Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Brain ; 17(1): 25, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773624

ABSTRACT

A growing body of evidence indicates intra- and inter-regional heterogeneity of astrocytes in the brain. However, because of a lack of an efficient method for isolating astrocytes from the spinal cord, little is known about how much spinal cord astrocytes are heterogeneous in adult mice. In this study, we developed a new method for isolating spinal astrocytes from adult mice using a cold-active protease from Bacillus licheniformis with an astrocyte cell surface antigen-2 (ACSA-2) antibody. Using fluorescence-activated cell sorting, isolated spinal ACSA-2+ cells were divided into two distinct populations, ACSA-2high and ACSA-2low. By analyzing the expression of cell-type marker genes, the ACSA-2high and ACSA-2low populations were identified as astrocytes and ependymal cells, respectively. Furthermore, ACSA-2high cells had mRNAs encoding genes that were abundantly expressed in the gray matter (GM) but not white matter astrocytes. By optimizing enzymatic isolation procedures, the yield of GM astrocytes also increased. Therefore, our newly established method enabled the selective and efficient isolation of GM astrocytes from the spinal cord of adult mice and may be useful for bulk- or single-cell RNA-sequencing under physiological and pathological conditions.


Subject(s)
Astrocytes , Cell Separation , Gray Matter , Spinal Cord , Animals , Astrocytes/metabolism , Astrocytes/cytology , Spinal Cord/cytology , Cell Separation/methods , Mice, Inbred C57BL , Mice , Male , RNA, Messenger/metabolism , RNA, Messenger/genetics , Aging
2.
Int J Neuropsychopharmacol ; 26(11): 773-783, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-37715955

ABSTRACT

BACKGROUND: Cannabis edibles are an increasingly popular form of cannabis consumption. Oral consumption of cannabis has distinct physiological and behavioral effects compared with injection or inhalation. An animal model is needed to understand the pharmacokinetics and physiological effects of oral cannabis consumption in rodents as a model for human cannabis edible use. METHODS: Adult male and female C57BL/6 mice received a single dose of commercially available cannabis oil (5 mg/kg Δ9-tetrahydrocannabinol [THC]) by oral gavage. At 0.5, 1, 2, 3, and 6 hours post exposure, plasma, hippocampus, and adipose tissue were collected for THC, 11-OH-THC, and THC-COOH measures. RESULTS: We report delayed time to peak THC and 11-OH-THC concentrations in plasma, brain, and adipose tissue, which is consistent with human pharmacokinetics studies. We also found sex differences in the cannabis tetrad: (1) female mice had a delayed hypothermic effect 6 hours post consumption, which was not present in males; (2) females had stronger catalepsy than males; (3) males were less mobile following cannabis exposure, whereas female mice showed no difference in locomotion but an anxiogenic effect at 3 hours post exposure; and (4) male mice displayed a longer-lasting antinociceptive effect of oral cannabis. CONCLUSIONS: Oral cannabis consumption is a translationally relevant form of administration that produces similar physiological effects as injection or vaping administration and thus should be considered as a viable approach for examining the physiological effects of cannabis moving forward. Furthermore, given the strong sex differences in metabolism of oral cannabis, these factors should be carefully considered when designing animal studies on the effects of cannabis.


Subject(s)
Cannabinoids , Cannabis , Hallucinogens , Adult , Humans , Female , Male , Mice , Animals , Dronabinol/pharmacology , Sex Characteristics , Mice, Inbred C57BL , Cannabinoid Receptor Agonists , Adipose Tissue
3.
Brain Behav Immun ; 110: 276-287, 2023 05.
Article in English | MEDLINE | ID: mdl-36898418

ABSTRACT

Pain transmission and processing in the nervous system are modulated by various biologically active substances, including lysophospholipids, through direct and indirect actions on the somatosensory pathway. Lysophosphatidylglucoside (LysoPtdGlc) was recently identified as a structurally unique lysophospholipid that exerts biological actions via the G protein-coupled receptor GPR55. Here, we demonstrated that GPR55-knockout (KO) mice show impaired induction of mechanical pain hypersensitivity in a model of spinal cord compression (SCC) without the same change in the models of peripheral tissue inflammation and peripheral nerve injury. Among these models, only SCC recruited peripheral inflammatory cells (neutrophils, monocytes/macrophages, and CD3+ T-cells) in the spinal dorsal horn (SDH), and GPR55-KO blunted these recruitments. Neutrophils were the first cells recruited to the SDH, and their depletion suppressed the induction of SCC-induced mechanical hypersensitivity and inflammatory responses in compressed SDH. Furthermore, we found that PtdGlc was present in the SDH and that intrathecal administration of an inhibitor of secretory phospholipase A2 (an enzyme required for producing LysoPtdGlc from PtdGlc) reduced neutrophil recruitment to compressed SDH and suppressed pain induction. Finally, by screening compounds from a chemical library, we identified auranofin as a clinically used drug with an inhibitory effect on mouse and human GPR55. Systemically administered auranofin to mice with SCC effectively suppressed spinal neutrophil infiltration and pain hypersensitivity. These results suggest that GPR55 signaling contributes to the induction of inflammatory responses and chronic pain after SCC via the recruitment of neutrophils and may provide a new target for reducing pain induction after spinal cord compression, such as spinal canal stenosis.


Subject(s)
Chronic Pain , Spinal Cord Compression , Humans , Mice , Animals , Neutrophil Infiltration , Spinal Cord Compression/metabolism , Auranofin/metabolism , Spinal Cord Dorsal Horn/metabolism , Chronic Pain/metabolism , Spinal Cord/metabolism , Receptors, Cannabinoid/metabolism
4.
J Neurosci Res ; 100(1): 251-264, 2022 01.
Article in English | MEDLINE | ID: mdl-34075613

ABSTRACT

Opioids are potent analgesics, but their pain-relieving effects diminish with repeated use. The reduction in analgesic potency is a hallmark of opioid analgesic tolerance, which hampers opioid pain therapy. In the central nervous system, opioid analgesia is critically modulated by adenosine, a purine nucleoside implicated in the beneficial and detrimental actions of opioid medications. Here, we focus on the A3 adenosine receptor (A3 AR) in opioid analgesic tolerance. Intrathecal administration of the A3 AR agonist MRS5698 with daily systemic morphine in male rats attenuated the reduction in morphine antinociception over 7 days. In rats with established morphine tolerance, intrathecal MRS5698 partially restored the antinociceptive effects of morphine. However, when MRS5698 was discontinued, these animals displayed a reduced antinociceptive response to morphine. Our results suggest that MRS5698 acutely and transiently potentiates morphine antinociception in tolerant rats. By contrast, in morphine-naïve rats MRS5698 treatment did not impact thermal nociceptive threshold or affect antinociceptive response to a single injection of morphine. Furthermore, we found that morphine-induced adenosine release in cerebrospinal fluid was blunted in tolerant animals, but total spinal A3 AR expression was not affected. Collectively, our findings indicate that spinal A3 AR activation acutely potentiates morphine antinociception in the opioid tolerant state.


Subject(s)
Analgesics, Opioid , Morphine , Adenosine/metabolism , Adenosine/pharmacology , Analgesics, Opioid/pharmacology , Animals , Drug Tolerance , Injections, Spinal , Male , Morphine/pharmacology , Rats , Receptors, Purinergic P1/metabolism , Spinal Cord/metabolism
5.
Front Immunol ; 12: 626884, 2021.
Article in English | MEDLINE | ID: mdl-33897685

ABSTRACT

Increased afferent input resulting from painful injury augments the activity of central nociceptive circuits via both neuron-neuron and neuron-glia interactions. Microglia, resident immune cells of the central nervous system (CNS), play a crucial role in the pathogenesis of chronic pain. This study provides a framework for understanding how peripheral joint injury signals the CNS to engage spinal microglial responses. During the first week of monosodium iodoacetate (MIA)-induced knee joint injury in male rats, inflammatory and neuropathic pain were characterized by increased firing of peripheral joint afferents. This increased peripheral afferent activity was accompanied by increased Iba1 immunoreactivity within the spinal dorsal horn indicating microglial activation. Pharmacological silencing of C and A afferents with co-injections of QX-314 and bupivacaine, capsaicin, or flagellin prevented the development of mechanical allodynia and spinal microglial activity after MIA injection. Elevated levels of ATP in the cerebrospinal fluid (CSF) and increased expression of the ATP transporter vesicular nucleotide transporter (VNUT) in the ipsilateral spinal dorsal horn were also observed after MIA injections. Selective silencing of primary joint afferents subsequently inhibited ATP release into the CSF. Furthermore, increased spinal microglial reactivity, and alleviation of MIA-induced arthralgia with co-administration of QX-314 with bupivacaine were recapitulated in female rats. Our results demonstrate that early peripheral joint injury activates joint nociceptors, which triggers a central spinal microglial response. Elevation of ATP in the CSF, and spinal expression of VNUT suggest ATP signaling may modulate communication between sensory neurons and spinal microglia at 2 weeks of joint degeneration.


Subject(s)
Arthritis, Experimental/physiopathology , Microglia/physiology , Neurons, Afferent/physiology , Spinal Cord/physiopathology , Adenosine Triphosphate/physiology , Animals , Arthralgia/therapy , Disease Models, Animal , Female , Hyperalgesia/physiopathology , Iodoacetic Acid/pharmacology , Male , Rats , Rats, Sprague-Dawley
6.
Nat Neurosci ; 23(11): 1376-1387, 2020 11.
Article in English | MEDLINE | ID: mdl-33020652

ABSTRACT

Astrocytes are critical regulators of CNS function and are proposed to be heterogeneous in the developing brain and spinal cord. Here we identify a population of astrocytes located in the superficial laminae of the spinal dorsal horn (SDH) in adults that is genetically defined by Hes5. In vivo imaging revealed that noxious stimulation by intraplantar capsaicin injection activated Hes5+ SDH astrocytes via α1A-adrenoceptors (α1A-ARs) through descending noradrenergic signaling from the locus coeruleus. Intrathecal norepinephrine induced mechanical pain hypersensitivity via α1A-ARs in Hes5+ astrocytes, and chemogenetic stimulation of Hes5+ SDH astrocytes was sufficient to produce the hypersensitivity. Furthermore, capsaicin-induced mechanical hypersensitivity was prevented by the inhibition of descending locus coeruleus-noradrenergic signaling onto Hes5+ astrocytes. Moreover, in a model of chronic pain, α1A-ARs in Hes5+ astrocytes were critical regulators for determining an analgesic effect of duloxetine. Our findings identify a superficial SDH-selective astrocyte population that gates descending noradrenergic control of mechanosensory behavior.


Subject(s)
Astrocytes/physiology , Hyperalgesia/physiopathology , Locus Coeruleus/physiology , Neurons/physiology , Nociception/physiology , Spinal Cord Dorsal Horn/physiology , Adrenergic Neurons/physiology , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/analysis , Female , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neural Pathways/physiology , Receptors, Adrenergic, alpha-1/physiology , Repressor Proteins/analysis , Spinal Cord Dorsal Horn/metabolism
7.
Brain Behav Immun ; 89: 389-399, 2020 10.
Article in English | MEDLINE | ID: mdl-32717400

ABSTRACT

Chronic pain is one of the main symptoms of spinal disorders such as spinal canal stenosis. A major cause of this pain is related to compression of the spinal cord, and chronic pain can develop at the level of the compressed spinal segment. However, in many patients chronic pain arises in an area that does not correspond to the compressed segment, and the underlying mechanism involved remains unknown. This was investigated in the present study using a mouse model of spinal cord compression in which mechanical pain of the hindpaws develops after compression of the first lumbar segment (L1) of the spinal cord. Compression induced the activation of astrocytes in the L1 spinal dorsal horn (SDH)-but not the L4 SDH that corresponds to the hindpaws-and activated signal transducer and activator of transcription 3 (STAT3). Suppressing reactive astrocytes by expressing a dominant negative form of STAT3 (dnSTAT3) in the compressed SDH prevented mechanical pain. Expression of interleukin (IL)-6 was also upregulated in the compressed SDH, and it was inhibited by astrocytic expression of dnSTAT3. Intrathecal administration of a neutralizing anti-IL-6 antibody reversed the compression-induced mechanical pain. These results suggest that astrocytic STAT3 and IL-6 in the compressed SDH are involved in remote mechanical pain observed in the lower extremity, and may provide a target for treating chronic pain associated with spinal cord compression such as spinal canal stenosis.


Subject(s)
Interleukin-6 , STAT3 Transcription Factor , Astrocytes/metabolism , Humans , Hyperalgesia , Interleukin-6/metabolism , Lower Extremity , Pain , STAT3 Transcription Factor/metabolism , Spinal Cord/metabolism
8.
J Allergy Clin Immunol ; 145(1): 183-191.e10, 2020 01.
Article in English | MEDLINE | ID: mdl-31787267

ABSTRACT

BACKGROUND: Chronic itch is a highly debilitating symptom among patients with inflammatory skin diseases. Recent studies have revealed that gastrin-releasing peptide (GRP) and its receptor (gastrin-releasing peptide receptor [GRPR]) in the spinal dorsal horn (SDH) play a central role in itch transmission. OBJECTIVE: We aimed to investigate whether GRP-GRPR signaling is altered in SDH neurons in a mouse model of chronic itch and to determine the potential mechanisms underlying these alterations. METHODS: Patch-clamp recordings from enhanced green fluorescent protein (EGFP)-expressing (GRPR+) SDH neurons were used to examine GRP-GRPR signaling in spinal cord slices obtained from Grpr-EGFP mice. Immunohistochemical, genetic (gene expression and editing through adeno-associated virus vectors), and behavioral approaches were also used for in vivo experiments. RESULTS: We observed potentiation of GRP-evoked excitation in the GRPR+ SDH neurons of mice with contact dermatitis, without concomitant changes in GRPR expression. Interestingly, increases in excitation were attenuated by suppressing the reactive state of SDH astrocytes, which are known to be reactive in patients with chronic itch conditions. Furthermore, CRISPR-Cas9-mediated astrocyte-selective in vivo editing of a gene encoding lipocalin-2 (LCN2), an astrocytic factor implicated in chronic itch, suppressed increases in GRP-induced excitation of GRPR+ neurons, repetitive scratching, and skin damage in mice with contact dermatitis. Moreover, LCN2 potentiated GRP-induced excitation of GRPR+ neurons in normal mice. CONCLUSION: Our findings indicate that, under chronic itch conditions, the GRP-induced excitability of GRPR+ SDH neurons is enhanced through a non-cell-autonomous mechanism involving LCN2 derived from reactive astrocytes.


Subject(s)
Astrocytes/immunology , Gastrin-Releasing Peptide/immunology , Posterior Horn Cells/immunology , Pruritus/immunology , Receptors, Bombesin/immunology , Signal Transduction/immunology , Animals , Astrocytes/pathology , Chronic Disease , Disease Models, Animal , Gastrin-Releasing Peptide/genetics , Male , Mice , Mice, Transgenic , Posterior Horn Cells/pathology , Pruritus/genetics , Pruritus/pathology , Receptors, Bombesin/genetics , Signal Transduction/genetics
9.
Biol Pharm Bull ; 41(7): 1096-1102, 2018.
Article in English | MEDLINE | ID: mdl-29962405

ABSTRACT

Neuropathic pain, a highly debilitating chronic pain following nerve damage, is a reflection of the aberrant functioning of a pathologically altered nervous system. Previous studies have implicated activated microglia in the spinal dorsal horn (SDH) as key cellular intermediaries in neuropathic pain. Microgliosis is among the dramatic cellular alterations that occur in the SDH in models of neuropathic pain established by peripheral nerve injury (PNI), but detailed characterization of SDH microgliosis has yet to be realized. In the present study, we performed a short-pulse labeling of proliferating cells with ethynyldeoxyuridine (EdU), a marker of the cell cycle S-phase, and found that EdU+ microglia in the SDH were rarely observed 32 h after PNI, but rapidly increased to the peak level at 40 h post-PNI. Numerous EdU+ microglia persisted for the next 20 h (60 h post-PNI) and decreased to the baseline on day 7. These results demonstrate a narrow time window for rapidly inducing a proliferation burst of SDH microglia after PNI, and these temporally restricted kinetics of microglial proliferation may help identify the molecule that causes microglial activation in the SDH, which is crucial for understanding and managing neuropathic pain.


Subject(s)
Gliosis/physiopathology , Microglia/pathology , Neuralgia/physiopathology , Peripheral Nerve Injuries/physiopathology , Spinal Cord Dorsal Horn/pathology , Animals , Cell Proliferation , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Neuralgia/etiology , Peripheral Nerve Injuries/complications , Rats , Rats, Wistar , Spinal Cord Dorsal Horn/cytology , Time Factors
10.
J Pharmacol Sci ; 137(1): 101-104, 2018 May.
Article in English | MEDLINE | ID: mdl-29773517

ABSTRACT

The role of astrocytes in the spinal dorsal horn (SDH) for sensory information processing under normal conditions is poorly understood. In this study, we investigated whether SDH astrocytes respond to noxious and innocuous stimuli to the skin of normal mice using in vivo two-photon Ca2+ imaging under anesthesia. We found that noxious stimulation evoked by intraplantar formalin injection provoked an elevation in intracellular Ca2+ levels in SDH astrocytes. By contrast, neither instantaneous noxious pinching nor innocuous stimuli (cooling or brushing) to the hindpaw elicited astrocytic Ca2+ responses. Thus, SDH astrocytes could respond preferentially to a strong and/or sustained noxious stimulus.


Subject(s)
Astrocytes/metabolism , Astrocytes/physiology , Calcium/metabolism , Formaldehyde/adverse effects , Sensation/drug effects , Sensation/physiology , Skin Physiological Phenomena , Skin/drug effects , Spinal Cord Dorsal Horn/cytology , Animals , Formaldehyde/administration & dosage , Injections, Subcutaneous , Male , Mice, Inbred C57BL , Stimulation, Chemical
11.
eNeuro ; 5(1)2018.
Article in English | MEDLINE | ID: mdl-29468190

ABSTRACT

Neuropathic pain is caused by peripheral nerve injury (PNI). One hallmark symptom is allodynia (pain caused by normally innocuous stimuli), but its mechanistic underpinning remains elusive. Notably, whether selective stimulation of non-nociceptive primary afferent Aß fibers indeed evokes neuropathic pain-like sensory and emotional behaviors after PNI is unknown, because of the lack of tools to manipulate Aß fiber function in awake, freely moving animals. In this study, we used a transgenic rat line that enables stimulation of non-nociceptive Aß fibers by a light-activated channel (channelrhodopsin-2; ChR2). We found that illuminating light to the plantar skin of these rats with PNI elicited pain-like withdrawal behaviors that were resistant to morphine. Light illumination to the skin of PNI rats increased the number of spinal dorsal horn (SDH) Lamina I neurons positive to activity markers (c-Fos and phosphorylated extracellular signal-regulated protein kinase; pERK). Whole-cell recording revealed that optogenetic Aß fiber stimulation after PNI caused excitation of Lamina I neurons, which were normally silent by this stimulation. Moreover, illuminating the hindpaw of PNI rats resulted in activation of central amygdaloid neurons and produced an aversion to illumination. Thus, these findings provide the first evidence that optogenetic activation of primary afferent Aß fibers in PNI rats produces excitation of Lamina I neurons and neuropathic pain-like behaviors that were resistant to morphine treatment. This approach may provide a new path for investigating circuits and behaviors of Aß fiber-mediated neuropathic allodynia with sensory and emotional aspects after PNI and for discovering novel drugs to treat neuropathic pain.


Subject(s)
Emotions/physiology , Neuralgia/physiopathology , Neuralgia/psychology , Neurons, Afferent/physiology , Spinal Nerves/injuries , Animals , Avoidance Learning/physiology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Conditioning, Psychological/physiology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Lumbar Vertebrae , Male , Neuralgia/etiology , Neuralgia/pathology , Neurons, Afferent/pathology , Optogenetics/methods , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , Rats, Transgenic , Skin/physiopathology , Spinal Nerves/pathology , Spinal Nerves/physiopathology , Tissue Culture Techniques
12.
Sci Rep ; 7(1): 4739, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28680103

ABSTRACT

Inhibitory interneurons in the spinal dorsal horn (SDH) are crucial for processing somatosensory information originating in the periphery. However, the effects of the acute and selective inactivation of GABAergic SDH interneurons on pain processing are not fully understood. In this study, we used designer receptors exclusively activated by designer drugs (DREADD) technology and vesicular GABA transporter-Cre (Vgat-Cre) mice to selectively express a modified human muscarinic Gi protein-coupled receptor (hM4Di) in Vgat-Cre + GABAergic SDH interneurons in the fourth lumbar segment. We found that clozapine-N-oxide (CNO) treatment rapidly hyperpolarized these neurons and induced spontaneous nocifensive behaviours in these mice. In Vgat-Cre neg lamina II neurons, CNO produced facilitation of A fibre-mediated polysynaptic excitatory responses, an effect that required N-methyl-D-aspartate (NMDA) receptor activation. The CNO-induced nocifensive behaviours were also reduced by NMDA receptor antagonism. Moreover, these nocifensive behaviours were suppressed by pregabalin but resistant to morphine. Our findings indicate that Vgat-Cre + SDH interneurons play an important role in morphine-resistant nocifensive behaviours and suggest that this approach may provide a useful model for understanding the mechanisms of opioid-resistant pain signalling and for developing novel analgesics.


Subject(s)
Clozapine/analogs & derivatives , GABAergic Neurons/drug effects , Interneurons/drug effects , Spinal Cord Dorsal Horn/drug effects , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , Cell Polarity/drug effects , Clozapine/administration & dosage , Clozapine/pharmacology , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Interneurons/cytology , Interneurons/metabolism , Mice , Morphine/pharmacology , Pregabalin/administration & dosage , Pregabalin/pharmacology , Spinal Cord Dorsal Horn/cytology , Spinal Cord Dorsal Horn/metabolism
14.
Bio Protoc ; 7(9): e2252, 2017 May 05.
Article in English | MEDLINE | ID: mdl-34541241

ABSTRACT

Neuropathic pain is one of the highly debilitating chronic pain conditions, for which, currently, there is no therapeutic treatment. In order to reveal the underlying mechanism for neuropathic pain, various animal models have been established ( Burma et al., 2016 ). This protocol describes how to prepare spinal nerve injury model (Kim and Chung, 1992; Rigaud et al., 2008 ; Masuda et al., 2016 ), one of the most frequently-used and highly reproducible models in which multiple alterations occur both in the peripheral and central nervous system.

15.
Nat Commun ; 7: 13102, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739425

ABSTRACT

Diurnal variations in pain hypersensitivity are common in chronic pain disorders, but the underlying mechanisms are enigmatic. Here, we report that mechanical pain hypersensitivity in sciatic nerve-injured mice shows pronounced diurnal alterations, which critically depend on diurnal variations in glucocorticoids from the adrenal glands. Diurnal enhancement of pain hypersensitivity is mediated by glucocorticoid-induced enhancement of the extracellular release of ATP in the spinal cord, which stimulates purinergic receptors on microglia in the dorsal horn. We identify serum- and glucocorticoid-inducible kinase-1 (SGK-1) as the key molecule responsible for the glucocorticoid-enhanced release of ATP from astrocytes. SGK-1 protein levels in spinal astrocytes are increased in response to glucocorticoid stimuli and enhanced ATP release by opening the pannexin-1 hemichannels. Our findings reveal an unappreciated circadian machinery affecting pain hypersensitivity caused by peripheral nerve injury, thus opening up novel approaches to the management of chronic pain.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/drug effects , Glucocorticoids/pharmacology , Spinal Cord/drug effects , Adrenalectomy , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Circadian Rhythm , Corticosterone/blood , Corticosterone/pharmacology , Gene Expression Profiling , Glucocorticoids/blood , Hyperalgesia/physiopathology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Ligation , Male , Mice, Inbred ICR , Neuralgia/physiopathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sciatic Nerve/surgery , Spinal Cord/cytology , Spinal Cord/metabolism
16.
Nat Commun ; 7: 12529, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27515581

ABSTRACT

Activation of purinergic receptors in the spinal cord by extracellular ATP is essential for neuropathic hypersensitivity after peripheral nerve injury (PNI). However, the cell type responsible for releasing ATP within the spinal cord after PNI is unknown. Here we show that PNI increases expression of vesicular nucleotide transporter (VNUT) in the spinal cord. Extracellular ATP content ([ATP]e) within the spinal cord was increased after PNI, and this increase was suppressed by exocytotic inhibitors. Mice lacking VNUT did not show PNI-induced increase in [ATP]e and had attenuated hypersensitivity. These phenotypes were recapitulated in mice with specific deletion of VNUT in spinal dorsal horn (SDH) neurons, but not in mice lacking VNUT in primary sensory neurons, microglia or astrocytes. Conversely, ectopic VNUT expression in SDH neurons of VNUT-deficient mice restored PNI-induced increase in [ATP]e and pain. Thus, VNUT is necessary for exocytotic ATP release from SDH neurons which contributes to neuropathic pain.


Subject(s)
Adenosine Triphosphate/metabolism , Neuralgia/pathology , Nucleotide Transport Proteins/metabolism , Peripheral Nerve Injuries/pathology , Posterior Horn Cells/pathology , Animals , Astrocytes/metabolism , Disease Models, Animal , Exocytosis/drug effects , Exocytosis/physiology , Female , Humans , Hypersensitivity/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Neuralgia/etiology , Nucleotide Transport Proteins/genetics , Peripheral Nerve Injuries/etiology , Posterior Horn Cells/metabolism , Sensory Receptor Cells/metabolism , Tetanus Toxin/pharmacology
17.
Sci Rep ; 6: 23701, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27005516

ABSTRACT

Accumulating evidence indicates that peripheral nerve injury (PNI) activates spinal microglia that are necessary for neuropathic pain. Recent studies using bone marrow (BM) chimeric mice have reported that after PNI, circulating BM-derived cells infiltrate into the spinal cord and differentiate into microglia-like cells. This raises the possibility that the population of spinal microglia after PNI may be heterogeneous. However, the infiltration of BM cells in the spinal cord remains controversial because of experimental adverse effects of strong irradiation used for generating BM chimeric mice. In this study, we evaluated the PNI-induced spinal infiltration of BM-derived cells not only by irradiation-induced myeloablation with various conditioning regimens, but also by parabiosis and mice with genetically labelled microglia, models without irradiation and BM transplantation. Results obtained from these independent approaches provide compelling evidence indicating little contribution of circulating BM-derived cells to the population of spinal microglia after PNI.


Subject(s)
Bone Marrow Cells/cytology , Microglia/pathology , Neuralgia/etiology , Peripheral Nerve Injuries/complications , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Bone Marrow Transplantation , Disease Models, Animal , Gamma Rays/adverse effects , Male , Mice , Microglia/cytology , Microglia/metabolism , Peripheral Nerve Injuries/etiology
18.
Sci Rep ; 5: 14306, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26387932

ABSTRACT

Noninvasive gene delivery to the spinal dorsal horn (SDH) remains challenging because existing methods to directly microinject vectors require laminectomy, which leads to tissue damage and inflammation. Such responses might hamper accurate readouts of cellular and behavioural effects of an introduced gene. Here we develop a new minimally-invasive SDH microinjection technique without the need of laminectomy in which a microcapillary is inserted into the SDH parenchyma through an intervertebral space. Using this method, we microinjected adeno-associated virus with an astrocytic promoter into the SDH and achieved efficient gene expression in an astrocyte-specific manner without gliosis, neuronal loss or inflammation. Furthermore, astrocytic loss- and gain-of-function of the transcription factor STAT3 by expressing a dominant-negative form and a constitutive-active form of STAT3, respectively, demonstrated the necessity and sufficiency of astrocytic STAT3 in the maintenance of neuropathic pain following peripheral nerve injury, a debilitating chronic pain state in which currently available treatments are frequently ineffective. Thus, our technique enables manipulation of gene expression in cell type- and spatial-specific manners without adverse effects, and may be useful for research in SDH physiology and pathology.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Microinjections/methods , Spinal Cord Dorsal Horn , Animals , Astrocytes/metabolism , Female , Male , Mice , STAT3 Transcription Factor/genetics , Transgenes
19.
Nat Med ; 21(8): 927-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26193341

ABSTRACT

Chronic itch is an intractable symptom of inflammatory skin diseases, such as atopic and contact dermatitis. Recent studies have revealed neuronal pathways selective for itch, but the mechanisms by which itch turns into a pathological chronic state are poorly understood. Using mouse models of atopic and contact dermatitis, we demonstrate a long-term reactive state of astrocytes in the dorsal horn of the spinal segments that corresponds to lesioned, itchy skin. We found that reactive astrogliosis depended on the activation of signal transducer and activator of transcription 3 (STAT3). Conditional disruption of astrocytic STAT3 suppressed chronic itch, and pharmacological inhibition of spinal STAT3 ameliorated the fully developed chronic itch. Mice with atopic dermatitis exhibited an increase in scratching elicited by intrathecal administration of the itch-inducer gastrin-releasing peptide (GRP), and this enhancement was normalized by suppressing STAT3-mediated reactive astrogliosis. Moreover, we identified lipocalin-2 (LCN2) as an astrocytic STAT3-dependent upregulated factor that was crucial for chronic itch, and we demonstrated that intrathecal administration of LCN2 to normal mice increased spinal GRP-evoked scratching. Our findings indicate that STAT3-dependent reactive astrocytes act as critical amplifiers of itching through a mechanism involving the enhancement of spinal itch signals by LCN2, thereby providing a previously unrecognized target for treating chronic itch.


Subject(s)
Pruritus/etiology , STAT3 Transcription Factor/physiology , Spinal Cord Dorsal Horn/pathology , Acute-Phase Proteins/physiology , Animals , Astrocytes/physiology , Chronic Disease , Gastrin-Releasing Peptide/physiology , Lipocalin-2 , Lipocalins/physiology , Male , Mice , Mice, Inbred C57BL , Oncogene Proteins/physiology
20.
Brain ; 134(Pt 4): 1127-39, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21371995

ABSTRACT

Neuropathic pain, a debilitating pain condition, is a common consequence of damage to the nervous system. Optimal treatment of neuropathic pain is a major clinical challenge because the underlying mechanisms remain unclear and currently available treatments are frequently ineffective. Emerging lines of evidence indicate that peripheral nerve injury converts resting spinal cord glia into reactive cells that are required for the development and maintenance of neuropathic pain. However, the mechanisms underlying reactive astrogliosis after nerve injury are largely unknown. In the present study, we investigated cell proliferation, a critical process in reactive astrogliosis, and determined the temporally restricted proliferation of dorsal horn astrocytes in rats with spinal nerve injury, a well-known model of neuropathic pain. We found that nerve injury-induced astrocyte proliferation requires the Janus kinase-signal transducers and activators of transcription 3 signalling pathway. Nerve injury induced a marked signal transducers and activators of transcription 3 nuclear translocation, a primary index of signal transducers and activators of transcription 3 activation, in dorsal horn astrocytes. Intrathecally administering inhibitors of Janus kinase-signal transducers and activators of transcription 3 signalling to rats with nerve injury reduced the number of proliferating dorsal horn astrocytes and produced a recovery from established tactile allodynia, a cardinal symptom of neuropathic pain that is characterized by pain hypersensitivity evoked by innocuous stimuli. Moreover, recovery from tactile allodynia was also produced by direct suppression of dividing astrocytes by intrathecal administration of the cell cycle inhibitor flavopiridol to nerve-injured rats. Together, these results imply that the Janus kinase-signal transducers and activators of transcription 3 signalling pathway are critical transducers of astrocyte proliferation and maintenance of tactile allodynia and may be a therapeutic target for neuropathic pain.


Subject(s)
Astrocytes/metabolism , Cell Proliferation , Janus Kinases/metabolism , Neuralgia/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Spinal Nerves/injuries , Analysis of Variance , Animals , Behavior, Animal/physiology , Immunohistochemistry , Male , Neuralgia/etiology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Spinal Nerves/metabolism , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...