1
|
Claure N, Tolosa J, Jain D, Schott A, Aguilar AC, Dormishian A, Bancalari E. Effect of Intermittent Hypoxemia and Hyperoxemia during the Neonatal Period on Control of Breathing Function among Infants Born Extremely Preterm. J Pediatr 2025; 281:114542. [PMID: 40090544 DOI: 10.1016/j.jpeds.2025.114542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE To evaluate the association between intermittent hypoxemia (IH) and hyperoxemia (HOX) during the first 28 days with peripheral and central chemoreception at 36 weeks of postmenstrual age among infants born extremely preterm. STUDY DESIGN For this observational study, 52 infants born at 23-28 weeks of gestational age were enrolled. Mean daily IH frequency (arterial oxygen saturation <80% for ≥10 seconds) and percent of time in HOX (arterial oxygen saturation ≥98% while the fraction of inspired oxygen was >0.21) were calculated for the first 28 days of life. At 36 weeks of postmenstrual age, respiratory control tests assessed peripheral chemoreception by ventilatory response to 100% O2 for 30 seconds in which decreased ventilation caused by inhibition of peripheral chemoreceptors reflects their contribution to respiratory drive. Central chemoreception was evaluated by ventilatory response to 4% inspired CO2 for 10 minutes. RESULTS Multivariable generalized linear models showed increasing IH and HOX were independently associated with an attenuated ventilatory response to 100% O2 at 36 weeks of postmenstrual age. IH and HOX were not significantly associated with an attenuated ventilatory response to CO2. CONCLUSIONS In these infants born extremely preterm, neonatal IH and HOX were independently associated with attenuated peripheral chemoreception at near-term corrected age. This may reflect reduced peripheral chemoreceptor oxygen sensitivity and may be in part responsible for persistence of respiratory instability in infants born preterm. Neonatal IH or HOX were not associated with reduced central chemoreception.
Collapse
Affiliation(s)
- Nelson Claure
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL; Biomedical Engineering, College of Engineering, University of Miami, Miami, FL.
| | - Jose Tolosa
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL
| | - Deepak Jain
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL
| | - Alini Schott
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL
| | - Ana Cecilia Aguilar
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL
| | - Alaleh Dormishian
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL; Biomedical Engineering, College of Engineering, University of Miami, Miami, FL
| | - Eduardo Bancalari
- Division of Neonatology, Departments of Pediatrics, Miller School of Medicine, Miami, FL
| |
Collapse
|
2
|
Apicella R, Mazzone GL, Taccola G. Acute intermittent hypoxia in neonatal rodent central nervous system facilitates respiratory frequency through the recruitment of hypothalamic areas. Exp Physiol 2025. [PMID: 40083110 DOI: 10.1113/ep092303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025]
Abstract
Moderate and acute intermittent hypoxia (IH) facilitates respiration in adults, mostly by recruiting peripheral chemo-/baroreceptors. As central chemoreceptors are widely expressed in immature brains, we hypothesized that IH modulates respiration at birth through a purely neurogenic mechanism involving the hypothalamus. The central nervous system (CNS) isolated from 0- to 3-day-old rats was perfused with four to eight brief (5 min) bouts of mild-hypoxic/normocapnic modified Krebs solution, intermingled with 5-min normoxic episodes, during continuous electrophysiological recordings from upper cervical ventral roots. An IH protocol did not modify bath pH, but superficial ventrolateral medulla and hypothalamic areas experienced lowered oxygen tension, more severe after the second postnatal day, with a partial recovery after each bout. Single exposures to mild hypoxia were well tolerated, and at birth often triggered a spontaneous epoch of irregular baseline activity (< 1 min) superimposed on respiratory events in both whole CNS preparations and spinal cords. Conversely, IH largely halted breathing activity after the second postnatal day, while at birth IH transiently increased the amplitude of respiratory bursts and stably sped up rhythm only when intact suprapontine structures were present. Rhythm acceleration was not directly correlated to instantaneous changes in tissue oxygen tension. After IH, respiratory frequency remained 260% higher than pre-IH control for up to 60 min. Identical modulatory effects were observed with IH supplied through a HEPES buffer solution. Interestingly, IH increased electrical activity and cFos expression in hypothalamic areas without altering total cell number. These observations cast some light on the mechanisms of IH during development, with important insights about pediatric effects of repeated hypoxic episodes.
Collapse
Affiliation(s)
- Rosamaria Apicella
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
- Applied Neurophysiology and Neuropharmacology Lab, Istituto di Medicina Fisica e Riabilitazione (IMFR), Udine, Italy
| | - Graciela L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Buenos Aires, Argentina
| | - Giuliano Taccola
- Neuroscience Department, International School for Advanced Studies (SISSA), Trieste, Italy
- Applied Neurophysiology and Neuropharmacology Lab, Istituto di Medicina Fisica e Riabilitazione (IMFR), Udine, Italy
| |
Collapse
|
3
|
Rüegger CM, Dawson JA, Cracknell J, Fiander M, Davis PG, Gaertner VD. Air versus supplemental oxygen for resuscitation of term or late preterm infants at birth. Cochrane Database Syst Rev 2024; 12:CD014781. [PMID: 39704284 PMCID: PMC11660226 DOI: 10.1002/14651858.cd014781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: Primary objective To assess the benefits and harms of air compared with supplemental oxygen for resuscitation of term or late preterm infants at birth in reducing rates of mortality and long-term neurodevelopmental impairment (NDI). Secondary objectives To assess whether the benefits and harms of air compared with supplemental oxygen differ according to different oxygen concentrations, gestational age (GA), whether oxygen was titrated to saturation curves and the income of the study country.
Collapse
Affiliation(s)
- Christoph M Rüegger
- Newborn Research, Department of Neonatology, University Hospital and University of Zürich, Zürich, Switzerland
| | - Jennifer A Dawson
- Newborn Research Centre, Royal Women's Hospital, Melbourne, Australia
- Murdoch Children's Research Institute, Melbourne, Australia
| | | | | | - Peter G Davis
- Murdoch Children's Research Institute, Melbourne, Australia
- Newborn Research Centre and Neonatal Services, The Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne, Melbourne, Australia
| | - Vincent D Gaertner
- Division of Neonatology, Dr. von Hauner'sches Children's Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| |
Collapse
|
4
|
Cheung EC, Nilsson A, Venter I, Kowalik G, Ribeiro C, Rodriguez J, Kuraoka K, Russo R, Escobar JB, Alber BR, Mendelowitz D, Kay MW, Schunke KJ. Sex differences in cardiac transcriptomic response to neonatal sleep apnea. Physiol Rep 2024; 12:e16110. [PMID: 38981849 PMCID: PMC11233197 DOI: 10.14814/phy2.16110] [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: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Pediatric obstructive sleep apnea poses a significant health risk, with potential long-term consequences on cardiovascular health. This study explores the dichotomous nature of neonatal cardiac response to chronic intermittent hypoxia (CIH) between males and females, aiming to fill a critical knowledge gap in the understanding of sex-specific cardiovascular consequences of sleep apnea in early life. Neonates were exposed to CIH until p28 and underwent comprehensive in vivo physiological assessments, including whole-body plethysmography, treadmill stress-tests, and echocardiography. Results indicated that male CIH rats weighed 13.7% less than age-matched control males (p = 0.0365), while females exhibited a mild yet significant increased respiratory drive during sleep (93.94 ± 0.84 vs. 95.31 ± 0.81;p = 0.02). Transcriptomic analysis of left ventricular tissue revealed a substantial sex-based difference in the cardiac response to CIH, with males demonstrating a more pronounced alteration in gene expression compared to females (5986 vs. 3174 genes). The dysregulated miRNAs in males target metabolic genes, potentially predisposing the heart to altered metabolism and substrate utilization. Furthermore, CIH in males was associated with thinner left ventricular walls and dysregulation of genes involved in the cardiac action potential, possibly predisposing males to CIH-related arrhythmia. These findings emphasize the importance of considering sex-specific responses in understanding the cardiovascular implications of pediatric sleep apnea.
Collapse
Affiliation(s)
- Emily C. Cheung
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Anna Nilsson
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Ian Venter
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Grant Kowalik
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Caitlin Ribeiro
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Jeannette Rodriguez
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Kiralee Kuraoka
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Rebekah Russo
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Joan B. Escobar
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Bridget R. Alber
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - David Mendelowitz
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Matthew W. Kay
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Kathryn J. Schunke
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| |
Collapse
|
5
|
Apicella R, Taccola G. Passive limb training modulates respiratory rhythmic bursts. Sci Rep 2023; 13:7226. [PMID: 37142670 PMCID: PMC10160044 DOI: 10.1038/s41598-023-34422-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/29/2023] [Indexed: 05/06/2023] Open
Abstract
Exercise modifies respiratory functions mainly through the afferent feedback provided by exercising limbs and the descending input from suprapontine areas, two contributions that are still underestimated in vitro. To better characterize the role of limb afferents in modulating respiration during physical activity, we designed a novel experimental in vitro platform. The whole central nervous system was isolated from neonatal rodents and kept with hindlimbs attached to an ad-hoc robot (Bipedal Induced Kinetic Exercise, BIKE) driving passive pedaling at calibrated speeds. This setting allowed extracellular recordings of a stable spontaneous respiratory rhythm for more than 4 h, from all cervical ventral roots. BIKE reversibly reduced the duration of single respiratory bursts even at lower pedaling speeds (2 Hz), though only an intense exercise (3.5 Hz) modulated the frequency of breathing. Moreover, brief sessions (5 min) of BIKE at 3.5 Hz augmented the respiratory rate of preparations with slow bursting in control (slower breathers) but did not change the speed of faster breathers. When spontaneous breathing was accelerated by high concentrations of potassium, BIKE reduced bursting frequency. Regardless of the baseline respiratory rhythm, BIKE at 3.5 Hz always decreased duration of single bursts. Surgical ablation of suprapontine structures completely prevented modulation of breathing after intense training. Albeit the variability in baseline breathing rates, intense passive cyclic movement tuned fictive respiration toward a common frequency range and shortened all respiratory events through the involvement of suprapontine areas. These observations contribute to better define how the respiratory system integrates sensory input from moving limbs during development, opening new rehabilitation perspectives.
Collapse
Affiliation(s)
- Rosamaria Apicella
- Neuroscience Department, International School for Advanced Studies (SISSA), Via Bonomea 265, Trieste, Italy
- Applied Neurophysiology and Neuropharmacology Lab, Istituto Di Medicina Fisica E Riabilitazione (IMFR), Via Gervasutta 48, Udine, UD, Italy
| | - Giuliano Taccola
- Neuroscience Department, International School for Advanced Studies (SISSA), Via Bonomea 265, Trieste, Italy.
- Applied Neurophysiology and Neuropharmacology Lab, Istituto Di Medicina Fisica E Riabilitazione (IMFR), Via Gervasutta 48, Udine, UD, Italy.
| |
Collapse
|
6
|
da Silva Junior CA, Patrone LGA, Biancardi V, Vilela-Costa HH, Marques DA, Cristina-Silva C, da Costa Silva KS, Bícego KC, Szawka RE, Gargaglioni LH. Sexually dimorphic effects of prenatal diazepam exposure on respiratory control and the monoaminergic system of neonate and young rats. Pflugers Arch 2022; 474:1185-1200. [DOI: 10.1007/s00424-022-02730-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/05/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022]
|
7
|
Biancardi V, Patrone LGA, Vicente MC, Marques DA, Bicego KC, Funk GD, Gargaglioni LH. Prenatal fluoxetine has long lasting, differential effects on respiratory control in male and female rats. J Appl Physiol (1985) 2022; 133:371-389. [PMID: 35708704 DOI: 10.1152/japplphysiol.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Serotonin (5-HT) is an important modulator of brain networks that control breathing. The selective serotonin reuptake inhibitor fluoxetine (FLX) is the first-line antidepressant drug prescribed during pregnancy. We investigated the effects of prenatal FLX on baseline breathing, ventilatory and metabolic responses to hypercapnia and hypoxia as well as number of brainstem 5-HT and tyrosine hydroxylase (TH) neurons of rats during postnatal development (P0-82). Prenatal FLX exposure of males showed a lower baseline that appeared in juveniles and remained in adulthood, with no sleep-wake state dependency. Prenatal FLX exposure of females did not affect baseline breathing. Juvenile male FLX rats showed increased CO2 and hypoxic ventilatory responses, normalizing by adulthood. Alterations in juvenile-FLX treated males were associated with greater number of 5-HT neurons in the ROB and RMAG. Adult FLX-exposed males showed greater number of 5-HT neurons in the RPA and TH neurons in the A5, while reduced number of TH neurons in A7. Prenatal FLX exposure of female rats was associated with greater hyperventilation induced by hypercapnia at P0-2 and juveniles whereas P12-14 and adult FLX (NREM sleep) rats showed an attenuation of the hypercapnic hyperventilation.FLX-exposed females had fewer 5-HT neurons in the RPA and reduced TH A6 density at P0-2; and greater number of TH neurons in the A7 at P12-14. These data indicate that prenatal FLX exposure affects the number of neurons of some monoaminergic regions in the brain and results in long lasting, sex specific changes in baseline breathing pattern and ventilatory responses to respiratory challenges.
Collapse
Affiliation(s)
- Vivian Biancardi
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,Department of Physiology, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Mariane C Vicente
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Danuzia A Marques
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,Department of Pediatrics, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec, QC, Canada
| | - Kênia C Bicego
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Gregory D Funk
- Department of Physiology, Faculty of Medicine and Dentistry, Women and Children's Health Research Institute, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| |
Collapse
|
8
|
Kinkead R, Gagnon M, Joseph V, Sériès F, Ambrozio-Marques D. Stress and Loss of Ovarian Function: Novel Insights into the Origins of Sex-Based Differences in the Manifestations of Respiratory Control Disorders During Sleep. Clin Chest Med 2021; 42:391-405. [PMID: 34353446 DOI: 10.1016/j.ccm.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The respiratory system of women and men develops and functions in distinct neuroendocrine milieus. Despite differences in anatomy and neural control, homeostasis of arterial blood gases is ensured in healthy individuals regardless of sex. This convergence in function differs from the sex-based differences observed in many respiratory diseases. Sleep-disordered breathing (SDB) results mainly from episodes of upper airway closure. This complex and multifactorial respiratory disorder shows significant sexual dimorphism in its clinical manifestations and comorbidities. Guided by recent progress from basic research, this review discusses the hypothesis that stress is necessary to reveal the sexual dimorphism of SDB.
Collapse
Affiliation(s)
- Richard Kinkead
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada.
| | - Marianne Gagnon
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada
| | - Vincent Joseph
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada
| | - Frédéric Sériès
- Department of Medicine, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Québec, Canada
| | - Danuzia Ambrozio-Marques
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada
| |
Collapse
|
9
|
Adenosine A2a receptors modulate TrkB receptor-dependent respiratory plasticity in neonatal rats. Respir Physiol Neurobiol 2021; 294:103743. [PMID: 34273553 DOI: 10.1016/j.resp.2021.103743] [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: 04/06/2021] [Revised: 07/06/2021] [Accepted: 07/11/2021] [Indexed: 11/24/2022]
Abstract
Neuroplasticity is a fundamental property of the respiratory control system, enabling critical adaptations in breathing to meet the challenges, but little is known whether neonates express neuroplasticity similar to adults. We tested the hypothesis that, similar to adults, tyrosine receptor kinase B (TrkB) or adenosine A2a receptor activation in neonates are independently sufficient to elicit respiratory motor facilitation, and that co-induction of TrkB and A2a receptor-dependent plasticity undermines respiratory motor facilitation. TrkB receptor activation with 7,8-dihydroxyflavone (DHF) in neonatal brainstem-spinal cord preparations induced a long-lasting increase in respiratory motor output in 55 % of preparations, whereas adenosine A2a receptor activation with CGS21680 only sporadically induced respiratory motor plasticity. CGS21680 and DHF co-application prevented DHF-dependent respiratory motor facilitation, whereas co-application of MSX-3 (adenosine A2a receptor antagonist) and DHF more rapidly induced respiratory motor plasticity. Collectively, these data suggest that mechanisms underlying respiratory neuroplasticity may be only partially operational in early neonatal life, and that adenosine A2a receptor activation undermines TrkB-induced respiratory plasticity.
Collapse
|
10
|
Anunziata F, Macchione AF, Alcalde AA, Tejerina DN, Amigone JL, Wille-Bille A, Trujillo V, Molina JC. Ethanol's disruptive effects upon early breathing plasticity and blood parameters associated with hypoxia and hypercapnia. Exp Neurol 2021; 344:113796. [PMID: 34224736 DOI: 10.1016/j.expneurol.2021.113796] [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: 04/15/2021] [Revised: 06/16/2021] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
Early ethanol exposure affects respiratory neuroplasticity; a risk factor associated with the Sudden Infant Death Syndrome. High and chronic ethanol doses exert long-lasting effects upon respiratory rates, apneic episodes and ventilatory processes triggered by hypoxia. The present study was performed in 3-9-day-old rat pups. Respiratory processes under normoxic and hypoxic conditions were analyzed in pups intoxicated with different ethanol doses which were pre-exposed or not to the drug. A second major goal was to examine if acute and/or chronic early ethanol exposure affects blood parameters related with hypercapnic or hypoxic states. In Experiment 1, at postnatal day 9, animals previously treated with ethanol (2.0 g/kg) or vehicle (0.0 g/kg) were tested sober or intoxicated with 0.75, 1.37 or 2.00 g/kg ethanol. The test involved sequential air conditions defined as initial normoxia, hypoxia and recovery normoxia. Motor activity was also evaluated. In Experiment 2, blood parameters indicative of possible hypoxic and hypercapnic states were assessed as a function of early chronic or acute experiences with the drug. The main results of Experiment 1 were as follows: i) ethanol's depressant effects upon respiratory rates increased as a function of sequential treatment with the drug (sensitization); ii) ethanol inhibited apneic episodes even when employing the lowest dose at test (0.75 g/kg); iii) the hyperventilatory response caused by hypoxia negatively correlated with the ethanol dose administered at test; iv) ventilatory long-term facilitation (LTF) during recovery normoxia was observed in pups pre-exposed to the drug and in pups that received the different ethanol doses at test; v) self-grooming increased in pups treated with either 1.37 or 2.00 g/kg ethanol. The main result of Experiment 2 indicated that acute as well as chronic ethanol exposure results in acidosis-hypercapnia. The results indicate that early and brief experiences with ethanol are sufficient to affect different respiratory plasticity processes as well as blood biomarkers indicative of acidosis-hypercapnia. An association between the LTF process and the acidosis-hypercapnic state caused by ethanol seems to exist. The mentioned experiences with the drug are sufficient to result in an anomalous programming of respiratory patterns and metabolic conditions.
Collapse
Affiliation(s)
- Florencia Anunziata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana F Macchione
- Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto de Investigaciones Psicológicas, IIPsi-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - Asier Angulo Alcalde
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Procesos Psicológicos Básicos y su Desarrollo, Facultad de Psicología, Universidad del País Vasco UPV-EHU, Donostia-San Sebastián, Guipúzcoa, Spain
| | - David N Tejerina
- Laboratorio de Bioquímica Clínica, Hospital Privado de Córdoba, Córdoba, Argentina
| | - José L Amigone
- Laboratorio de Bioquímica Clínica, Hospital Privado de Córdoba, Córdoba, Argentina
| | - Aranza Wille-Bille
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Verónica Trujillo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Juan C Molina
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
11
|
Tenorio-Lopes L, Kinkead R. Sex-Specific Effects of Stress on Respiratory Control: Plasticity, Adaptation, and Dysfunction. Compr Physiol 2021; 11:2097-2134. [PMID: 34107062 DOI: 10.1002/cphy.c200022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As our understanding of respiratory control evolves, we appreciate how the basic neurobiological principles of plasticity discovered in other systems shape the development and function of the respiratory control system. While breathing is a robust homeostatic function, there is growing evidence that stress disrupts respiratory control in ways that predispose to disease. Neonatal stress (in the form of maternal separation) affects "classical" respiratory control structures such as the peripheral O2 sensors (carotid bodies) and the medulla (e.g., nucleus of the solitary tract). Furthermore, early life stress disrupts the paraventricular nucleus of the hypothalamus (PVH), a structure that has emerged as a primary determinant of the intensity of the ventilatory response to hypoxia. Although underestimated, the PVH's influence on respiratory function is a logical extension of the hypothalamic control of metabolic demand and supply. In this article, we review the functional and anatomical links between the stress neuroendocrine axis and the medullary network regulating breathing. We then present the persistent and sex-specific effects of neonatal stress on respiratory control in adult rats. The similarities between the respiratory phenotype of stressed rats and clinical manifestations of respiratory control disorders such as sleep-disordered breathing and panic attacks are remarkable. These observations are in line with the scientific consensus that the origins of adult disease are often found among developmental and biological disruptions occurring during early life. These observations bring a different perspective on the structural hierarchy of respiratory homeostasis and point to new directions in our understanding of the etiology of respiratory control disorders. © 2021 American Physiological Society. Compr Physiol 11:1-38, 2021.
Collapse
Affiliation(s)
- Luana Tenorio-Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
12
|
Drummond SE, Burns DP, O'Connor KM, Clarke G, O'Halloran KD. The role of NADPH oxidase in chronic intermittent hypoxia-induced respiratory plasticity in adult male mice. Respir Physiol Neurobiol 2021; 292:103713. [PMID: 34116239 DOI: 10.1016/j.resp.2021.103713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/18/2021] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
Reactive oxygen species (ROS) are proposed as mediators of chronic intermittent hypoxia (CIH)-induced respiratory plasticity. We sought to determine if NADPH oxidase 2 (NOX2)-derived ROS underpin CIH-induced maladaptive changes in respiratory control. Adult male mice (C57BL/6 J) were assigned to one of three groups: normoxic controls (sham); chronic intermittent hypoxia-exposed (CIH, 12 cycles/hour, 8 h/day for 14 days); and CIH + apocynin (NOX2 inhibitor, 2 mM) given in the drinking water throughout exposure to CIH. In addition, we studied sham and CIH-exposed NOX2-null mice (B6.129S-CybbTM1Din/J). Whole-body plethysmography was used to measure breathing and metabolic parameters. Ventilation (V̇I/V̇CO2) during normoxia was unaffected by CIH, but apnoea index was increased, which was prevented by apocynin, but not by NOX2 deletion. The ventilatory response to hypercapnia following exposure to CIH was potentiated in NOX2-null mice. Our results reveal ROS-dependent influences on the control of breathing and point to antioxidant intervention as a potential adjunctive therapeutic strategy in respiratory control disorders.
Collapse
Affiliation(s)
- Sarah E Drummond
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - David P Burns
- 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; APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, School of Medicine, College of Medicine & Health, 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
|
13
|
Di Fiore JM, Raffay TM. The relationship between intermittent hypoxemia events and neural outcomes in neonates. Exp Neurol 2021; 342:113753. [PMID: 33984336 DOI: 10.1016/j.expneurol.2021.113753] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/06/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022]
Abstract
This brief review examines 1) patterns of intermittent hypoxemia in extremely preterm infants during early postnatal life, 2) the relationship between neonatal intermittent hypoxemia exposure and outcomes in both human and animal models, 3) potential mechanistic pathways, and 4) future alterations in clinical care that may reduce morbidity. Intermittent hypoxemia events are pervasive in extremely preterm infants (<28 weeks gestation at birth) during early postnatal life. An increased frequency of intermittent hypoxemia events has been associated with a range of poor neural outcomes including language and cognitive delays, motor impairment, retinopathy of prematurity, impaired control of breathing, and intraventricular hemorrhage. Neonatal rodent models have shown that exposure to short repetitive cycles of hypoxia induce a pathophysiological cascade. However, not all patterns of intermittent hypoxia are deleterious and some may even improve neurodevelopmental outcomes. Therapeutic interventions include supplemental oxygen, pressure support and pharmacologic drugs but prolonged hyperoxia and pressure exposure have been associated with cardiopulmonary morbidity. Therefore, it becomes imperative to distinguish high risk from neutral and/or even beneficial patterns of intermittent hypoxemia during early postnatal life. Identification of such patterns could improve clinical care with targeted interventions for high-risk patterns and minimal or no exposure to treatment modalities for low-risk patterns.
Collapse
Affiliation(s)
- Juliann M Di Fiore
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, OH, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States of America.
| | - Thomas M Raffay
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, OH, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
14
|
Taxini CL, Marques DA, Bícego KC, Gargaglioni LH. A5 noradrenergic neurons and breathing control in neonate rats. Pflugers Arch 2021; 473:859-872. [PMID: 33855632 DOI: 10.1007/s00424-021-02550-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/16/2021] [Accepted: 02/25/2021] [Indexed: 10/21/2022]
Abstract
The pontine A5 noradrenergic group contributes to the maturation of the respiratory system before birth in rats. These neurons are connected to the neural network responsible for respiratory rhythmogenesis. In the present study, we investigated the participation of A5 noradrenergic neurons in neonates (P7-8 and P14-15) in the control of ventilation during hypoxia and hypercapnia in in vivo experiments using conjugated saporin anti-dopamine beta-hydroxylase (DβH-SAP) to specifically ablate noradrenergic neurons. Thus, DβH-SAP (420 ng/μL) or saporin (SAP, control) was injected into the A5 region of neonatal male Wistar rats. Hypoxia reduced respiratory variability in control animals; however, A5 lesion prevented this effect in P7-8 rats. Our data suggest that noradrenergic neurons of the A5 region in neonate rats do not participate in the control of ventilation under baseline and hypercapnic conditions, but exert an inhibitory modulation on breathing variability under hypoxic challenge in early life (P7-8).
Collapse
Affiliation(s)
- Camila L Taxini
- Department of Morphology and Animal Physiology - FCAV, São Paulo State University (UNESP), Via de acesso Paulo Donato Castellane s/n, Jaboticabal, SP, 14870-900, Brazil
| | - Danuzia A Marques
- Department of Pediatrics, Centre de Recherche de L'Institut Universitaire de Cardiologie Et de Pneumologie de Québec, Université Laval, Québec, G1V 4G5, Canada
| | - Kênia C Bícego
- Department of Morphology and Animal Physiology - FCAV, São Paulo State University (UNESP), Via de acesso Paulo Donato Castellane s/n, Jaboticabal, SP, 14870-900, Brazil
| | - Luciane H Gargaglioni
- Department of Morphology and Animal Physiology - FCAV, São Paulo State University (UNESP), Via de acesso Paulo Donato Castellane s/n, Jaboticabal, SP, 14870-900, Brazil.
| |
Collapse
|
15
|
Abstract
The development of the control of breathing begins in utero and continues postnatally. Fetal breathing movements are needed for establishing connectivity between the lungs and central mechanisms controlling breathing. Maturation of the control of breathing, including the increase of hypoxia chemosensitivity, continues postnatally. Insufficient oxygenation, or hypoxia, is a major stressor that can manifest for different reasons in the fetus and neonate. Though the fetus and neonate have different hypoxia sensing mechanisms and respond differently to acute hypoxia, both responses prevent deviations to respiratory and other developmental processes. Intermittent and chronic hypoxia pose much greater threats to the normal developmental respiratory processes. Gestational intermittent hypoxia, due to maternal sleep-disordered breathing and sleep apnea, increases eupneic breathing and decreases the hypoxic ventilatory response associated with impaired gasping and autoresuscitation postnatally. Chronic fetal hypoxia, due to biologic or environmental (i.e. high-altitude) factors, is implicated in fetal growth restriction and preterm birth causing a decrease in the postnatal hypoxic ventilatory responses with increases in irregular eupneic breathing. Mechanisms driving these changes include delayed chemoreceptor development, catecholaminergic activity, abnormal myelination, increased astrocyte proliferation in the dorsal respiratory group, among others. Long-term high-altitude residents demonstrate favorable adaptations to chronic hypoxia as do their offspring. Neonatal intermittent hypoxia is common among preterm infants due to immature respiratory systems and thus, display a reduced drive to breathe and apneas due to insufficient hypoxic sensitivity. However, ongoing intermittent hypoxia can enhance hypoxic sensitivity causing ventilatory overshoots followed by apnea; the number of apneas is positively correlated with degree of hypoxic sensitivity in preterm infants. Chronic neonatal hypoxia may arise from fetal complications like maternal smoking or from postnatal cardiovascular problems, causing blunting of the hypoxic ventilatory responses throughout at least adolescence due to attenuation of carotid body fibers responses to hypoxia with potential roles of brainstem serotonin, microglia, and inflammation, though these effects depend on the age in which chronic hypoxia initiates. Fetal and neonatal intermittent and chronic hypoxia are implicated in preterm birth and complicate the respiratory system through their direct effects on hypoxia sensing mechanisms and interruptions to the normal developmental processes. Thus, precise regulation of oxygen homeostasis is crucial for normal development of the respiratory control network. © 2021 American Physiological Society. Compr Physiol 11:1653-1677, 2021.
Collapse
Affiliation(s)
- Gary C. Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Satyan Lakshminrusimha
- Department of Pediatrics, UC Davis Children’s Hospital, UC Davis Health, UC Davis, Davis, California, USA
| | - Girija G. Konduri
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Children’s Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
16
|
Macchione AF, Trujillo V, Anunziata F, Sahonero M, Anastasia A, Abate P, Molina JC. Early ethanol pre-exposure alters breathing patterns by disruptions in the central respiratory network and serotonergic balance in neonate rats. Behav Brain Res 2020; 396:112908. [PMID: 32961215 DOI: 10.1016/j.bbr.2020.112908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 08/04/2020] [Accepted: 09/13/2020] [Indexed: 10/23/2022]
Abstract
Early ethanol exposure alters neonatal breathing plasticity. Respiratory EtOH's effects are attributed to central respiratory network disruptions, particularly in the medullary serotonin (5HT) system. In this study we evaluated the effects of neonatal pre-exposure to low/moderate doses upon breathing rates, activation patterns of brainstem's nuclei and expression of 5HT 2A and 2C receptors. At PD9, breathing frequencies, tidal volumes and apneas were examined in pups pre-exposed to vehicle or ethanol (2.0 g/kg) at PDs 3, 5 and 7. This developmental stage is equivalent to the 3rd human gestational trimester, characterized by increased levels of synaptogenesis. Pups were tested under sobriety or under the state of ethanol intoxication and when subjected to normoxia or hypoxia. Number of c-Fos and 5HT immunolabelled cells and relative mRNA expression of 5HT 2A and 2C receptors were quantified in the brainstem. Under normoxia, ethanol pre-exposed pups exhibited breathing depressions and a high number of apneas. An opposite phenomenon was found in ethanol pre-treated pups tested under hypoxia where an exacerbated hypoxic ventilatory response (HVR) was observed. The breathing depression was associated with an increase in the neural activation levels of the raphe obscurus (ROb) and a high mRNA expression of the 5HT 2A receptor in the brainstem while desactivation of the ROb and high activation levels in the solitary tract nucleus and area postrema were associated to the exacerbated HVR. In summary, early ethanol experience induces respiratory disruptions indicative of sensitization processes. Neuroadaptive changes in central respiratory areas under consideration appear to be strongly associated with changes in their respiratory plasticity.
Collapse
Affiliation(s)
- A F Macchione
- Instituto De Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional De Córdoba, Córdoba, Argentina; Facultad De Psicología, Universidad Nacional De Córdoba, Córdoba, Argentina; Instituto De Investigaciones Psicológicas, IIPsi-CONICET-Universidad Nacional De Córdoba, Córdoba, Argentina.
| | - V Trujillo
- Instituto De Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional De Córdoba, Córdoba, Argentina
| | - F Anunziata
- Instituto De Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional De Córdoba, Córdoba, Argentina
| | - M Sahonero
- Facultad De Psicología, Universidad Nacional De Córdoba, Córdoba, Argentina
| | - A Anastasia
- Instituto De Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional De Córdoba, Córdoba, Argentina; Facultad De Psicología, Universidad Nacional De Córdoba, Córdoba, Argentina
| | - P Abate
- Facultad De Psicología, Universidad Nacional De Córdoba, Córdoba, Argentina; Instituto De Investigaciones Psicológicas, IIPsi-CONICET-Universidad Nacional De Córdoba, Córdoba, Argentina
| | - J C Molina
- Instituto De Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET- Universidad Nacional De Córdoba, Córdoba, Argentina; Facultad De Psicología, Universidad Nacional De Córdoba, Córdoba, Argentina.
| |
Collapse
|
17
|
O'Connor KM, Lucking EF, Bastiaanssen TFS, Peterson VL, Crispie F, Cotter PD, Clarke G, Cryan JF, O'Halloran KD. Prebiotic administration modulates gut microbiota and faecal short-chain fatty acid concentrations but does not prevent chronic intermittent hypoxia-induced apnoea and hypertension in adult rats. EBioMedicine 2020; 59:102968. [PMID: 32861200 PMCID: PMC7475129 DOI: 10.1016/j.ebiom.2020.102968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/22/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Evidence is accruing to suggest that microbiota-gut-brain signalling plays a regulatory role in cardiorespiratory physiology. Chronic intermittent hypoxia (CIH), modelling human sleep apnoea, affects gut microbiota composition and elicits cardiorespiratory morbidity. We investigated if treatment with prebiotics ameliorates cardiorespiratory dysfunction in CIH-exposed rats. METHODS Adult male rats were exposed to CIH (96 cycles/day, 6.0% O2 at nadir) for 14 consecutive days with and without prebiotic supplementation (fructo- and galacto-oligosaccharides) beginning two weeks prior to gas exposures. FINDINGS CIH increased apnoea index and caused hypertension. CIH exposure had modest effects on the gut microbiota, decreasing the relative abundance of Lactobacilli species, but had no effect on microbial functional characteristics. Faecal short-chain fatty acid (SCFA) concentrations, plasma and brainstem pro-inflammatory cytokine concentrations and brainstem neurochemistry were unaffected by exposure to CIH. Prebiotic administration modulated gut microbiota composition and diversity, altering gut-metabolic (GMMs) and gut-brain (GBMs) modules and increased faecal acetic and propionic acid concentrations, but did not prevent adverse CIH-induced cardiorespiratory phenotypes. INTERPRETATION CIH-induced cardiorespiratory dysfunction is not dependant upon changes in microbial functional characteristics and decreased faecal SCFA concentrations. Prebiotic-related modulation of microbial function and resultant increases in faecal SCFAs were not sufficient to prevent CIH-induced apnoea and hypertension in our model. Our results do not exclude the potential for microbiota-gut-brain axis involvement in OSA-related cardiorespiratory morbidity, but they demonstrate that in a relatively mild model of CIH, sufficient to evoke classic cardiorespiratory dysfunction, such changes are not obligatory for the development of morbidity, but may become relevant in the elaboration and maintenance of cardiorespiratory morbidity with progressive disease. FUNDING Department of Physiology and APC Microbiome Ireland, University College Cork, Ireland. APC Microbiome Ireland is funded by Science Foundation Ireland, through the Government's National Development Plan.
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
| | - 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
| | | | - Fiona Crispie
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, 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
|
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
|
Beyeler SA, Hodges MR, Huxtable AG. Impact of inflammation on developing respiratory control networks: rhythm generation, chemoreception and plasticity. Respir Physiol Neurobiol 2020; 274:103357. [PMID: 31899353 DOI: 10.1016/j.resp.2019.103357] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/17/2019] [Accepted: 12/02/2019] [Indexed: 10/25/2022]
Abstract
The respiratory control network in the central nervous system undergoes critical developmental events early in life to ensure adequate breathing at birth. There are at least three "critical windows" in development of respiratory control networks: 1) in utero, 2) newborn (postnatal day 0-4 in rodents), and 3) neonatal (P10-13 in rodents, 2-4 months in humans). During these critical windows, developmental processes required for normal maturation of the respiratory control network occur, thereby increasing vulnerability of the network to insults, such as inflammation. Early life inflammation (induced by LPS, chronic intermittent hypoxia, sustained hypoxia, or neonatal maternal separation) acutely impairs respiratory rhythm generation, chemoreception and increases neonatal risk of mortality. These early life impairments are also greater in young males, suggesting sex-specific impairments in respiratory control. Further, neonatal inflammation has a lasting impact on respiratory control by impairing adult respiratory plasticity. This review focuses on how inflammation alters respiratory rhythm generation, chemoreception and plasticity during each of the three critical windows. We also highlight the need for additional mechanistic studies and increased investigation into how glia (such as microglia and astrocytes) play a role in impaired respiratory control after inflammation. Understanding how inflammation during critical windows of development disrupt respiratory control networks is essential for developing better treatments for vulnerable neonates and preventing adult ventilatory control disorders.
Collapse
Affiliation(s)
- Sarah A Beyeler
- Department of Human Physiology, University of Oregon, Eugene, OR, 97403, United States
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Adrianne G Huxtable
- Department of Human Physiology, University of Oregon, Eugene, OR, 97403, United States.
| |
Collapse
|
20
|
McDonald FB, Dempsey EM, O'Halloran KD. The impact of preterm adversity on cardiorespiratory function. Exp Physiol 2019; 105:17-43. [PMID: 31626357 DOI: 10.1113/ep087490] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the topic of this review? We review the influence of prematurity on the cardiorespiratory system and examine the common sequel of alterations in oxygen tension, and immune activation in preterm infants. What advances does it highlight? The review highlights neonatal animal models of intermittent hypoxia, hyperoxia and infection that contribute to our understanding of the effect of stress on neurodevelopment and cardiorespiratory homeostasis. We also focus on some of the important physiological pathways that have a modulatory role on the cardiorespiratory system in early life. ABSTRACT Preterm birth is one of the leading causes of neonatal mortality. Babies that survive early-life stress associated with immaturity have significant prevailing short- and long-term morbidities. Oxygen dysregulation in the first few days and weeks after birth is a primary concern as the cardiorespiratory system slowly adjusts to extrauterine life. Infants exposed to rapid alterations in oxygen tension, including exposures to hypoxia and hyperoxia, have altered redox balance and active immune signalling, leading to altered stress responses that impinge on neurodevelopment and cardiorespiratory homeostasis. In this review, we explore the clinical challenges posed by preterm birth, followed by an examination of the literature on animal models of oxygen dysregulation and immune activation in the context of early-life stress.
Collapse
Affiliation(s)
- Fiona B McDonald
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland.,Irish Centre for Fetal and Neonatal Translational Research (INFANT) Research Centre, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- Irish Centre for Fetal and Neonatal Translational Research (INFANT) Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics & Child Health, School of Medicine, College of Medicine & Health, Cork University Hospital, Wilton, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland.,Irish Centre for Fetal and Neonatal Translational Research (INFANT) Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
21
|
Bittencourt‐Silva PG, Menezes MF, Mendonça‐Junior BA, Karlen‐Amarante M, Zoccal DB. Postnatal intermittent hypoxia enhances phrenic and reduces vagal upper airway motor activities in rats by epigenetic mechanisms. Exp Physiol 2019; 105:148-159. [DOI: 10.1113/ep087928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/08/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Paloma G. Bittencourt‐Silva
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Miguel Furtado Menezes
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Bolival A. Mendonça‐Junior
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Marlusa Karlen‐Amarante
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Daniel B. Zoccal
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| |
Collapse
|
22
|
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
|
23
|
Abstract
Intermittent hypoxemia (IH) events are common during early postnatal life, particularly in preterm infants. These events have been associated with multiple morbidities, including retinopathy of prematurity, sleep disordered breathing, neurodevelopmental impairment, and mortality. The relationship between IH and poor outcomes may depend on the patterns (frequency, duration, and timing) of the IH events. Current treatment modalities used in the clinical setting have been only partially successful in reducing the incidence of apnea and accompanying IH, but the risks and benefits of more aggressive interventions should include knowledge of the relationship between IH and morbidity.
Collapse
Affiliation(s)
- Juliann M. Di Fiore
- Case Western Reserve University, Rainbow Babies & Children’s Hospital, Division of Neonatology, Suite RBC 3100, Cleveland, OH 44106-6010
| | - Peter M MacFarlane
- Case Western Reserve University, Rainbow Babies & Children's Hospital, 11100 Euclid Avenue, Suite RBC 3100, Cleveland, OH 44106-6010, USA
| | - Richard J Martin
- Case Western Reserve University, Rainbow Babies & Children's Hospital, 11100 Euclid Avenue, Suite RBC 3100, Cleveland, OH 44106-6010, USA
| |
Collapse
|
24
|
Kiernan EA, Wang T, Vanderplow AM, Cherukuri S, Cahill ME, Watters JJ. Neonatal Intermittent Hypoxia Induces Lasting Sex-Specific Augmentation of Rat Microglial Cytokine Expression. Front Immunol 2019; 10:1479. [PMID: 31333645 PMCID: PMC6615134 DOI: 10.3389/fimmu.2019.01479] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023] Open
Abstract
Sleep disordered breathing (SDB) affects 3-5% of the pediatric population, including neonates who are highly susceptible due to an underdeveloped ventilatory control system, and REM-dominated sleep. Although pediatric SDB is associated with poor cognitive outcomes, very little research has focused on models of pediatric SDB, particularly in neonates. In adults and neonates, intermittent hypoxia (IH), a hallmark of SDB, recapitulates multiple physiological aspects of severe SDB, including neuronal apoptosis, sex-specific cognitive deficits, and neuroinflammation. Microglia, resident CNS immune cells, are important mediators of neurodevelopment and neuroinflammation, but to date, no studies have examined the molecular properties of microglia in the context of neonatal IH. Here, we tested the hypothesis that neonatal IH will enhance microglial inflammation and sex-specifically lead to long-term changes in working memory. To test this hypothesis, we exposed post-natal day (P1) neonates with dams to an established adult model of pathological IH consisting of 2 min cycles of 10.5% O2 followed by 21% O2, 8 h/day for 8 days. We then challenged the offspring with bacterial lipopolysaccharide (LPS) at P9 or at 6-8 weeks of age and immunomagnetically isolated microglia for gene expression analyses and RNA-sequencing. We also characterized neonatal CNS myeloid cell populations by flow cytometry analyses. Lastly, we examined working memory performance using a Y-maze in the young adults. Contrary to our hypothesis, we found that neonatal IH acutely augmented basal levels of microglial anti-inflammatory cytokines, attenuated microglial responses to LPS, and sex-specifically altered CNS myeloid populations. We identified multiple sex differences in basal neonatal microglial expression of genes related to chemotaxis, cognition, and aging. Lastly, we found that basal, but not LPS-induced, anti-inflammatory cytokines were augmented sex-specifically in the young adults, and that there was a significant interaction between sex and IH on basal working memory. Our results support the idea that neonates may be able to adapt to IH exposures that are pathological in adults. Further, they suggest that male and female microglial responses to IH are sex-specific, and that these sex differences in basal microglial gene expression may contribute to sexual dimorphisms in vulnerability to IH-induced cognitive disruption.
Collapse
Affiliation(s)
- Elizabeth A Kiernan
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Tao Wang
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Amanda M Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Sneha Cherukuri
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Michael E Cahill
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Jyoti J Watters
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
25
|
Johnson SM, Randhawa KS, Baker TL, Watters JJ. Respiratory frequency plasticity during development. Respir Physiol Neurobiol 2019; 266:54-65. [PMID: 31055188 DOI: 10.1016/j.resp.2019.04.014] [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: 02/13/2019] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 01/20/2023]
Abstract
Respiratory frequency plasticity is a long-lasting increase in breathing frequency due to a perturbation. Mechanisms underlying respiratory frequency are poorly understood, and there is little evidence of frequency plasticity in neonates. This hybrid review/research article discusses available literature regarding frequency plasticity and highlights potential research opportunities. Also, we include data demonstrating a model of frequency plasticity using isolated neonatal rat brainstem-spinal cord preparations. Specifically, substance P (SubP) application induced a long-lasting (>60 min) increase in spontaneous respiratory motor burst frequency, particularly in brainstem-spinal cords with the pons attached; there were no male/female differences. SubP-induced frequency plasticity is dependent on the application pattern, such that intermittent (rather than sustained) SubP applications induce more frequency plasticity. SubP-induced frequency plasticity was blocked by a neurokinin-1 receptor antagonist. Thus, the newborn rat respiratory control system has the capacity to express frequency plasticity. Identifying mechanisms that induce frequency plasticity may lead to novel methods to safely treat breathing disorders in premature and newborn infants.
Collapse
Affiliation(s)
- Stephen M Johnson
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, United States.
| | - Karanbir S Randhawa
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, United States
| | - Tracy L Baker
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, United States
| | - Jyoti J Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, United States
| |
Collapse
|
26
|
Bavis RW, Millström AH, Kim SM, MacDonald CA, O'Toole CA, Asklof K, McDonough AB. Combined effects of intermittent hyperoxia and intermittent hypercapnic hypoxia on respiratory control in neonatal rats. Respir Physiol Neurobiol 2018; 260:70-81. [PMID: 30439529 DOI: 10.1016/j.resp.2018.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/27/2018] [Accepted: 11/08/2018] [Indexed: 01/28/2023]
Abstract
Chronic exposure to intermittent hyperoxia causes abnormal carotid body development and attenuates the hypoxic ventilatory response (HVR) in neonatal rats. We hypothesized that concurrent exposure to intermittent hypercapnic hypoxia would influence this plasticity. Newborn rats were exposed to alternating bouts of hypercapnic hypoxia (10% O2/6% CO2) and hyperoxia (30-40% O2) (5 cycles h-1, 24 h d-1) through 13-14 days of age; the experiment was run twice, once in a background of 21% O2 and once in a background of 30% O2 (i.e., "relative hyperoxia"). Hyperoxia had only small effects on carotid body development when combined with intermittent hypercapnic hypoxia: the carotid chemoafferent response to hypoxia was reduced, but this did not affect the HVR. In contrast, sustained exposure to 30% O2 reduced carotid chemoafferent activity and carotid body size which resulted in a blunted HVR. When given alone, chronic intermittent hypercapnic hypoxia increased carotid body size and reduced the hypercapnic ventilatory response but did not affect the HVR. Overall, it appears that intermittent hypercapnic hypoxia counteracted the effects of hyperoxia on the carotid body and prevented developmental plasticity of the HVR.
Collapse
Affiliation(s)
- Ryan W Bavis
- Department of Biology, Bates College, Lewiston, ME 04240 USA.
| | | | - Song M Kim
- Department of Biology, Bates College, Lewiston, ME 04240 USA
| | | | | | - Kendra Asklof
- Department of Biology, Bates College, Lewiston, ME 04240 USA
| | - Amy B McDonough
- Department of Biology, Bates College, Lewiston, ME 04240 USA
| |
Collapse
|
27
|
Elliot-Portal E, Laouafa S, Arias-Reyes C, Janes TA, Joseph V, Soliz J. Brain-derived erythropoietin protects from intermittent hypoxia-induced cardiorespiratory dysfunction and oxidative stress in mice. Sleep 2018; 41:4985474. [PMID: 29697839 PMCID: PMC6047438 DOI: 10.1093/sleep/zsy072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
Study Objectives Based on the fact that erythropoietin (Epo) administration in rodents protects against spatial learning and cognitive deficits induced by chronic intermittent hypoxia (CIH)-mediated oxidative damage, here we tested the hypothesis that Epo in the brain protects against cardiorespiratory disorders and oxidative stress induced by CIH in adult mice. Methods Adult control and transgenic mice overexpressing Epo in the brain only (Tg21) were exposed to CIH (21%-10% O2-10 cycles/hour-8 hours/day-7 days) or room air. After CIH exposure, we used the tail cuff method to measure arterial pressure, and whole-body plethysmography to assess the frequency of apneic episodes at rest, minute ventilation, and ventilatory responses to hypoxia and hypercapnia. Finally, the activity of pro-oxidant (XO-xanthine oxidase, and NADPH) and antioxidant (super oxide dismutase) enzymes was evaluated in the cerebral cortex and brainstem. Results Exposure of control mice to CIH significantly increased the heart rate and arterial pressure, the number of apneic events, and the ventilatory response to hypoxia and hypercapnia. Furthermore, CIH increased the ratio of pro-oxidant to antioxidant enzymes in cortex and brainstem tissues. Both physiological and molecular changes induced by CIH were prevented in transgenic Tg21 mice. Conclusions We conclude that the neuroprotective effect of Epo prevents oxidative damage in the brain and cardiorespiratory disorders induced by CIH. Considering that Epo is used in clinics to treat chronic kidney disease and stroke, our data show convincing evidence suggesting that Epo may be a promising alternative drug to treat sleep-disorder breathing.
Collapse
Affiliation(s)
- Elizabeth Elliot-Portal
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Sofien Laouafa
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Christian Arias-Reyes
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Tara Adele Janes
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Vincent Joseph
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Jorge Soliz
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
28
|
Macchione AF, Anunziata F, Haymal BO, Abate P, Molina JC. Brief ethanol exposure and stress-related factors disorganize neonatal breathing plasticity during the brain growth spurt period in the rat. Psychopharmacology (Berl) 2018; 235:983-998. [PMID: 29464303 DOI: 10.1007/s00213-017-4815-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/13/2017] [Indexed: 12/24/2022]
Abstract
RATIONALE The effects of early ethanol exposure upon neonatal respiratory plasticity have received progressive attention given a multifactorial perspective related with sudden infant death syndrome or hypoxia-associated syndromes. The present preclinical study was performed in 3-9-day-old pups, a stage in development characterized by a brain growth spurt that partially overlaps with the 3rd human gestational trimester. METHODS Breathing frequencies and apneas were examined in pups receiving vehicle or a relatively moderate ethanol dose (2.0 g/kg) utilizing a whole body plethysmograph. The experimental design also considered possible associations between drug administration stress and exteroceptive cues (plethysmographic context or an artificial odor). Ethanol exposure progressively exerted a detrimental effect upon breathing frequencies. A test conducted at PD9 when pups were under the state of sobriety confirmed ethanol's detrimental effects upon respiratory plasticity (breathing depression). RESULTS Pre-exposure to the drug also resulted in a highly disorganized respiratory response following a hypoxic event, i.e., heightened apneic episodes. Associative processes involving drug administration procedures and placement in the plethysmographic context also affected respiratory plasticity. Pups that experienced intragastric administrations in close temporal contiguity with such a context showed diminished hyperventilation during hypoxia. In a 2nd test conducted at PD9 while pups were intoxicated and undergoing hypoxia, an attenuated hyperventilatory response was observed. In this test, there were also indications that prior ethanol exposure depressed breathing frequencies during hypoxia and a recovery normoxia phase. CONCLUSION As a whole, the results demonstrated that brief ethanol experience and stress-related factors significantly disorganize respiratory patterns as well as arousal responses linked to hypoxia in neonatal rats.
Collapse
Affiliation(s)
- A F Macchione
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.,Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - F Anunziata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - B O Haymal
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - P Abate
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.,Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - J C Molina
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina. .,Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba, Argentina. .,Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, USA.
| |
Collapse
|
29
|
Patrone LGA, Biancardi V, Marques DA, Bícego KC, Gargaglioni LH. Brainstem catecholaminergic neurones and breathing control during postnatal development in male and female rats. J Physiol 2018; 596:3299-3325. [PMID: 29479699 DOI: 10.1113/jp275731] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/21/2018] [Indexed: 01/23/2023] Open
Abstract
KEY POINTS The brainstem catecholaminergic (CA) modulation on ventilation changes with development. We determined the role of the brainstem CA system in ventilatory control under normocapnic and hypercapnic conditions during different phases of development [postnatal day (P)7-8, P14-15 and P20-21] in male and female Wistar rats. Brainstem CA neurones produce a tonic inhibitory drive that affects breathing frequency in P7-8 rats and provide an inhibitory drive during hypercapnic conditions in both males and females at P7-8 and P14-15. In pre-pubertal rats, brainstem CA neurones become excitatory for the CO2 ventilatory response in males but remain inhibitory in females. Diseases such as sudden infant death syndrome, congenital central hypoventilation syndrome and Rett syndrome have been associated with abnormalities in the functioning of CA neurones; therefore, the results of the present study contribute to a better understanding of this system. ABSTRACT The respiratory network undergoes significant development during the postnatal phase, including the maturation of the catecholaminergic (CA) system. However, postnatal development of this network and its effect on the control of pulmonary ventilation ( V̇E ) is not fully understood. We investigated the involvement of brainstem CA neurones in respiratory control during postnatal development [postnatal day (P)7-8, P14-15 and P20-21], in male and female rats, through chemical injury with conjugated saporin anti-dopamine β-hydroxylase (DβH-SAP). Thus, DβH-SAP (420 ng μL-1 ), saporin (SAP) or phosphate buffered solution (PBS) was injected into the fourth ventricle of neonatal Wistar rats of both sexes. V̇E and oxygen consumption were recorded 1 week after the injections in unanaesthetized neonatal and juvenile rats during room air and hypercapnia. The resting ventilation was higher in both male and female P7-8 lesioned rats by 33%, with a decrease in respiratory variability being observed in males. The hypercapnic ventilatory response (HCVR) was altered in male and female lesioned rats at all postnatal ages. At P7-8, the HCVR for males and females was increased by 37% and 30%, respectively. For both sexes at P14-15 rats, the increase in V̇E during hypercapnia was 37% higher for lesioned rats. A sex-specific difference in HCRV was observed at P20-21, with lesioned males showing a 33% decrease, and lesioned females showing an increase of 33%. We conclude that brainstem CA neurones exert a tonic inhibitory effect on V̇E in the early postnatal days of the life of a rat, increase variability in P7-8 males and modulate HCRV during the postnatal phase.
Collapse
Affiliation(s)
- Luis Gustavo A Patrone
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Vivian Biancardi
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Danuzia A Marques
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, Sao Paulo State University - UNESP/FCAV at Jaboticabal, SP, Brazil
| |
Collapse
|
30
|
Uppari NP, Joseph V, Bairam A. Respiratory responses to progesterone and allopregnanolone following chronic caffeine treatment in newborn female rats. Respir Physiol Neurobiol 2017; 240:32-40. [DOI: 10.1016/j.resp.2017.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 12/14/2016] [Accepted: 01/02/2017] [Indexed: 11/16/2022]
|
31
|
Kouchi H, Uppari N, Joseph V, Bairam A. Sex-specific respiratory effects of acute and chronic caffeine administration in newborn rats. Respir Physiol Neurobiol 2017; 240:8-16. [DOI: 10.1016/j.resp.2017.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/19/2017] [Accepted: 01/28/2017] [Indexed: 01/01/2023]
|
32
|
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
|
33
|
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
|
34
|
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]
|
35
|
Cardiorespiratory events in preterm infants: interventions and consequences. J Perinatol 2016; 36:251-8. [PMID: 26583943 DOI: 10.1038/jp.2015.165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 01/09/2023]
Abstract
Stabilization of respiration and oxygenation continues to be one of the main challenges in clinical care of the neonate. Despite aggressive respiratory support including mechanical ventilation, continuous positive airway pressure, oxygen and caffeine therapy to reduce apnea and accompanying intermittent hypoxemia, the incidence of intermittent hypoxemia events continues to increase during the first few months of life. Even with improvements in clinical care, standards for oxygen saturation targeting and modes of respiratory support have yet to be identified in this vulnerable infant cohort. In addition, we are only beginning to explore the association between the incidence and pattern of cardiorespiratory events during early postnatal life and both short- and long-term morbidity including retinopathy of prematurity, growth, sleep-disordered breathing and neurodevelopmental impairment. Part 1 of this review included a summary of lung development and diagnostic methods of cardiorespiratory monitoring. In Part 2 we focus on clinical interventions and the short- and long-term consequences of cardiorespiratory events in preterm infants.
Collapse
|
36
|
McDonald FB, Dempsey EM, O'Halloran KD. Early Life Exposure to Chronic Intermittent Hypoxia Primes Increased Susceptibility to Hypoxia-Induced Weakness in Rat Sternohyoid Muscle during Adulthood. Front Physiol 2016; 7:69. [PMID: 26973537 PMCID: PMC4777899 DOI: 10.3389/fphys.2016.00069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/12/2016] [Indexed: 12/30/2022] Open
Abstract
Intermittent hypoxia is a feature of apnea of prematurity (AOP), chronic lung disease, and sleep apnea. Despite the clinical relevance, the long-term effects of hypoxic exposure in early life on respiratory control are not well defined. We recently reported that exposure to chronic intermittent hypoxia (CIH) during postnatal development (pCIH) causes upper airway muscle weakness in both sexes, which persists for several weeks. We sought to examine if there are persistent sex-dependent effects of pCIH on respiratory muscle function into adulthood and/or increased susceptibility to re-exposure to CIH in adulthood in animals previously exposed to CIH during postnatal development. We hypothesized that pCIH would cause long-lasting muscle impairment and increased susceptibility to subsequent hypoxia. Within 24 h of delivery, pups and their respective dams were exposed to CIH: 90 s of hypoxia reaching 5% O2 at nadir; once every 5 min, 8 h per day for 3 weeks. Sham groups were exposed to normoxia in parallel. Three groups were studied: sham; pCIH; and pCIH combined with adult CIH (p+aCIH), where a subset of the pCIH-exposed pups were re-exposed to the same CIH paradigm beginning at 13 weeks. Following gas exposures, sternohyoid and diaphragm muscle isometric contractile and endurance properties were examined ex vivo. There was no apparent lasting effect of pCIH on respiratory muscle function in adults. However, in both males and females, re-exposure to CIH in adulthood in pCIH-exposed animals caused sternohyoid (but not diaphragm) weakness. Exposure to this paradigm of CIH in adulthood alone had no effect on muscle function. Persistent susceptibility in pCIH-exposed airway dilator muscle to subsequent hypoxic insult may have implications for the control of airway patency in adult humans exposed to intermittent hypoxic stress during early life.
Collapse
Affiliation(s)
- Fiona B McDonald
- Health Sciences Centre, School of Medicine and Medical Science, University College Dublin Dublin, Ireland
| | - Eugene M Dempsey
- Department of Paediatrics and Child Health, Cork University Maternity Hospital and the Irish Centre for Fetal and Neonatal Translational Research, University College Cork Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, University College Cork Cork, Ireland
| |
Collapse
|
37
|
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
|
38
|
Prabhakar NR, Peng YJ, Kumar GK, Nanduri J. Peripheral chemoreception and arterial pressure responses to intermittent hypoxia. Compr Physiol 2016; 5:561-77. [PMID: 25880505 DOI: 10.1002/cphy.c140039] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Carotid bodies are the principal peripheral chemoreceptors for detecting changes in arterial blood oxygen levels, and the resulting chemoreflex is a potent regulator of blood pressure. Recurrent apnea with intermittent hypoxia (IH) is a major clinical problem in adult humans and infants born preterm. Adult patients with recurrent apnea exhibit heightened sympathetic nerve activity and hypertension. Adults born preterm are predisposed to early onset of hypertension. Available evidence suggests that carotid body chemoreflex contributes to hypertension caused by IH in both adults and neonates. Experimental models of IH provided important insights into cellular and molecular mechanisms underlying carotid body chemoreflex-mediated hypertension. This article provides a comprehensive appraisal of how IH affects carotid body function, underlying cellular, molecular, and epigenetic mechanisms, and the contribution of chemoreflex to the hypertension.
Collapse
Affiliation(s)
- Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology for O2 Sensing, Biological Sciences Division, University of Chicago, Illinois, USA
| | | | | | | |
Collapse
|
39
|
Uppari N, Joseph V, Bairam A. Inhibitory respiratory responses to progesterone and allopregnanolone in newborn rats chronically treated with caffeine. J Physiol 2015; 594:373-89. [PMID: 26497835 DOI: 10.1113/jp270914] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/21/2015] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS In premature newborns, recurrent apnoea is systematically treated with caffeine to prevent long-term neurocognitive disorders, but a substantial percentage of apnoea persists particularly in neonates born before 28 weeks of gestation. Progesterone has been proposed as a respiratory stimulant potentially suitable for the treatment of newborn apnoea persistent to caffeine. Accordingly we asked whether acute progesterone administration reduces apnoea frequency in newborn rats treated with caffeine. Surprisingly our results show that in newborn rats treated with caffeine, administration of progesterone inhibits breathing and increases apnoea frequency. Additional experiments showed an enhanced GABAergic inhibitory drive on breathing after caffeine treatment, and that progesterone is converted to allopregnanolone (an allosteric modulator of GABAA receptors) to inhibit breathing. We conclude that combining progesterone and chronic caffeine is not an option in preterm neonates, unless the effects of allopregnanolone can be counteracted. ABSTRACT Caffeine is the main treatment for apnoea in preterm neonates, but its interactions with other respiratory stimulants like progesterone are unknown. We tested the hypothesis that the addition of progesterone to caffeine treatments further stimulates ventilation. Newborn rats were treated with water (control) or caffeine (15 mg kg(-1)) by daily gavage between postnatal day (P)3 and P12. At P4 and P12, we measured apnoea frequency, ventilatory responses and metabolic parameters under both normoxia and hypoxia (12% O2, 20 min) following an acute administration of either saline or progesterone (4 mg kg(-1); i.p.). Progesterone injection increased the serum levels of both progesterone and its neuroactive metabolite allopregnanolone. Progesterone had no effect on ventilation in control rats under normoxia. Progesterone depressed ventilation in P12 caffeine-treated rats under normoxia and hypoxia and increased apnoea frequency in both P4 and P12 rats. Because allopregnanolone is an allosteric modulator of GABAA receptors and caffeine may enhance GABAergic inhibition in newborns, we studied the effects of the GABAA receptor antagonist bicuculline at 0, 1, 2 and 3 mg kg(-1) doses and allopregnanolone (10 mg kg(-1) dose) in P12 rats. In caffeine-treated rats, bicuculline enhanced ventilation, while allopregnanolone decreased ventilation and increased total apnoea time. Progesterone had no effect on ventilation and apnoea frequency in caffeine-treated rats injected with finasteride, which blocks the conversion of progesterone to allopregnanolone. We conclude that combining progesterone and chronic caffeine therapy is not an option for the treatment of persistent apnoea in preterm neonates, unless the effects of allopregnanolone can be counteracted.
Collapse
Affiliation(s)
- NagaPraveena Uppari
- Unité de recherche en périnatologie, Centre Hospitalier Universitaire de Québec, Hôpital Saint-François d'Assise, Département de Pédiatrie, Université Laval, Québec, QC, Canada
| | - Vincent Joseph
- Unité de recherche en périnatologie, Centre Hospitalier Universitaire de Québec, Hôpital Saint-François d'Assise, Département de Pédiatrie, Université Laval, Québec, QC, Canada
| | - Aida Bairam
- Unité de recherche en périnatologie, Centre Hospitalier Universitaire de Québec, Hôpital Saint-François d'Assise, Département de Pédiatrie, Université Laval, Québec, QC, Canada
| |
Collapse
|
40
|
Abstract
Hypoxic episodes are troublesome components of bronchopulmonary dysplasia (BPD) in preterm infants. Immature respiratory control seems to be the major contributor, superimposed on abnormal respiratory function. Relatively short respiratory pauses may precipitate desaturation and bradycardia. This population is predisposed to pulmonary hypertension; it is likely that pulmonary vasoconstriction also plays a role. The natural history has been well-characterized in the preterm population at risk for BPD; however, the consequences are less clear. Proposed associations of intermittent hypoxia include retinopathy of prematurity, sleep disordered breathing, and neurodevelopmental delay. Future study should address whether these associations are causal relationships.
Collapse
Affiliation(s)
- Richard J. Martin
- Case Western Reserve University School of Medicine, Rainbow Babies & Children’s Hospital, 11100 Euclid Avenue, Suite RBC 3100, Cleveland, Ohio 44106-6010, Ph: 216-844-3387/Fax: 216-844-3380
| | - Juliann M. Di Fiore
- Case Western Reserve University, Rainbow Babies & Children’s Hospital, 11100 Euclid Avenue, Suite RBC 3100, Cleveland, Ohio 44106-6010, Ph: 216-368-1245/Fax: 216-844-3380
| | - Michele C. Walsh
- Case Western Reserve University School of Medicine, Rainbow Babies & Children’s Hospital, 11100 Euclid Avenue, Suite RBC 3100, Cleveland, Ohio 44106-6010, Ph: 216-844-3759/Fax: 216-844-3380
| |
Collapse
|
41
|
Magnusson J, Cummings KJ. Plasticity in breathing and arterial blood pressure following acute intermittent hypercapnic hypoxia in infant rat pups with a partial loss of 5-HT neurons. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1273-84. [PMID: 26354844 DOI: 10.1152/ajpregu.00241.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/05/2015] [Indexed: 12/29/2022]
Abstract
The role of serotonin (5-HT) neurons in cardiovascular responses to acute intermittent hypoxia (AIH) has not been studied in the neonatal period. We hypothesized that a partial loss of 5-HT neurons would reduce arterial blood pressure (BP) at rest, increase the fall in BP during hypoxia, and reduce the long-term facilitation of breathing (vLTF) and BP following AIH. We exposed 2-wk-old, 5,7-dihydroxytryptamine-treated and controls to AIH (10% O2; n = 13 control, 14 treated), acute intermittent hypercapnia (5% CO2; n = 12 and 11), or acute intermittent hypercapnic hypoxia (AIHH; 10% O2, 5% CO2; n = 15 and 17). We gave five 5-min challenges of AIH and acute intermittent hypercapnia, and twenty ∼20-s challenges of AIHH to mimic sleep apnea. Systolic BP (sBP), diastolic BP, mean arterial pressure, heart rate (HR), ventilation (V̇e), and metabolic rate (V̇o2) were continuously monitored. 5,7-Dihydroxytryptamine induced an ∼35% loss of 5-HT neurons from the medullary raphe. Compared with controls, pups deficient in 5-HT neurons had reduced resting sBP (∼6 mmHg), mean arterial pressure (∼5 mmHg), and HR (56 beats/min), and experienced a reduced drop in BP during hypoxia. AIHH induced vLTF in both groups, reflected in increased V̇e and V̇e/V̇o2, and decreased arterial Pco2. The sBP of pups deficient in 5-HT neurons, but not controls, was increased 1 h following AIHH. Our data suggest that a relatively small loss of 5-HT neurons compromises resting BP and HR, but has no influence on ventilatory plasticity induced by AIHH. AIHH may be useful for reversing cardiorespiratory defects related to partial 5-HT system dysfunction.
Collapse
Affiliation(s)
- Jennifer Magnusson
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Kevin J Cummings
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
42
|
McDonald FB, Williams R, Sheehan D, O'Halloran KD. Early life exposure to chronic intermittent hypoxia causes upper airway dilator muscle weakness, which persists into young adulthood. Exp Physiol 2015; 100:947-66. [PMID: 26096367 DOI: 10.1113/ep085003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/07/2015] [Accepted: 06/02/2015] [Indexed: 12/31/2022]
Abstract
NEW FINDINGS What is the central question of this study? Chronic intermittent hypoxia (CIH) is a dominant feature of respiratory control disorders, which are common. We sought to examine the effects of exposure to CIH during neonatal development on respiratory muscle form and function in male and female rats. What is the main finding and its importance? Exposure to CIH during neonatal development caused sternohyoid muscle weakness in both sexes; an effect that persisted into young adult life upon return to normoxia. Upper airway dilator muscle dysfunction in vivo could predispose to airway collapse, leading to impaired respiratory homeostasis. Chronic intermittent hypoxia (CIH) is a feature of sleep-disordered breathing, which is very common. Exposure to CIH is associated with aberrant plasticity in the respiratory control system including the final effector organs, the striated muscles of breathing. We reasoned that developmental age and sex are key factors determining the functional response of respiratory muscle to CIH. We tested the hypothesis that exposure to CIH causes persistent impairment of sternohyoid muscle function due to oxidative stress and that males are more susceptible to CIH-induced muscle impairment than females. Wistar rat litters (with respective dams) were exposed to intermittent hypoxia for 12 cycles per hour, 8 h per day for 3 weeks from the first day of life [postnatal day (P) 0]. Sham experiments were run in parallel. Half of each litter was studied on P22; the other half was returned to normoxia and studied on P42. Functional properties of the sternohyoid muscle were determined ex vivo. Exposure to CIH significantly decreased sternohyoid muscle force in both sexes; an effect that persisted into young adult life. Chronic intermittent hypoxia had no effect on sternohyoid muscle endurance. Chronic intermittent hypoxia did not affect sternohyoid myosin fibre type, succinate dehydrogenase or glycerol-3-phosphate dehydrogenase activities, or protein free thiol and carbonyl content. Muscles exposed to CIH had smaller cross-sectional areas, consistent with the observation of muscle weakness. In human infants with disordered breathing, CIH-induced upper airway dilator muscle weakness could increase the propensity for airway narrowing or collapse, which could serve to perpetuate impaired respiratory homeostasis.
Collapse
Affiliation(s)
- Fiona B McDonald
- School of Medicine and Medical Science, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Robert Williams
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| | - David Sheehan
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- School of Medicine and Medical Science, Health Sciences Centre, University College Dublin, Dublin, Ireland.,Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
43
|
Comparative ventilatory strategies of acclimated rats and burrowing plateau pika (Ochotona curzoniae) in response to hypoxic-hypercapnia. Comp Biochem Physiol A Mol Integr Physiol 2015; 187:103-10. [PMID: 25988712 DOI: 10.1016/j.cbpa.2015.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/16/2015] [Accepted: 05/07/2015] [Indexed: 11/22/2022]
Abstract
The objective of this study was to compare the different ventilatory strategies that help in coping with hypoxic-hypercapnia environment among two species: use acclimated rats and plateau pikas (Ochotona curzoniae) that live in Tibetan plateaus, and have been well adjusted to high altitude. Arterial blood samples taken at 4100 m of elevation in acclimatized rats and adapted pikas revealed inter-species differences with lower hemoglobin and hematocrit and higher blood pH in pikas. A linear and significant increase in minute ventilation was observed in pikas, which help them to cope with hypoxic-hypercapnia. Pikas also displayed a high inspiratory drive and an invariant respiratory timing regardless of the conditions. Biochemical analysis revealed that N-methyl-D-aspartate receptor (NMDA) receptor gene and nNOS gene are highly conserved between rats and pikas, however pikas have higher expression of NMDA receptors and nNOS compared to rats at the brainstem level. Taken together, these results suggest that pikas have developed a specific ventilatory pattern supported by a modification of the NMDA/NO ventilatory central pathways to survive in extreme conditions imposed on the Tibetan plateaus. These physiological adaptive strategies help in maintaining a better blood oxygenation despite high CO2 concentration in burrows at high altitude.
Collapse
|
44
|
Bairam A, Uppari N, Mubayed S, Joseph V. An Overview on the Respiratory Stimulant Effects of Caffeine and Progesterone on Response to Hypoxia and Apnea Frequency in Developing Rats. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 860:211-20. [DOI: 10.1007/978-3-319-18440-1_23] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
45
|
Nanduri J, Prabhakar NR. Epigenetic Regulation of Carotid Body Oxygen Sensing: Clinical Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 860:1-8. [PMID: 26303461 PMCID: PMC4870818 DOI: 10.1007/978-3-319-18440-1_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Recurrent apnea with intermittent hypoxia (IH) is a major clinical problem in infants born preterm. Recent epidemiological studies showed that adults who were born preterm exhibit increased incidence of sleep-disordered breathing and hypertension. Thus, apnea of prematurity predisposes individuals to autonomic dysfunction in adulthood. Experimental studies showed that adult rats exposed to IH as neonates exhibit augmented carotid body and adrenal chromaffin cells (AMC) response to hypoxia and irregular breathing with apneas and hypertension. The enhanced hypoxic sensitivity of the carotid body and AMC in adult rats exposed to neonatal IH was associated with increased oxidative stress, decreased expression of genes encoding anti-oxidant enzymes, and increased expression of pro-oxidant enzymes. Epigenetic mechanisms including DNA methylation leads to long-term changes in gene expression. The decreased expression of the Sod2 gene, which encodes the anti-oxidant enzyme, superoxide dismutase 2, was associated with DNA hypermethylation of a single CpG dinucleotide close to the transcription start site. Treating neonatal rats with decitabine, an inhibitor of DNA methylation, during IH exposure prevented the oxidative stress, enhanced hypoxic sensitivity, and autonomic dysfunction in adult rats. These findings suggest that epigenetic mechanisms, especially DNA methylation contributes to neonatal programming of hypoxic sensitivity and the ensuing autonomic dysfunction in adulthood.
Collapse
|
46
|
Potvin C, Rossignol O, Uppari N, Dallongeville A, Bairam A, Joseph V. Reduced hypoxic ventilatory response in newborn mice knocked-out for the progesterone receptor. Exp Physiol 2014; 99:1523-37. [DOI: 10.1113/expphysiol.2014.080986] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Catherine Potvin
- Department of Pediatrics, CR-CHU de Québec; Université Laval; Québec Canada
| | - Orlane Rossignol
- Department of Pediatrics, CR-CHU de Québec; Université Laval; Québec Canada
| | | | | | - Aida Bairam
- Department of Pediatrics, CR-CHU de Québec; Université Laval; Québec Canada
| | - Vincent Joseph
- Department of Pediatrics, CR-CHU de Québec; Université Laval; Québec Canada
| |
Collapse
|
47
|
Mayer CA, Di Fiore JM, Martin RJ, Macfarlane PM. Vulnerability of neonatal respiratory neural control to sustained hypoxia during a uniquely sensitive window of development. J Appl Physiol (1985) 2013; 116:514-21. [PMID: 24371020 DOI: 10.1152/japplphysiol.00976.2013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The first postnatal weeks represent a period of development in the rat during which the respiratory neural control system may be vulnerable to aberrant environmental stressors. In the present study, we investigated whether sustained hypoxia (SH; 11% O2) exposure starting at different postnatal ages differentially modifies the acute hypoxic (HVR) and hypercapnic ventilatory response (HCVR). Three different groups of rat pups were exposed to 5 days of SH, starting at either postnatal age 1 (SH1-5), 11 (SH11-15), or 21 (SH21-25) days. Whole body plethysmography was used to assess the HVR and HCVR the day after SH exposure ended. The primary results indicated that 1) the HVR and HCVR of SH11-15 rats were absent or attenuated (respectively) compared with age-matched rats raised in normoxia; 2) there was a profoundly high (∼84% of pups) incidence of unexplained mortality in the SH11-15 rats; and 3) these phenomena were unique to the SH11-15 group with no comparable effect of the SH exposure on the HVR, HCVR, or mortality in the younger (SH1-5) or older (SH21-25) rats. These results share several commonalities with the risk factors thought to underlie the etiology of sudden infant death syndrome, including 1) a vulnerable neonate; 2) a critical period of development; and 3) an environmental stressor.
Collapse
Affiliation(s)
- C A Mayer
- Department of Pediatrics, Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, Ohio
| | | | | | | |
Collapse
|
48
|
Iqbal W, Ciriello J. Effect of maternal chronic intermittent hypoxia during gestation on offspring growth in the rat. Am J Obstet Gynecol 2013; 209:564.e1-9. [PMID: 23981681 DOI: 10.1016/j.ajog.2013.08.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 07/10/2013] [Accepted: 08/23/2013] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Obstructive sleep apnea, a breathing disorder caused by the repetitive collapse of the upper airway during sleep, results in a state of chronic intermittent hypoxia (CIH). Although the etiology and consequences of CIH are extensively investigated in the adult, the developmental ramifications of this disease process are unknown. DESIGN This study was done to investigate the effect of CIH during gestation on offspring development. Pregnant female Spraque-Dawley rats were exposed to daily CIH throughout the gestational period. RESULTS Postnatal day-1 offspring from CIH mothers were asymmetrically growth restricted, with decreased body weights and elevated brain-weight:liver-weight ratios. Furthermore, CIH newborns had elevated heart- and brain-weight:body weight ratios, and decreased liver-weight:body weight ratios. By adulthood, body weights of growth restricted offspring were significantly greater, as were the liver-weight:body weight ratios. CIH offspring also had greater body fat deposition, were hyperglycemic and had elevated plasma levels of insulin during development into adults. CONCLUSION These data suggest that alteration of the maternal intrauterine environment by gestational CIH effects the long-term development of the offspring and increases the risk of the offspring to metabolic diseases in adulthood.
Collapse
Affiliation(s)
- Waseem Iqbal
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | |
Collapse
|
49
|
Gestational stress promotes pathological apneas and sex-specific disruption of respiratory control development in newborn rat. J Neurosci 2013; 33:563-73. [PMID: 23303936 DOI: 10.1523/jneurosci.1214-12.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Recurrent apneas are important causes of hospitalization and morbidity in newborns. Gestational stress (GS) compromises fetal brain development. Maternal stress and anxiety during gestation are linked to respiratory disorders in newborns; however, the mechanisms remain unknown. Here, we tested the hypothesis that repeated activation of the neuroendocrine response to stress during gestation is sufficient to disrupt the development of respiratory control and augment the occurrence of apneas in newborn rats. Pregnant dams were displaced and exposed to predator odor from days 9 to 19 of gestation. Control dams were undisturbed. Experiments were performed on male and female rats aged between 0 and 4 d old. Apnea frequency decreased with age but was consistently higher in stressed pups than controls. At day 4, GS augmented the proportion of apneas with O(2) desaturations by 12%. During acute hypoxia (12% O(2)), the reflexive increase in breathing augmented with age; however, this response was lower in stressed pups. Instability of respiratory rhythm recorded from medullary preparations decreased with age but was higher in stressed pups than controls. GS reduced medullary serotonin (5-HT) levels in newborn pups by 32%. Bath application of 5-HT and injection of 8-OH-DPAT [(±)-8-hydroxy-2-di-(n-propylamino) tetralin hydrobromide; 5-HT(1A) agonist; in vivo] reduced respiratory instability and apneas; these effects were greater in stressed pups than controls. Sex-specific effects were observed. We conclude that activation of the stress response during gestation is sufficient to disrupt respiratory control development and promote pathological apneas in newborn rats. A deficit in medullary 5-HT contributes to these effects.
Collapse
|
50
|
Julian CG, Vargas E, Gonzales M, Dávila RD, Ladenburger A, Reardon L, Schoo C, Powers RW, Lee-Chiong T, Moore LG. Sleep-disordered breathing and oxidative stress in preclinical chronic mountain sickness (excessive erythrocytosis). Respir Physiol Neurobiol 2013; 186:188-96. [PMID: 23380170 DOI: 10.1016/j.resp.2013.01.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 11/26/2022]
Abstract
Chronic mountain sickness (CMS) is considered to be a loss of ventilatory acclimatization to high altitude (>2500m) resulting in marked arterial hypoxemia and polycythemia. This case-control study explores the possibility that sleep-disordered breathing (SDB) and associated oxidative stress contribute to the etiology of CMS. Nocturnal respiratory and [Formula: see text] patterns were measured using standard polysomnography techniques and compared between male high-altitude residents (aged 18-25) with preclinical CMS (excessive erythrocytosis (EE), n=20) and controls (n=19). Measures of oxidative stress and antioxidant status included isoprostanes (8-iso-PGF2alpha), superoxide dismutase and ascorbic acid. EE cases had a greater apnea-hypopnea index, a higher frequency of apneas (central and obstructive) and hypopneas during REM sleep, and lower nocturnal [Formula: see text] compared to controls. 8-iso-PGF2alpha was greater in EE than controls, negatively associated with nocturnal [Formula: see text] , and positively associated with hemoglobin concentration. Mild sleep-disordered breathing and oxidative stress are evident in preclinical CMS, suggesting that the resolution of nocturnal hypoxemia or antioxidant treatment may prevent disease progression.
Collapse
Affiliation(s)
- Colleen Glyde Julian
- Altitude Research Center, Department of Emergency Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|