17 results on '"Yakhnitsa, V."'
Search Results
2. Spontaneous Activity Does Not Predict Morphological Type in Cerebellar Interneurons
- Author
-
Haar, S., primary, Givon-Mayo, R., additional, Barmack, N. H., additional, Yakhnitsa, V., additional, and Donchin, O., additional
- Published
- 2015
- Full Text
- View/download PDF
3. Modulated discharge of Purkinje and stellate cells persists after unilateral loss of vestibular primary afferent mossy fibers in mice
- Author
-
Barmack, N. H., primary and Yakhnitsa, V., additional
- Published
- 2013
- Full Text
- View/download PDF
4. Microlesions of the Inferior Olive Reduce Vestibular Modulation of Purkinje Cell Complex and Simple Spikes in Mouse Cerebellum
- Author
-
Barmack, N. H., primary and Yakhnitsa, V., additional
- Published
- 2011
- Full Text
- View/download PDF
5. Functions of Interneurons in Mouse Cerebellum
- Author
-
Barmack, N. H., primary and Yakhnitsa, V., additional
- Published
- 2008
- Full Text
- View/download PDF
6. Cerebellar Nodulectomy Impairs Spatial Memory of Vestibular and Optokinetic Stimulation in Rabbits
- Author
-
Barmack, N. H., primary, Errico, P., additional, Ferraresi, A., additional, Fushiki, H., additional, Pettorossi, V. E., additional, and Yakhnitsa, V., additional
- Published
- 2002
- Full Text
- View/download PDF
7. Vestibular Signals in the Parasolitary Nucleus
- Author
-
Barmack, N. H., primary and Yakhnitsa, V., additional
- Published
- 2000
- Full Text
- View/download PDF
8. Beneficial Effects of Ginger Root Extract on Pain Behaviors, Inflammation, and Mitochondrial Function in the Colon and Different Brain Regions of Male and Female Neuropathic Rats: A Gut-Brain Axis Study.
- Author
-
Santos JM, Deshmukh H, Elmassry MM, Yakhnitsa V, Ji G, Kiritoshi T, Presto P, Antenucci N, Liu X, Neugebauer V, and Shen CL
- Subjects
- Animals, Male, Female, Rats, Inflammation, Behavior, Animal drug effects, Plant Roots chemistry, Neuroinflammatory Diseases drug therapy, Disease Models, Animal, Zingiber officinale chemistry, Mitochondria drug effects, Mitochondria metabolism, Colon metabolism, Colon drug effects, Neuralgia drug therapy, Neuralgia metabolism, Brain metabolism, Brain drug effects, Plant Extracts pharmacology, Rats, Sprague-Dawley, Brain-Gut Axis drug effects
- Abstract
Background: Neuroinflammation and mitochondrial dysfunction have been implicated in the progression of neuropathic pain (NP) but can be mitigated by supplementation with gingerol-enriched ginger (GEG). However, the exact benefits of GEG for each sex in treating neuroinflammation and mitochondrial homeostasis in different brain regions and the colon remain to be determined., Objective: Evaluate the effects of GEG on emotional/affective pain and spontaneous pain behaviors, neuroinflammation, as well as mitochondria homeostasis in the amygdala, frontal cortex, hippocampus, and colon of male and female rats in the spinal nerve ligation (SNL) NP model., Methods: One hundred rats (fifty males and fifty females) were randomly assigned to five groups: sham + vehicle, SNL + vehicle, and SNL with three different GEG doses (200, 400, and 600 mg/kg BW) for 5 weeks. A rat grimace scale and vocalizations were used to assess spontaneous and emotional/affective pain behaviors, respectively. mRNA gene and protein expression levels for tight junction protein, neuroinflammation, mitochondria homeostasis, and oxidative stress were measured in the amygdala, frontal cortex, hippocampus, and colon using qRT-PCR and Western blot (colon)., Results: GEG supplementation mitigated spontaneous pain in both male and female rats with NP while decreasing emotional/affective responses only in male NP rats. GEG supplementation increased intestinal integrity (claudin 3) and suppressed neuroinflammation [glial activation (GFAP, CD11b, IBA1) and inflammation (TNFα, NFκB, IL1β)] in the selected brain regions and colon of male and female NP rats. GEG supplementation improved mitochondrial homeostasis [increased biogenesis (TFAM, PGC1α), increased fission (FIS, DRP1), decreased fusion (MFN2, MFN1) and mitophagy (PINK1), and increased Complex III] in the selected brain regions and colon in both sexes. Some GEG dose-response effects in gene expression were observed in NP rats of both sexes., Conclusions: GEG supplementation decreased emotional/affective pain behaviors of males and females via improving gut integrity, suppressing neuroinflammation, and improving mitochondrial homeostasis in the amygdala, frontal cortex, hippocampus, and colon in both male and female SNL rats in an NP model, implicating the gut-brain axis in NP. Sex differences observed in the vocalizations assay may suggest different mechanisms of evoked NP responses in females.
- Published
- 2024
- Full Text
- View/download PDF
9. Cells and circuits for amygdala neuroplasticity in the transition to chronic pain.
- Author
-
Kiritoshi T, Yakhnitsa V, Singh S, Wilson TD, Chaudhry S, Neugebauer B, Torres-Rodriguez JM, Lin JL, Carrasquillo Y, and Neugebauer V
- Subjects
- Animals, Neuralgia physiopathology, Neurons physiology, Mice, Male, Mice, Inbred C57BL, Neuronal Plasticity physiology, Chronic Pain physiopathology, Amygdala physiopathology
- Abstract
Maladaptive plasticity is linked to the chronification of diseases such as pain, but the transition from acute to chronic pain is not well understood mechanistically. Neuroplasticity in the central nucleus of the amygdala (CeA) has emerged as a mechanism for sensory and emotional-affective aspects of injury-induced pain, although evidence comes from studies conducted almost exclusively in acute pain conditions and agnostic to cell type specificity. Here, we report time-dependent changes in genetically distinct and projection-specific CeA neurons in neuropathic pain. Hyperexcitability of CRF projection neurons and synaptic plasticity of parabrachial (PB) input at the acute stage shifted to hyperexcitability without synaptic plasticity in non-CRF neurons at the chronic phase. Accordingly, chemogenetic inhibition of the PB→CeA pathway mitigated pain-related behaviors in acute, but not chronic, neuropathic pain. Cell-type-specific temporal changes in neuroplasticity provide neurobiological evidence for the clinical observation that chronic pain is not simply the prolonged persistence of acute pain., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Dysfunction of Small-Conductance Ca 2+ -Activated Potassium (SK) Channels Drives Amygdala Hyperexcitability and Neuropathic Pain Behaviors: Involvement of Epigenetic Mechanisms.
- Author
-
Yakhnitsa V, Thompson J, Ponomareva O, Ji G, Kiritoshi T, Mahimainathan L, Molehin D, Pruitt K, and Neugebauer V
- Subjects
- Animals, Male, Rats, Behavior, Animal drug effects, DNA Methylation genetics, Neurons metabolism, Rats, Sprague-Dawley, Amygdala metabolism, Amygdala physiopathology, Epigenesis, Genetic, Neuralgia metabolism, Neuralgia genetics, Neuralgia physiopathology, Small-Conductance Calcium-Activated Potassium Channels metabolism, Small-Conductance Calcium-Activated Potassium Channels genetics
- Abstract
Neuroplasticity in the amygdala and its central nucleus (CeA) is linked to pain modulation and pain behaviors, but cellular mechanisms are not well understood. Here, we addressed the role of small-conductance Ca
2+ -activated potassium (SK) channels in pain-related amygdala plasticity. The facilitatory effects of the intra-CeA application of an SK channel blocker (apamin) on the pain behaviors of control rats were lost in a neuropathic pain model, whereas an SK channel activator (NS309) inhibited pain behaviors in neuropathic rats but not in sham controls, suggesting the loss of the inhibitory behavioral effects of amygdala SK channels. Brain slice electrophysiology found hyperexcitability of CeA neurons in the neuropathic pain condition due to the loss of SK channel-mediated medium afterhyperpolarization (mAHP), which was accompanied by decreased SK2 channel protein and mRNA expression, consistent with a pretranscriptional mechanisms. The underlying mechanisms involved the epigenetic silencing of the SK2 gene due to the increased DNA methylation of the CpG island of the SK2 promoter region and the change in methylated CpG sites in the CeA in neuropathic pain. This study identified the epigenetic dysregulation of SK channels in the amygdala (CeA) as a novel mechanism of neuropathic pain-related plasticity and behavior that could be targeted to control abnormally enhanced amygdala activity and chronic neuropathic pain.- Published
- 2024
- Full Text
- View/download PDF
11. Ginger Polyphenols Reverse Molecular Signature of Amygdala Neuroimmune Signaling and Modulate Microbiome in Male Rats with Neuropathic Pain: Evidence for Microbiota-Gut-Brain Axis.
- Author
-
Shen CL, Santos JM, Elmassry MM, Bhakta V, Driver Z, Ji G, Yakhnitsa V, Kiritoshi T, Lovett J, Hamood AN, Sang S, and Neugebauer V
- Abstract
Emerging evidence shows that the gut microbiota plays an important role in neuropathic pain (NP) via the gut-brain axis. Male rats were divided into sham, spinal nerve ligation (SNL), SNL + 200 mg GEG/kg BW (GEG200), and SNL + 600 mg GEG/kg BW (GEG600) for 5 weeks. The dosages of 200 and 600 mg GEG/kg BW for rats correspond to 45 g and 135 g raw ginger for human daily consumption, respectively. Both GEG groups mitigated SNL-induced NP behavior. GEG-supplemented animals had a decreased abundance of Rikenella , Muribaculaceae , Clostridia UCG-014 , Mucispirillum schaedleri , RF39 , Acetatifactor , and Clostridia UCG-009 , while they had an increased abundance of Flavonifactor , Hungatella , Anaerofustis stercorihominis , and Clostridium innocuum group. Relative to sham rats, Fos and Gadd45g genes were upregulated, while Igf1 , Ccl2 , Hadc2 , Rtn4rl1 , Nfkb2 , Gpr84 , Pik3cg , and Abcc8 genes were downregulated in SNL rats. Compared to the SNL group, the GEG200 group and GEG600 group had increases/decreases in 16 (10/6) genes and 11 (1/10) genes, respectively. GEG downregulated Fos and Gadd45g genes and upregulated Hdac2 genes in the amygdala. In summary, GEG alleviates NP by modulating the gut microbiome and reversing a molecular neuroimmune signature.
- Published
- 2024
- Full Text
- View/download PDF
12. Gingerol-Enriched Ginger Supplementation Mitigates Neuropathic Pain via Mitigating Intestinal Permeability and Neuroinflammation: Gut-Brain Connection.
- Author
-
Shen CL, Wang R, Yakhnitsa V, Santos JM, Watson C, Kiritoshi T, Ji G, Hamood AN, and Neugebauer V
- Abstract
Objectives: Emerging evidence suggests an important role of the gut-brain axis in the development of neuropathic pain (NP). We investigated the effects of gingerol-enriched ginger (GEG) on pain behaviors, as well as mRNA expressions of inflammation via tight junction proteins in GI tissues (colon) and brain tissues (amygdala, both left and right) in animals with NP. Methods: Seventeen male rats were randomly divided into three groups: Sham, spinal nerve ligation (SNL, pain model), and SNL+0.375% GEG (wt/wt in diet) for 4 weeks. Mechanosensitivity was assessed by von Frey filament tests and hindpaw compression tests. Emotional responsiveness was measured from evoked audible and ultrasonic vocalizations. Ongoing spontaneous pain was measured in rodent grimace tests. Intestinal permeability was assessed by the lactulose/D-mannitol ratio in urine. The mRNA expression levels of neuroinflammation (NF-κB, TNF-α) in the colon and amygdala (right and left) were determined by qRT-PCR. Data was analyzed statistically. Results: Compared to the sham group, the SNL group had significantly greater mechanosensitivity (von Frey and compression tests), emotional responsiveness (audible and ultrasonic vocalizations to innocuous and noxious mechanical stimuli), and spontaneous pain (rodent grimace tests). GEG supplementation significantly reduced mechanosensitivity, emotional responses, and spontaneous pain measures in SNL rats. GEG supplementation also tended to decrease SNL-induced intestinal permeability markers. The SNL group had increased mRNA expression of NF-κB and TNF-α in the right amygdala and colon; GEG supplementation mitigated these changes in SNL-treated rats. Conclusion: This study suggests GEG supplementation palliated a variety of pain spectrum behaviors in a preclinical NP animal model. GEG also decreased SNL-induced intestinal permeability and neuroinflammation, which may explain the behavioral effects of GEG., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Shen, Wang, Yakhnitsa, Santos, Watson, Kiritoshi, Ji, Hamood and Neugebauer.)
- Published
- 2022
- Full Text
- View/download PDF
13. Kappa Opioid Receptor Blockade in the Amygdala Mitigates Pain Like-Behaviors by Inhibiting Corticotropin Releasing Factor Neurons in a Rat Model of Functional Pain.
- Author
-
Yakhnitsa V, Ji G, Hein M, Presto P, Griffin Z, Ponomareva O, Navratilova E, Porreca F, and Neugebauer V
- Abstract
Functional pain syndromes (FPS) occur in the absence of identifiable tissue injury or noxious events and include conditions such as migraine, fibromyalgia, and others. Stressors are very common triggers of pain attacks in various FPS conditions. It has been recently demonstrated that kappa opioid receptors (KOR) in the central nucleus of amygdala (CeA) contribute to FPS conditions, but underlying mechanisms remain unclear. The CeA is rich in KOR and encompasses major output pathways involving extra-amygdalar projections of corticotropin releasing factor (CRF) expressing neurons. Here we tested the hypothesis that KOR blockade in the CeA in a rat model of FPS reduces pain-like and nocifensive behaviors by restoring inhibition of CeA-CRF neurons. Intra-CeA administration of a KOR antagonist (nor-BNI) decreased mechanical hypersensitivity and affective and anxiety-like behaviors in a stress-induced FPS model. In systems electrophysiology experiments in anesthetized rats, intra-CeA application of nor-BNI reduced spontaneous firing and responsiveness of CeA neurons to peripheral stimulation. In brain slice whole-cell patch-clamp recordings, nor-BNI increased feedforward inhibitory transmission evoked by optogenetic and electrical stimulation of parabrachial afferents, but had no effect on monosynaptic excitatory transmission. Nor-BNI decreased frequency, but not amplitude, of spontaneous inhibitory synaptic currents, suggesting a presynaptic action. Blocking KOR receptors in stress-induced FPS conditions may therefore represent a novel therapeutic strategy., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Yakhnitsa, Ji, Hein, Presto, Griffin, Ponomareva, Navratilova, Porreca and Neugebauer.)
- Published
- 2022
- Full Text
- View/download PDF
14. Fear extinction learning ability predicts neuropathic pain behaviors and amygdala activity in male rats.
- Author
-
Ji G, Yakhnitsa V, Kiritoshi T, Presto P, and Neugebauer V
- Subjects
- Acoustic Stimulation, Action Potentials physiology, Amygdala pathology, Analysis of Variance, Animals, Conditioning, Classical physiology, Disease Models, Animal, Male, Mood Disorders etiology, Neuralgia psychology, Neurons physiology, Pain Measurement, Pain Threshold physiology, Physical Stimulation adverse effects, Rats, Rats, Sprague-Dawley, Extinction, Psychological physiology, Fear psychology, Learning Disabilities etiology, Learning Disabilities pathology, Neuralgia complications
- Abstract
Background The amygdala plays a key role in fear learning and extinction and has emerged as an important node of emotional-affective aspects of pain and pain modulation. Impaired fear extinction learning, which involves prefrontal cortical control of amygdala processing, has been linked to neuropsychiatric disorders. Here, we tested the hypothesis that fear extinction learning ability can predict the magnitude of neuropathic pain. Results We correlated fear extinction learning in naive adult male rats with sensory and affective behavioral outcome measures (mechanical thresholds, vocalizations, and anxiety- and depression-like behaviors) before and after the induction of the spinal nerve ligation model of neuropathic pain compared to sham controls. Auditory fear conditioning, extinction learning, and extinction retention tests were conducted after baseline testing. All rats showed increased freezing responses after fear conditioning. During extinction training, the majority (75%) of rats showed a decline in freezing level to 50% in 5 min (fear extinction+), whereas 25% of the rats maintained a high freezing level (>50%, fear extinction-). Fear extinction- rats showed decreased open-arm preference in the elevated plus maze, reflecting anxiety-like behavior, but there were no significant differences in sensory thresholds, vocalizations, or depression-like behavior (forced swim test) between fear extinction+ and fear extinction- types. In the neuropathic pain model (four weeks after spinal nerve ligation), fear extinction- rats showed a greater increase in vocalizations and anxiety-like behavior than fear extinction+ rats. Fear extinction- rats, but not fear extinction+ rats, also developed depression-like behavior. Extracellular single unit recordings of amygdala (central nucleus) neurons in behaviorally tested rats (anesthetized with isoflurane) found greater increases in background activity, bursting, and evoked activity in fear extinction- rats than fear extinction+ rats in the spinal nerve ligation model compared to sham controls. Conclusion The data may suggest that fear extinction learning ability predicts the magnitude of neuropathic pain-related affective rather than sensory behaviors, which correlates with differences in amygdala activity changes.
- Published
- 2018
- Full Text
- View/download PDF
15. Long-term climbing fibre activity induces transcription of microRNAs in cerebellar Purkinje cells.
- Author
-
Barmack NH, Qian Z, and Yakhnitsa V
- Subjects
- 14-3-3 Proteins genetics, Animals, Eye Movements physiology, Humans, MicroRNAs genetics, MicroRNAs metabolism, Photic Stimulation, Protein Kinase C metabolism, Rabbits, 14-3-3 Proteins metabolism, Afferent Pathways physiology, MicroRNAs physiology, Models, Neurological, Purkinje Cells physiology, Transcriptional Activation physiology
- Abstract
Synaptic activation of central neurons is often evoked by electrical stimulation leading to post-tetanic potentiation, long-term potentiation or long-term depression. Even a brief electrical tetanus can induce changes in as many as 100 proteins. Since climbing fibre activity is often associated with cerebellar behavioural plasticity, we used horizontal optokinetic stimulation (HOKS) to naturally increase synaptic input to floccular Purkinje cells in mice for hours, not minutes, and investigated how this activity influenced the transcription of microRNAs, small non-coding nucleotides that reduce transcripts of multiple, complementary mRNAs. A single microRNA can reduce the translation of as many as 30 proteins. HOKS evoked increases in 12 microRNA transcripts in floccular Purkinje cells. One of these microRNAs, miR335, increased 18-fold after 24 h of HOKS. After HOKS stopped, miR335 transcripts decayed with a time constant of approximately 2.5 h. HOKS evoked a 28-fold increase in pri-miR335 transcripts compared with an 18-fold increase in mature miR335 transcripts, confirming that climbing fibre-evoked increases in miR335 could be attributed to increases in transcription. We used three screens to identify potential mRNA targets for miR335 transcripts: (i) nucleotide complementarity, (ii) detection of increased mRNAs following microinjection of miR335 inhibitors into the cerebellum, and (iii) detection of decreased mRNAs following HOKS. Two genes, calbindin and 14-3-3-θ, passed these screens. Transfection of N2a cells with miR335 inhibitors or precursors inversely regulated 14-3-3-θ transcripts. Immunoprecipitation of 14-3-3-θ co-immunoprecipitated PKC-γ and GABAAγ2. Knockdown of either 14-3-3-θ or PKC-γ decreased the serine phosphorylation of GABAAγ2, suggesting that 14-3-3-θ and PKC-γ under the control of miR335 homeostatically regulate the phosphorylation and insertion of GABAAγ2 into the Purkinje cell post-synaptic membrane., (© 2014 The Author(s) Published by the Royal Society. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
16. Activity-dependent expression of acyl-coenzyme a-binding protein in retinal muller glial cells evoked by optokinetic stimulation.
- Author
-
Barmack NH, Bilderback TR, Liu H, Qian Z, and Yakhnitsa V
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Brain Chemistry, Carrier Proteins genetics, Gene Expression Profiling, Immunohistochemistry, In Situ Hybridization, Molecular Sequence Data, Photic Stimulation methods, Physical Stimulation methods, Protein Subunits metabolism, RNA, Messenger metabolism, Rabbits, Receptors, GABA-A metabolism, Retina metabolism, Two-Hybrid System Techniques, Carrier Proteins metabolism, Diazepam Binding Inhibitor metabolism, Neuroglia metabolism, Nystagmus, Optokinetic physiology, Retina physiology
- Abstract
Long-term horizontal optokinetic stimulation (HOKS) decreases the gain of the horizontal optokinetic reflex and evokes the second phase of optokinetic afternystagmus (OKAN-II). We investigated the possible molecular constituents of this adaptation. We used a differential display reverse transcriptase-PCR screen for mRNAs isolated from retinas of rabbits that received HOKS. In each rabbit, we compared mRNAs from the retina stimulated in the posterior-->anterior (preferred) direction with mRNAs from the retina stimulated in the anterior-->posterior (null) direction. Acyl-CoA-binding protein (ACBP) mRNA was one of four mRNAs selected by this screen, the proteins of which interact with GABA receptors. HOKS in the preferred direction increased ACBP mRNA transcription and ACBP protein expression. ACBP was localized to Muller glial cells by hybridization histochemistry and by immunohistochemistry. ACBP interacts with the alpha1-subunit of the GABA(A) receptor, as determined by a yeast two-hybrid technique. This interaction was confirmed by coimmunoprecipitation of ACBP and the alpha1-subunit of the GABA(A) receptor using an antibody to GABA(A)alpha1. The interaction was also confirmed by a "pull-down" assay in which histidine-tagged ACBP was used to pull down the GABA(A)alpha1. ACBP does not cross the blood-brain barrier. However, smaller truncated proteolytic fragments of ACBP do, increasing the excitability of central cortical neurons. Muller cells may secrete ACBP in the inner plexiform layer, thereby decreasing the sensitivity of GABA(A) receptors expressed on the surface of ganglion cell dendrites. Because retinal directional sensitivity is linked to GABAergic transmission, HOKS-induced expression of ACBP could provide a molecular basis for adaptation to HOKS and for the genesis of OKAN-II.
- Published
- 2004
- Full Text
- View/download PDF
17. Cerebellar climbing fibers modulate simple spikes in Purkinje cells.
- Author
-
Barmack NH and Yakhnitsa V
- Subjects
- Afferent Pathways, Animals, Cerebellum cytology, Ear, Inner surgery, Models, Neurological, Olivary Nucleus anatomy & histology, Olivary Nucleus cytology, Rabbits, Vestibular Nerve cytology, Vestibular Nerve physiology, Action Potentials, Cerebellum physiology, Nerve Fibers physiology, Purkinje Cells physiology
- Abstract
Purkinje cells have two action potentials: Climbing fiber responses (CFRs) and simple spikes (SSs). CFRs reflect the discharge of a single climbing fiber at multiple synaptic sites on the proximal dendrite of the Purkinje cell. SSs reflect the summed action of a subset of parallel fiber synapses on Purkinje cell dendritic spines. Because mossy fiber afferents terminate on granule cells, the ascending axons of which bifurcate, giving rise to parallel fibers, the modulation of SSs has been attributed to mossy fiber afferent signals. This inference has never been tested. Conversely, the low discharge frequency of CFRs has led many to conclude that they have a unique and intermittent role in cerebellar signal processing. We examine the relative potency of vestibularly modulated mossy fiber and climbing fiber signals in evoking CFRs and SSs in Purkinje cells of the uvula-nodulus in chloralose-urethane-anesthetized rabbits. Vestibular primary afferents were blocked by unilateral labyrinthectomy (UL). A UL destroys the vestibular primary afferent signal to the ipsilateral uvula-nodulus, while leaving intact the vestibular climbing fiber signal from the contralateral inferior olive. After UL, vestibular stimulation modulated CFRs and SSs in ipsilateral uvula-nodular Purkinje cells, demonstrating that the primary vestibular afferent mossy fiber input to the ipsilateral uvula-nodulus was not necessary for SS modulation. Unilateral microlesions of the caudal half of the beta-nucleus of the inferior olive reduced a modulated climbing fiber signal to the contralateral uvula-nodulus, causing loss of both vestibularly modulated CFRs and SSs in contralateral Purkinje cells. Vestibular climbing fibers not only evoke low-frequency CFRs, but also indirectly modulate higher-frequency SSs. This modulation must be attributed to cerebellar interneurons. Golgi cell inhibition of granule cells may provide the interneuronal mechanism for CFR-induced SS modulation.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.