ArticlePDF Available

P18 Proteome adaptations in ETHE1 deficient mice indicate a role of sulfide signaling in lipid catabolism and cytoskeleton organization via post-translational protein modifications

Authors:

Abstract and Figures

Hydrogen sulfide is a physiologically relevant signaling molecule. However, circulating levels of this highly biologically active substance have to be maintained within tightly controlled limits in order to avoid toxic side effects. In patients suffering from ethylmalonic encephalopathy a block in sulfide oxidation at the level of the sulfur dioxygenase ETHE1 leads to severe dysfunctions in microcirculation and cellular energy metabolism. We used an Ethe1 deficient mouse model to investigate the effect of increased sulfide and persulfide concentrations on liver, kidney, muscle and brain proteomes. Major disturbances in post-translational protein modifications indicate a crucial role of the mitochondrial sulfide oxidation pathway in sulfide signaling. Our results confirm the involvement of sulfide in redox regulation and cytoskeleton dynamics. In addition, they suggest that sulfide signaling specifically regulates mitochondrial catabolism of fatty acids and branched-chain amino acids.
Content may be subject to copyright.
Biosci. Rep. (2013) / 33 / art:e00052 / doi 10.1042/BSR20130051
Proteome adaptations in Ethe1-deficient mice
indicate a role in lipid catabolism and
cytoskeleton organization via post-translational
protein modifications
Tatjana M. HILDEBRANDT*1, Ivano DI MEO†, Massimo ZEVIANI†‡, Carlo VISCOMI† and Hans-Peter BRAUN*
*Institut f¨
ur Pflanzengenetik, Leibniz Universit¨
at Hannover, Herrenh¨
auser Straße 2, 30419 Hannover, Germany, †IRCCS Foundation
Neurological Institute ‘C. Besta’, Via Temolo 4, 20126 Milano, Italy, and ‡Medical Research Council Mitochondrial Biology Unit, Wellcome
Trust/MRC Building, Hills Road, Cambridge CB2 0XY, U.K.
Synopsis
Hydrogen sulfide is a physiologically relevant signalling molecule. However, circulating levels of this highly biologic-
ally active substance have to be maintained within tightly controlled limits in order to avoid toxic side effects. In
patients suffering from EE (ethylmalonic encephalopathy), a block in sulfide oxidation at the level of the SDO (sulfur
dioxygenase) ETHE1 leads to severe dysfunctions in microcirculation and cellular energy metabolism. We used an
Ethe1-deficient mouse model to investigate the effect of increased sulfide and persulfide concentrations on liver,
kidney, muscle and brain proteomes. Major disturbances in post-translational protein modifications indicate that the
mitochondrial sulfide oxidation pathway could have a crucial function during sulfide signalling most probably via
the regulation of cysteine S-modifications. Our results confirm the involvement of sulfide in redox regulation and
cytoskeleton dynamics. In addition, they suggest that sulfide signalling specifically regulates mitochondrial catabol-
ism of FAs (fatty acids) and BCAAs (branched-chain amino acids). These findings are particularly relevant in the context
of EE since they may explain major symptoms of the disease.
Key words: branched-chain amino acid oxidation, ethylmalonic encephalopathy, hydrogen sulfide, mitochondria, redox
regulation, sulfur dioxygenase
Cite this article as: Hildebrandt, T.M., Di Meo, I., Zeviani, M., Viscomi, C. and Braun, H-P. (2013) Proteome adaptations in Ethe1
deficient mice indicate a role in lipid catabolism and cytoskeleton organization via post-translational protein modifications. Biosci.
Rep. 33(4), art:e00052.doi:10.1042/BSR20130051
INTRODUCTION
ETHE1 is a mitochondrial SDO (sulfur dioxygenase) [1], which
takes part in sulfide detoxification by oxidizing persulfides to
sulfite [2]. Animal cells produce H2S during the catabolism of
sulfur-containing amino acids [3], and an additional source is the
large intestine where anaerobic bacteria reduce sulfate to sulf-
ide [4]. Endogenous H2S functions as a gaseous messenger and
has been suggested to be involved in the regulation of several
physiological processes such as vascular tone, insulin secretion
and inflammation, whereas elevated sulfide concentrations are
............................................................................................................................................................................................................................................................................................................
Abbreviations used: BCAA, branched-chain amino acid; BCKDH, branched-chain α-keto acid dehydrogenase; COX, cytochrome coxidase; DTT, dithiothreitol; EE, ethylmalonic
encephalopathy; ETF, electron transfer flavoprotein; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; IEF, isoelectric focusing; IPG, immobilized pH gradient; KGDH, 3-oxoglutarate
dehydrogenase; PDH, pyruvate dehydrogenase; pI, isoelectric point; PTM, post-translational modification; SCAD, short/branched-chain acyl-CoA dehydrogenase; SDO, sulfur
dioxygenase; QPCR, quantitative PCR.
1To whom correspondence should be addressed (email hildebrandt@genetik.uni-hannover.de).
highly toxic. The exact mechanism of sulfide signalling is largely
unknown and thought to be based on the activation of transcrip-
tion factors as well as direct cysteine S-sulfhydration of target
proteins [5,6].
Mutations that lead to a loss of function in ETHE1 dis-
rupt mitochondrial sulfide oxidation and thereby cause the fatal
metabolic disorder, EE (ethylmalonic encephalopathy; OMIM
#602473) [7,8]. In patients the vascular endothelium is severely
damaged by toxic sulfide concentrations in the bloodstream lead-
ing to the main symptoms of EE: rapidly progressive neurological
failure due to multiple necrotic and haemorrhagic brain lesions,
chronic haemorrhagic diarrhoea, vascular petechial purpura and
c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
575
Bioscience Reports www.bioscirep.org
T. M. Hildebrandt and others
orthostatic acrocyanosis [9]. The biochemical profile, an increase
in urinary and plasmatic lactate, acylcarnitine, acylglycine and
ethylmalonic acid levels, indicates disturbances in mitochondrial
energy metabolism. This can at least partially be explained by
sulfide toxicity. H2S is a well-known direct inhibitor of COX
(cytochrome coxidase) [10], and in addition chronic exposure
destabilizes subunits of the enzyme leading to a severe COX
deficiency in muscle, brain and colonic mucosa of EE patients
[11]. The accumulation of ethylmalonic acid, which is a derivat-
ive of butyrate, as well as elevated C4 and C5 acylcarnitines and
acylglycines, reflect a block in oxidative metabolism of BCAAs
(branched-chain amino acids). Indeed H2S has been shown to in-
hibit one enzyme of this pathway, SCAD (short-chain acyl CoA
dehydrogenase), in vitro [1].
The severe systemic consequences of a dysfunction in the
sulfide oxidation system become obvious in patients suffering
from EE. The aim of this study was to identify the effects
of elevated sulfide concentrations on the protein composition of
different tissues in order to shed light on the role of sulfide in
cellular metabolism. For this purpose, the proteomes of liver, kid-
ney, skeletal muscle and brain were analysed in a recombinant
Ethe1-deficient mouse model, which faithfully recapitulates the
clinical and biochemical features of EE [1].
EXPERIMENTAL
Animals
Recombinant Ethe1 /mice were obtained as described [1]. The
mice were maintained on a C57BL6/129Sv mixed background.
Four-week-old Ethe1/and Ethe1 +/+female littermates were
considered in this study. Animal studies were in accordance with
the Italian Law D.L. 116/1992 and the EU directive 86/609/CEE.
Mice were maintained in a temperature- and humidity-controlled
animal-care facility, with a 12 h light/dark cycle and free access to
water and food (Standard Diet). Mice were killed by dislocation
of the neck.
Protein extraction and 2D gel electrophoresis
Tissue samples were frozen in liquid nitrogen and ground in
a shaker mill (Retsch). For IEF (isoelectric focusing) 4 mg of
each sample were suspended in 350 μl rehydration buffer (6 M
urea, 2 M thiourea, 50 mM DTT (dithiothreitol), 2 % CHAPS
(w/v), 5 % IPG (immobilized pH gradient) buffer 3–11 nl (v/v),
12 μl/ml DeStreak reagent and a trace of bromphenol blue) and
homogenized by sonication (3×20 s). Samples were centrifuged
at 17000 gfor 10 min and the protein content was adjusted
to 2.14 mg/ml (Quantkit, GE Healthcare). IEF was carried out
on Immobiline DryStrip gels (18 cm, non-linear gradient pH 3–
11) using the Ettan IPGphor 3 system (GE Healthcare). For the
second dimension (SDS/PAGE) IPG strips were equilibrated in
6 M urea, 30 % glycerol (87 %, v/v), 2 % SDS, 50 mM Tris/HCl
pH 8.8, bromphenol blue with (i) 1% DTT (w/v) and (ii) 2.5 %
Iodacetamide (w/v) and transferred horizontally onto 16.5 % Tri-
cine gels. Electrophoresis was carried out for 20 h at 35 mA/mm
gel layer in a Protean IIXL gel system (Biorad) using a broad-
range protein molecular mass marker (10–225 kDa, Promega) as
molecular mass standard.
Gel image analysis
Gels were stained overnight with colloidal CBB (Coomassie Bril-
liant Blue), CBB-250 G (Merck), scanned and analysed with
Delta2D software version 4.2 (Decodon). Six gels were used
for each tissue (three biological replicates) to compare the pro-
tein abundance between Ethe1 knockout mice and wild-type lit-
termates. Only protein spots with significant (Student’s ttest,
P<0.05), at least 1.5-fold differences in the relative spot volume
were assumed to be of different abundance.
Protein identification by MS
Spots were excised from the 2D gels using a manual spot picker
(Genetix), washed with 0.1 M ammonium bicarbonate and de-
hydrated with acetonitrile. For tryptic digestion the gel pieces
were incubated overnight at 37C(2μg/ml trypsin in 0.1 M
ammonium bicarbonate, Promega). Tryptic peptides were ex-
tracted by successive incubation with (i) 50% (v/v) acetoni-
trile, 5 % (v/v) formic acid (ii +iii) 50 % (v/v) acetonitrile, 1 %
(v/v) formic acid (iv) 100% acetonitrile for 15 min at 37 C.
Supernatants were pooled and dried by vacuum centrifugation.
Peptides were analysed using an EASY-nLC-system (Proxeon)
coupled to a MicrOTOF-Q-II mass spectrometer (Bruker Dalton-
ics) according to the protocol described in [12]. Proteins were
identified using the MASCOT search algorithm against Swiss-
Prot (www.uniprot.org) and NCBI non-redundant protein data-
base (www.ncbi.nlm.nih.gov/protein).
Data analysis
A co-expression analysis for the Ethe1 gene (Mm.29553,
1417203_at) was performed with Genevestigator (www.
genevestigator.com; Nebion). All 2910 wild-type genetic back-
ground samples available in the mouse 40 k microarray platform
of the manually curated database of Genevestigator were used
for the query. Co-expressed genes were identified by using the
Pearson correlation coefficient as the measure of similarity. The
bioinformatics tool DAVID [13] was used to identify enriched
functional annotation terms within the datasets.
Western blotting
Mouse tissues were homogenized in 10×(v/w)10 mM potassium
phosphate buffer, pH 7.4 and centrifuged at 800 gfor 10 min
in the presence of protease inhibitors. For the analysis of mito-
chondrial proteins, an additional high-speed centrifugation step
was used to collect the mitochondria. Samples were frozen and
thawed twice in liquid nitrogen. Approximately 60 μg of proteins
was used for each sample in denaturing SDS/PAGE. Western blot
..........................................................................................................................................................................................................................................................................................................................................................................
576 c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
Proteome changes in Ethe1 deficiency
Figure 1 Changes in protein abundance in Ethe1-deficient mouse tissues
Total proteins (0.75 mg) from liver (A), kidney (B), brain (C) and muscle (D) of three knockout mice and three wild-type
littermates were separated by IEF/SDS/PAGE (pH 3–11, 16% acr ylamide). The Coomassie Brilliant Blue stained gels were
analysed with Delta2D, and significantly different spots (P<0.05) with at least 1.5-fold changes in volume are numbered
in the fused images. Proteins were identified via nanoLC-MS/MS (see Table 1, Supplementary Table S1).
analysis was performed with α-ETHE1 [1], α-ETF (α-electron
transfer flavoprotein) [14], α-SCAD (abcam), α-SDH-A (Invitro-
gen), α-GAPDH (α-glyceraldehyde 3-phosphate dehydrogenase)
(Millipore), α-Actin (Millipore) and α-ACTA1 (Dako) antibod-
ies, using the ECL (enhanced chemiluminescence) (Amersham),
as described elsewhere [11].
PCR
Tissue-derived RNA was isolated with TRIzol reagent (Invitro-
gen). Two micrograms of total RNA was treated with RNase-free
DNase and retro-transcribed by using the Cloned AMV First-
strand cDNA Synthesis kit and protocol (Invitrogen). Approx-
imately 2–5 ng of cDNA was used for SYBR-GREEN based
real-time PCR using primers specific for the amplification of
the genes of interest (oligo sequences available on request) ac-
cording to the ABI-Primer Express software. Standard transcript
HPRT (hypoxanthine-guanine phosphoribosyltransferase) was
co-amplified using suitable primers. Real-time QPCR (quantitat-
ive PCR) was carried out using an ABI PRISM 7000 Sequence
Detection System. The amplification profile was according to
a two-step protocol: one cycle at 50C for 2 min, one cycle at
95 C for 10 min and then 40 cycles of 95C for 15 s and 60 C
for 1 min. A final dissociation step (95 C for 15 s, 60 C for 20 s,
95for 15 s) was added to assess for unspecific primer–dimer
amplifications.
RESULTS
Effects of Ethe1 deficiency on mouse liver, kidney,
brain and muscle proteomes
Total protein extracts from liver, kidney, brain and skeletal muscle
were separated by 2D IEF/SDS/PAGE resulting in 70 spots with
significantly different volumes in Ethe1-deficient mice com-
pared with wild-type littermates (Figure 1) that contained 81
proteins (Tab l e 1 , Supplementary Table S1 available at http://
www.bioscirep.org/bsr/033/bsr033e052add.htm). These proteins
identified in spots with a changed volume will be described by the
shorter term ‘changed/affected proteins’ throughout the paper. In
parallel, an unbiased co-expression analysis based on microar-
ray data available in public repositories was carried out using
the Genevestigator software. The 81 top scoring genes for co-
expression with Ethe1 in wild-type mouse tissues were identified
..........................................................................................................................................................................................................................................................................................................................................................................
c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
577
T. M. Hildebrandt and others
Table 1 Proteins with potentially changed abundance in liver,
kidney, brain or muscle of Ethe1-deficient mice
Proteins were separated by IEF/SDS/PAGE (Figure 1) and spots
with at least 1.5-fold changes in volume (k.o./wt) were analysed by
nanoLC-MS/MS. For additional information see Supplementary Table
S1 (available at http://www.bioscirep.org/bsr/033/bsr033e052add.
htm).
Spot Accession Name k.o./wt
Liver
1 P60710 β-Actin 1 0.43
2 Q64374 Regucalcin 0.53
3 Q91Y97 Fructose-bisphosphate aldolase B 0.54
Q8VCH0 3-ketoacyl-CoA thiolase B 0.54
4 P97823 Acyl-protein thioesterase 1 0.57
5 Q8R086 Sulfite oxidase 0.57
Q63836 Selenium-binding protein 2 0.57
6 P29758 Ornithine aminotransferase 0.59
7 Intermediate filament protein 0.62
O08709 Peroxiredoxin 6 0.62
8 Q80W21 Glutathione S-transferase Mu 7 0.62
9 P30416 Peptidyl-prolyl cis-trans isomerase 0.66
10 Q03265 ATP synthase subunit alpha 1.57
11 P10649 GST Mu 1 1.58
12 P63038 HSP 60 1.66
13 P55264 Adenosine kinase 1.68
14 Q8BH95 Enoyl-CoA hydratase 2.15
15 Q9DCW4 Electron transfer flavoprotein 2.62
16 Q9QXF8 Glycine N-methyltransferase 2.85
17 P35700 Peroxiredoxin 1 3.01
Kidney
18 Q60866 Phosphotriesterase 0.34
19 P28825 Meprin A subunit alpha 0.37
20 Q9JHW2 Omega-amidase NIT2 0.41
21 Q9QUM9 Proteasome subunit alpha 0.42
22 Q9DBJ1 Phosphoglycerate mutase 1 0.42
23 Q9QYR9 Acyl-coenzyme A thioesterase 0.47
24 Q8CAQ8 Mitofilin 0.51
25 Q99J99 3-mercaptopyruvate sulfurtransferase 0.52
26 Q9WU78 PCD 6-interacting protein 0.53
27 Q60866 Phosphotriesterase 0.53
28 P11352 Glutathione peroxidase 1 0.57
29 P27773 Protein disulfide-isomerase A3 0.58
30 P28825 Meprin A 0.58
31 P47738 Aldehyde DH 0.59
32 P28825 Meprin A 0.61
33 P09103 Protein disulfide-isomerase A1 0.61
34 Q9D964 Glycine amidinotransferase 0.66
35 Q9JK42 PDH kinase 1.64
36 Q9DBL1 SCAD 1.64
37 Q62433 Protein NDRG1 1.67
38 Q6NZD1 Sulfotransferase 1.70
39 Q91VR2 ATP synthase subunit gamma 1.74
Q64669 NAD(P)H DH 1.74
Brain
40 P09103 Protein disulfide-isomerase A1 0.44
Table 1 Continue
Spot Accession Name k.o./wt
41 Q03265 ATP synthase subunit alpha 0.47
42 P14206 40S ribosomal protein SA 0.56
Q61937 Nucleophosmin 0.56
43 O55131 Septin-7 0.59
44 Q64332 Synapsin-2b 0.59
P06745 Glucose-6-phosphate isomerase 0.59
45 P52480 Pyr uvate kinase 0.64
46 P17751 Triosephosphate isomerase 1.54
47 Q03265 ATP synthase subunit alpha 1.58
48 Q61792 LIM and SH3 domain protein 1 1.59
49 P38647 Stress-70 protein 1.61
50 P35505 Fumarylacetoacetase 1.65
51 P80317 T-complex protein 1 1.87
52 P27773 Protein disulfide-isomerase A3 1.88
53 Q91YT0 NADH DH flavoprotein 1 1.94
P10126 Elongation factor 1-alpha 1 1.94
O55131 Septin-7 1.94
54 P16858 GAPDH 2.08
55 Q60932 VDAC protein 1 2.14
P00920 Carbonic anhydrase 2 2.14
56 P17742 Peptidyl-prolyl cis-trans isomerase A 2.19
57 P63101 14-3-3 protein ζ/δ3.06
58 Q9D2G2 2-oxoglutarate DH E2 3.45
Muscle
59 Q9JKS4 LIM domain-binding protein 3 0.28
60 Q9JIF9 Myotilin 0.34
61 Q9D0F9 Phosphoglucomutase-1 0.41
62 Q62167 RNA helicase DDX3X 0.53
63 P19157 GST P1 0.61
64 Q00898 α-1-antitrypsin 1-5 0.65
P62737 Actin, aortic smooth muscle 0.65
65 P62737 Actin, aor tic smooth muscle 1.50
P07758 α-1-antitrypsin 1-1 1.50
66 Q9D6R2 Isocitrate DH 1.58
Q9JHR4 myosin heavy chain IIB 1.58
67 Q99JY0 3-ketoacyl-CoA-thiolase 1.61
68 P14602 HSPβ1 1.61
69 P97457 Myosin regulator y light chain 2 1.67
70 P04117 FA-binding protein 2.25
(Supplementary Table S2 available at http://www.bioscirep.org/
bsr/033/bsr033e052add.htm) and will be called ‘co-expressed
genes/proteins’.
In order to obtain a general overview on which pathways or
functional categories are associated with Ethe1, we carried out en-
richment analysis with the software tool DAVID on the two com-
plete data sets generated by either proteomics or co-expression
analysis. The majority of differentially abundant proteins were
those regulated by PTMs (post-translational modifications, acet-
ylation and phosphorylation) or with specific subcellular local-
ization (cytosolic and mitochondrial) (Tab l e 2 ). While the latter
..........................................................................................................................................................................................................................................................................................................................................................................
578 c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
Proteome changes in Ethe1 deficiency
Table 2 Functional annotation analysis
Proteins with different abundance in Ethe1 deficient compared with wild-type mouse liver, kidney, brain and muscle (proteomics) and of genes that
are co-expressed with Ethe1 in wild-type mouse samples (co-expression) were analysed with DAVID.
Proteomics Co-expression
Functional annotation Number of proteins Fold enrichment Number of proteins Fold enrichment
Acetylation 48 5.0 21 2.0
Phosphorylation 48 1.8 Not enriched Not enriched
Cytosol 37 2.9 Not enriched Not enriched
Mitochondrion 30 4.9 38 5.7
Figure 2 Biological processes affected by Ethe1 deficiency
Proteins with different abundance in Ethe1 deficient compared with
wild-type mouse liver (black bars), kidney (white bars), brain (light grey
bars) or muscle (dark grey bars) were assigned to pathways according to
the information provided by UniProt. Coex., number of proteins from the
categories listed that are co-expressed with Ethe1 in wild-type mouse
samples.
result was somewhat expected, given the mitochondrial localiz-
ation of Ethe1, the former suggests a role for Ethe1 in PTMs.
In particular, proteins regulated by acetylation were five times
enriched in our sample compared with the whole proteome. In-
terestingly, similar results were obtained by clustering the genes
from co-expression analysis, confirming a close functional con-
nection between Ethe1 and PTMs.
Next, we sorted all proteins into biological processes accord-
ing to the information available in the Uniprot database (www.
uniprot.org) (Figure 2). Among proteins significantly changed,
only four were enzymes directly related to sulfur metabol-
ism, namely sulfite oxidase, glycine N-methyltransferase, 3-
mercaptopyruvate sulfurtransferase and a sulfotransferase. The
largest group of proteins, including 17 enzymes from the proteo-
mic experiment and 14 from co-expression analysis, catalyse re-
actions during the post-translational processing of proteins, some
taking part in general protein handling such as folding (several
chaperones, peptidyl-prolyl cis–trans isomerase) and degradation
(proteasome subunits, proteases), other being involved in specific
PTMs (ubiquitinylation, glycosylation, methylation, sialylation,
glutathionylation and acylation).
In brain and muscle, we found evidence for an influence on
proteins related to cell structure. Actin and myosin were iden-
tified in four spots with a changed volume in Ethe1-less mouse
muscle. In addition, several proteins that take part in the organiz-
ation of the cytoskeleton, such as chaperones [T-complex protein
1andHSPβ1 (heat-shock protein β1)], LIM domain containing
or binding proteins, a septin and myotilin, were affected, sug-
gesting that alterations of the cytoskeleton may be involved in
the pathogenesis of EE.
Our analysis shows that Ethe1 has a key role in energy meta-
bolism affecting central processes as different as the glycolysis,
the TCA (tricarboxylic acid) cycle and the OXPHOS system.
Interestingly, the correlation to lipid metabolism was most pro-
nounced. Six enzymes, all catalysing catabolic reactions, were
affected by Ethe1 deficiency. Co-expression analysis resulted in
19 genes related to lipid metabolism, including 14 enzymes of
FA (fatty acid) or BCAA oxidation, two proteins involved in the
regulation of lipid catabolism, two in FA binding and transport
and only one enzyme of FA synthesis (Supplementary Table S2).
These findings prompted us to investigate further the potential
role of Ethe1 in the oxidative catabolism of FAs and BCAAs.
Effect of Ethe1 deficiency on FA/BCAA catabolic
enzymes
The BCAAs valine, leucine and isoleucine are oxidized in the
mitochondria, and some of the reactions overlap with FA β
oxidation (Figure 3)[15]. Enzymes catalysing these common
steps were most clearly correlated to Ethe1. As an example, the
tissue distribution of Ethe1, SCAD and the committed step in
BCAA oxidation, BCKDH (branched-chain keto acid dehydro-
genase), on expression level is shown in Figure 4. Four enzymes
of FA/BCAA oxidation, SCAD, ETF, enoyl-CoA hydratase and
3-ketoacyl-CoA thiolase, are co-expressed with Ethe1 and also
present in spots with an increased volume (1.6- to 2.6-fold) in
Ethe1-deficient mice (Figure 3). Separation on the basis of the
pI (isoelectric point) detects not only differences in total protein
abundance but also some PTMs that modify the charge of the
protein. We therefore tested whether these enzymes were actu-
ally up-regulated by measuring transcript expression and protein
amount (Figure 5). No differences were detected in either ex-
pression level or total abundance of SCAD, ETF or enoyl-CoA
hydratase indicating an influence on PTMs. In contrast, BCKDH
subunits E2 and E3, but not E1, were significantly less expressed
in the Ethe1-less mice. Similar to PDH (pyruvate dehydrogenase)
and KGDH (3-oxoglutarate dehydrogenase), BCKDH is a large
protein complex consisting of three subunits present in multiple
copies, subunit E3 being common to all three dehydrogenases.
However, the expression of PDH and KGDH E2 subunits was
..........................................................................................................................................................................................................................................................................................................................................................................
c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
579
T. M. Hildebrandt and others
Figure 3 Correlation of Ethe1 and FA/BCAA
The enzymes catalysing common reactions in BCAA and FA oxidative metabolism are affected by Ethe1 deficiency (arrows
mark increased abundance in Ethe1-deficient mouse liver, kidney, brain or muscle) and also co-expressed with Ethe1
(asterisks). Valine oxidation is shown in solid lines, leucine oxidation in dashed lines and isoleucine oxidation in dotted
lines. BCKDH, branched-chain α-keto acid dehydrogenase; SCAD, short/branched-chain acyl-CoA dehydrogenase; ETF,
electron transfer flavoprotein.
Figure 4 Tissue distribution of Ethe1 and BCAA catabolic
enzymes
Similar expression pattern of BCKDH (E1 α-subunit), SCAD and ETHE1:
gene expression in wild-type mouse skeletal muscle, brain, liver and
kidney was analysed using Genevestigator.
unaffected (results not shown). Notably, these findings clarify
the original clinical description of the disease that was thought to
be a defect in the β-oxidation pathway.
Effect of Ethe1 deficiency on PTMs
GAPDH and actin are both known to be regulated by diverse
forms of PTMs. Among all proteins affected by Ethe1 deficiency,
these two have the highest number of annotations in Uniprot,
with GAPDH being subject to ADP-ribosylation, acetylation,
methylation, phosphorylation, S-nitrosylation and ubiquitin con-
jugation, and actin being annotated for acetylation, methylation,
nitration, oxidation, phosphorylation and ubiquitin conjugation
(Supplementary Table S1). In addition, both proteins are reg-
ulated by different forms of cysteine S-modifications such as
glutathionylation, palmitoylation and most interestingly sulfhy-
dration, which is not yet included in the annotations [16]. The
IEF/SDS results indicate an influence of elevated sulfide and per-
sulfide concentrations on these PTMs. GAPDH was present in
a 2.1-fold larger spot in Ethe1-deficient mouse brain that was
located at a more acidic pH than the calculated pI of the protein.
Such a shift can be caused by different modifications that either
introduce an additional negative charge, such as phosphorylation,
or mask a positive charge, as in lysine acetylation. A pI shift was
also detectable for αactin in muscle with a 1.5-fold decrease in
spot 64 and a matching increase in spot 65 at a slightly lower
pH (Figure 1). Expression levels of GAPDH and actin were con-
sistently elevated in Ethe1-deficient mice, whereas an increase in
total protein abundance was only detectable for muscle GAPDH
(Figure 6).
DISCUSSION
The pathogenic mechanism of EE is related to the accumulation
of high levels of hydrogen sulfide in tissues and body fluids of
the patients and of the animal model of the disease. While the
effects of sulfide on COX have been thoroughly investigated, and
shown to be because of an increased degradation of COX subunits
[11], the additional systemic consequences are not completely
understood.
Sulfide in protein regulation
Our results suggest that Ethe1 and the sulfide oxidation pathway
have a major role in post-translational protein modifications. Pro-
teins that are regulated by such modifications were enriched in
both lists of proteins, those affected by Ethe1 deficiency as well as
those co-expressed with Ethe1. The data sets also contained many
enzymes involved in the catalysis of reactions during the post-
translational processing of proteins. Two examples of changed
PTM patterns were directly detectable on our 2D gels via acidic
spot shifts (actin and septin).
Apart from direct inhibition, cysteine S-sulfhydration is the
only known mechanism of enzyme regulation by hydrogen
sulfide. Cysteine thiol groups can be converted to hydropersulf-
ides (-SSH), which mostly leads to increased protein activity
[16]. Since Ethe1 oxidizes persulfide groups, it could play a role
as a negative regulator of S-sulfhydration. The activity as well
as stability of enzymes can be regulated by an interaction of
..........................................................................................................................................................................................................................................................................................................................................................................
580 c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
Proteome changes in Ethe1 deficiency
Figure 5 Effect of Ethe1 deficiency on the expression and protein content of enzymes involved in BCAA and FA oxidation
in mouse liver
(A) Gene expression was analysed by QPCR and expression in Ethe/samples (grey bars) is shown in relation to the
corresponding wild-type samples (black bars). (B) Protein abundance was estimated by Western blot. Numbers indicate the
relative densities of the bands (percentage of wild-type). E1, E2, E3, subunits of the BCKDH; SCAD, short/branched-chain
acyl-CoA dehydrogenase; ETF, electron transfer flavoprotein; ECH, enoyl-CoA hydratase; SDH-A, subunit a of succinate
dehydrogenase complex. *Significantly different from wild-type (P<0.05).
Figure 6 Effect of Ethe1 deficiency on the expression and protein content of GAPDH and actin
(A,C) Gene expression was analysed by QPCR and expression in Ethe1/samples (grey bars) is shown in relation to the
corresponding wild-type samples (black bars). (B,D) Protein abundance was estimated by Western blot. Numbers indicate
the relative densities of the bands (percentage of wild-type). (A,B) mouse liver, (C,D) mouse skeletal muscle. GAPDH,
glyceraldehyde-3-phosphate dehydrogenase; ACTA1, actin α1; SDH-A, subunit a of succinate dehydrogenase complex.
*Significantly different from wild-type (P<0.05).
different cysteine S-modifications such as S-nitrosylation, S-
acylation and S-glutathionylation. We found that acyl-protein
thioesterase, which hydrolyses regulatory FAs bound to cysteine
thiols, as well as several GSTs (glutathione transferases), which
remove cysteine S-glutathionylations, were affected by the
knock-out of Ethe1. Thus, our results indicate a general disturb-
ance in the modification of regulatory cysteines, as a consequence
of a block in sulfide and persulfide oxidation.
GAPDH is a well-studied example of enzyme regulation by
different forms of cysteine S-modifications. It is inactivated by
binding of GSH, NO (nitric oxide) or palmitic acid to specific
cysteine residues, whereas the activity is 7-fold increased after
sulfhydration of Cys150 [1619]. The acidic shift of GAPDH on
our 2D gels indicates differences in the PTM pattern. GAPDH
expression was increased in liver and muscle of Ethe1-less mice
with no concomitant increase in total protein abundance in liver.
While in muscle glycolysis is probably up-regulated by the
block in mitochondrial respiration, dysregulation might lead to
decreased stability of GAPDH in the liver. Similar discrepan-
cies between expression level, total protein abundance and 2D
spot size, were also detected for actin and several enzymes of
BCAA/FA oxidation, which will be discussed in subsequent para-
graphs.
Sulfide in BCAA/FA catabolism
The metabolite profile in blood and urine of EE patients and
Ethe1-deficient mice, characterized by the accumulation of ethyl-
malonic acid and C4–C5 acylcarnitines, indicates major disturb-
ances in the oxidation of BCAAs and FAs. A similar profile is
indeed also present in defects of acyl-CoA dehydrogenases such
as SCAD, isobutyryl-CoA dehydrogenase and 2-methylbutyryl-
CoA dehydrogenase [2022]. Thus a simple and elegant explan-
ation for the effect of Ethe1 deficiency on lipid metabolism is the
inhibition of these enzymes by accumulating sulfide or persulf-
ides. SCAD is directly inhibited by sulfide [1] and, in addition,
CoA persulfide might play a role, as a powerful inhibitor of acyl-
CoA dehydrogenases [23,24].
The results presented here reveal an additional level of interac-
tion between Ethe1 and the metabolism of FAs and BCAAs that
may explain the original clinical description of the disease as a β-
oxidation defect. Lipid catabolism is most clearly co-expressed
with Ethe1 and several related protein abundances change as a
..........................................................................................................................................................................................................................................................................................................................................................................
c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
581
T. M. Hildebrandt and others
result of the knock-out of this gene. Interestingly, the prevalence
of Ethe1 in liver and muscle parallels the tissue distribution of
FA/BCAA oxidizing enzymes [25]. In addition, there is evidence
for an influence on the regulatory level. Gene expression for two
of the three subunits of the BCKDH complex was decreased in
Ethe1/liver. As the flux-generating step in the catabolism of
BCAAs BCKDH is tightly regulated [26]. In the knockout mice
either sulfide or, more likely, the accumulating reaction products
of BCKDH down-regulate E2 and E3 subunits. The expression
of enzymes catalysing subsequent steps in FA/BCAA oxidation
was unchanged so that in this case an effect of sulfide or other
metabolites on transcription factors can be excluded. Also sulfide
does not interfere with the stability of ETF subunits or SCAD as
the block in sulfide oxidation had no influence on total protein
abundance. Since direct enzyme inhibition would not be visible
on the gels, the differences detected in the 2D proteome are most
probably caused by an altered pattern of PTMs. Mitochondrial FA
βoxidation has been shown to be regulated by lysine acetylation
[27]. Also the enzymes catalysing common steps in FA/BCAA
oxidation are subject to different kinds of cysteine S modific-
ations [28,29]. Accumulating sulfide or persulfide probably in-
terferes with this regulatory system. However, the mechanism
of this interference will have to be further elucidated in order
to fully understand the physiological role of the SDO in lipid
metabolism.
A general connection between sulfide and lipid metabolism
has previously been reported. Sulfide concentrations were de-
creased in the plasma of overweight compared with lean men and
low sulfide levels are associated with the development of insulin
resistance in Type 2 diabetes [30]. Baiges et al. [31] found that
Ethe1 and sulfide quinone oxidoreductase, which catalyses the
first step in the mitochondrial sulfide oxidation pathway, were
decreased by more than 50 % in rats fed with a high-fat diet.
Thus there seems to be a reciprocal influence of FA and sulfide
metabolism with a block of lipid catabolism by high sulfide con-
centrations and a down-regulation of sulfide metabolism by high
concentrations of FAs.
Sulfide in cell structure
EE patients and Ethe1-deficient mice show severe defects in mo-
tor activity and rapidly progressive encephalopathy, both of which
are not fully understood yet. Our results show that the increase in
sulfide and persulfide concentrations elicits changes in structural
proteins as well as in enzymes involved in the organization of the
cytoskeleton in brain and muscle, indicating an organ-specific
role similar to the isolated COX deficiency, which is also preval-
ent in these tissues. Accordingly, co-expression analysis provided
no hints for a general correlation of genes associated with the cell
structure and Ethe1. The reasons for this tissue-specificity are
currently unknown. However, it should be mentioned that neither
brain nor muscle restricted Ethe1 knockout mice, show a
clinical phenotype, in spite of tissue-specific COX deficiency,
suggesting that the pathogenesis of the disease relies on the ab-
sence of Ethe1 in other tissues, particularly in the colon [11].
In fact, the combined administration of NAC (N-acetylcysteine),
a glutathione precursor and metronidazole, a bactericidal agent
against the anaerobic bacteria of the colon, ameliorates the clin-
ical conditions of both mice and patients [32]. Di Meo et al.
recently demonstrated that a gene therapy approach based on
adeno-associated viral vectors specifically targeting the liver in
the very same mouse model used in this study is highly effect-
ive in prolonging the lifespan, likely by scavenging most of the
circulating sulfide [33].
Our results indicate a role of PTMs in the regulation of cyto-
skeleton dynamics by sulfide. An intermediate filament protein
and actin were present in smaller spots in Ethe1-less mouse liver,
but no changes were detected in total actin. Similarly, the actin
content of knockout mouse muscle tissue was unchanged al-
though gene expression was increased, and the IEF/SDS results
show a higher ratio of modified α-actin. Mustafa et al. identified
actin as one of the three most abundant sulfhydrated proteins in
sulfide treated mouse liver and estimated that about 12% of the
actin molecules are sulfhydrated under basic physiological con-
ditions [16]. Sulfhydration enhances actin polymerization in vitro
and incubation with 100 μM NaHS leads to rearrangements of the
actin cytoskeleton in HEK-293 (human embryonic kidney 293).
Sulfhydration itself would not be visible on IEF/SDS gels as the
charge of the protein is not changed. However, there could be
direct or indirect effects on other forms of modifications such as
phosphorylation or acetylation that possibly influence its stabil-
ity. Sulfide has been shown to induce a reorganization of the
actin cytoskeleton in human vascular endothelial cells via
the small GTPase Rac1 [34]. Interestingly, we also found a pH
shift of the cytoskeletal GTPase Septin 7 in the brain of Ethe1-
deficient mice (Figure 1,Tab l e 1 , spots 43 and 53) indicating
again dysregulation. Additional hints confirming that accumulat-
ing sulfide interferes with the organization of the cell structure
are changes in abundance of structure specific chaperones and
several regulatory cytoskeletal elements. Therefore, the proteo-
mic results show that sulfide signalling must be considered an
important factor in the control of cytoskeleton dynamics.
Additional aspects
Surprisingly, effects of Ethe1 deficiency on sulfur metabolic en-
zymes were limited. Sulfite oxidase and β-mercaptopyruvate
sulfurtransferase were both detected in spots with a decreased
volume. A down-regulation of these enzymes would indeed make
sense since sulfite concentrations are decreased in the knockout
mice and the sulfurtransferase produces additional sulfide. It has
been suggested that probably alternative pathways for sulfide de-
toxification exist at least in liver or kidney, which are currently
unknown [1]. However, there is no hint so far about a possible
involvement of one of the enzymes found in increased spots in
these tissues, glycine-N-methyltransferase or sulfotransferase in
this process.
H2S acts as a cytoprotective agent by activating Nrf2 (nuclear
factor-erythroid 2 p45 subunit-related factor 2), which induces the
expression of several antioxidant enzymes [6]. In addition, the
sulfide molecule itself has antioxidant properties. Our results
show that increased sulfide concentrations due to disruption of
..........................................................................................................................................................................................................................................................................................................................................................................
582 c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
Proteome changes in Ethe1 deficiency
the sulfide oxidation pathway have several effects on the antiox-
idant system (e.g. glutathione peroxidase, peroxiredoxin), thus
confirming the findings of Palmfeldt et al., who detected alter-
ations in the redox mitochondrial proteome from cultured skin
fibroblasts of six EE patients [35].
Conclusions
Our results provide evidence for a crucial role of the mitochon-
drial sulfide oxidation pathway in sulfide signalling. Increased
sulfide concentrations, due to a block in this pathway at the level
of the SDO, lead to major disturbances in post-translational pro-
tein modifications, which can explain the pleiotropic devastating
effects of EE. This study confirms the involvement of sulfide in
redox regulation and cytoskeleton dynamics. In addition, we sug-
gest that sulfide signalling also specifically regulates mitochon-
drial BCAA/FA catabolism and thus takes part in the coordina-
tion of the energy source used by the organism. The mechanism
of this regulatory function is most likely based on cysteine S-
modifications. Increased sulfide concentrations promote cysteine
S-sulfhydrations and thereby shift the balance between different
forms of PTMs. In addition, ETHE1 as an SDO could take part in
the removal of sulfhydrations by oxidizing the cysteine-derived
persulfide group.
AUTHOR CONTRIBUTION
Tatjana Hildebrandt designed the study, performed the proteomics
experiments and co-expression analysis, analysed and interpreted
the data, and wrote the paper. Ivano Di Meo performed the qPCR
and Western blot analysis. Carlo Viscomi provided the mouse tis-
sues, analysed and interpreted the data, and wrote the paper.
Massimo Zeviani and Hans-Peter Braun supervised the work and
revised the paper.
FUNDING
We acknowledge support by Deutsche Forschungsgemeinschaft
and Open Access Publishing Fund of Leibniz Universit¨
at Hannover.
This work was supported by the Deutsche Forschungsgemeinschaft
[grant number HI 1471/1-1 (to T.H.)]; Telethon grant [grant numbers
GPP10005 and AFM15927 (to M.Z.)]; and the Pierfranco and Luisa
Mariani Foundation Italy.
REFERENCES
1 Tiranti, V., Viscomi, C., Hildebrandt, T., Di Meo, I., Mineri, R.,
Tiveron, C., D Levitt, M., Prelle, A., Fagiolari, G., Rimoldi, M. et al.
(2009) Loss of ETHE1, a mitochondrial dioxygenase, causes fatal
sulfide toxicity in ethylmalonic encephalopathy. Nat. Med. 15,
200–205
2 Hildebrandt, T. M. and Grieshaber, M. K. (2008) Three enzymatic
activities catalyze the oxidation of sulfide to thiosulfate in
mammalian and invertebrate mitochondria. FEBS J. 275,
3352–3361
3 Kabil, O. and Banerjee, R. (2010) Redox biochemistry of hydrogen
sulfide. J. Biol. Chem. 285, 21903–21907
4 Flannigan, K. L., McCoy, K. D. and Wallace, J. L. (2011) Eukaryotic
and prokaryotic contributions to colonic hydrogen sulfide
synthesis. Am. J. Physiol. Gastrointest. Liver Physiol. 301, G188
5 Li, L., Rose, P. and Moore, P. K. (2011) Hydrogen sulfide and cell
signaling. Annu. Rev. Pharmacol. Toxicol. 51, 169–187
6 Predmore, B. L., Lefer, D. J. and Gojon, G. (2012) Hydrogen sulfide
in biochemistry and medicine. Antioxid. Redox. Signal. 17,
119–140
7 Burlina, A., Zacchello, F., Dionisi-Vici, C., Bertini, E., Sabetta, G.,
Bennet, M., Hale, D., Schmidt-Sommerfeld, E. and Rinaldo, P.
(1991) New clinical phenotype of branched-chain acyl-CoA
oxidation defect. Lancet 338, 1522–1523
8 Mineri, R., Rimoldi, M., Burlina, A. B., Koskull, S., Perletti, C.,
Heese, B., Dobeln, U. von, Mereghetti, P., Di Meo, I., Invernizzi, F.
et al. (2008) Identification of new mutations in the ETHE1 gene in
a cohort of 14 patients presenting with ethylmalonic
encephalopathy. J. Med. Genet. 45, 473–478
9 Giordano, C., Viscomi, C., Or landi, M., Papoff, P., Spalice, A.,
Burlina, A., Meo, I., Tiranti, V., Leuzzi, V., d’Amati, G. et al. (2012)
Morphologic evidence of diffuse vascular damage in human and in
the experimental model of ethylmalonic encephalopathy. J. Inherit.
Metab. Dis. 35, 451–458
10 Cooper, C. E. and Brown, G. C. (2008) The inhibition of
mitochondrial cytochrome oxidase by the gases carbon monoxide,
nitric oxide, hydrogen cyanide and hydrogen sulfide: chemical
mechanism and physiological significance. J. Bioenerg. Biomembr.
40, 533–539
11 Di Meo, I., Fagiolari, G., Prelle, A., Viscomi, C., Zeviani, M. and
Tiranti, V. (2011) Chronic exposure to sulfide causes accelerated
degradation of cytochrome coxidase in ethylmalonic
encephalopathy. Antioxid. Redox. Signal. 15, 353–362
12 Klodmann, J., Senkler, M., Rode, C. and Braun, H.-P. (2011)
Defining the protein complex proteome of plant mitochondria. Plant
Physiol. 157, 587–598
13 Huang, D. W., Sherman, B. T. and Lempicki, R. A. (2008)
Systematic and integrative analysis of large gene lists using DAVID
bioinformatics resources. Nat. Protoc. 4, 44–57
14 Finocchiaro, G., Colombo, I., Garavaglia, B., Gellera, C., Valdameri,
G., Garbuglio, N. and Didonato, S. (1993) cDNA cloning and
mitochondrial import of the beta-subunit of the human
electron-transfer flavoprotein. Eur. J. Biochem. 213, 1003–1008
15 Brosnan, J. T. and Brosnan, M. E. (2006) Branched-chain amino
acids: enzyme and substrate regulation. J. Nutr. 136, 2075–2115
16 Mustafa, A. K., Gadalla, M. M., Sen, N., Kim, S., Mu, W., Gazi,
S. K., Barrow, R. K., Yang, G., Wang, R. and Snyder, S. H. (2009)
H2S Signals through protein S-sulfhydration. Sci. Signal. 2,ra72
17 Hara,M.R.,Agrawal,N.,Kim,S.F.,Cascio,M.B.,Fujimuro,M.,
Ozeki, Y., Takahashi, M., Cheah, J. H., Tankou, S. K., Hester, L. D.
et al. (2005) S-nitrosylated GAPDH initiates apoptotic cell death by
nuclear translocation following Siah1 binding. Nat. Cell Biol. 7,
665–674
18 Yang, J., Gibson, B., Snider, J., Jenkins, C. M., Han, X. and Gross,
R. W. (2005) Submicromolar concentrations of palmitoyl-CoA
specifically thioesterify cysteine 244 in glyceraldehyde-3-phosphate
dehydrogenase inhibiting enzyme activity: a novel mechanism
potentially underlying fatty acid induced insulin resistance.
Biochemistry 44, 11903–11912
19 Yap, L., Garcia, J. V., Han, D. S. and Cadenas, E. (2010) Role of
nitric oxide-mediated glutathionylation in neuronal function:
potential regulation of energy utilization. Biochem. J. 428, 85–93
..........................................................................................................................................................................................................................................................................................................................................................................
c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
583
T. M. Hildebrandt and others
20 Greger sen, N. (1998) Identification of four new mutations in the
short-chain acyl-CoA dehydrogenase (SCAD) gene in two patients:
one of the variant alleles, 511CT, is present at an unexpectedly
high frequency in the general population, as was the case for
625GA, together conferring susceptibility to ethylmalonic
aciduria. Hum. Mol. Genet. 7, 619–627
21 Koeberl, D. D., Young, S. P., Gregersen, N., Vockley, J., Smith,
W. E., Benjamin, D. K., An, Y., Weavil, S. D., Chaing, S. H., Bali, D.
et al. (2003) Rare disorders of metabolism with elevated butyryl-
and isobutyryl-carnitine detected by tandem mass spectrometry
newborn screening. Pediatr. Res. 54, 219–223
22 Sass, J. O., Ensenauer, R., R¨
oschinger, W., Reich, H., Steuerwald,
U., Schirrmacher, O., Engel, K., H¨
aberle, J., Andresen, B. S.,
M´
egarban´
e, A. et al. (2008) 2-methylbutyryl-coenzyme A
dehydrogenase deficiency: functional and molecular studies on a
defect in isoleucine catabolism. Mol. Genet. Metab. 93, 30–35
23 Kabil, O. and Banerjee, R. (2012) Characterization of patient
mutations in human persulfide dioxygenase (ETHE1) involved in
H2S catabolism. J. Biol. Chem. 287, 44561–44567
24 Shaw, L. and Engel, P. C. (1987) CoA-persulphide: a possible in vivo
inhibitor of mammalian short-chain acyl-CoA dehydrogenase.
Biochim. Biophys. Acta 919, 171–174
25 Sur yawan, A., Hawes, J. W., Harris, R. A., Shimomura, Y., Jenkins,
A. E. and Hutson, S. M. (1998) A molecular model of human
branched-chain amino acid metabolism. Am. J. Clin. Nutr. 68,
72–81
26 Har ris, R. A., Kobayashi, R., Murakami, T. and Shimomura, Y.
(2001) Regulation of branched-chain alpha-keto acid
dehydrogenase kinase expression in rat liver. J. Nutr. 131,
841S–845S
27 Hir schey, M. D., Shimazu, T., Goetzman, E., Jing, E., Schwer, B.,
Lombard, D. B., Grueter, C. A., Harris, C., Biddinger, S., Ilkayeva,
O. R. et al. (2010) SIRT3 regulates mitochondrial fatty-acid
oxidation by reversible enzyme deacetylation. Nature 464,
121–125
28 Doulias, P.-T., Tenopoulou, M., Greene, J. L., Raju, K. and
Ischiropoulos, H. (2013) Nitric oxide regulates mitochondrial fatty
acid metabolism through reversible protein S-nitrosylation. Sci.
Signal. 6,rs1
29 Kostiuk, M. A., Corvi, M. M., Keller, B. O., Plummer, G., Prescher,
J. A., Hangauer, M. J., Bertozzi, C. R., Rajaiah, G., Falck, J. R. and
Berthiaume, L. G. (2007) Identification of palmitoylated
mitochondrial proteins using a bio-orthogonal azido-palmitate
analogue. FASEB J. 22, 721–732
30 Whiteman, M., Gooding, K. M., Whatmore, J. L., Ball, C. I.,
Mawson, D., Skinner, K., Tooke, J. E. and Shore, A. C. (2010)
Adiposity is a major determinant of plasma levels of the novel
vasodilator hydrogen sulphide. Diabetologia 53,
1722–1726
31 Baiges, I., Palmfeldt, J., Blade, C., Gregersen, N. and Arola, L.
(2010) Lipogenesis is decreased by grape seed proanthocyanidins
according to liver proteomics of rats fed a high fat diet. Mol. Cell.
Proteomics 9, 1499–1513
32 Viscomi, C., Burlina, A. B., Dweikat, I., Savoiardo, M., Lamperti,
C., Hildebrandt, T., Tiranti, V. and Zeviani, M. (2010) Combined
treatment with oral metronidazole and N-acetylcysteine is effective
in ethylmalonic encephalopathy. Nat. Med. 16, 869–871
33 Di Meo, I., Auricchio, A., Lamperti, C., Bur lina, A., Viscomi, C. and
Zeviani, M. (2012) Effective AAV-mediated gene therapy in a
mouse model of ethylmalonic encephalopathy. EMBO Mol. Med. 4,
1008–1014
34 Zhang, L.-J., Tao, B.-B., Wang, M.-J., Jin, H.-M., Zhu, Y.-C. and Yang,
C.-M. (2012) PI3K p110αisoform-dependent Rho GTPase Rac1
activation mediates H2S-promoted endothelial cell migration via
actin cytoskeleton reorganization. PLoS ONE 7, e44590
35 Palmfeldt, J., Vang, S., Stenbroen, V., Pavlou, E., Baycheva, M.,
Buchal, G., Monavari, A. A., Augoustides-Savvopoulou, P., Mandel,
H. and Gregersen, N. (2011) Proteomics reveals that redox
regulation is disrupted in patients with ethylmalonic
encephalopathy. J. Proteome Res. 10, 2389–2396
Received 15 May 2013; accepted 29 May 2013
Published as Immediate Publication 26 June 2013, doi 10.1042/BSR20130051
..........................................................................................................................................................................................................................................................................................................................................................................
584 c
2013 The Author(s) This is an Open Access article distributed under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0)
which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.
... Fig. 4). Sulfide:quinone oxidoreductase (SQR), persulfide dioxygenase (PDO), and thiosulfate sulfurtransferase (TST) catalyze individual steps in the H 2 S oxidative pathway producing glutathione and other low-molecular weight persulfides that are further converted into sulfite and thiosulfate (Hildebrandt et al., 2013;Libiad et al., 2014). Again, no biologically relevant change was observed for those proteins. ...
... Both plasma and urine were stored at -80 °C until further measurement. Sulfite and thiosulfate were determined using monobromobimane derivatization and subsequent HPLC analysis (Hildebrandt et al., 2013). 15 µl of sample were mixed with 15 µl 160 mM HEPES, pH 8.0, 16 mM EDTA, 15 µl acetonitrile and 3 µl 46 mM monobromobimane in acetonitrile. ...
Preprint
Full-text available
Sulfite oxidase (SOX) deficiency is a rare inborn error of cysteine metabolism resulting in severe neurological damage. In patients, sulfite accumulates to toxic levels causing a raise in downstream products S-sulfocysteine (SSC), mediating excitotoxicity, and thiosulfate, a catabolic intermediate/product of H2S metabolism. Here, we report a full-body knock-out mouse model for SOX deficiency (SOXD) with a severely impaired phenotype. Amongst the urinary biomarkers, thiosulfate showed a 45-fold accumulation in SOXD mice representing the major excreted S-metabolite. Consistently, we found increased plasma H2S, which was derived from sulfite-induced release from persulfides as demonstrated in vitro and in vivo. Mass spectrometric analysis of total protein persulfidome identified a major loss of persulfidation in 20% of the proteome affecting enzymes in amino acids and fatty acid metabolism. Urinary amino acid profiles indicate metabolic rewiring suggesting partial reversal of the TCA cycle thus identifying a novel contribution of H2S metabolism and persulfidation in SOXD.
... BCKDH is the flux-generating step in the oxidation of branched-chain amino acids (valine, leucine and isoleucine), which occurs in the mitochondria, and has some steps overlapping with mtFAO [745]. ...
... In order to develop an animal model of NAFL, two diets were purchased from the company "Research Diets": one diet of standard composition (SD) for the control group (D12451) and one diet enriched in fat and sucrose (HF/HS) (45% fat, 20% protein, 35% carbohydrates of which 17.4% corresponded to sucrose) for the model of NAFL (D12450J). The HF/HS diet has been used in rodents to increase blood glycemia, body weight, adiposity and IR [743][744][745]. Indeed, it is thought that this diet combination closely resembles the diet that has dramatically increased obesity levels in humans over the past few decades. ...
Thesis
Hydrogen sulfide (H2S) is the third gasotransmitter described in mammals. Colourless and water-soluble, H2S is a highly effective inhibitor of mitochondrial cytochrome c oxidase when present at high concentrations. However, when present at low concentrations, H2S can act as an inorganic energetic substrate for mammalian mitochondria. To oxidize H2S, mitochondria need the sulfide oxidizing unit (SOU), a set of three specific enzymes: sulfide quinone reductase (SQR), dioxygenase (ETHE1) and thiosulfate sulfurtransferase (TST). Liver metabolism, finely regulated by hormones and nutrients, is central to energy homeostasis, and liver metabolic inflexibility is known to be associated with several metabolic diseases, such as Non-Alcoholic Fatty Liver Disease (NAFLD). The unique position of this organ makes it likely to be exposed to high levels of H2S coming from both exogenous (gastrointestinal tract) and endogenous (metabolism of sulfur-containing amino acids) sources. Recently, impaired liver H2S biosynthesis has been reported in animal models of NAFLD, and in vivo supplementation of H2S donors prevented the further escalation of the illness into steatohepatitis (NASH). Almost all studies exploring the hepatic pathological relevance of H2S are usually focused on H2S biosynthesis pathway and/or using exogenous donors. As intrahepatic H2S levels can also be controlled by its mitochondrial oxidation, the objectives of my PhD were to investigate the pathophysiological importance of this pathway in liver metabolism and in the development of NAFLD. First, we showed that, physiologically, the liver nutritional status regulates hepatic mitochondrial H2S oxidation capacity and SQR protein expression, both being downregulated by fasting while overnight refeeding abolished the fasting inhibitory effect. Adenovirus-mediated overexpression of human SQR in mouse liver clearly demonstrated that SQR is the key regulatory enzyme of mitochondrial H2S oxidation. Enhancing this pathway i) increased in vivo glucose tolerance and liver insulin signalling, ii) stimulated glucose metabolism in primary cultures of mouse hepatocytes (13C-glucose fluoxomics), and iii) decreased liver fatty acid oxidation capacity. Second, when exploring the context of NAFLD, we observed that liver mitochondrial H2S oxidation capacity was downregulated in mice fed high fat/high sucrose (HF/HS) diet (NAFL model) and surprisingly was upregulated in mice fed methionine-choline deficient (MCD) diet (NASH model). In vivo supplementation with sodium thiosulfate (STS), an H2S donor, abrogated the inhibitory effect of HF/HS diet while it had no impact in the NASH model. Additionally, STS supplementation had no effect on body weight, glucose tolerance and insulin sensitivity in the NAFL model, and on liver steatosis in both NAFL and NASH models. However, STS supplementation did have an impact in the composition of gut microbiota. Similarly to the NAFLD mouse models, in morbid obese patients with simple steatosis (NAFL), liver SQR protein expression was found decreased, while in individuals with NASH SQR expression was found increased. Altogether, these studies, which confirmed that SQR is the key rate-limiting enzyme for liver mitochondrial H2S oxidation, clearly demonstrated for the first time that this pathway is finely regulated by the nutritional status of the liver and is altered in animal models of NAFLD and obese NAFLD patients. The novel observation that increasing hepatic mitochondrial H2S oxidation capacity increases hepatic glucose metabolism opens a new door in the field of liver pathophysiology, with this pathway being a potential protective target against the development of liver insulin resistance.
... ETHE1 has sulfur dioxygenase activity and can catalyze the dioxygenation of glutathione persulfide to glutathione and sulfite to facilitate hydrogen sulfide (H 2 S) catabolism in the mitochondrial matrix [8,9]. This process is essential for mitochondrial metabolic homeostasis [10,11]. Loss of or mutations in ETHE1 leads to ethylmalonic encephalopathy in humans, characterized by brain damage, lactic acidemia, and even death [6,12,13]. ...
Article
Full-text available
Triple-negative breast cancer (TNBC) is the most fatal subtype of breast cancer; however, effective treatment strategies for TNBC are lacking. Therefore, it is important to explore the mechanism of TNBC metastasis and identify its therapeutic targets. Dysregulation of ETHE1 leads to ethylmalonic encephalopathy in humans; however, the role of ETHE1 in TNBC remains elusive. Stable cell lines with ETHE1 overexpression or knockdown were constructed to explore the biological functions of ETHE1 during TNBC progression in vitro and in vivo. Mass spectrometry was used to analyze the molecular mechanism through which ETHE1 functions in TNBC progression. ETHE1 had no impact on TNBC cell proliferation and xenograft tumor growth but promoted TNBC cell migration and invasion in vitro and lung metastasis in vivo. The effect of ETHE1 on TNBC cell migratory potential was independent of its enzymatic activity. Mechanistic investigations revealed that ETHE1 interacted with eIF2α and enhanced its phosphorylation by promoting the interaction between eIF2α and GCN2. Phosphorylated eIF2α in turn upregulated the expression of ATF4, a transcriptional activator of genes involved in cell migration and tumor metastasis. Notably, inhibition of eIF2α phosphorylation through ISRIB or ATF4 knockdown partially abolished the tumor-promoting effect of ETHE1 overexpression. ETHE1 has a functional and mechanistic role in TNBC metastasis and offers a new therapeutic strategy for targeting ETHE1-propelled TNBC using ISRIB.
... Here, we identified the expression of SMAD4 in 24 h-differentiated cells (Supplementary Table 1) and found that LOX was differentially abundant at the protein level in these induced cells (Supplementary Table 2). The remaining identified 10 Stem Cells International proteins (ATP1A1, ETHE1, PACSIN2, and BUB3) may play important roles in mitochondrial metabolism, lipid metabolism, vesicular traffic, cell migration, and cell cycle regulation [82][83][84][85]. ...
Article
Full-text available
Background. Obesity is characterized as a disease that directly affects the whole-body metabolism and is associated with excess fat mass and several related comorbidities. Dynamics of adipocyte hypertrophy and hyperplasia play an important role in health and disease, especially in obesity. Human adipose-derived stem cells (hASC) represent an important source for understanding the entire adipogenic differentiation process. However, little is known about the triggering step of adipogenesis in hASC. Here, we performed a proteogenomic approach for understanding the protein abundance alterations during the initiation of the adipogenic differentiation process. Methods. hASC were isolated from adipose tissue of three donors and were then characterized and expanded. Cells were cultured for 24 hours in adipogenic differentiation medium followed by protein extraction. We used shotgun proteomics to compare the proteomic profile of 24 h-adipogenic, differentiated, and undifferentiated hASC. We also used our previous next-generation sequencing data (RNA-seq) of the total and polysomal mRNA fractions of hASC to study posttranscriptional regulation during the initial steps of adipogenesis. Results. We identified 3420 proteins out of 48,336 peptides, of which 92 proteins were exclusively identified in undifferentiated hASC and 53 proteins were exclusively found in 24 h-differentiated cells. Using a stringent criterion, we identified 33 differentially abundant proteins when comparing 24 h-differentiated and undifferentiated hASC (14 upregulated and 19 downregulated, respectively). Among the upregulated proteins, we shortlisted several adipogenesis-related proteins. A combined analysis of the proteome and the transcriptome allowed the identification of positive correlation coefficients between proteins and mRNAs. Conclusions. These results demonstrate a specific proteome profile related to adipogenesis at the beginning (24 hours) of the differentiation process in hASC, which advances the understanding of human adipogenesis and obesity. Adipogenic differentiation is finely regulated at the transcriptional, posttranscriptional, and posttranslational levels. 1. Introduction Obesity is the main risk factor for several diseases such as hypertension, diabetes, insulin resistance, dyslipidemia, heart diseases, and some cancer types; it is also related to increased overall mortality [1]. Human adipose tissue is a primary regulator of metabolism and energy balance and is composed of several cell types, including adipocytes, preadipocytes, mesenchymal stem cells, fibroblasts, macrophages, and endothelial and blood cells [2, 3]. The pathophysiology of obesity is directly linked to increased adipocyte hypertrophy, adipocyte hyperplasia, or both in adipose tissue. Therefore, adipocyte size and the counterbalance between adipocyte loss and their formation through differentiation of preadipocytes are critical for human health. Therefore, adipogenesis plays a key role in the development of obesity, being the hormonal and nutritional status critical for the maintenance and development of body fat tissue [4]. Over the past two decades, adipogenesis has been extensively studied at the cellular and molecular levels, both in vitro and in vivo [4–8]. Adipose tissue is an abundant source of human adipose-derived mesenchymal stem cells (hASC), which have the ability to differentiate into adipocytes, osteoblasts, and chondrocytes [2, 9]. The adipogenic differentiation process is separated into two stages: the commitment of hASC into preadipocytes and the terminal differentiation of preadipocytes into mature adipocytes [10, 11]. Both phases are highly regulated at the transcriptional, posttranscriptional, translational, and posttranslational levels [12–14]. However, few studies have focused on molecular regulation at the earliest steps in hASC differentiation into preadipocytes [15, 16]. Thus, understanding how hASC become committed to preadipocytes and adipocyte lineages is a principal concern in the study of adipose tissue homeostasis and development. Different techniques have been used to understand molecular changes at a global level (e.g., transcriptomics and proteomics) to better understand the initial stage of adipogenesis [17–19]. In a previous study, we used ribosome profiling (Ribo-seq) to identify the translational regulation of mRNAs after 72 hours of hASC adipogenesis. We observed a significant reduction in cell size and migration, in addition to a reduction in protein synthesis after induction [20]. Similarly, we used total fraction and polysome profiling followed by RNA-seq to show the downregulation of cell cycle- and proliferation-related genes after 24 hours of induction [21]. Using these distinct high-throughput sequencing techniques of different cell fractions (i.e., bulk RNA-seq and polysomal and ribosome-associated mRNAs) may reflect a better understanding of the posttranscriptional and translational regulation of RNAs during cell differentiation, making possible a further comparison of gene expression at the RNA level with proteomics data (i.e., protein level). However, as far as we know, only one study discloses proteomic results from the early stages of human adipogenesis, focusing only on the expression of transcription factors [22, 23]. With this as a motivation, we performed a proteogenomic analysis of the early steps of adipogenesis (24 hours) in hASC. Our findings include the identification of proteins regulated at the beginning of the adipogenic differentiation process in hASC. This gene expression regulation is exerted at multiple levels, showing that human adipogenesis is a complex process and that combined strategies must be used for a better understanding. 2. Material and Methods 2.1. Ethical Statement Tissue collection and cell isolation were performed after donors provided informed consent in accordance with guidelines for research involving human subjects and with the approval of the Ethics Committee of the Oswaldo Cruz Foundation, Brazil (approval number CAAE: 48374715.8.0000.5248). Human adipose-derived stem cells (hASC) were obtained from adipose tissue from lipoaspirate samples from three female donors (biological replicates) with a mean (±SD) age of and BMI of (Table 1). We randomly selected the donors since no differences in morphology, immunophenotype characteristics, proliferative rates, and differentiation potential between hASC isolated from young and old subjects were demonstrated [24]. Subjects Donor 1 Donor 2 Donor 3 Age 46 20 48 Gender F F F F Weight (kg) 74.5 75 90 Height (cm) 166 174 175 BMI 27.04 24.77 29.39
... BCKDH is the flux-generating step in the oxidation of branched-chain amino acids (valine, leucine and isoleucine), which occurs in the mitochondria, and has some steps overlapping with mitochondrial FA ß-oxidation. 99 Although H 2 S production and oxidation are both important to maintain steady-state levels of H 2 S, the way by which these pathways control H 2 S actions in a cell may vary. H 2 S production impacts on H 2 S ability to modify cysteine residues and regulate protein expression/activity, whereas H 2 S oxidation participates in mitochondrial respiration, controlling oxygen levels as well as the production of reactive sulphur species, which have lately gained recognition as powerful cell signalling mediators. ...
Article
Full-text available
Background For a long time, hydrogen sulfide (H2S) was considered only as a toxic gas, inhibiting mitochondrial respiration at the level of cytochrome c oxidase, and an environmental pollutant. Nowadays, H2S is recognized as the third mammalian gasotransmitter, playing an important role in inflammation, septic shock, ischemia reperfusion events, cardiovascular disease, and more recently in liver physiology and chronic liver diseases such as Non-Alcoholic Fatty Liver Disease (NAFLD). Methods This narrative review is based on literature search using PubMed. Results From a bioenergetic perspective, H2S is a very unique molecule, serving as a mitochondrial poison at high concentrations or as an inorganic mitochondrial substrate at low concentrations. By using transgenic animal models to specifically modulate liver H2S biosynthesis or exogenous compounds that release H2S, several studies demonstrated that H2S is a key player in liver glucose and lipid metabolism. Liver H2S content and biosynthesis were also altered in NAFLD animal models with the in vivo administration of H2S-releasing molecules preventing the further escalation into Non-Alcoholic-Steatohepatitis. Liver steady-state levels of H2S, and hence its cell signalling properties, are controlled by a tight balance between its biosynthesis, mainly through the transsulfuration pathway, and its mitochondrial oxidation via the sulfide oxidizing unit. However, studies investigating mitochondrial H2S oxidation in liver dysfunction still remain scarce. Conclusions Since H2S emerges as a key regulator of liver metabolism and metabolic flexibility, further understanding the physiological relevance of mitochondrial H2S oxidation in liver energy homeostasis and its potential implication in chronic liver diseases are of great interest.
... In this disease, ETHE1, a gene encoding sulfur dioxygenase, which metabolizes H 2 S in collaboration with SQR, is deficient, and H 2 S levels are increased to suppress cytochrome c oxidase [93]. High levels of H 2 S and persulfides also suppress acyl-protein thioesterase in the mouse model of this disease [94,95]. The brain is very sensitive to oxygen deprivation, i.e., hypoxia. ...
Article
Full-text available
Since the first description of hydrogen sulfide (H2S) as a toxic gas in 1713 by Bernardino Ramazzini, most studies on H2S have concentrated on its toxicity. In 1989, Warenycia et al. demonstrated the existence of endogenous H2S in the brain, suggesting that H2S may have physiological roles. In 1996, we demonstrated that hydrogen sulfide (H2S) is a potential signaling molecule, which can be produced by cystathionine β-synthase (CBS) to modify neurotransmission in the brain. Subsequently, we showed that H2S relaxes vascular smooth muscle in synergy with nitric oxide (NO) and that cystathionine γ-lyase (CSE) is another producing enzyme. This study also opened up a new research area of a crosstalk between H2S and NO. The cytoprotective effect, anti-inflammatory activity, energy formation, and oxygen sensing by H2S have been subsequently demonstrated. Two additional pathways for the production of H2S with 3-mercaptopyruvate sulfurtransferase (3MST) from l- and d-cysteine have been identified. We also discovered that hydrogen polysulfides (H2Sn, n ≥ 2) are potential signaling molecules produced by 3MST. H2Sn regulate the activity of ion channels and enzymes, as well as even the growth of tumors. S-Sulfuration (S-sulfhydration) proposed by Snyder is the main mechanism for H2S/H2Sn underlying regulation of the activity of target proteins. This mini review focuses on the key findings on H2S/H2Sn signaling during the first 25 years.
Article
Full-text available
Hydrogen sulfide (H2S) and polysulfides (H2Sn, n ≥ 2) produced by enzymes play a role as signalling molecules regulating neurotransmission, vascular tone, cytoprotection, inflammation, oxygen sensing, and energy formation. H2Sn, which have additional sulfur atoms to H2S, and other S-sulfurated molecules such as cysteine persulfide and S-sulfurated cysteine residues of proteins, are produced by enzymes including 3-mercaptopyruvate sulfurtransferase (3MST). H2Sn are also generated by the chemical interaction of H2S with NO, or to a lesser extent with H2O2. S-sulfuration (S-sulfhydration) has been proposed as a mode of action of H2S and H2Sn to regulate the activity of target molecules. Recently, we found that H2S/H2S2 regulate the release of neurotransmitters, such as GABA, glutamate, and D-serine, a co-agonist of N-methyl-D-aspartate (NMDA) receptors. H2S facilitates the induction of hippocampal long-term potentiation, a synaptic model of memory formation, by enhancing the activity of NMDA receptors, while H2S2 achieves this by activating transient receptor potential ankyrin 1 (TRPA1) channels in astrocytes, potentially leading to the activation of nearby neurons. The recent findings show the other aspects of TRPA1 channels—that is, the regulation of the levels of sulfur-containing molecules and their metabolizing enzymes. Disturbance of the signalling by H2S/H2Sn has been demonstrated to be involved in various diseases, including cognitive and psychiatric diseases. The physiological and pathophysiological roles of these molecules will be discussed.
Article
Significance: Hydrogen sulfide (H2S), the third member of the gasotransmitter family, has a broad spectrum of biological activities, including antioxidant and cytoprotective actions, as well as vasodilatory, anti-inflammatory and antifibrotic effects. New, significant aspects of H2S biology in the kidney continue to emerge, underscoring the importance of this signaling molecule in kidney homeostasis, function, and disease. Recent Advances: H2S signals via three main mechanisms, by maintaining redox balance through its antioxidant actions, by post-translational modifications of cellular proteins (S-sulfhydration), and by binding to protein metal centers. Important renal functions such as glomerular filtration, renin release, or sodium reabsorption have been shown to be regulated by H2S, using either exogenous donors or by the endogenous-producing systems. Critical Issues: Lower H2S levels are observed in many renal pathologies, including renal ischemia-reperfusion injury and obstructive, diabetic, or hypertensive nephropathy. Unraveling the molecular targets through which H2S exerts its beneficial effects would be of great importance not only for understanding basic renal physiology, but also for identifying new pharmacological interventions for renal disease. Future Directions: Additional studies are needed to better understand the role of H2S in the kidney. Mapping the expression pattern of H2S-producing and -degrading enzymes in renal cells and generation of cell-specific knockout mice based on this information will be invaluable in the effort to unravel additional roles for H2S in kidney (patho)physiology. With this knowledge, novel targeted more effective therapeutic strategies for renal disease can be designed.
Article
Full-text available
Impaired hepatic glucose and lipid metabolism are hallmarks of type 2 diabetes. Increased sulfide production or sulfide donor compounds may beneficially regulate hepatic metabolism. Disposal of sulfide through the sulfide oxidation pathway (SOP) is critical for maintaining sulfide within a safe physiological range. We show that mice lacking the liver- enriched mitochondrial SOP enzyme thiosulfate sulfurtransferase (Tst−/− mice) exhibit high circulating sulfide, increased gluconeogenesis, hypertriglyceridemia, and fatty liver. Unexpectedly, hepatic sulfide levels are normal in Tst−/− mice because of exaggerated induction of sulfide disposal, with associated suppression of global protein persulfidation and nuclear respiratory factor 2 target protein levels. Hepatic proteomic and persulfidomic profiles converge on gluconeogenesis and lipid metabolism, revealing a selective deficit in medium-chain fatty acid oxidation in Tst−/− mice. We reveal a critical role of TST in hepatic metabolism that has implications for sulfide donor strategies in the context of metabolic disease.
Article
Background Sulfide is a well-known toxicant widely distributed in the culture environment. As a representative burrowing benthic bivalve, the razor clam Sinonovacula constricta is highly sulfide tolerant. Mitochondrial sulfide oxidation is an important way for sulfide detoxification, where sulfur dioxygenase (SDO) is the second key enzyme.Objective To investigate the mechanism of sulfide tolerance in S. constricta, the molecular characterization of its SDO (designated as ScSDO) was studied.Methods The cDNA sequence of ScSDO was cloned by RACE technique. The response of ScSDO in gills and livers of S. constricta was investigated during sulfide exposure (50, 150, and 300 μM sulfide) for 0, 3, 6, 12, 24, 48, 72, and 96 h by qRT-PCR. Moreover, the temporal expression of ScSDO protein in S. constricta gills after exposure to 150 μM sulfide was detected by Western blot. The subcellular location of ScSDO was identified by TargetP 1.1 prediction and Western Blot analysis.ResultsThe full-length cDNA of ScSDO was 2914 bp, encoding a protein of 304 amino acids. The deduced ScSDO protein was highly conserved, containing the signature HXHXDH motif of the metallo-β-lactamase superfamily and two metal-binding sites, of which metal-binding site I is known to be the catalytically active center. Subcellular localization confirmed that ScSDO was located only in the mitochondria. Responding to the sulfide exposure, distinct time-dependent increases in ScSDO expression were detected at both mRNA and protein levels. Moreover, the gills exhibited a higher ScSDO expression level than the livers.Conclusions All of our results suggest that ScSDO plays an important role in mitochondrial sulfide oxidation during sulfide stress, making S. constricta highly sulfide tolerant. In addition, as a respiratory tissue, the gills play a more critical role in sulfide detoxification.
Article
Full-text available
DAVID bioinformatics resources consists of an integrated biological knowledgebase and analytic tools aimed at systematically extracting biological meaning from large gene/protein lists. This protocol explains how to use DAVID, a high-throughput and integrated data-mining environment, to analyze gene lists derived from high-throughput genomic experiments. The procedure first requires uploading a gene list containing any number of common gene identifiers followed by analysis using one or more text and pathway-mining tools such as gene functional classification, functional annotation chart or clustering and functional annotation table. By following this protocol, investigators are able to gain an in-depth understanding of the biological themes in lists of genes that are enriched in genome-scale studies.
Article
Full-text available
Cysteine S-nitrosylation is a posttranslational modification by which nitric oxide regulates protein function and signaling. Studies of individual proteins have elucidated specific functional roles for S-nitrosylation, but knowledge of the extent of endogenous S-nitrosylation, the sites that are nitrosylated, and the regulatory consequences of S-nitrosylation remains limited. We used mass spectrometry-based methodologies to identify 1011 S-nitrosocysteine residues in 647 proteins in various mouse tissues. We uncovered selective S-nitrosylation of enzymes participating in glycolysis, gluconeogenesis, tricarboxylic acid cycle, and oxidative phosphorylation, indicating that this posttranslational modification may regulate metabolism and mitochondrial bioenergetics. S-nitrosylation of the liver enzyme VLCAD [very long chain acyl-coenzyme A (CoA) dehydrogenase] at Cys(238), which was absent in mice lacking endothelial nitric oxide synthase, improved its catalytic efficiency. These data implicate protein S-nitrosylation in the regulation of β-oxidation of fatty acids in mitochondria.
Article
Full-text available
Hydrogen sulfide (H2S) is a recently described endogenously produced gaseous signaling molecule that influences various cellular processes in the central nervous system, cardiovascular system, and gastrointestinal tract. The biogenesis of H2S involves the cytoplasmic transsulfuration enzymes, cystathionine β-synthase and γ-cystathionase, whereas its catabolism occurs in the mitochondrion and couples to the energy-yielding electron transfer chain. Low steady-state levels of H2S appear to be controlled primarily by efficient oxygen-dependent catabolism via sulfide quinone oxidoreductase, persulfide dioxygenase (ETHE1), rhodanese, and sulfite oxidase. Mutations in the persulfide dioxgenase, i.e. ETHE1, result in ethylmalonic encephalopathy, an inborn error of metabolism. In this study, we report the biochemical characterization and kinetic properties of human persulfide dioxygenase and describe the biochemical penalties associated with two patient mutations, T152I and D196N. Steady-state kinetic analysis reveals that the T152I mutation results in a 3-fold lower activity, which is correlated with a 3-fold lower iron content compared with the wild-type enzyme. The D196N mutation results in a 2-fold higher Km for the substrate, glutathione persulfide.
Article
Full-text available
Hydrogen sulfide (H(2)S) is now considered as the third gaseotransmitter, however, the signaling pathways that modulate the biomedical effect of H(2)S on endothelial cells are poorly defined. In the present study, we found in human endothelial cells that H(2)S increased cell migration rates and induced a marked reorganization of the actin cytoskeleton, which was prevented by depletion of Rac1. Pharmacologic inhibiting vascular endothelial growth factor receptor (VEGFR) and phosphoinositide 3-kinase (PI3K) both blunted the activation of Rac1 and the promotion of cell migration induced by H(2)S. Moreover, H(2)S-induced Rac1 activation was selectively dependent on the presence of the PI3K p110α isoform. Activated Rac1 by H(2)S thus in turn resulted in the phosphorylation of the F-actin polymerization modulator, cofilin. Additionally, inhibiting of extracellular signal-regulated kinase (ERK) decreased the augmented cell migration rate by H(2)S, but had no effect on Rac1 activation. These results indicate that Rac1 conveys the H(2)S signal to microfilaments inducing rearrangements of actin cytoskeleton that regulates cell migration. VEGFR-PI3K was found to be upstream pathway of Rac1, while cofilin acted as a downstream effector of Rac1. ERK was also shown to be involved in the action of H(2)S on endothelial cell migration, but independently of Rac1.
Article
Full-text available
Ethylmalonic encephalopathy (EE) is an invariably fatal disease, characterized by the accumulation of hydrogen sulfide (H(2)S), a highly toxic compound. ETHE1, encoding sulfur dioxygenase (SDO), which takes part in the mitochondrial pathway that converts sulfide into harmless sulfate, is mutated in EE. The main source of H(2)S is the anaerobic bacterial flora of the colon, although in trace amount it is also produced by tissues, where it acts as a 'gasotransmitter'. Here, we show that AAV2/8-mediated, ETHE1-gene transfer to the liver of a genetically, metabolically and clinically faithful EE mouse model resulted in full restoration of SDO activity, correction of plasma thiosulfate, a biomarker reflecting the accumulation of H(2)S, and spectacular clinical improvement. Most of treated animals were alive and well >6-8 months after birth, whereas untreated individuals live 26 ± 7 days. Our results provide proof of concept on the efficacy and safety of AAV2/8-mediated livergene therapy for EE, and alike conditions caused by the accumulation of harmful compounds in body fluids and tissues, which can directly be transferred to the clinic.
Article
Full-text available
An abundance of experimental evidence suggests that hydrogen sulfide (H(2)S) plays a prominent role in physiology and pathophysiology. Many targets exist for H(2)S therapy. The molecular targets of H(2)S include proteins, enzymes, transcription factors, and membrane ion channels. RECENT ADVANCES: Novel H(2)S precursors are being synthesized and discovered that are capable of releasing H(2)S in a slow and sustained manner. This presents a novel and advantageous approach to H(2)S therapy for treatment of chronic conditions associated with a decline in endogenous H(2)S, such as diabetes and cardiovascular disease. While H(2)S is cytoprotective at physiological concentrations, it is not universally cytoprotective, as it appears to have pro-apoptotic actions in cancer cells and is well known to be toxic at supraphysiological concentrations. Many of the pleiotropic effects of H(2)S on health are associated with the inhibition of inflammation and upregulation of prosurvival pathways. The powerful anti-inflammatory, cytoprotective, immunomodulating, and trophic effects of H(2)S on the vast majority of normal cells seem to be mediated mainly by its actions as an extremely versatile direct and indirect antioxidant and free radical scavenger. While the overall effects of H(2)S on transformed (i.e., malignant) cells can be characterized as pro-oxidant and pro-apoptotic, they contrast sharply with the cytoprotective effects on most normal cells. H(2)S has become a molecule of great interest, and several slow-releasing H(2)S prodrugs are currently under development. We believe that additional agents regulating H(2)S bioavailability will be developed during the next 10 years.
Article
Full-text available
Ethylmalonic encephalopathy (EE) is a rare autosomal recessive disorder characterized by early onset encephalopathy, chronic diarrhoea, petechiae, orthostatic acrocyanosis and defective cytochrome c oxidase (COX) in muscle and brain. High levels of lactic, ethylmalonic and methylsuccinic acids are detected in body fluids. EE is caused by mutations in ETHE1, a mitochondrial sulphur dioxygenase. By studying a suitable mouse model, we found that loss of ETHE1 leads to accumulation of sulphide, which is a poison for COX and other enzymatic activities thus accounting for the main features of EE. We report here the first autopsy case of a child with a genetically confirmed diagnosis of EE, and compare the histological, histochemical and immunohistochemical findings with those of the constitutive Ethe1 −/− mice. In addition to COX depleted cells, widespread endothelial lesions of arterioles and capillaries of the brain and gastrointestinal tract were the pathologic hallmarks in both organisms. Our findings of diffuse vascular damage of target critical organs are in keeping with the hypothesis that the pathologic effects of ETHE1 deficiency may stem from high levels of circulating hydrogen sulphide rather than the inability of specific organs to detoxify its endogenous production.
Article
Full-text available
A classical approach, protein separation by two-dimensional blue native/sodium dodecyl sulfate-polyacrylamide gel electrophoresis, was combined with tandem mass spectrometry and up-to-date computer technology to characterize the mitochondrial "protein complex proteome" of Arabidopsis (Arabidopsis thaliana) in so far unrivaled depth. We further developed the novel GelMap software package to annotate and evaluate two-dimensional blue native/sodium dodecyl sulfate gels. The software allows (1) annotation of proteins according to functional and structural correlations (e.g. subunits of a distinct protein complex), (2) assignment of comprehensive protein identification lists to individual gel spots, and thereby (3) selective display of protein complexes of low abundance. In total, 471 distinct proteins were identified by mass spectrometry, several of which form part of at least 35 different mitochondrial protein complexes. To our knowledge, numerous protein complexes were described for the first time (e.g. complexes including pentatricopeptide repeat proteins involved in nucleic acid metabolism). Discovery of further protein complexes within our data set is open to everybody via the public GelMap portal at www.gelmap.de/arabidopsis_mito.
Article
Full-text available
We have shown previously that a variant allele of the short-chain acyl-CoA dehydrogenase ( SCAD ) gene, 625G-->A, is present in homozygous form in 7% of control individuals and in 60% of 135 patients with elevated urinary excretion of ethylmalonic acid (EMA). We have now characterized three disease-causing mutations (confirmed by lack of enzyme activity after expression in COS-7 cells) and a new susceptibility variant in the SCAD gene of two patients with SCAD deficiency, and investigated their frequency in patients with elevated EMA excretion. The first SCAD-deficient patient was a compound heterozygote for two mutations, 274G-->T and 529T-->C. These mutations were not present in 98 normal control alleles, but the 529T-->C mutation was found in one allele among 133 patients with elevated EMA excretion. The second patient carried a 1147C-->T mutation and the 625G-->A polymorphism in one allele, and a single point mutation, 511C-->T, in the other. The 1147C-->T mutation was not present in 98 normal alleles, but was detected in three alleles of 133 patients with elevated EMA excretion, consistently as a 625A-1147T allele. On the other hand, the 511C-->T mutation was present in 13 of 130 and 15 of 67 625G alleles, respectively, of normal controls and patients with elevated EMA excretion, and was never associated with the 625A variant allele. This over-representation of the haplotype 511T-625G among the common 625G alleles in patients compared with controls was significant ( P < 0.02), suggesting that the allele 511T-625G-like 511C-625A-confers susceptibility to ethylmalonic aciduria. Expression of the variant R147W SCAD protein, encoded by the 511T-625G allele, in COS-7 cells showed 45% activity at 37 degrees C in comparison with the wild-type protein, comparable levels of activity at 26 degrees C, and 13% activity when incubated at 41 degrees C. This temperature profile is different from that observed for the variant G185S SCAD protein, encoded by the 511C-625A allele, where higher than normal activity was found at 26 and 37 degrees C, and 58% activity was present at 41 degrees C. These results corroborate the notion that the 511C-625A variant allele is one of the possible underlying causes of ethylmalonic aciduria, and suggest that the 511C-->T mutation represents a second susceptibility variation in the SCAD gene. We conclude that ethylmalonic aciduria, a commonly detected biochemical phenotype, is a complex multifactorial/polygenic condition where, in addition to the emerging role of SCAD susceptibility alleles, other genetic and environmental factors are involved.