Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 784
Filter
1.
Nature ; 631(8021): 601-609, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987587

ABSTRACT

Exaggerated airway constriction triggered by repeated exposure to allergen, also called hyperreactivity, is a hallmark of asthma. Whereas vagal sensory neurons are known to function in allergen-induced hyperreactivity1-3, the identity of downstream nodes remains poorly understood. Here we mapped a full allergen circuit from the lung to the brainstem and back to the lung. Repeated exposure of mice to inhaled allergen activated the nuclei of solitary tract (nTS) neurons in a mast cell-, interleukin-4 (IL-4)- and vagal nerve-dependent manner. Single-nucleus RNA sequencing, followed by RNAscope assay at baseline and allergen challenges, showed that a Dbh+ nTS population is preferentially activated. Ablation or chemogenetic inactivation of Dbh+ nTS neurons blunted hyperreactivity whereas chemogenetic activation promoted it. Viral tracing indicated that Dbh+ nTS neurons project to the nucleus ambiguus (NA) and that NA neurons are necessary and sufficient to relay allergen signals to postganglionic neurons that directly drive airway constriction. Delivery of noradrenaline antagonists to the NA blunted hyperreactivity, suggesting noradrenaline as the transmitter between Dbh+ nTS and NA. Together, these findings provide molecular, anatomical and functional definitions of key nodes of a canonical allergen response circuit. This knowledge informs how neural modulation could be used to control allergen-induced airway hyperreactivity.


Subject(s)
Allergens , Brain Stem , Bronchial Hyperreactivity , Dopamine beta-Hydroxylase , Lung , Neurons , Animals , Female , Male , Mice , Allergens/immunology , Asthma/immunology , Asthma/physiopathology , Brain Stem/cytology , Brain Stem/physiology , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Interleukin-4/immunology , Lung/drug effects , Lung/immunology , Lung/innervation , Lung/physiopathology , Mast Cells/immunology , Neurons/enzymology , Neurons/physiology , Norepinephrine/antagonists & inhibitors , Norepinephrine/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Vagus Nerve/cytology , Vagus Nerve/physiology , Medulla Oblongata/cytology , Medulla Oblongata/drug effects , Ganglia, Autonomic/cytology , Dopamine beta-Hydroxylase/metabolism
2.
Nature ; 632(8025): 585-593, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987598

ABSTRACT

The most successful obesity therapeutics, glucagon-like peptide-1 receptor (GLP1R) agonists, cause aversive responses such as nausea and vomiting1,2, effects that may contribute to their efficacy. Here, we investigated the brain circuits that link satiety to aversion, and unexpectedly discovered that the neural circuits mediating these effects are functionally separable. Systematic investigation across drug-accessible GLP1R populations revealed that only hindbrain neurons are required for the efficacy of GLP1-based obesity drugs. In vivo two-photon imaging of hindbrain GLP1R neurons demonstrated that most neurons are tuned to either nutritive or aversive stimuli, but not both. Furthermore, simultaneous imaging of hindbrain subregions indicated that area postrema (AP) GLP1R neurons are broadly responsive, whereas nucleus of the solitary tract (NTS) GLP1R neurons are biased towards nutritive stimuli. Strikingly, separate manipulation of these populations demonstrated that activation of NTSGLP1R neurons triggers satiety in the absence of aversion, whereas activation of APGLP1R neurons triggers strong aversion with food intake reduction. Anatomical and behavioural analyses revealed that NTSGLP1R and APGLP1R neurons send projections to different downstream brain regions to drive satiety and aversion, respectively. Importantly, GLP1R agonists reduce food intake even when the aversion pathway is inhibited. Overall, these findings highlight NTSGLP1R neurons as a population that could be selectively targeted to promote weight loss while avoiding the adverse side effects that limit treatment adherence.


Subject(s)
Anti-Obesity Agents , Avoidance Learning , Glucagon-Like Peptide-1 Receptor , Neural Pathways , Rhombencephalon , Satiety Response , Animals , Female , Male , Mice , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/pharmacology , Area Postrema/metabolism , Area Postrema/drug effects , Avoidance Learning/drug effects , Avoidance Learning/physiology , Eating/drug effects , Eating/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Mice, Inbred C57BL , Neural Pathways/drug effects , Neurons/metabolism , Neurons/physiology , Neurons/drug effects , Obesity/metabolism , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Rhombencephalon/physiology , Satiety Response/drug effects , Satiety Response/physiology , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Food
3.
CNS Neurosci Ther ; 30(6): e14808, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887205

ABSTRACT

OBJECTIVE: Phenylethanolamine N-methyltransferase (PNMT)-expressing neurons in the nucleus tractus solitarii (NTS) contribute to the regulation of autonomic functions. However, the neural circuits linking these neurons to other brain regions remain unclear. This study aims to investigate the connectivity mechanisms of the PNMT-expressing neurons in the NTS (NTSPNMT neurons). METHODS: The methodologies employed in this study included a modified rabies virus-based retrograde neural tracing technique, conventional viral anterograde tracing, and immunohistochemical staining procedures. RESULTS: A total of 43 upstream nuclei projecting to NTSPNMT neurons were identified, spanning several key brain regions including the medulla oblongata, pons, midbrain, cerebellum, diencephalon, and telencephalon. Notably, dense projections to the NTSPNMT neurons were observed from the central amygdaloid nucleus, paraventricular nucleus of the hypothalamus, area postrema, and the gigantocellular reticular nucleus. In contrast, the ventrolateral medulla, lateral parabrachial nucleus, and lateral hypothalamic area were identified as the primary destinations for axon terminals originating from NTSPNMT neurons. Additionally, reciprocal projections were evident among 21 nuclei, primarily situated within the medulla oblongata. CONCLUSION: Our research findings demonstrate that NTSPNMT neurons form extensive connections with numerous nuclei, emphasizing their essential role in the homeostatic regulation of vital autonomic functions.


Subject(s)
Neurons , Phenylethanolamine N-Methyltransferase , Solitary Nucleus , Animals , Phenylethanolamine N-Methyltransferase/metabolism , Phenylethanolamine N-Methyltransferase/genetics , Solitary Nucleus/enzymology , Solitary Nucleus/metabolism , Solitary Nucleus/cytology , Neurons/metabolism , Neurons/enzymology , Male , Efferent Pathways/enzymology , Afferent Pathways/enzymology , Rats, Sprague-Dawley , Brain Mapping/methods , Rats
4.
Nature ; 624(7990): 130-137, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37993711

ABSTRACT

The termination of a meal is controlled by dedicated neural circuits in the caudal brainstem. A key challenge is to understand how these circuits transform the sensory signals generated during feeding into dynamic control of behaviour. The caudal nucleus of the solitary tract (cNTS) is the first site in the brain where many meal-related signals are sensed and integrated1-4, but how the cNTS processes ingestive feedback during behaviour is unknown. Here we describe how prolactin-releasing hormone (PRLH) and GCG neurons, two principal cNTS cell types that promote non-aversive satiety, are regulated during ingestion. PRLH neurons showed sustained activation by visceral feedback when nutrients were infused into the stomach, but these sustained responses were substantially reduced during oral consumption. Instead, PRLH neurons shifted to a phasic activity pattern that was time-locked to ingestion and linked to the taste of food. Optogenetic manipulations revealed that PRLH neurons control the duration of seconds-timescale feeding bursts, revealing a mechanism by which orosensory signals feed back to restrain the pace of ingestion. By contrast, GCG neurons were activated by mechanical feedback from the gut, tracked the amount of food consumed and promoted satiety that lasted for tens of minutes. These findings reveal that sequential negative feedback signals from the mouth and gut engage distinct circuits in the caudal brainstem, which in turn control elements of feeding behaviour operating on short and long timescales.


Subject(s)
Appetite Regulation , Brain Stem , Eating , Feedback, Physiological , Food , Satiation , Stomach , Appetite Regulation/physiology , Brain Stem/cytology , Brain Stem/physiology , Eating/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/metabolism , Prolactin-Releasing Hormone/metabolism , Satiation/physiology , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Stomach/physiology , Taste/physiology , Time Factors , Animals , Mice
5.
J Physiol ; 601(10): 1881-1896, 2023 05.
Article in English | MEDLINE | ID: mdl-36975145

ABSTRACT

Circadian regulation of autonomic reflex pathways pairs physiological function with the daily light cycle. The brainstem nucleus of the solitary tract (NTS) is a key candidate for rhythmic control of the autonomic nervous system. Here we investigated circadian regulation of NTS neurotransmission and synaptic throughput using patch-clamp electrophysiology in brainstem slices from mice. We found that spontaneous quantal glutamate release onto NTS neurons showed strong circadian rhythmicity, with the highest rate of release during the light phase and the lowest in the dark, that were sufficient to drive day/night differences in constitutive postsynaptic action potential firing. In contrast, afferent evoked action potential throughput was enhanced during the dark and diminished in the light. Afferent-driven synchronous release pathways showed a similar decrease in release probability that did not explain the enhanced synaptic throughput during the night. However, analysis of postsynaptic membrane properties revealed diurnal changes in conductance, which, when coupled with the circadian changes in glutamate release pathways, tuned synaptic throughput between the light and dark phases. These coordinated pre-/postsynaptic changes encode nuanced control over synaptic performance and pair NTS action potential firing and vagal throughput with time of day. KEY POINTS: Vagal afferent neurons relay information from peripheral organs to the brainstem nucleus of the solitary tract (NTS) to initiate autonomic reflex pathways as well as providing important controls of food intake, digestive function and energy balance. Vagally mediated reflexes and behaviours are under strong circadian regulation. Diurnal fluctuations in presynaptic vesicle release pathways and postsynaptic membrane conductances provide nuanced control over NTS action potential firing and vagal synaptic throughput. Coordinated pre-/postsynaptic changes represent a fundamental mechanism mediating daily changes in vagal afferent signalling and autonomic function.


Subject(s)
Circadian Rhythm , Glutamic Acid , Solitary Nucleus , Synapses , Circadian Rhythm/physiology , Glutamic Acid/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Synapses/metabolism , Neurons, Afferent/metabolism , Vagus Nerve/cytology , Vagus Nerve/physiology , Action Potentials , Male , Animals , Mice , Nodose Ganglion/metabolism , Signal Transduction , Electric Conductivity , Patch-Clamp Techniques
6.
Nature ; 609(7928): 761-771, 2022 09.
Article in English | MEDLINE | ID: mdl-36071158

ABSTRACT

Infections induce a set of pleiotropic responses in animals, including anorexia, adipsia, lethargy and changes in temperature, collectively termed sickness behaviours1. Although these responses have been shown to be adaptive, the underlying neural mechanisms have not been elucidated2-4. Here we use of a set of unbiased methodologies to show that a specific subpopulation of neurons in the brainstem can control the diverse responses to a bacterial endotoxin (lipopolysaccharide (LPS)) that potently induces sickness behaviour. Whole-brain activity mapping revealed that subsets of neurons in the nucleus of the solitary tract (NTS) and the area postrema (AP) acutely express FOS after LPS treatment, and we found that subsequent reactivation of these specific neurons in FOS2A-iCreERT2 (also known as TRAP2) mice replicates the behavioural and thermal component of sickness. In addition, inhibition of LPS-activated neurons diminished all of the behavioural responses to LPS. Single-nucleus RNA sequencing of the NTS-AP was used to identify LPS-activated neural populations, and we found that activation of ADCYAP1+ neurons in the NTS-AP fully recapitulates the responses elicited by LPS. Furthermore, inhibition of these neurons significantly diminished the anorexia, adipsia and locomotor cessation seen after LPS injection. Together these studies map the pleiotropic effects of LPS to a neural population that is both necessary and sufficient for canonical elements of the sickness response, thus establishing a critical link between the brain and the response to infection.


Subject(s)
Brain Stem , Illness Behavior , Neurons , Animals , Anorexia/complications , Area Postrema/cytology , Area Postrema/metabolism , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/physiology , Illness Behavior/drug effects , Lethargy/complications , Lipopolysaccharides/pharmacology , Mice , Neurons/drug effects , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/metabolism
7.
Nature ; 609(7926): 320-326, 2022 09.
Article in English | MEDLINE | ID: mdl-36045291

ABSTRACT

The nervous system uses various coding strategies to process sensory inputs. For example, the olfactory system uses large receptor repertoires and is wired to recognize diverse odours, whereas the visual system provides high acuity of object position, form and movement1-5. Compared to external sensory systems, principles that underlie sensory processing by the interoceptive nervous system remain poorly defined. Here we developed a two-photon calcium imaging preparation to understand internal organ representations in the nucleus of the solitary tract (NTS), a sensory gateway in the brainstem that receives vagal and other inputs from the body. Focusing on gut and upper airway stimuli, we observed that individual NTS neurons are tuned to detect signals from particular organs and are topographically organized on the basis of body position. Moreover, some mechanosensory and chemosensory inputs from the same organ converge centrally. Sensory inputs engage specific NTS domains with defined locations, each containing heterogeneous cell types. Spatial representations of different organs are further sharpened in the NTS beyond what is achieved by vagal axon sorting alone, as blockade of brainstem inhibition broadens neural tuning and disorganizes visceral representations. These findings reveal basic organizational features used by the brain to process interoceptive inputs.


Subject(s)
Brain Stem , Sensation , Brain Stem/anatomy & histology , Brain Stem/cytology , Brain Stem/physiology , Calcium/metabolism , Posture/physiology , Sensation/physiology , Sensory Receptor Cells/physiology , Solitary Nucleus/anatomy & histology , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Vagus Nerve/physiology
8.
Int J Mol Sci ; 22(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201760

ABSTRACT

Obstructive sleep apnea (OSA) is recurrent obstruction of the upper airway due to the loss of upper airway muscle tone during sleep. OSA is highly prevalent, especially in obesity. There is no pharmacotherapy for OSA. Previous studies have demonstrated the role of leptin, an adipose-tissue-produced hormone, as a potent respiratory stimulant. Leptin signaling via a long functional isoform of leptin receptor, LEPRb, in the nucleus of the solitary tract (NTS), has been implicated in control of breathing. We hypothesized that leptin acts on LEPRb positive neurons in the NTS to increase ventilation and maintain upper airway patency during sleep in obese mice. We expressed designer receptors exclusively activated by designer drugs (DREADD) selectively in the LEPRb positive neurons of the NTS of Leprb-Cre-GFP mice with diet-induced obesity (DIO) and examined the effect of DREADD ligand, J60, on tongue muscle activity and breathing during sleep. J60 was a potent activator of LEPRb positive NTS neurons, but did not stimulate breathing or upper airway muscles during NREM and REM sleep. We conclude that, in DIO mice, the stimulating effects of leptin on breathing during sleep are independent of LEPRb signaling in the NTS.


Subject(s)
Neurons/metabolism , Receptors, Drug/metabolism , Receptors, Leptin/metabolism , Sleep Apnea Syndromes/physiopathology , Solitary Nucleus/cytology , Animals , Electromyography , Leptin/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Obesity/etiology , Obesity/physiopathology , Sleep, REM , Solitary Nucleus/metabolism
9.
J Neurosci ; 40(37): 7054-7064, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32817248

ABSTRACT

Leptin signaling within the nucleus of the solitary tract (NTS) contributes to the control of food intake, and injections of leptin into the NTS reduce meal size and increase the efficacy of vagus-mediated satiation signals. Leptin receptors (LepRs) are expressed by vagal afferents as well as by a population of NTS neurons. However, the electrophysiological properties of LepR-expressing NTS neurons have not been well characterized, and it is unclear how leptin might act on these neurons to reduce food intake. To address this question, we recorded from LepR-expressing neurons in horizontal brain slices containing the NTS from male and female LepR-Cre X Rosa-tdTomato mice. We found that the vast majority of NTS LepR neurons received monosynaptic innervation from vagal afferent fibers and LepR neurons exhibited large synaptic NMDA receptor (NMDAR)-mediated currents compared with non-LepR neurons. During high-frequency stimulation of vagal afferents, leptin increased the size of NMDAR-mediated currents, but not AMPAR-mediated currents. Leptin also increased the size of evoked EPSPs and the ability of low-intensity solitary tract stimulation to evoke action potentials in LepR neurons. These effects of leptin were blocked by bath applying a competitive NMDAR antagonist (DCPP-ene) or by an NMDAR channel blocker applied through the recording pipette (MK-801). Last, feeding studies using male rats demonstrate that intra-NTS injections of DCPP-ene attenuate reduction of overnight food intake following intra-NTS leptin injection. Our results suggest that leptin acts in the NTS to reduce food intake by increasing NMDAR-mediated currents, thus enhancing NTS sensitivity to vagal inputs.SIGNIFICANCE STATEMENT Leptin is a hormone that critically impacts food intake and energy homeostasis. The nucleus of the solitary tract (NTS) is activated by vagal afferents from the gastrointestinal tract, which promotes termination of a meal. Injection of leptin into the NTS inhibits food intake, while knockdown of leptin receptors (LepRs) in NTS neurons increases food intake. However, little was known about how leptin acts in the NTS neurons to inhibit food intake. We found that leptin increases the sensitivity of LepR-expressing neurons to vagal inputs by increasing NMDA receptor-mediated synaptic currents and that NTS NMDAR activation contributes to leptin-induced reduction of food intake. These findings suggest a novel mechanism by which leptin, acting in the NTS, could potentiate gastrointestinal satiation signals.


Subject(s)
Excitatory Postsynaptic Potentials , Leptin/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Solitary Nucleus/metabolism , Vagus Nerve/metabolism , Animals , Dizocilpine Maleate/pharmacology , Eating , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Proline/analogs & derivatives , Proline/pharmacology , Pyridines/pharmacology , Rats , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Synapses/metabolism , Synapses/physiology , Vagus Nerve/cytology , Vagus Nerve/physiology
10.
PLoS Genet ; 16(8): e1008925, 2020 08.
Article in English | MEDLINE | ID: mdl-32790785

ABSTRACT

Taste receptor cells use multiple signaling pathways to detect chemicals in potential food items. These cells are functionally grouped into different types: Type I cells act as support cells and have glial-like properties; Type II cells detect bitter, sweet, and umami taste stimuli; and Type III cells detect sour and salty stimuli. We have identified a new population of taste cells that are broadly tuned to multiple taste stimuli including bitter, sweet, sour, and umami. The goal of this study was to characterize these broadly responsive (BR) taste cells. We used an IP3R3-KO mouse (does not release calcium (Ca2+) from internal stores in Type II cells when stimulated with bitter, sweet, or umami stimuli) to characterize the BR cells without any potentially confounding input from Type II cells. Using live cell Ca2+ imaging in isolated taste cells from the IP3R3-KO mouse, we found that BR cells are a subset of Type III cells that respond to sour stimuli but also use a PLCß signaling pathway to respond to bitter, sweet, and umami stimuli. Unlike Type II cells, individual BR cells are broadly tuned and respond to multiple stimuli across different taste modalities. Live cell imaging in a PLCß3-KO mouse confirmed that BR cells use this signaling pathway to respond to bitter, sweet, and umami stimuli. Short term behavioral assays revealed that BR cells make significant contributions to taste driven behaviors and found that loss of either PLCß3 in BR cells or IP3R3 in Type II cells caused similar behavioral deficits to bitter, sweet, and umami stimuli. Analysis of c-Fos activity in the nucleus of the solitary tract (NTS) also demonstrated that functional Type II and BR cells are required for normal stimulus induced expression.


Subject(s)
Taste Buds/cytology , Taste , Afferent Pathways/cytology , Animals , Calcium Signaling , Cells, Cultured , Female , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Phospholipase C beta/metabolism , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Taste Buds/metabolism , Taste Buds/physiology , Taste Perception
11.
Brain Res ; 1746: 147006, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32628919

ABSTRACT

Neuronatin (Nnat) is involved in the regulation of cellular molecular signaling and appears to be also linked to metabolic processes. The gastrointestinal peptides cholecystokinin (CCK) and bombesin (BN) have an effect on the short-term inhibition of food intake and induce neuronal activation in different brain nuclei, prominently in the nucleus of the solitary tract (NTS) involved in the modulation of food intake. The aim of the study was to examine if Nnat immunoreactivity is detectable in the NTS, and whether peripheral CCK-8S or BN cause c-Fos activation of Nnat neurons. Non-fasted male Sprague-Dawley rats received an intraperitoneal (i.p.) injection of 5.2 or 8.7 nmol CCK-8S/kg or 26 or 32 nmol BN/kg (n = 4 all groups) or vehicle solution (0.15 M NaCl; n = 7). The number of c-Fos neurons was determined 90 min post injection in the NTS and dorsal motor nucleus of the vagus (DMV). We observed Nnat immunoreactive neurons in the NTS and DMV. CCK-8S (25-fold and 51-fold, p = 0.025 and p = 0.001) and BN (31-fold and 59-fold, p = 0.007 and p = 0.001) at both doses increased the number of c-Fos positive neurons in the NTS. CCK and BN did not show a significant effect in the DMV. Both doses of CCK-8S (24-fold and 48-fold p = 0.011 and p = 0.001) and bombesin (31-fold and 56-fold, p = 0.002 and p = 0.001) increased the number of activated Nnat neurons in the NTS (p = 0.001) compared to the vehicle group, while in the DMV no significant increase of c-Fos activation was detected. In conclusion, i.p. injected CCK-8S or BN induce an increased neuronal activity in NTS Nnat neurons, giving rise that Nnat may play a role in the regulation of food intake mediated by peripheral CCK-8S or BN.


Subject(s)
Bombesin/pharmacology , Cholecystokinin/pharmacology , Neurons/drug effects , Peptide Fragments/pharmacology , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Animals , Male , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Solitary Nucleus/metabolism
12.
J Neuroendocrinol ; 32(6): e12855, 2020 06.
Article in English | MEDLINE | ID: mdl-32436241

ABSTRACT

Phoenixin (PNX) is a neuropeptide shown to play roles in the control of reproduction. The nucleus of the solitary tract (NTS), a critical autonomic integrating centre in the hindbrain, is one of many areas with dense expression of PNX. Using coronal NTS slices obtained from male Sprague-Dawley rats, the present study characterised the effects of PNX on both spike frequency and membrane potential of NTS neurones. Extracellular recordings demonstrated that bath-applied 10 nmol L-1 PNX increased the firing frequency in 32% of NTS neurones, effects which were confirmed with patch-clamp recordings showing that 50% of NTS neurones tested depolarised in response to application of the peptide. Surprisingly, the responsiveness to PNX in NTS neurones then declined suddenly to 9% (P < 0.001). This effect was subsequently attributed to stress associated with construction in our animal care facility because PNX responsiveness was again observed in slices from rats delivered and maintained in a construction-free facility. We then examined whether this loss of PNX responsiveness could be replicated in rats placed on a chronic stress regimen involving ongoing corticosterone (CORT) treatment in the construction-free facility. Slices from animals treated in this way showed a similar lack of neuronal responsiveness to PNX (9.1 ± 3.9%) within 2 weeks of CORT treatment. These effects were specific to PNX responsiveness because CORT treatment had no effect on the responsiveness of NTS neurones to angiotensin II. These results are the first to implicate PNX with respect to directly controlling the excitability of NTS neurones and also provide intriguing data showing the plasticity of these effects associated with environmental and glucocorticoid stress levels of the animal.


Subject(s)
Cellular Microenvironment , Glucocorticoids/adverse effects , Neurons/drug effects , Peptide Hormones/pharmacology , Solitary Nucleus/drug effects , Action Potentials/drug effects , Animals , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Electric Stimulation , Electrophysiological Phenomena/drug effects , Male , Membrane Potentials/drug effects , Neurons/physiology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Stress, Physiological/drug effects , Stress, Physiological/physiology
13.
Mol Metab ; 39: 101024, 2020 09.
Article in English | MEDLINE | ID: mdl-32446875

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are used as anti-diabetic drugs and are approved for obesity treatment. However, GLP-1RAs also affect heart rate (HR) and arterial blood pressure (ABP) in rodents and humans. Although the activation of GLP-1 receptors (GLP-1R) is known to increase HR, the circuits recruited are unclear, and in particular, it is unknown whether GLP-1RAs activate preproglucagon (PPG) neurons, the brain source of GLP-1, to elicit these effects. METHODS: We investigated the effect of GLP-1RAs on heart rate in anaesthetized adult mice. In a separate study, we manipulated the activity of nucleus tractus solitarius (NTS) PPG neurons (PPGNTS) in awake, freely behaving transgenic Glu-Cre mice implanted with biotelemetry probes and injected with AAV-DIO-hM3Dq:mCherry or AAV-mCherry-FLEX-DTA. RESULTS: Systemic administration of the GLP-1RA Ex-4 increased resting HR in anaesthetized or conscious mice, but had no effect on ABP in conscious mice. This effect was abolished by ß-adrenoceptor blockade with atenolol, but unaffected by the muscarinic antagonist atropine. Furthermore, Ex-4-induced tachycardia persisted when PPGNTS neurons were ablated, and Ex-4 did not induce expression of the neuronal activity marker cFos in PPGNTS neurons. PPGNTS ablation or acute chemogenetic inhibition of these neurons via hM4Di receptors had no effect on resting HR. In contrast, chemogenetic activation of PPGNTS neurons increased resting HR. Furthermore, the application of GLP-1 within the subarachnoid space of the middle thoracic spinal cord, a major projection target of PPG neurons, increased HR. CONCLUSIONS: These results demonstrate that both systemic application of Ex-4 or GLP-1 and chemogenetic activation of PPGNTS neurons increases HR. Ex-4 increases the activity of cardiac sympathetic preganglionic neurons of the spinal cord without recruitment of PPGNTS neurons, and thus likely recapitulates the physiological effects of PPG neuron activation. These neurons therefore do not play a significant role in controlling resting HR and ABP but are capable of inducing tachycardia and so are likely involved in cardiovascular responses to acute stress.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Heart Rate , Neurons/metabolism , Proglucagon/biosynthesis , Solitary Nucleus/physiology , Tachycardia/etiology , Tachycardia/metabolism , Animals , Disease Models, Animal , Electrocardiography , Exenatide/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Heart Rate/drug effects , Mice , Mice, Transgenic , Neurons/drug effects , Solitary Nucleus/cytology , Spinal Cord/drug effects , Spinal Cord/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Tachycardia/diagnosis
14.
Cell Metab ; 31(2): 301-312.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31955990

ABSTRACT

To understand hindbrain pathways involved in the control of food intake, we examined roles for calcitonin receptor (CALCR)-containing neurons in the NTS. Ablation of NTS Calcr abrogated the long-term suppression of food intake, but not aversive responses, by CALCR agonists. Similarly, activating CalcrNTS neurons decreased food intake and body weight but (unlike neighboring CckNTS cells) failed to promote aversion, revealing that CalcrNTS neurons mediate a non-aversive suppression of food intake. While both CalcrNTS and CckNTS neurons decreased feeding via projections to the PBN, CckNTS cells activated aversive CGRPPBN cells while CalcrNTS cells activated distinct non-CGRP PBN cells. Hence, CalcrNTS cells suppress feeding via non-aversive, non-CGRP PBN targets. Additionally, silencing CalcrNTS cells blunted food intake suppression by gut peptides and nutrients, increasing food intake and promoting obesity. Hence, CalcrNTS neurons define a hindbrain system that participates in physiological energy balance and suppresses food intake without activating aversive systems.


Subject(s)
Eating , Energy Metabolism , Neurons/metabolism , Receptors, Calcitonin/physiology , Solitary Nucleus/metabolism , Animals , Body Weight , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Solitary Nucleus/cytology
15.
Cell Metab ; 31(2): 313-326.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31839488

ABSTRACT

Glucose is the essential energy source for the brain, whose deficit, triggered by energy deprivation or therapeutic agents, can be fatal. Increased appetite is the key behavioral defense against hypoglycemia; however, the central pathways involved are not well understood. Here, we describe a glucoprivic feeding pathway by tyrosine hydroxylase (TH)-expressing neurons from nucleus of solitary tract (NTS), which project densely to the hypothalamus and elicit feeding through bidirectional adrenergic modulation of agouti-related peptide (AgRP)- and proopiomelanocortin (POMC)-expressing neurons. Acute chemogenetic inhibition of arcuate nucleus (ARC)-projecting NTSTH neurons or their target, AgRP neurons, impaired glucoprivic feeding induced by 2-Deoxy-D-glucose (2DG) injection. Neuroanatomical tracing results suggested that ARC-projecting orexigenic NTSTH neurons are largely distinct from neighboring catecholamine neurons projecting to parabrachial nucleus (PBN) that promotes satiety. Collectively, we describe a circuit organization in which an ascending pathway from brainstem stimulates appetite through key hunger neurons in the hypothalamus in response to hypoglycemia.


Subject(s)
Agouti-Related Protein/metabolism , Appetite Regulation , Hypoglycemia/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Solitary Nucleus/metabolism , Animals , Female , Hypothalamus/cytology , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Solitary Nucleus/cytology
16.
Glia ; 68(6): 1241-1254, 2020 06.
Article in English | MEDLINE | ID: mdl-31880353

ABSTRACT

A role for glial cells in brain circuits controlling feeding has begun to be identified with hypothalamic astrocyte signaling implicated in regulating energy homeostasis. The nucleus of the solitary tract (NTS), within the brainstem dorsal vagal complex (DVC), integrates vagal afferent information from the viscera and plays a role in regulating food intake. We hypothesized that astrocytes in this nucleus respond to, and influence, food intake. Mice fed high-fat chow for 12 hr during the dark phase showed NTS astrocyte activation, reflected in an increase in the number (65%) and morphological complexity of glial-fibrillary acidic protein (GFAP)-immunoreactive cells adjacent to the area postrema (AP), compared to control chow fed mice. To measure the impact of astrocyte activation on food intake, we delivered designer receptors exclusively activated by designer drugs (DREADDs) to DVC astrocytes (encompassing NTS, AP, and dorsal motor nucleus of the vagus) using an adeno-associated viral (AAV) vector (AAV-GFAP-hM3Dq_mCherry). Chemogenetic activation with clozapine-N-oxide (0.3 mg/kg) produced in greater morphological complexity in astrocytes and reduced dark-phase feeding by 84% at 4 hr postinjection compared with vehicle treatment. hM3Dq-activation of DVC astrocytes also reduced refeeding after an overnight fast (71% lower, 4 hr postinjection) when compared to AAV-GFAP-mCherry expressing control mice. DREADD-mediated astrocyte activation did not impact locomotion. hM3Dq activation of DVC astrocytes induced c-FOS in neighboring neuronal feeding circuits (including in the parabrachial nucleus). This indicates that NTS astrocytes respond to acute nutritional excess, are involved in the integration of peripheral satiety signals, and can reduce food intake when activated.


Subject(s)
Astrocytes/metabolism , Brain Stem/metabolism , Eating/physiology , Hypothalamus/metabolism , Neurons/metabolism , Animals , Glial Fibrillary Acidic Protein/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/metabolism , Solitary Nucleus/cytology
17.
World J Gastroenterol ; 25(40): 6077-6093, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31686764

ABSTRACT

BACKGROUND: Central sensitization plays a pivotal role in the maintenance of chronic pain induced by chronic pancreatitis (CP). We hypothesized that the nucleus tractus solitarius (NTS), a primary central site that integrates pancreatic afferents apart from the thoracic spinal dorsal horn, plays a key role in the pathogenesis of visceral hypersensitivity in a rat model of CP. AIM: To investigate the role of the NTS in the visceral hypersensitivity induced by chronic pancreatitis. METHODS: CP was induced by the intraductal injection of trinitrobenzene sulfonic acid (TNBS) in rats. Pancreatic hyperalgesia was assessed by referred somatic pain via von Frey filament assay. Neural activation of the NTS was indicated by immunohistochemical staining for Fos. Basic synaptic transmission within the NTS was assessed by electrophysiological recordings. Expression of vesicular glutamate transporters (VGluTs), N-methyl-D-aspartate receptor subtype 2B (NR2B), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subtype 1 (GluR1) was analyzed by immunoblotting. Membrane insertion of NR2B and GluR1 was evaluated by electron microscopy. The regulatory role of the NTS in visceral hypersensitivity was detected via pharmacological approach and chemogenetics in CP rats. RESULTS: TNBS treatment significantly increased the number of Fos-expressing neurons within the caudal NTS. The excitatory synaptic transmission was substantially potentiated within the caudal NTS in CP rats (frequency: 5.87 ± 1.12 Hz in CP rats vs 2.55 ± 0.44 Hz in sham rats, P < 0.01; amplitude: 19.60 ± 1.39 pA in CP rats vs 14.71 ± 1.07 pA in sham rats; P < 0.01). CP rats showed upregulated expression of VGluT2, and increased phosphorylation and postsynaptic trafficking of NR2B and GluR1 within the caudal NTS. Blocking excitatory synaptic transmission via the AMPAR antagonist CNQX and the NMDAR antagonist AP-5 microinjection reversed visceral hypersensitivity in CP rats (abdominal withdraw threshold: 7.00 ± 1.02 g in CNQX group, 8.00 ± 0.81 g in AP-5 group and 1.10 ± 0.27 g in saline group, P < 0.001). Inhibiting the excitability of NTS neurons via chemogenetics also significantly attenuated pancreatic hyperalgesia (abdominal withdraw threshold: 13.67 ± 2.55 g in Gi group, 2.00 ± 1.37 g in Gq group, and 2.36 ± 0.67 g in mCherry group, P < 0.01). CONCLUSION: Our findings suggest that enhanced excitatory transmission within the caudal NTS contributes to pancreatic pain and emphasize the NTS as a pivotal hub for the processing of pancreatic afferents, which provide novel insights into the central sensitization of painful CP.


Subject(s)
Chronic Pain/physiopathology , Enteric Nervous System/physiopathology , Hyperalgesia/physiopathology , Pancreatitis, Chronic/complications , Solitary Nucleus/physiopathology , Afferent Pathways/physiopathology , Animals , Chronic Pain/etiology , Disease Models, Animal , Humans , Hyperalgesia/etiology , Male , Neurons/physiology , Pancreas/innervation , Pancreatitis, Chronic/chemically induced , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Solitary Nucleus/cytology , Stereotaxic Techniques , Synaptic Transmission/physiology , Trinitrobenzenesulfonic Acid/toxicity
18.
J Neurosci ; 39(49): 9767-9781, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31666353

ABSTRACT

Stress responses are coordinated by widespread neural circuits. Homeostatic and psychogenic stressors activate preproglucagon (PPG) neurons in the caudal nucleus of the solitary tract (cNTS) that produce glucagon-like peptide-1; published work in rodents indicates that these neurons play a crucial role in stress responses. While the axonal targets of PPG neurons are well established, their afferent inputs are unknown. Here we use retrograde tracing with cholera toxin subunit b to show that the cNTS in male and female mice receives axonal inputs similar to those reported in rats. Monosynaptic and polysynaptic inputs specific to cNTS PPG neurons were revealed using Cre-conditional pseudorabies and rabies viruses. The most prominent sources of PPG monosynaptic input include the lateral (LH) and paraventricular (PVN) nuclei of the hypothalamus, parasubthalamic nucleus, lateral division of the central amygdala, and Barrington's nucleus (Bar). Additionally, PPG neurons receive monosynaptic vagal sensory input from the nodose ganglia and spinal sensory input from the dorsal horn. Sources of polysynaptic input to cNTS PPG neurons include the hippocampal formation, paraventricular thalamus, and prefrontal cortex. Finally, cNTS-projecting neurons within PVN, LH, and Bar express the activation marker cFOS in mice after restraint stress, identifying them as potential sources of neurogenic stress-induced recruitment of PPG neurons. In summary, cNTS PPG neurons in mice receive widespread monosynaptic and polysynaptic input from brain regions implicated in coordinating behavioral and physiological stress responses, as well as from vagal and spinal sensory neurons. Thus, PPG neurons are optimally positioned to integrate signals of homeostatic and psychogenic stress.SIGNIFICANCE STATEMENT Recent research has indicated a crucial role for glucagon-like peptide-1-producing preproglucagon (PPG) neurons in regulating both appetite and behavioral and autonomic responses to acute stress. Intriguingly, the central glucagon-like peptide-1 system defined in rodents is conserved in humans, highlighting the translational importance of understanding its anatomical organization. Findings reported here indicate that PPG neurons receive significant monosynaptic and polysynaptic input from brain regions implicated in autonomic and behavioral responses to stress, as well as direct input from vagal and spinal sensory neurons. Improved understanding of the neural pathways underlying the recruitment of PPG neurons may facilitate the development of novel therapies for the treatment of stress-related disorders.


Subject(s)
Neurons/physiology , Proglucagon/physiology , Synapses/physiology , Vagus Nerve/physiology , Animals , Axons/physiology , Female , Hypothalamus/physiology , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Neurons, Afferent/physiology , Posterior Horn Cells/physiology , Reflex, Monosynaptic/physiology , Restraint, Physical , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Stress, Psychological/physiopathology , Thalamus/physiology
19.
J Neurosci ; 39(41): 8038-8050, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31471471

ABSTRACT

Integration and modulation of primary afferent sensory information begins at the first terminating sites within the CNS, where central inhibitory circuits play an integral role. Viscerosensory information is conveyed to the nucleus of the solitary tract (NTS) where it initiates neuroendocrine, behavioral, and autonomic reflex responses that ensure optimal internal organ function. This excitatory input is modulated by diverse, local inhibitory interneurons, whose functions are not clearly understood. Here we show that, in male rats, 65% of somatostatin-expressing (SST) NTS neurons also express GAD67, supporting their likely role as inhibitory interneurons. Using whole-cell recordings of NTS neurons, from horizontal brainstem slices of male and female SST-yellow fluorescent protein (YFP) and SST-channelrhodopsin 2 (ChR2)-YFP mice, we quantified the impact of SST-NTS neurons on viscerosensory processing. Light-evoked excitatory photocurrents were reliably obtained from SST-ChR2-YFP neurons (n = 16) and the stimulation-response characteristics determined. Most SST neurons (57%) received direct input from solitary tract (ST) afferents, indicating that they form part of a feedforward circuit. All recorded SST-negative NTS neurons (n = 72) received SST-ChR2 input. ChR2-evoked PSCs were largely inhibitory and, in contrast to previous reports, were mediated by both GABA and glycine. When timed to coincide, the ChR2-activated SST input suppressed ST-evoked action potentials at second-order NTS neurons, demonstrating strong modulation of primary viscerosensory input. These data indicate that the SST inhibitory network innervates broadly within the NTS, with the potential to gate viscerosensory input to powerfully alter autonomic reflex function and other behaviors.SIGNIFICANCE STATEMENT Sensory afferent input is modulated according to state. For example the baroreflex is altered during a stress response or exercise, but the basic mechanisms underpinning this sensory modulation are not fully understood in any sensory system. Here we demonstrate that the neuronal processing of viscerosensory information begins with synaptic gating at the first central synapse with second-order neurons in the NTS. These data reveal that the somatostatin subclass of inhibitory interneurons are driven by visceral sensory input to play a major role in gating viscerosensory signals, placing them within a feedforward circuit within the NTS.


Subject(s)
Nerve Net/physiology , Neurons/physiology , Sensation/physiology , Sensory Gating/physiology , Somatostatin/physiology , Animals , Feedback, Physiological , Female , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/physiology , Glycine/physiology , Interneurons/physiology , Male , Mice , Nerve Net/cytology , Photic Stimulation , Rats , Rats, Sprague-Dawley , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Visceral Afferents/physiology , gamma-Aminobutyric Acid/physiology
20.
Int J Obes (Lond) ; 43(12): 2370-2380, 2019 12.
Article in English | MEDLINE | ID: mdl-31152154

ABSTRACT

BACKGROUND: Elevated circulating levels of the divergent transforming growth factor-beta (TGFb) family cytokine, growth differentiation factor 15 (GDF15), acting through its CNS receptor, glial-derived neurotrophic factor receptor alpha-like (GFRAL), can cause anorexia and weight loss leading to anorexia/cachexia syndrome of cancer and other diseases. Preclinical studies suggest that administration of drugs based on recombinant GDF15 might be used to treat severe obesity. However, the role of the GDF15-GFRAL pathway in the physiological regulation of body weight and metabolism is unclear. The critical site of action of GFRAL in the CNS has also not been proven beyond doubt. To investigate these two aspects, we have inhibited the actions of GDF15 in mice started on high-fat diet (HFD). METHODS: The actions of GDF15 were inhibited using two methods: (1) Groups of 8 mice under HFD had their endogenous GDF15 neutralised by monoclonal antibody treatment, (2) Groups of 15 mice received AAV-shRNA to knockdown GFRAL at its hypothesised major sites of action, the hindbrain area postrema (AP) and the nucleus of the solitary tract (NTS). Metabolic measurements were determined during both experiments. CONCLUSIONS: Treating mice with monoclonal antibody to GDF15 shortly after commencing HFD results in more rapid gain of body weight, adiposity and hepatic lipid deposition than the control groups. This is accompanied by reduced glucose and insulin tolerance and greater expression of pro-inflammatory cytokines in adipose tissue. Localised AP and NTS shRNA-GFRAL knockdown in mice commencing HFD similarly caused an increase in body weight and adiposity. This effect was in proportion to the effectiveness of GFRAL knockdown, indicated by quantitative analysis of hindbrain GFRAL staining. We conclude that the GDF15-GFRAL axis plays an important role in resistance to obesity in HFD-fed mice and that the major site of action of GDF15 in the CNS is GFRAL-expressing neurons in the AP and NTS.


Subject(s)
Adiposity , Glial Cell Line-Derived Neurotrophic Factor Receptors , Growth Differentiation Factor 15 , Rhombencephalon , Adiposity/genetics , Adiposity/physiology , Animals , Area Postrema/cytology , Area Postrema/metabolism , Area Postrema/physiology , Body Weight/physiology , Diet, High-Fat , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Obesity/metabolism , Rhombencephalon/cytology , Rhombencephalon/metabolism , Rhombencephalon/physiology , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Solitary Nucleus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL