Dying cells, apoptotic or necrotic, are swiftly eliminated by professional phagocytes. We previously reported that CD47 engagement by CD47 mAb or thrombospondin induced caspase-independent cell death of chronic lymphocytic leukemic B cells (B-CLL). Here we show that human immature dendritic cells (iDCs) phagocytosed the CD47 mAb–killed leukemic cells in the absence of caspases 3, 7, 8, and 9 activation in the malignant lymphocytes. Yet the dead cells displayed the cytoplasmic features of apoptosis, including cell shrinkage, phosphatidylserine exposure, and decreased mitochondrial transmembrane potential (ΔΨm). CD47 mAb–induced cell death also occurred in normal resting and activated lymphocytes, with B-CLL cells demonstrating the highest susceptibility. Importantly, iDCs and CD34+ progenitors were resistant. Structure-function studies in cell lines transfected with various CD47 chimeras demonstrated that killing exclusively required the extracellular and transmembrane domains of the CD47 molecule. Cytochalasin D, an inhibitor of actin polymerization, and antimycin A, an inhibitor of mitochondrial electron transfer, completely suppressed CD47-induced phosphatidylserine exposure. Interestingly, CD47 ligation failed to induce cell death in mononuclear cells isolated from Wiskott-Aldrich syndrome (WAS) patients, suggesting the involvement of Cdc42/WAS protein (WASP) signaling pathway. We propose that CD47-induced caspase-independent cell death be mediated by cytoskeleton reorganization. This form of cell death may be relevant to maintenance of homeostasis and as such might be explored for the development of future therapeutic approaches in lymphoid malignancies.

Apoptosis is a noninflammatory destruction process essential to the regulation of immune system and homeostasis.1,2 It provides molecular basis for T- and B-cell development,3 induction of immune tolerance, and termination of normal immune response.4 By contrast, necrosis is perceived by the immune system as a danger signal that triggers inflammatory response and acquired immunity. Classically, the activation and function of a set of proteinases, the caspases, is the key event in apoptosis, with mitochondria playing a central role.5,6 A critical event, besides the execution phase of apoptosis, is the engulfment of dying cells by professional antigen-presenting cells (APCs) before the dead cells disgorge their toxic materials in the surrounding milieu.7 

In recent years, cell death other than necrosis was reported to occur in the absence of caspase activation in hematopoietic cells; it included glucocorticoid-induced death of thymocytes, Bax-mediated cell death, and death of cell lines induced by growth factor withdrawal.8-10 A growing number of surface molecules were shown to be involved in the induction of this nonclassical caspase-independent cell death. Engagement of CD2, CD45, CD47, CD99, and MHC (major histocompatibility complex) class II and I activated this death process.11-17Caspase-independent cell death induced by CD47 ligation in B-chronic lymphocytic leukemic (B-CLL) cells was characterized by cell shrinkage, exposure of phosphatidylserine (PS), and mitochondrial matrix swelling in complete absence of nuclear degradation.13 

CD47 antigen is ubiquitously expressed on hematopoietic and nonhematopoietic cells.18 It serves as a receptor for thrombospondin (TSP) and as a ligand for transmembrane signal regulatory protein (SIRP-α), mainly expressed on myeloid and neuronal cells.19,20 Through its association with integrins of β1, β2, and β3 families, it initiated heterotrimeric G-protein signaling and thus modulated cell motility, leukocyte adhesion and migration, phagocytosis, and platelet activation.21 On immune cells, CD47 ligation by soluble mAbs was shown to inhibit cytokine production by APCs and interleukin-12 (IL-12) responsiveness by neonatal and adult T cells.22-25 When immobilized, CD47 mAb costimulated T-cell receptor (TCR)–activated T cells.26,27 Thus, the biologic consequences of CD47 activation seemingly vary according to (1) the way the molecule is engaged, (2) the surface molecules it interacts with, and (3) its conformation and membrane localization, all of which depend on the cell type on which CD47 is expressed.26,28-32 

The molecular basis for caspase-independent cell death remains elusive. Apoptosis-inducing factor (AIF), released by mitochondria, induced caspase-independent nuclear degradation.33 In most of the studies, sole PS externalization in the presence of the broad caspase inhibitor Z-VAD.fmk (benzyloxycarbonyl–Val-Ala-Asp–OMe–fluoromethyl ketone) was used to define caspase-independent cell death pathway. Coexistence of caspase-dependent and -independent pathways occurred in various systems, including CD2 and CD95 ligation.11,34,35 For instance, in CD2-mediated apoptosis of T lymphocytes, the mitochondrial release of AIF preceded the dissipation of transmembrane potential (ΔΨm), the release of cytochrome c (cyt c), and the caspase-dependent execution phase of apoptosis.36 

Our present findings confirm and extend to other cell populations our previous observations that CD47 ligation exclusively induced the cytoplasmic events of apoptosis in B-CLL cells. We here show that DCs recognized and eliminated B-CLL dead cells and present evidence that cytoskeleton rearrangement was a triggering event in ΔΨm loss and PS exposure after CD47 ligation on human cells.

Patients

Diagnosis of B-CLL patients included in this study was based on clinical examination and peripheral blood count. CLL was defined as > 5000/μL lymphocytes expressing CD5, CD20, and CD23. Authorized consent forms for patients were obtained before blood collection. Patients with Wiskott-Aldrich syndrome (WAS) exhibited typical features, that is, thrombocytopenia with small platelets, eczema, and susceptibility to infections, a severe clinical phenotype. In patient 1, A nucleotide insertion at position 147 in exon 2 of the WAS gene was detected. In patients 2 and 3 there was a C to T missense mutation at nucleotide 631 in exon 7 and 155 in exon 1, respectively. In patient 4, a G to A mutation at position 777 in intron 8 was found. Blood samples were obtained following informed consent of parents according to the Declaration of Helsinki, and the study was approved by the institutional ethics committee of the Centre de Recherche du CHUM and Hôpital Necker.

Cell lines and transfectants

RAJI is a Burkitt lymphoma; RPMI 8226 and RPMI 8866 are B-lymphocytic cell lines; Jurkat is a T-cell hybridoma; KU812 is a granulocytic cell line; U937 and THP-1 are monocytic cell lines; K-562 is a chronic myelogenic leukemia cell; OV10 is an ovarian carcinoma transfected with CD47 cDNA37; and JinB8 is a CD47-negative Jurkat cell line.26 The different cDNAs and constructs used in this study were previously described.26Transfected Jurkat, JinB8, and U937 cells were sorted for high transgene expression using a FACSort (Lysys II Software, Becton Dickinson, Mississauga, ON, Canada).

Reagents

Recombinant human IL-4, soluble CD40 ligand, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were kindly provided by Immunex (Seattle, WA) and Dr D. Bron (Institut Bordet, Brussels, Belgium), respectively. Dr R.-P. Sekaly (Université de Montréal, Montreal, Canada) kindly provided polyclonal anti–caspase-3 Ab. Anti–human CD3 (UCHT-1) was provided by P. Beverley (University College and Middlesex School of Medicine, London, United Kingdom). Isotype-matched negative control mAb (mouse IgG1) was prepared in our laboratory.

The other antibodies and reagents used in this study were purchased from manufacturers as indicated: anti-CD47 mAbs (clone B6H12), Bioscience (Camarillo, CA); polyclonal anti–caspase-8, Upstate Biotechnology (Lake Placid, NY); polyclonal antibody against caspase-9 and anti–mouse CD8-α (clone 53-6.7), Pharmingen (San Diego, CA); and anti–caspase-7 mAb, Transduction Laboratories (Lexington, KY).

Cell preparation and culture conditions

B cells were isolated from CLL patients or from tonsils by density gradient centrifugation of heparinized blood or cell suspension, respectively, using Lymphoprep (Nycomed, Olso, Norway) followed by one cycle of rosetting with S-(2 aminoethyl) isothiouronium bromide–treated (Aldrich, Milwaukee, WI) sheep red blood cells to deplete T cells. B-cell purity was shown to be > 98% by flow cytometry (FACSort, Becton Dickinson). Highly purified T cells were obtained from monocyte-depleted peripheral blood mononuclear cells (PBMCs) from healthy volunteers by rosetting with aminoethylisothiouronium bromide (AET)–treated sheep red blood cells (SRBC), followed by treatment of rosette-forming cells with Lympho-Kwik T (One Lambda, Los Angeles, CA), following manufacturer's recommendations. Cell purity was assessed by flow cytometry using phycoerythrin (PE)–conjugated anti-CD3, anti-CD4, or anti-CD8 mAbs (Ancell, London, ON, Canada) and was shown to be > 98%. CD34+ cells were obtained from heparinized cord blood using the Dynal CD34 progenitor cell selection system according to the manufacturer's instructions (Dynal Skøyen, Oslo, Norway). Human monocyte-derived iDCs were prepared exactly as described.28 

Highly purified lymphocytes were cultured at 4 × 106/mL, iDC, lines, and transfectants at 2 × 106/mL in 100 μL of HB101 serum-free synthetic medium (Irvine Scientific, Edmonton, AB, Canada) or in special medium when indicated, on flat-bottomed 96-well plates (Nunc, Edmonton, AB, Canada) in the presence of soluble or immobilized mAbs. Plates were precoated with anti-CD47 or anti–CD8-α mAbs at 10 μg/mL in 100 μL 0.1M NaHCO3, pH 9, overnight at 4°C, then washed and blocked with medium. The following inhibitors were added 15 minutes before plating cells when indicated; only cytochalasin D–treated cells were washed before plating: antimycin A (30 μM) (Sigma, St Louis, MO), pertussis toxin (25-100 ng/mL), cytochalasin D (20 μM), rottlerin (5-20 μM) (Calbiochem, San Diego, CA), cyclosporin A (5 μM), aristolochic acid (50 μM), and bongkrekic acid (50 μM) (Biomol, Plymouth Meeting, PA).

For potassium efflux experiments, cells were cultured in 1% fetal calf serum (FCS) Na+K+-free RPMI medium, supplemented with normal or inverted [Na+]/[K+] ratio as described.38 

Phagocytosis assay

Freshly purified B-CLL cells were stained with PKH26 red fluorescent cell linker (Sigma) according to the manufacturer's instructions before induction of apoptosis by culture for 16 hours on immobilized CD47 mAb, hydrocortisone (HC) (5 × 10−4 M), or soluble CD47 mAb as negative control. Cells were washed and given to iDCs (2 × 105/mL) as a phagocytic meal (10 apoptotic cells per iDC) for 3 hours at 37°C. Endocytosis of PKH26-stained B cells was determined by fluorescence-activated cell-sorter scanner (FACS) after gating on live iDCs.

Flow cytometry analysis

CD47 and constructs expression was assessed using a 2-step procedure. Briefly, cells were incubated for 1 hour at 4°C with biotinylated CD47 mAb (B6H12) or unconjugated anti-CD8α (clone no. 53-6.7) or isotype-matched control mAb (5 μg/mL). After washing, cells were incubated with PE-labeled streptavidin or fluorescein isothiocyanate (FITC)–conjugated antirat mAb (Ancell) and analyzed by flow cytometry (FACSort, Becton Dickinson).

Assays for apoptosis

Detection of PS exposure, caspase 3 activity, and decrease in ΔΨm were performed by flow cytometry using a FACSort (Lysys II Software, Becton Dickinson). For detection of PS exposure, cells were double-stained with FITC-labeled annexin-V (R&D Systems, Minneapolis, MN) and propidium iodide (PI) at 2 μg/mL (Sigma). Decrease of ΔΨm was assessed using 3,3′–dihexyloxacarbocyanine iodide (DiOC6) (Molecular Probes, Hornby, ON, Canada). Caspase 3 activity was detected by flow cytometry in unfixed cells using a fluorogenic substrate (PhiPhilux, OncoImmunium, Gaithersburg, MD). Caspases 3, 7, and 9 cleavage products and caspase 8 expression were analyzed by Western blotting. Briefly, 5 × 106 cells were directly lysed in hot sample buffer containing 10% β-mercaptoethanol. Samples were then boiled for 5 minutes, electrophorezed on 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE), and transferred onto polyvinylidenefluoride (PVDF) membranes (Millipore, Mississauga, MN). Membranes were probed with indicated antibodies, and immunoreactive products were revealed using enhanced chemiluminescence (ECL; Amersham, Baie d'Urfe, QC, Canada).

CD47-induced cell death was calculated as follows:

(%of live cells in control)(%of live cells onCD47mAb)(%of live cells in control)×100

Statistical analysis

The Student paired t test was used for statistical analysis.

Engulfment by professional phagocytes of CD47 mAb-treated B-CLL cells dying in the absence of caspase activation

We previously reported that CD47 ligation by immobilized anti-CD47 monoclonal antibodies (CD47 mAbs) or its natural ligand, TSP,13 induced caspase-independent cell death in B-CLL cells. This cell death was characterized by the cytoplasmic features of apoptosis. They included cell shrinking, exposure of PS to the outer leaflet membrane and, as shown in Figure1A, a drop in mitochondrial membrane potential (ΔΨm) as demonstrated by decreased 3,3′–dihexyloxacarbocyanine iodide (DiOC6) staining. This cell death was considered to be caspase independent since the nucleus of the leukemic dead cells remained intact and PS exposure was not prevented by the presence of the broad caspase inhibitor z-Val-Ala-Asp-(OMe)–fluoromethylketone (zVAD-fmk).13 To formally demonstrate that the caspase remained inactive during this process, we performed Western blot analysis (Figure 1B) to search for the cleavage products of caspases in CD47 mAb-treated leukemic cells. We failed to detect any cleavage products of caspases 3, 7, and 9, and similar amounts of procaspase-8 were found in immobilized CD47 mAb and control mAb-treated B-CLL cells. The absence of caspases 3/7 activity in individual CD47-treated cells was confirmed by flow cytometry using a fluorogenic caspase substrate (Figure 1C). HC-treated B-CLL cells displayed caspase 3 activity. Note that B-CLL cells with a low level of spontaneous apoptosis were selected for these experiments.

Fig. 1.

Immature dendritic cells efficiently phagocytosed B-CLL cells killed by CD47-induced caspase-independent pathway.

Freshly isolated B-CLL cells were cultured in the presence of soluble or immobilized CD47 mAb (10 μg/mL), control mAb (10 μg/mL), or HC (5 × 10−4 M). (A) Cells were double-stained with FITC-labeled annexin-V and PI (upper graphs) or with DiOC6(lower graphs) and analyzed by flow cytometry. Shown are percent of dead cells (annexin-V+ or DiOC6low). (B) Soluble (lane 1) or immobilized (lane 2) CD47 mAb-treated cells were lysed, and Western blot analysis was performed for detection of caspases 3, 7, 8, and 9 cleavage products (as indicated by *). Asterisks indicate “expected molecular weight” of caspase cleavage products; arrowheads, molecular weight of procaspases. (C) Caspase 3 activity was measured after 48 hours by flow cytometry using the cell permeable fluorogenic substrate DEVDase. (D) B-CLL cells were stained in red with PKH26 linker before treatment overnight with soluble or immobilized CD47 mAb or HC. Cells were then cocultured for 3 hours with iDCs at a 10:1 ratio. The mixture was analyzed by flow cytometry for red fluorescence (FL2) after gating on iDCs. Shown are percent of iDCs that have phagocytosed red dead cells. One representative experiment of 3 is shown.

Fig. 1.

Immature dendritic cells efficiently phagocytosed B-CLL cells killed by CD47-induced caspase-independent pathway.

Freshly isolated B-CLL cells were cultured in the presence of soluble or immobilized CD47 mAb (10 μg/mL), control mAb (10 μg/mL), or HC (5 × 10−4 M). (A) Cells were double-stained with FITC-labeled annexin-V and PI (upper graphs) or with DiOC6(lower graphs) and analyzed by flow cytometry. Shown are percent of dead cells (annexin-V+ or DiOC6low). (B) Soluble (lane 1) or immobilized (lane 2) CD47 mAb-treated cells were lysed, and Western blot analysis was performed for detection of caspases 3, 7, 8, and 9 cleavage products (as indicated by *). Asterisks indicate “expected molecular weight” of caspase cleavage products; arrowheads, molecular weight of procaspases. (C) Caspase 3 activity was measured after 48 hours by flow cytometry using the cell permeable fluorogenic substrate DEVDase. (D) B-CLL cells were stained in red with PKH26 linker before treatment overnight with soluble or immobilized CD47 mAb or HC. Cells were then cocultured for 3 hours with iDCs at a 10:1 ratio. The mixture was analyzed by flow cytometry for red fluorescence (FL2) after gating on iDCs. Shown are percent of iDCs that have phagocytosed red dead cells. One representative experiment of 3 is shown.

Close modal

Both caspase activation and PS exposure generally have been considered as prerequisites for recognition and phagocytosis of cells dying by apoptosis.39,40 It was therefore important to determine whether CD47 mAb–killed B-CLL cells could signal their death to phagocytes and be eliminated. To test this hypothesis, we performed a phagocytosis assay using human monocyte-derived iDCs as professional phagocytes.41 B-CLL cells were stained by PKH26, a nontoxic red fluorescent cell-linker, and exposed for 16 hours to immobilized CD47 mAb or HC, a drug that induced caspase-dependent apoptosis in the leukemic cells.42 Soluble CD47 mAb did not induce cell death13 nor Fc-mediated phagocytosis23 and was used as negative control. Treated samples were cocultured with iDCs for 3 hours. Phagocytosis was assessed by flow cytometry by quantifying FL2 fluorescence after gating on iDCs (by size scatter). Results indicated that B-CLL killed by immobilized CD47 mAb were phagocytosed as efficiently as HC-treated cells (39% vs 46% FL2-positive cells) (Figure 1D). Note that each cell preparation was stained separately with FITC-labeled annexin-V to evaluate their percentage of annexin-V–positive cells (39%, 75%, and 83% for soluble, immobilized CD47 mAb, and HC, respectively).

Taken together, these results indicate that the caspase-independent death signal delivered by CD47 ligation to B-CLL cells is sufficient to allow their elimination by iDCs.

Blood cell susceptibility to CD47-induced cell death

Since CD47 Ag is ubiquitously expressed on hematopoietic cells, we assessed the in vitro sensitivity of normal blood cells to CD47-induced cell death. From the perspective of a potential therapeutic use of this mAb, we first examined CD34+ cells since they represent the pool of hematopoietic progenitors.43,44 As depicted in Figure 2A, CD34+ cells remained totally insensitive to CD47 mAb–induced killing (0% of cell-death induction, n = 6), despite their high level of CD47 expression. Immature DCs are major players in the clearance of apoptotic cells. Like CD34+ cells, they were resistant to CD47-induced cell death (0%, n = 4) (Figure 2A).

Fig. 2.

Differential susceptibility of normal human blood cells to CD47-induced cell death.

(A-B) Cord-blood CD34+ cells (stem cells) (n = 6), monocyte-derived iDCs (n = 4), resting and activated tonsillar B cells (n = 4), and peripheral T cells (n = 4) were stained for CD47 expression (B6H12 mAb) and analyzed by flow cytometry: isotype-control mAb (open histogram), CD47 mAb (shaded histogram). (A) Stem cells and iDCs were cultured in the absence or presence of immobilized CD47 mAbs (10 μg/mL) for 18 hours and double-stained with FITC-labeled annexin-V and PI. Shown are percent of viable cells (annexin-V/PI). (B) Resting T and B cells were left unstimulated or activated for 48 hours with plastic-coated anti-CD3 (3 μg/mL) or sCD40L (1 μg/mL) + IL-4 (10 ng/mL), respectively, and then cultured overnight in the absence or presence of immobilized CD47 mAbs. The mean percent of CD47-induced cell death, calculated as indicated in “Patients, materials, and methods” are shown.

Fig. 2.

Differential susceptibility of normal human blood cells to CD47-induced cell death.

(A-B) Cord-blood CD34+ cells (stem cells) (n = 6), monocyte-derived iDCs (n = 4), resting and activated tonsillar B cells (n = 4), and peripheral T cells (n = 4) were stained for CD47 expression (B6H12 mAb) and analyzed by flow cytometry: isotype-control mAb (open histogram), CD47 mAb (shaded histogram). (A) Stem cells and iDCs were cultured in the absence or presence of immobilized CD47 mAbs (10 μg/mL) for 18 hours and double-stained with FITC-labeled annexin-V and PI. Shown are percent of viable cells (annexin-V/PI). (B) Resting T and B cells were left unstimulated or activated for 48 hours with plastic-coated anti-CD3 (3 μg/mL) or sCD40L (1 μg/mL) + IL-4 (10 ng/mL), respectively, and then cultured overnight in the absence or presence of immobilized CD47 mAbs. The mean percent of CD47-induced cell death, calculated as indicated in “Patients, materials, and methods” are shown.

Close modal

We next observed a differential induction of cell death in normal resting and activated B and T lymphocytes. The percentage of CD47-induced cell death in normal resting B and T cells was 36% (n = 7, P < .01) and 49% (n = 14,P < .01), respectively (Figure 2B). B-CLL cells displayed the highest susceptibility (64%, n = 10). Unexpectedly, anti–CD3-stimulated T lymphocytes became almost insensitive to CD47 mAb-killing, whereas T-cell–dependent B-cell activation (sCD40L + IL-4) significantly increased the level of death induction (P < .01). Activation did not modulate CD47 expression as detected by CD47 mAb (clone B6H12) (Figure 2B).

Human cell lines represent the malignant counterpart of lymphoid and myeloid cells at different stages of maturation. In this context, we examined 4 lymphoid cell lines (RAJI, RPMI-8226, 8866, and Jurkat), 3 myeloid cell lines (U937, THP-1, and K-562), one granulocytic cell line (KU812), as well as the ovarian carcinoma cell line, OV10,37 transfected with CD47 cDNA. As shown in Figure3, these cell lines displayed differential sensitivity to CD47 mAb, with no correlation, as in untransformed cells, with the level of CD47 expression. In addition, the level of CD47-induced cell death induction did not discriminate between specific cell lineage, RAJI, THP-1, and K562 being resistant. Of interest, OV10 CD47 transfectants were efficiently killed by CD47 mAb (44% of death induction, n = 5, P < .001), indicating that CD47 expression was sufficient to confer cell death susceptibility in a cell that did not primarily express the CD47 molecule.

Fig. 3.

Susceptibility of human cell lines to CD47-induced cell death: lack of correlation between CD47 expression and CD47-induced cell death.

Various cell lines were stained for CD47 expression using B6H12 mAb: isotype-control mAb (open histogram) and CD47 mAb (shaded histogram). Cells were cultured overnight on immobilized control or CD47 mAb analyzed by flow cytometry. Shown are the mean percents of CD47-induced cell death, calculated as indicated in “Patients, materials, and methods.”

Fig. 3.

Susceptibility of human cell lines to CD47-induced cell death: lack of correlation between CD47 expression and CD47-induced cell death.

Various cell lines were stained for CD47 expression using B6H12 mAb: isotype-control mAb (open histogram) and CD47 mAb (shaded histogram). Cells were cultured overnight on immobilized control or CD47 mAb analyzed by flow cytometry. Shown are the mean percents of CD47-induced cell death, calculated as indicated in “Patients, materials, and methods.”

Close modal

From this data, we conclude that normal as well as transformed cells display differential susceptibility to CD47-induced cell death. CD34+ cells and immature DCs are resistant, whereas cell activation differentially modulates the intensity of the response, regardless of the level of CD47 expression.

CD47 extracellular and multiply membrane-spanning domains are necessary to signal cell death

As indicated above, Jurkat and U937 cell lines were sensitive to CD47-induced cell death. These cell lines were therefore suitable to perform structure-function studies to determine which portion(s) of the CD47 molecule was dispensable to mediate the cell death signaling. CD47 molecule is made of an extracellular immunoglobulinlike domain (IgV), a 5-membrane spanning domain (multiply membrane spanning [MMS]), and a cytoplasmic tail displaying 4 alternatively spliced isoforms (form 1 to 4 in order of increasing length).18,45 Form 2 of CD47 is predominant in the hematopoietic lineage, while the nearly tailless form 1 (only 3 amino acids) is expressed in keratinocytes and endothelial cells.

We first examined whether the cytoplasmic tail of CD47 was required. We used JinB8 cells, a CD47-negative Jurkat cell line,46 transfected with either of the 2 native splice forms (hIAP-form 1 and 2) or a chimera, CD8C2, made of the extracellular membrane domain of mouse CD8α fused to the cytoplasmic tail of CD47 form 2. Some experiments were performed on a U937 cell line transfected with CD8C2 cDNA. Since we observed that Jurkat sensitivity to CD47-induced cell death was augmented by pretreatment with phorbol 12-myristate 13-acetate (PMA) for 24 hours, we used PMA-activated cells in the next experiments. Results shown in Figure 4B indicate that the cytoplasmic tail of CD47 is not required to signal cell death. First, engagement of hIAP-form 1 and 2 molecules, equally expressed on JinB8, by immobilized CD47 mAb induced similar level of cell death (29% vs 21% death induction). Secondly, ligation of the chimera CD8C2 by CD8 mAb did not mediate cell death, either in JinB8, U937 or Jurkat (Figure 4B and data not shown). Of interest, PMA-activated Jurkat-CD8C2 cells did spread when endogenous CD47 or CD8C2 chimera was engaged by immobilized CD47 or anti-CD8 mAb, respectively (Figure 4C), indicating that the failure to induce cell death was not due to inappropriate chimera ligation.

Fig. 4.

Requirement for both extracellular and transmembrane domains for CD47-induced cell death.

(A) Various constructs or/and chimeras of the CD47 molecule (described in “Patients, materials, and methods”) were transfected into U937, Jurkat, or JinB8 (CD47−/−) cell lines. (B) Expression of the CD47 products (gray histograms) was determined by staining with B6H12 mAb (for hIAP form 1 and 2, IAP/CD7) or antimouse CD8α mAb (for CD8-MC2 and CD8-C2). Expression of endogenous CD47 (Jurkat and U937): cells were cultured overnight on immobilized antimouse CD8-α or CD47 mAb (10 μg/mL). *Jurkat and JinB8 transfectants were pretreated with PMA for 24 hours (2 ng/mL) before killing. CD47-induced cell death was calculated as described in “Patients, materials, and methods.” (C) Light microscopy of PMA-activated Jurkat cells transfected with CD8-C2 construct. Untreated cells (i), cells cultured for 18 hours with soluble (ii), immobilized CD47 mAb (iii) or immobilized antimouse CD8α mAb (iv).

Fig. 4.

Requirement for both extracellular and transmembrane domains for CD47-induced cell death.

(A) Various constructs or/and chimeras of the CD47 molecule (described in “Patients, materials, and methods”) were transfected into U937, Jurkat, or JinB8 (CD47−/−) cell lines. (B) Expression of the CD47 products (gray histograms) was determined by staining with B6H12 mAb (for hIAP form 1 and 2, IAP/CD7) or antimouse CD8α mAb (for CD8-MC2 and CD8-C2). Expression of endogenous CD47 (Jurkat and U937): cells were cultured overnight on immobilized antimouse CD8-α or CD47 mAb (10 μg/mL). *Jurkat and JinB8 transfectants were pretreated with PMA for 24 hours (2 ng/mL) before killing. CD47-induced cell death was calculated as described in “Patients, materials, and methods.” (C) Light microscopy of PMA-activated Jurkat cells transfected with CD8-C2 construct. Untreated cells (i), cells cultured for 18 hours with soluble (ii), immobilized CD47 mAb (iii) or immobilized antimouse CD8α mAb (iv).

Close modal

Previous studies on the CD47 molecule have demonstrated that IgV and MMS domains are generally necessary for CD47-induced cell spreading and costimulation.26,27,46,47 We therefore explored the requirement of the IgV or the MMS domains in CD47-induced cell death. To this end, we used 2 constructs: IAP/CD7, made of the CD47 IgV domain fused to the membrane and cytoplasmic domains of human CD7, and CD8MC2, made of the extracellular domain of mouse CD8α fused to the MMS and cytoplasmic tail of CD47 form 2 (Figure 4A). The following data indicate that neither CD47 IgV nor MMS domain alone was sufficient to mediate cell death signaling. Engagement of CD8MC2 chimera by immobilized anti-CD8 on either U937 or PMA-activated Jurkat transfectants or ligation of IAP/CD7 chimera by CD47 mAb on JinB8 transfectants did not trigger cell death (0%, n = 4 and 2%, n = 6, respectively). Thus, we conclude that CD47-induced cell death signaling requires both the IgV and MMS domains of the molecule, whereas the cytoplasmic tail is not required.

PS externalization in CD47-induced cell death is down-regulated by K+ efflux impairment and antimycin A treatment

Cell shrinkage, PS externalization, and drop in ΔΨm are major hallmarks of apoptosis. Cell volume loss has been demonstrated to result in potassium [K+] and sodium [Na+] efflux.38,48 Furthermore, K+ efflux and drop in ΔΨm are tightly coupled.49,50 To examine whether CD47-induced cell death was dependent on K+ efflux, we performed cell cultures in RPMI medium in which Na+/K+ concentrations had been inverted, resulting in K+ efflux impairment by osmotic forces.38 Under these conditions, CD47-induced PS externalization, together with cell shrinkage, were delayed following CD47 ligation in B-CLL cells and the U937 cell line when compared to that observed in cultures with medium reconstituted with a normal [Na+]/[K+] ratio (Figure5A and not shown). ΔΨm was also slightly decreased (V.M. and M.S., unpublished observations, May 2000).

Fig. 5.

Down-regulation of CD47-induced PS exposure by K+ efflux or antimycin A treatment: lack of effect of PKC inhibitor rottlerin.

Freshly isolated B-CLL cells and the U937 cell line were cultured with or without immobilized CD47 mAbs. (A) Cultures were performed in RPMI medium containing normal [Na+] or inverted [Na+]/[K+] ratios [K+]. Shown are percent of viable cells (annexin-V/PI). (B-C) Cells were cultured in the presence or absence of immobilized CD47 mAbs with or without antimycin A (30 μM) or increasing concentrations of rottlerin. Shown is the percent of dead cells (annexin-V+ or DiOC6low cells); one representative experiment of 4 is shown.

Fig. 5.

Down-regulation of CD47-induced PS exposure by K+ efflux or antimycin A treatment: lack of effect of PKC inhibitor rottlerin.

Freshly isolated B-CLL cells and the U937 cell line were cultured with or without immobilized CD47 mAbs. (A) Cultures were performed in RPMI medium containing normal [Na+] or inverted [Na+]/[K+] ratios [K+]. Shown are percent of viable cells (annexin-V/PI). (B-C) Cells were cultured in the presence or absence of immobilized CD47 mAbs with or without antimycin A (30 μM) or increasing concentrations of rottlerin. Shown is the percent of dead cells (annexin-V+ or DiOC6low cells); one representative experiment of 4 is shown.

Close modal

Classically, PS exposure is dependent on caspase activation, and PS externalization can be blocked by antimycin A, an inhibitor of the mitochondrial respiratory chain. As depicted in Figure 5B, antimycin A totally prevented PS exposure in CD47-treated cells, whereas it had no effect on decreased ΔΨm, strongly suggesting that these 2 events are uncoupled. Mechanisms involved in PS exposure are rather complex and not entirely understood. For example, in some models of apoptosis, long-term PS exposure has been shown to be the net result of increased scramblase (which has no selectivity for the direction of bilayer lipids movement) and decreased translocase activity (which selectively transports PS from the outer leaflet back to the inner leaflet). Protein kinase C (PKC)δ reportedly activated scramblase in apoptotic cells.51 We observed CD47-induced cell death in the presence of rottlerin, an inhibitor of new PKC family isoforms, notably of PKCδ (Figure 5C). These data exclude a role for PKCδ without ruling out the involvement of other PKC isoforms. Indeed, immobilized CD47 mAb activated PKCθ in T cells.27 

These results support the hypothesis that CD47-induced cell death involves cell volume dysregulation, which is partly associated with K+ efflux, and that PS exposure is intimately linked to cell shrinkage but may be uncoupled from decreased ΔΨm and is independent of PKCδ activation.

CD47-induced cell death involves cytoskeleton rearrangement

It was previously reported that T- and B-cell lines46,52 as well as B-CLL cells13 attach to and spread on CD47 mAb-coated surfaces, suggesting that CD47 ligation induces a change in cytoskeleton organization. Moreover, immobilized CD47 mAb reportedly induced F-actin polymerization in activated T cells.27 To assess the role of cytoskeleton in CD47-triggered cell death, we examined the effect of cytochalasin D, an inhibitor of actin polymerization. Treatment with cytochalasin D prevented both PS exposure and ΔΨm loss in CD47 mAb–treated U937 and B-CLL cells (Figure 6A). These data confirm and extend a previous report showing that cytochalasin D inhibited CD47-induced caspase-independent cell death in normal TCR-activated T cells.14 

Fig. 6.

Link between CD47-induced PS exposure and cytoskeleton rearrangement.

(A) Freshly isolated B-CLL cells or the U937 cell line were cultured overnight in the presence or absence of immobilized CD47 mAbs with or without cytochalasin D (Cyto D; 20 μM). Shown is percent of dead cells (annexin-V+ or DiOC6lowcells); one representative experiment of 4 is shown. (B-D) PBMCs were isolated from 4 WAS patients and cultured in the absence (Bi) or presence (Bii) of immobilized CD47 mAb. Annexin-V/PI and DiOC6 staining (patient no. 3) (B) and CD47 expression of PBMCs from patient no. 3 (B6H12) (C); percent viable cells (annexin-V/PI) in PBMCs of 4 WAS patients and 1 control donor (D).

Fig. 6.

Link between CD47-induced PS exposure and cytoskeleton rearrangement.

(A) Freshly isolated B-CLL cells or the U937 cell line were cultured overnight in the presence or absence of immobilized CD47 mAbs with or without cytochalasin D (Cyto D; 20 μM). Shown is percent of dead cells (annexin-V+ or DiOC6lowcells); one representative experiment of 4 is shown. (B-D) PBMCs were isolated from 4 WAS patients and cultured in the absence (Bi) or presence (Bii) of immobilized CD47 mAb. Annexin-V/PI and DiOC6 staining (patient no. 3) (B) and CD47 expression of PBMCs from patient no. 3 (B6H12) (C); percent viable cells (annexin-V/PI) in PBMCs of 4 WAS patients and 1 control donor (D).

Close modal

The motility of B-cell lines plated on CD47 mAb–coated surfaces requires the GTPase Cdc42.52 Since the Cdc42/WASP pathway is defective in cells from patients with WAS, we examined the effect of CD47 ligation in PBMCs isolated from WAS patients. We tested PBMCs of 4 characterized patients carrying WAS gene mutations, all leading to impaired WASP expression. As depicted in Figure 6B,D, we consistently failed to observe PS exposure and ΔΨm loss following CD47 ligation in WAS PBMCs, strongly suggesting a role for the Cdc42/WASP signaling pathway in CD47-induced cell death.

Taken together, these data indicate that cytoskeleton rearrangement is a key event in the triggering of CD47-mediated PS externalization and ΔΨm loss.

The present findings indicate that caspase-independent cell death induced by CD47 stimulation is sufficient to trigger a signal for phagocytosis in human DCs. The CD47-induced cell death involves PS exposure, disruption of mitochondrial function, and cytoskeleton rearrangement possibly linked to the Cdc42/WAS protein (WASP) signaling pathway. This observation challenges the classical concept that both caspase activation and PS exposure triggered by caspases are prerequisites for phagocytosis. Elimination of dead cells in the absence of caspase activation and nuclear degradation has been previously reported. For instance, constitutive but caspase-independent death of platelets, anucleated blood cells, leads to PS exposure and clearance by phagocytes.53 However, caspase-independent or -dependent PS exposure appears to be a critical event to allow engulfment of dead cells by phagocytic cells.54 A PS receptor was recently identified and cloned, and anti–PS receptor mAb inhibited elimination of apoptotic cells.55Blocking PS exposure without suppression of mitochondrial collapse and nuclear degradation prevented phagocytosis.56 But cells dying by delayed necrosis upon exposure to staurosporine and z-VAD-fmK were eliminated without PS externalization, underlying the potential role of other candidates for recognition and elimination of dead cells by APCs.57 

Caspases play a central role in the execution of cell death. These enzymes are recruited and activated by either receptors of the tumor necrosis factor receptor (TNFR) family or by apoptogenic molecules released from the intermembrane space of the mitochondria.58 Nevertheless, apart from CD47, stimulation of several surface antigens resulted in the activation of caspase-independent PS exposure in human cells. This includes CD2, MHC class I, and MHC class II.11,16,17 In contrast to CD47, ligation of these antigens may induce either caspase-dependent or -independent death according to the epitope triggered. So far, CD47 ligation by soluble, immobilized, or cross-linked mAbs recognizing at least 3 different epitopes exclusively induced caspase-independent cell death (Mateo et al,13 Pettersen et al,14 and data not shown).

During apoptosis, mitochondrial changes result in the dissipation of the mitochondrial transmembrane potential (ΔΨm). The loss of ΔΨm is generally mediated by opening of the mitochondrial permeability transition (PT) pores and as a consequence, the release of apoptogenic molecules such as AIF and cyt c59with subsequent caspase-independent or -dependent nuclear degradation. The engagement of CD47 results in cell shrinkage and ΔΨm loss, and this occurs in the absence of caspase activation and DNA degradation likely excluding the involvement of AIF and cyt c. Indeed, AIF release ultimately leads to caspase-independent DNA degradation60 and cyt c to caspase activation.61 However, inactivation of cyt c by at least HSP27 binding has been shown to prevent apoptosome formation and caspase activation.62 We therefore postulate that either cyt c is inactivated and translocation of AIF to the nucleus impaired or that there is no release of apoptogenic molecules during CD47 stimulation. Our unpublished observations indicate that inhibitors of permeability transition pore complex (PTPC) opening failed to prevent CD47-induced cell death. They include cyclosporin A in combination with aristolochic acid (which acts on cyclophilin D and PLA2, respectively), bongkrekic acid (inhibitor of adenine nucleotide transfer [ANT]), and Ca++chelators.59 In support of the latter hypothesis, B-cell receptor (BCR)–mediated apoptosis of immature B-cell line resulted in ΔΨm loss but did not induce cyt c release and caspase activation.63 Interestingly, cyt crelease may occur in the absence of ΔΨm loss by a direct effect of the proapoptotic molecule Bid/Bik.64 

We also provide evidence that CD47-induced PS exposure may be dissociated from ΔΨm loss but is intimately linked to cell shrinkage. Cell volume decrease during apoptosis is an active mechanism that was shown to be dependent on K+ channels (reviewed in Gomez-Angelats et al48 and Yu and Choi65). Maeno et al66 observed apoptotic volume decrease in the presence of zVAD-fmk, suggesting that K+ efflux is caspase independent. Furthermore, it was demonstrated that this phenomenon may occur upstream of caspase activation. In the present study, mitochondrial inhibitors such as antimycin A inhibited CD47-induced PS exposure but not dissipation of ΔΨm. Similar observations were made by Zhuang et al in ectoposide orN-tosyl-l-phenylalanine chloromethyl ketone (TPCK)–induced apoptosis of THP-1 monocyte cell line.56 Of interest, antimycin A alone induced ΔΨm in B-CLL and U937 cell lines. In that respect, antimycin A mimicked BH3 cell-death domain and induced mitochondrial swelling and ΔΨm loss.67 Taken together, these results strongly suggest that factors other than cyt c. released by the mitochondria and caspases may directly or indirectly affect PS exposure in CD47-induced cell death.

As described for CD47-mediated cell spreading and costimulation in T-cell lines, both extracellular and multispan transmembrane domains of CD47 molecule were required to induce killing. The short cytoplasmic domain was dispensable. In T cells, ∼ 65% of the CD47 molecule is localized in membrane rafts, where it regulates TCR-dependent and -independent T-cell activation. Immobilized CD47 mAb reportedly controlled the activation of heterotrimeric G proteins and triggered F-actin polymerization and PKCθ translocation.27However, CD47 mAb–induced killing was observed in the presence of G protein inhibitor (that is, pertussis toxin) (V.M. and M.S., data not shown) or new PKC family (δ, ε, η, θ, and μ) inhibitor (ie, rottlerin). PKCθ expression was low in B-cell lines, largely excluding its involvement in CD47-induced cell death.

Yoshida et al previously reported that CD47 regulated human B-cell motility through Cdc42.52 We observed that engagement of CD47 induced spreading of B-CLL cells. Cdc42 belongs to the small Rho GTPases family, which includes Rho and Rac, known to regulate formation of actin structures in many cell types.68 Among others, Cdc42 appears to have a unique role in actin remodeling during T-cell activation and endocytosis of immature DCs. WASP, uniquely expressed on hematopoietic cells,69 is the specific effector of Cdc42. WASP binds Cdc42 and controls actin polymerization by distinct domains.70 WAS-immunodeficiency syndrome is characterized by abnormalities in cytoskeletal function, resulting in abnormal chemotaxis, phagocytosis, and T-cell responses.69 

Two observations strongly suggest a direct role of cytoskeleton rearrangement in CD47-induced cell death: (1) inhibition of both PS exposure and ΔΨm loss by cytochalasin D, an inhibitor of actin polymerization, and (2) absence of CD47-induced death in PBMCs of WAS patients. As depicted in a schematic model in Figure7, we propose that CD47 ligation induces killing by 2 nonmutually exclusive pathways. The initial and common event would be the triggering of actin polymerization, perhaps via Cdc42/WASP pathway. This will lead to either mitochondrial changes including matrix swelling and ΔΨm loss, followed by PS exposure (pathway #1) and/or to a direct effect on PS externalization, bypassing mitochondria (pathway #2).

Fig. 7.

Hypothetical model for CD47-induced PS exposure.

Two nonmutually exclusive pathways leading to PS exposure and initiated by a common triggering event: Cdc42/WASP signaling pathway and F-actin polymerization. Inhibition by cytochalasin D (Ctyo D). Pathway no. 1 (thin arrow): loss in ΔΨm followed by PS externalization. Antimycin A inhibits PS but not ΔΨm loss. Pathway no. 2 (thick arrow): bypass of mitochondria and direct induction of PS externalization. Elevated extracellular K+ slows down PS exposure. Absence of caspase activation and nuclear degradation in CD47-induced cell death. Dotted arrows indicate hypothetical inhibitory pathways. ROS indicates reactive oxygen species; AIF, apoptosis-inducing factor; Cyt c, cytochrome c.

Fig. 7.

Hypothetical model for CD47-induced PS exposure.

Two nonmutually exclusive pathways leading to PS exposure and initiated by a common triggering event: Cdc42/WASP signaling pathway and F-actin polymerization. Inhibition by cytochalasin D (Ctyo D). Pathway no. 1 (thin arrow): loss in ΔΨm followed by PS externalization. Antimycin A inhibits PS but not ΔΨm loss. Pathway no. 2 (thick arrow): bypass of mitochondria and direct induction of PS externalization. Elevated extracellular K+ slows down PS exposure. Absence of caspase activation and nuclear degradation in CD47-induced cell death. Dotted arrows indicate hypothetical inhibitory pathways. ROS indicates reactive oxygen species; AIF, apoptosis-inducing factor; Cyt c, cytochrome c.

Close modal

When blood cell susceptibility was examined, immature DCs, activated T cells, and CD34+ precursors appeared to be much less sensitive and virtually resistant to CD47-induced killing. Similar differences in APC susceptibility have been reported in HLA-DR–induced caspase-independent cell death.71 Pettersen et al14 reported that CD47 mAb preferentially induces killing in normal activated but not resting T cells, and this was inhibited by cytochalasin D. We failed to induce cell death in anti-CD3–activated T cells. The use of different mAbs in the 2 studies and the regulation of CD47 conformation during T-cell activation (M.S. and V.M., unpublished data, January 2000) may provide one explanation for this discrepancy.

The role of CD47-induced killing in the regulation of immune response remains poorly understood. One may envision that it at least participates in the maintenance of homeostasis. Indeed, apoptosis is a self-destruction process which, in contrast to necrosis, does not lead to inflammatory response and may be involved in the induction of tolerance. Thrombospondin, the natural ligand of CD47, establishes a molecular bridge between apoptotic cells and APCs, facilitating phagocytosis of apoptotic cells.72 We reported that TSP, like CD47 mAb, induced caspase-independent cell death via CD47 binding but also acted on APCs to down-regulate the production of the proinflammatory molecule IL-12 and prevent DC maturation.23 It is important to mention that additional mechanisms inhibit inflammation during phagocytosis of apoptotic cells. They include induction of transforming growth factor (TGF)-β production by apoptotic cells themselves73 and by APCs upon their interaction with apoptotic cells.74 Also, TSP was found to be the main activator of TGFβ, and, reciprocally, TGFβ induces TSP production.75,76 

CD47 was recently reported to be a marker of self on murine cells that prevented clearance of intact red blood and lymphoid cells by CD47+ APCs through the engagement of its counterstructure SIRP-α, selectively expressed on APCs.77 Also, CD47/SIRP-α interactions negatively regulated APC functions.28 However, apoptotic cells (highly expressing CD4778) are known to be efficiently cleared by APCs, and this is not inhibited by CD47 mAb.23,79 Whether CD47 expressed on apoptotic cells has a particular conformation in order not to deliver a negative signal for phagocytosis to APCs via SIRP-α or, conversely, whether SIRP-α/CD47 interactions can induce cell death remain open questions.

Taken together, we propose that TSP-induced/enhanced PS exposure via CD47 on lymphoid cells, followed by their elimination by professional phagocytes, represent a process that continuously takes place in peripheral tissues to ensure the maintenance of tissue and host homeostasis. Additionally, the anti-inflammatory activity of TSP, combined with its ability to facilitate the clearance of apoptotic cells, may further contribute to homeostasis and induction of tolerance by avoiding inappropriate immune response to self-antigens.

Prepublished online as Blood First Edition Paper, June 14, 2002; DOI 10.1182/blood-2001-12-0217.

Supported by grants from Fondation Medic and the Leukemia Research Foundation of Canada.

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.

1
Rathmell
JC
Thompson
CB
The central effectors of cell death in the immune system.
Annu Rev Immunol.
17
1999
781
828
2
Vaux
DL
Korsmeyer
SJ
Cell death in development.
Cell.
96
1999
245
254
3
Scaffidi
C
Kirchhoff
S
Krammer
PH
Peter
ME
Apoptosis signaling in lymphocytes.
Curr Opin Immunol.
11
1999
277
285
4
Larsson
M
Fonteneau
JF
Bhardwaj
N
Dendritic cells resurrect antigens from dead cells.
Trends Immunol.
22
2001
141
148
5
Strasser
A
O'Connor
L
Dixit
VM
Apoptosis signaling.
Annu Rev Biochem.
69
2000
217
245
6
Kroemer
G
Dallaporta
B
Resche-Rigon
M
The mitochondrial death/life regulation in apoptosis and necrosis.
Annu Rev Physiol.
60
1998
619
642
7
Savill
J
Fadok
V
Corpse clearance defines the meaning of cell death.
Nature.
407
2000
784
788
8
Sarin
A
Wu
ML
Henkart
PA
Different interleukin-1 beta converting enzyme (ICE) family protease requirements for the apoptotic death of T lymphocytes triggered by diverse stimuli.
J Exp Med.
184
1996
2445
2450
9
Xiang
J
Chao
DT
Korsmeyer
SJ
BAX-induced cell death may not require interleukin 1 beta-converting enzyme-like proteases.
Proc Natl Acad Sci U S A.
93
1996
14559
14563
10
Kim
K
Lee
C
Sayers
TJ
Muegge
K
Durum
SK
The trophic action of IL-7 on pro-T cells: inhibition of apoptosis of pro-T1, -T2, -T3 cells correlates with Bcl-2 and Bax levels and is independent of Fas and p53 pathways.
J Immunol.
160
1998
5735
5741
11
Deas
O
Dumont
C
MacFarlane
M
et al
Caspase-independent cell death induced by anti-CD2 or staurosporine in activated human peripheral T lymphocytes.
J Immunol.
161
1998
3375
3383
12
Lesage
S
Steff
AM
Philippoussis
F
et al
CD4+ CD8+ thymocytes are preferentially induced to die following CD45 cross-linking, through a novel apoptotic pathway.
J Immunol.
159
1997
4762
4771
13
Mateo
V
Lagneaux
L
Bron
D
et al
CD47 ligation induces caspase-independent cell death in chronic lymphocytic leukemia.
Nat Med.
5
1999
1277
1284
14
Pettersen
RD
Hestdal
K
Olafsen
MK
Lie
SO
Lindberg
FP
CD47 signals T cell death.
J Immunol.
162
1999
7031
7040
15
Pettersen
RD
Bernard
G
Olafsen
MK
Pourtein
M
Lie
SO
Cd99 signals caspase-independent T cell death.
J Immunol.
166
2001
4931
4942
16
Skov
S
Klausen
P
Claesson
MH
Ligation of major histocompatability complex (MHC) class I molecules on human T cells induces cell death through PI-3 kinase-induced c-Jun NH2-terminal kinase activity: a novel apoptotic pathway distinct from Fas-induced apoptosis.
J Cell Biol.
139
1997
1523
1531
17
Drenou
B
Blancheteau
V
Burgess
DH
Fauchet
R
Charron
DJ
Mooney
NA
A caspase-independent pathway of MHC class II antigen-mediated apoptosis of human B lymphocytes.
J Immunol.
163
1999
4115
4124
18
Reinhold
MI
Lindberg
FP
Plas
D
Reynolds
S
Peters
MG
Brown
EJ
In vivo expression of alternatively spliced forms of integrin-associated protein (CD47).
J Cell Sci.
108
1995
3419
3425
19
Gao
AG
Lindberg
FP
Finn
MB
Blystone
SD
Brown
EJ
Frazier
WA
Integrin-associated protein is a receptor for the C-terminal domain of thrombospondin.
J Biol Chem.
271
1996
21
24
20
Vernon-Wilson
EF
Kee
WJ
Willis
AC
Barclay
AN
Simmons
DL
Brown
MH
CD47 is a ligand for rat macrophage membrane signal regulatory protein SIRP (OX41) and human SIRPalpha 1.
Eur J Immunol.
30
2000
2130
2137
21
Brown
EJ
Frazier
WA
Integrin-associated protein (CD47) and its ligands.
Trends Cell Biol.
11
2001
130
135
22
Armant
M
Avice
MN
Hermann
P
et al
CD47 ligation selectively downregulates human interleukin 12 production.
J Exp Med.
190
1999
1175
1182
23
Demeure
CE
Tanaka
H
Mateo
V
Rubio
M
Delespesse
G
Sarfati
M
CD47 engagement inhibits cytokine production and maturation of human dendritic cells.
J Immunol.
164
2000
2193
2199
24
Avice
MN
Rubio
M
Sergerie
M
Delespesse
G
Sarfati
M
CD47 ligation selectively inhibits the development of human naive T cells into Th1 effectors.
J Immunol.
165
2000
4624
4631
25
Avice
MN
Rubio
M
Sergerie
M
Delespesse
G
Sarfati
M
Role of cd47 in the induction of human naive T cell anergy.
J Immunol.
167
2001
2459
2468
26
Reinhold
MI
Lindberg
FP
Kersh
GJ
Allen
PM
Brown
EJ
Costimulation of T cell activation by integrin-associated protein (CD47) is an adhesion-dependent, CD28-independent signaling pathway.
J Exp Med.
185
1997
1
11
27
Rebres
RA
Green
JM
Reinhold
MI
Ticchioni
M
Brown
EJ
Membrane raft association of CD47 is necessary for actin polymerization and PKCθ translocation in T cells.
J Biol Chem.
15
2000
2
28
Latour
S
Tanaka
H
Demeure
C
et al
Bidirectional negative regulation of human T and dendritic cells by CD47 and its cognate receptor signal-regulator protein-alpha: down-regulation of IL-12 responsiveness and inhibition of dendritic cell activation.
J Immunol.
167
2001
2547
2554
29
Ticchioni
M
Deckert
M
Mary
F
Bernard
G
Brown
EJ
Bernard
A
Integrin-associated protein (CD47) is a comitogenic molecule on CD3-activated human T cells.
J Immunol.
158
1997
677
684
30
Waclavicek
M
Majdic
O
Stulnig
T
et al
T cell stimulation via CD47: agonistic and antagonistic effects of CD47 monoclonal antibody 1/1A4.
J Immunol.
159
1997
5345
5354
31
Green
JM
Zhelesnyak
A
Chung
J
Role of cholesterol in formation and function of a signaling complex involving alphaVbeta3, integrin-associated protein (CD47), and heterotrimeric G proteins.
J Cell Biol.
146
1999
673
682
32
Zhou
M
Brown
EJ
Leukocyte response integrin and integrin-associated protein act as a signal transduction unit in generation of a phagocyte respiratory burst.
J Exp Med.
178
1993
1165
1174
33
Susin
SA
Daugas
E
Ravagnan
L
et al
Two distinct pathways leading to nuclear apoptosis.
J Exp Med.
192
2000
571
580
34
Kawahara
A
Ohsawa
Y
Matsumura
H
Uchiyama
Y
Nagata
S
Caspase-independent cell killing by Fas-associated protein with death domain.
J Cell Biol.
143
1998
1353
1360
35
Holler
N
Zaru
R
Micheau
O
et al
Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule.
Nat Immunol.
1
2000
489
495
36
Dumont
C
Durrbach
A
Bidere
N
Caspase-independent commitment phase to apoptosis in activated blood T lymphocytes: reversibility at low apoptotic insult.
Blood.
96
2000
1030
1038
37
Blystone
SD
Lindberg
FP
Williams
MP
McHugh
KP
Brown
EJ
Inducible tyrosine phosphorylation of the beta3 integrin requires the alphaV integrin cytoplasmic tail.
J Biol Chem.
271
1996
31458
31462
38
Bortner
CD
Hughes
FM
Cidlowski
JA
A primary role for K+ and Na+ efflux in the activation of apoptosis.
J Biol Chem.
272
1997
32436
32442
39
Samali
A
Zhivotovsky
B
Jones
D
Nagata
S
Orrenius
S
Apoptosis: cell death defined by caspase activation.
Cell Death Differ.
6
1999
495
496
40
Krahling
S
Callahan
MK
Williamson
P
Schlegel
RA
Exposure of phosphatidylserine is a general feature in the phagocytosis of apoptotic lymphocytes by macrophages.
Cell Death Differ.
6
1999
183
189
41
Albert
ML
Pearce
SF
Francisco
LM
et al
Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes.
J Exp Med.
188
1998
1359
1368
42
Chandra
J
Gilbreath
J
Freireich
EJ
et al
Protease activation is required for glucocorticoid-induced apoptosis in chronic lymphocytic leukemic lymphocytes.
Blood.
90
1997
3673
3681
43
Holyoake
TL
Alcorn
MJ
CD34+ positive haemopoietic cells: biology and clinical applications.
Blood Rev.
8
1994
113
124
44
Messner
HA
Assessment and characterization of hemopoietic stem cells.
Stem Cells.
13(suppl 3)
1995
13
18
45
Lindberg
FP
Gresham
HD
Schwarz
E
Brown
EJ
Molecular cloning of integrin-associated protein: an immunoglobulin family member with multiple membrane-spanning domains implicated in alpha v beta 3-dependent ligand binding.
J Cell Biol.
123
1993
485
496
46
Reinhold
MI
Green
JM
Lindberg
FP
Ticchioni
M
Brown
EJ
Cell spreading distinguishes the mechanism of augmentation of T cell activation by integrin-associated protein/CD47 and CD28.
Int Immunol.
11
1999
707
718
47
Lindberg
FP
Gresham
HD
Reinhold
MI
Brown
EJ
Integrin-associated protein immunoglobulin domain is necessary for efficient vitronectin bead binding.
J Cell Biol.
134
1996
1313
1322
48
Gomez-Angelats
M
Bortner
CD
Cidlowski
JA
Cell volume regulation in immune cell apoptosis.
Cell Tissue Res.
301
2000
33
42
49
Dallaporta
B
Hirsch
T
Susin
SA
et al
Potassium leakage during the apoptotic degradation phase.
J Immunol.
160
1998
5605
5615
50
Bortner
CD
Cidlowski
JA
Caspase independent/dependent regulation of K(+), cell shrinkage, and mitochondrial membrane potential during lymphocyte apoptosis.
J Biol Chem.
274
1999
21953
21962
51
Frasch
SC
Henson
PM
Kailey
JM
et al
Regulation of phospholipid scramblase activity during apoptosis and cell activation by protein kinase Cdelta.
J Biol Chem.
275
2000
23065
23073
52
Yoshida
H
Tomiyama
Y
Ishikawa
J
et al
Integrin-associated protein/CD47 regulates motile activity in human B-cell lines through CDC42.
Blood.
96
2000
234
241
53
Brown
SB
Clarke
MCH
Magowan
L
Sanderson
H
Savill
J
Constitutive death of platelets leading to scavenger receptor-mediated phagocytosis: a caspase-independent cell clearance program.
J Biol Chem.
275
2000
5987
5996
54
Fadok
VA
de Cathelineau
A
Daleke
DL
Henson
PM
Bratton
DL
Loss of phospholipid asymmetry and surface exposure of phosphatidylserine is required for phagocytosis of apoptotic cells by macrophages and fibroblasts.
J Biol Chem.
276
2001
1071
1077
55
Fadok
VA
Bratton
DL
Rose
DM
Pearson
A
Ezekewitz
RA
Henson
PM
A receptor for phosphatidylserine-specific clearance of apoptotic cells.
Nature.
405
2000
85
90
56
Zhuang
J
Ren
Y
Snowden
RT
et al
Dissociation of phagocyte recognition of cells undergoing apoptosis from other features of the apoptotic program.
J Biol Chem.
273
1998
15628
15632
57
Hirt
UA
Gantner
F
Leist
M
Phagocytosis of nonapoptotic cells dying by caspase-independent mechanisms.
J Immunol.
164
2000
6520
6529
58
Thornberry
NA
Lazebnik
Y
Caspases: enemies within.
Science.
281
1998
1312
1316
59
Kroemer
G
Reed
JC
Mitochondrial control of cell death.
Nat Med.
6
2000
513
519
60
Daugas
E
Susin
SA
Zamzami
N
et al
Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis.
FASEB J.
14
2000
729
739
61
Reed
JC
Cytochrome c: can't live with it—can't live without it.
Cell.
91
1997
559
562
62
Bruey
J-M
Ducasset
C
Bonniaud
P
et al
Hsp27 negatively regulates cell death by interacting with cytochrome c.
Nat Cell Biol.
2
2000
645
652
63
Katz
E
Deehan
MR
Seatter
S
Lord
C
Sturrock
RD
Harnett
MM
B cell receptor-stimulated mitochondrial phospholipase A2 activation and resultant disruption of mitochondrial membrane potential correlate with the induction of apoptosis in WEHI-231 B cells.
J Immunol.
166
2001
137
147
64
Shimizu
S
Tsujimoto
Y
Proapoptotic BH3-only Bcl-2 family members induce cytochrome c release, but not mitochondrial membrane potential loss, and do not directly modulate voltage-dependent anion channel activity.
Proc Natl Acad Sci U S A.
97
2000
577
582
65
Yu
SP
Choi
DW
Ions, cell volume, and apoptosis.
Proc Natl Acad Sci U S A.
97
2000
9360
9362
66
Maeno
E
Ishizaki
Y
Kanaseki
T
Hazama
A
Okada
Y
Normotonic cell shrinkage because of disordered volume regulation is an early prerequisite to apoptosis.
Proc Natl Acad Sci U S A.
97
2000
9487
9492
67
Tzung
SP
Kim
KM
Basanez
G
et al
Antimycin A mimics a cell-death-inducing Bcl-2 homology domain 3.
Nat Cell Biol.
3
2001
183
191
68
Erickson
JW
Cerione
RA
Multiple roles for Cdc42 in cell regulation.
Curr Opin Cell Biol.
13
2001
153
157
69
Thrasher
AJ
Burns
S
Lorenzi
R
Jones
GE
The Wiskott-Aldrich syndrome: disordered actin dynamics in haematopoietic cells.
Immunol Rev.
178
2000
118
128
70
Machesky
LM
Insall
RH
Scar1 and the related Wiskott-Aldrich syndrome protein, WASP, regulate the actin cytoskeleton through the Arp2/3 complex.
Curr Biol.
8
1998
1347
1356
71
Bertho
N
Drenou
B
Laupeze
B
et al
HLA-DR-mediated apoptosis susceptibility discriminates differentiation stages of dendritic/monocytic APC.
J Immunol.
164
2000
2379
2385
72
Savill
J
Recognition and phagocytosis of cells undergoing apoptosis.
Br Med Bull.
53
1997
491
508
73
Chen
W
Frank
ME
Jin
W
Wahl
SM
TGF-beta released by apoptotic T cells contributes to an immunosuppressive milieu.
Immunity.
14
2001
715
725
74
Henson
PM
Bratton
DL
Fadok
V
The phosphatidylserine receptor: a crucial molecular switch?
Nat Rev Mol Cell Biol.
2
2001
627
633
75
Crawford
SE
Stellmach
V
Murphy-Ullrich
JE
Hynes
RO
Boivin
GP
Bouck
N
Thrombospondin-1 is a major activator of TGF-β1 in vivo.
Cell.
93
1998
1159
1170
76
Masli
S
Turpie
B
Hecker
KH
Streilein
JW
Expression of thrombospondin in TGFβ-treated APCs and its relevance to their immune deviation-promoting properties.
J Immunol.
168
2002
2264
2273
77
Oldenborg
PA
Zheleznyak
A
Fang
YF
Lagenaur
CF
Gresham
HD
Lindberg
FP
Role of CD47 as a marker of self on red blood cells.
Science.
288
2000
2051
2054
78
Guo
K
Wolf
V
Dharmarajan
AM
et al
Apoptosis-associated gene expression in the corpus luteum of the rat.
Biol Reprod.
58
1998
739
746
79
Savill
J
Dransfield
I
Hogg
N
Haslett
C
Vitronectin receptor-mediated phagocytosis of cells undergoing apoptosis.
Nature.
343
1990
170
173

Author notes

Marika Sarfati, Centre de Recherche du CHUM, Hôpital Notre-Dame, Laboratoire d'Immunorégulation (M4211-K), 1560 Sherbrooke St East, Montréal, QC H2L 4M1, Canada; e-mail: m.sarfati@umontreal.ca.

Sign in via your Institution