Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.203
1.
Article En | MEDLINE | ID: mdl-38780270

Spinal cord injury is associated with spinal vascular disruptions that result in spinal ischemia and tissue hypoxia. This study evaluated the therapeutic efficacy of normobaric hyperoxia on spinal cord oxygenation and circulatory function at the acute stage of cervical spinal cord injury. Adult male Sprague Dawley rats underwent dorsal cervical laminectomy or cervical spinal cord contusion. At 1-2 days after spinal surgery, spinal cord oxygenation was monitored in anesthetized and spontaneously breathing rats through optical recording of oxygen sensor foils placed on the cervical spinal cord and pulse oximetry. The arterial blood pressure, heart rate, blood gases, and peripheral oxyhemoglobin saturation were also measured under hyperoxic (50% O2) and normoxic (21% O2) conditions. The results showed that contused animals had significantly lower spinal cord oxygenation levels than uninjured animals during normoxia. Peripheral oxyhemoglobin saturation, arterial oxygen partial pressure, and mean arterial blood pressure are significantly reduced following cervical spinal cord contusion. Notably, spinal oxygenation of contused rats could be improved to a level comparable to uninjured animals under hyperoxia. Furthermore, acute hyperoxia elevated blood pressure, arterial oxygen partial pressure, and peripheral oxyhemoglobin saturation. These results suggest that normobaric hyperoxia can significantly improve spinal cord oxygenation and circulatory function in the acute phase after cervical spinal cord injury. We propose that adjuvant normobaric hyperoxia combined with other hemodynamic optimization strategies may prevent secondary damage after spinal cord injury and improve functional recovery.


Hyperoxia , Rats, Sprague-Dawley , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/metabolism , Male , Hyperoxia/physiopathology , Hyperoxia/blood , Rats , Oxygen/blood , Oxygen/metabolism , Spinal Cord/metabolism , Spinal Cord/blood supply , Spinal Cord/physiopathology , Cervical Cord/injuries , Cervical Cord/metabolism , Blood Pressure/physiology , Oxyhemoglobins/metabolism , Heart Rate/physiology
2.
Clin Auton Res ; 34(2): 233-252, 2024 Apr.
Article En | MEDLINE | ID: mdl-38709357

PURPOSE: We conducted a meta-analysis to determine the effect of hyperoxia on muscle sympathetic nerve activity in healthy individuals and those with cardio-metabolic diseases. METHODS: A comprehensive search of electronic databases was performed until August 2022. All study designs (except reviews) were included: population (humans; apparently healthy or with at least one chronic disease); exposures (muscle sympathetic nerve activity during hyperoxia or hyperbaria); comparators (hyperoxia or hyperbaria vs. normoxia); and outcomes (muscle sympathetic nerve activity, heart rate, blood pressure, minute ventilation). Forty-nine studies were ultimately included in the meta-analysis. RESULTS: In healthy individuals, hyperoxia had no effect on sympathetic burst frequency (mean difference [MD] - 1.07 bursts/min; 95% confidence interval [CI] - 2.17, 0.04bursts/min; P = 0.06), burst incidence (MD 0.27 bursts/100 heartbeats [hb]; 95% CI - 2.10, 2.64 bursts/100 hb; P = 0.82), burst amplitude (P = 0.85), or total activity (P = 0.31). In those with chronic diseases, hyperoxia decreased burst frequency (MD - 5.57 bursts/min; 95% CI - 7.48, - 3.67 bursts/min; P < 0.001) and burst incidence (MD - 4.44 bursts/100 hb; 95% CI - 7.94, - 0.94 bursts/100 hb; P = 0.01), but had no effect on burst amplitude (P = 0.36) or total activity (P = 0.90). Our meta-regression analyses identified an inverse relationship between normoxic burst frequency and change in burst frequency with hyperoxia. In both groups, hyperoxia decreased heart rate but had no effect on any measure of blood pressure. CONCLUSION: Hyperoxia does not change sympathetic activity in healthy humans. Conversely, in those with chronic diseases, hyperoxia decreases sympathetic activity. Regardless of disease status, resting sympathetic burst frequency predicts the degree of change in burst frequency, with larger decreases for those with higher resting activity.


Hyperoxia , Muscle, Skeletal , Sympathetic Nervous System , Humans , Hyperoxia/physiopathology , Sympathetic Nervous System/physiology , Sympathetic Nervous System/physiopathology , Muscle, Skeletal/physiology , Muscle, Skeletal/innervation , Heart Rate/physiology
3.
Am J Physiol Heart Circ Physiol ; 326(6): H1544-H1549, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38700471

Numerous studies have shown that oxidative stress plays an important role in peripheral artery disease (PAD). Prior reports suggested autonomic dysfunction in PAD. We hypothesized that responses of the autonomic nervous system and coronary tone would be impaired in patients with PAD during exposure to acute hyperoxia, an oxidative stressor. In 20 patients with PAD and 16 healthy, sex- and age-matched controls, beat-by-beat heart rate (HR, from ECG) and blood pressure (BP, with Finometer) were recorded for 10 min during room air breathing and 5 min of hyperoxia. Cardiovagal baroreflex sensitivity and HR variability (HRV) were evaluated as measures of autonomic function. Transthoracic coronary echocardiography was used to assess peak coronary blood flow velocity (CBV) in the left anterior descending coronary artery. Cardiovagal baroreflex sensitivity at rest was lower in PAD than in healthy controls. Hyperoxia raised BP solely in the patients with PAD, with no change observed in healthy controls. Hyperoxia induced an increase in cardiac parasympathetic activity assessed by the high-frequency component of HRV in healthy controls but not in PAD. Indices of parasympathetic activity were lower in PAD than in healthy controls throughout the trial as well as during hyperoxia. Hyperoxia induced coronary vasoconstriction in both groups, while the coronary perfusion time fraction was lower in PAD than in healthy controls. These results suggest that the response in parasympathetic activity to hyperoxia (i.e., oxidative stress) is blunted and the coronary perfusion time is shorter in patients with PAD.NEW & NOTEWORTHY Patients with peripheral artery disease (PAD) showed consistently lower parasympathetic activity and blunted cardiovagal baroreflex sensitivity compared with healthy individuals. Notably, hyperoxia, which normally boosts parasympathetic activity in healthy individuals, failed to induce this response in patients with PAD. These data suggest altered autonomic responses during hyperoxia in PAD.


Baroreflex , Blood Pressure , Heart Rate , Hyperoxia , Peripheral Arterial Disease , Humans , Male , Female , Hyperoxia/physiopathology , Aged , Peripheral Arterial Disease/physiopathology , Middle Aged , Coronary Circulation , Coronary Vessels/physiopathology , Coronary Vessels/diagnostic imaging , Autonomic Nervous System/physiopathology , Case-Control Studies , Oxidative Stress
4.
Exp Gerontol ; 191: 112435, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38636569

Oxygen supplementation is a widely used treatment for ICU patients. However, it can lead to hyperoxia, which in turn can result in oxidative stress, cardiac remodeling, and even mortality. This paper expands upon previous research conducted by our lab to establish time-dependent cardiac changes under hyperoxia. In this study, both young and aged mice (male and female) underwent 72 h of hyperoxia exposure and were monitored at 24-hour intervals for cardiac electrophysiological and functional parameters using ECG and electrocardiogram data. Our analysis showed that young male mice experienced significant weight loss as well as significant lung edema by 48 h. Although young male mice were highly susceptible to physical changes, they were resistant to early cardiac functional and electrophysiological changes compared to the other groups. Both young and aged female and aged males developed functional impairments by 24 h of hyperoxia exposure. Furthermore, sex and age differences were noted in the onset of electrophysiological changes. While some groups could resist early cardiac remodeling, our data suggests that 72 h of hyperoxia exposure is sufficient to induce significant cardiac remodeling across all age and sex groups. Our data establishes that time-dependent cardiac changes due to oxygen supplementation can have devastating consequences even with short exposure periods. These findings can aid in developing clinical practices for individuals admitted to the ICU by elucidating the impact of aging, sex, and length of stay under mechanical ventilation to limit hyperoxia-induced cardiac remodeling.


Disease Models, Animal , Hyperoxia , Animals , Hyperoxia/physiopathology , Female , Male , Mice , Mice, Inbred C57BL , Sex Factors , Electrocardiography , Age Factors , Aging/physiology , Pulmonary Edema/physiopathology , Oxygen Inhalation Therapy/methods , Heart/physiopathology , Heart/physiology , Time Factors , Ventricular Remodeling/physiology , Oxidative Stress
5.
Front Immunol ; 13: 792716, 2022.
Article En | MEDLINE | ID: mdl-35173718

Prematurity and bronchopulmonary dysplasia (BPD) increase the risk of asthma later in life. Supplemental oxygen therapy is a risk factor for chronic respiratory symptoms in infants with BPD. Hyperoxia induces cell injury and release of damage-associated molecular patterns (DAMPs). Cytoskeletal filamentous actin (F-actin) is a DAMP which binds Clec9a, a C-type lectin selectively expressed on CD103+ dendritic cells (DCs). Co-stimulation of Clec9a and TLR3 induces maximal proinflammatory responses. We have shown that neonatal hyperoxia (a model of BPD) increases lung IL-12+Clec9a+CD103+ DCs, pro-inflammatory responses and airway hyperreactivity following rhinovirus (RV) infection. CD103+ DCs and Clec9a are required for these responses. Hyperoxia increases F-actin levels in bronchoalveolar lavage fluid (BALF). We hypothesized that the F-actin severing protein gelsolin attenuates neonatal hyperoxia-induced Clec9a+CD103+ DC-dependent pro-inflammatory responses to RV and preserves alveolarization. We exposed neonatal mice to hyperoxia and treated them with gelsolin intranasally. Subsequently we inoculated the mice with RV intranasally. Alternatively, we inoculated normoxic neonatal mice with BALF from hyperoxia-exposed mice (hyperoxic BALF), RV and gelsolin. We analyzed lung gene expression two days after RV infection. For in vitro studies, lung CD11c+ cells were isolated from C57BL/6J or Clec9agfp-/- mice and incubated with hyperoxic BALF and RV. Cells were analyzed by flow cytometry. In neonatal mice, gelsolin blocked hyperoxia-induced Il12p40, TNF-α and IFN-γ mRNA and protein expression in response to RV infection. Similar effects were observed when gelsolin was co-administered with hyperoxic BALF and RV. Gelsolin decreased F-actin levels in hyperoxic BALF in vitro and inhibited hyperoxia-induced D103lo DC expansion and inflammation in vivo. Gelsolin also attenuated hyperoxia-induced hypoalveolarization. Further, incubation of lung CD11c+ cells from WT and Clec9agfp-/- mice with hyperoxic BALF and RV, showed Clec9a is required for maximal hyperoxic BALF and RV induced IL-12 expression in CD103+ DCs. Finally, in tracheal aspirates from mechanically ventilated human preterm infants the F-actin to gelsolin ratio positively correlates with FiO2, and gelsolin levels decrease during the first two weeks of mechanical ventilation. Collectively, our findings demonstrate a promising role for gelsolin, administered by inhalation into the airway to treat RV-induced exacerbations of BPD and prevent chronic lung disease.


Bronchopulmonary Dysplasia/drug therapy , Gelsolin/administration & dosage , Hyperoxia/physiopathology , Lectins, C-Type/metabolism , Picornaviridae Infections/drug therapy , Receptors, Immunologic/metabolism , Administration, Inhalation , Animals , Animals, Newborn/metabolism , Antigens, CD/metabolism , Bronchopulmonary Dysplasia/virology , Female , Humans , Infant, Newborn , Integrin alpha Chains/metabolism , Interleukin-12/metabolism , Lectins, C-Type/genetics , Lung/metabolism , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Oxygen Inhalation Therapy/adverse effects , Picornaviridae Infections/virology , Receptors, Immunologic/genetics , Respiratory Function Tests , Rhinovirus/isolation & purification
6.
Int J Mol Sci ; 23(3)2022 Jan 23.
Article En | MEDLINE | ID: mdl-35163176

Premature infants are born with developing lungs burdened by surfactant deficiency and a dearth of antioxidant defense systems. Survival rate of such infants has significantly improved due to advances in care involving mechanical ventilation and oxygen supplementation. However, a significant subset of such survivors develops the chronic lung disease, Bronchopulmonary dysplasia (BPD), characterized by enlarged, simplified alveoli and deformed airways. Among a host of factors contributing to the pathogenesis is oxidative damage induced by exposure of the developing lungs to hyperoxia. Recent data indicate that hyperoxia induces aberrant sphingolipid signaling, leading to mitochondrial dysfunction and abnormal reactive oxygen species (ROS) formation (ROS). The role of sphingolipids such as ceramides and sphingosine 1-phosphate (S1P), in the development of BPD emerged in the last decade. Both ceramide and S1P are elevated in tracheal aspirates of premature infants of <32 weeks gestational age developing BPD. This was faithfully reflected in the murine models of hyperoxia and BPD, where there is an increased expression of sphingolipid metabolites both in lung tissue and bronchoalveolar lavage. Treatment of neonatal pups with a sphingosine kinase1 specific inhibitor, PF543, resulted in protection against BPD as neonates, accompanied by improved lung function and reduced airway remodeling as adults. This was accompanied by reduced mitochondrial ROS formation. S1P receptor1 induced by hyperoxia also aggravates BPD, revealing another potential druggable target in this pathway for BPD. In this review we aim to provide a detailed description on the role played by sphingolipid signaling in hyperoxia induced lung injury and BPD.


Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Lung Injury/metabolism , Sphingolipids/physiology , Airway Remodeling , Animals , Animals, Newborn , Ceramides/metabolism , Disease Models, Animal , Humans , Hyperoxia/metabolism , Hyperoxia/physiopathology , Infant , Infant, Newborn , Lung/pathology , Lung Injury/pathology , Lysophospholipids/metabolism , Methanol/pharmacology , Mice , Oxidative Stress/physiology , Pulmonary Alveoli/metabolism , Pyrrolidines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sulfones/pharmacology
7.
Article En | MEDLINE | ID: mdl-34626804

Aquatic hypoxia is both a naturally-occurring and anthropogenically-generated event. Fish species have evolved different adaptations to cope with hypoxic environments, including gill modifications and air breathing. However, little is known about the molecular mechanisms involved in the respiration of embryonic and larval fishes during critical windows of development. We assessed expression of the genes hif-1α, fih-1, nhe1, epo, gr and il8 using the developing tropical gar as a piscine model during three developmental periods (fertilization to hatch, 1 to 6 days post hatch (dph) and 7 to 12 dph) when exposed to normoxia (~7.43 mg/L DO), hypoxia (~2.5 mg/L DO) or hyperoxia (~9.15 mg/L DO). All genes had higher expression when fish were exposed to either hypoxia or hyperoxia during the first two developmental periods. However, fish continuously exposed to hypoxia had increased expression of the six genes by hatching and 6 dph, and by 12 dph only hif-1α still had increased expression. The middle developmental period was the most hypoxia-sensitive, coinciding with several changes in physiology and morphology. The oldest larvae were the most resilient to gene expression change, with little variation in expression of the six genes compared. This study is the first to relate the molecular response of an air-breathing fish to oxygen availability to developmental critical windows and contributes to our understanding of some molecular responses of developing fish to changes in oxygen availability.


Fish Diseases/genetics , Fishes/genetics , Hyperoxia/veterinary , Hypoxia/veterinary , Animals , Aquaculture , Erythropoietin/genetics , Female , Fish Diseases/physiopathology , Fish Proteins/genetics , Fishes/growth & development , Fishes/physiology , Gene Expression Regulation, Developmental , Hyperoxia/genetics , Hyperoxia/physiopathology , Hypoxia/genetics , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-8/genetics , Male , Receptors, Glucocorticoid/genetics , Respiratory Physiological Phenomena , Sodium-Hydrogen Exchanger 1/genetics
8.
Hypertension ; 79(3): 575-587, 2022 03.
Article En | MEDLINE | ID: mdl-34961326

BACKGROUND: Individuals born preterm present left ventricle changes and increased risk of cardiac diseases and heart failure. The pathophysiology of heart disease after preterm birth is incompletely understood. Mitochondria dysfunction is a hallmark of cardiomyopathy resulting in heart failure. We hypothesized that neonatal hyperoxia in rats, a recognized model simulating preterm birth conditions and resulting in oxygen-induced cardiomyopathy, induce left ventricle mitochondrial changes in juvenile rats. We also hypothesized that humanin, a mitochondrial-derived peptide, would be reduced in young adults born preterm. METHODS: Sprague-Dawley pups were exposed to room air (controls) or 80% O2 at postnatal days 3 to 10 (oxygen-induced cardiomyopathy). We studied left ventricle mitochondrial changes in 4 weeks old males. In a cohort of young adults born preterm (n=55) and age-matched term (n=54), we compared circulating levels of humanin. RESULTS: Compared with controls, oxygen-exposed rats showed smaller left ventricle mitochondria with disrupted integrity on electron microscopy, decreased oxidative phosphorylation, increased glycolysis markers, and reduced mitochondrial biogenesis and abundance. In oxygen-exposed rats, we observed lipid deposits, increased superoxide production (isolated cardiomyocytes), and reduced Nrf2 gene expression. In the cohort, left ventricle ejection fraction and peak global longitudinal strain were similar between groups however humanin levels were lower in preterm and associated with left ventricle ejection fraction and peak global longitudinal strain. CONCLUSIONS: In conclusion, neonatal hyperoxia impaired left ventricle mitochondrial structure and function in juvenile animals. Serum humanin level was reduced in preterm adults. This study suggests that preterm birth-related conditions entail left ventricle mitochondrial alterations that may underlie cardiac changes perpetuated into adulthood. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03261609.


Cardiomyopathies/etiology , Hyperoxia/complications , Mitochondria/metabolism , Premature Birth , Ventricular Dysfunction, Left/etiology , Adolescent , Adult , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Female , Humans , Hyperoxia/metabolism , Hyperoxia/physiopathology , Intracellular Signaling Peptides and Proteins/blood , Male , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , Rats , Rats, Sprague-Dawley , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Young Adult
9.
Invest Ophthalmol Vis Sci ; 62(14): 4, 2021 11 01.
Article En | MEDLINE | ID: mdl-34730793

Purpose: To investigate the retinal vascular response to hyperoxia in patients with primary open-angle glaucoma (POAG) using optical coherence tomography angiography (OCTA). Methods: This prospective study included 27 eyes in 27 patients with POAG and 14 eyes in 14 age- and sex-matched healthy participants. Retinal radial peripapillary capillary (RPC) perfusion was measured by OCTA before and after inhaling oxygen in all participants. Systemic hemodynamic variables were also examined and recorded before and after hyperoxia. Results: Hyperoxia significantly reduced the perfused vessel density (PVD) of RPCs in both healthy controls (baseline and hyperoxia: 54.2 ± 4.1 and 51.0 ± 4.4, respectively, P < 0.001) and patients with POAG (baseline and hyperoxia: 44.7 ± 6.1 and 43.2 ± 5.4, respectively, P = 0.001). However, the changes in peripapillary PVD between the two gas conditions in patients with POAG were significantly lower than in healthy controls, including both the absolute change (baseline-hyperoxia: 1.5 ± 2.0 and 3.2 ± 1.2, respectively, P = 0.006) and relative change (ratio of absolute change and baseline value: 3.0% ± 4.6% and 6.0% ± 2.4%, respectively, P = 0.04). Conclusions: Retinal microvasculature responds to hyperoxia by reducing RPC perfusion in both healthy participants and patients with POAG. However, this vasoreactivity capacity was significantly impaired in patients with POAG.


Glaucoma, Open-Angle/physiopathology , Hyperoxia/physiopathology , Microcirculation/physiology , Optic Disk/blood supply , Retinal Vessels/physiology , Adult , Antihypertensive Agents/therapeutic use , Female , Fluorescein Angiography , Glaucoma, Open-Angle/diagnosis , Glaucoma, Open-Angle/drug therapy , Humans , Intraocular Pressure/physiology , Male , Middle Aged , Nerve Fibers/pathology , Prospective Studies , Retinal Ganglion Cells/pathology , Tomography, Optical Coherence , Visual Acuity/physiology , Visual Fields/physiology
10.
Sci Rep ; 11(1): 23086, 2021 11 29.
Article En | MEDLINE | ID: mdl-34845324

Oxygen supplementation, although a cornerstone of emergency and cardiovascular medicine, often results in hyperoxia, a condition characterized by excessive tissue oxygen which results in adverse cardiac remodeling and subsequent injurious effects to physiological function. Cardiac remodeling is further influenced by various risk factors, including pre-existing conditions and sex. Thus, the purpose of this experiment was to investigate cardiac remodeling in Type I Diabetic (Akita) mice subjected to hyperoxic treatment. Overall, we demonstrated that Akita mice experience distinct challenges from wild type (WT) mice. Specifically, Akita males at both normoxia and hyperoxia showed significant decreases in body and heart weights, prolonged PR, QRS, and QTc intervals, and reduced %EF and %FS at normoxia compared to WT controls. Moreover, Akita males largely resemble female mice (both WT and Akita) with regards to the parameters studied. Finally, statistical analysis revealed hyperoxia to have the greatest influence on cardiac pathophysiology, followed by sex, and finally genotype. Taken together, our data suggest that Type I diabetic patients may have distinct cardiac pathophysiology under hyperoxia compared to uncomplicated patients, with males being at high risk. These findings can be used to enhance provision of care in ICU patients with Type I diabetes as a comorbid condition.


Cardiovascular Diseases/complications , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/physiopathology , Hyperoxia/physiopathology , Animals , Cardiovascular Diseases/etiology , Disease Models, Animal , Echocardiography , Electrophysiology , Female , Heart/physiopathology , Heart Rate , Heterozygote , Male , Mice , Mice, Inbred C57BL , Oxygen , Sex Factors , Treatment Outcome
11.
Sci Rep ; 11(1): 20823, 2021 10 21.
Article En | MEDLINE | ID: mdl-34675332

Peripheral chemoreceptors' (PCh) hyperactivity increases sympathetic tone. An augmented acute ventilatory response to hypoxia, being a marker of PCh oversensitivity, was also identified as a marker of poor prognosis in HF. However, not much is known about the tonic (chronic) influence of PCh on cardio-respiratory parameters. In our study 30 HF patients and 30 healthy individuals were exposed to 100% oxygen for 1 min during which minute ventilation and hemodynamic parameters were non-invasively recorded. Systemic vascular resistance (SVR) and mean arterial pressure (MAP) responses to acute hyperoxia differed substantially between HF and control. In HF hyperoxia caused a significant drop in SVR in early stages with subsequent normalization, while increase in SVR was observed in controls. MAP increased in controls, but remained unchanged in HF. Bilateral carotid bodies excision performed in two HF subjects changed the response to hyperoxia towards the course seen in healthy individuals. These differences may be explained by the domination of early vascular reaction to hyperoxia in HF by vasodilation due to the inhibition of augmented tonic activity of PCh. Otherwise, in healthy subjects the vasoconstrictive action of oxygen remains unopposed. The magnitude of SVR change during acute hyperoxia may be used as a novel method for tonic PCh activity assessment.


Chemoreceptor Cells/pathology , Heart Failure/complications , Hyperoxia/complications , Vascular Resistance , Aged , Female , Heart Failure/pathology , Heart Failure/physiopathology , Hemodynamics , Humans , Hyperoxia/pathology , Hyperoxia/physiopathology , Male , Middle Aged
12.
Article En | MEDLINE | ID: mdl-34455084

Hypoxia and hyperoxia are disparate stressors which can have destructive influences on fish growth and physiology. It is yet to be determined if hypoxia and hyperoxia have a cumulative effect in aquatic ecosystems that affect biological parameters in fish, and to understand if this is associated with gene expression. Here we address whether growth performance and expressions of growth, immune system and stress related genes were affected by hypoxia and hyperoxia in fish. Rainbow trout was chosen as the study organism due to its excellent service as biomonitor. After an acclimatization period, fish were exposed to hypoxia (4.0 ± 0.5 ppm O2), normoxia (7.5 ± 0.5 ppm O2) and hyperoxia (12 ± 1.2 ppm O2) for 28 days. At 6 h, 12 h, 24 h, 48 h, 72 h and 28 days, samples were collected. Hypoxia and hyperoxia negatively affected weight gain (WG), specific growth rate (SGR), survival rate (SR) and feed conversion ratio (FCR). The best WG, SGR, SR and FCR values occurred in fish exposed to normoxia, whereas hypoxia was most suppressive on growth and hyperoxia showed intermediate suppression of these parameters. Gene expression analyses were performed in liver and results revealed that long term exposure caused reduced growth hormone-I (GH-I) and insulin like growth factor I-II (IGF I-II) levels in both hypoxia and hyperoxia-treated fish. Heat shock protein (HSP70) levels increased in both hypoxia and hyperoxia treatment, and both exposures caused elevation of leptin (LEP) expression in long-term exposure. Overall data indicate that both hypoxia and hyperoxia cause stress in rainbow trout and negatively affects growth parameters.


Hyperoxia/metabolism , Hypoxia/metabolism , Oncorhynchus mykiss/metabolism , Oxygen/metabolism , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation , Growth Hormone/genetics , Growth Hormone/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hyperoxia/genetics , Hyperoxia/immunology , Hyperoxia/physiopathology , Hypoxia/genetics , Hypoxia/immunology , Hypoxia/physiopathology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Leptin/genetics , Leptin/metabolism , Liver/metabolism , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/growth & development , Oncorhynchus mykiss/immunology , Stress, Physiological , Weight Gain
13.
Physiol Res ; 70(4): 489-499, 2021 08 31.
Article En | MEDLINE | ID: mdl-34062074

The use of oxygen therapy (high doses of oxygen - hyperoxia) in the treatment of premature infants results in their survival. However, it also results in a high incidence of chronic lung disease known as bronchopulmonary dysplasia, a disease in which airway hyper-responsiveness and pulmonary hypertension are well known as consequences. In our previous studies, we have shown that hyperoxia causes airway hyper-reactivity, characterized by an increased constrictive and impaired airway smooth muscle relaxation due to a reduced release of relaxant molecules such as nitric oxide, measured under in vivo and in vitro conditions (extra- and intrapulmonary) airways. In addition, the relaxation pathway of the vasoactive intestinal peptide (VIP) and/or pituitary adenylate cyclase activating peptide (PACAP) is another part of this system that plays an important role in the airway caliber. Peptide, which activates VIP cyclase and pituitary adenylate cyclase, has prolonged airway smooth muscle activity. It has long been known that VIP inhibits airway smooth muscle cell proliferation in a mouse model of asthma, but there is no data about its role in the regulation of airway and tracheal smooth muscle contractility during hyperoxic exposure of preterm newborns.


Bronchopulmonary Dysplasia/etiology , Hyperoxia/etiology , Infant, Premature , Lung/metabolism , Muscle, Smooth/metabolism , Oxygen Inhalation Therapy/adverse effects , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Premature Birth , Vasoactive Intestinal Peptide/metabolism , Airway Remodeling , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/physiopathology , Disease Models, Animal , Gestational Age , Humans , Hyperoxia/metabolism , Hyperoxia/physiopathology , Infant, Newborn , Lung/physiopathology , Muscle, Smooth/physiopathology , Signal Transduction
14.
BMC Cardiovasc Disord ; 21(1): 210, 2021 04 26.
Article En | MEDLINE | ID: mdl-33902450

BACKGROUND: Little is known regarding the effect of cardiopulmonary bypass (CPB) reoxygenation on cardiac function following tetralogy of Fallot repair. We hypothesized that hyperoxic reoxygenation would be more strongly associated with myocardial dysfunction in children with tetralogy of Fallot. METHODS: We investigated the association of perfusate oxygenation (PpO2) associated with myocardial dysfunction among children aged 6-72 months who underwent complete repair of tetralogy of Fallot in 2012-2018. Patients were divided into two groups: lower PpO2 group (≤ 250 mmHg) and higher PpO2 (> 250 mmHg) group based on the highest value of PpO2 during aortic occlusion. The odd ratio (ORs) and 95% confidence intervals (CIs) were estimated by logistic regression models. RESULTS: This study included 163 patients perfused with lower PpO2 and 213 with higher PpO2, with median age at surgery 23.3 (interquartile range [IQR] 12.5-39.4) months, 164 female (43.6%), and median body mass index 15.59 (IQR 14.3-16.9) kg/m2. After adjustment for baseline, clinical and procedural variables, patients with higher PpO2 were associated with higher risk of myocardial dysfunction than those with lower PpO2 (OR 1.770; 95% CI 1.040-3.012, P = 0.035). Higher PpO2, lower SpO2, lower pulmonary annular Z-score, and longer CPB time were independent risk factors for myocardial dysfunction. CONCLUSIONS: Association exists between higher PpO2 and myocardial dysfunction risk in patients with tetralogy of Fallot, highlighting the modulation of reoxygenation during aortic occlusion to reduce cardiovascular damage following tetralogy of Fallot repair. TRIAL REGISTRATION: Clinical Trials. gov number NCT03568357. June 26, 2018.


Cardiac Surgical Procedures/adverse effects , Cardiomyopathies/etiology , Cardiopulmonary Bypass/adverse effects , Hyperoxia/etiology , Tetralogy of Fallot/surgery , Cardiomyopathies/blood , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Child , Child, Preschool , Female , Humans , Hyperoxia/blood , Hyperoxia/diagnosis , Hyperoxia/physiopathology , Infant , Male , Myocardium/metabolism , Myocardium/pathology , Oxygen Saturation , Retrospective Studies , Risk Assessment , Risk Factors , Tetralogy of Fallot/diagnostic imaging , Tetralogy of Fallot/physiopathology , Treatment Outcome
15.
Nat Commun ; 12(1): 1565, 2021 03 10.
Article En | MEDLINE | ID: mdl-33692365

During late lung development, alveolar and microvascular development is finalized to enable sufficient gas exchange. Impaired late lung development manifests as bronchopulmonary dysplasia (BPD) in preterm infants. Single-cell RNA sequencing (scRNA-seq) allows for assessment of complex cellular dynamics during biological processes, such as development. Here, we use MULTI-seq to generate scRNA-seq profiles of over 66,000 cells from 36 mice during normal or impaired lung development secondary to hyperoxia with validation of some of the findings in lungs from BPD patients. We observe dynamic populations of cells, including several rare cell types and putative progenitors. Hyperoxia exposure, which mimics the BPD phenotype, alters the composition of all cellular compartments, particularly alveolar epithelium, stromal fibroblasts, capillary endothelium and macrophage populations. Pathway analysis and predicted dynamic cellular crosstalk suggest inflammatory signaling as the main driver of hyperoxia-induced changes. Our data provides a single-cell view of cellular changes associated with late lung development in health and disease.


Hyperoxia/genetics , Hyperoxia/physiopathology , Lung/metabolism , Lung/pathology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Genotype , Male , Mice
16.
Physiol Rep ; 9(3): e14590, 2021 02.
Article En | MEDLINE | ID: mdl-33565273

The pulmonary endothelium is an immediate recipient of high oxygen concentrations upon oxygen therapy and mediates down-stream responses. Cyclic collapse and reopening of atelectatic lung areas during mechanical ventilation with high fractions of inspired oxygen result in the propagation of oxygen oscillations in the hypoxic/hyperoxic range. We used primary murine lung endothelial cell cultures to investigate cell responses to constant and oscillating oxygen conditions in the hypoxic to hyperoxic range. Severe constant hyperoxia had pro-inflammatory and cytotoxic effects including an increase in expression of ICAM1, E-selectin, and RAGE at 24 hr exposure. The coagulative/fibrinolytic system responded by upregulation of uPA, tPA, and vWF and PAI1 under constant severe hyperoxia. Among antioxidant enzymes, the upregulation of SOD2, TXN1, TXNRD3, GPX1, and Gstp1 at 24 hr, but downregulation of SOD3 at 72 hr constant hyperoxia was evident. Hypoxic/hyperoxic oscillating oxygen conditions induced pro-inflammatory cytokine release to a lesser extent and later than constant hyperoxia. Gene expression analyses showed upregulation of NFKB p65 mRNA at 72 hr. More evident was a biphasic response of NOS3 and ACE1 gene expression (downregulation until 24 hr and upregulation at 72 hr). ACE2 mRNA was upregulated until 72 hr, but shedding of the mature protein from the cell surface favored ACE1. Oscillations resulted in severe production of peroxynitrite, but apart from upregulation of Gstp1 at 24 hr responses of antioxidative proteins were less pronounced than under constant hyperoxia. Oscillating oxygen in the hypoxic/hyperoxic range has a characteristical impact on vasoactive mediators like NOS3 and on the activation of the renin-angiotensin system in the lung endothelium.


Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hyperoxia/metabolism , Hypoxia/metabolism , Lung/blood supply , Oxygen/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Blood Coagulation , Cell Hypoxia , Cells, Cultured , Cytokines/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Hyperoxia/pathology , Hyperoxia/physiopathology , Hypoxia/pathology , Hypoxia/physiopathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Necrosis , Renin-Angiotensin System , Time Factors
17.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L688-L704, 2021 05 01.
Article En | MEDLINE | ID: mdl-33502939

Early therapeutic effect of intratracheally (IT)-administered extracellular vesicles secreted by mesenchymal stem cells (MSC-EVs) has been demonstrated in a rat model of bronchopulmonary dysplasia (BPD) involving hyperoxia exposure in the first 2 postnatal weeks. The aim of this study was to evaluate the protective effects of IT-administered MSC-EVs in the long term. EVs were produced from MSCs following GMP standards. At birth, rats were distributed in three groups: (a) animals raised in ambient air for 6 weeks (n = 10); and animals exposed to 60% hyperoxia for 2 weeks and to room air for additional 4 weeks and treated with (b) IT-administered saline solution (n = 10), or (c) MSC-EVs (n = 10) on postnatal days 3, 7, 10, and 21. Hyperoxia exposure produced significant decreases in total number of alveoli, total surface area of alveolar air spaces, and proliferation index, together with increases in mean alveolar volume, mean linear intercept and fibrosis percentage; all these morphometric changes were prevented by MSC-EVs treatment. The medial thickness index for <100 µm vessels was higher for hyperoxia-exposed/sham-treated than for normoxia-exposed rats; MSC-EV treatment significantly reduced this index. There were no significant differences in interstitial/alveolar and perivascular F4/8-positive and CD86-positive macrophages. Conversely, hyperoxia exposure reduced CD163-positive macrophages both in interstitial/alveolar and perivascular populations and MSC-EV prevented these hyperoxia-induced reductions. These findings further support that IT-administered EVs could be an effective approach to prevent/treat BPD, ameliorating the impaired alveolarization and pulmonary artery remodeling also in a long-term model. M2 macrophage polarization could play a role through anti-inflammatory and proliferative mechanisms.


Bronchopulmonary Dysplasia/complications , Disease Models, Animal , Extracellular Vesicles/physiology , Lung Injury/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Administration, Inhalation , Animals , Animals, Newborn , Female , Hyperoxia/physiopathology , Lung Injury/etiology , Lung Injury/pathology , Male , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Trachea
18.
Pediatr Res ; 90(4): 776-783, 2021 10.
Article En | MEDLINE | ID: mdl-33469177

BACKGROUND: Perinatal antibiotic treatment alters intestinal microbiota and augments hyperoxia-induced lung injury in mice offspring. The effect of maternal antibiotic treatment (MAT) during pregnancy on the lung microbiota and its relationship with lung injury remains unknown. METHODS: We fed timed-pregnant C57BL/6N mice sterile drinking water containing antibiotics from gestational day 15 to delivery. Neonatal mice were reared in either room air (RA) or hyperoxia (85% O2) from postnatal days 1 to 7. Four study groups were obtained: control + RA, control + O2, MAT + RA, and MAT + O2. On postnatal day 7, lung and intestinal microbiota were sampled from the left lung and lower gastrointestinal tract. The right lung was harvested for histology and cytokine analysis. RESULTS: MAT during pregnancy significantly reduced the total number of commensal bacteria in the intestine and birth body weight of newborn mice compared with control newborn mice. Neonatal hyperoxia exposure impaired alveolarization and angiogenesis, which was exacerbated by MAT. Neonatal hyperoxia altered the composition and diversity of intestinal and lung microbiota and MAT further exacerbated neonatal hyperoxia-induced intestinal and lung dysbiosis. CONCLUSIONS: MAT during pregnancy exacerbates hyperoxia-induced lung injury probably through the modulation of intestinal and lung microbiota in neonatal mice. IMPACT: MAT during pregnancy reduced the total number of commensal bacteria in the intestine. Neonatal hyperoxia altered the composition and diversity of intestinal and lung microbiota. MAT exacerbated neonatal hyperoxia-induced intestinal and lung dysbiosis. Neonatal hyperoxia exposure impaired alveolarization and angiogenesis, which was exacerbated by MAT. Avoiding and carefully using antibiotics during pregnancy is a potential therapeutic target for preventing lung injury in hyperoxia-exposed infants.


Hyperoxia/physiopathology , Lung Injury/etiology , Lung/microbiology , Maternal Exposure , Microbiota/drug effects , Animals , Animals, Newborn , Birth Weight , Female , Gastrointestinal Microbiome/drug effects , Hyperoxia/complications , Mice , Mice, Inbred C57BL , Survival Rate
19.
Am J Physiol Lung Cell Mol Physiol ; 320(3): L451-L466, 2021 03 01.
Article En | MEDLINE | ID: mdl-33404366

Supplemental O2 (hyperoxia) is necessary for preterm infant survival but is associated with development of bronchial airway hyperreactivity and childhood asthma. Understanding early mechanisms that link hyperoxia to altered airway structure and function are key to developing advanced therapies. We previously showed that even moderate hyperoxia (50% O2) enhances intracellular calcium ([Ca2+]i) and proliferation of human fetal airway smooth muscle (fASM), thereby facilitating bronchoconstriction and remodeling. Here, we introduce cellular clock biology as a novel mechanism linking early oxygen exposure to airway biology. Peripheral, intracellular clocks are a network of transcription-translation feedback loops that produce circadian oscillations with downstream targets highly relevant to airway function and asthma. Premature infants suffer circadian disruption whereas entrainment strategies improve outcomes, highlighting the need to understand relationships between clocks and developing airways. We hypothesized that hyperoxia impacts clock function in fASM and that the clock can be leveraged to attenuate deleterious effects of O2 on the developing airway. We report that human fASM express core clock machinery (PER1, PER2, CRY1, ARNTL/BMAL1, CLOCK) that is responsive to dexamethasone (Dex) and altered by O2. Disruption of the clock via siRNA-mediated PER1 or ARNTL knockdown alters store-operated calcium entry (SOCE) and [Ca2+]i response to histamine in hyperoxia. Effects of O2 on [Ca2+]i are rescued by driving expression of clock proteins, via effects on the Ca2+ channels IP3R and Orai1. These data reveal a functional fASM clock that modulates [Ca2+]i regulation, particularly in hyperoxia. Harnessing clock biology may be a novel therapeutic consideration for neonatal airway diseases following prematurity.


Bronchi/metabolism , Bronchial Hyperreactivity/metabolism , Calcium/metabolism , Circadian Clocks , Hyperoxia/physiopathology , Muscle, Smooth/metabolism , Oxygen/metabolism , Animals , Bronchi/pathology , Bronchial Hyperreactivity/pathology , Cell Proliferation , Cells, Cultured , Female , Fetus/metabolism , Fetus/pathology , Humans , Infant, Newborn , Male , Mice, Inbred C57BL , Muscle, Smooth/pathology
20.
Am J Physiol Lung Cell Mol Physiol ; 320(2): L193-L204, 2021 02 01.
Article En | MEDLINE | ID: mdl-33112186

Premature infants, especially those with bronchopulmonary dysplasia (BPD), develop recurrent severe respiratory viral illnesses. We have shown that hyperoxic exposure of immature mice, a model of BPD, increases lung IL-12-producing Clec9a+ CD103+ dendritic cells (DCs), pro-inflammatory responses, and airway hyperreactivity following rhinovirus (RV) infection. However, the requirement for CD103+ DCs and Clec9a, a DAMP receptor that binds necrotic cell cytoskeletal filamentous actin (F-actin), for RV-induced inflammatory responses has not been demonstrated. To test this, 2-day-old C57BL/6J, CD103+ DC-deficient Batf3-/- or Clec9agfp-/- mice were exposed to normoxia or hyperoxia for 14 days. Also, selected mice were treated with neutralizing antibody against CD103. Immediately after hyperoxia, the mice were inoculated with RV intranasally. We found that compared with wild-type mice, hyperoxia-exposed Batf3-/- mice showed reduced levels of IL-12p40, IFN-γ, and TNF-α, fewer IFN-γ-producing CD4+ T cells, and decreased airway responsiveness following RV infection. Similar effects were observed in anti-CD103-treated and Clec9agfp-/- mice. Furthermore, hyperoxia increased airway dead cell number and extracellular F-actin levels. Finally, studies in preterm infants with respiratory distress syndrome showed that tracheal aspirate CLEC9A expression positively correlated with IL12B expression, consistent with the notion that CLEC9A+ cells are responsible for IL-12 production in humans as well as mice. We conclude that CD103+ DCs and Clec9a are required for hyperoxia-induced pro-inflammatory responses to RV infection. In premature infants, Clec9a-mediated activation of CD103+ DCs may promote pro-inflammatory responses to viral infection, thereby driving respiratory morbidity.


Antigens, CD/metabolism , Dendritic Cells/immunology , Hyperoxia/physiopathology , Integrin alpha Chains/metabolism , Lectins, C-Type/physiology , Lung/immunology , Pneumonia/immunology , Receptors, Immunologic/physiology , Respiratory Distress Syndrome, Newborn/immunology , Animals , Animals, Newborn , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/physiology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Integrin alpha Chains/genetics , Lung/metabolism , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Picornaviridae Infections/complications , Picornaviridae Infections/virology , Pneumonia/virology , Repressor Proteins/physiology , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Rhinovirus/isolation & purification
...