Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.997
Filter
1.
BMC Musculoskelet Disord ; 25(1): 331, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38725009

ABSTRACT

BACKGROUND: The development of neuropathic pain (NP) is one of the reasons why the pain is difficult to treat, and microglial activation plays an important role in NP. Recently, platelet-rich plasma (PRP) has emerged as a novel therapeutic method for knee osteoarthritis (KOA). However, it's unclarified whether PRP has analgesic effects on NP induced by KOA and the underlying mechanisms unknown. PURPOSE: To observe the analgesic effects of PRP on NP induced by KOA and explore the potential mechanisms of PRP in alleviating NP. METHODS: KOA was induced in male rats with intra-articular injections of monosodium iodoacetate (MIA) on day 0. The rats received PRP or NS (normal saline) treatment at days 15, 17, and 19 after modeling. The Von Frey and Hargreaves tests were applied to assess the pain-related behaviors at different time points. After euthanizing the rats with deep anesthesia at days 28 and 42, the corresponding tissues were taken for subsequent experiments. The expression of activating transcription factor 3 (ATF3) in dorsal root ganglia (DRG) and ionized-calcium-binding adapter molecule-1(Iba-1) in the spinal dorsal horn (SDH) was detected by immunohistochemical staining. In addition, the knee histological assessment was performed by hematoxylin-eosin (HE) staining. RESULTS: The results indicated that injection of MIA induced mechanical allodynia and thermal hyperalgesia, which could be reversed by PRP treatment. PRP downregulated the expression of ATF3 within the DRG and Iba-1 within the SDH. Furthermore, an inhibitory effect on cartilage degeneration was observed in the MIA + PRP group only on day 28. CONCLUSION: These results indicate that PRP intra-articular injection therapy may be a potential therapeutic agent for relieving NP induced by KOA. This effect could be attributed to downregulation of microglial activation and reduction in nerve injury.


Subject(s)
Down-Regulation , Microglia , Neuralgia , Osteoarthritis, Knee , Platelet-Rich Plasma , Rats, Sprague-Dawley , Animals , Male , Neuralgia/therapy , Neuralgia/metabolism , Microglia/metabolism , Rats , Osteoarthritis, Knee/therapy , Activating Transcription Factor 3/metabolism , Ganglia, Spinal/metabolism , Disease Models, Animal , Injections, Intra-Articular , Calcium-Binding Proteins/metabolism , Iodoacetic Acid/toxicity , Microfilament Proteins
2.
FASEB J ; 38(10): e23661, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38733310

ABSTRACT

Itching is an aversive somatosensation that triggers the desire to scratch. Transient receptor potential (TRP) channel proteins are key players in acute and chronic itch. However, whether the modulatory effect of fibroblast growth factor 13 (FGF13) on acute and chronic itch is associated with TRP channel proteins is unclear. Here, we demonstrated that conditional knockout of Fgf13 in dorsal root ganglion neurons induced significant impairment in scratching behaviors in response to acute histamine-dependent and chronic dry skin itch models. Furthermore, FGF13 selectively regulated the function of the TRPV1, but not the TRPA1 channel on Ca2+ imaging and electrophysiological recordings, as demonstrated by a significant reduction in neuronal excitability and current density induced by TRPV1 channel activation, whereas TRPA1 channel activation had no effect. Changes in channel currents were also verified in HEK cell lines. Subsequently, we observed that selective modulation of TRPV1 by FGF13 required its microtubule-stabilizing effect. Furthermore, in FGF13 knockout mice, only the overexpression of FGF13 with a tubulin-binding domain could rescue TRP channel function and the impaired itch behavior. Our findings reveal a novel mechanism by which FGF13 is involved in TRPV1-dependent itch transduction and provide valuable clues for alleviating pathological itch syndrome.


Subject(s)
Fibroblast Growth Factors , Mice, Knockout , Microtubules , Pruritus , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Pruritus/metabolism , Pruritus/genetics , Animals , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Mice , Humans , HEK293 Cells , Microtubules/metabolism , Ganglia, Spinal/metabolism , Male , Mice, Inbred C57BL , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics
3.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731951

ABSTRACT

Distal sensory polyneuropathy (DSP) and distal neuropathic pain (DNP) remain significant challenges for older people with HIV (PWH), necessitating enhanced clinical attention. HIV and certain antiretroviral therapies (ARTs) can compromise mitochondrial function and impact mitochondrial DNA (mtDNA) replication, which is linked to DSP in ART-treated PWH. This study investigated mtDNA, mitochondrial fission and fusion proteins, and mitochondrial electron transport chain protein changes in the dorsal root ganglions (DRGs) and sural nerves (SuNs) of 11 autopsied PWH. In antemortem standardized assessments, six had no or one sign of DSP, while five exhibited two or more DSP signs. Digital droplet polymerase chain reaction was used to measure mtDNA quantity and the common deletions in isolated DNA. We found lower mtDNA copy numbers in DSP+ donors. SuNs exhibited a higher proportion of mtDNA common deletion than DRGs in both groups. Mitochondrial electron transport chain (ETC) proteins were altered in the DRGs of DSP+ compared to DSP- donors, particularly Complex I. These findings suggest that reduced mtDNA quantity and increased common deletion abundance may contribute to DSP in PWH, indicating diminished mitochondrial activity in the sensory neurons. Accumulated ETC proteins in the DRG imply impaired mitochondrial transport to the sensory neuron's distal portion. Identifying molecules to safeguard mitochondrial integrity could aid in treating or preventing HIV-associated peripheral neuropathy.


Subject(s)
DNA, Mitochondrial , HIV Infections , Humans , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Male , HIV Infections/metabolism , HIV Infections/virology , HIV Infections/genetics , Pilot Projects , Female , Middle Aged , Aged , Ganglia, Spinal/metabolism , Ganglia, Spinal/virology , Mitochondria/metabolism , Mitochondria/genetics , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Chain Complex Proteins/genetics , Peripheral Nerves/metabolism , Peripheral Nerves/virology , Peripheral Nerves/pathology , Adult , Sural Nerve/metabolism , Sural Nerve/pathology
4.
Traffic ; 25(5): e12936, 2024 May.
Article in English | MEDLINE | ID: mdl-38725127

ABSTRACT

Endosomal trafficking of TrkA is a critical process for nerve growth factor (NGF)-dependent neuronal cell survival and differentiation. The small GTPase ADP-ribosylation factor 6 (Arf6) is implicated in NGF-dependent processes in PC12 cells through endosomal trafficking and actin cytoskeleton reorganization. However, the regulatory mechanism for Arf6 in NGF signaling is largely unknown. In this study, we demonstrated that EFA6A, an Arf6-specific guanine nucleotide exchange factor, was abundantly expressed in PC12 cells and that knockdown of EFA6A significantly inhibited NGF-dependent Arf6 activation, TrkA recycling from early endosomes to the cell surface, prolonged ERK1/2 phosphorylation, and neurite outgrowth. We also demonstrated that EFA6A forms a protein complex with TrkA through its N-terminal region, thereby enhancing its catalytic activity for Arf6. Similarly, we demonstrated that EFA6A forms a protein complex with TrkA in cultured dorsal root ganglion (DRG) neurons. Furthermore, cultured DRG neurons from EFA6A knockout mice exhibited disturbed NGF-dependent TrkA trafficking compared with wild-type neurons. These findings provide the first evidence for EFA6A as a key regulator of NGF-dependent TrkA trafficking and signaling.


Subject(s)
ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors , Endosomes , Guanine Nucleotide Exchange Factors , Nerve Growth Factor , Neuronal Outgrowth , Receptor, trkA , Animals , PC12 Cells , Receptor, trkA/metabolism , Nerve Growth Factor/metabolism , Rats , Endosomes/metabolism , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Mice , Protein Transport , Ganglia, Spinal/metabolism , Mice, Knockout
5.
J Toxicol Sci ; 49(5): 241-248, 2024.
Article in English | MEDLINE | ID: mdl-38692911

ABSTRACT

Methylmercury is an environmental polluting organometallic compound that exhibits neurotoxicity, as observed in Minamata disease patients. Methylmercury damages peripheral nerves in Minamata patients, causing more damage to sensory nerves than motor nerves. Peripheral nerves are composed of three cell types: dorsal root ganglion (DRG) cells, anterior horn cells (AHCs), and Schwann cells. In this study, we compared cultured these three cell types derived from the rat for susceptibility to methylmercury cytotoxicity, intracellular accumulation of mercury, expression of L-type amino acid transporter 1 (LAT1), which transports methylmercury into cells, and expression of multidrug resistance-associated protein 2 (MRP2), which transports methylmercury-glutathione conjugates into the extracellular space. Of the cells examined, we found that DRG cells were the most susceptible to methylmercury with markedly higher intracellular accumulation of mercury. The constitutive level of LAT1 was higher and that of MRP2 lower in DRG cells compared with those in AHC and Schwann cells. Additionally, decreased cell viability caused by methylmercury was significantly reduced by either the LAT1 inhibitor, JPH203, or siRNA-mediated knockdown of LAT1. On the other hand, an MRP2 inhibitor, MK571, significantly intensified the decrease in the cell viability caused by methylmercury. Our results provide a cellular basis for sensory neve predominant injury in the peripheral nerves of Minamata disease patients.


Subject(s)
ATP-Binding Cassette Transporters , Cell Survival , Ganglia, Spinal , Methylmercury Compounds , Schwann Cells , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Methylmercury Compounds/toxicity , Schwann Cells/drug effects , Schwann Cells/metabolism , Cell Survival/drug effects , Cells, Cultured , Large Neutral Amino Acid-Transporter 1/metabolism , Large Neutral Amino Acid-Transporter 1/genetics , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Peripheral Nerves/metabolism , Peripheral Nerves/drug effects , Male , Rats , Multidrug Resistance-Associated Protein 2
6.
J Clin Invest ; 134(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38690736

ABSTRACT

Pain and inflammation are biologically intertwined responses that warn the body of potential danger. In this issue of the JCI, Defaye, Bradaia, and colleagues identified a functional link between inflammation and pain, demonstrating that inflammation-induced activation of stimulator of IFN genes (STING) in dorsal root ganglia nociceptors reduced pain-like behaviors in a rodent model of inflammatory pain. Utilizing mice with a gain-of-function STING mutation, Defaye, Bradaia, and colleagues identified type I IFN regulation of voltage-gated potassium channels as the mechanism of this pain relief. Further investigation into mechanisms by which proinflammatory pathways can reduce pain may reveal druggable targets and insights into new approaches for treating persistent pain.


Subject(s)
Ganglia, Spinal , Membrane Proteins , Pain , Animals , Mice , Ganglia, Spinal/metabolism , Pain/genetics , Pain/metabolism , Pain/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Humans , Nociceptors/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/immunology , Interferon Type I/metabolism , Interferon Type I/genetics , Interferon Type I/immunology
7.
J Clin Invest ; 134(9)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38690737

ABSTRACT

Inflammation and pain are intertwined responses to injury, infection, or chronic diseases. While acute inflammation is essential in determining pain resolution and opioid analgesia, maladaptive processes occurring during resolution can lead to the transition to chronic pain. Here we found that inflammation activates the cytosolic DNA-sensing protein stimulator of IFN genes (STING) in dorsal root ganglion nociceptors. Neuronal activation of STING promotes signaling through TANK-binding kinase 1 (TBK1) and triggers an IFN-ß response that mediates pain resolution. Notably, we found that mice expressing a nociceptor-specific gain-of-function mutation in STING exhibited an IFN gene signature that reduced nociceptor excitability and inflammatory hyperalgesia through a KChIP1-Kv4.3 regulation. Our findings reveal a role of IFN-regulated genes and KChIP1 downstream of STING in the resolution of inflammatory pain.


Subject(s)
Membrane Proteins , Nociceptors , Animals , Mice , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nociceptors/metabolism , Ganglia, Spinal/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Inflammation/genetics , Inflammation/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pain/metabolism , Pain/genetics , Signal Transduction , Male
8.
Mol Pain ; 20: 17448069241249455, 2024.
Article in English | MEDLINE | ID: mdl-38597175

ABSTRACT

Although the molecular mechanisms of chronic pain have been extensively studied, a global picture of alternatively spliced genes and events in the peripheral and central nervous systems of chronic pain is poorly understood. The current study analyzed the changing pattern of alternative splicing (AS) in mouse brain, dorsal root ganglion, and spinal cord tissue under inflammatory and neuropathic pain. In total, we identified 6495 differentially alternatively spliced (DAS) genes. The molecular functions of shared DAS genes between these two models are mainly enriched in calcium signaling pathways, synapse organization, axon regeneration, and neurodegeneration disease. Additionally, we identified 509 DAS in differentially expressed genes (DEGs) shared by these two models, accounting for a small proportion of total DEGs. Our findings supported the hypothesis that the AS has an independent regulation pattern different from transcriptional regulation. Taken together, these findings indicate that AS is one of the important molecular mechanisms of chronic pain in mammals. This study presents a global description of AS profile changes in the full path of neuropathic and inflammatory pain models, providing new insights into the underlying mechanisms of chronic pain and guiding genomic clinical diagnosis methods and rational medication.


Subject(s)
Alternative Splicing , Gene Expression Profiling , Inflammation , Mice, Inbred C57BL , Neuralgia , Transcriptome , Animals , Neuralgia/genetics , Neuralgia/metabolism , Alternative Splicing/genetics , Inflammation/genetics , Transcriptome/genetics , Male , Ganglia, Spinal/metabolism , Mice , Spinal Cord/metabolism , Spinal Cord/pathology , Gene Expression Regulation , Disease Models, Animal
9.
Int Immunopharmacol ; 133: 112113, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38657498

ABSTRACT

BACKGROUND: Phytosphingosine and its derivative are known for their skin-protective properties. While mYG-II-6, a phytosphingosine derivative, has shown anti-inflammatory and antipsoriatic effects, its potential antipruritic qualities have yet to be explored. This study aimed to investigate mYG-II-6's antipruritic properties. METHODS: The calcium imaging technique was employed to investigate the activity of ion channels and receptors. Mast cell degranulation was confirmed through the ß-hexosaminidase assay. Additionally, in silico molecular docking and an in vivo mouse scratching behavior test were utilized. RESULTS: Using HEK293T cells transfected with H1R and TRPV1, we examined the impact of mYG-II-6 on histamine-induced intracellular calcium rise, a key signal in itch-mediating sensory neurons. Pretreatment with mYG-II-6 significantly reduced histamine-induced calcium levels and inhibited TRPV1 activity, suggesting its role in blocking the calcium influx channel. Additionally, mYG-II-6 suppressed histamine-induced calcium increase in primary cultures of mouse dorsal root ganglia, indicating its potential antipruritic effect mediated by histamine. Interestingly, mYG-II-6 exhibited inhibitory effects on human MRGPRX2, a G protein-coupled receptor involved in IgE-independent mast cell degranulation. However, it did not inhibit mouse MrgprB2, the ortholog of human MRGPRX2. Molecular docking analysis revealed that mYG-II-6 selectively interacts with the binding pocket of MRGPRX2. Importantly, mYG-II-6 suppressed histamine-induced scratching behaviors in mice. CONCLUSIONS: Our findings show that mYG-II-6 can alleviate histamine-induced itch sensation through dual mechanisms. This underscores its potential as a versatile treatment for various pruritic conditions.


Subject(s)
Cell Degranulation , Histamine , Mast Cells , Molecular Docking Simulation , Receptors, G-Protein-Coupled , TRPV Cation Channels , Animals , Mast Cells/drug effects , Mast Cells/immunology , Humans , TRPV Cation Channels/metabolism , Cell Degranulation/drug effects , HEK293 Cells , Histamine/metabolism , Receptors, G-Protein-Coupled/metabolism , Mice , Male , Pruritus/drug therapy , Calcium/metabolism , Antipruritics/pharmacology , Antipruritics/therapeutic use , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Neuropeptide/metabolism , Mice, Inbred C57BL
10.
Brain Res ; 1834: 148915, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38582414

ABSTRACT

Bestrophin-1 and anoctamin-1 are members of the calcium-activated chloride channels (CaCCs) family and are involved in inflammatory and neuropathic pain. However, their role in pain hypersensitivity induced by REM sleep deprivation (REMSD) has not been studied. This study aimed to determine if anoctamin-1 and bestrophin-1 are involved in the pain hypersensitivity induced by REMSD. We used the multiple-platform method to induce REMSD. REM sleep deprivation for 48 h induced tactile allodynia and a transient increase in corticosterone concentration at the beginning of the protocol (12 h) in female and male rats. REMSD enhanced c-Fos and α2δ-1 protein expression but did not change activating transcription factor 3 (ATF3) and KCC2 expression in dorsal root ganglia and dorsal spinal cord. Intrathecal injection of CaCCinh-A01, a non-selective bestrophin-1 blocker, and T16Ainh-A01, a specific anoctamin-1 blocker, reverted REMSD-induced tactile allodynia. However, T16Ainh-A01 had a higher antiallodynic effect in male than female rats. In addition, REMSD increased bestrophin-1 protein expression in DRG but not in DSC in male and female rats. In marked contrast, REMSD decreased anoctamin-1 protein expression in DSC but not in DRG, only in female rats. Bestrophin-1 and anoctamin-1 promote pain and maintain tactile allodynia induced by REM sleep deprivation in both male and female rats, but their expression patterns differ between the sexes.


Subject(s)
Bestrophins , Ganglia, Spinal , Hyperalgesia , Sleep Deprivation , Spinal Cord , Animals , Sleep Deprivation/metabolism , Sleep Deprivation/complications , Hyperalgesia/metabolism , Male , Female , Rats , Ganglia, Spinal/metabolism , Spinal Cord/metabolism , Bestrophins/metabolism , Chloride Channels/metabolism , Sleep, REM/physiology , Rats, Wistar , Anoctamin-1 , Calcium Channels, L-Type
11.
Toxins (Basel) ; 16(4)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38668612

ABSTRACT

Accidents caused by Bothrops jararaca (Bj) snakes result in several local and systemic manifestations, with pain being a fundamental characteristic. The inflammatory process responsible for hyperalgesia induced by Bj venom (Bjv) has been studied; however, the specific roles played by the peripheral and central nervous systems in this phenomenon remain unclear. To clarify this, we induced hyperalgesia in rats using Bjv and collected tissues from dorsal root ganglia (DRGs) and spinal cord (SC) at 2 and 4 h post-induction. Samples were labeled for Iba-1 (macrophage and microglia), GFAP (satellite cells and astrocytes), EGR1 (neurons), and NK1 receptors. Additionally, we investigated the impact of minocycline, an inhibitor of microglia, and GR82334 antagonist on Bjv-induced hyperalgesia. Our findings reveal an increase in Iba1 in DRG at 2 h and EGR1 at 4 h. In the SC, markers for microglia, astrocytes, neurons, and NK1 receptors exhibited increased expression after 2 h, with EGR1 continuing to rise at 4 h. Minocycline and GR82334 inhibited venom-induced hyperalgesia, highlighting the crucial roles of microglia and NK1 receptors in this phenomenon. Our results suggest that the hyperalgesic effects of Bjv involve the participation of microglial and astrocytic cells, in addition to the activation of NK1 receptors.


Subject(s)
Bothrops , Crotalid Venoms , Ganglia, Spinal , Hyperalgesia , Receptors, Neurokinin-1 , Animals , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Crotalid Venoms/toxicity , Male , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Receptors, Neurokinin-1/metabolism , Minocycline/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/genetics , Microglia/drug effects , Microglia/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Rats , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Microfilament Proteins/metabolism , Neurokinin-1 Receptor Antagonists/pharmacology , Rats, Sprague-Dawley
12.
EMBO Rep ; 25(5): 2375-2390, 2024 May.
Article in English | MEDLINE | ID: mdl-38594391

ABSTRACT

Cancer patients undergoing treatment with antineoplastic drugs often experience chemotherapy-induced neuropathic pain (CINP), and the therapeutic options for managing CINP are limited. Here, we show that systemic paclitaxel administration upregulates the expression of neurotrophin-3 (Nt3) mRNA and NT3 protein in the neurons of dorsal root ganglia (DRG), but not in the spinal cord. Blocking NT3 upregulation attenuates paclitaxel-induced mechanical, heat, and cold nociceptive hypersensitivities and spontaneous pain without altering acute pain and locomotor activity in male and female mice. Conversely, mimicking this increase produces enhanced responses to mechanical, heat, and cold stimuli and spontaneous pain in naive male and female mice. Mechanistically, NT3 triggers tropomyosin receptor kinase C (TrkC) activation and participates in the paclitaxel-induced increases of C-C chemokine ligand 2 (Ccl2) mRNA and CCL2 protein in the DRG. Given that CCL2 is an endogenous initiator of CINP and that Nt3 mRNA co-expresses with TrkC and Ccl2 mRNAs in DRG neurons, NT3 likely contributes to CINP through TrkC-mediated activation of the Ccl2 gene in DRG neurons. NT3 may be thus a potential target for CINP treatment.


Subject(s)
Chemokine CCL2 , Ganglia, Spinal , Neuralgia , Neurons , Neurotrophin 3 , Paclitaxel , Receptor, trkC , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Neuralgia/chemically induced , Neuralgia/metabolism , Neuralgia/genetics , Paclitaxel/adverse effects , Paclitaxel/pharmacology , Neurotrophin 3/metabolism , Neurotrophin 3/genetics , Male , Mice , Neurons/metabolism , Neurons/drug effects , Female , Receptor, trkC/metabolism , Receptor, trkC/genetics , Antineoplastic Agents/adverse effects , RNA, Messenger/metabolism , RNA, Messenger/genetics
13.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673862

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most prevalent and dose-limiting complications in chemotherapy patients. One identified mechanism underlying CIPN is neuroinflammation. Most of this research has been conducted in only male or female rodent models, making direct comparisons regarding the role of sex differences in the neuroimmune underpinnings of CIPN limited. Moreover, most measurements have focused on the dorsal root ganglia (DRG) and/or spinal cord, while relatively few studies have been aimed at characterizing neuroinflammation in the brain, for example the periaqueductal grey (PAG). The overall goals of the present study were to determine (1) paclitaxel-associated changes in markers of inflammation in the PAG and DRG in male and female C57Bl6 mice and (2) determine the effect of prophylactic administration of an anti-inflammatory cannabinoid, cannabigerol (CBG). In Experiment 1, male and female mice were treated with paclitaxel (8-32 mg/kg/injection, Days 1, 3, 5, and 7) and mechanical sensitivity was measured using Von Frey filaments on Day 7 (Cohort 1) and Day 14 (Cohort 2). Cohorts were euthanized on Day 8 or 15, respectively, and DRG and PAG were harvested for qPCR analysis of the gene expression of markers of pain and inflammation Aig1, Gfap, Ccl2, Cxcl9, Tlr4, Il6, and Calca. In Experiment 2, male and female mice were treated with vehicle or 10 mg/kg CBG i.p. 30 min prior to each paclitaxel injection. Mechanical sensitivity was measured on Day 14. Mice were euthanized on Day 15, and PAG were harvested for qPCR analysis of the gene expression of Aig1, Gfap, Ccl2, Cxcl9, Tlr4, Il6, and Calca. Paclitaxel produced a transient increase in potency to produce mechanical sensitivity in male versus female mice. Regarding neuroinflammation, more gene expression changes were apparent earlier in the DRG and at a later time point in the PAG. Also, more changes were observed in females in the PAG than males. Overall, sex differences were observed for most markers at both time points and regions. Importantly, in both the DRG and PAG, most increases in markers of neuroinflammation and pain occurred at paclitaxel doses higher than those associated with significant changes in the mechanical threshold. Two analytes that demonstrated the most compelling sexual dimorphism and that changed more in males were Cxcl9 and Ccl2, and Tlr4 in females. Lastly, prophylactic administration of CBG protected the male and female mice from increased mechanical sensitivity and female mice from neuroinflammation in the PAG. Future studies are warranted to explore how these sex differences may shed light on the mechanisms of CIPN and how non-psychoactive cannabinoids such as CBG may engage these targets to prevent or attenuate the effects of paclitaxel and other chemotherapeutic agents on the nervous system.


Subject(s)
Mice, Inbred C57BL , Paclitaxel , Animals , Paclitaxel/adverse effects , Female , Male , Mice , Cannabinoids/pharmacology , Cannabinoids/administration & dosage , Neuroinflammatory Diseases/drug therapy , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Sex Factors , Hyperalgesia/drug therapy , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Sex Characteristics , Inflammation/drug therapy , Inflammation/metabolism
14.
Cell Rep ; 43(4): 114014, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38568807

ABSTRACT

The transmembrane channel-like (TMC) protein family comprises eight members, with TMC1 and TMC2 being extensively studied. This study demonstrates substantial co-expression of TMC7 with the mechanosensitive channel Piezo2 in somatosensory neurons. Genetic deletion of TMC7 in primary sensory ganglia neurons in vivo enhances sensitivity in both physiological and pathological mechanosensory transduction. This deletion leads to an increase in proportion of rapidly adapting (RA) currents conducted by Piezo2 in dorsal root ganglion (DRG) neurons and accelerates RA deactivation kinetics. In HEK293 cells expressing both proteins, TMC7 significantly suppresses the current amplitudes of co-expressed Piezo2. Our findings reveal that TMC7 and Piezo2 exhibit physical interactions, and both proteins also physically interact with cytoskeletal ß-actin. We hypothesize that TMC7 functions as an inhibitory modulator of Piezo2 in DRG neurons, either through direct inhibition or by disrupting the transmission of mechanical forces from the cytoskeleton to the channel.


Subject(s)
Ganglia, Spinal , Ion Channels , Mechanotransduction, Cellular , Sensory Receptor Cells , Humans , Sensory Receptor Cells/metabolism , Animals , Ion Channels/metabolism , Ion Channels/genetics , Ganglia, Spinal/metabolism , HEK293 Cells , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Actins/metabolism
15.
Cell Commun Signal ; 22(1): 240, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664711

ABSTRACT

BACKGROUND: The repair of peripheral nerve injury poses a clinical challenge, necessitating further investigation into novel therapeutic approaches. In recent years, bone marrow mesenchymal stromal cell (MSC)-derived mitochondrial transfer has emerged as a promising therapy for cellular injury, with reported applications in central nerve injury. However, its potential therapeutic effect on peripheral nerve injury remains unclear. METHODS: We established a mouse sciatic nerve crush injury model. Mitochondria extracted from MSCs were intraneurally injected into the injured sciatic nerves. Axonal regeneration was observed through whole-mount nerve imaging. The dorsal root ganglions (DRGs) corresponding to the injured nerve were harvested to test the gene expression, reactive oxygen species (ROS) levels, as well as the degree and location of DNA double strand breaks (DSBs). RESULTS: The in vivo experiments showed that the mitochondrial injection therapy effectively promoted axon regeneration in injured sciatic nerves. Four days after injection of fluorescently labeled mitochondria into the injured nerves, fluorescently labeled mitochondria were detected in the corresponding DRGs. RNA-seq and qPCR results showed that the mitochondrial injection therapy enhanced the expression of Atf3 and other regeneration-associated genes in DRG neurons. Knocking down of Atf3 in DRGs by siRNA could diminish the therapeutic effect of mitochondrial injection. Subsequent experiments showed that mitochondrial injection therapy could increase the levels of ROS and DSBs in injury-associated DRG neurons, with this increase being correlated with Atf3 expression. ChIP and Co-IP experiments revealed an elevation of DSB levels within the transcription initiation region of the Atf3 gene following mitochondrial injection therapy, while also demonstrating a spatial proximity between mitochondria-induced DSBs and CTCF binding sites. CONCLUSION: These findings suggest that MSC-derived mitochondria injected into the injured nerves can be retrogradely transferred to DRG neuron somas via axoplasmic transport, and increase the DSBs at the transcription initiation regions of the Atf3 gene through ROS accumulation, which rapidly release the CTCF-mediated topological constraints on chromatin interactions. This process may enhance spatial interactions between the Atf3 promoter and enhancer, ultimately promoting Atf3 expression. The up-regulation of Atf3 induced by mitochondria further promotes the expression of downstream regeneration-associated genes and facilitates axon regeneration.


Subject(s)
Activating Transcription Factor 3 , Axons , DNA Breaks, Double-Stranded , Ganglia, Spinal , Mesenchymal Stem Cells , Mitochondria , Nerve Regeneration , Reactive Oxygen Species , Sciatic Nerve , Up-Regulation , Animals , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Reactive Oxygen Species/metabolism , Axons/metabolism , Nerve Regeneration/genetics , Up-Regulation/genetics , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Ganglia, Spinal/metabolism , Mice, Inbred C57BL , Male
16.
Cell Rep ; 43(4): 114058, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38614084

ABSTRACT

Proteasomes are critical for peripheral nervous system (PNS) function. Here, we investigate mammalian PNS proteasomes and reveal the presence of the neuronal membrane proteasome (NMP). We show that specific inhibition of the NMP on distal nerve fibers innervating the mouse hind paw leads to reduction in mechanical and pain sensitivity. Through investigating PNS NMPs, we demonstrate their presence on the somata and proximal and distal axons of a subset of dorsal root ganglion (DRG) neurons. Single-cell RNA sequencing experiments reveal that the NMP-expressing DRGs are primarily MrgprA3+ and Cysltr2+. NMP inhibition in DRG cultures leads to cell-autonomous and non-cell-autonomous changes in Ca2+ signaling induced by KCl depolarization, αß-meATP, or the pruritogen histamine. Taken together, these data support a model whereby NMPs are expressed on a subset of somatosensory DRGs to modulate signaling between neurons of distinct sensory modalities and indicate the NMP as a potential target for controlling pain.


Subject(s)
Ganglia, Spinal , Proteasome Endopeptidase Complex , Sensory Receptor Cells , Animals , Sensory Receptor Cells/metabolism , Proteasome Endopeptidase Complex/metabolism , Ganglia, Spinal/metabolism , Mice , Mice, Inbred C57BL , Nociception , Male , Cell Membrane/metabolism , Calcium Signaling
17.
Neuromolecular Med ; 26(1): 12, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600344

ABSTRACT

The role of circular RNAs (circRNAs) in neuropathic pain is linked to the fundamental physiological mechanisms involved. However, the exact function of circRNAs in the context of neuropathic pain is still not fully understood. The functional impact of circGRIN2B on the excitability of dorsal root ganglion (DRG) neurons was investigated using siRNA or overexpression technology in conjunction with fluorescence in situ hybridization and whole-cell patch-clamp technology. The therapeutic efficacy of circGRIN2B in treating neuropathic pain was confirmed by assessing the pain threshold in a chronic constrictive injury (CCI) model. The interaction between circGRIN2B and NF-κB was examined through RNA pulldown, RIP, and mass spectrometry assays. CircGRIN2B knockdown significantly affected the action potential discharge frequency and the sodium-dependent potassium current flux (SLICK) in DRG neurons. Furthermore, knockdown of circGRIN2B dramatically reduced the SLICK channel protein and mRNA expression in vivo and in vitro. Our research confirmed the interaction between circGRIN2B and NF-κB. These findings demonstrated that circGRIN2B promotes the transcription of the SLICK gene by binding to NF-κB. In CCI rat models, the overexpression of circGRIN2B has been shown to hinder the progression of neuropathic pain, particularly by reducing mechanical and thermal hyperalgesia. Additionally, this upregulation significantly diminished the levels of the inflammatory cytokines IL-1ß, IL-6, and TNF-α in the DRG. Upon reviewing these findings, it was determined that circGRIN2B may mitigate the onset of neuropathic pain by modulating the NF-κB/SLICK pathway.


Subject(s)
NF-kappa B , Neuralgia , Rats , Animals , NF-kappa B/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Circular/therapeutic use , Rats, Sprague-Dawley , In Situ Hybridization, Fluorescence , Neuralgia/therapy , Neuralgia/drug therapy , Hyperalgesia/drug therapy , Ganglia, Spinal/metabolism
18.
J Ethnopharmacol ; 330: 118218, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38677570

ABSTRACT

ETHNOPHARMACOLOGY RELEVANCE: Zanthoxylum bungeanum Maxim. (Z. bungeanum), a member of the Rutaceae family, has a rich history of traditional use in Asia for treating arthritis and toothache conditions. As characteristic chemical components, numerous kinds of alkaloids have been extracted from plants and their diverse biological activities have been reported. However, research on the isoquinoline alkaloid, a specific type of alkaloids, in Z. bungeanum was scarce. AIM OF THE STUDY: The study aimed to isolate a novel isoquinoline alkaloid from Z. bungeanum and explore its pharmacological activity in vitro and analgesic activity in vivo. MATERIALS AND METHODS: Isoquinoline alkaloid isolation and identification from Z. bungeanum were conducted using chromatographic and spectroscopic methods. The whole-cell patch-clamp technique was applied to assess its impact on neuronal excitability, and endogenous voltage-gated potassium (Kv) and sodium (Nav) currents in acutely isolated mouse small-diameter dorsal root ganglion (DRG) neurons. Its inhibitory impacts on channels were further validated with HEK293 cells stably expressing Nav1.7 and Nav1.8, and Chinese hamster ovary (CHO) cells transiently expressing Kv2.1. The formalin inflammatory pain model was utilized to evaluate the potential analgesic activity in vivo. RESULTS: A novel isoquinoline alkaloid named HJ-69 (N-13-(3-methoxyprop-1-yl)rutaecarpine) was isolated and identified from Z. bungeanum for the first time. HJ-69 significantly suppressed the firing frequency and amplitudes of action potentials in DRG neurons. Consistently, it state-dependently inhibited endogenous Nav currents of DRG neurons, with half maximal inhibitory concentration (IC50) values of 13.06 ± 2.06 µM and 30.19 ± 2.07 µM for the inactivated and resting states, respectively. HJ-69 significantly suppressed potassium currents in DRG neurons, which notably inhibited the delayed rectifier potassium (IK) currents (IC50 = 6.95 ± 1.29 µM) and slightly affected the transient outward potassium (IA) currents (IC50 = 523.50 ± 39.16 µM). Furtherly, HJ-69 exhibited similar potencies on heterologously expressed Nav1.7, Nav1.8, and Kv2.1 channels, which correspondingly represent the main components in neurons. Notably, intraperitoneal administration of 30 mg/kg and 100 mg/kg HJ-69 significantly alleviated pain behaviors in the mouse inflammatory pain model induced by formalin. CONCLUSION: The study concluded that HJ-69 is a novel and active isoquinoline alkaloid, and the inhibition of Nav and Kv channels contributes to its analgesic activity. HJ-69 may be a promising prototype for future analgesic drug discovery based on the isoquinoline alkaloid.


Subject(s)
Analgesics , Ganglia, Spinal , Pain , Zanthoxylum , Animals , Zanthoxylum/chemistry , Humans , HEK293 Cells , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/isolation & purification , Analgesics/therapeutic use , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Mice , Male , Pain/drug therapy , Isoquinolines/pharmacology , Isoquinolines/isolation & purification , Isoquinolines/chemistry , Alkaloids/pharmacology , Alkaloids/isolation & purification , Alkaloids/chemistry , Alkaloids/therapeutic use , Potassium Channel Blockers/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Inflammation/drug therapy , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/isolation & purification , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/drug effects , Neurons/drug effects , Neurons/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/chemistry , Mice, Inbred C57BL , Cricetulus
19.
Exp Neurol ; 376: 114750, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492636

ABSTRACT

Nerve injury often leads to severe dysfunction because of the lack of axon regeneration in adult mammal. Intriguingly a series of extracellular vesicles (EVs) have the obvious ability to accelerate the nerve repair. However, the detailed molecular mechanisms to describe that EVs switch neuron from a transmitter to a regenerative state have not been elucidated. This study elucidated the microRNA (miRNA) expression profiles of two types of EVs that promote nerve regeneration. The functions of these miRNAs were screened in vitro. Among the 12 overlapping miRNAs, miR-25-3p was selected for further analysis as it markedly promoted axon regeneration both in vivo and in vitro. Furthermore, knockdown experiments confirmed that PTEN and Klf4, which are the major inhibitors of axon regeneration, were the direct targets of miR-25-3p in dorsal root ganglion (DRG) neurons. The utilization of luciferase reporter assays and functional tests provided evidence that miR-25-3p enhances axon regeneration by targeting Tgif1. Additionally, miR-25-3p upregulated the phosphorylation of Erk. Furthermore, Rapamycin modulated the expression of miR-25-3p in DRG neurons. Finally, the pro-axon regeneration effects of EVs were confirmed by overexpressing miR-25-3p and Tgif1 knockdown in the optic nerve crush model. Thus, the enrichment of miR-25-3p in EVs suggests that it regulates axon regeneration, proving a potential cell-free treatment strategy for nerve injury.


Subject(s)
Axons , Extracellular Vesicles , Ganglia, Spinal , Homeodomain Proteins , MicroRNAs , Nerve Regeneration , Schwann Cells , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Regeneration/physiology , Nerve Regeneration/genetics , Extracellular Vesicles/metabolism , Axons/physiology , Schwann Cells/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Skin/metabolism , Kruppel-Like Factor 4 , Mice, Inbred C57BL , Stem Cells/metabolism
20.
ACS Chem Neurosci ; 15(6): 1063-1073, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38449097

ABSTRACT

Chronic pain is a growing global health problem affecting at least 10% of the world's population. However, current chronic pain treatments are inadequate. Voltage-gated sodium channels (Navs) play a pivotal role in regulating neuronal excitability and pain signal transmission and thus are main targets for nonopioid painkiller development, especially those preferentially expressed in dorsal root ganglial (DRG) neurons, such as Nav1.6, Nav1.7, and Nav1.8. In this study, we screened in virtual hits from dihydrobenzofuran and 3-hydroxyoxindole hybrid molecules against Navs via a veratridine (VTD)-based calcium imaging method. The results showed that one of the molecules, 3g, could inhibit VTD-induced neuronal activity significantly. Voltage clamp recordings demonstrated that 3g inhibited the total Na+ currents of DRG neurons in a concentration-dependent manner. Biophysical analysis revealed that 3g slowed the activation, meanwhile enhancing the inactivation of the Navs. Additionally, 3g use-dependently blocked Na+ currents. By combining with selective Nav inhibitors and a heterozygous expression system, we demonstrated that 3g preferentially inhibited the TTX-S Na+ currents, specifically the Nav1.7 current, other than the TTX-R Na+ currents. Molecular docking experiments implicated that 3g binds to a known allosteric site at the voltage-sensing domain IV(VSDIV) of Nav1.7. Finally, intrathecal injection of 3g significantly relieved mechanical pain behavior in the spared nerve injury (SNI) rat model, suggesting that 3g is a promising candidate for treating chronic pain.


Subject(s)
Chronic Pain , Indoles , Neuralgia , Rats , Animals , Molecular Docking Simulation , NAV1.8 Voltage-Gated Sodium Channel , Neuralgia/drug therapy , Neuralgia/metabolism , Ganglia, Spinal/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...