Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 32(5): 896-906, 2012 May.
Article in English | MEDLINE | ID: mdl-22293985

ABSTRACT

Activation of the basal forebrain (BF), the primary source of acetylcholine (ACh) in the cortex, broadly increases cortical cerebral blood flow (CBF), a response downstream to ACh release. Although endothelial nitric oxide and cholinoceptive GABA (γ-aminobutyric acid) interneurons have been implicated, little is known about the role of pyramidal cells in this response and their possible interaction with astrocytes. Using c-Fos immunohistochemistry as a marker of neuronal activation and laser-Doppler flowmetry, we measured changes in CBF evoked by BF stimulation following pharmacological blockade of c-Fos-identified excitatory pathways, astroglial metabolism, or vasoactive mediators. Pyramidal cells including those that express cyclooxygenase-2 (COX-2) displayed c-Fos upregulation. Glutamate acting via NMDA, AMPA, and mGlu receptors was involved in the evoked CBF response, NMDA receptors having the highest contribution (~33%). In contrast, nonselective and selective COX-2 inhibition did not affect the evoked CBF response (+0.4% to 6.9%, ns). The metabolic gliotoxins fluorocitrate and fluoroacetate, the cytochrome P450 epoxygenase inhibitor MS-PPOH and the selective epoxyeicosatrienoic acids (EETs) antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE) all blocked the evoked CBF response by ~50%. Together, the data demonstrate that the hyperemic response to BF stimulation is largely mediated by glutamate released from activated pyramidal cells and by vasoactive EETs, likely originating from activated astrocytes.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Evoked Potentials/physiology , Pyramidal Cells/metabolism , 8,11,14-Eicosatrienoic Acid/antagonists & inhibitors , 8,11,14-Eicosatrienoic Acid/metabolism , Acetylcholine/metabolism , Amides/pharmacology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blood Flow Velocity/drug effects , Blood Flow Velocity/physiology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Citrates/toxicity , Cyclooxygenase 2/metabolism , Evoked Potentials/drug effects , Fluoroacetates/toxicity , Gene Expression Regulation/drug effects , Male , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyramidal Cells/cytology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , gamma-Aminobutyric Acid/metabolism
2.
J Neurosci ; 31(27): 9836-47, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734275

ABSTRACT

The whisker-to-barrel cortex is widely used to study neurovascular coupling, but the cellular basis that underlies the perfusion changes is still largely unknown. Here, we identified neurons recruited by whisker stimulation in the rat somatosensory cortex using double immunohistochemistry for c-Fos and markers of glutamatergic and GABAergic neurons, and investigated in vivo their contribution along with that of astrocytes in the evoked perfusion response. Whisker stimulation elicited cerebral blood flow (CBF) increases concomitantly with c-Fos upregulation in pyramidal cells that coexpressed cyclooxygenase-2 (COX-2) and GABA interneurons that coexpressed vasoactive intestinal polypeptide and/or choline acetyltransferase, but not somatostatin or parvalbumin. The evoked CBF response was decreased by blockade of NMDA (MK-801, -37%), group I metabotropic glutamate (MPEP+LY367385, -40%), and GABA-A (picrotoxin, -31%) receptors, but not by GABA-B, VIP, or muscarinic receptor antagonism. Picrotoxin decreased stimulus-induced somatosensory evoked potentials and CBF responses. Combined blockade of GABA-A and NMDA receptors yielded an additive decreasing effect (-61%) of the evoked CBF compared with each antagonist alone, demonstrating cooperation of both excitatory and inhibitory systems in the hyperemic response. Blockade of prostanoid synthesis by inhibiting COX-2 (indomethacin, NS-398), expressed by ∼40% of pyramidal cells but not by astrocytes, impaired the CBF response (-50%). The hyperemic response was also reduced (-40%) after inhibition of astroglial oxidative metabolism or epoxyeicosatrienoic acids synthesis. These results demonstrate that changes in pyramidal cell activity, sculpted by specific types of inhibitory GABA interneurons, drive the CBF response to whisker stimulation and, further, that metabolically active astrocytes are also required.


Subject(s)
Cerebrovascular Circulation/physiology , Neurogenesis/physiology , Pyramidal Cells/physiology , Somatosensory Cortex/cytology , Vibrissae/innervation , Afferent Pathways/physiology , Analysis of Variance , Animals , Cerebrovascular Circulation/drug effects , Cholinergic Antagonists/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Drug Interactions , Electroencephalography , Enzyme Inhibitors , Evoked Potentials, Somatosensory/drug effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Functional Laterality , GABA Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Laser-Doppler Flowmetry/methods , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurogenesis/drug effects , Organophosphorus Compounds/pharmacology , Patch-Clamp Techniques/methods , Physical Stimulation/methods , Picrotoxin/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Scopolamine/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
3.
Brain Res ; 1407: 62-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21764040

ABSTRACT

Nitric oxide (NO), a potent vasodilator and nontraditional neurotransmitter, is an important mediator of the changes in cerebral blood flow (CBF) associated with increased neuronal activity (neurovascular coupling). In the present work, we investigated the role of NO and of its newly recognized precursor, nitrite, in neurovascular coupling using a well-established rat model of somatosensory stimulation. Biological synthesis of NO of neuronal origin was inhibited pharmacologically. Following the initial uncoupling of neuronal and hemodynamic responses to somatosensory stimulation, the NO donor sodium nitroprusside, added within the range of physiological concentrations, significantly increased, but did not fully restore the functional CBF response. In contrast, nitrite at its physiological concentration fully recovered neurovascular coupling to its original magnitude. The magnitude of the effect is, however, dose-dependent. Sub-physiological concentrations of nitrite were not enough to entirely restore neurovascular coupling and supra-physiological concentrations acted more as a local vasodilator that changed resting CBF and interfered with the functional CBF response. These results suggest that nitrite can be efficiently converted into NO and utilized to support normal cerebrovascular physiology.


Subject(s)
Blood Vessels/innervation , Blood Vessels/physiology , Neurons/physiology , Nitric Oxide/physiology , Nitrites/pharmacology , Anesthesia , Anesthetics, Intravenous , Animals , Blood Gas Analysis , Blood Pressure/physiology , Body Temperature , Cerebrovascular Circulation/physiology , Chloralose , Data Interpretation, Statistical , Electroencephalography/drug effects , Evoked Potentials, Somatosensory/drug effects , Laser-Doppler Flowmetry , Male , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitroprusside/pharmacology , Rats , Rats, Sprague-Dawley
4.
Ann N Y Acad Sci ; 1225 Suppl 1: E171-81, 2011 May.
Article in English | MEDLINE | ID: mdl-21599695

ABSTRACT

The pattern of myelination over the cerebral cortex, termed myeloarchitecture, is an established and often-used feature to visualize cortical organization with histology in a variety of primate species. In this paper, we use in vivo magnetic resonance imaging (MRI) and advanced image processing using surface rendering to visualize and characterize myeloarchitecture in a small nonhuman primate, the common marmoset (Callithrix jacchus). Through images made in four female adult marmosets, we produce a representative 3D map of marmoset myeloarchitecture and flatten and annotate this map to show the location and extent of a variety of major areas of the cortex, including the primary visual, auditory, and somatosensory areas. By treating our MRI data as a surface, we can measure the surface area of cortical areas, and we present these measurements here to summarize cortical organization in the marmoset.


Subject(s)
Brain/anatomy & histology , Callithrix/anatomy & histology , Meninges/anatomy & histology , Aging , Animals , Auditory Cortex/anatomy & histology , Auditory Cortex/growth & development , Brain/cytology , Brain/growth & development , Brain Mapping/methods , Callithrix/growth & development , Female , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Meninges/cytology , Meninges/growth & development , Models, Anatomic , Myelin Sheath , Organ Size , Somatosensory Cortex/anatomy & histology , Somatosensory Cortex/growth & development , Surface Properties , Visual Cortex/anatomy & histology , Visual Cortex/growth & development
5.
J Neurosci Methods ; 185(1): 15-22, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19737577

ABSTRACT

Myeloarchitecture, the pattern of myelin density across the cerebral cortex, has long been visualized in histological sections to identify distinct anatomical areas of the cortex. In humans, two-dimensional (2D) magnetic resonance imaging (MRI) has been used to visualize myeloarchitecture in select areas of the cortex, such as the stripe of Gennari in the primary visual cortex and Heschl's gyrus in the primary auditory cortex. Here, we investigated the use of MRI contrast based on longitudinal relaxation time (T(1)) to visualize myeloarchitecture in vivo over the entire cortex of the common marmoset (Callithrix jacchus), a small non-human primate that is becoming increasingly important in neuroscience and neurobiology research. Using quantitative T(1) mapping, we found that T(1) at 7T in a cortical region with a high myelin content was 15% shorter than T(1) in a region with a low myelin content. To maximize this T(1) contrast for imaging cortical myelination patterns, we optimized a magnetization-prepared rapidly acquired gradient echo (MP-RAGE) sequence. In whole-brain, 3D T(1)-weighted images made in vivo with the sequence, we identified six major cortical areas with high myelination and confirmed the results with histological sections stained for myelin. We also identified several subtle features of myeloarchitecture, showing the sensitivity of our technique. The ability to image myeloarchitecture over the entire cortex may prove useful in studies of longitudinal changes of the topography of the cortex associated with development and neuronal plasticity, as well as for guiding and confirming the location of functional measurements.


Subject(s)
Callithrix/anatomy & histology , Cerebral Cortex/anatomy & histology , Magnetic Resonance Imaging/methods , Nerve Fibers, Myelinated/ultrastructure , Neural Pathways/anatomy & histology , Anatomy, Comparative/methods , Animals , Auditory Cortex/anatomy & histology , Auditory Cortex/physiology , Brain Mapping/methods , Callithrix/physiology , Cerebral Cortex/growth & development , Female , Image Processing, Computer-Assisted/methods , Male , Nerve Fibers, Myelinated/physiology , Neural Pathways/physiology , Neuronal Plasticity/physiology , Species Specificity , Time Factors , Visual Cortex/anatomy & histology , Visual Cortex/physiology
6.
J Neurosci Methods ; 181(2): 212-26, 2009 Jul 30.
Article in English | MEDLINE | ID: mdl-19454294

ABSTRACT

Glial cell Ca2+ signals play a key role in glial-neuronal and glial-glial network communication. Numerous studies have thus far utilized cell-permeant and injected Ca2+ indicator dyes to investigate glial Ca2+ signals in vitro and in situ. Genetically encoded fluorescent Ca2+ indicators have emerged as novel probes for investigating cellular Ca2+ signals. We have expressed one such indicator protein, the YC 3.60 cameleon, under the control of the S100beta promoter and directed its expression predominantly in astrocytes and Schwann cells. Expression of YC 3.60 extended into the entire cellular cytoplasmic compartment and the fine terminal processes of protoplasmic astrocytes and Schwann cell Cajal bands. In the brain, all the cells known to express S100beta in the adult or during development, expressed YC 3.60. While expression was most extensive in astrocytes, other glial cell types that express S100beta, such as NG2 and CNP-positive oligodendrocyte progenitor cells (OP cells), microglia, and some of the large motor neurons in the brain stem, also contained YC 3.60 fluorescence. Using a variety of known in situ and in vivo assays, we found that stimuli known to elicit Ca2+ signals in astrocytes caused substantial and rapid Ca2+ signals in the YC 3.60-expressing astrocytes. In addition, forepaw stimulation while imaging astrocytes through a cranial window in the somatosensory cortex in live mice, revealed robust evoked and spontaneous Ca2+ signals. These results, for the first time, show that genetically encoded reporter is capable of recording activity-dependent Ca2+ signals in the astrocyte processes, and networks.


Subject(s)
Astrocytes/metabolism , Calcium Signaling/physiology , Schwann Cells/metabolism , Somatosensory Cortex/metabolism , Animals , Astrocytes/drug effects , Calcium Signaling/drug effects , Cells, Cultured , Cerebellum/metabolism , Evoked Potentials, Somatosensory/physiology , Fluorescent Dyes , Glutamic Acid/pharmacology , Hippocampus/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Schwann Cells/drug effects , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects
7.
J Cereb Blood Flow Metab ; 29(5): 976-86, 2009 May.
Article in English | MEDLINE | ID: mdl-19337274

ABSTRACT

Functional neuroimaging signals are generated, in part, by increases in cerebral blood flow (CBF) evoked by mediators, such as nitric oxide and arachidonic acid derivatives that are released in response to increased neurotransmission. However, it is unknown whether the vascular and metabolic responses within a given brain area differ when local neuronal activity is evoked by an activity in the distinct neuronal networks. In this study we assessed, for the first time, the differences in neuronal responses and changes in CBF and oxygen consumption that are evoked after the activation of two different inputs to a single cortical area. We show that, for a given level of glutamatergic synaptic activity, corticocortical and thalamocortical inputs evoked activity in pyramidal cells and different classes of interneurons, and produced different changes in oxygen consumption and CBF. Furthermore, increases in stimulation intensities either turned off or activated additional classes of inhibitory interneurons immunoreactive for different vasoactive molecules, which may contribute to increases in CBF. Our data imply that for a given cortical area, the amplitude of vascular signals will depend critically on the type of input, and that a positive blood oxygen level-dependent (BOLD) signal may be a consequence of the activation of both pyramidal cells and inhibitory interneurons.


Subject(s)
Cerebrovascular Circulation/physiology , Neurons/metabolism , Oxygen Consumption/physiology , Somatosensory Cortex/blood supply , Somatosensory Cortex/metabolism , Animals , Electric Stimulation , Evoked Potentials, Somatosensory/physiology , Immunohistochemistry , Interneurons/metabolism , Interneurons/physiology , Laser-Doppler Flowmetry , Male , Neurons/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Rats , Rats, Wistar , Somatosensory Cortex/physiology , Synaptic Potentials/physiology , gamma-Aminobutyric Acid/metabolism
8.
NMR Biomed ; 22(7): 730-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19322808

ABSTRACT

MRI at 7 Tesla has been used to investigate the accumulation of manganese in the occipital cortex of common marmoset monkeys (Callithrix jacchus) after administering four fractionated injections of 30 mg/kg MnCl(2) . 4H(2)O in the tail vein. We found a statistically significant decrease in T(1) in the primary (V1) and secondary (V2) areas of the visual cortex caused by an accumulation of manganese. The larger T(1) shortening in V1 (DeltaT(1) = 640 ms) relative to V2 (DeltaT(1) = 490 ms) allowed us to robustly detect the V1/V2 border in vivo using heavily T(1)-weighted MRI. Furthermore, the dorso-medial (DM) and middle-temporal (MT) areas of the visual pathway could be identified by their T(1)-weighted enhancement. We showed by comparison to histological sections stained for cytochrome oxidase (CO) activity that the extent of V1 is accurately identified throughout the visual cortex by manganese-enhanced MRI (MEMRI). This provides a means of visualizing functional cortical regions in vivo and could be used in longitudinal studies of phenomena such as cortical plasticity, and for non-destructive localization of cortical regions to guide in the implementation of functional techniques.


Subject(s)
Callithrix/anatomy & histology , Magnetic Resonance Imaging/methods , Manganese/administration & dosage , Manganese/pharmacology , Visual Cortex/anatomy & histology , Visual Cortex/drug effects , Animals , Female , Humans , Male , Visual Cortex/metabolism
9.
J Cereb Blood Flow Metab ; 28(2): 221-31, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17895909

ABSTRACT

Neurovascular coupling, or the tight coupling between neuronal activity and regional cerebral blood flow (CBF), seems largely driven by the local processing of incoming afferent signals within the activated area. To test if cortical gamma-aminobutyric acid (GABA) interneurons-the local integrators of cortical activity-are involved in this coupling, we stimulated the basalocortical pathway in vivo, monitored cortical CBF, and identified the activated interneurons (c-Fos-immunopositive) and the neuromediators involved in this response. Basal forebrain (BF) stimulation induced ipsilateral increases in CBF and selective activation of layers II to VI somatostatin- and/or neuropeptide Y-containing, as well as layer I GABA interneurons. Nitric oxide synthase interneurons displayed weak bilateral activation, whereas vasoactive intestinal polypeptide- or acetylcholine (ACh)-containing GABA interneurons were not activated. Selective cholinergic deafferentation indicated that ACh released from stimulated BF afferents triggered the CBF response, but the latter was mediated, in part, by the local release of GABA from cholinoceptive cortical interneurons, and through GABA-A receptor-mediated transmission. These data show that activation of specific subsets of GABA interneurons and their GABA-A-mediated effects on neuronal, vascular, and/or astroglial targets are necessary for the full expression of the hemodynamic response to BF stimulation. Further, these findings highlight the importance of understanding the cellular networks and circuitry that underlie hemodynamic signals, as only specific subsets of neurons may be activated by a given stimulus, depending on the afferent inputs they receive and integrate.


Subject(s)
Blood Vessels/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/physiology , Cerebrovascular Circulation/physiology , Interneurons/physiology , Prosencephalon/blood supply , Prosencephalon/physiology , gamma-Aminobutyric Acid/physiology , Acetylcholine/physiology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Blood Vessels/innervation , Capillaries/metabolism , Cerebral Cortex/drug effects , Cerebrovascular Circulation/drug effects , Denervation , Electric Stimulation , Electrophysiology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Immunohistochemistry , Interneurons/drug effects , Male , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type III/physiology , Parasympathetic Nervous System/physiology , Prosencephalon/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/physiology , Somatostatin/metabolism
10.
J Neurosci ; 25(48): 11165-74, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16319316

ABSTRACT

The roles of oxidative stress and structural alterations in the cerebrovascular dysfunctions associated with Alzheimer's disease (AD) were investigated in transgenic mice overexpressing amyloid precusor protein (APP+) or transforming growth factor-beta1 (TGF+). Age-related impairments and their in vitro reversibility were evaluated, and underlying pathogenic mechanisms were assessed and compared with those seen in AD brains. Vasoconstrictions to 5-HT and endothelin-1 were preserved, except in the oldest (18-21 months of age) TGF+ mice. Despite unaltered relaxations to sodium nitroprusside, acetylcholine (ACh) and calcitonin gene-related peptide-mediated dilatations were impaired, and there was an age-related deficit in the basal availability of nitric oxide (NO) that progressed more gradually in TGF+ mice. The expression and progression of these deficits were unrelated to the onset or extent of thioflavin-S-positive vessels. Manganese superoxide dismutase (SOD2) was upregulated in pial vessels and around brain microvessels of APP+ mice, pointing to a role of superoxide in the dysfunctions elicited by amyloidosis. In contrast, vascular wall remodeling associated with decreased levels of endothelial NO synthase and cyclooxygenase-2 and increased contents of vascular endothelial growth factor and collagen-I and -IV characterized TGF+ mice. Exogenous SOD or catalase normalized ACh dilatations and NO availability in vessels from aged APP+ mice but had no effect in those of TGF+ mice. Increased perivascular oxidative stress was not evidenced in AD brains, but vascular wall alterations compared well with those seen in TGF+ mice. We conclude that brain vessel remodeling and associated alterations in levels of vasoactive signaling molecules are key contributors to AD cerebrovascular dysfunctions.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/blood supply , Oxidative Stress , Transforming Growth Factors/metabolism , Aged , Alzheimer Disease/metabolism , Amyloid Neuropathies/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Blood Vessels/metabolism , Blood Vessels/physiopathology , Brain Diseases/metabolism , Catalase/pharmacology , Cerebral Cortex/blood supply , Female , Humans , Male , Mice , Mice, Transgenic , Microcirculation , Superoxide Dismutase/pharmacology , Transforming Growth Factors/genetics , Vasomotor System/drug effects , Vasomotor System/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...