1
|
Onimaru H, Koyanagi Y, Iigaya K, Ikeda K, Izumizaki M. Intrinsic responses to hypoxia and hypercapnia of neurons in the cardiorespiratory center of the ventral medulla of newborn rats. Pflugers Arch 2025; 477:685-705. [PMID: 40119920 DOI: 10.1007/s00424-025-03077-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The rostral ventrolateral medulla (RVLM) includes a variety of neurons essential for cardiorespiratory control. Although some of these neurons are thought to be intrinsically sensitive to hypercapnia and/or hypoxia, relationships between types of neurons and responses to hypoxia and/or hypercapnia are not well understood. Tyrosine hydroxylase (TH) is one of the cell-type markers of the RVLM neurons. Here, we report effects of hypoxia and hypercapnia on TH-positive or -negative neurons in the RVLM of newborn rats. Brainstem-spinal cord preparations were isolated from 0-3-day-old Wistar rats and superfused with artificial cerebrospinal fluid equilibrated with 95% O2 and 5% CO2, pH 7.4 at 25-26 °C. Membrane potential responses to hypoxia (95% → 0% O2) and/or hypercapnia (2% → 8% CO2) were examined in the presence of tetrodotoxin (TTX) after identification of the firing pattern. We found that TH-positive C1 neurons in the RVLM were sensitive to hypoxia with membrane depolarization but less sensitive to hypercapnia. TH-negative neurons in the C1 area showed responses similar to those of C1 neurons. Moreover, C1 area neurons remained depolarized by hypoxia in the presence of TTX plus gliotransmitter blockers. In contrast, Phox2b-positive and TH-negative neurons in the parafacial respiratory group were intrinsically sensitive to CO2 but not sensitive to hypoxia. Respiratory-related neurons (Phox2b and TH negative) showed a variable response to hypoxia: unchanging, depolarizing, or hyperpolarizing. Our findings suggest that C1 area neurons in the RVLM are intrinsically sensitive to hypoxia and belong to one of the elements constituting central hypoxic sensors.
Collapse
Affiliation(s)
- Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan.
| | - Yui Koyanagi
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Kamon Iigaya
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Keiko Ikeda
- Department of Oral Physiology, Showa University School of Dentistry, Tokyo, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| |
Collapse
|
2
|
Amorim MR, Williams NR, Aung O, Alexis Ruiz M, Anokye-Danso F, de Deus JL, Xiong J, Dergacheva O, Bevans-Fonti S, Lee SM, Berger JS, Wu MN, Ahima RS, Mendelowitz D, Polotsky VY. Targeting melanocortin 4 receptor to treat sleep disordered breathing in mice. J Clin Invest 2025; 135:e177823. [PMID: 40232848 DOI: 10.1172/jci177823] [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: 04/17/2025] Open
Abstract
Weight loss medications are emerging candidates for pharmacotherapy of sleep disordered breathing (SDB). A melanocortin receptor 4 (MC4R) agonist, setmelanotide (SET), is used to treat obesity caused by abnormal melanocortin and leptin signaling. We hypothesized that SET can treat SDB in diet induced obese mice. We performed a proof-of-concept randomized crossover trial of a single dose of SET vs vehicle and a two-week daily SET vs vehicle trial, examined co-localization of Mc4r mRNAs with markers of CO2 sensing neurons Phox2b and neuromedin-B in the brainstem, and expressed Cre-dependent designer receptors exclusively activated by designer drugs or caspase in obese Mc4r-Cre mice. SET increased minute ventilation across sleep/wake states, enhanced the hypercapnic ventilatory response (HCVR) and abolished apneas during sleep. Phox2b+ neurons in the nucleus of the solitary tract (NTS) and the parafacial region expressed Mc4r. Chemogenetic stimulation of the MC4R+ neurons in the parafacial region, but not in the NTS, augmented HCVR without any changes in metabolism. Caspase elimination of the parafacial MC4R+ neurons abolished effects of SET on HCVR. Parafacial MC4R+ neurons projected to the respiratory pre-motor neurons retrogradely labeled from C3-C4. In conclusion, MC4R agonists enhance the HCVR and treat SDB by acting on the parafacial MC4R+ neurons.
Collapse
Affiliation(s)
- Mateus R Amorim
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, United States of America
| | - Noah R Williams
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, United States of America
| | - O Aung
- Department of Medicine, Medical College of Wisconsin, Milwaukee, United States of America
| | - Melanie Alexis Ruiz
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, United States of America
| | | | - Junia Lara de Deus
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, Washington, United States of America
| | - Jiali Xiong
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Washington, United States of America
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, United States of America
| | - Shannon Bevans-Fonti
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, United States of America
| | - Sean M Lee
- Office of Clinical Research, George Washington University, Washington, United States of America
| | - Jeffrey S Berger
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, United States of America
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Rexford S Ahima
- Department of Medicine, Johns Hopkins University, Baltimore, United States of America
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, United States of America
| | - Vsevolod Y Polotsky
- Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, United States of America
| |
Collapse
|
3
|
Moreira TS, Takakura AC, Falquetto B, Ramirez JM, Oliveira LM, Silva PE, Araujo EV. Neuroanatomical and neurochemical organization of brainstem and forebrain circuits involved in breathing regulation. J Neurophysiol 2025; 133:1116-1137. [PMID: 40059616 DOI: 10.1152/jn.00475.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/19/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Breathing regulation depends on a highly intricate and precise network within the brainstem, requiring the identification of all neuronal elements in the brainstem respiratory circuits and a comprehensive understanding of their organization into distinct functional compartments. These compartments play a pivotal role by providing essential input to three main targets: cranial motoneurons that regulate airway control, spinal motoneurons that activate the inspiratory and expiratory muscles, and higher brain structures that influence breathing behavior and integrate it with other physiological and behavioral processes. This review offers a comprehensive examination of the phenotypes, connections, and functional roles of the major compartments within the brainstem and forebrain respiratory circuits. In addition, it summarizes the diverse neurotransmitters used by neurons in these regions, highlighting their contributions to the coordination and modulation of respiratory activity.
Collapse
Affiliation(s)
- Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Barbara Falquetto
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, United States
- Department of Neurological Surgery, University of Washington, Seattle, Washington, United States
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, United States
| | - Phelipe E Silva
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Emmanuel V Araujo
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
4
|
Sato Y, Hayashi S, Oe S, Koike T, Nakano Y, Seki‐Omura R, Iwashita H, Hirahara Y, Kitada M. Chromosomal localization of PHOX2B during M-phase is disrupted in disease-associated mutants. Dev Growth Differ 2025; 67:136-148. [PMID: 39933489 PMCID: PMC11997733 DOI: 10.1111/dgd.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
In the M-phase, the nuclear membrane is broken down, nucleosomes are condensed as mitotic chromosomes, and transcription factors are generally known to be dislocated from their recognition sequences and dispersed to the cytoplasm. However, some transcription factors have recently been reported to remain on mitotic chromosomes and facilitate the rapid re-activation of the target genes in early G1-phase. Paired-like homeobox 2B (PHOX2B) is a transcription factor exhibiting chromosomal localization during M-phase. PHOX2B mutations are associated with congenital central hypoventilation syndrome, Hirschsprung disease, and neuroblastoma. In this study, we investigated PHOX2B chromosomal localization during M-phase through immunostaining and fluorescence recovery after photobleaching analysis to determine whether the chromosomal localization of disease-associated PHOX2B mutants is altered during M-phase. Missense mutations in the homeodomain and the frameshift mutation in the C-terminal domain disrupted the chromosomal localization of PHOX2B in M-phase, leading to its dispersion in the cell. Furthermore, a PHOX2B mutant with polyalanine expansion showed a line-shaped localization to the restricted region of mitotic chromosomes. Our findings suggest an association between the disease-associated mutations and defective chromosomal localization of transcription factors during M-phase. Further investigations of PHOX2B chromosomal localization during M-phase could reveal pathogenic mechanisms of such diseases.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Shinichi Hayashi
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Souichi Oe
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Taro Koike
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Yousuke Nakano
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Ryohei Seki‐Omura
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Hikaru Iwashita
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| | - Yukie Hirahara
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
- Faculty of Nursing, Kansai Medical UniversityOsakaJapan
| | - Masaaki Kitada
- Department of Anatomy, Faculty of MedicineKansai Medical UniversityOsakaJapan
| |
Collapse
|
5
|
Moreira TS, Burgraff NJ, Takakura AC, Oliveira LM, Araujo EV, Guan S, Ramirez JM. Functional Modulation of Retrotrapezoid Neurons Drives Fentanyl-Induced Respiratory Depression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635295. [PMID: 39975139 PMCID: PMC11838384 DOI: 10.1101/2025.01.28.635295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The primary cause of death from opioid overdose is opioid-induced respiratory depression (OIRD), characterized by severe suppression of respiratory rate, destabilized breathing patterns, hypercapnia, and heightened risk of apnea. The retrotrapezoid nucleus (RTN), a critical chemosensitive brainstem region in the rostral ventrolateral medullary reticular formation contains Phox2b + /Neuromedin-B ( Nmb ) propriobulbar neurons. These neurons, stimulated by CO 2 /H + , regulate breathing to prevent respiratory acidosis. Since the RTN shows limited expression of opioid-receptors, we expected that opioid-induced hypoventilation should activate these neurons to restore ventilation and stabilize arterial blood gases. However, the ability of the RTN to stimulate ventilation during OIRD has never been tested. We used optogenetic and pharmacogenetic approaches, to activate and inhibit RTN Phox2B + / Nmb + neurons before and after fentanyl administration. As expected, fentanyl (500 µg/kg, ip) suppressed respiratory rate and destabilized breathing. Before fentanyl, optogenetic stimulation of Phox2b + / Nmb + or chemogenetic inhibition of Nmb + cells increased and decreased breathing activity, respectively. Surprisingly, optogenetic stimulation after fentanyl administration caused a significantly greater increase in breathing activity compared to pre-fentanyl levels. By contrast chemogenetic ablation of RTN Nmb neurons caused profound hypoventilation and breathing instability after fentanyl. The results suggest that fentanyl does not inhibit the ability of Phox2b + / Nmb + cells within the RTN region to stimulate breathing. Thus, this study highlights the potential of stimulating RTN neurons as a therapeutic approach to restore respiratory function in cases of OIRD.
Collapse
|
6
|
Souza GMPR, Stornetta DS, Abbott SBG. Interactions between Arousal State and CO 2 Determine the Activity of Central Chemoreceptor Neurons That Drive Breathing. J Neurosci 2025; 45:e1587242024. [PMID: 39510833 DOI: 10.1523/jneurosci.1587-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/11/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
The homeostatic regulation of pulmonary ventilation, and ultimately arterial PCO2, depends on interactions between respiratory chemoreflexes and arousal state. The ventilatory response to CO2 is triggered by neurons in the retrotrapezoid nucleus (RTN) that function as sensors of central pH, which can be identified in adulthood by the expression of Phox2b and neuromedin B. Here, we examine the dynamic response of genetically defined RTN neurons to hypercapnia and arousal state in freely behaving adult male and female mice using the calcium indicator jGCaMP7 and fiber photometry. We found that hypercapnia vigorously activates RTN neurons with a low CO2 recruitment threshold and with response kinetics that match respiratory activity whereas hypoxia had little effect. RTN activity increased transiently during wakefulness and respiratory-related arousals and rose persistently during rapid eye movement sleep, and their CO2 response persisted under anesthesia. Complementary studies using inhibitory optogenetics show that RTN activity supports eupneic breathing under anesthesia as well as during states of high arousal, but their activity is redundant for voluntary breathing patterns. Collectively, this study demonstrates that CO2-activated RTN neurons are exquisitely sensitive to the arousal state, which determines their contribution to alveolar ventilation in relation to arterial PCO2.
Collapse
Affiliation(s)
- George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Daniel S Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| |
Collapse
|
7
|
Gasparini S, Almeida‐Pereira G, Munuzuri ASP, Resch JM, Geerling JC. Molecular Ontology of the Nucleus of Solitary Tract. J Comp Neurol 2024; 532:e70004. [PMID: 39629676 PMCID: PMC11615840 DOI: 10.1002/cne.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
The nucleus of the solitary tract (NTS) receives visceral information and regulates appetitive, digestive, and cardiorespiratory systems. Within the NTS, diverse processes operate in parallel to sustain life, but our understanding of their cellular composition is incomplete. Here, we integrate histologic and transcriptomic analysis to identify and compare molecular features that distinguish neurons in this brain region. Most glutamatergic neurons in the NTS and area postrema co-express the transcription factors Lmx1b and Phox2b, except for a ventral band of neurons in the far-caudal NTS, which include the Gcg-expressing neurons that produce glucagon-like peptide 1 (GLP-1). GABAergic interneurons intermingle through the Lmx1b+Phox2b macropopulation, and dense clusters of GABAergic neurons surround the NTS. The Lmx1b+Phox2b macropopulation includes subpopulations with distinct distributions expressing Grp, Hsd11b2, Npff, Pdyn, Pou3f1, Sctr, Th, and other markers. These findings highlight Lmx1b-Phox2b co-expression as a common feature of glutamatergic neurons in the NTS and improve our understanding of the organization and distribution of neurons in this critical brain region.
Collapse
Affiliation(s)
| | | | | | - Jon M. Resch
- Department of Neuroscience and PharmacologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| | - Joel C. Geerling
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
8
|
Moreira TS, Mulkey DK, Takakura AC. Update on vascular control of central chemoreceptors. Exp Physiol 2024; 109:1837-1843. [PMID: 38153366 PMCID: PMC11522829 DOI: 10.1113/ep091329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
At least four mechanisms have been proposed to elucidate how neurons in the retrotrapezoid (RTN) region sense changes in CO2/H+ to regulate breathing (i.e., function as respiratory chemosensors). These mechanisms include: (1) intrinsic neuronal sensitivity to H+ mediated by TASK-2 and GPR4; (2) paracrine activation of RTN neurons by CO2-responsive astrocytes (via a purinergic mechanism); (3) enhanced excitatory synaptic input or disinhibition; and (4) CO2-induced vascular contraction. Although blood flow can influence tissue CO2/H+ levels, there is limited understanding of how control of vascular tone in central CO2 chemosensitive regions might contribute to respiratory output. In this review, we focus on recent evidence that CO2/H+-induced purinergic-dependent vasoconstriction in the ventral parafacial region near RTN neurons supports respiratory chemoreception. This mechanism appears to be unique to the ventral parafacial region and opposite to other brain regions, including medullary chemosensor regions, where CO2/H+ elicits vasodilatation. We speculate that this mechanism helps to maintain CO2/H+ levels in the vicinity of RTN neurons, thereby maintaining the drive to breathe. Important next steps include determining whether disruption of CO2/H+ vascular reactivity contributes to or can be targeted to improve breathing problems in disease states, such as Parkinson's disease.
Collapse
Affiliation(s)
- Thiago S. Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias BiomedicasUniversidade de Sao PauloSao PauloBrazil
| | - Daniel K. Mulkey
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsConnecticutUSA
| | - Ana C. Takakura
- Department of Pharmacology, Instituto de Ciencias BiomedicasUniversidade de Sao PauloSão PauloBrazil
| |
Collapse
|
9
|
Naccarato MC, Oliveira LM, Ferreira CB, Moreira TS, Takakura AC. Nucleus of the solitary tract neuronal degeneration and impaired hypoxia response in a model of Parkinson's disease. Exp Neurol 2024; 380:114924. [PMID: 39147260 DOI: 10.1016/j.expneurol.2024.114924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Parkinson's disease (PD) involves the degeneration of dopaminergic neurons in the substantia nigra (SNpc) and manifests with both classic and non-classic motor symptoms, including respiratory failure. Our study aims to investigate the involvement of the commissural and intermediate nucleus of the solitary tract (cNTS and iNTS) in the attenuated respiratory response to hypoxia in PD. Using a PD rat model induced by bilateral injection of 6-hydroxydopamine (6-OHDA) into the striatum of male Wistar rats, we explored potential alterations in the population of Phox2b neurons or hypoxia-activated neurons in the NTS projecting to the retrotrapezoid nucleus (RTN). Additionally, we explored neuronal connectivity between SNpc and cNTS. Projections pathways were assessed using unilateral injection of the retrograde tracer Fluorogold (FG) in the cNTS and RTN. Neuronal activation was evaluated by analyzing fos expression in rats exposed to hypoxia. In the PD model, the ventilatory response, measured through whole-body plethysmography, was impaired at both baseline and in response to hypoxia. A reduction in Phox2b-expressing neurons or hypoxia-activated neurons projecting to the RTN was observed. Additionally, we identified an indirect pathway linking the SNpc and cNTS, which passes through the periaqueductal gray (PAG). In conclusion, our findings suggest impairment in the SNpc-PAG-cNTS pathway in the PD model, explaining the loss of Phox2b-expressing neurons or hypoxia-activated neurons in the cNTS and subsequent respiratory impairment during hypoxic stimulation. We propose that the reduced population of Phox2b-expressing neurons in the NTS may include the same neurons activated by hypoxia and projecting to the RTN.
Collapse
Affiliation(s)
- Monique C Naccarato
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil
| | - Luiz M Oliveira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil; Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Avenue, Seattle, WA 98101, USA
| | - Caroline B Ferreira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil; Department of Neurobiology, University of Pittsburgh School of Medicine, USA
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508 Sao Paulo, SP, Brazil.
| |
Collapse
|
10
|
Gonye EC, Shi Y, Li K, Clements RT, Xu W, Bayliss DA. Intrinsic Molecular Proton Sensitivity Underlies GPR4 Effects on Retrotrapezoid Nucleus Neuronal Activation and CO 2-Stimulated Breathing. J Neurosci 2024; 44:e0799242024. [PMID: 39107057 PMCID: PMC11376338 DOI: 10.1523/jneurosci.0799-24.2024] [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: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024] Open
Abstract
An interoceptive homeostatic reflex monitors levels of CO2/H+ to maintain blood gas homeostasis and rapidly regulate tissue acid-base balance by driving lung ventilation and CO2 excretion-this CO2-evoked increase in respiration is the hypercapnic ventilatory reflex (HCVR). Retrotrapezoid nucleus (RTN) neurons provide crucial excitatory drive to downstream respiratory rhythm/pattern-generating circuits, and their activity is directly modulated by changes in CO2/H+ RTN neurons express GPR4 and TASK-2, global deletion of which abrogates CO2/H+ activation of RTN neurons and the HCVR. It has not been determined if the intrinsic pH sensitivity of these proton detectors is required for these effects. We used CRISPR/Cas9 genome editing to generate mice with mutations in either of two pH-sensing histidine residues in GPR4 to determine effects on RTN neuronal CO2/H+ sensitivity and the HCVR. In global GPR4(H81F) and GPR4(H167F) mice, CO2-stimulated breathing and CO2-induced RTN neuronal activation were strongly blunted, with no effect on hypoxia-stimulated breathing. In brainstem slices from GPR4(H81F) mice, peak firing of RTN neurons during bath acidification was significantly reduced compared with GPR4 wild-type mice, and a subpopulation of RTN neurons was rendered pH-insensitive, phenocopying previous results from GPR4-deleted mice. These effects were independent of changes in RTN number/distribution, neuronal excitability or transcript levels for GPR4 and TASK-2. CO2-stimulated breathing was reduced to a similar extent in GPR4(H81F) and TASK-2-deleted mice, with combined mutation yielding no additional deficit in the HCVR. Together, these data demonstrate that the intrinsic pH sensitivity of GPR4 is necessary for full elaboration of the HCVR.
Collapse
Affiliation(s)
- Elizabeth C Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Rachel T Clements
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Wenhao Xu
- Genetically Engineered Mouse Model Core, University of Virginia, Charlottesville, Virginia 22903
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
11
|
Gu B. Linking Respiratory Challenges in KCNQ2 Encephalopathy to "Phox2b" Neurons in the Retrotrapezoid Nucleus. Epilepsy Curr 2024; 24:289-291. [PMID: 39309053 PMCID: PMC11412407 DOI: 10.1177/15357597241253680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 09/25/2024] Open
Abstract
Phox2b-Expressing Neurons Contribute to Breathing Problems in Kcnq2 Loss- and Gain-of-Function Encephalopathy Models Soto-Perez J, Cleary CM, Sobrinho CR, Mulkey SB, Carroll JL, Tzingounis AV, Mulkey DK. Nat Commun. 2023;14:8059. doi:10.1038/s41467-023-43834-7 Loss- and gain-of-function variants in the gene encoding KCNQ2 channels are a common cause of developmental and epileptic encephalopathy, a condition characterized by seizures, developmental delays, breathing problems, and early mortality. To understand how KCNQ2 dysfunction impacts behavior in a mouse model, we focus on the control of breathing by neurons expressing the transcription factor Phox2b which includes respiratory neurons in the ventral parafacial region. We find Phox2b-expressing ventral parafacial neurons express Kcnq2 in the absence of other Kcnq isoforms, thus clarifying why disruption of Kcnq2 but not other channel isoforms results in breathing problems. We also find that Kcnq2 deletion or expression of a recurrent gain-of-function variant R201C in Phox2b-expressing neurons increases baseline breathing or decreases the central chemoreflex, respectively, in mice during the light/inactive state. These results uncover mechanisms underlying breathing abnormalities in KCNQ2 encephalopathy and highlight an unappreciated vulnerability of Phox2b-expressing ventral parafacial neurons to KCNQ2 pathogenic variants.
Collapse
Affiliation(s)
- Bin Gu
- Department of Neuroscience, Ohio State University
| |
Collapse
|
12
|
Furdui A, da Silveira Scarpellini C, Montandon G. Anatomical distribution of µ-opioid receptors, neurokinin-1 receptors, and vesicular glutamate transporter 2 in the mouse brainstem respiratory network. J Neurophysiol 2024; 132:108-129. [PMID: 38748514 DOI: 10.1152/jn.00478.2023] [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: 12/21/2023] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 07/03/2024] Open
Abstract
µ-Opioid receptors (MORs) are responsible for mediating both the analgesic and respiratory effects of opioid drugs. By binding to MORs in brainstem regions involved in controlling breathing, opioids produce respiratory depressive effects characterized by slow and shallow breathing, with potential cardiorespiratory arrest and death during overdose. To better understand the mechanisms underlying opioid-induced respiratory depression, thorough knowledge of the regions and cellular subpopulations that may be vulnerable to modulation by opioid drugs is needed. Using in situ hybridization, we determined the distribution and coexpression of Oprm1 (gene encoding MORs) mRNA with glutamatergic (Vglut2) and neurokinin-1 receptor (Tacr1) mRNA in medullary and pontine regions involved in breathing control and modulation. We found that >50% of cells expressed Oprm1 mRNA in the preBötzinger complex (preBötC), nucleus tractus solitarius (NTS), nucleus ambiguus (NA), postinspiratory complex (PiCo), locus coeruleus (LC), Kölliker-Fuse nucleus (KF), and the lateral and medial parabrachial nuclei (LBPN and MPBN, respectively). Among Tacr1 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, Bötzinger complex (BötC), PiCo, LC, raphe magnus nucleus, KF, LPBN, and MPBN, whereas among Vglut2 mRNA-expressing cells, >50% coexpressed Oprm1 mRNA in the preBötC, NTS, NA, BötC, PiCo, LC, KF, LPBN, and MPBN. Taken together, our study provides a comprehensive map of the distribution and coexpression of Oprm1, Tacr1, and Vglut2 mRNA in brainstem regions that control and modulate breathing and identifies Tacr1 and Vglut2 mRNA-expressing cells as subpopulations with potential vulnerability to modulation by opioid drugs.NEW & NOTEWORTHY Opioid drugs can cause serious respiratory side-effects by binding to µ-opioid receptors (MORs) in brainstem regions that control breathing. To better understand the regions and their cellular subpopulations that may be vulnerable to modulation by opioids, we provide a comprehensive map of Oprm1 (gene encoding MORs) mRNA expression throughout brainstem regions that control and modulate breathing. Notably, we identify glutamatergic and neurokinin-1 receptor-expressing cells as potentially vulnerable to modulation by opioid drugs and worthy of further investigation using targeted approaches.
Collapse
Affiliation(s)
- Andreea Furdui
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Gaspard Montandon
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Jones AA, Marino GM, Arble DM. Time-restricted feeding reveals a role for neural respiratory clocks in optimizing daily ventilatory-metabolic coupling in mice. Am J Physiol Endocrinol Metab 2024; 327:E111-E120. [PMID: 38836780 PMCID: PMC11390118 DOI: 10.1152/ajpendo.00111.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
The master circadian clock, located in the suprachiasmatic nuclei (SCN), organizes the daily rhythm in minute ventilation (V̇e). However, the extent that the daily rhythm in V̇e is secondary to SCN-imposed O2 and CO2 cycles (i.e., metabolic rate) or driven by other clock mechanisms remains unknown. Here, we experimentally shifted metabolic rate using time-restricted feeding (without affecting light-induced synchronization of the SCN) to determine the influence of metabolic rate in orchestrating the daily V̇e rhythm. Mice eating predominantly at night exhibited robust daily rhythms in O2 consumption (V̇o2), CO2 production (V̇co2), and V̇e with similar peak times (approximately ZT18) that were consistent with SCN organization. However, feeding mice exclusively during the day separated the relative timing of metabolic and ventilatory rhythms, resulting in an approximately 8.5-h advance in V̇co2 and a disruption of the V̇e rhythm, suggesting opposing circadian and metabolic influences on V̇e. To determine if the molecular clock of cells involved in the neural control of breathing contributes to the daily V̇e rhythm, we examined V̇e in mice lacking BMAL1 in Phox2b-expressing respiratory cells (i.e., BKOP mice). The ventilatory and metabolic rhythms of predominantly night-fed BKOP mice did not differ from wild-type mice. However, in contrast to wild-type mice, exclusive day feeding of BKOP mice led to an unfettered daily V̇e rhythm with a peak time aligning closely with the daily V̇co2 rhythm. Taken together, these results indicate that both daily V̇co2 changes and intrinsic circadian time-keeping within Phox2b respiratory cells are predominant orchestrators of the daily rhythm in ventilation.NEW & NOTEWORTHY The master circadian clock organizes the daily rhythm in ventilation; however, the extent that this rhythm is driven by SCN regulation of metabolic rate versus other clock mechanisms remains unknown. We report that metabolic rate alone is insufficient to explain the daily oscillation in ventilation and that neural respiratory clocks within Phox2b-expressing cells additionally optimize breathing. Collectively, these findings advance our mechanistic understanding of the circadian rhythm in ventilatory control.
Collapse
Affiliation(s)
- Aaron A Jones
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Gabriella M Marino
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Deanna M Arble
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| |
Collapse
|
14
|
Diana D, Pirone L, Russo L, D'Abrosca G, Madheswaran M, Benfante R, Di Lascio S, Caldinelli L, Fornasari D, Acconcia C, Corvino A, Ventserova N, Pollegioni L, Isernia C, Di Gaetano S, Malgieri G, Pedone EM, Fattorusso R. Structural characterization of PHOX2B and its DNA interaction shed light on the molecular basis of the +7Ala variant pathogenicity in CCHS. Chem Sci 2024; 15:8858-8872. [PMID: 38873078 PMCID: PMC11168103 DOI: 10.1039/d3sc06427a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/12/2024] [Indexed: 06/15/2024] Open
Abstract
An expansion of poly-alanine up to +13 residues in the C-terminus of the transcription factor PHOX2B underlies the onset of congenital central hypoventilation syndrome (CCHS). Recent studies demonstrated that the alanine tract expansion influences PHOX2B folding and activity. Therefore, structural information on PHOX2B is an important target for obtaining clues to elucidate the insurgence of the alanine expansion-related syndrome and also for defining a viable therapy. Here we report by NMR spectroscopy the structural characterization of the homeodomain (HD) of PHOX2B and HD + C-terminus PHOX2B protein, free and in the presence of the target DNA. The obtained structural data are then exploited to obtain a structural model of the PHOX2B-DNA interaction. In addition, the variant +7Ala, responsible for one of the most frequent forms of the syndrome, was analysed, showing different conformational proprieties in solution and a strong propensity to aggregation. Our data suggest that the elongated poly-alanine tract would be related to disease onset through a loss-of-function mechanism. Overall, this study paves the way for the future rational design of therapeutic drugs, suggesting as a possible therapeutic route the use of specific anti-aggregating molecules capable of preventing variant aggregation and possibly restoring the DNA-binding activity of PHOX2B.
Collapse
Affiliation(s)
- Donatella Diana
- CNR - Institute of Biostructures and Bioimaging Via Pietro Castellino 111 80131 Naples Italy
| | - Luciano Pirone
- CNR - Institute of Biostructures and Bioimaging Via Pietro Castellino 111 80131 Naples Italy
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Gianluca D'Abrosca
- Department of Clinical and Experimental Medicine - University of Foggia Viale Luigi Pinto 71122 Foggia Italy
| | - Manoj Madheswaran
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Roberta Benfante
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano Milan Italy
- CNR - Institute of Neuroscience Vedano Al Lambro (MB) Italy
- NeuroMi - Milan Center for Neuroscience, University of Milano Bicocca Milan Italy
| | - Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano Milan Italy
| | - Laura Caldinelli
- Department of Biotechnology and Life Sciences, University of Insubria Via J.H. Dunant 3 21100 Varese Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano Milan Italy
| | - Clementina Acconcia
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Andrea Corvino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Nataliia Ventserova
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria Via J.H. Dunant 3 21100 Varese Italy
| | - Carla Isernia
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Sonia Di Gaetano
- CNR - Institute of Biostructures and Bioimaging Via Pietro Castellino 111 80131 Naples Italy
| | - Gaetano Malgieri
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| | - Emilia M Pedone
- CNR - Institute of Biostructures and Bioimaging Via Pietro Castellino 111 80131 Naples Italy
| | - Roberto Fattorusso
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies - University of Campania "Luigi Vanvitelli" Via Vivaldi 43 81100 Caserta Italy
| |
Collapse
|
15
|
Cardani S, Janes TA, Betzner W, Pagliardini S. Knockdown of PHOX2B in the retrotrapezoid nucleus reduces the central CO 2 chemoreflex in rats. eLife 2024; 13:RP94653. [PMID: 38727716 PMCID: PMC11087052 DOI: 10.7554/elife.94653] [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: 05/12/2024] Open
Abstract
PHOX2B is a transcription factor essential for the development of different classes of neurons in the central and peripheral nervous system. Heterozygous mutations in the PHOX2B coding region are responsible for the occurrence of Congenital Central Hypoventilation Syndrome (CCHS), a rare neurological disorder characterised by inadequate chemosensitivity and life-threatening sleep-related hypoventilation. Animal studies suggest that chemoreflex defects are caused in part by the improper development or function of PHOX2B expressing neurons in the retrotrapezoid nucleus (RTN), a central hub for CO2 chemosensitivity. Although the function of PHOX2B in rodents during development is well established, its role in the adult respiratory network remains unknown. In this study, we investigated whether reduction in PHOX2B expression in chemosensitive neuromedin-B (NMB) expressing neurons in the RTN altered respiratory function. Four weeks following local RTN injection of a lentiviral vector expressing the short hairpin RNA (shRNA) targeting Phox2b mRNA, a reduction of PHOX2B expression was observed in Nmb neurons compared to both naive rats and rats injected with the non-target shRNA. PHOX2B knockdown did not affect breathing in room air or under hypoxia, but ventilation was significantly impaired during hypercapnia. PHOX2B knockdown did not alter Nmb expression but it was associated with reduced expression of both Task2 and Gpr4, two CO2/pH sensors in the RTN. We conclude that PHOX2B in the adult brain has an important role in CO2 chemoreception and reduced PHOX2B expression in CCHS beyond the developmental period may contribute to the impaired central chemoreflex function.
Collapse
Affiliation(s)
- Silvia Cardani
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
- Women and Children’s Health Research Institute, University of AlbertaEdmontonCanada
| | - Tara A Janes
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
- Women and Children’s Health Research Institute, University of AlbertaEdmontonCanada
| | - William Betzner
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
| | - Silvia Pagliardini
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
- Women and Children’s Health Research Institute, University of AlbertaEdmontonCanada
- Neuroscience and Mental Health Institute, University of AlbertaEdmontonCanada
| |
Collapse
|
16
|
Janes TA, Cardani S, Saini JK, Pagliardini S. Etonogestrel promotes respiratory recovery in an in vivo rat model of central chemoreflex impairment. Acta Physiol (Oxf) 2024; 240:e14093. [PMID: 38258900 DOI: 10.1111/apha.14093] [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: 08/08/2023] [Revised: 10/11/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024]
Abstract
AIM The central CO2 chemoreflex is a vital component of respiratory control networks, providing excitatory drive during resting conditions and challenges to blood gas homeostasis. The retrotrapezoid nucleus is a crucial hub for CO2 chemosensitivity; its ablation or inhibition attenuates CO2 chemoreflexes and diminishes restful breathing. Similar phenotypes characterize certain hypoventilation syndromes, suggesting underlying retrotrapezoid nucleus impairment in these disorders. Progesterone stimulates restful breathing and CO2 chemoreflexes. However, its mechanisms and sites of actions remain unknown and the experimental use of synthetic progestins in patients and animal models have been met with mixed respiratory outcomes. METHODS We investigated whether acute or chronic administration of the progestinic drug, etonogestrel, could rescue respiratory chemoreflexes following selective lesion of the retrotrapezoid nucleus with saporin toxin. Adult female Sprague Dawley rats were grouped based on lesion size determined by the number of surviving chemosensitive neurons, and ventilatory responses were measured by whole body plethysmography. RESULTS Ventilatory responses to hypercapnia (but not hypoxia) were compromised in a lesion-dependent manner. Chronic etonogestrel treatment improved CO2 chemosensitivity selectively in rats with moderate lesion, suggesting that a residual number of chemosensitive neurons are required for etonogestrel-induced CO2 chemoreflex recovery. CONCLUSION This study provides new evidence for the use of progestins as respiratory stimulants under conditions of central hypoventilation and provides a new testable model for assessing the mechanism of action of progestins in the respiratory network.
Collapse
Affiliation(s)
- Tara A Janes
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Silvia Cardani
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jasmeen K Saini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Silvia Pagliardini
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Souza GMPR, Abbott SBG. Loss-of-function of chemoreceptor neurons in the retrotrapezoid nucleus: What have we learned from it? Respir Physiol Neurobiol 2024; 322:104217. [PMID: 38237884 PMCID: PMC10922619 DOI: 10.1016/j.resp.2024.104217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
Central respiratory chemoreceptors are cells in the brain that regulate breathing in relation to arterial pH and PCO2. Neurons located at the retrotrapezoid nucleus (RTN) have been hypothesized to be central chemoreceptors and/or to be part of the neural network that drives the central respiratory chemoreflex. The inhibition or ablation of RTN chemoreceptor neurons has offered important insights into the role of these cells on central respiratory chemoreception and the neural control of breathing over almost 60 years since the original identification of acid-sensitive properties of this ventral medullary site. Here, we discuss the current definition of chemoreceptor neurons in the RTN and describe how this definition has evolved over time. We then summarize the results of studies that use loss-of-function approaches to evaluate the effects of disrupting the function of RTN neurons on respiration. These studies offer evidence that RTN neurons are indispensable for the central respiratory chemoreflex in mammals and exert a tonic drive to breathe at rest. Moreover, RTN has an interdependent relationship with oxygen sensing mechanisms for the maintenance of the neural drive to breathe and blood gas homeostasis. Collectively, RTN neurons are a genetically-defined group of putative central respiratory chemoreceptors that generate CO2-dependent drive that supports eupneic breathing and stimulates the hypercapnic ventilatory reflex.
Collapse
|
18
|
Soto-Perez J, Cleary CM, Sobrinho CR, Mulkey SB, Carroll JL, Tzingounis AV, Mulkey DK. Phox2b-expressing neurons contribute to breathing problems in Kcnq2 loss- and gain-of-function encephalopathy models. Nat Commun 2023; 14:8059. [PMID: 38052789 PMCID: PMC10698053 DOI: 10.1038/s41467-023-43834-7] [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: 04/20/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023] Open
Abstract
Loss- and gain-of-function variants in the gene encoding KCNQ2 channels are a common cause of developmental and epileptic encephalopathy, a condition characterized by seizures, developmental delays, breathing problems, and early mortality. To understand how KCNQ2 dysfunction impacts behavior in a mouse model, we focus on the control of breathing by neurons expressing the transcription factor Phox2b which includes respiratory neurons in the ventral parafacial region. We find Phox2b-expressing ventral parafacial neurons express Kcnq2 in the absence of other Kcnq isoforms, thus clarifying why disruption of Kcnq2 but not other channel isoforms results in breathing problems. We also find that Kcnq2 deletion or expression of a recurrent gain-of-function variant R201C in Phox2b-expressing neurons increases baseline breathing or decreases the central chemoreflex, respectively, in mice during the light/inactive state. These results uncover mechanisms underlying breathing abnormalities in KCNQ2 encephalopathy and highlight an unappreciated vulnerability of Phox2b-expressing ventral parafacial neurons to KCNQ2 pathogenic variants.
Collapse
Affiliation(s)
- J Soto-Perez
- Dept of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - C M Cleary
- Dept of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - C R Sobrinho
- Dept of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - S B Mulkey
- Prenatal Pediatrics Institute, Children's National Hospital, Departments of Neurology and Pediatrics, The George Washington Univ. School of Medicine and Health Sciences, Washington, DC, USA
| | - J L Carroll
- Dept. of Pediatrics, Univ. Arkansas for Medical Sciences, Little Rock, AR, USA
| | - A V Tzingounis
- Dept of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.
| | - D K Mulkey
- Dept of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
19
|
Turk AZ, Millwater M, SheikhBahaei S. Whole-brain analysis of CO 2 chemosensitive regions and identification of the retrotrapezoid and medullary raphé nuclei in the common marmoset ( Callithrix jacchus). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.558361. [PMID: 37986845 PMCID: PMC10659419 DOI: 10.1101/2023.09.26.558361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Respiratory chemosensitivity is an important mechanism by which the brain senses changes in blood partial pressure of CO2 (PCO2). It is proposed that special neurons (and astrocytes) in various brainstem regions play key roles as CO2 central respiratory chemosensors in rodents. Although common marmosets (Callithrix jacchus), New-World non-human primates, show similar respiratory responses to elevated inspired CO2 as rodents, the chemosensitive regions in marmoset brain have not been defined yet. Here, we used c-fos immunostainings to identify brain-wide CO2-activated brain regions in common marmosets. In addition, we mapped the location of the retrotrapezoid nucleus (RTN) and raphé nuclei in the marmoset brainstem based on colocalization of CO2-induced c-fos immunoreactivity with Phox2b, and TPH immunostaining, respectively. Our data also indicated that, similar to rodents, marmoset RTN astrocytes express Phox2b and have complex processes that create a meshwork structure at the ventral surface of medulla. Our data highlight some cellular and structural regional similarities in brainstem of the common marmosets and rodents.
Collapse
Affiliation(s)
- Ariana Z. Turk
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Marissa Millwater
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| | - Shahriar SheikhBahaei
- Neuron-Glia Signaling and Circuits Unit, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, 20892 MD, USA
| |
Collapse
|
20
|
Gonye EC, Bayliss DA. Criteria for central respiratory chemoreceptors: experimental evidence supporting current candidate cell groups. Front Physiol 2023; 14:1241662. [PMID: 37719465 PMCID: PMC10502317 DOI: 10.3389/fphys.2023.1241662] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
An interoceptive homeostatic system monitors levels of CO2/H+ and provides a proportionate drive to respiratory control networks that adjust lung ventilation to maintain physiologically appropriate levels of CO2 and rapidly regulate tissue acid-base balance. It has long been suspected that the sensory cells responsible for the major CNS contribution to this so-called respiratory CO2/H+ chemoreception are located in the brainstem-but there is still substantial debate in the field as to which specific cells subserve the sensory function. Indeed, at the present time, several cell types have been championed as potential respiratory chemoreceptors, including neurons and astrocytes. In this review, we advance a set of criteria that are necessary and sufficient for definitive acceptance of any cell type as a respiratory chemoreceptor. We examine the extant evidence supporting consideration of the different putative chemoreceptor candidate cell types in the context of these criteria and also note for each where the criteria have not yet been fulfilled. By enumerating these specific criteria we hope to provide a useful heuristic that can be employed both to evaluate the various existing respiratory chemoreceptor candidates, and also to focus effort on specific experimental tests that can satisfy the remaining requirements for definitive acceptance.
Collapse
Affiliation(s)
- Elizabeth C. Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| | | |
Collapse
|
21
|
Brady CT, Marshall A, Zhang C, Parker MD. NBCe1-B/C-knockout mice exhibit an impaired respiratory response and an enhanced renal response to metabolic acidosis. Front Physiol 2023; 14:1201034. [PMID: 37405134 PMCID: PMC10315466 DOI: 10.3389/fphys.2023.1201034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
The sodium-bicarbonate cotransporter (NBCe1) has three primary variants: NBCe1-A, -B and -C. NBCe1-A is expressed in renal proximal tubules in the cortical labyrinth, where it is essential for reclaiming filtered bicarbonate, such that NBCe1-A knockout mice are congenitally acidemic. NBCe1-B and -C variants are expressed in chemosensitive regions of the brainstem, while NBCe1-B is also expressed in renal proximal tubules located in the outer medulla. Although mice lacking NBCe1-B/C (KOb/c) exhibit a normal plasma pH at baseline, the distribution of NBCe1-B/C indicates that these variants could play a role in both the rapid respiratory and slower renal responses to metabolic acidosis (MAc). Therefore, in this study we used an integrative physiologic approach to investigate the response of KOb/c mice to MAc. By means of unanesthetized whole-body plethysmography and blood-gas analysis, we demonstrate that the respiratory response to MAc (increase in minute volume, decrease in pCO2) is impaired in KOb/c mice leading to a greater severity of acidemia after 1 day of MAc. Despite this respiratory impairment, the recovery of plasma pH after 3-days of MAc remained intact in KOb/c mice. Using data gathered from mice housed in metabolic cages we demonstrate a greater elevation of renal ammonium excretion and greater downregulation of the ammonia recycling enzyme glutamine synthetase in KOb/c mice on day 2 of MAc, consistent with greater renal acid-excretion. We conclude that KOb/c mice are ultimately able to defend plasma pH during MAc, but that the integrated response is disturbed such that the burden of work shifts from the respiratory system to the kidneys, delaying the recovery of pH.
Collapse
Affiliation(s)
- Clayton T. Brady
- Jacobs School of Medicine and Biomedical Sciences, Department of Physiology and Biophysics, The State University of New York: The University at Buffalo, Buffalo, NY, United States
| | - Aniko Marshall
- Jacobs School of Medicine and Biomedical Sciences, Department of Physiology and Biophysics, The State University of New York: The University at Buffalo, Buffalo, NY, United States
| | - Chen Zhang
- Jacobs School of Medicine and Biomedical Sciences, Department of Physiology and Biophysics, The State University of New York: The University at Buffalo, Buffalo, NY, United States
- Department of Biological Sciences, The State University of New York: The University at Buffalo, Buffalo, NY, United States
| | - Mark D. Parker
- Jacobs School of Medicine and Biomedical Sciences, Department of Physiology and Biophysics, The State University of New York: The University at Buffalo, Buffalo, NY, United States
- Jacobs School of Medicine and Biomedical Sciences, Department of Ophthalmology, The State University of New York: The University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
22
|
Jones AA, Marino GM, Spears AR, Arble DM. The Molecular Circadian Clock of Phox2b-expressing Cells Drives Daily Variation of the Hypoxic but Not Hypercapnic Ventilatory Response in Mice. FUNCTION 2023; 4:zqad023. [PMID: 37342417 PMCID: PMC10278984 DOI: 10.1093/function/zqad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/22/2023] Open
Abstract
While the suprachiasmatic nucleus (SCN) controls 24-h rhythms in breathing, including minute ventilation (VE), the mechanisms by which the SCN drives these daily changes are not well understood. Moreover, the extent to which the circadian clock regulates hypercapnic and hypoxic ventilatory chemoreflexes is unknown. We hypothesized that the SCN regulates daily breathing and chemoreflex rhythms by synchronizing the molecular circadian clock of cells. We used whole-body plethysmography to assess ventilatory function in transgenic BMAL1 knockout (KO) mice to determine the role of the molecular clock in regulating daily rhythms in ventilation and chemoreflex. Unlike their wild-type littermates, BMAL1 KO mice exhibited a blunted daily rhythm in VE and failed to demonstrate daily variation in the hypoxic ventilatory response (HVR) or hypercapnic ventilatory response (HCVR). To determine if the observed phenotype was mediated by the molecular clock of key respiratory cells, we then assessed ventilatory rhythms in BMAL1fl/fl; Phox2bCre/+ mice, which lack BMAL1 in all Phox2b-expressing chemoreceptor cells (hereafter called BKOP). BKOP mice lacked daily variation in HVR, similar to BMAL1 KO mice. However, unlike BMAL1 KO mice, BKOP mice exhibited circadian variations in VE and HCVR comparable to controls. These data indicate that the SCN regulates daily rhythms in VE, HVR, and HCVR, in part, through the synchronization of the molecular clock. Moreover, the molecular clock of Phox2b-expressing cells is specifically necessary for daily variation in the hypoxic chemoreflex. These findings suggest that disruption of circadian biology may undermine respiratory homeostasis, which, in turn, may have clinical implications for respiratory disease.
Collapse
Affiliation(s)
- Aaron A Jones
- Department of Biological Sciences, Marquette University, WI 53233, USA
| | | | - Allison R Spears
- Department of Biological Sciences, Marquette University, WI 53233, USA
| | | |
Collapse
|
23
|
Abstract
The rhythmicity of breath is vital for normal physiology. Even so, breathing is enriched with multifunctionality. External signals constantly change breathing, stopping it when under water or deepening it during exertion. Internal cues utilize breath to express emotions such as sighs of frustration and yawns of boredom. Breathing harmonizes with other actions that use our mouth and throat, including speech, chewing, and swallowing. In addition, our perception of breathing intensity can dictate how we feel, such as during the slow breathing of calming meditation and anxiety-inducing hyperventilation. Heartbeat originates from a peripheral pacemaker in the heart, but the automation of breathing arises from neural clusters within the brainstem, enabling interaction with other brain areas and thus multifunctionality. Here, we document how the recent transformation of cellular and molecular tools has contributed to our appreciation of the diversity of neuronal types in the breathing control circuit and how they confer the multifunctionality of breathing.
Collapse
Affiliation(s)
- Kevin Yackle
- Department of Physiology, University of California, San Francisco, California, USA;
| |
Collapse
|
24
|
Pereyra K, Díaz-Jara E, Arias P, Bravo L, Toledo C, Schwarz K, Del Rio R. Role of Peripheral Chemoreceptors on Enhanced Central Chemoreflex Drive in Nonischemic Heart Failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1427:107-114. [PMID: 37322341 DOI: 10.1007/978-3-031-32371-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Heart failure (HF) is a prevalent disease in elderly population. Potentiation of the ventilatory chemoreflex drive plays a pivotal role in disease progression, at least in part, through their contribution to the generation/maintenance of breathing disorders. Peripheral and central chemoreflexes are mainly regulated by carotid body (CB) and the retrotrapezoid nuclei (RTN), respectively. Recent evidence showed an enhanced central chemoreflex drive in rats with nonischemic HF along with breathing disorders. Importantly, increase activity from RTN chemoreceptors contribute to the potentiation of central chemoreflex response to hypercapnia. The precise mechanism driving RTN potentiation in HF is still elusive. Since interdependency of RTN and CB chemoreceptors has been described, we hypothesized that CB afferent activity is required to increase RTN chemosensitivity in the setting of HF. Accordingly, we studied central/peripheral chemoreflex drive and breathing disorders in HF rats with and without functional CBs (CB denervation). We found that CB afferent activity was required to increase central chemoreflex drive in HF. Indeed, CB denervation restored normal central chemoreflex drive and reduced the incidence of apneas by twofold. Our results support the notion that CB afferent activity plays an important role in central chemoreflex potentiation in rats with HF.
Collapse
Affiliation(s)
- Katherin Pereyra
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Arias
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Liena Bravo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karla Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
25
|
Schottelkotte KM, Crone SA. Forebrain control of breathing: Anatomy and potential functions. Front Neurol 2022; 13:1041887. [PMID: 36388186 PMCID: PMC9663927 DOI: 10.3389/fneur.2022.1041887] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/11/2022] [Indexed: 01/25/2023] Open
Abstract
The forebrain plays important roles in many critical functions, including the control of breathing. We propose that the forebrain is important for ensuring that breathing matches current and anticipated behavioral, emotional, and physiological needs. This review will summarize anatomical and functional evidence implicating forebrain regions in the control of breathing. These regions include the cerebral cortex, extended amygdala, hippocampus, hypothalamus, and thalamus. We will also point out areas where additional research is needed to better understand the specific roles of forebrain regions in the control of breathing.
Collapse
Affiliation(s)
- Karl M. Schottelkotte
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Steven A. Crone
- Division of Pediatric Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
26
|
Bhandare A, van de Wiel J, Roberts R, Braren I, Huckstepp R, Dale N. Analyzing the brainstem circuits for respiratory chemosensitivity in freely moving mice. eLife 2022; 11:e70671. [PMID: 36300918 PMCID: PMC9643001 DOI: 10.7554/elife.70671] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/12/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of systemic PCO2 is a life-preserving homeostatic mechanism. In the medulla oblongata, the retrotrapezoid nucleus (RTN) and rostral medullary Raphe are proposed as CO2 chemosensory nuclei mediating adaptive respiratory changes. Hypercapnia also induces active expiration, an adaptive change thought to be controlled by the lateral parafacial region (pFL). Here, we use GCaMP6 expression and head-mounted mini-microscopes to image Ca2+ activity in these nuclei in awake adult mice during hypercapnia. Activity in the pFL supports its role as a homogenous neuronal population that drives active expiration. Our data show that chemosensory responses in the RTN and Raphe differ in their temporal characteristics and sensitivity to CO2, raising the possibility these nuclei act in a coordinated way to generate adaptive ventilatory responses to hypercapnia. Our analysis revises the understanding of chemosensory control in awake adult mouse and paves the way to understanding how breathing is coordinated with complex non-ventilatory behaviours.
Collapse
Affiliation(s)
- Amol Bhandare
- School of Life Sciences, University of WarwickCoventryUnited Kingdom
| | | | - Reno Roberts
- School of Life Sciences, University of WarwickCoventryUnited Kingdom
| | - Ingke Braren
- University Medical Center Eppendorf, Vector Facility, Institute of Experimental Pharmacology and ToxicologyHamburgGermany
| | - Robert Huckstepp
- School of Life Sciences, University of WarwickCoventryUnited Kingdom
| | - Nicholas Dale
- School of Life Sciences, University of WarwickCoventryUnited Kingdom
| |
Collapse
|
27
|
Souza GMPR, Stornetta DS, Vitali AJ, Wildner H, Zeilhofer HU, Campbell JN, Abbott SBG. Chemogenetic activation of noradrenergic A5 neurons increases blood pressure and visceral sympathetic activity in adult rats. Am J Physiol Regul Integr Comp Physiol 2022; 323:R512-R531. [PMID: 35993562 PMCID: PMC9602699 DOI: 10.1152/ajpregu.00119.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022]
Abstract
In mammals, the pontine noradrenergic system influences nearly every aspect of central nervous system function. A subpopulation of pontine noradrenergic neurons, called A5, are thought to be important in the cardiovascular response to physical stressors, yet their function is poorly defined. We hypothesized that activation of A5 neurons drives a sympathetically mediated increase in blood pressure (BP). To test this hypothesis, we conducted a comprehensive assessment of the cardiovascular effects of chemogenetic stimulation of A5 neurons in male and female adult rats using intersectional genetic and anatomical targeting approaches. Chemogenetic stimulation of A5 neurons in freely behaving rats elevated BP by 15 mmHg and increased cardiac baroreflex sensitivity with a negligible effect on resting HR. Importantly, A5 stimulation had no detectable effect on locomotor activity, metabolic rate, or respiration. Under anesthesia, stimulation of A5 neurons produced a marked elevation in visceral sympathetic nerve activity (SNA) and no change in skeletal muscle SNA, showing that A5 neurons preferentially stimulate visceral SNA. Interestingly, projection mapping indicates that A5 neurons target sympathetic preganglionic neurons throughout the spinal cord and parasympathetic preganglionic neurons throughout in the brainstem, as well as the nucleus of the solitary tract, and ventrolateral medulla. Moreover, in situ hybridization and immunohistochemistry indicate that a subpopulation of A5 neurons coreleases glutamate and monoamines. Collectively, this study suggests A5 neurons are a central modulator of autonomic function with a potentially important role in sympathetically driven redistribution of blood flow from the visceral circulation to critical organs and skeletal muscle.
Collapse
Affiliation(s)
- George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Daniel S Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Alexander J Vitali
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Hanns U Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - John N Campbell
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
28
|
Silva TM, Wasinski F, Flor KC, List EO, Kopchick JJ, Takakura AC, Donato J, Moreira TS. The effect of central growth hormone action on hypoxia ventilatory response in conscious mice. Brain Res 2022; 1791:147995. [PMID: 35779583 PMCID: PMC10938300 DOI: 10.1016/j.brainres.2022.147995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
Growth hormone (GH)-responsive neurons regulate several homeostatic behaviors including metabolism, energy balance, arousal, and stress response. Therefore, it is possible that GH-responsive neurons play a role in other responses such as CO2/H+-dependent breathing behaviors. Here, we investigated whether central GH receptor (GHR) modulates respiratory activity in conscious unrestrained mice. First, we detected clusters of GH-responsive neurons in the tyrosine hydroxylase-expressing cells in the rostroventrolateral medulla (C1 region) and within the locus coeruleus (LC). No significant expression was detected in phox2b-expressing cells in the retrotrapezoid nucleus. Whole body plethysmography revealed a reduction in the tachypneic response to hypoxia (FiO2 = 0.08) without changing baseline breathing and the hypercapnic ventilatory response. Contrary to the physiological findings, we did not find significant differences in the number of fos-activated cells in the nucleus of the solitary tract (NTS), C1, LC and paraventricular nucleus of the hypothalamus (PVH). Our finding suggests a possible secondary role of central GH action in the tachypneic response to hypoxia in conscious mice.
Collapse
Affiliation(s)
- Talita M Silva
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Frederick Wasinski
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Karine C Flor
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), 05508-000 São Paulo/SP, Brazil.
| |
Collapse
|
29
|
Chemogenetic inhibition of Phox2-expressing neurons in the commissural NTS decreases blood pressure in anesthetized spontaneously hypertensive rats. Neurosci Lett 2022; 787:136817. [PMID: 35905886 DOI: 10.1016/j.neulet.2022.136817] [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: 03/27/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/21/2022]
Abstract
Interruption of the activity of neurons in the commissural portion of the nucleus of the solitary tract (cNTS) decreases blood pressure (BP) in experimental models of hypertension, such as the spontaneously hypertensive (SH) rat. To examine whether PHOX2B expressing cNTS neurons are involved in maintaining the elevated BP, we used replication-deficient viruses with a modified Phox2 binding site promoter to express the inhibitory chemogenetic allatostatin receptor or green fluorescent protein in the cNTS. Following administration of allatostatin, we observed a depressor and bradycardic response in anesthetized SH rats that expressed the allatostatin receptor. Injection of allatostatin did not affect BP or heart rate (HR) in control SH rats expressing green fluorescent protein in the cNTS. Immunohistochemistry showed that the majority of transduced cNTS neurons were PHOX2B-immunoreactive and some also expressed tyrosine hydroxylase. We conclude that in anesthetized SH rat, the Phox2B expressing cNTS neurons maintain elevated BP.
Collapse
|
30
|
Alturkustani M, Walker AD, Tran I, Snuderl M, Cotter JA. Spectrum of paired-like homeobox 2b immunoexpression in pediatric brain tumors with embryonal morphology. Pathol Int 2022; 72:402-410. [PMID: 35763016 PMCID: PMC9544136 DOI: 10.1111/pin.13255] [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: 02/02/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
Paired‐like homeobox 2b (PHOX2B) is an established immunomarker for peripheral neuroblastoma and autonomic nervous system cells. We aimed to evaluate the utility of PHOX2B immunostaining in central nervous system (CNS) tumors with embryonal morphology. Fifty‐one tumors were stained with PHOX2B and submitted for whole slide image analysis: 35 CNS tumors with embryonal morphology (31 CNS embryonal tumors and four gliomas); and 16 peripheral neuroblastomas were included for comparison. Diffuse nuclear immunopositivity was observed in all (16/16) neuroblastomas (primary and metastatic). Among CNS embryonal tumors, focal immunoreactivity for PHOX2B was observed in most (5/7) embryonal tumors with multilayered rosettes (ETMR) and a single high‐grade neuroepithelial tumor (HGNET) with PLAGL2 amplification; the remaining 27 CNS tumors were essentially immunonegative (<0.05% positive). Among ETMR, PHOX2B expression was observed in a small overall proportion (0.04%–4.94%) of neoplastic cells but focally reached up to 39% in 1 mm ‘hot spot’ areas. In the PLAGL2‐amplified case, 0.09% of the total neoplastic population was immunoreactive, with 0.53% in the ‘hot spot’ area. Care should be taken in interpreting PHOX2B immunopositivity in a differential diagnosis that includes metastatic neuroblastoma and CNS tumors; focal or patchy expression should not be considered definitively diagnostic of metastatic peripheral neuroblastoma.
Collapse
Affiliation(s)
- Murad Alturkustani
- Department of Pathology, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | - Adam D Walker
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Ivy Tran
- Department of Pathology, New York University Langone Medical Center, New York, New York, USA
| | - Matija Snuderl
- Department of Pathology, New York University Langone Medical Center, New York, New York, USA
| | - Jennifer A Cotter
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
31
|
Zyuzin J, Jendzjowsky N. Neuroanatomic and neurophysiologic evidence of pulmonary nociceptor and carotid chemoreceptor convergence in the nucleus tractus solitarius and nucleus ambiguus. J Neurophysiol 2022; 127:1511-1518. [PMID: 35443145 PMCID: PMC9142158 DOI: 10.1152/jn.00125.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pulmonary vagal nociceptors defend the airways. Cardiopulmonary vagal nociceptors synapse in the nucleus tractus solitarius (NTS). Evidence has demonstrated the convergence of cardiopulmonary nociceptors with afferents from carotid chemoreceptors. Whether sensory convergence occurs in motor nuclei and how sensory convergence affects reflexive efferent motor output directed toward the airways are critical knowledge gaps. Here, we show that distinct tracer injection into the pulmonary nociceptors and carotid chemoreceptors leads to co-labeled neurons in the nucleus tractus solitarius and nucleus ambiguus. Precise simultaneous stimulation delivered to pulmonary nociceptors and carotid chemoreceptors doubled efferent vagal output, enhanced phrenic pause, and subsequently augmented phrenic motor activity. These results suggest that multiple afferents are involved in protecting the airways and concurrent stimulation enhances airway defensive reflex output. NEW & NOTEWORTHY Sensory afferents have been shown to converge onto nucleus tractus solitarius primary neurons. Here, we show sensory convergence of two distinct sets of sensory afferents in motor nuclei of the nucleus ambiguus, which results in augmentation of airway defense motor output.
Collapse
Affiliation(s)
- Jekaterina Zyuzin
- Respiratory and Critical Care Medicine and Physiology and, Neurotherapeutics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance California, United States
| | - Nicholas Jendzjowsky
- Respiratory and Critical Care Medicine and Physiology and, Neurotherapeutics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance California, United States
| |
Collapse
|
32
|
Shi Y, Sobrinho CR, Soto-Perez J, Milla BM, Stornetta DS, Stornetta RL, Takakura AC, Mulkey DK, Moreira TS, Bayliss DA. 5-HT7 receptors expressed in the mouse parafacial region are not required for respiratory chemosensitivity. J Physiol 2022; 600:2789-2811. [PMID: 35385139 PMCID: PMC9167793 DOI: 10.1113/jp282279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/23/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract A brainstem homeostatic system senses CO2/H+ to regulate ventilation, blood gases and acid–base balance. Neurons of the retrotrapezoid nucleus (RTN) and medullary raphe are both implicated in this mechanism as respiratory chemosensors, but recent pharmacological work suggested that the CO2/H+ sensitivity of RTN neurons is mediated indirectly, by raphe‐derived serotonin acting on 5‐HT7 receptors. To investigate this further, we characterized Htr7 transcript expression in phenotypically identified RTN neurons using multiplex single cell qRT‐PCR and RNAscope. Although present in multiple neurons in the parafacial region of the ventrolateral medulla, Htr7 expression was undetectable in most RTN neurons (Nmb+/Phox2b+) concentrated in the densely packed cell group ventrolateral to the facial nucleus. Where detected, Htr7 expression was modest and often associated with RTN neurons that extend dorsolaterally to partially encircle the facial nucleus. These dorsolateral Nmb+/Htr7+ neurons tended to express Nmb at high levels and the intrinsic RTN proton detectors Gpr4 and Kcnk5 at low levels. In mouse brainstem slices, CO2‐stimulated firing in RTN neurons was mostly unaffected by a 5‐HT7 receptor antagonist, SB269970 (n = 11/13). At the whole animal level, microinjection of SB269970 into the RTN of conscious mice blocked respiratory stimulation by co‐injected LP‐44, a 5‐HT7 receptor agonist, but had no effect on CO2‐stimulated breathing in those same mice. We conclude that Htr7 is expressed by a minor subset of RTN neurons with a molecular profile distinct from the established chemoreceptors and that 5‐HT7 receptors have negligible effects on CO2‐evoked firing activity in RTN neurons or on CO2‐stimulated breathing in mice. Key points Neurons of the retrotrapezoid nucleus (RTN) are intrinsic CO2/H+ chemosensors and serve as an integrative excitatory hub for control of breathing. Serotonin can activate RTN neurons, in part via 5‐HT7 receptors, and those effects have been implicated in conferring an indirect CO2 sensitivity. Multiple single cell molecular approaches revealed low levels of 5‐HT7 receptor transcript expression restricted to a limited population of RTN neurons. Pharmacological experiments showed that 5‐HT7 receptors in RTN are not required for CO2/H+‐stimulation of RTN neuronal activity or CO2‐stimulated breathing. These data do not support a role for 5‐HT7 receptors in respiratory chemosensitivity mediated by RTN neurons.
Collapse
Affiliation(s)
- Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Cleyton R Sobrinho
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jaseph Soto-Perez
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Brenda M Milla
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Daniel S Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Daniel K Mulkey
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
33
|
Souza GMPR, Stornetta RL, Stornetta DS, Guyenet PG, Abbott SBG. Adrenergic C1 neurons monitor arterial blood pressure and determine the sympathetic response to hemorrhage. Cell Rep 2022; 38:110480. [PMID: 35263582 DOI: 10.1016/j.celrep.2022.110480] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 11/16/2021] [Accepted: 02/11/2022] [Indexed: 11/03/2022] Open
Abstract
Hemorrhage initially triggers a rise in sympathetic nerve activity (SNA) that maintains blood pressure (BP); however, SNA is suppressed following severe blood loss causing hypotension. We hypothesized that adrenergic C1 neurons in the rostral ventrolateral medulla (C1RVLM) drive the increase in SNA during compensated hemorrhage, and a reduction in C1RVLM contributes to hypotension during decompensated hemorrhage. Using fiber photometry, we demonstrate that C1RVLM activity increases during compensated hemorrhage and falls at the onset of decompensated hemorrhage. Using optogenetics combined with direct recordings of SNA, we show that C1RVLM activation mediates the rise in SNA and contributes to BP stability during compensated hemorrhage, whereas a suppression of C1RVLM activity is associated with cardiovascular collapse during decompensated hemorrhage. Notably, re-activating C1RVLM during decompensated hemorrhage restores BP to normal levels. In conclusion, C1 neurons are a nodal point for the sympathetic response to blood loss.
Collapse
Affiliation(s)
- George M P R Souza
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA, USA
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA, USA
| | - Daniel S Stornetta
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA, USA
| | - Patrice G Guyenet
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA, USA
| | - Stephen B G Abbott
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA, USA.
| |
Collapse
|
34
|
Hirsch D, Kohl A, Wang Y, Sela-Donenfeld D. Axonal Projection Patterns of the Dorsal Interneuron Populations in the Embryonic Hindbrain. Front Neuroanat 2022; 15:793161. [PMID: 35002640 PMCID: PMC8738170 DOI: 10.3389/fnana.2021.793161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Unraveling the inner workings of neural circuits entails understanding the cellular origin and axonal pathfinding of various neuronal groups during development. In the embryonic hindbrain, different subtypes of dorsal interneurons (dINs) evolve along the dorsal-ventral (DV) axis of rhombomeres and are imperative for the assembly of central brainstem circuits. dINs are divided into two classes, class A and class B, each containing four neuronal subgroups (dA1-4 and dB1-4) that are born in well-defined DV positions. While all interneurons belonging to class A express the transcription factor Olig3 and become excitatory, all class B interneurons express the transcription factor Lbx1 but are diverse in their excitatory or inhibitory fate. Moreover, within every class, each interneuron subtype displays its own specification genes and axonal projection patterns which are required to govern the stage-by-stage assembly of their connectivity toward their target sites. Remarkably, despite the similar genetic landmark of each dINs subgroup along the anterior-posterior (AP) axis of the hindbrain, genetic fate maps of some dA/dB neuronal subtypes uncovered their contribution to different nuclei centers in relation to their rhombomeric origin. Thus, DV and AP positional information has to be orchestrated in each dA/dB subpopulation to form distinct neuronal circuits in the hindbrain. Over the span of several decades, different axonal routes have been well-documented to dynamically emerge and grow throughout the hindbrain DV and AP positions. Yet, the genetic link between these distinct axonal bundles and their neuronal origin is not fully clear. In this study, we reviewed the available data regarding the association between the specification of early-born dorsal interneuron subpopulations in the hindbrain and their axonal circuitry development and fate, as well as the present existing knowledge on molecular effectors underlying the process of axonal growth.
Collapse
Affiliation(s)
- Dana Hirsch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.,Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
35
|
Smith JC. Respiratory rhythm and pattern generation: Brainstem cellular and circuit mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:1-35. [PMID: 35965022 DOI: 10.1016/b978-0-323-91534-2.00004-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Breathing movements in mammals are driven by rhythmic neural activity automatically generated within spatially and functionally organized brainstem neural circuits comprising the respiratory central pattern generator (CPG). This chapter reviews up-to-date experimental information and theoretical studies of the cellular and circuit mechanisms of respiratory rhythm and pattern generation operating within critical components of this CPG in the lower brainstem. Over the past several decades, there have been substantial advances in delineating the spatial architecture of essential medullary regions and their regional cellular and circuit properties required to understand rhythm and pattern generation mechanisms. A fundamental concept is that the circuits in these regions have rhythm-generating capabilities at multiple cellular and circuit organization levels. The regional cellular properties, circuit organization, and control mechanisms allow flexible expression of neural activity patterns for a repertoire of respiratory behaviors under various physiologic conditions that are dictated by requirements for homeostatic regulation and behavioral integration. Many mechanistic insights have been provided by computational modeling studies driven by experimental results and have advanced understanding in the field. These conceptual and theoretical developments are discussed.
Collapse
Affiliation(s)
- Jeffrey C Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
36
|
Aquino YC, Cabral LM, Miranda NC, Naccarato MC, Falquetto B, Moreira TS, Takakura AC. Respiratory disorders of Parkinson's disease. J Neurophysiol 2022; 127:1-15. [PMID: 34817281 DOI: 10.1152/jn.00363.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra, mainly affecting people over 60 yr of age. Patients develop both classic symptoms (tremors, muscle rigidity, bradykinesia, and postural instability) and nonclassical symptoms (orthostatic hypotension, neuropsychiatric deficiency, sleep disturbances, and respiratory disorders). Thus, patients with PD can have a significantly impaired quality of life, especially when they do not have multimodality therapeutic follow-up. The respiratory alterations associated with this syndrome are the main cause of mortality in PD. They can be classified as peripheral when caused by disorders of the upper airways or muscles involved in breathing and as central when triggered by functional deficits of important neurons located in the brainstem involved in respiratory control. Currently, there is little research describing these disorders, and therefore, there is no well-established knowledge about the subject, making the treatment of patients with respiratory symptoms difficult. In this review, the history of the pathology and data about the respiratory changes in PD obtained thus far will be addressed.
Collapse
Affiliation(s)
- Yasmin C Aquino
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Laís M Cabral
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Nicole C Miranda
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Monique C Naccarato
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Bárbara Falquetto
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Abstract
Brain PCO2 is sensed primarily via changes in [H+]. Small pH changes are detected in the medulla oblongata and trigger breathing adjustments that help maintain arterial PCO2 constant. Larger perturbations of brain CO2/H+, possibly also sensed elsewhere in the CNS, elicit arousal, dyspnea, and stress, and cause additional breathing modifications. The retrotrapezoid nucleus (RTN), a rostral medullary cluster of glutamatergic neurons identified by coexpression of Phoxb and Nmb transcripts, is the lynchpin of the central respiratory chemoreflex. RTN regulates breathing frequency, inspiratory amplitude, and active expiration. It is exquisitely responsive to acidosis in vivo and maintains breathing autorhythmicity during quiet waking, slow-wave sleep, and anesthesia. The RTN response to [H+] is partly an intrinsic neuronal property mediated by proton sensors TASK-2 and GPR4 and partly a paracrine effect mediated by astrocytes and the vasculature. The RTN also receives myriad excitatory or inhibitory synaptic inputs including from [H+]-responsive neurons (e.g., serotonergic). RTN is silenced by moderate hypoxia. RTN inactivity (periodic or sustained) contributes to periodic breathing and, likely, to central sleep apnea. RTN development relies on transcription factors Egr2, Phox2b, Lbx1, and Atoh1. PHOX2B mutations cause congenital central hypoventilation syndrome; they impair RTN development and consequently the central respiratory chemoreflex.
Collapse
Affiliation(s)
- Patrice G Guyenet
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States.
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
38
|
Kajiwara R, Nakamura S, Ikeda K, Onimaru H, Yoshida A, Tsutsumi Y, Nakayama K, Mochizuki A, Dantsuji M, Nishimura A, Tachikawa S, Iijima T, Inoue T. Intrinsic properties and synaptic connectivity of Phox2b-expressing neurons in rat rostral parvocellular reticular formation. Neurosci Res 2021; 178:41-51. [PMID: 34973291 DOI: 10.1016/j.neures.2021.12.009] [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: 08/27/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
The paired-like homeobox 2b gene (Phox2b) is critical for the development of the autonomic nervous system. We have previously demonstrated the distinct characteristics of Phox2b-expressing (Phox2b+) neurons in the reticular formation dorsal to the trigeminal motor nucleus (RdV), which are likely related to jaw movement regulation. In this study, we focused on Phox2b+ neurons in the rostral parvocellular reticular formation (rPCRt), a critical region for controlling orofacial functions, using 2-11-day-old Phox2b-EYFP rats. Most Phox2b+ rPCRt neurons were glutamatergic, but not GABAergic or glycinergic. Approximately 65 % of Phox2b+ rPCRt neurons fired at a low frequency, and approximately 24 % of Phox2b+ rPCRt neurons fired spontaneously, as opposed to Phox2b+ RdV neurons. Stimulation of the RdV evoked inward postsynaptic currents in more than 50 % of Phox2b+ rPCRt neurons, while only one Phox2b+ rPCRt neuron responded to stimulation of the nucleus of the solitary tract. Five of the 10 Phox2b+ neurons sent their axons that ramified within the trigeminal motor nucleus (MoV). Of these, the axons of the two neurons terminated within both the MoV and rPCRt. Our findings suggest that Phox2b+ rPCRt neurons have distinct electrophysiological and synaptic properties that may be involved in the motor control of feeding behavior.
Collapse
Affiliation(s)
- Risa Kajiwara
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan; Department of Perioperative Medicine, Division of Anesthesiology, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Keiko Ikeda
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Atsushi Yoshida
- Department of Oral Anatomy and Neurobiology, Osaka University Graduate School of Dentistry, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Yumi Tsutsumi
- Department of Oral Anatomy and Neurobiology, Osaka University Graduate School of Dentistry, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Kiyomi Nakayama
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Ayako Mochizuki
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Masanori Dantsuji
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Akiko Nishimura
- Department of Perioperative Medicine, Division of Anesthesiology, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Satoshi Tachikawa
- Department of Perioperative Medicine, Division of Anesthesiology, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Takehiko Iijima
- Department of Perioperative Medicine, Division of Anesthesiology, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Tomio Inoue
- Department of Oral Physiology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| |
Collapse
|
39
|
Adolescent Congenital Central Hypoventilation Syndrome: An Easily Overlooked Diagnosis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182413402. [PMID: 34949014 PMCID: PMC8703802 DOI: 10.3390/ijerph182413402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022]
Abstract
Congenital central hypoventilation syndrome (CCHS), also known as Ondine’s curse, is a rare, potentially fatal genetic disease, manifesting as a lack of respiratory drive. Most diagnoses are made in pediatric patients, however late-onset cases have been rarely reported. Due to the milder symptoms at presentation that might easily go overlooked, these late-onset cases can result in serious health consequences later in life. Here, we present a case report of late-onset CCHS in an adolescent female patient. In this review we summarize the current knowledge about symptoms, as well as clinical management of CCHS, and describe in detail the molecular mechanism responsible for this disorder.
Collapse
|
40
|
Guyenet PG, Stornetta RL. Rostral ventrolateral medulla, retropontine region and autonomic regulations. Auton Neurosci 2021; 237:102922. [PMID: 34814098 DOI: 10.1016/j.autneu.2021.102922] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/08/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022]
Abstract
The rostral half of the ventrolateral medulla (RVLM) and adjacent ventrolateral retropontine region (henceforth RVLMRP) have been divided into various sectors by neuroscientists interested in breathing or autonomic regulations. The RVLMRP regulates respiration, glycemia, vigilance and inflammation, in addition to blood pressure. It contains interoceptors that respond to acidification, hypoxia and intracranial pressure and its rostral end contains the retrotrapezoid nucleus (RTN) which is the main central respiratory chemoreceptor. Acid detection by the RTN is an intrinsic property of the principal neurons that is enhanced by paracrine influences from surrounding astrocytes and CO2-dependent vascular constriction. RTN mediates the hypercapnic ventilatory response via complex projections to the respiratory pattern generator (CPG). The RVLM contributes to autonomic response patterns via differential recruitment of several subtypes of adrenergic (C1) and non-adrenergic neurons that directly innervate sympathetic and parasympathetic preganglionic neurons. The RVLM also innervates many brainstem and hypothalamic nuclei that contribute, albeit less directly, to autonomic responses. All lower brainstem noradrenergic clusters including the locus coeruleus are among these targets. Sympathetic tone to the circulatory system is regulated by subsets of presympathetic RVLM neurons whose activity is continuously restrained by the baroreceptors and modulated by the respiratory CPG. The inhibitory input from baroreceptors and the excitatory input from the respiratory CPG originate from neurons located in or close to the rhythm generating region of the respiratory CPG (preBötzinger complex).
Collapse
Affiliation(s)
- Patrice G Guyenet
- University of Virginia School of Medicine, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908-0735, USA.
| | - Ruth L Stornetta
- University of Virginia School of Medicine, Department of Pharmacology, 1340 Jefferson Park Avenue, Charlottesville, VA 22908-0735, USA.
| |
Collapse
|
41
|
Malheiros-Lima MR, Silva TM, Takakura AC, Moreira TS. A5 noradrenergic-projecting C1 neurons activate sympathetic and breathing outputs in anaesthetized rats. Exp Physiol 2021; 107:147-160. [PMID: 34813109 DOI: 10.1113/ep089691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 11/16/2021] [Indexed: 12/18/2022]
Abstract
NEW FINDINGS What is the central question of this study? C1 neurons innervate pontine noradrenergic cell groups, including the A5 region: do A5 noradrenergic neurons contribute to the activation of sympathetic and respiratory responses produced by selective activation of the C1 group of neurons. What is the main finding and its importance? The increase in sympathetic and respiratory activities elicited by selective stimulation of C1 neurons is reduced after blockade of excitatory amino acid within the A5 region, suggesting that the C1-A5 pathway might be important for sympathetic-respiratory control. ABSTRACT Adrenergic C1 neurons innervate and excite pontine noradrenergic cell groups, including the ventrolateral pontine noradrenergic region (A5). Here, we tested the hypothesis that C1 activates A5 neurons through the release of glutamate and this effect is important for sympathetic and respiratory control. Using selective tools, we restricted the expression of channelrhodopsin2 under the control of the artificial promoter PRSx8 to C1 neurons (69%). Transduced catecholaminergic terminals within the A5 region are in contact with noradrenergic A5 neurons and the C1 terminals within the A5 region are predominantly glutamatergic. In a different group of animals, we performed retrograde lesion of C1 adrenergic neurons projecting to the A5 region with unilateral injection of the immunotoxin anti-dopamine β-hydroxylase-saporin (anti-DβH-SAP) directly into the A5 region during the hypoxic condition. As expected, hypoxia (8% O2 , 3 h) induced a robust increase in fos expression within the catecholaminergic C1 and A5 regions of the brainstem. Depletion of C1 cells projecting to the A5 regions reduced fos immunoreactivity induced by hypoxia within the C1 region. Physiological experiments showed that bilateral injection of kynurenic acid (100 mM) into the A5 region reduced the rise in mean arterial pressure, and sympathetic and phrenic nerve activities produced by optogenetic stimulation of C1 cells. In conclusion, the C1 neurons activate the ventrolateral pontine noradrenergic neurons (A5 region) possibly via the release of glutamate and might be important for sympathetic and respiratory outputs in anaesthetized rats.
Collapse
Affiliation(s)
- Milene R Malheiros-Lima
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, SP, Brazil
| | - Talita M Silva
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, SP, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, SP, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
42
|
Baizer JS, Webster CJ, Witelson SF. Individual variability in the size and organization of the human arcuate nucleus of the medulla. Brain Struct Funct 2021; 227:159-176. [PMID: 34613435 DOI: 10.1007/s00429-021-02396-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
The arcuate nucleus (Arc) of the medulla is found in almost all human brains and in a small percentage of chimpanzee brains. It is absent in the brains of other mammalian species including mice, rats, cats, and macaque monkeys. The Arc is classically considered a precerebellar relay nucleus, receiving input from the cerebral cortex and projecting to the cerebellum via the inferior cerebellar peduncle. However, several studies have found aplasia of the Arc in babies who died of SIDS (Sudden Infant Death Syndrome), and it was suggested that the Arc is the locus of chemosensory neurons critical for brainstem control of respiration. Aplasia of the Arc, however, has also been reported in adults, suggesting that it is not critical for survival. We have examined the Arc in closely spaced Nissl-stained sections in thirteen adult human cases to acquire a better understanding of the degree of variability of its size and location in adults. We have also examined immunostained sections to look for neurochemical compartments in this nucleus. Caudally, neurons of the Arc are ventrolateral to the pyramidal tracts (py); rostrally, they are ventro-medial to the py and extend up along the midline. In some cases, the Arc is discontinuous, with a gap between sections with the ventrolaterally located and the ventromedially located neurons. In all cases, there is some degree of left-right asymmetry in Arc position, size, and shape at all rostro-caudal levels. Somata of neurons in the Arc express calretinin (CR), neuronal nitric oxide synthase (nNOS), and nonphosphorylated neurofilament protein (NPNFP). Calbindin (CB) is expressed in puncta whereas there is no expression of parvalbumin (PV) in somata or puncta. There is also immunostaining for GAD and GABA receptors suggesting inhibitory input to Arc neurons. These properties were consistent among cases. Our data show differences in location of caudal and rostral Arc neurons and considerable variability among cases in the size and shape of the Arc. The variability in size suggests that "hypoplasia" of the Arc is difficult to define. The discontinuity of the Arc in many cases suggests that establishing aplasia of the Arc requires examination of many closely spaced sections through the brainstem.
Collapse
Affiliation(s)
- Joan S Baizer
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 123 Sherman Hall, South Campus, Buffalo, NY, 14214, USA.
| | - Charles J Webster
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 123 Sherman Hall, South Campus, Buffalo, NY, 14214, USA
| | - Sandra F Witelson
- Department of Psychiatry and Behavioural Neurosciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, L8S 4K1, Canada
| |
Collapse
|
43
|
Magalhães KS, da Silva MP, Mecawi AS, Paton JFR, Machado BH, Moraes DJA. Intrinsic and synaptic mechanisms controlling the expiratory activity of excitatory lateral parafacial neurones of rats. J Physiol 2021; 599:4925-4948. [PMID: 34510468 DOI: 10.1113/jp281545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
Active expiration is essential for increasing pulmonary ventilation during high chemical drive (hypercapnia). The lateral parafacial (pFL ) region, which contains expiratory neurones, drives abdominal muscles during active expiration in response to hypercapnia. However, the electrophysiological properties and synaptic mechanisms determining the activity of pFL expiratory neurones, as well as the specific conditions for their emergence, are not fully understood. Using whole cell electrophysiology and single cell quantitative RT-PCR techniques, we describe the intrinsic electrophysiological properties, the phenotype and the respiratory-related synaptic inputs to the pFL expiratory neurones, as well as the mechanisms for the expression of their expiratory activity under conditions of hypercapnia-induced active expiration, using in situ preparations of juvenile rats. We also evaluated whether these neurones possess intrinsic CO2 /[H+ ] sensitivity and burst generating properties. GABAergic and glycinergic inhibition during inspiration and expiration suppressed the activity of glutamatergic pFL expiratory neurones in normocapnia. In hypercapnia, these neurones escape glycinergic inhibition and generate burst discharges at the end of expiration. Evidence for the contribution of post-inhibitory rebound, CaV 3.2 isoform of T-type Ca2+ channels and intracellular [Ca2+ ] is presented. Neither intrinsic bursting properties, mediated by persistent Na+ current, nor CO2 /[H+ ] sensitivity or expression of CO2 /[H+ ] sensitive ion channels/receptors (TASK or GPR4) were observed. On the other hand, hyperpolarisation-activated cyclic nucleotide-gated and twik-related K+ leak channels were recorded. Post-synaptic disinhibition and the intrinsic electrophysiological properties of glutamatergic neurones play important roles in the generation of the expiratory oscillations in the pFL region during hypercapnia in rats. KEY POINTS: Hypercapnia induces active expiration in rats and the recruitment of a specific population of expiratory neurones in the lateral parafacial (pFL ) region. Post-synaptic GABAergic and glycinergic inhibition both suppress the activity of glutamatergic pFL neurones during inspiratory and expiratory phases in normocapnia. Hypercapnia reduces glycinergic inhibition during expiration leading to burst generation by pFL neurones; evidence for a contribution of post-inhibitory rebound, voltage-gated Ca2+ channels and intracellular [Ca2+ ] is presented. pFL glutamatergic expiratory neurones are neither intrinsic burster neurones, nor CO2 /[H+ ] sensors, and do not express CO2 /[H+ ] sensitive ion channels or receptors. Post-synaptic disinhibition and the intrinsic electrophysiological properties of glutamatergic neurones both play important roles in the generation of the expiratory oscillations in the pFL region during hypercapnia in rats.
Collapse
Affiliation(s)
- Karolyne S Magalhães
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Melina P da Silva
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - André S Mecawi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Julian F R Paton
- Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Park Road, Grafton, Auckland, New Zealand
| | - Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Davi J A Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
44
|
Díaz-Jara E, Díaz HS, Rios-Gallardo A, Ortolani D, Andrade DC, Toledo C, Pereyra KV, Schwarz K, Ramirez G, Ortiz FC, Andía ME, Del Rio R. Exercise training reduces brainstem oxidative stress and restores normal breathing function in heart failure. Free Radic Biol Med 2021; 172:470-481. [PMID: 34216779 DOI: 10.1016/j.freeradbiomed.2021.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 10/21/2022]
Abstract
Enhanced central chemoreflex drive and irregular breathing are both hallmarks in heart failure (HF) and closely related to disease progression. Central chemoreceptor neurons located within the retrotrapezoid nucleus (RTN) are known to play a role in breathing alterations in HF. It has been shown that exercise (EX) effectively reduced reactive oxygen species (ROS) in HF rats. However, the link between EX and ROS, particularly at the RTN, with breathing alterations in HF has not been previously addressed. Accordingly, we aimed to determine: i) ROS levels in the RTN in HF and its association with chemoreflex drive, ii) whether EX improves chemoreflex/breathing function by reducing ROS levels, and iii) determine molecular alterations associated with ROS generation within the RTN of HF rats and study EX effects on these pathways. Adult male Sprague-Dawley rats were allocated into 3 experimental groups: Sham (n = 5), volume overloaded HF (n = 6) and HF (n = 8) rats that underwent EX training for 6 weeks (60 min/day, 25 m/min, 10% inclination). At 8 weeks post-HF induction, breathing patterns and chemoreflex function were analyzed by unrestrained plethysmography. ROS levels and anti/pro-oxidant enzymes gene expression were analyzed in the RTN. Our results showed that HF rats have high ROS levels in the RTN which were closely linked to the enhanced central chemoreflex and breathing disorders. Also, HF rats displayed decreased expression of antioxidant genes in the RTN compared with control rats. EX training increases antioxidant defense in the RTN, reduces ROS formation and restores normal central chemoreflex drive and breathing regularity in HF rats. This study provides evidence for a role of ROS in central chemoreception in the setting of HF and support the use of EX to reduce ROS in the brainstem of HF animals and reveal its potential as an effective mean to normalize chemoreflex and breathing function in HF.
Collapse
Affiliation(s)
- Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Angélica Rios-Gallardo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 621-0427, Punta Arenas, Chile.
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Fisiología y Medicina de Altura, Facultad de Ciencias de la Salud, Universidad de Antofagasta, 1270300, Antofagasta, Chile.
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 621-0427, Punta Arenas, Chile.
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Karla Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Gigliola Ramirez
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fernando C Ortiz
- Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile.
| | - Marcelo E Andía
- Radiology Department & ANID - Millennium Nucleus for Cardiovascular Magnetic Resonance, 8331150, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, 621-0427, Punta Arenas, Chile; Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| |
Collapse
|
45
|
Getsy PM, Sundararajan S, Lewis SJ. Carotid sinus nerve transection abolishes the facilitation of breathing that occurs upon cessation of a hypercapnic gas challenge in male mice. J Appl Physiol (1985) 2021; 131:821-835. [PMID: 34236243 DOI: 10.1152/japplphysiol.01031.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Arterial pCO2 elevations increase minute ventilation via activation of chemosensors within the carotid body (CB) and brainstem. Although the roles of CB chemoafferents in the hypercapnic (HC) ventilatory response have been investigated, there are no studies reporting the role of these chemoafferents in the ventilatory responses to a HC challenge or the responses that occur upon return to room air, in freely moving mice. This study found that an HC challenge (5% CO2, 21% O2, 74% N2 for 15 min) elicited an array of responses, including increases in frequency of breathing (accompanied by decreases in inspiratory and expiratory times), and increases in tidal volume, minute ventilation, peak inspiratory and expiratory flows, and inspiratory and expiratory drives in sham-operated (SHAM) adult male C57BL6 mice, and that return to room air elicited a brief excitatory phase followed by gradual recovery of all parameters toward baseline values over a 15-min period. The array of ventilatory responses to the HC challenge in mice with bilateral carotid sinus nerve transection (CSNX) performed 7 days previously occurred more slowly but reached similar maxima as SHAM mice. A major finding was responses upon return to room air were dramatically lower in CSNX mice than SHAM mice, and the parameters returned to baseline values within 1-2 min in CSNX mice, whereas it took much longer in SHAM mice. These findings are the first evidence that CB chemoafferents play a key role in initiating the ventilatory responses to HC challenge in C57BL6 mice and are essential for the expression of post-HC ventilatory responses.NEW & NOTEWORTHY This study presents the first evidence that carotid body chemoafferents play a key role in initiating the ventilatory responses, such as increases in frequency of breathing, tidal volume, and minute ventilation that occur in response to a hypercapnic gas challenge in freely moving C57BL6 mice. Our study also demonstrates for the first time that these chemoafferents are essential for the expression of the ventilatory responses that occur upon return to room air in these mice.
Collapse
Affiliation(s)
- Paulina M Getsy
- Department of Pediatrics, Case Western University, Cleveland, Ohio
| | - Sripriya Sundararajan
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Stephen J Lewis
- Department of Pediatrics, Case Western University, Cleveland, Ohio.,Department of Pharmacology, Case Western University, Cleveland, Ohio
| |
Collapse
|
46
|
Chang DF, Gilliam EA, Nucho LMA, Garcia J, Shevchenko Y, Zuber SM, Squillaro AI, Maselli KM, Huang S, Spence JR, Grikscheit TC. NH 2-terminal deletion of specific phosphorylation sites on PHOX2B disrupts the formation of enteric neurons in vivo. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1054-G1066. [PMID: 33881351 DOI: 10.1152/ajpgi.00073.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mutations in the paired-like homeobox 2 b (PHOX2B) gene are associated with congenital central hypoventilation syndrome (CCHS), which is a rare condition in which both autonomic dysregulation with hypoventilation and an enteric neuropathy may occur. The majority of patients with CCHS have a polyalanine repeat mutation (PARM) in PHOX2B, but a minority of patients have nonpolyalanine repeat mutations (NPARMs), some of which have been localized to exon 1. A PHOX2B-Y14X nonsense mutation previously generated in a human pluripotent stem cell (hPSC) line results in an NH2-terminus truncated product missing the first 17 or 20 amino acids, possibly due to translational reinitiation at an alternate ATG start site. This NH2-terminal truncation in the PHOX2B protein results in the loss of two key phosphorylation residues. Though the deletion does not affect the potential for PHOX2BY14X/Y14X mutant hPSC to differentiate into enteric neural crest cells (ENCCs) in culture, it impedes in vivo development of neurons in an in vivo model of human aganglionic small intestine.NEW & NOTEWORTHY A mutation that affects only 17-20 NH2-terminal amino acids in the paired-like homeobox 2 b (PHOX2B) gene hinders the subsequent in vivo establishment of intestinal neuronal cells, but not the in vitro differentiation of these cells.
Collapse
Affiliation(s)
- David F Chang
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Elizabeth A Gilliam
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Laura-Marie A Nucho
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Jazmin Garcia
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Yevheniya Shevchenko
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Samuel M Zuber
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Anthony I Squillaro
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathryn M Maselli
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Sha Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Program of Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tracy C Grikscheit
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.,Division of Pediatric Surgery, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California.,Keck Medical School, University of Southern California, Los Angeles, California
| |
Collapse
|
47
|
Oliveira LM, Baertsch NA, Moreira TS, Ramirez JM, Takakura AC. Unraveling the Mechanisms Underlying Irregularities in Inspiratory Rhythm Generation in a Mouse Model of Parkinson's Disease. J Neurosci 2021; 41:4732-4747. [PMID: 33863785 PMCID: PMC8260248 DOI: 10.1523/jneurosci.2114-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder anatomically characterized by a progressive loss of dopaminergic neurons in the substantia nigra compacta (SNpc). Much less known, yet clinically very important, are the detrimental effects on breathing associated with this disease. Consistent with the human pathophysiology, the 6-hydroxydopamine hydrochloride (6-OHDA) rodent model of PD shows reduced respiratory frequency (fR) and NK1r-immunoreactivity in the pre-Bötzinger complex (preBötC) and PHOX2B+ neurons in the retrotrapezoid nucleus (RTN). To unravel mechanisms that underlie bradypnea in PD, we employed a transgenic approach to label or stimulate specific neuron populations in various respiratory-related brainstem regions. PD mice were characterized by a pronounced decreased number of putatively rhythmically active excitatory neurons in the preBötC and adjacent ventral respiratory column (VRC). Specifically, the number of Dbx1 and Vglut2 neurons was reduced by 47.6% and 17.3%, respectively. By contrast, inhibitory Vgat+ neurons in the VRC, as well as neurons in other respiratory-related brainstem regions, showed relatively minimal or no signs of neuronal loss. Consistent with these anatomic observations, optogenetic experiments identified deficits in respiratory function that were specific to manipulations of excitatory (Dbx1/Vglut2) neurons in the preBötC. We conclude that the decreased number of this critical population of respiratory neurons is an important contributor to the development of irregularities in inspiratory rhythm generation in this mouse model of PD.SIGNIFICANCE STATEMENT We found a decreased number of a specific population of medullary neurons which contributes to breathing abnormalities in a mouse model of Parkinson's disease (PD).
Collapse
Affiliation(s)
- Luiz M Oliveira
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508, Brazil
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
- Department of Pediatrics, University of Washington, Seattle, Washington 98101
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508, Brazil
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
- Department of Pediatrics, University of Washington, Seattle, Washington 98101
| | - Ana C Takakura
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508, Brazil
| |
Collapse
|
48
|
Cleary CM, Milla BM, Kuo FS, James S, Flynn WF, Robson P, Mulkey DK. Somatostatin-expressing parafacial neurons are CO 2/H + sensitive and regulate baseline breathing. eLife 2021; 10:e60317. [PMID: 34013884 PMCID: PMC8169115 DOI: 10.7554/elife.60317] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 05/19/2021] [Indexed: 01/16/2023] Open
Abstract
Glutamatergic neurons in the retrotrapezoid nucleus (RTN) function as respiratory chemoreceptors by regulating breathing in response to tissue CO2/H+. The RTN and greater parafacial region may also function as a chemosensing network composed of CO2/H+-sensitive excitatory and inhibitory synaptic interactions. In the context of disease, we showed that loss of inhibitory neural activity in a mouse model of Dravet syndrome disinhibited RTN chemoreceptors and destabilized breathing (Kuo et al., 2019). Despite this, contributions of parafacial inhibitory neurons to control of breathing are unknown, and synaptic properties of RTN neurons have not been characterized. Here, we show the parafacial region contains a limited diversity of inhibitory neurons including somatostatin (Sst)-, parvalbumin (Pvalb)-, and cholecystokinin (Cck)-expressing neurons. Of these, Sst-expressing interneurons appear uniquely inhibited by CO2/H+. We also show RTN chemoreceptors receive inhibitory input that is withdrawn in a CO2/H+-dependent manner, and chemogenetic suppression of Sst+ parafacial neurons, but not Pvalb+ or Cck+ neurons, increases baseline breathing. These results suggest Sst-expressing parafacial neurons contribute to RTN chemoreception and respiratory activity.
Collapse
Affiliation(s)
- Colin M Cleary
- Department of Physiology and Neurobiology, University of ConnecticutStorrsUnited States
| | - Brenda M Milla
- Department of Physiology and Neurobiology, University of ConnecticutStorrsUnited States
| | - Fu-Shan Kuo
- Department of Physiology and Neurobiology, University of ConnecticutStorrsUnited States
| | - Shaun James
- Department of Physiology and Neurobiology, University of ConnecticutStorrsUnited States
| | - William F Flynn
- The Jackson Laboratory for Genomic MedicineFarmingtonUnited States
| | - Paul Robson
- The Jackson Laboratory for Genomic MedicineFarmingtonUnited States
- Institute for Systems Genomics, University of ConnecticutFarmingtonUnited States
| | - Daniel K Mulkey
- Department of Physiology and Neurobiology, University of ConnecticutStorrsUnited States
| |
Collapse
|
49
|
Onimaru H, Yazawa I, Takeda K, Fukushi I, Okada Y. Calcium Imaging Analysis of Cellular Responses to Hypercapnia and Hypoxia in the NTS of Newborn Rat Brainstem Preparation. Front Physiol 2021; 12:645904. [PMID: 33841182 PMCID: PMC8027497 DOI: 10.3389/fphys.2021.645904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/08/2021] [Indexed: 01/13/2023] Open
Abstract
It is supposed that the nucleus of the solitary tract (NTS) in the dorsal medulla includes gas sensor cells responsive to hypercapnia or hypoxia in the central nervous system. In the present study, we analyzed cellular responses to hypercapnia and hypoxia in the NTS region of newborn rat in vitro preparation. The brainstem and spinal cord were isolated from newborn rat (P0-P4) and were transversely cut at the level of the rostral area postrema. To detect cellular responses, calcium indicator Oregon Green was pressure-injected into the NTS just beneath the cut surface of either the caudal or rostral block of the medulla, and the preparation was superfused with artificial cerebrospinal fluid (25–26°C). We examined cellular responses initially to hypercapnic stimulation (to 8% CO2 from 2% CO2) and then to hypoxic stimulation (to 0% O2 from 95% O2 at 5% CO2). We tested these responses in standard solution and in two different synapse blockade solutions: (1) cocktail blockers solution including bicuculline, strychnine, NBQX and MK-801 or (2) TTX solution. At the end of the experiments, the superfusate potassium concentration was lowered to 0.2 from 3 mM to classify recorded cells into neurons and astrocytes. Excitation of cells was detected as changes of fluorescence intensity with a confocal calcium imaging system. In the synaptic blockade solutions (cocktail or TTX solution), 7.6 and 8% of the NTS cells responded to hypercapnic and hypoxic stimulation, respectively, and approximately 2% of them responded to both stimulations. Some of these cells responded to low K+, and they were classified into astrocytes comprising 43% hypercapnia-sensitive cells, 56% hypoxia-sensitive cells and 54% of both stimulation-sensitive cells. Of note, 49% of the putative astrocytes identified by low K+ stimulation were sensitive to hypercapnia, hypoxia or both. In the presence of a glia preferential blocker, 5 mM fluoroacetate (plus 0.5 μM TTX), the percentage of hypoxia-sensitive cells was significantly reduced compared to those of all other conditions. This is the first study to reveal that the NTS includes hypercapnia and hypoxia dual-sensitive cells. These results suggest that astrocytes in the NTS region could act as a central gas sensor.
Collapse
Affiliation(s)
- Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Itaru Yazawa
- Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Kotaro Takeda
- Faculty of Rehabilitation, School of Healthcare, Fujita Health University, Toyoake, Japan
| | - Isato Fukushi
- Faculty of Health Sciences, Uekusa Gakuen University, Chiba, Japan.,Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan
| | - Yasumasa Okada
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan
| |
Collapse
|
50
|
Abbott SBG, Souza GMPR. Chemoreceptor mechanisms regulating CO 2 -induced arousal from sleep. J Physiol 2021; 599:2559-2571. [PMID: 33759184 DOI: 10.1113/jp281305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/16/2021] [Indexed: 12/24/2022] Open
Abstract
Arousal from sleep in response to CO2 is a life-preserving reflex that enhances ventilatory drive and facilitates behavioural adaptations to restore eupnoeic breathing. Recurrent activation of the CO2 -arousal reflex is associated with sleep disruption in obstructive sleep apnoea. In this review we examine the role of chemoreceptors in the carotid bodies, the retrotrapezoid nucleus and serotonergic neurons in the dorsal raphe in the CO2 -arousal reflex. We also provide an overview of the supra-medullary structures that mediate CO2 -induced arousal. We propose a framework for the CO2 -arousal reflex in which the activity of the chemoreceptors converges in the parabrachial nucleus to trigger cortical arousal.
Collapse
Affiliation(s)
- Stephen B G Abbott
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 29903, USA
| | - George M P R Souza
- Department of Pharmacology, University of Virginia, Charlottesville, VA, 29903, USA
| |
Collapse
|