only proteins further supports the non-proteolytic functions of caspases in the regulation of cell survival, proliferation, differentiation and inflammation.


Subject(s)
Caspases/physiology , Cell Differentiation , Cell Proliferation , Cell Survival , Animals , Caspases/chemistry , Caspases/genetics , Caspases/immunology , Humans , Inflammation/enzymology , Inflammation/immunology , NF-kappa B/metabolism , Phylogeny
9.
Cell Death Differ ; 12 Suppl 2: 1497-508, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16247497

ABSTRACT

Homeostasis implies a balance between cell growth and cell death. This balance is essential for the development and maintenance of multicellular organisms. Homeostasis is controlled by several mechanisms including apoptosis, a process by which cells condemned to death are completely eliminated. However, in some cases, total destruction and removal of dead cells is not desirable, as when they fulfil a specific function such as formation of the skin barrier provided by corneocytes, also known as terminally differentiated keratinocytes. In this case, programmed cell death results in accumulation of functional cell corpses. Previously, this process has been associated with apoptotic cell death. In this overview, we discuss differences and similarities in the molecular regulation of epidermal programmed cell death and apoptosis. We conclude that despite earlier confusion, apoptosis and cornification occur through distinct molecular pathways, and that possibly antiapoptotic mechanisms are implicated in the terminal differentiation of keratinocytes.


Subject(s)
Apoptosis/physiology , Cell Differentiation/physiology , Epidermal Cells , Keratinocytes/cytology , Animals , Cell Nucleus/physiology , Cytoskeleton/physiology , Epidermis/ultrastructure , Humans , Keratinocytes/ultrastructure , Mitochondrial Proteins/physiology , Peptide Hydrolases/physiology , Signal Transduction , Transcription Factors/physiology , Transglutaminases/physiology
11.
Cell Death Differ ; 9(11): 1207-11, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12404119

ABSTRACT

Caspases are crucial for the initiation, propagation and execution of apoptosis. They normally exist as proenzymes, which can be activated through recruitment into activating complexes and by proteolytic cleavage by other caspases or proteases. Perturbation of organelles such as nuclei, endoplasmatic reticulum and mitochondria results in the activation of caspases. A number of caspases (-2, -3, -8 and -9) were published as being localized in the intermembrane space of mitochondria. However, in three different models of apoptosis (anti-Fas-induced cell death in murine hepatocytes, Fas ligand-induced apoptosis in Jurkat cells and apoptosis induced by growth factor withdrawal in Ba/F3 cells) we could not identify a mitochondrial location of caspases, neither under control nor under apoptotic conditions. In all three apoptotic models caspases were found in the cytosolic (caspases-2, -3, -6, -7, -8, -9) and nuclear subcellular fractions (caspases-2, -3). In another approach we treated isolated liver mitochondria with truncated Bid. Although tBid-dependent release of Cytochrome c, AIF, adenylate kinase, Smac/DIABLO and Omi/HtrA2 could be demonstrated, none of the caspases were detectable both in the supernatant and the mitochondrial fraction after treatment. Our results demonstrate that, in contrast to previous studies, no caspases-2, -3, -8 and -9 are associated with the mitochondrial fraction. These findings support the concept of a separate compartmentalization between proapoptotic cofactors in the mitochondria and silent precursor caspases in the cytosol.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Mitochondria/metabolism , Animals , Biomarkers , Caspase 2 , Enzyme Precursors/metabolism , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL
13.
Cell Death Differ ; 9(3): 301-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11859412

ABSTRACT

A crucial event in the process of apoptosis is caspase-dependent generation of truncated Bid (tBid), inducing release of cytochrome c. In an in vitro reconstitution system we combined purified recombinant tBid with isolated liver mitochondria and identified the released proteins using a proteomic matrix-assisted laser desorption ionization post-source decay (MALDI-PSD) approach. In order to meet physiological conditions, the concentration of tBid was chosen such that it was unable to induce cytochrome c release in mitochondria derived from liver-specific Bcl-2-transgenic mice. Several mitochondrial proteins were identified to be released in a tBid-dependent way, among which cytochrome c, DIABLO/Smac, adenylate kinase 2, acyl-CoA-binding protein, endonuclease G, polypyrimidine tract-binding protein, a type-I RNA helicase, a WD-40 repeat-containing protein and the serine protease Omi. Western blotting confirmed the absence of adenylate kinase 3, a matrix mitochondrial protein. These results demonstrate that a physiologically relevant concentration of tBid is sufficient to induce release of particular intermembrane mitochondrial proteins belonging to a broad molecular-mass range.


