The human herpes virus 8 (HHV8)–encoded viral FLICE (Fas-associating protein with death domain–like interleukin-1–converting enzyme) inhibitory protein (vFLIP) is believed to protect cells against death receptor–mediated apoptosis. In the present study we demonstrate that expression of HHV8 vFLIP in a growth factor–dependent TF-1 leukemia cell line protects against growth factor withdrawal–induced apoptosis. Unlike vector-expressing cells, those expressing HHV8 vFLIP maintain their mitochondrial membrane potential upon withdrawal from growth factor and also exhibit a block in the activation of caspases. The protective effect of HHV8 vFLIP is associated with its ability to activate the nuclear factor–κ B (NF-κB) pathway and is missing in the vFLIP encoded by equine herpes virus 2 that lacks this activity. Inhibition of the NF-κB pathway by IκB superrepressor, lactacystin, MG132, arsenic trioxide, and phenylarsine oxide reverse the protection against growth factor withdrawal–induced apoptosis conferred by HHV8 vFLIP. HHV8 vFLIP up-regulates the expression of Bcl-xL, an antiapoptotic member of the Bcl2 family, which is a known target of the NF-κB pathway. Collectively, the above results suggest that HHV8 vFLIP–induced NF-κB activation may contribute to cellular transformation seen in association with HHV8 infection by preventing the apoptosis of cells destined to die because of growth factor deprivation.

Caspase-8 (FLICE [Fas-associating protein with death domain–like interleukin-1 (IL-1)–converting enzyme]/MACH or Mch5) is one of the apical caspases of the extrinsic (death receptor) apoptosis pathway and possesses a prodomain containing 2 homologous death effector domains (DEDs).1-3 In addition to its role in maintaining caspase-8 in its zymogen state, the prodomain of caspase-8 also helps in its recruitment to the aggregated complex of death receptors, which results in its activation via cross-proteolysis. Several viruses also encode proteins containing 2 DEDs, called viral FLICE inhibitory proteins (vFLIPs), which have been postulated to protect against death receptor–induced cell death by blocking the recruitment or activation of caspase-8.4-6 These vFLIPs include the orf-K13 from the human herpes virus 8/Kaposi sarcoma–associated herpes virus (HHV8/KSHV), MC159L, and MC160L from the molluscum contagiosum virus (MCV) and E8 from the equine herpes virus 2 (EHV2).4-6 We have previously demonstrated that the HHV8 vFLIP possesses the ability to activate the nuclear factor–κ B (NF-κB) pathway, a property that is not shared by other vFLIPs.7,8 In this report we describe the ability of HHV8 vFLIP to protect against growth factor withdrawal–induced apoptosis and link this property to its ability to activate the NF-κB pathway. These results have important implications for the pathogenesis of HHV8-associated malignancies and suggest that vFLIPs have functions beyond their known role as inhibitors of caspase-8.

Cell line and culture

TF-1 cell line (human acute myeloid leukemia) was obtained from the American Type Culture Collection (Manassas, VA) and maintained in RPMI medium supplemented with 10% fetal calf serum and 2 ng/mL granulocyte-macrophage colony-stimulating factor (GM-CSF).

Drugs and chemicals

Recombinant human GM-CSF and tumor necrosis factor α (TNFα) were purchased from R&D systems (Minneapolis, MN). Hoechst 33342, tetramethylrhodamine ethyl ester (TMRE), propidium iodide (PI), phenylarsine oxide (PAO), and arsenic trioxide (As203) were purchased from Sigma (St Louis, MO). MG132 and lactacystin were purchased from Calbiochem (San Diego, CA) and Biomol (Plymouth, PA), respectively.

Retrovirus and adenovirus constructs

Retrovirus constructs containing C-terminal Flag epitope–tagged HHV8 vFLIP (K13-Flag) and EHV2 vFLIP (E8-Flag) were generated in murine stem cell virus (MSCV) neo-based retroviral vector and amphotropic viruses generated and used for infection as described previously.8 Cells were selected in the presence of 1 mg/mL of G418 (Invitrogen, Carlsbad, CA). Adenoviral vectors encoding β-galactosidase and IκB superrepressor (DN-IκBα) were kindly provided by Dr Richard Gaynor of our institution.

Cell viability assay

For the measurement of cell viability, cells from exponentially growing cultures were washed twice with GM-CSF–free medium and plated in a flat-bottom 96-well plate at a density of 20 000 cells/well in the absence and presence of GM-CSF (2 ng/mL). Cell viability was measured after 48 hours using the MTS reagent (3-4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt) and following the instructions of the manufacturer (Promega, Madison, WI). Absorbance of viable cells was measured at 490 nm with 600 nm as a reference wavelength. Percent cell survival was calculated based on the reading of cells grown in the presence of GM-CSF as 100%.

Western blot analysis

Cells were lysed in a lysis buffer containing 20 mM sodium phosphate (pH 7.4), 150 mM NaCl, 0.1% Triton X-100, and 10% glycerol supplemented with a protease inhibitor cocktail tablet (Roche Molecular Biochemicals, Indianapolis, IN). Western blot analysis was performed essentially as described previously.8Quantitative analysis of the Western blots was carried out using the National Institutes of Health (NIH; Bethesda, MD) Image (version 1.61) software package. Primary antibody dilutions used in these experiments were Flag (Santa Cruz Biotechnology, Santa Cruz, CA; sc-807, 1:5000); cleaved caspase-9 (Cell Signaling, Beverly, MA; Asp 330 1:2000); cleaved caspase-3 (Cell Signaling; Asp 175, 1:2000); caspase-8 (Cell Signaling; 1C12, 1:2000); caspase-6 (Cell Signaling; 9762, 1:2000); actin (Santa Cruz Biotechnology; sc-1616, 1:1000); α-IκBα (Santa Cruz Biotechnology; SC-371, 1:2000) and p-IκBα (Cell Signaling; 9241S, 1:1000); Mcl-1 (Santa Cruz Biotechnology; S-19, 1:1000); and Bcl-x L/S (Santa Cruz Biotechnology; S-18, 1:1000).

Generation and characterization of TF-1 cells expressing vFLIPs

In order to study the effect of HHV8 vFLIP on growth factor withdrawal–induced apoptosis we used retroviral-mediated gene transfer to generate mass population of TF-1 cells with stable expression of Flag epitope–tagged HHV8 vFLIP (K13-Flag) or an empty vector. We selected TF-1 cell line for this study as this CD34+leukemia cell line is known to be responsive to multiple cytokines such as interleukin-3 (IL-3), erythropoietin, stem cell factor, and GM-CSF.9In addition to HHV8 vFLIP, we also generated TF-1 cells expressing E8-Flag to serve as a control. Expression of the transduced proteins was confirmed by Western blot analysis on the immunoprecipitated proteins (Figure1A).

Fig. 1.

Expression of HHV8 vFLIP protects against growth factor withdrawal–induced apoptosis in TF-1 cells.

(A) TF-1 cells were transduced with an empty retroviral vector or vectors expressing Flag epitope–tagged HHV8 vFLIP K13 and EHV2 vFLIP E8. Expression of the transduced proteins was confirmed by immunoprecipitation with Flag antibody beads (Sigma) followed by Western blot analysis with a rabbit polyclonal antibody against the Flag epitope tag. (B) HHV8 vFLIP K13 protects TF-1 cells against growth factor withdrawal–induced cell death. TF-1 cells expressing an empty vector, vFLIP K13, and vFLIP E8 were deprived of GM-CSF for 48 hours and cell viability measured as described in “Materials and methods.” The values shown are means ± SD of 3 independent experiments performed in triplicate. (C) Photomicrograph demonstrating the protective effect of vFLIP K13 against GM-CSF withdrawal–induced apoptosis. Cells were examined under a phase-contrast microscope and photographed. Original magnification, × 200. (D) Hoechst 33342 staining demonstrating the nuclear morphology of the indicated TF-1 cells grown in the absence and presence of GM-CSF. TF-1 vector and vFLIP-E8 cells demonstrate the classical nuclear features of apoptosis, such as nuclear condensation and fragmentation, which are significantly absent in vFLIP K13–expressing cells. Original magnification, × 200. (E) DNA content analysis demonstrating the appearance of a significant population of cells with sub-G0/G1content among TF-1 vector cells when deprived of GM-CSF for 48 hours that is markedly reduced in vFLIP K13–expressing cells. Cell pellets were fixed in 70% ethanol, and incubated at 4°C overnight. For staining, the cell pellets were resuspended in 0.5 mL of 0.05 mg/mL PI plus 0.2 mg/mL RNase A and incubated at 37°C for 30 minutes. Cell cycle distribution was analyzed using a flow cytometer.

Fig. 1.

Expression of HHV8 vFLIP protects against growth factor withdrawal–induced apoptosis in TF-1 cells.

(A) TF-1 cells were transduced with an empty retroviral vector or vectors expressing Flag epitope–tagged HHV8 vFLIP K13 and EHV2 vFLIP E8. Expression of the transduced proteins was confirmed by immunoprecipitation with Flag antibody beads (Sigma) followed by Western blot analysis with a rabbit polyclonal antibody against the Flag epitope tag. (B) HHV8 vFLIP K13 protects TF-1 cells against growth factor withdrawal–induced cell death. TF-1 cells expressing an empty vector, vFLIP K13, and vFLIP E8 were deprived of GM-CSF for 48 hours and cell viability measured as described in “Materials and methods.” The values shown are means ± SD of 3 independent experiments performed in triplicate. (C) Photomicrograph demonstrating the protective effect of vFLIP K13 against GM-CSF withdrawal–induced apoptosis. Cells were examined under a phase-contrast microscope and photographed. Original magnification, × 200. (D) Hoechst 33342 staining demonstrating the nuclear morphology of the indicated TF-1 cells grown in the absence and presence of GM-CSF. TF-1 vector and vFLIP-E8 cells demonstrate the classical nuclear features of apoptosis, such as nuclear condensation and fragmentation, which are significantly absent in vFLIP K13–expressing cells. Original magnification, × 200. (E) DNA content analysis demonstrating the appearance of a significant population of cells with sub-G0/G1content among TF-1 vector cells when deprived of GM-CSF for 48 hours that is markedly reduced in vFLIP K13–expressing cells. Cell pellets were fixed in 70% ethanol, and incubated at 4°C overnight. For staining, the cell pellets were resuspended in 0.5 mL of 0.05 mg/mL PI plus 0.2 mg/mL RNase A and incubated at 37°C for 30 minutes. Cell cycle distribution was analyzed using a flow cytometer.

Close modal

HHV8-encoded vFLIP K13 protects against growth factor withdrawal–induced apoptosis

We next studied the effect of the vFLIPs on GM-CSF withdrawal–induced cell death of TF-1 cells. TF-1 cells expressing the empty vector demonstrated rapid loss of viability when deprived of growth factors and demonstrated massive fragmentation into small apoptotic bodies (Figure 1B-C). In contrast, cells expressing the HHV8 vFLIP K13 demonstrated significant protection against growth factor withdrawal–induced apoptosis (Figure 1B-C). However, no protective effect was observed in cells expressing vFLIP E8, suggesting that this effect was unique to vFLIP K13 (Figure 1B-C).

Protective effect of HHV8 vFLIP on growth factor withdrawal–induced cell death was further characterized by nuclear staining with Hoechst 33342 dye. TF-1 cells expressing the control vector and vFLIP E8 demonstrated the classical nuclear features of apoptosis upon withdrawal from GM-CSF, such as nuclear condensation and fragmentation, which were markedly reduced in vFLIP K13–expressing cells (Figure 1D). Moreover, a DNA-content analysis revealed a significant decrease in the cells with sub-G0/G1 DNA content among vFLIP K13–expressing TF-1 cells compared with those expressing empty vector (Figure 1E). Collectively, the above results suggested that expression of the HHV8 vFLIP protected against growth factor withdrawal–induced cell death by blocking key nuclear and cytoplasmic features of apoptosis.

Previous studies have demonstrated the ability of vFLIPs to protect against apoptosis induced by death domain receptors belonging to the tumor necrosis factor receptor family.4-6Therefore, we were interested in checking whether expression of HHV8 vFLIP would also lead to protection against TNFα-induced apoptosis. However, as shown in Figure 2A, we failed to observe a significant difference in cell death induced by treatment with TNFα in the vector and vFLIP K13–expressing TF-1 cells. Similarly, expression of vFLIP K13 failed to protect against anticancer drug–induced apoptosis (Figure 2B). Taken together, the above results suggest that the ability of HHV8 vFLIP to protect against growth factor withdrawal–induced apoptosis does not extend to all forms of cell death.

Fig. 2.

vFLIP K13 fails to protect against TNFα- and anticancer drug–induced apoptosis.

(A) TF-1 cells expressing an empty vector or vFLIP K13 were treated with TNFα (10 ng/mL) for 48 hours and cell viability examined using the MTS assay. The values shown are means ± SD of 2 independent experiments performed in triplicate. (B) TF-1 cells expressing an empty vector or vFLIP K13 were treated with the indicated doses of drugs for 48 hours and cell viability examined using the MTS assay. The values shown are means ± SD of 2 independent experiments performed in triplicate.

Fig. 2.

vFLIP K13 fails to protect against TNFα- and anticancer drug–induced apoptosis.

(A) TF-1 cells expressing an empty vector or vFLIP K13 were treated with TNFα (10 ng/mL) for 48 hours and cell viability examined using the MTS assay. The values shown are means ± SD of 2 independent experiments performed in triplicate. (B) TF-1 cells expressing an empty vector or vFLIP K13 were treated with the indicated doses of drugs for 48 hours and cell viability examined using the MTS assay. The values shown are means ± SD of 2 independent experiments performed in triplicate.

Close modal

HHV8 vFLIP blocks loss of mitochondrial membrane potential and resultant caspase activation during growth factor withdrawal–induced apoptosis

Growth factor withdrawal–induced apoptosis is generally believed to be triggered by the loss of mitochondrial membrane potential with the resultant release of cytochrome c, which forms a multiprotein apoptosome complex with Apaf-1 and procaspase-9 in the presence of dATP.10-12 Procaspase-9 is activated upon recruitment to this complex and in turn activates the effector caspases, such as caspase-3. In order to understand the mechanism of protective effect of vFLIP K13 against growth factor withdrawal–induced apoptosis, we examined the status of mitochondrial membrane potential after staining with potentiometric dye TMRE.13 GM-CSF deprivation for 24 hours led to the appearance of a significant subpopulation of TF-1 vector cells with loss of TMRE staining, which was absent in the case of vFLIP K13–expressing cells (Figure3A). We also examined the status of caspase-9 and -3 in the vector- and vFLIP K13–expressing TF-1 cells upon withdrawal from GM-CSF for various time intervals using antibodies directed against the cleaved form of the above caspases. As shown in Figure 3B, significantly higher amounts of cleaved caspase-9 and -3 were observed in TF-1 vector cells compared with those expressing vFLIP K13, and this difference was most pronounced at 27 hours after withdrawal from GM-CSF. We also observed a significant difference in the cleavage of caspase-8 between TF-1 vector– and vFLIP K13–expressing cells, which was again most pronounced approximately 27 hours after GM-CSF withdrawal. Finally, we examined the status of caspase-6 in vector- and vFLIP K13–expressing cells upon withdrawal from GM-CSF. As shown in Figure 3C, we observed a decrease in the level of expression of caspase-6 in vector-expressing cells 27 hours after withdrawal from GM-CSF, which was not present in the vFLIP K13–expressing cells. Collectively, the above results suggest that vFLIP K13 protects against growth factor withdrawal–induced apoptosis by blocking the activation of caspases belonging to both the death receptors (extrinsic) and mitochondrial (intrinsic) apoptosis pathway.14,15 However, in light of the recent studies that suggest significant cross-talk among the 2 pathways of caspase activation,16-20 we cannot rule out the possibility that vFLIP K13 primarily blocks one of these pathways. For example, it was recently demonstrated that caspase-8 can be activated by caspase-3 and -6 and serves as an executioner caspase in the mitochondrial pathway during drug-induced apoptosis.16,18 Thus, it is conceivable that the observed inhibition of caspase-8 processing in vFLIP K13–expressing cells is due to the inhibition of the mitochondrial pathway of caspase activation.

Fig. 3.

vFLIP K13 blocks growth factor withdrawal–induced apoptosis by blocking loss of mitochondrial membrane potential and caspase-activation.

(A) vFLIP K13–expressing cells maintain their mitochondrial potential in the absence of GM-CSF. TF-1 vector– and vFLIP-K13–expressing cells were grown in the absence or presence of GM-CSF for 24 hours. Cells were stained with 100 nM TMRE for 30 minutes at 37°C and fluorescence determined using a flow cytometer. (B) Inhibition of caspase-9, -3, and -8 cleavage during GM-CSF withdrawal–induced apoptosis in TF-1 vFLIP K13 cells. TF-1 vector and vFLIP K13 cells were grown in the presence and absence of GM-CSF for the indicated time periods. Cell lysates were analyzed for cleavage of caspase-9, -3, and -8 by Western blot analysis using antibodies that can recognize their cleaved forms. The blot was reprobed with a polyclonal antibody against actin to show equal loading of all lanes. (C) Inhibition of caspase-6 cleavage during GM-CSF withdrawal–induced apoptosis in TF-1 vFLIP K13 cells. The experiment was performed essentially as described for panel B, except expression of caspase-6 was analyzed using an antibody that recognizes its full-length form.

Fig. 3.

vFLIP K13 blocks growth factor withdrawal–induced apoptosis by blocking loss of mitochondrial membrane potential and caspase-activation.

(A) vFLIP K13–expressing cells maintain their mitochondrial potential in the absence of GM-CSF. TF-1 vector– and vFLIP-K13–expressing cells were grown in the absence or presence of GM-CSF for 24 hours. Cells were stained with 100 nM TMRE for 30 minutes at 37°C and fluorescence determined using a flow cytometer. (B) Inhibition of caspase-9, -3, and -8 cleavage during GM-CSF withdrawal–induced apoptosis in TF-1 vFLIP K13 cells. TF-1 vector and vFLIP K13 cells were grown in the presence and absence of GM-CSF for the indicated time periods. Cell lysates were analyzed for cleavage of caspase-9, -3, and -8 by Western blot analysis using antibodies that can recognize their cleaved forms. The blot was reprobed with a polyclonal antibody against actin to show equal loading of all lanes. (C) Inhibition of caspase-6 cleavage during GM-CSF withdrawal–induced apoptosis in TF-1 vFLIP K13 cells. The experiment was performed essentially as described for panel B, except expression of caspase-6 was analyzed using an antibody that recognizes its full-length form.

Close modal

HHV8 vFLIP activates the NF-κB pathway in TF-1 cells

We and others have previously demonstrated constitutive NF-κB activation in HHV8-infected primary effusion lymphoma (PEL) cell lines.8,21 We have also demonstrated that HHV8-encoded K13 vFLIP can activate the NF-κB pathway while the EHV-encoded E8 vFLIP lacks this property.7 We were therefore interested in checking whether the differential ability of these vFLIPs to protect against growth factor withdrawal–induced apoptosis could be attributed to their differential ability to activate the NF-κB pathway in TF-1 cells. As shown in Figure 4A, an electrophoretic mobility shift assay demonstrated significant NF-κB binding activity in TF-1 cells expressing vFLIP K13 compared with those expressing an empty vector or vFLIP E8. NF-κB is usually present in the cytoplasm of cells in association with a family of inhibitory proteins, called IκB.22,23 Cytokine-inducible phosphorylation of the IκB proteins leads to their rapid ubiquitination and proteasome-mediated degradation, which releases NF-κB from its inhibitory influence.22,23 Consistent with the increased NF-κB binding activity in vFLIP K13–expressing TF-1 cells, we also observed an increase in the phosphorylated and a decrease in the total IκBα protein in these cells compared with those expressing an empty vector (Figure 4B).

Fig. 4.

HHV8 vFLIP activates the NF-κB pathway in TF-1 cells.

(A) Electrophoretic mobility shift assay demonstrating persistent NF-κB activation in TF-1 cells expressing vFLIP K13. The positions of the induced NF-κB complexes are marked with arrows while an asterisk (*) marks the position of a constitutive NF-κB complex. (B) Western blot analysis demonstrating increased phosphorylation (top panel) and decrease in the total IκBα protein (bottom panel) in TF-1 cells expressing vFLIP K13.

Fig. 4.

HHV8 vFLIP activates the NF-κB pathway in TF-1 cells.

(A) Electrophoretic mobility shift assay demonstrating persistent NF-κB activation in TF-1 cells expressing vFLIP K13. The positions of the induced NF-κB complexes are marked with arrows while an asterisk (*) marks the position of a constitutive NF-κB complex. (B) Western blot analysis demonstrating increased phosphorylation (top panel) and decrease in the total IκBα protein (bottom panel) in TF-1 cells expressing vFLIP K13.

Close modal

HHV8 vFLIP protects against growth factor withdrawal–induced apoptosis via the NF-κB pathway

In order to test the hypothesis that vFLIP K13 protects TF-1 cells against growth factor withdrawal–induced apoptosis via the activation of the NF-κB pathway, we took advantage of known inhibitors of this pathway. As shown in Figure 5A, infection of TF-1 vector– and vFLIP K13–expressing cells with an adenovirus encoding a phosphorylation-resistant dominant-negative form of IκBα (IκBα superrepressor), which is known to block NF-κB activation via diverse stimuli,24,25 led to an almost complete reversal of the protective effect afforded by vFLIP-K13. Similarly treatment of vector and vFLIP K13 cells with lactacystin and MG132, 2 proteasome inhibitors that block NF-κB activation by blocking the degradation of IκB,26 led to reversal of protective effect afforded by the expression of vFLIP in a dose-dependent fashion (Figure 5B-C). Essentially similar results were obtained upon treatment of TF-1 vector and vFLIP K13 cells with PAO and As2O3, 2 recently identified inhibitors of the NF-κB pathway (Figure 5D-E).27 

Fig. 5.

Inhibitors of the NF-κB pathway reverse the protective effect of vFLIP K13 against growth factor withdrawal–induced apoptosis.

(A) TF-1 vector and vFLIP K13 cells were infected with adenoviral vectors expressing β-galactosidase (control) or a superrepressor form of IκBα(DN-IκBα) at a multiplicity of infection of 1000. Approximately 48 hours after infection, cells were grown in the absence or presence of GM-CSF for another 48 hours and cell viability measured using the MTS assay. The values shown are means ± SD of 2 independent experiments performed in triplicate. (B-E) TF-1 vector and vFLIP K13 cells were grown without GM-CSF and in the presence of indicated doses of drugs for 48 hours. Cell viability was measured using the MTS assay and percent cell survival calculated using the viability of cells grown in the presence of GM-CSF as 100%. The values shown are means ± SD of a representative of 2 independent experiments performed in triplicate.

Fig. 5.

Inhibitors of the NF-κB pathway reverse the protective effect of vFLIP K13 against growth factor withdrawal–induced apoptosis.

(A) TF-1 vector and vFLIP K13 cells were infected with adenoviral vectors expressing β-galactosidase (control) or a superrepressor form of IκBα(DN-IκBα) at a multiplicity of infection of 1000. Approximately 48 hours after infection, cells were grown in the absence or presence of GM-CSF for another 48 hours and cell viability measured using the MTS assay. The values shown are means ± SD of 2 independent experiments performed in triplicate. (B-E) TF-1 vector and vFLIP K13 cells were grown without GM-CSF and in the presence of indicated doses of drugs for 48 hours. Cell viability was measured using the MTS assay and percent cell survival calculated using the viability of cells grown in the presence of GM-CSF as 100%. The values shown are means ± SD of a representative of 2 independent experiments performed in triplicate.

Close modal

HHV8 vFLIP maintains the expression of several antiapoptotic proteins during growth factor withdrawal–induced apoptosis

In order to understand the mechanism by which vFLIP K13–induced NF-κB activation might protect TF-1 cells against growth factor withdrawal–induced apoptosis we examined the expression of Bcl-xL, and Mcl-1 proteins, which are known to be up-regulated by the NF-κB pathway.28-31 The expression of the above proteins was normalized relative to the expression of actin to ensure equal loading of all lanes. As shown in Figure6, TF-1 cells expressing vFLIP K13 demonstrated a significant increase in the level of Bcl-xLprotein compared with the vector-expressing cells, and this difference was maintained up to 48 hours after withdrawal from GM-CSF. We also observed a difference in the level of expression of Mcl-1 protein between vector- and K13-expressing cells grown in the presence of GM-CSF, which was maintained up to 6 hours after withdrawal from GM-CSF. However, this difference had considerably narrowed at the 24 hour time point and the expression of the 2 proteins was not significantly different at 48 hours after GM-CSF withdrawal (Figure 6). Collectively, the above results suggest the involvement of Bcl-xL in the protective effect of vFLIP K13 against growth factor withdrawal–induced apoptosis. However, the role of additional antiapoptotic proteins, including Mcl-1, in this process cannot be ruled out and deserves further study.

Fig. 6.

Expression of Bcl-xL and Mcl-1 in vFLIP K13–expressing cells.

TF-1 vector and vFLIP K13 cells were grown in the presence and absence of GM-CSF for the indicated time periods. Cell lysates were analyzed for the presence of Bcl-xL, Mcl-1, and actin by Western blot analysis. Quantitative analysis of the blot was carried out using the NIH Image program. The numbers below the Bcl-xL and Mcl-1 blots represent the relative intensity of the protein bands after normalization with the actin control.

Fig. 6.

Expression of Bcl-xL and Mcl-1 in vFLIP K13–expressing cells.

TF-1 vector and vFLIP K13 cells were grown in the presence and absence of GM-CSF for the indicated time periods. Cell lysates were analyzed for the presence of Bcl-xL, Mcl-1, and actin by Western blot analysis. Quantitative analysis of the blot was carried out using the NIH Image program. The numbers below the Bcl-xL and Mcl-1 blots represent the relative intensity of the protein bands after normalization with the actin control.

Close modal

In addition to Kaposi sarcoma (KS), HHV8 genomes have been frequently detected in PELs, multicentric Castleman disease, angioimmunoblastic lymphadenopathy, and some cases of reactive lymphadenopathies.32-35 However, the exact role played by HHV8 in the pathogenesis of these disorders is still unclear. Although HHV8 is known to encode for homologs of several cytokines and their receptors, none of them is expressed in latently infected PEL cell lines or KS spindle cells.36 HHV8 vFLIP is one of the few viral proteins that is expressed in latently infected KS spindle and PEL cells, making it a prime candidate for the dysregulated cellular proliferation and transformation associated with infection by this virus.36-39 

Somatic cells in multicellular organisms are dependent on a continuous supply of survival signal from the neighboring cells, which limits the proliferative autonomy of any single cell.40,41 These survival signals are usually provided in the form of growth and survival factors within discrete somatic environments, thereby effectively trapping the somatic cells in specialized trophic microenvironments and acting as a major defense against metastatic spread.40,41 In the present study, we demonstrate that HHV8-encoded vFLIP can protect cells against growth factor withdrawal–induced apoptosis. Because growth factor independence is considered one of the hallmarks of cancer,42 these results provide a possible mechanism by which HHV8 might contribute to the development of lymphoproliferative disorders seen in association with infection by this virus.

Based on its sequence homology with the prodomain of caspase-8, earlier studies speculated that the main biologic function of HHV8 vFLIP might be to protect against death receptors–induced apoptosis by blocking the recruitment and activation of caspase-8.4-6However, we recently demonstrated that HHV8 vFLIP can also activate the NF-κB pathway, which is known to protect against apoptosis induced by diverse stimuli.7,8 Consistent with the above observation, the present study demonstrates the ability of HHV8 vFLIP to protect against growth factor withdrawal–induced apoptosis in a factor-dependent cell line. Interestingly, the protective effect of HHV8 vFLIP against growth factor withdrawal–induced apoptosis in the present study was associated with the activation of the NF-κB pathway and not accompanied by the concomitant protection against apoptosis mediated by death receptors or anticancer drugs. Collectively, the above results suggest that the main biologic function of HHV8 vFLIP might be to activate the NF-κB pathway rather than its previously speculated role as an inhibitor of caspase-8 activation during death receptor signaling.

Although the exact mechanism(s) via which HHV8 vFLIP–mediated NF-κB might protect against growth factor withdrawal–induced apoptosis is not entirely clear, our results suggest a role of the Bcl-xL in this process. This antiapoptotic protein is known to be up-regulated by the NF-κB pathway and has been shown to block the processing of caspase-9, -6, and -8.16,28,29 However, in addition to Bcl-xL, the NF-κB pathway is known to induce the expression of a number of other antiapoptotic genes, such as A1, cIAP1, cIAP2, XIAP, andIEX-1.31,43 Thus, it is conceivable that additional genes induced by the NF-κB pathway might contribute to the protective effect of HHV8 vFLIP against growth factor withdrawal–induced apoptosis.

In the present study we demonstrate the ability of the inhibitors of the NF-κB pathway to reverse the protective effect of HHV8 vFLIP against growth factor withdrawal–induced apoptosis. In addition to HHV-associated PEL, abnormal activation of the NF-κB pathway has been previously implicated in the pathogenesis of several lymphoproliferative disorders such as multiple myeloma, Hodgkin disease, Epstein-Barr virus–mediated Burkitt lymphoma, and human T-cell leukemia virus 1–induced adult T-cell leukemia.43A number of drugs targeting this pathway are in various stages of clinical development or in clinical use for the treatment of hematologic malignancies.27,44-46 Our study suggests that the inhibitors of the NF-κB pathway also deserve study in the treatment of HHV8-associated lymphoproliferative disorders.

We would like to thank Dr Richard Gaynor for providing the adenoviral vectors expressing β-galactosidase and IκB superrepressor and Dr Suwan Sinha for technical assistance.

Prepublished online as Blood First Edition Paper, October 24, 2002; DOI 10.1182/blood-2002-07-2072.

Supported by a grant from the National Institutes of Health (CA85177).

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
Boldin
MP
Goncharov
TM
Goltsev
YV
Wallach
D
Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1- and TNF receptor-induced cell death.
Cell.
85
1996
803
815
2
Muzio
M
Chinnaiyan
AM
Kischkel
FC
et al
FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex.
Cell.
85
1996
817
827
3
Fernandes-Alnemri
T
Armstrong
RC
Krebs
J
et al
In vitro activation of CPP32 and Mch3 by Mch4, a novel human apoptotic cysteine protease containing two FADD-like domains.
Proc Natl Acad Sci U S A.
93
1996
7464
7469
4
Thome
M
Schneider
P
Hofmann
K
et al
Viral FLICE-inhibitory proteins (FLIPs) prevent apoptosis induced by death receptors.
Nature.
386
1997
517
521
5
Bertin
J
Armstrong
RC
Ottilie
S
et al
Death effector domain-containing herpesvirus and poxvirus proteins inhibit both Fas- and TNFR1-induced apoptosis.
Proc Natl Acad Sci U S A.
94
1997
1172
1176
6
Hu
S
Vincenz
C
Buller
M
Dixit
VM
A novel family of viral death effector domain-containing molecules that inhibit both CD-95- and tumor necrosis factor receptor-1-induced apoptosis.
J Biol Chem.
272
1997
9621
9624
7
Chaudhary
PM
Jasmin
A
Eby
MT
Hood
L
Modulation of the NF-kappa B pathway by virally encoded death effector domains-containing proteins.
Oncogene.
18
1999
5738
5746
8
Liu
L
Eby
MT
Rathore
N
Sinha
SK
Kumar
A
Chaudhary
PM
The human herpes virus 8-encoded viral FLICE inhibitory protein physically associates with and persistently activates the Ikappa B kinase complex.
J Biol Chem.
277
2002
13745
13751
9
Kitamura
T
Tange
T
Terasawa
T
et al
Establishment and characterization of a unique human cell line that proliferates dependently on GM-CSF, IL-3, or erythropoietin.
J Cell Physiol.
140
1989
323
334
10
Liu
X
Kim
CN
Yang
J
Jemmerson
R
Wang
X
Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c.
Cell.
86
1996
147
157
11
Li
P
Nijhawan
D
Budihardjo
I
et al
Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade.
Cell.
91
1997
479
489
12
Wang
X
The expanding role of mitochondria in apoptosis.
Genes Dev.
15
2001
2922
2933
13
Bernardi
P
Scorrano
L
Colonna
R
Petronilli
V
Di Lisa
F
Mitochondria and cell death. Mechanistic aspects and methodological issues.
Eur J Biochem.
264
1999
687
701
14
Chang
HY
Yang
X
Proteases for cell suicide: functions and regulation of caspases.
Microbiol Mol Biol Rev.
64
2000
821
846
15
Los
M
Wesselborg
S
Schulze-Osthoff
K
The role of caspases in development, immunity, and apoptotic signal transduction: lessons from knockout mice.
Immunity.
10
1999
629
639
16
Engels
IH
Stepczynska
A
Stroh
C
et al
Caspase-8/FLICE functions as an executioner caspase-in anticancer drug-induced apoptosis.
Oncogene.
19
2000
4563
4573
17
Seki
K
Yoshikawa
H
Shiiki
K
Hamada
Y
Akamatsu
N
Tasaka
K
Cisplatin (CDDP) specifically induces apoptosis via sequential activation of caspase-8, -3 and -6 in osteosarcoma.
Cancer Chemother Pharmacol.
45
2000
199
206
18
Cowling
V
Downward
J
Caspase-6 is the direct activator of caspase-8 in the cytochrome c-induced apoptosis pathway: absolute requirement for removal of caspase-6 prodomain.
Cell Death Differ.
9
2002
1046
1056
19
Luo
X
Budihardjo
I
Zou
H
Slaughter
C
Wang
X
Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors.
Cell.
94
1998
481
490
20
Li
H
Zhu
H
Xu
CJ
Yuan
J
Cleavage of BID by caspase-8 mediates the mitochondrial damage in the Fas pathway of apoptosis.
Cell.
94
1998
491
501
21
Keller
SA
Schattner
EJ
Cesarman
E
Inhibition of NF-kappaB induces apoptosis of KSHV-infected primary effusion lymphoma cells.
Blood.
96
2000
2537
2542
22
Baldwin
AS
Jr
The NF-kappa B and I kappa B proteins: new discoveries and insights.
Annu Rev Immunol.
14
1996
649
683
23
Baeuerle
PA
Baltimore
D
NF-kappa B: ten years after.
Cell.
87
1996
13
20
24
Van Antwerp
DJ
Martin
SJ
Kafri
T
Green
DR
Verma
IM
Suppression of TNF-alpha-induced apoptosis by NF-kappaB.
Science.
274
1996
787
789
25
Ghosh
S
May
MJ
Kopp
EB
NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses.
Annu Rev Immunol.
16
1998
225
260
26
Grisham
MB
Palombella
VJ
Elliott
PJ
et al
Inhibition of NF-kappa B activation in vitro and in vivo: role of 26S proteasome.
Methods Enzymol.
300
1999
345
363
27
Estrov
Z
Manna
SK
Harris
D
et al
Phenylarsine oxide blocks interleukin-1 beta-induced activation of the nuclear transcription factor NF-kappaB, inhibits proliferation, and induces apoptosis of acute myelogenous leukemia cells.
Blood.
94
1999
2844
2853
28
Tsukahara
T
Kannagi
M
Ohashi
T
et al
Induction of Bcl-x(L) expression by human T-cell leukemia virus type 1 Tax through NF-kappaB in apoptosis-resistant T-cell transfectants with Tax.
J Virol.
73
1999
7981
7987
29
Khoshnan
A
Tindell
C
Laux
I
Bae
D
Bennett
B
Nel
AE
The NF-kappa B cascade is important in Bcl-xL expression and for the anti-apoptotic effects of the CD28 receptor in primary human CD4+ lymphocytes.
J Immunol.
165
2000
1743
1754
30
Huang
HM
Huang
CJ
Yen
JJ
Mcl-1 is a common target of stem cell factor and interleukin-5 for apoptosis prevention activity via MEK/MAPK and PI-3K/Akt pathways.
Blood.
96
2000
1764
1771
31
Aggarwal
BB
Apoptosis and nuclear factor-kappa B: a tale of association and dissociation.
Biochem Pharmacol.
60
2000
1033
1039
32
Cesarman
E
Chang
Y
Moore
PS
Said
JW
Knowles
DM
Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS- related body-cavity-based lymphomas [see comments].
N Engl J Med.
332
1995
1186
1191
33
Soulier
J
Grollet
L
Oksenhendler
E
et al
Kaposi's sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman's disease.
Blood.
86
1995
1276
1280
34
Gessain
A
Sudaka
A
Briere
J
et al
Kaposi sarcoma-associated herpes-like virus (human herpesvirus type 8) DNA sequences in multicentric Castleman's disease: is there any relevant association in non-human immunodeficiency virus-infected patients?
Blood.
87
1996
414
416
35
Luppi
M
Barozzi
P
Maiorana
A
et al
Human herpesvirus-8 DNA sequences in human immunodeficiency virus-negative angioimmunoblastic lymphadenopathy and benign lymphadenopathy with giant germinal center hyperplasia and increased vascularity.
Blood.
87
1996
3903
3909
36
Schulz
TF
Kaposi's sarcoma-associated herpesvirus (human herpesvirus-8).
J Gen Virol.
79
1998
1573
1591
37
Sturzl
M
Hohenadl
C
Zietz
C
et al
Expression of K13/v-FLIP gene of human herpesvirus 8 and apoptosis in Kaposi's sarcoma spindle cells.
J Natl Cancer Inst.
91
1999
1725
1733
38
Rainbow
L
Platt
GM
Simpson
GR
et al
The 222- to 234-kilodalton latent nuclear protein (LNA) of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) is encoded by orf73 and is a component of the latency-associated nuclear antigen.
J Virol.
71
1997
5915
5921
39
Sarid
R
Flore
O
Bohenzky
RA
Chang
Y
Moore
PS
Transcription mapping of the Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) genome in a body cavity-based lymphoma cell line (BC-1).
J Virol.
72
1998
1005
1012
40
Evan
G
Littlewood
T
A matter of life and cell death.
Science.
281
1998
1317
1322
41
Evan
GI
Vousden
KH
Proliferation, cell cycle and apoptosis in cancer.
Nature.
411
2001
342
348
42
Hanahan
D
Weinberg
RA
The hallmarks of cancer.
Cell.
100
2000
57
70
43
Mayo
MW
Baldwin
AS
The transcription factor NF-kappaB: control of oncogenesis and cancer therapy resistance.
Biochim Biophys Acta.
1470
2000
M55
M62
44
Adams
J
Elliott
PJ
New agents in cancer clinical trials.
Oncogene.
19
2000
6687
6692
45
Keifer
JA
Guttridge
DC
Ashburner
BP
Baldwin
AS
Jr
Inhibition of NF-kappa B activity by thalidomide through suppression of IkappaB kinase activity.
J Biol Chem.
276
2001
22382
22387
46
Roussel
RR
Barchowsky
A
Arsenic inhibits NF-kappaB-mediated gene transcription by blocking IkappaB kinase activity and IkappaBalpha phosphorylation and degradation.
Arch Biochem Biophys.
377
2000
204
212

Author notes

Preet M. Chaudhary, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas TX 75390-8593; e-mail:preet.chaudhary@utsouthwestern.edu.

Sign in via your Institution