Basal-like breast cancer is the most aggressive breast cancer subtype with the worst prognosis. Despite its high recurrence rate, chemotherapy is the only treatment for basal-like breast cancer, which lacks expression of hormone receptors. In contrast, luminal A tumors express ERα and can undergo endocrine therapy for treatment. Previous studies have tried to develop effective treatments for basal-like patients using various therapeutics but failed due to the complex and dynamic nature of the disease. In this study, we performed a transcriptomic analysis of patients with breast cancer to construct a simplified but essential molecular regulatory network model. Network control analysis identified potential targets and elucidated the underlying mechanisms of reprogramming basal-like cancer cells into luminal A cells. Inhibition of BCL11A and HDAC1/2 effectively drove basal-like cells to transition to luminal A cells and increased ERα expression, leading to increased tamoxifen sensitivity. High expression of BCL11A and HDAC1/2 correlated with poor prognosis in patients with breast cancer. These findings identify mechanisms regulating breast cancer phenotypes and suggest the potential to reprogram basal-like breast cancer cells to enhance their targetability.

Significance:

A network model enables investigation of mechanisms regulating the basal-to-luminal transition in breast cancer, identifying BCL11A and HDAC1/2 as optimal targets that can induce basal-like breast cancer reprogramming and endocrine therapy sensitivity.

Breast cancer has the highest cancer-related mortality rate among women worldwide (1). It has been categorized into various subtypes, including basal-like (or triple-negative), luminal A, luminal B, and HER2-enriched (2–4). Among these subtypes, luminal A is the least aggressive breast cancer (5). Almost 70% of patients with breast cancer are diagnosed as luminal A and have normal-like features such as expressing estrogen receptor alpha (ERα; ref 6) by which they are probable in undergoing ERα-targeted therapy as their first line of treatment (7, 8). In contrast, basal-like patients have the worst prognosis with a very high tumor recurrence rate despite its low prevalence ratio of 15% (9). They have highly elevated expression of the epidermal growth factor receptor (EGFR; ref. 10), which is an important characteristic feature of the basal-like subtype. Unfortunately, chemotherapy is the only available treatment option for basal-like patients because they are deficient in hormone receptors, including ERα, and clinical trials of all single anti-EGFR therapy have failed (11). Researchers have speculated that this might be due to interactions between the EGFR signaling pathway and epithelial–mesenchymal transition (EMT) regulators, including SLUG and ZEB1 (12), that are more likely to develop distant metastasis. As a result, several studies showed that blocking EMT regulators can abolish mesenchymal features of the basal-like subtype (13), or induce the expression of ERα (14), which allows therapeutic responsiveness to anti-ERα drugs. Nevertheless, there still remains a limitation in using conventional ERα-targeted therapies for basal-like patients because the increased expression of ERα is not a sufficient measure to consider these basal-like cells being reprogrammed into luminal A; there are complex interactions and feedback between genes and molecules that retain basal-like characteristics even after the expression of ERα is induced. To overcome this limitation, we investigated the regulatory mechanisms and underlying dynamics of differentially expressed genes (DEG) that are associated with the corresponding signaling cascades, which can possibly determine the responsiveness of anti-ERα therapy at a system level.

A cancer cell can be represented by a dynamical network model composed of various genes, proteins, and small molecules as nodes and their interactions as corresponding edges connecting them (15). A well-defined and validated logical network model can capture differential behaviors of cellular systems to unveil their hidden mechanisms and predict cellular responses when certain perturbations are given (16–18). In this study, we have constructed a molecular regulatory network model that can represent the biological conditions of basal-like and luminal A cells. We then applied a complex network control strategy to identify potential targets that can induce a subtype transition from the most invasive basal-like breast cancer subtype to the terminally differentiated luminal A subtype, which is termed basal-to-luminal A transition (BLT). We also elucidated the underlying mechanisms of BLT through analyzing the logical relationships between the key molecules in our network model. We identified B-cell lymphoma/leukemia 11A (BCL11A) and histone deacetylase 1/2 (HDAC1/2) in combination as novel targets that can reprogram extremely aggressive basal-like cells into luminal A cells by inducing the activity of luminal A phenotypic markers while reducing the activity of basal-like phenotypic markers. We further validated our simulation results by performing in vitro experiments and analyzing clinical data. Together, our study provides new insights into the subtype transition between breast cancer cells and a development of new treatment for patients with the aggressive breast cancer subtype.

Boolean network model construction for the BLT using DEG analysis

The Cancer Genome Atlas (TCGA) Breast Invasive Carcinoma (BRCA) RNA sequencing (RNA-seq) expression data were downloaded as Upper Quartile normalized fragments per kilobase of transcript per million mapped reads (FPKM-UQ) values as well as their genomic data from the Genomic Data Commons (GDC) data portal (https://portal.gdc.cancer.gov/). All genomic data as well as RNA-seq expression data from the Cancer Cell Lines Encyclopedia (CCLE; ref. 19) breast cancer cell lines were downloaded as reads per kilobase of transcript per million mapped reads (RPKM) values. These were classified according to subtype classification results from PAM50 (20). We then performed DEG analysis of the basal-like and luminal A subtypes of TCGA BRCA RNA-seq expression data using the DEGseq R package (1.38.0).

Input–output relationships of the BLT model through in silico simulation

Boolean network is a logical dynamic model with binary node states, active (ON) as 1 or inactive (OFF) as 0, that does not require detailed kinetic parameters (21, 22). One can adopt a logical Boolean network model to describe a network where each state of nodes is based on their regulatory logical rules by Boolean operators, “OR,” “NOT,” and “AND” (23). We found that every initial state of our network model reaches a stable state within 500-time steps. Hence, we updated our Boolean logical functions for 1,000 times to analyze the activities of network components according to various input settings. To represent the input frequency of each simulation, the input node is set on a cycle that capitulates a desired percentage of ON state. For example, if the input node is set to be 50% ON, it would be placed on a cycle of “01010101” or “10101010” that would remain constant throughout each simulation.

Attractor landscape and perturbation analysis using the BLT Boolean network model

Our BLT network model has 228 or 268,435,456 possible states that constitute the entire state space of the network with 28 nodes excluding input nodes. We randomly sampled 214 initial states, which was a sufficient number to cover the major attractors, that ultimately converge to either point or cyclic attractor with a basal-like or luminal A phenotype.

We calculate a phenotypic score PA to define the phenotype for each attractor A in the BLT network model as follows:

where NL is defined as luminal A marker nodes that are ON and NB as basal-like marker nodes that are ON for each attractor. The larger the PA as 1, the more luminal A phenotype an attractor has. The lower the PA as −1, the more basal-like phenotype an attractor has:

  • 0 ≤ PA ≤ 1: luminal A phenotype

  • −1 ≤ PA < 0: basal-like phenotype

For cyclic attractor, we first define the phenotype of existing states within a cyclic attractor by using PA, and then determine the phenotype as follows:

  • If CLCB, then a cyclic attractor has a luminal A phenotype,

  • If CL < CB, then a cyclic attractor has a basal-like phenotype

where CL is a number of luminal A attractors within a cyclic attractor and CB is a number of basal-like attractors within a cyclic attractor. We perform attractor analysis using the BoolNet R package (2.1.9).

Clinicopathologic analysis

To compare overall survival of patients with breast cancer with basal-like and/or luminal A patient groups, the Kaplan—Meier analysis and log-rank test were used. We downloaded clinical information of TCGA and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) patients with breast cancer from cBioPortal (http://www.cbioportal.org/; ref. 24). We first classified the patients into each patient group and then found upper, intermediate, and lower quartiles for each of BCL11A, HDAC1, and HDAC2 genes according to their Z-score normalized mRNA expressions per group. For this, we labeled and compared between high (upper quartile; above the 80th percentile) and low (lower quartile; below the 20th percentile) expressions of these genes across the patient samples in each group. The R package “survival” was used to perform overall survival analysis (25).

Gene set variation analysis with a list of gene sets

We downloaded the gene expression data of 356 patients with breast cancer with tamoxifen treatment, resistant (n = 203) and sensitive (n = 153), from the Gene Expression Omnibus (GEO) database (GSE158309). Considering our in vitro experimental conditions, we categorized the samples into four groups, BCL11A & HDAC1/2 high, BCL11A low, HDAC1/2 low, and BCL11A & HDAC1/2 low, based on the expression levels of our target genes. For this, the expression data of each gene was Z-score normalized and labeled as H (upper quartile), L (lower quartile), or I (middle) across the patient samples (lower quartile: ≤ −0.75, middle: −0.75 < x < 0.75, upper quartile: ≥0.75). The samples were then categorized according to their label as follows:

  • (a) BCL11A & HDAC1/2 high group if two or more of these gene expressions of a sample were labeled as H,

  • (b) BCL11A low group if BCL11A gene expression of a sample was labeled as I or L,

  • (c) HDAC1/2 low group if HDAC1/2 gene expressions of a sample were labeled as I or L,

  • (d) BCL11A & HDAC1/2 low group if two or more of these gene expressions of a sample were labeled as L.

We performed gene set variation analysis (GSVA) by using the R package “GSVA” (26) to assess gene set enrichment of these patient samples. A list of gene sets was downloaded from the Molecular Signature Databases (MSigDB; refs. 27, 28) and included in Supplementary Data S5. Additional information regarding any relevant data supporting the findings of this study is available from the authors upon request.

Cell culture and reagents

Human breast cancer cell lines, BT20, MDA-MB-231, HS578T, MCF7, and T47D, obtained from the Korean Cell Line Bank were cultured in DMEM purchased from WelGENE Inc. containing 10% fetal bovine serum (FBS, WelGENE Inc.) and antibiotics (100 U/mL of penicillin, 100 μg/mL of streptomycin, and 0.25 μg/mL of fungizone) purchased from Life Technologies Corp. at 37°C in a humidified atmosphere containing 5% CO2. 4-hydrotamoxifen or tamoxifen (ERα inhibitor) and romidepsin (HDAC1/2 inhibitor) were purchased from Sigma-Aldrich. Estradiol (E2) and EGF were purchased from Sigma Aldrich. shBCL11A was purchased from Sigma-Aldrich to specifically target the gene or scrambled shBCL11A-negative control. Mycoplasma infection was regularly checked using the e-Myco Mycoplasma PCR Detection Kit from iNtRON Biotechnology Inc.

Plasmid generation and virus production for overexpression experiments

We generated lentiviral particles by transfecting HEK293T cells with relevant lentiviral plasmid and packaging DNA mixture (pLP1, pLP2, and pLP/VSVG) using polyethylenimine (PEI; Invitrogen) according to the manufacturer's instructions. To perform overexpression experiments, we amplified the full-length of human HDAC1 and HDAC2 through PCR from the human kidney and colon cDNA library (Clontech), respectively. The full-length human BCL11A was purchased from Korea Human Gene Bank. The corresponding amplified gene or genes in combination were ligated into the pLentiM1.4 lentiviral vector. We then confirmed that ligation is successfully done by sequencing.

RNA extraction and real-time PCR analysis

RNA was extracted from cells using RNA-spin RNA Mini Kit purchased from iNtRON Biotechnology Inc. according to the manufacturer's instructions and treated with RNase-free DNase I (Thermo Fisher Scientific Inc.) to remove genomic DNA. Complementary DNA (cDNA) was synthesized by reverse transcription (RT) using a DiaStar RT kit purchased from Solgent. RT-PCR was performed using the PCR system (Veriti 96-well Thermal Cycler purchased from Applied Biosystems), and sequences of primers are listed in Supplementary File S1. All primers were purchased from Neoprobe. qRT-PCR analysis was performed using the QuantStudio 5 real-time PCR system (Applied Biosystems) with the primers.

Crystal violet assay

Cells were seeded at 1 × 104 cells/well in a 96-well plate or 1 × 105 cells/well in a 24-well plate for 24 hours. Cells were then treated with the indicated drugs and doses, and incubated for 72 hours. Cells were washed with phosphate-buffered saline (PBS) and stained with 0.5% (w/v) crystal violet from Sigma-Aldrich for 30 minutes at room temperature. Finally, plates were washed with tap water, air-dried, and photographed.

Western blot analysis

Cells were washed in PBS and lysed in lysis buffer (20 nmol/L of HEPES, 150 mmol/L of NaCl, 0.5% Triton X-100, 10% glycerol, 1 μg/mL of aprotinin, 1 μg of leupeptin, 1 mmol/L of Na3VO4, and 1 mmol/L of NaF). For immunoblotting, anti–phospho‐ERK1/2, anti-ERK1/2, anti–phospho-EGFR, and anti-EGFR purchased from Cell Signaling Technology Inc., anti–α‐actinin and anti-ERα purchased from Santa Cruz Biotechnology Inc. were used. For quantifying intensity of the protein bands, ImageJ software was used (http://imagej.nih.gov/ij) and normalized by GAPDH.

Hormone dependency and cell viability assay using automated live-cell imaging instrument and analysis

Breast cancer cells were seeded into a 96‐well plate at a density of 1 × 104 cells/well, and cultured in phenol red-free and serum-free medium overnight. The cells were then exposed to medium containing 10% FBS plus or minus growth factors, 0.1 nmol/L E2 or 100 ng/mL EGF, or DMSO for control. Then, the cells were collected for RNA/protein extraction after 24 hours, or cell growth was measured at each time point using IncuCyte ZOOM (Essen Biosciences) for 5 days. Images using the software were captured every 3-hour interval from three different regions per well with a 20× objective. For testing tamoxifen sensitivity, cells were seeded into a 96‐well plate at a density of 1 × 104 cells/well, cultured for 24 hours, and then treated with the indicated concentrations of tamoxifen and/or romidepsin for 72 hours. To analyze cell confluency, cells were measured using IncuCyte ZOOM to assess cell viability rates according to the manufacturer's instructions.

A mathematical model of the BLT

We summarized the workflow of this study in Supplementary Fig. S1. There are various regulatory feedback and interactions intertwined in cancer cells that the mechanism of driving the BLT may not be easily predictable. To systemically analyze this mechanism, we have constructed a mathematical model that can explain the dynamics between basal-like and luminal A breast cancer cells during the BLT. For this purpose, we have reconstructed our network model based on two major pathways, EGFR and ERα signaling, that govern cell growth, survival, and tumorigenesis in these cell types (29, 30) by using public databases such as Kyoto Encyclopedia of Genes and Genomes (31). We also used the activity of these receptors in the pathways, ERα and EGFR, as a major phenotypic marker node for determining a cellular state for the luminal A and basal-like subtype, respectively. We further identified and integrated DEGs for the corresponding subtypes as additional phenotypic markers in our network model (Supplementary Fig. S2A; Supplementary File S2). For this, we compared the mRNA expression profiles of TCGA basal-like and luminal A breast cancer patients, and we listed the top 10% ranked DEGs (Fig. 1A) that are highly expressed in one of the subtypes but lowly expressed from another for each subtype. The selected nodes and their associated links in our network model are literature-based and supported by experimental evidence obtained from public databases (Supplementary File S3). We also confirmed the expression patterns of these selected DEGs from the corresponding TCGA patient samples by using the CCLE and METABRIC (32) cohorts (Fig. 1B–G; Supplementary Fig. S2B–S2G). As a result, we have constructed a simplified but essential regulatory network model for the BLT, which consists of 30 nodes and 73 links that can present molecular interactions between the ERα and EGFR signaling pathways as well as DEGs of the corresponding subtypes (Fig. 2A).

Network modification specific to basal-like cell lines

Although cancer cell lines were originated from the same tissue, they have various differences depending on their genomic information, including copy number alteration (CNA), mRNA expression, and somatic mutation. Prior to computational simulation, we have modified our BLT network model to cell line–specific networks by analyzing genomic data of 21 basal-like breast cancer cell lines from the CCLE cohort. We determined the functional results of each genomic change for the nodes in our model (Fig. 2B–D). A result of hierarchical clustering using the functional genomic profile shows that the basal-like cell lines fall into two groups (Fig. 2E). The main difference between the two groups is that most cell lines have PI3K and/or PTEN alterations in Group 1, whereas KRAS alteration is more frequent in Group 2. Thus, we selected BT20 and MDA-MB-231 cell lines with the corresponding alterations from each of the two groups (Supplementary Fig. S3). To modify our BLT model to cell line–specific networks, we mapped the functional genomic alterations of these cell lines onto our model to differentially rewire the network model (Fig. 2F; Supplementary File 4).

Reflecting molecular features for the breast cancer subtypes to the BLT model

To affirm that our BLT network model can reproduce the biological condition of both luminal A and basal-like cells, we have performed in silico simulation and observed the activity change of our network components depending on the intensity of each input signal, estrogen (33) and EGF (34). Considering to perform these qualitative simulations dependent on incoming signals, one can investigate the dynamics of a network model by evaluating the output results based on different concentrations of input signals (35). For detailed information, please see Materials and Methods. We then compared our results with previous experimental data to validate our model. As a result, we were able to reproduce the physiological condition of luminal A cells, of which, the activities of luminal A DEGs (36–38), ERα (39), and ERα-downstream molecules, including c-Src, AKT, and ERK1/2 (40, 41), are dependent on the intensity of estrogen. In contrast, the activities of basal-like DEGs (42, 43), EGFR, and EGFR-downstream molecules, including PI3K, AKT, KRAS, and ERK1/2 (34), are dependent on the intensity of EGF (Supplementary Fig. S4A) in basal-like cells.

Furthermore, basal-like cancer cells are solely dependent on EGFs to activate the downstream molecules even when estrogens coexist due to ERα being absent on the cell membrane. To validate this physiologic condition of the cells, we performed the same simulation with differential input settings where EGF is increasing its intensity from 0% to 100% by 1% increments with constitutively active estrogen at 0%, 50%, or 100% (Supplementary Fig. S4B). At 0% of active estrogen, the basal-like DEGs, EGFR, and its downstream molecules respond to EGF and continuously increase their activity as the intensity of EGF increases. However, the activity of luminal A DEGs and ERα shows no change because there is no estrogen. At 50% of active estrogen, the activity of basal-like DEGs, EGFR, and EGFR-downstream molecules increases, whereas the activity of luminal A DEGs and ERα decreases. Likewise, at 100% of active estrogen, the activity of basal-like–related nodes is dependent on EGF and increases their activity, whereas the luminal A–related nodes decrease their activity even with constitutively active estrogen at the highest level of intensity. These results illustrate that our model can well reproduce the cellular responses of basal-like cancer cells as well as luminal A cancer cells according to various input settings. Therefore, it is pertinent to note that our model is adequate to investigate the mechanism of the BLT at a system level.

Identifying possible targets for BLT through a systems biological approach

Next, we employed a complex network control strategy called logical domain of influence (LDOI)-based target control strategy (44) to identify potential targets that can drive the BLT and uncover its underlying mechanisms in our network model. The LDOI of a node can be defined as nodes and their corresponding state that are influenced and ultimately become stabilized by the node regardless of any initial states of a system. The algorithm of LDOI-based target control strategy retracts back what state of nodes influences a specific target node of interest based on the provided logical rules of a network model. As a result, it revealed nodes and their corresponding states as solution sets that can drive a system to any desired state (44).

Our network model does not have a single node that defines whether an attractor has either luminal A or basal-like phenotype. Thus, we have defined the luminal A and basal-like phenotype according to the activity of phenotypic markers within attractors. For instance, we defined a state as luminal A if the activity of ERα and/or luminal A DEGs are active (ON), and EGFR signaling molecules and/or basal-like DEGs are inactive (OFF). Conversely, we defined a state as basal-like if the activity of EGFR signaling molecules and/or basal-like DEGs are ON, and ERα and/or luminal A DEGs are OFF. Hereafter, we denoted “∼” as OFF or negation of a node. For instance, [∼EGFR] corresponds to “EGFR OFF” or “NOT EGFR”. Due to complexity problem, we primarily considered identifying potential targets that can induce the activity of ERα and luminal A DEGs, and simultaneously block the activity of EGFR and its downstream molecules. Among the list of solution sets, we identified the optimal targets that can also predominantly block the activity of basal-like DEGs to prompt the BLT. From the BLT network without any mutation (nominal), we found a total of three potential target solution sets, which are [∼KRAS, ∼HDAC1/2], [∼PI3K, ∼HDAC1/2], and [∼HDAC1/2, ∼BCL11A] (Table 1). For modifying our model to cell line–specific networks, we fixed KRAS as ON for the MDA-MB-231 network according to our functional genomic alteration profile. Likewise, we fixed PI3K as ON and PTEN as OFF for the BT20 network. As a result, we found potential target solution sets as [∼PI3K, ∼HDAC1/2] and [∼HDAC1/2, ∼BCL11A] for the MDA-MB-231 network, and [∼KRAS, ∼HDAC1/2] and [∼HDAC1/2, ∼BCL11A] for the BT20 network. Interestingly, [∼HDAC1/2, ∼BCL11A] was commonly found in all our nominal and cell line–specific network models, and thereby is the optimal combinatorial targets that can sufficiently induce the BLT. These results were in accordance with our quantitative Boolean attractor simulation by perturbing every single and double node(s) in our network models (Supplementary Fig. S5).

Double KO of BCL11A and HDAC1/2 drives the cell-fate switching to luminal A phenotype

An expanded network provided by the LDOI-based target control strategy is similar to hypergraph that integrates regulatory interactions and dynamics of a network (44). We employed the expanded network of our BLT model to analyze how BCL11A and HDAC1/2 interact with the other network components. The resulting expanded network of our model encompasses every footprint of the network components, including all possible initial states (basal-like phenotype) to our desired state, luminal A (Fig. 3A). We then analyzed how the BLT is occurring by navigating the transition after each perturbation, HDAC1/2 KO and/or BCL11A KO.

KO of HDAC1/2 induced the activity of [ESR1], which prompted the activity of luminal A DEGs and ERα as well as directly initiated [∼EGFR] and [∼ERK1/2] (Fig. 3B). As soon as ERα was activated, its direct downstream molecule, [c-Src], was also activated and induced its further downstream molecules, including [AKT] and [ERK1/2]. Therefore, KO of HDAC1/2 initially induced the activity of ERα and luminal A DEGs but ultimately turned OFF their activity by activating the EGFR downstream molecules that are shared with the ERα signaling pathway (Fig. 3B). In contrast, KO of BCL11A directly downregulated [PI3K], [AKT], and some of the basal-like DEGs such as [SOX11], [ZEB1], and [SLUG] (Fig. 3C). It can also partially regulate the activity of HDAC1/2 that can be turned OFF only if both [AKT] and [ERK1/2] are inactive. Together, it is imperative that both KO of HDAC1/2 and BCL11A are required to induce the activity of ERα and luminal A DEGs, and simultaneously block the activity of EGFR signaling molecules and basal-like DEGs.

Modification of dynamic stability of the attractors after KO of BCL11A and HDAC1/2

To investigate dynamic stability of the states and to examine effectiveness of the targets for the BLT, we performed attractor landscape analysis by mathematically analyzing the basin of each attractor. An attractor is a steady state within a network model that can be defined by binary activity of molecules (23). The attractor can be either stabilized at a single state, which is named as a point attractor, or as a limit cycle of multiple states that are recurrently traversed by dynamics of a network, which is named as a cyclic attractor (45). Boolean network with n nodes has 2n different initial states as their entire state space. A basin of attractors is a set of initial states that will ultimately converge to a particular attractor determined by the interactions between network components. We can analyze the relative stability of those components by measuring the basin size of attractors (46, 47). For this, we first defined the phenotype of each attractor by using binary activities of the phenotypic marker nodes within each attractor (see Materials and Methods for details). We then calculated how many initial states converge to the attractors with a particular phenotype over time out of a total number of the initial states in each network before and after the target perturbation. In the MDA-MB-231 network (no perturbation), there were four attractors whose phenotype is basal-like according to their phenotypic score, and the basin of luminal A was 0% (Fig. 4A). After KO of BCL11A or HDAC1/2 from the network, there were distributed attractors, including point and cyclic attractors with repeatedly visited luminal A states, and the basin of luminal A phenotype was increased to 49.342% or 33.956%, respectively. When both BCL11A and HDAC1/2 were KO, the basin of the entire eight attractors was shown to have the luminal A phenotype with 100% basin size. These results were consistent with the BT20 network (Fig. 4B).

High expressions of BCL11A and/or HDAC1/2 correlated with poor prognosis

To validate our network analysis and simulation results, we performed Kaplan–Meier analysis of overall survival among basal-like and/or luminal A patients using TCGA (Fig. 4C–K) and METABRIC (Supplementary Fig. S6) cohorts. Among TCGA basal-like patients (n = 115), overall survival for BCL11A (high = 27, low = 29, P = 0.049), HDAC1 (high = 16, low = 28, P = 0.04), and HDAC2 (high = 17, low = 27, P = 0.012) showed that high expressions of these genes were correlated with poor prognosis (Fig. 4C–E). Similar results were shown from TCGA luminal A patients (n = 356; Fig. 4F–H). Finally, among basal-like and luminal A patients (n = 471), Kaplan–Meier analysis of overall survival for BCL11A (high = 76, low = 93, P = 0.049), HDAC1 (high = 51, low = 89, P = 0.024), and HDAC2 (high = 91, low = 94, P = 0.011) also showed that high expressions of the corresponding genes were correlated with poor prognosis (Fig. 4I–K). These results were in concordance with the results using the METABRIC dataset (Supplementary Fig. S6).

BCL11A and HDAC1/2 regulate protein activities and mRNA expression of luminal A and basal-like phenotypic markers

To further validate our analyses, we performed in vitro experiments for short hairpin RNA (shRNA) knockdown (KD) and drug treatment in three of the basal-like breast cancer cell lines, MDA-MB-231 (Fig. 5; Supplementary Fig. S7), BT20 (Fig. 5), and HS578T (Supplementary Fig. S7A and S7B). All experiments were repeated three times. We have observed the BLT through cell culture experiments (mRNA, protein expressions, and cell morphology), drug responsiveness, and growth factor–dependent cell growth. We used shRNA for blocking BCL11A gene (Fig. 5A and C), whereas romidepsin (an HDAC1/2-specific inhibitor; Fig. 5B and D) that biochemically binds to HDAC1/2 to prevent their protein activity. We also have used shRNA for blocking HDAC1/2 genes in the MDA-MB-231 cells to observe the expression of ESR1 and ERα (Supplementary Fig. S7C and S7D). The effect of BLT using shRNA was consistent with romidepsin through the mRNA expression of ESR1 and protein expression of ERα.

Blocking HDAC1/2 remarkably increased the mRNA expression of ESR1 by 4-fold in MDA-MB-231 cells (P = 0.048) and 2-fold in BT20 cells (P = 0.049; Fig. 5B and D). KD of BCL11A did not show much change for the expression of ESR1 in MDA-MB-231 cells (P = 0.015), whereas it increased the expression of ESR1 by 1.5-fold in BT20 cells (P = 0.035). After both BCL11A and HDAC1/2 were inhibited, the expression of ESR1 was increased by 10-fold in MDA-MB-231 cells (P = 0.005) and 3.5-fold in BT20 cells (P = 0.003). These results showed that the fold changes of ESR1 expression after inhibiting the target(s) can vary depending on the cell lines with different mutation profiles. However, it was consistent that inhibiting both BCL11A and HDAC1/2 can synergistically increase the expression of ESR1 to drive the BLT in both MDA-MB-231 and BT20 cells.

Next, we accessed the effect of BLT by measuring the protein expression of ERα, EGFR, and ERK1/2 and then quantified the results using ImageJ software. Using romidepsin to block HDAC1/2 considerably increased the expression of ERα in BT20 cells. Although the effect was not as substantial in shBCL11A-treated cells, the expression of ERα was increased by 15-fold in MDA-MB-231 cells (P = 0.031) and 18-fold in BT20 cells (P = 0.046) after inhibiting both BCL11A and HDAC1/2. Moreover, the expression of phosphorylated EGFR (pEGFR) was decreased by 10-fold in MDA-MB-231 cells (P = 0.047) and 100-fold in BT20 cells (P = 0.006) after inhibiting both BCL11A and HDAC1/2. Similarly, the expression of phosphorylated ERK1/2 (pERK1/2) was reduced by 3-fold in MDAMB231 cells (P < 0.001) and 114-fold in BT20 cells (P = 0.003; Fig. 5E and F).

BCL11A and HDAC1/2 regulate tamoxifen sensitivity in basal-like breast cancer cells

Increased protein expression of ERα in basal-like cells after blocking BCL11A and HDAC1/2 suggested that the cells have been reprogramed into luminal A cells. We hypothesized that these reprogrammed cells would now be sensitive to anti-hormone treatment targeting ERα. To validate this hypothesis, we used 5 μg/mL of 4-OH-tamoxifen (tamoxifen), which is an ERα-targeted drug for luminal breast cancer patients, to treat MDA-MB-231 and BT20 cells. While the control cells (shScrambled) showed no sensitivity, BCL11A-silenced cells (using shBCL11A) or HDAC1/2-inhibited cells (using romidepsin) showed some sensitivity to tamoxifen. As expected, the cells that were treated with both shBCL11A and romidepsin, which had the highest protein expression level of ERα, showed the most sensitivity to tamoxifen in both MDA-MB-231 (Fig. 5G and I) and BT20 (Fig. 5H and J) cells. These results support our hypothesis that depletion of both BCL11A and HDAC1/2 can sufficiently induce the BLT through their increased expression of ESR1 and ERα, and tamoxifen sensitivity.

Overexpression of BCL11A and HDAC1/2 can reactivate EGFR–ERK1/2 signaling in luminal A cells

To further validate, we conversely performed in vitro experiments for BCL11A and HDAC1/2 overexpression (OE), and drug treatment in two of the luminal A breast cancer cell lines, T47D (Fig. 6) and MCF7 (Supplementary Fig. S7E–S7I). Fold difference >1.5-fold increase was considered to be overexpression. BCL11A and/or HDAC1/2 OE did not show dramatic changes of ESR1 or EGFR mRNA expression levels in T47D cells (Fig. 6A–C). After BCL11A or HDAC1/2 OE, however, we observed decreased protein expressions of ERα as well as increased expressions of pEGFR and pERK1/2. Almost no expression of ERα was observed when all these genes were overexpressed in T47D cells (Fig. 6D). Similarly, BCL11A and HDAC1/2 OE synergistically decreased the expression of ESR1 for 2-fold and increased the EGFR expression for 30-fold in MCF7 cells (Supplementary Fig. S7E–S7G) as well as their protein expressions accordingly (Supplementary Fig. S7H). Moreover, BCL11A and/or HDAC1/2 OE also drastically changed the morphology of MCF7 cells towards a more mesenchymal-like phenotype (Supplementary Fig. S7I).

Decreased protein expression of ERα in luminal A cells after BCL11A and HDAC1/2 OE suggested that the cells possibly have been reprogramed into basal-like cells that are not responsive to tamoxifen. To validate our hypothesis, we used 10 μg/mL of tamoxifen to treat the cells. While the control cells responded to tamoxifen, BCL11A OE and/or HDAC1/2 OE cells showed no sensitivity to tamoxifen in T47D cells (Fig. 6E–G). These results further substantiate our hypothesis that both BCL11A and HDAC1/2 OE can conversely induce the reverse-BLT through the upregulated expression of pERK1/2 and pEGFR as well as the downregulated expression of ERα, which ultimately regulate tamoxifen sensitivity.

BCL11A and HDAC1/2 regulate cell growth dependency in basal-like and luminal A cells

To analyze cell growth dependency on different growth factors, cells were measured using IncuCyte ZOOM to assess cell viability rates. While the shScrambled basal-like cells showed no dependency on estradiol (E2) for growth, shBCL11A or shHDAC1/2 cells showed some dependency, and shBCL11A and shHDAC1/2 cells became fully dependent on E2 for growth (Supplementary Fig. S8A). When treated with EGF, the shScrambled cells were dependent on EGF for growth, whereas all of BCL11A- and/or HDAC1/2-silenced cells showed no dependency on EGF for growth (Supplementary Fig. S8B). This is due to increased protein expressions of ERα (Supplementary Fig. S8C) and decreased expressions of pEGFR and pERK1/2 (Supplementary Fig. S8D) after inhibiting BCL11A and HDAC1/2. Furthermore, E2-triggered basal-like cells showed increased RNA expression patterns of ERα downstream target genes after silencing the target(s) (Supplementary Fig. S8E), whereas the cells showed no change or decreased RNA expression patterns of EGFR downstream target genes even after EGF stimulation (Supplementary Fig. S8F). This indicates that inhibition of BCL11A and HDAC1/2 not only enhances targetability of basal-like cells but also switches their dependency from EGF to E2 for growth.

To further validate, we conversely analyzed cell growth dependency in luminal A cells. The results showed that the control cells had complete dependency on E2 for growth, whereas BCL11A and/or HDAC1/2 OE cells were not dependent on E2 for growth (Supplementary Fig. S8G). In addition, the control cells did not show any cell growth increase after EGF stimulation, whereas BCL11A and/or HDAC1/2 OE cells showed dependency on EGF for growth (Supplementary Fig. S8H). Although the OE cells still showed increased protein expressions of ERα after E2 stimulation (Supplementary Fig. S8I), they also showed increased expression of pEGFR and pERK1/2 (Supplementary Fig. S8J), which possibly had led them to depend on EGF for growth. Furthermore, E2-triggered BCL11A and/or HDAC1/2 OE luminal A cells showed no change in RNA expression patterns of the ERα-downstream target genes in contrast to their control (Supplementary Fig. S8K). However, the OE cells showed increased RNA expression patterns of the EGFR downstream target genes after EGF stimulation (Supplementary Fig. S8L). These results further support our hypothesis that depletion of both BCL11A and HDAC1/2 can sufficiently induce the BLT, whereas BCL11A and HDAC1/2 OE can conversely induce the reverse-BLT through activating the EGFR pathway.

Switching the signal flow from the EGFR- to ERα signaling pathway can make cells rely on the ERα cascade for their growth. This can be partially done by solely blocking BCL11A in our network model. The role of BCL11A KO is to inactivate the EGFR/AKT signaling pathway by blocking its interaction with PI3K, which is in accord with the previous study (48). To inactivate further downstream molecules such as ZEB1, SLUG, and SOX11, ERK1/2 also needs to be inactivated, which can be done by blocking HDAC1/2. KO of HDAC1/2 has two opposite roles in our network model. It initially induces the activity of ESR1 and thereby reduces the activity of EGFR, ERK1/2, and the basal-like DEGs, CDCA7 and ZEB1. This is consistent with previous studies that HDAC1/2 transcriptionally regulates various genes, including ESR1 (49, 50) and EGFR (47, 51). Inactivation of both ERK1/2 and AKT can terminate the activity of entire basal-like DEGs. Moreover, induction of ESR1 activity further activates ERα and most luminal A DEGs. As soon as the activity of ERα is induced, however, it starts to trigger the activity c-SRC and its downstream molecules that are shared between the EGFR and ERα signaling pathways. As a result, the EGFR signaling molecules as well as the basal-like DEGs become activated and, eventually, intervene positive feedback between the luminal A DEGs and ERα signaling pathway. Therefore, both BCL11A and HDAC1/2 are required to be terminated to completely reprogram basal-like cells into luminal A cells by turning OFF the basal-like phenotypic nodes and turning ON the luminal A phenotypic nodes.

To confirm that our results are clinically reasonable, we analyzed the gene expression data of 356 patients with breast cancer with tamoxifen treatment. We first categorized the patient samples into four groups, BCL11A & HDAC1/2 high, BCL11A low, HDAC1/2 low, and BCL11A & HDAC1/2 low, based on the expression of these genes. We then analyzed the relationship between tamoxifen sensitivity and the expression of our target genes according to each conditional subgroup. As a result, a higher percentage of the tamoxifen-resistant group was enriched within the BCL11A & HDAC1/2 high group, whereas the tamoxifen-sensitive group was highly enriched in the BCL11A & HDAC1/2 low group (Supplementary Fig. S9A). Thus, we further affirmed that high expressions of the target genes were enriched in the tamoxifen-resistant patient group and inhibiting them can possibly drive the patients to become sensitive to the drug.

Finally, we performed GSVA to assess gene set enrichment of these patient samples by calculating sample-wise gene set enrichment scores of various gene sets that are associated with the ERα and EGFR signaling pathways. GSVA scores of the gene sets that are upregulated in basal-like or luminal A breast cancer (Supplementary Fig. S9B), and are downstream genes of ERα or EGFR signaling pathway (Supplementary Fig. S9C) for each of the conditional subgroups. These results show that cancer cells with high expression levels of BCL11A and HDAC1/2 are more likely to be tamoxifen-resistant and dependent on the EGFR signaling pathway for growth. Conversely, cancer cells with low expression levels of BCL11A and HDAC1/2 are more likely to be tamoxifen-sensitive and dependent on the ERα signaling pathway for growth. Therefore, blocking BCL11A and HDAC1/2 will drive tamoxifen-resistant cells to activate the ERα signaling pathway, which may provide a vulnerability for endocrine therapy in basal-like patients.

HDAC1/2 is a well-known epigenetic regulator that governs the expression of ERα (49) by removing acetyl groups that are bound at the estrogen response element (ERE) site of ERα or from the promoter region of ESR1 (52). In addition, BCL11A is a transcription factor that regulates the cell cycle and apoptosis in lymphoma and hematopoietic stem cells (24, 53). BCL11A is also known for regulating various chromatin regulators such as histone deacetylases. A previous study showed that BCL11A can recruit endogenous SIRT1, the class III histone deacetylase, to the promotor template, which plays an important role in transcriptional regulation (54). In addition, BCL11A is a potential regulator of fetal hemoglobin that can ameliorate the major β-hemoglobin disorders by regulating the nucleosome remodeling deacetylase (NuRD) complex, including HDAC1/2, in erythroid cells (55, 56). Furthermore, its interaction with histone deacetylase complexes promotes tumorigenesis in triple-negative breast cancer (TNBC; ref. 57). Although BCL11A and HDAC1/2 were previously reported as highly expressed genes in TNBC (49, 58), their role in the cellular functions of breast cancer cells was not previously identified.

Cancer is a highly heterogeneous disease with various genetic and epigenetic modifications. Although we are only considering two subtypes within breast cancer, our analysis has shown that even these relatively homogeneous groups of patients with breast cancer and cell lines could be very heterogeneous with their diversified mutation profiles. For instance, we found out from our in vitro experiments that interactive relationships between BCL11A and HDAC1/2 could be different at the gene expression level and independently related to cancer subtypes or cell types with different mutation profiles. In addition, the scope of this study was limited to in vitro experiments to validate the identified targets using the systems biological approach. Even with our effort to overcome these limitations, it is still hard to rationalize that our network can recapitulate the entire basal-like and luminal A breast cancer subtypes with diverse genetic backgrounds. Thus, it is imperative to study the relationship between BCL11A and HDAC1/2 more in-depth using animal models as well as to more precisely represent each of patient samples and cell lines.

Here, we suggest that highly expressed BCL11A may recruit more frequently existing HDAC1/2 to the promoter region of ESR1 to block its transcription as well as its target genes. Moreover, we show how double inhibition of BCL11A and HDAC1/2 can regulate the ERα and EGFR signaling cascades as well as the basal-like and luminal DEGs to synergistically reprogram basal-like cells into luminal A cells, and vice versa. Therefore, our study demonstrates that the systems biological approach to identify such targets can be a useful tool for analyzing and governing the repertoire of complex biological networks of biological phenomena.

No disclosures were reported.

S.R. Choi: Resources, data curation, formal analysis, validation, investigation, visualization, methodology, writing–original draft. C.Y. Hwang: Resources, formal analysis, validation, investigation, visualization, methodology. J. Lee: Software, formal analysis, investigation, visualization, methodology. K.-H. Cho: Conceptualization, formal analysis, supervision, funding acquisition, validation, investigation, writing–original draft, project administration, writing–review and editing.

This work was supported by the National Research Foundation of Korea (NRF) funded by the Korea Government, the Ministry of Science and ICT (2020R1A2B5B03094920), and by Electronics and Telecommunications Research Institute (ETRI) grants (21ZS1100, Core Technology Research for Self-Improving Integrated Artificial Intelligence System). It was also supported by the KAIST Grand Challenge 30 Project. We thank Dongkwan Shin and Jae Il Joo for their analytic support.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Torre
LA
,
Islami
F
,
Siegel
RL
,
Ward
EM
,
Jemal
A
. 
Global cancer in women: Burden and trend
.
Cancer Epidem Biomar
2017
;
26
:
444
57
.
2.
Perou
CM
,
Sorlie
T
,
Eisen
MB
,
van de Rijn
M
,
Jeffrey
SS
,
Rees
CA
, et al
Molecular portraits of human breast tumours
.
Nature
2000
;
406
:
747
52
.
3.
Prat
A
,
Perou
CM
. 
Mammary development meets cancer genomics
.
Nat Med
2009
;
15
:
842
4
.
4.
Lim
E
,
Vaillant
F
,
Wu
D
,
Forrest
NC
,
Pal
B
,
Hart
AH
, et al
Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers
.
Nat Med
2009
;
15
:
907
13
.
5.
Dai
X
,
Cheng
H
,
Chen
X
,
Li
T
,
Zhang
J
,
Jin
G
, et al
FOXA1 is prognostic of triple negative breast cancers by transcriptionally suppressing SOD2 and IL6
.
Int J Biol Sci
2019
;
15
:
1030
41
.
6.
Esteva
FJ
,
Sahin
AA
,
Cristofanilli
M
,
Arun
B
,
Hortobagyi
GN
. 
Molecular prognostic factors for breast cancer metastasis and survival
.
Semin Radiat Oncol
2002
;
12
:
319
28
.
7.
Voduc
KD
,
Cheang
MC
,
Tyldesley
S
,
Gelmon
K
,
Nielsen
TO
,
Kennecke
H
. 
Breast cancer subtypes and the risk of local and regional relapse
.
J Clin Oncol
2010
;
28
:
1684
91
.
8.
Howlader
N
,
Altekruse
SF
,
Li
CI
,
Chen
VW
,
Clarke
CA
,
Ries
LA
, et al
US incidence of breast cancer subtypes defined by joint hormone receptor and HER2 status
.
J Natl Cancer Inst
2014
;
106
::
dju055
.
9.
Millikan
RC
,
Newman
B
,
Tse
CK
,
Moorman
PG
,
Conway
K
,
Dressler
LG
, et al
Epidemiology of basal-like breast cancer
.
Breast Cancer Res Treat
2008
;
109
:
123
39
.
10.
Jiang
W
,
Wang
X
,
Zhang
C
,
Xue
L
,
Yang
L
. 
Expression and clinical significance of MAPK and EGFR in triple-negative breast cancer
.
Oncol Lett
2020
;
19
:
1842
8
.
11.
Nakai
K
,
Hung
MC
,
Yamaguchi
H
. 
A perspective on anti-EGFR therapies targeting triple-negative breast cancer
.
Am J Cancer Res
2016
;
6
:
1609
23
.
12.
Majorini
MT
,
Manenti
G
,
Mano
M
,
De Cecco
L
,
Conti
A
,
Pinciroli
P
, et al
cIAP1 regulates the EGFR/Snai2 axis in triple-negative breast cancer cells
.
Cell Death Differ
2018
;
25
:
2147
64
.
13.
Su
Y
,
Hopfinger
NR
,
Nguyen
TD
,
Pogash
TJ
,
Santucci-Pereira
J
,
Russo
J
. 
Epigenetic reprogramming of epithelial mesenchymal transition in triple negative breast cancer cells with DNA methyltransferase and histone deacetylase inhibitors
.
J Exp Clin Cancer Res
2018
;
37
:
314
.
14.
Zhao
H
,
Agazie
YM
. 
Inhibition of SHP2 in basal-like and triple-negative breast cells induces basal-to-luminal transition, hormone dependency, and sensitivity to anti-hormone treatment
.
BMC Cancer
2015
;
15
:
109
.
15.
Kwon
YK
,
Cho
KH
. 
Boolean dynamics of biological networks with multiple coupled feedback loops
.
Biophys J
2007
;
92
:
2975
81
.
16.
Fumia
HF
,
Martins
ML
. 
Boolean network model for cancer pathways: predicting carcinogenesis and targeted therapy outcomes
.
PLoS One
2013
;
8
:
e69008
.
17.
Shin
SY
,
Kim
T
,
Lee
HS
,
Kang
JH
,
Lee
JY
,
Cho
KH
, et al
The switching role of beta-adrenergic receptor signalling in cell survival or death decision of cardiomyocytes
.
Nat Commun
2014
;
5
:
5777
.
18.
Kim
JR
,
Kim
J
,
Kwon
YK
,
Lee
HY
,
Heslop-Harrison
P
,
Cho
KH
. 
Reduction of complex signaling networks to a representative kernel
.
Sci Signal
2011
;
4
:
ra35
.
19.
Barretina
J
,
Caponigro
G
,
Stransky
N
,
Venkatesan
K
,
Margolin
AA
,
Kim
S
, et al
The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity
.
Nature
2012
;
483
:
603
7
.
20.
Parker
JS
,
Mullins
M
,
Cheang
MC
,
Leung
S
,
Voduc
D
,
Vickery
T
, et al
Supervised risk predictor of breast cancer based on intrinsic subtypes
.
J Clin Oncol
2009
;
27
:
1160
7
.
21.
Saez-Rodriguez
J
,
Alexopoulos
LG
,
Epperlein
J
,
Samaga
R
,
Lauffenburger
DA
,
Klamt
S
, et al
Discrete logic modelling as a means to link protein signalling networks with functional analysis of mammalian signal transduction
.
Mol Syst Biol
2009
;
5
:
331
.
22.
Kim
J
,
Park
SM
,
Cho
KH
. 
Discovery of a kernel for controlling biomolecular regulatory networks
.
Sci Rep
2013
;
3
:
2223
.
23.
Kauffman
SA
. 
Metabolic stability and epigenesis in randomly constructed genetic nets
.
J Theor Biol
1969
;
22
:
437
67
.
24.
Gao
Y
,
Wu
H
,
He
D
,
Hu
X
,
Li
Y
. 
Downregulation of BCL11A by siRNA induces apoptosis in B lymphoma cell lines
.
Biomed Rep
2013
;
1
:
47
52
.
25.
Gentleman
RC
,
Carey
VJ
,
Bates
DM
,
Bolstad
B
,
Dettling
M
,
Dudoit
S
, et al
Bioconductor: open software development for computational biology and bioinformatics
.
Genome Biol
2004
;
5
:
R80
.
26.
Hanzelmann
S
,
Castelo
R
,
Guinney
J
. 
GSVA: gene set variation analysis for microarray and RNA-seq data
.
BMC Bioinformatics
2013
;
14
:
7
.
27.
Liberzon
A
,
Birger
C
,
Thorvaldsdottir
H
,
Ghandi
M
,
Mesirov
JP
,
Tamayo
P
. 
The Molecular Signatures Database (MSigDB) hallmark gene set collection
.
Cell Syst
2015
;
1
:
417
25
.
28.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
29.
Cheang
MCU
,
Voduc
D
,
Bajdik
C
,
Leung
S
,
McKinney
S
,
Chia
SK
, et al
Basal-like breast cancer defined by five biomarkers has superior prognostic value then triple-negative phenotype
.
Clin Cancer Res
2008
;
14
:
1368
76
.
30.
Arnal
JF
,
Lenfant
F
,
Metivier
R
,
Flouriot
G
,
Henrion
D
,
Adlanmerini
M
, et al
Membrane and nuclear estrogen receptor alpha actions: From tissue specificity to medical implications
.
Physiol Rev
2017
;
97
:
1045
87
.
31.
Kanehisa
M
,
Goto
S
. 
KEGG: kyoto encyclopedia of genes and genomes
.
Nucleic Acids Res
2000
;
28
:
27
30
.
32.
Pereira
B
,
Chin
SF
,
Rueda
OM
,
Vollan
HK
,
Provenzano
E
,
Bardwell
HA
, et al
The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes
.
Nat Commun
2016
;
7
:
11479
.
33.
Welboren
WJ
,
Stunnenberg
HG
,
Sweep
FC
,
Span
PN
. 
Identifying estrogen receptor target genes
.
Mol Oncol
2007
;
1
:
138
43
.
34.
Zakaria
Z
,
Zulkifle
MF
,
Wan Hasan
WAN
,
Azlah
AK
,
Raub
SHA
,
Eswaran
J
, et al
Epidermal growth factor receptor (EGFR) gene alteration and protein overexpression in Malaysian triple-negative breast cancer (TNBC) cohort
.
Onco Targets Ther
2019
;
12
:
7749
56
.
35.
Helikar
T
,
Kochi
N
,
Kowal
B
,
Dimri
M
,
Naramura
M
,
Raja
SM
, et al
A comprehensive, multi-scale dynamical model of ErbB receptor signal transduction in human mammary epithelial cells
.
PLoS One
2013
;
8
:
e61757
.
36.
Chaudhary
S
,
Krishna
BM
,
Mishra
SK
. 
A novel FOXA1/ESR1 interacting pathway: A study of Oncomine breast cancer microarrays
.
Oncol Lett
2017
;
14
:
1247
64
.
37.
Fang
SH
,
Chen
Y
,
Weigel
RJ
. 
GATA-3 as a marker of hormone response in breast cancer
.
J Surg Res
2009
;
157
:
290
5
.
38.
Witkiewicz
AK
,
Balaji
U
,
Knudsen
ES
. 
Systematically defining single-gene determinants of response to neoadjuvant chemotherapy reveals specific biomarkers
.
Clin Cancer Res
2014
;
20
:
4837
48
.
39.
Stoica
GE
,
Franke
TF
,
Moroni
M
,
Mueller
S
,
Morgan
E
,
Iann
MC
, et al
Effect of estradiol on estrogen receptor-alpha gene expression and activity can be modulated by the ErbB2/PI 3-K/Akt pathway
.
Oncogene
2003
;
22
:
7998
8011
.
40.
Osborne
CK
,
Shou
J
,
Massarweh
S
,
Schiff
R
. 
Crosstalk between estrogen receptor and growth factor receptor pathways as a cause for endocrine therapy resistance in breast cancer
.
Clin Cancer Res
2005
;
11
:
865s
70s
.
41.
Presti
D
,
Quaquarini
E
. 
The PI3K/AKT/mTOR and CDK4/6 pathways in endocrine resistant HR+/HER2- metastatic breast cancer: biological mechanisms and new treatments
.
Cancers
2019
;
11
:
1242
.
42.
Citro
S
,
Miccolo
C
,
Meloni
L
,
Chiocca
S
. 
PI3K/mTOR mediate mitogen-dependent HDAC1 phosphorylation in breast cancer: a novel regulation of estrogen receptor expression
.
J Mol Cell Biol
2015
;
7
:
132
42
.
43.
Khaled
WT
,
Lee
SC
,
Stingl
J
,
Chen
XF
,
Ali
HR
,
Rueda
OM
, et al
BCL11A is a triple-negative breast cancer gene with critical functions in stem and progenitor cells
.
Nat Commun
2015
;
6
:
5987
.
44.
Yang
G
,
Zanudo
JGT
,
Albert
R
. 
Target control in logical models using the domain of influence of nodes
.
Front Physiol
2018
;
9
:
454
.
45.
Huang
S
,
Eichler
G
,
Bar-Yam
Y
,
Ingber
DE
. 
Cell fates as high-dimensional attractor states of a complex gene regulatory network
.
Phys Rev Lett
2005
;
94
:
128701
.
46.
Joo
JI
,
Zhou
JX
,
Huang
S
,
Cho
KH
. 
Determining relative dynamic stability of cell states using boolean network model
.
Sci Rep
2018
;
8
:
12077
.
47.
Chou
CW
,
Wu
MS
,
Huang
WC
,
Chen
CC
. 
HDAC inhibition decreases the expression of EGFR in colorectal cancer cells
.
PLoS One
2011
;
6
:
e18087
.
48.
Zhang
CH
,
Wang
J
,
Zhang
LX
,
Lu
YH
,
Ji
TH
,
Xu
L
, et al
Shikonin reduces tamoxifen resistance through long non-coding RNA uc.57
.
Oncotarget
2017
;
8
:
88658
69
.
49.
Muller
BM
,
Jana
L
,
Kasajima
A
,
Lehmann
A
,
Prinzler
J
,
Budczies
J
, et al
Differential expression of histone deacetylases HDAC1, 2 and 3 in human breast cancer–overexpression of HDAC2 and HDAC3 is associated with clinicopathological indicators of disease progression
.
BMC Cancer
2013
;
13
:
215
.
50.
Bicaku
E
,
Marchion
DC
,
Schmitt
ML
,
Munster
PN
. 
Selective inhibition of histone deacetylase 2 silences progesterone receptor-mediated signaling
.
Cancer Res
2008
;
68
:
1513
9
.
51.
Liu
H
,
Chen
S
,
Yao
X
,
Li
Y
,
Chen
CH
,
Liu
J
, et al
Histone deacetylases 1 and 2 regulate the transcriptional programs of nephron progenitors and renal vesicles
.
Development
2018
;
145
:
dev153619
.
52.
Linares
A
,
Dalenc
F
,
Balaguer
P
,
Boulle
N
,
Cavailles
V
. 
Manipulating protein acetylation in breast cancer: a promising approach in combination with hormonal therapies?
J Biomed Biotechnol
2011
;
2011
:
856985
.
53.
Luc
S
,
Huang
JL
,
McEldoon
JL
,
Somuncular
E
,
Li
D
,
Rhodes
C
, et al
Bcl11a deficiency leads to hematopoietic stem cell defects with an aging-like phenotype
.
Cell Rep
2016
;
16
:
3181
94
.
54.
Senawong
T
,
Peterson
VJ
,
Leid
M
. 
BCL11A-dependent recruitment of SIRT1 to a promoter template in mammalian cells results in histone deacetylation and transcriptional repression
.
Arch Biochem Biophys
2005
;
434
:
316
25
.
55.
Sankaran
VG
,
Menne
TF
,
Xu
J
,
Akie
TE
,
Lettre
G
,
Van Handel
B
, et al
Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A
.
Science
2008
;
322
:
1839
42
.
56.
Bauer
DE
,
Orkin
SH
. 
Hemoglobin switching's surprise: the versatile transcription factor BCL11A is a master repressor of fetal hemoglobin
.
Curr Opin Genet Dev
2015
;
33
:
62
70
.
57.
Moody
RR
,
Lo
MC
,
Meagher
JL
,
Lin
CC
,
Stevers
NO
,
Tinsley
SL
, et al
Probing the interaction between the histone methyltransferase/deacetylase subunit RBBP4/7 and the transcription factor BCL11A in epigenetic complexes
.
J Biol Chem
2018
;
293
:
2125
36
.
58.
Khaled
WT
,
Lee
SC
,
Stingl
J
,
Chen
X
,
Ali
HR
,
Rueda
OM
, et al
BCL11A is a triple-negative breast cancer gene with critical functions in stem and progenitor cells
.
Nat Commun
2015
;
6
:
5987
.