Basic Study Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Psychiatry. Aug 19, 2024; 14(8): 1254-1266
Published online Aug 19, 2024. doi: 10.5498/wjp.v14.i8.1254
Botulinum toxin type A-targeted SPP1 contributes to neuropathic pain by the activation of microglia pyroptosis
Li-Ping Chen, Fu-Hai Ji, Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
Xiao-Die Gui, Wen-Di Tian, Jin-Zhao Huang, Department of Pain, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
Hou-Ming Kan, Faculty of Medicine, Macao University of Science and Technology, Macau 999078, China
ORCID number: Li-Ping Chen (0009-0002-2056-0681); Fu-Hai Ji (0000-0001-6649-665X).
Author contributions: Chen LP and Ji FH designed the research study; Chen LP, Gui XD, and Tian WD performed the research; Chen LP, Kan HM, and Huang JZ analyzed the data; Chen LP and Ji FH wrote the manuscript. All authors have read and approved the final manuscript.
Institutional animal care and use committee statement: All procedures involving animals were reviewed and approved by the Institutional Animal Care and Use Committee of the Xuzhou Medical University (No. SC225179).
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: No additional data are available.
ARRIVE guidelines statement: The authors have read the ARRIVE guidelines, and the manuscript was prepared and revised according to the ARRIVE guidelines.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Fu-Hai Ji, MD, Professor, Department of Anesthesiology, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215006, Jiangsu Province, China. jifuhai@suda.edu.cn
Received: April 3, 2024
Revised: May 29, 2024
Accepted: July 2, 2024
Published online: August 19, 2024
Processing time: 130 Days and 18.4 Hours

Abstract
BACKGROUND

Neuropathic pain (NP) is the primary symptom of various neurological conditions. Patients with NP often experience mood disorders, particularly depression and anxiety, that can severely affect their normal lives. Microglial cells are associated with NP. Excessive inflammatory responses, especially the secretion of large amounts of pro-inflammatory cytokines, ultimately lead to neuroinflammation. Microglial pyroptosis is a newly discovered form of inflammatory cell death associated with immune responses and inflammation-related diseases of the central nervous system.

AIM

To investigate the effects of botulinum toxin type A (BTX-A) on microglial pyroptosis in terms of NP and associated mechanisms.

METHODS

Two models, an in vitro lipopolysaccharide (LPS)-stimulated microglial cell model and a selective nerve injury model using BTX-A and SPP1 knockdown treatments, were used. Key proteins in the pyroptosis signaling pathway, NLRP3-GSDMD, were assessed using western blotting, real-time quantitative polymerase chain reaction, and immunofluorescence. Inflammatory factors [interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α] were assessed using enzyme-linked immunosorbent assay. We also evaluated microglial cell proliferation and apoptosis. Furthermore, we measured pain sensation by assessing the delayed hind paw withdrawal latency using thermal stimulation.

RESULTS

The expression levels of ACS and GSDMD-N and the mRNA expression of TNF-α, IL-6, and IL-1β were enhanced in LPS-treated microglia. Furthermore, SPP1 expression was also induced in LPS-treated microglia. Notably, BTX-A inhibited SPP1 mRNA and protein expression in the LPS-treated microglia. Additionally, depletion of SPP1 or BTX-A inhibited cell viability and induced apoptosis in LPS-treated microglia, whereas co-treatment with BTX-A enhanced the effect of SPP1 short hairpin (sh)RNA in LPS-treated microglia. Finally, SPP1 depletion or BTX-A treatment reduced the levels of GSDMD-N, NLPRP3, and ASC and suppressed the production of inflammatory factors.

CONCLUSION

Notably, BTX-A therapy and SPP1 shRNA enhance microglial proliferation and apoptosis and inhibit microglial death. It improves pain perception and inhibits microglial activation in rats with selective nerve pain.

Key Words: Botulinum toxin A, SPP1, Microglia, Pyroptosis, Neuropathic pain

Core Tip: Neuropathic pain (NP) arises from structural lesions that induce functional abnormalities in the central and peripheral nervous systems. This condition plagues approximately 10% of the population. In this study, we found that microglial pyroptosis was closely correlated with NP progression and that botulinum toxin type A (BTX-A) treatment notably alleviated this. Our mechanistic study identified that SPP1 may be positively correlated with microglial pyroptosis and inflammation during NP and that it is targeted by BTX-A. These findings provide novel evidence for the application of BTX-A in the treatment of NP.



INTRODUCTION

Neuropathic pain (NP) arises from structural lesions that induce functional abnormalities in the central and peripheral nervous systems. This condition is prevalent in an estimated 10% of the general population[1] and constitutes a frequent disorder. Patients with NP often experience mood disorders, such as depression and anxiety. Chronic pain can lead to depressive symptoms, such as low mood, loss of interest, sleep disturbances, and reduced energy. The association between NP and depression has been confirmed in multiple studies. For instance, a review by Meda et al[2] indicated a high prevalence of depression among patients with NP, and antidepressants were effective in treating chronic pain. Additionally, structural and functional changes in the brain regions associated with emotion regulation, such as the prefrontal cortex, amygdala, and hippocampus, have been observed in patients with NP, similar to those observed in patients with depression[3]. Furthermore, anxiety is a common comorbidity among patients with NP. Chronic pain may lead to a persistent state of anxiety, which could be associated with adaptive changes in the neural circuits involved in the stress response[4]. Notably, NP is the primary symptom of various neurological conditions, including post-herpetic and trigeminal neuralgia, encompassing these painful states[1]. Effective management of NP poses a significant therapeutic challenge for clinicians. Various pharmacological and non-pharmacological interventions have been suggested to yield varying degrees of benefit[5]. Pharmacological approaches include anticonvulsants, nonsteroidal anti-inflammatory drugs, antidepressants, and opioids despite the potential limitations arising from their side effects[6]. Surprisingly, < 60% of patients experience partial relief from recently approved agents targeting NP[7]. Therefore, the search for novel treatments and therapeutic targets commonly used for various indications persists.

Microglia play a dual role in preventing tissue damage or responding to stimuli caused by pathogen infections[8,9]. Alterations in microglial signaling pathways associated with NP involve inflammatory reactions[10]. Excessive inflammatory responses, especially the secretion of abundant pro-inflammatory cytokines, ultimately lead to neural inflammation[11,12]. Microglial pyroptosis, a newly discovered form of inflammatory cell death, is associated with immune responses and inflammation-related disorders of the central nervous system, including spinal cord injury and depression[13,14].

Initially limited to specific neurological disturbances, botulinum toxin type A (BTX-A) has been extensively used in various medical domains, including neurological, urological, gastroenterological, surgical, dermatological, and cosmetic applications. Within neurorehabilitation, BTX serves as a valuable adjunct to other interventions, particularly in aiding the treatment of individuals with neurological disability by primarily targeting spasticity reduction[15]. While historically associated with motor neurons, research indicates the potential entry of BTX into diverse neuron types, prompting studies exploring the efficacy of BTX-A in treating neurological disorders not solely associated with muscle hyperactivity[16-18]. Furthermore, BTX-A has been used to manage several painful conditions, including NP[19-22]. Notably, although both BTX-A and BTX-B are commercially available, clinical investigations have predominantly focused on the use of BTX-A for pain relief. In this study, we determined the effects of BTX-A on microglial growth and pyroptosis and investigated their potential mechanisms.

MATERIALS AND METHODS
Cell culture

Mouse glial cells BV2 (adherent cells) were cultured in Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum and 1% penicillin/streptomycin. The cells were cultured in a 37 °C incubator with 5% CO2. For lipopolysaccharide (LPS) induction, BV2 cells were seeded in six-well plates and cultured to 70 % confluence. Subsequently, LPS (5 mg/mL) was added to the culture medium and incubated for 8 hours. For BTX-A treatment, BTX-A (0.1 U/mL) was added to the culture medium for 24 hours.

Cell transfection

Lipofectamine 2000 reagent was used to conduct RNA-silencing experiments on microglia, according to the manufacturer’s protocol. Briefly, cells were seeded in a six-well plate and incubated overnight at 37 °C in a cell culture incubator until they reached 70% confluence. The culture medium was replaced with serum-free medium for 2 hours to induce cell starvation. Subsequently, 200 pmol of short hairpin (sh)RNA was mixed with lipofectamine 2000 in 50 μL of Opti-MEM medium. The mixture was incubated at room temperature for 15 minutes and then added to a cell culture medium. Cells were incubated at 37 °C for 48 hours and collected for further experimentation.

RNA extraction and polymerase chain reaction assay

RNA was extracted using TRIzol reagent (Invitrogen, Waltham, MA, United States). RNA was reverse transcribed to cDNA using a first-strand synthesis kit (TransGen, China). Subsequently, RNA was quantified using the SYBR Green kit (Takara, China) and calculated using the 2-∆∆Ct method. The primers used in this study were as follows: Tumor necrosis factor (TNF)-α sense: 5’-CAGGCGGTGCCTATGTCTC-3’; TNF-α anti-sense: 5’-CGATCACCCCGAAGTTCAGTAG-3’. Tnterleukin (IL)-6 sense: 5’-CTGCAAGAGACTTCCATCCAG-3’; IL-6 anti-sense: 5’-AGTGGTATAGACAGGTCTGTTGG-3’. IL-1β sense: 5’-GAAATGCCACCTTTTGACAGTG-3’; IL-1β anti-sense: 5’-TGGATGCTCTCATCAGGACAG-3’. Spp1 sense: 5’-AGAGCGGTGAGTCTAAGGAGT-3’; Spp1 anti-sense: 5’-TGCCCTTTCCGTTGTTGTCC-3’. GAPDH sense: 5’-GGAGCGAGATCCCTCCAAAAT-3’; GAPDH anti-sense: 5’-GGCTGTTGTCATACTTCTCATGG-3’.

Cell culture

Cells of different treatments were collected and suspended in complete media, and 100 μL cell suspension buffer was added to each well of the 96-well plate so that the number of cells per well was approximately 104, with five replicates in each group. The 96-well plate was incubated at 37 °C for 24 hours, and 10 μL of cell counting kit-8 reagent (Beyotime, China) was added to each well and cultured for 2 hours. The absorbance of each well was measured at 450 nm using a microplate reader (Thermo, Waltham, MA, United States).

Cell apoptosis

Apoptosis was detected using an Annexin V/propidium iodide apoptosis detection kit (Beyotime, China). Cells were collected and re-suspended in 1 × binding buffer at a density of approximately 1 × 106 cells/mL. Subsequently, 5 μL each of FITC-Annexin V and propidium iodide staining reagent were added into each well and incubated for 30 minutes in the dark. The cells were then centrifuged at 800 rpm for 5 minutes, re-suspended with 500 μL phosphate-buffered saline (PBS), and examined using a flow cytometer (BD Biosciences, United States).

Spared nerve injury surgery

The spared nerve injury (SNI) model was constructed as previously described[23]. Sprague-Dawley rats were purchased from the Beijing Vital River Laboratory. Animals were anesthetized with 100 mg/kg ketamine, and an incision was made on the thigh skin, reaching through the biceps femoris muscle to expose the three terminal branches of the sciatic nerve: The sural, common peroneal, and tibial nerves. The common peroneal and tibial nerves were securely ligated using 6.0 silk and then sectioned distal to the ligation, removing 2-4 mm of the distal nerve stump to separate the layers of the muscle and skin. Subsequently, BoNT/A (10 U/kg/day) was injected into the cheek for three consecutive days for treatment. Behavioral assessments commenced the day after surgery and continued for 14 days after surgery.

Thermal stimulation

The thermal withdrawal thresholds were determined following established protocols[19]. Animals were given 10 to 15 minutes to acclimate to the apparatus (Ugo Basile, Varese, Italy) before testing commenced. The rats were housed in a clear plastic chamber (18 cm × 29 cm × 12.5 cm) with a glass floor for 5 minutes to familiarize themselves with the environment. Initially, the rats displayed exploratory behavior; however, they later settled and stood quietly with occasional bouts of grooming. After acclimation, a radiant heat source was placed directly beneath the hind paw through a glass floor. Each trial began when the switch activated the radiant heat source, and an electronic timer was initiated. This assay measured the latency in seconds until hind paw withdrawal. The heat intensity was calibrated to provide an average latency of 8-10 seconds in naive, untreated animals; a maximum cut-off value of 20 seconds was established to prevent tissue injury. For each animal, the withdrawal latency was calculated as the average of three separate determinations, with at least 2 minutes intervals between trials.

Tissue collection

The rats were anesthetized with ketamine and transcardially perfused. Brief perfusion was conducted using a saline solution containing 5000 IU/mL heparin, followed by perfusion with a 4 % PFA solution. The glabrous skin of the hind paw and spinal cord were then dissected, fixed in PFA solution for 2 hours, and subsequently transferred into 30% sucrose solution for at least 24 hours. Subsequently, tissues were sectioned into 40 μm thick slices.

Enzyme-linked immunosorbent assay

The levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β) were detected by enzyme-linked immunosorbent assay kits (Elabscience, China) in accordance with the manufacturer’s protocol.

Immunofluorescence staining

The cells and tissue samples were incubated with 4% paraformaldehyde for 10 minutes and then with 0.1% Triton X-100 in PBS solution for 10 minutes. The cells were incubated with PBS containing 1% bovine serum albumin for 30 minutes to block nonspecific proteins. The samples were incubated with anti-NLRP3 and anti-ASC specific antibodies overnight in a refrigerator at 4 °C. Fluorescent secondary antibodies were then incubated for 1 hours. Nuclei were stained with a 4’,6-diamidino-2-phenylindole staining solution for 10 minutes. The images were captured using a confocal microscope (Nikon, Tokyo, Japan).

Statistical analyses

Data presented in this study are the averages of three independent experiments. Statistical analyses were conducted using SPSS 20.0 and GraphPad Prism 7.0 software. Statistical significance was defined based on standard deviation. Student’s t-test or one-way analysis of variance were used for statistical analyses between two or multiple groups. Statistical significance was set at P < 0.05.

RESULTS
SPP1 and pyroptosis are activated in LPS-induced microglia

To investigate the correlation of SPP1 with microglia pyroptosis, the levels of pyroptosis markers, inflammatory factors, and SPP1 were analyzed in LPS-treated microglia. The expression levels of ACS and GSDMD-N were enhanced in LPS-treated microglia (Figure 1A). Consistently, the mRNA expression of TNF-α, IL-6, and IL-1β was upregulated in LPS-treated microglia (Figure 1B-D). Importantly, SPP1 expression was induced in LPS-treated microglia (Figure 1E), indicating that SPP1 expression may positively correlate with microglial pyroptosis and inflammation.

Figure 1
Figure 1 SPP1 and pyroptosis are activated in lipopolysaccharide-induced microglia. A-E: The microglia were treated with lipopolysaccharide. The expression of ACS, GSDMD, and GSDMD-N was measured by western blot (A). The levels of tumor necrosis factor-α, interleukin (IL)-6, and IL-1β were detected by quantitative real-time polymerase chain reaction (B-D). The expression of SPP1 was analyzed by quantitative real-time polymerase chain reaction (E). aP < 0.001. LPS: Lipopolysaccharide; TNF: Tumor necrosis factor; IL: Interleukin; NC: Negative control.
SPP1 targeted by BTX-A promotes proliferation and represses apoptosis of LPS-induced microglia

BTX-A is a gram-positive anaerobic Clostridium botulinum exotoxin that is widely used clinically for aesthetics and dystonia. Some clinical studies have provided evidence of the effect of BTX-A on the attenuation of NP. Our data showed that BTX-A inhibited SPP1 mRNA and protein expression in LPS-treated microglia (Figure 2A and B). In addition, the effectiveness of SPP1 depletion by shRNA was validated in microglia (Figure 2A and B). Functionally, the depletion of SPP1 or BTX-A inhibited cell viability and induced apoptosis of LPS-treated microglia, and co-treatment with BTX-A enhanced the effect of SPP1 shRNA on LPS-treated microglia (Figure 2C-E), indicating that SPP1 targeted by BTX-A promotes proliferation and represses apoptosis in LPS-induced microglia.

Figure 2
Figure 2 SPP1 targeted by botulinum toxin type A promotes proliferation and represses apoptosis of lipopolysaccharide-induced microglia. A-E: Lipopolysaccharide-treated microglia were treated with control short hairpin RNA (shRNA) or SPP1 shRNA, or co-treated with botulinum toxin type A. The expression of SPP1 was analyzed by quantitative real-time polymerase chain reaction (A). The expression of SPP1 was measured by western blot (B). Cell viability was detected by cell counting kit-8 assay (C). Cell apoptosis was examined by flow cytometry (D and E). aP < 0.05, bP < 0.01, cP < 0.001. BTX-A: Botulinum toxin type A.
SPP1 targeted by BTX-A contributes to pyroptosis of LPS-induced microglia

We assessed the effect of SPP1 targeted by BTX-A on microglial pyroptosis. We observed that the levels of GSDMD-N, NLPRP3, and ASC were repressed by SPP1 knockdown or BTX-A treatment in LPS-treated microglia and that co-treatment with BTX-A enhanced the effect of SPP1 shRNA on pyroptosis in LPS-treated microglia (Figure 3A-C). Consistently, SPP1 depletion or BTX-A treatment was able to inhibit the levels of TNF-α, IL-6, and IL-1β in LPS-treated microglia, and SPP1 shRNA and BTX-A co-treatment presented a more observable effect on the phenotype (Figure 3D-F), indicating that SPP1 targeted by BTX-A contributes to pyroptosis of LPS-induced microglia.

Figure 3
Figure 3 SPP1 targeted by botulinum toxin type A contributes to pyroptosis of lipopolysaccharide-induced microglia. A-F: Lipopolysaccharide-treated rat microglia were treated with control short hairpin RNA (shRNA) or SPP1 shRNA, or co-treated with botulinum toxin type A. The expression of GSDMD-N was measured by western blot (A). The levels of NLRP3 and ASC were detected by immunofluorescence (B and C). The levels of tumor necrosis factor-α, interleukin (IL)-6, and IL-1β were analyzed by enzyme-linked immunosorbent assay (D-F). aP < 0.05, bP < 0.01, cP < 0.001. TNF: Tumor necrosis factor; IL: Interleukin; BTX-A: Botulinum toxin type A.
SPP1 targeted by BTX-A enhances microglial pyroptosis during NP induced by SNI in rat

Next, we evaluated the effect of SPP1 targeting by BTX-A on NP in a rat model of SNI. We observed that the mechanical withdrawal threshold and thermal withdrawal latency were promoted by SPP1 knockdown or BTX-A in rats with SNI, whereas co-treatment with BTX-A enhanced the effect of SPP1 shRNA (Figure 4A and B). The levels of SPP1, IBA1, ACS, and GSDMD-N were inhibited by SPP1 depletion or BTX-A in rats with SNI, and co-treatment with BTX-A and SPP1 depletion had a more observable effect (Figure 4C and D). Consistently, the levels of TNF-α, IL-6, and IL-1β were repressed by BTX-A or SPP1 depletion in rats with SNI, and co-treatment with BTX-A could enhance the effect of SPP1 depletion (Figure 4E-G).

Figure 4
Figure 4 SPP1 targeted by botulinum toxin type A enhances microglia pyroptosis during neuropathic pain induced by spared nerve injury in rat. A-G: The spared nerve injury model was established in SD rats and the rats were treated with control short hairpin RNA (shRNA) or SPP1 shRNA, or co-treated with botulinum toxin type A. The mechanical withdrawal threshold and thermal withdrawal latency were analyzed in the rats (A and B). The expression of SPP1, ACS, and GSDMD-N was measured by western blot in spinal cord of the rats (C). The levels of IBA1 and SPP1 were detected by immunofluorescence in spinal cord of the rats (D). The levels of tumor necrosis factor-α, interleukin (IL)-6, and IL-1β were analyzed by enzyme-linked immunosorbent assay (E-G). aP < 0.05, bP < 0.01, cP < 0.001. TNF: Tumor necrosis factor; IL: Interleukin; BTX-A: Botulinum toxin type A.
DISCUSSION

In the present study, we investigated the potential mechanisms by which BTX-A alleviates PN. By establishing models of LPS-induced microglial pyroptosis and SNI, we demonstrated a significant elevation in SPP1 within microglial cells. Moreover, SPP1 knockdown inhibited microglial pyroptosis and alleviated pain in rats with nerve injury. Microglial pyroptosis is a novel form of cell death mediated by inflammasomes and is associated with immune and inflammatory diseases of the central nervous system, including depression, radiation-induced brain injury, NP, and spinal cord injury[24-27]. Notably, NLRP3 inflammasomes play a crucial role in microglial activation, are predominantly expressed in microglial cells, and have been extensively studied in the context of NP[28-31]. Recent studies have indicated the involvement of NLRP3 and pyroptosis in depression and anxiety, two primary symptoms of NP[32]. For example, Li et al[33] reported that isoliquiritin protects primary microglia by suppressing NLRP3-mediated pyroptosis and possesses potent antidepressant properties[33]. The primary components of the NLRP3 inflammasome include the pattern recognition receptor NLRP3, adaptor protein ASC, and pro-caspase-1 enzyme[34]. Activation of NLRP3 by various stimuli and ligands triggers the assembly of the NLRP3-ASC inflammasome complex, activating caspase-1. Activated caspase-1 cleaves IL-1β and IL-18, increasing the secretion of mature IL-1β and IL-18[34]. Additionally, activated caspase-1 cleaves GSDMD to produce an N-terminal fragment, which forms pores in the cell membrane. This leads to the secretion of IL-1β and IL-18 and an influx of water molecules, ultimately resulting in excessive inflammatory responses and cell pyroptosis[35]. In this study, we observed that BTX-A treatment alleviated the production of pro-inflammatory cytokines and repressed the level of GSDMD-N in LPS-induced microglia, suggesting that BTX-A affects NP by modulating microglial pyroptosis.

Aberrantly activated microglia and innate immune cells exacerbate inflammatory pain by upregulating inflammatory factors[36,37]. A wealth of research indicates that NLRP3 inflammasome activation triggers functional alterations in microglia, contributing to the onset and progression of chronic pain[24,38]. Previous studies have suggested that silicate agents modulate microglial pyroptosis by inhibiting the activation of the NLRP3-caspase-1-GSDMD pathway, thereby regulating chronic neuroinflammation and NP[24]. Moreover, both acute and chronic pain have associations with pyroptosis in inflammatory cells[39,40]. Consequently, targeting microglial pyroptosis holds promise in alleviating inflammatory damage and mitigating NP. Accumulating evidence suggests a role for the NLRP3 inflammasome in inflammatory pain[41,42]. For example, NLRP3 knockout male mice were protected from surgery-induced postoperative inflammation and neuron-sensitized postoperative inflammatory pain[43]. Furthermore, complete Freund’s adjuvant injection induced the activation of the NLRP3 inflammasome in claw skin macrophages and promoted the maturation of inflammatory cytokine IL-1β through the cleavage of caspase-1[44]. Therefore, NLRP3-mediated pyroptosis may mediate the therapeutic effects of BTX-A in NP.

Notably, BTX-A has been reported to modulate pain. Currently, the only approved use of BTX-A in relation to pain is for the treatment of chronic migraine. However, controlled clinical studies have shown promising results for neuropathic and other chronic pain disorders[45,46]. Several studies have revealed potential mechanisms underlying BTX-A treatment. For example, in the sensory ganglia of injured nerves, BoNT/A reduces pain-evoked upregulated protein expression of nociception-related ion channels such as TRPV1 and purinoceptor P2X3, and reduces the mRNA expression of pronociceptive peptides such as preprodynorphin[47]. Intraplantar injection of BTX-A in neuropathic mice improved the sciatic index and weight bearing, along with increased cell division cycle 2 protein expression and Schwann cell proliferation and maturation[48]. In this study, we found that the RNA and protein levels of SPP1 were significantly increased during LPS-induced pyroptosis of microglia. Knockdown of SPP1 synergistically downregulated the expression of inflammatory factors and pyroptosis signaling with BTX-A treatment to inhibit microglial viability and improve pain in rats with nerve injury. Notably, SPP1, also known as osteopontin, has been widely studied in several diseases, and is a multifunctional glycoprotein that was originally thought to be a pro-inflammatory cytokine secreted by T cells. It was later found to be expressed in macrophages in different tissues and is associated with the active clearance of apoptotic cells, chemotaxis, and macrophage migration[49,50]. In the brain, SPP1 expression is highly regulated in a spatiotemporal and cell type-specific manner, depending on the environment, age, and brain region. In the perinatal and prenatal brain, SPP1 is expressed by microglia. It is associated with the axon tract of the corpus callosum. In contrast, in the adult posterior brain, SPP1 expression is limited to glutaminergic and γ-aminobutyric acid neurons[51-55]. Consistent with previous reports, we elucidated the important role of SPP1 in BTX-A-induced NP relief.

CONCLUSION

In the present study, we revealed the therapeutic potential of BTX-A against NP and identified SPP1 as a potential target of BTX-A. Our findings provide novel evidence for the application of BTX-A to NP. These findings provide novel insights into the treatment of NP and may contribute to the improvement of psychiatric disorders, such as depression and anxiety, which are correlated with NP. The exploration of other potential regulatory mechanisms and validation of BTX-A for clinical use are necessitated in future studies.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Psychiatry

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Giannouli V S-Editor: Wang JJ L-Editor: A P-Editor: Yuan YY

References
1.  van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain. 2014;155:654-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 799]  [Cited by in F6Publishing: 957]  [Article Influence: 87.0]  [Reference Citation Analysis (0)]
2.  Meda RT, Nuguru SP, Rachakonda S, Sripathi S, Khan MI, Patel N. Chronic Pain-Induced Depression: A Review of Prevalence and Management. Cureus. 2022;14:e28416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
3.  Descalzi G, Mitsi V, Purushothaman I, Gaspari S, Avrampou K, Loh YE, Shen L, Zachariou V. Neuropathic pain promotes adaptive changes in gene expression in brain networks involved in stress and depression. Sci Signal. 2017;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 110]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
4.  Argoff CE. The coexistence of neuropathic pain, sleep, and psychiatric disorders: a novel treatment approach. Clin J Pain. 2007;23:15-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 104]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
5.  Magrinelli F, Zanette G, Tamburin S. Neuropathic pain: diagnosis and treatment. Pract Neurol. 2013;13:292-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
6.  Finnerup NB, Otto M, McQuay HJ, Jensen TS, Sindrup SH. Algorithm for neuropathic pain treatment: an evidence based proposal. Pain. 2005;118:289-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 959]  [Cited by in F6Publishing: 775]  [Article Influence: 40.8]  [Reference Citation Analysis (0)]
7.  Dray A. Neuropathic pain: emerging treatments. Br J Anaesth. 2008;101:48-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 123]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
8.  Inoue K, Tsuda M. Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential. Nat Rev Neurosci. 2018;19:138-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 365]  [Article Influence: 60.8]  [Reference Citation Analysis (0)]
9.  Fiore NT, Debs SR, Hayes JP, Duffy SS, Moalem-Taylor G. Pain-resolving immune mechanisms in neuropathic pain. Nat Rev Neurol. 2023;19:199-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
10.  Kohno K, Shirasaka R, Yoshihara K, Mikuriya S, Tanaka K, Takanami K, Inoue K, Sakamoto H, Ohkawa Y, Masuda T, Tsuda M. A spinal microglia population involved in remitting and relapsing neuropathic pain. Science. 2022;376:86-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 92]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
11.  Ding R, Li H, Liu Y, Ou W, Zhang X, Chai H, Huang X, Yang W, Wang Q. Activating cGAS-STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis. J Neuroinflammation. 2022;19:137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 58]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
12.  Hua T, Yang M, Song H, Kong E, Deng M, Li Y, Li J, Liu Z, Fu H, Wang Y, Yuan H. Huc-MSCs-derived exosomes attenuate inflammatory pain by regulating microglia pyroptosis and autophagy via the miR-146a-5p/TRAF6 axis. J Nanobiotechnology. 2022;20:324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 49]  [Reference Citation Analysis (0)]
13.  Xu S, Wang J, Zhong J, Shao M, Jiang J, Song J, Zhu W, Zhang F, Xu H, Xu G, Zhang Y, Ma X, Lyu F. CD73 alleviates GSDMD-mediated microglia pyroptosis in spinal cord injury through PI3K/AKT/Foxo1 signaling. Clin Transl Med. 2021;11:e269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 108]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
14.  Wu X, Wan T, Gao X, Fu M, Duan Y, Shen X, Guo W. Microglia Pyroptosis: A Candidate Target for Neurological Diseases Treatment. Front Neurosci. 2022;16:922331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
15.  Intiso D. Therapeutic use of botulinum toxin in neurorehabilitation. J Toxicol. 2012;2012:802893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
16.  Gilio F, Iacovelli E, Frasca V, Gabriele M, Giacomelli E, Picchiori F, Soldo P, Cipriani AM, Ruoppolo G, Inghilleri M. Botulinum toxin type A for the treatment of sialorrhoea in amyotrophic lateral sclerosis: a clinical and neurophysiological study. Amyotroph Lateral Scler. 2010;11:359-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
17.  Mancini F, Zangaglia R, Cristina S, Sommaruga MG, Martignoni E, Nappi G, Pacchetti C. Double-blind, placebo-controlled study to evaluate the efficacy and safety of botulinum toxin type A in the treatment of drooling in parkinsonism. Mov Disord. 2003;18:685-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 131]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
18.  Reid SM, Johnstone BR, Westbury C, Rawicki B, Reddihough DS. Randomized trial of botulinum toxin injections into the salivary glands to reduce drooling in children with neurological disorders. Dev Med Child Neurol. 2008;50:123-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 96]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
19.  Apalla Z, Sotiriou E, Lallas A, Lazaridou E, Ioannides D. Botulinum toxin A in postherpetic neuralgia: a parallel, randomized, double-blind, single-dose, placebo-controlled trial. Clin J Pain. 2013;29:857-864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 68]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
20.  Hu Y, Guan X, Fan L, Li M, Liao Y, Nie Z, Jin L. Therapeutic efficacy and safety of botulinum toxin type A in trigeminal neuralgia: a systematic review. J Headache Pain. 2013;14:72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
21.  Diener HC, Dodick DW, Turkel CC, Demos G, Degryse RE, Earl NL, Brin MF. Pooled analysis of the safety and tolerability of onabotulinumtoxinA in the treatment of chronic migraine. Eur J Neurol. 2014;21:851-859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
22.  Brown EA, Schütz SG, Simpson DM. Botulinum toxin for neuropathic pain and spasticity: an overview. Pain Manag. 2014;4:129-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
23.  Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain. 2000;87:149-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1478]  [Cited by in F6Publishing: 1619]  [Article Influence: 67.5]  [Reference Citation Analysis (0)]
24.  Chen R, Yin C, Fang J, Liu B. The NLRP3 inflammasome: an emerging therapeutic target for chronic pain. J Neuroinflammation. 2021;18:84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 57]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
25.  Cai X, Zhang ZY, Yuan JT, Ocansey DKW, Tu Q, Zhang X, Qian H, Xu WR, Qiu W, Mao F. hucMSC-derived exosomes attenuate colitis by regulating macrophage pyroptosis via the miR-378a-5p/NLRP3 axis. Stem Cell Res Ther. 2021;12:416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 64]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
26.  He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, Yang ZH, Zhong CQ, Han J. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res. 2015;25:1285-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1061]  [Cited by in F6Publishing: 1588]  [Article Influence: 176.4]  [Reference Citation Analysis (0)]
27.  Li J, Tian M, Hua T, Wang H, Yang M, Li W, Zhang X, Yuan H. Combination of autophagy and NFE2L2/NRF2 activation as a treatment approach for neuropathic pain. Autophagy. 2021;17:4062-4082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
28.  Zhang MD, Su J, Adori C, Cinquina V, Malenczyk K, Girach F, Peng C, Ernfors P, Löw P, Borgius L, Kiehn O, Watanabe M, Uhlén M, Mitsios N, Mulder J, Harkany T, Hökfelt T. Ca2+-binding protein NECAB2 facilitates inflammatory pain hypersensitivity. J Clin Invest. 2018;128:3757-3768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
29.  Colvin LA, Bull F, Hales TG. Perioperative opioid analgesia-when is enough too much? A review of opioid-induced tolerance and hyperalgesia. Lancet. 2019;393:1558-1568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 275]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
30.  Asami T, Ishii M, Fujii H, Namkoong H, Tasaka S, Matsushita K, Ishii K, Yagi K, Fujiwara H, Funatsu Y, Hasegawa N, Betsuyaku T. Modulation of murine macrophage TLR7/8-mediated cytokine expression by mesenchymal stem cell-conditioned medium. Mediators Inflamm. 2013;2013:264260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
31.  Huh Y, Ji RR, Chen G. Neuroinflammation, Bone Marrow Stem Cells, and Chronic Pain. Front Immunol. 2017;8:1014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 95]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
32.  Mokhtari T, Uludag K. Role of NLRP3 Inflammasome in Post-Spinal-Cord-Injury Anxiety and Depression: Molecular Mechanisms and Therapeutic Implications. ACS Chem Neurosci. 2024;15:56-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
33.  Li Y, Song W, Tong Y, Zhang X, Zhao J, Gao X, Yong J, Wang H. Isoliquiritin ameliorates depression by suppressing NLRP3-mediated pyroptosis via miRNA-27a/SYK/NF-κB axis. J Neuroinflammation. 2021;18:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 148]  [Article Influence: 49.3]  [Reference Citation Analysis (0)]
34.  Ren J, Liu N, Sun N, Zhang K, Yu L. Mesenchymal Stem Cells and their Exosomes: Promising Therapeutics for Chronic Pain. Curr Stem Cell Res Ther. 2019;14:644-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
35.  Chen HX, Liang FC, Gu P, Xu BL, Xu HJ, Wang WT, Hou JY, Xie DX, Chai XQ, An SJ. Exosomes derived from mesenchymal stem cells repair a Parkinson's disease model by inducing autophagy. Cell Death Dis. 2020;11:288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 137]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
36.  Su DJ, Li LF, Wang SY, Yang Q, Wu YJ, Zhao MG, Yang L. Pra-C exerts analgesic effect through inhibiting microglial activation in anterior cingulate cortex in complete Freund's adjuvant-induced mouse model. Mol Pain. 2021;17:1744806921990934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
37.  Yu S, Zhao G, Han F, Liang W, Jiao Y, Li Z, Li L. Muscone relieves inflammatory pain by inhibiting microglial activation-mediated inflammatory response via abrogation of the NOX4/JAK2-STAT3 pathway and NLRP3 inflammasome. Int Immunopharmacol. 2020;82:106355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
38.  He W, Long T, Pan Q, Zhang S, Zhang Y, Zhang D, Qin G, Chen L, Zhou J. Microglial NLRP3 inflammasome activation mediates IL-1β release and contributes to central sensitization in a recurrent nitroglycerin-induced migraine model. J Neuroinflammation. 2019;16:78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 123]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
39.  Wang G, Yuan J, Cai X, Xu Z, Wang J, Ocansey DKW, Yan Y, Qian H, Zhang X, Xu W, Mao F. HucMSC-exosomes carrying miR-326 inhibit neddylation to relieve inflammatory bowel disease in mice. Clin Transl Med. 2020;10:e113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 69]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
40.  Shiue SJ, Rau RH, Shiue HS, Hung YW, Li ZX, Yang KD, Cheng JK. Mesenchymal stem cell exosomes as a cell-free therapy for nerve injury-induced pain in rats. Pain. 2019;160:210-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 125]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
41.  Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol. 2021;22:550-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 463]  [Article Influence: 154.3]  [Reference Citation Analysis (0)]
42.  Wang L, Hauenstein AV. The NLRP3 inflammasome: Mechanism of action, role in disease and therapies. Mol Aspects Med. 2020;76:100889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 215]  [Article Influence: 53.8]  [Reference Citation Analysis (0)]
43.  Matsuoka Y, Yamashita A, Matsuda M, Kawai K, Sawa T, Amaya F. NLRP2 inflammasome in dorsal root ganglion as a novel molecular platform that produces inflammatory pain hypersensitivity. Pain. 2019;160:2149-2160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
44.  Gao F, Xiang HC, Li HP, Jia M, Pan XL, Pan HL, Li M. Electroacupuncture inhibits NLRP3 inflammasome activation through CB2 receptors in inflammatory pain. Brain Behav Immun. 2018;67:91-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 65]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
45.  Ranoux D, Attal N, Morain F, Bouhassira D. Botulinum toxin type A induces direct analgesic effects in chronic neuropathic pain. Ann Neurol. 2008;64:274-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 310]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
46.  Attal N, de Andrade DC, Adam F, Ranoux D, Teixeira MJ, Galhardoni R, Raicher I, Üçeyler N, Sommer C, Bouhassira D. Safety and efficacy of repeated injections of botulinum toxin A in peripheral neuropathic pain (BOTNEP): a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2016;15:555-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 136]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
47.  Xiao L, Cheng J, Zhuang Y, Qu W, Muir J, Liang H, Zhang D. Botulinum toxin type A reduces hyperalgesia and TRPV1 expression in rats with neuropathic pain. Pain Med. 2013;14:276-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
48.  Marinelli S, Luvisetto S, Cobianchi S, Makuch W, Obara I, Mezzaroma E, Caruso M, Straface E, Przewlocka B, Pavone F. Botulinum neurotoxin type A counteracts neuropathic pain and facilitates functional recovery after peripheral nerve injury in animal models. Neuroscience. 2010;171:316-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
49.  Rittling SR. Osteopontin in macrophage function. Expert Rev Mol Med. 2011;13:e15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 98]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
50.  Weber GF, Cantor H. The immunology of Eta-1/osteopontin. Cytokine Growth Factor Rev. 1996;7:241-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 102]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
51.  Kim JS, Kolesnikov M, Peled-Hajaj S, Scheyltjens I, Xia Y, Trzebanski S, Haimon Z, Shemer A, Lubart A, Van Hove H, Chappell-Maor L, Boura-Halfon S, Movahedi K, Blinder P, Jung S. A Binary Cre Transgenic Approach Dissects Microglia and CNS Border-Associated Macrophages. Immunity. 2021;54:176-190.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
52.  Zeisel A, Hochgerner H, Lönnerberg P, Johnsson A, Memic F, van der Zwan J, Häring M, Braun E, Borm LE, La Manno G, Codeluppi S, Furlan A, Lee K, Skene N, Harris KD, Hjerling-Leffler J, Arenas E, Ernfors P, Marklund U, Linnarsson S. Molecular Architecture of the Mouse Nervous System. Cell. 2018;174:999-1014.e22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1775]  [Cited by in F6Publishing: 1505]  [Article Influence: 250.8]  [Reference Citation Analysis (0)]
53.  Li Q, Cheng Z, Zhou L, Darmanis S, Neff NF, Okamoto J, Gulati G, Bennett ML, Sun LO, Clarke LE, Marschallinger J, Yu G, Quake SR, Wyss-Coray T, Barres BA. Developmental Heterogeneity of Microglia and Brain Myeloid Cells Revealed by Deep Single-Cell RNA Sequencing. Neuron. 2019;101:207-223.e10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 439]  [Cited by in F6Publishing: 604]  [Article Influence: 100.7]  [Reference Citation Analysis (0)]
54.  Shen X, Qiu Y, Wight AE, Kim HJ, Cantor H. Definition of a mouse microglial subset that regulates neuronal development and proinflammatory responses in the brain. Proc Natl Acad Sci U S A. 2022;119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
55.  Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A, Walker AJ, Gergits F, Segel M, Nemesh J, Marsh SE, Saunders A, Macosko E, Ginhoux F, Chen J, Franklin RJM, Piao X, McCarroll SA, Stevens B. Single-Cell RNA Sequencing of Microglia throughout the Mouse Lifespan and in the Injured Brain Reveals Complex Cell-State Changes. Immunity. 2019;50:253-271.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 808]  [Cited by in F6Publishing: 1154]  [Article Influence: 192.3]  [Reference Citation Analysis (0)]