ArticlePDF Available

Identification of Frataxin as a regulator of ferroptosis

Authors:
  • Zhejiang Provincial People’s Hospital

Abstract and Figures

Ferroptosis is a newly discovered form of non-apoptotic regulated cell death and is characterized by iron-dependent and lipid peroxidation. Due to the enhanced dependence on iron in cancer cells, induction of ferroptosis is becoming a promising therapeutic strategy. However, the precise underlying molecular mechanism and regulation process of ferroptosis remains largely unknown. In the present study, we demonstrated that the molecular protein Frataxin (FXN) is a key regulator of ferroptosis by modulating iron homeostasis and mitochondrial function. Suppression of FXN expression specifically repressed the proliferation, destroyed mitochondrial morphology, impeded Fe–S cluster assembly and activated iron starvation stress. Moreover, suppression of FXN expression significantly enhanced erastin-induced cell death through accelerating free iron accumulation, lipid peroxidation and resulted in dramatic mitochondria morphological damage including enhanced fragmentation and vanished cristae. In addition, this type of cell death was confirmed to be ferroptosis, since it could be pharmacologically restored by ferroptotic inhibitor Fer-1 or GSH, but not by inhibitors of apoptosis, necrosis. Vice versa, enforced expression of FXN blocked iron starvation response and erastin induced ferroptosis. More importantly, pharmacological or genetic blocking the signal of iron starvation could completely restore the resistance to ferroptosis in FXN knockdown cells and xenograft graft in vivo. This paper suggests that FXN is a novel ferroptosis modulator, as well as a potential provided target to improve the antitumor activity based on ferroptosis.
Content may be subject to copyright.
Contents lists available at ScienceDirect
Redox Biology
journal homepage: www.elsevier.com/locate/redox
Identication of Frataxin as a regulator of ferroptosis
Jing Du
a,1
, Yi Zhou
c,f,1
, Yanchun Li
c,1
, Jun Xia
a
, Yongjian Chen
a
, Sufeng Chen
a
, Xin Wang
d
,
Weidong Sun
e,h
, Tongtong Wang
f
, Xueying Ren
a
, Xu Wang
g
, Yihan An
h
, Kang Lu
h
, Wanye Hu
h
,
Siyuan Huang
c
, Jianghui Li
g
, Xiangmin Tong
a,b,c,d,g,h,∗∗
, Ying Wang
b,d,h,
a
Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
b
Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
c
The Second Clinical Medical School of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
d
Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
e
Department of Hematology, Shaoxing Central Hospital, Shaoxing, Zhejiang, 312030, China
f
Department of Wangjiangshan, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
g
School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
h
Bengbu Medical College, Bengbu, Anhui, 233000, China
ARTICLE INFO
Keywords:
Ferroptosis
Frataxin
Iron-sulfur cluster
Mitochondria
ABSTRACT
Ferroptosis is a newly discovered form of non-apoptotic regulated cell death and is characterized by iron-de-
pendent and lipid peroxidation. Due to the enhanced dependence on iron in cancer cells, induction of ferroptosis
is becoming a promising therapeutic strategy. However, the precise underlying molecular mechanism and
regulation process of ferroptosis remains largely unknown. In the present study, we demonstrate that the protein
Frataxin (FXN) is a key regulator of ferroptosis by modulating iron homeostasis and mitochondrial function.
Suppression of FXN expression specically repressed the proliferation, destroyed mitochondrial morphology,
impeded FeS cluster assembly and activated iron starvation stress. Moreover, suppression of FXN expression
signicantly enhanced erastin-induced cell death through accelerating free iron accumulation, lipid peroxidation
and resulted in dramatic mitochondria morphological damage including enhanced fragmentation and vanished
cristae. In addition, this type of cell death was conrmed to be ferroptosis, since it could be pharmacologically
restored by ferroptotic inhibitor Fer-1 or GSH, but not by inhibitors of apoptosis, necrosis. Vice versa, enforced
expression of FXN blocked iron starvation response and erastin-induced ferroptosis. More importantly, phar-
macological or genetic blocking the signal of iron starvation could completely restore the resistance to ferrop-
tosis in FXN knockdown cells and xenograft graft in vivo. This paper suggests that FXN is a novel ferroptosis
modulator, as well as a potential provided target to improve the antitumor activity based on ferroptosis.
1. Introduction
Despite success in chemotherapy drugs clinically, drug toxicity and
resistance continue to be the principal limiting factor to achieving cures
in patients with cancer. Activation of regulated cell death and ex-
ploration therapy targets with less toxicity is a potential anticancer
treatment strategy. Numerous therapy targets have been discovered for
that cancer is a heterogeneous disease dened by various genetic and
epigenetic variations. But little is known about the molecular biological
characteristics of mitochondrial protein Frataxin (FXN) in cancer.
Iron is an indispensable element for various metabolic and physio-
logical functions in living organisms. It functions as a cofactor for vital
iron-containing enzymes that are involved in DNA synthesis, ATP pro-
duction, heme synthesis and many other physiological activities [1]. In
general, a certain amount of iron is crucial for cell survival, growth and
proliferation. Therefore, iron deciency is undoubtedly deleterious,
leading to a multisystem disorder with innumerable eects, including
anemia [2]. Conversely, too much iron is also detrimental. The aberrant
accumulation of iron causes excess free radical generation through
Fenton reaction and subsequent results in signicant alterations in the
https://doi.org/10.1016/j.redox.2020.101483
Received 27 November 2019; Received in revised form 15 February 2020; Accepted 28 February 2020
Corresponding author. Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou,
Zhejiang, 310014, China.
∗∗
Corresponding author. Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou,
Zhejiang, 310014, China.
E-mail addresses: tongxiangming@hmc.edu.cn (X. Tong), wangying@hmc.edu.cn (Y. Wang).
1
These authors contributed equally to this work.
Redox Biology 32 (2020) 101483
Available online 02 March 2020
2213-2317/ © 2020 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
T
cellular redox homeostasis, thus leading to the damage of DNA, proteins
or other biomolecules. Interestingly, relative to healthy cells, cancer
cells do exhibit an enhanced dependence on iron to enable its growth,
which may act as a target for cancer therapy [3]. It is noted that iron
overload can cause a specic cell death termed as ferroptosis, which is a
recently discovered form of non-apoptotic regulated necrosis [4]. Fer-
roptosis has been implicated in the pathological process associated with
carcinogenesis, degenerative diseases, stroke, and kidney ischemia/re-
perfusion (I/R) injury [58]. However, the precise underlying mole-
cular mechanism and potential anticancer therapeutic signicance of
ferroptosis remain to be elucidated.
The execution of ferroptosis closely involves the iron-dependent
peroxidation of polyunsaturated fatty acids (PUFAs) [9]. Since mi-
tochondria employ the majority of cellular iron, it plays a critical role in
the iron homeostasis. The excess iron leads to a disturbance in mi-
tochondrial dynamics and even results in ferroptosis. The cystine/glu-
tamate antiporter inhibitor erastin and GPX4 inhibitor RSL3 accelerate
the progression of ferroptosis, which inhibited by the iron chelator,
GSH, ferrostatin-1 and its analogs. In addition, degradation of ferritin
by ferritinophagy can release free iron and initiate the process of fer-
roptosis [10,11]. Cancer cells harboring oncogenic Ras exhibit sensi-
tivity to ferroptosis and can be selectively killed. Our previous study has
provided experimental evidence that dihydroartemisinin, the semi-
synthetic derivatives, represents a promising therapeutic eect to pre-
ferentially target AML cells by inducing ferroptotic cell death [12].
Daolin Tang and co-workers also discovered the ferroptosis inducer
erastin enhanced the sensitivity of AML cells to chemotherapeutic
agents [13]. Here we focus on exploring targets to improve the sensi-
tivity for ferroptosis in cancer cells.
The ancient and conservative cofactors, iron-sulfur cluster (ISC), are
essential for biological processes such as redox reactions, iron home-
ostasis, enzymatic catalysis, heme synthesis and regulation of gene
expression [14]. More than 60 human proteins bind with dierent types
of ISCs to execute their functions. ISC assembly genes encode in the
nuclear genome and translocate into mitochondria for creating cofac-
tors. In humans, the de novo of FeS cluster biogenesis is initiated with
the removal of sulfur from cysteine by the cysteine desulfurase complex
Nfs1-Isd11, a process activated by frataxin [15]. The removed persul-
de intermediately reduces to sulde with the accomplished of ferre-
doxin reductase and ferredoxin 2, and delivers to scaold protein ISCU
to form a [2Fe2S] cluster with ferrous iron (Fe
2+
). Neosynthesized
ISCs are then transferred to mitochondrial recipients or further as-
sembled into [4Fe4S] with the help of the HSC20/HSPA9 chaperone
system [16]. The exact molecular function of FXN is unclear, but it has
been proposed to be an iron chaperone or an allosteric activator of
cysteine binding to NFS1 [15]. Defects in the biogenesis of Fe/S protein
are associated with human disorders, which called FeS diseases. An
expansion of the GAA repeat in the rst intron of the FXN gene would
result in the diminished expression of the encoded protein, thus leading
to Friedreich's ataxia (FRDA), a most common recessive ataxia in the
Caucasian population. Complete depletion of FXN is embryonically le-
thal in mice demonstrating its extremely vital biological function [17].
Decreased expression of FXN in FRDA is characterized as mitochondrial
iron accumulation, mitochondrial dysfunction and increased oxidative
stress [18]. The newly published paper demonstrated that ferroptosis
inhibitors might have therapeutic potential in primary FRDA patient-
derived broblasts [19]. Despite the central role of FXN in FeS as-
sembly and FRDA, it remains unclear whether it plays a pivotal role in
cancer biologic behavior and prognosis.
Indeed, our study demonstrated suppression of FXN expression
specically destroyed mitochondrial morphology, impeded FeS cluster
assembly and enhanced cysteine deprivation-induced cell death in HT-
1080 cells. This type of cell death could be pharmacologically restored
by ferroptotic inhibitor Fer-1 or GSH, but not by inhibitors of apoptosis,
necrosis. Moreover, the observed phenotypes of increased lipid ROS,
accumulation of free iron and mitochondria dysfunction could be
rescued by ectopic expression of FXN. Additionally, pharmacological or
genetic blocking the signal of iron starvation could completely restore
the resistance to ferroptosis in FXN knockdown cells. This study is the
rst to discover the role of FXN in molecular biological characteristics
of cancer and to identify the link between FXN and ferroptosis.
Meanwhile, our work sheds new light on the molecular mechanisms
underlying FXN induced ferroptosis through mitochondrial dysfunction
as well as activation of iron starvation response, and discoveries that
the ISC assembly is tightly correlated with the cancer progression,
which can be developed as a potential therapeutic strategy.
2. Materials and methods
2.1. Antibodies and reagents
The antibody to Frataxin (ab219414), to Aconitase 2 (ab129069), to
Aconitase 1 (ab126595), to IREB2 (ab181153), to NDUFV2
(ab183717), to FECH (ab137042), to Transferrin Receptor (ab80194),
SDHB (ab14714), to Ferritin Heavy Chain (ab65080), to beta Actin
(ab8226) and N-acetylcysteine (NAC), glutathione (GSH) and DCFH-DA
were obtained from Abcam (Cambridge, MA). CCK-8 Assay Kit was
obtained from Meilunbio (Dalian, China). CFDA-SE, GSH Assay Kit was
purchased from Beyotime (Shanghai, China). Erastin, Sorafenib,
Ferrostatin-1, Z-VAD-FMK, Necrosuifonamide were obtained from
Selleck Chemicals (Houston, TX). C11-BODIPY (581/591), TMRE,
MitoTracker were obtained from Thermo Fisher Scientic (Waltham,
MA). The Cell Cycle Staining Kit was purchased from MultiSciences
(Hangzhou, China).
2.2. Cell culture
Human brosarcoma HT-1080 cells were obtained from the Cell
Bank of Chinese Academy of Sciences (Shanghai, China) and cultured in
DMEM medium (Hyclone, Logan, UT, USA) containing 10% fetal bo-
vine serum (Gibco, Grand Island, NY, USA), supplementary with 100 U/
mL penicillin and 100 μg/ML streptomycin. Cells were maintained in an
incubator with a humidied atmosphere of 5% CO
2
at 37 °C.
2.3. Cell viability assay
Cell viability assay was evaluated using the CCK8 Assay Kit. HT-
1080 cells were seeded at a density of 4 × 10
4
cells/well in 100 μl
DMEM medium in the 96-well plate for overnight, followed by treat-
ment with dierent doses of erastin for 12 h. 10 μl CCK-8 was added to
each well and the cells were subsequently incubated at 37 °C for 2 h.
Absorbance was measured at 450 nm using the microplate reader.
2.4. PI staining
Cells were plated in 96-well plates at a density of 4 × 10
4
cells/well
and treated with the indicated concentration of erastin. Propidium io-
dide (PI) was then added to each well at a nal concentration of 10 μg/
mL for 10 min in the dark, and cells were subjected to uorescence
microscopy.
2.5. Transwell assay
HT-1080 cells (4 × 10
4
) in 200 μl serum-free DMEM medium were
seeded onto the upper transwell chambers, while 500 μl medium con-
taining 5% FBS was added to the lower chambers. The cells were cul-
tured at 37 °C in an incubator for 24 h. The transwell chambers were
then removed, washed with PBS twice, xed in 4% paraformaldehyde
for 30 min and stained with 0.1% crystal violet for 20 min at room
temperature. Next, PBS was used to wash chambers, and the un-mi-
grated cells in the upper transwell membrane were gently wiped o
using a cotton swab. Images were observed and photographed using an
J. Du, et al. Redox Biology 32 (2020) 101483
2
inverted light microscope.
2.6. Cell growth assay
HT-1080 cells were seeded in a 96-well plate at a density of
3×10
3
cells/well in 100 μl DMEM medium containing 10% FBS. The
cells were incubated overnight at 37 °C in an incubator. From the
second day after plating, the numbers of cells were measured at the
specied time using the CCK8 Assay Kit. Next, a microplate reader was
used to measure the absorbance at 450 nm.
2.7. Cell proliferation assay
Cell proliferation was detected using a CFDA-SE probe. Briey, HT-
1080 cells stained with CFDA-SE according to the manufacturer's pro-
tocol and seeded in 6-well plates. Then, cells were exposed to the same
concentration of erastin. CFDA-SE uorescence was detected by the
ow cytometry.
2.8. Cell cycle analysis
Cell cycle proles were examined by the Cell cycle staining Kit
(MultiSciences, Hangzhou, China) according to the manufacturer's in-
structions. Briey, cells were harvested, washed and xed in ice-cold
70% ethanol at 4 °C overnight. Then they were washed, treated with
100 μl RNase A at 37 °C for 30 min and stained with 400 μl PI at 4 °C in
the dark for 30 min. Finally, the cells were washed and analyzed by
ow cytometry.
2.9. Colony formation assay
3×10
3
cells were seeded in a 6-well plate. After incubation for 2
weeks, the colonies were washed with PBS, xed with 4% paraf-
ormaldehyde, and then stained with crystal violet. The colonies were
photographed by the inverted microscope.
2.10. Soft agar colony formation assay
6-well plates were covered with a bottom layer of 2 ml 0.8% agar in
10% FBS-DMEM. Then 3 × 10
3
cells were suspended in 2 ml of 10%
FBS-DMEM containing 0.4% agarose and seeded to the top of a 0.8%
agarose. After two weeks, colonies were xed with 4% paraformalde-
hyde for 30 min, stained with dilute crystal violet for 20 min. The co-
lonies were photographed by the inverted microscope.
2.11. Determination of ROS production
ROS levels in cells were measured by using 2ʹ,7ʹ-dichlorouore scin
diacetate (DCFH-DA). After treatment with erastin(5 μM), cells were
washed with serum-free culture medium and incubated with 4 μM
DCFH-DA in the dark at 37 °C for 30 min. Cells were then washed and
harvested, suspended in serum-free culture and uorescence intensity
was measured by ow cytometry.
2.12. Lipid peroxidation
Lipid peroxidation was measured with C11-BODIPY (581/591).
When lipid peroxidation increased, the uorescence is undergoing a
shift from red to green uorescence emission. HT-1080 cells in 6 well
plates were incubated with the indicated concentrations of erastin.
2.5 μM C11-BODIPY(581/591) was added and incubated at 37 °C for
30 min. Excess C11-BODIPY was removed by washing the cells twice
with Hanks buer. Cells were harvested and resuspended for ow cy-
tometry analysis.
2.13. Confocal microscopy assay
The cells were seeded in a chamber confocal dish and treated with
the indicated concentration of erastin. Then 581/591 C11- BODIPY
(5 μM) or MitoTracker(100 nM) or RPA(4 μM) were co-staining with
DAPI(10 mg/ml) in the dark for a 30 min incubation. Representative
images were viewed under a confocal microscope.
2.14. Measurement of mitochondrial membrane potential (MMP)
The changes of mitochondrial membrane potential (MMP) were
observed by tetramethylrhodamine methyl ester (TMRE) probe. HT-
1080 cells were treated with 5 μM erastin for 12 h and then stained with
TMRE (100 nM) for 30 min. The stained cells were washed with PBS
twice, and analyzed using the ow cytometry.
2.15. Iron assay
The mitochondrial chelatable iron pool was assessed using
Rhodamine B-[(1, 10-phenanthroline-5-yl)-aminocarbonyl]benzyl ester
(RPA), a Fe
2+
specicuorescent sensor. After treatment, cells were
harvested and incubated with 2 μM RPA for 15 min at 37 °C in Hanks
balanced salt solution(HBSS), then washed subsequently three times
with HBSS. The samples were incubated with dye-free HBSS for another
15 min following washing once with HBSS. Mitochondrial iron was
measured using ow cytometry.
2.16. Determination of iron concentrations in isolated mitochondria and
cytoplasm by ICP-MS
Total iron concentration was measured using inductively coupled
plasma mass spectrometry (ICP-MS, Agilent, Varian) as previously de-
scribed [20]. In brief, indicated cells were washed, trypsinized and
subjected to the subcellular fractions isolation kit (Beyotime, Shanghai,
China). Fractions were lysed and quantied by BCA Protein Assay Kit.
Each aliquot of the lysed sample was prepared for ICP-MS analyses.
Quadruplicate determinations of iron concentration were performed for
each sample. The results were conversions from raw ppb values and
expressed as pmol/100 mg protein.
2.17. GSH detection
Intracellular GSH levels were examined by using a GSH Assay Kit
(Beyotime, Shanghai, China). HT-1080 cells were treated with erastin
for 24 h. Then, the subsequent procedures were performed according to
the manufacturer's instructions. The experimental data were obtained
by a microplate reader.
2.18. Transmission electron microscope (TEM)
After indicated treatment, The cells were xed by the 2.5% glutar-
aldehyde solution at 4 °C overnight. After xation, the samples were
dehydrated by a graded series of ethanol, then dehydrated by alcohol
and eventually transferred to absolute acetone. Following Inltration
with absolute acetone and the nal Spurr resin mixture, the samples
were embedded, ultrathin sectioned and stained. Finally, the samples
were observed in the Hitachi Model H-7650 transmission electron mi-
croscope.
2.19. Plasmids
For knockdown of FXN, target shRNA sequences were subcloned
into pLVX-shRNA Lentivector (Takara). The two shRNA knockdown
sequences for FXN were forward: 5- GATCCGCTGGACTCTTTAGCAG
AGTTTTCAAGAGAAACTCTGCTAAAGAGTCCAGCTTTTTTG-3, and 5-
GATCCGCAGACGCCAAACAAGCAAATTTCAAGAGAATTTGCTTGTTTG
J. Du, et al. Redox Biology 32 (2020) 101483
3
GCGTCTGCTTTTTTG-3. Human full-length FXN or FTH cDNA was
amplied by RTPCR using HEK-293 mRNA and veried by sequen-
cing. Then the cDNA was subcloned into pLVX -Neo lentivirus vector
(Takara, Dalian, China) by ClonFast Seamless Cloning kit (obio,
Nanjing, China). The plasmid ofshRNA resistant form of FXN (Res-FXN)
was generated according to the described methods by introduced silent
changes in the coding region targeted by the shRNA [21].
2.20. Lentiviral transduction
The recombinant lentiviral plasmids were veried by sequencing
and co-transfected with pMD2G, pSPAX2 into HEK293 cells to produce
recombinant lentiviral. Lentivirus infections were carried out as de-
scribed previously [12]. Briey, the cell seeded in 24-well plates
reached 7080% conuence, the 10%-DMEM medium was removed.
Cells were then transfected with the corresponding lentivirus. After two
days, puromycin or G418 were added for screening . Then the stable
cells were maintained in puromycin or G418. The expression eciency
was evaluated by RT-PCR and western blot analysis.
2.21. Western blotting
Following treatment, the cells were lysed in RIPA buer after
washing with PBS and incubated on ice for 30 min. Then cellular debris
was removed by centrifugation and the protein concentration was
quantied with BCA Protein Assay Kit. Subsequently, equal amounts of
protein were separated by SDSPAGE and transferred to PVDF mem-
branes. The membranes were blocked with 5% skim milk for 1 h and
incubated with the primary antibodies at 4 °C overnight. After washing
three times with TBST, the membranes were incubated with the sec-
ondary antibodies at room temperature for 1 h and washed again. The
blots were visualized using a chemiluminescence detection kit ECL-
PLUS.
2.22. RNA isolation and quantitative real-time PCR (RT-PCR)
Cells were lysed using Trizol reagent (Invitrogen, USA) and total
RNA was extracted with chloroform and isopropyl alcohol. cDNA was
then synthesized using a reverse transcription reagent kit (TaKaRa,
Dalian, China) according to the manufacturer's protocols. The SYBR
Green Master Mix Kit was used for relative quantication of RNA levels
according to the manufacturer's instructions. GAPDH was chosen as an
internal control. The sequences of the primers were as follows: GAPDH,
forward, 5-GCACCGTCAAGGCTGAGAAC, reverse, 5-ATGGTGGTGAA
GACGCCAGT; FXN, forward, 5-TAGCAGAGGAAACGCTGGAC, reverse,
5-ACGCTTAGGTCCACTGGATG. The expression level was normalized
to the internal control and determined by a 2
-ΔΔCT
method.
2.23. Determining mitochondrial DNA (mtDNA) copy number
Quantitation of the mitochondrial DNA copy number relative to the
nuclear DNA was carried out by using real-time PCR. Primer specic for
HGB1 genes were used for the determination of nuclear DNA (nDNA).
This primer sequences were used as follows: forward primer, 5-GTGC
ACCTGACTCCTGAGGAGA-3; reverse primer, 5-CCTTGATACCAACCT
GCCCAG-3. And another primer (ND-1) for the detection of mtDNA.
The primer sequences were as follows: forward primer, 5-CCCTAAAA
CCCGCCACATCT-3; reverse primer, 5-GAGCGATGGTGAGAGCTAA
GGT-3. Q-PCR was performed and the mtDNA copy number was cal-
culated. The thermal cycling conditions for the nDNA and mtDNA
amplication were 95 °C for 5 min, followed by 40 cycles of 95 °C for
15 s, 55 °C for 15 s, and 72 °C for 1 min.
2.24. Mouse xenograft model
46 weeks old male BALB/c nude mice were used to construct
xenograft models. 2.5 × 10
6
HT-1080 cells suspended in 0.1 mL PBS
were injected subcutaneously into the nude mice. After 7 days, tumor
growth was detectable and monitored every 2 days. Tumor volume in
mm
3
was determined by measuring the longest diameter (a) and
shortest width (b) and calculated by using the following formula: vo-
lume (mm
3
)=0.5×a×b
2
. On the 12th day, mice were euthanized
and tumors were isolated.
2.25. H&E analysis
Tumors collected from mice were xed in 4% paraformaldehyde.
The paran-embedded samples were cut to 4 μm thickness and stained
with H&E (Sigma). Stained sections were viewed and photographed
under a microscope.
2.26. Immunohistochemistry (IHC)
Tissues were xed with 4% paraformaldehyde and embedded in
paran. The paran-embedded block tissues were cut into 4 μm sec-
tions and followed dewaxed, hydrated and antigen retrieval. After
washing with PBS three times, the slides were treated with 3% hy-
drogen peroxide for 15 min, washed with PBS, blocked with BSA for
15 min at room temperature. Subsequently, anti-FXN antibody (1:200),
anti-FTH antibody (1:100) were added to the sections at 4 °C for
overnight. The streptavidin peroxidase method was used for signal
detection and then stained by diaminobenzidine (DAB) and counter-
stained with hematoxylin. The sections were observed and photo-
graphed under the light microscope. All slides were scored by two in-
dependent observers in a blinded fashion.
2.27. Statistical analysis
All statistical calculations were performed using GraphPad Prism
(version 7.0). All results were presented as the mean ± standard de-
viation (SD). The dierences between the two groups were performed
by the Student's t-test. Comparisons among multiple groups were ana-
lyzed by the one-way ANOVA. P< 0.05 was considered to be sig-
nicant.
3. Results
3.1. FXN knockdown repressed the proliferation of HT-1080 cells
FXN stable knockdown cells were generated through lentivirus
transduction by two separate short hairpin RNA sequences. Western
blot and real-time PCR were used to detect FXN expression. As ex-
pected, the knockdown approach dramatically reduced RNA levels and
protein expression of FXN in the shRNA cells (Fig. 1A, B). Using these
cells, we found that FXN depletion did not lead to cell death, but instead
inhibited cellular proliferation (Fig. 1C). To further determine whether
the decreased cell proliferation was aected by the cell cycle distribu-
tion or not. Cell cycle distribution assay was performed followed by
ow cytometry and showed cells were arrested at G0/G1 phase
(Fig. 1D, E). Furthermore, the capacity of long-term cell viability be-
tween the stably FXN knockdown and control cells was detected by
colony formation and soft agar assay (Fig. 1F-I). The results showed
FXN suppressed signicantly inhibited the capacity of colony forma-
tion. Besides, transwell assays were performed to determine the mi-
gration of HT-1080 cells and revealed that FXN knockdown dramati-
cally inhibited the migration of HT-1080 cells (Fig. 1J, K). Notably, the
high FXN expression indicated a signicantly poorer overall survival
rate in sarcoma, acute myeloid leukemia, bladder urothelial carcinoma,
and adrenocortical carcinoma patients based on The Cancer Genome
Atlas (TCGA) datasets (Fig. S1). Taken together, these results discover
that FXN plays a vital role in the molecular biological characteristics of
cancer and might be a potential indicator of poor prognosis.
J. Du, et al. Redox Biology 32 (2020) 101483
4
3.2. FXN knockdown induced the dysfunction of mitochondria and
accumulation of free iron
As FXN is a conserved mitochondrial protein, we next examined the
mitochondrial function in the FXN suppressed HT-1080 cells. Firstly,
mitochondria were labeled with the mitotracker probe and observed by
a confocal laser scanning microscope. The control cells showed a net-
work of elongated mitochondria, in contrast, the FXN knockdown cells
appeared fragmentation and accumulation around the nucleus, in-
dicating that FXN depletion resulted in the dysfunction of mitochon-
drial homeostasis (Fig. 2A). For the close association of mitochondrial
morphology with functionality, we next monitored the changes of mi-
tochondrial membrane potential (MMP) through TMRE staining fol-
lowed by ow cytometry. The results conrmed that the MMP of HT-
1080 cells with two dierent FXN knockdown sequences appeared
signicant hyperpolarization (Fig. 2B). Earlier published paper has
proved that excessive elevating of MMP does not help to accelerate ATP
production, but assist the production of free radicals exponentially
[22].
The above studies demonstrate that FXN plays a role in maintaining
mitochondrial function; it is unclear the regulation loop between FXN
and the activity of oxidative phosphorylation. We next tested the mi-
tochondrial oxygen consumption rate (OCR) by extracellular ux ana-
lyzer, which reected the main mitochondrial function of energy re-
spiration. As showed in Fig. 2C, knockdown of FXN remarkably
decreased the oxygen consumption in mitochondria. The basal re-
spiration, maximal respiration and spare respiration, three indices that
represent mitochondrial respiration ux revealed a signicant decline,
demonstrating that FXN knockdown disrupted the process of oxidative
phosphorylation (Fig. 2D, E, F). Moreover, ATP production was also
calculated and a pronounced loss of ATP exhibited in the FXN knock-
down cells, which was unable to meet the cellular ATP demands for
supporting cell growth (Fig. 2G). Collectively, the mitochondrial OCR
tested by Seahorse XF96 Analyzer uncovered an important link between
FXN and oxidative phosphorylation.
Besides energy respiration, ISC assembly is another important pro-
cess that takes place in mitochondria and considered to be essential for
viability. Thus, ISC turnover in iron-sulfur protein was monitored in
several ways. Above all, the activity of mitochondrial and cytoplasmic
aconitase, important [4Fe4S] proteins that catalyze the reaction from
Fig. 1. FXN knockdown repressed the proliferation
of HT-1080 cells. (A) Western blot analysis of FXN
expression in indicated FXN knockdown and control
HT-1080 cells. Tom20 was served as internal
loading control. (B) Quantitative real-time PCR
analysis of the mRNA of FXN in the three indicated
HT-1080 cells. (C) The ability of proliferation was
detected by CCK8 assay between control and sh-
FXN cells. Induction of cycle arrest in HT-1080 cells
was detected by ow cytometry (D-E). Eects of
FXN suppression in HT-1080 cells on the prolifera-
tion capacity were measured by plate clone forma-
tion assay (F-G) and soft agar colony formation
assay (H-I), corresponding histograms were shown
on the right . Migration of HT-1080 cells was eval-
uated by transwell assay; corresponding histograms
were shown on the right (J-K). All histograms were
represented as mean ± SD.
P < 0.05,
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
5
citric acid to isocitrate, were analyzed by the in-gel activity assay. And
the breakdown of mitochondrial ISC assembly resulted in a more dra-
matic decrease of cytoplasmic aconitase activity than m-aconitase ac-
tivity, without obvious changes of protein level, demonstrating FXN
depletion promoted the loss of the aconitase ISCs (Figs. 2H, I, S2A). It
has been reported cellular iron-responsive protein1 (IRP1) and IRP2
play a central role in the regulation of iron metabolism. Apo form of
cytoplasmic aconitase transformed into IRP1, which could promote the
degradation of ferritin heavy chain (FTH1) mRNA and stabilize trans-
ferrin receptor 1 (TFR1) mRNA through IRP-IRE mechanism, thus
leading to the iron starvation response. Increased iron starvation re-
sponse was also conrmed by the upregulated IRP2, TFR and down-
regulated FTH (Fig. 2J). For the importance of FXN in the de novo ISC
biogenesis and impaired mitochondrial OXPHOS in FXN suppressed
cells, we also measured the expression of SDHB and NDUFV2, subunits
of complexes I and II whose electron transfer function rely on the intact
ISC cluster. Pronounced depletion of SDHB and NDUFV2 was detected
in FXN knockdown cells. In addition, the [2Fe2S] binding FECH, a
rate-limiting enzyme in heme biosynthesis, and lipoic acid harbored in
PDH E2, KGDH E2 which represented the activity of [4Fe4S] binding
lipoic acid synthase were also signicantly decreased (Fig. 2K). These
data conclude that FXN depletion reduce steady-state levels of FeS
cluster dependent proteins as well as reactions and subsequent activa-
tion of the iron starvation stress. Besides, in line with previous studies
that NRF2, an important transcription factor in regulating cellular
redox homeostasis, was downregulated in FXN suppressed cells
[23,24]. The mechanism involved may be transcription-independent
(protein-protein interaction), for the increased levels of cytosolic Kelch-
like ECH-associated protein 1 and a similar level of NRF2 mRNA under
FXN suppression (Fig. S 2B, C).
Fig. 2. Suppression of FXN induced the dysfunction
of mitochondria and accumulation of free iron. (A)
To evaluate mitochondrial morphology, HT-
1080 cells were stained with MitoTracker probe
(100 nM) and DAPI (10 mg/ml), and then photo-
graphed by confocal laser microscope. Scale bars:
75 μm. (B) Flow cytometry was performed to mea-
sure mitochondrial membrane potential (MMP) by
TMRE probe (100 nM) in indicated FXN knockdown
and control HT-1080 cells. (C) OCR rate was carried
out by extracellular ux analyzer after the additions
of oligomycin, FCCP and antimycin A. (D-G) The
basal respiration, maximal respiration, spare re-
spiration and ATP production were calculated. (H)
Mitochondrial and cytoplasmic aconitase activity
was analyzed by the in-gel activity assay. (I) The
protein levels of mitochondrial and cytoplasmic
aconitase were detected by western blot assay. β-
actin and Tom20 were used as a loading control. (J-
K) Western blot analysis of iron-starvation stress
and ISC turnover in iron-sulfur protein. (L) HT-
1080 cells were stained with Rhodamine B-[(1,10-
phenanthroline-5-yl)-aminocarbonyl]benzyl ester
(RPA) probe (4 μM) and DAPI (10 mg/ml) to detect
the cellular labile iron by confocal laser microscope.
Scale bars: 75 μm. (M) The quantitative PCR ana-
lysis was performed to measure mitochondrial DNA
copy number. Primer specic for HGB1 genes were
used for the determination of nuclear DNA (nDNA)
and another primer (ND-1) for the detection of
mtDNA.
P < 0.05,
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
6
We next used the selective yield uorescence probe RPA, whose
cationic uorophore could rapidly quench by Fe
2+
ions, to detect the
cellular labile iron. As showed in Fig. 2L, FXN knockdown induced a
pronounced accumulation of free Fe
2+
ions with the decreased uor-
escence of RPA. The quantitation of the RPA uorescence was shown in
Fig. S2D. For the observed reduction of OCR, we subsequently mon-
itored the mtDNA copy number, which encodes 13 polypeptides playing
crucial roles in the OXPHOS. Consistent with this nding, the mtDNA
copy number appeared a clearly decrease in the FXN knockdown cells
(Fig. 2M). Collectively, these ndings provide a pivotal role of FXN in
maintaining the homeostasis of mitochondrial and keep the balance of
iron metabolism.
3.3. Suppression of FXN enhanced erastin-induced ferroptosis
We wondered whether suppression of FXN accelerated the process
of ferroptosis cell death. Erastin, system X
c
cystine/glutamate anti-
porter (xCT) inhibitor, which can depress cysteine levels, was applied to
induce ferroptosis in HT-1080 cells. FXN knockdown and control cells
were exposed to erastin treatment for 12 h and suppression of FXN
expression signicantly enhanced erastininduced cell death. The con-
centrations of IC50 values for control were evidently higher than that of
two FXN knockdown cells, which calculated results were 4.130 μM and
5.527 μM, respectively (Fig. 3A). We obtained a similar phenomenon in
U266 and Kasumi-1 cells (Fig. S3 A, B), indicating that FXN sensitized
cells to ferroptosis was not restricted to only a single cell lineage. Of
note, IKE, another cystine/glutamate antiporter inhibitor, also co-
operate with FXN suppression to induce cell death. On the contrary,
suppression of FXN expression did not aect cell death induced by other
ferroptosis inducers, including RSL3 (GPX4 inhibitor) and BSO (GSH
synthase inhibitor) (Fig. S3 C).
Erastin-induced ferroptosis would lead to a disruption of cell
membrane permeability, therefore enabled propidium iodide (PI)
staining to monitor the progress. The PI staining assay manifested that
suppression of FXN expression remarkably induced HT-1080 cell death
following erastin treatment. Phase-contrast microscopy also showed
FXN depleted cells became shrinking followed by condensation of cy-
toplasmic constituents, like a ballooningphenotype, upon erastin
treatment (Fig. 3B). These ndings indicated FXN knockdown sig-
nicantly augmented erastininduced ferroptotic cell death. In addition,
we investigated the proliferation rate of diverse cells exposed to a low
concentration of erastin and carboxyuorescein diacetate succinimidyl
ester (CFDA-SE) uorescence intensity which exponentially decreased
with cell proliferation and division was analyzed by ow cytometry.
The result indicated that erastin obviously attenuated cell proliferation
of HT-1080 cells, which was much more signicant upon FXN depletion
cells (Fig. 3C). Furthermore, Dramatic morphological changes of mi-
tochondria were observed by transmission electron microscopy in FXN
depletion cells comprising mitochondrial fragmentation, vacuolization
and cristae enlargement, which was aggravated under treatment with
erastin (Fig. 3D).
As the accumulation of lipid peroxidation and free iron level were
two critical drivers of ferroptosis. The uorescent staining of BODIPY
C11, a sensitive uorescent reporter for lipid peroxidation, was used to
quantify the formation of lipid peroxidation in live cells by ow cyt-
ometer and confocal laser microscope. Increasing levels of lipid per-
oxides were monitored in indicated HT-1080 cells after erastin treated
for 12 h, and FXN deletion signicantly promoted lipid peroxide for-
mation upon erastin treatment with red uorescence of the BODIPY
shifted to green uorescence. But FXN depletion alone could not sti-
mulate the accumulation of lipid peroxidation (Fig. 3E, H). Quantita-
tion of the confocal uorescence was provided in Fig. S3 D. We also
discovered similar results that FXN accelerated the accumulation of
lipid peroxidation and ROS under the exposure of erastin in U266 cells
(Fig. S3 E, F). We next asked whether alteration of GSH level partici-
pated in the FXN dependent ferroptosis, and found FXN depletion alone
was not enough to reduce GSH level. But under ferroptotic stress, such
as erastin treatment, FXN depletion accelerated the decrease of GSH
(Fig. S3 G). Given that iron enables to produce superoxide radicals via
the Fenton reaction and result in lipid peroxidation. We next assessed
the cellular labile iron pool by RPA probe and found FXN suppression
robustly activated the labile iron level under the erastin exposure (Fig.
S3 H). In consistent with RPA uorescence, we also found that FXN
depletion activated the iron-starvation response, which was far more
robust under the treatment of erastin (Fig. S3 I).
Mitochondrial dysfunction and oxidative stress are the other two
characteristics of ferroptosis. We hypothesized that FXN accelerated
ferroptosis was associated with the increased of MMP and cytoplasm
ROS. Indeed, the elevated levels of MMP and cytoplasm ROS induced by
erastin were signicantly aggravating by the suppression of FXN
(Fig. 3F, G). To further validate that FXN knockdown specically en-
hanced the ferroptotic process, we used dierent signal pathway in-
hibitors to observe the rescue eect. Notably, erastin-induced cell death
was able to reverse by the ferroptosis inhibitor ferrostatin-1 and GSH,
but not by the apoptosis inhibitor ZVAD-FMK and necroptosis inhibitor
necrosulfonamide (Fig. 3I).
Several studies have revealed that autophagy-dependent degrada-
tion of ferritin, a process named ferritinophagy, takes place in ferrop-
tosis. To distinguish whether FXN regulates ferroptosis through mod-
ulating autophagy. The conversion of LC3 was detected in FXN
depletion and control cells in the absence or presence of erastin.
Alterations of FXN expression did not aect the erastin-induced for-
mation of lipidated LC3B (Fig. S3 J). A similar phenomenon was
showed that pharmacological inhibition of autophagy by BafA1 could
not reverse the FXN induced ferroptosis (Fig. 3I), indicating autophagy
was not involved in the FXN dependent ferroptosis. Together, these
studies demonstrate that FXN depletion is capable of accelerating era-
stin induced ferroptosis, most likely due to the breakdown of the ISC
biosynthetic machinery and mitochondrial dysfunction.
3.4. Overexpress of FXN contributed to ferroptosis resistance
To shed more light on the specic order of events in modulating
mitochondrial function and ferroptosis by FXN, ectopic expression of
FNX cell was generated and the proliferation rate was analyzed by the
CFDA-SE probe. The results showed that FXN overexpression obviously
increased cell proliferation of HT-1080 cells (Fig. 4A). In contrast with
mitochondrial dysfunction in FXN suppression cells, ectopic expression
of FXN markedly increased mtDNA copy number and decreased the
liable iron level (Figs. 4B, D, S4A). Next, we asked whether over-
expression of FXN in HT-1080 cells could diminish erastin-induced
ferroptosis. As shown in Fig. 4C, cell viability assays of the indicated
cells experienced that ectopic expression of FXN sensibly abolished
erastin-induced growth inhibition compared to the control and FXN
depletion cells. To further verify the specicity of the observed phe-
nomenon, we generated an shRNA-resistant FXN cDNA (Res-FXN) and
co-transduced with shFXN. Indeed, overexpression of an shRNA re-
sistant form of FXN eciently restored the growth inhibition of erastin
(Figs. S4B and C). Similarly, expression of Res-FXN conferred resistance
to xCT inhibitors, IKE, but not to RSL3 and BSO (Fig. S4D). In consistent
with the results of RPA, mitochondrial and cytoplasmic iron measured
by ICP-MS demonstrated that knockdown of FXN induced the accu-
mulation of iron in both fractions, while ectopic expression of Res-FXN
decreased the level of iron(Figs. S4E and F). Collectively, these ndings
indicate that expression of FXN is directly responsible for the observed
phenotypes and excluding the possibility of o-target eects. More
details were found with the mitochondrial morphological changes
through MitoTracker uorescence staining and transmission electron
microscopy. The results showed that FXN expression could keep mi-
tochondrial morphology integrity, without severe mitochondrial frag-
mentation or vacuolization changes under erastin treatment (Fig. 4E,
F).
J. Du, et al. Redox Biology 32 (2020) 101483
7
As FXN expression robustly decreased cellular free iron contents,
another important question was whether FXN expression could rescue
the erastininduced accumulation of lipid peroxidation. In line with
previous ndings, FXN expression restored the erastinchallenged lipid
peroxidation (Figs. 4G, H, S4G). In addition, ferroptosis inhibitors
(ferrostatin-1 and GSH) and DFO restored much less in the FXN over-
expression cells than knockdown cells (Fig. 4I). Collectively, these re-
sults imply that FXN expression signicantly suppresses erastin-induced
ferroptosis.
3.5. Free iron depletion enhanced the resistance to ferroptosis in the FXN
knockdown cells
The above results suggested that FXN suppression dramatically in-
duced mitochondria dysfunction and activated the iron-starvation re-
sponse, thus accelerating erastin-challenged ferroptosis. Since ferritin
heavy chain (FTH) has ferroxidase activity and oxidizes Fe
2+
to
catalytically inactive Fe
3+
and plays a vital role in maintaining iron
homeostasis by storing iron in a soluble, non-toxic form. We, therefore,
hypothesized that overexpression of FTH might restore dysregulated
iron homeostasis in the FXN suppression HT-1080 cells. To test this
hypothesis, we reconstituted the expression of iron storage protein FTH
in FXN suppression HT-1080 cells and rstly explored the eects on the
cell proliferation and mitochondrial function. As expected, the FTH
overexpressed cells eliminated the proliferation arrest induced by FXN
suppression and accelerated the formation of cell colony, manifesting
that iron homeostasis regulation could modulate the injury derive from
FXN knockdown (Fig. 5A, B). The quantication of free iron level re-
vealed that FTH expression sensibly blocked the iron accumulation
stimulated by FXN suppression (Figs. 5C, D, S5A). Similarity, mito-
tracker staining also showed a signicantly increased network of tu-
bule-shaped morphology, which characteristic for healthy and func-
tional mitochondria under the recombinant expression of FTH in FXN
suppression cells (Fig. 5E). Additionally, we evaluated the
Fig. 3. Suppression of FXN accelerated erastin in-
duced ferroptosis. (A) HT-1080 cells were exposed
to various concentrations of erastin for 12 h re-
spectively and followed by CCK8 assay. (B)
Propidium iodide positive cells were stained and
observed by uorescent microscopy after erastin
exposure for 12 h. Scale bars: 30 μm. (C) CFDA-SE
probe (5 μM) stained HT-1080 cells were exposed to
2.5 μM erastin and cultured for 3 days, then sub-
jected to ow cytometry. (D) The morphological
changes of mitochondria were detected by trans-
mission electron microscopy (TEM) in the absence
or presence of erastin. Flow cytometry was per-
formed to measure lipid peroxides (E), cytoplasm
ROS (F), mitochondrial membrane potential(G)
after erastin treatment for 12 h in HT-1080 cells. (H)
Representative images of BODIPY staining in erastin
treated HT-1080 cells which observed by confocal
laser microscope. Scale bars: 75 μm. (I) HT-
1080 cells were treated with erastin (10 μM) with or
without small molecule inhibitor for 12 h and re-
spective cell viability was detected by CCK8. (fer-
roptosis inhibitor ferrostatin-1,0.5 μM; GSH, 1 mM;
apoptosis inhibitor Z-VAD-FMK, 4 μM; necroptosis
inhibitor Necrosulfonamide, 0.5 μM; autophagy in-
hibitor BafA1, 20 nM ). All histograms were re-
presented as mean ± SD.
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
8
mitochondrial DNA copy and found that FTH recombinant expression
could reverse the decrease of mitochondrial DNA copy (Fig. 5F). In
summary, these data emphasize the fact that FTH expression restores
mitochondrial impairment induced by FXN suppression.
Based on the observed results, we next studied whether FTH ex-
pression could rescue the ferroptosis induced by erastin in FXN sup-
pression cells. And the results showed suppression of FXN increased
erastin-induced cell death, while reconstituted expression of FTH sig-
nicantly restored the ferroptosis cell death (Fig. 6A). CCK8 assay was
used to evaluate the potency of FTH to restored erastin-induced cell
death and results showed FTH fully restored the erastin-induced phe-
notypes (Fig. 6B). Cells transduced with shFXN displayed noticeable
altered mitochondrial shape and size, including mitochondrial frag-
mentation, vacuolization and loss of cristae enlargement, whereas these
phenotypes were reversed by FTH overexpression through reducing
cellular iron even with the treatment with erastin (Fig. 6C).
In addition, overexpression of FTH was able to rescue the accu-
mulation of lipid peroxidation and cytoplasm ROS in the FXN knock-
down cells upon erastin exposure (Fig. 6D, E). Consistent with these
results, pharmacologically chelate free iron by DFO also prevented cell
death of FXN suppression cells to a similar extent as the control under
the treatment of erastin (Fig. 4I). In summary, our results emphasize the
decisive role of FXN in the regulation of cysteine deprivation-induced
ferroptosis through modulating iron homeostasis, which alleviating by
the iron depleted.
3.6. Knockdown of FXN inhibited the growth of xenograft in vivo
To evaluate the eect of FXN knockdown on proliferation in vivo,
FXN suppression HT-1080 cells with or without FTH recombinant ex-
pression were subcutaneously injected into nude mice. The growth
condition of xenograft tumors was traced, which manifested that FXN
silencing indeed blocked the proliferation of tumor in vivo, while
overexpression of FXN or FTH accelerated the tumor growth (Fig. 7A).
Further, H&E and IHC staining were performed and results showed that
there were large necrosis areas in FXN silenced tumor tissues, accom-
panied by nuclear condensation, fragmentation and a signicantly de-
creased Ki-67 expression (Fig. 7B). Inversely FXN overexpression not
only maintained the tumor morphology but also promoted the expres-
sion of Ki-67. FTH expression was also detected by IHC staining and
Fig. 4. Enforced expression of FXN contributed to
ferroptosis resistance. (A) The proliferation rate was
analyzed by the CFDA-SE probe between the in-
dicated cells. (B) Quantitative PCR analysis was
performed to measure mtDNA copy number.
Primers specic for the HGB1 gene were used for
the determination of nuclear DNA (nDNA) and pri-
mers specic for ND-1 were used to detect mtDNA.
(C) Indicated HT-1080 cells were incubated with
dierent concentrations of erastin for 12 h and cell
viability was assayed by CCK8 assay. (D)
Intracellular Fe
2+
was measured by the staining of
RPA and photographed by the confocal microscope.
Scale bars: 75 μm. (E) MitoTracker Red labeled HT-
1080 cells were subjected to the confocal micro-
scope for observing the changes of mitochondrial
morphology. Scale bars: 75 μm. (F) The morpholo-
gical changes of mitochondria were detected by
transmission electron microscopy (TEM) in the ab-
sence or presence of erastin. Lower scale bars:
0.2 μm. (G-H) To assess lipid ROS production, HT-
1080 cells were treated with 5 μM erastin and
loaded with BODIPY C11 probe for 30 min followed
by ow cytometry measurement (G) and confocal
laser microscope (H). Scale bars: 75 μm. (I)
Indicated HT-1080 cells were treated with erastin
(10 μM) with or without small molecule inhibitor
and respective cell viability was detected by CCK8.
(ferrostatin-1,0.5 μM; GSH, 1 mM; iron chelator
DFO, 100 μM; Z-VAD-FMK, 4 μM;
Necrosulfonamide, 0.5 μM). All histograms were
represented as mean ± SD. (For interpretation of
the references to colour in this gure legend, the
reader is referred to the Web version of this article.)
P < 0.05,
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
9
consistent with the data in vitro that FXN knockdown robustly activated
iron starving stress and decreased the FTH expression, which sig-
nicantly restored by FXN overexpression. In addition, FTH expression
could also reverse the phenomenons exhibited in FXN knockdown xe-
nograft, including inhibited tumors growth and decreased Ki-67 ex-
pression. These observations lead to the hypothesis that FXN or the
downstream iron homeostasis associated protein can be developed as a
therapeutic target, leaving cancer cells at increased risk of ferroptotic
cell death.
4. Discussion
Iron is an essential metallic element for all eukaryotes with im-
portant functions in the synthesis of iron-sulfur cluster (FeS), heme
and other cofactors [2527]. In mammalian cells, Iron homeostasis is
tightly regulated by iron regulatory proteins 1 (IRP1) and 2(IRP2)
through post-transcriptionally controlling iron metabolism genes via
the IRE-IRP mechanism. As shown in several studies, iron excess is
closely related with tumor genesis in some types of human cancers [28].
There is a phenomenon termed iron addiction, which is to say, cancer
cells do exhibit an enhanced dependence on iron relatively to healthy
cells to enable its growth [3,29]. Supersaturation ferric ion or iron
exposure may increase cancer risk. Like two sides of a coin, excess iron
loading may also lead to toxicity due to the ability of Fe participating in
the production of ROS via Fenton reactions, which may be more pro-
nounced in the highly redox-active mitochondrion. Therefore, the high
level of iron may act as a target for cancer therapy [8].
Ferroptosis is a recently identied form of regulated cell death that
is distinct from the other types of cell death at morphological, bio-
chemical, and genetic levels and eciently recovered by ferrostatin-1,
GSH and DFO, et, al [4]. Recent study demonstrated that cancer cells
depended on high levels of the ISC biosynthetic enzyme NFS1, which
was indispensable for FeS clusters present in multiple cell essential
proteins upon exposure to oxidative damage. Suppression of NFS ro-
bustly activates the accumulation of iron and triggers ferroptosis co-
operates with cystine/glutamate antiporter inhibitor [30]. Our present
Fig. 5. Overexpress of FTH eliminated the pro-
liferation arrest and mitochondria dysfunction in-
duced by FXN suppression. (A-B) The ability of cell
proliferation was measured by CCK8 assay (A) and
plate clone formation assay(B) between the three
indicated cells. (C-D) To assess intracellular Fe
2+
,
cells were stained with RPA probe for 30 min and
photographed by a confocal laser microscope(C) or
subjected to ow cytometry (D). (E) Cells were
stained by MitoTracker Red and mitochondrial
morphology changes were observed under the con-
focal microscope. Scale bars: 75 μm. (F) The quan-
titative PCR analysis was performed to measure
mtDNA copy number. (For interpretation of the re-
ferences to colour in this gure legend, the reader is
referred to the Web version of this article.)
P < 0.05,
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
10
study demonstrated that FXN was a key regulator of ferroptosis by
modulating the iron homeostasis and mitochondrial function. Genetic
inhibition of FXN signicantly repressed the proliferation, destroyed
mitochondrial morphology, impeded FeS cluster assembly and ex-
acerbated iron accumulation. Erastin further induced dramatic mi-
tochondria morphological damage in FXN suppressed cells such as en-
hanced fragmentation and vanished cristae, a phenotype as the
hallmarks of ferroptosis, and was consistent with early research. Vice
versa, enforced expression of FXN blocked the iron starvation response,
MMP hyperpolarization, mitochondrial fragmentation and erastin-in-
duced ferroptosis, conrming that the function of FXN shared benecial
mechanisms for cell survival which acted at upstream of ferroptosis. In
particular, both pharmacological and genetic inhibition of iron starva-
tion stress also wholly restored the resistance to ferroptosis in FXN
knockdown cells. These results supporte a mechanistic hypothesis that
FXN deciency accelerate erastin activated ferroptotic cell death.
FXN is a highly conserved protein localizes in the mitochondrial
matrix and participates in the biosynthesis of FeS cluster. A recent
study shows that FXN can activation NFS1 and accelerating a rate-
limiting sulfur transfer step of FeS cluster assembly by inducing an
unusual rearrangement of protein subunits in the de novo assembly
complex [15]. In line with the previous nding, suppression of NFS1
make cancer cell sensitive to ferroptosis in vitro and slow tumor growth
[30]. The main mechanism of FXN medicated ferroptosis may be the
breakdown of the ISC biosynthetic machinery. Mutation of human FXN
causes FRDA, a devastating, multi-systemic inherited degenerative
syndrome that aects 1 in 50,000 people. Although there is clear evi-
dence demonstrating that FRDA is associated with mitochondrial
dysfunction, mitochondrial iron accumulation, and increased oxidative
stress [31], the exact mechanism remains uncertain. Kevin Kemp [32]
and Irazusta V et al. [33] demonstrated that cells derived from FRDA
patients had deciencies in defenses against oxidative stress and de-
creased in superoxide dismutase (SOD) activity. Therefore, Hongting
Zhao et al. [34] discovered that mitochondrion-targeted peptide SS-31,
a novel mitochondrion-targeted antioxidant, improved the function of
mitochondria in FRDA patient-derived cells and might potentially be a
new drug for the early treatment of FRDA. Furthermore, Amy Anzovino
et al. [23] revealed signicant alterations in the cellular redox home-
ostasis were mediated by Nrf2 deciency through increased cytosolic
Keap1 levels in FRDA mouse model. Similar to earlier studies, Tslil Ast
et al. [35] also veried that FXN depletion resulted in the activation of
ATF4-dependent integrated stress response and loss of anti-oxidant
NRF2 signaling, which was compromised under hypoxia. NRF2 is an
important transcription factor in regulating cellular redox homeostasis
through binding antioxidant response elements (AREs). It has been
reported NRF2 plays a critical role in mitigating lipid peroxidation
[36,37] and its inhibition reverses the resistance to ferroptosis [6,38].
We also identied the downregulation of NRF2 in FXN suppression
cells, which might be another factor giving rise to the cells sensitive to
ferroptosis. Despite the central role of FXN in oxidative metabolism, it
remains unclear whether it plays a central role in cancer progression. In
our work, we examined the expression of FXN based on The Cancer
Genome Atlas (TCGA) datasets and conrmed that the expression of
FXN was much higher in various cancer tissue. And high FXN expres-
sion level indicated poor survival of Sarcoma, ACC, BLCA, LAML pa-
tients. Overall, the present study exposes FXN as the molecular link
Fig. 6. FTH expression restored the resistance to
ferroptosis in FXN knockdown cells. (A) Indicated
cells were treated with erastin (010 μM) for 12 h
and cell viability was assayed by CCK8. (B) HT-
1080 cells were treated with erastin (10 μM) for
12 h with or without small molecule inhibitor and
respective cell viability was detected by CCK8. (C)
The morphological changes of mitochondria were
detected by transmission electron microscopy
(TEM) in the absence or presence of erastin. Lower
scale bars: 0.2 μm. (D) Cells were treated with 5 μM
erastin and loaded with DCFH-DA (4 μM) probe for
30 min followed by ow cytometry to assess cyto-
plasm ROS formation. (E) Indicated HT-1080 cells
were treated with erastin (5 μM) for 12 h. Levels of
lipid ROS production were detected with BODIPY
C11 (2.5 μM) probe by ow cytometry measure-
ment.
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
11
between the oxidative stress signaling and ferroptosis.
It is currently under debate whether mitochondria involved in fer-
roptosis. Dixon and his co-workers found that mtDNA depleted cancer
cell lines did not show a signicant dierence in ferroptosis sensitivity
[39]. Gaschler conrmed that ferrostatin analogs executed the anti-
death potency without located in mitochondria [40]. These observa-
tions provided evidence arguing against the potential involvement of
mitochondria in ferroptosis. By contrast, we evaluated mitochondria
from both its morphology and function. Abnormal mitochondrial
morphology with decreased or vanished mitochondria cristae was de-
tected in the FXN knockdown cells. The subsequent copy number of
mtDNA, encodes 13 polypeptides which play crucial roles in the OX-
PHOS, was signicantly decreased following the knockdown of FXN. As
published before, ISCs undergo spontaneous degradation upon ex-
posure to oxygen and other oxidative stress conditions [41]. Upon FXN
suppression, breakdown of the ISC biosynthetic machinery cannot meet
the increased demand, ultimately leading to the reduced steady-state
levels of FeS cluster dependent proteins such as the FeS cluster
binding subunits of mitochondrial complex and aconitase. All the above
data were consistent with the nding FXN deciency led to dramati-
cally decline of mitochondrial oxygen consumption rate, thus resulting
in the insucient generation of ATP. The detailed investigation of
mitochondrial dysfunction provide novel insights into the role of mi-
tochondrial damage in ferroptosis.
It's interesting that FXN suppression stimulates the hyperpolariza-
tion of MMP, followed by the eventual collapse leading to cell death,
Fig. 7. Knockdown of FXN inhibited the growth of xenograft in vivo. (A) The tumor volumes were measured every 2 days and signicant inhibition of tumor growth
was observed after FXN knockdown. Representative photographs showed the tumor size in mice after the mice were sacriced. (B) H&E and IHC analysis for Ki67,
FXN and FTH in indicated tumor specimens. Scale bars: 50 μm. (C-E) The Specimens were scored by relative integrated optical density (IOD) value.
P < 0.05,
★★
P < 0.01 versus control.
J. Du, et al. Redox Biology 32 (2020) 101483
12
which is in accordance with previous study [35]. It has been reported
that excess hyperpolarization of MMP did not accelerate ATP produc-
tion, but help to the production of free radicals [22]. In addition, upon
pharmacological inhibition of GPX4 or GSH synthase, the downstream
components of the ferroptosis signal pathway, ferroptosis can still be
triggered independently of FXN depletion. It is likely that iron-starva-
tion stress and mitochondrial dysfunction, associated with FXN de-
ciency, act at the upstream of ferroptosis, making FXN dispensable for
GPX4 inhibition induced ferroptosis. This is in general agreement with
earlier reports published by Yang et al., which revealed that mi-
tochondria was involved in the cysteine deprivation-induced ferroptosis
but not in glutathione peroxidase-4 (GPX4) inhibition-induced ferrop-
tosis [42]. Collectively, the oxidative stress caused by mitochondrial
dysregulation in FXN suppressed cells together with the defective uti-
lization of mitochondrial iron enhancing the erastin-induced ferrop-
tosis, which might potentially be a new drug target.
Declaration of competing interest
The authors declare that there is no conict of interest.
Acknowledgments
This research was supported by National Science and Technology
Major Project for New Drug (No. 2017ZX301033), National Natural
Science Foundation of China (81971172), The Key Research and
Development Program of Zhejiang Province (WKJ-ZJ-1914), Zhejiang
Public Welfare Technology Application Research Project (Grant No.
LGF19H080006, LGF20H080005, LY18C090004, 2017C33091),
Medical and Health Science and Technology Project of Zhejiang
Province (No. 2019RC014, 2019RC115, 2018KY003, 2017KY006,
2017KY209), Zhejiang Students' Science and Technology Innovation
Activity Plan (No. 2019R410053).
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://
doi.org/10.1016/j.redox.2020.101483.
List of abbreviations
FXN frataxin
shRNA short hairpin small interfering RNA
PUFAs polyunsaturated fatty acids
FRDA Friedreich's ataxia
ISC Iron-sulfur cluster
ROS reactive oxygen species
DFO deferoxamine
GSH glutathione
TCGA The Cancer Genome Atlas
MMP mitochondrial membrane potential
OCR oxygen consumption rate
IRP1 iron responsive protein 1
FTH Ferritin heavy chain
TFR1 transferrin receptor 1
CFDA-SE carboxyuorescein diacetate succinimidyl ester
Fer-1 ferrostatin-1
AREs antioxidant response elements
GPX4 glutathione peroxidase-4
PI propidium iodide
SOD superoxide dismutase
References
[1] S. Puig, L. Ramos-Alonso, A.M. Romero, M.T. Martinez-Pastor, The elemental role
of iron in DNA synthesis and repair, Metallomics 9 (2017) 14831500.
[2] K. Furuyama, K. Kaneko, Iron metabolism in erythroid cells and patients with
congenital sideroblastic anemia, Int. J. Hematol. 107 (2018) 4454.
[3] D.H. Manz, N.L. Blanchette, B.T. Paul, F.M. Torti, S.V. Torti, Iron and cancer: recent
insights, Ann. N. Y. Acad. Sci. 1368 (2016) 149161.
[4] Scott J. Dixon, Kathryn M. Lemberg, Michael R. Lamprecht, R. Skouta, Eleina
M. Zaitsev, Caroline E. Gleason, Darpan N. Patel, Andras J. Bauer, Alexandra
M. Cantley, Wan S. Yang, et al., Ferroptosis: an iron-dependent form of non-
apoptotic cell death, Cell 149 (2012) 10601072.
[5] P. A, I. Fl, M. Jm, C. Pj, Regulated necrosis in kidney ischemia-reperfusion injury,
Kidney Int. 96 (2019) 291301.
[6] D. Shin, E.H. Kim, J. Lee, J.L. Roh, Nrf2 inhibition reverses resistance to GPX4
inhibitor-induced ferroptosis in head and neck cancer, Free Radical Biol. Med. 129
(2018) 454462.
[7] Y. Mou, J. Wang, J. Wu, D. He, C. Zhang, C. Duan, B. Li, Ferroptosis, a new form of
cell death: opportunities and challenges in cancer, J. Hematol. Oncol. (2019),
https://doi.org/10.1186/s13045-019-0720-y.
[8] B. Hassannia, P. Vandenabeele, T. Vanden Berghe, Targeting ferroptosis to iron out
cancer, Canc. Cell 35 (2019) 830849.
[9] T. Hirschhorn, B.R. Stockwell, The development of the concept of ferroptosis, Free
Radical Biol. Med. (2018), https://doi.org/10.1016/j.freeradbiomed.2018.09.043.
[10] M. Gao, P. Monian, Q. Pan, W. Zhang, J. Xiang, X. Jiang, Ferroptosis is an autop-
hagic cell death process, Cell Res. 26 (2016) 10211032.
[11] W. Hou, Y. Xie, X. Song, X. Sun, M.T. Lotze, H.J. Zeh 3rd, R. Kang, D. Tang,
Autophagy promotes ferroptosis by degradation of ferritin, Autophagy 12 (2016)
14251428.
[12] J. Du, T. Wang, Y. Li, Y. Zhou, X. Wang, X. Yu, X. Ren, Y. An, Y. Wu, W. Sun, DHA
inhibits proliferation and induces ferroptosis of leukemia cells through autophagy
dependent degradation of ferritin, Free Radical Biol. Med. (2018), https://doi.org/
10.1016/j.freeradbiomed.2018.12.011.
[13] Y. Yu, Y. Xie, L. Cao, L. Yang, M. Yang, M.T. Lotze, H.J. Zeh, R. Kang, D. Tang, The
ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to
chemotherapeutic agents, Mol. Cell. Oncol. 2 (2015) e1054549.
[14] C. Jc, L.B. Ne, Redox-sensing iron-sulfur cluster regulators, Antioxidants Redox
Signal. 29 (2018) 18091829.
[15] S. Patra, D.P. Barondeau, Mechanism of activation of the human cysteine de-
sulfurase complex by frataxin, Proc. Natl. Acad. Sci. U.S.A. 116 (2019)
1942119430.
[16] S. Pp, V. V, B. K, D.S. P, Iron-sulfur protein assembly in human cells, Rev. Physiol.
Biochem. Pharmacol. 174 (2018) 2565.
[17] C. M, P. H, G. A, K. H, D. A, L. M, F. K, D. P, K. M, Inactivation of the Friedreich
ataxia mouse gene leads to early embryonic lethality without iron accumulation,
Hum. Mol. Genet. 9 (2000) 12191226.
[18] F. Lupoli, T. Vannocci, G. Longo, N. Niccolai, A. Pastore, The role of oxidative stress
in Friedreich's ataxia, FEBS Lett. 592 (2018) 718727.
[19] M.G. Cotticelli, S. Xia, D. Lin, T. Lee, L. Terrab, P. Wipf, D. Huryn, R. Wilson,
Ferroptosis as a novel therapeutic target for Friedreich's ataxia, J. Pharmacol. Exp.
Therapeut. (2019), https://doi.org/10.1124/jpet.118.252759.
[20] U. Mühlenho, J.A. Stadler, N. Richhardt, A. Seubert, T. Eickhorst, R.J. Schweyen,
R. Lill, G. Wiesenberger, A specic role of the yeast mitochondrial carriers Mrs3/4p
in mitochondrial iron acquisition under iron-limiting conditions, J. Biol. Chem. 278
(2003) 4061240620.
[21] Z. L, B. U, R. Jl, An ecient one-step site-directed and site-saturation mutagenesis
protocol, Nucleic Acids Res. 32 (2004), https://doi.org/10.1093/nar/gnh110.
[22] M. Hüttemann, I. Lee, A. Pecinova, P. Pecina, K. Przyklenk, J.W. Doan, Regulation
of oxidative phosphorylation, the mitochondrial membrane potential, and their role
in human disease, J. Bioenerg. Biomembr. 40 (2008) 445456.
[23] A. Anzovino, S. Chiang, B.E. Brown, C.L. Hawkins, D.R. Richardson, M.L. Huang,
Molecular alterations in a mouse cardiac model of Friedreich ataxia: an impaired
Nrf2 response mediated via upregulation of Keap1 and activation of the Gsk3beta
Axis, Am. J. Pathol. 187 (2017) 28582875.
[24] Y. Shan, R.A. Schoenfeld, G. Hayashi, E. Napoli, T. Akiyama, M. Iodi Carstens,
E.E. Carstens, M.A. Pook, G.A. Cortopassi, Frataxin deciency leads to defects in
expression of antioxidants and Nrf2 expression in Dorsal Root Ganglia of the
Friedreich's Ataxia YG8R mouse model, Antioxidants Redox Signal. 19 (2013)
14811493.
[25] S. Toyokuni, F. Ito, K. Yamashita, Y. Okazaki, S. Akatsuka, Iron and thiol redox
signaling in cancer: an exquisite balance to escape ferroptosis, Free Radical Biol.
Med. 108 (2017) 610626.
[26] H. Harigae, K. Hino, S. Toyokuni, Iron as soul of life on earth revisited: from che-
mical reaction, ferroptosis to therapeutics, Free Radical Biol. Med. 133 (2019) 12.
[27] D.A. Stoyanovsky, Y.Y. Tyurina, I. Shrivastava, I. Bahar, V.A. Tyurin,
O. Protchenko, S. Jadhav, S.B. Bolevich, A.V. Kozlov, Y.A. Vladimirov, et al., Iron
catalysis of lipid peroxidation in ferroptosis: regulated enzymatic or random free
radical reaction? Free Radical Biol. Med. 133 (2019) 153161.
[28] S.V. Torti, F.M. Torti, Iron and cancer: more ore to be mined, Nat. Rev. Canc. 13
(2013) 342, https://doi.org/10.1038/nrc3495.
[29] C. Raggi, E. Gammella, M. Correnti, P. Buratti, E. Forti, J.B. Andersen, G. Alpini,
S. Glaser, D. Alvaro, P. Invernizzi, Dysregulation of iron metabolism in cholangio-
carcinoma stem-like cells, Sci. Rep. 7 (2017) 17667.
[30] S.W. Alvarez, V.O. Sviderskiy, E.M. Terzi, T. Papagiannakopoulos, A.L. Moreira,
S. Adams, D.M. Sabatini, K. Birsoy, R. Possemato, NFS1 undergoes positive selection
in lung tumours and protects cells from ferroptosis, Nature (2017), https://doi.org/
10.1038/nature24637.
[31] R. Abeti, M.H. Parkinson, I.P. Hargreaves, P.R. Angelova, C. Sandi, M.A. Pook,
P. Giunti, A.Y. Abramov, Mitochondrial energy imbalance and lipid peroxidation
cause cell death in Friedreich's ataxia, Cell Death Dis. 7 (2016) e2237, https://doi.
J. Du, et al. Redox Biology 32 (2020) 101483
13
org/10.1038/cddis.2016.111.
[32] K. Kemp, R. Dey, A. Cook, N. Scolding, A. Wilkins, Mesenchymal stem cell-derived
factors restore function to human frataxin-decient cells, Cerebellum 16 (2017)
840851.
[33] I. V, O. E, M.-C. A, C. E, R. J, T. J, Yeast frataxin mutants display decreased su-
peroxide dismutase activity crucial to promote protein oxidative damage, Free
Radical Biol. Med. 48 (2010) 411420.
[34] H. Zhao, H. Li, S. Hao, J. Chen, J. Wu, C. Song, M. Zhang, T. Qiao, K. Li, Peptide SS-
31 upregulates frataxin expression and improves the quality of mitochondria: im-
plications in the treatment of Friedreich ataxia, Sci. Rep. 7 (2017) 9840, https://
doi.org/10.1038/s41598-017-10320-2.
[35] T. Ast, J.D. Meisel, S. Patra, H. Wang, R.M.H. Grange, S.H. Kim, S.E. Calvo,
L.L. Orece, F. Nagashima, F. Ichinose, et al., Hypoxia rescues frataxin loss by re-
storing iron sulfur cluster biogenesis, Cell 177 (2019) 15071521.
[36] M. Dodson, R. Castro-Portuguez, D.D. Zhang, NRF2 plays a critical role in miti-
gating lipid peroxidation and ferroptosis, Redox Biol. (2019), https://doi.org/10.
1016/j.redox.2019.101107.
[37] X. Sun, Z. Ou, R. Chen, X. Niu, D. Chen, R. Kang, D. Tang, Activation of the p62-
Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells,
Hepatology 63 (2016) 173184.
[38] J.L. Roh, E.H. Kim, H. Jang, D. Shin, Nrf2 inhibition reverses the resistance of
cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis,
Redox Biol. 11 (2017) 254262.
[39] D. Sj, L. Km, L. Mr, S. R, Z. Em, G. Ce, P. Dn, B. Aj, C. Am, Y. Ws, et al., Ferroptosis:
an iron-dependent form of nonapoptotic cell death, Cell 149 (2012) 10601072.
[40] G. Mm, H. F, F. H, L. A, M. W, S. Br, Determination of the subcellular localization
and mechanism of action of ferrostatins in suppressing ferroptosis, ACS Chem. Biol.
13 (2018) 10131020.
[41] D.R. Crooks, N. Maio, A.N. Lane, M. Jarnik, R.M. Higashi, R.G. Haller, Y. Yang,
T.W. Fan, W.M. Linehan, T.A. Rouault, Acute loss of iron-sulfur clusters results in
metabolic reprogramming and generation of lipid droplets in mammalian cells, J.
Biol. Chem. 293 (2018) 82978311.
[42] M. Gao, J. Yi, J. Zhu, A.M. Minikes, P. Monian, C.B. Thompson, X. Jiang, Role of
mitochondria in ferroptosis, Mol. Cell. 73 (2019) 354363.
J. Du, et al. Redox Biology 32 (2020) 101483
14
... Here, the electroporation protocols stimulated a synergistic interaction between the iron compounds and 5-FU. In each case, the compounds did not decrease the progression of frataxin expression over time, meaning that no ferroptosis induction occurred [31]. ...
Article
Full-text available
These authors contributed equally to this work. Abstract: Colon cancer (CC) management includes surgery, radio-and chemotherapy based on treatment with 5-fluorouracil (5-FU) or its derivatives. However, its application is limited to low-grade carcinomas. Thus, much research has been conducted to introduce new techniques and drugs to the therapy. CC mostly affects older people suffering from cardiac diseases, where iron compounds are commonly used. Ferric citrate and iron (III)-EDTA complexes have proven to be effective in colon cancer in vitro. This study aimed to determine the potency and action of iron-containing compounds in colon cancer treatment by chemo-and electrochemotherapy in both nano-and microsecond protocols. The viability of the cells was assessed after standalone iron (III) citrate and iron (III)-EDTA incubation. Both compounds were also assessed with 5-FU to determine the combination index. Additionally, frataxin expression was taken as the quantitative response to the exposition of iron compounds. Each of the substances exhibited a cytotoxic effect on the LoVo cell line. Electroporation with standalone drugs revealed the potency of 5-FU and iron(III)-EDTA in CC treatment. The combination of 5-FU with iron(III)-EDTA acted synergistically, increasing the viability of the cells in the nanosecond electrochemotherapy protocol. Iron(III)-EDTA decreased the frataxin expression, thus inducing ferroptosis. Iron(III) citrate induced the progression of cancer; therefore, it should not be considered as a potential therapeutic option. The relatively low stability of iron(III) citrate leads to the delivery of citrate anions to cancer cells, which could increase the Krebs cycle rate and promote progression.
... As intracellular iron levels are closely controlled, the available iron pool can also be used for the synthesis of iron-sulfur clusters that usually function as cofactors in numerous mitochondrial processes [30] and also in non-redox reactions [31]. Interestingly, defective synthesis of iron-sulfur clusters imposed by nitrogen fixation 1 (NFS1) inhibition/deletion or frataxin (FXN) suppression can activate an iron starvation response and sensitize cells to ferroptosis [32,33]. Importantly, NFS1 expression is selected in lung cancer to protect against ferroptosis in highly oxygenated tissues [34]. ...
Article
Full-text available
Ferroptosis is a newly identified iron‐dependent type of regulated cell death that can also be regarded as death caused by the specific collapse of the lipid antioxidant defence machinery. Ferroptosis has gained increasing attention as a potential therapeutic strategy for therapy‐resistant cancer types. However, many ferroptosis‐inducing small molecules do not reach the pharmacokinetic requirements for their effective clinical use yet. Nevertheless, their clinical optimization is under development. In this review, we summarize the current understanding of molecular pathways regulating ferroptosis, how cells protect themselves from the induction of ferroptotic cell death, and how a better understanding of cancer cell metabolism can represent vulnerabilities for ferroptosis‐based therapies. Lastly, we discuss the context‐dependent effect of ferroptosis on various cell types within the tumor microenvironment and address controversies on how tissue ferroptosis might impact systemic cancer immunity in a paracrine manner.
Article
Full-text available
Molecular hybridization is a widely employed technique in medicinal chemistry for drug modification, aiming to enhance pharmacological activity and minimize side effects. The combination of an indole ring and imidazole[2,1-b]thiazole has shown promising potential as a group that exhibits potent anti-inflammatory effects. In this study, we designed and synthesized a series of derivatives comprising indole-2-formamide benzimidazole[2,1-b]thiazole to evaluate their impact on LPS-induced production of pro-inflammatory cytokines NO, IL-6, and TNF-α release, as well as iron death in RAW264.7 cells. The findings revealed that most compounds effectively inhibited LPS-induced production of pro-inflammatory cytokines NO, IL-6, and TNF-α release in RAW264.7 cells. Compound 13b exhibited the most potent anti-inflammatory activity among the tested compounds. The results of the cytotoxicity assay indicated that compound 13b was nontoxic. Additionally, compound 13b was found to elevate the levels of ROS, MDA, and Fe²⁺, while reducing GSH content, thereby facilitating the iron death process. Consequently, compound 13b showed promise for future development as an anti-inflammatory drug.
Article
Full-text available
Bladder cancer (BC) is one of the 3 common malignant tumors in the urinary system, with high incidence, easy metastasis, poor therapeutic efficacy, and poor prognosis, which seriously threatens the health of human. Tumor cells exhibit a strong demand for iron, and iron overload can induce ferroptosis, which is an iron dependent cell death caused by lipid peroxidation and cell membrane damage. Therefore, ferroptosis has strong anti-tumor potential. The molecular mechanisms of ferroptosis is associated with abnormalities in cellular phospholipid metabolism and iron metabolism, and dysregulation of antioxidant and non-antioxidant systems Xc-/glutathione (GSH)/glutathione peroxidase 4 (GPX4). Ferroptosis relevant molecules play important roles in the occurrence and development, metastasis, drug resistance, and immune response of BC, and are expected to become targets for the treatment of BC.
Article
Full-text available
BACKGROUND Gastric cancer (GC) is a common malignancy of the digestive system. According to global 2018 cancer data, GC has the fifth-highest incidence and the third-highest fatality rate among malignant tumors. More than 60% of GC are linked to infection with Helicobacter pylori (H. pylori) , a gram-negative, active, microaerophilic, and helical bacterium. This parasite induces GC by producing toxic factors, such as cytotoxin-related gene A, vacuolar cytotoxin A, and outer membrane proteins. Ferroptosis, or iron-dependent programmed cell death, has been linked to GC, although there has been little research on the link between H. pylori infection-related GC and ferroptosis. AIM To identify coregulated differentially expressed genes among ferroptosis-related genes (FRGs) in GC patients and develop a ferroptosis-related prognostic model with discrimination ability. METHODS Gene expression profiles of GC patients and those with H. pylori -associated GC were obtained from The Cancer Genome Atlas and Gene Expression Omnibus (GEO) databases. The FRGs were acquired from the FerrDb database. A ferroptosis-related gene prognostic index (FRGPI) was created using least absolute shrinkage and selection operator–Cox regression. The predictive ability of the FRGPI was validated in the GEO cohort. Finally, we verified the expression of the hub genes and the activity of the ferroptosis inducer FIN56 in GC cell lines and tissues. RESULTS Four hub genes were identified (NOX4, MTCH1, GABARAPL2, and SLC2A3 ) and shown to accurately predict GC and H. pylori -associated GC. The FRGPI based on the hub genes could independently predict GC patient survival; GC patients in the high-risk group had considerably worse overall survival than did those in the low-risk group. The FRGPI was a significant predictor of GC prognosis and was strongly correlated with disease progression. Moreover, the gene expression levels of common immune checkpoint proteins dramatically increased in the high-risk subgroup of the FRGPI cohort. The hub genes were also confirmed to be highly overexpressed in GC cell lines and tissues and were found to be primarily localized at the cell membrane. The ferroptosis inducer FIN56 inhibited GC cell proliferation in a dose-dependent manner. CONCLUSION In this study, we developed a predictive model based on four FRGs that can accurately predict the prognosis of GC patients and the efficacy of immunotherapy in this population.
Article
Full-text available
Significance In humans, essential iron-sulfur (Fe-S) cluster cofactors are synthesized using an assembly complex that depends on the protein frataxin (FXN). The physiological function of FXN, which is linked to the fatal neurodegenerative disease Friedreich’s ataxia (FRDA), is still under debate. Here, we show that FXN accelerates Fe-S cluster formation by positioning a mobile loop cysteine of the assembly complex to interact with the substrate and function as an acid, nucleophile, and sulfur carrier during the reaction. Our architectural switch model suggests that FXN controls Fe-S cluster biosynthesis by inducing an unusual rearrangement of protein subunits in the assembly complex. These results provide mechanistic insights into this critical biological process and establish a foundation for the design of new FRDA treatments.
Article
Full-text available
Ferroptosis is a novel type of cell death with distinct properties and recognizing functions involved in physical conditions or various diseases including cancers. The fast-growing studies of ferroptosis in cancer have boosted a perspective for its usage in cancer therapeutics. Here, we review the current findings of ferroptosis regulation and especially focus on the function of ncRNAs in mediating the process of cell ferroptotic death and on how ferroptosis was in relation to other regulated cell deaths. Aberrant ferroptosis in diverse cancer types and tissues were summarized, and we elaborated recent data about the novel actors of some “conventional” drugs or natural compounds as ferroptosis inducers in cancer. Finally, we deliberate future orientation for ferroptosis in cancer cells and current unsettled issues, which may forward the speed of clinical use of ferroptosis induction in cancer treatment.
Article
Full-text available
The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is a key regulator of the cellular antioxidant response, controlling the expression of genes that counteract oxidative and electrophilic stresses. Many pathological conditions are linked to imbalances in redox homeostasis, illustrating the important role of antioxidant defense systems in preventing the pathogenic effects associated with the accumulation of reactive species. In particular, it is becoming increasingly apparent that the accumulation of lipid peroxides has an important role in driving the pathogenesis of multiple disease states. A key example of this is the recent discovery of a novel form of cell death termed ferroptosis. Ferroptosis is an iron-dependent, lipid peroxidation-driven cell death cascade that has become a key target in the development of anti-cancer therapies, as well as the prevention of neurodegenerative and cardiovascular diseases. In this review, we will provide a brief overview of lipid peroxidation, as well as key components involved in the ferroptotic cascade. We will also highlight the role of the NRF2 signaling pathway in mediating lipid peroxidation and ferroptosis, focusing on established NRF2 target genes that mitigate these pathways, as well as the relevance of the NRF2-lipid peroxidation-ferroptosis axis in disease.
Article
One of the key challenges in cancer research is how to effectively kill cancer cells while leaving the healthy cells intact. Cancer cells often have defects in cell death executioner mechanisms, which is one of the main reasons for therapy resistance. To enable growth, cancer cells exhibit an increased iron demand compared with normal, non-cancer cells. This iron dependency can make cancer cells more vulnerable to iron-catalyzed necrosis, referred to as ferroptosis. The identification of FDA-approved drugs as ferroptosis inducers creates high expectations for the potential of ferroptosis to be a new promising way to kill therapy-resistant cancers.
Article
Friedreich's ataxia (FRDA) is a devastating, multisystemic disorder caused by recessive mutations in the mitochondrial protein frataxin (FXN). FXN participates in the biosynthesis of Fe-S clusters and is considered to be essential for viability. Here we report that when grown in 1% ambient O2, FXN null yeast, human cells, and nematodes are fully viable. In human cells, hypoxia restores steady-state levels of Fe-S clusters and normalizes ATF4, NRF2, and IRP2 signaling events associated with FRDA. Cellular studies and in vitro reconstitution indicate that hypoxia acts through HIF-independent mechanisms that increase bioavailable iron as well as directly activate Fe-S synthesis. In a mouse model of FRDA, breathing 11% O2 attenuates the progression of ataxia, whereas breathing 55% O2 hastens it. Our work identifies oxygen as a key environmental variable in the pathogenesis associated with FXN depletion, with important mechanistic and therapeutic implications.
Article
Ischemia-reperfusion injury (IRI) is the outcome of an inflammatory process that is triggered when an organ undergoes a transient reduction or cessation of blood flow, followed by re-establishment of perfusion. In the clinical setting, IRI contributes to significant acute kidney injury, patient morbidity and mortality, and adverse outcomes in transplantation. Tubular cell death by necrosis and apoptosis is a central feature of renal IRI. Recent research has challenged traditional views of cell death by identifying new pathways in which cells die in a regulated manner but with the morphologic features of necrosis. This regulated necrosis (RN) takes several forms, with necroptosis and ferroptosis being the best described. The precise mechanisms and relationships between the RN pathways in renal IRI are currently the subject of active research. The common endpoint of RN is cell membrane rupture, resulting in the release of cytosolic components with subsequent inflammation and activation of the immune system. We review the evidence and mechanisms of RN in the kidney following renal IRI, and discuss the use of small molecule inhibitors and genetically modified mice to better understand this process and guide potentially novel therapeutic interventions.
Article
Friedreich ataxia (FRDA) is a progressive neuro- and cardio-degenerative disorder characterized by ataxia, sensory loss, and hypertrophic cardiomyopathy. In most cases, the disorder is caused by GAA repeat expansions in the first introns of both alleles of the FXN gene, resulting in decreased expression of the encoded protein, frataxin. Frataxin localizes to the mitochondrial matrix and is required for iron-sulfur-cluster biosynthesis. Decreased expression of frataxin is associated with mitochondrial dysfunction, mitochondrial iron accumulation, and increased oxidative stress. Ferropotosis is a recently identified pathway of regulated, iron-dependent cell death, which is biochemically distinct from apoptosis. We evaluated whether there is evidence for ferroptotic pathway activation in cellular models of FRDA. We found that primary patient-derived fibroblasts, murine fibroblasts with FRDA-associated mutations, and murine fibroblasts in which a repeat expansion had been introduced (knockin/knockout) were more sensitive than normal control cells to erastin, a known ferroptosis inducer. We also found that the ferroptosis inhibitors ethyl 3-(benzylamino)-4-(cyclohexylamino)benzoate (SRS11-92) and ethyl 3-amino-4-(cyclohexylamino)benzoate, used at 500 nM, were efficacious in protecting human and mouse cellular models of FRDA treated with ferric ammonium citrate (FAC) and an inhibitor of glutathione synthesis [L-buthionine (S,R)-sulfoximine (BSO)], whereas caspase-3 inhibitors failed to show significant biologic activity. Cells treated with FAC and BSO consistently showed decreased glutathione-dependent peroxidase activity and increased lipid peroxidation, both hallmarks of ferroptosis. Finally, the ferroptosis inhibitor SRS11-92 decreased the cell death associated with frataxin knockdown in healthy human fibroblasts. Taken together, these data suggest that ferroptosis inhibitors may have therapeutic potential in FRDA.
Article
Ferroptosis is a regulated necrosis process driven by iron-dependent lipid peroxidation. Although ferroptosis and cellular metabolism interplay with one another, whether mitochondria are involved in ferroptosis is under debate. Here, we demonstrate that mitochondria play a crucial role in cysteinedeprivation- induced ferroptosis but not in that induced by inhibiting glutathione peroxidase-4 (GPX4), the most downstream component of the ferroptosis pathway. Mechanistically, cysteine deprivation leads to mitochondrial membrane potential hyperpolarization and lipid peroxide accumulation. Inhibition of mitochondrial TCA cycle or electron transfer chain (ETC) mitigated mitochondrial membrane potential hyperpolarization, lipid peroxide accumulation, and ferroptosis. Blockage of glutaminolysis had the same inhibitory effect, which was counteracted by supplying downstream TCA cycle intermediates. Importantly, loss of function of fumarate hydratase, a tumor suppressor and TCA cycle component, confers resistance to cysteine-deprivation- induced ferroptosis. Collectively, this work demonstrates the crucial role of mitochondria in cysteine-deprivation-induced ferroptosis and implicates ferroptosis in tumor suppression.
Article
Dihydroartemisinin (DHA) has been shown to be capable of inhibiting cancer growth, whereas it remains largely elusive that the underlying molecular mechanism of DHA induced acute myeloid leukemia (AML) cell death. In the present study, we examined the effects of DHA on the proliferation and ferroptosis of AML cells as well as to elucidate the underlying molecular mechanisms. We found that DHA strongly inhibited the viability of AML cell lines and arrest cell cycle at G0/G1 phase. Further studies found that DHA effectively induced AML cells ferroptosis, which was iron-dependent and accompanied by mitochondrial dysfunction. Mechanistically, DHA induced autophagy by regulating the activity of AMPK/mTOR/p70S6k signaling pathway, which accelerated the degradation of ferritin, increased the labile iron pool, promoted the accumulation of cellular ROS and eventually led to ferroptotic cell death. Over expression of ISCU (Iron-sulfur cluster assembly enzyme, a mitochondrial protein) significantly attenuated DHA induced ferroptosis by regulating iron metabolism, rescuing the mitochondrial function and increasing the level of GSH. Meanwhile, FTH reconstituted AML cells also exhibited the reduced lipid peroxides content and restored the DHA-induced ferroptosis. In summary, these results provide experimental evidences on the detailed mechanism of DHA-induced ferroptosis and reveal that DHA might represent a promising therapeutic agent to preferentially target AML cells.