Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
EMBO J ; 23(23): 4679-89, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15526035

ABSTRACT

Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein that, after apoptosis induction, translocates to the nucleus where it participates in apoptotic chromatinolysis. Here, we show that human or mouse cells lacking AIF as a result of homologous recombination or small interfering RNA exhibit high lactate production and enhanced dependency on glycolytic ATP generation, due to severe reduction of respiratory chain complex I activity. Although AIF itself is not a part of complex I, AIF-deficient cells exhibit a reduced content of complex I and of its components, pointing to a role of AIF in the biogenesis and/or maintenance of this polyprotein complex. Harlequin mice with reduced AIF expression due to a retroviral insertion into the AIF gene also manifest a reduced oxidative phosphorylation (OXPHOS) in the retina and in the brain, correlating with reduced expression of complex I subunits, retinal degeneration, and neuronal defects. Altogether, these data point to a role of AIF in OXPHOS and emphasize the dual role of AIF in life and death.


Subject(s)
Membrane Proteins/deficiency , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis , Apoptosis Inducing Factor , Brain/metabolism , Cells, Cultured , Electron Transport Complex I/biosynthesis , Electron Transport Complex III/biosynthesis , Flavoproteins/genetics , Flavoproteins/metabolism , Glucose/metabolism , Humans , Lactic Acid/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocardium/metabolism , Organ Specificity , Oxidative Phosphorylation , Phylogeny , RNA, Small Interfering/metabolism , Retina/metabolism , Yeasts/genetics , Yeasts/growth & development , Yeasts/metabolism
2.
J Cell Biol ; 166(7): 969-74, 2004 Sep 27.
Article in English | MEDLINE | ID: mdl-15381687

ABSTRACT

Apoptosis-inducing factor (AIF), a key regulator of cell death, is essential for normal mammalian development and participates in pathological apoptosis. The proapoptotic nature of AIF and its mode of action are controversial. Here, we show that the yeast AIF homologue Ynr074cp controls yeast apoptosis. Similar to mammalian AIF, Ynr074cp is located in mitochondria and translocates to the nucleus of yeast cells in response to apoptotic stimuli. Purified Ynr074cp degrades yeast nuclei and plasmid DNA. YNR074C disruption rescues yeast cells from oxygen stress and delays age-induced apoptosis. Conversely, overexpression of Ynr074cp strongly stimulates apoptotic cell death induced by hydrogen peroxide and this effect is attenuated by disruption of cyclophilin A or the yeast caspase YCA1. We conclude that Ynr074cp is a cell death effector in yeast and rename it AIF-1 (Aif1p, gene AIF1).


Subject(s)
Flavoproteins/metabolism , Membrane Proteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Apoptosis/genetics , Apoptosis Inducing Factor , Caspase Inhibitors , Caspases/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Cellular Senescence/genetics , Cyclophilin A/antagonists & inhibitors , Cyclophilin A/metabolism , DNA/genetics , DNA/metabolism , DNA, Complementary/analysis , DNA, Complementary/genetics , Enzyme Inhibitors/pharmacology , Flavoproteins/genetics , Flavoproteins/isolation & purification , Hydrogen Peroxide/pharmacology , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mitochondria/genetics , Mitochondria/metabolism , Molecular Sequence Data , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/isolation & purification , Oxidative Stress/genetics , Protein Transport/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
3.
Clin Cancer Res ; 10(17): 5665-9, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15355891

ABSTRACT

The proapoptotic protein apoptosis protein activating factor-1 (Apaf-1), which is normally located in the cytoplasm, can translocate to the nucleus before non-small cell lung carcinoma (NSCLC) cells manifest signs of apoptosis such as mitochondrial damage, caspase activation, or chromatin condensation. This may indicate a stage of imminent apoptosis. Importantly, we found that 24% (15 of 62) of resected stage I NSCLC (T(1)N(0)M(0) or T(2)N(0)M(0)), manifested a marked nuclear localization of Apaf-1 (Apaf-1(Nuc)), as compared with the mostly cytoplasmic localization of Apaf-1 found in the remaining tumors (Apaf-1(Cyt)). After a median follow-up of 6.31 years, the actuarial 5-year overall survival rates were 89% (56-98%) in the Apaf-1(Nuc) group and 54% (36-71%) in the Apaf-1(Cyt) group (P = 0.039). No correlation between the subcellular localization of Apaf-1 and that of p53 and Hsp70 could be established. Thus, the subcellular location of Apaf-1 (but not that of p53 or Hsp70) constitutes an accurate prognostic factor for overall survival in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Cell Nucleus/enzymology , Lung Neoplasms/enzymology , Proteins/metabolism , Adult , Aged , Aged, 80 and over , Apoptotic Protease-Activating Factor 1 , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/surgery , Caspases/metabolism , Chromatin/metabolism , Cytoplasm/metabolism , Enzyme Activation , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Mitochondria/metabolism , Neoplasm Staging , Predictive Value of Tests , Protein Transport , Subcellular Fractions , Survival Rate
4.
J Cell Sci ; 117(Pt 19): 4461-8, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15316071

ABSTRACT

Stress granules (SG) are dynamic cytoplasmic foci in which stalled translation initiation complexes accumulate. In conditions of acute cellular redox, stress cells manipulated to lose the expression of apoptosis-inducing factor (AIF) nucleate SG signature proteins (e.g. TIA-1, PABP1) more efficiently than AIF-positive controls. AIF also inhibited SG formation induced by the RasGAP-associated endoribonuclease G3BP. Retransfection of mouse AIF into cells subjected to human AIF-specific siRNA revealed that only AIF imported into mitochondria could repress SGs and that redox-active domains of AIF, which are dispensable for its apoptogenic action, were required for SG inhibition. In response to oxidative stress, AIF-negative cells were found to deplete non-oxidized glutathione more rapidly than AIF-expressing cells. Exogenous supplementation of glutathione inhibited SG formation elicited by arsenate or G3BP. Together, these data suggest that the oxidoreductase function of AIF is required for the maintenance of glutathione levels in stress conditions and that glutathione is a major regulator of SG.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Flavoproteins/metabolism , Glutathione/analogs & derivatives , Membrane Proteins/metabolism , Organelles/metabolism , Protein Transport/physiology , Acetylcysteine/toxicity , Apoptosis/drug effects , Apoptosis Inducing Factor , Arsenates/toxicity , DNA Helicases , Glutathione/metabolism , Glutathione/toxicity , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Organelles/drug effects , Oxidation-Reduction/drug effects , Poly-ADP-Ribose Binding Proteins , Protein Transport/drug effects , RNA Helicases , RNA Recognition Motif Proteins , RNA, Small Interfering/metabolism
5.
Oncogene ; 23(8): 1514-21, 2004 Feb 26.
Article in English | MEDLINE | ID: mdl-14716299

ABSTRACT

Cyclophilin A (CypA) was determined to interact with apoptosis-inducing factor (AIF) by mass spectroscopy, coimmunoprecipitation, pull-down assays, and molecular modeling. During the initial, caspase-independent stage of chromatin condensation that accompanies apoptosis, AIF and CypA were found to coimmunolocalize in the nucleus. Recombinant AIF and CypA proteins synergized in vitro in the degradation of plasmid DNA, as well as in the capacity to induce DNA loss in purified nuclei. The apoptogenic cooperation between AIF and CypA did not rely on the CypA peptidyl-prolyl cis-trans isomerase activity. In Cyp-expressing cells, AIF overexpression augmented apoptotic chromatinolysis. The AIF-dependent large-scale DNA fragmentation was less pronounced in CypA knockout cells as compared to controls. AIF mutants lacking the CypA-binding domain were inefficient apoptosis sensitizers in transfection experiments. Moreover, AIF failed to sensitize CypA knockout cells to apoptosis induction, and this defect in the AIF response was reversed by reintroduction of the CypA gene into CypA-deficient cells. In summary, AIF and CypA collaborate in chromatinolysis.


Subject(s)
Apoptosis/drug effects , Chromatin/metabolism , Cyclophilin A/metabolism , Flavoproteins/metabolism , Membrane Proteins/metabolism , Apoptosis Inducing Factor , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Culture Media, Serum-Free , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , HeLa Cells , Humans , Jurkat Cells , Luminescent Proteins/metabolism , Mass Spectrometry , Models, Molecular , Recombinant Proteins/metabolism , Sequence Deletion , Staurosporine/pharmacology , Vimentin/metabolism
6.
Cancer Res ; 63(23): 8233-40, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14678980

ABSTRACT

Heat shock protein 70 (HSP70) inhibits apoptosis and thereby increases the survival of cells exposed to a wide range of lethal stimuli. HSP70 has also been shown to increase the tumorigenicity of cancer cells in rodent models. The protective function of this chaperone involves interaction and neutralization of the caspase activator apoptotic protease activation factor-1 and the mitochondrial flavoprotein apoptosis-inducing factor (AIF). In this work, we determined by deletional mutagenesis that a domain of AIF comprised between amino acids 150 and 228 is engaged in a molecular interaction with the substrate-binding domain of HSP70. Computer calculations favored this conclusion. On the basis of this information, we constructed an AIF-derived protein, which is cytosolic, noncytotoxic, yet maintains its capacity to interact with HSP70. This protein, designated ADD70, sensitized different human cancer cells to apoptosis induced by a variety of death stimuli by its capacity to interact with HSP70 and therefore to sequester HSP70. Thus, its chemosensitizing effect was lost in cells in which inducible HSP70 genes had been deleted. These data delineate a novel strategy for the selective neutralization of HSP70.


Subject(s)
Flavoproteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Apoptosis/physiology , Apoptosis Inducing Factor , Caspase 3 , Caspase 9 , Caspases/metabolism , Computer Simulation , Flavoproteins/genetics , Green Fluorescent Proteins , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Membrane Proteins/genetics , Models, Molecular , Mutagenesis , Peptide Mapping , Protein Conformation , Protein Structure, Tertiary , Transfection
7.
Oncogene ; 22(43): 6669-78, 2003 Oct 02.
Article in English | MEDLINE | ID: mdl-14555980

ABSTRACT

Heat shock protein 70 (HSP70) can inhibit apoptosis by neutralizing and interacting with apoptosis-inducing factor (AIF), a mitochondrial flavoprotein that translocates upon apoptosis induction to the nucleus, via the cytosol. Here, we show that only members of the HSP70 family interact with AIF. Systematic deletion mapping revealed the existence of three distinct functional regions in the AIF protein: (1) a region between amino acids 150 and 228 that binds HSP70, (2) a domain between residues 367 and 459 that includes a nuclear localization sequence (NLS) and (3) a C-terminal domain beyond residue 567 required for its chromatin-condensing activity. Deletion of the 150-268 domain completely abolished HSP70 binding and facilitated the nuclear import of AIF, resulting in a gain-of-function phenotype with enhanced AIF-mediated chromatin condensation as compared to wild-type AIF. This gain-of-function phenotype was observed in wild-type control cells (which express low but significant levels of HSP70), yet was lost when AIFDelta150-268 was introduced into HSP70 knockout cells, underscoring the functional importance of the AIF-HSP70 interaction. Altogether, our data demonstrate that AIF inhibition by HSP70 involves cytosolic retention of AIF. Moreover, it appears that endogenous HSP70 protein levels are sufficiently elevated to modulate the lethal action of AIF.


Subject(s)
Active Transport, Cell Nucleus , Apoptosis , Flavoproteins/chemistry , HSP70 Heat-Shock Proteins/physiology , Membrane Proteins/chemistry , Animals , Apoptosis Inducing Factor , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , DNA, Complementary/metabolism , Flavoproteins/metabolism , Green Fluorescent Proteins , HSP70 Heat-Shock Proteins/metabolism , Immunoblotting , Luminescent Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitochondria/metabolism , Models, Genetic , Peptides/chemistry , Phenotype , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/metabolism , Subcellular Fractions
8.
Cell Cycle ; 2(6): 579-84, 2003.
Article in English | MEDLINE | ID: mdl-14512773

ABSTRACT

Stress or heat shock proteins (HSPs) such as HSP27 and HSP70 are expressed in response to a wide variety of physiological and environmental insults including heat, reactive oxygen species or anticancer drugs. Their overexpression allows cells to survive to otherwise lethal conditions. Several different mechanisms may account for the cytoprotective activity of HSP27 and HSP70. First, both proteins are powerful chaperones. Second, both inhibit key effectors of the apoptotic machinery including the apoptosome, the caspase activation complex (both HSP27 and HSP70), and apoptosis inducing factor (only HSP70). Third, they both play a role in the proteasome-mediated degradation of apoptosis-regulatory proteins. HSP27 and HSP70 may participate in oncogenesis, as suggested by the fact that overexpression of heat shock proteins can increase the tumorigenic potential of tumor cells. The down-regulation or selective inhibition of HSP70 might constitute a valuable strategy for the treatment of cancer.


Subject(s)
Apoptosis/physiology , HSP70 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Animals , Caspases/metabolism , Cysteine Endopeptidases/metabolism , Enzyme Activation , HSP70 Heat-Shock Proteins/genetics , Humans , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex
9.
J Neurochem ; 86(2): 306-17, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12871572

ABSTRACT

Apoptosis-inducing factor (AIF) triggers apoptosis in a caspase-independent manner. Here we report for the first time involvement of AIF in neuronal death induced by cerebral ischemia. Unilateral cerebral hypoxia-ischemia (HI) was induced in 7-day-old rats by ligation of the left carotid artery and hypoxia (7.7% O2) for 55 min. AIF release from mitochondria and AIF translocation to nuclei was detected immediately after HI, and only in damaged areas, as judged by the concurrent loss of MAP-2. AIF release was detected earlier than that of cytochrome c. Cells with AIF-positive nuclei displayed nuclear condensation and signs of DNA damage. The number of AIF-positive nuclei showed a positive correlation with the infarct volume 72 h post-HI, and this was not changed by treating the animals with boc-Asp-fmk (BAF), a multicaspase inhibitor. BAF treatment reduced the activity of caspase-3, -2 and -9 (78, 73 and 33%, respectively), and prevented caspase-dependent fodrin cleavage in vivo, but did not affect AIF release from mitochondria or the frequency of positive nuclear AIF or DNA damage 72 h post-HI, indicating that these processes occurred in a caspase-independent fashion. In summary, AIF-mediated cell death may be an important mechanism of HI-induced neuronal loss in the immature brain.


Subject(s)
Apoptosis , Brain/metabolism , Flavoproteins/metabolism , Hypoxia-Ischemia, Brain/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis Inducing Factor , Brain/drug effects , Brain/pathology , Brain Infarction/complications , Brain Infarction/pathology , Carrier Proteins/metabolism , Caspase Inhibitors , Caspases/metabolism , Cell Count , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/metabolism , Disease Models, Animal , Female , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/pathology , Male , Microfilament Proteins/metabolism , Mitochondria/metabolism , Neurons/pathology , Rats , Rats, Wistar
10.
Am J Respir Cell Mol Biol ; 28(3): 339-46, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12594060

ABSTRACT

Proteinase-activated receptor (PAR)-2 is cleaved within its aminoterminal extracellular domain by serine proteinases such as trypsin, unmasking a new aminoterminus starting with the sequence SLIGKV, which binds intramolecularly and activates the receptor. PAR-2 has been reported to be involved in inflammation within the lungs. We show that PAR-2 is expressed not only by human alveolar (A549), but also by bronchial (16HBE) epithelial cell lines, using RT-PCR and flow cytometry with a PAR-2 antibody whose epitope maps over the trypsin cleavage site. PAR-2 activation by trypsin and by the activating peptide SLIGKV-NH(2) leads to intracellular calcium mobilization in both lung epithelial cells. During lung inflammation, airspaces are burdened by neutrophils that release elastase and cathepsin G, two serine proteinases. We demonstrate that these proteinases do not activate PAR-2, but rather disarm the receptor, preventing activation by trypsin but not by SLIGKV-NH(2). Preincubation of a PAR-2-transfected cell line, as well as 16HBE and A549 cells, with either proteinase led to the disappearance of the cleavage/activation epitope recognized by the PAR-2 antibody. We hypothesize that elastase and cathepsin G disarm PAR-2 by proteolysis of the extracellular domain downstream from the trypsin cleavage/activation site, while leaving unmodified the SLIGKV-NH(2)-binding site. These findings suggest that the neutrophil serine proteinases may play a role in PAR-2-mediated lung inflammation.


Subject(s)
Epithelial Cells/metabolism , Neutrophils/enzymology , Receptors, Thrombin/physiology , Serine Endopeptidases/physiology , Cathepsin G , Cathepsins/physiology , Cell Line , Enzyme Activation , Epithelial Cells/enzymology , Humans , Leukocyte Elastase/physiology , Neutrophil Activation , Neutrophils/physiology , Peptide Fragments/metabolism , RNA, Messenger/analysis , Receptor, PAR-2 , Serine Endopeptidases/pharmacology , Tumor Cells, Cultured
11.
J Cell Sci ; 115(Pt 24): 4727-34, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12432061

ABSTRACT

Numerous pro-apoptotic signal transducing molecules act on mitochondria and provoke the permeabilization of the outer mitochondrial membrane, thereby triggering the release of potentially toxic mitochondrial proteins. One of these proteins, apoptosis-inducing factor (AIF), is a phylogenetically old flavoprotein which, in healthy cells, is confined to the mitochondrial intermembrane space. Upon lethal signaling, AIF translocates, via the cytosol, to the nucleus where it binds to DNA and provokes caspase-independent chromatin condensation. The crystal structures of both human and mouse AIF have been determined, and the fine mechanisms accounting for its oxidoreductase activity and its electrostatic interaction with double-stranded DNA have been elucidated. Importantly, the apoptogenic and oxidoreductase functions of AIF can be dissociated. Thus, mutations that abolish the AIF-DNA interaction suppress AIF-induced chromatin condensation, yet have no effect on the NADH oxidase activity. Recent studies suggest AIF to be a major factor determining caspase-independent neuronal death, emphasizing the central role of mitochondria in the control of physiological and pathological cell demise.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Flavoproteins/physiology , Membrane Proteins/physiology , Animals , Apoptosis Inducing Factor , Flavoproteins/chemistry , Flavoproteins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phylogeny
12.
Nat Struct Biol ; 9(9): 680-4, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12198487

ABSTRACT

The execution of apoptosis or programmed cell death comprises both caspase-dependent and caspase-independent processes. Apoptosis inducing factor (AIF) was identified as a major player in caspase-independent cell death. It induces chromatin condensation and initial DNA cleavage via an unknown molecular mechanism. Here we report the crystal structure of human AIF at 1.8 A resolution. The structure reveals the presence of a strong positive electrostatic potential at the AIF surface, although the calculated isoelectric point for the entire protein is neutral. We show that recombinant AIF interacts with DNA in a sequence-independent manner. In addition, in cells treated with an apoptotic stimulus, endogenous AIF becomes co-localized with DNA at an early stage of nuclear morphological changes. Structure-based mutagenesis shows that DNA-binding defective mutants of AIF fail to induce cell death while retaining nuclear translocation. The potential DNA-binding site identified from mutagenesis also coincides with computational docking of a DNA duplex. These observations suggest that AIF-induced nuclear apoptosis requires a direct interaction with DNA.


Subject(s)
Apoptosis/physiology , DNA/metabolism , Flavoproteins/physiology , Membrane Proteins/physiology , Animals , Apoptosis Inducing Factor , Cells, Cultured , Crystallography, X-Ray , Flavoproteins/chemistry , Membrane Proteins/chemistry , Mice , Protein Binding , Protein Conformation , Static Electricity
13.
Biochimie ; 84(2-3): 215-22, 2002.
Article in English | MEDLINE | ID: mdl-12022952

ABSTRACT

Apoptosis-inducing factor (AIF) is a phylogenetically ancient mitochondrial intermembrane flavoprotein endowed with the unique capacity to induce caspase-independent peripheral chromatin condensation and large-scale DNA fragmentation when added to purified nuclei. In addition to its apoptogenic activity on nuclei, AIF can also participate in the regulation of apoptotic mitochondrial membrane permeabilization and exhibits an NADH oxidase activity. Under normal circumstances, AIF is secluded behind the outer mitochondrial membrane. However, upon apoptosis induction AIF translocates to the cytosol and the nucleus. Injection of anti-AIF antibodies or knockout of the AIF gene have demonstrated that AIF may be required for cell death occurring in response to some stimuli. In particular, inactivation of AIF renders embryonic stem cells resistant to cell death following growth factor withdrawal. Moreover, AIF is essential for programmed cell death during cavitation of embryoid bodies, the very first wave of (caspase-independent) cell death indispensable for mouse morphogenesis. We have recently found that AIF is neutralized by heat-shock protein (HSP) 70, in a reaction that appears to be independent of ATP or the ATP-binding domain (ABD) of HSP70 and thus differs from the previously described Apaf-1/HSP70 interaction (which requires ATP and the HSP70 ABD). Intriguingly, HSP70 lacking ABD (HSP70 Delta ABD) inhibits apoptosis induced by serum withdrawal, staurosporin, and menadione, three models of apoptosis which are also affected by micro-injection of anti-AIF antibody or genetic ablation of AIF. Altogether, these data suggest that AIF plays a role in the regulation of caspase-independent cell death.


Subject(s)
Apoptosis/physiology , Flavoproteins/physiology , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/physiology , Mitochondria/physiology , Animals , Apoptosis Inducing Factor , Apoptotic Protease-Activating Factor 1 , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Humans , Proteins
SELECTION OF CITATIONS
SEARCH DETAIL