Skip to main content
Erschienen in: Molecular Brain 1/2015

Open Access 01.12.2015 | Research

Heterogeneity of tremor mechanisms assessed by tremor-related cortical potential in mice

verfasst von: Young-Gyun Park, Jee Hyun Choi, Chungki Lee, Sehyun Kim, Youngsoo Kim, Ki-Young Chang, Sun Ha Paek, Daesoo Kim

Erschienen in: Molecular Brain | Ausgabe 1/2015

Abstract

Background

Identifying a neural circuit mechanism that is differentially involved in tremor would aid in the diagnosis and cure of such cases. Here, we demonstrate that tremor-related cortical potential (TRCP) is differentially expressed in two different mouse models of tremor.

Results

Hybrid tremor analysis of harmaline-induced and genetic tremor in mice revealed that two authentic tremor frequencies for each type of tremor were conserved and showed an opposite dependence on CaV3.1 T-type Ca2+ channels. Electroencephalogram recordings revealed that α1−/−;α1G-/- mice double-null for the GABA receptor α1 subunit (Gabra1) and CaV3.1 T-type Ca2+ channels (Cacna1g), in which the tremor caused by the absence of Gabra1 is potentiated by the absence of Cacna1g, showed a coherent TRCP that exhibited an onset that preceded the initiation of behavioral tremor by 3 ms. However, harmaline-induced tremor, which is known to be abolished by α1G−/−, showed no TRCP.

Conclusions

Our results demonstrate that the α1−/−;α1G−/− double-knockout tremor model is useful for studying cortical mechanisms of tremor.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YGP and DSK designed experiments, performed data analysis, and were primarily responsible for preparing the figures and manuscript. JHC, YK, and SHP designed electrophysiological experiment and participated in data analysis. CL and SK performed EEG data analysis. KYC performed pharmacogenetic experiments. All authors read and approved the final manuscript.
Abkürzungen
α1−/−
mice null for Gabra1
α1-/-;α1G-/-
mice double-null for Gabra1 and Cacna1g
EEG
Electroencephalography
EMG
Electromyography
ERP
Event-related potential
TRCP
Tremor-related cortical potential

Background

Tremor is an involuntary rhythmic oscillation of body parts [1,2]. Various mechanism ranges from reflex loop to central oscillation has been proposed for the origin of oscillatory activity generating tremor [1,2]. Animal models of tremor have been contributed to examine those hypotheses.
Harmaline is a plant-derived metabolite used to induce tremor in animals [3-5]. Harmaline tremor is an action-induced tremor and coherent with pathological oscillation in olivocerebellar pathway, which propagates to reticulospinal pathway and entrains muscles with synchronized oscillation [4,6,7]. Either knockout of the Cacna1g gene encoding the CaV3.1 channel α1G subunit (α1G−/− mice) or pharmacological blockage of T-type Ca2+ channels in the inferior olive abolishes 10–15 Hz harmaline-induced tremor and the pathological oscillation [8], suggesting that activity of T-type calcium channel is critical for generating harmaline tremor.
T-type calcium channel has a contradictory role in the other model of action-induced tremor. In Gabra1 model of action tremor, knockout mice of α1 subunit of the γ-aminobutyric acid (GABA) receptor, additional knockout of α1G enhances action-induced tremor [9]. Considering that T-type calcium channel is involved in thalamocortical oscillations [10,11] and cortical lesion has no effect on harmaline tremor [7,12], we hypothesized that two tremor models has different involvement of cortical mechanisms.
Here, using pharmacogenetic dissection of tremor frequencizes and high-resolution electroencephalograzphic (EEG) techniques [13] combining with event-related potential analysis(ERP) [14-17], we reveal that abnormal cortical rhythmicity in the form of tremor-related cortical potential (TRCP) is differentially expressed between harmaline and genetic models of action tremor.

Results

Pharmacogenetic determination of tremor frequency independence

To test the independence of mechanism between harmaline and genetic tremors (α1−/− and α1−/−;α1G−/−) [9], we performed pharmacogenetic experiment in which two types of tremor were produced in a mouse. We predicted two possibilities. The first is that the two tremor mechanisms interact to generate a hybrid tremor according to the dominant/recessive relationship between them. Alternatively, the two tremor frequencies may emerge independently, like the difference in behavior between two genes on separate chromosomes.
After harmaline administration (9 mg/kg), wild-type (WT) mice exhibited only 10–15 Hz tremor (Figure 1A-C, left), whereas α1−/−;α1G−/− mice showed only 15–30 Hz tremor (Figure 1A-C, right). In contrast, harmaline-treated α1−/− mice exhibited two separable band frequencies, 10–15 and 15–30 Hz, reflecting the coexistence of authentic harmaline and genetic tremor (Figure 1A-C, middle, and Figure 1D). Considering that the different tremor frequencies reflect the involvement of different neural circuit mechanisms [1,2], the two action tremor models appear to have distinct neural circuit mechanisms.
Using high-resolution EEG covering the whole frontal and occipital cortex (Figure 2A) and forelimb electromyography (EMG), we measured the cortical activity associated with forelimb tremor in α1−/−;α1G−/− mice. We isolated cortical potentials associated with tremor onset by averaging EEG activity corresponding to the first discharge of each tremor epoch. Tremor onset-related potentials in α1−/−;α1G−/− mice initiated approximately 3 ms prior to behavioral tremor onset and were followed by 15–30 Hz oscillations corresponding to the accompanying muscular tremor discharges (Figure 2B and C). The resulting TRCPs were phase-locked to the 15–30 Hz tremor (Figure 2B and C), although their amplitudes and phases were variable among channels (Figure 2B and C), indicating the differential involvement of cortical subdomains. Power spectral analyses also confirmed that TRCPs were initiated upon tremor onset (Figure 2D, upper, and 2E) and coincided with tremor events (Figure 2D, lower). These results indicate that the α1-/-;α1G−/− tremor includes the cortical circuit mechanism, although the origin remains unknown.

No TRCPs in harmaline tremor

We then compared TRCPs in the two tremor models. When we aligned and averaged EEG traces along the timing of tremor discharges in α1-/-;α1G−/− mice, TRCPs were obtained as an oscillatory pattern (Figure 3A, left). However, TRCPs were not found in harmaline tremor (Figure 3A, right) or in non-tremor contractions of α1−/−;α1G−/− mice (Figure 3A, middle). The power and coherence of TRCPs were significantly lower in harmaline tremor than in α1−/−;α1G−/− tremor (Figure 3B-D). These results suggest that TRCPs can be a robust measure for discriminating these two types of tremor models.

Discussion

Essential tremor is a neurological condition characterized by action-induced tremors [2,19]. Considerable evidence suggests that essential tremor is heterogeneous with respect to affected body parts, tremor frequency, and drug sensitivity [20-24]. The heterogeneity appears to reflect the involvement of different neural substrates. While some studies have reported that abnormal cortical rhythms are associated with tremor in a subset of essential tremor patients [25,26], another clinical study failed to find such a correlation [27], necessitating in-depth comparative studies on cortical involvement in the generation of essential tremor [25-28].
Two action-induced tremor models used in present study has been proposed as an animal models of essential tremor [4,9]. By employing event-related potential analysis, we identified TRCPs in α1-/-;α1G-/- mice. However, we failed to find out tremor-related activity in harmaline tremor, suggesting that two models of tremor have different cortical dependency.
Whereas harmaline and α1−/− tremors are ameliorated by propranolol [29,30], α1−/−;α1G−/− tremor is resistant to propranolol [9]. Deuschl et al. reported that the patients with propranolol susceptibility show synchronous activity between agonist and antagonist muscles during tremor, whereas the patients with propranolol resistance exhibit reciprocal antagonist muscle activity [20]. Notably, harmaline tremor, which is sensitive to propranolol [30], also shows co-contraction of agonist and antagonist muscle pairs during tremor [31]. Thus, it is plausible that the TRCP serves a role in generating propranolol-resistant or reciprocal agonist–antagonist contracting subtypes of essential tremor. A comparative study on the cortical activity of essential tremor patients with different phenotypes would effectively test this hypothesis.
The two essential tremor models in this study are also different with respect to their underlying neural circuit mechanism. The CaV3.1 channel is expressed in various motor pathways and may serve different functions [32]. In the thalamocortical pathway, where both Gabra1 and Ca V 3.1 genes are co-expressed [33], CaV3.1 may play an inhibitory role in tremorogenesis. In contrast, CaV3.1 is involved in the generation of tremor in the inferior olive nucleus [8,32]. Inferior olive neurons are connected to muscles through the cerebelloreticulospinal pathway [6,34]. This means that harmaline could induce tremor while bypassing the involvement of the thalamocortical pathway. Indeed, evidence shows that neither the thalamus nor the motor cortex is associated with harmaline-induced tremor [7,12].
The TRCP in α1-/-;α1G-/- mice is not merely a sensory feedback or a movement artifact for a number of reasons. First, it occurred prior to the onset of tremor, at least in some channels, suggesting the possibility that TRCPs are involved in the mechanism responsible for tremor onset (Figure 2). Moreover, TRCPs were not observed in harmaline tremor, which have an intensity similar to that of α1−/−;α1G−/− tremor (Figure 3). Although tremoring muscle is known to provide sensory feedback to the cortex, it is not likely that our method is sensitive enough to detect the low level of associated neural activity. These TRCP findings suggest that the two types of tremor are differentially associated with physiological and neural circuit mechanisms.
In conclusion, we have demonstrated the presence of TRCP only in a certain animal model of essential tremor, suggesting its mechanistic heterogeneity. The TRCP characterized in our study could be a useful readout for the diagnosis of essential tremor type. Notably, in patients showing a high level of TRCP, therapeutic strategies that target thalamocortical pathways, rather than T-type Ca2+ channel blockers, might be effective. Furthermore, the α1−/−;α1G−/− mouse should be a useful model for revealing how the cerebral cortex can generate tremor.

Methods

Animals

Animal care and handling conformed to the institutional guidelines of the Animal Care and Use Committee of the Korea Advanced Institute of Science and Technology (KAIST). Mice heterozygous for GABAA receptor α1 deletion (α1+/−) in a C57BL/6 J genetic background were obtained from Dr. Gregg E. Homanics at the University of Pittsburgh. Crossing α1+/− and α1G+/− mice yielded double-heterozygous animals (α1+/−;α1G+/−), which were mated to produce WT (α1+/+;α1G+/+), α1 mutant (α1−/−;α1G+/+), and double-mutant (α1−/−;α1G−/−) mice. WT mice were utilized for the harmaline-induced tremor model. All experiments were performed using adult mice (>10 weeks old). Mice were housed under a 12-hour light/dark cycle (7 a.m. to 7 p.m.) with free access to water and food.

High-resolution EEG

EEG electrode arrays were prepared as previously described [13]. For surgical anesthesia, ketamine (100 mg/kg) mixed with xylazine (5 mg/kg) was administered intraperitoneally. After surgical anesthesia had developed, the head of the mouse was fixed on a stereotaxic apparatus (David Kopf Instruments, CA, USA), and an incision of ~2.5 cm was made on the scalp. The skull was exposed using micro-clamps, and tissue debris was removed by carefully wiping with a 70% ethanol-soaked cotton ball. After moisturizing the skull surface with a drop of tap water, a polyamide-based, high-density film electrode was placed on the skull and carefully positioned so that the vertical midline was aligned with the skull midline and the bregma met the upper edge of the third branch. After positioning the electrode, two screws were implanted onto the skull, and dental acrylate was coated over the electrode array. The viscosity of the dental acrylate mix used was carefully examined before application. Mice were allowed to recover under an infrared lamp and housed singly until the experiment.

Electromyography

The electrodes for electromyography (EMG) in mice were made with Teflon-coated stainless steel wires (A-M Systems, WA, USA) and implanted as previously described [35]. Briefly, two wires were combined and twisted together with a knot 4 cm from the end. The Teflon insulation was removed from 0.5-mm-long segments 1 or 2 mm from the knot in both wires to create two exposures located 1 mm apart. The ends of both wires were crumpled into the shaft of a 26-gauge needle. The opposite ends of the wires were cut ~8 cm from the knot, bared, and soldered to a connector. After the fur in the dorsal neck region was shaved away, small incisions were made in the skin above the left and right forelimb muscles (triceps brachii). EMG electrodes were inserted under the skin from the neck incision to the muscles. The needle harboring the electrode was inserted into the target muscle until the knot proximal to the bared regions was firmly butted against the surface of the muscle. The distal end of the electrode exiting the muscle was loosely knotted, and the knot was moved to the muscle surface and tightened. Incisions in the limbs were closed with silk suturing material.

Analysis of tremor

After habituation in the recording room for 30 minutes, the mouse was placed on a stainless steel plate (25 cm diameter) suspended by two elastic rubber bands. The edge of the plate was surrounded by paper (5 cm wide) to prevent mice from falling off. The behavior of the mouse was monitored using a video camera positioned above the platform. For induction of harmaline tremor, harmaline hydrochloride dissolved in 0.9% saline was injected intraperitoneally (15 mg/kg). A DC response accelerometer (Model No. 3711D1FA3G; Piezotronics, NY, USA) was mounted below the steel plate to aid in tremor detection.

Acquisition of EEG and EMG signals

High-resolution EEG, EMG, and accelerometer signals were obtained using a SynAmps2 amplifier (Neuroscan Inc., TX, USA) with a 2000-Hz sampling rate. Channel 34 of the EEG was used as a reference and channel 33 was used as a ground. Contact impedances of EEG channels were measured using Scan 4.3 software (Neuroscan Inc.) with 30-Hz test pulses. EEG channels with abnormal impedance (>500 kOhm) were excluded from further analyses.

Pre-processing

All signal processing and analyses were performed using the Matlab-based open source toolbox EEGLAB [36] or custom Matlab codes. Signals were loaded onto EEGLAB and filtered with a 100–Hz high-pass filter for EMG signals and a 1–200 Hz band-pass filter for EEG and accelerometer signals. All filters used were finite-impulse response filters. EEG signals were re-referenced by common average reference using an EEGLAB plug-in algorithm.

Tremor detection

EMG traces obtained from the left or right forelimb triceps brachii muscle were used for measuring tremor. Raw EMG traces were rectified and smoothed using a moving-average window. A cluster of EMG action potentials with peak amplitudes 2.5-fold greater than the standard deviation of baseline EMG activity and separated from others by more than 25 ms was defined as an ‘EMG discharge’. Rhythmic and successive EMG discharges in a specific frequency range (15–30 Hz for α1−/−;α1G−/− and 10–15 Hz for harmaline tremor) were identified as a ‘tremor epoch’. Each EMG discharge in a tremor epoch was defined as a ‘tremor discharge’. The ‘tremor onset’ was determined as the onset time of the first tremor discharge in a tremor epoch. Irregular EMG discharges temporally separated from the tremor epoch by more than 1 second in α1−/−;α1G−/− mice were identified as ‘non-tremor discharges’. Tremor onset is used for ERP analysis in Figure 2. However, we used tremor discharge itself for comparing TRCP in two tremor models (Figure 3) because single tremor epoch is much longer in harmaline tremor so that it was hard to collect enough number of tremor epochs for ERP analysis. Event detection was performed in the left and right forelimb independently using Matlab codes, and was confirmed by visual inspection.
EEG channel activity during each event (500 ms pre- and post-event) was collected, and a baseline correction was applied to the pre-event period for each event and channel. For the calculation of ERPs, the waveforms of collected events (>100 for tremor discharge and non-tremor discharge, and >50 for tremor onset) were averaged, and the ERP from each channel was plotted in a different color. ERPs of tremor onset, tremor discharge and non-tremor discharge for α1−/−;α1G−/− and harmaline tremor models were calculated, and ERPs of tremor onset and tremor discharge were identified as TRCPs. Tremor events that occurred in only one forelimb at a given moment (>0.5 seconds apart from contralateral limb tremor events) were used for ERP calculations. Inter-trial coherence is a frequency-domain measure of activity synchronization between event-related trials [36]. Inter-trial coherence value indicates degree of consistency of oscillatory phase across trials at certain latency and frequencym [37] and calculated by EEGLab [36].

Statistical analysis

All data are presented as mean values ± SEM, and differences between experimental groups were analyzed for statistical significance using Sigmastat 3.1.

Acknowledgements

This work was supported by grants from the National Leading Research Laboratory Program and KAIST Future Systems Healthcare Project of the Ministry of Science, ICT and Future Planning (No. 20120008795, 2012 K001117, to D. Kim), the Global Frontier R&D Program (NRF-M1AXA003-2011-0031525, to J-H Choi), and Korea Healthcare Technology R&D Project, Ministry of Health & Welfare and the Korea Institute of Planning & Evaluation for Technology in Food, Agriculture, Forestry, and Fisheries (No. HI12C02050101 and 311011-05-3-SB020, to S. H. Paek).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YGP and DSK designed experiments, performed data analysis, and were primarily responsible for preparing the figures and manuscript. JHC, YK, and SHP designed electrophysiological experiment and participated in data analysis. CL and SK performed EEG data analysis. KYC performed pharmacogenetic experiments. All authors read and approved the final manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Deuschl G, Raethjen J, Lindemann M, Krack P. The pathophysiology of tremor. Muscle Nerve. 2001;24:716–35.PubMedCrossRef Deuschl G, Raethjen J, Lindemann M, Krack P. The pathophysiology of tremor. Muscle Nerve. 2001;24:716–35.PubMedCrossRef
3.
Zurück zum Zitat Deuschl G, Elble RJ. The pathophysiology of essential tremor. Neurology. 1999;54:S14–20. Deuschl G, Elble RJ. The pathophysiology of essential tremor. Neurology. 1999;54:S14–20.
4.
Zurück zum Zitat Le Doux M. Harmaline Tremor. Movement Disorders: Genetics and Models 2005;361-68. Le Doux M. Harmaline Tremor. Movement Disorders: Genetics and Models 2005;361-68.
5.
Zurück zum Zitat Poirier L, Sourkes T, Bouvier G, Boucher R, Carabin S. Striatal amines, experimental tremor and the effect of harmaline in the monkey. Brain. 1966;89:37–52.PubMedCrossRef Poirier L, Sourkes T, Bouvier G, Boucher R, Carabin S. Striatal amines, experimental tremor and the effect of harmaline in the monkey. Brain. 1966;89:37–52.PubMedCrossRef
6.
Zurück zum Zitat De Montigny C, Lamarre Y. Rhythmic activity induced by harmaline in the olivo-cerebello-bulbar system of the cat. Brain Res. 1973;53:81–95.PubMedCrossRef De Montigny C, Lamarre Y. Rhythmic activity induced by harmaline in the olivo-cerebello-bulbar system of the cat. Brain Res. 1973;53:81–95.PubMedCrossRef
7.
Zurück zum Zitat Lamarre Y, Joffroy A, Dumont M, De Montigny C, Grou F, Lund J. Central mechanisms of tremor in some feline and primate models. Can J Neurol Sci. 1975;2:227–33.PubMed Lamarre Y, Joffroy A, Dumont M, De Montigny C, Grou F, Lund J. Central mechanisms of tremor in some feline and primate models. Can J Neurol Sci. 1975;2:227–33.PubMed
8.
Zurück zum Zitat Park Y-G, Park H-Y, Lee CJ, Choi S, Jo S, Choi H, et al. CaV3. 1 is a tremor rhythm pacemaker in the inferior olive. Proc Natl Acad Sci. 2010;107:10731–6.PubMedCentralPubMedCrossRef Park Y-G, Park H-Y, Lee CJ, Choi S, Jo S, Choi H, et al. CaV3. 1 is a tremor rhythm pacemaker in the inferior olive. Proc Natl Acad Sci. 2010;107:10731–6.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Chang K-Y, Park Y-G, Park H-Y, Homanics GE, Kim J, Kim D. Lack of CaV3.1 channels causes severe motor coordination defects and an age-dependent cerebellar atrophy in a genetic model of essential tremor. Biochem Biophys Res Commun. 2011;410:19–23.PubMedCrossRef Chang K-Y, Park Y-G, Park H-Y, Homanics GE, Kim J, Kim D. Lack of CaV3.1 channels causes severe motor coordination defects and an age-dependent cerebellar atrophy in a genetic model of essential tremor. Biochem Biophys Res Commun. 2011;410:19–23.PubMedCrossRef
10.
Zurück zum Zitat Huguenard J. Low-threshold calcium currents in central nervous system neurons. Annu Rev Physiol. 1996;58:329–48.PubMedCrossRef Huguenard J. Low-threshold calcium currents in central nervous system neurons. Annu Rev Physiol. 1996;58:329–48.PubMedCrossRef
11.
Zurück zum Zitat Kim D, Song I, Keum S, Lee T, Jeong MJ, Kim SS, et al. Lack of the burst firing of thalamocortical relay neurons and resistance to absence seizures in mice lacking α1G T-type Ca2+ channels. Neuron. 2001;31:35–45.PubMedCrossRef Kim D, Song I, Keum S, Lee T, Jeong MJ, Kim SS, et al. Lack of the burst firing of thalamocortical relay neurons and resistance to absence seizures in mice lacking α1G T-type Ca2+ channels. Neuron. 2001;31:35–45.PubMedCrossRef
12.
Zurück zum Zitat Battista AF, Nakatani S, Goldstein M, Anagnoste B. Effect of harmaline in monkeys with central nervous system lesions. Exp Neurol. 1970;28:513–24.PubMedCrossRef Battista AF, Nakatani S, Goldstein M, Anagnoste B. Effect of harmaline in monkeys with central nervous system lesions. Exp Neurol. 1970;28:513–24.PubMedCrossRef
13.
Zurück zum Zitat Choi JH, Koch KP, Poppendieck W, Lee M, Shin H-S. High resolution electroencephalography in freely moving mice. J Neurophysiol. 2010;104:1825–34.PubMedCrossRef Choi JH, Koch KP, Poppendieck W, Lee M, Shin H-S. High resolution electroencephalography in freely moving mice. J Neurophysiol. 2010;104:1825–34.PubMedCrossRef
14.
Zurück zum Zitat Handy TC. Event-related potentials: a methods handbook. Cambridge, MA: MIT press; 2005. Handy TC. Event-related potentials: a methods handbook. Cambridge, MA: MIT press; 2005.
15.
Zurück zum Zitat Johnson R. Event-related potential insights into the neurobiology of memory systems. Handbook Neuropsychol. 1995;10:135-135. Johnson R. Event-related potential insights into the neurobiology of memory systems. Handbook Neuropsychol. 1995;10:135-135.
16.
Zurück zum Zitat Luck SJ. An introduction to the event-related potential technique. MA: MIT press Cambridge; 2005. Luck SJ. An introduction to the event-related potential technique. MA: MIT press Cambridge; 2005.
17.
Zurück zum Zitat Walter W, Cooper R, Aldridge V, McCallum W, Winter A. Contingent negative variation: an electric sign of sensori-motor association and expectancy in the human brain. Nature. 1964;203:380–4.PubMedCrossRef Walter W, Cooper R, Aldridge V, McCallum W, Winter A. Contingent negative variation: an electric sign of sensori-motor association and expectancy in the human brain. Nature. 1964;203:380–4.PubMedCrossRef
18.
Zurück zum Zitat Lee M, Kim D, Shin H-S, Sung H-G, Choi JH. High-density EEG recordings of the freely moving mice using polyimide-based microelectrode. J Visualized Exp. 2011;47:1–4. Lee M, Kim D, Shin H-S, Sung H-G, Choi JH. High-density EEG recordings of the freely moving mice using polyimide-based microelectrode. J Visualized Exp. 2011;47:1–4.
20.
Zurück zum Zitat Deuschl G, Lücking C, Schenck E. Essential tremor: electrophysiological and pharmacological evidence for a subdivision. J Neurol Neurosurg Psychiatry. 1987;50:1435–41.PubMedCentralPubMedCrossRef Deuschl G, Lücking C, Schenck E. Essential tremor: electrophysiological and pharmacological evidence for a subdivision. J Neurol Neurosurg Psychiatry. 1987;50:1435–41.PubMedCentralPubMedCrossRef
21.
22.
Zurück zum Zitat Louis E, Vonsattel J, Honig L, Ross G, Lyons K, Pahwa R. Neuropathologic findings in essential tremor. Neurology. 2006;66:1756–9.PubMedCrossRef Louis E, Vonsattel J, Honig L, Ross G, Lyons K, Pahwa R. Neuropathologic findings in essential tremor. Neurology. 2006;66:1756–9.PubMedCrossRef
23.
Zurück zum Zitat Louis ED, Ford B, Barnes LF. Clinical subtypes of essential tremor. Arch Neurol. 2000;57:1194.PubMedCrossRef Louis ED, Ford B, Barnes LF. Clinical subtypes of essential tremor. Arch Neurol. 2000;57:1194.PubMedCrossRef
24.
Zurück zum Zitat Testa CM. Key issues in essential tremor genetics research: Where are we now and how can we move forward? Tremor Other Hyperkinetic Movements. 2013;3:1–19. Testa CM. Key issues in essential tremor genetics research: Where are we now and how can we move forward? Tremor Other Hyperkinetic Movements. 2013;3:1–19.
25.
Zurück zum Zitat Hellwig B, Häußler S, Schelter B, Lauk M, Guschlbauer B, Timmer J, et al. Tremor-correlated cortical activity in essential tremor. Lancet. 2001;357:519–23.PubMedCrossRef Hellwig B, Häußler S, Schelter B, Lauk M, Guschlbauer B, Timmer J, et al. Tremor-correlated cortical activity in essential tremor. Lancet. 2001;357:519–23.PubMedCrossRef
26.
Zurück zum Zitat Raethjen J, Govindan R, Kopper F, Muthuraman M, Deuschl G. Cortical involvement in the generation of essential tremor. J Neurophysiol. 2007;97:3219–28.PubMedCrossRef Raethjen J, Govindan R, Kopper F, Muthuraman M, Deuschl G. Cortical involvement in the generation of essential tremor. J Neurophysiol. 2007;97:3219–28.PubMedCrossRef
27.
Zurück zum Zitat Halliday D, Conway B, Farmer S, Shahani U, Russell A, Rosenberg J. Coherence between low-frequency activation of the motor cortex and tremor in patients with essential tremor. Lancet. 2000;355:1149–53.PubMedCrossRef Halliday D, Conway B, Farmer S, Shahani U, Russell A, Rosenberg J. Coherence between low-frequency activation of the motor cortex and tremor in patients with essential tremor. Lancet. 2000;355:1149–53.PubMedCrossRef
28.
Zurück zum Zitat Schnitzler A, Münks C, Butz M, Timmermann L, Gross J. Synchronized brain network associated with essential tremor as revealed by magnetoencephalography. Mov Disord. 2009;24:1629–35.PubMedCrossRef Schnitzler A, Münks C, Butz M, Timmermann L, Gross J. Synchronized brain network associated with essential tremor as revealed by magnetoencephalography. Mov Disord. 2009;24:1629–35.PubMedCrossRef
29.
Zurück zum Zitat Kralic JE, Criswell HE, Osterman JL, O’Buckley TK, Wilkie ME, Matthews DB, et al. Genetic essential tremor in γ-aminobutyric acidA receptor α1 subunit knockout mice. J Clin Investig. 2005;115:774–9.PubMedCentralPubMedCrossRef Kralic JE, Criswell HE, Osterman JL, O’Buckley TK, Wilkie ME, Matthews DB, et al. Genetic essential tremor in γ-aminobutyric acidA receptor α1 subunit knockout mice. J Clin Investig. 2005;115:774–9.PubMedCentralPubMedCrossRef
30.
Zurück zum Zitat Paterson NE, Malekiani SA, Foreman MM, Olivier B, Hanania T. Pharmacological characterization of harmaline-induced tremor activity in mice. Eur J Pharmacol. 2009;616:73–80.PubMedCrossRef Paterson NE, Malekiani SA, Foreman MM, Olivier B, Hanania T. Pharmacological characterization of harmaline-induced tremor activity in mice. Eur J Pharmacol. 2009;616:73–80.PubMedCrossRef
31.
Zurück zum Zitat Milner TE, Cadoret G, Lessard L, Smith AM. EMG analysis of harmaline-induced tremor in normal and three strains of mutant mice with Purkinje cell degeneration and the role of the inferior olive. J Neurophysiol. 1995;73:2568–77.PubMed Milner TE, Cadoret G, Lessard L, Smith AM. EMG analysis of harmaline-induced tremor in normal and three strains of mutant mice with Purkinje cell degeneration and the role of the inferior olive. J Neurophysiol. 1995;73:2568–77.PubMed
33.
Zurück zum Zitat Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2006;445:168–76.PubMedCrossRef Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2006;445:168–76.PubMedCrossRef
34.
Zurück zum Zitat Lamarre Y, Mercier L-A. Neurophysiological studies of harmaline-induced tremor in the cat. Can J Physiol Pharmacol. 1971;49:1049–58.PubMedCrossRef Lamarre Y, Mercier L-A. Neurophysiological studies of harmaline-induced tremor in the cat. Can J Physiol Pharmacol. 1971;49:1049–58.PubMedCrossRef
35.
Zurück zum Zitat Pearson K, Acharya H, Fouad K. A new electrode configuration for recording electromyographic activity in behaving mice. J Neurosci Methods. 2005;148:36–42.PubMedCrossRef Pearson K, Acharya H, Fouad K. A new electrode configuration for recording electromyographic activity in behaving mice. J Neurosci Methods. 2005;148:36–42.PubMedCrossRef
36.
Zurück zum Zitat Delorme A, Makeig S. EEGLAB: an open source toolbox for analysis of single-trial EEG dynamics including independent component analysis. J Neurosci Methods. 2004;134:9–21.PubMedCrossRef Delorme A, Makeig S. EEGLAB: an open source toolbox for analysis of single-trial EEG dynamics including independent component analysis. J Neurosci Methods. 2004;134:9–21.PubMedCrossRef
37.
Zurück zum Zitat Shin Y-W, Krishnan G, Hetrick WP, Brenner CA, Shekhar A, Malloy FW, et al. Increased temporal variability of auditory event-related potentials in schizophrenia and Schizotypal Personality Disorder. Schizophr Res. 2010;124:110–8.PubMedCentralPubMedCrossRef Shin Y-W, Krishnan G, Hetrick WP, Brenner CA, Shekhar A, Malloy FW, et al. Increased temporal variability of auditory event-related potentials in schizophrenia and Schizotypal Personality Disorder. Schizophr Res. 2010;124:110–8.PubMedCentralPubMedCrossRef
Metadaten
Titel
Heterogeneity of tremor mechanisms assessed by tremor-related cortical potential in mice
verfasst von
Young-Gyun Park
Jee Hyun Choi
Chungki Lee
Sehyun Kim
Youngsoo Kim
Ki-Young Chang
Sun Ha Paek
Daesoo Kim
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Molecular Brain / Ausgabe 1/2015
Elektronische ISSN: 1756-6606
DOI
https://doi.org/10.1186/s13041-015-0093-2

Weitere Artikel der Ausgabe 1/2015

Molecular Brain 1/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Update Neurologie

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