Skip to main content
The Journal of Biological Chemistry logoLink to The Journal of Biological Chemistry
. 2020 May 28;295(34):12188–12202. doi: 10.1074/jbc.RA119.010710

Combined p53- and PTEN-deficiency activates expression of mesenchyme homeobox 1 (MEOX1) required for growth of triple-negative breast cancer

Mari Gasparyan 1, Miao-Chia Lo 1, Hui Jiang 3, Chang-Ching Lin 1, Duxin Sun 1,2,*
PMCID: PMC7443492  PMID: 32467227

Abstract

Triple-negative breast cancer (TNBC) is an aggressive cancer subtype for which effective therapies are unavailable. TNBC has a high frequency of tumor protein p53 (Tp53/p53)- and phosphatase and tensin homolog (PTEN) deficiencies, and combined p53- and PTEN-deficiency is associated with poor prognosis and poor response to anticancer therapies. In this study, we discovered that combined p53- and PTEN-deficiency in TNBC activates expression of the transcription factor mesenchyme homeobox 1 (MEOX1). We found that MEOX1 is expressed only in TNBC cells with frequent deficiencies in p53 and PTEN, and that its expression is undetectable in luminal A, luminal B, and HER2+ subtypes, as well as in normal breast cells with wild-type (WT) p53 and PTEN. Notably, siRNA knockdown of both p53 and PTEN activated MEOX1 expression in breast cancer cells, whereas individual knockdowns of either p53 or PTEN had only minimal effects on MEOX1 expression. MEOX1 knockdown abolished cell proliferation of p53- and PTEN-deficient TNBC in vitro and inhibited tumor growth in vivo, but had no effect on the proliferation of luminal and HER2+ cancer cells and normal breast cells. RNA-Seq and immunoblotting analyses showed that MEOX1 knockdown decreased expression of tyrosine kinase 2 (TYK2), signal transducer and activator of transcription 5B (STAT5B), and STAT6 in p53- and PTEN-deficient TNBC cells. These results reveal the effects of combined p53- and PTEN-deficiency on MEOX1 expression and TNBC cell proliferation, suggesting that MEOX1 may serve as a potential therapeutic target for managing p53- and PTEN-deficient TNBC.

Keywords: p53, phosphatase and tensin homolog (PTEN), cancer therapy, breast cancer, cancer biology, cancer therapeutics, cell proliferation, homeobox transcription, mesenchyme homeobox 1 (MEOX1), triple-negative breast cancer (TNBC)


Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that lacks effective therapy. Among four subtypes of breast cancers (luminal A, ER+/PR+/HER2−; luminal B, ER+/PR+/HER2 low; HER2+, ER−/PR−/HER2+; TNBC, ER−/PR−/HER2−), TNBC accounts for 10–20% of all breast cancers (15). TNBC has high rates of metastasis, and metastatic TNBC patients have poor 5-year survival (30%) (5, 6). Although tremendous advances have been made for therapies against the other three subtypes of breast cancers (714), TNBC still lacks effective treatment options.

TNBC has high frequencies of tumor protein p53 (Tp53/p53) and phosphatase and tensin homolog (PTEN) wild-type (WT) deficiency (9, 1523). To ascertain new therapeutic alternatives, researchers have set out to explore the complex heterogeneity of TNBC tumors and its evolving biology; in doing so studies demonstrate frequent genetic aberrations in tumor suppressor genes of p53 and PTEN as critical “driver mutations” conferring a selective advantage for TNBC aggressive tumorigenesis, drug resistance, and poor prognosis (9, 15, 1923). p53 deficiency is observed in ∼80% of TNBC tumors (15, 16). These include loss-/gain-of-function mutations (16, 24, 25) or loss of expression of p53 by epigenetic alteration of p53, which increases tumor formation and metastasis (2629). PTEN deficiency is observed in 35% of TNBC tumors (15). These include a small percentage of PTEN mutations (2–9%) and the majority of cases with reduced or loss of expression (3032), which is due to loss of heterozygosity (∼40%) and aberrant promoter epigenetic hyper-methylation (∼30-50%) (30, 33, 34). Heterozygous PTEN+/− mice (61–83%) develop basal-like breast cancer (3537).

Combined p53- and PTEN-deficiency is seen in 20–30% of TNBC, which is associated with poor prognosis and survival (19, 23). Analysis of human TNBC tumors show patients with low p53 and PTEN expressions display poorer prognosis with worse metastatic free survival relative to patients with normal p53 and PTEN levels (19). In addition, p53 and PTEN deletion in normal mammary epithelia of mice induces the formation of TNBC tumors (19, 23). These tumors manifest fast and aggressive TNBC hallmark characteristics with increased metastasis (19, 23). Furthermore, mice with loss of both p53 and PTEN in normal mammary epithelia exhibit decreased tumor free survival relative to loss of either p53 or PTEN alone (19, 23). In normal breast cells (MCF-10A), combined p53 and PTEN knockdown transforms these normal breast cells to aggressive metastatic cancer cells in vitro and in mice (38).

Combined p53- and PTEN-deficiency in TNBC is associated with poor responses to various therapies. Chemotherapy is the first line of treatment for TNBC. However, molecular profiles of TNBC residual disease unresponsive to neoadjuvant chemotherapy show high frequencies of genetic aberrations of both p53 and PTEN (9). Although PTEN-deficiency activates the PI3K/AKT/mTOR signaling pathway that can be inhibited by respective inhibitors to show promising efficacy in some breast cancers (39, 40), clinical evaluation of the pathway inhibitors (such as Everolimus and Ipatasertib) achieves limited success in TNBC patients (4143). Combined p53- and PTEN-deficiency in TNBC may result in a unique molecular signaling pathway, which may be distinct from the deficiency of p53 or PTEN alone as well as p53 and PTEN WT.

In this paper, we intended to identify molecular targets in TNBC with combined p53- and PTEN-deficiency, as well as offer insight into its poorly understood molecular biology. Our data show that combined p53- and PTEN-deficiency in TNBC activates a specific molecular target mesenchyme homeobox 1 (MEOX1), which may serve as a therapeutic target for p53- and PTEN-deficient TNBC. MEOX1 expression was surveyed in four subtypes of breast cancer cell lines, which show it is only expressed in TNBC cells with frequent p53- and PTEN-deficiencies, but undetectable in luminal A, luminal B, and HER2+ breast cancer subtypes, as well as in normal breast cells with WT p53 and PTEN. Single and double siRNA knockdowns of p53 and PTEN were used to investigate how they regulate MEOX1 expression in breast cancer cells. Furthermore, we studied the function of MEOX1 in p53- and PTEN-deficient TNBC in regulating cell proliferation and invasion in vitro and tumor growth in vivo. Finally, we also explored the downstream targets of MEOX1 in TNBC cells. These data suggest that MEOX1 may serve as a potential therapeutic target for p53- and PTEN-deficient TNBC.

Results

TNBC p53- and PTEN-deficiency is associated with MEOX1 expression

We examined MEOX1 expression by RT-qPCR and found that MEOX1 is only expressed in TNBC cells with frequent p53- and PTEN-deficiencies, but undetectable in normal breast cells of MCF-10A and three other breast cancer subtypes (Fig. 1A). Data shows TNBC cell lines of MDA-MB-231, BT-549, SUM149, MDA-MB-468, SUM159, and MDA-MB-453 have high expression of MEOX1; as expected, these in vitro TNBC cell lines show high incidences of p53 and PTEN genetic aberrations conferring loss of WT function for both tumor-suppressor genes (Table 1) (36, 4452). In contrast, results show no expression of MEOX1 in the luminal subtypes of MCF-7, T-47D, and ZR-75-1. Similarly, no expression of MEOX1 is seen in the HER2+ subtypes of BT-474, MDA-MB-361, and ZR-75-30.

Figure 1.

Figure 1.

MEOX1 is up-regulated in TNBC; combined loss of both p53 and PTEN significantly increase expression of MEOX1. A, MEOX1 expression is up-regulated in TNBC cell lines of MDA-MB-231, BT-549, SUM149, MDA-MB-468, SUM159, and MDA-MB-453. No expression of MEOX1 is seen in luminal cell lines of MCF-7, T-47D, and ZR-75-1. No expression of MEOX1 is also seen in HER2+ cell lines of BT-474, MDA-MB-361, and ZR-75-30. B-D, siRNA knockdown experiments with p53 and PTEN in normal immortalized MCF-10A, luminal MCF-7, and triple-negative SUM159 show p53 and PTEN negatively regulate MEOX1 expression. The most significant increase in MEOX1 expression is seen with dual knockdown of both tumor-suppressor genes, compared with individual knockdown of either tumor-suppressor gene alone. Results are shown as mean ± S.D., n = 3, one-way ANOVA statistical analysis with Dunnett's multiple comparisons test, **, p ≤ 0.01; ****, p ≤ 0.0001.

Table 1.

Summary of p53 and PTEN genetic aberrations in different subtypes of in vitro breast cancer cell lines

Breast cancer subtype Cell line TP53 mutation status TP53 expression PTEN mutation status PTEN expression
Normal Immortalized MCF-10A WT WT expression WT WT expression
Luminal MCF-7 WT WT expression WT WT expression
T-47D Homozygous c.580C > T, p.L194F Mutant expression WT WT expression
ZR-75-1 WT WT expression Homozygous c.323T>G, p.L108R Mutant expression
HER2-positive BT-474 Homozygous c.853G>A, p.E285K Mutant expression WT WT expression
MDA-MB-361 WT/homozygous c.166G>T, p.E56 ×? WT/mutant expression? WT WT expression
ZR-75-30 WT WT expression WT WT expression
Triple negative MDA-MB-231 Homozygous c.839G>A, p.R280K Mutant expression WT WT expression
BT-549 Homozygous c.747G>C, p.R249S Mutant expression Homozygous c.823delG, p.V275fsX1 Null
SUM149 Homozygous c.711G>A, p.M237I Mutant expression WT/mutation? WT/null?
MDA-MB-468 Homozygous c.818G>A, p.R273H Mutant expression Homozygous c.253 + 1G>T, p.A72fsX5 Null
SUM159 Homozygous c.472_473ins3, p.S158insS Mutant expression WT WT expression
MDA-MB-453 Homozygous c.991_1182del192, p.0 Null Heterozygous c.919G>A, p.E307K WT and mutant expression

Combined p53- and PTEN-deficiency activates MEOX1 expression compared with WT and single deficiency of p53 or PTEN

To confirm if p53 and PTEN regulates MEOX1 expression, we used siRNA to knockdown p53 and/or PTEN. The knockdown efficiency of p53 and PTEN was confirmed using RT-qPCR (Fig. S1). The data show that siRNA knockdown of both p53 and PTEN in normal immortalized MCF-10A, luminal MCF-7, and triple-negative SUM159 increases MEOX1 expression (Fig. 1, B–D), whereas siRNA knockdown of either p53 or PTEN alone only have minor effects on MEOX1 expression in all three different breast cancer subtypes.

It is important to note that normal immortalized MCF-10A and luminal MCF-7 cells are used because both have normal WT function and expression of p53 and PTEN (Table 1) (36, 4452). However, triple-negative SUM159 cells harbor loss of WT function for p53 with normal function of PTEN (Table 1) (36, 4452). Finding a triple-negative cell line with no genetic aberrations in p53 is challenging, as such SUM159 cells are just used as proof of concept. Nevertheless, data clearly shows the expression of MEOX1 is specific to TNBC and negatively regulated by both tumor suppressor genes of p53 and PTEN.

Interestingly, HER2 also negatively regulates MEOX1 expression. In luminal MCF-7, stable knockdown of PTEN alone increased MEOX1 expression, however, stable overexpression of HER2 down-regulated its expression (Fig. S2A). Furthermore, HER2 expression inhibited MEOX1 expression when PTEN was knocked down in MCF-7 cells. Similar results were observed in triple-negative SUM159 cells. Stable knockdown of PTEN alone in SUM159 increased the expression of MEOX1, but stable overexpression of HER2 down-regulated its expression (Fig. S2B). As seen with MCF-7, overexpression of HER2 inhibited MEOX1 expression when PTEN was concurrently knocked down in SUM159. This point was further established when performing HER2, p53, and PTEN siRNA experiments in the HER2+ breast cancer cell line BT-474. Knockdown of p53 or PTEN alone had no effect on MEOX1 expression in BT-474, and neither did dual knockdown of both p53 and PTEN, where HER2 levels are high in BT-474 (Fig. S2C). Only during dual knockdown of p53 with HER2 or PTEN with HER2 did expression of MEOX1 start to increase. However, the most significant increase of MEOX1 expression was seen when all three genes of HER2, p53, and PTEN were down-regulated.

MEOX1 knockdown decreases in vitro cell proliferation of p53- and PTEN-deficient TNBC

To assess MEOX1's function on cell proliferation, we used three different siRNA treatments to knockdown MEOX1 in p53- and PTEN-deficient TNBC (claudin-low BT-549 and basal-like MDA-MB-468). These two different intrinsic subtypes of TNBC harbor inherent genetic aberrations in p53 and PTEN conferring loss of WT function for both tumor-suppressor genes (Table 1) (36, 4452). The siRNA knockdown efficiency of MEOX1 was confirmed using RT-qPCR (Fig. S3). The data show that knocking down MEOX1 using two single siRNAs and a pooled mixure of four different siRNAs significantly decrease cell proliferation of these two TNBC cell lines (Fig. 2, A and B). However, two single siRNAs and a pooled mixure of four different siRNAs show no significant change in cell proliferation in normal immortalized MCF-10A, luminal ZR-75-1, and HER2+ BT-474 (Fig. 2, C–E). These data suggest that MEOX1 expression may be critical and specific to regulating cell proliferation in p53- and PTEN-deficient TNBC. In addition, the siRNA experiments in MCF-10A, ZR-75-1 and BT-474 also suggest that these siRNAs of MEOX1 have no off-target effects on these cell lines, which have undetectable levels of MEOX1. These data may also suggest these cells do not rely on MEOX1 for their proliferation.

Figure 2.

Figure 2.

MEOX1 knockdown specifically decreases in vitro cell proliferation of p53- and PTEN-deficient TNBC. A and B, using three different siRNA treatments, MEOX1 knockdown in triple-negative cells (claudin-low BT-549 and basal-like MDA-MB-468) significantly decreases cell proliferation of these p53- and PTEN-deficient TNBCs. C-E, using three different siRNA treatments, MEOX1 knockdown in normal immortalized MCF-10A, luminal ZR-75-1, and HER2+ BT-474 shows no significant change in cell proliferation. Results are shown as mean ± S.D., n = 6, two-way ANOVA statistical analysis with Dunnett's multiple comparisons test, ****, p ≤ 0.0001.

Knockdown of MEOX1 decreases in vitro cell self-renewal of p53- and PTEN-deficient TNBC

To evaluate if MEOX1 also regulates cell self-renewal of p53- and PTEN-deficient TNBC, we used 3D mammosphere formation efficiency for consecutive passages in vitro with three different MEOX1 siRNA knockdown treatments previously described (53). Mammosphere formation assays show knocking down MEOX1 in p53- and PTEN-deficient TNBC (claudin-low BT-549 and basal-like MDA-MB-468) decreases both primary and secondary consecutive mammosphere formations (Fig. 3, A–D and F). As such, MEOX1 knockdown decreases cell self-renewal function of p53- and PTEN-deficient TNBC. In contrast, stable overexpression of MEOX1 in normal immortalized MCF-10A increases mammosphere formation, regulating an increase in cell self-renewal (Fig. 3, E and G). The overexpression efficieny of MEOX1 in MCF-10A was confirmed using RT-qPCR (Fig. S4).

Figure 3.

Figure 3.

Knockdown of MEOX1 decreases in vitro cell self-renewal of p53- and PTEN-deficient TNBC. A–D and F, using different siRNA treatments, a decrease in MEOX1 expression decreases primary and secondary consecutive mammosphere formations in triple-negative cells (claudin-low BT-549 and basal-like MDA-MB-468). As such, knockdown of MEOX1 decreases self-renewal properties of p53- and PTEN-deficient TNBCs. E and G, normal immortalized MCF-10A cells with stable overexpression of MEOX1 increases mammosphere formation, regulating an increase in cell self-renewal. Results are shown as mean ± S.D., one-way ANOVA statistical analysis with Dunnett's multiple comparisons test was used for A-D, unpaired t test statistical analysis for E, **, p ≤ 0.01; ***, p ≤ 0.001; ****, p ≤ 0.0001.

MEOX1 knockdown decreases in vitro cell migration and invasion of p53- and PTEN-deficient TNBC

Cell directional migration and invasion through an extracellular matrix are important properties of metastasis (5456). In vitro cell migration and invasion assays were conducted to ascertain the role of MEOX1 as a metastatic regulator of p53- and PTEN-deficient TNBC. Knocking down MEOX1 in both claudin-low BT-549 and basal-like MDA-MB-468 decreases both migration and invasion of TNBC cells (Fig. 4, A, C, D, and F). Furthermore, the invasion index decreases by ∼50% in claudin-low BT-549 and ∼30% in basal-like MDA-MB-468 cells (Fig. 4, B and E).

Figure 4.

Figure 4.

MEOX1 knockdown decreases in vitro cell migration and invasion of p53- and PTEN-deficient TNBC. A, C, D, and F, following MEOX1 siRNA treatments, cell migration and invasion are decreased in triple-negative cells (claudin-low BT-549 and basal-like MDA-MB-468). B and E, invasion index decreases by ∼50% in claudin-low BT-549 and ∼30% in basal-like MDA-MB-468 cells after MEOX1 siRNA treatments. The cells were transfected with siRNA of MEOX1 for 72 h, then cells were collected and only live viable cells were plated to test for migration and invasion in 24 h. Results are shown as mean ± S.D., n = 2, two-way ANOVA statistical analysis with Dunnett's multiple comparisons test, *, p ≤ 0.05; **, p ≤ 0.01; ***, p ≤ 0.001; ****, p ≤ 0.0001.

It is worth noting that after 72 h of siRNA transfection, cells were trypsinized and counted three times to make certain only live viable cells were plated for migration and Matrigel invasion assays. In addition, these live cells plated were serum starved to establish cell synchronization, minimizing any potential differences in cell migration and invasion that may be seen due to varying cell proliferation rates and serum components. Furthermore, we also calculated the invasion index, which normalized the ratio of the cells invading through Matrigel insert to cells migrating through control insert (% Invasion = mean # of cells invading through Matrigel insert/mean # of cells migrating through control insert; Invasion Index = % invasion in MEOX1 siRNA treatment/% invasion in control siRNA treatment). These results indicate MEOX1 regulates metastatic properties of cell migration and invasion in p53- and PTEN-deficient TNBC.

Inducible knockdown of MEOX1 inhibits in vivo tumor growth of p53- and PTEN-deficient TNBC

To evaluate MEOX1 function on tumor growth in vivo, we used a doxycycline-inducible MEOX1 shRNA knockdown system in basal-like TNBC cells MDA-MB-468 with p53- and PTEN-deficiency. The inducible knockdown efficiency of MEOX1 by doxycycline was confirmed using RT-qPCR (Fig. S5). Using NOD/SCID mice, we used two treatment regimens (adjuvant and advanced treatment) to test MEOX1 function on xenograft tumor growth in orthotopic mammary fat pads. In an adjuvant regimen, MEOX1 shRNA knockdown was induced with administration of doxycycline 3 days following surgery. In an advanced treatment regimen setting, MEOX1 shRNA knockdown was induced with administration of doxycycline after palpable or visible tumor growth. Data shows that regardless of when doxycycline was administered to induce shRNA, in an adjuvant or advanced treatment regimen, MEOX1 knockdown decreases in vivo tumor growth of p53- and PTEN-deficient TNBC (Fig. 5, A and B). However, knockdown of MEOX1 seems to have a greater impact on decreasing tumor growth in an adjuvant setting.

Figure 5.

Figure 5.

Inducible knockdown of MEOX1 inhibits in vivo tumor growth of p53- and PTEN-deficient TNBC. A, in an adjuvant setting, MEOX1 shRNA knockdown was induced with doxycycline 3 days after orthotopic mammary fat pad injections. B, in an advanced setting, MEOX1 shRNA knockdown was induced with doxycycline after palpable or visible tumor growth following orthotopic mammary fat pad injections. In both settings, knocking down MEOX1 decreases growth of basal-like MDA-MB-468 p53- and PTEN-deficient TNBC xenograft tumors in NOD/SCID mice. However, a greater impact on decreasing tumor growth with MEOX1 knockdown is seen in the adjuvant setting. Results are shown as mean ± S.D., n = 5, two-way ANOVA statistical analysis with Dunnett's multiple comparisons test, ****, p ≤ 0.0001.

MEOX1 regulates JAK/STAT signaling in p53- and PTEN-deficient TNBC

We used RNA-seq to ascertain mechanisms involved in MEOX1 functional regulation and potential downstream targets of p53- and PTEN-deficient TNBC. IPA was used to assess how RNA-seq differential gene expression between MEOX1 knockdown versus negative control effects biological signaling pathways in p53- and PTEN-deficient TNBCs.

RNA-seq IPA comparison and individual analyses of p53- and PTEN-deficient claudin-low BT-549 and basal-like MDA-MB-468 show important biological signaling pathways are perturbed following MEOX1 knockdown (Fig. 6, A–C). This includes well-known JAK/STATs, MAPK, p70S6K, CXCR4, actin cytoskeleton signaling, integrin signaling, Gαq, RhoA, and Rho family of GTPases. These pathways offer insight to potential mechanistic transductions involved in MEOX1 regulation of proliferation and metastasis in these p53- and PTEN-deficient TNBC cells.

Figure 6.

Figure 6.

MEOX1 knockdown perturbs important canonical biological signaling pathways in p53- and PTEN-deficient TNBC. A, IPA was utilized to assess and compare biological signaling pathways significantly altered in p53- and PTEN-deficient TNBCs of claudin-low BT-549 and basal-like MDA-MB-468 upon MEOX1 knockdown. B, individual IPA assessment of MEOX1 knockdown in p53- and PTEN-deficient claudin-low BT-549 shows activation and inactivation of canonical signaling pathways. C, individual IPA assessment of MEOX1 knockdown in p53- and PTEN-deficient basal-like MDA-MB-468 shows inactivation of canonical signaling pathways. JAK/STAT signaling pathways are inactivated in both TNBC cell lines.

In a comparison analysis or individual analysis of p53- and PTEN-deficient TNBCs signaling of the JAK/STAT pathway as a whole is disrupted (Fig. S6). Although inactivation of JAK/STAT signaling has a significantly high p value of 0.0000089 in BT-549 and a close to significant p value of 0.060 in MDA-MB-468, inactivation of the STAT3 signaling pathway has significant p values < 0.05 and z-scores < −2 in both cell lines. (Table S1). To confirm these findings, we performed Western blotting analysis for JAK/STAT pathways in MEOX1 siRNA knockdown cells in two TNBC cell lines. The data show that knockdown of MEOX1 decreases JAK1 and TYK2 protein levels in claudin-low BT-549, but only decreases TYK2 protein levels in basal-like MDA-MB-468 (Fig. 7, A and B). Analyzing the STAT family of proteins shows an increase in STAT1 as well as decreases in STAT5B and STAT6 protein levels in claudin-low BT-549 (Fig. 7A). Similarly, decreases in STAT5B and STAT6 protein levels are also observed in basal-like MDA-MB-468 (Fig. 7B). Further analysis of phosphorylated STATs shows a decrease of P-STAT3 (Tyr-705) in claudin-low BT-549, however, no change in phosphorylated STATs are seen in basal-like MDA-MB-468 cells (Fig. 7, A and B). In analyzing inhibitory JAK/STAT signaling pathways, which were reported to be associated with p53 and PTEN (38), no significant changes by siRNA knockdown of MEOX1 were found in PIAS1, PIAS3, PIAS4, SOCS2, and SOCS3 (Fig. S7).

Figure 7.

Figure 7.

MEOX1 regulates JAK/STAT signaling in p53- and PTEN-deficient TNBC. A, knockdown of MEOX1 in p53- and PTEN-deficient TNBC of claudin-low BT-549 decreases JAK1, TYK2, STAT5B, STAT6, and P-STAT3 (Tyr-705) protein levels; however, an increase in STAT1 protein levels are observed. B, knockdown of MEOX1 in p53- and PTEN-deficient TNBC of basal-like MDA-MB-468 decreases TYK2, STAT5B, and STAT6 protein levels.

In summary, Western blotting analysis for the JAK/STAT pathway shows that MEOX1 knockdown has a significant impact on decreasing JAK1, TYK2, STAT5B, STAT6, and P-STAT3 (Tyr-705) protein levels in claudin-low BT-549, but only has a significant impact on decreasing TYK2, STAT5B, and STAT6 protein levels in basal-like MDA-MB-468. Interestingly, STAT1 protein levels increase in claudin-low BT-549 after MEOX1 knockdown.

Discussion

In this article, insight is offered into the complex and poorly understood molecular biology of p53- and PTEN-deficient TNBC by studying the functional and mechanistic role of MEOX1, presenting a specific therapeutic target. A homeobox transcription factor, MEOX1 is well known for its role in somitogenesis of the developing vertebrate embryo (5761). Although mainly expressed and studied in the context of development, recent research demonstrates a link between MEOX1 and its activation in cancer. Thiaville and colleagues (62) show evidence involving MEOX1 in ovarian cancer, describing MEOX1 as an important pre-B-cell leukemia homeobox-1 (PBX1) cofactor and target gene for tumor cell growth. Additional research links the function of MEOX1 to increased breast cancer cell proliferation and correlates nuclear expression of MEOX1 with poor overall breast cancer patient survival, as well as increased lymph node metastasis and high tumor stage (43).

In investigating the role of homeobox transcription factor MEOX1 in breast cancer, RNA expression analysis of in vitro breast cancer cell lines show MEOX1 is up-regulated in TNBC and no expression of MEOX1 is seen in luminal or HER2+ subtypes. Furthermore, RNA expression analysis also show that MEOX1 is negatively regulated by both tumor-suppressor genes of p53 and PTEN. In vitro p53 and PTEN siRNA knockdown experiments demonstrate that only with combined loss of p53 and PTEN does expression of MEOX1 significantly increase. High expression of MEOX1 in p53- and PTEN-deficient TNBC has a crucial role for regulating cell proliferation and cell self-renewal. Experiments with siRNA knockdown show a decrease in MEOX1 expression significantly decreases in vitro cell proliferation and cell self-renewal of two different intrinsic subtypes of claudin-low and basal-like p53- and PTEN-deficient TNBC. Additionally, no effect on cell proliferation is seen in normal immortalized, luminal, or HER2+ breast cancer cells upon knockdown of the transcription factor; these data suggest that targeting MEOX1 can specifically decrease the aggressive proliferative behavior of p53- and PTEN-deficient TNBC with minimal potential of off-target effects. In vivo experiments corroborate in vitro results, where tumor xenograft mouse models using doxycycline-inducible shRNA show knockdown of MEOX1 decreases tumor growth of basal-like p53- and PTEN-deficient TNBC in an adjuvant and advanced treatment regimen. However, a greater inhibition of tumor growth is seen in an adjuvant regimen. As such, MEOX1 targets particularly in an adjuvant setting can benefit to decrease the rapid proliferative property of TNBC tumors with p53- and PTEN-deficiency. Moreover, results also show that MEOX1 regulates metastasis; knockdown experiments using siRNA for MEOX1 decreases metastatic functions of migration and invasion in both claudin-low and basal-like intrinsic subtypes. Thus, along with decreasing rapid cell proliferation, targeting MEOX1 can also have a crucial role in reducing the aggressive metastatic potential of p53- and PTEN-deficient TNBC.

To ascertain the mechanistic biological signaling pathways involved in MEOX1 function of regulating cell proliferation and metastasis in p53- and PTEN-deficient TNBC, RNA-seq was utilized. RNA-seq mechanistic investigation of IPA comparison and individual analyses of both claudin-low and basal-like p53- and PTEN-deficient TNBC shows JAK/STAT signaling pathways are inactivated upon MEOX1 knockdown. Indeed, investigation of the JAK/STAT pathway using Western blotting analysis show MEOX1 knockdown increases STAT1 but decreases JAK1, TYK2, STAT5B, STAT6, and P-STAT3 (Tyr-705) protein levels in claudin-low, as well as decreases TYK2, STAT5B, and STAT6 protein levels in basal-like p53- and PTEN-deficient TNBC.

Although still undergoing extensive research and scrutiny, the JAK family proteins of JAK1 and TYK2, as well as STAT family proteins of STAT1, STAT3, STAT5, and STAT6 have been implicated in multiple cancers, including breast tumorigenic pathways of proliferation and metastasis (6370). Research shows aberrations in JAK/STAT signaling promote oncogenic properties of proliferation and metastasis, such as increased cell growth, migration, and invasion (6365). In addition, research has also previously shown a link between aberrations in JAK/STAT signaling with loss of p53 and PTEN WT function in TNBC cells (38).

Kim and colleagues (38) demonstrate in vitro stable knockdown of p53 and PTEN in normal immortalized MCF-10A transforms cells to resemble basal-like and claudin-low TNBCs. These cells display increased activation of the IL6-STAT3-NFκB signaling pathway through proteolytic degradation of SOCS3, as well as manifest aggressive proliferative metastatic tumors following xenograft orthotropic mammary fat pad injections in NOD/SCID mice (38). However, in this paper, siRNA knockdown of MEOX1 in p53- and PTEN-deficient TNBC did not alter SOCS3 protein levels significantly. Although these results offer some potential mechanistic insight for MEOX1 regulation of p53- and PTEN-deficient TNBC through JAK/STAT signaling, more research is required to assess how exactly the interplay of individual JAK/STAT proteins affect functions of proliferation and metastasis, especially in different intrinsic subtypes of claudin-low and basal-like.

It is worth noting that the expression of MEOX1 was detected by its RNA levels using only RT-qPCR in this study. No antibody for MEOX1 was found to be sensitive and specific enough to confirm its protein levels in various cells and confirm siRNA or shRNA knockdown. In addition, while the siRNAs and shRNAs showed similar knockdown efficiency for MEOX1 RNA, it is not known if these knockdown treatments will have similar efficiency in down-regulating MEOX1 protein levels because knockdown of RNA and protein are not always consistent (71). These discrepancies need to be further investigated in the future.

Establishing effective therapeutic targets will require analyzing the interplay of signaling pathways transformed by multiple combined genetic aberrations, which ultimately govern TNBC tumorigenesis and heterogeneity. Studying MEOX1 offers biological insight and presents an opportunity for specified targeting of combined p53- and PTEN-deficient TNBC, which lacks actionable targets. Given the functional role of MEOX1 to decrease tumorigenic properties of proliferation and metastasis, targeting MEOX1 can help decrease the aggressive proliferative and metastatic potential of p53- and PTEN-deficient TNBC. Since genetic aberrations in tumor-suppressor genes of p53 and PTEN are frequent common driver mutations for tumorigenesis in TNBC, targeting MEOX1 may provide a potential therapeutic strategy. Given that MEOX1 is expressed during development and not known to be present in adult tissues, therapies utilizing the latest treatment modalities in the clinic to target MEOX1 are suspected to be specific in p53- and PTEN-deficient TNBC.

Materials and Methods

In vitro cell culture growth conditions

Gibco Life Technologies base media was used to grow in vitro breast cancer cell lines and ATCC recommended media formulation guidelines were followed. MCF-10A cells were grown in DMEM/F-12 supplemented with antibiotic-antimycotic, 5% horse serum, 20 ng/ml of epidermal growth factor (EGF), 100 ng/ml of cholera toxin, 500 ng/ml of hydrocortisone, and 10 µg/ml of insulin. MCF-7 cells were grown in Earle's minimal essential medium supplemented with antibiotic-antimycotic, 10% fetal bovine serum, 10 µg/ml of insulin, and 1 mm sodium pyruvate. T-47D cells were grown in RPMI supplemented with antibiotic-antimycotic, 10% fetal bovine serum, 5 µg/ml of insulin, 1 mm sodium pyruvate, and 10 mm HEPES. ZR-75-1, BT-474, and ZR-75-30 cells were grown in RPMI supplemented with antibiotic-antimycotic, 10% fetal bovine serum, 1 mm sodium pyruvate, and 10 mm HEPES. MDA-MB-231, MDA-MB-453, and MDA-MB-468 cells were grown in DMEM supplemented with antibiotic-antimycotic, 10% fetal bovine serum, and 1 mm sodium pyruvate. MDA-MB-361 cells were grown in DMEM supplemented with antibiotic-antimycotic, 20% fetal bovine serum, and 1 mm sodium pyruvate. BT-549 cells were grown in RPMI supplemented with antibiotic-antimycotic, 10% fetal bovine serum, 0.85 µg/ml of insulin, 1 mm sodium pyruvate, and 10 mm HEPES. SUM149 and SUM159 cells were grown in Ham's F-12 supplemented with antibiotic-antimycotic, 5% fetal bovine serum, 1 µg/ml of hydrocortisone, 5 µg/ml of insulin, and 10 mm HEPES. All cells were grown in a humidified 37 °C incubator with 5% CO2.

In vitro cell culture transient transfection for siRNA knockdown

Each breast cancer cell line was plated onto 6-well–plates at ∼50% confluence per well. For each well, 2 × 105 cells of BT-549, MDA-MB-468, SUM159, MCF-10A; 6 × 105 cells of ZR-75-1; and 9 × 105 cells of BT-474 were required to achieve 50% confluence. After 24 h of plating, cells were transfected with siRNA using Invitrogen Lipofectamine RNAiMAX Reagent (No. 13778-150); transfection was conducted according to the manufacturer's instructions and antibiotic-antimycotic was omitted from the media to increase transfection efficiency as well avoid unnecessary toxicity. For MEOX1 knockdown experiments, 50 nm Negative Control siRNA and 50 nm MEOX1 siRNA for each treatment group was utilized. For HER2, p53, and PTEN knockdown experiments, 30 nm Negative Control siRNA and 30 nm HER2, p53, and PTEN siRNA was utilized. Cells were incubated with transfection reagent and siRNA only 24 h to avoid toxicity, after which media was removed and fresh media without antibiotic-antimycotic was added. Following 48-72 h from the start of transfection, cells were trypsinized or harvested for RNA, protein, or for specified experimental analysis.

All siRNAs used to conduct experiments were purchased from Qiagen. Negative Control siRNA (No. 1027281) contained target sequence, 5′-CAGGGTATCGACGATTACAAA-3′; sense strand, 5′-GGGUAUCGACGAUUACAAAUU-3′; and antisense strand, 5′-UUUGUAAUCGUCGAUACCCUG-3′. Three different MEOX1 siRNA treatments were used to validate results. MEOX1 siRNA 1 (No. SI00630266) contained target sequence, 5′-CAGGCTTGACTGGGTGGACAA-3′; sense strand, 5′-GGCUUGACUGGGUGGACAATT-3′; and antisense strand, 5′-UUGUCCACCCAGUCAAGCCTG-3′. MEOX1 siRNA 2 (No. SI00630280) contained target sequence, 5′-AAGCTAATTGTGCGAGCTCAA-3′; sense strand, 5′-GCUAAUUGUGCGAGCUCAATT-3'; and antisense strand, 5′-UUGAGCUCGCACAAUUAGCTT-3′. MEOX1 siRNA Mixture is a pool of four different MEOX1 siRNAs mixed together for a final concentration of 50 nm, matching the concentration of MEOX1 siRNA 1 and MEOX1 siRNA 2. This is a technique commonly carried out using four different siRNAs each at low concentrations to ensure there are no off-target effects that may be caused when using one siRNA alone. The MEOX1 siRNA mixture pools together 12.5 nm of each MEOX1 siRNA 1, MEOX1 siRNA 2, and MEOX1 siRNA 3 (No. SI03145205) with target sequence, 5′-AGCTGGCGACTCGGAAAGTAA-3′; sense strand, 5′-CUGGCGACUCGGAAAGUAATT-3′; and antisense strand, 5′-UUACUUUCCGAGUCGCCAGCT-3′, as well as MEOX1 siRNA 4 (No. SI04293310) with target sequence, 5′-TCCACGATTTCTGGATTGAAA-3′; sense strand, 5′-CACGAUUUCUGGAUUGAAATT-3′; and antisense strand, 5′-UUUCAAUCCAGAAAUCGUGGA-3′.

To knockdown HER2, Qiagen FlexiTube GeneSolution (No. GS2064) was tested. Functionally verified HER2 siRNA (No. SI02223571) validated by Qiagen to knockdown HER2 contained target sequence, 5′-AACAAAGAAATCTTAGACGAA-3′; sense strand, 5′-CAAAGAAAUCUUAGACGAATT-3′; and antisense strand, 5′-UUCGUCUAAGAUUUCUUUGTT-3′. To knockdown p53, Qiagen FlexiTube GeneSolution (No. GS7157) was tested. Functionally verified p53 siRNA (No. SI02655170) validated by Qiagen to knockdown p53 contained target sequence, 5′-AAGGAAATTTGCGTGTGGAGT-3′; sense strand, 5′-GGAAAUUUGCGUGUGGAGUTT-3′; and antisense strand, 5′-ACUCCACACGCAAAUUUCCTT-3′. To knockdown PTEN, Qiagen FlexiTube GeneSolution (No. GS5728) was tested. Functionally verified PTEN siRNA (No. SI00301504) validated by Qiagen to knockdown PTEN contained target sequence, 5′-AAGGCGTATACAGGAACAATA-3′; sense strand, 5′-GGCGUAUACAGGAACAAUATT-3′; and antisense strand, 5′-UAUUGUUCCUGUAUACGCCTT-3′.

In vitro cell culture stable transduction for lentiviral overexpression

Normal immortalized MCF-10A cells were used to overexpress MEOX1 with lentiviral vectors. MCF-10A cells were virally transduced with Abmgood pLenti-CMV-RFP-2A-Puro-Blank Control (No. LV591) and Abmgood pLenti-GIII-CMV-hMEOX1-RFP-2A-Puro (No. LV217710) lentiviral vectors. Viral particles were generated at the University of Michigan Vector Core. Cells were infected with lentiviral particles for 24 h using 8 μg/ml of Polybrene. Following 72 h from the start of infection, selection for infected cells was conducted using FACS of RFP-positive MCF-10A pLenti-CMV-RFP-2A-Puro-Blank Control and RFP-positive MCF-10A pLenti-GIII-CMV-hMEOX1-RFP-2A-Puro–transduced cells. MCF-10A cells with pLenti-CMV-RFP-2A-Puro-Blank Control and pLenti-GIII-CMV-hMEOX1-RFP-2A-Puro are hereafter labeled as pLenti control and pLenti MEOX1 overexpression, respectively.

Two-Step reverse transcription-quantitative PCR (RT-qPCR)

RNA was isolated from cells using either Life Technologies TRIzol reagent (No. 15596018) or Qiagen RNeasy Kit (No. 74104). RNA isolation was conducted according to the manufacturer's instructions. Invitrogen SuperScript III First-Strand Synthesis SuperMix (No. 11752-050) was used to make cDNA from isolated RNA. The maximum amount of RNA allotted by the kit was utilized, 1 μg, to make cDNA. Afterward, cDNA was diluted no more than 5-fold to perform qPCR. Applied Biosystems SYBR Green PCR Master Mix (No. 4309155) was used to perform qPCR. All primers were obtained from Integrated DNA Technologies (IDT) and used at a final concentration of 500 nm. The ACTB primer (No. Hs.PT.39a.22214847) was used with forward sequence of 5′-CCTTGCACATGCCGGAG-3′ and reverse sequence of 5′-ACAGAGCCTCGCCTTTG-3′. Although different MEOX1 primers were used to validate results, the main primer used for data obtained in this paper was MEOX1 primer (No. Hs.PT.58.26021003.g) with forward sequence of 5′-CTCAGTGAAGATGTGCTCCTC-3′ and reverse sequence of 5′-CAGACTTCCTGGCGACA-3′.

It is important to note that when performing RT-qPCR for MEOX1, extra care must be taken to handle the RNA samples, MEOX1 RNA expression in breast cancer cell lines is fairly low. MEOX1 was only detected at RNA levels using in vitro breast cancer cell lines; although many attempts were made, no successful antibody was found to detect MEOX1 expression at the protein level.

Cell proliferation assay

Following 48 h with transfection of 50 nm Negative Control siRNA and 50 nm MEOX1 siRNA treatments, cells were trypsinized and counted three times so as to ensure accurate plating of cell number. Using 96-well–plates, each cell line required plating a different number of cells per well to achieve normal exponential growth within 5 days of analysis. As such, for each well, 2 × 103 cells of BT-549, MDA-MB-468, MCF-10A; 6 × 103 cells of ZR-75-1; and 9 × 103 cells of BT-474 were plated. For each siRNA treatment, cells were plated into six different wells for statistical analysis. Invitrogen CyQUANT Cell Proliferation Assay (No. C35011) was conducted for a period of 5 days.

Mammosphere formation assay

Following 48 h with transfection of 50 nm Negative Control siRNA and 50 nm MEOX1 siRNA treatments, cells were trypsinized and counted three times. Cells were resuspended to a final concentration of 1 × 106 cells/ml and a syringe was utilized with a 23-gauge needle to gently aspirate and dissociate the cells three times to achieve a single cell suspension. Stem Cell Technologies MammoCult Medium (No. 05620) was utilized and supplemented with 4 μg/ml of heparin and 0.48 μg/ml of hydrocortisone. Ultra-Low attachment 6-well–plates were necessary for this experiment to ensure cells would not adhere to the bottom but grow suspended in media. To each well, 3 ml of complete MammoCult medium was added and 5 × 103 cells were plated; each treatment group was plated into at least three wells for statistical analysis. Primary mammospheres were allowed to grow undisturbed for 5 days, after which mammospheres >25 μm were counted under a microscope. After counting, mammospheres for each treatment group were collected and combined for secondary mammosphere experiments. Combined wells for each treatment group were trypsinized and counted three times. Cells were resuspended to a final concentration of 1 × 106 cells/ml, and again a 23-guage needle was utilized to gently aspirate and dissociate the cells three times to achieve a single cell suspension. Using Ultra-Low attachment 6-well–plates, 3 ml of complete MammoCult medium was added per well and to each well 5 × 103 cells were plated; each treatment group was plated into at least two wells for statistical analysis. All cells from the primary mammospheres were plated for secondary mammosphere formation. Secondary mammospheres were allowed to grow undisturbed for 5 days, after which mammospheres >25 μm were counted under a microscope. Mammosphere formation efficiency (%) was calculated as (number of mammospheres per well/number of cells plated per well) × 100 (53).

Cell migration and invasion assay

Following 72 h with transfection of 50 nm Negative Control siRNA and 50 nm MEOX1 siRNA treatments, Corning BioCoat Control Inserts (No. 354578) and Corning BioCoat Matrigel Invasion Chamber (No. 354480) 24-well–plate was utilized for cell migration and invasion assays. Protocol was conducted according to the manufacturer's instructions with one exception, an optimized seeding density of 7.5 × 104 cells per 24-well chamber were plated. For microscopy counting and analysis, cells were fixed and stained using Thermo Scientific Shandon Kwik-Diff Stain Kit (No. 9990701).

Cloning for short hairpin RNA (shRNA) stable cell lines

The Addgene Tet-pLKO-puro plasmid (No. 21915) was used to clone doxycycline-inducible MEOX1 shRNA, a gift from Dmitri Wiederschain (72). As such, protocols to clone and establish stable doxycycline-inducible MEOX1 shRNA cell lines were conducted according to Wiederschain and Addgene instruction manuals. Sequences for MEOX1 shRNA were obtained from the Mission shRNA Library of The RNAi Consortium; the sequences used for MEOX1 cloning were TRCN0000016108 and TRCN0000016110. TRCN0000016108 contained a top sequence of 5′-CCGGGCTGTTATTGTGGGAGGAAATCTCGAGATTTCCTCCCACAATAACAGCTTTTT-3′ and a bottom sequence of 5′-AATTAAAAAGCTGTTATTGTGGGAGGAAATCTCGAGATTTCCTCCCACAATAACAGC-3′. TRCN0000016110 contained a top sequence of 5′-CCGGCCAATGAGACAGAGAAGAAATCTCGAGATTTCTTCTCTGTCTCATTGGTTTTT-3′ and a bottom sequence of 5′-AATTAAAAACCAATGAGACAGAGAAGAAATCTCGAGATTTCTTCTCTGTCTCATTGG-3′. The Addgene Tet-pLKO-puro-scrambled plasmid (No. 47541) was used as a negative control shRNA, a gift from Charles Rudin (73). The Tet-pLKO-puro-scrambled contained negative control shRNA top sequence of 5′-CCGGCCTAAGGTTAAGTCGCCCTCGCTCGAGCGAGGGCGACTTAACCTTAGGTTTTT-3′ and a bottom sequence of 5′-AATTAAAAACCTAAGGTTAAGTCGCCCTCGCTCGAGCGAGGGCGACTTAACCTTAGG-3′. The following stable cell lines were established using the basal-like MDA-MB-468 TNBC cells: MDA-MB-468 Tet-pLKO-puro-scrambled (labeled as Scrambled Control shRNA), MDA-MB-468 Tet-pLKO-puro MEOX1 shRNA TRCN0000016108 (labeled as MEOX1 shRNA 1), and MDA-MB-468 Tet-pLKO-puro MEOX1 shRNA TRCN0000016110 (labeled as MEOX1 shRNA 2).

Mammary fat pad injections for in vivo tumor growth

All animal experimental protocols were approved by the Institutional Animal Care and Use Committee (IACUC) at the University of Michigan. Female non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice at 8-10 weeks of age were used for orthotopic fourth inguinal mammary fat pad injections. Xenograft tumor models were established by injecting 5 × 104 human breast cancer cells suspended in a 2:3 ratio of 1× PBS/Matrigel, respectively. Three groups of cells were injected, the MDA-MB-468 Tet-pLKO-puro-scrambled (labeled as Scrambled Control shRNA), MDA-MB-468 Tet-pLKO-puro MEOX1 shRNA TRCN0000016108 (labeled as MEOX1 shRNA 1), and MDA-MB-468 Tet-pLKO-puro MEOX1 shRNA TRCN0000016110 (labeled as MEOX1 shRNA 2). Five mice per group were injected for statistical analysis.

To induce MEOX1 shRNA knockdown, Envigo doxycycline (625 mg/kg) pellet diet (No. TD.08541) was administered to the mice once a week. In an adjuvant setting, MEOX1 shRNA knockdown was induced with doxycycline 3 days following surgery. In an advanced treatment setting, MEOX1 shRNA knockdown was induced with doxycycline after palpable or visible tumor growth.

RNA-Seq

BT-549 and MDA-MB-468 cell lines were transfected with 50 nm Negative Control siRNA and 50 nm MEOX1 siRNA mixture. Using Qiagen RNeasy Kit (No. 74104), cells were harvested to extract RNA following 72 h from the start of transfection. To afford better statistical analysis, two biological replicates along with the two technical replicates of each biological replicate were used for sequencing RNA. Samples were submitted for library preparation and sequencing RNA at the University of Michigan DNA Core. Libraries were prepared for mRNA, nonstrand-specific, using poly(A) selection, ∼120–nucleotide fragment lengths. RNA-seq was conducted using Illumina Sequencing with the HiSeq-4000 platform. Samples were multiplexed together and split into two lanes. Single-end sequencing was conducted with 50 nucleotide read lengths. The sequencing reads were mapped to human transcripts annotated in GENCODE using Bowtie, only uniquely mapped reads were used for further analysis. Gene expression levels were estimated as reads/kilobase/million mapped reads (RPKM) using rSeq, genes with an average RPKM less than 1.0 across all the samples were removed from analysis. Calculations for log2 (fold-change), p value, and false discovery rate were conducted using edgeR to ascertain differential gene expression between MEOX1 knockdown versus negative control.

Integrative pathway analysis (IPA) was used to assess how differential gene expression between MEOX1 knockdown versus negative control effects biological pathways. Using IPA, differential gene expressions were filtered with log2 (fold-change) >1.5 and false discovery rate <0.05. To ensure statistically significant predicted pathways are analyzed, IPA advises the use of canonical pathway activated z-scores >2 and canonical pathway inactivated z-scores of < −2. Further statistical significance is ensured by analyzing canonical pathways scoring p values <0.05.

Western blots

BT-549 and MDA-MB-468 cell lines were transfected with 50 nm Negative Control siRNA and 50 nm MEOX1 siRNA mixture. Following 72 h from the start of transfection, cells were washed twice with cold 1× PBS and harvested by scraping. Harvested cells were pelleted using centrifugation and Pierce RIPA buffer (No. 89900) with Thermo Scientific protease inhibitor mixture (No. 78410), Halt phosphatase inhibitor mixture (No. 78420), and 5 mm EDTA were used to lyse cells for protein extraction. Pierce BCA Assay Kit (No. 23227) was performed to ascertain protein concentrations. A total of 20 μg of protein was used for SDS-PAGE. Nitrocellulose or polyvinylidene difluoride membranes were utilized for protein transfers. Depending on the antibody, blocking was performed for 1 h with either 5% milk or 5% BSA. Primary antibodies were incubated rotating overnight at 4 °C. Following several washes, appropriate secondary antibodies conjugated with horseradish peroxidase were added and incubated rotating for 1 h at room temperature. After several washes, enhanced chemiluminescence (ECL) Western blotting substrate was added and blots were imaged using film. JAK/STAT pathway antibodies were purchased from Cell Signaling Technology, which were supplied as STAT Antibody Sampler Kit (No. 9939), Phospho-STAT Antibody Sampler Kit (No. 9914), Phospho-JAK Family Antibody Sampler Kit (No. 97999), and JAK/STAT Pathway Inhibitors Antibody Sampler Kit (No. 8343). Through Santa Cruz Biotechnology, antibodies for STAT5A (No. sc-271542), STAT5B (No. sc-1656), and loading control β-actin (No. sc-47778) were purchased. The manufacturer's instructions were followed for all antibodies to use specific concentrations required for optimal staining.

Statistical analysis

GraphPad Prism 7.0 was used for calculations of all statistical analysis. An unpaired t test was conducted when comparing 2 means of two unmatched groups. One-way ANOVA was conducted when measuring one variable and comparing three or more means. Two-way ANOVA was conducted when determining a change in response based on two factors and comparing three or more means.

Data availability

The RNA-seq data are located in a repository (GSE133927). Other data are to be shared upon request from Duxin Sun (duxins@umich.edu), University of Michigan.

Supplementary Material

Supporting Information

This article contains supporting information.

Author contributions—M. G., M.-C. L., and D. S. conceptualization; M. G., M.-C. L., H. J., and D. S. formal analysis; M. G. validation; M. G., M.-C. L., H. J., C.-C. L., and D. S. visualization; M. G., M.-C. L., C.-C. L., and D. S. methodology; M. G. writing-original draft; M.-C. L. and D. S. supervision; D. S. resources; D. S. investigation; D. S. project administration; D. S. writing-review and editing.

Conflict of interestThe authors declare that they have no conflicts of interest with the contents of this article.

Abbreviations—The abbreviations used are:
TNBC
triple-negative breast cancer
MEOX1
molecular target mesenchyme homeobox 1
PTEN
phosphatase and tensin homolog deleted on chromosome 10
qPCR
quantitative PCR
shRNA
short hairpin RNA
DMEM
Dulbecco's modified Eagle's medium
NOD/SCID
non-obese diabetic/severe combined immunodeficiency
IPA
integrative pathway analysis
ANOVA
analysis of variance.

References

  • 1. Lehmann B. D., Bauer J. A., Chen X., Sanders M. E., Chakravarthy A. B., Shyr Y., and Pietenpol J. A. (2011) Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 121, 2750–2767 10.1172/JCI45014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Bertucci F., Finetti P., Cervera N., Esterni B., Hermitte F., Viens P., and Birnbaum D. (2008) How basal are triple-negative breast cancers? Int. J. Cancer 123, 236–240 10.1002/ijc.23518 [DOI] [PubMed] [Google Scholar]
  • 3. Rakha E. A., Elsheikh S. E., Aleskandarany M. A., Habashi H. O., Green A. R., Powe D. G., El-Sayed M. E., Benhasouna A., Brunet J. S., Akslen L. A., Evans A. J., Blamey R., Reis-Filho J. S., Foulkes W. D., and Ellis I. O. (2009) Triple-negative breast cancer: distinguishing between basal and nonbasal subtypes. Clin. Cancer Res. 15, 2302–2310 10.1158/1078-0432.CCR-08-2132 [DOI] [PubMed] [Google Scholar]
  • 4. Carey L. A., Perou C. M., Livasy C. A., Dressler L. G., Cowan D., Conway K., Karaca G., Troester M. A., Tse C. K., Edmiston S., Deming S. L., Geradts J., Cheang M. C., Nielsen T. O., Moorman P. G., et al. (2006) Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA 295, 2492–2502 10.1001/jama.295.21.2492 [DOI] [PubMed] [Google Scholar]
  • 5. Dent R., Trudeau M., Pritchard K. I., Hanna W. M., Kahn H. K., Sawka C. A., Lickley L. A., Rawlinson E., Sun P., and Narod S. A. (2007) Triple-negative breast cancer: clinical features and patterns of recurrence. Clin. Cancer Res. 13, 4429–4434 10.1158/1078-0432.CCR-06-3045 [DOI] [PubMed] [Google Scholar]
  • 6. Haffty B. G., Yang Q., Reiss M., Kearney T., Higgins S. A., Weidhaas J., Harris L., Hait W., and Toppmeyer D. (2006) Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J. Clin. Oncol. 24, 5652–5657 10.1200/JCO.2006.06.5664 [DOI] [PubMed] [Google Scholar]
  • 7. Wiggans R. G., Woolley P. V., Smythe T., Hoth D., Macdonald J. S., Green L., and Schein P. S. (1979) Phase-II trial of tamoxifen in advanced breat cancer. Cancer Chemother. Pharmacol. 3, 45–48 10.1007/BF00254419 [DOI] [PubMed] [Google Scholar]
  • 8. Pegram M. D., Lipton A., Hayes D. F., Weber B. L., Baselga J. M., Tripathy D., Baly D., Baughman S. A., Twaddell T., Glaspy J. A., and Slamon D. J. (1998) Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. J. Clin. Oncol. 16, 2659–2671 10.1200/JCO.1998.16.8.2659 [DOI] [PubMed] [Google Scholar]
  • 9. Balko J. M., Giltnane J. M., Wang K., Schwarz L. J., Young C. D., Cook R. S., Owens P., Sanders M. E., Kuba M. G., Sánchez V., Kurupi R., Moore P. D., Pinto J. A., Doimi F. D., Gómez H., et al. (2014) Molecular profiling of the residual disease of triple-negative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets. Cancer Discov. 4, 232–245 10.1158/2159-8290.CD-13-0286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Lehmann B. D., Jovanović B., Chen X., Estrada M. V., Johnson K. N., Shyr Y., Moses H. L., Sanders M. E., and Pietenpol J. A. (2016) Refinement of triple-negative breast cancer molecular subtypes: implications for neoadjuvant chemotherapy selection. PLoS ONE 11, e0157368 10.1371/journal.pone.0157368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Carey L. A., Dees E. C., Sawyer L., Gatti L., Moore D. T., Collichio F., Ollila D. W., Sartor C. I., Graham M. L., and Perou C. M. (2007) The triple negative paradox: primary tumor chemosensitivity of breast cancer subtypes. Clin. Cancer Res. 13, 2329–2334 10.1158/1078-0432.CCR-06-1109 [DOI] [PubMed] [Google Scholar]
  • 12. Liedtke C., Mazouni C., Hess K. R., André F., Tordai A., Mejia J. A., Symmans W. F., Gonzalez-Angulo A. M., Hennessy B., Green M., Cristofanilli M., Hortobagyi G. N., and Pusztai L. (2008) Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 26, 1275–1281 10.1200/JCO.2007.14.4147 [DOI] [PubMed] [Google Scholar]
  • 13. von Minckwitz G., Untch M., Blohmer J. U., Costa S. D., Eidtmann H., Fasching P. A., Gerber B., Eiermann W., Hilfrich J., Huober J., Jackisch C., Kaufmann M., Konecny G. E., Denkert C., Nekljudova V., et al. (2012) Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J. Clin. Oncol. 30, 1796–1804 10.1200/JCO.2011.38.8595 [DOI] [PubMed] [Google Scholar]
  • 14. Cortazar P., Zhang L., Untch M., Mehta K., Costantino J. P., Wolmark N., Bonnefoi H., Cameron D., Gianni L., Valagussa P., Swain S. M., Prowell T., Loibl S., Wickerham D. L., Bogaerts J., et al. (2014) Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 384, 164–172 10.1016/S0140-6736(13)62422-8 [DOI] [PubMed] [Google Scholar]
  • 15. Cancer Genome Atlas, N. (2012) Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 10.1038/nature11412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Powell E., Piwnica-Worms D., and Piwnica-Worms H. (2014) Contribution of p53 to metastasis. Cancer Discov. 4, 405–414 10.1158/2159-8290.CD-13-0136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Marty B., Maire V., Gravier E., Rigaill G., Vincent-Salomon A., Kappler M., Lebigot I., Djelti F., Tourdès A., Gestraud P., Hupé P., Barillot E., Cruzalegui F., Tucker G. C., Stern M. H., et al. (2008) Frequent PTEN genomic alterations and activated phosphatidylinositol 3-kinase pathway in basal-like breast cancer cells. Breast Cancer Res. 10, R101 10.1186/bcr2204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. López-Knowles E., O'Toole S. A., McNeil C. M., Millar E. K. A., Qiu M. R., Crea P., Daly R. J., Musgrove E. A., and Sutherland R. L. (2010) PI3K pathway activation in breast cancer is associated with the basal-like phenotype and cancer-specific mortality. Int. J. Cancer 126, 1121–1131 10.1002/ijc.24831 [DOI] [PubMed] [Google Scholar]
  • 19. Liu J. C., Voisin V., Wang S., Wang D. Y., Jones R. A., Datti A., Uehling D., Al-Awar R., Egan S. E., Bader G. D., Tsao M., Mak T. W., and Zacksenhaus E. (2014) Combined deletion of PTEN and p53 in mammary epithelium accelerates triple-negative breast cancer with dependency on eEF2K. EMBO Mol. Med. 6, 1542–1560 10.15252/emmm.201404402 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Shah S. P., Roth A., Goya R., Oloumi A., Ha G., Zhao Y., Turashvili G., Ding J., Tse K., Haffari G., Bashashati A., Prentice L. M., Khattra J., Burleigh A., Yap D., et al. (2012) The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature 486, 395–399 10.1038/nature10933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Lawrence R. T., Perez E. M., Hernández D., Miller C. P., Haas K. M., Irie H. Y., Lee S. I., Blau C. A., and Villén J. (2015) The proteomic landscape of triple-negative breast cancer. Cell Rep. 11, 630–644 10.1016/j.celrep.2015.03.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Magee J. A., Piskounova E., and Morrison S. J. (2012) Cancer stem cells: impact, heterogeneity, and uncertainty. Cancer Cell 21, 283–296 10.1016/j.ccr.2012.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Wang S., Liu J. C., Kim D., Datti A., and Zacksenhaus E. (2016) Targeted PTEN deletion plus p53-R270H mutation in mouse mammary epithelium induces aggressive claudin-low and basal-like breast cancer. Breast Cancer Res. 18, 9 10.1186/s13058-015-0668-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Iwakuma T., and Lozano G. (2007) Crippling p53 activities via knock-in mutations in mouse models. Oncogene 26, 2177–2184 10.1038/sj.onc.1210278 [DOI] [PubMed] [Google Scholar]
  • 25. Muller P. A., Vousden K. H., and Norman J. C. (2011) p53 and its mutants in tumor cell migration and invasion. J. Cell Biol. 192, 209–218 10.1083/jcb.201009059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Liu G., McDonnell T. J., Montes de Oca Luna R., Kapoor M., Mims B., El-Naggar A. K., and Lozano G. (2000) High metastatic potential in mice inheriting a targeted p53 missense mutation. Proc. Natl. Acad. Sci. U.S.A. 97, 4174–4179 10.1073/pnas.97.8.4174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Olive K. P., Tuveson D. A., Ruhe Z. C., Yin B., Willis N. A., Bronson R. T., Crowley D., and Jacks T. (2004) Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 119, 847–860 10.1016/j.cell.2004.11.004 [DOI] [PubMed] [Google Scholar]
  • 28. Lang G. A., Iwakuma T., Suh Y. A., Liu G., Rao V. A., Parant J. M., Valentin-Vega Y. A., Terzian T., Caldwell L. C., Strong L. C., El-Naggar A. K., and Lozano G. (2004) Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell 119, 861–872 10.1016/j.cell.2004.11.006 [DOI] [PubMed] [Google Scholar]
  • 29. Harvey M., McArthur M. J., Montgomery C. A. Jr., Butel J. S., Bradley A., and Donehower L. A. (1993) Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nat. Genet. 5, 225–229 10.1038/ng1193-225 [DOI] [PubMed] [Google Scholar]
  • 30. Hollander M. C., Blumenthal G. M., and Dennis P. A. (2011) PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat. Rev. Cancer 11, 289–301 10.1038/nrc3037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Zhang H. Y., Liang F., Jia Z. L., Song S. T., and Jiang Z. F. (2013) PTEN mutation, methylation and expression in breast cancer patients. Oncol Lett 6, 161–168 10.3892/ol.2013.1331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Luo S., Chen J., and Mo X. (2016) The association of PTEN hypermethylation and breast cancer: a meta-analysis. OncoTargets Ther. 9, 5643–5650 10.2147/OTT.S111684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Jones N., Bonnet F., Sfar S., Lafitte M., Lafon D., Sierankowski G., Brouste V., Banneau G., Tunon de Lara C., Debled M., MacGrogan G., Longy M., and Sevenet N. (2013) Comprehensive analysis of PTEN status in breast carcinomas. Int. J. Cancer 133, 323–334 10.1002/ijc.28021 [DOI] [PubMed] [Google Scholar]
  • 34. Lynch E. D., Ostermeyer E. A., Lee M. K., Arena J. F., Ji H., Dann J., Swisshelm K., Suchard D., MacLeod P. M., Kvinnsland S., Gjertsen B. T., Heimdal K., Lubs H., Møller P., and King M. C. (1997) Inherited mutations in PTEN that are associated with breast cancer, cowden disease, and juvenile polyposis. Am. J. Hum. Genet. 61, 1254–1260 10.1086/301639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Knobbe C. B., Lapin V., Suzuki A., and Mak T. W. (2008) The roles of PTEN in development, physiology and tumorigenesis in mouse models: a tissue-by-tissue survey. Oncogene 27, 5398–5415 10.1038/onc.2008.238 [DOI] [PubMed] [Google Scholar]
  • 36. Saal L. H., Gruvberger-Saal S. K., Persson C., Lövgren K., Jumppanen M., Staaf J., Jönsson G., Pires M. M., Maurer M., Holm K., Koujak S., Subramaniyam S., Vallon-Christersson J., Olsson H., Su T., et al. (2008) Recurrent gross mutations of the PTEN tumor suppressor gene in breast cancers with deficient DSB repair. Nat. Genet. 40, 102–107 10.1038/ng.2007.39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Korkaya H., Paulson A., Charafe-Jauffret E., Ginestier C., Brown M., Dutcher J., Clouthier S. G., and Wicha M. S. (2009) Regulation of mammary stem/progenitor cells by PTEN/Akt/β-catenin signaling. PLos Biol. 7, e1000121 10.1371/journal.pbio.1000121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Kim G., Ouzounova M., Quraishi A. A., Davis A., Tawakkol N., Clouthier S. G., Malik F., Paulson A. K., D'Angelo R. C., Korkaya S., Baker T. L., Esen E. S., Prat A., Liu S., Kleer C. G., et al. (2015) SOCS3-mediated regulation of inflammatory cytokines in PTEN and p53 inactivated triple negative breast cancer model. Oncogene 34, 671–680 10.1038/onc.2014.4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Lin J., Sampath D., Nannini M. A., Lee B. B., Degtyarev M., Oeh J., Savage H., Guan Z., Hong R., Kassees R., Lee L. B., Risom T., Gross S., Liederer B. M., Koeppen H., et al. (2013) Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models. Clin. Cancer Res. 19, 1760–1772 10.1158/1078-0432.CCR-12-3072 [DOI] [PubMed] [Google Scholar]
  • 40. Vivanco I., and Sawyers C. L. (2002) The phosphatidylinositol 3-kinase–AKT pathway in human cancer. Nat. Rev. Cancer 2, 489–501 10.1038/nrc839 [DOI] [PubMed] [Google Scholar]
  • 41. Lee J. J., Loh K., and Yap Y.-S. (2015) PI3K/Akt/mTOR inhibitors in breast cancer. Cancer Biol. Med. 12, 342–354 10.7497/j.issn.2095-3941.2015.0089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kim S.-B., Dent R., Im S.-A., Espié M., Blau S., Tan A. R., Isakoff S. J., Oliveira M., Saura C., Wongchenko M. J., Kapp A. V., Chan W. Y., Singel S. M., Maslyar D. J., and Baselga J, LOTUS investigators, (2017) Ipatasertib plus paclitaxel versus placebo plus paclitaxel as first-line therapy for metastatic triple-negative breast cancer (LOTUS): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 18, 1360–1372 10.1016/S1470-2045(17)30450-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Sun L., Burnett J., Gasparyan M., Xu F., Jiang H., Lin C.-C., Myers I., Korkaya H., Liu Y., Connarn J., He H., Zhang N., Wicha M. S., and Sun D. (2016) Novel cancer stem cell targets during epithelial to mesenchymal transition in PTEN-deficient trastuzumab-resistant breast cancer. Oncotarget 7, 51408–51422 10.18632/oncotarget.9839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Barnabas N., and Cohen D. (2013) Phenotypic and molecular characterization of MCF10DCIS and SUM breast cancer cell lines. Int. J. Breast Cancer 2013, 872743 10.1155/2013/872743 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Concin N., Zeillinger C., Tong D., Stimpfl M., König M., Printz D., Stonek F., Schneeberger C., Hefler L., Kainz C., Leodolter S., Haas O. A., and Zeillinger R. (2003) Comparison of p53 mutational status with mRNA and protein expression in a panel of 24 human breast carcinoma cell lines. Breast Cancer Res. Treat. 79, 37–46 10.1023/A:1023351717408 [DOI] [PubMed] [Google Scholar]
  • 46. Hollestelle A., Nagel J. H. A., Smid M., Lam S., Elstrodt F., Wasielewski M., Ng S. S., French P. J., Peeters J. K., Rozendaal M. J., Riaz M., Koopman D. G., ten Hagen T. L. M., de Leeuw B. H. C. G. M., Zwarthoff E. C., et al. (2010) Distinct gene mutation profiles among luminal-type and basal-type breast cancer cell lines. Breast Cancer Res. Treat. 121, 53–64 10.1007/s10549-009-0460-8 [DOI] [PubMed] [Google Scholar]
  • 47. Hu X., Stern H. M., Ge L., O'Brien C., Haydu L., Honchell C. D., Haverty P. M., Peters B. A., Wu T. D., Amler L. C., Chant J., Stokoe D., Lackner M. R., and Cavet G. (2009) Genetic alterations and oncogenic pathways associated with breast cancer subtypes. Mol. Cancer Res. 7, 511–522 10.1158/1541-7786.MCR-08-0107 [DOI] [PubMed] [Google Scholar]
  • 48. Leroy B., Girard L., Hollestelle A., Minna J. D., Gazdar A. F., and Soussi T. (2014) Analysis of TP53 mutation status in human cancer cell lines: a reassessment. Hum. Mutat. 35, 756–765 10.1002/humu.22556 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Meric-Bernstam F., Akcakanat A., Chen H. Q., Do K. A., Sangai T., Adkins F., Gonzalez-Angulo A. M., Rashid A., Crosby K., Dong M., Phan A. T., Wolff R. A., Gupta S., Mills G. B., and Yao J. (2012) PIK3CA/PTEN mutations and Akt activation as markers of sensitivity to allosteric mTOR inhibitors. Clin. Cancer Res. 18, 1777–1789 10.1158/1078-0432.CCR-11-2123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Neve R. M., Chin K., Fridlyand J., Yeh J., Baehner F. L., Fevr T., Clark L., Bayani N., Coppe J. P., Tong F., Speed T., Spellman P. T., DeVries S., Lapuk A., Wang N. J., et al. (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10, 515–527 10.1016/j.ccr.2006.10.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wasielewski M., Elstrodt F., Klijn J. G. M., Berns E. M. J. J., and Schutte M. (2006) Thirteen new p53 gene mutants identified among 41 human breast cancer cell lines. Breast Cancer Res. Treat. 99, 97–101 10.1007/s10549-006-9186-z [DOI] [PubMed] [Google Scholar]
  • 52. Weigelt B., Warne P. H., and Downward J. (2011) PIK3CA mutation, but not PTEN loss of function, determines the sensitivity of breast cancer cells to mTOR inhibitory drugs. Oncogene 30, 3222–3233 10.1038/onc.2011.42 [DOI] [PubMed] [Google Scholar]
  • 53. Shaw F. L., Harrison H., Spence K., Ablett M. P., Simões B. M., Farnie G., and Clarke R. B. (2012) A detailed mammosphere assay protocol for the quantification of breast stem cell activity. J. Mammary Gland Biol. 17, 111–117 10.1007/s10911-012-9255-3 [DOI] [PubMed] [Google Scholar]
  • 54. Fife C. M., McCarroll J. A., and Kavallaris M. (2014) Movers and shakers: cell cytoskeleton in cancer metastasis. Brit. J. Pharmacol. 171, 5507–5523 10.1111/bph.12704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Olson M. F., and Sahai E. (2009) The actin cytoskeleton in cancer cell motility. Clin. Exp. Metastasis 26, 273–287 10.1007/s10585-008-9174-2 [DOI] [PubMed] [Google Scholar]
  • 56. Yamaguchi H., and Condeelis J. (2007) Regulation of the actin cytoskeleton in cancer cell migration and invasion. Biochim. Biophys. Acta 1773, 642–652 10.1016/j.bbamcr.2006.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Candia A. F., Hu J., Crosby J., Lalley P. A., Noden D., Nadeau J. H., and Wright C. V. (1992) Mox-1 and Mox-2 define a novel homeobox gene subfamily and are differentially expressed during early mesodermal patterning in mouse embryos. Development 116, 1123–1136 [DOI] [PubMed] [Google Scholar]
  • 58. Candia A. F., and Wright C. V. (1995) The expression pattern of Xenopus Mox-2 implies a role in initial mesodermal differentiation. Mech. Dev. 52, 27–36 10.1016/0925-4773(95)00384-D [DOI] [PubMed] [Google Scholar]
  • 59. Candia A. F., and Wright C. V. (1996) Differential localization of Mox-1 and Mox-2 proteins indicates distinct roles during development. Int. J. Dev. Biol. 40, 1179–1184 [PubMed] [Google Scholar]
  • 60. Mankoo B. S., Collins N. S., Ashby P., Grigorieva E., Pevny L. H., Candia A., Wright C. V., Rigby P. W., and Pachnis V. (1999) Mox2 is a component of the genetic hierarchy controlling limb muscle development. Nature 400, 69–73 10.1038/21892 [DOI] [PubMed] [Google Scholar]
  • 61. Mankoo B. S., Skuntz S., Harrigan I., Grigorieva E., Candia A., Wright C. V., Arnheiter H., and Pachnis V. (2003) The concerted action of Meox homeobox genes is required upstream of genetic pathways essential for the formation, patterning and differentiation of somites. Development 130, 4655–4664 10.1242/dev.00687 [DOI] [PubMed] [Google Scholar]
  • 62. Thiaville M. M., Stoeck A., Chen L., Wu R. C., Magnani L., Oidtman J., Shih Ie M., Lupien M., and Wang T. L. (2012) Identification of PBX1 target genes in cancer cells by global mapping of PBX1 binding sites. PLoS ONE 7, e36054 10.1371/journal.pone.0036054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Calo V., Migliavacca M., Bazan V., Macaluso M., Buscemi M., Gebbia N., and Russo A. (2003) STAT proteins: from normal control of cellular events to tumorigenesis. J. Cell. Physiol. 197, 157–168 10.1002/jcp.10364 [DOI] [PubMed] [Google Scholar]
  • 64. Yu H., and Jove R. (2004) The STATs of cancer–new molecular targets come of age. Nat. Rev. Cancer 4, 97–105 10.1038/nrc1275 [DOI] [PubMed] [Google Scholar]
  • 65. O'Shea J. J., Schwartz D. M., Villarino A. V., Gadina M., McInnes I. B., and Laurence A. (2015) The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu. Rev. Med. 66, 311–328 10.1146/annurev-med-051113-024537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Zhang W. J., Li B. H., Yang X. Z., Li P. D., Yuan Q., Liu X. H., Xu S. B., Zhang Y., Yuan J., Gerhard G. S., Masker K. K., Dong C., Koltun W. A., and Chorney M. J. (2008) IL-4-induced Stat6 activities affect apoptosis and gene expression in breast cancer cells. Cytokine 42, 39–47 10.1016/j.cyto.2008.01.016 [DOI] [PubMed] [Google Scholar]
  • 67. Ferbeyre G., and Moriggl R. (2011) The role of Stat5 transcription factors as tumor suppressors or oncogenes. Biochim. Biophys. Acta 1815, 104–114 10.1016/j.bbcan.2010.10.004 [DOI] [PubMed] [Google Scholar]
  • 68. Carpenter R. L., and Lo H. W. (2014) STAT3 target genes relevant to human cancers. Cancers 6, 897–925 10.3390/cancers6020897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Banerjee K., and Resat H. (2016) Constitutive activation of STAT3 in breast cancer cells: a review. Int. J. Cancer 138, 2570–2578 10.1002/ijc.29923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Leitner N. R., Witalisz-Siepracka A., Strobl B., and Müller M. (2017) Tyrosine kinase 2-surveillant of tumours and bona fide oncogene. Cytokine 89, 209–218 10.1016/j.cyto.2015.10.015 [DOI] [PubMed] [Google Scholar]
  • 71. Mainland R. L., Lyons T. A., Ruth M. M., and Kramer J. M. (2017) Optimal RNA isolation method and primer design to detect gene knockdown by qPCR when validating Drosophila transgenic RNAi lines. BMC Res. Notes 10, 647 10.1186/s13104-017-2959-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Wiederschain D., Wee S., Chen L., Loo A., Yang G. Z., Huang A., Chen Y., Caponigro G., Yao Y. M., Lengauer C., Sellers W. R., and Benson J. D. (2009) Single-vector inducible lentiviral RNAi system for oncology target validation. Cell Cycle 8, 498–504 10.4161/cc.8.3.7701 [DOI] [PubMed] [Google Scholar]
  • 73. Rudin C. M., Durinck S., Stawiski E. W., Poirier J. T., Modrusan Z., Shames D. S., Bergbower E. A., Guan Y. H., Shin J., Guillory J., Rivers C. S., Foo C. K., Bhatt D., Stinson J., Gnad F., et al. (2012) Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat. Genet 44, 1111–1116 10.1038/ng.2405 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Data Availability Statement

The RNA-seq data are located in a repository (GSE133927). Other data are to be shared upon request from Duxin Sun (duxins@umich.edu), University of Michigan.


Articles from The Journal of Biological Chemistry are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES