Dexamethasone Induces Apoptosis in Proliferative Chondrocytes through Activation of Caspases and Suppression of the Akt-Phosphatidylinositol 3′-Kinase Signaling Pathway (original) (raw)

Journal Article

,

1Department of Woman and Child Health (D.C., F.Z., A.S.C., L.S.), Pediatric Endocrine Unit, Astrid Lindgren Children’s Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden

*Address all correspondence and requests for reprints to: Dionisios Chrysis, Pediatric Endocrinology Unit, Q2: 08, Astrid Lindgren Children’s Hospital, Karolinska Hospital, SE-17176 Stockholm, Sweden.

Search for other works by this author on:

,

1Department of Woman and Child Health (D.C., F.Z., A.S.C., L.S.), Pediatric Endocrine Unit, Astrid Lindgren Children’s Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden

Search for other works by this author on:

,

1Department of Woman and Child Health (D.C., F.Z., A.S.C., L.S.), Pediatric Endocrine Unit, Astrid Lindgren Children’s Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden

Search for other works by this author on:

,

2Department of Biochemistry and Molecular Dentistry (T.M.), Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8525, Japan

Search for other works by this author on:

1Department of Woman and Child Health (D.C., F.Z., A.S.C., L.S.), Pediatric Endocrine Unit, Astrid Lindgren Children’s Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden

Search for other works by this author on:

Accepted:

24 November 2004

Cite

Dionisios Chrysis, Farasat Zaman, Andrei S. Chagin, Masaharu Takigawa, Lars Sävendahl, Dexamethasone Induces Apoptosis in Proliferative Chondrocytes through Activation of Caspases and Suppression of the Akt-Phosphatidylinositol 3′-Kinase Signaling Pathway, Endocrinology, Volume 146, Issue 3, 1 March 2005, Pages 1391–1397, https://doi.org/10.1210/en.2004-1152
Close

Navbar Search Filter Mobile Enter search term Search

Although glucocorticoids are known to induce apoptosis in chondrocytes, the mechanisms for this effect and the potential antiapoptotic role of IGF-I are unknown. To address this, we studied the effects of dexamethasone (Dexa) on apoptosis in the HCS-2/8 chondrocytic cell line. Dexa (25 μm) increased apoptosis (cell death ELISA) by 39% and 45% after 48 and 72 h, respectively (P < 0.01 and P < 0.05, respectively). IGF-I (100 ng/ml) decreased Dexa-induced apoptosis to levels similar to control cells. Apoptosis was associated with cleavage of poly-ADP-ribose polymerase (PARP) and α-fodrin and activation of caspases-8, -9, and -3 (Western), an effect that was counteracted when chondrocytes were cocultured with Dexa + IGF-I. Inhibitors for caspases-8, -9, and -3 (50 μm each) equally suppressed Dexa-induced apoptosis (P < 0.01). Time-response experiments showed that caspase-8 was activated earlier (at 12 h) than caspase-9 (at 36 h). We studied the phosphatidylinositol 3′-kinase (PI3K) pathway to further investigate the mechanisms of Dexa-induced apoptosis. Dexa decreased Akt phosphorylation by 93% (P < 0.001) without affecting total Akt and increased the p85α subunit 4-fold. The Akt inhibitor SH-6 (10 μm) increased apoptosis by 54% (P < 0.001). When combining Dexa with SH-6, apoptosis was not further increased, showing that Dexa-induced apoptosis is mediated through inhibition of the PI3K pathway. Addition of IGF-I to SH-6- or Dexa + SH-6-treated cells decreased apoptosis by 21.2% (P < 0.001) and 20.6% (P < 0.001), respectively. We conclude that Dexa-induced apoptosis is caspase dependent with an early activation of caspase-8. IGF-I can rescue chondrocytes from Dexa-induced apoptosis partially through the activation of other pathways than the PI3K signaling pathway. Based on our in vitro data, we speculate that in vivo treatment with glucocorticoids may diminish longitudinal growth by increasing apoptosis of proliferative growth plate chondrocytes.

THE USE OF glucocorticoids in humans and animals is well known to be associated with impaired longitudinal growth (1, 2). The observed growth retardation is accompanied by morphological changes in the growth plate, the structure responsible for longitudinal growth. It becomes thinner as a result of decreased chondrocyte proliferation and increased apoptosis, mainly of terminal hypertrophic chondrocytes (1, 35). In vivo and in vitro studies have shown that glucocorticoids decrease chondrocyte proliferation, alter their synthetic capacity, and subsequently affect the cartilage matrix (3, 4, 6).

We have previously shown in rats that in vivo administration of dexamethasone (Dexa) increases apoptosis mainly in terminally differentiated hypertrophic chondrocytes (5). In these experiments, we also observed that Dexa increased apoptosis of resting and proliferative chondrocytes even though the magnitude was less than in hypertrophic cells. A premature loss of early proliferative growth plate chondrocytes by apoptosis could diminish the growth potential and contribute to the incomplete catch-up growth often observed after long-term treatment with glucocorticoids (7, 8).

The apoptotic pathway is complex with multiple components. Important steps in this cascade are the caspase enzymes, a family of proteins responsible for the degradation of targeted cells to undergo apoptosis. Apoptosis can occur after the activation of the initiator caspase-8, which subsequently activates caspase executors (extrinsic) or after the activation of the initiator caspase-9 (intrinsic), or through interplay between these two pathways (9). In addition to these two caspase-dependent pathways, there is also caspase-independent apoptosis (10). The phosphatidylinositol 3′-kinase (PI3K)/Akt pathway is viewed as a key player for cell survival in different cell systems (for reviews, see Refs.11 and 12). Inhibition of the PI3K pathway and subsequently Akt phosphorylation seem to be important mechanisms of Dexa-induced apoptosis (13, 14). Moreover, growth retardation has been reported in knockout mice for Akt1, which demonstrates that Akt is also important for normal growth (15).

IGF-I promotes growth by increasing cell proliferation and survival. IGF-I counterbalances glucocorticoid-mediated growth retardation and increases body weight in Dexa-treated rats (16), most likely by affecting proliferation, hypertrophy, and cell survival. The antiapoptotic actions of IGF-I are well established mainly in vitro but also in vivo (1719). IGF-I exerts these actions in a variety of cells challenged to different apoptotic stimuli, mainly through the activation of the Akt signaling pathway (20).

This study was designed to investigate whether Dexa causes apoptosis in proliferative chondrocytes, the mechanisms for this action, and the potential antiapoptotic role of IGF-I. Because of technical limitations of in vivo experiments in the growth plate, we chose to study the effects of Dexa in proliferative HCS-2/8 cells. This is a well-described human chondrocytic cell line that maintains all the characteristics of chondrocytes and can be differentiated into hypertrophic chondrocytes (21).

Materials and Methods

Reagents

Dexa, IGF-1, and BSA were purchased from Sigma-Aldrich, Inc. (St. Louis, MO). The stock solution of Dexa (25 mm) was initially prepared in ethanol and diluted 1:1000 to reach a final medium concentration of 25 μm. Human recombinant IGF-I was prepared as a stock solution (100 μg/ml; in PBS with 0.1% BSA) and diluted 1:1000 (100 ng/ml in test medium). Inhibitors for caspase-3 (Z-DEVD-FMK), caspase-8 (Z-IETD-FMK), and caspase-9 (Z-LEHD-FMK) were all purchased from R&D Systems (Minneapolis, MN), dissolved in dimethylsulfoxide and diluted (1:1000) to reach a final concentration of 50 μm in the medium. Wortmannin KY12420 (PI3K inhibitor) and SH-6 (Akt inhibitor) were purchased from Calbiochem (San Diego, CA) and both used at a final concentration of 10 μm. Polyvinylidene difluoride membrane, and the ECLplus chemiluminescent system for Western immunoblots were purchased from Amersham Biosciences UK Ltd. (Buckinghamshire, UK). Protein molecular standards from Bio-Rad Laboratories (Hercules, CA).

Chondrocyte cell culture

The chondrocytic cell line, HCS-2/8 (human chondrosarcoma derived) established by Takigawa et al. (21), was maintained in DMEM mixed with F12 media (DMEM/F12) supplemented with 20% fetal bovine serum (FBS) and 20 μg/ml gentamycin in humidified air with 5% CO2. The cells were subcultivated every week and given fresh medium every 3–4 d. For Western immunoblotting, the cells were seeded in 75-cm2 tissue culture flasks (1.5 × 106 cells/flask) in DMEM/F12 medium containing 20% FBS. After 72 h, when cells were approximately 80% confluent, they were washed with 1× PBS and the medium was changed to test medium specific for each experiment. Trypsine, PBS, EDTA, FBS, and DMEM/F12 medium were all purchased from Invitrogen Life Technologies (Paisley, Scotland, UK).

Cell proliferation assay

We used the cell proliferation reagent 4-[3-(4-iodophenyl)-2(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1) kit purchased from Roche Diagnostics GmbH (Mannheim, Germany), which is a colorimetric assay for the quantification of cell proliferation and cell viability. This assay is based on a water-soluble tetrazolium salt, which is cleaved to formazan by mitochondrial enzymes. The amount of the formazan dye formed is directly correlated to the number of metabolically active cells. The cells were plated in 96-well plates (2 × 104 cells/well), and cultured for 72 h in DMEM/F12 with 20% FBS added. The cells were then washed once with 1× PBS before the medium was changed to test medium, which contained 1% FBS and for each experiment specific concentrations of effectors. After treatment, WST-1 was added, 20 μl/well (final dilution 1:10), and the reaction mixture was incubated for 1 h at 37 C before reading absorbance at 450 nm. Each experiment included three wells and was repeated four times.

Cell death detection ELISA

Apoptosis was studied with the detection and quantification of cytoplasmic histone-associated DNA fragments (mono- and oligonucleasomes) by photometric enzyme immunoassay (Cell Death Detection ELISAPLUS, Roche Diagnostics). The cells were first treated as described for the proliferation assay and then lysated (80 μl lysis buffer) and centrifuged. The supernatant was removed for analysis of cytoplasmic histone-associated DNA fragments according to the manufacturer’s instructions. In preliminary experiments, we excluded necrosis to occur under our experimental conditions by the negative determination of mono- and oligonucleasomes in the supernatant of the cell cultures. Three samples were analyzed for each experiment, which was repeated four times.

RT-PCR for type II and X collagens

Extraction of total RNA from HCS-2/8 proliferative chondrocytes treated for 24, 48, and 72 h with and without Dexa (25 μm) was performed by using QIAGEN RNeasy mini kit (VWR International AB, Stockholm, Sweden). The following pair of primers were used to generate the respective cDNAs: type II collagen, 5′-ATGACAATCTGGCTCCCAACACTGC-3′ and 5′-GACCGGCCCTATGTCCACACCGAAT-3′ giving a PCR product of 364 bp; human type X collagen, 5′-AGCCAGGGTTGCCAGGACCA-3′ and 5′-TTTTCCCACTCCAGGAGGGC-3′ (387 bp). The housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH), 5′-CAATgACCCCTTCATTgACC-3′ and 5′-CCTgCTTCACCACCTTCTTg-3′ (694 bp), was used as control to normalize the data. RT-PCR was performed as it has been previously described (22). The amplification conditions for type II and type X collagen were as follows: 96 C for 5 min, 96 C for 30 sec with 35 cycles of denaturation, annealing for 1 min at 55 C (30 sec at 54 C, 30 cycles for GAPDH) extension for 1 min at 72 C (1 min at 72 C for GAPDH) and final extension for 5 min at 72 C. PCR products were separated by electrophoresis in a 2% agarose gel with ethidium bromide (15 μg/10 ml). RNA from HCS-2/8 chondrocytes, 18 d in culture under differentiation conditions was used as a positive control for type X collagen.

Western immunoblotting

After each treatment, both floating and adherent cells were lysated in RIPA buffer [150 mm NaCl, 10 mm Tris (pH 7.2), 0.1% sodium dodecyl sulfate, 1.0% Triton X-100, 1% deoxycholate, and 5 mm EDTA] and the protein concentration was measured by the Bradford protein assay (Bio-Rad Laboratories AB, Sundbyberg, Sweden). Proteins were separated on 7.5% Tris-HCl or 4–20% gradient acrylamide gels (Bio-Rad Laboratories). Proteins were transferred on Hybond-P polyvinylidene difluoride-transfer membrane. The membranes were blocked with 5% Blotto, nonfat dry milk (Santa Cruz Biotechnology Inc., Santa Cruz, CA) in Tris-buffered saline with 0.1% Tween 20, for 1 h at room temperature. The membranes were probed overnight at 4 C (gentle shaking) with primary antibodies against PARP (1:1000 dilution), caspase-3 (1:1000 dilution), Akt 1/2 (1:1500 dilution) or the p85α subunit of PI3K (1:1000 dilution), all purchased from Santa Cruz Biotechnology Inc. Additional primary antibodies including caspase-8 (1:1000 dilution), human-specific caspase-9 (1:500 dilution), α-fodrin (Asp 1185, 1:500 dilution), and phospho-Akt (Ser-473, 1:1000 dilution) were purchased from Cell Signaling Technology (Beverly, MA). The blots were probed with the corresponding secondary antibody, goat-antirabbit Ig-horseradish peroxidase (HRP) (Transduction Laboratories, Lexington, KY, 1:10000 or 1:15000 dilutions), goat-antimouse IgG-HRP (1:10000), or rabbit-antigoat IgG-HRP (1:5000 dilution), all purchased from Santa Cruz Biotechnology, Inc. The ECLplus Western blotting detection system was used according to the manufacturer’s instructions. The resulting bands were confirmed by comparing the size of the protein in the cell extract with known molecular weight markers. The antigen-antibody complexes were then detected by chemiluminescence. After films had been developed, blots were stained with Coomassie Blue to ensure equal loading of total protein. Each experiment was repeated at least three times.

Statistical analysis

Results are presented as the mean ± sem. Differences between the groups were tested by one-way ANOVA. All P values were calculated using the Newman-Keuls posttest. A P value <0.05 was considered significant.

Results

Effect of Dexa on chondrocyte proliferation and apoptosis

HCS-2/8 chondrocytes were cultured under semiconfluent conditions for 24, 48, and 72 h in the presence of 25 μm Dexa. The treatment with Dexa decreased cell proliferation rate after 24 h (60.4 ± 10.90%, P < 0.05 _vs._ control), 48 h (61.30 ± 0.65%, _P_ < 0.01 _vs._ control), and 72 h (63.70 ± 1.85%, _P_ < 0.001 _vs._ control). Apoptosis was significantly increased after treatment with Dexa (25 μm) for 48 h (139.5 ± 8.90%; _P_ < 0.01 _vs._ control) and 72 h (145.1 ± 15.47%; _P_ < 0.05 _vs._ control) but not after 24 h (114.1 ± 6.24%; _P_ > 0.05 vs. control; Fig. 1). The Dexa-induced decrease in chondrocyte proliferation and increase in apoptosis showed clear dose-dependencies (Fig. 2, A and B).

HCS-2/8 cells were treated with 25 μm Dexa for 24, 48, and 72 h. Apoptosis was measured with cell death ELISA, and the levels are expressed as percent of control-treated cells. Dexa increased apoptosis after 48 h (**, P < 0.01) by 39.5%, and after 72 h (*, P < 0.05) by 45% when compared with the control group.

Fig. 1.

HCS-2/8 cells were treated with 25 μm Dexa for 24, 48, and 72 h. Apoptosis was measured with cell death ELISA, and the levels are expressed as percent of control-treated cells. Dexa increased apoptosis after 48 h (**, P < 0.01) by 39.5%, and after 72 h (*, P < 0.05) by 45% when compared with the control group.

Dose-response effects of Dexa (72 h) on cell proliferation (A) and apoptosis (B). Cell proliferation was measured with the WST-1 cell proliferation assay and apoptosis with a cell death ELISA; see Materials and Methods. Results are expressed as percent of control-treated cells. Dexa had a clear dose-dependent inhibitory effect on cell viability (A) and a dose-dependent proapoptotic effect (B). *, P < 0.05; **, P < 0.01 vs. control, respectively.

Fig. 2.

Dose-response effects of Dexa (72 h) on cell proliferation (A) and apoptosis (B). Cell proliferation was measured with the WST-1 cell proliferation assay and apoptosis with a cell death ELISA; see Materials and Methods. Results are expressed as percent of control-treated cells. Dexa had a clear dose-dependent inhibitory effect on cell viability (A) and a dose-dependent proapoptotic effect (B). *, P < 0.05; **, P < 0.01 vs. control, respectively.

To rule out whether our experimental conditions may influence the differentiation process of HCS-2/8 cells, we analyzed by PCR the expression of type II and X collagen after 24, 48, and 72 h of culture. We found that type II collagen was expressed at all time points and that type X collagen, a marker of differentiating chondrocytes, was not expressed at any time point for both control and Dexa-treated cells. These data indicate that Dexa treatment did not influence the rate of chondrocyte differentiation when exposed for up to 72 h. Control experiments showed that HCS-2/8 chondrocytes can express type X collagen when cultured under conditions promoting cell differentiation for 18 d (data not shown).

We then investigated whether IGF-I can counteract the antiproliferative and apoptotic actions of Dexa in cultured HCS-2/8 chondrocytes. Initial experiments with IGF-I only (100 ng/ml) showed that cell proliferation was stimulated (112.5 ± 3.4% of control; P < 0.05 _vs._ control; Fig. 3A) and spontaneous apotosis was decreased (85.0 ± 5.8% of control; _P_ < 0.05 _vs._ control; Fig. 3B). When IGF-I (100 ng/ml) was combined with Dexa (25 μm), the antiproliferative effect of Dexa was partially counteracted (_P_ < 0.05 _vs._ Dexa; Fig. 3A). Whereas cells cultured with Dexa only demonstrated increased apoptosis (134.8 ± 1.28%; _P_ < 0.001 _vs._ control), those cocultured with IGF-I plus Dexa had less apoptosis (110.2 ± 4.42% of control cells; _P_ < 0.001 _vs._ Dexa; _P_ > 0.05 vs. control; Fig. 3B).

Effects of IGF-I on HCS-2/8 cell proliferation (WST-1 cell proliferation assay) and apoptosis (cell death ELISA) after 72 h of cotreatment with or without Dexa. A, IGF-I (100 ng/ml) alone increased basal cell proliferation by 13% (*, P < 0.05 vs. control). The addition of IGF-I to Dexa (25 μm)-treated cells improved cell proliferation by 17% (*, P < 0.05 vs. Dexa), but still cell viability was significantly less than in control (**, P < 0.01). B, IGF-I alone decreased basal apoptosis by 15% (*, P < 0.05 vs. control). Cotreatment with Dexa and IGF-I decreased apoptosis by 18% (***, P < 0.001 vs. Dexa) and reached levels similar to control (P > 0.05), whereas Dexa increased apoptosis by 35% (***, P < 0.001 vs. control).

Fig. 3.

Effects of IGF-I on HCS-2/8 cell proliferation (WST-1 cell proliferation assay) and apoptosis (cell death ELISA) after 72 h of cotreatment with or without Dexa. A, IGF-I (100 ng/ml) alone increased basal cell proliferation by 13% (*, P < 0.05 _vs._ control). The addition of IGF-I to Dexa (25 μm)-treated cells improved cell proliferation by 17% (*, _P_ < 0.05 _vs._ Dexa), but still cell viability was significantly less than in control (**, _P_ < 0.01). B, IGF-I alone decreased basal apoptosis by 15% (*, _P_ < 0.05 _vs._ control). Cotreatment with Dexa and IGF-I decreased apoptosis by 18% (***, _P_ < 0.001 _vs._ Dexa) and reached levels similar to control (_P_ > 0.05), whereas Dexa increased apoptosis by 35% (***, P < 0.001 vs. control).

To confirm our apoptosis data (cell death detection ELISA), we studied the proteins PARP and α-fodrin, both well known to be cleaved during caspase-dependent apoptosis. Indeed, Dexa-induced apoptosis was accompanied by a 2.5-fold increase in cleaved PARP (28.0 ± 1.3 and 11.0 ± 0.5 optical units in Dexa and control groups, respectively; P < 0.001 vs. control; Fig. 4A) and a 3-fold increase in cleaved α-fodrin (40.0 ± 2.4 and 13.0 ± 0.6 optical units in Dexa and control groups, respectively; P < 0.001 vs. control; Fig. 4B). In contrast, IGF-I prevented Dexa-induced cleavages of PARP and α-fodrin, and the levels were similar as in control-treated cells (Fig. 4, A and B).

Cells were treated for 72 h with Dexa (25 μm), IGF-I (100 ng/ml), or the combination of Dexa and IGF-I. A, Western immunoblots revealed that Dexa induced cleavage of PARP, whereas the addition of IGF-I restored it. B, The same results were obtained with another protein substrate for caspases, fodrin-α. C, Representative Western blots for caspases-8, -9, and -3. The cleaved products of all these caspases were increased after treatment with Dexa and decreased by the addition of IGF-I.

Fig. 4.

Cells were treated for 72 h with Dexa (25 μm), IGF-I (100 ng/ml), or the combination of Dexa and IGF-I. A, Western immunoblots revealed that Dexa induced cleavage of PARP, whereas the addition of IGF-I restored it. B, The same results were obtained with another protein substrate for caspases, fodrin-α. C, Representative Western blots for caspases-8, -9, and -3. The cleaved products of all these caspases were increased after treatment with Dexa and decreased by the addition of IGF-I.

Dexa activates the caspase cascade

We then studied whether Dexa-induced apoptosis is associated with caspase activation (Western immunoblotting). We found that Dexa (25 μm; 72 h incubation) increased cleavage of caspase-3, whereas significantly less cleaved caspase-3 was detected in cells treated with Dexa + IGF-I (16.0 ± 0.9 vs. 4.0 ± 0.2 optical units; P < 0.001; Fig. 4C). Cells treated with IGF-I (100 ng/ml) alone had no detectable cleaved caspsase-3 (Fig. 4C). Our next step was to analyze the activation of the caspase initiators, caspases-8 and -9. We found that Dexa (25 μm; 72 h) activated both these caspase initiators and that cotreatment with IGF-I (100 ng/ml) decreased the levels to what was detected in control cells. The 41-kDa fragment of caspase-8 was increased 3-fold in the Dexa group when compared with control (34.0 ± 1.5 and 11.0 ± 0.8 optical units, respectively; P < 0.001; Fig. 4C) and the addition of IGF-I returned it to control levels. Similarly, the cleaved product of caspase-9 (37-kDa fragment) was increased 2.7-fold by Dexa and cotreatment with IGF-I significantly decreased cleaved caspase-9 (100.0 ± 4.8 and 37.0 ± 2.9 optical units, respectively; P < 0.001) to levels similar as in control cells (Fig. 4C).

We performed experiments with caspase-inhibitors to further confirm that Dexa-induced apoptosis is caspase dependent (Fig. 5). Cells treated with Dexa alone (25 μm, 72 h), had increased apoptosis compared with control-treated cells (151.0 ± 4.1% of control, P < 0.001). On the other hand, cells cotreated with Dexa and caspase-3 inhibitor had less apoptosis (72.7 ± 4.1% of control; _P_ < 0.001 _vs._ Dexa, _P_ > 0.05 vs. control). Similar results were obtained with the inhibitors for caspase-8 (100.2 ± 17.5% of control, P < 0.01 vs. Dexa), and caspase-9 (108.7 ± 13.9% of control, P < 0.01 vs. Dexa). None of the caspase inhibitors affected cell proliferation (data not shown). When the HCS-2/8 chondrocytes were treated with caspase inhibitors alone, apoptosis was slightly decreased when compared with control-treated cells (caspase-8 inhibitor: 71.6 ± 3.1%, caspase-9 inhibitor: 87.9 ± 5.4%, and caspase-3 inhibitor: 79.1 ± 10.5%, P < 0.01 vs. control).

Cells were treated for 72 h with Dexa (25 μm) alone, with Dexa and caspase inhibitors (50 μm; filled bars), or with each caspase inhibitor alone (hatched bars). Dexa alone increased apoptosis by 51.3%, whereas all caspase inhibitors in coculture with Dexa decreased apoptosis to control levels (**, P < 0.01 vs. Dexa; ***, P < 0.001 vs. Dexa). Caspase inhibitors alone slightly decreased spontaneous apoptosis compared with control-treated cells (P < 0.01).

Fig. 5.

Cells were treated for 72 h with Dexa (25 μm) alone, with Dexa and caspase inhibitors (50 μm; filled bars), or with each caspase inhibitor alone (hatched bars). Dexa alone increased apoptosis by 51.3%, whereas all caspase inhibitors in coculture with Dexa decreased apoptosis to control levels (**, P < 0.01 vs. Dexa; ***, P < 0.001 vs. Dexa). Caspase inhibitors alone slightly decreased spontaneous apoptosis compared with control-treated cells (P < 0.01).

To rule out which caspase-initiator is activated first, we determined the levels of caspase-8 and -9 in HCS-2/8 chondrocytes treated with Dexa (25 μm) for 0, 12, 24, 36, or 48 h. As seen in Fig. 6, these studies showed a clear time dependency where caspase-8 was activated earlier (12 h) than caspase-9 (36 h).

To assess which caspase initiator is activated first, HCS 2/8 cells were cultured with 25 μm Dexa for 12, 24, 36, and 48 h. The control was untreated cells at time point 0. Western blots were performed at all time points for caspase-8 (A) and caspase-9 (B). Cleaved products of caspase-8 (p41 kDa) were detected as early as after 12 h, whereas those from caspase-9 (p37 kDa) were detected first after 36 h.

Fig. 6.

To assess which caspase initiator is activated first, HCS 2/8 cells were cultured with 25 μm Dexa for 12, 24, 36, and 48 h. The control was untreated cells at time point 0. Western blots were performed at all time points for caspase-8 (A) and caspase-9 (B). Cleaved products of caspase-8 (p41 kDa) were detected as early as after 12 h, whereas those from caspase-9 (p37 kDa) were detected first after 36 h.

Inhibition of Akt phosphorylation by Dexa induces apoptosis

The PI3K/Akt pathway is considered as a key component of cell survival in different cell systems. To investigate whether the PI3K pathway is important for the apoptotic actions of Dexa in chondrocytes, Akt phosphorylation was studied. Phosphorylated Akt was significantly decreased in Dexa- (7.0 ± 2.9% of control, P < 0.001 vs. control) and Dexa/IGF-I-treated cells (26.7 ± 2.9% of control, P < 0.001 vs. control, P < 0.01 by t test vs. Dexa), whereas total Akt did not change (Fig. 7A). IGF-I alone increased phosphorylated Akt by 23% (P < 0.01 vs. control). Moreover, the p85α subunit of PI3K, well known to inhibit Akt phosphorylation, was increased more than 4-fold after treatment with Dexa (21.0 ± 3.4 and 5.0 ± 1.0 optical units in Dexa and control groups, respectively; P < 0.01).

Dexa caused apoptosis in HCS-2/8 cells through inhibition of Akt phosphorylation. A, Cells were treated for 72 h with 25 μm Dexa, with Dexa plus IGF-I (100 ng/ml), or IGF-I alone. Western immunoblots revealed no changes of total Akt. In contrast, Dexa inhibited phosphorylation of Akt (***, P < 0.001 vs. control) and the addition of IGF-I had only a weak effect (***, P < 0.001 vs. control, **, P < 0.01 by t test vs. Dexa). IGF-I alone, increased phosphorylation of Akt compared with control cells (**, P < 0.01 vs. control). B, The PI3K pathway was blocked with the specific inhibitor of Akt phosphorylation SH-6 (10 μm) and apoptosis was studied by cell death ELISA. SH-6 caused a significant increase in apoptosis (P < 0.001 vs. control) and cotreatment with IGF-I (100 ng/ml) decreased apoptosis (***, P < 0.001 vs. SH-6). On the other hand, addition of Dexa (25 μm) to SH-6-treated cells did not change apoptosis (P > 0.05 vs. SH-6), but again IGF-I added to SH-6/Dexa-treated cells decreased apoptosis (***, P < 0.001 vs. SH-6/Dexa) to a similar level as in cells treated with SH-6/IGF-I (P > 0.05). Dexa alone increased apoptosis to the same degree as SH-6- or SH-6/Dexa-treated cells. IGF-I alone (100 ng/ml) decreased spontaneous apoptosis compared with control cells (**, P < 0.01 vs. control).

Fig. 7.

Dexa caused apoptosis in HCS-2/8 cells through inhibition of Akt phosphorylation. A, Cells were treated for 72 h with 25 μm Dexa, with Dexa plus IGF-I (100 ng/ml), or IGF-I alone. Western immunoblots revealed no changes of total Akt. In contrast, Dexa inhibited phosphorylation of Akt (***, P < 0.001 _vs._ control) and the addition of IGF-I had only a weak effect (***, _P_ < 0.001 _vs._ control, **, _P_ < 0.01 by _t_ test _vs._ Dexa). IGF-I alone, increased phosphorylation of Akt compared with control cells (**, _P_ < 0.01 _vs._ control). B, The PI3K pathway was blocked with the specific inhibitor of Akt phosphorylation SH-6 (10 μm) and apoptosis was studied by cell death ELISA. SH-6 caused a significant increase in apoptosis (_P_ < 0.001 _vs._ control) and cotreatment with IGF-I (100 ng/ml) decreased apoptosis (***, _P_ < 0.001 _vs._ SH-6). On the other hand, addition of Dexa (25 μm) to SH-6-treated cells did not change apoptosis (_P_ > 0.05 vs. SH-6), but again IGF-I added to SH-6/Dexa-treated cells decreased apoptosis (***, P < 0.001 _vs._ SH-6/Dexa) to a similar level as in cells treated with SH-6/IGF-I (_P_ > 0.05). Dexa alone increased apoptosis to the same degree as SH-6- or SH-6/Dexa-treated cells. IGF-I alone (100 ng/ml) decreased spontaneous apoptosis compared with control cells (**, P < 0.01 vs. control).

Our next step was to study apoptosis after blocking Akt phosphorylation with the specific Akt inhibitor SH-6. Under our experimental conditions, phosphorylation of Akt was almost abolished when cells were treated with SH-6, whereas IGF-I weakly increased Akt phosphorylation (Western immunoblotting; data not shown). As seen in Fig. 7B, SH-6 alone (10 μm) increased apoptosis compared with control-treated cells (154.0 ± 0.7% of control, P < 0.001), an effect that was counteracted when SH-6 was combined with IGF-I (121.2 ± 0.7% of control, _P_ < 0.001 _vs._ SH-6). On the other hand, the addition of Dexa to SH-6-treated cells had no further effect on apoptosis (156.6 ± 1.4% of control, _P_ > 0.05 vs. SH-6; Fig. 7B). In contrast, the addition of IGF-I to SH-6/Dexa-treated cells decreased apoptosis compared with SH-6/Dexa (124.2 ± 1.4% of control; P < 0.001 vs. SH-6/Dexa; Fig. 7B), but still apoptosis remained higher than in control (P < 0.001). As expected, Dexa alone increased apoptosis (151.6 ± 4.6%; P < 0.001 vs. control), whereas IGF-I decreased apoptosis (82.6 ± 3.1%; P < 0.01 vs. control).

We used another PI3K inhibitor, Wortmannin, to confirm that IGF-I is able to exert antiapoptotic actions despite the Akt pathway had been blocked. After 72 h in culture with 10 μm Wortmannin, apoptosis was significantly increased compared with control-treated cells (327.0 ± 3.2% of control; P < 0.001 vs. control), an effect that was partially prevented by IGF-I (239.5 ± 5.7% of control; P < 0.001 vs. Wortmannin) but still remained higher than in control (P < 0.001). As expected, total Akt was not changed, but phosphorylation of Akt was almost abolished by Wortmannin and partially restored by IGF-I (data not shown).

Discussion

We here show that Dexa not only decreases chondrocyte proliferation but also increases apoptosis, whereas IGF-I as a growth-promoting factor counteracts both these effects. The observed apoptosis is caspase dependent and includes activation of both caspase initiators: caspase-8 and -9. We also demonstrate that inhibition of Akt phosphorylation is a crucial step involved in Dexa-induced apoptosis.

Or data show that Dexa induces apoptosis in the human HCS-2/8 chondrocytic cell line in a dose-dependent manner. In addition, this was not an early effect because increased apoptosis was detected first after 48 h treatment, as it has been observed in bovine articular chondrocytes (23). Moreover, human growth plate proliferative chondrocytes are known to express caspase-8 (24), and rat proliferative chondrocytes express caspases-3 and -6 (25). Altogether, this indicates that these cells have the machinery for apoptosis and have the potential to activate it after being exposed to a suitable stimulus. Loss of proliferative chondrocytes by apoptosis after treatment with glucocorticoids could have important clinical significance because it may diminish growth potential in children. This premature loss of proliferative chondrocytes could account for the incomplete catch-up growth and diminished final height which has been reported after glucocorticoid excess (8). In this study, we focused on a pharmacological dose of Dexa (25 μm), not only because it reflects the clinical use of glucocorticoids and therefore their side effects, but also because Dexa has been reported to have anabolic effects at lower concentrations (26). Although our present in vitro data are in correlation with other in vitro (23) and in vivo studies (5), one should be careful when drawing definitive conclusions from in vitro data and extrapolating these to human disorders.

We here show that apoptosis is caspase dependent in HCS-2/8 chondrocytes because the caspase initiators, caspase-8 and -9, and the executor caspase-3 are all activated. This is further supported by our finding that coincubation with each caspase inhibitor totally blocked Dexa-induced apoptosis. Furthermore, our experiments reveal that caspase-8 is activated earlier than caspase-9. This suggests that caspase-9 is activated through caspase-8 as it has been reported in other cell systems after Dexa treatment (27). The delay in caspase-9 activation could be because it requires the cleavage of Bid by caspase-8, which then translocates to the mitochondria and there releases cytochrome c (28).

Several mechanisms have been reported by which Dexa causes apoptosis in different cell lines, including inhibition of the PI3K pathway and decreased phosphorylation of Akt (13, 14, 2931). Our experiments clearly show that Dexa inhibits Akt phosphorylation in HCS-2/8 chondrocytes. In addition, when we blocked the Akt signaling pathway, Dexa had no additive effect on apoptosis. The p85α subunit, which inhibits the catalytic activity of the PI3K, has previously been reported to be increased by Dexa (14, 32). We investigated weather the abrogation of Akt phosphorylation is a result of an increase in the p85α subunit. Indeed, we found that Dexa causes a dramatic increase in the p85α subunit, thus providing a mechanism for Dexa-induced inhibition of the PI3K signaling pathway and subsequent inhibition of Akt phosphorylation.

IGF-I has antiapoptotic effects in many cell types in response to different apoptotic stimuli mainly through the activation of the PI3K/Akt pathway (17, 20). Therefore, we hypothesized that IGF-I can rescue chondrocytes from Dexa-induced apoptosis. Our data demonstrate that IGF-I counteracts the antiproliferative and proapoptotic actions of Dexa in the human chondrocytic cell line HCS-2/8. The IGF-I prevention of Dexa-induced apoptosis was associated with restoration of PARP and α-fodrin cleavage, which are products of caspase activation, and abrogation of caspases-8, -9, and -3 activation. Several studies have shown that the PI3K signaling pathway and subsequently the phosphorylation of Akt are very important targets for the antiapoptotic actions of IGF-I (17, 20, 33, 34). The involvement of other pathways facilitating the antiapoptotic actions of IGF-I have been demonstrated also in other cell types (3537). Our findings in HCS-2/8 chondrocytes clearly demonstrate that IGF-I protects chondrocytes from Dexa-induced apoptosis. This effect is most likely partially mediated through other pathways than the PI3K/Akt signaling pathway because, when we blocked this pathway with SH-6, IGF-I was able to significantly reduce apoptosis. On the other hand, we observed some restoration of Akt phosphorylation after IGF-I cotreatment and Wortmannin significantly reduced the antiapoptotic actions of IGF-I, suggesting that the PI3K/Akt signaling pathway may mediate some of the antiapoptotic actions of IGF-I.

In summary, our results show that Dexa decreases not only proliferation of chondrocytes but also increases apoptosis through inhibition of Akt phosphorylation and therefore inhibition of the PI3K/Akt signaling pathway. IGF-I counteracts these actions of Dexa resulting in increased chondrocyte proliferation and decreased apoptosis. Based on our data in HCS chondrocytes, we speculate that treatment with IGF-I or GH may rescue growth plate chondrocytes from apoptosis and thereby prevent loss of growth potential during long-term treatment with glucocorticoids. Further in vivo studies are needed to confirm whether this is the case.

Abbreviations

References

1

Altman

A

,

Hochberg

Z

,

Silbermann

M

1992

Interactions between growth hormone and dexamethasone in skeletal growth and bone structure of the young mouse.

Calcif Tissue Int

51

:

298

304

2

Allen

DB

1996

Growth suppression by glucocorticoids therapy.

Endocrinol Metab Clin North Am

25

:

699

717

3

Annefeld

M

1992

Changes in the rat epiphyseal cartilage after treatment with dexamethasone and glycosaminoglycan-peptide complex.

Pathol Res Pract

188

:

649

652

4

Kember

NF

,

Walker

KV

1971

Control of bone growth in rats.

Nature

229

:

428

429

5

Chrysis

D

,

Ritzen

EM

,

Sävendahl

L

2003

Growth retardation induced by dexamethasone is associated with increased apoptosis of the growth plate chondrocytes.

J Endocrinol

176

:

331

337

6

Dearden

LC

,

Mosier

HD

,

Brundage Jr

M

,

Thai

C

,

Jansons

R

1986

The effects of different steroids on costal and epiphyseal cartilage of fetal and adult rats.

Cell Tissue Res

246

:

401

412

7

Baron

J

,

Klein

KO

,

Colli

MJ

,

Yanovski

JA

,

Novosad

JA

,

Bacher

JD

,

Cutler Jr

GB

1994

Catch-up growth after glucocorticoid excess: a mechanism intrinsic to the growth plate.

Endocrinology

135

:

1367

1371

8

Magiakou

MA

,

Mastorakos

G

,

Chrousos

GP

1994

Final stature in patients with endogenous Cushing’s syndrome.

J Clin Endocrinol Metab

79

:

1082

1085

9

Barnhart

BC

,

Alappat

EC

,

Peter

ME

2003

The CD95 typeI/type II model.

Sem Immunol

15

:

185

193

10

Lockshin

RA

,

Zakeri

Z

2004

Caspase-independent cell death?

Oncogene

23

:

2766

2773

11

Datta

SR

,

Brunet

A

,

Greenberg

ME

1999

Cellular survival: a play in three Akts.

Genes Dev

13

:

2905

2927

12

Kandel

ES

,

Hay

N

1999

The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB.

Exp Cell Res

253

:

210

229

13

Leis

H

,

Page

A

,

Ramirez

A

,

Bravo

A

,

Segrelles

C

,

Paramio

J

,

Barettino

D

,

Jorcano

JL

,

Perez

P

2004

Glucocorticoid receptor counteracts tumorigenic activity of Akt in skin through interference with the phosphatidylinositol 3-kinase signailing pathway.

Mol Endocrinol

18

:

303

311

14

Singleton

JR

,

Baker

BL

,

Thorburn

A

2000

Dexamethasone inhibits insulin-like growth factor signaling and potentiates myoblast apoptosis.

Endocrinology

141

:

2945

2950

15

Chen

WS

,

Xu

PZ

,

Gottlob

K

,

Chen

ML

,

Sokol

K

,

Shiyanova

T

,

Roninson

I

,

Weng

W

,

Suzuki

R

,

Tobe

K

,

Kadowaki

T

,

Hay

N

2001

Growth retardation and increased apoptosis in mice with homozygous disruption of the akt1 gene.

Genes Dev

15

:

2203

2208

16

Tomas

FM

,

Knowles

SE

,

Owens

PC

,

Chandler

CS

,

Francis

GL

,

Read

LC

,

Ballard

FJ

1992

Insulin-like growth factor-I (IGF-I) and especially IGF-I variants are anabolic in dexamethasone-treated rats.

Biochem J

282

:

91

97

17

Butt

AJ

,

Firth

SM

,

Baxter

RC

1999

The IGF axis and programmed cell death.

Immunol Cell Biol

77

:

256

262

18

Chrysis

D

,

Calikoglu

AS

,

Ye

P

,

D’Ercole

AJ

2001

Insulin-like growth factor-I overexpression attenuates cerebellar apoptosis by altering the expression of Bcl family proteins in a developmentally specific manner.

J Neurosci

21

:

1481

1489

19

Wilkins

HR

,

Ohneda

K

,

Keku

TO

,

D’Ercole

AJ

,

Fuller

CR

,

Williams

KL

,

Lund

PK

2002

Reduction of spontaneous and irradiation-induced apoptosis in small intestine of IGF-I transgenic mice

.

Am J Physiol

283

:

G457

G464

20

Vincent

AM

,

Feldman

EL

2002

Control of cell survival by IGF signaling pathways.

Growth Horm IGF Res 2002

12

:

193

197

21

Takigawa,

M

,

Tajima

K

,

Pan

HO

,

Enomoto

M

,

Kinoshita

A

,

Suzuki

F

,

Takano

Y

,

Mori

Y

1989

Establishment of a clonal human chondrosarcoma cell line with cartilage phenotypes.

Cancer Res

49

:

3996

4002

22

Martensson

K

,

Chrysis

D

,

Savendahl

L

2004

Interleukin-1β and TNF-α act in synergy to inhibit longitudinal growth in fetal rat metatarsal bones.

J Bone Miner Res

19

:

1805

1812

23

Van Offel

JF

,

Schuerwegh

AJ

,

Bridts

CH

,

Stevens

WJ

,

De Clerck

LS

2002

Effect of biphosphonates on viability, proliferation, and dexamethasone-induced apoptosis in articular chondrocytes.

Ann Rheum Dis

61

:

925

928

24

Cetin

E

,

Girsch

W

,

Brand

G

,

Thurnher

D

,

Cetin

EM

,

Trieb

K

2004

Distinct expression of APO-1/Fas and caspase-8 in the human growth plate.

Calcif Tissue Int

74

:

181

186

25

Chrysis

D

,

Nilsson

O

,

Ritzen

EM

,

Savendahl

L

2002

Apoptosis is developmentally regulated in rat growth plate.

Endocrine

18

:

271

278

26

Kubota

S

,

Moritani

NH

,

Kawaki

H

,

Mimura

H

,

Minato

M

Takigawa

M

2003

Transcriptional induction of connective tissue growth factor/hypertrophic chondrocyte specific 24 (CTGF/Hcs24) gene by dexamethasone in human chondrocytic cells.

Bone

33

:

694

702

27

Marchetti

MC

,

Di Marco

B

,

Cifone

G

,

Migliorati

G

,

Riccardi

C

2003

Dexamethasone-induced apoptosis of thymocytes: role of glucocorticoids receptor-associated Src kinase and caspase-8 activation.

Blood

101

:

585

593

28

Li

H

,

Zhu

H

,

Xu

CJ

,

Yuan

J

1998

Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis.

Cell

94

:

491

501

29

Distelhorst

CW

2002

Recent insights into the mechanism of glucocorticoids-induced apoptosis.

Cell Death Differ

9

:

6

19

30

Ogawa

M

,

Nishiura

T

,

Oritani

K

,

Yoshida

H

,

Yoshimura

M

,

Okajima

Y

,

Ishikawa

J

,

Hashimoto

K

,

Matsumura

I

,

Tomiyama

Y

,

Matsuzawa

Y

2002

Cytokines prevent dexamethasone-induced apoptosis via the activation of mitogen-activated protein kinase and phosphatidylinositol 3-kinase pathways in a new multiple myeloma cell line.

Cancer Res

60

:

4262

4269

31

Hsu

JH

,

Shi

Y

,

Hu

L

,

Fisher

M

,

Franke

TF

,

Lichtenstein

A

2002

Role of the AKT kinase in expansion of multiple myeloma clones: effects on cytokine-dependent proliferative and survival responses.

Oncogene

21

:

1391

1400

32

Giorgino

F

,

Pedrini

MT

,

Matera

L

,

Smith

RJ

1997

Specific increase in p85α expression in response to dexamethasone is associated with inhibition of insulin-like growth factor I stimulated phosphatidylinositol 3-kinase activity in cultured muscle cells.

J Biol Chem

272

:

7455

7463

.

33

Dudek

H

,

Datta

SR

,

Franke

TF

,

Birnbaum

MJ

,

Yao

R

,

Cooper

GM

,

Segal

RA

,

Kaplan

DR

,

Greenberg

ME

1997

Regulation of neuronal survival by the serine-threonine protein kinase Akt.

Science

275

:

661

665

34

Oh

CD

,

Chun

JS

2003

Signaling mechanisms leading to the regulation of differentiation and apoptosis of articular chondrocytes by insulin-like growth factor-1.

J Biol Chem

278

:

36563

36571

35

Parizzas

M

,

Saltiel

AR

,

LeRoith

D

1997

Insulin-like growth factor I inhibits apoptosis using the phosphatidylinositol 3′-kinase and mitogen-activated protein kinase pathways.

J Biol Chem

272

:

154

161

36

Grey

A

,

Chen

Q

,

Xu

X

,

Callon

K

,

Cornish

J

2003

Parallel phosphatidylinositol-3 kinase and p42/44 mitogen-activated protein kinase signaling pathways subserve the mitogenic and antiapoptotic actions of insulin-like growth factor I in osteoblastic cells.

Endocrinology

144

:

4886

4893

37

Thimmaiah

KN

,

Easton

J

,

Huang

S

,

Veverka

KA

,

Germain

GS

,

Harwood

FC

,

Houghton

PJ

2003

Insulin-like growth factor I-mediated protection from rapamycin-induced apoptosis is independent of Ras-Erk1-Erk2 and phosphatidylinositol 3′-kinase-Akt signaling pathways.

Cancer Res

63

:

364

374

Copyright © 2005 by The Endocrine Society

Citations

Views

Altmetric

Metrics

Total Views 1,809

1,293 Pageviews

516 PDF Downloads

Since 2/1/2017

Month: Total Views:
February 2017 7
March 2017 2
April 2017 2
May 2017 7
June 2017 1
July 2017 7
August 2017 10
September 2017 4
October 2017 2
November 2017 4
December 2017 18
January 2018 19
February 2018 9
March 2018 14
April 2018 19
May 2018 13
June 2018 19
July 2018 15
August 2018 15
September 2018 15
October 2018 7
November 2018 13
December 2018 17
January 2019 11
February 2019 14
March 2019 32
April 2019 29
May 2019 30
June 2019 15
July 2019 21
August 2019 22
September 2019 21
October 2019 45
November 2019 16
December 2019 24
January 2020 25
February 2020 16
March 2020 25
April 2020 19
May 2020 8
June 2020 18
July 2020 17
August 2020 24
September 2020 18
October 2020 24
November 2020 41
December 2020 30
January 2021 23
February 2021 19
March 2021 33
April 2021 21
May 2021 22
June 2021 19
July 2021 16
August 2021 11
September 2021 18
October 2021 17
November 2021 20
December 2021 15
January 2022 17
February 2022 17
March 2022 15
April 2022 26
May 2022 22
June 2022 12
July 2022 29
August 2022 27
September 2022 20
October 2022 25
November 2022 26
December 2022 22
January 2023 15
February 2023 5
March 2023 30
April 2023 33
May 2023 30
June 2023 41
July 2023 25
August 2023 18
September 2023 23
October 2023 25
November 2023 36
December 2023 20
January 2024 21
February 2024 14
March 2024 30
April 2024 41
May 2024 29
June 2024 27
July 2024 35
August 2024 23
September 2024 12

Citations

121 Web of Science

×

Email alerts

More on this topic

Citing articles via

More from Oxford Academic