The caspase inhibitor and RING finger-containing protein cellular inhibitor of apoptosis protein 1 (c-IAP1) has been shown to be involved in both apoptosis inhibition and signaling by members of the tumor necrosis factor (TNF) receptor family. The protein is regulated transcriptionally (eg, is a target for nuclear factor-κB [NF-κB]) and can be inhibited by mitochondrial proteins released in the cytoplasm upon apoptotic stimuli. The present study indicates that an additional level of regulation of c-IAP1 may be cell compartmentalization. The protein is present in the nucleus of undifferentiated U937 and THP1 monocytic cell lines. When these cells undergo differentiation under phorbol ester exposure, c-IAP1 translocates to the cytoplasmic side of the Golgi apparatus. This redistribution involves a nuclear export signal (NES)-mediated, leptomycin B-sensitive mechanism. Using site-directed mutagenesis, we localized the functional NES motif in the caspase recruitment domain (CARD) of c-IAP1. A nucleocytoplasmic redistribution of the protein was also observed in human monocytes as well as in tumor cells from epithelial origin when undergoing differentiation. c-IAP1 does not translocate from the nucleus of cells whose differentiation is blocked (ie, in cell lines and monocytes from transgenic mice overexpressing B-cell lymphoma 2 [Bcl-2] and in monocytes from patients with chronic myelomonocytic leukemia). Altogether, these observations associate c-IAP1 cellular location with cell differentiation, which opens new perspectives on the functions of the protein. (Blood. 2004;104:2035-2043)

The inhibitors of apoptosis proteins (IAPs) have been initially defined as natural cellular inhibitors of cell death. These proteins were identified in baculoviral genome as regulators of host-cell viability during virus infection,1  and cellular orthologues were subsequently described in yeast, nematodes, drosophila, and mammals. The human genome encodes at least 8 IAPs (X-linked IAP [XIAP], cellular IAP1 [c-IAP1], c-IAP2, melanoma IAP [ML-IAP], neuronal apoptosis inhibitory protein [NAIP], survivin, IAP-like protein 2 [ILP-2], Apollon).2  All of these proteins have in common the presence of 1 to 3 copies of a baculovirus IAP repeat (BIR) domain.1  These domains are essential for the antiapoptotic properties of the IAPs, which have been attributed to the direct binding and inhibition of caspases. XIAP binds the small subunit of caspase-9 through its BIR3 domain3  and masks the active site of caspase-3 and -7 through a distinct segment, which is immediately amino-terminal to its BIR2 domain.4,5  c-IAP1 and c-IAP2 bind caspase-3 and -7 but their inhibitory effect on caspases is 2- to 3-log lower than that of XIAP.6  Some of the BIR-containing proteins do not have clear links with apoptosis and several members of the family have demonstrated distinct functions including cell cycle regulation,7  protein degradation,8  and caspase-independent signal transduction.9-12 

In addition to the BIR domains, several IAPs, including XIAP, c-IAP1, and c-IAP2, contain a highly conserved carboxy-terminal RING finger domain that confers them an enzyme 3 (E3) function in the protein ubiquitylation process. Several proteins specifically targeted for ubiquitylation by IAPs have been identified. At least in vitro, XIAP and c-IAP2 direct the ubiquitylation of caspase-3 and caspase-7,13,14  whereas c-IAP1 and c-IAP2 mediate ubiquitylation of second mitochondria-derived activator of caspase (Smac)/DIABLO, an antagonist of IAPs.15  c-IAP1 and c-IAP2 are also components of the type 2 tumor necrosis factor (TNF) receptor complex through interaction with the signaling intermediates TNF receptor-associated factor 1 (TRAF1) and TRAF2.9  c-IAP1 could induce the ubiquitylation of TRAF2 and participated in the TNF-α-mediated proteasomal degradation of TRAF2,16  and c-IAP2 has been involved in the TNF-α signaling leading to nuclear factor-κB (NF-κB) activation.17 

The expression and activity of IAPs are regulated at several levels. The transcription factor NF-κB enhances the expression of c-IAP1, c-IAP2, and XIAP, which may contribute to the prosurvival effect exerted in many situations by this transcription factor.18,19  XIAP translation can be enhanced through the use of an internal ribosomal entry site in the 5′-untranslated region of its messenger RNA.20  IAPs could regulate their own degradation through autoubiquitylation,8  whereas the IAP-interacting proteins Smac/DIABLO and Omi/HtrA2 neutralize XIAP and possibly other IAPs when released from the mitochondria under apoptotic stimuli.21 

Another level of regulation of IAP functions is the modulation of their subcellular location. Such a regulation has been described for XIAP whose interaction with the protein XIAP-associated factor 1 (XAF1) induces its sequestration in the nucleus and suppresses its caspase-inhibitory function.22  The present study demonstrates that c-IAP1 is located in the nucleus of various undifferentiated cells and migrates to the cytoplasm, more specifically to the Golgi apparatus, when these cells undergo differentiation. This redistribution of c-IAP1 involves a nuclear export signal (NES) located in its caspase recruitment domain (CARD). Overexpression of c-IAP1 interferes with 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced differentiation of leukemic cells, a process also inhibited by the nuclear export inhibitor leptomycin B (LMB). Altogether, these observations suggest a role for c-IAP1 in cell differentiation.

Antibodies and chemicals

We used mouse monoclonal antibodies (mAbs) directed against c-IAP1 (PharMingen, La Jolla, CA); Golgin 97 (clone CDF4; Molecular Probes, Eugene, OR); mitochondrial HSP70 (mHSP70; Affinity BioReagent, Golden, CO); HSC70 (Santa Cruz Biotechnology, Santa Cruz, CA); GM130 (Golgi Matrix protein of 130 kDa; fluorescein isothiocyanate [FITC]-conjugated antibody; Transduction Laboratories, Lexington, KY); rabbit polyclonal Abs targeting c-IAP1 (Santa Cruz Biotechnology and R&D Systems, Abington, United Kingdom); macrophage antigen-1 (Mac-1; phycoerythrin [PE]-conjugated antibody; Pharmingen, Becton Dickinson, Heidelberg, Germany); Bcl-2 (FITC-conjugated antibody; Pharmingen, Becton Dickinson); CD1a (FITC-conjugated antibody; Pharmingen, Becton Dickinson), CD71 (FITC-conjugated antibody; Pharmingen, Becton Dickinson); poly-(adenosine diphosphate-ribose) polymerase (PARP; Boehringer-Mannheim, Mannheim, Germany); XIAP (R&D Systems and Stressgen Biotech, San Diego, CA); protein disulfide isomerase (PDI; Calbiochem, La Jolla, CA); green fluorescent protein (GFP; Invitrogen, Cergy Pontoise, France); and survivin (Novus Biologicals, Littleton, CO). Macrophage colony-stimulating factor (M-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), and interleukin-4 (IL-4) were obtained from R&D Systems; erythropoietin (EPO) was from Amgen (Thousand Oaks, CA); TPA was from Sigma-Aldrich Laboratories (St Quentin Fallavier, France); brefeldin A (BFA) and nocodazole were from Alexis Biochemicals (Lausen, Switzerland); and trypsin-EDTA (ethylenediaminetetraacetic acid) was from Gibco-BRL (Carlsbad, CA). LMB was kindly provided by Dr M. Yoshida (Tokyo, Japan) and thrombopoietin (TPO) was kindly provided by Kirin Brewery (Tokyo, Japan).

Cell culture and differentiation

Cell lines were obtained from the American Type Culture Collection (ATCC, Rockville, MD) and cultured as described.23  We also tested the previously described Bcl-2-transfected U937 and HT29 cells and HT29-MTX cells.23-25  The TPA-resistant variant of U937 cells were kindly provided by Prof P. J. Parker (London, United Kingdom).26  Monocytes from human peripheral blood were obtained with informed consent from healthy donors and 7 patients with chronic myelomonocytic leukemia (CMML) and purified using an isolation kit (Miltenyi Biotec, Paris, France) following the manufacturer's instructions. Cells were differentiated into macrophages or dendritic cells and checked for the expression of differentiation marker CD71 and CD1a as described.23  Peripheral blood CD34+ cells were cultured in liquid conditions in the presence of cytokines to generate megakaryocytes or erythroid cells as described.27,28  The Bcl-2 transgenic mice were obtained from Irv Weismann (Stanford, CA).29  Bcl-2 overexpression in Mac-1+ cells of transgenic mice was verified by flow cytometry using a FACSCalibur cytometer and Cell Quest software (Pharmingen, Becton Dickinson). Femoral bone marrow cells were isolated from 6- to 8-week-old control and transgenic FVB/N female mice and cultured for 4 hours on plastic plates before culturing adherent cells for 6 days in the presence of 10% L929 cell-conditioned medium as source of CSF-1. Macrophage differentiation was assessed by May-Grünwald-Giemsa staining.

Immunofluorescence studies

Cells were fixed in paraformaldehyde (PFA; 2%) for 10 minutes at room temperature, washed twice, saturated in phosphate-buffered saline (PBS) containing 0.1% saponin and 5% nonfat milk, and incubated overnight at room temperature in the presence of primary Ab diluted in PBS containing 0.1% saponin and 0.5% bovine serum albumin (BSA). After washing, cells were incubated for 30 minutes with 488-alexa goat antirabbit or antimouse Ab (Molecular Probes) and washed 3 times with PBS. Nuclei were stained by Hoechst 33342 (Sigma-Aldrich). To demonstrate colocalization of c-IAP1 with Golgin 97 or GM130, cells were first incubated with anti-c-IAP1 Ab overnight at 4°C, then with the secondary biotinylated-immunoglobulin (Ig; 1:100; Amersham Biosciences, Orsay, France) for 1 hour at room temperature, then with a streptavidin-texas red-conjugated Ab (Molecular Probes; 1:2000) for 1 hour. Cells were subsequently incubated for 1 hour at room temperature with anti-GM130-FITC (1:100) or anti-Golgin 97 (1:100), then with FITC-conjugated antimouse Ab. Fluorescence was preserved using the FluorSave mounting medium (Calbiochem). Analysis was performed using either a fluorescence (Nikon Eclipse 80i; Nikon, Champigny, France) or a confocal (Leica TCS SP2; Leica, Bron, France) microscope (objective × 50; original magnification × 500). The images were captured by a 3 CCD (charge-coupled device) color video camera (Sony, Paris, France), digitally saved using Archimed-Pro software (Microvision Instruments, Evry, France), and further processed using Photoshop software (Adobe Systems France, Paris, France).

Preparation of cellular extracts and Western blot analysis

Whole-cell lysates and nuclear-free extracts were prepared as described.23  Nuclear and cytoplasmic fractions were obtained by lysing the cells in lysis buffer (10 mM Hepes [N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid], 10 mM KCl, 0.1 mM EDTA, 0.1 mM EGTA [ethyleneglycoltetraacetic acid], 1 mM DTT [dithiothreitol], 0.6% NP-40 [nonidet P-40]) in the presence of the protease inhibitors. Cell lysate was centrifuged at 1200g for 10 minutes. The supernatant was carefully collected (cytoplasmic fraction [C]) and the pellet was washed once then resuspended in lysis buffer (nuclear fraction [N]). Further cell fractionation was performed as described.30  All fractions were stored at -80°C until Western blotting analysis, and protein concentration was measured using the Bio-Rad DC protein assay kit (Hercules, CA). Western blot experiments were performed as previously described.23 

Trypsin digestion of microsomal proteins

Proteins from reticular/microsomal-enriched fraction were digested by 0.05% trypsin in the presence of 0.02% EDTA for 30 minutes at 37°C and analyzed by Western blotting for c-IAP1 content.31 

Plasmid constructs

Plasmid-enhanced GFP (pEGFP)-c-IAP1 was constructed by subcloning full-length c-IAP1 cDNA (kindly provided by J. C. Reed, La Jolla, CA) into the BglII/SalI site of pEGFP-C1 (Clontech, Palo Alto, CA). Sense and antisense oligonucleotides corresponding to leucine-rich motif (LRM) putative NES were as follows: LRM1 sense, 5′-GAT CTT TTT TGG AAA ATT CTC TAG AAA CTC TGA GGA-3′; LRM1 antisense, 5′-GAT CTC CTC AGA GTT TCT AGA GAA TTT TCC AAA AAA-3′; LRM2 sense, 5′-GAT CTC TCT TTC AAC AAT TGA CAT GTG TGC TTC CTA TCC TGG ATA ATC TTT TAA-3′; LRM2 antisense, 5′-GAT CTT AAA AGA TTA TCC AGG ATA GGA AGC ACA CAT GTC AAT TGT TGA AAG AGA-3′; LRM3 sense, 5′-GAT CTC TGT CAC TGG AAG AAC AAT TGA GGA GGT TGC AAA-3′; and LRM3 antisense, 5′-GAT CTT TGC AAC CTC CTC AAT TGT TCT TCC AGT GAC AGA-3′ (Proligo France SAS, Paris, France). Complementary oligonucleotides were annealed and cloned in a sense orientation into the BglII site of pEGFP-C1 (Clontech). All sequences are expressed at the C-terminus of GFP. Full-length c-IAP1 mutants (GFP-c-IAP1-LRM1*,-LRM2*, and -LRM3*) were obtained by mutagenesis of LRM1, 2, and 3, separately or in combination (leucine were replaced by alanine) using the Quick-Change Site-directed Mutagenesis Kit (Stratagene, La Jolla, CA). All constructs were sequenced to ensure the accuracy of the reading frames and the site-directed mutations.

Cell transfection

HeLa cells were transfected 24 hours after seeding using Superfect transfection reagent (Qiagen, Valencia, CA) following the manufacturer's instructions. Cells were studied 24 hours after transient transfection: nuclei were stained with Hoechst 33342 and cells were fixed with 2% PFA for 5 minutes before studying the subcellular distribution of GFP fusion protein using a fluorescence (Nikon) or a confocal (Leica) microscope. THP1 cells were transiently transfected using the AMAXA nucleofector kit (Amaxa, Köln, Germany) and transfected cells were enriched by a 10-day geneticin selection (0.7 μg/mL) before expansion and treatment.

TPA-induced differentiation of human monocytic cell lines is associated with the redistribution of c-IAP1 and XIAP from the nucleus into the cytoplasm

It has been previously shown that exposure of U937 cells to 20 nM TPA induced their differentiation into macrophage-like cells. Cells become adherent to the culture flask and the expression of CD11b at their plasma membrane increases.23  We used Western blotting to analyze the expression of XIAP, c-IAP1, c-IAP2, and survivin, 4 proteins that belong to the IAP family, in U937 cells undergoing TPA-induced differentiation (Figure 1A). c-IAP2 could not be detected in undifferentiated U937 cells and remained undetectable at all steps of the differentiation process (not shown). Survivin expression was limited to the nucleus of undifferentiated cells and disappeared upon differentiation. This may be related to the differentiation-associated cell cycle exit since this protein, which has an evolutionarily conserved role as a mitotic spindle checkpoint protein, is expressed mainly in dividing cells.7  The expression of XIAP and c-IAP1 was poorly influenced by the differentiation process when studied in whole-cell lysates (Figure 1A left). However, c-IAP1, and to a lesser extent XIAP, progressively accumulated in nuclear-free extracts as the cells underwent differentiation (Figure 1A right). The present study focused on c-IAP1 redistribution.

Figure 1.

c-IAP1 redistribution in human leukemia cell lines undergoing TPA-induced differentiation. U937 (A-B) and THP1 (C-D) cells were treated for indicated times with 20 nM TPA to induce a macrophage-like differentiation. (A,C) Western blot analysis of indicated proteins in whole-cell, cytoplasmic, and nuclear extracts. HSC70 was used as a loading control. (B,D) Fluorescence microscopy analysis of c-IAP1 (green), as observed using an anti-c-IAP1 mAb (Pharmingen). Nuclei, labeled with Hoechst 33342, are stained in blue. Magnification × 300.

Figure 1.

c-IAP1 redistribution in human leukemia cell lines undergoing TPA-induced differentiation. U937 (A-B) and THP1 (C-D) cells were treated for indicated times with 20 nM TPA to induce a macrophage-like differentiation. (A,C) Western blot analysis of indicated proteins in whole-cell, cytoplasmic, and nuclear extracts. HSC70 was used as a loading control. (B,D) Fluorescence microscopy analysis of c-IAP1 (green), as observed using an anti-c-IAP1 mAb (Pharmingen). Nuclei, labeled with Hoechst 33342, are stained in blue. Magnification × 300.

Close modal

Differentiation-associated redistribution of c-IAP1 from the nucleus to the cytoplasm was further confirmed by Western blotting analysis of c-IAP1 expression in TPA-treated THP1 cells (Figure 1C) and by fluorescent microscopy analysis of the 2 cell lines (Figure 1B,D). c-IAP1 was located mainly in the nucleus of U937 and THP1 undifferentiated cells and in the cytoplasm of TPA-differentiated cells. A kinetic analysis identified a transient diffuse staining of the cytoplasm in the first hours of TPA treatment. As the cells progressed toward the differentiation process, a more patchy staining close to the nucleus was observed (see THP1 cells in Figure 1D).

c-IAP1 colocalizes with the Golgi apparatus of differentiated cells

To precisely determine the subcellular localization of c-IAP1 in TPA-differentiated cells, we performed Western blot experiments in enriched cellular fractions. Figure 2A shows that c-IAP1 is localized in the nucleus of undifferentiated U937 cells and in the reticular fraction of TPA-differentiated U937 cells. Thus, in accordance with Figure 1A, the protein migrates from the nucleus to the cytoplasm. A similar observation was made by comparing cellular fractions of undifferentiated and differentiated THP1 cells (not shown). Fluorescence microscopy experiments indicated that c-IAP1 colocalized with Golgin 97, a Golgi matrix protein, in TPA-differentiated THP1 (Figure 2B) and U937 (not shown) cells. c-IAP1 also colocalized, although less precisely, with GM130, a protein associated with the cis-Golgi (Figure 2B). Addition of either BFA, a fungal metabolite that causes disintegration of Golgi structure through inhibition of adenosine diphosphate (ADP)-ribosylation factor (ARF) guanosine 5′-triphosphate (GTP)-binding proteins,32  or nocodazole, a microtubule-depolarizing agent, suppressed the patchy staining of c-IAP1 in TPA-differentiated THP1 (Figure 2C) and U937 (not shown) cells. In the tested conditions, BFA did not modify calnexin C subcellular localization, indicating that the endoplasmic reticulum was not altered (not shown). Altogether, these observations indicated that c-IAP1 was redistributed to the Golgi apparatus in cells undergoing differentiation.

Figure 2.

c-IAP1 is localized to the Golgi apparatus in differentiated cells. (A) Western blot analysis of c-IAP1 (pAb; Santa Cruz Biotechnology) expression in the mitochondrial (M), cytosolic (C), reticular/microsomal (R), and nuclear (N) fractions obtained from U937 cells before (Co) and after exposure to 20 nM TPA for 72 hours. The expression of poly(ADP-ribose)polymerase (PARP), Golgin 97, and mitochondrial HSP70 was used to assess the enrichment of each cell fraction. (B) THP1 cells were treated with TPA for 48 hours before analyzing the expression of c-IAP1 (red), Golgin 97 (green), or GM130 (green) by confocal microscopy (magnification × 300). (Insets) Increased magnification of Golgi labeling (magnification × 3000). (C) c-IAP1 expression in TPA-differentiated THP1 cells before (Co) and after exposure to either brefeldin A (BFA; 5 μg/mL; 2 h 30 min) or nocodazole (10 μM; 1 h). c-IAP1 expression was observed by fluorescence microscopy (magnification ×700) using an anti-c-IAP1 mAb (Pharmingen). (D) Western blot analysis of c-IAP1 expression under limited proteolytic digestion of the reticular/microsomal fraction of TPA-differentiated U937 cells. Golgin 97 and protein disulfide isomerase (PDI) are used as positive and negative controls, respectively.

Figure 2.

c-IAP1 is localized to the Golgi apparatus in differentiated cells. (A) Western blot analysis of c-IAP1 (pAb; Santa Cruz Biotechnology) expression in the mitochondrial (M), cytosolic (C), reticular/microsomal (R), and nuclear (N) fractions obtained from U937 cells before (Co) and after exposure to 20 nM TPA for 72 hours. The expression of poly(ADP-ribose)polymerase (PARP), Golgin 97, and mitochondrial HSP70 was used to assess the enrichment of each cell fraction. (B) THP1 cells were treated with TPA for 48 hours before analyzing the expression of c-IAP1 (red), Golgin 97 (green), or GM130 (green) by confocal microscopy (magnification × 300). (Insets) Increased magnification of Golgi labeling (magnification × 3000). (C) c-IAP1 expression in TPA-differentiated THP1 cells before (Co) and after exposure to either brefeldin A (BFA; 5 μg/mL; 2 h 30 min) or nocodazole (10 μM; 1 h). c-IAP1 expression was observed by fluorescence microscopy (magnification ×700) using an anti-c-IAP1 mAb (Pharmingen). (D) Western blot analysis of c-IAP1 expression under limited proteolytic digestion of the reticular/microsomal fraction of TPA-differentiated U937 cells. Golgin 97 and protein disulfide isomerase (PDI) are used as positive and negative controls, respectively.

Close modal

c-IAP1 is located to the cytoplasmic side of the Golgi apparatus in differentiated cells

To determine the topologic orientation of c-IAP1 in the Golgi compartment of differentiated cells, we isolated the microsomal fraction from TPA-treated U937 cells and submitted this fraction to tryptic-limited digestion before Western blotting analysis (Figure 2D). Addition of trypsin resulted in complete digestion of Golgin 97, which is located on the cytoplasmic side of the Golgi apparatus, whereas the endoplasmic reticulum-lumenal PDI was resistant to trypsin digestion. In these conditions, trypsin completely digested c-IAP1, indicating that the protein may be located to the external cytoplasmic side of the Golgi apparatus.

The differentiation-induced nuclear export of c-IAP1 involves a nuclear export signal

To characterize the mechanisms that are responsible for the nuclear export of c-IAP1, we first used LMB, a specific inhibitor of exportin 1 (also known as CRM1 [chromosomal region maintenance-1]), which is the receptor for leucine-rich NES.33  Addition of 100 nM LMB for 24 hours to TPA-treated THP1 (Figure 3A) and U937 (not shown) cells prevented the redistribution of c-IAP1. To confirm the ability of LMB to inhibit the nuclear export of c-IAP1, we used a construct encoding full-length c-IAP1 associated, through its N-terminus, to GFP. This construct was transiently expressed in HeLa and 293T cell lines, in which the transfection rate was much higher than in leukemic cell lines. Twenty-four hours after transfection of GFP-c-IAP1 construct in these cells, the fluorescence was detected in both the nucleus and the cytoplasm. In the presence of LMB, the protein accumulated in the nucleus (see HeLa cells on Figure 3B). These results suggested that the nuclear export of c-IAP1 involved an NES and CRM1.

Figure 3.

c-IAP1 redistribution involves a leptomycin B-sensitive mechanism. (A) Fluorescence microscopy analysis of c-IAP1 expression (Pharmingen mAb; green) in THP1 cells treated with 20 nM TPA for 24 hours in the presence or absence of 100 nM leptomycin B (LMB). Hoechst 33342 was used to stain the nuclei (blue). (B) HeLa cells were transiently transfected with a GFP-c-IAP1 construct before staining the nuclei with Hoechst 33342 and fluorescence microscopy analysis. When indicated, LMB (200 nM) was added 3 hours before analysis. Magnification is × 500 for panels A and B.

Figure 3.

c-IAP1 redistribution involves a leptomycin B-sensitive mechanism. (A) Fluorescence microscopy analysis of c-IAP1 expression (Pharmingen mAb; green) in THP1 cells treated with 20 nM TPA for 24 hours in the presence or absence of 100 nM leptomycin B (LMB). Hoechst 33342 was used to stain the nuclei (blue). (B) HeLa cells were transiently transfected with a GFP-c-IAP1 construct before staining the nuclei with Hoechst 33342 and fluorescence microscopy analysis. When indicated, LMB (200 nM) was added 3 hours before analysis. Magnification is × 500 for panels A and B.

Close modal

A leucine-rich motif in the CARD behaves as a nuclear export signal

A software-based search in the protein sequence of c-IAP1 identified 3 hydrophobic LRMs that were consensus sequences for potential NESs. The first one was located in the BIR2 domain (LRM1), the second one in the CARD (LRM2), and the last one between the CARD and the RING domain (LRM3; Figure 4A). To determine whether one or several of these motifs played a role in c-IAP1 nuclear export, we cloned the sequences encoding these motifs in a GFP-encoding vector and expressed them by transient transfection in HeLa and 293T cells. The subcellular location of GFP fusion proteins was examined 24 hours after transfection by conventional (Figure 4B) and confocal laser (not shown) microscopy. While GFP-associated LRM1 and LRM3 were expressed in both the nucleus and the cytoplasm, GFP-associated LRM2 was almost exclusively expressed in the cytoplasm (Figure 4B-C). In addition, exposure to leptomycin B induced accumulation of the GFP-LRM2 protein in the nucleus (Figure 4C). These results indicated that LRM2 was the only sequence to behave as a functional NES.

Figure 4.

Identification of a potential nuclear export sequence in c-IAP1. (A) (Top) Schematic representation of amino acid motifs in c-IAP1 protein (619 amino acids). Leucine-rich motifs (LRMs) that could behave as nuclear export signal (NES) are indicated (BIR indicates baculovirus IAP repeat; CARD, caspase recruitment domain). (Bottom) Amino acid sequence of regions containing a potential LRM (underlined). (B-C) The cDNA sequences encoding the 3 LRMs were fused to GFP in the pEGFP-C1 vector. These constructs were transiently transfected into HeLa cells and microscopy analyses were performed 24 hours later. Nuclei were stained with Hoechst 33342 (magnification × 500). When indicated, LMB (200 nM) was added 3 hours before analysis.

Figure 4.

Identification of a potential nuclear export sequence in c-IAP1. (A) (Top) Schematic representation of amino acid motifs in c-IAP1 protein (619 amino acids). Leucine-rich motifs (LRMs) that could behave as nuclear export signal (NES) are indicated (BIR indicates baculovirus IAP repeat; CARD, caspase recruitment domain). (Bottom) Amino acid sequence of regions containing a potential LRM (underlined). (B-C) The cDNA sequences encoding the 3 LRMs were fused to GFP in the pEGFP-C1 vector. These constructs were transiently transfected into HeLa cells and microscopy analyses were performed 24 hours later. Nuclei were stained with Hoechst 33342 (magnification × 500). When indicated, LMB (200 nM) was added 3 hours before analysis.

Close modal

To determine whether this potential NES was functional in the whole protein, a series of mutants were prepared in which leucine amino acids in the LRMs were replaced by alanine residues. The mutated constructs fused to GFP in a plasmid vector were transiently transfected in HeLa (Figure 5) and THP1 (not shown) cells, and their subcellular location was analyzed by fluorescence microscopy and Western blotting 24 hours later. As shown previously (Figure 3B), overexpressed wild-type c-IAP1 in HeLa cells demonstrated a cytoplasmic and nuclear pattern of expression. Mutations in either LRM1 or LRM3 or both did not affect the cellular distribution of the protein, whereas all the LRM2 mutants accumulated in the nucleus (Figure 5A). For unknown reasons, LRM2 mutant was less expressed than wild-type protein and other mutants. These observations were confirmed by immunoblotting the nuclear and cytoplasmic fractions of transiently transfected HeLa (Figure 5B) and THP1 (not shown) cells with an anti-GFP Ab. These experiments indicated that overexpressed wild-type c-IAP1 was detected mainly in the cytoplasmic fraction. When leucine residues in LRM2 were mutated, GFP-associated protein was located in the nucleus. When leucine residues in LRM1 or LRM3 were mutated, GFP-associated c-IAP1 demonstrated a cytoplasmic and nuclear expression similar to that of the wild-type protein. Altogether, these results indicated that LRM2 was a functional NES in c-IAP1.

Figure 5.

LRM2 is the functional nuclear export signal in c-IAP1. (A) Fluorescence microscopy analysis of HeLa cells transfected for 24 hours with constructs encoding wild-type or mutated GFP-c-IAP1 (nuclei were stained with Hoechst 33342). Leucine residues in LRMs (LRM1*: Leu250, Leu254, Leu257; LRM2*: Leu468, Leu472, Leu476, Leu483; LRM3*: Leu556, Leu558, Leu562, Leu565) were replaced by alanine residues using site-directed mutagenesis. *Mutated constructs (magnification ×1000). (B) Western blot analysis of GFP expression in nuclear (N) and cytoplasmic (C) extracts from HeLa cells transfected 24 hours before with indicated constructs.

Figure 5.

LRM2 is the functional nuclear export signal in c-IAP1. (A) Fluorescence microscopy analysis of HeLa cells transfected for 24 hours with constructs encoding wild-type or mutated GFP-c-IAP1 (nuclei were stained with Hoechst 33342). Leucine residues in LRMs (LRM1*: Leu250, Leu254, Leu257; LRM2*: Leu468, Leu472, Leu476, Leu483; LRM3*: Leu556, Leu558, Leu562, Leu565) were replaced by alanine residues using site-directed mutagenesis. *Mutated constructs (magnification ×1000). (B) Western blot analysis of GFP expression in nuclear (N) and cytoplasmic (C) extracts from HeLa cells transfected 24 hours before with indicated constructs.

Close modal

Overexpressed c-IAP1 interferes with the differentiation process

LMB was observed to prevent TPA-induced differentiation of THP1, as demonstrated by studying the CD11b marker (Figure 6A), which suggested that a nucleocytoplasmic redistribution of proteins was required for the differentiation process. In an attempt to determine whether c-IAP1 was one of the proteins whose nuclear export was a key event in this process, we transiently overexpressed the GFP-tagged LRM2 mutant of c-IAP1 in THP1 cells. The protein was located mainly in the nucleus of transfected cells (Figure 6B) and TPA exposure failed to increase CD11b expression in GFP-tagged cells (Figure 6C-D). In addition, adhesion of GFP-positive cells to the culture flasks was delayed (not shown). However, similar results were obtained when wild-type c-IAP1 was transiently overexpressed in THP1 cells (Figure 6B-C). These results indicated that c-IAP1 overexpression could interfere with cell differentiation.

Figure 6.

Overexpressed c-IAP1 interferes with TPA-induced THP1 cell differentiation. (A) Flow cytometry analysis of CD11b membrane expression in THP1 cells treated with 20 nM TPA for indicated times (h) in the presence or absence of 100 nM leptomycin B (LMB). Gray histograms indicate treated cells; and white histograms, untreated cells. (B) Western blot analysis of GFP expression in nuclear (N) and cytoplasmic (C) extracts from THP1 cells transfected with wild-type (wt-c-IAP1) and LRM2-mutated (c-IAP1-LRM2*) GFP-c-IAP1. (C) Flow cytometry analysis of CD11b expression in GFP-positive THP1 cells transfected with pEGFP empty vector (vector) or wild-type (wt-c-IAP1) or LRM2-mutated (c-IAP1-LRM2*) GFP-c-IAP1 constructs. Cells were treated with 20 nM TPA for indicated times (h). Gray histograms indicate treated cells; and black lines, control untreated cells. (D) Fluorescence microscopy analysis of THP1 cells transfected with pEGFP empty vector (vector) or LRM2-mutated GFP-c-IAP1 construct (c-IAP1-LRM2*). Cells were incubated with 20 nM TPA for 48 hours and labeled with an anti-CD11b Ab (red). Nuclei were stained with Hoechst 33342 (blue). Magnification × 600.

Figure 6.

Overexpressed c-IAP1 interferes with TPA-induced THP1 cell differentiation. (A) Flow cytometry analysis of CD11b membrane expression in THP1 cells treated with 20 nM TPA for indicated times (h) in the presence or absence of 100 nM leptomycin B (LMB). Gray histograms indicate treated cells; and white histograms, untreated cells. (B) Western blot analysis of GFP expression in nuclear (N) and cytoplasmic (C) extracts from THP1 cells transfected with wild-type (wt-c-IAP1) and LRM2-mutated (c-IAP1-LRM2*) GFP-c-IAP1. (C) Flow cytometry analysis of CD11b expression in GFP-positive THP1 cells transfected with pEGFP empty vector (vector) or wild-type (wt-c-IAP1) or LRM2-mutated (c-IAP1-LRM2*) GFP-c-IAP1 constructs. Cells were treated with 20 nM TPA for indicated times (h). Gray histograms indicate treated cells; and black lines, control untreated cells. (D) Fluorescence microscopy analysis of THP1 cells transfected with pEGFP empty vector (vector) or LRM2-mutated GFP-c-IAP1 construct (c-IAP1-LRM2*). Cells were incubated with 20 nM TPA for 48 hours and labeled with an anti-CD11b Ab (red). Nuclei were stained with Hoechst 33342 (blue). Magnification × 600.

Close modal

The nucleocytoplasmic redistribution of c-IAP1 is observed in several differentiation pathways

c-IAP1 was observed to be present mainly in the nucleus of the CD34+ progenitor, in both the nucleus and the cytoplasm of peripheral blood monocytes, and exclusively in the cytoplasm of macrophages and dendritic cells obtained by ex vivo differentiation of monocytes (Figure 7A) as well as erythroblasts and megakaryocytes obtained by ex vivo differentiation of CD34+ cells (Figure 7B).27,28  As previously observed in TPA-differentiated cells (Figure 1B), c-IAP1 demonstrated a punctuated expression in the peri-nuclear zone and colocalized with Golgin 97 in macrophages (Figure 7C) and dendritic cells (not shown) obtained by differentiation of normal peripheral blood monocytes. A differentiation-associated redistribution of c-IAP1 from the nucleus to the cytoplasm was also observed in nonhematopoietic cells (ie, in HT29 human colon carcinoma cells undergoing partial differentiation when grown at confluence).34  c-IAP1 also demonstrated a cytoplasmic expression in the well-differentiated, mucus-secreting HT29/MTX clone (Figure 7D).25 

Figure 7.

c-IAP1 redistribution is a differentiation-associated event in various cell types. (A) Fluorescence microscopy analysis of c-IAP1 (mAb; Pharmingen) in peripheral blood CD34+ cells and monocytes (Mo) obtained from healthy donors and in macrophages (MΦ) and dendritic cells (DC) obtained from monocytes cultured for 6 days in the presence of M-CSF or GM-CSF/IL-4, respectively. The top panels show c-IAP1 alone (green); the bottom panels, c-IAP1 (green) + Hoechst 33352-labeled nuclei (blue). (B) Fluorescence microscopy analysis of c-IAP1 (mAb; Pharmingen; green) in CD34+ cell-derived erythoblasts (Ery) and megakaryocytes (Meg). Nuclei were labeled simultaneously with Hoechst 33352. (C) Colocalization of Golgin 97 (green) and c-IAP1 (red) in macrophages derived from peripheral blood monocytes as described for panel A. (D) Fluorescence microscopy analysis of c-IAP1 (pAb; Santa Cruz Biotechnology) in HT29 cells studied before (control) and after (confluent) reaching confluence in culture and in a methotrexate-resistant, well-differentiated derivative cell clone (HT29-MTX). (E) Fluorescence microscopy analysis of c-IAP1 (pAb; Santa Cruz Biotechnology) in control (Co), Bcl-2-overexpressing (Bcl-2), and TPA-resistant U937 cells exposed for 72 hours to 20 nM TPA. (F) May-Grünwald-Giemsa staining (MGG) and fluorescence microscopy analysis of c-IAP1 (pAb; Santa Cruz Biotechnology; Bcl-2) in bone marrow monocytes from control (Co) and Bcl-2 transgenic (Bcl-2) mice, cultured for 3 days in the presence of CSF-1-containing medium. (G) Peripheral blood mononuclear cells obtained from patients with chronic myelomonocytic leukemia (CMML) were cultured for 6 days in the presence of M-CSF or GM-CSF/IL-4. The top panels show fluorescence microscopy analysis of c-IAP1; the bottom panels, flow cytometry analysis of CD71 and CD1a membrane expression. White histograms indicate CMML patient; and gray histograms, healthy donor. One representative of 7 studied patients is shown. Magnification: × 700 (A-C, E, G) and × 500 (D, F).

Figure 7.

c-IAP1 redistribution is a differentiation-associated event in various cell types. (A) Fluorescence microscopy analysis of c-IAP1 (mAb; Pharmingen) in peripheral blood CD34+ cells and monocytes (Mo) obtained from healthy donors and in macrophages (MΦ) and dendritic cells (DC) obtained from monocytes cultured for 6 days in the presence of M-CSF or GM-CSF/IL-4, respectively. The top panels show c-IAP1 alone (green); the bottom panels, c-IAP1 (green) + Hoechst 33352-labeled nuclei (blue). (B) Fluorescence microscopy analysis of c-IAP1 (mAb; Pharmingen; green) in CD34+ cell-derived erythoblasts (Ery) and megakaryocytes (Meg). Nuclei were labeled simultaneously with Hoechst 33352. (C) Colocalization of Golgin 97 (green) and c-IAP1 (red) in macrophages derived from peripheral blood monocytes as described for panel A. (D) Fluorescence microscopy analysis of c-IAP1 (pAb; Santa Cruz Biotechnology) in HT29 cells studied before (control) and after (confluent) reaching confluence in culture and in a methotrexate-resistant, well-differentiated derivative cell clone (HT29-MTX). (E) Fluorescence microscopy analysis of c-IAP1 (pAb; Santa Cruz Biotechnology) in control (Co), Bcl-2-overexpressing (Bcl-2), and TPA-resistant U937 cells exposed for 72 hours to 20 nM TPA. (F) May-Grünwald-Giemsa staining (MGG) and fluorescence microscopy analysis of c-IAP1 (pAb; Santa Cruz Biotechnology; Bcl-2) in bone marrow monocytes from control (Co) and Bcl-2 transgenic (Bcl-2) mice, cultured for 3 days in the presence of CSF-1-containing medium. (G) Peripheral blood mononuclear cells obtained from patients with chronic myelomonocytic leukemia (CMML) were cultured for 6 days in the presence of M-CSF or GM-CSF/IL-4. The top panels show fluorescence microscopy analysis of c-IAP1; the bottom panels, flow cytometry analysis of CD71 and CD1a membrane expression. White histograms indicate CMML patient; and gray histograms, healthy donor. One representative of 7 studied patients is shown. Magnification: × 700 (A-C, E, G) and × 500 (D, F).

Close modal

c-IAP1 does not translocate from the nucleus when cell differentiation is inhibited

The redistribution of c-IAP1 observed in parental U937 cells when undergoing TPA-induced differentiation was not identified in a TPA-resistant U937 cell clone treated under the same conditions (Figure 7E). We have previously shown that Bcl-2 overexpression prevented TPA-induced differentiation in U937 human leukemia cells.23  The nucleocytoplasmic redistribution of c-IAP1 observed in TPA-treated parental cells transfected with an empty vector was not identified in Bcl-2-overexpressing U937 cells treated under similar conditions (Figure 7E). To confirm this latter observation, we tested the differentiation of bone marrow monocytes obtained from control and transgenic FVB/N mice overexpressing Bcl-2 in Mac-1+ cells. Whereas c-IAP1 translocation to the cytoplasm was observed in control cells induced to differentiate into macrophages by ex vivo culture in the presence of CSF-1-containing medium, no redistribution of the protein could be detected in cells from transgenic FVB/N mice cultured under similar conditions (Figure 7F). We also cultured peripheral blood mononuclear cells from 7 patients with chronic myelomonocytic leukemia (CMML) in the presence of M-CSF or the GM-CSF/IL-4 combination for 6 days. Cell differentiation was assessed morphologically and confirmed by studying cell phenotype using CD71 and CD1a to identify macrophages and dendritic cells, respectively. Mononuclear cells from these patients failed to differentiate, and this blockade in cell differentiation was associated with a lack of c-IAP1 nucleus export (see Figure 7G for example).

Two main functions have been assigned to c-IAP1. The protein is involved in the signaling induced by engagement of several members of the TNF receptor family including TNF-R2,9  CD40,35  and the lymphotoxin-β receptor.36  c-IAP1 was also described as an endogenous inhibitor of apoptosis through direct binding to the active sites of caspase-3 and -7.37  The recently described role of the protein in ubiquitylation may contribute to both functions (eg, by regulating the cellular level of the adaptor molecule TRAF216  or the apoptosis inducer Smac/DIABLO).15  These functions imply a cytoplasmic localization of the protein. We show here that c-IAP1 is present almost exclusively in the nucleus of the studied undifferentiated cells, translocates to the cytoplasm when these cells undergo differentiation, and localizes mainly to the Golgi apparatus in differentiated cells.

Several arguments suggest a translocation of the protein rather than a degradation followed by a synthesis in a distinct cellular compartment. First, c-IAP1 mRNA (not shown) and protein levels remain stable along the differentiation. Secondly, by immunofluorescence analysis, the protein was identified in the nucleus of undifferentiated cells, in the cytosol of cells undergoing differentiation, and in the Golgi of differentiated cells, suggesting a migration. Third, we have identified a functional NES in the protein. The nuclear transport of proteins through nuclear pores requires the presence of specific signals such as nuclear localization signals (NLSs) and NESs. The size of c-IAP1 prevents it from passively diffusing through nuclear pores38  and we did not identify any classical NLSs in the protein, which suggests that the protein is either carried by a cofactor or enters the nucleus using a nonconventional mechanism of active import. On the other hand, the protein export may involve an NES-mediated, LMB-sensitive mechanism. The retention of c-IAP1 in the nucleus of undifferentiated cells suggests inhibition of this NES-mediated export. Activation of such an active export mechanism has been shown in other proteins to require an event that promotes their interaction with CRM1 such as phosphorylation,39  monoubiquitylation,40  conformational changes,41  or proteolytic cleavage.42  So far, we did not detect any posttranslational modification of the protein in cells undergoing differentiation. The location of the functional NES of c-IAP1 in its CARD, a motif involved in protein-protein interactions, could also indicate a role for an unidentified protein partner in the modulation of c-IAP1 translocation. Such a protein partner has been identified for XIAP, which can be retained in the nucleus through interaction with XAF1.22 

We have observed that c-IAP1 was located in the Golgi apparatus of differentiated cells. Another IAP, referred to as BRUCE in mice and Apollon in humans, also localizes to the Golgi compartment and the vesicular system.43  In addition to being related to IAPs through a BIR motif, this giant protein is an ubiquitin-conjugating enzyme (E2).44  However, the functions of both Apollon and c-IAP1 in the Golgi apparatus remain to be elucidated. Proteins that accumulate in the Golgi structure can be secreted.45  We have observed that c-IAP1 did not accumulate in cholesterol- and sphingomyelin-enriched fractions of differentiated cells (data not shown), suggesting that the protein did not associate with microdomains described in the secretory pathway.46  c-IAP1 was shown to alter the cellular distribution of coexpressed reaper and grim drosophila proteins in mammalian cells, suggesting that c-IAP1 could modulate signaling pathways by sequestration of proteins in cell compartments.47 

c-IAP1 is not the only IAP to translocate from the nucleus to the cytoplasm. The main isoform of survivin, a single-BIR-containing IAP survivin involved in the control of the mitotic spindle checkpoint,7  can be exported from the nucleus through an LMB-sensitive mechanism, whereas its alternative isoform survivin-ΔEx3 is driven to the nucleus by a C-terminal NLS.48  In the present study, we have observed that XIAP was also exported from the nucleus in U937 cells undergoing differentiation, together with c-IAP1. Of importance, survivin is ubiquitylated and degraded when cells exit the cell cycle, whereas c-IAP1 and XIAP protein levels remain stable along the differentiation process, which suggest that cellular redistribution of these latter proteins may be essential in their regulation.

Two IAPs have previously been shown to interfere with cell differentiation (ie, overexpressed human NAIP prevents neuronal differentiation of PC12 cells,49  whereas transgenic mice overexpressing XIAP in their lymphocytes demonstrate altered T-cell maturation).50  Here, we show that overexpression of either c-IAP1 or its NES-mutated protein interferes with TPA-induced differentiation of human leukemic cells. How these IAPs interfere with cell differentiation remains unidentified. The first function assigned to c-IAP1 has been its ability to interact with and to inhibit caspases.38  These enzymes have been involved in cell differentiation processes, both in humans23,27  and drosophila.51  Obviously, caspase activity must be carefully controlled when associated with cell differentiation to prevent cell death by apoptosis and it is attractive to speculate that IAPs play a role in this control. In accordance with this hypothesis, the BIR-containing protein dBRUCE was proposed to bind to and degrade caspases involved in drosophila spermatogenesis.51  We have recently shown that a limited activation of several caspases was required for the differentiation of human peripheral blood monocytes into macrophages, whereas their differentiation into dendritic cells did not depend on caspase activation.23  Since the nucleocytoplasmic translocation of c-IAP1 was observed in both caspase-dependent (macrophages) and -independent (dendritic cells) pathways of differentiation, the negative control of caspase activity may not be the function of c-IAP1 shuttling. In addition, caspases have been observed moving from the cytoplasm to other cell compartments in leukemic cells undergoing differentiation but were not identified to associate with the Golgi apparatus.52  Finally, we have shown previously that the postmitochondrial pathway to apoptosis remained functional in TPA-differentiated U937 cells, indicating that redistributed c-IAP1 did not prevent caspase activation by cytochrome-c released from the mitochondria.

Another function assigned to c-IAP1 and other IAPs containing a RING finger motif is ubiquitylation of proteins. c-IAP1 catalyzes its own ubiquitylation in vitro,8  promotes ubiquitylation of the apoptosis inducer Smac/DIABLO,15  and modulates cell response to TNF-α through the regulation of the intracellular level of TRAF216  and NF-κB essential modulator (NEMO).53  Preliminary studies suggest that c-IAP1 down-regulation decreases the proliferation rate of THP1 cells (data not shown). Together with the role of the ubiquitin/proteasome pathway in the regulation of cell cycle, this observation could indicate a connection between c-IAP1 redistribution and growth inhibition in cells undergoing differentiation.

The nucleocytoplasmic traffic of proteins modulates cellular functions.54  Accordingly, LMB prevents TPA-induced differentiation of U937 and THP1 cells. However, the role of the inhibition of c-IAP1 translocation in LMB-induced inhibition of cell differentiation remains to be determined. On the other hand, inhibition of the differentiation process correlates with a lack of nuclear export of c-IAP1, as observed in Bcl-2-overexpressing cells, in TPA-resistant U937 cells, and in monocytes from patients with CMML that do not respond to M-CSF-induced differentiation. Bcl-2 and related proteins were involved in the regulation of differentiation (eg, in restricting lineage determination during hematopoietic differentiation).55  The ability of overexpressed Bcl-2 to prevent the translocation of c-IAP1 could be a nonspecific consequence of its influence on the differentiation process. However, we cannot rule out that Bcl-2 directly interferes with the translocation of c-IAP1, since the protein is expressed at multiple cellular sites including the nuclear outer membrane56  and the nucleus.57 

Altogether, the present study suggests that c-IAP1 functions may be regulated in part by the subcellular location of the protein that is present mainly in the nucleus of various types of undifferentiated cells and translocates to the cytoplasm when these cells undergo differentiation. Ongoing studies may indicate whether this redistribution of c-IAP1 plays an active role in the differentiation process or confers specific functions to differentiated cells.

Prepublished online as Blood First Edition Paper, June 8, 2004; DOI 10.1182/blood-2004-01-0065.

Supported by grants from the Ligue Nationale Contre le Cancer.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

The author thanks M. Yoshida for kindly providing LMB; J. C. Reed for c-IAP1 cDNA; P. J. Parker, T. Lesuffleur, and J. Breard for cell lines; Irving Weismann and Eric Lagasse for the MRP8BCL-2 transgenic mice; and B. Goud, S. Khochbin, O. Hermine, and M. Fontenay for fruitful discussions.

1
Uren AG, Coulson EJ, Vaux DL. Conservation of baculovirus inhibitor of apoptosis repeat proteins (BIRPs) in viruses, nematodes, vertebrates and yeasts.
Trends Biochem Sci
.
1998
;
23
:
159
-162.
2
Salvesen GS, Duckett CS. IAP proteins: blocking the road to death's door.
Nat Rev Mol Cell Biol
.
2002
;
3
:
401
-410.
3
Srinivasula SM, Hegde R, Saleh A, et al. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis.
Nature
.
2001
;
410
:
112
-116.
4
Chai J, Shiozaki E, Srinivasula SM, et al. Structural basis of caspase-7 inhibition by XIAP.
Cell
.
2001
;
104
:
769
-780.
5
Riedl SJ, Renatus M, Schwarzenbacher R, et al. Structural basis for the inhibition of caspase-3 by XIAP.
Cell
.
2001
;
104
:
791
-800.
6
Roy N, Deveraux QL, Takahashi R, Salvesen GS, Reed JC. The c-IAP-1 and c-IAP-2 proteins are direct inhibitors of specific caspases.
EMBO J
.
1997
;
16
:
6914
-6925.
7
Li F, Ambrosini G, Chu EY, et al. Control of apoptosis and mitotic spindle checkpoint by survivin.
Nature
.
1998
;
396
:
580
-584.
8
Yang Y, Fang S, Jensen JP, Weissman AM, Ashwell JD. Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli.
Science
.
2000
;
288
:
874
-877.
9
Rothe M, Pan MG, Henzel WJ, Ayres TM, Goeddel DV. The TNFR2-TRAF signaling complex contains two novel proteins related to baculoviral inhibitor of apoptosis proteins.
Cell
.
1995
;
83
:
1243
-1252.
10
Yamaguchi K, Nagai S, Ninomiya-Tsuji J, et al. XIAP, a cellular member of the inhibitor of apoptosis protein family, links the receptors to TAB1-TAK1 in the BMP signaling pathway.
EMBO J
.
1999
;
18
:
179
-187.
11
Birkey Reffey S, Wurthner JU, Parks WT, Roberts AB, Duckett CS. X-linked inhibitor of apoptosis protein functions as a cofactor in transforming growth factor-beta signaling.
J Biol Chem
.
2001
;
276
:
26542
-26549.
12
Sanna MG, da Silva Correia J, Ducrey O, et al. IAP suppression of apoptosis involves distinct mechanisms: the TAK1/JNK1 signaling cascade and caspase inhibition.
Mol Cell Biol
.
2002
;
22
:
1754
-1766.
13
Huang H, Joazeiro CA, Bonfoco E, Kamada S, Leverson JD, Hunter T. The inhibitor of apoptosis, c-IAP2, functions as a ubiquitin-protein ligase and promotes in vitro monoubiquitination of caspases 3 and 7.
J Biol Chem
.
2000
;
275
:
26661
-26664.
14
Suzuki Y, Nakabayashi Y, Takahashi R. Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its anti-apoptotic effect in Fas-induced cell death.
Proc Natl Acad Sci U S A
.
2001
;
98
:
8662
-8667.
15
Hu S, Yang X. Cellular inhibitor of apoptosis 1 and 2 are ubiquitin ligases for the apoptosis inducer Smac/DIABLO.
J Biol Chem
.
2003
;
278
:
10055
-10060.
16
Li X, Yang Y, Ashwell JD. TNF-RII and c-IAP1 mediate ubiquitination and degradation of TRAF2.
Nature
.
2002
;
416
:
345
-347.
17
Chu ZL, McKinsey TA, Liu L, Gentry JJ, Malim MH, Ballard DW. Suppression of tumor necrosis factor-induced cell death by inhibitor of apoptosis c-IAP2 is under NF-kappaB control.
Proc Natl Acad Sci U S A
.
1997
;
94
:
10057
-10062.
18
Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS Jr. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
Science
.
1998
;
281
:
1680
-1683.
19
Stehlik C, de Martin R, Kumabashiri I, Schmid JA, Binder BR, Lipp J. Nuclear factor (NF)-kappaB-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha-induced apoptosis.
J Exp Med
.
1998
;
188
:
211
-216.
20
Holcik M, Lefebvre C, Yeh C, Chow T, Korneluk RG. A new internal-ribosome-entry-site motif potentiates XIAP-mediated cytoprotection.
Nat Cell Biol
.
1999
;
1
:
190
-192.
21
Verhagen AM, Ekert PG, Pakusch M, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins.
Cell
.
2000
;
102
:
43
-53.
22
Liston P, Fong WG, Kelly NL, et al. Identification of XAF1 as an antagonist of XIAP anti-Caspase activity.
Nat Cell Biol
.
2001
;
3
:
128
-133.
23
Sordet O, Rebe C, Plenchette S, et al. Specific involvement of caspases in the differentiation of monocytes into macrophages.
Blood
.
2002
;
100
:
4446
-4453.
24
Lacour S, Micheau O, Hammann A, et al. Chemotherapy enhances TNF-related apoptosis-inducing ligand DISC assembly in HT29 human colon cancer cells.
Oncogene
.
2003
;
22
:
1807
-1816.
25
Lesuffleur T, Porchet N, Aubert JP, et al. Differential expression of the human mucin genes MUC1 to MUC5 in relation to growth and differentiation of different mucus-secreting HT-29 cell subpopulations.
J Cell Sci
.
1993
;
106
(pt 3):
771
-783.
26
Kiley SC, Adams PD, Parker PJ. Cloning and characterization of phorbol ester differentiation-resistant U937 cell variant.
Cell Growth Differ
.
1997
;
8
:
221
-230.
27
Zermati Y, Garrido C, Amsellem S, et al. Caspase activation is required for terminal erythroid differentiation.
J Exp Med
.
2001
;
193
:
247
-254.
28
De Botton S, Sabri S, Daugas E, et al. Platelet formation is the consequence of caspase activation within megakaryocytes.
Blood
.
2002
;
100
:
1310
-1317.
29
Lagasse E, Weissman IL. bcl-2 inhibits apoptosis of neutrophils but not their engulfment by macrophages.
J Exp Med
.
1994
;
179
:
1047
-1052.
30
Sordet O, Rebe C, Leroy I, et al. Mitochondria-targeting drugs arsenic trioxide and lonidamine bypass the resistance of TPA-differentiated leukemic cells to apoptosis.
Blood
.
2001
;
97
:
3931
-3940.
31
Rao RV, Hermel E, Castro-Obregon S, et al. Coupling endoplasmic reticulum stress to the cell death program: mechanism of caspase activation.
J Biol Chem
.
2001
;
276
:
33869
-33874.
32
Togawa A, Morinaga N, Ogasawara M, Moss J, Vaughan M. Purification and cloning of a brefeldin A-inhibited guanine nucleotide-exchange protein for ADP-ribosylation factors.
J Biol Chem
.
1999
;
274
:
12308
-12315.
33
Fornerod M, Ohno M, Yoshida M, Mattaj IW. CRM1 is an export receptor for leucine-rich nuclear export signals.
Cell
.
1997
;
90
:
1051
-1060.
34
Pizao PE, Lyaruu DM, Peters GJ, et al. Growth, morphology and chemosensitivity studies on postconfluent cells cultured in `V'-bottomed microtiter plates.
Br J Cancer
.
1992
;
66
:
660
-665.
35
Werneburg BG, Zoog SJ, Dang TT, Kehry MR, Crute JJ. Molecular characterization of CD40 signaling intermediates.
J Biol Chem
.
2001
;
276
:
43334
-43342.
36
Kuai J, Nickbarg E, Wooters J, Qiu Y, Wang J, Lin LL. Endogenous association of TRAF2, TRAF3, c-IAP1, and Smac with lymphotoxin beta receptor reveals a novel mechanism of apoptosis.
J Biol Chem
.
2003
;
278
:
14363
-14369.
37
Deveraux QL, Roy N, Stennicke HR, et al. IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases.
EMBO J
.
1998
;
17
:
2215
-2223.
38
Gorlich D. Transport into and out of the cell nucleus.
EMBO J
.
1998
;
17
:
2721
-2727.
39
Kao HY, Verdel A, Tsai CC, Simon C, Juguilon H, Khochbin S. Mechanism for nucleocytoplasmic shuttling of histone deacetylase 7.
J Biol Chem
.
2001
;
276
:
47496
-47507.
40
Lohrum MA, Woods DB, Ludwig RL, Balint E, Vousden KH. C-terminal ubiquitination of p53 contributes to nuclear export.
Mol Cell Biol
.
2001
;
21
:
8521
-8532.
41
Perander M, Bjorkoy G, Johansen T. Nuclear import and export signals enable rapid nucleocytoplasmic shuttling of the atypical protein kinase C lambda.
J Biol Chem
.
2001
;
276
:
13015
-13024.
42
Ura S, Masuyama N, Graves JD, Gotoh Y. Caspase cleavage of MST1 promotes nuclear translocation and chromatin condensation.
Proc Natl Acad Sci U S A
.
2001
;
98
:
10148
-10153.
43
Chen Z, Naito M, Hori S, Mashima T, Yamori T, Tsuruo T. A human IAP-family gene, apollon, expressed in human brain cancer cells.
Biochem Biophys Res Commun
.
1999
;
264
:
847
-854.
44
Hauser HP, Bardroff M, Pyrowolakis G, Jentsch S. A giant ubiquitin-conjugating enzyme related to IAP apoptosis inhibitors.
J Cell Biol
.
1998
;
141
:
1415
-1422.
45
Tooze SA, Martens GJ, Huttner WB. Secretory granule biogenesis: rafting to the SNARE.
Trends Cell Biol
.
2001
;
11
:
116
-122.
46
Gkantiragas I, Brugger B, Stuven E, et al. Sphingomyelin-enriched microdomains at the Golgi complex.
Mol Biol Cell
.
2001
;
12
:
1819
-1833.
47
McCarthy JV, Dixit VM. Apoptosis induced by Drosophila reaper and grim in a human system: attenuation by inhibitor of apoptosis proteins (c-IAPs).
J Biol Chem
.
1998
;
273
:
24009
-24015.
48
Rodriguez JA, Span SW, Ferreira CG, Kruyt FA, Giaccone G. CRM1-mediated nuclear export determines the cytoplasmic localization of the antiapoptotic protein Survivin.
Exp Cell Res
.
2002
;
275
:
44
-53.
49
Gotz R, Karch C, Digby MR, Troppmair J, Rapp UR, Sendtner M. The neuronal apoptosis inhibitory protein suppresses neuronal differentiation and apoptosis in PC12 cells.
Hum Mol Genet
.
2000
;
9
:
2479
-2489.
50
Conte D, Liston P, Wong JW, Wright KE, Korneluk RG. Thymocyte-targeted overexpression of xiap transgene disrupts T lymphoid apoptosis and maturation.
Proc Natl Acad Sci U S A
.
2001
;
98
:
5049
-5054.
51
Arama E, Agapite J, Steller H. Caspase activity and a specific cytochrome C are required for sperm differentiation in Drosophila.
Dev Cell
.
2003
;
4
:
687
-697.
52
Sordet O, Rebe C, Dubrez-Daloz L, Boudard D, Solary E. Intracellular redistribution of procaspases during TPA-induced differentiation of U937 human leukemic cells.
Leukemia
.
2002
;
16
:
1569
-1570.
53
Tang ED, Wang CY, Xiong Y, Guan KL. A role for NEMO/IKKgamma ubiquitination in the activation of the Ikappa B kinase complex by TNF-alpha.
J Biol Chem
.
2003
;
278
:
37297
-37305.
54
Weis K. Regulating access to the genome: nucleocytoplasmic transport throughout the cell cycle.
Cell
.
2003
;
112
:
441
-451.
55
Haughn L, Hawley RG, Morrison DK, von Boehmer H, Hockenbery DM. BCL-2 and BCL-XL restrict lineage choice during hematopoietic differentiation.
J Biol Chem
.
2003
;
278
:
25158
-25165.
56
Krajewski S, Tanaka S, Takayama S, Schibler MJ, Fenton W, Reed JC. Investigation of the subcellular distribution of the bcl-2 oncoprotein: residence in the nuclear envelope, endoplasmic reticulum, and outer mitochondrial membranes.
Cancer Res
.
1993
;
53
:
4701
-4714.
57
Hoetelmans R, van Slooten HJ, Keijzer R, Erkeland S, van de Velde CJ, Dierendonck JH. Bcl-2 and Bax proteins are present in interphase nuclei of mammalian cells.
Cell Death Differ
.
2000
;
7
:
384
-392.
Sign in via your Institution