Article

Cholinergic And Non-Cholinergic Afferents Of The Caudolateral Parabrachial Nucleus: A Role In The Long-Term Enhancement Of Rapid Eye Movement Sleep

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

A single microinjection of the cholinergic agonist carbachol into the feline caudolateral parabrachial nucleus produces an immediate increase in state-independent ipsilateral ponto-geniculooccipital waves, followed by a long-term rapid eye movement sleep enhancement lasting 7-10 days. Using retrogradely-transported fluorescent carbachol-conjugated nanospheres and choline acetyltransferase immunohistochemistry, afferent projections to this injection site for long-term rapid eye movement sleep enhancement were mapped and quantified. Six regions in the brain stem contained retrogradely-labelled cells: the raphe nuclei, locus coeruleus, laterodorsal tegmental nucleus, pedunculopontine tegmental nucleus, parabrachial nucleus, and the pontine reticular formation. The retrogradely-labelled (rhodamine+) cells in the pontine reticular formation and pedunculopontine tegmental nucleus contributed the predominant input to the parabrachial nucleus injection site (34.3 +/- 5.3% and 28.4 +/- 5.6%, respectively), compared to the laterodorsal tegmental nucleus (5.8 +/- 3.8%), parabrachial nucleus (13.5 +/- 3.1%), raphe nuclei (12.9 +/- 2.7%), and locus coeruleus (5.1 +/- 2.4%). By comparison with findings of afferent input to the induction site for short-latency rapid eye movement sleep in the anterodorsal pontine reticular formation, the parabrachial nucleus injection site is characterized by a similar proportion of afferents, except that the raphe nuclei were found to provide more than a two-fold greater input. Retrogradely-labelled neurons quantified in these nuclear regions consisted of 21.5% double-labelled (rhodamine+/choline acetyltransferase+) cholinergic and 78.5% noncholinergic (rhodamine+/choline acetyltransferase-) cells. The pedunculopontine tegmental nucleus contributed the predominant (51.7 +/- 8.2%) cholinergic input, compared to laterodorsal tegmental nucleus (20.7 +/- 10.2%), parabrachial nucleus (23.1 +/- 7.5%), and pontine reticular formation (4.4 +/- 2.1%). A comparative analysis of the total retrogradely-labelled cells within each nuclear region which were also double-labelled showed the highest proportion in the laterodorsal tegmental nucleus (76.2 +/- 7.5%) compared to pedunculopontine tegmental nucleus (39.4 +/- 3.6%), parabrachial nucleus (37.3 +/- 2.8%), and pontine reticular formation (3.2 +/- 2.1%). These data indicate that while pedunculopontine tegmental nucleus and laterodorsal tegmental nucleus neurons exert a powerful cholinergic influence on the injection site for long-term rapid eye movement enhancement, a major component of the afferent circuitry is non-cholinergic. Since the non-cholinergic input includes contributions from the locus coeruleus and raphe nuclei, it is probable that the caudolateral parabrachial nucleus contains cholinergic and aminergic afferent systems that participate in the long-term enhancement of rapid eye movement sleep.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... The main ascending inputs to the lateral parabrachial nucleus come from the spinal cord and NTS (Palmiter, 2018). Other inputs come from the pre-Bötzinger and Bötzinger complexes (Tan et al., 2010;Yang and Feldman, 2018), Kölliker-Fuse nucleus (Song et al., 2012a;Geerling et al., 2017), rVRG (Holstege, 1989), NTS (Beckstead et al., 1980;Herbert et al., 1990;McGovern et al., 2015b;Kawai, 2018;Yu et al., 2022), retrotrapezoid nucleus (Rosin et al., 2006;Bochorishvili et al., 2012;Silva et al., 2016a), locus coeruleus Yang et al., 2021), dorsal raphe (Petrov et al., 1992;Quattrochi et al., 1998;Kaur et al., 2020), bed nucleus of stria terminalis , central amygdala Yang et al., 2021), paraventricular nucleus (Zheng et al., 1995;Geerling et al., 2010;Singh et al., 2022), spinal trigeminal nucleus (Panneton et al., 2006;Zhang et al., 2018), paratrigeminal nucleus (Saxon and Hopkins, 1998;Caous et al., 2001;McGovern et al., 2015b;Driessen et al., 2018), PPTg (Quattrochi et al., 1998;Lima et al., 2019b), and fastigial nucleus (Teune et al., 2000). ...
... The main ascending inputs to the lateral parabrachial nucleus come from the spinal cord and NTS (Palmiter, 2018). Other inputs come from the pre-Bötzinger and Bötzinger complexes (Tan et al., 2010;Yang and Feldman, 2018), Kölliker-Fuse nucleus (Song et al., 2012a;Geerling et al., 2017), rVRG (Holstege, 1989), NTS (Beckstead et al., 1980;Herbert et al., 1990;McGovern et al., 2015b;Kawai, 2018;Yu et al., 2022), retrotrapezoid nucleus (Rosin et al., 2006;Bochorishvili et al., 2012;Silva et al., 2016a), locus coeruleus Yang et al., 2021), dorsal raphe (Petrov et al., 1992;Quattrochi et al., 1998;Kaur et al., 2020), bed nucleus of stria terminalis , central amygdala Yang et al., 2021), paraventricular nucleus (Zheng et al., 1995;Geerling et al., 2010;Singh et al., 2022), spinal trigeminal nucleus (Panneton et al., 2006;Zhang et al., 2018), paratrigeminal nucleus (Saxon and Hopkins, 1998;Caous et al., 2001;McGovern et al., 2015b;Driessen et al., 2018), PPTg (Quattrochi et al., 1998;Lima et al., 2019b), and fastigial nucleus (Teune et al., 2000). ...
... The target areas of the PPTg are not restricted to those mentioned above, but also include the Kölliker-Fuse nucleus (Lima et al., 2019b), lateral parafacial nucleus (Biancardi et al., 2021), hypoglossal, trigeminal and facial motor nuclei (Woolf and Butcher, 1989), locus coeruleus (Woolf and Butcher, 1989), dorsal and caudal raphe (Woolf and Butcher, 1989;Hermann et al., 1997;Ogawa et al., 2014;Lima et al., 2019b), lateral hypothalamus (Woolf and Butcher, 1986), bed nucleus of the stria terminalis (Ni et al., 2016), central amygdala (Dautan et al., 2016), lateral parabrachial nucleus (Quattrochi et al., 1998;Lima et al., 2019b), spinal trigeminal nucleus (Woolf and Butcher, 1989), and all three cerebellar nuclei (Woolf and Butcher, 1989;Ruggiero et al., 1997;Vitale et al., 2016). ...
Article
Full-text available
Respiration is a brain function on which our lives essentially depend. Control of respiration ensures that the frequency and depth of breathing adapt continuously to metabolic needs. In addition, the respiratory control network of the brain has to organize muscular synergies that integrate ventilation with posture and body movement. Finally, respiration is coupled to cardiovascular function and emotion. Here, we argue that the brain can handle this all by integrating a brainstem central pattern generator circuit in a larger network that also comprises the cerebellum. Although currently not generally recognized as a respiratory control center, the cerebellum is well known for its coordinating and modulating role in motor behavior, as well as for its role in the autonomic nervous system. In this review, we discuss the role of brain regions involved in the control of respiration, and their anatomical and functional interactions. We discuss how sensory feedback can result in adaptation of respiration, and how these mechanisms can be compromised by various neurological and psychological disorders. Finally, we demonstrate how the respiratory pattern generators are part of a larger and integrated network of respiratory brain regions.
... Furthermore, it should not be possible to dissociate phasic activity is a distinguishing component of REM sleep in PGO activity from REM. the feline and has been regarded by many as a REM sleep We now report that at sites within the caudolateral unitary event propagated throughout the brain (Malcolm et al. peribrachial area (C-PB), just 0.4 mm anterior to those 1970; Hobson and Steriade 1986; Callaway et al. 1987). Because triggering long-term REM enhancement (LTRE) (Datta et al. it occurs so early in the transition from slow wave sleep (SWS) 1991, 1992; Calvo et al. 1992; Quattrochi et al. 1998), a to REM (defined as SWS with PGO activity, or SP), it has single microinjection of carbachol produces a long-term PGO been thought to be necessary to trigger REM sleep. enhancement (LTPE) lasting 5 days, but without REM sleep As PGO activity begins antecedent to the onset of REM enhancement. ...
... Seven to 10 days after surgery, cats were adapted and fluorescent labeled boundary contours of the carbachol to unrestrained freely moving recording conditions before nanosphere injection sites were plotted using a DEC computer microinjection trials with carbachol-conjugated rhodamine imaging system and software (Digital Equipment Corp., USA). nanospheres (Quattrochi et al. 1989Quattrochi et al. , 1998; Quattrochi and Hobson 1990) were begun. RESULTS In order to establish baseline control values, polygraphic recordings were performed during three consecutive 4-h We performed a quantitative analysis of PGO waves recorded sessions between 12.00 and 16.00 h for each cat. ...
... Nanospheres are noncytotoxic and the rhodamine label following cholinoceptive activation at the C-PB site described in remains with no transneuronal diffusion. Neither is there this study of LTPE, carbachol at this site gives no such LTRE, evidence of local dendritic uptake, or uptake from unbroken as was seen with carbachol microinjected at a site 0.4 mm more axons of passage (Quattrochi et al. 1998). Each of our injection posterior. ...
Article
This study presents new findings of carbachol-induced long-term ponto-geniculo-occipital (PGO) enhancement lasting five days, but without REM sleep enhancement. A quantitative analysis of the number and types of bilateral PGO wave events during slow wave sleep with PGO activity (SP) and REM was performed in each of four cats over a period of six days following a single unilateral microinjection of carbachol nanospheres into the caudolateral peribrachial area. The results demonstrate increases in the summed total of all PGO wave events to continue for five days postcarbachol reaching a peak sixfold increase on day three in SP and REM. The tendency of PGO waves to occur in clusters of greater than three waves increased sevenfold on day three in SP and fourfold during REM. These findings indicate a dissociation of long-term PGO enhancement from long-term REM enhancement, and suggest that even a sixfold increase in PGO activity alone is not, in itself, sufficient to produce the cholinergic orchestration of REM sleep enhancement.
... In contrast, a single micro-injection of carbachol into the caudolateral PMT elicits a very different effect of REM sleep enhancement within normal sleep time lasting 6-10 days [18]. Using microinjections of a carbachol-conjugated latex nanosphere delivery system (LNDS) [46,67] that limits the diffusion of drug at these pontomesencephalic sites, our neuroanatomical studies identified cholinergic and non-cholinergic afferent neurons within the PMT associated with this long-term REM enhancement [68]. ...
... All electroencephalographic (EEG) leads were connected to an Amphenol 33-pin connector plug that was attached to the vertex of the skull with dental acrylic cement. Bilateral stainless steel guide cannulae (26 gauge) were stereotaxically implanted and aimed 5 mm above the injection sites in the caudolateral PMT (P3.6, L4.5, H-2.5) according to Berman [4] and as described previously [68]. A jugular intravenous (iv) catheter was inserted for later anesthesia and perfusion. ...
... Microinjection procedures consisting of vehicle (unconjugated nanospheres, 0.9% saline) and carbachol-conjugated nanospheres have been detailed previously [67,68]. All microinjections were performed with a 1-μl Hamilton syringe attached to a 31-gauge stainless steel injector cannula 5 mm longer than the implanted guide cannulae and with a tip diameter of 40 μm. ...
Article
Full-text available
It is not known how the brain modifies its regulatory systems in response to the application of a drug, especially over the long term of weeks and months. We have developed a model system approach to this question by manipulating cholinergic cell groups of the laterodorsal and pedunculopontine tegmental (LDT/PPT) nuclei in the pontomesencephalic tegmentum (PMT), which are known to be actively involved in the timing and quantity of rapid eye movement (REM) sleep. In a freely moving feline model, a single microinjection of the cholinergic agonist carbachol conjugated to a latex nanosphere delivery system into the caudolateral PMT elicits a long-term enhancement of one distinguishing phasic event of REM sleep, ponto-geniculo-occipital (PGO) waves, lasting 5 days but without any significant change in REM sleep or other behavioral state. Here, we test the hypothesis that cholinergic activation within the caudolateral PMT alters the postsynaptic excitability of the PGO network, stimulating the prolonged expression of c-fos that underlies this long-term PGO enhancement (LTPE) effect. Using quantitative Fos immunohistochemistry, we found that the number of Fos-immunoreactive (Fos-IR) neurons surrounding the caudolateral PMT injection site decreased sharply by postcarbachol day 03, while the number of Fos-IR neurons in the more rostral LDT/PPT increased >30-fold and remained at a high level following the course of LTPE. These results demonstrate a sustained c-fos expression in response to pharmacological stimulation of the brain and suggest that carbachol's acute effects induce LTPE via cholinergic receptors, with subsequent transsynaptic activation of the LDT/PPT maintaining the LTPE effect.
... Possibly, in wakefulness states or during REM sleep, the stimulation of PPTg produces the activation of a circuit involving inhibitory neurons, producing a reduction of respiratory activity. PPTg neurons have ipsilateral and contralateral projections to the parabrachial nucleus and this network can certainly modulate and increase REM sleep (Quattrochi et al., 1998). Our results are in agreement with the literature, since PPTg neurons seem to activate the pontine respiratory centers such as the Kölliker-Fuse and Parabrachial Complex that are involved in expiratory activity (Saponjic et al., 2006). ...
... In addition to the two forebrain hotspots of NAc and VP, there is also some evidence for a brainstem hedonic hotspot within the parabrachial nucleus (PBN) of the pons [108]. Although best known as a visceral/ taste sensory relay [38,79], the PBN has additional functions, including food intake [28,40,124,127], establishing a conditioned taste aversion [28,36,130], and REM sleep [90,120]. ...
Chapter
Food is one of the intense pleasures in life. Food “liking” and “wanting” systems interact with regulatory mechanisms of hunger and satiety to control eating. So, what brain systems produce taste pleasure or “liking”? And what brain systems convert pleasure into “wanting” to eat? Neuroscientists have begun to answer these questions combining information from brain manipulation experiments in laboratory animals together with neuroimaging experiments in humans. In this chapter we review recent progress in (1) the identification of brain reward substrates of food pleasure and desire and how food becomes “liked” and “wanted”; (2) how “wanting” and “liking” systems interact with hunger and satiety mechanisms to influence eating; and (3) how dysfunction of “wanting” and “liking” systems might lead to overeating.
... Given that the NTS does not directly innervate cortical areas, chronic VNS-induced EEG changes are likely mediated by modulation of pathways indirectly involved in the genesis of cortical rhythms. Indeed, the parabrachial nucleus, which receives NTS efferents (Quattrocchi et al., 1998) projects to several thalamic nuclei that contribute to EEG activity and receives an important input from the locus coeruleus (LC). In addition, the integrity of the LC is important for the antiepileptic, desynchronizing, and arousal-promoting effects of VNS (Krahl et al., 1998). ...
Article
Objective: Though vagus nerve stimulation (VNS) is an important option in pharmacoresistant epilepsy, its mechanism of action remains unclear. The observation that VNS desynchronised the EEG activity in animals suggested that this mechanism could be involved in VNS antiepileptic effects in humans. Indeed VNS decreases spiking bursts, whereas its effects on the EEG background remain uncertain. The objective of the present study is to investigate how VNS affects local and inter regional syncronization in different frequencies in pharmacoresistent partial epilepsy. Methods: Digital recordings acquired in 11 epileptic subjects 1 year and 1 week before VNS surgery were compared with that obtained 1 month and 1 year after VNS activation. Power spectrum and synchronization were then analyzed and compared with an epileptic group of 10 patients treated with AEDs only. Results: VNS decreases the synchronization of theta frequencies (P!0.01), whereas it increases gamma power spectrum and synchronization (!0.001 and 0.01, respectively). Conclusions: The reduction of theta frequencies and the increase in power spectrum and synchronization of gamma bands can be related to VNS anticonvulsant mechanism. In addition, gamma modulation could also play a seizure-independent role in improving attentional performances. Significance: These results suggest that some antiepileptic mechanisms affected by VNS can be modulated by or be the reflection of EEG changes.
... Therefore, it is likely that GLUT-induced behavioral changes as observed in this study were predominantly caused by the activation of neurons in the immediate vicinity of the injection site, the PeF, where GLUT was possibly maximally effective and where clusters of orexin neurons have been reported [22]. However, based on earlier reports from us [17] and others [23], the injected GLUT possibly diffused to ∼800-900 M diameter and therefore, it is plausible that the activation of neurons in the extended diffusion field, i.e., the neurons located within the boundaries of adjacent dorsal, medial, ventral and lateral hypothalamic areas, but to a relatively lesser extent, contributed to GLUT-induced behavioral effects as well. The above interpretations are based on the observations that in this study PeF has consistently been the core of the injection sites and it is an unlikely possibility that in all experiments GLUT had diffused uniformly and simultaneously to both extremes from medial to lateral and ventral to dorsal areas, where neuronal activation does not produce uniform and identical effects (rather often they induce opposite effects) on sleep-waking [18]. ...
... In addition to the two forebrain hotspots of NAc and VP, there is also some evidence for a brainstem hedonic hotspot within the parabrachial nucleus (PBN) of the pons (Soderpalm and Berridge, 2000a). Although best known as a visceral/taste sensory relay (Norgren and Leonard, 1971;Di Lorenzo and Monroe, 1997), the PBN has additional functions, including food intake (Wilson et al., 2003;DiPatrizio and Simansky, 2008;Wu et al., 2009;Carter et al., 2013b), establishing a conditioned taste aversion (Yamamoto, 2007;Carter et al., 2013b;Dayawansa et al., 2013), and REM sleep (Quattrochi et al., 1998;Torterolo et al., 2011). ...
... This peculiar type of cognition was originally attributed to the loss of noradrenergic modulation of the cortex (Hartmann, 1973) or the spontaneous activity of the brain as a closed system in absence of environmental inputs and voluntary self-regulation mechanisms (Rechtschaffen, 1978;Antrobus, 1991). More recently, it was correlated to the ponto-geniculo-occipital waves that have been recorded when cholinergic hyperactivity triggers REM sleep in the absence of aminergic inhibition (Quattrochi et al. 1998). In terms of functional activation in this stage of sleep, where bizarreness has been found to be highest, recent findings suggest a relationship between dream bizarreness and the decrease in temporal coupling between executive and perceptual regions of the cortex (Corsi-Cabrera et al., 2003). ...
Article
Cognitive bizarreness has been shown to be equally elevated in the dream and waking mentation of acutely symptomatic inpatients diagnosed with affective and non-affective psychoses. Although some studies have reported on dream content in non-psychotic depression, no study has previously measured this formal aspect of cognition in patients hospitalized for Psychotic Major Depression (PMD). 65 dreams and 154 waking fantasy reports were collected from 11 PMD inpatients and 11 age- and sex-matched healthy controls. All narrative reports were scored by judges blind to diagnosis in terms of formal aspects of cognition (Bizarreness). Dream content was also scored (Hall/Van de Castle scoring system). Unlike controls, PMD patients had similar levels of cognitive bizarreness in their dream and waking mentation. Dreams of PMD patients also differed from those of controls in terms of content variables. In particular, Happiness, Apprehension and Dynamism were found to differ between the two groups. Whereas dream content reflects a sharp discontinuity with the depressive state, cognitive bizarreness adequately measures the stability of cognition across dreams and wakefulness in PMD inpatients.
... The parabrachial region surrounding the brachium conjunctivum is divided into 13 distinct subnuclei in the rat, each associated with a unique set of afferents, efferents and neurotransmitters (Saper, 1995). The area where we observed Fos immunoreactivity during NREM is identified as the CLPB (Datta, 1997;Quattrochi et al.). This area includes part of the marginal nucleus of the brachium conjunctivum as well as the dorsal region of the Kolliker Fuse nucleus (Berman, 1968). ...
Article
Full-text available
The principal site that generates both rapid eye movement (REM) sleep and wakefulness is located in the mesopontine reticular formation, whereas non-rapid eye movement (NREM) sleep is primarily dependent upon the functioning of neurons that are located in the preoptic region of the hypothalamus. In the present study, we were interested in determining whether the occurrence of NREM might also depend on the activity of mesopontine structures, as has been shown for wakefulness and REM sleep. Adult cats were maintained in one of the following states: quiet wakefulness (QW), alert wakefulness (AW), NREM, or REM sleep induced by microinjections of carbachol into the nucleus pontis oralis (REM-carbachol). Subsequently, they were euthanized and single-labeling immunohistochemical studies were undertaken to determine state-dependent patterns of neuronal activity in the brainstem based upon the expression of the protein Fos. In addition, double-labeling immunohistochemical studies were carried out to detect neurons that expressed Fos as well as choline acetyltransferase, tyrosine hydroxylase, or GABA. During NREM, only a few Fos-immunoreactive cells were present in different regions of the brainstem; however, a discrete cluster of Fos+ neurons was observed in the caudolateral parabrachial region (CLPB). The number of Fos+ neurons in the CLPB during NREM was significantly greater (67.9±10.9, P<0.0001) compared with QW (8.0±6.7), AW (5.2±4.2), or REM-carbachol (8.0±4.7). In addition, there was a positive correlation (R=0.93) between the time the animals spent in NREM and the number of Fos+ neurons in the CLPB. Fos-immunoreactive neurons in the CLPB were neither cholinergic nor catecholaminergic; however, about 50% of these neurons were GABAergic. We conclude that a group of GABAergic and unidentified neurons in the CLPB are active during NREM and likely involved in the control of this behavioral state. These data open new avenues for the study of NREM, as well as for the explorations of interactions between these neurons that are activated during NREM and cells of the adjacent pontine tegmentum that are involved in the generation of REM sleep.
... Therefore, it is possible that the cardiovascular and respiratory responses we observed following PPT glutamate injections resulted from activation of monosynaptic projections from the PPT to the RVLM, although polysynaptic pathways cannot be ruled out. For example, the PPT reciprocally innervates the parabrachial complex (Gilbert and Lydic, 1991;Quattrochi et al. 1998;Semba and Fibiger, 1992) and neurons of the parabrachical complex together with the Kolliker-Fuse nucleus regulate respiratory phase switching. In addition, stimulation of the medial parabrachial or Kölliker-Fuse areas produces expiratory facilitation and apnea, whereas activation of the lateral parabrachial region can produce tachypnea (Chamberlin and Saper, 1994;Dick and Coles, 2000;Takayama and Miura, 1993). ...
Article
Functionally distinct areas were mapped within the pedunculopontine tegmentum (PPT) of 42 ketamine/xylazine anesthetized rats using local stimulation by glutamate microinjection (10 mM, 5-12 nl). Functional responses were classified as: (1) apnea; (2) tachypnea; (3) hypertension (HTN); (4) sinus tachycardia; (5) genioglossus electromyogram activation or (6) pontine-waves (p-waves) activation.We found that short latency apneas were predominantly elicited by stimulation in the lateral portion of the PPT, in close proximity to cholinergic neurons. Tachypneic responses were elicited from ventral regions of the PPT and HTN predominated in the ventral portion of the antero-medial PPT. We observed sinus tachycardia after stimulation of the most ventral part of the medial PPT at the boundary with nucleus reticularis pontis oralis, whereas p-waves were registered predominantly following stimulation in the dorso-caudal portion of the PPT. Genioglossus EMG activation was evoked from the medial PPT. Our results support the existence of the functionally distinct areas within the PPT affecting respiration, cardiovascular function, EEG and genioglossus EMG.
... There is a major projection of CRH pathways from the amygdala to pontine areas, 32,33 where REMon cells are dominant in the cholinergic system. 34 CRH is capable of activating these areas to induce muscle atonia that is one of the characteristics accompanying REM sleep. 35 When the amygdala receives either electric stimulation or cholinergic activation, prolonged REM sleep occurs. ...
Article
Full-text available
Impaired sleep and enhanced stress hormone secretion are the hallmarks of stress-related disorders, including major depression. The central neuropeptide, corticotropin-releasing hormone (CRH), is a key hormone that regulates humoral and behavioral adaptation to stress. Its prolonged hypersecretion is believed to play a key role in the development and course of depressive symptoms, and is associated with sleep impairment. To investigate the specific effects of central CRH overexpression on sleep, we used conditional mouse mutants that overexpress CRH in the entire central nervous system (CRH-COE-Nes) or only in the forebrain, including limbic structures (CRH-COE-Cam). Compared with wild-type or control mice during baseline, both homozygous CRH-COE-Nes and -Cam mice showed constantly increased rapid eye movement (REM) sleep, whereas slightly suppressed non-REM sleep was detected only in CRH-COE-Nes mice during the light period. In response to 6-h sleep deprivation, elevated levels of REM sleep also became evident in heterozygous CRH-COE-Nes and -Cam mice during recovery, which was reversed by treatment with a CRH receptor type 1 (CRHR1) antagonist in heterozygous and homozygous CRH-COE-Nes mice. The peripheral stress hormone levels were not elevated at baseline, and even after sleep deprivation they were indistinguishable across genotypes. As the stress axis was not altered, sleep changes, in particular enhanced REM sleep, occurring in these models are most likely induced by the forebrain CRH through the activation of CRHR1. CRH hypersecretion in the forebrain seems to drive REM sleep, supporting the notion that enhanced REM sleep may serve as biomarker for clinical conditions associated with enhanced CRH secretion.
... Given that the NTS does not directly innervate cortical areas, chronic VNS-induced EEG changes are likely mediated by modulation of pathways indirectly involved in the genesis of cortical rhythms. Indeed, the parabrachial nucleus, which receives NTS efferents ( Quattrocchi et al., 1998) projects to several thalamic nuclei that contribute to EEG activity and receives an important input from the locus coeruleus (LC). In addition, the integrity of the LC is important for the antiepileptic, desynchronizing, and arousal-promoting effects of VNS ( Krahl et al., 1998). ...
Article
Though vagus nerve stimulation (VNS) is an important option in pharmaco-resistant epilepsy, its mechanism of action remains unclear. The observation that VNS desynchronised the EEG activity in animals suggested that this mechanism could be involved in VNS antiepileptic effects in humans. Indeed VNS decreases spiking bursts, whereas its effects on the EEG background remain uncertain. The objective of the present study is to investigate how VNS affects local and inter regional syncronization in different frequencies in pharmaco-resistant partial epilepsy. Digital recordings acquired in 11 epileptic subjects 1 year and 1 week before VNS surgery were compared with that obtained 1 month and 1 year after VNS activation. Power spectrum and synchronization were then analyzed and compared with an epileptic group of 10 patients treated with AEDs only. VNS decreases the synchronization of theta frequencies (P < 0.01), whereas it increases gamma power spectrum and synchronization (< 0.001 and 0.01, respectively). The reduction of theta frequencies and the increase in power spectrum and synchronization of gamma bands can be related to VNS anticonvulsant mechanism. In addition, gamma modulation could also play a seizure-independent role in improving attentional performances. These results suggest that some antiepileptic mechanisms affected by VNS can be modulated by or be the reflection of EEG changes.
... Therefore, it is likely that GLUT-induced behavioral changes as observed in this study were predominantly caused by the activation of neurons in the immediate vicinity of the injection site, the PeF, where GLUT was possibly maximally effective and where clusters of orexin neurons have been reported [22]. However, based on earlier reports from us [17] and others [23], the injected GLUT possibly diffused to ∼800-900 M diameter and therefore, it is plausible that the activation of neurons in the extended diffusion field, i.e., the neurons located within the boundaries of adjacent dorsal, medial, ventral and lateral hypothalamic areas, but to a relatively lesser extent, contributed to GLUT-induced behavioral effects as well. The above interpretations are based on the observations that in this study PeF has consistently been the core of the injection sites and it is an unlikely possibility that in all experiments GLUT had diffused uniformly and simultaneously to both extremes from medial to lateral and ventral to dorsal areas, where neuronal activation does not produce uniform and identical effects (rather often they induce opposite effects) on sleep-waking [18]. ...
Article
The orexinergic neurons, localized in the perifornical hypothalamic area (PeF), are active during waking and quiet during non-rapid eye movement (non-REM) and REM sleep. Orexins promote arousal and suppress non-REM and REM sleep. Although in vitro studies suggest that PeF-orexinergic neurons are under glutamatergic influence, the sleep-wake behavioral consequences of glutamatergic activation of those neurons are not known. We examined the effects of bilateral glutamatergic activation of neurons in and around the PeF on sleep-wake parameters in freely behaving rats. Nine male Wistar rats were surgically prepared for electrophysiological sleep-wake recording and with bilateral guide cannulae targeting the PeF for microinjection. The sleep-wake profiles of each rat were recorded for 8h under baseline (without injection), and after bilateral microinjections of 200nl saline and 200nl saline containing 20 or 40ng of l-glutamic acid (GLUT) using a remote-controlled pump and without disturbing the animals. The injection of 40ng GLUT into the PeF (n=6) significantly increased mean time spent in waking (F=85.11, p<0.001) and concomitantly decreased mean time spent in non-REM (F=19.67, p<0.001) and REM sleep (F=38.72, p<0.001). The increase in waking and decreases in non-REM and REM sleep were due to significantly increased durations of waking episodes (F=24.64; p<0.001) and decreased durations of non-REM (F=12.96; p=0.002) and REM sleep events (F=13.82; p=0.001), respectively. These results suggest that the activation of neurons in and around the PeF including those of orexin neurons contribute to the promotion of arousal and suppression of non-REM and REM sleep.
Article
Nearly 1% of the global population suffers from epilepsy. Drug‐resistant epilepsy (DRE) affects one‐third of epileptic patients who are unable to treat their condition with existing drugs. For the treatment of DRE, neuromodulation offers a lot of potential. The background, mechanism, indication, application, efficacy, and safety of each technique are briefly described in this narrative review, with an emphasis on three approved neuromodulation therapies: vagus nerve stimulation (VNS), deep brain stimulation of the anterior nucleus of the thalamus (ANT‐DBS), and closed‐loop responsive neurostimulation (RNS). Neuromodulatory approaches involving direct or induced electrical currents have been developed to lessen seizure frequency and duration in patients with DRE since the notion of electrical stimulation as a therapy for neurologic diseases originated in the early nineteenth century. Although few people have attained total seizure independence for more than 12 months using these treatments, more than half have benefitted from a 50% drop in seizure frequency over time. Although promising outcomes in adults and children with DRE have been achieved, challenges such as heterogeneity among epilepsy types and etiologies, optimization of stimulation parameters, a lack of biomarkers to predict response to neuromodulation therapies, high‐level evidence to aid decision‐making, and direct comparisons between neuromodulatory approaches remain. To solve these existing gaps, authorize new kinds of neuromodulation, and develop personalized closed‐loop treatments, further research is needed. Finally, both invasive and non‐invasive neuromodulation seems to be safe. Implantation‐related adverse events for invasive stimulation primarily include infection and pain at the implant site. Intracranial hemorrhage is a frequent adverse event for DBS and RNS. Other stimulation‐specific side‐effects are mild with non‐invasive stimulation.
Article
The data outlined in this chapter provides evidence to support a concept that the activation of pontine-wave (P-wave) generating neurons plays a critical role in long-term memory formation. The P-wave, generated by the phasic activation of glutamatergic neurons in the pons, is one of the most prominent phasic events of REM sleep. These P-wave generating neurons project to the hippocampus, amygdala, entorhinal cortex and many other regions of the brain known to be involved in cognitive processing. These P-wave generating glutamatergic neurons remain silent during wakefulness and slow-wave sleep (SWS), but during the transition from SWS to REM sleep and throughout REM sleep these neurons discharge high-frequency spike bursts in the background of tonically increased firing rates. Activation of these P-wave generating neurons increases glutamate release and activates postsynaptic N-methyl-D-aspartic acid (NMDA) receptors in the dorsal hippocampus. Activation of P-wave generating neurons increases phosphorylation of transcription factor cAMP response element binding protein (CREB) in the dorsal hippocampus and amygdala by activating intracellular protein kinase A (PKA). The P-wave generating neurons activation-dependent PKA-CREB phosphorylation increases the expression of activity-regulated cytoskeletal-associated protein (Arc), brain-derived neurotrophic factor (BDNF), and early growth response-1 (Egr-1) genes in the dorsal hippocampus and amygdala. The P-wave generator activation-induced increased activation of PKA and expression of pCREB, Arc, BDNF, and Egr-1 in the dorsal hippocampus is shown to be necessary for REM sleep-dependent memory processing. Continued research on P-wave generation and its functions may provide new advances in understanding memory and treating its disorders.
Article
Rapid eye movement (REM) sleep shares many underlying mechanisms with wakefulness, to a much greater extent than does non-REM, especially those relating to feeding behaviours, appetite, curiosity, exploratory (locomotor) activities, as well as aspects of emotions, particularly 'fear extinction'. REM is most evident in infancy, thereafter declining in what seems to be a dispensable manner that largely reciprocates increasing wakefulness. However, human adults retain more REM than do other mammals, where for us it is most abundant during our usual final REM period (fREMP) of the night, nearing wakefulness. The case is made that our REM is unusual, and that (i) fREMP retains this 'dispensability', acting as a proxy for wakefulness, able to be forfeited (without REM rebound) and substituted by physical activity (locomotion) when pressures of wakefulness increase; (ii) REM's atonia (inhibited motor output) may be a proxy for this locomotion; (iii) our nocturnal sleep typically develops into a physiological fast, especially during fREMP, which is also an appetite suppressant; (iv) REM may have 'anti-obesity' properties, and that the loss of fREMP may well enhance appetite and contribute to weight gain ('overeating') in habitually short sleepers; (v) as we also select foods for their hedonic (emotional) values, REM may be integral to developing food preferences and dislikes; and (vii) REM seems to have wider influences in regulating energy balance in terms of exercise 'substitution' and energy (body heat) retention. Avenues for further research are proposed, linking REM with feeding behaviours, including eating disorders, and effects of REM-suppressant medications. Copyright © 2015 Elsevier B.V. All rights reserved.
Article
Brain activity explains the essential features of near-death experience, including the perceptions of envelopment by light, out-of-body, and meeting deceased loved ones or spiritual beings. To achieve their fullest expression, such near-death experiences require a confluence of events and draw upon more than a single physiological or biochemical system, or one anatomical structure. During impaired cerebral blood flow from syncope or cardiac arrest that commonly precedes near-death, the boundary between consciousness and unconsciousness is often indistinct and a person may enter a borderland and be far more aware than is appreciated by others. Consciousness can also come and go if blood flow rises and falls across a crucial threshold. During crisis the brain's prime biologic purpose to keep itself alive lies at the heart of many spiritual experiences and inextricably binds them to the primal brain. Brain ischemia can disrupt the physiological balance between conscious states by leading the brainstem to blend rapid eye movement (REM) and waking into another borderland of consciousness during near-death. Evidence converges from many points to support this notion, including the observation that the majority of people with a near-death experience possess brains predisposed to fusing REM and waking consciousness into an unfamiliar reality, and are as likely to have out-of-body experience while blending REM and waking consciousness as they are to have out-of-body experience during near-death.
Article
This study explored the efficacy and safety of transcutaneous vagus nerve stimulation (t-VNS) in patients with pharmacoresistant epilepsy. A total of 60 patients were randomly divided into two groups based on the stimulation zone: the Ramsay-Hunt zone (treatment group) and the earlobe (control group). Before and after the 12-month treatment period, all patients completed the Self-Rating Anxiety Scale (SAS), the Self-Rating Depression Scale (SDS), the Liverpool Seizure Severity Scale (LSSS), and the Quality of Life in Epilepsy Inventory (QOLIE-31). Seizure frequency was determined according to the patient's seizure diary. During our study, the antiepileptic drugs were maintained at a constant level in all subjects. After 12months, the monthly seizure frequency was lower in the treatment group than in the control group (8.0 to 4.0; P=0.003). This reduction in seizure frequency was correlated with seizure frequency at baseline and duration of epilepsy (both P>0.05). Additionally, all patients showed improved SAS, SDS, LSSS, and QOLIE-31 scores that were not correlated with a reduction in seizure frequency. The side effects in the treatment group were dizziness (1 case) and daytime drowsiness (3 cases), which could be relieved by reducing the stimulation intensity. In the control group, compared with baseline, there were no significant changes in seizure frequency (P=0.397), SAS, SDS, LESS, or QOLIE-31. There were also no complications in this group.
Article
Disorders of consciousness are due to failure of the arousal system. In this review, the authors introduce the spectrum of disorders of consciousness and describe the structures, projections, and neurotransmitters involved in the generation and maintenance of arousal. Next, they discuss the neurologic diseases frequently associated with arousal failure. Evaluation of patients with disorders of arousal is summarized, including the neurologic exam, electrophysiological studies, biochemical testing, and imaging modalities. Finally, they review treatment options, including therapeutic hypothermia, medications, and deep brain and spinal cord stimulation.
Article
A 15-year-old girl had no REM sleep presumably due to a pontine cavernous hemangioma was reported. Her brain MRI revealed a cavernous hemangioma extending from the dorsal pontine to the medulla. She manifested truncal ataxia, facial nerve palsy, and ocular motor apraxia. She could not sleep in the supine position due to the sleep apnea accompanied with loud snoring. Overnight polysomnography (PSG) was performed for detection of obstructive sleep apnea syndrome (OSAS). In addition to severe OSAS and Cheyne-Stokes-like respiration at wake after sleep onset, her 1st PSG study revealed no periods with rapid eye movement, EEG characteristic of REM sleep, atonia and variation on respiratory and heart rate. Even after effective therapy for OSAS with non-invasive positive airway pressure ventilation (NPPV), her 2nd PSG also failed to show stage REM. These findings suggest that this pontine cavernous hemangioma disturbed her REM-on system. This is the first report of an individual with long-term loss of REM sleep and a valuable case for the understanding of anatomical structures of the REM-on system and the role of REM in memory consolidation.
Article
Given that vagal afferents project to brainstem regions that promote alertness, the authors tested the hypothesis that vagus nerve stimulation (VNS) would improve daytime sleepiness in patients with epilepsy. Sixteen subjects with medically refractory seizures underwent polysomnography and multiple sleep latency tests (MSLT) and completed the Epworth Sleepiness Scale (ESS), a measure of subjective daytime sleepiness, before and after 3 months of VNS. Most subjects (>80%) were maintained on constant doses of antiepileptic medications. In the 15 subjects who completed baseline and treatment MSLT, the mean sleep latency (MSL) improved from 6.4 +/- 4.1 minutes to 9.8 +/- 5.8 minutes (+/- SD; p = 0.033), indicating reduced daytime sleepiness. All subjects with stimulus intensities of < or =1.5 mA showed improved MSL. In the 16 subjects who completed baseline and treatment ESS, the mean ESS score decreased from 7.2 +/- 4.4 to 5.6 +/- 4.5 points (p = 0.049). Improvements in MSLT and ESS were not correlated with reduction in seizure frequency. Sleep-onset REM periods occurred more frequently in treatment naps as compared to baseline naps (p < 0.008; Cochran-Mantel-Haenszel test). The amount of REM sleep or other sleep stages recorded on overnight polysomnography did not change with VNS treatment. Treatment with VNS at low stimulus intensities improves daytime sleepiness, even in subjects without reductions in seizure frequency. Daytime REM sleep is enhanced with VNS. These findings support the role of VNS in activating cholinergic and other brain regions that promote alertness.
Article
Full-text available
Sleep and wakefulness are instinctive behaviours that are present across the animal species. Rapid eye movement (REM) sleep is a unique biological phenomenon expressed during sleep. It evolved about 300 million years ago and is noticed in the more evolved animal species. Although it has been objectively identified in its present characteristic form about half a century ago, the mechanics of how REM is generated, and what happens upon its loss are not known. Nevertheless, extensive research has shown that norepinephrine plays a crucial role in its regulation. The present knowledge that has been reviewed in this manuscript suggests that neurons in the brain stem are responsible for controlling this state and presence of excess norepinephrine in the brain does not allow its generation. Furthermore, REM sleep loss increases levels of norepinephrine in the brain that affects several factors including an increase in Na-K ATPase activity. It has been argued that such increased norepinephrine is ultimately responsible for REM sleep deprivation, associated disturbances in at least some of the physiological conditions leading to alteration in behavioural expression and settling into pathological conditions.
Article
The pontine oral reticular nucleus, gigantocellular reticular nucleus (Gi) and dorsal paragigantocellular nucleus (DPGi) of the medulla are key elements of a brainstem-reticulospinal inhibitory system that participates in rapid eye movement (REM) sleep atonia. Our recent study has shown that excitation of these brainstem nuclei in decerebrate rats inhibits locus coeruleus cells and the midbrain locomotor region neurons related to muscle tone facilitation. In the present study we have examined the influences of electrical and chemical stimulation of Gi and DPGi inhibitory sites on the activity of neurons located in the magnocellular part of the red nucleus (RMC), a cell group that participates in both the tonic and phasic regulation of motor output. A total of 192 RMC neurons were recorded in precollicular-premammillary decerebrate rats with muscle rigidity and induced locomotion. Thirty-three RMC neurons were identified antidromically as rubrospinal (RMC-spinal) cells by stimulation of the contralateral dorsolateral funiculus at the L2 level. A total of 141 RMC neurons (88.7 %) and all RMC-spinal neurons were inhibited during electrical stimulation of Gi and DPGi inhibitory sites. This cessation of activity was correlated with bilateral muscle atonia or blockage of locomotion. Six RMC cells (3.8 %) were excited (224 +/- 50 %, n = 6, minimum = 98, maximum = 410, P < 0.05) and 11 cells (7 %) gave no response to Gi and DPGi stimulation. Microinjections of kainic acid (100 microM, 0.2 microl) into Gi and DPGi inhibitory sites, previously identified by electrical stimulation, produced a short-latency (35 +/- 3.5 s, n = 11) decrease of rigid hindlimb muscle tone and inhibition of all tested RMC (n = 7) and RMC-spinal (n = 5) neurons. These results, combined with our recent published data, suggest that inhibition of motor function during activation of the brainstem inhibitory system is related to both the descending inhibition of spinal motoneurons and suppression of activity in supraspinal motor facilitatory systems. These two mechanisms acting synergistically may cause generalized motor inhibition during REM sleep and cataplexy.
Article
Full-text available
Our study aimed to evaluate the existence and entity of changes in sleep structure following vagus nerve stimulation in patients with refractory epilepsy. A polysomnographic study was performed on the nocturnal sleep of 10 subjects with refractory epilepsy. Subjects were recorded both in baseline conditions and after chronic vagus nerve stimulation. Sleep parameters of the entire night were evaluated. Mean power value of slow-wave activity was computed in the first non-rapid eye movement sleep cycle. A sleep-wake diary evaluated quantity of both nocturnal and daytime sleep, while visual-analog scales assessed quality of sleep and wake. The differences between the 2 conditions underwent parametric and nonparametric statistical evaluation. Vagus nerve stimulation produced a significant reduction in REM sleep (in all subjects with vagus nerve stimulus intensity greater than 1.5 milliampere, but not in the only patient with a stimulus intensity less than 1.5 milliampere), along with an increase in the number of awakenings, percentage of wake after sleep onset, and stage 1 sleep. Data from a sleep-wake questionnaire show a decrease in both nocturnal sleep and daytime naps and an increased daytime alertness, while the quality of wakefulness is globally improved. Spectral analysis shows an enhancement of delta power during non-rapid eye movement sleep. Our data demonstrate major effects of vagus nerve stimulation on both daytime alertness (which is improved) and nocturnal rapid eye movement sleep (which is reduced). These effects could be interpreted as the result of a destabilizing action of vagus nerve stimulation on neural structures regulating sleep-wake and rapid eye movement/non-rapid eye movement sleep cycles. Lower intensity vagus nerve stimulation seems only to improve alertness; higher intensity vagus nerve stimulation seems able to exert an adjunctive rapid eye movement sleep-attenuating effect.
Article
Full-text available
The noradrenergic (NA-ergic) rapid eye movement (REM)-OFF neurons in locus coeruleus (LC) and cholinergic REM-ON neurons in laterodorsal/pedunculopontine tegmentum show a reciprocal firing pattern. The REM-ON neurons fire during REM sleep whereas REM-OFF neurons stop firing during REM sleep. The cessation of firing of REM-OFF neurons is a pre-requisite for the generation of REM sleep and non-cessation of those neurons result in REM sleep loss that is characterized by symptoms like loss of memory retention, irritation, hypersexuality, etc. There is an intricate interplay between the REM-OFF and REM-ON neurons for REM sleep regulation. Acetylcholine from REM-ON neurons excites the GABA-ergic interneurons in the LC that in turn inhibit the REM-OFF neurons. The cessation of firing of REM-OFF neurons withdraws the inhibition from the REM-ON neurons, and facilitates the excitation of these neurons resulting in the initiation of REM sleep. GABA modulates the generation of REM sleep in pedunculopontine tegmentum (PPT) by acting pre-synaptically on the NA-ergic terminals that synapse on the REM-ON neurons whereas in LC it modulates the maintenance of REM sleep by acting post-synaptically on REM-OFF neurons. The activity of REM sleep related neurons is modulated by wakefulness (midbrain reticular formation/ascending reticular activating system) and sleep inducing (caudal brainstem/medullary reticular formation) areas. Thus, during wakefulness the wake-active neurons keep on firing that excites the REM-OFF neurons, which in turn keeps the REMON neurons inhibited; therefore, during wakefulness REM sleep episodes are not expressed. Additionally, the wakefulness inducing area keeps the REM-ON neurons inhibited. In contrast, the sleep inducing area excites the REM-ON neurons. Thus, the wakefulness inducing area excites and inhibits the REM-OFF and REM-ON neurons, respectively, while the sleep inducing area excites the REM-ON neurons that facilitate the generation of REM sleep.
Article
Full-text available
The only mesopontine neurons previously described as involved in the transfer of ponto-geniculo-occipital (PGO) waves from the brain stem to the thalamus were termed PGO-on bursting cells. We have studied, in chronically implanted cats, neuronal activities in brain-stem peribrachial (PB) and laterodorsal tegmental (LDT) cholinergic nuclei in relation to PGO waves recorded from the lateral geniculate (LG) thalamic nucleus during rapid-eye-movement (REM) sleep. We constructed peri-PGO histograms of PB/LDT cells' discharges and analyzed the interspike interval distribution during the period of increased neuronal activity related to PGO waves. Six categories of PGO-related PB/LDT neurons with identified thalamic projections were found: 4 classes of PGO-on cells: PGO-off but REM-on cells: and post-PGO cells. The physiological characteristics of a given cell class were stable even during prolonged recordings. One of these cell classes (1) represents the previously described PGO-on bursting neurons, while the other five (2–6) are newly discovered neuronal types. (1) Some neurons (16% of PGO-related cells) discharged stereotyped low-frequency (120– 180 Hz) spike bursts preceding the negative peak of the LG-PGO waves by 20–40 msec. These neurons had low firing rates (0.5–3.5 Hz) during all states. (2) A distinct cell class (22% of PGO-related neurons) fired high-frequency spike bursts (greater than 500 Hz) about 20–40 msec prior to the thalamic PGO wave. These bursts were preceded by a period (150–200 msec) of discharge acceleration on a background of tonically increased activity during REM sleep. (3) PGO-on tonic neurons (20% of PGO-related neurons) discharged trains of repetitive single spikes preceding the thalamic PGO waves by 100–150 msec, but never fired high- frequency spike bursts. (4) Other PGO-on neurons (10% of PGO-related neurons) discharged single spikes preceding thalamic PGO waves by 15–30 msec. On the basis of parallel intracellular recordings in acutely prepared, reserpine-treated animals, we concluded that the PGO-on single spikes arise from conventional excitatory postsynaptic potentials and do not reflect tiny postinhibitory rebounds. (5) A peculiar cellular class, termed PGO-off elements (8% of PGO-related neurons), consisted of neurons with tonic, high discharge rates (greater than 30 Hz) during REM sleep. These neurons stopped firing 100– 200 msec before and during the thalamic PGO waves. (6) Finally, other neurons discharged spike bursts or tonic spike trains 100–300 msec after the initially negative peak of the thalamic PGO field potential (post-PGO elements, 23% of PGO-related neurons).(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Full-text available
The only mesopontine neurons previously described as involved in the transfer of ponto-geniculo-occipital (PGO) waves from the brain stem to the thalamus were termed PGO-on bursting cells. We have studied, in chronically implanted cats, neuronal activities in brain-stem peribrachial (PB) and laterodorsal tegmental (LDT) cholinergic nuclei in relation to PGO waves recorded from the lateral geniculate (LG) thalamic nucleus during rapid-eye-movement (REM) sleep. We constructed peri-PGO histograms of PB/LDT cells' discharges and analyzed the interspike interval distribution during the period of increased neuronal activity related to PGO waves. Six categories of PGO-related PB/LDT neurons with identified thalamic projections were found: 4 classes of PGO-on cells: PGO-off but REM-on cells: and post-PGO cells. The physiological characteristics of a given cell class were stable even during prolonged recordings. One of these cell classes (1) represents the previously described PGO-on bursting neurons, while the other five (2-6) are newly discovered neuronal types. (1) Some neurons (16% of PGO-related cells) discharged stereotyped low-frequency (120-180 Hz) spike bursts preceding the negative peak of the LG-PGO waves by 20-40 msec. These neurons had low firing rates (0.5-3.5 Hz) during all states. (2) A distinct cell class (22% of PGO-related neurons) fired high-frequency spike bursts (greater than 500 Hz) about 20-40 msec prior to the thalamic PGO wave. These bursts were preceded by a period (150-200 msec) of discharge acceleration on a background of tonically increased activity during REM sleep. (3) PGO-on tonic neurons (20% of PGO-related neurons) discharged trains of repetitive single spikes preceding the thalamic PGO waves by 100-150 msec, but never fired high-frequency spike bursts. (4) Other PGO-on neurons (10% of PGO-related neurons) discharged single spikes preceding thalamic PGO waves by 15-30 msec. On the basis of parallel intracellular recordings in acutely prepared, reserpine-treated animals, we concluded that the PGO-on single spikes arise from conventional excitatory postsynaptic potentials and do not reflect tiny postinhibitory rebounds. (5) A peculiar cellular class, termed PGO-off elements (8% of PGO-related neurons), consisted of neurons with tonic, high discharge rates (greater than 30 Hz) during REM sleep. These neurons stopped firing 100-200 msec before and during the thalamic PGO waves. (6) Finally, other neurons discharged spike bursts or tonic spike trains 100-300 msec after the initially negative peak of the thalamic PGO field potential (post-PGO elements, 23% of PGO-related neurons).(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Full-text available
The distribution of acetylcholine neurons in the brainstem of the cat was studied by choline acetyltransferase (ChAT) immunohistochemistry and compared to that of catecholamine neurons examined in the same or adjacent sections by tyrosine hydroxylase (TH) immunohistochemistry. The largest group of ChAT-positive (+) neurons was located in the lateral pontomesencephalic tegmentum within the pedunculopontine tegmental nucleus and the laterodorsal tegmental nucleus rostrally and within the parabrachial nuclei and locus coeruleus nucleus more caudally. TH+ neurons were found to be coextensive and intermingled with ChAT+ neurons in the dorsolateral pontomesencephalic tegmentum, where the number of ChAT+ cells (approximately 18,500) exceeded that of the TH+ cells (approximately 12,000). In the caudal pons, scattered ChAT+ neurons were situated in the ventrolateral tegmentum together with TH+ neurons. In the medulla, numerous ChAT+ cells were located in the lateral tegmental field, where they extended in a radial column from the dorsal motor nucleus of the vagus to the ventrolateral tegmentum around the facial and ambiguus nuclei, occupying the position of preganglionic parasympathetic neurons of the 7th, 9th, and 10th cranial nerves. TH+ cells were also present in this field. Neurons within the general visceral, special visceral, and somatic motor cranial nerve nuclei were all immunoreactive to ChAT. Scattered ChAT+ neurons were also present within the medullary gigantocellular and magnocellular tegmental fields together with a small number of TH+ neurons. Other groups of ChAT+ cells were identified within the periolivary nuclei, parabigeminal nucleus, prepositus hypoglossi nucleus, and the medial and inferior vestibular nuclei. Acetylcholine neurons thus constitute a heterogeneous population of cells in the brainstem, which in addition to including the somatic and visceral efferent systems, comprises many other discrete systems and represents an important component of the brainstem reticular formation. The proximity to and interdigitation with catecholamine neurons within these systems may be of important functional significance.
Article
Full-text available
This study documents the time course profiles for simultaneous measures of: the electrographic signs of sleep and wakefulness, ponto-geniculo-occipital (PGO) waves, and extracellular discharge potentials for single cells in the dorsal raphe nucleus (DRN). These measures were obtained from intact, undrugged cats across 177 sleep cycles. Ninety-one of these sleep cycles were recorded with no prior forced activity. Forced activity previously has been shown to powerfully alter the temporal organization of sleep by shortening the duration of both the sleep cycle and the ultradian rhythm of DRN discharge. The present paper evaluated the hypothesis that DRN discharge time course might regulate the sleep cycle. These experiments documented the phase relationship between the time course of DRN discharge and the electrographic signs of sleep. These phase relationship were examined by determining whether forced locomotor activity could dissociate the time course profile for behavioral states, PGO waves, and DRN discharge. The results revealed that the time course of DRN discharge and PGO waves were always phase-locked to the time course of the ultradian sleep cycle. Furthermore, the results show that changes in DRN discharge consistently precede changes in PGO waves, and behavioral state. Since a cause must precede an effect, these data are consistent with the hypothesis that the DRN may be causally involved in sleep cycle regulation. These temporal data also provide parameter values for the continued evaluation of cellularly based, mathematical models of sleep cycle control.
Article
1. Neurons were recorded in the parabrachial (PB) area, located in the dorsolateral region of the pons (with the use of extracellular micropipette), in the anesthetized rat. Parabrachioamygdaloid (PA) neurons (n = 67) were antidromically identified after stimulation in the centralis nucleus of the amygdala (Ce). The axons of these neurons exhibit a very slow conduction velocity, between 0.26 and 1.1 m/s, i.e., in the unmyelinated range. 2. These PA neurons were located in a restricted region of the PB area: the subnuclei external lateral (PBel) and external medial (PBem). A relative somatotopic organization was found in this region. 3. These units were separated into two groups: 1) a group of nociceptive-specific (NS) neurons (69%), which responded exclusively to noxious stimuli, and 2) a group of nonresponsive (NR) neurons (31%). 4. The NS neurons exhibited low or lacked spontaneous activity. They responded exclusively to mechanical (pinch or squeeze) and/or thermal (waterbath or waterjet greater than 44 degrees C) noxious stimuli with a marked and sustained activation with a rapid onset and generally without afterdischarge. Noxious thermal stimuli generally induced a stronger response than the noxious mechanical stimuli. These neurons exhibited a clear capacity to encode thermal stimuli in the noxious range: 1) the stimulus-response function was always positive and monotonic; 2) the slope of the curve progressively increased up to a maximum where it was very steep, then the steepness of the slope decreased close to the maximum response; and 3) the mean threshold was 44.1 +/- 2 degrees C, and the point of steepest slope of the mean curve was around 47 degrees C. 5. The excitatory receptive fields of the NS neurons were large in the majority (70%) of the cases and included several areas of the body. A more marked activation was often obtained from stimuli applied to one part of the body, denoted as the preferential receptive field (PRF). In the other cases (30%), the excitatory receptive field was relatively small (SRF) and restricted to one part of the body (the tail, a paw, a hemiface, or the tongue). Both the PRF and SRF were more often located on the contralateral side. In addition, noxious stimuli applied outside the excitatory receptive field were found to strongly inhibit the responses of NS neurons. 6. All the NS neurons responded to intense transcutaneous electrical stimulation applied to the PRF or SRF with two peaks of activation.(ABSTRACT TRUNCATED AT 400 WORDS)
Book
A monograph communicating the current realities and future possibilities of unifying basic studies on anatomy and cellular physiology with investigations of the behavioral and physiological events of waking and sleep. Steriade established the Laboratory of Neurophysiology at Laval U., Quebec; McCarl
Chapter
The sections in this article are:
Article
The efferent connections of the Edinger-Westphal (EW) nucleus of the cat have been examined using the autoradiographic anterograde axonal transport technique. Following injections of [3H]amino acids into the EW nucleus, fibers could be traced from this region to a number of sites in the caudal brain stem and spinal cord heretofore not known to receive an afferent input from this nucleus. Two descending pathways have been identified. One pathway travels in the medialmost aspect to the medial longitudinal bundle and terminates in the dorsal accessory olive. The other pathway leaves the nucleus laterally, coursing through the medial tegmentum, and then shifts to a ventrolateral position in the rostral rhombencephalon. Some fibers of this lateral pathway curve dorsally and terminate in the medial parabrachial nucleus. The remainder of this fiber system lies ventral to the spinal trigeminal complex, with some axons terminating in the subtrigeminal nucleus, while other fibers continue ventral to the caudal part of the spinal trigeminal nucleus and appear to terminate in the marginal layer and ventromedial part of this nucleus. Another component of this system terminates between the gracile and medial cuneate nuclei. The main pathways from the EW nucleus to the spinal cord include (1) some fibers which course through the dorsal column nuclei into the ventromedial part of the dorsal columns, and (2) other fibers which continue caudally immediately along the ventrolateral aspect of the spinal trigeminal nucleus and then proceed to the spinal cord in the region between the dorsal horn and the lateral cervical nucleus. These EW fibers appear to terminate mainly in Rexed's lamina I (marginal layer); other fibers, from both tracts appear to terminate in lamina V. There was no evidence for any ascending projections from the EW nucleus.
Article
The efferent connections of the lateral hypothalamic area (LHA) have been analyzed in a series of 30 rat brains with injections of 3H-amino acids into different parts of the area and the surrounding regions. Our findings indicate that all parts of the LHA contribute ascending and descending fibers to the medial forebrain bundle, and also project medially to certain of the adjoining hypothalamic nuclei. All levels of the LHA appear to send some fibers to a continuous group of structures that extends from the medial septal-diagonal band complex rostrally, through the lateral preoptic and lateral hypothalamic areas to the mammillary complex and the ventral tegmental area caudally. In addition, it is evident that cells at different levels within the LHA may have differential projections. Thus, the anterior and lateral parts of the LHA also appear to project substantially to the anterior hypothalamic area, the ventromedial and dorsomedial hypothalamic nuclei, the parataenial and paraventricular nuclei of the thalamus, and the medial part of the lateral habenular nucleus. Similarly, cells in the tuberal and posterior parts of the LHA project to the central gray, the longest projections from the posterior region reaching as far caudally as the central tegmental field, the parabrachial nucleus, the locus coeruleus, and the superior central and dorsal nuclei of the raphe. Viewed as a whole, the LHA is therefore well-suited to integrate inputs from the limbic system and brainstem and to relay them on the one hand to the medial zone of the hypothalamus and on the other to virtually every structure closely associated with the medial forebrain bundle and to the nuclei of origin of the major ascending monoaminergic systems.
Article
To define the role of the rostral pons in the control of release of ACTH, we stimulated electrically (30 sec, 200 muA, 50 Hz) 128 sites in the dorsal rostral pons of 20 cats anesthetized with chloralose/urethane. Responses of arterial pressure to electrical stimulation were prevented by lesions placed previously in the medulla. Plasma concentrations of ACTH were measured by radioimmunoassay. Active areas consisted of three regions: 1) lateral inhibitory: Locus subcoeruleus and anteroventral locus coeruleus (mean deltaACTH: -189, -164, -145 pg/ml at 1.5,3.0 and 6.0 min respectively, P less than 0.01);2) intermediate facilitatory:principal locus coeruleus and lateral ventral tegmental nucleus (mean deltaACTH: +81, +68, +37 pg/ml; P less than 0.05); and 3) medial inhibitory: dorsal tegmental nucleus, dorsal raphé and medial ventral tegmental nucleus (mean deltaACTH; -211, -212, -115 pg/ml; P less than 0.01). The former two areas received direct projections from medullary neurons activated or inhibited by atrial stretch, and, in turn, give rise to adrenergic and cholinergic projections to the medial hypothalamus. Since the release of ACTH is inversely correlated with right atrial stretch, the results suggest that the lateral inhibitory area and the intermediate facilitatory area are involved in mediation of changes in release of ACTH in response to hemodynamic changes.
Article
The efferent fiber connections of the nuclei of the amygdaloid complex with subcortical structures in the basal telencephalon, hypothalamus, midbrain, and pons have been studied in the rat and cat, using the autoradiographic method for tracing axonal connections. The cortical and thalamic projections of these nuclei have been described in previous papers (Krettek and Price, ′77b,c). Although the subcortical connections of the amygdaloid nuclei are widespread within the basal forebrain and brain stem, the projections of each nucleus have been found to be well defined, and distinct from those of the other amygdaloid nuclei. The basolateral amygdaloid nucleus projects heavily to the lateral division of the bed nucleus of the stria terminalis (BNST), to the caudal part of the substantia innominata, and to the ventral part of the corpus striatum (nucleus accumbens and ventral putamen) and the olfactory tubercle; it projects more lightly to the lateral hypothalamus. The central nucleus also projects to the lateral division of the BNST and the lateral hypothalamus, but in addition it sends fibers to the lateral part of the substantia nigra and the marginal nucleus of the brachium conjunctivum. The basomedial nucleus has projections to the ventral striatum and olfactory tubercle which are similar to those of the basolateral nucleus, but it also projects to the core of the ventromedial hypothalamic nucleus and the premammillary nucleus, and to a central zone of the BNST which overlaps the medial and lateral divisions. The medial nucleus also projects to the core of the ventromedial nucleus and the premammillary nucleus, but sends fibers to the medial division of the BNST and does not project to the ventral striatum. The posterior cortical nucleus projects to the premammillary nucleus and to the medial division of the BNST, but a projection from this nucleus to the ventromedial nucleus has not been demonstrated. Projections to the “shell” of the ventromedial nucleus have been found only from the ventral part of the subiculum and from a structure at the junction of the amygdala and the hippocampal formation, which has been termed the amygdalo‐hippocampal area (AHA). The AHA also sends fibers to the medial part of the BNST and the premammillary nucleus. Virtually no subcortical projections outside the amygdala itself have been demonstrated from the lateral nucleus, or from the olfactory cortical areas around the amygdala (the anterior cortical nucleus, the periamygdaloid cortex, and the posterior prepiriform cortex). However, portions of the endopiriform nucleus deep to the prepiriform cortex project to the ventral putamen, and to the lateral hypothalamus.
Article
Using a recently developed retrograde tracer technique with horseradish peroxidase (HRP), attempts were made to identify afferent projections to the dorso-lateral part of the pontomesencephalic tegmental areas, including the nucleus locus coeruleus (LC), locus subcoeruleus (LSC), parabrachialis lateralis (Pbl), Kölliker-Fuse (K-F), reticularis pontis oralis (RPO), reticularis pontis caudalis (RPC), as well as an area rostral to the Pbl and dorsolateral to the brachium conjunctivum (mesencephalic reticular formation (MRF) area). It was revealed that the nucleus raphe dorsalis projects widely to all the studied pontomesencephalic tegmental nuclei. In addition, the LC was found to project to all the contralateral pontine tegmental structures which were studied. Following the injection of HRP into the dorsomedial part of the LC (‘principal LC’ or ‘LC’), HRP labeled neurons were observed almost restricted to the nucleus raphe dorsalis. In contrast, following the injection into the ventrolateral part of the LC (LCα) or into other tegmental areas, the HRP containing neurons were observed widely distributed in the brain, extending from the diencephalon to the rhombencephalon. Especially in the case where injection was made into the LCα, numerous HRP positive cells appeared in the nucleus raphe pontis, magnus and substantia nigra, and were also identified in other brain structures, the topography of which corresponded to that of the catecholamine-containing neurons of the rat (group A1–A14). The present results confirm some previous reports on the afferent connections of the LC described in the rat, cat and rabbit, and further indicate the richness of afferent projections to the dorsolateral pontine tegmental areas. The present study also shows the heterogeneity existing between the main LC areas and the subcoeruleus areas, as well as between the dorsomedial part of the main LC (principal LC) and the ventrolateral part of this nucleus (LCα).
Article
Discharge patterns of the nucleus parabrachialis lateralis (PbL) neurons located in the dorsolateral pontine tegmentum were investigated in 7 unrestrained, freely behaving cats with a multiwire electrode bundle method. Among 84 PbL units recorded so far, 31% of the units exhibited a marked reduction or complete cessation of firing during paradoxical sleep (PS) (PS-off cells), while 55% of the units exhibited an increase of firing during PS (PS-on cells) as compared with quiet wakefulness (QW) and slow wave sleep (SWS). The remaining 14% of the PbL units exhibited no remarkable change of firing rate and pattern during the sleep-walking cycle. The PbL PS-off cells were further characterized by a slow and regular firing pattern during QW and SWS, and they were similar in several respects to the PS-off cells reported in the nucleus raphe dorsalis and locus coeruleus complex. Six of the PbL PS-on cells were tightly phase-locked with PGO waves, and the spikes preceded by 5-25 msec the onset of the PGO waves recorded in the lateral geniculate nucleus. Seven other PbL PS-on cells were related to the locomotor activity observed in active wakefulness. In light of the present results, the functional significance of the PbL units has been discussed.
Article
Increasingly strong evidence suggests that cholinergic neurons in the mesopontine tegmentum play important roles in the control of wakefulness and sleep. To understand better how the activity of these neurons is regulated, the potential afferent connections of the laterodorsal (LDT) and pedunculopontine tegmental nuclei (PPT) were investigated in the rat. This was accomplished by using retrograde and anterograde axonal transport methods and NADPH-diaphorase histochemistry. Immunohistochemistry was also used to identify the transmitter content of some of the retrogradely identified afferents.
Article
We examined the functional organization of the parabrachial complex (PB) by mapping the cardiovascular and respiratory responses to PB microstimulation in anesthetized rats. The PB was explored with 100 microns resolution, at threshold doses of electrical current (5 microA) and glutamate (10-500 pmols), and the locations of stimulation sites were identified by small iontophoretic or pressure injections of biocytin or Phaseolus vulgaris leucoagglutinin. Threshold doses of either L-glutamate or electrical current pulses caused pressor-tachycardic responses that mapped to the outer edge of the external lateral subnucleus while depressor bradycardic responses were elicited from stimuli near the dorsal lateral subnucleus. Pressor responses persisted in paralyzed, ventilated animals and were thus not dependent upon concomitant respiratory changes. Cardiac arrhythmias sometimes occurred during large pressor responses and during augmented breaths that occurred during or following PB stimulation. These observations indicate that the PB contains at least two distinct neuronal systems that are potently and opposingly involved in cardiovascular control. The locations of the sites giving the most potent responses implicate specific ascending and descending pathways as substrates for the cardiovascular responses.
Article
The hypothesis that REM sleep is cholinergically mediated is supported by the identification of a cholinoceptive trigger zone in the FTG. Since this trigger zone is devoid of cholinergic neurons, the aim of the present study was to test the hypothesis that a cholinergic drive for REM sleep may come from the cholinergic cells of the PBL region. Chronically implanted freely moving cats with electrodes for sleep and PGO wave recordings were used. Guide tubes were implanted for carbachol microinjections (4 micrograms/250 nl) in the PBL and FTG. All microinjections were delivered in close vicinity of ChAT+ cholinergic cells in the PBL region. Results showed that a single unilateral carbachol microinjection into the PBL induced sustained (24 hr) state-independent ipsilateral PGO wave activity. This PGO wave activity was followed by a prolonged enhancement of REM sleep lasting for more than six days. We also observed that REM enhancement was followed by a delayed but marked enhancement of S sleep episodes with PGO waves (SP), which are normally brief transitions from S to REM sleep. Our findings strongly support the hypothesis that cholinergic drive for REM sleep comes from the lateral pontine tegmentum and we suggest that the PBL region plays a major role in both PGO wave generation and long-term regulation of REM sleep induction.
Article
The cholinergic agonist carbachol was injected into the pontine Pb area where PGO bursting cells have been recorded. When microinjections were localized to the ventrolateral aspect of the caudal Pb nucleus near aggregates of ChAT immunolabeled cholinergic neurons, carbachol produced an immediate onset of state-independent PGO waves in the ipsilateral LGB. These state-independent PGO waves persisted for 3-4 days. After the first 24 hrs PGO wave activity increasingly became associated with REM sleep and with REM transitional SP sleep as both of these PGO-related states increased in amount to 3-4 times baseline levels. The increase in amount of PGO-related states peaked on days 2-4 following one carbachol injection and persisted for 10-12 days. These results suggest a two stage process: stage one, PGO enhancement, is the direct consequence of the membrane activation of cholinoceptive PGO burst neurons by carbachol; stage two, REM enhancement, is the consequence of metabolic activation of endogenous cholinergic neurons. This experimental preparation is a useful model for the study of the electrophysiology and functional significance of PGO wave and REM sleep generation.
Article
We examined the subnuclear organization of projections to the parabrachial nucleus (PB) from the nucleus of the solitary tract (NTS), area postrema, and medullary reticular formation in the rat by using the anterograde and retrograde transport of wheat germ agglutinin‐horseradish peroxidase conjugate and anterograde tracing with Phaseolus vulgaris ‐leucoagglutinin. Different functional regions of the NTS/area postrema complex and medullary reticular formation were found to innervate largely nonoverlapping zones in the PB. The general visceral part of the NTS , including the medial, parvicellular, intermediate, and commissural NTS subnuclei and the core of the area postrema, projects to restricted terminal zones in the inner portion of the external lateral PB, the central and dorsal lateral PB subnuclei, and the “waist” area. The dorsomedial NTS subnucleus and the rim of the area postrema specifically innervate the outer portion of the external lateral PB subnucleus. In addition, the medial NTS innervates the caudal lateral part of the external medial PB subnucleus. The respiratory part of the NTS , comprising the ventrolateral, intermediate, and caudal commissural subnuclei, is reciprocally connected with the Kölliker‐Fuse nucleus, and with the far lateral parts of the dorsal and central lateral PB subnuclei. There is also a patchy projection to the caudal lateral part of the external medial PB subnucleus from the ventrolateral NTS. The rostral, gustatory part of the NTS projects mainly to the caudal medial parts of the PB complex, including the “waist” area, as well as more rostrally to parts of the medial, external medial, ventral, and central lateral PB subnuclei. The connections of different portions of the medullary reticular formation with the PB complex reflect the same patterns of organization, but are reciprocal. The periambiguus region is reciprocally connected with the same PB subnuclei as the ventrolateral NTS; the rostral ventrolateral reticular nucleus with the same PB subnuclei as both the ventrolateral (respiratory) and medial (general visceral) NTS; and the parvicellular reticular area , adjcent to the rostral NTS, and with parts of the central and ventral lateral and the medial PB subnuclei that also receive rostral (gustatory) NTS input. In addition, the rostral ventrolateral reticular nucleus and the parvicellular reticular formation have more extensive connections with parts of the rostal PB and the subjacent reticular formation that recieve little if any NTS input. The PB contains a series of topographically complex terminal domains reflecting the functional organization of its afferent sources in the NTS and medullary reticular formation.
Article
A six day long increase in rapid eye movement (REM) sleep followed the unilateral microinjection of a single dose of the cholinergic agonist drug carbachol into the brain stem of cats. Effective drug injection sites were localized to the pontine peribrachial region containing cholinergic choline acetyltransferase (ChAT) labeled neurons. At the peak of the effect, which occurred 24-28 h post-injection, the relative amount of time devoted to REM sleep tripled, resulting in an absolute time increase from 3.12 to 11.28 h REM sleep per day. This pronounced and prolonged REM sleep increase was associated with marked enhancement of ponto-geniculo-occipital (PGO) waves and with PGO burst cell activity unilateral to the site of injection.
Article
Mapping neuronal populations that induce behavioral state changes after pharmacological activation requires discrete localization of drug injection sites, and is limited by widespread diffusion of molecular drugs. Nanospheres with diameters of 50-100 nm can reduce diffusion significantly because of their relatively large sizes. The cholinergic agonist carbachol was radiolabeled with methyl14C and incorporated within a latex nanosphere delivery system (LNDS). We quantitatively compared diffusion of 14C-carbachol within these nanospheres with that of free 14C-carbachol, demonstrating approximately ten-fold reduced radial diffusion by nanospheres 10 min to 24 h post-injection; approximately 90% of injected radioactivity was restricted to regions within approximately 100-150 microns and 1400-1500 microns respectively. Thus, incorporation of active agents such as drugs within nanospheres dramatically increases the precision of their delivery in-vivo (here about 1,000-fold by volume).
Article
Carbachol, a long-acting cholinergic agonist, was microinjected (4 micrograms/250 nl per 90 s) into 90 sites within the anterodorsal pontine tegmentum of four cats and the time to onset and percentage of time spent in a desynchronized sleep-like state during 40 min postinjection were calculated. Compared with more posteroventral pontine sites, the shorter latencies and higher percentages observed confirmed earlier predictions of a sensitive cholinoceptive zone in the anterodorsal pons. In 27 trials a desynchronized sleep-like state was observed within 5 min; in 31 trials the latency was 5-10 min and in the remaining 32 trials, greater than 10 min. Plotting the desynchronized sleep-like state latency and the desynchronized sleep-like state percentage as a function of the three-dimensional coordinates revealed that injection sites with short latency (less than 5 min) and high percentage (greater than 80%) were concentrated between the coordinates of P 1.0 to 3.5 and V -3.5 to -5.5, at the lateral coordinate L 2.0. On the frontal plane, the short desynchronized sleep-like state latency and high desynchronized sleep-like state percentage sites begin in the pontine tegmental region just lateral to the ventral tegmental nucleus and extend 3 mm ventrocaudally. A regression plot of the data in sagittal plane 2.0 revealed a short latency axis, around which the short latency sites cluster, running in a slightly dorsoventral direction from about P 1.0 to V -4.0 to P 4.0 to V -5.5. This observation suggests that the sensitive zone might approximate a cylinder in shape, a hypothesis supported by the correlation of longer latencies and lower percentages at increasing radial distance from the axis. The non-linear relationship between cholinergic potency and distance from the short latency axis suggests that the desynchronized sleep-like state latency is a function of two factors; a variable diffusion-based delay of carbachol to distant neuronal populations involved in the desynchronized sleep-like state production, and a fixed recruitment-based delay following activation of neurons in the sensitive zone. Interpretation of these findings in light of earlier studies involving microstimulation of the pontine tegmentum argue in favor of a distributed network of discrete neuronal populations as the source of desynchronized sleep generation.
Article
1. Neurons were recorded in the parabrachial (PB) area, located in the dorsolateral region of the pons (with the use of extracellular micropipette), in the anesthetized rat. Parabrachioamygdaloid (PA) neurons (n = 67) were antidromically identified after stimulation in the centralis nucleus of the amygdala (Ce). The axons of these neurons exhibit a very slow conduction velocity, between 0.26 and 1.1 m/s, i.e., in the unmyelinated range. 2. These PA neurons were located in a restricted region of the PB area: the subnuclei external lateral (PBel) and external medial (PBem). A relative somatotopic organization was found in this region. 3. These units were separated into two groups: 1) a group of nociceptive-specific (NS) neurons (69%), which responded exclusively to noxious stimuli, and 2) a group of nonresponsive (NR) neurons (31%). 4. The NS neurons exhibited low or lacked spontaneous activity. They responded exclusively to mechanical (pinch or squeeze) and/or thermal (waterbath or waterjet greater than 44 degrees C) noxious stimuli with a marked and sustained activation with a rapid onset and generally without afterdischarge. Noxious thermal stimuli generally induced a stronger response than the noxious mechanical stimuli. These neurons exhibited a clear capacity to encode thermal stimuli in the noxious range: 1) the stimulus-response function was always positive and monotonic; 2) the slope of the curve progressively increased up to a maximum where it was very steep, then the steepness of the slope decreased close to the maximum response; and 3) the mean threshold was 44.1 +/- 2 degrees C, and the point of steepest slope of the mean curve was around 47 degrees C. 5. The excitatory receptive fields of the NS neurons were large in the majority (70%) of the cases and included several areas of the body. A more marked activation was often obtained from stimuli applied to one part of the body, denoted as the preferential receptive field (PRF). In the other cases (30%), the excitatory receptive field was relatively small (SRF) and restricted to one part of the body (the tail, a paw, a hemiface, or the tongue). Both the PRF and SRF were more often located on the contralateral side. In addition, noxious stimuli applied outside the excitatory receptive field were found to strongly inhibit the responses of NS neurons. 6. All the NS neurons responded to intense transcutaneous electrical stimulation applied to the PRF or SRF with two peaks of activation.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
The ascending projections of cholinergic neurons in the laterodorsal tegmental nucleus (TLD) were investigated in the rat by using Phaseolus vulgaris leucoagglutinin (PHA‐L) and wheat germ agglutinin‐conjugated horseradish peroxidase (WGA‐HRP) anterograde tracing techniques. Two ascending pathways were identified after iontophoretic injections of PHA‐L into the TLD. A long projection system courses through the dorsomedial tegmentum, caudal diencephalon, medial forebrain bundle, and diagonal band. Different branches of this system innervate the midbrain (superior colliculus, interstitial magnocellular nucleus of the posterior commissure, and anterior pretectal nucleus), the diencephalon (lateral habenular nucleus, parafascicular, anteroventral, anterodorsal, mediodorsal, and intralaminar thalamic nuclei), and the telencephalon (lateral septum and medial prefrontal cortex). The second system is shorter and more diffuse and innervates the median raphe, interpeduncular, and lateral mammillary nuclei. Retrograde tracing with WGA‐HRP, combined with choline acetyltransferase immunohistochemistry, revealed that most of the TLD projections to the tectum, pretectum, thalamus, lateral septum, and medial prefrontal cortex are cholinergic. Afferents to the TLD were studied by anterograde and retrograde tracing techniques. Injection of tracers into the TLD retrogradely labelled neurons bilaterally in the midbrain reticular formation, the periaqueductal gray, the medial preoptic nucleus, the anterior hypothalamic nucleus, and the perifornical and lateral hypothalamic areas. Retrogradely labelled cells were also located bilaterally in the premammillary nucleus, paraventricular hypothalamic nucleus, zona incerta, and lateral habenular nucleus. In the telencephalon, the nucleus of the diagonal band and the medial prefrontal cortex contained retrogradely labelled neurons ipsilateral to the TLD injection site. The projections of the medial prefrontal cortex, the bed nucleus of the stria terminalis, and the lateral habenular nucleus to the TLD were confirmed in anterograde tracing studies. These findings indicate that the TLD gives rise to several ascending cholinergic projections that innervate diverse regions of the forebrain. Afferents to the TLD arise in hypothalamic and limbic forebrain regions, some of which appear to have reciprocal connections with the TLD. The latter include the lateral habenular nucleus and medial prefrontal cortex.
Article
The cholinergic agonist carbachol was conjugated to latex microspheres that were fluorescently labeled with rhodamine and used as neuroanatomical probes that show little diffusion from their injection site and retrogradely label neurons projecting to the injection site. Microinjection of this pharmacologically active probe into the gigantocellular field of the cat pontine brain stem caused the awake cats to fall into rapid movement (REM) sleep indistinguishable from that produced by free carbachol. Three-dimensional computer reconstruction of the retrogradely labeled neurons revealed a widely distributed neuronal network in the pontine tegmentum. These pharmacologically active microspheres permit a new precision in the characterization and mapping of neurons associated with the control of behavioral state and of other cholinergic networks.
Article
The pedunculopontine tegmental nucleus (PPTn) was originally defined on cytoarchitectonic grounds in humans. We have employed cytoarchitectonic, cytochemical, and connectional criteria to define a homologous cell group in the rat. A detailed cytoarchitectonic delineation of the mesopontine tegmentum, including the PPTn, was performed employing tissue stained for Nissl substance. Choline acetyltransferase (ChAT) immunostained tissue was then analyzed in order to investigate the relationship of cholinergic perikarya, dendritic arborizations, and axonal trajectories within this cytoarchitectonic scheme. To confirm some of our cytoarchitectonic delineations, the relationships between neuronal elements staining for ChAT and tyrosine hydroxylase were investigated on tissue stained immunohistochemically for the simultaneous demonstration of these two enzymes. The PPTn consists of large, multipolar neurons, all of which stain immunohistochemically for ChAT. It is present within cross‐sections that also include the A‐6 through A‐9 catecholamine cell groups and is traversed by catecholaminergic axons within the dorsal tegmental bundle and central tegmental tract. The dendrites of PPTn neurons respect several nuclear boundaries and are oriented perpendicularly to several well‐defined fiber tracts. Cholinergic axons ascend from the mesopontine tegmentum through the dorsal tegmental bundle and a more lateral dorsal ascending pathway. A portion of the latter terminates within the lateral geniculate nucleus. It has been widely believed that the PPTn is reciprocally connected with several extrapyramidal structures, including the globus pallidus and substantia nigra pars reticulata. Therefore, the relationships of pallidotegmental and nigrotegmental pathways to the PPTn were investigated employing the anterograde autoradiographic methodology. The reciprocity of tegmental connections with the substantia nigra and entopeduncular nucleus was investigated employing combined WGA‐HRP injections and ChAT immunohistochemistry. The pallido‐ and nigrotegmental terminal fields did not coincide with the PPTn, but, rather, were located just medial and dorsomedial to it (the midbrain extrapyramidal area). The midbrain extrapyramidal area, but not the PPTn, was reciprocally connected with the substantia nigra and entope‐duncular nucleus. We discuss these results in light of other cytoarchitec‐tonic, cytochemical, connectional, and physiologic studies of the functional anatomy of the mesopontine tegmentum.
Article
The immunohistochemical localization of the neurotransmitter synthesizing enzymes choline acetyltransferase, tyrosine hydroxylase and dopamine-beta-hydroxylase was examined in the feline pontomesencephalic tegmentum. Examination of adjacent sections stained for either choline acetyltransferase, tyrosine hydroxylase or dopamine-beta-hydroxylase immunoreactivity, as well as individual sections doubly stained for both choline acetyltransferase and tyrosine hydroxylase immunoreactivity, unequivocally demonstrated that noradrenergic and cholinergic neurons were extensively intermingled in the brainstem tegmentum of the cat. This contrasts with the situation in various other species, where neurons utilizing these two neurotransmitters are discretely localized in distinct nuclei. Furthermore, the present studies demonstrate the existence of two types of choline acetyltransferase immunoreactive neurons in the feline tegmentum: the magnocellular neurons of the pedunculopontine and laterodorsal tegmental nuclei which stain histochemically for NADPH diaphorase, plus a population of small spindle-shaped neurons in the medial and lateral parabrachial nuclei which do not stain positively for NADPH diaphorase. The data are discussed with respect to several influential hypotheses of sleep cycle control.
Article
In the present study we examined the distribution of cholinergic and catecholaminergic neurons, in the feline brainstem, as defined by choline acetyltransferase (ChAT) and tyrosine hydroxylase (TH) immunohistochemistry. In the dorsal tegmentum, ChAT immunoreactive neurons were distributed in the parabrachial area [the pedunculopontine group (PPG)] and along the medial adjacent central gray [the lateral dorsal tegmental group (LDT)]. The cholinergic neurons in the LDT area were larger than those in the PPG. When adjacent tissue sections were labeled with TH we noted extensive overlap between catecholamine and cholinergic neurons in the PPG, suggesting that REM sleep may occur as a result of an interaction between these transmitters in this area rather than the medial pontine reticular formation where no cholinergic or catecholamine neurons were found. Cholinergic neurons were also found in the cranial nerve nuclei and the nucleus ambiguus. The presence of cholinergic neurons in the PPG and LDT suggest that these neurons may play an important role in the generation of some of the tonic and phasic components of REM sleep, such as cortical desynchronization, pontogeniculo occipital waves, and muscle atonia.
Article
The laterodorsal tegmental nucleus (ntdl) contains a cluster of cells located just medial to the locus coeruleus in the pontine brainstem. The ntdl has been shown to project both rostrally to the forebrain and diencephalon and caudally to the spinal cord. In an effort to characterize this region neurochemically, the present study was conducted to identify a variety of neurochemicals localized within perikarya and fibers of the ntdl and surrounding nuclei. Rats were perfused with formalin, and brain sections were processed for fluorescence immunocytochemistry and acetylcholinesterase (AChE). Of the neurochemicals screened, atrial natriuretic factor (ANF), choline acetyltransferase (ChAT), cholecystokinin (CCK), calcitonin gene-related peptide (CGRP), dynorphin B (Dyn B), galanin, somatostatin, substance P, neurotensin (NT), neuropeptide Y (NPY), vasopressin, vasoactive intestinal polypeptide (VIP), serotonin (5HT), glutamic acid decarboxylase (GAD), and tyrosine hydroxylase (TH) were studied. AChE and ChAT staining revealed that the ntdl contains mostly cholinergic neurons. In addition, brightly reactive substance P and galanin and paler staining CRF, ANF, CGRP, NT, VIP, and Dyn B cell bodies were found within the ntdl. Varicose fibers in this nucleus also contained these peptides in addition to CCK, GAD, TH, 5HT, and NPY. The dorsal tegmental nucleus, dorsal raphe nucleus, locus coeruleus, and the parabrachial region contained a dense and varied assortment of peptides with distinct positions and patterns. This multiplicity of neurochemicals within this area suggests a possible influence on a variety of functions modulated by the ntdl and other closely associated tegmental nuclei.
Article
A data acquisition and analysis system based on an Apple II microcomputer has been developed for use in sleep studies in the adult cat. The system reliably counts delta, spindle, and EMG waveforms, PGO waves, and REMs using amplitude and frequency criteria. These data can be used to algorithmically score sleep-wake state with high reliability (greater than 90% agreement with manual scoring). This method allows for automatic and quantitative analysis of selected EEG waveforms and sleep-wake states with less expense, more time savings, and greater convenience than manual scoring.
Article
This study demonstrates that the laterodorsal tegmental nucleus (LDT) and pedunculopontine tegmental nucleus (PPT) are sources of cholinergic projections to the cat pontine reticular formation gigantocellular tegmental field (PFTG). Neurons of the LDT and PPT were double-labeled utilizing choline acetyltransferase immunohistochemistry combined with retrograde transport of horseradish peroxidase conjugated with wheat germ agglutinin (WGA-HRP). In the LDT the percentage of cholinergic neurons retrogradely labeled from PFTG was 10.2% ipsilaterally and 3.7% contralaterally, while in the PPT the percentages were 5.2% ipsilaterally and 1.3% contralaterally. These projections from the LDT and PPT to the PFTG were confirmed and their course delineated with anterograde labeling utilizing Phaseolus vulgaris leucoagglutinin (PHA-L) anterograde transport.
Article
The distribution of neurons displaying choline acetyltransferase (ChAT) immunoreactivity was examined in the feline brain using a monoclonal antibody. Groups of ChAT-immunoreactive neurons were detected that have not been identified previously in the cat or in any other species. These included small, weakly stained cells found in the lateral hypothalamus, distinct from the magnocellular rostral column cholinergic neurons. Other small, lightly stained cells were also detected in the parabrachial nuclei, distinct from the caudal cholinergic column. Many small ChAT-positive cells were also found in the superficial layers of the superior colliculus. Other ChAT-immunoreactive neurons previously detected in rodent and primate, but not in cat, were observed in the present study. These included a dense cluster of cells in the medial habenula, together with outlying cells in the lateral habenula. Essentially all of the cells in the parabigeminal nucleus were found to be ChAT-positive. Additional ChAT-positive neurons were detected in the periolivary portion of the superior olivary complex, and scattered in the medullary reticular formation. In addition to these new observations, many of the cholinergic cell groups that have been previously identified in the cat as well as in rodent and primate brain such as motoneurons, striatal interneurons, the magnocellular rostral cholinergic column in the basal forebrain and the caudal cholinergic column in the midbrain and pontine tegmentum were confirmed. Together, these observations suggest that the feline central cholinergic system may be much more extensive than previous studies have indicated.
Article
Microinjection of cholinergic agonists into the pontine tegmentum was used to evoke a state which was polygraphically and behaviorally similar to desynchronized (D) sleep. This study was designed to test the hypothesis that the production of this pharmacologically induced D sleep-like state (D-ACh) was dependent upon the anatomical locus of drug administration within the pontine tegmentum. Four dependent variables of D sleep were measured: D latency, D percentage, D duration and D frequency. Multiple regression analysis and analysis of variance were performed to evaluate the relationship between the three-dimensional coordinates of the injection site (posterior, vertical and lateral) and these 4 dependent measures. The intrapontine site of drug administration accounted for a statistically significant amount of the variance in D latency, D percentage and D duration. There was no significant relationship between the anatomical site of saline injection and the dependent measures of D sleep. A significant increase in D frequency following microinjection of cholinergic agonists was found to be independent of injection site. Pontine injection sites which yielded the shortest D latencies were found to be the same sites from which the highest D percentages were evoked. Rostrodorsal pontine tegmental injection sites were most effective in producing the highest percentages of D-ACh with the shortest latencies to onset. Injections made more caudally and ventrally within the pontine tegmentum produced lower percentages of D-ACh with longer latencies to onset. These data suggest the existence of an anatomical gradient within the pontine tegmentum for the cholinoceptive evocation of a D sleep-like state, and further support the concept that D sleep is generated, in part, by pontine cholinergic mechanisms.
Article
Electrical stimulation of sites within the parabrachial nucleus (PBN; 0.2 ms pulses at 50 hz for 10 s, 100 microA) of chloralose-anesthetized rats elicited increases in mean arterial pressure (+ 34 +/- 2 mm Hg, n = 13). In rats in which the electrode tip was located in the dorsal and lateral portions of the PBN, electrical stimulation also produced a small increase in plasma levels of arginine vasopressin (VP; 6.7 +/- 0.9 pg/ml before stimulation to 16.5 +/- 3.9 pg/ml after, n = 7, P less than 0.05). No change in VP levels was observed with stimulation of more medial and ventral portions of the PBN. Following blockade of the autonomic nervous system (by i.v. injection of chlorisondamine plus infusion of phenylephrine to prevent hypotension) VP release in response to stimulation of the lateral and dorsal portions of the PBN was greatly enhanced (8.8 +/- 1.4 pg/ml following ganglionic blockade but before PBN stimulation and 82 +/- 27 pg/ml following stimulation, P less than 0.01) and this increase in VP release was sufficient to increase arterial pressure (+ 18 +/- 3 mm Hg, P less than 0.01). Electrical stimulation of the locus coeruleus (LC) produced a pressor response of similar magnitude to that produced by PBN stimulation. LC stimulation also increased plasma VP release (5.6 +/- 0.3 pg/ml before stimulation, 13 +/- 3 pg/ml following stimulation, and 30 +/- 8 pg/ml following stimulation in the ganglionic blocked state; n = 5).(ABSTRACT TRUNCATED AT 250 WORDS)
Article
The ascending cholinergic projections of the pedunculopontine and dorsolateral tegmental nuclei, referred to collectively as the pontomesencephalotegmental (PMT) cholinergic complex, were investigated by use of fluorescent tracer histology in combination with choline-O-acetyltransferase (ChAT) immunohistochemistry and acetylcholinesterase (AChE) pharmacohistochemistry. Propidium iodide, true blue, or Evans blue was infused into the anterior, reticular, mediodorsal, central medial, and posterior nuclear areas of the thalamus; the habenula; lateral geniculate; superior colliculus; pretectal/parafascicular area; subthalamic nucleus; caudate-putamen complex; globus pallidus; entopeduncular nucleus; substantia nigra; medial septal nucleus/vertical limb of the diagonal band area; magnocellular preoptic/ventral pallidal area; and lateral hypothalamus. In some animals, separate injections of propidium iodide and true blue were made into two different regions in the same rat brain, usually a dorsal and a ventral target, in order to assess collateralization patterns. Retrogradely transported fluorescent labels and ChAT and/or AChE were analyzed microscopically on the same brain section. All of the above-delimited targets were found to receive cholinergic input from the PMT cholinergic complex, but some regions were preferentially innervated by either the pedunculopontine or dorsolateral tegmental nucleus. The former subdivision of the PMT cholinergic complex projected selectively to extrapyramidal structures and the superior colliculus, whereas the dorsolateral tegmental nucleus was observed to provide cholinergic input preferentially to anterior thalamic regions and rostral portions of the basal forebrain. The PMT cholinergic neurons showed a tendency to collateralize extensively.
Article
The lateral parabrachial nucleus (LPBN) has been shown to be anatomically linked to a number of forebrain nuclei and medullary structures implicated in the control of body fluid balance and cardiovascular regulation. Although these connections suggest a role for the LPBN in body fluid homeostasis, there is currently little or no physiological or behavioral data to support this notion. The purpose of the present series of experiments was to determine the importance of the ventrolateral region of the LPBN (VLLPBN) in the behavioral response to various thirst challenges. Rats with electrolytic lesions of the VLLPBN and control rats were studied after administration of angiotensin II (ANG II) (1.5 and 3.0 mg/kg), isoproterenol (30 and 100 micrograms/kg), polyethylene glycol (20%) and hypertonic saline (4 and 12%). It was found that rats with lesions drank more in response to ANG II and isoproterenol administration than did control animals.
Article
The utility of computers and computer graphics as aids in the study of nervous system architecture is growing. However, modern histologic, immunocytologic and biochemical methods for revealing the underlying microarchitecture and macroarchitecture of the nervous system yield data formats requiring disparate computer acquisition, analysis and display approaches, capable of spanning many orders of magnitude of scale. This paper describes the Image Graphics Laboratory data acquisition, processing and display system, whose various components and programs may be used singly or in concert to enable definition of various tissue properties at different levels of resolution and integration. Examples are given of the system's use in light microscopic two-dimensional and three-dimensional reconstructions, autoradiographic reconstructions, reconstructions from projected images, reconstructions of impregnated cells (e.g., whole neurons) and peripheral nerve image analysis.
Article
Our previous anatomical and electrophysiological studies demonstrated that first-order hepatic and gustatory afferents project to separate regions of the solitary nucleus (NST) and no intra-NST interaction of these two sensory systems could be demonstrated. However, iontophoretic injections of horseradish peroxidase into physiologically identified zones of the NST revealed that both of these regions send overlapping projections to the immediately subjacent parvocellular reticular formation as well as the postero-medial parabrachial nucleus (PBN). The present electrophysiological studies demonstrate that an interstitial zone of neurons in the caudal, medial PBN, indeed, receive convergent input from second-order gustatory and vagal afferents. Co-activation of these PBN units by the simultaneous arrival of both input sources frequently resulted in an additive interaction of evoked activity. PBN units lateral and caudal to this zone responded to vagal stimulation only, while units in the anterior and extreme medial portion of the PBN only responded to gustatory stimulation. By virtue of the efferent projections of the PBN, one might speculate that the convergence of information at this locus may, eventually, play a role in directing long term feeding behavior patterns such as learned taste aversion as well as the more transient changes in taste preference with visceral loading.
Article
Monoclonal antibodies to choline acetyltransferase and a histochemical method for the concurrent demonstration of acetylcholinesterase and horseradish peroxidase were used to investigate the organization of ascending cholinergic pathways in the central nervous system of the rat. The cortical mantle, the amygdaloid complex, the hippocampal formation, the olfactory bulb and the thalamic nuclei receive their cholinergic innervation principally, from cholinergic projection neurons of the basal forebrain and upper brainstem. On the basis of connectivity patterns, we subdivided these cholinergic neurons into six major sectors. The Ch1 and Ch2 sectors are contained within the medial septal nucleus and the vertical limb nucleus of the diagonal band, respectively. They provide the major cholinergic projections of the hippocampus. The Ch3 sector is contained mostly within the lateral portion of the horizontal limb nucleus of the diagonal band and provides the major cholinergic innervation to the olfactory bulb. The Ch4 sector includes cholinergic neurons in the nucleus basalis, and also within parts of the diagonal band nuclei. Neurons of the Ch4 sector provide the major cholinergic innervation of the cortical mantle and the amygdala. The Ch5-Ch6 sectors are contained mostly within the pedunculopontine nucleus of the pontomesencephalic reticular formation (Ch5) and within the laterodorsal tegmental gray of the periventricular area (Ch6). These sectors provide the major cholinergic innervation of the thalamus. The Ch5-Ch6 neurons also provide a minor component of the corticopetal cholinergic innervation. These central cholinergic pathways have been implicated in a variety of behaviors and especially in memory function. It appears that the age-related changes of memory function as well as some of the behavioral disturbances seen in the dementia of Alzheimer's Disease may be related to pathological alterations along central cholinergic pathways.
Article
In summary, we have demonstrated the subnuclear organization of PB, and correlated this with the origins of its efferent connections. In general, PBm projects primarily to the insular, infralimbic and lateral frontal cortex, and to associated areas in the thalamus, hypothalamus and amygdala. PBl chiefly innervates the autonomic nuclei of the hypothalamus and related portions of the amygdala and the bed nucleus of the stria terminalis. KF is the main source of descending projections from PB to the region of the nucleus of the solitary tract, the ventrolateral medulla and the intermediolateral cell column in the thoracic spinal cord. Further subnuclear organization of the origins of these projections within the major PB subdivisions has been described in detail. While PB afferents tend to terminate in specific subnuclei, one cannot reliably predict from the functional properties of the major inputs to a subnucleus what information will be carried in its efferents. Further anatomical and physiological studies of the input-output relationships of single PB neurons will be necessary to help resolve this enigma. However, recent immunohistochemical observations suggest that the subnuclear organization of PB afferent and efferent connections may reflect, at least in part, their biochemical specificity.
Article
Spontaneously firing units in the region of parabrachial nuclei (PB) and Kölliker-Fuse nuclei (KF) of 19 chloralose-anesthetized cats were monitored for changes in firing frequency during electrical stimulation of carotid sinus (CSN) and aortic depressor (ADN) nerves, of central nucleus of the amygdala (ACE), and of paraventricular nuclei of the hypothalamus (PVH). In the ipsilateral PB 64 of 189 and in the contralateral PB 9 of 103 units responded to CSN stimulation; 18 of 185 ipsilaterally and 7 of 97 contralaterally responded to ADN stimulation. Responses were primarily excitatory, and units were located primarily in the ventrolateral portion of the PB. Only 9 of 267 units responded to stimulation of both CSN and ADN. Stimulation of the ACE and PVH antidromically activated 9 and 7 units, respectively, in PB and approximately half of these also responded to buffer nerve stimulation. In the ipsilateral PB 56 of 207 and in the contralateral PB 11 of 103 units responded orthodromically to ACE stimulation, and 23 of 177 ipsilaterally and 2 of 103 contralaterally responded orthodromically to PVH stimulation with primarily excitatory responses and were located primarily in the ventrolateral portion of the PB and KF. Of these units approximately half also responded to buffer nerve stimulation. These results suggest an important role for PB-KF in mediating ascending and descending cardiovascular and respiratory control signals.
Article
The fiber connections of the nucleus tegmenti pedunculopontinus, pars compacta (TPc) were studied by means of the anterograde autoradiographic and retrograde horseradish peroxidase tracer methods. The limits of TPc, which is not a cytoarchitecturally distinct cell group in the cat, were estimated on the basis of autoradiographic experiments in which we plotted the distribution of afferent connections from the entopeduncular nucleus, motor cortex, substantia nigra, and subthalamic nucleus. We concluded that in the cat, TPc can most readily be identified by the terminal distribution of fibers originating in the substantia nigra. Deposits of radiolabel in the motor-pre-motor cortex, the entopeduncular nucleus, and subthalamic nucleus weakly labeled the same tegmental region. Parallel horseradish peroxidase experiments suggested that neurons near the entopeduncular nucleus, especially in the lateral hypothalamus and subthalamus, contribute to the pallido-TPc pathway; that entopeduncular neurons projecting to TPc are most numerous in the ventral part of the nucleus; and that of the neocortical motor fields, both areas 4 and 6 project to TPc.
Article
The efferent connections of the parabrachial nucleus have been analyzed in the rat using the anterograde autoradiographic method. Fibers originating from the lateral parabrachial nucleus (PBI) ascend in the periventricular system, the dorsal tegmental bundle and the central tegmental tract. The PBl projects to the dorsal raphe nucleus, the superior central raphe nucleus, and the Edinger-Westphal nucleus. It also innervates the intralaminar (centromedian, centrolateral, paracentral, parafascicular), the midline (paraventricular, reuniens), and the ventromedial basal (VMb) thalamic nuclei as well as much of the hypothalamus, including the dorsomedial, the ventromedial, the arcuate and the paraventricular nuclei, the lateral hypothalamic and the lateral preoptic areas. The PBl sends fibers via the ansa peduncularis into the amygdala, innervating the anterior, the central, the medial, the basomedial, and the posterior basolateral nuclei. In addition, it projects to the lateral part of the bed nucleus of the stria terminalis. Descending PBl fibers travel mainly through the ventrolateral medulla, passing through the region of the A1 and A5 catecholamine cell groups, the ventrolateral reticular formation and the region that contains parasympathetic preganglionic neurons. A small component travels in Probst's bundle to the ventral part of the nucleus of the solitary tract. Only a few PBl axons continue caudally into the lateral funiculus of the spinal cord, but these could not be followed beyond the first few cervical segments.
Article
The afferent projections to nucleus parabrachialis (NPB) and nearby pontine areas from the lower brainstem were studied in cats using retrograde horseradish peroxidase (HRP) and anterograde autoradiographic tracing techniques. Two groups of medullary neurons send major projections to NPB and the Kölliker‐Fuse nucleus (KF): (1) the solitary complex, especially the medial nucleus of the solitary tract (SM), nearby smaller cells of the dorsal motor nucleus of the vagus (DMV) and the commissural nucleus; and (2) the lateral tegmental field (FTL), or parvocellular reticular formation. Autoradiographic tracing from these areas demonstrated terminal fields in NPB/KF and emphasized a ventrolateral route to NPB from both sources, with axons ascending between the facial nerve and superior olive and passing rostral to the trigeminal nuclei. Minor projections to NPB/KF originate in the ventrolateral nucleus of the solitary tract, area subpostrema, the alaminar spinal trigeminal nucleus, the gigantocellular and magnocellular tegmental fields, and an area dorsal to the ipsilateral inferior olive. Topographical features of the major projections were studied by correlating the locus of overlap of injection sites with the locations of HRP‐positive cells. Medial areas of SM/DMV project mostly to medial parts of NPB, while lateral areas near the solitary tract project to lateral parts of NPB and KF. Crossing projections from SM/DMV favor dorsolateral NPB and KF. FTL neurons in dorsomedial areas project more to medial NPB, and ventrolateral areas project to lateral NPB/KF. Using a new coordinate system to locate and normalize the positions of FTL neurons, data from many brains were collated. FTL cells projecting to NPB/KF were found to be on discrete longitudinal sheets, running radially with respect to the fourth ventricle. This substructure and related evidence suggest a preferred pattern for neuroanatomical connections and information processing in the lateral reticular areas of the brainstem, and help in understanding the topography of the projections to NPB/KF.