In the present study, treatment of HEK-293 cells with the synthetic small molecule N-iodoacetyl-tryptophan (I-Trp) at submicromolar concentrations efficiently induced cell apoptosis as judged from the accumulation of sub-G0 cells and intracellular DNA fragmentation. Activation of all intracellular caspases, except caspase-1, was detected in I-Trp–treated cells. Proteomic analysis revealed that β-tubulin acted as a specific intracellular target of I-Trp. Protein fingerprinting analysis indicated that the Cys354 residue in the peptide fragment TAVCDIPPR of β-tubulin, which is located at the binding interface with chaperonin containing TCP1-β (CCT-β), was alkylated by I-Trp. Moreover, site-directed mutagenesis of Cys354 (Cys-Ala) abolished the incorporation of I-Trp into β-tubulin, suggesting Cys354 is indeed the targeting site of I-Trp. Immunoprecipitation showed that the β-tubulin/CCT-β complex was constitutively formed but disrupted after treatment with I-Trp. Overexpression of the truncated β-tubulin (T351-S364) or treatment with I-Trp or the synthetic peptide Myr-TAVCDIPPRG caused more severe cell apoptosis in multidrug-resistant MES-SA/Dx5 cancer cells due to higher levels of CCT-β relative to wild-type MES-SA cancer cells. Silencing the expression of CCT-β rendered MES-SA/Dx5 cells less sensitive to I-Trp–induced apoptotic cell death. These findings suggest that the β-tubulin/CCT-β complex may serve as an effective chemotherapeutic target for treating clinical tubulin-binding agent-resistant or CCT-β–overexpressing tumors. [Cancer Res 2009;69(17):6879–88]

Iodoacetamide-based derivatives are known to interact with the low pKa cysteine residues of intracellular proteins, such as tubulin (1), myosin (2), actin (3), HMG-CoA reductase (4), sarcoplasmic reticulum Ca2+-ATPase (5), and protein tyrosine phosphatase 1B (6), to yield S-carboxymethyl cysteine derivatives. Recently, 3-(iodoacetamido)-benzoyl ethyl ester (3-IAABE) was found to inhibit tubulin polymerization; cause a double blockade in the cell cycle at the G1-S transition and M phase through the increased kinase activities of cyclin E, cyclin-dependent kinase 2, cyclin B, and cdc2; and induce activation of caspase-9, caspase-3, and caspase-6, as well as the caspase-3 downstream effector, poly(ADP-ribose) polymerase (PARP), and accompanied cellular DNA fragmentation in CEM cells (7). In addition, 3-IAABE also inhibits the growth of human hepatocarcinoma in nude mice by a 72% decrease in tumor volume. This inhibition is more robust than treatment with vincristine (7), a tubulin-binding and antimitotic agent indicating the feasibility of using iodoacetamide-based derivatives in clinical cancer therapies.

Tubulin is a subunit of intracellular microtubules and is composed of an α and β heterodimer (8). Unlike α-tubulin, which binds GTP irreversibly, β-tubulin binds GTP reversibly and hydrolyzes it to GDP (9). Changes in the concentration of intracellular GDP-bound β-tubulin is known to affect intracellular microtubule dynamics (10), which play critical role in regulating the separation of chromosomes during meiosis and mitosis (11, 12). Several anticancer agents, such as paclitaxel (13), vinblastine (14), and colchicine (15), were designed to disrupt the α/β tubulin heterodimer so as to inhibit microtubule dynamics (16). However, occurrence of drug-resistant tumor cells limits the use of these antimitotic agents in current clinical cancer therapies. Although the molecular mechanisms promoting the induction of drug-resistant tumor cells remain unclear, the overexpression (1720) or mutation (21, 22) of β-tubulin isotypes is thought to be crucial for this event.

Chaperonin containing TCP1 (CCT) is involved in the folding of cytoskeleton proteins, actin and tubulin (23), and other intracellular proteins, such as cyclin E1 (24), histone deacetylase (25), and protein phosphatase PP2A regulatory subunit B (26). CCT is divided into eight different subunits (α, β, γ, δ, ε, ζ, η, and 𝛉), each with an approximate molecular mass of 60 kDa. Although the functions of CCT containing complexes are still not well understood, the interaction of CCT with tubulin has been illustrated using cryoelectron microscopic (CryoEM) analysis (27). The docking model reveals that CCT subunits interact with different regions of α/β tubulin heterodimers to stabilize the microtubular structure (27). In the case of β-tubulin, for example, CCT-β interacts with the fragment V353-P357 within its COOH terminal region (27).

Although eukaryotic CCT plays an important function in maintaining cellular homoeostasis by assisting the folding of many proteins, the nature of those proteins and the involved cellular pathways have not yet been established. The present study offers a detailed illustration of β-tubulin/CCT-β complex interruption by a synthetic small molecule, N-iodoacetyl-tryptophan (I-Trp), via its incorporation at the Cys354 residue of β-tubulin. Furthermore, this interaction elicited caspase-dependent signaling and consequent cellular apoptosis. Our results also show that interrupting the constitutive β-tubulin/CCT-β complex causes more severe cell apoptosis in the multidrug-resistant MES-SA/Dx5 uterine cancer cells when compared with wild-type MES-SA cells. These observations indicate that targeting the β-tubulin/CCT-β complex may serve as a novel chemotherapeutic strategy for treating clinical multidrug-resistant tumors.

Materials. All the chemicals for organic synthesis were purchased from Aldrich-Sigma. N-iodoacetylated amino acids (1), dansylated I-Trp (I-Trp-Dan) (2), and biotinylated I-Trp (I-Trp-B) (3) were synthesized according to Supplementary Scheme S1. The human cDNA and pET32Xa/Lic vector were purchased from Invitrogen. Tubulin polymerization kit was purchased from Cytoskeleton. Protein A-agarose and antibodies against β-tubulin and CCT-β were purchased from Santa Cruz Biotechnology. Antibodies against caspase-7, caspase-9, and PARP were purchased from Cell Signaling. HEK-293 cells and MES-SA and MES-SA/Dx-5 uterine cancer cells were obtained from the Bioresource Collection and Research Center in Taiwan and cultivated in accordance with manufacturer's guideline.

Plasmid construction and protein expression. The full-length β-tubulin cDNA was subcloned into pET32 Xa/LIC vector according to manufacturer's protocol (Invitrogen). The constructed pET32 Xa/LIC/β-tubulin plasmid was used as a DNA template for site-directed mutagenesis of the Cys354 residue to Ala. Site-directed mutagenesis was carried out by using QuikChange II Site-Directed Mutagenesis kit (Strategene). Recombinant wild-type β-tubulin and C354A mutant β-tubulin proteins were overexpressed in the Escherichia coli strain BL-21(DE3) as COOH terminally hexa-His-tagged proteins.

Protein purification. Cell lysates obtained from a 2-L culture of E. coli strain BL-21(DE3) were resuspended in 80 mL buffer A [12 mmol/L Tris-HCl (pH 7.5), 120 mmol/L NaCl] and loaded onto a 20-mL Ni-NTA resin that was equilibrated with buffer A. The column was washed with excess buffer A containing 10 mmol/L imidazole. His-tagged β-tubulin was eluted with buffer A containing 300 mmol/L imidazole. His-tagged β-tubulin protein was then digested with thrombin protease to remove the peptide tag.

For identifying I-Trp-B–incorporated protein(s), cell lysate (20 mg) of HEK-293 cells was prepared in 1 mL buffer [25 mmol/L Tris-HCl (pH 7.4), 150 mmol/L NaCl] and incubated with I-Trp-B compound (200 μmol/L) overnight at 4°C. Proteins linked with I-Trp-B were purified by affinity chromatography using a HiTrap Streptavidin affinity column according to manufacturer's guideline (GE Healthcare).

Mass spectrometric analysis. HEK-293 cells or recombinant β-tubulin (100 μg) were treated with I-Trp-Dan (1 μmol/L) for 24 h at 37°C or 2 h at room temperature in the dark, respectively. I-Trp-Dan–treated cell lysates (100 μg) or recombinant β-tubulin (30 μg) were subjected to electrophoresis on a 10% SDS-PAGE gel. The fluorescence-labeled protein band, visualized under a UV lamp, was excised from the gel and subjected to in-gel tryptic digestion and liquid chromatography-electrospray ionization-tandem mass spectrometric analysis. The raw data were converted to the Mascot (Matrix Science) generic format and searched against the NCBInr human protein database. For protein fingerprinting analysis, the resultant m/z values of each peptide fragment were compared with predicted molecular weight of peptide fragment derived from β-tubulin IVb subtype to assess possible I-Trp-Dan–mediated alkylation of cysteines.

Cell transfection. The gene encoding the β-tubulin T351-S364 fragment was synthesized by Mission Biotech and subcloned into an EGFP-IRES2 vector with an XhoI/EcoRI restriction site. The transfection of plasmid DNA or CCT-β small interfering RNA (siRNA) was performed according to Lipofectamine transfection guidelines (Invitrogen).

SDS-PAGE and Western blotting. Cell lysate (100 μg) or recombinant β-tubulin (20 μg) was subjected to SDS-PAGE. In the case of I-Trp-Dan compound treatment, proteins were detected by exposure to UV light (280 nm) before Coomassie blue staining. For Western blotting, proteins were transferred onto a polyvinylidene difluoride membrane (Millipore) and incubated with an antibody against β-tubulin, CCT-β, caspase-7, caspase-9, or PARP. After incubation with horseradish peroxidase–conjugated secondary antibody, immunoreactive protein bands were visualized using the enhanced chemiluminescence system (Amersham Bioscience).

Immunoprecipitation. Cell lysates (200 μg) were diluted in 1 mL of cell lysis buffer [10 mmol/L Tris-HCl (pH 7.4), 140 mmol/L NaCl, 3 mmol/L MgCl2, 2 mmol/L EDTA, 5 mmol/L EGTA, 0.5% Triton X-100, 4% protease inhibitor cocktail; Merck Biosciences] and incubated with 2 μg of anti–β-tubulin antibody for 2 h at 4°C, followed by precipitation with 20 μL of protein A-agarose beads for 1 h at 4°C. The immunoprecipitates were analyzed by SDS-PAGE/Western blotting using a CCT-β antibody. In addition, the blots were stripped using a commercial kit (Millipore) and reprobed with an anti–β-tubulin antibody.

Flow cytometric analysis. Cells (6 × 105) were fixed in 70% ice-cold ethanol for 30 min at room temperature. Cells were washed with PBS once and then incubated with propidium iodide (PI) staining solution (PBS containing 0.1% bovine serum albumin, 0.1% RNase A, and 20 ng/mL of PI) for 30 min at room temperature in the dark. After the incubation, the cells were analyzed by flow cytometry to assess their DNA content.

Determination of caspase activities. The caspase activity assay was performed according to the manufacturer's guidelines (BioVision). Fold increases in caspase activity was determined by comparing these results with the level of the untreated control.

Computer modeling. Molecular docking of I-Trp compounds into the β-tubulin protein structure was evaluated using GEMDOCK software (28). Intermolecular interactions were predicted using Swiss-PdbViewer 3.7.

Synthesis of N-iodoacetylated amino acids and evaluation of their cytotoxic effects. A series of N-iodoacetylated amino acids were synthesized in accordance with the procedure described in Supplementary Scheme S1. The identities of all synthesized compounds were confirmed by mass spectrometric and nuclear magnetic resonance analyses (data not shown). The treatment of HEK-293 cells with 1 μmol/L I-Asp, I-Glu, I-Ser, and I-Trp (chemical structures shown in Fig. 1A), but not I-Gly, I-Lys, or iodoacetamide, induced the accumulation of sub-G0 cell population compared with untreated cells (Supplementary Fig. S1). Effects of these compounds on sub-G0 cell accumulation of HEK-293 cells were dose dependent at concentrations ranging from 0.001 to 1 μmol/L (Fig. 1B,, left) and also caused intracellular DNA fragmentation (Fig. 1B , right), indicating intracellular apoptotic signaling. Based on flow cytometric analysis, I-Trp exhibited the most potent cytotoxic effect on HEK-293 cells. Thereafter, we used the I-Trp compound in subsequent experiments to elucidate the mechanism by which it induced cell apoptosis.

Figure 1.

Effects of N-iodoacetylated amino acids on cellular survival and activation of caspases in HEK-293 cell culture. A, chemical structures of I-Asp, I-Glu, I-Ser, and I-Trp. B, populations of HEK-293 sub-G0 cells treated with iodoacetamide (IDAM; control), I-Asp, I-Glu, I-Ser, or I-Trp at indicated dosages for 24 h (left). Genomic DNA extracted from the HEK-293 cells treated without or with iodoacetamide (IDAM), I-Asp, I-Glu, I-Ser, or I-Trp at 1 μmol/L for 48 h (right). C, HEK-293 cells were treated without (open column) as the control group or with iodoacetamide (gray column) or I-Trp (black column) at 1 μmol/L for 24 h. Cell lysates were subjected to caspase activity assays. D, HEK-293 cells were pretreated with caspase-3/caspase-7–specific peptide inhibitor (DEVD) at designated concentrations for at least 40 min before the treatment without or with I-Trp at 1 μmol/L for 24 h. The column height represents mean percentage of sub-G0 cells among total cells derived from three independent experiments.

Figure 1.

Effects of N-iodoacetylated amino acids on cellular survival and activation of caspases in HEK-293 cell culture. A, chemical structures of I-Asp, I-Glu, I-Ser, and I-Trp. B, populations of HEK-293 sub-G0 cells treated with iodoacetamide (IDAM; control), I-Asp, I-Glu, I-Ser, or I-Trp at indicated dosages for 24 h (left). Genomic DNA extracted from the HEK-293 cells treated without or with iodoacetamide (IDAM), I-Asp, I-Glu, I-Ser, or I-Trp at 1 μmol/L for 48 h (right). C, HEK-293 cells were treated without (open column) as the control group or with iodoacetamide (gray column) or I-Trp (black column) at 1 μmol/L for 24 h. Cell lysates were subjected to caspase activity assays. D, HEK-293 cells were pretreated with caspase-3/caspase-7–specific peptide inhibitor (DEVD) at designated concentrations for at least 40 min before the treatment without or with I-Trp at 1 μmol/L for 24 h. The column height represents mean percentage of sub-G0 cells among total cells derived from three independent experiments.

Close modal

Because caspase activation is thought to be critical for intracellular apoptotic signaling (29), the activities of intracellular caspases were determined in I-Trp–treated HEK-293 cells. Except caspase-1, the increased activities of caspase-2 through caspase-10 were detected after HEK-293 cells were treated for 24 hours with I-Trp (1 μmol/L), but not iodoacetamide (Fig. 1C). Especially, I-Trp induced an approximate of 2.5-fold elevation in caspase-3/caspase-7 activities in HEK-293 cells compared with control cells (Fig. 1C). Furthermore, the inclusion of caspase-3/caspase-7 peptide inhibitor (DEVD) at concentrations ranging from 0.3 to 3 μmol/L significantly (P < 0.05) suppressed the I-Trp–induced sub-G0 cell accumulation of HEK-293 cells in a dose-dependent manner (Fig. 1D).

Identification of intracellular protein target(s) of I-Trp in HEK-293 cells. To identify the intracellular protein target(s) of I-Trp, a dansylated I-Trp compound (I-Trp-Dan; Fig. 2A,, insert) was synthesized and given to HEK-293 cell cultures. As shown in Fig. 2A, I-Trp-Dan induced generation of apoptotic cells in a dose-dependent manner at concentrations ranging from 0.05 to 2 μmol/L. SDS-PAGE analysis of cell lysates obtained from I-Trp-Dan (1 μmol/L)–treated cells revealed a fluorescent protein band with a molecular weight of ∼55 kDa after exposure of the gel to UV light (Fig. 2B). The fluorescent protein band was excised from gel and subjected to mass spectrometric analysis. As shown in Supplementary Table S1, CCT-β subunit, tubulin α/β subunits, and other proteins were identified in the mass spectrometric analysis. These proteins all have similar molecular weights and run on SDS-PAGE at similar positions. To identify the real target, an I-Trp-B compound (Fig. 2C,, insert) was synthesized and incubated with HEK-293 cell lysates. Subsequently, proteins covalently linked to this I-Trp-B compound were purified by affinity chromatography using a streptavidin-immobilized column. Large amounts of unbound proteins were washed from the column, and a small quantity of streptavidin-bound protein was eluted using a urea solution (Fig. 2D). Western blot analysis revealed that β-tubulin, not α-tubulin or CCT-β, was present in the eluted solution (Fig. 2D), suggesting that β-tubulin acts as an intracellular protein target of I-Trp in HEK-293 cells.

Figure 2.

Identification of the intracellular target of the I-Trp compound. A, apoptotic HEK-293 cells treated with the I-Trp-Dan compound (insert) at indicated dosages for 24 h. After the incubation, cells were collected and subjected to PI staining and then analyzed by flow cytometry. B, HEK-293 cells were treated without or with I-Trp-Dan at 1 μmol/L for 24 h, and the cell lysates were analyzed by SDS-PAGE stained with Coomassie blue (left). The dansylated protein band was visualized under UV light (right). C, I-Trp-B compound (insert)–incorporated proteins of HEK-293 cells were purified by affinity chromatography using a streptavidin column. D, streptavidin-unbound (flow-through, FT) and bound (Elute) proteins were analyzed by SDS-PAGE and Western blot analysis using α-tubulin, β-tubulin, or CCT-β–specific antibodies.

Figure 2.

Identification of the intracellular target of the I-Trp compound. A, apoptotic HEK-293 cells treated with the I-Trp-Dan compound (insert) at indicated dosages for 24 h. After the incubation, cells were collected and subjected to PI staining and then analyzed by flow cytometry. B, HEK-293 cells were treated without or with I-Trp-Dan at 1 μmol/L for 24 h, and the cell lysates were analyzed by SDS-PAGE stained with Coomassie blue (left). The dansylated protein band was visualized under UV light (right). C, I-Trp-B compound (insert)–incorporated proteins of HEK-293 cells were purified by affinity chromatography using a streptavidin column. D, streptavidin-unbound (flow-through, FT) and bound (Elute) proteins were analyzed by SDS-PAGE and Western blot analysis using α-tubulin, β-tubulin, or CCT-β–specific antibodies.

Close modal

To further confirm that β-tubulin was an authentic target of I-Trp, recombinant β-tubulin was produced as described in Materials and Methods. As shown in Fig. 3A, the treatment of recombinant β-tubulin, not CCT-β (Supplementary Fig. S2), with the I-Trp-Dan compound generated a fluorescent protein band on SDS-PAGE after UV light exposure. Furthermore, the incubation of recombinant β-tubulin with I-Asp, I-Glu, I-Ser, I-Trp, or I-Trp-B compounds before the I-Trp-Dan treatment abrogated generation of the fluorescent protein band (Fig. 3B), indicating that those synthesized compounds likely interacted and alkylated the same cysteine residue of β-tubulin.

Figure 3.

Identification of the I-Trp incorporating site in β-tubulin. A, recombinant human β-tubulin was incubated without or with I-Trp-Dan and analyzed by SDS-PAGE (left) or pretreated with I-Asp, I-Glu, I-Ser, I-Trp, or I-Trp-B before I-Trp-Dan treatment (right). B, I-Trp-Dan–treated β-tubulin protein excised from the gel was subjected to mass spectrometric analysis. A distinct peak with m/z = 1532.275 was assigned as dansylated β-tubulin (arrow). C, wild-type (wt) and C354A mutant (mt) β-tubulin (100 μg) were incubated with I-Trp-Dan at designated concentrations and subsequently analyzed by SDS-PAGE. Dansylated proteins were visualized under UV light (top) or stained with Coomassie blue (bottom). D, cell lysates of HEK-293 cells treated without or with I-Trp (1 μmol/L) were immunoprecipitated with β-tubulin antibody and then immunoblotted with CCT-β antibody (top). The same blots were reprobed with β-tubulin antibody to quantify protein input in immunoprecipitation.

Figure 3.

Identification of the I-Trp incorporating site in β-tubulin. A, recombinant human β-tubulin was incubated without or with I-Trp-Dan and analyzed by SDS-PAGE (left) or pretreated with I-Asp, I-Glu, I-Ser, I-Trp, or I-Trp-B before I-Trp-Dan treatment (right). B, I-Trp-Dan–treated β-tubulin protein excised from the gel was subjected to mass spectrometric analysis. A distinct peak with m/z = 1532.275 was assigned as dansylated β-tubulin (arrow). C, wild-type (wt) and C354A mutant (mt) β-tubulin (100 μg) were incubated with I-Trp-Dan at designated concentrations and subsequently analyzed by SDS-PAGE. Dansylated proteins were visualized under UV light (top) or stained with Coomassie blue (bottom). D, cell lysates of HEK-293 cells treated without or with I-Trp (1 μmol/L) were immunoprecipitated with β-tubulin antibody and then immunoblotted with CCT-β antibody (top). The same blots were reprobed with β-tubulin antibody to quantify protein input in immunoprecipitation.

Close modal

Furthermore, protein fingerprinting analyses indicated that the Cys354 residue within the β-tubulin fragment TAVCDIPPR was alkylated with the I-Trp-Dan compound. This observation is based on the resultant mass spectrometer peak at m/z = 1532.751, approximately equal to the predicted mass values of the TAVCDIPPR peptide fragment incorporated with an N-acetyl-Trp-Dan group (Fig. 3B). In contrast to the wild-type β-tubulin, a Cys354Ala mutant failed to produce a fluorescently labeled band on SDS-PAGE (Fig. 3C), consistent with the conclusion that Cys354 reacted with I-Trp. Interestingly, the Cys354 residue is located within the peptide fragment VCDIP of β-tubulin (the magenta region in Supplementary Fig. S3), which serves as the CCT-β binding site (27). We further found that I-Trp caused the disruption of the constitutively formed β-tubulin/CCT-β complex, as judged by the reduced immunoprecipitates of β-tubulin/CCT-β complex in cell lysates of I-Trp–treated HEK-293 cells (Fig. 3D).

Intermolecular interactions between the I-Trp compound and β-tubulin. Because the I-Trp compound disrupts constitutively associated β-tubulin/CCT-β complex by alkylating the Cys354 residue of β-tubulin, computer modeling was used to simulate the intermolecular interactions between I-Trp and β-tubulin. Docking experiments display that I-Trp binds to β-tubulin at the region near the Cys354 residue (Fig. 4A). The intermolecular distance between the iodo group of I-Trp and the SH group of Cys354 was calculated to be 3.24 Å (enlarged in Fig. 4A). Possible hydrogen bond interactions with the Asp355 residue and hydrophobic interactions with Leu42, Pro243, Gly244, and Gln245 residues of β-tubulin may also be formed (Supplementary Fig. S4B). These interactions ensure the specific incorporation of I-Trp into this region of β-tubulin. Similar binding modes of I-Trp-Dan and I-Trp-B with β-tubulin were also predicted in the same region (Supplementary Fig. S4A and B).

Figure 4.

Molecular docking model of I-Trp in β-tubulin and its effect on tubulin polymerization. A, molecular docking of I-Trp in β-tubulin [protein data bank (PDB) entry 1Z2B] was performed using GEMDOCK software. The intermolecular distance between the iodo group of I-Trp and the SH group of the Cys354 residue was determined to be 3.24 Å (enlarged figure) using Swiss-PdbViewer 3.7. B, the structure of β-tubulin complexed with Taxol was taken from PDB entry 1TUB and analyzed using Pymol software. The β-tubulin subunit is in blue. The peptide VCDIP (353–357) of β-tubulin, which interacts with CCT-β, is in magenta. The Cys354 residue is in yellow. Taxol (TXL) located in the β-tubulin subunit is in orange. C, recombinant β-tubulin proteins (100 μg) were preincubated with paclitaxel or I-Trp (1 μmol/L of each) before the addition of I-Trp-Dan (1 μmol/L). Dansylated protein was visualized under a UV light (top) or by staining with Coomassie blue (bottom), as shown in the inserted figures. An in vitro tubulin polymerization assay was performed in the presence of paclitaxel (10 nmol/L), I-Trp (1 μmol/L), or DMSO (0.1%) as a solvent control. The results obtained from triplicate assays were normalized relative to time 0.

Figure 4.

Molecular docking model of I-Trp in β-tubulin and its effect on tubulin polymerization. A, molecular docking of I-Trp in β-tubulin [protein data bank (PDB) entry 1Z2B] was performed using GEMDOCK software. The intermolecular distance between the iodo group of I-Trp and the SH group of the Cys354 residue was determined to be 3.24 Å (enlarged figure) using Swiss-PdbViewer 3.7. B, the structure of β-tubulin complexed with Taxol was taken from PDB entry 1TUB and analyzed using Pymol software. The β-tubulin subunit is in blue. The peptide VCDIP (353–357) of β-tubulin, which interacts with CCT-β, is in magenta. The Cys354 residue is in yellow. Taxol (TXL) located in the β-tubulin subunit is in orange. C, recombinant β-tubulin proteins (100 μg) were preincubated with paclitaxel or I-Trp (1 μmol/L of each) before the addition of I-Trp-Dan (1 μmol/L). Dansylated protein was visualized under a UV light (top) or by staining with Coomassie blue (bottom), as shown in the inserted figures. An in vitro tubulin polymerization assay was performed in the presence of paclitaxel (10 nmol/L), I-Trp (1 μmol/L), or DMSO (0.1%) as a solvent control. The results obtained from triplicate assays were normalized relative to time 0.

Close modal

Effect of the I-Trp compound on tubulin polymerization. The binding site of I-Trp in β-tubulin (Fig. 4B, peptide fragment VCDIP in magenta and Cys354 in yellow) is different from those of other β-tubulin–binding agents, such as Taxol (Fig. 4B,, TXL), colchicine, and vinblastine (Supplementary Fig. S3). As shown in the insert in Fig. 4C, the pretreatment of recombinant β-tubulin with paclitaxel, as well as colchicine and vinblastine (data not shown), did not affect the incorporation of I-Trp-Dan into β-tubulin. Unlike paclitaxel, the treatment with I-Trp failed to induce in vitro tubulin polymerization (Fig. 4C).

Effects of interrupting the β-tubulin/CCT-β interaction in multidrug-resistant cancer cells. Cys354 residue of β-tubulin is located within the CCT-β–binding sequence VCDIP, which is conserved in all β-tubulin isotypes of mammalian cells (Supplementary Fig. S5). Therefore, MES-SA/Dx5 cell line, a multidrug-resistant derivative from uterine MES-SA sarcoma cell line (30), was used to examine whether interrupting β-tubulin/CCT-β association contributes a new strategy to treat tumors with drug resistance to β-tubulin–binding agents, such as paclitaxel, vinblastine, and colchicine. MES-SA and MES-SA/Dx5 cells were cultivated and transfected with the gene encoding the peptide fragment TAVCDIPPR to block the formation of the intracellular β-tubulin/CCT-β complex. As shown in Fig. 5A, overexpression of this peptide fragment caused a more predominant accumulation of sub-G0 cells in MES-SA/Dx5 cells compared with the wild-type MES-SA cells. Particularly, in the cells transfected with 2 μg of plasmid containing this truncated β-tubulin gene, an approximate of 10-fold larger population of sub-G0 cells was observed in MES-SA/Dx5 cells relative to MES-SA cells (Fig. 5A,, left) and intracellular DNA fragmentation (right) was detected. Similar observations were made using 1 μmol/L I-Trp or 10 to 100 μmol/L myristoylated synthetic peptide (Myr-TAVCDIPPRG; Supplementary Fig. S6). Furthermore, immunoprecipition revealed that overexpression of the truncated β-tubulin reduced the formation of intracellular β-tubulin/CCT-β complex in MES-SA/Dx5 cells (Fig. 5B). Surprisingly, Western blot analysis revealed that an increased CCT-β, not β-tubulin, protein expression was detected in MES-SA/Dx5 cells compared with the wild-type MES-SA cells (Fig. 5C). This could explain the predominant activity of overexpressed β-tubulin fragment in causing sub-G0 cell accumulation in MES-SA/Dx5 cells. The knockdown of CCT-β expression using CCT-β–specific siRNA markedly suppressed endogenous CCT-β protein levels (Fig. 5D,, top) and significantly (P < 0.05) inhibited I-Trp compound–induced sub-G0 cell accumulation in MES-SA/Dx5 cells compared with untreated and control experiments (Fig. 5D).

Figure 5.

Effects of β-tubulin/CCT-β complex interruption by the overexpression of peptide (T351-S364) in MES-SA and MES-SA/Dx-5 cells. A, MES-SA and MES-SA/Dx-5 cells were transfected without (Untreated) or with the pIRES2-EGFP vector (Vector only) or the pIRES2-EGFP vector containing the truncated β-tubulin peptide fragment T351-S364 (p[β-Tub (T351-S364)]). The mean percentage of the sub-G0 cell population was obtained from three independent experiments (left). The isolated genomic DNA from MES-SA/Dx5 cells transfected without (Untreated) or with vector only or the p(β-Tub [T351-S364]) plasmid (2 μg of each) for 72 h was analyzed by electrophoresis using a 1.8% agarose gel (right). B, cell lysates were extracted from MES-SA/Dx-5 cells transfected without or with the p(β-Tub [T351-S364]) plasmid (2 μg) for 48 h and then immunoprecipitated with β-tubulin–specific antibodies and immunoblotted using a CCT-β antibody. The same blots were reprobed with the β-tubulin antibody to quantify protein input in immunoprecipitation. C, cell lysates obtained from MES-SA and MES-SA/Dx-5 cells were analyzed by SDS-PAGE/Western blot using CCT-β or β-tubulin antibodies. D, MES-SA/Dx-5 cells were transfected without or with CCT-β siRNA or nontargeting control at 100 nmol/L for 48 h before treating with I-Trp at 1 μmol/L for another 24 h. Cell lysates were analyzed by SDS-PAGE/Western blot using CCT-β or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) antibodies (top). GAPDH was used as an internal control for protein loading. The above cells were also subjected to PI staining and flow cytometric analysis.

Figure 5.

Effects of β-tubulin/CCT-β complex interruption by the overexpression of peptide (T351-S364) in MES-SA and MES-SA/Dx-5 cells. A, MES-SA and MES-SA/Dx-5 cells were transfected without (Untreated) or with the pIRES2-EGFP vector (Vector only) or the pIRES2-EGFP vector containing the truncated β-tubulin peptide fragment T351-S364 (p[β-Tub (T351-S364)]). The mean percentage of the sub-G0 cell population was obtained from three independent experiments (left). The isolated genomic DNA from MES-SA/Dx5 cells transfected without (Untreated) or with vector only or the p(β-Tub [T351-S364]) plasmid (2 μg of each) for 72 h was analyzed by electrophoresis using a 1.8% agarose gel (right). B, cell lysates were extracted from MES-SA/Dx-5 cells transfected without or with the p(β-Tub [T351-S364]) plasmid (2 μg) for 48 h and then immunoprecipitated with β-tubulin–specific antibodies and immunoblotted using a CCT-β antibody. The same blots were reprobed with the β-tubulin antibody to quantify protein input in immunoprecipitation. C, cell lysates obtained from MES-SA and MES-SA/Dx-5 cells were analyzed by SDS-PAGE/Western blot using CCT-β or β-tubulin antibodies. D, MES-SA/Dx-5 cells were transfected without or with CCT-β siRNA or nontargeting control at 100 nmol/L for 48 h before treating with I-Trp at 1 μmol/L for another 24 h. Cell lysates were analyzed by SDS-PAGE/Western blot using CCT-β or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) antibodies (top). GAPDH was used as an internal control for protein loading. The above cells were also subjected to PI staining and flow cytometric analysis.

Close modal

Caspase activation in MES-SA/Dx5 cells transfected with truncated β-tubulin. Further experiments were performed to examine the activation of caspase proteins during apoptosis of MES-SA/Dx5 cells induced by the overexpression of the β-tubulin peptide fragment T351-S364. As shown in Fig. 6A, except caspase-1, all the other caspases were activated in the MES-SA/Dx5 cells transfected with the truncated β-tubulin. The cleaved forms of caspase-3, caspase-7, and caspase-9 (Fig. 6B) and the fragmentation of PARP, an intracellular downstream target of caspase-3/caspase-7 (Fig. 6C), were observed in the MES-SA/Dx5 cells transfected (T) with truncated β-tubulin gene compared with the untreated (U) control. Interestingly, the profile of caspase activation in the truncated β-tubulin–expressing MES-SA/Dx5 cells was similar to that in the I-Trp–treated HEK-293 cells, indicating that a unique intracellular caspase cascade is involved in mediating apoptotic signaling induced by interrupting constitutive β-tubulin/CCT-β complexes (see Discussion).

Figure 6.

Caspase activation in MES-SA/Dx-5 cells transfected with the truncated β-tubulin peptide fragment T351-S364. A, cell lysates obtained from MES-SA/Dx-5 cells transfected without (untreated) or with the pIRES2-EGFP vector (mock) or the pIRES2-EGFP vector containing the truncated β-tubulin peptide fragment T351-S364 (β-tub (T351-S364); 2 μg of each) for 48 h and subjected to caspase activity assays. B, MES-SA/Dx-5 cells were transfected without (U) or with (T) the β-tub (T351-S364) (2 μg) for 48 h. Cell lysates were subjected to SDS-PAGE and Western blot analyses using caspase-3, caspase-7, or caspase-9–specific antibodies. C, the blots were also analyzed by Western blot with a PARP-specific antibody. F and C represent full-length and cleaved proteins, respectively.

Figure 6.

Caspase activation in MES-SA/Dx-5 cells transfected with the truncated β-tubulin peptide fragment T351-S364. A, cell lysates obtained from MES-SA/Dx-5 cells transfected without (untreated) or with the pIRES2-EGFP vector (mock) or the pIRES2-EGFP vector containing the truncated β-tubulin peptide fragment T351-S364 (β-tub (T351-S364); 2 μg of each) for 48 h and subjected to caspase activity assays. B, MES-SA/Dx-5 cells were transfected without (U) or with (T) the β-tub (T351-S364) (2 μg) for 48 h. Cell lysates were subjected to SDS-PAGE and Western blot analyses using caspase-3, caspase-7, or caspase-9–specific antibodies. C, the blots were also analyzed by Western blot with a PARP-specific antibody. F and C represent full-length and cleaved proteins, respectively.

Close modal

CryoEM analysis reveals that CCT-β interacts with amino acid residues 353 to 357 (VCDIP) of β-tubulin (27), a region highly conserved in β-tubulin isotypes (see Supplementary Fig. S5). By mass spectrometric analysis, the conserved Cys354 residue of β-tubulin was identified to serve as the incorporation site of I-Trp compound. Furthermore, treatment with I-Trp or overexpression of truncated β-tubulin (T351-S364) indeed reduced the formation of intracellular β-tubulin/CCT-β complexes and caused cell apoptosis. Because I-Trp did not affect in vitro tubulin polymerization (Fig. 4C), the disruption of intracellular β-tubulin/CCT-β complexes by I-Trp may induce cell apoptosis by causing microtubule disassembly in accordance with the results presented in a previous study (31).

Recently, tubulin-binding agent–resistant tumors were considered to be correlated with the occurrence of β-tubulin mutations (22) or overexpression of individual β-tubulin isotypes (17, 19, 20) such as class III β-tubulin. Our data indicate that CCT-β is highly expressed in multidrug-resistant MES-SA/Dx-5 uterine cancer cells, and the destruction of intracellular β-tubulin/CCT-β complex by the small molecule I-Trp, synthetic peptide Myr-TAVCDIPPRG (T351-G360), or overexpression of the β-tubulin fragment T351-S364 causes a predominant cell apoptosis response in MES-SA/Dx-5 cells when compared with wild-type MES-SA cells. Conversely, the knockdown of CCT-β protein expression rendered MES-SA/Dx5 cells less sensitive to apoptotic signals elicited through interrupting constitutive β-tubulin/CCT-β complex by I-Trp compound. Because increased CCT-β expression is also detectable in several malignant tumors (32) and might occur commonly in drug-resistant tumors, the constitutive β-tubulin/CCT-β complex may serve as a useful chemotherapeutic target for treating clinical multidrug-resistant or CCT-β-overexpressing tumors.

The interaction of microtubules with mitochondria was identified in myocytes (33) and brain cells (34). Recently, microtubular depolymerization was found to induce mitochondria-dependent apoptotic signaling accompanied with activation of caspase-9 and caspase-3 (35). On the other hand, the treatment with tubulin-binding agent Taxol transactivates the intracellular domain of the Notch receptor and modulates the Notch signaling pathway–dependent survival of several cancer cells (36, 37). Besides, the association of tubulin with other receptors, including the muscarinic receptor (38), γ-aminobutyric acid receptor (39), purinergic receptor (40), and interleukin-2 receptor (41), was also previously documented. This information supports our data indicating that the disruption of the constitutive β-tubulin/CCT-β complex by treatment with I-Trp or transfection with the β-tubulin fragment (T351-S364) activates both intrinsic (caspase-3, caspase-6, caspase-7, and caspase-9) and extrinsic (caspase-2, caspase-8, and caspase-10) caspase-dependent apoptotic signaling.

Although caspase-1 clusters phylogenetically with caspase-4 and caspase-5 and is a member of the group of the so-called inflammatory caspases, caspase-4 and caspase-5–dependent apoptotic signaling is not mediated by caspase-1 (42). Furthermore, the in vitro cleavage of pro-caspase-3 by caspase-4 and caspase-5 suggests that caspase-4 and caspase-5 might be involved in the intracellular apoptotic signaling through activated caspase-3 (4244). In the present study, either I-Trp treatment or the truncated β-tubulin transfection induced the activation of intracellular caspase-4 and caspase-5, but not caspase-1. A reasonable interpretation is that caspase-1 might be specialized to mediate extracellular inflammatory signals (45, 46), because caspase-1 expression was detectable in HEK-293 cells (47, 48). On the other hand, recent studies show that caspase-4 is localized to the surface of the endoplasmic reticulum (ER) and is involved in ER stress–induced apoptosis. Besides, it was well known that intracellular ER movement is dependent on microtubule mobilization (49, 50). Thus, caspase-4 activation may reflect the involvement of ER-dependent apoptotic signaling during β-tubulin/CCT-β complex–dependent apoptosis.

In conclusion, the interaction of iodoacetamide derivatives with the sulfhydryl group of cysteine residues provides a useful approach for us to discover a novel intracellular target for cancer therapy. By using the I-Trp-Dan compound, which was specifically incorporated into β-tubulin in HEK-293 cells, we define a novel β-tubulin/CCT-β–dependent apoptotic event mediated by intrinsic, extrinsic, and ER-dependent caspase signaling cascades. Our data also show that disrupting constitutive β-tubulin/CCT-β complexes via overexpression of truncated β-tubulin causes a more severe cell apoptosis in multidrug-resistant MES-SA/Dx5 cells compared with wild-type MES-SA cells. Because the binding site of CCT-β is conserved among β-tubulin isotypes and is located away from the vinblastine-, colchicine-, and taxol-binding sites, the modulation of β-tubulin/CCT-β complex dynamics may serve as a useful chemotherapeutic strategy for treating tubulin-binding agent–resistant tumors.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

We thank the Core Facilities for Proteomics at Academia Sinica in Taiwan for the mass technical service and Shih-Hsun Chen for his assistance in computer modeling.

1
Sharma J, Luduena RF. Use of N,N′-polymethylenebis (iodoacetamide) derivatives as probes for the detection of conformational differences in tubulin isotypes.
J Protein Chem
1994
;
13
:
165
–76.
2
Criddle AH, Geeves MA, Jeffries T. The use of actin labelled with N-(1-pyrenyl)iodoacetamide to study the interaction of actin with myosin subfragments and troponin/tropomyosin.
Biochem J
1985
;
232
:
343
–9.
3
Grazi E. Polymerization of N-(1-pyrenyl) iodoacetamide-labelled actin: the fluorescence signal is not directly proportional to the incorporation of the monomer into the polymer.
Biochem Biophys Res Commun
1985
;
128
:
1058
–63.
4
Roitelman J, Shechter I. Studies on the catalytic site of rat liver HMG-CoA reductase: interaction with CoA-thioesters and inactivation by iodoacetamide.
J Lipid Res
1989
;
30
:
97
–107.
5
Wawrzynow A, Collins JH, Coan C. An iodoacetamide spin-label selectively labels a cysteine side chain in an occluded site on the sarcoplasmic reticulum Ca(2+)-ATPase.
Biochemistry
1993
;
32
:
10803
–11.
6
Wu Y, Kwon KS, Rhee SG. Probing cellular protein targets of H2O2 with fluorescein-conjugated iodoacetamide and antibodies to fluorescein.
FEBS Lett
1998
;
440
:
111
–5.
7
Jiang JD, Denner L, Ling YH, et al. Double blockade of cell cycle at g(1)-s transition and m phase by 3-iodoacetamido benzoyl ethyl ester, a new type of tubulin ligand.
Cancer Res
2002
;
62
:
6080
–8.
8
Sullivan KF. Structure and utilization of tubulin isotypes.
Annu Rev Cell Biol
1988
;
4
:
687
–716.
9
Linse K, Mandelkow EM. The GTP-binding peptide of β-tubulin. Localization by direct photoaffinity labeling and comparison with nucleotide-binding proteins.
J Biol Chem
1988
;
263
:
15205
–10.
10
Mandelkow E, Mandelkow EM. Microtubular structure and tubulin polymerization.
Curr Opin Cell Biol
1990
;
2
:
3
–9.
11
Sharp DJ, Rogers GC, Scholey JM. Microtubule motors in mitosis.
Nature
2000
;
407
:
41
–7.
12
Sawin KE, Endow SA. Meiosis, mitosis and microtubule motors.
BioEssays
1993
;
15
:
399
–407.
13
Vyas DM, Kadow JF. Paclitaxel: a unique tubulin interacting anticancer agent.
Prog Med Chem
1995
;
32
:
289
–337.
14
Gigant B, Wang C, Ravelli RB, et al. Structural basis for the regulation of tubulin by vinblastine.
Nature
2005
;
435
:
519
–22.
15
Uppuluri S, Knipling L, Sackett DL, Wolff J. Localization of the colchicine-binding site of tubulin.
Proc Natl Acad Sci U S A
1993
;
90
:
11598
–602.
16
Bhattacharyya B, Panda D, Gupta S, Banerjee M. Anti-mitotic activity of colchicine and the structural basis for its interaction with tubulin.
Med Res Rev
2008
;
28
:
155
–83.
17
Lee KM, Cao D, Itami A, et al. Class III β-tubulin, a marker of resistance to paclitaxel, is overexpressed in pancreatic ductal adenocarcinoma and intraepithelial neoplasia.
Histopathology
2007
;
51
:
539
–46.
18
Montgomery RB, Guzman J, O'Rourke DM, Stahl WL. Expression of oncogenic epidermal growth factor receptor family kinases induces paclitaxel resistance and alters β-tubulin isotype expression.
J Biol Chem
2000
;
275
:
17358
–63.
19
Urano N, Fujiwara Y, Doki Y, et al. Clinical significance of class III β-tubulin expression and its predictive value for resistance to docetaxel-based chemotherapy in gastric cancer.
Int J Oncol
2006
;
28
:
375
–81.
20
Umezu T, Shibata K, Kajiyama H, et al. Taxol resistance among the different histological subtypes of ovarian cancer may be associated with the expression of class III β-tubulin.
Int J Gynecol Pathol
2008
;
27
:
207
–12.
21
Hari M, Wang Y, Veeraraghavan S, Cabral F. Mutations in α- and β-tubulin that stabilize microtubules and confer resistance to colcemid and vinblastine.
Mol Cancer Ther
2003
;
2
:
597
–605.
22
Yin S, Cabral F, Veeraraghavan S. Amino acid substitutions at proline 220 of β-tubulin confer resistance to paclitaxel and colcemid.
Mol Cancer Ther
2007
;
6
:
2798
–806.
23
Liou AK, Willison KR. Elucidation of the subunit orientation in CCT (chaperonin containing TCP1) from the subunit composition of CCT micro-complexes.
EMBO J
1997
;
16
:
4311
–6.
24
Won KA, Schumacher RJ, Farr GW, Horwich AL, Reed SI. Maturation of human cyclin E requires the function of eukaryotic chaperonin CCT.
Mol Cell Biol
1998
;
18
:
7584
–9.
25
Guenther MG, Yu J, Kao GD, Yen TJ, Lazar MA. Assembly of the SMRT-histone deacetylase 3 repression complex requires the TCP-1 ring complex.
Genes Dev
2002
;
16
:
3130
–5.
26
Siegers K, Bolter B, Schwarz JP, Bottcher UM, Guha S, Hartl FU. TRiC/CCT cooperates with different upstream chaperones in the folding of distinct protein classes.
EMBO J
2003
;
22
:
5230
–40.
27
Llorca O, Martin-Benito J, Gomez-Puertas P, et al. Analysis of the interaction between the eukaryotic chaperonin CCT and its substrates actin and tubulin.
J Struct Biol
2001
;
135
:
205
–18.
28
Yang JM, Chen CC. GEMDOCK: a generic evolutionary method for molecular docking.
Proteins
2004
;
55
:
288
–304.
29
Fan TJ, Han LH, Cong RS, Liang J. Caspase family proteases and apoptosis.
Acta Biochim Biophys Sin (Shanghai)
2005
;
37
:
719
–27.
30
Harker WG, Sikic BI. Multidrug (pleiotropic) resistance in doxorubicin-selected variants of the human sarcoma cell line MES-SA.
Cancer Res
1985
;
45
:
4091
–6.
31
Jiang JD, Davis AS, Middleton K, et al. 3-(Iodoacetamido)-benzoylurea: a novel cancericidal tubulin ligand that inhibits microtubule polymerization, phosphorylates bcl-2, and induces apoptosis in tumor cells.
Cancer Res
1998
;
58
:
5389
–95.
32
Yokota S, Yamamoto Y, Shimizu K, et al. Increased expression of cytosolic chaperonin CCT in human hepatocellular and colonic carcinoma.
Cell Stress Chaperones
2001
;
6
:
345
–50.
33
Saetersdal T, Greve G, Dalen H. Associations between β-tubulin and mitochondria in adult isolated heart myocytes as shown by immunofluorescence and immunoelectron microscopy.
Histochemistry
1990
;
95
:
1
–10.
34
Jung D, Filliol D, Miehe M, Rendon A. Interaction of brain mitochondria with microtubules reconstituted from brain tubulin and MAP2 or TAU.
Cell Motil Cytoskeleton
1993
;
24
:
245
–55.
35
Ling X, Cheng Q, Black JD, Li F. Forced expression of survivin-2B abrogates mitotic cells and induces mitochondria-dependent apoptosis by blockade of tubulin polymerization and modulation of Bcl-2, Bax, and survivin.
J Biol Chem
2007
;
282
:
27204
–14.
36
Yeh TS, Hsieh RH, Shen SC, et al. Nuclear βII-tubulin associates with the activated notch receptor to modulate notch signaling.
Cancer Res
2004
;
64
:
8334
–40.
37
Ferrari-Toninelli G, Bonini SA, Bettinsoli P, Uberti D, Memo M. Microtubule stabilizing effect of notch activation in primary cortical neurons.
Neuroscience
2008
;
154
:
946
–52.
38
Popova JS, Rasenick MM. Muscarinic receptor activation promotes the membrane association of tubulin for the regulation of Gq-mediated phospholipase Cβ(1) signaling.
J Neurosci
2000
;
20
:
2774
–82.
39
Mansuy V, Boireau W, Fraichard A, Schlick JL, Jouvenot M, Delage-Mourroux R. GEC1, a protein related to GABARAP, interacts with tubulin and GABA(A) receptor.
Biochem Biophys Res Commun
2004
;
325
:
639
–48.
40
Guimaraes MZ. Isoform specificity of P2X2 purinergic receptor C-terminus binding to tubulin.
Neurochem Int
2008
;
52
:
314
–20.
41
Goebel J, Forrest K, Wills-Karp M, Roszman TL. Tubulin polymerization modulates interleukin-2 receptor signal transduction in human T cells.
J Recept Signal Transduct Res
2006
;
26
:
87
–106.
42
Kamada S, Funahashi Y, Tsujimoto Y. Caspase-4 and caspase-5, members of the ICE/CED-3 family of cysteine proteases, are CrmA-inhibitable proteases.
Cell Death Differ
1997
;
4
:
473
–8.
43
Hitomi J, Katayama T, Eguchi Y, et al. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Aβ-induced cell death.
J Cell Biol
2004
;
165
:
347
–56.
44
Kim SJ, Zhang Z, Hitomi E, Lee YC, Mukherjee AB. Endoplasmic reticulum stress-induced caspase-4 activation mediates apoptosis and neurodegeneration in INCL.
Hum Mol Genet
2006
;
15
:
1826
–34.
45
Yu HB, Finlay BB. The caspase-1 inflammasome: a pilot of innate immune responses.
Cell Host Microbe
2008
;
4
:
198
–208.
46
Gemma C, Bickford PC. Interleukin-1β and caspase-1: players in the regulation of age-related cognitive dysfunction.
Rev Neurosci
2007
;
18
:
137
–48.
47
Kahns S, Kalai M, Jakobsen LD, Clark BF, Vandenabeele P, Jensen PH. Caspase-1 and caspase-8 cleave and inactivate cellular parkin.
J Biol Chem
2003
;
278
:
23376
–80.
48
Gudipaty L, Munetz J, Verhoef PA, Dubyak GR. Essential role for Ca2+ in regulation of IL-1β secretion by P2X7 nucleotide receptor in monocytes, macrophages, and HEK-293 cells.
Am J Physiol Cell Physiol
2003
;
285
:
C286
–99.
49
Aihara Y, Inoue T, Tashiro T, Okamoto K, Komiya Y, Mikoshiba K. Movement of endoplasmic reticulum in the living axon is distinct from other membranous vesicles in its rate, form, and sensitivity to microtubule inhibitors.
J Neurosci Res
2001
;
65
:
236
–46.
50
McIlvain JM, Jr., Lamb C, Dabora S, Sheetz MP. Microtubule motor-dependent formation of tubulovesicular networks from endoplasmic reticulum and Golgi membranes.
Methods Cell Biol
1993
;
39
:
227
–36.

Supplementary data