Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 89(Pt A): 106962, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33039970

ABSTRACT

BACKGROUND: Neuroinflammation in the spinal cord following acute brachial plexus injury (BPI) remains a vital cause that leads to motor dysfunction and neuropathic pain. In this study, we aim to explore the role of long non-coding RNA JHDM1D antisense 1 (JHDM1D-AS1) in mediating BPI-induced neuroinflammation and neuronal injury. METHODS: A total brachial plexus root avulsion (tBPRA) model in adult rats and IL-1ß-treated motor neuron-like NSC-34 cells and LPS-treated microglia cell line BV2 were conducted for in vivo and in vitro experiments, respectively. The expressions of JHDM1D-AS1, miR-101-3p and DUSP1, p38, NF-κB, TNF-α, IL-1ß, and IL-6 were detected by RT-PCR and western blot seven days after tBPI. Immunohistochemistry (IHC) was used to detect neuronal apoptosis. CCK8 assay, Tunel assay and LDH kit were used for the detection of neuronal injury. The targeted relationships between JHDM1D-AS1 and miR-101-3p, miR-101-3p and DUSP1 were verified by RNA immunoprecipitation (RIP) and dual-luciferase reporter gene assay. RESULTS: We found significant downregulated expression of JHDM1D-AS1 and DUSP1 but upregulated expression of miR-101-3p in the spinal cord after tBPI. Overexpression of JHDM1D-AS1 had a prominent neuroprotective effect by suppressing neuronal apoptosis and microglial inflammation through reactivation of DUSP1. Further exploration revealed that JHDM1D-AS1 may act as a competitive endogenous RNA targeting miR-101-3p, which bound on the 3'UTR of DUSP1 mRNA. In addition, overexpression of miR-101-3p could reverse the neuroprotective effects of JHDM1D-AS1 upregulation by blocking DUSP1. CONCLUSIONS: JHDM1D-AS1 exerted neuroprotective and anti-inflammatory effects in a rat model of tBPI by regulating miR-101-3p/DUSP1 axis.


Subject(s)
Brachial Plexus Neuropathies/enzymology , MicroRNAs/metabolism , Microglia/enzymology , Motor Neurons/enzymology , Myelitis/enzymology , RNA, Long Noncoding/metabolism , Spinal Cord/enzymology , Animals , Apoptosis , Brachial Plexus Neuropathies/genetics , Brachial Plexus Neuropathies/pathology , Brachial Plexus Neuropathies/physiopathology , Cell Line , Disease Models, Animal , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Mice , MicroRNAs/genetics , Microglia/pathology , Motor Neurons/pathology , Myelitis/genetics , Myelitis/pathology , Myelitis/physiopathology , RNA, Long Noncoding/genetics , Rats , Signal Transduction , Spinal Cord/pathology , Spinal Cord/physiopathology , Up-Regulation
2.
J Comp Neurol ; 525(16): 3414-3428, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28649695

ABSTRACT

In both acute and chronic pain conditions, women tend to be more sensitive than men. This sex difference may be regulated by estrogens, such as estradiol, that are synthesized in the spinal cord and brainstem and act locally to influence pain processing. To identify a potential cellular source of local estrogen, here we examined the expression of aromatase, the enzyme that catalyzes the conversion of testosterone to estradiol. Our studies focused on primary afferent neurons and on their central targets in the spinal cord and medulla as well as in the nucleus of the solitary tract, the target of nodose ganglion-derived visceral afferents. Immunohistochemical staining in an aromatase reporter mouse revealed that many neurons in laminae I and V of the spinal cord dorsal horn and caudal spinal trigeminal nucleus and in the nucleus of the solitary tract express aromatase. The great majority of these cells also express inhibitory interneuron markers. We did not find sex differences in aromatase expression and neither the pattern nor the number of neurons changed in a sciatic nerve transection model of neuropathic pain or in the Complete Freund's adjuvant model of inflammatory pain. A few aromatase neurons express Fos after cheek injection of capsaicin, formalin, or chloroquine. In total, given their location, these aromatase neurons are poised to engage nociceptive circuits, whether it is through local estrogen synthesis or inhibitory neurotransmitter release.


Subject(s)
Aromatase/genetics , Aromatase/metabolism , Gene Expression Regulation , Medulla Oblongata/cytology , Neurons/enzymology , Sciatica/enzymology , Spinal Cord Dorsal Horn/cytology , Afferent Pathways/physiology , Animals , Disease Models, Animal , Freund's Adjuvant/toxicity , Mice , Mice, Transgenic , Myelitis/chemically induced , Myelitis/enzymology , Nerve Tissue Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Stilbamidines/metabolism , TRPV Cation Channels/metabolism
3.
Neurosci Res ; 71(3): 210-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21802455

ABSTRACT

In our previous study, peripheral inflammatory stimulation evoked production of macrophage migration inhibitory factor (MIF) in the spinal cord and found spinal microglia are the major source of MIF in this context. Given the contribution of the activated-microglia to the inflammatory neuropathy plus the role for upregulated COX 2 expression and PGE(2) production in the severity of clinical manifestations of these neuroinflammatory conditions, we herein tested the hypothesis that in vitro MIF stimulation to spinal microglia could result in an activation of COX 2-PGE(2) system by MIF-CD74 interaction. We found MIF played roles in evoking COX 2 mRNA and protein expression in a dose-dependent manner correspondingly in changes in PGE(2) level in the cultured rat microglia, but these changes could be inhibited by genetic deletion of CD74. Finally, MIF-induced COX 2-PGE(2) activation could be blocked by selective inhibitors of p44/p42 and p38 MAPKs. These data highlight MIF/CD74 interaction induces upregulation of COX 2 expression and PGE(2) secretion in primary rodent microglia, and further this effect is associated with downstream activation of p38 and p44/p42 signaling cascades, and favors the role of MIF as a novel pathway for microglia-associated neuroinflammation.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gliosis/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Microglia/metabolism , Myelitis/metabolism , Animals , Animals, Newborn , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/physiology , Cyclooxygenase 2/genetics , Dinoprostone/physiology , Enzyme Activation/genetics , Gliosis/enzymology , Gliosis/pathology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/physiology , Microglia/enzymology , Microglia/pathology , Myelitis/enzymology , Myelitis/pathology , Neural Pathways/pathology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Up-Regulation/genetics
4.
J Neurotrauma ; 28(6): 1035-49, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21355819

ABSTRACT

We tested the hypothesis that a selective phosphodiesterase type 4 inhibitor (PDE4-I; IC486051) would attenuate early inflammatory and oxidative processes following spinal cord injury (SCI) when delivered during the first 3 days after injury. Rats receiving a moderately severe thoracic-clip-compression SCI were treated with the PDE4-I (0.5, 1.0, and 3.0 mg/kg IV) in bolus doses from 2-60 h post-injury. Doses at 0.5 mg/kg and 1.0 mg/kg significantly decreased myeloperoxidase (MPO) enzymatic activity (neutrophils), expression of a neutrophil-associated protein and of ED-1 (macrophages), and estimates of lipid peroxidation in cord lesion homogenates at 24 h and 72 h post-injury by 25-40%. The 3.0 mg/kg dose had small or no effects on these measures. The PDE4-I treatment (0.5 or 1.0 mg/kg) reduced expression of the oxidative enzymes gp91(phox), inducible nitric oxide synthase, and cyclooxygenase-2, and diminished free radical generation by up to 40%. Treatment with 0.5 mg/kg PDE4-I improved motor function (as assessed by the Basso-Beattie-Bresnahan scale) significantly from 4-8 weeks after SCI (average difference 1.3 points). Mechanical allodynia elicited from the hindpaw decreased by up to 25%. The PDE4-I treatment also increased white matter volume near the lesion at 8 weeks after SCI. In conclusion, the PDE4-I reduced key markers of oxidative stress and leukocyte infiltration, producing cellular protection, locomotor improvements, and a reduction in neuropathic pain. Early inhibition of PDE4 is neuroprotective after SCI when given acutely and briefly at sufficient doses.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Nerve Degeneration/prevention & control , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Chemotaxis, Leukocyte/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Female , Male , Myelitis/drug therapy , Myelitis/enzymology , Myelitis/pathology , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Oxidative Stress/physiology , Phosphodiesterase 4 Inhibitors/therapeutic use , Rats , Rats, Wistar , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology
5.
J Biol Chem ; 286(3): 2331-42, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21075851

ABSTRACT

A major immunological response during neuroinflammation is the activation of microglia, which subsequently release proinflammatory mediators such as prostaglandin E(2) (PGE(2)). Besides its proinflammatory properties, cyclooxygenase-2 (COX-2)-derived PGE(2) has been shown to exhibit anti-inflammatory effects on innate immune responses. Here, we investigated the role of microsomal PGE(2) synthase-1 (mPGES-1), which is functionally coupled to COX-2, in immune responses using a model of lipopolysaccharide (LPS)-induced spinal neuroinflammation. Interestingly, we found that activation of E-prostanoid (EP)2 and EP4 receptors, but not EP1, EP3, PGI(2) receptor (IP), thromboxane A(2) receptor (TP), PGD(2) receptor (DP), and PGF(2) receptor (FP), efficiently blocked LPS-induced tumor necrosis factor α (TNFα) synthesis and COX-2 and mPGES-1 induction as well as prostaglandin synthesis in spinal cultures. In vivo, spinal EP2 receptors were up-regulated in microglia in response to intrathecally injected LPS. Accordingly, LPS priming reduced spinal synthesis of TNFα, interleukin 1ß (IL-1ß), and prostaglandins in response to a second intrathecal LPS injection. Importantly, this reduction was only seen in wild-type but not in mPGES-1-deficient mice. Furthermore, intrathecal application of EP2 and EP4 agonists as well as genetic deletion of EP2 significantly reduced spinal TNFα and IL-1ß synthesis in mPGES-1 knock-out mice after LPS priming. These data suggest that initial inflammation prepares the spinal cord for a negative feedback regulation by mPGES-1-derived PGE(2) followed by EP2 activation, which limits the synthesis of inflammatory mediators during chronic inflammation. Thus, our data suggest a role of mPGES-1-derived PGE(2) in resolution of neuroinflammation.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Microglia/metabolism , Myelitis/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intramolecular Oxidoreductases/genetics , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Myelitis/chemically induced , Myelitis/genetics , Prostaglandin-E Synthases , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandins/genetics , Prostaglandins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
6.
J Neurol Sci ; 294(1-2): 29-37, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20488464

ABSTRACT

The 70-kDa family of heat shock proteins (HSP70), in particular, plays a vital role in cellular protection and has been detected in various tissues subject to stress. HSPA12B is the newest member of the HSP70 family but is distinct from the HSP70 family. In this study, we elucidated the dynamic expression changes and localization of HSPA12B in lipopolysaccharide (LPS)-induced neuroinflammatory processes in adult rats. HSPA12B expression was strongly induced in active microglial cells in inflamed spinal cord. In vitro studies indicated that the up-regulation of HSPA12B may be involved in the subsequent microglia activation following LPS challenge. The elevated HSPA12B expression was regulated by activation of MAPK-p38 and ERK1/2 pathways, less contribution of the SAPK/JNK pathway in microglial cells. Collectively, these results suggested HSPA12B may be important for host defense in microglia-mediated immune response. Understanding the cell signal pathway may provide a novel strategy against inflammatory and immune reaction in neuroinflammtion in CNS.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , MAP Kinase Signaling System , Microglia/metabolism , Myelitis/metabolism , Spinal Cord/metabolism , Animals , Cell Line , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/immunology , Lumbar Vertebrae , Male , Microglia/enzymology , Microglia/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myelitis/enzymology , Rats , Rats, Sprague-Dawley , Spinal Cord/enzymology , Spinal Cord/immunology , Time Factors , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Anesthesiology ; 110(6): 1379-89, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19417613

ABSTRACT

BACKGROUND: This study explores the underlying mechanism of the antiinflammatory effect of amitriptyline in chronic morphine-infused rats. METHODS: Male Wistar rats were implanted with two intrathecal catheters. One catheter was for the continuous infusion of saline, amitriptyline (15 microg/h), morphine (15 microg/h), p38 mitogen-activated protein kinase inhibitor SB203580 (0.5 microg/h), morphine plus amitriptyline, or morphine plus amitriptyline plus SB203580 for 5 days. The other catheter was used for daily intrathecal injection of anti-interleukin-10 (IL-10) antibody or heme oxygenase-1 inhibitor zinc protoporphyrin for 5 days. RESULTS: Amitriptyline/morphine coinfusion upregulated IL-10 protein expression in microglia; this was not observed in morphine-infused rats. Anti-IL-10 antibody effectively neutralized the amitriptyline-induced IL-10 expression in chronic morphine-infused rats. In addition, coinfusion of amitriptyline restored the antinociceptive effect of morphine (a 4.8-fold right-shift of the morphine dose-response curve compared to a 77.8-fold right-shift in its absence), and the injection of anti-IL-10 antibody or coinfusion of SB203580 partially reversed the effect of amitriptyline on the antinociceptive effect of morphine in morphine-infused rats (a 17.9-fold and 15.1-fold right-shift in morphine dose-response curves). Anti-IL-10 antibody and SB203580 significantly inhibited the amitriptyline-induced p38 mitogen-activated protein kinase and heme oxygenase-1 expression and the associated antiinflammatory effect of amitriptyline. Daily injection of zinc protoporphyrin also demonstrated that it reverses the effect of amitriptyline in morphine's antinociception and antiinflammation in chronic morphine-infused rats. CONCLUSIONS: These results suggest that the antiinflammatory effect of amitriptyline on morphine tolerance, probably acting by increasing IL-10 expression, is mediated by p38 mitogen-activated protein kinase heme oxygenase-1 signal transduction cascade.


Subject(s)
Amitriptyline/pharmacology , Analgesics, Opioid/pharmacology , Anti-Inflammatory Agents, Non-Steroidal , Antidepressive Agents, Tricyclic/pharmacology , Heme Oxygenase-1/physiology , Interleukin-10/physiology , Morphine/pharmacology , Myelitis/enzymology , Myelitis/physiopathology , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies, Blocking/pharmacology , Drug Tolerance , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Imidazoles/pharmacology , Interleukin-10/antagonists & inhibitors , Male , Pain Measurement/drug effects , Protoporphyrins/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Spinal Cord/pathology
8.
Brain Res ; 1271: 114-20, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19332038

ABSTRACT

Ca(2+)/calmodulin-dependent protein kinase II alpha (CaMKIIalpha) is abundant in the central nervous system, where it plays important roles in regulating neuronal plasticity and survival. However, the role of CaMKIIalpha activation in traumatically injured spinal cords remains unclear. This study examined the effects of clip compression injury on levels of phosphorylated CaMKIIalpha (pCaMKIIalpha) and its cellular localization in rat spinal cords. Western blot analysis showed that the pCaMKIIalpha levels in both rostral (days 7, 14, and 21 post-injury) and caudal (days 4, 7, 14, and 21 post-injury) areas of the injury site were more than twice the levels in the non-injured controls. Immunohistochemical examination revealed constitutive localization of pCaMKIIalpha in the superficial lamina of the dorsal horn and neurons in normal spinal cord controls. After spinal cord injury, levels of the same components were markedly increased in both rostral and caudal regions approximately 3 mm from the center of the spinal cord lesions. However, pCaMKIIalpha was very rare in inflammatory cells in the injured spinal cords. In this animal model, CaMKIIalpha may play an important role in the spontaneous reversal of spinal cord dysfunction, thus restoring locomotor activity, possibly by functioning in the reconstruction of synaptic transmission and in protecting neurons from spinal cord injury.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Enzyme Activation/physiology , Spinal Cord Compression/enzymology , Spinal Cord/enzymology , Up-Regulation/physiology , Animals , Cell Survival/physiology , Cytoprotection/physiology , Disease Models, Animal , Female , Immunohistochemistry , Motor Activity/physiology , Myelitis/enzymology , Myelitis/pathology , Myelitis/physiopathology , Neurons/enzymology , Neurons/pathology , Neurosurgical Procedures/instrumentation , Neurosurgical Procedures/methods , Phosphorylation , Posterior Horn Cells/enzymology , Posterior Horn Cells/pathology , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Compression/pathology , Spinal Cord Compression/physiopathology , Surgical Instruments , Synaptic Transmission/physiology
9.
Glycoconj J ; 26(1): 19-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18677561

ABSTRACT

Beta1,4-galactosyltransferase-I (beta1,4-GalT-I) is one of the best studied glycosyltransferases. Previous studies demonstrated that beta1,4-GalT-I was a major galactosyltransferase responsible for selectin-ligand biosynthesis and that inflammatory responses of beta1,4-GalT-I deficient mice were impaired. In this study, we investigate the expression of beta1,4-GalT-I in lipopolysaccharide (LPS)-induced neuroinflammatory processes. The results of this study demonstrated that beta1,4-GalT-I was strongly induced by intraspinal administration of LPS. More than 90% galactose-containing glycans and beta1,4-GalT-I were expressed in immune cells. The ELISA assay shows focal injection LPS also induces TNF-alpha alteration. Double staining indicated beta1,4-GalT-I overlapped with TNF-alpha. Moreover, RT-PCR for beta1,4-GalT-I mRNA showed that beta1,4-GalT-I mRNA in microglia in vitro was affected in a dose- and time dependent manner in response to LPS or TNF-alpha stimulation. All these results indicated that the increase of beta1,4-GalT-I might attribute to the effect of TNF-alpha excreting during inflammation. E-selectin, which ligand was modified by beta1,4-GalT-I, was correlated with galactose-containing glycans following injecting LPS into spinal cord. We therefore suggest that beta1,4-GalT-I may play an important role in regulating immune cell migration into the inflammatory site.


Subject(s)
Galactosyltransferases/biosynthesis , Lipopolysaccharides/toxicity , Myelitis/enzymology , Animals , Cell Movement/drug effects , Cell Movement/immunology , E-Selectin/immunology , E-Selectin/metabolism , Enzyme Induction/drug effects , Enzyme Induction/immunology , Galactosyltransferases/immunology , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Male , Mice , Myelitis/chemically induced , Myelitis/immunology , Myelitis/pathology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
10.
Brain Res ; 1145: 19-27, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17331478

ABSTRACT

Protein kinase D (PKD), a newly described serine/threonine kinase, has been implicated in many signal transduction pathways. The present study was designed to determine whether and how PKD is activated in inflammation. The results demonstrated that lipopolysaccharide (LPS, 30 microg/ml) stimulated PKD and protein kinase C (PKC) phosphorylation in spinal neurons within 0.5 h, and the activation reached a maximum at 3 or 8 h and declined at 12 h. The phosphorylation could be inhibited by the selective inhibitors for PKC (100 nM), mainly for PKCalpha and PKCbeta, suggesting the involvement of the PKC pathway. Particularly, PKCalpha might be critical for LPS-induced PKD activation since the PKCbeta inhibitor (100 nM) observed no effect on the phosphorylation of PKD. Furthermore, the expression of interleukin-1beta (IL-1beta) was significantly induced by LPS within 0.5 h, and reached a maximum at 8 h. IL-1 receptor antagonist inhibited PKD and PKCs activation induced by LPS at a concentration of 50 nM and achieved maximum at 1000 nM. These results demonstrated for the first time that PKD could be activated by LPS in spinal neurons, might via the IL-1beta/PKCalpha pathway. Additionally, immunostaining showed an increase in number of phosphorylated PKD-immunoreactive cells of adult spinal dorsal horn induced by intraplantar injected carrageenan (2 microg/100 microl), and antisense oligodeoxynucleotide to IL-1 receptor type I (50 microg/10 microl, intrathecal injected) inhibited the PKD activation, suggesting an involvement of IL-1beta/PKD pathway in inflammation in adult spinal cord.


Subject(s)
Interleukin-1beta/metabolism , Myelitis/enzymology , Protein Kinase C/metabolism , Spinal Cord/enzymology , Animals , Carrageenan/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Inflammation Mediators/pharmacology , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Male , Myelitis/chemically induced , Myelitis/immunology , Neurons/drug effects , Neurons/enzymology , Neurons/immunology , Oligonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/immunology , Protein Kinase C-alpha/drug effects , Protein Kinase C-alpha/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/drug effects , Receptors, Interleukin-1/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/drug effects , Spinal Cord/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
11.
J Neuroimmunol ; 111(1-2): 122-30, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11063829

ABSTRACT

Acute inflammation following spinal cord injury results in secondary injury and pathological reorganisation of the central nervous system (CNS) architecture. Cyclooxygenases (Prostaglandin Endoperoxide H Synthases, PGH) are key enzymes in the conversion of arachidonic acid into prostanoids which mediate immunomodulation, mitogenesis, apoptosis, blood flow, secondary injury (lipid peroxygenation) and inflammation. Here, we report cyclooxygenase-1 (COX-1) expression following spinal cord injury. In control spinal cords, COX-1 expression was localized by immunohistochemistry to ependymal cells, some neurons, inclusive dorsal and ventral root ganglion cells, few endothelial cells but rarely to brain microglia/macrophages. In injured spinal cords, COX-1(+) microglia/macrophages accumulated highly significantly (P<0.0001) at peri-lesional areas and in the developing necrotic core early after injury. Here numbers of COX-1(+) cells remained persistently elevated up to 4 weeks following injury. Further, COX-1(+) cells were located in perivascular Virchow-Robin spaces, between spared axons and in areas of Wallerian degeneration. Double labeling experiments confirmed co-expression of COX-1 by ED-1(+) and OX-42(+) microglia/macrophages. Transiently after infarction most COX-1(+) microglia/macrophages coexpress the activation antigen OX-6 (MHC class II). However, the prolonged accumulation of COX-1(+) microglia/macrophages at the lesion site enduring the acute post injury inflammatory response points to a role of COX-1 in tissue remodeling or secondary injury. We have identified and localized persistent accumulation of COX-1 expressing cells which might be a potential pharmacological target following spinal cord injury. Therefore, we suggest that approaches based on: (i) short-term; and (ii) selective COX-2 blocking alone might not be a sufficient tool to suppress the local synthesis of prostanoids.


Subject(s)
Isoenzymes/metabolism , Macrophages/enzymology , Microglia/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Spinal Cord Injuries/metabolism , Animals , Cyclooxygenase 1 , Endothelium/cytology , Endothelium/metabolism , Male , Membrane Proteins , Myelitis/enzymology , Myelitis/immunology , Myelitis/pathology , Prostaglandins/metabolism , Rats , Rats, Inbred Lew , Spinal Cord/enzymology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
12.
Neuroscience ; 95(2): 525-33, 2000.
Article in English | MEDLINE | ID: mdl-10658633

ABSTRACT

Several lines of evidence have shown a role for the nitric oxide/cyclic guanosine monophosphate signaling pathway in the development of spinal hyperalgesia. However, the roles of effectors for cyclic guanosine monophosphate are not fully understood in the processing of pain in the spinal cord. The present study showed that cyclic guanosine monophosphate-dependent protein kinase Ialpha but not Ibeta was localized in the neuronal bodies and processes, and was distributed primarily in the superficial laminae of the spinal cord. Intrathecal administration of a selective inhibitor of cyclic guanosine monophosphate-dependent protein kinase Ialpha, Rp-8-[(4-chlorophenyl)thio]-cGMPS triethylamine, produced a significant antinociception demonstrated by the decrease in the number of flinches and shakes in the formalin test. This was accompanied by a marked reduction in formalin-induced c-fos expression in the spinal dorsal horn. Moreover, cyclic guanosine monophosphate-dependent protein kinase Ialpha protein expression was dramatically increased in the lumbar spinal cord 96 h after injection of formalin into a hindpaw, which occurred mainly in the superficial laminae on the ipsilateral side of a formalin-injected hindpaw. This up-regulation of cyclic guanosine monophosphate-dependent protein kinase Ialpha expression was completely blocked not only by a neuronal nitric oxide synthase inhibitor, 7-nitroindazole, and a soluble guanylate cyclase inhibitor, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, but also by an N-methyl-D-aspartate receptor antagonist, dizocilpine maleate (MK-801). The present results indicate that noxious stimulation not only initially activates but also later up-regulates cyclic guanosine monophosphate-dependent protein kinase Ialpha expression in the superficial laminae via an N-methyl-D-aspartate-nitric oxide-cyclic guanosine monophosphate signaling pathway, suggesting that cyclic guanosine monophosphate-dependent protein kinase Ialpha may play an important role in the central mechanism of formalin-induced inflammatory hyperalgesia in the spinal cord.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Hyperalgesia/enzymology , Myelitis/physiopathology , Spinal Cord/enzymology , Spinal Cord/physiopathology , Animals , Antibodies , Behavior, Animal/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/analysis , Cyclic GMP-Dependent Protein Kinases/immunology , Dizocilpine Maleate/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Formaldehyde , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/immunology , Male , Myelitis/enzymology , N-Methylaspartate/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nociceptors/drug effects , Nociceptors/physiology , Pain/chemically induced , Pain/physiopathology , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley , Spinal Cord/immunology , Thionucleotides/pharmacology
13.
Brain ; 116 ( Pt 6): 1425-50, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8293279

ABSTRACT

Quinolinic acid (QUIN), kynurenic acid (KYNA) and L-kynurenine (L-KYN) are neuroactive kynurenine pathway metabolites that accumulate in inflammatory neurological diseases. These increases were attributed to the induction of indoleamine-2,3-dioxygenase (IDO), the enzyme that converts L-tryptophan into L-KYN. Direct conversion of L-tryptophan into QUIN by brain tissue occurs in conditions of CNS inflammation, but not by normal brain tissue. To investigate whether increased activity of enzymes distal to IDO may determine L-KYN conversion to QUIN, rhesus macaques were inoculated with poliovirus directly into the spinal cord, as a model of focal inflammatory neurological disease (FASEB J. 6, 2977-2989, 1992). Induction of spinal cord IDO (35.9-fold) accompanied smaller, but proportional increases in kynurenine-3-hydroxylase (2.4-fold) and kynureninase (2.3-fold) activities, which were correlated to CSF and tissue QUIN levels, as well as to measures of inflammatory lesions. 3-Hydroxyanthranilate-3,4-dioxygenase activity was unchanged. Cerebrospinal fluid KYNA levels increased in proportion to both IDO activity and L-KYN accumulation, though kynurenine aminotransferase activity was unaffected. Cerebrospinal fluid neopterin, a marker of macrophage and immune activation, accumulated in proportion to the responsive enzymes and metabolites. The cell types involved in producing QUIN were investigated in vitro. Human foetal brain cultures consisting of astrocytes and neurons converted large quantities of [13C6]L-tryptophan into L-KYN when stimulated by gamma-interferon, but very little [13C6]QUIN was formed unless macrophages (THP-1 cells) were first added to the cultures (to model a key component of brain inflammation). [13C6]L-Tryptophan was converted into [13C6]QUIN by either gamma-interferon stimulated macrophages, or following intracisternal administration into poliovirus-infected macaques. Inhibitors of the kynurenine pathway, 6-chlorotryptophan and 4-chloro-3-hydroxyanthranilic acid, attenuated [13C6]QUIN formation by macrophages, and when co-infused with [13C6]L-tryptophan into poliovirus-infected macaques. These results suggest roles for increased activities of IDO, kynurenine-3-hydroxylase and kynureninase in accelerating the synthesis of QUIN, L-KYN and KYNA in conditions of brain inflammation. Macrophage infiltrates, and perhaps microglia, are important sources of QUIN, whereas constitutive brain cells and macrophages are sources of L-KYN. Drugs that inhibit kynurenine pathway enzymes attenuate QUIN formation in the CNS, and provide tools to examine the consequences of reduced QUIN accumulation.


Subject(s)
Encephalitis/metabolism , Kynurenic Acid/metabolism , Kynurenine/metabolism , Myelitis/metabolism , Quinolinic Acid/metabolism , 3-Hydroxyanthranilic Acid/analogs & derivatives , 3-Hydroxyanthranilic Acid/pharmacology , Animals , Biopterins/analogs & derivatives , Biopterins/analysis , Brain/metabolism , Encephalitis/enzymology , Fetus/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenic Acid/analysis , Kynurenine/analysis , Macaca mulatta , Myelitis/enzymology , Neopterin , Poliomyelitis/enzymology , Poliomyelitis/metabolism , Quinolinic Acid/analysis , Tryptophan/analogs & derivatives , Tryptophan/metabolism , Tryptophan/pharmacology , Tryptophan Oxygenase/metabolism
15.
Pol Tyg Lek ; 45(27-28): 553-6, 1990.
Article in Polish | MEDLINE | ID: mdl-2287577

ABSTRACT

The study aimed at assessing the value lysozyme assay in CFS as indicator of the damage to blood-cerebrospinal fluid barrier and intensification of inflammatory process in the course of meningitis. The study involved 20 patients with suppurative and 66 with viral meningitis. Control group included 26 patients without nervous system disease. To estimate the degree of blood-cerebrospinal fluid barrier damage albumin and lysozyme indicators were calculated. It was proved, that CSF lysozyme levels are bigger in the suppurative meningitis than in viral meningitis. According to the author, CSF lysozyme levels the value of lysozyme indicator may inform on intensification of the inflammatory process and the degree of blood-cerebrospinal fluid barrier damage in suppurative meningitis, whereas in the viral meningitis they inform on degree of blood-cerebrospinal fluid barrier damage, only.


Subject(s)
Meningitis/enzymology , Muramidase/cerebrospinal fluid , Myelitis/enzymology , Adolescent , Adult , Child , Child, Preschool , Humans , Meningitis, Viral/enzymology , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...