2017
DOI: 10.1126/scisignal.aao6604
|View full text |Cite
|
Sign up to set email alerts
|

Oncogenic PI3K promotes methionine dependency in breast cancer cells through the cystine-glutamate antiporter xCT

Abstract: The precursor homocysteine is metabolized either through the methionine cycle to produce methionine or through the transsulfuration pathway to synthesize cysteine. Alternatively, cysteine can be obtained through uptake of its oxidized form, cystine. Many cancer cells exhibit methionine dependency such that their proliferation is impaired in growth media in which methionine is replaced by homocysteine. Here, we showed that oncogenic PIK3CA and decreased expression of SLC7A11, a gene that encodes a cystine trans… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

3
81
0

Year Published

2018
2018
2023
2023

Publication Types

Select...
8
1

Relationship

0
9

Authors

Journals

citations
Cited by 80 publications
(84 citation statements)
references
References 57 publications
3
81
0
Order By: Relevance
“…The differential metabolic fates of homocysteine in methionine-dependent (transsulfuration) and -independent cells (mainly methionine cycle) observed in breast cancer cell systems may be a general feature of the Hoffman effect. Indeed, similar redirection of homocysteine into the transsulfuration branch is induced by oncogenic PI3K mutations, which induce a methionine-dependent phenotype when expressed in methionine-independent cells (Figure 2A) [10]. It is interesting to note that the transsulfuration pathway is largely restricted to pancreas, liver, and kidney, but is active in cancer cells [8,[35][36][37], which could contribute to the differential response of cancer cells to growth in homocysteine medium.…”
Section: Methionine Metabolism and Cancermentioning
confidence: 86%
See 1 more Smart Citation
“…The differential metabolic fates of homocysteine in methionine-dependent (transsulfuration) and -independent cells (mainly methionine cycle) observed in breast cancer cell systems may be a general feature of the Hoffman effect. Indeed, similar redirection of homocysteine into the transsulfuration branch is induced by oncogenic PI3K mutations, which induce a methionine-dependent phenotype when expressed in methionine-independent cells (Figure 2A) [10]. It is interesting to note that the transsulfuration pathway is largely restricted to pancreas, liver, and kidney, but is active in cancer cells [8,[35][36][37], which could contribute to the differential response of cancer cells to growth in homocysteine medium.…”
Section: Methionine Metabolism and Cancermentioning
confidence: 86%
“…As indicated above, generation of methionine stress resistant cancer cells is usually coupled with loss of tumorigenic properties such as the ability of anchorage independent growth and proliferation in 1% serum. Conversely, oncogenic mutations in phosphoinositide 3-kinase (PI3K) and H-ras expression have been shown to promote methionine dependence [10,33]. Whether other oncogenes induce a similar metabolic dependence has not been systematically investigated, but these experiments indicate a tight link between methionine dependence and tumorigenicity.…”
Section: Methionine Metabolism and Cancermentioning
confidence: 99%
“…The component of class III phosphatidylinositol 3-kinase complex BECN1 binds to xCT when it is phosphorylated by AMPK and negatively regulates xCT activity (49). The activity of xCT is also negatively regulated by phosphorylation on serine 26 in the N-terminal cytosolic region (50,51), although it is unclear whether this affects xCT protein stability.…”
Section: Discussionmentioning
confidence: 99%
“…For example, epidermal growth factor receptor 11 and CD44 variants 12 directly interact with xCT and stabilize its expression at the cell surface, which enhances antioxidant capacity. In contrast, oncogenic PI3KCA inhibits xCT and promotes methionine dependency in mammary epithelial tumors 13 , and 5′ adenosine monophosphate-activated protein kinase (AMPK)-mediated phosphorylation of Beclin-1 promote ferroptosis via complexation with xCT 14 . xCT expression is transcriptionally activated by nuclear factor erythroid 2related factor 2 (Nrf2) and activating transcription factor 4 (ATF4) under conditions of oxidative stress or hypoxia 15,16 , whereas it is inhibited by the tumor suppressor TP53 following reactive oxygen species (ROS)induced stress independent of its functions regulating apoptosis and senescence 17 . Taken together, these findings indicate that xCT expression is precisely regulated by various growth-regulating signals to meet genetic, metabolic, and environmental needs.…”
Section: System X Cmentioning
confidence: 99%