1
|
Souza E Silva LF, Siena A, Yuzawa JM, Rosenstock TR. SIRTUINS MODULATORS MITIGATE HYPOXIA-INDUCED CELL DEATH DUE TO CHANGES IN HISTONE 3 ACETYLATION, AND MITOCHONDRIAL FUNCTION, DYNAMICS, AND CONTENT. Neuropharmacology 2025:110484. [PMID: 40315982 DOI: 10.1016/j.neuropharm.2025.110484] [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: 01/21/2025] [Revised: 04/01/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
Hypoxia is a key environmental factor linked to neurodevelopmental complications, primarily through its impact on mitochondrial dysfunction. Given that sirtuins regulate mitochondrial and cellular metabolism, we aimed to investigate whether pharmacological modulation of sirtuins could protect neurons from hypoxia-induced mitochondrial dysfunction and cell death. To explore this, primary cortical neurons from male Wistar rats (control) and Spontaneously Hypertensive Rats (a model for neonatal hypoxia and schizophrenia) were exposed to cobalt chloride (CoCl2) to chemically induce hypoxia. Neurons were also treated with Nicotinamide (50 μM), Resveratrol (0.5 μM), and Sirtinol (5 μM) to modulate sirtuin activity. We first assessed histone deacetylation, cell death, mitochondrial calcium retention capacity, mitochondrial membrane potential, and levels of reactive oxygen species (ROS). In addition, we analysed the expression of genes related to mitochondrial metabolism, dynamics, and biogenesis, as well as high-energy compound levels. Our data indicate that both chemical and neonatal hypoxia caused mitochondrial depolarization, reduced calcium retention, increased ROS levels, and elevated Nfe2l2 expression in primary cortical neurons. Hypoxia also led to increased expression of genes associated with mitochondrial biogenesis and fission, as well as reduced ATP levels and elevated pyruvate and lactate levels. Importantly, treatment with sirtuin modulators enhanced neuron viability, likely by further increasing Nfe2l2 expression and reducing ROS production. These modulators also improved metabolic outcomes, including higher ATP levels, and normalized pyruvate and lactate production, as well as mitochondrial fusion gene expression. Collectively, our findings suggest that sirtuin modulators could mitigate hypoxia-induced damage and may represent a potential therapeutic strategy for managing neurodevelopmental disorders.
Collapse
Affiliation(s)
- Luiz Felipe Souza E Silva
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil; Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Amanda Siena
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jessica Mayumi Yuzawa
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Tatiana Rosado Rosenstock
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil; Department of Bioscience, In-vitro Neuroscience, Sygnature Discovery, Nottingham, United Kingdom.
| |
Collapse
|
2
|
Martinez S, Chen Z, Di Fiore JM, Nguyen C, Minich NM, Hibbs AM. Neonatal intermittent hypoxemia events are associated with later systemic hypertension. Pediatr Res 2025:10.1038/s41390-025-03881-w. [PMID: 39885241 DOI: 10.1038/s41390-025-03881-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/09/2024] [Accepted: 01/05/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Approximately 5% of very premature infants delivered at less than 30 weeks' gestation have systemic hypertension. In adult human and animal models, intermittent hypoxemia events are associated with systemic hypertension. In neonates, intermittent hypoxemia events are associated with adverse outcomes, but it is unknown if they are a risk factor for hypertension. We hypothesize that early intermittent hypoxemia events in very preterm neonates are associated with systemic hypertension at 34-36 weeks' postmenstrual age. METHODS Secondary analysis of a single-center cohort study of 164 infants, <31 weeks' gestational age. Intermittent hypoxemia events were continuously recorded during the first 21 days of age. RESULTS There was a significant association between the number of intermittent hypoxemia events (per 100) and systemic hypertension (OR (95% CI) = 1.08 (1.01-1.15)), and both the number of intermittent hypoxemia events (per 100 β (95% CI) = 0.22 (0.10-0.34)) and percent of time with hypoxemia (β (95% CI) = 0.10 (0.01-0.19)) and systolic blood pressure at 34-36 weeks' postmenstrual age. CONCLUSION This study demonstrated a higher incidence of early intermittent hypoxemia events in preterm infants with hypertension. Decreasing intermittent hypoxemia during this critical period may reduce incidence of later vascular stress in this population. IMPACT Intermittent hypoxemia events are very common in premature infants and increased frequency of intermittent hypoxemia events is associated with morbidity. Intermittent hypoxemia events in adult human as well as adult and neonatal animal models are associated with systemic hypertension. This study demonstrated an association between early intermittent hypoxemia events and systemic hypertension in very preterm neonates, adding to the body of literature of possible morbidities caused by intermittent hypoxemia events. This study addresses the common, though under-recognized, issue of neonatal hypertension, and suggests increased intermittent hypoxemia events may be contributory.
Collapse
Affiliation(s)
- Stephanie Martinez
- Division of Neonatology, University Hospitals of Cleveland, Rainbow Babies & Children's Hospital, Cleveland, OH, USA.
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Zhengyi Chen
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Juliann M Di Fiore
- Division of Neonatology, University Hospitals of Cleveland, Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Christina Nguyen
- Division of Neonatology, University Hospitals of Cleveland, Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Nori M Minich
- Division of Neonatology, University Hospitals of Cleveland, Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Anna Maria Hibbs
- Division of Neonatology, University Hospitals of Cleveland, Rainbow Babies & Children's Hospital, Cleveland, OH, USA
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
3
|
Appiah CB, Gardner JJ, Farmer GE, Cunningham RL, Cunningham JT. Chronic intermittent hypoxia-induced hypertension: the impact of sex hormones. Am J Physiol Regul Integr Comp Physiol 2024; 326:R333-R345. [PMID: 38406843 PMCID: PMC11381015 DOI: 10.1152/ajpregu.00258.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
Obstructive sleep apnea, a common form of sleep-disordered breathing, is characterized by intermittent cessations of breathing that reduce blood oxygen levels and contribute to the development of hypertension. Hypertension is a major complication of obstructive sleep apnea that elevates the risk of end-organ damage. Premenopausal women have a lower prevalence of obstructive sleep apnea and cardiovascular disease than men and postmenopausal women, suggesting that sex hormones play a role in the pathophysiology of sleep apnea-related hypertension. The lack of protection in men and postmenopausal women implicates estrogen and progesterone as protective agents but testosterone as a permissive agent in sleep apnea-induced hypertension. A better understanding of how sex hormones contribute to the pathophysiology of sleep apnea-induced hypertension is important for future research and possible hormone-based interventions. The effect of sex on the pathophysiology of sleep apnea and associated intermittent hypoxia-induced hypertension is of important consideration in the screening, diagnosis, and treatment of the disease and its cardiovascular complications. This review summarizes our current understanding of the impact of sex hormones on blood pressure regulation in sleep apnea with a focus on sex differences.
Collapse
Affiliation(s)
- Cephas B Appiah
- Department of Physiology and Anatomy, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| | - Jennifer J Gardner
- Department of Physiology and Anatomy, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| | - George E Farmer
- Department of Physiology and Anatomy, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, System College of Pharmacy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, School of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, United States
| |
Collapse
|
4
|
Gozal D. Early life postnatal intermittent hypoxia: a case for (Mal)adaptive cardiorespiratory plasticity, inflammation, and epigenetics. Sleep 2023; 46:zsad065. [PMID: 36883695 PMCID: PMC10171623 DOI: 10.1093/sleep/zsad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Indexed: 03/09/2023] Open
Affiliation(s)
- David Gozal
- Department of Child Health and Child Health Research Institute, MU Children’s Hospital, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
5
|
Felipe Souza E Silva L, Siena Dos Santos A, Mayumi Yuzawa J, Luiz de Barros Torresi J, Ziroldo A, Rosado Rosenstock T. SIRTUINS MODULATORS COUNTERACT MITOCHONDRIAL DYSFUNCTION IN CELLULAR MODELS OF HYPOXIA: RELEVANCE TO SCHIZOPHRENIA. Neuroscience 2023:S0306-4522(23)00200-2. [PMID: 37169164 DOI: 10.1016/j.neuroscience.2023.04.027] [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: 10/24/2022] [Revised: 04/16/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Schizophrenia (SZ) is a neurodevelopmental-associated disorder strongly related to environmental factors, such as hypoxia. Because there is no cure for SZ or any pharmacological approach that could revert hypoxia-induced cellular damages, we evaluated whether modulators of sirtuins could abrogate hypoxia-induced mitochondrial deregulation as a neuroprotective strategy. Firstly, astrocytes from control (Wistar) and Spontaneously Hypertensive Rats (SHR), a model of both SZ and neonatal hypoxia, were submitted to chemical hypoxia. Then, cells were exposed to different concentrations of Nicotinamide (NAM), Resveratrol (Resv), and Sirtinol (Sir) for 48hrs. Our data indicate that sirtuins modulation reduces cell death increasing the acetylation of histone 3. This outcome is related to the rescue of loss of mitochondrial membrane potential, changes in mitochondrial calcium buffering capacity, decreased O2-• levels and increased expression of metabolic regulators (Nrf-1 and Nfe2l2) and mitochondrial content. Such findings are relevant not only for hypoxia-associated conditions, named pre-eclampsia but also for SZ since prenatal hypoxia is a relevant environmental factor related to this burdensome neuropsychiatric disorder.
Collapse
Affiliation(s)
- Luiz Felipe Souza E Silva
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Amanda Siena Dos Santos
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jessica Mayumi Yuzawa
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | | | - Alan Ziroldo
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Tatiana Rosado Rosenstock
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil; Dept. of Bioscience, In-vitro Neuroscience, Sygnature Discovery, Nottingham, United Kingdom.
| |
Collapse
|
6
|
Ramirez LA, Mohamed R, Marin T, Brands MW, Snyder E, Sullivan JC. Perinatal intermittent hypoxia increases early susceptibility to ANG II-induced hypertension in adult male but not in female Sprague-Dawley rats. Am J Physiol Renal Physiol 2023; 324:F483-F493. [PMID: 36951371 PMCID: PMC10151053 DOI: 10.1152/ajprenal.00308.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/15/2023] [Accepted: 03/16/2023] [Indexed: 03/24/2023] Open
Abstract
Prenatal, perinatal, and adulthood exposure to chronic intermittent hypoxia (IH) increases blood pressure in rodents. Males exposed to chronic IH have higher blood pressure versus females. However, it is unknown if this same-sex difference exists with acute perinatal IH. We tested the hypothesis that acute perinatal IH increases baseline blood pressure and enhances sensitivity to angiotensin II (ANG II)-induced hypertension in male Sprague-Dawley rats. Male and female pups were randomized to control (room air) or IH (10 min of ∼10% O2 for 3 times/day) for the first 8 days of life. IH decreased oxygen saturation, as confirmed via a pulse oximeter. Pups were weaned at postnatal day 21. Blood pressure was measured via telemetry beginning at 14 wk of age and analyzed separately into light and dark phases to assess circadian rhythm. Osmotic minipumps to deliver ANG II were implanted at 15 wk of age. Perinatal IH exposure did not alter baseline blood pressure. One week of ANG II treatment increased blood pressure in light and dark periods in males exposed to IH versus control; there was no effect in females. Blood pressure among the groups was comparable following 2 wk of ANG II infusion. Perinatal IH did not change the circadian rhythm. Following ANG II treatment, indexes of renal injury were measured. Perinatal IH did not alter kidney size, structure, nephron number, or creatinine clearance. These data indicate that acute perinatal IH enhances early ANG II-induced hypertension in males, independent of nephron loss or decreases in body weight or kidney function.NEW & NOTEWORTHY The impact of acute intermittent hypoxia (IH) in early life on blood pressure in adulthood is unknown. This study used a new model exposing female and male rat pups to acute IH in the first 8 days of life, without exposing the dam. