You are on page 1of 10

Oncogene

https://doi.org/10.1038/s41388-019-0751-4

ARTICLE

Mitochondrial GPT2 plays a pivotal role in metabolic adaptation to


the perturbation of mitochondrial glutamine metabolism
1,2,3
Minjoong Kim ●
Jihye Gwak1,2,3 Sunsook Hwang1,2,3 Seungyeon Yang1,2,3 Seung Min Jeong1,2,3
● ● ●

Received: 19 October 2018 / Revised: 11 January 2019 / Accepted: 31 January 2019


© Springer Nature Limited 2019

Abstract
Cancer cells exhibit metabolic dependence on mitochondrial glutamine metabolism that provides them with the substrates
required for rapid proliferation. Despite the extensive efforts to target this glutamine addiction for therapeutic purposes, the
adaptive metabolic responses and the mechanisms whereby cells maintain their unlimited growth remain areas of active
investigation. Here we report that mitochondrial glutamate-pyruvate transaminase 2 (GPT2) contributes to cell survival and
growth by sustaining the tricarboxylic acid (TCA) cycle anaplerosis after the inhibition of glutaminase (GLS), the first
enzyme for mitochondrial glutamine metabolism. We found that elevated reactive oxygen species upon GLS inhibition
1234567890();,:
1234567890();,:

induce GPT2 expression via activating transcription factor 4. Moreover, inhibition of GPT2 synergized with suppression of
GLS activity to induce a pronounced reduction in proliferation and an increase in cell death of cancer cells. Our data uncover
GPT2 as an important component of the adaptive metabolic response for glutamine deprivation and indicate that targeting
this pathway in combination with GLS inhibition may be an effective therapeutic approach for cancer treatment.

Introduction and lipids for their unlimited proliferation [3]. However,


continuous use of TCA cycle intermediates would lead to
The dysregulation of cell metabolism is a defining factor of the loss of mitochondrial functions and limit the pro-
many cancers [1]. For example, cancer cells undergo gly- liferative capacity of cells. Accruing evidence suggests that
colysis even in the presence of ample oxygen and this glutamine (Gln) provides a carbon source to refill the TCA
preferential use of aerobic glycolysis has emerged as a cycle, which is critical for the maintenance of mitochondrial
hallmark of cancer [2]. Cancer cells also use precursors integrity, cell growth and survival [3, 4].
derived from the tricarboxylic acid (TCA) cycle to produce In mitochondria, Gln is converted to glutamate via glu-
biomass building blocks including amino acids, nucleotides, taminase (GLS) and further metabolized to the TCA cycle
intermediate α-ketoglutarate (αKG) via glutamate dehy-
drogenase 1 (GLUD1) or transaminases such as glutamate-
oxaloacetate transaminase 2 (GOT2) or glutamate-pyruvate
Supplementary information The online version of this article (https:// transaminase 2 (GPT2). The incorporation of αKG into the
doi.org/10.1038/s41388-019-0751-4) contains supplementary TCA cycle through Gln metabolism is the major anaplerotic
material, which is available to authorized users. step in proliferating cells [5]. Recent studies have demon-
* Seung Min Jeong
strated that mitochondrial Gln metabolism is also an
smjeong@catholic.ac.kr important regulator of cellular redox state and thus its
defects lead to the generation of reactive oxygen species
1
Department of Biochemistry, College of Medicine, The Catholic (ROS) [6–8].
University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591,
Given the importance of mitochondrial Gln metabolism
Republic of Korea
2
to support anabolic processes that fuel cell proliferation, it is
Department of Biomedicine & Health Sciences, College of
not surprising that many cancer cells are addicted to Gln
Medicine, The Catholic University of Korea, 222, Banpo-daero,
Seocho-gu, Seoul 06591, Republic of Korea and exhibit an increased Gln anaplerosis and high GLS
3 activity [3, 5]. As GLS is the most proximal enzyme in
Institute for Aging and Metabolic Diseases, College of Medicine,
The Catholic University of Korea, 222, Banpo-daero, Seocho-gu,
Seoul 06591, Republic of Korea
M. Kim et al.

mitochondrial Gln metabolism, drugs targeting GLS are Mitochondrial GPT2 is required for cell growth and
being developed and tested for cancer treatment [9, 10]. survival upon GLS inhibition
However, these drugs suffer from poor bioavailability and
cytotoxicity at higher doses [9]. Moreover, many cancer Given the significant induction of GPT2 upon GLS inhi-
cells retain the capacity for metabolic adaptation to sustain bition, we speculated that GPT2 may be required for cell
cell growth and survival periods of impaired Gln metabo- growth and/or survival when mitochondrial Gln metabolism
lism by inducing alternative metabolic pathways. For is impaired. To test this concept, we inhibited GPT2 by
example, Gln withdrawal activates serine biosynthesis treating cells with aminooxyacetate (AOA), a transaminase
pathway, which is important for cell survival by facilitating inhibitor [16], and examined whether this would affect cell
glutathione (GSH) production and nucleic acid synthesis growth after BPTES treatment. First, we found that BPTES
[11, 12]. It has been also shown that asparagine synthetase or AOA treatment alone had a mild growth-suppressive
(ASNS) suppresses Gln deprivation-induced cell death by effect (Fig. 2a, b and Supplementary Fig. 2A, B).
maintaining intracellular asparagine (Asn) levels [13–15]. Remarkably, when these two inhibitors are combined, this
However, the adaptive molecular events that maintain Gln effect was notably augmented, indicating that cells are
anaplerosis upon GLS inhibition are still unclear. markedly more sensitive to GLS inhibition when GPT2
In this study, we describe a role of mitochondrial GPT2 activity is suppressed (Fig. 2a, b and Supplementary Fig.
in the regulation of cell growth and survival after the 2A, B). When we measured cell viability and clonogenic
inhibition of mitochondrial Gln metabolism, and provide growth, similar synergistic effects were observed (Fig. 2c, d
evidence that targeting this compensatory response may be and Supplementary Fig. 2C, D). Near-identical results were
an important therapeutic strategy for multiple cancer cells. also observed with CB-839 or with another GPT2 inhibitor,
cycloserine (Supplementary Fig. 2E, F) [17]. Consistent
with these results, GLS knockdown also significantly atte-
Results nuated AOA treated cell viability (Fig. 2e). As further
confirmation of the importance of GPT2 upon GLS inhi-
Mitochondrial GPT2 is induced by Gln deprivation bition, we suppressed GPT2, GLUD1 and GOT2 expression
using shRNAs and assessed cell viability under GLS
Gln, the most abundant amino acid in the body, plays an inhibited conditions. In line with our hypothesis, GLUD1 or
essential role for cell proliferation [4]. To investigate how GOT2 knockdown had no effect on cell viability (Supple-
cells might cope with impaired Gln metabolism, we first mentary Fig. 2G, H). In contrast, GPT2 knockdown
examined the expression of metabolic enzymes which are robustly diminished cell viability, when mitochondrial Gln
involved in mitochondrial Gln metabolism after Gln metabolism is repressed by BPTES treatment (Fig. 2f).
deprivation. We found that GPT2 messenger RNA (mRNA) Mitochondrial Gln metabolism refuels the TCA cycle,
and protein levels were highly induced upon Gln with- which is essential for cell survival [4]. Thus, we next
drawal, whereas the expressions of other mitochondrial Gln explored the role of GPT2 on cell survival. We found that
metabolism-related enzymes were not changed (Fig. 1a–c AOA treatment or knockdown of GPT2 significantly
and Supplementary Fig. 1A, B). Notably, we observed no induced cell death, when GLS activity is inhibited by
obvious change in GPT2 expression under glucose-deprived BPTES treatment (Fig. 2g, h). Similarly, we found that
conditions (Fig. 1b, c and Supplementary Fig. 1A), indi- GPT2 impaired cells exhibited significantly increased cell
cating that the induction of GPT2 is a specific response death in Gln-free media compared to control cells (Sup-
under Gln-starved conditions and not an indirect con- plementary Fig. 2I, J). Together, these data demonstrate that
sequence of the metabolic crisis. In mitochondria, Gln is GPT2 contributes to cell growth and survival upon GLS
catabolized into αKG to replenish the TCA cycle. When inhibition.
mitochondrial Gln metabolism was impaired by adding bis-
2-(5-phenylacetoamido-1,2,4-thiadiazol-2-yl)ethylsulfide ATF4 regulates the transcription of GPT2 upon GLS
(BPTES), an inhibitor of GLS [9], we found that GPT2 was inhibition
highly induced (Fig. 1d, e and Supplementary Fig. 1C). We
also observed a similar induction of GPT2 with another We next examined the mechanisms underlying increased
GLS inhibitor, CB-839 (Supplementary Fig. 1D) [10]. GPT2 expression after GLS inhibition. Given the essential
Moreover, GLS knockdown using short hairpin RNAs role of mitochondrial Gln metabolism in maintaining the
(shRNAs) also markedly increased GPT2 expression (Fig. cellular redox homeostasis [6], we speculated that elevated
1f, g). These data hint that GPT2 may have an important, ROS production by GLS inhibition may affect this adaptive
previously undetermined role when mitochondrial Gln metabolic response. Indeed, cellular ROS production was
metabolism is suppressed. significantly induced by Gln deprivation or GLS inhibition
Mitochondrial GPT2 plays a pivotal role in metabolic adaptation to the perturbation of mitochondrial. . .

A B
***

Relative mRNA expression levels


5 Control 6
***

Relative GPT2 mRNA levels


-Gln 12h
-Gln 24h
4

4
3

2
2
1

0 0
GLS1 GLUD1 GOT2 GPT2
Glc + + -
Gln + - +
C D E
3
**

Relative GPT2 mRNA levels GPT2


GPT2 2

β-actin β-actin

Glc + + - 1
BPTES - +

Gln + - +

0
BPTES - +

F G
*
Relative mRNA expression levels

3 shGFP
shGLS1 #1 ***
shGLS1 #2
GLS1
2
GPT2

β-actin
1
shRNA GFP GLS1 #1 GLS1 #2

0
GLS1 GPT2

Fig. 1 Glutamine starvation or GLS inhibition induces expression of d, e Relative GPT2 mRNA (d) and protein (e) levels in HeLa cells
GPT2. a Relative mRNA levels of GLS1, GLUD1, GOT2, and GPT2 treated with BPTES (10 μM) for 24 h. f, g Relative mRNA (f) and
in HeLa cells incubated with or without Gln for 12 or 24 h (n = 3). β- protein (g) levels of GLS1 and GPT2 in HeLa cells expressing a
actin was used as an endogenous control for qRT-PCR. b, c Relative control shRNA or two independent shRNAs to GLS1. All error bars,
GPT2 mRNA (b) and protein (c) levels in HeLa cells incubated with or ± SEM. *p < 0.05, **p < 0.01 and ***p < 0.001
without Gln or Glc for 12 h. β-actin serves as a loading control.

(Supplementary Fig. 3A). Thus, we first treated cells with significantly reduced GPT2 expression (Fig. 3a, b). NAC
the antioxidant N-acetylcysteine (NAC) in order to probe also inhibited GPT2 induction by Gln withdrawal (Fig. 3c
the model that suppressing ROS could block the induction and Supplementary Fig. 3B). To directly test whether GLS
of GPT2 upon GLS inhibition. Indeed, whereas GLS- inhibition increases GPT2 gene transcription through ROS,
impaired cells had higher levels of GPT2, NAC treatment we cloned the human GPT2 promoter region into a
M. Kim et al.

A B C
15 DM SO 1.2
BPTES
AOA
***

Relative cell viability


Cell number x 104 BPTES + AOA
10 0.8

***
5 0.4

0 0.0
Day 0 Day 1 Day 2 Day 3 Day 4
BPTES - + - +
AOA - - + +
D E F
1.8 1.2 *** 1.2 ***
*** ***
***
Relative colony number

Relative cell viability


Relative cell viability

1.2 0.8 0.8

0.6 0.4 0.4

0.0 0.0 0.0


BPTES - + - + AOA - + - + - + BPTES - + - + - +
AOA - - + + shGFP shGLS1 #1 shGLS1 #2 shGFP shGPT2 #1 shGPT2 #2

GLS1 GPT2

β-actin β-actin

shRNA GFP GLS1 #1 GLS1 #2 shRNA GFP GPT2 #1 GPT2 #2

G H ***
40 30 shGFP
*** shGPT2 #1
shGPT2 #2
30
Cell death (%)
Cell death (%)

20

20

10
10

0 0
DMSO BPTES
BPTES - + - +
AOA - - + +
Fig. 2 GPT2 knockdown sensitizes cells to the inhibition of mito- shRNAs to GLS1 treated with or without AOA (top) (n = 4). Western
chondrial Gln metabolism. a Growth curves of HeLa cells treated with blot confirmed the knockdown of GLS1 expression (bottom). f Rela-
or without BPTES and/or AOA (125 μM). Statistical differences were tive cell viability of HeLa cells expressing a control shRNA or two
determined using a two-way ANOVA. Data represent the mean ± SD independent shRNAs to GPT2 treated with or without BPTES (top)
(n = 3). b HeLa cells were treated with or without BPTES and/or AOA (n = 4). Western blot confirmed the knockdown of GPT2 expression
as indicated for 72 h. Phase microscopy was used to observe cell (bottom). g Survival of HeLa cells cultured with or without BPTES
proliferation. c Relative cell viability of HeLa cells treated with or and/or AOA for 72 h (n = 3). h Survival of HeLa cells expressing a
without BPTES (5 μM) and/or AOA for 72 h (n = 4). d Relative clo- control shRNA or two independent shRNAs to GPT2 incubated with
nogenic growth of HeLa cells treated with or without BPTES and/or or without BPTES for 72 h (n = 3). All error bars (except growth
AOA. The number of colonies was counted (n = 3). e Relative cell curves), ± SEM. ***p < 0.001
viability of HeLa cells expressing a control shRNA or two independent
Mitochondrial GPT2 plays a pivotal role in metabolic adaptation to the perturbation of mitochondrial. . .

luciferase reporter construct. We found that the increased A B


5

Relative GPT2 mRNA levels


GPT2 promoter activity upon GLS inhibition was restored **
4
by NAC treatment (Fig. 3d). Similar results were observed
under Gln-free conditions (Fig. 3e). Thus, these data 3
GPT2

demonstrate that increased ROS production by GLS inhi- β-actin


2
bition induces GPT2 expression.
BPTES - - + +
As GLS inhibition positively regulates the transcription 1
NAC - + - +
of GPT2, we next attempted to identify the transcription
0
factor that might be responsible for GPT2 induction. Recent
BPTES - - + +
studies reported that activating transcription factor 4 (ATF4) NAC - + - +
is a critical regulator of genes involved in amino acid
metabolism including GPT2 and an enhanced ROS gen- C D
15 4

Relative GPT2 mRNA levels


eration upregulates ATF4 expression [18–21]. Thus, we

Relative GPT2 Luc activity


** ***
reasoned that ATF4 may regulate GPT2 transcription upon 3
10
GLS inhibition. First, we observed that GLS inhibition
resulted in a marked increase in ATF4 expression at the 2

transcriptional level, which was completely reverse by NAC 5


treatment (Fig. 3f). Next, we found that mutating a motif 1

matching ATF4 consensus-binding site, located in a GPT2


0 0
promoter region (Supplementary Fig. 3C), significantly
Gln + + - - BPTES - - + +
abrogated BPTES or Gln deprivation-induced increase of NAC - + - + NAC - + - +
GPT2 promoter activity (Fig. 3g, h). Taken together, these
data demonstrate that an increased ROS production caused E F
10 ** 4

Relative ATF4 mRNA levels


Relative GPT2 Luc activity

by GLS inhibition induces GPT2 expression via ATF4. ***


8
3
Maintaining TCA cycle anaplerosis by GPT2 supports 6
cell survival during GLS inhibition 2
4

Given the important role of GPT2 in converting glutamate 1


2
into αKG to support mitochondrial carbon pool, we
0 0
speculated that GPT2-mediated TCA cycle anaplerosis
Gln + + - - BPTES - - + +
could support cell growth and survival upon GLS inhibi- NAC - + - + NAC - + - +
tion. To test this concept, we treated cells with membrane
permeable dimethyl-αKG (DMKG) and examined whether G H
5 10
***
this would affect cell growth and survival of GPT2 impaired
Relative GPT2 Luc activity

Relative GPT2 Luc activity

***
4 8
cells upon GLS inhibition. Importantly, DMKG supplement
almost completely restored decreased viability and increase 3 6
cell death in GLS inhibited or Gln-deprived cells upon
2 4
AOA treatment (Fig. 4a, b and Supplementary Fig. 4A–C).
Consistent with these results, DMKG treatment also 1 2
recovered cell survival upon GPT2 knockdown (Fig. 4c).
0 0
Similar results were observed when we treated another TCA BPTES - + - + Gln + - + -
cycle intermediate, oxaloacetate (OAA) (Supplementary WT Mut WT Mut

Fig. 4D), demonstrating that GPT2 is required for sustain-


ing TCA cycle anaplerosis upon GLS inhibition.
Several groups have provided evidence that the addition As GPT2 converts glutamate to αKG, glutamate sup-
of Asn rescued cell viability in the absence of Gln [13, 15]. plement is necessary for GPT2 function. Recently, it has
However, we found that Asn supplement failed to restore been reported that the re-accumulation of glutamate by
cell survival when GPT2 activity is suppressed (Supple- alternative metabolic pathways after GLS inhibition [22].
mentary Fig. 4E). Additionally, we found that alanine (Ala), Indeed, we found that ASNS, which can produce glutamate
another byproduct of GPT2, or antioxidants treatment had in a GLS independent manner, is highly elevated upon GLS
minor effect on cell survival under these conditions (Fig. 4d inhibition (Fig. 4e). Moreover, non-Gln derived glutamate
and Supplementary Fig. 4F–H). producing enzymes, including branched chain
M. Kim et al.

Fig. 3 Mitochondrial Gln metabolism inhibition induces GPT2 via Discussion


ROS-ATF4 pathway. a, b Relative GPT2 mRNA (a) and protein (b)
levels in HeLa cells treated with or without BPTES and/or NAC (5
In this study, we demonstrate a novel dependence on
mM) for 24 h. c Relative GPT2 mRNA levels in HeLa cells treated
with or without Gln and/or NAC for 24 h (n = 3). d, e GPT2 promoter mitochondrial GPT2 for cell survival and growth after the
luciferase activity. HeLa cells were co-transfected with the pGL3- repression of mitochondrial Gln metabolism. We dis-
GPT2 promoter and Renilla reporter constructs. Renilla luciferase covered that Gln deprivation or GLS inhibition-mediated
activity was measured to normalize the transfection efficiency. Luci-
ROS production induces the transcription of GPT2 via
ferase activity was measured in cells treated with or without BPTES
and/or NAC (d), or with or without Gln and/or NAC (e) for 24 h (n = ATF4 (Fig. 5e). Significantly, the induction of GPT2 is
3). f Relative ATF4 mRNA levels in HeLa cells treated with or without required for supplying cellular αKG upon GLS inhibition,
BPTES and/or NAC for 24 h (n = 3). g, h HeLa cells were co- highlighting the potential importance of GPT2-mediated
transfected with ATF4-binding-site-intact (WT) or ATF4-binding-site-
adaptive metabolic response in cell survival and growth.
mutated (Mut) GPT2 promoter. Luciferase activity was measured in
cells incubated with or without BPTES (g), and with or without Gln This idea is further validated by the finding that GPT2
(h) for 24 h (n = 3). All error bars, ± SEM. **p < 0.01 and ***p < inhibition sensitizes cancer cells to the repression of mito-
0.001 chondrial Gln metabolism.
Many therapeutic approaches have been developed to
target metabolic pathways of cancer cells [1]. As GLS is the
aminotransferase 1 (BCAT1), Gamma-Glutamyl hydrolase most proximal enzyme in mitochondrial Gln metabolism,
(GGH) and 5-oxoprolinase (OPLAH), were also increased GLS inhibition has been used to treat cancers [9, 10].
in these cells (Fig. 4e). Furthermore, BPTES treatment had However, cancer cells have the abilities to adapt to meta-
only a modest impact on intracellular glutamate levels, bolic challenges by activating alternative metabolic path-
indicating that alternate metabolic pathways can recover ways or by scavenging for fuel sources [15, 22, 23]. Indeed,
cellular glutamate upon GLS inhibition (Fig. 4f). Taken we found that chronic GLS inhibition had a minor growth-
together, these results demonstrate the essentiality of GPT2 suppressive effect on several cancer cells. We demonstrate
in cell survival by supplying the TCA cycle anaplerosis that GPT2 is intimately implicated in adaptive metabolic
upon GLS inhibition. responses to exposure of GLS inhibitors. Importantly, AOA
treatment even at low doses significantly sensitizes cells to
Inhibition of GPT2 synergizes with GLS inhibitors to GLS inhibition-induced growth arrest and cell death. This
suppress cancer cell survival finding may have important therapeutic implications, given
that its cellular toxicity at higher doses limits the clinical use
Our data indicate that GPT2 has an important role in the of AOA [4]. The essentiality of GPT2 on cell survival upon
adaptive metabolic response to GLS inhibition. Until GLS inhibition and the fact that GLUD1 and GOT2 are
recently, drugs targeting GLS are being developed for dispensable under these conditions may provide potential
cancer therapy [9, 10]. However, poor bioavailability and therapeutic strategies for cancer treatment.
low potency mitigate its use as a viable therapeutic strategy Our studies reveal the important impact of a mitochon-
[22]. Thus, these observations suggest that inhibition of drial transaminase GPT2 on cell survival and proliferation
GPT2 may have therapeutic implications by sensitizing by supplying αKG in GLS-impaired cells. It has been
cancer cells to GLS inhibition. To test this idea, we inhib- shown that a transaminase GOT2 generates aspartate,
ited GPT2 activity and examined whether this would allowing Kras-driven cancer cells to avoid cell cycle arrest
synergize with BPTES treatment in multiple cancer cell upon Gln starvation by producing nucleotides [24].
lines including MiaPaCa2, HCT116, A549 and MCF-7. Although GOT2 also can fulfill TCA cycle by producing
Although each cancer line exhibited different sensitivity to αKG, it consumes OAA, another TCA cycle intermediate,
either AOA or BPTES, the combinational treatment pro- to convert glutamate to αKG. Thus, cells may prefer to use
duces synthetic lethality in all tested cancer cell lines (Fig. pyruvate to produce αKG via GPT2 upon GLS inhibition.
5a–d). To further confirm the importance of GPT2 activity In support of this idea, we show that GPT2, but not GOT2,
upon GLS inhibition in tumorigenic properties of cancer is markedly induced in GLS-impaired cells (Fig. 1).
cells, we assessed their clonogenic growth. As expected, the GLUD1, which produces αKG without the need for other
treatment with AOA potently synergized with BPTES to amino acids, has been shown to be required for cell survival
diminish the clonogenic properties of cancer cells when glucose is scarce [25]. Thus, it will be interesting for
(Supplementary Fig. 5A–D). In conclusion, our results future work to investigate whether the relative abundance of
demonstrate that the combination treatments aimed at pyruvate is a determinant of these metabolic choices.
inhibiting GLS and GPT2 potently suppress cancer cell Recent studies have shed light on the mechanisms
survival. through which mitochondrial Gln metabolism regulates
Mitochondrial GPT2 plays a pivotal role in metabolic adaptation to the perturbation of mitochondrial. . .

A B
1.2 50

***
40
Relative cell viability ***

Cell death (%)


0.8
30

20
0.4

10

0.0 0
BPTES - + - + + BPTES - + - + +
AOA - - + + + AOA - - + + +
DM KG - - - - + DM KG - - - - +

C ***
D
30 DM SO 1.2
BPTES
BPTES + DM KG

Relative cell viability


**
Cell death (%)

20 0.8

10 0.4
**
ns

0 0.0
shGFP shGPT2 #1 shGPT2 #2
BPTES - + - + + +
AOA - - + + + +
NAC - - - - - +
Ala - - - - + -

E F ns
1.2 ns
Relative mRNA expression levels

15 DM SO
*** BPTES
Relative glutamate levels

10 0.8

5 0.4
**
ns *

0 0.0
ASNS BCAT1 GGH OPLAH
BPTES 0 5 10
(μM )

Fig. 4 αKG rescue cell survival treated with BPTES and AOA. a 3). d Relative cell viability of HeLa cells treated with BPTES, AOA,
Relative cell viability of HeLa cells treated with BPTES, AOA and/or NAC and/or Ala (4 mM) (n = 4). e Relative mRNA levels of ASNS,
DMKG (4 mM) for 72 h (n = 4). b Survival of HeLa cells cultured BCAT1, GGH and OPLAH in HeLa cells treated with or without
with BPTES, AOA and/or DMKG for 72 h (n = 3). c Survival of HeLa BPTES for 24 h (n = 3). f Relative Glu levels in HeLa cells treated
cells expressing a control shRNA or two independent shRNAs to with or without BPTES (n = 4). All error bars ± SEM. n.s., not sig-
GPT2 incubated with or without BPTES and/or DMKG for 72 h (n = nificant. *p < 0.05, **p < 0.01 and ***p < 0.001
M. Kim et al.

A MiaPaCa2 B HCT116 mitochondrial integrity [5], which can increase mitochon-


1.2 1.2 *** drial ROS production. Given the importance of Gln ana-
plerosis in maintaining cellular redox state, it has been

Relative cell viability


Relative cell viability

reported that growth defects caused by Gln deprivation were


0.8 ** 0.8 rescued by the treatment of antioxidants such as NAC or
*** GSH [6]. However, we found that elevated ROS upon GLS
inhibition induces GPT2 expression, which is important for
0.4 0.4 cell growth and survival. Moreover, when GPT2 is
impaired, NAC treatment has minimal effects on GLS
inhibition-mediated growth suppression. These findings are
0.0 0.0 in line with a previous work showing that an enhanced
BPTES - + - + BPTES - + - + ROS production contributes to cell survival of Gln-
AOA - - + + AOA - - + + depleted cancer cells by promoting transcriptional
activation of Sestrin2 [23]. Thus, further studies are
C A549 E needed for delineating the roles of ROS in Gln-deprived
1.2
conditions.
Relative cell viability

***
0.8
Materials and methods

Cell culture
0.4

HeLa and HCT116 cell lines used in this work have been
described before [27]. MiaPaCa2 and MCF-7 cell lines were
0.0
provided by Dr. Alec C. Kimmelman (NYU Langone
BPTES - + - +
Medical Center, New York, NY, USA). A549 cell line was
AOA - - + +
provided by Dr. Jeong-Hwa Lee (The Catholic University
D MCF-7 of Korea, Seoul, Korea). Primary MEFs were immortalized
1.2 with retrovirus expression SV40 large T antigen at passage
3. MEFs, HeLa, MiaPaCa2, HCT116, MCF-7, and A549
Relative cell viability

were cultured in Dulbecco’s modified Eagle’s medium


***
0.8 (Biowest, Nuaillé, France) supplemented with 10% fetal
bovine serum (FBS, Biowest) and penicillin/streptomycin
(Biowest). All cell lines were tested routinely for myco-
0.4 plasma contamination by using a commercially available kit
(Takara Bio Inc, Shinga, Japan).

0.0 Constructs and reagents


BPTES - + - +
AOA - - + + The following antibodies were used: GLS1 (ab93434,
Fig. 5 Multiple cancer cell lines suffer the consequence of synthetic Abcam, Cambridge, MA, USA), GLUD1 (H00002746-
lethality by combined BPTES and AOA. a–d Relative cell viability of M01, Abnova, Taipei, Taiwan), GOT2 (CSB-PA912447,
MiaPaCa2 (a), HCT116 (b), A549 (c), and MCF-7 (d) cell lines Cusabio biotech, Wuhan, China), GPT2 (16757-1-AP,
treated with or without BPTES and/or AOA (n = 4). e A proposed
model illustrating the regulation of cell survival via ROS-ATF4-GPT2
Proteintech Group, Rosemont, IL, USA) and β-actin
during mitochondrial Gln metabolism-inhibited conditions. All error (A4700, Sigma, St. Louis, MO, USA). Ala, glutamate, Asn,
bars, ± SEM. **p < 0.01 and ***p < 0.001 OAA, cycloserine, AOA, DMKG, GSH and NAC were
purchased from Sigma. BPTES was purchased from Cay-
man Chemical (Ann Arbor, MI, USA). CB-839 was
cellular ROS production [6]. For example, Gln metabolism obtained from Selleckchem (Houston, TX, USA). shRNAs
provides the metabolic precursors necessary for de novo for GPT2 (TRCN0000035026 and TRCN0000035028)
synthesis of GSH, an intracellular antioxidant [26]. Addi- were purchased from Sigma. Other shRNAs were described
tionally, inhibition of Gln anaplerosis results in the loss of previously [6].
Mitochondrial GPT2 plays a pivotal role in metabolic adaptation to the perturbation of mitochondrial. . .

Quantitative RT-PCR ROS determination

Total RNA was prepared with Ribospin kit (GeneAll, The levels of cellular ROS were assessed by using DCFDA
Seoul, Korea) according to the manufacturer’s instructions. (Invitrogen, Grand Island, NY, USA) according to the
In all, 0.5 μg of total RNA was reverse-transcribed using the manufacturer’s instructions. Cells were washed with PBS
iScript cDNA synthesis kit (Bio-Rad, Hercules, CA, USA). and then incubated at 37 °C for 30 min in Hank’s balanced
Diluted cDNAs were analyzed by real-time PCR using salt solution (Gibco, Big Cabin, OK, USA) containing 5 μM
SYBR Green I Mastermix on a Lightcycler 480 (Roche, DCFDA. Cells were trypsinized, pelleted by centrifugation
South San Francisco, CA, USA). The level of gene and resuspended in Hank’s balanced salt solution. Fluor-
expression was normalized to β-actin. The primer sequences escence was measured by flow cytometry using a FACS
were: TTCCAGAAGGCACAGACATG and GGCTCAG Canto (BD Biosciences, San Jose, CA, USA).
TACTCTTTCACCAG for human GLS1; AGGAATGA
CACCAGGGTTTG and TCAGACTCACCAACAGCAA Cell viability assay
TAC for human GLUD1; GTTTGCCTCTGCCAATCA
TATG and GAGGGTTGGAATACATGGGAC for human Cells were plated into 96-well plates at 1000 cells per well
GOT2; CATGGACATTGTCTGAACC and TTACCCAG in 100 μl of growth media. The following day, the cells
GACCGACTCCTT for human GPT2; CAGCTGAAA were treated with drugs or were replaced by growth media.
GAAGCCCAAGT and AGAGCCTGAATGCCTTCCTC Parallel plates were analyzed at 3 days by Cell Titer Glo
for human ASNS; TGAGGCTTGGCTTTTGTGAA and analysis (Promega, Madison, Wisconsin, USA), per the
GGCTCTGGTGTAACAAAGCC for human BCAT1; manufacturer’s instruction.
CCCAGTGCTACTTTGAGGGG and CTGTTACTGTC
GATGATGAGGC for human OPLAH; GAGTCTG Flow cytometric measurement of cell death
CAGGTGCGAGAGTTGTA and TTTGGCCACTTTAG
CATAATCTGAGC for human GGH; CCAACAA Cells at less than 80% confluence were treated with drugs.
CAGCAAGGAGGAT and AGTGTCATCCAACGTG After 72 h, cells were harvested by trypsinization, pelleted
GTCA for human ATF4; CTACGTCGCCC TGGA by centrifugation, and resuspended in PBS. The measure-
CTTCGAGC and GATGGAGCCGCCGATCCACACGG ment of cell death was performed by flow cytometry using
for human β-actin; ATTATGACTCCAGAACAGCCC and propidiumiodide (PI) staining, as previously described [28].
AGTGTAGTGCTTCATCCATGG for mouse GLS1;
AGGAATGACACCAGGCTTTG and CTCCAACACC Reporter assay
AACACATTTAGC for mouse GLUD1; GATCGG
CATGTTCTGTTTCAC and CATGTAGACCGAGAAC The human GPT2 promoter was cloned into pGL3 basic
TCCTTG for mouse GOT2; CTGCACCTACCCAAACC vector to obtain a pGL3-GPT2 promoter plasmid. pGL3-
TAC and TGCCACATCTTCACGGATAC for mouse GPT2 promoter was co-transfect with Renilla luciferase-
GPT2; and AGCCATGTACGTAGCCATCC and CTCTC expressing pRL-TK. After 18 h of transfection, the medium
AGCTGTGGTGGTGAA for mouse β-actin. was changed to complete or Gln-free medium for 24 h.
Luciferase activity was then assayed according to the
Western blotting manufacturer’s instructions (Promega). Firefly luciferase
activity was normalized to Renilla luciferase activity.
Cells were lysed with EZ-RIPA lysis buffer (ATTO, WSE- Mutations in the GPT2 promoter were generated with the
7420, Tokyo, Japan) supplemented with protease inhibitor Quickchange kit (Agilent Technologies, CA, USA) and
cocktail (Roche) and phosphatase inhibitors (Sigma). Cell primer sequences were: CCGTGTGGCCTTGGACCCT
lysates were separated by sodium dodecyl sulfate- GAAACTCGGGGCG and CGCCCCGAGTTTCAGGGT
polyacrylamide gel electrophoresis (SDS-PAGE) and CCAAGGCCACACGG.
immunoblotting.
Intracellular glutamate detection
Clonogenic assay
Cells were plated into 96-well plates in 100 μl of growth
Cells were plated in 6-well plates at 300 cells per well in 2 media. The following day, the cells were supplemented
ml of media and treated with drugs the day after seeding. media with or without BPTES. Parallel plates were ana-
After 8–12 days, colonies were fixed with 80% methanol lyzed at 3 days by Glutamate-Glo™ Assay Kit (Promega),
and stained with 0.2% crystal violet. Media was not chan- per the manufacturer’s instruction. Glutamate levels nor-
ged throughout the course of the experiment. malized to the number of cells in each well.
M. Kim et al.

Statistical analysis 11. Polet F, Corbet C, Pinto A, Rubio LI, Martherus R, Bol V, et al.
Reducing the serine availability complements the inhibition of the
glutamine metabolism to block leukemia cell growth. Oncotarget.
Statistical analysis was performed using GraphPad Prism
2016;7:1765–76.
5 software (GraphPad Software, Inc, San Diego, CA, USA). 12. Sun L, Song L, Wan Q, Wu G, Li X, Wang Y, et al. cMyc-
The graphs show the mean from independent experiments mediated activation of serine biosynthesis pathway is critical for
unless otherwise specified (SD is standard deviation and cancer progression under nutrient deprivation conditions. Cell
Res. 2015;25:429–44.
SEM is standard error of the mean). Sample size and
13. Toda K, Kawada K, Iwamoto M, Inamoto S, Sasazuki T, Shir-
number of independent experiments for each experiment is asawa S, et al. Metabolic Alterations Caused by KRAS Mutations
indicated in the appropriate figure legend. Unless otherwise in Colorectal Cancer Contribute to Cell Adaptation to Glutamine
noted, n equals the number of independent biological Depletion by Upregulation of Asparagine Synthetase. Neoplasia.
2016;18:654–65.
replicates. All experiments were performed at least twice.
14. Ye J, Kumanova M, Hart LS, Sloane K, Zhang H, De Panis DN,
Statistically significant differences were calculated using et al. The GCN2-ATF4 pathway is critical for tumour cell survival
unpaired two-tailed Student’s t-test or two-way ANOVA. A and proliferation in response to nutrient deprivation. EMBO J.
p-value of < 0.05 was considered statistically significant. 2010;29:2082–96.
15. Zhang J, Fan J, Venneti S, Cross JR, Takagi T, Bhinder B, et al.
Asparagine plays a critical role in regulating cellular adaptation to
Acknowledgements This research was supported by Basic Science
glutamine depletion. Mol Cell. 2014;56:205–18.
Research Program through the National Research Foundation of Korea
16. Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY,
(NRF) funded by the Ministry of Education (2015R1C1A1A01052548
Pfeiffer HK, et al. Myc regulates a transcriptional program that
and 2018R1D1A1B07040961).
stimulates mitochondrial glutaminolysis and leads to glutamine
addiction. Proc Natl Acad Sci USA. 2008;105:18782–7.
Compliance with ethical standards 17. Wong DT, Fuller RW, Molloy BB. Inhibition of amino acid
transaminases by L-cycloserine. Adv Enzym Regul. 1973;11:
Conflict of interest The authors declare that they have no conflict of 139–54.
interest. 18. Ferguson J, Smith M, Zudaire I, Wellbrock C, Arozarena I.
Glucose availability controls ATF4-mediated MITF suppression
to drive melanoma cell growth. Oncotarget. 2017;8:32946–59.
Publisher’s note: Springer Nature remains neutral with regard to
19. Hao Y, Samuels Y, Li Q, Krokowski D, Guan BJ, Wang C, et al.
jurisdictional claims in published maps and institutional affiliations.
Oncogenic PIK3CA mutations reprogram glutamine metabolism
in colorectal cancer. Nat Commun. 2016;7:11971.
References 20. Salgado MC, Meton I, Anemaet IG, Baanante IV. Activating
transcription factor 4 mediates up-regulation of alanine amino-
1. Tennant DA, Duran RV, Gottlieb E. Targeting metabolic trans- transferase 2 gene expression under metabolic stress. Biochim
formation for cancer therapy. Nat Rev Cancer. 2010;10:267–77. Biophys Acta. 2014;1839:288–96.
2. Vander Heiden MG, Cantley LC, Thompson CB. Understanding 21. Wang SF, Chen MS, Chou YC, Ueng YF, Yin PH, Yeh TS, et al.
the Warburg effect: the metabolic requirements of cell prolifera- Mitochondrial dysfunction enhances cisplatin resistance in human
tion. Science. 2009;324:1029–33. gastric cancer cells via the ROS-activated GCN2-eIF2alpha-
3. Wise DR, Thompson CB. Glutamine addiction: a new therapeutic ATF4-xCT pathway. Oncotarget. 2016;7:74132–51.
target in cancer. Trends Biochem Sci. 2010;35:427–33. 22. Biancur DE, Paulo JA, Malachowska B, Quiles Del Rey M, Sousa
4. Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: glutamine CM, Wang X, et al. Compensatory metabolic networks in pan-
metabolism to cancer therapy. Nat Rev Cancer. 2016;16:619–34. creatic cancers upon perturbation of glutamine metabolism. Nat
5. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, Commun. 2017;8:15965.
Wehrli S, et al. Beyond aerobic glycolysis: transformed cells can 23. Byun JK, Choi YK, Kim JH, Jeong JY, Jeon HJ, Kim MK, et al.
engage in glutamine metabolism that exceeds the requirement for A Positive feedback loop between Sestrin2 and mTORC2 Is
protein and nucleotide synthesis. Proc Natl Acad Sci USA. required for the survival of glutamine-depleted lung cancer cells.
2007;104:19345–50. Cell Rep. 2017;20:586–99.
6. Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. 24. Patel D, Menon D, Bernfeld E, Mroz V, Kalan S, Loayza D, et al.
Glutamine supports pancreatic cancer growth through a KRAS- Aspartate rescues S-phase arrest caused by suppression of gluta-
regulated metabolic pathway. Nature. 2013;496:101–5. mine utilization in KRas-driven cancer cells. J Biol Chem.
7. Yang S, Hwang S, Kim M, Seo SB, Lee JH, Jeong SM. Mito- 2016;291:9322–9.
chondrial glutamine metabolism via GOT2 supports pancreatic 25. Shanware NP, Mullen AR, DeBerardinis RJ, Abraham RT. Glu-
cancer growth through senescence inhibition. Cell death & Dis. tamine: pleiotropic roles in tumor growth and stress resistance. J
2018;9:55. Mol Med. 2011;89:229–36.
8. Panieri E, Santoro MM. ROS homeostasis and metabolism: a 26. Lu SC. Regulation of glutathione synthesis. Mol Asp Med.
dangerous liason in cancer cells. Cell death & Dis. 2016;7:e2253 2009;30:42–59.
9. Robinson MM, McBryant SJ, Tsukamoto T, Rojas C, Ferraris 27. Jeong SM, Hwang S, Park K, Yang S, Seong RH. Enhanced
DV, Hamilton SK, et al. Novel mechanism of inhibition of rat mitochondrial glutamine anaplerosis suppresses pancreatic cancer
kidney-type glutaminase by bis-2-(5-phenylacetamido-1,2,4-thia- growth through autophagy inhibition. Sci Rep. 2016;6:30767.
diazol-2-yl)ethyl sulfide (BPTES). Biochem J. 2007;406:407–14. 28. Jeong SM, Xiao C, Finley LW, Lahusen T, Souza AL, Pierce K,
10. Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, et al. SIRT4 has tumor-suppressive activity and regulates the
Goyal B, et al. Antitumor activity of the glutaminase inhibitor CB-839 cellular metabolic response to DNA damage by inhibiting mito-
in triple-negative breast cancer. Mol Cancer Ther. 2014;13:890–901. chondrial glutamine metabolism. Cancer Cell. 2013;23:450–63.

You might also like