Osteoblasts are the principal bone-forming cells. As such, osteoblasts have enhanced demand for amino acids to sustain high rates of matrix synthesis associated with bone formation. The precise systems utilized by osteoblasts to meet these synthetic demands are not well understood. WNT signaling is known to rapidly stimulate glutamine uptake during osteoblast differentiation. Using a cell biology approach, we identified two amino acid transporters, γ(+)-LAT1 and ASCT2 (encoded by Slc7a7 and Slc1a5, respectively), as the primary transporters of glutamine in response to WNT. ASCT2 mediates the majority of glutamine uptake, whereas γ(+)-LAT1 mediates the rapid increase in glutamine uptake in response to WNT. Mechanistically, WNT signals through the canonical β-catenin (CTNNB1)-dependent pathway to rapidly induce Slc7a7 expression. Conversely, Slc1a5 expression is regulated by the transcription factor ATF4 downstream of the mTORC1 pathway. Targeting either Slc1a5 or Slc7a7 using shRNA reduced WNT-induced glutamine uptake and prevented osteoblast differentiation. Collectively, these data highlight the critical nature of glutamine transport for WNT-induced osteoblast differentiation.

This article has an associated First Person interview with the joint first authors of the paper.

Osteoblasts are the primary bone forming cell responsible for producing and secreting type I collagen and other proteins that comprise the bone matrix. A constant supply of amino acids is required to maintain high rates of protein and matrix synthesis associated with bone anabolism. To fulfill this demand, osteoblasts must maximize the production or acquisition of amino acids. Indeed, recent evidence links amino acid uptake and metabolism to osteoblast function (Elefteriou et al., 2006; Karner et al., 2015; Rached et al., 2010; Yu et al., 2019). However, little is known about the transporters mediating amino acid uptake, nor their regulation during bone development.

The WNT family of secreted glycoproteins are critical regulators of osteoblast differentiation (Babij et al., 2003; Bennett et al., 2005; Day et al., 2005; Gong et al., 2001; Hu et al., 2005; Rodda and McMahon, 2006; Tu et al., 2007). WNTs activate multiple intracellular signaling cascades to induce osteoblast differentiation and modulate osteoblast activity. In the canonical pathway, WNT regulates the stability of the transcriptional coactivator β-catenin (CTNNB1), a critical regulator of osteoblast specification and differentiation (Day et al., 2005; Hu et al., 2005; MacDonald and He, 2012; Rodda and McMahon, 2006). In the absence of WNT, β-catenin is phosphorylated by the β-catenin destruction complex and targeted for proteasomal degradation. WNT stimulation inhibits the destruction complex, resulting in stabilization of β-catenin, which can then translocate into the nucleus and induce expression of target genes (for example, Tcf7) (Angers and Moon, 2009; Clevers, 2006; Clevers and Nusse, 2012; Roose et al., 1999). WNTs can also regulate osteoblast differentiation independently of β-catenin through the serine/threonine kinase mechanistic target of rapamycin (mTOR) (Chen et al., 2014; Inoki et al., 2006; Karner et al., 2015). Indeed, mTORC1 is critical for preosteoblasts to increase protein synthesis and differentiate into mature osteoblasts (Chen and Long, 2015; Fitter et al., 2017; Karner et al., 2017; Lim et al., 2016; Xian et al., 2012). We previously discovered that WNT stimulates glutamine uptake and catabolism necessary for osteoblast differentiation and bone formation (Karner et al., 2016, 2015). Mechanistically, it is not clear how WNT stimulates glutamine uptake in osteoblasts.

Glutamine transport is facilitated by a diverse array of membrane-tethered amino acid transporters categorized into discrete transport systems based on substrate specificity, kinetics and ion and pH dependence (Pochini et al., 2014). Glutamine uptake can occur in a Na+-dependent or -independent manner mediated by Systems ASC, A, γ(+)-L and N, or System L, respectively (Biltz et al., 1983; Bode, 2001; Jacob et al., 1986; Mackenzie et al., 2003; Tamarappoo et al., 1992, 1997; Taylor et al., 1992). Na+-dependent transport is subdivided by the strict requirement of Na+ [Systems ASC, A and γ(+)-L] or the substitution of Li+ for Na+ (System N). Finally, inhibitors can further subdivide Na+-dependent transporters, with System A being sensitive to 2-(methylamino)isobutyric acid (MeAIB) and System ASC to γ-glutamyl-p-nitroanilide (GPNA) (Esslinger et al., 2005; Freeman et al., 1999; Mackenzie et al., 2003). Although a great deal is known about the functional characteristics of these transporter systems, little is known about the specific transporters mediating glutamine uptake or their regulation in osteoblasts.

Here, we describe the biphasic regulation of glutamine uptake in response to WNT. WNT rapidly stimulates glutamine uptake that is sustained throughout osteoblast differentiation. Mechanistically, WNT activates β-catenin to rapidly stimulate glutamine uptake through Slc7a7, whereas mTORC1 regulates basal glutamine uptake through Slc1a5. These data highlight previously unknown roles for the amino acid transporters encoded by Slc7a7 and Slc1a5 and their regulation by WNT during osteoblast differentiation.

WNT rapidly stimulates glutamine uptake during osteoblast differentiation

We previously determined that WNT stimulates glutamine uptake associated with osteoblast differentiation (Karner et al., 2015). In order to understand how WNT regulates glutamine consumption, we first evaluated the kinetics of glutamine uptake in ST2 cells, a bone marrow-derived cell line that undergoes osteoblast differentiation in response to WNT (Bennett et al., 2005; Kang et al., 2007; Karner et al., 2016, 2015; Otsuka et al., 1999; Tu et al., 2007) (Fig. 1A). ST2 cells were treated with recombinant WNT3a (rWNT3a) for up to 96 h, and glutamine uptake was quantified using L-(2,3,4-3H)-glutamine. We observed a rapid and sustained increase in glutamine consumption beginning as rapidly as 6 h after rWNT3a stimulation (Fig. 1B). Glutamine consumption continued to increase throughout the course of the experiment, even 96 h after rWNT3a stimulation (Fig. 1B). Basal glutamine transport occurred primarily in a Na+-dependent manner that could not be rescued by Li+ or inhibited by the amino acid analog MeAIB in ST2 cells (Fig. 1C). These data indicate that glutamine uptake occurs primarily in a Na+-dependent manner characteristic of the amino acid transport systems System ASC and System γ(+)-L (Fig. 1D). Likewise, WNT-stimulated glutamine uptake in ST2 cells was Na+ dependent and insensitive to either MeAIB or Li+ rescue (Fig. 1C). These data indicate that WNT-induced glutamine uptake is not the result of enhanced activity of other transport systems, rather it results from increased System ASC and/or γ(+)-L activity (Fig. 1D) (Jacob et al., 1986; Tamarappoo et al., 1997; Taylor et al., 1992).

Fig. 1.

WNT signaling rapidly increaseNa+-dependent glutamine consumption in ST2 cells. (A) Alkaline phosphatase (ALPL) and von Kossa staining in ST2 cells treated with 25 ng/ml rWnt3a or vehicle (VEH) for 72 h followed by osteogenic medium for 6 days. (B) Timecourse of radiolabeled glutamine uptake in response to rWNT3A. n=6 independent trials for each time point. (C) Glutamine uptake assays performed in the presence (+) or absence (−) of sodium (Na+), MeAIB or lithium (Li+) in the uptake media in ST2 cells treated with rWNT3A or vehicle for 24 h. n=4 for each condition. (D) Relative system activity calculated from experiments in C. Graphs depict mean±s.d. *P≤0.05 (unpaired two-tailed Student's t-test).

Fig. 1.

WNT signaling rapidly increaseNa+-dependent glutamine consumption in ST2 cells. (A) Alkaline phosphatase (ALPL) and von Kossa staining in ST2 cells treated with 25 ng/ml rWnt3a or vehicle (VEH) for 72 h followed by osteogenic medium for 6 days. (B) Timecourse of radiolabeled glutamine uptake in response to rWNT3A. n=6 independent trials for each time point. (C) Glutamine uptake assays performed in the presence (+) or absence (−) of sodium (Na+), MeAIB or lithium (Li+) in the uptake media in ST2 cells treated with rWNT3A or vehicle for 24 h. n=4 for each condition. (D) Relative system activity calculated from experiments in C. Graphs depict mean±s.d. *P≤0.05 (unpaired two-tailed Student's t-test).

Slc1a5 and Slc7a7 regulate basal and WNT stimulated glutamine uptake

To determine how WNT signaling regulated glutamine uptake, we first evaluated the mRNA expression of genes encoding glutamine transporters in ST2 cells. To do this, we analyzed our previously generated RNAseq dataset of ST2 cells treated with rWNT3a for up to 72 h (Karner et al., 2016). In unstimulated ST2 cells, the System ASC transporter alanine serine cysteine transporter 2 (ASCT2, encoded by Slc1a5) was the highest expressed glutamine transporter in either System ASC or γ(+)-L (Table 1). Conversely, the System γ(+)-L transporter γ(+)-L transporter 1 [γ(+)-LAT1, encoded by Slc7a7] was expressed at low levels in unstimulated ST2 cells (Table 1). It is important to note that ASCT2 and γ(+)-LAT1 are the only members of their respective transport systems that accept glutamine as a substrate. Despite being expressed at lower levels in unstimulated ST2 cells, Slc7a7 was significantly increased by WNT treatment at 6, 24 and 72 h (Table 1). On the other hand, expression of Slc1a5 was not significantly increased until after 72 h WNT treatment (Table 1). We confirmed the temporal regulation of both Slc1a5 and Slc7a7 using reverse transcription qPCR (RT-qPCR). Both Slc7a7 and Slc1a5 were significantly induced by WNT signaling, albeit with differing kinetics (Fig. 2A). Slc7a7 was induced rapidly, with peak induction occurring within 6–24 h of WNT stimulation. By 96 h, Slc7a7 had returned to baseline expression (Fig. 2A). Conversely, Slc1a5 was significantly induced beginning 72 h after WNT stimulation (Fig. 2A). These data demonstrate that WNT regulates two amino acid transporters with differing kinetics in ST2 cells.

Table 1. Amino acid transporter expression in ST2 cells

Table 1. Amino acid transporter expression in ST2 cells
Table 1. Amino acid transporter expression in ST2 cells
Fig. 2.

Wnt signaling induces Slc7a7 and Slc1a5 with disparate kinetics. (A) RT-qPCR analyses of Slc7a7 and Slc1a5 expression in ST2 cells treated with WNT3a for up to 96 h. Fold induction±s.d. relative to 0 h time point for three independent experiments. (B) Measurements of radiolabeled glutamine uptake in response to WNT3a in the presence of GPNA. (C) Graphical depiction of WNT-induced glutamine uptake calculated from data shown in B (Veh, vehicle). (D) Western blotting analyses of ASCT2 protein expression level in ST2 cells infected with shRNA targeting Slc1a5 or RFP as a control. ASCT2 normalized to β-actin. Fold change±s.d. relative to shRFP control is shown for three independent experiments. (E,F) Effect of Slc1a5 knockdown (1a5) on glutamine uptake in response to WNT3a. (G) Western blotting analyses of γ(+)-LAT1 protein expression in ST2 cells infected with shRNA targeting Slc7a7 or RFP as a control. γ(+)-LAT1 normalized to β-actin. Fold change±s.d. relative to shRFP control is shown for three independent experiments. (H,I) Effect of Slc7a7 knockdown (7a7) on glutamine uptake in response to WNT3a. Three independent trials were performed in each experiment. *P≤0.05, **P≤0.005, ***P≤0.0005, ****P≤0.00005, by one-way ANOVA with Tukey's post hoc test (B,E,H) or by an unpaired two-tailed Student's t-test (A,C,D,F,G,I).

Fig. 2.

Wnt signaling induces Slc7a7 and Slc1a5 with disparate kinetics. (A) RT-qPCR analyses of Slc7a7 and Slc1a5 expression in ST2 cells treated with WNT3a for up to 96 h. Fold induction±s.d. relative to 0 h time point for three independent experiments. (B) Measurements of radiolabeled glutamine uptake in response to WNT3a in the presence of GPNA. (C) Graphical depiction of WNT-induced glutamine uptake calculated from data shown in B (Veh, vehicle). (D) Western blotting analyses of ASCT2 protein expression level in ST2 cells infected with shRNA targeting Slc1a5 or RFP as a control. ASCT2 normalized to β-actin. Fold change±s.d. relative to shRFP control is shown for three independent experiments. (E,F) Effect of Slc1a5 knockdown (1a5) on glutamine uptake in response to WNT3a. (G) Western blotting analyses of γ(+)-LAT1 protein expression in ST2 cells infected with shRNA targeting Slc7a7 or RFP as a control. γ(+)-LAT1 normalized to β-actin. Fold change±s.d. relative to shRFP control is shown for three independent experiments. (H,I) Effect of Slc7a7 knockdown (7a7) on glutamine uptake in response to WNT3a. Three independent trials were performed in each experiment. *P≤0.05, **P≤0.005, ***P≤0.0005, ****P≤0.00005, by one-way ANOVA with Tukey's post hoc test (B,E,H) or by an unpaired two-tailed Student's t-test (A,C,D,F,G,I).

We next sought to determine the necessity of these amino acid transporters for glutamine uptake. We first evaluated the role of Slc1a5 in glutamine uptake because it was highly expressed in ST2 cells (Table 1). To do this, we used GPNA, a specific inhibitor of Slc1a5/ASCT2 (Esslinger et al., 2005). GPNA treatment reduced basal glutamine uptake by 77% indicating the majority of glutamine uptake is mediated by Slc1a5/ASCT2 (Fig. 2B). We next sought to determine the effects on WNT-induced glutamine uptake. Interestingly, despite significantly reducing basal glutamine uptake, glutamine consumption was stimulated equally by WNT in the presence or absence of GPNA (Fig. 2B,C). To confirm the specificity of GPNA, we targeted Slc1a5 using shRNA. This approach significantly reduced both ASCT2 protein and basal glutamine uptake, similar to the effects of GPNA treatment (Fig. 2D,E). Similar to the results of the GPNA experiment, glutamine uptake was stimulated equally by WNT in both control and ASCT2-knockdown cells (Fig. 2E,F). These data indicate that Slc1a5/ASCT2 mediates the majority of glutamine uptake in ST2 cells but is not responsible for the acute increase in glutamine consumption in response to WNT. We next sought to determine whether Slc7a7 mediates glutamine uptake in ST2 cells. To do this, we targeted Slc7a7 using shRNA. This approach effectively reduced γ(+)-LAT1 protein levels (Fig. 2G). Slc7a7 knockdown minimally affected basal glutamine uptake but completely abrogated increased glutamine consumption in response to WNT stimulation (Fig. 2H,I). Collectively these data indicate WNT signaling rapidly increases Slc7a7 expression to increase glutamine uptake whereas Slc1a5 is responsible for the majority of glutamine uptake in ST2 cells.

Glutamine uptake and Slc7a7 are regulated by canonical WNT signaling

We next sought to understand how WNT signaling regulated Slc7a7 expression. The rapid induction kinetics of Slc7a7 expression suggested it may be regulated directly by β-catenin. Inhibition of LRP5 and LRP6 using recombinant DKK1 (rDKK1) reduced WNT-induced β-catenin stabilization and prevented the induction of alkaline phosphatase (Fig. 3A). Moreover, DKK1 treatment prevented WNT induction of known β-catenin target genes (e.g. Tcf7) as well as of Slc7a7 (Fig. 3B,C). Expression of a dominant-negative form of TCF4 that is unable to interact with β-catenin also prevented induction of Slc7a7, suggesting that Slc7a7 is a β-catenin target gene (data not shown). To test the role of β-catenin directly, we targeted the gene encoding β-catenin (Catnnb1) using shRNA (Fig. 3D). β-catenin knockdown prevented osteoblast differentiation, as shown by inhibition of alkaline phosphatase induction (Fig. 3D). Importantly, β-catenin knockdown significantly reduced the basal expression of Slc7a7 and prevented the induction of Slc7a7 in response to WNT treatment (Fig. 3E). Moreover, β-catenin knockdown reduced WNT-induced glutamine uptake without affecting Slc1a5 expression (Fig. 3F and data not shown). These data indicate that β-catenin is necessary for the induction of Slc7a7 and acute glutamine consumption in response to WNT. We next sought to determine the sufficiency of β-catenin by inhibiting GSK3β using LiCl (Klein and Melton, 1996). LiCl treatment stabilized β-catenin and induced both alkaline phosphatase staining and Tcf7 mRNA expression similar to WNT treatment (Fig. 3G,H). Moreover, LiCl stimulated both Slc7a7 mRNA and γ(+)-Lat1 protein expression and increased glutamine uptake similar to WNT treatment (Fig. 3G–I). Importantly, LiCl did not increase Slc1a5 mRNA expression (Fig. 3H). These data indicate that β-catenin-dependent WNT signaling rapidly stimulates glutamine uptake via transcriptional upregulation of Slc7a7.

Fig. 3.

Slc7a7 is regulated by canonical WNT signaling. (A–C) Effect of rDKK1 treatment on WNT3a-induced β-catenin expression and alkaline phosphatase (ALPL) staining (A) or Tcf7 (B) and Slc7a7 (C) gene expression in ST2 cells, compared with vehicle-treated controls (VEH). (D–F) Effect of β-catenin knockdown (shCatnnb1) on WNT3a-induced β-catenin expression and alkaline phosphatase staining (D), Slc7a7 gene expression (E), or glutamine uptake (F), compared with the effect of control shRNA treatment (shLacz, shRFP). (G–I) Effect of LiCl treatment on protein expression, alkaline phosphatase staining (G), gene expression (H) and glutamine uptake (I). In A,D,G, β-catenin and γ(+)-LAT1 levels are normalized to α-tubulin, and fold change±s.d. relative to untreated negative control samples are shown for three independent experiments (N.D., not determined). Data in B,C,E,F,H,I are mean±s.d. of three independent experiments. *P≤0.05, #P≤0.05; **P≤0.005, ***P≤0.0005, ****P≤0.00005 [one-way ANOVA with Tukey's post hoc test (A–F) or unpaired two-tailed Student's t-test (G–I)].

Fig. 3.

Slc7a7 is regulated by canonical WNT signaling. (A–C) Effect of rDKK1 treatment on WNT3a-induced β-catenin expression and alkaline phosphatase (ALPL) staining (A) or Tcf7 (B) and Slc7a7 (C) gene expression in ST2 cells, compared with vehicle-treated controls (VEH). (D–F) Effect of β-catenin knockdown (shCatnnb1) on WNT3a-induced β-catenin expression and alkaline phosphatase staining (D), Slc7a7 gene expression (E), or glutamine uptake (F), compared with the effect of control shRNA treatment (shLacz, shRFP). (G–I) Effect of LiCl treatment on protein expression, alkaline phosphatase staining (G), gene expression (H) and glutamine uptake (I). In A,D,G, β-catenin and γ(+)-LAT1 levels are normalized to α-tubulin, and fold change±s.d. relative to untreated negative control samples are shown for three independent experiments (N.D., not determined). Data in B,C,E,F,H,I are mean±s.d. of three independent experiments. *P≤0.05, #P≤0.05; **P≤0.005, ***P≤0.0005, ****P≤0.00005 [one-way ANOVA with Tukey's post hoc test (A–F) or unpaired two-tailed Student's t-test (G–I)].

Basal glutamine uptake and Slc1a5 expression is regulated by mTORC1 signaling

We next evaluated the regulation of Slc1a5 expression by WNT. Unlike Slc7a7 and other canonical WNT target genes, which are rapidly induced by WNT, Slc1a5 mRNA levels were not significantly increased until 72 h after WNT treatment (Fig. 2A and Table 1). Indeed, we observed no change in Slc1a5 expression in β-catenin-knockdown cells or in response to LiCl treatment, suggesting Slc1a5 is not regulated by canonical WNT–β-catenin signaling (Fig. 3H and data not shown). Rather, the induction of Slc1a5 followed a timecourse reminiscent of protein anabolism genes induced by WNT through secondary activation of the transcription factor ATF4 (Karner et al., 2015). We have previously determined that WNT signals through mTORC1 to activate ATF4 and protein anabolism genes during osteoblast differentiation (Karner et al., 2015). Here, we observed a significant increase in ATF4 protein expression after 72 h of WNT treatment (Fig. 4A). Inhibition of mTORC1 signaling using rapamycin prevented ATF4 protein induction by WNT (Fig. 4A). Importantly, rapamycin treatment reduced ASCT2 expression in both unstimulated and WNT treated ST2 cells (Fig. 4A). Consistent with decreased ASCT2 expression, rapamycin significantly reduced glutamine uptake under both basal conditions and in response to WNT stimulation (Fig. 4B). Despite a significant reduction in overall glutamine uptake, WNT was able to stimulate glutamine consumption in the presence of rapamycin (Fig. 4B,C). This is likely due to normal induction of Slc7a7, because rapamycin treatment did not affect WNT induction of Slc7a7 despite completely abrogating Slc1a5 induction by WNT (Fig. 4D,E). These data indicate that mTORC1 activity is required for both glutamine uptake and the normal expression of both ATF4 and ASCT2. We next sought to determine whether ATF4 activity is required for Slc1a5 induction by WNT. Atf4 knockdown using shRNA reduced both ATF4 protein expression and Slc1a5 mRNA expression in ST2 cells (Fig. 4F,G). Moreover, Atf4 expression was necessary for Slc1a5 induction by WNT, because Atf4 knockdown completely prevented the induction of Slc1a5 by WNT (Fig. 4F,G). Likewise, Atf4 knockdown significantly reduced basal glutamine uptake (Fig. 4H). Similar to the rapamycin treatment, Atf4 knockdown did not affect either WNT-induced glutamine uptake or Slc7a7 induction by WNT (Fig. 4I,J). Collectively, these data indicate that ATF4 is critical for the majority of glutamine uptake by regulating Slc1a5 mRNA expression downstream of mTORC1.

Fig. 4.

Slc1a5 is regulated by non-canonical WNT signaling. (A–D) Effect of rapamycin treatment on WNT3a-induced protein expression (A), glutamine uptake (B,C) or mRNA expression (D,E). In A, S6 phosphorylation at Ser240/244 (S6 pSer240/244) is shown as a positive control for rapamycin treatment. pSer240/244 S6 normalized to total S6, ATF4 and ASCT2 normalized to β-actin. Fold change±s.d. relative to untreated negative control samples are shown, calculated from three independent experiments (N.D., not determined). In B, data are glutamine uptake as mean±s.d. relative to vehicle-treated negative control samples, calculated from three independent experiments. In C–E, data are fold change±s.d. for WNT3a over vehicle calculated from three independent experiments. (F) Western blotting analysis of ATF4 expression following the indicated shRNA treatments. β-actin is shown as a loading control. (G–J) Effect of Atf4 knockdown on WNT-induced Slc1a5 expression (G), glutamine uptake (H,I) or Slc7a7 expression (I), compared with the effect of control shRNA treatment (shLacZ, shRFP). Data are presented as mean±s.d from three independent experiments. *P≤0.05, **P≤0.005, ***P≤0.0005, ****P≤0.00005 [one-way ANOVA with Tukey's post hoc test (A,B,G,H) or unpaired two-tailed Student's t-test (C–F,I,J)].

Fig. 4.

Slc1a5 is regulated by non-canonical WNT signaling. (A–D) Effect of rapamycin treatment on WNT3a-induced protein expression (A), glutamine uptake (B,C) or mRNA expression (D,E). In A, S6 phosphorylation at Ser240/244 (S6 pSer240/244) is shown as a positive control for rapamycin treatment. pSer240/244 S6 normalized to total S6, ATF4 and ASCT2 normalized to β-actin. Fold change±s.d. relative to untreated negative control samples are shown, calculated from three independent experiments (N.D., not determined). In B, data are glutamine uptake as mean±s.d. relative to vehicle-treated negative control samples, calculated from three independent experiments. In C–E, data are fold change±s.d. for WNT3a over vehicle calculated from three independent experiments. (F) Western blotting analysis of ATF4 expression following the indicated shRNA treatments. β-actin is shown as a loading control. (G–J) Effect of Atf4 knockdown on WNT-induced Slc1a5 expression (G), glutamine uptake (H,I) or Slc7a7 expression (I), compared with the effect of control shRNA treatment (shLacZ, shRFP). Data are presented as mean±s.d from three independent experiments. *P≤0.05, **P≤0.005, ***P≤0.0005, ****P≤0.00005 [one-way ANOVA with Tukey's post hoc test (A,B,G,H) or unpaired two-tailed Student's t-test (C–F,I,J)].

Glutamine uptake is required for osteoblast differentiation

We next sought to determine the role of these glutamine transporters during WNT-induced osteoblast differentiation. Knockdown of Slc7a7 using shRNA reduced the induction of both early osteoblast marker genes (exemplified by Akp2, also known as Alpl) and terminal osteoblast marker genes (exemplified by Bglap) and prevented matrix mineralization in response to WNT (Fig. 5A–D). Interestingly, Slc1a5 knockdown did not affect early osteoblast marker gene induction (e.g. Akp2) but specifically reduced the induction of terminal osteoblast marker genes (e.g. Bglap) and prevented matrix mineralization in response to WNT (Fig. 5E–H). These data indicate that both Slc7a7 and Slc1a5 are required for WNT-induced osteoblast differentiation in ST2 cells.

Fig. 5.

Slc7a7 and Slc1a5 are both required for WNT-induced osteoblast differentiation. Effect of Slc7a7 (A–D) or Slc1a5 knockdown (E–H) on WNT-induced gene expression (A–C,E–G), or alkaline phosphatase (ALPL) and von Kossa staining (D,H), compared with the effects of control shRNA treatment (shRFP). Data are presented as mean fold change±s.d. for three independent experiments. *P≤0.05 (one-way ANOVA with Tukey's post hoc test).

Fig. 5.

Slc7a7 and Slc1a5 are both required for WNT-induced osteoblast differentiation. Effect of Slc7a7 (A–D) or Slc1a5 knockdown (E–H) on WNT-induced gene expression (A–C,E–G), or alkaline phosphatase (ALPL) and von Kossa staining (D,H), compared with the effects of control shRNA treatment (shRFP). Data are presented as mean fold change±s.d. for three independent experiments. *P≤0.05 (one-way ANOVA with Tukey's post hoc test).

Increased matrix synthesis associated with bone formation increases the demand for amino acids. It stands to reason that osteoblasts must increase amino acid production or acquisition to meet this biosynthetic demand. Here, we describe the intricate regulation of glutamine acquisition by WNT signaling in an osteoblast progenitor cell line. We identified genes encoding two amino acid transporters, Slc1a5 and Slc7a7, responsible for the majority of glutamine uptake in ST2 cells undergoing osteoblast differentiation in response to WNT. Slc1a5 encodes the primary glutamine transporter in ST2 cells whereas Slc7a7 mediates WNT-stimulated glutamine uptake associated with osteoblast differentiation. These transporters appear to work in concert to provide sufficient glutamine, and likely other amino acids, to initiate and sustain osteoblast differentiation and matrix production in vitro.

Osteoblast differentiation is characterized by rapid proliferation of osteoblast progenitors followed by differentiation into mature matrix-producing osteoblasts. This process is associated with increased glutamine consumption and metabolism (Fig. 1) (Karner et al., 2015; Yu et al., 2019). Blocking glutamine consumption (Fig. 5) or metabolism prevents osteoblast differentiation and matrix formation (Brown et al., 2011; Karner et al., 2015; Yu et al., 2019). It is not clear why osteoblasts have such an acute requirement for glutamine to facilitate these processes. Osteoblast differentiation is associated with altered energetic and biosynthetic demands (Guntur et al., 2014; Karner and Long, 2017; Riddle and Clemens, 2017). For example, proliferating cells require nucleotides and must duplicate their cell mass in order to divide (Hosios et al., 2016). Similarly, differentiation and matrix production is associated with increased protein synthesis and secretion, which may increase reactive oxygen species and oxidative stress detrimental to osteoblast differentiation (Almeida et al., 2007; Mody et al., 2001). Glutamine is a multifunctional amino acid uniquely suited to fulfill these anabolic demands. For example, glutamine-derived nitrogen is important for the de novo synthesis of both nucleotides and amino acids. Similarly, glutamine-derived carbon is used for the synthesis of both amino acids and the antioxidant glutathione. Finally, glutamine-derived α-ketoglutarate can provide energy to fulfill energetic demands through entry into the TCA cycle (Newsholme et al., 2003). Thus, osteoblasts likely utilize glutamine disparately during differentiation to fulfill distinct metabolic purposes.

WNT is a potent regulator of osteoblast differentiation and bone formation (Cui et al., 2011; Hill et al., 2005). One of the earliest events associated with WNT-induced osteoblast differentiation is increased glutamine consumption (Fig. 1; Karner et al., 2015). It is interesting to note that glutamine uptake is not only rapidly increased but also sustained throughout the differentiation process. Here, we identified two glutamine transporters encoded by Slc1a5 and Slc7a7 that mediate glutamine uptake in ST2 cells. Slc1a5 is critical for the majority of glutamine uptake (Fig. 2). Conversely, Slc7a7 is responsible for mediating the acute increase in glutamine uptake in response to WNT. Importantly, the transcription of these transporters is regulated disparately by WNT. First, expression of Slc7a7 is upregulated within 6 h, whereas Slc1a5 expression is not increased until 72 h after WNT stimulation. Mechanistically, β-catenin is both necessary and sufficient for the rapid induction of Slc7a7 expression and the acute increase in glutamine uptake in response to WNT. Conversely, β-catenin is dispensable for Slc1a5 expression and basal glutamine consumption in ST2 cells (Fig. 3 and data not shown). Rather, Slc1a5 expression is regulated downstream of mTORC1 and ATF4 in ST2 cells (Fig. 4). We previously determined that WNT induces a GCN2-dependent integrated stress response (ISR) through mTORC1 (Karner et al., 2015). We show here that the ISR transcriptional effector ATF4 is critical for both basal and WNT-induced Slc1a5 expression and glutamine uptake. This is consistent with recent data demonstrating that Slc1a5 is directly regulated by ATF4 (Han et al., 2013; Hu et al., 2020).

Collectively, these data support a biphasic model in which WNT signaling regulates glutamine consumption via canonical and noncanonical pathways to facilitate osteoblast differentiation. Interestingly, inhibiting Slc7a7, but not Slc1a5, inhibited Akp2 induction, whereas inhibition of either transporter inhibited terminal osteoblast differentiation and matrix mineralization. The precise mechanism underlying this discrepancy is not clear; however, it is important to note that in addition to glutamine, ASCT2 can transport alanine, serine and asparagine. Conversely, γ(+)-Lat1 mediates the influx of ornithine, arginine and lysine in exchange for the efflux of cationic amino acids (Chillaron et al., 1996; Pfeiffer et al., 1999; Torrents et al., 1999), which may be contributing to the disparate early osteoblast differentiation phenotypes observed. It will be important to elucidate the substrates of these amino acid transporters in osteoblast progenitors and determine the precise role they play during WNT-induced osteoblast differentiation.

Cell culture

The bone marrow-derived cell line, ST2 (RRID: CVCL_2205) was plated at 40,000 cells/ml in α-MEM (GIBCO) supplemented with 10% FBS (Invitrogen). In WNT treatment experiments, 25 ng/ml WNT3a (TIME Bioscience) or vehicle control (0.1% BSA in PBS) was supplemented in the α-MEM. For LiCl treatment, 20 mM LiCl or vehicle control (20 mM NaCl) was added in the growth medium. When inhibitors were used, cells were pretreated with corresponding inhibitors for 30 min before any other treatment. In indicated experiments, growth medium was supplemented with 100 nM rapamycin (Sigma-Aldrich), 250 ng/ml DKK1 (R&D) or 0.03 mM GPNA (MP Biomedicals), or the respective vehicle (DMSO, 0.1% BSA in PBS or 1 M HCl for Rapamycin, DKK1 and GPNA, respectively) as a control. For WNT3a-induced mineralization, cells were treated with WNT3a for 72 h followed by osteogenic medium [α-MEM supplemented with 50 mg/ml ascorbic acid (Sigma-Aldrich) and 10 mM β-glycerophosphate (Sigma-Aldrich)] for 6 days. Osteoblast differentiation was assayed by visualizing alkaline phosphatase activity using 5-bromo-4-chloro-3′-indolyl-phosphate/Nitro Blue tetrazolium (BCIP/NPT; Katagiri et al., 1994) or von Kossa staining of deposited calcium phosphate (Rungby et al., 1993). The alkaline phosphatase assay was performed 24 h after WNT stimulation. Von Kossa staining was performed 6 days after addition of osteogenic medium.

Glutamine uptake assay

Cells were washed with PBS and two additional washes with Krebs Ringer Hepes (KRH; 120 mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 25 mM NaHCO3, 5 mM HEPES, pH 8 and 1 mM D-glucose). Cells were then treated for 5 min with KRH containing 4 μCi/ml L-(2,3,4-3H)-glutamine. Glutamine uptake and cellular metabolism was terminated by washing with ice-cold KRH and scraping cells with 1 ml ice-cold dH2O. The lysate was centrifuged, and counts per minute (CPM) were measured using a Beckman LS6500 scintillation counter. All results are normalized to cell number. In experiments to measure glutamine transport system activity, the recipe for KRH was altered as follows: complete KRH (120 mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 0.4 mM KH2PO4, 5 mM HEPES, 1 mM D-glucose, pH 8), complete KRH plus MeAIB (120 mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 0.4 mM KH2PO4, 5 mM HEPES, 5 mM MeAIB, 1 mM D-glucose, pH 8), complete KRH plus GPNA (120 mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 0.4 mM KH2PO4, 5 mM HEPES, 0.3 mM GPNA, 1 mM D-glucose, pH 8), Na+-free KRH (120 mM choline chloride, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 0.4 mM KH2PO4, 5 mM HEPES, 1 mM D-glucose, pH 8), Na+-free plus Li+ KRH (120 mM LiCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 0.4 mM KH2PO4, 5 mM HEPES, 1 mM D-glucose, pH 8). We calculated the relative glutamine transport system activity as follows:
formula
formula
formula
formula

shRNA knockdowns

Lentiviral vectors were obtained from the shRNA consortium at Washington University School of Medicine. All knockdown results were confirmed using two or more unrelated shRNA constructs. The shRNA sequence listed first was shown in the results. The lentiviral vector pLKOpuro (RRID: Addgene_8453) was modified to express shRNAs targeting Slc1a5 (5′-CCTGTAGAGTTCTCTACCCTT-3′, 5′-GCAGTGTTCATCGCACAACTA-3′), Slc7a7 (5′-GCTACATGTTTCAGACTTCAT-3′, 5′-GCCATCTGTATGGTTCATG-3′), Atf4 (5′-CCAGAGCATTCCTTTAGTTTA-3′, 5′-CCTCTAGTCCAAGAGACTAAT-3′), Catnnb1 (5′-GCGTTATCAAACCCTAGCCTT-3′, 5′-CCATCACAGATGTTGAAACAT-3′), or either RFP (5′-ACAACAGCCACAACGTCTATA-3′) or LacZ (5′-GCGATCGTAATCACCCGAGTG-3′) as a negative control. The shRNA-expressing lentiviral vector was co-transfected in 293T cells with the plasmids pMD2.g (RRID: Addgene_12259) and psPax2 (RRID: Addgene_12260). Virus-containing medium was collected and filtered. ST2 cells were infected for 24 h and recovered for 24 h in regular medium prior to further treatment.

RNA isolation and RT-qPCR

Total RNA was isolated using an RNAeasy kit with on-column DNase treatment (Qiagen). 500 ng of total RNA was reverse transcribed using the Iscript cDNA Synthesis kit (Bio-Rad). Reactions were set up in technical and biological triplicate in a 96-well format on an ABI Quantstudio 3 using SYBR Green chemistry (SsoAdvanced; Bio-Rad). The PCR conditions were 95°C for 3 min followed by 35 cycles of 95°C for 10 s and 60°C for 30 s. Gene expression was normalized to 18S rRNA, and relative expression was calculated using the 2−(ΔΔCt) method. Primers were used at 0.1 µM, and their sequences are listed in Table S1. PCR efficiency was optimized, and melting curve analyses of products were performed to ensure reaction specificity.

Western blotting

ST2 cells were scraped in lysis buffer containing 50 mM Tris (pH 7.4), 15 mM NaCl, 0.5% NP-40, 0.1% SDS and 0.1% sodium deoxycholate with a protease and phosphatase inhibitor tablet (Roche). Protein concentration was quantified using the BCA method (Pierce). Proteins (20 µg) were resolved on 12% polyacrylamide gels and transferred onto Immuno-Blot PVDF membrane (Bio-Rad). Proteins were detected using the following specific antibodies: anti-ASCT2 (RRID: AB_10891440), anti-Slc7a7 (RRID: AB_2576546), anti-α-tubulin (RRID: AB_2619646), anti-ATF4 (RRID: AB_2058752), anti-β-actin (RRID: AB_330288), anti-phospho-S6 (Ser240/244) (RRID: AB_10694233), anti-S6 (RRID: AB_331355) and anti-β-catenin (RRID: AB_634603). Further antibody information is provided in Table S2. The membranes were blocked for 1 h at room temperature in 5% milk powder in TBS with 0.1% Tween (TBST) and then incubated at 4°C with the primary antibody overnight. Membranes were washed three times with TBST and further incubated with anti-rabbit IgG, HRP-linked antibody (RRID: AB_2099233) in 5% milk (in TBST) for 1 h at room temperature. All blots were developed using enhanced chemiluminescence (Clarity Substrate Kit, Bio-Rad). Each experiment was repeated with a minimum of three independently prepared protein samples.

Statistics

Statistical significance was determined by one-way ANOVA or by an unpaired two-tailed Student's t-test.

The authors would like to thank all members of the Karner laboratory for critical comments on the manuscript.

Author contributions

Conceptualization: C.M.K.; Methodology: L.S., D.S.; Validation: L.S., D.S.; Formal analysis: L.S., D.S.; Investigation: L.S., D.S., Y.Y., C.M.K.; Resources: F.L., C.M.K.; Data curation: L.S., C.M.K.; Writing - original draft: L.S., D.S., C.M.K.; Writing - review & editing: L.S., D.S., C.M.K.; Visualization: L.S., D.S., C.M.K.; Supervision: C.M.K.; Project administration: C.M.K.; Funding acquisition: F.L., C.M.K.

Funding

This work was supported by the National Institutes of Health (AR060456 to F.L.; AR071967 and AR076325 to C.M.K.). Deposited in PMC for release after 12 months.

Almeida
,
M.
,
Han
,
L.
,
Martin-Millan
,
M.
,
O'Brien
,
C. A.
and
Manolagas
,
S. C.
(
2007
).
Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting β-catenin from T cell factor- to forkhead box O-mediated transcription
.
J. Biol. Chem.
282
,
27298
-
27305
.
Angers
,
S.
and
Moon
,
R. T.
(
2009
).
Proximal events in Wnt signal transduction
.
Nat. Rev. Mol. Cell Biol.
10
,
468
-
477
.
Babij
,
P.
,
Zhao
,
W.
,
Small
,
C.
,
Kharode
,
Y.
,
Yaworsky
,
P. J.
,
Bouxsein
,
M. L.
,
Reddy
,
P. S.
,
Bodine
,
P. V.
,
Robinson
,
J. A.
,
Bhat
,
B.
et al. 
(
2003
).
High bone mass in mice expressing a mutant LRP5 gene
.
J. Bone Miner. Res.
18
,
960
-
974
.
Bennett
,
C. N.
,
Longo
,
K. A.
,
Wright
,
W. S.
,
Suva
,
L. J.
,
Lane
,
T. F.
,
Hankenson
,
K. D.
and
MacDougald
,
O. A.
(
2005
).
Regulation of osteoblastogenesis and bone mass by Wnt10b
.
Proc. Natl. Acad. Sci. USA
102
,
3324
-
3329
.
Biltz
,
R. M.
,
Letteri
,
J. M.
,
Pellegrino
,
E. D.
,
Palekar
,
A.
and
Pinkus
,
L. M.
(
1983
).
Glutamine metabolism in bone
.
Miner. Electrolyte Metab.
9
,
125
-
131
.
Bode
,
B. P.
(
2001
).
Recent molecular advances in mammalian glutamine transport
.
J. Nutr.
131
,
2475S
-
2485S
,
discussion 2486S-7S
.
Brown
,
P. M.
,
Hutchison
,
J. D.
and
Crockett
,
J. C.
(
2011
).
Absence of glutamine supplementation prevents differentiation of murine calvarial osteoblasts to a mineralizing phenotype
.
Calcif. Tissue Int.
89
,
472
-
482
.
Chen
,
J.
and
Long
,
F.
(
2015
).
mTORC1 signaling promotes osteoblast differentiation from preosteoblasts
.
PLoS ONE
10
,
e0130627
.
Chen
,
J.
,
Tu
,
X.
,
Esen
,
E.
,
Joeng
,
K. S.
,
Lin
,
C.
,
Arbeit
,
J. M.
,
Rüegg
,
M. A.
,
Hall
,
M. N.
,
Ma
,
L.
,
Long
,
F.
et al. 
(
2014
).
WNT7B promotes bone formation in part through mTORC1
.
PLoS Genet.
10
,
e1004145
.
Chillarón
,
J.
,
Estévez
,
R.
,
Mora
,
C.
,
Wagner
,
C. A.
,
Suessbrich
,
H.
,
Lang
,
F.
,
Gelpí
,
J. L.
,
Testar
,
X.
,
Busch
,
A. E.
,
Zorzano
,
A.
et al. 
(
1996
).
Obligatory amino acid exchange via systems bo,+-like and y+L-like. A tertiary active transport mechanism for renal reabsorption of cystine and dibasic amino acids
.
J. Biol. Chem.
271
,
17761
-
17770
.
Clevers
,
H.
(
2006
).
Wnt/β-catenin signaling in development and disease
.
Cell
127
,
469
-
480
.
Clevers
,
H.
and
Nusse
,
R.
(
2012
).
Wnt/β-catenin signaling and disease
.
Cell
149
,
1192
-
1205
.
Cui
,
Y.
,
Niziolek
,
P. J.
,
MacDonald
,
B. T.
,
Zylstra
,
C. R.
,
Alenina
,
N.
,
Robinson
,
D. R.
,
Zhong
,
Z.
,
Matthes
,
S.
,
Jacobsen
,
C. M.
,
Conlon
,
R. A.
et al. 
(
2011
).
Lrp5 functions in bone to regulate bone mass
.
Nat. Med.
17
,
684
-
691
.
Day
,
T. F.
,
Guo
,
X.
,
Garrett-Beal
,
L.
and
Yang
,
Y.
(
2005
).
Wnt/β-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis
.
Dev. Cell
8
,
739
-
750
.
Elefteriou
,
F.
,
Benson
,
M. D.
,
Sowa
,
H.
,
Starbuck
,
M.
,
Liu
,
X.
,
Ron
,
D.
,
Parada
,
L. F.
and
Karsenty
,
G.
(
2006
).
ATF4 mediation of NF1 functions in osteoblast reveals a nutritional basis for congenital skeletal dysplasiae
.
Cell Metab.
4
,
441
-
451
.
Esslinger
,
C. S.
,
Cybulski
,
K. A.
and
Rhoderick
,
J. F.
(
2005
).
Nγ-aryl glutamine analogues as probes of the ASCT2 neutral amino acid transporter binding site
.
Bioorg. Med. Chem.
13
,
1111
-
1118
.
Fitter
,
S.
,
Matthews
,
M. P.
,
Martin
,
S. K.
,
Xie
,
J.
,
Ooi
,
S. S.
,
Walkley
,
C. R.
,
Codrington
,
J. D.
,
Ruegg
,
M. A.
,
Hall
,
M. N.
,
Proud
,
C. G.
et al. 
(
2017
).
mTORC1 plays an important role in skeletal development by controlling preosteoblast differentiation
.
Mol. Cell Biol.
37
,
e00668-e00616
.
Freeman
,
T. L.
,
Ngo
,
H. Q.
and
Mailliard
,
M. E.
(
1999
).
Inhibition of system A amino acid transport and hepatocyte proliferation following partial hepatectomy in the rat
.
Hepatology
30
,
437
-
444
.
Gong
,
Y.
,
Slee
,
R. B.
,
Fukai
,
N.
,
Rawadi
,
G.
,
Roman-Roman
,
S.
,
Reginato
,
A. M.
,
Wang
,
H.
,
Cundy
,
T.
,
Glorieux
,
F. H.
,
Lev
,
D.
et al. 
(
2001
).
LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development
.
Cell
107
,
513
-
523
.
Guntur
,
A. R.
,
Le
,
P. T.
,
Farber
,
C. R.
and
Rosen
,
C. J.
(
2014
).
Bioenergetics during calvarial osteoblast differentiation reflect strain differences in bone mass
.
Endocrinology
155
,
1589
-
1595
.
Han
,
J.
,
Back
,
S. H.
,
Hur
,
J.
,
Lin
,
Y.-H.
,
Gildersleeve
,
R.
,
Shan
,
J.
,
Yuan
,
C. L.
,
Krokowski
,
D.
,
Wang
,
S.
,
Hatzoglou
,
M.
et al. 
(
2013
).
ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death
.
Nat. Cell Biol.
15
,
481
-
490
.
Hill
,
T. P.
,
Später
,
D.
,
Taketo
,
M. M.
,
Birchmeier
,
W.
and
Hartmann
,
C.
(
2005
).
Canonical Wnt/β-catenin signaling prevents osteoblasts from differentiating into chondrocytes
.
Dev. Cell
8
,
727
-
738
.
Hosios
,
A. M.
,
Hecht
,
V. C.
,
Danai
,
L. V.
,
Johnson
,
M. O.
,
Rathmell
,
J. C.
,
Steinhauser
,
M. L.
,
Manalis
,
S. R.
and
Vander Heiden
,
M. G.
(
2016
).
Amino acids rather than glucose account for the majority of cell mass in proliferating mammalian cells
.
Dev. Cell
36
,
540
-
549
.
Hu
,
H.
,
Hilton
,
M. J.
,
Tu
,
X.
,
Yu
,
K.
,
Ornitz
,
D. M.
and
Long
,
F.
(
2005
).
Sequential roles of Hedgehog and Wnt signaling in osteoblast development
.
Development
132
,
49
-
60
.
Hu
,
G.
,
Yu
,
Y.
,
Tang
,
Y. J.
,
Wu
,
C.
,
Long
,
F.
and
Karner
,
C. M.
(
2020
).
The amino acid sensor Eif2ak4/GCN2 is required for proliferation of osteoblast progenitors in mice
.
J. Bone Miner. Res.
35
,
2004
-
2014
.
Inoki
,
K.
,
Ouyang
,
H.
,
Zhu
,
T.
,
Lindvall
,
C.
,
Wang
,
Y.
,
Zhang
,
X.
,
Yang
,
Q.
,
Bennett
,
C.
,
Harada
,
Y.
,
Stankunas
,
K.
et al. 
(
2006
).
TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth
.
Cell
126
,
955
-
968
.
Jacob
,
R.
,
Rosenthal
,
N.
and
Barrett
,
E. J.
(
1986
).
Characterization of glutamine transport by liver plasma membrane vesicles
.
Am. J. Physiol.
251
,
E509
-
E514
.
Kang
,
S.
,
Bennett
,
C. N.
,
Gerin
,
I.
,
Rapp
,
L. A.
,
Hankenson
,
K. D.
and
Macdougald
,
O. A.
(
2007
).
Wnt signaling stimulates osteoblastogenesis of mesenchymal precursors by suppressing CCAAT/enhancer-binding protein α and peroxisome proliferator-activated receptor γ
.
J. Biol. Chem.
282
,
14515
-
14524
.
Karner
,
C. M.
and
Long
,
F.
(
2017
).
Wnt signaling and cellular metabolism in osteoblasts
.
Cell Mol. Life Sci.
74
,
1649
-
1657
.
Karner
,
C. M.
,
Esen
,
E.
,
Okunade
,
A. L.
,
Patterson
,
B. W.
and
Long
,
F.
(
2015
).
Increased glutamine catabolism mediates bone anabolism in response to WNT signaling
.
J. Clin. Investig.
125
,
551
-
562
.
Karner
,
C. M.
,
Esen
,
E.
,
Chen
,
J.
,
Hsu
,
F.-F.
,
Turk
,
J.
and
Long
,
F.
(
2016
).
Wnt protein signaling reduces nuclear Acetyl-CoA levels to suppress gene expression during osteoblast differentiation
.
J. Biol. Chem.
291
,
13028
-
13039
.
Karner
,
C. M.
,
Lee
,
S.-Y.
and
Long
,
F.
(
2017
).
Bmp induces osteoblast differentiation through both Smad4 and mTORC1 signaling
.
Mol. Cell Biol.
37
,
e00253
-
e00216
.
Katagiri
,
T.
,
Yamaguchi
,
A.
,
Komaki
,
M.
,
Abe
,
E.
,
Takahashi
,
N.
,
Ikeda
,
T.
,
Rosen
,
V.
,
Wozney
,
J. M.
,
Fujisawa-Sehara
,
A.
,
Suda
,
T.
et al. 
(
1994
).
Bone morphogenetic protein-2 converts the differentiation pathway of C2C12 myoblasts into the osteoblast lineage
.
J. Cell Biol.
127
,
1755
-
1766
.
Klein
,
P. S.
and
Melton
,
D. A.
(
1996
).
A molecular mechanism for the effect of lithium on development
.
Proc. Natl. Acad. Sci. USA
93
,
8455
-
8459
.
Lim
,
J.
,
Shi
,
Y.
,
Karner
,
C. M.
,
Lee
,
S.-Y.
,
Lee
,
W.-C.
,
He
,
G.
and
Long
,
F.
(
2016
).
Dual function of Bmpr1a signaling in restricting preosteoblast proliferation and stimulating osteoblast activity in mouse
.
Development
143
,
339
-
347
.
MacDonald
,
B. T.
and
He
,
X.
(
2012
).
Frizzled and LRP5/6 receptors for Wnt/beta-catenin signaling
.
Cold Spring Harb. Perspect. Biol.
4
,
a007880
.
Mackenzie
,
B.
,
Schäfer
,
M. K.-H.
,
Erickson
,
J. D.
,
Hediger
,
M. A.
,
Weihe
,
E.
and
Varoqui
,
H.
(
2003
).
Functional properties and cellular distribution of the system A glutamine transporter SNAT1 support specialized roles in central neurons
.
J. Biol. Chem.
278
,
23720
-
23730
.
Mody
,
N.
,
Parhami
,
F.
,
Sarafian
,
T. A.
and
Demer
,
L. L.
(
2001
).
Oxidative stress modulates osteoblastic differentiation of vascular and bone cells
.
Free Radic. Biol. Med.
31
,
509
-
519
.
Newsholme
,
P.
,
Procopio
,
J.
,
Lima
,
M. M. R.
,
Pithon-Curi
,
T. C.
and
Curi
,
R.
(
2003
).
Glutamine and glutamate—their central role in cell metabolism and function
.
Cell Biochem. Funct.
21
,
1
-
9
.
Otsuka
,
E.
,
Yamaguchi
,
A.
,
Hirose
,
S.
and
Hagiwara
,
H.
(
1999
).
Characterization of osteoblastic differentiation of stromal cell line ST2 that is induced by ascorbic acid
.
Am. J. Physiol.
277
,
C132
-
C138
.
Pfeiffer
,
R.
,
Rossier
,
G.
,
Spindler
,
B.
,
Meier
,
C.
,
Kühn
,
L.
and
Verrey
,
F.
(
1999
).
Amino acid transport of y+L-type by heterodimers of 4F2hc/CD98 and members of the glycoprotein-associated amino acid transporter family
.
EMBO J.
18
,
49
-
57
.
Pochini
,
L.
,
Scalise
,
M.
,
Galluccio
,
M.
and
Indiveri
,
C.
(
2014
).
Membrane transporters for the special amino acid glutamine: structure/function relationships and relevance to human health
.
Front. Chem.
2
,
61
.
Rached
,
M.-T.
,
Kode
,
A.
,
Xu
,
L.
,
Yoshikawa
,
Y.
,
Paik
,
J.-H.
,
Depinho
,
R. A.
and
Kousteni
,
S.
(
2010
).
FoxO1 is a positive regulator of bone formation by favoring protein synthesis and resistance to oxidative stress in osteoblasts
.
Cell Metab.
11
,
147
-
160
.
Riddle
,
R. C.
and
Clemens
,
T. L.
(
2017
).
Bone cell bioenergetics and skeletal energy homeostasis
.
Physiol. Rev.
97
,
667
-
698
.
Rodda
,
S. J.
and
McMahon
,
A. P.
(
2006
).
Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors
.
Development
133
,
3231
-
3244
.
Roose
,
J.
,
Huls
,
G.
,
van Beest
,
M.
,
Moerer
,
P.
,
van der Horn
,
K.
,
Goldschmeding
,
R.
,
Logtenberg
,
T.
and
Clevers
,
H.
(
1999
).
Synergy between tumor suppressor APC and the beta-catenin-Tcf4 target Tcf1
.
Science
285
,
1923
-
1926
.
Rungby
,
J.
,
Kassem
,
M.
,
Eriksen
,
E. F.
and
Danscher
,
G.
(
1993
).
The von Kossa reaction for calcium deposits: silver lactate staining increases sensitivity and reduces background
.
Histochem. J.
25
,
446
-
451
.
Tamarappoo
,
B. K.
,
Handlogten
,
M. E.
,
Laine
,
R. O.
,
Serrano
,
M. A.
,
Dugan
,
J.
and
Kilberg
,
M. S.
(
1992
).
Identification of the protein responsible for hepatic system N amino acid transport activity
.
J. Biol. Chem.
267
,
2370
-
2374
.
Tamarappoo
,
B. K.
,
Raizada
,
M. K.
and
Kilberg
,
M. S.
(
1997
).
Identification of a system N-like Na(+)-dependent glutamine transport activity in rat brain neurons
.
J. Neurochem.
68
,
954
-
960
.
Taylor
,
P. M.
,
Mackenzie
,
B.
,
Low
,
S. Y.
and
Rennie
,
M. J.
(
1992
).
Expression of rat liver glutamine transporters in Xenopus laevis oocytes
.
J. Biol. Chem.
267
,
3873
-
3877
.
Torrents
,
D.
,
Mykkänen
,
J.
,
Pineda
,
M.
,
Feliubadaló
,
L.
,
Estévez
,
R.
,
de Cid
,
R.
,
Sanjurjo
,
P.
,
Zorzano
,
A.
,
Nunes
,
V.
,
Huoponen
,
K.
et al. 
(
1999
).
Identification of SLC7A7, encoding y+LAT-1, as the lysinuric protein intolerance gene
.
Nat. Genet.
21
,
293
-
296
.
Tu
,
X.
,
Joeng
,
K. S.
,
Nakayama
,
K. I.
,
Nakayama
,
K.
,
Rajagopal
,
J.
,
Carroll
,
T. J.
,
McMahon
,
A. P.
and
Long
,
F.
(
2007
).
Noncanonical Wnt signaling through G protein-linked PKCδ activation promotes bone formation
.
Dev. Cell
12
,
113
-
127
.
Xian
,
L.
,
Wu
,
X.
,
Pang
,
L.
,
Lou
,
M.
,
Rosen
,
C. J.
,
Qiu
,
T.
,
Crane
,
J.
,
Frassica
,
F.
,
Zhang
,
L.
,
Rodriguez
,
J. P.
et al. 
(
2012
).
Matrix IGF-1 maintains bone mass by activation of mTOR in mesenchymal stem cells
.
Nat. Med.
18
,
1095
-
1101
.
Yu
,
Y.
,
Newman
,
H.
,
Shen
,
L.
,
Sharma
,
D.
,
Hu
,
G.
,
Mirando
,
A. J.
,
Zhang
,
H.
,
Knudsen
,
E.
,
Zhang
,
G.-F.
,
Hilton
,
M. J.
et al. 
(
2019
).
Glutamine metabolism regulates proliferation and lineage allocation in skeletal stem cells
.
Cell Metab.
29
,
966
-
978.e4
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information