S-nitrosoglutathione reductase (GSNOR) represents the best-documented denitrosylase implicated in regulating the levels of proteins posttranslationally modified by nitric oxide on cysteine residues by S-nitrosylation. GSNOR controls a diverse array of physiologic functions, including cellular growth and differentiation, inflammation, and metabolism. Chromosomal deletion of GSNOR results in pathologic protein S-nitrosylation that is implicated in human hepatocellular carcinoma (HCC). Here we identify a metabolic hallmark of aberrant S-nitrosylation in HCC and exploit it for therapeutic gain. We find that hepatocyte GSNOR deficiency is characterized by mitochondrial alteration and by marked increases in succinate dehydrogenase (SDH) levels and activity. We find that this depends on the selective S-nitrosylation of Cys501 in the mitochondrial chaperone TRAP1, which mediates its degradation. As a result, GSNOR-deficient cells and tumors are highly sensitive to SDH inhibition, namely to α-tocopheryl succinate, an SDH-targeting molecule that induced RIP1/PARP1-mediated necroptosis and inhibited tumor growth. Our work provides a specific molecular signature of aberrant S-nitrosylation in HCC, a novel molecular target in SDH, and a first-in-class therapy to treat the disease. Cancer Res; 76(14); 4170–82. ©2016 AACR.

S-nitrosylation is a cysteine posttranslational modification emerging as the main modification underlying nitric oxide (NO) bioactivity (1). The extent of S-nitrosocysteines (SNO) depends on: (i) NO production by NO synthase (NOS); (ii) how NO is conveyed by S-nitrosylase complexes on target proteins (2); (iii) SNO removal by denitrosylases, among which S-nitrosoglutathione reductase (GSNOR) is the best-characterized example (3, 4). GSNOR catalyzes S-nitrosoglutathione (GSNO) reduction, thus indirectly controlling cellular levels of protein SNOs (PSNO; refs. 3, 5).

GSNOR-KO mice develop hepatocellular carcinoma (HCC) in association with S-nitrosylation and proteasomal degradation of the DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT; ref. 6). Studies performed on human patients with HCC showed a significant decrease of GSNOR protein levels and activity (7), arguing for a functional link between GSNOR-dependent S-nitrosylation and HCC existing also in humans.

We recently observed that skeletal muscle of GSNOR-KO mice show altered mitochondrial network (8), suggesting that excessive S-nitrosylation affects mitochondrial homeostasis. This is in line with the evidence arguing for mitochondria representing a preferential target of NO (9), and respiratory chain being susceptible to S-nitrosylation (10).

Here, we demonstrate that GSNOR deficiency induces mitochondrial complex II (succinate dehydrogenase, SDH) upregulation due to S-nitrosylation and subsequent degradation of the TNF receptor–associated protein 1 (TRAP1). TRAP1 is a mitochondrial chaperone: (i) suppressing mitochondrial ROS production (11, 12); (ii) inhibiting permeability transition pore opening; (iii) preserving mitochondrial bioenergetics (11, 13); and (iv) acting as SDH activity inhibitor (14, 15). We provide evidence that TRAP1 degradation by S-nitrosylation upregulates SDH and sensitizes GSNOR-deficient HCC cells to SDH inhibitors, paving the way for the development of novel chemotherapeutic approaches.

Cell culture and treatments

HepG2 cell line was obtained from the Banca Biologica e Cell Factory and grown in RPMI1640. Hepa1-6 and Huh-7-12 were purchased from Sigma and grown in DMEM. Media were supplemented with 10% FBS at 37°C in 5% CO2. All cell lines were passaged for fewer than 2 months after resuscitation and were used from the third to the fifteenth passage in culture. Cell line validation was carried out by the manufacturer by means of DNA Profile STR (short tandem repeat). Cell lines were routinely screened for mycoplasma contamination by a PCR-based assay. Primary hepatocytes were obtained from GSNOR-KO and WT mice as described previously (16) and cultured in mammary epithelial cell growth medium (MEGM). Compounds and relative concentrations used in this study are as follows: 40 or 60 μmol/L α-TOS, 20 μmol/L 3BrPA, 5 mmol/L NAC, 200 μmol/L TROLOX, 100 μmol/L DHQ, 1 μmol/L 3AP, 1 mmol/L DTT, 50 μmol/L CHX, 500 μmol/L L-NAME (Sigma). 5 μmol/L DHE, 5 μmol/L H2DCFDA and 20 μmol/L DAF-FM-DA (Life Technologies); 10 μmol/L MG132 (Merck Millipore); 0.5 μmol/L epoxomicin and 10 μmol/L necrostatin-1 (NEC-1; Santa Cruz Biotechnology); 20 μmol/L z-VAD (MP Biomedicals). Cells transfections, Western blot analysis, and qRT-PCR analysis are described in Supplementary Materials.

Analysis of cell viability, survival, and apoptosis

Dead cells were evaluated by direct cell count upon Trypan blue staining or by fluorescence microscopy upon staining with LIVE/DEAD Cell Imaging Kit (488/570; Life Technologies). Cell viability was determined following fluorescence emission at 590 nm after 4-hour incubation with AlamarBlue Reagent (Life Technologies). Apoptotic cells were evaluated upon staining with 50 μmol/L propidium iodide (Sigma) by a FACSCalibur flow cytometer (BD Biosciences; ref. 17). Apoptotic cells refer to the hypodiploid nuclei (sub-G1) population. Clonogenic survival assay was performed as described previously (18). Electron and immunofluorescence microscopy analyses are described in Supplementary Materials.

ROS and nitric oxide evaluation

Thirty minutes before the end of treatment with SDH inhibitors (6 hours), cells were incubated with DHE or H2DCFDA at 37°C for the detection of superoxide or H2O2, respectively. Endogenous nitric oxide production was assessed in cells 24 hours after plating by 30-minute incubation with DAF-FM-DA. Fluorescence intensity was analyzed cytofluorometrically.

Enzymatic activities

Succinate:coenzyme Q oxidoreductase activity was assayed spectrophotometrically in purified mitochondria after the reduction of 2,6-dichlorophenolindophenol, DCPIP (Sigma) as reported previously (19). Lactate dehydrogenase (LDH) activity was measured in cell media after the oxidation of NADH (Roche). GSNOR activity was assayed as described by Jensen and colleagues (5).

Detection of PSNOs and TRAP1 S-nitrosylation

PSNOs were evaluated by biotin switch assay as described previously (8, 20). To identify TRAP1 S-nitrosylated sites, human recombinant TRAP1 (ADI-SPP-848; Enzo Life Sciences) was incubated with 2 mmol/L DTT for 1 hour, washed with PBS, and then maintained for 2 hours with 5 mmol/L DPTA-NONOate (Sigma). As control, equal amount of protein was left in DTT. Proteins were then subjected to biotin switch assay and digested with endopeptidase LysC (Wako) followed by trypsin (sequencing grade, Promega; ref. 21). Resulted peptides were analyzed by nanoLC-MS/MS as in ref. 22 and measurements were performed on an EASY-nLC system (Proxeon Biosystems) connected to the Q-Exactive mass spectrometer (Thermo Scientific) equipped with a nanoelectrospray ion source (Proxeon Biosystems). The Q-Exactive was operated in data-dependent mode with the MS survey scan ranging from 300 to 1,750 m/z and a resolution of 70,000. The 12 most abundant ions with charge ≥ 2+ from each survey scan were sequentially isolated (isolation window of 2 m/z) and subjected to MS/MS fragmentation by high-energy collisional dissociation (HCD; ref. 23). Raw data were processed with MaxQuant software suite version 1.3.0.5 searching peak lists agains UniProt human database. Methylthiolation and HPDP-Biotinylation were manually configured as variable peptide modifications. Maximum false discovery rates were set to 0.01 for proteins, peptides, and modification sites.

Animal experimentation

Mouse experiments were conducted at the mouse facility of the Danish Cancer Society Research Center with the approval of the Regional Committee for Medical Research Ethics in Denmark. GSNOR-KO mice were generated in J.S. Stamler's laboratory on C57BL/6J background (4). Livers were collected from 2-month-old mice and used for Western blot analyses or processed to evaluate mitochondrial oxygen consumption. Xenograft experiments were conducted on NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (SCID) mice strains. Tumors were generated by subcutaneous injection, into the right lower flank, of 5 × 106 HepG2 cells stably downregulating GSNOR (n = 16) or scrambled counterparts (n = 16). When palpable tumors were recognized, each group was split into α-TOS–treated (n = 10) and vehicle-treated (n = 6) groups. α-TOS was dissolved in Corn oil/4% ethanol as reported previously (24) and used at 5 μmol (2 injections/week). Treatments started when tumor diameter reached 5 mm, and tumor size was measured twice per week for 2 weeks.

ATP measurement

ATP levels were measured by the ATP Bioluminescence Assay Kit CLS II (Roche) using a microplate luminometer (PerkinElmer).

Analyses of mitochondrial mass and transmembrane potential (Δψm)

Total mitochondrial mass and Δψm were analyzed cytofluorometrically by incubating the cells with 50 nmol/L MitoTracker Green-FM and 200 nmol/L MitoTracker Red CMXRos (Life Technologies), respectively. Alternatively, Δψm was evaluated by fluorescence microscopy upon JC-1 staining (Life Technologies; ref. 8).

Cell fractionation and oxygen consumption

Mitochondria were isolated and oxygen consumption determined as described previously (25) in intact mitochondria, or in mitochondrial membranes obtained upon osmotic lysis in water as previously reported (26). P:O ratio calculated in intact mitochondria of WT and GSNOR-KO livers was 2.34 ± 0.13 versus 2.09 ± 0.13, respectively.

Protein determination

Protein concentration was determined by the method of Lowry and colleagues (27).

Statistical analysis

Values are expressed as means ± SD (or SEM when indicated). The statistical significance of the differences between means was assessed by independent Student t test or one-way ANOVA test using GraphPad Prism.

GSNOR downregulation results in mitochondrial defects

GSNOR-KO mice spontaneously develop HCC (7) and show mitochondrial fragmentation in skeletal muscle (8). On the basis of this, we investigated whether mitochondria of liver obtained from 2-month-old GSNOR-KO mice (which did show no sign of HCC hitherto) exhibited defects in mitochondrial respiration and function. Respiration, measured upon malate/glutamate addition (involving complex I), was significantly dampened in GSNOR-KO mitochondria, whereas respiration measured upon succinate addition (starting from complex II) was about 40% higher than the WT counterparts, notwithstanding complex IV efficiency was decreased (Fig. 1A, left). To untie complex I and II activities from complex IV oxygen consumption, we repeated the analyses in mitochondrial membranes obtained upon osmotic lysis. Figure 1A (right) shows no difference of complex I activity between the two genotypes. In contrast, complex II activity was even more increased in GSNOR-KO samples, suggesting that this effect was masked in intact mitochondria due to the downstream inhibition of complex IV. These results were strengthened by Western blot analysis of complex IV (subunit I), which were reduced in GSNOR-KO liver extracts, and complex II (as revealed by its subunit A) that, conversely, significantly increased (Fig. 1B).

Figure 1.

GSNOR deficiency results in mitochondrial alteration without affecting NO production. A, oxygen consumption analyses performed on mitochondria purified from livers of GSNOR wild-type (WT) and deficient (KO) mouse (left). Five mmol/L malate and 5 mmol/L glutamate (Mal/Glu), 5 mmol/L succinate or 5 mmol/L TMPD/ascorbate were added to mitochondria to evaluate the activity of complex I, II and IV, respectively. Right, oxygen consumption performed in osmotically lysed mitochondria measured upon NADH or succinate addition. 5 μmol/L rotenone (Rot) and 20 μmol/L 3-nitropropionate (3NP) were used to irreversibly inhibit complex I and II, respectively. n = 3; *P ≤ 0.05. B, Western blots of GSNOR and subunits of mitochondrial complexes, NDUFB8 (complex I), SDHA (complex II), and subunit I (complex IV) performed in GSNOR WT and KO liver homogenates. Western blot analysis of GSNOR and NOS isoforms (C) and qRT-PCR (D) in HepG2 cells transfected with scrambled siRNA (siScr) or with two siRNAs against GSNOR (siGSNOR 1 and siGSNOR 2). n ≥ 3; ***, P < 0.0001. E, evaluation of PSNOs. Densitometry of PSNOs signal is shown on the left; *, P = 0.0491. F, cytofluorometric evaluation of NO. G, Western blot analysis and densitometry of immunoreactive bands of complex II. n = 3; ***, P = 0.001. H, analyses of enzymatic activity of complex II. n = 3; **, P = 0.0051; *, P = 0.043.

Figure 1.

GSNOR deficiency results in mitochondrial alteration without affecting NO production. A, oxygen consumption analyses performed on mitochondria purified from livers of GSNOR wild-type (WT) and deficient (KO) mouse (left). Five mmol/L malate and 5 mmol/L glutamate (Mal/Glu), 5 mmol/L succinate or 5 mmol/L TMPD/ascorbate were added to mitochondria to evaluate the activity of complex I, II and IV, respectively. Right, oxygen consumption performed in osmotically lysed mitochondria measured upon NADH or succinate addition. 5 μmol/L rotenone (Rot) and 20 μmol/L 3-nitropropionate (3NP) were used to irreversibly inhibit complex I and II, respectively. n = 3; *P ≤ 0.05. B, Western blots of GSNOR and subunits of mitochondrial complexes, NDUFB8 (complex I), SDHA (complex II), and subunit I (complex IV) performed in GSNOR WT and KO liver homogenates. Western blot analysis of GSNOR and NOS isoforms (C) and qRT-PCR (D) in HepG2 cells transfected with scrambled siRNA (siScr) or with two siRNAs against GSNOR (siGSNOR 1 and siGSNOR 2). n ≥ 3; ***, P < 0.0001. E, evaluation of PSNOs. Densitometry of PSNOs signal is shown on the left; *, P = 0.0491. F, cytofluorometric evaluation of NO. G, Western blot analysis and densitometry of immunoreactive bands of complex II. n = 3; ***, P = 0.001. H, analyses of enzymatic activity of complex II. n = 3; **, P = 0.0051; *, P = 0.043.

Close modal

To investigate the effects of GSNOR deficiency in the context of HCC, we silenced GSNOR in the human HCC cell line HepG2 (siGSNOR HepG2) with two different pools of siRNAs (Fig. 1C and D), inducing 2-fold increase in PSNOs content (Fig. 1E). However, they did not affect NOS isoform expression pattern (with nNOS being undetectable, see Fig. 1C), or alter NO production (Fig. 1F), indicating that changes in S-nitrosylation were independent on NO synthesis.

In line with results obtained in liver extracts, SDHA levels and Complex II activity were upregulated in GSNOR-silenced cells (Fig. 1G and H). However, mitochondrial proteins and mass (Supplementary Fig. S1A and S1B) and gene expression of all four SDH subunits (Supplementary Fig. S1C) did not change, indicating that SDH upregulation was not due to any transcriptional regulation or change in the number of mitochondria. We also observed that ATP levels tend to decrease (Supplementary Fig. S1D and S1E), whereas Δψm significantly dropped with respect to siScr cells (Supplementary Fig. S1F and S1G).

GSNOR downregulation modulates SDH by affecting TRAP1 stability via S-nitrosylation at Cys501

To unveil whether SDH upregulation upon GSNOR depletion was related to S-nitrosylation, we focused on the mitochondrial chaperone TRAP1, which has recently been found to affect SDH activity (14). Western blot analysis revealed that TRAP1 protein levels were reduced in GSNOR-depleted cells (Fig. 2A; Supplementary Table S3), despite no change in mRNA was detected (Supplementary Fig. S2A). We then hypothesized that TRAP1 could be destabilized and undergo degradation owing to S-nitrosylation, with this assumption strengthening by the evidence that: (i) incubations with the proteasome inhibitor MG132 completely restored TRAP1 levels in siGSNOR cells (Fig. 2B) and (ii) pretreatment with the NOS inhibitor Nω-nitro-l-arginine methyl ester (L-NAME) prevented TRAP1 decrease and SDHA upregulation (Supplementary Fig. S2B). We then evaluated TRAP1 protein levels by Western blot analysis on a pool of PSNOs obtained by biotin switch assays. Results shown in Fig. 2C indicate that S-nitrosylated TRAP1 accumulated upon proteasome inhibition only in siGSNOR cells, arguing for S-nitrosylation being associated with TRAP1-selective degradation. Consistently, siGSNOR or siTRAP1 HepG2 cells showed the same expression pattern as SDHA and TRAP1, with the former upregulated and the latter decreased (Fig. 2D). Moreover, TRAP1 overexpression was able to downregulate SDH (Fig. 2E) and to accelerate its turnover in the presence of the protein translation inhibitor cycloheximide (Fig. 2F).

Figure 2.

GSNOR downregulation modulates SDH by affecting TRAP1 stability via S-nitrosylation at Cys501. A, Western blot analysis of TRAP1 in total cell lysates from siScr and siGSNOR HepG2. B, same analyses as in A performed in the presence or absence of 10 μmol/L MG132 (6 hours). C, detection of S-nitrosylated TRAP1 (SNO-TRAP1) upon biotin switch assay, followed by TRAP1 immunoblot. Total input is also shown as control. D, Western blots of SDHA and TRAP1 in siScr, siGSNOR, and siTRAP1 cells. E, Western blots of TRAP1 and SDHA upon overexpression of WT TRAP1. F, Western blot analysis of SDHA in total cell lysates from mock (empty vector) and WT TRAP1-overexpressing cells upon incubation with 50 μmol/L cycloheximide (CHX) for the indicated times. Western blot analysis of cytosolic (C) and mitochondrial (M) extracts of siScr and siGSNOR cells incubated for 6 hours with 10 μmol/L MG132 or with vehicle alone (DMSO; G) or GSNOR-KO or WT livers (H). TOM20 and LDH were selected as both loading and purity controls. I, Western blot analysis of TRAP1 upon incubation with 10 μmol/L MG132 or 0.5 μmol/L epoxomicin (EPOX). J, Western blot analysis of TRAP1 in cells incubated with 50 μmol/L cycloheximide for the indicated times. K, schematic model of biotin switch assay coupled with mass spectrometry. L, representative nano LC/MS-MS spectrum indicating SNO-modification at Cys501 as demonstrated by the presence of biotin-Cys label. M, Western blot analysis of TRAP1 in siGSNOR cells overexpressing WT or TRAP1-C501S incubated with 50 μmol/L cycloheximide for the indicated times. N, Western blot analysis of SDHA and TRAP1 in mock and TRAP1-C501S-overexpressing cells. O, Western blot analysis of SDHA in same cells as in N upon treatment with 50 μmol/L cycloheximide for the indicated times. P, complex II activity in same cells as in N. Graph shown represent the mean of data ± SD. n = 3; **, P = 0.0047.

Figure 2.

GSNOR downregulation modulates SDH by affecting TRAP1 stability via S-nitrosylation at Cys501. A, Western blot analysis of TRAP1 in total cell lysates from siScr and siGSNOR HepG2. B, same analyses as in A performed in the presence or absence of 10 μmol/L MG132 (6 hours). C, detection of S-nitrosylated TRAP1 (SNO-TRAP1) upon biotin switch assay, followed by TRAP1 immunoblot. Total input is also shown as control. D, Western blots of SDHA and TRAP1 in siScr, siGSNOR, and siTRAP1 cells. E, Western blots of TRAP1 and SDHA upon overexpression of WT TRAP1. F, Western blot analysis of SDHA in total cell lysates from mock (empty vector) and WT TRAP1-overexpressing cells upon incubation with 50 μmol/L cycloheximide (CHX) for the indicated times. Western blot analysis of cytosolic (C) and mitochondrial (M) extracts of siScr and siGSNOR cells incubated for 6 hours with 10 μmol/L MG132 or with vehicle alone (DMSO; G) or GSNOR-KO or WT livers (H). TOM20 and LDH were selected as both loading and purity controls. I, Western blot analysis of TRAP1 upon incubation with 10 μmol/L MG132 or 0.5 μmol/L epoxomicin (EPOX). J, Western blot analysis of TRAP1 in cells incubated with 50 μmol/L cycloheximide for the indicated times. K, schematic model of biotin switch assay coupled with mass spectrometry. L, representative nano LC/MS-MS spectrum indicating SNO-modification at Cys501 as demonstrated by the presence of biotin-Cys label. M, Western blot analysis of TRAP1 in siGSNOR cells overexpressing WT or TRAP1-C501S incubated with 50 μmol/L cycloheximide for the indicated times. N, Western blot analysis of SDHA and TRAP1 in mock and TRAP1-C501S-overexpressing cells. O, Western blot analysis of SDHA in same cells as in N upon treatment with 50 μmol/L cycloheximide for the indicated times. P, complex II activity in same cells as in N. Graph shown represent the mean of data ± SD. n = 3; **, P = 0.0047.

Close modal

The possibility that TRAP1 could be degraded via the proteasome has been suggested (28). However, mitochondrial proteins are usually targets of specialized mitochondrial proteases (29). Therefore, to clarify whether S-nitrosylation–induced degradation of TRAP1 was mediated by the proteasome, we purified cytosolic and mitochondrial fractions of HepG2 cells and mouse livers, confirming that TRAP1 significantly decreased only in mitochondria of GSNOR-deficient systems (Fig. 2G and H). Moreover, incubation with MG132 showed that TRAP1 accumulated exclusively in the cytosol of siGSNOR cells (Fig. 2G), where the levels of ubiquitylated TRAP1 increased even in the absence of MG132 (Supplementary Fig. S2C). The evidence that the highly selective proteasome inhibitor epoxomicin produced the same effects of MG132 (Fig. 2I) reinforced the hypothesis that TRAP1 degradation occurred via the proteasome. In agreement, incubations with cycloheximide resulted in a more rapid turnover of TRAP1 in siGSNOR cells (Fig. 2J).

To identify the cysteine residue(s) being modified by NO, we treated recombinant human TRAP1 with DETA-NONOate and performed a biotin switch assay, followed by analysis with high accuracy mass spectrometry (Fig. 2K). The mass spectrometric measurements unambiguously identified TRAP1 peptide bearing biotinylation at Cys501 residue, implementing Cys501 as S-nitrosylation site (Fig. 2L), and confirming previous high-throughput proteomic studies (30). On the basis of these results, we generated a human TRAP1-C501S mutant and induced its expression in siGSNOR cells. Western blots, obtained upon incubation with cycloheximide, indicate that TRAP1-C501S was more stable (Fig. 2M), confirming that S-nitrosylation of Cys501 prepares the way for TRAP1 degradation.

To understand whether Cys-to-Ser substitution was still able to maintain TRAP1 functional relationship with SDH, we overexpressed TRAP1-C501S mutant in parental HepG2. Under these conditions, SDH protein levels were reduced (Fig. 2N) and its turnover accordingly decreased (Fig. 2O). In agreement, TRAP1-C501S was able to revert the increase of Complex II activity observed in siGSNOR cells (Fig. 2P).

GSNOR downregulation sensitizes HepG2 cells to oxidative stress and cytotoxicity induced by SDH-targeting mitocans

We attempted to exploit the unusual TRAP1 decrease and take advantage by SDH upregulation to selectively target HCC cells. We analyzed the occurrence of cell death upon treatment with two different SDH inhibitors listed as mitocans, namely: 3-bromopyruvate (3-BrPA), which alkylates SDH sulfhydryl groups (31), and α-tocopheryl succinate (α-TOS), which specifically binds to ubiquinone binding site of SDH (32). As negative control, we used malonate, the physiologic SDH inhibitor, at a concentration of 20 mmol/L to abolish Complex II activity. Direct cell counts showed that siGSNOR cells were insensitive to malonate, but more susceptible to treatments with α-TOS and 3-BrPA used at concentrations in a range that provides a similar extent of cell death (Supplementary Fig. S3A). Further cell viability and clonogenic survival assays performed with α-TOS produced similar results, confirming that GSNOR downregulation sensitized HepG2 cells to α-TOS treatment (Fig. 3A and B).

Figure 3.

GSNOR downregulation sensitizes HepG2 cells to SDH-targeting mitocans cytotoxicity via ROS production. A, cell viability fluorescent assay performed in siScr and siGSNOR cells treated with 40 or 60 μmol/L α-TOS. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. Data were analyzed using ANOVA with Dunnett multiple comparisons test; **, P = 0.0048; ***, P = 0.0006 with respect to DMSO-treated siGSNOR cells. B, representative clonogenic assay of cells treated as in A (left); statistical analyses of colony number upon α-TOS treatments shown as percent of vehicle (DMSO)-treated cells (right); ***, P ≤ 0.001; *, P ≤ 0.05. Cytofluorimetric assays of superoxide (C) and H2O2 (D) in cells preincubated with 100 μmol/L TROLOX, overexpressing SOD2 or a mitochondrial form of catalase (mitoCAT) and treated with 40 μmol/L α-TOS. n = 3, *, P ≤ 0.05; **, P ≤ 0.01. E, Western blot analysis of protein carbonyls of siGSNOR cells treated as in C and D. F, cell viability of siGSNOR cells treated as in C and D. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. ***, P ≤ 0.001 with respect to α-TOS–treated siGSNOR cells. G, representative clonogenic assay of siGSNOR cells treated as in C and D (left); statistical analyses (right); **, P ≤ 0.01. H, cell viability assay of siGSNOR cells, overexpressing TRAP1-C501S or the empty vector, treated with 40 μmol/L α-TOS. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. ***, P ≤ 0.001 with respect to mock cells treated with α-TOS. I, representative clonogenic assay of TRAP1-C501S–overexpressing siGSNOR cells treated as in H; right, statistical analyses; ***, P ≤ 0.001.

Figure 3.

GSNOR downregulation sensitizes HepG2 cells to SDH-targeting mitocans cytotoxicity via ROS production. A, cell viability fluorescent assay performed in siScr and siGSNOR cells treated with 40 or 60 μmol/L α-TOS. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. Data were analyzed using ANOVA with Dunnett multiple comparisons test; **, P = 0.0048; ***, P = 0.0006 with respect to DMSO-treated siGSNOR cells. B, representative clonogenic assay of cells treated as in A (left); statistical analyses of colony number upon α-TOS treatments shown as percent of vehicle (DMSO)-treated cells (right); ***, P ≤ 0.001; *, P ≤ 0.05. Cytofluorimetric assays of superoxide (C) and H2O2 (D) in cells preincubated with 100 μmol/L TROLOX, overexpressing SOD2 or a mitochondrial form of catalase (mitoCAT) and treated with 40 μmol/L α-TOS. n = 3, *, P ≤ 0.05; **, P ≤ 0.01. E, Western blot analysis of protein carbonyls of siGSNOR cells treated as in C and D. F, cell viability of siGSNOR cells treated as in C and D. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. ***, P ≤ 0.001 with respect to α-TOS–treated siGSNOR cells. G, representative clonogenic assay of siGSNOR cells treated as in C and D (left); statistical analyses (right); **, P ≤ 0.01. H, cell viability assay of siGSNOR cells, overexpressing TRAP1-C501S or the empty vector, treated with 40 μmol/L α-TOS. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. ***, P ≤ 0.001 with respect to mock cells treated with α-TOS. I, representative clonogenic assay of TRAP1-C501S–overexpressing siGSNOR cells treated as in H; right, statistical analyses; ***, P ≤ 0.001.

Close modal

Oxidative stress is one of the main effects induced by mitocans, thus contributing to their antitumor activity (24, 33). Therefore, we measured superoxide and H2O2 levels and found that both increased significantly when GSNOR was downregulated (Fig. 4C and D). Interestingly, when we preincubated the cells with the ROS scavenger TROLOX, as well as when we ectopically overexpressed either mitochondrial superoxide dismutase (SOD2) or catalase fused with a mitochondrial import sequence (mitoCAT; Supplementary Fig. S3C and S3D), ROS production was inhibited and protein oxidative damage was prevented (Fig. 3C–E). This was associated with a complete recovery of cell viability (Fig. 3F and G and Supplementary Fig. S3B). We then transfected HepG2 cells with siRNA against GSNOR in combination with siRNAs against each SDH subunit (SDH A–D). In these conditions, superoxide production was prevented and α-TOS cytotoxicity was completely abolished (Supplementary Fig. S3E and S3F), confirming the selectivity of α-TOS towards mitochondrial complex II. Moreover, TRAP1-C501S rendered siGSNOR cells insensitive to α-TOS toxicity (Fig. 3H and I) and abolished superoxide production (Supplementary Fig. S3G), implying that TRAP1 S-nitrosylation is required for α-TOS toxicity. In agreement, TRAP1 silencing enhanced α-TOS vulnerability of parental HepG2 cells (Supplementary Fig. S3H).

Figure 4.

Cell death induced by SDH-directed mitocans proceeds via the PARP1/RIP1-dependent necroptosis. A, Western blots of caspase-9 (Casp9), caspase-3 (Casp3), and PARP1 in siScr and siGSNOR cells treated with 40 μmol/L α-TOS for 9 and 24 hours. B, LDH activity of extracellular media of cells treated for 24 hours with 40 μmol/L α-TOS. n = 3; *, P = 0.0118. C, analyses of cell death upon Trypan blue staining of cells transfected separately with siScr, siGSNOR, siPARP1, or with a siGSNOR/siPARP1 combination and treated for 24 hours with 40 μmol/L α-TOS. n ≥ 3; ***, P = 0.0001. D, same analyses as in C of α-TOS–treated cells incubated with PARP1 inhibitors 3-aminophthalhydrazide (3AP, 1 mmol/L; n = 3; **, P = 0.0048) or 1,5-dihydroxyisoquinoline (DHQ, 100 μmol/L; n = 3; **, P = 0.0021). E, Western blot analysis of RIP1 in α-TOS–treated cells. Immunoreactive bands densitometry is shown on the top. n = 3; **, P = 0.0011. F, analyses of cell death upon Trypan blue staining of α-TOS–treated cells incubated with 10 μmol/L NEC-1. n = 3; **, P = 0.001. G, Western blot analysis of RIP1 in α-TOS–treated cells incubated with 100 μmol/L DHQ or transfected with siPARP1. Immunoreactive bands densitometry is shown at the top. n = 3; ***, P = 0.0006; **, P = 0.0018. H, representative clonogenic survival assay of siGSNOR cells preincubated with 10 μmol/L NEC-1 or 1 mmol/L DTT and treated with 40 μmol/L α-TOS (top); bottom, statistical analyses; ***, P ≤ 0.001. I, electron microscopy analyses of cells treated for 18 hours with 40 μmol/L α-TOS. High magnification insets are shown. Arrowheads, mitochondria; n, nucleus. Scale bar, 5 μm.

Figure 4.

Cell death induced by SDH-directed mitocans proceeds via the PARP1/RIP1-dependent necroptosis. A, Western blots of caspase-9 (Casp9), caspase-3 (Casp3), and PARP1 in siScr and siGSNOR cells treated with 40 μmol/L α-TOS for 9 and 24 hours. B, LDH activity of extracellular media of cells treated for 24 hours with 40 μmol/L α-TOS. n = 3; *, P = 0.0118. C, analyses of cell death upon Trypan blue staining of cells transfected separately with siScr, siGSNOR, siPARP1, or with a siGSNOR/siPARP1 combination and treated for 24 hours with 40 μmol/L α-TOS. n ≥ 3; ***, P = 0.0001. D, same analyses as in C of α-TOS–treated cells incubated with PARP1 inhibitors 3-aminophthalhydrazide (3AP, 1 mmol/L; n = 3; **, P = 0.0048) or 1,5-dihydroxyisoquinoline (DHQ, 100 μmol/L; n = 3; **, P = 0.0021). E, Western blot analysis of RIP1 in α-TOS–treated cells. Immunoreactive bands densitometry is shown on the top. n = 3; **, P = 0.0011. F, analyses of cell death upon Trypan blue staining of α-TOS–treated cells incubated with 10 μmol/L NEC-1. n = 3; **, P = 0.001. G, Western blot analysis of RIP1 in α-TOS–treated cells incubated with 100 μmol/L DHQ or transfected with siPARP1. Immunoreactive bands densitometry is shown at the top. n = 3; ***, P = 0.0006; **, P = 0.0018. H, representative clonogenic survival assay of siGSNOR cells preincubated with 10 μmol/L NEC-1 or 1 mmol/L DTT and treated with 40 μmol/L α-TOS (top); bottom, statistical analyses; ***, P ≤ 0.001. I, electron microscopy analyses of cells treated for 18 hours with 40 μmol/L α-TOS. High magnification insets are shown. Arrowheads, mitochondria; n, nucleus. Scale bar, 5 μm.

Close modal

SDH inhibitors induce PARP1/RIP1-dependent necroptosis in GSNOR downregulating HepG2 cells

By looking at the molecular mechanisms underlying the cell death process observed, we analyzed the possible occurrence of apoptosis. Western blot analyses indicated that cleavage of caspase-9 and caspase-3, as well as proteolysis of PARP1, were not induced in α-TOS–treated siGSNOR cells (Fig. 4A). No increase of apoptotic cells was observed (Supplementary Fig. S4A), and no rescue of cell viability was achieved upon the inhibition of caspases (Supplementary Fig. S4B). Subcellular localization analyses of the apoptosis inducing factor 1 (AIF1) also excluded a caspase-independent apoptosis (Supplementary Fig. S4C). Conversely, we observed an increase of LDH release in the extracellular medium (Fig. 4B), suggesting that treatment with α-TOS reasonably induced necrosis in siGSNOR cells.

Several findings have reported that caspases undergo S-nitrosylation and inactivation (34–36). Consistently, we observed that caspase-3 underwent massive S-nitrosylation in siGSNOR cells (Supplementary Fig. S4D), suggesting that apoptosis, even though induced, could be inhibited by excessive S-nitrosylation. To confirm this hypothesis, we preincubated the cells with the thiol-reducing agent dithiotreitol (DTT) and treated them with higher concentration of α-TOS to also induce a significant extent of cell death in siScr cells. DTT significantly switched cell death response in siGSNOR cells from full necrosis to apoptosis at the same extent measured in siScr cells (Supplementary Fig. S4E), thus arguing for the denitrosylating effect of DTT being entirely compensatory of GSNOR deficiency. In agreement, PARP1 was cleaved, signaling the effective accomplishment of the apoptotic program (Supplementary Fig. S4F).

In the search for the molecular determinants of mitocan-induced necrosis, we pondered the evidence that full-length PARP1 seemed actually to be induced upon α-TOS treatment (see Fig. 4A). Consistently, polyADP-ribosylated (pADPr) protein levels also increased in siGSNOR cells, even in the absence of α-TOS (Supplementary Fig. S4G). Long-lasting PARP1 hyperactivation results in necrosis (37). Therefore, we investigated whether PARP1 could have a role in α-TOS–induced cell death. Figure 4C and D indicate that the extent of dead cells significantly decreased by inhibiting PARP1 activity either genetically (siRNA against PARP1), or pharmacologically (pretreatments with PARP1 inhibitors).

We then verified whether PARP1 triggered programmed necrosis by the activation of the receptor-activating protein 1 (RIP1; ref. 38). Particularly, we observed that RIP1 was induced in siGSNOR cells treated with α-TOS (Fig. 4E) and that preincubation with the RIP1 inhibitor NEC-1 reverted cell death (Fig. 4F). Moreover, PARP1 inhibition reduced RIP1 levels (Fig. 4G), confirming the crucial role of PARP1/RIP1 signaling axis in transducing death signals under nitrosative stress. Indeed, both NEC-1 and DTT restored clonogenic survival of siGSNOR cells treated with α-TOS (Fig. 4H). To widen the biologic significance of our findings, we also performed experiments with 3-BrPa obtaining similar results (Supplementary Fig. S4H).

To finally confirm the occurrence of necroptosis, we analyzed the cell ultrastructure through electron microscopy (39). siGSNOR cells exposed to α-TOS generally display mitochondrial swelling and damage, whose severity increased in a dose-dependent manner (Fig. 4I). Particularly, upon treatment with 40 μmol/L α-TOS, the normal structure of inner mitochondrial cristae appeared almost completely lost. Nuclei were still intact but swollen and cytoplasmic volume was substantially reduced. Conversely, in siScr cells, α-TOS induced changes in subcellular structures only at the highest concentrations used (60 μmol/L), with many of them resembling the canonical features of apoptosis (Supplementary Fig. S4I).

GSNOR downregulation selectively sensitizes HCC cells to α-TOS in an SDH-dependent manner

We next tested α-TOS effects on cell viability of other two different HCC cell lines, HuH7 and Hepa1-6, along with primary murine hepatocytes (Fig. 5A). α-TOS did not affect cell viability of hepatocytes as previously reported (40), but induced only a mild cytostatic effect above 80 μmol/L, with this being independent on GSNOR expression (Fig. 5B and C). In contrast, α-TOS induced cell death in HCC cells, although to different extents. In particular, Hepa1-6 were less susceptible, displaying an EC50 = 175.04 μmol/L versus 74.07 and 60.06 μmol/L of HuH7 and HepG2, respectively. Furthermore, GSNOR downregulation did not enhance α-TOS toxicity in Hepa1-6 cells (Fig. 5D). Going into depth on this different response, we found that Hepa1-6 cells displayed a halved mitochondrial content and 8-fold decrease of SDH activity (Supplementary Fig. S5A and Fig. 5E), confirming the role of mitochondria as elective target for HCC selective killing. On the other hand, GSNOR-downregulating HuH7 cells recapitulated what previously observed in HepG2. In particular, GSNOR deficiency resulted in TRAP1 downregulation and SDH increase (Fig. 5F), condition enhancing α-TOS toxicity (Fig. 5G and H). Cell death was reverted by preincubation with DTT and NEC-1, as well as by overexpressing TRAP1-C501S mutant (Fig. 5I). Similarly, α-TOS induced cell death by means of massive superoxide and H2O2 production that were dampened upon preincubation with TROLOX or by overexpression of SOD2 and mitoCAT (Fig. 5K and L and Supplementary Fig. S5B and S5C).

Figure 5.

SDH levels are predictive of GSNOR-deficient HCC sensitivity to α-TOS. A, dose–response curves to α-TOS of HCC cell lines HepG2, HuH7, and Hepa1-6 and primary mouse hepatocytes. EC values are shown at the top of the graph. B, analyses of cell death of α-TOS–treated GSNOR-KO and WT mouse hepatocytes upon staining with Alamar blue. C, cell viability assay of α-TOS–treated mouse hepatocytes. D, analyses of cell death of siScr and siGSNOR Hepa1-6 cells treated with α-TOS upon staining with Alamar blue. E, complex II activity in HepG2, HuH7, and Hepa1-6. n = 3; ***, P ≤ 0.0001. F, Western blots of SDHA, TRAP1, and GSNOR in HuH7 cells. G, analyses of cell death upon Trypan blue staining in α-TOS–treated HuH7 cells. n = 3; **, P = 0.0091. H, cell viability assay performed on α-TOS–treated HuH7 cells. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments; ***, P ≤ 0.0001 with respect to α-TOS-treated siScr cells. I, analyses of cell death upon Trypan blue staining of α-TOS–treated HuH7 cells preincubated with 1 mmol/L DTT, 10 μmol/L NEC-1, or overexpressing TRAP1-C501S. n = 3; **, P ≤ 0.005. Cytofluorimetric assays of superoxide (J) and H2O2 (K) of α-TOS–treated-HuH7 cells preincubated with 100 μmol/L TROLOX, overexpressing SOD2 or mCAT. n = 3, *, P ≤ 0.05; **, P ≤ 0.01. L, viability assay of cells treated as in J and K. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. ***, P ≤ 0.001 with respect to α-TOS–treated cells.

Figure 5.

SDH levels are predictive of GSNOR-deficient HCC sensitivity to α-TOS. A, dose–response curves to α-TOS of HCC cell lines HepG2, HuH7, and Hepa1-6 and primary mouse hepatocytes. EC values are shown at the top of the graph. B, analyses of cell death of α-TOS–treated GSNOR-KO and WT mouse hepatocytes upon staining with Alamar blue. C, cell viability assay of α-TOS–treated mouse hepatocytes. D, analyses of cell death of siScr and siGSNOR Hepa1-6 cells treated with α-TOS upon staining with Alamar blue. E, complex II activity in HepG2, HuH7, and Hepa1-6. n = 3; ***, P ≤ 0.0001. F, Western blots of SDHA, TRAP1, and GSNOR in HuH7 cells. G, analyses of cell death upon Trypan blue staining in α-TOS–treated HuH7 cells. n = 3; **, P = 0.0091. H, cell viability assay performed on α-TOS–treated HuH7 cells. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments; ***, P ≤ 0.0001 with respect to α-TOS-treated siScr cells. I, analyses of cell death upon Trypan blue staining of α-TOS–treated HuH7 cells preincubated with 1 mmol/L DTT, 10 μmol/L NEC-1, or overexpressing TRAP1-C501S. n = 3; **, P ≤ 0.005. Cytofluorimetric assays of superoxide (J) and H2O2 (K) of α-TOS–treated-HuH7 cells preincubated with 100 μmol/L TROLOX, overexpressing SOD2 or mCAT. n = 3, *, P ≤ 0.05; **, P ≤ 0.01. L, viability assay of cells treated as in J and K. Percentage of dead (red) cells is shown and represents the mean count ± SEM of n ≥ 3 different fields of n = 3 independent experiments. ***, P ≤ 0.001 with respect to α-TOS–treated cells.

Close modal

α-TOS induces tumor regression in GSNOR-downregulating HepG2 tumor xenografts

We next moved to HepG2 xenografts implanted in SCID mice, and evaluated α-TOS antitumor efficacy. We generated HepG2 cell lines stably expressing shRNAs against GSNOR (shGSNOR) (Fig. 6A). Then, we selected two clones showing TRAP1 downregulation, SDHA increase (Fig. 6B), and sensitivity to α-TOS cytotoxicity (Fig. 6C) to exclude any cellular compensatory mechanism(s) emerging during clonal selection. We injected subcutaneously shGSNOR or control shSrc HepG2 cells and once tumor masses reached 5 mm diameter, we treated the mice for 2 weeks by intraperitoneal administrations of α-TOS, provided at low doses (5 μmol each three days). Results shown in Fig. 6D and E indicate that volumes, as well as final weights and size, were significantly reduced in shGSNOR HCC xenografts with respect to the vehicle-treated counterparts, whereas no significant difference was observed in shScr xenografts. shGSNOR xenografts treated with α-TOS showed increased protein carbonylation (Fig. 6F), and induction of RIP1 expression (Fig. 6G), confirming that α-TOS susceptibility was due to necroptosis-like cell death induced by oxidative stress.

Figure 6.

α-TOS induces tumor regression in GSNOR downregulating HepG2 tumor xenografts via oxidative stress and RIP1 activation. A, qRT-PCR analyses of GSNOR in HepG2 cells transfected with scrambled shRNA (shScr) or with two shRNAs against GSNOR (shGSNOR-A and shGSNOR-B). n = 3; ***, P < 0.0001. B, Western blots of GSNOR, TRAP1, and SDHA. Immunoreactive bands densitometry is shown on the top. n = 3; *P < 0.0001. C, analyses of cell death upon staining with Alamar blue. n = 3; α-TOS 40 μmol/L: shGSNOR-A; **, P = 0.0094; shGSNOR-B **, P = 0.023; α-TOS 60 μmol/L: shGSNOR-A; ***, P = 0.0004; shGSNOR-B; ***, P = 0.0009. D, volume evaluation of shScr and shGSNOR HCC xenografts in SCID mice during 2-week treatment with 5 μmol α-TOS. All treatments started when tumor volume was 62.5 mm3 (approx. diameter of 5 mm). **, P = 0.0059; ***, P < 0.0001. E, tumor xenograft weight (left; **, P = 0.0015). Representative images of tumor xenografts (right). Histograms show the mean of data ± SEM obtained only from animals that developed tumor mass (n = 5 for untreated shScr; n = 7 for α-TOS-treated shScr; n = 6 for untreated shGSNOR; n = 9 for α-TOS–treated shGSNOR). Western blots of protein carbonyls (F) and RIP1 (G) of α-TOS–treated HepG2 xenografts. Stain-free gel (Bio-Rad) was used as loading control. Densitometry is shown at the bottom. n = 3; Carbonyls: ***, P = 0.0002; **, P = 0.0026; RIP1: ***, P = 0.0009; **, P = 0.0052.

Figure 6.

α-TOS induces tumor regression in GSNOR downregulating HepG2 tumor xenografts via oxidative stress and RIP1 activation. A, qRT-PCR analyses of GSNOR in HepG2 cells transfected with scrambled shRNA (shScr) or with two shRNAs against GSNOR (shGSNOR-A and shGSNOR-B). n = 3; ***, P < 0.0001. B, Western blots of GSNOR, TRAP1, and SDHA. Immunoreactive bands densitometry is shown on the top. n = 3; *P < 0.0001. C, analyses of cell death upon staining with Alamar blue. n = 3; α-TOS 40 μmol/L: shGSNOR-A; **, P = 0.0094; shGSNOR-B **, P = 0.023; α-TOS 60 μmol/L: shGSNOR-A; ***, P = 0.0004; shGSNOR-B; ***, P = 0.0009. D, volume evaluation of shScr and shGSNOR HCC xenografts in SCID mice during 2-week treatment with 5 μmol α-TOS. All treatments started when tumor volume was 62.5 mm3 (approx. diameter of 5 mm). **, P = 0.0059; ***, P < 0.0001. E, tumor xenograft weight (left; **, P = 0.0015). Representative images of tumor xenografts (right). Histograms show the mean of data ± SEM obtained only from animals that developed tumor mass (n = 5 for untreated shScr; n = 7 for α-TOS-treated shScr; n = 6 for untreated shGSNOR; n = 9 for α-TOS–treated shGSNOR). Western blots of protein carbonyls (F) and RIP1 (G) of α-TOS–treated HepG2 xenografts. Stain-free gel (Bio-Rad) was used as loading control. Densitometry is shown at the bottom. n = 3; Carbonyls: ***, P = 0.0002; **, P = 0.0026; RIP1: ***, P = 0.0009; **, P = 0.0052.

Close modal

Conflicting lines of evidence argue that NO can carry out anti- or protumor activities, as well as having a still not clarified role in tumorigenesis. HCC is a case in point, as iNOS is often increased in pathologies predisposing to HCC (41) but iNOS−/− mice revealed only very little effects of NO in hepatocarcinogenesis (42). In reconciling these apparently contradictory results, several pieces of evidence about the establishment of excessive S-nitrosylation in HCC due to GSNOR downregulation are accumulating (6). Moreover, human GSNOR gene is located at approximately 4q23, a region frequently subjected to chromosomal deletion in HCC (43–45) and GSNOR downregulation has been found associated with de novo hepatocarcinogenesis after tumor resection and a poor prognosis in patients with HCC (46). Finally, meta-analyses present in the Oncomine platform (Supplementary Fig. S6A) and quantitative evaluation of human HCC samples obtained by The Human Protein Atlas collection by IHC profiler (Supplementary Fig. S6B; ref. 47) show a direct correlation between GSNOR underexpression and HCC.

Here, we have characterized that GSNOR downregulation induces a rearrangement of the mitochondrial electron transfer chain. Although the downregulation of complex IV was already reported as being negatively affected by S-nitrosylation, and, for this reason, expected to occur in conditions of GSNOR deficiency, the evidence that SDH is upregulated is new and noteworthy, and may constitute a mechanism of cell adaptation. In support of this hypothesis, it should be noticed that, notwithstanding the fact that GSNOR-downregulating cells show a decreased Δψ, ATP levels are relatively preserved, indicating that a metabolic reprogramming is nevertheless activated to sustain cell energetic needs.

From a mechanistic point of view, we have demonstrated that TRAP1 S-nitrosylation at Cys501 is the causative event underlying the increase of SDH levels and activity. TRAP1 is essential for mitochondrial protein quality control (48, 49). As it preferentially accumulates in the matrix (50), it is plausible that TRAP1 could interact with SDH, mainly with subunits A and B that are exposed to this mitochondrial compartment, and, in turn, regulate SDH turnover. This hypothesis would support our data, and would also partly agree with recent results highlighting a negative correlation between TRAP1 and SDH activity (14). However, the role of TRAP1 downregulation that we have here outlined as responsible for HCC sensitivity to mitochondrial drugs, to the best of our knowledge does not represent a condition concurring to HCC onset. This would be inconsistent with the current literature that, in contrast, indicates that TRAP1 is upregulated in several cancers (51, 52). We do not currently know the kinetics of TRAP1 modulation during the complex process of hepatocarcinogenesis. We only indicate that at least in the GSNOR-downregulating subtype of HCC, TRAP1 is target of S-nitrosylation and subjected to accelerated degradation. This condition, resulting in SDH upregulation, makes it possible to envision that SDH inhibitors might selectively target this HCC subtype.

It is worthwhile noting that excessive S-nitrosylation, because of GSNOR deficiency, also has an effect on the modality of cell death induced by SDH inhibitors, with the RIP1-mediated necroptosis activated in place of apoptosis. This depends both on the high levels of ROS produced and on the incapability of GSNOR-downregulating HCC cells to correctly activate caspase-3, as caspases undergo S-nitrosylation on catalytic cysteine and inactivation of their proteolytic activity (34, 35). The incapability to correctly activate apoptosis could be, in principle, another factor that contributes to hepatocarcinogenesis in conditions of GSNOR deficiency.

Altogether, if on the one hand, our results provide the molecular basis for designing highly selective SDH-targeting drugs aimed at improving chemotherapy against HCC, on the one other they also set the stage for comprehending a redox-based regulation of TRAP1 stability.

J.S. Stamler reports receiving a commercial research grant from Novartis and has ownership interest (including patents) in Adamas and Lifehealth. No potential conflicts of interest were disclosed by the other authors.

Conception and design: S. Rizza, G. Filomeni

Development of methodology: B. Blagoev, J.S. Stamler

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Rizza, C. Montagna, E. Maiani, V. Sanchez-Quiles, B. Blagoev, D.D. Zio

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Rizza, V. Sanchez-Quiles, B. Blagoev, A. Rasola, G. Filomeni

Writing, review, and/or revision of the manuscript: S. Rizza, S. Cardaci, B. Blagoev, A. Rasola, J.S. Stamler, F. Cecconi, G. Filomeni

Study supervision: B. Blagoev, F. Cecconi, G. Filomeni

Other (performed oxygen consumption measurements): G.D. Giacomo

Other (mass spectrometry analysis): V. Sanchez-Quiles

The authors thank Vanda Turcanova for laboratory assistance; Petra Hamerlik and Elena Papaleo for assistance and discussion; and Magdalena Acuña Villa, Laila Fisher, and Martin W. Bennett for secretarial and editorial work.

This work has been supported by grants from the Danish Cancer Society (KBVU R72-A4408 to F. Cecconi; KBVU R72-A4647 to G. Filomeni); Danish Research Foundation (CARD); the Italian Association for Cancer Research, (AIRC-MFAG 2011 n.11452 to G. Filomeni; IG2013 to F. Cecconi; IG2014 n.15863 to A. Rasola); the Italian Ministry of Health (GR-2008-1138121 to G. Filomeni). We are also grateful to the Bjarne Saxhof Foundation, Lundbeck Foundation (R167-2013-16100 and R52-A4895 to F. Cecconi), NovoNordisk (7559 to F. Cecconi), FNU (DFF – 1323-00191 to B. Blagoev).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Hess
DT
,
Matsumoto
A
,
Kim
SO
,
Marshall
HE
,
Stamler
JS
. 
Protein S-nitrosylation: purview and parameters
.
Nat Rev Mol Cell Biol
2005
;
6
:
150
66
.
2.
Jia
J
,
Arif
A
,
Terenzi
F
,
Willard
B
,
Plow
EF
,
Hazen
SL
, et al
Target-selective protein s-nitrosylation by sequence motif recognition
.
Cell
2014
;
159
:
623
34
.
3.
Liu
L
,
Hausladen
A
,
Zeng
M
,
Que
L
,
Heitman
J
,
Stamler
JS
. 
A metabolic enzyme for S-nitrosothiol conserved from bacteria to humans
.
Nature
2001
;
410
:
490
4
.
4.
Liu
L
,
Yan
Y
,
Zeng
M
,
Zhang
J
,
Hanes
MA
,
Ahearn
G
, et al
Essential roles of S-nitrosothiols in vascular homeostasis and endotoxic shock
.
Cell
2004
;
116
:
617
28
.
5.
Jensen
DE
,
Belka
GK
,
Du Bois
GC
. 
S-Nitrosoglutathione is a substrate for rat alcohol dehydrogenase class III isoenzyme
.
Biochem J
1998
;
331
(
Pt 2
):
659
68
.
6.
Wei
W
,
Li
B
,
Hanes
MA
,
Kakar
S
,
Chen
X
,
Liu
L
. 
S-nitrosylation from GSNOR deficiency impairs DNA repair and promotes hepatocarcinogenesis
.
Sci Transl Med
2010
;
2
:
19ra13
.
7.
Wei
W
,
Yang
Z
,
Tang
CH
,
Liu
L
. 
Targeted deletion of GSNOR in hepatocytes of mice causes nitrosative inactivation of O6-alkylguanine-DNA alkyltransferase and increased sensitivity to genotoxic diethylnitrosamine
.
Carcinogenesis
2011
;
32
:
973
7
.
8.
Montagna
C
,
Di Giacomo
G
,
Rizza
S
,
Cardaci
S
,
Ferraro
E
,
Grumati
P
, et al
S-nitrosoglutathione reductase deficiency-induced S-nitrosylation results in neuromuscular dysfunction
.
Antioxid Redox Signal
2014
;
21
:
570
87
.
9.
Moncada
S
,
Erusalimsky
JD
. 
Does nitric oxide modulate mitochondrial energy generation and apoptosis?
Nat Rev Mol Cell Biol
2002
;
3
:
214
20
.
10.
Piantadosi
CA
. 
Regulation of mitochondrial processes by protein S-nitrosylation
.
Biochim Biophys Acta
2012
;
1820
:
712
21
.
11.
Guzzo
G
,
Sciacovelli
M
,
Bernardi
P
,
Rasola
A
. 
Inhibition of succinate dehydrogenase by the mitochondrial chaperone TRAP1 has anti-oxidant and anti-apoptotic effects on tumor cells
.
Oncotarget
2014
;
5
:
11897
908
.
12.
Montesano Gesualdi
N
,
Chirico
G
,
Pirozzi
G
,
Costantino
E
,
Landriscina
M
,
Esposito
F
. 
Tumor necrosis factor-associated protein 1 (TRAP-1) protects cells from oxidative stress and apoptosis
.
Stress
2007
;
10
:
342
50
.
13.
Xiang
F
,
Huang
YS
,
Shi
XH
,
Zhang
Q
. 
Mitochondrial chaperone tumour necrosis factor receptor-associated protein 1 protects cardiomyocytes from hypoxic injury by regulating mitochondrial permeability transition pore opening
.
FEBS J
2010
;
277
:
1929
38
.
14.
Sciacovelli
M
,
Guzzo
G
,
Morello
V
,
Frezza
C
,
Zheng
L
,
Nannini
N
, et al
The mitochondrial chaperone TRAP1 promotes neoplastic growth by inhibiting succinate dehydrogenase
.
Cell Metab
2013
;
17
:
988
99
.
15.
Yoshida
S
,
Tsutsumi
S
,
Muhlebach
G
,
Sourbier
C
,
Lee
MJ
,
Lee
S
, et al
Molecular chaperone TRAP1 regulates a metabolic switch between mitochondrial respiration and aerobic glycolysis
.
Proc Natl Acad Sci U S A
2013
;
110
:
E1604
12
.
16.
Li
WC
,
Ralphs
KL
,
Tosh
D
. 
Isolation and culture of adult mouse hepatocytes
.
Methods Mol Biol
2010
;
633
:
185
96
.
17.
Riccardi
C
,
Nicoletti
I
. 
Analysis of apoptosis by propidium iodide staining and flow cytometry
.
Nat Protoc
2006
;
1
:
1458
61
.
18.
Franken
NA
,
Rodermond
HM
,
Stap
J
,
Haveman
J
,
van Bree
C
. 
Clonogenic assay of cells in vitro
.
Nat Protoc
2006
;
1
:
2315
9
.
19.
Lemarie
A
,
Grimm
S
. 
Mutations in the heme b-binding residue of SDHC inhibit assembly of respiratory chain complex II in mammalian cells
.
Mitochondrion
2009
;
9
:
254
60
.
20.
Forrester
MT
,
Foster
MW
,
Benhar
M
,
Stamler
JS
. 
Detection of protein S-nitrosylation with the biotin-switch technique
.
Free Radic Biol Med
2009
;
46
:
119
26
.
21.
Chylek
LA
,
Akimov
V
,
Dengjel
J
,
Rigbolt
KT
,
Hu
B
,
Hlavacek
WS
, et al
Phosphorylation site dynamics of early T-cell receptor signaling
.
PLoS One
2014
;
9
:
e104240
.
22.
Akimov
V
,
Henningsen
J
,
Hallenborg
P
,
Rigbolt
KT
,
Jensen
SS
,
Nielsen
MM
, et al
StUbEx: Stable tagged ubiquitin exchange system for the global investigation of cellular ubiquitination
.
J Proteome Res
2014
;
13
:
4192
204
.
23.
Francavilla
C
,
Rigbolt
KT
,
Emdal
KB
,
Carraro
G
,
Vernet
E
,
Bekker-Jensen
DB
, et al
Functional proteomics defines the molecular switch underlying FGF receptor trafficking and cellular outputs
.
Mol Cell
2013
;
51
:
707
22
.
24.
Dong
LF
,
Jameson
VJ
,
Tilly
D
,
Cerny
J
,
Mahdavian
E
,
Marin-Hernandez
A
, et al
Mitochondrial targeting of vitamin E succinate enhances its pro-apoptotic and anti-cancer activity via mitochondrial complex II
.
J Biol Chem
2011
;
286
:
3717
28
.
25.
Frezza
C
,
Cipolat
S
,
Scorrano
L
. 
Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts
.
Nat Protoc
2007
;
2
:
287
95
.
26.
Caplan
AI
,
Greenawalt
JW
. 
The effects of osmotic lysis on the oxidative phosphorylation and compartmentation of rat liver mitochondria
.
J Cell Biol
1968
;
36
:
15
31
.
27.
Lowry
OH
,
Rosebrough
NJ
,
Farr
AL
,
Randall
RJ
. 
Protein measurement with the Folin phenol reagent
.
J Biol Chem
1951
;
193
:
265
75
.
28.
Landriscina
M
,
Laudiero
G
,
Maddalena
F
,
Amoroso
MR
,
Piscazzi
A
,
Cozzolino
F
, et al
Mitochondrial chaperone Trap1 and the calcium binding protein Sorcin interact and protect cells against apoptosis induced by antiblastic agents
.
Cancer Res
2010
;
70
:
6577
86
.
29.
Goard
CA
,
Schimmer
AD
. 
Mitochondrial matrix proteases as novel therapeutic targets in malignancy
.
Oncogene
2014
;
33
:
2690
9
.
30.
Forrester
MT
,
Thompson
JW
,
Foster
MW
,
Nogueira
L
,
Moseley
MA
,
Stamler
JS
. 
Proteomic analysis of S-nitrosylation and denitrosylation by resin-assisted capture
.
Nat Biotechnol
2009
;
27
:
557
9
.
31.
Pereira da Silva
AP
,
El-Bacha
T
,
Kyaw
N
,
dos Santos
RS
,
da-Silva
WS
,
Almeida
FC
, et al
Inhibition of energy-producing pathways of HepG2 cells by 3-bromopyruvate
.
Biochem J
2009
;
417
:
717
26
.
32.
Dong
LF
,
Low
P
,
Dyason
JC
,
Wang
XF
,
Prochazka
L
,
Witting
PK
, et al
Alpha-tocopheryl succinate induces apoptosis by targeting ubiquinone-binding sites in mitochondrial respiratory complex II
.
Oncogene
2008
;
27
:
4324
35
.
33.
Neuzil
J
. 
Vitamin E succinate and cancer treatment: a vitamin E prototype for selective antitumour activity
.
Br J Cancer
2003
;
89
:
1822
6
.
34.
Melino
G
,
Bernassola
F
,
Knight
RA
,
Corasaniti
MT
,
Nistico
G
,
Finazzi-Agro
A
. 
S-nitrosylation regulates apoptosis
.
Nature
1997
;
388
:
432
3
.
35.
Mannick
JB
,
Schonhoff
C
,
Papeta
N
,
Ghafourifar
P
,
Szibor
M
,
Fang
K
, et al
S-Nitrosylation of mitochondrial caspases
.
J Cell Biol
2001
;
154
:
1111
6
.
36.
Maejima
Y
,
Adachi
S
,
Morikawa
K
,
Ito
H
,
Isobe
M
. 
Nitric oxide inhibits myocardial apoptosis by preventing caspase-3 activity via S-nitrosylation
.
J Mol Cell Cardiol
2005
;
38
:
163
74
.
37.
van Wijk
SJ
,
Hageman
GJ
. 
Poly(ADP-ribose) polymerase-1 mediated caspase-independent cell death after ischemia/reperfusion
.
Free Radic Biol Med
2005
;
39
:
81
90
.
38.
Festjens
N
,
Vanden Berghe
T
,
Cornelis
S
,
Vandenabeele
P
. 
RIP1, a kinase on the crossroads of a cell's decision to live or die
.
Cell Death Differ
2007
;
14
:
400
10
.
39.
Krysko
DV
,
Vanden Berghe
T
,
D'Herde
K
,
Vandenabeele
P
. 
Apoptosis and necrosis: detection, discrimination and phagocytosis
.
Methods
2008
;
44
:
205
21
.
40.
Neuzil
J
,
Weber
T
,
Gellert
N
,
Weber
C
. 
Selective cancer cell killing by alpha-tocopheryl succinate
.
Br J Cancer
2001
;
84
:
87
9
.
41.
McNaughton
L
,
Puttagunta
L
,
Martinez-Cuesta
MA
,
Kneteman
N
,
Mayers
I
,
Moqbel
R
, et al
Distribution of nitric oxide synthase in normal and cirrhotic human liver
.
Proc Natl Acad Sci U S A
2002
;
99
:
17161
6
.
42.
Denda
A
,
Kitayama
W
,
Kishida
H
,
Murata
N
,
Tamura
K
,
Kusuoka
O
, et al
Expression of inducible nitric oxide (NO) synthase but not prevention by its gene ablation of hepatocarcinogenesis with fibrosis caused by a choline-deficient, L-amino acid-defined diet in rats and mice
.
Nitric Oxide
2007
;
16
:
164
76
.
43.
Patil
MA
,
Gutgemann
I
,
Zhang
J
,
Ho
C
,
Cheung
ST
,
Ginzinger
D
, et al
Array-based comparative genomic hybridization reveals recurrent chromosomal aberrations and Jab1 as a potential target for 8q gain in hepatocellular carcinoma
.
Carcinogenesis
2005
;
26
:
2050
7
.
44.
Wong
N
,
Lai
P
,
Lee
SW
,
Fan
S
,
Pang
E
,
Liew
CT
, et al
Assessment of genetic changes in hepatocellular carcinoma by comparative genomic hybridization analysis: relationship to disease stage, tumor size, and cirrhosis
.
Am J Pathol
1999
;
154
:
37
43
.
45.
Nagai
H
,
Pineau
P
,
Tiollais
P
,
Buendia
MA
,
Dejean
A
. 
Comprehensive allelotyping of human hepatocellular carcinoma
.
Oncogene
1997
;
14
:
2927
33
.
46.
Hoshida
Y
,
Villanueva
A
,
Kobayashi
M
,
Peix
J
,
Chiang
DY
,
Camargo
A
, et al
Gene expression in fixed tissues and outcome in hepatocellular carcinoma
.
N Engl J Med
2008
;
359
:
1995
2004
.
47.
Varghese
F
,
Bukhari
AB
,
Malhotra
R
,
De
A
. 
IHC Profiler: an open source plugin for the quantitative evaluation and automated scoring of immunohistochemistry images of human tissue samples
.
PLoS One
2014
;
9
:
e96801
.
48.
Bender
T
,
Lewrenz
I
,
Franken
S
,
Baitzel
C
,
Voos
W
. 
Mitochondrial enzymes are protected from stress-induced aggregation by mitochondrial chaperones and the Pim1/LON protease
.
Mol Biol Cell
2011
;
22
:
541
54
.
49.
Kriegenburg
F
,
Ellgaard
L
,
Hartmann-Petersen
R
. 
Molecular chaperones in targeting misfolded proteins for ubiquitin-dependent degradation
.
FEBS J
2012
;
279
:
532
42
.
50.
Cechetto
JD
,
Gupta
RS
. 
Immunoelectron microscopy provides evidence that tumor necrosis factor receptor-associated protein 1 (TRAP-1) is a mitochondrial protein which also localizes at specific extramitochondrial sites
.
Exp Cell Res
2000
;
260
:
30
9
.
51.
Rasola
A
,
Neckers
L
,
Picard
D
. 
Mitochondrial oxidative phosphorylation TRAP(1)ped in tumor cells
.
Trends Cell Biol
2014
;
24
:
455
63
.
52.
Agorreta
J
,
Hu
J
,
Liu
D
,
Delia
D
,
Turley
H
,
Ferguson
DJ
, et al
TRAP1 regulates proliferation, mitochondrial function, and has prognostic significance in NSCLC
.
Mol Cancer Res
2014
;
12
:
660
9
.