Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nature ; 534(7608): 494-9, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27281198

ABSTRACT

Voltage-gated sodium (Nav) channels initiate action potentials in most neurons, including primary afferent nerve fibres of the pain pathway. Local anaesthetics block pain through non-specific actions at all Nav channels, but the discovery of selective modulators would facilitate the analysis of individual subtypes of these channels and their contributions to chemical, mechanical, or thermal pain. Here we identify and characterize spider (Heteroscodra maculata) toxins that selectively activate the Nav1.1 subtype, the role of which in nociception and pain has not been elucidated. We use these probes to show that Nav1.1-expressing fibres are modality-specific nociceptors: their activation elicits robust pain behaviours without neurogenic inflammation and produces profound hypersensitivity to mechanical, but not thermal, stimuli. In the gut, high-threshold mechanosensitive fibres also express Nav1.1 and show enhanced toxin sensitivity in a mouse model of irritable bowel syndrome. Together, these findings establish an unexpected role for Nav1.1 channels in regulating the excitability of sensory nerve fibres that mediate mechanical pain.


Subject(s)
NAV1.1 Voltage-Gated Sodium Channel/metabolism , Nociception/drug effects , Nociceptors/drug effects , Nociceptors/metabolism , Spider Venoms/pharmacology , Stress, Mechanical , Animals , Disease Models, Animal , Female , Ganglia, Sensory/cytology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Irritable Bowel Syndrome/metabolism , Male , Myelin Sheath/metabolism , NAV1.1 Voltage-Gated Sodium Channel/chemistry , Nerve Fibers/drug effects , Nerve Fibers/metabolism , Oocytes/metabolism , Pain/chemically induced , Pain/metabolism , Protein Structure, Tertiary , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Spiders/chemistry , Substrate Specificity/drug effects , Temperature
2.
J Allergy Clin Immunol ; 140(2): 454-464.e2, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28232084

ABSTRACT

BACKGROUND: Despite recent insights into the pathophysiology of acute and chronic itch, chronic itch remains an often intractable condition. Among major contributors to chronic itch is dysfunction of spinal cord gamma aminobutyric acidergic (GABAergic) inhibitory controls. OBJECTIVES: We sought to test the hypothesis that selective GABA agonists as well as cell transplant-derived GABA are antipruritic against acute itch and in a transgenic mouse model of atopic dermatitis produced by overexpression of the TH2 cell-associated cytokine, IL-31 (IL-31Tg mice). METHODS: We injected wild-type and IL-31Tg mice with combinations of GABA-A (muscimol) or GABA-B (baclofen) receptor agonists 15 to 20 minutes prior to injection of various pruritogens (histamine, chloroquine, or endothelin-1) and recorded spontaneous scratching before and after drug administration. We also tested the antipruritic properties of intraspinal transplantation of precursors of GABAergic interneurons in the IL-31Tg mice. RESULTS: Systemic muscimol or baclofen are antipruritic against both histamine-dependent and -independent pruritogens, but the therapeutic window using either ligand alone was very small. In contrast, combined subthreshold doses of baclofen and muscimol produced a significant synergistic antipruritic effect, with no sedation. Finally, transplant-mediated long-term enhancement of GABAergic signaling not only reduced spontaneous scratching in the IL-31Tg mice but also dramatically resolved the associated skin lesions. CONCLUSIONS: Although additional research is clearly needed, existing approved GABA agonists should be considered in the management of chronic itch, notably atopic dermatitis.


Subject(s)
Antipruritics/therapeutic use , Baclofen/therapeutic use , Dermatitis, Atopic/drug therapy , GABA-A Receptor Agonists/therapeutic use , GABA-B Receptor Agonists/therapeutic use , Muscimol/therapeutic use , Animals , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/therapy , Disease Models, Animal , Drug Synergism , Gastrin-Releasing Peptide/genetics , Glutamate Decarboxylase/genetics , Interleukins/genetics , Interneurons/drug effects , Male , Median Eminence/cytology , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, Bombesin/genetics , Receptors, GABA-A/genetics , Receptors, GABA-B/genetics , Receptors, Neurokinin-1/genetics , Skin/drug effects , Skin/pathology , Spinal Cord/drug effects , Spinal Cord/metabolism , Stem Cell Transplantation
3.
J Neurosci ; 36(46): 11634-11645, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27852772

ABSTRACT

Spinal cord transplants of embryonic cortical GABAergic progenitor cells derived from the medial ganglionic eminence (MGE) can reverse mechanical hypersensitivity in the mouse models of peripheral nerve injury- and paclitaxel-induced neuropathic pain. Here, we used electrophysiology, immunohistochemistry, and electron microscopy to examine the extent to which MGE cells integrate into host circuitry and recapitulate endogenous inhibitory circuits. Whether the transplants were performed before or after nerve injury, the MGE cells developed into mature neurons and exhibited firing patterns characteristic of subpopulations of cortical and spinal cord inhibitory interneurons. Conversely, the transplanted cells preserved cortical morphological and neurochemical properties. We also observed a robust anatomical and functional synaptic integration of the transplanted cells into host circuitry in both injured and uninjured animals. The MGE cells were activated by primary afferents, including TRPV1-expressing nociceptors, and formed GABAergic, bicuculline-sensitive, synapses onto host neurons. Unexpectedly, MGE cells transplanted before injury prevented the development of mechanical hypersensitivity. Together, our findings provide direct confirmation of an extensive, functional synaptic integration of MGE cells into host spinal cord circuits. This integration underlies normalization of the dorsal horn inhibitory tone after injury and may be responsible for the prophylactic effect of preinjury transplants. SIGNIFICANCE STATEMENT: Spinal cord transplants of embryonic cortical GABAergic interneuron progenitors from the medial ganglionic eminence (MGE), can overcome the mechanical hypersensitivity produced in different neuropathic pain models in adult mice. Here, we examined the properties of transplanted MGE cells and the extent to which they integrate into spinal cord circuitry. Using electrophysiology, immunohistochemistry, and electron microscopy, we demonstrate that MGE cells, whether transplanted before or after nerve injury, develop into inhibitory neurons, are activated by nociceptive primary afferents, and form GABA-A-mediated inhibitory synapses with the host. Unexpectedly, cells transplanted into naive spinal cord prevented the development of nerve-injury-induced mechanical hypersensitivity. These results illustrate the remarkable plasticity of adult spinal cord and the potential of cell-based therapies against neuropathic pain.


Subject(s)
GABAergic Neurons/pathology , Hyperalgesia/physiopathology , Hyperalgesia/therapy , Neural Stem Cells/transplantation , Spinal Cord Regeneration/physiology , Spinal Cord/physiology , Synapses/pathology , Animals , GABAergic Neurons/metabolism , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Prosencephalon/cytology , Stem Cell Transplantation/methods , Synapses/metabolism , Treatment Outcome
4.
Nature ; 462(7273): 651-5, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19915548

ABSTRACT

Mechanical pain contributes to the morbidity associated with inflammation and trauma, but primary sensory neurons that convey the sensation of acute and persistent mechanical pain have not been identified. Dorsal root ganglion (DRG) neurons transmit sensory information to the spinal cord using the excitatory transmitter glutamate, a process that depends on glutamate transport into synaptic vesicles for regulated exocytotic release. Here we report that a small subset of cells in the DRG expresses the low abundance vesicular glutamate transporter VGLUT3 (also known as SLC17A8). In the dorsal horn of the spinal cord, these afferents project to lamina I and the innermost layer of lamina II, which has previously been implicated in persistent pain caused by injury. Because the different VGLUT isoforms generally have a non-redundant pattern of expression, we used Vglut3 knockout mice to assess the role of VGLUT3(+) primary afferents in the behavioural response to somatosensory input. The loss of VGLUT3 specifically impairs mechanical pain sensation, and in particular the mechanical hypersensitivity to normally innocuous stimuli that accompanies inflammation, nerve injury and trauma. Direct recording from VGLUT3(+) neurons in the DRG further identifies them as a poorly understood population of unmyelinated, low threshold mechanoreceptors (C-LTMRs). The analysis of Vglut3(-/-) mice now indicates a critical role for C-LTMRs in the mechanical hypersensitivity caused by injury.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Ganglia, Spinal/metabolism , Hypersensitivity/genetics , Hypersensitivity/physiopathology , Mechanoreceptors/physiology , Pain/genetics , Wounds and Injuries/physiopathology , Amino Acid Transport Systems, Acidic/genetics , Animals , Behavior, Animal/physiology , Female , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
J Allergy Clin Immunol ; 133(2): 448-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24373353

ABSTRACT

BACKGROUND: Although the cytokine IL-31 has been implicated in inflammatory and lymphoma-associated itch, the cellular basis for its pruritic action is yet unclear. OBJECTIVE: We sought to determine whether immune cell-derived IL-31 directly stimulates sensory neurons and to identify the molecular basis of IL-31-induced itch. METHODS: We used immunohistochemistry and quantitative real-time PCR to determine IL-31 expression levels in mice and human subjects. Immunohistochemistry, immunofluorescence, quantitative real-time PCR, in vivo pharmacology, Western blotting, single-cell calcium imaging, and electrophysiology were used to examine the distribution, functionality, and cellular basis of the neuronal IL-31 receptor α in mice and human subjects. RESULTS: Among all immune and resident skin cells examined, IL-31 was predominantly produced by TH2 and, to a significantly lesser extent, mature dendritic cells. Cutaneous and intrathecal injections of IL-31 evoked intense itch, and its concentrations increased significantly in murine atopy-like dermatitis skin. Both human and mouse dorsal root ganglia neurons express IL-31RA, largely in neurons that coexpress transient receptor potential cation channel vanilloid subtype 1 (TRPV1). IL-31-induced itch was significantly reduced in TRPV1-deficient and transient receptor channel potential cation channel ankyrin subtype 1 (TRPA1)-deficient mice but not in c-kit or proteinase-activated receptor 2 mice. In cultured primary sensory neurons IL-31 triggered Ca(2+) release and extracellular signal-regulated kinase 1/2 phosphorylation, inhibition of which blocked IL-31 signaling in vitro and reduced IL-31-induced scratching in vivo. CONCLUSION: IL-31RA is a functional receptor expressed by a small subpopulation of IL-31RA(+)/TRPV1(+)/TRPA1(+) neurons and is a critical neuroimmune link between TH2 cells and sensory nerves for the generation of T cell-mediated itch. Thus targeting neuronal IL-31RA might be effective in the management of TH2-mediated itch, including atopic dermatitis and cutaneous T-cell lymphoma.


Subject(s)
Interleukins/immunology , Pruritus/immunology , Receptors, Interleukin/immunology , Th2 Cells/immunology , Animals , Calcium Channels/immunology , Cells, Cultured , Female , Ganglia, Spinal/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/immunology , Receptors, Interleukin/genetics , Sensory Receptor Cells/immunology , Skin/immunology , TRPA1 Cation Channel , TRPV Cation Channels/genetics , TRPV Cation Channels/immunology , Transient Receptor Potential Channels/immunology
6.
Proc Natl Acad Sci U S A ; 107(51): 22296-301, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21135246

ABSTRACT

Dorsal root ganglia (DRG) neurons, including the nociceptors that detect painful thermal, mechanical, and chemical stimuli, transmit information to spinal cord neurons via glutamatergic and peptidergic neurotransmitters. However, the specific contribution of glutamate to pain generated by distinct sensory modalities or injuries is not known. Here we generated mice in which the vesicular glutamate transporter 2 (VGLUT2) is ablated selectively from DRG neurons. We report that conditional knockout (cKO) of the Slc17a6 gene encoding VGLUT2 from the great majority of nociceptors profoundly decreased VGLUT2 mRNA and protein in these neurons, and reduced firing of lamina I spinal cord neurons in response to noxious heat and mechanical stimulation. In behavioral assays, cKO mice showed decreased responsiveness to acute noxious heat, mechanical, and chemical (capsaicin) stimuli, but responded normally to cold stimulation and in the formalin test. Strikingly, although tissue injury-induced heat hyperalgesia was lost in the cKO mice, mechanical hypersensitivity developed normally. In a model of nerve injury-induced neuropathic pain, the magnitude of heat hypersensitivity was diminished in cKO mice, but both the mechanical allodynia and the microgliosis generated by nerve injury were intact. These findings suggest that VGLUT2 expression in nociceptors is essential for normal perception of acute pain and heat hyperalgesia, and that heat and mechanical hypersensitivity induced by peripheral injury rely on distinct (VGLUT2 dependent and VGLUT2 independent, respectively) primary afferent mechanisms and pathways.


Subject(s)
Ganglia, Spinal/metabolism , Gene Expression Regulation , Hot Temperature , Hyperalgesia/metabolism , Nociceptors/metabolism , Pain/metabolism , Vesicular Glutamate Transport Protein 2/biosynthesis , Animals , Behavior, Animal/drug effects , Capsaicin/pharmacology , Cold Temperature , Fixatives/pharmacology , Formaldehyde/pharmacology , Hyperalgesia/genetics , Mice , Mice, Knockout , Pain/genetics , Sensory System Agents/pharmacology , Vesicular Glutamate Transport Protein 2/genetics
7.
Neuron ; 110(22): 3711-3726.e16, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36087583

ABSTRACT

Axon degeneration is an early pathological event in many neurological diseases. The identification of the nicotinamide adenine dinucleotide (NAD) hydrolase SARM1 as a central metabolic sensor and axon executioner presents an exciting opportunity to develop novel neuroprotective therapies that can prevent or halt the degenerative process, yet limited progress has been made on advancing efficacious inhibitors. We describe a class of NAD-dependent active-site SARM1 inhibitors that function by intercepting NAD hydrolysis and undergoing covalent conjugation with the reaction product adenosine diphosphate ribose (ADPR). The resulting small-molecule ADPR adducts are highly potent and confer compelling neuroprotection in preclinical models of neurological injury and disease, validating this mode of inhibition as a viable therapeutic strategy. Additionally, we show that the most potent inhibitor of CD38, a related NAD hydrolase, also functions by the same mechanism, further underscoring the broader applicability of this mechanism in developing therapies against this class of enzymes.


Subject(s)
Armadillo Domain Proteins , NAD , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , NAD/metabolism , Neuroprotection , Cytoskeletal Proteins/metabolism , Axons/metabolism , Hydrolases/metabolism
8.
J Neurosci ; 29(17): 5508-15, 2009 Apr 29.
Article in English | MEDLINE | ID: mdl-19403818

ABSTRACT

Activation of primary afferent nociceptors produces acute, short-lived pain, and tissue or nerve injury induces long-term enhancement of nociceptive processing, manifested as hypersensitivity to thermal and mechanical stimulation. Here we used a chemical-genetic and pharmacological approach to study the contribution of the receptor tyrosine kinase, type 2 (TrkB) to the generation and maintenance of injury-induced persistent pain. We performed the studies in wild-type mice and transgenic (TrkB(F616A)) mice that express mutant but fully functional TrkB receptors. By injecting a small molecule derivative of the protein kinase inhibitor protein phosphatase 1 (1NM-PP1), it is possible to produce highly selective inhibition of TrkB autophosphorylation in adult mice, without interfering with the activity of other protein kinases. We report that oral administration of 1NM-PP1, at doses that blocked phosphorylation of TrkB in the spinal cord, had no effect in behavioral tests of acute heat, mechanical, or chemical pain sensitivity. However, the same pretreatment with 1NM-PP1 prevented the development of tissue- or nerve injury-induced heat and mechanical hypersensitivity. Established hypersensitivity was transiently reversed by intraperitoneal injection of 1NM-PP1. Although interfering with TrkB signaling altered neither acute capsaicin nor formalin-induced pain behavior, the prolonged mechanical hypersensitivity produced by these chemical injuries was prevented by 1NM-PP1 inhibition of TrkB signaling. We conclude that TrkB signaling is not only an important contributor to the induction of heat and mechanical hypersensitivity produced by tissue or nerve injury but also to the persistence of the pain.


Subject(s)
Neuralgia/metabolism , Receptor, trkB/biosynthesis , Signal Transduction/physiology , Animals , Chronic Disease , Female , Male , Mice , Mice, Transgenic , Neuralgia/genetics , Pain Measurement/drug effects , Pain Measurement/methods , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Signal Transduction/drug effects , Stimulation, Chemical
9.
eNeuro ; 6(3)2019.
Article in English | MEDLINE | ID: mdl-31122949

ABSTRACT

Reelin (Reln) and Disabled-1 (Dab1) participate in the Reln-signaling pathway and when either is deleted, mutant mice have the same spinally mediated behavioral abnormalities, increased sensitivity to noxious heat and a profound loss in mechanical sensitivity. Both Reln and Dab1 are highly expressed in dorsal horn areas that receive and convey nociceptive information, Laminae I-II, lateral Lamina V, and the lateral spinal nucleus (LSN). Lamina I contains both projection neurons and interneurons that express Neurokinin-1 receptors (NK1Rs) and they transmit information about noxious heat both within the dorsal horn and to the brain. Here, we ask whether the increased heat nociception in Reln and dab1 mutants is due to incorrectly positioned dorsal horn neurons that express NK1Rs. We found more NK1R-expressing neurons in Reln-/- and dab1-/- Laminae I-II than in their respective wild-type mice, and some NK1R neurons co-expressed Dab1 and the transcription factor Lmx1b, confirming their excitatory phenotype. Importantly, heat stimulation in dab1-/- mice induced Fos in incorrectly positioned NK1R neurons in Laminae I-II. Next, we asked whether these ectopically placed and noxious-heat responsive NK1R neurons participated in pain behavior. Ablation of the superficial NK1Rs with an intrathecal injection of a substance P analog conjugated to the toxin saporin (SSP-SAP) eliminated the thermal hypersensitivity of dab1-/- mice, without altering their mechanical insensitivity. These results suggest that ectopically positioned NK1R-expressing neurons underlie the heat hyperalgesia of Reelin-signaling pathway mutants, but do not contribute to their profound mechanical insensitivity.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hyperalgesia/physiopathology , Nerve Tissue Proteins/physiology , Posterior Horn Cells/physiology , Receptors, Neurokinin-1/physiology , Serine Endopeptidases/physiology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Hot Temperature , Male , Mice, Knockout , Nerve Tissue Proteins/genetics , Posterior Horn Cells/metabolism , Receptors, Neurokinin-1/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction , Spinal Cord/physiopathology
10.
Nat Neurosci ; 19(1): 94-101, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26642091

ABSTRACT

Although microglia have been implicated in nerve injury-induced neuropathic pain, the manner by which injured sensory neurons engage microglia remains unclear. We found that peripheral nerve injury induced de novo expression of colony-stimulating factor 1 (CSF1) in injured sensory neurons. CSF1 was transported to the spinal cord, where it targeted the microglial CSF1 receptor (CSF1R). Cre-mediated sensory neuron deletion of Csf1 completely prevented nerve injury-induced mechanical hypersensitivity and reduced microglial activation and proliferation. In contrast, intrathecal injection of CSF1 induced mechanical hypersensitivity and microglial proliferation. Nerve injury also upregulated CSF1 in motoneurons, where it was required for ventral horn microglial activation and proliferation. Downstream of CSF1R, we found that the microglial membrane adaptor protein DAP12 was required for both nerve injury- and intrathecal CSF1-induced upregulation of pain-related microglial genes and the ensuing pain, but not for microglial proliferation. Thus, both CSF1 and DAP12 are potential targets for the pharmacotherapy of neuropathic pain.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Gene Expression Regulation , Macrophage Colony-Stimulating Factor/metabolism , Microglia/metabolism , Motor Neurons/metabolism , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Sensory Receptor Cells/metabolism , Animals , Male , Mice , Up-Regulation
11.
PLoS One ; 10(12): e0142906, 2015.
Article in English | MEDLINE | ID: mdl-26630489

ABSTRACT

Although central serotonergic systems are known to influence responses to noxious stimuli, mechanisms underlying serotonergic modulation of pain responses are unclear. We proposed that serotonin 2C receptors (5-HT2CRs), which are expressed within brain regions implicated in sensory and affective responses to pain, contribute to the serotonergic modulation of pain responses. In mice constitutively lacking 5-HT2CRs (2CKO mice) we found normal baseline sensory responses to noxious thermal, mechanical and chemical stimuli. In contrast, 2CKO mice exhibited a selective enhancement of affect-related ultrasonic afterdischarge vocalizations in response to footshock. Enhanced affect-related responses to noxious stimuli were also exhibited by 2CKO mice in a fear-sensitized startle assay. The extent to which a brief series of unconditioned footshocks produced enhancement of acoustic startle responses was markedly increased in 2CKO mice. As mesolimbic dopamine pathways influence affective responses to noxious stimuli, and these pathways are disinhibited in 2CKO mice, we examined the sensitivity of footshock-induced enhancement of startle to dopamine receptor blockade. Systemic administration of the dopamine D2/D3 receptor antagonist raclopride selectively reduced footshock-induced enhancement of startle without influencing baseline acoustic startle responses. We propose that 5-HT2CRs regulate affective behavioral responses to unconditioned aversive stimuli through mechanisms involving the disinhibition of ascending dopaminergic pathways.


Subject(s)
Fear/physiology , Receptor, Serotonin, 5-HT2C/physiology , Reflex, Startle/physiology , Vocalization, Animal/physiology , Animals , Dopamine Antagonists/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Raclopride/pharmacology , Receptors, Dopamine D2/chemistry , Reflex, Startle/drug effects , Ultrasonics , Vocalization, Animal/drug effects , Vocalization, Animal/radiation effects
12.
Pain ; 156(6): 1084-1091, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25760475

ABSTRACT

Decreased spinal cord GABAergic inhibition is a major contributor to the persistent neuropathic pain that can follow peripheral nerve injury. Recently, we reported that restoring spinal cord GABAergic signaling by intraspinal transplantation of cortical precursors of GABAergic interneurons from the embryonic medial ganglionic eminence (MGE) can reverse the mechanical hypersensitivity (allodynia) that characterizes a neuropathic pain model in the mouse. We show that MGE cell transplants are also effective against both the mechanical allodynia and the heat hyperalgesia produced in a paclitaxel-induced chemotherapy model of neuropathic pain. To test the necessity of GABA release by the transplants, we also studied the utility of transplanting MGE cells from mice with a deletion of VGAT, the vesicular GABA transporter. Transplants from these mice, in which GABA is synthesized but cannot be stored or released, had no effect on mechanical hypersensitivity or heat hyperalgesia in the paclitaxel model. Taken together, these results demonstrate the therapeutic potential of GABAergic precursor cell transplantation in diverse neuropathic pain models and support our contention that restoration of inhibitory controls through release of GABA from the transplants is their mode of action.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Cell Transplantation/methods , Hyperalgesia , Paclitaxel/toxicity , Spinal Cord/metabolism , gamma-Aminobutyric Acid/metabolism , Activating Transcription Factor 3/metabolism , Animals , Cell Count , Disease Models, Animal , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/surgery , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain Measurement , Pain Threshold , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
13.
Neuron ; 82(3): 522-36, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24811377

ABSTRACT

The original formulation of Gate Control Theory (GCT) proposed that the perception of pain produced by spinal cord signaling to the brain depends on a balance of activity generated in large (nonnociceptive)- and small (nociceptive)-diameter primary afferent fibers. The theory proposed that activation of the large-diameter afferent "closes" the gate by engaging a superficial dorsal horn interneuron that inhibits the firing of projection neurons. Activation of the nociceptors "opens" the gate through concomitant excitation of projection neurons and inhibition of the inhibitory interneurons. Sixty years after publication of the GCT, we are faced with an ever-growing list of morphologically and neurochemically distinct spinal cord interneurons. The present Review highlights the complexity of superficial dorsal horn circuitry and addresses the question whether the premises outlined in GCT still have relevance today. By examining the dorsal horn circuits that underlie the transmission of "pain" and "itch" messages, we also address the extent to which labeled lines can be incorporated into a contemporary view of GCT.


Subject(s)
Nerve Net/physiology , Pain/physiopathology , Pruritus/physiopathology , Sensory Gating/physiology , Spinal Cord/physiology , Synaptic Transmission/physiology , Animals , Humans , Nerve Net/pathology , Pain/diagnosis , Pruritus/diagnosis , Spinal Cord/pathology
14.
Neuron ; 78(2): 312-24, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23622066

ABSTRACT

To what extent dorsal horn interneurons contribute to the modality specific processing of pain and itch messages is not known. Here, we report that loxp/cre-mediated CNS deletion of TR4, a testicular orphan nuclear receptor, results in loss of many excitatory interneurons in the superficial dorsal horn but preservation of primary afferents and spinal projection neurons. The interneuron loss is associated with a near complete absence of supraspinally integrated pain and itch behaviors, elevated mechanical withdrawal thresholds and loss of nerve injury-induced mechanical hypersensitivity, but reflex responsiveness to noxious heat, nerve injury-induced heat hypersensitivity, and tissue injury-induced heat and mechanical hypersensitivity are intact. We conclude that different subsets of dorsal horn excitatory interneurons contribute to tissue and nerve injury-induced heat and mechanical pain and that the full expression of supraspinally mediated pain and itch behaviors cannot be generated solely by nociceptor and pruritoceptor activation of projection neurons; concurrent activation of excitatory interneurons is essential.


Subject(s)
Interneurons/physiology , Pain/genetics , Pain/pathology , Pruritus/pathology , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Spinal Nerve Roots/pathology , Animals , Cell Death/genetics , Cognition Disorders/etiology , Cognition Disorders/genetics , Disease Models, Animal , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/metabolism , Hyperalgesia/genetics , Hyperalgesia/pathology , Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins v-fos/metabolism , Pain Threshold/physiology , Phosphopyruvate Hydratase/metabolism , Pruritus/genetics , Reaction Time/genetics , Receptors, Steroid/deficiency , Receptors, Thyroid Hormone/deficiency , Substance P/metabolism
15.
Nat Neurosci ; 16(9): 1284-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23872594

ABSTRACT

TMEM16C belongs to the TMEM16 family, which includes the Ca(2+)-activated Cl(-) channels TMEM16A and TMEM16B and a small-conductance, Ca(2+)-activated, nonselective cation channel (SCAN), TMEM16F. We found that in rat dorsal root ganglia (DRG) TMEM16C was expressed mainly in the IB4-positive, non-peptidergic nociceptors that also express the sodium-activated potassium (K(Na)) channel Slack. Together these channel proteins promote K(Na) channel activity and dampen neuronal excitability. DRG from TMEM16C knockout rats had diminished Slack expression, broadened action potentials and increased excitability. Moreover, the TMEM16C knockout rats, as well as rats with Slack knockdown by intrathecal injection of short interfering RNA, exhibited increased thermal and mechanical sensitivity. Experiments involving heterologous expression in HEK293 cells further showed that TMEM16C modulated the single-channel activity of Slack channels and increased its sodium sensitivity. Our study thus reveals that TMEM16C enhances K(Na) channel activity in DRG neurons and regulates the processing of pain messages.


Subject(s)
Chloride Channels/metabolism , Nerve Tissue Proteins/metabolism , Pain Threshold/physiology , Potassium Channels/metabolism , Sensory Receptor Cells/physiology , Sodium/metabolism , Animals , Cadmium/pharmacology , Cells, Cultured , Chloride Channels/deficiency , Chloride Channels/genetics , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Humans , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mutation/physiology , Nerve Tissue Proteins/genetics , New Brunswick , Pain Measurement , Pain Threshold/drug effects , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , RNA, Small Interfering/pharmacology , Rats , Rats, Transgenic , Sensory Receptor Cells/drug effects , Sodium Chloride/pharmacology , Spinal Cord/cytology
16.
Life Sci ; 88(13-14): 590-7, 2011 Mar 28.
Article in English | MEDLINE | ID: mdl-21277866

ABSTRACT

AIMS: Immunoglobulin superfamily (IgSF) proteins play a critical role in development of the nervous system. Here, a new member of IgSF gene family was cloned from rat brain, which was subsequently identified as rat homolog of Drosophila Kirre. This new molecule was named as rat Kirre (rKirre). We aimed to reveal the developmental expression of rKirre, both at mRNA and protein levels, in the central nervous system. The deduced amino acid sequence of rKirre showed a putative PDZ binding motif at the C-terminus, which provided a rationale for analyzing the co-localization of rKirre and post-synaptic density protein 95 (PSD-95) in cultured rat cortical neurons. MAIN METHODS: cDNA library screening was used in the isolation of cDNA. Northern blotting and Western blotting were used to reveal the levels of rKirre expression. In situ hybridization and immuno-fluorescent staining were used to determine the localization of rKirre. KEY FINDINGS: The rKirre gene was found to be highly expressed in the cerebrum, hippocampus, cerebellum, brain stem and spinal cord of adult rats. In parallel, the protein level of rKirre was also increased in a developing cerebral cortex. In cultured rat cortical neurons, the amount of rKirre was significantly increased during neuronal differentiation. Immuno-cytofluorescent staining indicated that rKirre was present along the neurites of cortical neurons, and was co-localized with PSD-95. SIGNIFICANCE: These results suggested that rKirre might play an essential role in neuronal differentiation and development in the central nervous system.


Subject(s)
Central Nervous System/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Western , Cell Differentiation , Cells, Cultured , Central Nervous System/embryology , Central Nervous System/growth & development , Cloning, Molecular , DNA, Complementary/genetics , Disks Large Homolog 4 Protein , Gene Library , Immunohistochemistry , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/genetics , Male , Molecular Sequence Data , Neurons/metabolism , PDZ Domains , Rats , Rats, Sprague-Dawley , Sequence Alignment
17.
Pain ; 146(1-2): 130-40, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19748740

ABSTRACT

Using the chronic constriction injury (CCI) model of neuropathic pain, we profiled gene expression in the rat spinal cord, and identified SIP30 as a gene whose expression was elevated after CCI. SIP30 was previously shown to interact with SNAP25, but whose function was otherwise unknown. We now show that in the spinal cord, SIP30 was present in the dorsal horn laminae where the peripheral nociceptive inputs first synapse, co-localizing with nociception-related neuropeptides CGRP and substance P. With the onset of neuropathic pain after CCI surgery, SIP30 mRNA and protein levels increased in the ipsilateral side of the spinal cord, suggesting a potential association between SIP30 and neuropathic pain. When CCI-upregulated SIP30 was inhibited by intrathecal antisense oligonucleotide administration, neuropathic pain was attenuated. This neuropathic pain-reducing effect was observed both during neuropathic pain onset following CCI, and after neuropathic pain was fully established, implicating SIP30 involvement in the development and maintenance phases of neuropathic pain. Using a secretion assay in PC12 cells, anti-SIP30 siRNA decreased the total pool of synaptic vesicles available for exocytosis, pointing to a potential function for SIP30. These results suggest a role of SIP30 in the development and maintenance of peripheral nerve injury-induced neuropathic pain.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Nerve Tissue Proteins/physiology , Pain/pathology , Peripheral Nerves/pathology , Peripheral Nervous System Diseases/pathology , Animals , Blotting, Western , Chromosomal Proteins, Non-Histone/genetics , Chronic Disease , Constriction, Pathologic , Exocytosis/drug effects , Hot Temperature , Immunohistochemistry , Male , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , Pain/psychology , Pain Measurement , Peripheral Nervous System Diseases/psychology , Physical Stimulation , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Neuropathy/pathology , Spinal Cord/metabolism
18.
Eur J Neurosci ; 22(5): 1090-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16176350

ABSTRACT

Neuropathic pain from nerve injury by trauma, disease or surgery often causes prolonged suffering. To explore the molecular mechanisms that underlie neuropathic pain, we used mRNA from the L4--5 segments of the lumbar spinal cord of rats with chronic constriction injury (CCI)-induced neuropathic pain, and differentially screened a cDNA library from the rat brain. A novel gene, termed RSEP1 (Rat Spinal cord Expression Protein 1), was identified. Northern blots revealed that RSEP1 was expressed mainly in the central nervous system including the cerebral cortex, hippocampus, brainstem and spinal cord, as well as in the kidney and ovary. In situ hybridization showed a high level of RSEP1 expression in the CA1, CA3 and dentate gyrus regions of the hippocampus and the small sensory neurons in the dorsal horn, as well as the large neurons in the ventral horn of the spinal cord. Intrathecal injection of RSEP1 antisense oligonucleotide into the spinal cord lumbar enlargement attenuated neuropathic pain behaviours in CCI rats, suggesting a functional involvement of RSEP1 in neuropathic pain.


Subject(s)
Nerve Tissue Proteins/physiology , Pain/metabolism , Sciatic Neuropathy/metabolism , Animals , Behavior, Animal , Blotting, Northern/methods , Brain/metabolism , Cloning, Molecular/methods , Disease Models, Animal , Green Fluorescent Proteins/metabolism , Hyperalgesia/drug therapy , In Situ Hybridization/methods , Indoles/metabolism , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligonucleotide Array Sequence Analysis/methods , Pain/etiology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Sciatic Neuropathy/complications , Sequence Analysis, DNA/methods , Sequence Analysis, Protein/methods , Time Factors
19.
Acta Biochim Biophys Sin (Shanghai) ; 36(7): 501-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15248025

ABSTRACT

The low-abundantly expressed genes composed the majorities of the mRNAs expressed in the central nervous system (CNS), and were thought to be important for the normal brain functions. Through differential screening a low-abundance cDNA sublibrary with mRNA from neuropathic pain of chronic constriction injury (CCI) model, we have identified a novel rat gene, rat spinal-cord expression protein 4 gene (RSEP4). The total length of RSEP4 cDNA is 2006 bp, with a 501 nucleotide open reading frame (ORF) that encodes a 167 amino acid polypeptide. Northern blot revealed that RSEP4 was expressed specifically in the CNS. In situ hybridization showed that the mRNA of RSEP4 was strongly expressed in the CA1, CA2, CA3 and DG regions of hippocampus, the Purkinje cells of cerebellum, and the small sensory neurons of dorsal horn and large motor neurons of ventral horn of spinal cord. Over-expression of RSEP4-EGFP fusion protein in the human embryonic kidney 293T cells showed that RSEP4 protein was mainly localized in the cell cytoplasm. These results suggest that RSEP4 may play some roles in the CNS.


Subject(s)
Nerve Tissue Proteins/metabolism , Spinal Cord/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , DNA, Complementary , In Situ Hybridization , Male , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL