Lipogenesis in cancer progression (Review)

  • Authors:
    • Catherine Mounier
    • Lamia Bouraoui
    • Eric Rassart
  • View Affiliations

  • Published online on: May 13, 2014     https://doi.org/10.3892/ijo.2014.2441
  • Pages: 485-492
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In normal tissues, energy-providing lipids come principally from circulating lipids. However, in growing tumors, energy supply is mainly provided by lipids coming from de novo synthesis. It is not surprising to see elevated expression of several lipogenic genes in tumors from different origins. The role of lipogenic genes in the establishment of the primary tumor has been clearly established. A large number of studies demonstrate a role of fatty acid synthase in the activation of cell cycle and inhibition of apoptosis in tumor cells. Other lipogenic genes such as the acetyl CoA carboxylase (ACC) and the stearoyl CoA desaturase 1 (SCD1) are highly expressed in primary tumors and also appear to play a role in their development. However, the role of lipogenesis in the metastatic process is less clear. In the present review, we aim to present the most recent evidences for the key role of lipogenic enzymes in the metastatic process and in epithelial to mesenchymal transition.

1. Introduction

Solid tumors require high levels of energy for growth and membrane synthesis. Lipids provide this energy. In normal tissues lipids come from circulating lipids, while cancer cells mainly use de novo synthesized lipids (1). As a result, the rate of lipogenesis is highly induced (2).

Lipogenesis occurs both in liver and adipose tissues resulting in the synthesis of de novo fatty acids from acetyl CoA synthesized by glycolysis (Fig. 1). Acetyl CoA is then carboxylated by ACC forming malonyl CoA. Malonyl CoA and acetyl CoA are further processed by fatty acid synthase (FAS) in palmitic acid, which is then transformed by Elvol6 into stearic acid (3). SCD1 catalyzes the formation of palmitoleoyl-CoA and oleoyl-CoA from palmitoyl-CoA and stearoyl-CoA, respectively (4), which are preferentially transformed in triglycerides for storage in adipose tissue or phospholipids for membrane formation (5).

Expressions of ACC, FAS and SCD1 are under the control of the transcription factors LXR and the sterol regulatory element-binding protein-1c (SREBP-1c) (3). The 5′ AMP-activated protein kinase (AMPK) has been implicated in the control of hepatic lipogenesis (6) through inactivation of ACC (7) and SREBP-1c (6). ACC and FAS are overexpressed in numerous types of cancers (2,8) while high levels of mono unsaturated fatty acids (MUFA) were found in tumors (9) as a result of increased SCD1 expression and activity. SREBP1 has also been implicated in tumor growth (10). Therefore, high rate of lipogenesis is probably associated with tumorogenesis.

High lipogenic activity was also associated with cancer progression and metastasis (11). As suggested, high SREBP1 expression may explain at least in part the increased expression of lipogenic genes associated with stratification of the malignancy. However, independently of SREBP1, the lipogenic enzymes expression correlates with the state of malignancy. The most recent findings on the role of lipogenesis in cancer progression and metastatic process are presented.

2. Lipogenesis, tumor growth and apoptosis

An elevated FAS expression induces progression of cancer cells into S phase (12). In contrast, inhibition of FAS expression decreases tumor growth and it induces apoptosis of cancer cells (8,13). Elevated ACC expression is observed in the early stage of breast cancer (14) and silencing its expression results in growth inhibition and apoptosis of cancer cells (1517). High SCD1 expression is associated with cancer cell proliferation (18) and with a decrease in cell death (1821). SREBP1c also plays a role in the transformation of normal cells (22).

Lipogenesis is induced in cancer cells by EGF through activation of the HER2/neu receptor (23) and the PI3-kinase/Akt pathway (2426) targeting SREBP-1 (24,27). As AMPK inhibits ACC by phosphorylation, its inactivation diminishes lipid supply and blocks cell cycle decreasing cell division and tumor growth (28). Therefore, lipogenesis probably provides energy supply to cancer cells stimulating cell division and survival leading to tumor growth.

3. The metastatic process

Metastasis is a complex multi-step process. Tumor cells need to escape from the primary tumor and to enter in the blood or in the lymphatic system. Most of the circulating cells undergo apoptosis (29) but some of them survive and invade new tissues (30). Epithelial to mesenchymal transition (EMT) has been associated with tumor progression and metastasis (31,32). It dissolves tight junctions between epithelial cells, the extracellular matrix and the adherent basolateral junctions leading to a disorganized and mobile mesenchymal cell population (32).

One of the first events of EMT is the loss of E-cadherin, a transmembrane protein implicated in formation of the tight junctions (33). This is the result of increased expression of its transcriptional repressors (34,35). In normal conditions, GSK3β phosphorylates E-cadherin transcriptional repressors targeting them to proteasome degradation thus allowing transcription of E-cadherin (36) (Fig. 2). In cancers, activation of TGF-β, Wnt, RTK and integrin pathways (37,38) leads to inhibition of GSK3β (39). Consequently, E-cadherin transcriptional repressors are no longer phosphorylated nor degraded leading to inhibition of E-cadherin expression (40). Another consequence is the loss of β-catenin phosphorylation that cannot be targeted to the proteasome and accumulates in the cytosol. It further translocates to the nucleus where it activates the transcription of genes such as c-myc, an important cell cycle regulator (41,42).

E-cadherin is also implicated in the actin cytoskeleton organization. Its direct binding to actin filament or to β-catenin maintains cell polarity and tissue architecture (43). In cancer cells, once the E-cadherin/β-catenin complexes disappear, the actin network is disrupted modifying cell migration (44,45). Nuclear β-catenin will also increase expression of mesenchymal proteins (46,47). These molecular events will allow EMT inducing the migration and invasion of cancer cells and metastasis.

4. Implication of fatty acid synthase

Among the lipogenic enzymes implicated in the development of metastasis, FAS is certainly the most studied protein (Table I). Prognostic and survival of patients with cancer are mainly predicted by the presence of metastasis (48) and overexpression of FAS has been associated with poor prognostics in several hormone-dependent cancers (4952). The direct association between FAS expression and metastasis has also been observed in prostate cancers (53) and breast carcinomas (49).

Table I

Evidence for a role of fatty acid synthase in the development of metastasis in various types of cancer. These studies were performed in patients, in mice and transformed cancer cell lines.

Table I

Evidence for a role of fatty acid synthase in the development of metastasis in various types of cancer. These studies were performed in patients, in mice and transformed cancer cell lines.

RoleType of cancerSystemImplicated mechanismsRefs.
Synthesis of palmitateOvarian neoplasmsPatientsOestrogen(52)
A2780 and SKOV3 cellsAkt-USP2a(25,60)
Breast carcinomasPatientsEGF-oestrogen(23,48,49,51)
MDA-MB-231, MCF-10A, BT474, and SKBr3 cellsHER2/neu(23)
MCF7 cellsHER2/neu(62)
SKRB3 cellsLipid rafts(64)
PI3K/Akt and ERK1/2
Endometrial carcinomasPatientsOestrogen(51)
Prostate cancersPatientsAndrogen(50)
Mouse TRAMPAndrogen(53,54)
iPrECs cellsAndrogen(55)
LNCaP/Androgen
MDA-PCa-2a cellsSREBP1/SCAP(57)
PI3K/Akt pathway(58)
USP2a(59)
Renal cancersPatients(73)
Caki-1 cellsHER2/neu-STAT3(13)
Renca cells
Head and neck neoplasmsPatientsEGFR(71)
Lung carcinomasPatients(74)
Colon cancersMice xenograph modelsMET-AKT-FAK(76,77)
MelanomasMelanoma cell line B16-F10(75)
RetinoblastomasPatients(69)

In the transgenic adenocarcinoma of the mouse prostate (TRAMP) model which closely mirrors the progression of prostate cancer observed in human, FAS expression and activity are high compared to control littermates (54). Injection of immunodeficient mice with human prostate cancer cells overexpressing FAS and the androgen receptor (AR) leads to invasive adenocarcinomas (55). Androgens stimulate FAS expression in prostate cancers (55) by increasing the nuclear levels of SREBP (57). This is probably the result of increased SCAP expression that exports SREBP from the endoplasmic reticulum to the Golgi where it is activated by cleavage. As a consequence of androgen action on SREBP, expression of lipogenic genes is increased (56,57). Downstream of the AR, the PI3KAkt pathway has been implicated in FAS activation (58). In prostate cancer, the isopeptidase USP2a has been also implicated in the activation of FAS expression by inhibiting its proteosomal degradation (59).

In ovarian cancer cells, proteolysis degradation of FAS and focal adhesion kinase (FAK) cause a strong reduction of the vascular endothelial growth factor (VEGF)-mediated cell migration and invasion (60), in the same study, the isopeptidase USP2a was also shown to stabilize FAS. In breast cancer cells, the green tea extract EGCg causes accumulation of β-catenin in the cytosol and decreased expression of E-cadherin (61). EGCg appears to disturb cell adhesion by modifying FAS and the EGF receptor (EGFR) signaling pathway.

FAS has also been implicated in the transformation of breast cancer cells through an effect on the EGFR (HER2/neu isoform) expression (62). HER2/neu is a proto-oncogene associated with the development of metastasis in breast cancer (63). In HER2-positive cells, elevated FAS expression stabilizes the lipid rafts and consequently, HER2/neu expression is increased activating downstream signaling pathways (64). EGF also increases FAS transcription establishing a positive feedback loop between FAS and EGF (6567).

FAS expression is stimulated by estrogen in both endometrial and breast cancer cells (51). However, this increase is probably associated with the establishment of the primary tumor as the presence of estrogen and progesterone receptors in tumors provide better prognostic for the patients than those expressing HER2/neu (63,68).

Correlative associations between FAS expression and poor prognosis for patients were also observed in non-hormone-dependent cancers (6971) as well as with metastasis (72).

In metastatic renal cancer, FAS expression is strongly induced compared to non-transformed tumors (73). In human pancreatic cells, invasiveness was abolished by C75, a synthetic FAS inhibitor possibly through downregulation of HER2/neu and/or STAT3 phosphorylation (72). In a mouse model of spontaneous melanoma metastasis, direct IP injection of Orlistat, a natural FAS inhibitor, inhibits metastasis in lymph nodes by more than 50% (75). In xenograft models of advanced colon cancer, inhibition of FAS decreased hepatic metastasis (76,77) implicating AKT downstream of FAS. Inhibition of FAS also attenuates the activation of the MET receptor and FAK, two proteins implicated in adhesion, migration and invasion of cancer cells (60).

The above studies point to a key role of FAS in cancer progression, probably through modulation of lipid raft formation leading to activation of EGFR, HER2/neu and MET. Consequently, downstream signaling pathways are activated increasing nuclear localization of SREBP1c activating FAS and other lipogenic genes describing a positive feedback loop.

5. Implication of stearoyl CoA desaturase 1

An increased content of MUFAs has been observed in transformed cells suggesting a role for SCD1 in tumorigenesis (9). The fatty acid profile and particularly the balance between saturated fatty acids (SFA) and MUFA can be used as predictor for breast cancer (7880). It was recently demonstrated that silencing of SCD1 in breast cancer cells does not affect cell viability but inhibits cell cycle progression (81). In these conditions, expressions of key proteins involved in cell cycle progression are decreased. The degree of SCD1 inhibition appears directly correlated with inhibition of cancer cells proliferation (19) decreasing the amount of SFA (SCD1 substrates), the main inhibitors of ACC (82).

Others studies point for a role of SCD1 in cancer progression and metastasis (Table II). It was shown that MUFA content in cholesterol esters is associated with higher death rate in cancer patients (83) while elevated levels of oleic acid were observed in breast cancers with metastasis (84), suggesting an increased activity of SCD1. Low content of stearic acid (SCD1 substrate) in phosphatodylcholine were also measured in breast tumors associated with subsequent metastasis (9). In breast adipose tissues, no difference in MUFA content was observed between benign tumors and normal tissues, but a positive correlation was observed between MUFA concentration and metastasis (85). Alteration of SFA/MUFA ratio in breast tumors does not reflect the dietary intake of patients but rather the change in fatty acid metabolism in cells (86) underlying a role for de novo synthesized MUFA and SCD1.

Table II

Role of SCD1 in metastatic cancers.

Table II

Role of SCD1 in metastatic cancers.

RoleType of cancerSystemImplicated mechanismsRefs.
Fatty acid desaturationGeneralPatient(83)
Formation of palmitoleate and oleateBreast cancersPatients(9,7880,8386)
MCF-7ACC/AMPK(19)
MDA-MB-231 GSK3β/β-catenin(81)
MDA-MB-435, MDA-MB-468, SKBR3, BT-474(11)
Lung cancerSV40-WI38PPARγ(18)
A549AMPK/ACC(19)
AKT/GSK3β(20)

In lung adenocarcinomas, SCD1 knockdown inhibits AKT phosphorylation and activity (20) known to be associated with cancer progression (87). Silencing SCD1 in SV40-transformed lung fibroblasts and in breast cancer cells inhibits GSK3β phosphorylation (20).

Consequently, nuclear β-catenin translocation is decreased leading to lower expression of cyclin D1 and vimentin, two proteins associated with a mesenchymal phenotype (88). Silencing SCD1 in MCF7 and MDA-MB-231 breast cancer cells also increased E-cadherin expression associated with changes in cellular morphological aspects and decreased migration (81). It was also shown that palmitoleic acid (SCD1 product) is required to modify Wnt proteins leading to activation of the Wnt signaling pathway (89).

In breast cancer cells, we observed that the induction of β-catenin nuclear translocation by TGFβ is abrogated upon SCD1 silencing (Mounier et al, unpublished data). TGFβ acts as a tumor suppressor, but when cells become resistant to its action, it acts as a potent stimulator of malignant conversion (32). TGF-β activates SCD1 expression through a Smad-dependent pathway (88). Constitutive activation of the EGF signaling pathways through the ErbB receptors has been associated with metastasis and poor prognostic for patients (91). Paradoxally, incubation of breast cancer cells with oleic acid inhibits the expression of HER2/neu suggesting an anti-metastatic effect of the product of SCD1 (92).

The role of SCD1 in EMT probably involves GSK3β activation and downstream cellular events modifying cell adhesion and migration. Certain evidence also point for a role of TGFβ and EGF in mediating SCD1 expression in metastasic cancer cells.

6. Implication of other lipogenic genes

General modification of the lipid profile during cancer progression is associated with increased expression of several genes involved in lipid metabolism (84). Apart from FAS and SCD1, the expression of other genes was modulated such as ACC, INSIG1 (insulin-induced gene 1), SCAP (sterol regulatory element-binding protein cleavage-activating protein) and THRSP (thyroid hormone-responsive protein).

THRSP, also known as Spot14, is a nuclear protein that activates lipogenic genes (93). Low Spot14 expression was associated with prolonged survival in invasive breast cancers suggesting that Spot14 may not be a key player in EMT (93). Another study suggests that as breast cancer cells do not express lipoprotein lipase, lipids must be provided by a local environment such as breast lipids explaining why cancer cells with low Spot14 levels cannot survive in a low lipids concentration environment such as lymph nodes (94). The authors even suggest that elevated expression of Spot14 in cancer cells may provide a unique explanation for the elevated lipid synthesis in cancer cells.

Elevated ACC expression was also associated with a higher risk of infiltration in breast cancer (49). Amplification in ACC gene copy number was observed in breast cancer patients with reduced survival (95). Mutations in BCRA1, a gene associated with predisposition of inherited cancer, disrupt BCRA1 interaction with the inactive phosphorylated ACC. Consequently, ACC is dephosphorylated and activated (96). AMPK that phosphorylates ACC was also associated with malignancy providing energy for cancer cells (28). Adiponectin, an adipocytokine described as an anti-metastatic agent, inhibits ACC by increasing AMPK activity (97). In breast cancer cells, ACC is also regulated by a ubiquitin-dependent degradation process through its interaction with AKR1B10 (aldoketo reductase family 1 B10) (14). Pharmaceutical inhibition of ACC in cancer cells inhibits invadopodia formation, a membrane protrusion that facilitates matrix degradation and cellular invasion (98) but SCD1 is not required for invadopodia formation suggesting a different pathway. However, malonyl CoA decreases expression of the HER2/neu gene suggesting an anti-metastasic effect of ACC (99). This emphasizes the role of FAS in EMT as FAS decreases malonyl CoA content in cells.

Prostate cancer development and progression is often dependent of androgen and evidence indicates that androgen activation of the SREBP-dependent pathway may explain most of the androgen effects on lipogenesis (100). Androgen activates the cleavage of SREBP by increasing the expression of SCAP and INSIG (57). SREBP1 also increases reactive oxygen species (ROS) production (101) inducing tumor progression (102). A similar role of progesterone and EGF on SREBP cleavage and expression was reported in breast cancer cells (22,65,67).

7. Conclusions

Increased lipogenesis is an important hallmark of cancer progression and metastasis. Part of this effect is probably mediated by SREBP1. However, evidence points to a role of lipogenic enzymes independent of SREBP1. ACC activity is regulated in cancer cells through phosphorylation by AMPK or by interaction with BCRA1. However, malonyl CoA, the product of ACC, has an anti-metastatic effect suggesting a more direct role of FAS on EMT. As such, a role for palmitate in the formation of membranes and rafts was suggested. Rafts allow recruitment and stabilization of receptors such as EGF, HER2/neu and MET increasing cancer progression. In contrast to FAS, SCD1 is not involved in the formation of invadopodia suggesting that if both enzymes are involved in EMT, they probably act through different mechanisms. Increased SCD1 activity decreases the level of SFA, the main ACC inhibitors activating lipogenesis in cancer cells. SCD1 is also probably directly involved in EMT. FAS and SCD1 may be the most interesting targets for the treatment of metastasic cancers, and pharmaceutical inhibitors already exist that could be used readily for the treatment of patients.

References

1 

Medes G, Thomas A and Weinhouse S: Metabolism of neoplastic tissue. IV A study of lipid synthesis in neoplastic tissue slices in vitro. Cancer Res. 13:27–29. 1953.PubMed/NCBI

2 

Swinnen JV, Brusselmans K and Verhoeven G: Increased lipogenesis in cancer cells: new players, novel targets. Curr Opin Clin Nutr Metab Care. 9:358–365. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Postic C and Girard J: The role of the lipogenic pathway in the development of hepatic steatosis. Diabetes Metab. 34:643–648. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Ntambi JM: The regulation of stearoyl-CoA desaturase (SCD). Prog Lipid Res. 34:139–150. 1995. View Article : Google Scholar : PubMed/NCBI

5 

Neuschwander-Tetri BA: Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology. 52:774–788. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Viollet B, Guigas B, Leclerc J, et al: AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives. Acta Physiol (Oxf). 196:81–98. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Viollet B, Foretz M, Guigas B, et al: Activation of AMP-activated protein kinase in the liver: a new strategy for the management of metabolic hepatic disorders. J Physiol. 574:41–53. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Kuhajda FP: Fatty-acid synthase and human cancer: new perspectives on its role in tumor biology. Nutrition. 16:202–208. 2000. View Article : Google Scholar

9 

Bougnoux P, Chajes V, Lanson M, et al: Prognostic significance of tumor phosphatidylcholine stearic acid level in breast carcinoma. Breast Cancer Res Treat. 20:185–194. 1992. View Article : Google Scholar : PubMed/NCBI

10 

Guo D, Reinitz F, Youssef M, et al: An LXR agonist promotes glioblastoma cell death through inhibition of an EGFR/AKT/SREBP-1/LDLR-dependent pathway. Cancer Discov. 1:442–456. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Nieva C, Marro M, Santana-Codina N, Rao S, Petrov D and Sierra A: The lipid phenotype of breast cancer cells characterized by Raman microspectroscopy: towards a stratification of malignancy. PLoS One. 7:e464562012. View Article : Google Scholar

12 

Pizer ES, Chrest FJ, DiGiuseppe JA and Han WF: Pharmacological inhibitors of mammalian fatty acid synthase suppress DNA replication and induce apoptosis in tumor cell lines. Cancer Res. 58:4611–4615. 1998.PubMed/NCBI

13 

Horiguchi A, Asano T, Asano T, Ito K, Sumitomo M and Hayakawa M: Pharmacological inhibitor of fatty acid synthase suppresses growth and invasiveness of renal cancer cells. J Urol. 180:729–736. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Ma J, Yan R, Zu X, et al: Aldo-keto reductase family 1 B10 affects fatty acid synthesis by regulating the stability of acetyl-CoA carboxylase-alpha in breast cancer cells. J Biol Chem. 283:3418–3423. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Chajes V, Cambot M, Moreau K, Lenoir GM and Joulin V: Acetyl-CoA carboxylase alpha is essential to breast cancer cell survival. Cancer Res. 66:5287–5294. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Brusselmans K, De Schrijver E, Verhoeven G and Swinnen JV: RNA interference-mediated silencing of the acetyl-CoA-carboxylase-alpha gene induces growth inhibition and apoptosis of prostate cancer cells. Cancer Res. 65:6719–6725. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Zhan Y, Ginanni N, Tota MR, et al: Control of cell growth and survival by enzymes of the fatty acid synthesis pathway in HCT-116 colon cancer cells. Clin Cancer Res. 14:5735–5742. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Scaglia N, Caviglia JM and Igal RA: High stearoyl-CoA desaturase protein and activity levels in simian virus 40 transformed-human lung fibroblasts. Biochim Biophys Acta. 1687:141–151. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Scaglia N, Chisholm JW and Igal RA: Inhibition of stearoylCoA desaturase-1 inactivates acetyl-CoA carboxylase and impairs proliferation in cancer cells: role of AMPK. PloS One. 4:e68122009. View Article : Google Scholar : PubMed/NCBI

20 

Scaglia N and Igal RA: Inhibition of stearoyl-CoA desaturase 1 expression in human lung adenocarcinoma cells impairs tumorigenesis. Int J Oncol. 33:839–850. 2008.PubMed/NCBI

21 

Morgan-Lappe SE, Tucker LA, Huang X, et al: Identification of Ras-related nuclear protein, targeting protein for xenopus kinesin-like protein 2, and stearoyl-CoA desaturase 1 as promising cancer targets from an RNAi-based screen. Cancer Res. 67:4390–4398. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Guo D, Prins RM, Dang J, et al: EGFR signaling through an Akt-SREBP-1-dependent, rapamycin-resistant pathway sensitizes glioblastomas to antilipogenic therapy. Sci Signal. 2:ra822009.PubMed/NCBI

23 

Zhang D, Tai LK, Wong LL, Chiu LL, Sethi SK and Koay ES: Proteomic study reveals that proteins involved in metabolic and detoxification pathways are highly expressed in HER-2/neu-positive breast cancer. Mol Cell Proteomics. 4:1686–1696. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Porstmann T, Griffiths B, Chung YL, et al: PKB/Akt induces transcription of enzymes involved in cholesterol and fatty acid biosynthesis via activation of SREBP. Oncogene. 24:6465–6481. 2005.PubMed/NCBI

25 

Wang HQ, Altomare DA, Skele KL, et al: Positive feedback regulation between AKT activation and fatty acid synthase expression in ovarian carcinoma cells. Oncogene. 24:3574–3582. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Bandyopadhyay S, Pai SK, Watabe M, et al: FAS expression inversely correlates with PTEN level in prostate cancer and a PI 3-kinase inhibitor synergizes with FAS siRNA to induce apoptosis. Oncogene. 24:5389–5395. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Chang Y, Wang J, Lu X, Thewke DP and Mason RJ: KGF induces lipogenic genes through a PI3K and JNK/SREBP-1 pathway in H292 cells. J Lipid Res. 46:2624–2635. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Swinnen JV, Beckers A, Brusselmans K, et al: Mimicry of a cellular low energy status blocks tumor cell anabolism and suppresses the malignant phenotype. Cancer Res. 65:2441–2448. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Mehes G, Witt A, Kubista E and Ambros PF: Circulating breast cancer cells are frequently apoptotic. Am J Pathol. 159:17–20. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Iwatsuki M, Mimori K, Yokobori T, et al: Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci. 101:293–299. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Polyak K and Weinberg RA: Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat Rev Cancer. 9:265–273. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Voulgari A and Pintzas A: Epithelial-mesenchymal transition in cancer metastasis: mechanisms, markers and strategies to overcome drug resistance in the clinic. Biochim Biophys Acta. 1796:75–90. 2009.PubMed/NCBI

34 

Yang J, Mani SA, Donaher JL, et al: Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 117:927–939. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Batlle E, Sancho E, Franci C, et al: The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol. 2:84–89. 2000. View Article : Google Scholar : PubMed/NCBI

36 

Darnell JE Jr: Transcription factors as targets for cancer therapy. Nature reviews Cancer. 2:740–749. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Moustakas A and Heldin CH: Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 98:1512–1520. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Blobe GC, Schiemann WP and Lodish HF: Role of transforming growth factor beta in human disease. N Engl J Med. 342:1350–1358. 2000. View Article : Google Scholar

39 

Stambolic V and Woodgett JR: Mitogen inactivation of glycogen synthase kinase-3 beta in intact cells via serine 9 phosphorylation. Biochem J. 303:701–704. 1994.

40 

Peinado H, Portillo F and Cano A: Transcriptional regulation of cadherins during development and carcinogenesis. Int J Dev Biol. 48:365–375. 2004. View Article : Google Scholar

41 

Hamada F and Bienz M: The APC tumor suppressor binds to C-terminal binding protein to divert nuclear beta-catenin from TCF. Dev Cell. 7:677–685. 2004. View Article : Google Scholar : PubMed/NCBI

42 

He TC, Sparks AB, Rago C, et al: Identification of c-MYC as a target of the APC pathway. Science. 281:1509–1512. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Takeichi M, Nakagawa S, Aono S, Usui T and Uemura T: Patterning of cell assemblies regulated by adhesion receptors of the cadherin superfamily. Philos Trans R Soc Lond B Biol Sci. 355:885–890. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Efstathiou JA and Pignatelli M: Modulation of epithelial cell adhesion in gastrointestinal homeostasis. Am J Pathol. 153:341–347. 1998. View Article : Google Scholar

45 

Wijnhoven BP and Pignatelli M: E-cadherin-catenin: more than a ‘sticky’ molecular complex. Lancet. 354:356–357. 1999.

46 

Gilles C, Polette M, Mestdagt M, et al: Transactivation of vimentin by beta-catenin in human breast cancer cells. Cancer Res. 63:2658–2664. 2003.PubMed/NCBI

47 

Takahashi M, Tsunoda T, Seiki M, Nakamura Y and Furukawa Y: Identification of membrane-type matrix metalloproteinase-1 as a target of the beta-catenin/Tcf4 complex in human colorectal cancers. Oncogene. 21:5861–5867. 2002. View Article : Google Scholar : PubMed/NCBI

48 

Rakha EA: Pitfalls in outcome prediction of breast cancer. J Clin Pathol. 66:458–464. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Milgraum LZ, Witters LA, Pasternack GR and Kuhajda FP: Enzymes of the fatty acid synthesis pathway are highly expressed in in situ breast carcinoma. Clin Cancer Res. 3:2115–2120. 1997.PubMed/NCBI

50 

Epstein JI, Carmichael M and Partin AW: OA-519 (fatty acid synthase) as an independent predictor of pathologic state in adenocarcinoma of the prostate. Urology. 45:81–86. 1995. View Article : Google Scholar : PubMed/NCBI

51 

Lupu R and Menendez JA: Targeting fatty acid synthase in breast and endometrial cancer: An alternative to selective estrogen receptor modulators? Endocrinology. 147:4056–4066. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Davidson B, Smith Y, Nesland JM, Kaern J, Reich R and Trope CG: Defining a prognostic marker panel for patients with ovarian serous carcinoma effusion. Hum Pathol. 44:2449–2460. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Pflug BR, Pecher SM, Brink AW, Nelson JB and Foster BA: Increased fatty acid synthase expression and activity during progression of prostate cancer in the TRAMP model. Prostate. 57:245–254. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Greenberg NM, DeMayo F, Finegold MJ, et al: Prostate cancer in a transgenic mouse. Proc Natl Acad Sci USA. 92:3439–3443. 1995. View Article : Google Scholar : PubMed/NCBI

55 

Migita T, Ruiz S, Fornari A, Fiorentino M, Priolo C, Zadra G, et al: Fatty acid synthase: a metabolic enzyme and candidate oncogene in prostate cancer. J Natl Cancer Inst. 101:519–532. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Swinnen JV, Esquenet M, Goossens K, Heyns W and Verhoeven G: Androgens stimulate fatty acid synthase in the human prostate cancer cell line LNCaP. Cancer Res. 57:1086–1090. 1997.PubMed/NCBI

57 

Heemers H, Maes B, Foufelle F, Heyns W, Verhoeven G and Swinnen JV: Androgens stimulate lipogenic gene expression in prostate cancer cells by activation of the sterol regulatory element-binding protein cleavage activating protein/sterol regulatory element-binding protein pathway. Mol Endocrinol. 15:1817–1828. 2001. View Article : Google Scholar

58 

Van de Sande T, De Schrijver E, Heyns W, Verhoeven G and Swinnen JV: Role of the phosphatidylinositol 3′-kinase/PTEN/Akt kinase pathway in the overexpression of fatty acid synthase in LNCaP prostate cancer cells. Cancer Res. 62:642–646. 2002.

59 

Graner E, Tang D, Rossi S, et al: The isopeptidase USP2a regulates the stability of fatty acid synthase in prostate cancer. Cancer Cell. 5:253–261. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Selvendiran K, Ahmed S, Dayton A, et al: HO-3867, a synthetic compound, inhibits the migration and invasion of ovarian carcinoma cells through downregulation of fatty acid synthase and focal adhesion kinase. Mol Cancer Res. 8:1188–1197. 2010. View Article : Google Scholar

61 

Hsu YC and Liou YM: The anti-cancer effects of (−)-epigallocatechin-3-gallate on the signaling pathways associated with membrane receptors in MCF-7 cells. J Cell Physiol. 226:2721–2730. 2011.

62 

Menendez JA, Vellon L, Mehmi I, et al: Inhibition of fatty acid synthase (FAS) suppresses HER2/neu (erbB-2) oncogene overexpression in cancer cells. Proc Natl Acad Sci USA. 101:10715–10720. 2004. View Article : Google Scholar : PubMed/NCBI

63 

Hartkopf AD, Banys M and Fehm T: HER2-positive DTCs/CTCs in breast cancer. Recent Results Cancer Res. 195:203–215. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Lee JS, Yoon IS, Lee MS, et al: Anticancer activity of pristimerin in epidermal growth factor receptor 2-positive SKBR3 human breast cancer cells. Biol Pharm Bull. 36:316–325. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Swinnen JV, Heemers H, Deboel L, Foufelle F, Heyns W and Verhoeven G: Stimulation of tumor-associated fatty acid synthase expression by growth factor activation of the sterol regulatory element-binding protein pathway. Oncogene. 19:5173–5181. 2000. View Article : Google Scholar

66 

Oskouian B: Overexpression of fatty acid synthase in SKBR3 breast cancer cell line is mediated via a transcriptional mechanism. Cancer Lett. 149:43–51. 2000. View Article : Google Scholar

67 

Kumar-Sinha C, Ignatoski KW, Lippman ME, Ethier SP and Chinnaiyan AM: Transcriptome analysis of HER2 reveals a molecular connection to fatty acid synthesis. Cancer Res. 63:132–139. 2003.PubMed/NCBI

68 

Nicolini A, Giardino R, Carpi A, et al: Metastatic breast cancer: an updating. Biomed Pharmacother. 60:548–556. 2006. View Article : Google Scholar : PubMed/NCBI

69 

Camassei FD, Cozza R, Acquaviva A, et al: Expression of the lipogenic enzyme fatty acid synthase (FAS) in retinoblastoma and its correlation with tumor aggressiveness. Invest Ophthalmol Vis Sci. 44:2399–2403. 2003. View Article : Google Scholar : PubMed/NCBI

70 

Rashid A, Pizer ES, Moga M, et al: Elevated expression of fatty acid synthase and fatty acid synthetic activity in colorectal neoplasia. Am J Pathol. 150:201–208. 1997.PubMed/NCBI

71 

Kalyankrishna S and Grandis JR: Epidermal growth factor receptor biology in head and neck cancer. J Clin Oncol. 24:2666–2672. 2006. View Article : Google Scholar : PubMed/NCBI

72 

Qiu Z, Huang C, Sun J, et al: RNA interference-mediated signal transducers and activators of transcription 3 gene silencing inhibits invasion and metastasis of human pancreatic cancer cells. Cancer Sci. 98:1099–1106. 2007. View Article : Google Scholar

73 

Horiguchi A, Asano T, Asano T, Ito K, Sumitomo M and Hayakawa M: Fatty acid synthase over expression is an indicator of tumor aggressiveness and poor prognosis in renal cell carcinoma. J Urol. 180:1137–1140. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Piyathilake CJ, Frost AR, Manne U, Bell WC, Weiss H, Heimburger DC, et al: The expression of fatty acid synthase (FASE) is an early event in the development and progression of squamous cell carcinoma of the lung. Hum Pathol. 31:1068–1073. 2000. View Article : Google Scholar : PubMed/NCBI

75 

Carvalho MA, Zecchin KG, Seguin F, et al: Fatty acid synthase inhibition with Orlistat promotes apoptosis and reduces cell growth and lymph node metastasis in a mouse melanoma model. Int J Cancer. 123:2557–2565. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Murata S, Yanagisawa K, Fukunaga K, et al: Fatty acid synthase inhibitor cerulenin suppresses liver metastasis of colon cancer in mice. Cancer Sci. 101:1861–1865. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Zaytseva YY, Rychahou PG, Gulhati P, et al: Inhibition of fatty acid synthase attenuates CD44-associated signaling and reduces metastasis in colorectal cancer. Cancer Res. 72:1504–1517. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Chajes V, Hulten K, Van Kappel AL, et al: Fatty-acid composition in serum phospholipids and risk of breast cancer: an incident case-control study in Sweden. Int J Cancer. 83:585–590. 1999. View Article : Google Scholar : PubMed/NCBI

79 

Chajes V, Thiebaut AC, Rotival M, et al: Association between serum trans-monounsaturated fatty acids and breast cancer risk in the E3N-EPIC study. Am J Epidemiol. 167:1312–1320. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Pala V, Krogh V, Muti P, et al: Erythrocyte membrane fatty acids and subsequent breast cancer: a prospective Italian study. J Natl Cancer Inst. 93:1088–1095. 2001. View Article : Google Scholar : PubMed/NCBI

81 

Mauvoisin D, Charfi C, Lounis AM, Rassart E and Mounier C: Decreasing stearoyl-CoA desaturase-1 expression inhibits beta-catenin signaling in breast cancer cells. Cancer Sci. 104:36–42. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Goodridge AG: Regulation of the activity of acetyl coenzyme A carboxylase by palmitoyl coenzyme A and citrate. J Biol Chem. 247:6946–6952. 1972.PubMed/NCBI

83 

Zureik M, Ducimetiere P, Warnet JM and Orssaud G: Fatty acid proportions in cholesterol esters and risk of premature death from cancer in middle aged French men. BMJ. 311:1251–1254. 1995. View Article : Google Scholar : PubMed/NCBI

84 

Petrek JA, Hudgins LC, Ho M, Bajorunas DR and Hirsch J: Fatty acid composition of adipose tissue, an indication of dietary fatty acids, and breast cancer prognosis. J Clin Oncol. 15:1377–1384. 1997.PubMed/NCBI

85 

Zhu ZR, Agren J, Mannisto S, et al: Fatty acid composition of breast adipose tissue in breast cancer patients and in patients with benign breast disease. Nutr Cancer. 24:151–160. 1995. View Article : Google Scholar : PubMed/NCBI

86 

Simonsen NR, Fernandez-Crehuet Navajas J, Martin-Moreno JM, et al: Tissue stores of individual monounsaturated fatty acids and breast cancer: the EURAMIC study. European Community Multicenter Study on Antioxidants, Myocardial Infarction, and Breast. Cancer Am J Clin Nutr. 68:134–141. 1998.PubMed/NCBI

87 

Lamouille S and Derynck R: Emergence of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin axis in transforming growth factor-beta-induced epithelial-mesenchymal transition. Cells Tissues Organs. 193:8–22. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Lin SY, Xia W, Wang JC, et al: Beta-catenin, a novel prognostic marker for breast cancer: its roles in cyclin D1 expression and cancer progression. Proc Natl Acad Sci USA. 97:4262–4266. 2000. View Article : Google Scholar : PubMed/NCBI

89 

Rios-Esteves J and Resh MD: Stearoyl CoA desaturase is required to produce active, lipid-modified Wnt proteins. Cell Rep. 4:1072–1081. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Samuel W, Nagineni CN, Kutty RK, et al: Transforming growth factor-beta regulates stearoyl coenzyme A desaturase expression through a Smad signaling pathway. J Biol Chem. 277:59–66. 2002. View Article : Google Scholar : PubMed/NCBI

91 

McIntyre E, Blackburn E, Brown PJ, Johnson CG and Gullick WJ: The complete family of epidermal growth factor receptors and their ligands are co-ordinately expressed in breast cancer. Breast Cancer Res Treat. 122:105–110. 2010. View Article : Google Scholar : PubMed/NCBI

92 

Menendez JA, Vazquez-Martin A, Ropero S, Colomer R and Lupu R: HER2 (erbB-2)-targeted effects of the omega-3 polyunsaturated fatty acid, alpha-linolenic acid (ALA; 18:3n-3), in breast cancer cells: the ‘fat features’ of the ‘Mediterranean diet’ as an ‘anti-HER2 cocktail’. Clin Transl Oncol. 8:812–820. 2006.PubMed/NCBI

93 

Wells WA, Schwartz GN, Morganelli PM, Cole BF, Gibson JJ and Kinlaw WB: Expression of ‘Spot 14’ (THRSP) predicts disease free survival in invasive breast cancer: immunohistochemical analysis of a new molecular marker. Breast Cancer Res Treat. 98:231–240. 2006.

94 

Kinlaw WB, Quinn JL, Wells WA, Roser-Jones C and Moncur JT: Spot 14: A marker of aggressive breast cancer and a potential therapeutic target. Endocrinology. 147:4048–4055. 2006. View Article : Google Scholar : PubMed/NCBI

95 

Chin K, DeVries S, Fridlyand J, et al: Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell. 10:529–541. 2006. View Article : Google Scholar : PubMed/NCBI

96 

Moreau K, Dizin E, Ray H, et al: BRCA1 affects lipid synthesis through its interaction with acetyl-CoA carboxylase. J Biol Chem. 281:3172–3181. 2006. View Article : Google Scholar : PubMed/NCBI

97 

Saxena NK and Sharma D: Metastasis suppression by adiponectin: LKB1 rises up to the challenge. Cell Adh Migr. 4:358–362. 2010. View Article : Google Scholar : PubMed/NCBI

98 

Scott KE, Wheeler FB, Davis AL, Thomas MJ, Ntambi JM, Seals DF, et al: Metabolic regulation of invadopodia and invasion by acetyl-CoA carboxylase 1 and de novo lipogenesis. PloS One. 7:e297612012. View Article : Google Scholar : PubMed/NCBI

99 

Menendez JA and Lupu R: Mediterranean dietary traditions for the molecular treatment of human cancer: anti-oncogenic actions of the main olive oil’s monounsaturated fatty acid oleic acid (18:1n-9). Curr Pharm Biotechnol. 7:495–502. 2006.PubMed/NCBI

100 

Swinnen JV, Ulrix W, Heyns W and Verhoeven G: Coordinate regulation of lipogenic gene expression by androgens: evidence for a cascade mechanism involving sterol regulatory element binding proteins. Proc Natl Acad Sci USA. 94:12975–12980. 1997. View Article : Google Scholar

101 

Huang WC, Li X, Liu J, Lin J and Chung LW: Activation of androgen receptor, lipogenesis, and oxidative stress converged by SREBP-1 is responsible for regulating growth and progression of prostate cancer cells. Mol Cancer Res. 10:133–142. 2012. View Article : Google Scholar

102 

Bhandary B, Marahatta A, Kim HR and Chae HJ: Mitochondria in relation to cancer metastasis. J Bioenerg Biomembr. 44:623–627. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2014
Volume 45 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mounier C, Bouraoui L and Rassart E: Lipogenesis in cancer progression (Review). Int J Oncol 45: 485-492, 2014
APA
Mounier, C., Bouraoui, L., & Rassart, E. (2014). Lipogenesis in cancer progression (Review). International Journal of Oncology, 45, 485-492. https://doi.org/10.3892/ijo.2014.2441
MLA
Mounier, C., Bouraoui, L., Rassart, E."Lipogenesis in cancer progression (Review)". International Journal of Oncology 45.2 (2014): 485-492.
Chicago
Mounier, C., Bouraoui, L., Rassart, E."Lipogenesis in cancer progression (Review)". International Journal of Oncology 45, no. 2 (2014): 485-492. https://doi.org/10.3892/ijo.2014.2441