Abstract Acute inflammatory responses often involve the production of reactive oxygen and nitrogen species by innate immune cells, particularly macrophages. How activated macrophages protect themselves in the face of oxidative-inflammatory stress remains a long-standing question. Recent evidence implicates reactive sulfur species (RSS) in inflammatory responses; however, how endogenous RSS affect macrophage function and response to oxidative and inflammatory insults remains poorly understood. In this study, we investigated the endogenous pathways of RSS biogenesis and clearance in macrophages, with a particular focus on exploring how hydrogen sulfide (H[2]S)-mediated S-persulfidation influences macrophage responses to oxidative-inflammatory stress. We show that classical activation of mouse or human macrophages using lipopolysaccharide and interferon-γ (LPS/IFN-γ) triggers substantial production of H[2]S/RSS, leading to widespread protein persulfidation. Biochemical and proteomic analyses revealed that this surge in cellular S-persulfidation engaged ∼2% of total thiols and modified over 800 functionally diverse proteins. S-persulfidation was found to be largely dependent on the cystine importer xCT and the H[2]S-generating enzyme cystathionine γ-lyase and was independent of changes in the global proteome. We further investigated the role of the sulfide-oxidizing enzyme sulfide quinone oxidoreductase (SQOR), and found that it acts as a negative regulator of S-persulfidation. Elevated S-persulfidation following LPS/IFN-γ stimulation or SQOR inhibition was associated with increased resistance to oxidative stress. Upregulation of persulfides also inhibited the activation of the macrophage NLRP3 inflammasome and provided protection against inflammatory cell death. Collectively, our findings shed light on the metabolism and effects of RSS in macrophages and highlight the crucial role of persulfides in enabling macrophages to withstand and alleviate oxidative-inflammatory stress. Keywords: M1 macrophages, Persulfides, Oxidative stress, Inflammation, Cell death 1. Introduction Innate inflammatory responses are characterized by the production of reactive oxygen and nitrogen species (ROS and RNS) thus being closely associated with oxidative/nitrosative stress [[31]1,[32]2]. Innate immune cells, in particular macrophages, utilize ROS/RNS, such as hydrogen peroxide (H[2]O[2]) and nitric oxide (NO), to kill microbes and tumor cells. In addition to their cytotoxic function, ROS/RNS act as versatile signaling molecules in macrophages and other inflammatory cells, where they regulate diverse processes including metabolism, motility, cell division and differentiation [[33][3], [34][4], [35][5]]. Recent evidence shows that hydrogen sulfide (H[2]S) and other reactive sulfur species (RSS) are also generated during inflammatory processes [[36][6], [37][7], [38][8], [39][9]]. The evidence further suggests that similar to ROS and RNS, RSS also exert regulatory effects on cellular inflammatory responses [[40][6], [41][7], [42][8], [43][9]]. However, while the roles and mechanisms of action of innate immune cell-derived ROS/RNS have been extensively studied and well characterized, those of endogenous RSS have been less studied and characterized. In recent years, the cellular mechanisms of H[2]S synthesis and metabolism have been delineated in great detail [[44][10], [45][11], [46][12], [47][13]]. H[2]S is synthesized in cells by cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS) in the transsulfuration pathway, and 3-mercaptopyruvate sulfurtransferase (MPST), involved in cysteine (Cys) catabolism. H[2]S is primarily metabolized via the mitochondrial sulfide oxidation pathway. The first enzyme in this pathway is sulfide quinone oxidoreductase (SQOR), which catalyzes the oxidation of H[2]S to reactive persulfide species and reduces coenzyme Q (CoQ). The biochemical interplay between H[2]S and various RSS has become a topic of increased research interest. Among RSS, sulfane sulfur (S^0) species, such as hydropersulfides (RSSH) and polysulfides (RSS(n)SR, n ≥ 1), are increasingly recognized as potential biological mediators [[48][14], [49][15], [50][16]]. In this regard, evidence indicates that the above-mentioned H[2]S biosynthetic and degrading enzymes influence endogenous persulfide levels; however, the precise contribution of each enzyme remains to be fully explored, and it may vary depending on the cell type and context. Furthermore, despite their emerging importance, there is limited knowledge about the metabolism and roles of persulfides in macrophages and related cell types. Protein S-persulfidation (P-SSH), the process in which protein Cys thiols are converted to persulfides, has recently emerged as an important mechanism of H[2]S-based signaling [[51]17,[52]18]. The available evidence indicates that persulfidation of proteins serves two main roles: (1) the reversible modulation of protein function, and (2) the protection of protein thiols against overoxidation [[53][17], [54][18], [55][19], [56][20]]. Persulfidation has been shown to modulate the activity of various classes of proteins, including transcription factors, proteases, kinases/phosphatases, and others [[57]17,[58][21], [59][22], [60][23], [61][24]]. Nevertheless, the mechanisms that govern persulfide formation in cells are incompletely understood, and the cell-specific roles of protein persulfidation largely remain to be characterized. The kinetics and magnitude of S-persulfidation are determined by several factors, including cellular H[2]S fluxes and the redox milieu. Yet the exact mechanisms by which proteins undergo persulfidation in cells are not well defined and are a subject of intensive research [[62][14], [63][15], [64][16], [65][17], [66][18],[67]25]. In general, de-novo, non-enzymatic protein persulfidation is thought to occur via one of two mechanisms: (i) the reaction of oxidized Cys derivatives with sulfide, or (ii) sulfide oxidation products reacting with Cys thiols. In addition, recent findings support enzyme-catalyzed protein persulfidation [[68]26,[69]27]. In this context, the role of SQOR is of particular interest. On the one hand, SQOR maintains low levels of H[2]S and can thus limit the generation of P-SSH; conversely, SQOR-catalyzed H[2]S oxidation in itself is source of persulfides, in particular glutathione persulfide (GSSH), which could then promote P-SSH formation [[70]28]. These divergent scenarios remain to be tested in different cell types and conditions. In particular, the role of SQOR in regulating persulfidation processes in macrophages is so far unknown, and the potential functional consequences of such regulation remain to be determined. In this study, we investigated the formation, metabolism and roles of H[2]S/persulfides in M1/classically-activated macrophages. Our results reveal that activation of human and mouse macrophages induces robust H[2]S generation and widespread S-persulfidation. We provide evidence that SQOR negatively regulates endogenous S-persulfidation and show that elevated levels of persulfides are associated with enhanced macrophage survival in response to oxidant or inflammatory stress. Altogether, our findings indicate that the upregulation of persulfides in activated macrophages serves as a self-protective mechanism to mitigate and cope with oxidative and inflammatory stress. 2. Materials and methods 2.1. Antibodies and reagents The following primary antibodies were used in this study: anti-SQOR (17256-1-AP) and anti-SLC7A11/xCT (26864-1-AP) from Proteintech, anti-caspase 1 (SC-515) from Santa Cruz Biotechnology, anti-NLRP3 (ag-20b-0014) and anti-ASC (AG-25B-0006) from AdipoGen, anti-IL-1β (5129-100) from Biovision, anti-GAPDH (ab9484) from Abcam. LPS from E. coli 055:B5 (L4005), PMA (P8139), dl-propargylglycine (p7888), aminooxyacetate ([71]C13408) and sulfasalazine (S0883) were purchased from Sigma. Mouse IFN-γ (485-MI-100) and human IFN-γ (285-IF-100) were from R&D Systems. Poly (dA:dT) (tlrl-patn-1) and flagellin (tlrl-stfla) were from InvivoGen. EZ-Link Maleimide-PEG2-Biotin (21901BID), streptavidin-agarose resin (20361) and disuccinimidyl suberate (DSS) (21655) were from Thermo Scientific. The fluorescent probes WSP-5 and SSP4 were a kind gift of Prof. Ming Xian (Brown University). The MPST inhibitor I3-MT-3 was a kind gift of Prof. Kenjiro Hanaoka (Keio University, Tokyo, Japan). Tissue culture media and reagents were from Biological Industries (Beit Haemek, Israel). 2.2. Preparation of Cys persulfide solution A Cys persulfide (Cys-SSH) solution was prepared based on published protocols for persulfide synthesis by reacting cystine with Na[2]S [[72][29], [73][30], [74][31]]. In brief, freshly prepared degassed solutions of 20 mM cystine (pH 8.8) and 40 mM Na[2]S were mixed 1:1 and incubated for 30 min at 37 °C and then promptly aliquoted and frozen at −20 °C. The solution sulfane sulfur content was determined by cold cyanolysis conducted at pH 8.8 and colorimetric detection of the resulting ferric thiocyanate complex [[75]32]. Under these conditions, cystine alone did not generate any detectable sulfur product in the cold cyanolysis assay. According to literature [[76]33], the Cys-SSH solution thus prepared contains several species in metastable equilibrium, namely, Cys-SSH, Cys–SSS–Cys, cystine, Cys, and H[2]S. Absorbance measurements at 335 nm [[77]31,[78]32] indicated that Cys-SSH was the major species in the solution. Furthermore, when the solution was kept on ice, no appreciable absorbance changes were seen for at least 1 h. Regardless, the Cys-SSH solution was used immediately after the preparation or immediately after thawing from the frozen state. 2.3. Cell culture and treatments In this work, we have used human and mouse monocytes/macrophages as follows. THP-1 cells (human monocyte cell line) were obtained from American Type Culture Collection (ATCC) and maintained in RPMI 1640, supplemented with 10% (v/v) fetal bovine serum (FBS), 1% penicillin/streptomycin and 1% sodium pyruvate. The cells were sub-cultured using fresh media every 2–4 days and maintained in an incubator (37 °C, 5% CO[2]). THP-1 monocytes were differentiated to adherent macrophages (TDM) by incubation with PMA (166 nM) for 48 h at 37 °C. THP-1 cell differentiation was enhanced by removing the PMA-containing media and adding fresh media for a further 24 h. The mouse macrophage cell line J774 was obtained from ATCC and maintained in DMEM containing 10% FBS, 1% penicillin/streptomycin, 1% sodium pyruvate and 1% glutamine, at 37 °C in 5% CO[2]. Bone marrow-derived macrophages (BMDM) from 8- to 12-week-old C57BL/6 male mice were generated using established protocols. In brief, femurs of mice were flushed, and a single-cell suspension of the bone marrow was prepared in DMEM medium. The suspension was centrifuged at 1500 rpm for 5 min to pellet the cells. The cells were then resuspended in DMEM supplemented with 30% (v/v) conditioned L929 (ATCC number: CCL-1) medium at a density of 8x10^6 cells/well. On the seventh day of culture, the cells were transferred to a new plate and resuspended in DMEM supplemented with 10% FBS, 1% penicillin/streptomycin and 1% glutamine, at 37 °C in 5% CO[2]. For the induction of M1 macrophages, cells were stimulated for 24 h with LPS (0.5 μg/ml) and IFN-γ (50 ng/ml or 20 ng/ml for mouse and human cells, respectively). In order to activate the NLRP3 inflammasome, macrophages and monocytes were exposed to LPS (0.5 μg/ml) for 4 h, followed by stimulation with 10 μM nigericin or 5 mM ATP. In experiments assessing the activation of caspase-1 and IL-1β, cell stimulation was performed in serum-free-medium. In order to activate the NLRC4 and AIM2 inflammasomes LPS-primed macrophages were transfected with flagellin (1 μg/ml) or poly (dA:dT) (2 μg/ml), respectively, using Lipofectamine RNAiMAX (Thermo Fisher Scientific). Procaspase-1 and pro-IL-1β in the cell extracts and their active forms secreted in the culture supernatant precipitated with 10% trichloroacetic acid were analyzed by immunoblotting. 2.4. Measurement of intracellular cysteine The intracellular Cys content was determined by Gaitonde's acid ninhydrin method [[79]34]. In brief, after treatments, the cells were washed with cold PBS, and then 5% sulfosalicylic acid (SSA) was added to each well. The cells were collected and centrifuged for 15 min at 15000 g. The pellets were used for protein quantification and the supernatants were used for the assay. The pH of SSA supernatant was adjusted to 8.3 using NaOH and then the samples were reduced with 5 mM DTT for 15 min. Next, 0.1 ml of glacial acetic acid was added to 100 μl sample followed by addition of 100 μl of acid ninhydrin reagent. After heating (100 °C for 10 min) and cooling, the samples were diluted 1:2 with 100% ethanol. Sample absorbance was measured at 560 nm, and Cys levels were quantified using Cys standards dissolved in 5% SSA and processed in the same manner as the samples. 2.5. Measurement of intracellular H[2]S, sulfane sulfur and ROS Intracellular H[2]S and S^0 levels were evaluated using the fluorescent dyes WSP-5 and SSP4, respectively. After treatments, the cells were loaded with the probes (50 μM WSP-5 or 20 μM SSP4) in the presence of 500 μM hexadecyltrimethylammonium bromide (CTAB) for 30 min (for WSP-5) or 15 min (for SSP4), at 37 °C, in the dark. Thereafter, the cells were washed with PBS and transferred to FACS tubes for analysis. The fluorescence intensity was acquired by LSRFortessa™ (BD Biosciences) and flow cytometry data were analyzed using FCS express software. For fluorescence quantification, samples were acquired in triplicate, and 10,000 events were registered from each sample. Intensity of specific fluorescence was expressed as the median fluorescence intensity (MFI), from the average of at least three repeated experiments. To provide a general assessment of ROS, the cell permeable fluorescent dye, 2′,7′-dichlorofluorescin diacetate (H2DCFDA) was employed as detailed before [[80]35]. In brief, the cells were loaded with 10 μM H2DCFDA and incubated for 60 min at 37 °C. After a wash step, the cells were exposed to treatments in serum-free and phenol red-free medium and fluorescence was detected using a microplate reader at 493 nm excitation and 523 nm emission. 2.6. H[2]S consumption assay After treatments, cells were washed twice with PBS and then collected in 1 ml of buffer A (PBS/20 mM HEPES, 5 mM d-glucose, pH 7.5) into pre-weighed microfuge tubes. Then, the cells were centrifuged (1,700 g, 5 min, 4 °C), the supernatants were removed and cell pellets were resuspended in 20 vol of buffer A. The cell suspensions were then incubated with fresh Na[2]S solution (100 μM) prepared in 0.2 M HEPES (pH 7.5) and incubated at 37 °C for 15 min. Thereafter, the cells were centrifuged (3,000 g, 1 min, 4 °C) and 50 μl of the supernatant was mixed with 50 μl of 25 μM 7-Azido-4-Methylcoumarin (AzMC) fluorescent dye (Sigma, 802409) in a 96-well microplate and incubated for 30 min at room temperature. Finally, the samples were analyzed using fluorescence plate reader (Ex: 356 nm, Em: 450 nm). The sulfide concentrations were determined by comparison of the fluorescence intensity to a standard curve prepared using Na[2]S (0–100 μM). 2.7. Assessment of protein persulfidation using the biotin thiol assay The BTA was performed according to the original protocol of Gao et al. [[81]36] with minor modifications. In brief, whole cell lysates were prepared using RIPA buffer (100 mm Tris-HCl, 150 mm NaCl, 1 mm EDTA, 0.5% Triton X-100, 0.5% deoxycholic acid, with protease inhibitors, pH 7.5) and protein concentrations were adjusted to 0.8 mg/ml. A total of 1 mg protein was used for each reaction. Thiol and persulfide groups were modified with EZ-Link Maleimide-PEG2-Biotin at a final concentration of 0.1 μM for 30 min at room temperature. After acetone precipitation, the protein pellets were washed with 70% cold acetone three times, and then resuspended in resuspension buffer (50 mM Tris–HCl, 150 mM NaCl, 1 mM EDTA, 0.5% Triton X-100, 1% SDS, pH 7.5). Thereafter, biotinylated proteins were captured by streptavidin-agarose resin overnight at 4 °C. After extensive washes of the beads, disulfide-linked proteins (formerly P-SSH) were eluted with 20 mm DTT for 30 min at room temperature as described in detail in Ref. [[82]36]. Eluates were subjected to SDS-PAGE or immunoblotting as indicated in the figures. 2.8. Sample preparation for proteomic analysis Persulfide and total proteome analysis was performed using THP-1 cells to allow for the scalability needed for a proteome-wide analysis. Persulfidated proteins were isolated using the BTA method detailed above. The proteins in the gel were reduced with 3 mM DTT (60 °C for 30 min), modified with 10 mM iodoacetamide in 100 mM ammonium bicarbonate (in the dark, room temperature for 30 min) and digested in 10% acetonitrile and 10 mM ammonium bicarbonate with modified trypsin (Promega) at a 1:10 enzyme-to-substrate ratio, overnight at 37 °C. An additional second trypsinization was done for 4 h at a 1:20 enzyme-to-substrate ratio. 2.9. Liquid chromatography–MS/MS The tryptic peptides obtained above were desalted using C18 tips (homemade stage tips) and were subjected to MS analysis, as follows. The peptides were resolved by LC-MS/MS using a Q Exactive HF mass spectrometer (Thermo) fitted with a capillary HPLC (Evosep) with EV1137 column, ID: 150 μm, ReproSil-Pur C18, 1.5 μm beads (Dr Maisch, GmbH, Germany). The peptides were separated with the built-in 15 samples per day method (88 min gradient). MS was performed in a positive mode using repetitively full MS scan (m/z 300–1,500, resolution 120,000 for MS1 and 15,000 for MS2) followed by 20 data-dependent MS/MS scans at 27% normalized collision energy (HCD). The automatic gain control settings were 3x10^6 for survey MS scans and 1x10^5 for MS/MS scans. A dynamic exclusion list was enabled with exclusion duration of 20 s. Tryptic peptides from input samples were resolved by reverse-phase chromatography on 0.075 X 180-mm fused silica capillaries (J&W) packed with Reprosil reversed phase material (Dr Maisch GmbH, Germany). The peptides were eluted with linear 180-min gradient of 5–28% acetonitrile with 0.1% of formic acid followed by 15-min gradient of 28–95% and 25 min at 95% acetonitrile with 0.1% formic acid in water at flow rates of 0.15 μl/min. MS/MS analysis was performed using the Q Exactive HFX mass spectrometer (Thermo) in a positive mode using the same settings as above, expect for selecting the 30 most dominant ions from the first MS scan. 2.10. Proteomic data analysis The mass spectrometry data were analyzed using the MaxQuant software (version 1.5.2.8) [[83]37,[84]38] for peak picking and identification using the Andromeda search engine, searching against the human UniProt database with mass tolerance of 6 ppm for the precursor masses and 20 ppm for the fragment ions. Oxidation on methionine and protein N-terminus acetylation were accepted as variable modifications and carbamidomethyl on Cys was accepted as static modifications. Minimal peptide length was set to 7 amino acids and a maximum of two miscleavages was allowed. Peptide- and protein-level false discovery rates (FDRs) were filtered to 1% using the target-decoy strategy. Protein tables were filtered to eliminate the identifications from the reverse database, and common contaminants. Statistical analysis was performed using the Perseus software platform (version 1.6.7.0) [[85]39]. Proteins showing at least 4-fold change of intensities between conditions with p-value below 0.05 were considered as differentially persulfidated. A smaller list of differentially persulfidated proteins (p < 0.01) were subjected to bioinformatic analysis. Functional enrichment analysis was performed using Matecaspe ([86]http://metascape.org) with the Custom Analysis module (Minimum Overlap: 3, p-value Cutoff: 0.01, Minimum enrichment: 1.5) [[87]40]. The enrichment background list included a total of 3717 genes, obtained from the total proteome analysis. 2.11. Assessment of cellular persulfide content using a lead acetate-based assay Cells were extracted with NP-40 containing hypotonic buffer (10 mM HEPES, 10 mM KCl, 0.1 mM EDTA, 3 mM MgCl[2], 0.5 % NP-40, with protease inhibitors, pH 7.5), centrifuged to remove cellular debris, and diluted to 2 mg/ml using HEN buffer (25 mM HEPES, 1 mM EDTA, 10 μM neocuproine, pH 7.5). Next, equal amount of samples (∼3 mg protein in 600 μL) were incubated with 0.1% MMTS for 15 min at room temperature with rotation. Thereafter, protein samples and Cys-SSH standard solutions were placed in a round bottom deep-well plate and DTT (1 mM) was added to each well, followed by a 2-h incubation at 37 °C with gentle shaking. The released sulfide was subsequently detected and quantified using a lead acetate embedded filter paper, essentially as described by Hine and Mitchell [[88]41]. In some experiments, low- and high-mass persulfides were isolated by centrifugal filtration of lysates using an Amicon Ultra-4 10 kDa centrifugal concentrator (Millipore), where the low-mass fraction represented the flow through and high-mass fraction was the retentate. The sample fractionation was performed essentially as described elsewhere [[89]42]. 2.12. siRNA knockdown Knockdown of SQOR was accomplished by electroporation of small interfering RNA (siRNA) oligos. The siRNA oligos were purchased from Horizon Discovery and included the following: ON-TARGETplus Non-targeting Control Pool (D-001210-02) and ON-TARGETplus Human SQOR siRNA (J-008271-10 and J-008271-12). The siRNA oligos (3 μM) were electroporated into 4x10^6 THP-1 cells in 100 μL Opti-MEM (275 V, 1.5 ms, NEPA21, Nepa Gene Co., Ltd., Japan). Analyses were performed 72 h after electroporation. In some experiments, a second electroporation was conducted after 24 h, and cells were then cultured for 48 h. 2.13. Assessment of cell death Cells were plated at 40,000 cells/well in 96-well plates and grown overnight. After a medium change, the cells were treated as indicated in the figures legends. Cell death was evaluated using lactate dehydrogenase (LDH) release assay, which is based on the principle that upon permeabilization of plasma membrane, LDH, which is a cytosolic enzyme, is released into the culture supernatants. LDH release into the medium was measured using the Cytotox96 cytotoxicity kit as per manufacturer instructions (Promega). 2.14. Assessment of ASC oligomerization and speck formation ASC oligomerization was analyzed using a method adapted from Shi et al. [[90]43]. After treatment with indicated stimuli, cells were washed with cold PBS, resuspended in a PBS buffer containing 1% NP-40 and EDTA-free protease inhibitor cocktail, and lysed by shearing 10 times through a 21G needle. Nuclei and unlysed cells were removed by centrifugation at 250g for 5 min. The cell lysates were then centrifuged at 6000 g for 15 min at 4 °C, and the pellets and supernatants were used as the NP-40-insoluble fractions and NP-40-soluble fractions, respectively. For the detection of ASC oligomerization, the NP-40-insoluble pellets were washed twice with PBS and then were resuspended in 300 μl of the lysis buffer. The resuspended pellets underwent crosslinkage for 30 min at room temperature with 2 mM disuccinimidyl suberate (DSS) and then were centrifuged at 3500 g for 5 min at 4 °C. The supernatants were discarded and the pellets were dissolved in 2X SDS sample buffer. Finally, the samples were boiled at 95 °C for 10 min, centrifuged at 16,000 g for 2 min, and the supernatants were used for immunoblotting with anti-ASC antibody. For ASC speck formation, macrophages were seeded on chamber slides and allowed to attach overnight. The next day, the cells were treated with indicated stimulators. The cells were then fixed in 4% paraformaldehyde followed by permeabilization with 0.3% Triton X-100 for 10 min. The slides were blocked with 5% bovine serum albumin, followed by ASC and DAPI staining. Images were acquired on a Confocal LSM 880 microscope. 2.15. Statistics All data are presented as the mean ± S.D. Statistical differences were analyzed by one-way analysis of variance (ANOVA) using Prism software (GraphPad). A p-value <0.05 was considered statistically significant. 2.16. Data availability The mass spectrometry proteomics data have been deposited to the PRIDE Archive ([91]http://www.ebi.ac.uk/pride/archive/) via the PRIDE partner repository with the data set identifier PXD044043. 3. Results 3.1. Enhanced H[2]S/RSS production in M1-activated monocytes/macrophages We initially set out to examine the biosynthesis of H[2]S/RSS in response to stimulation of macrophages with lipopolysaccharide and interferon-γ (LPS/IFN-γ), which drives classical activation of macrophages and polarizes them towards an M1-like phenotype [[92]44]. We first measured levels of Cys, a major precursor of H[2]S/RSS, under basal conditions and in response to LPS/IFN-γ stimulation. We found that Cys levels were significantly increased (∼2 fold) in LPS/IFN-γ stimulated THP-1 cells (human monocytes) or J774 cells (mouse macrophages) compared to non-stimulated cells ([93]Fig. 1A). This effect was abolished by cotreatment with sulfasalazine (SSZ), an inhibitor of the cystine transporter, xCT ([94]Fig. 1A). We also observed that xCT expression was increased in response to LPS/IFN-γ stimulation of these cells ([95]Fig. 1B). Next, we used the fluorescent probes WSP-5 and SSP4 to assess cellular levels of H[2]S and S^0 species, respectively [[96]45,[97]46]. We found that LPS/INF-γ stimulation triggered a significant increase in H[2]S and S^0 levels in THP-1 cells, THP-1-derived macrophages (TDM), J774 macrophages and primary bone marrow derived mouse macrophages (BMDM) ([98]Fig. 1C and D and [99]Supplementary Fig. S1). Fig. 1. [100]Fig. 1 [101]Open in a new tab Increased generation of reactive sulfur species in LPS/IFN-γ-activated monocytes/macrophages. (A) THP-1 monocytes or J774 macrophages were either left untreated or treated for 24 h with LPS/IFN-γ in the absence or presence of sulfasalazine (SSZ), followed by determination of intracellular Cys content. (B) THP-1 or J774 cells were treated for 24 h with LPS/IFN-γ followed by analysis of xCT expression by immunoblotting. (C) THP-1 monocytes, THP-1-derived macrophages (TDM), J774 cells, or bone-marrow-derived macrophages (BMDM) were either left untreated or treated for 24 h with LPS/IFN-γ and cellular H[2]S levels were assessed by fluorescent dye WSP-5 staining followed by flow cytometry analysis. (D) Cells were treated as in C and S^0 levels were assessed by fluorescent dye SSP4 staining followed by flow cytometry analysis. Graphs (A, C, D) show mean ± SD (n ≥ 3). MFI, mean fluorescence intensity. 3.2. Widespread protein S-persulfidation in M1-activated macrophages We next focused our attention to H[2]S-mediated protein modification, i.e., persulfidation. Using the biotin thiol assay (BTA) of persulfidation analysis [[102]36] we observed a substantial increase in protein persulfidation in M1-activated THP-1 cells and TDM ([103]Fig. 2A). BTA reactions without DTT were performed in parallel as a negative control for assay specificity and non-specific binding ([104]Fig. 2A). Using TDM and J774 macrophages, we next examined the effects of several inhibitors of enzymes/proteins that may affect cellular persulfide formation. We found that macrophage treatment with the CSE inhibitor dl-propargylglycine (PAG) markedly lowered protein persulfidation, while in comparison, the CBS inhibitor aminooxyacetate (AOAA) and the MPST inhibitor (I3-MT-3) exerted only minor effects ([105]Fig. 2B). Notably, the xCT inhibitor SSZ greatly decreased protein persulfidation in the activated macrophages ([106]Fig. 2B). Taken together, the results thus far indicate that LPS/IFN-γ activation of macrophages leads to increased cystine import and intracellular Cys pools, followed by CSE-dependent persulfidation of a large number of proteins. Fig. 2. [107]Fig. 2 [108]Open in a new tab Enhanced S-persulfidation in LPS/IFN-γ-activated macrophages. (A) THP-1 monocytes or THP-1-derived macrophages (TDM) cells were either left untreated or treated for 24 h with LPS/IFN-γ. Thereafter, protein persulfidation was analyzed using the biotin thiol assay (BTA). (B) TDM or J774 cells were either left untreated or treated for 24 h with LPS/IFN-γ ± inhibitors and thereafter protein persulfidation was analyzed using BTA. The inhibitors used were dl-propargylglycine (PAG, 3 mM), I3-MT-3 (10 μM), aminooxyacetate (AOAA, 1 mM) or sulfasalazine (SSZ, 400 μM). (C) Schematic illustration of the assay for measuring total cellular persulfide content. In this assay, thiols and persulfides are first capped with S-methyl methanethiosulfonate (MMTS). The samples are then transferred to microwells and incubated with DTT. Sulfide released from the nascent Cys-S-S-Me groups is detected using lead-acetate paper. (D) TDM were either left untreated or treated with LPS/IFN-γ (24 h) or with 50 μM Cys-SSH (15 min). Persulfide content in the cell lysates was determined using the lead acetate-coupled assay depicted in C. (E) Lysates were prepared and analyzed as in D. Size separation was performed using a centrifugal filter with a molecular mass cutoff of 10,000 Da. We sought to obtain a more quantitative assessment of cellular persulfide content in resting/activated macrophages. To this end, we developed a simple biochemical assay (schematically depicted in [109]Fig. 2C) in which thiols and persulfides are initially blocked with S-methyl methanethiosulfonate (MMTS), followed by DTT treatment and capture of the released sulfide by lead acetate paper (see Material and methods for further details). Using this assay in TDM, we observed that persulfide levels were essentially undetectable in lysates obtained from untreated cells; in contrast, they were readily detected in samples from LPS/IFN-γ-stimulated cells or from cells treated with the persulfidating agent Cys persulfide (Cys-SSH) ([110]Fig. 2D). Using Cys-SSH standard solutions, we then calculated the persulfide content to be 3.25 ± 1.2 nmol/mg protein in the LPS/IFN-γ samples. It was previously reported that total thiol content in these cells is ∼180 nmol/mg [[111]35,[112]47]; therefore, from these results we estimate that in the activated TDM nearly 2% of thiols have undergone persulfidation. The level of persulfidation in LPS/IFN-γ-challenged cells was comparable to that of Cys–SSH–treated cells ([113]Fig. 2D). Using the same methodology in J774 cells, we estimated the persulfide content to be 1.1 ± 0.4 nmol/mg and 4.0 ± 0.25 nmol/mg in the control and LPS/IFN-γ samples, respectively. It should be considered that the Cys-SSH solution used in our experiments is a mixture of several species and likely contains some amount of cystine, Cys and Na[2]S (see further comments in the Materials and methods section). In control experiments we observed that cell exposure to Na[2]S, cystine or Cys did not detectibly increase S-persulfidation levels ([114]Supplementary Figs. S2A and B), supporting the conclusion that Cys-SSH was responsible for the above mentioned effects. However, it remains possible that polysulfides present in the Cys-SSH solution [[115]33] may partially contribute to the observed effects. It is likely that in cells, P-SSH are in equilibrium with low mass persulfides such as Cys-SSH and GSSH; however, the nature of this equilibrium is not established. Using ultrafiltration fractionation, we found that the majority (over 75%) of the RSSH could be ascribed to molecules of high mass (>10,000 Da; [116]Fig. 2E). While the determination of individual low mass persulfides in macrophages awaits further investigation, these results nonetheless suggest that cellular RSSH are mostly associated with protein rather than small thiol. 3.3. Large-scale analysis of the macrophage persulfide proteome We next set out to identify protein targets of persulfidation in resting and LPS/IFN-γ-stimulated cells. For this purpose, we performed a proteomic analysis of persulfidation, employing BTA enrichment coupled to LC-MS/MS analysis (see Materials and methods for details). In parallel, a global proteome analysis was conducted. These experiments were performed using THP-1 cells to allow for the scalability needed for a proteome-wide analysis. LC-MS/MS analysis of three biological replicates resulted in the identification of 3818 proteins (in the BTA pulldown) across the treatment groups. A total of 2849 proteins were included in the final analysis following filtration for proteins found in at least two out of three replicates in at least one experimental group ([117]Table S1). In addition, the proteins in the starting lysate were subjected to LC-MS/MS analysis. In the whole-cell proteome, a total of 3927 proteins were identified across the samples; of these, 3717 proteins were found in at least two out of three replicates in at least one experimental group and were retained for further analysis ([118]Table S2). Next, a more detailed analysis of the persulfide and global proteomes was undertaken in order to identify differentially persulfidated or expressed proteins (see Materials and methods section), the results of which are summarized in the volcano plots shown in [119]Fig. 3A and [120]Supplementary Fig. S3A. To determine significant differences in protein persulfidation/expression, we applied a threshold of log2 fold-change >2 and p-value <0.05 (t-test). This analysis demonstrated the significant and widespread effect of LPS/IFN-γ stimulation on persulfidation, with ∼800 proteins displaying significantly increased persulfidation ([121]Fig. 3A). In sharp contrast, we did not detect proteins displaying significantly decreased persulfidation. Compared to the persulfide proteome, changes in the global proteome were rather small, with only 40 proteins identified as upregulated and 14 as downregulated in response to LPS/IFN-γ stimulation (log2 fold-change >2; p < 0.05) ([122]Supplementary Fig. S3A). These latter results are in good agreement with previous studies that analyzed the total proteome of M1-activated monocytes/macrophages, particularly considering that the experimental conditions as well as the MS analysis parameters were not identical among the different studies [[123][48], [124][49], [125][50]]. In particular, there is a substantial overlap between proteins identified as being upregulated in M1-activated cells in our study and the previous studies ([126]Supplementary Fig. S3A). Notably, our finding that the redox proteome is more dynamic than the total proteome extends recent work by us and others [[127]35,[128]51]. Fig. 3. [129]Fig. 3 [130]Open in a new tab Alterations in the persulfide proteome following exposure of THP-1 cells to LPS/IFN-γ. (A) Volcano plot visualization of the persulfidation profile of 2849 unique proteins following cell treatment with LPS/IFN-γ. Proteins with significantly increased persulfidation (log2 fold change >2; adjusted p-value <0.05) are highlighted in red. (B) Highly enriched biological processes of the LPS/IFN-γ-regulated persulfide proteome as determined by Metascape. The categories with gene count >6 are shown in ascending order of p-values. (C) Metascape visualization of the interactome network of the LPS/IFN-γ-regulated persulfide proteome. (For interpretation of the references to color in