Skip to main content
Erschienen in: Cancer Cell International 1/2011

Open Access 01.12.2011 | Primary research

Upstream molecular signaling pathways of p27 (Kip1) expression in human breast cancer cells in vitro: differential effects of 4-hydroxytamoxifen and deficiency of either D-(+)-glucose or L-leucine

verfasst von: Isao Eto

Erschienen in: Cancer Cell International | Ausgabe 1/2011

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

The objective of this study was to investigate whether the levels of glucose or certain amino acids could regulate the expression of a cell cycle repressor protein p27(Kip1), thereby dictating the risk of cancer in either obesity or caloric/dietary restriction. Previously, we identified and reported four different upstream molecular signaling pathways of p27 expression in human breast cancer cells. We called these four pathways as pathway #1, #2, #3 and #4. We found that 4-hydroxytamoxifen - but not tamoxifen - up-regulated the expression of p27 using pathway #1 which consisted mainly of receptor tyrosine kinases and mTORC1. We now investigate, using 4-hydroxytamoxifen as a reference anti-cancer agents, whether (a) the moderate increase in the concentration of D-(+)-glucose could down-regulate and, conversely, (b) the deficiency of D-(+)-glucose or certain L-amino acids could up-regulate the expression of p27 in these cells using pathway #2 which consists mainly of AMPK and mTORC1.

Results

Using human MDA-MB-231 breast cancer cells in vitro, these hypotheses were tested experimentally by performing p27-luciferase reporter transfection assays and western immunoblot analyses. The results obtained are consistent with these hypotheses. Furthermore, the results indicated that, although 4-hydroxytamoxifen used primarily pathway #1 to down-regulate the phosphorylation of 4E-BP1 and up-regulate the expression of p27, it also secondarily down-regulated the phosphorylation of S6K1. In contrast, the deficiency of D-(+)-glucose or L-leucine used primarily pathway #2 to down-regulate the phosphorylation of S6K1, but they also secondarily down-regulated the phosphorylation of 4E-BP1 and up-regulated the expression of p27. Finally, deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxytamoxifen - up-regulated the expression of mitochondrial ATP5A and SIRT3.

Conclusions

(a) 4-Hydroxitamoxifen used primarily pathway #1 to up-regulate the expression of p27. (b) Moderate increase in the concentration of D-(+)-glucose used primarily pathway #2 to down-regulate the expression of p27. (c) Deficiency of D-(+)-glucose or L-leucine also used primarily pathway #2 to up-regulate the expression of p27. (d) Deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxytamoxifen - up-regulated the expression of mitochondrial ATP5A in the Complex V of respiratory oxidation-phosphorylation chain and mitochondrial SIRT3. The SIRT3 is one of the seven mammalian anti-aging as well as anti-metabolic sirtuins.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1475-2867-11-31) contains supplementary material, which is available to authorized users.

Competing interests

The author declares that they have no competing interests.
Abkürzungen
p27
p27Kip1
p21
p21Cip1/Waf1
AMPK
5'-AMP-activated protein kinase
TSC
tuberous sclerosis complex
mTORC1
mammalian target of rapamycin complex 1
RTK
receptor tyrosine kinase
PI3K
phosphoinositide 3-kinase
PKB
protein kinase B
AMPK
5'-AMP-activated protein kinase
MAPK
mitogen-activated protein kinase
MEK
mitogen-activated protein (MAP) kinase kinase
ERK
ERK MAP kinase
MNK
MAP kinase interacting kinase
m7G
7-methylguanosine
CDK
cyclin-dependent kinase
CDI
cyclin-dependent kinase inhibitor
MNU
N-methyl-N-nitrosourea
ER
estrogen-receptor
5'-UTR
5'-untranslated region
IRES
internal ribosome entry site
DMSO
dimethyl sulfoxide
pGL3
pGL3 luciferase reporter vector
AdoMet or SAM
S-(5'-adenosyl)-L-methionine
AdoHcy or SAH
S-(5'-adenosyl)-L-homocysteine
4E-BP1
eukaryotic translation initiation factor 4E binding protein 1
S6K
p70 S6 kinase
AICAR
5-amino-4-imidazolecarboxamide aminoimidazole carboxamide ribonucleotide
Glc
D-(+)-glucose
Ser
L-serine
Thr
L-threonine
Met
L-methionine
Cys
L-cysteine
Leu
L-leucine
Tyr
L-tyrosine
eIF4E
eukaryotic translation initiation factor 4E
uORF
5'-upstream open reading frame
FBS
fetal bovine serum
DMEM
Dulbecco's modified Eagle's medium
EDTA
ethylenediaminetetraacetic acid
CPRG
chlorophenol red-β-D-galactopyranoside
βGal
β-galactosidase
Luc
firefly luciferase
GAPDH
glyceraldehydes phosphate dehydrogenase
mTORC1
mammalian target of rapamycin complex 1
HIF-1α
hypoxia-inducible factor 1α
SREBP1
sterol regulatory element binding protein-1
eEF2k
eukaryotic elongation-factor-2 kinase
ATP5A
mitochondrial ATP Synthase α chain in the Complex V of respiratory oxidation-phosphorylation chain
SIRT3
sirtuin 3
SIRT1
sirtuin 1
OxPhos
mitochondrial oxidation-phosphorylation (repiratory electron transfer) chain, ROS, reactive oxygen species.

Background

The risk of developing cancer is increased in obesity where the serum levels of glucose, certain amino acids, insulin and other growth factors tend to be elevated. Conversely, the risk of developing cancer is decreased in caloric/dietary restriction where the serum levels of these metabolites tend to be reduced. The objective of this study was to investigate whether the levels of glucose or certain amino acids could regulate the expression of a cell cycle repressor protein p27(Kip1), thereby dictating the risk of cancer in either obesity or caloric/dietary restriction.
p27 is a member of the family of cyclin-dependent kinase (CDK) inhibitors (CDIs). p27 binds to certain cyclin/CDK complexes, arrests the cell cycle progression from G1 to S phase and inhibit DNA replication. It is known that a relatively large number of nutritional and chemopreventive anti-cancer agents - including 4-hydroxytamoxifen - specifically up-regulate the expression of p27 in both estrogen receptor (ER)-positive and -negative human breast cancer cells in vitro[1, 2]. It is also known that some other anti-cancer agents specifically up-regulate the expression of p27 in either ER-positive or -negative human breast cancer cells in vitro[1, 2].
p27 exhibits a set of unique characteristics that are not seen in other G1-to-S phase cell cycle regulatory proteins [1, 2]. First, various anti-cancer agents specifically up-regulate the expression of p27 without directly affecting expression of other G1-to-S phase cell cycle regulatory proteins including INK4s, p57(Kip2), p21(Cip1Waf1), D-type cyclins, cyclin E, cyclin A, CDK2, CDK4 and CDK6 [13]. Secondly, the degree of up-regulation of the expression of p27 in human breast cancer cell lines in vitro by these anti-cancer agents linearly and positively correlates with the degree of inhibition of methylnitrosourea (MNU)-induced rat mammary adenocarcinoma in vivo by the same anti-cancer agents [2]. This linear and positive correlation could not be held if a particular anti-cancer agent must be converted to an active metabolite in vivo in order to up-regulate the expression of p27. An example of such anti-cancer agent is tamoxifen which must be converted to 4-hydroxytamoxifen in vivo to specifically up-regulate the expression of p27. Lastly, unlike other G1-to-S phase cell cycle regulatory proteins, expression of p27 is not regulated at the level of transcription, but primarily at the level of translation. It was observed in the 1980s and 1990s that, during the progression of cell cycle, the level of p27 protein expression oscillated cyclically, but the level of p27 mRNA remained constant. This observation led investigators to suggest that, during the cell cycle, expression of p27 is regulated primarily at the level of translation [410]. It was also proposed that the expression of p27 during the progression of cell cycle could be regulated by various other post-translational mechanisms including ubiquitin-proteasome-induced degradation [1114], complex formation [15], subcellular localization [1621] and phosphorylation [2123]. Based on the results of our previous studies [1, 2], we believe that a large number of anti-cancer agents up-regulate the expression of p27 primarily by activating the rate of translation initiation of p27 mRNA.
Despite all these information, however, very little is known about the upstream molecular signaling pathways of how various anti-cancer agents specifically up-regulate the expression of p27 in human breast cancer cells in vitro. Previously, we identified and reported four different upstream molecular signaling pathways of p27 expression by using p27-luciferase reporter plasmids, western immunoblot analysis and numerous specific inhibitors and stimulators of p27 expression [1, 2]. (We will call these four pathways as #1, #2, #3 and #4.).
We also reported previously that, in both ER-positive and -negative human breast cancer cells in vitro, 4-hydroxytamoxifen (4-OH-tamoxifen) - but not tamoxifen - up-regulated the expression of p27 by using pathway #1 which consists mainly of receptor tyrosine kinases (RTKs) and mammalian target of rapamycin complex 1 (mTORC1) [2].
We now hypothesize that moderate increase in the concentration of D-(+)-glucose down-regulates the expression of p27 in human breast cancer cells in vitro by using pathway #2 which consists mainly of 5'-AMP-activated protein kinase (AMPK) and mTORC1. Conversely, we also hypothesize that deficiency of D-(+)-glucose or certain L-amino acids up-regulates the expression of p27 in these cells by using the pathway #2
To test these hypotheses, we performed p27-luciferase reporter transfection assays and western immunoblot analyses using ER and LKB1-double negative human MDA-MB-231 breast cancer cell in vitro. The results obtained were consistent with these hypotheses. Additional results were also obtained that indicated that deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxytamoxifen - up-regulated the expression of mitochondrial ATP Synthase α chain (ATP5A) in the Complex V of respiratory oxidation-phosphorylation chain and mitochondrial SIRT3 in these cells. The SIRT3 is one of the seven mammalian anti-aging and anti-metabolic sirtuins.

Results

4-Hydroxytamoxifen - but not tamoxifen - up-regulated the expression of p27 in estrogen receptor (ER) and LKB1-double negative human MDA-MB-231 breast cancer cells in vitro

The effects of 4-hydroxytamoxifen (4-OH-tamoxifen) and tamoxifen on the expression of p27 in human breast cancer cells in vitro were investigated using p27-luciferase reporter plasmids containing the following proximal 5'-upstream regions of p27 gene, namely -1797 p27 (p27-Kpn I), -774 p27 (p27-Apa I), and -575 p27 (p27-5'UTR) (Figure 1a). These plasmids were transfected into ER and LKB1-double negative human MDA-MB-231 breast cancer cells in vitro and then the transfected cells were exposed to DMSO or 1 μM each of tamoxifen or 4-OH-tamoxifen for 24 hours. The results indicated that tamoxifen did not up-regulate the relative luciferase activity of p27 (Figure 1b), but 4-OH-tamoxifen up-regulated it in these cells (Figure 1c). Previously, we reported essentially the same results using ER and LKB1-double positive human MCF7 breast cancer cells in vitro[1, 2].
Additionally, the results of these studies, along with those of our previous studies [1, 2], were consistent with the hypothesis that the expression of p27 is regulated primarily at the level of translation. For more information about this issue, please go to the Methods section below,
Based on these results, we constructed a schematic diagram (Figure 1d) showing the outline of how 4-OH-tamoxifen could up-regulate the expression of p27, down-regulate the cell cycle progression from G1 to S phase, thereby inhibiting the DNA replication of the human breast cancer cells in vitro.

Moderate increase in the concentration of D-(+)-glucose down-regulated the expression of p27, and, conversely, deficiency of D-(+)-glucose, L-leucine, L-methionine, L-cysteine or combination of L-methionine and L-cysteine up-regulated the expression of p27 in human MDA-MB-231 breast cancer cells in vitro

The effects of (a) moderate increase in the concentration of D-(+)-glucose and (b) deficiency of D-(+)-glucose, L-leucine, L-methionine, L-cysteine or combination of L-methionine and L-cysteine on the expression of p27 in MDA-MB-231 cells were investigated using one of the luciferase reporter plasmids containing a proximal 5'-upstream region of p27 gene (-575 p27-5'UTR) (Figure 2a).
The results indicated that moderate increase in the concentration of D-(+)-glucose down-regulated the relative luciferase activity of -575 p27-5'UTR in MDA-MB-231 cells (Figures 2b). In contrast, deficiency of D-(+)-glucose, L-leucine, L-methionine, L-cysteine or combination of L-methionine and L-cysteine up-regulated the relative luciferase activity of p27 in these cells (Figure 2c). It should be noted that deficiency of the combination of L-methionine and L-cysteine up-regulated the relative luciferase activity of p27 more than the deficiency of individual amino acids.
The results (Figure 2b) also indicated that (a) rotenone (inhibitor of NADH dehydrogenase (Complex 1) of the mitochondrial respiratory oxidation-phosphorylation chain) and AICA riboside (inhibitor of AMPK (5'-AMP-activated protein kinase)) up-regulated the relative luciferase activity of p27 in MDA-MB-231 cells, but (b) compound C (activator of AMPK) down-regulated the relative luciferase activity of p27 in these cells. Metformin did not either up or down-regulate the relative luciferase activity of p27 probably because MDA-MB-231 cells lack LKB1.

Differential effects of 4-hydroxytamoxifen and deficiency of D-(+)-glucose on the upstream molecular signaling pathways of the expression of p27: pathways immediately downstream of mTORC1 (mammalian target of rapamycin complex 1)

Previously, we identified and reported four different upstream molecular signaling pathways of p27 expression that could lead to either activation or inactivation of the translation initiation of p27 mRNA through its unusually long 5'-untranslated region (5'-UTR) (-575) of p27 mRNA (Figure 3a and 3b) [2]. We also reported previously that 4-hydroxytamoxifen (4-OH-tamoxifen) up-regulated the expression of p27 by using pathway #1 which consists mainly of receptor tyrosine kinases (RTKs) and mTORC1 (Figure 3a) [2]. We now hypothesize that (a) moderate increase in the concentration of D-(+)-glucose down-regulates and, conversely, (b) deficiency of D-(+)-glucose or certain L-amino acids up-regulates the expression of p27 by using pathway #2 which consists mainly of AMPK (5'-AMP-activated protein kinase) and mTORC1 (mammalian target of rapamycin complex 1) (Figure 3a).
To begin to test these hypotheses, we first performed the western immunoblot analysis of the expression of p27 protein itself. The results (Figures 3c, d and 3e) indicated that 4-OH-tamoxifen and deficiency of D-(+)-glucose or L-leucine up-regulated the expression of p27 protein, but deficiency of L-methionine or L-cysteine did not in MDA-MB-231 cells.
In order to look more closely into the effects of 4-OH-tamoxifen and deficiency of D-(+)-glucose or certain L-amino acids on the upstream molecular signaling pathways #1 and #2 of the expression of p27, western immunoblot analyses were performed to investigate the proteins immediately downstream of mTORC1, namely eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1) and p70 S6 kinase 1 (S6K1).

(a) Differential effects on the phosphorylation of 4E-BP1

Figure 4a to 4e show that (a) 4-OH-tamoxifen and (b) deficiency of D-(+)-glucose or L-leucine did not either down or up-regulate the expression of total 4E-BP1, but they down-regulated the phosphorylated 4E-BP1. As summarized in Figure 4f, the degree of down-regulation of the phosphorylated 4E-BP1 appeared to be positively and linearly correlated with the degree of expression of p27.

(b) Differential effects on the phosphorylation of S6K1

Figure 5a to 5e show that (a) 4-OH-tamoxifen and (b) deficiency of D-(+)-glucose, L-leucine or L-methionine did not influence the expression of total S6K1, but they down-regulated the phosphorylated S6K1. As summarized in the Figure 5f, the degree of down-regulation of the phosphorylated S6K1 did not appear to be correlated with the degree of expression of p27.
It should be noted that (a) 4-OH-tamoxifen and (b) deficiency of D-(+)-glucose or certain L-amino acids exerted differential effects on the degree of down-regulation of either the phosphorylated 4E-BP1 or phosphorylated S6K1. For example, (a) 4-OH-tamoxifen preferentially down-regulated the phosphorylation of 4E-BP1 over S6K1. Conversely, (b) D-(+)-glucose deficiency preferentially down-regulated the phosphorylation of S6K1 over 4E-BP1. (c) L-Leucine deficiency significantly down-regulated the phosphorylation of both 4E-BP1 and S6K1, but to a much lesser extent. (d) L-Methionine deficiency significantly down-regulated the phosphorylation of only S6K1 and to a much lesser extent; but it did not significantly down-regulate the phosphorylation of 4E-BP1. Lastly, (e) L-cysteine deficiency did not significantly down-regulate the phosphorylation of either 4E-BP1 or S6K1.

Differential effects of 4-hydroxytamoxifen and deficiency of D-(+)-glucose on the upstream molecular signaling pathways of p27 expression: pathways further downstream of mTORC1

Next, we investigated the effects of tamoxifen, 4-OH-tamoxifen, and the deficiency of D-(+)-glucose on the pathways further downstream of mTORC1. They were (a) hypoxia-inducible factor 1α (HIF-1α), (b) sterol regulatory element binding protein-1 (SREBP-1) and (c) phosphorylation of eukaryotic elongation-factor-2 kinase (eEF2k). The results of the western immunoblot analyses are presented in Figure 6a to 6e. The effects of L-amino acid deficiencies were not investigated because they exerted either only a moderate or no effect on the phosphorylation of 4E-BP1 or S6K1.

(a) Differential effects on HIF-1α

HIF-1α has been variably characterized in the literature as being a protein downstream of 4E-BP1, S6K1 or both. The results of our western immunoblot analyses presented in Figure 6a and 6e indicated that D-(+)-glucose deficiency significantly down-regulated the expression of HIF-1α; but 4-OH-tamoxifen did not. These results are consistent with the hypothesis that HIF-1α is a protein primarily downstream of S6K1.

(b) Differential effects on SREBP-1 and phosphorylated eEF2k

No controversy exists in the literature as to the SREBP1 and eEF2k; the consensus is that they are the proteins primarily downstream of S6K1. The results of our western immunoblot analyses of SREBP1 (Figure 6b and 6e) and phosphorylated eEF2k at Ser366 (Figure 6c, d and 6e) are consistent with this consensus.

Differential effects of 4-hydroxytamoxifen and deficiency of D-(+)-glucose or L-leucine on the upstream molecular signaling pathways of p27 expression: pathways upstream of mTORC1

The results presented above (Figure 2b) suggested that NADH dehydrogenase (Complex 1) in the mitochondrial respiratory oxidation-phosphorylation chain and 5'-AMP-activated protein kinase (AMPK) are the two critical components of the pathway #2 upstream of mTORC1. In addition to these two proteins, we investigated two other proteins that also appeared to be associated with the pathway #2 upstream of mTORC1. They were mitochondrial ATP Synthase α chain (ATP5A) in the Complex V of respiratory oxidation-phosphorylation chain and mitochondrial SIRT3.

(a) Differential effects on the mitochondrial ATP5A

During our preliminary proteomic analysis of the hepatic proteins of genetically obese mice and long-lived dwarf mice, we observed that mitochondrial ATP5A was most significantly down-regulated in the liver of leptin-deficient obese mice relative to the lean control mice. Conversely, we also observed that mitochondrial ATP5A was most significantly up-regulated in the liver of long-lived Ames dwarf mice compared to the normal Ames mice. Based on these preliminary observations, we decided to investigate the effects of 4-OH-tamoxifen and deficiency of D-(+)-glucose or certain L-amino acids on the expression of mitochondrial ATP5A in the human MDA-MB-231 breast cancer cells in vitro.
The results of our western immunoblot analyses (Figure 7a, b and 7e) indicated that 4-OH-tamoxifen did not influence the expression of mitochondrial ATP5A, but deficiency of D-(+)-glucose, L-leucine or L-methionine up-regulated it. Deficiency of L-cysteine did not alter the expression of mitochondrial ATP5A.

(b) Differential effects on the mitochondrial SIRT3

Mitochondrial SIRT3 is one of the seven mammalian anti-aging and anti-metabolic sirtuins. It was reported recently that mitochondrial ATP5A forms complex with and interacts with mitochondrial SIRT3 [24]. Based on this report, we decided to investigate the effects of 4-OH-tamoxifen and deficiency of D-(+)-glucose or certain L-amino acids on the expression of mitochondrial SIRT3 in the human MDA-MB-231breast cancer cells in vitro.
The results of our western immunoblot analyses (Figure 7c, d and 7e) indicated that deficiency of D-(+)-glucose or L-leucine - but not 4-OH-tamoxifen - up-regulated the expression of mitochondrial SIRT3 in these cells. Deficiency of L-methionine or L-cysteine, however, did not either up or down-regulate the expression of SIRT3.
Finally, 4-OH-tamoxifen and deficiency of D-(+)-glucose or certain L-amino acids did not regulate the expression of nuclear anti-aging and anti-metabolic protein SIRT1 in these cells (results not shown).

Discussion

Based on the results presented above, a schematic diagram is presented in Figure 8 that outlines the effects of 4-hydroxitamoxifen, moderate increase in the concentration of D-(+)-glucose and deficiency of D-(+)-glucose or L-leucine on the pathways #1 and #2 of the upstream molecular signaling pathways of the expression of p27 in human breast cancer cells in vitro. The results presented are consistent with the following hypotheses, namely:
(a) 4-Hydroxitamoxifen up-regulates the expression p27 in human breast cancer cells in vitro primarily by using pathway #1. The pathway #1 consists mainly of receptor tyrosine kinases/phosphoinositide-3-kinase/Akt/tuberous sclerosis complex/mammalian target of rapamycin complex 1/eukaryotic translation initiation factor 4E-binding protein 1 (RTKs/PI3K/Akt/TSC/mTORC1/4E-BP1).
(b) Moderate increase in the concentration of D-(+)-glucose or certain amino acids down-regulates and deficiency of D-(+)-glucose or L-leucine up-regulates the expression of p27 in human breast cancer cells in vitro primarily by using pathway #2. The pathway #2 consists mainly of 5'-AMP-activated protein kinase/tuberous sclerosis complex/mammalian target of rapamycin complex 1/p70 S6 kinase I (AMPK/TSC/mTORC1/S6K1). The pathway #2 also modulates the phosphorylation of 4E-BP1 thereby regulating the expression of p27, but this effect is secondary to its effect on the phosphorylation of S6K1.
Down-regulation of the phosphorylation of S6K1 in the pathway #2 by the deficiency of D-(+)-glucose resulted in the (i) down-regulation (de-stabilization) of HIF-1α, (ii) up-regulation (stabilization) of SREBP1 and (iii) down-regulation of the phosphorylation of eEF2k.
The SREBP1 is of particular interest here because SREBP1 has recently been implicated in the insulin resistance of type 2 diabetes. It is well known that, in the liver of many insulin-resistant mouse models, insulin fails to suppress D-(+)-glucose production (gluconeogenesis) but continue to promote lipid synthesis. It is also known that mTORC1 down-regulates (activates or de-stabilizes) SREBP-1 and thereby uncouples lipogenesis from gluconeogenesis [25]. This down-regulation of the expression of SREBP1 by mTORC1 appears to be achieved at least in part by promoting its posttranslational processing via S6K1, which in turn leads to the increased transcription of genes involved in sterol and lipid biosynthesis and the oxidative arm of the pentose-phosphate pathway [26, 27]. mTORC1-mediated decrease in (or activation or de-stabilization of) the expression of SREBP-1 also appears to be necessary for lipogenesis induced by Akt [28]. In summary, our results suggested that the deficiency of D-(+)-glucose could suppress insulin resistance and restore insulin sensitivity by down-regulating the phosphorylation of S6K1 and up-regulating (de-activating or stabilizing) the expression of SREBP1,

Deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxitamoxifen - up-regulates the expression of mitochondrial ATP Synthase α chain (ATP5A) in the Complex V of respiratory oxidation-phosphorylation chain

During our preliminary proteomic analysis of the hepatic proteins of leptin-deficient obese mice and long-lived dwarf mice, we observed that the expression of mitochondrial ATP5A protein was most significantly down-regulated in the liver of leptin-deficient obese mice relative to the lean control mice. We also observed that ATP5A protein was most significantly up-regulated in the liver of long-lived Ames dwarf mice relative to the normal Ames mice.
Based on these findings, we decided to investigate and, in fact, reported, as described above in the results section, that the deficiency of D-(+)-glucose, L-leucine or L-methionine up-regulated the expression of mitochondrial ATP5A in the human MDA-MB-231 breast cancer cells in vitro as well.
As to the possible molecular mechanisms of the up-regulation of ATP5A, we realized that almost no study was published in the literature. One exception was the study published in 2010, where authors speculated that, as wakefulness continues in mice, the maintenance of ATP becomes more challenging and likely to involve additional nuclear transcriptional mechanisms [29]. The authors further stated that, initially, the demand for increased ATP during wakefulness is met by increased activity of the mitochondrial respiratory oxidation-phosphorylation (OxPhos) system (including ATP5B and probably ATP5A as well). This would eventually lead to an increase in the production of reactive oxygen species (ROS) during extended wakefulness that would then lead to uncoupling with at least temporary decline in ATP and increase in AMP resulting in the activation (increased phosphorylation) of 5'-AMP-dependent protein kinase (AMPK).
This study [29] places the molecular basis of the metabolic up-regulation of the expression of ATP5A by the deficiency of D-(+)-glucose or L-leucine at the AMPK, which is one of the essential components of the pathway #2 in the upstream molecular signaling pathways of p27 expression.

Deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxitamoxifen - up-regulates the expression of mitochondrial SIRT3, one of the seven mammalian anti-aging and anti-metabolic sirtuins

Mitochondrial SIRT3 is one of the seven mammalian sirtuins that are involved in anti-aging and other metabolic processes. Recently, it was reported that mitochondrial SIRT3 forms complex with and interacts with mitochondrial ATP5A [24]. Since SIRT3 is known to be present ubiquitously in the body, we speculated that SIRT3 could also be present in the human MDA-MB-231 breast cancer cells in vitro and, in fact, as described in the results section above, w e found that deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxytamoxifen - up-regulated the expression of SIRT3 in these cells.
Sirtuins are a family of NAD+-dependent protein deacetylases that regulate cellular functions through deacetylation of a wide range of protein targets [24, 3034]. Overexpression of Sir2, the first gene discovered in this family, is able to extend the life span in various organisms. The anti-aging effects of human homologues of sirtuins, SIRT1-7, have also been suggested by animal and human studies.
The results of our study are consistent with the notion that deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxytamoxifen - could exert anti-aging and other metabolic effects through the pathway that involves AMPK, ATP5A and SIRT3. It has been reported, in fact, that the expression of mitochondrial SIRT3 may be up-regulated in caloric restriction and down-regulated in obesity and diabetes [35, 36]. It has also been reported that the activation (increased phosphorylation) of AMPK may lead to up-regulation of the expression of mitochondrial SIRT3 [37].
Thus, the origin of the metabolic up-regulation of the expression of mitochondrial SIRT3 by the deficiency of D-(+)-glucose or L-leucine could be traced to AMPK, which is again one of the essential components of the pathway #2 in the upstream molecular signaling pathways of p27. expression.

Conclusions

Previously, we identified and reported four different upstream molecular signaling pathways - we called them pathway #1, #2, #3 and #4 - of the expression of p27 in human breast cancer cells in vitro. Based on the results presented above, we conclude that:
(a) 4-Hydroxitamoxifen uses primarily pathway #1 to up-regulate the expression of p27. The pathway #1 consists mainly of receptor tyrosine kinases (RTKs) and mammalian target of rapamycin complex 1 (mTORC1).
(b) Moderate increase in the concentration of D-(+)-glucose is likely to use primarily pathway #2 to down-regulate the expression of p27. The pathway #2 consists mainly of 5'-AMP-activated protein kinase (AMPK) and mTORC1 protein kinase.
(c) Deficiency of D-(+)-glucose or L-leucine uses primarily pathway #2 to up-regulate the expression of p27.
(d) Additionally, deficiency of D-(+)-glucose or L-leucine - but not 4-hydroxytamoxifen - also up-regulates the expression of mitochondrial ATP5A in the Complex V of respiratory oxidation-phosphorylation chain and mitochondrial anti-aging as well as anti-metabolic SIRT3.

Methods

Reagents

4-Hydroxytamoxifen, tamoxifen, D-(+)-glucose, and rotenone were obtained from Sigma-Aldrich (St. Louis, MO, USA). Compound C and metformin were obtained from Calbiochem/EMD (San Diego, CA, USA). AICA riboside was purchased from Phoenix Pharmaceuticals, Inc. (Belmont, CA, USA). Dulbecco's Modified Eagle's Medium (DMEM) Labeling Kit was purchased initially from Chemicon International (Temecula, CA, USA) and later from EMD Millipore (Billerica, MA, USA).
The following antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA): namely (a) total 4E-BP1 and phospho-4E-BP1 (Thr37/46); (b) total S6K1 and phospho-S6K1 (Thr389); and (c) total eEF2k and phospho-eEF2k (Ser366). Additionally, the following antibodies were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA): namely (a) p27, (b) GAPDH, (c) ATP5A, (d) SIRT3, (e) SIRT1, (f) SREBP-1, and (g) HIF-1α.

Cell Cultures

Human MDA-MB-231 breast cancer cells (estrogen receptor (ER) and LKB1-double negative) were purchased from the American Type Culture Collection (Rockville, MD, USA). The cells were grown in Dulbecco's Modified Eagle's Medium (DMEM) containing 4.5 g/L of D-(+)-glucose, supplemented with 10% heat-inactivated FBS, 2% L-glutamine, and antibiotics/antimycotics. Incubation of the cells was carried out at 37°C in a 5% CO2 humidified chamber. The cells were subcultured after trypsinization with 0.05% trypsin-0.02% EDTA solution. The cells were always maintained below confluency and checked periodically for mycoplasmal infection by DNA fluorochrome staining.

Plasmids

Luciferase reporter plasmids containing one of the following proximal 5'-upstream regions of the p27 gene were used to transfect the human MDA-MB-231 breast cancer cells: -1797 p27 (p27-Kpn I) [38], -774 p27 (p27-Apa I) [38], and -575 p27 (p27-5'UTR) [4, 8]. The control luciferase reporter plasmids not containing these inserts were also prepared and used to test if 24-hour treatment of the cells with DMSO, 4-hydroxytamoxifen, tamoxifen, excess D-(+)-glucose, or the deficiency of D-(+)-glucose, L-leucine, L-methionine, L-cysteine, or combination of L-methionine and L-cysteine was exerting any spurious effects on the backbone, rather than the insert, of the luciferase reporter plasmids. None of these treatments were found to exert any spurious effects on the backbone of the plasmids in the human MDA-MB-231 breast cancer cells.

Transfection and Luciferase Assay

Transfections were performed according to the published protocol [39] using FuGENE 6 obtained from Roche Applied Science (Indianapolis, IN, USA). In brief, 24 hours before reporter transfection, the cells were seeded into a 60-mm tissue culture dish at a density of 1.5 × 105 cells/dish and incubated at 37°C in a 5% CO2 humidified chamber. Transfection of the luciferase reporter plasmid was then carried out with 1 μg of luciferase reporter plasmid and 0.2 μg of pSV-β-galactosidase internal control plasmid (Promega, Madison, WI, USA) mixed with 3 μL of FuGENE 6 solution in 3 mL of FBS-free DMEM supplemented with only 2% L-glutamine. A minimum of 5-hour incubation at 37°C was needed for transient transfection, followed by 18-hour incubation in DMEM with 10% FBS for recovery. The transfected cells were then starved in DMEM with 0.2% FBS for 24 hours. Subsequently, the resulting cells were cultured either (a) in the presence of DMSO, tamoxifen, or 4-hydroxytamoxifen in the regular DMEM with 0.2% FBS, (b) in the presence of a moderate increase in the concentration of D-(+)-glucose or (c) deficiency of D-(+)-glucose, L-leucine, L-methionine, L-cysteine or combination of L-methionine and L-cysteine in the appropriately supplemented basal DMEM Labeling Kit as described in the figure legends. After 24 hours, the treated cells were collected and lysed using Reporter Lysis Buffer (Promega, Madison, WI) and the resulting cell lysates were assayed for luciferase activity using Luciferase Assay Kit (Promega, Madison, WI, USA) and TD-20/20 Luminometer (Turner Designs, Sunnyvale, CA, USA). β-Galactosidase activity was measured using chlorophenol red-β-D-galactopyranoside (CPRG) (Sigma-Aldrich, St. Louis, MO, USA) as a substrate.
Each luciferase activity driven by a specific proximal 5'-upstream region of the p27 gene was normalized to β-galactosidase activity in order to control for variations in transfection efficiency.
As for the issue of whether the expression of p27 was regulated primarily at the level of translation, we performed the following three different studies:
(a) The various deletion constructs of -1797 p27-Kpn1 luciferase reporter plasmids were used to determine the core element of the activation of the proximal 5'-upstream region (-1797) of p27 gene [1, 2]. The results indicated that various nutritional and chemopreventive anti-cancer agents, including tamoxifen and 4-hydroxytamoxifen, activated the proximal 5'-upstream region (-1797) of p27 gene through its 5'-untranslated region (5'UTR) (-575). It is well established that this region mediates the cap-independent translation initiation of p27 mRNA [410].
(b) To investigate if -575 p27 (p27-5'UTR) contains any cryptic transcription factor binding sites - in other words, if the expression of p27 is regulated primarily at the level of transcription - the luciferase activity of the region was stimulated with 4-hydroxytamoxifen in the presence of an adequate dose of antibiotic actinomycin D [1, 2]. Actinomycin D is a well-known inhibitor of transcription. The results indicated that the -575 p27 (5'-untranslated region (5'UTR)) is unlikely to contain any cryptic transcription factor binding sites. This assay was performed not only with tamoxifen and 4-hydroxytamoxifen, but also with many other anti-cancer agents.
(c) Depending on the cell types, it was observed from time to time that control vector expression was affected by each treatment and also there could be cell cycle effects probably changing with treatment. To exclude these possibilities, the p27 luciferase reporter vector that does not contain and insert of the specific proximal 5'-upstream region of the p27 gene was prepared and tested using the same anti-cancer agents and cell types [1, 2]. In these exceptional cases, the following formula was used to correct this false increase in the relative luciferase activity:
Relative luciferase activity (% ) = (Experimental luciferase activity/Control luciferase activity) × 100,
where,
(1)
Experimental luciferase activity = {Test compound/None}{Luciferase reporter vector containing a specific insert},
 
(2)
Control luciferase activity = {Test compound/None}{Luciferase reporter vector NOT containing a specific promoter insert}, and
 
(3)
Test compound/None = [Luc(Test)/βGal(Test)]/[Luc(None)/βGal(None)].
 
The human MDA-MB-231breast cancer cells that were used in this study did not present any of these exceptional problems. For additional information on this and related issues, please refer to the reference #40 [40].

Western Immunoblot Analysis

Western immunoblot analysis of the upstream molecular signaling pathways of the expression of p27 was performed using estrogen receptor (ER) and LKB1-double negative human MDA-MB-231 breast cancer cells in vitro. This analysis was performed without either transfecting the cells with various proximal 5'-upstream region of p27 gene-luciferase reporter plasmids or adding any growth factors to avoid the artificial stimulation of the cell proliferation.
In brief, the cells were seeded at a density of 5.5 × 106 cells/dish into a 100-mm tissue culture dish containing 10 mL of DMEM supplemented with 10% heat-inactivated fetal bovine serum (FBS), 2% L-glutamine, and antibiotic/antimycotic solution and incubated at 37°C in a 5% CO2 humidified chamber for 24 hours. After 24 hours, the cells were partially synchronized for another 24 hours in DMEM containing only 0.2% (v/v) of FBS. Subsequently, the resulting cells were cultured either (a) in the presence of DMSO, tamoxifen, or 4-hydroxytamoxifen in the regular DMEM with 0.2% FBS or (b) in the presence or absence of D-(+)-glucose, L-leucine, L-methionine, or L-cysteine in the appropriately supplemented basal DMEM Labeling Kit as described in the figure legends. After 24 hours, the cells were washed twice with cold 1× PBS and scraped in 1× RIPA Lysis Buffer (Santa Cruz Biotechnology, Santa Cruz, CA, USA) containing phenylmethylsulphonyl fluoride (PMSF), protease inhibitor cocktail, sodium orthovanadate and 50 mM NaF. The cells were then sonicated and the supernatant was collected by centrifugation and stored at -80°C.
The supernatants (60 μg protein/lane) were applied to the SDS-PAGE and, after fractionation, proteins were transferred to nitrocellulose membrane, which was then blocked and incubated in a solution containing first primary antibody. After shaking overnight at 4°C, the target proteins bound to the first primary antibody were further incubated in a solution containing alkaline phosphatase (AP)-conjugated secondary anti-immunoglobulin antibody and detected by chemiluminescence using TROPIX Western-Star Kit (Applied Biosystems, Foster City, CA, U.S.A.). After exposure to X-ray film, the blots were stripped using Western Re-Probe Solution (G-Biosciences, St. Louis, MO, U.S.A.), checked for removal of the chemiluminescence and then re-probed with second primary antibody.
Densitometric measurement of the intensity of the bands on the X-ray film was performed using UN-SCAN-IT Gel & Graph Digitizing Software Version 6.1 (Silk Scientific Corporation, Orem, UT, U.S.A.). Background corrections were done by four corner interpolation and optical density calculations were performed

Statistical Analysis

An experimental value with statistical significance of P ≤ 0.05 compared to the control by t test is indicated as a single asterisk on top of the vertical bar.

Acknowledgements

The author is grateful to Dr. Sakai (Kyoto Prefectural University of Medicine, Kyoto, Japan) for the gift of luciferase reporter plasmids -1797 p27Kip1 (p27-Kpn I) and -774 p27Kip1 (p27-Apa I) and Dr. Hengst (Max-Planck-Institut fur Biochemie, Martinsried bei Munchen, Germany) for the gift of luciferase reporter plasmid -575 p27Kip1 (p27-5'-UTR).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The author declares that they have no competing interests.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Literatur
1.
Zurück zum Zitat Eto I: Nutritional and chemopreventive anti-cancer agents up-regulate expression of p27Kip1, a cyclin-dependent kinase inhibitor, in mouse JB6 epidermal and human MCF7, MDA-MB-321 and AU565 breast cancer cells. Cancer Cell Int. 2006, 6: 20-10.1186/1475-2867-6-20. (pp. 1-19)PubMedCentralCrossRefPubMed Eto I: Nutritional and chemopreventive anti-cancer agents up-regulate expression of p27Kip1, a cyclin-dependent kinase inhibitor, in mouse JB6 epidermal and human MCF7, MDA-MB-321 and AU565 breast cancer cells. Cancer Cell Int. 2006, 6: 20-10.1186/1475-2867-6-20. (pp. 1-19)PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Eto I: Upstream molecular signaling pathways of p27(Kip1) expression: Effects of 4-hydroxytamoxifen, dexamethasone, and retinoic acids. Cancer Cell Int. 2010, 10: 3-10.1186/1475-2867-10-3. (pp. 1-19)PubMedCentralCrossRefPubMed Eto I: Upstream molecular signaling pathways of p27(Kip1) expression: Effects of 4-hydroxytamoxifen, dexamethasone, and retinoic acids. Cancer Cell Int. 2010, 10: 3-10.1186/1475-2867-10-3. (pp. 1-19)PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Eto I: G1 cell cycle regulatory proteins in chemically induced rat mammary adenocarcinomas in vivo and tumor promotion-sensitive, -resistant, and transformed mouse epidermal cells in vitro. Cell Cycle. 2003, 2: 149-156.CrossRefPubMed Eto I: G1 cell cycle regulatory proteins in chemically induced rat mammary adenocarcinomas in vivo and tumor promotion-sensitive, -resistant, and transformed mouse epidermal cells in vitro. Cell Cycle. 2003, 2: 149-156.CrossRefPubMed
4.
Zurück zum Zitat Goepfert U, Kullmann M, Hengst L: Cell cycle-dependent translation of p27 involves a responsive element in its 5'-UTR that overlaps with a uORF. Hum Mol Genet. 2003, 12: 1767-1779. 10.1093/hmg/ddg177.CrossRef Goepfert U, Kullmann M, Hengst L: Cell cycle-dependent translation of p27 involves a responsive element in its 5'-UTR that overlaps with a uORF. Hum Mol Genet. 2003, 12: 1767-1779. 10.1093/hmg/ddg177.CrossRef
5.
Zurück zum Zitat Agrawal D, Hauser P, McPherson F, Dong F, Garcia A, Pledger WJ: Repression of p27(kip1) synthesis by PDGF in balb/c 3T3 cells. Mol Cell Biol. 1996, 16: 4327-4336.PubMedCentralCrossRefPubMed Agrawal D, Hauser P, McPherson F, Dong F, Garcia A, Pledger WJ: Repression of p27(kip1) synthesis by PDGF in balb/c 3T3 cells. Mol Cell Biol. 1996, 16: 4327-4336.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Hengst L, Reed SI: Translational control of p27Kip1 accumulation during the cell cycle. Science. 1996, 271: 1861-1864. 10.1126/science.271.5257.1861.CrossRefPubMed Hengst L, Reed SI: Translational control of p27Kip1 accumulation during the cell cycle. Science. 1996, 271: 1861-1864. 10.1126/science.271.5257.1861.CrossRefPubMed
7.
Zurück zum Zitat Millard SS, Yan JS, Nguyen H, Pagano M, Kiyokawa H, Koff A: Enhanced ribosomal association of p27(Kip1) mRNA is a mechanism contributing to accumulation during growth arrest. J Biol Chem. 1997, 272: 7093-7098. 10.1074/jbc.272.11.7093.CrossRefPubMed Millard SS, Yan JS, Nguyen H, Pagano M, Kiyokawa H, Koff A: Enhanced ribosomal association of p27(Kip1) mRNA is a mechanism contributing to accumulation during growth arrest. J Biol Chem. 1997, 272: 7093-7098. 10.1074/jbc.272.11.7093.CrossRefPubMed
8.
Zurück zum Zitat Kullmann M, Goepfert U, Siewe B, Hengst L: ELAV/Hu proteins inhibit p27 translation via an IRES element in the p27 5'UTR. Genes Dev. 2002, 16: 3087-3099. 10.1101/gad.248902.PubMedCentralCrossRefPubMed Kullmann M, Goepfert U, Siewe B, Hengst L: ELAV/Hu proteins inhibit p27 translation via an IRES element in the p27 5'UTR. Genes Dev. 2002, 16: 3087-3099. 10.1101/gad.248902.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Miskimins WK, Wang G, Hawkinson M, Miskimins R: Control of cyclin-dependent kinase inhibitor p27 expression by cap-independent translation. Mol Cell Biol. 2001, 21: 4960-4967. 10.1128/MCB.21.15.4960-4967.2001.PubMedCentralCrossRefPubMed Miskimins WK, Wang G, Hawkinson M, Miskimins R: Control of cyclin-dependent kinase inhibitor p27 expression by cap-independent translation. Mol Cell Biol. 2001, 21: 4960-4967. 10.1128/MCB.21.15.4960-4967.2001.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Millard SS, Vidal A, Markus M, Koff A: A U-rich element in the 5' untranslated region is necessary for the translation of p27 mRNA. Mol Cell Biol. 2000, 20: 5947-5959. 10.1128/MCB.20.16.5947-5959.2000.PubMedCentralCrossRefPubMed Millard SS, Vidal A, Markus M, Koff A: A U-rich element in the 5' untranslated region is necessary for the translation of p27 mRNA. Mol Cell Biol. 2000, 20: 5947-5959. 10.1128/MCB.20.16.5947-5959.2000.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Pagano M, Tam SW, Theodoras AM, Beer-Romero P, Del Sal G, Chau V, Yew PR, Draetta GF, Rolfe M: Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 1995, 269: 682-685. 10.1126/science.7624798.CrossRefPubMed Pagano M, Tam SW, Theodoras AM, Beer-Romero P, Del Sal G, Chau V, Yew PR, Draetta GF, Rolfe M: Role of the ubiquitin-proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science. 1995, 269: 682-685. 10.1126/science.7624798.CrossRefPubMed
12.
Zurück zum Zitat Hara T, Kamura T, Nakayama K, Oshikawa K, Hatakeyama S: Degradation of p27(Kip1) at the G(0)-G(1) transition mediated by a Skp2-independent ubiquitination pathway. J Biol Chem. 2001, 276: 48937-48943. 10.1074/jbc.M107274200.CrossRefPubMed Hara T, Kamura T, Nakayama K, Oshikawa K, Hatakeyama S: Degradation of p27(Kip1) at the G(0)-G(1) transition mediated by a Skp2-independent ubiquitination pathway. J Biol Chem. 2001, 276: 48937-48943. 10.1074/jbc.M107274200.CrossRefPubMed
13.
Zurück zum Zitat Malek NP, Sundberg H, McGrew S, Nakayama K, Kyriakides TR, Roberts JM, Kyriakidis TR: A mouse knock-in model exposes sequential proteolytic pathways that regulate p27Kip1 in G1 and S phase. Nature. 2001, 413: 323-327. 10.1038/35095083.CrossRefPubMed Malek NP, Sundberg H, McGrew S, Nakayama K, Kyriakides TR, Roberts JM, Kyriakidis TR: A mouse knock-in model exposes sequential proteolytic pathways that regulate p27Kip1 in G1 and S phase. Nature. 2001, 413: 323-327. 10.1038/35095083.CrossRefPubMed
14.
Zurück zum Zitat Hengst L: A second RING to destroy p27 Kip1 . Nature Cell Biol. 2004, 6: 1153-1155. 10.1038/ncb1204-1153.CrossRefPubMed Hengst L: A second RING to destroy p27 Kip1 . Nature Cell Biol. 2004, 6: 1153-1155. 10.1038/ncb1204-1153.CrossRefPubMed
15.
Zurück zum Zitat Soos TJ, Kiyokawa H, Yan JS, Rubin MS, Giordano A, DeBlasio A, Bottega S, Wong B, Mendelsohn J, Koff A: Formation of p27-CDK complexes during the human mitotic cell cycle. Cell Growth Differ. 1996, 7: 135-146.PubMed Soos TJ, Kiyokawa H, Yan JS, Rubin MS, Giordano A, DeBlasio A, Bottega S, Wong B, Mendelsohn J, Koff A: Formation of p27-CDK complexes during the human mitotic cell cycle. Cell Growth Differ. 1996, 7: 135-146.PubMed
16.
Zurück zum Zitat Rodier G, Montagnoli A, Di Marcotullio L, Coulombe P, Draetta GF, Pagano M, Meloche S: p27 cytoplasmic localization is regulated by phosphorylation on Ser10 and is not a prerequisite for its proteolysis. EMBO J. 2001, 20: 6672-6682. 10.1093/emboj/20.23.6672.PubMedCentralCrossRefPubMed Rodier G, Montagnoli A, Di Marcotullio L, Coulombe P, Draetta GF, Pagano M, Meloche S: p27 cytoplasmic localization is regulated by phosphorylation on Ser10 and is not a prerequisite for its proteolysis. EMBO J. 2001, 20: 6672-6682. 10.1093/emboj/20.23.6672.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Viglietto G, Motti ML, Bruni P, Melillo RM, D'Alessio A, Califano D, Vinci F, Chiappetta G, Tsichlis P, Bellacosa A, Fusco A, Santoro M: Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kip1) by PKB/Akt-mediated phosphorylation in breast cancer. Nat Med. 2002, 8: 1136-1144. 10.1038/nm762.CrossRefPubMed Viglietto G, Motti ML, Bruni P, Melillo RM, D'Alessio A, Califano D, Vinci F, Chiappetta G, Tsichlis P, Bellacosa A, Fusco A, Santoro M: Cytoplasmic relocalization and inhibition of the cyclin-dependent kinase inhibitor p27(Kip1) by PKB/Akt-mediated phosphorylation in breast cancer. Nat Med. 2002, 8: 1136-1144. 10.1038/nm762.CrossRefPubMed
18.
Zurück zum Zitat Liang J, Zubovitz J, Petrocelli T, Kotchetkov R, Connor MK, Han K, Lee JH, Ciarallo S, Catzavelos C, Beniston R, Liang J, Zubovitz J, Petrocelli T, Kotchetkov R, Connor MK, Han K, Lee JH, Ciarallo S, Catzavelos C, Beniston R, Franssen E, Slingerland JM: PKB/Akt phosphorylates p27, impairs nuclear import of p27 and opposes p27-mediated G1 arrest. Nat Med. 2002, 8: 1153-1160. 10.1038/nm761.CrossRefPubMed Liang J, Zubovitz J, Petrocelli T, Kotchetkov R, Connor MK, Han K, Lee JH, Ciarallo S, Catzavelos C, Beniston R, Liang J, Zubovitz J, Petrocelli T, Kotchetkov R, Connor MK, Han K, Lee JH, Ciarallo S, Catzavelos C, Beniston R, Franssen E, Slingerland JM: PKB/Akt phosphorylates p27, impairs nuclear import of p27 and opposes p27-mediated G1 arrest. Nat Med. 2002, 8: 1153-1160. 10.1038/nm761.CrossRefPubMed
19.
Zurück zum Zitat Shin I, Yakes FM, Rojo F, Shin NY, Bakin AV, Baselga J, Arteaga CL: PKB/Akt mediates cell-cycle progression by phosphorylation of p27(Kip1) at threonine 157 and modulation of its cellular localization. Nat Med. 2002, 8: 1145-1152. 10.1038/nm759.CrossRefPubMed Shin I, Yakes FM, Rojo F, Shin NY, Bakin AV, Baselga J, Arteaga CL: PKB/Akt mediates cell-cycle progression by phosphorylation of p27(Kip1) at threonine 157 and modulation of its cellular localization. Nat Med. 2002, 8: 1145-1152. 10.1038/nm759.CrossRefPubMed
20.
Zurück zum Zitat Connor MK, Kotchetkov R, Cariou S, Resch A, Lupetti R, Beniston RG, Melchior F, Hengst L, Slingerland JM: CRM1/RAN-mediated nuclear export of p27 Kip1 involves a nuclear export signal and links p27 export and proteolysis. Mol Biol Cell. 2003, 14: 201-213. 10.1091/mbc.E02-06-0319.PubMedCentralCrossRefPubMed Connor MK, Kotchetkov R, Cariou S, Resch A, Lupetti R, Beniston RG, Melchior F, Hengst L, Slingerland JM: CRM1/RAN-mediated nuclear export of p27 Kip1 involves a nuclear export signal and links p27 export and proteolysis. Mol Biol Cell. 2003, 14: 201-213. 10.1091/mbc.E02-06-0319.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Ciarallo S, Subramanian V, Hung W, Lee JH, Kotchetkov R, Sandhu C, Milic A, Slingerland JM: Altered p27Kip1 phosphorylation, localization, and function in human epithelial cells resistant to transforming growth factor β-mediated G1 arrest. Mol Cell Biol. 2002, 22: 2993-3002. 10.1128/MCB.22.9.2993-3002.2002.PubMedCentralCrossRefPubMed Ciarallo S, Subramanian V, Hung W, Lee JH, Kotchetkov R, Sandhu C, Milic A, Slingerland JM: Altered p27Kip1 phosphorylation, localization, and function in human epithelial cells resistant to transforming growth factor β-mediated G1 arrest. Mol Cell Biol. 2002, 22: 2993-3002. 10.1128/MCB.22.9.2993-3002.2002.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Chu I, Sun J, Arnaout A, Kahn H, Hanna W, Narod S, Sun P, Tan CK, Hengst L, Slingerland J: p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2. Cell. 2007, 128: 281-294. 10.1016/j.cell.2006.11.049.PubMedCentralCrossRefPubMed Chu I, Sun J, Arnaout A, Kahn H, Hanna W, Narod S, Sun P, Tan CK, Hengst L, Slingerland J: p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2. Cell. 2007, 128: 281-294. 10.1016/j.cell.2006.11.049.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Kazi A, Carie A, Blaskovich MA, Bucher C, Thai V, Moulder S, Peng H, Carrico D, Pusateri E, J. Pledger WJ, Berndt N, Hamilton A, Sebti SM: Blockade of protein geranylgeranylation inhibits Cdk2-dependent p27Kip1 phosphorylation on Thr187 and accumulates p27Kip1 in the nucleus: Implications for breast cancer therapy. Mol Cell Biol. 2009, 29: 2254-2263. 10.1128/MCB.01029-08.PubMedCentralCrossRefPubMed Kazi A, Carie A, Blaskovich MA, Bucher C, Thai V, Moulder S, Peng H, Carrico D, Pusateri E, J. Pledger WJ, Berndt N, Hamilton A, Sebti SM: Blockade of protein geranylgeranylation inhibits Cdk2-dependent p27Kip1 phosphorylation on Thr187 and accumulates p27Kip1 in the nucleus: Implications for breast cancer therapy. Mol Cell Biol. 2009, 29: 2254-2263. 10.1128/MCB.01029-08.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Law IKM, Liu L, Xu A, Lam KSL, Vanhoutte PM, Che CM, Leung PTY, Wang Y: Identification and characterization of proteins interacting with SIRT1 and SIRT3: implications in the anti-aging and metabolic effects of sirtuins. Proteomics. 2009, 9: 2444-2456. 10.1002/pmic.200800738.CrossRefPubMed Law IKM, Liu L, Xu A, Lam KSL, Vanhoutte PM, Che CM, Leung PTY, Wang Y: Identification and characterization of proteins interacting with SIRT1 and SIRT3: implications in the anti-aging and metabolic effects of sirtuins. Proteomics. 2009, 9: 2444-2456. 10.1002/pmic.200800738.CrossRefPubMed
25.
Zurück zum Zitat Laplante M, Sabatini DM: mTORC1 activates SREBP-1c and uncouples lipogenesis from gluconeogenesis. Proc Natl Acad Sci US. 2010, 107: 3281-3282. 10.1073/pnas.1000323107.CrossRef Laplante M, Sabatini DM: mTORC1 activates SREBP-1c and uncouples lipogenesis from gluconeogenesis. Proc Natl Acad Sci US. 2010, 107: 3281-3282. 10.1073/pnas.1000323107.CrossRef
26.
Zurück zum Zitat Sengupta S, Peterson TR, Sabatini DM: Regulation of the mTOR Complex 1 Pathway by Nutrients, Growth Factors, and Stress. Molecular Cell. 2010, 40: 310-322. 10.1016/j.molcel.2010.09.026.PubMedCentralCrossRefPubMed Sengupta S, Peterson TR, Sabatini DM: Regulation of the mTOR Complex 1 Pathway by Nutrients, Growth Factors, and Stress. Molecular Cell. 2010, 40: 310-322. 10.1016/j.molcel.2010.09.026.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Düvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD: Activation of a metabolic gene regulatory network downstream of mTOR complex. Molecular Cell. 2010, 39: 171-183. 10.1016/j.molcel.2010.06.022.PubMedCentralCrossRefPubMed Düvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD: Activation of a metabolic gene regulatory network downstream of mTOR complex. Molecular Cell. 2010, 39: 171-183. 10.1016/j.molcel.2010.06.022.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Porstmann T, Santos CR, Griffiths B, Cully M, Wu M, Leevers S, Griffiths JR, Chung YL, Schulze A: SREBP activity Is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008, 8: 224-236. 10.1016/j.cmet.2008.07.007.PubMedCentralCrossRefPubMed Porstmann T, Santos CR, Griffiths B, Cully M, Wu M, Leevers S, Griffiths JR, Chung YL, Schulze A: SREBP activity Is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 2008, 8: 224-236. 10.1016/j.cmet.2008.07.007.PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Naidoo N, Zhang L, Romer M, Cater JR, Scharf MT, Raymond J. Galante RJ, Pack AI: Changes in components of energy regulation in mouse cortex with Increases in wakefulness. SLEEP. 2010, 33: 889-900.PubMedCentralPubMed Naidoo N, Zhang L, Romer M, Cater JR, Scharf MT, Raymond J. Galante RJ, Pack AI: Changes in components of energy regulation in mouse cortex with Increases in wakefulness. SLEEP. 2010, 33: 889-900.PubMedCentralPubMed
30.
Zurück zum Zitat Finley LWS, Carracedo A, Lee J, Souza A, Egia A, Zhang J, Teruya-Feldstein J, Moreira PI, Cardoso SM, Clish CB, Pandolf PP, Haigis MC: SIRT3 opposes reprogramming of cancer cell metabolism through HIF1a destabilization. Cancer Cell. 2011, 19: 416-428. 10.1016/j.ccr.2011.02.014.PubMedCentralCrossRefPubMed Finley LWS, Carracedo A, Lee J, Souza A, Egia A, Zhang J, Teruya-Feldstein J, Moreira PI, Cardoso SM, Clish CB, Pandolf PP, Haigis MC: SIRT3 opposes reprogramming of cancer cell metabolism through HIF1a destabilization. Cancer Cell. 2011, 19: 416-428. 10.1016/j.ccr.2011.02.014.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Giralt A, Hondares E, Villena JA, Ribas F, Díaz-Delfín J, Giralt M, Iglesias R, Villarroya F: Peroxisome proliferator-activated receptor-γ coactivator-1α controls transcription of the Sirt3 gene, an essential component of the thermogenic brown adipocyte phenotype. J Biol Chem. 2011, 286: 16958-16966. 10.1074/jbc.M110.202390.PubMedCentralCrossRefPubMed Giralt A, Hondares E, Villena JA, Ribas F, Díaz-Delfín J, Giralt M, Iglesias R, Villarroya F: Peroxisome proliferator-activated receptor-γ coactivator-1α controls transcription of the Sirt3 gene, an essential component of the thermogenic brown adipocyte phenotype. J Biol Chem. 2011, 286: 16958-16966. 10.1074/jbc.M110.202390.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Kim HS, Patel K, Muldoon-Jacobs K, Bisht KS, Aykin-Burns NA, Pennington JD, van der Meer R, Nguyen P, Savage J, Owens KM, Vassilopoulos A, Ozden O, Park SH, Singh KK, Sarki A. Abdulkadir SA, Douglas R. Spitz DR, Chu-Xia Deng CX, Gius D: SIRT3 Is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell. 2010, 17: 41-52. 10.1016/j.ccr.2009.11.023.PubMedCentralCrossRefPubMed Kim HS, Patel K, Muldoon-Jacobs K, Bisht KS, Aykin-Burns NA, Pennington JD, van der Meer R, Nguyen P, Savage J, Owens KM, Vassilopoulos A, Ozden O, Park SH, Singh KK, Sarki A. Abdulkadir SA, Douglas R. Spitz DR, Chu-Xia Deng CX, Gius D: SIRT3 Is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell. 2010, 17: 41-52. 10.1016/j.ccr.2009.11.023.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang Y: Sirtuin 3, a new target of PGC-1a, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE. 2010, 5: e11707-10.1371/journal.pone.0011707.PubMedCentralCrossRefPubMed Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang Y: Sirtuin 3, a new target of PGC-1a, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE. 2010, 5: e11707-10.1371/journal.pone.0011707.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Liu Y, Zhang D, Chen D: SIRT3: Striking at the heart of aging. Aging. 2010, 12: 914-923. Liu Y, Zhang D, Chen D: SIRT3: Striking at the heart of aging. Aging. 2010, 12: 914-923.
35.
Zurück zum Zitat Choudhury M, Jonscher KR, Friedman JE: Reduced mitochondrial function in obesity-associated fatty liver: SIRT3 takes on the fat. AGING. 2011, 3: 175-178.PubMedCentralPubMed Choudhury M, Jonscher KR, Friedman JE: Reduced mitochondrial function in obesity-associated fatty liver: SIRT3 takes on the fat. AGING. 2011, 3: 175-178.PubMedCentralPubMed
36.
Zurück zum Zitat Huang JY, Hirschey MD, Shimazu T, Ho L, Verdin E: Mitochondrial sirtuins. Biochimica et Biophysica Acta (BBA) - Proteins & Proteomics. 2010, 1804: 1645-1651. 10.1016/j.bbapap.2009.12.021.CrossRef Huang JY, Hirschey MD, Shimazu T, Ho L, Verdin E: Mitochondrial sirtuins. Biochimica et Biophysica Acta (BBA) - Proteins & Proteomics. 2010, 1804: 1645-1651. 10.1016/j.bbapap.2009.12.021.CrossRef
37.
Zurück zum Zitat Palacios OM, Carmona JJ, Michan S, Ke KY, Manabe Y, Ward III, Goodyear LJ, Tong Q: Diet and exercise signals regulate SIRT3 and activate AMPK and PGC-1α in skeletal muscle. AGING. 2009, 1: 771-783.PubMedCentralPubMed Palacios OM, Carmona JJ, Michan S, Ke KY, Manabe Y, Ward III, Goodyear LJ, Tong Q: Diet and exercise signals regulate SIRT3 and activate AMPK and PGC-1α in skeletal muscle. AGING. 2009, 1: 771-783.PubMedCentralPubMed
38.
Zurück zum Zitat Minami S, Ohtani-Fujita N, Igata E, Tamaki T, Sakai T: Molecular cloning and characterization of the human p27Kip1 gene promoter. FEBS Lett. 1997, 411: 1-6. 10.1016/S0014-5793(97)00660-1.CrossRefPubMed Minami S, Ohtani-Fujita N, Igata E, Tamaki T, Sakai T: Molecular cloning and characterization of the human p27Kip1 gene promoter. FEBS Lett. 1997, 411: 1-6. 10.1016/S0014-5793(97)00660-1.CrossRefPubMed
39.
Zurück zum Zitat Hsu TC, Nair R, Tulsian P, Camalier CE, Hegamyer GA, Young MR, Colburn NH: Transformation nonresponsive cells owe their resistance to lack of p65/nuclear factor-κB activation. Cancer Res. 2001, 61: 4160-4168.PubMed Hsu TC, Nair R, Tulsian P, Camalier CE, Hegamyer GA, Young MR, Colburn NH: Transformation nonresponsive cells owe their resistance to lack of p65/nuclear factor-κB activation. Cancer Res. 2001, 61: 4160-4168.PubMed
40.
Zurück zum Zitat Vidal A, S. Millard S, Miller JP, Koff A: Rho activity can alter the translation of p27 mRNA and Is important for RasV12-induced transformation in a manner dependent on p27 status. J Biol Chem. 2002, 277: 16433-16440. 10.1074/jbc.M112090200.CrossRefPubMed Vidal A, S. Millard S, Miller JP, Koff A: Rho activity can alter the translation of p27 mRNA and Is important for RasV12-induced transformation in a manner dependent on p27 status. J Biol Chem. 2002, 277: 16433-16440. 10.1074/jbc.M112090200.CrossRefPubMed
Metadaten
Titel
Upstream molecular signaling pathways of p27 (Kip1) expression in human breast cancer cells in vitro: differential effects of 4-hydroxytamoxifen and deficiency of either D-(+)-glucose or L-leucine
verfasst von
Isao Eto
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2011
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/1475-2867-11-31

Weitere Artikel der Ausgabe 1/2011

Cancer Cell International 1/2011 Zur Ausgabe

Update Onkologie

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