Tryparedoxin peroxidase-deficiency commits trypanosomes to ferroptosis-type cell death

  1. Marta Bogacz
  2. R Luise Krauth-Siegel  Is a corresponding author
  1. Biochemie-Zentrum der Universität Heidelberg, Germany
8 figures, 1 table and 2 additional files

Figures

Figure 1 with 1 supplement
Ferroptosis inhibitors protect the Px I-III KO cells.

The mutant cells were either kept in Trolox-supplemented medium or transferred into medium containing 100 µM Dfx, 100 nM ferrostatin-1 or 200 nM liproxstatin-1 or no addition. (A) Every hour viable cells were counted. The data are the mean ± SD of three independent experiments. (B) The cells were kept for 2 hr in medium ± Trolox, incubated for 30 min with 10 µM H2DCFDA in medium + Trolox and subjected to flow cytometry measuring DCF fluorescence. (C) BODIPY was added and after 2 hr incubation the cells were analyzed by flow cytometry. (B and C) show representative histograms from two independent experiments.

https://doi.org/10.7554/eLife.37503.003
Figure 1—figure supplement 1
Protection of Px I-III KO cells by Trolox, Dfx and α-Toc.

The cells were incubated in the presence of (A) different concentrations of Trolox, (B) various concentrations of Dfx, (C) in medium supplemented with 10 µM Trolox or 20 µM Dfx alone or in combination and (D) different concentrations of α-Toc. (E) The cells were pre-cultured for 18 hr in the presence or absence of 10 µM α-Toc in medium containing Trolox, washed with PBS and transferred into medium ± α-Toc. After different times, viable cells were counted and the percentage relative to the start cell density was calculated. The data are the mean ± SD of three independent experiments.

https://doi.org/10.7554/eLife.37503.004
Figure 2 with 1 supplement
Px I-III KO cells are affected in mitochondrial matrix – but not outer membrane – immuno-staining.

The parasites were incubated for 1 or 2 hr in medium ± Trolox, treated with MitoTracker (red) and subjected to immunofluorescence microscopy using antibodies against (A) mtTXNPx (green) or (B) VDAC (green). Nuclear and kinetoplast DNA were visualized by DAPI staining (blue). Merge, overlay of the respective three signals. Phase, phase contrast image. Scale bare 10 µm.

https://doi.org/10.7554/eLife.37503.005
Figure 2—figure supplement 1
Mitochondrial damage precedes plasma membrane leakage.

The Px I-III KO cells were incubated ± Trolox for up to 4 hr, stained with MitoTracker or propidium iodide and subjected to flow cytometry. (A) Forward- (FSC) and side-scatter (SSC) DotPlots of MitoTracker-treated cells. The cells were gated in three sub-populations (P1-3) representing cells with normal FSC and SSC (P1; green), cells with reduced SSC (P2; magenta), and mainly dead cells (P3; grey). (B) Histograms of MitoTracker and propidium iodide signals of all single cells. The figure shows representative results of two independent experiments.

https://doi.org/10.7554/eLife.37503.006
Figure 3 with 1 supplement
PC Px I-III KO T. brucei undergo strinking ultrastructural changes at their single mitochondrion.

Parasites kept in the presence or absence of Trolox were fixed, processed and subjected to transmission electron microscopy as described under Materials and methods. Electron micrographs of representative cells incubated for (A) 2 hr in Trolox-containing medium and (B and C) 2 hr in Trolox-free medium. N, nucleus; LP, lipid droplet; G, glycosome; ER, endoplasmic reticulum; K, kinetoplast; GA, Golgi apparatus; M, mitochondrion; A, acidocalcisome; black arrow heads, normal cristae; white arrow heads, enlarged cristae. The inserts in (A) and (C) show higher magnifications of mitochondria to highlight the normal and altered morphology, respectively. The double white arrow heads point to the three membranes that surround a dilated cristae. Scale bars 500 nm. (D) Quantification of different phenotypes observed. In the presence of Trolox or for ≤0.5 hr without the antioxidant, the mutant cells were indistinguishable from wild type parasites (not shown). The cytosol was densely packed and the mitochondrion had a comparable or even lower electron density, defined as normal. In the absence of Trolox, the Px I-III KO parasites displayed a time-dependent darkening of the mitochondrion and lightening of the cytosol. Only parasites that clearly displayed an elongated cell body in the electron micrographs were incorporated in the analysis. The number of cells inspected at the different time points is given above the columns. The percentage of each phenotype in the total number of inspected cells is depicted.

https://doi.org/10.7554/eLife.37503.007
Figure 3—figure supplement 1
Short-term incubation of Px I-III KO cells in Trolox-free medium results in mitochondrial alterations without changes at other organelles, whereas prolonged incubation can finally lead to plasma membrane blebs.

Electron micrographs of Px I-III KO parasites kept for (A) 1 hr and (B) 2 hr in Trolox-free medium. (A) The intact elongated mitochondrion is darker than the cytosol. Other subcellular structures appear to be unaffected. (B) One of the very few cells that after 2 hr in Trolox-free medium displayed already a plasma membrane damage. The high magnification shows vesicular structures blebbing out of the plasma membrane (double arrow heads). N, nucleus; LP, lipid droplet; G, glycosome; ER, endoplasmic reticulum; M, mitochondrion; A, acidocalcisome. Scale bar 500 nm.

https://doi.org/10.7554/eLife.37503.008
Figure 4 with 1 supplement
MitoSOX Red exerts a dual effect on the Px I-III KO cells, sensing oxidant production and acting as protecting agent.

(A) The cells were pre-loaded with MitoSOX in Trolox-containing medium, transferred into medium ± Trolox, incubated for 2 hr, and stained with MitoTracker Green. Nuclear (large dot) and kinetoplast (small dot) DNA were visualized by Hoechst 33342 staining. Representative fluorescence microscopy images are depicted. Merge, overlay of all three signals. Phase, phase contrast image. Cells kept in the presence of Trolox displayed a very small red MitoSOX signal that co-localized with the kinetoplast (upper panel). After 2 hr in the absence of Trolox, most cells still had normal morphology but many of them displayed a more intense kinetoplast MitoSOX signal (lower two panels). Scale bar 10 µm. (B–D) The cells were incubated ± MitoSOX in Trolox-containing medium, transferred into medium ± Trolox, incubated for up to 5 hr, stained with DAPI, and subjected to flow cytometry. (B) Representative histograms of the 488:585/42 (ex:em) channel (MitoSOX signal of the treated cells and auto-fluorescence of non-treated cells) and DAPI signal of all single cells from samples kept for 5 hr ± Trolox, with or without loading with MitoSOX. (C) MitoSOX fluorescence change between cells kept in the absence and presence of Trolox. The data represent the mean ± error of the mean of two independent experiments. (D) Representative FSC:SSC DotPlots of cells kept for 5 hr in the presence or absence of Trolox, with or without loading with MitoSOX. The cells were gated in three sub-populations (P1-3) representing cells with normal FSC and SSC (P1; green), those with reduced SSC (P2; magenta), and severely damaged or dead cells (P3; grey).

https://doi.org/10.7554/eLife.37503.009
Figure 4—figure supplement 1
The P2 sub-population displays the strongest increase in MitoSOX fluorescence.

The Px I-III KO cells were loaded with MitoSOX for 10 min in Trolox-supplemented medium, washed with PBS and re-suspended in medium ± Trolox. After 5 hr incubation, the cells were stained with DAPI and subjected to flow cytometry. The histograms represent overlays of the DAPI (green) and MitoSOX (magenta) fluorescence signals of cells kept in the presence (tinted peak) or absence (transparent peak) of Trolox. The upper, central and lower panels show the fluorescence intensities for the P1, P2, and P3 subsets, representing cells with normal FSC:SSC, decreased SSC and dead cells, respectively, as defined in Figure 4. The percentage of cells with increased signals (black range line) are given in each histogram. The figure shows representative results of one of two independent experiments.

https://doi.org/10.7554/eLife.37503.010
Cellular ATP levels rapidly decrease when Px I-III KO cells are in medium lacking a protecting agent.

The cells were incubated ± Trolox in (A) SDM-79 medium or (B) MEM-Pros medium. After 0 to 4 hr, aliquots of each sample were removed and (A) cells with normal morphology or only highly motile cells were counted, or (B) the cells were treated with PI and the fluorescence was measured by flow cytometry. The remaining cells were treated with ATPlite one-step solution and the luminescence was measured in the plate reader. The data are given as percentage of the respective value at 0 hr that was set as 100%. They represent the mean ± SD of three independent experiments.

https://doi.org/10.7554/eLife.37503.011
Lipid peroxidation in the Px I-III KO cells originates at the mitochondrion.

(A) Px I-III KO parasites in Trolox-supplemented medium were incubated for 1.5 hr with MitoPerOx, stained with MitoTracker Green and subjected to life cell fluorescence microscopy. Representative images are depicted. Merge, overlay of both signals. Phase, phase contrast image. Scale bar 10 µm. (B) The cells were transferred into medium ± Trolox. BODIPY or MitoPerOx was added and after different times cells were analyzed by flow cytometry. Histograms of BODIPY (upper panel) and MitoPerOx (lower panel) fluorescence of all single cells at 520 nm from a representative experiment are depicted. (C) Quantitative analysis of the percentage of cells with increased BODIPY and MitoPerOx signals (gating as depicted in B). The data represent the mean ± SD of three independent experiments.

https://doi.org/10.7554/eLife.37503.012
Px I-III KO cells that overexpress mitochondrial SODA require less Dfx for survival.

(A) Total lysates of 5 × 106 Px I-III KO/SODA-myc cells cultured for 18 hr ± tetracycline (Tet) were subjected to Western blot analysis using antibodies against myc as well as aldolase for loading control. (B) Px I-III KO/SODA-myc cells cultured for 18 hr in the presence of Tet were treated with MitoTracker (red, upper panel) followed by antibodies against Myc (green) or simultaneously with antibodies against Myc and cytosolic 2-Cys-peroxiredoxin (cTXNPx) (red; lower panel). Nuclear and kinetoplast DNA were stained with DAPI (blue) and the cells were subjected to immunofluorescence microscopy. Merge, overlay of the Myc with the MitoTracker or cTXNPx signal. Phase + DAPI, phase contrast image with the nucleus (large dot) and the kinetoplast (small dot) visualized by DAPI staining. Scale bar 10 µm. (C) Px I-III KO and Px I-III KO/SODA-myc cells cultured for 18 hr ± Tet in Trolox-supplemented medium were transferred into Trolox-free medium and incubated ± Tet and ± 25 µM Dfx. Every hour viable cells were counted. The data represent the mean ± SD of three independent experiments. (D) Px I-III KO/SODA-myc cells cultured for 18 hr ± Tet in Trolox-supplemented medium were transferred into Trolox-free medium and incubated ± Tet and ± 25 µM Dfx for 1–4 hr starting with the longest time point. Cells were stained with PI and subjected to flow cytometry. Data show the mean + SEM of the PI fluorescence of all single cells from two experiments. Data were analyzed by two-way ANOVA with Bonferroni post-test, *p<0.05.

https://doi.org/10.7554/eLife.37503.013
Mitochondrial iron is involved in the death phenotype of Px I-III-deficient cells.

(A) Cells were pre-loaded with 150 nM RPA or 10 nM RPAC in PBS, re-transferred into Trolox-supplemented medium, stained with MitoTracker Green and subjected to life cell imaging. Merge, overlay of the RPA and RPAC fluorescence, respectively, with the MitoTracker signal. Phase, phase contrast image. (B) Cells pre-loaded with 150 nM RPA or 10 nM RPAC were transferred into medium ± Trolox and incubated for up to 3 hr. The upper panels show the RPA or RPAC fluorescence, the lower ones the respective phase contrast pictures. (A and B) Representative parasites are depicted. Scale bars 10 µm. (C) The cells were pre-loaded with 0.15, 0.5 or 1.5 µM RPA, transferred into medium – Trolox and incubated for 6 hr. Every hour, viable cells were counted. Cells not treated with RPA (no addition) served as control. The data are the mean ± SD of three independent experiments. (D) FSC:SSC dot plots of cells that were treated with 150 nM RPA or RPAC and incubated for 3 hr ± Trolox. P1 (green), P2 (magenta) and P3 (grey) gates determined as defined in Figure 4. A representative histogram of two independent experiments is shown. (E) RPA and RPAC fluorescence of cells stained with 50 nM RPA or 1 nM RPAC and incubated ± Trolox for 0–3 hr. (F) RPA fluorescence of cells incubated for 2 hr in medium supplemented with 100 µM Trolox ± 100 µM Fe(III)/HQ complex, 100 nM ferrostatin-1 or 100 µM Dfx and stained with 50 nM RPA. (G) RPA fluorescence of Px I-III KO/SODA-myc cells grown for 20 hr in the presence or absence of Tet and stained with 50 nM RPA. (E–G) Representative histograms from one of at least three independent experiments are depicted.

https://doi.org/10.7554/eLife.37503.014

Tables

Key resources table
Reagent type
(species) or
resource
DesignationSource or referenceIdentifiersAdditional information
Gene
(Trypanosoma
brucei brucei)
sodATriTrypDatabase ID:
Tb427.05.3350
Cell line
(Trypanosoma
brucei brucei)
WT PC and BSPMID: 9108552WT PC and BS 449
Lister strain 427
Culture-adapted T. brucei
strains stably expressing
the tetracycline repressor
Cell line
(Trypanosoma
brucei brucei)
PC Px I-III KOPMID: 26374473PC WT cells in which both
alleles of the complete
px locus are replaced by
resistance casettes
(blasticidin and puromycin)
Cell line
(Trypanosoma
brucei brucei)
PC Px I-III KO/SODA-mycthis workPC Px I-III KO cells that
contain a Tet-inducible
construct for SODA-c-myc2
overexpression
AntibodyRabbit anti-cTXNPxPMID: 20826821IF (1:1000)
AntibodyGuinea pig anti-mTXNPxPMID: 29413965IF (1:1000)
AntibodyRabbit anti-aldolaseChristine Clayton,
Heidelberg, Germany
WB (1:20000)
AntibodyRabbit anti-VDACAndré Schneider,
Bern, Switzerland
IF (1:500)
AntibodyMouse anti-c-Myc
(monoclonal)
Santa Cruz
Biotechnology
sc-40,
RRID:AB_627268
WB, IF (1:200)
AntibodyHRP-conjugated
goat anti-mouse IgGs
Santa Cruz
Biotechnology
sc-2005,
RRID:AB_631736
WB (1:5000)
AntibodyHRP-conjugated
goat anti-rabbit IgGs
Santa Cruz
Biotechnology
sc-2004,
RRID:AB_631746
WB (1:10000)
AntibodyAlexa Fluor 488
goat anti-guinea pig IgGs
Thermo Fisher
Scientific
A11073,
RRID:AB_142018
IF (1:1000)
AntibodyAlexa Fluor 488
goat anti-mouse IgGs
Thermo Fisher
Scientific
A11001,
RRID:AB_2534069
IF (1:250)
AntibodyAlexa Fluor 488
goat anti-rabbit IgGs
Thermo Fisher
Scientific
A11008,
RRID:AB_143165
IF (1:1000)
AntibodyAlexa Fluor 594
goat anti-rabbit IgGs
Thermo Fisher
Scientific
A11012,
RRID:AB_141359
IF (1:1000)
Recombinant
DNA reagent
pHD1700/grx2-
c-myc2 (plasmid)
PMID: 20826822
Recombinant
DNA reagent
pHD1700/sodA-
c-myc2 (plasmid)
this work
Sequence-based
reagent
5'CGATAAGCTTATG
AGGTCTGTCATGATGC3'
this workPrimer for amplification
of sodA from genomic
T. brucei DNA
Sequence-based
reagent
5'CGATGGATCCCTT
CATAGCCTGTTCATAC3'
this workPrimer for amplification
of sodA from genomic
T. brucei DNA
Peptide,
recombinant
protein
T. brucei
trypanothione
reductase (TR)
PMID: 24788386
Peptide,
recombinant
protein
T. brucei
tryparedoxin (Tpx)
PMID: 22275351
Peptide,
recombinant
protein
T. brucei
peroxidase (Px)
PMID: 18684708
Chemical
compound, drug
trypanothione
and trypanothione
disulfide
PMID:19477177
Chemical
compound, drug
Chlorhexidine
diacetate
Monohydrate
Fluka24800
Chemical
compound, drug
(1S,3R)-RSL3José Pedro Friedmann
Angeli and Marcus
Conrad, Munich,
Germany and Cayman
Chemical
CAS: 1219810-16-8
Chemical
compound, drug
Racemic
mixture of RSL3
José Pedro Friedmann
Angeli and Marcus
Conrad, Munich,
Germany
Chemical
compound, drug
Ferrostatin-1Sigma-AldrichSML0583
Chemical
compound, drug
Liproxstatin-1Sigma-AldrichSML1414
Chemical
compound, drug
TroloxSigma-Aldrich238813
Chemical
compound, drug
Iron(III) chloride
hexahydrate,
FeCl3 × 6H2O
Merk31232
Chemical
compound, drug
Deferoxamine
mesylate
Sigma-AldrichD-9533
Chemical
compound, drug
8-HydroxyquinolineSigma-Aldrich252565
Chemical
compound, drug
(±)-α-TocopherolSigma-AldrichT3251
Chemical
compound, drug
ATPlite 1step solutionPerkin Elmer6016731
Chemical
compound, drug
Hoechst 33342Walter Nickel,
Heidelberg,
Germany
Chemical
compound, drug
4’,6-Diamidino-2
-phenylindole (DAPI)
Sigma-AldrichD-8417
Chemical
compound, drug
Rhodamine
B-[(1,10-phenanthroline-5-yl)-
aminocarbonyl]benzyl
ester (RPA)
Squarix
Biotechnology
ME043.1
Chemical
compound, drug
Rhodamine
B-[(phenanthren-9-yl)
-aminocarbonyl]-
benzylester (RPAC)
Squarix
Biotechnology
ME046.1
Chemical
compound, drug
BODIPY 581/591 C11Molecular ProbesD3861
Chemical
compound, drug
MitoPerOxMike Murphy,
Cambridge, UK
Chemical
compound, drug
MitoTracker Red CMXRosMolecular ProbesM7512
Chemical
compound, drug
MitoTracker Green FMMolecular ProbesM7514
Chemical
compound, drug
MitoSOX RedMolecular ProbesM36008
Chemical
compound, drug
Propidium iodide (PI)Molecular ProbesP3566
Chemical
compound, drug
2',7'-Dichlorodihydro
fluorescein-diacetate
(H2DCFDA)
Molecular ProbesD399

Additional files

Supplementary file 1

Trypanocidal activity and in vitro inhibitory potency of RSL3.

(A) Bloodstream T. brucei were cultured for 1, 2 or 3 days in the presence of 100 µM Trolox, 200 nM Liproxstatin-1 or 100 nM Ferrostatin-1 as well as different RSL3 concentrations and then subjected to plate reader-based ATPlite measurements. Chlorhexidine, a trypanocidal compound and known inhibitor of trypanothione reductase (TR) (Meiering et al., 2005; Beig et al., 2015), served as positive control. 1The data are the mean of at least two independent series of experiments each conducted in triplicate with standard deviations (SD). 2The values are the mean of an experiment conducted in triplicate with SD. (B) NADPH, T(SH)2, TR, and Tpx ± Px were incubated with 40 µM RSL3. After different times, the assays were started by adding (a) 100 µM H2O2 or (b) Px and H2O2. The data were derived from at least double determinations which varied by ≤ 10%.

https://doi.org/10.7554/eLife.37503.015
Transparent reporting form
https://doi.org/10.7554/eLife.37503.016

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Marta Bogacz
  2. R Luise Krauth-Siegel
(2018)
Tryparedoxin peroxidase-deficiency commits trypanosomes to ferroptosis-type cell death
eLife 7:e37503.
https://doi.org/10.7554/eLife.37503