scispace - formally typeset
Search or ask a question

Showing papers in "The Journal of Comparative Neurology in 2006"


Journal ArticleDOI
TL;DR: Ghrelin is a hormone that stimulates growth hormone secretion and signals energy insufficiency via interaction with its receptor, the growth hormone secretagogue receptor (GHSR), and its distribution in the CNS, as assessed by in situ hybridization histochemistry (ISHH), has been described previously in a few mammalian species.
Abstract: Ghrelin is a hormone that stimulates growth hormone secretion and signals energy insufficiency via interaction with its receptor, the growth hormone secretagogue receptor (GHSR). The GHSR is located in both the central nervous system and the periphery. Its distribution in the CNS, as assessed by in situ hybridization histochemistry (ISHH), has been described previously in a few mammalian species, although these studies were limited by either the detail provided or the extent of the regions examined. In the present study, we systematically examined the distribution of GHSR mRNA in the adult rat and mouse brains and cervical spinal cords by using ISHH with novel cRNA probes specific for the mRNA encoding functional GHSR (the type 1a variant). We confirmed GHSR mRNA expression in several hypothalamic nuclei, many of which have long been recognized as playing roles in body weight and food intake. GHSR also was found in several other regions previously unknown to express GHSR mRNA, including many parasympathetic preganglionic neurons. Additionally, we found GHSR mRNA within all three components of the dorsal vagal complex, including the area postrema, the nucleus of the solitary tract, and the dorsal motor nucleus of the vagus. Finally, we examined the coexpression of GHSR with tyrosine hydroxylase and cholecystokinin and demonstrate a high degree of GHSR mRNA expression within dopaminergic, cholecystokinin-containing neurons of the substantia nigra and ventral tegmental area.

953 citations


Journal ArticleDOI
TL;DR: Staining patterns after monocular enucleation revealed that the projections of these cells are overwhelmingly crossed except for the projection to the SCN, which is bilaterally symmetrical, and that other ganglion cells do contribute at least some retinal input to these targets.
Abstract: A rare type of ganglion cell in mammalian retina is directly photosensitive. These novel retinal photoreceptors express the photopigment melanopsin. They send axons directly to the suprachiasmatic nucleus (SCN), intergeniculate leaflet (IGL), and olivary pretectal nucleus (OPN), thereby contributing to photic synchronization of circadian rhythms and the pupillary light reflex. Here, we sought to characterize more fully the projections of these cells to the brain. By targeting tau-lacZ to the melanopsin gene locus in mice, ganglion cells that would normally express melanopsin were induced to express, instead, the marker enzyme beta-galactosidase. Their axons were visualized by X-gal histochemistry or anti-beta-galactosidase immunofluorescence. Established targets were confirmed, including the SCN, IGL, OPN, ventral division of the lateral geniculate nucleus (LGv), and preoptic area, but the overall projections were more widespread than previously recognized. Targets included the lateral nucleus, peri-supraoptic nucleus, and subparaventricular zone of the hypothalamus, medial amygdala, margin of the lateral habenula, posterior limitans nucleus, superior colliculus, and periaqueductal gray. There were also weak projections to the margins of the dorsal lateral geniculate nucleus. Co-staining with the cholera toxin B subunit to label all retinal afferents showed that melanopsin ganglion cells provide most of the retinal input to the SCN, IGL, and lateral habenula and much of that to the OPN, but that other ganglion cells do contribute at least some retinal input to these targets. Staining patterns after monocular enucleation revealed that the projections of these cells are overwhelmingly crossed except for the projection to the SCN, which is bilaterally symmetrical.

856 citations


Journal ArticleDOI
TL;DR: The observations suggest that the orexin neurons may integrate a variety of interoceptive and homeostatic signals to increase behavioral arousal in response to hunger, stress, circadian signals, and autonomic challenges.
Abstract: Emotions, stress, hunger, and circadian rhythms all promote wakefulness and behavioral arousal. Little is known about the pathways mediating these influences, but the orexin-producing neurons of the hypothalamus may play an essential role. These cells heavily innervate many wake-promoting brain regions, and mice lacking the orexin neurons have narcolepsy and fail to rouse in response to hunger (Yamanaka et al. [2003] Neuron 38:701-713). To identify the afferents to the orexin neurons, we first injected a retrograde tracer into the orexin neuron field of rats. Retrogradely labeled neurons were abundant in the allocortex, claustrum, lateral septum, bed nucleus of the stria terminalis, and in many hypothalamic regions including the preoptic area, dorsomedial nucleus, lateral hypothalamus, and posterior hypothalamus. Retrograde labeling in the brainstem was generally more modest, but labeling was strong in the periaqueductal gray matter, dorsal raphe nucleus, and lateral parabrachial nucleus. Injection of an anterograde tracer confirmed that most of these regions directly innervate the orexin neurons, with some of the heaviest input coming from the lateral septum, preoptic area, and posterior hypothalamus. In addition, hypothalamic regions preferentially innervate orexin neurons in the medial and perifornical parts of the field, but most projections from the brainstem target the lateral part of the field. Inputs from the suprachiasmatic nucleus are mainly relayed via the subparaventricular zone and dorsomedial nucleus. These observations suggest that the orexin neurons may integrate a variety of interoceptive and homeostatic signals to increase behavioral arousal in response to hunger, stress, circadian signals, and autonomic challenges.

566 citations


Journal ArticleDOI
TL;DR: This work provides a basic description of the organization of the adult human SVZ and provides ultrastructural criteria to identify the different cells types including three distinct types of astrocytes and a group of displaced ependymal cells between Layers II and III.
Abstract: The lateral wall of the lateral ventricle in the human brain contains neural stem cells throughout adult life. We conducted a cytoarchitectural and ultrastructural study in complete postmortem brains (n = 7) and in postmortem (n = 42) and intraoperative tissue (n = 43) samples of the lateral walls of the human lateral ventricles. With varying thickness and cell densities, four layers were observed throughout the lateral ventricular wall: a monolayer of ependymal cells (Layer I), a hypocellular gap (Layer II), a ribbon of cells (Layer III) composed of astrocytes, and a transitional zone (Layer IV) into the brain parenchyma. Unlike rodents and nonhuman primates, adult human glial fibrillary acidic protein (GFAP)+ subventricular zone (SVZ) astrocytes are separated from the ependyma by the hypocellular gap. Some astrocytes as well as a few GFAP-cells in Layer II in the SVZ of the anterior horn and the body of the lateral ventricle appear to proliferate based on proliferating cell nuclear antigen (PCNA) and Ki67 staining. However, compared to rodents, the adult human SVZ appears to be devoid of chain migration or large numbers of newly formed young neurons. It was only in the anterior SVZ that we found examples of elongated Tuj1+ cells with migratory morphology. We provide ultrastructural criteria to identify the different cells types in the human SVZ including three distinct types of astrocytes and a group of displaced ependymal cells between Layers II and III. Ultrastructural analysis of this layer revealed a remarkable network of astrocytic and ependymal processes. This work provides a basic description of the organization of the adult human SVZ. J. Comp. Neurol. 494:415–434, 2006. © 2005 Wiley-Liss, Inc.

533 citations


Journal ArticleDOI
TL;DR: An age‐dependent decrease in SGZ proliferation in homozygous PDAPP mice is reported, suggesting altered neurogenesis in the PDAPP mouse may contribute to the age‐related cognitive deficits reported in this model of AD and may be a useful adjunct target for assessing the impact of AD therapies.
Abstract: Abnormal subgranular zone (SGZ) neurogenesis is proposed to contribute to Alzheimer's disease (AD)-related decreases in hippocampal function. Our goal was to examine hippocampal neurogenesis in the PDAPP mouse, a model of AD with age-dependent accumulation of amyloid-beta(42) (Abeta(42))-containing plaques that is well studied with regard to AD therapies. A secondary goal was to determine whether altered neurogenesis in the PDAPP mouse is associated with abnormal maturation or number of mature cells. A tertiary goal was to provide insight into why hippocampal neurogenesis appears to be increased in AD post-mortem tissue and decreased in most AD mouse models. We report an age-dependent decrease in SGZ proliferation in homozygous PDAPP mice. At 1 year of age, PDAPP mice also had new dentate gyrus granule neurons with abnormal maturation and fewer dying cells relative to control mice. In contrast to decreased SGZ cell birth, PDAPP mice had increased birth of immature neurons in the outer portion of the granule cell layer (oGCL), providing insight into why some studies link AD with increased neurogenesis. However, these ectopic oGCL cells were still rare compared with SGZ proliferating cells, emphasizing that the primary characteristic of PDAPP mice is decreased neurogenesis. The decrease in SGZ neurogenesis was not associated with an age-dependent loss of dentate granule neurons. The altered neurogenesis in the PDAPP mouse may contribute to the age-related cognitive deficits reported in this model of AD and may be a useful adjunct target for assessing the impact of AD therapies.

357 citations


Journal ArticleDOI
TL;DR: It is proposed that HASs, which can retain HA on the cell surface, may act as a link between PNNs and neurons, and HAS and link proteins might be key molecules for PNN formation and stability.
Abstract: The decrease in plasticity that occurs in the central nervous system during postnatal development is accompanied by the appearance of perineuronal nets (PNNs) around the cell body and dendrites of many classes of neuron. These structures are composed of extracellular matrix molecules, such as chondroitin sulfate proteoglycans (CSPGs), hyaluronan (HA), tenascin-R, and link proteins. To elucidate the role played by neurons and glial cells in constructing PNNs, we studied the expression of PNN components in the adult rat cerebellum by immunohistochemistry and in situ hybridization. In the deep cerebellar nuclei, only large excitatory neurons were surrounded by nets, which contained the CSPGs aggrecan, neurocan, brevican, versican, and phosphacan, along with tenascin-R and HA. Whereas both net-bearing neurons and glial cells were the sources of CSPGs and tenascin-R, only the neurons expressed the mRNA for HA synthases (HASs), cartilage link protein, and link protein Bral2. In the cerebellar cortex, Golgi neurons possessed PNNs and also synthesized HASs, cartilage link protein, and Bral2 mRNAs. To see whether HA might link PNNs to the neuronal cell surface by binding to a receptor, we investigated the expression of the HA receptors CD44, RHAMM, and LYVE-1. No immunolabelling for HA receptors on the membrane of net-bearing neurons was found. We therefore propose that HASs, which can retain HA on the cell surface, may act as a link between PNNs and neurons. Thus, HAS and link proteins might be key molecules for PNN formation and stability. J. Comp. Neurol. 494: 559 –577, 2006. © 2005 Wiley-Liss, Inc.

298 citations


Journal ArticleDOI
TL;DR: The topography and connections of Dl are remarkably similar to those of the hippocampus of tetrapods, whereas the topography of Dm is similar toThose of the amygdala.
Abstract: Biotinylated dextran amine and fluorescent carbocyanine dye (DiI) were used to examine connections of the lateral (Dl) and medial (Dm) divisions of the goldfish pallium. Besides numerous intrinsic telencephalic connections to Dl and Dm, major ascending projections to these pallial divisions arise in the preglomerular complex of the posterior tuberculum, rather than in the dorsal thalamus. The rostral subnucleus of the lateral preglomerular nucleus receives auditory input via the medial pretoral nucleus, lateral line input via the ventrolateral toral nucleus, and visual input via the optic tectum, and it projects to both Dl and Dm. The anterior preglomerular nucleus and caudal subnucleus of the lateral preglomerular nucleus receive auditory input via the central toral nucleus and project to Dm. This pallial division also receives chemosensory information via the medial preglomerular nucleus. The central posterior (CP) nucleus, which receives both auditory and visual inputs, also projects to Dm and is the only dorsal thalamic nucleus projecting to the pallium. Thus, both Dl and Dm clearly receive multisensory inputs. Major projections of CP and projections of all other dorsal thalamic nuclei are to the subpallium, however. Descending projections of Dl are primarily to the preoptic area and the caudal hypothalamus, whereas descending projections of Dm are more extensive and particularly heavy to the anterior tuber and nucleus diffusus of the hypothalamus. The topography and connections of Dl are remarkably similar to those of the hippocampus of tetrapods, whereas the topography and connections of Dm are similar to those of the amygdala.

276 citations


Journal ArticleDOI
TL;DR: This cytoarchitectonic mapping study was to determine whether consistent regions and borders can be found within the cortex of the anterior IPS in a population of 10 post‐mortem human brains and concludes that the human IPS has a more finely grained parcellation than shown in Brodmann's map.
Abstract: Anatomical studies in the macaque cortex and functional imaging studies in humans have demonstrated the existence of different cortical areas within the intraparietal sulcus (IPS). Such functional segregation, however, does not correlate with presently available architectonic maps of the human brain. This is particularly true for the classical Brodmann map, which is still widely used as an anatomical reference in functional imaging studies. The aim of this cytoarchitectonic mapping study was to use previously defined algorithms to determine whether consistent regions and borders can be found within the cortex of the anterior IPS in a population of 10 post-mortem human brains. Two areas, the human intraparietal area 1 (hIP1) and the human intraparietal area 2 (hIP2), were delineated in serial histological sections of the anterior, lateral bank of the human IPS. The region hIP1 is located posterior and medial to hIP2, and the former is always within the depths of the IPS. The latter, on the other hand, sometimes reaches the free surface of the superior parietal lobule. The delineations were registered to standard reference space, and probabilistic maps were calculated, thereby quantifying the intersubject variability in location and extent of both areas. In the future, they can be a tool for analyzing structure-function relationships and a basis for determining degrees of homology in the IPS among anthropoid primates. We conclude that the human IPS has a more finely grained parcellation than shown in Brodmann's map.

270 citations


Journal ArticleDOI
TL;DR: The overall pattern of axonal projections from the rat BSTam was analyzed with the Phaseolus vulgaris‐leucoagglutinin anterograde pathway tracing method and suggests that the BSTam is part of a striatopallidal differentiation involved in coordinating neuroendocrine, autonomic, and behavioral or somatic responses associated with maintaining energy balance homeostasis.
Abstract: The anteromedial area of the bed nuclei of the stria terminalis (BSTam) is the relatively undifferentiated region of the anterior medial (anteromedial) group of the bed nuclei of the stria terminalis (BSTamg), which also includes the more distinct dorsomedial, magnocellular, and ventral nuclei. The overall pattern of axonal projections from the rat BSTam was analyzed with the PHAL anterograde pathway tracing method. Brain areas receiving relatively moderate to strong inputs from the BSTam fall into five general categories: neuroendocrine system (regions containing pools of magnocellular oxytocin neurons, and parvicellular corticotropin-releasing hormone, thyrotropin-releasing hormone, somatostatin, and dopamine neurons); central autonomic control network (central amygdalar nucleus, descending paraventricular nucleus, and ventrolateral periaqueductal gray); hypothalamic visceromotor pattern generator network (5 of 6 known components); behavior control column (descending paraventricular nucleus and associated arcuate nucleus; ventral tegmental area and associated nucleus accumbens and substantia innominata); and behavioral state control (supramammillary and tuberomammillary nuclei). The BSTam projects lightly to thalamocortical feedback loops (via the medial-midline-intralaminar thalamus). Its pattern of axonal projections, combined with its pattern of neural inputs (the most varied of all BST cell groups), suggest that the BSTam is part of a striatopallidal differentiation involved in coordinating neuroendocrine, autonomic, and behavioral or somatic responses associated with maintaining energy balance homeostasis.

269 citations


Journal ArticleDOI
TL;DR: Patterns of neurodegeneration and intraparenchymal inflammation after traumatic spinal cord injury (SCI) in mice known to exhibit varying degrees of EAE susceptibility are described and genetic predisposition to EAE is predicted but not lesion size/length or locomotor recovery.
Abstract: Susceptibility to neuroinflammatory disease is influenced in part by genetics. Recent data indicate that survival of traumatized neurons is strain dependent and influenced by polygenic loci that control resistance/susceptibility to experimental autoimmune encephalomyelitis (EAE), a model of CNS autoimmune disease. Here, we describe patterns of neurodegeneration and intraparenchymal inflammation after traumatic spinal cord injury (SCI) in mice known to exhibit varying degrees of EAE susceptibility [EAE-resistant (r) or EAE-susceptible (s) mice]. Spinal cords from C57BL/6 (EAE-s), C57BL/10 (EAE-r), BALB/c (EAE-r), and B10.PL (EAE-s) mice were prepared for stereological and immunohistochemical analysis at 6 hours or 3, 7, 14, 28, or 42 days following midthoracic (T9) spinal contusion injury. In general, genetic predisposition to EAE predicted the magnitude of intraparenchymal inflammation but not lesion size/length or locomotor recovery. Specifically, microglia/macrophage activation, recruitment of neutrophils and lymphocytes, and de novo synthesis of MHC class II were greatest in C57BL/6 mice and least in BALB/c mice at all times examined. However, lesion volume and axial spread of neurodegeneration were similar in C57BL/6 and BALB/c mice and were significantly greater than in C57BL/10 or B10.PL mice. Strains with marked intraspinal inflammation also developed the most intense lesion fibrosis. Thus, strain-dependent neuroinflammation was observed after SCI, but without a consistent relationship to EAE susceptibility or lesion progression. Only in C57BL/6 mice was the magnitude of intraspinal inflammation predictive of secondary neurodegeneration, functional recovery, or fibrosis.

263 citations


Journal ArticleDOI
TL;DR: The results suggest that dividing, NG2‐expressing progenitor cells are vulnerable to injury, but a separate, immature population of neural stem and/or progenitors is activated by injury and rapidly divides to replace this vulnerable population.
Abstract: The adult mammalian spinal cord contains neural stem and/or progenitor cells that slowly multiply throughout life and differentiate exclusively into glia. The contribution of adult progenitors to repair has been highlighted in recent studies, demonstrating extensive cell proliferation and gliogenesis following central nervous system (CNS) trauma. The present experiments aimed to determine the relative roles of endogenously dividing progenitor cells versus quiescent progenitor cells in posttraumatic gliogenesis. Using the mitotic indicator bromodeoxyuridine (BrdU) and a retroviral vector, we found that, in the adult female Fisher 344 rat, endogenously dividing neural progenitors are acutely vulnerable in response to T8 dorsal hemisection spinal cord injury. We then studied the population of cells that divide postinjury in the injury epicenter by delivering BrdU or retrovirus at 24 hours after spinal cord injury. Animals were euthanized at five timepoints postinjury, ranging from 6 hours to 9 weeks after BrdU delivery. At all timepoints, we observed extensive proliferation of ependymal and periependymal cells that immunohistochemically resembled stem/progenitor cells. BrdU+ incorporation was noted to be prominent in NG2-immunoreactive progenitors that matured into oligodendrocytes, and in a transient population of microglia. Using a green fluorescence protein (GFP) hematopoietic chimeric mouse, we determined that 90% of the dividing cells in this early proliferation event originate from the spinal cord, whereas only 10% originate from the bone marrow. Our results suggest that dividing, NG2-expressing progenitor cells are vulnerable to injury, but a separate, immature population of neural stem and/or progenitor cells is activated by injury and rapidly divides to replace this vulnerable population.

Journal ArticleDOI
TL;DR: The nucleus reuniens (RE) is the largest of the midline nuclei of the thalamus and exerts strong excitatory actions on the hippocampus and medial prefrontal cortex, and efferent projections of RE as well as those of the rhomboid nucleus (RH) located dorsal to RE are examined.
Abstract: The nucleus reuniens (RE) is the largest of the midline nuclei of the thalamus and exerts strong excitatory actions on the hippocampus and medial prefrontal cortex. Although RE projections to the hippocampus have been well documented, no study using modern tracers has examined the totality of RE projections. With the anterograde anatomical tracer Phaseolus vulgaris leuccoagglutinin, we examined the efferent projections of RE as well as those of the rhomboid nucleus (RH) located dorsal to RE. Control injections were made in the central medial nucleus (CEM) of the thalamus. We showed that the output of RE is almost entirely directed to the hippocampus and "limbic" cortical structures. Specifically, RE projects strongly to the medial frontal polar, anterior piriform, medial and ventral orbital, anterior cingulate, prelimbic, infralimbic, insular, perirhinal, and entorhinal cortices as well as to CA1, dorsal and ventral subiculum, and parasubiculum of the hippocampus. RH distributes more widely than RE, that is, to several RE targets but also significantly to regions of motor, somatosensory, posterior parietal, retrosplenial, temporal, and occipital cortices; to nucleus accumbens; and to the basolateral nucleus of amygdala. The ventral midline thalamus is positioned to exert significant control over fairly widespread regions of the cortex (limbic, sensory, motor), hippocampus, dorsal and ventral striatum, and basal nuclei of the amygdala, possibly to coordinate limbic and sensorimotor functions. We suggest that RE/RH may represent an important conduit in the exchange of information between subcortical-cortical and cortical-cortical limbic structures potentially involved in the selection of appropriate responses to specific and changing sets of environmental conditions.

Journal ArticleDOI
TL;DR: There is a close functional relationship between dynorphin and NKB peptides within the arcuate nucleus of the rat, which may include an autofeedback loop mediated through NK3R.
Abstract: Considerable evidence suggests that dynorphin and neurokinin B (NKB) neurons in the hypothalamic arcuate nucleus participate in the sex-steroid regulation of reproduction. In the present study, we used dual-label immunofluorescence to explore the distribution of prodynorphin and proNKB immunoreactivity in the rat hypothalamus. Additionally, we investigated whether arcuate prodynorphin-ir (immunoreactive) neurons expressed the neurokinin 3 receptor (NK3R) or nuclear estrogen receptor-alpha (ERalpha). We found that the majority of prodynorphin-ir neurons in the rat arcuate nucleus expressed proNKB, whereas nearly all (99%) of the proNKB neurons were immunoreactive for prodynorphin. The arcuate nucleus was the only site in the hypothalamus where neuronal somata coexpressing prodynorphin and proNKB-immunoreactivity were identified. A dense plexus of double-labeled prodynorphin/proNKB-ir fibers was found within the arcuate nucleus extending to the median eminence and throughout the periventricular zone of the hypothalamus. Prodynorphin/proNKB fibers were also identified in the paraventricular nucleus, anterior hypothalamic area, medial preoptic area, median preoptic nucleus, anteroventral periventricular nucleus, and bed nucleus of the stria terminalis in a distribution consistent with previously described arcuate nucleus projections. Interestingly, the majority of prodynorphin-ir neurons in the arcuate nucleus expressed NK3R, and nearly 100% of the prodynorphin-ir neurons contained nuclear ERalpha. Our results suggest that there is a close functional relationship between dynorphin and NKB peptides within the arcuate nucleus of the rat, which may include an autofeedback loop mediated through NK3R. The diverse hypothalamic projections of fibers expressing both prodynorphin and proNKB provide evidence that these neurons may participate in a variety of homeostatic and neuroendocrine processes.

