Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 266
Filter
Add more filters

Publication year range
1.
Mol Pain ; 16: 1744806920943685, 2020.
Article in English | MEDLINE | ID: mdl-32865105

ABSTRACT

Neuropathic pain can be generated by chronic compression of dorsal root ganglion (CCD). Stimulation of primary motor cortex can disrupt the nociceptive sensory signal at dorsal root ganglion level and reduce pain behaviors. But the mechanism behind it is still implicit. Protein kinase C gamma is known as an essential enzyme for the development of neuropathic pain, and specific inhibitor of protein kinase C gamma can disrupt the sensory signal and reduce pain behaviors. Optogenetic stimulation has been emerged as a new and promising conducive method for refractory neuropathic pain. The aim of this study was to provide evidence whether optical stimulation of primary motor cortex can modulate chronic neuropathic pain in CCD rat model. Animals were randomly divided into CCD group, sham group, and control group. Dorsal root ganglion-compressed neuropathic pain model was established in animals, and knocking down of protein kinase C gamma was also accomplished. Pain behavioral scores were significantly improved in the short hairpin Protein Kinase C gamma knockdown CCD animals during optic stimulation. Ventral posterolateral thalamic firing inhibition was also observed during light stimulation on motor cortex in CCD animal. We assessed alteration of pain behaviors in pre-light off, stimulation-light on, and post-light off state. In vivo extracellular recording of the ventral posterolateral thalamus, viral expression in the primary motor cortex, and protein kinase C gamma expression in dorsal root ganglion were investigated. So, optical cortico-thalamic inhibition by motor cortex stimulation can improve neuropathic pain behaviors in CCD animal, and knocking down of protein kinase C gamma plays a conducive role in the process. This study provides feasibility for in vivo optogenetic stimulation on primary motor cortex of dorsal root ganglion-initiated neuropathic pain.


Subject(s)
Ganglia, Spinal/metabolism , Motor Cortex/metabolism , Neuralgia/metabolism , Optogenetics/methods , Protein Kinase C/metabolism , Thalamus/metabolism , Animals , Behavior Rating Scale , Behavior, Animal/physiology , Female , Ganglia, Spinal/enzymology , Ganglia, Spinal/injuries , Gene Knockdown Techniques , Immunohistochemistry , Motor Cortex/enzymology , Motor Cortex/radiation effects , Neuralgia/genetics , Optical Fibers , Protein Kinase C/genetics , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Thalamus/enzymology
2.
Mol Pain ; 16: 1744806920956480, 2020.
Article in English | MEDLINE | ID: mdl-32909881

ABSTRACT

To develop non-opioid therapies for postoperative incisional pain, we must understand its underlying molecular mechanisms. In this study, we assessed global gene expression changes in dorsal root ganglia neurons in a model of incisional pain to identify pertinent molecular pathways. Male, Sprague-Dawley rats underwent infiltration of 1% capsaicin or vehicle into the plantar hind paw (n = 6-9/group) 30 min before plantar incision. Twenty-four hours after incision or sham (control) surgery, lumbar L4-L6 dorsal root ganglias were collected from rats pretreated with vehicle or capsaicin. RNA was isolated and sequenced by next generation sequencing. The genes were then annotated to functional networks using a knowledge-based database, Ingenuity Pathway Analysis. In rats pretreated with vehicle, plantar incision caused robust hyperalgesia, up-regulated 36 genes and downregulated 90 genes in dorsal root ganglias one day after plantar incision. Capsaicin pretreatment attenuated pain behaviors, caused localized denervation of the dermis and epidermis, and prevented the incision-induced changes in 99 of 126 genes. The pathway analyses showed altered gene networks related to increased pro-inflammatory and decreased anti-inflammatory responses in dorsal root ganglias. Insulin-like growth factor signaling was identified as one of the major gene networks involved in the development of incisional pain. Expression of insulin-like growth factor -2 and IGFBP6 in dorsal root ganglia were independently validated with quantitative real-time polymerase chain reaction. We discovered a distinct subset of dorsal root ganglia genes and three key signaling pathways that are altered 24 h after plantar incision but are unchanged when incision was made after capsaicin infiltration in the skin. Further exploration of molecular mechanisms of incisional pain may yield novel therapeutic targets.


Subject(s)
Capsaicin/pharmacology , Ganglia, Spinal/metabolism , Pain, Postoperative/drug therapy , Signal Transduction/drug effects , Somatomedins/metabolism , Transcriptome/genetics , Animals , Behavior Rating Scale , Capsaicin/therapeutic use , Computational Biology , Down-Regulation , Ganglia, Spinal/injuries , Gene Expression Profiling , Gene Regulatory Networks , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Male , RNA-Seq , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Somatomedins/genetics , Surgical Wound/complications , Up-Regulation
3.
Mol Pain ; 16: 1744806920955103, 2020.
Article in English | MEDLINE | ID: mdl-32880221

ABSTRACT

Neuropathic pain is a chronic disease state resulting from injury to the nervous system. This type of pain often responds poorly to standard treatments and occasionally may get worse instead of better over time. Patients who experience neuropathic pain report sensitivity to cold and mechanical stimuli. Since the nociceptive system of African naked mole-rats contains unique adaptations that result in insensitivity to some pain types, we investigated whether naked mole-rats may be resilient to sensitivity following nerve injury. Using the spared nerve injury model of neuropathic pain, we showed that sensitivity to mechanical stimuli developed similarly in mice and naked mole-rats. However, naked mole-rats lacked sensitivity to mild cold stimulation after nerve injury, while mice developed robust cold sensitivity. We pursued this response deficit by testing behavior to activators of transient receptor potential (TRP) receptors involved in detecting cold in naïve animals. Following mustard oil, a TRPA1 activator, naked mole-rats responded similarly to mice. Conversely, icilin, a TRPM8 agonist, did not evoke pain behavior in naked mole-rats when compared with mice. Finally, we used RNAscope to probe for TRPA1 and TRPM8 messenger RNA expression in dorsal root ganglia of both species. We found increased TRPA1 messenger RNA, but decreased TRPM8 punctae in naked mole-rats when compared with mice. Our findings likely reflect species differences due to evolutionary environmental responses that are not easily explained by differences in receptor expression between the species.


Subject(s)
Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , Neuralgia/metabolism , TRPA1 Cation Channel/metabolism , TRPM Cation Channels/metabolism , Animals , Cold Temperature , Disease Models, Animal , Female , Ganglia, Spinal/injuries , Male , Mice , Mole Rats , Mustard Plant , Neurons/metabolism , Neurons/physiology , Nociception , Pain Measurement , Plant Oils/pharmacology , Pyrimidinones/pharmacology , TRPA1 Cation Channel/genetics , TRPM Cation Channels/agonists , TRPM Cation Channels/genetics
4.
Neuromodulation ; 23(2): 167-176, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32103594

ABSTRACT

OBJECTIVE: The role of the nucleus accumbens (NAc) in chronic neuropathic pain has been suggested, but the role of the NAc in dorsal root ganglion (DRG) neuropathic pain remains unclear. The objective of this study was to determine whether optogenetic stimulation of the NAc influences DRG compression-induced neuropathic pain. MATERIALS AND METHODS: We established sham or DRG lesions in female Sprague-Dawley rats by L4-5 DRG root compression, and the animals received unilateral injections of optogenetic virus in the NAc core. We employed reflexive pain tests to assess the alterations between the groups at the light on/off states. To determine thalamic firing, we performed single-unit in vivo extracellular recording. For statistical analysis, we used one- or two-way repeated-measures analysis of variance. RESULTS: Compared to sham-operated rats, chronic compressed DRG rats showed elevated behavioral sensitivity and sustained neuronal hyperexcitability in the thalamus. NAc optic stimulation improved pain behaviors and lowered thalamic discharge from ventral posterolateral thalamic nuclei. CONCLUSIONS: The NAc core impacts the reward and motivational aspects of chronic neuropathic pain influenced by limbic behaviors to thalamic discharge. Increased thalamic firing activity may result in chronic compressed DRG-induced neuropathic pain, and optogenetic neuromodulation of the NAc can ease chronic pain and thalamic discharge.


Subject(s)
Ganglia, Spinal/injuries , Laser Therapy/methods , Nerve Compression Syndromes/therapy , Neuralgia/therapy , Nucleus Accumbens/physiology , Optical Fibers , Animals , Disease Models, Animal , Female , Ganglia, Spinal/physiopathology , Nerve Compression Syndromes/physiopathology , Neuralgia/physiopathology , Pain Management/methods , Rats , Rats, Sprague-Dawley
5.
J Neurosci ; 38(27): 6090-6101, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29875269

ABSTRACT

Nerve injury-induced hyperactivity of primary sensory neurons in the dorsal root ganglion (DRG) contributes to chronic pain development, but the underlying epigenetic mechanisms remain poorly understood. Here we determined genome-wide changes in DNA methylation in the nervous system in neuropathic pain. Spinal nerve ligation (SNL), but not paclitaxel treatment, in male Sprague Dawley rats induced a consistent low-level hypomethylation in the CpG sites in the DRG during the acute and chronic phases of neuropathic pain. DNA methylation remodeling in the DRG occurred early after SNL and persisted for at least 3 weeks. SNL caused DNA methylation changes at 8% of CpG sites with prevailing hypomethylation outside of CpG islands, in introns, intergenic regions, and repetitive sequences. In contrast, SNL caused more gains of methylation in the spinal cord and prefrontal cortex. The DNA methylation changes in the injured DRGs recapitulated developmental reprogramming at the neonatal stage. Methylation reprogramming was correlated with increased gene expression variability. A diet deficient in methyl donors induced hypomethylation and pain hypersensitivity. Intrathecal administration of the DNA methyltransferase inhibitor RG108 caused long-lasting pain hypersensitivity. DNA methylation reprogramming in the DRG thus contributes to nerve injury-induced chronic pain. Restoring DNA methylation may represent a new therapeutic approach to treat neuropathic pain.SIGNIFICANCE STATEMENT Epigenetic mechanisms are critically involved in the transition from acute to chronic pain after nerve injury. However, genome-wide changes in DNA methylation in the nervous system and their roles in neuropathic pain development remain unclear. Here we used digital restriction enzyme analysis of methylation to quantitatively determine genome-wide DNA methylation changes caused by nerve injury. We showed that nerve injury caused DNA methylation changes at 8% of CpG sites with prevailing hypomethylation outside of CpG islands in the dorsal root ganglion. Reducing DNA methylation induced pain hypersensitivity, whereas increasing DNA methylation attenuated neuropathic pain. These findings extend our understanding of the epigenetic mechanism of chronic neuropathic pain and suggest new strategies to treat nerve injury-induced chronic pain.


Subject(s)
Chronic Pain/metabolism , DNA Methylation/physiology , Ganglia, Spinal/metabolism , Neuralgia/metabolism , Animals , Chronic Pain/genetics , Epigenesis, Genetic/genetics , Ganglia, Spinal/injuries , Male , Neuralgia/genetics , Rats , Rats, Sprague-Dawley
6.
Neurochem Res ; 44(5): 1214-1227, 2019 May.
Article in English | MEDLINE | ID: mdl-30859436

ABSTRACT

Neuropathic pain (NP) has complicated pathogenesis as it mainly involves a lesion or dysfunction of the somatosensory nervous system and its clinical treatment remains challenging. Chronic constriction injury (CCI) model is a widely used neuropathic pain model and involved in mechanisms including both nerve inflammatory and injury. Cytokines and their receptors play essential roles in the occurrence and persistence of neuropathic pain, but the underlying mechanisms have not well been understood. Therefore, Interleukin-1 receptor-associated kinase 1 (IRAK1) is chosen to explore the possible mechanisms of NP. In the present study, IRAK1 was found to persistently increase in the dorsal root ganglion (DRG) and spinal cord (SC) during CCI detected by western blot. The staining further confirmed that IRAK1 was mainly co-located in the DRG astrocytes or SC neurons, but less in the DRG microglia or SC astrocytes. Moreover, the region of increased IRAK1 expression was observed in superficial laminae of the spinal dorsal horn, which was the nociceptive neuronal expression domain, suggesting that IRAK1 may mediated CCI-induced pain by nociceptive primary afferent. In addition, intrathecal injection of Toll-like receptor 4 (TLR4) inhibitor or IRAK1 siRNA decreased the expression of IRAK1 accompanied with the alleviation of CCI-induced neuropathic pain. The upregulation of p-NF-κB expression was reversed by IRAK1 siRNA in SC, and intrathecal injection of p-NF-κB inhibitor relieved neuropathic pain. Taking together, targeting IRAK1 may be a potential treatment for chronic neuropathic pain.


Subject(s)
Ganglia, Spinal/metabolism , Neuralgia/metabolism , Neuralgia/physiopathology , Sciatic Nerve/injuries , Animals , Chronic Disease , Constriction , Ganglia, Spinal/injuries , Hyperalgesia/metabolism , Male , Microglia/metabolism , Nociceptors/metabolism , Rats, Sprague-Dawley , Receptors, Interleukin-1/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology
7.
Neurol India ; 67(Supplement): S32-S37, 2019.
Article in English | MEDLINE | ID: mdl-30688230

ABSTRACT

Peripheral nerve and brachial plexus injuries typically cause severe impairment in the affected limb. The incidence of neuropathic pain is high, reaching up to 95% of cases, especially if cervical root avulsion has occurred. Neuropathic pain results from damage to the somatosensory system, and its progression towards chronicity depends upon disruptions affecting both the peripheral and central nervous system. Managing these painful conditions is complex and must be accomplished by a multidisciplinary team, starting with first-line pharmacological therapies like tricyclic antidepressants and calcium channel ligands, combined physical and occupational therapy, transcutaneous electrical stimulation and psychological support. For patients refractory to the initial measures, several neurosurgical options are available, including nerve decompression or reconstruction and ablative/modulatory procedures.


Subject(s)
Brachial Plexus/injuries , Neuralgia/therapy , Peripheral Nerve Injuries/complications , Brachial Plexus/physiopathology , Ganglia, Spinal/injuries , Ganglia, Spinal/physiopathology , Humans , Neuralgia/etiology , Neuralgia/physiopathology , Peripheral Nerve Injuries/physiopathology , Treatment Outcome
8.
J Recept Signal Transduct Res ; 38(3): 198-203, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29932348

ABSTRACT

OBJECTIVES: Here, using rat model, we investigated the roles of gardenoside in the chronic constriction injury (CCI) of the ischiadic nerve. METHODS: Bennett and Xie's unilateral sciatic nerve CCI model was used in this study. A total of 60 rats were divided into control group (CN), sham group (Sham), CCI group, and gardenoside administrated CCI group. An aliquot of 5 mL gardenoside solution was administrated through gavage once per day for 14 d. Mechanical withdrawal threshold (MWT) and the thermal withdrawal latency (TWL) were detected. The levels of inducible nitric oxide synthase (iNOS), interleukin-1ß (IL-1ß), and tumor necrosis factor-α (TNF-α) in spinal fluid were detected by ELISA. By using real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot, we analyzed the expression of P2X purinoceptor 3 and 7 (P2X3 and P2X7 receptors) in different groups. The expression of p-ERK/ERK and p-p38/p38 were also detected by western blot. RESULTS: We found out that gardenoside could significantly improve the sciatica by partially restore the decrease of MWT and TWL in CCI rats. The levels of iNOS, IL-1ß, and TNF-α were higher in CCI group (p < .05). The expressions of P2X3 and P2X7 were significantly increased in the CCI rats compared to control rats (p < .05). The levels of p-ERK/ERK and p-p38/p38 were also obviously increased in CCI group (p < .05). After treated with the gardenoside, these increases were decreased. CONCLUSIONS: These results indicated that gardenoside may be able to relief CCI-induced neuropathic pain by regulating the P2X3 and the P2X7 expression on the ischiadic nerve.


Subject(s)
Iridoids/administration & dosage , Neuralgia/drug therapy , Receptors, Purinergic P2X3/genetics , Receptors, Purinergic P2X7/genetics , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Gene Expression Regulation , Humans , Interleukin-1beta , Neuralgia/genetics , Neuralgia/physiopathology , Nitric Oxide Synthase Type II , Pain Threshold , Rats , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Tumor Necrosis Factor-alpha
9.
J Biol Chem ; 291(16): 8475-85, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26917724

ABSTRACT

The µ-opioid receptor (MOR, encoded by Oprm1) agonists are the mainstay analgesics for treating moderate to severe pain. Nerve injury causes down-regulation of MORs in the dorsal root ganglion (DRG) and diminishes the opioid effect on neuropathic pain. However, the epigenetic mechanisms underlying the diminished MOR expression caused by nerve injury are not clear. G9a (encoded by Ehmt2), a histone 3 at lysine 9 methyltransferase, is a key chromatin regulator responsible for gene silencing. In this study, we determined the role of G9a in diminished MOR expression and opioid analgesic effects in animal models of neuropathic pain. We found that nerve injury in rats induced a long-lasting reduction in the expression level of MORs in the DRG but not in the spinal cord. Nerve injury consistently increased the enrichment of the G9a product histone 3 at lysine 9 dimethylation in the promoter of Oprm1 in the DRG. G9a inhibition or siRNA knockdown fully reversed MOR expression in the injured DRG and potentiated the morphine effect on pain hypersensitivity induced by nerve injury. In mice lacking Ehmt2 in DRG neurons, nerve injury failed to reduce the expression level of MORs and the morphine effect. In addition, G9a inhibition or Ehmt2 knockout in DRG neurons normalized nerve injury-induced reduction in the inhibitory effect of the opioid on synaptic glutamate release from primary afferent nerves. Our findings indicate that G9a contributes critically to transcriptional repression of MORs in primary sensory neurons in neuropathic pain. G9a inhibitors may be used to enhance the opioid analgesic effect in the treatment of chronic neuropathic pain.


Subject(s)
Analgesia , Ganglia, Spinal/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, Opioid, mu/metabolism , Sensory Receptor Cells/metabolism , Transcription, Genetic/drug effects , Animals , Female , Ganglia, Spinal/injuries , Ganglia, Spinal/pathology , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Male , Mice , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/genetics
10.
J Biol Chem ; 290(23): 14647-55, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25925949

ABSTRACT

Pannexin-1 (Panx1) is a large-pore membrane channel involved in the release of ATP and other signaling mediators. Little is known about the expression and functional role of Panx1 in the dorsal root ganglion (DRG) in the development of chronic neuropathic pain. In this study, we determined the epigenetic mechanism involved in increased Panx1 expression in the DRG after nerve injury. Spinal nerve ligation in rats significantly increased the mRNA and protein levels of Panx1 in the DRG but not in the spinal cord. Immunocytochemical labeling showed that Panx1 was primarily expressed in a subset of medium and large DRG neurons in control rats and that nerve injury markedly increased the number of Panx1-immunoreactive DRG neurons. Nerve injury significantly increased the enrichment of two activating histone marks (H3K4me2 and H3K9ac) and decreased the occupancy of two repressive histone marks (H3K9me2 and H3K27me3) around the promoter region of Panx1 in the DRG. However, nerve injury had no effect on the DNA methylation level around the Panx1 promoter in the DRG. Furthermore, intrathecal injection of the Panx1 blockers or Panx1-specific siRNA significantly reduced pain hypersensitivity induced by nerve injury. In addition, siRNA knockdown of Panx1 expression in a DRG cell line significantly reduced caspase-1 release induced by neuronal depolarization. Our findings suggest that nerve injury increases Panx1 expression levels in the DRG through altered histone modifications. Panx1 up-regulation contributes to the development of neuropathic pain and stimulation of inflammasome signaling.


Subject(s)
Connexins/genetics , Ganglia, Spinal/injuries , Nerve Tissue Proteins/genetics , Neuralgia/etiology , Neuralgia/genetics , Up-Regulation , Animals , Connexins/metabolism , DNA Methylation , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Male , Nerve Tissue Proteins/metabolism , Neuralgia/pathology , Promoter Regions, Genetic , Rats, Sprague-Dawley
11.
BMC Complement Altern Med ; 16(1): 402, 2016 Oct 22.
Article in English | MEDLINE | ID: mdl-27770785

ABSTRACT

BACKGROUND: Bogijetong decoction (BGJTD) is a herbal drug formulation used in the traditional Asian medicine to treat neuropathic insults associated with diabetes and anticancer therapy. To understand the biological basis of BGJTD on protective effects against neuropathy, we investigated physiological and biochemical responses of the sciatic nerves deranged by taxol injection or crush injury in the rats. METHODS: Dissociated Schwann cells and neurons were prepared from the sciatic nerve and dorsal root ganglia (DRG) respectively and were treated with taxol and BGJTD. The sciatic nerve in the rat was injected with taxol or given crush injury. Animals were then administered orally with BGJTD. Effects of BGJTD treatment on cultured cells and in vivo sciatic nerves and DRG tissues were examined by immunofluorescence staining and western blot analysis. Sciatic nerve regeneration was assessed by histological observation using retrograde tracing technique and by behavioral hot plate test. Eighteen different herbal components of BGJTD were divided into 4 subgroups and were used to select herbal drugs that enhanced neurite outgrowth in cultured neurons. RESULTS: Morphological abnormalities in the sciatic nerve axons and DRG tissue caused by taxol injection were largely improved by BGJTD treatment. BGJTD treatment enhanced neurite outgrowth in cultured DRG neurons and improved Schwann cell survival. Phospho-Erk1/2 levels were elevated by BGJTD administration in the injured- or taxol-injected sciatic nerves. Vimentin phosphorylation catalyzed by cell division cycle 2 (Cdc2) kinase was induced from Schwann cells in the sciatic nerves after taxol injection and crush injury, and phospho-vimentin levels were further upregulated by BGJTD treatment. Retrograde tracing of DiI-labeled DRG sensory neurons revealed growth-promoting activity of BGJTD on axonal regeneration. A drug group (Be) composed of 4 active herbal components which were selected by neurite growth-enhancing activity was as effective as BGJDT for the recovery of thermal sensitivity of the hind paws which had been suppressed by taxol administration. CONCLUSIONS: These data suggest that BGJTD and its active herbal components may protects the peripheral nerve from damage caused by taxol injection and nerve crush.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Nerve Regeneration/drug effects , Peripheral Nerve Injuries , Protective Agents/pharmacology , Animals , Drugs, Chinese Herbal/chemistry , Ganglia, Spinal/drug effects , Ganglia, Spinal/injuries , Male , Mice , Mice, Inbred BALB C , Nerve Crush , Neurites/drug effects , Paclitaxel/adverse effects , Protective Agents/chemistry , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/injuries
12.
Dev Neurosci ; 37(1): 66-77, 2015.
Article in English | MEDLINE | ID: mdl-25592862

ABSTRACT

In contrast to adult rat nerve injury models, neonatal sciatic nerve crush leads to massive motor and sensory neuron death. Death of these neurons results from both the loss of functional contact between the nerve terminals and their targets, and the inability of immature Schwann cells in the distal stump of the injured nerve to sustain regeneration. However, current dogma holds that little to no motoneuron death occurs in response to nerve crush at postnatal day 5 (P5). The purpose of the current study was to fully characterize the extent of motor and sensory neuronal death and functional recovery following sciatic nerve crush at mid-thigh level in rats at postnatal days 3-30 (P3-P30), and then compare this to adult injured animals. Following nerve crush at P3, motoneuron numbers were reduced to 35% of that of naïve uninjured animals. Animals in the P5 and P7 group also displayed statistically fewer motoneurons than naïve animals. Animals that were injured at P30 or earlier displayed statistically lower sensory neuron counts in the dorsal root ganglion than naïve controls. Surprisingly, complete behavioral recovery was observed exclusively in the P30 and adult injured groups. Similar results were observed in muscle twitch/tetanic force analysis, motor unit number estimation and wet muscle weights. Rats in both the P5 and P7 injury groups displayed significant neuronal death and impaired functional recovery following injury, challenging current dogma and suggesting that severe deficits persist following nerve injury during this early postnatal developmental period. These findings have important implications concerning the timing of neonatal nerve injury in rats.


Subject(s)
Ganglia, Spinal/injuries , Motor Neurons/pathology , Nerve Crush , Nerve Regeneration/physiology , Sciatic Nerve/injuries , Animals , Animals, Newborn , Cell Death , Ganglia, Spinal/pathology , Nerve Crush/methods , Rats, Inbred Lew , Sciatic Nerve/pathology
14.
J Neurosci ; 33(41): 16236-48, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107955

ABSTRACT

Macrophages have been implicated in peripheral nerve regeneration for some time, supposedly through their involvement in Wallerian degeneration, the process by which the distal nerve degenerates after axotomy and is cleared by phagocytosis. Thus, in several studies in which macrophage accumulation in the distal nerve was reduced and Wallerian degeneration inhibited, regeneration was delayed. However, this interpretation ignores the more recent findings that macrophages also accumulate around axotomized cell bodies. The function of macrophage action at this second site has not been clear. In two mutant strains of mice, the slow Wallerian degeneration (Wld(s)) mouse and the chemokine receptor CCR2 knock-out mouse, we report that macrophage accumulation after axotomy was abolished in both the dorsal root ganglion (DRG) and the distal sciatic nerve. To measure neurite outgrowth, DRG neurons were given a conditioning lesion, and outgrowth was measured in vitro 7 d later in the absence of the distal nerve segment. The increased growth normally seen after a conditioning lesion did not occur or was reduced in Wld(s) or CCR2(-/-) mice. In the superior cervical ganglion (SCG), particularly in Wld(s) mice, macrophage accumulation was reduced but not abolished after axotomy. In SCG neurons from Wld(s) mice, the conditioning lesion response was unchanged; however, in CCR2(-/-) mice in which the effect on macrophage accumulation was greater, SCG neurite outgrowth was significantly reduced. These results indicate that macrophages affect neurite outgrowth by acting at the level of peripheral ganglia in addition to any effects they might produce by facilitation of Wallerian degeneration.


Subject(s)
Ganglia, Spinal/physiology , Macrophages , Nerve Regeneration/physiology , Neurites/physiology , Sciatic Nerve/physiology , Animals , Axotomy , Female , Ganglia, Spinal/injuries , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/physiology , Real-Time Polymerase Chain Reaction , Sciatic Nerve/injuries
15.
Glia ; 62(5): 763-77, 2014 May.
Article in English | MEDLINE | ID: mdl-24616056

ABSTRACT

Satellite glial cells (SGCs) surrounding primary sensory neurons are similar to astrocytes of the central nervous system in that they buffer the extracellular environment via potassium and calcium channels and express the intermediate filament glial fibrillary acidic protein (GFAP). Peripheral nerve injury induces a reactive state in SGCs that includes SGC proliferation, increased SGC/SGC coupling via gap junctions, decreased inward rectifying potassium channel 4.1 (Kir 4.1) expression and increased expression of GFAP and the common neurotrophin receptor, p75NTR. In contrast, neuronal p75NTR expression, normally detected in ∼80% of adult rat sensory neurons, decreases in response to peripheral axotomy. Given the differential regulation of p75NTR expression in neurons versus SGCs with injury, we hypothesized that reduced signaling via neuronal p75NTR contributes to the induction of a reactive state in SGCs. We found that reducing neuronal p75NTR protein expression in uninjured sensory neurons by intrathecal subarachnoid infusion of p75NTR-selective anti-sense oligodeoxynucleotides for one week was sufficient to induce a "reactive-like" state in the perineuronal SGCs akin to that normally observed following peripheral nerve injury. This reactive state included significantly increased SGC p75NTR, GFAP and gap junction protein connexin-43 protein expression, increased numbers of SGCs surrounding individual sensory neurons and decreased SGC Kir 4.1 channel expression. Collectively, this supports the tenet that reductions in target-derived trophic support leading to, or as a consequence of, reduced neuronal p75NTR expression plays a critical role in switching the SGC to a reactive state.


Subject(s)
Ganglia, Spinal/injuries , Ganglia, Spinal/metabolism , Neuroglia/metabolism , Receptors, Nerve Growth Factor/biosynthesis , Satellite Cells, Perineuronal/metabolism , Sensory Receptor Cells/metabolism , Animals , Ganglia, Spinal/drug effects , Gene Expression Regulation , Injections, Spinal , Male , Nerve Tissue Proteins , Neuroglia/drug effects , Oligonucleotides, Antisense/administration & dosage , Rats , Rats, Wistar , Receptors, Growth Factor , Satellite Cells, Perineuronal/drug effects , Sensory Receptor Cells/drug effects
16.
Spinal Cord ; 52(9): 658-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25023860

ABSTRACT

OBJECTIVES: To compare changes in lower urinary tract (LUT) function with modifications in pathways that regulate LUT function using two different animal models (incomplete and complete) of spinal cord injury (SCI). METHODS: Female Sprague-Dawley rats were used. SCI was made at Th8/9 by a contusion injury (contusion, n=9) or a complete transection (transection, n=9). Unoperated rats were used as normal controls (normal, n=6). LUT function was evaluated by micturition behavior in metabolic cages for 24 h and cystometry in awake animals. Immunocytochemical staining at the L6 spinal cord, spinal areas associated with LUT, was performed to identify descending modulatory fibers and dorsal root afferents that project to the L6 spinal cord. RESULTS: Volume/micturition in metabolic cages gradually increased in both contusion and transection groups compared with normals, and operated groups did not differ from each other. Urodynamic parameters from cystometry were significantly different in contusion and transection groups compared with normals, but again there was no significant difference between contusion and transection groups. Immunocytochemical analyses at the L6 spinal cord showed no serotonergic or noradrenergic fibers in transection group, but some descending fibers remained in contusion group, indicating sparing. Small dorsal root afferents were denser in both contusion and transection groups than in normals, indicating sprouting. CONCLUSIONS: Although differences were not found in LUT function in operated animals, supraspinal and dorsal root projections to the L6 spinal cord responded differently to contusion and transection. This suggests that the benefits of pharmacologic treatments may be different in two lesion models.


Subject(s)
Spinal Cord Injuries/complications , Urinary Tract/physiopathology , Animals , Contusions , Disease Models, Animal , Ganglia, Spinal/injuries , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Nerve Roots/injuries , Urination/physiology , Urodynamics
17.
Bull Exp Biol Med ; 157(5): 637-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25257430

ABSTRACT

The development of dissociated cells from rat embryonic spinal ganglion after transplantation to damaged nerve of adult animals was studied using immunohistochemical differentiation markers of neural and glial cells. The cell suspension obtained after dissociation of rat embryonic spinal ganglia (embryonic day 15) was injected into the proximal segment of crushed sciatic nerve. The nerve was damaged by ligation for 40 sec. Progenitor cells were labeled with 5-bromo-2'-deoxyuridine (BrdU) before transplantation. BrdU-immunopositive cells were detected in the nerve trunks of recipients on days 1, 21, and 28 after transplantation. Dissociated cells of rat embryonic spinal ganglion (embryonic day 15) survived for at least 4 weeks after transplantation to the nerve and differentiate into NeuN-immunopositive neurons with morphological properties of sensory neurons and satellite cells containing S100 protein.


Subject(s)
Ganglia, Spinal/cytology , Animals , Ganglia, Spinal/embryology , Ganglia, Spinal/injuries , Rats , Rats, Wistar
18.
J Neurosci ; 32(21): 7325-35, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623678

ABSTRACT

Growing and regenerating axons need to interact with the molecules in the extracellular matrix as they traverse through their environment. An important group of receptors that serve this function is the integrin superfamily of cell surface receptors, which are evolutionarily conserved αß heterodimeric transmembrane proteins. The function of integrins is controlled by regulating the affinity for ligands (also called "integrin activation"). Previous results have shown that CNS inhibitory molecules inactivate axonal integrins, while enhancing integrin activation can promote axon growth from neurons cultured on inhibitory substrates. We tested two related molecules, kindlin-1 and kindlin-2 (Fermitin family members 1 and 2), that can activate ß1, ß2, and ß3 integrins, for their effects on integrin signaling and integrin-mediated axon growth in rat sensory neurons. We determined that kindlin-2, but not kindlin-1, is endogenously expressed in the nervous system. Knocking down kindlin-2 levels in cultured sensory neurons impaired their ability to extend axons, but this was partially rescued by kindlin-1 expression. Overexpression of kindlin-1, but not kindlin-2, in cultured neurons increased axon growth on an inhibitory aggrecan substrate. This was found to be associated with enhanced integrin activation and signaling within the axons. Additionally, in an in vivo rat dorsal root injury model, transduction of dorsal root ganglion neurons to express kindlin-1 promoted axon regeneration across the dorsal root entry zone and into the spinal cord. These animals demonstrated improved recovery of thermal sensation following injury. Our results therefore suggest that kindlin-1 is a potential tool for improving axon regeneration after nervous system lesions.


Subject(s)
Aggrecans/pharmacology , Axons/physiology , Ganglia, Spinal/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/physiology , Retinal Ganglion Cells/physiology , Sensory Receptor Cells/physiology , Animals , Axons/metabolism , Brain/metabolism , Brain/physiology , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/injuries , Ganglia, Spinal/metabolism , Gene Knockdown Techniques , Hippocampus/metabolism , Integrins/metabolism , Laminin/pharmacology , Nerve Regeneration/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Primary Cell Culture , Purkinje Cells/metabolism , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
19.
J Neurosci ; 32(22): 7745-57, 2012 05 30.
Article in English | MEDLINE | ID: mdl-22649252

ABSTRACT

Following CNS injury, microglial phagocytosis of damaged endogenous tissue is thought to play an important role in recovery and regeneration. Previous work has focused on delineating mechanisms of clearance of neurons and myelin. Little, however, is known of the mechanisms underlying phagocytosis of axon debris. We have developed a novel microfluidic platform that enables coculture of microglia with bundles of CNS axons to investigate mechanisms of microglial phagocytosis of axons. Using this platform, we find that axon degeneration results in the induction of type-1 interferon genes within microglia. Pharmacologic and genetic disruption of Toll/interleukin-1 receptor domain-containing adapter inducing interferon-ß (TRIF), a Toll-like receptor adapter protein, blocks induction of the interferon response and inhibits microglial phagocytosis of axon debris in vitro. In vivo, microglial phagocytosis of axons following dorsal root axotomy is impaired in mice in which TRIF has been genetically deleted. Furthermore, we identify the p38 mitogen-activated protein kinase (MAPK) cascade as a signaling pathway downstream of TRIF following axon degeneration and find that inhibition of p38 MAPK by SB203580 (4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole) also blocked clearance of axon debris. Finally, we find that TRIF-dependent microglial clearance of unmyelinated axon debris facilitates axon outgrowth. Overall, we provide evidence that TRIF-mediated signaling plays an unexpected role in axonal debris clearance by microglia, thereby facilitating a more permissive environment for axonal outgrowth. Our study has significant implications for the development of novel regenerative and restorative strategies for the many traumatic, neuroinflammatory, and neurodegenerative conditions characterized by CNS axon degeneration.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Interferon-beta/metabolism , Microglia/physiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Phagocytosis/physiology , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/deficiency , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Animals, Newborn , Axons/pathology , Axotomy , CD11b Antigen/metabolism , Cells, Cultured , Chemokine CXCL10/metabolism , Coculture Techniques , Disease Models, Animal , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/injuries , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/cytology , Humans , Imidazoles/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Microfluidic Analytical Techniques , Nerve Degeneration/chemically induced , Neurons/cytology , Neurons/drug effects , Nitric Oxide/pharmacology , Peptides/pharmacology , Phagocytosis/genetics , Pyridines/pharmacology , Quaternary Ammonium Compounds/toxicity , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/metabolism , Time Factors , Transfection
20.
J Biol Chem ; 287(8): 6002-13, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22187436

ABSTRACT

High voltage-activated calcium channels (HVACCs) are essential for synaptic and nociceptive transmission. Although blocking HVACCs can effectively reduce pain, this treatment strategy is associated with intolerable adverse effects. Neuronal HVACCs are typically composed of α(1), ß (Cavß), and α(2)δ subunits. The Cavß subunit plays a crucial role in the membrane expression and gating properties of the pore-forming α(1) subunit. However, little is known about how nerve injury affects the expression and function of Cavß subunits in primary sensory neurons. In this study, we found that Cavß(3) and Cavß(4) are the most prominent subtypes expressed in the rat dorsal root ganglion (DRG) and dorsal spinal cord. Spinal nerve ligation (SNL) in rats significantly increased mRNA and protein levels of the Cavß(3), but not Cavß(4), subunit in the DRG. SNL also significantly increased HVACC currents in small DRG neurons and monosynaptic excitatory postsynaptic currents of spinal dorsal horn neurons evoked from the dorsal root. Intrathecal injection of Cavß(3)-specific siRNA significantly reduced HVACC currents in small DRG neurons and the amplitude of monosynaptic excitatory postsynaptic currents of dorsal horn neurons in SNL rats. Furthermore, intrathecal treatment with Cavß(3)-specific siRNA normalized mechanical hyperalgesia and tactile allodynia caused by SNL but had no significant effect on the normal nociceptive threshold. Our findings provide novel evidence that increased expression of the Cavß(3) subunit augments HVACC activity in primary sensory neurons and nociceptive input to dorsal horn neurons in neuropathic pain. Targeting the Cavß(3) subunit at the spinal level represents an effective strategy for treating neuropathic pain.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Neuralgia/metabolism , Neuralgia/pathology , Nociception , Sensory Receptor Cells/metabolism , Up-Regulation , Animals , Base Sequence , Down-Regulation , Ganglia, Spinal/injuries , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Glutamic Acid/metabolism , Male , Neuralgia/genetics , Neuralgia/physiopathology , Posterior Horn Cells/metabolism , Posterior Horn Cells/pathology , Protein Transport , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Sensory Receptor Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL