The ubiquitin ligases CBL and CBL-B are negative regulators of tyrosine kinase signaling with established roles in the immune system. However, their physiological roles in epithelial tissues are unknown. Here, we used MMTV-Cre-mediated Cbl gene deletion on a Cbl-b null background, as well as a tamoxifen-inducible mammary stem cell (MaSC)-specific Cbl and Cbl-b double knockout (Cbl/Cbl-b DKO) using Lgr5-EGFP-IRES-CreERT2, to demonstrate a mammary epithelial cell-autonomous requirement of CBL and CBL-B in the maintenance of MaSCs. Using a newly engineered tamoxifen-inducible Cbl and Cbl-b deletion model with a dual fluorescent reporter (Cblflox/flox; Cbl-bflox/flox; Rosa26-CreERT; mT/mG), we show that Cbl/Cbl-b DKO in mammary organoids leads to hyperactivation of AKT-mTOR signaling with depletion of MaSCs. Chemical inhibition of AKT or mTOR rescued MaSCs from Cbl/Cbl-b DKO-induced depletion. Our studies reveal a novel, cell-autonomous requirement of CBL and CBL-B in epithelial stem cell maintenance during organ development and remodeling through modulation of mTOR signaling.

Postnatal organ development and maintenance require organ-resident stem cells. While mechanisms of hematopoietic stem cell (HSC) maintenance are well understood, those that maintain epithelial stem cells have only recently become a focus of investigation. This is in part due to the rapid turnover of many epithelia and an emerging interest in the stem cell-like hierarchy of cancers, the clear majority of which originate in epithelial tissues. The mammary gland (MG) provides an excellent model to examine the mechanisms of adult epithelial stem cell maintenance, since a large part of ductal growth and branching takes place during pubertal development (Robinson et al., 2001; Gjorevski and Nelson, 2011).

Recently developed approaches to isolate and propagate mammary stem cells (MaSCs) in vitro, and to examine their self-renewal, differentiation and organ-regenerating abilities, make them excellent models to investigate mechanisms of epithelial stem cell homeostasis (Stingl et al., 2006a,b; Visvader, 2009). A better understanding of these mechanisms is important given the similarities of normal and cancer stem cell hierarchies and gene expression (Beck and Blanpain, 2013; Nguyen et al., 2012), as well as the prognostic significance of MaSC transcriptional signatures in breast cancer patients (Pece et al., 2010; Siegel and Muller, 2010; Soady et al., 2015).

Signaling pathways that regulate MG development also regulate MaSCs. For example, the Wnt signaling pathway regulates mammary development as well as MaSC self-renewal (van Amerongen et al., 2012). Wnt ligands help propagate MaSCs in vitro (Kessenbrock et al., 2013; Wang et al., 2013; Zeng and Nusse, 2010) and partly mediate the hormonal regulation of MaSCs (Cai et al., 2014). The Wnt pathway target gene Lgr5, which marks epithelial stem cells in the intestine, skin and other tissues (Barker et al., 2007, 2013), was recently shown to mark MaSCs (Plaks et al., 2013; Rios et al., 2014). Lgr5+ MaSCs can regenerate a MG upon transplantation (Plaks et al., 2013). Recently, Lgr5 was reported to be among the top 5% of 329 genes that are highly expressed in the MaSCs present among basal cells, as compared with other mammary epithelial cell (MEC) populations (Soady et al., 2015).

Receptor tyrosine kinases (RTKs), including members of the ErbB family, c-MET, RON, IGFR1/2, ephrin A2, among others, regulate MG development and MaSC maintenance (Gjorevski and Nelson, 2011; Hynes and Watson, 2010). FGFR2, for example, is crucial for MaSC maintenance (Pond et al., 2013) and RTK ligands are required for in vitro culture of MaSCs (Dontu et al., 2003; Guo et al., 2012). Dysregulation of precise signaling from RTKs and other receptors often leads to oncogenesis (Hynes and Watson, 2010; Korkaya et al., 2008).

Members of the CBL family (CBL, CBL-B and CBL-C in mammals) of ubiquitin ligases serve as negative regulators of protein tyrosine kinases (PTKs), including RTKs and non-receptor PTKs (Mohapatra et al., 2013). In contrast to substantial evidence supporting key physiological roles of CBL proteins (CBL/CBL-B) in hematopoietic and immune systems (An et al., 2015; Duan et al., 2004; Naramura et al., 2010; Thien and Langdon, 2005), their roles in epithelial tissues are essentially unknown. Cbl-c (also known as Cblc) mRNA is primarily expressed in epithelia, but a protein product has not been clearly demonstrated in such epithelia, and germline Cbl-c deletion is without an overt phenotype (Griffiths et al., 2003). A mammary epithelium-intrinsic role of CBL family proteins remains unknown. Transcriptome data show that CBL and CBL-B are expressed in the mammary epithelium, with CBL-B expression enriched in MaSCs (Lim et al., 2010). The embryonic lethality of germline Cbl and Cbl-b (also known as Cblb) double knockout (Cbl/Cbl-b DKO) in mice (Naramura et al., 2002), the exaggeration of immune phenotypes of Cbl-b deficiency by conditional Cbl deletion in immune cells (Kitaura et al., 2007; Naramura et al., 2002), a myeloproliferative disorder (MPD) upon Cbl/Cbl-b DKO in HSCs (An et al., 2015; Naramura et al., 2010), and the apparent lack of mammary epithelial-intrinsic and other epithelial phenotypes in Cbl−/− or Cbl-b−/− mice strongly suggest redundant functions of CBL and CBL-B in epithelia.

To investigate the epithelial cell-intrinsic roles of CBL and CBL-B, we used a conditional DKO model in which floxed Cbl was selectively deleted in the mammary epithelium on a germline Cbl-b−/− background using MMTV-Cre (Wagner et al., 1997). Since concomitant DKO in a small fraction of HSCs in this model leads to a MPD (An et al., 2015; Naramura et al., 2010), we characterized the MG development prior to significant MPD and by using a transplant approach. These analyses revealed a redundant but essential epithelium-intrinsic requirement for CBL and CBL-B in pubertal MG development. Cbl/Cbl-b DKO mammary epithelium exhibited shrinkage of the MaSC-containing basal compartment, which led us to develop a novel MaSC-specific Cbl/Cbl-b DKO model in which floxed Cbl is inducibly deleted only in Lgr5+ MaSCs. We also generated a novel mouse model in which floxed Cbl and Cbl-b can be inducibly deleted in isolated basal MECs upon tamoxifen treatment (Goetz et al., 2016). Complementary evidence from these genetic models establishes that CBL and CBL-B are redundantly required to maintain MaSCs, apparently by controlling the level of AKT-mTOR signaling.

MMTV-Cre-mediated Cbl deletion on a Cbl-b null background (conditional DKO) leads to impaired mouse MG development

Real-time qPCR analyses of FACS-purified luminal and basal cell fractions of the mouse MG confirmed that all three CBL family genes are expressed in epithelial compartments (Fig. S1A). Since an endogenous CBL-C protein remains to be demonstrated (Mohapatra et al., 2013), while strong evidence supports redundant but crucial roles of CBL and CBL-B (Mohapatra et al., 2013; Naramura et al., 2002), we investigated the impact of mammary epithelial-intrinsic Cbl and Cbl-b DKO using Cbl-b null mice with MMTV-Cre-induced mammary epithelial deletion of floxed Cbl and expression of lacZ reporter (Naramura et al., 2010). The Cre+Cblf/+; Cbl-b+/− littermates served as Cre controls. X-gal staining of MG whole-mounts at 5-6 weeks of age indicated efficient Cre-mediated recombination in both control and DKO mice (Fig. S1B). Concurrent nuclear Fast Red and X-gal staining confirmed recombination in both luminal and basal compartments (Fig. S1C). Separately, the expression of a GFP reporter confirmed the MMTV-Cre-mediated gene deletion in the DKO and Cre control mice (Fig. S1D).

Since MMTV-Cre-induced DKO leads to MPD by 10 weeks of age, we analyzed the postnatal MG growth in 5-, 7- and 9-week-old virgin females. Compared with littermate controls, the DKO mice exhibited significantly retarded mammary ductal outgrowths (Fig. 1A), with significant reduction in the number of branch points, ductal length and fat pad filling (Fig. 1B-D). Western blotting (WB) confirmed the CBL deletion and the lack of CBL-B expression in DKO MGs (Fig. 1E), and immunohistochemical (IHC) staining revealed this to be in the mammary epithelium (Fig. 1F). Cbl-b KO mice show a compensatory increase in CBL expression in several tissues, including MG (Fig. 1E).

Fig. 1.

Impaired mammary gland (MG) development in Cbl/Cbl-b DKO mice. (A) Carmine alum-stained whole-mounts of Cbl/Cbl-b DKO, Cbl KO and Cbl-b KO virgin female mice at 5, 7 and 9 weeks of age. (B-D) Images as in A were analyzed to quantify differences in ductal length (B), number of branch points (C) and percentage of fat pad filling (D). Data represent mean±s.e.m. of four animals per group. *P≤0.05, ***P≤0.001 (Student's t-test). (E) Whole MG lysates from 6-week-old control, Cbl-b KO or Cbl KO, and 6- or 8-week-old Cbl/Cbl-b DKO mice were blotted with antibodies against CBL, CBL-B or HSC70 (HSPA8) to reveal deletion of CBL and CBL-B according to genotype. (F) IHC of MG sections from 6-week-old control, Cbl/Cbl-b DKO and Cbl KO mice with anti-CBL monoclonal antibody was performed using avidin-biotin amplification. CBL staining (brown) is seen in epithelial cells lining the ducts in control but not in DKO MG. Inset shows magnified images of brown staining seen in control not in DKO.

Fig. 1.

Impaired mammary gland (MG) development in Cbl/Cbl-b DKO mice. (A) Carmine alum-stained whole-mounts of Cbl/Cbl-b DKO, Cbl KO and Cbl-b KO virgin female mice at 5, 7 and 9 weeks of age. (B-D) Images as in A were analyzed to quantify differences in ductal length (B), number of branch points (C) and percentage of fat pad filling (D). Data represent mean±s.e.m. of four animals per group. *P≤0.05, ***P≤0.001 (Student's t-test). (E) Whole MG lysates from 6-week-old control, Cbl-b KO or Cbl KO, and 6- or 8-week-old Cbl/Cbl-b DKO mice were blotted with antibodies against CBL, CBL-B or HSC70 (HSPA8) to reveal deletion of CBL and CBL-B according to genotype. (F) IHC of MG sections from 6-week-old control, Cbl/Cbl-b DKO and Cbl KO mice with anti-CBL monoclonal antibody was performed using avidin-biotin amplification. CBL staining (brown) is seen in epithelial cells lining the ducts in control but not in DKO MG. Inset shows magnified images of brown staining seen in control not in DKO.

Hematoxylin and Eosin (H&E) staining of sections revealed sparser ducts in the MGs of 6-week-old DKO mice, but the overall basal and luminal layers were intact (Fig. S2A), a conclusion confirmed by immunofluorescence staining for basal [keratin 5 (K5)] and luminal [keratin 8 (K8)] cytokeratins (Fig. S2B).

The smaller size of DKO MGs could reflect reduced proliferation, increased apoptosis, or both, as their balance is important in mammary morphogenesis (Humphreys et al., 1996; McCaffrey and Macara, 2009). Ki67 staining showed reduced proliferation in DKO mammary ducts but not in terminal end buds (TEBs), compared with controls (Fig. S2C-F). Cleaved caspase-3 staining revealed a comparable level of apoptosis in the TEBs of control and DKO mice (Fig. S2G).

Although the analyses above were performed prior to overt MPD development, we used a transplantation approach (Deome et al., 1959; Stingl et al., 2006a,b) to rule out the possibility that subclinical MPD might have impaired MG development. Contralateral transplantation of control and DKO mammary tissue was performed in NSG hosts. Carmine alum and X-gal staining of MG whole-mounts 10 weeks post-transplant revealed significantly less fat pad filling and reduced branching in DKO transplants (Fig. S3A,B). Compared with the extensive growth and alveolar differentiation of control transplants during pregnancy, DKO transplants were dramatically smaller with less fat pad filling and reduced ductal elongation, although the alveolar differentiation appeared intact (Fig. S3B).

Next, to address whether the DKO phenotype was MEC-autonomous, FACS-sorted lineage-negative (Lin; epithelial and stromal cells) or Lin/EpCAM+ (epithelial cells only) MECs from 6-week-old Cbl/Cbl-b DKO versus control donors were transplanted in NSG mice. Whereas all the control Lin or Lin/EpCAM+ MEC transplants led to mammary outgrowths, none of the DKO transplants showed any outgrowths (Fig. S3C,D). Together, our results conclusively demonstrate that deletion of Cbl and Cbl-b in MECs leads to a cell-autonomous impairment of MG growth and branching.

Cbl/Cbl-b DKO shrinks the MaSC-containing basal MEC population

Impaired MG development in Cbl/Cbl-b DKO mice and their inability to regenerate the mammary epithelium upon transplant suggested potential impairment of DKO MaSCs. We therefore analyzed dissociated MECs from 6- or 8-week-old control or DKO mice by FACS, with staining for CD29 (ITGB1), CD24 and EpCAM, which separate MECs into basal (CD29hi CD24+ or EpCAMlow CD29hi) and luminal (CD29low CD24+ or EpCAMhi CD29low) populations (Shehata et al., 2012; Smalley et al., 2012). The former includes MaSCs and mature myoepithelial cells, whereas the latter includes luminal progenitors and mature luminal cells (Shackleton et al., 2006; Stingl et al., 2006a,b). Although we used both CD24/CD29 and EpCAM/CD29 combinations, better separation of subsets was seen in FACS plots with EpCAM/CD29 combination (Fig. S4A,B). This analysis revealed that Cbl/Cbl-b DKO MGs yielded significantly fewer Lin/EpCAM+ MECs in total, consistent with reduced MG size in these mice (Fig. S5A,B). Notably, the basal cell percentage was significantly lower, both at 6 and 8 weeks, with a corresponding increase in the proportion of luminal cells (Fig. 2A-D). However, the absolute number of luminal cells was reduced in DKO MGs (Fig. S5B,C). Given the overall reduction in the number of MECs in DKO mice (Fig. S5B,D), the smaller size of their basal MEC compartment suggested a particularly severe reduction in all or some of the cell types that comprise this compartment (Yamaji et al., 2009).

Fig. 2.

Isolated MMTV-Cre-induced Cbl/Cbl-b DKO mammary epithelial cells (MECs) show a reduction in the proportion of basal cells with a relative increase in the proportion of luminal cells. (A,C) Representative pseudocolored FACS plots, gating on basal (CD29hi EpCAMlow) and luminal (CD29low EpCAMhi) populations, show a smaller basal and a larger luminal subset in DKO versus control mice. (B,D) Quantitation of the relative percentages at 6 weeks (B, n=9) and 8 weeks (D, n=5) of age. (E) GFP+ basal and luminal cell subsets of control versus Cbl/Cbl-b DKO mice bearing a GFP reporter, indicating a reduction in the basal population and increase in the luminal population in DKO compared with control. (F) Quantitation of the percentage of basal and luminal GFP+ cells in Cbl/Cbl-b DKO compared with control mice (n=4) (as in E). (G) FACS dot plots showing mammary stem cell (MaSC), luminal progenitor (LP), mature luminal cell (LC) and myoepithelial cell (ME) populations. Cbl/Cbl-b DKO mice show a reduction in the proportions of stem cells, LCs and MEs, and an increase in the proportion of LPs compared with control mice (numbers within quadrants indicate percentage of total). (H) Quantitation of the relative percentages of stem cell, LP, LC and ME populations (as in G) in control versus DKO mice (n=6). Data are mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01 (Student's t-test). Note that differences in the percentage of cells within the stem cell and LP compartments are significant.

Fig. 2.

Isolated MMTV-Cre-induced Cbl/Cbl-b DKO mammary epithelial cells (MECs) show a reduction in the proportion of basal cells with a relative increase in the proportion of luminal cells. (A,C) Representative pseudocolored FACS plots, gating on basal (CD29hi EpCAMlow) and luminal (CD29low EpCAMhi) populations, show a smaller basal and a larger luminal subset in DKO versus control mice. (B,D) Quantitation of the relative percentages at 6 weeks (B, n=9) and 8 weeks (D, n=5) of age. (E) GFP+ basal and luminal cell subsets of control versus Cbl/Cbl-b DKO mice bearing a GFP reporter, indicating a reduction in the basal population and increase in the luminal population in DKO compared with control. (F) Quantitation of the percentage of basal and luminal GFP+ cells in Cbl/Cbl-b DKO compared with control mice (n=4) (as in E). (G) FACS dot plots showing mammary stem cell (MaSC), luminal progenitor (LP), mature luminal cell (LC) and myoepithelial cell (ME) populations. Cbl/Cbl-b DKO mice show a reduction in the proportions of stem cells, LCs and MEs, and an increase in the proportion of LPs compared with control mice (numbers within quadrants indicate percentage of total). (H) Quantitation of the relative percentages of stem cell, LP, LC and ME populations (as in G) in control versus DKO mice (n=6). Data are mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01 (Student's t-test). Note that differences in the percentage of cells within the stem cell and LP compartments are significant.

The purity of FACS-sorted basal or luminal cells was verified by K5 or K8 expression, respectively (Lim et al., 2010) (Fig. S6A,B). qPCR analysis of purified cell populations showed that Cbl and Cbl-b were expressed in control MECs but undetectable (Cbl signal <0.02% and Cbl-b signal <0.003% of wild type) in DKO MECs (Fig. S6C). Cbl-b expression was enriched in the basal compartment of control mice, consistent with previous microarray data (Lim et al., 2010). Recent studies have shown that the basal K5+ postnatal mouse MEC population contains bi-potent MaSCs, whereas the luminal K8+ population includes the luminal lineage-restricted progenitors (Rios et al., 2014). Quantification of mRNA levels in FACS-sorted populations by qPCR revealed reduced K5 levels in the basal compartment and increased K8 levels in the luminal compartment of DKO versus control mice (Fig. S6D,E).

We confirmed these findings using 6-week-old GFP reporter-bearing control and DKO mice, in which the GFP+ MECs represent the progeny of MMTV-Cre-expressing cells. These analyses also demonstrated a reduction in the basal cell population and a relative increase in the luminal fraction in DKO mice (Fig. 2E,F).

CD61 (ITGB3) and CD29 co-staining separates virgin MG MECs into populations that harbor MaSCs (CD29hi CD61), luminal progenitors (CD29low CD61+), mature luminal cells (CD29low CD61) and mature myoepithelial cells (CD29hi CD61+) (Carr et al., 2012; Shackleton et al., 2006; Stingl et al., 2006a,b). CD29 and CD61 co-staining and FACS analysis of MECs from 6-week-old control and DKO mice revealed that DKO MECs exhibited a significant increase in the proportion of luminal progenitors and a marked reduction of MaSCs, suggesting that Cbl/Cbl-b DKO promotes a shift of MaSCs towards luminal progenitors (Fig. 2G,H). However, we did not observe a proportionate increase in the mature luminal cell pool, suggesting that the DKO luminal progenitors are defective in their capacity to generate mature cells. The proportion of mature myoepithelial cells was comparable between control and DKO MECs.

Cbl/Cbl-b DKO basal MECs show reduced stem cell activity in organoid cultures in vitro

Organoid-forming ability in Matrigel reflects the stemness of MECs, with the MaSC-containing basal population yielding solid organoids and the luminal progenitor-containing luminal population yielding acini (Lim et al., 2009; Shackleton et al., 2006). Indeed, Matrigel culture of basal and luminal MEC populations, FACS sorted based on CD29/EpCAM (plus GFP reporter for DKO mice), showed that control MaSC-enriched basal cells form solid organoids with ∼6% efficiency, whereas luminal cells formed acini with hollow lumens at higher efficiencies (than 6%) but solid organoids only rarely (Fig. 3A). The ability of DKO basal MECs to form solid organoids was significantly reduced, whereas acini formation of DKO luminal cells was comparable to that of controls (Fig. 3B). Similar results were obtained after harvesting and replating the first passage organoids or acini for a second passage (Fig. 3C). qPCR at the end of passage 1 confirmed lack of Cbl and Cbl-b mRNA in organoids generated from DKO basal and luminal cells, excluding the possibility of organoids/acini having arisen from precursors in which floxed Cbl was not deleted (Fig. 3D). Altogether, these results further established that loss of CBL and CBL-B in the mammary epithelium impairs the ability of MaSCs to self-renew.

Fig. 3.

Demonstration of impaired MaSC renewal in MMTV-Cre-driven Cbl/Cbl-b DKO basal MECs using the in vitro organoid-forming assay. (A) Basal and luminal MEC populations were isolated from MGs of control [Cblf/f; Cbl-b+/−; MMTV-Cre(0/0)], Cre control [Cblf/+; Cbl-b+/−; MMTV-Cre(Tg/0)] and Cbl/Cbl-b DKO [Cblf/f; Cbl-b−/−; MMTV-Cre(Tg/0)] mice using FACS sorting with CD29 and EpCAM markers. Note that MMTV-Cre(Tg/0) refers to the Cre+ animal maintained as hemizygous transgene and MMTV-Cre(0/0) refers to Cre. Sorted cells were cultured in suspension and passaged a second time. Representative images of filled organoids formed by basal cells and acinar organoids formed by luminal cell populations are shown. Scale bar applies to all images in A. (B,C) The numbers of organoids greater than 100 µm in diameter were counted in three independent samples and expressed per 1000 seeded cells at first (B) and second (C) passage. (D-G) RNA isolated from organoids was used for qPCR to confirm the deletion of Cbl and lack of expression of Cbl-b. Data shown are mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Fig. 3.

Demonstration of impaired MaSC renewal in MMTV-Cre-driven Cbl/Cbl-b DKO basal MECs using the in vitro organoid-forming assay. (A) Basal and luminal MEC populations were isolated from MGs of control [Cblf/f; Cbl-b+/−; MMTV-Cre(0/0)], Cre control [Cblf/+; Cbl-b+/−; MMTV-Cre(Tg/0)] and Cbl/Cbl-b DKO [Cblf/f; Cbl-b−/−; MMTV-Cre(Tg/0)] mice using FACS sorting with CD29 and EpCAM markers. Note that MMTV-Cre(Tg/0) refers to the Cre+ animal maintained as hemizygous transgene and MMTV-Cre(0/0) refers to Cre. Sorted cells were cultured in suspension and passaged a second time. Representative images of filled organoids formed by basal cells and acinar organoids formed by luminal cell populations are shown. Scale bar applies to all images in A. (B,C) The numbers of organoids greater than 100 µm in diameter were counted in three independent samples and expressed per 1000 seeded cells at first (B) and second (C) passage. (D-G) RNA isolated from organoids was used for qPCR to confirm the deletion of Cbl and lack of expression of Cbl-b. Data shown are mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Lgr5-Cre-based conditional Cbl/Cbl-b DKO leads to shrinkage of the MaSC pool and their rapid transition into luminal progenitors

The Wnt target gene Lgr5 was recently found to mark MaSCs within the basal MEC compartment (de Visser et al., 2012; Plaks et al., 2013); such cells were shown to be bi-potent and to regenerate the MG upon transplant (Plaks et al., 2013; Rios et al., 2014). We crossed the Lgr5-EGFP-IRES-CreERT2 mice with Cblf/f; Cbl-b−/− mice to create mice for MaSC-selective and tamoxifen-inducible Cbl/Cbl-b DKO within the mammary epithelium. FACS analysis of MECs from parental Lgr5-GFP-Cre mice confirmed the presence of a small pool of Lgr5+ (Lgr5-driven GFP+) cells nearly exclusively in the basal population (Plaks et al., 2013) (Fig. S7A,B). Importantly, within 5 days of initiating tamoxifen-induced Cbl deletion in the DKO model, the Lgr5+ cells underwent a significant shrinkage compared with control (Cblf/+; Cbl-b+/−) mice (Fig. 4A,B). CD29/EpCAM-based assignment of GFP+ cells to basal and luminal compartments indicated that the proportion of Lgr5+ cells in the basal compartment decreased upon tamoxifen induction, whereas the proportion in the luminal compartment increased significantly (Fig. 4C), indicating a severe depletion of MaSCs, with a proportion of the DKO MaSCs transitioning into the luminal compartment. The tamoxifen-induced deletion of Cbl (and constitutive deletion of Cbl-b) in the sorted GFP+, but not GFP, MEC fraction was confirmed by qPCR (Fig. 4D,E), clearly establishing the MaSC-selective DKO in the Lgr5-Cre-based model.

Fig. 4.

Lgr5-CreERT-driven, tamoxifen-inducible MaSC-specific Cbl/Cbl-b DKO leads to shrinkage of the MaSC pool. (A) Timeline of tamoxifen (TAM) induction and analysis. Five-week-old Cblf/+; Cbl-b+/−; Lgr5-CreERT (MaSC Cbl control) and Cblf/f; Cbl-b−/−; Lgr5-GFP-CreERT (MaSC DKO mice) were given three daily injections of tamoxifen and analyzed 5 days after the last injection. (B) Representative FACS plots of isolated Lin MECs from MaSC control and MaSC DKO analyzed for GFP and CD29/EpCAM staining. Middle panel shows gating for Lgr5+ (GFP+) population and right panel represents compartmentalization of Lgr5+ cells within the basal and luminal populations. Percentage of cells within each population is indicated. (C) Percentage of total GFP+ cells, and of GFP+ cells within the basal and luminal populations, in MaSC DKO compared with control mice (n=6). (D,E) RNA isolated from FACS-sorted GFP+ epithelial cells from MaSC control mice and GFP+ and GFP fractions of MaSC DKO Lin cells was used for qPCR using primers specific to Cbl (D) and Cbl-b (E) to confirm Lgr5+ cell-specific deletion of Cbl as well as lack of expression of Cbl-b in MaSC DKO mice. Data shown are mean±s.e.m. *P≤0.05, **P≤0.01 (Student's t-test).

Fig. 4.

Lgr5-CreERT-driven, tamoxifen-inducible MaSC-specific Cbl/Cbl-b DKO leads to shrinkage of the MaSC pool. (A) Timeline of tamoxifen (TAM) induction and analysis. Five-week-old Cblf/+; Cbl-b+/−; Lgr5-CreERT (MaSC Cbl control) and Cblf/f; Cbl-b−/−; Lgr5-GFP-CreERT (MaSC DKO mice) were given three daily injections of tamoxifen and analyzed 5 days after the last injection. (B) Representative FACS plots of isolated Lin MECs from MaSC control and MaSC DKO analyzed for GFP and CD29/EpCAM staining. Middle panel shows gating for Lgr5+ (GFP+) population and right panel represents compartmentalization of Lgr5+ cells within the basal and luminal populations. Percentage of cells within each population is indicated. (C) Percentage of total GFP+ cells, and of GFP+ cells within the basal and luminal populations, in MaSC DKO compared with control mice (n=6). (D,E) RNA isolated from FACS-sorted GFP+ epithelial cells from MaSC control mice and GFP+ and GFP fractions of MaSC DKO Lin cells was used for qPCR using primers specific to Cbl (D) and Cbl-b (E) to confirm Lgr5+ cell-specific deletion of Cbl as well as lack of expression of Cbl-b in MaSC DKO mice. Data shown are mean±s.e.m. *P≤0.05, **P≤0.01 (Student's t-test).

In vivo Cbl/Cbl-b DKO in MaSCs impairs their organoid-forming ability in vitro

To assess the impact of in vivo Lgr5-Cre-based Cbl/Cbl-b DKO in MaSCs on their stemness, GFP+ (Lgr5+) MECs sorted from Lgr5-EGFP-Cre+ (MaSC control) or conditional DKO (MaSC DKO) mice 10 days post-tamoxifen were subjected to organoid-forming assays. Whereas Lgr5+ cells from MaSC control MGs formed solid organoids, Lgr5+ cells from tamoxifen-treated MaSC DKO mice produced significantly fewer and smaller organoids (Fig. 5A-C).

Fig. 5.

In vivo or in vitro induction of Lgr5-CreERT to impart Lgr5+ MaSC-selective Cbl/Cbl-b DKO leads to impaired MaSC self-renewal. (A-C) MECs isolated 10 days post-induction of Lgr5-GFP-IRES-CreERT mice were assayed for organoid-forming ability. (A) Representative images of organoids cultured for 9 days in 100% Matrigel (insets are enlarged images). (B) Quantification of organoid numbers; n=3. (C) Quantification of organoid size (in µm); n=3. (D,G) FACS-sorted Lgr5+ (GFP+) MECs isolated from non-induced Lgr5-GFP-IRES-CreERT control and Lgr5-GFP-CreERT; Cblf/f; Cbl-b−/− mice were cultured in 100% Matrigel for 10 days with 4-OH-T (400 nM final) added for the last 96 h. Comparable numbers of organoids were seen in MaSC control and DKO, as shown in representative pictures of organoids before 4-OH-T induction (D) and quantitation of results (G). The scale bar applies to all images in D. (E) The organoids were then dissociated and 2000 cells/well were cultured in 96-well ultra low-attachment plates in the presence of 5% Matrigel for 8 days, and imaged (first passage). (F) Likewise, secondary plating was performed. (H) Quantification of organoids after 4-OH-T induction of control and conditional DKO Lgr5+ MaSCs at passage 1 and 2. (I,J) WB analysis of 4-OH-T induced control and DKO organoids at the end of the second passage for CBL, CBL-B and HSC70. Data (n=3) are shown as mean±s.e.m. ns, P≥0.05; ***P≤0.001 (Student's t-test).

Fig. 5.

In vivo or in vitro induction of Lgr5-CreERT to impart Lgr5+ MaSC-selective Cbl/Cbl-b DKO leads to impaired MaSC self-renewal. (A-C) MECs isolated 10 days post-induction of Lgr5-GFP-IRES-CreERT mice were assayed for organoid-forming ability. (A) Representative images of organoids cultured for 9 days in 100% Matrigel (insets are enlarged images). (B) Quantification of organoid numbers; n=3. (C) Quantification of organoid size (in µm); n=3. (D,G) FACS-sorted Lgr5+ (GFP+) MECs isolated from non-induced Lgr5-GFP-IRES-CreERT control and Lgr5-GFP-CreERT; Cblf/f; Cbl-b−/− mice were cultured in 100% Matrigel for 10 days with 4-OH-T (400 nM final) added for the last 96 h. Comparable numbers of organoids were seen in MaSC control and DKO, as shown in representative pictures of organoids before 4-OH-T induction (D) and quantitation of results (G). The scale bar applies to all images in D. (E) The organoids were then dissociated and 2000 cells/well were cultured in 96-well ultra low-attachment plates in the presence of 5% Matrigel for 8 days, and imaged (first passage). (F) Likewise, secondary plating was performed. (H) Quantification of organoids after 4-OH-T induction of control and conditional DKO Lgr5+ MaSCs at passage 1 and 2. (I,J) WB analysis of 4-OH-T induced control and DKO organoids at the end of the second passage for CBL, CBL-B and HSC70. Data (n=3) are shown as mean±s.e.m. ns, P≥0.05; ***P≤0.001 (Student's t-test).

In vitro induction of Cbl/Cbl-b DKO in Lgr5+ MaSCs impairs their stemness

The number and size of organoids formed after 7 days of Matrigel culture from FACS-sorted GFP+ (Lgr5+) MECs from uninduced MaSC control or conditional MaSC Cbl/Cbl-b DKO mice were comparable (Fig. 5D,G). Next, 4-hydroxytamoxifen (4-OH-T) was added on day 7 of culture and organoids were harvested after 96 h, replated, and examined after 8 days. Compared with uninduced cultures, 4-OH-T induction of Lgr5+ MECs significantly impaired the ability to form organoids upon replating (Fig. 5E,H). Further passaging confirmed these results (Fig. 5F,H). WB of harvested organoids verified CBL and CBL-B deletion (Fig. 5I,J). Organoid assays on Lgr5+ cells isolated from Lgr5-GFP-Cre+ conditional DKO mice and cultured in the presence of 4-OH-T also showed reduced organoid-forming ability, as compared culture in the absence of 4-OH-T, owing to Cbl deletion (as confirmed by qPCR; Fig. 6A-C).

Fig. 6.

Impairment of organoid-forming ability of Lgr5+ MECs isolated from Lgr5-CreERT-based conditional Cbl/Cbl-b DKO mice requires tamoxifen induction. Lgr5+ (GFP+) cells were FACS sorted from Lgr5-GFP-CreERT; Cblf/f; Cbl-b−/− mice and organoid cultures performed as in Fig. 5E,F for 9 days either without (A, top) or with (A, bottom) 4-OH-T. (B) Quantitation of organoid numbers as in A. (C) qPCR analysis verified 4-OH-T-induced reduction in Cbl expression and lack of Cbl-b expression in both non-induced and induced organoids. (D-G) qPCR (D-F) and immunofluorescence (G) analyses were performed to assess the expression of basal (K5, D; K14, E,G) and luminal (K8, F,G) cytokeratins. (H) Quantitation of immunofluorescence staining of basal and luminal cytokeratins in organoids as in G. Data (n=3) are shown as mean±s.e.m. *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Fig. 6.

Impairment of organoid-forming ability of Lgr5+ MECs isolated from Lgr5-CreERT-based conditional Cbl/Cbl-b DKO mice requires tamoxifen induction. Lgr5+ (GFP+) cells were FACS sorted from Lgr5-GFP-CreERT; Cblf/f; Cbl-b−/− mice and organoid cultures performed as in Fig. 5E,F for 9 days either without (A, top) or with (A, bottom) 4-OH-T. (B) Quantitation of organoid numbers as in A. (C) qPCR analysis verified 4-OH-T-induced reduction in Cbl expression and lack of Cbl-b expression in both non-induced and induced organoids. (D-G) qPCR (D-F) and immunofluorescence (G) analyses were performed to assess the expression of basal (K5, D; K14, E,G) and luminal (K8, F,G) cytokeratins. (H) Quantitation of immunofluorescence staining of basal and luminal cytokeratins in organoids as in G. Data (n=3) are shown as mean±s.e.m. *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Consistent with results in the MMTV-Cre-based DKO model, qPCR analysis showed that the levels of K5 and K14 were reduced in 4-OH-T-induced DKO organoids compared with non-induced controls, whereas the expression of K8 increased (Fig. 6D-F). Immunofluorescence analyses showed an increase in the proportion of K8+ cells and a decrease in the proportion of K14+ cells in MaSC DKO organoids (Fig. 6G,H).

Depletion of MaSCs upon Cbl and Cbl-b deletion is mediated through hyperactivation of AKT-mTOR signaling

RTK signaling is crucial for MG development and the maintenance of MaSCs (Brisken et al., 2000; van Amerongen et al., 2012). PI3K-AKT-mTOR is a key signaling node downstream of activated RTK signaling (Sun and Bernards, 2014), and studies in cell models have shown that loss of Cbl or Cbl-b increases the levels of phosphorylated (p) AKT (An et al., 2015; Fang and Liu, 2001). Importantly, hyperactivation of AKT-mTOR signaling has been shown to promote differentiation and stem cell exhaustion in hematopoietic, neural and epidermal stem cell systems (Iglesias-Bartolome et al., 2012; Kalaitzidis et al., 2012; Magri et al., 2011). In view of reduced MG development and of MaSC depletion upon Cbl/Cbl-b DKO, we assessed whether AKT-mTOR signaling is hyperactivated in DKO MECs. As the number of cells available from the Lgr5-Cre-driven model was small, we used a fully tamoxifen-inducible Cbl and Cbl-b deletion model (Goetz et al., 2016) in which both Cbl and Cbl-b are floxed, and the ROSA26 locus CreERT and dual fluorescent reporter (mT/mG) alleles allow verifiable 4-OH-T-inducible in vitro Cbl and Cbl-b deletion. The MaSC-containing basal cells from Cblf/f; Cbl-bf/f; CreERT; mT/mG or Cblf/f; Cbl-bf/+; CreERT; mT/mG mice were plated in organoid cultures and induced with 4-OH-T (Fig. 7A). Whereas uninduced organoids were uniformly RFP+, 4-OH-T induction led to a switch to GFP+ organoids, indicating robust CreERT-dependent gene deletion. WB verified CBL and CBL-B deletion, concurrent with upregulation of the levels of total and pEGFR, pAKT and pmTOR in 4-OH-T-induced DKO organoids as compared with uninduced organoids (Fig. 8A). Replating of uninduced versus induced Cblf/f; Cbl-bf/f; CreERT; mT/mG organoids showed a substantial reduction in organoid growth upon in vitro Cbl and Cbl-b deletion (Fig. 7B,C), consistent with results of the MMTV-Cre and Lgr5-Cre models. PCR analysis of Cbl and Cbl-b expression showed significant reduction upon 4-OH-T induction (Fig. 7D,E). Egfr mRNA levels were not significantly upregulated upon 4-OH-T induction (Fig. 7F). Basal cell-derived organoids from Cblf/f; Cbl-bf/+; CreERT; mT/mG mice showed only a slight reduction in organoid numbers upon induction (Fig. 7C, lower panels). qPCR analysis showed lower levels of K14 and increased levels of K8 in induced versus non-induced organoids, indicating differentiation towards luminal lineages (Fig. 7G,H). Luminal cells from Cblf/f; Cbl-bf/f; CreERT; mT/mG mice yielded comparable numbers of organoids with and without 4-OH-T induction (Fig. S8A), and WB analysis showed no changes in cyclin D1 (proliferation marker) expression (Fig. S8B). WB analyses also verified CBL and CBL-B depletion and an increase in pEGFR but without an increase in total EGFR, but relatively little impact on pAKT or pERK levels (Fig. S8B). Collectively, the results of inducible deletion of both Cbl and Cbl-b further support our conclusion that CBL and CBL-B are required for MaSC maintenance, and show that their deletion hyperactivates the AKT-mTOR signaling axis.

Fig. 7.

Impaired stem cell renewal upon tamoxifen-induced, CreERT-mediated in vitro deletion in a Cbl/Cbl-b double-floxed model. (A) Schematic of basal MEC isolation from 8-week-old Cblf/f; Cbl-bf/f; CreERT; mT/mG (conditional DKO) and Cblf/f; Cbl-bf/+; CreERT; mT/mG (control) mice. Organoid cultures were established and treated with or without 4-OH-T to induce Cbl/Cbl-b deletion, tracked using a dual fluorescent reporter. (B,C) The non-induced or 4-OH-T-induced primary organoids formed by isolated basal cells from Cblf/f; Cbl-bf/f; CreERT; mT/mG (conditional DKO) and Cblf/f; Cbl-bf/+; CreERT; mT/mG (control) mice were replated (2° organoids) to assess self-renewal ability. (B) Representative images showing loss of red (RFP) and gain of green (GFP) fluorescence indicating successful CreERT activation and Cbl/Cbl-b deletion (confirmed by WB, not shown). (C) Quantitation of organoid numbers from B. Note the dramatic reduction in organoid numbers in 4-OH-T-treated double-floxed organoids only. (D-F) qPCR analyses verify the deletion of Cbl and Cbl-b, with no significant upregulation of Egfr. (G,H) Relative expression of basal (K14, G) and luminal (K8, H) cytokeratins in 4-OH-T-induced versus non-induced organoids following replating. Data (n=3) are shown as mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Fig. 7.

Impaired stem cell renewal upon tamoxifen-induced, CreERT-mediated in vitro deletion in a Cbl/Cbl-b double-floxed model. (A) Schematic of basal MEC isolation from 8-week-old Cblf/f; Cbl-bf/f; CreERT; mT/mG (conditional DKO) and Cblf/f; Cbl-bf/+; CreERT; mT/mG (control) mice. Organoid cultures were established and treated with or without 4-OH-T to induce Cbl/Cbl-b deletion, tracked using a dual fluorescent reporter. (B,C) The non-induced or 4-OH-T-induced primary organoids formed by isolated basal cells from Cblf/f; Cbl-bf/f; CreERT; mT/mG (conditional DKO) and Cblf/f; Cbl-bf/+; CreERT; mT/mG (control) mice were replated (2° organoids) to assess self-renewal ability. (B) Representative images showing loss of red (RFP) and gain of green (GFP) fluorescence indicating successful CreERT activation and Cbl/Cbl-b deletion (confirmed by WB, not shown). (C) Quantitation of organoid numbers from B. Note the dramatic reduction in organoid numbers in 4-OH-T-treated double-floxed organoids only. (D-F) qPCR analyses verify the deletion of Cbl and Cbl-b, with no significant upregulation of Egfr. (G,H) Relative expression of basal (K14, G) and luminal (K8, H) cytokeratins in 4-OH-T-induced versus non-induced organoids following replating. Data (n=3) are shown as mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Fig. 8.

Hyperactivation of AKT-mTOR signaling mediates loss of MaSC self-renewal ability upon conditional Cbl/Cbl-b deletion. (A) Basal mammary cells were isolated from 8-week-old Cblf/f; Cbl-bf/f; CreERT; mT/mG mice, plated for organoid culture for 10 days with or without 4-OH-T for the last 4 days. WB of non-induced versus induced primary organoids which exhibit CBL/CBL-B deletion and an increase in the levels of total EGFR, pEGFR, pAKT and pmTOR. (B) Basal mammary cells plated for 10 days, with or without 4-OH tamoxifen or the inhibitors MK-2206 or rapamycin at the indicated concentrations for the last 4 days. WB demonstrates the reduction in pAKT or pmTOR levels as indicators of AKT or mTOR inhibition, respectively, after primary organoid culture. Rapamycin-induced loss of pAKT signals reflects the feedback loop. Anti-CBL/CBL-B blotting verified the expected deletions. (C) Representative scanned images of induced (GFP+, green) and non-induced (RFP+, red) secondary organoid cultures without or with the indicated inhibitor treatment in primary culture. (D) Quantification of the number of organoids formed by induced (green bars) versus non-induced (red bars) MaSCs and the reversal of the effect of Cbl/Cbl-b deletion by AKT or mTOR inhibition. Data (n=3) are shown as mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

Fig. 8.

Hyperactivation of AKT-mTOR signaling mediates loss of MaSC self-renewal ability upon conditional Cbl/Cbl-b deletion. (A) Basal mammary cells were isolated from 8-week-old Cblf/f; Cbl-bf/f; CreERT; mT/mG mice, plated for organoid culture for 10 days with or without 4-OH-T for the last 4 days. WB of non-induced versus induced primary organoids which exhibit CBL/CBL-B deletion and an increase in the levels of total EGFR, pEGFR, pAKT and pmTOR. (B) Basal mammary cells plated for 10 days, with or without 4-OH tamoxifen or the inhibitors MK-2206 or rapamycin at the indicated concentrations for the last 4 days. WB demonstrates the reduction in pAKT or pmTOR levels as indicators of AKT or mTOR inhibition, respectively, after primary organoid culture. Rapamycin-induced loss of pAKT signals reflects the feedback loop. Anti-CBL/CBL-B blotting verified the expected deletions. (C) Representative scanned images of induced (GFP+, green) and non-induced (RFP+, red) secondary organoid cultures without or with the indicated inhibitor treatment in primary culture. (D) Quantification of the number of organoids formed by induced (green bars) versus non-induced (red bars) MaSCs and the reversal of the effect of Cbl/Cbl-b deletion by AKT or mTOR inhibition. Data (n=3) are shown as mean±s.e.m. ns, P≥0.05; *P≤0.05, **P≤0.01, ***P≤0.001 (Student's t-test).

To assess whether AKT-mTOR hyperactivation might mediate the loss of MaSCs upon Cbl and Cbl-b deletion, we tested if specific AKT or mTOR inhibitors could rescue MaSCs from Cbl/Cbl-b DKO-induced depletion. The allosteric AKT inhibitor MK-2206 (Hirai et al., 2010; Yap et al., 2011) was added to preformed organoids generated from a Cblf/f; Cbl-bf/f; CreERT; mT/mG basal cell population with or without concurrent 4-OH-T induction at 50 nM or 100 nM, based on IC50 values reported in other systems (Sangai et al., 2012). MK-2206 treatment markedly reduced AKT-pS473 levels, and partially reduced the levels of pmTOR (Fig. 8B). Direct mTOR inhibition with rapamycin, at 1 nM and 10 nM, led to reduction of pmTOR as well as AKT-pS473 (Fig. 8B), the latter an expected effect of prolonged rapamycin treatment causing inhibition of mTORC1 and mTORC2 complex activity (Ballou and Lin, 2008; Sarbassov et al., 2006; Weichhart et al., 2015). Replating of untreated versus inhibitor-treated organoids showed an inhibitor dose-dependent partial (AKT inhibitor) or complete (mTOR inhibitor) rescue of secondary organoid formation by Cbl/Cbl-b DKO cells as compared with the untreated control (Fig. 8C,D). These results support the conclusion that hyperactivation of mTOR signaling downstream of RTKs, in part due to hyperactivity of AKT, promotes the depletion of MaSCs in Cbl/Cbl-b DKO mammary epithelium.

CBL family ubiquitin ligases target active pools of PTKs for ubiquitylation and degradation, thus serving as activation-dependent negative-feedback regulators of cellular signaling. Although the role of CBL-C is unclear, mouse knockouts have revealed key physiological roles of CBL or CBL-B, individually as well as in combination, especially as negative regulators of immune responses and hematopoiesis (Duan et al., 2004; Mohapatra et al., 2013; Thien and Langdon, 2005). Relatively little is known about the role(s) of CBL proteins in mammalian epithelia despite their broad epithelial expression and apparent redundancy. In this study we used three complementary conditional mouse gene deletion models to reveal a novel and crucial requirement for CBL and CBL-B in the maintenance of MaSCs. Multiple lines of evidence support our conclusion.

Mammary epithelium-selective Cbl/Cbl-b DKO via floxed Cbl deletion with MMTV-Cre (Wagner et al., 1997) on a whole-body Cbl-b null background led to a significant block in MG development (Fig. 1A). Transplantation of MMTV-Cre DKO MECs demonstrated that the MG developmental block was not due to preclinical MPD driven by MMTV-Cre-induced DKO HSCs (An et al., 2015; Naramura et al., 2010) (Fig. S3B). No defect in MG development was seen in Cbl null or Cbl-b null mice, indicating that CBL and CBL-B play a redundant, cell-autonomous regulatory role in the mammary epithelium. Redundancy between CBL and CBL-B and a compensatory increase in the expression of family members, as seen in Cbl-b null MGs (Fig. 1E), is likely to account for the lack of MG phenotypes in MGs of single-KO mice. Increased mammary branching as previously reported in Cbl null mice (Murphy et al., 1998) was later shown to be due to a non-cell-autonomous increase in growth factor secretion by the stroma (Crowley et al., 2005).

Surface marker-based analyses revealed a significant reduction in the overall numbers of MECs and their basal and luminal subsets in DKO mice, with a more substantial reduction in the MaSC-containing basal population, leading to a relative increase in the proportion of luminal cells (Fig. 2A-D). Interestingly, CD61/CD29-based MEC subset analysis indicated an increase in luminal progenitors but not the mature luminal cell pool. Together with a smaller basal population, this finding is consistent with a more rapid transition of DKO MaSCs into luminal progenitors. We suspect that such cells are functionally impaired since there was not a proportionate increase in mature luminal cells (Fig. 2G,H). Importantly, in vitro and in vivo assays on basal MECs derived from the MMTV-Cre-driven DKO mice demonstrated impaired MaSC self-renewal and ability to regenerate a MG.

The requirement of CBL and CBL-B for the maintenance of MaSCs was independently confirmed by selectively inducing Cbl/Cbl-b DKO in Lgr5+ MECs, which have recently been established as bi-potent MaSCs with MG-regenerating ability (Barker et al., 2007, 2013). Tamoxifen-induced, MaSC-specific Cbl/Cbl-b DKO led to a rapid depletion of Lgr5+ cells (Fig. 4B), with the redistribution of a significant proportion of the remaining Lgr5+ MECs into luminal compartment. The reduction in MaSC numbers and their apparent transition into luminal cells appear to represent an abnormally accelerated differentiation of MaSCs at the expense of their self-renewal, as demonstrated by organoid studies (Fig. 5A,B,E,F,H, Fig. 6A,B). In vitro deletion of Cbl and Cbl-b in basal MECs from CreERT; Cblf/f; Cbl-bf/f mice also led to reduced organoid-forming ability, complementing results from the other models (Fig. 7B,C). Although our immunofluorescence and qPCR analyses in multiple models support the idea of differentiation of MaSCs towards luminal lineages, mammary lineage tracing in Cblflox/Cbl-bflox mice bearing the dual reporter, possibly using K14-Cre (Van Keymeulen and Blanpain, 2012; Visvader and Stingl, 2014), will be needed to definitively test this idea.

Since negative regulation of PTKs is the primary biochemical function of CBL family proteins (Mohapatra et al., 2013), we surmised that dysregulated signaling downstream of PTK-coupled receptors in MaSCs was responsible for the rapid MaSC depletion upon Cbl and Cbl-b deletion. As multiple potential CBL/CBL-B target RTKs, including ErbB receptors, c-MET, IGF1/2 receptors and FGF receptors, are known to regulate MG development (Gjorevski and Nelson, 2011; Hynes and Watson, 2010; Sternlicht, 2006), we focused on AKT-mTOR signaling because it is a pivotal signaling axis downstream of all RTKs and non-receptor PTKs (Sun and Bernards, 2014), and its hyperactivity is associated with stem cell exhaustion in hematopoietic, neural and epidermal stem cells (Iglesias-Bartolome et al., 2012; Kalaitzidis et al., 2012; Magri et al., 2011). Indeed, inducible CreERT-mediated Cbl/Cbl-b DKO demonstrated an upregulation of pAKT and pmTOR in addition to the expected increase in pEGFR and EGFR levels, as organoids are cultured in EGF (Fig. 8A). CBL/CBL-B proteins are well-established negative regulators of RTKs. We chose EGFR as an index RTK since the organoids are grown in the presence of EGF and FGF2. As the use of an EGFR inhibitor (or lack of addition of EGF in the culture medium) would make the organoid assay unfeasible, we focused on downstream signaling pathways that are shared between RTKs activated by the added ligands as well as those that might be produced by MaSCs themselves. Our results raised the possibility that hyperactive mTOR signaling might commit Cbl/Cbl-b DKO MaSCs to premature differentiation, thereby reducing their self-renewing pool. A chemical inhibitor strategy supported such a role of hyperactive AKT-mTOR signaling. However, whether hyperactive mTOR is sufficient for MaSC depletion needs to be directly tested; our attempts to test this by expressing constitutively active or dominant-negative RHEB (Nie et al., 2010) were unsuccessful due to technical difficulties in lentiviral infection of primary organoids. The partial rescue of MaSC self-renewal by AKT inhibition versus an essentially complete rescue by mTOR inhibition (Fig. 8B-D) supports a model in which the hyperactivity of AKT and other upstream signaling pathways increases mTOR signaling to deplete MaSCs by promoting their rapid differentiation. Among the upstream pathways that might add to increased mTOR activity, ERK is a likely candidate as it is hyperphosphorylated when Cbl and Cbl-b are genetically deleted in HSCs (An et al., 2015) or depleted by shRNA in human MECs (Duan et al., 2011). Culture of human mammary organoids with amphiregulin has been found to maintain MECs in a less differentiated state, whereas growth in EGF led to myoepithelial differentiation; this dichotomy was shown to be related to sustained ERK/RSK signaling by EGF (Pasic et al., 2011).

Although our results support the likelihood that hyperactive mTOR results in rapid differentiation and loss of MaSCs, it is possible that the increased differentiation is a consequence rather than a cause of MaSC depletion. In this context, CBL is unequally distributed in daughter cells of asymmetrically dividing neural stem cells, with higher CBL correlating with lower EGFR levels in daughter cells that retain stemness (Ferron et al., 2010). Future studies to investigate a potential role of CBL and CBL-B in promoting asymmetric division in MaSCs will therefore be of interest.

Conclusions

Our studies provide the first evidence for a cell-intrinsic and redundant requirement of CBL and CBL-B in epithelial stem cell self-renewal and maintenance, provide a mechanistic basis for this requirement and suggest that fine-tuning of mTOR signaling is crucial for MaSC maintenance. These insights, together with models generated here, should help uncover an unanticipated positive role of CBL family proteins in epithelial stem cell maintenance, with implications for epithelial stem cell biology and cancers.

Antibodies

The following antibodies were used for WB: mouse monoclonal antibodies to CBL (clone 17/c-Cbl, BD Biosciences, 610442; 1:1000), HSC70 (clone B6, Santa Cruz, SC-7298; 1:10,000), cytokeratin 8 (Troma-1, Developmental Studies Hybridoma Bank; 1:200) and Ki67 (Alexa 488 conjugated, BD Biosciences, B56 clone, 558616; 1:200); rabbit monoclonal antibodies to CBL-B [clone D3C12, Cell Signaling Technology (CST), 9498; 1:1000], phospho-AKT (Ser473, CST, 4060; 1:500), pan-AKT (CST, 4691; 1:1000), phospho-mTOR (Ser2448, CST, 5536; 1:1000), mTOR (7C10, CST, 2983; 1:1000) and cyclin D1 (SP4, Abcam, ab16663; 1:200) and rabbit polyclonal antibodies to cytokeratin 5 (Covance, PRB-106P; 1:500), cytokeratin 14 (Covance, PRB-155P; 1:200), cleaved caspase 3 (CST, 9662; 1:200), EGFR (sc-03, Santa Cruz, 1005; 1:500) and phospho-EGF receptor (Tyr1068, CST, 2234; 1:1000). Anti-CD24-PE (clone M1/69, BD Biosciences, 553262; 1:50), anti-EpCAM-PE (eBioscience, 12-5791-82; 1:50), anti-CD29-APC (clone eBioHMb1-1, eBiosciences, 17-0291-82; 1:50) and anti-CD61-BV421 (clone 2C9.G2, BD Horizon, 562917; 1:50) were used for flow cytometry. Biotinylated anti-CD45 (clone 30-F11, BD Biosciences, 553077; 1:50), anti-Ter119 (clone ter119, BD Biosciences, 553672, 1:50) and anti-CD31 (clone MEC 13.3, BD Biosciences, 553371; 1:50) antibodies were used for AUTOMACS depletion of Lin+ cells.

Media, reagents and chemicals

All reagents and media used for primary mammary epithelial isolation and in vitro organoid and mammosphere cultures were from Stem Cell Technologies, unless otherwise specified. ROCK inhibitor was from Tocris Bioscience. Tamoxifen, 4-OH-T, recombinant EGF, FGF and rapamycin (R0395) were from Sigma. MK-2206 2HCl was from Selleckchem (S1078). Reduced growth factor Matrigel was from BD Biosciences.

Mouse strains

Mice carrying MMTV-Cre [stock Tg(MMTV-Cre)4 Mam/J] for conditional floxed Cbl deletion (Cbltm2Hua/Cblf/f) (Naramura et al., 2010) on a Cbl-b null (Cbl-btm1Hua) background (Cbl/Cbl-b DKO) (all genotypes are listed in Table S1) as well as Cbl KO (Cbltm1Hua) (Naramura et al., 1998) and Cbl-b KO (Cbl-btm1Hua) (Chiang et al., 2000) mice have been described previously. ROSA26 locus lacZ reporter [B6.129S4-Gt(ROSA)26Sortm1Sor/J] and EGFP reporter (CAG-CAT-EGFP) (Naramura et al., 2010) were incorporated to track cells with Cre-dependent target gene deletion. To generate a tamoxifen-inducible MaSC Cbl/Cbl-b DKO model, the Lgr5-GFP-IRES-CreERT allele (The Jackson Laboratory) was incorporated into Cblf/f; Cbl-b−/−; R26R-lacZ mice. Control mice were littermates obtained during breeding. We engineered a Cbl-bf/f mouse model and used it to generate a Cblf/f; Cbl-bf/f; CreERT; mT/mG strain that was used for in vitro Cbl and Cbl-b deletion in MaSCs in certain experiments (Goetz et al., 2016). Three-week-old female NSG mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) (The Jackson Laboratory) were used as recipients for mammary transplantation. All mouse strains were maintained on a C57BL/6J background under specific pathogen-free conditions and genotyped using tail DNA PCR (sequences are provided in Table S2). All experimental analyses were carried out on female mice. The Institutional Animal Care and Use Committee of University of Nebraska Medical Center approved all mouse experiments.

MG harvest and whole-mount staining

Fourth (inguinal) MGs were excised and processed according to a standard protocol (LaMarca and Rosen, 2007). Whole-mounts were photographed and various parameters (branch points, ductal length and fat pad filling) were quantified using Nikon Elements software. Whole MG X-gal staining, to visualize lacZ-encoded β-galactosidase activity as a reporter of gene deletion, was performed as described (LaMarca and Rosen, 2007). At least four animals were analyzed per genotype and time point.

Immunofluorescence analyses and western blotting (WB)

Staining of paraffin-embedded MG sections and mammary organoids with fluorescently labeled antibodies was carried out using standard protocols as previously described (Pasic et al., 2011). WB of freshly isolated MG and organoids was performed as described in the supplementary Materials and Methods.

Immunohistochemistry (IHC)

Formalin-fixed and paraffin-embedded tissue sections were stained with anti-CBL antibody using the Mouse on Mouse (MOM) and ABC Amplification IHC Kit (Vector Laboratories) as per the manufacturer's instructions. The slides were counterstained with Hematoxylin. In all cases, a species-matched (and isotype-matched for mouse monoclonal antibodies) control IgG was used as a negative control.

Transplantation assay

The mammary epithelial tissue between the nipple and lymph node of the fourth MG of 3-week-old immune-compromised NSG female mice was removed to yield epithelium-cleared fat pads (Deome et al., 1959). Complete removal of the mammary epithelium was verified by whole-mount staining. Recipient mice received a control mammary transplant on one side and a Cbl/Cbl-b DKO transplant on the contralateral side. The transplants were harvested from virgin hosts 8 weeks post-transplantation. Alternatively, the recipients were bred with fertile males and the glands harvested at various points of pregnancy. The whole-mounts of transplants were stained with carmine alum or X-gal.

MG repopulating activity

To test MG repopulating activity, 50,000 Lin single MECs isolated from 6-week-old DKO or control MGs were suspended in 100 μl 1:1 mix of Matrigel and Complete Mouse Epicult-B medium (STEMCELL Technologies, 05610) and then injected into epithelium-cleared mammary fat pads of 3-week-old NSG mice. In other experiments, 5000 Lin/EpCAM+ cells were directly sorted into Epicult-B medium, centrifuged (400 g for 5 min at 4°C) and resuspended in 50 μl 1:1 Epicult-B medium and Matrigel, and injected into epithelium-cleared fat pads. Mammary fat pads were harvested 8 weeks after transplant and stained with carmine alum to assess the extent of mammary outgrowths.

Isolation of primary mouse MECs and flow cytometry

Single cells were prepared from mammary tissue as previously described (Stingl et al., 2006a,b). Single-cell suspensions were stained with a combination of biotinylated anti-CD45, anti-Ter119 and anti-CD31 antibodies followed by anti-biotin microbeads (Miltenyi Biotech) to magnetically deplete the Lin+ cells using AUTOMACS (Miltenyi Biotech). Lin cells were co-stained with the indicated combinations of anti-CD24-PE, anti-EpCAM-PE, anti-CD29-APC and anti-CD61-BV421. FACS analysis and cell sorting were performed on a FACSAria II (BD Biosciences) and the results analyzed using FlowJo software.

In vivo Lgr5-Cre-mediated gene deletion

To induce Lgr5-CreERT-mediated floxed Cbl deletion in vivo, 4-week-old mice received three consecutive daily intraperitoneal injections of tamoxifen (1 mg/20 g body weight) in corn oil (10 mg/ml stock). Mice were analyzed at day 5 or 10 as indicated.

In vitro mammary epithelial organoid culture assay for MaSC renewal

In vitro mammary epithelial organoid culture was performed as described (Guo et al., 2012) with minor modifications. FACS sorting of single MECs was used to prepare the GFP+ fraction from control or DKO mice carrying the Lgr5-GFP-CreERT allele, or the RFP+ basal cell fraction from Cblf/f; Cbl-bf/f; CreERT; mT/mG or Cblf/f; Cbl-bf/+; CreERT; mT/mG (control) mice. Cells were seeded at 2000-5000 per well in 100% Matrigel in 8-well Lab-Tek plates (Nunc) precoated with Matrigel and fed with complete Epicult-B medium and 5 μM Y-27632 (ROCK inhibitor; TOCRIS, 1254). The medium was replaced every 2 days. Organoids (>100 μm in diameter) were imaged and counted 7-14 days after seeding. For immunofluorescence analyses, organoid cultures were fixed with 4% paraformaldehyde, permeabilized with 0.5% Triton X-100 in PBS and processed as described previously (Ewald et al., 2008).

Mammosphere assay of MaSC renewal

In vitro mammosphere assay of MaSC renewal was performed as described (Guo et al., 2012). GFP+ basal (CD29hi EpCAMlow) and luminal (CD29low EpCAMhi) cell populations from DKO and control mice carrying the MMTV-Cre-GFP allele or the GFP+ fraction of single MECs from control or DKO mice carrying the Lgr5-GFP-CreERT allele, or the RFP+ basal cells sorted from Cblf/f; Cbl-bf/f; CreERT; mT/mG and Cblf/f; Cbl-bf/+; CreERT; mT/mG (control) were prepared by FACS sorting and seeded in complete Epicult-B medium containing 5% Matrigel, as above, at 1000-2000 per well in 96-well ultra low-attachment plates (Corning). Mammospheres were imaged and counted (>100 μm in diameter) 7-10 days after seeding. For serial passaging, mammospheres were harvested using 5 mg/ml dispase I (Stem Cell Technologies) and dissociated in 0.25% trypsin-EDTA (Gibco). Single cells were seeded (2000 per well) for 7-10 days followed by imaging, counting, and further passaging of mammospheres.

In vitro Lgr5-Cre-mediated Cbl and Cbl-b deletion

Sorted GFP+ MECs from Cblfl/+; Cbl-b+/−; Lgr5-CreERT (control) or Cblfl/fl; Cbl-b−/−; Lgr5-CreERT (DKO) mice were plated in 100% Matrigel in Matrigel-coated 8-well chambers and cultured in Epicult-B medium with ROCK inhibitor for 7 days (Guo et al., 2012). The medium was replaced every 2 days. For in vitro Cre activation, 400 nM 4-OH-T in Epicult-B medium was added for 96 h beginning at day 3, replaced with regular medium for 1 day, and organoids harvested. 2000 cells were then replated per well in 96-well ultra low-attachment plates to assess mammosphere-forming efficiency.

In vitro Rosa26-CreERT induction and inhibitor treatment

RFP+ basal and luminal cells were isolated from Cblfl/f; Cbl-bf/f; CreERT; mT/mG and Cblfl/f; Cbl-bf/+; CreERT; mT/mG mice and plated in 96-well ultra low-attachment plates for 7 days as described above. 400 nM 4-OH-T was added in Epicult-B medium for 96 h. GFP expression in organoids indicates induction of Cre and deletion of floxed genes. WBs were performed to test the signaling events and CBL and CBL-B deletion. Basal organoids were treated with MK-2206 (50 nM, 100 nM) or rapamycin (1 nM, 10 nM) with and without 4-OH-T induction for 4 days. Single cells were replated, and organoids imaged and quantified. Non-induced cultures were treated with inhibitors alone to control for any non-specific effects. DMSO was used as a vehicle control.

Real-time quantitative PCR (qPCR)

FACS-sorted cells were directly collected in Trizol reagent (Ambion, Life Technologies) for RNA extraction. Detailed procedures are provided in the supplementary Materials and Methods and qPCR primers are listed in Table S3.

Statistical analyses

Quantified results of whole-mounts, qPCR, IHC and flow cytometry were compared between groups using Student's t-test and are presented as mean±s.e.m., with P≤0.05 deemed significant. Statistical analysis and graphical representation of data were performed using Prism 4.0c (GraphPad).

We thank the H.B. laboratory members for helpful discussions, Kay-Uwe Wagner and Aleata Triplett for advice on mammary transplants, the staff of UNMC Cores for assistance and Dr Andrew Ewald (Johns Hopkins School of Medicine) for sharing his protocol for mammary organoid staining.

Author contributions

B.M., N.Z.: conceptualization and design, collection and/or assembly of data, data analysis and interpretation, manuscript writing; W.A., B.G., P.A., T.A.B., I.M., M.D.S.: collection and assembly of data, data analysis and interpretation, manuscript editing; J.L.M.: data analysis and interpretation; V.B.: conceptualization and design, manuscript editing; H.B.: conception and design, data analysis and interpretation, manuscript writing.

Funding

This work was supported by the National Institutes of Health (CA105489, CA87986, CA99163 to H.B.; CA96844, CA144027 to V.B.); the U.S. Department of Defense (W81WH-11-1-0167 to H.B.; W81XWH-07-1-0351, W81XWH-11-1-0171 to V.B.); the Nebraska Department of Health and Human Services LB606 (18123-Y3 to H.B.); an Institutional Development Award (IDeA) from the National Institute of General Medical Sciences (P30 GM106397); and a National Cancer Institute Core Support Grant (P30CA036727) to the Fred & Pamela Buffett Cancer Center and Nebraska Research Initiative. We thank the Dr Raphael Bonita Fund for partial support of graduate students W.A. and B.G. in the H.B. lab. B.M., N.Z., W.A. and P.A. received University of Nebraska Medical Center graduate assistantships. B.G. was a trainee under the National Cancer Institute Cancer Biology Training Grant. Deposited in PMC for release after 12 months.

An
,
W.
,
Nadeau
,
S. A.
,
Mohapatra
,
B. C.
,
Feng
,
D.
,
Zutshi
,
N.
,
Storck
,
M. D.
,
Arya
,
P.
,
Talmadge
,
J. E.
,
Meza
,
J. L.
,
Band
,
V.
, et al. 
(
2015
).
Loss of Cbl and Cbl-B ubiquitin ligases abrogates hematopoietic stem cell quiescence and sensitizes leukemic disease to chemotherapy
.
Oncotarget
6
,
10498
-
10509
.
Ballou
,
L. M.
and
Lin
,
R. Z.
(
2008
).
Rapamycin and mTOR kinase inhibitors
.
J. Chem. Biol.
1
,
27
-
36
.
Barker
,
N.
,
van Es
,
J. H.
,
Kuipers
,
J.
,
Kujala
,
P.
,
van den Born
,
M.
,
Cozijnsen
,
M.
,
Haegebarth
,
A.
,
Korving
,
J.
,
Begthel
,
H.
,
Peters
,
P. J.
, et al. 
(
2007
).
Identification of stem cells in small intestine and colon by marker gene Lgr5
.
Nature
449
,
1003
-
1007
.
Barker
,
N.
,
Tan
,
S.
and
Clevers
,
H.
(
2013
).
Lgr proteins in epithelial stem cell biology
.
Development
140
,
2484
-
2494
.
Beck
,
B.
and
Blanpain
,
C.
(
2013
).
Unravelling cancer stem cell potential
.
Nat. Rev. Cancer
13
,
727
-
738
.
Brisken
,
C.
,
Heineman
,
A.
,
Chavarria
,
T.
,
Elenbaas
,
B.
,
Tan
,
J.
,
Dey
,
S. K.
,
McMahon
,
J. A.
,
McMahon
,
A. P.
and
Weinberg
,
R. A.
(
2000
).
Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling
.
Genes Dev.
14
,
650
-
654
.
Cai
,
C.
,
Yu
,
Q. C.
,
Jiang
,
W.
,
Liu
,
W.
,
Song
,
W.
,
Yu
,
H.
,
Zhang
,
L.
,
Yang
,
Y.
and
Zeng
,
Y. A.
(
2014
).
R-Spondin1 is a novel hormone mediator for mammary stem cell self-renewal
.
Genes Dev.
28
,
2205
-
2218
.
Carr
,
J. R.
,
Kiefer
,
M. M.
,
Park
,
H. J.
,
Li
,
J.
,
Wang
,
Z.
,
Fontanarosa
,
J.
,
DeWaal
,
D.
,
Kopanja
,
D.
,
Benevolenskaya
,
E. V.
,
Guzman
,
G.
, et al. 
(
2012
).
FoxM1 regulates mammary luminal cell fate
.
Cell Rep.
1
,
715
-
729
.
Chiang
,
Y. J.
,
Kole
,
H. K.
,
Brown
,
K.
,
Naramura
,
M.
,
Fukuhara
,
S.
,
Hu
,
R. J.
,
Jang
,
I. K.
,
Gutkind
,
J. S.
,
Shevach
,
E.
and
Gu
,
H
. (
2000
).
Cbl-B regulates the CD28 dependence of T-cell activation
.
Nature
403
,
216
-
220
.
Crowley
,
M. R.
,
Bowtell
,
D.
and
Serra
,
R.
(
2005
).
TGF-beta, c-Cbl, and PDGFR-alpha the in mammary stroma
.
Dev. Biol.
279
,
58
-
72
.
de Visser
,
K. E.
,
Ciampricotti
,
M.
,
Michalak
,
E. M.
,
Tan
,
D. W.
,
Speksnijder
,
E. N.
,
Hau
,
C. S.
,
Clevers
,
H.
,
Barker
,
N.
and
Jonkers
,
J.
(
2012
).
Developmental stage-specific contribution of LGR5(+)cells to basal and luminal epithelial lineages in the postnatal mammary gland
.
J. Pathol.
228
,
300
-
309
.
Deome
,
K. B.
,
Faulkin
,
L. J.
, Jr
,
Bern
,
H. A.
and
Blair
,
P. B.
(
1959
).
Development of mammary tumors from hyperplastic alveolar nodules transplanted into gland-free mammary fat pads of female C3H mice
.
Cancer Res.
19
,
515
-
520
.
Dontu
,
G.
,
Abdallah
,
W. M.
,
Foley
,
J. M.
,
Jackson
,
K. W.
,
Clarke
,
M. F.
,
Kawamura
,
M. J.
and
Wicha
,
M. S.
(
2003
).
In Vitro Propagation and transcriptional profiling of human mammary stem/progenitor cells
.
Genes Dev.
17
,
1253
-
1270
.
Duan
,
L.
,
Reddi
,
A. L.
,
Ghosh
,
A.
,
Dimri
,
M.
and
Band
,
H.
(
2004
).
The Cbl family and other ubiquitin ligases: destructive forces in control of antigen receptor signaling
.
Immunity
21
,
7
-
17
.
Duan
,
L.
,
Raja
,
S. M.
,
Chen
,
G.
,
Virmani
,
S.
,
Williams
,
S. H.
,
Clubb
,
R. J.
,
Mukhopadhyay
,
C.
,
Rainey
,
M. A.
,
Ying
,
G.
,
Dimri
,
M.
, et al. 
(
2011
).
Negative regulation of EGFR-Vav2 signaling axis by Cbl ubiquitin ligase controls EGF receptor-mediated epithelial cell adherens junction dynamics and cell migration
.
J. Biol. Chem.
286
,
620
-
633
.
Ewald
,
A. J.
,
Brenot
,
A.
,
Duong
,
M.
,
Chan
,
B. S.
and
Werb
,
Z.
(
2008
).
Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis
.
Dev. Cell
14
,
570
-
581
.
Fang
,
D.
and
Liu
,
Y.-C.
(
2001
).
Proteolysis-independent regulation of PI3K by Cbl-B-mediated ubiquitination in T cells
.
Nat. Immunol.
2
,
870
-
875
.
Ferron
,
S. R.
,
Pozo
,
N.
,
Laguna
,
A.
,
Aranda
,
S.
,
Porlan
,
E.
,
Moreno
,
M.
,
Fillat
,
C.
,
de la Luna
,
S.
,
Sánchez
,
P.
,
Arbones
,
M. L.
, et al. 
(
2010
).
Regulated segregation of kinase Dyrk1A during asymmetric neural stem cell division is critical for EGFR-mediated biased signaling
.
Cell Stem Cell
7
,
367
-
379
.
Gjorevski
,
N.
and
Nelson
,
C. M.
(
2011
).
Integrated morphodynamic signalling of the mammary gland
.
Nat. Rev. Mol. Cell Biol.
12
,
581
-
593
.
Goetz
,
B.
,
An
,
W.
,
Mohapatra
,
B.
,
Zutshi
,
N.
,
Iseka
,
F.
,
Storck
,
M. D.
,
Meza
,
J.
,
Sheinin
,
Y.
,
Band
,
V.
and
Band
,
H.
(
2016
).
A novel CBL-Bflox/flox mouse model allows tissue-selective fully conditional CBL/CBL-B double-knockout: CD4-Cre mediated CBL/CBL-B deletion occurs in both T-cells and hematopoietic stem cells
.
Oncotarget
7
,
51107
-
51123
.
Griffiths
,
E. K.
,
Sanchez
,
O.
,
Mill
,
P.
,
Krawczyk
,
C.
,
Hojilla
,
C. V.
,
Rubin
,
E.
,
Nau
,
M. M.
,
Khokha
,
R.
,
Lipkowitz
,
S.
,
Hui
,
C. C.
, et al. 
(
2003
).
Cbl-3-deficient mice exhibit normal epithelial development
.
Mol. Cell. Biol.
23
,
7708
-
7718
.
Guo
,
W.
,
Keckesova
,
Z.
,
Donaher
,
J. L.
,
Shibue
,
T.
,
Tischler
,
V.
,
Reinhardt
,
F.
,
Itzkovitz
,
S.
,
Noske
,
A.
,
Zürrer-Härdi
,
U.
,
Bell
,
G.
, et al. 
(
2012
).
Slug and Sox9 cooperatively determine the mammary stem cell state
.
Cell
148
,
1015
-
1028
.
Hirai
,
H.
,
Sootome
,
H.
,
Nakatsuru
,
Y.
,
Miyama
,
K.
,
Taguchi
,
S.
,
Tsujioka
,
K.
,
Ueno
,
Y.
,
Hatch
,
H.
,
Majumder
,
P. K.
,
Pan
,
B.-S.
, et al. 
(
2010
).
MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo
.
Mol. Cancer. Ther.
9
,
1956
-
1967
.
Humphreys
,
R. C.
,
Krajewska
,
M.
,
Krnacik
,
S.
,
Jaeger
,
R.
,
Weiher
,
H.
,
Krajewski
,
S.
,
Reed
,
J. C.
and
Rosen
,
J. M.
(
1996
).
Apoptosis in the terminal endbud of the murine mammary gland: a mechanism of ductal morphogenesis
.
Development
122
,
4013
-
4022
.
Hynes
,
N. E.
and
Watson
,
C. J.
(
2010
).
Mammary gland growth factors: roles in normal development and in cancer
.
Cold Spring Harb. Perspect. Biol.
2
,
a003186
.
Iglesias-Bartolome
,
R.
,
Patel
,
V.
,
Cotrim
,
A.
,
Leelahavanichkul
,
K.
,
Molinolo
,
A. A.
,
Mitchell
,
J. B.
and
Gutkind
,
J. S.
(
2012
).
mTOR inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis
.
Cell Stem Cell
11
,
401
-
414
.
Kalaitzidis
,
D.
,
Sykes
,
S. M.
,
Wang
,
Z.
,
Punt
,
N.
,
Tang
,
Y.
,
Ragu
,
C.
,
Sinha
,
A. U.
,
Lane
,
S. W.
,
Souza
,
A. L.
,
Clish
,
C. B.
, et al. 
(
2012
).
mTOR complex 1 plays critical roles in hematopoiesis and Pten-loss-evoked leukemogenesis
.
Cell Stem Cell
11
,
429
-
439
.
Kessenbrock
,
K.
,
Dijkgraaf
,
G. J. P.
,
Lawson
,
D. A.
,
Littlepage
,
L. E.
,
Shahi
,
P.
,
Pieper
,
U.
and
Werb
,
Z.
(
2013
).
A role for matrix metalloproteinases in regulating mammary stem cell function via the wnt signaling pathway
.
Cell Stem Cell
13
,
300
-
313
.
Kitaura
,
Y.
,
Jang
,
I. K.
,
Wang
,
Y.
,
Han
,
Y.-C.
,
Inazu
,
T.
,
Cadera
,
E. J.
,
Schlissel
,
M.
,
Hardy
,
R. R.
and
Gu
,
H.
(
2007
).
Control of the B cell-intrinsic tolerance programs by ubiquitin ligases Cbl and Cbl-b
.
Immunity
26
,
567
-
578
.
Korkaya
,
H.
,
Paulson
,
A.
,
Iovino
,
F.
and
Wicha
,
M. S.
(
2008
).
HER2 regulates the mammary stem/progenitor cell population driving tumorigenesis and invasion
.
Oncogene
27
,
6120
-
6130
.
LaMarca
,
H. L.
and
Rosen
,
J. M.
(
2007
).
Estrogen regulation of mammary gland development and breast cancer: amphiregulin takes center stage
.
Breast Cancer Res.
9
,
304
.
Lim
,
E.
,
Vaillant
,
F.
,
Wu
,
D.
,
Forrest
,
N. C.
,
Pal
,
B.
,
Hart
,
A. H.
,
Asselin-Labat
,
M.-L.
,
Gyorki
,
D. E.
,
Ward
,
T.
,
Partanen
,
A.
, et al. 
(
2009
).
Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers
.
Nat. Med.
15
,
907
-
913
.
Lim
,
E.
,
Wu
,
D.
,
Pal
,
B.
,
Bouras
,
T.
,
Asselin-Labat
,
M.-L.
,
Vaillant
,
F.
,
Yagita
,
H.
,
Lindeman
,
G. J.
,
Smyth
,
G. K.
and
Visvader
,
J. E.
(
2010
).
Transcriptome analyses of mouse and human mammary cell subpopulations reveal multiple conserved genes and pathways
.
Breast Cancer Res.
12
,
R21
.
Magri
,
L.
,
Cambiaghi
,
M.
,
Cominelli
,
M.
,
Alfaro-Cervello
,
C.
,
Cursi
,
M.
,
Pala
,
M.
,
Bulfone
,
A.
,
Garcìa-Verdugo
,
J. M.
,
Leocani
,
L.
,
Minicucci
,
F.
, et al. 
(
2011
).
Sustained activation of mTOR pathway in embryonic neural stem cells leads to development of tuberous sclerosis complex-associated lesions
.
Cell Stem Cell
9
,
447
-
462
.
McCaffrey
,
L. M.
and
Macara
,
I. G.
(
2009
).
The Par3/aPKC interaction is essential for end bud remodeling and progenitor differentiation during mammary gland morphogenesis
.
Genes Dev.
23
,
1450
-
1460
.
Mohapatra
,
B.
,
Ahmad
,
G.
,
Nadeau
,
S.
,
Zutshi
,
N.
,
An
,
W.
,
Scheffe
,
S.
,
Dong
,
L.
,
Feng
,
D.
,
Goetz
,
B.
,
Arya
,
P.
, et al. 
(
2013
).
Protein tyrosine kinase regulation by ubiquitination: critical roles of Cbl-family ubiquitin ligases
.
Biochim. Biophys. Acta
1833
,
122
-
139
.
Murphy
,
M. A.
,
Schnall
,
R. G.
,
Venter
,
D. J.
,
Barnett
,
L.
,
Bertoncello
,
I.
,
Thien
,
C. B.
,
Langdon
,
W. Y.
and
Bowtell
,
D. D. L.
(
1998
).
Tissue hyperplasia and enhanced T-cell signalling via ZAP-70 in C-Cbl-deficient mice
.
Mol. Cell. Biol.
18
,
4872
-
4882
.
Naramura
,
M.
,
Kole
,
H. K.
,
Hu
,
R. J.
and
Gu
,
H.
(
1998
).
Altered thymic positive selection and intracellular signals in Cbl-deficient mice
.
Proc. Natl. Acad. Sci. USA
95
,
15547
-
15552
. https://doi.org/10.1073/pnas.95.26.15547
Naramura
,
M.
,
Jang
,
I.-K.
,
Kole
,
H.
,
Huang
,
F.
,
Haines
,
D.
and
Gu
,
H.
(
2002
).
C-Cbl and Cbl-B regulate T cell responsiveness by promoting ligand-induced TCR down-modulation
.
Nat. Immunol.
3
,
1192
-
1199
.
Naramura
,
M.
,
Nandwani
,
N.
,
Gu
,
H.
,
Band
,
V.
and
Band
,
H.
(
2010
).
Rapidly fatal myeloproliferative disorders in mice with deletion of casitas B-cell lymphoma (Cbl) and Cbl-B in hematopoietic stem cells
.
Proc. Natl. Acad. Sci. USA
107
,
16274
-
16279
.
Nguyen
,
L. V.
,
Vanner
,
R.
,
Dirks
,
P.
and
Eaves
,
C. J.
(
2012
).
Cancer stem cells: an evolving concept
.
Nat. Rev. Cancer
12
,
133
-
143
. https://doi.org/10.1038/nrc3184
Nie
,
D.
,
Di Nardo
,
A.
,
Han
,
J. M.
,
Baharanyi
,
H.
,
Kramvis
,
I.
,
Huynh
,
T. T.
,
Dabora
,
S.
,
Codeluppi
,
S.
,
Pandolfi
,
P. P.
,
Pasquale
,
E. B.
, et al. 
(
2010
).
Tsc2-Rheb signaling regulates EphA-mediated axon guidance
.
Nat. Neurosci.
13
,
163
-
172
.
Pasic
,
L.
,
Eisinger-Mathason
,
T. S. K.
,
Velayudhan
,
B. T.
,
Moskaluk
,
C. A.
,
Brenin
,
D. R.
,
Macara
,
I. G.
and
Lannigan
,
D. A.
(
2011
).
Sustained activation of the HER1-ERK1/2-RSK signaling pathway controls myoepithelial cell fate in human mammary tissue
.
Genes Dev.
25
,
1641
-
1653
.
Pece
,
S.
,
Tosoni
,
D.
,
Confalonieri
,
S.
,
Mazzarol
,
G.
,
Vecchi
,
M.
,
Ronzoni
,
S.
,
Bernard
,
L.
,
Viale
,
G.
,
Pelicci
,
P. G.
and
Di Fiore
,
P. P.
(
2010
).
Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content
.
Cell
140
,
62
-
73
.
Plaks
,
V.
,
Brenot
,
A.
,
Lawson
,
D. A.
,
Linnemann
,
J. R.
,
Van Kappel
,
E. C.
,
Wong
,
K. C.
,
de Sauvage
,
F.
,
Klein
,
O. D.
and
Werb
,
Z.
(
2013
).
Lgr5-expressing cells are sufficient and necessary for postnatal mammary gland organogenesis
.
Cell Rep.
3
,
70
-
78
.
Pond
,
A. C.
,
Bin
,
X.
,
Batts
,
T.
,
Roarty
,
K.
,
Hilsenbeck
,
S.
and
Rosen
,
J. M.
(
2013
).
Fibroblast growth factor receptor signaling is essential for normal mammary gland development and stem cell function
.
Stem Cells
31
,
178
-
189
.
Rios
,
A. C.
,
Fu
,
N. Y.
,
Lindeman
,
G. J.
and
Visvader
,
J. E.
(
2014
).
In situ identification of bipotent stem cells in the mammary gland
.
Nature
506
,
322
-
327
.
Robinson
,
G. W.
,
Wagner
,
K. U.
and
Hennighausen
,
L
. (
2001
).
Functional mammary gland development and oncogene-induced tumor formation are not affected by the absence of the retinoblastoma gene
.
Oncogene
20
,
7115
-
7119
.
Sangai
,
T.
,
Akcakanat
,
A.
,
Chen
,
H.
,
Tarco
,
E.
,
Wu
,
Y.
,
Do
,
K.-A.
,
Miller
,
T. W.
,
Arteaga
,
C. L.
,
Mills
,
G. B.
,
Gonzalez-Angulo
,
A. M.
, et al. 
(
2012
).
Biomarkers of response to Akt inhibitor MK-2206 in breast cancer
.
Clin. Cancer Res.
18
,
5816
-
5828
.
Sarbassov
,
D. D.
,
Ali
,
S. M.
,
Sengupta
,
S.
,
Sheen
,
J.-H.
,
Hsu
,
P. P.
,
Bagley
,
A. F.
,
Markhard
,
A. L.
and
Sabatini
,
D. M.
(
2006
).
Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB
.
Mol. Cell
22
,
159
-
168
.
Shackleton
,
M.
,
Vaillant
,
F.
,
Simpson
,
K. J.
,
Stingl
,
J.
,
Smyth
,
G. K.
,
Asselin-Labat
,
M.-L.
,
Wu
,
L.
,
Lindeman
,
G. J.
and
Visvader
,
J. E.
(
2006
).
Generation of a functional mammary gland from a single stem cell
.
Nature
439
,
84
-
88
.
Shehata
,
M.
,
Teschendorff
,
A.
,
Sharp
,
G.
,
Novcic
,
N.
,
Russell
,
I. A.
,
Avril
,
S.
,
Prater
,
M.
,
Eirew
,
P.
,
Caldas
,
C.
,
Watson
,
C. J.
, et al. 
(
2012
).
Phenotypic and functional characterisation of the luminal cell hierarchy of the mammary gland
.
Breast Cancer Res.
14
,
R134
.
Siegel
,
P. M.
and
Muller
,
W. J.
(
2010
).
Transcription factor regulatory networks in mammary epithelial development and tumorigenesis
.
Oncogene
29
,
2753
-
2759
.
Smalley
,
M. J.
,
Kendrick
,
H.
,
Sheridan
,
J. M.
,
Regan
,
J. L.
,
Prater
,
M. D.
,
Lindeman
,
G. J.
,
Watson
,
C. J.
,
Visvader
,
J. E.
and
Stingl
,
J.
(
2012
).
Isolation of mouse mammary epithelial subpopulations: a comparison of leading methods
.
J. Mammary Gland Biol. Neoplasia
17
,
91
-
97
.
Soady
,
K. J.
,
Kendrick
,
H.
,
Gao
,
Q.
,
Tutt
,
A.
,
Zvelebil
,
M.
,
Ordonez
,
L. D.
,
Quist
,
J.
,
Tan
,
D. W.
,
Isacke
,
C. M.
,
Grigoriadis
,
A.
, et al. 
(
2015
).
Mouse mammary stem cells express prognostic markers for triple-negative breast cancer
.
Breast Cancer Res.
17
,
31
.
Sternlicht
,
M. D.
(
2006
).
Key stages in mammary gland development: the cues that regulate ductal branching morphogenesis
.
Breast Cancer Res.
8
,
201
.
Stingl
,
J.
,
Eirew
,
P.
,
Ricketson
,
I.
,
Shackleton
,
M.
,
Vaillant
,
F.
,
Choi
,
D.
,
Li
,
H. I.
and
Eaves
,
C. J.
(
2006a
).
Purification and unique properties of mammary epithelial stem cells
.
Nature
439
,
993
-
997
. https://doi.org/10.1038/nature04496
Stingl
,
J.
,
Raouf
,
A.
,
Eirew
,
P.
and
Eaves
,
C. J.
(
2006b
).
Deciphering the mammary epithelial cell hierarchy
.
Cell Cycle
5
,
1519
-
1522
.
Sun
,
C.
and
Bernards
,
R.
(
2014
).
Feedback and redundancy in receptor tyrosine kinase signaling: relevance to cancer therapies
.
Trends Biochem. Sci.
39
,
465
-
474
.
Thien
,
C. B. F.
and
Langdon
,
W. Y.
(
2005
).
c-Cbl and Cbl-b ubiquitin ligases: substrate diversity and the negative regulation of signalling responses
.
Biochem. J.
391
,
153
-
166
.
van Amerongen
,
R.
,
Bowman
,
A. N.
and
Nusse
,
R.
(
2012
).
Developmental stage and time dictate the fate of Wnt/beta-catenin-responsive stem cells in the mammary gland
.
Cell Stem Cell
11
,
387
-
400
.
Van Keymeulen
,,
A.
and
Blanpain
,
C.
(
2012
).
Tracing epithelial stem cells during development, homeostasis, and repair
.
J. Cell Biol.
197
,
575
-
584
.
Visvader
,
J. E.
(
2009
).
Keeping abreast of the mammary epithelial hierarchy and breast tumorigenesis
.
Genes Dev.
23
,
2563
-
2577
.
Visvader
,
J. E.
and
Stingl
,
J.
(
2014
).
Mammary stem cells and the differentiation hierarchy: current status and perspectives
.
Genes Dev.
28
,
1143
-
1158
.
Wagner
,
K.-U.
,
Wall
,
R. J.
,
St-Onge
,
L.
,
Gruss
,
P.
,
Wynshaw-Boris
,
A.
,
Garrett
,
L.
,
Li
,
M.
,
Furth
,
P. A.
and
Hennighausen
,
L.
(
1997
).
Cre-mediated gene deletion in the mammary gland
.
Nucleic Acids Res.
25
,
4323
-
4330
.
Wang
,
Y.
,
Dong
,
J.
,
Li
,
D.
,
Lai
,
L.
,
Siwko
,
S.
,
Li
,
Y.
and
Liu
,
M.
(
2013
).
Lgr4 regulates mammary gland development and stem cell activity through the pluripotency transcription factor Sox2
.
Stem Cells
31
,
1921
-
1931
.
Weichhart
,
T.
,
Hengstschlager
,
M.
and
Linke
,
M.
(
2015
).
Regulation of innate immune cell function by mTOR
.
Nat. Rev. Immunol.
15
,
599
-
614
.
Yamaji
,
D.
,
Na
,
R.
,
Feuermann
,
Y.
,
Pechhold
,
S.
,
Chen
,
W.
,
Robinson
,
G. W.
and
Hennighausen
,
L.
(
2009
).
Development of mammary luminal progenitor cells is controlled by the transcription factor STAT5A
.
Genes Dev.
23
,
2382
-
2387
.
Yap
,
T. A.
,
Yan
,
L.
,
Patnaik
,
A.
,
Fearen
,
I.
,
Olmos
,
D.
,
Papadopoulos
,
K.
,
Baird
,
R. D.
,
Delgado
,
L.
,
Taylor
,
A.
,
Lupinacci
,
L.
, et al. 
(
2011
).
First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors
.
J. Clin. Oncol.
29
,
4688
-
4695
.
Zeng
,
Y. A.
and
Nusse
,
R.
(
2010
).
Wnt proteins are self-renewal factors for mammary stem cells and promote their long-term expansion in culture
.
Cell Stem Cell
6
,
568
-
577
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information