Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Research

Metformin counteracts stimulatory effects induced by insulin in primary breast cancer cells

verfasst von: Domenica Scordamaglia, Francesca Cirillo, Marianna Talia, Maria Francesca Santolla, Damiano Cosimo Rigiracciolo, Lucia Muglia, Azzurra Zicarelli, Salvatore De Rosis, Francesca Giordano, Anna Maria Miglietta, Ernestina Marianna De Francesco, Veronica Vella, Antonino Belfiore, Rosamaria Lappano, Marcello Maggiolini

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

Abstract

Background

Metabolic disorders are associated with increased incidence, aggressive phenotype and poor outcome of breast cancer (BC) patients. For instance, hyperinsulinemia is an independent risk factor for BC and the insulin/insulin receptor (IR) axis is involved in BC growth and metastasis. Of note, the anti-diabetic metformin may be considered in comprehensive therapeutic approaches in BC on the basis of its antiproliferative effects obtained in diverse pre-clinical and clinical studies.

Methods

Bioinformatics analysis were performed using the information provided by The Invasive Breast Cancer Cohort of The Cancer Genome Atlas (TCGA) project. The naturally immortalized BC cell line, named BCAHC-1, as well as cancer-associated fibroblasts (CAFs) derived from BC patients were used as model systems. In order to identify further mechanisms that characterize the anticancer action of metformin in BC, we performed gene expression and promoter studies as well as western blotting experiments. Moreover, cell cycle analysis, colony and spheroid formation, actin cytoskeleton reorganization, cell migration and matrigel drops evasion assays were carried out to provide novel insights on the anticancer properties of metformin.

Results

We first assessed that elevated expression and activation of IR correlate with a worse prognostic outcome in estrogen receptor (ER)-positive BC. Thereafter, we established that metformin inhibits the insulin/IR-mediated activation of transduction pathways, gene changes and proliferative responses in BCAHC-1 cells. Then, we found that metformin interferes with the insulin-induced expression of the metastatic gene CXC chemokine receptor 4 (CXCR4), which we found to be associated with poor disease-free survival in BC patients exhibiting high levels of IR. Next, we ascertained that metformin prevents a motile phenotype of BCAHC-1 cells triggered by the paracrine liaison between tumor cells and CAFs upon insulin activated CXCL12/CXCR4 axis.

Conclusions

Our findings provide novel mechanistic insights regarding the anti-proliferative and anti-migratory effects of metformin in both BC cells and important components of the tumor microenvironment like CAFs. Further investigations are warranted to corroborate the anticancer action of metformin on the tumor mass toward the assessment of more comprehensive strategies halting BC progression, in particular in patients exhibiting metabolic disorders and altered insulin/IR functions.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-022-03463-y.
Domenica Scordamaglia, Francesca Cirillo and Marianna Talia contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACTB
Actin beta
AMPK
Adenosine monophosphate activated protein kinase
AKT
Protein kinase B
BC
Breast cancer
BCA
Bicinchoninic acid
CAFs
Cancer-associated fibroblasts
CM
Conditioned medium
CXCL12
C-X-C Motif Chemokine Ligand 12
CXCR4
CXC chemokine receptor 4
DAPI
4′,6-Diamidino-2-phenylindole
DFI
Disease-free interval
DMEM/F-12
Dulbecco’s Modified Eagle Medium/Nutrient Mixture F-12
DN/c-Fos
Dominant negative c-Fos
EMT
Epithelial-to-mesenchymal transition
ER
Estrogen receptor
ERα46
46 kDa ERα splice variant
ERK1/2
Extracellular signal-regulated kinases 1 and 2
FAPα
Fibroblast activation protein alpha
FBS
Fetal bovine serum
FLNA
Filamin-A
GFR
Growth Factor Reduced
IGF-1R
Insulin-like growth factor 1 receptor
IGFs
Insulin-like growth factors
IR
Insulin receptor
IR-A
IR isoform A
IR-B
IR isoform B
ISR1
Insulin receptor substrate 1
MAPK
Mitogen-activated protein kinase
MEK
MAPK/ERK kinase
mTOR
Mechanistic target of rapamycin
ORR
Objective response rate
OS
Overall survival
PBS
Phosphate-buffered saline
PCR
Polymerase chain reaction
PI
Propidium iodide
PI3K
Phosphoinositide 3-kinase
pRL-TK
HSV-thymidine kinase promoter
RSEM
RNA-Seq by expectation–maximization
SDS
Sodium dodecyl sulfate
T2D
Type 2 diabetes
TCGA
The Invasive Breast Cancer Cohort of the Cancer Genome Atlas

Background

Breast cancer (BC) represents the most diagnosed malignancy and leading cause of cancer death in women worldwide [1]. Despite many improvements gained in early detection and treatment of BC, the advanced disease still remains a main challenge [25]. Metabolic traits that are usually linked with type 2 diabetes (T2D), including hyperinsulinemia, dysglycemia, dyslipidemia, inflammation, oxidative stress and obesity, are well-known risk factors for BC and major contributors to its progression and metastatic dissemination [6, 7]. For instance, high plasma levels of insulin are associated with increased BC incidence and relapse, resistance to conventional and targeted therapies as well as poor outcome even in the absence of diabetes [8, 9]. Insulin regulates multiple signaling pathways implicated in the growth and metastatic features of BC by binding to and activating either the cognate receptor (IR) or the insulin-like growth factor 1 receptor (IGF-1R) [1013]. In this regard, it should be mentioned that approximately 80% of BCs express high levels of IR and its autophosphorylation is associated with high BC mortality [1416]. In particular, the isoform A of IR (IR-A) is frequently overexpressed in BC, where it regulates epithelial-to-mesenchymal transition (EMT), stem-like cell phenotype, cell invasion, metastasis and resistance to therapies [10, 17, 18]. Epidemiological and retrospective clinical data have demonstrated that a first-line drug for T2D, namely metformin, is associated with a low risk of BC and reduced cancer-related mortality rate in T2D patients [1923]. Accordingly, extensive preclinical studies have indicated that metformin may exert antitumor effects in BC cells through both direct (insulin-independent) and indirect (insulin-dependent) mechanisms [2426]. One of the well-accepted insulin-independent effects of metformin involves the activation of adenosine monophosphate activated protein kinase (AMPK), which in turn downregulates main insulin-stimulated transduction pathways, such as IRS1 and PI3K/AKT/mTOR transduction signaling [25]. Furthermore, it has been reported that IR expression status, which is frequently related to insulin levels, may represent a predictive factor of the antitumoral activity of metformin [23, 26].
Here, we provide evidence regarding novel mechanisms by which metformin may elicit anti-proliferative, anti-migratory and anti-invasive effects in a naturally immortalized BC cell line named BCAHC‐1 [25]. In particular, we show that the anti-cancer action triggered by metformin relies on its ability to inhibit the insulin/IR-mediated transduction pathway as well as the insulin-generated feedforward loop that couples CXCL12 induction by CAFs to CXCR4 expression by BCAHC‐1 cells.

Methods

Reagents

Insulin and Metformin were purchased from Merck Life Science (Milan, Italy). The MEK inhibitor trametinib, the insulin receptor inhibitor OSI-906 and the PI3Kα inhibitor alpelisib were obtained from MedChemExpress (DBA, Milan, Italy). The CXCR4 antagonist AMD3100 was purchased from Santa Cruz Biotechnology (DBA, Milan, Italy). All compounds were dissolved in DMSO, except insulin, metformin and AMD3100 that were solubilized in water.

Cell cultures

BCAHC-1 cells were isolated and characterized as previously described [27], and maintained in DMEM/F-12 with phenol red, supplemented with 5% FBS and 100 μg/ml penicillin/streptomycin. Cells were grown at 37 °C in a humidified 5% CO2 and switched to a medium without serum and phenol red the day before treatments to be processed for experiments. CAFs were obtained as previously described [27, 28] from 10 invasive ductal breast carcinomas and pooled for the subsequent studies. Briefly, specimens were cut into 1–2 mm diameter pieces, placed in a digestion solution comprising 400 IU collagenase, 100 IU hyaluronidase, 10% serum, antibiotics, and antimycotics, and incubated overnight at 37 °C. After centrifugation at 90×g for 2 min, the supernatant containing fibroblasts was centrifuged at 485×g for 8 min; the pellet obtained was suspended in Medium 199 and Ham’s F12 mixed 1:1 (supplemented with 10% FBS and 100 μg/ml penicillin/streptomycin). CAFs were then expanded into 10-cm Petri dishes and stored as cells passaged for three population doublings within a total of 7 to tissue dissociation. Primary cell cultures of fibroblasts were characterized by immunofluorescence with human anti-vimentin (V9) and human anti-cytokeratin 14 (LL001) (Santa Cruz Biotechnology, DBA, Milan, Italy) (data not shown). FAPα antibody (H-56, Santa Cruz Biotechnology, DBA, Milan, Italy) was used to characterize activated fibroblasts (data not shown). We used CAFs passaged for up to 10 population doublings for the experiments to minimize clonal selection and culture stress, which could occur during extended tissue culture.

Gene expression studies and PCR arrays

Total RNA was extracted, and cDNA was synthesized by reverse transcription as previously described [29]. The expression of selected genes was quantified by real-time PCR using platform Quant Studio7 Flex Real-Time PCR System (Thermo Fisher Scientific, Milan, Italy). Gene-specific primers were designed using Primer Express version 2.0 software (Applied Biosystems) and are as follows: 5′-CGAGCCCTTTGATGACTTCCT-3′ (c-Fos forward) and 5′-GGAGCGGGCTGTCTCAGA-3′ (c-Fos reverse); 5′-AGCTGTGCATCTACACCGAC-3′ (cyclin D1 forward) and 5′-GAAATCGTGCGGGGTCATTG-3′ (cyclin D1 reverse); 5′-AAGCCACCCCACTTCTCTCTAA-3′ (ACTB forward) and 5′-CACCTCCCCTGTGTGGACTT-3′ (ACTB reverse); 5′-CCTTGGAGCCAAATTTAAAACCT-3′ (CXCR4 forward) and 5′-GCTGGACCCTCTGCTCACA-3′ (CXCR4 reverse); 5′-CAGATGCCCATGCCGATTCT-3′ (CXCL12 forward) and 5′-TTCTTCAGCCGGGCTACAAT-3′ (CXCL12 reverse). Assays were performed in triplicate and the results were normalized for actin beta (ACTB) expression and then calculated as fold induction of RNA expression.
PCR arrays were performed using a TaqMan™ Human Tumor Metastasis Array (Thermo Fisher Scientific, Milan, Italy) according to the manufacturer’s instructions. The amplification reaction and the results analysis were carried out using platform Quant Studio7 Flex Real-Time PCR System (Thermo Fisher Scientific, Milan, Italy).

Reporter gene assays

For reporter gene assays, cells (1 × 105) were plated into 24-well dishes with 500 mL/well culture medium containing 5% FBS and transfected for 18 h using X-treme GENE 9 DNA Transfection Reagent, as recommended by the manufacture (Merck Life Science, Milan, Italy). A mixture containing 0.5 mg of gene reporter (Fos-luc, kindly obtained from Dr. K. Nose, Department of Microbiology, Showa University School of Pharmaceutical Sciences, Hatanodai, Shinagawa-ku, Tokyo, Japan; Cyclin D1-luc, kindly obtained from Dr. R. G. Pestell, Kimmel Cancer Center, Department of Cancer Biology and Jefferson Stem Cell and Regenerative Medicine Center, Thomas Jefferson University, Philadelphia, PA), and 5 ng of pRL-TK was then transfected, after 18 h cells were treated with insulin in the presence or absence of metformin, OSI-906, trametinib and alpelisib for additional 12 h. Luciferase activity was measured using the Dual Luciferase Kit (Promega, Milan, Italy) according to the manufacturer’s recommendations. Firefly luciferase activity was normalized to the internal transfection control provided by the Renilla luciferase activity. Normalized relative light unit values obtained from cells treated with vehicle (−) were set as onefold induction, upon which the activity induced by treatments was calculated. For gene silencing experiments, cells were plated onto 10-cm dishes and transfected by X-treme GENE 9 DNA Transfection Reagent for 24 h before treatments with a control vector and the plasmid DN/c-Fos (kindly obtained from Dr. C. Vinson, NIH, Bethesda, USA) encoding a c-Fos mutant that heterodimerizes with c-Fos dimerization partners but not allowing DNA binding.

Western blot analysis

Cells were grown in 10-cm dishes, exposed to treatments and then lysed in 500 μl RIPA buffer with protease inhibitors (1.7 mg/ml aprotinin, 1 mg/ml leupeptin, 200 mmol/l phenylmethylsulfonyl fluoride, 200 mmol/l sodium orthovanadate and 100 mmol/l sodium fluoride). Samples were then centrifuged at 13,000 rpm for 10 min and protein concentrations were determined using BCA protein assay according to the manufacturer’s instructions (Thermo Fisher Scientific, Milan, Italy). Equal amounts of whole-protein extract were resolved on 8–10% SDS polyacrylamide gels and transferred to a nitrocellulose membrane (Merck Life Science, Milan, Italy), which were probed with primary antibodies against: phosphorylated ERK1/2 (E4), ERK2 (C-14), AKT, c-Fos (E-8), CXCR4 (4G10), Insulin Rα (N-20) and β-actin (AC-15) (Santa Cruz Biotechnology, DBA, Milan, Italy), cyclin D1 (TA801655) (OriGene Technologies, DBA, Milan, Italy), p-IR (Y1146), p-AKT (D9E) and CXCL12 (3740) (Cell Signaling Technology, Euroclone, Milan, Italy). Proteins were detected by horseradish peroxidase-linked secondary antibodies (Bio-Rad, Milan, Italy) and then revealed using the chemiluminescent substrate for western blotting Clarity Western ECL Substrate (Bio-Rad, Milan, Italy).

Acetone precipitation of proteins

Protein precipitation from conditioned medium derived from CAFs was carried out using the precipitation method with acetone [30]. Briefly, four volumes of ice-cold acetone (Merck Life Science, Milan, Italy) were added to one volume of sample. The mixture was vortexed and incubated at − 20 °C overnight. This was followed by centrifugation at 10,000×g for 15 min at 4 °C. Afterwards, the supernatant was discarded, the pellet was air dried, then it was dissolved in 2× Laemmli buffer and used in the appropriate experiments. In western blot analysis, the protein loading of conditioned medium samples was checked by Ponceau red staining [0.1% Ponceau S (w/v) in 5% acetic acid] of the blotted membranes.

Immunofluorescence microscopy

Cells were grown on a cover slip, exposed to treatments and then fixed in 4% paraformaldehyde in PBS, permeabilized with 0.2% Triton X-100, washed 3 times with PBS and incubated at 4 °C overnight with primary antibodies anti-CXCL12 (3740) (Cell Signaling Technology, Euroclone, Milan, Italy). After incubation, the slides were extensively washed with PBS, probed with Alexa Fluor conjugated secondary antibodies (Thermo Fisher Scientific, Milan, Italy) for 1 h at room temperature. Finally, cells were washed three times with PBS, incubated with DAPI (4′,6-diamidino-2-phenylindole) (1:1000) for 3 min and, after washing, immunofluorescence images were obtained using the Cytation 3 Cell Imaging Multimode reader (BioTek, AHSI, Milan Italy) and analyzed by the Gen5 software (BioTek, AHSI, Milan Italy).

Phalloidin staining

Cells were exposed to treatments, washed twice with PBS, fixed in 4% paraformaldehyde in PBS for 10 min, washed briefly with PBS and then incubated with Phalloidin-Fluorescent Conjugate (Santa Cruz Biotechnology, DBA, Milan, Italy). The images were obtained using the Cytation 3 Cell Imaging Multimode reader (BioTek, AHSI, Milan Italy) and analyzed by the Gen5 software (BioTek, AHSI, Milan Italy).

Cell cycle analysis

To analyze cell cycle distribution, BCAHC-1 cells (1 × 105) were cultured in regular medium in 6 well plates and shifted in medium without serum. Next, BCAHC-1 cells were exposed to treatments, then pelleted, once washed with PBS and fixed in 50% methanol overnight at − 20 °C, before to stain with a solution containing 50 µg/ml propidium iodide (PI) in 1×PBS, 20 U/ml RNAse-A and 0.1% Triton (Merck Life Science, Milan, Italy). Cell phases were estimated as a percentage of a total of 10 000 events. Samples were then analyzed with CytoFLEX flow cytometry (Beckman-Coulter, Milan, Italy).

Proliferation assay

BCAHC-1 cells (1 × 104) were seeded in 24-well plates in regular growth medium, washed once they had attached, incubated in medium containing 2.5% charcoal-stripped FBS and then exposed to treatments. Treatments were renewed every day. The proliferation rate was calculated counting the cells on day 5 using the Countess Automated Cell Counter, as recommended by the manufacturer’s protocol (Thermo Fisher Scientific, Milan, Italy).

Colony formation assay

BCAHC-1 cells were cultured in regular growth medium to 90% confluence. Cells were then trypsinized, counted and seeded (1 × 103) in 6-well plates in medium containing 2.5% charcoal-stripped FBS and then exposed to treatments, as indicated. Treatments were renewed every 3 days. After 10 days, cells were washed with PBS, fixed in acetone:methanol (1:1) for 3 min at room temperature and then stained with Giemsa (Merck Life Science, Milan, Italy) for 10 min. A total of 10 pictures for each condition was detected using a digital camera and colony number was measured by ImageJ program.

Spheroid formation assay

For spheroid generation, 100 μl/well of BCAHC-1 cell suspensions (1 × 104) were placed into 2% agar coated 24-well plates in medium containing 2.5% charcoal stripped FBS. Three days after seeding, tumor spheroids (a single spheroid per well) were exposed to treatments and a 50% medium and treatment renewal was performed every 2 days. Images were obtained on day 20 using a conventional inverted microscope, thereafter cell number per spheroid was determined by trypsinizing three different spheroids, mixing the cell suspension with trypan blue and counting the number of viable cells. The total number of cells obtained was divided by the number of trypsinized spheroids.

Conditioned medium

CAFs were placed in medium without serum and treated with vehicle (−) or 10 nM insulin for 4 h. Thereafter, CAFs were washed and fresh medium was added without serum for additional 8 h. The supernatants were then collected, centrifuged at 3500 rpm for 5 min to remove cell debris and used as conditioned medium in the appropriate experiments.

Migration assay

Transwell 8 μm polycarbonate membranes (Costar, Merck Life Science, Milan, Italy) were used to evaluate in vitro cell migration. In 300 μl serum free medium, BCAHC-1 cells (5 × 104) previously exposed to treatments were seeded in the upper chamber. Conditioned medium of CAFs was added to the bottom chambers. 8 h after seeding, cells on the upper surface of the membrane were then removed by wiping with Q-tip, and migrated cells were fixed with 100% methanol, stained with Giemsa (Merck Life Science, Milan, Italy), photographed using a digital camera and counted using the WCIF ImageJ software.

Matrigel evasion assay

The Matrigel drop assay was performed as previously described [31, 32] and modified by suspending 3 × 104/drop of BCAHC-1 cells, previously exposed to treatments, in 15 μl of complete medium and gently mixed with 15 μl of Corning® Matrigel® Growth Factor Reduced (GFR) Basement Membrane Matrix (Biogenerica, Catania, Italy). The cell/matrigel suspension was layered onto the surface of 12-well plate to form a well-defined drop and placed at 37 °C to solidify. 2 ml of conditioned media collected from CAFs previously treated with vehicle (−) or 10 nM insulin were placed over the drop. Cells were observed at specified time points and drops were photographed using the ImageFocus Plus V2 software of the inverted Oxion Inverso microscope (Euromex Microscopen bv, The Netherlands). The number of cells migrated out of the drop was measured.

TCGA database and data collection

Bioinformatics studies were performed using the Invasive Breast Cancer Cohort data of the publicly available dataset The Cancer Genome Atlas (TCGA). The patient clinical information, gene expression data (RNA Seq V2 RSEM) and the pathway activity (z-scores) were retrieved from UCSC Xena (https://​xenabrowser.​net/​) on the 7th March 2022. Samples (n. 1247) were filtered for missing values.

Survival analysis

The survival analyses were performed using the IR-mediated signaling pathway activity and the IR and CXCR4 gene expression data of the TCGA patients along with the overall survival (OS), and disease-free interval (DFI) information. The survivALL package was employed to examine Cox proportional hazards for all possible points of separation (low–high cut-points). The cut-point with the lowest p-value was selected [33], therefore dividing the patients with high (n = 488) and low (n = 87) IR-mediated signaling pathway activity, high (n = 166) and low (n = 90) IR expression or high (n = 145) and low (n = 107) CXCR4 expression. The Kaplan Meier survival curves were generated using the R survival and survminer packages.

Statistical analysis

The statistical analysis was performed using ANOVA followed by NewmanKeuls’ test to determine differences in means. Densitometric analysis was performed using the freeware software ImageJ that allowed to quantify the band intensity of the protein of interest with respect to the band intensity of the loading control. All bioinformatics analyses were carried out using R Studio. Box plots were performed with the tidyverse package and the related statistical analysis was carried out by using the t-test. Heatmaps were drawn on the log2 fold changes of gene expression using the pheatmap package. p‐values < 0.05 were considered statistically significant.

Results

Metformin prevents the growth effects triggered by insulin/IR signaling in BCAHC-1 cells

Previous studies have shown that a crosstalk between estrogen and insulin transduction pathways promotes the progression of BC [27, 34, 35]. Therefore, we began our study analyzing the expression and activity of IR in the TCGA cohort of ER-positive and negative BC patients. First, we ascertained that the IR mRNA expression as well as the IR-mediated signaling pathway are significantly higher in ER-positive than ER-negative BCs (Fig. 1A, B). In addition, stratifying the BC patients on the basis of the PAM50 gene signature, both IR expression and signaling were found higher in the BC luminal A and luminal B patients respect to the other BC subtypes (Additional file 1: Fig. S1A, B). In accordance with these data, was found enriched and associated with a worse overall survival (OS) (Fig. 1C) in ER-positive respect to ER-negative BC patients. Pre‑clinical and currently ongoing clinical trials indicate that the insulin-sensitizer metformin elicits anti-tumor effects in ER-positive BCs and is associated with a lower risk of developing BC in patients with T2D respect to control patients [11, 20, 3640]. Hence, we aimed to ascertain whether metformin may inhibit insulin-dependent signaling pathways like phosphoinositide 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK). To this end, we used as a model system a BC cell line namely BCAHC-1, which is characterized by the peculiar expression of IR and the 46 kDa ERα splice variant (ERα46) [27, 41, 42]. First, the insulin-induced IR phosphorylation was prevented using the IR inhibitor OSI-906, but not in the presence of the PI3K inhibitor alpelisib and the MEK inhibitor trametinib (Fig. 1D). The activation of AKT upon insulin exposure was totally abrogated by OSI-906 and alpelisib, but only partially dampened by trametinib (Fig. 1E) as previously reported [43], whereas the insulin-promoted activation of the ERK1/2 pathway was inhibited by OSI-906 and trametinib but not using alpelisib (Fig. 1F). Next, we assessed that metformin reduces the IR phosphorylation prompted by insulin, without any change in total IR protein expression (Fig. 1G). Moreover, we determined that metformin prevents the activation of both AKT and ERK1/2 upon insulin exposure in BCAHC-1 cells (Fig. 1H, I). On the basis of these findings and in line with previous studies showing that PI3K/AKT and ERK1/2 transduction pathways mediate the expression of insulin target genes, such as c‐Fos and Cyclin D1 [27, 4446], we found that insulin induces c-Fos expression in BCAHC-1 cells at both mRNA (data not shown) and protein levels (Fig. 2A–C). Worthy, OSI-906, alpelisib, trametinib and metformin abrogated either the protein increase (Fig. 2A–C) or the activation of a promoter construct of c-Fos transiently transfected in BCAHC-1 cells (Fig. 2B–D). Similarly, we established that OSI-906, alpelisib and trametinib prevent the Cyclin D1 protein increase and the transactivation of a Cyclin D1 promoter construct upon insulin exposure (Fig. 2E, F). Reminiscing our previous findings showing that c-Fos is involved in the regulation of Cyclin D1 [27], we demonstrated that the protein induction of Cyclin D1 by insulin is prevented transfecting BCAHC-1 cells with a DN/c-Fos expression vector (Fig. 2G). Furthermore, we showed that metformin abolishes the Cyclin D1 protein increase (Fig. 2H) and the activation of a Cyclin D1 promoter construct (Fig. 2I) triggered by insulin. Taken together, these findings indicate that in BCAHC-1 cells metformin inhibits the insulin-induced IR/PI3K/MAPK/c-Fos transduction pathway, thus preventing the up-regulation of Cyclin D1. In line with the acknowledged role of Cyclin D1 as a regulator of G1/S transition in the cell cycle [47], we found that metformin leads to the arrest within the G0/G1 phase of the cell cycle in BCAHC-1 cells (Fig. 3A, B). Interestingly, the results of flow cytometric analysis of propidium iodide (PI)-staining showed that insulin induces S-phase entry in BCAHC-1 cells, however this effect is no longer evident in the presence of metformin (Fig. 3A, B) and using OSI-906, alpelisib or trametinib (data not shown). In accordance with these observations, the proliferation and colony-forming ability of BCAHC-1 cells upon insulin treatment were abolished by metformin (Fig. 3C–E) as well as in the presence of OSI-906, alpelisib, trametinib (Additional file 2: Fig. S2A–C). In addition, the spheroid expansion of BCAHC-1 cells prompted by insulin was inhibited by metformin (Fig. 3F, G) as well as OSI-906, alpelisib and trametinib (Additional file 2: Fig. S2D, E). Overall, these data indicate that metformin impairs the growth responses induced by insulin in BCAHC-1 cells.

Metformin inhibits the up-regulation of the pro-tumorigenic chemokine receptor CXCR4 triggered by insulin in BCAHC-1 cells

Considering that a negative correlation between metformin and the metastatic spread of BC has been reported [4850], we then assessed whether metformin may abolish the capability of insulin to induce a metastatic gene signature in BCAHC-1 cells. In this vein, we performed a TaqMan Gene Expression Assay consisting of a Human Tumor Metastasis Array in BCAHC-1 cells exposed to insulin in the presence or absence of metformin. Among the genes that showed at least a 0.25 log2 fold change upon insulin treatment respect to vehicle and the reduction of this increase in the presence of metformin, we focused on the C-X-C Motif Chemokine Receptor 4 (CXCR4) due to its acknowledged role as prognostic marker and main driver of BC metastasis [5153]. Evaluating the expression of CXCR4 in ER-positive BC samples of the TCGA dataset, we assessed by pairwise comparison that CXCR4 levels are significantly higher in ER-positive BCs respect to matched normal tissues (Fig. 4A). Thereafter, we ascertained whether the mRNA expression of both CXCR4 and IR would be predictive of the outcome of ER-positive BC patients. In this regard, survival analyses revealed that high IR levels are associated with a short disease-free interval (DFI) in BC patients showing increased CXCR4 expression (Fig. 4B). Similarly, a correlation between high CXCR4 levels and poor prognosis characterizes BCs displaying enhanced IR levels (Fig. 4C). Considering that insulin modulates pro-metastatic mediators involved in the regulation of CXCR4 in BC cells [54, 55], we ascertained that insulin increases both mRNA (Fig. 4D) and protein levels (Fig. 4E) of CXCR4 in BCAHC-1 cells. Next, immunoblot experiments showed that either the IR inhibitor OSI-906 (Fig. 4F) or metformin (Fig. 4G) prevent the up-regulation of CXCR4 triggered by insulin, suggesting that metformin prevents the expression of CXCR4 mediated by insulin/IR axis in BCAHC-1 cells.

Insulin induces the secretion of the CXCR4 ligand CXCL12 in cancer-associated fibroblasts (CAFs)

A crosstalk between tumor cells and cancer‐associated fibroblasts (CAFs) may fuel the growth and metastasis of BC [5659]. In particular, the CAF-secreted CXCR4 ligand, namely CXCL12 (also known as SDF-1), is an important factor linking stromal and BC cells toward aggressive malignant features [5964]. On the basis of the aforementioned findings and considering that insulin has been implicated in the release of pro-tumorigenic and inflammatory mediators by stromal cells [65, 66], we ascertained whether insulin may promote the expression and secretion of CXCL12 in breast CAFs. By real-time PCR, immunoblotting and immunofluorescence experiments we demonstrated that the expression of CXCL12 increases at both mRNA (Fig. 5A) and protein levels (Fig. 5B, C) in CAFs exposed to insulin. Moreover, CXCL12 was found up-regulated in conditioned medium (CM) of CAFs treated with insulin (Fig. 5D), as ascertained by acetone precipitation assays. Altogether, these data suggest that insulin induces the increase of the CXCR4 ligand CXCL12 and stimulates its release by CAFs.

Metformin suppresses the insulin-driven feedforward loop linking CXCR4 induction in BCAHC-1 cells to CXCL12 expression and secretion by CAFs

Considering the pivotal role played by the CXCL12/CXCR4 axis in BC development and metastasis [51, 64, 67, 68], we evaluated whether insulin may engage the CXCL12/CXCR4 signaling, leading to aggressive features in BCAHC-1 cells. Hence, in order to visualize the F-actin pattern CM from CAFs exposed to vehicle and insulin was collected and used as culture medium in BCAHC-1 cells, which were previously treated with vehicle and insulin. These experimental conditions allowed us to observe a marked increase in F-actin assembly in BCAHC-1 cells treated with insulin and cultured with CM from insulin-stimulated CAFs (Fig. 6A, B). These effects were prevented using the specific CXCR4 antagonist AMD3100 and metformin (Fig. 6A, B). The aforementioned findings were corroborated by transwell migration assays performed in BCAHC-1 cells that were previously treated with insulin and cultured as described above. Of note, the migration of BCAHC-1 cells prompted by insulin was further potentiated culturing cells with CM obtained from insulin-treated CAFs (Fig. 6C, D), however this response was no longer evident treating BCAHC-1 cells with AMD3100 and metformin (Fig. 6C, D). Similarly, three-dimensional motility assays showed that AMD3100 and metformin prevent the outflow from the matrigel drop of insulin-treated BCAHC-1 cells cultured with CM from insulin-stimulated CAFs (Fig. 7A, B). Taken together, these results suggest that the insulin-activated CXCL12/CXCR4 axis sustains a paracrine feedforward loop coupling cancer and stromal cells, thus fostering a motile phenotype in BCAHC-1 cells.

Discussion

Insulin-resistance and related metabolic disorders, such as obesity, T2D and metabolic syndrome, contribute to cancer development and progression dysregulating many oncogenic pathways mediated by hormones, growth factors and cytokines [69, 70]. For instance, an early indicator of metabolic dysfunction as hyperinsulinemia, has been considered an independent risk and prognostic factor for BC [7173]. In this regard, insulin has been shown to facilitate BC progression through multiple signaling pathways that promote mitogenic and metastatic responses [13, 74, 75]. An additional insight toward a better understanding of insulin action in BC derives from the observation that the IR isoform A (IR-A) is frequently overexpressed in BC patients [17]. Contrary to the isoform B (IR-B) that is predominantly expressed in adult tissues and mediates metabolic effects of insulin, IR-A is involved in fetal development and is re-expressed in malignant tissues, where it triggers the oncogenic responses to insulin [17, 18, 76]. In addition, it should be mentioned that the dysregulation of the IR-A/IR-B ratio may be involved in pro-tumorigenic effects elicited by insulin [17, 77, 78]. In this scenario, we have recently isolated a naturally immortalized BC cell line named BCAHC‐1, which is distinguished by a peculiar receptor expression profile that may allow to better dissect the mechanisms underlying the stimulatory effects of insulin/IR-A axis in BC [27]. Considering that insulin and IGFs are able to bind to and activate each other’s receptors and share common downstream signaling pathways [78, 79], BCAHC‐1 cells may represent a valuable model system for a comprehensive evaluation of insulin/IR signaling in BC.
Metformin (1,1-dimethylbiguanide hydrochloride) is the most commonly prescribed drug for T2D treatment worldwide, displaying a remarkable balance between efficacy and safety profile [80]. The effectiveness of metformin in improving the sensitivity to insulin, and therefore in lowering insulin circulating levels, may provide a rationale to investigate the potential benefits of metformin in cancer patients affected by metabolic disorders [81]. As it concerns BC, several preclinical findings have demonstrated that metformin can interfere with cell proliferation and induce cell cycle arrest and apoptosis [25, 82]. Moreover, metformin may modify the expression of main pro-tumorigenic genes and exert a synergic action with chemotherapeutics in BC cells [8387]. Therefore, metformin has been suggested as a potential antitumoral agent in BC also considering the results of epidemiological studies in both diabetic and non-diabetic women [20, 26, 8890]. Cumulatively, these data recently paved the way for numerous trials aimed at investigating whether patients with diverse BC subtypes may take advantage from the use of metformin either alone or in combination with chemotherapeutics. In this regard, a recent meta-analysis revealed an increased objective response rate (ORR) in metformin-treated patients with inoperable BCs [91]. Moreover, data from a prospective study evidenced that the use of metformin is associated with a decreased risk of progression in ER-positive BC associated with T2D, whereas this beneficial effect was absent in ER-negative BC associated with T2D [40]. These investigations may suggest that ER is involved in the reduced risk of BC by metformin, likely through the inhibition of either ER expression or ER-mediated transcriptional activity [92]. Accordingly, we have previously demonstrated that a bi-directional crosstalk between ERα46 and IR may occur in BCAHC‐1 cells upon estrogen and insulin stimulation toward growth and pulmonary metastases [27], in line with the cooperation between ERα and IR transduction pathways assessed in BC cells [34, 35, 93].
In this study, we provide novel insights on the ability of metformin to prevent BC growth and motility stimulated by insulin/IR signaling. First, an integrated bioinformatics analysis allowed us to show that the expression and activation of IR are both associated with a worse outcome in ER-positive respect to ER-negative BC patients. Next, we focused on the characterization of the molecular mechanisms by which metformin might inhibit the stimulatory responses triggered by the insulin/IR axis in an IGF-deficient model system as BCAHC‐1 cells. In line with previous evidence demonstrating that metformin lowers IR phosphorylation [94], we observed that metformin inhibits the insulin-dependent activation of IR in BCAHC‐1 cells, without changes in IR expression. Previous studies have also reported a reduction or even an increase of IR protein levels upon metformin treatment, suggesting a differential regulation of IR expression depending on the cell context [9597]. We next ascertained that in BCAHC‐1 cells metformin prevents the activation of two main insulin-stimulated signaling pathways, namely PI3K/AKT and MAPK [41, 42], leading to the downregulation of insulin target genes like c-Fos and Cyclin D1 [27]. In accordance with these data, metformin was shown to down-regulate the expression of transcription factors as well as cell-cycle regulators [98, 99]. Nicely recapitulating these observations, we found that metformin inhibits the proliferative responses triggered by insulin in BCAHC-1 cells, including cell cycle progression, spheroids expansion and colony formation. Notably, metformin increased the proportion of BCAHC-1 cells in G0/G1 phase, reduced cell growth and inhibited the rate of colony formation. Reminiscing previous studies revealing that metformin exerts per se anti-proliferative effects in BC through diverse mechanisms [83], our current findings may suggest the potential usefulness of metformin toward new comprehensive strategies targeting BC.
The tumor microenvironment has been recognized as an important player in BC progression toward the acquisition of aggressive features [100]. CAFs, which are the largest population of stromal cells of BC, influence cancer growth and metastatic spread through the secretion of a variety of hormones, growth factors and inflammatory cytokines [101]. In this vein, a distinctive pro-inflammatory molecule released by CAFs is the chemokine CXCL12, which drives a metastatic phenotype interacting with the cognate receptor CXCR4 [5964, 102]. CXCR4 is a G protein-coupled chemokine receptor implicated in hemopoietic cell trafficking from bone marrow and lymphoid organs [103, 104]. In addition, emerging data have strengthened the prominent role of CXCR4 in regulating many aspects of BC progression as growth, invasion, angiogenesis, metastasis and resistance to therapies [51, 105]. In particular, the signaling cascade triggered by the CXCL12/CXCR4 axis orchestrates the chemotaxis of BC cells towards CXCL12-enriched tissues, which may therefore arrange the metastatic niche [51, 64, 67, 68, 106108]. In the present study, CXCR4 emerged as one of the most induced metastatic genes in BCAHC-1 cells exposed to insulin. In line with previous findings showing an association between CXCR4 expression and poor outcome in BC patients [109112], we performed bioinformatic analysis in ER-positive BC cohorts of patients. Notably, we found that high levels of both CXCR4 and IR are correlated with a worse DFI in ER-positive BC patients. On this basis and considering that a crosstalk between IGF and CXCR4 promotes migratory responses in BC cells [113], we assessed that the up-regulation of CXCR4 expression induced by insulin in BCAHC-1 cells is prevented by metformin. Of note, we also found that insulin triggers CXCL12 induction and release in CAFs. Consistent with the recognized ability of stromal cells to acquire a metastatic secretory phenotype upon insulin exposure [65, 66], we next demonstrated that insulin may drive a feed-forward loop coupling CXCL12 secretion by CAFs to CXCR4 induction in BCAHC-1 cells. Remarkably, we showed that metformin inhibits the acquisition of a migratory and invasive phenotype by BCAHC-1 cells sustained through the aforementioned cooperative network (as schematically depicted in Fig. 8). Nevertheless, further studies are warranted to determine whether metformin may prevent BC motility also via other mechanisms, for instance interfering with recently-identified targets of insulin signaling like the actin filament cross-linking protein filamin-A (FLNA) [114], which has been implicated in the growth and metastatic spread of BC and other hormone-dependent tumors [114118]. Overall, our data may provide novel insights on the usefulness of metformin in combination therapies targeting the growth effects and motility of the insulin/IR axis in BC.

Acknowledgements

We would like to thank the special award, namely “Department of Excellence 2018–2022” (Italian Law 232/2016) at the Department of Pharmacy, Health and Nutritional Sciences of the University of Calabria (Italy) and the “Sistema Integrato di Laboratori per L’Ambiente—(SILA) PONa3_00341”.

Declarations

All procedures conformed to the Helsinki Declaration for the research on humans. Signed informed consent was obtained from all patients and the experimental research has been performed with the ethical approval provided by the “Comitato Etico Ospedale Regionale, Cosenza, Italy” (approval code: 166, December 2nd, 2016).
All of the authors have consented to publication of this research.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.PubMed Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.PubMed
3.
Zurück zum Zitat Hendrick RE, Helvie MA, Monticciolo DL. Breast cancer mortality rates have stopped declining in US women younger than 40 years. Radiology. 2021;299:143–9.CrossRefPubMed Hendrick RE, Helvie MA, Monticciolo DL. Breast cancer mortality rates have stopped declining in US women younger than 40 years. Radiology. 2021;299:143–9.CrossRefPubMed
4.
Zurück zum Zitat Lei S, Zheng R, Zhang S, Wang S, Chen R, Sun K, et al. Global patterns of breast cancer incidence and mortality: a population-based cancer registry data analysis from 2000 to 2020. Cancer Commun. 2021;41:1183–94.CrossRef Lei S, Zheng R, Zhang S, Wang S, Chen R, Sun K, et al. Global patterns of breast cancer incidence and mortality: a population-based cancer registry data analysis from 2000 to 2020. Cancer Commun. 2021;41:1183–94.CrossRef
5.
Zurück zum Zitat Pei J, Wang Y, Li Y. Identification of key genes controlling breast cancer stem cell characteristics via stemness indices analysis. J Transl Med. 2020;18:74.CrossRefPubMedPubMedCentral Pei J, Wang Y, Li Y. Identification of key genes controlling breast cancer stem cell characteristics via stemness indices analysis. J Transl Med. 2020;18:74.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Lohmann AE, Goodwin PJ. Diabetes, metformin and breast cancer: a tangled web. Ann Oncol. 2021;32:285–6.CrossRefPubMed Lohmann AE, Goodwin PJ. Diabetes, metformin and breast cancer: a tangled web. Ann Oncol. 2021;32:285–6.CrossRefPubMed
7.
Zurück zum Zitat Godsland IF. Insulin resistance and hyperinsulinaemia in the development and progression of cancer. Clin Sci. 2009;118:315–32.CrossRef Godsland IF. Insulin resistance and hyperinsulinaemia in the development and progression of cancer. Clin Sci. 2009;118:315–32.CrossRef
8.
9.
Zurück zum Zitat Lawlor DA, Smith GD, Ebrahim S. Hyperinsulinaemia and increased risk of breast cancer: findings from the British women’s heart and health study. Cancer Causes Control. 2004;15:267–75.CrossRefPubMed Lawlor DA, Smith GD, Ebrahim S. Hyperinsulinaemia and increased risk of breast cancer: findings from the British women’s heart and health study. Cancer Causes Control. 2004;15:267–75.CrossRefPubMed
12.
Zurück zum Zitat Chan JY, LaPara K, Yee D. Disruption of insulin receptor function inhibits proliferation in endocrine-resistant breast cancer cells. Oncogene. 2016;35:4235–43.CrossRefPubMedPubMedCentral Chan JY, LaPara K, Yee D. Disruption of insulin receptor function inhibits proliferation in endocrine-resistant breast cancer cells. Oncogene. 2016;35:4235–43.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Rose DP, Vona-Davis L. The cellular and molecular mechanisms by which insulin influences breast cancer risk and progression. Endocr Relat Cancer. 2012;19:R225–41.CrossRefPubMed Rose DP, Vona-Davis L. The cellular and molecular mechanisms by which insulin influences breast cancer risk and progression. Endocr Relat Cancer. 2012;19:R225–41.CrossRefPubMed
14.
Zurück zum Zitat Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, et al. Insulin receptor isoforms in physiology and disease: an updated view. Endocr Rev. 2017;38:379–431.CrossRefPubMedPubMedCentral Belfiore A, Malaguarnera R, Vella V, Lawrence MC, Sciacca L, Frasca F, et al. Insulin receptor isoforms in physiology and disease: an updated view. Endocr Rev. 2017;38:379–431.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Vella V, Malaguarnera R, Nicolosi ML, Morrione A, Belfiore A. Insulin/IGF signaling and discoidin domain receptors: an emerging functional connection. Biochim Biophys Acta Mol Cell Res. 2019;1866: 118522.CrossRefPubMed Vella V, Malaguarnera R, Nicolosi ML, Morrione A, Belfiore A. Insulin/IGF signaling and discoidin domain receptors: an emerging functional connection. Biochim Biophys Acta Mol Cell Res. 2019;1866: 118522.CrossRefPubMed
16.
Zurück zum Zitat Sciacca L, Costantino A, Pandini G, Mineo R, Frasca F, Scalia P, et al. Insulin receptor activation by IGF-II in breast cancers: evidence for a new autocrine/paracrine mechanism. Oncogene. 1999;18:2471–9.CrossRefPubMed Sciacca L, Costantino A, Pandini G, Mineo R, Frasca F, Scalia P, et al. Insulin receptor activation by IGF-II in breast cancers: evidence for a new autocrine/paracrine mechanism. Oncogene. 1999;18:2471–9.CrossRefPubMed
18.
Zurück zum Zitat Aljada A, Saleh AM, Al-Aqeel SM, Shamsa HB, Al-Bawab A, Al Dubayee M, et al. Quantification of insulin receptor mRNA splice variants as a diagnostic tumor marker in breast cancer. Cancer Biomark. 2015;15:653–61.CrossRefPubMed Aljada A, Saleh AM, Al-Aqeel SM, Shamsa HB, Al-Bawab A, Al Dubayee M, et al. Quantification of insulin receptor mRNA splice variants as a diagnostic tumor marker in breast cancer. Cancer Biomark. 2015;15:653–61.CrossRefPubMed
19.
Zurück zum Zitat Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JMM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620–5.CrossRefPubMedPubMedCentral Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JMM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620–5.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Bosco JLF, Antonsen S, Sørensen HT, Pedersen L, Lash TL. Metformin and incident breast cancer among diabetic women: a population-based case–control study in Denmark. Cancer Epidemiol Biomark Prev. 2011;20:101–11.CrossRef Bosco JLF, Antonsen S, Sørensen HT, Pedersen L, Lash TL. Metformin and incident breast cancer among diabetic women: a population-based case–control study in Denmark. Cancer Epidemiol Biomark Prev. 2011;20:101–11.CrossRef
21.
Zurück zum Zitat Tang GH, Satkunam M, Pond GR, Steinberg GR, Blandino G, Schünemann HJ, et al. Association of metformin with breast cancer incidence and mortality in patients with type II diabetes: a GRADE-assessed systematic review and meta-analysis. Cancer Epidemiol Biomark Prev. 2018;27:627–35.CrossRef Tang GH, Satkunam M, Pond GR, Steinberg GR, Blandino G, Schünemann HJ, et al. Association of metformin with breast cancer incidence and mortality in patients with type II diabetes: a GRADE-assessed systematic review and meta-analysis. Cancer Epidemiol Biomark Prev. 2018;27:627–35.CrossRef
22.
Zurück zum Zitat Wang L, Zhang H-J, Liu Y-F, Chen G-Y. Impact of insulin use on outcomes of diabetic breast cancer patients: a systematic review and meta-analysis. Eur Rev Med Pharmacol Sci. 2021;25:3507–18.PubMed Wang L, Zhang H-J, Liu Y-F, Chen G-Y. Impact of insulin use on outcomes of diabetic breast cancer patients: a systematic review and meta-analysis. Eur Rev Med Pharmacol Sci. 2021;25:3507–18.PubMed
23.
Zurück zum Zitat Dowling RJO, Niraula S, Chang MC, Done SJ, Ennis M, McCready DR, et al. Changes in insulin receptor signaling underlie neoadjuvant metformin administration in breast cancer: a prospective window of opportunity neoadjuvant study. Breast Cancer Res. 2015;17:32.CrossRefPubMedPubMedCentral Dowling RJO, Niraula S, Chang MC, Done SJ, Ennis M, McCready DR, et al. Changes in insulin receptor signaling underlie neoadjuvant metformin administration in breast cancer: a prospective window of opportunity neoadjuvant study. Breast Cancer Res. 2015;17:32.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Cirillo F, Pellegrino M, Talia M, Perrotta ID, Rigiracciolo DC, Spinelli A, et al. Estrogen receptor variant ERα46 and insulin receptor drive in primary breast cancer cells growth effects and interleukin 11 induction prompting the motility of cancer-associated fibroblasts. Clin Transl Med. 2021;11: e516.CrossRefPubMedPubMedCentral Cirillo F, Pellegrino M, Talia M, Perrotta ID, Rigiracciolo DC, Spinelli A, et al. Estrogen receptor variant ERα46 and insulin receptor drive in primary breast cancer cells growth effects and interleukin 11 induction prompting the motility of cancer-associated fibroblasts. Clin Transl Med. 2021;11: e516.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Lappano R, Talia M, Cirillo F, Rigiracciolo DC, Scordamaglia D, Guzzi R, et al. The IL1β-IL1R signaling is involved in the stimulatory effects triggered by hypoxia in breast cancer cells and cancer-associated fibroblasts (CAFs). J Exp Clin Cancer Res. 2020;39:153.CrossRefPubMedPubMedCentral Lappano R, Talia M, Cirillo F, Rigiracciolo DC, Scordamaglia D, Guzzi R, et al. The IL1β-IL1R signaling is involved in the stimulatory effects triggered by hypoxia in breast cancer cells and cancer-associated fibroblasts (CAFs). J Exp Clin Cancer Res. 2020;39:153.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Lappano R, Rosano C, Santolla MF, Pupo M, De Francesco EM, De Marco P, et al. Two novel GPER agonists induce gene expression changes and growth effects in cancer cells. Curr Cancer Drug Targets. 2012;12:531–42.CrossRefPubMed Lappano R, Rosano C, Santolla MF, Pupo M, De Francesco EM, De Marco P, et al. Two novel GPER agonists induce gene expression changes and growth effects in cancer cells. Curr Cancer Drug Targets. 2012;12:531–42.CrossRefPubMed
30.
Zurück zum Zitat Fic E, Kedracka-Krok S, Jankowska U, Pirog A, Dziedzicka-Wasylewska M. Comparison of protein precipitation methods for various rat brain structures prior to proteomic analysis. Electrophoresis. 2010;31:3573–9.CrossRefPubMed Fic E, Kedracka-Krok S, Jankowska U, Pirog A, Dziedzicka-Wasylewska M. Comparison of protein precipitation methods for various rat brain structures prior to proteomic analysis. Electrophoresis. 2010;31:3573–9.CrossRefPubMed
31.
Zurück zum Zitat Szymczak M, Murray M, Petrovic N. Modulation of angiogenesis by ω-3 polyunsaturated fatty acids is mediated by cyclooxygenases. Blood. 2008;111:3514–21.CrossRefPubMed Szymczak M, Murray M, Petrovic N. Modulation of angiogenesis by ω-3 polyunsaturated fatty acids is mediated by cyclooxygenases. Blood. 2008;111:3514–21.CrossRefPubMed
32.
Zurück zum Zitat Aslan M, Hsu E-C, Liu S, Stoyanova T. Quantifying the invasion and migration ability of cancer cells with a 3D Matrigel drop invasion assay. Biol Methods Protoc. 2021;6: bpab014.CrossRefPubMedPubMedCentral Aslan M, Hsu E-C, Liu S, Stoyanova T. Quantifying the invasion and migration ability of cancer cells with a 3D Matrigel drop invasion assay. Biol Methods Protoc. 2021;6: bpab014.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Vella V, De Francesco EM, Lappano R, Muoio MG, Manzella L, Maggiolini M, et al. Microenvironmental determinants of breast cancer metastasis: focus on the crucial interplay between estrogen and insulin/insulin-like growth factor signaling. Front Cell Dev Biol. 2020;8: 608412.CrossRefPubMedPubMedCentral Vella V, De Francesco EM, Lappano R, Muoio MG, Manzella L, Maggiolini M, et al. Microenvironmental determinants of breast cancer metastasis: focus on the crucial interplay between estrogen and insulin/insulin-like growth factor signaling. Front Cell Dev Biol. 2020;8: 608412.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat De Marco P, Cirillo F, Vivacqua A, Malaguarnera R, Belfiore A, Maggiolini M. Novel aspects concerning the functional cross-talk between the insulin/IGF-I system and estrogen signaling in cancer cells. Front Endocrinol. 2015;6:30.CrossRef De Marco P, Cirillo F, Vivacqua A, Malaguarnera R, Belfiore A, Maggiolini M. Novel aspects concerning the functional cross-talk between the insulin/IGF-I system and estrogen signaling in cancer cells. Front Endocrinol. 2015;6:30.CrossRef
36.
Zurück zum Zitat Marinello PC, Panis C, Silva TNX, Binato R, Abdelhay E, Rodrigues JA, et al. Metformin prevention of doxorubicin resistance in MCF-7 and MDA-MB-231 involves oxidative stress generation and modulation of cell adaptation genes. Sci Rep. 2019;9:5864.CrossRefPubMedPubMedCentral Marinello PC, Panis C, Silva TNX, Binato R, Abdelhay E, Rodrigues JA, et al. Metformin prevention of doxorubicin resistance in MCF-7 and MDA-MB-231 involves oxidative stress generation and modulation of cell adaptation genes. Sci Rep. 2019;9:5864.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Jang SY, Kim A, Kim JK, Kim C, Cho Y-H, Kim J-H, et al. Metformin inhibits tumor cell migration via down-regulation of MMP9 in tamoxifen-resistant breast cancer cells. Anticancer Res. 2014;34:4127–34.PubMed Jang SY, Kim A, Kim JK, Kim C, Cho Y-H, Kim J-H, et al. Metformin inhibits tumor cell migration via down-regulation of MMP9 in tamoxifen-resistant breast cancer cells. Anticancer Res. 2014;34:4127–34.PubMed
38.
Zurück zum Zitat Bodmer M, Meier C, Krähenbühl S, Jick SS, Meier CR. Long-term metformin use is associated with decreased risk of breast cancer. Diabetes Care. 2010;33:1304–8.CrossRefPubMedPubMedCentral Bodmer M, Meier C, Krähenbühl S, Jick SS, Meier CR. Long-term metformin use is associated with decreased risk of breast cancer. Diabetes Care. 2010;33:1304–8.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Goodwin PJ, Pritchard KI, Ennis M, Clemons M, Graham M, Fantus IG. Insulin-lowering effects of metformin in women with early breast cancer. Clin Breast Cancer. 2008;8:501–5.CrossRefPubMed Goodwin PJ, Pritchard KI, Ennis M, Clemons M, Graham M, Fantus IG. Insulin-lowering effects of metformin in women with early breast cancer. Clin Breast Cancer. 2008;8:501–5.CrossRefPubMed
40.
Zurück zum Zitat Park Y-MM, Bookwalter DB, O’Brien KM, Jackson CL, Weinberg CR, Sandler DP. A prospective study of type 2 diabetes, metformin use, and risk of breast cancer. Ann Oncol. 2021;32:351–9.CrossRefPubMed Park Y-MM, Bookwalter DB, O’Brien KM, Jackson CL, Weinberg CR, Sandler DP. A prospective study of type 2 diabetes, metformin use, and risk of breast cancer. Ann Oncol. 2021;32:351–9.CrossRefPubMed
41.
Zurück zum Zitat Hopkins BD, Goncalves MD, Cantley LC. Insulin-PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nat Rev Endocrinol. 2020;16:276–83.CrossRefPubMedPubMedCentral Hopkins BD, Goncalves MD, Cantley LC. Insulin-PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nat Rev Endocrinol. 2020;16:276–83.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Lu C-C, Chu P-Y, Hsia S-M, Wu C-H, Tung Y-T, Yen G-C. Insulin induction instigates cell proliferation and metastasis in human colorectal cancer cells. Int J Oncol. 2017;50:736–44.CrossRefPubMed Lu C-C, Chu P-Y, Hsia S-M, Wu C-H, Tung Y-T, Yen G-C. Insulin induction instigates cell proliferation and metastasis in human colorectal cancer cells. Int J Oncol. 2017;50:736–44.CrossRefPubMed
43.
Zurück zum Zitat Shaw J, Kirshenbaum LA. Prime time for JNK-mediated Akt reactivation in hypoxia-reoxygenation. Circ Res. 2006;98:7–9.CrossRefPubMed Shaw J, Kirshenbaum LA. Prime time for JNK-mediated Akt reactivation in hypoxia-reoxygenation. Circ Res. 2006;98:7–9.CrossRefPubMed
44.
Zurück zum Zitat Ikeda K, Horie-Inoue K, Inoue S. Identification of estrogen-responsive genes based on the DNA binding properties of estrogen receptors using high-throughput sequencing technology. Acta Pharmacol Sin. 2015;36:24–31.CrossRefPubMed Ikeda K, Horie-Inoue K, Inoue S. Identification of estrogen-responsive genes based on the DNA binding properties of estrogen receptors using high-throughput sequencing technology. Acta Pharmacol Sin. 2015;36:24–31.CrossRefPubMed
45.
Zurück zum Zitat Keeton AB, Amsler MO, Venable DY, Messina JL. Insulin signal transduction pathways and insulin-induced gene expression. J Biol Chem. 2002;277:48565–73.CrossRefPubMed Keeton AB, Amsler MO, Venable DY, Messina JL. Insulin signal transduction pathways and insulin-induced gene expression. J Biol Chem. 2002;277:48565–73.CrossRefPubMed
46.
Zurück zum Zitat Gao F, Zhou L, Li M, Liu W, Yang S, Li W. Inhibition of ERKs/Akt-mediated c-Fos expression is required for piperlongumine-induced cyclin D1 downregulation and tumor suppression in colorectal cancer cells. Onco Targets Ther. 2020;13:5591–603.CrossRefPubMedPubMedCentral Gao F, Zhou L, Li M, Liu W, Yang S, Li W. Inhibition of ERKs/Akt-mediated c-Fos expression is required for piperlongumine-induced cyclin D1 downregulation and tumor suppression in colorectal cancer cells. Onco Targets Ther. 2020;13:5591–603.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Wang J-C, Li G-Y, Wang B, Han S-X, Sun X, Jiang Y-N, et al. Metformin inhibits metastatic breast cancer progression and improves chemosensitivity by inducing vessel normalization via PDGF-B downregulation. J Exp Clin Cancer Res. 2019;38:235.CrossRefPubMedPubMedCentral Wang J-C, Li G-Y, Wang B, Han S-X, Sun X, Jiang Y-N, et al. Metformin inhibits metastatic breast cancer progression and improves chemosensitivity by inducing vessel normalization via PDGF-B downregulation. J Exp Clin Cancer Res. 2019;38:235.CrossRefPubMedPubMedCentral
49.
50.
Zurück zum Zitat Cerezo M, Tichet M, Abbe P, Ohanna M, Lehraiki A, Rouaud F, et al. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Mol Cancer Ther. 2013;12:1605–15.CrossRefPubMed Cerezo M, Tichet M, Abbe P, Ohanna M, Lehraiki A, Rouaud F, et al. Metformin blocks melanoma invasion and metastasis development in AMPK/p53-dependent manner. Mol Cancer Ther. 2013;12:1605–15.CrossRefPubMed
52.
Zurück zum Zitat Chen IX, Chauhan VP, Posada J, Ng MR, Wu MW, Adstamongkonkul P, et al. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc Natl Acad Sci USA. 2019;116:4558–66.CrossRefPubMedPubMedCentral Chen IX, Chauhan VP, Posada J, Ng MR, Wu MW, Adstamongkonkul P, et al. Blocking CXCR4 alleviates desmoplasia, increases T-lymphocyte infiltration, and improves immunotherapy in metastatic breast cancer. Proc Natl Acad Sci USA. 2019;116:4558–66.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Li Z-H, Xiong Q-Y, Xu L, Duan P, Yang QO, Zhou P, et al. miR-29a regulated ER-positive breast cancer cell growth and invasion and is involved in the insulin signaling pathway. Oncotarget. 2017;8:32566–75.CrossRefPubMedPubMedCentral Li Z-H, Xiong Q-Y, Xu L, Duan P, Yang QO, Zhou P, et al. miR-29a regulated ER-positive breast cancer cell growth and invasion and is involved in the insulin signaling pathway. Oncotarget. 2017;8:32566–75.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Al-Souhibani N, Al-Ghamdi M, Al-Ahmadi W, Khabar KSA. Posttranscriptional control of the chemokine receptor CXCR4 expression in cancer cells. Carcinogenesis. 2014;35:1983–92.CrossRefPubMedPubMedCentral Al-Souhibani N, Al-Ghamdi M, Al-Ahmadi W, Khabar KSA. Posttranscriptional control of the chemokine receptor CXCR4 expression in cancer cells. Carcinogenesis. 2014;35:1983–92.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Lappano R, Maggiolini M. GPER is involved in the functional liaison between breast tumor cells and cancer-associated fibroblasts (CAFs). J Steroid Biochem Mol Biol. 2018;176:49–56.CrossRefPubMed Lappano R, Maggiolini M. GPER is involved in the functional liaison between breast tumor cells and cancer-associated fibroblasts (CAFs). J Steroid Biochem Mol Biol. 2018;176:49–56.CrossRefPubMed
57.
Zurück zum Zitat Mehraj U, Dar AH, Wani NA, Mir MA. Tumor microenvironment promotes breast cancer chemoresistance. Cancer Chemother Pharmacol. 2021;87:147–58.CrossRefPubMed Mehraj U, Dar AH, Wani NA, Mir MA. Tumor microenvironment promotes breast cancer chemoresistance. Cancer Chemother Pharmacol. 2021;87:147–58.CrossRefPubMed
58.
Zurück zum Zitat De Marco P, Lappano R, De Francesco EM, Cirillo F, Pupo M, Avino S, et al. GPER signalling in both cancer-associated fibroblasts and breast cancer cells mediates a feedforward IL1β/IL1R1 response. Sci Rep. 2016;6:24354.CrossRefPubMedPubMedCentral De Marco P, Lappano R, De Francesco EM, Cirillo F, Pupo M, Avino S, et al. GPER signalling in both cancer-associated fibroblasts and breast cancer cells mediates a feedforward IL1β/IL1R1 response. Sci Rep. 2016;6:24354.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Lappano R, Rigiracciolo DC, Belfiore A, Maggiolini M, De Francesco EM. Cancer associated fibroblasts: role in breast cancer and potential as therapeutic targets. Expert Opin Ther Targets. 2020;24:559–72.CrossRefPubMed Lappano R, Rigiracciolo DC, Belfiore A, Maggiolini M, De Francesco EM. Cancer associated fibroblasts: role in breast cancer and potential as therapeutic targets. Expert Opin Ther Targets. 2020;24:559–72.CrossRefPubMed
60.
Zurück zum Zitat Zhang F, Cui J-Y, Gao H-F, Yu H, Gao F-F, Chen J-L, et al. Cancer-associated fibroblasts induce epithelial–mesenchymal transition and cisplatin resistance in ovarian cancer via CXCL12/CXCR4 axis. Future Oncol. 2020;16:2619–33.CrossRefPubMed Zhang F, Cui J-Y, Gao H-F, Yu H, Gao F-F, Chen J-L, et al. Cancer-associated fibroblasts induce epithelial–mesenchymal transition and cisplatin resistance in ovarian cancer via CXCL12/CXCR4 axis. Future Oncol. 2020;16:2619–33.CrossRefPubMed
61.
Zurück zum Zitat Mousavi A. CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted-therapy. Immunol Lett. 2020;217:91–115.CrossRefPubMed Mousavi A. CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted-therapy. Immunol Lett. 2020;217:91–115.CrossRefPubMed
62.
Zurück zum Zitat Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin Cancer Res. 2010;16:2927–31.CrossRefPubMed Teicher BA, Fricker SP. CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin Cancer Res. 2010;16:2927–31.CrossRefPubMed
63.
Zurück zum Zitat Britton C, Poznansky MC, Reeves P. Polyfunctionality of the CXCR4/CXCL12 axis in health and disease: implications for therapeutic interventions in cancer and immune-mediated diseases. FASEB J. 2021;35: e21260.CrossRefPubMed Britton C, Poznansky MC, Reeves P. Polyfunctionality of the CXCR4/CXCL12 axis in health and disease: implications for therapeutic interventions in cancer and immune-mediated diseases. FASEB J. 2021;35: e21260.CrossRefPubMed
64.
Zurück zum Zitat Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–48.CrossRefPubMed Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:335–48.CrossRefPubMed
65.
Zurück zum Zitat Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE. Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer. 2018;17:66.CrossRefPubMedPubMedCentral Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE. Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer. 2018;17:66.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Yang J, Waldron RT, Su H-Y, Moro A, Chang H-H, Eibl G, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311:G675–87.CrossRefPubMedPubMedCentral Yang J, Waldron RT, Su H-Y, Moro A, Chang H-H, Eibl G, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2016;311:G675–87.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Sun Y, Mao X, Fan C, Liu C, Guo A, Guan S, et al. CXCL12-CXCR4 axis promotes the natural selection of breast cancer cell metastasis. Tumour Biol. 2014;35:7765–73.CrossRefPubMed Sun Y, Mao X, Fan C, Liu C, Guo A, Guan S, et al. CXCL12-CXCR4 axis promotes the natural selection of breast cancer cell metastasis. Tumour Biol. 2014;35:7765–73.CrossRefPubMed
68.
Zurück zum Zitat Burger JA, Kipps TJ. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006;107:1761–7.CrossRefPubMed Burger JA, Kipps TJ. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood. 2006;107:1761–7.CrossRefPubMed
69.
Zurück zum Zitat Xue F, Michels KB. Diabetes, metabolic syndrome, and breast cancer: a review of the current evidence. Am J Clin Nutr. 2007;86:s823–35.CrossRefPubMed Xue F, Michels KB. Diabetes, metabolic syndrome, and breast cancer: a review of the current evidence. Am J Clin Nutr. 2007;86:s823–35.CrossRefPubMed
71.
72.
Zurück zum Zitat Tsujimoto T, Kajio H, Sugiyama T. Association between hyperinsulinemia and increased risk of cancer death in nonobese and obese people: a population-based observational study. Int J Cancer. 2017;141:102–11.CrossRefPubMedPubMedCentral Tsujimoto T, Kajio H, Sugiyama T. Association between hyperinsulinemia and increased risk of cancer death in nonobese and obese people: a population-based observational study. Int J Cancer. 2017;141:102–11.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Senapati P, Kato H, Lee M, Leung A, Thai C, Sanchez A, et al. Hyperinsulinemia promotes aberrant histone acetylation in triple-negative breast cancer. Epigenet Chromatin. 2019;12:44.CrossRef Senapati P, Kato H, Lee M, Leung A, Thai C, Sanchez A, et al. Hyperinsulinemia promotes aberrant histone acetylation in triple-negative breast cancer. Epigenet Chromatin. 2019;12:44.CrossRef
75.
Zurück zum Zitat Siklova-Vitkova M, Polak J, Klimcakova E, Vrzalova J, Hejnova J, Kovacikova M, et al. Effect of hyperinsulinemia and very-low-calorie diet on interstitial cytokine levels in subcutaneous adipose tissue of obese women. Am J Physiol Endocrinol Metab. 2009;297:E1154–61.CrossRefPubMed Siklova-Vitkova M, Polak J, Klimcakova E, Vrzalova J, Hejnova J, Kovacikova M, et al. Effect of hyperinsulinemia and very-low-calorie diet on interstitial cytokine levels in subcutaneous adipose tissue of obese women. Am J Physiol Endocrinol Metab. 2009;297:E1154–61.CrossRefPubMed
76.
Zurück zum Zitat Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30:586–623.CrossRefPubMed Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr Rev. 2009;30:586–623.CrossRefPubMed
77.
Zurück zum Zitat Frasca F, Pandini G, Scalia P, Sciacca L, Mineo R, Costantino A, et al. Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol Cell Biol. 1999;19:3278–88.CrossRefPubMedPubMedCentral Frasca F, Pandini G, Scalia P, Sciacca L, Mineo R, Costantino A, et al. Insulin receptor isoform A, a newly recognized, high-affinity insulin-like growth factor II receptor in fetal and cancer cells. Mol Cell Biol. 1999;19:3278–88.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Belfiore A. The role of insulin receptor isoforms and hybrid insulin/IGF-I receptors in human cancer. Curr Pharm Des. 2007;13:671–86.CrossRefPubMed Belfiore A. The role of insulin receptor isoforms and hybrid insulin/IGF-I receptors in human cancer. Curr Pharm Des. 2007;13:671–86.CrossRefPubMed
79.
Zurück zum Zitat Singh P, Alex JM, Bast F. Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: novel treatment strategies for cancer. Med Oncol. 2014;31:805.CrossRefPubMed Singh P, Alex JM, Bast F. Insulin receptor (IR) and insulin-like growth factor receptor 1 (IGF-1R) signaling systems: novel treatment strategies for cancer. Med Oncol. 2014;31:805.CrossRefPubMed
81.
Zurück zum Zitat Saengboonmee C, Sanlung T, Wongkham S. Repurposing metformin for cancer treatment: a great challenge of a promising drug. Anticancer Res. 2021;41:5913–8.CrossRefPubMed Saengboonmee C, Sanlung T, Wongkham S. Repurposing metformin for cancer treatment: a great challenge of a promising drug. Anticancer Res. 2021;41:5913–8.CrossRefPubMed
82.
Zurück zum Zitat Malki A, Youssef A. Antidiabetic drug metformin induces apoptosis in human MCF breast cancer via targeting ERK signaling. Oncol Res. 2011;19:275–85.CrossRefPubMed Malki A, Youssef A. Antidiabetic drug metformin induces apoptosis in human MCF breast cancer via targeting ERK signaling. Oncol Res. 2011;19:275–85.CrossRefPubMed
84.
Zurück zum Zitat Farahi A, Abedini MR, Javdani H, Arzi L, Chamani E, Farhoudi R, et al. Crocin and Metformin suppress metastatic breast cancer progression via VEGF and MMP9 downregulations: in vitro and in vivo studies. Mol Cell Biochem. 2021;476:3341–51.CrossRefPubMed Farahi A, Abedini MR, Javdani H, Arzi L, Chamani E, Farhoudi R, et al. Crocin and Metformin suppress metastatic breast cancer progression via VEGF and MMP9 downregulations: in vitro and in vivo studies. Mol Cell Biochem. 2021;476:3341–51.CrossRefPubMed
85.
Zurück zum Zitat Cha J-H, Yang W-H, Xia W, Wei Y, Chan L-C, Lim S-O, et al. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol Cell. 2018;71:606-620.e7.CrossRefPubMedPubMedCentral Cha J-H, Yang W-H, Xia W, Wei Y, Chan L-C, Lim S-O, et al. Metformin promotes antitumor immunity via endoplasmic-reticulum-associated degradation of PD-L1. Mol Cell. 2018;71:606-620.e7.CrossRefPubMedPubMedCentral
86.
Zurück zum Zitat Cheng L, Zhang X, Huang Y-Z, Zhu Y-L, Xu L-Y, Li Z, et al. Metformin exhibits antiproliferation activity in breast cancer via miR-483-3p/METTL3/m6A/p21 pathway. Oncogenesis. 2021;10:7.CrossRefPubMedPubMedCentral Cheng L, Zhang X, Huang Y-Z, Zhu Y-L, Xu L-Y, Li Z, et al. Metformin exhibits antiproliferation activity in breast cancer via miR-483-3p/METTL3/m6A/p21 pathway. Oncogenesis. 2021;10:7.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Col NF, Ochs L, Springmann V, Aragaki AK, Chlebowski RT. Metformin and breast cancer risk: a meta-analysis and critical literature review. Breast Cancer Res Treat. 2012;135:639–46.CrossRefPubMed Col NF, Ochs L, Springmann V, Aragaki AK, Chlebowski RT. Metformin and breast cancer risk: a meta-analysis and critical literature review. Breast Cancer Res Treat. 2012;135:639–46.CrossRefPubMed
89.
Zurück zum Zitat Niraula S, Dowling RJO, Ennis M, Chang MC, Done SJ, Hood N, et al. Metformin in early breast cancer: a prospective window of opportunity neoadjuvant study. Breast Cancer Res Treat. 2012;135:821–30.CrossRefPubMed Niraula S, Dowling RJO, Ennis M, Chang MC, Done SJ, Hood N, et al. Metformin in early breast cancer: a prospective window of opportunity neoadjuvant study. Breast Cancer Res Treat. 2012;135:821–30.CrossRefPubMed
90.
Zurück zum Zitat Hadad SM, Coates P, Jordan LB, Dowling RJO, Chang MC, Done SJ, et al. Evidence for biological effects of metformin in operable breast cancer: biomarker analysis in a pre-operative window of opportunity randomized trial. Breast Cancer Res Treat. 2015;150:149–55.CrossRefPubMed Hadad SM, Coates P, Jordan LB, Dowling RJO, Chang MC, Done SJ, et al. Evidence for biological effects of metformin in operable breast cancer: biomarker analysis in a pre-operative window of opportunity randomized trial. Breast Cancer Res Treat. 2015;150:149–55.CrossRefPubMed
91.
Zurück zum Zitat Wu Z, Qu B, Huang X, Song Y, Gao P, Shi J, et al. The potential adjunctive benefit of adding metformin to standard treatment in inoperable cancer patients: a meta-analysis of randomized controlled trials. Ann Transl Med. 2020;8:1404.CrossRefPubMedPubMedCentral Wu Z, Qu B, Huang X, Song Y, Gao P, Shi J, et al. The potential adjunctive benefit of adding metformin to standard treatment in inoperable cancer patients: a meta-analysis of randomized controlled trials. Ann Transl Med. 2020;8:1404.CrossRefPubMedPubMedCentral
92.
Zurück zum Zitat Kim J, Lee J, Jang SY, Kim C, Choi Y, Kim A. Anticancer effect of metformin on estrogen receptor-positive and tamoxifen-resistant breast cancer cell lines. Oncol Rep. 2016;35:2553–60.CrossRefPubMedPubMedCentral Kim J, Lee J, Jang SY, Kim C, Choi Y, Kim A. Anticancer effect of metformin on estrogen receptor-positive and tamoxifen-resistant breast cancer cell lines. Oncol Rep. 2016;35:2553–60.CrossRefPubMedPubMedCentral
93.
Zurück zum Zitat Lanzino M, Morelli C, Garofalo C, Panno ML, Mauro L, Andò S, et al. Interaction between estrogen receptor alpha and insulin/IGF signaling in breast cancer. Curr Cancer Drug Targets. 2008;8:597–610.CrossRefPubMed Lanzino M, Morelli C, Garofalo C, Panno ML, Mauro L, Andò S, et al. Interaction between estrogen receptor alpha and insulin/IGF signaling in breast cancer. Curr Cancer Drug Targets. 2008;8:597–610.CrossRefPubMed
94.
Zurück zum Zitat Quinn BJ, Dallos M, Kitagawa H, Kunnumakkara AB, Memmott RM, Hollander MC, et al. Inhibition of lung tumorigenesis by metformin is associated with decreased plasma IGF-I and diminished receptor tyrosine kinase signaling. Cancer Prev Res. 2013;6:801–10.CrossRef Quinn BJ, Dallos M, Kitagawa H, Kunnumakkara AB, Memmott RM, Hollander MC, et al. Inhibition of lung tumorigenesis by metformin is associated with decreased plasma IGF-I and diminished receptor tyrosine kinase signaling. Cancer Prev Res. 2013;6:801–10.CrossRef
95.
Zurück zum Zitat Sarfstein R, Friedman Y, Attias-Geva Z, Fishman A, Bruchim I, Werner H. Metformin downregulates the insulin/IGF-I signaling pathway and inhibits different uterine serous carcinoma (USC) cells proliferation and migration in p53-dependent or -independent manners. PLoS ONE. 2013;8: e61537.CrossRefPubMedPubMedCentral Sarfstein R, Friedman Y, Attias-Geva Z, Fishman A, Bruchim I, Werner H. Metformin downregulates the insulin/IGF-I signaling pathway and inhibits different uterine serous carcinoma (USC) cells proliferation and migration in p53-dependent or -independent manners. PLoS ONE. 2013;8: e61537.CrossRefPubMedPubMedCentral
96.
Zurück zum Zitat Memmott RM, Mercado JR, Maier CR, Kawabata S, Fox SD, Dennis PA. Metformin prevents tobacco carcinogen-induced lung tumorigenesis. Cancer Prev Res. 2010;3:1066–76.CrossRef Memmott RM, Mercado JR, Maier CR, Kawabata S, Fox SD, Dennis PA. Metformin prevents tobacco carcinogen-induced lung tumorigenesis. Cancer Prev Res. 2010;3:1066–76.CrossRef
97.
Zurück zum Zitat Villalva-Pérez JM, Ramírez-Vargas MA, Serafín-Fabían JI, Ramírez M, Elena Moreno-Godínez M, Espinoza-Rojo M, et al. Characterization of Huh7 cells after the induction of insulin resistance and post-treatment with metformin. Cytotechnology. 2020;72:499–511.CrossRefPubMedPubMedCentral Villalva-Pérez JM, Ramírez-Vargas MA, Serafín-Fabían JI, Ramírez M, Elena Moreno-Godínez M, Espinoza-Rojo M, et al. Characterization of Huh7 cells after the induction of insulin resistance and post-treatment with metformin. Cytotechnology. 2020;72:499–511.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Akinyeke T, Matsumura S, Wang X, Wu Y, Schalfer ED, Saxena A, et al. Metformin targets c-MYC oncogene to prevent prostate cancer. Carcinogenesis. 2013;34:2823–32.CrossRefPubMedPubMedCentral Akinyeke T, Matsumura S, Wang X, Wu Y, Schalfer ED, Saxena A, et al. Metformin targets c-MYC oncogene to prevent prostate cancer. Carcinogenesis. 2013;34:2823–32.CrossRefPubMedPubMedCentral
99.
Zurück zum Zitat Gwak H, Kim Y, An H, Dhanasekaran DN, Song YS. Metformin induces degradation of cyclin D1 via AMPK/GSK3β axis in ovarian cancer. Mol Carcinog. 2017;56:349–58.CrossRefPubMed Gwak H, Kim Y, An H, Dhanasekaran DN, Song YS. Metformin induces degradation of cyclin D1 via AMPK/GSK3β axis in ovarian cancer. Mol Carcinog. 2017;56:349–58.CrossRefPubMed
100.
Zurück zum Zitat Luo H, Tu G, Liu Z, Liu M. Cancer-associated fibroblasts: a multifaceted driver of breast cancer progression. Cancer Lett. 2015;361:155–63.CrossRefPubMed Luo H, Tu G, Liu Z, Liu M. Cancer-associated fibroblasts: a multifaceted driver of breast cancer progression. Cancer Lett. 2015;361:155–63.CrossRefPubMed
101.
Zurück zum Zitat Kim HM, Jung WH, Koo JS. Expression of cancer-associated fibroblast related proteins in metastatic breast cancer: an immunohistochemical analysis. J Transl Med. 2015;13:222.CrossRefPubMedPubMedCentral Kim HM, Jung WH, Koo JS. Expression of cancer-associated fibroblast related proteins in metastatic breast cancer: an immunohistochemical analysis. J Transl Med. 2015;13:222.CrossRefPubMedPubMedCentral
102.
Zurück zum Zitat Rubie C, Frick VO, Ghadjar P, Wagner M, Justinger C, Faust SK, et al. CXC receptor-4 mRNA silencing abrogates CXCL12-induced migration of colorectal cancer cells. J Transl Med. 2011;9:22.CrossRefPubMedPubMedCentral Rubie C, Frick VO, Ghadjar P, Wagner M, Justinger C, Faust SK, et al. CXC receptor-4 mRNA silencing abrogates CXCL12-induced migration of colorectal cancer cells. J Transl Med. 2011;9:22.CrossRefPubMedPubMedCentral
103.
104.
Zurück zum Zitat Möhle R, Bautz F, Rafii S, Moore MA, Brugger W, Kanz L. The chemokine receptor CXCR-4 is expressed on CD34+ hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood. 1998;91:4523–30.CrossRefPubMed Möhle R, Bautz F, Rafii S, Moore MA, Brugger W, Kanz L. The chemokine receptor CXCR-4 is expressed on CD34+ hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood. 1998;91:4523–30.CrossRefPubMed
105.
Zurück zum Zitat Xiang J, Hurchla MA, Fontana F, Su X, Amend SR, Esser AK, et al. CXCR4 protein epitope mimetic antagonist POL5551 disrupts metastasis and enhances chemotherapy effect in triple-negative breast cancer. Mol Cancer Ther. 2015;14:2473–85.CrossRefPubMedPubMedCentral Xiang J, Hurchla MA, Fontana F, Su X, Amend SR, Esser AK, et al. CXCR4 protein epitope mimetic antagonist POL5551 disrupts metastasis and enhances chemotherapy effect in triple-negative breast cancer. Mol Cancer Ther. 2015;14:2473–85.CrossRefPubMedPubMedCentral
106.
107.
Zurück zum Zitat Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, Reyes-Soto G, Ortiz-Sánchez E, Herrera-Montalvo LA. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med. 2020;18:1.CrossRefPubMedPubMedCentral Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, Reyes-Soto G, Ortiz-Sánchez E, Herrera-Montalvo LA. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med. 2020;18:1.CrossRefPubMedPubMedCentral
108.
Zurück zum Zitat Fusi A, Liu Z, Kümmerlen V, Nonnemacher A, Jeske J, Keilholz U. Expression of chemokine receptors on circulating tumor cells in patients with solid tumors. J Transl Med. 2012;10:52.CrossRefPubMedPubMedCentral Fusi A, Liu Z, Kümmerlen V, Nonnemacher A, Jeske J, Keilholz U. Expression of chemokine receptors on circulating tumor cells in patients with solid tumors. J Transl Med. 2012;10:52.CrossRefPubMedPubMedCentral
109.
Zurück zum Zitat Zhao H, Guo L, Zhao H, Zhao J, Weng H, Zhao B. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6:5022–40.CrossRefPubMed Zhao H, Guo L, Zhao H, Zhao J, Weng H, Zhao B. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6:5022–40.CrossRefPubMed
110.
Zurück zum Zitat Mukherjee D, Zhao J. The role of chemokine receptor CXCR4 in breast cancer metastasis. Am J Cancer Res. 2013;3:46–57.PubMedPubMedCentral Mukherjee D, Zhao J. The role of chemokine receptor CXCR4 in breast cancer metastasis. Am J Cancer Res. 2013;3:46–57.PubMedPubMedCentral
111.
Zurück zum Zitat Zhang Z, Ni C, Chen W, Wu P, Wang Z, Yin J, et al. Expression of CXCR4 and breast cancer prognosis: a systematic review and meta-analysis. BMC Cancer. 2014;14:49.CrossRefPubMedPubMedCentral Zhang Z, Ni C, Chen W, Wu P, Wang Z, Yin J, et al. Expression of CXCR4 and breast cancer prognosis: a systematic review and meta-analysis. BMC Cancer. 2014;14:49.CrossRefPubMedPubMedCentral
112.
Zurück zum Zitat Zhang L, Ye S-B, Ma G, Tang X-F, Chen S-P, He J, et al. The expressions of MIF and CXCR4 protein in tumor microenvironment are adverse prognostic factors in patients with esophageal squamous cell carcinoma. J Transl Med. 2013;11:60.CrossRefPubMedPubMedCentral Zhang L, Ye S-B, Ma G, Tang X-F, Chen S-P, He J, et al. The expressions of MIF and CXCR4 protein in tumor microenvironment are adverse prognostic factors in patients with esophageal squamous cell carcinoma. J Transl Med. 2013;11:60.CrossRefPubMedPubMedCentral
113.
Zurück zum Zitat Akekawatchai C, Holland JD, Kochetkova M, Wallace JC, McColl SR. Transactivation of CXCR4 by the insulin-like growth factor-1 receptor (IGF-1R) in human MDA-MB-231 breast cancer epithelial cells. J Biol Chem. 2005;280:39701–8.CrossRefPubMed Akekawatchai C, Holland JD, Kochetkova M, Wallace JC, McColl SR. Transactivation of CXCR4 by the insulin-like growth factor-1 receptor (IGF-1R) in human MDA-MB-231 breast cancer epithelial cells. J Biol Chem. 2005;280:39701–8.CrossRefPubMed
114.
Zurück zum Zitat Aizen D, Pasmanik-Chor M, Sarfstein R, Laron Z, Bruchim I, Werner H. Genome-wide analyses identify filamin-A as a novel downstream target for insulin and IGF1 action. Front Endocrinol. 2018;9:105.CrossRef Aizen D, Pasmanik-Chor M, Sarfstein R, Laron Z, Bruchim I, Werner H. Genome-wide analyses identify filamin-A as a novel downstream target for insulin and IGF1 action. Front Endocrinol. 2018;9:105.CrossRef
115.
Zurück zum Zitat Castoria G, D’Amato L, Ciociola A, Giovannelli P, Giraldi T, Sepe L, et al. Androgen-induced cell migration: role of androgen receptor/filamin A association. PLoS ONE. 2011;6: e17218.CrossRefPubMedPubMedCentral Castoria G, D’Amato L, Ciociola A, Giovannelli P, Giraldi T, Sepe L, et al. Androgen-induced cell migration: role of androgen receptor/filamin A association. PLoS ONE. 2011;6: e17218.CrossRefPubMedPubMedCentral
117.
Zurück zum Zitat Zhou J, Wu L, Xu P, Li Y, Ji Z, Kang X. Filamin A is a potential driver of breast cancer metastasis via regulation of MMP-1. Front Oncol. 2022;12: 836126.CrossRefPubMedPubMedCentral Zhou J, Wu L, Xu P, Li Y, Ji Z, Kang X. Filamin A is a potential driver of breast cancer metastasis via regulation of MMP-1. Front Oncol. 2022;12: 836126.CrossRefPubMedPubMedCentral
118.
Zurück zum Zitat Jiang X, Yue J, Lu H, Campbell N, Yang Q, Lan S, et al. Inhibition of filamin-A reduces cancer metastatic potential. Int J Biol Sci. 2013;9:67–77.CrossRefPubMed Jiang X, Yue J, Lu H, Campbell N, Yang Q, Lan S, et al. Inhibition of filamin-A reduces cancer metastatic potential. Int J Biol Sci. 2013;9:67–77.CrossRefPubMed
Metadaten
Titel
Metformin counteracts stimulatory effects induced by insulin in primary breast cancer cells
verfasst von
Domenica Scordamaglia
Francesca Cirillo
Marianna Talia
Maria Francesca Santolla
Damiano Cosimo Rigiracciolo
Lucia Muglia
Azzurra Zicarelli
Salvatore De Rosis
Francesca Giordano
Anna Maria Miglietta
Ernestina Marianna De Francesco
Veronica Vella
Antonino Belfiore
Rosamaria Lappano
Marcello Maggiolini
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03463-y

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.