- Research
- Open access
- Published:
Increased TSPO alleviates neuropathic pain by preventing pyroptosis via the AMPK-PGC-1α pathway
The Journal of Headache and Pain volume 26, Article number: 16 (2025)
Abstract
Neuropathic pain poses a significant clinical challenge, largely due to the incomplete understanding of its molecular mechanisms, particularly the role of mitochondrial dysfunction. Bioinformatics analysis revealed that pyroptosis and inflammatory responses induced by spared nerve injury (SNI) in the spinal dorsal horn play a critical role in the initiation and persistence of neuropathic pain. Among the factors involved, TSPO (translocator protein) emerged as a key regulator. Our experimental findings showed that TSPO expression was upregulated during neuropathic pain, accompanied by mitochondrial dysfunction, specifically manifested as impaired mitochondrial biogenesis, disrupted mitochondrial dynamics (including insufficient expression of mitochondrial biogenesis and fusion-related proteins, as well as significantly increased expression of fission-related proteins), and activation of pyroptosis. Pharmacological upregulation of TSPO, but not its downregulation, effectively alleviated SNI-induced pain hypersensitivity, improving mitochondrial function and reducing pyroptosis. Immunofluorescence staining confirmed that TSPO was primarily localized in astrocytes, and its expression mirrored the protective effects on mitochondrial health and pyroptosis prevention. PCR array analysis suggested a strong association between TSPO and the mitochondrial regulation pathway AMPK-PGC-1α. Notably, inhibition of AMPK-PGC-1α abolished TSPO effects on mitochondrial balance and pyroptosis suppression. Furthermore, Mendelian randomization analysis of GWAS data indicated that increased TSPO expression was linked to pain relief. Through drug screening, molecular docking, and behavioral assays, we identified zopiclone as a promising TSPO-targeting drug for pain treatment. In summary, this study enhances our understanding of the molecular interplay between TSPO, mitochondrial health, and neuropathic pain, highlighting TSPO as a potential therapeutic target for pain management.
Graphical Abstract

Background
Neuropathic pain is a chronic condition caused by nerve injury or disease, characterized by hyperexcitability in somatosensory nociceptive circuits that severely affects quality of life [1, 2]. Despite extensive research, effective treatments for this debilitating condition remain limited in clinical practice [3]. A key feature of neuropathic pain is central sensitization, driven by maladaptive synaptic plasticity in the spinal dorsal horn, which, together with widespread neuroinflammation, amplifies pain hypersensitivity and facilitates pain spread [4, 5]. Throughout neuropathic pain progression, activated glial cells release cytokines and other inflammatory mediators, intensifying neuroinflammation and reinforcing central sensitization [4]. This interaction between central sensitization and neuroinflammation not only exacerbates pain but also makes it more challenging to control. Therefore, identifying therapeutic targets that can reduce neuroinflammation and modulate glial cell function may be critical to effectively managing neuropathic pain.
Emerging evidence suggests that pyroptosis, an inflammatory form of programmed cell death, plays a significant role in the development of neuropathic pain [6,7,8,9]. Mitochondria serve as central regulators of pyroptosis: under pathological conditions, damaged mitochondria release reactive oxygen species (ROS), amplifying pyroptosis and promoting inflammation. Furthermore, activated gasdermin D (GSDMD) forms pores in the mitochondrial membrane, leading to the release of pro-inflammatory mitochondrial contents, including cytochrome c and mitochondrial DNA (mtDNA), thereby disrupting mitochondrial integrity and creating a cycle of damage and inflammation [10]. Maintaining mitochondrial health is essential, as these organelles rely on biogenesis, fusion, and fission to meet cellular energy needs and maintain their structure. Disruption of these dynamics impairs mitochondrial function, increases ROS production, and leads to mtDNA damage, further intensifying pyroptosis. Pathways such as AMPK-PGC-1α regulate mitochondrial biogenesis and maintain mitochondrial dynamics, and dysregulation of these pathways exacerbates mitochondrial damage [10].
The 18 kDa translocator protein (TSPO), formerly known as the peripheral benzodiazepine receptor (PBR), is a high-affinity binding site for benzodiazepine drugs located predominantly on the outer mitochondrial membrane. Unlike central benzodiazepine receptors, TSPO exhibits distinct pharmacological and structural characteristics [11]. Discovered in rat kidneys in 1977, TSPO was subsequently identified in the brains of mammals, where it is expressed at low levels, predominantly in microglial cells, astrocytes, endothelial cells, vascular smooth muscle cells, and peripheral macrophages that infiltrate the central nervous system [12,13,14]. TSPO plays a pivotal role in inflammatory responses, particularly neuroinflammation, and serves as an important biomarker for brain inflammation imaging techniques such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) [15,16,17]. It is also implicated in various inflammatory conditions, including retinal neuroinflammation [18], post-hemorrhagic brain inflammation [19], heart dysfunction [20], and liver inflammation [21, 22]. In addition, TSPO is critical in mitochondrial functions, such as the regulation of oxidative stress, iron homeostasis, and cholesterol transport [22]. It is involved in processes like apoptosis, cell death, inflammation, and immune responses [13, 23, 24]. TSPO deficiency can result in mitochondrial dysfunction, contributing to hypoxia, angiogenesis, and a metabolic shift toward glycolysis in glioblastoma [25]. Furthermore, TSPO activation has been shown to alleviate pain in various neuropathic pain models [26, 27].
Despite these insights, the precise mechanisms by which TSPO regulates inflammation remain incompletely understood, and further research is needed to fully elucidate its role in these processes. In this study, through bioinformatics analysis of spinal dorsal horn tissue following SNI, we identified pyroptosis, especially TSPO closely associated with SNI-induced neuropathic pain. Experimentally, we found that TSPO was upregulated after SNI, along with mitochondrial dysfunction as well as pyroptosis. Pharmacological increase but not decrease of TSPO alleviated neuropathic pain, along with improved mitochondrial health and pyroptosis. By immunofluorescence staining, TSPO was mainly located in astrocytes. Upregulation of TSPO specific in astrocytes mirrored the protective effects. PCR array analysis showed that TSPO supported mitochondrial biogenesis and regulated mitochondrial fusion and fission via the AMPK-PGC-1α pathway. Intriguingly, Mendelian randomization analysis indicated that increase of TSPO was linked to pain relief. Therefore, to facilitate clinical translation, we identified TSPO-targeted drugs, zopiclone, through drug databases, highlighting the potential of repurposing existing drugs for pain treatment.
Materials
Data processing and download of the SNI dataset
To examine the causal mechanisms linked to SNI, we obtained the microarray dataset GSE18803 from the Gene Expression Omnibus (GEO) [28]. We carried out differential expression analysis to identify differentially expressed genes (DEGs) between SNI samples (n = 6) and control samples (n = 6) using the “limma” R package [29], applying significance thresholds of P < 0.05 and |log2(fold change, FC)|> 1. We generated a volcano plot and heatmap using R 4.0.3 software. Enrichment analyses for Gene Ontology (GO) and KEGG pathways were performed utilizing the ‘clusterProfiler’ R package [30]. Additionally, gene set enrichment analysis (GSEA) was executed with GSEA 4.1.0 software, referencing gene sets from the MSigDB database. We identified a total of 1,576 mitochondria-related genes (Mito-RGs) from the MSigDB [31]. Using the “WGCNA” R package, we constructed a co-expression network based on gene expression data from GSE18803, designating SNI and control samples as trait data [32]. Sample clustering was conducted with the hclust function to detect outliers, utilizing the parameter “method = average.” We approximated the optimal soft threshold for establishing a scale-free network and employed a dynamic shear tree algorithm to segment modules. Correlation analysis helped identify SNI-associated modules, and SNI genes (SNIGs) were derived through Module Membership (MM) and Gene Significance (GS) analyses.
Drugs and reagents
4',6-Diamidino-2-phenylindole (DAPI) was sourced from Beyotime Biotechnology (China). Ro 5–4864 (4'-Chlorodiazepam; MW: 319.19; purity: ≥ 99.19%) [33], PK 11195 (RP 52028; MW: 352.86; purity: ≥ 99.05%) [34], Compound C (MW: 399.49; purity: ≥ 98.14%) [35] and Zopiclone (N-Desmethyl zopiclone-d8,MW:374.78; purity: ≥ 99.79%) [36] were obtained from MedChem Express (USA). Lipopolysaccharide (LPS, L4391) was obtained from Sigma. DMEM was obtained from GIBCO/BRL, Life Technologies (Carlsbad, CA, USA). Donkey anti-Rabbit IgG H&L (Alexa Fluor® 488), anti-Mouse IgG H&L (Alexa Fluor® 546), and anti-Goat IgG H&L (Alexa Fluor® 546) were obtained from Invitrogen (United Kingdom; Cat # A-31573, A-31571, and A-21447). Primary antibodies against AMPK, NLRP3, p-AMPK, TFAM, c-Fos, and Cleaved Gasdermin D were sourced from Cell Signaling Technology (USA; Cat# 2532, 15,101, 50,081, 7495, 4384, and 32,112). An anti-TSPO antibody was obtained from Novus Biologicals (USA; Cat# NB100-41398). Antibodies against NeuN, GFAP, and Iba1 were obtained from Abcam (United Kingdom; Cat# ab104224, ab7260, and ab178846). Antibodies against GAPDH and Caspase-1 were obtained from Proteintech Group (USA; Cat# 10,494–1 and 22,915–1). Antibodies against DRP1, PGC-1α, and MFN2 were obtained from Affinity Biosciences (USA; Cat# DF7037, AF5395, and DF8106).
Animals and ethics statement
Adult male C57BL/6 J mice, with an average weight of 20 ± 2.5 g, were obtained from Liaoning Changsheng Biotechnology in Liaoning, China. The mice were maintained on a 12-h light–dark cycle (from 8 a.m. to 8 p.m.) and had continuous access to food and water. All animal studies were conducted in compliance with the US National Institutes of Health (NIH) Guide for the Care and Use of Laboratory Animals and received approval from the Jilin University Administration Committee of Experimental Animals (Approval No. 20230609–01). Testing was performed in a double-blinded manner.
Spared nerve injury (SNI) model
The SNI surgery was conducted according to the method previously outlined [37]. In summary, the animals were induced with isoflurane anesthesia at 2.5% and maintained at 2%. A surgical incision was made in the left lateral thigh to access the sciatic nerve, which was carefully separated from the surrounding muscle to expose its main trunk and three branches. SNI was induced by ligating and cutting segments of the common peroneal and tibial nerves measuring 2–4 mm. In the sham-operated mice, the sciatic nerve and its branches were exposed, but no further intervention was performed.
Von Frey filament assay
The sensitivity of the mechanical paw withdrawal threshold (PWT) in mice was examined using calibrated von Frey filaments (Stoelting, Wood Dale, IL). These filaments were positioned perpendicular to the plantar surface of the hind paw and pressed with adequate force to create a bend. A withdrawal of the paw occurring within 4 s was categorized as a positive response. If the initial filament did not elicit a response, the filament with the next higher force was tested; if a response was observed, the filament with the next lower force was applied instead. Filaments ranging from 0.04 to 1.4 g were utilized for this assessment. The force needed to achieve a 50% probability of a withdrawal response was calculated employing the "up-down" technique. PWT measurements were recorded before the SNI procedure (day -1) and on postoperative days 3, 7, 14, 21, and 28.
Astrocyte-specific TSPO constructs (AS-TSPO)
Recombinant adeno-associated viruses (rAAVs) designed to express enhanced green fluorescent protein (EGFP) and TSPO were utilized for both broad-spectrum and astrocyte-specific overexpression of TSPO. These viruses were produced by Heyuan Biotechnology Co., Ltd (OBIO, Shanghai, China), with the following stock titers: pCAAV-CMV-Tspo-linker-EGFP-3 × FLAG-WPRE at 4.4 × 1012 and pAAV-GfaABC1D-Tspo-linker-EGFP-3 × FLAG-WPRE also at 4.4 × 1012.
Virus vector delivery
The AAV vector was administered into the spinal dura mater of anesthetized mice, adhering to a previously established protocol [38]. The target vertebral column was identified through palpation, and the paraspinal muscles on the left side of the T13-L1 intervertebral space were excised, followed by a partial laminectomy to facilitate injection. Using a micro syringe (RWD 79013, Shenzhen, China), 0.6 μL of AAV was injected at a rate of 50 nL/min. A stereotaxic apparatus (RWD 69100, Shenzhen, China) was employed to position the injection needle approximately 500 μm from the midline, with the tip advanced into the spinal dorsal horn at a depth of 250 μm from the dorsal root entry zone surface. The needle was retained in the spinal cord for 10 min prior to being removed. The incision was closed with interrupted sutures, and iodine disinfectant was applied to the area.
Tissue extraction
For the Western blot (WB) experiments, we evaluated the expression changes of TSPO and pyroptosis-related proteins at different time points, including sham, day 3, day 7, day 14, and day 21 post-SNI. Tissue samples for WB analysis were collected at these respective time points. For all other experiments, including immunofluorescence, tissue samples were consistently collected on day 7 post-SNI. All mice were anesthetized and perfused via the left ventricle with 0.9% saline to remove blood, as confirmed by observing the liver turning pale.
For WB experiments: The spinal cord was exposed by removing the lamina, and the right side of the spinal cord was marked for orientation. The entire spinal cord was carefully extracted, with the lumbar enlargement identified at the T13-L1 vertebrae. The spinal cord was halved along the midline, discarding the right half. The left half was dissected on ice to isolate the dorsal horn, comprising approximately one-quarter of the spinal cord tissue, while discarding the ventral horn. Rapid handling was maintained throughout to preserve tissue integrity.
For IF experiments: Following perfusion with 0.9% saline, 20 ml of 4% paraformaldehyde was used for further fixation. After observing whole-body rigidity, the spinal column was extracted, and the tissue was post-fixed in 4% paraformaldehyde overnight. The spinal cord was processed similarly by removing the lamina, marking the right side for orientation, and isolating the lumbar enlargement. The tissue was halved along the midline, retaining the left dorsal horn while discarding the right half and ventral horn. The dorsal horn tissue was dehydrated in 30% sucrose until it sank, then embedded in OCT compound. Frozen sections were prepared using a cryostat for subsequent staining.
Intrathecal injection
Drugs were administered via intrathecal injection. Under anesthesia, a 0.25 mm insulin needle was carefully inserted into the L5-L6 intervertebral space. Proper placement was confirmed by observing a tail flick response. A solution containing the drug dissolved in 10 µl of saline was injected, and the needle was held in place for 15 s to prevent leakage.
Western blotting
Astrocytes and left spinal dorsal horn tissue were lysed using RIPA buffer (C500008; Sangon Biotech, Shanghai, China), and protein concentrations were assessed with a BCA assay (P0010, Beyotime Biotechnology). The samples were subjected to separation via 10% SDS-PAGE and subsequently transferred to PVDF membranes (IPVH00010, Millipore, USA). The membranes were blocked in 5% BSA in TBS-T for 2 h at room temperature and then incubated overnight at 4 °C with primary antibodies, including p-AMPK, AMPK, NLRP3, Cleaved Gasdermin D, pro-Caspase1, Caspase-1, PGC-1α, DRP1, MFN2, TFAM, and GAPDH (all at a dilution of 1:1000). After incubation, the membranes were scanned using a GS800 densitometer (Fusion FX6-XT, Wheelabrator, USA) and analyzed with Multi Gauge software (Fuji, Tokyo, Japan). Band intensities were quantified with FIJI software (NIH ImageJ), normalized to GAPDH, and presented as fold changes relative to the control samples.
Immunofluorescence staining
Tissue samples from the left dorsal horn of the spinal cord were fixed in 4% paraformaldehyde (PFA) overnight. After fixation, samples were dehydrated, embedded, and sectioned at 30 µm for later staining. The sections underwent PBS washing and were blocked for 1 h at room temperature with a solution containing 5% BSA and 1% Triton X-100. They were then incubated overnight at 4 °C with the following primary antibody combinations: TSPO (1:200) with NeuN (1:400), TSPO (1:200) with GFAP (1:400), TSPO (1:200) with Iba1 (1:400), or c-Fos (1:200) with NeuN (1:400). After an additional washing step, sections were exposed to secondary antibodies at 37 °C for 1 h, counterstained with DAPI, washed again, and observed using a Laser Confocal Electron Microscope (Nikon, Japan).
Isolation and culture of astrocytes
Spinal cord tissue was obtained from 24-h-old mice, minced, homogenized, and passed through a 70 µm filter before being transferred to culture flasks. On the third day, cultures were shaken at 200 rpm for 20 min, and the medium was subsequently refreshed. This process was repeated on the sixth day. Astrocyte purity was evaluated using GFAP staining. Cultures were maintained in DMEM supplemented with 10% FBS and 1% (v/v) penicillin/streptomycin at 37 °C in a humidified incubator with 5% CO₂. Cells at passages 6 to 8 were utilized for all experiments. To induce pyroptosis, astrocytes were primed with lipopolysaccharide (LPS, 100 ng/mL) for 24 h, followed by treatment with ATP (5 mM) for 30 min [39]. Ro 5–4864 (100 µg/mL) was added 2 h post-pyroptosis induction. For investigating the AMPK pathway, astrocytes were pretreated with 5 µg/mL Compound C 1 h prior to Ro 5–4864 administration [40, 41].
Mitochondrial assessment in astrocytes
The JC-1 fluorescent probe (Beyotime Biotechnology, China) was used to assess mitochondrial membrane potential (MMP) changes. Astrocytes were incubated with 1 µM JC-1 in culture medium for 30 min at 37 °C. After incubation, cells were washed to eliminate excess dye and then visualized with a laser-scanning confocal microscope (Nikon, Tokyo, Japan). The red and green fluorescence intensities were quantified using ImageJ software (version 4.1; Bethesda, MD, USA), with data presented as the red-to-green fluorescence ratio. To evaluate mitochondrial ROS levels, MitoSox Red dye (Invitrogen, M36008) was applied, while Mito Tracker Red (Invitrogen, M24426) was used to assess mitochondrial function. After treatments, astrocytes were incubated with MitoSox (20 µM) or Mito Tracker (50 µM) for 30 min at 37 °C. Cells were then washed three times with PBS, followed by Hoechst staining for nuclei at 37 °C. Images were acquired using a fluorescence microscope.
Transmission electron microscope
Spinal tissue samples were perfused and preserved in 2.5% glutaraldehyde. After fixation, the sections were exposed to 1% osmium tetroxide for 1 h, then washed and progressively dehydrated through an ascending series of alcohol concentrations. The specimens were embedded in Epon 812 epoxy resin, sectioned into ultrathin slices of 60 nm thickness, stained with uranyl acetate and lead citrate, and examined under a transmission electron microscope (JEOL Ltd., Tokyo, Japan).
Cell viability and LDH detection
Cell viability was evaluated using the Cell Counting Kit-8 (CCK-8, Epizyme, China) according to the manufacturer’s guidelines, while LDH release was quantified with the LDH Cytotoxicity Assay Kit (Beyotime) as instructed in the protocol, with absorbance recorded at 490 nm.
Enzyme-linked immunosorbent assay (ELISA)
Protein samples were extracted, and concentrations were determined as previously outlined. Levels of mature IL-1β and IL-18 in the spinal dorsal horn were measured using commercial ELISA kits (R&D Systems, Minneapolis, MN, USA) following the manufacturer’s instructions.
Oxidative stress and Reactive Oxygen Species (ROS) assessment in astrocytes
The DCFH-DA probe (S0035S, Beyotime Biotechnology) was used to quantify cytosolic reactive oxygen species (ROS) in astrocytes. Following specified treatments, cells were stained based on established protocols, and images were captured with a fluorescence microscope. Additionally, lysates from the spinal dorsal horn and astrocytes were analyzed for total superoxide dismutase (SOD) activity, glutathione (GSH) levels, and malondialdehyde (MDA) concentration. The following kits were utilized: Total Superoxide Dismutase Assay Kit (WST-8, Cat#S0101, Beyotime Biotechnology), Total Glutathione Assay Kit (Cat#S0052, Beyotime Biotechnology), and Lipid Peroxidation MDA Assay Kit (Cat#S0131, Beyotime Biotechnology).
Quantitative real-time PCR for mitochondrial DNA content
Mitochondrial DNA (mtDNA) copy number was measured through real-time quantitative RT-PCR using the mtDNA Copy Number Kit (Cat# MCN3, Detroit R&D). DNA was isolated from spinal dorsal horn tissues and astrocytes with the Qiagen DNeasy Blood and Tissue Kit. The total DNA concentration was determined using a NanoDrop 1000 (Thermo Fisher Scientific). Mitochondrial DNA levels were quantified by normalizing the mitochondrial gene (cytochrome b) against the nuclear gene (GAPDH). Quantitative real-time PCR was employed to assess mtDNA content in both tissue homogenates and cultured cells.
qRT-PCR array
Mouse mitochondrial gene expression profiles were assessed using the Mitochondria PCR Array, following the protocol provided by the manufacturer (Wcgene Biotech, Shanghai, China). Data analysis was conducted using R software (version 4.3.2).
Mendelian randomization and SMR analysis of TSPO and pain phenotypes
We extracted genome-wide association study (GWAS) data for the TSPO gene from the eQTLGen database (https://eqtlgen.org/), applying a significance threshold of P < 5e-8 to identify robust instrumental variables. Next, we retrieved GWAS data on multiple pain phenotypes from the IEU database (https://gwas.mrcieu.ac.uk/), specifically for back pain (ukb-b-9838), facial pain (ukb-b-17107), headache (ukb-b-12181), hip pain (ukb-b-7289), knee pain (ukb-b-16254), and neck or shoulder pain (ukb-b-18596). We used the TwoSampleMR R package to conduct Mendelian Randomization (MR) analysis, assessing the causal relationships between TSPO and the selected pain phenotypes [42]. To further validate the results, Summary-based Mendelian Randomization (SMR) analysis (https://yanglab.westlake.edu.cn/software/SMR/) was conducted [43]. For significant MR results, we performed Bayesian colocalization analysis to further eliminate false positive results.
Drug screening and molecular docking
We searched for TSPO-related compounds in three drug databases: DGIdb (https://www.dgidb.org/), DrugBank (https://go.drugbank.com/), and TTD (https://db.idrblab.net/ttd/), selecting compounds present in all three for molecular docking studies. Using PubChem database (http://pubchem.ncbi.nlm.nih.gov/), we retrieved and converted 2D ligand structures into 3D with ChemOffice 20.0, saving them in mol2 format. The TSPO protein sequence (Uniprotkb: P30536) was sourced from UniProt database (https://www.uniprot.org/), and a crystal model was built using AlphaFold (https://alphafold.ebi.ac.uk), saved in PDB format. Molecular Operating Environment (MOE) 2019 was used for energy minimization, target protein preprocessing, and active site identification. Docking simulations were conducted in MOE 2019 with 50 runs per experiment, with binding affinities assessed by docking scores and visualized through PyMOL 2.6.0 and Discovery Studio 2019.
Statistical analysis
Statistical analyses for bioinformatics were conducted using the R language (version 4.3.2). Data analysis and graph construction were carried out with GraphPad Prism 8.3.0. Results are presented as mean values ± standard error of the mean (SEM). Two-tailed Student’s t-tests were utilized for comparisons between two groups, while one-way or two-way ANOVA with Tukey’s multiple comparisons was employed for analyses involving multiple groups. Significance levels are indicated as follows: *p < 0.05, **p < 0.01, ***p < 0.001.
Results
Characterization of SNI-induced changes in the spinal dorsal horn
To characterize the changes of spinal dorsal horn after SNI, we conducted a differential gene expression analysis and identified 91 DEGs (Fig. 1A), Among them, 90 genes upregulated and 1 downregulated (p-value < 0.05, |log2(fold change)|> 1), with a volcano plot illustrating their distribution (Fig. 1B). The DEGs were enriched in inflammation and immune-related biological processes by GO analyses (Supplementary Fig. 1A). KEGG revealed an enrichment of Fc gamma R-mediated phagocytosis, NOD-like receptor signaling, chemokine signaling, and cytosolic DNA-sensing pathways (Fig. 1C). The NOD-like receptor (NLR) signaling pathway, particularly the NLRP3 inflammasome, was notably activated after SNI. GSEA results further supported these findings, highlighting a strong association with NLRP3 inflammasome, cytokine signaling, and pyroptosis in SNI (Fig. 1D-F). These results suggest that pyroptosis and mitochondrial dysfunction play critical roles in neuropathic pain process after SNI.
Characterization of SNI-induced changes in the spinal dorsal horn. A Heatmap showing differential gene expression in Sham and SNI. B Volcano plot showing the significantly upregulated and downregulated genes between SNI and Sham groups. C Pathways enriched by KEGG showed important biological pathways associated with differentially expressed genes. D-F GSEA results for enrichment of NLRP3 inflammasome pathway, Cytokine signaling in immune systems and Pyroptosis. G Module-trait relationship analysis by WGCNA showed correlations between gene modules and clinical traits
To identify SNI-related genes, we performed Weighted Gene Co-Expression Network Analysis (WGCNA). Sample clustering was well-defined (Supplementary Fig. 1B), and we selected a soft threshold of 7 basing on a scale-free topology model fit (R2 > 0.9) and high mean connectivity (Supplementary Fig. 1C, D). After merging highly correlated modules using a clustering height cut-off of 0.25, 24 gene modules were identified for further analysis (Supplementary Fig. 1E). Module-trait relationships showed that the yellow module exhibited a strong negative correlation with the control group (r = -0.98, p = 4e-08) and a positive correlation with SNI (r = 0.98, p = 4e-08) (Fig. 1G; Supplementary Fig. 1F). Furthermore, it was demonstrated that these gene networks were independent of each other (Supplementary Fig. 1G). GO and KEGG analyses of yellow module genes revealed significant enrichment in inflammatory pathways, consistent with differential expression patterns (Supplementary Fig. 1I, J). These findings further highlight the essential role of inflammation in the spinal dorsal horn in response to SNI.
TSPO was correlated to pyroptosis and inflammation in neuropathic pain
Previous studies have shown that pyroptosis and inflammasome activation are strongly linked to mitochondrial dysfunction [44]. Consistently, our GSEA analysis confirmed the existence of mitochondrial impairment after SNI (Fig. 2A-C). Moreover, we performed a Venn analysis of DEGs, SNI-related genes (yellow module), and mitochondrial genes (Fig. 2D), which identified TSPO and UCP2 as the critical genes involved in neuropathic pain (Supplementary Fig. 2A, B). Existing evidence indicates that TSPO plays a role in pain modulation; however, the underlying precise mechanisms remain to be elucidated.
TSPO was correlated to pyroptosis and inflammation in neuropathic pain. A-C GSEA results for enrichment of mitochondrial complex I assembly, enrichment of the electron transport chain and mitochondrial complex III assembly. D Venn diagram illustrating the intersection between SNI-related genes, mitochondrial genes, and differentially expressed genes (DEGs). E Schematic diagram of the experimental flow. F Von Frey results for the Sham and SNI groups at different time points (days 0, 3, 7, 14, and 21), n = 10 per group. G Western blot showing the expression levels of TSPO, NLRP3, Caspase-1, and GSDMD-N at various time points post-SNI. H–K Quantification of the relative expression of TSPO, NLRP3, Caspase-1, and GSDMD-N at various time points post-SNI, n = 10 per group. L, M ELISA results showing the levels of IL-1β and IL-18, n = 6 per group. Data are represented as mean ± SEM, ***p < 0.001
Therefore, we validated the bioinformatics findings through experimental approaches. Neuropathic pain was induced using the SNI model, and pain thresholds in mice were assessed on days 0 (baseline), 3, 7, 14, and 21 post-surgeries (Fig. 2E). To control potential confounding effects of the surgical procedure, a sham-operated and control group was included in the study (Supplementary Fig. 8). As expected, SNI mice showed increased sensitivity to mechanical stimuli compared to sham controls (Fig. 2F). Western blot analysis revealed that TSPO expression was elevated on day 7 post-SNI and maintained till day 21 or longer. At the same time, pyroptosis markers (NLRP3, GSDMD-N, Caspase-1) were also significantly elevated on day 7 and maintained high levels thereafter (Fig. 2G-K). This suggests that day 7 post-SNI is a critical time point for these processes. ELISA analysis further confirmed that inflammatory cytokines IL-1β and IL-18 were upregulated on day 7 (Fig. 2L, M). These results demonstrate a positive correlation between TSPO expression and pyroptosis.
Pharmacological increase but not decrease of TSPO alleviated neuropathic pain
To investigate the relationship between TSPO and neuropathic pain, we modulated TSPO expression pharmacologically (Fig. 3A). As TSPO was positively correlated with pyroptosis, we firstly intrathecally administered PK 11195, a TSPO inhibitor at 2 μg (in 10 μl saline) daily from the first day post-SNI [27]. Von Frey tests were conducted two hours later after each administration, and the results revealed that inhibition of TSPO amplified the hypersensitivity to mechanical stimuli in SNI mice (Fig. 3B). This result was also supported by immunofluorescence results, which showed a significant increase in the number of c-Fos + neurons after TSPO inhibition Fig. 3C; Supplementary Fig. 2C). PK 11195 administration resulted in elevated levels of inflammatory cytokines IL-1β and IL-18 (Fig. 3D, E), as well as increased pyroptosis-related proteins, including NLRP3, GSDMD-N, and Caspase-1 (Fig. 3F; Supplementary Fig. 2D-G). The above results indicate that the decreased expression of TSPO weakens its inhibitory effect on pyroptosis.
Pharmacological increase but not decrease of TSPO alleviated neuropathic pain. A Schematic diagram of the experimental flow. B Behavioral changes following TSPO inhibition by PK 11195 were assessed using von Frey testing at multiple time points (days 0, 3, 7, 14, and 21), n = 10 per group. C Representative immunofluorescence images showing c-Fos and NeuN co-staining after PK 11195 treatment, Scale bar = 50 μm. D, E ELISA results showing the levels of IL-1β and IL-18 after PK 11195 administration, n = 6 per group. F Western blot showing the expression levels of TSPO, NLRP3, Caspase-1 and GSDMD-N after PK 11195 treatment. G Behavioral tests of mice receiving different concentrations of Ro 5–4864, n = 6 per group. H Behavioral changes following TSPO activation by Ro 5–4864 were assessed using von Frey testing at multiple time points (days 0, 3, 7, 14, and 21), n = 10 per group. I Western blot showing the expression levels of TSPO, NLRP3, Caspase-1 and GSDMD-N after Ro 5–4864 treatment. J Representative immunofluorescence images showing c-Fos and NeuN co-staining after Ro 5–4864 treatment, Scale bar = 50 μm. K ELISA results showing the levels of the IL-1β and IL-18 after Ro 5–4864 treatment, n = 6 per group. Data are represented as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001
To further verify the role of TSPO, we upregulated its expression using Ro 5–4864. The results showed that Ro 5–4864 at 1 μg and 3 μg significantly reduced pain sensitivity, while 0.25 μg had no effect. Besides, no differences were detected between the 1 μg and 3 μg doses over the 21-day observation period (Fig. 3G). As a result, 1 μg was selected for further experiments. Von Frey tests show that Ro 5–4864 effectively alleviated mechanical allodynia (Fig. 3H). Administration of Ro 5–4864 resulted in the upregulation of TSPO (Fig. 3I; Supplementary Fig. 2H), which was shown to decrease the expression of pyroptosis-related proteins (NLRP3, Caspase-1, and GSDMD-N) by Western blot analysis (Fig. 3I; Supplementary Fig. 2I-K). Immunofluorescence staining indicated a reduction in c-Fos + neurons (Fig. 3J; Supplementary Fig. 2L), and ELISA results demonstrated lower levels of IL-1β and IL-18 (Fig. 3K, L). To exclude the potential interference of the drug itself on the results, we conducted a separate experiment to rule out its effect (Supplementary Fig. 7). Collectively, these findings suggest that upregulated TSPO alleviates neuropathic pain by modulating pyroptosis.
Genetic increase of TSPO in astrocytes alleviated neuropathic pain by reducing pyroptosis
To determine whether the function of TSPO was cell-type specific, immunofluorescence staining was performed on the spinal dorsal horn using anti-TSPO, NeuN, GFAP, and Iba1 antibodies. Interestingly, the results demonstrated that TSPO was predominantly co-expressed with GFAP + astrocytes, with minimal expression observed in neurons or microglia (Fig. 4A). Therefore, we constructed astrocyte-specific TSPO adeno-associated virus (AS-TSPO) to upregulate the expression of TSPO in astrocytes. AS-TSPO was injected into the left spinal dorsal horn in mice (Fig. 4B), and viral effectiveness was verified two weeks later by observing green fluorescence at the injection site and co-localization with astrocytes (Fig. 4C). Western blot confirmed the upregulation of TSPO by AS-TSPO (Fig. 4D; Supplementary Fig. 3B). After that, we investigated the role of TSPO overexpression in astrocytes during neuropathic pian by injection AS-TSPO two weeks before SNI. Von Frey tests indicated that AS-TSPO alleviated mechanical allodynia (Fig. 4E) and reduced the number of c-Fos + neurons (Fig. 4F; Supplementary Fig. 3C). Additionally, inflammatory cytokines, IL-1β and IL-18 (Fig. 4G, H), as well as pyroptosis-related proteins NLRP3, Caspase-1, and GSDMD-N were all suppressed (Fig. 4I; Supplementary Fig. 3D-F).
Genetic increase of TSPO in astrocytes alleviated neuropathic pain by reducing pyroptosis. A Representative immunofluorescence images showing TSPO/NeuN, TSPO/GFAP, and TSPO/Iba1 co-staining, Scale bar = 50 μm. B Schematic diagram of the experimental flow. C Representative immunofluorescence images showing AS-TSPO colocalized with GFAP + astrocytes, scale bar = 50 μm. D Western blot shows the expression levels of TSPO in AS-TSPO or AS-TSPO-Ctrl treated conditions. E Behavioral changes following TSPO activation by AS-TSPO were assessed using von Frey tests at multiple time points (days 0, 3, 7, 14, and 21), n = 10 per group. F Representative immunofluorescence images showing c-Fos and NeuN co-staining after AS-TSPO treatment, Scale bar = 50 μm. G, H ELISA results showing the levels of IL-1β and IL-18 after AS-TSPO treatment, n = 6 per group. I Western blot showing the expression levels of TSPO, NLRP3, Caspase-1 and GSDMD-N after AS-TSPO treatment. Data are represented as mean ± SEM, ***p < 0.001
Mitochondial dysfuction and AMPK-PGC-1α activation were observed after SNI
Since TSPO has been associated with pyroptosis in alleviating neuropathic pain, we next aimed to investigate the mechanisms by which TSPO regulates pyroptosis. Transcriptome analysis revealed that SNI induces mitochondrial dysfunction and activates oxidative stress (Figs. 2A-C and 5A). Mitochondrial integrity is crucial in regulating pyroptosis, with adenosine 5'-monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) identified as key regulatory molecules.
Mitochondial dysfuction and AMPK-PGC-1α activation were observed after SNI. A GSEA results for enrichment of oxidative stress response. B Schematic diagram of the experimental flow. C Volcano plot showing the significantly upregulated and downregulated genes between SNI and AS-TSPO groups, n = 3 per group. D PCR array heat map illustrating the relative mRNA levels of mitochondrial injury-related genes, including those associated with fusion and fission, between the SNI and AS-TSPO groups, n = 3 per group. E Western blot showing the expression levels of p-AMPK, AMPK, PGC-1α, MFN2 and TFAM in SNI and AS-TSPO groups. F–H Levels of antioxidant stress markers (GSH, SOD) and oxidative stress marker (MDA) in different groups, n = 6 per group. I Representative transmission electron microscopy images of mitochondria in the spinal dorsal horn of indicated groups, scale bar = 2 μm. Data are represented as mean ± SEM, ***p < 0.001
By conducting PCR array analysis (Fig. 5B), the overall expression and the up- and down- regulation distribution of mitochondria-related genes were displayed (Fig. 5C; Supplementary Fig. 3A). A heat map showed the expression levels of mitochondrial injury, fusion, fission, and pathway-related genes in the SNI and AS-TSPO groups on day 7 in the spinal dorsal horn (Fig. 5D). Specifically, compared to AS-TSPO, we found that SNI significantly upregulated the expression of mitochondrial damage-related genes (Bak1, Bid) and fission genes (Fis1, Drp1). In contrast, AS-TSPO treatment notably increased the expression of fusion genes (Mfn2, Mfn1, Opa1). This suggests that AS-TSPO treatment may induce a trend where fission gene expression decreases and fusion gene expression increases. Regarding the AMPK-PGC-1α pathway, AS-TSPO treatment significantly upregulated the expression of genes associated with this pathway.
At the protein level, AS-TSPO treatment upregulated AMPK-PGC-1α pathway proteins, MFN2, and TFAM, while decreased DRP1 expression (Fig. 5E; Supplementary Fig. 3G-K). It also reduced oxidative stress levels, indicated by lower MDA and increased antioxidant markers (GSH, SOD) (Fig. 5F-H), and improved mitochondrial health, as shown by elevated mtDNA copy numbers (Supplementary Fig. 3L). Through transmission electron microscopy results, we found that AS-TSPO alleviated mitochondrial damage caused by SNI (Fig. 5I). Overall, TSPO overexpression activated the AMPK-PGC-1α pathway, supporting mitochondrial biogenesis, maintaining the balance of fusion and fission, reducing oxidative stress levels, and sustaining mitochondrial function, collectively influencing pyroptosis and the release of inflammatory factors.
TSPO protected against mitochondrial dysfunction and pyroptosis by enhancing AMPK-PGC1α pathway
To make sure that TSPO regulates mitochondrial function and mitigate pyroptosis and inflammation via the AMPK-PGC-1α pathway, the AMPK inhibitor Compound C was used to block this pathway. Compound C 6 μg/day in 10 μl saline was intrathecal administrated from the first day after SNI [45] (Fig. 6A), and the results showed that Compound C effectively inhibited AMPK-PGC-1α pathway, reducing the expression of mitochondrial biogenesis protein TFAM and fusion protein MFN2, while increasing mitochondrial fission protein DRP1 Fig. 6B; Supplementary Fig. 4A-F). These changes led to elevated MDA levels, reduced antioxidant factors (GSH and SOD), (Supplementary Fig. 4G-I) and mtDNA copy number (Supplementary Fig. 4 J), thereby exacerbating mitochondrial damage (Fig. 6C). Inhibition of the AMPK-PGC-1α pathway also elevated pyroptosis-related inflammatory cytokines IL-1β and IL-18 (Fig. 6D, E), as well as pyroptosis proteins NLRP3, Caspase-1, and GSDMD-N (Fig. 6F; Supplementary Fig. 4 K-M). These changes ultimately led to behavioral alterations, as the c-Fos + neurons (Fig. 6G; Supplementary Fig. 4N) and the analgesic effect of AS-TSPO was significantly diminished by Compound C (Fig. 6H). These findings indicate that TSPO regulates mitochondrial fusion and fission, influencing cellular pyroptosis and alleviating neuropathic pain via the AMPK-PGC-1α pathway.
TSPO protected against mitochondrial dysfunction and pyroptosis by enhancing AMPK-PGC1α pathway. A Schematic diagram of the experimental flow. B Western blot showing the expression levels of TSPO, p-AMPK, AMPK, PGC-1α, MFN2 and TFAM after inhibition of AMPK. C Representative transmission electron microscopy images of mitochondria after inhibition of AMPK, scale bar = 2 μm. D, E ELISA results showing the levels of the IL-1β and the IL- after inhibition of AMPK, n = 6 per group. F Western blot showing the expression levels of NLRP3, Caspase-1 and GSDMD-N after inhibition of AMPK. G Representative immunofluorescence images showing c-Fos and NeuN co-staining after inhibition of AMPK. H Behavioral testing of von Frey after inhibition of AMPK at different time points (days 0, 3, 7, 14, and 21), n = 10 per group. Data are represented as mean ± SEM, ***p < 0.001
Mitochondrial dysfunction was observed during pyroptosis induction in primary astrocytes
To validate the astrocyte-specific role of TSPO, primary astrocytes were isolated (Fig. 7A). Astrocytes were treated with LPS (100 ng/mL) for 24 h, followed by a 30-min pulse of 5 mM ATP to induce pyroptosis [39]. The pyroptosis model was validated by elevated IL-1β and IL-18 levels in the supernatant (Fig. 7B, C), an increase in PI-stained pyroptotic astrocytes (Fig. 7D; Supplementary Fig. 5A), and higher LDH levels, indicating membrane rupture and enhanced permeability (Supplementary Fig. 5B). TSPO and pyroptosis-related proteins (NLRP3, Caspase-1 and GSDMD-N) were notably upregulated, consistent with findings from animal models (Fig. 7E; Supplementary Fig. 5C-F). From the perspective of mitochondria, after pyroptosis occurs in astrocytes, there is a significant increase in ROS both within the astrocytes and in the mitochondria (Fig. 7F, G; Supplementary Fig. 5G, H). This is accompanied by a decreased membrane potential (Fig. 7H; Supplementary Fig. 5I), reduced antioxidant markers (GSH, SOD), elevated MDA levels (Supplementary Fig. 5 J-L), and suppressed mtDNA levels (Supplementary Fig. 5 M). Additionally, observable mitochondrial network fragmentation occurs (Fig. 7I, J).
Mitochondrial dysfunction was observed during pyroptosis induction in primary astrocytes. A Representative images of GFAP immunofluorescence staining to identify astrocytes, scale bar = 50 μm. B, C ELISA showing the levels of IL-1β and IL-18 in different groups, n = 6 per group. D Representative images of PI staining demonstrate the number of pyroptotic cells in the different groups, scale bar = 50 μm. E Western blot showing the expression levels of p-AMPK, PGC1, MFN2, and TFAM after ATP + LPS treatment. F Representative images of ROS staining indicate the level of ROS produced by astrocytes after ATP + LPS treatment, scale bar = 50 μm. G Representative images of MitoSOX staining show the level of mitochondrial ROS levels after ATP + LPS treatment, scale bar = 50 μm. H Representative images of JC-1 staining show a shift from aggregates (red) to monomers (green), indicating mitochondrial depolarization, scale bar = 50 μm. I Representative images of Mito-Tracker staining show mitochondrial morphology, scale bar = 20 μm. J Quantification of mitochondrial aspect ratio, n = 6 per group. K Western blot showing the expression levels of TSPO, NLRP3, Caspase-1, and GSDMD-N after ATP + LPS treatment. Data are represented as mean ± SEM, ***p < 0.001
Meanwhile, TSPO was also significantly increased in the pyroptosis group, consistent with our animal experimental results. In the pyroptosis model, there was an imbalance in oxidative stress, with elevated reactive oxygen species (ROS) and mitochondrial damage, which aligns with the findings from the animal experiments. Additionally, we observed a significant increase in proteins related to mitochondrial biogenesis (TFAM, mitochondrial copy number), fusion (MFN2), and fission (DRP1) compared to the normal group. Based on previous experimental results, we speculate that this change may be associated with insufficient upregulation of AMPK-PGC-1α in the pyroptosis model (Fig. 7K; Supplementary Fig. 5N-R).
TSPO-driven modulation of mitochondrial function and pyroptosis in astrocytes via the AMPK-PGC-1α pathway
Next, we investigated the effects of TSPO and the AMPK-PGC-1α pathway on astrocytes. Pyroptotic astrocytes were treated with Ro 5–4864 at various concentrations (0, 10, 50, 100, and 300 nM) for 2 h. CCK-8 assay showed that 100 nM Ro 5–4864 had the most pronounced effect (Supplementary Fig. 6A), which was selected as the standard condition for subsequent treatments. Supplement of Ro 5–4864 significantly reduced the proliferation of pyroptotic astrocytes, as shown by PI staining, and decreased the levels of IL-1β, IL-18, and LDH in the supernatant (Fig. 8A, B; Supplementary Fig. 6B, C). TSPO was elevated with the activation of the AMPK-PGC-1α pathway, which significantly increased the expression of the mitochondrial fusion protein MFN2 and the biogenesis marker TFAM, while attenuating the increase in the fission protein DRP1 (Fig. 8C; Supplementary Fig. 6D-I).
TSPO-driven modulation of mitochondrial function and pyroptosis in astrocytes via the AMPK-PGC-1α pathway. A Representative images of PI staining showed the number of pyroptosis cells in different groups, scale bar = 50 μm. B ELISA showing the levels of IL-1β and IL-18 in different groups, n = 6 per group. C Western blot showing the expression levels of TSPO, p-AMPK, PGC1, MFN2, and TFAM in different groups. D Representative images of ROS staining indicates the level of ROS produced by cells in different groups, scale bar = 50 μm. E Representative images of MitoSOX staining shows the level of mitochondrial ROS levels in different groups, scale bar = 50 μm. F Representative images of JC-1 staining shows a shift from aggregates (red) to monomers (green), indicating mitochondrial depolarization, scale bar = 50 μm. G Quantitative analysis of the relative intensity of JC-1 in different groups, n = 6 per group. H Quantification of mitochondrial aspect ratio, n = 6 per group. I Representative images of Mito-Tracker staining show mitochondrial morphology, scale bar = 20 μm. J Representative images of TSPO and Mito-tracker co-staining, scale bar = 20 μm. K Western blot showing the expression levels of NLRP3, Caspase-1, and GSDMD-N in different groups. Data are represented as mean ± SEM, **p < 0.01, ***p < 0.001
Ro 5–4864 improved cellular antioxidant capacity, evidenced by increased GSH and SOD levels, reduced MDA, and restored mtDNA levels (Supplementary Fig. 6 J-M). It also lowered intracellular and mitochondrial ROS (Fig. 8D, E; Supplementary Fig. 6N, O), restored mitochondrial membrane potential (Fig. 8F, G), and reduced mitochondrial network disruption and fragmentation, as confirmed by TSPO and Mito Tracker co-staining (Fig. 8H-J). Pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD-N) were ultimately downregulated (Fig. 8K; Supplementary Fig. 6P-R). However, treatment with the AMPK inhibitor Compound C significantly reduced AMPK and PGC-1α expression, disrupted mitochondrial fusion-fission balance, and impaired mitochondrial function, thereby abolishing the protective effect of TSPO on pyroptosis-related proteins. These findings confirm that TSPO enhances mitochondrial function and regulates pyroptosis in astrocytes through the AMPK-PGC-1α pathway.
Association of TSPO gene with multiple pain phenotypes and validation of potential targeted drugs
To gain the value of this study in clinic, we conducted Mendelian randomization (MR) analysis using GWAS data to explore the association between the TSPO gene and various types of pain, including headache, knee pain, neck and shoulder pain, and back pain (Fig. 9A). The MR analysis indicated that TSPO was associated with neck, shoulder, and back pain. Unexpectedly, we found that the direction of the β-value was consistent and negatively correlated across all pain phenotypes (Fig. 9B), which suggested a negative correlation between TSPO and pain. Increased expression of TSPO was linked to pain relief, further supporting the effectiveness of our strategy to upregulate TSPO for pain alleviation, as demonstrated from a human whole-genome perspective.
Association of TSPO gene with multiple pain phenotypes and validation of potential targeted drugs. A Schematic diagram showing the localization of TSPO and its association with different pain types, including neck and shoulder pain, back pain, knee pain, and foot pain. B MR results depicting the association of TSPO with various pain phenotypes. β-values and 95% confidence intervals (CI) for each phenotype are shown, with significant associations highlighted in red. C Manhattan plot from the SMR analysis of TSPO expression with neck and shoulder pain, showing the association signals across the genome. D Venn diagram showing the intersection of TSPO-targeted drugs from three different drug databases. Chlormezanone and Zopiclone were identified as common drugs across all databases. E, F Molecular docking of Chlormezanone and Zopiclone with TSPO, showing their respective binding energies. G Behavioral changes at different concentrations of Zopiclone were assessed using the von Frey test at multiple time points (days 0, 3, 7, 14, and 21), n = 10 per group. Data are represented as mean ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001
Next, we further confirmed this causal relationship by performing SMR analysis on TSPO, focusing on various types of pain (Supplementary Fig. 8). Since both MR and SMR tests showed positive results for neck or shoulder pain, we performed Bayesian colocalization analysis to exclude potential pleiotropic effects. The results, by integrating GWAS and eQTL data, revealed that TSPO expression is a causal factor for the neck or shoulder pain phenotype across all analyses (Fig. 9C and Table 1)
To uncover clinical drugs associated with TSPO and assess their potential in pain treatment, we performed a comprehensive search in three major drug target databases to identify TSPO-targeting compounds. This search highlighted two drugs—chlormezanone and zopiclone—that were consistently present across all three databases (Fig. 9D). Subsequent molecular docking studies confirmed that both compounds bind within TSPO with binding affinities of -5.6267 kcal/mol for chlormezanone and -6.9176 kcal/mol for zopiclone, signifying favorable binding activity (Fig. 9E, F). The superior binding affinity of zopiclone warranted its selection for further experimental investigation. We evaluated the therapeutic potential of zopiclone through intrathecal administration in SNI model. Initial testing showed that a dose of 0.25 µg was ineffective for pain relief, whereas a 5 µg dosage induced side effects, including drowsiness, complicating behavioral assessments. After dose optimization, a 2 µg dose was identified as the most effective dosage, achieving approximately 50% pain relief by day 21, though with limited efficacy during the early treatment phase (Fig. 9G).
Discussions
This study elucidates the critical roles of mitochondrial dysfunction, pyroptosis, and inflammation in neuropathic pain following SNI in spinal dorsal horn. Our findings highlight TSPO, a mitochondrial gene, as a key regulator of pyroptosis. Upregulation of TSPO reduces the expression of pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD-N) and decreases the release of inflammatory factors (IL-1β, IL-18). Mechanistic studies demonstrate that TSPO regulates mitochondrial biogenesis and the dynamic balance of fusion and fission through the AMPK-PGC1α pathway, enhancing antioxidant capacity, reducing both cellular and mitochondrial ROS levels, and maintaining mitochondrial homeostasis. To further enhance the clinical relevance of this research, we utilized human whole-genome sequencing data to validate TSPO upregulation as a therapeutic strategy for pain relief, identifying clinically relevant drugs targeting TSPO and providing new avenues for repurposing existing medications.
Pyroptosis is a programmed cell death mechanism reliant on inflammasomes, characterized by cell membrane rupture and the release of pro-inflammatory cytokines such as IL-1β and IL-18, which are significant in inflammatory diseases [46]. In neuropathic pain, inflammation plays a key role in inducing hypersensitivity and persistent pain. Numerous studies have shown that inflammatory factors, including TNF-α, IL-6, and IL-1β, are closely related to the onset and maintenance of pain [47,48,49,50,51]. These cytokines enhance pain signal transmission by activating nociceptive neurons or altering synaptic plasticity in the central nervous system. However, despite extensive research on the connection between inflammation and pain, the role of pyroptosis in pain has not been sufficiently explored. Only a few studies have preliminarily indicated that pyroptosis may exacerbate pain by activating inflammatory pathways in the nervous system [52]. Our study highlights the pivotal role of pyroptosis in the induction of pain. We observed elevated expression of pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD-N) and inflammatory factors (IL-1β, IL-18) in the spinal dorsal horn following SNI, which correlates with increased pain sensitivity. In contrast, the upregulation of TSPO mitigates pain behaviors in mice, as evidenced by reduced levels of pyroptosis-related proteins and inflammatory factors.
Research indicates that mitochondrial dysfunction is a primary trigger of pyroptosis. In lumbar disc degeneration, oxidative stress in nucleus pulposus cells results in mitochondrial damage, which causes the release of mitochondrial DNA into the cytoplasm. This event subsequently activates the cGAS-STING-NLRP3 axis, leading to the induction of pyroptosis. Therefore, maintaining mitochondrial homeostasis is crucial for regulating pyroptosis [53]. Mitochondrial biogenesis, fusion, and fission are essential for this homeostasis [54,55,56]. Although literature confirms increased expression of mitochondrial fission proteins in the spinal dorsal horn after SNI [57], focusing solely on mitochondrial fission without considering the overall dynamic balance (biogenesis, fusion, and fission) is insufficient to address fundamental issues. External stimuli, such as spinal nerve injury (SNI), cause mitochondrial damage that triggers mitochondrial fission, while mitochondrial biogenesis and fusion respond to maintain mitochondrial health. However, these processes are critical and should not be overlooked, as they are essential for preserving mitochondrial function. In our study, the results indicate an excessive increase in the mitochondrial fission protein DRP1, coupled with an insufficient elevation of proteins related to mitochondrial biogenesis (TFAM) and fusion (MFN2) following SNI. This imbalance contributed to heightened oxidative stress, a decline in mitochondrial membrane potential, elevated reactive oxygen species (ROS) levels, and exacerbated mitochondrial damage.
These changes are significantly regulated by the AMPK-PGC-1α pathway. As a cellular energy sensor, AMPK responds to metabolic and oxidative stress by activating PGC-1α, which upregulates the expression of mitochondrial-related genes, promoting mitochondrial DNA replication, transcription, and biogenesis, thereby improving mitochondrial function under stress conditions [58,59,60]. However, in the SNI group, although AMPK and PGC-1α were upregulated, they were insufficient to restore the balance of mitochondrial fission and fusion. Specifically, the high expression of mitochondrial fission proteins persisted, while the expression of fusion and biogenesis proteins remained inadequately elevated, indicating insufficient activation of the AMPK-PGC-1α pathway following injury. When we upregulated TSPO, we observed a reduction in the expression of mitochondrial fission proteins (DRP1) and an increase in the expression of mitochondrial biogenesis (TFAM) and fusion proteins (MFN2). This restoration of the balance between mitochondrial fusion and fission helped normalize mitochondrial function, reduce oxidative stress, lower ROS production, restore mitochondrial membrane potential, and ultimately improve overall mitochondrial health. These changes resulted in reduced pyroptosis and significantly alleviated pain behaviors in mice.
The downstream regulation of mitochondrial dynamics by the AMPK-PGC-1α signaling pathway is an important and intriguing area of focus. Studies have shown that the AMPK-PGC-1α axis regulates the astrocytic glutathione system to enhance mitochondrial function, thereby protecting it from oxidative and metabolic damage [61]. AMPK also modulates mitochondrial dynamics by activating autophagic flux in neurodegenerative diseases [62]. Furthermore, research suggests that the AMPK-PGC-1α-SIRT3 axis plays a critical role in regulating mitochondrial homeostasis and redox balance [63]. Currently, research primarily highlights the involvement of the glutathione system, autophagic flux, and SIRT3 in mitochondrial dynamics regulation. The regulation of mitochondria by the AMPK-PGC-1α axis remains underexplored and presents an exciting scientific question worthy of further investigation.
Additionally, through human Mendelian randomization analysis, we confirmed the significant association between TSPO expression and pain phenotypes, highlighting its clinical potential in treating neuropathic pain. Using drug databases, we identified Chlormezanone and Zopiclone as promising compounds for TSPO regulation. Chlormezanone, a central muscle relaxant effective for anxiety and muscle tension, is often combined with other analgesics to enhance pain relief in neuropathic conditions, suggesting a potential synergistic effect [64,65,66].
Zopiclone, a commonly used hypnotic for the treatment of insomnia, belongs to the class of non-benzodiazepine sedative-hypnotics [67]. Studies have demonstrated that zopiclone exerts its sedative effects primarily by binding to GABA-A receptors and potentially through interactions with TSPO, modulating cellular responses to enhance its sedative and sleep-promoting effects, thereby improving sleep structure and quality [68, 69]. Clinically, zopiclone is widely used to treat various types of insomnia, including difficulty in falling asleep, maintaining sleep, and early awakening. It is particularly suitable for adults, elderly patients, and individuals with hepatic impairment, offering fewer side effects compared to traditional benzodiazepines. Clinical studies have also revealed that zopiclone can provide sustained relief from chronic neuropathic pain and improve sleep quality in affected patients [70], with enhanced sleep quality potentially contributing to reduced pain perception [71]. Furthermore, evidence suggests that zopiclone, when used in combination with other analgesics, can enhance analgesic efficacy, supporting its role in multimodal pain management strategies [71, 72].
Our molecular docking analysis demonstrated that zopiclone exhibits superior binding energy compared to chlormezanone, corroborating its analgesic effects observed in mice. Behavioral experiments revealed that high doses of zopiclone induce sedation, impacting behavioral assessments, while lower doses showed minimal effects. An intrathecal dose of 2 µg was identified as the most effective, achieving approximately 50% pain relief on postoperative day 21 in the spared nerve injury (SNI) model. However, its limited efficacy during the early treatment phase may be attributed to rapid metabolism, with low doses transiently affecting TSPO expression. Prolonged TSPO expression, necessitating a cumulative effect, appears critical for sustained analgesia. While zopiclone represents a potential TSPO-targeted therapeutic for pain management, its development pathway remains challenging. Our findings provide new insights into its application for neuropathic pain and lay a foundation for future clinical trials and TSPO-focused therapies, although significant efforts are still required for clinical translation.
Despite the significant progress achieved in this study, several limitations remain that need to be addressed in future research. First, our results primarily focus on demonstrating how TSPO regulates mitochondrial function through the AMPK-PGC-1 pathway, influencing pyroptosis and contributing to neuropathic pain. However, multiple factors drive pyroptosis, and our study only investigates the mitochondrial aspect. For instance, Levo-tetrahydropalmatine has been reported to alleviate neuropathic pain by regulating pyroptosis through the Clec7a-MAPK/NF-κB pathway [9]. Additionally, autophagy regulation has been shown to reduce NLRP3 inflammasome-dependent pyroptosis, thereby mitigating neuropathic pain [6, 8]. Other factors, such as potassium efflux, calcium signaling, and lysosomal leakage, have also been shown to influence the regulation of pyroptosis by mitochondria [73]. This limitation underscores the need for future studies to explore alternative pathways and mechanisms contributing to pyroptosis in neuropathic pain. Second, although we identified potential TSPO-targeted drugs through database screening and molecular docking, along with preliminary behavioral assessments, further in-depth research is needed to comprehensively evaluate their efficacy and safety. Future studies should include detailed analyses of drug metabolism, structural assessments, and rigorous preclinical testing to establish their therapeutic potential. Additionally, the potential side effects of these drugs must be investigated to facilitate the translation of these findings into clinical applications. Conducting more extensive research into the clinical utility of TSPO-targeted therapies for neuropathic pain is critical for advancing this field. Addressing these limitations in future studies will contribute to a deeper understanding of pyroptosis mechanisms and enhance therapeutic strategies for neuropathic pain.
Data availability
No datasets were generated or analysed during the current study.
References
Costigan M, Scholz J, Woolf CJ (2009) Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci 32:1–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.neuro.051508.135531
Cohen SP, Mao J (2014) Neuropathic pain: mechanisms and their clinical implications. BMJ 348:f7656–f7656. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/bmj.f7656
Li L, Ru Q, Lu Y et al (2023) Tiam1 coordinates synaptic structural and functional plasticity underpinning the pathophysiology of neuropathic pain. Neuron 111:2038–2050.e6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuron.2023.04.010
Ji R-R, Nackley A, Huh Y et al (2018) Neuroinflammation and Central Sensitization in Chronic and Widespread Pain. Anesthesiology 129:343–366. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/ALN.0000000000002130
Woolf CJ, Salter MW (2000) Neuronal plasticity: increasing the gain in pain. Science 288:1765–1768. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.288.5472.1765
Hua T, Yang M, Song H et al (2022) Huc-MSCs-derived exosomes attenuate inflammatory pain by regulating microglia pyroptosis and autophagy via the miR-146a-5p/TRAF6 axis. J Nanobiotechnol 20:324. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12951-022-01522-6
Gao X, Gao L-F, Zhang Z-Y et al (2024) miR-99b-3p/Mmp13 axis regulates NLRP3 inflammasome-dependent microglial pyroptosis and alleviates neuropathic pain via the promotion of autophagy. Int Immunopharmacol 126:111331. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2023.111331
Wu D, Zhang Y, Zhao C et al (2022) Disruption of C/EBPβ-Clec7a axis exacerbates neuroinflammatory injury via NLRP3 inflammasome-mediated pyroptosis in experimental neuropathic pain. J Transl Med 20:583. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-022-03779-9
Wu D, Wang P, Zhao C et al (2023) Levo-tetrahydropalmatine ameliorates neuropathic pain by inhibiting the activation of the Clec7a-MAPK/NF-κB-NLRP3 inflammasome axis. Phytomedicine 121:155075. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.phymed.2023.155075
Su L, Zhang J, Gomez H et al (2022) Mitochondria ROS and mitophagy in acute kidney injury. Autophagy 19:401–414. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/15548627.2022.2084862
Lacapere J-J, Duma L, Finet S et al (2020) Insight into the Structural Features of TSPO: Implications for Drug Development. Trends Pharmacol Sci 41:110–122. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tips.2019.11.005
Gehlert DR, Yamamura HI, Wamsley JK (1985) Autoradiographic localization of “peripheral-type” benzodiazepine binding sites in the rat brain, heart and kidney. N-s Arch Pharmacol 328:454–460. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/BF00692915
Nutma E, Ceyzériat K, Amor S et al (2021) Cellular sources of TSPO expression in healthy and diseased brain. Eur J Nucl Med Mol Imaging 49:146–163. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00259-020-05166-2
Saraste M, Matilainen M, Vuorimaa A et al (2023) Association of serum neurofilament light with microglial activation in multiple sclerosis. J Neurol Neurosurg Psychiatry 94:698–706. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jnnp-2023-331051
De Picker LJ, Morrens M, Branchi I et al (2023) TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun 113:415–431. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbi.2023.07.023
Kraguljac NV, McDonald WM, Widge AS et al (2021) Neuroimaging Biomarkers in Schizophrenia. Am J Psychiatry 178:509–521. https://doiorg.publicaciones.saludcastillayleon.es/10.1176/appi.ajp.2020.20030340
Gallus M, Roll W, Dik A, et al (2023) Translational imaging of TSPO reveals pronounced innate inflammation in human and murine CD8 T cell-mediated limbic encephalitis. Sci Adv 9:eabq7595. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abq7595
Tang Q, Cheng Z, Liu S et al (2024) FGF1ΔHBS ameliorates retinal inflammation via suppressing TSPO signal in a type 2 diabetes mouse model. Biochem Pharmacol 221:116039. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2024.116039
Bonsack F, Alleyne CH, Sukumari-Ramesh S (2016) Augmented expression of TSPO after intracerebral hemorrhage: a role in inflammation? J Neuroinflamm 13:151. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-016-0619-2
Li X, Chen X, Yang F-Y et al (2024) Effect of mitochondrial translocator protein TSPO on LPS-induced cardiac dysfunction. J Adv Res S2090–1232(24):00437–00445. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jare.2024.10.004
Jin G-L, Liu H-P, Huang Y-X et al (2022) Koumine regulates macrophage M1/M2 polarization via TSPO, alleviating sepsis-associated liver injury in mice. Phytomedicine 107:154484. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.phymed.2022.154484
Zhang D, Man D, Lu J et al (2023) Mitochondrial TSPO Promotes Hepatocellular Carcinoma Progression through Ferroptosis Inhibition and Immune Evasion. Adv Sci 10:e2206669. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/advs.202206669
Betlazar C, Middleton RJ, Banati R, Liu G-J (2020) The Translocator Protein (TSPO) in Mitochondrial Bioenergetics and Immune Processes. Cells 9:512. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells9020512
Menevse AN, Ammer L-M, Vollmann-Zwerenz A et al (2023) TSPO acts as an immune resistance gene involved in the T cell mediated immune control of glioblastoma. Acta Neuropathol Commun 11:75. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40478-023-01550-9
Fu Y, Wang D, Wang H et al (2020) TSPO deficiency induces mitochondrial dysfunction, leading to hypoxia, angiogenesis, and a growth-promoting metabolic shift toward glycolysis in glioblastoma. Neuro Oncol 22:240–252. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/neuonc/noz183
Wei X-H, Wei X, Chen F-Y et al (2013) The upregulation of translocator protein (18 kDa) promotes recovery from neuropathic pain in rats. J Neurosci 33:1540–1551. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.0324-12.2013
Liu X, Liu H, Xu S et al (2016) Spinal translocator protein alleviates chronic neuropathic pain behavior and modulates spinal astrocyte-neuronal function in rats with L5 spinal nerve ligation model. Pain 157:103–116. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/j.pain.0000000000000339
Costigan M, Moss A, Latremoliere A et al (2009) T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci 29:14415–14422. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.4569-09.2009
Ritchie ME, Phipson B, Wu D et al (2015) limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res 43:e47–e47. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nar/gkv007
Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS: J Integr Biol 2012;16:284–287. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/omi.2011.0118
Liberzon A, Subramanian A, Pinchback R et al (2011) Molecular signatures database (MSigDB) 3.0. Method Biochem Anal 27:1739–1740. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btr260
Langfelder P, Horvath S (2008) WGCNA: an R package for weighted correlation network analysis. BMC Bioinf 9:559. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2105-9-559
Bowling AC, DeLorenzo RJ (1982) Micromolar affinity benzodiazepine receptors: identification and characterization in central nervous system. Science 216:1247–1250. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.6281893
Fan Z, Brooks DJ, Okello A, Edison P (2017) An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain 140:aww349. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/brain/aww349
Zhou G, Myers R, Li Y et al (2001) Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108:1167–1174. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI13505
Russak EM, Bednarczyk EM (2018) Impact of Deuterium Substitution on the Pharmacokinetics of Pharmaceuticals. Ann Pharmacother 53:211–216. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/1060028018797110
Decosterd I, Woolf CJ (2000) Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 87:149–158. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0304-3959(00)00276-1
Kohro Y, Sakaguchi E, Tashima R et al (2015) A new minimally-invasive method for microinjection into the mouse spinal dorsal horn. Sci Rep 5:14306. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep14306
Li F, Jiang S-Y, Tian T et al (2022) Kir6.1/K-ATP channel in astrocytes is an essential negative modulator of astrocytic pyroptosis in mouse model of depression. Theranostics 12:6611–6625. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.77455
Zhang Y, Miao J-M (2018) Ginkgolide K promotes astrocyte proliferation and migration after oxygen-glucose deprivation via inducing protective autophagy through the AMPK/mTOR/ULK1 signaling pathway. Eur J Pharmacol 832:96–103. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2018.05.029
Cheng J, Zhou Y, Qiao H et al (2023) Curcumin protects from LPS-induced activation of astrocytes via AMPK pathway. NeuroReport 34:748–758. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/WNR.0000000000001950
Hemani G, Zheng J, Elsworth B et al (2018) The MR-Base platform supports systematic causal inference across the human phenome. Elife 7:e34408. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.34408
Zhu Z, Zheng Z, Zhang F et al (2018) Causal associations between risk factors and common diseases inferred from GWAS summary data. Nat Commun 9:224. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-017-02317-2
Xian H, Watari K, Sanchez-Lopez E et al (2022) Oxidized DNA fragments exit mitochondria via mPTP- and VDAC-dependent channels to activate NLRP3 inflammasome and interferon signaling. Immunity 55:1370–1385.e8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.immuni.2022.06.007
Gabriel KA, Streicher JM (2023) HSP90 inhibition in the mouse spinal cord enhances opioid signaling by suppressing an AMPK-mediated negative feedback loop. Sci Signaling 16:eade2438. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/scisignal.ade2438
Yu P, Zhang X, Liu N et al (2021) Pyroptosis: mechanisms and diseases. Signal Transduction Targeted Ther 6:128. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41392-021-00507-5
Sommer C, Leinders M, Üçeyler N (2017) Inflammation in the pathophysiology of neuropathic pain. Pain 159:595–602. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/j.pain.0000000000001122
Mitsui K, Hishiyama S, Jain A et al (2023) Role of macrophage autophagy in postoperative pain and inflammation in mice. J Neuroinflamm 20:102. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-023-02795-w
Petrikonis K, Bernatoniene J, Kopustinskiene DM et al (2024) The Antinociceptive Role of Nrf2 in Neuropathic Pain: From Mechanisms to Clinical Perspectives. Pharmaceutics 16:1068. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/pharmaceutics16081068
Fitzcharles M-A, Cohen SP, Clauw DJ et al (2021) Nociplastic pain: towards an understanding of prevalent pain conditions. Lancet 397:2098–2110. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0140-6736(21)00392-5
Mai C-L, Tan Z, Xu Y-N et al (2021) CXCL12-mediated monocyte transmigration into brain perivascular space leads to neuroinflammation and memory deficit in neuropathic pain. Theranostics 11:1059–1078. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.44364
Hao W, Feng C (2023) Research progress on pyroptosis and its effect on the central nervous system. Neurobiol Dis 188:106333. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2023.106333
Zhang W, Li G, Luo R et al (2022) Cytosolic escape of mitochondrial DNA triggers cGAS-STING-NLRP3 axis-dependent nucleus pulposus cell pyroptosis. Exp Mol Med 54:129–142. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s12276-022-00729-9
Johnson J, Mercado-Ayon E, Mercado-Ayon Y et al (2021) Mitochondrial dysfunction in the development and progression of neurodegenerative diseases. Arch Biochem Biophys 702:108698. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.abb.2020.108698
Meyer JN, Leuthner TC, Luz AL (2017) Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology 391:42–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tox.2017.07.019
Wu L, Fan Z, Gu L et al (2023) QiShenYiQi dripping pill alleviates myocardial ischemia-induced ferroptosis via improving mitochondrial dynamical homeostasis and biogenesis. J Ethnopharmacol 308:116282. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jep.2023.116282
Zhang K-L, Li S-J, Pu X-Y et al (2022) Targeted up-regulation of Drp1 in dorsal horn attenuates neuropathic pain hypersensitivity by increasing mitochondrial fission. Redox Biol 49:102216. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2021.102216
Ozaki Y, Ohashi K, Otaka N et al (2023) Myonectin protects against skeletal muscle dysfunction in male mice through activation of AMPK/PGC1α pathway. Nat Commun 14:4675. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-40435-2
Fan H, Ding R, Liu W et al (2021) Heat shock protein 22 modulates NRF1/TFAM-dependent mitochondrial biogenesis and DRP1-sparked mitochondrial apoptosis through AMPK-PGC1α signaling pathway to alleviate the early brain injury of subarachnoid hemorrhage in rats. Redox Biol 40:101856. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2021.101856
Herzig S, Shaw RJ (2017) AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol 19:121–135. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm.2017.95
Guo X, Jiang Q, Tuccitto A et al (2018) The AMPK-PGC-1α signaling axis regulates the astrocyte glutathione system to protect against oxidative and metabolic injury. Neurobiol Dis 113:59–69. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2018.02.004
Shah SZA, Zhao D, Hussain T, Yang L (2017) Role of the AMPK pathway in promoting autophagic flux via modulating mitochondrial dynamics in neurodegenerative diseases: Insight into prion diseases. Ageing Res Rev 40:51–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.arr.2017.09.004
Peerapanyasut W, Kobroob A, Palee S et al (2019) N-Acetylcysteine Attenuates the Increasing Severity of Distant Organ Liver Dysfunction after Acute Kidney Injury in Rats Exposed to Bisphenol A. Antioxidants (Basel) 8:497. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox8100497
Devulder J (2010) Flupirtine in pain management: pharmacological properties and clinical use. CNS Drugs 24:867–881. https://doiorg.publicaciones.saludcastillayleon.es/10.2165/11536230-000000000-00000
Simiand J, Keane PE, Biziere K, Soubrie P (1989) Comparative study in mice of tetrazepam and other centrally active skeletal muscle relaxants. Arch Int Pharmacodyn Ther 297:272–285
Voicu VA, Mircioiu C, Plesa C et al (2019) Effect of a New Synergistic Combination of Low Doses of Acetylsalicylic Acid, Caffeine, Acetaminophen, and Chlorpheniramine in Acute Low Back Pain. Front Pharmacol 10:607. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2019.00607
Kelly F, O’Grady J, Champey Y (1990) Zopiclone Lancet 335:1033–1034. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0140-6736(90)91095-r
Noble S, Langtry HD, Lamb HM (1998) Zopiclone. An update of its pharmacology, clinical efficacy and tolerability in the treatment of insomnia. Drugs 55:277–302. https://doiorg.publicaciones.saludcastillayleon.es/10.2165/00003495-199855020-00015
Yue J-L, Chang X-W, Zheng J-W et al (2023) Efficacy and tolerability of pharmacological treatments for insomnia in adults: A systematic review and network meta-analysis. Sleep Med Rev 68:101746. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.smrv.2023.101746
Pick CG, Chernes Y, Rigai T et al (2005) The antinociceptive effect of zolpidem and zopiclone in mice. Pharmacol Biochem Behav 81:417–423. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pbb.2005.02.013
Bamgbade OA, Tai-Osagbemi J, Bamgbade DO et al (2022) Clonidine is better than zopiclone for insomnia treatment in chronic pain patients. J Clin Sleep Med 18:1565–1571. https://doiorg.publicaciones.saludcastillayleon.es/10.5664/jcsm.9930
Giron SE, Lai G, Griffis CA, Zhang SJ (2022) Demystifying Buprenorphine with Current Evidence-Based Practice in Acute and Chronic Pain Management. AANA J 90:225–233
Li B, Guo J, Zhou X et al (2023) The emerging role of pyroptosis in neuropathic pain. Int Immunopharmacol 121:110562. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2023.110562
Funding
This research was funded by National Natural Science Foundation of China (82271414, 82471410), Health Scientific Research Talents Special Project of Jilin Province (2023SCZ07, 2023SCZ01), Scientific and Technological Development Program of Jilin Province (YDZJ202301ZYTS017), Jilin Industrial Technology Research and Development Project (2023C040-4, 2024C011-6), and Science and technology innovation platform construction project of Jilin Province (YDZJ202302CXJD060).
Author information
Authors and Affiliations
Contributions
Baolong Li and Xiongyao Zhou wrote the draft manuscript and designed studies; Kaiming Yu, Weiye Li, Jialu Sun, Weizhen Li and Ningning Wang conducted Western blot and qRT-PCR; Le Qi, Tuo Yang performed animal preparation and analyzed data; Xiaosong Gu, Rangjuan Cao and Shusen Cui supervised the experiments and revised the manuscript. All the authors read and approved the final manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
10194_2025_1953_MOESM1_ESM.tif
Supplementary Material 1. Supplementary Fig. 1. WGCNA and pathway enrichment results for SNI and Sham groups. (A) Results of GO analysis of differentially expressed genes. (B) Sample dendrogram and trait heatmap between SNI and Sham groups. (C, D) Soft-thresholding power analysis is used to determine the appropriate power for network construction. (E) Module-trait relationships for gene expression modules in relation to the experimental conditions. (F) Scatter plot of module membership versus gene significance for the yellow module. (G) Heatmap of the correlation matrix for different modules. (H) Dendrogram of gene modules and associated heatmap. (I) Results of GO analysis of yellow module genes. (J) KEGG showing important biological pathways associated with the yellow module genes.
10194_2025_1953_MOESM2_ESM.tif
Supplementary Material 2. Supplementary Fig. 2. Expression levels of UCP2 and TSPO were assessed in both Sham and SNI groups, along with an analysis of c-Fos and pyroptosis-related proteins following TSPO modulation. (A, B) Expression of UCP2 and TSPO in Sham versus SNI group, n = 6 per group. (C) Number of c-Fos+ neurons after PK 11195 treatment, n = 6 per group. (D-G) Quantification of the relative expression of TSPO, NLRP3, Caspase-1, and GSDMD-N after PK 11195 treatment, n = 3 per group. (H-K) Quantification of the relative expression of TSPO, NLRP3, Caspase-1, and GSDMD-N after Ro 5-4864 treatment, n = 3 per group. (L) Number of c-Fos+ neurons after Ro 5-4864 treatment, n = 6 per group. Data are represented as mean ± SEM, **p < 0.01, ***p < 0.001.
10194_2025_1953_MOESM3_ESM.tif
Supplementary Material 3. Supplementary Fig. 3. Genetic upregulation of TSPO expression alleviates neuropathic pain by decreasing astrocytic pyroptosis. (A) Heatmap showing differential expression of PCR array mitochondria-related genes in different groups. (B) Relative expression of TSPO after AS-TSPO AAV treatment in sham and SNI mice. (C) Number of c-Fos+ neurons after AS-TSPO treatment, n = 6 per group. (D-K) Quantification of the relative expression of TSPO, NLRP3, Caspase-1, GSDMD-N, p-AMPK, PGC-1, DRP1, MFN2 and TFAM after AS-TSPO treatment, n = 3 per group. (L) The copy number of mtDNA in the indicated groups, n = 6 per group. Data are represented as mean ± SEM, ***p < 0.001.
10194_2025_1953_MOESM4_ESM.tif
Supplementary Material 4. Supplementary Fig. 4. TSPO regulates the AMPK-PGC-1α pathway to reduce pyroptosis and mitochondrial dysfunction following SNI. (A-F) Quantification of the relative expression of TSPO, p-AMPK, PGC-1, DRP1, MFN2 and TFAM in different groups following AMPK inhibition, n = 3 per group. (G-I) Levels of antioxidant stress markers (GSH, SOD) and oxidative stress marker (MDA) in different groups, n = 6 per group. (J) The copy number of mtDNA in the indicated groups, n = 6 per group. (K-M) Quantification of the relative expression of NLRP3, Caspase-1, and GSDMD-N in different groups following AMPK inhibition, n = 3 per group. (N) Number of c-Fos+ neurons in different groups following AMPK inhibition, n = 6 per group. Data are represented as mean ± SEM, *p < 0.05, **p < 0.01, ***p < 0.001.
10194_2025_1953_MOESM5_ESM.tif
Supplementary Material 5. Supplementary Fig. 5. Impact of TSPO on mitochondrial health and pyroptosis in astrocytic models. (A) Quantification of PI positive cells after ATP+LPS treatment, n = 6 per group. (B) LDH release in culture supernatants after ATP+LPS treatment, n = 6 per group. (C-F) Quantification of the relative expression of TSPO, NLRP3, Caspase-1, and GSDMD-N after ATP+LPS treatment, n = 3 per group. (G) Quantification of relative fluorescence intensity of MitoSOX after ATP+LPS treatment, n = 6 per group. (H) Quantification of relative fluorescence intensity of ROS after ATP+LPS treatment, n = 6 per group. (I) Quantitative analysis of the relative intensity of JC-1 after ATP+LPS treatment, n = 6 per group. (J-L) Levels of antioxidant stress markers (GSH, SOD) and oxidative stress marker (MDA) in different groups, n = 6 per group. (M) The copy number of mtDNA after ATP+LPS treatment, n = 6 per group. (N-R) Quantification of relative expression of p-AMPK, PGC1, DRP1, MFN2 and TFAM after ATP+LPS treatment, n = 3 per group. Data are represented as mean ± SEM, ***p < 0.001.
10194_2025_1953_MOESM6_ESM.tif
Supplementary Material 6. Supplementary Fig. 6. TSPO-driven modulation of mitochondrial function and pyroptosis in astrocytes via the AMPK-PGC-1α pathway. (A) Viability of astrocytes after 2 h of Ro 5-4864 treatment, n = 6 per group. (B) Quantification of PI positive cells in different groups, n = 6 per group. (C) LDH release in culture supernatants in different groups, n = 6 per group. (D-I) Quantification of relative expression of TSPO, p-AMPK, PGC1, DRP1, MFN2 and TFAM in different groups, n = 3 per group. (J-L) Levels of antioxidant stress markers (GSH, SOD) and oxidative stress marker (MDA) in different groups of astrocytes, n = 6 per group. (M) The copy number of mtDNA in different groups, n = 6 per group. (N) Quantification of relative fluorescence intensity of ROS in different groups, n = 6 per group. (O) Quantification of relative fluorescence intensity of MitoSOX in different groups, n = 6 per group. (P-R) Quantification of the relative expression of NLRP3, Caspase-1 and GSDMD-N in different groups, n = 3 per group. Data are represented as mean ± SEM, **p < 0.01, ***p < 0.001.
10194_2025_1953_MOESM7_ESM.tif
Supplementary Material 7. Supplementary Fig. 7. The individual use of TSPO agonists and inhibitors did not have a significant impact on the experimental results. (A) Behavioral changes following TSPO inhibition by PK 11195 were assessed using von Frey testing at multiple time points (days 0, 3, 7, 14, and 21), n = 10 per group. (B, C) ELISA results showing the levels of IL-1β and IL-18 after PK 11195 administration, n = 6 per group. (D) Western blot showing the expression levels of TSPO, NLRP3, Caspase-1 and GSDMD-N after PK 11195 treatment. (E-H) Quantification of relative expression of TSPO, NLRP3, Caspase-1 and GSDMD-N after PK 11195 treatment, n = 3 per group. (I) Behavioral changes following TSPO activation by Ro 5-4864 were assessed using von Frey testing at multiple time points (days 0, 3, 7, 14, and 21), n = 10 per group. (J, K) ELISA results showing the levels of IL-1β and IL-18 after Ro 5-4864 administration, n = 6 per group. (L) Western blot showing the expression levels of TSPO, NLRP3, Caspase-1 and GSDMD-N after Ro 5-4864 treatment. (M-P) Quantification of relative expression of TSPO, NLRP3, Caspase-1 and GSDMD-N after Ro 5-4864 treatment, n = 3 per group. Data are represented as mean ± SEM. ns, no significant difference, ***p < 0.001.
10194_2025_1953_MOESM8_ESM.tif
Supplementary Material 8. Supplementary Fig. 8. On day 7 after SNI, the expression changes of pyroptosis-related proteins and mitochondrial-related proteins in the control, sham, and SNI groups were observed. (A) Western blot showing the expression levels of p-AMPK, PGC1, DRP1, MFN2 and TFAM in different groups. (B-F) Quantification of relative expression of T p-AMPK, PGC1, DRP1, MFN2 and TFAM in different groups, n = 3 per group. (G) Western blot showing the expression levels of TSPO, NLRP3, Caspase-1 and GSDMD-N a in different groups. (H-K) Quantification of relative expression of TSPO, NLRP3, Pro-Caspase 1, Caspase-1 and GSDMD-N in different groups, n = 3 per group. Data are represented as mean ± SEM. ns, no significant difference, ***p < 0.001.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Li, B., Yu, K., Zhou, X. et al. Increased TSPO alleviates neuropathic pain by preventing pyroptosis via the AMPK-PGC-1α pathway. J Headache Pain 26, 16 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10194-025-01953-0
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s10194-025-01953-0