You are on page 1of 3

Cancer Cell

Previews

ROS Promotes Cancer Cell Survival


through Calcium Signaling
Colleen R. Reczek1 and Navdeep S. Chandel1,*
1Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA

*Correspondence: nav@northwestern.edu
https://doi.org/10.1016/j.ccell.2018.05.010

To avoid reactive oxygen species (ROS)-induced cell death, cancer cells increase their antioxidant defense
system. In this issue of Cancer Cell, Takahashi et al. identify a novel, non-canonical oxidative stress defense
mechanism involving TRPA1, a redox-sensitive Ca2+ channel, and the upregulation of anti-apoptotic path-
ways to promote cancer cell survival.

Cancer cells rely on the signaling capa- In this issue of Cancer Cell, Takahashi defense programs have not been
bilities of reactive oxygen species et al. report an unconventional oxidative examined.
(ROS) for cell migration, proliferation, stress defense mechanism whereby tran- Takahashi et al. (2018) systematically
and survival. To increase ROS produc- sient receptor potential ankyrin 1 (TRPA1) determined whether and how ROS-
tion, cancer cells acquire oncogenic mediates the upregulation of calcium induced TRPA1 activation and Ca2+ influx
mutations, lose tumor suppressors, and (Ca2+)-dependent anti-apoptotic path- affect the oxidative stress defense pro-
accelerate their metabolism. Hydrogen ways to prevent ROS-induced cancer grams in cancer cells to promote cancer
peroxide (H2O2), a type of ROS, pro- cell death (Takahashi et al., 2018). cell survival. The authors first examined
motes pro-tumorigenic signaling by TRPA1 is one member of the TRP family whether TRPA1 was upregulated and
mediating the cysteine oxidation of of ion channel proteins. It is a plasma functional in a variety of cancer types;
target proteins to alter their function. membrane-bound protein well known this would be expected if cancer cells
Importantly, if ROS levels are too high, for its chemosensing ability, mediating use TRPA1 to tolerate oxidative stress.
ROS can promote anti-tumorigenic neurogenic pain and inflammation (Paul- Indeed, Takahashi et al. found increased
signaling and induce cancer cell senes- sen et al., 2015). TRPA1 has been shown TRPA1 mRNA and protein expression in
cence and cell death (Reczek and Chan- to be activated by numerous noxious breast and lung tumors. Moreover, by us-
del, 2017). Therefore, it is critical for compounds, including H2O2, which oxi- ing the TRPA1 agonist mustard oil in
cancer cells to maintain an optimal dizes hyper-reactive cysteine residues combination with genetic (TRPA1 knock-
ROS level. To prevent excessive ROS on its cytoplasmic side (Bessac et al., down) and pharmacological (AP-18, a
accumulation, cancer cells increase their 2008; Paulsen et al., 2015). Activation of TRPA1-specific inhibitor) approaches,
antioxidant capacity (Gorrini et al., TRPA1 leads to the influx of Ca2+, which the authors showed that TRPA1 medi-
2013). This canonical ROS-neutralizing can serve as an important signaling mole- ates an increase in the intracellular Ca2+
mechanism includes upregulation of the cule to regulate numerous physiological concentration in numerous cancer cell
redox-sensitive transcription factor nu- and biological responses, including cell lines. Next, the authors showed that a
clear factor erythroid 2-related factor 2 proliferation and survival (Bessac et al., TRPA1-mediated Ca2+ influx was
(NRF2). NRF2 protein stabilization 2008; Déliot and Constantin, 2015; Paul- required for the increased survival of
during oxidative stress allows for the sen et al., 2015). Importantly, these TRPA1-enriched cancer cells in response
induction of numerous antioxidants, Ca2+-dependent cellular responses are to H2O2. Furthermore, ectopic expres-
including enzymes involved in gluta- also implicated in tumorigenesis, sug- sion of TRPA1 was sufficient to improve
thione (GSH) synthesis and utilization, gesting a role for TRPA1 in cancer. Previ- the cell survival of TRPA1-low-express-
glutathione peroxidase, and peroxire- ously, the antibacterial agent triclosan ing cancer cells. Using a more physiolog-
doxin (Gorrini et al., 2013). Given that was shown to induce TRPA1 activation ically relevant model, Takahashi et al.
cancer cells require ROS homeostasis, and Ca2+ influx in human prostate cancer found that the inner cells of tumor spher-
anti-cancer therapies that manipulate stromal cells, resulting in vascular endo- oids generated high levels of ROS, which
ROS levels seem attractive. Inhibiting thelial growth factor (VEGF) secretion triggered the TRPA1-mediated Ca2+
key antioxidant enzymes may elevate and increased prostate cancer epithelial influx required for inner cell survival.
ROS levels to promote oxidative stress- cell growth (Derouiche et al., 2017). Notably, TRPA1 inhibition did not alter
induced cancer cell death. However, it Moreover, through its interaction with the ROS levels in inner spheroid cells.
is important to consider with this FGFR2, TRPA1 was shown to support Thus, in a mechanism distinct from a
approach that normal, non-transformed lung adenocarcinoma cell proliferation ROS scavenging antioxidant defense
cells are also sensitive to high ROS and metastasis (Berrout et al., 2017). program, TRPA1 oxidization under high
levels; therefore, a viable therapeutic Although TRPA1 has been implicated in levels of ROS allows for an increase in
strategy should involve selectively tar- cancer, its redox-sensing capabilities Ca2+ into cells to promote cancer cell
geting cancer cells. and potential effect on oxidative stress survival.

Cancer Cell 33, June 11, 2018 ª 2018 Elsevier Inc. 949
Cancer Cell

Previews

through TRPA1. Because TRPA1 was


overexpressed relative to corresponding
normal tissue in numerous cancer types
(Takahashi et al., 2018), targeting
TRPA1, as opposed to directly manipu-
lating ROS levels through antioxidant
inhibition, may be a more selective and
effective anti-cancer therapy. To date,
several TRPA1 inhibitors have been
developed as pain and respiratory thera-
pies; however, their pharmacokinetics
are poor, with a high IC50 and a short
half-life (Chen and Hackos, 2015). Thus,
future work should be done to improve
the efficacy and utility of TRPA1 inhibi-
tors as anti-cancer agents. Combination
therapy may increase the efficacy of in-
hibiting TRPA1, because Takahashi
et al. (2018) showed in vivo that TRPA1
inhibitor treatment in combination with
carboplatin reduced tumor growth more
than either drug treatment alone.
Figure 1. ROS-Mediated TRPA1-Dependent Ca2+ Signaling Promotes Cancer Cell Survival Furthermore, because TRPA1 was
Cancer cells need to adapt to high levels of reactive oxygen species (ROS) in order to survive. The shown to be activated by ROS in a
transcription factor NRF2 induces expression of the TRPA1 channel. Hydrogen peroxide (H2O2), a type of
NRF2-dependent manner, this study
ROS produced by the mitochondria, can oxidize critical cysteine residues on TRPA1 to activate the
protein. As a result, intracellular calcium (Ca2+) concentrations increase, activating Ca2+-dependent anti- warrants the potential use of a TRPA1 in-
apoptotic and pro-survival signaling cascades involving the MCL-1 protein. Thus, ROS can promote hibitor with selectivity for hypoxic tumor
cancer cell survival through Ca2+ signaling. cells, which have high levels of ROS, or
for tumor cells with a constitutively active
To better understand how TRPA1 sup- reverse-phase protein array, the authors NRF2, like a subset of lung adenocarci-
ports cancer cell survival, Takahashi et al. identified without bias that TRPA1 noma cells. Additionally, it will be of in-
(2018) examined the role of TRPA1 in activates Ca2+-dependent pro-survival terest to determine whether other cal-
anchorage-independent growth, a key and anti-apoptotic programs involving cium channels can be activated by
feature of anoikis resistance, cell survival MCL-1 to mediate oxidative stress toler- ROS to promote cancer cell survival.
in the absence of extracellular matrix ance in cancer cells. Specifically, Nevertheless, the findings from Takaha-
(ECM) attachment, and tumor metas- TRPA1 increased the phosphorylation of shi et al. reveal the existence of a novel,
tasis. The authors used the antioxidant RAS-ERK, PI3K/AKT, and mTOR pro- ROS-controlled TRPA1-mediated cell
N-Acetyl-L-cysteine (NAC) to show that survival signaling proteins and increased survival mechanism and open up an
ECM detachment-mediated ROS pro- the expression of the anti-apoptotic pro- exciting new avenue for the treatment
duction was required for the increase in tein MCL-1 in a Ca2+-CaM/PYK2-depen- of cancer.
intracellular Ca2+ by TRPA1. Further- dent manner. Moreover, Takahashi et al.
more, in a soft agar assay, inhibition of found that TRPA1 expression in cancer ACKNOWLEDGMENTS
TRPA1 reduced the colony number and cells was directly regulated by the
average colony size of HCC1569 breast redox-sensitive antioxidant master regu- This work was supported by the NIH
cancer cells, indicating the necessity of lator NRF2. Thus, in response to H2O2, (5R35CA197532-02 to N.S.C.).
TRPA1 for anchorage-independent NRF2 is stabilized and translocated to
growth. These data suggest that TRPA1 the nucleus, where it induces the expres- REFERENCES
promotes oxidative stress tolerance and sion of both canonical (antioxidants) and
cancer cell survival in part by suppress- non-canonical (TRPA1) oxidative stress Berrout, J., Kyriakopoulou, E., Moparthi, L., Ho-
gea, A.S., Berrout, L., Ivan, C., Lorger, M., Boyle,
ing anoikis. Takahashi et al. also found defense proteins. J., Peers, C., Muench, S., et al. (2017). TRPA1-
that TRPA1-mediated Ca2+ influx pro- An intriguing finding of this study was FGFR2 binding event is a regulatory oncogenic
moted cancer cell resistance to ROS- that high levels of ROS oxidized specific driver modulated by miRNA-142-3p. Nat. Com-
mun. 8, 947.
producing therapies like carboplatin. cysteine residues of TRPA1, not to in-
Consistent with their in vitro data, the au- crease cancer cell antioxidant capacity Bessac, B.F., Sivula, M., von Hehn, C.A., Escalera,
J., Cohn, L., and Jordt, S.E. (2008). TRPA1 is a
thors used immunocompromised mice but to promote cancer cell survival
major oxidant sensor in murine airway sensory
to show that genetic knockdown and through the upregulation of Ca2+-depen- neurons. J. Clin. Invest. 118, 1899–1910.
pharmacological inhibition of TRPA1 sup- dent anti-apoptotic pathways (Figure 1).
Chen, J., and Hackos, D.H. (2015). TRPA1 as
pressed tumor growth and chemoresist- Thus, ROS can function as signaling mol- a drug target–promise and challenges. Naunyn
ance in vivo. Furthermore, by using ecules to promote cancer cell survival Schmiedebergs Arch. Pharmacol. 388, 451–463.

950 Cancer Cell 33, June 11, 2018


Cancer Cell

Previews
Déliot, N., and Constantin, B. (2015). Plasma human prostate cancer stromal cells. Cancer Reczek, C.R., and Chandel, N.S. (2017). The two
membrane calcium channels in cancer: Alter- Prev. Res. (Phila.) 10, 177–187. faces of reactive oxygen species in cancer. Annu.
ations and consequences for cell proliferation Rev. Cancer Biol. 1, 79–98.
and migration. Biochim. Biophys. Acta 1848 Gorrini, C., Harris, I.S., and Mak, T.W. (2013). Mod-
(10 Pt B), 2512–2522. ulation of oxidative stress as an anticancer strat- Takahashi, N., Chen, H.Y., Harris, I.S., Stover,
egy. Nat. Rev. Drug Discov. 12, 931–947. D.G., Selfors, L.M., Bronson, R.T., Deraedt, T.,
Derouiche, S., Mariot, P., Warnier, M., Vancauwen- Cichowski, K., Welm, A.L., Mori, Y., et al.
berghe, E., Bidaux, G., Gosset, P., Mauroy, B., Paulsen, C.E., Armache, J.P., Gao, Y., Cheng, Y., (2018). Cancer cells co-opt the neuronal redox-
Bonnal, J.L., Slomianny, C., Delcourt, P., et al. and Julius, D. (2015). Structure of the TRPA1 ion sensing channel TRPA1 to promote oxidative
(2017). Activation of TRPA1 channel by antibacte- channel suggests regulatory mechanisms. Nature stress tolerance. Cancer Cell 33, this issue,
rial agent triclosan induces VEGF secretion in 520, 511–517. 985–1003.

Epigenetic ConFUSION: SS18-SSX Fusion


Rewires BAF Complex to Activate
Bivalent Genes in Synovial Sarcoma
Marc A. Morgan1 and Ali Shilatifard1,*
1Simpson Querrey Center for Epigenetics, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of

Medicine, 320 E. Superior St., Chicago, IL 60611, USA


*Correspondence: ash@northwestern.edu
https://doi.org/10.1016/j.ccell.2018.05.011

In this issue of Cancer Cell, McBride and colleagues report that the synovial sarcoma SS18-SSX fusion drives
BAF complex recruitment to bivalent domains repressed by PRC2 complex to orchestrate aberrant transcrip-
tional activation. Redistribution of BAF localization is a major driver of synovial sarcoma proliferation and pre-
sents a promising therapeutic target.

‘‘Uninvited guests are often most SSX’s deviant gain-of-function behavior SSX’s status as a bona fide oncoprotein
welcome when they leave.’’ is the primary requirement for halting (Haldar et al., 2007), until recently it re-
synovial sarcoma proliferation, rather mained largely unknown how SS18-SSX
-Aesop
than restoring BAF47 (also known as drives synovial sarcoma formation. In
SMARCB1, SNF5, or INI1) to the BAF 2013, Kadoch and Crabtree published
A group of friends (the BAF complex) has complex. These findings shed new light their breakthrough discovery that SS18-
gathered for a dinner party, but on this onto synovial sarcoma pathogenesis and SSX alters the composition of BAF com-
particular evening, one member (SS18) have important implications for the devel- plexes by evicting both wild-type SS18
has brought along an uninvited and prob- opment of targeted therapies. and the tumor suppressor BAF47, thus
lematic guest (SSX). One friend (BAF47) Synovial sarcoma is a soft-tissue tumor earmarking them for proteolytic degrada-
cannot abide this unwelcome new- characterized by a reciprocal t(X;18) tion (Kadoch and Crabtree, 2013). These
comer’s antics and leaves the party. The translocation involving the SS18 gene biochemical alterations are accompanied
interloper hijacks the conversation and and one of three related SSX genes by the transcriptional activation of
takes it to inappropriate areas (Bivalent (SSX1, SSX2, or SSX4) that occurs pre- oncogenic factors such as SOX2, which
Polycomb repressed genes). This derails dominantly in young adults. SS18-SSX fu- are normally targeted for repression by
the entire evening and leaves a lingering sions contain most of the SS18 protein the Polycomb Repressive Complex 2
malignant mood. This allegorical scene sequence followed by additional amino (PRC2). Importantly, removing SS18-
describes molecular mechanisms for the acids derived from SSX. SS18 normally SSX restores wild-type SS18 and BAF47
intruding SS18-SSX fusion oncoprotein participates as a component of the to the BAF complex and is sufficient to
in synovial sarcoma, as revealed in this multi-subunit BAF (mammalian SWI/ inhibit synovial sarcoma proliferation
issue of Cancer Cell (McBride et al., SNF) ATP-dependent chromatin-remod- (Kadoch and Crabtree, 2013), thus mak-
2018). Expelling the unwanted visitor, eling complex, thus implicating the dereg- ing SS18-SSX an attractive therapeutic
SS18-SSX, restores normal BAF complex ulation of chromatin-based gene-regula- target because of its involvement in both
function and reestablishes Polycomb tory systems as a central driver of tumor initiation and tumor maintenance.
repression of bivalent genes. Importantly, synovial sarcoma pathogenesis (Kadoch In their current study, McBride and col-
the authors reveal that eliminating SS18- and Crabtree, 2015). Despite SS18- leagues use patient-derived cell lines to

Cancer Cell 33, June 11, 2018 ª 2018 Published by Elsevier Inc. 951

You might also like