Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.349
Filter
1.
Epigenetics ; 19(1): 2380930, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39066680

ABSTRACT

In mammals, the molecular mechanisms underlying transgenerational inheritance of phenotypic traits in serial generations of progeny after ancestral environmental exposures, without variation in DNA sequence, remain elusive. We've recently described transmission of a beneficial trait in rats and mice, in which F0 supplementation of methyl donors, including folic acid, generates enhanced axon regeneration after sharp spinal cord injury in untreated F1 to F3 progeny linked to differential DNA methylation levels in spinal cord tissue. To test whether the transgenerational effect of folic acid is transmitted via the germline, we performed whole-genome methylation sequencing on sperm DNA from F0 mice treated with either folic acid or vehicle control, and their F1, F2, and F3 untreated progeny. Transgenerational differentially methylated regions (DMRs) are observed in each consecutive generation and distinguish folic acid from untreated lineages, predominate outside of CpG islands and in regions of the genome that regulate gene expression, including promoters, and overlap at both the differentially methylated position (DMP) and gene levels. These findings indicate that molecular changes between generations are caused by ancestral folate supplementation. In addition, 29,719 DMPs exhibit serial increases or decreases in DNA methylation levels in successive generations of untreated offspring, correlating with a serial increase in the phenotype across generations, consistent with a 'wash-in' effect. Sibship-specific DMPs annotate to genes that participate in axon- and synapse-related pathways.


Subject(s)
Axons , DNA Methylation , Folic Acid , Spermatozoa , Folic Acid/pharmacology , Folic Acid/administration & dosage , Animals , Male , Mice , Spermatozoa/drug effects , Spermatozoa/metabolism , Axons/metabolism , Axons/drug effects , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , CpG Islands , Female , Nerve Regeneration/drug effects , Epigenesis, Genetic , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/cytology
2.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063088

ABSTRACT

This study investigated the effects of cilostazol on motor dysfunction, spinal motor neuron abnormalities, and schwannopathy in rats with diabetes. Diabetes mellitus (DM) was induced in rats via femoral intravenous streptozotocin (STZ) injection (60 mg/kg). After successful DM induction, cilostazol was administered on day 15 via oral gavage (100 mg/kg/day) for 6 weeks until sacrifice. Behavioral assays, including motor function, were performed weekly. The sciatic nerve, L5 spinal cord, and spinal ventral root were collected to evaluate the expression of the glial fibrillary acidic protein (GFAP), myelin protein zero (P0), and choline acetyltransferase (ChAT) by immunofluorescence and Western blotting. DM rats displayed decreased running speeds, running distances, and toe spread but increased foot pressure. In addition, loss of non-myelinating Schwann cells and myelin sheaths was observed in the sciatic nerve and L5 spinal ventral root. Reduced numbers of motor neurons were also found in the L5 spinal ventral horn. Cilostazol administration significantly potentiated running speed and distance; increased hind paw toe spread; and decreased foot pressure. In the sciatic nerve and L5 spinal ventral root, cilostazol treatment significantly improved non-myelinated Schwann cells and increased myelin mass. ChAT expression in motor neurons in the spinal ventral horn was improved, but not significantly. Cilostazol administration may protect sensorimotor function in diabetic rats.


Subject(s)
Cilostazol , Diabetes Mellitus, Experimental , Schwann Cells , Sciatic Nerve , Animals , Cilostazol/pharmacology , Cilostazol/therapeutic use , Schwann Cells/drug effects , Schwann Cells/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Rats , Male , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Choline O-Acetyltransferase/metabolism , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism , Motor Neurons/drug effects , Motor Neurons/metabolism , Glial Fibrillary Acidic Protein/metabolism , Myelin P0 Protein/metabolism , Streptozocin
3.
Biomed Res ; 45(4): 151-161, 2024.
Article in English | MEDLINE | ID: mdl-39010191

ABSTRACT

Linalool and linalyl acetate are major components of lavender essential oil. These substances possess many biological activities, such as anti-inflammatory activity, analgesic and anxiolytic effects, and anticonvulsant properties, and they also induce modulation of neuronal activity in the autonomic nervous system. However, there are no reports of the direct effects of linalool on respiratory activity. In the present study, we analyzed the effects of linalool and linalyl acetate on central respiratory activity in the brainstem-spinal cord preparation isolated from newborn rats. Linalool dose-dependently decreased the rate of respiratory activity. This effect was reversed by bicuculline, suggesting that linalool enhanced inhibitory synaptic connections via GABAA receptors. In addition, linalool reduced the coefficient of variation of inspiratory burst intervals and thus could work to stabilize the respiratory rhythm. Linalyl acetate did not cause inhibitory effects as observed in linalool treatment. Linalool depressed burst activity of pre-inspiratory neurons in the medullary respiratory networks and increased the amplitude of inspiratory inhibitory postsynaptic potentials of pre-inspiratory neurons. We concluded that linalool caused inhibitory effects on respiratory rhythm generation mainly through activation of presynaptic GABAA receptors of pre-inspiratory neurons.


Subject(s)
Acyclic Monoterpenes , Animals, Newborn , Brain Stem , Monoterpenes , Neurons , Spinal Cord , Animals , Acyclic Monoterpenes/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Rats , Monoterpenes/pharmacology , Neurons/drug effects , Neurons/metabolism , Brain Stem/drug effects , Brain Stem/physiology , Receptors, GABA-A/metabolism , Respiration/drug effects , Bicuculline/pharmacology
4.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000569

ABSTRACT

Regulation of neuroinflammation is critical for maintaining central nervous system (CNS) homeostasis and holds therapeutic promise in autoimmune diseases such as multiple sclerosis (MS). Previous studies have highlighted the significance of selective innate signaling in triggering anti-inflammatory mechanisms, which play a protective role in an MS-like disease, experimental autoimmune encephalomyelitis (EAE). However, the individual intra-CNS administration of specific innate receptor ligands or agonists, such as for toll-like receptor 7 (TLR7) and nucleotide-binding oligomerization-domain-containing protein 2 (NOD2), failed to elicit the desired anti-inflammatory response in EAE. In this study, we investigated the potential synergistic effect of targeting both TLR7 and NOD2 simultaneously to prevent EAE progression. Our findings demonstrate that simultaneous intrathecal administration of NOD2- and TLR7-agonists led to synergistic induction of Type I IFN (IFN I) and effectively suppressed EAE in an IFN I-dependent manner. Suppression of EAE was correlated with a significant decrease in the infiltration of monocytes, granulocytes, and natural killer cells, reduced demyelination, and downregulation of IL-1ß, CCL2, and IFNγ gene expression in the spinal cord. These results underscore the therapeutic promise of concurrently targeting the TLR7 and NOD2 pathways in alleviating neuroinflammation associated with MS, paving the way for novel and more efficacious treatment strategies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Nod2 Signaling Adaptor Protein , Toll-Like Receptor 7 , Animals , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/agonists , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Mice , Mice, Inbred C57BL , Immunity, Innate/drug effects , Female , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/drug effects , Membrane Glycoproteins/metabolism , Interferon Type I/metabolism , Signal Transduction/drug effects
5.
CNS Neurosci Ther ; 30(7): e14829, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961264

ABSTRACT

AIMS: Paclitaxel (PTX) is extensively utilized in the management of diverse solid tumors, frequently resulting in paclitaxel-induced peripheral neuropathy (PIPN). The present study aimed to investigate sex differences in the behavioral manifestations and underlying pathogenesis of PIPN and search for clinically efficacious interventions. METHODS: Male and female C57BL/6 mice (5-6 weeks and 12 months, weighing 18-30 g) were intraperitoneally (i.p.) administered paclitaxel diluted in saline (NaCl 0.9%) at a dose of 2 mg/kg every other day for a total of 4 injections. Von Frey and hot plate tests were performed before and after administration to confirm the successful establishment of the PIPN model and also to evaluate the pain of PIPN and the analgesic effect of PD-L1. On day 14 after PTX administration, PD-L1 protein (10 ng/pc) was injected into the PIPN via the intrathecal (i.t.) route. To knock down TRPV1 in the spinal cord, adeno-associated virus 9 (AAV9)-Trpv1-RNAi (5 µL, 1 × 1013 vg/mL) was slowly injected via the i.t. route. Four weeks after AAV9 delivery, the downregulation of TRPV1 expression was verified by immunofluorescence staining and Western blotting. The levels of PD-L1, TRPV1 and CGRP were measured via Western blotting, RT-PCR, and immunofluorescence staining. The levels of TNF-α and IL-1ß were measured via RT-PCR. RESULTS: TRPV1 and CGRP protein and mRNA levels were higher in the spinal cords of control female mice than in those of control male mice. PTX-induced nociceptive behaviors in female PIPN mice were greater than those in male PIPN mice, as indicated by increased expression of TRPV1 and CGRP. The analgesic effects of PD-L1 on mechanical hyperalgesia and thermal sensitivity were significantly greater in female mice than in male mice, with calculated relative therapeutic levels increasing by approximately 2.717-fold and 2.303-fold, respectively. PD-L1 and CGRP were partly co-localized with TRPV1 in the dorsal horn of the mouse spinal cord. The analgesic effect of PD-L1 in PIPN mice was observed to be mediated through the downregulation of TRPV1 and CGRP expression following AAV9-mediated spinal cord specific decreased TRPV1 expression. CONCLUSIONS: PTX-induced nociceptive behaviors and the analgesic effect of PD-L1 in PIPN mice were sexually dimorphic, highlighting the significance of incorporating sex as a crucial biological factor in forthcoming mechanistic studies of PIPN and providing insights for potential sex-specific therapeutic approaches.


Subject(s)
B7-H1 Antigen , Calcitonin Gene-Related Peptide , Mice, Inbred C57BL , Paclitaxel , Peripheral Nervous System Diseases , Sex Characteristics , TRPV Cation Channels , Animals , Paclitaxel/toxicity , Male , Female , Mice , Calcitonin Gene-Related Peptide/metabolism , TRPV Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , B7-H1 Antigen/metabolism , Peripheral Nervous System Diseases/chemically induced , Antineoplastic Agents, Phytogenic/toxicity , Spinal Cord/drug effects , Spinal Cord/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism
6.
Pharm Res ; 41(7): 1401-1411, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38981901

ABSTRACT

PURPOSE: Serotonin (5-HT3) receptor antagonists are promising agents for treatment of neuropathic pain. However, insufficient drug exposure at the central nervous system (CNS) might result in lack of efficacy. The goal of this study was to evaluate the impact of administration of a Pgp inhibitor (tariquidar) on ondansetron exposure in the brain, spinal cord, and cerebrospinal fluid in a wild-type rat model. METHODS: Ondansetron (10 mg/kg) and tariquidar (7.5 mg/kg) were administered intravenously, plasma and tissue samples were collected and analyzed by HPLC. A mathematical model with brain, spinal cord, cerebrospinal fluid and two systemic disposition compartments was developed to describe the data. RESULTS: The results demonstrate that tariquidar at 7.5 mg/kg resulted in a complete inhibition of Pgp efflux of ondansetron in the brain and spinal cord. The compartmental model successfully captured pharmacokinetics of ondansetron in wild type and Pgp knockout (KO) animals receiving the drug alone or in wild type animals receiving the ondansetron and tariquidar combination. CONCLUSIONS: The study provided important quantitative information on enhancement of CNS exposure to ondansetron using co-administration of Pgp Inhibitor in a rat model, which will be further utilized in conducting a clinical study. Tariquidar co-administration resulted in ondansetron CNS exposure comparable to observed in Pgp KO rats. Results also highlighted the effect of tariquidar on plasma disposition of ondansetron, which may not be dependent on Pgp inhibition, and should be evaluated in future studies.


Subject(s)
Ondansetron , Quinolines , Spinal Cord , Animals , Ondansetron/pharmacokinetics , Rats , Male , Spinal Cord/metabolism , Spinal Cord/drug effects , Quinolines/pharmacokinetics , Quinolines/administration & dosage , Rats, Sprague-Dawley , Brain/metabolism , Brain/drug effects , Models, Biological , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Central Nervous System/metabolism , Central Nervous System/drug effects , Serotonin 5-HT3 Receptor Antagonists/pharmacokinetics , Serotonin 5-HT3 Receptor Antagonists/pharmacology
8.
Exp Neurol ; 379: 114886, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996862

ABSTRACT

Spinal cord injury (SCI) induces the disruption of the blood-spinal cord barrier (BSCB) and the failure of axonal growth. SCI activates a complex series of responses, including cell apoptosis and endoplasmic reticulum (ER) stress. Pericytes play a critical role in maintaining BSCB integrity and facilitating tissue growth and repair. However, the roles of pericytes in SCI and the potential mechanisms underlying the improvements in functional recovery in SCI remain unclear. Recent evidence indicates that irisflorentin exerts neuroprotective effects against Parkinson's disease; however, whether it has potential protective roles in SCI or not is still unknown. In this study, we found that the administration of irisflorentin significantly inhibited pericyte apoptosis, protected BSCB integrity, promoted axonal growth, and ultimately improved locomotion recovery in a rat model of SCI. In vitro, we found that the positive effects of irisflorentin on axonal growth were likely to be mediated by regulating the crosstalk between pericytes and neurons. Furthermore, irisflorentin effectively ameliorated ER stress caused by incubation with thapsigargin (TG) in pericytes. Meanwhile, the protective effect of irisflorentin on BSCB disruption is strongly related to the reduction of pericyte apoptosis via inhibition of ER stress. Collectively, our findings demonstrate that irisflorentin is beneficial for functional recovery after SCI and that pericytes are a valid target of interest for future SCI therapies.


Subject(s)
Neuroprotective Agents , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord Injuries , Animals , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Recovery of Function/drug effects , Recovery of Function/physiology , Rats , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Axons/drug effects , Pericytes/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Female , Spinal Cord/drug effects , Apoptosis/drug effects , Cells, Cultured
9.
Aging (Albany NY) ; 16(13): 11062-11071, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38975935

ABSTRACT

OBJECTIVE: To investigate the effect of sevoflurane on neuropathic pain induced by chronic constriction injury (CCI) of sciatic nerve in mice, and to elucidate its mechanism by animal experiments. METHODS AND RESULTS: Thirty-two C57BL/6 mice were randomly divided into four groups: Sham group, Model group, Control group and Sevoflurane group. First, a mouse model of neuropathic pain was established. Then, the mice in each group were killed on Day 14 after operation to harvest the enlarged lumbosacral spinal cord. In contrast with the Model group, the Sevoflurane group displayed a significantly increased paw withdrawal mechanical threshold (PWMT) and significantly prolonged paw withdrawal thermal latency (PWTL) from Day 5 after operation. The morphological changes of lumbosacral spinal cord were observed by hematoxylin-eosin (HE) staining and transmission electron microscopy. Pathological results showed that sevoflurane reduced nuclear pyknosis in lumbosacral spinal cord tissue, with a large number of mitochondrial crista disappearance and mitochondrial swelling. The results of Western blotting showed that sevoflurane significantly decreased the protein expressions of phosphorylated phospholipase Cγ (p-PLCγ), phosphorylated calcium/calmodulin-dependent protein kinase II (p-CaMKII) and phosphorylated inositol 1,4,5-triphosphate receptor (p-IP3R), and reduced the protein expressions of endoplasmic reticulum (ER) stress proteins glucose-regulated protein 78 (GRP78) and GRP94, oxidative stress-related proteins P22 and P47 and inflammatory factors nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), interleukin-1 ß (IL-1ß), and tumor necrosis factor-α (TNF-α). CONCLUSIONS: Sevoflurane inhibits neuropathic pain by maintaining ER stress and oxidative stress homeostasis through inhibiting the activation of the PLCγ/CaMKII/IP3R signaling pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Inositol 1,4,5-Trisphosphate Receptors , Mice, Inbred C57BL , Neuralgia , Oxidative Stress , Phospholipase C gamma , Sevoflurane , Signal Transduction , Animals , Sevoflurane/pharmacology , Endoplasmic Reticulum Stress/drug effects , Neuralgia/metabolism , Neuralgia/drug therapy , Signal Transduction/drug effects , Oxidative Stress/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Mice , Phospholipase C gamma/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Homeostasis/drug effects , Disease Models, Animal , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/injuries
10.
Nat Commun ; 15(1): 6264, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048565

ABSTRACT

Opioid withdrawal is a liability of chronic opioid use and misuse, impacting people who use prescription or illicit opioids. Hyperactive autonomic output underlies many of the aversive withdrawal symptoms that make it difficult to discontinue chronic opioid use. The locus coeruleus (LC) is an important autonomic centre within the brain with a poorly defined role in opioid withdrawal. We show here that pannexin-1 (Panx1) channels expressed on microglia critically modulate LC activity during opioid withdrawal. Within the LC, we found that spinally projecting tyrosine hydroxylase (TH)-positive neurons (LCspinal) are hyperexcitable during morphine withdrawal, elevating cerebrospinal fluid (CSF) levels of norepinephrine. Pharmacological and chemogenetic silencing of LCspinal neurons or genetic ablation of Panx1 in microglia blunted CSF NE release, reduced LC neuron hyperexcitability, and concomitantly decreased opioid withdrawal behaviours in mice. Using probenecid as an initial lead compound, we designed a compound (EG-2184) with greater potency in blocking Panx1. Treatment with EG-2184 significantly reduced both the physical signs and conditioned place aversion caused by opioid withdrawal in mice, as well as suppressed cue-induced reinstatement of opioid seeking in rats. Together, these findings demonstrate that microglial Panx1 channels modulate LC noradrenergic circuitry during opioid withdrawal and reinstatement. Blocking Panx1 to dampen LC hyperexcitability may therefore provide a therapeutic strategy for alleviating the physical and aversive components of opioid withdrawal.


Subject(s)
Connexins , Locus Coeruleus , Nerve Tissue Proteins , Probenecid , Spinal Cord , Substance Withdrawal Syndrome , Animals , Locus Coeruleus/metabolism , Locus Coeruleus/drug effects , Connexins/metabolism , Connexins/genetics , Connexins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/drug therapy , Mice , Male , Rats , Spinal Cord/metabolism , Spinal Cord/drug effects , Probenecid/pharmacology , Morphine/pharmacology , Microglia/drug effects , Microglia/metabolism , Analgesics, Opioid/pharmacology , Norepinephrine/metabolism , Neurons/metabolism , Neurons/drug effects , Mice, Inbred C57BL , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism , Mice, Knockout
11.
Yi Chuan ; 46(6): 478-489, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38886151

ABSTRACT

Metronidazole (MTZ), a commonly used anti-infective drug in clinical practice, has also been employed as a prodrug in cell-targeted ablation systems in scientific research, exhibiting significant application value. However, it has been demonstrated that MTZ can induce neurotoxic symptoms to some extent during its use, and there is currently a lack of effective means to circumvent its toxicity in both clinical and research settings, which limits its application. Therefore, exploring the specific mechanisms underlying MTZ-induced neurotoxic symptoms and elucidating countermeasures will enhance the practical value of MTZ. In this study, using a zebrafish spinal cord injury regeneration model, we confirmed that MTZ neurotoxicity leads to impaired axon regeneration in the central nervous system. By overexpressing il34 in the central nervous system of zebrafish, we eliminated the inhibitory effect of MTZ on axonal regeneration and demonstrated that the pro-regenerative effect against MTZ neurotoxicity is not caused by excessive macrophages/microglia chemoattracted by interleukin 34(Il34). Transcriptome sequencing analysis and GO enrichment analysis of differentially expressed genes between groups revealed that Il34 may counteract MTZ neurotoxicity and promote spinal cord injury repair through biological processes that enhance cellular adhesion and cell location. In summary, our work uncovers a possible cause of MTZ neurotoxicity and provides a new perspective for eliminating MTZ toxicity.


Subject(s)
Metronidazole , Spinal Cord Injuries , Spinal Cord Regeneration , Zebrafish , Animals , Metronidazole/pharmacology , Metronidazole/adverse effects , Spinal Cord Regeneration/drug effects , Spinal Cord Injuries/metabolism , Interleukins/genetics , Interleukins/metabolism , Central Nervous System/drug effects , Central Nervous System/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism
12.
J Neuroimmune Pharmacol ; 19(1): 28, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38862787

ABSTRACT

Despite antiretroviral therapy (ART), HIV-associated peripheral neuropathy remains one of the most prevalent neurologic manifestations of HIV infection. The spinal cord is an essential component of sensory pathways, but spinal cord sampling and evaluation in people with HIV has been very limited, especially in those on ART. The SIV/macaque model allows for assessment of the spinal cord at key time points throughout infection with and without ART. In this study, RNA was isolated from the spinal cord of uninfected, SIV+, and SIV + ART animals to track alterations in gene expression using global RNA-seq. Next, the SeqSeek platform was used to map changes in gene expression to specific cell types. Pathway analysis of differentially expressed genes demonstrated that highly upregulated genes in SIV-infected spinal cord aligned with interferon and viral response pathways. Additionally, this upregulated gene set significantly overlapped with those expressed in myeloid-derived cells including microglia. Downregulated genes were involved in cholesterol and collagen biosynthesis, and TGF-b regulation of extracellular matrix. In contrast, enriched pathways identified in SIV + ART animals included neurotransmitter receptors and post synaptic signaling regulators, and transmission across chemical synapses. SeqSeek analysis showed that upregulated genes were primarily expressed by neurons rather than glia. These findings indicate that pathways activated in the spinal cord of SIV + ART macaques are predominantly involved in neuronal signaling rather than proinflammatory pathways. This study provides the basis for further evaluation of mechanisms of SIV infection + ART within the spinal cord with a focus on therapeutic interventions to maintain synaptodendritic homeostasis.


Subject(s)
Neuroglia , Neurons , Simian Acquired Immunodeficiency Syndrome , Spinal Cord , Animals , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/drug therapy , Spinal Cord/metabolism , Spinal Cord/drug effects , Spinal Cord/virology , Neuroglia/metabolism , Neuroglia/drug effects , Neuroglia/virology , Neurons/metabolism , Neurons/drug effects , Neurons/virology , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/pharmacology , Simian Immunodeficiency Virus/drug effects , Macaca mulatta , Gene Expression/drug effects , Male , Gene Expression Regulation/drug effects
13.
Sci Rep ; 14(1): 14715, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926482

ABSTRACT

Opioids are the gold standard for the treatment of chronic pain but are limited by adverse side effects. In our earlier work, we showed that Heat shock protein 90 (Hsp90) has a crucial role in regulating opioid signaling in spinal cord; Hsp90 inhibition in spinal cord enhances opioid anti-nociception. Building on these findings, we injected the non-selective Hsp90 inhibitor KU-32 by the intrathecal route into male and female CD-1 mice, showing that morphine anti-nociceptive potency was boosted by 1.9-3.5-fold in acute and chronic pain models. At the same time, tolerance was reduced from 21-fold to 2.9 fold and established tolerance was rescued, while the potency of constipation and reward was unchanged. These results demonstrate that spinal Hsp90 inhibition can improve the therapeutic index of morphine. However, we also found that systemic non-selective Hsp90 inhibition blocked opioid pain relief. To avoid this effect, we used selective small molecule inhibitors and CRISPR gene editing to identify 3 Hsp90 isoforms active in spinal cord (Hsp90α, Hsp90ß, and Grp94) while only Hsp90α was active in brain. We thus hypothesized that a systemically delivered selective inhibitor to Hsp90ß or Grp94 could selectively inhibit spinal cord Hsp90 activity, resulting in enhanced opioid therapy. We tested this hypothesis using intravenous delivery of KUNB106 (Hsp90ß) and KUNG65 (Grp94), showing that both drugs enhanced morphine anti-nociceptive potency while rescuing tolerance. Together, these results suggest that selective inhibition of spinal cord Hsp90 isoforms is a novel, translationally feasible strategy to improve the therapeutic index of opioids.


Subject(s)
Analgesics, Opioid , HSP90 Heat-Shock Proteins , Morphine , Spinal Cord , Animals , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Spinal Cord/metabolism , Spinal Cord/drug effects , Mice , Analgesics, Opioid/pharmacology , Male , Female , Morphine/pharmacology , Protein Isoforms/metabolism , Drug Tolerance , Chronic Pain/drug therapy , Chronic Pain/metabolism , Disease Models, Animal , Injections, Spinal
14.
PLoS One ; 19(6): e0305173, 2024.
Article in English | MEDLINE | ID: mdl-38875300

ABSTRACT

Chlorpyrifos is an organophosphate pesticide associated with numerous health effects including motor performance decrements. While many studies have focused on the health effects following acute chlorpyrifos poisonings, almost no studies have examined the effects on motoneurons following occupational-like exposures. The main objective of this study was to examine the broad effects of repeated occupational-like chlorpyrifos exposures on spinal motoneuron soma size relative to motor activity. To execute our objective, adult rats were exposed to chlorpyrifos via oral gavage once a day, five days a week for two weeks. Chlorpyrifos exposure effects were assessed either three days or two months following the last exposure. Three days following the last repeated chlorpyrifos exposure, there were transient effects in open-field motor activity and plasma cholinesterase activity levels. Two months following the chlorpyrifos exposures, there were delayed effects in sensorimotor gating, pro-inflammatory cytokines and spinal lumbar motoneuron soma morphology. Overall, these results offer support that subacute repeated occupational-like chlorpyrifos exposures have both short-term and longer-term effects in motor activity, inflammation, and central nervous system mechanisms.


Subject(s)
Chlorpyrifos , Motor Activity , Motor Neurons , Animals , Chlorpyrifos/toxicity , Motor Neurons/drug effects , Motor Neurons/pathology , Rats , Male , Motor Activity/drug effects , Insecticides/toxicity , Spinal Cord/drug effects , Spinal Cord/pathology , Rats, Sprague-Dawley , Lumbosacral Region , Cholinesterases/metabolism , Cholinesterases/blood , Cholinesterase Inhibitors/toxicity
15.
Neuromolecular Med ; 26(1): 26, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907170

ABSTRACT

Spinal cord injury (SCI) causes irreversible cell loss and neurological dysfunctions. Presently, there is no an effective clinical treatment for SCI. It can be the only intervention measure by relieving the symptoms of patients such as pain and fever. Free radical-induced damage is one of the validated mechanisms in the complex secondary injury following primary SCI. Hydrogen sulfide (H2S) as an antioxidant can effectively scavenge free radicals, protect neurons, and improve SCI by inhibiting the p38MAPK/mTOR/NF-κB signaling pathway. In this report, we analyze the pathological mechanism of SCI, the role of free radical-mediated the p38MAPK/mTOR/NF-κB signaling pathway in SCI, and the role of H2S in scavenging free radicals and improving SCI.


Subject(s)
Free Radical Scavengers , Hydrogen Sulfide , NF-kappa B , Signal Transduction , Spinal Cord Injuries , TOR Serine-Threonine Kinases , p38 Mitogen-Activated Protein Kinases , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/metabolism , Animals , Free Radical Scavengers/therapeutic use , Free Radical Scavengers/pharmacology , Signal Transduction/drug effects , Rats , Mice , Free Radicals/metabolism , Antioxidants/therapeutic use , Antioxidants/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Humans
16.
Cells ; 13(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38920670

ABSTRACT

Proinflammatory T-lymphocytes recruited into the brain and spinal cord mediate multiple sclerosis (MS) and currently there is no cure for MS. IFN-γ-producing Th1 cells induce ascending paralysis in the spinal cord while IL-17-producing Th17 cells mediate cerebellar ataxia. STAT1 and STAT3 are required for Th1 and Th17 development, respectively, and the simultaneous targeting of STAT1 and STAT3 pathways is therefore a potential therapeutic strategy for suppressing disease in the spinal cord and brain. However, the pharmacological targeting of STAT1 and STAT3 presents significant challenges because of their intracellular localization. We have developed a STAT-specific single-domain nanobody (SBT-100) derived from camelids that targets conserved residues in Src homolog 2 (SH2) domains of STAT1 and STAT3. This study investigated whether SBT-100 could suppress experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show that SBT-100 ameliorates encephalomyelitis through suppressing the expansion of Th17 and Th1 cells in the brain and spinal cord. Adoptive transfer experiments revealed that lymphocytes from SBT-100-treated EAE mice have reduced capacity to induce EAE, indicating that the immunosuppressive effects derived from the direct suppression of encephalitogenic T-cells. The small size of SBT-100 makes this STAT-specific nanobody a promising immunotherapy for CNS autoimmune diseases, including multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Single-Domain Antibodies , Th17 Cells , Animals , Female , Mice , Camelids, New World , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Mice, Inbred C57BL , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/drug therapy , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/therapeutic use , Spinal Cord/pathology , Spinal Cord/drug effects , Spinal Cord/immunology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Th1 Cells/immunology , Th1 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/drug effects
17.
Neurosci Lett ; 836: 137885, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38914276

ABSTRACT

To investigate the precise mechanism of xenon (Xe), pharmacologically isolated AMPA/KA and NMDA receptor-mediated spontaneous (s) and evoked (e) excitatory postsynaptic currents (s/eEPSCAMPA/KA and s/eEPSCNMDA) were recorded from mechanically isolated single spinal sacral dorsal commissural nucleus (SDCN) neurons attached with glutamatergic nerve endings (boutons) using conventional whole-cell patch-clamp technique. We analysed kinetic properties of both s/eEPSCAMPA/KA and s/eEPSCNMDA by focal single- and/or paired-pulse electrical stimulation to compare them. The s/eEPSCNMDA showed smaller amplitude, slower rise time, and slower 1/e decay time constant (τDecay) than those of s/eEPSCAMPA/KA. We previously examined how Xe modulates s/eEPSCAMPA/KA, therefore, examined the effects on s/eEPSCNMDA in the present study. Xe decreased the frequency and amplitude of sEPSCNMDA, and decreased the amplitude but increased the failure rate and paired-pulse ratio of eEPSCNMDA without affecting their τDecay. It was concluded that Xe might suppress NMDA receptor-mediated synaptic transmission via both presynaptic and postsynaptic mechanisms.


Subject(s)
Excitatory Postsynaptic Potentials , Neurons , Receptors, N-Methyl-D-Aspartate , Xenon , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Xenon/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Rats , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiology , Synapses/drug effects , Synapses/physiology , Rats, Sprague-Dawley , Patch-Clamp Techniques , Receptors, AMPA/metabolism , Receptors, AMPA/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Male
18.
CNS Neurosci Ther ; 30(6): e14692, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38872258

ABSTRACT

AIM: Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease characterized by progressive death of upper and lower motor neurons, leading to generalized muscle atrophy, paralysis, and even death. Mitochondrial damage and neuroinflammation play key roles in the pathogenesis of ALS. In the present study, the efficacy of A-1, a derivative of arctigenin with AMP-activated protein kinase (AMPK) and silent information regulator 1 (SIRT1) activation for ALS, was investigated. METHODS: A-1 at 33.3 mg/kg was administrated in SOD1G93A transgenic mice orally from the 13th week for a 6-week treatment period. Motor ability was assessed before terminal anesthesia. Muscle atrophy and fibrosis, motor neurons, astrocytes, and microglia in the spinal cord were evaluated by H&E, Masson, Sirius Red, Nissl, and immunohistochemistry staining. Protein expression was detected with proteomics analysis, Western blotting, and ELISA. Mitochondrial adenosine triphosphate (ATP) and malondialdehyde (MDA) levels were measured using an assay kit. RESULTS: A-1 administration in SOD1G93A mice enhanced mobility, decreased skeletal muscle atrophy and fibrosis, mitigated loss of spinal motor neurons, and reduced glial activation. Additionally, A-1 treatment improved mitochondrial function, evidenced by elevated ATP levels and increased expression of key mitochondrial-related proteins. The A-1 treatment group showed decreased levels of IL-1ß, pIκBα/IκBα, and pNF-κB/NF-κB. CONCLUSIONS: A-1 treatment reduced motor neuron loss, improved gastrocnemius atrophy, and delayed ALS progression through the AMPK/SIRT1/PGC-1α pathway, which promotes mitochondrial biogenesis. Furthermore, the AMPK/SIRT1/IL-1ß/NF-κB pathway exerted neuroprotective effects by reducing neuroinflammation. These findings suggest A-1 as a promising therapeutic approach for ALS.


Subject(s)
AMP-Activated Protein Kinases , Amyotrophic Lateral Sclerosis , Furans , Interleukin-1beta , Mice, Transgenic , NF-kappa B , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Mice , NF-kappa B/metabolism , AMP-Activated Protein Kinases/metabolism , Furans/pharmacology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Interleukin-1beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Lignans/pharmacology , Lignans/therapeutic use , Signal Transduction/drug effects , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Male , Motor Neurons/drug effects , Motor Neurons/pathology , Motor Neurons/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/metabolism
19.
Biomed Pharmacother ; 176: 116856, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38852510

ABSTRACT

Fibromyalgia is characterised by widespread chronic pain and is often accompanied by comorbidities such as sleep disorders, anxiety, and depression. Because it is often accompanied by many adverse symptoms and lack of effective treatment, it is important to search for the pathogenesis and treatment of fibromyalgia. Astaxanthin, a carotenoid pigment known for its anti-inflammatory and antioxidant properties, has demonstrated effective analgesic effects in neuropathic pain. However, its impact on fibromyalgia remains unclear. Therefore, in this study, we constructed a mouse model of fibromyalgia and investigated the effect of astaxanthin on chronic pain and associated symptoms through multiple intragastrical injections. We conducted behavioural assessments to detect pain and depression-like states in mice, recorded electroencephalograms to monitor sleep stages, examined c-Fos activation in the anterior cingulate cortex, measured activation of spinal glial cells, and assessed levels of inflammatory factors in the brain and spinal cord, including interleukin (IL)-1ß, IL-6, and tumour necrosis factor- α(TNF-α).Additionally, we analysed the expression levels of IL-6, IL-10, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), Apoptosis-associated speck-like protein containing CARD, and Caspase-1 proteins. The findings revealed that astaxanthin significantly ameliorated mechanical and thermal pain in mice with fibromyalgia and mitigated sleep disorders and depressive-like symptoms induced by pain. A potential mechanism underlying these effects is the anti-inflammatory action of astaxanthin, likely mediated through the inhibition of the NLRP3 inflammasome, which could be one of the pathways through which astaxanthin alleviates fibromyalgia. In conclusion, our study suggests that astaxanthin holds promise as a potential analgesic medication for managing fibromyalgia and its associated symptoms.


Subject(s)
Depression , Fibromyalgia , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Xanthophylls , Animals , Xanthophylls/pharmacology , Fibromyalgia/drug therapy , Fibromyalgia/complications , Fibromyalgia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Inflammasomes/metabolism , Inflammasomes/antagonists & inhibitors , Depression/drug therapy , Depression/metabolism , Mice , Male , Mice, Inbred C57BL , Disease Models, Animal , Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Chronic Pain/drug therapy , Chronic Pain/metabolism , Cytokines/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Behavior, Animal/drug effects
20.
J Neurosci ; 44(29)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38886057

ABSTRACT

Calcineurin inhibitors, such as cyclosporine and tacrolimus (FK506), are commonly used immunosuppressants for preserving transplanted organs and tissues. However, these drugs can cause severe and persistent pain. GluA2-lacking, calcium-permeable AMPA receptors (CP-AMPARs) are implicated in various neurological disorders, including neuropathic pain. It is unclear whether and how constitutive calcineurin, a Ca2+/calmodulin protein phosphatase, controls synaptic CP-AMPARs. In this study, we found that blocking CP-AMPARs with IEM-1460 markedly reduced the amplitude of AMPAR-EPSCs in excitatory neurons expressing vesicular glutamate transporter-2 (VGluT2), but not in inhibitory neurons expressing vesicular GABA transporter, in the spinal cord of FK506-treated male and female mice. FK506 treatment also caused an inward rectification in the current-voltage relationship of AMPAR-EPSCs specifically in VGluT2 neurons. Intrathecal injection of IEM-1460 rapidly alleviated pain hypersensitivity in FK506-treated mice. Furthermore, FK506 treatment substantially increased physical interaction of α2δ-1 with GluA1 and GluA2 in the spinal cord and reduced GluA1/GluA2 heteromers in endoplasmic reticulum-enriched fractions of spinal cords. Correspondingly, inhibiting α2δ-1 with pregabalin, Cacna2d1 genetic knock-out, or disrupting α2δ-1-AMPAR interactions with an α2δ-1 C terminus peptide reversed inward rectification of AMPAR-EPSCs in spinal VGluT2 neurons caused by FK506 treatment. In addition, CK2 inhibition reversed FK506 treatment-induced pain hypersensitivity, α2δ-1 interactions with GluA1 and GluA2, and inward rectification of AMPAR-EPSCs in spinal VGluT2 neurons. Thus, the increased prevalence of synaptic CP-AMPARs in spinal excitatory neurons plays a major role in calcineurin inhibitor-induced pain hypersensitivity. Calcineurin and CK2 antagonistically regulate postsynaptic CP-AMPARs through α2δ-1-mediated GluA1/GluA2 heteromeric assembly in the spinal dorsal horn.


Subject(s)
Calcineurin , Casein Kinase II , Receptors, AMPA , Spinal Cord , Tacrolimus , Animals , Receptors, AMPA/metabolism , Mice , Calcineurin/metabolism , Male , Female , Tacrolimus/pharmacology , Spinal Cord/metabolism , Spinal Cord/drug effects , Casein Kinase II/metabolism , Neurons/metabolism , Neurons/drug effects , Mice, Inbred C57BL , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Synapses/drug effects , Synapses/metabolism , Synapses/physiology , Calcineurin Inhibitors/pharmacology , Phenotype , Calcium Channels
SELECTION OF CITATIONS
SEARCH DETAIL