Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Small ; 19(9): e2203464, 2023 03.
Article in English | MEDLINE | ID: mdl-36526612

ABSTRACT

Three-dimensional (3D) bioprinting, which is being increasingly used in tissue engineering, requires bioinks with tunable mechanical properties, biological activities, and mechanical strength for in vivo implantation. Herein, a growth-factor-holding poly(organophosphazene)-based thermo-responsive nanocomposite (TNC) bioink system is developed. The mechanical properties of the TNC bioink are easily controlled within a moderate temperature range (5-37 °C). During printing, the mechanical properties of the TNC bioink, which determine the 3D printing resolution, can be tuned by varying the temperature (15-30 °C). After printing, TNC bioink scaffolds exhibit maximum stiffness at 37 °C. Additionally, because of its shear-thinning and self-healing properties, TNC bioinks can be extruded smoothly, demonstrating good printing outcomes. TNC bioink loaded with bone morphogenetic protein-2 (BMP-2) and transforming growth factor-beta1 (TGF-ß1), key growth factors for osteogenesis, is used to print a scaffold that can stimulate biological activity. A biological scaffold printed using TNC bioink loaded with both growth factors and implanted on a rat calvarial defect model reveals significantly improved bone regenerative effects. The TNC bioink system is a promising next-generation bioink platform because its mechanical properties can be tuned easily for high-resolution 3D bioprinting with long-term stability and its growth-factor holding capability has strong clinical applicability.


Subject(s)
Bioprinting , Nanocomposites , Animals , Rats , Tissue Scaffolds , Tissue Engineering/methods , Bioprinting/methods , Printing, Three-Dimensional , Bone Regeneration
2.
Mol Pain ; 15: 1744806919843046, 2019.
Article in English | MEDLINE | ID: mdl-30900515

ABSTRACT

Spinal D-serine plays an important role in nociception via an increase in phosphorylation of the N-Methyl-D-aspartate (NMDA) receptor GluN1 subunit (pGluN1). However, the cellular mechanisms underlying this process have not been elucidated. Here, we investigate the possible role of neuronal nitric oxide synthase (nNOS) in the D-serine-induced potentiation of NMDA receptor function and the induction of neuropathic pain in a chronic constriction injury (CCI) model. Intrathecal administration of the serine racemase inhibitor, L-serine O-sulfate potassium salt (LSOS) or the D-serine degrading enzyme, D-amino acid oxidase (DAAO) on post-operative days 0-3 significantly reduced the CCI-induced increase in nitric oxide (NO) levels and nicotinamide adenine dinucleotide phosphate-diaphorase staining in lumbar dorsal horn neurons, as well as the CCI-induced decrease in phosphorylation (Ser847) of nNOS (pnNOS) on day 3 post-CCI surgery. LSOS or DAAO administration suppressed the CCI-induced development of mechanical allodynia and protein kinase C (PKC)-dependent (Ser896) phosphorylation of GluN1 on day 3 post-surgery, which were reversed by the co-administration of the NO donor, 3-morpholinosydnonimine hydrochloride (SIN-1). In naïve mice, exogenous D-serine increased NO levels via decreases in pnNOS. D-serine-induced increases in mechanical hypersensitivity, NO levels, PKC-dependent pGluN1, and NMDA-induced spontaneous nociception were reduced by pretreatment with the nNOS inhibitor, 7-nitroindazole or with the NMDA receptor antagonists, 7-chlorokynurenic acid and MK-801. Collectively, we show that spinal D-serine modulates nNOS activity and concomitant NO production leading to increases in PKC-dependent pGluN1 and ultimately contributing to the induction of mechanical allodynia following peripheral nerve injury.


Subject(s)
Astrocytes/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Nitric Oxide Synthase Type I/metabolism , Serine/pharmacology , Animals , Blotting, Western , D-Amino-Acid Oxidase/metabolism , Hyperalgesia/etiology , Male , Mice , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , N-Methylaspartate/metabolism , Neuralgia/etiology , Phosphorylation/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/analogs & derivatives , Serine/metabolism
3.
Biol Pharm Bull ; 39(12): 1922-1931, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27601184

ABSTRACT

We recently demonstrated that activation of spinal sigma-1 receptors (Sig-1Rs) induces pain hypersensitivity via the activation of neuronal nitric oxide synthase (nNOS) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2). However, the potential direct interaction between nNOS-derived nitric oxide (NO) and Nox2-derived reactive oxygen species (ROS) is poorly understood, particularly with respect to the potentiation of N-methyl-D-aspartate (NMDA) receptor activity in the spinal cord associated with the development of central sensitization. Thus, the main purpose of this study was to investigate whether Sig-1R-induced and nNOS-derived NO modulates spinal Nox2 activation leading to an increase in ROS production and ultimately to the potentiation of NMDA receptor activity and pain hypersensitivity. Intrathecal pretreatment with the nNOS inhibitor, 7-nitroindazole or with the Nox inhibitor, apocynin significantly inhibited the mechanical and thermal hypersensitivity induced by intrathecal administration of the Sig-1R agonist, 2-(4-morpholinethyl) 1-phenylcyclohexanecarboxylate hydrochloride (PRE084). Conversely, pretreatment with 5,10,15,20-tetrakis-(4-sulphonatophenyl)-porphyrinato iron(III) (FeTPPS; a scavenger of peroxynitrite, a toxic reaction product of NO and superoxide) had no effect on the PRE084-induced pain hypersensitivity. Pretreatment with 7-nitroindazole significantly reduced the PRE084-induced increase in Nox2 activity and concomitant ROS production in the lumbar spinal cord dorsal horn, whereas apocynin did not alter the PRE084-induced changes in nNOS phosphorylation. On the other hand pretreatment with apocynin suppressed the PRE084-induced increase in the protein kinase C (PKC)-dependent phosphorylation of NMDA receptor GluN1 subunit (pGluN1) at Ser896 site in the dorsal horn. These findings demonstrate that spinal Sig-1R-induced pain hypersensitivity is mediated by nNOS activation, which leads to an increase in Nox2 activity ultimately resulting in a ROS-induced increase in PKC-dependent pGluN1 expression.


Subject(s)
Hyperalgesia/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/metabolism , Animals , Hot Temperature , Male , Mice, Inbred ICR , NADPH Oxidase 2 , Nitric Oxide/metabolism , Pain/metabolism , Physical Stimulation , Spinal Cord Dorsal Horn/metabolism , Sigma-1 Receptor
4.
Pharmacol Res ; 100: 353-64, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26316425

ABSTRACT

We have previously demonstrated that activation of the spinal sigma-1 receptor (Sig-1R) plays an important role in the development of mechanical allodynia (MA) via secondary activation of the N-methyl-d-aspartate (NMDA) receptor. Sig-1Rs have been shown to localize to astrocytes, and blockade of Sig-1Rs inhibits the pathologic activation of astrocytes in neuropathic mice. However, the mechanism by which Sig-1R activation in astrocytes modulates NMDA receptors in neurons is currently unknown. d-serine, synthesized from l-serine by serine racemase (Srr) in astrocytes, is an endogenous co-agonist for the NMDA receptor glycine site and can control NMDA receptor activity. Here, we investigated the role of d-serine in the development of MA induced by spinal Sig-1R activation in chronic constriction injury (CCI) mice. The production of d-serine and Srr expression were both significantly increased in the spinal cord dorsal horn post-CCI surgery. Srr and d-serine were only localized to astrocytes in the superficial dorsal horn, while d-serine was also localized to neurons in the deep dorsal horn. Moreover, we found that Srr exists in astrocytes that express Sig-1Rs. The CCI-induced increase in the levels of d-serine and Srr was attenuated by sustained intrathecal treatment with the Sig-1R antagonist, BD-1047 during the induction phase of neuropathic pain. In behavioral experiments, degradation of endogenous d-serine with DAAO, or selective blockade of Srr by LSOS, effectively reduced the development of MA, but not thermal hyperalgesia in CCI mice. Finally, BD-1047 administration inhibited the development of MA and this inhibition was reversed by intrathecal treatment with exogenous d-serine. These findings demonstrate for the first time that the activation of Sig-1Rs increases the expression of Srr and d-serine in astrocytes. The increased production of d-serine induced by CCI ultimately affects dorsal horn neurons that are involved in the development of MA in neuropathic mice.


Subject(s)
Astrocytes/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Receptors, sigma/metabolism , Serine/metabolism , Animals , Astrocytes/drug effects , Disease Models, Animal , Ethylenediamines/pharmacology , Male , Mice , Mice, Inbred ICR , Posterior Horn Cells/metabolism , Racemases and Epimerases/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Sigma-1 Receptor
5.
Mol Pain ; 10: 2, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24401144

ABSTRACT

BACKGROUND: We previously developed a thrombus-induced ischemic pain (TIIP) animal model, which was characterized by chronic bilateral mechanical allodynia without thermal hyperalgesia (TH). On the other hand we had shown that intraplantar injection of acidic saline facilitated ATP-induced pain, which did result in the induction of TH in normal rats. Because acidic pH and increased ATP are closely associated with ischemic conditions, this study is designed to: (1) examine whether acidic saline injection into the hind paw causes the development of TH in TIIP, but not control, animals; and (2) determine which peripheral mechanisms are involved in the development of this TH. RESULTS: Repeated intraplantar injection of pH 4.0 saline, but not pH 5.5 and 7.0 saline, for 3 days following TIIP surgery resulted in the development of TH. After pH 4.0 saline injections, protein levels of hypoxia inducible factor-1α (HIF-1α) and carbonic anhydrase II (CA II) were elevated in the plantar muscle indicating that acidic stimulation intensified ischemic insults with decreased tissue acidity. At the same time point, there were no changes in the expression of TRPV1 in hind paw skin, whereas a significant increase in TRPV1 phosphorylation (pTRPV1) was shown in acidic saline (pH 4.0) injected TIIP (AS-TIIP) animals. Moreover, intraplantar injection of chelerythrine (a PKC inhibitor) and AMG9810 (a TRPV1 antagonist) effectively alleviated the established TH. In order to investigate which proton- or ATP-sensing receptors contributed to the development of TH, amiloride (an ASICs blocker), AMG9810, TNP-ATP (a P2Xs antagonist) or MRS2179 (a P2Y1 antagonist) were pre-injected before the pH 4.0 saline. Only MRS2179 significantly prevented the induction of TH, and the increased pTRPV1 ratio was also blocked in MRS2179 injected animals. CONCLUSION: Collectively these data show that maintenance of an acidic environment in the ischemic hind paw of TIIP rats results in the phosphorylation of TRPV1 receptors via a PKC-dependent pathway, which leads to the development of TH mimicking what occurs in chronic ischemic patients with severe acidosis. More importantly, peripheral P2Y1 receptors play a pivotal role in this process, suggesting a novel peripheral mechanism underlying the development of TH in these patients.


Subject(s)
Hindlimb/blood supply , Hyperalgesia/complications , Ischemia/etiology , Pain/etiology , Receptors, Purinergic P2Y1/metabolism , TRPV Cation Channels/metabolism , Thrombosis/complications , Acids , Acrylamides/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Benzophenanthridines/pharmacology , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Disease Models, Animal , Diterpenes/pharmacology , Hindlimb/pathology , Hot Temperature , Hyperalgesia/metabolism , Hyperalgesia/pathology , Hypoxia/etiology , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Injections , Ion Channels/metabolism , Ischemia/metabolism , Ischemia/pathology , Pain/metabolism , Pain/pathology , Phosphorylation/drug effects , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Sodium Chloride/administration & dosage , Sodium Chloride/pharmacology , Thrombosis/metabolism , Thrombosis/pathology , Tissue Extracts
6.
Acta Physiol (Oxf) ; : e14236, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39324481

ABSTRACT

AIM: This investigation addresses Piezo1's expression and mechanistic role in dorsal root ganglion (DRG) neurons and delineates its participation in mechanical and inflammatory pain modulation. METHODS: We analyzed Piezo1's expression patterns in DRG neurons and utilized Piezo1-specific shRNA to modulate its activity. Electrophysiological assessments of mechanically activated (MA) currents in DRG neurons and behavioral analyses in mouse models of inflammatory pain were conducted to elucidate Piezo1's functional implications. Additionally, we investigated the excitability of TRPV1-expressing DRG neurons, particularly under inflammatory conditions. RESULTS: Piezo1 was preferentially expressed in DRG neurons co-expressing the TRPV1 nociceptor marker. Knockdown of Piezo1 attenuated intermediately adapting MA currents and lessened tactile pain hypersensitivity in models of inflammatory pain. Additionally, silencing Piezo1 modified the excitability of TRPV1-expressing neurons under inflammatory stress. CONCLUSION: Piezo1 emerges as a key mediator in the transmission of mechanical and inflammatory pain, indicating its potential as a novel target for pain management therapies. Our finding not only advances the understanding of nociceptive signaling but also emphasizes the therapeutic potential of modulating Piezo1 in the treatment of pain.

7.
Mol Brain ; 17(1): 50, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095918

ABSTRACT

Neuroactive steroids (NASs) directly affect neuronal excitability. Despite their role in the nervous system is intimately linked to pain control, knowledge is currently limited. This study investigates the peripheral involvement of NASs in chronic ischemic pain by targeting the cytochrome P450 side-chain cleavage enzyme (P450scc). Using a rat model of hind limb thrombus-induced ischemic pain (TIIP), we observed an increase in P450scc expression in the ischemic hind paw skin. Inhibiting P450scc with intraplantar aminoglutethimide (AMG) administration from post-operative day 0 to 3 significantly reduced the development of mechanical allodynia. However, AMG administration from post-operative day 3 to 6 did not affect established mechanical allodynia. In addition, we explored the role of the peripheral sigma-1 receptor (Sig-1R) by co-administering PRE-084 (PRE), a Sig-1R agonist, with AMG. PRE reversed the analgesic effects of AMG during the induction phase. These findings indicate that inhibiting steroidogenesis with AMG alleviates peripheral ischemic pain during the induction phase via Sig-1Rs.


Subject(s)
Disease Models, Animal , Hyperalgesia , Ischemia , Rats, Sprague-Dawley , Receptors, sigma , Animals , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Hyperalgesia/complications , Male , Ischemia/complications , Ischemia/pathology , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Sigma-1 Receptor , Pain/drug therapy , Pain/complications , Pain/etiology , Pain/pathology , Hindlimb/drug effects , Rats , Cytochrome P-450 Enzyme System/metabolism
8.
Pharmacol Res ; 74: 56-67, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732704

ABSTRACT

We have recently demonstrated that spinal sigma-1 receptors (Sig-1Rs) mediate pain hypersensitivity in mice and neuropathic pain in rats. In this study, we examine the role of NADPH oxidase 2 (Nox2)-induced reactive oxygen species (ROS) on Sig-1R-induced pain hypersensitivity and the induction of chronic neuropathic pain. Neuropathic pain was produced by chronic constriction injury (CCI) of the right sciatic nerve in rats. Mechanical allodynia and thermal hyperalgesia were evaluated in mice and CCI-rats. Western blotting and dihydroethidium (DHE) staining were performed to assess the changes in Nox2 activation and ROS production in spinal cord, respectively. Direct activation of spinal Sig-1Rs with the Sig-1R agonist, PRE084 induced mechanical allodynia and thermal hyperalgesia, which were dose-dependently attenuated by pretreatment with the ROS scavenger, NAC or the Nox inhibitor, apocynin. PRE084 also induced an increase in Nox2 activation and ROS production, which were attenuated by pretreatment with the Sig-1R antagonist, BD1047 or apocynin. CCI-induced nerve injury produced an increase in Nox2 activation and ROS production in the spinal cord, all of which were attenuated by intrathecal administration with BD1047 during the induction phase of neuropathic pain. Furthermore, administration with BD1047 or apocynin reversed CCI-induced mechanical allodynia during the induction phase, but not the maintenance phase. These findings demonstrate that spinal Sig-1Rs modulate Nox2 activation and ROS production in the spinal cord, and ultimately contribute to the Sig-1R-induced pain hypersensitivity and the peripheral nerve injury-induced induction of chronic neuropathic pain.


Subject(s)
Hyperalgesia/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Neuralgia/metabolism , Receptors, sigma/metabolism , Animals , Ethylenediamines/pharmacology , Hot Temperature , Male , Mice , Mice, Inbred ICR , Morpholines/pharmacology , NADPH Oxidase 2 , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Spinal Cord/metabolism , Touch , Sigma-1 Receptor
9.
Neurobiol Aging ; 132: 175-184, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37837733

ABSTRACT

The anti-inflammatory efficacy of radiation therapy (RT) with single fractions below 1.0 Gy has been demonstrated in Alzheimer's disease mouse models. As neuroinflammation is also a major pathological feature of Parkinson's disease (PD), RT may also be effective in PD treatment. Therefore, this study aimed to investigate the anti-inflammatory effect of low-moderate dose RT (LMDRT, 0.6 Gy/single dose, for 5 days) exposure in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 30 mg/kg, intraperitoneally, for 5 consecutive days)-induced PD mouse model. Importantly, LMDRT reduced the levels of glial fibrillary acidic protein and intercellular adhesion molecule-1 (CD54) in the striatum region, which increased following MPTP administration. LMDRT also modulated inflammatory gene expression patterns in the substantia nigra region of the MPTP-treated mice. However, LMDRT had no direct effects on the severe loss of dopaminergic neurons and impaired motor behavior in the rotarod test. These results indicate that LMDRT has anti-inflammatory effects by modulating neuroinflammatory factors, including glial fibrillary acidic protein and intercellular adhesion molecule-1, but showed no behavioral improvements or neuroprotection in the MPTP-induced mouse model of PD.


Subject(s)
Brain , Glial Fibrillary Acidic Protein , Parkinson Disease , Animals , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/therapeutic use , Brain/metabolism , Brain/radiation effects , Disease Models, Animal , Dopaminergic Neurons/pathology , Glial Fibrillary Acidic Protein/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/pharmacology , Intercellular Adhesion Molecule-1/therapeutic use , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/radiotherapy , Substantia Nigra/metabolism
10.
Biomed Pharmacother ; 166: 115312, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37567072

ABSTRACT

Histone deacetylases (HDACs) are key epigenetic regulators and classified into four subtypes. Despite the various roles of each HDAC isoform, the lack of selective HDAC inhibitors has limited the elucidation of their roles in biological systems. HDAC11, the sole class-IV HDAC, is highly expressed in the brain, however, the role of HDAC11 in microglia is not fully understood. Based on the modification of MC1568, we developed a novel HDAC inhibitor, 5. Interestingly, 5 suppresses lipopolysaccharide-induced microglial activation by the initiation of autophagy and subsequent inhibition of nitric oxide production. Furthermore, we demonstrated that 5 significantly alleviates depression-like behavior by inhibiting microglial activation in mouse brain. Our discovery reveals that specific pharmacological regulation of HDAC11 induces autophagy and reactive nitrogen species balance in microglia for the first time, which makes HDAC11 a new therapeutic target for depressive disorder.


Subject(s)
Depression , Histone Deacetylase Inhibitors , Microglia , Animals , Mice , Brain/drug effects , Brain/metabolism , Depression/drug therapy , Depression/genetics , Depression/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Microglia/drug effects , Microglia/metabolism
11.
Neuropharmacology ; 149: 169-180, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30797030

ABSTRACT

While evidence indicates that sigma-1 receptors (Sig-1Rs) play an important role in the induction of peripheral neuropathic pain, there is limited understanding of the role that the neurosteroidogenic enzymes, which produce Sig-1R endogenous ligands, play during the development of neuropathic pain. We examined whether sciatic nerve injury upregulates the neurosteroidogenic enzymes, cytochrome P450c17 and 3ß-hydroxysteroid dehydrogenase (3ß-HSD), which modulate the expression and/or activation of Sig-1Rs leading to the development of peripheral neuropathic pain. Chronic constriction injury (CCI) of the sciatic nerve induced a significant increase in the expression of P450c17, but not 3ß-HSD, in the ipsilateral lumbar spinal cord dorsal horn at postoperative day 3. Intrathecal administration of the P450c17 inhibitor, ketoconazole during the induction phase of neuropathic pain (day 0 to day 3 post-surgery) significantly reduced the development of mechanical allodynia and thermal hyperalgesia in the ipsilateral hind paw. However, administration of the 3ß-HSD inhibitor, trilostane had no effect on the development of neuropathic pain. Sciatic nerve injury increased astrocyte Sig-1R expression as well as dissociation of Sig-1Rs from BiP in the spinal cord. These increases were suppressed by administration of ketoconazole, but not by administration of trilostane. Co-administration of the Sig-1R agonist, PRE084 restored the development of mechanical allodynia originally suppressed by the ketoconazole administration. However, ketoconazole-induced inhibition of thermal hyperalgesia was not affected by co-administration of PRE084. Collectively these results demonstrate that early activation of P450c17 modulates the expression and activation of astrocyte Sig-1Rs, ultimately contributing to the development of mechanical allodynia induced by peripheral nerve injury.


Subject(s)
Hyperalgesia/metabolism , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, sigma/metabolism , Spinal Cord/enzymology , Steroid 17-alpha-Hydroxylase/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Animals , Astrocytes , Dihydrotestosterone/analogs & derivatives , Dihydrotestosterone/pharmacology , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/enzymology , Hyperalgesia/prevention & control , Ketoconazole/pharmacology , Male , Mice , Mice, Inbred ICR , Neuralgia/enzymology , Neurosteroids/metabolism , Peripheral Nerve Injuries/chemically induced , Peripheral Nerve Injuries/enzymology , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Receptors, sigma/agonists , Sciatic Nerve/enzymology , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Spinal Cord/drug effects , Spinal Cord Dorsal Horn/metabolism , Sigma-1 Receptor
12.
J Vet Sci ; 19(5): 708-715, 2018 Sep 30.
Article in English | MEDLINE | ID: mdl-29929357

ABSTRACT

Respiratory inflammation is a frequent and fatal pathologic state encountered in veterinary medicine. Although diluted bee venom (dBV) has potent anti-inflammatory effects, the clinical use of dBV is limited to several chronic inflammatory diseases. The present study was designed to propose an acupoint dBV treatment as a novel therapeutic strategy for respiratory inflammatory disease. Experimental pleurisy was induced by injection of carrageenan into the left pleural space in mouse. The dBV was injected into a specific lung meridian acupoint (LU-5) or into an arbitrary non-acupoint located near the midline of the back in mouse. The inflammatory responses were evaluated by analyzing inflammatory indicators in pleural exudate. The dBV injection into the LU-5 acupoint significantly suppressed the carrageenan-induced increase of pleural exudate volume, leukocyte accumulation, and myeloperoxidase activity. Moreover, dBV acupoint treatment effectively inhibited the production of interleukin 1 beta, but not tumor necrosis factor alpha in the pleural exudate. On the other hand, dBV treatment at non-acupoint did not inhibit the inflammatory responses in carrageenan-induced pleurisy. The present results demonstrate that dBV stimulation in the LU-5 lung meridian acupoint can produce significant anti-inflammatory effects on carrageenan-induced pleurisy suggesting that dBV acupuncture may be a promising alternative medicine therapy for respiratory inflammatory diseases.


Subject(s)
Acupuncture Points , Bee Venoms/therapeutic use , Inflammation/therapy , Pleurisy/therapy , Animals , Carrageenan/pharmacology , Inflammation/chemically induced , Lung/immunology , Male , Mice , Mice, Inbred BALB C , Pleurisy/chemically induced
13.
Neuroscience ; 372: 181-191, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29289721

ABSTRACT

Aromatase is a key enzyme responsible for the biosynthesis of estrogen from testosterone. Although recent evidence indicates that spinal cord aromatase participates in nociceptive processing, the mechanisms underlying its regulation and its involvement in nociception remain unclear. The present study focuses on the potential role of astrocyte aromatase in formalin-induced acute pain and begins to uncover one mechanism by which spinal aromatase activation is controlled. Following intraplantar formalin injection, nociceptive responses were quantified and immunohistochemistry/co-immunoprecipitation assays were used to investigate the changes in spinal Fos expression and the phospho-serine levels of spinal aromatase. Intrathecal (i.t.) injection of letrozole (an aromatase inhibitor) mitigated both the late phase formalin-induced nociceptive responses and formalin-induced spinal Fos expression. Furthermore, formalin-injected mice showed significantly reduced phospho-serine levels of aromatase, which is associated with the rapid activation of this enzyme. However, sigma-1 receptor inhibition with i.t. BD1047 blocked the dephosphorylation of aromatase and potentiated the pharmacological effect of letrozole on formalin-induced nociceptive responses. In addition, i.t. administration of a sub-effective dose of BD1047 potentiated the pharmacological effect of cyclosporin A (a calcineurin inhibitor) on both the formalin-induced reduction in phospho-serine levels of aromatase and nociceptive behavior. These results suggest that dephosphorylation is an important regulatory mechanism involved in the rapid activation of aromatase and that spinal sigma-1 receptors mediate this dephosphorylation of aromatase through an intrinsic calcineurin pathway.


Subject(s)
Aromatase/metabolism , Astrocytes/metabolism , Inflammation/metabolism , Nociceptive Pain/metabolism , Spinal Cord/metabolism , Animals , Aromatase Inhibitors/pharmacology , Astrocytes/drug effects , Astrocytes/pathology , Calcineurin/metabolism , Formaldehyde , Glial Fibrillary Acidic Protein/metabolism , Inflammation/drug therapy , Inflammation/pathology , Letrozole , Male , Mice, Inbred ICR , Nitriles/pharmacology , Nociceptive Pain/drug therapy , Nociceptive Pain/pathology , Oncogene Proteins v-fos/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Receptors, sigma/metabolism , Spinal Cord/drug effects , Spinal Cord/pathology , Triazoles/pharmacology , Sigma-1 Receptor
14.
Br J Pharmacol ; 175(3): 558-572, 2018 02.
Article in English | MEDLINE | ID: mdl-29172248

ABSTRACT

BACKGROUND AND PURPOSE: Although we have recently demonstrated that spinal astrocyte gap junctions mediate the development of mirror-image pain (MIP), it is still unclear which astrocyte-derived factor is responsible for the development of MIP and how its production is controlled. In the present study, we focused on the role of ipsilateral versus contralateral D-serine in the development of MIP and investigated the possible involvement of σ1 receptors and gap junctions in astrocyte D-serine production. EXPERIMENTAL APPROACH: Following carrageenan injection, mechanical allodynia was tested at various time points to examine the effect of individual drugs. Immunohistochemistry and Western blot analyses were performed to clarify the expression levels of spinal D-serine, serine racemase, σ1 receptors and connexin 43. KEY RESULTS: The expression of ipsilateral D-serine was up-regulated during the early phase of inflammation, while contralateral D-serine increased during the later phase of inflammation. The pharmacological inhibition of D-serine during the early phase blocked the development of both ipsilateral and contralateral mechanical allodynia. However, the inhibition of D-serine during the later phase of inflammation blocked contralateral, but not ipsilateral mechanical allodynia. Furthermore, the inhibition of σ1 receptors during the earlier phase of inflammation inhibited the increase in ipsilateral D-serine. Conversely, the blockade of astrocyte gap junctions suppressed the up-regulation of contralateral D-serine during the later phase of inflammation. CONCLUSION AND IMPLICATIONS: Spinal astrocyte D-serine plays an important role in the development of mirror-image pain. Furthermore, σ1 receptors and astrocyte gap junction signalling mediate ipsilateral and contralateral D-serine production respectively.


Subject(s)
Astrocytes/physiology , Carrageenan/toxicity , Gap Junctions/physiology , Pain/drug therapy , Receptors, sigma/physiology , Serine/administration & dosage , Animals , Astrocytes/drug effects , Dose-Response Relationship, Drug , Gap Junctions/drug effects , Injections, Spinal , Male , Pain/chemically induced , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/physiology , Sigma-1 Receptor
15.
Brain Res Bull ; 130: 165-172, 2017 04.
Article in English | MEDLINE | ID: mdl-28153540

ABSTRACT

Although interleukin-1ß (IL-1ß) is a prototypical pro-inflammatory cytokine, the specific mechanisms underlying the role of its cognate receptor, the interleukin-1 receptor (IL-1R) in peripheral sensitization remain to be investigated. Since emerging evidence in the literature indicates that IL-1ß can modulate membrane-bound receptors, we decided to examine the involvement of P2Y1 receptor (P2Y1R) in IL-1ß induced pain and the potential interaction of P2Y1Rs and IL-1Rs in both naïve and carrageenan injected rats. Intraplantar (i.pl) injection of IL-1ß dose-dependently produced mechanical and thermal hypersensitivity in naïve rats. Pre-treatment with IL-1ra (i.pl, 30 and 100ng), an endogenous IL-1R antagonist, prevented the IL-1ß induced mechanical and thermal hypersensitivity. Pre-treatment with MRS2500 (i.pl, 1 and 3nmol), a specific P2Y1R antagonist, dose-dependently reduced IL-1ß induced thermal hypersensitivity, but did not affect the development of mechanical hypersensitivity. Conversely coadministration of MRS2500 (i.pl, 0.1nmol, sub-effective dose) together with IL-1ra (10nmol, sub-effective dose) significantly reduced IL-1ß induced thermal, but not mechanical hypersensitivity. We next used immunohistochemistry to demonstrate that P2Y1 and IL-1 type I receptors co-localize predominantly in small diameter neurons in the dorsal root ganglion. We also performed experiments to examine the interaction of P2Y1Rs and IL-1Rs under the inflammatory conditions induced by 2% carrageenan. Intraplantar coadministration of MRS2500 (3nmol, sub-effective dose) and IL-1ra (30ng, sub-effective dose) significantly reduced inflammatory thermal, but not mechanical, hypersensitivity. These data indicate the involvement of P2Y1Rs in IL-1ß mediated pain in both naive and carrageenan injected rats. There is a positive interaction between peripheral P2Y1Rs and IL-1Rs in both IL-1ß and carrageenan-induced thermal hypersensitivity.


Subject(s)
Hyperalgesia/physiopathology , Interleukin-1beta/physiology , Receptors, Interleukin-1/physiology , Receptors, Purinergic P2Y1/physiology , Animals , Carrageenan/administration & dosage , Hyperalgesia/chemically induced , Interleukin-1beta/administration & dosage , Male , Pain Threshold , Rats, Sprague-Dawley
16.
J Pain ; 18(4): 415-427, 2017 04.
Article in English | MEDLINE | ID: mdl-27986591

ABSTRACT

We have recently shown that spinal sigma-1 receptor (Sig-1R) activation facilitates nociception via an increase in phosphorylation of the N-methyl-D-aspartate (NMDA) receptor GluN1 subunit (pGluN1). The present study was designed to examine whether the Sig-1R-induced facilitative effect on NMDA-induced nociception is mediated by D-serine, and whether D-serine modulates spinal pGluN1 expression and the development of neuropathic pain after chronic constriction injury (CCI) of the sciatic nerve. Intrathecal administration of the D-serine degrading enzyme, D-amino acid oxidase attenuated the facilitation of NMDA-induced nociception induced by the Sig-1R agonist, 2-(4-morpholinethyl)1-phenylcyclohexane carboxylate. Exogenous D-serine increased protein kinase C (PKC)-dependent (Ser896) pGluN1 expression and facilitated NMDA-induced nociception, which was attenuated by preteatment with the PKC inhibitor, chelerythrine. In CCI mice, administration of the serine racemase inhibitor, L-serine O-sulfate potassium salt or D-amino acid oxidase on postoperative days 0 to 3 suppressed CCI-induced mechanical allodynia (MA) and pGluN1 expression on day 3 after CCI surgery. Intrathecal administration of D-serine restored MA as well as the GluN1 phosphorylation on day 3 after surgery that was suppressed by the Sig-1R antagonist, N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide or the astrocyte inhibitor, fluorocitrate. In contrast, D-serine had no effect on CCI-induced thermal hyperalgesia or GluN1 expression. These results indicate that spinal D-serine: 1) mediates the facilitative effect of Sig-1R on NMDA-induced nociception, 2) modulates PKC-dependent pGluN1 expression, and 3) ultimately contributes to the induction of MA after peripheral nerve injury. PERSPECTIVE: This report shows that reducing D-serine suppresses central sensitization and significantly alleviates peripheral nerve injury-induced chronic neuropathic pain and that this process is modulated by spinal Sig-1Rs. This preclinical evidence provides a strong rationale for using D-serine antagonists to treat peripheral nerve injury-induced neuropathy.


Subject(s)
Hyperalgesia/etiology , Nerve Tissue Proteins/metabolism , Neuralgia/complications , Protein Kinase C/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, sigma/metabolism , Serine/pharmacology , Animals , D-Amino-Acid Oxidase/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Ethylenediamines/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred ICR , Morpholines/pharmacology , N-Methylaspartate/pharmacology , N-Methylaspartate/toxicity , Phosphorylation/drug effects , Physical Stimulation/adverse effects , Receptors, sigma/antagonists & inhibitors , Spinal Cord/drug effects , Sigma-1 Receptor
17.
Exp Neurol ; 287(Pt 1): 1-13, 2017 01.
Article in English | MEDLINE | ID: mdl-27776252

ABSTRACT

Although we have recently demonstrated that carrageenan-induced inflammation upregulates the expression of spinal interleukin (IL)-1ß, which inhibits spinal astrocyte activation and results in the delayed development of Mirror-Image Pain (MIP), little is known regarding the mechanisms that underlie how spinal IL-1ß inhibits the astrocyte activation. In this study, we examined the effect of spinal IL-1ß on astrocyte gap junctions (GJ) and the development of MIP. Following unilateral carrageenan (CA) injection, mechanical allodynia (MA) was evaluated at various time points. Immunohistochemistry and Western blot analysis were used to determine changes in the expression of GFAP and connexins (Cx) in the spinal cord dorsal horn. Carrageenan rats showed a delayed onset of contralateral MA, which mimicked the temporal expression pattern of spinal Cx43 (an astrocyte gap junctional protein) and GFAP. Intrathecal administration of an interleukin-1 receptor antagonist (IL-1ra) twice-a-day on post-carrageenan injection days 0 to 3 caused a significant increase in contralateral MA and spinal Cx43 and GFAP expression. In addition, co-administration of IL-1ß with IL-1ra blocked the IL-1ra-induced increase in contralateral MA and the upregulated expression of spinal Cx43 and GFAP. Finally, co-administration of carbenoxolone (CBX; a GJ decoupler) or Gap26 (a specific Cx43 mimetic blocking peptide) with IL-1ra significantly blocked the IL-1ra-induced early development of contralateral MA and the associated upregulation of spinal Cx43 and GFAP expression. These results demonstrate that spinal IL-1ß suppresses Cx43 expression and astrocyte activation during the early phase of CA-induced inflammation resulting in the delayed onset of contralateral MA. These findings imply that spinal IL-1ß can inhibit astrocyte activation and regulate the time of induction of contralateral MA through modulation of spinal Cx43 expression.

18.
Neuropharmacology ; 111: 34-46, 2016 12.
Article in English | MEDLINE | ID: mdl-27567941

ABSTRACT

We have previously shown using a spinal cord injury (SCI) model that gap junctions contribute to the early spread of astrocyte activation in the lumbar spinal cord and that this astrocyte communication plays critical role in the induction of central neuropathic pain. Sigma-1 receptors (Sig-1Rs) have been implicated in spinal astrocyte activation and the development of peripheral neuropathic pain, yet their contribution to central neuropathic pain remains unknown. Thus, we investigated whether SCI upregulates spinal Sig-1Rs, which in turn increase the expression of the astrocytic gap junction protein, connexin 43 (Cx43) leading to the induction of central neuropathic pain. A thoracic spinal cord hemisection significantly increased both astrocyte activation and Cx43 expression in lumbar dorsal horn. Sig-1Rs were also increased in lumbar dorsal horn astrocytes, but not neurons or microglia. Intrathecal injection of an astrocyte metabolic inhibitor (fluorocitrate); a gap junction/hemichannel blocker (carbenoxolone); or a Cx43 mimetic peptide (43Gap26) significantly reduced SCI-induced bilateral below-level mechanical allodynia. Blockade of Sig-1Rs with BD1047 during the induction phase of pain significantly suppressed the SCI-induced development of mechanical allodynia, astrocyte activation, increased expression of Cx43 in both total and membrane levels, and increased association of Cx43 with Sig-1R. However, SCI did not change the expression of oligodendrocyte (Cx32) or neuronal (Cx36) gap junction proteins. These findings demonstrate that SCI activates astrocyte Sig-1Rs leading to increases in the expression of the gap junction protein, Cx43 and astrocyte activation in the lumbar dorsal horn, and ultimately contribute to the induction of bilateral below-level mechanical allodynia.


Subject(s)
Astrocytes/metabolism , Connexin 43/metabolism , Hyperalgesia/metabolism , Neuralgia/metabolism , Receptors, sigma/metabolism , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Injuries/complications , Animals , Connexin 43/physiology , Disease Models, Animal , Ethylenediamines/administration & dosage , Hyperalgesia/complications , Male , Mice , Mice, Inbred ICR , Neuralgia/complications , Receptors, sigma/antagonists & inhibitors , Sigma-1 Receptor
19.
Brain Res Bull ; 121: 227-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26876754

ABSTRACT

The role of peripheral neurosteroids and their related mechanisms on nociception have not been thoroughly investigated. Based on emerging evidence in the literature indicating that neurosteroids and their main target receptors, i.e., sigma-1, GABAA and NMDA, affect P2X-induced changes in neuronal activity, this study was designed to investigate the effect of peripherally injected dehydroepiandrosterone sulphate (DHEAS) and pregnenolone sulfate (PREGS) on P2X receptor-mediated mechanical allodynia in rats. Intraplantar injection of either neurosteroids alone did not produced any detectable changes in paw withdrawal frequency to the innocuous mechanical stimulation in naïve rats. However, When DHEAS or PREGS were co-injected with a sub-effective dose of αßmeATP, mechanical allodynia was developed and this was dose dependently blocked by pre-injection of the P2X antagonist, TNP-ATP. These results demonstrates that DHEAS and PREGS potentiate the activity of P2X receptors which results in the enhancement of αßmeATP-induced mechanical allodynia. In order to investigate the potential role of peripheral sigma-1, GABAA and NMDA receptors in this facilitatory action, we pretreated animals with BD-1047 (a sigma-1 antagonist), muscimol (a GABAA agonist) or MK-801 (a NMDA antagonist) prior to DHEAS or PREGS+αßmeATP injection. Only BD-1047 effectively prevented the facilitatory effects induced by neurosteroids on αßmeATP-induced mechanical allodynia. Collectively, we have shown that peripheral neurosteroids potentiate P2X-induced mechanical allodynia and that this action is mediated by sigma-1, but not by GABAA nor NMDA, receptors.


Subject(s)
Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Receptors, Purinergic P2X/metabolism , Receptors, sigma/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Dehydroepiandrosterone Sulfate/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Ethylenediamines/pharmacology , Hyperalgesia/drug therapy , Male , Pain Measurement , Pregnenolone/toxicity , Purinergic P2X Receptor Agonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, sigma/antagonists & inhibitors , Time Factors
20.
Pain ; 156(6): 1046-1059, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25749305

ABSTRACT

Damage on one side of the body can also result in pain on the contralateral unaffected side, called mirror-image pain (MIP). Currently, the mechanisms responsible for the development of MIP are unknown. In this study, we investigated the involvement of spinal microglia and interleukin-1ß (IL-1ß) in the development of MIP using a peripheral inflammatory pain model. After unilateral carrageenan injection, mechanical allodynia (MA) in both hind paws and the expression levels of spinal Iba-1, IL-1ß, and GFAP were evaluated. Ipsilateral MA was induced beginning at 3 hours after carrageenan injection, whereas contralateral MA showed a delayed onset occurring 5 days after injection. A single intrathecal (i.t.) injection of minocycline, a tetracycline derivative that displays selective inhibition of microglial activation, or an interleukin-1 receptor antagonist (IL-1ra) on the day of carrageenan injection caused an early temporary induction of contralateral MA, whereas repeated i.t. treatment with these drugs from days 0 to 3 resulted in a long-lasting contralateral MA, which was evident in its advanced development. We further showed that IL-1ß was localized to microglia and that minocycline inhibited the carrageenan-induced increases in spinal Iba-1 and IL-1ß expression. Conversely, minocycline or IL-1ra pretreatment increased GFAP expression as compared with that of control rats. However, i.t. pretreatment with fluorocitrate, an astrocyte inhibitor, restored minocycline- or IL-1ra-induced contralateral MA. These results suggest that spinal IL-1ß derived from activated microglia temporarily suppresses astrocyte activation, which can ultimately prevent the development of contralateral MA under inflammatory conditions. These findings imply that microglial IL-1ß plays an important role in regulating the induction of inflammatory MIP.


Subject(s)
Astrocytes/physiology , Functional Laterality/physiology , Hyperalgesia/prevention & control , Interleukin-1beta/metabolism , Microglia/metabolism , Spinal Cord Dorsal Horn/cytology , Animals , Astrocytes/metabolism , Carrageenan/toxicity , Citrates/pharmacology , Disease Models, Animal , Gene Expression Regulation/drug effects , Hyperalgesia/etiology , Inflammation/chemically induced , Inflammation/complications , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Male , Microglia/drug effects , Minocycline/therapeutic use , Nerve Tissue Proteins/metabolism , Pain/drug therapy , Pain/etiology , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1 Type I/metabolism , Spinal Cord/pathology
SELECTION OF CITATIONS
SEARCH DETAIL