Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Curr Biol ; 25(19): 2479-92, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26387716

ABSTRACT

Autophagy plays key roles in development, oncogenesis, cardiovascular, metabolic, and neurodegenerative diseases. Hence, understanding how autophagy is regulated can reveal opportunities to modify autophagy in a disease-relevant manner. Ideally, one would want to functionally define autophagy regulators whose enzymatic activity can potentially be modulated. Here, we describe the STK38 protein kinase (also termed NDR1) as a conserved regulator of autophagy. Using STK38 as bait in yeast-two-hybrid screens, we discovered STK38 as a novel binding partner of Beclin1, a key regulator of autophagy. By combining molecular, cell biological, and genetic approaches, we show that STK38 promotes autophagosome formation in human cells and in Drosophila. Upon autophagy induction, STK38-depleted cells display impaired LC3B-II conversion; reduced ATG14L, ATG12, and WIPI-1 puncta formation; and significantly decreased Vps34 activity, as judged by PI3P formation. Furthermore, we observed that STK38 supports the interaction of the exocyst component Exo84 with Beclin1 and RalB, which is required to initiate autophagosome formation. Upon studying the activation of STK38 during autophagy induction, we found that STK38 is stimulated in a MOB1- and exocyst-dependent manner. In contrast, RalB depletion triggers hyperactivation of STK38, resulting in STK38-dependent apoptosis under prolonged autophagy conditions. Together, our data establish STK38 as a conserved regulator of autophagy in human cells and flies. We also provide evidence demonstrating that STK38 and RalB assist the coordination between autophagic and apoptotic events upon autophagy induction, hence further proposing a role for STK38 in determining cellular fate in response to autophagic conditions.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Beclin-1 , Cell Line, Tumor , Cells, Cultured , Drosophila , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Protein Binding , Two-Hybrid System Techniques
2.
EMBO J ; 34(8): 1025-41, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25586377

ABSTRACT

To obtain mechanistic insights into the cross talk between lipolysis and autophagy, two key metabolic responses to starvation, we screened the autophagy-inducing potential of a panel of fatty acids in human cancer cells. Both saturated and unsaturated fatty acids such as palmitate and oleate, respectively, triggered autophagy, but the underlying molecular mechanisms differed. Oleate, but not palmitate, stimulated an autophagic response that required an intact Golgi apparatus. Conversely, autophagy triggered by palmitate, but not oleate, required AMPK, PKR and JNK1 and involved the activation of the BECN1/PIK3C3 lipid kinase complex. Accordingly, the downregulation of BECN1 and PIK3C3 abolished palmitate-induced, but not oleate-induced, autophagy in human cancer cells. Moreover, Becn1(+/-) mice as well as yeast cells and nematodes lacking the ortholog of human BECN1 mounted an autophagic response to oleate, but not palmitate. Thus, unsaturated fatty acids induce a non-canonical, phylogenetically conserved, autophagic response that in mammalian cells relies on the Golgi apparatus.


Subject(s)
Autophagy/drug effects , Fatty Acids, Unsaturated/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Beclin-1 , Caenorhabditis elegans , Cells, Cultured , Female , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , Saccharomyces cerevisiae , Up-Regulation/drug effects
3.
Methods Enzymol ; 543: 73-88, 2014.
Article in English | MEDLINE | ID: mdl-24924128

ABSTRACT

Macroautophagy (hereafter referred to as autophagy), a central mechanism mediating the lysosomal degradation of cytoplasmic components, can be stimulated by a wide panel of adverse stimuli, including a panoply of anticancer agents. The central autophagic organelle is the autophagosome, a double membrane-bound vacuole that sequesters the cytoplasmic material destined to disposal. The ultimate destiny of the autophagosome is to fuse with a lysosome, resulting in the degradation of the autophagic cargo. In this setting, it is important to discriminate whether a particular stimulus actually promotes autophagy or it simply blocks the fusion of autophagosomes with lysosomes. To this aim, the methods that assess autophagy should assess not only the number of autophagosomes but also the so-called autophagic flux, that is, the clearance of the autophagy cargo from the lysosomal compartment. Here, we present a compendium of methods to assess the autophagic flux in cultured malignant cells. This approach should allow for the study of the intimate link between autophagy and oncometabolism in several experimental paradigms.


Subject(s)
Autophagy , Neoplasms/pathology , Cell Line, Tumor , Humans , Molecular Probes , Neoplasms/immunology
4.
Autophagy ; 10(7): 1341-2, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24852146

ABSTRACT

We recently reported that BAG6/BAT3 (BCL2-associated athanogene 6) is essential for basal and starvation-induced autophagy in E18.5 bag6(-/-) mouse embryos and in mouse embryonic fibroblasts (MEFs) through the modulation of the EP300/p300-dependent acetylation of TRP53 and autophagy-related (ATG) proteins. We observed that BAG6 increases TRP53 acetylation during starvation and pro-autophagic TRP53-target gene expression. BAG6 also decreases the EP300 dependent-acetylation of ATG5, ATG7, and LC3-I, posttranslational modifications that inhibit autophagy. In addition, in the absence of BAG6 or when using a mutant of BAG6 exclusively located in the cytoplasm, autophagy is inhibited, ATG7 is hyperacetylated, TRP53 acetylation is abrogated, and EP300 accumulates in the cytoplasm indicating that BAG6 is involved in the regulation of the nuclear localization of EP300. We also reported that the interaction between BAG6 and EP300 occurs in the cytoplasm rather than the nucleus. Moreover, during starvation, EP300 is transported to the nucleus in a BAG6-dependent manner. We concluded that BAG6 regulates autophagy by controlling the localization of EP300 and its accessibility to nuclear (TRP53) and cytoplasmic (ATGs) substrates.


Subject(s)
Autophagy , E1A-Associated p300 Protein/metabolism , Intracellular Space/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Acetylation , Animals , Mice , Models, Biological , Protein Transport , Tumor Suppressor Protein p53/metabolism
5.
Proc Natl Acad Sci U S A ; 111(11): 4115-20, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24591579

ABSTRACT

Autophagy is regulated by posttranslational modifications, including acetylation. Here we show that HLA-B-associated transcript 3 (BAT3) is essential for basal and starvation-induced autophagy in embryonic day 18.5 BAT3(-/-) mouse embryos and in mouse embryonic fibroblasts (MEFs) through the modulation of p300-dependent acetylation of p53 and ATG7. Specifically, BAT3 increases p53 acetylation and proautophagic p53 target gene expression, while limiting p300-dependent acetylation of ATG7, a mechanism known to inhibit autophagy. In the absence of BAT3 or when BAT3 is located exclusively in the cytosol, autophagy is abrogated, ATG7 is hyperacetylated, p53 acetylation is abolished, and p300 accumulates in the cytosol, indicating that BAT3 regulates the nuclear localization of p300. In addition, the interaction between BAT3 and p300 is stronger in the cytosol than in the nucleus and, during starvation, the level of p300 decreases in the cytosol but increases in the nucleus only in the presence of BAT3. We conclude that BAT3 tightly controls autophagy by modulating p300 intracellular localization, affecting the accessibility of p300 to its substrates, p53 and ATG7.


Subject(s)
Autophagy/physiology , E1A-Associated p300 Protein/metabolism , Embryo, Mammalian/physiology , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Autophagy/genetics , Autophagy-Related Protein 7 , Cell Fractionation , Cell Nucleus/metabolism , Cytosol/metabolism , DNA Primers/genetics , Embryo, Mammalian/metabolism , Immunoprecipitation , Mice , Mice, Knockout , Molecular Chaperones/genetics , Nuclear Proteins/genetics , Real-Time Polymerase Chain Reaction
6.
Mol Cell ; 53(5): 710-25, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24560926

ABSTRACT

Acetyl-coenzyme A (AcCoA) is a major integrator of the nutritional status at the crossroads of fat, sugar, and protein catabolism. Here we show that nutrient starvation causes rapid depletion of AcCoA. AcCoA depletion entailed the commensurate reduction in the overall acetylation of cytoplasmic proteins, as well as the induction of autophagy, a homeostatic process of self-digestion. Multiple distinct manipulations designed to increase or reduce cytosolic AcCoA led to the suppression or induction of autophagy, respectively, both in cultured human cells and in mice. Moreover, maintenance of high AcCoA levels inhibited maladaptive autophagy in a model of cardiac pressure overload. Depletion of AcCoA reduced the activity of the acetyltransferase EP300, and EP300 was required for the suppression of autophagy by high AcCoA levels. Altogether, our results indicate that cytosolic AcCoA functions as a central metabolic regulator of autophagy, thus delineating AcCoA-centered pharmacological strategies that allow for the therapeutic manipulation of autophagy.


Subject(s)
Acetyl Coenzyme A/chemistry , Autophagy , Cytosol/enzymology , Gene Expression Regulation, Enzymologic , Adenosine Triphosphate/chemistry , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , E1A-Associated p300 Protein/chemistry , Green Fluorescent Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , Ketoglutaric Acids/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitochondria/metabolism , RNA, Small Interfering/metabolism
7.
Autophagy ; 9(6): 850-60, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23575388

ABSTRACT

Amino acids, leucine in particular, are known to inhibit autophagy, at least in part by their ability to stimulate MTOR-mediated signaling. Evidence is presented showing that glutamate dehydrogenase, the central enzyme in amino acid catabolism, contributes to leucine sensing in the regulation of autophagy. The data suggest a dual mechanism by which glutamate dehydrogenase activity modulates autophagy, i.e., by activating MTORC1 and by limiting the formation of reactive oxygen species.


Subject(s)
Autophagy/drug effects , Glutamate Dehydrogenase/metabolism , Leucine/pharmacology , Gene Knockdown Techniques , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Valine/pharmacology
8.
Autophagy ; 9(5): 714-29, 2013 May.
Article in English | MEDLINE | ID: mdl-23519090

ABSTRACT

Breast cancer tissue contains a small population of cells that have the ability to self-renew; these cells are known as cancer stem-like cells (CSCs). We have recently shown that autophagy is essential for the tumorigenicity of these CSCs. Salinomycin (Sal), a K (+) /H (+) ionophore, has recently been shown to be at least 100 times more effective than paclitaxel in reducing the proportion of breast CSCs. However, its mechanisms of action are still unclear. We show here that Sal blocked both autophagy flux and lysosomal proteolytic activity in both CSCs and non-CSCs derived from breast cancer cells. GFP-LC3 staining combined with fluorescent dextran uptake and LysoTracker-Red staining showed that autophagosome/lysosome fusion was not altered by Sal treatment. Acridine orange staining provided evidence that lysosomes display the characteristics of acidic compartments in Sal-treated cells. However, tandem mCherry-GFP-LC3 assay indicated that the degradation of mCherry-GFP-LC3 is blocked by Sal. Furthermore, the protein degradation activity of lysosomes was inhibited, as demonstrated by the rate of long-lived protein degradation, DQ-BSA assay and measurement of cathepsin activity. Our data indicated that Sal has a relatively greater suppressant effect on autophagic flux in the ALDH (+) population in HMLER cells than in the ALDH (-) population; moreover, this differential effect on autophagic flux correlated with an increase in apoptosis in the ALDH (+) population. ATG7 depletion accelerated the proapoptotic capacity of Sal in the ALDH (+) population. Our findings provide new insights into how the autophagy-lysosomal pathway contributes to the ability of Sal to target CSCs in vitro.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Pyrans/pharmacology , Acridine Orange/metabolism , Aldehyde Dehydrogenase/metabolism , Apoptosis/drug effects , Autophagy-Related Protein 7 , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Green Fluorescent Proteins/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , MCF-7 Cells , Membrane Fusion/drug effects , Microtubule-Associated Proteins/metabolism , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Phagosomes/drug effects , Phagosomes/metabolism , Proteolysis/drug effects , Recombinant Fusion Proteins/metabolism , Staining and Labeling , Ubiquitin-Activating Enzymes/metabolism
9.
Cell Res ; 23(4): 508-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23337583

ABSTRACT

Lysosome is a key subcellular organelle in the execution of the autophagic process and at present little is known whether lysosomal function is controlled in the process of autophagy. In this study, we first found that suppression of mammalian target of rapamycin (mTOR) activity by starvation or two mTOR catalytic inhibitors (PP242 and Torin1), but not by an allosteric inhibitor (rapamycin), leads to activation of lysosomal function. Second, we provided evidence that activation of lysosomal function is associated with the suppression of mTOR complex 1 (mTORC1), but not mTORC2, and the mTORC1 localization to lysosomes is not directly correlated to its regulatory role in lysosomal function. Third, we examined the involvement of transcription factor EB (TFEB) and demonstrated that TFEB activation following mTORC1 suppression is necessary but not sufficient for lysosomal activation. Finally, Atg5 or Atg7 deletion or blockage of the autophagosome-lysosome fusion process effectively diminished lysosomal activation, suggesting that lysosomal activation occurring in the course of autophagy is dependent on autophagosome-lysosome fusion. Taken together, this study demonstrates that in the course of autophagy, lysosomal function is upregulated via a dual mechanism involving mTORC1 suppression and autophagosome-lysosome fusion.


Subject(s)
Autophagy/drug effects , Lysosomes/drug effects , Phagosomes/drug effects , TOR Serine-Threonine Kinases/genetics , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/agonists , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Indoles/pharmacology , Lysosomes/genetics , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Membrane Fusion/drug effects , Membrane Fusion/genetics , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Naphthyridines/pharmacology , Phagosomes/genetics , Purines/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
10.
Neurobiol Aging ; 34(3): 770-90, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22892312

ABSTRACT

Using cultured cortical neurons, we show that the blockade of protein phosphatase 2A (PP2A), either pharmacologically by okadaic acid or by short hairpin RNA (shRNA)-mediated silencing of PP2A catalytic subunit, inhibited basal autophagy and autophagy induced in several experimental settings (including serum deprivation, endoplasmic reticulum stress, rapamycin, and proteasome inhibition) at early stages before autophagosome maturation. Conversely, PP2A upregulation by PP2A catalytic subunit overexpression stimulates neuronal autophagy. In addition, PP2A blockade resulted in the activation of the negative regulator of autophagy mammalian target of rapamycin complex 1 and 5' adenosine monophosphate (AMP)-activated protein kinase (AMPK) and led to intraneuronal accumulation of p62- and ubiquitin-positive protein inclusions, likely due to autophagy downregulation. These data are consistent with previous findings showing that specific invalidation of the autophagy process in the nervous system of mouse resulted in the accumulation of p62- and ubiquitin-positive protein inclusion bodies. Furthermore, we showed that PP2A inhibition alters the distribution of the microtubule-associated protein 1 light chain(LC) 3-I (MAP LC3-I), a key component of the autophagy molecular machinery. Whether MAP LC3-I distribution in the cell accounts for autophagy regulation remains to be determined. These data are important to human neurodegenerative diseases, especially Alzheimer's disease, because they provide links for the first time between the pathological features of Alzheimer's disease:PP2A downregulation, autophagy disruption, and protein aggregation.


Subject(s)
Autophagy/physiology , Neurons/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Ubiquitinated Proteins/metabolism , Animals , Cells, Cultured , Gene Silencing , Microtubule-Associated Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , RNA, Small Interfering , Rats , Rats, Wistar
11.
J Biol Chem ; 285(33): 25570-81, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20529838

ABSTRACT

Gossypol, a natural Bcl-2 homology domain 3 mimetic compound isolated from cottonseeds, is currently being evaluated in clinical trials. Here, we provide evidence that gossypol induces autophagy followed by apoptotic cell death in both the MCF-7 human breast adenocarcinoma and HeLa cell lines. We first show that knockdown of the Bcl-2 homology domain 3-only protein Beclin 1 reduces gossypol-induced autophagy in MCF-7 cells, but not in HeLa cells. Gossypol inhibits the interaction between Beclin 1 and Bcl-2 (B-cell leukemia/lymphoma 2), antagonizes the inhibition of autophagy by Bcl-2, and hence stimulates autophagy. We then show that knockdown of Vps34 reduces gossypol-induced autophagy in both cell lines, and consistent with this, the phosphatidylinositol 3-phosphate-binding protein WIPI-1 is recruited to autophagosomal membranes. Further, Atg5 knockdown also reduces gossypol-mediated autophagy. We conclude that gossypol induces autophagy in both a canonical and a noncanonical manner. Notably, we found that gossypol-mediated apoptotic cell death was potentiated by treatment with the autophagy inhibitor wortmannin or with small interfering RNA against essential autophagy genes (Vps34, Beclin 1, and Atg5). Our findings support the notion that gossypol-induced autophagy is cytoprotective and not part of the cell death process induced by this compound.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Gossypol/pharmacology , Membrane Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Androstadienes/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Autophagy-Related Protein 5 , Beclin-1 , Blotting, Western , Cell Line, Tumor , Contraceptive Agents, Male/pharmacology , Gossypol/chemistry , HeLa Cells , Humans , Immunoprecipitation , Membrane Proteins/genetics , Microscopy, Electron , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Wortmannin
12.
J Biol Chem ; 285(14): 10850-61, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20123989

ABSTRACT

A group of phosphoinositide 3-kinase (PI3K) inhibitors, such as 3-methyladenine (3-MA) and wortmannin, have been widely used as autophagy inhibitors based on their inhibitory effect on class III PI3K activity, which is known to be essential for induction of autophagy. In this study, we systematically examined and compared the effects of these two inhibitors on autophagy under both nutrient-rich and deprivation conditions. To our surprise, 3-MA is found to promote autophagy flux when treated under nutrient-rich conditions with a prolonged period of treatment, whereas it is still capable of suppressing starvation-induced autophagy. We first observed that there are marked increases of the autophagic markers in cells treated with 3-MA in full medium for a prolonged period of time (up to 9 h). Second, we provide convincing evidence that the increase of autophagic markers is the result of enhanced autophagic flux, not due to suppression of maturation of autophagosomes or lysosomal function. More importantly, we found that the autophagy promotion activity of 3-MA is due to its differential temporal effects on class I and class III PI3K; 3-MA blocks class I PI3K persistently, whereas its suppressive effect on class III PI3K is transient. Because 3-MA has been widely used as an autophagy inhibitor in the literature, understanding the dual role of 3-MA in autophagy thus suggests that caution should be exercised in the application of 3-MA in autophagy study.


Subject(s)
Adenine/analogs & derivatives , Autophagy , Embryo, Mammalian/drug effects , Fibroblasts/drug effects , Phosphoinositide-3 Kinase Inhibitors , Adenine/pharmacology , Androstadienes/pharmacology , Animals , Autophagy-Related Protein 7 , Blotting, Western , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Fibroblasts/cytology , Fibroblasts/enzymology , Immunoprecipitation , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Phosphatidylinositol 3-Kinases/classification , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphodiesterase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Wortmannin
13.
Cancer Res ; 69(24): 9346-53, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19934311

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite involved in cancer development through stimulation of cell survival, proliferation, migration, and angiogenesis. Irreversible degradation of S1P is catalyzed by S1P lyase (SPL). The human SGPL1 gene that encodes SPL maps to a region often mutated in cancers. To investigate the effect of SPL deficiency on cell survival and transformation, the susceptibility to anticancer drugs of fibroblasts generated from SPL-deficient mouse embryos (Sgpl1(-/-)) was compared with that of cells from heterozygous (Sgpl1(+/-)) or wild-type (Sgpl1(+/+)) embryos. First, loss of SPL caused resistance to the toxic effects of etoposide and doxorubicin. Interestingly, heterozygosity for the Sgpl1 gene resulted in partial resistance to apoptosis. Secondly, doxorubicin-induced apoptotic signaling was strongly inhibited in Sgpl1(-/-) cells (phosphatidylserine externalization, caspase activation, and cytochrome c release). This was accompanied by a strong increase in Bcl-2 and Bcl-xL protein content. Whereas correction of SPL deficiency in Sgpl1(-/-) cells led to downregulation of antiapoptotic proteins, Bcl-2 and Bcl-xL small interfering RNA-mediated knockdown in SPL-deficient cells resulted in increased sensitivity to doxorubicin, suggesting that Bcl-2 upregulation mediates SPL protective effects. Moreover, SPL deficiency led to increased cell proliferation, anchorage-independent cell growth, and formation of tumors in nude mice. Finally, transcriptomic studies showed that SPL expression is downregulated in human melanoma cell lines. Thus, by affecting S1P metabolism and the expression of Bcl-2 members, the loss of SPL enhances cell resistance to anticancer regimens and results in an increased ability of cells to acquire a transformed phenotype and become malignant.


Subject(s)
Aldehyde-Lyases/deficiency , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-X Protein/biosynthesis , Aldehyde-Lyases/biosynthesis , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Down-Regulation , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Dosage , Humans , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Up-Regulation , bcl-X Protein/genetics
14.
Autophagy ; 5(4): 558-60, 2009 May.
Article in English | MEDLINE | ID: mdl-19337026

ABSTRACT

Ceramide is a sphingolipid bioactive molecule that induces apoptosis and other forms of cell death, and triggers macroautophagy (referred to below as autophagy). Like amino acid starvation, ceramide triggers autophagy by interfering with the mTOR-signaling pathway, and by dissociating the Beclin 1:Bcl-2 complex in a c-Jun N-terminal kinase 1 (JNK1)-mediated Bcl-2 phosphorylation-dependent manner. Dissociation of the Beclin 1:Bcl-2 complex, and the subsequent stimulation of autophagy have been observed in various contexts in which the cellular level of long-chain ceramides was increased. It is notable that the conversion of short-chain ceramides (C(2)-ceramide and C(6)-ceramide) into long-chain ceramide via the activity of ceramide synthase is required to trigger autophagy. The dissociation of the Beclin 1:Bcl-2 complex has also been observed in response to tamoxifen and PDMP (an inhibitor of the enzyme that converts ceramide to glucosylceramide), drugs that increase the intracellular level of long-chain ceramides. However, and in contrast to starvation, overexpression of Bcl-2 does not blunt ceramide-induced autophagy. Whether this autophagy that is unchecked by forced dissociation of the Beclin 1:Bcl-2 complex is related to the ability of ceramide to trigger cell death remains an open question. More generally, the question of whether ceramide-induced autophagy is a dedicated cell death mechanism deserves closer scrutiny.


Subject(s)
Autophagy/drug effects , Ceramides/pharmacology , Cytoprotection/drug effects , Humans , Models, Biological
15.
Methods Enzymol ; 452: 47-61, 2009.
Article in English | MEDLINE | ID: mdl-19200875

ABSTRACT

Macroautophagy is a three-step process: (1) autophagosomes form and mature, (2) the autophagosomes fuse with lysosomes, and (3) the autophagic cargo is degraded in the lysosomes. It is this lysosomal degradation of the autophagic cargo that constitutes the autophagic flux. As in the case of metabolic pathways, the steady-state concentration of the intermediary autophagic structures alone is insufficient for investigating the flux. Assaying the degradation of long-lived proteins as described in this chapter is one of the methods that can be used to measure autophagic flux.


Subject(s)
Autophagy/physiology , Biological Assay/methods , Proteins/metabolism , Animals , Cells, Cultured , HT29 Cells , Hepatocytes/metabolism , Humans , Lysosomes/metabolism , Rats
16.
J Biol Chem ; 284(5): 2719-2728, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19029119

ABSTRACT

Macroautophagy is a vacuolar lysosomal catabolic pathway that is stimulated during periods of nutrient starvation to preserve cell integrity. Ceramide is a bioactive sphingolipid associated with a large range of cell processes. Here we show that short-chain ceramides (C(2)-ceramide and C(6)-ceramide) and stimulation of the de novo ceramide synthesis by tamoxifen induce the dissociation of the complex formed between the autophagy protein Beclin 1 and the anti-apoptotic protein Bcl-2. This dissociation is required for macroautophagy to be induced either in response to ceramide or to starvation. Three potential phosphorylation sites, Thr(69), Ser(70), and Ser(87), located in the non-structural N-terminal loop of Bcl-2, play major roles in the dissociation of Bcl-2 from Beclin 1. We further show that activation of c-Jun N-terminal protein kinase 1 by ceramide is required both to phosphorylate Bcl-2 and to stimulate macroautophagy. These findings reveal a new aspect of sphingolipid signaling in up-regulating a major cell process involved in cell adaptation to stress.


Subject(s)
Autophagy/physiology , Ceramides/physiology , Mitogen-Activated Protein Kinase 8/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Humans , Immunoprecipitation , Phosphorylation
17.
Autophagy ; 3(4): 390-2, 2007.
Article in English | MEDLINE | ID: mdl-17471012

ABSTRACT

The NF-kappaB transcription factor is an important anti-apoptotic factor, which is frequently deregulated in cancer cells. We have recently demonstrated that NF-kappaB activation mediates the repression of autophagy in response to TNFa in three models of cancer cell lines. In contrast, in the absence of NF-kappaB activation, TNFa induces macroautophagy (autophagy), which requires reactive oxygen species (ROS) production and participates in the TNFalpha-induced apoptotic signaling pathway. Autophagy-dependent apoptosis was also observed following direct addition of ROS to cells. Moreover, addition of rapamycin to TNFalpha renders these cells susceptible to the cytotoxic effect of this cytokine. These findings highlight the regulation of autophagy by oxidative stress and support the idea that repression of autophagy by NF-kappaB may constitute a novel anti-apoptotic function of this transcription factor. We also bring evidence that direct stimulation of autophagy may represent a new therapeutic strategy for overcoming the NF-kappaB-dependent chemoresistance of cancer cells.


Subject(s)
Autophagy/physiology , Gene Expression Regulation/physiology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Apoptosis/physiology , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Models, Biological , Tumor Necrosis Factor-alpha/pharmacology
18.
J Biol Chem ; 281(46): 34870-9, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16990266

ABSTRACT

Interruption of mTOR-dependent signaling by rapamycin is known to stimulate autophagy, both in mammalian cells and in yeast. Because activation of AMPK also inhibits mTOR-dependent signaling one would expect stimulation of autophagy by AMPK activation. According to the literature, this is true for yeast but, unexpectedly, not for mammalian cells on the basis of the use of AICAR, a pharmacological activator of AMPK. In the present study, carried out with hepatocytes, HT-29 cells, and HeLa cells, we have reexamined the possible role of AMPK in the control of mammalian autophagy. Inhibition of AMPK activity by compound C or by transfection with a dominant negative form of AMPK almost completely inhibited autophagy. These results suggest that the inhibition of autophagy by AICAR is not related to its ability to activate AMPK. We conclude that in mammalian cells, as in yeast, AMPK is required for autophagy.


Subject(s)
Autophagy/physiology , Hepatocytes/metabolism , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Autophagy/drug effects , HT29 Cells , HeLa Cells , Hepatocytes/drug effects , Humans , Male , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Wistar , Ribonucleotides/pharmacology
19.
J Biol Chem ; 281(41): 30373-82, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16857678

ABSTRACT

Activation of NF-kappaB and autophagy are two processes involved in the regulation of cell death, but the possible cross-talk between these two signaling pathways is largely unknown. Here, we show that NF-kappaB activation mediates repression of autophagy in tumor necrosis factor-alpha (TNFalpha)-treated Ewing sarcoma cells. This repression is associated with an NF-kappaB-dependent activation of the autophagy inhibitor mTOR. In contrast, in cells lacking NF-kappaB activation, TNFalpha treatment up-regulates the expression of the autophagy-promoting protein Beclin 1 and subsequently induces the accumulation of autophagic vacuoles. Both of these responses are dependent on reactive oxygen species (ROS) production and can be mimicked in NF-kappaB-competent cells by the addition of H2O2. Small interfering RNA-mediated knockdown of beclin 1 and atg7 expression, two autophagy-related genes, reduced TNFalpha- and reactive oxygen species-induced apoptosis in cells lacking NF-kappaB activation and in NF-kappaB-competent cells, respectively. These findings demonstrate that autophagy may amplify apoptosis when associated with a death signaling pathway. They are also evidence that inhibition of autophagy is a novel mechanism of the antiapoptotic function of NF-kappaB activation. We suggest that stimulation of autophagy may be a potential way bypassing the resistance of cancer cells to anti-cancer agents that activate NF-kappaB.


Subject(s)
Autophagy , NF-kappa B p50 Subunit/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Membrane Proteins/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Reactive Oxygen Species
20.
J Biol Chem ; 279(18): 18384-91, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14970205

ABSTRACT

The sphingolipid ceramide is involved in the cellular stress response. Here we demonstrate that ceramide controls macroautophagy, a major lysosomal catabolic pathway. Exogenous C(2)-ceramide stimulates macroautophagy (proteolysis and accumulation of autophagic vacuoles) in the human colon cancer HT-29 cells by increasing the endogenous pool of long chain ceramides as demonstrated by the use of the ceramide synthase inhibitor fumonisin B(1). Ceramide reverted the interleukin 13-dependent inhibition of macroautophagy by interfering with the activation of protein kinase B. In addition, C(2)-ceramide stimulated the expression of the autophagy gene product beclin 1. Ceramide is also the mediator of the tamoxifen-dependent accumulation of autophagic vacuoles in the human breast cancer MCF-7 cells. Monodansylcadaverine staining and electron microscopy showed that this accumulation was abrogated by myriocin, an inhibitor of de novo synthesis ceramide. The tamoxifen-dependent accumulation of vacuoles was mimicked by 1-phenyl-2-decanoylamino-3-morpholino-1-propanol, an inhibitor of glucosylceramide synthase. 1-Phenyl-2-decanoylamino-3-morpholino-1-propanol, tamoxifen, and C(2)-ceramide stimulated the expression of beclin 1, whereas myriocin antagonized the tamoxifen-dependent up-regulation. Tamoxifen and C(2)-ceramide interfere with the activation of protein kinase B, whereas myriocin relieved the inhibitory effect of tamoxifen. In conclusion, the control of macroautophagy by ceramide provides a novel function for this lipid mediator in a cell process with major biological outcomes.


Subject(s)
Autophagy/drug effects , Ceramides/pharmacology , Protein Biosynthesis , Protein Serine-Threonine Kinases , Proteins , Proto-Oncogene Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins , Beclin-1 , Cell Line, Tumor , Ceramides/physiology , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Humans , Membrane Proteins , Microscopy, Electron , Proto-Oncogene Proteins c-akt , Tamoxifen/pharmacology , Up-Regulation , Vacuoles
SELECTION OF CITATIONS
SEARCH DETAIL
...