Journal ArticleDOI
TL;DR: Analysis of the double‐labeled sections indicates that NTPDase2 immunoreactivity is found on cell processes that often envelop other taste cells, reminiscent of type I cells, indicating that the presence of ecto‐ATPase in taste buds likely reflects the importance of ATP as an intercellular signaling molecule in this system.
Abstract: The presence of one or more calcium-dependent ecto-ATPases (enzymes that hydrolyze extracellular 5′-triphosphates) in mammalian taste buds was first shown histochemically. Recent studies have established that dominant ecto-ATPases consist of enzymes now called nucleoside triphosphate diphosphohydrolases (NTPDases). Massively parallel signature sequencing (MPSS) from murine taste epithelium provided molecular evidence suggesting that NTPDase2 is the most likely member present in mouse taste papillae. Immunocytochemical and enzyme histochemical staining verified the presence of NTPDase2 associated with plasma membranes in a large number of cells within all mouse taste buds. To determine which of the three taste cell types expresses this enzyme, double label assays were performed using antisera directed against the glial glutamate/aspartate transporter, (GLAST), the transduction pathway proteins phospholipaseC β2 (PLCβ2) or the G protein subunit α-gustducin, and serotonin (5HT) as markers of type I, II or III taste cells, respectively. Analysis of the double labeled sections indicates that NTPDase2 immunoreactivity is found on cell processes that often envelop other taste cells, reminiscent of type I cells. In agreement with this observation, NTPDase2 was located to the same membrane as GLAST, indicating that this enzyme is present in type I cells. The presence of ecto-ATPase in taste buds likely reflects the importance of ATP as an intercellular signaling molecule in this system.

Journal ArticleDOI
TL;DR: The results indicate that parallel subcortical pathways target the core and medial belt regions and that RM and CM represent functionally distinct areas within the medial belt auditory cortex.
Abstract: The auditory cortex of primates contains a core region of three primary areas surrounded by a belt region of secondary areas. Recent neurophysiological studies suggest that the belt areas medial to the core have unique functional roles, including multisensory properties, but little is known about their connections. In this study and its companion, the cortical and subcortical connections of the core and medial belt regions of marmoset monkeys were compared to account for functional differences between areas and refine our working model of the primate auditory cortex. Anatomical tracer injections targeted two core areas (A1 and R) and two medial belt areas (rostromedial [RM] and caudomedial [CM]). RM and CM had topographically weighted connections with all other areas of the auditory cortex ipsilaterally, but these were less widespread contralaterally. CM was densely connected with caudal auditory fields, the retroinsular (Ri) area of the somatosensory cortex, the superior temporal sulcus (STS), and the posterior parietal and entorhinal cortex. The connections of RM favored rostral auditory areas, with no clear somatosensory inputs. RM also projected to the lateral nucleus of the amygdala and tail of the caudate nucleus. A1 and R had topographically weighted connections with medial and lateral belt regions, infragranular inputs from the parabelt, and weak connections with fields outside the auditory cortex. The results indicated that RM and CM are distinct areas of the medial belt region with direct inputs from the core. CM also has somatosensory input and may correspond to an area on the posteromedial transverse gyrus of humans and the anterior auditory field of other mammals.

Journal ArticleDOI
TL;DR: The data reported here show that behavior‐ or seizure‐induced Arc expression in the hippocampus, primary somatosensory cortex, and dorsal striatum of rats colocalizes only with neuronal (NeuN‐positive) and not with glial (GFAP‐ positive) cells, consistent with the hypothesis that Arc and CaMKII act as plasticity partners to promote functional and/or structural synaptic modifications that accompany learning.
Abstract: Active behavior, such as exploring a novel environment, induces the expression of the immediate-early gene Arc (activity-regulated cytoskeletal associated protein, or Arg 3.1) in many brain regions, including the hippocampus, neocortex, and striatum. Arc messenger ribonucleic acid and protein are localized in activated dendrites, and Arc protein is required for the maintenance of long-term potentiation and memory consolidation. Although previous evidence suggests that Arc is expressed in neurons, there is no direct demonstration that only neurons can express Arc. Furthermore, there is no characterization of the main neuronal types that express Arc. The data reported here show that behavior- or seizure-induced Arc expression in the hippocampus, primary somatosensory cortex, and dorsal striatum of rats colocalizes only with neuronal (NeuN-positive) and not with glial (GFAP-positive) cells. Furthermore, Arc was found exclusively in non-GABAergic alpha-CaMKII-positive hippocampal and neocortical neurons of rats that had explored a novel environment. Some GAD65/67-positive neurons in these regions were observed to express Arc, but only after a very strong stimulus (electroconvulsive seizure). In the dorsal striatum, spatial exploration induced Arc only in GABAergic and alpha-CaMKII-positive neurons. Combined, these results show that although a very strong stimulus (seizure) can induce Arc in a variety of neurons, behavior induces Arc in the CaMKII-positive principal neurons of the hippocampus, neocortex, and dorsal striatum. These results, coupled with recent in vitro findings of interactions between Arc and CaMKII, are consistent with the hypothesis that Arc and CaMKII act as plasticity partners to promote functional and/or structural synaptic modifications that accompany learning.

Journal ArticleDOI
TL;DR: The overall projection pattern of a tiny bed nuclei of the stria terminalis anteromedial group differentiation, the dorsomedial nucleus (BSTdm), was analyzed with the Phaseolus vulgaris‐leucoagglutinin anterograde pathway tracing method in rats and suggests that the BSTdm is part of a striatopallidal differentiation involved in coordinating the homeostatic and behavioral responses associated thirst and salt appetite.
Abstract: The overall projection pattern of a tiny bed nuclei of the stria terminalis anteromedial group differentiation, the dorsomedial nucleus (BSTdm), was analyzed with the Phaseolus vulgaris-leucoagglutinin anterograde pathway tracing method in rats. Many brain regions receive a relatively moderate to strong input from the BSTdm. They fall into eight general categories: humeral sensory-related (subfornical organ and median preoptic nucleus, involved in initiating drinking behavior and salt appetite), neuroendocrine system (magnocellular: oxytocin, vasopressin; parvicellular: gonadotropin-releasing hormone, somatostatin, thyrotropin-releasing hormone, corticotropin-releasing hormone), central autonomic control network (central amygdalar nucleus, BST anterolateral group, descending paraventricular hypothalamic nucleus, retrochiasmatic area, ventrolateral periaqueductal gray, Barrington's nucleus), hypothalamic visceromotor pattern-generator network (five of six known components), behavior control column (ingestive: descending paraventricular nucleus; reproductive: lateral medial preoptic nucleus; defensive: anterior hypothalamic nucleus; foraging: ventral tegmental area, along with interconnected nucleus accumbens and substantia innominata), orofacial motor control (retrorubral area), thalamocortical feedback loops (paraventricular, central medial, intermediodorsal, and medial mediodorsal nuclei; nucleus reuniens), and behavioral state control (subparaventricular zone, ventrolateral preoptic nucleus, tuberomammillary nucleus, supramammillary nucleus, lateral habenula, and raphe nuclei). This pattern of axonal projections, and what little is known of its inputs suggest that the BSTdm is part of a striatopallidal differentiation involved in coordinating the homeostatic and behavioral responses associated thirst and salt appetite, although clearly it may relate them to other functions as well. The BSTdm generates the densest known inputs directly to the neuroendocrine system from any part of the cerebral hemispheres. J. Comp. Neurol. 494:75–107, 2006. © 2005 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: The results indicate that olfactory input in the honeybee is processed via two separate, mainly uPN pathways to the MB calyx and LH and several pathway to the lateral protocerebrum.
Abstract: The antennal lobes (ALs) are the primary olfactory centers in the insect brain. In the AL of the honeybee, olfactory glomeruli receive input via four antennal sensory tracts (T1–4). Axons of projection neurons (PNs) leave the AL via several antenno-cerebral tracts (ACTs). To assign the input–output connectivity of all glomeruli, we investigated the spatial relationship of the antennal tracts and two prominent AL output tracts (medial and lateral ACT) mainly formed by uniglomerular (u) PNs using fluorescent tracing, confocal microscopy, and 3D analyses. Furthermore, we investigated the projections of all ACTs in higher olfactory centers, the mushroom-bodies (MB) and lateral horn (LH). The results revealed a clear segregation of glomeruli into two AL hemispheres specifically supplied by PNs of the medial and lateral ACT. PNs of the lateral ACT innervate glomeruli in the ventral-rostral AL and primarily receive input from T1 (plus a few glomeruli from T2 and T3). PNs of the medial ACT innervate glomeruli in the dorsal-caudal hemisphere, and mainly receive input from T3 (plus a few glomeruli from T2 and T4). The PNs of the m- and l-ACT terminate in different areas of the MB calyx and LH and remain largely segregated. Tracing of three mediolateral (ml) ACTs mainly formed by multiglomerular PNs revealed terminals in distinct compartments of the LH and in three olfactory foci within the lateral protocerebrum. The results indicate that olfactory input in the honeybee is processed via two separate, mainly uPN pathways to the MB calyx and LH and several pathways to the lateral protocerebrum. J. Comp. Neurol. 499:933–952, 2006. © 2006 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: The results suggest that whether or not the ML is specialized for CO2 sensing, its complex neuropil likely regulates the activity of RTN chemosensitive neurons and the chemossensitive neurons of RTn may provide a chemical drive to multiple respiratory outflows, insofar as RTN innervates the entire VRC.
Abstract: The rat retrotrapezoid nucleus (RTN) contains candidate central chemoreceptors that have extensive dendrites within the marginal layer (ML). This study describes the axonal projections of RTN neurons and their probable synaptic inputs. The ML showed a dense plexus of nerve terminals immunoreactive (ir) for markers of glutamatergic (vesicular glutamate transporters VGLUT1-3), gamma-aminobutyric acid (GABA)-ergic, adrenergic, serotonergic, cholinergic, and peptidergic transmission. The density of VGLUT3-ir terminals tracked the location of RTN chemoreceptors. The efferent and afferent projections of RTN were studied by placing small iontophoretic injections of anterograde (biotinylated dextran amine; BDA) and retrograde (cholera toxin B) tracers where RTN chemoreceptors have been previously recorded. BDA did not label the nearby C1 cells. BDA-ir varicosities were found in the solitary tract nucleus (NTS), all ventral respiratory column (VRC) subdivisions, A5 noradrenergic area, parabrachial complex, and spinal cord. In each target region, a large percentage of the BDA-ir varicosities was VGLUT2-ir (41-83%). Putative afferent input to RTN originated from spinal cord, caudal NTS, area postrema, VRC, dorsolateral pons, raphe nuclei, lateral hypothalamus, central amygdala, and insular cortex. The results suggest that 1) whether or not the ML is specialized for CO(2) sensing, its complex neuropil likely regulates the activity of RTN chemosensitive neurons; 2) the catecholaminergic, cholinergic, and serotonergic innervation of RTN represents a possible substrate for the known state-dependent control of RTN chemoreceptors; 3) VGLUT3-ir terminals are a probable marker of RTN; and 4) the chemosensitive neurons of RTN may provide a chemical drive to multiple respiratory outflows, insofar as RTN innervates the entire VRC.

Journal ArticleDOI
TL;DR: A systematic analysis of the visual projection neurons that connect the optic lobe and the central brain of Drosophila melanogaster identified 44 pathways that form columnar arborization in the lobula and nine terminate in the ventrolateral protocerebrum.
Abstract: In insects, visual information is processed in the optic lobe and conveyed to the central brain. Although neural circuits within the optic lobe have been studied extensively, relatively little is known about the connection between the optic lobe and the central brain. To understand how visual information is read by the neurons of the central brain, and what kind of centrifugal neurons send the control signal from the central brain to the optic lobe, we performed a systematic analysis of the visual projection neurons that connect the optic lobe and the central brain of Drosophila melanogaster. By screening ∼4,000 GAL4 enhancer-trap strains we identified 44 pathways. The overall morphology and the direction of information of each pathway were investigated by expressing cytoplasmic and presynapsis-targeted fluorescent reporters. A canonical nomenclature system was introduced to describe the area of projection in the central brain. As the first part of a series of articles, we here describe 14 visual projection neurons arising specifically from the lobula. Eight pathways form columnar arborization in the lobula, whereas the remaining six form tangential or tree-like arborization. Eleven are centripetal pathways, among which nine terminate in the ventrolateral protocerebrum. Terminals of each columnar pathway form glomerulus-like structures in different areas of the ventrolateral protocerebrum. The posterior lateral protocerebrum and the optic tubercle were each contributed by a single centripetal pathway. Another pathway connects the lobula on each side of the brain. Two centrifugal pathways convey signals from the posterior lateral protocerebrum to the lobula. J. Comp. Neurol. 497:928–958, 2006. © 2006 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: In both white and gray matter, the periods of high GluR2 deficiency correspond to those of regional susceptibility to hypoxic/ischemic injury in each of the two rat strains, supporting prior studies suggesting a critical role for Ca2+‐permeable AMPARs in excitotoxic cellular injury and epileptogenesis.
Abstract: The immature brain is highly susceptible to hypoxia/ischemia (H/I), and perinatal H/I brain injury represents a major cause of neurodevelopmental disorders in both preterm and term infants (Volpe, 2001; Ferriero, 2004). In preterm infants, H/I causes primarily white matter injury, termed periventricular leukomalacia (PVL; Banker and Larroche, 1962; Okumura et al., 1997; Volpe, 2001). In contrast, H/I in term newborns causes predominantly gray matter lesions and seizures (Hauser et al., 1993; Maller et al., 1998; Roland et al., 1998; Saliba et al., 1999; Volpe, 2001). Rodent models of perinatal H/I brain injury reflect similar age-dependent regional differences, despite slight age variations in different rat strains. During the first week of life (postnatal day P1–P7), H/I results in selective white matter injury, characterized by loss of premyelinating oligodendrocytes (pre-OLs), followed by hypomyelination (Sheldon et al., 1996; Follett et al., 2000; Cai et al., 2001; Back et al., 2002; Liu et al., 2002). During the second postnatal week (P8–14), H/I causes spontaneous electro-graphic and behavioral seizures (Jensen et al., 1991; Owens et al., 1997), as well as extensive cortical and hippocampal neuronal loss (Towfighi et al., 1997; Chen et al., 1998). H/I causes glutamate accumulation in both gray and white matter structures in the developing rat brain (Benveniste et al., 1984; Andine et al., 1991; Silverstein et al., 1991; Hagberg, 1992; Loeliger et al., 2003), implying a key role for glutamate receptors (GluRs) in the pathophysiology of perinatal H/I brain injury. Glutamate receptor subtypes include the N-methyl-D-aspartate receptors (NMDARs), the α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs), and kainate receptors (KARs; Hollmann and Heinemann, 1994; Michaelis, 1998). Whereas neurons and astrocytes express both NMDARs and non-NMDARs (Petralia and Wenthold, 1992; Petralia et al., 1994; Conti et al., 1994; Shelton and McCarthy, 1999; Schipke et al., 2001), OLs express primarily non-NMDARs (Patneau et al., 1994; Gallo et al., 1994; Rosenberg et al., 2003). Compelling evidence for a critical role of GluRs in perinatal H/I injury is provided by experimental therapeutic trials. AMPAR antagonists are highly protective to developing OLs against H/I injury at P7 (Follett et al., 2000, 2004) and are effective in suppressing hypoxia-induced seizures at P10–P12 (Jensen et al., 1995; Koh and Jensen, 2001). Similarly, in P7–P10 rats, NMDAR and AMPAR antagonists have been shown to attenuate H/I-induced neuronal injury (Olney et al., 1989; Hagberg et al., 1994; Chen et al., 1998). AMPAR-mediated signaling and excitotoxicity depend on the functional properties of the receptor complex, such as Ca2+ permeability (Gu et al., 1996; Friedman and Koudinov, 1999; Sanchez et al., 2001, 2005; Jensen et al., 2001; Deng et al., 2003; Follett et al., 2004), which in turn are dictated by subunit composition. AMPARs are heteromeric complexes composed of four subunits (GluR1 through GluR4), and receptors lacking the GluR2 subunit are Ca2+-permeable, whereas those including the GluR2 subunit are impermeable to Ca2+ (Burnashev et al., 1992; Seeburg, 1993; Jonas et al., 1994; Washburn et al., 1997). In the immature rat brain, GluR2 expression is low relative to non-GluR2 subunits (Pellegrini-Giampietro et al., 1991, 1992; Sanchez et al., 2001; Kumar et al., 2002), suggesting that AMPARs with increased Ca2+ permeability are abundantly expressed during this time window. Indeed, functional studies in situ in immature rodent brain have confirmed increased Ca2+ influx through AMPARs expressed on pre-OLs (Fulton et al., 1992; Bergles et al., 2000; Follett et al., 2004) and developing hippocampal and pyramidal neurons (Sanchez et al., 2001; Kumar et al., 2002), supporting a close correlation between GluR2 expression level and receptor function. In this two-part series of studies, we examine evidence for a relationship between differential distribution of GluR2-lacking (Ca2+-permeable) AMPARs and selective white and gray matter vulnerability to H/I in both rodent (part I) and human (Talos et al., 2006). We hypothesize that the GluR2-lacking AMPARs represent a key factor in age-dependent regional susceptibility to H/I, so regional and temporal expression of these receptors would correspond in a cell-specific manner to patterns of susceptibility to H/I. In addition, we hypothesize that subtle differences in age windows of susceptibility between rat strains are due to strain-dependent differences in temporal onset and progression of AMPAR subunits on specific cell types. In part I, we analyzed the developmental profile of each AMPAR subunit in both white matter and cortex from Long Evans (LE) rats during the first 3 postnatal weeks (postnatal days P1–P21) and specifically evaluated the developmental regulation of the GluR2 subunit relative to other AMPAR subunits by immunoblotting and immunofluorescence double labeling. To examine strain-dependent differences in AMPAR subunit expression, a subset of immunofluorescence double-labeling experiments for specific AMPAR subunits was conducted in parallel in both LE and Sprague Dawley (SD) rat pups, ages P1–P14. In part II (Talos et al., 2006), human parietal white matter and cortex from cases ranging between 18 postconceptional weeks (PCW) and 210 PCW (approximately 3.3 years) were similarly evaluated for age-dependent variations in AMPAR subunit expression by immunoblotting and immunofluorescence double labeling.

Journal ArticleDOI
TL;DR: A comprehensive projection map of the auditory receptor cells (Johnston's organ neurons: JONs) from the antennae to the primary auditory center of the Drosophila brain is established, suggesting that each JON group, and hence each zone of thePrimary auditory center, might sense different aspects of sensory signals.
Abstract: We established a comprehensive projection map of the auditory receptor cells (Johnston's organ neurons: JONs) from the antennae to the primary auditory center of the Drosophila brain. We found 477 ± 24 cell bodies of JONs, which are arranged like a “bottomless bowl” within the auditory organ. The target of the JONs in the brain comprises five spatially segregated zones, each of which is contributed by bundles of JON axons that gradually branch out from the antennal nerve. Four zones are confined in the antennal mechanosensory and motor center, whereas one zone further extends over parts of the ventrolateral protocerebrum and the subesophageal ganglion. Single-cell labeling with the FLP-out technique revealed that most JONs innervate only a single zone, indicating that JONs can be categorized into five groups according to their target zones. Within each zone, JONs innervate various combinations of subareas. We classified these five zones into 19 subareas according to the branching patterns and terminal distributions of single JON axons. The groups of JONs that innervate particular zones or subareas of the primary auditory center have their cell bodies in characteristic locations of the Johnston's organ in the antenna, e.g., in concentric rings or in paired clusters. Such structural organization suggests that each JON group, and hence each zone of the primary auditory center, might sense different aspects of sensory signals. J. Comp. Neurol. 499:317–356, 2006. © 2006 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: The results indicate that VGLUT2 is expressed in subsets of A10 and A11 dopamine neurons, which might release dopamine and glutamate separately from different varicosities in the majority of their single axons.
Abstract: Vesicular glutamate transporters (VGLUT1, -2, and -3) mediate the accumulation of transmitter glutamate into synaptic vesicles in glutamatergic neurons. VGLUT1 and VGLUT2 are more reliable glutamatergic neuron markers, since VGLUT3 also exists in other neuron types. To study whether the dopaminergic neuron uses glutamate as a cotransmitter, we analyzed VGLUTs expression in dopamine neurons of adult male rats by in situ hybridization and immunohistochemistry. In the ventral midbrain, in situ hybridization analysis revealed no VGLUT1 mRNA expression, a widespread but discrete pattern of VGLUT2 mRNA expression, and a highly limited expression of VGLUT3 mRNA. Reverse-transcriptase polymerase chain reaction analysis detected full-length VGLUT2 gene transcripts in the ventral midbrain. Using in situ hybridization combined with tyrosine hydroxylase (TH) immunostaining, only VGLUT2 signals were detectable in some TH-labeled neurons of A10 dopamine neuron groups, with the highest incidence (20%) in the rostral linear nucleus of the ventral tegmental area. In the forebrain, VGLUT2 signals were demonstrated in half of the A11 TH-labeled neurons in the hypothalamus. Double-label immunostaining for VGLUT2 and vesicular monoamine transporter 2 or TH showed that double-labeled varicosities are rarely observed in any target regions examined of A10 and A11 dopamine neuron groups. These results indicate that VGLUT2 is expressed in subsets of A10 and A11 dopamine neurons, which might release dopamine and glutamate separately from different varicosities in the majority of their single axons.

Journal ArticleDOI
TL;DR: An enhancer trap marker called R32 is described that specifically reveals several previously undescribed aspects of the fly's central neuronal pacemakers, and it is shown that the neuropeptide IPNamide is specifically expressed by this DN1 subclass, which implicateIPNamide as a second candidate circadian transmitter in the Drosophila brain.
Abstract: Overt biological rhythms such as the predictable daily leaf movements of plants and the sleep/wake cycle of animals are ultimately driven by molecular oscillations. Throughout the living world such oscillations are sustained by intracellular transcriptional/translational feedback loops (reviewed by Dunlap, 1999). In Drosophila many components of the molecular clock are known in detail. Products of the clock genes period (per), timeless (tim), Clock, cycle, vrille, and PAR domain protein 1 (Pdp1) form two interconnected and self-sustained transcriptional feedback loops, the kinetics of which are regulated by clock protein interactions and attendant kinases (recently reviewed by Hardin, 2004; Schoning and Staiger, 2005; Taghert and Lin, 2005). Although the core clock genes are expressed rhythmically throughout the fly's body, only a few small groups of clock-expressing neurons in the central brain (termed pacemakers) are necessary and sufficient for the organization and maintenance of rhythmic locomotor activity (reviewed by Hall, 2005; Helfrich-Forster, 2005). When these pacemaker neurons of Drosophila are electrically silenced their molecular oscillations are lost under constant conditions, suggesting that electrical activity at the cell membrane is required for the endogenous molecular clockwork (Nitabach et al., 2002). Approximately 150 neurons express the dynamic molecular clockwork in the adult brain and are divided into six groups based on location and size (Ewer et al., 1992; Frisch et al., 1994; Kaneko and Hall, 2000; Helfrich-Forster, 2003) (Fig. 1A). These are the large and small ventrolateral neurons (the l-LNvs and s-LNvs, respectively), the dorsolateral neurons (LNds), and three groups of dorsal neurons (DNs), the DN1s, DN2s, and DN3s. All six neuronal classes have been recognized for more than a decade (Ewer et al., 1992; Frisch et al., 1994) and no new classes of clock-neurons (i.e., neurons that support clock-gene oscillations under light:dark cycles or constant conditions) have been described in the central brain since that time (Hall, 2005). Hundreds more glial cells express clock products throughout the brain (Ewer et al., 1992), although the details of the molecular oscillations within glia and their role in directing behavioral rhythms is currently undefined. Fig. 1 R32-mediated LacZ expression in the adult brain. A: A projected Z-series montage of an adult R32 brain labeled for LacZ and imaged through its posterior surface. Cell classes are labeled in the left hemisphere. The dashed line represents the midline and ... The extent to which locomotor rhythms require specific neuronal classes has been investigated by the targeted deletion of clock neuron classes or the specific rescue of clock function within them (reviewed by Hall, 2005; Helfrich-Forster, 2005). The LNv and LNd are necessary and sufficient for the maintenance of locomotor rhythms in the absence of environmental times cues (Frisch et al., 1994; Helfrich-Forster, 1998; Renn et al., 1999) and for normal crepuscular organization of locomotion under light:dark (LD) conditions (Renn et al., 1999). Recent mosaic analysis suggests that the morning peak of activity is controlled by the s-LNv, while the evening peak is governed by a group of neurons that include the LNd and a subset of the DN1s (Stoleru et al., 2004; Grima et al., 2004). The specific functions of the l-LNvs are not known. DNs are not required for locomotor rhythms under constant darkness (DD) and temperature, but contribute to the organization and/or entrainment of such behavior under LD conditions (Veleri et al., 2003; Klarsfeld et al., 2004). Notwithstanding the ability to assign functions to most classes of clock neurons, the physiological and synaptic basis of these functions is poorly understood. For example, only one neurochemical output has been identified for clock neurons, a neuropeptide called pigment dispersing factor (PDF). PDF is expressed in both the large and small LNvs and is required for the maintenance of locomotor rhythms under constant conditions and the normal timing of locomotion under LD (Renn et al., 1999). The transmitter phenotypes of the remaining clock-neuron classes remain a mystery, as do the synaptic interactions that organize neuronal timekeeping. To provide ongoing studies of circadian pacemaker networks with greater cellular resolution, we reanalyzed the identities, anatomical position, and cellular properties of the clock neurons in the fly. In the present study we employ the detection of a local enhancer by a LacZ insertion called P{wF6-84,R32} (“R32”; Schneider et al., 1993) to describe the clock-neuron classes of Drosophila's central brain. First, we show that R32 is a limited and highly accurate reporter for neuronal PER in the adult brain. Next we present evidence based on R32 expression for a previously unappreciated class of pacemakers that we have called lateral posterior neurons (LPNs), based on their earlier description as Timeless (TIM)-expressing, nonpacemakers by Kaneko and Hall (2000). We define developmental, cellular, and anatomical criteria by which identified neuronal subclasses can be recognized within the LNd, DN1, DN2, and DN3 classes. Finally, we identify a second candidate neuropeptide transmitter within the Drosophila pacemaker network, IPNamide (IPNa), a product of the gene neuropeptide-like-precursor 1 (nplp1; Baggerman et al., 2002; Verleyen et al., 2004).

Journal ArticleDOI
TL;DR: The data indicate that the synaptology of basolateral amygdalar pyramidal cells is remarkably similar to that of cortical pyramsidal cells and that PV+ interneurons provide a robust inhibition of both the perisomatic and the distal dendritic domains of these principal neurons.
Abstract: The generation of emotional responses by the basolateral amygdala is determined largely by the balance of excitatory and inhibitory inputs to its principal neurons, the pyramidal cells. The activity of these neurons is tightly controlled by gamma-aminobutyric acid (GABA)-ergic interneurons, especially a parvalbumin-positive (PV(+)) subpopulation that constitutes almost half of all interneurons in the basolateral amygdala. In the present semiquantitative investigation, we studied the incidence of synaptic inputs of PV(+) axon terminals onto pyramidal neurons in the rat basolateral nucleus (BLa). Pyramidal cells were identified by using calcium/calmodulin-dependent protein kinase II (CaMK) immunoreactivity as a marker. To appreciate the relative abundance of PV(+) inputs compared with excitatory inputs and other non-PV(+) inhibitory inputs, we also analyzed the proportions of asymmetrical (presumed excitatory) synapses and symmetrical (presumed inhibitory) synapses formed by unlabeled axon terminals targeting pyramidal neurons. The results indicate that the perisomatic region of pyramidal cells is innervated almost entirely by symmetrical synapses, whereas the density of asymmetrical synapses increases as one proceeds from thicker proximal dendritic shafts to thinner distal dendritic shafts. The great majority of synapses with dendritic spines are asymmetrical. PV(+) axon terminals form mainly symmetrical synapses. These PV(+) synapses constitute slightly more than half of the symmetrical synapses formed with each postsynaptic compartment of BLa pyramidal cells. These data indicate that the synaptology of basolateral amygdalar pyramidal cells is remarkably similar to that of cortical pyramidal cells and that PV(+) interneurons provide a robust inhibition of both the perisomatic and the distal dendritic domains of these principal neurons.

Journal ArticleDOI
TL;DR: It is demonstrated that Nurr1 deficiency in dopaminergic neurons is associated with the intracellular pathology in both synucleinopathies and tauopathies.
Abstract: In mammals, the transcription factor Nurr1 is expressed early in development and continues to be detectable throughout the organism's lifetime. Nurr1 is involved in the establishment and maintenance of the dopaminergic phenotype within specific central nervous system neuronal subpopulations including the nigrostriatal dopamine system. This protein is reduced over the course of normal aging, which is a major risk factor for Parkinson's disease (PD). However, whether Nurr1 expression is affected by PD has not been documented. The present study examined the role of Nurr1 in the maintenance of the dopaminergic phenotype within neurons in substantia nigra in PD compared with patients with diagnoses of progressive supranuclear palsy (PSP) or Alzheimer's disease (AD) or age-matched-matched controls. In PD, the optical density (OD) of Nurr1 immunofluorescence was significantly decreased in nigral neurons containing alpha-synuclein-immunoreactive inclusions. Similarly, the OD of Nurr1 immunofluorescence intensity in the nigra of AD cases was decreased in neurons with neurofibrillary tangles (NFTs). In contrast to PD and AD, the OD of Nurr1 immunofluorescence intensity was severely decreased in the neurons with or without NFTs in PSP cases. Decline of Nurr1-ir neuronal number and OD was observed within substantia nigra (SN) neurons in PD but not within hippocampal neurons. The decline in Nurr1-ir expression was correlated with loss of tyrosine hydroxylase immunofluorescence across the four groups. These data demonstrate that Nurr1 deficiency in dopaminergic neurons is associated with the intracellular pathology in both synucleinopathies and tauopathies.

Journal ArticleDOI
TL;DR: The efferent association fibers from the caudal part of the prefrontal cortex to posterior cortical areas course via several pathways: the three components of the superior longitudinal fasciculus (SLF I, SLF II, and SLF III), the arcuate fascicule (AF), the fronto‐occipitals (FOF), the cingulate fasciculi (CING F), and the extreme capsule (Extm C).
Abstract: The efferent association fibers from the caudal part of the prefrontal cortex to posterior cortical areas course via several pathways: the three components of the superior longitudinal fasciculus (SLF I, SLF II, and SLF III), the arcuate fasciculus (AF), the fronto-occipital fasciculus (FOF), the cingulate fasciculus (CING F), and the extreme capsule (Extm C). Fibers from area 8Av course via FOF and SLF II, merging in the white matter of the inferior parietal lobule (IPL) and terminating in the caudal intraparietal sulcus (IPS). A group of these fibers turns ventrally to terminate in the caudal superior temporal sulcus (STS). Fibers from the rostral part of area 8Ad course via FOF and SLF II to the IPS and IPL and via the AF to the caudal superior temporal gyrus and STS. Some fibers from the rostral part of area 8Ad are conveyed to the medial parieto-occipital region via FOF, to the STS via Extm C, and to the caudal cingulate gyrus via CING F. Fibers from area 8B travel via SLF I to the supplementary motor area and area 31 in the caudal dorsal cingulate region and via the CING F to cingulate areas 24 and 23 and the cingulate motor areas. Fibers from area 9/46d course via SLF I to the superior parietal lobule and medial parieto-occipital region, via SLF II to the IPL. Fibers from area 9/46v travel via SLF III to the rostral IPL and the frontoparietal opercular region and via the CING F to the cingulate gyrus.

Journal ArticleDOI
TL;DR: Data show an important species difference in the chemical distinction of inhibitory neuron subtypes, and indicate that colocalization of CR in SST cells correlates with different morphological and physiological features.
Abstract: Mammalian cortex contains a diversity of inhibitory neuron types, each with distinct morphological, immunochemical, and/or physiological properties. In rat cortex, chemical markers distinguish at least four distinct and nonoverlapping neuron classes based on expression of parvalbumin (PV), somatostatin (SST), calretinin (CR), and cholecystokinin (CCK). It has generally been assumed that these classifications should also apply to other rodent species. In mouse cortex, however, we found significant colocalization of SST and CR in inhibitory neurons; about 30% of SST-positive cells contained CR, and about 33% of CR-positive cells contained SST across frontal, somatosensory (S1), and visual cortex (V1). The SST and CR colocalized cells were concentrated in layer 2/3. We further characterized morphological and physiological properties of the mouse cortical inhibitory neuron types that express SST by using "GIN" transgenic mice, in which GFP is expressed in a subset of SST inhibitory neurons (see Oliva et al. [2000] J Neurosci 20:3354-3368). Generally, both SST/CR+ cells and SST/CR- cells exhibited morphological features of Martinotti cells as described in rat cortex, and they also had similar accommodating spike-firing patterns. However, they differed significantly in quantitative comparisons of morphology and spike shapes. SST/CR+ cells had more horizontally extended dendritic fields and more primary process than did SST/CR- cells; and SST/CR- cells had narrower action potential widths and faster afterhyperpolarization than did SST/CR+ cells. Thus, our data show an important species difference in the chemical distinction of inhibitory neuron subtypes, and indicate that colocalization of CR in SST cells correlates with different morphological and physiological features.

Journal ArticleDOI
TL;DR: It is hypothesized that a transient overexpression in activated microglial density occurs normally in the cerebral white matter of the human fetus during the peak window of vulnerability for PVL, suggesting a potential “priming” of this area for diverse brain insults characterized by activation of microglia, particularly PVL.
Abstract: Although microglial activation may be an initial beneficial response to a variety of insults, prolonged activation can release toxic substances and lead to cell death. Microglial activation secondary to hypoxia-ischemia and/or infection in immature cerebral white matter is important in the pathogenesis of periventricular leukomalacia (PVL), the major pathological substrate of cerebral palsy in the premature infant. We hypothesize that a transient overexpression in activated microglial density occurs normally in the cerebral white matter of the human fetus during the peak window of vulnerability for PVL. Such an increase could render this region susceptible to insults that cause prolonged microglial activation, as conceptualized in PVL. To examine the developmental profile of microglia in the human fetus and infant brain, immunocytochemistry with microglial specific markers were used in 23 control (non-PVL) cases ranging from 20 to 183 postconceptional (PC) weeks. Tomato lectin, used to identify microglial morphology, revealed that the cerebral white matter of the human fetus and infant is densely populated with intermediate and amoeboid microglia; the latter is indicative of an activated state. Quantitative analysis with CD68 showed increased density of activated microglia in the cerebral white matter of the fetus (<37 PC weeks) relative to the neonate/infant (≥37 PC weeks) and to the overlying cortex of either age group (P = 0.01). The primary finding of a transient, developmental-dependent overabundance of CD68-activated microglia in the cerebral white matter of the fetus suggests a potential “priming” of this area for diverse brain insults characterized by activation of microglia, particularly PVL. J. Comp. Neurol. 497:199–208, 2006. © 2006 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: It is shown that, in addition to the extrasynaptic pool, there is a pool of α5‐GABAARs that concentrates at the GABAergic synapses in dendrites of hippocampal pyramidal cells that might play a role in the phasic GABAergic inhibition of Pyramidal neurons in hippocampus and cerebral cortex.
Abstract: The alpha5 subunit of the GABA(A) receptors (GABA(A)Rs) has a restricted expression in the brain. Maximum expression of this subunit occurs in the hippocampus, cerebral cortex, and olfactory bulb. Hippocampal pyramidal cells show high expression of alpha5 subunit-containing GABA(A)Rs (alpha5-GABA(A)Rs) both in culture and in the intact brain. A large pool of alpha5-GABA(A)Rs is extrasynaptic and it has been proposed to be involved in the tonic GABAergic inhibition of the hippocampus. Nevertheless, there are no studies on the localization of the alpha5-GABA(A)Rs at the electron microscope (EM) level. By using both immunofluorescence of cultured hippocampal pyramidal cells and EM postembedding immunogold of the intact hippocampus we show that, in addition to the extrasynaptic pool, there is a pool of alpha5-GABA(A)Rs that concentrates at the GABAergic synapses in dendrites of hippocampal pyramidal cells. The results suggest that the synaptic alpha5-GABA(A)Rs might play a role in the phasic GABAergic inhibition of pyramidal neurons in hippocampus and cerebral cortex.