Thyroid Hormone Activates Adenosine 5 -Monophosphate-Activated ...

7 downloads 342 Views 667KB Size Report
Jan 10, 2008 - ... Fukushi Kambe, Xia Cao, Xiuli Lu, Yasuko Kozaki, Yutaka Oiso, and Hisao Seo .... line web site at http://mend.endojournals.org), similar .... signal densities were determined by Multi Gauge software on the LAS-1000 system.
0888-8809/08/$15.00/0 Printed in U.S.A.

Molecular Endocrinology 22(4):893–903 Copyright © 2008 by The Endocrine Society doi: 10.1210/me.2007-0249

Thyroid Hormone Activates Adenosine 5ⴕ-Monophosphate-Activated Protein Kinase via Intracellular Calcium Mobilization and Activation of Calcium/Calmodulin-Dependent Protein Kinase Kinase-␤ Masako Yamauchi, Fukushi Kambe, Xia Cao, Xiuli Lu, Yasuko Kozaki, Yutaka Oiso, and Hisao Seo Department of Endocrinology (M.Y., F.K., X.C., X.L., Y.K., H.S.), Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; and Department of Endocrinology and Diabetes (M.Y., Y.O.), Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan AMP-activated protein kinase (AMPK) is a key regulator of glucose and fatty acid homeostasis. In muscle cells, AMPK stimulates mitochondrial fatty acid oxidation and ATP production. The thyroid hormone T3 increases cellular oxygen consumption and is considered to be a major regulator of mitochondrial activities. In this study, we examined the possible involvement of AMPK in the stimulatory action of T3 on mitochondria. Treatment of C2C12 myoblasts with T3 rapidly led to phosphorylation of AMPK. Acetyl-coenzyme A carboxylase, a direct target of AMPK, was also phosphorylated after T3 treatment. Similar results were obtained with 3T3-L1, FRTL-5, and HeLa cells. Stable expression of T3 receptor (TR)-␣ or TR␤ in Neuro2a cells enhanced this effect of T3, indicating the involvement of TRs. Because HeLa cells express only Ca2ⴙ/calmodulin-dependent protein kinase kinase-␤ (CaMKK␤), one of two known AMPK kinases, it was suggested that the effect of T3 is

mediated by CaMKK␤. Indeed, experiments using a CaMKK inhibitor, STO-609, and an isoformspecific small interfering RNA demonstrated the CaMKK␤-dependent phosphorylation of AMPK. Furthermore, T3 was found to rapidly induce intracellular Ca2ⴙ mobilization in HeLa cells, and a Ca2ⴙ chelator, 1,2-bis(2-aminophenoxy)ethaneN,N,Nⴕ,Nⴕ-tetraacetic acid (BAPTA), suppressed T3- as well as ionomycin-dependent phosphorylation of AMPK. In addition, T3-dependent oxidation of palmitic acids was attenuated by BAPTA, STO-609, and the small interfering RNA for CaMKK␤, indicating that T3-induced activation of AMPK leads to increased fatty acid oxidation. These results demonstrate that T3 nontranscriptionally activates AMPK via intracellular Ca2ⴙ mobilization and CaMKK␤ activation, thereby stimulating mitochondrial fatty acid oxidation. (Molecular Endocrinology 22: 893–903, 2008)

A

heat shock, hypoxia, ischemia, and muscle contraction induce AMPK activation. AMPK is a heterotrimeric complex composed of a catalytic ␣-subunit and regulatory ␤- and ␥-subunits and is allosterically activated by the phosphorylation of Thr-172 on the ␣-subunit, which is catalyzed by upstream kinases such as LKB1. Binding of AMP to the ␥-subunit makes AMPK a better substrate for LKB1 (3). Recently, Ca2⫹/calmodulin-dependent protein kinase kinase-␤ (CaMKK␤) was identified as another upstream kinase (4–6). Upon an elevation of intracellular Ca2⫹, CaMKK␤ is activated and directly phosphorylates the ␣-subunit of AMPK at Thr-172. This action does not require intracellular AMP elevation. The role played by AMPK in the regulation of fatty acid oxidation has been intensively studied in muscle cells (2). Activated AMPK directly phosphorylates Ser-79 in acetyl-coenzyme A (CoA) carboxylase (ACC) and down-regulates its enzymatic activity, resulting in

MP-ACTIVATED protein kinase (AMPK) is a serine/threonine kinase that acts as a sensor of cellular energy status; that is, AMPK is activated by a rise in intracellular AMP/ATP ratio (1, 2). Once activated, AMPK increases energy supply by switching on ATP-generating pathways and decreases energy demand by switching off ATP-consuming pathways. Not only cellular ATP depletion but also various cellular and metabolic stresses such as glucose deprivation, First Published Online January 10, 2008 Abbreviations: ACC, Acetyl-coenzyme A carboxylase; AM, acetoxymethyl ester; AMPK, AMP-activated protein kinase; 2APB, 2-aminoethoxydiphenyl borate; BAPTA, 1,2-bis(2aminophenoxy)ethane-N,N,N⬘,N⬘-tetraacetic acid; CaMKK␤, Ca2⫹/calmodulin-dependent protein kinase kinase-␤; CoA, coenzyme A; IP3, inositol 1,4,5-triphosphate; TH, thyroid hormone; TR, T3 receptor. Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community. 893

894 Mol Endocrinol, April 2008, 22(4):893–903

a decrease in intracellular malonyl-CoA levels. Because malonyl-CoA inhibits fatty acid uptake into mitochondria by preventing carnitine:palmitoyl-CoA acyl transferase present on the mitochondrial membrane, its decrease allows fatty acid uptake, leading to increased fatty acid oxidation and ATP production. The major thyroid hormone (TH) produced by the thyroid gland is L-T4 (T4), which is deiodinated to T3 in peripheral tissues. TH impacts on a number of physiological events, including growth, development, differentiation, thermogenesis, and metabolism. T3 receptor (TR) belongs to the steroid/thyroid hormone receptor superfamily and functions as a ligand-dependent transcription factor. Two separate genes, THRA and THRB, encode TR␣ and TR␤, respectively. Multiple isoforms are generated from each gene by alternative splicing and promoter usage. Some distinct isoforms of TR␣ are localized to mitochondria (7). TH stimulates cellular oxygen consumption and is considered to be a major regulator of mitochondrial activities (8, 9). Although some controversy remains, TH increases mitochondrial oxidative phosphorylation, membrane potential (⌬␺), and uncoupling proton transport that generates heat. TH concomitantly enhances ATP production capacity as a result of increased oxidative phosphorylation (10). Although the molecular bases underlying these TH actions are still not fully understood, TH stimulates the expression of components of oxidative phosphorylation (11–13), proteins involved in uncoupling proton transport (14), and factors regulating mitochondrial activities (15). These proteins are encoded by nuclear and mitochondrial genes, and T3 modulates their expression via TRs present in the respective organelles. Furthermore, T3 stimulates mitochondrial DNA replication (8). In addition to such long-term, probably mainly transcriptional effects, TH exerts nontranscriptional, short-term effects on mitochondria. T3 rapidly increases oxygen consumption and oxidative phosphorylation in mitochondria, in a process that does not require protein synthesis (16). Several mitochondrial proteins have been shown to bind to TH with high affinity. However, their roles in the short-term effects of TH appear to be controversial (17). In the present study, we examined the possible involvement of AMPK in the stimulatory actions of T3 on mitochondria. We provide evidence that T3 rapidly activates AMPK via intracellular Ca2⫹ mobilization and CaMKK␤ activation.

RESULTS T3 Rapidly Induces Phosphorylation of Thr-172 in the ␣-Subunit of AMPK Because the involvement of AMPK in fatty acid oxidation has been well documented in muscle and fat cells (1), we first chose myoblast C2C12 cells and preadipocyte 3T3-L1 cells to test the effect of T3 on AMPK. Differentiated cells were kept in serum-deprived media

Yamauchi et al. • Thyroid Hormone Activates AMPK

for 12 h and treated with 10 nM T3 for 30 min. Western blot analysis (Fig. 1A) revealed that serum deprivation induced a slight but significant increase in phosphorylation of Thr-172 in the ␣-subunit of AMPK. T3 treatment further increased the phosphorylation. Similar

Fig. 1. T3 Induces the Phosphorylation of Thr-172 in the ␣-Subunit of AMPK Differentiated C2C12, 3T3-L1, and FRTL-5 cells were cultured with serum [S(⫹)] in serum-deprived media for 12 h [S(⫺)]. A, The cells were treated with 10 nM T3 for 30 min. Whole-cell lysates were subjected to Western blot analysis using antibodies directed against phospho-AMPK ␣-subunit (pT172), AMPK ␣-subunit (total), and actin. Most experiments were performed in duplicate cultures. Similar results were obtained from separate experiments. In densitometric analysis, the phospho-AMPK levels were normalized to the actin levels and expressed as percentages of the levels in T3treated cells. Results are shown as means ⫾ SD (n ⫽ 4). *, P ⬍ 0.05 vs. the levels in S(⫹) cells; #, P ⬍ 0.05 vs. the levels in S(⫺) cells. B, C2C12 cells were treated with 10 nM T3 for the indicated times, and Western blot analysis was performed. C, C2C12 cells were treated with 1 nM to 1 ␮M T3 for 30 min, and Western blot analysis was performed.

Yamauchi et al. • Thyroid Hormone Activates AMPK

results were obtained with thyroid follicular FRTL-5 cells, indicating that T3-induced phosphorylation of AMPK is not a phenomenon that is restricted to a particular cell type. A time-course experiment (Fig. 1B) showed that T3 induced the phosphorylation of AMPK within 5 min and that its effect lasted for more than 120 min. The phosphorylation of AMPK by T3 was dose dependent, as shown in Fig. 1C. TR␣ and TR␤ Mediate the T3-Induced Phosphorylation of AMPK Several nontranscriptional effects of T3 have been demonstrated to be mediated by TRs (18–22). Thus, we studied the possible involvement of TR in the T3induced phosphorylation of AMPK by using N2aTR␣ and N2aTR␤ cells, which stably express TR␣1 and TR␤1, respectively. As shown in Fig. 2A, serum deprivation significantly increased the phosphorylation of Thr-172 in the ␣-subunit of AMPK in wild-type N2a cells, but subsequent treatment with T3 had only a marginal effect. A similar tendency was observed for Ser-79 phosphorylation of ACC. By contrast, N2aTR␣ and N2aTR␤ cells markedly responded to T3. Serum deprivation increased the phosphorylation of AMPK and ACC, and T3 further increased this phosphorylation. Immunocytochemical analysis using anti-phosphoThr-172 AMPK ␣-subunit showed that T3 significantly increased the fluorescence intensity in the cytoplasm, indicating that T3-induced phosphorylation of AMPK mainly occurred in the cytoplasm (Fig. 2B). A similar increase was observed in the cells treated with oligomycin, an ATP synthase inhibitor. The expression profiles of TR isoforms in wild-type N2a, N2aTR␣, and N2aTR␤ cells are shown in Fig. 2C. Together, these results indicate that both TR␣ and -␤ isoforms can mediate the T3-induced phosphorylation of AMPK. The TR mRNA levels in N2a cells were compared with those in C2C12 and 3T3-L1 cells by RT-PCR. As shown in supplemental Fig. A (published as supplemental data on The Endocrine Society’s Journals Online web site at http://mend.endojournals.org), similar levels of TR mRNA were detected in all three cell types. Thus, it is unlikely that the lower response of N2a cells to T3 is due to a difference in TR expression. Some components of the downstream signaling pathway might be less functional in N2a cells. CaMKK␤ Mediates the T3-Induced Phosphorylation of AMPK To explore the signaling cascade underlying the T3 action, we first examined the expression of LKB1 and CaMKK␤ in the cells used in this study. As shown in Fig. 3A, RT-PCR analysis revealed that whereas CaMKK␤ was expressed in all of the cell types tested, LKB1 was not expressed in HeLa cells, consistent with a previous report (6). Thus, we used HeLa cells to study whether LKB1 is required for the T3-induced phosphorylation of AMPK. TR␣ and TR␤1 expression

Mol Endocrinol, April 2008, 22(4):893–903 895

was detected by RT-PCR in HeLa cells (data not shown). As shown in Fig. 3B, T3 induced a significant increase in the phosphorylation of Thr-172 in the ␣-subunit of AMPK. Ser-79 phosphorylation of ACC was also increased by T3 in HeLa cells, indicating that LKB1 was dispensable for the T3 action. We then tested a CaMKK inhibitor, STO-609. Hawley et al. (4) reported that 10 ␮M STO-609 inhibits a broad range of kinases, but 1 ␮M STO-609 is relatively selective for CaMKK␣ and -␤. As shown in Fig. 3B, 1 ␮M STO-609 was sufficient to prevent the effect of T3 on AMPK and ACC, suggesting the involvement of CaMKK␤. This notion was validated by the following experiment using small interfering RNA (siRNA). The effects of CaMKK␤ and control siRNAs on the expression of CaMKK␤, CaMKK␣, and TR␤1 in HeLa cells were examined by RT-PCR (Fig. 3C). CaMKK␤ siRNA markedly silenced CaMKK␤ expression, whereas it did not affect the expression of CaMKK␣ and TR␤1. A nontargeting, control siRNA had no effect on the expression of these three proteins. To confirm the silencing effect of CaMKK␤ siRNA, HeLa cells were treated with a Ca2⫹ ionophore, ionomycin, and the levels of AMPK and ACC phosphorylation were determined. As shown in Fig. 3D, the introduction of CaMKK␤ siRNA markedly suppressed the ionomycin-dependent phosphorylation of AMPK and ACC, whereas control siRNA had no effect, demonstrating that the CaMKK␤ siRNA successfully functioned in HeLa cells. When HeLa cells transfected with CaMKK␤ siRNA were treated with T3, the phosphorylation of AMPK and ACC was markedly inhibited. Together, these results demonstrate that CaMKK␤ mediates the T3-induced phosphorylation of AMPK. T3-Dependent Intracellular Ca2ⴙ Mobilization Is Associated with AMPK Phosphorylation We next studied T3-dependent intracellular Ca2⫹ mobilization in HeLa cells by using a fluorescent indicator, Fluo-4. As shown in Fig. 4A, T3 induced a rapid and significant increase in intracellular Ca2⫹ levels, which gradually decreased after T3 withdrawal. When the cells were preincubated with 1,2-bis(2-aminophenoxy)ethane-N,N,N⬘,N⬘-tetraacetic acid (BAPTA), a Ca2⫹ chelator, the fluorescence signals from Ca2⫹/ Fluo-4 complexes were markedly suppressed, possibly due to the competition of BAPTA with Fluo-4 to bind Ca2⫹. We repeated the experiment by using a ratiometric Ca2⫹ indicator, Indo-1 (Fig. 4B). The profiles of Ca2⫹ increase were similar to those obtained with Fluo-4. The lower degree of Ca2⫹ increase may be due to the lower sensitivity of Indo-1. These results demonstrate the T3 induction of intracellular Ca2⫹ mobilization in HeLa cells. Then, the effects of BAPTA on the phosphorylation of AMPK and ACC were examined, as shown in Fig. 5A. BAPTA suppressed both ionomycin- and T3-dependent phosphorylation of AMPK and ACC,

896 Mol Endocrinol, April 2008, 22(4):893–903

Yamauchi et al. • Thyroid Hormone Activates AMPK

Fig. 2. TR Mediates the T3-Induced Phosphorylation of AMPK A, Wild-type N2a cells and N2aTR␣ and N2aTR␤ cells were cultured with [S(⫹)] or without [S(⫺)] serum for 12 h and were then treated with 10 nM T3 for 30 min. Whole-cell lysates were subjected to Western blot analysis using antibodies directed against phospho-AMPK ␣-subunit (pT172), AMPK ␣-subunit (total), phospho-ACC (pS79), and actin. Most experiments were performed in duplicate cultures. Similar results were obtained from separate experiments. In densitometric analysis, the phospho-AMPK and phospho-ACC levels were normalized to the actin levels and expressed as percentages of the levels in T3-treated N2aTR␣ cells. Results are means ⫾ SD (n ⫽ 4). *, P ⬍ 0.05 vs. the levels in S(⫹) cells; #, P ⬍ 0.05 vs. the levels in S(⫺) cells. B, N2aTR␣ cells cultured without serum for 12 h [S(⫺)] were treated with 10 nM T3 or 10 ␮M oligomycin for 30 min. Immunocytochemical analysis was performed using rabbit anti-phospho-AMPK ␣-subunit (pT172) antibody and antirabbit IgG antibody conjugated to Alexa Fluor 488 (green fluorescence). Propidium iodide staining was performed to identify nuclei (red fluorescence). Representative images are shown. Quantitative data on the amounts of phospho-AMPK in the cytoplasm are also presented. Several images were randomly captured, and the cytoplasmic signal densities were determined by Multi Gauge software on the LAS-1000 system. The values are expressed in arbitrary units, means ⫾ SD (n ⫽ 20). *, P ⬍ 0.05 vs. S(⫺). Similar results were obtained from separate experiments. C, The mRNA levels of TR␣ and TR␤1 in wild-type N2a, N2aTR␣, and N2aTR␤ cells were determined by RT-PCR. N2aTR␣ and N2aTR␤ cells are stable transformants expressing chicken TR␣1 and human TR␤1, respectively. The PCR products were subjected to agarose gel electrophoresis and visualized by ethidium bromide.

strongly suggesting that T3-dependent intracellular Ca2⫹ mobilization leads to CaMKK␤ activation, followed by AMPK phosphorylation. Similarly, T3-dependent AMPK phosphorylation in N2aTR␤ cells

was suppressed by BAPTA and STO-609 (supplemental Fig. B). We then studied the effects of an antagonist to inositol 1,4,5-triphosphate (IP3) receptor, 2-amino-

Yamauchi et al. • Thyroid Hormone Activates AMPK

Mol Endocrinol, April 2008, 22(4):893–903 897

Fig. 3. CaMKK␤ Mediates the T3-Induced Phosphorylation of AMPK A, The mRNA levels of LKB1 and CaMKK␤ in C2C12, 3T3-L1, FRTL-5, N2a, and HeLa cells were determined by RT-PCR. The products were subjected to agarose gel electrophoresis and visualized by ethidium bromide. No band was detected when the reverse transcriptase was omitted (data not shown). B, HeLa cells were cultured without serum for 12 h. They were pretreated for 15 min with 1 ␮M STO-609 and treated for 30 min with 10 nM T3 in the presence of STO-609. Whole-cell lysates were subjected to Western blot analysis using antibodies against phospho-AMPK ␣-subunit (pT172), AMPK ␣-subunit (Total), phospho-ACC (pS79), and actin. Experiments were performed in duplicate cultures. Similar results were obtained from separate experiments. In densitometric analysis, the phospho-AMPK and phospho-ACC levels were normalized to the actin levels and expressed as percentages of the levels in cells treated with T3 alone. Results are means ⫾ SD (n ⫽ 4). *, P ⬍ 0.05 vs. the control levels; #, P ⬍ 0.05 vs. the levels in cells treated with T3 alone. C, CaMKK␤ siRNA and nontargeting, negative control siRNA were transfected into HeLa cells. The expression of CaMKK␤, CaMKK␣, and TR␤1 was determined by RT-PCR 48 h after transfection. D, HeLa cells transfected with CaMKK␤ or control siRNA for 48 h were cultured without serum for 12 h and then treated with 1 ␮M ionomycin or 10 nM T3 for 30 min. Whole-cell lysates were subjected to Western blot analysis using antibodies against phospho-AMPK ␣-subunit (pT172), phospho-ACC (pS79), and actin. The experiments were performed in duplicate cultures. Similar results were obtained from separate experiments.

ethoxydiphenyl borate (2APB) (23), on T3- and IP3dependent AMPK phosphorylation (Fig. 5B). AMPK phosphorylation was markedly suppressed by 2APB, suggesting that Ca2⫹ release from IP3 receptor is involved in the T3-induced phosphorylation of AMPK. T3-Dependent Fatty Acid Oxidation Is Mediated by CaMKK␤ The above studies showed that T3 induced Ser-79 phosphorylation of ACC via AMPK activation, which would result in ACC inactivation and increase fatty acid oxidation (2). To confirm this notion, the effects of T3 on the oxidation of labeled palmitic acids were assessed in HeLa cells. As shown in Fig. 6A, the basal oxidation levels were significantly decreased by BAPTA, but not by STO-609, suggesting that intracellular Ca2⫹, but not CaMKK, might contribute to the basal fatty acid oxidation. T3 significantly increased the fatty acid oxidation. This increase was attenuated not only by BAPTA but also by STO-609. When HeLa cells transfected with CaMKK␤ siRNA were subjected to the fatty acid oxidation assay, the T3-dependent increase was significantly reduced (Fig. 6B). These

results indicate that T3-dependent fatty acid oxidation is mediated by CaMKK␤ in HeLa cells. As shown in Fig. 6C, T3-dependent fatty acid oxidation was also decreased by BAPTA and STO-609 in C2C12 cells that endogenously express both CaMKK␤ and LKB1. Accordingly, T3-induced phosphorylation of AMPK was suppressed by BAPTA and STO-609 (supplemental Fig. C), suggesting that the effect of T3 on AMPK in C2C12 cells is mainly mediated by CaMKK. Similarly, the phosphorylation of AMPK by 2-deoxyglucose was also suppressed by BAPTA and STO-609 (supplemental Fig. C). Together, these results demonstrate that a signaling pathway involving CaMKK is important for the activation of AMPK by multiple stimuli such as T3 and 2-deoxyglucose in C2C12 cells.

DISCUSSION In the present study, we show for the first time that T3 stimulates the phosphorylation of AMPK that is critical for kinase activation. This action occurred rapidly, within several minutes of the start of T3 treatment and

898 Mol Endocrinol, April 2008, 22(4):893–903

Fig. 4. T3 Induces Intracellular Ca2⫹ Mobilization A, HeLa cells labeled with Fluo-4-AM (F) or cells treated with both BAPTA-AM and Fluo-4-AM (E) were superfused with HEPES-buffered Ringer solution (see Materials and Methods ). Five consecutive images with 6-sec intervals were obtained before T3 treatment. Cells were then treated with 10 nM T3 for 180 sec, and 45 consecutive images were obtained during and after T3 treatment for 270 sec. Representative images before (control) and 150 sec after T3 treatment are presented. Changes in fluorescence intensities are indicated by pseudo-colors. The fluorescence intensities of individual cells were measured and are expressed as a ratio of the averaged intensities obtained from five images before T3 stimulation. Results are means ⫾ SE (n ⫽ 38–45). B, HeLa cells labeled with the ratiometric Ca2⫹ indicator Indo-1-AM (F) or cells treated with both BAPTA-AM and Indo-1-AM (E) were treated with T3 as described above. Indo-1 was excited at 351 nm. Emissions at 400–445 nm (FS) and longer than 445 nm (FL) were captured simultaneously. Indo-1 ratios (a ratio of FS to FL) in individual cells were calculated and expressed relative to the averaged ratio obtained from five images before T3 stimulation. Results are means ⫾ SE (n ⫽ 30–35).

was observed in cells in which the basal phosphorylation levels were already elevated by 12-h serum starvation. AMPK activation during long-term serum starvation is thought to be mainly due to cellular ATP

Yamauchi et al. • Thyroid Hormone Activates AMPK

depletion. It was thus suggested that the additive effect of T3 would be mediated by the enhancement of the signaling pathway elicited by ATP depletion or the activation of an alternative pathway leading to AMPK phosphorylation. The fact that the effect of T3 was extremely rapid does not support the involvement of alterations in intracellular adenine nucleotide levels. Using cells lacking LKB1, a CaMKK inhibitor, and an isoform-specific siRNA, we provide evidence that the T3-induced phosphorylation of AMPK is mediated by a signaling pathway involving CaMKK␤. CaMKK␤ was first identified as a kinase upstream of Ca2⫹/calmodulin-dependent protein kinase I and IV (24). Recent extensive studies involving yeast genetics have revealed that CaMKK␤ can also phosphorylate AMPK in mammalian cells (4–6). These studies showed that ionomycin, hydrogen peroxide, sorbitol, and 2-deoxyglucose stimulated AMPK via CaMKK␤ activation. These reagents mimic the situations of intracellular Ca2⫹ mobilization, oxidant stress, osmotic stress, and energy deprivation, respectively, thus suggesting that CaMKK␤-dependent AMPK activation would play an important role in cells exposed to various physiological and pathological conditions. However, the physiological stimuli activating the CaMKK␤AMPK cascade remain poorly identified. It was recently reported that thrombin can activate this cascade in vein endothelial cells (25). Moreover, antigen receptor activation in T lymphocytes was reported to stimulate this cascade (26). T3 is another physiological stimulus activating AMPK via CaMKK␤. T3 has been shown to nontranscriptionally modulate a variety of proteins, including ERKs, the sodium/proton exchanger, a voltage-dependent potassium channel (ether-a-go-go related gene channel), the sodium/ potassium ATPase, and phosphoinositide 3 kinase (PI3K) (20, 22, 27–29). These proteins do not directly bind T3, so their functions are regulated by T3 indirectly, most likely through T3/TR complexes. The involvement of TR in the nontranscriptional effect of T3 was demonstrated in mice possessing a mutant TR␤ that selectively lacks DNA-binding activity (30). Recent in vitro studies including ours have shown that TR directly interacts with a regulatory subunit of PI3K, suggesting an essential role of TR in the regulation of PI3K by T3 (19–22). In addition, it was reported that T3-induced activation of Akt, a kinase downstream of PI3K, was greatly impaired in TR␣ and -␤ doubleknockout mice (21). In the current study, the involvement of TR␣ and TR␤ in the T3-induced activation of AMPK was suggested by using cells stably expressing TR␣ or TR␤. However, unlike the case of PI3K, T3/TR complexes would not directly contribute to AMPK activation; rather, they triggered intracellular Ca2⫹ mobilization. This study demonstrates that T3 rapidly, within a minute, increases intracellular Ca2⫹ levels and that this is associated with the phosphorylation of AMPK and ACC. Several recent reports describe acute effects of T3 on intracellular Ca2⫹. D’Arezzo et al. (28)

Yamauchi et al. • Thyroid Hormone Activates AMPK

Mol Endocrinol, April 2008, 22(4):893–903 899

Fig. 5. T3-Dependent Intracellular Ca2⫹ Mobilization Is Associated with the Phosphorylation of AMPK and ACC A, HeLa cells were cultured with [S(⫹)] or without [S(⫺)] serum for 12 h. They were pretreated for 15 min with 10 ␮M BAPTA and treated for 30 min with 10 nM T3 or 1 ␮M ionomycin (Iono) in the presence of BAPTA. Whole-cell lysates were subjected to Western blot analysis using antibodies against phospho-AMPK ␣-subunit (pT172), AMPK ␣-subunit (total), phospho-ACC (pS79), and actin. The experiments were performed in duplicate cultures. Similar results were obtained from separate experiments. In densitometric analysis, the phospho-AMPK and phospho-ACC levels were normalized to the actin levels and expressed as percentages of the levels in cells treated with T3 or ionomycin alone. Results are means ⫾ SD (n ⫽ 4). *, P ⬍ 0.05 vs. the levels in S(⫺) cells; #, P ⬍ 0.05 vs. the levels in cells treated with T3 or ionomycin alone. B, HeLa cells were cultured with [S(⫹)] or without [S(⫺)] serum for 12 h. They were pretreated for 15 min with 100 ␮M 2APB and treated with 10 nM T3 for 30 min or 1 ␮M IP3 for 5 min in the presence of 2APB. Whole-cell lysates were subjected to Western blot analysis using antibodies against phosphoAMPK ␣-subunit (pT172) and actin. The experiments were performed in duplicate cultures. Similar results were obtained from separate experiments. In densitometric analysis, the phospho-AMPK levels were normalized to the actin levels and expressed as percentages of the levels in cells treated with T3 or IP3 alone. Results are means ⫾ SD (n ⫽ 4). *, P ⬍ 0.05 vs. the levels of S(⫺); #, P ⬍ 0.05 vs. the levels of T3 alone or the levels of IP3 alone.

900 Mol Endocrinol, April 2008, 22(4):893–903

Yamauchi et al. • Thyroid Hormone Activates AMPK

showed in the L-6 rat myoblast cell line that T3 induces intracellular Ca2⫹ mobilization within a few minutes, through activation of IP3 receptors and without contributions of extracellular Ca2⫹ and ryanodine receptors. Interestingly, T3-bound agarose, which cannot enter cells, reproduced this effect of T3, suggesting that its action is initiated at the plasma membrane, but the nature of T3-binding protein on the membrane was not identified. Saelim et al. (18) also reported that T3 increases the IP3-mediated Ca2⫹ wave period and amplitude in Xenopus oocytes expressing mitochondrial forms of TR and that this increase is mediated by mitochondrial Ca2⫹ uptake. They further showed that increased mitochondrial membrane potential (⌬␺) contributes to the uptake. These reports indicate that T3 induces intracellular Ca2⫹ mobilization by cooperatively stimulating the endoplasmic reticulum and mitochondria. The current study further shows that the T3-induced phosphorylation of AMPK leads to increased fatty acid oxidation, which would subsequently increase mitochondrial acetyl-CoA levels and stimulate oxidative phosphorylation. T3 exerts profound effects on cell growth, differentiation, and thermogenesis, processes that may consume a considerable amount of ATP or dissipate mitochondrial membrane potential. Thus, cells with limited energy turnover could not properly respond to T3. The activation of AMPK would allow cells to switch on ATP-generating pathways and render them acceptable for T3.

MATERIALS AND METHODS Cell Culture

Fig. 6. T3-Dependent Fatty Acid Oxidation Is Mediated by CaMKK␤ A, HeLa cells were preincubated for 2 h in DMEM containing 0.5% fatty acid-free BSA and were incubated for 3 h in DMEM containing 2% fatty acid-free BSA, 0.3 mM palmitic acid, and 1 ␮Ci/ml [3H]palmitic acid in the presence or absence of 10 nM T3, 10 ␮M BAPTA (BAP), and 1 ␮M STO-609 (STO). After incubation, the media were collected, and the labeled palmitic acids remaining in the media were removed by precipitation with perchloric acid. The radioactivity of the supernatants was counted using a liquid scintillation counter. The levels of radioactive water production are expressed as percentages of the nontreated, control levels. Results are means ⫾ SD (n ⫽ 3). *, P ⬍ 0.05 vs. the levels of the control; #, P ⬍ 0.05 vs. the levels in cells treated with T3 alone. Similar results were obtained from separate experiments. B, HeLa cells transfected with CaMKK␤ or control siRNA for 48 h were subjected to the fatty acid oxidation assay as described above. Results are means ⫾ SD (n ⫽ 3). *, P ⬍ 0.05 vs. the

The mouse myoblast cell line C2C12 [American Type Culture Collection (ATCC), Manassas, VA; CRL-1772], the mouse preadipocyte cell line 3T3-L1 (ATCC CL-173), and the human cervical carcinoma cell line HeLa (ATCC CCL-2) were cultured in DMEM supplemented with 10% fetal bovine serum. The mouse neuroblastoma cell line Neuro 2a (ATCC CCL131) was cultured in DMEM/F12 medium supplemented with 10% fetal bovine serum. The rat thyroid cell line FRTL-5 (ATCC CRL-1468) was cultured in Coon’s modified Ham’s F-12 medium supplemented with 5% heat-inactivated calf serum and six hormones as described previously (31). Neuro 2a cells stably transfected with chicken TR␣1 (N2aTR␣) were a generous gift from Professor J. Bernal (Instituto de Investigaciones Biomedicas, Madrid, Spain) (32). Neuro 2a cells stably transfected with human TR␤1 (N2aTR␤) were a generous gift from Professor J. Puymirat (Human Genetics Re-

levels in cells treated with the control siRNA and T3 (⫺); #, P ⬍ 0.05 vs. the levels in cells treated with the control siRNA and T3 (⫹). Similar results were obtained from separate experiments. C, Differentiated C2C12 cells were subjected to the fatty acid oxidation assay as described above. Results are means ⫾ SD (n ⫽ 3). *, P ⬍ 0.05 vs. the levels of the control; #, P ⬍ 0.05 vs. the levels in cells treated with T3 alone. Similar results were obtained from separate experiments.

Yamauchi et al. • Thyroid Hormone Activates AMPK

search Unit, Laval University, Quebec, Canada) (33). Differentiation of C2C12 cells into myotubes was induced by mitogen withdrawal (DMEM with 2% horse serum) and was confirmed by troponin T expression. The differentiation of 3T3-L1 cells into adipocytes was induced by combined treatment of the cells with 3-isobutyl-1-methylxanthine, dexamethasone, and insulin (34). Near-confluent cells were cultured in serum-deprived media for 12 h and treated with various hormones and reagents such as T3, oligomycin, ionomycin, STO-609, BAPTA-acetoxymethyl ester (BAPTA-AM), IP3, and 2APB, alone or in combination. These reagents were purchased from Sigma-Aldrich (St. Louis, MO), except for BAPTA-AM and IP3 (Dojindo Laboratories, Kumamoto, Japan).

Mol Endocrinol, April 2008, 22(4):893–903 901

rat TR␤1, 5⬘-ACAGAAAATGGCCTTCCAGC-3⬘ and 5⬘-TCTTGCTGTCATCCAGCACCA-3⬘; and primers for GAPDH (glyceraldehyde-3-phosphate dehydrogenase), 5⬘-GCACCGTCAAGGCTGAGAAC-3⬘ and 5⬘-GCCTTCTCCATGGTGGTGAA-3⬘. siRNA A predesigned siRNA directed against CaMKK␤ (ID 110916, ␤3) and Silencer Negative Control no. 1 (nontargeting siRNA) were purchased from Ambion (Austin, TX). HeLa cells were seeded in six-well plates 24 h before transfection. siRNA (final concentration 80 nM) was introduced into cells using siPORT Lipid (Ambion).

Western Blot Analysis

Measurement of Intracellular Ca2ⴙ Levels

The procedures used for preparation of whole-cell lysates and Western blot analysis were described in our previous report (22). In brief, whole-cell lysates (40 ␮g/lane) were separated by SDS-PAGE and transferred onto polyvinylidene difluoride membranes (Amersham Pharmacia, Piscataway, NJ). The blots were probed with the first antibodies described below, followed by incubation with horseradish peroxidaseconjugated antirabbit IgG antibody. Rabbit anti-phosphoAMPK ␣-subunit (Thr-172), anti-AMPK ␣-subunit, and antiphospho-ACC (Ser-79) antibodies were purchased from Cell Signaling (Beverly, MA). Rabbit anti-actin antibody was purchased from Sigma-Aldrich. Proteins were visualized using enhanced chemiluminescence (ECL) reagents (Pierce, Rockford, IL). Images of the membranes were obtained using an LAS-1000 lumino-image analyzer (Fuji Film, Tokyo, Japan), and densitometric analysis was performed using software on the LAS-1000 system.

Intracellular Ca2⫹ changes were measured using a fluorescent dye technique. The detailed procedures were described in our previous report (37). In brief, HeLa cells cultured on glass coverslips were treated for 45 min with 5 ␮M Fluo-4-AM (Molecular Probes), 0.02% pluronic acid F127 (Molecular Probes), and 1.25 mM probenecid in HEPES-buffered Ringer solution. In some experiments, cells were preincubated with 10 ␮M BAPTA-AM for 15 min; then, 5 ␮M Fluo-4 was added and incubated for 45 min. The coverslips were placed in a chamber mounted on the stage of an inverted microscope (Axiovert, Zeiss) equipped with a confocal laser scanning system (Oz; Noran, Middleton, WT) and then superfused with HEPES-buffered Ringer solution at a flow rate of 3 ml/min. Five consecutive images with 6-sec intervals were obtained before T3 stimulation. The cells were then treated with 10 nM T3 for 180 sec, and 45 consecutive images were obtained during and after T3 treatment for 270 sec. The averages of all the fluorescence intensities of individual cells were analyzed using the Intervision 2D program (Noran). A ratiometric calcium indicator, Indo-1-AM (Dojindo), was also used instead of Fluo-4-AM. When Indo-1 binds Ca2⫹, the fluorescence emission at 405 nm increases, and the emission at 495 nm decreases. Measurement of the ratio of these emission intensities allows accurate estimation of intracellular Ca2⫹ concentrations, independent of intracellular dye concentrations. Indo-1-AM was loaded using the same protocol as Fluo-4-AM. To prevent photodegradation of Indo-1, 10 mM Trolox (EMD Biosciences, La Jolla, CA) was added to HEPES-Ringer. Indo-1 was excited at 351 nm. The emissions at 400–445 nm (FS) and longer than 445 nm (FL) were captured simultaneously using a dichroic mirror (445 nm) and a 400-nm longpass barrier filter, and the Indo-1 ratios (the ratio of FS to FL) in individual cells were calculated.

Immunocytochemical Analysis The procedures used for immunocytochemical analysis were described in our previous report (35). After fixation and blocking, cells were incubated with anti-phospho-AMPK (Thr-172) antibody followed by incubation with antirabbit IgG antibody conjugated to Alexa Fluor 488 (Molecular Probes, Eugene, OR). Counterstaining was performed using propidium iodide. Images were obtained using a confocal laser microscope (LSM510; Carl Zeiss, Jena, Germany) with the same set of optical parameters as described previously (36). Several images were captured, and densitometric analysis of the amounts of phosphorylated AMPK was performed using Multi Gauge software on the LAS-1000 system. Merged images were obtained using Adobe Photoshop software (version 7.0.1; Adobe Systems, San Jose, CA). RT-PCR

Measurement of Fatty Acid Oxidation

The expression of LKB1, CaMKK␤, TR␣, and TR␤1 was examined by RT-PCR using total RNA. The primer sequences are as follows: sense and antisense primers for LKB1, 5⬘-CTCATCGGCAAGTACCTGATG-3⬘ and 5⬘TGGGAAACGCTTCTCCGGCAC-3⬘; primers for mouse CaMKK-␤, 5⬘- TAGAGTACTTGCATTACCAGAAGA-3⬘ and 5⬘- TGGGTTTTTGTCCAACATCCGGGT-3⬘; primers for human CaMKK-␤, 5⬘- TCGAGTACTTGCACTGCCAGAAGATC-3⬘ and 5⬘-GGGGTTCTTGTCCAGCATACGGGT-3⬘; primers for human CaMKK-␣, 5⬘-TCGAGTACTTGCACTGCCAGAAGATC-3⬘ and 5⬘-GGGATTCTTGTCTAACATCTTCAG-3⬘; primers for TR␣, 5⬘-AAGCCAAGCAAGGTGGAGTGT-3⬘ and 5⬘-ATGCACTTCTTGAAGCGGCACA-3⬘; primers for chicken TR␣, 5⬘-GGTCGTGTCTGATGCCATCT-3⬘ and 5⬘- CTCCTGGTCCTCGAAGACCT-3⬘; primers for human TR␤1, 5⬘-GCTATGACCCAGAAAGTGAG-3⬘ and 5⬘-TGTCACCTTCATCAGGAGTT-3⬘; primers for mouse and

Fatty acid oxidation was determined using [9,10(n)3 H]palmitic acid (GE Healthcare Amersham, Buckinghamshire, UK). Palmitic acids were conjugated to fatty acid-free BSA (Sigma-Aldrich) by incubating DMEM containing 2% fatty acid-free BSA, 0.3 mM palmitic acid (Sigma-Aldrich), and 1 ␮Ci/ml [3H]palmitic acid at 45 C. HeLa cells seeded in six-well plates on the day before the experiment were preincubated for 2 h in DMEM containing 0.5% fatty acid-free BSA. Then they were incubated for 3 h in DMEM containing [3H]palmitic acids conjugated to BSA in the presence or absence of 10 nM T3, 10 ␮M BAPTA, and 1 ␮M STO-609. After incubation, the media were collected, and the labeled palmitic acids remaining in the media were removed by precipitation with perchloric acid. Precipitation was performed twice. The radioactivity of the supernatants was determined using a liquid scintillation counter to quantify the radioactive water production.

902 Mol Endocrinol, April 2008, 22(4):893–903

Yamauchi et al. • Thyroid Hormone Activates AMPK

Statistical Analysis Statistical analysis was performed using ANOVA followed by Bonferroni’s multiple t test; a P value ⬍ 0.05 was considered to be statistically significant.

14.

Acknowledgments

16.

Received May 10, 2007. Accepted January 2, 2008. Address all correspondence and requests for reprints to: Fukushi Kambe, M.D., Ph.D., Department of Endocrinology, Research Institute of Environmental Medicine (RIEM), Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan. E-mail: [email protected]. This work was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Science, Sports, and Culture, Japan. Disclosure Statement: The authors have nothing to disclose.

15.

17.

18.

19.

20.

REFERENCES 21. 1. Hardie DG, Sakamoto K 2006 AMPK: a key sensor of fuel and energy status in skeletal muscle. Physiology (Bethesda) 21:48–60 2. Kahn BB, Alquier T, Carling D, Hardie DG 2005 AMPactivated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab 1:15–25 3. Hardie DG 2005 New roles for the LKB13AMPK pathway. Curr Opin Cell Biol 17:167–173 4. Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM, Frenguelli BG, Hardie DG 2005 Calmodulindependent protein kinase kinase-␤ is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab 2:9–19 5. Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, Carlson M, Carling D 2005 Ca2⫹/ calmodulin-dependent protein kinase kinase-␤ acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab 2:21–33 6. Hurley RL, Anderson KA, Franzone JM, Kemp BE, Means AR, Witters LA 2005 The Ca2⫹/calmodulin-dependent protein kinase kinases are AMP-activated protein kinase kinases. J Biol Chem 280:29060–29066 7. Bassett JH, Harvey CB, Williams GR 2003 Mechanisms of thyroid hormone receptor-specific nuclear and extra nuclear actions. Mol Cell Endocrinol 213:1–11 8. Wrutniak-Cabello C, Casas F, Cabello G 2001 Thyroid hormone action in mitochondria. J Mol Endocrinol 26: 67–77 9. Goglia F, Silvestri E, Lanni A 2002 Thyroid hormones and mitochondria. Biosci Rep 22:17–32 10. Short KR, Nygren J, Barazzoni R, Levine J, Nair KS 2001 T3 increases mitochondrial ATP production in oxidative muscle despite increased expression of UCP2 and -3. Am J Physiol Endocrinol Metab 280:E761–E769 11. Izquierdo JM, Cuezva JM 1993 Thyroid hormones promote transcriptional activation of the nuclear gene coding for mitochondrial ␤-F1-ATPase in rat liver. FEBS Lett 323:109–112 12. Dummler K, Muller S, Seitz HJ 1996 Regulation of adenine nucleotide translocase and glycerol 3-phosphate dehydrogenase expression by thyroid hormones in different rat tissues. Biochem J 317(Pt 3):913–918 13. Li R, Luciakova K, Zaid A, Betina S, Fridell E, Nelson BD 1997 Thyroid hormone activates transcription from the promoter regions of some human nuclear-encoded

22.

23.

24.

25.

26.

27.

28.

29.

30.

genes of the oxidative phosphorylation system. Mol Cell Endocrinol 128:69–75 Lanni A, Moreno M, Lombardi A, Goglia F 2003 Thyroid hormone and uncoupling proteins. FEBS Lett 543:5–10 Weitzel JM, Iwen KA, Seitz HJ 2003 Regulation of mitochondrial biogenesis by thyroid hormone. Exp Physiol 88:121–128 Sterling K, Brenner MA, Sakurada T 1980 Rapid effect of triiodothyronine on the mitochondrial pathway in rat liver in vivo. Science 210:340–342 Goglia F, Moreno M, Lanni A 1999 Action of thyroid hormones at the cellular level: the mitochondrial target. FEBS Lett 452:115–120 Saelim N, John LM, Wu J, Park JS, Bai Y, Camacho P, Lechleiter JD 2004 Nontranscriptional modulation of intracellular Ca2⫹ signaling by ligand stimulated thyroid hormone receptor. J Cell Biol 167:915–924 Furuya F, Hanover JA, Cheng SY 2006 Activation of phosphatidylinositol 3-kinase signaling by a mutant thyroid hormone beta receptor. Proc Natl Acad Sci USA 103:1780–1785 Storey NM, Gentile S, Ullah H, Russo A, Muessel M, Erxleben C, Armstrong DL 2006 Rapid signaling at the plasma membrane by a nuclear receptor for thyroid hormone. Proc Natl Acad Sci USA 103:5197–5201 Hiroi Y, Kim HH, Ying H, Furuya F, Huang Z, Simoncini T, Noma K, Ueki K, Nguyen NH, Scanlan TS, Moskowitz MA, Cheng SY, Liao JK 2006 Rapid nongenomic actions of thyroid hormone. Proc Natl Acad Sci USA 103: 14104–14109 Cao X, Kambe F, Moeller LC, Refetoff S, Seo H 2005 Thyroid hormone induces rapid activation of Akt/protein kinase B-mammalian target of rapamycin-p70S6K cascade through phosphatidylinositol 3-kinase in human fibroblasts. Mol Endocrinol 19:102–112 Maruyama T, Kanaji T, Nakade S, Kanno T, Mikoshiba K 1997 2APB, 2-aminoethoxydiphenyl borate, a membrane-penetrable modulator of Ins(1,4,5)P3-induced Ca2⫹ release. J Biochem (Tokyo) 122:498–505 Anderson KA, Means RL, Huang QH, Kemp BE, Goldstein EG, Selbert MA, Edelman AM, Fremeau RT, Means AR 1998 Components of a calmodulin-dependent protein kinase cascade. Molecular cloning, functional characterization and cellular localization of Ca2⫹/calmodulindependent protein kinase kinase ␤. J Biol Chem 273: 31880–31889 Stahmann N, Woods A, Carling D, Heller R 2006 Thrombin activates AMP-activated protein kinase in endothelial cells via a pathway involving Ca2⫹/calmodulin-dependent protein kinase kinase ␤. Mol Cell Biol 26:5933–5945 Tamas P, Hawley SA, Clarke RG, Mustard KJ, Green K, Hardie DG, Cantrell DA 2006 Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2⫹ in T lymphocytes. J Exp Med 203:1665–1670 Lin HY, Davis FB, Gordinier JK, Martino LJ, Davis PJ 1999 Thyroid hormone induces activation of mitogenactivated protein kinase in cultured cells. Am J Physiol 276:C1014–C1024 D’Arezzo S, Incerpi S, Davis FB, Acconcia F, Marino M, Farias RN, Davis PJ 2004 Rapid nongenomic effects of 3,5,3⬘-triiodo-L-thyronine on the intracellular pH of L-6 myoblasts are mediated by intracellular calcium mobilization and kinase pathways. Endocrinology 145: 5694–5703 Lei J, Mariash CN, Ingbar DH 2004 3,3⬘,5-Triiodo-L-thyronine up-regulation of Na,K-ATPase activity and cell surface expression in alveolar epithelial cells is Src kinase- and phosphoinositide 3-kinase-dependent. J Biol Chem 279:47589–47600 Shibusawa N, Hashimoto K, Nikrodhanond AA, Liberman MC, Applebury ML, Liao XH, Robbins JT, Refetoff S, Cohen RN, Wondisford FE 2003 Thyroid hormone action

Yamauchi et al. • Thyroid Hormone Activates AMPK

31.

32. 33. 34.

in the absence of thyroid hormone receptor DNA-binding in vivo. J Clin Invest 112:588–597 Kambe F, Nomura Y, Okamoto T, Seo H 1996 Redox regulation of thyroid-transcription factors, Pax-8 and TTF-1, is involved in their increased DNA-binding activities by thyrotropin in rat thyroid FRTL-5 cells. Mol Endocrinol 10:801–812 Pastor R, Bernal J, Rodriguez-Pena A 1994 Unliganded c-erbA/thyroid hormone receptor induces trkB expression in neuroblastoma cells. Oncogene 9:1081–1089 Lebel JM, Dussault JH, Puymirat J 1994 Overexpression of the ␤1 thyroid receptor induces differentiation in neuro-2a cells. Proc Natl Acad Sci USA 91:2644–2648 Green H, Kehinde O 1975 An established preadipose cell line and its differentiation in culture. II. Factors affecting the adipose conversion. Cell 5:19–27

Mol Endocrinol, April 2008, 22(4):893–903 903

35. Cao X, Kambe F, Lu X, Kobayashi N, Ohmori S, Seo H 2005 Glutathionylation of two cysteine residues in paired domain regulates DNA binding activity of Pax-8. J Biol Chem 280:25901–25906 36. Lu X, Kambe F, Cao X, Yoshida T, Ohmori S, Murakami K, Kaji T, Ishii T, Zadworny D, Seo H 2006 DHCR24knockout embryonic fibroblasts are susceptible to serum withdrawal-induced apoptosis because of dysfunction of caveolae and insulin-Akt-Bad signaling. Endocrinology 147:3123–3132 37. Kozaki Y, Kambe F, Hayashi Y, Ohmori S, Seo H, Kumazawa T, Mizumura K 2007 Molecular cloning of prostaglandin EP3 receptors from canine sensory ganglia and their facilitatory action on bradykinin-induced mobilization of intracellular calcium. J Neurochem 100:1636–1647

Molecular Endocrinology is published monthly by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the endocrine community.