Cell context is key for cell state. Using physiologically relevant models of laminin-rich extracellular matrix (lrECM) induction of mammary epithelial cell quiescence and differentiation, we provide a landscape of the key molecules for the proliferation–quiescence decision, identifying multiple layers of regulation at the mRNA and protein levels. Quiescence occurred despite activity of Fak (also known as PTK2), Src and phosphoinositide 3-kinases (PI3Ks), suggesting the existence of a disconnecting node between upstream and downstream proliferative signalling. Pten, a lipid and protein phosphatase, fulfils this role, because its inhibition increased proliferation and restored signalling via the Akt, mTORC1, mTORC2 and mitogen-activated protein kinase (MAPK) pathways. Pten and laminin levels were positively correlated in developing murine mammary epithelia, and Pten localized apicolaterally in luminal cells in ducts and near the nascent lumen in terminal end buds. Consistently, in three-dimensional acinogenesis models, Pten was required for triggering and sustaining quiescence, polarity and architecture. The multilayered regulatory circuitry that we uncovered provides an explanation for the robustness of quiescence within a growth-suppressive microenvironment, which could nonetheless be disrupted by perturbations in master regulators such as Pten.

The mammary gland is unparalleled with regard to development and differentiation. A rudimentary epithelial tree at birth, the mammary gland further branches in puberty and will only fully grow and differentiate during pregnancy and lactation. Upon weaning, the mammary gland goes through involution and massive remodelling, returning to a histological architecture similar to the one found before pregnancy (Sternlicht et al., 2006). This cycle repeats as many times as pregnancy, lactation and weaning occur in a female mammal lifetime (Inman et al., 2015; Sternlicht et al., 2006). All these events are coordinated by local and systemic stimuli (Sternlicht et al., 2006). Therefore, the developing mammary gland offers an invaluable tool for the study of spatiotemporal control of cell behaviour by the cell microenvironment.

As a complex and widely diverse mesh of proteins and soluble factors, the extracellular matrix (ECM) not only confers physical support for cells, but also provides biochemical and mechanical cues perceived by the cells (Nelson and Bissell, 2006; Sternlicht et al., 2006), which can respond accordingly by altering cell phenotype and, in turn, remodelling the ECM in a dynamic and reciprocal process (Bissell et al., 1982). Cells attach to specific ECM molecules via surface receptors, including integrins and the dystroglycan receptor (encoded by Dag1) (Cloutier et al., 2019; Pozzi and Zent, 2011). Intracellularly, these receptors are physically linked to a range of cytoskeleton and signalling proteins; hence, they mechanically and biochemically connect cells to insoluble extracellular cues (Nelson and Bissell, 2006; Pozzi and Zent, 2011).

Thus, the composition and mechanical properties of the ECM, along with the receptors and downstream effectors expressed by the cells, will dictate cell behaviour, modulating processes such as proliferation, quiescence, differentiation, motility and polarity (Bissell et al., 1982; Nelson and Bissell, 2006).

Epithelial cells reside on top of a specialized ECM compartment, the basement membrane (BM), the formation and microarchitecture of which is nucleated and stabilized mainly by laminins (LeBleu et al., 2007). The BM physically separates epithelial cells from the surrounding stroma, in which the ECM is mainly composed of type I collagen and fibronectin (LeBleu et al., 2007). An intact BM is essential to sustain epithelial homeostasis (Gaiko-Shcherbak et al., 2015; Khalilgharibi and Mao, 2021).

During pubertal mammary epithelial development, the BM is thicker in ducts than in terminal end buds (TEBs), (Sternlicht et al., 2006) and this has been extensively linked to the spatiotemporal regulation of mammary gland morphogenesis in vivo and in cell culture models such as ECM overlays and three-dimensional (3D) cell cultures, where cells retain the ability to respond to laminin-triggered quiescence and differentiation, even in the presence of mitogens and growth factors (Bhat et al., 2016; Fiore et al., 2017; Inman et al., 2015; Spencer et al., 2011).

In this work, we provide a molecular landscape of key regulators of laminin-rich ECM (lrECM)-triggered mammary epithelial cell quiescence and differentiation, which we found involves multiple layers of regulation at the mRNA and protein level. Interestingly, cell cycle arrest was achieved despite the active status of Fak (also known as PTK2), Src and phosphoinositide 3-kinases (PI3Ks), indicating the existence of a ‘disconnecting node’ between upstream and downstream proliferative signalling in laminin-induced quiescence. Our data indicate that Pten, a tumour suppressor that counteracts PI3K activity, fulfils this role: Pten was upregulated in lrECM-treated mammary epithelial cells, and Pten inhibition hindered quiescence by restoring activity of the Akt, mTORC1 and mTORC2 pathways, as well as mitogen-activated protein kinase (MAPK) signalling via Mek [here referring collectively to Mek1 (MAP2K1) and Mek2 (MAP2K2)] and Erk [here referring collectively to Erk1 (MAPK3) and Erk2 (MAPK1)]. In the developing mammary gland, the levels and tissue distribution of Pten were positively correlated to the degree of laminin staining and the presence of lumen. Furthermore, Pten inhibition in mammary 3D acini increased cell proliferation, disrupted polarity and led to loss of the lumen in both forming and mature structures. Taken together, these data highlight the core role of Pten in mammary gland quiescence regulation, with Pten downregulation being necessary for epithelial cell growth and Pten upregulation sustaining quiescence and tissue architecture.

Laminin-induced quiescence is characterized by alterations in cell size and morphology, G1 arrest, and decreased cyclin-dependent kinase activity

When the BM that envelopes the mammary epithelia is mimicked in culture systems using lrECM, either on a two-dimensional ECM overlay or in 3D assays, primary epithelial cells and some established non-malignant epithelial cell lines, such as murine EpH4 cells, exit the cell cycle (Mroue and Bissell, 2012), thus representing a physiological and suitable model for the study of microenvironmentally regulated cellular quiescence.

We further characterized this model using flow cytometry, cell cycle fluorescence probes and quantitative image analysis. As previously reported, lrECM overlay on two-dimensional cultures triggered remarkable morphological alterations and G1 arrest in EpH4 cells (Mroue and Bissell, 2012; Spencer et al., 2011; Xu et al., 2010), reducing the overall cell number without affecting cell viability (Fig. 1A–F; Fig. S1). LrECM-induced G1 arrest was consistently observed either by analysing DNA content via flow cytometry (Fig. 1D) or via fluorescence microscopy of EpH4 cells expressing the FUCCI reporter system (EpH4-ES-FUCCI) (Fig. 1F). The FUCCI system is based on the cyclic degradation of fluorescent probes labelling Cdt1 (mCherry) and geminin (mCitrine). Cdt1 accumulates during G1 and is degraded in early S phase, whereas geminin builds up over the S, G2 and M phases (S/G2/M); as such, FUCCI cells express mCherry during G1 and mCitrine during S/G2/M, allowing for monitoring of the cell cycle in live cells (Zielke and Edgar, 2015).

Fig. 1.

LrECM treatment of mammary epithelial cells induces G1 cell cycle arrest, reduces cell number and leads to morphological changes without inducing cell death. (A) General workflow of the lrECM-overlay model (figure created with Biorender.com). GIH, medium supplemented with gentamycin, insulin and hydrocortisone (see Materials and Methods); Prl, prolactin. (B) Bright-field images of freely cycling (Ctrl, control) or quiescent (lrECM-treated) EpH4 cells. Scale bar: 50 µm. (C) Cell density of lrECM-treated EpH4 cells (lrECM +), relative to that of control EpH4 cells (dots, normalized mean for each replicate; line, mean±s.e.m. of all replicates; n=3; two-tailed unpaired Student's t-test). (D) Cell cycle analysis (PI staining and flow cytometry). Left: representative histograms displaying count (y-axis) versus PI fluorescence intensity (x-axis). Intensity ranges corresponding to cell cycle phases and the percentage of cells within each range are shown. Right: distribution of cells across the cell cycle phases according to DNA content (sub-G1, G1, S, G2/M and >4C) in freely cycling (control, left) or quiescent (lrECM +, right) cells (mean±s.e.m.; n=3; two-way ANOVA with Sidak's multiple comparisons test). (E) Percentage of live and dead cells in control (left) and lrECM-treated (right) conditions (mean±s.e.m.; n=3; two-way ANOVA with Sidak's multiple comparisons test; ns, not significant). (F) Cell cycle analysis using live imaging of EpH4 cells expressing the FUCCI sensor and subjected to lrECM overlay assay for 48 h. n=3 biological replicates (number of cells analysed per biological replicate: control, 10,010±429; lrECM, 3567±409; mean±s.e.m.). Right: representative fluorescence images of live EpH4-ES-FUCCI cells that were either freely cycling (control) or quiescent (lrECM). mCitrine-positive cells (green) are in S/G2/M, whereas mCherry-positive cells (red) are in G1. Scale bar: 50 µm. Left: distribution of cells across the cell cycle according to the expression of the FUCCI sensors (G1, S/G2/M, and just finished M or very early G1) in control or lrECM-treated cells (mean±s.e.m.; two-way ANOVA with Sidak's multiple comparisons test). (G,H) Cdk4 and Cdk2 activity in EpH4 cells upon lrECM overlay treatment for 48 h. n=3 biological replicates (number of cells analysed per biological replicate: control, 19,658±3443; lrECM, 6334±1008; mean±s.e.m.). (G) Fluorescence images of live EpH4 cells expressing the nuclear marker histone H2B–mTurquoise (H2B–mTurq; cyan) and activity sensors for Cdk2 (DHB–Ven, DHB–mVenus; green) and Cdk4 (CDK4.KTR–mCh, mCherry–Cdk4KTR; red) that were either freely cycling (control) or quiescent (lrECM-treated). Merged image shows DHB–mVenus and mCherry–Cdk4KTR. Scale bar: 50 µm. (H) Cdk4 and Cdk2 activity (lighter points, values for individual cells; darker points, mean of each biological replicate; line, mean; two-tailed unpaired Student's t-test). (I) Size of cells in G1 for EpH4 cells treated or not with lrECM overlay for 48 h (lighter points, values for individual cells; darker points, mean of each biological replicate; line, mean; two-tailed unpaired Student's t-test). Left: cell size estimated using flow cytometry forward scatter area (FSC-A). n=3 biological replicates (number of cells analysed per biological replicate: control, 3777±68; lrECM, 6951±210; mean±s.e.m.). Right: cell size estimated by microscopy (nuclear area, µm2). n=3 biological replicates (number of cells analysed per biological replicate: control, 1447±179.2; lrECM, 1814±147; mean±s.e.m.).

Fig. 1.

LrECM treatment of mammary epithelial cells induces G1 cell cycle arrest, reduces cell number and leads to morphological changes without inducing cell death. (A) General workflow of the lrECM-overlay model (figure created with Biorender.com). GIH, medium supplemented with gentamycin, insulin and hydrocortisone (see Materials and Methods); Prl, prolactin. (B) Bright-field images of freely cycling (Ctrl, control) or quiescent (lrECM-treated) EpH4 cells. Scale bar: 50 µm. (C) Cell density of lrECM-treated EpH4 cells (lrECM +), relative to that of control EpH4 cells (dots, normalized mean for each replicate; line, mean±s.e.m. of all replicates; n=3; two-tailed unpaired Student's t-test). (D) Cell cycle analysis (PI staining and flow cytometry). Left: representative histograms displaying count (y-axis) versus PI fluorescence intensity (x-axis). Intensity ranges corresponding to cell cycle phases and the percentage of cells within each range are shown. Right: distribution of cells across the cell cycle phases according to DNA content (sub-G1, G1, S, G2/M and >4C) in freely cycling (control, left) or quiescent (lrECM +, right) cells (mean±s.e.m.; n=3; two-way ANOVA with Sidak's multiple comparisons test). (E) Percentage of live and dead cells in control (left) and lrECM-treated (right) conditions (mean±s.e.m.; n=3; two-way ANOVA with Sidak's multiple comparisons test; ns, not significant). (F) Cell cycle analysis using live imaging of EpH4 cells expressing the FUCCI sensor and subjected to lrECM overlay assay for 48 h. n=3 biological replicates (number of cells analysed per biological replicate: control, 10,010±429; lrECM, 3567±409; mean±s.e.m.). Right: representative fluorescence images of live EpH4-ES-FUCCI cells that were either freely cycling (control) or quiescent (lrECM). mCitrine-positive cells (green) are in S/G2/M, whereas mCherry-positive cells (red) are in G1. Scale bar: 50 µm. Left: distribution of cells across the cell cycle according to the expression of the FUCCI sensors (G1, S/G2/M, and just finished M or very early G1) in control or lrECM-treated cells (mean±s.e.m.; two-way ANOVA with Sidak's multiple comparisons test). (G,H) Cdk4 and Cdk2 activity in EpH4 cells upon lrECM overlay treatment for 48 h. n=3 biological replicates (number of cells analysed per biological replicate: control, 19,658±3443; lrECM, 6334±1008; mean±s.e.m.). (G) Fluorescence images of live EpH4 cells expressing the nuclear marker histone H2B–mTurquoise (H2B–mTurq; cyan) and activity sensors for Cdk2 (DHB–Ven, DHB–mVenus; green) and Cdk4 (CDK4.KTR–mCh, mCherry–Cdk4KTR; red) that were either freely cycling (control) or quiescent (lrECM-treated). Merged image shows DHB–mVenus and mCherry–Cdk4KTR. Scale bar: 50 µm. (H) Cdk4 and Cdk2 activity (lighter points, values for individual cells; darker points, mean of each biological replicate; line, mean; two-tailed unpaired Student's t-test). (I) Size of cells in G1 for EpH4 cells treated or not with lrECM overlay for 48 h (lighter points, values for individual cells; darker points, mean of each biological replicate; line, mean; two-tailed unpaired Student's t-test). Left: cell size estimated using flow cytometry forward scatter area (FSC-A). n=3 biological replicates (number of cells analysed per biological replicate: control, 3777±68; lrECM, 6951±210; mean±s.e.m.). Right: cell size estimated by microscopy (nuclear area, µm2). n=3 biological replicates (number of cells analysed per biological replicate: control, 1447±179.2; lrECM, 1814±147; mean±s.e.m.).

LrECM-induced quiescence was also accompanied by a decrease in the activity of the cyclin-dependent kinases (Cdks) Cdk4 and Cdk2 (Fig. 1G,H). Cdk2 and Cdk4 activities were evaluated using specific fluorescent reporters and fluorescence microscopy (Spencer et al., 2013; Yang et al., 2020). In this system, cells express a nuclear marker (histone H2B–mTurquoise) and probes for Cdk2 (DHB–mVenus) and Cdk4 (mCherry–Cdk4KTR; this probe also detects Cdk6 activity, but in mammary cells, Cdk4 is the predominant kinase) activities. Upon phosphorylation by Cdk2, or by Cdk4 or Cdk6, the respective fluorescent probe is translocated from the nucleus to the cytoplasm. Therefore, high Cdk activity results in the probe being translocated to the cytoplasm, and thus, Cdk activity can be estimated by the ratio of the cytoplasmic and nuclear mean fluorescence signal of a given probe (Spencer et al., 2013; Yang et al., 2020).

Quiescent cells were smaller than freely cycling cells, even when only cells in G1 were considered (Fig. 1I). This was consistent when size was estimated via flow cytometry (Fig. 1I, left) or via microscopy (Fig. 1I, right). Despite these differences in estimated cell size between quiescent and cycling G1 cells, the correlations between nuclear area and Cdk activity, for both Cdk4 and Cdk2, in freely cycling and lrECM-treated cells were weak (Fig. S2), and thus, discrimination between cycling G1 cells and G1-arrested cells based solely on nuclear area was not feasible. A possible explanation for this observation is that newly born cells would be small and would present low Cdk activity, but, unlike quiescent cells, their cell size and Cdk activity would build up over time (Rhind, 2021; Spencer et al., 2013), whereas quiescent cells would remain small and with low Cdk activity.

Many molecular circuits could be involved in the acquisition and maintenance of the quiescent phenotype, and this probably accounts for the morphological and functional diversity amongst quiescent cells in nature (O'Farrell et al., 2011). We sought to delineate a landscape of key molecules involved in cell cycle progression that are regulated during laminin-induced quiescence in mammary epithelial cells.

Laminin-induced quiescence and differentiation in mammary epithelial cells are accompanied by alterations in cell cycle regulators at the transcriptional and protein levels

In addition to becoming quiescent in the presence of lrECM, upon concomitant exposure to prolactin, EpH4 cells become functionally differentiated for lactogenesis and produce milk-related proteins (Xu et al., 2009). Thus, the EpH4 model is largely used for the study of cellular and molecular mechanisms underlying mammary gland functional differentiation. In this sense, untreated (proliferating) cells could be considered a parallel for the proliferative TEBs found in the developing mammary gland, whereas lrECM-treated cells would be comparable to the quiescent cells found in the ducts and immature alveoli. Similarly, cells treated with both lrECM and prolactin would represent cells found in the alveoli of a fully mature, milk-producing mammary gland.

We compared proliferating, quiescent and lactogenic EpH4 cells with respect to some key cell cycle regulators, initially at the transcriptional level (Fig. 2). The mRNA levels of several cell cycle regulators dramatically changed upon lrECM treatment, regardless of co-treatment with prolactin (Fig. 2A; Fig. S3). Confirming the functional differentiation state towards a lactogenic programme (Xu et al., 2009), only exposure to both lrECM and prolactin triggered the expression of Csn2, the gene encoding β-casein (Fig. 2A). The mRNA expression of most positive regulators of the cell cycle (such as cyclins and Cdks) plummeted upon lrECM treatment (Fig. 2A), but surprisingly, so did the expression levels of some negative regulators of the cell cycle (such as Trp53, Cdkn1a, Cdkn2c and Cdkn2d). On the other hand, the mRNA levels of Pten, Dag1 and Cdkn1b were unchanged across all treatments (Fig. 2A). These data confirm some previous findings indicating that upon laminin-induced quiescence acquisition there is a general repression of transcription (Spencer et al., 2011; Fiore et al., 2017). But how are these cells arrested if, apparently, expression of negative regulators of the cell cycle is either unchanged or also downregulated? The lack of alteration or even the decrease observed in mRNA levels of cell cycle negative regulators might indicate that these molecules are most likely to be controlled at the protein level during lrECM-induced quiescence (Koepp, 2014); therefore, we assessed the protein levels of a range of known positive and negative regulators of the cell cycle by western blotting (Fig. 2B–F).

Fig. 2.

Quiescent epithelial cells sustain the activity of Fak, Src and PI3K but display decreased signalling in downstream positive regulators of cellular proliferation. (A–F) EpH4 cells were treated or not with lrECM overlay and/or prolactin (Prl) for 48 h and evaluated for mRNA and protein expression levels of some key cell cycle regulators (n=3). (A) Relative mRNA expression levels assessed by RT-qPCR. Colour-code indicates log2 fold change (log2FC) and the size of circles indicate significance (pVal; one-way ANOVA with Dunnett's multiple comparison test comparing all samples to the control condition of no lrECM and no Prl). (B) Immunoblotting of key proliferation and cell cycle regulators for assessment of protein levels. Left: upstream regulators of cell proliferation. Middle: intermediate mediators of cell proliferation. Right: downstream positive and negative regulators of the cell cycle. Lamin B and tubulin are shown as loading controls. Estimated molecular masses of bands are indicated in kDa. α-DG, α-dystroglycan; β-DG, β-dystroglycan; p, phospho. (C–F) Volcano plots displaying direct comparisons between pairs of conditions regarding the expression levels of proteins quantified by western blotting (log2FC versus –log10P-value). Comparisons between two groups were performed by two-tailed unpaired Student's t-test. P<0.05 (−log10P-value=1.30, grey line) was considered significant. Dashed lines mark a 25% decrease (blue) or increase (red) in protein level. (C) lrECM-treated (quiescent) versus control (Ctrl; proliferative) cells. (D) Cells treated with lrECM and Prl (quiescent and lactogenic) versus cells treated only with Prl (proliferative). (E) Prl (proliferative) versus Ctrl (proliferative) cells. (F) Cells treated with lrECM and Prl (quiescent and lactogenic) versus lrECM-treated (quiescent) cells. Points shown in blue represent proteins that were significantly decreased; points shown in red represent proteins that were significantly increased; points shown in grey represent proteins that were not altered. a-DG, α-dystroglycan; b-DG, β-dystroglycan.

Fig. 2.

Quiescent epithelial cells sustain the activity of Fak, Src and PI3K but display decreased signalling in downstream positive regulators of cellular proliferation. (A–F) EpH4 cells were treated or not with lrECM overlay and/or prolactin (Prl) for 48 h and evaluated for mRNA and protein expression levels of some key cell cycle regulators (n=3). (A) Relative mRNA expression levels assessed by RT-qPCR. Colour-code indicates log2 fold change (log2FC) and the size of circles indicate significance (pVal; one-way ANOVA with Dunnett's multiple comparison test comparing all samples to the control condition of no lrECM and no Prl). (B) Immunoblotting of key proliferation and cell cycle regulators for assessment of protein levels. Left: upstream regulators of cell proliferation. Middle: intermediate mediators of cell proliferation. Right: downstream positive and negative regulators of the cell cycle. Lamin B and tubulin are shown as loading controls. Estimated molecular masses of bands are indicated in kDa. α-DG, α-dystroglycan; β-DG, β-dystroglycan; p, phospho. (C–F) Volcano plots displaying direct comparisons between pairs of conditions regarding the expression levels of proteins quantified by western blotting (log2FC versus –log10P-value). Comparisons between two groups were performed by two-tailed unpaired Student's t-test. P<0.05 (−log10P-value=1.30, grey line) was considered significant. Dashed lines mark a 25% decrease (blue) or increase (red) in protein level. (C) lrECM-treated (quiescent) versus control (Ctrl; proliferative) cells. (D) Cells treated with lrECM and Prl (quiescent and lactogenic) versus cells treated only with Prl (proliferative). (E) Prl (proliferative) versus Ctrl (proliferative) cells. (F) Cells treated with lrECM and Prl (quiescent and lactogenic) versus lrECM-treated (quiescent) cells. Points shown in blue represent proteins that were significantly decreased; points shown in red represent proteins that were significantly increased; points shown in grey represent proteins that were not altered. a-DG, α-dystroglycan; b-DG, β-dystroglycan.

LrECM-induced quiescent cells displayed dramatic changes in protein expression and/or phosphorylation levels of key cell cycle regulators (Fig. 2B–F). Again, lrECM-treated cells exhibited similar protein expression profiles concerning cell cycle regulators, regardless of prolactin supplementation (Fig. 2B,F), and likewise, cells treated only with prolactin exhibited a protein expression profile similar to that of untreated cells (Fig. 2B,E).

Quiescent cells displayed decreased protein levels of positive regulators of proliferation, such cyclins and Cdks (Fig. 2B–D), matching their transcriptional repression (Fig. 2A), which resulted in the near absence of hyperphosphorylated retinoblastoma protein (Rb, also known as RB1) in lrECM-treated quiescent cells (Fig. 2B–D). Quiescent cells also had increased protein levels of some negative regulators of the cell cycle, such as Pten (Worby and Dixon, 2014), p27 (also known as CDKN1B) (Sherr and Roberts, 1999) and the dystroglycan receptor (Sgambato et al., 2004), which showed no alterations at the transcript level (Fig. 2A). The balance between cyclins and cyclin-dependent kinase inhibitors (CKIs) is believed to be one of the components that dictates cell cycle progression (Gérard and Goldbeter, 2012; Yang et al., 2017); thus, the decreased activity of Cdk4 and Cdk2 observed in lrECM-treated cells (Fig. 1G,H) could be, at least in part, attributed to the lower levels of cyclins and Cdks and the increased levels of p27 found in quiescent cells (Fig. 2B–D).

LrECM treatment attenuated many proliferative signalling pathways, such as the MAPK pathway (Fiore et al., 2017), as evidenced by reduced levels of phosphorylated Mek and phosphorylated Erk, and the PI3K–Akt–mTORC1 and mTORC2 (mTORC1/2) pathways (Fiore et al., 2017), as evidenced by reduced levels of phosphorylated Akt kinase (herein referring collectively to Akt1, Akt2 and Akt3) and phosphorylated S6 (also known as RPS6) (Fig. 2B–D). Surprisingly though, lrECM-treated quiescent cells displayed increased or unchanged levels of active (phosphorylated) Fak, Src and PI3K (Fig. 2B–D). However, downstream signalling pathways triggered by these kinases were widely downregulated in the presence of lrECM (Fig. 2B–D). These included the PI3K–Akt–mTORC1/2 axis and the MAPK pathway (Fig. 2B–D). Fak, Src and PI3K are components of adhesion complexes and are activated upon integrin binding to ECM molecules (Glukhova and Streuli, 2013). Fak, Src and PI3K also take part in the control of mitogenic signalling triggered by the binding of growth factors to receptor tyrosine kinases (RTKs) (Streuli and Akhtar, 2009). There is significant cross-activation between Fak, Src and PI3K, and although each of them has specific functions in cell behaviour, their activation usually triggers Akt–mTORC1/2 and MAPK signalling (Moreno-Layseca and Streuli, 2014), which places them as positive regulators of cell proliferation.

Decreased phosphorylation levels of Akt and Mek–Erk have been previously reported in EpH4 acini (Janda et al., 2002; Tognon et al., 2011) and also in non-malignant human mammary epithelial S1 (Beliveau et al., 2010; Fiore et al., 2017; Onodera et al., 2014) and MCF 10A (Debnath et al., 2003; Watt et al., 2017) cells undergoing laminin-induced quiescence.

The Ras–Raf–Mek–Erk axis is the most crucial MAPK signalling pathway for the positive regulation of cell proliferation in epithelial cells (Sun et al., 2015). It is manly triggered by the binding of mitogens and growth factors to RTKs, but it is also modulated by the ECM (Moreno-Layseca and Streuli, 2014; Streuli and Akhtar, 2009; Sun et al., 2015). Complementarily, mTOR controls cell growth by promoting anabolism and suppressing catabolism, and its function might be different depending on upstream signalling (Kim and Guan, 2019). Consistent with the smaller cell size observed in quiescent cells (Fig. 1I), we observed decreased levels of phosphorylation of S6 (at S240 and/or S244) and Akt (at S473), reflecting the activities of mTORC1 and mTORC2, respectively.

Another feature of quiescent cells that captured our attention was the presence of phosphorylated PI3K p55 only in lrECM-treated cells (Fig. 2B). The PI3K complex is comprised of catalytic (p110) and regulatory (p85 or its alternative spliced versions p50 and p55) subunits. Specific functions for different PI3K isoforms in mammary gland development have been described, including a role of p110γ (encoded by Pik3cg) in lumen formation (Peng et al., 2015) and for p55α and/or p50α (encoded by Pik3r1) in cell death induction during mammary gland involution (Pensa et al., 2014). Thus, phosphorylated PI3K p55 might have functions beyond the regulation of proliferation in lrECM-induced quiescence, such as suppression of apoptosis when mammary epithelial cells are exposed to a reconstituted BM (Boudreau et al., 1995).

Pharmacological inhibition of key mediators identifies Pten as a disrupting node for proliferative signalling during lrECM-induced quiescence

The data on protein expression prompted us to assess the contribution of different signalling pathways to the cell cycle arrest upon lrECM treatment by using commercially available inhibitors (Fig. 3; Table S3).

Fig. 3.

Inhibition of upstream and downstream regulators of the cell cycle affects the balance between proliferation and quiescence in mammary epithelial cells. (A,B) FUCCI-based cell cycle distribution (top left) and relative cell density (bottom left) in EpH4-ES-FUCCI cells treated or not with lrECM overlay and specific inhibitors for 24 h, and representative images (right) displaying mCherry-positive cells (G1) and mCitrine-positive cells (S/G2/M) for each condition. Scale bars: 50 µm. Cells were treated (A) only with the indicated inhibitors or (B) with lrECM and inhibitors simultaneously. Experiments were performed in two biological replicates with two replicate wells per biological replicate. Graphs show mean±s.e.m. of n=2. Relative cell density (normalized to the control condition) was analysed by one-way ANOVA followed by multiple comparisons using the two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli (control versus inhibitor treatments), where q<0.05 was considered significant (blue asterisks). FUCCI-based cell cycle analysis was performed using two-way ANOVA followed by multiple comparisons using the two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli (control versus inhibitor treatments for each cell cycle phase), where q<0.05 was considered significant. Blue hash symbol indicates statistically significant differences for G1, S/G2/M and just finished M/very early G1. Red asterisks indicate statistically significant differences for G1. Green asterisks indicate statistically significant differences for S/G2/M. Black asterisks indicate statistically significant differences for just finished M/very early G1. Dashed lines in the cell cycle distribution graphs indicate the percentage of G1 cells in the control condition. Faki: FAK inhibitor 14 (1 µM); Srci: PP2 (2 µM); ILKi: CPD22 (0.25 µM); Jak/Stati: Tofacitinib (20 µM); PI3Ki: LY294002 (20 µM); Pteni: bpV(pic) (2.5 µM); mTORC1i: rapamycin (0.2 µM); mTORC1/2i: Torin 2 (0.4 µM); Akti: MK 2206 (2 µM); Meki: PD98059 (20 µM); Cdk4i: Palbociclib (1 µM), Cdk2i: Cdk2 inhibitor III (60 µM).

Fig. 3.

Inhibition of upstream and downstream regulators of the cell cycle affects the balance between proliferation and quiescence in mammary epithelial cells. (A,B) FUCCI-based cell cycle distribution (top left) and relative cell density (bottom left) in EpH4-ES-FUCCI cells treated or not with lrECM overlay and specific inhibitors for 24 h, and representative images (right) displaying mCherry-positive cells (G1) and mCitrine-positive cells (S/G2/M) for each condition. Scale bars: 50 µm. Cells were treated (A) only with the indicated inhibitors or (B) with lrECM and inhibitors simultaneously. Experiments were performed in two biological replicates with two replicate wells per biological replicate. Graphs show mean±s.e.m. of n=2. Relative cell density (normalized to the control condition) was analysed by one-way ANOVA followed by multiple comparisons using the two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli (control versus inhibitor treatments), where q<0.05 was considered significant (blue asterisks). FUCCI-based cell cycle analysis was performed using two-way ANOVA followed by multiple comparisons using the two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli (control versus inhibitor treatments for each cell cycle phase), where q<0.05 was considered significant. Blue hash symbol indicates statistically significant differences for G1, S/G2/M and just finished M/very early G1. Red asterisks indicate statistically significant differences for G1. Green asterisks indicate statistically significant differences for S/G2/M. Black asterisks indicate statistically significant differences for just finished M/very early G1. Dashed lines in the cell cycle distribution graphs indicate the percentage of G1 cells in the control condition. Faki: FAK inhibitor 14 (1 µM); Srci: PP2 (2 µM); ILKi: CPD22 (0.25 µM); Jak/Stati: Tofacitinib (20 µM); PI3Ki: LY294002 (20 µM); Pteni: bpV(pic) (2.5 µM); mTORC1i: rapamycin (0.2 µM); mTORC1/2i: Torin 2 (0.4 µM); Akti: MK 2206 (2 µM); Meki: PD98059 (20 µM); Cdk4i: Palbociclib (1 µM), Cdk2i: Cdk2 inhibitor III (60 µM).

As expected, treatment with lrECM increased the percentage of cells in G1 (quiescent), and treatment with pharmacological inhibitors of intermediate and downstream positive regulators of the cell cycle (such as mTORC1/2, Akt, Mek, Cdk4 and Cdk2) mostly decreased cell cycle progression and, in general, increased the percentage of Eph4-ES-FUCCI cells arrested in G1 in both control and lrECM conditions (Fig. 3A and B, respectively).

Even though both cyclinE1–Cdk2 and cyclinD1–Cdk4 were less active and had lower expression levels in quiescent cells (Figs 1G,H, 2A–D), the effects of Cdk2 inhibition on cell cycle and cell number were more prominent than those observed for Cdk4 inhibition (Fig. 3B). Cells can often bypass loss of cyclins or Cdks and progress through the cell cycle by activating alternative cyclin–Cdk complexes (Malumbres and Barbacid, 2009; Shapiro, 2006; Whittaker et al., 2017). Particularly, mature cyclinD–Cdk4 complexes are refractory to palbociclib, the Cdk4 inhibitor used in this work. Palbociclib is only functional if binding to Cdk4 by itself, before Cdk4 complexes with cyclinD, and therefore inhibits cell cycle progression by preventing the assembly of new cyclin D–Cdk4 complexes and their activity, and also by indirectly inhibiting Cdk2 (Guiley et al., 2019). Furthermore, the smooth-edged and compact morphology of cell clusters treated with Cdk2 inhibitor (Fig. 3A, Cdk2i) closely resembled those formed by quiescent lrECM-treated cells (Fig. 3B, lrECM), which was not the case for cells treated with the Cdk4 inhibitor (Fig. 3A, Cdk4i). These data could be indicative that Cdk2 and Cdk4 have distinct roles in lrECM-induced quiescence.

Inhibition of Src, PI3K, mTORC1/2 and Akt all gave rise to small, G1-arrested cells, whereas Fak inhibition arrested cells in G2 (Fig. 3A,B). G2 arrest upon Fak inhibition was evidenced by increased mCitrine signal along with decreased cell number (Fig. 3A,B), and this is in agreement with previous reports (Aboubakar Nana et al., 2019; Cabrita et al., 2011). Inhibition of the Jak–Stat pathway or Ilk had no or only minor effects on the cell cycle (Fig. 3A,B). This is consistent with previous reports that have proposed a role for Jak–Stat signalling in milk production rather than proliferation (Xu et al., 2009). Similarly, Fak, Akt and Ilk signalling have all been linked to cell survival and/or polarity (Keely, 2011; Lee and Streuli, 2014), whereas Src and PI3K–Akt–mTORC1/2 signalling are involved in the control of cell size (Cohen, 2005; Kim and Guan, 2019).

Another key observation from this assay was that, despite the high levels of phosphorylated Fak, Src and PI3K proteoforms found in quiescent cells (Fig. 2), inhibition of these molecules changed cell proliferation (arresting cells in G1 for Src and PI3K inhibition and in G2 for Fak inhibition) (Fig. 3). G1 arrest upon Src and PI3K inhibition (Chu et al., 2007; Liu et al., 2004), as well as G2 arrest in response to Fak inhibition (Aboubakar Nana et al., 2019; Cabrita et al., 2011) have been previously reported. Therefore, the activity of these signalling pathways, as a default, contributes to proliferation in mammary epithelial cells.

How could Fak, Src and PI3K be active, but their downstream proliferative pathways be downregulated in quiescent lrECM-treated cells? There must be a ‘disconnecting point’ between upstream and downstream proliferative signalling triggered lrECM treatment.

Pten, which was found to be upregulated in lrECM-treated cells (Fig. 2B–D) could fulfil this role: its lipid phosphatase function counterbalances PI3K activity by converting phosphatidylinositol (3,4,5)-trisphosphate (PIP3) to phosphatidylinositol (4,5)-bisphosphate (PIP2) (Worby and Dixon, 2014), whereas its protein phosphatase activity can target Fak (Tamura et al., 1999). Strikingly, Pten inhibition led to increased proliferation, increased cell number, a reduced percentage of cells in G1 and an increased percentage of cells in S/G2/M (Fig. 3). Alterations induced by Pten inhibition were clearer in lrECM-treated cells, in which the distribution of cells across the phases of the cell cycle resembled that observed for freely cycling (control) cells (Fig. 3A,B).

Furthermore, inhibition of Pten increased the levels of active Fak (phospho-FakY397) and Src (phospho-SrcY416), and partially restored PI3K (phospho-AktT308), MAPK (phospho-Erk), mTORC1 (phospho-S6) and mTORC2 (phospho-AktS473) signalling in lrECM-treated cells (Fig. 4). Treatment with Pten inhibitor in the absence of lrECM had overall milder effects in further increasing phosphorylated levels of Fak, Akt, Erk and S6 (Fig. 4), which is consistent with the notion that sub confluent freely cycling (control) cells in culture are in exponential growth, near the maximum of their proliferation rates (https://www.atcc.org/resources/culture-guides/animal-cell-culture-guide#Cell).

Fig. 4.

Inhibition of Pten reverts lrECM-induced quiescence by partially restoring PI3K, Src, Erk, mTORC1 and mTORC2 signalling. (A) Immunoblots of EpH4 cells treated or not with lrECM overlay and/or Pten inhibitor [Pteni; bpV(pic), 2.5 µM] for 24 h (representative of n=3), probed for the indicated proteins and phosphoproteins (p, phospho). Estimated molecular masses of bands are indicated in kDa. Tubulin is shown as a loading control. (B) Left: quantification of blots as displayed in A (n=3; points show data for each biological replicate, normalized to the control condition, and connecting lines link samples from a given replicate). p-FakY397, Pten target; phospho-SrcY416; phospho-AktT308, PKD1 substrate; phospho-AktS473, mTORC2 substrate; phospho-S6S240/244, mTORC1 substrate; phospho-ErkT202/Y204, MAPK pathway. Right: quantification of Pten expression as assayed in A, normalized to the control condition (mean±s.e.m., n=3). Statistical differences were assessed by one-way ANOVA with Tukey post hoc test for multiple comparisons.

Fig. 4.

Inhibition of Pten reverts lrECM-induced quiescence by partially restoring PI3K, Src, Erk, mTORC1 and mTORC2 signalling. (A) Immunoblots of EpH4 cells treated or not with lrECM overlay and/or Pten inhibitor [Pteni; bpV(pic), 2.5 µM] for 24 h (representative of n=3), probed for the indicated proteins and phosphoproteins (p, phospho). Estimated molecular masses of bands are indicated in kDa. Tubulin is shown as a loading control. (B) Left: quantification of blots as displayed in A (n=3; points show data for each biological replicate, normalized to the control condition, and connecting lines link samples from a given replicate). p-FakY397, Pten target; phospho-SrcY416; phospho-AktT308, PKD1 substrate; phospho-AktS473, mTORC2 substrate; phospho-S6S240/244, mTORC1 substrate; phospho-ErkT202/Y204, MAPK pathway. Right: quantification of Pten expression as assayed in A, normalized to the control condition (mean±s.e.m., n=3). Statistical differences were assessed by one-way ANOVA with Tukey post hoc test for multiple comparisons.

These data, in combination with the protein expression analysis (Fig. 2B–D), indicated that Pten could act as a disrupting node for Fak–Src–PI3K proliferative signalling in mammary epithelial cells, especially upon quiescence induction by a laminin-rich microenvironment.

Pten correlates with laminin deposition in the developing mammary gland and displays different distribution and polarization patterns in ducts and TEBs

Our data obtained for mammary epithelial cells pinpointed Pten as being upregulated upon lrECM treatment and as an important regulator of laminin-induced quiescence. To investigate whether this holds true in vivo, we assessed the correlation between laminin deposition and Pten staining in developing murine mammary glands (Fig. 5). Ducts, which are laminin-rich, mostly quiescent epithelial structures in the tubuloalveolar mammary network (Sternlicht, 2005) had higher levels of Pten (Fig. 5B,C). TEBs, which are the laminin-poor, invasive and proliferative tip of the growing epithelia in the mammary gland (Sternlicht, 2005) had overall lower levels and an unclear pattern of Pten staining (Fig. 5B,C). Furthermore, Pten intensity was found to be positively correlated with the degree of laminin deposition (Fig. 5B,D).

Fig. 5.

Pten levels are positively correlated with laminin-111 deposition in the developing mammary gland. (A–D) Immunofluorescence analysis of Pten and laminin-111 levels in mammary glands of 5-week-old female mice (n=3 mammary fat pads from three different mice). (A) Schematic view of a developing mammary gland from a 5-week-old female mouse. An example of areas corresponding to a duct (top) and a TEB (bottom) are highlighted by dashed rectangles. (B) Representative images of a duct (top) and a TEB (bottom) stained for Pten (green) and laminin-111 (LN-111, magenta). Nuclei are stained using DAPI (blue). Scale bars: 25 µm. (C) Mean fluorescence intensity of Pten in cells found in TEBs (green) or ducts (pink) (n=3 mice). Each open symbol corresponds to the mean of the mean fluorescence intensity of Pten per cell in a given field of view (TEB, 46 fields; duct, 41 fields), whereas filled symbol represent the mean fluorescence intensity of Pten for all fields of view in a given mouse (n=3 mice), and the line marks the mean fluorescence intensity of Pten for ducts or TEBs amongst all mice. The number of cells contained in each field of view ranged from 56 to 248 for ducts and 155 to 335 for TEBs. Statistical differences were evaluated by two-tailed unpaired Student's t-test. (D) Correlation analysis (Pearson) of Pten levels and laminin-111 deposition in TEBs (green) and ducts (pink) (n=3 mice). Each point represents a TEB or a duct analysed. Data are plotted as log10 fluorescence intensity. Pearson r and linear regression line are marked on the graph. For further details on data analysis, please refer to the Materials and Methods.

Fig. 5.

Pten levels are positively correlated with laminin-111 deposition in the developing mammary gland. (A–D) Immunofluorescence analysis of Pten and laminin-111 levels in mammary glands of 5-week-old female mice (n=3 mammary fat pads from three different mice). (A) Schematic view of a developing mammary gland from a 5-week-old female mouse. An example of areas corresponding to a duct (top) and a TEB (bottom) are highlighted by dashed rectangles. (B) Representative images of a duct (top) and a TEB (bottom) stained for Pten (green) and laminin-111 (LN-111, magenta). Nuclei are stained using DAPI (blue). Scale bars: 25 µm. (C) Mean fluorescence intensity of Pten in cells found in TEBs (green) or ducts (pink) (n=3 mice). Each open symbol corresponds to the mean of the mean fluorescence intensity of Pten per cell in a given field of view (TEB, 46 fields; duct, 41 fields), whereas filled symbol represent the mean fluorescence intensity of Pten for all fields of view in a given mouse (n=3 mice), and the line marks the mean fluorescence intensity of Pten for ducts or TEBs amongst all mice. The number of cells contained in each field of view ranged from 56 to 248 for ducts and 155 to 335 for TEBs. Statistical differences were evaluated by two-tailed unpaired Student's t-test. (D) Correlation analysis (Pearson) of Pten levels and laminin-111 deposition in TEBs (green) and ducts (pink) (n=3 mice). Each point represents a TEB or a duct analysed. Data are plotted as log10 fluorescence intensity. Pearson r and linear regression line are marked on the graph. For further details on data analysis, please refer to the Materials and Methods.

One feature that became clear while analysing the images of the developing mammary glands was that Pten presented distinct staining patterns in ducts and TEBs, and amongst specific regions or cell types. In the ducts, Pten showed strong apicolateral staining in the membrane of luminal cells (Fig. 5B, top); this pattern was only seen in cells surrounding the nascent lumen in TEBs (Fig. 5B, bottom).

Once we observed a predominance of apicolateral Pten staining, particularly in polarized luminal epithelial cells in the duct, we next stained developing mammary glands for Pten and F-actin, and imaged ducts and TEBs optically sliced at different focal planes using confocal and super-resolution microscopy (Fig. 6). In ducts, Pten appeared in puncta at the basal region (portion of the cell laying on the BM) and at the membrane along with the actin belt in the apical region (facing the lumen) (Fig. 6A). In TEBs (Fig. 6B), which mostly lack polarization, there was not a clear pattern of Pten staining. The exceptions were cap cells, which are found at the front edge of TEBs, where Pten was mainly found in the nuclei (Fig. 6B, middle row), and cells surrounding the nascent lumen, which presented a staining pattern of Pten and F-actin similar to that observed in the apical region of ducts (Fig. 6B, top row).

Fig. 6.

Pten displays distinct localization patterns within different compartments of ducts and TEBs. (A,B) Confocal and super-resolution microscopy images of epithelial cells in the developing mammary gland stained for F-actin (red), laminin-111 (LN-111, magenta), Pten (green) and counterstained with DAPI (nuclei, blue) (n=3 mammary fat pads from three different mice). (A) Localization within ducts. Left: schematic representation of a duct, displaying to which z position in the structure the microscopy images correspond to (dashed lines and corresponding images 1–3). 1, central; 2, near the lumen (apical domain of luminal cells); 3, near the BM (basal). A key to cell types in the schematic is shown at the top of the figure. Right: microscopy images of a duct at different z positions. The merged image displayed on the left was taken with a confocal microscope, and the dashed rectangles highlight the areas selected for Airyscan super-resolution imaging (images of separate channels displayed on the right). Note Pten apical staining in images from z positions 1 and 2. Scale bars: confocal images, 20 µm; Airyscan images, 5 µm. (B) Localization within TEBs. Left: schematic representation of a TEB, displaying to which z position in the structure the microscopy images correspond to (dashed lines and corresponding images 1 and 2). 1, central; 2, closer to the BM. A key to cell types in the schematic is shown at the top of the figure. Right: microscopy images of the TEB at different z positions. The merged image displayed on the left was taken with a confocal microscope, and the dashed rectangles highlight the areas selected for Airyscan super-resolution imaging (images of separate channels displayed on the right) In the TEB at z position 1, note apical Pten staining in cells near the nascent lumen (top row) and nuclear Pten staining in cap cells (middle row). Images for different heights were not necessarily taken in the same field or structure. Scale bars: confocal images, 20 µm; Airyscan images, 5 µm. (C,D) Quantification of Pten fluorescence in cells from different compartments of ducts (luminal or basal cells) and TEBs [cells near the nascent lumen, body cells or basal (cap) cells] (n=3 mice, one TEB and one duct per mouse). A two-tailed paired Student's t-test was used to analyse the differences between duct luminal versus duct basal cells, and a repeated measures one-way ANOVA with Tukey post hoc test was used to analyse differences between TEB near lumen versus TEB body versus TEB basal (cap) cells. (C) Mean fluorescence intensity of Pten amongst the different regions analysed. Lines connect ductal or TEB compartments from the same mouse. (D) Cytoplasmic:nuclear ratio of Pten fluorescence amongst the different regions analysed. Open symbols represent individual field of view, filled symbols represent the mean from all fields of view from a given mouse, and the line marks the mean from the three mice analysed.

Fig. 6.

Pten displays distinct localization patterns within different compartments of ducts and TEBs. (A,B) Confocal and super-resolution microscopy images of epithelial cells in the developing mammary gland stained for F-actin (red), laminin-111 (LN-111, magenta), Pten (green) and counterstained with DAPI (nuclei, blue) (n=3 mammary fat pads from three different mice). (A) Localization within ducts. Left: schematic representation of a duct, displaying to which z position in the structure the microscopy images correspond to (dashed lines and corresponding images 1–3). 1, central; 2, near the lumen (apical domain of luminal cells); 3, near the BM (basal). A key to cell types in the schematic is shown at the top of the figure. Right: microscopy images of a duct at different z positions. The merged image displayed on the left was taken with a confocal microscope, and the dashed rectangles highlight the areas selected for Airyscan super-resolution imaging (images of separate channels displayed on the right). Note Pten apical staining in images from z positions 1 and 2. Scale bars: confocal images, 20 µm; Airyscan images, 5 µm. (B) Localization within TEBs. Left: schematic representation of a TEB, displaying to which z position in the structure the microscopy images correspond to (dashed lines and corresponding images 1 and 2). 1, central; 2, closer to the BM. A key to cell types in the schematic is shown at the top of the figure. Right: microscopy images of the TEB at different z positions. The merged image displayed on the left was taken with a confocal microscope, and the dashed rectangles highlight the areas selected for Airyscan super-resolution imaging (images of separate channels displayed on the right) In the TEB at z position 1, note apical Pten staining in cells near the nascent lumen (top row) and nuclear Pten staining in cap cells (middle row). Images for different heights were not necessarily taken in the same field or structure. Scale bars: confocal images, 20 µm; Airyscan images, 5 µm. (C,D) Quantification of Pten fluorescence in cells from different compartments of ducts (luminal or basal cells) and TEBs [cells near the nascent lumen, body cells or basal (cap) cells] (n=3 mice, one TEB and one duct per mouse). A two-tailed paired Student's t-test was used to analyse the differences between duct luminal versus duct basal cells, and a repeated measures one-way ANOVA with Tukey post hoc test was used to analyse differences between TEB near lumen versus TEB body versus TEB basal (cap) cells. (C) Mean fluorescence intensity of Pten amongst the different regions analysed. Lines connect ductal or TEB compartments from the same mouse. (D) Cytoplasmic:nuclear ratio of Pten fluorescence amongst the different regions analysed. Open symbols represent individual field of view, filled symbols represent the mean from all fields of view from a given mouse, and the line marks the mean from the three mice analysed.

Cells near the nascent lumen seemed to present higher overall levels of Pten in comparison to the majority of the cells within the TEB, namely body cells (Fig. 6C). Furthermore, Pten was found predominantly in the cytoplasm in luminal cells in the ducts and in cells near the nascent lumen in TEBs (Fig. 6D), whereas body cells in the TEB often presented a more even distribution of Pten between the cytoplasmic and nuclear compartments (Fig. 6D). Basal cells in ducts also presented a more even distribution of Pten across both compartments, whereas basal (cap) cells in TEBs most often presented a predominance of nuclear Pten (Fig. 6D).

The presence of Pten in the membrane is directly related to its phosphatase activity (Brito et al., 2015) and might therefore indicate not only that Pten activity is higher in ducts and near the nascent lumen, but also that this activity is predominantly polarized to the apical surface. The fact that cap cells, which are believed to be multipotent mammary stem cells (Paine and Lewis, 2017; Visvader and Stingl, 2014), presented high Pten levels in the nuclei might be related to the contribution of Pten to genomic stability (Ho et al., 2020), which would be particularly important for progenitor cells, as mutations in these cells would be passed on to all their descendant cells.

Pten inhibition leads to cell proliferation and loss of cell polarity in both developing and mature mammary epithelial 3D acini

The distinct levels and cell and tissue localization of Pten in ducts and in TEBs prompted us to use 3D acini cell culture models to evaluate the contribution of Pten to inducing and sustaining cell cycle arrest, cell polarity and lumen formation. Upon lrECM exposure in 3D, EpH4 cells give rise to quiescent polarized acini with a central lumen, recapitulating many cellular and molecular events observed during mammary gland morphogenesis in vivo (Xu et al., 2009).

To better understand the role of Pten in the interplay between quiescence acquisition and maintenance, as well for establishing and sustaining apicobasal polarity and tissue architecture, we pharmacologically inhibited Pten in 3D epithelial acini at two distinct stages: during development and in mature structures (Fig. 7A,F).

Fig. 7.

Pten inhibition affects quiescence and polarity in both developing and mature mammary 3D acini. (A) Schematic representation of the ‘on top’ 3D acinogenesis model used in experiments depicted in B–E (figure created with Biorender.com). EpH4-ES-FUCCI cells were seeded on top of lrECM (Matrigel) bedding and treated or not with 2.5 µM bpV(pic) (Pten inhibitor, Pteni) on the day of seeding (Pteni d0) or when the 3D structures had already established quiescence at day 4 (Pteni d4) and imaged alongside the control at day 5. (B) Merged bright-field and fluorescence images of representative structures for each treatment (control, Ctrl; Pteni d0; and Pteni d4). Red: Cdt1–mCherry FUCCI probe (cells in G1); green: geminin–mCitrine FUCCI probe (cells in S/G2/M). Scale bar: 25 µm. (C) Cell cycle quantification based on the expression of FUCCI probes. The proportion of mCitrine-positive (mCit+) and mCherry-positive (mCh+) cells is shown for each condition (mean±s.e.m.; n=3). *P<0.05 (one-way ANOVA followed by Dunnett's multiple comparisons test). (D) Values of roundness of individual structures (each light-coloured point represents an individual 3D structure, darker points are the mean of a given biological replicate, and the line marks the mean) Total number of structures analysed per condition: Ctrl, 775; Pteni d0, 812; Pteni d4, 875 (from n=3). (E) Normalized area (relative to the control), plotted as log2 area (each light-coloured point represents an individual 3D structure, darker points are the mean of a given biological replicate, and the line marks the mean). Total number of structures analysed per condition: Ctrl, 775; Pteni d0, 812; Pteni d4, 875 (from n=3). (F) Schematic representation of the 3D lrECM in polyHEMA-coated plates acinogenesis model used in experiments depicted in G–I (figure created with Biorender.com). EpH4 cell clusters grown in polyHEMA-treated plates were subjected to lrECM treatment (Ctrl), along with pharmacological inhibition of Pten with 2.5 µM bpV(pic) during acinar morphogenesis (inhibitor added on the same day structures were treated with lrECM, Pteni d0) or when the structures were already mature (inhibitor added after 5 days of lrECM treatment, Pteni d5). Control and treated structures were collected and processed 6 days post lrECM treatment for immunofluorescence (IF) staining. (G) Confocal images of 3D structures stained for integrin α6 (red, basal marker), ZO-1 (green, apical marker) and Ki-67 (magenta, proliferation marker). Nuclei are stained using DAPI (blue). Scale bar: 10 µm. (H) Percentage of Ki-67-positive signal relative to the total area in each sample (mean±s.e.m., n=2). (I) Percentage of structures with lumen (mean±s.e.m.). Total number of structures analysed per condition: Ctrl, 1747; Pteni d0, 1610; Pteni d5, 1482 (from n=2). In D, E, H and I, statistical significance was evaluated using one-way ANOVA followed by Dunnett’s multiple comparisons test, with P<0.05 being considered significant.

Fig. 7.

Pten inhibition affects quiescence and polarity in both developing and mature mammary 3D acini. (A) Schematic representation of the ‘on top’ 3D acinogenesis model used in experiments depicted in B–E (figure created with Biorender.com). EpH4-ES-FUCCI cells were seeded on top of lrECM (Matrigel) bedding and treated or not with 2.5 µM bpV(pic) (Pten inhibitor, Pteni) on the day of seeding (Pteni d0) or when the 3D structures had already established quiescence at day 4 (Pteni d4) and imaged alongside the control at day 5. (B) Merged bright-field and fluorescence images of representative structures for each treatment (control, Ctrl; Pteni d0; and Pteni d4). Red: Cdt1–mCherry FUCCI probe (cells in G1); green: geminin–mCitrine FUCCI probe (cells in S/G2/M). Scale bar: 25 µm. (C) Cell cycle quantification based on the expression of FUCCI probes. The proportion of mCitrine-positive (mCit+) and mCherry-positive (mCh+) cells is shown for each condition (mean±s.e.m.; n=3). *P<0.05 (one-way ANOVA followed by Dunnett's multiple comparisons test). (D) Values of roundness of individual structures (each light-coloured point represents an individual 3D structure, darker points are the mean of a given biological replicate, and the line marks the mean) Total number of structures analysed per condition: Ctrl, 775; Pteni d0, 812; Pteni d4, 875 (from n=3). (E) Normalized area (relative to the control), plotted as log2 area (each light-coloured point represents an individual 3D structure, darker points are the mean of a given biological replicate, and the line marks the mean). Total number of structures analysed per condition: Ctrl, 775; Pteni d0, 812; Pteni d4, 875 (from n=3). (F) Schematic representation of the 3D lrECM in polyHEMA-coated plates acinogenesis model used in experiments depicted in G–I (figure created with Biorender.com). EpH4 cell clusters grown in polyHEMA-treated plates were subjected to lrECM treatment (Ctrl), along with pharmacological inhibition of Pten with 2.5 µM bpV(pic) during acinar morphogenesis (inhibitor added on the same day structures were treated with lrECM, Pteni d0) or when the structures were already mature (inhibitor added after 5 days of lrECM treatment, Pteni d5). Control and treated structures were collected and processed 6 days post lrECM treatment for immunofluorescence (IF) staining. (G) Confocal images of 3D structures stained for integrin α6 (red, basal marker), ZO-1 (green, apical marker) and Ki-67 (magenta, proliferation marker). Nuclei are stained using DAPI (blue). Scale bar: 10 µm. (H) Percentage of Ki-67-positive signal relative to the total area in each sample (mean±s.e.m., n=2). (I) Percentage of structures with lumen (mean±s.e.m.). Total number of structures analysed per condition: Ctrl, 1747; Pteni d0, 1610; Pteni d5, 1482 (from n=2). In D, E, H and I, statistical significance was evaluated using one-way ANOVA followed by Dunnett’s multiple comparisons test, with P<0.05 being considered significant.

In both cases, Pten inhibition led to increased proliferation, which was assessed via the expression of FUCCI sensors (Fig. 7B,C) or by Ki-67 (MKI67) staining (Fig. 7G,H). These data indicate that in mammary epithelial cells, Pten activity is necessary to induce quiescence during development and to sustain quiescence in mature structures (Fig. 7B,C,G,H). Structures treated earlier (and therefore for longer) with Pten inhibitor were larger and less spherical than those that were exposed to the inhibitor later, although in both conditions the inhibitor-treated structures were larger and more irregular than those observed in control conditions (Fig. 7B,D,E).

Differentiated EpH4 acini express apicobasal markers such as ZO-1 (apical marker; also known as TJP1) and integrin α6 (basal marker; also known as ITGA6) (Fig. 7G, left). Pten inhibition, either during development or in mature acini, unleashed proliferation and disrupted tissue architecture, as evidenced by the presence of irregular structures that lacked a lumen and had widespread membrane staining for ZO-1 and integrin α6 (Fig. 7B–D,G–I), consistent with a role of Pten in inducing and sustaining quiescence and tissue architecture.

By using a relevant cell culture model where, even in the presence of growth factors, exposure to a BM surrogate (Kleinman and Martin, 2005) triggers quiescence and differentiation in mammary epithelial cells, akin to what occurs in vivo (Fiore et al., 2017; Paine and Lewis, 2017; Sternlicht et al., 2006; Xu et al., 2010), we have provided a snapshot of the molecular landscape of cell cycle regulators in lrECM-induced quiescent cells, which in combination with microscopy data derived from fluorescent cell cycle indicators and perturbations made using specific inhibitors, allowed us to investigate in detail the signalling networks that might lay the molecular foundation for specific traits of quiescent cells.

For example, the complexity of alterations found in this molecular landscape helps to explain why overexpressing a CKI or downregulating cyclins and/or Cdks might lead to irreversible cell cycle arrest, not fully recapitulating the transient state of quiescence (Lim and Kaldis, 2012; Sang et al., 2008).

Also, in order to proliferate, cells need to grow throughout G1 until they achieve a specific target size (Gokhale and Shingleton, 2015; Lloyd, 2013; Vollmer et al., 2017). Thus, although cell growth and cell proliferation can be separable events, there is a high degree of coupling between cell size and cell cycle progression (Kafri et al., 2013). Mechanisms underlying cell size control are poorly understood (Lloyd, 2013), but mTOR is a clear positive regulator of cell growth (Kim and Guan, 2019). The reduced size observed in quiescent cells, along with reduced mTOR signalling upon lrECM treatment and induction of cell cycle arrest by mTORC1 and mTORC1/2 inhibitors, combined with previous findings (Kafri et al., 2013; Kim and Guan, 2019), indicate that reduced cell growth might be one of the mechanisms behind cell cycle arrest in BM-induced quiescence.

Another intriguing observation was the differential effect of Cdk2 and Cdk4 inhibition in cell proliferation and morphology, together with the interplay between cyclins, Cdks and CKIs during lrECM-induced quiescence. A few things need to be considered to better explore these data. First, in the classical model of cell cycle progression, in response to mitogenic stimuli during G1, cyclinD–Cdk4 complexes phosphorylate and inactivate Rb, which releases E2F transcription factors that, in turn, switch on the expression of a range of positive regulators of the cell cycle, including cyclinE, which in complex with Cdk2 further phosphorylates and inhibits Rb. This process culminates in Rb hyperphosphorylation, at the ‘restriction point’ of the cell cycle, after which cells are committed to proliferation and will progress through cell cycle regardless of mitogenic stimuli (Blagosklonny and Pardee, 2002; Gérard and Goldbeter, 2012; Pardee, 1974). It has been recently demonstrated, however, that molecular events occurring much earlier than Rb hyperphosphorylation, including the degree of Cdk2 activity in newly born cells, are the determinant factors behind cell cycle commitment (Spencer et al., 2013). Second, although the CKIs p21 (also known as CDKN1A) and p27 supress Cdk2 activity, they are required for the assembly of cyclinD–Cdk4 complexes. In this context, p21 is generally an inhibitor or a poor activator, whereas p27, depending on its phosphorylation status, can be either an inhibitor or a strong activator for cyclinD–Cdk4 complexes (Guiley et al., 2019; Ray et al., 2009). Thus, in addition to kinase activity, cyclinD–Cdk4–p27 complexes would contribute to cell cycle progression by titrating p27 away from Cdk2 complexes (Guiley et al., 2019; Ray et al., 2009).

Although a much more in-depth analysis of these mechanisms would be necessary to draw any conclusions, it is tempting to speculate that during lrECM-induced cell cycle arrest, inhibition of Cdk2 activity could be a core event for acquiring and sustaining the quiescent phenotype. LrECM-induced inhibition of Cdk2 activity, due to both reduced levels of cyclinE and Cdk2, along with increased p27, in a context where cyclinD1 and Cdk4 were also reduced, would allow for cell cycle arrest even in the presence of mitogens. Which of these events comes first, how this is regulated at the molecular level and whether this stands true for lrECM-induced quiescence would need extensive investigation and is beyond the scope of the present work. Moreover, although sufficient to map the overall molecular landscape of cells induced to quiescence by the ECM, our current bulk analysis relying on RT-qPCR and western blotting might not have completely uncovered important aspects of the decision signalling network made at the single-cell level. By employing single-cell level methodologies in future investigations, we could gain enhanced granularity, thereby elucidating cell subpopulations and distinct cellular dynamics concerning Cdks and other key signalling molecules.

The apparent discrepancy between the activation status of some upstream (Fak, Src and PI3K) and downstream (Akt, mTORC1/2 and MAPK) regulators of cell cycle progression could also be consistent with the idea that proliferative signals from the ECM are sensed by quiescent cells, but that proliferative signalling effectors are actively shut down by laminin-triggered alterations. Depending on spatiotemporal regulation, signalling integration and subcellular localization, Fak, Src and PI3K are involved in a range of cellular processes other than proliferation, such as cell survival (Aksamitiene et al., 2012), invasion (Levental et al., 2009), migration (Gehler et al., 2013), polarity (Liu et al., 2004) and lumen formation (Peng et al., 2015). Shutting down the proliferative signalling arms of these pathways might open space to the rise of other signalling routes and, for instance, result in a shift in cell phenotype towards differentiation. Our findings indicate that Pten might, at least in part, fulfil this role. Pten was upregulated at the protein level in quiescent cells (Fig. 2), and its inhibition led to increased proliferation, restored Akt–mTORC1/2 and MAPK signalling in the presence of quiescence-inducing signals from lrECM in mammary epithelial cells, and disrupted quiescence, polarity and tissue architecture in 3D acini (Figs 3, 4 and 7).

Pten is a well-known tumour suppressor, and its role in restraining cell proliferation has been extensively demonstrated in a wide range of tissues and cell types (Lee et al., 2018). The PI3K pathway is one of the most mutated pathways in cancer, with Pten figuring, in fact, as the most frequently altered PI3K pathway member (Millis et al., 2016). Pten loss has been associated with increased proliferation due to upregulation of PI3K–Akt–mTORC1/2 and MAPK signalling pathways (Gu et al., 1998; Worby and Dixon, 2014), but even discreet alterations in Pten levels can trigger dramatic changes in cell behaviour and increase the risk of developing cancer (Lee et al., 2018).

The mechanisms by which laminin upregulates Pten and whether this regulation follows the rules of dynamic reciprocity (Bissell et al., 1982) remain unclear, but, in 3D culture of mammary epithelial cells, Pten levels have been linked to increased dystroglycan receptor expression (Muschler et al., 2002), as well as to E-cadherin (CDH1) expression and localization (Fournier et al., 2008). Tissue architecture integrity and the levels and localization of Pten have been pointed out to exist in dynamic reciprocity (Fournier et al., 2008). We did find increased levels of dystroglycan receptor upon lrECM-induced quiescence (Fig. 2A,D,E). The dystroglycan receptor has two subunits encoded by a single transcript (Moore and Winder, 2012). The extracellular subunit α-dystroglycan is subjected to posttranslational modifications through glycosylation, which confer its ability to bind laminin (Cloutier et al., 2019; Pozzi and Zent, 2011). The intracellular subunit β-dystroglycan usually binds to α-dystroglycan and to cytoskeleton proteins, although it can also be found in the nucleus (Cloutier et al., 2019; Gracida-Jiménez et al., 2017). Dystroglycan receptors are associated with attenuation of integrin signalling (Ferletta et al., 2003), cell cycle arrest (Sgambato et al., 2004) and polarity (Muschler et al., 2002) in mammary epithelial cells treated with lrECM. Therefore, increased expression of Pten and dystroglycan receptor might cooperate to trigger and sustain quiescence in growth-suppressive microenvironments.

Although the BM is deposited mainly by basal cells, cells across the entire epithelial structure are under the influence of cues triggered by it, in both physiological and pathological conditions (Allinen et al., 2004; Gudjonsson et al., 2002). In this sense, correlations between BM thickness and some regulators of proliferation and migration, including nuclear actin (Spencer et al., 2011) and nuclear galectin-1 (Bhat et al., 2016), have been reported in the developing mammary gland, and our data indicate that Pten might also fall into this category. Pten displayed differential levels and staining patterns in TEBs versus ducts of the developing mammary gland in vivo, which correlated with the degree of laminin deposition in these structures (Figs 5, 6). Myoepithelial cells are organized as a network that does not cover luminal cells completely (Gieniec and Davis, 2022), allowing for luminal cell contact with the BM in ducts, which could be linked to the overall higher levels of Pten observed in ducts in comparison to levels in TEBs. Interestingly, cells surrounding the nascent lumen in TEBs exhibited apicolateral Pten staining, the same pattern found in differentiated luminal cells in the ducts (Figs 5, 6). These data might indicate that in TEBs, cells surrounding the lumen already show some degree of polarization and maybe even differentiation, whereas cells in other areas do not. It has been shown that even though the TEB is a highly proliferative structure within the developing mammary epithelia, proliferation is not observed in luminal epithelial cells surrounding the nascent lumen (Ewald et al., 2012), and our data indicate that Pten might have a role in this phenomenon. These cells near the nascent lumen in TEBs clearly are not in contact with the BM; thus, other cues, such as signalling through cell–cell adhesions, might be involved in their differentiation programme (Runswick et al., 2001). Based on these observations, it is tempting to speculate that mammary gland differentiation involves two parallel and converging events: one occurring at the luminal interface and one coming from the basal surface, both involving Pten upregulation, with Pten polarization to the apicolateral region possibly being involved in commitment to the luminal lineage.

Pten in the apicolateral membrane, either in ducts or in cells surrounding the lumen in TEBs, strongly colocalized with the actin belt (Fig. 6), resembling the staining of other classical apicolateral markers of polarized cells, such as ZO-1 and E-cadherin (Laprise et al., 2002; Weaver et al., 1997). Whether cell differentiation precedes or follows Pten apicolateral polarization remains to be experimentally demonstrated. Nevertheless, Pten most likely acts on the onset of polarity acquisition rather than being a late factor recruited to mature junctional complexes.

It has been shown that Par3 (also known as PARD3), which has a core role in the establishment of cell polarity, and E-cadherin, a key regulator of epithelial tissue integrity, are both able to recruit Pten to cell–cell junctions, which is required for cell differentiation (Feng et al., 2008; Fournier et al., 2009). Polarization of Pten to the apicolateral membrane might contribute for the establishment of lipid domains (Comer and Parent, 2007; Pinal et al., 2006) and for the compartmentalization of PI3K activity in quiescent, differentiated cells (Fournier et al., 2009). In fact, PIP2, the lipid product of Pten phosphatase activity, has been proposed to be an apical marker itself (Comer and Parent, 2007; Martin-Belmonte et al., 2007). EpH4 acini display active PI3K polarized to the basolateral membrane, and activation of the PI3K–Rac1 axis is required for complete differentiation and milk protein synthesis (Xu et al., 2009, 2010), thus, polarized activity of PI3K to the basal surface might trigger survival and differentiation, rather than proliferation.

Our data support a role for Pten not only in proliferation inhibition, but also in lumen assembly and maintenance in the developing mammary gland, and they are consistent with previous findings (Berglund et al., 2013; Martin-Belmonte et al., 2007). Apicolateral Pten has been reported in several 3D polarized epithelial structures, such as Madin–Darby canine kidney (MDCK) cell and T84 (colon) cell cysts (Martin-Belmonte et al., 2007), and the chick epiblast (Leslie et al., 2007), as well as in human and murine mammary epithelial cells (Berglund et al., 2013; Fournier et al., 2008); however, to our knowledge, the results we present here are the first report of apicolateral Pten in the developing mammary gland in vivo.

Facing these results, two old but not fully understood questions come to mind. What comes first: polarity or cell cycle arrest? And at what point does Pten come into play? Cell polarity and cell cycle arrest are interrelated, PI3K-modulated, but molecularly distinct events, and inhibition of the PI3K downstream effectors Akt and Rac1 has been linked to proliferation arrest and polarity, respectively (Liu et al., 2004). Similarly, by using 3D acini, Leslie and collaborators (Berglund et al., 2013) have shown that Pten requires both lipid phosphatase and protein phosphatase activities to control lumen formation through a mechanism that does not correlate with its ability to control Akt. Interestingly, in murine NMuMG acini, despite displaying increased cell proliferation, Pten knockdown or overexpression of p110α (PIK3CA) oncogenic mutants disrupts lumen formation, whereas a constitutively activated Akt does not (Berglund et al., 2013). These data further support the notion that different pathways downstream of PI3K govern the regulation of lumen formation and proliferation, and that Pten might be a key regulator of both. In this sense, our data in 3D acini models provide further evidence that Pten simultaneously coordinates proliferation and normal tissue architecture during acini development, and sustains quiescence and tissue architecture in mature, quiescent acini (Fig. 8).

Fig. 8.

Proposed regulatory mechanisms underlying the mammary epithelial cell quiescence–proliferation decision, highlighting the role of Pten. (A) Molecular landscape of crucial cell cycle and proliferation regulators in laminin-induced quiescent mammary epithelial cells. Names of molecules classically associated with cell cycle progression are depicted in green, whereas names of molecules classically associated with cell cycle arrest are in red. Blue boxes: molecules that were found to be downregulated during lrECM-induced quiescence; red boxes: molecules that were found to be upregulated during lrECM-induced quiescence; grey boxes: molecules that exhibited no alteration during lrECM-induced quiescence; white boxes: not assessed; box with dashed outline: protein complex, not assessed. α-DG, α-dystroglycan; β-DG, β-dystroglycan; p, phospho (B) Interplay between Pten and laminin-111 during mammary gland development to coordinate differentiation and cell cycle arrest. Figure created with Biorender.com.

Fig. 8.

Proposed regulatory mechanisms underlying the mammary epithelial cell quiescence–proliferation decision, highlighting the role of Pten. (A) Molecular landscape of crucial cell cycle and proliferation regulators in laminin-induced quiescent mammary epithelial cells. Names of molecules classically associated with cell cycle progression are depicted in green, whereas names of molecules classically associated with cell cycle arrest are in red. Blue boxes: molecules that were found to be downregulated during lrECM-induced quiescence; red boxes: molecules that were found to be upregulated during lrECM-induced quiescence; grey boxes: molecules that exhibited no alteration during lrECM-induced quiescence; white boxes: not assessed; box with dashed outline: protein complex, not assessed. α-DG, α-dystroglycan; β-DG, β-dystroglycan; p, phospho (B) Interplay between Pten and laminin-111 during mammary gland development to coordinate differentiation and cell cycle arrest. Figure created with Biorender.com.

Taken together, the data we present here provide evidence that laminin, found in the BM, induces quiescence and differentiation in mammary epithelial cells by differentially modulating the expression and activity of cell cycle regulators at the transcriptional and protein levels. This panorama, along with the extensive previous work done by many others, provides some hints of the molecules, pathways and mechanisms involved in microenvironmental control of quiescence in higher organisms, which could be explored in studies to come. The notion that lrECM-triggered quiescence involves several pathways and has many layers of regulation provides an explanation for the robustness of this system in suppressing epithelial cell proliferation, which could ultimately be linked to cancer incidence being lower than expected (Bissell and Hines, 2011) and even for the need of more than one perturbation to bypass microenvironmentally induced suppression of cancer development (Knudson, 2001). In this sense, disruptions in master regulators such as Pten, which regulate several signalling pathways and phenotypical features required for tissue homeostasis and tumour suppression, could indeed be drivers of malignant transformation.

Reagents

Primary and secondary antibodies used in this work are listed in Table S1 and Table S2, respectively. Table S3 shows information on the pharmacological inhibitors used. Additional reagents used are listed in Table S4 and plasmids are listed in Table S6.

Cell culture

Murine mammary epithelial cells EpH4 were a gift from Mina J. Bissell (Lawrence Berkeley National Laboratory, Berkeley, CA, USA) and were routinely tested for mycoplasma contamination and verified to express specific markers for mammary epithelial cells. The parental EpH4 and the EpH4-ES-FUCCI and EpH4-DHB-mVen-mCh-Cdk4.KTR-H2B-mTurq (expressing DHB–mVenus, mCherry–Cdk4KTR and histone H2B–mTurquoise) sublines were routinely grown in DMEM/F12 containing 2% FBS, 5 µg/ml insulin and 50 µg/ml gentamycin at 37°C and 5% CO2 in a humidified incubator. Cells were passaged when necessary (every 2–3 days) using trypsin-EDTA (0.05%). The cell sublines generated in this work can be provided upon request.

Laminin-rich ECM overlay differentiation assay

EpH4 cell quiescence induction and lactogenesis differentiation upon overlay treatment with lrECM has been described previously (Bridgewater et al., 2017; Spencer et al., 2011). Briefly, cells were seeded at 2000 cells/cm2 (unless otherwise specified) in uncoated plastic wells or culture dishes. The following day (day 2), the medium was changed for GIH medium (DMEM/F12, 50 µg/ml gentamycin, 5 µg/ml insulin, 1 µg/ml hydrocortisone). Then, 24 h later (day 3), cells were treated or not with 2% growth factor-reduced Matrigel (lrECM) and/or 3 µg/ml prolactin (Prl and lrECM+Prl conditions), and where appropriate, with specific inhibitors. Cells were harvested or imaged 24–48 h post treatment (day 4 or day 5, depending on the assay). Fig. 1A depicts a general workflow for the lrECM overlay model used here.

Cell cycle analysis via propidium iodide staining and flow cytometry

EpH4 cells were subjected or not to lrECM overlay treatment for 48 h. Cells were then detached and pelleted by centrifugation, washed with cold PBS, fixed in 70% cold ethanol and stored for 24 h at −20°C. Fixed cells were incubated with RNase A (0.2 mg/ml) at 37°C for 30 min and stained with propidium iodide (PI) (1 μg/ml). DNA content of cells (20,000 events) was analysed using an ACCURI C6 flow cytometer. Data were plotted using GraphPad Prism, and statistical differences between treatments were assessed by two-tailed unpaired Student's t-test. P<0.05 was considered significant.

Cell density and live–dead assay

EpH4 cells were subjected or not to lrECM overlay treatment for 48 h. Cells were then stained with the DNA-binding dyes PI (5 μg/ml) and Hoechst 33342 (5 μg/ml) for 15 min. Cells were then imaged on a customized TissueFAXS i-Fluo system (TissueGnostics) mounted on a Zeiss AxioObserver 7 microscope (Zeiss), using a 20× Plan-Neofluar (NA 0.5) objective and an ORCA Flash 4.0 v3 camera (Hamamatsu). 8×8 adjacent microscope fields of view were acquired per well using automated autofocus and image acquisition settings. Thousands of cells per sample were analysed using a customized pipeline on StrataQuest software (TissueGnostics), which can be made available upon request. Details of parameters used for nuclei segmentation and detection on StrataQuest were as described previously (Russo et al., 2021). Individual nuclei for all cells were detected in the Hoechst channel, which was used to build a nuclear mask where the PI signal (dead cells) was detected. The relative cell density (number of cells based on the Hoescht signal/total area imaged) was calculated for control (normalized to 1) or lrECM-treated cells. The percentage of dead cells (PI-positive cells) was calculated in relation to the total number of cells (Hoechst-positive cells). Data were plotted using GraphPad Prism and statistical differences between treatments were assessed by two-tailed unpaired Student's t-test. P<0.05 was considered significant.

FUCCI-based cell cycle analysis

For these experiments, EpH4 cells constitutively expressing the fluorescence cell cycle indicator FUCCI (Sakaue-Sawano et al., 2008) were used. EpH4-ES-FUCCI cells were generated by transfecting EpH4 cells with the ES-FUCCI plasmid (Table S6), followed by hygromycin B selection (500 µg/ml, 10 days) and aseptic cell sorting of fluorescent cells.

EpH4-ES-FUCCI cells were seeded into 96-well plates and subjected to the lrECM overlay assay (control and lrECM). At 48 h post treatment, cells were incubated with Hoechst 33342 (5 μg/ml) for 15 min and imaged on a Leica DMi8 wide-field fluorescence microscope (E-signal lab, University of São Paulo) using the LasX Navigator Application (Leica Microsystems). The DMi8 microscope was coupled with an incubation system that allowed live-cell imaging at 37°C and 5% CO2. Each well was imaged in 3×3 adjacent microscope fields, using the 10× objective and specific fluorescence filters (DAPI for Hoescht, Txr for mCherry and YFP for mCitrine). Images were analysed on StrataQuest software (TissueGnostics) using a customized pipeline, which can be provided upon request. Details of parameters used for nuclei segmentation and detection on StrataQuest were as described previously (Russo et al., 2021). In brief, nuclei were detected in the Hoechst channel and a mask was created, allowing the measurement of the mean fluorescent intensity signal of the FUCCI probes (Cdt1–mCherry and geminin–mCitrine) for thousands of cells per sample. The percentage of mCherry-positive cells (cells in G1), mCitrine-positive cells or double-positive cells (cells in S/G2/M), or double-negative cells (cells that had just finished M) was determined for each well. Data were plotted using GraphPad Prism, and statistical differences between treatments for each cell cycle phase were assessed by two-tailed unpaired Student's t-test. P<0.05 was considered significant. These experiments were performed in triplicate wells in three biological replicates.

Cdk2 and Cdk4 activity

Cdk2 and Cdk4 activities were evaluated using fluorescent reporters (Spencer et al., 2013; Yang et al., 2020). EpH4-DHB-mVen-mCh-Cdk4.KTR-H2B-mTurq cells were generated by simultaneous infection of EpH4 cells with lentiviruses containing either DHB-mVen-mCh-Cdk4.KTR or the H2B-mTurq constructs in the presence of 8 µg/ml polybrene. Lentiviruses were previously packaged by HEK293 FT cells (gift from Mina J. Bissell, Lawrence Berkeley National Laboratory, Berkeley, CA, USA) transfected with polyethylenimine (PEI), the packaging plasmids psPAX2 and pMD2.G and pLenti-DHB-mVenus-p2a-mCherry-Cdk4KTR or CSII-EF1-H2B-mTurquoise (Table S6), following instructions from Addgene. Cells were then subjected to aseptic sorting for selection of double-infected cells (expressing mVenus and mCherry, as well as mTurquoise).

EpH4-DHB-mVen-mCh-Cdk4.KTR-H2B-mTurq cells were plated into 96-well plates and subjected to lrECM overlay assay (control and lrECM). At 48 h post treatment, cells were imaged on a Leica DMi8 wide-field fluorescence microscope (E-signal lab, University of São Paulo) using the LasX Navigator Application (Leica Microsystems). Each well was imaged in 3×3 adjacent fields of view, using the 10× objective and specific fluorescence filters (CFP for mTurquoise, Txr for mCherry and YFP for mVenus). Cdk2 and Cdk4 activities were calculated based on the guidelines from Spencer et al. (2013) and Yang et al. (2020), the developers of these specific probes. Images were analysed on StrataQuest software (TissueGnostics) using a customized pipeline, also available upon request. Details of parameters used for nuclei segmentation and detection on StrataQuest were as described previously (Russo et al., 2021). In brief, nuclei were detected in the mTurq channel and a nuclear mask was created. In order to measure the cytoplasmic intensity of Cdk2 and Cdk4 probes, another mask, corresponding to a cytoplasmic ring was created (2 µm around the nucleus, or less if touching another cytoplasmic ring or nucleus, starting 0.5 µm away from the nucleus). The mean intensity of fluorescence signals (mVenus for Cdk2 and mCherry for Cdk4) were obtained for the nucleus and for the cytoplasmic ring for thousands of individual cells per sample. For each cell, Cdk2 activity was calculated as the ratio of the mean fluorescent intensity of mVenus in the cytoplasmic ring and in the nucleus. Because Cdk2 can to some extent phosphorylate the Cdk4 probe (Yang et al., 2020), Cdk4 activity was calculated as the cytoplasmic:nuclear ratio of the mean fluorescent intensity of mCherry, minus the Cdk2 activity in that cell, multiplied by a correction factor that was previously calculated by Yang et al. (2020): Cdk4 activity=[(mCherry cytoplasmic/mCherry nuclear) – Cdk2 activity]×0.05. Data were plotted as a ‘Superplot’ (Lord et al., 2020) using GraphPad Prism, and statistical differences between treatments were assessed by two-tailed unpaired Student's t-test. P<0.05 was considered significant. These experiments were performed in triplicate wells in three biological replicates.

Inference of cell size via flow cytometry and microscopy

EpH4 cells were subjected to lrECM overlay assay for 48 h, and cell size was estimated according to the forward scatter area (FSC-A) determined using a BD Accuri C6 flow cytometer. As freely cycling cells (control) would have significantly more cells in S/G2/M (which are larger in size) than quiescent (lrECM-treated) cells, in order to do a fair comparison, both control and lrECM-treated cells were gated in G1 according to DNA content (assessed using PI staining, see above).

Similarly, EpH4-FUCCI cells were subjected to lrECM overlay assay for 48 h, counterstained with Hoechst 33342 (5 µg/ml, 15 min) and imaged (as described in the ‘FUCCI-based cell cycle analysis’ section above). Cell size was estimated according to the area of the nucleus (µm2). Images were analysed on StrataQuest software (TissueGnostics), and once again, only cells in G1 (mCherry positive) were considered for this analysis.

Data were plotted using GraphPad Prism, and statistical differences between treatments were assessed by two-tailed unpaired Student's t-test. P<0.05 was considered significant. These experiments were performed in three biological replicates.

For the analysis of the correlation between Cdk activity and nuclear area (Fig. S2), the data regarding Cdk4, Cdk2 and nuclear area were obtained from the experiments shown in Fig. 1G,H, plotted using the Python package Seaborn (version 0.12.2), and the Pearson's correlation r value was calculated using GraphPad Prism.

Gene expression analysis

EpH4 cells were subjected to lrECM overlay and/or prolactin treatment for 48 h. Cells were then harvested for RNA extraction, reverse transcription and RT-qPCR. This experiment was performed in three biological replicates.

Gene expression was assessed using RT-qPCR, following the MIQE guidelines (Bustin et al., 2009). Briefly, total RNA was extracted using an RNeasy kit, quantified and treated with a Turbo DNAse kit according to the manufacturer's instructions. DNAse-treated RNA was reserve transcribed into cDNA using the Superscript II reverse transcriptase, oligoDT and random hexamer primers (Thermo Fisher Scientific), following the manufacturer's guidelines.

RT-qPCR was performed in triplicates using 5 ng of cDNA, Power SYBR Green PCR Master Mix and specific primers (listed in Table S5) into a 10 µl reaction. The reactions were run in the AB-7500 real-time thermocycler (Applied Biosystems) using standard settings. B2m was used as the endogenous control, and mRNA fold changes (FCs) were calculated according to the Pfaffl method (Pfaffl, 2001).

Statistical analysis was carried out on GraphPad Prism using one-way ANOVA followed by Dunnett's multiple comparison test (comparing all samples to the control; P<0.05 was considered significant). Data were plotted as log2FC and –log10P-value using the online software Morpheus (Broad Institute; https://software.broadinstitute.org/morpheus/).

Protein expression analysis

EpH4 cells were subjected to lrECM overlay and/or prolactin treatment for 48 h. Cells were then lysed with boiling 1× Laemmli buffer (62.5 µM Tris, 2% SDS, 10% glycerol), and the obtained lysate was further incubated at 100°C for 15 min and stored at −20°C. Protein was quantified using the DC Protein Assay kit (Bio-Rad) according to the manufacturer's instructions.

The levels of specific proteins were then assessed by western blotting. For loading the gel, 1 µl of 0.15% Bromophenol Blue in 2-mercaptoethanol was added to 20 µg of protein in 1× Laemmli buffer for a total volume of 30 µl. Samples were boiled at 100°C for 10 min and run in on Tris–glycine polyacrylamide gels (8%, 10%, 12% or 15%, depending on the molecular mass of the protein of interest). Resolved proteins were transferred overnight onto a PVDF membrane (0.22 µm; Millipore) followed by 30 min incubation in blocking buffer (TBS, 0.2% Tween-20 with 5% BSA). Membranes were incubated overnight in blocking buffer containing primary antibodies (listed in Table S1), followed by incubation with the appropriate HRP-conjugated second antibody (listed in Table S2) for 1 h at room temperature. HRP was detected by SuperSignal West Dura Extended Duration Substrate (Thermo Fisher Scientific) and the chemiluminescence signal was captured with a ChemiDoc MP Imaging System (Bio-Rad).

When necessary, membranes were subjected to a stripping protocol to remove antibodies and allow the reprobing with antibodies against another protein (this was used particularly to assess the total levels of a given protein after assessing its phosphorylated levels). Stripping consisted of 2×10 min in distilled water, followed by 1 min in stripping buffer (25 mM glycine, 1% SDS, pH 2) and 4×5 min in TBS containing 0.2% Tween-20. Membranes were then blocked for 30 min and subjected to standard primary antibody and HRP-conjugated secondary antibody incubation and detection.

For the relative quantification of protein levels, the optical density of each band was determined using ImageJ (NIH, Bethesda, MD, USA). The endogenous control (lamin B or α-tubulin, depending on the experiment) was also quantified, and the relative expression was determined as the ratio between target protein level and the endogenous control. For phosphorylated proteins, we used the ratio of phosphorylated protein to total protein to endogenous control. Phospho-Rb and phospho-PI3K (p55) were normalized only against the endogenous control, as we could not assess the total levels of these proteins.

Data were analysed using GraphPad Prism. Fold change differences of a given protein between two groups were evaluated by two-tailed unpaired Student's t-test (P<0.05 was considered significant). The obtained data were compiled and displayed as volcano plots (log2FC versus –log10P-value). Comparisons between multiple groups were performed by one-way ANOVA followed by Tukey post-hoc test (P<0.05 was considered significant). This experiment has at least three biological replicates for each protein and condition analysed. Images of uncropped membranes can be found in Figs S4 and S5.

Treatment with inhibitors and live-cell imaging for FUCCI-based cell cycle analysis

EpH4-ES-FUCCI cells were seeded into 96-well plates (Corning) at a density of 2000 cells/well. The following day, the medium was changed to GIH without Phenol Red. Then, 24 h later, cells were treated or not with 2% Matrigel and/or specific inhibitors (see Table S3 for details and concentrations). At 24 h post treatment, cells were incubated with Hoechst 33342, imaged and analysed as described in the ‘FUCCI-based cell cycle analysis’ section above. The relative cell density (as described in the ‘Cell density and live–dead assay’ section above) was also determined for each well.

Data were analysed using GraphPad Prism. Relative cell density (normalized to the density of control or lrECM untreated cells) was analysed by one-way ANOVA followed by multiple comparisons (all conditions versus control) using the two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli (q<0.05 was considered significant). Differences in cell cycle based on the FUCCI probe expression were assessed by two-way ANOVA followed by multiple comparisons (all conditions versus control, for G1, S/G2/M and just finished M/early G1) using the two-stage linear step-up procedure of Benjamini, Krieger and Yekutieli (q<0.05 was considered significant). These experiments were performed at least two times with two technical replicates (two wells/condition).

Treatment with Pten inhibitor and analysis of downstream pathway activation

EpH4 cells were subjected to lrECM overlay assay with or without concomitant treatment with 2.5 µM bpV(pic) Pten inhibitor [control, Pten inhibitor (Pteni), lrECM and lrECM+Pteni]. At 24 h post treatment, cells were harvested for protein extraction and western blot analysis of protein expression. Data were analysed on GraphPad Prism using one-way ANOVA followed by Tukey's multiple comparisons test. These experiments were performed at least three times.

Immunofluorescence of murine mammary tissue sections

The mammary fat pads were harvested from 5-week-old female Balb/C mice obtained from Rede de Bioterios – University of São Paulo, and the experimental procedures involved in this work were approved by the Ethics Committee for Animal Welfare from the Institute of Chemistry, University of São Paulo (ethics number #218/2022).

Mammary fat pads were embedded in Tissue-Tek O. C. T., snap-frozen and stored at −80°C. The samples were sliced at a thickness of 25 µm using a cryostat (Leica). Slices were collected in StarFrost slides, dried briefly and were stored at −80°C until used.

For immunofluorescence staining, slices were thawed, fixed in 4% paraformaldehyde (PFA) for 10 min and quenched in PBS containing 0.3 M glycine. Antigen retrieval was performed by applying boiling Tris-EDTA (10 mM Tris, 1 mM EDTA, pH 9) directly onto the slides thrice with 5 min interval. Slices were then permeabilized in PBS containing 0.5% Triton X-100 (PBS-T) for 20 min, blocked in undiluted Odyssey Blocking buffer (LI-COR) containing 10% goat serum for 1 h at room temperature and incubated with primary antibodies (anti-Pten, anti-laminin-111; Table S1) overnight at 4°C. The following day, slides were washed and incubated with Alexa Fluor-conjugated secondary antibodies (Table S2) and, when pertinent, with Phalloidin–Alexa Fluor 647 (1:50) for 45 min at room temperature. Slides were counterstained with DAPI (0.5 µg/ml), mounted with Prolong Diamond antifade reagent, sealed and left overnight at 4°C to cure.

Mammary tissue section imaging and analysis

Epithelial structures of interest (TEBs and ducts) were either imaged with a Leica DMi8 wide-field fluorescence microscope (E-signal lab, University of São Paulo) or a Zeiss LSM880 Airyscan (INFABIC – National Institute of Science and Technology on Photonics Applied to Cell Biology at the State University of Campinas). Several ducts and TEBs from mammary glands obtained from at least three mice were used for these analyses.

Images obtained with the Leica microscope were taken using the 63×/1.4NA oil objective in z-stacks and adjacent fields (to cover larger structures comprising whole ducts and/or TEBs) using the LasX Navigator Application (Leica microsystems) in three channels (DAPI for nuclei, FITC for Pten and TXR for laminin-111). Images were initially processed in the LasX software, where adjacent fields were merged and 3D-blind deconvoluted. Five sequential z-stacks were then combined to obtain the maximal projection. The resulting images were used to analyse the association between laminin content and Pten levels in ducts and TEBs using a customized pipeline on StrataQuest software (TissueGnostics). Details of parameters used for nuclei segmentation and detection on StrataQuest were as described previously (Russo et al., 2021), and for this specific analysis, nuclei detection and segmentation were manually corrected for each image. In brief, images of ducts or TEBs were analysed separately; nuclei were detected in the DAPI channel, and the mean intensity of Pten signal was obtained for each cell in an area comprised by the nucleus and the cytoplasm, which was estimated by growing outward from the nuclear area until neighbouring areas touched each other or until the fluorescence signal of Pten reached background levels (a similar approach to estimate cell boundaries was used for ADP ribose signal by Russo et al., 2021). Total laminin-111 fluorescence intensity (sum of intensity) was obtained for the entire field of interest (epithelial structure) and this value was divided by the total number of cells detected in that field (mean laminin-111 fluorescence). The average of the mean fluorescence intensity of Pten was plotted against the associated mean laminin-111 intensity for that field using GraphPad Prism, and data were analysed by linear regression and Pearson's correlation. Differences between Pten intensity in ducts versus TEBs were analysed by two-tailed unpaired Student's t-test.

Images obtained using the Zeiss LSM880 Airyscan were taken using the 63×/1.4NA oil objective, and fluorescence was captured in four channels using the following laser lines for excitation: 405 nm (DAPI), 488 nm (Pten, Alexa Fluor 488), 543 nm (laminin-111, Alexa Fluor 546) and 633 nm (Phalloidin, Alexa Fluor 647). The detection spectra were adjusted manually for each channel to avoid crosstalk between the different channels. Both confocal and zoomed (3.4 zoom factor) super-resolution (Airyscan) images were obtained for ducts and TEBs at different focal planes, corresponding to the basal domain (closer to the laminin-rich BM) and apical domain (closer to the lumen).

Further analyses regarding to the fluorescence levels of Pten in different regions of the epithelial structure (Fig. 6C,D) were performed on ImageJ using confocal images from three different mice (one duct and one TEB per mouse). Briefly, regions of interest (ROIs) corresponding to luminal or basal cells in ducts and to cells surrounding the nascent lumen, body cells or basal (cap) cells in TEBs were manually assigned to ducts and TEBs, based on the position of the BM or lumen in these structures. Initially, the mean fluorescence intensity for each ROI was obtained, and the mean intensity of all ROIs for the same region was calculated for each mouse. To calculate the cytoplasmic/nuclear ratio, we used the DAPI channel to build a mask that was used to obtain the mean fluorescence intensity of Pten in the nucleus and outside the nucleus (cytoplasm). We then calculated the cytoplasmic:nuclear ratio for each ROI and the average for each mouse. Data were plotted in GraphPad Prism. Data from luminal versus basal cells in ducts were compared by two-tailed paired Student's t-test (P<0.05 considered significant). In TEBs, comparisons between cells near the nascent lumen versus body cells versus basal cells were conducted by repeated measures one-way ANOVA followed by Tukey's post-hoc test (P<0.05 was considered significant).

Analysis of quiescence and polarity in 3D acini upon Pten inhibition

Assaying proliferation, roundness and size using EpH4-ES-FUCCI cells in an ‘on top’ 3D acinogenesis model

EpH4-ES-FUCCI cells (5×103 cells/cm2) were plated into 24-well plates on top of a bedding of Matrigel as previously described (Mroue and Bissell, 2012). Wells were treated with 2.5 µM Pten inhibitor bpV(pic), either on the day of seeding (day 0 and again at day 2 and day 4) or when quiescent 3D structures were already established (day 4). On day 5, 3D structures were incubated with Hoechst 33342 and imaged in z stacks using a 10× objective in a fluorescence microscope (Leica DMI8 wide-field microscope; imaging mCherry, mCitrine and Hoechst). A general workflow for the ‘on top’ 3D model is shown in Fig. 7A. Representative structures were then imaged at 40×. Image analysis was conducted using the LASX application (Leica). Briefly, images were deconvoluted and the maximum-intensity projection image was obtained. Structure size and roundness (0–1, where 1 represents a perfect circle) were obtained for individual structures, based on a mask built using the Hoechst channel. FUCCI-based cell cycle was calculated according to the mCherry and mCitrine areas in each field. These experiments were performed three times in duplicate wells, and three microscope fields per well at 10× were used for image analysis. Data were plotted in GraphPad Prism and analysed by one-way ANOVA followed by Dunnett's multiple comparisons test between all groups (Control, Pten inhibitor at day 0 and Pten inhibitor at day 4).

Onset and maintenance of apicobasal polarity and lumen

EpH4 cells were seeded in low-adherence polyHEMA-coated plates (30,000 cells/cm2), incubated for 48 h to allow for cell clusters to be formed and replated in medium supplemented or not with 4% Matrigel. Plates were coated in-house with polyHEMA at 0.25 mg/cm2 (Xu et al., 2009). When indicated, cell clusters were treated with 2.5 µM Pten inhibitor bpV(pic) either on the day of seeding (day 0) or when mature acini were already present (day 5). On day 6, 3D structures were fixed and subjected to immunofluorescence staining for ZO-1 (apical marker), α6 integrin (basal marker), Ki-67 (proliferation marker) and counterstained with DAPI (nuclei). A general workflow for the 3D lrECM in polyHEMA plates is shown in Fig. 7F. Briefly, structures were fixed for 10 min by adding 32% PFA straight into the culture well (for a final concentration of 4%). Structures were then collected into microcentrifuge tubes, spun, resuspended and washed twice in PBS containing 0.3 M glycine (quenching), permeabilized with 0.5% Triton X-100 in PBS for 20 min and blocked in 10% goat serum in Odyssey blocking buffer. Structures were then incubated overnight with primary antibodies diluted in Odyssey blocking buffer, washed three times with PBS and incubated with Alexa Fluor-conjugated secondary antibodies for 45 min (details of primary and secondary antibodies can be found in Tables S1 and S2), washed twice in PBS, incubated with DAPI for 5 min and mounted on glass slides using Prolong Diamond. All centrifugation steps were performed at 200 g for 3 min, and all tubes and tips used were previously rinsed with 5% BSA in PBS. These experiments were performed in two biological replicates.

Proliferation was estimated by the ratio between the area of Ki-67-positive cells and total area (DAPI). Slides were scanned with a Leica DMi8 wide-field microscope at 10×/0.3 NA (200 fields per slide), using the LASX Navigator application (Leica). Image analysis was conducted using the LASX application (Leica), where the total area for Ki-67 and DAPI were obtained and used to calculate the ratio between Ki-67 (proliferative cells) and DAPI (all cells) for a given slide.

The presence of lumen was assessed by manually scanning and counting the number of structures with lumen and the total number structures, using a 40×/0.6 NA objective with a Leica DMi8 wide-field microscope. The percentage of structures with lumen was then calculated for each condition. At least 600 structures per slide were analysed. The data regarding proliferation and lumen were then plotted and analysed in GraphPad Prism using one-way ANOVA followed by Dunnett's multiple comparisons test.

Representative structures were imaged with a Leica SP8-STED FALCON microscope using the confocal mode, a 63×/1.4 NA oil objective and using the following laser lines for excitation: 405 nm (DAPI), 488 nm (Ki67, Alexa Fluor 488), 561 nm (integrin α6, Alexa Fluor 546), 633 nm (ZO-1, Alexa Fluor 647). The detection range for each emission spectrum was adjusted manually to prevent signal overlap.

The authors thank Hugo Armelin (Institute of Chemistry, University of São Paulo, Brazil), Patricia Gama (Institute of Biological Sciences, University of São Paulo, Brazil), William Festuccia (Institute of Biological Sciences, University of São Paulo, Brazil), Cyrus Ghajar (Fred Hutchinson Cancer Center, USA) and Sabrina Spencer (University of Colorado Boulder, USA) for the kind donation of antibodies, reagents and plasmids. We thank the National Institute of Science and Technology on Photonics Applied to Cell Biology (INFABIC) at the State University of Campinas for access to equipment and assistance; INFABIC is co-funded by FAPESP (#2014/50938-8) and CNPq (#465699/2014-6). The authors are also grateful to Nicolas Hoch and Deborah Schechtman for scientific discussions, advice on image analysis and insightful inputs.

Author contributions

Conceptualization: R.T., A.B.-C.; Methodology: R.T.; Validation: R.T.; Formal analysis: R.T., A.M.R., A.C.M., A.B.-C.; Investigation: R.T., A.M.R., A.C.M.; Data curation: R.T.; Writing - original draft: R.T.; Writing - review & editing: R.T., A.B.-C.; Visualization: R.T., A.B.-C.; Supervision: A.B.-C.; Project administration: R.T., A.B.-C.; Funding acquisition: A.B.-C.

Funding

This work was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP; 2014/10492-0 and 2019/26767-2) and Instituto Serrapilheira. The Leica DMi8 microscope used in this study was obtained with funding from FAPESP (2015/02654-3) and the SP8-STED FALCON Leica confocal microscope was funded by a Financiadora de Estudos e Projetos (FINEP) grant (0424/2016). R.T. was initially funded by Instituto Serrapilheira and subsequently by a Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) postdoctoral fellowship (88882.315500/2019-01). A.M.R. was supported by a PhD scholarship from CAPES (88882.332986/2019-01), and A.C.M. was supported by a PhD scholarship from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq; 141668/2019-9).

Data availability

All relevant data can be found within the article and its supplementary information.

Aboubakar Nana
,
F.
,
Lecocq
,
M.
,
Ladjemi
,
M. Z.
,
Detry
,
B.
,
Dupasquier
,
S.
,
Feron
,
O.
,
Massion
,
P. P.
,
Sibille
,
Y.
,
Pilette
,
C.
and
Ocak
,
S.
(
2019
).
Therapeutic potential of focal adhesion kinase inhibition in small cell lung cancer
.
Mol. Cancer Ther.
18
,
17
-
27
.
Aksamitiene
,
E.
,
Kiyatkin
,
A.
and
Kholodenko
,
B. N.
(
2012
).
Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: a fine balance
.
Biochem. Soc. Trans.
40
,
139
-
146
.
Allinen
,
M.
,
Beroukhim
,
R.
,
Cai
,
L.
,
Brennan
,
C.
,
Lahti-Domenici
,
J.
,
Huang
,
H.
,
Porter
,
D.
,
Hu
,
M.
,
Chin
,
L.
,
Richardson
,
A.
et al.
(
2004
).
Molecular characterization of the tumor microenvironment in breast cancer
.
Cancer Cell
6
,
17
-
32
.
Beliveau
,
A.
,
Mott
,
J. D.
,
Lo
,
A.
,
Chen
,
E. I.
,
Koller
,
A. A.
,
Yaswen
,
P.
,
Muschler
,
J.
and
Bissell
,
M. J.
(
2010
).
Raf-induced MMP9 disrupts tissue architecture of human breast cells in three-dimensional culture and is necessary for tumor growth in vivo
.
Genes Dev.
24
,
2800
-
2811
.
Berglund
,
F. M.
,
Weerasinghe
,
N. R.
,
Davidson
,
L.
,
Lim
,
J. C.
,
Eickholt
,
B. J.
and
Leslie
,
N. R.
(
2013
).
Disruption of epithelial architecture caused by loss of PTEN or by oncogenic mutant p110α/PIK3CA but not by HER2 or mutant AKT1
.
Oncogene
32
,
4417
-
4426
.
Bhat
,
R.
,
Belardi
,
B.
,
Mori
,
H.
,
Kuo
,
P.
,
Tam
,
A.
,
Hines
,
W. C.
,
Le
,
Q.-T.
,
Bertozzi
,
C. R.
and
Bissell
,
M. J.
(
2016
).
Nuclear repartitioning of galectin-1 by an extracellular glycan switch regulates mammary morphogenesis
.
Proc. Natl. Acad. Sci. USA
113
,
E4820
-
E4827
.
Bissell
,
M. J.
and
Hines
,
W. C.
(
2011
).
Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression
.
Nat. Med.
17
,
320
-
329
.
Bissell
,
M. J.
,
Hall
,
H. G.
and
Parry
,
G.
(
1982
).
How does the extracellular matrix direct gene expression
?
J. Theor. Biol.
99
,
31
-
68
.
Blagosklonny
,
M. V.
and
Pardee
,
A. B.
(
2002
).
The restriction point of the cell cycle
.
Cell Cycle
1
,
102
-
109
.
Boudreau
,
N.
,
Sympson
,
C. J.
,
Werb
,
Z.
and
Bissell
,
M. J.
(
1995
).
Suppression of ICE and apoptosis in mammary epithelial cells by extracellular matrix
.
Science
267
,
891
-
893
.
Bridgewater
,
R. E.
,
Streuli
,
C. H.
and
Caswell
,
P. T.
(
2017
).
Extracellular matrix promotes clathrin-dependent endocytosis of prolactin and STAT5 activation in differentiating mammary epithelial cells
.
Sci. Rep.
7
,
4572
.
Brito
,
M. B.
,
Goulielmaki
,
E.
and
Papakonstanti
,
E. A.
(
2015
).
Focus on PTEN regulation
.
Front. Oncol.
5
,
166
.
Bustin
,
S. A.
,
Benes
,
V.
,
Garson
,
J. A.
,
Hellemans
,
J.
,
Huggett
,
J.
,
Kubista
,
M.
,
Mueller
,
R.
,
Nolan
,
T.
,
Pfaffl
,
M. W.
,
Shipley
,
G. L.
et al.
(
2009
).
The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments
.
Clin. Chem.
55
,
611
-
622
.
Cabrita
,
M. A.
,
Jones
,
L. M.
,
Quizi
,
J. L.
,
Sabourin
,
L. A.
,
Mckay
,
B. C.
and
Addison
,
C. L.
(
2011
).
Focal adhesion kinase inhibitors are potent anti-angiogenic agents
.
Mol. Oncol.
5
,
517
-
526
.
Chu
,
I.
,
Sun
,
J.
,
Arnaout
,
A.
,
Kahn
,
H.
,
Hanna
,
W.
,
Narod
,
S.
,
Sun
,
P.
,
Tan
,
C.-K.
,
Hengst
,
L.
and
Slingerland
,
J.
(
2007
).
p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2
.
Cell
128
,
281
-
294
.
Cloutier
,
G.
,
Sallenbach-Morrissette
,
A.
and
Beaulieu
,
J. F.
(
2019
).
Non-integrin laminin receptors in epithelia
.
Tissue Cell
56
,
71
-
78
.
Cohen
,
D. M.
(
2005
).
SRC family kinases in cell volume regulation
.
Am. J. Physiol. Cell Physiol.
288
,
C483
-
C493
.
Comer
,
F. I.
and
Parent
,
C. A.
(
2007
).
Phosphoinositides specify polarity during epithelial organ development
.
Cell
128
,
239
-
240
.
Debnath
,
J.
,
Walker
,
S. J.
and
Brugge
,
J. S.
(
2003
).
Akt activation disrupts mammary acinar architecture and enhances proliferation in an mTOR-dependent manner
.
J. Cell Biol.
163
,
315
-
326
.
Ewald
,
A. J.
,
Huebner
,
R. J.
,
Palsdottir
,
H.
,
Lee
,
J. K.
,
Perez
,
M. J.
,
Jorgens
,
D. M.
,
Tauscher
,
A. N.
,
Cheung
,
K. J.
,
Werb
,
Z.
and
Auer
,
M.
(
2012
).
Mammary collective cell migration involves transient loss of epithelial features and individual cell migration within the epithelium
.
J. Cell Sci.
125
,
2638
-
2654
.
Feng
,
W.
,
Wu
,
H.
,
Chan
,
L. N.
and
Zhang
,
M.
(
2008
).
Par-3-mediated junctional localization of the lipid phosphatase PTEN is required for cell polarity establishment
.
J. Biol. Chem.
283
,
23440
-
23449
.
Ferletta
,
M.
,
Kikkawa
,
Y.
,
Yu
,
H.
,
Talts
,
J. F.
,
Durbeej
,
M.
,
Sonnenberg
,
A.
,
Timpl
,
R.
,
Campbell
,
K. P.
,
Ekblom
,
P.
and
Genersch
,
E.
(
2003
).
Opposing roles of integrin α6Aβ1 and dystroglycan in laminin-mediated extracellular signal-regulated kinase activation
.
Mol. Biol. Cell
14
,
2088
-
2103
.
Fiore
,
A. P. Z. P.
,
Spencer
,
V. A.
,
Mori
,
H.
,
Carvalho
,
H. F.
,
Bissell
,
M. J.
and
Bruni-Cardoso
,
A.
(
2017
).
Laminin-111 and the level of nuclear actin regulate epithelial quiescence via exportin-6
.
Cell Rep.
19
,
2102
-
2115
.
Fournier
,
A. K.
,
Campbell
,
L. E.
,
Castagnino
,
P.
,
Liu
,
W. F.
,
Chung
,
B. M.
,
Weaver
,
V. M.
,
Chen
,
C. S.
and
Assoian
,
R. K.
(
2008
).
Rac-dependent cyclin D1 gene expression regulated by cadherin- and integrin-mediated adhesion
.
J. Cell Sci.
121
,
226
-
233
.
Fournier
,
M. V.
,
Fata
,
J. E.
,
Martin
,
K. J.
,
Yaswen
,
P.
and
Bissell
,
M. J.
(
2009
).
Interaction of E-cadherin and PTEN regulates morphogenesis and growth arrest in human mammary epithelial cells
.
Cancer Res.
69
,
4545
-
4552
.
Gaiko-Shcherbak
,
A.
,
Fabris
,
G.
,
Dreissen
,
G.
,
Merkel
,
R.
,
Hoffmann
,
B.
and
Noetzel
,
E.
(
2015
).
The acinar cage: basement membranes determine molecule exchange and mechanical stability of human breast cell Acini
.
PLoS One
10
,
e0145174
.
Gehler
,
S.
,
Ponik
,
S. M.
,
Riching
,
K. M.
and
Keely
,
P. J.
(
2013
).
Bi-directional signaling: extracellular matrix and integrin regulation of breast tumor progression
.
Crit. Rev. Eukaryot. Gene Expr.
23
,
139
-
157
.
Gérard
,
C.
and
Goldbeter
,
A.
(
2012
).
From quiescence to proliferation: Cdk oscillations drive the mammalian cell cycle
.
Front. Physiol.
3
,
413
.
Gieniec
,
K. A.
and
Davis
,
F. M.
(
2022
).
Mammary basal cells: stars of the show
.
Biochim. Biophys. Acta Mol. Cell Res.
1869
,
119159
.
Glukhova
,
M. A.
and
Streuli
,
C. H.
(
2013
).
How integrins control breast biology
.
Curr. Opin. Cell Biol.
25
,
633
-
641
.
Gokhale
,
R. H.
and
Shingleton
,
A. W.
(
2015
).
Size control: the developmental physiology of body and organ size regulation
.
Wiley Interdiscip. Rev. Dev. Biol.
4
,
335
-
356
.
Gracida-Jiménez
,
V.
,
Mondragón-González
,
R.
,
Vélez-Aguilera
,
G.
,
Vásquez-Limeta
,
A.
,
Laredo-Cisneros
,
M. S.
,
Gómez-López
,
J. D. D.
,
Vaca
,
L.
,
Gourlay
,
S. C.
,
Jacobs
,
L. A.
,
Winder
,
S. J.
et al.
(
2017
).
Retrograde trafficking of β-dystroglycan from the plasma membrane to the nucleus
.
Sci. Rep.
7
,
9906
.
Gu
,
J.
,
Tamura
,
M.
and
Yamada
,
K. M.
(
1998
).
Tumor suppressor PTEN inhibits integrin- and growth factor-mediated mitogen-activated protein (MAP) kinase signaling pathways
.
J. Cell Biol.
143
,
1375
-
1383
.
Gudjonsson
,
T.
,
Rønnov-Jessen
,
L.
,
Villadsen
,
R.
,
Rank
,
F.
,
Bissell
,
M. J.
and
Petersen
,
O. W.
(
2002
).
Normal and tumor-derived myoepithelial cells differ in their ability to interact with luminal breast epithelial cells for polarity and basement membrane deposition
.
J. Cell Sci.
115
,
39
-
50
.
Guiley
,
K. Z.
,
Stevenson
,
J. W.
,
Lou
,
K.
,
Barkovich
,
K. J.
,
Kumarasamy
,
V.
,
Wijeratne
,
T. U.
,
Bunch
,
K. L.
,
Tripathi
,
S.
,
Knudsen
,
E. S.
,
Witkiewicz
,
A. K.
et al.
(
2019
).
P27 allosterically activates cyclin-dependent kinase 4 and antagonizes palbociclib inhibition
.
Science
366
,
eaaw2106
.
Ho
,
J.
,
Cruise
,
E. S.
,
Dowling
,
R. J. O.
and
Stambolic
,
V.
(
2020
).
PTEN nuclear functions
.
Cold Spring Harb. Perspect. Med.
10
,
a036079
.
Inman
,
J. L.
,
Robertson
,
C.
,
Mott
,
J. D.
and
Bissell
,
M. J.
(
2015
).
Mammary gland development: cell fate specification, stem cells and the microenvironment
.
Development
142
,
1028
-
1042
.
Janda
,
E.
,
Litos
,
G.
,
Grünert
,
S.
,
Downward
,
J.
and
Beug
,
H.
(
2002
).
Oncogenic Ras/Her-2 mediate hyperproliferation of polarized epithelial cells in 3D cultures and rapid tumor growth via the PI3K pathway
.
Oncogene
21
,
5148
-
5159
.
Kafri
,
R.
,
Levy
,
J.
,
Ginzberg
,
M. B.
,
Oh
,
S.
,
Lahav
,
G.
and
Kirschner
,
M. W.
(
2013
).
Dynamics extracted from fixed cells reveal feedback linking cell growth to cell cycle
.
Nature
494
,
480
-
483
.
Keely
,
P. J.
(
2011
).
Mechanisms by which the extracellular matrix and integrin signaling act to regulate the switch between tumor suppression and tumor promotion
.
J. Mammary Gland Biol. Neoplasia
16
,
205
-
219
.
Khalilgharibi
,
N.
and
Mao
,
Y.
(
2021
).
To form and function: on the role of basement membrane mechanics in tissue development, homeostasis and disease
.
Open Biol.
11
,
200360
.
Kim
,
J.
and
Guan
,
K. L.
(
2019
).
mTOR as a central hub of nutrient signalling and cell growth
.
Nat. Cell Biol.
21
,
63
-
71
.
Kleinman
,
H. K.
and
Martin
,
G. R.
(
2005
).
Matrigel: basement membrane matrix with biological activity
.
Semin. Cancer Biol.
15
,
378
-
386
.
Knudson
,
A. G.
(
2001
).
Two genetic hits (more or less) to cancer
.
Nat. Rev. Cancer
1
,
157
-
162
.
Koepp
,
D. M.
(
2014
).
Cell cycle regulation by protein degradation
.
Methods Mol. Biol.
1170
,
61
-
73
.
Laprise
,
P.
,
Chailler
,
P.
,
Houde
,
M.
,
Beaulieu
,
J.-F.
,
Boucher
,
M. J.
and
Rivard
,
N.
(
2002
).
Phosphatidylinositol 3-kinase controls human intestinal epithelial cell differentiation by promoting adherens junction assembly and p38 MAPK activation
.
J. Biol. Chem.
277
,
8226
-
8234
.
Lebleu
,
V. S.
,
Macdonald
,
B.
and
Kalluri
,
R.
(
2007
).
Structure and function of basement membranes
.
Exp. Biol. Med.
232
,
1121
-
1129
.
Lee
,
J. L.
and
Streuli
,
C. H.
(
2014
).
Integrins and epithelial cell polarity
.
J. Cell Sci.
127
,
3217
-
3225
.
Lee
,
Y. R.
,
Chen
,
M.
and
Pandolfi
,
P. P.
(
2018
).
The functions and regulation of the PTEN tumour suppressor: new modes and prospects
.
Nat. Rev. Mol. Cell Biol.
19
,
547
-
562
.
Leslie
,
N. R.
,
Yang
,
X.
,
Downes
,
C. P.
and
Weijer
,
C. J.
(
2007
).
PtdIns (3,4,5)P3-dependent and -independent roles for PTEN in the control of cell migration
.
Curr. Biol.
17
,
115
-
125
.
Levental
,
K. R.
,
Yu
,
H.
,
Kass
,
L.
,
Lakins
,
J. N.
,
Egeblad
,
M.
,
Erler
,
J. T.
,
Fong
,
S. F. T.
,
Csiszar
,
K.
,
Giaccia
,
A.
,
Weninger
,
W.
et al.
(
2009
).
Matrix crosslinking forces tumor progression by enhancing integrin signaling
.
Cell
139
,
891
-
906
.
Lim
,
S.
and
Kaldis
,
P.
(
2012
).
Loss of Cdk2 and Cdk4 induces a switch from proliferation to differentiation in neural stem cells
.
Stem Cells
30
,
1509
-
1520
.
Liu
,
H.
,
Radisky
,
D. C.
,
Wang
,
F.
and
Bissell
,
M. J.
(
2004
).
Polarity and proliferation are controlled by distinct signaling pathways downstream of PI3-kinase in breast epithelial tumor cells
.
J. Cell Biol.
164
,
603
-
612
.
Lloyd
,
A. C.
(
2013
).
The regulation of cell size
.
Cell
154
,
1194
-
1205
.
Lord
,
S. J.
,
Velle
,
K. B.
,
Dyche Mullins
,
R.
and
Fritz-Laylin
,
L. K.
(
2020
).
SuperPlots: communicating reproducibility and variability in cell biology
.
J. Cell Biol.
219
,
e202001064
.
Malumbres
,
M.
and
Barbacid
,
M.
(
2009
).
Cell cycle, CDKs and cancer: a changing paradigm
.
Nat. Rev. Cancer
9
,
153
-
166
.
Martin-Belmonte
,
F.
,
Gassama
,
A.
,
Datta
,
A.
,
Yu
,
W.
,
Rescher
,
U.
,
Gerke
,
V.
and
Mostov
,
K.
(
2007
).
PTEN-mediated apical segregation of phosphoinositides controls epithelial morphogenesis through Cdc42
.
Cell
128
,
383
-
397
.
Millis
,
S. Z.
,
Ikeda
,
S.
,
Reddy
,
S.
,
Gatalica
,
Z.
and
Kurzrock
,
R.
(
2016
).
Landscape of phosphatidylinositol-3-kinase pathway alterations across 19 784 diverse solid tumors
.
JAMA Oncol.
2
,
1565
.
Moore
,
C. J.
and
Winder
,
S. J.
(
2012
).
The inside and out of dystroglycan post-translational modification
.
Neuromuscul. Disord.
22
,
959
-
965
.
Moreno-Layseca
,
P.
and
Streuli
,
C. H.
(
2014
).
Signalling pathways linking integrins with cell cycle progression
.
Matrix Biol.
34
,
144
-
153
.
Mroue
,
R.
and
Bissell
,
M. J.
(
2012
).
Three-dimensional cultures of mouse mammary epithelial cells
.
Methods Mol. Biol.
945
,
221
-
250
.
Muschler
,
J.
,
Levy
,
D.
,
Boudreau
,
R.
,
Henry
,
M.
,
Campbell
,
K.
and
Bissell
,
M. J.
(
2002
).
A role for dystroglycan in epithelial polarization: loss of function in breast tumor cells
.
Cancer Res.
62
,
7102
-
7109
.
Nelson
,
C. M.
and
Bissell
,
M. J.
(
2006
).
Of extracellular matrix, scaffolds, and signaling: tissue architecture regulates development, homeostasis, and cancer
.
Annu. Rev. Cell Dev. Biol.
22
,
287
-
309
.
O'Farrell
,
P. H.
,
Farrell
,
P. H. O.
,
O'farrell
,
P. H.
and
Farrell
,
P. H. O.
(
2011
).
Quiescence: early evolutionary origins and universality do not imply uniformity
.
Philos. Trans. R. Soc. B: Biol. Sci.
366
,
3498
-
3507
.
Onodera
,
Y.
,
Nam
,
J.
and
Bissell
,
M. J.
(
2014
).
Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathways
.
J. Clin. Investig.
124
,
367
-
384
.
Paine
,
I. S.
and
Lewis
,
M. T.
(
2017
).
The terminal end bud: the little engine that could
.
J. Mammary Gland Biol. Neoplasia
22
,
93
-
108
.
Pardee
,
A. B.
(
1974
).
A restriction point for control of normal animal cell proliferation
.
Proc. Natl. Acad. Sci. USA
71
,
1286
-
1290
.
Peng
,
J.
,
Awad
,
A.
,
Sar
,
S.
,
Hamze Komaiha
,
O.
,
Moyano
,
R.
,
Rayal
,
A.
,
Samuel
,
D.
,
Shewan
,
A.
,
Vanhaesebroeck
,
B.
,
Mostov
,
K.
et al.
(
2015
).
Phosphoinositide 3-kinase p110 δ promotes lumen formation through the enhancement of apico-basal polarity and basal membrane organization
.
Nat. Commun.
6
,
5937
.
Pensa
,
S.
,
Neoh
,
K.
,
Resemann
,
H. K.
,
Kreuzaler
,
P. A.
,
Abell
,
K.
,
Clarke
,
N. J.
,
Reinheckel
,
T.
,
Kahn
,
C. R.
and
Watson
,
C. J.
(
2014
).
The PI3K regulatory subunits p55α and p50α regulate cell death in vivo
.
Cell Death Differ.
21
,
1442
-
1450
.
Pfaffl
,
M. W.
(
2001
).
A new mathematical model for relative quantification in real-time RT-PCR
.
Nucleic Acids Res.
29
,
45e
-
445
.
Pinal
,
N.
,
Goberdhan
,
D. C. I.
,
Collinson
,
L.
,
Fujita
,
Y.
,
Cox
,
I. M.
,
Wilson
,
C.
and
Pichaud
,
F.
(
2006
).
Regulated and polarized PtdIns (3,4,5)P3 accumulation is essential for apical membrane morphogenesis in photoreceptor epithelial cells
.
Curr. Biol.
16
,
140
-
149
.
Pozzi
,
A.
and
Zent
,
R.
(
2011
).
Extracellular matrix receptors in branched organs
.
Curr. Opin. Cell Biol.
23
,
547
-
553
.
Ray
,
A.
,
James
,
M. K.
,
Larochelle
,
S.
,
Fisher
,
R. P.
and
Blain
,
S. W.
(
2009
).
p27 Kip1 inhibits cyclin D-cyclin-dependent kinase 4 by two independent modes
.
Mol. Cell. Biol.
29
,
986
-
999
.
Rhind
,
N.
(
2021
).
Cell-size control
.
Curr. Biol.
31
,
R1414
-
R1420
.
Runswick
,
S. K.
,
O'hare
,
M. J.
,
Jones
,
L.
,
Streuli
,
C. H.
and
Garrod
,
D. R.
(
2001
).
Desmosomal adhesion regulates epithelial morphogenesis and cell positioning
.
Nat. Cell Biol.
3
,
823
-
830
.
Russo
,
L. C.
,
Tomasin
,
R.
,
Matos
,
I. A.
,
Manucci
,
A. C.
,
Sowa
,
S. T.
,
Dale
,
K.
,
Caldecott
,
K. W.
,
Lehtiö
,
L.
,
Schechtman
,
D.
,
Meotti
,
F. C.
et al.
(
2021
).
The SARS-CoV-2 Nsp3 macrodomain reverses PARP9/DTX3L-dependent ADP-ribosylation induced by interferon signaling
.
J. Biol. Chem.
297
,
101041
.
Sakaue-Sawano
,
A.
,
Kurokawa
,
H.
,
Morimura
,
T.
,
Hanyu
,
A.
,
Hama
,
H.
,
Osawa
,
H.
,
Kashiwagi
,
S.
,
Fukami
,
K.
,
Miyata
,
T.
,
Miyoshi
,
H.
et al.
(
2008
).
Visualizing spatiotemporal dynamics of multicellular cell-cycle progression
.
Cell
132
,
487
-
498
.
Sang
,
L.
,
Coller
,
H. A.
and
Roberts
,
J. M.
(
2008
).
Control of the reversibility of cellular quiescence by the transcriptional repressor HES1
.
Science
321
,
1095
-
1100
.
Sgambato
,
A.
,
Camerini
,
A.
,
Faraglia
,
B.
,
Pavoni
,
E.
,
Montanari
,
M.
,
Spada
,
D.
,
Losasso
,
C.
,
Brancaccio
,
A.
and
Cittadini
,
A.
(
2004
).
Increased expression of dystroglycan inhibits the growth and tumorigenicity of human mammary epithelial cells
.
Cancer Biol. Ther.
3
,
967
-
975
.
Shapiro
,
G. I.
(
2006
).
Cyclin-dependent kinase pathways as targets for cancer treatment
.
J. Clin. Oncol.
24
,
1770
-
1783
.
Sherr
,
C. J.
and
Roberts
,
J. M.
(
1999
).
CDK inhibitors: positive and negative regulators of G1-phase progression
.
Genes Dev.
13
,
1501
-
1512
.
Spencer
,
V. A.
,
Costes
,
S.
,
Inman
,
J. L.
,
Xu
,
R.
,
Chen
,
J.
,
Hendzel
,
M. J.
and
Bissell
,
M. J.
(
2011
).
Depletion of nuclear actin is a key mediator of quiescence in epithelial cells
.
J. Cell Sci.
124
,
123
-
132
.
Spencer
,
S. L.
,
Cappell
,
S. D.
,
Tsai
,
F. C.
,
Overton
,
K. W.
,
Wang
,
C. L.
and
Meyer
,
T.
(
2013
).
The proliferation-quiescence decision is controlled by a bifurcation in CDK2 activity at mitotic exit
.
Cell
155
,
369
-
383
.
Sternlicht
,
M. D.
(
2005
).
Key stages in mammary gland development: the cues that regulate ductal branching morphogenesis
.
Breast Cancer Res.
8
,
201
.
Sternlicht
,
M. D.
,
Kouros-Mehr
,
H.
,
Lu
,
P.
and
Werb
,
Z.
(
2006
).
Hormonal and local control of mammary branching morphogenesis
.
Differentiation
74
,
365
-
381
.
Streuli
,
C. H.
and
Akhtar
,
N.
(
2009
).
Signal co-operation between integrins and other receptor systems
.
Biochem. J.
418
,
491
-
506
.
Sun
,
Y.
,
Liu
,
W.-Z.
,
Liu
,
T.
,
Feng
,
X.
,
Yang
,
N.
and
Zhou
,
H.-F.
(
2015
).
Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis
.
J. Recept. Signal Transduct. Res.
35
,
600
-
604
.
Tamura
,
M.
,
Gu
,
J.
,
Danen
,
E. H. J.
,
Takino
,
T.
,
Miyamoto
,
S.
and
Yamada
,
K. M.
(
1999
).
PTEN interactions with focal adhesion kinase and suppression of the extracellular matrix-dependent phosphatidylinositol 3-kinase/Akt cell survival pathway
.
J. Biol. Chem.
274
,
20693
-
20703
.
Tognon
,
C. E.
,
Somasiri
,
A. M.
,
Evdokimova
,
V. E.
,
Trigo
,
G.
,
Uy
,
E. E.
,
Melnyk
,
N.
,
Carboni
,
J. M.
,
Gottardis
,
M. M.
,
Roskelley
,
C. D.
,
Pollak
,
M.
et al.
(
2011
).
ETV6-NTRK3–mediated breast epithelial cell transformation is blocked by targeting the IGF1R signaling pathway
.
Cancer Res.
71
,
1060
-
1070
.
Visvader
,
J. E.
and
Stingl
,
J.
(
2014
).
Mammary stem cells and the differentiation hierarchy: current status and perspectives
.
Genes Dev.
28
,
1143
-
1158
.
Vollmer
,
J.
,
Casares
,
F.
and
Iber
,
D.
(
2017
).
Growth and size control during development
.
Open Biol.
7
,
170190
.
Watt
,
L. F.
,
Panicker
,
N.
,
Mannan
,
A.
,
Copeland
,
B.
,
Kahl
,
R. G. S.
,
Dun
,
M. D.
,
Young
,
B.
,
Roselli
,
S.
and
Verrills
,
N. M.
(
2017
).
Functional importance of PP2A regulatory subunit loss in breast cancer
.
Breast Cancer Res. Treat.
166
,
117
-
131
.
Weaver
,
V. M.
,
Petersen
,
O. W.
,
Wang
,
F.
,
Larabell
,
C. A.
,
Briand
,
P.
,
Damsky
,
C.
and
Bissell
,
M. J.
(
1997
).
Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies
.
J. Cell Biol.
137
,
231
-
245
.
Whittaker
,
S. R.
,
Mallinger
,
A.
,
Workman
,
P.
and
Clarke
,
P. A.
(
2017
).
Inhibitors of cyclin-dependent kinases as cancer therapeutics
.
Pharmacol. Ther.
173
,
83
-
105
.
Worby
,
C. A.
and
Dixon
,
J. E.
(
2014
).
PTEN
.
Annu. Rev. Biochem.
83
,
641
-
669
.
Xu
,
R.
,
Nelson
,
C. M.
,
Muschler
,
J. L.
,
Veiseh
,
M.
,
Vonderhaar
,
B. K.
and
Bissell
,
M. J.
(
2009
).
Sustained activation of STAT5 is essential for chromatin remodeling and maintenance of mammary-specific function
.
J. Cell Biol.
184
,
57
-
66
.
Xu
,
R.
,
Spencer
,
V. A.
,
Groesser
,
D. L.
and
Bissell
,
M. J.
(
2010
).
Laminin regulates PI3K basal localization and activation to sustain STAT5 activation
.
Cell Cycle
9
,
4315
-
4322
.
Yang
,
H. W.
,
Chung
,
M.
,
Kudo
,
T.
and
Meyer
,
T.
(
2017
).
Competing memories of mitogen and p53 signalling control cell-cycle entry
.
Nature
549
,
404
-
408
.
Yang
,
H. W.
,
Cappell
,
S. D.
,
Jaimovich
,
A.
,
Liu
,
C.
,
Chung
,
M.
,
Daigh
,
L. H.
,
Pack
,
L. R.
,
Fan
,
Y.
,
Regot
,
S.
,
Covert
,
M.
et al.
(
2020
).
Stress-mediated exit to quiescence restricted by increasing persistence in cdk4/6 activation
.
Elife
9
,
e44571
.
Zielke
,
N.
and
Edgar
,
B. A.
(
2015
).
FUCCI sensors: powerful new tools for analysis of cell proliferation
.
Wiley Interdiscip. Rev. Dev. Biol.
4
,
469
-
487
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information