Paradoxic effects of metformin on endothelial cells and angiogenesis

Katiuscia Dallaglio, Antonino Bruno, Anna R Cantelmo, Alessia I Esposito, Luca Ruggiero, Stefania Orecchioni, Angelica Calleri, Francesco Bertolini, Ulrich Pfeffer, Douglas M Noonan, Adriana Albini, Katiuscia Dallaglio, Antonino Bruno, Anna R Cantelmo, Alessia I Esposito, Luca Ruggiero, Stefania Orecchioni, Angelica Calleri, Francesco Bertolini, Ulrich Pfeffer, Douglas M Noonan, Adriana Albini

Abstract

The biguanide metformin is used in type 2 diabetes management and has gained significant attention as a potential cancer preventive agent. Angioprevention represents a mechanism of chemoprevention, yet conflicting data concerning the antiangiogenic action of metformin have emerged. Here, we clarify some of the contradictory effects of metformin on endothelial cells and angiogenesis, using in vitro and in vivo assays combined with transcriptomic and protein array approaches. Metformin inhibits formation of capillary-like networks by endothelial cells; this effect is partially dependent on the energy sensor adenosine-monophosphate-activated protein kinase (AMPK) as shown by small interfering RNA knockdown. Gene expression profiling of human umbilical vein endothelial cells revealed a paradoxical modulation of several angiogenesis-associated genes and proteins by metformin, with short-term induction of vascular endothelial growth factor (VEGF), cyclooxygenase 2 and CXC chemokine receptor 4 at the messenger RNA level and downregulation of ADAMTS1. Antibody array analysis shows an essentially opposite regulation of numerous angiogenesis-associated proteins in endothelial and breast cancer cells including interleukin-8, angiogenin and TIMP-1, as well as selective regulation of angiopioetin-1, -2, endoglin and others. Endothelial cell production of the cytochrome P450 member CYP1B1 is upregulated by tumor cell supernatants in an AMPK-dependent manner, metformin blocks this effect. Metformin inhibits VEGF-dependent activation of extracellular signal-regulated kinase 1/2, and the inhibition of AMPK activity abrogates this event. Metformin hinders angiogenesis in matrigel pellets in vivo, prevents the microvessel density increase observed in obese mice on a high-fat diet, downregulating the number of white adipose tissue endothelial precursor cells. Our data show that metformin has an antiangiogenic activity in vitro and in vivo associated with a contradictory short-term enhancement of pro-angiogenic mediators, as well as with a differential regulation in endothelial and breast cancer cells.

Figures

Fig. 1.
Fig. 1.
Metformin inhibits endothelial cell morphogenesis. HUVE cells were treated with metformin at the indicated concentrations and seeded on the top of the matrigel layer previously solidified on 24-well plates. The formation of capillary-like networks was documented by photography after 6h (×5 magnification). (A) Metformin (Metf) inhibited morphogenesis in a dose-dependent manner. (B) Quantification of capillary-like structures (both number of segments and segments length) using the Angiogenesis Analyzer ImageJ toolkit showed significant differences in both parameters, P-values are shown, C− is the negative control. (C) HUVE cells were transfected with an AMPKα1-specific (siRNAα1 or α1) control (siCTR) siRNA, cell lysates were harvested and analyzed by western blotting for AMPKα1 expression; AMPKα1 was successfully downregulated in HUVE transfected cells as compared with control. (D) Transfected HUVE cells were seeded on top of a matrigel layer in presence or absence of metformin and analyzed at 6h as in (A). Metformin-mediated inhibition of capillary-like structures formation was reverted when AMPKα1 expression was downregulated. (E) Quantification of capillary-like structures as described previously, P-values are shown.
Fig. 2.
Fig. 2.
Metformin modulates several angiogenesis-related genes. Microarray analysis was performed on total RNAs isolated from HUVE cells from three different donors that were treated with either vehicle alone (CTR) or 6 or 24h with 10mM metformin (Metf). Data from the scans were normalized and analyzed for relative intensity of regulation of genes with metformin treatment. (A) Hierarchical clustering of human endothelial cells from the three different donors shows clear division into control, 6 and 24h metformin treatment, genes showing statistically significant regulation in all donors are shown. The genes are as follows: VEGF-A; PTGS2 (COX2); F3 (coagulation factor III, thromboplastin); ERG (ETS-related gene); FLT1 (VEGF receptor 1); SAT1 (spermidine/spermine N1-acetyltransferase 1); PRKD1 (protein kinase D1) CYP1B1 (cytochrome P450, family 1, subfamily B, polypeptide 1); WARS (tryptophanyl-tRNA synthetase); ADAMTS1; CXCR4; and CCL2 (CC chemokine ligand 2). (B) Real-time PCR validation of gene expression of the statistically significant angiogenesis-related genes up or downregulated by metformin in microarrays. Relative expression values are indicated as fold change over untreated HUVE cells (=1) assessed after normalization on glyceraldehyde 3-phosphate dehydrogenase, RNA polymerase II and glucose-6-phosphate dehydrogenase expression data obtained from reactions run in parallel. All amplifications were performed in triplicate. (C) Analysis of CXCR4, CYP1B1, COX2, VEGF-A and ADAMTS1 protein expression on HUVE cells, by western blotting. A table summarizing protein changes relative to control (Ctrl) is also shown (upregulated proteins are shown in red; downregulated proteins are shown in green). β-Actin was used as loading control.
Fig. 3.
Fig. 3.
Tumor cell supernatants upregulate CYP1B1 expression in HUVE cells, metformin abrogates this effect. HUVE cells were incubated with serum-free unconditioned medium (NC) or conditioned supernatants (conditioned medium, CM) from tumor cells (MDA-MB-231 for breast cancer; PC3 and DU-145 for prostate cancer), all harvested after 24h. (A) Western blotting shows a significant upregulation of CYP1B1 in HUVE cells cultured in presence of conditioned supernatants as compared with those treated with unconditioned medium. Addition of metformin significantly abrogates this effect. (B) HUVE cells were transfected with an AMPKα-specific or control (SiCTR) siRNAs, treated with or without 10mM metformin, lysed and analyzed by western blotting for CYP1B1 expression. CYP1B1 is significantly downregulated in AMPKα siRNA-transfected cells as compared with control siRNA. Metformin upregulates CYP1B1 levels in AMPKα siRNA-transfected HUVE cells, suggesting AMPK-dependent CYP1B1 expression in these cells. β-Actin was used as a loading control. The values in the western blot panels represented the ratio of the indicated protein and actin.
Fig. 4.
Fig. 4.
Expression of angiogenesis-related proteins in HUVE and breast cancer cells treated with metformin. HUVE and MCF-7 (breast cancer) cells were treated with vehicle or 10mM metformin for 24h. Cell culture medium were collected, centrifuged and applied to a human angiogenesis antibody array as described in Materials and methods section. Control proteins are located in duplicate in three corners of the arrays, whereas negative control is indicated as an empty box (lower right corner) in the array. The experiment was repeated in duplicate cell lysates. Modulated proteins in treated cells as compared with control are highlighted with squares and indicated by numbers. Each spot is spotted on the array membrane in duplicate. (A) Metformin modulates 13 angiogenesis-related proteins in HUVE cells. (B) Relative amounts of proteins in selected spots indicated in (A) showing mean and standard error of the three experiments in duplicate. (C) In MCF-7, metformin downregulates 19 angiogenesis protein with either antiangiogenic or pro-angiogenic function. (D) Relative amounts of proteins in selected spots indicated in (B) showing mean and standard error of the duplicate. (E) When we compare the modulation of several factors by metformin in the two cell types, we observed different effects. The phenomenon is summarized in the table: in green, downregulated protein; in red: upregulated protein; in white: no significant modulation.
Fig. 5.
Fig. 5.
Metformin inhibits VEGF-induced ERK1/2 activation in an AMPK-dependent manner. HUVE cells were plated onto six-well plates, treated with or without VEGF in presence or absence of 10mM metformin and/or 10 µM compound C and analyzed at different time points by western blotting. ERK1/2 phosphorylation was measured at 5, 15, 30min or 6h after treatment. (A) ERK1/2 activation was reduced in metformin-treated cells, starting at 15min after metformin addition to the medium. (B) Metformin inhibited phosphorylation of ERK1/2 and inhibited VEGF-induced ERK1/2 phosphorylation. When compound C (cC) was added to the medium, the effects of metformin inhibition and induction of phosphorylation by VEGF alone were counteracted suggesting an involvement of AMPK in ERK1/2 signaling. The experiments were performed twice in triplicate with similar results. The values in the western blot panels represented the ratio of the indicated protein and actin; β-actin was used as loading control.
Fig. 6.
Fig. 6.
Metformin inhibits angiogenesis in vivo in normal and obese mice. A cocktail of pro-angiogenic factors (VEGF-A, TNFα and heparin), either alone (Ctrl) or in combination with different concentrations of metformin as indicated was added into matrigel and injected subcutaneously into C57/BL6 mice. (A) Measurement of the hemoglobin content of the matrigel sponges (*P < 0.05, Mann–Whitney). Inset: matrigel pellets photographed 4 days postinjection. Six-week-old C57 mice were fed with either a control ND or a HFD. After 30 days of the respective diet, mice then received the same diet with metformin (0.5mg metformin/ml drinking water, leading to 2mg metformin/mouse/day) or without (control vehicle in drinking water) for further 60 consecutive days (n = 10 per study arm). On day 90, mice were killed. (B) Measurement of the weight of the WAT shows that the mice on the HFD were clearly obese (P < 0.0001, Mann–Whitney). Mice fed with ND or HFD treated with metformin or vehicle were injected with matrigel as described. (C) Immunofluorescence analysis of vascularization by staining matrigel plug sections with anti-CD31 antibody for endothelial cells in mice either on ND or HFD, with or without oral metformin. (D) Quantification of MVD in the sponges based on CD31 staining (shown as CD31-positive cells/field normalized to control). HFD increased the number of CD31-positive cells as compared with ND, whereas metformin abrogated this effect. (E and F) Blood and visceral WAT were collected for the enumeration of CD45−Sca1+CD34+CD31+ progenitor cells by flow cytometry as described previously (29). (E) Obesity was associated with higher numbers of CD45−Sca1+CD34+CD31+ progenitor cells in the WAT (P = 0.0009, Mann–Whitney). Metformin treatment of the obese mice drastically lowered the CD45−Sca1+CD34+CD31+ progenitor cells in the WAT (P < 0.0001, Mann–Whitney), yet had no effect on the numbers of these cells in mice on a ND. (F) Obesity and/or metformin treatment did not significantly alter the number of CD45−Sca1+CD34+CD31+ progenitor cells in the peripheral blood.

References

    1. Evans J.M., et al. (2005). Metformin and reduced risk of cancer in diabetic patients. BMJ, 330, 1304–1305
    1. Zhang P., et al. (2013). Association of metformin use with cancer incidence and mortality:a meta-analysis. Cancer Epidemiol., 37, 207–218
    1. Ben Sahra I., et al. (2008). The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene, 27, 3576–3586
    1. Phoenix K.N., et al. (2009). Therapeutic metformin/AMPK activation promotes the angiogenic phenotype in the ERalpha negative MDA-MB-435 breast cancer model. Breast Cancer Res. Treat., 113, 101–111
    1. Akinyeke T., et al. (2013) Metformin targets c-MYC oncogene to prevent prostate cancer. Carcinogenesis, 34, 2823–2832
    1. Eid A.A., et al. (2010) AMP-activated protein kinase (AMPK) negatively regulates Nox4-dependent activation of p53 and epithelial cell apoptosis in diabetes. J. Biol. Chem., 285, 37503–37512
    1. Tan B.K., et al. (2009). Metformin decreases angiogenesis via NF-kappaB and Erk1/2/Erk5 pathways by increasing the antiangiogenic thrombospondin-1. Cardiovasc. Res., 83, 566–574
    1. Xavier D.O., et al. (2010). Metformin inhibits inflammatory angiogenesis in a murine sponge model. Biomed. Pharmacother., 64, 220–225
    1. Esfahanian N., et al. (2012). Effect of metformin on the proliferation, migration, and MMP-2 and -9 expression of human umbilical vein endothelial cells. Mol. Med. Rep., 5, 1068–1074
    1. Albini A., et al. (2012). Cancer prevention by targeting angiogenesis. Nat. Rev. Clin. Oncol., 9, 498–509
    1. Chung A.S., et al. (2010). Targeting the tumour vasculature: insights from physiological angiogenesis. Nat. Rev. Cancer, 10, 505–514
    1. Mann J.R., et al. (2005). Mechanisms of disease:inflammatory mediators and cancer prevention. Nat. Clin. Pract. Oncol., 2, 202–210
    1. Albini A., et al. (2007). The tumour microenvironment as a target for chemoprevention. Nat. Rev. Cancer, 7, 139–147
    1. Ersoy C., et al. (2008). The effect of metformin treatment on VEGF and PAI-1 levels in obese type 2 diabetic patients. Diabetes Res. Clin. Pract., 81, 56–60
    1. Lund S.S., et al. (2008). Impact of metformin versus repaglinide on non-glycaemic cardiovascular risk markers related to inflammation and endothelial dysfunction in non-obese patients with type 2 diabetes. Eur. J. Endocrinol., 158, 631–641
    1. Hirsch H.A., et al. (2013). Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc. Natl Acad. Sci. USA, 110, 972–977
    1. Caballero A.E., et al. (2004). The differential effects of metformin on markers of endothelial activation and inflammation in subjects with impaired glucose tolerance: a placebo-controlled, randomized clinical trial. J. Clin. Endocrinol. Metab., 89, 3943–3948
    1. Abbasi F., et al. (2004). Effect of metformin treatment on multiple cardiovascular disease risk factors in patients with type 2 diabetes mellitus. Metabolism., 53, 159–164
    1. Cacicedo J.M., et al. (2004). AMPK inhibits fatty acid-induced increases in NF-kappaB transactivation in cultured human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun., 324, 1204–1209
    1. De Jager J., et al. (2005). Effects of short-term treatment with metformin on markers of endothelial function and inflammatory activity in type 2 diabetes mellitus: a randomized, placebo-controlled trial. J. Intern. Med., 257, 100–109
    1. Hadad S.M., et al. (2009). Therapeutic metformin/AMPK activation promotes the angiogenic phenotype in the ERalpha negative MDA-MB-435 breast cancer model. Breast Cancer Res. Treat., 114, 391.
    1. Martin M.J., et al. (2012). Metformin accelerates the growth of BRAF V600E-driven melanoma by upregulating VEGF-A. Cancer Discov., 2, 344–355
    1. Cittadini A., et al. (2012). Metformin prevents the development of chronic heart failure in the SHHF rat model. Diabetes, 61, 944–953
    1. Sena C.M., et al. (2011). Metformin restores endothelial function in aorta of diabetic rats. Br. J. Pharmacol., 163, 424–437
    1. Ziche M., et al. (2009). Molecular regulation of tumour angiogenesis by nitric oxide. Eur. Cytokine Netw., 20, 164–170
    1. Carpentier G. (2012). Contribution:angiogenesis analyzer. ImageJ News, 5 October 2012. (21 October 2013, date last accessed).
    1. Passaniti A., et al. (1992). A simple, quantitative method for assessing angiogenesis and antiangiogenic agents using reconstituted basement membrane, heparin, and fibroblast growth factor. Lab. Invest., 67, 519–528
    1. Balwierz A., et al. (2009). Angiogenesis in the New Zealand obese mouse model fed with high fat diet. Lipids Health Dis., 8, 13.
    1. Martin-Padura I., et al. (2012). The white adipose tissue used in lipotransfer procedures is a rich reservoir of CD34+ progenitors able to promote cancer progression. Cancer Res., 72, 325–334
    1. Mancuso P., et al. (2011). Circulating perivascular progenitors: a target of PDGFR inhibition. Int. J. Cancer, 129, 1344–1350
    1. Dagher Z., et al. (1999). The effect of AMP-activated protein kinase and its activator AICAR on the metabolism of human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun., 265, 112–115
    1. Ido Y., et al. (2002). Hyperglycemia-induced apoptosis in human umbilical vein endothelial cells: inhibition by the AMP-activated protein kinase activation. Diabetes, 51, 159–167
    1. Nagata D., et al. (2003) AMP-activated protein kinase (AMPK) signaling in endothelial cells is essential for angiogenesis in response to hypoxic stress. J. Biol. Chem., 278, 31000–31006
    1. Ouchi N., et al. (2004) Adiponectin stimulates angiogenesis by promoting cross-talk between AMP-activated protein kinase and Akt signaling in endothelial cells. J. Biol. Chem., 279, 1304–1309
    1. Shibata R., et al. (2004) Adiponectin stimulates angiogenesis in response to tissue ischemia through stimulation of AMP-activated protein kinase signaling. J. Biol. Chem., 279, 28670–28674
    1. Hattori Y., et al. (2006). Metformin inhibits cytokine-induced nuclear factor kappaB activation via AMP-activated protein kinase activation in vascular endothelial cells. Hypertension, 47, 1183–1188
    1. Baud V., et al. (2009) Is NF-kappaB a good target for cancer therapy? Hopes and pitfalls. Nat. Rev. Drug Discov., 8, 33–40
    1. Jalving M., et al. (2010). Metformin: taking away the candy for cancer? Eur. J. Cancer, 46, 2369–2380
    1. Wong C., et al. (2005) Protein kinase C-dependent protein kinase D activation modulates ERK signal pathway and endothelial cell proliferation by vascular endothelial growth factor. J. Biol. Chem., 280, 33262–33269
    1. To K.K., et al. (2006). The phosphorylation status of PAS-B distinguishes HIF-1alpha from HIF-2alpha in NBS1 repression. EMBO J., 25, 4784–4794
    1. Teicher B.A., et al. (2010). CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin. Cancer Res., 16, 2927–2931
    1. Tang Y., et al. (2010). CYP1B1 and endothelial nitric oxide synthase combine to sustain proangiogenic functions of endothelial cells under hyperoxic stress. Am. J. Physiol. Cell Physiol., 298, C665–C678
    1. Tang Y., et al. (2009). CYP1B1 expression promotes the proangiogenic phenotype of endothelium through decreased intracellular oxidative stress and thrombospondin-2 expression. Blood, 113, 744–754
    1. Palomba S., et al. (2013). Effects of metformin in women with polycystic ovary syndrome treated with gonadotrophins for in vitro fertilisation and intracytoplasmic sperm injection cycles: a systematic review and meta-analysis of randomised controlled trials. BJOG, 120, 267–276
    1. Rattan R., et al. (2011). Metformin suppresses ovarian cancer growth and metastasis with enhancement of cisplatin cytotoxicity in vivo . Neoplasia, 13, 483–491
    1. Liao H., et al. (2012). Luteinizing hormone facilitates angiogenesis in ovarian epithelial tumor cells and metformin inhibits the effect through the mTOR signaling pathway. Oncol. Rep., 27, 1873–1878
    1. Obika M., et al. (2012). Tumor growth inhibitory effect of ADAMTS1 is accompanied by the inhibition of tumor angiogenesis. Cancer Sci., 103, 1889–1897
    1. Ware W.R. (2009). Nutrition and the prevention and treatment of cancer: association of cytochrome P450 CYP1B1 with the role of fruit and fruit extracts. Integr. Cancer Ther., 8, 22–28
    1. Luby T.M. (2008). Targeting cytochrome P450 CYP1B1 with a therapeutic cancer vaccine. Expert Rev. Vaccines, 7, 995–1003
    1. Conway D.E., et al. (2009). Expression of CYP1A1 and CYP1B1 in human endothelial cells: regulation by fluid shear stress. Cardiovasc. Res., 81, 669–677
    1. Weis S.M., et al. (2011). Tumor angiogenesis: molecular pathways and therapeutic targets. Nat. Med., 17, 1359–1370
    1. Mikstacka R., et al. (2007). Inhibition of human recombinant cytochromes P450 CYP1A1 and CYP1B1 by trans-resveratrol methyl ethers. Mol. Nutr. Food Res., 51, 517–524
    1. Guedez L., et al. (2001). Tissue inhibitor of metalloproteinase-1 alters the tumorigenicity of Burkitt’s lymphoma via divergent effects on tumor growth and angiogenesis. Am. J. Pathol., 158, 1207–1215
    1. Gou S., et al. (2013). Low concentrations of metformin selectively inhibit CD133⁺ cell proliferation in pancreatic cancer and have anticancer action. PLoS One, 8, e63969.
    1. Ye J., et al. (2013). Inflammation during obesity is not all bad: evidence from animal and human studies. Am. J. Physiol. Endocrinol. Metab., 304, E466–E477

Source: PubMed

3
Tilaa