1
|
Khalilpour J, Alipour MR, Shahabi P. Chronic sustained hypoxia alters the pattern of diaphragm electrical activity in anaesthetized rats. Exp Physiol 2025; 110:599-609. [PMID: 39991980 PMCID: PMC11963899 DOI: 10.1113/ep092211] [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: 08/01/2024] [Accepted: 12/12/2024] [Indexed: 02/25/2025]
Abstract
Chronic sustained hypoxia (CSH) is known to induce functional and structural changes in the respiratory system. The diaphragm, as the main inspiratory muscle of mammals, is particularly important in the neuromotor regulation of respiration. Diaphragm electromyography (dEMG) records the sum of motor unit action potentials (MUAP) and provides information regarding motor unit recruitment and frequency coding during muscle contraction. We aimed to assess changes in dEMG activity following CSH. Herein, eight male Wistar rats (2-3 months) were subjected to CSH (10 ± 0.5% O2) for 10 successive days. In vivo dEMG recording was employed to assess changes in the diaphragm electrical activity. Filtered and rectified dEMGs were used for further analyses. Findings showed that CSH for 10 consecutive days significantly changed the pattern of dEMG signals. The slope of the rising phase of RMS-enveloped dEMG bursts was much steeper in CSH rats compared to normoxic control rats (rise time: 373 vs. 286 ms; P = 0.005). Burst frequency significantly decreased following CSH (59 vs. 42 bursts/min; P = 0.0001), which was associated with a significant increase in burst amplitude (P = 0.039) and inter-burst duration (0.65 vs. 0.88 s; P = 0.041). Power spectral density analyses showed that the mean frequency (293 vs. 266 Hz; P = 0.033) and high-frequency to low-frequency power ratio (P = 0.009) of dEMG signals significantly declined in CSH rats. Notably, the regularity of frequency and amplitude of dEMG signals did not change significantly following CSH.
Collapse
Affiliation(s)
- Jamal Khalilpour
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- National Medical Emergency OrganizationMinistry of Health & Medical EducationTehranIran
| | | | - Parviz Shahabi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
2
|
Rana S, Fusco AF, Witkin JM, Radin DP, Cerne R, Lippa A, Fuller DD. Pharmacological modulation of respiratory control: Ampakines as a therapeutic strategy. Pharmacol Ther 2025; 265:108744. [PMID: 39521442 PMCID: PMC11849399 DOI: 10.1016/j.pharmthera.2024.108744] [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: 08/04/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Ampakines are a class of compounds that are positive allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and enhance glutamatergic neurotransmission. Glutamatergic synaptic transmission and AMPA receptor activation are fundamentally important to the genesis and propagation of the neural impulses driving breathing, including respiratory motoneuron depolarization. Ampakines therefore have the potential to modulate the neural control of breathing. In this paper, we describe the influence of ampakines on respiratory motor output in health and disease. We dissect the molecular mechanisms underlying ampakine action, delineate the diverse targets of ampakines along the respiratory neuraxis, survey the spectrum of respiratory disorders in which ampakines have been tested, and culminate with an examination of how ampakines modulate respiratory function after spinal cord injury. Collectively, the studies reviewed here indicate that ampakines may be a useful adjunctive strategy to pair with conventional respiratory rehabilitation approaches in conditions with impaired neural activation of the respiratory muscles.
Collapse
Affiliation(s)
- Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America.
| | - Anna F Fusco
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America
| | - Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, United States of America; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, IN, United States of America; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - Daniel P Radin
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, St. Vincent's Hospital, Indianapolis, IN, United States of America; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America; Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, Ljubljana, Slovenia
| | - Arnold Lippa
- RespireRx Pharmaceuticals Inc, Glen Rock, NJ, United States of America
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, United States of America; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, United States of America; Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, United States of America
| |
Collapse
|
3
|
Lino-Alvarado A, Maia OAC, Oliveira MA, Takakura AC, Tavares-Lima W, Moriya HT, Moreira TS. Central and peripheral mechanisms underlying respiratory deficits in a mouse model of accelerated senescence. Pflugers Arch 2024; 476:1665-1676. [PMID: 39150501 DOI: 10.1007/s00424-024-03006-y] [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: 04/12/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Aging invariably decreases sensory and motor stimuli and affects several neuronal systems and their connectivity to key brain regions, including those involved in breathing. Nevertheless, further investigation is needed to fully comprehend the link between senescence and respiratory function. Here, we investigate whether a mouse model of accelerated senescence could develop central and peripheral respiratory abnormalities. Adult male Senescence Accelerated Mouse Prone 8 (SAMP8) and the control SAMR1 mice (10 months old) were used. Ventilatory parameters were assessed by whole-body plethysmography, and measurements of respiratory input impedance were performed. SAMP8 mice exhibited a reduction in the density of neurokinin-1 receptor immunoreactivity in the entire ventral respiratory column. Physiological experiments showed that SAMP8 mice exhibited a decreased tachypneic response to hypoxia (FiO2 = 0.08; 10 min) or hypercapnia (FiCO2 = 0.07; 10 min). Additionally, the ventilatory response to hypercapnia increased further due to higher tidal volume. Measurements of respiratory mechanics in SAMP8 mice showed decreased static compliance (Cstat), inspiratory capacity (IC), resistance (Rn), and elastance (H) at different ages (3, 6, and 10 months old). SAMP8 mice also have a decrease in contractile response to methacholine compared to SAMR1. In conclusion, our findings indicate that SAMP8 mice display a loss of the NK1-expressing neurons in the respiratory brainstem centers, along with impairments in both central and peripheral respiratory mechanisms. These observations suggest a potential impact on breathing in a senescence animal model.
Collapse
Affiliation(s)
| | - Octavio A C Maia
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria Aparecida Oliveira
- Deptartment of Pharmacology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Ana C Takakura
- Deptartment of Pharmacology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Wothan Tavares-Lima
- Deptartment of Pharmacology, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Henrique T Moriya
- Biomedical Engineering Laboratory, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
4
|
Khalilpour J, Soltani Zangbar H, Alipour MR, Shahabi P. The hypoxic respiratory response of the pre-Bötzinger complex. Heliyon 2024; 10:e34491. [PMID: 39114066 PMCID: PMC11305331 DOI: 10.1016/j.heliyon.2024.e34491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Since the discovery of the pre-Bötzinger Complex (preBötC) as a crucial region for generating the main respiratory rhythm, our understanding of its cellular and molecular aspects has rapidly increased within the last few decades. It is now apparent that preBötC is a highly flexible neuronal network that reconfigures state-dependently to produce the most appropriate respiratory output in response to various metabolic challenges, such as hypoxia. However, the responses of the preBötC to hypoxic conditions can be varied based on the intensity, pattern, and duration of the hypoxic challenge. This review discusses the preBötC response to hypoxic challenges at the cellular and network level. Particularly, the involvement of preBötC in the classical biphasic response of the respiratory network to acute hypoxia is illuminated. Furthermore, the article discusses the functional and structural changes of preBötC neurons following intermittent and sustained hypoxic challenges. Accumulating evidence shows that the preBötC neural circuits undergo substantial changes following hypoxia and contribute to several types of the respiratory system's hypoxic ventilatory responses.
Collapse
Affiliation(s)
- Jamal Khalilpour
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Kobayashi R, Negoro H. Acute effects of the 4-4-8 breathing technique on arterial stiffness in healthy young men. Cardiol J 2024; 31:418-426. [PMID: 38348911 PMCID: PMC11229811 DOI: 10.5603/cj.96299] [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: 07/01/2023] [Revised: 12/01/2023] [Accepted: 12/23/2023] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Increased arterial stiffness is a risk factor for cardiovascular disease. Slow, deep breathing decreases blood pressure related to arterial stiffness. The objective of the present study was to determine the acute effects of a single session of slow breathing on arterial stiffness, blood pressure, and cardiac autonomic function. METHODS Fifteen healthy men (20 ± 0 years) were administered (a) a slow breathing condition (12 consecutive breaths of 4 s of inhalation, 4 s of pause, and 8 s of exhalation through the nose, approximately 5 min per breath) and (b) a control, two-condition crossover design. Carotid-femoral artery pulse wave velocity (cfPWV), brachial-ankle PWV (baPWV), brachial blood pressure, high frequency (HF) and low frequency (LF) were measured at baseline, 30 min, 60 min and 24 h after respiratory control. RESULTS Brachial-ankle PWV and brachial systolic pressure on the 4-4-8 breathing trial decreased after 30 min of respiratory control compared to baseline (p < 0.05), but did not change on the CON trial. Carotid-femoral PWV on both trials was unchanged; HF on the 4-4-8 breathing trial increased (p < 0.05) and LF decreased (p < 0.05) after 30 min of respiratory control compared to baseline, but was unchanged on the CON trial. CONCLUSIONS These results suggest that slow breathing techniques may be effective in modulating autonomic function and improving arterial stiffness in healthy young adults.
Collapse
Affiliation(s)
- Ryota Kobayashi
- Department of Natural and Environmental Science, Teikyo University of Science, Tokyo, Japan.
| | - Hideyuki Negoro
- Department of Medicine, Nara Medical University, Nara, Japan
| |
Collapse
|
6
|
Severs LJ, Bush NE, Quina LA, Hidalgo-Andrade S, Burgraff NJ, Dashevskiy T, Shih AY, Baertsch NA, Ramirez JM. Purinergic signaling mediates neuroglial interactions to modulate sighs. Nat Commun 2023; 14:5300. [PMID: 37652903 PMCID: PMC10471608 DOI: 10.1038/s41467-023-40812-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 08/10/2023] [Indexed: 09/02/2023] Open
Abstract
Sighs prevent the collapse of alveoli in the lungs, initiate arousal under hypoxic conditions, and are an expression of sadness and relief. Sighs are periodically superimposed on normal breaths, known as eupnea. Implicated in the generation of these rhythmic behaviors is the preBötzinger complex (preBötC). Our experimental evidence suggests that purinergic signaling is necessary to generate spontaneous and hypoxia-induced sighs in a mouse model. Our results demonstrate that driving calcium increases in astrocytes through pharmacological methods robustly increases sigh, but not eupnea, frequency. Calcium imaging of preBötC slices corroborates this finding with an increase in astrocytic calcium upon application of sigh modulators, increasing intracellular calcium through g-protein signaling. Moreover, photo-activation of preBötC astrocytes is sufficient to elicit sigh activity, and this response is blocked with purinergic antagonists. We conclude that sighs are modulated through neuron-glia coupling in the preBötC network, where the distinct modulatory responses of neurons and glia allow for both rhythms to be independently regulated.
Collapse
Affiliation(s)
- Liza J Severs
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA.
| | - Nicholas E Bush
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Lely A Quina
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Skyler Hidalgo-Andrade
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Nicholas J Burgraff
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Tatiana Dashevskiy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98195, USA.
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA.
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
7
|
Tacke C, Bischoff AM, Harb A, Vafadari B, Hülsmann S. Fiber optical imaging of astroglial calcium signaling in the respiratory network in the working heart brainstem preparation. Front Physiol 2023; 14:1237376. [PMID: 37693007 PMCID: PMC10484401 DOI: 10.3389/fphys.2023.1237376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
The neuronal activity in the respiratory network strongly depends on a variety of different neuromodulators. Given the essential role of astrocytes in stabilizing respiratory network activity generated by neurons in the preBötzinger complex (preBötC), our aim was to investigate astrocytic calcium signaling in the working heart brainstem preparation using fiber-optical imaging. By using transgenic mice that express GCaMP6s specifically in astrocytes, we successfully recorded astrocytic calcium signals in response to norepinephrine from individual astrocytes.
Collapse
Affiliation(s)
| | | | | | | | - Swen Hülsmann
- Department of Anesthesiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
8
|
Bolte KN, Wealing JC, Revill AL. Arginine vasopressin potentiates inspiratory bursting in hypoglossal motoneurons of neonatal mice. Respir Physiol Neurobiol 2023; 314:104087. [PMID: 37269889 PMCID: PMC10443434 DOI: 10.1016/j.resp.2023.104087] [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: 02/02/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/05/2023]
Abstract
Vasopressin (AVP) acts as a neurotransmitter and its activity can potentiate respiratory activity. Hypoglossal (XII) motoneurons that innervate the tongue express V1a vasopressin receptors, which are excitatory. Therefore, we hypothesized that V1a receptor activation at XII motoneurons would potentiate inspiratory bursting. We developed this study to determine whether AVP can potentiate inspiratory bursting in rhythmic medullary slice preparations in neonatal (postnatal, P0-5) mice. Bath or local application of AVP potentiated inspiratory bursting compared to baseline XII inspiratory burst amplitude. Antagonizing V1a receptors revealed significant attenuation of the AVP-mediated potentiation of inspiratory bursting, while antagonism of oxytocin receptors (at which AVP has similar binding affinity) revealed a trend to attenuate AVP-mediated potentiation of inspiratory bursting. Finally, we discovered that the AVP-mediated potentiation of inspiratory bursting increases significantly with postnatal maturation from P0-5. Overall, these data support that AVP potentiates inspiratory bursting directly at XII motoneurons.
Collapse
Affiliation(s)
- K N Bolte
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, United States
| | - J C Wealing
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, United States
| | - A L Revill
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, United States; Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ, United States.
| |
Collapse
|
9
|
Bauer J, Devinsky O, Rothermel M, Koch H. Autonomic dysfunction in epilepsy mouse models with implications for SUDEP research. Front Neurol 2023; 13:1040648. [PMID: 36686527 PMCID: PMC9853197 DOI: 10.3389/fneur.2022.1040648] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
Epilepsy has a high prevalence and can severely impair quality of life and increase the risk of premature death. Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death in drug-resistant epilepsy and most often results from respiratory and cardiac impairments due to brainstem dysfunction. Epileptic activity can spread widely, influencing neuronal activity in regions outside the epileptic network. The brainstem controls cardiorespiratory activity and arousal and reciprocally connects to cortical, diencephalic, and spinal cord areas. Epileptic activity can propagate trans-synaptically or via spreading depression (SD) to alter brainstem functions and cause cardiorespiratory dysfunction. The mechanisms by which seizures propagate to or otherwise impair brainstem function and trigger the cascading effects that cause SUDEP are poorly understood. We review insights from mouse models combined with new techniques to understand the pathophysiology of epilepsy and SUDEP. These techniques include in vivo, ex vivo, invasive and non-invasive methods in anesthetized and awake mice. Optogenetics combined with electrophysiological and optical manipulation and recording methods offer unique opportunities to study neuronal mechanisms under normal conditions, during and after non-fatal seizures, and in SUDEP. These combined approaches can advance our understanding of brainstem pathophysiology associated with seizures and SUDEP and may suggest strategies to prevent SUDEP.
Collapse
Affiliation(s)
- Jennifer Bauer
- Department of Epileptology and Neurology, RWTH Aachen University, Aachen, Germany,Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Orrin Devinsky
- Departments of Neurology, Neurosurgery and Psychiatry, NYU Langone School of Medicine, New York, NY, United States
| | - Markus Rothermel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Henner Koch
- Department of Epileptology and Neurology, RWTH Aachen University, Aachen, Germany,*Correspondence: Henner Koch ✉
| |
Collapse
|
10
|
Mediation of Sinusoidal Network Oscillations in the Locus Coeruleus of Newborn Rat Slices by Pharmacologically Distinct AMPA and KA Receptors. Brain Sci 2022; 12:brainsci12070945. [PMID: 35884751 PMCID: PMC9321180 DOI: 10.3390/brainsci12070945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 02/04/2023] Open
Abstract
Brain control by locus coeruleus (LC) neurons involves afferent glutamate (Glu) inputs. In newborns, LC Glu receptors and responses may be sparse due to immaturity of the brain circuits providing such input. However, we reported, using newborn rat brain slices, that Glu and its ionotropic receptor (iGluR) agonist NMDA transform spontaneous local field potential (LFP) rhythm. Here, we studied whether α-amino-3-hydroxy-5-methyl-4-isoxazole propionic-acid (AMPA) and kainate (KA) iGluR subtypes also transform the LFP pattern. AMPA (0.25–0.5 µM) and KA (0.5–2.5 µM) merged ~0.2 s-lasting bell-shaped LFP events occurring at ~1 Hz into ~40% shorter and ~4-fold faster spindle-shaped and more regular sinusoidal oscillations. The AMPA/KA effects were associated with a 3.1/4.3-fold accelerated phase-locked single neuron spiking due to 4.0/4.2 mV depolarization while spike jitter decreased to 64/42% of the control, respectively. Raising extracellular K+ from 3 to 9 mM increased the LFP rate 1.4-fold or elicited slower multipeak events. A blockade of Cl−-mediated inhibition with gabazine (5 μM) plus strychnine (10 μM) affected neither the control rhythm nor AMPA/KA oscillations. GYKI-53655 (25 μM) blocked AMPA (but not KA) oscillations whereas UBP-302 (25 μM) blocked KA (but not AMPA) oscillations. Our findings revealed that AMPA and KA evoke a similar novel neural network discharge pattern transformation type by acting on pharmacologically distinct AMPAR and KA receptors. This shows that already the neonatal LC can generate oscillatory network behaviors that may be important, for example, for responses to opioids.
Collapse
|
11
|
Oliveira LM, Fernandes-Junior SA, Cabral LMC, Miranda NCS, Czeisler CM, Otero JJ, Moreira TS, Takakura AC. Regulation of blood vessels by ATP in the ventral medullary surface in a rat model of Parkinson's disease. Brain Res Bull 2022; 187:138-154. [PMID: 35777704 DOI: 10.1016/j.brainresbull.2022.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/26/2022] [Accepted: 06/25/2022] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) patients often experience impairment of autonomic and respiratory functions. These include conditions such as orthostatic hypotension and sleep apnea, which are highly correlated with dysfunctional central chemoreception. Blood flow is a fundamental determinant of tissue CO2/H+, yet the extent to which blood flow regulation within chemoreceptor regions contributes to respiratory behavior during neurological disease remains unknown. Here, we tested the hypothesis that 6-hydroxydopamine injection to inducing a known model of PD results in dysfunctional vascular homeostasis, biochemical dysregulation, and glial morphology of the ventral medullary surface (VMS). We show that hypercapnia (FiCO2 = 10%) induced elevated VMS pial vessel constriction in PD animals through a P2-receptor dependent mechanism. Similarly, we found a greater CO2-induced vascular constriction after ARL67156 (an ectonucleotidase inhibitor) in control and PD-induced animals. In addition, we also report that weighted gene correlational network analysis of the proteomic data showed a protein expression module differentially represented between both groups. This module showed that gene ontology enrichment for components of the ATP machinery were reduced in our PD-model compared to control animals. Altogether, our data indicate that dysfunction in purinergic signaling, potentially through altered ATP bioavailability in the VMS region, may compromise the RTN neuroglial vascular unit in a PD animal model.
Collapse
Affiliation(s)
- Luiz M Oliveira
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | - Silvio A Fernandes-Junior
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil; The Ohio State University College of Medicine, Department of Pathology, USA
| | - Laís M C Cabral
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | - Nicole C S Miranda
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | | | - José J Otero
- The Ohio State University College of Medicine, Department of Pathology, USA
| | - Thiago S Moreira
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil
| | - Ana C Takakura
- Departamento de Farmacologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
12
|
Camacho-Hernández P, Lorea-Hernández JJ, Pinedo-Vargas L, Peña-Ortega F. Perinatal inflammation and gestational intermittent hypoxia disturbs respiratory rhythm generation and long-term facilitation in vitro: partial protection by acute minocycline. Respir Physiol Neurobiol 2021; 297:103829. [PMID: 34921999 DOI: 10.1016/j.resp.2021.103829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 10/31/2021] [Accepted: 12/13/2021] [Indexed: 01/04/2023]
Abstract
Perinatal inflammation triggers breathing disturbances early in life and affects the respiratory adaptations to challenging conditions, including the generation of amplitude long-term facilitation (LTF) by acute intermittent hypoxia (AIH). Some of these effects can be avoided by anti-inflammatory treatments like minocycline. Since little is known about the effects of perinatal inflammation on the inspiratory rhythm generator, located in the preBötzinger complex (preBötC), we tested the impact of acute lipopolysaccharide (LPS) systemic administration (sLPS), as well as gestational LPS (gLPS) and gestational chronic IH (gCIH), on respiratory rhythm generation and its long-term response to AIH in a brainstem slice preparation from neonatal mice. We also evaluated whether acute minocycline administration could influence these effects. We found that perinatal inflammation induced by sLPS or gLPS, as well as gCIH, modulate the frequency, signal-to-noise ratio and/or amplitude (and their regularity) of the respiratory rhythm recorded from the preBötC in the brainstem slice. Moreover, all these perinatal conditions inhibited frequency LTF and amplitude long-term depression (LTD); gCIH even induced frequency LTD of the respiratory rhythm after AIH. Some of the alterations were not observed in slices pre-treated in vitro with minocycline, when compared with slices obtained from naïve pups, suggesting that ongoing inflammatory conditions affect respiratory rhythm generation and its plasticity. Thus, it is likely that alterations in the inspiratory rhythm generator and its adaptive responses could contribute to the respiratory disturbances observed in neonates that suffered from perinatal inflammatory challenges.
Collapse
Affiliation(s)
- Polet Camacho-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Jonathan Julio Lorea-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Laura Pinedo-Vargas
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico.
| |
Collapse
|
13
|
Sharma HS, Lafuente JV, Feng L, Muresanu DF, Menon PK, Castellani RJ, Nozari A, Sahib S, Tian ZR, Buzoianu AD, Sjöquist PO, Patnaik R, Wiklund L, Sharma A. Methamphetamine exacerbates pathophysiology of traumatic brain injury at high altitude. Neuroprotective effects of nanodelivery of a potent antioxidant compound H-290/51. PROGRESS IN BRAIN RESEARCH 2021; 266:123-193. [PMID: 34689858 DOI: 10.1016/bs.pbr.2021.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Military personnel are often exposed to high altitude (HA, ca. 4500-5000m) for combat operations associated with neurological dysfunctions. HA is a severe stressful situation and people frequently use methamphetamine (METH) or other psychostimulants to cope stress. Since military personnel are prone to different kinds of traumatic brain injury (TBI), in this review we discuss possible effects of METH on concussive head injury (CHI) at HA based on our own observations. METH exposure at HA exacerbates pathophysiology of CHI as compared to normobaric laboratory environment comparable to sea level. Increased blood-brain barrier (BBB) breakdown, edema formation and reductions in the cerebral blood flow (CBF) following CHI were exacerbated by METH intoxication at HA. Damage to cerebral microvasculature and expression of beta catenin was also exacerbated following CHI in METH treated group at HA. TiO2-nanowired delivery of H-290/51 (150mg/kg, i.p.), a potent chain-breaking antioxidant significantly enhanced CBF and reduced BBB breakdown, edema formation, beta catenin expression and brain pathology in METH exposed rats after CHI at HA. These observations are the first to point out that METH exposure in CHI exacerbated brain pathology at HA and this appears to be related with greater production of oxidative stress induced brain pathology, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Per-Ove Sjöquist
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Buchholz KJ, Burgraff NJ, Neumueller SE, Hodges MR, Pan LG, Forster HV. Physiological and neurochemical adaptations following abrupt termination of chronic hypercapnia in goats. J Appl Physiol (1985) 2021; 130:1259-1273. [PMID: 33539265 PMCID: PMC8262788 DOI: 10.1152/japplphysiol.00909.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic hypercapnia (CH) is a hallmark of respiratory diseases such as chronic obstructive pulmonary disease. In such patients, mechanical ventilation is often used to restore normal blood-gas homeostasis. However, little is known regarding physiological changes and neuroplasticity within physiological control networks after termination of CH. Utilizing our goat model of increased inspired CO2-induced CH, we determined whether termination of CH elicits time-dependent physiological and neurochemical changes within brain stem sites of physiological control. Thirty days of CH increased [Formula: see text] (+15 mmHg) and steady-state ventilation (SS V̇i; 283% of control). Within 24 h after terminating CH, SS V̇i, blood gases, arterial [H+], and most physiological measurements returned to control. However, the acute ventilatory chemoreflex (ΔV̇i/Δ[H+]) was greater than control, and measured SS V̇i exceeded ventilation predicted by arterial [H+] and ΔV̇i/Δ[H+]. Potentially contributing to these differences were increased excitatory neuromodulators serotonin and norepinephrine in the nucleus tractus solitarius, which contrasts with minimal changes observed at 24 h and 30 days of hypercapnia. Similarly, there were minimal changes found in markers of neuroinflammation and glutamate receptor-dependent neuroplasticity upon termination of CH, which were previously increased following 24 h of hypercapnia. Thus, following termination of CH: 1) ventilatory, renal, and other physiological functions rapidly return to control; 2) neuroplasticity within the ventilatory control network may contribute to the difference between measured vs. predicted ventilation and the elevation in the acute ventilatory [H+] chemoreflex; and 3) neuroplasticity is fundamentally distinct from acclimatization to CH.NEW & NOTEWORTHY In healthy adult goats, steady-state ventilation and most physiological measures return to control within 24 h after termination of chronic hypercapnia (CH). However, the acute [H+] chemoreflex is increased, and measured ventilation exceeds predicted ventilation. At 24 h of recovery, excitatory neuromodulators are above control, but other measured markers of neuroplasticity are unchanged from control. Our data suggest that CH elicits persistent physiological and neurochemical changes for up to 24 h after termination of CH.
Collapse
Affiliation(s)
- Kirstyn J. Buchholz
- 1Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nicholas J. Burgraff
- 5Center for Integrated Brain Research, Seattle Children’s Research Institute, Seattle, Washington
| | | | - Matthew Robert Hodges
- 1Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin,3Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lawrence G. Pan
- 2Department of Physical Therapy, Marquette University, Milwaukee, Wisconsin
| | - Hubert V. Forster
- 1Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin,3Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin,4Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
15
|
Michel-Flutot P, Mansart A, Deramaudt TB, Jesus I, Lee KZ, Bonay M, Vinit S. Permanent diaphragmatic deficits and spontaneous respiratory plasticity in a mouse model of incomplete cervical spinal cord injury. Respir Physiol Neurobiol 2021; 284:103568. [DOI: 10.1016/j.resp.2020.103568] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/21/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022]
|
16
|
Juárez-Vidales JDJ, Pérez-Ortega J, Lorea-Hernández JJ, Méndez-Salcido F, Peña-Ortega F. Configuration and dynamics of dominant inspiratory multineuronal activity patterns during eupnea and gasping generation in vitro. J Neurophysiol 2021; 125:1289-1306. [PMID: 33502956 DOI: 10.1152/jn.00563.2020] [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] [Indexed: 12/14/2022] Open
Abstract
The pre-Bötzinger complex (preBötC), located within the ventral respiratory column, produces inspiratory bursts in varying degrees of synchronization/amplitude. This wide range of population burst patterns reflects the flexibility of the preBötC neurons, which is expressed in variations in the onset/offset times of their activations and their activity during the population bursts, with respiratory neurons exhibiting a large cycle-to-cycle timing jitter both at the population activity onset and at the population activity peak, suggesting that respiratory neurons are stochastically activated before and during the inspiratory bursts. However, it is still unknown whether this stochasticity is maintained while evaluating the coactivity of respiratory neuronal ensembles. Moreover, the preBötC topology also remains unknown. In this study, by simultaneously recording tens of preBötC neurons and using coactivation analysis during the inspiratory periods, we found that the preBötC has a scale-free configuration (mixture of not many highly connected nodes, hubs, with abundant poorly connected elements) exhibiting the rich-club phenomenon (hubs more likely interconnected with each other). PreBötC neurons also produce multineuronal activity patterns (MAPs) that are highly stable and change during the hypoxia-induced reconfiguration. Moreover, preBötC contains a coactivating core network shared by all its MAPs. Finally, we found a distinctive pattern of sequential coactivation of core network neurons at the beginning of the inspiratory periods, indicating that, when evaluated at the multicellular level, the coactivation of respiratory neurons seems not to be stochastic.NEW & NOTEWORTHY By means of multielectrode recordings of preBötC neurons, we evaluated their configuration in normoxia and hypoxia, finding that the preBötC exhibits a scale-free configuration with a rich-club phenomenon. preBötC neurons produce multineuronal activity patterns that are highly stable but change during hypoxia. The preBötC contains a coactivating core network that exhibit a distinctive pattern of coactivation at the beginning of inspirations. These results reveal some network basis of inspiratory rhythm generation and its reconfiguration during hypoxia.
Collapse
Affiliation(s)
- Josué de Jesús Juárez-Vidales
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Mexico
| | - Jesús Pérez-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Mexico
| | - Jonathan Julio Lorea-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Mexico
| | - Felipe Méndez-Salcido
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Mexico
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro, Mexico
| |
Collapse
|
17
|
Shen Y, Ma HX, Lu H, Zhao HT, Sun JL, Cheng Y, Zhang HH. Central deficiency of norepinephrine synthesis and norepinephrinergic neurotransmission contributes to seizure-induced respiratory arrest. Biomed Pharmacother 2021; 133:111024. [PMID: 33232929 DOI: 10.1016/j.biopha.2020.111024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 11/19/2022] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the leading cause of mortality in patients with intractable epilepsy. However, the pathogenesis of SUDEP seems to be poorly understood. Our previous findings showed that the incidence of seizure-induced respiratory arrest (S-IRA) was markedly reduced by atomoxetine in a murine SUDEP model. Because the central norepinephrine α-1 receptor (NEα-1R) plays a vital role in regulating respiratory function, we hypothesized that the suppression of S-IRA by atomoxetine was mediated by NE/NEα-1R interactions that can be reversed by NEα-1R antagonism. We examined whether atomoxetine-mediated suppression of S-IRA evoked by either acoustic stimulation or pentylenetetrazole (PTZ) in DBA/1 mice can be reversed by intraperitoneal (IP) and intracerebroventricular (ICV) administration of prazosin, a selective antagonist of NEα-1R. The content and activity of tyrosine hydroxylase (TH), a rate-limiting enzyme for NE synthesis, in the lower brainstem was measured by ELISA. Electroencephalograms (EEG) were obtained from using the PTZ-evoked SUDEP model. In our models, atomoxetine-mediated suppression of S-IRA evoked by either acoustic stimulation or PTZ was significantly reversed by low doses of IP and ICV prazosin. Neither repetitive acoustic stimulation nor S-IRA reduced TH levels in lower brainstem. However, the enzyme activity of TH levels in lower brainstem was significantly increased by mechanical ventilation with DBA/1 mice, which makes the dying DBA/1 mice suffering from S-IRA and SUDEP recover. EEG data showed that although the protective effect of atomoxetine was reversed by prazosin, neither drug suppressed EEG activity. These data suggest that deficient synthesis of NE and norepinephrinergic neurotransmission contributed to S-IRA and that the NEα-1R is a potential therapeutic target for the prevention of SUDEP.
Collapse
Affiliation(s)
- Yue Shen
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Department of Anesthesiology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, 310006, China
| | - Hai Xiang Ma
- Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Han Lu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Ting Zhao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jian Liang Sun
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yuan Cheng
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Hong Hai Zhang
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Department of Anesthesiology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, 310006, China; Department of Anesthesiology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China.
| |
Collapse
|
18
|
O'Connor KM, Lucking EF, Cryan JF, O'Halloran KD. Bugs, breathing and blood pressure: microbiota-gut-brain axis signalling in cardiorespiratory control in health and disease. J Physiol 2020; 598:4159-4179. [PMID: 32652603 DOI: 10.1113/jp280279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
There is clear evidence of physiological effects of the gut microbiota on whole-body function in health and disease. Microbiota-gut-brain axis signalling is recognised as a key player in behavioural disorders such as depression and anxiety. Recent evidence suggests that the gut microbiota affects neurocontrol networks responsible for homeostatic functions that are essential for life. We consider the evidence suggesting the potential for the gut microbiota to shape cardiorespiratory homeostasis. In various animal models of disease, there is an association between cardiorespiratory morbidity and perturbed gut microbiota, with strong evidence in support of a role of the gut microbiota in the control of blood pressure. Interventions that target the gut microbiota or manipulate the gut-brain axis, such as short-chain fatty acid supplementation, prevent hypertension in models of obstructive sleep apnoea. Emerging evidence points to a role for the microbiota-gut-brain axis in the control of breathing and ventilatory responsiveness, relevant to cardiorespiratory disease. There is also evidence for an association between the gut microbiota and disease severity in people with asthma and cystic fibrosis. There are many gaps in the knowledge base and an urgent need to better understand the mechanisms by which gut health and dysbiosis contribute to cardiorespiratory control. Nevertheless, there is a growing consensus that manipulation of the gut microbiota could prove an efficacious adjunctive strategy in the treatment of common cardiorespiratory diseases, which are the leading causes of morbidity and mortality.
Collapse
Affiliation(s)
- Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland.,Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
Patrone LGA, Capalbo AC, Marques DA, Bícego KC, Gargaglioni LH. An age- and sex-dependent role of catecholaminergic neurons in the control of breathing and hypoxic chemoreflex during postnatal development. Brain Res 2019; 1726:146508. [PMID: 31606412 DOI: 10.1016/j.brainres.2019.146508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022]
Abstract
The respiratory system undergoes significant development during the postnatal phase. Maturation of brainstem catecholaminergic (CA) neurons is important for the control and modulation of respiratory rhythmogenesis, as well as for chemoreception in early life. We demonstrated an inhibitory role for CA neurons in CO2 chemosensitivity in neonatal and juvenile male and female rats, but information regarding their role in the hypoxic ventilatory response (HVR) is lacking. We evaluated the contribution of brainstem CA neurons in the HVR during postnatal (P) development (P7-8, P14-15 and P20-21) in male and female rats through chemical injury with conjugated saporin anti-dopamine beta-hydroxylase (DβH-SAP, 420 ng·μL-1) injected in the fourth ventricle. Ventilation (V̇E) and oxygen consumption were recorded one week after the lesion in unanesthetized rats during exposure to normoxia and hypoxia. Hypoxia reduced breathing variability in P7-8 control rats of both sexes. At P7-8, the HVR for lesioned males and females increased 27% and 24%, respectively. Additionally, the lesion reduced the normoxic breathing variability in both sexes at P7-8, but hypoxia partially reverted this effect. For P14-15, the increase in V̇E during hypoxia was 30% higher for male and 24% higher for female lesioned animals. A sex-specific difference was detected at P20-21, as lesioned males exhibited a 24% decrease in the HVR, while lesioned females experienced a 22% increase. Furthermore, the hypoxia-induced body temperature reduction was attenuated in P20-21 lesioned females. We conclude that brainstem CA neurons modulate the HRV during the postnatal phase, and possibly thermoregulation during hypoxia.
Collapse
Affiliation(s)
- Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Aretuza C Capalbo
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Danuzia A Marques
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV, Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University, UNESP/FCAV, Jaboticabal, SP, Brazil.
| |
Collapse
|
20
|
The pre-Bötzinger complex: Generation and modulation of respiratory rhythm. NEUROLOGÍA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.nrleng.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
21
|
Garcia AJ, Viemari JC, Khuu MA. Respiratory rhythm generation, hypoxia, and oxidative stress-Implications for development. Respir Physiol Neurobiol 2019; 270:103259. [PMID: 31369874 DOI: 10.1016/j.resp.2019.103259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/15/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Encountered in a number of clinical conditions, repeated hypoxia/reoxygenation during the neonatal period can pose both a threat to immediate survival as well as a diminished quality of living later in life. This review focuses on our current understanding of central respiratory rhythm generation and the role that hypoxia and reoxygenation play in influencing rhythmogenesis. Here, we examine the stereotypical response of the inspiratory rhythm from the preBötzinger complex (preBötC), basic neuronal mechanisms that support rhythm generation during the peri-hypoxic interval, and the physiological consequences of inspiratory network responsivity to hypoxia and reoxygenation, acute and chronic intermittent hypoxia, and oxidative stress. These topics are examined in the context of Sudden Infant Death Syndrome, apneas of prematurity, and neonatal abstinence syndrome.
Collapse
Affiliation(s)
- Alfredo J Garcia
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, Chicago, 60637, IL, United States
| | - Jean Charles Viemari
- Institut de Neurosciences de la Timone, P3M team, UMR7289 CNRS & AMU, Faculté de Médecine de la Timone, 27 Bd Jean Moulin, Marseille, 13005, France
| | - Maggie A Khuu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, Chicago, 60637, IL, United States
| |
Collapse
|
22
|
Oliveira LM, Oliveira MA, Moriya HT, Moreira TS, Takakura AC. Respiratory disturbances in a mouse model of Parkinson's disease. Exp Physiol 2019; 104:729-739. [PMID: 30758090 DOI: 10.1113/ep087507] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the central question of this study? Clinical reports have described and suggested central and peripheral respiratory abnormalities in Parkinson's disease (PD) patients; however, these reports have never addressed the occurrence of these abnormalities in an animal model. What is the main finding and its importance? A mouse model of PD has reduced neurokinin-1 receptor immunoreactivity in the pre-Bӧtzinger complex and Phox2b-expressing neurons in the retrotrapezoid nucleus. The PD mouse has impairments of respiratory frequency and the hypercapnic ventilatory response. Lung collagen deposition and ribcage stiffness appear in PD mice. ABSTRACT Parkinson's disease (PD) is a neurodegenerative motor disorder characterized by dopaminergic deficits in the brain. Parkinson's disease patients may experience shortness of breath, dyspnoea, breathing difficulties and pneumonia, which can be linked as a cause of morbidity and mortality of those patients. The aim of the present study was to clarify whether a mouse model of PD could develop central brainstem and lung respiratory abnormalities. Adult male C57BL/6 mice received bilateral injections of 6-hydroxydopamine (10 μg μl-1 ; 0.5 μl) or vehicle into the striatum. Ventilatory parameters were assessed in the 40 days after induction of PD, by whole-body plethysmography. In addition, measurements of respiratory input impedance (closed and opened thorax) were performed. 6-Hydroxydopamine reduced the number of tyrosine hydroxylase neurons in the substantia nigra pars compacta, the density of neurokinin-1 receptor immunoreactivity in the pre-Bӧtzinger complex and the number of Phox2b neurons in the retrotrapezoid nucleus. Physiological experiments revealed a reduction in resting respiratory frequency in PD animals, owing to an increase in expiratory time and a blunted hypercapnic ventilatory response. Measurements of respiratory input impedance showed that only PD animals with the thorax preserved had increased viscance, indicating that the ribcage could be stiff in this animal model of PD. Consistent with stiffened ribcage mechanics, abnormal collagen deposits in alveolar septa and airways were observed in PD animals. Our data showed that our mouse model of PD presented with neurodegeneration in respiratory brainstem centres and disruption of lung mechanical properties, suggesting that both central and peripheral deficiencies contribute to PD-related respiratory pathologies.
Collapse
Affiliation(s)
- Luiz M Oliveira
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Maria A Oliveira
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Henrique T Moriya
- Biomedical Engineering Laboratory, University of São Paulo, São Paulo, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
23
|
Lucking EF, O'Connor KM, Strain CR, Fouhy F, Bastiaanssen TFS, Burns DP, Golubeva AV, Stanton C, Clarke G, Cryan JF, O'Halloran KD. Chronic intermittent hypoxia disrupts cardiorespiratory homeostasis and gut microbiota composition in adult male guinea-pigs. EBioMedicine 2018; 38:191-205. [PMID: 30446434 PMCID: PMC6306383 DOI: 10.1016/j.ebiom.2018.11.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/26/2018] [Accepted: 11/05/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Carotid body (peripheral oxygen sensor) sensitisation is pivotal in the development of chronic intermittent hypoxia (CIH)-induced hypertension. We sought to determine if exposure to CIH, modelling human sleep apnoea, adversely affects cardiorespiratory control in guinea-pigs, a species with hypoxia-insensitive carotid bodies. We reasoned that CIH-induced disruption of gut microbiota would evoke cardiorespiratory morbidity. METHODS Adult male guinea-pigs were exposed to CIH (6.5% O2 at nadir, 6 cycles.hour-1) for 8 h.day-1 for 12 consecutive days. FINDINGS CIH-exposed animals established reduced faecal microbiota species richness, with increased relative abundance of Bacteroidetes and reduced relative abundance of Firmicutes bacteria. Urinary corticosterone and noradrenaline levels were unchanged in CIH-exposed animals, but brainstem noradrenaline concentrations were lower compared with sham. Baseline ventilation was equivalent in CIH-exposed and sham animals; however, respiratory timing variability, sigh frequency and ventilation during hypoxic breathing were all lower in CIH-exposed animals. Baseline arterial blood pressure was unaffected by exposure to CIH, but β-adrenoceptor-dependent tachycardia and blunted bradycardia during phenylephrine-induced pressor responses was evident compared with sham controls. INTERPRETATION Increased carotid body chemo-afferent signalling appears obligatory for the development of CIH-induced hypertension and elevated chemoreflex control of breathing commonly reported in mammals, with hypoxia-sensitive carotid bodies. However, we reveal that exposure to modest CIH alters gut microbiota richness and composition, brainstem neurochemistry, and autonomic control of heart rate, independent of carotid body sensitisation, suggesting modulation of breathing and autonomic homeostasis via the microbiota-gut-brainstem axis. The findings have relevance to human sleep-disordered breathing. FUNDING The Department of Physiology, and APC Microbiome Ireland, UCC.
Collapse
Affiliation(s)
- Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Conall R Strain
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Fiona Fouhy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Anna V Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
24
|
Ramirez JM, Baertsch N. Defining the Rhythmogenic Elements of Mammalian Breathing. Physiology (Bethesda) 2018; 33:302-316. [PMID: 30109823 PMCID: PMC6230551 DOI: 10.1152/physiol.00025.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 01/08/2023] Open
Abstract
Breathing's remarkable ability to adapt to changes in metabolic, environmental, and behavioral demands stems from a complex integration of its rhythm-generating network within the wider nervous system. Yet, this integration complicates identification of its specific rhythmogenic elements. Based on principles learned from smaller rhythmic networks of invertebrates, we define criteria that identify rhythmogenic elements of the mammalian breathing network and discuss how they interact to produce robust, dynamic breathing.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine , Seattle, Washington
| | - Nathan Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, University of Washington School of Medicine , Seattle, Washington
| |
Collapse
|
25
|
Mueller SG, Nei M, Bateman LM, Knowlton R, Laxer KD, Friedman D, Devinsky O, Goldman AM. Brainstem network disruption: A pathway to sudden unexplained death in epilepsy? Hum Brain Mapp 2018; 39:4820-4830. [PMID: 30096213 DOI: 10.1002/hbm.24325] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/05/2018] [Accepted: 07/12/2018] [Indexed: 12/16/2022] Open
Abstract
Observations in witnessed Sudden Unexpected Death in Epilepsy (SUDEP) suggest that a fatal breakdown of the central autonomic control could play a major role in SUDEP. A previous MR study found volume losses in the mesencephalon in focal epilepsy that were more severe and extended into the lower brainstem in two patients who later died of SUDEP. The aims of this study were to demonstrate an association (1) between brainstem volume loss and impaired autonomic control (reduced heart rate variability [HRV]); (2) between brainstem damage and time to SUDEP in patients who later died of SUDEP. Two populations were studied: (1) Autonomic system function population (ASF, 18 patients with focal epilepsy, 11 controls) with HRV measurements and standardized 3 T MR exams. (2) SUDEP population (26 SUDEP epilepsy patients) with clinical MRI 1-10 years before SUDEP. Deformation-based morphometry of the brainstem was used to generate profile similarity maps from the resulting Jacobian determinant maps that were further characterized by graph analysis to identify regions with excessive expansion indicating significant volume loss or atrophy. The total number of regions with excessive expansion in ASF was negatively correlated with HRV (r = -.37, p = .03), excessive volume loss in periaqueductal gray/medulla oblongata autonomic nuclei explained most of the HRV associated variation (r/r2 = -.82/.67, p < .001). The total number of regions with excessive expansion in SUDEP was negatively correlated with time to SUDEP (r = -.39, p = .03), excessive volume loss in the raphe/medulla oblongata at the obex level explained most of the variation of the time between MRI to SUDEP (r/r2 = -.60/.35,p = .001). Epilepsy is associated with brainstem atrophy that impairs autonomic control and can increase the risk for SUDEP if it expands into the mesencephalon.
Collapse
Affiliation(s)
- Susanne G Mueller
- Department of Radiology, University of California, San Francisco, California
| | - Maromi Nei
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | - Robert Knowlton
- Department of Neurology, University of California, San Francisco, California
| | - Kenneth D Laxer
- Pacific Epilepsy Program, California Pacific Medical Center, San Francisco, California
| | | | | | | |
Collapse
|
26
|
Abstract
Rhythmicity is a universal timing mechanism in the brain, and the rhythmogenic mechanisms are generally dynamic. This is illustrated for the neuronal control of breathing, a behavior that occurs as a one-, two-, or three-phase rhythm. Each breath is assembled stochastically, and increasing evidence suggests that each phase can be generated independently by a dedicated excitatory microcircuit. Within each microcircuit, rhythmicity emerges through three entangled mechanisms: ( a) glutamatergic transmission, which is amplified by ( b) intrinsic bursting and opposed by ( c) concurrent inhibition. This rhythmogenic triangle is dynamically tuned by neuromodulators and other network interactions. The ability of coupled oscillators to reconfigure and recombine may allow breathing to remain robust yet plastic enough to conform to nonventilatory behaviors such as vocalization, swallowing, and coughing. Lessons learned from the respiratory network may translate to other highly dynamic and integrated rhythmic systems, if approached one breath at a time.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98101, USA;
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98101, USA;
| |
Collapse
|
27
|
Peña-Ortega F. Neural Network Reconfigurations: Changes of the Respiratory Network by Hypoxia as an Example. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1015:217-237. [PMID: 29080029 DOI: 10.1007/978-3-319-62817-2_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neural networks, including the respiratory network, can undergo a reconfiguration process by just changing the number, the connectivity or the activity of their elements. Those elements can be either brain regions or neurons, which constitute the building blocks of macrocircuits and microcircuits, respectively. The reconfiguration processes can also involve changes in the number of connections and/or the strength between the elements of the network. These changes allow neural networks to acquire different topologies to perform a variety of functions or change their responses as a consequence of physiological or pathological conditions. Thus, neural networks are not hardwired entities, but they constitute flexible circuits that can be constantly reconfigured in response to a variety of stimuli. Here, we are going to review several examples of these processes with special emphasis on the reconfiguration of the respiratory rhythm generator in response to different patterns of hypoxia, which can lead to changes in respiratory patterns or lasting changes in frequency and/or amplitude.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM-Campus Juriquilla, Boulevard Juriquilla 3001, Querétaro, 76230, Mexico.
| |
Collapse
|
28
|
Garcia AJ, Dashevskiy T, Khuu MA, Ramirez JM. Chronic Intermittent Hypoxia Differentially Impacts Different States of Inspiratory Activity at the Level of the preBötzinger Complex. Front Physiol 2017; 8:571. [PMID: 28936176 PMCID: PMC5603985 DOI: 10.3389/fphys.2017.00571] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/24/2017] [Indexed: 11/13/2022] Open
Abstract
The preBötzinger complex (preBötC) is a medullary brainstem network crucially involved in the generation of different inspiratory rhythms. In the isolated brainstem slice, the preBötC reconfigures to produce different rhythms that we refer to as "fictive eupnea" under baseline conditions (i.e., carbogen), and "fictive gasping" in hypoxia. We recently demonstrated that fictive eupnea is irregular following exposure to chronic intermittent hypoxia (CIH). However, it is unknown how CIH impacts fictive gasping. To address this, brain slices containing the preBötC were prepared from control and CIH exposed mice. Electrophysiological recordings of rhythmogenesis were obtained during the perihypoxic interval. We examined how CIH affects various dynamic aspects of the rhythm characterized by: (1) the irregularity score (IrS), to assess burst-to-variability; (2) the fluctuation value (χ), to quantify the gain of oscillations throughout the time series; and (3) Sample Entropy (sENT), to characterize the pattern/structure of oscillations in the time series. In baseline conditions, CIH increased IrS of amplitude (0.21 ± 0.2) and χ of amplitude (0.34 ± 0.02) but did not affect sENT of amplitude. This indicated that CIH increased burst-to-burst irregularity and the gain of amplitude fluctuations but did not affect the overall pattern/structure of amplitude oscillations. During the transition to hypoxia, 33% of control rhythms whereas 64% of CIH-exposed rhythms showed no doubling of period, suggesting that the probability for stable rhythmogenesis during the transition to hypoxia was greater following CIH. While 29% of control rhythms maintained rhythmicity throughout hypoxia, all slices from CIH exposed mice exhibited rhythms throughout the hypoxic interval. During hypoxia, differences in χ for amplitude were no longer observed between groups. To test the contribution of the persistent sodium current, we examined how riluzole influenced rhythmogenesis following CIH. In networks exposed to CIH, riluzole reduced the IrS of amplitude (-24 ± 14%) yet increased IrS of period (+49 ± 17%). Our data indicate that CIH affects the preBötC, in a manner dependent on the state of the oxygenation. Along with known changes that CIH has on peripheral sensory organs, the effects of CIH on the preBötC may have important implications for sleep apnea, a condition characterized by rapid transitions between normoxia and hypoxia.
Collapse
Affiliation(s)
- Alfredo J. Garcia
- Institute for Integrative Physiology, The University of ChicagoChicago, IL, United States
- Department of Medicine, Section of Emergency Medicine, The University of ChicagoChicago, IL, United States
| | - Tatiana Dashevskiy
- Center for Integrative Brain Research, Seattle Children's Research InstituteSeattle, WA, United States
| | - Maggie A. Khuu
- Institute for Integrative Physiology, The University of ChicagoChicago, IL, United States
- Department of Medicine, Section of Emergency Medicine, The University of ChicagoChicago, IL, United States
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research InstituteSeattle, WA, United States
- Departments of Neurological Surgery and Pediatrics, University of WashingtonSeattle, WA, United States
| |
Collapse
|
29
|
Langer TM, Neumueller SE, Crumley E, Burgraff NJ, Talwar S, Hodges MR, Pan L, Forster HV. Effects on breathing of agonists to μ-opioid or GABA A receptors dialyzed into the ventral respiratory column of awake and sleeping goats. Respir Physiol Neurobiol 2017; 239:10-25. [PMID: 28137700 PMCID: PMC5996971 DOI: 10.1016/j.resp.2017.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Pulmonary ventilation (V̇I) in awake and sleeping goats does not change when antagonists to several excitatory G protein-coupled receptors are dialyzed unilaterally into the ventral respiratory column (VRC). Concomitant changes in excitatory neuromodulators in the effluent mock cerebral spinal fluid (mCSF) suggest neuromodulatory compensation. Herein, we studied neuromodulatory compensation during dialysis of agonists to inhibitory G protein-coupled or ionotropic receptors into the VRC. Microtubules were implanted into the VRC of goats for dialysis of mCSF mixed with agonists to either μ-opioid (DAMGO) or GABAA (muscimol) receptors. We found: (1) V̇I decreased during unilateral but increased during bilateral dialysis of DAMGO, (2) dialyses of DAMGO destabilized breathing, (3) unilateral dialysis of muscimol increased V̇I, and (4) dialysis of DAMGO decreased GABA in the effluent mCSF. We conclude: (1) neuromodulatory compensation can occur during altered inhibitory neuromodulator receptor activity, and (2) the mechanism of compensation differs between G protein-coupled excitatory and inhibitory receptors and between G protein-coupled and inotropic inhibitory receptors.
Collapse
Affiliation(s)
- Thomas M Langer
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Suzanne E Neumueller
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Emma Crumley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Nicholas J Burgraff
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Sawan Talwar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Lawrence Pan
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Department of Physical Therapy, Marquette University, Milwaukee, WI 53226, United States
| | - Hubert V Forster
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53226, United States.
| |
Collapse
|
30
|
O'Halloran KD, Lewis P, McDonald F. Sex, stress and sleep apnoea: Decreased susceptibility to upper airway muscle dysfunction following intermittent hypoxia in females. Respir Physiol Neurobiol 2016; 245:76-82. [PMID: 27884793 DOI: 10.1016/j.resp.2016.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/19/2016] [Accepted: 11/20/2016] [Indexed: 12/21/2022]
Abstract
Obstructive sleep apnoea syndrome (OSAS) is a devastating respiratory control disorder more common in men than women. The reasons for the sex difference in prevalence are multifactorial, but are partly attributable to protective effects of oestrogen. Indeed, OSAS prevalence increases in post-menopausal women. OSAS is characterized by repeated occlusions of the pharyngeal airway during sleep. Dysfunction of the upper airway muscles controlling airway calibre and collapsibility is implicated in the pathophysiology of OSAS, and sex differences in the neuro-mechanical control of upper airway patency are described. It is widely recognized that chronic intermittent hypoxia (CIH), a cardinal feature of OSAS due to recurrent apnoea, drives many of the morbid consequences characteristic of the disorder. In rodents, exposure to CIH-related redox stress causes upper airway muscle weakness and fatigue, associated with mitochondrial dysfunction. Of interest, in adults, there is female resilience to CIH-induced muscle dysfunction. Conversely, exposure to CIH in early life, results in upper airway muscle weakness equivalent between the two sexes at 3 and 6 weeks of age. Ovariectomy exacerbates the deleterious effects of exposure to CIH in adult female upper airway muscle, an effect partially restored by oestrogen replacement therapy. Intriguingly, female advantage intrinsic to upper airway muscle exists with evidence of substantially greater loss of performance in male muscle during acute exposure to severe hypoxic stress. Sex differences in upper airway muscle physiology may have relevance to human OSAS. The oestrogen-oestrogen receptor α axis represents a potential therapeutic target in OSAS, particularly in post-menopausal women.
Collapse
Affiliation(s)
- Ken D O'Halloran
- Department of Physiology, University College Cork, Cork, Ireland.
| | - Philip Lewis
- Department of Physiology, University College Cork, Cork, Ireland; Institute and Policlinic for Occupational Medicine, Environmental Medicine and Preventative Research, University of Cologne, Germany
| | - Fiona McDonald
- Physiology, School of Medicine, University College Dublin, Dublin, Ireland; School of Clinical Sciences, Bristol University, Bristol, United Kingdom
| |
Collapse
|
31
|
Morgan BJ, Bates ML, Rio RD, Wang Z, Dopp JM. Oxidative stress augments chemoreflex sensitivity in rats exposed to chronic intermittent hypoxia. Respir Physiol Neurobiol 2016; 234:47-59. [PMID: 27595979 DOI: 10.1016/j.resp.2016.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
Abstract
Chronic exposure to intermittent hypoxia (CIH) elicits plasticity of the carotid sinus and phrenic nerves via reactive oxygen species (ROS). To determine whether CIH-induced alterations in ventilation, metabolism, and heart rate are also dependent on ROS, we measured responses to acute hypoxia in conscious rats after 14 and 21 d of either CIH or normoxia (NORM), with or without concomitant administration of allopurinol (xanthine oxidase inhibitor), combined allopurinol plus losartan (angiotensin II type 1 receptor antagonist), or apocynin (NADPH oxidase inhibitor). Carotid body nitrotyrosine production was measured by immunohistochemistry. CIH produced an increase in the ventilatory response to acute hypoxia that was virtually eliminated by all three pharmacologic interventions. CIH caused a robust increase in carotid body nitrotyrosine production that was greatly attenuated by allopurinol plus losartan and by apocynin but unaffected by allopurinol. CIH caused a decrease in metabolic rate and a reduction in hypoxic bradycardia. Both of these effects were prevented by allopurinol, allopurinol plus losartan, and apocynin.
Collapse
Affiliation(s)
- Barbara J Morgan
- John Rankin Laboratory of Pulmonary Medicine, School of Medicine and Public Health: University of Wisconsin, Madison, WI 53706, USA; Department of Orthopedics and Rehabilitation, School of Medicine and Public Health; University of Wisconsin, Madison, WI 53706, USA.
| | - Melissa L Bates
- Department of Health and Human Physiology, College of Liberal Arts and Sciences; University of Iowa, Iowa City, IA 52242, USA
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Universidad Autónoma de Chile, Santiago, Chile
| | - Zunyi Wang
- Department of Surgical Sciences, School of Veterinary Medicine; University of Wisconsin, Madison, WI 53706, USA
| | - John M Dopp
- Pharmacy Practice Division, School of Pharmacy; University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
32
|
Microcircuits in respiratory rhythm generation: commonalities with other rhythm generating networks and evolutionary perspectives. Curr Opin Neurobiol 2016; 41:53-61. [PMID: 27589601 DOI: 10.1016/j.conb.2016.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
Abstract
Rhythmicity is critical for the generation of rhythmic behaviors and higher brain functions. This review discusses common mechanisms of rhythm generation, including the role of synaptic inhibition and excitation, with a focus on the mammalian respiratory network. This network generates three phases of breathing and is highly integrated with brain regions associated with numerous non-ventilatory behaviors. We hypothesize that during evolution multiple rhythmogenic microcircuits were recruited to accommodate the generation of each breathing phase. While these microcircuits relied primarily on excitatory mechanisms, synaptic inhibition became increasingly important to coordinate the different microcircuits and to integrate breathing into a rich behavioral repertoire that links breathing to sensory processing, arousal, and emotions as well as learning and memory.
Collapse
|
33
|
Muñoz-Ortiz J, Muñoz-Ortiz E, López-Meraz ML, Beltran-Parrazal L, Morgado-Valle C. Pre-Bötzinger complex: Generation and modulation of respiratory rhythm. Neurologia 2016; 34:461-468. [PMID: 27443242 DOI: 10.1016/j.nrl.2016.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 03/14/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION In mammals, the preBötzinger complex (preBötC) is a bilateral and symmetrical neural network located in the brainstem which is essential for the generation and modulation of respiratory rhythm. There are few human studies about the preBötC and, its relationship with neurological diseases has not been described. However, the importance of the preBötC in neural control of breathing and its potential participation in neurological diseases in humans, has been suggested based on pharmacological manipulation and lesion of the preBötC in animal models, both in vivo and in vitro. METHOD In this review, we describe the effects of some drugs on the inspiratory activity in vitro in a transverse slice that contains the preBötC, as well as some in vivo experiments. Drugs were classified according to their effects on the main neurotransmitter systems and their importance as stimulators or inhibitors of preBötC activity and therefore for the generation of the respiratory rhythm. CONCLUSION Clinical neurologists will find this information relevant to understanding how the central nervous system generates the respiratory rhythm and may also relate this information to the findings made in daily practice.
Collapse
Affiliation(s)
- J Muñoz-Ortiz
- Doctorado en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz, México
| | - E Muñoz-Ortiz
- Doctorado en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz, México
| | - M L López-Meraz
- Centro de Investigaciones Cerebrales, Dirección General de Investigaciones, Universidad Veracruzana, Xalapa de Enríquez, Veracruz, México
| | - L Beltran-Parrazal
- Centro de Investigaciones Cerebrales, Dirección General de Investigaciones, Universidad Veracruzana, Xalapa de Enríquez, Veracruz, México
| | - C Morgado-Valle
- Centro de Investigaciones Cerebrales, Dirección General de Investigaciones, Universidad Veracruzana, Xalapa de Enríquez, Veracruz, México.
| |
Collapse
|
34
|
Tree K, Viemari JC, Cayetanot F, Peyronnet J. Growth restriction induced by chronic prenatal hypoxia affects breathing rhythm and its pontine catecholaminergic modulation. J Neurophysiol 2016; 116:1654-1662. [PMID: 27486108 DOI: 10.1152/jn.00869.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 07/08/2016] [Indexed: 11/22/2022] Open
Abstract
Impaired transplacental supply of oxygen leads to intrauterine growth restriction, one of the most important causes of perinatal mortality and respiratory morbidity. Breathing rhythm depends on the central respiratory network modulated by catecholamines. We investigated the impact of growth restriction, using prenatal hypoxia, on respiratory frequency, on central respiratory-like rhythm, and on its catecholaminergic modulation after birth. At birth, respiratory frequency was increased and confirmed in en bloc medullary preparations, where the frequency of the fourth cervical (C4) ventral root discharge was increased, and in slice preparations containing the pre-Bötzinger complex with an increased inspiratory rhythm. The inhibition of C4 burst discharge observed in pontomedullary preparations was stronger in the growth-restricted group. These results cannot be directly linked by the tyrosine hydroxylase activity increase of A1/C1 and A2/C2 cell groups in the medulla since blockade of α1- and α2-adrenergic receptors did not abolish the difference between both groups. However, in pontomedullary preparations, the stronger inhibition of C4 burst discharge is probably supported by an increased inhibition of A5, a respiratory rhythm inhibitor pontine group of neurons displaying increased tyrosine hydroxylase activity, because blockade of α2-adrenergic receptors abolished the difference between the two groups. Taken together, these results indicate that growth restriction leads to a perturbation of the breathing frequency, which finds, at least in part, its origin in the modification of catecholaminergic modulation of the central breathing network.
Collapse
Affiliation(s)
- K Tree
- UMR 7289, Institut de Neurosciences de la Timone, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | - J C Viemari
- UMR 7289, Institut de Neurosciences de la Timone, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | - F Cayetanot
- UMR 7289, Institut de Neurosciences de la Timone, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | - J Peyronnet
- UMR 7289, Institut de Neurosciences de la Timone, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| |
Collapse
|
35
|
Kuo FS, Falquetto B, Chen D, Oliveira LM, Takakura AC, Mulkey DK. In vitro characterization of noradrenergic modulation of chemosensitive neurons in the retrotrapezoid nucleus. J Neurophysiol 2016; 116:1024-35. [PMID: 27306669 DOI: 10.1152/jn.00022.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/09/2016] [Indexed: 01/08/2023] Open
Abstract
Chemosensitive neurons in the retrotrapezoid nucleus (RTN) regulate breathing in response to CO2/H(+) changes and serve as an integration center for other autonomic centers, including brain stem noradrenergic neurons. Norepinephrine (NE) contributes to respiratory control and chemoreception, and, since disruption of NE signaling may contribute to several breathing disorders, we sought to characterize effects of NE on RTN chemoreception. All neurons included in this study responded similarly to CO2/H(+) but showed differential sensitivity to NE; we found that NE activated (79%), inhibited (7%), or had no effect on activity (14%) of RTN chemoreceptors. The excitatory effect of NE on RTN chemoreceptors was dose dependent, retained in the presence of neurotransmitter receptor blockers, and could be mimicked and blocked by pharmacological manipulation of α1-adrenergic receptors (ARs). In addition, NE-activation was blunted by XE991 (KCNQ channel blocker), and partially occluded the firing response to serotonin, suggesting involvement of KCNQ channels. However, in whole cell voltage clamp, activation of α1-ARs decreased outward current and conductance by what appears to be a mixed effect on multiple channels. The inhibitory effect of NE on RTN chemoreceptors was blunted by an α2-AR antagonist. A third group of RTN chemoreceptors was insensitive to NE. We also found that chemosensitive RTN astrocytes do not respond to NE with a change in voltage or by releasing ATP to enhance activity of chemosensitive neurons. These results indicate NE modulates subsets of RTN chemoreceptors by mechanisms involving α1- and α2-ARs.
Collapse
Affiliation(s)
- Fu-Shan Kuo
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut; and
| | - Bárbara Falquetto
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, Brazil
| | - Dawei Chen
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut; and
| | - Luiz M Oliveira
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana C Takakura
- Department of Pharmacology, University of Sao Paulo, Sao Paulo, Brazil
| | - Daniel K Mulkey
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut; and
| |
Collapse
|
36
|
Oliveira LM, Moreira TS, Kuo FS, Mulkey DK, Takakura AC. α1- and α2-adrenergic receptors in the retrotrapezoid nucleus differentially regulate breathing in anesthetized adult rats. J Neurophysiol 2016; 116:1036-48. [PMID: 27306670 DOI: 10.1152/jn.00023.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/09/2016] [Indexed: 02/07/2023] Open
Abstract
Norepinephrine (NE) is a potent modulator of breathing that can increase/decrease respiratory activity by α1-/α2-adrenergic receptor (AR) activation, respectively. The retrotrapezoid nucleus (RTN) is known to contribute to central chemoreception, inspiration, and active expiration. Here we investigate the sources of catecholaminergic inputs to the RTN and identify respiratory effects produced by activation of ARs in this region. By injecting the retrograde tracer Fluoro-Gold into the RTN, we identified back-labeled catecholaminergic neurons in the A7 region. In urethane-anesthetized, vagotomized, and artificially ventilated male Wistar rats unilateral injection of NE or moxonidine (α2-AR agonist) blunted diaphragm muscle activity (DiaEMG) frequency and amplitude, without changing abdominal muscle activity. Those inhibitory effects were reduced by preapplication of yohimbine (α2-AR antagonist) into the RTN. Conversely, unilateral RTN injection of phenylephrine (α1-AR agonist) increased DiaEMG amplitude and frequency and facilitated active expiration. This response was blocked by prior RTN injection of prazosin (α1-AR antagonist). Interestingly, RTN injection of propranolol (β-AR antagonist) had no effect on respiratory inhibition elicited by applications of NE into the RTN; however, the combined blockade of α2- and β-ARs (coapplication of propranolol and yohimbine) revealed an α1-AR-dependent excitatory response to NE that resulted in increase in DiaEMG frequency and facilitation of active expiration. However, blockade of α1-, α2-, or β-ARs in the RTN had minimal effect on baseline respiratory activity, on central or peripheral chemoreflexes. These results suggest that NE signaling can modulate RTN chemoreceptor function; however, endogenous NE signaling does not contribute to baseline breathing or the ventilatory response to central or peripheral chemoreceptor activity in urethane-anesthetized rats.
Collapse
Affiliation(s)
- Luiz M Oliveira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil; and
| | - Fu-Shan Kuo
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Daniel K Mulkey
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil;
| |
Collapse
|
37
|
O’Halloran KD. Chronic intermittent hypoxia creates the perfect storm with calamitous consequences for respiratory control. Respir Physiol Neurobiol 2016; 226:63-7. [DOI: 10.1016/j.resp.2015.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 11/27/2022]
|
38
|
Bacak BJ, Kim T, Smith JC, Rubin JE, Rybak IA. Mixed-mode oscillations and population bursting in the pre-Bötzinger complex. eLife 2016; 5:e13403. [PMID: 26974345 PMCID: PMC4846382 DOI: 10.7554/elife.13403] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 03/11/2016] [Indexed: 11/17/2022] Open
Abstract
This study focuses on computational and theoretical investigations of neuronal activity arising in the pre-Bötzinger complex (pre-BötC), a medullary region generating the inspiratory phase of breathing in mammals. A progressive increase of neuronal excitability in medullary slices containing the pre-BötC produces mixed-mode oscillations (MMOs) characterized by large amplitude population bursts alternating with a series of small amplitude bursts. Using two different computational models, we demonstrate that MMOs emerge within a heterogeneous excitatory neural network because of progressive neuronal recruitment and synchronization. The MMO pattern depends on the distributed neuronal excitability, the density and weights of network interconnections, and the cellular properties underlying endogenous bursting. Critically, the latter should provide a reduction of spiking frequency within neuronal bursts with increasing burst frequency and a dependence of the after-burst recovery period on burst amplitude. Our study highlights a novel mechanism by which heterogeneity naturally leads to complex dynamics in rhythmic neuronal populations. DOI:http://dx.doi.org/10.7554/eLife.13403.001 Each breath we take removes carbon dioxide from the body and exchanges it for oxygen. A structure called the brainstem, which connects the brain with the spinal cord, generates the breathing rhythm and controls its rate. While this process normally occurs automatically, we can also control our breathing voluntarily, such as when singing or speaking. Within the brainstem, a group of neurons in the area known as the pre-Bötzinger complex is responsible for ensuring that an animal breathes in at regular intervals. Recordings of the electrical activity from slices of brainstem show that pre-Bötzinger neurons display rhythmic activity with characteristic patterns called “mixed-mode oscillations”. These rhythms consist of bursts of strong activity (“large amplitude bursts”), essential for triggering regular breathing, separated by a series of bursts of weak activity (“small amplitude bursts”). However, it is not clear how mixed-mode oscillations arise. Bacak, Kim et al. now provide insights into this process by developing two computational models of the pre-Bötzinger complex. The first model consists of a diverse population of 100 neurons joined by a relatively small number of weak connections to form a network. The second model is a simplified version of the first, consisting of just three neurons. By manipulating the properties of the simulated networks, and analysing the data mathematically, Bacak, Kim et al. identify the properties of the neurons that allow them to generate mixed-mode oscillations and thus rhythmic breathing. The models suggest that mixed-mode oscillations result from the synchronization of many neurons with different levels of activity (excitability). Neurons with low excitability have low bursting frequencies, but generate strong activity and recruit other neurons, ultimately producing large amplitude bursts that cause breathing. Many parts of the nervous system are also made up of networks of neurons with diverse excitability. A challenge for future studies is thus to investigate whether other networks of neurons similar to the pre-Bötzinger complex generate rhythms that control other repetitive actions, such as walking and chewing. DOI:http://dx.doi.org/10.7554/eLife.13403.002
Collapse
Affiliation(s)
- Bartholomew J Bacak
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, United States
| | - Taegyo Kim
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, United States
| | - Jeffrey C Smith
- Cellular and Systems Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Jonathan E Rubin
- Department of Mathematics, University of Pittsburgh, Pittsburgh, United States
| | - Ilya A Rybak
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, United States
| |
Collapse
|
39
|
Morgan BJ, Adrian R, Wang ZY, Bates ML, Dopp JM. Chronic intermittent hypoxia alters ventilatory and metabolic responses to acute hypoxia in rats. J Appl Physiol (1985) 2016; 120:1186-95. [PMID: 26917692 DOI: 10.1152/japplphysiol.00015.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/22/2016] [Indexed: 12/18/2022] Open
Abstract
We determined the effects of chronic exposure to intermittent hypoxia (CIH) on chemoreflex control of ventilation in conscious animals. Adult male Sprague-Dawley rats were exposed to CIH [nadir oxygen saturation (SpO2), 75%; 15 events/h; 10 h/day] or normoxia (NORM) for 21 days. We assessed the following responses to acute, graded hypoxia before and after exposures: ventilation (V̇e, via barometric plethysmography), V̇o2 and V̇co2 (analysis of expired air), heart rate (HR), and SpO2 (pulse oximetry via neck collar). We quantified hypoxia-induced chemoreceptor sensitivity by calculating the stimulus-response relationship between SpO2 and the ventilatory equivalent for V̇co2 (linear regression). An additional aim was to determine whether CIH causes proliferation of carotid body glomus cells (using bromodeoxyuridine). CIH exposure increased the slope of the V̇e/V̇co2/SpO2 relationship and caused hyperventilation in normoxia. Bromodeoxyuridine staining was comparable in CIH and NORM. Thus our CIH paradigm augmented hypoxic chemosensitivity without causing glomus cell proliferation.
Collapse
Affiliation(s)
- Barbara J Morgan
- John Rankin Laboratory of Pulmonary Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin; Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin;
| | - Russell Adrian
- John Rankin Laboratory of Pulmonary Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin; Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Zun-Yi Wang
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin
| | - Melissa L Bates
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa; and
| | - John M Dopp
- Pharmacy Practice Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
40
|
Garcia AJ, Zanella S, Dashevskiy T, Khan SA, Khuu MA, Prabhakar NR, Ramirez JM. Chronic Intermittent Hypoxia Alters Local Respiratory Circuit Function at the Level of the preBötzinger Complex. Front Neurosci 2016; 10:4. [PMID: 26869872 PMCID: PMC4740384 DOI: 10.3389/fnins.2016.00004] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/07/2016] [Indexed: 01/08/2023] Open
Abstract
Chronic intermittent hypoxia (CIH) is a common state experienced in several breathing disorders, including obstructive sleep apnea (OSA) and apneas of prematurity. Unraveling how CIH affects the CNS, and in turn how the CNS contributes to apneas is perhaps the most challenging task. The preBötzinger complex (preBötC) is a pre-motor respiratory network critical for inspiratory rhythm generation. Here, we test the hypothesis that CIH increases irregular output from the isolated preBötC, which can be mitigated by antioxidant treatment. Electrophysiological recordings from brainstem slices revealed that CIH enhanced burst-to-burst irregularity in period and/or amplitude. Irregularities represented a change in individual fidelity among preBötC neurons, and changed transmission from preBötC to the hypoglossal motor nucleus (XIIn), which resulted in increased transmission failure to XIIn. CIH increased the degree of lipid peroxidation in the preBötC and treatment with the antioxidant, 5,10,15,20-Tetrakis (1-methylpyridinium-4-yl)-21H,23H-porphyrin manganese(III) pentachloride (MnTMPyP), reduced CIH-mediated irregularities on the network rhythm and improved transmission of preBötC to the XIIn. These findings suggest that CIH promotes a pro-oxidant state that destabilizes rhythmogenesis originating from the preBötC and changes the local rhythm generating circuit which in turn, can lead to intermittent transmission failure to the XIIn. We propose that these CIH-mediated effects represent a part of the central mechanism that may perpetuate apneas and respiratory instability, which are hallmark traits in several dysautonomic conditions.
Collapse
Affiliation(s)
- Alfredo J Garcia
- Center for Integrative Brain Research, Seattle Children's Research Institute Seattle, WA, USA
| | - Sebastien Zanella
- Center for Integrative Brain Research, Seattle Children's Research Institute Seattle, WA, USA
| | - Tatiana Dashevskiy
- Center for Integrative Brain Research, Seattle Children's Research Institute Seattle, WA, USA
| | - Shakil A Khan
- Institute for Integrative Physiology, The University of Chicago Chicago, IL, USA
| | - Maggie A Khuu
- Center for Integrative Brain Research, Seattle Children's Research Institute Seattle, WA, USA
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology, The University of Chicago Chicago, IL, USA
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research InstituteSeattle, WA, USA; Departments of Neurological Surgery and Pediatrics, University of WashingtonSeattle, WA, USA
| |
Collapse
|
41
|
Gulati IK, Shubert TR, Sitaram S, Wei L, Jadcherla SR. Effects of birth asphyxia on the modulation of pharyngeal provocation-induced adaptive reflexes. Am J Physiol Gastrointest Liver Physiol 2015; 309:G662-9. [PMID: 26272260 PMCID: PMC4609929 DOI: 10.1152/ajpgi.00204.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/10/2015] [Indexed: 01/31/2023]
Abstract
Perinatal asphyxia and aerodigestive symptoms are troublesome. We tested the hypothesis that pharyngeal provocation alters proximal and distal aerodigestive reflex coordination and kinetics in infants with hypoxic ischemic encephalopathy (HIE), compared with healthy controls. Specifically, we characterized the sensory-motor properties of pharyngeal provocation-induced effects on upper esophageal sphincter (UES) and lower esophageal sphincter (LES) reflexes. Ten orally fed controls (32.0 ± 1.5 wk gestation) and 25 infants with HIE (38.1 ± 0.4 wk gestation) were evaluated at 39.7 ± 0.9 and 41.9 ± 0.6 wk postmenstrual age respectively. Pharyngo-esophageal reflexes evoked upon graded water stimuli were tested using water-perfusion micromanometry methods. Analysis included sensory-motor characteristics of pharyngeal reflexive swallow (PRS), pharyngo-UES-contractile reflex (PUCR), esophageal body-waveform kinetics, and pharyngo-LES-relaxation reflex (PLESRR). For controls vs. infants with HIE, median appearance, pulse, grimace, activity, respiration (APGAR) scores were 6 vs. 1 at 1 min (P < 0.001) and 8 vs. 3 at 5 min (P < 0.001). Upon pharyngeal- stimulation, HIE infants (vs. controls) had frequent PUCR (P = 0.01); increased UES basal tone (P = 0.03); decreased LES basal tone (P = 0.002); increased pharyngeal-waveforms per stimulus (P = 0.03); decreased frequency of LES relaxation (P = 0.003); and decreased proximal esophageal contractile amplitude (P = 0.002), with prolonged proximal esophageal contractile duration (P = 0.008). Increased tonicity and reactivity of the UES and dysregulation of LES may provide the pathophysiological basis for pooling of secretions, improper bolus clearance, and aspiration risk. Deficits in function at the nuclear or supranuclear level involving glossopharyngeal and vagal neural networks and respiratory regulatory pathways involved with aerodigestive protection may be contributory.
Collapse
Affiliation(s)
- Ish K. Gulati
- 1Sections of Neonatology, Pediatric Gastroenterology, and Nutrition, Department of Pediatrics, The Ohio State University College of Medicine, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; ,2The Neonatal and Infant Feeding Disorders Program, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| | - Theresa R. Shubert
- 2The Neonatal and Infant Feeding Disorders Program, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| | - Swetha Sitaram
- 2The Neonatal and Infant Feeding Disorders Program, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| | - Lai Wei
- 3Center for Biostatistics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Sudarshan R. Jadcherla
- 1Sections of Neonatology, Pediatric Gastroenterology, and Nutrition, Department of Pediatrics, The Ohio State University College of Medicine, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; ,2The Neonatal and Infant Feeding Disorders Program, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| |
Collapse
|
42
|
Muere C, Neumueller S, Olesiak S, Miller J, Langer T, Hodges MR, Pan L, Forster HV. Combined unilateral blockade of cholinergic, peptidergic, and serotonergic receptors in the ventral respiratory column does not affect breathing in awake or sleeping goats. J Appl Physiol (1985) 2015; 119:308-20. [PMID: 26023224 DOI: 10.1152/japplphysiol.00145.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/23/2015] [Indexed: 12/21/2022] Open
Abstract
Previous work in intact awake and sleeping goats has found that unilateral blockade of excitatory inputs in the ventral respiratory column (VRC) elicits changes in the concentrations of multiple neurochemicals, including serotonin (5-HT), substance P, glycine, and GABA, while increasing or having no effect on breathing. These findings are consistent with the concept of interdependence between neuromodulators, whereby attenuation of one modulator elicits compensatory changes in other modulators to maintain breathing. Because there is a large degree of redundancy and multiplicity of excitatory inputs to the VRC, we herein tested the hypothesis that combined unilateral blockade of muscarinic acetylcholine (mACh), neurokinin-1 (NK1, the receptor for substance P), and 5-HT2A receptors would elicit changes in multiple neurochemicals, but would not change breathing. We unilaterally reverse-dialyzed a cocktail of antagonists targeting these receptors into the VRC of intact adult goats. Breathing was continuously monitored while effluent fluid from dialysis was collected for quantification of neurochemicals. We found that neither double blockade of mACh and NK1 receptors, nor triple blockade of mACh, NK1, and 5-HT2A receptors significantly affected breathing (P ≥ 0.05) in goats that were awake or in non-rapid eye movement (NREM) sleep. However, both double and triple blockade increased the effluent concentration of substance P (P < 0.001) and decreased GABA concentrations. These findings support our hypothesis and, together with past data, suggest that both in wakefulness and NREM sleep, multiple neuromodulator systems collaborate to stabilize breathing when a deficit in one or multiple excitatory neuromodulators exists.
Collapse
Affiliation(s)
- Clarissa Muere
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Suzanne Neumueller
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Samantha Olesiak
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Justin Miller
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| | - Thomas Langer
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lawrence Pan
- Department of Physical Therapy, Marquette University, Milwaukee, Wisconsin; and
| | - Hubert V Forster
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
43
|
Koch H, Caughie C, Elsen FP, Doi A, Garcia AJ, Zanella S, Ramirez JM. Prostaglandin E2 differentially modulates the central control of eupnoea, sighs and gasping in mice. J Physiol 2014; 593:305-19. [PMID: 25556802 DOI: 10.1113/jphysiol.2014.279794] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/24/2014] [Indexed: 01/10/2023] Open
Abstract
Prostaglandin E2 (PGE2) augments distinct inspiratory motor patterns, generated within the preBötzinger complex (preBötC), in a dose-dependent way. The frequency of sighs and gasping are stimulated at low concentrations, while the frequency of eupnoea increases only at high concentrations. We used in vivo microinjections into the preBötC and in vitro isolated brainstem slice preparations to investigate the dose-dependent effects of PGE2 on the preBötC activity. Synaptic measurements in whole cell voltage clamp recordings of inspiratory neurons revealed no changes in inhibitory or excitatory synaptic transmission in response to PGE2 exposure. In current clamp recordings obtained from inspiratory neurons of the preBötC, we found an increase in the frequency and amplitude of bursting activity in neurons with intrinsic bursting properties after exposure to PGE2. Riluzole, a blocker of the persistent sodium current, abolished the effect of PGE2 on sigh activity, while flufenamic acid, a blocker of the calcium-activated non-selective cation conductance, abolished the effect on eupnoeic activity caused by PGE2. Prostaglandins are important regulators of autonomic functions in the mammalian organism. Here we demonstrate in vivo that prostaglandin E2 (PGE2) can differentially increase the frequency of eupnoea (normal breathing) and sighs (augmented breaths) when injected into the preBötzinger complex (preBötC), a medullary area that is critical for breathing. Low concentrations of PGE2 (100-300 nm) increased the sigh frequency, while higher concentrations (1-2 μm) were required to increase the eupnoeic frequency. The concentration-dependent effects were similarly observed in the isolated preBötC. This in vitro preparation also revealed that riluzole, a blocker of the persistent sodium current (INap), abolished the modulatory effect on sighs, while flufenamic acid, an antagonist for the calcium-activated non-selective cation conductance (ICAN ) abolished the effect of PGE2 on fictive eupnoea at higher concentrations. At the cellular level PGE2 significantly increased the amplitude and frequency of intrinsic bursting in inspiratory neurons. By contrast PGE2 affected neither excitatory nor inhibitory synaptic transmission. We conclude that PGE2 differentially modulates sigh, gasping and eupnoeic activity by differentially increasing INap and ICAN currents in preBötC neurons.
Collapse
Affiliation(s)
- Henner Koch
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Neurological Surgery, University of Washington, Seattle, WA, 98104, USA; Department of Neurology, University of Tübingen, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Cropper EC, Friedman AK, Jing J, Perkins MH, Weiss KR. Neuromodulation as a mechanism for the induction of repetition priming. Curr Opin Neurobiol 2014; 29:33-8. [PMID: 25261622 DOI: 10.1016/j.conb.2014.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 11/25/2022]
Abstract
It is becoming apparent that the activity of many neural networks is shaped by effects of endogenous neuromodulators. Modulators exert second messenger-mediated actions that persist. We consider how this may impact network function and its potential role in the induction of repetition priming (increased performance when behavior is repeated). When effects of modulators persist and modulatory substances are repeatedly released, their effects will accumulate (summate) and become more pronounced. If this enhances the ability of a network to generate a particular output, performance will improve. We review data that support this model, and consider its implications for task switching. This model predicts that priming of one type of network activity will negatively impact the rapid transition to an incompatible type.
Collapse
Affiliation(s)
- Elizabeth C Cropper
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States.
| | - Allyson K Friedman
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States
| | - Jian Jing
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Jiangsu 210093, China
| | - Matthew H Perkins
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States
| | - Klaudiusz R Weiss
- Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, United States
| |
Collapse
|