Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Respir Physiol Neurobiol ; 185(1): 94-104, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22640932

ABSTRACT

Preterm infants often experience hyperoxia while receiving supplemental oxygen. Prolonged exposure to hyperoxia during development is associated with pathologies such as bronchopulmonary dysplasia and retinopathy of prematurity. Over the last 25 years, however, experiments with animal models have revealed that moderate exposures to hyperoxia (e.g., 30-60% O(2) for days to weeks) can also have profound effects on the developing respiratory control system that may lead to hypoventilation and diminished responses to acute hypoxia. This plasticity, which is generally inducible only during critical periods of development, has a complex time course that includes both transient and permanent respiratory deficits. Although the molecular mechanisms of hyperoxia-induced plasticity are only beginning to be elucidated, it is clear that many of the respiratory effects are linked to abnormal morphological and functional development of the carotid body, the principal site of arterial O(2) chemoreception for respiratory control. Specifically, developmental hyperoxia reduces carotid body size, decreases the number of chemoafferent neurons, and (at least transiently) diminishes the O(2) sensitivity of individual carotid body glomus cells. Recent evidence suggests that hyperoxia may also directly or indirectly impact development of the central neural control of breathing. Collectively, these findings emphasize the vulnerability of the developing respiratory control system to environmental perturbations.


Subject(s)
Carotid Body/embryology , Hyperoxia/physiopathology , Animals , Humans
2.
Respir Physiol Neurobiol ; 180(2-3): 193-203, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22138179

ABSTRACT

Rats reared in hyperoxia have smaller carotid bodies as adults. To study the time course and mechanisms underlying these changes, rats were reared in 60% O(2) from birth and their carotid bodies were harvested at various postnatal ages (P0-P7, P14). The carotid bodies of hyperoxia-reared rats were smaller than those of age-matched controls beginning at P4. In contrast, 7d of 60% O(2) had no effect on carotid body size in rats exposed to hyperoxia as adults. Bromodeoxyuridine (BrdU) and TdT-mediated dUTP nick end labeling (TUNEL) were used to assess cell proliferation and DNA fragmentation at P2, P4, and P6. Hyperoxia reduced the proportion of glomus cells undergoing cell division at P4; although a similar trend was evident at P2, hyperoxia no longer affected cell proliferation by P6. The proportion of TUNEL-positive glomus cells was modestly increased by hyperoxia. We did not detect changes in mRNA expression for proapoptotic (Bax) or antiapoptotic (Bcl-X(L)) genes or transcription factors that regulate cell cycle checkpoints (p53 or p21), although mRNA levels for cyclin B1 and cyclin B2 were reduced. Collectively, these data indicate that hyperoxia primarily attenuates postnatal growth of the carotid body by inhibiting glomus cell proliferation during the first few days of exposure.


Subject(s)
Carotid Body/growth & development , Carotid Body/pathology , Hyperoxia/pathology , Animals , Animals, Newborn , Antimetabolites , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Blood Vessels/anatomy & histology , Bromodeoxyuridine , Carotid Body/blood supply , Cell Death/physiology , Cell Proliferation/drug effects , Chronic Disease , DNA Fragmentation/drug effects , Female , Gene Expression/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Lung/cytology , Lung/physiology , Pregnancy , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Regional Blood Flow/physiology , p21-Activated Kinases/biosynthesis
3.
Respir Physiol Neurobiol ; 177(3): 342-6, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21601659

ABSTRACT

Chronic postnatal hyperoxia attenuates the hypoxic ventilatory response (HVR) of rats. To determine whether the ability to detect deficits in the HVR depends on the degree of hypoxia, we assessed the HVR at several levels of hypoxia in adult rats reared in 60% O(2) for the first two postnatal weeks. Hyperoxia-treated rats exhibited smaller increases in ventilation than control rats at 12% O(2) (30±8 vs. 53±4% baseline, mean±SEM; P=0.02) but not at 10% O(2) (83±11 vs. 96±14% baseline; P=0.47). Interestingly, 10% O(2) was used as the test gas in the only study to assess HVR in mice exposed to developmental hyperoxia, and that study reported normal HVR (Dauger et al., Chest 123 (2003), 530-538). Therefore, we assessed the HVR at 12.5% O(2) in adult mice reared in 60% O(2) for the first two postnatal weeks. Hyperoxia-treated mice exhibited smaller increases in ventilation (28±7 vs. 58±8% baseline; P<0.01) and smaller carotid bodies than control mice. We conclude that hyperoxia impairs the HVR in both rats and mice, but this effect is most evident at moderate levels of hypoxia.


Subject(s)
Hyperoxia/physiopathology , Respiratory Mechanics/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Blood Gas Analysis , Blood Volume/physiology , Carotid Body/blood supply , Female , Male , Mice , Mice, Inbred C57BL , Plethysmography/methods , Pulmonary Ventilation , Rats , Rats, Sprague-Dawley , Wakefulness
4.
Respir Physiol Neurobiol ; 177(1): 47-55, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21420511

ABSTRACT

Chronic postnatal hyperoxia blunts the hypoxic ventilatory response (HVR) in rats, an effect that persists for months after return to normoxia. To determine whether decreased carotid body O(2) sensitivity contributes to this lasting impairment, single-unit chemoafferent nerve and glomus cell calcium responses to hypoxia were recorded from rats reared in 60% O(2) through 7d of age (P7) and then returned to normoxia. Single-unit nerve responses were attenuated by P4 and remained low through P7. After return to normoxia, hypoxic responses were partially recovered within 3d and fully recovered within 7-8d (i.e., at P14-15). Glomus cell calcium responses recovered with a similar time course. Hyperoxia altered carotid body mRNA expression for O(2)-sensitive K(+) channels TASK-1, TASK-3, and BK(Ca), but only TASK-1 mRNA paralleled changes in chemosensitivity (i.e., downregulation by P7, partial recovery by P14). Collectively, these data do not support a role for reduced O(2) sensitivity of individual chemoreceptor cells in long-lasting reduction of the HVR after developmental hyperoxia.


Subject(s)
Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Hyperoxia/metabolism , Animals , Animals, Newborn , Calcium Signaling/physiology , Carotid Body/physiopathology , Hyperoxia/physiopathology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Nerve Tissue Proteins/biosynthesis , Patch-Clamp Techniques , Potassium Channels/biosynthesis , Potassium Channels, Tandem Pore Domain/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
5.
Respir Physiol Neurobiol ; 175(2): 220-7, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21094282

ABSTRACT

Chronic exposure to hyperoxia alters the postnatal development and innervation of the rat carotid body. We hypothesized that this plasticity is related to changes in the expression of neurotrophic factors or related proteins. Rats were reared in 60% O(2) from 24 to 36h prior to birth until studied at 3d of age (P3). Protein levels for brain-derived neurotrophic factor (BDNF) were significantly reduced (-70%) in the P3 carotid body, while protein levels for its receptor, tyrosine kinase B, and for glial cell line-derived neurotrophic factor (GDNF) were unchanged. Transcript levels in the carotid body were downregulated for the GDNF receptor Ret (-34%) and the neuropeptide Vgf (-67%), upregulated for Cbln1 (+205%), and unchanged for Fgf2; protein levels were not quantified for these genes. Immunohistochemical analysis revealed that Vgf and Cbln1 proteins are expressed within the carotid body glomus cells. These data suggest that BDNF, and perhaps other neurotrophic factors, contribute to abnormal carotid body function following perinatal hyperoxia.


Subject(s)
Carotid Body/metabolism , Hyperoxia/metabolism , Nerve Growth Factors/metabolism , Animals , Animals, Newborn , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkB/metabolism
6.
Respir Physiol Neurobiol ; 160(3): 301-12, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18069076

ABSTRACT

Perinatal hyperoxia attenuates the hypoxic ventilatory response in rats by altering development of the carotid body and its chemoafferent neurons. In this study, we tested the hypothesis that hyperoxia elicits this plasticity through the increased production of reactive oxygen species (ROS). Rats were born and raised in 60% O(2) for the first two postnatal weeks while treated with one of two antioxidants: vitamin E (via milk from mothers whose diet was enriched with 1000 IU vitamin E kg(-1)) or a superoxide dismutase mimetic, manganese(III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride (MnTMPyP; via daily intraperitoneal injection of 5-10 mg kg(-1)); rats were subsequently raised in room air until studied as adults. Peripheral chemoreflexes, assessed by carotid sinus nerve responses to cyanide, asphyxia, anoxia and isocapnic hypoxia (vitamin E experiments) or by hypoxic ventilatory responses (MnTMPyP experiments), were reduced after perinatal hyperoxia compared to those of normoxia-reared controls (all P<0.01); antioxidant treatment had no effect on these responses. Similarly, the carotid bodies of hyperoxia-reared rats were only one-third the volume of carotid bodies from normoxia-reared controls (P <0.001), regardless of antioxidant treatment. Protein carbonyl concentrations in the blood plasma, measured as an indicator of oxidative stress, were not increased in neonatal rats (2 and 8 days of age) exposed to 60% O(2) from birth. Collectively, these data do not support the hypothesis that perinatal hyperoxia impairs peripheral chemoreceptor development through ROS-mediated oxygen toxicity.


Subject(s)
Antioxidants/administration & dosage , Carotid Sinus/drug effects , Hyperoxia/pathology , Hyperoxia/prevention & control , Metalloporphyrins/administration & dosage , Analysis of Variance , Animals , Animals, Newborn , Anorexia/physiopathology , Anorexia/prevention & control , Asphyxia/physiopathology , Asphyxia/prevention & control , Dose-Response Relationship, Drug , Drug Interactions , Phrenic Nerve/drug effects , Phrenic Nerve/physiopathology , Protein Carbonylation/drug effects , Rats , Rats, Sprague-Dawley , Sodium Cyanide/pharmacology , Vitamin E/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...