scispace - formally typeset
Search or ask a question
Posted ContentDOI

Expression and function of SLC38A5, an amino acid-coupled Na+/H+ exchanger, in triple-negative breast cancer and its relevance to macropinocytosis.

TL;DR: In this paper, the authors examined the impact of SLC38A5 on macropinocytosis in triple-negative breast cancer (TNBC) cells and found that the transport function of the amino acid transporter is coupled to induction of protein-protein interactions.
Abstract: Metabolic reprogramming in cancer cells necessitates increased amino acid uptake, which is accomplished by upregulation of specific amino acid transporters. Since amino acid transporters differ in substrate selectivity, mode of transport, and driving forces, not all tumors rely on any single amino acid transporter for this purpose. Here we report on the differential upregulation of the amino acid transporter SLC38A5 in triple-negative breast cancer (TNBC). The upregulation is evident in primary TNBC tumors, conventional TNBC cell lines, patient-derived xenograft TNBC cell lines, and a mouse model of spontaneous mammary tumor representing TNBC. The upregulation is confirmed by functional assays. SLC38A5 is an amino acid-dependent Na+/H+ exchanger which transports Na+ and amino acids into cells coupled with H+ efflux. Since the traditional Na+/H+ exchanger is an established inducer of macropinocytosis, an endocytic process for cellular uptake of bulk fluid and its components, we examined the impact of SLC38A5 on macropinocytosis in TNBC cells. We found that the transport function of SLC38A5 is coupled to induction of macropinocytosis. Surprisingly, the transport function of SLC38A5 is inhibited by amilorides, the well-known inhibitors of Na+/H+ exchanger, possibly related to the amino acid-dependent Na+/H+ exchange function of SLC38A5. The Cancer Genome Atlas database corroborates SLC38A5 upregulation in TNBC. This represents the first report on the selective expression of SLC38A5 in TNBC and its role as an inducer of macropinocytosis, thus revealing a novel, hitherto unsuspected, function for an amino acid transporter that goes beyond amino acid delivery but is still relevant to cancer cell nutrition. Summary Statement SLC38A5 is an amino acid-coupled Na+/H+ exchanger that is upregulated in triple-negative breast cancer, and its function in cancer cells goes beyond amino acid delivery; it promotes macropinocytosis, a distinct form of endocytic process for cellular uptake of proteins and other nutrients present in extracellular fluid.

Summary (3 min read)

Jump to: [Introduction][Materials][Human tissues][Animals][Uptake assays][RT-PCR][Macropinocytosis assay][Statistics][Results][Discussion] and [Figure Legends]

Introduction

  • Cancer cells reprogram their metabolism to best suit their need for accelerated proliferation associated with enhanced protein/lipid synthesis, DNA/RNA synthesis, and potentiated machinery for protection against oxidative stress and apoptosis [1-3].
  • This requires increased supply of nutrients to feed into the altered metabolic pathways; amino acids represent an important group among such nutrients.
  • Only recently, a couple of studies have focused on SLC38A2 in breast cancer [16,17].

Materials

  • Ethylisopropyl amiloride and hexamethylene amiloride were from Sigma Aldrich (St. Louis, MO, USA).
  • Amiloride, benzyl amiloride and harmaline were from Cayman Chemical (Ann Arbor, MI, USA).

Human tissues

  • Human breast cancer tissues and the surrounding normal tissues were obtained from the Augusta University Tumor Bank.
  • The Tumor Bank collects and maintains a repository of de-identified tumor tissues and matched normal tissues; the tumor tissue collection has the approval from the Institutional Review Board and the Human Assurance Committee.
  • These tissues are available to investigators without a separate approval from the Institutional Review Board.

Animals

  • The authors used three different transgenic mouse models of spontaneous breast cancer for the analysis of Slc38a5 expression: MMTV-Neu (a model for HER2-positive breast cancer), MMTV-HRAS (a model for Ras activation-associated breast cancer), and MMTV-PyMT (polyoma middle T antigen-driven breast cancer, which starts initially as estrogen receptor positive and subsequently turns into estrogen receptor-negative).
  • The authors used four human non-transformed mammary epithelial cell lines and seven estrogen receptornegative breast cancer cell lines.
  • MCF10A and MCF12A cells were cultured in a special medium consisting of Dulbecco’s Modified Eagle’s Medium and Ham’s F12 medium, in a 1:1 ratio, supplemented with 20 ng/ml human EGF, 0.01 mg/ml bovine insulin, 500 ng/ml hydrocortisone, and 100 ng/ml cholera toxin.
  • All media contained 10% fetal bovine serum.

Uptake assays

  • Uptake of [3H]-serine was used to monitor the transport function of SLC38A5.
  • As there are several amino acid transporters, even for serine that is used as the substrate in the present study, that are Na+-coupled, the authors cannot specifically monitor the function of SLC38A5 by using Na+-containing uptake buffer.
  • Cells were seeded in 24-well culture plates (2 x 105 cells/well) with the culture medium.
  • The uptake medium (250 µl) containing [3H]-serine was added to the cells.
  • On the day of the uptake assay, cells were incubated with NaCl-buffer (same as the uptake buffer except that LiCl or NMDGCl was replaced with NaCl), pH 7.5.

RT-PCR

  • RNA was extracted from cells and mammary tissues from mice (normal and tumor) by TRIzol reagent (Thermo Fisher Scientific), and the RNA was reverse-transcribed using High-capacity cDNA reverse transcription kit (Thermo Fisher Scientific).
  • PCR and quantitative-PCR were performed with Takara Taq Hot Start Version (TaKaRa Biotechnology, Shiga, Japan) or Power SYBR Green PCR master mix (Thermo Fisher Scientific).
  • Human-specific primers were used for cell lines whereas mouse-specific primers were used for mouse tissues.
  • The relative mRNA expression was determined by the 2-ΔΔCt method.
  • HPRT (hypoxanthine/guanine phosphoribosyl transferase) was used as a housekeeping gene for normalization.

Macropinocytosis assay

  • Cells were plated on coverslips, placed in wells in a 12 well-plate, at a density of 1x105 cells/well, and cultured with 5% CO2 at 37 oC using the culture media (with 10% fetal bovine serum) recommended for the respective cell lines.
  • About 16 h prior to macropinocytosis assay, the medium was removed and fresh culture medium without the fetal bovine serum was added.
  • The images represent a maximum projection intensity derived from a Z-stack.
  • The fluorescence quantification was done by measuring the Corrected Total Cell fluorescence (CTCF) using Image J and the following formula: CTCF = (integrated density) - (area of cell of interest) x (mean fluorescence of background).

Statistics

  • RT-PCR and uptake studies were done in triplicates and repeated twice.
  • Statistical analysis was performed with a two-tailed, paired Student’s t-test for single comparison and a p value < 0.05 was considered statistically significant.
  • For quantification of fluorescence signals in image analysis related to micropinocytosis and for analysis of inhibition of serine uptake by amiloride and its derivatives, ANOVA followed by Dunn’s test was used to determine the significance of difference among the different groups.

Results

  • Differential expression of SLC6A14 and SLC38A5 in estrogen receptor-positive (ER+) breast cancer and triple-negative (TNBC) breast cancer Functional evidence for SLC38A5 expression in TNBC cells A unique feature that distinguishes SLC38A5 from other Na+-coupled amino acid transporters is its ability to function in the presence of Li+ in place of Na+.
  • When Na+ is removed from the medium, serine uptake does not occur via SLC38A5 because its transport activity is obligatorily coupled to Na+ (or Li+) and at the same time Na+/H+ exchanger functions in the opposite direction.
  • To further confirm the role of SLC38A5 as a promoter of macropinocytosis, the authors employed experimental conditions in which serine uptake occurs solely via SLC38A5.
  • The authors found the expression of SLC38A5 to be significantly higher in primary breast tumor tissues than in normal breast tissue (Fig. S3A).

Discussion

  • The Na+/Cl- -coupled broad-selective amino acid transporter SLC6A14 is expressed ER+ breast cancer cell lines but not in TNBC cell lines [25].
  • Whenever amino acid transporters in cancer are investigated, the sole focus is on the role of these transporters in the provision of amino acids to cancer cells.
  • There are about three dozen amino acid transporters in mammalian cells, and to the best of their knowledge, no amino acid transporter other than SLC38A3 and SLC38A5 possesses this interesting feature of Na+/H+ exchange.
  • Even though SLC38A3 exhibits functional features that are almost identical to those of SLC38A5 [7], the expression of the former is actually decreased in breast cancer, and the decrease is independent of the hormone receptor status of the tumor (TCGA database).
  • This glutamine is then released into the extracellular fluid via SLC38A3 and SLC38A5, which is then taken up by neurons via SLC38A1 and SLC38A2 for subsequent conversion into glutamate for reuse as the neurotransmitter.

Figure Legends

  • Relative expression of the amino acid transporters SLC6A14, SLC38A5, and SLC1A5 in paired tissues samples of breast cancer (ER+ breast cancer and ER-negative breast cancer) and normal mammary gland.
  • (B) Transport activity of SLC38A5 in the TNBC cell line MB231 as monitored by the uptake of serine, glycine, and glutamine as the substrates for the transporter.
  • The transport function of SLC38A5 was monitored in an uptake buffer (pH 8.5) containing 5 mM tryptophan to suppress the involvement of SLC7A5 in the uptake and by comparing the uptake in the presence and absence of Li+.
  • Mice used the study did not go through breeding or pregnancy.
  • (B) Quantification of the fluorescence signals, and statistical significance among the groups.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
Expression and function of SLC38A5, an amino acid-coupled Na
+
/H
+
exchanger, in triple-negative breast cancer and its relevance to
macropinocytosis
Sabarish RAMACHANDRAN
1
, Souad SENNOUNE
1
, Monica SHARMA
2
, Muthusamy
THANGARAJU
3
, Varshini SURESH
4
, Yangzom D. BHUTIA
1
, Kevin PRUITT
2
, and Vadivel
GANAPATHY
1,
*
1
Department of Cell Biology and Biochemistry, and
2
Department of Immunology and Molecular
Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
3
Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta
University, Augusta, GA 30912, USA
4
Department of Biology, Texas Tech University, Lubbock, TX 79409, USA
Corresponding author: Vadivel Ganapathy, Ph. D., Department of Cell Biology and
Biochemistry, Texas Tech University Health Sciences Center, 3601 4
th
street, Lubbock, TX
79430, USA. Email: Vadivel.ganapathy@ttuhsc.edu
Keywords: glutamine addiction, one-carbon metabolism, intracellular alkalinization, breast
cancer, macropinocytosis, amiloride
Short Title: SLC38A5 in breast cancer and its relevance to macropinocytosis
Summary Statement
SLC38A5 is an amino acid-coupled Na
+
/H
+
exchanger that is upregulated in triple-negative breast
cancer, and its function in cancer cells goes beyond amino acid delivery; it promotes
macropinocytosis, a distinct form of endocytic process for cellular uptake of proteins and other
nutrients present in extracellular fluid.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted June 18, 2021. ; https://doi.org/10.1101/2021.06.17.448844doi: bioRxiv preprint

2
Abstract
Metabolic reprogramming in cancer cells necessitates increased amino acid uptake, which is
accomplished by upregulation of specific amino acid transporters. Since amino acid transporters
differ in substrate selectivity, mode of transport, and driving forces, not all tumors rely on any
single amino acid transporter for this purpose. Here we report on the differential upregulation of
the amino acid transporter SLC38A5 in triple-negative breast cancer (TNBC). The upregulation is
evident in primary TNBC tumors, conventional TNBC cell lines, patient-derived xenograft TNBC
cell lines, and a mouse model of spontaneous mammary tumor representing TNBC. The
upregulation is confirmed by functional assays. SLC38A5 is an amino acid-dependent Na
+
/H
+
exchanger which transports Na
+
and amino acids into cells coupled with H
+
efflux. Since the
traditional Na
+
/H
+
exchanger is an established inducer of macropinocytosis, an endocytic process
for cellular uptake of bulk fluid and its components, we examined the impact of SLC38A5 on
macropinocytosis in TNBC cells. We found that the transport function of SLC38A5 is coupled to
induction of macropinocytosis. Surprisingly, the transport function of SLC38A5 is inhibited by
amilorides, the well-known inhibitors of Na
+
/H
+
exchanger, possibly related to the amino acid-
dependent Na
+
/H
+
exchange function of SLC38A5. The Cancer Genome Atlas database
corroborates SLC38A5 upregulation in TNBC. This represents the first report on the selective
expression of SLC38A5 in TNBC and its role as an inducer of macropinocytosis, thus revealing a
novel, hitherto unsuspected, function for an amino acid transporter that goes beyond amino acid
delivery but is still relevant to cancer cell nutrition.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted June 18, 2021. ; https://doi.org/10.1101/2021.06.17.448844doi: bioRxiv preprint

3
Introduction
Cancer cells reprogram their metabolism to best suit their need for accelerated proliferation
associated with enhanced protein/lipid synthesis, DNA/RNA synthesis, and potentiated machinery
for protection against oxidative stress and apoptosis [1-3]. This requires increased supply of
nutrients to feed into the altered metabolic pathways; amino acids represent an important group
among such nutrients. Because of their hydrophilic nature, amino acids cannot permeate plasma
membrane by diffusion; they enter the cells via selective transporters. There are more than three
dozen amino acid transporters in mammalian cells [4]. They are grouped into seven different solute
carrier gene families, based on the similarity in primary amino acid sequence. The increased
demand for amino acids in cancer cells is met by upregulation of selective amino acid transporters.
The most studied transporters for their role in cancer include SLC1A5 (Alanine-Serine-Cysteine
Transporter 2 or ASCT2) [5-8], SLC7A5 (system L Amino acid Transporter 1 or LAT1) [5-9],
SLC7A11 (system x
-
c
transporter or xCT) [5,6,10,11], and SLC6A14 (Amino acid Transporter B
0,+
or ATB
0,+
) [5-8,12-14]. The contribution of each of these transporters to tumor promotion is the
provision of selective amino acids to cancer cells, which might impact on a multitude of
downstream metabolic pathways and signaling cascades.
SLC38 family contains several amino acid transporters [15], but there is little or no information in
the literature on the role of these transporters in cancer. Only recently, a couple of studies have
focused on SLC38A2 in breast cancer [16,17]. This transporter represents the classical Na
+
-
coupled neutral amino acid transporter known as system A, cloned and functionally characterized
in our laboratory [18]. It was called “system A” because of its preference for the amino acid
alanine. However, it transports not only alanine but also other small, hydrophilic, amino acids such
as glycine and serine. Interestingly, glutamine is an excellent substrate for this transporter.
SLC38A2 is induced by hypoxia and its expression is related to endocrine resistance in breast
cancer [16]. In triple-negative breast cancer (TNBC), the level of SLC38A2 expression is
associated with worse prognosis [17]. Both studies have implicated SLC38A2 in supplying
glutamine to cancer cells. Notwithstanding these data suggesting a role for this transporter in breast
cancer progression, its expression is actually downregulated in breast cancer, particularly in TNBC
(p<1.5x10
-8
) (TCGA database). The decreased expression in cancer is paradoxical and
counterintuitive, given the proposed role of the transporter as a tumor promoter.
In the present study, we focused on another member of the SLC38 family, namely SLC38A5. This
transporter was also cloned in our laboratory [19,20]. It is referred to as SN2 transporter, meaning
that it is the second isoform with functional features defined selectively for the amino acid
transport system N. This system is Na
+
-coupled and accepts as substrates amino acids that contain
nitrogen in the side chain (i.e., glutamine, asparagine, and histidine). However, functional
characterization of the cloned transporter revealed two interesting features: (i) it also transports
glycine, serine and methionine, with highest affinity towards serine, and (ii) it utilizes an outward-
directed H
+
gradient as a driving force in addition to an inward-directed Na
+
gradient [19,20]. The
transport mechanism involves the transfer of Na
+
and amino acid substrate in one direction coupled
to the transfer of H
+
in the opposite direction, with the overall transport process being
electroneutral. Stated differently, SLC38A5 is an amino acid-coupled Na
+
/H
+
exchanger. The
rationale for the present study to evaluate the expression and function of SLC38A5 in breast cancer
is the following: (i) SLC38A5 is a target for the oncogene c-Myc [21]; (ii) the substrate selectivity
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted June 18, 2021. ; https://doi.org/10.1101/2021.06.17.448844doi: bioRxiv preprint

4
of SLC38A5 suggests a potentially a key role for this transporter in cancer-associated metabolic
pathways such as glutaminolysis (glutamine) and one-carbon metabolism (serine, glycine, and
methionine) [14]; (iii) the efflux of H
+
coupled to amino acid entry fulfills dual needs in cancer
cells, namely supply of amino acids and removal of H
+
[19,20], and (iv) Na
+
/H
+
exchanger is
known to promote macropinocytosis, a novel mechanism for cellular entry of nutrients from
extracellular medium [22,23], suggesting a possibility that SLC38A5 as an amino acid-coupled
Na
+
/H
+
exchanger might also promote macropinocytosis in cancer cells.
Materials and Methods
Materials
[2,3-
3
H]-L-Serine (specific radioactivity, >5 Ci/mmol) was purchased from Moravek, Inc. (Brea,
CA, USA). [2-
3
H]-Glycine (specific radioactivity, x Ci/mmol) and [3,4-
3
H]-glutamine (specific
radioactivity, x Ci/mmol) were purchased from Perkin Elmer Corp. (Waltham, MA, USA).
Ethylisopropyl amiloride and hexamethylene amiloride were from Sigma Aldrich (St. Louis, MO,
USA). Amiloride, benzyl amiloride and harmaline were from Cayman Chemical (Ann Arbor, MI,
USA).
Human tissues
Human breast cancer tissues and the surrounding normal tissues were obtained from the Augusta
University Tumor Bank. The Tumor Bank collects and maintains a repository of de-identified
tumor tissues and matched normal tissues; the tumor tissue collection has the approval from the
Institutional Review Board and the Human Assurance Committee. These tissues are available to
investigators without a separate approval from the Institutional Review Board.
Animals
We used three different transgenic mouse models of spontaneous breast cancer for the analysis of
Slc38a5 expression: MMTV-Neu (a model for HER2-positive breast cancer), MMTV-HRAS (a
model for Ras activation-associated breast cancer), and MMTV-PyMT (polyoma middle T
antigen-driven breast cancer, which starts initially as estrogen receptor positive and subsequently
turns into estrogen receptor-negative). These mouse lines were originally obtained from the
Jackson Laboratory (Bar Harbor, ME, USA): MMTV-Neu, stock # 002376; MMTV-HRAS, stock
# x; MMTV-PyMT, stock # 002374). Since all mice used in the present study never went through
pregnancy, we used mammary tissues from adult (~12-week-old) virgin wild type mice as a control
for comparison. These mice form spontaneous tumors in mammary glands at different ages: 8-10
months in MMTV-Neu and MMTV-HRAS mouse lines; 2-3 months in the MMTV-PyMT mouse
line). The protocol was approved by the Institutional Animal Care and Use Committee of the
Texas Tech University Health Sciences Center, Lubbock, TX, USA (IACUC approval number:
15002 for breeding protocol and 18005 for experimental protocol). All the experiments described
in this study, including the animal experiments, were conducted at this institution. At the
termination of the study, mice were killed by cervical dislocation under CO
2
anesthesia in
accordance with the guidelines from the American Veterinary Medical Association.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted June 18, 2021. ; https://doi.org/10.1101/2021.06.17.448844doi: bioRxiv preprint

5
Cell lines and culture conditions
We used four human non-transformed mammary epithelial cell lines and seven estrogen receptor-
negative breast cancer cell lines. The non-transformed cell lines are: HMEC (ATCC, Cat. no. PCS-
600-010), HBL100 (ATCC, Cat. no. HTB-124), MCF10A (ATCC, Cat. no. CRL-10317), and
MCF12A (ATCC, Cat. no. CRL10782). HMEC cells were cultured in Mammary Epithelial Cell
Basal Medium (ATCC, Cat. no. PCS-600-030) and HBL100 cells were cultured in modified
McCoy’s 5a medium (ATCC, Cat. no. 30-2007). MCF10A and MCF12A cells were cultured in a
special medium consisting of Dulbecco’s Modified Eagle’s Medium and Ham’s F12 medium, in
a 1:1 ratio, supplemented with 20 ng/ml human EGF, 0.01 mg/ml bovine insulin, 500 ng/ml
hydrocortisone, and 100 ng/ml cholera toxin. All media contained 10% fetal bovine serum. Six of
the seven estrogen receptor-negative cell lines were obtained from ATCC: BT20 (Cat. no. HTB-
19), HCC1937 (Cat. no. CRL-2336), MDA-MB231 (Cat. no. CRM-HTB-26), MDA-MB436 (Cat.
no. HTB-136), MDA-MB453 (Cat. no. HTB-131) and MDA-MB468 (Cat. no. HTB-132).
SUM1315MO2 cell line was obtained from Expasy (Cat. no. CVCL_5589). BT20 cells were
cultured in Eagle’s Minimum Essential Medium (ATCC, Cat. no. 30-2003); HCC1937 cells were
cultured in RPMI-1640 medium (ATCC, Cat. no. 30-2001); MDA-MB436 cells were cultured in
Leibovitz’s L-15 medium (ATCC, Cat. no. 30-2008), supplemented with 10 µg/ml insulin and 16
µg/ml glutathione. MDA-MB231, MDA-MB453, and MDA-MB468 cells were cultured in
Leibovitz’s L-15 medium. SUMO1315MO2 cells were cultured in Ham’s F12 medium containing
10 ng/ml EGF and 5 µg/ml insulin. All media contained 10% fetal bovine serum. All cell lines
were mycoplasma-free. Three patient-derived xenograft cell lines were provided by the TTUHSC
Cancer Center; these are identified as TXBR-100, TXBR-237 and TXBR-247. These cell lines
were cultured in the special medium described above for MCF10A.
Uptake assays
Uptake of [
3
H]-serine was used to monitor the transport function of SLC38A5. Since SLC38A5 is
a Na
+
-coupled transporter with involvement of H
+
movement in the opposite direction, the uptake
assays were done using a pH 8.5 buffer to create an outwardly directly H
+
gradient across the
plasma membrane. As there are several amino acid transporters, even for serine that is used as the
substrate in the present study, that are Na
+
-coupled, we cannot specifically monitor the function of
SLC38A5 by using Na
+
-containing uptake buffer. However, unlike other Na
+
-coupled amino acid
transporters, SLC38A5 is tolerant to Li
+
, meaning that this transporter functions when Na
+
is
replaced with Li
+
[19,20]. Therefore, we used an uptake buffer with LiCl in place of NaCl. The
composition of the uptake buffer was 25 mM Tris/Hepes, pH 8.5, containing 140 mM LiCl, 5.4
mM KCl, 1.8 mM CaCl
2
, 0.8 mM MgSO
4
, and 5 mM D-glucose. Serine is also a substrate for
SLC7A5 (LAT1), which is a Na
+
-independent transporter; therefore, uptake via this transporter
will contribute to the total uptake measured in the LiCl-containing buffer. To suppress serine
uptake that might occur via SLC7A5, the uptake buffer contained 5 mM tryptophan to compete
with and block the transport of serine mediated by SLC7A5; SLC38A5 does not transport
tryptophan and therefore SLC38A5-mediated serine uptake is not affected by tryptophan. To
determine the contribution of diffusion to the total uptake of serine, the same uptake buffer but
with LiCl replaced isosmotically with N-methyl-D-glucamine chloride (NMDGCl) was used.
Serine uptake was measured in two buffers: (i) LiCl-buffer, pH 8.5 with 5 mM tryptophan; (ii)
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted June 18, 2021. ; https://doi.org/10.1101/2021.06.17.448844doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
22 Jan 2022-Cancers
TL;DR: The metabolic and molecular pathways that are linked to dysregulated glutamine metabolism in multiple cancer types are discussed and their potential for clinical applications are discussed.
Abstract: Simple Summary Dysregulated glutamine metabolism is one of the metabolic features evident in cancer cells when compared to normal cells. Cancer cells utilize glutamine for energy generation as well as the synthesis of other molecules that are critical for cancer growth and progression. Therefore, drugs targeting glutamine metabolism have been extensively investigated. However, inhibition of glutamine metabolism in cancer cells results in the activation of other metabolic pathways enabling cancer cells to survive. In this review, we summarize and discuss the targets in glutamine metabolism, which has been probed in the development of anticancer drugs in preclinical and clinical studies. We further discuss pathways activated in response to glutamine metabolism inhibition, enabling cancer cells to survive the challenge. Finally, we put into perspective combined treatment strategies targeting glutamine metabolism along with other pathways as potential treatment options. Abstract Tumor growth and metastasis strongly depend on adapted cell metabolism. Cancer cells adjust their metabolic program to their specific energy needs and in response to an often challenging tumor microenvironment. Glutamine metabolism is one of the metabolic pathways that can be successfully targeted in cancer treatment. The dependence of many hematological and solid tumors on glutamine is associated with mitochondrial glutaminase (GLS) activity that enables channeling of glutamine into the tricarboxylic acid (TCA) cycle, generation of ATP and NADPH, and regulation of glutathione homeostasis and reactive oxygen species (ROS). Small molecules that target glutamine metabolism through inhibition of GLS therefore simultaneously limit energy availability and increase oxidative stress. However, some cancers can reprogram their metabolism to evade this metabolic trap. Therefore, the effectiveness of treatment strategies that rely solely on glutamine inhibition is limited. In this review, we discuss the metabolic and molecular pathways that are linked to dysregulated glutamine metabolism in multiple cancer types. We further summarize and review current clinical trials of glutaminolysis inhibition in cancer patients. Finally, we put into perspective strategies that deploy a combined treatment targeting glutamine metabolism along with other molecular or metabolic pathways and discuss their potential for clinical applications.

22 citations

Journal ArticleDOI
TL;DR: It is concluded that intracellular Ca2+ responses are amplified in breast cancer tissue, but not in colon cancer tissue and that nucleotide signaling stimulates breast cancer cell proliferation within the extracellular concentration range typical for solid cancer tissue.
Abstract: Intracellular Ca2+ dynamics shape malignant behaviors of cancer cells. Whereas previous studies focused on cultured cancer cells, we here used breast organoids and colonic crypts freshly isolated from human and murine surgical biopsies. We performed fluorescence microscopy to evaluate intracellular Ca2+ concentrations in breast and colon cancer tissue with preferential focus on intracellular Ca2+ release in response to purinergic and cholinergic stimuli. Inhibition of the sarco‐/endoplasmic reticulum Ca2+ ATPase with cyclopiazonic acid elicited larger Ca2+ responses in breast cancer tissue, but not in colon cancer tissue, relative to respective normal tissue. The resting intracellular Ca2+ concentration was elevated, and ATP, UTP and acetylcholine induced strongly augmented intracellular Ca2+ responses in breast cancer tissue compared with normal breast tissue. In contrast, resting intracellular Ca2+ levels and acetylcholine‐induced increases in intracellular Ca2+ concentrations were unaffected and ATP‐ and UTP‐induced Ca2+ responses were smaller in colon cancer tissue compared with normal colon tissue. In accordance with the amplified Ca2+ responses, ATP and UTP substantially increased proliferative activity—evaluated by bromodeoxyuridine incorporation—in breast cancer tissue, whereas the effect was minimal in normal breast tissue. ATP caused cell death—identified with ethidium homodimer‐1 staining—in breast cancer tissue only at concentrations above the expected pathophysiological range. We conclude that intracellular Ca2+ responses are amplified in breast cancer tissue, but not in colon cancer tissue, and that nucleotide signaling stimulates breast cancer cell proliferation within the extracellular concentration range typical for solid cancer tissue.

4 citations

Journal ArticleDOI
TL;DR: In this paper , the authors used the platinum nanocluster (short for nano-Pt) for optical imaging without the help of other fluorescent probes and possess targeted antitumor activity as well as low systemic toxicity.

3 citations

Journal ArticleDOI
TL;DR: In this article , a review examines the cell biological mechanisms that drive macropinocytosis, as well as the complex signaling pathways -GTPases, lipid and protein kinases and phosphatases, and actin regulatory proteins -that regulate macropinosome formation, internalization, and disposition.
Abstract: Macropinocytosis is defined as an actin-dependent but coat- and dynamin-independent endocytic uptake process, which generates large intracellular vesicles (macropinosomes) containing a non-selective sampling of extracellular fluid. Macropinocytosis provides an important mechanism of immune surveillance by dendritic cells and macrophages, but also serves as an essential nutrient uptake pathway for unicellular organisms and tumor cells. This review examines the cell biological mechanisms that drive macropinocytosis, as well as the complex signaling pathways - GTPases, lipid and protein kinases and phosphatases, and actin regulatory proteins - that regulate macropinosome formation, internalization, and disposition.

2 citations

Journal ArticleDOI
TL;DR: The present review highlights the unusual involvement of selective amino acid transporters in macropinocytosis and diet-induced obesity/metabolic syndrome.
Abstract: Amino acid transporters are expressed in mammalian cells not only in the plasma membrane but also in intracellular membranes. The conventional function of these transporters is to transfer their amino acid substrates across the lipid bilayer; the direction of the transfer is dictated by the combined gradients for the amino acid substrates and the co-transported ions (Na+, H+, K+ or Cl−) across the membrane. In cases of electrogenic transporters, the membrane potential also contributes to the direction of the amino acid transfer. In addition to this expected traditional function, several unconventional functions are known for some of these amino acid transporters. This includes their role in intracellular signaling, regulation of acid–base balance, and entry of viruses into cells. Such functions expand the biological roles of these transporters beyond the logical amino acid homeostasis. In recent years, two additional unconventional biochemical/metabolic processes regulated by certain amino acid transporters have come to be recognized: macropinocytosis and obesity. This adds to the repertoire of biological processes that are controlled and regulated by amino acid transporters in health and disease. In the present review, we highlight the unusual involvement of selective amino acid transporters in macropinocytosis (SLC38A5/SLC38A3) and diet-induced obesity/metabolic syndrome (SLC6A19/SLC6A14/SLC6A6).

2 citations

References
More filters
Journal ArticleDOI
TL;DR: This Perspective has organized known cancer-associated metabolic changes into six hallmarks: deregulated uptake of glucose and amino acids, use of opportunistic modes of nutrient acquisition, useof glycolysis/TCA cycle intermediates for biosynthesis and NADPH production, increased demand for nitrogen, alterations in metabolite-driven gene regulation, and metabolic interactions with the microenvironment.

3,565 citations

Journal ArticleDOI
TL;DR: It is reported that the transcriptional regulatory properties of the oncogene Myc coordinate the expression of genes necessary for cells to engage in glutamine catabolism that exceeds the cellular requirement for protein and nucleotide biosynthesis, resulting in the reprogramming of mitochondrial metabolism to depend on glutaminolysis to sustain cellular viability and TCA cycle anapleurosis.
Abstract: Mammalian cells fuel their growth and proliferation through the catabolism of two main substrates: glucose and glutamine. Most of the remaining metabolites taken up by proliferating cells are not catabolized, but instead are used as building blocks during anabolic macromolecular synthesis. Investigations of phosphoinositol 3-kinase (PI3K) and its downstream effector AKT have confirmed that these oncogenes play a direct role in stimulating glucose uptake and metabolism, rendering the transformed cell addicted to glucose for the maintenance of survival. In contrast, less is known about the regulation of glutamine uptake and metabolism. Here, we report that the transcriptional regulatory properties of the oncogene Myc coordinate the expression of genes necessary for cells to engage in glutamine catabolism that exceeds the cellular requirement for protein and nucleotide biosynthesis. A consequence of this Myc-dependent glutaminolysis is the reprogramming of mitochondrial metabolism to depend on glutamine catabolism to sustain cellular viability and TCA cycle anapleurosis. The ability of Myc-expressing cells to engage in glutaminolysis does not depend on concomitant activation of PI3K or AKT. The stimulation of mitochondrial glutamine metabolism resulted in reduced glucose carbon entering the TCA cycle and a decreased contribution of glucose to the mitochondrial-dependent synthesis of phospholipids. These data suggest that oncogenic levels of Myc induce a transcriptional program that promotes glutaminolysis and triggers cellular addiction to glutamine as a bioenergetic substrate.

1,703 citations

Journal ArticleDOI
06 Feb 2009-Cell
TL;DR: It is shown that cellular uptake of L-glutamine and its subsequent rapid efflux in the presence of essential amino acids (EAA) is the rate-limiting step that activates mTOR.

1,540 citations

Journal ArticleDOI
TL;DR: The PyMT mouse model is demonstrated to be an excellent one to understand the biology of tumor progression in humans, and its comparison to human breast tumors is compared.
Abstract: Animal models are powerful tools to analyze the mechanism of the induction of human breast cancer. Here we report a detailed analysis of mammary tumor progression in one mouse model of breast cancer caused by expression of the polyoma middle T oncoprotein (PyMT) in the mammary epithelium, and its comparison to human breast tumors. In PyMT mice, four distinctly identifiable stages of tumor progression from premalignant to malignant stages occur in a single primary tumor focus and this malignant transition is followed by a high frequency of distant metastasis. These stages are comparable to human breast diseases classified as benign or in situ proliferative lesions to invasive carcinomas. In addition to the morphological similarities with human breast cancer, the expression of biomarkers in PyMT-induced tumors is also consistent with those associated with poor outcome in humans. These include a loss of estrogen and progesterone receptors as well as integrin-beta1 expression and the persistent expression of ErbB2/Neu and cyclinD1 in PyMT-induced tumors as they progress to the malignant stage. An increased leukocytic infiltration was also closely associated with the malignant transition. This study demonstrates that the PyMT mouse model is an excellent one to understand the biology of tumor progression in humans.

986 citations

Journal ArticleDOI
TL;DR: Because of the reliance of MYC-driven cancers on specific metabolic pathways, synthetic lethal interactions between MYC overexpression and specific enzyme inhibitors provide novel cancer therapeutic opportunities.
Abstract: The MYC oncogene encodes a transcription factor, MYC, whose broad effects make its precise oncogenic role enigmatically elusive. The evidence to date suggests that MYC triggers selective gene expression amplification to promote cell growth and proliferation. Through its targets, MYC coordinates nutrient acquisition to produce ATP and key cellular building blocks that increase cell mass and trigger DNA replication and cell division. In cancer, genetic and epigenetic derangements silence checkpoints and unleash MYC9s cell growth– and proliferation-promoting metabolic activities. Unbridled growth in response to deregulated MYC expression creates dependence on MYC-driven metabolic pathways, such that reliance on specific metabolic enzymes provides novel targets for cancer therapy. Significance: MYC9s expression and activity are tightly regulated in normal cells by multiple mechanisms, including a dependence upon growth factor stimulation and replete nutrient status. In cancer, genetic deregulation of MYC expression and loss of checkpoint components, such as TP53, permit MYC to drive malignant transformation. However, because of the reliance of MYC-driven cancers on specific metabolic pathways, synthetic lethal interactions between MYC overexpression and specific enzyme inhibitors provide novel cancer therapeutic opportunities. Cancer Discov; 5(10); 1024–39. ©2015 AACR.

833 citations

Frequently Asked Questions (20)
Q1. What is the effect of serine on macropinocytosis?

Since macropinocytosis is promoted by alkalinization on the cytoplasmic surface of the plasma membrane, which alters actin polymerization and hence impacts formation of macropinosomes, the decrease in the cellular uptake of the fluorescent marker for macropinocytosis is expected in a Na+-free extracellular medium. 

The amino acid-dependent Na+/H+ exchange activity of SLC38A5 promotes macropinocytosis, which may contribute significantly to amino acid nutrition in cancer cells. 

In this paper, the authors focused on another member of the SLC38 family, namely SLC 38A5. 

Tumors with high expression of SLC38A5 might be more sensitive to such drugs than tumors with low expression of SLC38A5 ; this is however only a hypothesis at present, but it is a concept that deserves further investigation in the future. 

Because of their hydrophilic nature, amino acids cannot permeate plasma membrane by diffusion; they enter the cells via selective transporters. 

During the preincubation with NH4Cl, intracellular pH increases because of the conversion of NH3 to NH4+ inside the cells and then the pH comes down to physiological pH because of the various pH regulatory mechanisms. 

Since macropinocytosis facilitates the cellular entry of macromolecules, including proteins, present in the extracellular fluid, SLC38A5 contributes to amino acid nutrition in TNBC by two totally independent mechanisms, firstly by the traditional delivery of amino acids via the function of SLC38A5 as an amino acid transporter, and secondly by the promotion of macropinocytosis via the function of SLC38A5 as an amino acid-dependent Na+/H+ exchanger. 

RNA was used for RT-PCR to monitor the expression pattern of several amino acid transporters that might be of relevance to tumor growth. 

Considering that glutamine, glycine, and cysteine are good substrates for SLC38A5, efflux of these amino acids via the transporter might result in decreased cellular levels of the antioxidant glutathione (g-glutamylcysteinyl-glycine) in advanced tumors. 

When Na+ is removed from the medium, serine uptake does not occur via SLC38A5 because its transport activity is obligatorily coupled to Na+ (or Li+) and at the same time Na+/H+ exchanger functions in the opposite direction. 

Since SLC38A5 also functions as a Na+/H+ exchanger but in an amino acid-dependent manner, the authors wondered whether the promotion of macropinocytosis caused by serine in a NaCl-containing extracellular medium was sensitive to inhibition by amiloride. 

Amiloride and its derivatives are known inhibitors of macropinocytosis because of their ability to inhibit Na+/H+ exchanger [22, 23]. 

The authors focused on the transporters for glutamine because of its multiple biological functions in cancer cell proliferation and growth. 

Glutamine uptake and glycine uptake were also increased in the presence of Li+, but the fold-stimulation elicited by Li+ was smaller compared to what was seen with serine uptake. 

It is possible that SLC38A5 plays a role in the transport of amino acids into cancer cells during initial stages of carcinogenesis when the extracellular pH is not acidic. 

Serine uptake was measured in two buffers: (i) LiCl-buffer, pH 8.5 with 5 mM tryptophan; (ii)6NMDGCl-buffer, pH 8.5 with 5 mM tryptophan. 

BT20 cells were cultured in Eagle’s Minimum Essential Medium (ATCC, Cat. no. 30-2003); HCC1937 cells were cultured in RPMI-1640 medium (ATCC, Cat. no. 30-2001); MDA-MB436 cells were cultured in Leibovitz’s L-15 medium (ATCC, Cat. no. 30-2008), supplemented with 10 µg/ml insulin and 16 µg/ml glutathione. 

(B) Transport activity of SLC38A5 in the TNBC cell line TXBR-100 as monitored by the uptake of serine, glycine, and glutamine as the substrates for the transporter. 

For this, the authors used the NH4Cl prepulse method to cause intracellular acidification, another strategy to alter the transmembrane H+ gradient. 

In normal physiology, this potential for bidirectional function of SLC38A3 and SLC38A5 is important in the glutamine-glutamate cycle that occurs in the brain between glutamatergic neurons and the surrounding astrocytes [37, 38].