Subject(s)
Apoptosis/physiology , Carrier Proteins/pharmacology , Mitochondria, Liver/drug effects , Mitochondrial Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Adenylate Kinase/analysis , Adenylate Kinase/metabolism , Animals , Apoptosis Regulatory Proteins , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/analysis , Carrier Proteins/metabolism , Cytochrome c Group/analysis , Cytochrome c Group/metabolism , Diazepam Binding Inhibitor/analysis , Endodeoxyribonucleases/analysis , Endodeoxyribonucleases/metabolism , High-Temperature Requirement A Serine Peptidase 2 , Isoenzymes/analysis , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/analysis , Polypyrimidine Tract-Binding Protein , RNA-Binding Proteins/analysis , RNA-Binding Proteins/metabolism , Recombinant Proteins/pharmacology , Ribonucleoproteins/analysis , Ribonucleoproteins/metabolism , Serine Endopeptidases/analysis , Serine Endopeptidases/metabolism
14.
Cell Death Differ ; 9(1): 20-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11803371

ABSTRACT

Proteome analysis of supernatant of isolated mitochondria exposed to recombinant tBid, a proapoptotic Bcl-2 member, revealed the presence of the serine protease Omi, also called HtrA2. This release was prevented in mitochondria derived from Bcl-2-transgenic mice. Release of Omi under apoptotic conditions was confirmed in vivo in livers from mice injected with agonistic anti-Fas antibodies and was prevented in livers from Bcl-2 transgenic mice. Omi release also occurs in apoptotic dying but not in necrotic dying fibrosarcoma L929 cells, treated with anti-Fas antibodies and TNF, respectively. The amino acid sequence reveals the presence of an XIAP interaction motif at the N-terminus of mature Omi. We demonstrate an interaction between endogeneous Omi and recombinant XIAP. Furthermore we show that endogenous Omi is involved in enhanced activation of caspases in cytosolic extracts.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Mitochondria/metabolism , Proteins/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/pharmacology , Cells, Cultured , Cytosol/metabolism , Enzyme Activation , High-Temperature Requirement A Serine Peptidase 2 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins , Molecular Sequence Data , Translocation, Genetic/drug effects , X-Linked Inhibitor of Apoptosis Protein
15.
Cell Death Differ ; 8(12): 1136-42, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753562

ABSTRACT

A hallmark of apoptosis is the fragmentation of nuclear DNA. Although this activity involves the caspase-3-dependent DNAse CAD (caspase-activated DNAse), evidence exists that DNA fragmentation can occur independently of caspase activity. Here we report on the ability of truncated Bid (tBid) to induce the release of a DNAse activity from mitochondria. This DNAse activity was identified by mass spectrometry as endonuclease G, an abundant 30 kDa protein released from mitochondria under apoptotic conditions. No tBid-induced endonuclease G release could be observed in mitochondria from Bcl-2-transgenic mice. The in vivo occurrence of endonuclease G release from mitochondria during apoptosis was confirmed in the liver from mice injected with agonistic anti-Fas antibody and is completely prevented in Bcl-2 transgenic mice. These data indicate that endonuclease G may be involved in CAD-independent DNA fragmentation during cell death pathways in which truncated Bid is generated.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , DNA Fragmentation , Endodeoxyribonucleases/physiology , Mitochondrial Proteins/physiology , Animals , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/pharmacology , Cytochrome c Group/metabolism , Endodeoxyribonucleases/metabolism , Genes, bcl-2/physiology , Mice , Mitochondrial Proteins/metabolism
16.
Cell Signal ; 13(10): 717-25, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11602182

ABSTRACT

TT-232 is a somatostatin analogue containing a five-residue ring structure. The present report describes TT-232-induced signalling events in A431 cells, where a 4-h preincubation with the peptide irreversibly induced a cell death program, which involves DNA-laddering and the appearance of shrunken nuclei, but is unrelated to somatostatin signalling. Early intracellular signals of TT-232 include a transient two-fold activation of the extracellular signal-regulated kinase (ERK2) and a strong and sustained activation of the stress-activated protein kinases c-Jun NH(2)-terminal kinase (JNK)/SAPK and p38MAPK. Blocking the signalling to ERK or p38MAPK activation had no effect on the TT-232-induced cell killing. At the commitment time for inducing cell death, TT-232 decreased EGFR-tyrosine phosphorylation and prevented epidermial growth factor (EGF)-induced events like cRaf-1 and ERK2 activation. Signalling to ERK activation by FCS, phorbol 12-myristate 13-acetate (PMA) and platelet-derived growth factor (PDGF) was similarly blocked. Our data suggest that TT-232 triggers an apoptotic type of cell death, concomitant with a strong activation of JNK and a blockade of cellular ERK2 activation pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Peptides, Cyclic/pharmacology , Drug Antagonism , Epidermal Growth Factor/pharmacology , Humans , Kinetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 8 , Neoplasms/enzymology , Neoplasms/pathology , Somatostatin/pharmacology , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
17.
Photochem Photobiol ; 74(2): 133-42, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11547546

ABSTRACT

In this study we show that overexpression of Bcl-2 in PC60R1R2 cells reveals a caspase-dependent mechanism of cytochrome c release following photodynamic therapy (PDT) with hypericin. Bcl-2 overexpression remarkably delayed cytochrome c release, procaspase-3 activation and poly(adenosine diphosphate-ribose)polymerase cleavage during PDT-induced apoptosis while it did not protect against PDT-induced necrosis. PDT-treated cells showed a reduction in the mitochondrial membrane potential which occurred with similar kinetics in PC60R1R2 and PC60R1R2/Bcl-2 cells, and was affected neither by the permeability transition pore inhibitor cyclosporin A nor by the caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (zVAD-fmk). Hypericin-induced mitochondrial depolarization coincided with cytochrome c release in PC60R1R2 cells while it precedes massive cytochrome c efflux in PC60R1R2/Bcl-2 cells. Preincubation of PC60R1R2 cells with zVAD-fmk or cyclosporin A did not prevent the mitochondrial efflux of cytochrome c, and caspase inhibition only partially protected the cells from PDT-induced apoptosis. In contrast, in PC60R1R2/Bcl-2 cells cytochrome c release and apoptosis were suppressed by addition of zVAD-fmk or cyclosporin A. These observations suggest that the progression of the PDT-induced apoptotic process in Bcl-2-overexpressing cells involves a caspase-dependent feed-forward amplification loop for the release of cytochrome c.


Subject(s)
Cytochrome c Group/metabolism , Perylene/analogs & derivatives , Perylene/pharmacology , Photochemotherapy , Viral Proteins , Animals , Anthracenes , Caspase 3 , Caspases/metabolism , Cell Death/drug effects , Cell Line , Enzyme Activation/drug effects , Enzyme Precursors/metabolism , Genes, bcl-2 , Hybridomas , Membrane Potentials/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Rats , Serpins/genetics , Transfection
18.
Cell Death Differ ; 8(8): 829-40, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11526436

ABSTRACT

In L929sAhFas cells, tumor necrosis factor (TNF) leads to necrotic cell death, whereas agonistic anti-Fas antibodies elicit apoptotic cell death. Apoptosis, but not necrosis, is correlated with a rapid externalization of phosphatidylserine and the appearance of a hypoploid population. During necrosis no cytosolic and organelle-associated active caspase-3 and -7 fragments are detectable. The necrotic process does not involve proteolytic generation of truncated Bid; moreover, no mitochondrial release of cytochrome c is observed. Bcl-2 overexpression slows down the onset of necrotic cell death. In the case of apoptosis, active caspases are released to the culture supernatant, coinciding with the release of lactate dehydrogenase. Following necrosis, mainly unprocessed forms of caspases are released. Both TNF-induced necrosis and necrosis induced by anti-Fas in the presence of the caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone are prevented by the serine protease inhibitor N-tosyl-L-phenylalanine chloromethylketone and the oxygen radical scavenger butylated hydroxyanisole, while Fas-induced apoptosis is not affected.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Mitochondria/metabolism , Necrosis , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , Animals , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Caspases/drug effects , Cytochrome c Group/metabolism , Humans , Kinetics , Mice , Mitochondria/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor/drug effects , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Time Factors , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Necrosis Factor-alpha/pharmacology , fas Receptor/drug effects , fas Receptor/metabolism
19.
Mech Dev ; 104(1-2): 89-98, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11404083

ABSTRACT

At weaning, milk producing mammary epithelial cells undergo apoptosis and are removed by phagocytosis. Here, we show that mouse mammary gland involution is associated with mitochondrial cytochrome c release and processing of numerous caspases, including caspase-1, -3, -7, -8 and -9. Induction of caspase-3-like activity paralleled cleavage of poly-(ADP--ribose) polymerase. Dexamethasone inhibited processing of caspase-3, -7 and -8 and apoptosis, but had no effect on caspase-1 accumulation and cytochrome c release. In Bcl-2 transgenic animals, cytochrome c release, caspase activation and apoptosis were impaired. Thus, the pro-apoptotic signaling pathway in mammary epithelial cells during involution involves the release of cytochrome c and activation of caspases. It is inhibited by Bcl-2 at the mitochondrial level and by dexamethasone at a post-mitochondrial level.


Subject(s)
Caspases/metabolism , Cytochrome c Group/metabolism , Mammary Glands, Animal/physiology , Weaning , Animals , Apoptosis , Blotting, Western , Caspase 1/metabolism , Caspase 3 , Caspase 7 , Caspase 8 , Caspase 9 , Dexamethasone/pharmacology , Enzyme Activation , Glucocorticoids/pharmacology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Spectrometry, Fluorescence , Time Factors
20.
J Biol Chem ; 276(29): 27098-103, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11371574

ABSTRACT

Incubation of murine tumor necrosis factor (mTNF) at subnanomolar concentrations results in partial dissociation of the trimers, coinciding with a decrease in bioactivity. Using size-exclusion chromatography, we observed that the conversion of labeled mTNF to monomers is not only prevented by coincubation with an excess of unlabeled mTNF but also with unlabeled human TNF (hTNF). Moreover, after coincubation of mTNF and hTNF four different TNF complexes were revealed by native polyacrylamide gel electrophoresis, viz. homotrimeric mTNF and hTNF, as well as two complexes with an intermediate migration pattern. Analytical gel filtration in combination with native polyacrylamide gel electrophoresis and Western blot immunodetection indicated that these new complexes consisted of heterotrimeric TNF molecules. We conclude that an exchange of monomers takes place during coincubation of two different species of TNF, which results in homotrimeric and heterotrimeric TNF. To assess receptor interaction in vitro, TNF heterotrimeric molecules were used as obtained after incubation of mTNF with labeled hTNF (which only binds to mTNF receptor I) or with labeled mutein mTNF75 (specific for mTNF receptor II). These heterotrimers were retained by both mTNF receptors, which means that the mTNF subunits incorporated in heterotrimeric complexes still can bind to both types of TNF receptor. In addition, the gradual decrease in mTNF bioactivity during preincubation at subnanomolar concentrations was prevented by the presence of mutein mTNF75, which is inactive in an L929 cytotoxicity assay, indicating that heterotrimerization can influence the overall bioactivity.


Subject(s)
Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies, Monoclonal/immunology , Biopolymers , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Humans , Mice , Species Specificity , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...