Skip to main content
Genome Biology logoLink to Genome Biology
. 2002 Jul 31;3(8):research0042.1–research0042.22. doi: 10.1186/gb-2002-3-8-research0042

Identification of genes involved in ceramide-dependent neuronal apoptosis using cDNA arrays

Charles Decraene 1,4, Bernard Brugg 2, Merle Ruberg 3, Eric Eveno 1, Christiane Matingou 1, Fariza Tahi 1,5, Jean Mariani 2, Charles Auffray 1, Geneviève Pietu 1,4,
PMCID: PMC126236  PMID: 12186649

Short abstract

A cell-culture model was used to establish a profile of gene expression during the effector phase of ceramide-mediated cell death. Of the 239 genes that met the criteria for differential hybridization, 10 correspond to genes previously involved in C2-ceramide or TNF-α signaling pathways and 20 in neuronal disorders, oncogenesis or more broadly in the regulation of proliferation.

Abstract

Background

Ceramide is important in many cell responses, such as proliferation, differentiation, growth arrest and apoptosis. Elevated ceramide levels have been shown to induce apoptosis in primary neuronal cultures and neuronally differentiated PC 12 cells.

Results

To investigate gene expression during ceramide-dependent apoptosis, we carried out a global study of gene expression in neuronally differentiated PC 12 cells treated with C2-ceramide using an array of 9,120 cDNA clones. Although the criteria adopted for differential hybridization were stringent, modulation of expression of 239 genes was identified during the effector phase of C2-ceramide-induced cell death. We have made an attempt at classifying these genes on the basis of their putative functions, first with respect to known effects of ceramide or ceramide-mediated transduction systems, and then with respect to regulation of cell growth and apoptosis.

Conclusions

Our cell-culture model has enabled us to establish a profile of gene expression during the effector phase of ceramide-mediated cell death. Of the 239 genes that met the criteria for differential hybridization, 10 correspond to genes previously involved in C2-ceramide or TNF-α signaling pathways and 20 in neuronal disorders, oncogenesis or more broadly in the regulation of proliferation. The remaining 209 genes, with or without known functions, constitute a pool of genes potentially implicated in the regulation of neuronal cell death.

Background

Ceramide is an intracellular lipid second messenger generated in response to a large number of extracellular signals [1,2]. These include tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), ionizing and ultraviolet radiation, anti-cancer drugs, growth-factor withdrawal, infection by human immunodeficiency virus (HIV) or bacteria. It is reported to participate in cell differentiation [3], senescence [4], growth arrest or programmed cell death [1,2], depending on the cell type.

The role of ceramide in programmed cell death or apoptosis has been described in lymphocytes [5], macrophages [6], neurons in primary culture [7,8] and neuronally differentiated PC12 cells [9,10,11]. A number of downstream targets of ceramide have been identified. The best documented are the ceramide-activated protein phosphatases (CAPP) and the ceramide-activated protein kinase (CAPK). The former, represented by the PP1 and PP2A families, mediate the effect of ceramide on the transcription factors c-Myc [12] and c-Jun [13]. CAPK is involved in the mitogen-activated protein (MAP) kinase (MAPK) cascades that include the extracellular-signal regulated kinases (ERK), the c-Jun N-terminal kinases or stress-activated kinases (JNK/ SNK/SAPK) and the p38 family [14].

Recently, it has been shown that C2-ceramide rapidly decreases phosphorylation of ERKs, but increases p38 and JNK phosphorylation, activating the transcription factors c-Fos, c-Jun and p53, during the effector phase of apoptosis in primary cortical neurons [15]. It also regulates the protein kinase B (Akt/PKB)-dependent survival pathways, inactivating Akt by dephosphorylation and activating the Bcl-2-related protein BAD by phosphorylation [16,17,18]. Ceramide-induced apoptosis in neurons or in neuronally differentiated PC12 cells has been associated with mitochondrially produced reactive oxygen species (ROS) as well as activation and nuclear translocation of the transcription factor NFκB [10,11,19]. All these molecular events are observed during the effector phase of ceramide-induced apoptosis which also includes gene expression and new protein synthesis required for ceramide-mediated cell death, as it has been shown that neuronal cell death can be inhibited by cycloheximide [7].

The genes that are transcriptionally regulated during ceramide-mediated cell death are still poorly documented. To study gene expression during neuronal cell death, we carried out a differential screen of an array of 9,120 cDNA clones from a human infant brain library (library 1NIB [20]) with complex cDNA targets derived from neuronally differentiated rat pheocytochroma PC12 cells treated with C2-ceramide compared to control PC12 cells. This model is particularly suitable for establishing a gene-expression profile during ceramide-mediated neuronal death because first, the neuronal cell population is synchronized and homogeneous, unlike brain tissue or primary neuronal cultures, and second, because the use of exogenous C2-ceramide eliminates the risk of interference by transcripts activated by signal transducers upstream of ceramide in the cell-death pathway or in pathways activated in parallel.

Results

Cell death induced in neuronally differentiated PC12 cells by C2-ceramide

The morphological characteristics of differentiated PC12 cells after 24 hours in the presence of 25 μM C2-ceramide were compatible with cell death by apoptosis. Compared with control cultures, as viewed by phase-contrast microscopy (Figure 1a), C2-ceramide-treated cells lost their neurites and became rounded and shrunken after 24 hours of treatment (Figure 1b). The cells that remained viable in the C2-ceramide-treated cultures were refringent (Figure 1b), like those in the control cultures (Figure 1a), and excluded the vital marker propidium iodide (Figure 1c), whereas the dead cells took up propidium iodide that intercalated into their DNA (Figure 1d), revealing condensed and fragmented nuclei. As previously described, when neuronally differentiated PC12 cells or primary cultures of mesencephalic neurons were treated with cell-permeant C2-ceramide (10-50 μM), they died in a dose-dependent manner [7,10]. At 25 μM no significant cell death was observed until 12 hours after the initiation of treatment (Figure 2a). After 24 hours, 50% of the cells had died. By 48 hours, no viable cells remained. Furthermore, we observed activation of caspase-3/CPP32, a member of the cysteine-activated aspartate family of cell-death proteases [21], that started 8 hours after the beginning of ceramide treatment and was five times the control value by 18 hours (Figure 2b). No significant cell death and caspase-3/CPP32 activity were observed using the inactive C2 analog of ceramide, C2-dihydroceramide (Figure 2).

Figure 1.

Figure 1

Morphological characteristics of nerve growth factor (NGF)-differentiated PC12 cells during C2-ceramide-induced apoptosis. (a) Control cultures of PC12 cells after 6 days in the presence of NGF viewed by phase-contrast microscopy; (b) NGF-differentiated PC12 cells after 24 h treatment with 25 μM C2-ceramide. Open arrows, viable cells; white arrows, dead cells. (c,d) Condensed and fragmented nuclei of dead cells in (c) control and (d) NGF-differentiated PC12 cells visualized by intercalation of propidium iodide into DNA were viewed under epifluorescence illumination.

Figure 2.

Figure 2

Characterization of C2-ceramide-induced apoptosis. (a) Time course of cell death induced by 25 μM C2-ceramide (circles) or by 25 μM C2-dihydro-ceramide (triangles). Cells were counted in at least 10 randomly chosen fields with a 20x objective. The percentage of cells excluding the vital dye propidium iodide was calculated at each time point after the beginning of C2-ceramide treatment with respect to the corresponding control. (b) Time course of caspase-3-like activity after 25 μM C2-ceramide (circles) or 25 μM C2-dihydroceramide (triangles) treatment. Data are mean ± SEM (bars) values of at least three experiments, performed in triplicate. The black arrows indicate the time of C2-ceramide treatment of the cell cultures used in the expression study.

Validation of hybridization signals

Hybridization of 9,120 cDNA clones with complex cDNA targets from poly(A)+ RNA extracted from C2-ceramide-treated or control cells produced signals of varying intensities (Figure 3a). In order to eliminate clones for which no reproducible hybridization signals were obtained, the signal-intensity values were validated as described in Materials and methods. Thus, 7% of the clones hybridized with the control cDNA target (634) and 14% of clones hybridized with the C2-ceramide-treated cDNA target (1,297) were excluded from further analysis. The remaining 6,494 clones were analyzed for differential hybridization.

Figure 3.

Figure 3

Hybridization signal analysis. (a) Macroarray of 9,120 cDNA clones hybridized with complex cDNA targets derived from mRNA of neuronally differentiated PC12 cells without C2-ceramide treatment (control) or treated with C2-ceramide (stimulated). (b) Distribution of the hybridization signal intensities between control and stimulated cells. Some genes identified in the present study are indicated.

Differential gene expression in neuronally differentiated PC12 cells treated with C2-ceramide compared to controls

Changes in gene expression were analyzed during the effector phase of neuronal death, 7 hours after the beginning of C2-ceramide treatment. This time point was chosen because on the one hand it is preceded by the activation of the transcription factor NFκB and c-Jun observed 4 to 6 hours after C2-ceramide treatment in PC12 cells [10,22], and on the other, the apoptotic process is still not induced by caspase-3 activation, which occurs 8 hours after the beginning of C2-ceramide treatment.

Hybridization between the rat PC12 cell-derived targets and the human cDNA macroarray was carried out as described in Materials and methods. Modulation of gene expression was quantitated by calculating the ratio of the intensity of the normalized hybridization signal obtained with the C2-ceramide cDNA target to that obtained with the control target. Clones were considered to be differentially hybridized in C2-ceramide-treated cells compared to control cells if the ratio between the corresponding hybridization intensity values was ≥ 2 (up-hybridized clones) or ≤ 0.5 (down-hybridized clones) which are the limits of confidence for the method. To decrease the risk of false-positive results, clones with hybridization signals that were less than twofold above background were also excluded, resulting in the elimination of 538 clones. In addition, the remaining clones were hybridized with complex cDNA targets from poly(A)+ RNA extracted from C2-dihydroceramide-treated cells used as negative control and compared to untreated cells. No modulation of expression was observed (except for one clone excluded from the analysis) in the presence of this inactive analog of C2-ceramide (data not shown). Among the 239 clones that met the criteria for differential hybridization, 132 were up-hybridized in C2-ceramide-treated cells and 107 were down-hybridized. The distribution of the hybridization-intensity values between the control and the C2-ceramide complex cDNA targets is presented in Figure 3b. Approximately 55% (72/132) of the up-hybridized clones were hybridized 3-6-fold more in C2-ceramide-treated cells than in the control and 40% (41/107) of the down-hybridized clones were hybridized 3-9-fold less.

Partial 5' and 3' sequences of the 239 clones were compared with all the sequences in the database developed in our laboratory (the Genexpress Index [23]) and in public databases. Of the 239 clones, 179 clones corresponded to already identified human genes, 113 of which have defined functions. The remaining 60 clones corresponded to genes with limited characterization. Under the hypothesis that differential hybridization of the clones reflects linear modulation of expression of the corresponding genes, we assume that we have detected differential gene expression using cDNA array technology that can be interpreted according to the information available.

Ten differentially expressed genes encode proteins with a role in ceramide or TNF-α signaling pathways (Figure 4, Table 1; see [24] for links to database entries for each gene). Two of these genes, PLA2G4C [25] and CLN3 [26,27] seem to have a role in ceramide-mediated cell death or survival. Two upregulated genes (ETV5 [28], NPTX2 [29,30]) and two downregulated genes (COL1SA1 [31,32], TNFAIP1 [33]) encode proteins that are modulated by TNF-α. Four genes, three upregulated (AXL [34], BIRC1 [35], RSU1 [36]) and one downregulated (MAPK10 [37]) encode proteins with a role in the TNF-α signaling pathway.

Figure 4.

Figure 4

Differentially expressed genes that encode proteins with functions involved in ceramide-dependent apoptosis. Black boxes, Genes involved in the ceramide signaling pathway; gray boxes, genes transcriptionally stimulated by TNF-α; white boxes, genes involved in the TNF-α signaling pathway.

Table 1.

Genes differentially expressed in ceramide-dependent apoptosis and involved in the ceramide and TNF-α pathways

Clone ID GENX GenBank accession number Unigene C. int. C. SD S. int. S. SD Ratio Similarity Gene symbol
Genes involved in the C2-ceramide signaling pathway
 yf59e08 5705 R13531 Hs.18858 3.56 0.54 12.01 0.86 3.37 Phospholipase A2, group IVC (cytosolic, calcium-independent) PLA2G4C
 yf71a08 115123 R12998; R40387 Hs.194660 1.14 0.23 2.53 0.15 2.21 Ceroid-lipofuscinosis, neuronal 3, juvenile (Batten, Spielmeyer-Vogt disease) CLN3
Genes transcriptionally stimulated by TNF-α
 yg86b08 4272 R53048; R53135 Hs.43697 1.72 0.18 8.77 0.58 5.09 Ets variant gene 5 (ets-related molecule) ETV5
 yc86d06 5838 F12910; T75064 Hs.3281 3.61 0.48 11.54 0.61 3.19 Neuronal pentraxin II NPTX2
 yc88f11 197 F10424; F12821 Hs.78409 1.18 0.21 0.55 0.11 0.47 Collagen, type XVIII, alpha 1 COL18A1
 yf78g01 200888 R14176; R40470 Hs.76090 1.19 0.28 0.54 0.06 0.46 TNF-α-induced protein 1 (endothelial) TNFAIP1
Genes involved in the TNF-α signaling pathway
 yf76d09 1017 R13424; R40936 Hs.83341 1.49 0.23 5.45 0.54 3.65 AXL receptor tyrosine kinase AXL
 yg49f10 116415 R20716 Hs.79019 4.54 0.13 12.69 1.38 2.80 Baculoviral IAP repeat-containing 1 BIRC1
 c-26g10 1350 F07467 Hs.75551 1.37 0.27 2.81 0.56 2.05 Ras suppressor protein 1 RSU1
 c-08d10 4997 F05370; Z38358 Hs.151051 4.94 1.06 2.41 0.51 0.49 Mitogen-activated protein kinase 10 MAPK10

Clone ID, clone name according to the public databases. GENX, cluster name including the corresponding cDNA sequence in the Genexpress Index 2 ([23] and R. Mariage-Samson et al., unpublished data). UniGene, cluster name in the UniGene database [85]; C. int., mean of the normalized and validated intensity values obtained after filter hybridization with complex cDNA target derived from control mRNA. C. SD, standard deviation derived from the C.int. S. int., mean of the normalized and validated intensity values obtained after filter hybridization with complex cDNA target derived from ceramide-stimulated cultured cell mRNA. S. SD, standard deviation derived from the S. int. Ratio, ratio of S. int. to C. int. Similarity, gene similarity.

Twenty clones correspond to genes encoding proteins that have been involved in the regulation of apoptosis and/or cell growth (Figure 5, Table 2, see [24]). Fourteen are up-hybridized and six are down-hybridized by C2-ceramide. Ten of the upregulated and two of the downregulated genes encode proteins stimulating apoptosis and/or growth arrest. The other genes (four upregulated and four downregulated) encode proteins downregulating apoptosis and/or stimulating growth.

Figure 5.

Figure 5

Differentially expressed genes that encode proteins involved in the regulation of apoptosis and/or cell growth. Gray boxes, genes stimulating apoptosis and/or growth arrest; white boxes, genes downregulating apoptosis and/or stimulating growth.

Table 2.

Differentially expressed genes that encode proteins involved in the regulation of apoptosis and/or cell growth

Clone ID GENX GenBank accession number Unigene C. int. C. SD S. int. S. SD Ratio Similarity Gene symbol
Proteins stimulating apoptosis and/or growth arrest
 yg01b10 2112 R18353; R42557 Hs.286 1.68 0.17 7.95 0.55 4.74 Ribosomal protein L4 RPL4
 yl73h11 567 H06473 Hs.9663 1.60 0.15 7.42 0.54 4.64 Programmed cell death 6-interacting protein PDCD6IP
 yc92h11 673 F13260; T77039 Hs.75709 2.12 0.33 9.83 0.95 4.64 Mannose-6-phosphate receptor (cation dependent) M6PR
 yg94h08 6030 R56149 Hs.78776 1.96 0.34 7.09 1.25 3.61 Putative transmembrane protein NMA
 yf90d04 25970 R15366 Hs.20912 1.34 0.22 4.71 0.58 3.51 Adenomatous polyposis coli like APCL
 yg76b02 3804 R51346; R51453 Hs.78935 0.95 0.18 2.89 0.40 3.03 Methionine aminopeptidase; eIF-2-associated p67 METAP2
 yd01h06 9451 R39334; T78769 Hs.274348 1.98 0.16 5.89 0.66 2.97 HLA-B associated transcript-3 BAT3
 c-22F12 2915 F08770 Hs.75323 1.44 0.23 3.26 0.29 2.26 Prohibitin PHB
 yf69g07 115124 R14126 Hs.132955 1.82 0.30 3.99 0.96 2.19 BCL2/adenovirus E1B 19 kD-interacting protein 3-like BNIP3L
 yd02b11 115910 T79985 Hs.63984 0.88 0.19 1.88 0.30 2.14 Cadherin 13, H-cadherin (heart) CDH13
 c-3ke04 781 F10823; F13223 Hs.12409 1.17 0.10 ND ND 0.43 Somatostatin SST
 yg64g08 115205 R35542; R51110 Hs.288986 3.01 0.41 0.90 0.15 0.30 Survival of motor neuron 1 1, telomeric SMN1
Proteins downregulating apoptosis and/or stimulating growth
 yg44d03 408 R25503 Hs.155212 1.74 0.40 7.67 0.40 4.40 Methylmalonyl coenzyme A mutase MUT
 yg68d10 2957 R36284; R49571 Hs.89582 1.80 0.25 7.26 0.57 4.04 Glutamate receptor, ionotropic, AMPA 2 GRIA2
 yl81d04 9379 H05457; H07007 Hs.150423 2.64 0.36 8.72 1.06 3.31 Cyclin-dependent kinase 9 (CDC2-related kinase) CDK9
 yd02a11 78693 T79973 Hs.107911 2.52 0.42 5.38 0.44 2.14 ATP-binding cassette, sub-family B (MDR/TAP), member 6 ABCB6
 yg51a11 17820 R21694; R46587 Hs.223014 1.09 0.24 ND ND 0.46 Antizyme inhibitor OAZIN
 yh10g09 4858 R61276; R61277 Hs.8073 1.59 0.23 0.66 0.16 0.41 Septin 3 SEP3
 yf53a12 3165 R12025; R37093 Hs.356245 1.14 0.21 0.29 0.02 0.25 Apoptosis regulator LOC51283
 yg67b12 115951 R35827; Hs.285754 2.38 0.48 0.50 0.12 0.21 Met proto-oncogene MET

Abbreviations and column headings are as in Table 1.

The remaining 83 clones corresponding to 82 genes with known or putative functions have no obvious relation to the apoptosis process (Table 3, see [24]). Of the total number of differentially hybridized clones, 66 correspond to mRNA sequences (Table 4, see [24]) and 60 to poorly characterized genes (Table 5, see [24]) that encode proteins without known function.

Table 3.

Known genes differentially expressed in ceramide-dependent apoptosis with no identified direct interaction with the ceramide-dependent apoptosis process

Clone ID GENX GenBank accession number Unigene C. int. C. SD S. int. S. SD Ratio Similarity Gene symbol
Signal transduction
 yl85b10 1842 H05211 Hs.22003 1.62 0.40 7.99 0.37 4.94 Solute carrier family 6 (neurotransmitter transporter, GABA), member 1 SLC6A1
 yf77g11 3900 R14207; R37490 Hs.75819 1.12 0.12 5.02 0.43 4.46 Glycoprotein M6A GPM6A
 yg63f10 1552 R26636; R49665 Hs.24212 1.01 0.15 4.02 0.61 3.98 Latrophilin KIAA0786
 c-2ee07 116218 Z45003 Hs.107979 1.75 0.35 6.17 0.85 3.52 Small membrane protein 1 SMP1
 yf60h11 12653 R13771 Hs.61628 1.43 0.18 4.68 0.61 3.28 Calcium binding atopy-related autoantigen 1 CBARA1
 yf88a09 9668 R15201 Hs.181326 4.01 0.50 11.65 2.01 2.90 Myotubularin-related protein 2 MTMR2
 yg11b08 107475 R17181; R41731 Hs.5462 0.72 0.12 1.54 0.33 2.14 Solute carrier family 4, sodium bicarbonate cotransporter, member 4 SLC4A4
 c-2mh12 1997 Z41050; Z45338 Hs.108787 1.08 0.21 0.52 0.04 0.47 Phosphatidylinositol glycan, class N PIGN
 yc87e10 115203 F10343; F12737 Hs.173717 1.24 0.17 0.50 0.06 0.40 Phosphatidic acid phosphatase type 2B PPAP2B
 yf48c10 9043 R12286; R12797 Hs.10842 1.10 0.24 0.43 0.03 0.39 RAN, member RAS oncogene family RAN
 yd09f12 2991 R39085 Hs.306359 2.39 0.46 0.90 0.22 0.38 Hect domain and RCC1 (CHC1)-like domain (RLD) 1 HERC1
 c-3ie05 5307 F10685; F13091 Hs.9347 1.48 0.24 0.53 0.02 0.36 Regulator of G-protein signaling 14 RGS14
 yg16c08 5294 R17962; R43452 Hs.1440 1.05 0.23 0.29 0.04 0.27 Gamma-aminobutyric acid (GABA) A receptor, beta 3 GABRB3
 yf50c04 1366 R11777; R37698 Hs.5985 1.13 0.12 0.17 0.01 0.15 Non-kinase Cdc42 effector protein SPEC2 LOC56990
Transcription/translation
 yf71g02 5232 R40420 Hs.16313 0.90 0.13 2.30 0.15 2.55 Kruppel-like zinc-finger protein GLIS2 GLIS2
 c-26a02 451 F07446 Hs.13993 1.64 0.38 3.39 0.73 2.07 TBP-like 1 TBPL1
 c-05c07 4917 Z38284; Z41997 Hs.26973 1.21 0.20 2.45 0.52 2.02 Bromodomain adjacent to zinc-finger domain, 2B BAZ2B
 c-24a11 114423 F07382 Hs.75678 1.38 0.23 0.66 0.16 0.47 FBJ murine osteosarcoma viral oncogene homolog B FOSB
 yg90e12 10904 R56427; R56428 Hs.239 1.28 0.23 0.59 0.03 0.46 Forkhead box M1 FOXM1
 yf61e03 4401 R13803; R37662 Hs.182447 7.20 1.01 2.75 0.60 0.38 Heterogeneous nuclear ribonucleoprotein C (C1/C2) HNRPC
 yf64g02 993 R37803 Hs.6151 4.87 0.77 1.87 0.46 0.38 Pumilio homolog 2 (Drosophila) PUM2
 yg53f10 1678 R62465; R25720 Hs.520 1.41 0.20 ND ND 0.35 Nuclear receptor subfamily 2, group C, member 2 NR2C2
 yg47e10 1548 R21283; R45373 Hs.14520 1.55 0.26 0.53 0.13 0.34 Eukaryotic translation initiation factor 2C, 1 EIF2C1
 yg36d06 1872 R24568; R44373 Hs.76177 10.91 1.23 3.67 0.10 0.34 Transcription factor CP2 TFCP2
 yg60b12 303 R35123; R49511 Hs.2186 3.12 0.63 0.94 0.07 0.30 Eukaryotic translation elongation factor 1 gamma EEF1G
 yg27a08 4127 R43968 Hs.278589 9.43 1.24 2.76 0.40 0.29 General transcription factor II, i, pseudogene 1 GTF2IP1
Cellular traffic or structure proteins
 yg19f05 200119 R20424; R43544 Hs.169793 1.51 0.36 7.37 1.32 4.86 Ribosomal protein L32 RPL32
 yc86h03 2760 F12918; T75229 Hs.182625 2.38 0.26 7.78 1.17 3.27 Vamp (vesicle-associated membrane protein)-associated protein B and C VAPB
 yc87f04 5084 R38549; T75126 Hs.22826 1.83 0.11 5.59 0.64 3.06 Tropomodulin 3 (ubiquitous) TMOD3
 yf98g01 8512 R18713 Hs.75196 2.96 0.63 9.29 0.80 3.14 Ankyrin repeat-containing protein G9A
 yh17e09 1304 R59488; R59489 Hs.30991 0.78 0.19 2.32 0.11 2.97 Ankyrin repeat domain 6 ANKRD6
 yf76d11 424 R13426; R40938 Hs.119324 0.84 0.08 2.07 0.35 2.48 Kinesin-like 4 KNSL4
 c-27f03 1382 F07488 Hs.89497 2.32 0.31 5.60 0.64 2.42 Lamin B1 LMNB1
 yc96a12 11155 F13331; T77651 Hs.159613 4.50 0.32 10.84 2.13 2.41 Thyroid hormone receptor binding protein AIB3
 yf57c11 1225 R12822; r20734 Hs.1501 0.94 0.22 2.25 0.22 2.39 Syndecan 2 SDC2
 yl71a06 10804 H05894 Hs.6682 1.33 0.11 2.94 0.20 2.21 Solute carrier family 7, cationic amino acid transporter, y+ system, member 11 SLC7A11
 yc99f07 11082 T78361 Hs.103042 2.21 0.07 0.98 0.19 0.44 Microtubule-associated protein 1B MAP1B
 yf72e08 2558 R13080; R40510 Hs.7979 2.05 0.34 0.80 0.15 0.39 Likely ortholog of mouse synaptic vesicle glycoprotein 2a SV2
 yc87h12 2952 F10545; F12946 Hs.21611 5.68 0.59 1.93 0.17 0.34 Kinesin family member 3C KIF3C
 yg54d05 604 R25813; R46810 Hs.117977 1.62 0.33 0.50 0.11 0.31 Kinesin 2 (60-70 kD) KNS2
 yf91b02 1980 R16352; R42300 Hs.103042 3.50 0.41 1.01 0.24 0.29 Microtubule-associated protein 1B MAP1B
 yf72a03 115963 R13048; R40479 Hs.187958 1.46 0.34 0.40 0.06 0.28 Solute carrier family 6, member 8, accessory proteins BAP31/BAP29 SLC6A8, DXS1357E
Immunity/inflammatory response
 yg75d06 25621 R54423 Hs.179661 1.88 0.18 8.01 1.02 4.26 FK506-binding protein 1A (12 kD) FKBP1A
 yg65b03 2453 R35324 Hs.9688 0.86 0.13 3.67 0.60 4.26 Leukocyte membrane antigen IRC1
 yg57f05 190007 R34428 Hs.181244 3.83 0.24 9.76 1.22 2.55 MHC class I gene family
 yf51e08 2563 R12005; R39844 Hs.75682 0.89 0.04 2.05 0.21 2.31 Autoantigen RCD-8
 c-2bh04 190137 F03851; F07604 Hs.284394 1.13 0.07 0.56 0.07 0.50 Complement component 3 C3
 yf59h02 5580 R13549; R20669 Hs.82689 1.05 0.21 0.47 0.10 0.44 Tumor rejection antigen (gp96) 1 TRA1
 yc86g03 8628 F10456; F12856 Hs.302749 1.51 0.35 0.58 0.09 0.39 FK506-binding protein 9 (63 kD) FKBP9
Protein processing
 yf68a10 1071 R40190; Hs.75890 0.55 0.13 2.09 0.23 3.80 Site-1 protease (subtilisin-like, sterol-regulated, cleaves sterol regulatory element binding proteins) S1P
 c-2na07 2001 F04230; F07978 Hs.102 1.01 0.16 0.46 0.10 0.45 Aminomethyltransferase (glycine cleavage system protein T) AMT
 yc85d05 6301 F10498; F12892 Hs.170197 1.45 0.22 0.59 0.10 0.41 Glutamic-oxaloacetic transaminase 2, mitochondrial (aspartate aminotransferase 2) GOT2
 c-2ge12 2793 Z40826; Z46090 Hs.183212 1.14 0.19 0.45 0.09 0.39 Isoprenylcysteine carboxyl methyltransferase ICMT
 yg52f04 202164 R21082; R46258 Hs.235887 1.55 0.31 0.54 0.08 0.34 HMT1 (hnRNP methyltransferase, Saccharomyces cerevisiae)-like 1 HRMT1L1
Proteases
 yf64f07 2813 R13707; R37801 Hs.171501 1.27 0.24 ND ND 0.39 Ubiquitin specific protease 11 USP11
Metabolism
 yc97f08 1805 R39698; T78043 Hs.2838 2.73 0.24 9.35 0.86 3.42 Malic enzyme 3, NADP(+)-dependent, mitochondrial ME3
 yg97d06 3929 R59198; R59256 Hs.78989 0.68 0.07 1.99 0.18 2.94 Alcohol dehydrogenase 5 (class III), chi polypeptide ADH5
 c-2ca07 1549 F03858; F07608 Hs.180616 1.04 0.12 0.50 ND 0.48 CD36 antigen (collagen type I receptor, thrombospondin receptor)-like 1 CD36L1
 yc95g06 3164 R39463; T77281 Hs.155247 1.01 0.20 0.31 0.05 0.30 Aldolase C, fructose-bisphosphate ALDOC
Miscellaneous
 yg24g06 3097 R19249; R44514 Hs.22654 0.48 0.08 2.93 0.67 6.12 Sodium channel, voltage-gated, type I, alpha polypeptide SCNIA
 yc89d05 10816 F10796; F13191 Hs.12365 1.26 0.20 5.46 0.36 4.34 Synaptotagmin XIII SYT13
 c-28e05 4334 F07514 Hs.6126 2.04 0.50 7.19 1.11 3.52 Mannosidase, beta A, lysosomal-like MANBAL
 yg35g09 4463 R20330 Hs.88764 3.30 0.19 9.88 2.40 2.99 Male-specific lethal-3 (Drosophila)-like 1 MSL3L1
 yg36c01 292 R24560; R44360 Hs.6430 1.50 0.22 4.40 0.78 2.93 Protein with polyglutamine repeat; calcium (Ca2+) homeostasis endoplasmic reticulum protein ERPROT213-21
 yc98a06 1475 R37847; T78111 Hs.301789 1.55 0.27 4.52 0.35 2.92 Capping protein (actin filament) muscle Z-line, alpha 1 CAPZA
 yf54h02 924 R11969 Hs.4865 1.41 0.17 3.05 0.18 2.17 Voltage-gated sodium channel beta-3 subunit (scn3b gene) HSA243396
 yf91a04 434 R16348; R42296 Hs.12152 1.00 0.24 2.03 0.16 2.04 APMCF1 protein APMCF1
 c-1ia09 4199 Z39718; Z43661 Hs.8834 0.64 0.15 1.30 0.19 2.02 Ring finger protein 3 RNF3
 yg83b04 4211 R53332; R53937 Hs.7022 1.19 0.28 0.59 0.09 0.50 Dedicator of cytokinesis 3 DOCK3
 yf57d02 4610 R12627; R20528 Hs.334688 1.51 0.25 0.75 0.13 0.50 Phytanoyl-CoA hydroxylase interacting protein PHYHIP
 yg36h06 3087 R24595; R44400 Hs.7122 1.08 0.13 ND ND 0.46 Scrapie responsive protein 1 SCRG1
 yf99b05 2822 R18211; R42149 Hs.79284 1.07 0.21 0.48 0.07 0.45 Mesoderm specific transcript (mouse) homolog MEST
 c-24h06 92359 Z40467; Z44591 Hs.171545 1.02 0.20 0.45 0.11 0.44 HIV-1 Rev binding protein HRB
 yd05d01 2346 R38832; T80384 Hs.13493 1.06 0.24 0.44 0.06 0.42 Like mouse brain protein E46 E46L
 yf74e11 2106 R13277; R40723 Hs.334851 1.95 0.41 0.80 0.17 0.41 LIM and SH3 protein 1 LASP1
 yd01e11 3181 T78746 Hs.168640 1.17 0.27 0.47 0.10 0.40 Homolog of mouse Ank ANK
 yf48e09 414 R12292; R12804 Hs.21050 1.22 0.26 0.47 0.07 0.38 g20 protein LOC51161
 yg16d07 1087 R43459; R17969 Hs.87125 9.91 2.06 3.35 0.51 0.34 EH-domain containing 3 EHD3
 yf57d07 12763 R12632; R20533 Hs.109706 1.78 0.23 0.58 0.09 0.33 Hematological and neurological expressed I HN1
 c-2Ia12 200991 F04056; F07796 Hs.74376 1.00 0.07 0.33 0.01 0.33 Olfactomedin related ER localized protein NOE1
 yf61b05 1882 R13783; R37641 Hs.297743 2.04 0.16 0.63 0.12 0.31 Carbonic anhydrase X CA10
 yf61a10 3960 R39112; R13989 Hs.2288 12.49 1.82 3.38 0.84 0.27 Visinin-like 1 VSNL1
 yg53b12 8924 R25707; R62451 Hs.169047 2.34 0.46 0.54 0.12 0.23 Chondroitin sulfate proteoglycan 3 (neurocan) CSPG3

Abbreviations and column headings are as in Table 1.

Table 4.

Messenger RNA or protein sequences differentially expressed in ceramide-dependent apoptosis

Clone ID GENX GenBank accession number Unigene C. int. C. SD S. int. S. SD Ratio Similarity

Upregulated clones
 yg51f11 229 R21710 Hs.64691 1.32 0.16 6.34 0.43 4.80 KIAA0483 protein
 yg30b04 5093 R44721 Hs.12896 1.31 0.12 5.99 0.29 4.57 KIAA1034 protein
 yf53g09 13 R12046 Hs.90424 1.56 0.10 7.09 0.67 4.55 Homo sapiens cDNA: FLJ23285 fis, clone HEP09071
 yc94b11 223 F13362; T77404 Hs.101375 1.40 0.15 6.38 1.10 4.55 cDNA DKFZp434H205 (from clone DKFZp434H205)
 yg37d06 2008 R19640 Hs.264636 1.70 0.39 7.39 1.60 4.34 KIAA0781 protein
 yf75c06 425 R13300; R40783 Hs.26409 0.83 0.18 3.55 0.67 4.27 cDNA DKFZp547K204 (from clone DKFZp547K204)
 yf79e07 2479 R14269; R40562 Hs.19150 0.93 0.15 3.91 0.25 4.22 cDNA DKFZp564A2164 (from clone DKFZp564A2164)
 yf49e01 6657 R11708 Hs.21710 1.64 0.20 6.61 1.00 4.04 Hypothetical protein DKFZp761G0313
 yg07h12 200578 R22668 Hs.7734 1.00 0.09 3.71 0.56 3.70 H. sapiens cDNA: FLJ21380 fis, clone COL03329
 yg32e10 5469 R23681 Hs.106825 1.38 0.15 5.09 0.55 3.68 Hypothetical protein FLJ20300
 yf90d07 6203 R15369; R42110 Hs.323396 1.28 0.20 4.66 0.41 3.63 Hypothetical protein RP1-317E23 (LOC56181)
 yg91g03 1624 R56083; R56195 Hs272814 0.83 0.15 2.89 0.30 3.48 Chromosome 20 open reading frame 67
 yg11e11 2086 R17284 Hs.106210 4.25 0.56 14.04 2.67 3.30 Hypothetical protein FLJ10813
 ym11b06 6715 H11788 Hs.125034 1.98 0.16 6.52 0.85 3.29 H. sapiens cDNA FLJ10733 fis, clone NT2RP3001392
 yf80c08 772 R14304; R40254 Hs.59236 1.21 0.25 3.94 0.63 3.24 Hypothetical protein DKFZp434L0718
 yg18e11 4391 R20224 Hs.41185 3.49 0.86 11.27 1.23 3.23 cDNA DKFZp564O1262 (from clone DKFZp564O1262)
 yc90h10 1343 F13218; T75433 Hs.141003 1.32 0.24 4.19 1.05 3.18 H. sapiens cDNA: FLJ21691 fis, clone COL09555
 yg42a11 200671 R24764; R45496 Hs.288368 0.45 0.10 1.41 0.11 3.14 H. sapiens cDNA: FLJ21314 fis, clone COL02248
 yc85f03 255 F12760; T74722 Hs.318401 3.03 0.42 9.46 0.80 3.12 HSPC039 protein (LOC51124)
 yc86g12 32 F12859; T75226 Hs.180948 4.61 0.28 14.27 1.89 3.10 KIAA0729 protein
 c-2lb03 1917 Z45263 Hs.155182 6.48 1.55 19.98 3.34 3.08 KIAA1036 protein
 yf94d09 2836 R16328; R41404 Hs.6343; HS.306400 3.16 0.48 9.58 0.97 3.03 KIAA1464 protein
 yf49g10 2696 R11887 Hs.40094 4.42 0.68 13.38 1.70 3.03 Human DNA sequence from clone 167A19 on chromosome 1p32.1-33
 yg67h02 1136 R35733; R49366 Hs. 325825 3.76 0.34 11.27 0.80 3.00 H. sapiens cDNA: FLJ20848 fis, clone ADKA01732
 yc89d09 2388 F13194; T75317 Hs.22109 3.52 0.07 10.11 1.40 2.87 KIAA0945 protein
 yf72d11 4469 R13137; R40616 Hs.6311 2.38 0.54 6.80 0.92 2.86 H. sapiens cDNA: FLJ20859 fis, clone ADKA01617
 yg73c09 51540 R51740 Hs.288959 1.31 0.18 3.70 0.65 2.83 H. sapiens cDNA: FLJ20920 fis, clone ADSE00877
 yf50h09 9583 R11919; Hs.11637 3.87 0.33 10.71 1.31 2.77 H. sapiens mRNA; cDNA DKFZp547J125 (from clone DKFZp547J125)
 c-2ba02 4345 Z41723; Z44845 Hs.15921 1.93 0.37 5.31 1.02 2.75 Hypothetical protein FLJ10759
 yg36f12 11000 R25011; R45019 Hs.118983 1.13 0.27 3.00 0.46 2.65 H. sapiens cDNA FLJ12150 fis, clone MAMMA1000422
 c-24b10 1689 Z44563 Hs.154919 2.67 0.43 6.60 1.44 2.47 KIAA0625 protein
 yf76a11 1849 R13420; R40930 Hs.7822 1.12 0.09 2.73 0.33 2.43 cDNA DKFZp564C1216 (from clone DKFZp564C1216)
 yc91c07 162 F10758; F13156 Hs.140833 0.61 0.14 1.46 0.32 2.41 H. sapiens mRNA full length insert cDNA clone EUROIMAGE 29222
 yc94c08 4310 R38361; T77413 Hs.119004 0.53 0.05 1.25 0.22 2.36 KIAA0665 gene product
 yg57f04 3072 R34427; R48960 Hs.326416 0.90 0.16 1.98 0.35 2.20 cDNA DKFZp564H1916 (from clone DKFZp564H1916)
 yg15g12 5559 R18075; R42970 Hs.22370 0.66 0.14 1.45 0.21 2.19 cDNA DKFZp564O0122 (from clone DKFZp564O0122)
 yg97d02 1018 R59194; R59252 Hs.5324 0.64 0.07 1.32 0.16 2.06 Hypothetical protein (CL25022)
 yc95f04 3851 F13386; R39459 Hs.7888 0.58 0.07 1.16 0.25 2.02 H. sapiens clone 23736 mRNA sequence
Downregulated clones
 yg42e05 4312 R45416; R25077 Hs.169330 1.05 0.23 0.52 0.05 0.49 Neuronal protein (NP25)
 yg89f11 2081 R55970; R55969 Hs.16443 1.16 0.27 0.56 0.06 0.49 H. sapiens cDNA: FLJ21721 fis, clone COLF0381
 yg33e09 5446 R20455; R44158 Hs.333389 1.39 0.19 0.67 0.17 0.48 Hypothetical protein MGC13090
 c-2aa11 1485 Z40609; Z44824 Hs.13485 1.44 0.23 0.70 0.16 0.48 KIAA1918 protein
 yf65e06 5690 R13865; R37007 Hs.301685 1.03 0.21 ND ND 0.48 KIAA0620 protein
 yl76d07 37588 H05960; H06010 Hs.92418; Hs.63510 3.95 0.73 1.85 0.18 0.47 KIAA0141
 c-2cg09 201091 F03885; F07635 Hs.288361 1.06 0.10 0.49 0.02 0.47 H. sapiens cDNA: FLJ22696 fis, clone HSI11696
 yg64h02 2829 R35543; R51112 Hs.12239 2.94 0.61 1.35 0.30 0.46 CGI-10 protein (LOC51004)
 yf49c08 23982 R11699; R17677 Hs.322844 1.29 0.32 0.58 0.12 0.45 Hypothetical protein DKFZp564A176
 yg33g08 636 R20203; R44989 Hs.7750 8.39 1.43 3.60 0.64 0.43 Novel human gene mapping to chromosome 1
 yf53d08 532 R11837; R36955 Hs.246885 1.07 0.05 0.44 0.08 0.42 Hypothetical protein FLJ20783
 yg65h10 10701 R35431; R49229 Hs.222746 1.04 0.25 0.42 0.09 0.40 KIAA1610 protein
 yg69e11 1257 R36317; R49249 Hs.216958 1.16 0.28 0.44 0.10 0.38 KIAA0194 protein
 yf79f12 5599 R14349; R40677 Hs.179946 2.55 0.37 0.86 0.20 0.34 KIAA1100 protein
 yf86c11 1909 R15181; R41632 Hs.286013 1.06 0.14 0.34 0.08 0.32 Short coiled-coil protein
 yf78c09 1664 R14217; R40635 Hs.351029 10.44 1.29 3.36 0.75 0.32 H. sapiens cDNA FLJ31803 fis, clone NT2RI2009101
 yf61c10 1067 R13997; R39120 Hs.5008; Hs.21515 1.11 0.09 0.35 0.08 0.32 CG-87 protein
 yd06g01 2455 R38891; T81283 Hs.165570 1.41 0.07 0.45 0.10 0.32 H. sapiens clone 25052 mRNA sequence
 yf64f10 111134 R36936 Hs.80285 8.72 0.83 2.76 0.35 0.32 mRNA cDNA DKFZp586C1723 (from clone DKFZp586C1723)
 yc91e03 11140 F13018; T77597 Hs.337629 5.27 1.08 1.67 0.22 0.32 cDNA DKFZp434D115 (from clone DKFZp434D115)
 yf65b02 8918 R13839; R36985 Hs.227913 1.09 0.24 0.32 0.05 0.30 API5-like 1
 yf60a03 1485 R13618; R38474 Hs.13485 2.13 0.36 0.65 0.09 0.30 KIAA1918 protein
 yf56a05 5140 R12419 Hs.7132 1.97 0.33 0.57 0.08 0.29 KIAA0574 protein
 yg78h08 884 R51917; R54309 Hs.6449 1.20 0.28 0.34 0.08 0.28 CGI-87 protein (LOC51112)
 yf69g12 713 R40161 Hs.288776 1.21 0.23 0.29 0.04 0.24 H. sapiens cDNA: FLJ21304 fis, clone COL02111
 yf93b11 2192 R16295; R40219 Hs.108504 1.25 0.16 0.24 0.05 0.20 Hypothetical protein FLJ20113
 yf51g08 2572 R12017; R39856 Hs.20977 1.11 0.20 0.19 0.04 0.17 Human DNA sequence from clone RP5-881L22 on chromosome 20
 yg26c11 904 R19006; R44076 Hs.226396 3.13 0.50 0.35 0.08 0.11 Hypothetical protein FLJ11126

Abbreviations and column headings are as in Table 1.

Table 5.

Unknown genes differentially expressed in ceramide-dependent apoptosis

Clone ID GENX GenBank accession number Unigene C. int. C. SD S. int. S. SD Ratio Similarity
Upregulated clones
 yf66a04 17755 R18781 1.04 0.24 5.19 0.59 4.98 ESTs
 yf88d07 3024 R15141; R41563 Hs.12381 1.21 0.09 5.83 0.85 4.80 ESTs
 yc85h07 11132 F12902; T74741 1.38 0.16 5.75 0.53 4.16 ESTs
 yg38a10 2564 R19870; R45098 Hs.182503 1.32 0.23 5.46 0.63 4.15 ESTs
 c-25h01 1301 Z44625 Hs.29672 4.51 0.68 18.31 3.99 4.06 ESTs
 yg53c11 5862 R25710; R62454 1.83 0.19 7.36 0.59 4.02 ESTs
 yg02a02 5691 R18381; R42444 Hs.240816 1.52 0.23 6.08 0.94 4.00 ESTs
 yh09g12 4411 R61781; R61782 1.20 0.30 4.55 0.24 3.78 ESTs
 yf80c09 943 R14362 2.24 0.32 8.34 0.16 3.73 ESTs
 yd02e05 761 R39357; T80134 Hs.306425; Hs.327350 1.20 0.19 4.47 0.73 3.72 ESTs
 yg17c05 5291 R18746; R43067 Hs.238956 1.09 0.11 3.79 0.39 3.48 ESTs
 c-2ef12 1659 F07687 3.08 0.02 10.53 2.42 3.42 ESTs
 yf58e03 1072 R12737; R39789 Hs.119714 3.06 0.60 10.30 0.39 3.36 ESTs
 yl69a01 160 H00104 Hs.21417 3.24 0.54 10.72 1.23 3.31 ESTs
 yc93d09 438 T77119 Hs.21417 2.08 0.45 6.77 1.60 3.25 ESTs
 c-28f03 1425 F07517; Z40576 2.49 0.35 7.77 0.52 3.12 ESTs
 yg60e11 2509 R35134 4.12 0.92 12.87 1.02 3.12 ESTs
 yl96g09 11047 H09060 2.97 0.51 9.23 1.00 3.11 ESTs
 yg02f03 2758 R18419 HS.18585 3.51 0.62 10.83 1.03 3.09 ESTs
 yf94d10 11844 R16329; R41405 Hs.197143 2.73 0.46 8.41 1.14 3.08 ESTs
 yf63f02 201117 R13594 Hs.155639 1.92 0.25 5.75 0.77 3.00 ESTs
 yf98b09 16058 R18177; R42241 Hs.106359 1.07 0.24 3.17 0.27 2.95 ESTs
 yf80c07 1885 R14303 Hs.32565 0.76 0.04 2.18 0.33 2.85 ESTs
 yc92a01 11141 F13028; T76925 4.87 0.12 13.72 2.61 2.82 ESTs
 yf76a02 711 R13339 Hs.7913 5.21 0.78 14.52 3.06 2.79 ESTs
 yf55h04 664 R12357 3.64 0.18 9.99 1.24 2.75 ESTs
 yc85h06 11131 F12901; T74740 5.20 0.54 14.15 2.43 2.72 ESTs
 yc88c03 10642 F12878; R38624 Hs.106313 1.50 0.30 4.04 0.70 2.70 ESTs
 yg39a10 10317 R19899; R45120 Hs.89388 4.91 0.67 12.91 2.40 2.63 ESTs
 yh15d09 6818 R61465 4.60 0.37 11.88 1.21 2.58 ESTs
 yg02g01 1987 R18425; R42486 Hs.4983 1.11 0.27 2.75 0.51 2.47 ESTs
 yg08h03 201114 R22721; R43427 Hs.244482 0.70 0.17 1.60 0.08 2.28 ESTs, moderately similar to alternatively spliced product using exon 13A (H. sapiens)
 yg33b02 4208 R20161; R44947 Hs.22905 0.91 0.19 2.05 0.17 2.26 ESTs
 yg44c04 3106 R25497; R45563 None 1.11 0.27 2.60 0.50 2.33 ESTs
 yg46g12 5388 R20696; R45358 Hs.311444; Hs.6591 0.90 0.17 1.95 0.48 2.16 ESTs
 yg42a06 2573 R25050; R45389 Hs.23558 0.57 0.14 1.22 0.22 2.13 ESTs
 yf63f11 5521 R36919 Hs.25205 0.99 0.14 2.11 0.19 2.13 ESTs
Downregulated clones
 c-2eg10 1662 F03955; F07692 1.04 0.19 0.51 0.08 0.49 ESTs
 c-29f04 201571 Z40598; Z44804 Hs.184780 1.06 0.15 0.52 0.11 0.49 ESTs
 c-2la08 1913 Z40977; Z45261 Hs.125266 1.03 0.22 ND ND 0.49 ESTs
 c-2ch10 3050 F03889; F07637 Hs.27278 2.42 0.49 1.12 0.24 0.46 ESTs, weakly similar to chain A, cyclophilin A complexed with cyclosporin A (H. sapiens)
 yg83b05 20476 R53938; R53333 1.27 0.10 0.56 0.13 0.44 ESTs
 yg36f04 5214 R24580 Hs.27104 2.18 0.53 0.95 0.17 0.43 ESTs
 yf60a12 3065 R38592; R13746 6.52 1.15 2.61 0.42 0.40 ESTs
 yc86e07 2935 F10326; F12716 Hs.227993 7.76 0.94 3.02 0.59 0.39 ESTs
 yc90f10 10752 F10679; F13085 Hs.12395 1.12 0.20 0.43 0.08 0.38 ESTs
 yc97e12 4395 T78036 Hs.23213 1.11 0.20 0.41 0.05 0.37 ESTs
 yf50h10 477 R11920; R39108 Hs.6777 2.39 0.56 0.82 0.16 0.34 ESTs
 yf74a06 16024 R13206; R40294 1.32 0.28 0.45 0.10 0.34 ESTs
 yg96d11 3143 R59141; R59142 1.30 0.19 0.43 0.08 0.33 ESTs
 yf51a04 958 R11976; R39818 Hs.4241 1.25 0.23 0.40 0.09 0.32 ESTs
 yg51e05 5025 R46483; R21387 Hs.23187 6.26 0.92 1.98 0.19 0.32 ESTs
 yg02h09 2969 R17514; R42608 Hs.139270 10.09 0.93 3.19 0.46 0.32 ESTs
 yf66f03 978 R37086 Hs.23210 1.72 0.18 ND ND 0.29 ESTs
 yf67b06 115094 R18860 Hs.203213 1.72 0.28 ND ND 0.29 ESTs
 yl91f12 4185 H08130; H08131 Hs.19515 2.86 0.33 0.70 0.17 0.25 ESTs
 yg14a03 2782 R17432; R42778 Hs.22217 1.57 0.27 0.34 0.06 0.22 ESTs
 yf52e12 4147 R12228; R39947 Hs.7237 1.57 0.30 0.34 0.06 0.22 ESTs
 yf50g11 1829 R11917; R39107 Hs.352354; Hs.244624 2.37 0.24 0.48 0.10 0.20 ESTs
 yf84f08 2237 R14545; R41206 Hs.349648 1.06 0.17 0.19 0.03 0.18 ESTs, weakly similar to KIAA1157 protein (H. sapiens)

Abbreviations and column headings are as in Table 1.

To confirm the results obtained by macroarray analysis, differentially expressed transcripts representing upregulated or downregulated genes were analyzed for differential expression by reverse transcription PCR (RT-PCR) or northern blots. As shown in Figure 6, the upregulation of ETV5, M6PR and APCL was confirmed by RT-PCR, and the downregulation of two genes with unknown function (mRNA DKFZp586C1723 and GENX 2969) was confirmed by northern blotting.

Figure 6.

Figure 6

Confirmation of macroarray results by RT-PCR and northern blotting. The percentage of signal modulation (PCR amplification signal or hybridization signal) in relation to control cells (without C2-ceramide treatment) has been calculated in each condition to compare the expression of each gene in neuronally differentiated PC12 cells with (black boxes) or without (white boxes) C2-ceramide treatment. The PCR amplification signal and the hybridization signal for the positive controls (HPRT and 18S rRNA genes, respectively) are indicated.

Discussion

Extracellular signaling molecules such as cytokines, growth-factor deprivation and DNA damage caused by chemotherapeutic agents or irradiation activate ceramide-mediated signal transduction pathways leading to cell death. These pathways have been investigated in the immune system, where they are known to have an important role, and in neurons, as they are suspected to play a part in neurodegenerative disorders [1]. A number of steps in the signaling cascades have been elucidated. However, although the translation inhibitor cycloheximide inhibits the ceramide-mediated death of mesencephalic neurons [7], the expression patterns of genes modulated during ceramide-mediated cell death remain unknown. In a global approach to this question, we have used cDNA macroarray technology to determine the profile of gene expression in a neuronal model of cell death, neuronally differentiated and C2-ceramide-treated PC12 cells, in which ceramide-dependent changes in gene expression could be isolated from the effects of other transcription modulators.

Identification of genes closely implicated in the ceramide and/or TNF-α signaling pathway

We were able to detect differential expression of 10 genes known to be involved in the ceramide or TNF-α signaling pathways (see Figure 4, Table 1) thus validating our study. A summary illustration of the putative role of these genes is presented in Figure 7. Briefly, two genes, encoding phospholipase A2 group IVC (PLA2G4C) and ceroid-lipofuscinosis, neuronal 3, juvenile (CLN3) are already known to be involved in ceramide-mediated signal transduction. The first, PLA2G4C, belongs to the cytosolic phospholipase A2 gene family that encodes two different proteins: calcium-independent and calcium-dependent cytosolic phospholipases [38]. TNF-α regulates the expression of PLA2G4A mRNA in HeLa cells [39] and in human bronchial epithelial cells [40], which is indirect evidence of modulation by ceramide, but the role of ceramide was not demonstrated directly in these studies. However, ceramide was shown directly to upregulate the expression of the gene encoding cytosolic phospholipase A2 in the fibroblast cell line L929 [41]. Conversely, the activation of this gene was reported to be necessary for ceramide accumulation and cell death in the same cells [25]. We show for the first time that this gene is involved in neuronal apoptosis.

Figure 7.

Figure 7

Schematic illustrating the putative roles of the proteins encoded by the genes noted in Figures 4 and 5.

The second gene, CLN3, is expressed in a variety of human tissues including the brain, where the product is necessary for neuronal survival [26,27]. Interestingly, CLN3 does not inhibit C2-ceramide-induced apoptosis but modulates endogenous ceramide synthesis and suppresses apoptosis by preventing generation of ceramide [42]. Thus, C2-ceramide can activate a negative feedback mechanism regulating endogenous ceramide generation as well as activate the downstream targets of the endogenous lipid.

Four other genes or families of genes known to be transcriptionally regulated by TNF-α were also modulated by C2-ceramide in our model (Table 1). Of these, ETS variant 5 (ETV5) belongs to the family of ETS transcription factor genes. Increased expression of both ETS1 mRNA and the protein has been observed in human fibroblasts after TNF-α or IL-1β stimulation [28]. PEA3 (a mouse protein corresponding to ETV5) inhibits tumorigenesis in vivo [43]. Moreover, ETV5 and ETS1 can cooperate with c-Jun/c-Fos [44,45], potential regulators of apoptosis in many cell types and specially in the mammalian nervous system [46]. The second gene regulated by TNF-α is NPTX2, encoding neuronal pentraxin II. Pentraxins are a family of proteins that include C-reactive protein and serum amyloid P. They have been found in the brain plaques characteristic of Alzheimer's disease and are toxic to neuronal cell cultures [47,48]. Furthermore, the expression of NPTX3 is increased in response to TNF-α or IL-1β stimulation via activation of NFκB [29,30]. The regulation of the pentraxin gene family by C2-ceramide treatment is consistent with our previous studies showing NFκB activation by C2-ceramide in PC12 cells and in primary cultures of neurons [10,19]. The last two genes known to be regulated by TNF-α and identified in our model are COL18A1, encoding type XVIII collagen alpha 1, and TNFAIP1, encoding TNF-α-induced protein 1. These proteins, downregulated by C2-ceramide, are modulated by TNF-α in various cell types [31,32,33].

We also identified four genes encoding proteins known to participate in TNF-α-activated signal transduction pathways. Thus AXL, upregulated by a factor of 3.65 (Table 1), encodes a tyrosine kinase receptor. Signaling through this receptor is reported to protect against TNF-α-induced apoptosis in fibroblasts and its absence increases apoptosis after serum deprivation [34]. Interestingly, ARK, the mouse protein corresponding to AXL, activates the survival pathway mediated by the serine-threonine kinase Akt [49], which is negatively regulated by ceramide [16,17,50], and is also reported to modulate ceramide synthesis [51]. The second gene we identified is BIRC1, encoding baculoviral IAP repeat-containing 1 protein. This protein, putatively involved in spinal muscular atrophy [52], is an inhibitor of cell death induced by various apoptotic stimuli, including TNF-α [35]. The third identified gene, RSU1, encodes Ras suppressor protein 1, which is involved in TNF-α signaling by blocking the Ras-dependent response. Levels of both RSU1 mRNA and protein have been correlated with a decrease in growth rate and tumorigenic potential in U251 glioblastoma cells [53] and it induces growth arrest in PC12 cells [36]. This is consistent with the report that ceramide regulates apoptosis via modulation of the Ras signaling pathway [18]. In addition, RSU1 has been identified as an inhibitor of Jun kinase activation [37]. This point is interesting, as the fourth gene presenting in this group, MAPK10/J.NK3, encoding the JNK family member mitogen-activated protein kinase 10, is downregulated by C2-ceramide in our model.

The identification of these eight genes, which are involved in the TNF-α signaling pathway, in C2-ceramide treated PC12 cells, suggests that their modulation of expression by TNF-α could be the result of a ceramide-dependent mechanism.

Commitment to apoptosis: upregulation of pro-apoptotic genes and downregulation of anti-apoptotic genes by the ceramide pathway

Twenty genes regulated by C2-ceramide correspond to genes known to be involved in regulation of apoptosis and/or cell growth (Figure 5, Table 2). Twelve of these genes are known to be associated with oncogenesis and four with neuronal disorders. Of the upregulated genes, 10 out of 14 are known to be associated with a pro-apoptotic or anti-proliferation process and 3 out of 14 are mainly implicated in protection of the cell against cytotoxicity or damage. Of the downregulated genes, 4 out of 6 are associated with an anti-apoptotic or a proliferation process. This highlights the fact that the cells are engaged in programmed cell death. The putative roles of these genes are illustrated in Figure 7, which focuses on the pro-apoptotic or anti-proliferation process versus anti-apoptotic or proliferation processes.

Briefly, of the known pro-apoptotic or anti-proliferative genes that are upregulated in our model, RPL4 encodes the ribosomal protein L4 that has been shown to be transcriptionally stimulated prior to apoptosis induced by the 5-azacytidine in the PC12 cells [54]. PDCD6IP, upregulated by C2-ceramide in our model, encodes a protein that interacts with ALG2, a Ca2+-binding protein that is required for apoptosis induced by diverse stimuli, including ceramide treatment [55,56,57]. M6PR encodes the cation-dependent mannose-6-phosphate receptor, which has been implicated in retinoid-induced apoptosis [58]. NMA, encoding a putative transmembrane protein, is expressed at low levels in metastatic human melanoma cell lines and xenografts, and is completely absent in highly metastatic human melanoma cell lines [59]. APCL, encoding adenomatous polyposis coli like protein, is a tumor-suppressor gene [60]. METAP2 encodes methionine aminopeptidase eIF-2-associated p67, which interacts with eukaryotic translation initiation factor eIF-2 [61] and could regulate p53 signaling [62]. BAT3, downregulated in some transformed cells, encodes HLA-B associated transcript-3, which interacts with the tumor-suppressor protein DAN that contains growth or tumor suppressive activity in vitro [63]. PHB encodes the protein prohibitin, a potential tumor-suppressor protein that binds to the retinoblastoma (Rb) protein and represses E2F transcriptional activity [64,65]. BNIP3L, encoding BCL2/adenovirus E1B 19 kD-interacting protein 3-like, is a pro-apoptotic gene which has a growth-inhibitory effect on cancer cells [66]. CDH13, encoding cadherin 13, is significantly downregulated in human breast carcinoma cell lines and breast cancer, whereas its overexpression decreases tumor-cell growth [67,68].

Of the known anti-apoptotic or proliferative genes that are downregulated in our model, LOC51582 encodes an antizyme inhibitor, which regulates the antizyme activity proposed to be involved in the polyamine biosynthesis pathway [69,70]. Interestingly, overexpression of antizyme inhibits cell growth [71,72], whereas LOC51582 is downregulated in our model. This observation is consistent with a role of antizyme in the apoptotic process and suggests that ceramide can regulate its activity. LOC51283, a regulator of the activity of the Bcl-2 family proteins, encodes a novel apoptosis regulator, which has been identified as an inhibitor of Bax-induced cell death [73]. Its downregulation by C2-ceramide confirms its involvement in the ceramide-dependent regulation of cell death. The last gene presented here, MET, encodes the MET proto-oncogene, known to be a receptor of the hepatocyte growth factor that has been described to protect neuronal cells from apoptosis via the phosphatidylinositol-3 kinase/Akt pathway [74].

Four genes out of the other genes presented in Table 2 have already been implicated in neuronal disorders, suggesting that ceramide may be a key second messenger in these pathologies. The upregulation of the glutamate receptor gene (GRIA2) seems to be an indicator of tolerance to ischemia [75]. The absence of somatostatin, encoded by SST (downregulated in our model), is associated with apoptotic neurons in patients with Alzheimer's disease [76]. SMN, encoding Survival of motor neuron 2, downregulated by C2-ceramide, strongly contributes to the severity of the spinal muscular atrophy [77]. MUT mRNA is upregulated in ischemia, in relation to a decrease in the accumulation of its neurotoxic metabolite [78].

In conclusion, our cell culture model has enabled us to establish a profile of gene expression during the effector phase of ceramide-mediated cell death. In spite of the stringency of the criteria adopted for differential hybridization, a large number of cDNA clones, 239 of the 9,120 in our cDNA array derived from a normalized infant brain library, correspond to genes up- or downregulated by C2-ceramide treatment. Already-known genes account for 179 of the transcripts, 113 of which have a putative function.

On the basis of their putative functions, we have made an attempt at classifying these transcripts, first with respect to known effects of ceramide or ceramide-mediated transduction systems, then with respect to regulation of cell growth and apoptosis. The 30 genes in Tables 1 and 2 met these criteria, validating the approach and suggesting that the other modulated genes may also be relevant with regard to the progression of the cell-death mechanisms. These genes were classified as having no obvious relation to cell death or survival (Table 3), no known function (Table 4) or as poorly characterized (Table 5). As a result of our study, these genes now have tentative functions. The full list can be consulted with the relevant data on the dedicated website [24].

Interestingly, given the large number of genes known to be modulated by NFκB in the immune system [79], it was surprising that only pentraxin was detected in our model. This suggests either that NFκB is less important in neurons than in lymphocytes, or that its targets are different. Conversely, the transcriptional regulators responsible for the differential expression of the genes detected in our study remain to be discovered. In any case, our results show that transcriptional regulation plays an important role in ceramide-mediated cell death and that some of the modulated transcripts, in agreement with published studies, are involved in other cell-death mechanisms as well.

Materials and methods

Cell culture

Rat PC12 cells [80], which acquire a neuronal phenotype in the presence of nerve growth factor (NGF), were plated at a density of 2,000-3,000 cells/cm2 in 75 cm2 culture flasks coated with polyethylenimine (1 mg/ml) in Leibovitz modified L15 medium (Gibco BRL) supplemented with 2% horse serum and 150 ng/ml NGF (grade II; Alomone Labs, Jerusalem, Israel) as previously described [81]. Apoptosis was induced, after 6 days in the presence of NGF, with the cell-permeant C2 analog of ceramide (C2-ceramide), N-acetylsphingosine (Biomol Research Laboratories, Plymouth Meeting, PA), at a concentration of 25 μM. As negative control, an inactive C2 analog of ceramide (C2-dihydroceramide), N-acetylsphinganine (Biomol Research Laboratories), was used in the same condition as C2-ceramide.

Morphological characterization of apoptosis and cell counts

Neurite retraction and cell shrinkage were visualized by phase-contrast microscopy. Condensed and fragmented nuclei were made visible in situ as described in [7], by intercalation into nuclear DNA of the fluorescent probe propidium iodide. Propidium iodide, which only enters dead cells that have become permeable, was visualized by epifluorescence with a rhodamine filter (excitation, 548-580 nm; emission, 580-610 nm). Viability was quantified by counting cells in at least 10 randomly chosen fields with a 20x objective. The percentage of cells excluding the vital dye propidium iodide was calculated at each time point after the beginning of C2-ceramide or C2-dihydroceramide treatment with respect to the corresponding control.

Measurement of caspase-3-like activity

Caspase-3-like activity was measured using the CaspACE Assay system (Promega, Madison, WI). Cell extracts containing equivalent amounts of protein were used to measure DEVDase (caspase-3-like) activity: the chromophore p-nitroaniline (pNA), released from the colorimetric substrate (Ac-DEVD-pNA) upon cleavage by DEVDase produces a yellow color that is monitored by a photometer at 405 nm.

Preparation of the cDNA macroarray

cDNA clones from a normalized infant brain library (library 1NIB; [20]) were randomly selected to provide a set of 9,120 cDNA clones. The 3' and/or 5' ends of these clones had been previously sequenced [25]. The sequences, registered in GenBank [82], were compared to those in public data bases, permitting tentative identification of the corresponding gene transcripts. The cDNA clones were used to prepare PCR products using oligonucleotide primers complementary to sequences in the vector. They were spotted by robot (Flexis; Perkin Elmer, Shelton, CT) at medium density (25 PCR products/cm2) on nylon membranes (Hybond-N+; Amersham Biosciences, Uppsala, Sweden) as previously described [83]. The entire collection of 9,120 cDNA clones was spotted on a set of four filters.

Purification of poly(A)+ mRNA

Total RNA was extracted from control PC12 cultures and from PC12 cultures treated with C2-ceramide or C2-dihydroceramide (approximately 106 cells) with the RNeasy midi kit (Qiagen, Courtaboeuf, France), according to the manufacturer's instructions. The integrity of the RNA was confirmed by agarose gel electrophoresis. Poly(A)+ mRNA was extracted from total RNA with oligo(dT)-conjugated magnetic beads (Dynabeads; Dynal, Oslo, Norway), as described in the manufacturer's protocols.

Complex cDNA target synthesis

Complex cDNA targets were synthesised by reverse transcription of 500 ng poly(A)+ mRNA extracted from control, C2-dihydroceramide- or C2-ceramide-treated PC12 cells. The reaction was performed with the Superscript™ Preamplification System (Invitrogen) as previously described [84]. The reaction mixture contained random-oligonucleotide primers (500 ng), 50 μCi [α-33P]dATP, 3,000 Ci/mmol (Amersham), 500 μM d(T, C, G)TP (Amersham) and 50 μM dideoxyGTP (Invitrogen).

Filter hybridization

The filters were prehybridized at 68°C for 30 min in ExpressHyb hybridization solution (Clontech, Palo Alto, CA), hybridized for 2 h in the same solution to which the radiolabeled complex cDNA target was added, then washed twice for 30 min at 25°C in standard saline citrate (SSC) 1×/0.1% sodium dodecyl sulfate (SDS) and twice for 30 min at 25°C in SSC 0.1×/0.1% SDS. The washed filters were exposed to phosphorus screens (Molecular Dynamics, Sunnyvlae, CA) for 16 h.

Hybridization signal quantitation

Image acquisition was carried out with the Phosphorlmager (Molecular Dynamics). The hybridization signal corresponding to each cDNA clone was quantitated with a specifically designed software (XdotsReader; Cose, Dugny, France) and the local background signal was subtracted. The intensity of the hybridization signal for each clone was then divided by the average intensity of all the clones on each filter to obtain normalized values. Hybridization was done in quadruplicate so that, for each clone/target combination, four values were obtained, compared and validated if at least three out of the four values were similar (SD ± 25%). The final value assigned to each clone was the average of the validated values.

Northern blotting

Total RNA (20 μg) were fractionated under denaturing conditions in a 1.2% agarose gel and transferred onto a Hybond-N+ membrane (Amersham). Specific probes were generated from cDNA clones of interest by PCR using vector-specific primers. The PCR products were purified using the microcon kit (Amicon, Wageningen, The Netherlands) and radiolabeled by random priming (Gibco BRL). Oligonucleotides corresponding to 18S rRNA (control probe) were 32P-labeled using [γ-33P]ATP and T4 RNA kinase. For northern blot analysis, the blots were prehybridized 2 h in ULTRAhyb hybridization buffer (Ambion, Austin, TX), hybridized with the labelled probe (1-2 × 106 cpm/ml) for 16 h at 42°C in the same solution, and washed as for the high-density filters. The washed filters were exposed to phosphorus screens (Molecular Dynamics) for 48 h. The hybridization signal of the specific probes was analyzed with the ImageQuant software (Molecular Dynamics) and compared to the signal obtained with the control probe.

RT-PCR

Total RNA of PC12 cells cultured with or without C2-ceramide was purified according to the protocol described above. Total RNA (2 μg) were reverse transcribed using the Superscript™ Preamplification System (Invitrogen) according to the manufacturer's protocol. An aliquot of the reaction was then used for PCR amplification with the Advantage PCR kit (Clontech) and primers specific to the gene of interest. The amplification products were visualized after electrophoresis in a 1.5% agarose gel with ethidium bromide. The signals were analyzed with ImageQuant software and compared to HPRT (hypoxanthine phosphoribosyl transferase) as control gene.

Acknowledgments

Acknowledgements

This work was supported by the CNRS and grants from European Union to J.M. (TMR projet Neuril) and BIOMED2 programs (EURO-IMAGE Consortium, BMH4-CT-97-2284) to C.A. C.M. was supported by Genome Express and C.D. acknowledges fellowships from the Fédération Française des Groupements Parkinsoniens (FFGP) and the Association pour la Recherche sur le Cancer (ARC).

References

  1. Hannun YA. Functions of ceramide in coordinating cellular responses to stress. Science. 1996;274:1855–1859. doi: 10.1126/science.274.5294.1855. [DOI] [PubMed] [Google Scholar]
  2. Mathias S, Pena LA, Kolesnick RN. Signal transduction of stress via ceramide. Biochem J. 1998;335:465–480. doi: 10.1042/bj3350465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Furuya S, Mitoma J, Makino A, Hirabayashi Y. Ceramide and its interconvertible metabolite sphingosine function as indispensable lipid factors involved in survival and dendritic differentiation of cerebellar Purkinje cells. J Neurochem. 1998;71:366–377. doi: 10.1046/j.1471-4159.1998.71010366.x. [DOI] [PubMed] [Google Scholar]
  4. Obeid LM, Venable ME. Signal transduction in cellular senescence. J Am Geriatr Soc. 1997;45:361–366. doi: 10.1111/j.1532-5415.1997.tb00954.x. [DOI] [PubMed] [Google Scholar]
  5. Kolesnick R, Golde DW. The sphingomyelin pathway in tumor necrosis factor and interleukin-1 signaling. Cell. 1994;77:325–328. doi: 10.1016/0092-8674(94)90147-3. [DOI] [PubMed] [Google Scholar]
  6. Kyprianou N, Alexander RB, Isaacs JT. Activation of programmed cell death by recombinant human tumor necrosis factor plus topoisomerase II-targeted drugs in L929 tumor cells. J Natl Cancer Inst. 1991;83:346–350. doi: 10.1093/jnci/83.5.346. [DOI] [PubMed] [Google Scholar]
  7. Brugg B, Michel PP, Agid Y, Ruberg M. Ceramide induces apoptosis in cultured mesencephalic neurons. J Neurochem. 1996;66:733–739. doi: 10.1046/j.1471-4159.1996.66020733.x. [DOI] [PubMed] [Google Scholar]
  8. Mitoma J, Ito M, Furuya S, Hirabayashi Y. Bipotential roles of ceramide in the growth of hippocampal neurons: promotion of cell survival and dendritic outgrowth in dose- and developmental stage-dependent manners. J Neurosci Res. 1998;51:712–722. doi: 10.1002/(SICI)1097-4547(19980315)51:6<712::AID-JNR5>3.3.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  9. Hartfield PJ, Mayne GC, Murray AW. Ceramide induces apoptosis in PC12 cells. FEBS Lett. 1997;401:148–152. doi: 10.1016/s0014-5793(96)01460-3. [DOI] [PubMed] [Google Scholar]
  10. France-Lanord V, Brugg B, Michel PP, Agid Y, Ruberg M. Mitochondrial free radical signal in ceramide-dependent apoptosis: a putative mechanism for neuronal death in Parkinson's disease. J Neurochem. 1997;69:1612–1621. doi: 10.1046/j.1471-4159.1997.69041612.x. [DOI] [PubMed] [Google Scholar]
  11. Lambeng N, Michel PP, Brugg B, Agid Y, Ruberg M. Mechanisms of apoptosis in PC12 cells irreversibly differentiated with nerve growth factor and cyclic AMP. Brain Res. 1999;821:60–68. doi: 10.1016/s0006-8993(99)01061-6. [DOI] [PubMed] [Google Scholar]
  12. Wolff RA, Dobrowsky RT, Bielawska A, Obeid LM, Hannun YA. Role of ceramide-activated protein phosphatase in ceramide-mediated signal transduction. J Biol Chem. 1994;269:19605–19609. [PubMed] [Google Scholar]
  13. Reyes JG, Robayna IG, Delgado PS, Gonzalez IH, Aguiar JQ, Rosas FE, Fanjul LF, Galarreta CM. c-Jun is a downstream target for ceramide-activated protein phosphatase in A431 cells. J Biol Chem. 1996;271:21375–21380. doi: 10.1074/jbc.271.35.21375. [DOI] [PubMed] [Google Scholar]
  14. Su B, Karin M. Mitogen-activated protein kinase cascades and regulation of gene expression. Curr Opin Immunol. 1996;8:402–411. doi: 10.1016/s0952-7915(96)80131-2. [DOI] [PubMed] [Google Scholar]
  15. Willaime S, Vanhoutte P, Caboche J, Lemaigre-Dubreuil Y, Mariani J, Brugg B. Ceramide-induced apoptosis in cortical neurons is mediated by an increase in p38 phosphorylation and not by the decrease in ERK phosphorylation. Eur J Neurosci. 2001;13:2037–2046. doi: 10.1046/j.0953-816x.2001.01581.x. [DOI] [PubMed] [Google Scholar]
  16. Schubert KM, Scheid MP, Duronio V. Ceramide inhibits protein kinase B/Akt by promoting dephosphorylation of serine 473. J Biol Chem. 2000;275:13330–13335. doi: 10.1074/jbc.275.18.13330. [DOI] [PubMed] [Google Scholar]
  17. Salinas M, Lopez-Valdaliso R, Martin D, Alvarez A, Cuadrado A. Inhibition of PKB/Akt1 by C2-ceramide involves activation of ceramide-activated protein phosphatase in PC12 cells. Mol Cell Neurosci. 2000;15:156–169. doi: 10.1006/mcne.1999.0813. [DOI] [PubMed] [Google Scholar]
  18. Basu S, Bayoumy S, Zhang Y, Lozano J, Kolesnick R. BAD enables ceramide to signal apoptosis via Ras and Raf-1. J Biol Chem. 1998;273:30419–30426. doi: 10.1074/jbc.273.46.30419. [DOI] [PubMed] [Google Scholar]
  19. Hunot S, Brugg B, Ricard D, Michel PP, Muriel MP, Ruberg M, Faucheux BA, Agid Y, Hirsch EC. Nuclear translocation of NF-kappaB is increased in dopaminergic neurons of patients with Parkinson disease. Proc Natl Acad Sci USA. 1997;94:7531–7536. doi: 10.1073/pnas.94.14.7531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Soares MB, Bonaldo MF, Jelene P, Su L, Lawton L, Efstratiadis A. Construction and characterization of a normalized cDNA library. Proc Natl Acad Sci USA. 1994;91:9228–9232. doi: 10.1073/pnas.91.20.9228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Nicholson DW. Caspase structure, proteolytic substrates, and function during apoptotic cell death. Cell Death Differ. 1999;6:1028–1042. doi: 10.1038/sj/cdd/4400598. [DOI] [PubMed] [Google Scholar]
  22. Hartfield PJ, Bilney AJ, Murray AW. Neurotrophic factors prevent ceramide-induced apoptosis downstream of c-Jun N-terminal kinase activation in PC12 cells. J Neurochem. 1998;71:161–169. doi: 10.1046/j.1471-4159.1998.71010161.x. [DOI] [PubMed] [Google Scholar]
  23. Houlgatte R, Mariage-Samson R, Duprat S, Tessier A, Bentolila S, Lamy B, Auffray C. The Genexpress Index: a resource for gene discovery and the genic map of the human genome. Genome Res. 1995;5:272–304. doi: 10.1101/gr.5.3.272. [DOI] [PubMed] [Google Scholar]
  24. Identification of genes involved in ceramide-dependent neuronal apoptosis using cDNA array http://idefix.ers1984.vjf.cnrs.fr/APOPTOSE/ [DOI] [PMC free article] [PubMed]
  25. Jayadev S, Hayter HL, Andrieu N, Gamard CJ, Liu B, Balu R, Hayakawa M, Ito F, Hannun YA. Phospholipase A2 is necessary for tumor necrosis factor alpha-induced ceramide generation in L929 cells. J Biol Chem. 1997;272:17196–17203. doi: 10.1074/jbc.272.27.17196. [DOI] [PubMed] [Google Scholar]
  26. Janes RW, Munroe PB, Mitchison HM, Gardiner RM, Mole SE, Wallace BA. A model for Batten disease protein CLN3: functional implications from homology and mutations. FEBS Lett. 1996;399:75–77. doi: 10.1016/s0014-5793(96)01290-2. [DOI] [PubMed] [Google Scholar]
  27. The International Batten Disease Consortium Isolation of a novel gene underlying Batten disease, CLN3. Cell. 1995;82:949–957. doi: 10.1016/0092-8674(95)90274-0. [DOI] [PubMed] [Google Scholar]
  28. Gilles F, Raes MB, Stehelin D, Vandenbunder B, Fafeur V. The c-ets-1 proto-oncogene is a new early-response gene differentially regulated by cytokines and growth factors in human fibroblasts. Exp Cell Res. 1996;222:370–378. doi: 10.1006/excr.1996.0046. [DOI] [PubMed] [Google Scholar]
  29. Basile A, Sica A, d'Aniello E, Breviario F, Garrido G, Castellano M, Mantovani A, Introna M. Characterization of the promoter for the human long pentraxin PTX3. Role of NF-kappaB in tumor necrosis factor-alpha and interleukin-1beta regulation. J Biol Chem. 1997;272:8172–8178. doi: 10.1074/jbc.272.13.8172. [DOI] [PubMed] [Google Scholar]
  30. Mantovani A, Muzio M, Ghezzi P, Colotta C, Introna M. Regulation of inhibitory pathways of the interleukin-1 system. Ann NY Acad Sci. 1998;840:338–351. doi: 10.1111/j.1749-6632.1998.tb09573.x. [DOI] [PubMed] [Google Scholar]
  31. Solis-Herruzo JA, Brenner DA, Chojkier M. Tumor necrosis factor alpha inhibits collagen gene transcription and collagen synthesis in cultured human fibroblasts. J Biol Chem. 1988;263:5841–5845. [PubMed] [Google Scholar]
  32. Hernandez-Munoz I, de la Torre P, Sanchez-Alcazar JA, Garcia I, Santiago E, Munoz-Yague MT, Solis-Herruzo JA. Tumor necrosis factor alpha inhibits collagen alpha 1(I) gene expression in rat hepatic stellate cells through a G protein. Gastroenterology. 1997;113:625–640. doi: 10.1053/gast.1997.v113.pm9247485. [DOI] [PubMed] [Google Scholar]
  33. Wolf FW, Marks RM, Sarma V, Byers MG, Katz RW, Shows TB, Dixit VM. Characterization of a novel tumor necrosis factor-alpha-induced endothelial primary response gene. J Biol Chem. 1992;267:1317–1326. [PubMed] [Google Scholar]
  34. Bellosta P, Zhang Q, Goff SP, Basilico C. Signaling through the ARK tyrosine kinase receptor protects from apoptosis in the absence of growth stimulation. Oncogene. 1997;15:2387–2397. doi: 10.1038/sj/onc/1201419. [DOI] [PubMed] [Google Scholar]
  35. Liston P, Roy N, Tamai K, Lefebvre C, Baird S, Cherton-Horvat G, Farahani R, McLean M, Ikeda JE, MacKenzie A, Korneluk RG. Suppression of apoptosis in mammalian cells by NAIP and a related family of IAP genes. Nature. 1996;379:349–353. doi: 10.1038/379349a0. [DOI] [PubMed] [Google Scholar]
  36. Masuelli L, Ettenberg S, Vasaturo F, Vestergaard-Sykes K, Cutler ML. The ras suppressor, RSU-1, enhances nerve growth factor-induced differentiation of PC12 cells and induces p21CIP expression. Cell Growth Differ. 1999;10:555–564. [PubMed] [Google Scholar]
  37. Masuelli L, Cutler ML. Increased expression of the Ras suppressor Rsu-1 enhances Erk-2 activation and inhibits Jun kinase activation. Mol Cell Biol. 1996;16:5466–5476. doi: 10.1128/mcb.16.10.5466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Underwood KW, Song C, Kriz RW, Chang XJ, Knopf JL, Lin LL. A novel calcium-independent phospholipase A2, cPLA2-gamma, that is prenylated and contains homology to cPLA2. J Biol Chem. 1998;273:21926–21932. doi: 10.1074/jbc.273.34.21926. [DOI] [PubMed] [Google Scholar]
  39. Hoeck WG, Ramesha CS, Chang DJ, Fan N, Heller RA. Cytoplasmic phospholipase A2 activity and gene expression are stimulated by tumor necrosis factor: dexamethasone blocks the induced synthesis. Proc Natl Acad Sci USA. 1993;90:4475–4479. doi: 10.1073/pnas.90.10.4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Wu T, Ikezono T, Angus CW, Shelhamer JH. Tumor necrosis factor-alpha induces the 85-kDa cytosolic phospholipase A2 gene expression in human bronchial epithelial cells. Biochim Biophys Acta. 1996;1310:175–184. doi: 10.1016/0167-4889(95)00143-3. [DOI] [PubMed] [Google Scholar]
  41. Hayakawa M, Jayadev S, Tsujimoto M, Hannun YA, Ito F. Role of ceramide in stimulation of the transcription of cytosolic phospholipase A2 and cyclooxygenase 2. Biochem Biophys Res Commun. 1996;220:681–686. doi: 10.1006/bbrc.1996.0464. [DOI] [PubMed] [Google Scholar]
  42. Puranam KL, Guo WX, Qian WH, Nikbakht K, Boustany RM. CLN3 defines a novel antiapoptotic pathway operative in neurode-generation and mediated by ceramide. Mol Genet Metab. 1999;66:294–308. doi: 10.1006/mgme.1999.2834. [DOI] [PubMed] [Google Scholar]
  43. Xing X, Wang SC, Xia W, Zou Y, Shao R, Kwong KY, Yu Z, Zhang S, Miller S, Huang L, Hung MC. The ets protein PEA3 suppresses HER-2/neu overexpression and inhibits tumorigenesis. Nat Med. 2000;6:189–195. doi: 10.1038/72294. [DOI] [PubMed] [Google Scholar]
  44. Nakae K, Nakajima K, Inazawa J, Kitaoka T, Hirano T. ERM, a PEA3 subfamily of Ets transcription factors, can cooperate with c-Jun. J Biol Chem. 1995;270:23795–23800. doi: 10.1074/jbc.270.40.23795. [DOI] [PubMed] [Google Scholar]
  45. Basuyaux JP, Ferreira E, Stehelin D, Buttice G. The Ets transcription factors interact with each other and with the c-Fos/ c-Jun complex via distinct protein domains in a DNA-dependent and -independent manner. J Biol Chem. 1997;272:26188–26195. doi: 10.1074/jbc.272.42.26188. [DOI] [PubMed] [Google Scholar]
  46. Herdegen T, Leah JD. Inducible and constitutive transcription factors in the mammalian nervous system: control of gene expression by Jun, Fos and Krox, and CREB/ATF proteins. Brain Res Brain Res Rev. 1998;28:370–490. doi: 10.1016/s0165-0173(98)00018-6. [DOI] [PubMed] [Google Scholar]
  47. Duong T, Nikolaeva M, Acton PJ. C-reactive protein-like immunoreactivity in the neurofibrillary tangles of Alzheimer's disease. Brain Res. 1997;749:152–156. doi: 10.1016/s0006-8993(96)01359-5. [DOI] [PubMed] [Google Scholar]
  48. Duong T, Acton PJ, Johnson RA. The in vitro neuronal toxicity of pentraxins associated with Alzheimer's disease brain lesions. Brain Res. 1998;813:303–312. doi: 10.1016/s0006-8993(98)00966-4. [DOI] [PubMed] [Google Scholar]
  49. Allen MP, Zeng C, Schneider K, Xiong X, Meintzer MK, Bellosta P, Basilico C, Varnum B, Heidenreich KA, Wierman ME. Growth arrest-specific gene 6 (Gas6)/adhesion related kinase (Ark) signaling promotes gonadotropin-releasing hormone neuronal survival via extracellular signal-regulated kinase (ERK) and Akt. Mol Endocrinol. 1999;13:191–201. doi: 10.1210/mend.13.2.0230. [DOI] [PubMed] [Google Scholar]
  50. Zhou H, Summers SA, Birnbaum MJ, Pittman RN. Inhibition of Akt kinase by cell-permeable ceramide and its implications for ceramide-induced apoptosis. J Biol Chem. 1998;273:16568–16575. doi: 10.1074/jbc.273.26.16568. [DOI] [PubMed] [Google Scholar]
  51. Goswami R, Kilkus J, Dawson SA, Dawson G. Overexpression of Akt (protein kinase B) confers protection against apoptosis and prevents formation of ceramide in response to pro-apoptotic stimuli. J Neurosci Res. 1999;57:884–893. doi: 10.1002/(SICI)1097-4547(19990915)57:6<884::AID-JNR14>3.3.CO;2-1. [DOI] [PubMed] [Google Scholar]
  52. Roy N, Mahadevan MS, McLean M, Shutler G, Yaraghi Z, Farahani R, Baird S, Besner-Johnston A, Lefebvre C, Kang X, et al. The gene for neuronal apoptosis inhibitory protein is partially deleted in individuals with spinal muscular atrophy. Cell. 1995;80:167–178. doi: 10.1016/0092-8674(95)90461-1. [DOI] [PubMed] [Google Scholar]
  53. Tsuda T, Marinetti MR, Masuelli L, Cutler ML. The Ras suppressor RSU-1 localizes to 10p13 and its expression in the U251 glioblastoma cell line correlates with a decrease in growth rate and tumorigenic potential. Oncogene. 1995;11:397–403. [PubMed] [Google Scholar]
  54. Kajikawa S, Nakayama H, Suzuki M, Takashima A, Murayama O, Nishihara M, Takahashi M, Doi K. Increased expression of rat ribosomal protein L4 mRNA in 5-azacytidine-treated PC12 cells prior to apoptosis. Biochem Biophys Res Commun. 1998;252:220–224. doi: 10.1006/bbrc.1998.9633. [DOI] [PubMed] [Google Scholar]
  55. Vito P, Pellegrini L, Guiet C, D'Adamio L. Cloning of AIP1, a novel protein that associates with the apoptosis-linked gene ALG-2 in a Ca2+-dependent reaction. J Biol Chem. 1999;274:1533–1540. doi: 10.1074/jbc.274.3.1533. [DOI] [PubMed] [Google Scholar]
  56. Vito P, Lacana E, D'Adamio L. Interfering with apoptosis: Ca(2+)-binding protein ALG-2 and Alzheimer's disease gene ALG-3. Science. 1996;271:521–525. doi: 10.1126/science.271.5248.521. [DOI] [PubMed] [Google Scholar]
  57. Lacana E, Ganjei JK, Vito P, D'Adamio L. Dissociation of apoptosis and activation of IL-1beta-converting enzyme/Ced-3 proteases by ALG-2 and the truncated Alzheimer's gene ALG-3. J Immunol. 1997;158:5129–5135. [PubMed] [Google Scholar]
  58. Kang JX, Bell J, Beard RL, Chandraratna RA. Mannose 6-phosphate/insulin-like growth factor II receptor mediates the growth-inhibitory effects of retinoids. Cell Growth Differ. 1999;10:591–600. [PubMed] [Google Scholar]
  59. Degen WG, Weterman MA, van Groningen JJ, Cornelissen IM, Lemmers JP, Agterbos MA, Geurts van Kessel A, Swart GW, Bloemers HP. Expression of nma, a novel gene, inversely correlates with the metastatic potential of human melanoma cell lines and xenografts. Int J Cancer. 1996;65:460–465. doi: 10.1002/(SICI)1097-0215(19960208)65:4<460::AID-IJC12>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  60. Nakagawa H, Koyama K, Murata Y, Morito M, Akiyama T, Nakamura Y. APCL, a central nervous system-specific homologue of adenomatous polyposis coli tumor suppressor, binds to p53-binding protein 2 and translocates it to the perinucleus. Cancer Res. 2000;60:101–105. [PubMed] [Google Scholar]
  61. Ray MK, Datta B, Chakraborty A, Chattopadhyay A, Meza-Keuthen S, Gupta NK. The eukaryotic initiation factor 2-associated 67-kDa polypeptide (p67) plays a critical role in regulation of protein synthesis initiation in animal cells. Proc Natl Acad Sci USA. 1992;89:539–543. doi: 10.1073/pnas.89.2.539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Cuddihy AR, Li S, Tam NW, Wong AH, Taya Y, Abraham N, Bell JC, Koromilas AE. Double-stranded-RNA-activated protein kinase PKR enhances transcriptional activation by tumor suppressor p53. Mol Cell Biol. 1999;19:2475–2484. doi: 10.1128/mcb.19.4.2475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Ozaki T, Hanaoka E, Naka M, Nakagawara A, Sakiyama S. Cloning and characterization of rat BAT3 cDNA. DNA Cell Biol. 1999;18:503–512. doi: 10.1089/104454999315222. [DOI] [PubMed] [Google Scholar]
  64. Wang S, Nath N, Fusaro G, Chellappan S. Rb and prohibitin target distinct regions of E2F1 for repression and respond to different upstream signals. Mol Cell Biol. 1999;19:7447–7460. doi: 10.1128/mcb.19.11.7447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Wang S, Nath N, Adlam M, Chellappan S. Prohibitin, a potential tumor suppressor, interacts with RB and regulates E2F function. Oncogene. 1999;18:3501–3510. doi: 10.1038/sj/onc/1202684. [DOI] [PubMed] [Google Scholar]
  66. Matsushima M, Fujiwara T, Takahashi E, Minaguchi T, Eguchi Y, Tsujimoto Y, Suzumori K, Nakamura Y. Isolation, mapping, and functional analysis of a novel human cDNA (BNIP3L) encoding a protein homologous to human NIP3. Genes Chromosomes Cancer. 1998;21:230–235. [PubMed] [Google Scholar]
  67. Lee SW. H-cadherin, a novel cadherin with growth inhibitory functions and diminished expression in human breast cancer. Nat Med. 1996;2:776–782. doi: 10.1038/nm0796-776. [DOI] [PubMed] [Google Scholar]
  68. Lee SW, Reimer CL, Campbell DB, Cheresh P, Duda RB, Kocher O. H-cadherin expression inhibits in vitro invasiveness and tumor formation in vivo. Carcinogenesis. 1998;19:1157–1159. doi: 10.1093/carcin/19.6.1157. [DOI] [PubMed] [Google Scholar]
  69. Pegg AE, Wechter RS, Clark RS, Wiest L, Erwin BG. Acetylation of decarboxylated S-adenosylmethionine by mammalian cells. Biochemistry. 1986;25:379–384. doi: 10.1021/bi00350a016. [DOI] [PubMed] [Google Scholar]
  70. Tabor CW, Tabor H. Polyamines. Annu Rev Biochem. 1984;53:749–790. doi: 10.1146/annurev.bi.53.070184.003533. [DOI] [PubMed] [Google Scholar]
  71. Murakami Y, Ichiba T, Matsufuji S, Hayashi S. Cloning of antizyme inhibitor, a highly homologous protein to ornithine decarboxylase. J Biol Chem. 1996;271:3340–3342. doi: 10.1074/jbc.271.7.3340. [DOI] [PubMed] [Google Scholar]
  72. Iwata S, Sato Y, Asada M, Takagi M, Tsujimoto A, Inaba T, Yamada T, Sakamoto S, Yata J, Shimogori T, et al. Anti-tumor activity of antizyme which targets the ornithine decarboxylase (ODC) required for cell growth and transformation. Oncogene. 1999;18:165–172. doi: 10.1038/sj/onc/1202275. [DOI] [PubMed] [Google Scholar]
  73. Zhang H, Xu Q, Krajewski S, Krajewska M, Xie Z, Fuess S, Kitada S, Godzik A, Reed JC. BAR: An apoptosis regulator at the intersection of caspases and Bcl-2 family proteins. Proc Natl Acad Sci USA. 2000;97:2597–2602. doi: 10.1073/pnas.97.6.2597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Zhang L, Himi T, Morita I, Murota S. Hepatocyte growth factor protects cultured rat cerebellar granule neurons from apop-tosis via the phosphatidylinositol-3 kinase/Akt pathway. J Neurosci Res. 2000;59:489–496. doi: 10.1002/(SICI)1097-4547(20000215)59:4<489::AID-JNR3>3.3.CO;2-0. [DOI] [PubMed] [Google Scholar]
  75. Alsbo CW, Wrang ML, Moller F, Diemer NH. Is the AMPA receptor subunit GluR2 mRNA an early indicator of cell fate after ischemia? A quantitative single cell RT-PCR study. Brain Res. 2001;894:101–108. doi: 10.1016/s0006-8993(01)01985-0. [DOI] [PubMed] [Google Scholar]
  76. Li WP, Lai HW, Cheng SY, Yew DT. Somatostatin-positive neurons in the different parts of the brain in normal aging and Alzheimer's disease. Biol Signals. 1996;5:343–348. doi: 10.1159/000109207. [DOI] [PubMed] [Google Scholar]
  77. Lorson CL, Strasswimmer J, Yao JM, Baleja JD, Hahnen E, Wirth B, Le T, Burghes AH, Androphy EJ. SMN oligomerization defect correlates with spinal muscular atrophy severity. Nat Genet. 1998;19:63–66. doi: 10.1038/ng0598-63. [DOI] [PubMed] [Google Scholar]
  78. Narasimhan P, Sklar R, Murrell M, Swanson RA, Sharp FR. Methyl-malonyl-CoA mutase induction by cerebral ischemia and neurotoxicity of mitochondrial toxin methylmalonic acid. J Neurosci. 1996;22:7336–7346. doi: 10.1523/JNEUROSCI.16-22-07336.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Baeuerle PA, Henkel T. Function and activation of NF-kappa B in the immune system. Annu Rev Immunol. 1994;12:141–179. doi: 10.1146/annurev.iy.12.040194.001041. [DOI] [PubMed] [Google Scholar]
  80. Greene LA, Tischler AS. Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci USA. 1976;73:2424–2428. doi: 10.1073/pnas.73.7.2424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Michel PP, Vyas S, Agid Y. Synergistic differentiation by chronic exposure to cyclic AMP and nerve growth factor renders rat phaeochromocytoma PC12 cells totally dependent upon trophic support for survival. Eur J Neurosci. 1995;7:251–260. doi: 10.1111/j.1460-9568.1995.tb01061.x. [DOI] [PubMed] [Google Scholar]
  82. GenBank http://www.ncbi.nlm.nih.gov/Genbank
  83. Pietu G, Alibert O, Guichard V, Lamy B, Bois F, Leroy E, Mariage-Sampson R, Houlgatte R, Soularue P, Auffray C. Novel gene transcripts preferentially expressed in human muscles revealed by quantitative hybridization of a high density cDNA array. Genome Res. 1996;6:492–503. doi: 10.1101/gr.6.6.492. [DOI] [PubMed] [Google Scholar]
  84. Decraene C, Reguigne-Arnould I, Auffray C, Pietu G. Reverse transcription in the presence of dideoxynucleotides to increase the sensitivity of expression monitoring with cDNA arrays. Biotechniques. 1999;27:962–966. doi: 10.2144/99275st03. [DOI] [PubMed] [Google Scholar]
  85. UniGene http://www.ncbi.nlm.nih.gov/UniGene

Articles from Genome Biology are provided here courtesy of BMC

RESOURCES