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










Publication year range
1.
Cell Rep ; 37(5): 109936, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731609

ABSTRACT

Depression symptoms are often found in patients suffering from chronic pain, a phenomenon that is yet to be understood mechanistically. Here, we systematically investigate the cellular mechanisms and circuits underlying the chronic-pain-induced depression behavior. We show that the development of chronic pain is accompanied by depressive-like behaviors in a mouse model of trigeminal neuralgia. In parallel, we observe increased activity of the dopaminergic (DA) neuron in the midbrain ventral tegmental area (VTA), and inhibition of this elevated VTA DA neuron activity reverses the behavioral manifestations of depression. Further studies establish a pathway of glutamatergic projections from the spinal trigeminal subnucleus caudalis (Sp5C) to the lateral parabrachial nucleus (LPBN) and then to the VTA. These glutamatergic projections form a direct circuit that controls the development of the depression-like behavior under the state of the chronic neuropathic pain.


Subject(s)
Behavior, Animal , Chronic Pain/physiopathology , Depression/physiopathology , Parabrachial Nucleus/physiopathology , Trigeminal Neuralgia/physiopathology , Ventral Tegmental Area/physiopathology , Action Potentials , Animals , Chronic Pain/metabolism , Chronic Pain/psychology , Depression/metabolism , Depression/psychology , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/metabolism , Female , Glutamic Acid/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/metabolism , Neural Pathways/physiopathology , Parabrachial Nucleus/metabolism , Trigeminal Caudal Nucleus/metabolism , Trigeminal Caudal Nucleus/physiopathology , Trigeminal Neuralgia/metabolism , Trigeminal Neuralgia/psychology , Ventral Tegmental Area/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism
2.
Nat Neurosci ; 24(10): 1402-1413, 2021 10.
Article in English | MEDLINE | ID: mdl-34373644

ABSTRACT

Pain decreases the activity of many ventral tegmental area (VTA) dopamine (DA) neurons, yet the underlying neural circuitry connecting nociception and the DA system is not understood. Here we show that a subpopulation of lateral parabrachial (LPB) neurons is critical for relaying nociceptive signals from the spinal cord to the substantia nigra pars reticulata (SNR). SNR-projecting LPB neurons are activated by noxious stimuli and silencing them blocks pain responses in two different models of pain. LPB-targeted and nociception-recipient SNR neurons regulate VTA DA activity directly through feed-forward inhibition and indirectly by inhibiting a distinct subpopulation of VTA-projecting LPB neurons thereby reducing excitatory drive onto VTA DA neurons. Correspondingly, ablation of SNR-projecting LPB neurons is sufficient to reduce pain-mediated inhibition of DA release in vivo. The identification of a neural circuit conveying nociceptive input to DA neurons is critical to our understanding of how pain influences learning and behavior.


Subject(s)
Dopaminergic Neurons , Mesencephalon/physiopathology , Neural Pathways/physiopathology , Pain/physiopathology , Parabrachial Nucleus/physiopathology , Spinal Cord/physiopathology , Animals , Behavior, Animal , Brain Mapping , Male , Mice , Mice, Inbred C57BL , Neurons , Nociception , Optogenetics , Pain/psychology , Pain Management , Substantia Nigra/physiopathology , Ventral Tegmental Area/physiopathology
3.
J Neurophysiol ; 125(6): 2178-2190, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33909497

ABSTRACT

The chorda tympani is a gustatory nerve that fails to regenerate if sectioned in rats 10 days of age or younger. This early denervation causes an abnormally high preference for NH4Cl in adult rats, but the impact of neonatal chorda tympani transection on the development of the gustatory hindbrain is unclear. Here, we tested the effect of neonatal chorda tympani transection (CTX) on gustatory responses in the parabrachial nucleus (PbN). We recorded in vivo extracellular spikes in single PbN units of urethane-anesthetized adult rats following CTX at P5 (chronic CTX group) or immediately prior to recording (acute CTX group). Thus, all sampled PbN neurons received indirect input from taste nerves other than the CT. Compared to acute CTX rats, chronic CTX animals had significantly higher responses to stimulation with 0.1 and 0.5 M NH4Cl, 0.1 and 0.5 M NaCl, and 0.01 M citric acid. Activity to 0.5 M sucrose and 0.01 M quinine stimulation was not significantly different between groups. Neurons from chronic CTX animals also had larger interstimulus correlations and significantly higher entropy, suggesting that neurons in this group were more likely to be activated by stimulation with multiple tastants. Although neural responses were higher in the PbN of chronic CTX rats compared to acute-sectioned controls, taste-evoked activity was much lower than observed in previous reports, suggesting permanent deficits in taste signaling. These findings demonstrate that the developing gustatory hindbrain exhibits high functional plasticity following early nerve injury.NEW & NOTEWORTHY Early and chronic loss of taste input from the chorda tympani is associated with abnormal taste behaviors. We found that compared to when the chorda tympani is sectioned acutely, chronic nerve loss leads to amplification of spared inputs in the gustatory pons, with higher response to salty and sour stimuli. Findings point to plasticity that may compensate for sensory loss, but permanent deficits in taste signaling also occur following early denervation.


Subject(s)
Chorda Tympani Nerve/injuries , Neuronal Plasticity/physiology , Parabrachial Nucleus/physiopathology , Perceptual Disorders/physiopathology , Sensory Receptor Cells/physiology , Taste Perception/physiology , Taste/physiology , Action Potentials/physiology , Animals , Animals, Newborn , Denervation , Disease Models, Animal , Female , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
4.
Toxicol Appl Pharmacol ; 414: 115428, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33524449

ABSTRACT

Researches have shown that calcitonin gene-related peptide (CGRP) plays a pivotal role in pain modulation. Nociceptive information from the periphery is relayed from parabrachial nucleus (PBN) to brain regions implicated involved in pain. This study investigated the effects and mechanisms of CGRP and CGRP receptors in pain regulation in the PBN of naive and neuropathic pain rats. Chronic sciatic nerve ligation was used to model neuropathic pain, CGRP and CGRP 8-37 were injected into the PBN of the rats, and calcitonin receptor-like receptor (CLR), a main structure of CGRP receptor, was knocked down by lentivirus-coated CLR siRNA. The hot plate test (HPT) and the Randall Selitto Test (RST) was used to determine the latency of the rat hindpaw response. The expression of CLR was detected with RT-PCR and western blotting. We found that intra-PBN injecting of CGRP induced an obvious anti-nociceptive effect in naive and neuropathic pain rats in a dose-dependent manner, the CGRP-induced antinociception was significantly reduced after injection of CGRP 8-37, Moreover, the mRNA and protein levels of CLR, in PBN decreased significantly and the antinociception CGRP-induced was also significantly lower in neuropathic pain rats than that in naive rats. Knockdown CLR in PBN decreased the expression of CLR and the antinociception induced by CGRP was observably decreased. Our results demonstrate that CGRP induced antinociception in PBN of naive or neuropathic pain rats, CGRP receptor mediates this effect. Neuropathic pain induced decreases in the expression of CGRP receptor, as well as in CGRP-induced antinociception in PBN.


Subject(s)
Analgesics/pharmacology , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Receptor-Like Protein/agonists , Nociceptive Pain/prevention & control , Pain Threshold/drug effects , Parabrachial Nucleus/drug effects , Peptide Fragments/pharmacology , Receptors, Calcitonin Gene-Related Peptide/agonists , Sciatica/prevention & control , Animals , Calcitonin Receptor-Like Protein/genetics , Calcitonin Receptor-Like Protein/metabolism , Disease Models, Animal , Gene Expression Regulation , Male , Nociceptive Pain/genetics , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiopathology , Rats, Sprague-Dawley , Receptors, Calcitonin Gene-Related Peptide/genetics , Receptors, Calcitonin Gene-Related Peptide/metabolism , Sciatica/genetics , Sciatica/metabolism , Sciatica/physiopathology
5.
Nat Commun ; 11(1): 1729, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32265438

ABSTRACT

The TrkB receptor is critical for the control of energy balance, as mutations in its gene (NTRK2) lead to hyperphagia and severe obesity. The main neural substrate mediating the appetite-suppressing activity of TrkB, however, remains unknown. Here, we demonstrate that selective Ntrk2 deletion within paraventricular hypothalamus (PVH) leads to severe hyperphagic obesity. Furthermore, chemogenetic activation or inhibition of TrkB-expressing PVH (PVHTrkB) neurons suppresses or increases food intake, respectively. PVHTrkB neurons project to multiple brain regions, including ventromedial hypothalamus (VMH) and lateral parabrachial nucleus (LPBN). We find that PVHTrkB neurons projecting to LPBN are distinct from those to VMH, yet Ntrk2 deletion in PVH neurons projecting to either VMH or LPBN results in hyperphagia and obesity. Additionally, TrkB activation with BDNF increases firing of these PVH neurons. Therefore, TrkB signaling is a key regulator of a previously uncharacterized neuronal population within the PVH that impinges upon multiple circuits to govern appetite.


Subject(s)
Hyperphagia/metabolism , Membrane Glycoproteins/metabolism , Neurons/metabolism , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Appetite/genetics , Feeding Behavior/physiology , Female , Hyperphagia/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/genetics , Parabrachial Nucleus/cytology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiopathology , Protein-Tyrosine Kinases/genetics , Ventromedial Hypothalamic Nucleus/cytology , Ventromedial Hypothalamic Nucleus/metabolism
6.
J Neurosci ; 40(17): 3424-3442, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32217613

ABSTRACT

The parabrachial (PB) complex mediates both ascending nociceptive signaling and descending pain modulatory information in the affective/emotional pain pathway. We have recently reported that chronic pain is associated with amplified activity of PB neurons in a rat model of neuropathic pain. Here we demonstrate that similar activity amplification occurs in mice, and that this is related to suppressed inhibition to lateral parabrachial (LPB) neurons from the CeA in animals of either sex. Animals with pain after chronic constriction injury of the infraorbital nerve (CCI-Pain) displayed higher spontaneous and evoked activity in PB neurons, and a dramatic increase in after-discharges, responses that far outlast the stimulus, compared with controls. LPB neurons in CCI-Pain animals showed a reduction in inhibitory, GABAergic inputs. We show that, in both rats and mice, LPB contains few GABAergic neurons, and that most of its GABAergic inputs arise from CeA. These CeA GABA neurons express dynorphin, somatostatin, and/or corticotropin releasing hormone. We find that the efficacy of this CeA-LPB pathway is suppressed in chronic pain. Further, optogenetically stimulating this pathway suppresses acute pain, and inhibiting it, in naive animals, evokes pain behaviors. These findings demonstrate that the CeA-LPB pathway is critically involved in pain regulation, and in the pathogenesis of chronic pain.SIGNIFICANCE STATEMENT We describe a novel pathway, consisting of inhibition by dynorphin, somatostatin, and corticotropin-releasing hormone-expressing neurons in the CeA that project to the parabrachial nucleus. We show that this pathway regulates the activity of pain-related neurons in parabrachial nucleus, and that, in chronic pain, this inhibitory pathway is suppressed, and that this suppression is causally related to pain perception. We propose that this amygdalo-parabrachial pathway is a key regulator of both chronic and acute pain, and a novel target for pain relief.


Subject(s)
Amygdala/physiopathology , Chronic Pain/physiopathology , Neuralgia/physiopathology , Pain Perception/physiology , Parabrachial Nucleus/physiopathology , Action Potentials/physiology , Animals , Disease Models, Animal , Female , Male , Mice , Neural Pathways/physiopathology , Neurons/physiology , Pain Measurement , Pain Threshold/physiology
7.
Pain ; 161(1): 166-176, 2020 01.
Article in English | MEDLINE | ID: mdl-31479066

ABSTRACT

Dissecting the organization of circuit pathways involved in pain affect is pivotal for understanding behavior associated with noxious sensory inputs. The central nucleus of the amygdala (CeA) comprises distinct populations of inhibitory GABAergic neurons expressing a wide range of molecular markers. CeA circuits are associated with aversive learning and nociceptive responses. The CeA receives nociceptive signals directly from the parabrachial nucleus (PBn), contributing to the affective and emotional aspects of pain. Although the CeA has emerged as an important node in pain processing, key questions remain regarding the specific targeting of PBn inputs to different CeA subregions and cell types. We used a multifaceted approach involving transgenic reporter mice, viral vector-mediated optogenetics, and brain slice electrophysiology to delineate cell-type-specific functional organization of the PBn-CeA pathway. Whole-cell patch clamp recordings of molecularly defined CeA neurons while optogenetically driving long-range inputs originating from PBn revealed the direct monosynaptic excitatory inputs from PBn neurons to 3 major subdivisions of the CeA: laterocapsular (CeC), lateral (CeL), and medial (CeM). Direct monosynaptic excitatory inputs from PBn targeted both somatostatin-expressing (SOM+) and corticotropin-releasing hormone expressing (CRH+) neurons in the CeA. We find that monosynaptic PBn input is preferentially organized to molecularly specific neurons in distinct subdivisions of the CeA. The spared nerve injury model of neuropathic pain differentially altered PBn monosynaptic excitatory input to CeA neurons based on molecular identity and topographical location within the CeA. These results provide insight into the functional organization of affective pain pathways and how they are altered by chronic pain.


Subject(s)
Central Amygdaloid Nucleus/physiopathology , Neuralgia/physiopathology , Neurons/physiology , Parabrachial Nucleus/physiopathology , Peripheral Nerve Injuries/physiopathology , Animals , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Male , Mice , Mice, Transgenic , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neuralgia/metabolism , Neurons/metabolism , Parabrachial Nucleus/metabolism , Patch-Clamp Techniques , Peripheral Nerve Injuries/metabolism , Somatostatin/metabolism
8.
J Neurosci ; 39(42): 8225-8230, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619491

ABSTRACT

The parabrachial nucleus (PBN) has long been recognized as a sensory relay receiving an array of interoceptive and exteroceptive inputs relevant to taste and ingestive behavior, pain, and multiple aspects of autonomic control, including respiration, blood pressure, water balance, and thermoregulation. Outputs are known to be similarly widespread and complex. How sensory information is handled in PBN and used to inform different outputs to maintain homeostasis and promote survival is only now being elucidated. With a focus on taste and ingestive behaviors, pain, and thermoregulation, this review is intended to provide a context for analysis of PBN circuits involved in aversion and avoidance, and consider how information of various modalities, interoceptive and exteroceptive, is processed within PBN and transmitted to distinct targets to signal challenge, and to engage appropriate behavioral and physiological responses to maintain homeostasis.


Subject(s)
Body Temperature Regulation/physiology , Nociception/physiology , Pain/physiopathology , Parabrachial Nucleus/physiology , Taste/physiology , Animals , Humans , Neural Pathways/physiology , Neural Pathways/physiopathology , Neurons/physiology , Parabrachial Nucleus/physiopathology
9.
J Neurosci ; 39(11): 2065-2079, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30651329

ABSTRACT

There is strong evidence that spinoparabrachial neurons in the superficial dorsal horn contribute to persistent pain states, and that the lateral parabrachial complex (PB) conveys relevant nociceptive information to higher structures. The role of PB itself in hyperalgesia and how it recruits descending facilitation has nevertheless received significantly less attention. The current study is a first step toward delineating the functional dynamics of PB and its link to descending control in acute and persistent inflammatory pain. In lightly anesthetized rats, we recorded behavioral withdrawal evoked by mechanical stimulation of the hindpaw and, simultaneously, the activity of identified pain-modulating neurons, "ON-cells" and "OFF-cells," in the rostral ventromedial medulla (RVM). This was done before and after the inactivation of PB, contralateral or ipsilateral to an inflamed paw [1 h, 1 d, or 5-6 d after intraplantar injection of Complete Freund's Adjuvant (CFA)]. The inactivation of contralateral, but not ipsilateral, PB interfered with nociceptive input to RVM under basal conditions, as well as in acute inflammation. By contrast, blocking ipsilateral, but not contralateral, PB in established inflammation interfered with behavioral hyperalgesia and ON-cell and OFF-cell responses. The lesioning of contralateral PB before CFA injection prevented this recruitment of ipsilateral PB in persistent inflammation. These experiments show that contralateral PB is required to initiate hyperalgesia, which is then maintained by ipsilateral PB, most likely in both cases via the engagement of pain-modulating neurons of the RVM.SIGNIFICANCE STATEMENT The lateral parabrachial complex (PB) relays nociceptive information to brain circuits that are important for the transmission and modulation of pain, but its specific role in persistent pain and engagement of descending control mechanisms has received relatively little attention. We show here that PB contralateral and ipsilateral to an inflammatory insult demonstrate different functions as inflammation persists, likely by engaging pain-facilitating neurons of the rostral ventromedial medulla. While the contralateral PB, the target of the major spinoparabrachial pathway, relays acute nociceptive information, the ipsilateral PB is recruited or unmasked in persistent inflammation to maintain hyperalgesia. These data point to plasticity in the PB itself or its direct and indirect connections with pain-modulating systems as central to the development and maintenance of persistent pain.


Subject(s)
Inflammation/physiopathology , Neuronal Plasticity/physiology , Neurons/physiology , Nociception/physiology , Pain/physiopathology , Parabrachial Nucleus/physiopathology , Animals , Hyperalgesia/complications , Hyperalgesia/physiopathology , Inflammation/complications , Male , Neural Pathways/physiology , Pain/complications , Pain Threshold , Rats, Sprague-Dawley
10.
Elife ; 72018 08 28.
Article in English | MEDLINE | ID: mdl-30149836

ABSTRACT

Pain is a prevalent symptom of Parkinson's disease, and is effectively treated by deep brain stimulation of the subthalamic nucleus (STN). However, the link between pain and the STN remains unclear. In the present work, using in vivo electrophysiology in rats, we report that STN neurons exhibit complex tonic and phasic responses to noxious stimuli. We also show that nociception is altered following lesions of the STN, and characterize the role of the superior colliculus and the parabrachial nucleus in the transmission of nociceptive information to the STN, physiologically from both structures and anatomically in the case of the parabrachial nucleus. We show that STN nociceptive responses are abnormal in a rat model of PD, suggesting their dependence on the integrity of the nigrostriatal dopaminergic system. The STN-linked nociceptive network that we reveal is likely to be of considerable clinical importance in neurological diseases involving a dysfunction of the basal ganglia.


Subject(s)
Nerve Net/physiopathology , Nociception/physiology , Pain/physiopathology , Subthalamic Nucleus/physiopathology , Animals , Biomarkers/metabolism , Disease Models, Animal , Electrophysiological Phenomena , Male , Parabrachial Nucleus/physiopathology , Parkinson Disease/physiopathology , Reaction Time , Superior Colliculi
11.
Science ; 357(6352): 695-699, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28818946

ABSTRACT

Although itch sensation is an important protective mechanism for animals, chronic itch remains a challenging clinical problem. Itch processing has been studied extensively at the spinal level. However, how itch information is transmitted to the brain and what central circuits underlie the itch-induced scratching behavior remain largely unknown. We found that the spinoparabrachial pathway was activated during itch processing and that optogenetic suppression of this pathway impaired itch-induced scratching behaviors. Itch-mediating spinal neurons, which express the gastrin-releasing peptide receptor, are disynaptically connected to the parabrachial nucleus via glutamatergic spinal projection neurons. Blockade of synaptic output of glutamatergic neurons in the parabrachial nucleus suppressed pruritogen-induced scratching behavior. Thus, our studies reveal a central neural circuit that is critical for itch signal processing.


Subject(s)
Nerve Net/physiopathology , Parabrachial Nucleus/physiopathology , Pruritus/physiopathology , Sensation/physiology , Spinal Cord/physiopathology , Animals , Chronic Disease , Glutamates/metabolism , Male , Mice , Mice, Inbred C57BL , Optogenetics , Parabrachial Nucleus/cytology , Receptors, Bombesin/genetics , Receptors, Bombesin/metabolism , Sensation/genetics , Spinal Cord/metabolism , Vesicular Glutamate Transport Protein 2/genetics
12.
J Neurosci ; 37(36): 8678-8687, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28821663

ABSTRACT

To maintain energy homeostasis, orexigenic (appetite-inducing) and anorexigenic (appetite suppressing) brain systems functionally interact to regulate food intake. Within the hypothalamus, neurons that express agouti-related protein (AgRP) sense orexigenic factors and orchestrate an increase in food-seeking behavior. In contrast, calcitonin gene-related peptide (CGRP)-expressing neurons in the parabrachial nucleus (PBN) suppress feeding. PBN CGRP neurons become active in response to anorexigenic hormones released following a meal, including amylin, secreted by the pancreas, and cholecystokinin (CCK), secreted by the small intestine. Additionally, exogenous compounds, such as lithium chloride (LiCl), a salt that creates gastric discomfort, and lipopolysaccharide (LPS), a bacterial cell wall component that induces inflammation, exert appetite-suppressing effects and activate PBN CGRP neurons. The effects of increasing the homeostatic drive to eat on feeding behavior during appetite suppressing conditions are unknown. Here, we show in mice that food deprivation or optogenetic activation of AgRP neurons induces feeding to overcome the appetite suppressing effects of amylin, CCK, and LiCl, but not LPS. AgRP neuron photostimulation can also increase feeding during chemogenetic-mediated stimulation of PBN CGRP neurons. AgRP neuron stimulation reduces Fos expression in PBN CGRP neurons across all conditions. Finally, stimulation of projections from AgRP neurons to the PBN increases feeding following administration of amylin, CCK, and LiCl, but not LPS. These results demonstrate that AgRP neurons are sufficient to increase feeding during noninflammatory-based appetite suppression and to decrease activity in anorexigenic PBN CGRP neurons, thereby increasing food intake during homeostatic need.SIGNIFICANCE STATEMENT The motivation to eat depends on the relative balance of activity in distinct brain regions that induce or suppress appetite. An abnormal amount of activity in neurons that induce appetite can cause obesity, whereas an abnormal amount of activity in neurons that suppress appetite can cause malnutrition and a severe reduction in body weight. The purpose of this study was to determine whether a population of neurons known to induce appetite ("AgRP neurons") could induce food intake to overcome appetite-suppression following administration of various appetite-suppressing compounds. We found that stimulating AgRP neurons could overcome various forms of appetite suppression and decrease neural activity in a separate population of appetite-suppressing neurons, providing new insights into how the brain regulates food intake.


Subject(s)
Agouti-Related Protein/metabolism , Anorexia/physiopathology , Appetite Regulation , Eating , Neural Inhibition , Neurons/metabolism , Parabrachial Nucleus/physiopathology , Agouti-Related Protein/genetics , Animals , Anorexia/pathology , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Parabrachial Nucleus/pathology
13.
eNeuro ; 4(3)2017.
Article in English | MEDLINE | ID: mdl-28660248

ABSTRACT

The parabrachial complex (PB) is a functionally and anatomically complex structure involved in a range of homeostatic and sensory functions, including nociceptive transmission. There is also evidence that PB can engage descending pain-modulating systems, the best characterized of which is the rostral ventromedial medulla (RVM). Two distinct classes of RVM neurons, "ON-cells" and "OFF-cells," exert net pronociceptive and anti-nociceptive effects, respectively. PB was recently shown to be a relay of nociceptive information to RVM ON- and OFF-cells. The present experiments used optogenetic methods in a lightly anesthetized rat and an adult RVM slice to determine whether there are direct, functionally relevant inputs to RVM pain-modulating neurons from PB. Whole-cell patch-clamp recordings demonstrated that PB conveys direct glutamatergic and GABAergic inputs to RVM neurons. Consistent with this, in vivo recording showed that nociceptive-evoked responses of ON- and OFF-cells were suppressed by optogenetic inactivation of archaerhodopsin (ArchT)-expressing PB terminals in RVM, demonstrating that a net inhibitory input to OFF-cells and net excitatory input to ON-cells are engaged by acute noxious stimulation. Further, the majority of ON- and OFF-cells responded to optogenetic activation of channelrhodopsin (ChR2)-expressing terminals in the RVM, confirming a direct PB influence on RVM pain-modulating neurons. These data show that a direct connection from the PB to the RVM conveys nociceptive information to the pain-modulating neurons of RVM under basal conditions. They also reveal additional inputs from PB with the capacity to activate both classes of RVM pain-modulating neurons and the potential to be recruited under different physiological and pathophysiological conditions.


Subject(s)
Medulla Oblongata/physiopathology , Neurons/physiology , Nociceptive Pain/physiopathology , Pain Perception/physiology , Parabrachial Nucleus/physiopathology , Action Potentials , Animals , Glutamic Acid/metabolism , Male , Medulla Oblongata/pathology , Neural Pathways/pathology , Neural Pathways/physiopathology , Neurons/pathology , Nociceptive Pain/pathology , Optogenetics , Parabrachial Nucleus/pathology , Patch-Clamp Techniques , Rats, Sprague-Dawley , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
14.
Nat Commun ; 7: 11905, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27301688

ABSTRACT

The central nervous system controls food consumption to maintain metabolic homoeostasis. In response to a meal, visceral signals from the gut activate neurons in the nucleus of the solitary tract (NTS) via the vagus nerve. These NTS neurons then excite brain regions known to mediate feeding behaviour, such as the lateral parabrachial nucleus (PBN). We previously described a neural circuit for appetite suppression involving calcitonin gene-related protein (CGRP)-expressing PBN (CGRP(PBN)) neurons; however, the molecular identity of the inputs to these neurons was not established. Here we identify cholecystokinin (CCK) and noradrenergic, dopamine ß-hydroxylase (DBH)-expressing NTS neurons as two separate populations that directly excite CGRP(PBN) neurons. When these NTS neurons are activated using optogenetic or chemogenetic methods, food intake decreases and with chronic stimulation mice lose body weight. Our optogenetic results reveal that CCK and DBH neurons in the NTS directly engage CGRP(PBN) neurons to promote anorexia.


Subject(s)
Anorexia/genetics , Anorexia/physiopathology , Neural Pathways/physiopathology , Parabrachial Nucleus/physiopathology , Solitary Nucleus/physiopathology , Action Potentials , Animals , Anxiety/physiopathology , Calcitonin Gene-Related Peptide/metabolism , Cholecystokinin/metabolism , Dopamine beta-Hydroxylase/metabolism , Eating , Mice, Inbred C57BL , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism
15.
Int J Obes (Lond) ; 40(6): 921-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26984418

ABSTRACT

BACKGROUND/OBJECTIVE: Obesity and metabolic diseases are at an alarming level globally and increasingly affect children and adolescents. Gastric bypass and other bariatric surgeries have proven remarkably successful and are increasingly performed worldwide. Reduced desire to eat and changes in eating behavior and food choice account for most of the initial weight loss and diabetes remission after surgery, but the underlying mechanisms of altered gut-brain communication are unknown. SUBJECTS/METHODS: To explore the potential involvement of a powerful brainstem anorexia pathway centered around the lateral parabrachial nucleus (lPBN), we measured meal-induced neuronal activation by means of c-Fos immunohistochemistry in a new high-fat diet-induced obese mouse model of Roux-en-Y gastric bypass (RYGB) at 10 and 40 days after RYGB or sham surgery. RESULTS: Voluntary ingestion of a meal 10 days after RYGB, but not after sham surgery, strongly and selectively activates calcitonin gene-related peptide neurons in the external lPBN as well as neurons in the nucleus tractus solitarius, area postrema and medial amygdala. At 40 days after surgery, meal-induced activation in all these areas was greatly diminished and did not reach statistical significance. CONCLUSIONS: The neural activation pattern and dynamics suggest a role of the brainstem anorexia pathway in the early effects of RYGB on meal size and food intake that may lead to adaptive neural and behavioral changes involved in the control of food intake and body weight at a lower level. However, selective inhibition of this pathway will be required for a more causal implication.


Subject(s)
Anorexia/physiopathology , Disease Models, Animal , Eating , Feeding Behavior , Gastric Bypass , Parabrachial Nucleus/physiopathology , Animals , Diet, High-Fat , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Obesity/pathology , Obesity/surgery , Satiety Response
16.
J Neurophysiol ; 115(6): 2721-39, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26888105

ABSTRACT

A large majority of neurons in the superficial layer of the dorsal horn projects to the lateral parabrachial nucleus (LPB). LPB neurons then project to the capsular part of the central amygdala (CeA; CeC), a key structure underlying the nociception-emotion link. LPB-CeC synaptic transmission is enhanced in various pain models by using electrical stimulation of putative fibers of LPB origin in brain slices. However, this approach has limitations for examining direct monosynaptic connections devoid of directly stimulating fibers from other structures and local GABAergic neurons. To overcome these limitations, we infected the LPB of rats with an adeno-associated virus vector expressing channelrhodopsin-2 and prepared coronal and horizontal brain slices containing the amygdala. We found that blue light stimulation resulted in monosynaptic excitatory postsynaptic currents (EPSCs), with very small latency fluctuations, followed by a large polysynaptic inhibitory postsynaptic current in CeC neurons, regardless of the firing pattern type. Intraplantar formalin injection at 24 h before slice preparation significantly increased EPSC amplitude in late firing-type CeC neurons. These results indicate that direct monosynaptic glutamatergic inputs from the LPB not only excite CeC neurons but also regulate CeA network signaling through robust feed-forward inhibition, which is under plastic modulation in response to persistent inflammatory pain.


Subject(s)
Central Amygdaloid Nucleus/physiopathology , Neurons/physiology , Nociceptive Pain/physiopathology , Parabrachial Nucleus/physiopathology , Synapses/physiology , Action Potentials/physiology , Animals , Central Amygdaloid Nucleus/pathology , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/physiology , Male , Neural Pathways/pathology , Neural Pathways/physiopathology , Neuronal Plasticity/physiology , Neurons/pathology , Nociceptive Pain/pathology , Parabrachial Nucleus/pathology , Rats, Wistar , Synapses/pathology , Tissue Culture Techniques
18.
Am J Physiol Regul Integr Comp Physiol ; 310(1): R41-54, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26491097

ABSTRACT

The parabrachial nucleus is important for thermoregulation because it relays skin temperature information from the spinal cord to the hypothalamus. Prior work in rats localized thermosensory relay neurons to its lateral subdivision (LPB), but the genetic and neurochemical identity of these neurons remains unknown. To determine the identity of LPB thermosensory neurons, we exposed mice to a warm (36°C) or cool (4°C) ambient temperature. Each condition activated neurons in distinct LPB subregions that receive input from the spinal cord. Most c-Fos+ neurons in these LPB subregions expressed the transcription factor marker FoxP2. Consistent with prior evidence that LPB thermosensory relay neurons are glutamatergic, all FoxP2+ neurons in these subregions colocalized with green fluorescent protein (GFP) in reporter mice for Vglut2, but not for Vgat. Prodynorphin (Pdyn)-expressing neurons were identified using a GFP reporter mouse and formed a caudal subset of LPB FoxP2+ neurons, primarily in the dorsal lateral subnucleus (PBdL). Warm exposure activated many FoxP2+ neurons within PBdL. Half of the c-Fos+ neurons in PBdL were Pdyn+, and most of these project into the preoptic area. Cool exposure activated a separate FoxP2+ cluster of neurons in the far-rostral LPB, which we named the rostral-to-external lateral subnucleus (PBreL). These findings improve our understanding of LPB organization and reveal that Pdyn-IRES-Cre mice provide genetic access to warm-activated, FoxP2+ glutamatergic neurons in PBdL, many of which project to the hypothalamus.


Subject(s)
Fever/metabolism , Hypothermia/metabolism , Neurons/metabolism , Parabrachial Nucleus/metabolism , Skin Temperature , Thermosensing , Animals , Disease Models, Animal , Enkephalins/genetics , Enkephalins/metabolism , Fever/genetics , Fever/physiopathology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Genes, Reporter , Genotype , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothermia/genetics , Hypothermia/physiopathology , Integrases/genetics , Integrases/metabolism , Internal Ribosome Entry Sites , Male , Mice, Transgenic , Neuroanatomical Tract-Tracing Techniques , Parabrachial Nucleus/physiopathology , Phenotype , Protein Precursors/genetics , Protein Precursors/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Repressor Proteins/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
19.
J Neurophysiol ; 113(1): 58-70, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25298386

ABSTRACT

We tested the possibility that the trigeminoparabrachial tract (VcPbT), a projection thought to be importantly involved in nociception, might also contribute to sensation of itch. In anesthetized rats, 47 antidromically identified VcPbT neurons with receptive fields involving the cheek were characterized for their responses to graded mechanical and thermal stimuli and intradermal injections of pruritogens (serotonin, chloroquine, and ß-alanine), partial pruritogens (histamine and capsaicin), and an algogen (mustard oil). All pruriceptive VcPbT neurons were responsive to mechanical stimuli, and more than half were additionally responsive to thermal stimuli. The majority of VcPbT neurons were activated by injections of serotonin, histamine, capsaicin, and/or mustard oil. A subset of neurons were inhibited by injection of chloroquine. The large majority of VcPbT neurons projected to the ipsilateral and/or contralateral external lateral parabrachial and Kölliker-Fuse nuclei, as evidenced by antidromic mapping techniques. Analyses of mean responses and spike-timing dynamics of VcPbT neurons suggested clear differences in firing rates between responses to noxious and pruritic stimuli. Comparisons between the present data and those previously obtained from trigeminothalamic tract (VcTT) neurons demonstrated several differences in responses to some pruritogens. For example, responses of VcPbT neurons to injection of serotonin often endured for nearly an hour and showed a delayed peak in discharge rate. In contrast, responses of VcTT neurons endured for roughly 20 min and no delayed peak of firing was noted. Thus the longer duration responses to 5-HT and the delay in peak firing of VcPbT neurons better matched behavioral responses to stimulation in awake rats than did those of VcTT neurons. The results indicate that VcPbT neurons may have important roles in the signaling of itch as well as pain.


Subject(s)
Nociceptive Pain/physiopathology , Parabrachial Nucleus/physiopathology , Pruritus/physiopathology , Sensory Receptor Cells/physiology , Trigeminal Nerve/physiopathology , Action Potentials , Animals , Capsaicin , Cheek/physiopathology , Chloroquine , Histamine , Hot Temperature , Male , Mustard Plant , Neural Pathways/cytology , Neural Pathways/physiopathology , Nociceptive Pain/pathology , Parabrachial Nucleus/cytology , Physical Stimulation , Plant Oils , Pruritus/pathology , Rats, Sprague-Dawley , Sensory Receptor Cells/cytology , Serotonin , Touch , Trigeminal Nerve/cytology , beta-Alanine
20.
Neuropharmacology ; 86: 38-48, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24998751

ABSTRACT

The intricate relationships that associate pain, stress responses and emotional behavior have been well established. Acute stressful situations can decrease nociceptive sensations and conversely, chronic pain can enhance other pain experiences and heighten the emotional and behavioral consequences of stress. Accordingly, chronic pain is comorbid with a number of behavioral disorders including depression, anxiety abnormalities and associated stress-related disorders including post traumatic stress disorder (PTSD). The central nucleus of the amygdala (CeA) represents a convergence of pathways for pain, stress and emotion, and we have identified pituitary adenylate cyclase activating polypeptide (PACAP) immunoreactivity in fiber elements in the lateral capsular division of the CeA (CeLC). The PACAP staining patterns colocalized in part with those for calcitonin gene related peptide (CGRP); anterograde fiber tracing and excitotoxic lesion studies demonstrated that the CeLC PACAP/CGRP immunoreactivities represented sensory fiber projections from the lateral parabrachial nucleus (LPBn) along the spino-parabrachioamygdaloid tract. The same PBn PACAP/CGRP fiber system also projected to the BNST. As in the BNST, CeA PACAP signaling increased anxiety-like behaviors accompanied by weight loss and decreased feeding. But in addition to heightened anxiety-like responses, CeA PACAP signaling also altered nociception as reflected by decreased latency and threshold responses in thermal and mechanical sensitivity tests, respectively. From PACAP expression in major pain pathways, the current observations are novel and suggest that CeA PACAP nociceptive signaling and resulting neuroplasticity via the spino-parabrachioamygdaloid tract may represent mechanisms that associate chronic pain with sensory hypersensitivity, fear memory consolidation and severe behavioral disorders.


Subject(s)
Amygdala/physiopathology , Pain/physiopathology , Parabrachial Nucleus/physiopathology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Anxiety/physiopathology , Calcitonin Gene-Related Peptide/metabolism , Chronic Disease , Disease Models, Animal , Eating/physiology , Male , Neural Pathways/physiopathology , Nociception/physiology , Pain Threshold/physiology , Random Allocation , Rats, Sprague-Dawley , Stress, Psychological , Weight Loss/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...