Abstract
Immediate early gene X-1 (IEX-1) modulates apoptosis, cellular growth, mechanical strain-induced cardiac hypertrophy, and vascular intimal hyperplasia. To determine how IEX-1 alters apoptosis, we performed yeast two-hybrid studies using IEX-1 as the “bait” protein, and examined interactions between IEX-1 and proteins expressed by a human kidney cDNA expression library. We found that IEX-1 interacts with several proteins of which at least four are known to play a role in the regulation of apoptosis: (1) calcium-modulating cyclophilin ligand; (2) tumor necrosis factor-related apoptosis-inducing ligand (tumor necrosis factor superfamily, member 10); (3) ML-1 myeloid cell leukemia gene encoded protein; and (4) BAT3, a gene present in the major histo-compatibility complex. Our data suggest that IEX-1 may regulate apoptosis by directly interacting with various proteins involved in the control of apoptotic pathways.
Keywords: IEX-1; gly96; 1,25-Dihydroxyvitamin D; Apoptosis
The human immediate early gene X-1 (IEX-1, also known as Dif2, and murine orthologs gly96 and PRG1) plays a role in the control of apoptosis and cellular growth [1-20]. The gene (gly96) was initially identified in mouse fibroblasts as a serum-regulated, glycosylated, immediate early gene [1]. The messenger RNA of a similar gene (p22/PRG1) is induced in rat pancreatic cells by pituitary adenylate cyclase activating peptide (PACAP) [20]. The human ortholog of gly96 and PRG1, IEX-1, was identified as an X-radiation induced in fibroblasts by Kondratyev et al. [2], and as a UV-radiation and 1α, 25-dihydroxyvitamin D3-regulated gene in human keratinocytes [3,4].
Increased expression of IEX-1 is associated with an increase in the growth rate of keratinocytes and HeLa cells, and disruption of IEX-1 gene expression in HeLa cells and 293 cells is associated with a decrease in cellular proliferation [3,7,8,21]. IEX-1 has been shown to be induced in cardiomyocytes and vascular smooth muscle cells by mechanical stretch, and overexpression of IEX-1 in vascular smooth muscle cells protects against stretch-induced hypertrophy [9,15]. Injury-induced neo-intimal and vascular smooth muscle proliferation is reduced by transfection of carotid vessels with IEX-1 [10].
The rate of apoptosis is either enhanced or diminished depending upon the cell line and apoptosis-inducing reagent used. For example, in keratinocytes [22], HeLa cells [7], hepatocyte (Hc) [19], and 293 cells [8], IEX-1 increases the rate of apoptosis. With respect to the induction of apoptosis, the effect of IEX-1 can be explained partially by its inhibitory effect on survival signals. There is inhibition of TNF-α-induced activation and expression of phosphatidylinositol 3-kinase (PI3K)/Akt and blockade of expression of ML-1 myeloid cell leukemia gene encoded protein (MCL1), an anti-apoptotic BCL-2 family member, which is located downstream of Akt [19]. In some situations, expression of the gene in lymphocytes and Jurkat cells [11,12] results in a reduction of apoptosis and protection against TNF-induced apoptosis. Transgenic animals over-expressing IEX-1 in T-cells develop an extended duration of an effector phase of a specific immune response, and a lupus-like syndrome and apoptosis in T-lymphocytes is reduced [13,23].
IEX-1 is regulated by a variety of factors. IEX-1 expression is increased by serum, X-, and UV-irradiation, growth factors such as epidermal growth factor, inflammatory stimuli such as lipopolysaccharide and ceramide, retinoids such as all-trans-retinoic acid or cis-retinoic acid, and by over-expression of Sp1 in cells[1-3,6,10,11,20,24-31]. The gene is repressed by steroid hormones such as 1α, 25-dihydroxyvitamin D3 and by over-expression of p53 [3,4,30,32]. 1α, 25-Dihydroxyvitamin D3 also results in a redistribution of IEX-1 from the nucleus into the peri-nuclear and cytoplasmic space. The ratio of Sp1 to p53 within the cell appears to regulate the amount of IEX-1 expression present within the cell [30]. Slight modifications of IEX-1 transcription are observed by mutating putative elements for p300, Sox, NFκB, and AP4 within the promoter of the IEX-1 gene [30].
To gain further insights into the regulation of apoptosis by IEX-1, we performed yeast two-hybrid studies using IEX-1 as a “bait” protein, and a human kidney cDNA expression library as the source of expressed proteins. We now show that IEX-1 interacts with four proteins that are involved in the regulation of apoptosis, namely, calcium-modulating cyclophilin ligand (CAML), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL, tumor necrosis factor super family, member 10), ML-1, a myeloid leukemia cell gene encoded protein, and BAT3. A fifth protein, zyxin, which is part of the focal adhesion plaque, has recently been shown to modulate apoptosis as well.
Materials and methods
General
pGBKT7 and pACT-2 yeast expression vectors and a human kidney cDNA library (S1769, Lot No. OO60199) were purchased from Clontech (Palo Alto, CA). Protein A/G–Sepharose beads were purchased from Pierce (Rockford, IL). Anti-CAML Mouse Monoclonal Antibodies were generous gifts from Dr. Richard Bram (Mayo Clinic, Rochester). Antibodies against human IEX-l were generated as described earlier [33]. Protein concentrations were measured using the Bio-Rad reagent (Bio-Rad Laboratories, Hercules, CA).
Yeast two-hybrid screening
Yeast two-hybrid screening was performed as described by the manufacturer (Clontech Matchmaker Gal4 System 3, BD, Clontech, Palo Alto, CA). Full-length human IEX-1 complementary DNA generated by polymerase chain reaction methods and encompassing the IEX-1 coding region was ligated into EcoRI and BamHI sites of the yeast two-hybrid GAL-4 DNA-binding domain (BD) expression vector, pGBKT7 [34]. The chimeric plasmid was used to express the “bait” protein IEX-1 in Saccharomyces cerevisiae AH109 cells. A human kidney cDNA library was prepared by the manufacturer using the GAL-4 activation domain (AD) pACT-2 vector (Clontech BD Bioscience, S1769, Lot No. OO60199) and expressed proteins were examined for interactions with IEX-1. Preliminary experiments were carried out to show that IEX-1 expression does not activate transcription of genes itself in S. cerevisiae AH109 cells and that the IEX-1 protein is expressed in S. cerevisiae AH109 cells.
Saccharomyces cerevisiae
AH1O9 (MATa) cells were transformed with the chimeric IEX-1- pGBKT7 DNA-BD plasmid encoding full-length human IEX-l, and transformed cells were mated for 24 h at 30 °C with S. cerevisiae Y187 (MATα) cells pre-transformed with a human kidney cDNA library (S1769, Lot No. OO60199). The mating mixture was plated onto SD/-His/-Leu/-Trp selective medium plates. Plates were incubated for 8 days at 30 °C. For further selection of true positive clones, yeast colonies were re-plated onto SD/-Ade/-His/-Leu/-Trp plates containing X-α-Gal. The plates were incubated for 5 days at 30 °C. Positive blue colonies were picked and plasmid DNA was isolated and used to transfect Escherichia coli cells. Transformed E. coli cells were grown on an ampicillin containing nutrient agarose plate to select for clones containing the AD vector, pACT-2. Colonies were picked and the individual plasmids were amplified and purified [34]. Automated di-deoxy DNA sequencing [34,35] of purified plasmid DNA followed by analysis using the NCBI Blast Search program revealed the identity of insert DNA in positive clones.
Cell culture and stable transfection
HaCaT cells were routinely grown in high glucose Dulbecco’s modified Eagle’s media and supplemented with 10% FBS and 1% penicillin/streptomycin at 37 °C in 5% CO2 atmosphere. These cells were stably transfected with full-length human IEX-1 cloned into the pcDNA 3.1 Zeo (–) vector as described previously [3]. Zeocin (400 μg/ml) was used to select transformed cells.
Immunoprecipitation and immunoblotting
For immunoprecipitation, transfected HaCaT cells were washed with 1× PBS and lysed in lysis buffer (300 mM NaCl, 50 mM Tris, pH 7.4, 1% Triton X-100, 5 mM EDTA, 0.02% sodium azide, 1.0 mM phenylmethylsulfonyl fluoride, and protease inhibitor cocktail). Cell lysates were pre-cleared for 30 min at 4 °C with protein A/G beads. Immunoprecipitation was performed using protein A/G beads conjugated to either anti-CAML or anti-IEX-1 antibodies by incubation for 2 h at 4 °C. Pre-cleared lysates were mixed with antibody labeled beads and incubated overnight at 4 °C. Beads were centrifuged briefly at 4 °C and washed three times with ice-cold washing buffer (0.1% (w/v) Triton X-100, 50 mM Tris, pH 7.4, 300 mM NaCl, and 5 mM EDTA). Interacting proteins were heated at 100 °C in 30 μl of SDS-sample buffer, loaded onto 15% SDS–polyacrylamide gel, and analyzed by immunoblotting. The PVDF membrane was blocked in a 1% TBS blocking solution (Roche Molecular Biochemicals, Indianapolis, IN) for 1 h at room temperature with agitation. Membranes were blotted using anti-IEX-1 antibodies (1:1000) or anti-CAML (1:500) primary antibodies for 2 h at room temperature. Membranes were then washed and incubated with appropriate horseradish peroxidase (HRP) conjugated secondary antibodies for 1 h at room temperature. Proteins were detected using BM chemiluminescent detection solution (Roche Molecular Biochemicals).
Results and discussion
Sixty-three yeast colonies showed histidine, adenine, tryptophan, and leucine auxotrophy. Of these 63 colonies, 13 colonies grew and turned blue when grown on SD/-Trp/-Leu/-His/-Ade + X-α-Gal plates. The method used to verify interaction between proteins was stringent, in that initial colonies growing on media lacking in tryptophan, histidine, and leucine were further screened on media lacking tryptophan, histidine, leucine, and adenine. Finally, healthy colonies growing on the latter plates were further tested for their ability to hydrolyze X-α-Gal and generate blue colonies. These colonies were evaluated further by sequencing of inserts. The identity of the DNA inserts was determined by NCBI blast searches.
To further verify the authenticity of the interactions seen in the two-hybrid system we performed immunoprecipitation reactions with IEX-1 and CAML specific antibodies, which were used to immunoprecipitate the proteins from human keratinocytes (HaCaT cells). When lysates of these cells were treated with anti-IEX-1 antibody beads, CAML was immunoprecipitated with IEX-1 and was detected with an anti-CAML antibody. It is very likely that the interactions that were seen in this system are indeed authentic and of biological relevance.
Sequencing of inserts in the pACT2 plasmids showed the following inserts that encoded proteins of interest: calcium-modulating cyclophilin ligand, TNF-related apoptosis-inducing ligand (TRAIL), the BCL2 homolog MCL1, BAT3, zyxin, platelet factor 4 variant, HUEL (c4orf1), peroxisomal enoyl coA hydratase, small glutamine rich tetratricopeptide containing protein, integrin β5 precursor, and the β polypeptide of the sodium potassium ATPase. Remarkably, the first four proteins (out of a total of eleven) are involved in the regulation of apoptosis [36-63] (see Table 1 for a summary of the actions of these proteins). A fifth protein, zyxin, has also been implicated in the apoptotic response [64,65]. Since IEX-1 has been shown to modulate apoptosis, this is of great interest as it points to mechanisms by which IEX-1 may function in this regard.
Table 1.
Clone No. | Identity of DNA | Accession No. | Function of encoded protein—comments from NCBI summaries in nucleotide database |
---|---|---|---|
1 |
Homo sapiens calcium modulating ligand |
NM_001745 | The integral membrane protein encoded by this gene functions similar to cyclosporin A, binding to cyclophilin B and acting downstream of the TCR and upstream of calcineurin by causing an influx of calcium |
2 | TNF-related apoptosis-inducing ligand (TRAIL/Apo-2 ligand); Homo sapiens tumor necrosis factor (ligand) superfamily, member 10, (TNFSF10) |
NM_00381 | The protein encoded by this gene is a cytokine that belongs to the tumor necrosis factor (TNF) ligand family. TRAIL induces apoptosis in transformed and tumor cells, but does not normal cells. TRAIL binds to several members of TNF receptor superfamily |
3 |
Homo sapiens myeloid cell leukemia sequence 1 (BCL2-related) (MCL1) |
NM_182763 | The protein encoded by this gene belongs to the Bcl-2 family. Alternative splicing occurs at this locus and two transcript variants encoding distinct isoforms have been identified. The longer gene product (isoform 1) enhances cell survival by inhibiting apoptosis while the alternatively spliced,shorter gene product (isoform 2) promotes apoptosis and is death-inducing |
4 |
Homo sapiens HLA-B associated transcript 3 (BAT3) |
NM_080703 | The BAT1-BAT5 gene cluster is present in the vicinity of the genes for TNF-α and TNF-β. These genes are all within the human major histocompatibility complex class III region. The protein encoded by this gene is a nuclear protein. It has been implicated in the control of apoptosis and regulating heat shock protein. There are three alternatively spliced transcript variants described for this gene |
Calcium-modulating cyclophilin ligand is an integral membrane protein that appears to be a participant in the calcium-signaling pathway [36-41,43]. CAML functions in a manner similar to cyclosporine A, binding to cyclophylin B and acting downstream of the T cell receptor and upstream of calcineurin by causing an influx of calcium. CAML is thought to inhibit the rate of apoptosis in some cells [43]. TNF-related apoptosis-inducing ligand [TRAIL/Apo-2 ligand, tumor necrosis factor, member 10 (TNF SF10)] is a member of the tumor necrosis family that induces apoptosis [44-49]. The BCL2 homolog, MCL1, is expressed in human myeloid leukemia cell lines during phorbol ester-induced differentiation along the monocyte/macrophage pathway. The protein has sequence similarity to BCL2. Expression of the MCL1 gene is associated with inhibition of program cell death [50-53,55,56,58,60]. BAT3 is a 110 kDa protein that has an amino terminal ubiquitinlike domain, is rich in proline, and contains short tracks of polyproline, polyglycine, and charged amino acids. The gene for BAT3 is present within the major histo-compatability complex and is closely linked to TNF-α, TNF-β, and HLAb genes. Zyxin, which is a component of adhesion plaques, has been suggested to perform regulatory functions at these specialized regions of the plasma membrane. It displays a collection of proline rich sequences as well as three copies of the LIM domain, a zinc finger domain found in many signaling molecules. Recent findings suggest that zyxin may play a role in CD99 associated signaling in Ewing’s sarcoma cells. Treatment of ES cells with zyxin antisense oligonucleotides inhibited CD99-induced cell aggregation and apoptosis, suggesting a role for zyxin in CD99 function [64]. Additional data suggest that zyxin like domains may be involved in VASP and semaphorin cell signaling [65].
IEX-1 enhances apoptosis in some cells, whereas in other cell systems, IEX-1 protects against apoptosis[7,8,11-13,19,22,23]. Our findings show that IEX-1 may function in the control of apoptosis by directly binding to a variety of proteins associated with either the enhancement or inhibition of apoptosis and that differential binding and activation may provide an explanation for the varied effects of this protein in different cells. Further investigations of the mechanism of action of IEX-1 involving binding to pro- or anti-apoptotic molecules are warranted.
Acknowledgment
This work was supported by NIH Grant DK25409.
References
- [1].Charles CH, Yoon JK, Simske JS, Lau LF. Genomic structure, cDNA sequence, and expression of gly96, a growth factor-inducible immediate-early gene encoding a short-lived glycosylated protein. Oncogene. 1993;8:797–801. [PubMed] [Google Scholar]
- [2].Kondratyev AD, Chung KN, Jung MO. Identification and characterization of a radiation-inducible glycosylated human early-response gene. Cancer Res. 1996;56:1498–1502. [PubMed] [Google Scholar]
- [3].Kumar R, Kobayashi T, Warner GM, Wu Y, Salisbury JL, Lingle W, Pittelkow MR. A novel immediate early response gene, IEX-1, is induced by ultraviolet radiation in human keratinocytes. Biochem. Biophys. Res. Commun. 1998;253:336–341. doi: 10.1006/bbrc.1998.9692. [DOI] [PubMed] [Google Scholar]
- [4].Kobayashi T, Pittelkow MR, Warner GM, Squillace KA, Kumar R. Regulation of a novel immediate early response gene, IEX-1, in keratinocytes by 1α,25-dihydroxyvitamin D3. Biochem. Biophys. Res. Commun. 1998;251:868–873. doi: 10.1006/bbrc.1998.9556. [DOI] [PubMed] [Google Scholar]
- [5].Kumar R, Pittelkow MR, Salisbury JL, Grande JP, Im HJ, Feldmann KA, Schilling D. A novel vitamin D-regulated immediate-early gene, IEX-1, alters cellular growth and apoptosis. Recent Results Cancer Res. 2003;164:123–134. doi: 10.1007/978-3-642-55580-0_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Schafer H, Lettau P, Trauzold A, Banasch M, Schmidt WE. Human PACAP response gene 1 (p22/PRG1): proliferation-associated expression in pancreatic carcinoma cells. Pancreas. 1999;18:378–384. doi: 10.1097/00006676-199905000-00008. [DOI] [PubMed] [Google Scholar]
- [7].Arlt A, Grobe O, Sieke A, Kruse ML, Folsch UR, Schmidt WE, Schafer H. Expression of the NF-κB target gene IEX-1 (p22/PRG1) does not prevent cell death but instead triggers apoptosis in HeLa cells. Oncogene. 2001;20:69–76. doi: 10.1038/sj.onc.1204061. [DOI] [PubMed] [Google Scholar]
- [8].Arlt A, Kruse ML, Breitenbroich M, Gehrz A, Koc B, Minkenberg J, Folsch UR, Schafer H. The early response gene IEX-1 attenuates NF-κB activation in 293 cells, a possible counter-regulatory process leading to enhanced cell death. Oncogene. 2003;22:3343–3351. doi: 10.1038/sj.onc.1206524. [DOI] [PubMed] [Google Scholar]
- [9].De Keulenaer GW, Wang Y, Feng Y, Muangman S, Yamamoto K, Thompson JF, Turi TG, Landschutz K, Lee RT. Identification of IEX-1 as a biomechanically controlled nuclear factor-kappa B target gene that inhibits cardiomyocyte hypertrophy. Circ. Res. 2002;90:690–696. doi: 10.1161/01.res.0000012922.40318.05. [DOI] [PubMed] [Google Scholar]
- [10].Schulze PC, de Keulenaer GW, Kassik KA, Takahashi T, Chen Z, Simon DI, Lee RT. Biomechanically induced gene iex-1 inhibits vascular smooth muscle cell proliferation and neointima formation. Circ. Res. 2003;93:1210–1217. doi: 10.1161/01.RES.0000103635.38096.2F. [DOI] [PubMed] [Google Scholar]
- [11].Wu MX, Ao Z, Prasad KV, Wu R, Schlossman SF. IEX-1L, an apoptosis inhibitor involved in NF-κB-mediated cell survival. Science. 1998;281:998–1001. doi: 10.1126/science.281.5379.998. [DOI] [PubMed] [Google Scholar]
- [12].Wu MX. Roles of the stress-induced gene IEX-1 in regulation of cell death and oncogenesis. Apoptosis. 2003;8:11–18. doi: 10.1023/a:1021688600370. [DOI] [PubMed] [Google Scholar]
- [13].Zhang Y, Schlossman SF, Edwards RA, Ou CN, Gu J, Wu MX. Impaired apoptosis, extended duration of immune responses, a lupus-like autoimmune disease in IEX-1-transgenic mice. Proc. Natl. Acad. Sci. USA. 2002;99:878–883. doi: 10.1073/pnas.022326699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Pietzsch A, Buchler C, Schmitz G. Genomic organization, promoter cloning, and chromosomal localization of the Dif-2 gene. Biochem. Biophys. Res. Commun. 1998;245:651–657. doi: 10.1006/bbrc.1998.8500. [DOI] [PubMed] [Google Scholar]
- [15].Ohki R, Yamamoto K, Mano H, Lee RT, Ikeda U, Shimada K. Identification of mechanically induced genes in human monocytic cells by DNA microarrays. J. Hypertens. 2002;20:685–691. doi: 10.1097/00004872-200204000-00026. [DOI] [PubMed] [Google Scholar]
- [16].Lehoux S, Tedgui A. All strain, no gain: stretch keeps proliferation at bay via the NF-κB response gene iex-1. Circ. Res. 2003;93:1139–1141. doi: 10.1161/01.RES.0000108693.79326.A8. [DOI] [PubMed] [Google Scholar]
- [17].Grobe O, Arlt A, Ungefroren H, Krupp G, Folsch UR, Schmidt WE, Schafer H. Functional disruption of IEX-1 expression by concatemeric hammerhead ribozymes alters growth properties of 293 cells. FEBS Lett. 2001;494:196–200. doi: 10.1016/s0014-5793(01)02344-4. [DOI] [PubMed] [Google Scholar]
- [18].Garcia J, Ye Y, Arranz V, Letourneux C, Pezeron G, Porteu F. IEX-1: a new ERK substrate involved in both ERK survival activity and ERK activation. EMBO J. 2002;21:5151–5163. doi: 10.1093/emboj/cdf488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Osawa Y, Nagaki M, Banno Y, Brenner DA, Nozawa Y, Moriwaki H, Nakashima S. Expression of the NF-κ B target gene X-ray-inducible immediate early response factor-1 short enhances TNF-alpha-induced hepatocyte apoptosis by inhibiting Akt activation. J. Immunol. 2003;170:4053–4060. doi: 10.4049/jimmunol.170.8.4053. [DOI] [PubMed] [Google Scholar]
- [20].Schafer H, Trauzold A, Siegel EG, Folsch UR, Schmidt WE. PRG1: a novel early-response gene transcriptionally induced by pituitary adenylate cyclase activating polypeptide in a pancreatic carcinoma cell line. Cancer Res. 1996;56:2641–2648. [PubMed] [Google Scholar]
- [21].Schafer H, Arlt A, Trauzold A, Hunermann-Jansen A, Schmidt WE. The putative apoptosis inhibitor IEX-1L is a mutant nonspliced variant of p22(PRG1/IEX-1) and is not expressed in vivo. Biochem. Biophys. Res. Commun. 1999;262:139–145. doi: 10.1006/bbrc.1999.1131. [DOI] [PubMed] [Google Scholar]
- [22].Schilling D, Pittelkow MR, Kumar R. IEX-1, an immediate early gene, increases the rate of apoptosis in keratinocytes. Oncogene. 2001;20:7992–7997. doi: 10.1038/sj.onc.1204965. [DOI] [PubMed] [Google Scholar]
- [23].Zhang Y, Finegold MJ, Porteu F, Kanteti P, Wu MX. Development of T-cell lymphomas in Emu-IEX-1 mice. Oncogene. 2003;22:6845–6851. doi: 10.1038/sj.onc.1206707. [DOI] [PubMed] [Google Scholar]
- [24].Pietzsch A, Buchler C, Aslanidis C, Schmitz G. Identification and characterization of a novel monocyte/macrophage differentiation-dependent gene that is responsive to lipopolysaccharide, ceramide, and lysophosphatidylcholine. Biochem. Biophys. Res. Commun. 1997;235:4–9. doi: 10.1006/bbrc.1997.6715. [DOI] [PubMed] [Google Scholar]
- [25].Schafer H, Diebel J, Arlt A, Trauzold A, Schmidt WE. The promoter of human p22/PACAP response gene 1 (PRG1) contains functional binding sites for the p53 tumor suppressor and for NFκB. FEBS Lett. 1998;436:139–143. doi: 10.1016/s0014-5793(98)01109-0. [DOI] [PubMed] [Google Scholar]
- [26].Schafer H, Trauzold A, Sebens T, Deppert W, Folsch UR, Schmidt WE. p22/PACAP response gene 1 (PRG1): a putative target gene for the tumor suppressor p53. Ann. N Y Acad. Sci. 1998;865:27–36. doi: 10.1111/j.1749-6632.1998.tb11159.x. [DOI] [PubMed] [Google Scholar]
- [27].Schafer H, Trauzold A, Sebens T, Deppert W, Folsch UR, Schmidt WE. The proliferation-associated early response gene p22/PRG1 is a novel p53 target gene. Oncogene. 1998;16:2479–2487. doi: 10.1038/sj.onc.1201788. [DOI] [PubMed] [Google Scholar]
- [28].Schmidt WE, Arlt A, Trauzold A, Schafer H. p22/PRG1: a novel early response gene in pancreatic cancer cells regulated by p53 and NF κB. Ann. N Y Acad. Sci. 1999;880:147–156. doi: 10.1111/j.1749-6632.1999.tb09517.x. [DOI] [PubMed] [Google Scholar]
- [29].Huang YH, Wu JY, Zhang Y, Wu MX. Synergistic and opposing regulation of the stress-responsive gene IEX-1 by p53, c-Myc, and multiple NF-κB/rel complexes. Oncogene. 2002;21:6819–6828. doi: 10.1038/sj.onc.1205854. [DOI] [PubMed] [Google Scholar]
- [30].Im HJ, Pittelkow MR, Kumar R. Divergent regulation of the growth-promoting gene IEX-1 by the p53 tumor suppressor and Sp1. J. Biol. Chem. 2002;277:14612–14621. doi: 10.1074/jbc.M109414200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Arlt A, Minkenberg J, Kocs B, Grossmann M, Kruse ML, Folsch UR, Schafer H. The expression of immediate early gene X-1 (IEX-1) is differentially induced by retinoic acids in NB4 and KG1 cells: possible implication in the distinct phenotype of retinoic acid-responsive and -resistant leukemic cells. Leukemia. 2004 doi: 10.1038/sj.leu.2403481. [DOI] [PubMed] [Google Scholar]
- [32].Im HJ, Craig TA, Pittelkow MR, Kumar R. Characterization of a novel hexameric repeat DNA sequence in the promoter of the immediate early gene, IEX-1, that mediates 1α,25-dihydroxyvitamin D(3)-associated IEX-1 gene repression. Oncogene. 2002;21:3706–3714. doi: 10.1038/sj.onc.1205450. [DOI] [PubMed] [Google Scholar]
- [33].Feldmann KA, Pittelkow MR, Roche PC, Kumar R, Grande JP. Expression of an immediate early gene, IEX-1, in human tissues. Histochem. Cell Biol. 2001;115:489–497. doi: 10.1007/s004180100284. [DOI] [PubMed] [Google Scholar]
- [34].Ausubel FM, Brent R, Kingston RE, Moore DD, Seidman JG, Smith JA, Struhl K. Current Protocols in Molecular Biology. Wiley; Hoboken, NJ: 2003. [Google Scholar]
- [35].Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc. Natl. Acad. Sci. USA. 1977;74:5463–5467. doi: 10.1073/pnas.74.12.5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Bram RJ, Crabtree GR. Calcium signalling in T cells stimulated by a cyclophilin B-binding protein. Nature. 1994;371:355–358. doi: 10.1038/371355a0. [DOI] [PubMed] [Google Scholar]
- [37].Bram RJ, Valentine V, Shapiro DN, Jenkins NA, Gilbert DJ, Copeland NG. The gene for calcium-modulating cyclophilin ligand (CAMLG) is located on human chromosome 5q23 and a syntenic region of mouse chromosome 13. Genomics. 1996;31:257–260. doi: 10.1006/geno.1996.0044. [DOI] [PubMed] [Google Scholar]
- [38].Holloway MP, Bram RJ. A hydrophobic domain of Ca2+-modulating cyclophilin ligand modulates calcium influx signaling in T lymphocytes. J. Biol. Chem. 1996;271:8549–8552. doi: 10.1074/jbc.271.15.8549. [DOI] [PubMed] [Google Scholar]
- [39].von Bulow GU, Bram RJ. NF-AT activation induced by a CAML-interacting member of the tumor necrosis factor receptor superfamily. Science. 1997;278:138–141. doi: 10.1126/science.278.5335.138. [DOI] [PubMed] [Google Scholar]
- [40].Tovey SC, Bootman MD, Lipp P, Berridge MJ, Bram RJ. Calcium-modulating cyclophilin ligand desensitizes hormone-evoked calcium release. Biochem. Biophys. Res. Commun. 2000;276:97–100. doi: 10.1006/bbrc.2000.3442. [DOI] [PubMed] [Google Scholar]
- [41].von Bulow GU, Russell H, Copeland NG, Gilbert DJ, Jenkins NA, Bram RJ. Molecular cloning and functional characterization of murine transmembrane activator and CAML interactor (TACI) with chromosomal localization in human and mouse. Mamm. Genome. 2000;11:628–632. doi: 10.1007/s003350010125. [DOI] [PubMed] [Google Scholar]
- [42].Xia XZ, Treanor J, Senaldi G, Khare SD, Boone T, Kelley M, Theill LE, Colombero A, Solovyev I, Lee F, McCabe S, Elliott R, Miner K, Hawkins N, Guo J, Stolina M, Yu G, Wang J, Delaney J, Meng SY, Boyle WJ, Hsu H. TACI is a TRAF-interacting receptor for TALL-1, a tumor necrosis factor family member involved in B cell regulation. J. Exp. Med. 2000;192:137–143. doi: 10.1084/jem.192.1.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Feng P, Park J, Lee BS, Lee SH, Bram RJ, Jung JU. Kaposi’s sarcoma-associated herpesvirus mitochondrial K7 protein targets a cellular calcium-modulating cyclophilin ligand to modulate intracellular calcium concentration and inhibit apoptosis. J. Virol. 2002;76:11491–11504. doi: 10.1128/JVI.76.22.11491-11504.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Wiley SR, Schooley K, Smolak PJ, Din WS, Huang CP, Nicholl JK, Sutherland GR, Smith TD, Rauch C, Smith CA, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 1995;3:673–682. doi: 10.1016/1074-7613(95)90057-8. [DOI] [PubMed] [Google Scholar]
- [45].Jeremias I, Herr I, Boehler T, Debatin KM. TRAIL/Apo-2-ligand-induced apoptosis in human T cells. Eur. J. Immunol. 1998;28:143–152. doi: 10.1002/(SICI)1521-4141(199801)28:01<143::AID-IMMU143>3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
- [46].Griffith TS, Lynch DH. TRAIL: a molecule with multiple receptors and control mechanisms. Curr. Opin. Immunol. 1998;10:559–563. doi: 10.1016/s0952-7915(98)80224-0. [DOI] [PubMed] [Google Scholar]
- [47].Goke R, Goke A, Goke B, Chen Y. Regulation of TRAIL-induced apoptosis by transcription factors. Cell. Immunol. 2000;201:77–82. doi: 10.1006/cimm.2000.1650. [DOI] [PubMed] [Google Scholar]
- [48].Abe K, Kurakin A, Mohseni-Maybodi M, Kay B, Khosravi-Far R. The complexity of TNF-related apoptosis-inducing ligand. Ann. N Y Acad. Sci. 2000;926:52–63. doi: 10.1111/j.1749-6632.2000.tb05598.x. [DOI] [PubMed] [Google Scholar]
- [49].Baetu TM, Hiscott J. On the TRAIL to apoptosis. Cytokine Growth Factor Rev. 2002;13:199–207. doi: 10.1016/s1359-6101(02)00006-0. [DOI] [PubMed] [Google Scholar]
- [50].Reynolds JE, Yang T, Qian L, Jenkinson JD, Zhou P, Eastman A, Craig RW. Mcl-1, a member of the Bcl-2 family, delays apoptosis induced by c-Myc overexpression in Chinese hamster ovary cells. Cancer Res. 1994;54:6348–6352. [PubMed] [Google Scholar]
- [51].McDonnell TJ, Beham A, Sarkiss M, Andersen MM, Lo P. Importance of the Bcl-2 family in cell death regulation. Experientia. 1996;52:1008–1017. doi: 10.1007/BF01920110. [DOI] [PubMed] [Google Scholar]
- [52].Zhou P, Qian L, Kozopas KM, Craig RW. Mcl-1, a Bcl-2 family member, delays the death of hematopoietic cells under a variety of apoptosis-inducing conditions. Blood. 1997;89:630–643. [PubMed] [Google Scholar]
- [53].Moulding DA, Quayle JA, Hart CA, Edwards SW. Mcl-1 expression in human neutrophils: regulation by cytokines and correlation with cell survival. Blood. 1998;92:2495–2502. [PubMed] [Google Scholar]
- [54].Townsend KJ, Trusty JL, Traupman MA, Eastman A, Craig RW. Expression of the antiapoptotic MCL1 gene product is regulated by a mitogen activated protein kinase-mediated pathway triggered through microtubule disruption and protein kinase C. Oncogene. 1998;17:1223–1234. doi: 10.1038/sj.onc.1202035. [DOI] [PubMed] [Google Scholar]
- [55].Johnson AL. Mcl-1–just another antiapoptotic Bcl-2 Homolog? Endocrinology. 1999;140:5465–5468. doi: 10.1210/endo.140.12.7254. [DOI] [PubMed] [Google Scholar]
- [56].Leuenroth SJ, Grutkoski PS, Ayala A, Simms HH. Suppression of PMN apoptosis by hypoxia is dependent on Mcl-1 and MAPK activity. Surgery. 2000;128:171–177. doi: 10.1067/msy.2000.107609. [DOI] [PubMed] [Google Scholar]
- [57].Leuenroth SJ, Grutkoski PS, Ayala A, Simms HH. The loss of Mcl-1 expression in human polymorphonuclear leukocytes promotes apoptosis. J. Leukoc. Biol. 2000;68:158–166. [PubMed] [Google Scholar]
- [58].Moulding DA, Giles RV, Spiller DG, White MR, Tidd DM, Edwards SW. Apoptosis is rapidly triggered by antisense depletion of MCL-1 in differentiating U937 cells. Blood. 2000;96:1756–1763. [PubMed] [Google Scholar]
- [59].Zhang B, Gojo I, Fenton RG. Myeloid cell factor-1 is a critical survival factor for multiple myeloma. Blood. 2002;99:1885–1893. doi: 10.1182/blood.v99.6.1885. [DOI] [PubMed] [Google Scholar]
- [60].Jourdan M, Veyrune JL, Vos JD, Redal N, Couderc G, Klein B. A major role for Mcl-1 antiapoptotic protein in the IL-6-induced survival of human myeloma cells. Oncogene. 2003;22:2950–2959. doi: 10.1038/sj.onc.1206423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Mori M, Burgess DL, Gefrides LA, Foreman PJ, Opferman JT, Korsmeyer SJ, Cavalheiro EA, Naffah-Mazzacoratti MG, Noebels JL. Expression of apoptosis inhibitor protein Mcl1 linked to neuroprotection in CNS neurons. Cell Death Differ. 2004 doi: 10.1038/sj.cdd.4401483. [DOI] [PubMed] [Google Scholar]
- [62].Wu YH, Shih SF, Lin JY. Ricin triggers apoptotic morphological changes through caspase-3 cleavage ofBAT3. J. Biol. Chem. 2004;279:19264–19275. doi: 10.1074/jbc.M307049200. [DOI] [PubMed] [Google Scholar]
- [63].Manchen ST, Hubberstey AV. Human Scythe contains a functional nuclear localization sequence and remains in the nucleus during staurosporine-induced apoptosis. Biochem. Biophys. Res. Commun. 2001;287:1075–1082. doi: 10.1006/bbrc.2001.5701. [DOI] [PubMed] [Google Scholar]
- [64].Cerisano V, Aalto Y, Perdichizzi S, Bernard G, Manara MC, Benini S, Cenacchi G, Preda P, Lattanzi G, Nagy B, Knuutila S, Colombo MP, Bernard A, Picci P, Scotlandi K. Molecular mechanisms of CD99-induced caspase-independent cell death and cell-cell adhesion in Ewing’s sarcoma cells: actin and zyxin as key intracellular mediators. Oncogene. 2004;23:5664–5674. doi: 10.1038/sj.onc.1207741. [DOI] [PubMed] [Google Scholar]
- [65].Klostermann A, Lutz B, Gertler F, Behl C. The orthologous human and murine semaphorin 6A-1 proteins (SEMA6A-1/Sema6A-1) bind to the enabled/vasodilator-stimulated phosphoprotein-like protein (EVL) via a novel carboxyl-terminal zyxinlike domain. J. Biol. Chem. 2000;275:39647–39653. doi: 10.1074/jbc.M006316200. [DOI] [PubMed] [Google Scholar]