1
|
Hedley KE, Gomez HM, Kecelioglu E, Carroll OR, Jobling P, Horvat JC, Tadros MA. Neonatal Chlamydia muridarum respiratory infection causes neuroinflammation within the brainstem during the early postnatal period. J Neuroinflammation 2024; 21:158. [PMID: 38879567 PMCID: PMC11179230 DOI: 10.1186/s12974-024-03150-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
Respiratory infections are one of the most common causes of illness and morbidity in neonates worldwide. In the acute phase infections are known to cause wide-spread peripheral inflammation. However, the inflammatory consequences to the critical neural control centres for respiration have not been explored. Utilising a well characterised model of neonatal respiratory infection, we investigated acute responses within the medulla oblongata which contains key respiratory regions. Neonatal mice were intranasally inoculated within 24 h of birth, with either Chlamydia muridarum or sham-infected, and tissue collected on postnatal day 15, the peak of peripheral inflammation. A key finding of this study is that, while the periphery appeared to show no sex-specific effects of a neonatal respiratory infection, sex had a significant impact on the inflammatory response of the medulla oblongata. There was a distinct sex-specific response in the medulla coincident with peak of peripheral inflammation, with females demonstrating an upregulation of anti-inflammatory cytokines and males showing very few changes. Microglia also demonstrated sex-specificity with the morphology of females and males differing based upon the nuclei. Astrocytes showed limited changes during the acute response to neonatal infection. These data highlight the strong sex-specific impact of a respiratory infection can have on the medulla in the acute inflammatory phase.
Collapse
Affiliation(s)
- Kateleen E Hedley
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Henry M Gomez
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Eda Kecelioglu
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Olivia R Carroll
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Phillip Jobling
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jay C Horvat
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia.
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
2
|
Mishra D, Richard JE, Maric I, Shevchouk OT, Börchers S, Eerola K, Krieger JP, Skibicka KP. Lateral parabrachial nucleus astrocytes control food intake. Front Endocrinol (Lausanne) 2024; 15:1389589. [PMID: 38887265 PMCID: PMC11180714 DOI: 10.3389/fendo.2024.1389589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/03/2024] [Indexed: 06/20/2024] Open
Abstract
Food intake behavior is under the tight control of the central nervous system. Most studies to date focus on the contribution of neurons to this behavior. However, although previously overlooked, astrocytes have recently been implicated to play a key role in feeding control. Most of the recent literature has focused on astrocytic contribution in the hypothalamus or the dorsal vagal complex. The contribution of astrocytes located in the lateral parabrachial nucleus (lPBN) to feeding behavior control remains poorly understood. Thus, here, we first investigated whether activation of lPBN astrocytes affects feeding behavior in male and female rats using chemogenetic activation. Astrocytic activation in the lPBN led to profound anorexia in both sexes, under both ad-libitum feeding schedule and after a fasting challenge. Astrocytes have a key contribution to glutamate homeostasis and can themselves release glutamate. Moreover, lPBN glutamate signaling is a key contributor to potent anorexia, which can be induced by lPBN activation. Thus, here, we determined whether glutamate signaling is necessary for lPBN astrocyte activation-induced anorexia, and found that pharmacological N-methyl D-aspartate (NMDA) receptor blockade attenuated the food intake reduction resulting from lPBN astrocyte activation. Since astrocytes have been shown to contribute to feeding control by modulating the feeding effect of peripheral feeding signals, we further investigated whether lPBN astrocyte activation is capable of modulating the anorexic effect of the gut/brain hormone, glucagon like peptide -1, as well as the orexigenic effect of the stomach hormone - ghrelin, and found that the feeding effect of both signals is modulated by lPBN astrocytic activation. Lastly, we found that lPBN astrocyte activation-induced anorexia is affected by a diet-induced obesity challenge, in a sex-divergent manner. Collectively, current findings uncover a novel role for lPBN astrocytes in feeding behavior control.
Collapse
Affiliation(s)
- Devesh Mishra
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jennifer E. Richard
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Ivana Maric
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Olesya T. Shevchouk
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Stina Börchers
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Kim Eerola
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jean-Philippe Krieger
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich - VetSuisse, Zurich, Switzerland
| | - Karolina P. Skibicka
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
3
|
Zhu Y, Sun H, Wang H, Li N. Synaptic mechanisms underlying the elevated sympathetic outflow in fructose-induced hypertension. Front Physiol 2024; 15:1365594. [PMID: 38505704 PMCID: PMC10949223 DOI: 10.3389/fphys.2024.1365594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/05/2024] [Indexed: 03/21/2024] Open
Abstract
Metabolic syndrome is associated with cardiovascular dysfunction, including elevated sympathetic outflow. However, the underlying brain mechanisms are unclear. The nucleus tractus solitarius (NTS) critically regulates autonomic reflexes related to cardiovascular function and contains neurons projecting to the caudal ventrolateral medulla (CVLM). Nitric oxide (NO) is a diffusible free-radical messenger in the vascular, immune, and nervous systems. In this study, we determine if NO in the NTS is involved in the synaptic plasticity underlying the elevated sympathetic outflow in fructose-induced hypertension. We retrogradely labeled CVLM-projecting NTS neurons through the injection of FluoSpheres into the CVLM in a fructose-fed rat model to determine the cellular mechanism involved in increased sympathetic outflow. Fructose feeding increased the blood pressure and glucose levels, which represent metabolic syndrome. We found that fructose feeding reduces the NO precursor L-arginine-induced increase in the firing activity of CVLM-projecting NTS neurons. Furthermore, fructose feeding reduces the L-arginine-induced increase in presynaptic spontaneous glutamatergic synaptic inputs to NTS neurons, while NO donor DEA/NO produces an increase in glutamatergic synaptic inputs in fructose-fed rats similar to that in vehicle-treated rats. In addition, fructose feeding reduces the NO-induced depressor response and sympathoinhibition. These data suggested that fructose feeding reduced NO production and, thus, the subsequent NO-induced glutamate releases in the NTS and depressor response. The findings of this study provide new insights into the central mechanisms involved in the neural control of cardiovascular and autonomic functions in the NTS in metabolic syndrome.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiying Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjie Wang
- Department of Anesthesiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Na Li
- Department of Physiology, School of Basic Medical Sciences, Hebei University, Baoding, China
| |
Collapse
|
4
|
Hofmann GC, Gama de Barcellos Filho P, Khodadadi F, Ostrowski D, Kline DD, Hasser EM. Vagotomy blunts cardiorespiratory responses to vagal afferent stimulation via pre- and postsynaptic effects in the nucleus tractus solitarii. J Physiol 2024; 602:1147-1174. [PMID: 38377124 DOI: 10.1113/jp285854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Abstract
Viscerosensory information travels to the brain via vagal afferents, where it is first integrated within the brainstem nucleus tractus solitarii (nTS), a critical contributor to cardiorespiratory function and site of neuroplasticity. We have shown that decreasing input to the nTS via unilateral vagus nerve transection (vagotomy) induces morphological changes in nTS glia and reduces sighs during hypoxia. The mechanisms behind post-vagotomy changes are not well understood. We hypothesized that chronic vagotomy alters cardiorespiratory responses to vagal afferent stimulation via blunted nTS neuronal activity. Male Sprague-Dawley rats (6 weeks old) underwent right cervical vagotomy caudal to the nodose ganglion, or sham surgery. After 1 week, rats were anaesthetized, ventilated and instrumented to measure mean arterial pressure (MAP), heart rate (HR), and splanchnic sympathetic and phrenic nerve activity (SSNA and PhrNA, respectively). Vagal afferent stimulation (2-50 Hz) decreased cardiorespiratory parameters and increased neuronal Ca2+ measured by in vivo photometry and in vitro slice imaging of nTS GCaMP8m. Vagotomy attenuated both these reflex and neuronal Ca2+ responses compared to shams. Vagotomy also reduced presynaptic Ca2+ responses to stimulation (Cal-520 imaging) in the nTS slice. The decrease in HR, SSNA and PhrNA due to nTS nanoinjection of exogenous glutamate also was tempered following vagotomy. This effect was not restored by blocking excitatory amino acid transporters. However, the blunted responses were mimicked by NMDA, not AMPA, nanoinjection and were associated with reduced NR1 subunits in the nTS. Altogether, these results demonstrate that vagotomy induces multiple changes within the nTS tripartite synapse that influence cardiorespiratory reflex responses to afferent stimulation. KEY POINTS: Multiple mechanisms within the nucleus tractus solitarii (nTS) contribute to functional changes following vagal nerve transection. Vagotomy results in reduced cardiorespiratory reflex responses to vagal afferent stimulation and nTS glutamate nanoinjection. Blunted responses occur via reduced presynaptic Ca2+ activation and attenuated NMDA receptor expression and function, leading to a reduction in nTS neuronal activation. These results provide insight into the control of autonomic and respiratory function, as well as the plasticity that can occur in response to nerve damage and cardiorespiratory disease.
Collapse
Affiliation(s)
- Gabrielle C Hofmann
- Comparative Medicine, University of Missouri, Columbia, Missouri, USA
- Area Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Procopio Gama de Barcellos Filho
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Fateme Khodadadi
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Daniela Ostrowski
- Department of Pharmacology, A.T. Still University, Kirksville, Missouri, USA
| | - David D Kline
- Area Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Eileen M Hasser
- Area Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
5
|
Tao R, Liu S, Crawford J, Tao F. Gut-Brain Crosstalk and the Central Mechanisms of Orofacial Pain. Brain Sci 2023; 13:1456. [PMID: 37891825 PMCID: PMC10605055 DOI: 10.3390/brainsci13101456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/21/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Accumulated evidence has demonstrated that the gut microbiome can contribute to pain modulation through the microbiome-gut-brain axis. Various relevant microbiome metabolites in the gut are involved in the regulation of pain signaling in the central nervous system. In this review, we summarize recent advances in gut-brain interactions by which the microbiome metabolites modulate pain, with a focus on orofacial pain, and we further discuss the role of gut-brain crosstalk in the central mechanisms of orofacial pain whereby the gut microbiome modulates orofacial pain via the vagus nerve-mediated direct pathway and the gut metabolites/molecules-mediated indirect pathway. The direct and indirect pathways both contribute to the central regulation of orofacial pain through different brain structures (such as the nucleus tractus solitarius and the parabrachial nucleus) and signaling transmission across the blood-brain barrier, respectively. Understanding the gut microbiome-regulated pain mechanisms in the brain could help us to develop non-opioid novel therapies for orofacial pain.
Collapse
Affiliation(s)
| | | | | | - Feng Tao
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, 3302 Gaston Ave., Dallas, TX 75246, USA
| |
Collapse
|
6
|
Fozzato A, New LE, Griffiths JC, Patel B, Deuchars SA, Filippi BM. Manipulating mitochondrial dynamics in the NTS prevents diet-induced deficits in brown fat morphology and glucose uptake. Life Sci 2023; 328:121922. [PMID: 37423379 DOI: 10.1016/j.lfs.2023.121922] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
AIMS Brown adipose tissue (BAT) can produce heat by metabolizing glucose and fatty acids. Activation of BAT is controlled by the central nervous system (CNS) through sympathetic innervation. Dysregulation of signalling molecules in selective CNS areas such as the nucleus of tractus solitarius (NTS) are linked with altered BAT activity, obesity and diabetes. High-fat diet (HFD)-feeding increases mitochondrial fragmentation in the NTS, triggering insulin resistance, hyperphagia and weight gain. Here we sought to determine whether changes in mitochondrial dynamics in the NTS can affect BAT glucose uptake. MAIN METHODS Rats received DVC stereotactic surgery for local brain administration of viruses that express mutated Drp1 genes. BAT glucose uptake was measured with PET/CT scans. Biochemical assays and immunohistochemistry determined altered levels of key signalling molecules and neural innervation of BAT. KEY FINDINGS We show that short-term HFD-feeding decreases BAT glucose uptake. However, inhibiting mitochondrial fragmentation in NTS-astrocytes of HFD-fed rats partially restores BAT glucose uptake accompanied by lower blood glucose and insulin levels. Tyrosine Hydroxylase (TH) revealed that rats with inhibited mitochondrial fragmentation in NTS astrocytes had higher levels of catecholaminergic innervation in BAT compared to HFD-fed rats, and did not exhibit HFD-dependent infiltration of enlarged white fat droplets in the BAT. In regular chow-fed rats, increasing mitochondrial fragmentation in the NTS-astrocytes reduced BAT glucose uptake, TH immune-positive boutons and β3-adrenergic receptor levels. SIGNIFICANCE Our data suggest that targeting mitochondrial dynamics in the NTS-astrocytes could be a beneficial strategy to increase glucose utilization and protect from developing obesity and diabetes.
Collapse
Affiliation(s)
- Arianna Fozzato
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Lauryn E New
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Joanne C Griffiths
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Bianca Patel
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Susan A Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Beatrice M Filippi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
| |
Collapse
|
7
|
MacDonald AJ, Pye KR, Beall C, Ellacott KLJ. Impact of chemogenetic activation of dorsal vagal complex astrocytes in mice on adaptive glucoregulatory responses. J Neuroendocrinol 2023; 35:e13315. [PMID: 37370201 DOI: 10.1111/jne.13315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/13/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
The dorsal vagal complex (DVC) regulates diverse aspects of physiology including food intake and blood glucose homeostasis. Astrocytes play an active role in regulating DVC function and, by extension, physiological parameters. DVC astrocytes in ex vivo slices respond to low tissue glucose. The response of neurons to low glucose is conditional on intact astrocyte signalling in slice preparations, suggesting astrocytes are primary sensors of glucose deprivation (glucoprivation). Based on these published findings we hypothesised that in vivo DVC astrocyte manipulation with chemogenetics would be sufficient to alter physiological responses that control blood glucose. We found that 2-h after systemic 2-DG-induced glucoprivation there were no observable changes in morphology of glial fibrillary acidic protein (GFAP)-immunoreactive DVC cells, specifically those in the nucleus of the solitary tract (NTS). Chemogenetic activation of DVC astrocytes was sufficient to suppress nocturnal food intake by reducing both meal size and meal number and this manipulation also suppressed 2-DG-induced glucoprivic food intake. Chemogenetic activation of DVC astrocytes did not increase basal blood glucose nor protect against insulin-induced hypoglycaemia. In male mice, chemogenetic DVC astrocyte activation did not alter glucose tolerance. In female mice, the initial glucose excursion was reduced in a glucose tolerance test, suggesting enhanced glucose absorption. Based on our data and published work, we propose that DVC astrocytes may play an indispensable homeostatic role, that is, are necessary to maintain the function of glucoregulatory neuronal circuitry, but alone their bulk activation is not sufficient to result in adaptive glucoregulatory responses. It is possible that there are state-dependent effects and/or DVC astrocyte subsets that have this specialised role, but this was unresolvable using the experimental approaches employed here.
Collapse
Affiliation(s)
- Alastair J MacDonald
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Katherine R Pye
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Craig Beall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Kate L J Ellacott
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
8
|
Fabbri R, Spennato D, Conte G, Konstantoulaki A, Lazzarini C, Saracino E, Nicchia GP, Frigeri A, Zamboni R, Spray DC, Benfenati V. The emerging science of Glioception: Contribution of glia in sensing, transduction, circuit integration of interoception. Pharmacol Ther 2023; 245:108403. [PMID: 37024060 DOI: 10.1016/j.pharmthera.2023.108403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
Interoception is the process by which the nervous system regulates internal functions to achieve homeostasis. The role of neurons in interoception has received considerable recent attention, but glial cells also contribute. Glial cells can sense and transduce signals including osmotic, chemical, and mechanical status of extracellular milieu. Their ability to dynamically communicate "listening" and "talking" to neurons is necessary to monitor and regulate homeostasis and information integration in the nervous system. This review introduces the concept of "Glioception" and focuses on the process by which glial cells sense, interpret and integrate information about the inner state of the organism. Glial cells are ideally positioned to act as sensors and integrators of diverse interoceptive signals and can trigger regulatory responses via modulation of the activity of neuronal networks, both in physiological and pathological conditions. We believe that understanding and manipulating glioceptive processes and underlying molecular mechanisms provide a key path to develop new therapies for the prevention and alleviation of devastating interoceptive dysfunctions, among which pain is emphasized here with more focused details.
Collapse
Affiliation(s)
- Roberta Fabbri
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, viale del Risorgimento 2, 40136 Bologna, Italy.
| | - Diletta Spennato
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Giorgia Conte
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Aikaterini Konstantoulaki
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi, 2, 40126 Bologna, BO, Italy
| | - Chiara Lazzarini
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Emanuela Saracino
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Grazia Paola Nicchia
- School of Medicine, Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, BA, Italy; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Antonio Frigeri
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Roberto Zamboni
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - David C Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Valentina Benfenati
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy.
| |
Collapse
|
9
|
Ellacott KLJ. Gastrocytes and GLUttony - astrocyte regulation of calorie intake via glutamatergic modulation of gastric activity in rats. J Physiol 2023; 601:709-710. [PMID: 36704958 DOI: 10.1113/jp284264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Affiliation(s)
- Kate L J Ellacott
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
10
|
Jia S, Rybalchenko N, Kunwar K, Farmer GE, Little JT, Toney GM, Cunningham JT. Chronic intermittent hypoxia enhances glycinergic inhibition in nucleus tractus solitarius. J Neurophysiol 2022; 128:1383-1394. [PMID: 36321700 PMCID: PMC9678432 DOI: 10.1152/jn.00241.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Chronic intermittent hypoxia (CIH), an animal model of sleep apnea, has been shown to alter the activity of second-order chemoreceptor neurons in the caudal nucleus of the solitary tract (cNTS). Although numerous studies have focused on excitatory plasticity, few studies have explored CIH-induced plasticity impacting inhibitory inputs to NTS neurons, and the roles of GABAergic and glycinergic inputs on heightened cNTS excitability following CIH are unknown. In addition, changes in astrocyte function may play a role in cNTS plasticity responses to CIH. This study tested the effects of a 7-day CIH protocol on miniature inhibitory postsynaptic currents (mIPSCs) in cNTS neurons receiving chemoreceptor afferents. Normoxia-treated rats primarily displayed GABA mIPSCs, whereas CIH-treated rats exhibited a shift toward combined GABA/glycine-mediated mIPSCs. CIH increased glycinergic mIPSC amplitude and area. This shift was not observed in dorsal motor nucleus of the vagus neurons or cNTS cells from females. Immunohistochemistry showed that strengthened glycinergic mIPSCs were associated with increased glycine receptor protein and were dependent on receptor trafficking in CIH-treated rats. In addition, CIH altered astrocyte morphology in the cNTS, and inactivation of astrocytes following CIH reduced glycine receptor-mediated mIPSC frequency and overall mIPSC amplitude. In cNTS, CIH produced changes in glycine signaling that appear to reflect increased trafficking of glycine receptors to the cell membrane. Increased glycine signaling in cNTS associated with CIH also appears to be dependent on astrocytes. Additional studies will be needed to determine how CIH influences glycine receptor expression and astrocyte function in cNTS.NEW & NOTEWORTHY Chronic intermittent hypoxia (CIH) has been used to mimic the hypoxemia associated with sleep apnea and determine how these hypoxemias influence neural function. The nucleus of the solitary tract is the main site for chemoreceptor input to the CNS, but how CIH influences NTS inhibition has not been determined. These studies show that CIH increases glycine-mediated miniature IPSCs through mechanisms that depend on protein trafficking and astrocyte activation.
Collapse
Affiliation(s)
- Shuping Jia
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Nataliya Rybalchenko
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Kishor Kunwar
- 2Microscopy Core, Division of Research and Innovation, University of Texas Health Science Center, Fort Worth, Texas
| | - George E. Farmer
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Joel T. Little
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| | - Glenn M. Toney
- 3Department of Cellular & Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - J. Thomas Cunningham
- 1Department of Physiology and Anatomy, University of Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
11
|
Cai P, Huang SN, Lin ZH, Wang Z, Liu RF, Xiao WH, Li ZS, Zhu ZH, Yao J, Yan XB, Wang FD, Zeng SX, Chen GQ, Yang LY, Sun YK, Yu C, Chen L, Wang WX. Regulation of wakefulness by astrocytes in the lateral hypothalamus. Neuropharmacology 2022; 221:109275. [PMID: 36195131 DOI: 10.1016/j.neuropharm.2022.109275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 09/03/2022] [Accepted: 09/26/2022] [Indexed: 10/07/2022]
Abstract
The lateral hypothalamus (LH) is an important brain region mediating sleep-wake behavior. Recent evidence has shown that central nervous system astrocytes modulate the activity of adjacent neurons and participate in several physiological functions. However, the role of LH astrocytes in sleep-wake regulation remains unclear. Here, using synchronous recording of electroencephalogram/electromyogram in mice and calcium signals in LH astrocytes, we show that the activity of LH astrocytes is significantly increased during non-rapid eye movement (NREM) sleep-to-wake transitions and decreased during wake-to-NREM sleep transitions. Chemogenetic activation of LH astrocytes potently promotes wakefulness and maintains long-term arousal, while chemogenetic inhibition of LH astrocytes decreases the total amount of wakefulness in mice. Moreover, by combining chemogenetics with fiber photometry, we show that activation of LH astrocytes significantly increases the calcium signals of adjacent neurons, especially among GABAergic neurons. Taken together, our results clearly illustrate that LH astrocytes are a key neural substrate regulating wakefulness and encode this behavior through surrounding GABAergic neurons. Our findings raise the possibility that overactivity of LH astrocytes may be an underlying mechanism of clinical sleep disorders.
Collapse
Affiliation(s)
- Ping Cai
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Sheng-Nan Huang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhi-Hui Lin
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zewu Wang
- Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Ren-Fu Liu
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Wen-Hao Xiao
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhang-Shu Li
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhong-Hua Zhu
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Jing Yao
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiong-Bin Yan
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Fu-Dan Wang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Shun-Xing Zeng
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Guo-Qiang Chen
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Liu-Yun Yang
- School of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Yu-Kun Sun
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China.
| | - Li Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, Fujian, China.
| | - Wen-Xiang Wang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
12
|
Jaster JH, Ottaviani G. Acute Heart Failure, 90-Day Mortality, and Gravitational Ischemia in the Brain. Diagnostics (Basel) 2022; 12:diagnostics12061473. [PMID: 35741284 PMCID: PMC9221596 DOI: 10.3390/diagnostics12061473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/04/2022] [Accepted: 06/14/2022] [Indexed: 12/20/2022] Open
Abstract
During the 90 days following hospitalization for acute heart failure, the ejection fraction and type of discharge medications have been shown in clinical trials to have little effect on mortality. We examined the recent literature addressing brain-related etiologies of sudden death following heart failure. Two mechanisms of sudden unexpected death have been suggested to possibly result from four significant influences on pathophysiology in the brain. The two causes of sudden death are (1) severe cardiac arrhythmia and (2) neurogenic pulmonary edema. They are both mediated through the brainstem autonomic nuclei generally and executed specifically through the dorsal motor nucleus of the vagus nerve. The four significant influences on pathophysiology, all contributing to ischemia in the brainstem autonomic nuclei, are: (1) Hyper-stimulation of neurons in the solitary tract nucleus, increasing their metabolic requirements; (2) Inadequate blood flow at a vascular watershed terminus, perfusing the autonomic nuclei; (3) Additionally decreased blood flow, globally throughout the brain, following vasoconstriction related to relative hyperventilation and decreased pCO2 levels; (4) Gravitational ischemia in the brainstem caused by the weight of the cerebral hemispheres sitting above the brainstem. Changes in head tilt release gravitational ischemia in the brain. There is no specific head position relative to gravity that is considered favorable or unfavorable for an extended period of time, longer than 24 h. Even a small degree of head elevation, used in managing pulmonary congestion, may increase gravitational ischemia in the posterior fossa and brainstem. In this paper, we suggest a new and important research avenue for intervening in cardiac arrhythmias and preventing their occurrence through the non-invasive use of head-tilting and other head repositioning maneuvers. This could potentially help many geriatric patients with heart failure, who have decreased mobility in bed, and who tend to stay in one position longer, thereby increasing gravitational ischemia in the brain.
Collapse
Affiliation(s)
| | - Giulia Ottaviani
- Anatomic Pathology, Lino Rossi Research Center, Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-50320819; Fax: +39-02-50320823
| |
Collapse
|
13
|
Holt MK. The ins and outs of the caudal nucleus of the solitary tract: An overview of cellular populations and anatomical connections. J Neuroendocrinol 2022; 34:e13132. [PMID: 35509189 PMCID: PMC9286632 DOI: 10.1111/jne.13132] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
The body and brain are in constant two-way communication. Driving this communication is a region in the lower brainstem: the dorsal vagal complex. Within the dorsal vagal complex, the caudal nucleus of the solitary tract (cNTS) is a major first stop for incoming information from the body to the brain carried by the vagus nerve. The anatomy of this region makes it ideally positioned to respond to signals of change in both emotional and bodily states. In turn, the cNTS controls the activity of regions throughout the brain that are involved in the control of both behaviour and physiology. This review is intended to help anyone with an interest in the cNTS. First, I provide an overview of the architecture of the cNTS and outline the wide range of neurotransmitters expressed in subsets of neurons in the cNTS. Next, in detail, I discuss the known inputs and outputs of the cNTS and briefly highlight what is known regarding the neurochemical makeup and function of those connections. Then, I discuss one group of cNTS neurons: glucagon-like peptide-1 (GLP-1)-expressing neurons. GLP-1 neurons serve as a good example of a group of cNTS neurons, which receive input from varied sources and have the ability to modulate both behaviour and physiology. Finally, I consider what we might learn about other cNTS neurons from our study of GLP-1 neurons and why it is important to remember that the manipulation of molecularly defined subsets of cNTS neurons is likely to affect physiology and behaviours beyond those monitored in individual experiments.
Collapse
Affiliation(s)
- Marie K. Holt
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
14
|
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci 2022; 16:888232. [PMID: 35614970 PMCID: PMC9124899 DOI: 10.3389/fncel.2022.888232] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Although myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) has a specific and distinctive profile of clinical features, the disease remains an enigma because causal explanation of the pathobiological matrix is lacking. Several potential disease mechanisms have been identified, including immune abnormalities, inflammatory activation, mitochondrial alterations, endothelial and muscular disturbances, cardiovascular anomalies, and dysfunction of the peripheral and central nervous systems. Yet, it remains unclear whether and how these pathways may be related and orchestrated. Here we explore the hypothesis that a common denominator of the pathobiological processes in ME/CFS may be central nervous system dysfunction due to impaired or pathologically reactive neuroglia (astrocytes, microglia and oligodendrocytes). We will test this hypothesis by reviewing, in reference to the current literature, the two most salient and widely accepted features of ME/CFS, and by investigating how these might be linked to dysfunctional neuroglia. From this review we conclude that the multifaceted pathobiology of ME/CFS may be attributable in a unifying manner to neuroglial dysfunction. Because the two key features - post exertional malaise and decreased cerebral blood flow - are also recognized in a subset of patients with post-acute sequelae COVID, we suggest that our findings may also be pertinent to this entity.
Collapse
Affiliation(s)
- Herbert Renz-Polster
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Quebec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Joachim E. Fischer
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
15
|
Stojanovska V, Atta J, Kelly SB, Zahra VA, Matthews-Staindl E, Nitsos I, Moxham A, Pham Y, Hooper SB, Herlenius E, Galinsky R, Polglase GR. Increased Prostaglandin E2 in Brainstem Respiratory Centers Is Associated With Inhibition of Breathing Movements in Fetal Sheep Exposed to Progressive Systemic Inflammation. Front Physiol 2022; 13:841229. [PMID: 35309054 PMCID: PMC8928579 DOI: 10.3389/fphys.2022.841229] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/08/2022] [Indexed: 12/11/2022] Open
Abstract
Background Preterm newborns commonly experience apnoeas after birth and require respiratory stimulants and support. Antenatal inflammation is a common antecedent of preterm birth and inflammatory mediators, particularly prostaglandin E2 (PGE2), are associated with inhibition of vital brainstem respiratory centers. In this study, we tested the hypothesis that exposure to antenatal inflammation inhibits fetal breathing movements (FBMs) and increases inflammation and PGE2 levels in brainstem respiratory centers, cerebrospinal fluid (CSF) and blood plasma. Methods Chronically instrumented late preterm fetal sheep at 0.85 of gestation were randomly assigned to receive repeated intravenous saline (n = 8) or lipopolysaccharide (LPS) infusions (experimental day 1 = 300 ng, day 2 = 600 ng, day 3 = 1200 ng, n = 8). Fetal breathing movements were recorded throughout the experimental period. Sheep were euthanized 4 days after starting infusions for assessment of brainstem respiratory center histology. Results LPS infusions increased circulating and cerebrospinal fluid PGE2 levels, decreased arterial oxygen saturation, increased the partial pressure of carbon dioxide and lactate concentration, and decreased pH (p < 0.05 for all) compared to controls. LPS infusions caused transient reductions in the % of time fetuses spent breathing and the proportion of vigorous fetal breathing movements (P < 0.05 vs. control). LPS-exposure increased PGE2 expression in the RTN/pFRG (P < 0.05 vs. control) but not the pBÖTC (P < 0.07 vs. control) of the brainstem. No significant changes in gene expression were observed for PGE2 enzymes or caspase 3. LPS-exposure reduced the numbers of GFAP-immunoreactive astrocytes in the RTN/pFRG, NTS and XII of the brainstem (P < 0.05 vs. control for all) and increased microglial activation in the RTN/pFRG, preBÖTC, NTS, and XII brainstem respiratory centers (P < 0.05 vs. control for all). Conclusion Chronic LPS-exposure in late preterm fetal sheep increased PGE2 levels within the brainstem, CSF and plasma, and was associated with inhibition of FBMs, astrocyte loss and microglial activation within the brainstem respiratory centers. Further studies are needed to determine whether the inflammation-induced increase in PGE2 levels plays a key role in depressing respiratory drive in the perinatal period.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - John Atta
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Sharmony B. Kelly
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Valerie A. Zahra
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Eva Matthews-Staindl
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Ilias Nitsos
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Alison Moxham
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Yen Pham
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Stuart B. Hooper
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Eric Herlenius
- Department of Women’s and Children’s Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
- Astrid Lindgren Childrens Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Robert Galinsky
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- *Correspondence: Robert Galinsky,
| | - Graeme R. Polglase
- The Ritchie Center, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Graeme R. Polglase,
| |
Collapse
|
16
|
Glial Modulation of Energy Balance: The Dorsal Vagal Complex Is No Exception. Int J Mol Sci 2022; 23:ijms23020960. [PMID: 35055143 PMCID: PMC8779587 DOI: 10.3390/ijms23020960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/04/2023] Open
Abstract
The avoidance of being overweight or obese is a daily challenge for a growing number of people. The growing proportion of people suffering from a nutritional imbalance in many parts of the world exemplifies this challenge and emphasizes the need for a better understanding of the mechanisms that regulate nutritional balance. Until recently, research on the central regulation of food intake primarily focused on neuronal signaling, with little attention paid to the role of glial cells. Over the last few decades, our understanding of glial cells has changed dramatically. These cells are increasingly regarded as important neuronal partners, contributing not just to cerebral homeostasis, but also to cerebral signaling. Our understanding of the central regulation of energy balance is part of this (r)evolution. Evidence is accumulating that glial cells play a dynamic role in the modulation of energy balance. In the present review, we summarize recent data indicating that the multifaceted glial compartment of the brainstem dorsal vagal complex (DVC) should be considered in research aimed at identifying feeding-related processes operating at this level.
Collapse
|
17
|
Hofmann GC, Hasser EM, Kline DD. Unilateral vagotomy alters astrocyte and microglial morphology in the nucleus tractus solitarii of the rat. Am J Physiol Regul Integr Comp Physiol 2021; 320:R945-R959. [PMID: 33978480 PMCID: PMC8285617 DOI: 10.1152/ajpregu.00019.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022]
Abstract
The nucleus tractus solitarii (nTS) is the initial site of integration of sensory information from the cardiorespiratory system and contributes to reflex responses to hypoxia. Afferent fibers of the bilateral vagus nerves carry input from the heart, lungs, and other organs to the nTS where it is processed and modulated. Vagal afferents and nTS neurons are integrally associated with astrocytes and microglia that contribute to neuronal activity and influence cardiorespiratory control. We hypothesized that vagotomy would alter glial morphology and cardiorespiratory responses to hypoxia. Unilateral vagotomy (or sham surgery) was performed in rats. Prior to and seven days after surgery, baseline and hypoxic cardiorespiratory responses were monitored in conscious and anesthetized animals. The brainstem was sectioned and caudal, mid-area postrema (mid-AP), and rostral sections of the nTS were prepared for immunohistochemistry. Vagotomy increased immunoreactivity (-IR) of astrocytic glial fibrillary acidic protein (GFAP), specifically at mid-AP in the nTS. Similar results were found in the dorsal motor nucleus of the vagus (DMX). Vagotomy did not alter nTS astrocyte number, yet increased astrocyte branching and altered morphology. In addition, vagotomy both increased nTS microglia number and produced morphologic changes indicative of activation. Cardiorespiratory baseline parameters and hypoxic responses remained largely unchanged, but vagotomized animals displayed fewer augmented breaths (sighs) in response to hypoxia. Altogether, vagotomy alters nTS glial morphology, indicative of functional changes in astrocytes and microglia that may affect cardiorespiratory function in health and disease.
Collapse
Affiliation(s)
- Gabrielle C Hofmann
- Comparative Medicine, University of Missouri, Columbia, Missouri
- Area Pathobiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Eileen M Hasser
- Area Pathobiology, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - David D Kline
- Area Pathobiology, University of Missouri, Columbia, Missouri
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
18
|
Clyburn C, Browning KN. Glutamatergic plasticity within neurocircuits of the dorsal vagal complex and the regulation of gastric functions. Am J Physiol Gastrointest Liver Physiol 2021; 320:G880-G887. [PMID: 33730858 PMCID: PMC8202199 DOI: 10.1152/ajpgi.00014.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The meticulous regulation of the gastrointestinal (GI) tract is required for the coordination of gastric motility and emptying, intestinal secretion, absorption, and transit as well as for the overarching management of food intake and energy homeostasis. Disruption of GI functions is associated with the development of severe GI disorders and the alteration of food intake and caloric balance. Functional GI disorders as well as the dysregulation of energy balance and food intake are frequently associated with, or result from, alterations in the central regulation of GI control. The faithful and rapid transmission of information from the stomach and upper GI tract to second-order neurons of the nucleus of the tractus solitarius (NTS) relies on the delicate modulation of excitatory glutamatergic transmission, as does the relay of integrated signals from the NTS to parasympathetic efferent neurons of the dorsal motor nucleus of the vagus (DMV). Many studies have focused on understanding the physiological and pathophysiological modulation of these glutamatergic synapses, although their role in the control and regulation of GI functions has lagged behind that of cardiovascular and respiratory functions. The purpose of this review is to examine the current literature exploring the role of glutamatergic transmission in the DVC in the regulation of GI functions.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|