Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Neurochem ; 152(1): 72-91, 2020 01.
Article in English | MEDLINE | ID: mdl-31563141

ABSTRACT

Peripheral nerve injury elicits spinal microgliosis, contributing to neuropathic pain. The aurora kinases A (AURKA), B (AURKB), and C (AURKC) are potential therapeutic targets in proliferating cells. However, their role has not been clarified in microglia. The aim of this study was to examine the regulation of aurora kinases and their roles and druggability in spinal microgliosis and neuropathic pain. Sprague-Dawley rats received chronic constriction injury (CCI). Gene expression of aurora kinases A-C was evaluated by quantitative RT-PCR and western blot, respectively, in spinal cords at 1, 3, 7, and 14 days after CCI. AURKB gene and protein expression was up-regulated concomitantly with the development of spinal microgliosis and neuropathic pain. Using lentiviral over-expression and adeno-associated viral knockdown approaches, the function of AURKB was further investigated by western blot, immunohistochemistry, RNA sequencing, and pain behavior tests. We found that AURKB over-expression in naive rats caused spinal microgliosis and pain hypersensitivity, whereas AURKB knockdown reduced microgliosis and alleviated CCI-induced neuropathic pain. Accordingly, RNA sequencing data revealed down-regulation of genes critically involved in signaling pathways associated with spinal microgliosis and neuropathic pain after AURKB knockdown in CCI rats. To examine its therapeutic potential for treatment of neuropathic pain, animals were treated intrathecally with the pharmacological AURKB inhibitor AZD1152-HQPA resulting in the alleviation of CCI-induced pain. Taken together, our findings indicated that AURKB plays a critical role in spinal microgliosis and neuropathic pain. Targeting AURKB may be an efficient method for treatment of neuropathic pain subsequent to peripheral nerve injury.


Subject(s)
Aurora Kinase B/antagonists & inhibitors , Microglia/physiology , Neuralgia/therapy , Peripheral Nerve Injuries/physiopathology , Animals , Aurora Kinase B/genetics , Aurora Kinase B/physiology , Disease Models, Animal , Down-Regulation , Enzyme Inhibitors/therapeutic use , Gene Expression , Gene Knockdown Techniques , Male , Microglia/enzymology , Microglia/pathology , Neuralgia/enzymology , Peripheral Nerve Injuries/enzymology , Rats , Rats, Sprague-Dawley , Spinal Cord/enzymology , Spinal Cord/pathology
2.
Pharmacol Res ; 159: 105049, 2020 09.
Article in English | MEDLINE | ID: mdl-32598944

ABSTRACT

Brachial plexus axotomy is a common peripheral nerve trauma. Artemisinin, an FDA-approved antimalarial drug, has been described to possess neuroprotective properties. However, the specific mechanisms by which artemisinin protects neurons from axotomy-induced neurotoxicity remain to be elucidated. In this study, we assessed the neuroprotective effects of artemisinin on an experimental animal model of brachial plexus injury and explored the possible mechanisms involved. Artemisinin treatment restored both athletic ability and sensation of the affected upper limb, rescued motoneurons and attenuated the inflammatory response in the ventral horn of the spinal cord. Additionally, artemisinin inhibited the molecular signals of apoptosis, activated signaling pathways related to cell survival and induced NSCPs differentiation into NeuN-positive neurons. Further validation of the involved key signaling molecules, using an in vitro model of hydrogen peroxide-induced neurotoxicity, revealed that both the inhibition of PKA signaling pathway or the silencing of Akt reversed the neuroprotective action of artemisinin on motoneurons. Our results indicate that artemisinin provides neuroprotection against axotomy and hydrogen peroxide-induced neurotoxicity, an effect that might be mediated by the PKA-Akt signaling pathway.


Subject(s)
Apoptosis/drug effects , Artemisinins/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Motor Neurons/drug effects , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Peripheral Nerve Injuries/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord/drug effects , Animals , Axotomy , Behavior, Animal/drug effects , Brachial Plexus/surgery , Cells, Cultured , Disease Models, Animal , Mice, Inbred C57BL , Motor Neurons/enzymology , Motor Neurons/pathology , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Phosphorylation , Recovery of Function , Signal Transduction , Spinal Cord/enzymology , Spinal Cord/pathology , Spinal Cord/physiopathology
3.
J Neurochem ; 141(1): 37-47, 2017 04.
Article in English | MEDLINE | ID: mdl-27973735

ABSTRACT

Myelination in the peripheral nervous system (PNS) is controlled by both positive and negative regulators within Schwann cells to ensure timely onset and correct myelin thickness for saltatory conduction by neurons. Transcription factors such as Sox10, octamer-binding transcription factor 6 (Oct6) and Krox20 form a positive regulatory network, whereas negative regulators such as cJun and Sox2 oppose myelination in Schwann cells. The role of the p38 MAPK pathway has been studied in PNS myelination, but its precise function remains unclear, with both positive and negative effects of p38 activity reported upon both myelination and processes of nerve repair. To clarify the role of p38 MAPK in the PNS, we have analysed mice with a Schwann cell-specific ablation of the major p38 isoform, p38alpha. In line with previous findings of an inhibitory role for p38 MAPK, we observe acceleration of post-natal myelination in p38alpha null nerves, a delay in myelin down-regulation following injury, together with a small increase in levels of re-myelination following injury. Finally we explored roles for p38alpha in controlling axonal regeneration and functional repair following PNS injury and observe that loss of p38alpha function in Schwann cells does not appear to affect these processes as previously reported. These studies therefore provide further proof for a role of p38 MAPK signalling in the control of myelination by Schwann cells in the PNS, but do not show an apparent role for signalling by this MAP kinase in Schwann cells controlling other elements of Wallerian degeneration and functional repair following injury. Cover Image for this issue: doi: 10.1111/jnc.13793.


Subject(s)
Mitogen-Activated Protein Kinase 14/physiology , Nerve Fibers, Myelinated/enzymology , Peripheral Nerve Injuries/enzymology , Peripheral Nerves/enzymology , Recovery of Function/physiology , Schwann Cells/enzymology , Animals , Animals, Newborn , Cells, Cultured , Female , Male , Mice , Nerve Fibers, Myelinated/pathology , Peripheral Nerve Injuries/pathology , Peripheral Nerves/pathology , Rats , Schwann Cells/pathology
4.
Brain Behav Immun ; 66: 94-102, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28709913

ABSTRACT

Pain and depression often co-occur, but the underlying mechanisms have not been elucidated. Here, we used the spared nerve injury (SNI) model in mice to induce both neuropathic pain and depression-like behavior. We investigated whether brain interleukin (IL)-1 signaling and activity of kynurenine 3-monoxygenase (KMO), a key enzyme for metabolism of kynurenine into the neurotoxic NMDA receptor agonist quinolinic acid, are necessary for comorbid neuropathic pain and depression-like behavior. SNI mice showed increased expression levels of Il1b and Kmo mRNA in the contralateral side of the brain. The SNI-induced increase of Kmo mRNA was associated with increased KMO protein and elevated quinolinic acid and reduced kynurenic acid in the contralateral hippocampus. The increase in KMO-protein in response to SNI mostly took place in hippocampal NeuN-positive neurons rather than microglia. Inhibition of brain IL-1 signaling by intracerebroventricular administration of IL-1 receptor antagonist after SNI prevented the increase in Kmo mRNA and depression-like behavior measured by forced swim test. However, inhibition of brain IL-1 signaling has no effect on mechanical allodynia. In addition, intracerebroventricular administration of the KMO inhibitor Ro 61-8048 abrogated depression-like behavior without affecting mechanical allodynia after SNI. We show for the first time that the development of depression-like behavior in the SNI model requires brain IL-1 signaling and activation of neuronal KMO, while pain is independent of this pathway. Inhibition of KMO may represent a promising target for treating depression.


Subject(s)
Depression/enzymology , Kynurenine 3-Monooxygenase/metabolism , Neuralgia/enzymology , Neurons/enzymology , Animals , Depression/complications , Disease Models, Animal , Hippocampus/enzymology , Hyperalgesia/complications , Hyperalgesia/enzymology , Interleukin-1/metabolism , Kynurenine 3-Monooxygenase/genetics , Male , Mice, Inbred C57BL , Microglia/enzymology , Neuralgia/complications , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/enzymology , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation
5.
J Surg Res ; 200(2): 655-63, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26521098

ABSTRACT

BACKGROUND: CDP-choline (cytidine-5'-diphosphocholine) improves functional recovery, promotes nerve regeneration, and decreases perineural scarring in rat peripheral nerve injury. The aim of the present study was to investigate the mechanism of action of CDP-choline with regard to matrix metalloproteinase (MMP) activity in the rat-transected sciatic nerve injury model. MATERIALS AND METHODS: Male Wistar rats were randomized into Sham, Saline, and CDP-choline groups. Rats in Sham group received Sham surgery, whereas rats in Saline and CDP-choline groups underwent right sciatic nerve transection followed by immediate primary saturation and injected intraperitoneally with 0.9% NaCl (1 mL/kg) and CDP-choline (600 µg/kg), respectively. Sciatic nerve samples were obtained 1, 3, and 7 d after the surgery and analyzed for levels and activities of MMP-2 and MMP-9, levels of tissue inhibitor of metalloproteinases-1 (TIMP-1) and TIMP-3, and axonal regeneration. RESULTS: CDP-choline treatment decreased the levels and activities of MMP-2 and MMP-9, whereas increasing levels of TIMP-1 and TIMP-3 significantly on the third and seventh day after injury compared to Saline group. In addition, CDP-choline administration resulted in new axon formation and formation and advancement of myelination on newly formed islets (compartments) of axonal regrowth. CONCLUSIONS: Our data show, for the first time, that CDP-choline modulates MMP activity and promotes the expression of TIMPs to stimulate axonal regeneration. These data help to explain one mechanism by which CDP-choline provides neuroprotection in peripheral nerve injury.


Subject(s)
Cytidine Diphosphate Choline/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Nerve Regeneration/drug effects , Neuroprotective Agents/pharmacology , Peripheral Nerve Injuries/drug therapy , Sciatic Nerve/injuries , Animals , Biomarkers/metabolism , Blotting, Western , Cytidine Diphosphate Choline/therapeutic use , Injections, Intraperitoneal , Male , Nerve Regeneration/physiology , Neuroprotective Agents/therapeutic use , Peripheral Nerve Injuries/enzymology , Random Allocation , Rats , Rats, Wistar , Sciatic Nerve/drug effects , Sciatic Nerve/enzymology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism
6.
Neural Plast ; 2015: 601767, 2015.
Article in English | MEDLINE | ID: mdl-26457205

ABSTRACT

The insular cortex (IC) is associated with important functions linked with pain and emotions. According to recent reports, neural plasticity in the brain including the IC can be induced by nerve injury and may contribute to chronic pain. Continuous active kinase, protein kinase Mζ (PKMζ), has been known to maintain the long-term potentiation. This study was conducted to determine the role of PKMζ in the IC, which may be involved in the modulation of neuropathic pain. Mechanical allodynia test and immunohistochemistry (IHC) of zif268, an activity-dependent transcription factor required for neuronal plasticity, were performed after nerve injury. After ζ-pseudosubstrate inhibitory peptide (ZIP, a selective inhibitor of PKMζ) injection, mechanical allodynia test and immunoblotting of PKMζ, phospho-PKMζ (p-PKMζ), and GluR1 and GluR2 were observed. IHC demonstrated that zif268 expression significantly increased in the IC after nerve injury. Mechanical allodynia was significantly decreased by ZIP microinjection into the IC. The analgesic effect lasted for 12 hours. Moreover, the levels of GluR1, GluR2, and p-PKMζ were decreased after ZIP microinjection. These results suggest that peripheral nerve injury induces neural plasticity related to PKMζ and that ZIP has potential applications for relieving chronic pain.


Subject(s)
Cerebral Cortex/enzymology , Cerebral Cortex/physiopathology , Neuralgia/physiopathology , Neuronal Plasticity , Peripheral Nerve Injuries/physiopathology , Protein Kinase C/drug effects , Animals , Antigens, Nuclear/metabolism , Cell-Penetrating Peptides , Early Growth Response Protein 1/genetics , Glucose Transporter Type 2/genetics , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Lipopeptides/pharmacology , Male , Nerve Tissue Proteins/metabolism , Neuralgia/enzymology , Pain Measurement/drug effects , Peripheral Nerve Injuries/enzymology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Signal Transduction
7.
J Biol Chem ; 288(33): 24116-27, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23836894

ABSTRACT

Lipocalin 2 (LCN2), which is also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), binds small, hydrophobic ligands and interacts with cell surface receptor 24p3R to regulate diverse cellular processes. In the present study, we examined the role of LCN2 in the pathogenesis of neuropathic pain using a mouse model of spared nerve injury (SNI). Lcn2 mRNA levels were significantly increased in the dorsal horn of the spinal cord after SNI, and LCN2 protein was mainly localized in neurons of the dorsal and ventral horns. LCN2 receptor 24p3R was expressed in spinal neurons and microglia after SNI. Lcn2-deficient mice exhibited significantly less mechanical pain hypersensitivity during the early phase after SNI, and an intrathecal injection of recombinant LCN2 protein elicited mechanical pain hypersensitivity in naive animals. Lcn2 deficiency, however, did not affect acute nociceptive pain. Lcn2-deficient mice showed significantly less microglial activation and proalgesic chemokine (CCL2 and CXCL1) production in the spinal cord after SNI than wild-type mice, and recombinant LCN2 protein induced the expression of these chemokines in cultured neurons. Furthermore, the expression of LCN2 and its receptor was detected in neutrophils and macrophages in the sciatic nerve following SNI, suggesting the potential role of peripheral LCN2 in neuropathic pain. Taken together, our results indicate that LCN2 plays a critical role in the development of pain hypersensitivity following peripheral nerve injury and suggest that LCN2 mediates neuropathic pain by inducing chemokine expression and subsequent microglial activation.


Subject(s)
Acute-Phase Proteins/metabolism , Chemokines/metabolism , Lipocalins/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Oncogene Proteins/metabolism , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/metabolism , Signal Transduction , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/pathology , Chemokines/genetics , Gene Expression Regulation/drug effects , Hyperalgesia/complications , Hyperalgesia/metabolism , Hyperalgesia/pathology , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Models, Biological , Neuralgia/enzymology , Neuralgia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nociception/drug effects , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Exp Cell Res ; 319(15): 2384-94, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23830879

ABSTRACT

Rho GTPases are molecular switches that modulate multiple intracellular signaling processes by means of various effector proteins. As a result, Rho GTPase activities are tightly spatiotemporally regulated in order to ensure homeostasis within the cell. Though the roles of Rho GTPases during neural development have been well documented, their participation during neurodegeneration has been far less characterized. Herein we discuss our current knowledge of the role and function of Rho GTPases and regulators during neurodegeneration, and highlight their potential as targets for therapeutic intervention in common neurodegenerative disorders.


Subject(s)
Gene Expression Regulation , Neurites/metabolism , Neurodegenerative Diseases/genetics , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Humans , Isoenzymes/classification , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Nerve Regeneration/physiology , Neurites/pathology , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurogenesis/genetics , Optic Nerve Injuries/enzymology , Optic Nerve Injuries/genetics , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/genetics , rho GTP-Binding Proteins/classification , rho GTP-Binding Proteins/genetics
9.
J Neurosci ; 32(34): 11737-49, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915116

ABSTRACT

Currents through voltage-gated Ca²âº channels (I(Ca)) may be regulated by cytoplasmic Ca²âº levels ([Ca²âº](c)), producing Ca²âº-dependent inactivation (CDI) or facilitation (CDF). Since I(Ca) regulates sensory neuron excitability, altered CDI or CDF could contribute to pain generation after peripheral nerve injury. We explored this by manipulating [Ca²âº](c) while recording I(Ca) in rat sensory neurons. In uninjured neurons, elevating [Ca²âº](c) with a conditioning prepulse (-15 mV, 2 s) inactivated I(Ca) measured during subsequent test pulses (-15 mV, 5 ms). This inactivation was Ca²âº-dependent (CDI), since it was decreased with elimination of Ca²âº influx by depolarization to above the I(Ca) reversal potential, with high intracellular Ca²âº buffering (EGTA 10 mm or BAPTA 20 mm), and with substitution of Ba²âº for extracellular Ca²âº, revealing a residual voltage-dependent inactivation. At longer latencies after conditioning (>6 s), I(Ca) recovered beyond baseline. This facilitation also proved to be Ca²âº-dependent (CDF) using the protocols limiting cytoplasmic Ca²âº elevation. Ca²âº/calmodulin-dependent protein kinase II (CaMKII) blockers applied by bath (KN-93, myristoyl-AIP) or expressed selectively in the sensory neurons (AIP) reduced CDF, unlike their inactive analogues. Protein kinase C inhibition (chelerythrine) had no effect. Selective blockade of N-type Ca²âº channels eliminated CDF, whereas L-type channel blockade had no effect. Following nerve injury, CDI was unaffected, but CDF was eliminated in axotomized neurons. Excitability of sensory neurons in intact ganglia from control animals was diminished after a similar conditioning pulse, but this regulation was eliminated by injury. These findings indicate that I(Ca) in sensory neurons is subject to both CDI and CDF, and that hyperexcitability following injury-induced loss of CDF may result from diminished CaMKII activity.


Subject(s)
Biophysical Phenomena/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Neurons, Afferent/physiology , Peripheral Nerve Injuries/pathology , Signal Transduction/physiology , Analysis of Variance , Animals , Biophysical Phenomena/drug effects , Biophysics , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Dantrolene/pharmacology , Drug Interactions , Egtazic Acid/analogs & derivatives , Electric Stimulation , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Genetic Vectors/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Laminectomy , Male , Membrane Potentials/drug effects , Neurons, Afferent/drug effects , Pain Threshold/drug effects , Patch-Clamp Techniques , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/enzymology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
10.
Glycobiology ; 22(1): 107-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21840969

ABSTRACT

The polysialic acid (PSA) moiety of the neural cell adhesion molecule (NCAM) has been shown to support dynamic changes underlying peripheral nerve regeneration. Using transgenic mice expressing polysialyltransferase ST8SiaIV under control of a glial-specific (proteolipid protein, PLP) promoter (PLP-ST8SiaIV-transgenic mice), we tested the hypothesis that permanent synthesis of PSA in Schwann cells impairs functional recovery of lesioned peripheral nerves. After sciatic nerve crush, histomorphometric analyses demonstrated impaired remyelination of regenerated axons at the lesion site and in target tissue of PLP-ST8SiaIV-transgenic mice, though the number and size of regenerating unmyelinated axons were not changed. This was accompanied by slower mechanosensory recovery in PLP-ST8SiaIV-transgenic mice. However, the proportion of successfully mono-(re)innervated motor endplates in the foot pad muscle was significantly increased in PLP-ST8SiaIV-transgenic mice when compared with wild-type littermates, suggesting that long-term increase in PSA levels in regenerating nerves may favor selective motor target reinnervation. The combined negative and positive effects of a continuous polysialyltransferase overexpression observed during peripheral nerve regeneration suggest that an optimized time- and differentiation-dependent control of polysialyltransferase expression in Schwann cells may further improve recovery after peripheral nerves injury.


Subject(s)
Gene Expression , Schwann Cells/enzymology , Sciatic Nerve/enzymology , Sialyltransferases/metabolism , Animals , Axons/pathology , Cell Count , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/innervation , Nerve Regeneration , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/physiopathology , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schwann Cells/metabolism , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Sialic Acids/metabolism , Sialyltransferases/genetics
11.
Mol Pain ; 8: 8, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22296727

ABSTRACT

BACKGROUND: Platelet-activating factor (PAF; 1-alkyl-2-acetyl-sn-glycero-3-phosphocholine) is a lipid mediator derived from cell membrane. It has been reported that PAF is involved in various pathological conditions, such as spinal cord injury, multiple sclerosis, neuropathic pain and intrathecal administration of PAF leads to tactile allodynia. However, the expression of PAF synthases and its receptor in the spinal cord following peripheral nerve injury is unknown. METHODS: Using the rat spared nerve injury (SNI) model, we investigated the expression of PAF synthases (LPCAT1 and 2) and PAF receptor (PAFr) mRNAs in the spinal cord. Reverse transcription polymerase chain reaction (RT-PCR) and double-labeling analysis of in situ hybridization histochemistry (ISHH) with immunohistochemistry (IHC) were employed for the analyses. Pain behaviors were also examined with PAFr antagonist (WEB2086). RESULTS: RT-PCR showed that LPCAT2 mRNA was increased in the ipsilateral spinal cord after injury, but not LPCAT1 mRNA. Double-labeling of ISHH with IHC revealed that LPCAT1 and 2 mRNAs were constitutively expressed by a subset of neurons, and LPCAT2 mRNA was increased in spinal microglia after nerve injury. RT-PCR showed that PAFr mRNA was dramatically increased in the ipsilateral spinal cord after nerve injury. Double-labeling analysis of ISHH with IHC revealed that after injury PAFr mRNA was predominantly colocalized with microglia in the spinal cord. Continuous intrathecal administration of the PAFr antagonist suppressed mechanical allodynia following peripheral nerve injury. Delayed administration of a PAFr antagonist did not reverse the mechanical allodynia. CONCLUSIONS: Our data show the histological localization of PAF synthases and its receptor in the spinal cord following peripheral nerve injury, and suggest that PAF/PAFr signaling in the spinal cord acts in an autocrine or paracrine manner among the activated microglia and neurons, thus contributing to development of neuropathic pain.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Neuralgia/etiology , Peripheral Nerve Injuries/complications , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/enzymology , Up-Regulation , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Animals , Azepines/pharmacology , Azepines/therapeutic use , Enzyme Activation/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/pathology , Injections, Spinal , Male , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Neuralgia/enzymology , Neuralgia/pathology , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Spinal Cord/drug effects , Spinal Cord/pathology , Triazoles/pharmacology , Triazoles/therapeutic use , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Clin Invest ; 130(6): 2953-2965, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32134743

ABSTRACT

Platinum-based chemotherapy-induced peripheral neuropathy is one of the most common causes of dose reduction and discontinuation of life-saving chemotherapy in cancer treatment; it often causes permanent impairment of quality of life in cancer patients. The mechanisms that underlie this neuropathy are not defined, and effective treatment and prevention measures are not available. Here, we demonstrate that SIRT2 protected mice against cisplatin-induced peripheral neuropathy (CIPN). SIRT2 accumulated in the nuclei of dorsal root ganglion sensory neurons and prevented neuronal cell death following cisplatin treatment. Mechanistically, SIRT2, an NAD+-dependent deacetylase, protected neurons from cisplatin cytotoxicity by promoting transcription-coupled nucleotide excision repair (TC-NER) of cisplatin-induced DNA cross-links. Consistent with this mechanism, pharmacological inhibition of NER using spironolactone abolished SIRT2-mediated TC-NER activity in differentiated neuronal cells and protection of neurons from cisplatin-induced cytotoxicity and CIPN in mice. Importantly, SIRT2's protective effects were not evident in lung cancer cells in vitro or in tumors in vivo. Taken together, our results identified SIRT2's function in the NER pathway as a key underlying mechanism of preventing CIPN, warranting future investigation of SIRT2 activation-mediated neuroprotection during platinum-based cancer treatment.


Subject(s)
Cisplatin/adverse effects , DNA Repair/drug effects , Neurons , Peripheral Nerve Injuries , Sirtuin 2/metabolism , Animals , Cisplatin/pharmacology , Humans , Mice , Mice, Knockout , Neurons/enzymology , Neurons/pathology , PC12 Cells , Peripheral Nerve Injuries/chemically induced , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/prevention & control , Rats , Sirtuin 2/genetics
13.
Neurotherapeutics ; 17(3): 973-988, 2020 07.
Article in English | MEDLINE | ID: mdl-32236823

ABSTRACT

While the peripheral nervous system is able to repair itself following injury and disease, recovery is often slow and incomplete, with no available treatments to enhance the effectiveness of regeneration. Using knock-out and transgenic overexpressor mice, we previously reported that BACE1, an aspartyl protease, as reported by Hemming et al. (PLoS One 4:12, 2009), negatively regulates peripheral nerve regeneration. Here, we investigated whether pharmacological inhibition of BACE may enhance peripheral nerve repair following traumatic nerve injury or neurodegenerative disease. BACE inhibitor-treated mice had increased numbers of regenerating axons and enhanced functional recovery after a sciatic nerve crush while inhibition increased axonal sprouting following a partial nerve injury. In the SOD1G93A ALS mouse model, BACE inhibition increased axonal regeneration with improved muscle re-innervation. CHL1, a BACE1 substrate, was elevated in treated mice and may mediate enhanced regeneration. Our data demonstrates that pharmacological BACE inhibition accelerates peripheral axon regeneration after varied nerve injuries and could be used as a potential therapy.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Axons/physiology , Nerve Regeneration/physiology , Peripheral Nerve Injuries/enzymology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Animals , Axons/drug effects , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/genetics , Superoxide Dismutase/genetics
14.
Neuropsychopharmacology ; 45(3): 524-533, 2020 02.
Article in English | MEDLINE | ID: mdl-31590180

ABSTRACT

Neuropathic pain caused by nerve injury presents with severe spontaneous pain and a variety of comorbidities, including deficits in higher executive functions. None of these clinical problems are adequately treated with current analgesics. Targeting of the mitogen-activated protein kinase-interacting kinase (MNK1/2) and its phosphorylation target, the mRNA cap binding protein eIF4E, attenuates many types of nociceptive plasticity induced by inflammatory mediators and chemotherapeutic drugs but inhibiting this pathway does not alter nerve injury-induced mechanical allodynia. We used genetic manipulations and pharmacology to inhibit MNK-eIF4E activity in animals with spared nerve injury, a model of peripheral nerve injury (PNI)-induced neuropathic pain. We assessed the presence of spontaneous pain using conditioned place preference. We also tested performance in a medial prefrontal cortex (mPFC)-dependent rule-shifting task. WT neuropathic animals showed signs of spontaneous pain and were significantly impaired in the rule-shifting task while genetic and pharmacological inhibition of the MNK-eIF4E signaling axis protected against and reversed spontaneous pain and PNI-mediated cognitive impairment. Additionally, pharmacological and genetic inhibition of MNK-eIF4E signaling completely blocked and reversed maladaptive shortening in the length of axon initial segments (AIS) in the mPFC of PNI mice. Surprisingly, these striking positive outcomes on neuropathic pain occurred in the absence of any effect on mechanical allodynia, a standard test for neuropathic pain efficacy. Our results illustrate new testing paradigms for determining preclinical neuropathic pain efficacy and point to the MNK inhibitor tomivosertib (eFT508) as an important drug candidate for neuropathic pain treatment.


Subject(s)
Cognitive Dysfunction/therapy , Gene Targeting/methods , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuralgia/therapy , Peripheral Nerve Injuries/therapy , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Animals , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/genetics , Drug Delivery Systems/methods , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neuralgia/enzymology , Neuralgia/genetics , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology
15.
PLoS One ; 14(3): e0213586, 2019.
Article in English | MEDLINE | ID: mdl-30870492

ABSTRACT

We evaluated whether chronic administration of LIMK2-inhibitors could improve erectile function by alleviating CVOD through suppressing cavernosal fibrosis in a rat model of cavernosal nerve crush-injury (CNCI). Forty-two 12-week-old rats were equally categorized into the three groups: sham-surgery (S), CNCI (I), and CNCI treated with LIMK2-inhibitors (L). The L-group was treated with daily intraperitoneal injection of LIMK2-inhibitors (10.0 mg/kg) for 30-days after surgery. Erectile function was assessed using dynamic-infusion-cavernosometry (DIC). Penile tissue was processed for Masson's-trichrome staining, Western-blotting, and double immunofluorescence. The I-group showed significantly higher maintenance and drop rates as well as lower papaverine response, compared to the S-group. Chronic inhibition of LIMK2 in the L-group significantly improved the DIC parameters compared to those in the I-group, although the parameters were not completely restored to normal control values. Also, the I-group showed a reduced smooth muscle (SM)-to-collagen ratio, decreased immunohistochemical staining for α-SM-actin, increased number of fibroblasts positive for phosphorylated Cofilin, increased LIMK2/Cofilin phosphorylation and increased protein expression of Collagen-1 or Fibronectin, compared to the S-group. The L-group showed significant improvements in SM/collagen ratio and the deposition of Collagen-1 or Fibronectin compared to the I-group, although not completely normalized. According to the densitometry and confocal microscopy results, the L-group showed restoration of LIMK2/Cofilin phosphorylation and amount of fibroblasts positive for phosphorylated Cofilin to the normal control value. In conclusion, chronic inhibition of LIMK2 can improve CVOD and ED by alleviating cavernosal fibrosis via normalizing the LIMK2/Cofilin pathway.


Subject(s)
Erectile Dysfunction , Lim Kinases , Penis , Peripheral Nerve Injuries , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Disease Models, Animal , Erectile Dysfunction/drug therapy , Erectile Dysfunction/enzymology , Erectile Dysfunction/pathology , Fibrosis , Lim Kinases/antagonists & inhibitors , Lim Kinases/metabolism , Male , Penis/enzymology , Penis/pathology , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Rats
16.
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
17.
Int J Mol Med ; 42(1): 633-642, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29620141

ABSTRACT

Treatment of neuropathic pain (NPP) continues to be a major challenge, and the underlying mechanisms remain to be elucidated. Previous studies have demonstrated that histone methylation is important in synaptic plasticity of the nervous system and may affect nuclear factor­κB (NF­κB) signaling through epigenetic mechanisms. The present study aimed to investigate the role of Jumonji C domain 6 (JMJD6), a histone demethylase, in a chronic constriction injury (CCI) model of NPP. On the third day post­CCI surgery, a JMJD6 overexpressing lentiviral vector (LV­JMJD6) was intrathecally injected in the rats. Mechanical withdrawal threshold and thermal withdrawal latency were assessed prior surgery and on days 3, 7, 10 and 14 post­CCI. The results showed that intrathecal injection with the LV­JMJD6 attenuated CCI­induced pain facilitation. The expression of JMJD6 was lower following CCI surgery, and its expression was significantly increased following intrathecal injection with LV­JMJD6, compared with levels in normal saline (NS)­ and negative control lentiviral vector (NC)­treated rats. The expression of spinal NF­κB phosphorylated (p­)p65 subunit and its downstream pain­associated effectors, including interleukin 1ß (IL­1ß), tumor necrosis factor­α (TNF­α) and vascular endothelial growth factor (VEGF), were increased following CCI surgery. Intrathecal injection with LV­JMJD6 suppressed activation of the p­p65 subunit in CCI rats. In addition, expression levels of its downstream effectors IL­1ß, TNF­α and VEGF were attenuated by intrathecal treatment with LV­JMJD6, compared with those in the NS­ and NC­treated CCI rats. Furthermore, the JMJD6­ and p65­immunoreactive cells overlapped in the spinal dorsal horn, however, co­immunoprecipitation showed that JMJD6 and the NF­κB p65 subunit did not directly interact, indicating other functional connections may exist between these factors following CCI surgery. Collectively, these findings indicated an important mechanism underlying the pathogenesis of NPP. JMJD6 may exert its therapeutic function in NPP by regulating NF­κB following CCI.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , NF-kappa B/metabolism , Neuralgia/complications , Neuralgia/enzymology , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/enzymology , Animals , Behavior, Animal , Chronic Disease , Constriction, Pathologic , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Injections, Spinal , Lentivirus/metabolism , Male , Models, Biological , Neuralgia/pathology , Protein Binding , Protein Subunits/metabolism , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/pathology , Transfection , Vascular Endothelial Growth Factor A/metabolism
18.
Nat Cell Biol ; 20(3): 307-319, 2018 03.
Article in English | MEDLINE | ID: mdl-29434374

ABSTRACT

Reactive oxygen species (ROS) contribute to tissue damage and remodelling mediated by the inflammatory response after injury. Here we show that ROS, which promote axonal dieback and degeneration after injury, are also required for axonal regeneration and functional recovery after spinal injury. We find that ROS production in the injured sciatic nerve and dorsal root ganglia requires CX3CR1-dependent recruitment of inflammatory cells. Next, exosomes containing functional NADPH oxidase 2 complexes are released from macrophages and incorporated into injured axons via endocytosis. Once in axonal endosomes, active NOX2 is retrogradely transported to the cell body through an importin-ß1-dynein-dependent mechanism. Endosomal NOX2 oxidizes PTEN, which leads to its inactivation, thus stimulating PI3K-phosporylated (p-)Akt signalling and regenerative outgrowth. Challenging the view that ROS are exclusively involved in nerve degeneration, we propose a previously unrecognized role of ROS in mammalian axonal regeneration through a NOX2-PI3K-p-Akt signalling pathway.


Subject(s)
Axons/enzymology , Exosomes/enzymology , Ganglia, Spinal/enzymology , NADPH Oxidase 2/metabolism , Nerve Degeneration , Nerve Regeneration , Peripheral Nerve Injuries/enzymology , Reactive Oxygen Species/metabolism , Sciatic Nerve/enzymology , Spinal Cord Injuries/enzymology , Animals , Axons/pathology , CX3C Chemokine Receptor 1/metabolism , Cell Line , Disease Models, Animal , Dyneins/metabolism , Endocytosis , Endosomes/enzymology , Endosomes/pathology , Exosomes/pathology , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Macrophages/enzymology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2/deficiency , NADPH Oxidase 2/genetics , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Signal Transduction , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , beta Karyopherins
19.
Elife ; 72018 07 03.
Article in English | MEDLINE | ID: mdl-29968565

ABSTRACT

Neuropathic pain resulting from nerve injury can become persistent and difficult to treat but the molecular signaling responsible for its development remains poorly described. Here, we identify the neuronal stress sensor dual leucine zipper kinase (DLK; Map3k12) as a key molecule controlling the maladaptive pathways that lead to pain following injury. Genetic or pharmacological inhibition of DLK reduces mechanical hypersensitivity in a mouse model of neuropathic pain. Furthermore, DLK inhibition also prevents the spinal cord microgliosis that results from nerve injury and arises distant from the injury site. These striking phenotypes result from the control by DLK of a transcriptional program in somatosensory neurons regulating the expression of numerous genes implicated in pain pathogenesis, including the immune gene Csf1. Thus, activation of DLK is an early event, or even the master regulator, controlling a wide variety of pathways downstream of nerve injury that ultimately lead to chronic pain.


Subject(s)
Gliosis/genetics , Hyperalgesia/genetics , MAP Kinase Kinase Kinases/genetics , Neuralgia/genetics , Peripheral Nerve Injuries/genetics , Sensory Receptor Cells/enzymology , Animals , Disease Models, Animal , Female , Gene Expression Regulation , Gliosis/enzymology , Gliosis/pathology , Gliosis/prevention & control , Hyperalgesia/enzymology , Hyperalgesia/pathology , Hyperalgesia/prevention & control , MAP Kinase Kinase Kinases/deficiency , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Transgenic , Microglia/enzymology , Microglia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/enzymology , Neuralgia/pathology , Neuralgia/prevention & control , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Sciatic Nerve/enzymology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sensory Receptor Cells/pathology , Signal Transduction , Spinal Cord/enzymology , Spinal Cord/pathology , Touch , Transcription, Genetic
20.
Neuroscience ; 320: 93-104, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26851772

ABSTRACT

Although thrombin has an important role in both central and peripheral nerve diseases, characterization of the anatomical distribution of its proteolytic activity has been limited by available methods. This study presents the development, challenges, validation and implementation of a novel histochemical method for visualization of thrombin activity in the nervous system. The method is based on the cleavage of the substrate, Boc-Asp(OBzl)-Pro-Arg-4MßNA by thrombin to liberate free 4-methoxy-2-naphthylamine (4MßNA). In the presence of 5-nitrosalicylaldehyde, free 4MßNA is captured, yielding an insoluble yellow fluorescent precipitate which marks the site of thrombin activity. The sensitivity of the method was determined in vitro using known concentrations of thrombin while the specificity was verified using a highly specific thrombin inhibitor. Using this method we determined the spatial distribution of thrombin activity in mouse brain following transient middle cerebral artery occlusion (tMCAo) and in mouse sciatic nerve following crush injury. Fluorescence microscopy revealed well-defined thrombin activity localized to the right ischemic hemisphere in cortical areas and in the striatum compared to negligible thrombin activity contralaterally. The histochemical localization of thrombin activity following tMCAo was in good correlation with the infarct areas per triphenyltetrazolium chloride staining and to thrombin activity measured biochemically in tissue punches (85 ± 35 and 20 ± 3 mU/ml, in the cortical and striatum areas respectively, compared to 7 ± 2 and 13 ± 2 mU/ml, in the corresponding contralateral areas; mean ± SEM; p<0.05). In addition, 24 h following crush injury, focal areas of highly elevated thrombin activity were detected in teased sciatic fibers. This observation was supported by the biochemical assay and western blot technique. The histochemical method developed in this study can serve as an important tool for studying the role of thrombin in physiological and pathological conditions.


Subject(s)
Brain/enzymology , Histocytochemistry/methods , Peripheral Nerves/enzymology , Thrombin/analysis , Animals , Disease Models, Animal , Mice , Peripheral Nerve Injuries/enzymology , Sensitivity and Specificity , Stroke/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL