1
|
Cheng YY, Yao Q, Miao Y, Guan W. Metformin as a potential antidepressant: Mechanisms and therapeutic insights in depression. Biochem Pharmacol 2025; 233:116773. [PMID: 39894309 DOI: 10.1016/j.bcp.2025.116773] [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: 11/21/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
Depression is one of the most disabling psychiatric disorders, whose pathophysiology has not been fully understood. Increasing numbers of preclinical studies have highlighted that metformin, as the first-line hypoglycaemic agent, has a potential pleiotropic effect on depression. Moreover, there is emerging evidence that metformin shows antidepressant activity and improves depressive symptoms in rodent models of depression. However, the exact role and underlying mechanism of metformin in depression remain unclear and still need to be investigated. Recent studies suggest that metformin not only improves neuronal damage and structural plasticity in the hippocampus but also enhances the antidepressant effect of antidepressants. Therefore, in this review, we summarize the existing evidence for the use of metformin as a psychopharmaceutical and elaborate on the underlying mechanisms of metformin in mitigating the onset and progression of depression, as well as the associated biochemical signaling pathways and targets involved in the pathogenesis of depression. After reviewing several studies, we conclude that metformin helps reduce depressive symptoms by targeting multiple pathways, including the regulation of neurotransmitters, enhanced neurogenesis, anti-inflammatory effects, and changes in gut microbiota. We aim to gain a deeper understanding of the mechanism of action of metformin and provide new insights into its clinical value in the prevention and therapy of depression.
Collapse
Affiliation(s)
- Yuan-Yuan Cheng
- Department of Pharmacology, Nantong Stomatological Hospital, Nantong 226001 Jiangsu, China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong 226001 Jiangsu, China
| | - Yang Miao
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng 224000 Jiangsu, China.
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001 Jiangsu, China.
| |
Collapse
|
2
|
Singh J, Wilkins G, Goodman-Vincent E, Chishti S, Bonilla Guerrero R, Fiori F, Ameenpur S, McFadden L, Zahavi Z, Santosh P. Using Precision Medicine to Disentangle Genotype-Phenotype Relationships in Twins with Rett Syndrome: A Case Report. Curr Issues Mol Biol 2024; 46:8424-8440. [PMID: 39194714 DOI: 10.3390/cimb46080497] [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: 06/26/2024] [Revised: 07/20/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Rett syndrome (RTT) is a paediatric neurodevelopmental disorder spanning four developmental stages. This multi-system disorder offers a unique window to explore genotype-phenotype relationships in a disease model. However, genetic prognosticators of RTT have limited clinical value due to the disorder's heterogeneity on multiple levels. This case report used a precision medicine approach to better understand the clinical phenotype of RTT twins with an identical pathogenic MECP2 mutation and discordant neurodevelopmental profiles. Targeted genotyping, objective physiological monitoring of heart rate variability (HRV) parameters, and clinical severity were assessed in a RTT twin pair (5 years 7 months old) with an identical pathogenic MECP2 mutation. Longitudinal assessment of autonomic HRV parameters was conducted using the Empatica E4 wristband device, and clinical severity was assessed using the RTT-anchored Clinical Global Impression Scale (RTT-CGI) and the Multi-System Profile of Symptoms Scale (MPSS). Genotype data revealed impaired BDNF function for twin A when compared to twin B. Twin A also had poorer autonomic health than twin B, as indicated by lower autonomic metrics (autonomic inflexibility). Hospitalisation, RTT-CGI-S, and MPSS subscale scores were used as measures of clinical severity, and these were worse in twin A. Treatment using buspirone shifted twin A from an inflexible to a flexible autonomic profile. This was mirrored in the MPSS scores, which showed a reduction in autonomic and cardiac symptoms following buspirone treatment. Our findings showed that a combination of a co-occurring rs6265 BDNF polymorphism, and worse autonomic and clinical profiles led to a poorer prognosis for twin A compared to twin B. Buspirone was able to shift a rigid autonomic profile to a more flexible one for twin A and thereby prevent cardiac and autonomic symptoms from worsening. The clinical profile for twin A represents a departure from the disorder trajectory typically observed in RTT and underscores the importance of wider genotype profiling and longitudinal objective physiological monitoring alongside measures of clinical symptoms and severity when assessing genotype-phenotype relationships in RTT patients with identical pathogenic mutations. A precision medicine approach that assesses genetic and physiological risk factors can be extended to other neurodevelopmental disorders to monitor risk when genotype-phenotype relationships are not so obvious.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Georgina Wilkins
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Ella Goodman-Vincent
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Samiya Chishti
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | | | - Federico Fiori
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Shashidhar Ameenpur
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Leighton McFadden
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Zvi Zahavi
- Myogenes Limited, Borehamwood WD6 4PJ, UK
| | - Paramala Santosh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| |
Collapse
|
3
|
Fan YZ, Duan YL, Chen CT, Wang Y, Zhu AP. Advances in attenuating opioid-induced respiratory depression: A narrative review. Medicine (Baltimore) 2024; 103:e38837. [PMID: 39029082 PMCID: PMC11398798 DOI: 10.1097/md.0000000000038837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Opioids exert analgesic effects by agonizing opioid receptors and activating signaling pathways coupled to receptors such as G-protein and/or β-arrestin. Concomitant respiratory depression (RD) is a common clinical problem, and improvement of RD is usually achieved with specific antagonists such as naloxone; however, naloxone antagonizes opioid analgesia and may produce more unknown adverse effects. In recent years, researchers have used various methods to isolate opioid receptor-mediated analgesia and RD, with the aim of preserving opioid analgesia while attenuating RD. At present, the focus is mainly on the development of new opioids with weak respiratory inhibition or the use of non-opioid drugs to stimulate breathing. This review reports recent advances in novel opioid agents, such as mixed opioid receptor agonists, peripheral selective opioid receptor agonists, opioid receptor splice variant agonists, biased opioid receptor agonists, and allosteric modulators of opioid receptors, as well as in non-opioid agents, such as AMPA receptor modulators, 5-hydroxytryptamine receptor agonists, phosphodiesterase-4 inhibitors, and nicotinic acetylcholine receptor agonists.
Collapse
Affiliation(s)
- Yong-Zheng Fan
- The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, China
| | - Yun-Li Duan
- Xiangyang No. 4 Middle School Compulsory Education Department, Xiangyang, China
| | - Chuan-Tao Chen
- Taihe Country People's Hospital·The Taihe Hospital of Wannan Medical College, Fuyang, China
| | - Yu Wang
- The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, China
| | - An-Ping Zhu
- The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, China
| |
Collapse
|
4
|
Gonye EC, Dagli AV, Kumar NN, Clements RT, Xu W, Bayliss DA. Expression of endogenous epitope-tagged GPR4 in the mouse brain. eNeuro 2024; 11:ENEURO.0002-24.2024. [PMID: 38408869 DOI: 10.1523/eneuro.0002-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 02/28/2024] Open
Abstract
GPR4 is a proton-sensing G protein-coupled receptor implicated in many peripheral and central physiological processes. GPR4 expression has previously been assessed only via detection of the cognate transcript or indirectly, by use of fluorescent reporters. In this work, CRISPR/Cas9 knock-in technology was used to encode a hemagglutinin (HA) epitope tag within the endogenous locus of Gpr4 and visualize GPR4-HA in the mouse central nervous system using a specific, well characterized HA antibody; GPR4 expression was further verified by complementary Gpr4 mRNA detection. HA immunoreactivity was found in a limited set of brain regions, including in the retrotrapezoid nucleus (RTN), serotonergic raphe nuclei, medial habenula, lateral septum, and several thalamic nuclei. GPR4 expression was not restricted to cells of a specific neurochemical identity as it was observed in excitatory, inhibitory, and aminergic neuronal cell groups. HA immunoreactivity was not detected in brain vascular endothelium, despite clear expression of Gpr4 mRNA in endothelial cells. In the RTN, GPR4 expression was detected at the soma and in proximal dendrites along blood vessels and the ventral surface of the brainstem; HA immunoreactivity was not detected in RTN projections to two known target regions. This localization of GPR4 protein in mouse brain neurons corroborates putative sites of expression where its function has been previously implicated (e.g., CO2-regulated breathing by RTN), and provides a guide for where GPR4 could contribute to other CO2/H+ modulated brain functions. Finally, GPR4-HA animals provide a useful reagent for further study of GPR4 in other physiological processes outside of the brain.Significance Statement GPR4 is a proton-sensing G-protein coupled receptor whose expression is necessary for a number of diverse physiological processes including acid-base sensing in the kidney, immune function, and cancer progression. In the brain, GPR4 has been implicated in the hypercapnic ventilatory response mediated by brainstem neurons. While knockout studies in animals have clearly demonstrated its necessity for normal physiology, descriptions of GPR4 expression have been limited due to a lack of specific antibodies for use in mouse models. In this paper, we implemented a CRISPR/Cas9 knock-in approach to incorporate the coding sequence for a small epitope tag into the locus of GPR4. Using these mice, we were able to describe GPR4 protein expression directly for the first time.
Collapse
Affiliation(s)
- Elizabeth C Gonye
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| | - Alexandra V Dagli
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| | - Natasha N Kumar
- University of New South Wales Sydney, School of Biomedical Sciences, New South Wales, Australia
| | - Rachel T Clements
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| | - Wenhao Xu
- University of Virginia, Genetically Engineered Mouse Model Core, Charlottesville, VA, USA
| | - Douglas A Bayliss
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| |
Collapse
|
5
|
Cummings KJ, Leiter JC, Trachtenberg FL, Okaty BW, Darnall RA, Haas EA, Harper RM, Nattie EE, Krous HF, Mena OJ, Richerson GB, Dymecki SM, Kinney HC, Haynes RL. Altered 5-HT2A/C receptor binding in the medulla oblongata in the sudden infant death syndrome (SIDS): Part II. Age-associated alterations in serotonin receptor binding profiles within medullary nuclei supporting cardiorespiratory homeostasis. J Neuropathol Exp Neurol 2024; 83:144-160. [PMID: 38323418 PMCID: PMC10880067 DOI: 10.1093/jnen/nlae004] [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] [Indexed: 02/08/2024] Open
Abstract
The failure of chemoreflexes, arousal, and/or autoresuscitation to asphyxia may underlie some sudden infant death syndrome (SIDS) cases. In Part I, we showed that some SIDS infants had altered 5-hydroxytryptamine (5-HT)2A/C receptor binding in medullary nuclei supporting chemoreflexes, arousal, and autoresuscitation. Here, using the same dataset, we tested the hypotheses that the prevalence of low 5-HT1A and/or 5-HT2A/C receptor binding (defined as levels below the 95% confidence interval of controls-a new approach), and the percentages of nuclei affected are greater in SIDS versus controls, and that the distribution of low binding varied with age of death. The prevalence and percentage of nuclei with low 5-HT1A and 5-HT2A/C binding in SIDS were twice that of controls. The percentage of nuclei with low 5-HT2A/C binding was greater in older SIDS infants. In >80% of older SIDS infants, low 5-HT2A/C binding characterized the hypoglossal nucleus, vagal dorsal nucleus, nucleus of solitary tract, and nuclei of the olivocerebellar subnetwork (important for blood pressure regulation). Together, our findings from SIDS infants and from animal models of serotonergic dysfunction suggest that some SIDS cases represent a serotonopathy. We present new hypotheses, yet to be tested, about how defects within serotonergic subnetworks may lead to SIDS.
Collapse
Affiliation(s)
- Kevin J Cummings
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - James C Leiter
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | | | - Benjamin W Okaty
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert A Darnall
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Elisabeth A Haas
- Department of Research, Rady’s Children’s Hospital, San Diego, California, USA
| | - Ronald M Harper
- Department of Neurobiology and the Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Eugene E Nattie
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Henry F Krous
- Department of Pediatrics, University of California San Diego, San Diego, California, USA
- Departments of Pathology and Pediatrics, Rady Children’s Hospital, San Diego, California, USA
| | - Othon J Mena
- San Diego County Medical Examiner Office, San Diego, California, USA
| | - George B Richerson
- Departments of Neurology and Molecular Physiology & Biophysics, University of Iowa, Iowa City, Iowa, USA
| | - Susan M Dymecki
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Hannah C Kinney
- Department of Pathology, CJ Murphy Laboratory for SIDS Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Robin L Haynes
- Department of Pathology, CJ Murphy Laboratory for SIDS Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Aung O, Amorim MR, Mendelowitz D, Polotsky VY. Revisiting the Role of Serotonin in Sleep-Disordered Breathing. Int J Mol Sci 2024; 25:1483. [PMID: 38338762 PMCID: PMC10855456 DOI: 10.3390/ijms25031483] [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: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Serotonin or 5-hydroxytryptamine (5-HT) is a ubiquitous neuro-modulator-transmitter that acts in the central nervous system, playing a major role in the control of breathing and other physiological functions. The midbrain, pons, and medulla regions contain several serotonergic nuclei with distinct physiological roles, including regulating the hypercapnic ventilatory response, upper airway patency, and sleep-wake states. Obesity is a major risk factor in the development of sleep-disordered breathing (SDB), such as obstructive sleep apnea (OSA), recurrent closure of the upper airway during sleep, and obesity hypoventilation syndrome (OHS), a condition characterized by daytime hypercapnia and hypoventilation during sleep. Approximately 936 million adults have OSA, and 32 million have OHS worldwide. 5-HT acts on 5-HT receptor subtypes that modulate neural control of breathing and upper airway patency. This article reviews the role of 5-HT in SDB and the current advances in 5-HT-targeted treatments for SDB.
Collapse
Affiliation(s)
- O Aung
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA; (O.A.); (M.R.A.)
- Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC 20037, USA
| | - Mateus R. Amorim
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA; (O.A.); (M.R.A.)
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC 20037, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA;
| | - Vsevolod Y. Polotsky
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA; (O.A.); (M.R.A.)
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC 20037, USA
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA;
| |
Collapse
|
7
|
MacFarlane PM, Martin RJ, Di Fiore JM, Raffay TM, Tatsuoka C, Chen Z, Minich N, Quintas G, Sánchez-Illana Á, Kuligowski J, Piñeiro-Ramos JD, Vento M, Hibbs AM. Plasma serotonergic biomarkers are associated with hypoxemia events in preterm neonates. Pediatr Res 2023; 94:1436-1443. [PMID: 37188799 PMCID: PMC11414210 DOI: 10.1038/s41390-023-02620-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/15/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Hypoxemia is a physiological manifestation of immature respiratory control in preterm neonates, which is likely impacted by neurotransmitter imbalances. We investigated relationships between plasma levels of the neurotransmitter serotonin (5-HT), metabolites of tryptophan (TRP), and parameters of hypoxemia in preterm neonates. METHODS TRP, 5-HT, 5-hydroxyindoleacetic acid (5-HIAA), and kynurenic acid (KA) were analyzed in platelet-poor plasma at ~1 week and ~1 month of life from a prospective cohort of 168 preterm neonates <31 weeks gestational age (GA). Frequency of intermittent hypoxemia (IH) events and percent time hypoxemic (<80%) were analyzed in a 6 h window after the blood draw. RESULTS At 1 week, infants with detectable plasma 5-HT had fewer IH events (OR (95% CI) = 0.52 (0.29, 0.31)) and less percent time <80% (OR (95% CI) = 0.54 (0.31, 0.95)) compared to infants with undetectable 5-HT. A similar relationship occurred at 1 month. At 1 week, infants with higher KA showed greater percent time <80% (OR (95% CI) = 1.90 (1.03, 3.50)). TRP, 5-HIAA or KA were not associated with IH frequency at either postnatal age. IH frequency and percent time <80% were positively associated with GA < 29 weeks. CONCLUSIONS Circulating neuromodulators 5-HT and KA might represent biomarkers of immature respiratory control contributing to hypoxemia in preterm neonates. IMPACT Hypoxemia events are frequent in preterm infants and are associated with poor outcomes. Mechanisms driving hypoxemia such as immature respiratory control may include central and peripheral imbalances in modulatory neurotransmitters. This study found associations between the plasma neuromodulators serotonin and kynurenic acid and parameters of hypoxemia in preterm neonates. Imbalances in plasma biomarkers affecting respiratory control may help identify neonates at risk of short- and long-term adverse outcomes.
Collapse
Affiliation(s)
- Peter Mathew MacFarlane
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA.
| | - Richard John Martin
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Juliann Marie Di Fiore
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Thomas Michael Raffay
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Curtis Tatsuoka
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Zhengyi Chen
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Nori Minich
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Guillermo Quintas
- Health and Biomedicine, Leitat Technological Center, Carrer de la Innovació, 2, 08225, Terrassa, Spain
- Analytical Unit, Health Research Institute La Fe, Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Ángel Sánchez-Illana
- Neonatal Research Unit, Health Research Institute La Fe (IISLAFE), University & Polytechnic Hospita La Fe (HULAFE), Valencia, Spain
- Department of Analytical Chemistry, Chemistry Faculty, Universtitat de València, Burjassot, Spain
| | - Julia Kuligowski
- Neonatal Research Unit, Health Research Institute La Fe (IISLAFE), University & Polytechnic Hospita La Fe (HULAFE), Valencia, Spain
| | - José David Piñeiro-Ramos
- Neonatal Research Unit, Health Research Institute La Fe (IISLAFE), University & Polytechnic Hospita La Fe (HULAFE), Valencia, Spain
| | - Maximo Vento
- Neonatal Research Unit, Health Research Institute La Fe (IISLAFE), University & Polytechnic Hospita La Fe (HULAFE), Valencia, Spain
| | - Anna Maria Hibbs
- Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
8
|
Marciante AB, Lurk C, Mata L, Lewis J, Reznikov LR, Mitchell GS. Progressive tauopathy disrupts breathing stability and chemoreflexes during presumptive sleep in mice. Front Physiol 2023; 14:1272980. [PMID: 37811498 PMCID: PMC10551153 DOI: 10.3389/fphys.2023.1272980] [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: 08/04/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Rationale: Although sleep apnea occurs in over 50% of individuals with Alzheimer's Disease (AD) or related tauopathies, little is known concerning the potential role of tauopathy in the pathogenesis of sleep apnea. Here, we tested the hypotheses that, during presumptive sleep, a murine model of tauopathy (rTg4510) exhibits: 1) increased breathing instability; 2) impaired chemoreflex function; and 3) exacerbation of these effects with tauopathy progression. Methods: rTg4510 mice initially develop robust tauopathy in the hippocampus and cortex, and eventually progresses to the brainstem. Type I and II post-sigh apnea, Type III (spontaneous) apnea, sigh, and hypopnea incidence were measured in young adult (5-6 months; n = 10-14/group) and aged (13-15 months; n = 22-24/group) non-transgenic (nTg), monogenic control tetracycline transactivator, and bigenic rTg4510 mice using whole-body plethysmography during presumptive sleep (i.e., eyes closed, curled/laying posture, stable breathing for >200 breaths) while breathing room air (21% O2). Peripheral and central chemoreceptor sensitivity were assessed with transient exposures (5 min) to hyperoxia (100% O2) or hypercapnia (3% and 5% CO2 in 21% O2), respectively. Results: We report significant increases in Type I, II, and III apneas (all p < 0.001), sighs (p = 0.002) and hypopneas (p < 0.001) in aged rTg4510 mice, but only Type III apneas in young adult rTg4510 mice (p < 0.001) versus age-matched nTg controls. Aged rTg4510 mice exhibited profound chemoreflex impairment versus age matched nTg and tTA mice. In rTg4510 mice, breathing frequency, tidal volume and minute ventilation were not affected by hyperoxic or hypercapnic challenges, in striking contrast to controls. Histological examination revealed hyperphosphorylated tau in brainstem regions involved in the control of breathing (e.g., pons, medullary respiratory column, retrotrapezoid nucleus) in aged rTg4510 mice. Neither breathing instability nor hyperphosphorylated tau in brainstem tissues were observed in young adult rTg4510 mice. Conclusion: Older rTg4510 mice exhibit profound impairment in the neural control of breathing, with greater breathing instability and near absence of oxygen and carbon-dioxide chemoreflexes. Breathing impairments paralleled tauopathy progression into brainstem regions that control breathing. These findings are consistent with the idea that tauopathy per se undermines chemoreflexes and promotes breathing instability during sleep.
Collapse
Affiliation(s)
- Alexandria B. Marciante
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Carter Lurk
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Luz Mata
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Jada Lewis
- Center for Translational Research in Neurodegenerative Diseases, Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Leah R. Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | - Gordon S. Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
9
|
Haynes RL, Trachtenberg F, Darnall R, Haas EA, Goldstein RD, Mena OJ, Krous HF, Kinney HC. Altered 5-HT2A/C receptor binding in the medulla oblongata in the sudden infant death syndrome (SIDS): Part I. Tissue-based evidence for serotonin receptor signaling abnormalities in cardiorespiratory- and arousal-related circuits. J Neuropathol Exp Neurol 2023; 82:467-482. [PMID: 37226597 PMCID: PMC10209647 DOI: 10.1093/jnen/nlad030] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023] Open
Abstract
The sudden infant death syndrome (SIDS), the leading cause of postneonatal infant mortality in the United States, is typically associated with a sleep period. Previously, we showed evidence of serotonergic abnormalities in the medulla (e.g. altered serotonin (5-HT)1A receptor binding), in SIDS cases. In rodents, 5-HT2A/C receptor signaling contributes to arousal and autoresuscitation, protecting brain oxygen status during sleep. Nonetheless, the role of 5-HT2A/C receptors in the pathophysiology of SIDS is unclear. We hypothesize that in SIDS, 5-HT2A/C receptor binding is altered in medullary nuclei that are key for arousal and autoresuscitation. Here, we report altered 5-HT2A/C binding in several key medullary nuclei in SIDS cases (n = 58) compared to controls (n = 12). In some nuclei the reduced 5-HT2A/C and 5-HT1A binding overlapped, suggesting abnormal 5-HT receptor interactions. The data presented here (Part 1) suggest that a subset of SIDS is due in part to abnormal 5-HT2A/C and 5-HT1A signaling across multiple medullary nuclei vital for arousal and autoresuscitation. In Part II to follow, we highlight 8 medullary subnetworks with altered 5-HT receptor binding in SIDS. We propose the existence of an integrative brainstem network that fails to facilitate arousal and/or autoresuscitation in SIDS cases.
Collapse
Affiliation(s)
- Robin L Haynes
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | | | - Ryan Darnall
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Elisabeth A Haas
- Department of Research, Rady Children’s Hospital, San Diego, California, USA
| | - Richard D Goldstein
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Othon J Mena
- San Diego County Medical Examiner Office, San Diego, California, USA
| | - Henry F Krous
- University of California, San Diego, San Diego, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Hannah C Kinney
- CJ Murphy Laboratory for SIDS Research, Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Robert’s Program on Sudden Unexpected Death in Pediatrics, Division of General Pediatrics, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Hao S, Shi W, Liu W, Chen QY, Zhuo M. Multiple modulatory roles of serotonin in chronic pain and injury-related anxiety. Front Synaptic Neurosci 2023; 15:1122381. [PMID: 37143481 PMCID: PMC10151796 DOI: 10.3389/fnsyn.2023.1122381] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Chronic pain is long-lasting pain that often persists during chronic diseases or after recovery from disease or injury. It often causes serious side effects, such as insomnia, anxiety, or depression which negatively impacts the patient's overall quality of life. Serotonin (5-HT) in the central nervous system (CNS) has been recognized as an important neurotransmitter and neuromodulator which regulates various physiological functions, such as pain sensation, cognition, and emotions-especially anxiety and depression. Its widespread and diverse receptors underlie the functional complexity of 5-HT in the CNS. Recent studies found that both chronic pain and anxiety are associated with synaptic plasticity in the anterior cingulate cortex (ACC), the insular cortex (IC), and the spinal cord. 5-HT exerts multiple modulations of synaptic transmission and plasticity in the ACC and the spinal cord, including activation, inhibition, and biphasic actions. In this review, we will discuss the multiple actions of the 5-HT system in both chronic pain and injury-related anxiety, and the synaptic mechanisms behind them. It is likely that the specific 5-HT receptors would be new promising therapeutic targets for the effective treatment of chronic pain and injury-related anxiety in the future.
Collapse
Affiliation(s)
- Shun Hao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
- International Institute of Brain Research, Forevercheer Medicine Pharmac Inc., Qingdao, Shandong, China
| | - Wantong Shi
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Weiqi Liu
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qi-Yu Chen
- International Institute of Brain Research, Forevercheer Medicine Pharmac Inc., Qingdao, Shandong, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Min Zhuo
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
- International Institute of Brain Research, Forevercheer Medicine Pharmac Inc., Qingdao, Shandong, China
- Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Bateman JT, Saunders SE, Levitt ES. Understanding and countering opioid-induced respiratory depression. Br J Pharmacol 2023; 180:813-828. [PMID: 34089181 PMCID: PMC8997313 DOI: 10.1111/bph.15580] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/06/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Respiratory depression is the proximal cause of death in opioid overdose, yet the mechanisms underlying this potentially fatal outcome are not well understood. The goal of this review is to provide a comprehensive understanding of the pharmacological mechanisms of opioid-induced respiratory depression, which could lead to improved therapeutic options to counter opioid overdose, as well as other detrimental effects of opioids on breathing. The development of tolerance in the respiratory system is also discussed, as are differences in the degree of respiratory depression caused by various opioid agonists. Finally, potential future therapeutic agents aimed at reversing or avoiding opioid-induced respiratory depression through non-opioid receptor targets are in development and could provide certain advantages over naloxone. By providing an overview of mechanisms and effects of opioids in the respiratory network, this review will benefit future research on countering opioid-induced respiratory depression. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Jordan T Bateman
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Sandy E Saunders
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Erica S Levitt
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, Florida, USA
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Kim DS, Pessah IN, Santana CM, Purnell BS, Li R, Buchanan GF, Rumbeiha WK. Investigations into hydrogen sulfide-induced suppression of neuronal activity in vivo and calcium dysregulation in vitro. Toxicol Sci 2023; 192:kfad022. [PMID: 36882182 PMCID: PMC10109532 DOI: 10.1093/toxsci/kfad022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Acute exposure to high concentrations of hydrogen sulfide (H2S) leads to sudden death and, if survived, lingering neurological disorders. Clinical signs include seizures, loss of consciousness, and dyspnea. The proximate mechanisms underlying H2S-induced acute toxicity and death have not been clearly elucidated. We investigated electrocerebral, cardiac and respiratory activity during H2S exposure using electroencephalogram (EEG), electrocardiogram (EKG) and plethysmography. H2S suppressed electrocerebral activity and disrupted breathing. Cardiac activity was comparatively less affected. To test whether Ca2+ dysregulation contributes to H2S-induced EEG suppression, we developed an in vitro real-time rapid throughput assay measuring patterns of spontaneous synchronized Ca2+ oscillations in cultured primary cortical neuronal networks loaded with the indicator Fluo-4 using the fluorescent imaging plate reader (FLIPR-Tetra®). Sulfide >5 ppm dysregulated synchronous calcium oscillation (SCO) patterns in a dose-dependent manner. Inhibitors of NMDA and AMPA receptors magnified H2S-induced SCO suppression. Inhibitors of L-type voltage gated Ca2+ channels and transient receptor potential channels prevented H2S-induced SCO suppression. Inhibitors of T-type voltage gated Ca2+ channels, ryanodine receptors, and sodium channels had no measurable influence on H2S-induced SCO suppression. Exposures to > 5 ppm sulfide also suppressed neuronal electrical activity in primary cortical neurons measured by multi-electrode array (MEA), an effect alleviated by pretreatment with the nonselective transient receptor potential channel inhibitor, 2-APB. 2-APB also reduced primary cortical neuronal cell death from sulfide exposure. These results improve our understanding of the role of different Ca2+ channels in acute H2S-induced neurotoxicity and identify transient receptor potential channel modulators as novel structures with potential therapeutic benefits.
Collapse
Affiliation(s)
- Dong-Suk Kim
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616, USA
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616, USA
| | - Cristina M Santana
- VDPAM, College of Veterinary Medicine, Iowa State University, Ames, Iowa 50011, USA
- MRIGlobal, Kansas City, Missouri 64110, USA
| | - Benton S Purnell
- Department of Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52246, USA
- Department of Nerosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Rui Li
- Department of Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52246, USA
| | - Gordon F Buchanan
- Department of Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52246, USA
| | - Wilson K Rumbeiha
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
13
|
Joyal KG, Petrucci AN, Littlepage-Saunders MV, Boodhoo NA, Wendt LH, Buchanan GF. Selective Serotonin Reuptake Inhibitors and 5-HT 2 Receptor Agonists Have Distinct, Sleep-state Dependent Effects on Postictal Breathing in Amygdala Kindled Mice. Neuroscience 2023; 513:76-95. [PMID: 36702372 PMCID: PMC9974756 DOI: 10.1016/j.neuroscience.2023.01.016] [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: 08/24/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
Seizures can cause profound breathing disruptions. Seizures arising from sleep cause greater breathing impairment than those emerging from wakefulness and more often result in sudden unexpected death in epilepsy (SUDEP). The neurotransmitter serotonin (5-HT) plays a major role in respiration and sleep-wake regulation. 5-HT modulates seizure susceptibility and severity and is dysregulated by seizures. Thus, the impact of seizures on breathing dysregulation may be due to impaired 5-HT neurotransmission. We examined whether pharmacologically increasing 5-HT neurotransmission prior to seizures improves postictal breathing and how sleep-state during seizure induction contributes to these effects. We assessed breathing with whole-body plethysmography in 84 amygdala-kindled mice pre-treated with selective serotonin reuptake inhibitors (SSRI) or 5-HT2 receptor agonists. SSRIs and 5-HT2 agonists increased postictal breathing frequency (fR), tidal volume (VT), and minute ventilation (VE) at different timepoints following seizures induced during wakefulness. These effects were not observed following seizures induced during NREM sleep. SSRIs suppressed ictal and postictal apnea regardless of sleep state. The SSRI citalopram and the 5-HT2 agonists TCB-2 and MK-212 decreased breathing variability following wake-occurring seizures at different postictal timepoints. Only MK-212 decreased breathing variability when seizures were induced during NREM sleep. The 5-HT2A antagonist MDL-11939 reduced the effect of citalopram on fR, VT, and VE, and enhanced its effect on breathing variability in the initial period following a seizure. These results suggest that 5-HT mechanisms that are dependent on or independent from the 5-HT2 family of receptors impact breathing on different timescales during the recovery of eupnea, and that certain serotonergic treatments may be less effective at facilitating postictal breathing following seizures emerging from sleep.
Collapse
Affiliation(s)
- Katelyn G Joyal
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Alexandra N Petrucci
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Mydirah V Littlepage-Saunders
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Nicole A Boodhoo
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Linder H Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA 52242, USA.
| | - Gordon F Buchanan
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
14
|
Santana Maldonado CM, Kim DS, Purnell B, Li R, Buchanan GF, Smith J, Thedens DR, Gauger P, Rumbeiha WK. Acute hydrogen sulfide-induced neurochemical and morphological changes in the brainstem. Toxicology 2023; 485:153424. [PMID: 36610655 DOI: 10.1016/j.tox.2023.153424] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Hydrogen sulfide (H2S) is a toxin affecting the cardiovascular, respiratory, and central nervous systems. Acute H2S exposure is associated with a high rate of mortality and morbidity. The precise pathophysiology of H2S-induced death is a controversial topic; however, inhibition of the respiratory center in the brainstem is commonly cited as a cause of death. There is a knowledge gap on toxicity and toxic mechanisms of acute H2S poisoning on the brainstem, a brain region responsible for regulating many reflective and vital functions. Serotonin (5-HT), dopamine (DA), and γ-aminobutyric acid (GABA) play a role in maintaining a normal stable respiratory rhythmicity. We hypothesized that the inhibitory respiratory effects of H2S poisoning are mediated by 5-HT in the respiratory center of the brainstem. Male C57BL/6 mice were exposed once to an LCt50 concentration of H2S (1000 ppm). Batches of surviving mice were euthanized at 5 min, 2 h, 12 h, 24 h, 72 h, and on day 7 post-exposure. Pulmonary function, vigilance state, and mortality were monitored during exposure. The brainstem was analyzed for DA, 3,4-dehydroxyphenyl acetic acid (DOPAC), 5-HT, 5-hydroxyindoleatic acid (5-HIAA), norepinephrine (NE), GABA, glutamate, and glycine using HPLC. Enzymatic activities of monoamine oxidases (MAO) were also measured in the brainstem using commercial kits. Neurodegeneration was assessed using immunohistochemistry and magnetic resonance imaging. Results showed that DA and DOPAC were significantly increased at 5 min post H2S exposure. However, by 2 h DA returned to normal. Activities of MAO were significantly increased at 5 min and 2 h post-exposure. In contrast, NE was significantly decreased at 5 min and 2 h post-exposure. Glutamate was overly sensitive to H2S-induced toxicity manifesting a time-dependent concentration reduction throughout the 7 day duration of the study. Remarkably, there were no changes in 5-HT, 5-HIAA, glycine, or GABA concentrations. Cytochrome c oxidase activity was inhibited but recovered by 24 h. Neurodegeneration was observed starting at 72 h post H2S exposure in select brainstem regions. We conclude that acute H2S exposure causes differential effects on brainstem neurotransmitters. H2S also induces neurodegeneration and biochemical changes in the brainstem. Additional work is needed to fully understand the implications of both the short- and long-term effects of acute H2S poisoning on vital functions regulated by the brainstem.
Collapse
Affiliation(s)
- Cristina M Santana Maldonado
- Veterinary Diagnostic Production and Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA.
| | - Dong-Suk Kim
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| | - Benton Purnell
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| | - Rui Li
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| | - Gordon F Buchanan
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| | - Jodi Smith
- Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA.
| | - Daniel R Thedens
- Seamans Center for the Engineering Arts and Sciences, Iowa City, IA 52242, USA.
| | - Phillip Gauger
- Veterinary Diagnostic Production and Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA.
| | - Wilson K Rumbeiha
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| |
Collapse
|
15
|
Cáceres D, Ochoa M, González-Ortiz M, Bravo K, Eugenín J. Effects of Prenatal Cannabinoids Exposure upon Placenta and Development of Respiratory Neural Circuits. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:199-232. [PMID: 37466775 DOI: 10.1007/978-3-031-32554-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Cannabis use has risen dangerously during pregnancy in the face of incipient therapeutic use and a growing perception of safety. The main psychoactive compound of the Cannabis sativa plant is the phytocannabinoid delta-9-tetrahydrocannabinol (A-9 THC), and its status as a teratogen is controversial. THC and its endogenous analogues, anandamide (AEA) and 2-AG, exert their actions through specific receptors (eCBr) that activate intracellular signaling pathways. CB1r and CB2r, also called classic cannabinoid receptors, together with their endogenous ligands and the enzymes that synthesize and degrade them, constitute the endocannabinoid system. This system is distributed ubiquitously in various central and peripheral tissues. Although the endocannabinoid system's most studied role is controlling the release of neurotransmitters in the central nervous system, the study of long-term exposure to cannabinoids on fetal development is not well known and is vital for understanding environmental or pathological embryo-fetal or postnatal conditions. Prenatal exposure to cannabinoids in animal models has induced changes in placental and embryo-fetal organs. Particularly, cannabinoids could influence both neural and nonneural tissues and induce embryo-fetal pathological conditions in critical processes such as neural respiratory control. This review aims at the acute and chronic effects of prenatal exposure to cannabinoids on placental function and the embryo-fetal neurodevelopment of the respiratory pattern. The information provided here will serve as a theoretical framework to critically evaluate the teratogen effects of the consumption of cannabis during pregnancy.
Collapse
Affiliation(s)
- Daniela Cáceres
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Martín Ochoa
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Karina Bravo
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Facultad de Ingeniería, Universidad Autónoma de Chile, Providencia, Chile
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
16
|
Casciato A, Bianchi L, Reverdy M, Joubert F, Delucenay-Clarke R, Parrot S, Ramanantsoa N, Sizun E, Matrot B, Straus C, Similowski T, Cayetanot F, Bodineau L. Serotonin and the ventilatory effects of etonogestrel, a gonane progestin, in a murine model of congenital central hypoventilation syndrome. Front Endocrinol (Lausanne) 2023; 14:1077798. [PMID: 36896185 PMCID: PMC9989262 DOI: 10.3389/fendo.2023.1077798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/06/2023] [Indexed: 02/23/2023] Open
Abstract
INTRODUCTION Congenital Central Hypoventilation Syndrome, a rare disease caused by PHOX2B mutation, is associated with absent or blunted CO2/H+ chemosensitivity due to the dysfunction of PHOX2B neurons of the retrotrapezoid nucleus. No pharmacological treatment is available. Clinical observations have reported non-systematic CO2/H+ chemosensitivity recovery under desogestrel. METHODS Here, we used a preclinical model of Congenital Central Hypoventilation Syndrome, the retrotrapezoid nucleus conditional Phox2b mutant mouse, to investigate whether etonogestrel, the active metabolite of desogestrel, led to a restoration of chemosensitivity by acting on serotonin neurons known to be sensitive to etonogestrel, or retrotrapezoid nucleus PHOX2B residual cells that persist despite the mutation. The influence of etonogestrel on respiratory variables under hypercapnia was investigated using whole-body plethysmographic recording. The effect of etonogestrel, alone or combined with serotonin drugs, on the respiratory rhythm of medullary-spinal cord preparations from Phox2b mutants and wildtype mice was analyzed under metabolic acidosis. c-FOS, serotonin and PHOX2B were immunodetected. Serotonin metabolic pathways were characterized in the medulla oblongata by ultra-high-performance liquid chromatography. RESULTS We observed etonogestrel restored chemosensitivity in Phox2b mutants in a non-systematic way. Histological differences between Phox2b mutants with restored chemosensitivity and Phox2b mutant without restored chemosensitivity indicated greater activation of serotonin neurons of the raphe obscurus nucleus but no effect on retrotrapezoid nucleus PHOX2B residual cells. Finally, the increase in serotonergic signaling by the fluoxetine application modulated the respiratory effect of etonogestrel differently between Phox2b mutant mice and their WT littermates or WT OF1 mice, a result which parallels with differences in the functional state of serotonergic metabolic pathways between these different mice. DISCUSSION Our work thus highlights that serotonin systems were critically important for the occurrence of an etonogestrel-restoration, an element to consider in potential therapeutic intervention in Congenital Central Hypoventilation Syndrome patients.
Collapse
Affiliation(s)
- Alexis Casciato
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Lola Bianchi
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Manon Reverdy
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Fanny Joubert
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Roman Delucenay-Clarke
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Sandrine Parrot
- Centre de Recherche en Neurosciences, NeuroDialyTics, Bron, France
| | | | - Eléonore Sizun
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Boris Matrot
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Christian Straus
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Thomas Similowski
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Florence Cayetanot
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
| | - Laurence Bodineau
- Sorbonne Université, Inserm, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Paris, France
- *Correspondence: Laurence Bodineau,
| |
Collapse
|
17
|
Yang G, Wu G, Yao W, Guan L, Geng X, Liu J, Liu Z, Yang L, Huang Q, Zeng X, Yang P. 5-HT is associated with the dysfunction of regulating T cells in patients with allergic rhinitis. Clin Immunol 2022; 243:109101. [PMID: 36029976 DOI: 10.1016/j.clim.2022.109101] [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: 06/14/2022] [Revised: 08/06/2022] [Accepted: 08/20/2022] [Indexed: 11/03/2022]
Abstract
The dysfunction of regulating T lymphocytes (Treg) is associated with the pathogenesis of many diseases. Serotonin (5-HT) is capable of interacting with immune cells. The objective of the present study is to shed light on the role of 5-HT in regulating Treg activities. Blood samples were collected from patients with perennial allergic rhinitis (AR). Tregs were isolated from blood samples by magnetic cell sorting. The levels of 5-HT and other cytokines were determined by enzyme-linked immunosorbent assay. The results showed that serum 5-HT levels in patients with AR were higher than in healthy control (HC) subjects. A positive correlation was identified in the data between 5-HT concentrations and AR-related cytokine concentrations in the serum. A negative correlation was found between serum levels of 5-HT and the peripheral frequency of Treg. Exposure to 5-HT enhanced the expression of IL-6 and IL-21 in dendritic (DC) cells. Co-culture of 5-HT-primed DCs with Tregs led to the conversion of Th17 cells. STAT3 blockade efficiently abolished the 5-HT-associated conversion of Th17 cells from Tregs. In summary, patients with AR exhibited higher serum concentrations of 5-HT. 5-HT-primed DCs could convert Tregs to Th17 cells.
Collapse
Affiliation(s)
- Gui Yang
- Department of Otolaryngology, Longgang Central Hospital, Shenzhen, China
| | - Gaohui Wu
- Departments of Respirology and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wenkai Yao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Li Guan
- Departments of Respirology and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiaorui Geng
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T, Shenzhen, China
| | - Jiangqi Liu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T, Shenzhen, China
| | - Zhiqiang Liu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T, Shenzhen, China
| | - Liteng Yang
- Departments of Respirology and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qinmiao Huang
- Departments of Respirology and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T, Shenzhen, China
| | - Pingchang Yang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China.
| |
Collapse
|
18
|
Mouradian GC, Liu P, Nakagawa P, Duffy E, Gomez Vargas J, Balapattabi K, Grobe JL, Sigmund CD, Hodges MR. Patch-to-Seq and Transcriptomic Analyses Yield Molecular Markers of Functionally Distinct Brainstem Serotonin Neurons. Front Synaptic Neurosci 2022; 14:910820. [PMID: 35844900 PMCID: PMC9280690 DOI: 10.3389/fnsyn.2022.910820] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/10/2022] [Indexed: 01/22/2023] Open
Abstract
Acute regulation of CO2 and pH homeostasis requires sensory feedback from peripheral (carotid body) and central (central) CO2/pH sensitive cells - so called respiratory chemoreceptors. Subsets of brainstem serotonin (5-HT) neurons in the medullary raphe are CO2 sensitive or insensitive based on differences in embryonic origin, suggesting these functionally distinct subpopulations may have unique transcriptional profiles. Here, we used Patch-to-Seq to determine if the CO2 responses in brainstem 5-HT neurons could be correlated to unique transcriptional profiles and/or unique molecular markers and pathways. First, firing rate changes with hypercapnic acidosis were measured in fluorescently labeled 5-HT neurons in acute brainstem slices from transgenic, Dahl SS (SSMcwi) rats expressing T2/ePet-eGFP transgene in Pet-1 expressing (serotonin) neurons (SS ePet1-eGFP rats). Subsequently, the transcriptomic and pathway profiles of CO2 sensitive and insensitive 5-HT neurons were determined and compared by single cell RNA (scRNAseq) and bioinformatic analyses. Low baseline firing rates were a distinguishing feature of CO2 sensitive 5-HT neurons. scRNAseq of these recorded neurons revealed 166 differentially expressed genes among CO2 sensitive and insensitive 5-HT neurons. Pathway analyses yielded novel predicted upstream regulators, including the transcription factor Egr2 and Leptin. Additional bioinformatic analyses identified 6 candidate gene markers of CO2 sensitive 5-HT neurons, and 2 selected candidate genes (CD46 and Iba57) were both expressed in 5-HT neurons determined via in situ mRNA hybridization. Together, these data provide novel insights into the transcriptional control of cellular chemoreception and provide unbiased candidate gene markers of CO2 sensitive 5-HT neurons. Methodologically, these data highlight the utility of the patch-to-seq technique in enabling the linkage of gene expression to specific functions, like CO2 chemoreception, in a single cell to identify potential mechanisms underlying functional differences in otherwise similar cell types.
Collapse
Affiliation(s)
- Gary C. Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States,*Correspondence: Gary C. Mouradian Jr.,
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Erin Duffy
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Javier Gomez Vargas
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kirthikaa Balapattabi
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States,Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Matthew R. Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
19
|
Biancardi V, Patrone LGA, Vicente MC, Marques DA, Bicego KC, Funk GD, Gargaglioni LH. Prenatal fluoxetine has long lasting, differential effects on respiratory control in male and female rats. J Appl Physiol (1985) 2022; 133:371-389. [PMID: 35708704 DOI: 10.1152/japplphysiol.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Serotonin (5-HT) is an important modulator of brain networks that control breathing. The selective serotonin reuptake inhibitor fluoxetine (FLX) is the first-line antidepressant drug prescribed during pregnancy. We investigated the effects of prenatal FLX on baseline breathing, ventilatory and metabolic responses to hypercapnia and hypoxia as well as number of brainstem 5-HT and tyrosine hydroxylase (TH) neurons of rats during postnatal development (P0-82). Prenatal FLX exposure of males showed a lower baseline that appeared in juveniles and remained in adulthood, with no sleep-wake state dependency. Prenatal FLX exposure of females did not affect baseline breathing. Juvenile male FLX rats showed increased CO2 and hypoxic ventilatory responses, normalizing by adulthood. Alterations in juvenile-FLX treated males were associated with greater number of 5-HT neurons in the ROB and RMAG. Adult FLX-exposed males showed greater number of 5-HT neurons in the RPA and TH neurons in the A5, while reduced number of TH neurons in A7. Prenatal FLX exposure of female rats was associated with greater hyperventilation induced by hypercapnia at P0-2 and juveniles whereas P12-14 and adult FLX (NREM sleep) rats showed an attenuation of the hypercapnic hyperventilation.FLX-exposed females had fewer 5-HT neurons in the RPA and reduced TH A6 density at P0-2; and greater number of TH neurons in the A7 at P12-14. These data indicate that prenatal FLX exposure affects the number of neurons of some monoaminergic regions in the brain and results in long lasting, sex specific changes in baseline breathing pattern and ventilatory responses to respiratory challenges.
Collapse
Affiliation(s)
- Vivian Biancardi
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,Department of Physiology, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Mariane C Vicente
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Danuzia A Marques
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Kênia C Bicego
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Gregory D Funk
- Department of Physiology, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| |
Collapse
|
20
|
Pho H, Amorim MR, Qiu Q, Shin M, Kim LJ, Anokye‐Danso F, Jun JJ, Ahima RS, Branco LGS, Kuhn DM, Mateika JH, Polotsky VY. The effect of brain serotonin deficiency on breathing is magnified by age. Physiol Rep 2022; 10:e15245. [PMID: 35581741 PMCID: PMC9114658 DOI: 10.14814/phy2.15245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/25/2022] [Accepted: 03/13/2022] [Indexed: 11/24/2022] Open
Abstract
Serotonin is an important mediator modulating behavior, metabolism, sleep, control of breathing, and upper airway function, but the role of aging in serotonin-mediated effects has not been previously defined. Our study aimed to examine the effect of brain serotonin deficiency on breathing during sleep and metabolism in younger and older mice. We measured breathing during sleep, hypercapnic ventilatory response (HCVR), CO2 production (VCO2 ), and O2 consumption (VO2 ) in 16-18-week old and 40-44-week old mice with deficiency of tryptophan hydroxylase 2 (Tph2), which regulates serotonin synthesis specifically in neurons, compared to Tph2+/+ mice. As expected, aging decreased VCO2 and VO2 . Tph2 knockout resulted in an increase in both metabolic indexes and no interaction between age and the genotype was observed. During wakefulness, neither age nor genotype had an effect on minute ventilation. The genotype did not affect hypercapnic sensitivity in younger mice. During sleep, Tph2-/- mice showed significant decreases in maximal inspiratory flow in NREM sleep, respiratory rate, and oxyhemoglobin saturation in REM sleep, compared to wildtype, regardless of age. Neither serotonin deficiency nor aging affected the frequency of flow limited breaths (a marker of upper airway closure) or apneas. Serotonin deficiency increased the amount and efficiency of sleep only in older animals. In conclusion, younger Tph2-/- mice were able to defend their ventilation and phenotypically did not differ from wildtype during wakefulness. In contrast, both young and old Tph2-/- mice showed sleep-related hypoventilation, which was manifested by hypoxemia during REM sleep.
Collapse
Affiliation(s)
- Huy Pho
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mateus R. Amorim
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Qingchao Qiu
- Department of PhysiologyWayne State UniversityDetroitMichiganUSA
| | - Mi‐Kyung Shin
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lenise J. Kim
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Frederick Anokye‐Danso
- Division of Endocrinology, Diabetes, and MetabolismDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jonathan J. Jun
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Rexford S. Ahima
- Division of Endocrinology, Diabetes, and MetabolismDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Luiz G. S. Branco
- Dental School of Ribeirão PretoUniversity of São PauloSão PauloBrazil
| | - Donald M. Kuhn
- Department of Psychiatry and Behavioral NeurosciencesWayne State University School of MedicineDetroitMichiganUSA
- John D. Dingell Veterans Affairs Medical CenterDetroitMichiganUSA
| | - Jason H. Mateika
- Department of PhysiologyWayne State UniversityDetroitMichiganUSA
- John D. Dingell Veterans Affairs Medical CenterDetroitMichiganUSA
- Department of Internal MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
21
|
Wollman L, Hill A, Hasse B, Young C, Hernandez-De La Pena G, Levine RB, Fregosi RF. Influence of developmental nicotine exposure on serotonergic control of breathing-related motor output. Dev Neurobiol 2022; 82:175-191. [PMID: 35016263 PMCID: PMC8940681 DOI: 10.1002/dneu.22866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/06/2021] [Accepted: 01/04/2022] [Indexed: 11/07/2022]
Abstract
Serotonin plays an important role in the development of brainstem circuits that control breathing. Here, we test the hypothesis that developmental nicotine exposure (DNE) alters the breathing-related motor response to serotonin (5HT). Pregnant rats were exposed to nicotine or saline, and brainstem-spinal cord preparations from 1- to 5-day-old pups were studied in a split-bath configuration, allowing drugs to be applied selectively to the medulla or spinal cord. The activity of the fourth cervical ventral nerve roots (C4VR), which contain axons of phrenic motoneurons, was recorded. We applied 5HT alone or together with antagonists of 5HT1A, 5HT2A, or 5HT7 receptor subtypes. In control preparations, 5HT applied to the medulla consistently reduced C4VR frequency and this reduction could not be blocked by any of the three antagonists. In DNE preparations, medullary 5HT caused a large and sustained frequency increase (10 min), followed by a sustained decrease. Notably, the transient increase in frequency could be blocked by the independent addition of any of the antagonists. Experiments with subtype-specific agonists suggest that the 5HT7 subtype may contribute to the increased frequency response in the DNE preparations. Changes in C4VR burst amplitude in response to brainstem 5HT were uninfluenced by DNE. Addition of 5HT to the caudal chamber modestly increased phasic and greatly increased tonic C4VR activity, but there were no effects of DNE. The data show that DNE alters serotonergic signaling within brainstem circuits that control respiratory frequency but does not functionally alter serotonin signaling in the phrenic motoneuron pool.
Collapse
Affiliation(s)
- Lila Wollman
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ
| | - Andrew Hill
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ
| | - Brady Hasse
- Department of Neuroscience, University of Arizona, Tucson, AZ
| | - Christina Young
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ
| | | | - Richard B Levine
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ,Department of Neuroscience, University of Arizona, Tucson, AZ
| | - Ralph F. Fregosi
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ,Department of Neuroscience, University of Arizona, Tucson, AZ
| |
Collapse
|
22
|
The lamprey respiratory network: Some evolutionary aspects. Respir Physiol Neurobiol 2021; 294:103766. [PMID: 34329767 DOI: 10.1016/j.resp.2021.103766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/19/2021] [Accepted: 07/25/2021] [Indexed: 01/25/2023]
Abstract
Breathing is a complex behaviour that involves rhythm generating networks. In this review, we examine the main characteristics of respiratory rhythm generation in vertebrates and, in particular, we describe the main results of our studies on the role of neural mechanisms involved in the neuromodulation of the lamprey respiration. The lamprey respiratory rhythm generator is located in the paratrigeminal respiratory group (pTRG) and shows similarities with the mammalian preBötzinger complex. In fact, within the pTRG a major role is played by glutamate, but also GABA and glycine display important contributions. In addition, neuromodulatory influences are exerted by opioids, substance P, acetylcholine and serotonin. Both structures respond to exogenous ATP with a biphasic response and astrocytes there located strongly contribute to the modulation of the respiratory pattern. The results emphasize that some important characteristics of the respiratory rhythm generating network are, to a great extent, maintained throughout evolution.
Collapse
|
23
|
Cinelli E, Mutolo D, Pantaleo T, Bongianni F. Neural mechanisms underlying respiratory regulation within the preBötzinger complex of the rabbit. Respir Physiol Neurobiol 2021; 293:103736. [PMID: 34224867 DOI: 10.1016/j.resp.2021.103736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 11/29/2022]
Abstract
The preBötzinger complex (preBötC) is a medullary area essential for normal breathing and widely recognized as necessary and sufficient to generate the inspiratory phase of respiration. It has been studied mainly in rodents. Here we report the main results of our studies revealing the characteristics of the rabbit preBötC identified by means of neuronal recordings, D,L-homocysteic acid microinjections and histological controls. A crucial role in the respiratory rhythmogenesis within this neural substrate is played by excitatory amino acids, but also GABA and glycine display important contributions. Increases in respiratory frequency are induced by microinjections of neurokinins, somatostatin as well by serotonin (5-HT) through an action on 5-HT1A and 5-HT3 receptors or the disinhibition of a GABAergic circuit. Respiratory depression is observed in response to microinjections of the μ-opioid receptor agonist DAMGO. Our results show similarities and differences with the rodent preBötC and emphasize the importance of comparative studies on the mechanisms underlying respiratory rhythmogenesis in different animal species.
Collapse
Affiliation(s)
- Elenia Cinelli
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università Degli Studi di Firenze, Viale G.B. Morgagni 63, Firenze, 50134, Italy
| | - Donatella Mutolo
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università Degli Studi di Firenze, Viale G.B. Morgagni 63, Firenze, 50134, Italy
| | - Tito Pantaleo
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università Degli Studi di Firenze, Viale G.B. Morgagni 63, Firenze, 50134, Italy
| | - Fulvia Bongianni
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università Degli Studi di Firenze, Viale G.B. Morgagni 63, Firenze, 50134, Italy.
| |
Collapse
|
24
|
Mouradian GC, Kilby M, Alvarez S, Kaplan K, Hodges MR. Mortality and ventilatory effects of central serotonin deficiency during postnatal development depend on age but not sex. Physiol Rep 2021; 9:e14946. [PMID: 34228894 PMCID: PMC8259800 DOI: 10.14814/phy2.14946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/24/2022] Open
Abstract
Serotonin (5-HT) influences brain development and has predominantly excitatory neuromodulatory effects on the neural respiratory control circuitry. Infants that succumb to sudden infant death syndrome (SIDS) have reduced brainstem 5-HT levels and Tryptophan hydroxylase 2 (Tph2). Furthermore, there are age- and sex-dependent risk factors associated with SIDS. Here we utilized our established Dark Agouti transgenic rat lacking central serotonin KO to test the hypotheses that CNS 5-HT deficiency leads to: (1) high mortality in a sex-independent manner, (2) age-dependent alterations in other CNS aminergic systems, and (3) age-dependent impairment of chemoreflexes during post-natal development. KO rat pups showed high neonatal mortality but not in a sex-dependent manner and did not show altered hypoxic or hypercapnic ventilatory chemoreflexes. However, KO rat pups had increased apnea-related metrics during a specific developmental age (P12-16), which were preceded by transient increases in dopaminergic system activity (P7-8). These results support and extend the concept that 5-HT per se is a critical factor in supporting respiratory control during post-natal development.
Collapse
Affiliation(s)
- Gary C. Mouradian
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Madeline Kilby
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
| | - Santiago Alvarez
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
| | - Kara Kaplan
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
| | - Matthew R. Hodges
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWIUSA
| |
Collapse
|
25
|
James N, Bell A. Minimally invasive brain injections for viral-mediated transgenesis: New tools for behavioral genetics in sticklebacks. PLoS One 2021; 16:e0251653. [PMID: 33999965 PMCID: PMC8128275 DOI: 10.1371/journal.pone.0251653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023] Open
Abstract
Behavioral genetics in non-model organisms is currently gated by technological limitations. However, with the growing availability of genome editing and functional genomic tools, complex behavioral traits such as social behavior can now be explored in diverse organisms. Here we present a minimally invasive neurosurgical procedure for a classic behavioral, ecological and evolutionary system: threespine stickleback (Gasterosteus aculeatus). Direct brain injection enables viral-mediated transgenesis and pharmaceutical delivery which bypasses the blood-brain barrier. This method is flexible, fast, and amenable to statistically powerful within-subject experimental designs, making it well-suited for use in genetically diverse animals such as those collected from natural populations. Developing this minimally invasive neurosurgical protocol required 1) refining the anesthesia process, 2) building a custom surgical rig, and 3) determining the normal recovery pattern allowing us to clearly identify warning signs of failure to thrive. Our custom-built surgical rig (publicly available) and optimized anesthetization methods resulted in high (90%) survival rates and quick behavioral recovery. Using this method, we detected changes in aggression from the overexpression of either of two different genes, arginine vasopressin (AVP) and monoamine oxidase (MAOA), in outbred animals in less than one month. We successfully used multiple promoters to drive expression, allowing for tailored expression profiles through time. In addition, we demonstrate that widely available mammalian plasmids work with this method, lowering the barrier of entry to the technique. By using repeated measures of behavior on the same fish before and after transfection, we were able to drastically reduce the necessary sample size needed to detect significant changes in behavior, making this a viable approach for examining genetic mechanisms underlying complex social behaviors.
Collapse
Affiliation(s)
- Noelle James
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Evolution, Ecology and Behavior, University of Illinois at Urbana, Urbana, Illinois, United States of America
| | - Alison Bell
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Evolution, Ecology and Behavior, University of Illinois at Urbana, Urbana, Illinois, United States of America
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Program in Ecology, Evolution and Conservation Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| |
Collapse
|
26
|
Abstract
The development of the control of breathing begins in utero and continues postnatally. Fetal breathing movements are needed for establishing connectivity between the lungs and central mechanisms controlling breathing. Maturation of the control of breathing, including the increase of hypoxia chemosensitivity, continues postnatally. Insufficient oxygenation, or hypoxia, is a major stressor that can manifest for different reasons in the fetus and neonate. Though the fetus and neonate have different hypoxia sensing mechanisms and respond differently to acute hypoxia, both responses prevent deviations to respiratory and other developmental processes. Intermittent and chronic hypoxia pose much greater threats to the normal developmental respiratory processes. Gestational intermittent hypoxia, due to maternal sleep-disordered breathing and sleep apnea, increases eupneic breathing and decreases the hypoxic ventilatory response associated with impaired gasping and autoresuscitation postnatally. Chronic fetal hypoxia, due to biologic or environmental (i.e. high-altitude) factors, is implicated in fetal growth restriction and preterm birth causing a decrease in the postnatal hypoxic ventilatory responses with increases in irregular eupneic breathing. Mechanisms driving these changes include delayed chemoreceptor development, catecholaminergic activity, abnormal myelination, increased astrocyte proliferation in the dorsal respiratory group, among others. Long-term high-altitude residents demonstrate favorable adaptations to chronic hypoxia as do their offspring. Neonatal intermittent hypoxia is common among preterm infants due to immature respiratory systems and thus, display a reduced drive to breathe and apneas due to insufficient hypoxic sensitivity. However, ongoing intermittent hypoxia can enhance hypoxic sensitivity causing ventilatory overshoots followed by apnea; the number of apneas is positively correlated with degree of hypoxic sensitivity in preterm infants. Chronic neonatal hypoxia may arise from fetal complications like maternal smoking or from postnatal cardiovascular problems, causing blunting of the hypoxic ventilatory responses throughout at least adolescence due to attenuation of carotid body fibers responses to hypoxia with potential roles of brainstem serotonin, microglia, and inflammation, though these effects depend on the age in which chronic hypoxia initiates. Fetal and neonatal intermittent and chronic hypoxia are implicated in preterm birth and complicate the respiratory system through their direct effects on hypoxia sensing mechanisms and interruptions to the normal developmental processes. Thus, precise regulation of oxygen homeostasis is crucial for normal development of the respiratory control network. © 2021 American Physiological Society. Compr Physiol 11:1653-1677, 2021.
Collapse
Affiliation(s)
- Gary C. Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Satyan Lakshminrusimha
- Department of Pediatrics, UC Davis Children’s Hospital, UC Davis Health, UC Davis, Davis, California, USA
| | - Girija G. Konduri
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
27
|
Moreira TS, Sobrinho CR, Falquetto B, Oliveira LM, Lima JD, Mulkey DK, Takakura AC. The retrotrapezoid nucleus and the neuromodulation of breathing. J Neurophysiol 2020; 125:699-719. [PMID: 33427575 DOI: 10.1152/jn.00497.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Breathing is regulated by a host of arousal and sleep-wake state-dependent neuromodulators to maintain respiratory homeostasis. Modulators such as acetylcholine, norepinephrine, histamine, serotonin (5-HT), adenosine triphosphate (ATP), substance P, somatostatin, bombesin, orexin, and leptin can serve complementary or off-setting functions depending on the target cell type and signaling mechanisms engaged. Abnormalities in any of these modulatory mechanisms can destabilize breathing, suggesting that modulatory mechanisms are not overly redundant but rather work in concert to maintain stable respiratory output. The present review focuses on the modulation of a specific cluster of neurons located in the ventral medullary surface, named retrotrapezoid nucleus, that are activated by changes in tissue CO2/H+ and regulate several aspects of breathing, including inspiration and active expiration.
Collapse
Affiliation(s)
- Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, Brazil
| | - Cleyton R Sobrinho
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, Brazil
| | - Barbara Falquetto
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, Brazil
| | - Luiz M Oliveira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, Brazil
| | - Janayna D Lima
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, Brazil
| | - Daniel K Mulkey
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, Brazil
| |
Collapse
|
28
|
Avraam J, Wu Y, Richerson GB. Perinatal Nicotine Reduces Chemosensitivity of Medullary 5-HT Neurons after Maturation in Culture. Neuroscience 2020; 446:80-93. [PMID: 32818601 DOI: 10.1016/j.neuroscience.2020.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 01/19/2023]
Abstract
Perinatal exposure to nicotine produces ventilatory and chemoreflex deficits in neonatal mammals. Medullary 5-HT neurons are putative central chemoreceptors that innervate respiratory nuclei and promote ventilation, receive cholinergic input and express nicotinic acetylcholine receptors (nAChRs). Perforated patch clamp recordings were made from cultured 5-HT neurons dissociated from the medullary raphé of 0-3 day old mice expressing enhanced yellow fluorescent protein driven by the enhancer region for PET1 (ePet-EYFP). The effect of exposure to low (6 mg kg-1day-1) or high (60 mg kg-1day-1) doses of nicotine in utero (prenatal), in culture (postnatal), or both and the effect of acute nicotine exposure (10 μM), were examined on baseline firing rate (FR at 5% CO2, pH = 7.4) and the change in FR with acidosis (9% CO2, pH 7.2) in young (12-21 days in vitro, DIV) and older (≥22 DIV) acidosis stimulated 5-HT neurons. Nicotine exposed neurons exhibited ∼67% of the response to acidosis recorded in neurons given vehicle (p = 0.005), with older neurons exposed to high dose prenatal and postnatal nicotine, exhibiting only 28% of that recorded in the vehicle neurons (p < 0.01). In neurons exposed to low or high dose prenatal and postnatal nicotine, acute nicotine exposure led to a smaller increase in FR (∼+51% vs +168%, p = 0.026) and response to acidosis (+6% vs +67%, p = 0.014) compared to vehicle. These data show that exposure to nicotine during development reduces chemosensitivity of 5-HT neurons as they mature, an effect that may be related to the abnormal chemoreflexes reported in rodents exposed to nicotine in utero, and may cause a greater risk for sudden infant death syndrome (SIDS).
Collapse
Affiliation(s)
- Joanne Avraam
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States; Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Yuanming Wu
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States
| | - George Bradley Richerson
- Department of Neurology, University of Iowa, Iowa City, IA 52242, United States; Veteran's Affairs Medical Center, Iowa City, IA 52242, United States; Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
29
|
Quipazine Elicits Swallowing in the Arterially Perfused Rat Preparation: A Role for Medullary Raphe Nuclei? Int J Mol Sci 2020; 21:ijms21145120. [PMID: 32698469 PMCID: PMC7404031 DOI: 10.3390/ijms21145120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/06/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
Pharmacological neuromodulation of swallowing may represent a promising therapeutic option to treat dysphagia. Previous studies suggested a serotonergic control of swallowing, but mechanisms remain poorly understood. Here, we investigated the effects of the serotonergic agonist quipazine on swallowing, using the arterially perfused working heart-brainstem (in situ) preparation in rats. Systemic injection of quipazine produced single swallows with motor patterns and swallow-breathing coordination similar to spontaneous swallows, and increased swallow rate with moderate changes in cardiorespiratory functions. Methysergide, a 5-HT2 receptor antagonist, blocked the excitatory effect of quipazine on swallowing, but had no effect on spontaneous swallow rate. Microinjections of quipazine in the nucleus of the solitary tract were without effect. In contrast, similar injections in caudal medullary raphe nuclei increased swallow rate without changes in cardiorespiratory parameters. Thus, quipazine may exert an excitatory effect on raphe neurons via stimulation of 5-HT2A receptors, leading to increased excitability of the swallowing network. In conclusion, we suggest that pharmacological stimulation of swallowing by quipazine in situ represents a valuable model for experimental studies. This work paves the way for future investigations on brainstem serotonergic modulation, and further identification of neural populations and mechanisms involved in swallowing and/or swallow-breathing interaction.
Collapse
|
30
|
Varga AG, Maletz SN, Bateman JT, Reid BT, Levitt ES. Neurochemistry of the Kölliker-Fuse nucleus from a respiratory perspective. J Neurochem 2020; 156:16-37. [PMID: 32396650 DOI: 10.1111/jnc.15041] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
The Kölliker-Fuse nucleus (KF) is a functionally distinct component of the parabrachial complex, located in the dorsolateral pons of mammals. The KF has a major role in respiration and upper airway control. A comprehensive understanding of the KF and its contributions to respiratory function and dysfunction requires an appreciation for its neurochemical characteristics. The goal of this review is to summarize the diverse neurochemical composition of the KF, focusing on the neurotransmitters, neuromodulators, and neuropeptides present. We also include a description of the receptors expressed on KF neurons and transporters involved in each system, as well as their putative roles in respiratory physiology. Finally, we provide a short section reviewing the literature regarding neurochemical changes in the KF in the context of respiratory dysfunction observed in SIDS and Rett syndrome. By over-viewing the current literature on the neurochemical composition of the KF, this review will serve to aid a wide range of topics in the future research into the neural control of respiration in health and disease.
Collapse
Affiliation(s)
- Adrienn G Varga
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Sebastian N Maletz
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jordan T Bateman
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Brandon T Reid
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Erica S Levitt
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA.,Department of Physical Therapy, Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL, USA
| |
Collapse
|
31
|
Cinelli E, Mutolo D, Iovino L, Pantaleo T, Bongianni F. Key role of 5-HT 1A receptors in the modulation of the neuronal network underlying the respiratory rhythm generation in lampreys. Eur J Neurosci 2020; 52:3903-3917. [PMID: 32378271 DOI: 10.1111/ejn.14769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/06/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022]
Abstract
In mammals, 5-HTexcitatory respiratory effects imply 5-HT1A receptor-mediated disinhibition of pre-Bötzinger complex neurons. In the lamprey, 5-HT1A receptors are involved in the neural control of locomotion, but their role in the respiratory regulation, particularly at the level of the putative respiratory rhythm generator, the paratrigeminal respiratory group (pTRG), is not known. We here investigate the respiratory function of inhibitory 5-HT1A receptors within the pTRG of the isolated brainstem of the adult lamprey. The 5-HT1A receptor agonists either bath applied or microinjected into the pTRG did not cause significant effects. However, the selective 5-HT1A receptor antagonist (S)-WAY 100135 bath applied or microinjected into the pTRG induced depressing respiratory effects or even apnoea, thus revealing that 5-HT exerts a 5-HT1A receptor-mediated potent tonic influence on respiration and contributes to maintain baseline levels of respiratory activity. Microinjections of strychnine or bicuculline, either alone or in combination, into the pTRG prevented (S)-WAY 100135-induced apnoea. In addition, immunohistochemical studies corroborate the present findings suggesting that 5-HT1A receptors are widely expressed in close apposition to the soma of glycine-immunoreactive cells located within the pTRG region. The results show that in the lamprey respiratory network, 5-HT exerts a tonic influence on respiration by a potent inhibitory control on both GABAergic and glycinergic mechanisms. The observed disinhibitory effects resemble the excitatory respiratory modulation exerted by 5-HT1A receptor-mediated inhibition of glycinergic and/or GABAergic neurons present in mammals, supporting the notion that some features of the neuronal network subserving respiratory rhythm generation are highly conserved throughout phylogeny.
Collapse
Affiliation(s)
- Elenia Cinelli
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università degli Studi di Firenze, Firenze, Italy
| | - Donatella Mutolo
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università degli Studi di Firenze, Firenze, Italy
| | - Ludovica Iovino
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università degli Studi di Firenze, Firenze, Italy
| | - Tito Pantaleo
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università degli Studi di Firenze, Firenze, Italy
| | - Fulvia Bongianni
- Dipartimento di Medicina Sperimentale e Clinica, Sezione Scienze Fisiologiche, Università degli Studi di Firenze, Firenze, Italy
| |
Collapse
|
32
|
Charli JL, Rodríguez-Rodríguez A, Hernández-Ortega K, Cote-Vélez A, Uribe RM, Jaimes-Hoy L, Joseph-Bravo P. The Thyrotropin-Releasing Hormone-Degrading Ectoenzyme, a Therapeutic Target? Front Pharmacol 2020; 11:640. [PMID: 32457627 PMCID: PMC7225337 DOI: 10.3389/fphar.2020.00640] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
Thyrotropin releasing hormone (TRH: Glp-His-Pro-NH2) is a peptide mainly produced by brain neurons. In mammals, hypophysiotropic TRH neurons of the paraventricular nucleus of the hypothalamus integrate metabolic information and drive the secretion of thyrotropin from the anterior pituitary, and thus the activity of the thyroid axis. Other hypothalamic or extrahypothalamic TRH neurons have less understood functions although pharmacological studies have shown that TRH has multiple central effects, such as promoting arousal, anorexia and anxiolysis, as well as controlling gastric, cardiac and respiratory autonomic functions. Two G-protein-coupled TRH receptors (TRH-R1 and TRH-R2) transduce TRH effects in some mammals although humans lack TRH-R2. TRH effects are of short duration, in part because the peptide is hydrolyzed in blood and extracellular space by a M1 family metallopeptidase, the TRH-degrading ectoenzyme (TRH-DE), also called pyroglutamyl peptidase II. TRH-DE is enriched in various brain regions but is also expressed in peripheral tissues including the anterior pituitary and the liver, which secretes a soluble form into blood. Among the M1 metallopeptidases, TRH-DE is the only member with a very narrow specificity; its best characterized biological substrate is TRH, making it a target for the specific manipulation of TRH activity. Two other substrates of TRH-DE, Glp-Phe-Pro-NH2 and Glp-Tyr-Pro-NH2, are also present in many tissues. Analogs of TRH resistant to hydrolysis by TRH-DE have prolonged central efficiency. Structure-activity studies allowed the identification of residues critical for activity and specificity. Research with specific inhibitors has confirmed that TRH-DE controls TRH actions. TRH-DE expression by β2-tanycytes of the median eminence of the hypothalamus allows the control of TRH flux into the hypothalamus-pituitary portal vessels and may regulate serum thyrotropin secretion. In this review we describe the critical evidences that suggest that modification of TRH-DE activity in tanycytes, and/or in other brain regions, may generate beneficial consequences in some central and metabolic disorders and identify potential drawbacks and missing information needed to test these hypotheses.
Collapse
Affiliation(s)
- Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| | | | | | | | | | | | | |
Collapse
|
33
|
Erickson JT. Central serotonin and autoresuscitation capability in mammalian neonates. Exp Neurol 2020; 326:113162. [DOI: 10.1016/j.expneurol.2019.113162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/04/2019] [Accepted: 12/23/2019] [Indexed: 01/08/2023]
|
34
|
5-HT neurons and central CO2 chemoreception. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/b978-0-444-64125-0.00021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
35
|
Cummings KJ, Hodges MR. The serotonergic system and the control of breathing during development. Respir Physiol Neurobiol 2019; 270:103255. [PMID: 31362064 DOI: 10.1016/j.resp.2019.103255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/28/2019] [Accepted: 07/12/2019] [Indexed: 01/26/2023]
Abstract
Serotonin (5-hydroxytryptamine 5-HT) was first discovered in the late 1940's as an endogenous bioactive amine capable of inducing vasoconstriction, and in the mid-1950's was found in the brain. It was in these early years that some of the first demonstrations were made regarding a role for brain 5-HT in neurological function and behavior, including data implicating reduced brain levels of 5-HT in clinical depression. Since that time, advances in molecular biology and physiological approaches in basic science research have intensely focused on 5-HT in the brain, and the many facets of its role during embryonic development, post-natal maturation, and neural function in adulthood continues to be established. This review focuses on what is known about the developmental roles for the 5-HT system, which we define as the neurons producing 5-HT along with pre-and post-synaptic receptors, in a vital homeostatic motor behavior - the control of breathing. We will cover what is known about the embryonic origins and fate specification of 5-HT neurons, and how the 5-HT system influences pre- and post-natal maturation of the ventilatory control system. In addition, we will focus on the role of the 5-HT system in specific respiratory behaviors during fetal, neonatal and postnatal development, and the relevance of dysfunction in this system in respiratory-related human pathologies including Sudden Infant Death Syndrome (SIDS).
Collapse
Affiliation(s)
- Kevin J Cummings
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA.
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
36
|
Mu L, Xia DD, Michalkiewicz T, Hodges M, Mouradian G, Konduri GG, Wong-Riley MTT. Effects of neonatal hyperoxia on the critical period of postnatal development of neurochemical expressions in brain stem respiratory-related nuclei in the rat. Physiol Rep 2019. [PMID: 29516654 PMCID: PMC5842315 DOI: 10.14814/phy2.13627] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have identified a critical period of respiratory development in rats at postnatal days P12‐13, when inhibitory influence dominates and when the response to hypoxia is at its weakest. This critical period has significant implications for Sudden Infant Death Syndrome (SIDS), the cause of which remains elusive. One of the known risk factors for SIDS is prematurity. A common intervention used in premature infants is hyperoxic therapy, which, if prolonged, can alter the ventilatory response to hypoxia and induce sustained inhibition of lung alveolar growth and pulmonary remodeling. The goal of this study was to test our hypothesis that neonatal hyperoxia from postnatal day (P) 0 to P10 in rat pups perturbs the critical period by altering the normal progression of neurochemical development in brain stem respiratory‐related nuclei. An in‐depth, semiquantitative immunohistochemical study was undertaken at P10 (immediately after hyperoxia and before the critical period), P12 (during the critical period), P14 (immediately after the critical period), and P17 (a week after the cessation of hyperoxia). In agreement with our previous findings, levels of cytochrome oxidase, brain‐derived neurotrophic factor (BDNF), TrkB (BDNF receptor), and several serotonergic proteins (5‐HT1A and 2A receptors, 5‐HT synthesizing enzyme tryptophan hydroxylase [TPH], and serotonin transporter [SERT]) all fell in several brain stem respiratory‐related nuclei during the critical period (P12) in control animals. However, in hyperoxic animals, these neurochemicals exhibited a significant fall at P14 instead. Thus, neonatal hyperoxia delayed but did not eliminate the critical period of postnatal development in multiple brain stem respiratory‐related nuclei, with little effect on the nonrespiratory cuneate nucleus.
Collapse
Affiliation(s)
- Lianwei Mu
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Dong Dong Xia
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Matthew Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gary Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Girija G Konduri
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Margaret T T Wong-Riley
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
37
|
Iovino L, Mutolo D, Cinelli E, Contini M, Pantaleo T, Bongianni F. Breathing stimulation mediated by 5-HT1A and 5-HT3 receptors within the preBötzinger complex of the adult rabbit. Brain Res 2019; 1704:26-39. [DOI: 10.1016/j.brainres.2018.09.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/10/2018] [Accepted: 09/18/2018] [Indexed: 02/06/2023]
|
38
|
Forster HV. Julius H. Comroe Distinguished Lecture: Interdependence of neuromodulators in the control of breathing. J Appl Physiol (1985) 2018; 125:1511-1525. [PMID: 30138081 DOI: 10.1152/japplphysiol.00477.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In vitro and in vivo anesthetized studies led to the conclusion that "deficiencies in one neuromodulator are immediately compensated by the action of other neuromodulators," which suggests an interdependence among neuromodulators. This concept was the focus of the 2018 Julius H. Comroe Lecture to the American Physiological Society in which I summarized our published studies testing the hypothesis that if modulatory interdependence was robust, breathing would not decrease during dialysis of antagonists to G protein-coupled excitatory receptors or agonists to inhibitory receptors into the ventral respiratory column (VRC) or the hypoglossal motor nuclei (HMN). We found breathing was not decreased during unilateral VRC dialyses of antagonists to excitatory muscarinic, serotonergic, and neurokinin-1 receptors alone or in combinations nor was breathing decreased with unilateral VRC dialysis of a µ-opioid receptor agonist. Analyses of the effluent dialysate revealed locally increased serotonin (excitatory) during muscarinic receptor blockade and decreased γ-aminobutyric acid (inhibitory) during dialysis of opioid agonists, suggesting an interdependence of neuromodulators through release of compensatory neuromodulators. Bilateral dialysis of receptor antagonists or agonist in the VRC increased breathing, which does not support the concept that unchanged breathing with unilateral dialyses was due to contralateral compensation. In contrast, in the HMN neither unilateral nor bilateral dialysis of the excitatory receptor antagonists altered breathing, but unilateral dialysis of the opioid receptor agonist decreased breathing. We conclude: 1) there is site-dependent interdependence of neuromodulators during physiologic conditions, and 2) attributing physiologic effects to a specific receptor perturbation is complicated by local compensatory mechanisms.
Collapse
Affiliation(s)
- Hubert V Forster
- Department of Physiology, Medical College of Wisconsin, Neuroscience Research Center, Medical College of Wisconsin, Zablocki Veterans Affairs Medical Center , Milwaukee, Wisconsin
| |
Collapse
|
39
|
Bright FM, Vink R, Byard RW. The potential role of substance P in brainstem homeostatic control in the pathogenesis of sudden infant death syndrome (SIDS). Neuropeptides 2018; 70:1-8. [PMID: 29908886 DOI: 10.1016/j.npep.2018.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/25/2018] [Accepted: 02/25/2018] [Indexed: 12/30/2022]
Abstract
Victims of sudden infant death syndrome (SIDS) are believed to have an underlying dysfunction in medullary homeostatic control that impairs critical responses to life threatening challenges such as hypoxia, hypercarbia and asphyxia, often during a sleep period. This failure is thought to result from abnormalities in a network of neural pathways in the medulla oblongata that control respiration, chemosensitivity, autonomic function and arousal. Studies have mainly focused on the role of serotonin, 5-hydroxytyptamine (5HT), although the neuropeptide substance P (SP) has also been shown to play an integral role in the modulation of medullary homeostatic function, often in conjunction with 5-HT. Actions of SP include regulation of respiratory rhythm generation, integration of cardiovascular control, modulation of the baroreceptor reflex and mediation of the chemoreceptor reflex in response to hypoxia. Abnormalities in SP neurotransmission may, therefore, also play a significant role in homeostatic dysfunction of the neurotransmitter network in SIDS. This review focuses on the pathways within the medulla involving SP and its tachykinin NK1 receptor, their potential relationship with the medullary 5-HT system, and possible involvement in the pathogenesis of SIDS.
Collapse
Affiliation(s)
- Fiona M Bright
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, SA, Australia.
| | - Robert Vink
- Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Roger W Byard
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, SA, Australia
| |
Collapse
|
40
|
Ikoma Y, Kusumoto-Yoshida I, Yamanaka A, Ootsuka Y, Kuwaki T. Inactivation of Serotonergic Neurons in the Rostral Medullary Raphé Attenuates Stress-Induced Tachypnea and Tachycardia in Mice. Front Physiol 2018; 9:832. [PMID: 30050449 PMCID: PMC6050454 DOI: 10.3389/fphys.2018.00832] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/13/2018] [Indexed: 11/13/2022] Open
Abstract
The medullary raphé nuclei are involved in controlling cardiovascular, respiratory, and thermoregulatory functions, as well as mediating stress-induced tachycardia and hyperthermia. Although the serotonergic system of the medullary raphé has been suggested as the responsible entity, specific evidence has been insufficient. In the present study, we tested this possibility by utilizing an optogenetic approach. We used genetically modified mice [tryptophan hydroxylase 2 (Tph2); archaerhodopsin-T (ArchT) mice] in which ArchT, a green light-driven neuronal silencer, was selectively expressed in serotonergic neurons under the regulation of Tph2 promoters. We first confirmed that an intruder stress selectively activated medullary, but not dorsal or median raphé serotonergic neurons. This activation was suppressed by photo-illumination via a pre-implanted optical fiber, as evidenced by the decrease of a cellular activation marker protein in the neurons. Next, we measured electro cardiogram (ECG), respiration, body temperature (BT), and locomotor activity in freely moving mice during intruder and cage-drop stress tests, with and without photo-illumination. In the intruder test, photo inactivation of the medullary serotonergic neurons significantly attenuated tachycardia (362 ± 58 vs. 564 ± 65 bpm.min, n = 19, p = 0.002) and tachypnea (94 ± 82 vs. 361 ± 138 cpm.min, n = 9, p = 0.026), but not hyperthermia (1.0 ± 0.1 vs. 1.0 ± 0.1°C.min, n = 19, p = 0.926) or hyperlocomotion (17 ± 4 vs. 22 ± 4, arbitrary, n = 19, p = 0.089). Similar results were obtained from cage-drop stress testing. Finally, photo-illumination did not affect the basal parameters of the resting condition. We conclude that a subpopulation of serotonergic neurons in the medullary raphé specifically mediate stress-induced tachypnea and tachycardia, which have little involvement in the basal determination of respiratory frequency (Res) and heart rate (HR), specifically mediate stress-induced tachycardia and tachypnea.
Collapse
Affiliation(s)
- Yoko Ikoma
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ikue Kusumoto-Yoshida
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Youichirou Ootsuka
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.,Centre for Neuroscience, Discipline of Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Tomoyuki Kuwaki
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
41
|
Bright FM, Byard RW, Vink R, Paterson DS. Normative distribution of substance P and its tachykinin neurokinin-1 receptor in the medullary serotonergic network of the human infant during postnatal development. Brain Res Bull 2018; 137:319-328. [PMID: 29331576 DOI: 10.1016/j.brainresbull.2018.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/09/2018] [Indexed: 10/18/2022]
Abstract
Substance P (SP) and its tachykinin NK1 receptor (NK1R) function within key medullary nuclei to regulate cardiorespiratory and autonomic control. We examined the normative distribution of SP and NK1R in the serotonergic (5-Hydroxytryptamine, [5-HT]) network of the human infant medulla during postnatal development, to provide a baseline to facilitate future analysis of the SP/NK1R system and its interaction with 5-HT within pediatric brainstem disorders in early life. [125I] labelled Bolton Hunter SP (BH-SP) tissue receptor autoradiography (n = 15), single label immunohistochemistry (IHC) and double label immunofluorescence (IF) (n = 10) were used to characterize the normative distribution profile of SP and NK1R in the 5-HT network of the human infant medulla during postnatal development. Tissue receptor autoradiography revealed extensive distribution of SP and NK1R in nuclei intimately related to cardiorespiratory function and autonomic control, with significant co-distribution and co-localization with 5-HT in the medullary network in the normal human infant during development. A trend for NK1R binding to decrease with age was observed with significantly higher binding in premature and male infants. We provide further evidence to suggest a significant role for SP/NK1R in the early postnatal period in the modulation of medullary cardiorespiratory and autonomic control in conjunction with medullary 5-HT mediated pathways and provide a baseline for future analysis of the potential consequences of abnormalities in these brainstem neurotransmitter networks during development.
Collapse
Affiliation(s)
- Fiona M Bright
- Harvard University Medical School, Boston, MA, USA; School of Medicine, University of Adelaide SA, Australia; Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Roger W Byard
- School of Medicine, University of Adelaide SA, Australia
| | - Robert Vink
- Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - David S Paterson
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Matarazzo V, Caccialupi L, Schaller F, Shvarev Y, Kourdougli N, Bertoni A, Menuet C, Voituron N, Deneris E, Gaspar P, Bezin L, Durbec P, Hilaire G, Muscatelli F. Necdin shapes serotonergic development and SERT activity modulating breathing in a mouse model for Prader-Willi syndrome. eLife 2017; 6:32640. [PMID: 29087295 PMCID: PMC5711373 DOI: 10.7554/elife.32640] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/29/2017] [Indexed: 12/31/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a genetic neurodevelopmental disorder that presents with hypotonia and respiratory distress in neonates. The Necdin-deficient mouse is the only model that reproduces the respiratory phenotype of PWS (central apnea and blunted response to respiratory challenges). Here, we report that Necdin deletion disturbs the migration of serotonin (5-HT) neuronal precursors, leading to altered global serotonergic neuroarchitecture and increased spontaneous firing of 5-HT neurons. We show an increased expression and activity of 5-HT Transporter (SERT/Slc6a4) in 5-HT neurons leading to an increase of 5-HT uptake. In Necdin-KO pups, the genetic deletion of Slc6a4 or treatment with Fluoxetine, a 5-HT reuptake inhibitor, restored normal breathing. Unexpectedly, Fluoxetine administration was associated with respiratory side effects in wild-type animals. Overall, our results demonstrate that an increase of SERT activity is sufficient to cause the apneas in Necdin-KO pups, and that fluoxetine may offer therapeutic benefits to PWS patients with respiratory complications. Prader-Willi syndrome results from the disruption of a cluster of neighboring genes, including one called Necdin. Symptoms begin in early infancy and worsen with age. Affected children tend to develop an insatiable appetite, which often leads to obesity. They also experience serious problems with their breathing. Chest infections, high altitude and intense physical activity can be dangerous for children with Prader-Willi syndrome. This is because a slight shortage of oxygen may trigger breathing difficulties that could prove fatal. The brain cells that produce a chemical messenger called serotonin help to control breathing. Several lines of evidence suggest that loss of Necdin may trigger breathing difficulties in Prader-Willi syndrome via effects on the serotonin system. First, serotonin neurons produce the Necdin protein. Second, laboratory mice that lack the gene for Necdin have abnormally shaped serotonin neurons. Third, these mice show breathing difficulties like those of individuals with Prader-Willi syndrome. But while this implies a connection between serotonin, Necdin and breathing difficulties, it falls short of establishing a causal link. Matarazzo et al. now reveal an increase in the quantity and activity of a protein called the serotonin transporter in mutant mice that lacked the gene for Necdin compared to normal mice. Serotonin transporter proteins mop up the serotonin that neurons release when they signal to one another. Neurons in the mutant mice take up more serotonin than their counterparts in normal mice; this means they have less serotonin available for signaling. This may make it harder for the mutant mice to regulate their breathing. Drugs called selective serotonin-reuptake inhibitors (or SSRIs for short) can block the serotonin transporter. These drugs, which include Fluoxetine (also called Prozac), are antidepressants. Matarazzo et al. show that SSRIs temporarily restore normal breathing in young mice that lack the gene for Necdin. However, these drugs have harmful long-term effects on breathing in non-mutant mice. Further studies should test whether short-term use of SSRIs could offer immediate relief for breathing difficulties in infants and children with Prader-Willi syndrome.
Collapse
Affiliation(s)
| | | | | | - Yuri Shvarev
- Department of Women's and Children's Health, Karolinska Institute, Solna, Sweden
| | | | | | | | | | - Evan Deneris
- Department of Neurosciences, Case Western Reserve University, Cleveland, United States
| | - Patricia Gaspar
- UPMC Univ Paris 6, Institut du Fer à Moulin, INSERM, Paris, France
| | - Laurent Bezin
- Lyon Neuroscience Research Center, Université de Lyon, INSERM, CNRS, Lyon, France
| | | | | | | |
Collapse
|
43
|
Zhang H, Zhao H, Feng HJ. Atomoxetine, a norepinephrine reuptake inhibitor, reduces seizure-induced respiratory arrest. Epilepsy Behav 2017; 73:6-9. [PMID: 28605634 PMCID: PMC5545072 DOI: 10.1016/j.yebeh.2017.04.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 04/26/2017] [Accepted: 04/26/2017] [Indexed: 12/31/2022]
Abstract
Sudden unexpected death in epilepsy (SUDEP) is a devastating epilepsy complication, and no effective preventive strategies are currently available for this fatal disorder. Clinical and animal studies of SUDEP demonstrate that seizure-induced respiratory arrest (S-IRA) is the primary event leading to death after generalized seizures in many cases. Enhancing brain levels of serotonin reduces S-IRA in animal models relevant to SUDEP, including the DBA/1 mouse. Given that serotonin in the brain plays an important role in modulating respiration and arousal, these findings suggest that deficits in respiration and/or arousal may contribute to S-IRA. It is well known that norepinephrine is an important neurotransmitter that modulates respiration and arousal in the brain as well. Therefore, we hypothesized that enhancing noradrenergic neurotransmission suppresses S-IRA. To test this hypothesis, we examined the effect of atomoxetine, a norepinephrine reuptake inhibitor (NRI), on S-IRA evoked by either acoustic stimulation or pentylenetetrazole in DBA/1 mice. We report the original observation that atomoxetine specifically suppresses S-IRA without altering the susceptibility to seizures evoked by acoustic stimulation, and atomoxetine also reduces S-IRA evoked by pentylenetetrazole in DBA/1 mice. Our data suggest that the noradrenergic signaling is importantly involved in S-IRA, and that atomoxetine, a medication widely used to treat attention deficit hyperactivity disorder (ADHD), is potentially useful to prevent SUDEP.
Collapse
Affiliation(s)
- Honghai Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA,Department of Anesthesia, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, China
| | - Haiting Zhao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA,Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hua-Jun Feng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Feng HJ, Faingold CL. Abnormalities of serotonergic neurotransmission in animal models of SUDEP. Epilepsy Behav 2017; 71:174-180. [PMID: 26272185 PMCID: PMC4749463 DOI: 10.1016/j.yebeh.2015.06.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/05/2015] [Accepted: 06/06/2015] [Indexed: 12/25/2022]
Abstract
Sudden unexpected death in epilepsy (SUDEP) is a devastating event, and both DBA/1 and DBA/2 mice have been shown to be relevant animal models for studying SUDEP. DBA mice exhibit seizure-induced respiratory arrest (S-IRA), leading to cardiac arrest and subsequent sudden death after generalized audiogenic seizures (AGSs). This sequence of terminal events is also observed in the majority of witnessed human SUDEP cases. Several pathophysiological mechanisms, including respiratory/cardiac dysfunction, have been proposed to contribute to human SUDEP. Several (but not all) selective serotonin (5-HT) reuptake inhibitors (SSRIs), including fluoxetine, can reversibly block S-IRA, and abnormal expression of 5-HT receptors is found in the brainstem of DBA mice. DBA mice, which do not initially show S-IRA, exhibit S-IRA after treatment with a nonselective 5-HT antagonist. These studies suggest that abnormalities of 5-HT neurotransmission are involved in the pathogenesis of S-IRA in DBA mice. Serotonergic (5-HT) transmission plays an important role in normal respiration, and DBA mice exhibiting S-IRA can be resuscitated using a rodent ventilator. It is important and interesting to know if fluoxetine blocks S-IRA in DBA mice by enhancing respiratory ventilation. To test this, the effects of breathing stimulants, doxapram, and 5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine (PK-THPP) were compared with the effects of fluoxetine on S-IRA in DBA/1 mice. Although fluoxetine reduces the incidence of S-IRA in DBA/1 mice, as reported previously, the same dose of fluoxetine fails to enhance baseline respiratory ventilation in the absence of AGSs. Doxapram and PK-THPP augment the baseline ventilation in DBA/1 mice. However, these breathing stimulants are ineffective in preventing S-IRA in DBA/1 mice. These data suggest that fluoxetine blocks S-IRA in DBA/1 mice by cellular/molecular mechanisms other than enhancement of basal ventilation. Future research directions are also discussed. This article is part of a Special Issue entitled "Genetic and Reflex Epilepsies, Audiogenic Seizures and Strains: From Experimental Models to the Clinic".
Collapse
Affiliation(s)
- Hua-Jun Feng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Carl L. Faingold
- Department of Pharmacology, Southern Illinois University School of Medicine, P.O. Box 19629, Springfield, Illinois 62794, U.S.A
| |
Collapse
|
45
|
Andrzejewski K, Kaczyńska K, Zaremba M. Serotonergic system in hypoxic ventilatory response in unilateral rat model of Parkinson's disease. J Biomed Sci 2017; 24:24. [PMID: 28347345 PMCID: PMC5368902 DOI: 10.1186/s12929-017-0331-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/21/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Malfunctioning of the serotonergic system in Parkinson's disease may contribute to non-motor symptoms such as respiratory complications. Thus the aim of our study was to investigate the role of serotonin 5-HT2 receptors in the modulation of normoxic breathing and the hypoxic ventilatory response (HVR) in rat model of Parkinson's disease. METHODS Wistar rats were lesioned unilaterally with double 6-hydroxydopamine (6-OHDA) injection to the right medial forebrain bundle (MFB). Before lesion and two weeks later animals were put in whole body plethysmography chamber and exposed to hypoxia (8% O2). Before hypoxic tests animals received intraperitoneal injections of DOI and ketanserin. Efficacy of lesion was confirmed by cylinder test, assessing limb use asymmetry. RESULTS Degeneration of the nigrostriatal pathway augmented response of tidal volume and minute ventilation to hypoxia. DOI administration in control and lesion state caused a significant rise in normoxic respiratory rate and minute ventilation. Yet, ventilatory response of these parameters to hypoxia was attenuated. Post-DOI magnitude of HVR in lesioned state was decreased in compare to pre-lesion control. Subsequent ketanserin injection reverted DOI-induced respiratory effects. We demonstrated that 6-OHDA treatment decreased the content of serotonin in the injured striatum and on both sides of the brainstem, leaving the concentration of noradrenaline on unchanged level. CONCLUSIONS These observations showed that damage of the nigrostriatal system initiates changes in the serotonergic system, confirmed by reduced concentration of serotonin in the striatum and brainstem, which affects the magnitude of respiratory response to hypoxia after activation of 5-HT2 receptors.
Collapse
Affiliation(s)
- Kryspin Andrzejewski
- Laboratory of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Katarzyna Kaczyńska
- Laboratory of Respiration Physiology, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland
| | - Małgorzata Zaremba
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research (CePT), Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
46
|
Madsen PM, Sloley SS, Vitores AA, Carballosa-Gautam MM, Brambilla R, Hentall ID. Prolonged stimulation of a brainstem raphe region attenuates experimental autoimmune encephalomyelitis. Neuroscience 2017; 346:395-402. [PMID: 28147248 PMCID: PMC5337132 DOI: 10.1016/j.neuroscience.2017.01.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS), a neuroinflammatory disease, has few treatment options, none entirely adequate. We studied whether prolonged electrical microstimulation of a hindbrain region (the nucleus raphe magnus) can attenuate experimental autoimmune encephalomyelitis, a murine model of MS induced by MOG35-55 injection. Eight days after symptoms emerged, a wireless electrical stimulator with an attached microelectrode was implanted cranially, and daily intermittent stimulation was begun in awake, unrestrained mice. The thoracic spinal cord was analyzed for changes in histology (on day 29) and gene expression (on day 37), with a focus on myelination and cytokine production. Controls, with inactive implants, showed a phase of disease exacerbation on days 19-25 that stimulation for >16days eliminated. Prolonged stimulation also reduced numbers of infiltrating immune cells and increased numbers of myelinated axons. It additionally lowered genetic expression of some pro-inflammatory cytokines (interferon gamma and tumor necrosis factor) and platelet-derived growth factor receptor alpha, a marker of oligodendrocyte precursors, while raising expression of myelin basic protein. Studies of restorative treatments for MS might profitably consider ways to stimulate the raphe magnus, directly or via its inputs, or to emulate its serotonergic and peptidergic output.
Collapse
Affiliation(s)
- Pernille M Madsen
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stephanie S Sloley
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Alberto A Vitores
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | | | - Roberta Brambilla
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.
| | - Ian D Hentall
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.
| |
Collapse
|
47
|
Activity of Tachykinin1-Expressing Pet1 Raphe Neurons Modulates the Respiratory Chemoreflex. J Neurosci 2017; 37:1807-1819. [PMID: 28073937 DOI: 10.1523/jneurosci.2316-16.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/30/2016] [Accepted: 12/30/2016] [Indexed: 11/21/2022] Open
Abstract
Homeostatic control of breathing, heart rate, and body temperature relies on circuits within the brainstem modulated by the neurotransmitter serotonin (5-HT). Mounting evidence points to specialized neuronal subtypes within the serotonergic neuronal system, borne out in functional studies, for the modulation of distinct facets of homeostasis. Such functional differences, read out at the organismal level, are likely subserved by differences among 5-HT neuron subtypes at the cellular and molecular levels, including differences in the capacity to coexpress other neurotransmitters such as glutamate, GABA, thyrotropin releasing hormone, and substance P encoded by the Tachykinin-1 (Tac1) gene. Here, we characterize in mice a 5-HT neuron subtype identified by expression of Tac1 and the serotonergic transcription factor gene Pet1, referred to as the Tac1-Pet1 neuron subtype. Transgenic cell labeling showed Tac1-Pet1 soma resident largely in the caudal medulla. Chemogenetic [clozapine-N-oxide (CNO)-hM4Di] perturbation of Tac1-Pet1 neuron activity blunted the ventilatory response of the respiratory CO2 chemoreflex, which normally augments ventilation in response to hypercapnic acidosis to restore normal pH and PCO2Tac1-Pet1 axonal boutons were found localized to brainstem areas implicated in respiratory modulation, with highest density in motor regions. These findings demonstrate that the activity of a Pet1 neuron subtype with the potential to release both 5-HT and substance P is necessary for normal respiratory dynamics, perhaps via motor outputs that engage muscles of respiration and maintain airway patency. These Tac1-Pet1 neurons may act downstream of Egr2-Pet1 serotonergic neurons, which were previously established in respiratory chemoreception, but do not innervate respiratory motor nuclei.SIGNIFICANCE STATEMENT Serotonin (5-HT) neurons modulate physiological processes and behaviors as diverse as body temperature, respiration, aggression, and mood. Using genetic tools, we characterize a 5-HT neuron subtype defined by expression of Tachykinin1 and Pet1 (Tac1-Pet1 neurons), mapping soma localization to the caudal medulla primarily and axonal projections to brainstem motor nuclei most prominently, and, when silenced, observed blunting of the ventilatory response to inhaled CO2Tac1-Pet1 neurons thus appear distinct from and contrast previously described Egr2-Pet1 neurons, which project primarily to chemosensory integration centers and are themselves chemosensitive.
Collapse
|
48
|
Neurodevelopmental Effects of Serotonin on the Brainstem Respiratory Network. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:193-216. [DOI: 10.1007/978-3-319-62817-2_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
49
|
Beltrán-Castillo S, Morgado-Valle C, Eugenín J. The Onset of the Fetal Respiratory Rhythm: An Emergent Property Triggered by Chemosensory Drive? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:163-192. [PMID: 29080027 DOI: 10.1007/978-3-319-62817-2_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The mechanisms responsible for the onset of respiratory activity during fetal life are unknown. The onset of respiratory rhythm may be a consequence of the genetic program of each of the constituents of the respiratory network, so they start to interact and generate respiratory cycles when reaching a certain degree of maturation. Alternatively, generation of cycles might require the contribution of recently formed sensory inputs that will trigger oscillatory activity in the nascent respiratory neural network. If this hypothesis is true, then sensory input to the respiratory generator must be already formed and become functional before the onset of fetal respiration. In this review, we evaluate the timing of the onset of the respiratory rhythm in comparison to the appearance of receptors, neurotransmitter machinery, and afferent projections provided by two central chemoreceptive nuclei, the raphe and locus coeruleus nuclei.
Collapse
Affiliation(s)
- Sebastián Beltrán-Castillo
- Laboratorio de Sistemas Neurales, Facultad de Química y Biología, Departamento de Biología, Universidad de Santiago de Chile, USACH, PO 9170022, Santiago, Chile
| | - Consuelo Morgado-Valle
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Campus Xalapa, Berlin 7, Fracc., Monte Magno Animas, C.P. 91190, Xalapa, Veracruz, Mexico.
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Facultad de Química y Biología, Departamento de Biología, Universidad de Santiago de Chile, USACH, PO 9170022, Santiago, Chile.
| |
Collapse
|
50
|
Faingold CL, Randall M, Zeng C, Peng S, Long X, Feng HJ. Serotonergic agents act on 5-HT 3 receptors in the brain to block seizure-induced respiratory arrest in the DBA/1 mouse model of SUDEP. Epilepsy Behav 2016; 64:166-170. [PMID: 27743549 PMCID: PMC5123739 DOI: 10.1016/j.yebeh.2016.09.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 11/18/2022]
Abstract
Drugs that enhance the action of serotonin (5-hydroxytrypamine, 5-HT), including several selective serotonin reuptake inhibitors (SSRIs), reduce susceptibility to seizure-induced respiratory arrest (S-IRA) that leads to death in the DBA/1 mouse model of sudden unexpected death in epilepsy (SUDEP). However, it is not clear if specific 5-HT receptors are important in the action of these drugs and whether the brain is the major site of action of these agents in this SUDEP model. The current study examined the actions of agents that affect the 5-HT3 receptor subtype on S-IRA and whether intracerebroventricular (ICV) microinjection of an SSRI would reduce S-IRA susceptibility in DBA/1 mice. The data indicate that systemic administration of SR 57227, a 5-HT3 agonist, was effective in blocking S-IRA in doses that did not block seizures, and the S-IRA blocking effect of the SSRI, fluoxetine, was abolished by coadministration of a 5-HT3 antagonist, ondansetron. Intracerebroventricular administration of fluoxetine in the present study was also able to block S-IRA without blocking seizures. These findings suggest that 5-HT3 receptors play an important role in the block of S-IRA by serotonergic agents, such as SSRIs, which is consistent with the abnormal expression of 5-HT3 receptors in the brainstem of DBA mice observed previously. Taken together, these data indicate that systemically administered serotonergic agents act, at least, in part, in the brain, to reduce S-IRA susceptibility in DBA/1 mice and that 5-HT3 receptors may be important to this effect.
Collapse
Affiliation(s)
- Carl L Faingold
- Department of Pharmacology and Neurology, Southern Illinois University, School of Medicine, Springfield, IL, USA.
| | - Marcus Randall
- Department of Pharmacology and Neurology, Southern Illinois University, School of Medicine, Springfield, IL, USA
| | - Chang Zeng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| | - Shifang Peng
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Hua-Jun Feng
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|