Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2020 Jul 16;15(7):e0235850. doi: 10.1371/journal.pone.0235850

c-Src functionality controls self-renewal and glucose metabolism in MCF7 breast cancer stem cells

Víctor Mayoral-Varo 1,#, Annarica Calcabrini 1,¤a,#, María Pilar Sánchez-Bailón 1,¤b,#, Óscar H Martínez-Costa 1, Cristina González-Páramos 1, Sergio Ciordia 2, David Hardisson 3,4,5, Juan J Aragón 1, Miguel Ángel Fernández-Moreno 1,6,7, Jorge Martín-Pérez 1,5,*
Editor: Antimo Migliaccio8
PMCID: PMC7365443  PMID: 32673341

Abstract

Deregulation of Src kinases is associated with cancer. We previously showed that SrcDN conditional expression in MCF7 cells reduces tumorigenesis and causes tumor regression in mice. However, it remained unclear whether SrcDN affected breast cancer stem cell functionality or it reduced tumor mass. Here, we address this question by isolating an enriched population of Breast Cancer Stem Cells (BCSCs) from MCF7 cells with inducible expression of SrcDN. Induction of SrcDN inhibited self-renewal, and stem-cell marker expression (Nanog, Oct3-4, ALDH1, CD44). Quantitative proteomic analyses of mammospheres from MCF7-Tet-On-SrcDN cells (data are available via ProteomeXchange with identifier PXD017789, project DOI: 10.6019/PXD017789) and subsequent GSEA showed that SrcDN expression inhibited glycolysis. Indeed, induction of SrcDN inhibited expression and activity of hexokinase, pyruvate kinase and lactate dehydrogenase, resulting in diminished glucose consumption and lactate production, which restricted Warburg effect. Thus, c-Src functionality is important for breast cancer stem cell maintenance and renewal, and stem cell transcription factor expression, effects linked to glucose metabolism reduction.

Introduction

Mammary gland stem cells are capable of self-renewal and to generate all differentiated progeny of the gland [1, 2]. Because of their long lifespan, they can accumulate mutations to become breast cancer stem cells (BCSCs). BCSCs are slow dividing and represent 1–2% of a tumor biopsy; nevertheless, they reproduce the tumor upon transplantation in nude mice [3]. Breast tumors exhibiting basal phenotype, like the triple negative breast cancers (TNBC), are usually highly metastatic with poor prognosis, resistance to chemotherapy, and are similar to the earliest mammary gland progenitor cells, supporting the hypothesis that therapeutic failure results from inefficient BCSCs targeting [4]. In contrast, luminal A human mammary epithelial cells produce tumors in a hormone-dependent manner. As they express estrogen and progesterone receptors, but not HER2 (human epidermal growth factor 2), and they are poorly /non-metastatic, the MCF7 cell line is a cellular model representative of this type of mammary tumors [5].

The Src-family tyrosine kinases (SFKs) acts as intracellular mediators for signaling pathways from growth factors, estrogen and cytokine receptors activation, etc., mediating pathways involved in proliferation, survival, differentiation, adhesion, migration, and invasion [68]. SFKs is overexpressed and/or aberrantly activated in human tumors [7, 9, 10]. In breast cancer, SFKs has been associated with induction, progression, and metastasis [8, 1116]. c-Src has been involved in glycolysis regulation and Warburg effect [1720], which is the elevated rate of glycolysis and lactate production in aerobic conditions by tumor cells [21]. As initially shown, c-Src/v-Src phosphorylates several glycolytic enzymes, including enolase and lactate dehydrogenase (LDH) [22]. In 3T3 fibroblast, v-src transfection leads to increased expression of glucose uptake [23]. c-Src phosphorylates Y10-LDH, increasing its activity, which promotes invasion, anoikis resistance, and metastasis of breast cancer cells [19]. Similarly, c-Src phosphorylates hexokinase 1 and 2 (HK1, HK2) augmenting their activity, which stimulates glycolysis, cell proliferation, migration, invasion, and tumorigenesis [20]. In human glioblastoma U87 and U251 cells, EGFR activation leads to c-Src stimulation and subsequent Y59-Cdc25A phosphorylation, which dephosphorylates pyruvate kinase (PKM2), promoting its interaction with β-catenin, its transactivation and Myc transcription that induces Glut-1 (glucose transporter 1), PKM2 and LDHA expression and, consequently, the Warburg effect and tumorigenesis [18].

We previously showed in MCF7 cells that conditional Tet-On expression of SrcDN (Src-Dominant Negative) leads to inhibition of proliferation, attachment, spreading and migration in cultured cells. Inoculation of cells in nude mice generates tumors, while induction of SrcDN expression significantly reduces their tumorigenesis, and causes regression when induced in established tumors [24]. The Src-DN is a chicken paralog of c-Src with two mutations (c-Src-K295M/Y527F). The mutation K295M prevent binding of ATP to c-Src, avoiding its tyrosine kinase activity, the second point mutation Y527F at the C-terminal of the molecule, that is phosphorylated by CSK (C-terminal Src Kinase), provokes that c-Src is forced to maintain its open conformation, which implies full functionality of the SH2 and SH3 domains [68].

Here we addressed the question as to whether interfering SFKs functionality by SrcDN expression directly affects MCF7-BCSC renewal. Thus, here we isolate by FACS the enriched population of BCSCs (ESA+-CD44+-CD24-/Low cells, from now on CD24-) and the so-called tumor-differentiated cells (ESA+-CD44+-CD24+ cells, from now on CD24+) from MCF7-Tet-On-SrcDN [24], and test their capacity of self-renewal. Our findings show that c-Src is important for mammospheres self-renewal, which is associated with an alteration in glucose metabolism.

Materials and methods

Reagents

Information about antibodies used in these experiments is in S2 Table in S1 File (Antibodies). BCA protein assay (Termofisher); Acrylamide/bis 40% solution, 29:1 (3.3% C), ammonium persulfate and clarity immunoblot ECL substrate (Bio-Rad); trypan blue, doxycycline (Doxy), BSA, puromycin, and insulin (Sigma-Aldrich); EGF, and bFGF (PeproTech EC Ltd., London, UK); G418, versene, and trypsin (Invitrogen); tetracycline-free fetal bovine serum (Tet-Free-FCS, PAA Laboratories GmbH). Other chemical reagents and enzymes used were of analytical grade and purchased from Roche, GE Healthcare, or Sigma-Aldrich/Merck.

Cells and culture

MCF7 (HTB-22) were from ATCC. Cells were mycoplasma free and authenticated by short-tandem-repeat analysis (GenePrintR 10 System from Promega, and GeneMapper v3.7 STR profile analysis software, Life Technologies) (see Supplementary Information). Profiles obtained were checked against public databases ATCC and DSMZ. MCF7-Tet-On-SrcDN cells bearing a Doxy-inducible SrcDN (avian c-Src-K295M/Y527F) were previously generated [24] and maintained in DMEM, 5% Tet-Free-FCS, 2 mM glutamine, 100 IU/mL penicillin, 100 μg/mL streptomycin, and 0.2 mg/mL G418, 0.5 μg/mL puromycin for selection.

Isolation and culture of CD24- and CD24+ subpopulations from MCF7 cells

The enriched subpopulations of CD24- and CD24+ cells derived from MCF7-Tet-On-SrcDN were isolated by fluorescence-activated cell sorting (FACS), as described [3]. Briefly, 1x107 cells were detached from the culture plates with versene (5 min, 37°C), and then simultaneously labeled with antibodies to ESA-FITC, CD44-APC, CD24-PE, and with their respective isotypic immunoglobulins. Cells were washed and subjected to FACS with a FACS-Vantage cell sorter (BD, San Jose CA) equipped with an argon ion laser (emission at 488 nm) and a He-Ne laser (emission at 633 nm). Cells were gated on forward and side scatters properties and specific fluorescent signals were collected using 530 nm (FITC), 575 nm (PE) and 660 nm (APC) bandpass filters. About 1.6% of the cell population showed the CD24- (ESA+-CD44+-CD24-) phenotype and were isolated. In parallel, the CD24+ (ESA+-CD44+-CD24+) cells were also isolated as described [3].

CD24- cells were maintained in mammosphere media (1:1 DMEM/HAM’S F12, 2 mM glutamine, 100 IU/mL penicillin, 100 mg/mL streptomycin, 5 μg/mL Insulin, 20 ng/mL EGF, 10 ng/mL bFGF, 4 mg/ml BSA), and 0.2 mg/mL G418, 0.5 μg/mL puromycin to maintain selection for c-SrcDN expression [24], and they were grown in suspension in 6-multiwell ultralow attachment plates (Falcon 351146, BD); CD24+ cells were cultured in DMEM, 5% Tet-Free-FCS, 2 mM glutamine, 100 IU/mL penicillin, 100 mg/mL streptomycin, and 0.2 mg/mL of G418 and 0.5 μg/mL puromycin in standard plates (Falcon, BD). Also, another protocol was employed for BCSCs enrichment to enrich for BCSCs from MCF7-Tet-On-SrcDN, single cells obtained after trypsinization of adherent cultures were plated at 1x103 cells/mL and cultured as described above for CD24- [25].

Sphere Formation Efficiency (SFE)

SFE from MCF7-Tet-On-SrcDN was determined as described [26]. Briefly, single cell suspensions of adherent cultures were plated in 6-well ultralow attachment plates (Falcon, Corning Life Science) at 2x103 cells/well and maintained in serum-free DMEM/F12 media (1:1), BSA (4 mg/mL), EGF (20 ng/mL) and bFGF (20 ng/mL), insulin (5 μg/mL), hydrocortisone (5 μg/mL) to obtain mammospheres. Fifteen days later, mammospheres were dissociated into single cells that were plated in 6-well ultralow attachment plates at 2x103 cells/well in mammosphere culture media, 3 wells without Doxy (Control) and 3 wells with Doxy (2 μg/mL). During the experiments Doxy was renewed every 3 days in cultures. Each mammosphere generation was cultured about 15 days. Mammospheres (sphere-like structures with diameter ≥ 50 μm) were clearly detected by optical phase contrast microscope (Nikon-Eclipse TS100, 4x magnification). For mammosphere dissociation, they were collected in a sterile tube and allow them to sediment, the media was removed with a pipette and trypsinized in Tris-Saline pipetting up and down to facilitate mammosphere dissociation, cells were then collected by centrifugation to remove trypsin, washed with mammosphere media and counted. Single cells were seeded again for mammosphere formation in 6-well ultralow attachment plates at 2x103 cells/well. The experiment ended at 3rd generation of mammosphere formation. The cells were growth in absence (Control) or presence (Doxy) of 2 μg/ml Doxy for three generations, and renewed every three days. SFE was calculated as number of spheres formed per number of seeded cells and expressed as % means ± SD. The SFE experiments were repeated 5 times, each one of them was carried out in triplicates (p<0.05*).

Immunohistochemistry of mammospheres

Mammospheres derived from MCF7-Tet-On-SrcDN were fixed in 4% formaldehyde solution. Fixed samples were paraffin embedded, and 5 μm sections were analyzed by hematoxylin/eosin, and E-cadherin and P-cadherin staining were performed using Envision method [27].

Immunoblotting analysis

Preparation of cell lysates from mammospheres and immunoblotting analyses were carried out as previously described [24]. Briefly, mammospheres were collected by centrifugation at 200xg 5 min at room temperature, washed twice with cool PBS and lysed at 4°C with lysis buffer [10 mM Tris–HCl (pH 7.6), 50 mM NaCl, 30 mM sodium pyrophosphate, 5 mM EDTA, 5 mM EGTA, 0.1% SDS, 1% Triton X-100, 50 mM NaF, 0.1 mM Na3VO4, 1 mM PMSF, 1 mM benzamidine, 1 mM iodoacetamide and 1 mM phenantroline]. Cell lysates were obtained by centrifugation at 21,380xg for 30 min at 4°C; protein concentration in the supernatant was determined by BCA protein assay (Pierce, Rockford, IL), and lysates were adjusted to equivalent concentrations with lysis buffer. Aliquots of 10–40 μg of total cell lysate were then separated on SDS–PAGE. Proteins were transferred to PVDF membranes that were blocked overnight at 4°C with 5% non-fat milk in TTBS (TBS with 0.05% Tween-20). Incubation with primary specific antibodies was carried out overnight at 4°C, and horseradish peroxidase-conjugated secondary antibodies in blocking solution for 1 h at room temperature. Immunoreactive bands were visualized by ECL kit.

Quantitative proteomic analysis

Briefly, MCF7-Tet-On-SrcDN derived mammospheres at 3rd generation from two independent experiments cultured either in the absence (Control) or presence of Doxy (2 μg/mL) were collected by centrifugation, washed with PBS, and lysed in 9 M urea, 2 M thiourea, 5% CHAPS, and 2 mM TCEP. The samples were precipitated by methanol/chloroform and quantified by Pierce 660nm Protein Assay. Then 30 μg of protein from each sample were trypsinized, and the resulting peptides were labeled using iTRAQ 4-plex reagent (SCIEX, Foster City, CA, USA) according to the manufacturer's instructions and named as follows: Control-1, 114-tag; Doxy-1, 115-tag; Control-2, 116-tag; Doxy-2, 117-tag. Samples were pooled, evaporated to dryness and frozen. The pool was separated into 5 fractions in a reverse phase C18 chromatography at basic pH and cleaned with a StageTip-C18. The 5 fractions were quantified by fluorimetry, and 1 μg of each fraction was separated by C-18 reverse phase chromatography during 150 min. The eluted peptides passed then into 5600 Triple-TOF MS (SCIEX, Foster City, CA). The raw data of the combined 5 fractions was exported to the search-engine Mascot Server v2.5.1 (Matrix Science, London, UK) confronted to the Homo sapiens UniProt database. Peptide mass tolerance was set to ± 25 ppm for precursors and 0.05 Da for fragment masses. Frequency distribution histograms of protein ratios were obtained into Excel 2010. Log2 peptide ratios followed a normal distribution that was fitted using least squares regression. Mean and standard deviation values derived from the Gaussian fit were used to calculate p-values and quantification of False Discovery Rates (FDR). The confidence interval for protein identification was set to ≥ 95% (p<0.05) and only peptides with an individual ion score above the 1% False Discovery Rates (FDR) threshold were considered correctly identified. Only proteins having at least two quantitated peptides were considered in the quantitation. A 5% quantitation FDR threshold was estimated to consider the significant differentially expressed proteins.

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE [28] partner repository (Project accession number: PXD017789, project DOI:10.6019/PXD017789).

Enzymatic activity assays

Mammospheres at 3rd generation derived from MCF7-TET-ON-SrcDN cells were seeded into a six-well plate and cultured with fresh medium -/+ Doxy (2 μg/mL) for 12–24 h. Cells were harvested by centrifugation, washed twice with PBS and preincubated for 10 min in PBS. Then, cells were collected by centrifugation, suspended in fresh culture medium and incubated for another 4 h. Cells were harvested by centrifugation at room temperature at 0 and 4 h incubation and treated with 200 μL lysis buffer (Tris-HCl (pH 7.5), 1 mM EDTA, 0.5 mM dithiothreitol, 0.2% Triton X-100, 0.2 mg/mL deoxycholate, 0.2 mM phenylmethylsulfonyl fluoride, and 2.5 μg/mL leupeptin), and homogenized at 4°C by pipetting up and down. The homogenate was centrifuged at 21,380xg for 15 min at 4°C and the supernatant was used for enzymatic activity measurements. Hexokinase (HK) activity was determined spectrophotometrically at 340 nm by following the G6PDH dependent conversion of NADP to NADPH. The assay mixture contained 100 mM Tris-HCl (pH 8.0), 0.5 mM EDTA, 10 mM MgCl2, 5 mM MgATP, 2 mM glucose, 0.5 mM NADP+ and 10 U/mL glucose 6-phosphate dehydrogenase (G6PDH) in a final volume of 1 mL. HK activities were calculated according to the slope of the resulting curves in the log phase. The initial velocity of lactate dehydrogenase (LDH) was determined by measuring spectrophotometrically the NADH consumption at 340 nm in a 1 mL reaction mixture (50 mM HEPES pH 7.2, 100 mM KCl, 5 mM MgCl2, 0.15 mM NADH, and 2 mM pyruvate). The slopes of the initial linear decrease were used to calculate initial velocities. Pyruvate kinase (PK) activity was measured by following the production of pyruvate. The reaction mixture consisted of 50 mM HEPES (pH 7.2), 100 mM KCl, 5 mM MgCl2, 0.15 mM NADH, 1 mM MgADP, 2 mM phosphoenolpyruvate, and 1 U/mL of LDH as the auxiliary enzyme in a final volume of 1 mL. PK activity was determined spectrophotometrically as above by recording the change of 340 nm absorbance produced by NADH oxidation. One unit of enzyme activity is defined as the amount of enzyme catalyzing the conversion of 1 μmol of substrate/min at 25°C. Enzymatic activities of different samples were normalized against the corresponding protein concentrations in the lysates.

Lactate production and glucose consumption assays

Cells were treated as above. At 0 and 4 h incubation, cells were harvested by centrifugation at room temperature, and the culture medium was removed and stored at -70°C for lactate production and glucose consumption measurements. Cell pellets were homogenized in ice-cold 8% (v/v) perchloric acid in 40% ethanol. After centrifugation at 21,380xg for 15 min at 4°C, the supernatant was neutralized to pH 7.0 with a 2 M KOH and 0.5 M triethanolamine solution, the mixture was incubated on ice for 15 min, and precipitated KClO4 was removed by centrifugation. Glucose and lactate content in culture medium and levels of intracellular lactate were assessed by using standard enzymatic techniques [29]. Briefly, lactate was determined in a reaction containing 0.5 M Glycine, 0.2 M hydrazine, 3.4 mM EDTA, 1.5 mM NAD+, and 10 U/mL of LDH, pH 9.5. The reaction mixture for glucose measurement consisted of 0.1 M triethanolamine, 5 mM MgCl2, 0.5 mM NADP+, 1 mM ATP, 1 U/mL hexokinase and 3 U/mL G6PD, pH 7.4. The function of the glycolytic pathway was evaluated by the amount of lactate produced per hour after 4 h incubation. Glucose consumption rate (per hour) was calculated considering the difference between the amount in the medium at 0 h and that recovered in the medium after 4 h incubation. Glucose and lactate content were normalized to the protein concentration of cells cultured under similar conditions and obtained as described above for enzymatic activity.

Oxygen consumption and determination of ATP concentration

Mammospheres at 3rd generation cultured in the absence or presence of 2 μg/mL Doxy were dissociated to single-cell suspension as for SFE, resuspended in mammospheres media with or without pyruvate and without BSA to 106 cells/mL and the oxygen consumption of 2x106 cells was measured by high-resolution respirometry with the OROBOROS oxygraph-2k in a standard configuration, at 37°C and a 750-rpm stirrer speed as described [30]. The protocol includes the determination in a sequential manner of the aerobic metabolic activity under routine culture conditions with the physiological substrates in culture medium (Cr), the oligomycin-inhibited leak rate of respiration after inhibition of ATP synthase with 2 μg/mL oligomycin (CrO) and the maximum respiratory capacity of uncoupled mitochondria in nonpermeabilized cells (CrU), obtained by sequential addition of 0.5 μM boluses of trifluoro-carbonylcyanide phenylhydrazone (FCCP). The protonophore FCCP dissipates the proton inner membrane gradient making ETC to function at its maximal rate since oxygen and protons are not limiting for the ETC Complex IV catalysis. All values, including physiological conditions and the maximal respiratory capacity, are derived by subtracting non- mitochondrial respiration from the FCCP rate.

Measurements of total ATP concentration were made in mammospheres that were maintained for at least 15 days -/+ Doxy (2 μg/mL) at 0 and 4 h, as previously described [31], following instructions of the ATP Bioluminescence Assay Kit CLS II (Roche, Ref. 11699695001).

Statistical analyses

Mean values, standard deviation and statistical significance between data from two different experimental conditions were determined by two-tail Student t-test. In all figures, data represent the Mean from at least three separate biological repeats done in at least triplicates each ± SD (standard deviation), *p <0.05, **p < 0.01, and ***p < 0.001.

Results

Isolation and characterization of an enriched population of BCSCs from MCF7 cells

To determine the role of SrcDN in the self-renewal of the enriched subpopulation of BCSCs, we isolated CD24- (ESA+-CD44+-CD24-) and CD24+ (ESA+-CD44+-CD24+) cell populations derived from MCF7-Tet-On-SrcDN by FACS. Cells were first sorted as ESA+ and then for being CD44+-CD24- (1.6%), while the CD44+-CD24+ represented 98.4% of total cell population (Fig 1A, isotype controls shown in S1 Fig in S1 File). CD24- cells formed mammospheres (Fig 1B) that after staining with hematoxylin/eosin showed a monolayer cell-structure with their nuclei oriented towards the lumen. Furthermore, mammospheres expressed E-cadherin, but not P-cadherin (Fig 1B). These results suggested that both CD24- (mammospheres) and CD24+ cells maintained the epithelial phenotype of the MCF7 total population.

Fig 1. Cell sorting of CD24- and CD24+ subpopulations of MCF7 cells.

Fig 1

(A) Histograms of ESA-FITC positive cells that were later sorted for CD44-APC positive and CD24-PE negative (1.6%). (B) Microphotograph under light microscope of mammospheres from CD24- cells in culture. Representative images of spheres stained with hematoxylin/eosin, E-cadherin or P-cadherin. Scales bars are included. (C) Immunoblot of total c-Src, SrcDN, p-Y397-Fak/Fak, and E-cadherin, using α-Tubulin expression as loading control from CD24- cells forming mammospheres and CD24+ cells in absence (Control) or presence of Doxy (2 μg/mL) for induction of SrcDN during three passages. The ratios of the proteins Total c-Src, SrcDN, and E-cadherin with the loading control α-Tubulin, or pY397-Fak/Fak were calculated and referred to -Doxy (Control) considered as 1. These are representative results from 3 independent experiments.

To determine the functionality of SrcDN, protein extracts from mammospheres from CD24- and from CD24+ cell subpopulations were blotted for pY397-Fak, showing an increased phosphorylation upon SrcDN induction. This is consistent with previous results showing that SH2 domain from SrcDN binds pY397-Fak protecting from dephosphorylation [24, 32, 33], and with the reduction of pY925-Fak, p-Y-p130CAS, pY118-Paxillin phosphorylation by c-Src in MCF7 cells [24]. Expression of E-cadherin remained unaltered in CD24- and CD24+ upon SrcDN induction (Fig 1C). SrcDN expression was tested by detecting total c-Src and avian SrcDN (Fig 1C).

c-Src functionality is necessary for mammospheres self-renewal

To characterize the role of c-Src in the enriched subpopulation of BCSC self-renewal, we analyzed whether induction of SrcDN expression altered formation of mammospheres from MCF7-Tet-On-SrcDN. Self-renewal capacity was confirmed by three successive generations of mammospheres, where a progressive enrichment of mammosphere-forming cells (CD24-) was observed (S2 Fig in S1 File). Then, we determined Sphere Formation Efficiency (SFE) upon induction of SrcDN expression from the first generation and found that it significantly reduced SFE as compared to control (-Doxy) at the 3rd generation (Fig 2A). This effect was clearly evident from the 1st generation (S2 Fig in S1 File), although no morphological alterations were observed upon SrcDN induction (data not shown). Consistently, cyclin D1 expression at the 3rd generation was also reduced. However, SrcDN expression did not appear to cause apoptosis, as PARP degradation was undetectable. To determine the nature of cells that form mammospheres, we have dissociated mammospheres at the 3rd generation, labelled cells with anti-CD24-PE and CD44-APC monoclonal antibodies, as well as with isotypic immunoglobulins for control, and analyzed them by flow cytometry. S3 Fig in S1 File showed that in control mammospheres most of cells are CD44+-CD24-. In contrast, in Doxy-treated mammospheres a large number of cells were CD44+-CD24+. In addition, the analysis of the expression level of these markers (MFC, mean fluorescence channel) indicates that, along with an increase of expression of CD24, treated mammospheres show a reduced expression of CD44. Moreover, expression of pluripotency-related transcription factors involved in self-renewal Nanog, Oct3/4, and stem cell markers ALDH1, and CD44 [3436] were also reduced (Fig 2B). Together, these results suggested that c-Src functionality is relevant for MCF7-mammosphere renewal.

Fig 2. Self-renewal of mammospheres derived from MCF7.

Fig 2

(A) The Sphere Formation Efficiency (SFE) was determined in MCF7-Tet-On-SrcDN. Thus, single cell suspensions from adherent cells were plated in 6-well ultralow attachment plates at 2x103 cells/well and cultured in mammosphere medium. Fifteen days later, mammospheres were dissociated into single cells that were divided into two groups: Control (-Doxy) and Doxy treated (2 μg/mL). Doxy-treatment was maintained for three generations, and renewed every 3 days. SFE at 3rd generation was calculated as number of spheres formed per number of seeded cells and expressed as % means ± SD. The SFE experiments were repeated 5 times, each one of them was carried out in triplicates. (p<0.05*). (B) Total cell extracts from the 3rd generation of mammospheres treated as above (panel A) were used to determine by immunoblotting expression of cyclin D1, PARP, with α-Tubulin, as a loading control, and SrcDN, Nanog, Oct3/4, ALDH1, and CD44, employing GAPDH or β-Actin, as a loading control. The net quantification of the gel bands after subtracting the background was carried out with ImageJ program and expressed in arbitrary units. These are representative results from 3 independent experiments. The ratios of the proteins SrcDN, Nanog, Oct3/4, ALDH1, and CD44 and their loading controls GAPDH or ß-Actin, or pY10-LDHA/LDHA were calculated and referred to -Doxy (Control) considered as 1.

Proteomic analyses of mammospheres

Since c-Src functionality is important for SFE of MCF7-mammospheres, we analyzed whether SrcDN induction quantitatively modified protein expression. Proteomic analyses identified 2,759 proteins with at least two peptides, full data available at Proteome Xchange database (project accession number: PX017789, project DOI: 10.6019/PX017789). From those, only 101 proteins were differentially expressed with an FDR < 5% (S1 Table in S1 File). In the Fig 3 only proteins having at least two quantitated peptides and <5% quantitation FDR with the same tendency in both experiments were included. Thus, induction of SrcDN by Doxy (2 μg/mL) diminished the levels of fifteen proteins, and increased the levels of four of them (Fig 3, and S4 Fig in S1 File). The most increased protein in Doxy/Control is c-Src, which most likely should correspond to SrcDN induced by Doxy-treated mammospheres, as the identity of Homo Sapiens and Gallus-Gallus c-Src at the amino acid sequences is 94%. Results obtained were submitted to GSEA and found that glycolysis was significantly reduced in MCF7-mammospheres upon SrcDN induction (S5 Fig in S1 File), which was consistent with data from Fig 3.

Fig 3. Quantitative proteomic analysis of mammospheres derived from MCF7-Tet-On-SrcDN.

Fig 3

The analysis was carried out in two independent experiments from the total cell extract of the 3rd generation mammospheres either untreated (Control) or Doxy-treated. Only proteins having at least two quantitated peptides and <5% quantitation FDR in both experiments are included in the figure.

Role of c-Src in the regulation of glycolysis in MCF7-mammospheres

As glycolytic pathway was inhibited in mammospheres upon SrcDN induction, we investigated whether glucose consumption was altered, and found that it was significantly reduced in mammospheres expressing SrcDN (Fig 4A), which parallels a reduction in Glut-1 levels (Fig 4D), the major glucose transporter in tumor cells [37]. In cancer cells, Src enhances HK enzymatic activity, a rate-limiting enzyme of glycolysis, by direct phosphorylation [20]. Since SrcDN is a kinase-dead mutant and competes with endogenous Src kinases [38], HK enzymatic activity was significantly inhibited in Doxy-treated mammospheres (Fig 4C). In addition, HK2 levels were also diminished (Fig 4D), supporting proteomic results (Fig 3). However, we were unable to detect HK2 tyrosine phosphorylation. Similarly, PKM enzymatic activity was also inhibited upon SrcDN expression (Fig 4C). In head and neck and in breast cancer cells, Src phosphorylates and enhances LDHA [19]. We tested for SrcDN effects on LDHA enzymatic activity, expression, and phosphorylation. In Doxy-treated mammospheres, LDH activity was significantly reduced (Fig 4C), as it was pY10-LDHA/LDHA ratio (Fig 4D). It is well established that c-Myc and HIF-1 promote the expression of enzymes implicated in glycolysis regulation, including HK2 and LDHA [39, 40, 41]. Thus, we analyzed c-Myc and HIF-1 expression level in Control (Doxy-untreated) and Doxy-treated (2 μg/mL) 3rd mammosphere generations by SYBR Green-q-RT-PCR and by immunoblotting (see Supplementary Material). SrcDN induction did not modify c-Myc expression both at RNA and protein level while HIF-1 RNA expression was significantly reduced. In addition, we were not able to detect HIF-1 protein expression by immunoblotting as the protein is extremely labile (S6 Fig in S1 File). The decrease in glucose consumption was paralleled by a nearly stoichiometric reduction of lactate production (lactate/glucose ratio of 1.45 ± 0.12 and 1.58 ± 0.09 for wild-type and SrcDN-mammospheres, respectively). Thus, the production of lactate from glutaminolysis is very limited. Interestingly, although lactate production diminished, intracellular accumulation of this metabolite was increased (Fig 4B), a result consistent with the reduced expression of MCT-1 (SLC16A1, proton coupled monocarboxylate transporter 1) in Doxy-treated mammospheres (Fig 4D), the major lactate transporter in tumor cells [42].

Fig 4. Expression of SrcDN reduced glycolysis in MCF7 mammosphere-derived cells.

Fig 4

Control and Doxy-treated (2 μg/mL) mammospheres from the 3rd generation were used to study glycolysis. (A) Determination of glucose consumption. (B) Lactate production, intracellular lactate. (C) Enzymatic activity of hexokinase (HK), pyruvate kinase (PK), lactate dehydrogenase (LDH). (***p<0.001). (D) Expression of Glut-1, total c-Src, HK2, pY10-LDHA, LDHA, and MCT-1, was determined by immunoblotting, employing ß-Actin as a loading control. These are representative results from 3 independent experiments. The ratios refer to -Doxy considered as 1.

Effects of SrcDN on oxygen consumption and ATP production

Results showed that SrcDN expression inhibited glycolysis and altered the lactate levels in mammospheres. We next determined the effects of SrcDN in mitochondrial respiration capacity, either in culture without pyruvate, and in normal culture media, which containing saturating concentrations of pyruvate to the uncoupled mitochondria to glycolysis, pushing oxygen consumption to its highest. Basal oxygen consumption of cells dissociated from mammospheres in media containing pyruvate was unmodified by SrcDN expression, while the maximum was significantly inhibited (Fig 5A). Similarly, oxygen consumption carried out in media without pyruvate was also unaltered in basal conditions, but the maximum was highly reduced in mammospheres expressing SrcDN (Fig 5A, and S7 Fig in S1 File: OCR profiles). Furthermore, analyses of ATP production did not differ between cells dissociated from mammospheres expressing SrcDN and control cells (Fig 5B). Neither alteration in ROS level, nor modification of catalase or MnSOD expression was observed upon SrcDN induction in mammospheres, as compared to controls (Doxy-untreated) (S8 Fig in S1 File).

Fig 5. Effects of SrcDN on oxygen consumption, and ATP production in MCF7 mammosphere-derived cells.

Fig 5

(A) Oxygen consumption in normal media containing pyruvate or in media without pyruvate and (B) ATP production, they were determined in cells obtained from Control (Doxy-untreated) and Doxy-treated (2 μg/mL) 3rd generation mammospheres.

Discussion

Cancer stem cells, which represent only a small subpopulation in a tumor, possess the ability of self-renewal and to form new tumors in nude mice. In breast cancer, BCSCs are defined by expression of ESA, CD44 and low CD24 expression (ESA+/CD44+/CD24-), and high tumorigenic capabilities [3]. In this context, the low percentage of BCSCs (CD24- cells) that we found here, correlates with previous observations [3] and confirms that the isolation of the CD24- cells was properly done.

In this study we analyzed the effects of the induction of a dominant-negative mutant of the protooncogene c-Src, the SrcDN. We found that disrupting c-Src pathways by induction of SrcDN expression reduced the self-renewal capabilities (SFE) of the enriched population of BCSCs derived from MCF7. Interestingly, cytometric analyses of SrcDN expressing mammosphere cells showed a reduction of CD44 and increased CD24 expression as compared to control mammosphere cells (non-expressing SrcDN). In addition, SrcDN induction did not induce apoptosis as PARP degradation did not occur, instead cyclin D1 expression was reduced, while Myc expression was unaltered. In this respect, we previously observed that SrcDN expression and selective c-Src tyrosine-kinase inhibitors diminish cyclin D1 expression and cell cycle progression at G1-phase in breast cancer cells [24, 43, 44]. Moreover, these mammosphere cells also showed expression of Oct3/4, Nanog pluripotency-associated transcription factors, ALDH1, and CD44 stem-cell markers [3436], which were reduced upon SrcDN induction. These pluripotency-associated transcription factors are required for embryonic stem cell self-renewal [34], and they are also involved in tumorigenesis and metastasis [45]. Specifically, MDA-MB-231 cells exhibit high expression of Oct3/4, and low levels of Nanog compared to MCF7 and T47D [46]. Collectively, our results suggest that c-Src functionality is essential for the self-renewal ability of BCSCs derived from MCF7 to grow in vitro as mammospheres.

MCF7 cells have been used as a model for Luminal A type of hormonal-dependent breast tumors, they have epithelial characteristics, they express E-cadherin, and are low or non-metastatic [5]. Here, we showed that mammosphere-forming cells derived from MCF7, which represented about 1.6% of total cell population, maintained their epithelial phenotype as they showed E-cadherin, but not P-cadherin staining.

Quantitative proteomic analyses of untreated and Doxy-treated mammospheres identified up to 2.759 proteins; of these proteins, 101 were differentially expressed (FDR < 5%), but only 19 of them showed same tendency in two independent experiments. Among those proteins, 15 showed a reduced expression upon SrcDN induction. The gene set enrichment analysis (GSEA) of the reduced proteins revealed an inhibition of the glycolytic pathway in these tumor-initiating cells. Thus, SrcDN restricts the Warburg effect in MCF7-mammospheres, as it reduced expression of Glut-1, glucose consumption, and expression and activity of HK2, PKM, LDHA, and lactate production. Since HIF-1 and Myc are known transcription factors involved in the control of HK2 and LDHA expression [3941], we tested whether they were modulated by induction of SrcDN. While SrcDN inhibited expression of mRNA expression of HIF-1, Myc expression both at mRNA and protein levels was not altered. In human colon adenocarcinoma LS174T and in murine melanoma B16-F10 cell lines Warburg effect requires expression of both LDH A and B [47]. Interestingly, while LDH B is expressed in TNBC cells, it is undetectable in luminal A cells, such as MCF7 [48]. Since MCT-1 expression was also inhibited upon SrcDN induction in MCF7-mammospheres, lactate accumulated intracellularly. It has been recently shown that SrcDN reduced glucose catabolism required for proliferation, migration, invasion, and tumorigenesis in cancer cells [49]. c-Src phosphorylates HK1 and HK2, rate-limiting enzymes of glycolysis, and enhances their catalytic activity [20]. Moreover, c-Src also phosphorylates LDHA at Y10, increasing its activity, which correlates with metastasis progression of clinical tumor samples of breast cancer [19], while as shown here, the dominant-negative variant of c-Src, the SrcDN counteracted these effects. In contrast, the basal oxygen consumption, ATP production, ROS, catalase, and MnSOD levels were unmodified by SrcDN expression in mammospheres, indicating that under basal conditions, the mitochondrial functionality was unaltered by SrcDN expression.

These data strongly suggest that c-Src functionality is important for the self-renewal capacity in MCF7 breast cancer stem cells, linked to glucose metabolism.

Conclusions

The findings presented in our study support the role of c-Src in the self-renewal of the enriched subpopulation of MCF7 breast cancer stem cells (BCSCs). Furthermore, results showed that c-Src functionality is a requisite for BCSCs. Src regulates Warburg effect in BCSCs, which in turn somehow controls stem-cell factors mediating stem-cell renewal. Elucidating the pathways that regulate BCSC functionality is fundamental to achieve their specific targeting.

Supporting information

S1 File

(PDF)

Acknowledgments

Authors are grateful to L. del Peso for the GSEA of proteomics, S. Alcalá, B. Sainz, S. Guerra, C. Gamallo, P. Lastres, M.C. Mena, E. Martín-Forero, and J. Pérez for their contributions and support, L. Boscá, and J. Gonzalez-Castaño for their comments, suggestions, and reagents. Jorge Martín-Pérez and David Hardisson are members of the GEICAM (Grupo Español de Investigación en Cáncer de Mama).

Data Availability

Data have been uploaded to Proteome Xchange database (Project accession number: PXD017789, project DOI: 10.6019/PXD017789).

Funding Statement

This work has been supported by grand SAF2016–75991-R (MINECO, AEI/FEDER, UE) to Jorge Martín-Pérez and ISCIII [grand PI 16/00789] to Miguel Ángel Fernández-Moreno. Víctor Mayoral-Varo was supported by the grand SAF2016–75991-R (MINECO, AEI/FEDER, UE). We acknowledge support for publication fee by the CSIC Open Access Publication Support Initiative through its Unit for Information Resources for Research (URICI).

References

  • 1.Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, Asselin-Labat ML, et al. Generation of a functional mammary gland from a single stem cell. Nature. 2006;439(7072):84–8. 10.1038/nature04372 . [DOI] [PubMed] [Google Scholar]
  • 2.Stingl J, Eirew P, Ricketson I, Shackleton M, Vaillant F, Choi D, et al. Purification and unique properties of mammary epithelial stem cells. Nature. 2006;439(7079):993–7. 10.1038/nature04496 . [DOI] [PubMed] [Google Scholar]
  • 3.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100(7):3983–8. 10.1073/pnas.0530291100 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Marotta LL, Polyak K. Cancer stem cells: a model in the making. Curr Opin Genet Dev. 2009;19(1):44–50. 10.1016/j.gde.2008.12.003 . [DOI] [PubMed] [Google Scholar]
  • 5.Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011;13(4):215 Epub 2011/09/03. 10.1186/bcr2889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Thomas SM, Brugge JS. Cellular functions regulated by Src family kinases. Ann Rev Cell Dev Biol. 1997;13:513–609. [DOI] [PubMed] [Google Scholar]
  • 7.Guarino M. Src signaling in cancer invasion. J Cell Physiol. 2010;223(1):14–26. Epub 2010/01/06. 10.1002/jcp.22011 . [DOI] [PubMed] [Google Scholar]
  • 8.Espada J, Martin-Perez J. An Update on Src Family of Nonreceptor Tyrosine Kinases Biology. Int Rev Cell Mol Biol. 2017;331:83–122. 10.1016/bs.ircmb.2016.09.009 . [DOI] [PubMed] [Google Scholar]
  • 9.Johnson FM, Gallick GE. SRC family nonreceptor tyrosine kinases as molecular targets for cancer therapy. Anticancer Agents Med Chem. 2007;7(6):651–9. Epub 2007/11/30. 10.2174/187152007784111278 . [DOI] [PubMed] [Google Scholar]
  • 10.Wheeler DL, Iida M, Dunn EF. The role of Src in solid tumors. Oncologist. 2009;14(7):667–78. Epub 2009/07/08. theoncologist.2009-0009 [pii] 10.1634/theoncologist.2009-0009 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Guy CT, Muthuswamy SK, Cardiff RD, Soriano P, Muller WJ. Activation of the c-Src tyrosine kinase is required for the induction of mammary tumors in transgenic mice. Genes Dev. 1994;8(1):23–32. 10.1101/gad.8.1.23 [DOI] [PubMed] [Google Scholar]
  • 12.Verbeek BS, Vroom TM, Adriaansen-Slot SS, Ottenhoff-Kalff AE, Geertzema JG, Hennipman A, et al. c-Src protein expression is increased in human breast cancer. An immunohistochemical and biochemical analysis. J Pathol. 1996;180(4):383–8. [DOI] [PubMed] [Google Scholar]
  • 13.Finn RS. Targeting Src in breast cancer. Ann Oncol. 2008;19(8):1379–86. Epub 2008/05/20. 10.1093/annonc/mdn291 . [DOI] [PubMed] [Google Scholar]
  • 14.Zhang XH, Wang Q, Gerald W, Hudis CA, Norton L, Smid M, et al. Latent bone metastasis in breast cancer tied to SRC-dependent survival signals. Cancer Cell. 2009;16(1):67–78. Epub 2009/07/04. 10.1016/j.ccr.2009.05.017 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhang S, Huang WC, Zhang L, Zhang C, Lowery FJ, Ding Z, et al. SRC family kinases as novel therapeutic targets to treat breast cancer brain metastases. Cancer Res. 2013;73(18):5764–74. 10.1158/0008-5472.CAN-12-1803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527(7578):329–35. 10.1038/nature15756 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.DeNicola GM, Cantley LC. Cancer's Fuel Choice: New Flavors for a Picky Eater. Mol Cell. 2015;60(4):514–23. Epub 2015/11/23. 10.1016/j.molcel.2015.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Liang J, Cao R, Zhang Y, Xia Y, Zheng Y, Li X, et al. PKM2 dephosphorylation by Cdc25A promotes the Warburg effect and tumorigenesis. Nat Commun. 2016;7:12431 10.1038/ncomms12431 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jin L, Chun J, Pan C, Alesi GN, Li D, Magliocca KR, et al. Phosphorylation-mediated activation of LDHA promotes cancer cell invasion and tumour metastasis. Oncogene. 2017;36(27):3797–806. 10.1038/onc.2017.6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhang J, Wang S, Jiang B, Huang L, Ji Z, Li X, et al. c-Src phosphorylation and activation of hexokinase promotes tumorigenesis and metastasis. Nat Commun. 2017;8:13732 10.1038/ncomms13732 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Koppenol WH, Bounds PL, Dang CV. Otto Warburg's contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011;11(5):325–37. Epub 2011/04/22. 10.1038/nrc3038 . [DOI] [PubMed] [Google Scholar]
  • 22.Cooper JA, Reiss NA, Schwartz RJ, Hunter T. Three glycolytic enzymes are phosphorylated at tyrosine in cells transformed by Rous sarcoma virus. Nature. 1983;302(5905):218–23. 10.1038/302218a0 [DOI] [PubMed] [Google Scholar]
  • 23.Flier JS, Mueckler MM, Usher P, Lodish HF. Elevated levels of glucose transport and transporter messenger RNA are induced by ras or src oncogenes. Science. 1987;235(4795):1492–5. 10.1126/science.3103217 . [DOI] [PubMed] [Google Scholar]
  • 24.Gonzalez L, Agullo-Ortuno MT, Garcia-Martinez JM, Calcabrini A, Gamallo C, Palacios J, et al. Role of c-Src in Human MCF7 Breast Cancer Cell Tumorigenesis. J Biol Chem. 2006;281(30):20851–64. 10.1074/jbc.M601570200 [DOI] [PubMed] [Google Scholar]
  • 25.Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, et al. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res. 2005;65(13):5506–11. 10.1158/0008-5472.CAN-05-0626 . [DOI] [PubMed] [Google Scholar]
  • 26.Mayoral-Varo V, Calcabrini A, Sanchez-Bailon MP, Martin-Perez J. miR205 inhibits stem cell renewal in SUM159PT breast cancer cells. PLoS One. 2017;12(11):e0188637 10.1371/journal.pone.0188637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Munoz-Guerra MF, Marazuela EG, Fernandez-Contreras ME, Gamallo C. P-cadherin expression reduced in squamous cell carcinoma of the oral cavity: an indicatior of poor prognosis. Cancer. 2005;103(5):960–9. 10.1002/cncr.20858 . [DOI] [PubMed] [Google Scholar]
  • 28.Perez-Riverol Y, Csordas A, Bai J, Bernal-Llinares M, Hewapathirana S, Kundu DJ, et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 2019;47(D1):D442–D50. Epub 2018/11/06. 10.1093/nar/gky1106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bergmeyer HU, Bergmeyer J, Grassl M. Methods of enzymatic analysis. Methods of enzymatic analysis / editor-in-chief, Hans Ulrich Bergmeyer; editors, Jürgen Bergmeyer and Marianne Grassl Edition: 3rd ed. 1983;III. NLM ID: 8508151 [Book].
  • 30.Echevarria L, Clemente P, Hernandez-Sierra R, Gallardo ME, Fernandez-Moreno MA, Garesse R. Glutamyl-tRNAGln amidotransferase is essential for mammalian mitochondrial translation in vivo. Biochem J. 2014;460(1):91–101. Epub 2014/03/04. 10.1042/BJ20131107 . [DOI] [PubMed] [Google Scholar]
  • 31.Adan C, Matsushima Y, Hernandez-Sierra R, Marco-Ferreres R, Fernandez-Moreno MA, Gonzalez-Vioque E, et al. Mitochondrial transcription factor B2 is essential for metabolic function in Drosophila melanogaster development. J Biol Chem. 2008;283(18):12333–42. Epub 2008/03/01. 10.1074/jbc.M801342200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cobb BS, Schaller MD, Leu TH, Parsons JT. Stable association of pp60src and pp59fyn with the focal adhesion-associated protein tyrosine kinase, pp125FAK. Mol Cell Biol. 1994;14(1):147–55. 10.1128/mcb.14.1.147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Schaller MD, Hildebrand JD, Shannon JD, Fox JW, Vines RR, Parsons JT. Autophosphorylation of the focal adhesion kinase, pp125FAK, directs SH2- dependent binding of pp60src. Mol Cell Biol. 1994;14(3):1680–8. 10.1128/mcb.14.3.1680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122(6):947–56. 10.1016/j.cell.2005.08.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell. 2007;1(5):555–67. Epub 2008/03/29. 10.1016/j.stem.2007.08.014 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Crabtree JS, Miele L. Breast Cancer Stem Cells. Biomedicines. 2018;6(3):77 Epub 2018/07/19. 10.3390/biomedicines6030077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Oh S, Kim H, Nam K, Shin I. Glut1 promotes cell proliferation, migration and invasion by regulating epidermal growth factor receptor and integrin signaling in triple-negative breast cancer cells. BMB Rep. 2017;50(3):132–7. 10.5483/bmbrep.2017.50.3.189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Garcia-Martinez JM, Calcabrini A, Gonzalez L, Martin-Forero E, Agullo-Ortuno MT, Simon V, et al. A non-catalytic function of the Src family tyrosine kinases controls prolactin-induced Jak2 signaling. Cell Signal. 2010;22(3):415–26. Epub 2009/11/07. S0898-6568(09)00330-1 [pii] 10.1016/j.cellsig.2009.10.013 . [DOI] [PubMed] [Google Scholar]
  • 39.Aleshin A, Finn RS. SRC: a century of science brought to the clinic. Neoplasia. 2010;12(8):599–607. 10.1593/neo.10328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Augoff K, Hryniewicz-Jankowska A, Tabola R. Lactate dehydrogenase 5: an old friend and a new hope in the war on cancer. Cancer Lett. 2015;358(1):1–7. Epub 2014/12/22. 10.1016/j.canlet.2014.12.035 . [DOI] [PubMed] [Google Scholar]
  • 41.Jia D, Lu M, Jung KH, Park JH, Yu L, Onuchic JN, et al. Elucidating cancer metabolic plasticity by coupling gene regulation with metabolic pathways. Proc Natl Acad Sci U S A. 2019;116(9):3909–18. Epub 2019/02/09. 10.1073/pnas.1816391116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hong CS, Graham NA, Gu W, Espindola Camacho C, Mah V, Maresh EL, et al. MCT1 Modulates Cancer Cell Pyruvate Export and Growth of Tumors that Co-express MCT1 and MCT4. Cell Rep. 2016;14(7):1590–601. Epub 2016/02/16. 10.1016/j.celrep.2016.01.057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Acosta JJ, Munoz RM, Gonzalez L, Subtil-Rodriguez A, Dominguez-Caceres MA, Garcia-Martinez JM, et al. Src mediates PRL-dependent proliferation of T47D and MCF7 cells via the activation of Fak/Erk1/2 and PI3K pathways. Mol Endocrinol. 2003;17(11):2268–82. 10.1210/me.2002-0422 . [DOI] [PubMed] [Google Scholar]
  • 44.Sanchez-Bailon MP, Calcabrini A, Gomez-Dominguez D, Morte B, Martin-Forero E, Gomez-Lopez G, et al. Src kinases catalytic activity regulates proliferation, migration and invasiveness of MDA-MB-231 breast cancer cells. Cell Signal. 2012;24(6):1276–86. Epub 2012/05/10. 10.1016/j.cellsig.2012.02.011 ; PubMed Central PMCID: PMCPMID: 22570868. [DOI] [PubMed] [Google Scholar]
  • 45.Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet. 2008;40(5):499–507. Epub 2008/04/30. ng.127 [pii] 10.1038/ng.127 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ling GQ, Chen DB, Wang BQ, Zhang LS. Expression of the pluripotency markers Oct3/4, Nanog and Sox2 in human breast cancer cell lines. Oncol Lett. 2012;4(6):1264–8. 10.3892/ol.2012.916 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zdralevic M, Vucetic M, Daher B, Marchiq I, Parks SK, Pouyssegur J. Disrupting the 'Warburg effect' re-routes cancer cells to OXPHOS offering a vulnerability point via 'ferroptosis'-induced cell death. Adv Biol Regul. 2018;68:55–63. Epub 2018/01/08. 10.1016/j.jbior.2017.12.002 . [DOI] [PubMed] [Google Scholar]
  • 48.McCleland ML, Adler AS, Shang Y, Hunsaker T, Truong T, Peterson D, et al. An integrated genomic screen identifies LDHB as an essential gene for triple-negative breast cancer. Cancer Res. 2012;72(22):5812–23. Epub 2012/11/10. 10.1158/0008-5472.CAN-12-1098 . [DOI] [PubMed] [Google Scholar]
  • 49.Lincet H, Icard P. How do glycolytic enzymes favour cancer cell proliferation by nonmetabolic functions? Oncogene. 2015;34(29):3751–9. 10.1038/onc.2014.320 . [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Antimo Migliaccio

1 Apr 2020

PONE-D-20-06055

c-Src functionality controls self-renewal and glucose metabolism in MCF7 breast cancer stem cells

PLOS ONE

Dear Dr. Martín-Pérez,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

As you could see from attached comment the Reviewers several points that need to be addressed basically concerning the Figures 2 and 3.

We would appreciate receiving your revised manuscript by May 16 2020 11:59PM. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Antimo Migliaccio, M.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. We note that you have stated that you will provide repository information for your data at acceptance. Should your manuscript be accepted for publication, we will hold it until you provide the relevant accession numbers or DOIs necessary to access your data. If you wish to make changes to your Data Availability statement, please describe these changes in your cover letter and we will update your Data Availability statement to reflect the information you provide.

3. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other r equirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels.

In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions.

4. Please include your tables as part of your main manuscript and remove the individual files. Please note that supplementary tables (should remain/ be uploaded) as separate "supporting information" files

Additional Editor Comments (if provided):

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: No

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: N/A

Reviewer #2: I Don't Know

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: No

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors have previously constructed a line of MCF7 human breast cancer cells (MCF7-Tet-On-SrcDN cells) in which expression of a dominant-negative form of the avian c-Src protein tyrosine kinase (SrcDN) can be induced by doxycycline (DOX) treatment, and shown that SrcDN expression inhibited cell attachment, spreading, and migration and also reduced tumorigenesis. Here, they have extended their studies of the phenotypic consequences of inducibly expressed SrcDN, and examined the effect SrcDN expression on the stem cell-like properties of a population of sorted CD44-high/CD24-low cells (BCSCs) MCF7-Tet-On-SrcDN cells that can form mammospheres in culture and initiate tumor growth in vivo. They found that induction of SrcDN expression in mammospheres increased the level of pY397 FAK, as reported previously, but that the level of E-cadherin was unaffected. They went on to show that mammosphere formation efficiency was reduced ~40% by the third passage of CD44-high/CD24-low cells when SrcDN was induced, suggesting that Src function is needed for BCSC self-renewal. To define the effect of SrcDN induction on BCSCs, immunoblotting for proteins of interest and a global proteomic analysis was carried on control and SrcDN expressing mammospheres. Decreases in cyclin D1, Nanog, Oct4 and ALDH1 were noted, consistent with reduced stemness. The global proteomic analysis revealed decreased levels of 15 proteins, with proteins involved in glycolysis being prominent. Next they tested whether SrcDN expression affected glycolysis, and found reduced glucose uptake and lactate excretion, paralleling a reduction in GLUT1, HK2 and LDHA levels, and decreased hexokinase, pyruvate kinase and lactate dehydrogenase enzymatic activity, all consistent with a reduced glycolytic rate. They also found that there were reduced levels of MCT1, the lactate transporter, explaining the observed rise in the level of intracellular lactate. Finally, they showed that cells dissociated from SrcDN-expressing mammospheres had altered maximum oxygen consumption rates in cells cultured in pyruvate-containing medium, albeit without a significant effect on ATP levels.

The observed effects of SrcDN expression on the stemness and metabolic properties of the CD44-high/CD24-low population of stem cell-like cells isolated from cultured MCF7-SrcDN cells are quite interesting, and suggest that that Src family kinase activity is needed to maintain the stemness state and glycolytic properties of BCSC cells. The data are largely convincing, and imply an important role for Src famliy kinase signaling in maintaining a stem cell-like population, although no mechanistic insights are presented into how Src kinase activity results in the observed changes in stem cell and metabolic genes.

Points: 1. Figure 1: The CD44-high/CD24-low population of BCSCs was isolated from MCF7-Tet-On-SrcDN cultures without induction of SrcDN, and then cells were plated to form mammospheres. However, it is not clear from the legend how long the mammospheres, once formed, were treated with DOX prior to the immunoblotting analysis or what happened to the acinar structures during the DOX treatment period, e.g., do they maintain their morphology. It is also not clear what happens to the phenotype of BCSC cells once a mammosphere has been formed - do all the cells retain their BCSC phenotype or have some of them differentiated back into CD44-high/CD24-high cells. The authors report that when mammospheres were dissociated into single cells and then re-plated they could form mammospheres, but information about the efficiency of this process was not provided. If the dissociated cells are re-analyzed by FACS, what fraction are CD44-high/CD24-low and what fraction are CD44-high/CD24-high. If the cells are treated with DOX either prior to (i.e. as mammospheres) or during the re-plating process with DOX, how does this affect the efficiency of mammosphere formation (see point 2)?

2. Figure 2A: It is not clear from the legend or text exactly what was done in this experiment. Were the single cells that were plated obtained from mammospheres that had been pretreated with DOX, or were the cells continuously treated with DOX during plating and mammosphere outgrowth? This needs to be spelled out in the legend.

3. Figure 2B: How long was SrcDN induced for in the mammospheres prior to harvesting for the proteomic analyses?

4. Table 1: It is unclear from the text and the supplementary materials how many total proteins were identified through the MS analysis, i.e. the 15 protein decreases and 4 protein increases need to have a denominator of the total number of proteins identified. By comparative immunoblotting of control and DOX-induced MCF7-SrcDN mammospheres, the authors showed that the levels of several proteins involved in stem cell function and the glycolytic pathway were decreased upon SrcDN induction. However, it is not clear how many of these proteins were also found to be downregulated in their proteomic dataset - none of the proteins tested by immunoblotting is listed in Table S1, but I did not check the data deposited in the ProteomeXchange.

5. Figure 2: Have the authors carried out a parallel RNA-seq analysis on control and DOX-induced MCF7-SrcDN cells to determine which of the observed changes in protein levels can be attributed to decreased levels of the cognate mRNAs (such an analysis does not seem to be reported in their prior studies on MCF7-Tet-On-SrcDN cells). It would be particularly interesting to know whether SrcDN expression affects the levels of Myc and HIF1 RNA expression, since these transcription factors are known to promote expression of the HK2 and LDHA genes. Also, are there any changes in the levels of expression of RNAs encoding mitochondrial ETC genes? Likewise, analysis of the differences in RNA expression patterns between CD44-high/CD24-low BCSCs and the CD44-high/CD24-high cells would be informative.

6. Figure 3: Although an ~50% decrease in pY10 LDHA level upon DOX induction of SrcDN is reported in panel D, there is a concern about how accurate this value is - the low level of the pY10 immunoblot signal means that the percent decrease will be very dependent on how accurately the background signal, which needs to be subtracted, was estimated. A bigger decrease than the observed 50% in pY10 LDHA level might have been expected given the very high level of SrcDN expression compared to that of endogenous c-Src in the induced cells. In this regard, it has been reported (ref. 18) that the level of pY10 in MCF7 cells is very low compared to that in other more advanced breast cancer lines.

Minor points: 1. The first paragraph of the results is entitled “SrcDN reduced self-renewal of MCF7-BCSCs” but no such data are described in this section.

2. While many readers may know what MCF7 cells are, the authors should describe them in the Introduction, i.e. they are ER/PR-positive, HER2-negative, luminal A-type, non-metastatic human breast cancer cells.

3. Do the cellular phenotypes of DOX treated BCSCs revert when DOX is withdrawn, and if so how rapidly?

4. Page 12, second paragraph: The authors say that the endogenous c-Src cannot be distinguished from the exogenously expressed SrcDN by proteomic analysis, but this is not actually true, since they expressed chicken c-SrcDN in human cells, and there are a significant number of sequence differences between the two c-Src’s, which would mean that the chicken and human c-Src proteins would generate unique peptides that could be quantified.

5. Although technically more challenging because of the small cell numbers, it would have been interesting to analyze the sorted CD44-high/CD24-low BCSC MCF7 cell population directly for changes in protein levels, glycolysis rates, etc, compared to the CD44-high/CD24-high population.

Reviewer #2: In this study, the authors tried to investigate whether SrcDN reduces tumorigenesis of MCF7 cells via inhibiting breast cancer stem cells. They isolated BCSCs and non-BCSCs based on ESA, CD44, and CD24 markers and evaluated the effect of SrcDN on the metabolic changes. Though somewhat interesting, the data are underdeveloped and not supporting the conclusion. The major flaw is that they didn’t show that the changes in glycolysis and decrease in self-renewal were unique to the BCSCs. Except isolating these different cell populations and analysis of the E-Cad levels, there are no experiments done comparing these two groups. Without such analysis, the conclusion of the SrcDN on BSCS to inhibit tumorigenesis is not supported. There were several syntax/grammatical errors throughout the manuscript, which should be improved.

In addition, they should have performed direct analysis to address whether the SrcDN induced glycolysis changes are directly connected to self-renewal properties, since they showed it wasn’t related to ATP production or ROS.

Specific comments:

Figure 1:

1. Figure 1A, isotype control for setting up the gate for the FACS should be included.

2. Figure 1B, it is unclear the significance of this figure, since there is no comparison of spheres formed by CD24- and CD24+ groups. There is lack of scale bars.

3. Other methods have been used to isolate putative BCSCs, particularly using ALDH activity for the luminal breast cancers. To strengthen the claim on BCSCs, the authors should also consider using different ways to evaluate BCSCs.

Figure 2:

1. Figure 2A, possibly due to sample size or heterogeneity in the mammospheres, without seeing the full error bars and with the statistical significance being relatively large, this data is not very strong. It is also unclear what the bar and the error bar show. mean or median? SD or SEM?

2. In Figure 2B, the quantitation for the Nanog and Oct3/4 Western blots were the same, but the blot for Oct3/4 was far less apparent compared to Nanog, especially if there were compared to the same β-Actin loading control.

Figure 3:

1. A-C. As in Figure 2, the lack of full error bars and small sample size makes the data less convincing, especially because each graph has the same inferred statistical significance from very different measurements. Again, it is unclear what the error bars represent.

2. In Figure 3D, The blots for Glut-1 and pY10-LDHA, and LDHA are not as well defined as the others, although it could be due to the antibodies. The quantitative decreases in Glut-1, HK2, and pY10-LDHA/LDHA are not very significant and it is unclear whether such level of a decrease can cause changes in glycolytic activity/output. There are no functional validation or rescue experiments done to link the protein level changes to the glycolysis changes.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 Jul 16;15(7):e0235850. doi: 10.1371/journal.pone.0235850.r002

Author response to Decision Letter 0


9 Jun 2020

We have elaborated a large document that we named "Reviewers_Reply_03-06-2020" in PDF format where we have answered the questions raised by the Reviewers and the Editor. We must say that we are thankful to the comments raised, as we think that they help us to improve the quality of the manuscript.

Attachment

Submitted filename: Reviewers_Repply_03-06-2020.pdf

Decision Letter 1

Antimo Migliaccio

24 Jun 2020

c-Src functionality controls self-renewal and glucose metabolism in MCF7 breast cancer stem cells

PONE-D-20-06055R1

Dear Dr. Martín-Pérez,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Antimo Migliaccio, M.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: (No Response)

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors have satisfactorily addressed most of my comments, and have added some new data as supplementary figures, including Figure S3, where they showed that DOX treatment of MCF7-Tet-On-SrcDN cell-derived mammospheres to induce DN-cSrc expression resulted in a strong decrease in the numbers of CD44-high/CD24-low stem cell-like cells, which have high tumorigenicity, and a corresponding increase in the number of CD44-high/CD24-high cells, which show reduced tumorigenicity, consistent with the observed DOX-induced decrease in stem cell factors such as Nanog.

Reviewer #2: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: Tony Hunter

Reviewer #2: No

Acceptance letter

Antimo Migliaccio

30 Jun 2020

PONE-D-20-06055R1

c-Src functionality controls self-renewal and glucose metabolism in MCF7 breast cancer stem cells

Dear Dr. Martín-Pérez:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Antimo Migliaccio

Academic Editor

PLOS ONE

Associated Data

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

    Supplementary Materials

    S1 File

    (PDF)

    Attachment

    Submitted filename: Reviewers_Repply_03-06-2020.pdf

    Data Availability Statement

    Data have been uploaded to Proteome Xchange database (Project accession number: PXD017789, project DOI: 10.6019/PXD017789).


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES