Skip to main content
Erschienen in: Molecular Pain 1/2009

Open Access 01.12.2009 | Short report

Inhibitory coupling between inhibitory interneurons in the spinal cord dorsal horn

verfasst von: Charalampos Labrakakis, Louis-Etienne Lorenzo, Cyril Bories, Alfredo Ribeiro-da-Silva, Yves De Koninck

Erschienen in: Molecular Pain | Ausgabe 1/2009

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Local inhibitory interneurons in the dorsal horn play an important role in the control of excitability at the segmental level and thus determine how nociceptive information is relayed to higher structures. Regulation of inhibitory interneuron activity may therefore have critical consequences on pain perception. Indeed, disinhibition of dorsal horn neuronal networks disrupts the balance between excitation and inhibition and is believed to be a key mechanism underlying different forms of pain hypersensitivity and chronic pain states. In this context, studying the source and the synaptic properties of the inhibitory inputs that the inhibitory interneurons receive is important in order to predict the impact of drug action at the network level. To address this, we studied inhibitory synaptic transmission in lamina II inhibitory interneurons identified under visual guidance in spinal slices taken from transgenic mice expressing enhanced green fluorescent protein (EGFP) under the control of the GAD promoter. The majority of these cells fired tonically to a long depolarizing current pulse. Monosynaptically evoked inhibitory postsynaptic currents (eIPSCs) in these cells were mediated by both GABAA and glycine receptors. Consistent with this, both GABAA and glycine receptor-mediated miniature IPSCs were recorded in all of the cells. These inhibitory inputs originated at least in part from local lamina II interneurons as verified by simultaneous recordings from pairs of EGFP+ cells. These synapses appeared to have low release probability and displayed potentiation and asynchronous release upon repeated activation. In summary, we report on a previously unexamined component of the dorsal horn circuitry that likely constitutes an essential element of the fine tuning of nociception.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1744-8069-5-24) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CL and YDK conceived and designed the study. CL performed the experiments, LEL performed the immunostainings, CB performed Lucifer Yellow injections and 2D reconstruction of EGFP+ neurons. All authors contributed to the preparation of the final manuscript.

Findings

The superficial laminae of the dorsal horn act as a relay station for nociceptive sensory signals. They receive inputs from unmyelinated C- and thinly myelinated Aδ-fibers [13] and process this input through a network of local inhibitory and excitatory interneurons before relaying it to supraspinal centers. Inhibitory interneurons are key players in the regulation of spinal excitability, by releasing the neurotransmitter GABA and, in many cases, co-releasing glycine [4]. Their important role in pain processing is illustrated by the finding that local blockade of GABAA receptors (GABAARs) and glycine receptors (GlyRs) replicates symptoms of neuropathic and inflammatory pain [58]. Blocking this form of inhibition also allows low threshold input to be relayed to normally nociceptive specific neurons [912]. Despite this, very little is known about the functional circuitry of the dorsal horn and, in particular, on inhibitory connections onto GABAergic interneurons. This is an important aspect of the dorsal horn circuitry as inhibitory inputs onto inhibitory neurons would reduce their output leading to net disinhibition and spinal hyperexcitability.
Towards this, we used transgenic mice that express EGFP under the control of the glutamate decarboxylase 65 gene (GAD65), the enzyme that catalyzes the production of GABA[13], to be able to visually identify GABAergic interneurons and perform targeted recordings from them in live spinal cord slices. Figure 1A shows a high density of EGFP+ neurons in the superficial laminae as well as in the medial part of lamina V in these mice. Previous reports indicate that, in this transgenic line, virtually all (> 98%) of EGFP+ cells co-express GAD65 [14]. In addition, we found that 25% of the EGFP+ cells co-expressed parvalbumin (Figure 1B), consistent with findings in another mouse line [15]. Previous studies report that parvalbumin-immunoreactive GABAergic cells also express glycine [16], indicating that a subpopulation of EGFP+ cells in our study are also glycinergic. EGFP+ neuron morphology (Figure 1C) was comparable to that previously described for inhibitory interneurons in mice [15, 17] and rats [18, 19].
We then studied the firing responses of the cells to depolarizing pulses as previously described to further characterize the population [15, 17, 1922]. Of the 33 neurons tested, 23 showed a clear tonic firing pattern (Figure 1D). An additional 4 cells showed a firing pattern that could be interpreted, at first sight, as phasic or initial bursting [15, 20] (Figure 1D). Using isolated responses can however lead to misclassification. Indeed, in a previous study, we showed that the decisive classification criterion is the nature of the input-output curve of the cell: tonic cells display a characteristic linear encoding curve, while phasic cells encode non-linearly with an abrupt increase in the f-I curve at low stimulus intensity [20], yielding a discontinuous f-I. The latter difference in encoding properties of tonic vs. phasic neurons corresponds to class 1 vs. class 2 excitability as defined by Hodgkin's initial classification [23]. Using this classification criterion, we found that these 4 additional cells fell under the tonic class (Figure 1D). Previous work also suggests identity of the two populations based on the ability of individual neurons to convert from one pattern to the other [24]. The remaining 6 neurons had a single spike firing pattern (Figure 1D). These data are consistent with previous reports on dorsal horn inhibitory interneurons in mice [17, 22, 25] and rats [3, 18].
To test for inhibitory input to GAD65-EGFP+ neurons, we recorded responses to focal stimulation in the presence of CNQX and APV. Under these conditions, monosynaptic eIPSCs with complex decay kinetics were recorded in all cells: a fast component (12.6 ± 0.5 ms) and a slower component (200.8 ± 26.9 ms) (n = 11; Figure 2). The fast component was blocked by the GlyR antagonist strychnine (0.5 μM; n = 8; Figure 2) and the slow one by the GABAAR antagonist SR95531 (10 μM; n = 6; Figure 2). Complex eIPSC are the result of activation and release from several GABAergic and glycinergic axon terminals and thus complex GABAAR and GlyR mediated eIPSCs should not be misinterpreted as evidence of co-release of the two neurotransmitters from the same terminal.
To test whether IPSCs at individual synapses had similar properties, we studied the decay kinetics of mIPSCs (Figure 3A). Average mIPSCs displayed similar dual decay kinetics: τ1 = 12 ± 0.9 ms and τ2 = 156.8 ± 64.4 ms (n = 9). As for eIPSCs, strychnine administration confirmed the fast component was mediated by GlyRs (n = 6). Individual mIPSCs could be distinguished on the basis of their decay kinetics (Figure 3B): mIPSCs with monotonic fast decay kinetics made up the majority of events (66%). These were absent in strychnine confirming that they were mediated by GlyRs only. Another subpopulation was characterized by slow decay kinetics (27%), consistent with them being mediated by GABAARs [19, 2628]. A remaining small subset of events showed a dual decay kinetics (7%) composed by a fast (< 14 ms) and a slower component (Figure 3C); they were absent in the presence of strychnine, indicating that they corresponded to dual GABAAR- and GlyR-mediated mIPSCs, consistent with co-release from the same synapse [26]. Additional experiments using pipettes filled with high Cl- solutions to yield inward Cl- currents at a holding potential of -70 mV showed similar kinetics. Under these conditions, all inward currents were abolished by addition of strychnine and SR95531 indicating that the evoked responses were entirely mediated by GABAARs and GlyRs.
To study the behaviour of this inhibitory synapse in response to repetitive activation, we recorded eIPSCs following trains of focal stimuli (Figure 4A). Using this paradigm, three forms of plasticity were observed. First, repetitive train stimulation caused a significant increase in amplitude of the first eIPSC of each train (Figure 4B; p < 0.05, Friedman test, n = 20 cells). Second, within individual trains, short term plastic changes followed opposite trends, depending on whether the first eIPSC of the train was potentiated or not; there was facilitation when the first eIPSC was not-potentiated vs. depression when the first eIPSC was potentiated (Figure 4C; n = 20). Third, in 16 out of 20 cells asynchronous release occurred at the end of each train of stimuli (Figure 4D). This is shown by the observation that the frequency of spontaneous IPSCs changed from 0.5 ± 0.08 Hz during the second preceding the train to 2.9 ± 0.4 during the second following the end of the train (paired t-test, p < 0.05, n = 20). Such asynchronous release phenomenon was observed for both pharmacologically isolated GABAAR and GlyR-mediated IPSCs. These specific plastic properties have been observed at other, but not all inhibitory synapses in the CNS [2932].
The sources of the inhibitory inputs to inhibitory interneurons in lamina II could be other local interneurons, lamina III inhibitory interneurons [19, 22] and/or descending fibres from the brain [33]. To test whether local inhibitory interneurons can themselves be the source of the IPSCs recorded on inhibitory interneurons, we performed simultaneous whole-cell recordings from pairs of EGFP+ lamina II neurons. In recordings from 56 EGFP+ pairs, 3 pairs were found synaptically connected. In 2 of the 3 pairs, the direction of the synaptic connection was tested and was found unidirectional in both pairs. The latency between the peak of the action potential and the onset of the IPSC ranged between 1.6 and 3.4 ms (mean 2.3 ± 0.5 ms). In one of the pairs, some IPSCs had clear dual decay kinetics (Figure 5A), with a fast component comparable to that mediated by glycine receptors as described above indicative of mixed glycine- and GABAA IPSCs. In the other two pairs, IPSCs were lacking the fast decaying component, indicating GABAA-only IPSCs. All three synaptically coupled pairs displayed high degree of failure. Probability of release in response to the first action potential of the train ranged between 0.12–0.23 (Figure 5B). In 2 of the pairs, probability of release increased significantly in response to the subsequent action potentials in the train (χ2, p < 0.05). In the third pair, the probability of release was not different in response to the first three action potentials but increased later (Figure 5B–D). Thus, the connection between dorsal horn neurons is very selective as shown by the rarity of synaptically connected pairs in this and previous studies [34]. In addition, such connectivity between inhibitory interneurons appears to display low fidelity (< 25%), in contrast to connections between inhibitory islet cells and excitatory transient central cells which showed high fidelity (60–100%) [18, 34]. This suggests that our recordings are from different synapses from that previously described and thus consist in a novel component of the superficial dorsal horn circuitry.
Our results indicate that inhibitory interneurons impose inhibition locally on other inhibitory interneurons in the superficial dorsal horn. This synaptic arrangement can be the substrate of local disinhibition, which may play a key role in sensory processing. Indeed, GABAergic interneurons intercalated within the circuitry linking primary afferents and spinal projection neurons [25] may play a dual role in controlling network excitability: lamina II inhibitory interneurons can act, directly, to repress polysynaptic excitatory relay connections [11] or, indirectly, to potentiate these connections by silencing specific inhibitory components of the dorsal horn circuitry. Understanding how drugs may selectively affect such distinct inhibitory synapses is critical to predict the net result of a specific treatment on spinal output. Equally, the finding that inhibitory synapses impinging on inhibitory interneurons are characterized by specific properties opens the door for differential modulation of inhibitory vs. "disinhibitory" components of the spinal dorsal horn, yielding potentially more effective interventions.

Methods

All experiments were performed in accordance with regulations of the Canadian Council and Animal Care. Mice in this study were heterozygous C57BL/6 BAC transgenic mice expressing enhanced green fluorescent protein (EGFP) under the control of the GAD65 promoter [14, 35]. Adult transgenic mice (3–5 months) were anaesthetized with ketamine/xylazine and perfused intracardially with ice-cold oxygenated (95% O2, 5% CO2) sucrose substituted ACSF containing (mM) 252 sucrose, 2.5 KCl, 1.5 CaCl2, 6 MgCl2, 10 glucose, 26 NaHCO3, 1.25 NaH2PO4 and 5 kynurenic acid. Mice were decapitated, the spinal cord was removed by hydraulic extrusion and 250 μm thick parasagittal slices were cut from the lumbar portion [36]. Slices were kept in normal oxygenated ACSF (126 NaCl, 2.5 KCl, 2 CaCl2, 2 MgCl2, 10 glucose, 26 NaHCO3, 1.25 NaH2PO4) at room temperature until recording.
Slices were transferred in the recording chamber and continuously superfused at 2–3 ml/min with oxygenated ACSF at room temperature. Patch pipettes (7–8 MΩ) were filled with (in mM) 135 KCl, 10 HEPES, 2 MgCl2, 0.5 EGTA, 2 ATP, pH 7.2. For some recordings, KCl was substituted by 130 CsMeSO3/5 CsCl or by 135 CsCl. Whole cell patch clamp recordings were made using a Multiclamp 700B amplifier (Molecular Devices, Sunnyvale, California, UK). Data were low pass filtered at 10 kHz, digitized at 20–30 kHz and acquired with the Strathclyde electrophysiology software (WinWCP and WinEDR courtesy of Dr. J. Dempster, University of Strathclyde, Glasgow, UK).
Epifluorescence was used to identify EGFP+ neurons. All recordings in this study are from neurons with their somata located in lamina II. Action potential firing was analysed as previously described [20]. Only cells with resting membrane potential more negative than -50 mV were included in the study. Instantaneous firing frequency (f) was calculated as the reciprocal of the interspike interval.
The mIPSCs were detected and analysed using Mini Analysis (Synaptosoft, Decatur, Georgia, UK) and a locally designed software (YDK). Decay time constants were fitted using automated least square algorithms. The necessity to introduce additional exponential components to the fits was first judged on the basis of visual inspection. When the merit of additional components was not obvious, further statistical analysis was applied as previously described [26]. To simplify analysis, complex mIPSC that did not contain the strychnine sensitive, fast decay component (< 14 ms; i.e. mIPSCs that consisted of multiple GABAAR-mediated decay components), were treated as monotonic.
Monosynaptic IPSCs were evoked focally by electrical stimulation (30–70 μA, 250 μs) via patch pipette filled with ACSF placed 50–100 μm from the soma of the recorded cell as described previously [26]. Single stimuli or trains of stimuli (12 pulses at 20 Hz intraburst frequency) were delivered every 10 or 20 s, respectively. To isolate the complex decay components of the eIPSCs pharmacologically, all tested cells were treated with strychnine or SR95531. In all cases strychnine blocked only the fast component. Similarly, SR95531, blocked only the slow component in all cases. The relative contribution of each component to the peak current was calculated from the pharmacologically blocked portion (by subtraction) and the portion that was not blocked. In some experiments, strychnine and SR95531 were applied together and abolished all components of the evoked current. eIPSC amplitude measurements from train stimulations were measured from the point immediately before the stimulation artefact to take into account baseline changes due to IPSC summation.
In simultaneous recordings from pairs of neurons (located within < 250 μm of each other), one of the neurons, defined as presynaptic, was kept in current clamp mode and injected with a 100 ms-long depolarizing current pulse of 100 pA to elicit a train of 4–7 action potentials. The other cell, defined as postsynaptic, was held at -70 mV in voltage clamp mode and postsynaptic currents were recorded. These recordings were conducted using KCl filled pipettes to allow for action potential generation and maximum amplification of Cl--mediated IPSCs. All glutamatergic transmission was blocked by APV and CNQX, thus all synaptic connections were assumed to be monosynaptic and inhibitory. This protocol was repeated for 40–50 trials at 10 s intervals for all tested pairs.
For immunostainings, animals were anaesthetized and perfused intracardially with 4% paraformaldehyde (PFA) in 0.1 M phosphate buffer. Spinal cord segments L4–L5 were collected, postfixed for 60 minutes in PFA and cryoprotected in 30% sucrose in phosphate buffer overnight at 4°C. Fifty μm-thick transverse sections were cut on a freezing microtome (Leica SM2000R). Sections were collected into tissue culture plates with 24 wells, washed with phosphate-buffered saline (pH = 7.4) and 0.2% Triton (PBST), and pre-treated with 5% donkey normal serum in PBST for 10 minutes.
The sections were washed twice in PBST and incubated overnight in a mixture of a mouse anti-parvalbumin antibody (1:4000, Sigma) and a rabbit anti-GFP antibody (1:500, Clontech Living Colors, Mountain View, CA) in PBST. After washing in PBST, the tissue was incubated for 2 hrs at room temperature in a Rhodamine Red-X-conjugated donkey anti-mouse IgG (H+L) antibody (1:500, Jackson, West Grove, PA) and in Alexa Fluor 488-conjugated IgG (H+L) donkey anti-rabbit antibody (Invitrogen, Carlsbad, California, USA) in PBST. Lastly, sections were washed for 15 minutes (3 × 5) with PBS, mounted on gelatin-subbed slides, allowed to dry overnight at 4°C and cover-slipped using Aquapolymount (Polysciences, Warrington, PA). The sections were observed with a Zeiss LSM 510 confocal microscope (Zeiss Canada). Images were obtained using a multi-track approach for the detection of two signals (Alexa Fluor 488 and Rhodamine Red-X), with the help of a 20 × water immersion objective. Data were collected with Metamorph 7 (Molecular Devices) from 1815 cells counted from 20 sections of three mice. Only cells with visible nuclei were counted.
For morphological classification of EGFP+ neurons, in a subset of electrophysiological experiments, neurons (n = 6) were recorded and filled with patch pipettes containing 0.5% neurobiotin. Slices were then fixed in 4% PFA in PB. In addition, 200 μm thick lumbar parasagittal slices taken from 4% PFA fixed spinal cords were used to inject EGFP+ neurons with Lucifer Yellow (LY) as described previously [37]. LY-injected slices were then incubated with a rabbit anti-LY antibody (1:20000) and subsequently with biotinylated goat anti-rabbit (1:500) antibody. Biotinylated antibodies and Neurobiotin were visualized with a horseradish peroxidase reaction (ABC kit, Vector Laboratories) and nickel-diaminobenzidine as a substrate. Cells were reconstructed using a light microscope with a computerized tracing system (Neurolucida, MicroBrightField Inc). Cells were classified according to previous descriptions [21]. As the distinction between islet and central cells is sometimes difficult [15] and due to our small sample, they were treated as one group.
Drugs were obtained as follows: 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) and D(-)-2-amino-5-phosphonopentanoic acid (D-APV) from Tocris Cookson (Ballwin, Montana, USa), Gabazine (SR95531) and strychnine hydrochloride from Sigma (St. Louis, Missouri, USA), and tetrodotoxin (TTX) from Alomone Labs (Jerusalem, Israel).
Data are presented as means ± SEM, n numbers refer to number of cells tested unless otherwise indicated. Normality of the data was tested with the Shapiro-Wilk test. Paired t-tests were used to compare IPSC frequencies. χ2 tests for contingency tables were used to determine differences in release probabilities. Repeated measure comparison of non-parametric data were analysed using the non-parametric Friedman test and subsequently analysed with the Student-Newman-Keuls posthoc test.

Acknowledgements

Supported by Canadian Institute of Health Research (CIHR) grants MOP-12942 to YDK and MOP-79411 to ARS. CL and LEL were supported by post-doctoral fellowships from CIHR. YDK is a Chercheur National of the Fonds de la recherche en santé du Québec (FRSQ). We thank Dr. F. Ferrini for helpful discussions and comments on the manuscript and Dr. A. Castonguay for help with imaging. We also thank Drs. F. Erdelyi and G. Szabo (Institute of Experimental Medicine, Budapest, Hungary) for the generous gift of GAD-GFP mice.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CL and YDK conceived and designed the study. CL performed the experiments, LEL performed the immunostainings, CB performed Lucifer Yellow injections and 2D reconstruction of EGFP+ neurons. All authors contributed to the preparation of the final manuscript.
Literatur
1.
Zurück zum Zitat Light AR, Perl ER: Reexamination of the dorsal root projection to the spinal dorsal horn including observations on the differential termination of coarse and fine fibers. J Comp Neurol 1979, 186: 117–131. 10.1002/cne.901860202PubMedCrossRef Light AR, Perl ER: Reexamination of the dorsal root projection to the spinal dorsal horn including observations on the differential termination of coarse and fine fibers. J Comp Neurol 1979, 186: 117–131. 10.1002/cne.901860202PubMedCrossRef
2.
Zurück zum Zitat Sugiura Y, Lee CL, Perl ER: Central projections of identified, unmyelinated (C) afferent fibers innervating mammalian skin. Science 1986, 234: 358–361. 10.1126/science.3764416PubMedCrossRef Sugiura Y, Lee CL, Perl ER: Central projections of identified, unmyelinated (C) afferent fibers innervating mammalian skin. Science 1986, 234: 358–361. 10.1126/science.3764416PubMedCrossRef
3.
Zurück zum Zitat Ribeiro-da-Silva A, De Koninck Y: Morphological and neurochemical organization of the spinal dorsal horn. In The Science of Pain. Edited by: Basbaum AI, Bushnell MC. New York: Academic Press; 2008:279–310. Ribeiro-da-Silva A, De Koninck Y: Morphological and neurochemical organization of the spinal dorsal horn. In The Science of Pain. Edited by: Basbaum AI, Bushnell MC. New York: Academic Press; 2008:279–310.
4.
Zurück zum Zitat Todd AJ, Sullivan AC: Light microscope study of the coexistence of GABA-like and glycine-like immunoreactivities in the spinal cord of the rat. J Comp Neurol 1990, 296: 496–505. 10.1002/cne.902960312PubMedCrossRef Todd AJ, Sullivan AC: Light microscope study of the coexistence of GABA-like and glycine-like immunoreactivities in the spinal cord of the rat. J Comp Neurol 1990, 296: 496–505. 10.1002/cne.902960312PubMedCrossRef
5.
Zurück zum Zitat Sherman SE, Loomis CW: Morphine insensitive allodynia is produced by intrathecal strychnine in the lightly anesthetized rat. Pain 1994, 56: 17–29. 10.1016/0304-3959(94)90146-5PubMedCrossRef Sherman SE, Loomis CW: Morphine insensitive allodynia is produced by intrathecal strychnine in the lightly anesthetized rat. Pain 1994, 56: 17–29. 10.1016/0304-3959(94)90146-5PubMedCrossRef
6.
Zurück zum Zitat Sivilotti L, Woolf CJ: The contribution of GABAA and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. J Neurophysiol 1994, 72: 169–179.PubMed Sivilotti L, Woolf CJ: The contribution of GABAA and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. J Neurophysiol 1994, 72: 169–179.PubMed
7.
Zurück zum Zitat Sorkin LS, Puig S: Neuronal model of tactile allodynia produced by spinal strychnine: effects of excitatory amino acid receptor antagonists and a mu-opiate receptor agonist. Pain 1996, 68: 283–292. 10.1016/S0304-3959(96)03130-2PubMedCrossRef Sorkin LS, Puig S: Neuronal model of tactile allodynia produced by spinal strychnine: effects of excitatory amino acid receptor antagonists and a mu-opiate receptor agonist. Pain 1996, 68: 283–292. 10.1016/S0304-3959(96)03130-2PubMedCrossRef
8.
Zurück zum Zitat Yaksh TL: Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain 1989, 37: 111–123. 10.1016/0304-3959(89)90160-7PubMedCrossRef Yaksh TL: Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain 1989, 37: 111–123. 10.1016/0304-3959(89)90160-7PubMedCrossRef
9.
Zurück zum Zitat Schoffnegger D, Ruscheweyh R, Sandkuhler J: Spread of excitation across modality borders in spinal dorsal horn of neuropathic rats. Pain 2008, 135: 300–310. 10.1016/j.pain.2007.12.016PubMedCrossRef Schoffnegger D, Ruscheweyh R, Sandkuhler J: Spread of excitation across modality borders in spinal dorsal horn of neuropathic rats. Pain 2008, 135: 300–310. 10.1016/j.pain.2007.12.016PubMedCrossRef
10.
Zurück zum Zitat Baba H, Ji RR, Kohno T, Moore KA, Ataka T, Wakai A, Okamoto M, Woolf CJ: Removal of GABAergic inhibition facilitates polysynaptic A fiber-mediated excitatory transmission to the superficial spinal dorsal horn. Mol Cell Neurosci 2003, 24: 818–830. 10.1016/S1044-7431(03)00236-7PubMedCrossRef Baba H, Ji RR, Kohno T, Moore KA, Ataka T, Wakai A, Okamoto M, Woolf CJ: Removal of GABAergic inhibition facilitates polysynaptic A fiber-mediated excitatory transmission to the superficial spinal dorsal horn. Mol Cell Neurosci 2003, 24: 818–830. 10.1016/S1044-7431(03)00236-7PubMedCrossRef
11.
Zurück zum Zitat Torsney C, MacDermott AB: Disinhibition opens the gate to pathological pain signaling in superficial neurokinin 1 receptor-expressing neurons in rat spinal cord. J Neurosci 2006, 26: 1833–1843. 10.1523/JNEUROSCI.4584-05.2006PubMedCrossRef Torsney C, MacDermott AB: Disinhibition opens the gate to pathological pain signaling in superficial neurokinin 1 receptor-expressing neurons in rat spinal cord. J Neurosci 2006, 26: 1833–1843. 10.1523/JNEUROSCI.4584-05.2006PubMedCrossRef
12.
Zurück zum Zitat Keller AF, Beggs S, Salter MW, De Koninck Y: Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol Pain 2007, 3: 27. 10.1186/1744-8069-3-27PubMedCentralPubMedCrossRef Keller AF, Beggs S, Salter MW, De Koninck Y: Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol Pain 2007, 3: 27. 10.1186/1744-8069-3-27PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Erlander MG, Tillakaratne NJ, Feldblum S, Patel N, Tobin AJ: Two genes encode distinct glutamate decarboxylases. Neuron 1991, 7: 91–100. 10.1016/0896-6273(91)90077-DPubMedCrossRef Erlander MG, Tillakaratne NJ, Feldblum S, Patel N, Tobin AJ: Two genes encode distinct glutamate decarboxylases. Neuron 1991, 7: 91–100. 10.1016/0896-6273(91)90077-DPubMedCrossRef
14.
Zurück zum Zitat Lopez-Bendito G, Sturgess K, Erdelyi F, Szabo G, Molnar Z, Paulsen O: Preferential origin and layer destination of GAD65-GFP cortical interneurons. Cereb Cortex 2004, 14: 1122–1133. 10.1093/cercor/bhh072PubMedCrossRef Lopez-Bendito G, Sturgess K, Erdelyi F, Szabo G, Molnar Z, Paulsen O: Preferential origin and layer destination of GAD65-GFP cortical interneurons. Cereb Cortex 2004, 14: 1122–1133. 10.1093/cercor/bhh072PubMedCrossRef
15.
Zurück zum Zitat Heinke B, Ruscheweyh R, Forsthuber L, Wunderbaldinger G, Sandkuhler J: Physiological, neurochemical and morphological properties of a subgroup of GABAergic spinal lamina II neurones identified by expression of green fluorescent protein in mice. J Physiol 2004, 560: 249–266. 10.1113/jphysiol.2004.070540PubMedCentralPubMedCrossRef Heinke B, Ruscheweyh R, Forsthuber L, Wunderbaldinger G, Sandkuhler J: Physiological, neurochemical and morphological properties of a subgroup of GABAergic spinal lamina II neurones identified by expression of green fluorescent protein in mice. J Physiol 2004, 560: 249–266. 10.1113/jphysiol.2004.070540PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Laing I, Todd AJ, Heizmann CW, Schmidt HH: Subpopulations of GABAergic neurons in laminae I-III of rat spinal dorsal horn defined by coexistence with classical transmitters, peptides, nitric oxide synthase or parvalbumin. Neuroscience 1994, 61: 123–132. 10.1016/0306-4522(94)90065-5PubMedCrossRef Laing I, Todd AJ, Heizmann CW, Schmidt HH: Subpopulations of GABAergic neurons in laminae I-III of rat spinal dorsal horn defined by coexistence with classical transmitters, peptides, nitric oxide synthase or parvalbumin. Neuroscience 1994, 61: 123–132. 10.1016/0306-4522(94)90065-5PubMedCrossRef
17.
Zurück zum Zitat Hantman AW, Pol AN, Perl ER: Morphological and physiological features of a set of spinal substantia gelatinosa neurons defined by green fluorescent protein expression. J Neurosci 2004, 24: 836–842. 10.1523/JNEUROSCI.4221-03.2004PubMedCrossRef Hantman AW, Pol AN, Perl ER: Morphological and physiological features of a set of spinal substantia gelatinosa neurons defined by green fluorescent protein expression. J Neurosci 2004, 24: 836–842. 10.1523/JNEUROSCI.4221-03.2004PubMedCrossRef
18.
Zurück zum Zitat Lu Y, Perl ER: A specific inhibitory pathway between substantia gelatinosa neurons receiving direct C-fiber input. J Neurosci 2003, 23: 8752–8758.PubMed Lu Y, Perl ER: A specific inhibitory pathway between substantia gelatinosa neurons receiving direct C-fiber input. J Neurosci 2003, 23: 8752–8758.PubMed
19.
Zurück zum Zitat Yasaka T, Kato G, Furue H, Rashid MH, Sonohata M, Tamae A, Murata Y, Masuko S, Yoshimura M: Cell-type-specific excitatory and inhibitory circuits involving primary afferents in the substantia gelatinosa of the rat spinal dorsal horn in vitro. J Physiol 2007, 581: 603–618. 10.1113/jphysiol.2006.123919PubMedCentralPubMedCrossRef Yasaka T, Kato G, Furue H, Rashid MH, Sonohata M, Tamae A, Murata Y, Masuko S, Yoshimura M: Cell-type-specific excitatory and inhibitory circuits involving primary afferents in the substantia gelatinosa of the rat spinal dorsal horn in vitro. J Physiol 2007, 581: 603–618. 10.1113/jphysiol.2006.123919PubMedCentralPubMedCrossRef
20.
Zurück zum Zitat Prescott SA, De Koninck Y: Four cell types with distinctive membrane properties and morphologies in lamina I of the spinal dorsal horn of the adult rat. J Physiol 2002, 539: 817–836. 10.1113/jphysiol.2001.013437PubMedCentralPubMedCrossRef Prescott SA, De Koninck Y: Four cell types with distinctive membrane properties and morphologies in lamina I of the spinal dorsal horn of the adult rat. J Physiol 2002, 539: 817–836. 10.1113/jphysiol.2001.013437PubMedCentralPubMedCrossRef
21.
Zurück zum Zitat Grudt TJ, Perl ER: Correlations between neuronal morphology and electrophysiological features in the rodent superficial dorsal horn. J Physiol 2002, 540: 189–207. 10.1113/jphysiol.2001.012890PubMedCentralPubMedCrossRef Grudt TJ, Perl ER: Correlations between neuronal morphology and electrophysiological features in the rodent superficial dorsal horn. J Physiol 2002, 540: 189–207. 10.1113/jphysiol.2001.012890PubMedCentralPubMedCrossRef
22.
Zurück zum Zitat Zeilhofer HU, Studler B, Arabadzisz D, Schweizer C, Ahmadi S, Layh B, Bosl MR, Fritschy JM: Glycinergic neurons expressing enhanced green fluorescent protein in bacterial artificial chromosome transgenic mice. J Comp Neurol 2005, 482: 123–141. 10.1002/cne.20349PubMedCrossRef Zeilhofer HU, Studler B, Arabadzisz D, Schweizer C, Ahmadi S, Layh B, Bosl MR, Fritschy JM: Glycinergic neurons expressing enhanced green fluorescent protein in bacterial artificial chromosome transgenic mice. J Comp Neurol 2005, 482: 123–141. 10.1002/cne.20349PubMedCrossRef
23.
Zurück zum Zitat Prescott SA, De Koninck Y, Sejnowski TJ: Biophysical basis for three distinct dynamical mechanisms of action potential initiation. PLoS Comput Biol 2008, 4: e1000198. 10.1371/journal.pcbi.1000198PubMedCentralPubMedCrossRef Prescott SA, De Koninck Y, Sejnowski TJ: Biophysical basis for three distinct dynamical mechanisms of action potential initiation. PLoS Comput Biol 2008, 4: e1000198. 10.1371/journal.pcbi.1000198PubMedCentralPubMedCrossRef
24.
Zurück zum Zitat Dougherty KJ, Sawchuk MA, Hochman S: Properties of mouse spinal lamina I GABAergic interneurons. J Neurophysiol 2005, 94: 3221–3227. 10.1152/jn.00184.2005PubMedCentralPubMedCrossRef Dougherty KJ, Sawchuk MA, Hochman S: Properties of mouse spinal lamina I GABAergic interneurons. J Neurophysiol 2005, 94: 3221–3227. 10.1152/jn.00184.2005PubMedCentralPubMedCrossRef
25.
Zurück zum Zitat Daniele CA, MacDermott AB: Low-threshold primary afferent drive onto GABAergic interneurons in the superficial dorsal horn of the mouse. J Neurosci 2009, 29: 686–695. 10.1523/JNEUROSCI.5120-08.2009PubMedCentralPubMedCrossRef Daniele CA, MacDermott AB: Low-threshold primary afferent drive onto GABAergic interneurons in the superficial dorsal horn of the mouse. J Neurosci 2009, 29: 686–695. 10.1523/JNEUROSCI.5120-08.2009PubMedCentralPubMedCrossRef
26.
Zurück zum Zitat Chery N, De Koninck Y: Junctional versus extrajunctional glycine and GABA(A) receptor-mediated IPSCs in identified lamina I neurons of the adult rat spinal cord. J Neurosci 1999, 19: 7342–7355.PubMed Chery N, De Koninck Y: Junctional versus extrajunctional glycine and GABA(A) receptor-mediated IPSCs in identified lamina I neurons of the adult rat spinal cord. J Neurosci 1999, 19: 7342–7355.PubMed
27.
Zurück zum Zitat Keller AF, Coull JA, Chery N, Poisbeau P, De Koninck Y: Region-specific developmental specialization of GABA-glycine cosynapses in laminas I-II of the rat spinal dorsal horn. J Neurosci 2001, 21: 7871–7880.PubMed Keller AF, Coull JA, Chery N, Poisbeau P, De Koninck Y: Region-specific developmental specialization of GABA-glycine cosynapses in laminas I-II of the rat spinal dorsal horn. J Neurosci 2001, 21: 7871–7880.PubMed
28.
Zurück zum Zitat Gonzalez-Forero D, Alvarez FJ: Differential postnatal maturation of GABAA, glycine receptor, and mixed synaptic currents in Renshaw cells and ventral spinal interneurons. J Neurosci 2005, 25: 2010–2023. 10.1523/JNEUROSCI.2383-04.2005PubMedCrossRef Gonzalez-Forero D, Alvarez FJ: Differential postnatal maturation of GABAA, glycine receptor, and mixed synaptic currents in Renshaw cells and ventral spinal interneurons. J Neurosci 2005, 25: 2010–2023. 10.1523/JNEUROSCI.2383-04.2005PubMedCrossRef
29.
Zurück zum Zitat Jensen K, Jensen MS, Lambert JD: Post-tetanic potentiation of GABAergic IPSCs in cultured rat hippocampal neurones. J Physiol 1999,519(Pt 1):71–84. 10.1111/j.1469-7793.1999.0071o.xPubMedCentralPubMedCrossRef Jensen K, Jensen MS, Lambert JD: Post-tetanic potentiation of GABAergic IPSCs in cultured rat hippocampal neurones. J Physiol 1999,519(Pt 1):71–84. 10.1111/j.1469-7793.1999.0071o.xPubMedCentralPubMedCrossRef
30.
Zurück zum Zitat Jensen K, Lambert JD, Jensen MS: Tetanus-induced asynchronous GABA release in cultured hippocampal neurons. Brain Res 2000, 880: 198–201. 10.1016/S0006-8993(00)02746-3PubMedCrossRef Jensen K, Lambert JD, Jensen MS: Tetanus-induced asynchronous GABA release in cultured hippocampal neurons. Brain Res 2000, 880: 198–201. 10.1016/S0006-8993(00)02746-3PubMedCrossRef
31.
Zurück zum Zitat Caillard O, Ben Ari Y, Gaiarsa JL: Long-term potentiation of GABAergic synaptic transmission in neonatal rat hippocampus. J Physiol 1999,518(Pt 1):109–119. 10.1111/j.1469-7793.1999.0109r.xPubMedCentralPubMedCrossRef Caillard O, Ben Ari Y, Gaiarsa JL: Long-term potentiation of GABAergic synaptic transmission in neonatal rat hippocampus. J Physiol 1999,518(Pt 1):109–119. 10.1111/j.1469-7793.1999.0109r.xPubMedCentralPubMedCrossRef
32.
Zurück zum Zitat Hefft S, Jonas P: Asynchronous GABA release generates long-lasting inhibition at a hippocampal interneuron-principal neuron synapse. Nat Neurosci 2005, 8: 1319–1328. 10.1038/nn1542PubMedCrossRef Hefft S, Jonas P: Asynchronous GABA release generates long-lasting inhibition at a hippocampal interneuron-principal neuron synapse. Nat Neurosci 2005, 8: 1319–1328. 10.1038/nn1542PubMedCrossRef
33.
Zurück zum Zitat Kato G, Yasaka T, Katafuchi T, Furue H, Mizuno M, Iwamoto Y, Yoshimura M: Direct GABAergic and glycinergic inhibition of the substantia gelatinosa from the rostral ventromedial medulla revealed by in vivo patch-clamp analysis in rats. J Neurosci 2006, 26: 1787–1794. 10.1523/JNEUROSCI.4856-05.2006PubMedCrossRef Kato G, Yasaka T, Katafuchi T, Furue H, Mizuno M, Iwamoto Y, Yoshimura M: Direct GABAergic and glycinergic inhibition of the substantia gelatinosa from the rostral ventromedial medulla revealed by in vivo patch-clamp analysis in rats. J Neurosci 2006, 26: 1787–1794. 10.1523/JNEUROSCI.4856-05.2006PubMedCrossRef
34.
Zurück zum Zitat Lu Y, Perl ER: Modular organization of excitatory circuits between neurons of the spinal superficial dorsal horn (laminae I and II). J Neurosci 2005, 25: 3900–3907. 10.1523/JNEUROSCI.0102-05.2005PubMedCrossRef Lu Y, Perl ER: Modular organization of excitatory circuits between neurons of the spinal superficial dorsal horn (laminae I and II). J Neurosci 2005, 25: 3900–3907. 10.1523/JNEUROSCI.0102-05.2005PubMedCrossRef
35.
Zurück zum Zitat Erdélyi F, Sekerkova G, Katarova Z, Hájos N, Pálhalmi J, Freund TF, Mugnaini E, Szabó G: GAD65-GFP Transgenic Mice Expressing GFP In The GABAergic Nervous System. FENS Abstr 2002, 1: A011.2. Erdélyi F, Sekerkova G, Katarova Z, Hájos N, Pálhalmi J, Freund TF, Mugnaini E, Szabó G: GAD65-GFP Transgenic Mice Expressing GFP In The GABAergic Nervous System. FENS Abstr 2002, 1: A011.2.
36.
Zurück zum Zitat Chery N, Yu XH, De Koninck Y: Visualization of lamina I of the dorsal horn in live adult rat spinal cord slices. J Neurosci Methods 2000, 96: 133–142. 10.1016/S0165-0270(99)00195-8PubMedCrossRef Chery N, Yu XH, De Koninck Y: Visualization of lamina I of the dorsal horn in live adult rat spinal cord slices. J Neurosci Methods 2000, 96: 133–142. 10.1016/S0165-0270(99)00195-8PubMedCrossRef
37.
Zurück zum Zitat Buhl EH, Lubke J: Intracellular lucifer yellow injection in fixed brain slices combined with retrograde tracing, light and electron microscopy. Neuroscience 1989, 28: 3–16. 10.1016/0306-4522(89)90227-3PubMedCrossRef Buhl EH, Lubke J: Intracellular lucifer yellow injection in fixed brain slices combined with retrograde tracing, light and electron microscopy. Neuroscience 1989, 28: 3–16. 10.1016/0306-4522(89)90227-3PubMedCrossRef
Metadaten
Titel
Inhibitory coupling between inhibitory interneurons in the spinal cord dorsal horn
verfasst von
Charalampos Labrakakis
Louis-Etienne Lorenzo
Cyril Bories
Alfredo Ribeiro-da-Silva
Yves De Koninck
Publikationsdatum
01.12.2009
Verlag
BioMed Central
Erschienen in
Molecular Pain / Ausgabe 1/2009
Elektronische ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-5-24

Weitere Artikel der Ausgabe 1/2009

Molecular Pain 1/2009 Zur Ausgabe

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Hinter dieser Appendizitis steckte ein Erreger

23.04.2024 Appendizitis Nachrichten

Schmerzen im Unterbauch, aber sonst nicht viel, was auf eine Appendizitis hindeutete: Ein junger Mann hatte Glück, dass trotzdem eine Laparoskopie mit Appendektomie durchgeführt und der Wurmfortsatz histologisch untersucht wurde.

Ärztliche Empathie hilft gegen Rückenschmerzen

23.04.2024 Leitsymptom Rückenschmerzen Nachrichten

Personen mit chronischen Rückenschmerzen, die von einfühlsamen Ärzten und Ärztinnen betreut werden, berichten über weniger Beschwerden und eine bessere Lebensqualität.

Mehr Schaden als Nutzen durch präoperatives Aussetzen von GLP-1-Agonisten?

23.04.2024 Operationsvorbereitung Nachrichten

Derzeit wird empfohlen, eine Therapie mit GLP-1-Rezeptoragonisten präoperativ zu unterbrechen. Eine neue Studie nährt jedoch Zweifel an der Notwendigkeit der Maßnahme.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.