Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 79: 626-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24412674

ABSTRACT

Cocaine's main pharmacological actions are the inhibition of the dopamine, serotonin, and norepinephrine transporters. Its main behavioral effects are reward and locomotor stimulation, potentially leading to addiction. Using knock-in mice with a cocaine-insensitive dopamine transporter (DAT-CI mice) we have shown previously that inhibition of the dopamine transporter (DAT) is necessary for both of these behaviors. In this study, we sought to determine brain regions in which DAT inhibition by cocaine stimulates locomotor activity and/or produces reward. We used adeno-associated viral vectors to re-introduce the cocaine-sensitive wild-type DAT in specific brain regions of DAT-CI mice, which otherwise only express a cocaine-insensitive DAT globally. Viral-mediated expression of wild-type DAT in the rostrolateral striatum restored cocaine-induced locomotor stimulation and sensitization in DAT-CI mice. In contrast, the expression of wild-type DAT in the dorsal striatum, or in the medial nucleus accumbens, did not restore cocaine-induced locomotor stimulation. These data help to determine cocaine's molecular actions and anatomical loci that cause hyperlocomotion. Interestingly, cocaine did not produce significant reward - as measured by conditioned place-preference - in any of the three cohorts of DAT-CI mice with the virus injections. Therefore, the locus or loci underlying cocaine-induced reward remain underdetermined. It is possible that multiple dopamine-related brain regions are involved in producing the robust rewarding effect of cocaine.


Subject(s)
Cocaine/pharmacology , Conditioning, Classical/drug effects , Corpus Striatum/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Psychomotor Agitation/physiopathology , Animals , Cocaine-Related Disorders/physiopathology , Conditioning, Classical/physiology , Corpus Striatum/physiopathology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Gene Knock-In Techniques , HEK293 Cells , Humans , Mice , Mice, Transgenic , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Reward , Space Perception/drug effects , Space Perception/physiology
2.
Proc Natl Acad Sci U S A ; 105(11): 4242-6, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18334641

ABSTRACT

Primary cilia are ubiquitous cellular appendages that provide important yet not well understood sensory and signaling functions. Ciliary dysfunction underlies numerous human genetic disorders. However, the precise defects in cilia function and the basis of disease pathophysiology remain unclear. Here, we report that the proteins disrupted in the human ciliary disorder Bardet-Biedl syndrome (BBS) are required for the localization of G protein-coupled receptors to primary cilia on central neurons. We demonstrate a lack of ciliary localization of somatostatin receptor type 3 (Sstr3) and melanin-concentrating hormone receptor 1 (Mchr1) in neurons from mice lacking the Bbs2 or Bbs4 gene. Because Mchr1 is involved in the regulation of feeding behavior and BBS is associated with hyperphagia-induced obesity, our results suggest that altered signaling caused by mislocalization of ciliary signaling proteins underlies the BBS phenotypes. Our results also provide a potential molecular mechanism to link cilia defects with obesity.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Cilia/metabolism , Microtubule-Associated Proteins/metabolism , Proteins/metabolism , Receptors, Somatostatin/metabolism , Animals , Bardet-Biedl Syndrome/genetics , Cells, Cultured , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Mutation/genetics , Proteins/genetics
3.
Am J Physiol Regul Integr Comp Physiol ; 294(5): R1704-15, 2008 May.
Article in English | MEDLINE | ID: mdl-18321955

ABSTRACT

The rostral hypothalamus, especially the preoptic-anterior hypothalamus (POAH), contains temperature-sensitive and -insensitive neurons that form synaptic networks to control thermoregulatory responses. Previous studies suggest that the cyclic nucleotide cGMP is an important mediator in this neuronal network, since hypothalamic microinjections of cGMP analogs produce hypothermia in several species. In the present study, immunohistochemisty showed that rostral hypothalamic neurons contain cGMP, guanylate cyclase (necessary for cGMP synthesis), and CNG A2 (an important cyclic nucleotide-gated channel). Extracellular electrophysiological activity was recorded from different types of neurons in rat hypothalamic tissue slices. Each recorded neuron was classified according to its thermosensitivity as well as its firing rate response to 2-100 microM 8-bromo-cGMP (a membrane-permeable cGMP analog). cGMP has specific effects on different neurons in the rostral hypothalamus. In the POAH, the cGMP analog decreased the spontaneous firing rate in 45% of temperature-sensitive and -insensitive neurons, an effect that is likely due to cGMP-enhanced hyperpolarizing K(+) currents. This decreased POAH activity could attenuate thermoregulatory responses and produce hypothermia during exposures to cool or neutral ambient temperatures. Although 8-bromo-cGMP did not affect the thermosensitivity of most POAH neurons, it did increase the warm sensitivity of neurons in other hypothalamic regions located dorsal, lateral, and posterior to the POAH. This increased thermosensitivity may be due to pacemaker currents that are facilitated by cyclic nucleotides. If some of these non-POAH thermosensitive neurons promote heat loss or inhibit heat production, then their increased thermosensitivity could contribute to cGMP-induced decreases in body temperature.


Subject(s)
Cyclic GMP/pharmacology , Hypothalamus/physiology , Neurons/physiology , Animals , Body Temperature/physiology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/physiology , Electrophysiology , Guanylate Cyclase/metabolism , Hypothalamus/drug effects , Hypothalamus, Anterior/cytology , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/physiology , Immunohistochemistry , Ion Channel Gating/physiology , Male , Neurons/drug effects , Preoptic Area/cytology , Preoptic Area/drug effects , Preoptic Area/physiology , Rats , Rats, Sprague-Dawley
4.
J Comp Neurol ; 505(5): 562-71, 2007 Dec 10.
Article in English | MEDLINE | ID: mdl-17924533

ABSTRACT

Solitary primary cilia project from nearly every cell type in the human body. These organelles are considered to have important sensory and signaling functions. Although primary cilia have been detected throughout the mammalian brain, their functions are unknown. The study of primary cilia in the brain is constrained by the scarcity of specific markers for these organelles. We previously demonstrated that type III adenylyl cyclase (ACIII) is a marker for primary cilia on neonatal hippocampal neurons in vivo and in vitro. We further showed that ACIII localizes to cilia on cultured glial cells. Here, we report that ACIII is a marker for primary cilia throughout many regions of the adult mouse brain. Furthermore, we report that ACIII localizes to primary cilia on choroid plexus cells and some astrocytes in the brain, which to our knowledge is the first report of a marker for visualizing cilia on glia in vivo. Overall, our data indicate that ACIII is a prominent marker of primary cilia in the brain and will provide an important tool to facilitate further investigations into the functions of these organelles.


Subject(s)
Adenylyl Cyclases/metabolism , Astrocytes/metabolism , Biomarkers/metabolism , Brain/metabolism , Cilia/enzymology , Mice, Inbred Strains/metabolism , Adenylyl Cyclases/immunology , Age Factors , Animals , Antibodies , Astrocytes/ultrastructure , Brain/cytology , Choroid Plexus/cytology , Choroid Plexus/metabolism , Fluorescent Antibody Technique , Isoenzymes/immunology , Isoenzymes/metabolism , Mice , Rabbits
5.
J Neurosci Res ; 85(9): 1996-2005, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17471557

ABSTRACT

Corticotropin releasing factor (CRF) and its cognate receptors, defined as Type 1 and Type 2 have been localized within the cerebellum. The Type 2 CRF receptor (CRF-R2) is known to have both a full length (CRF-R2alpha) and a truncated (CRF-R2alpha-tr) isoform. A recent study documented CRF-R2alpha primarily in Bergann glia and astrocytes, as well as in populations of Purkinje cells in the adult cerebellum. The goal of the present study is to determine if CRF-R2alpha is present in the postnatal cerebellum, and if so to describe its cellular distribution. RT-PCR data showed that CRF-R2alpha is expressed in the mouse cerebellum from birth through postnatal day 21. Between birth and P14, CRF-R2alpha-immunoreactivity was localized within the somata of Purkinje cells, and migrating GABAergic interneurons. GFAP-immunoreactive astrocytes, including Bergmann glia, also expressed CRF-R2alpha-immunoreactivity from P3-P14. There is a change, however, in CRF-R2alpha immunolabeling within neurons as the cerebellum matures. Compared to its expression in the adult cerebellum, Purkinje cells, and GABAergic interneurons showed more extensive CRF-R2alpha immunolabeling during early postnatal development. We postulate that CRF-R2alpha could be involved in developmental events related to the survival and differentiation of Purkinje cells and GABAergic neurons, whereas in the adult, this isoform of the CRF receptor family is likely involved in modulating Bergmann glia that have been shown to play a role in regulating the synaptic environment around Purkinje neurons.


Subject(s)
Cerebellar Cortex/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Cerebellar Cortex/growth & development , Corticotropin-Releasing Hormone/metabolism , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Immunohistochemistry , Interneurons/physiology , Isomerism , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Purkinje Cells/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , gamma-Aminobutyric Acid/physiology
6.
J Neurosci Res ; 85(5): 1095-100, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17304575

ABSTRACT

Primary cilia are cellular appendages that provide important sensory functions and defects in primary ciliary signaling have been implicated in the pathophysiology of human diseases and developmental abnormalities. Almost all human cell types possess a primary cilium. Neurons throughout the brain possess primary cilia on which certain receptors localize, suggesting that neurons possess cilia-mediated signaling. However, the functional significance of neuronal cilia is unknown. Although there is a great deal of interest in understanding the functions of neuronal cilia, their study is hampered by the lack of an in vitro model system. We report that the majority of hippocampal neurons cultured from postnatal mice possess primary cilia in vitro. Further, we describe cilia proteins that can be labeled to readily visualize neuronal primary cilia in culture. These findings are the first characterization of neuronal primary cilia in vitro and should greatly facilitate further investigations into the function of these organelles.


Subject(s)
Cilia/ultrastructure , Hippocampus/cytology , Neurons/cytology , Adenylyl Cyclases/analysis , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Biomarkers/analysis , Biomarkers/metabolism , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Shape/physiology , Cells, Cultured , Cilia/metabolism , Hippocampus/metabolism , Immunohistochemistry/methods , Isoenzymes/analysis , Isoenzymes/metabolism , Mice , Neurons/metabolism , Receptors, Somatostatin/analysis , Receptors, Somatostatin/metabolism
7.
J Neurosci Res ; 84(6): 1255-69, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16955482

ABSTRACT

Corticotropin releasing factor (CRF), localized in afferent inputs to the cerebellum, binds to two receptors defined as the Type 1 (CRF-R1) and the Type 2 (CRF-R2alpha). CRF-R1 has been localized to the cerebellum, as has a truncated isoform of CRF-R2alpha. Evidence for the presence of the full length isoform of CRF-R2alpha in the cerebellum is conflicting. We used RT-PCR, immunohistochemical, and physiologic techniques to resolve this conflict. RT-PCR data show low levels of CRF-R2alpha in the vermis and hemisphere of the cerebellum. These observations were confirmed by the Gene Expression Nervous System Atlas (GENSAT) database. A CRF-R2alpha antibody was used to determine the cellular distribution of the receptor in the cerebellum. The vast majority of the receptors are localized to Bergmann glial cells located throughout the cerebellum, as well as astrocytes in the granule cell layer. Neuronal labeling is present in sub-populations of Purkinje cells, Golgi cells, basket cells, and cerebellar nuclear neurons. Physiologic data show that urocortin II, which binds selectively to CRF-R2alpha, increases the firing rate of both Purkinje cells and nuclear neurons; this response can be blocked by the CRF-R2alpha-specific antagonist, antisauvagine-30. The present results confirm that CRF-R2alpha is present in the cerebellum and functions in circuits that modulate the firing rate of Purkinje cells and cerebellar nuclear neurons. A comparative analysis showed that the patterns of distribution of CRF-R1, CRF-R2alpha and CRF-R2alpha-tr are distinct. These data indicate that the CRF family of peptides modulates cerebellar output by binding to multiple CRF receptors.


Subject(s)
Cerebellum/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Antibody Specificity , Blotting, Western , Cerebellum/cytology , Cytoplasmic Granules/physiology , Fluorescent Antibody Technique, Indirect , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Mice , Neurons/physiology , Purkinje Cells/physiology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical
8.
Am J Physiol Regul Integr Comp Physiol ; 291(3): R518-29, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16690776

ABSTRACT

Thermoregulatory responses are partially controlled by the preoptic area and anterior hypothalamus (PO/AH), which contains a mixed population of temperature-sensitive and insensitive neurons. Immunohistochemical procedures identified the extent of various ionic channels in rat PO/AH neurons. These included pacemaker current channels [i.e., hyperpolarization-activated cyclic nucleotide-gated channels (HCN)], background potassium leak channels (TASK-1 and TRAAK), and transient receptor potential channel (TRP) TRPV4. PO/AH neurons showed dense TASK-1 and HCN-2 immunoreactivity and moderate TRAAK and HCN-4 immunoreactivity. In contrast, the neuronal cell bodies did not label for TRPV4, but instead, punctate labeling was observed in traversing axons or their terminal endings. On the basis of these results and previous electrophysiological studies, Hodgkin-Huxley-like models were constructed. These models suggest that most PO/AH neurons have the same types of ionic channels, but different levels of channel expression can explain the inherent properties of the various types of temperature-sensitive and insensitive neurons.


Subject(s)
Body Temperature/physiology , Electric Conductivity , Hypothalamus/cytology , Ion Channels/metabolism , Neurons/metabolism , Action Potentials , Animals , Gene Expression Regulation , Ion Channels/genetics , Male , Models, Biological , Rats , Rats, Sprague-Dawley
9.
Hum Mol Genet ; 12(19): 2519-32, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12915461

ABSTRACT

The glial glutamate transporter EAAT2 is primarily responsible for clearance of glutamate from the synaptic cleft and loss of EAAT2 has been previously reported in amyotrophic lateral sclerosis (ALS) and Alzheimer's disease. The loss of functional EAAT2 could lead to the accumulation of extracellular glutamate, resulting in cell death known as excitotoxicity. However, it is still unknown whether it is a primary cause in the cascade leading to neuron degeneration or a secondary event to cell death. The goals of this study were to generate transgenic mice overexpressing EAAT2 and then to cross these mice with the ALS-associated mutant SOD1(G93A) mice to investigate whether supplementation of the loss of EAAT2 would delay or rescue the disease progression. We show that the amount of EAAT2 protein and the associated Na+-dependent glutamate uptake was increased about 2-fold in our EAAT2 transgenic mice. The transgenic EAAT2 protein was properly localized to the cell surface on the plasma membrane. Increased EAAT2 expression protects neurons from L-glutamate induced cytotoxicity and cell death in vitro. Furthermore, our EAAT2/G93A double transgenic mice showed a statistically significant (14 days) delay in grip strength decline but not in the onset of paralysis, body weight decline or life span when compared with G93A littermates. Moreover, a delay in the loss of motor neurons and their axonal morphologies as well as other events including caspase-3 activation and SOD1 aggregation were also observed. These results suggest that the loss of EAAT2 may contribute to, but does not cause, motor neuron degeneration in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cell Death/genetics , Excitatory Amino Acid Transporter 2/metabolism , Gene Expression , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Caspases/drug effects , Caspases/metabolism , Cell Membrane/metabolism , Crosses, Genetic , Disease Models, Animal , Enzyme Activation/drug effects , Excitatory Amino Acid Transporter 2/genetics , Glial Fibrillary Acidic Protein/genetics , Glutamic Acid/metabolism , Glutamic Acid/pharmacokinetics , Mice , Mice, Transgenic , Motor Neurons/enzymology , Motor Neurons/pathology , Promoter Regions, Genetic , Spinal Cord/metabolism , Spinal Cord/pathology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Time Factors , Transgenes
10.
J Neurocytol ; 32(3): 305-16, 2003 Mar.
Article in English | MEDLINE | ID: mdl-14724392

ABSTRACT

Corticotropin releasing factor (CRF) is present in the adult, as well as in the embryonic and postnatal rodent cerebellum. Further, the distribution of the type 1 CRF receptor has been described in adult and postnatal animals. The focus of the present study is to determine the distribution and cellular relationships of the type 1 CRF receptor (CRF-R1) during embryonic development of the cerebellum. Between embryonic day (E)11 and E12, CRF-R1 immunoreactive puncta are uniformly distributed in the ventricular zone, the site of origin of Purkinje cells, nuclear neurons, and GABAergic interneurons, as well as the germinal trigone, the birthplace of the precursors of granule cells. Between E13 and 18, the distribution of immunolabeled puncta decreases in both the ventricular zone and the germinal trigone and increases in the intermediate zone, as well as in the dorsal aspect of the cerebellar plate. Between E14 and 18, antibodies that label specific populations of cerebellar neurons were combined with the antibody for the receptor to determine the cellular elements that expressed CRF-R1. At E14, CRF-R1 immunoreactivity is co-localized in neurons immunolabeled with PAX-2, an antibody that is specific for GABAergic interneurons. These neurons continue to express CRF-R1 as they migrate dorsally toward the cerebellar surface. Between E16 and 18, Purkinje cells, immunolabeled with calbindin, near the dorsal surface of the cerebellum express CRF-R1 in their cell bodies and apical processes. CRF has been shown to have a depolarizing effect on adult and postnatal Purkinje cells. Further, CRF has been shown to contribute to excitability of hippocampal neurons during embryonic development by binding to CRF-R1; depolarization induced excitability appears to be critical for cell survival. The location of the type one CRF receptor and the presence of its primary ligand, CRF, in the germinal zones of the cerebellum and in migrating neurons suggest that this receptor/ligand interaction could be important in the regulation of neuronal survival through cellular mechanisms that lead to depolarization of embryonic cerebellar neurons.


Subject(s)
Cell Differentiation/physiology , Cerebellum/embryology , Cerebellum/metabolism , Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Antigens, Differentiation/metabolism , Cell Movement/physiology , Cerebellum/cytology , DNA-Binding Proteins/metabolism , Female , Fetus , Gestational Age , Immunohistochemistry , Interneurons/cytology , Interneurons/metabolism , Mice , PAX2 Transcription Factor , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Purkinje Cells/cytology , Purkinje Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Exp Neurol ; 178(2): 165-74, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12504876

ABSTRACT

Corticotropin-releasing factor (CRF), present in climbing and mossy fiber afferents to the adult mouse cerebellum, acts as a neuromodulator to enhance the spontaneous and amino-acid-induced firing rate of Purkinje cells. CRF also is present during development of the mouse cerebellum, at ages that precede synaptogenesis, which suggests that it may have a different function during development compared to its modulatory role in the adult. The intent of this study was to determine when CRF begins to affect the firing rate of Purkinje cells as well as the time course over which this effect matures. The earliest effect of CRF was elicited at postnatal day (P) 9 at which time a weak enhancement in the amplitude of the firing rate was recorded. However, the amplitude, time to peak, sustainability, and duration of the response were significantly different from that recorded in the older animals or adults. The excitatory effect of CRF became stronger and the duration of the response increased progressively from P9 until it was adult-like by P20. Purkinje cells in the posterior lobe vermis developed a mature response before those in the anterior lobe or hemispheres. Data from previous studies have shown that CRF and its type 1 receptor are present in the cerebellum before birth and that both undergo major reorganization around P10. Taken together, these immunohistochemical observations and the present physiologic data indicate that CRF does not modulate the activity of Purkinje cells until the peptide begins to assume an adult-like distribution in cerebellar afferents.


Subject(s)
Action Potentials/drug effects , Cerebellum/physiology , Corticotropin-Releasing Hormone/pharmacology , Purkinje Cells/drug effects , Action Potentials/physiology , Age Factors , Animals , Animals, Newborn , Cerebellum/cytology , Cerebellum/drug effects , Cerebellum/growth & development , Corticotropin-Releasing Hormone/physiology , Mice , Mice, Inbred C57BL , Purkinje Cells/physiology
12.
Exp Neurol ; 178(2): 175-85, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12504877

ABSTRACT

The distribution of corticotropin-releasing factor (CRF), the development of CRF-binding sites, and the age at which application of CRF elicits a physiological response have been described previously in the postnatal mouse cerebellum. The intent of the present study was to determine the cellular and subcellular distribution of the CRF type 1 receptor (CRF-R1) in the vermis of the postnatal mouse cerebellum and to correlate these data with those presented in previous studies. On P0, CRF-R1 is present in the apical processes of migrating Purkinje cells. Between P0 and P8, CRF-R1 immunostaining is confined to a supranuclear position in Purkinje cell bodies. Between P9 and P14, the receptor immunolabeling circumscribes Purkinje cell nuclei and extends into their primary dendrites. An adult-like distribution is achieved between P16 and P21. Between P0 and P14, the CRF-R1 antibody also labels processes of migrating GABAergic interneurons that are directed toward the pial surface. By P12, labeling begins to circumscribe the nucleus of GABAergic cells in the internal granule cell layer. Finally, astrocytic processes in the white matter, as well as radial glial processes, show focal labeling with the CRF-R1 antibody beginning at P3 and throughout postnatal development. A previous study demonstrated that CRF does not elicit a physiological response in Purkinje cells until P9. This observation, together with the data presented in this study, suggests that the binding of CRF to the type 1 receptor may be involved in regulating the development of cerebellar neurons and glia immediately after birth, before CRF assumes its function as a neuromodulator later in postnatal development and in the adult.


Subject(s)
Cerebellum/chemistry , Cerebellum/cytology , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Animals, Newborn , Astrocytes/chemistry , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/physiology , Cerebellum/growth & development , Cerebellum/metabolism , Humans , Mice , Mice, Inbred C57BL , Purkinje Cells/chemistry , Purkinje Cells/cytology , Purkinje Cells/metabolism , Purkinje Cells/physiology , Receptors, Corticotropin-Releasing Hormone/analysis
13.
J Chem Neuroanat ; 23(3): 157-70, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11861123

ABSTRACT

The intent of the present study was to use chemical or electrical stimulation of cerebellar afferents to determine how different stimulation paradigms affect the pattern of activation of different populations of neurons in the cerebellar cortex. Specifically, we analyzed immediate changes in neuronal activity, identified neurons affected by different stimulation paradigms, and determined the time course over which neuronal activity is altered. In the present study, we used either systemic (harmaline) or electrical stimulation of the inferior cerebellar peduncle (10 and 40 Hz) to alter the firing rate of climbing and mossy fiber afferents to the rat cerebellum and an antibody made against the proto-oncogene, c-fos, as a marker to identify activated neurons and glia. In control animals, only a few scattered granule cells express nuclear Fos-like immunoreactivity. Although no other cells show Fos-like immunoreactivity in their nuclei, Purkinje cells express Fos-like immunoreactivity within their somatic and dendritic cytoplasm in control animals. Within 15 min of chemical or electrical stimulation, numerous granule and glial cells express Fos-like immunoreactivity in their nuclei. Cells in the molecular layer express Fos-like immunoreactivity following harmaline stimulation in a time and lobule specific manner; they do not appear to be activated in the electrical stimulation paradigm. Following harmaline injections, there is an initial loss of Fos-like immunoreactivity in the cytoplasm of Purkinje cells; 90 min later, nuclear staining is observed in a few scattered Purkinje cells. Following electrical stimulation, the cytoplasmic staining in Purkinje cells is enhanced; it is never present in the nucleus. Data derived from this study reveal cell-specific temporal and spatial patterns of c-Fos activation that is unique to each paradigm. Further, it reveals the presence of an activity dependent protein in the cytoplasm of Purkinje cell somata and dendrites.


Subject(s)
Cerebellum/metabolism , Neuroglia/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Blotting, Western , Cerebellar Nuclei/physiology , Cerebellum/cytology , Electric Stimulation , Fluorescent Antibody Technique, Indirect , Harmaline/pharmacology , Immunoenzyme Techniques , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Tissue Fixation
SELECTION OF CITATIONS
SEARCH DETAIL
...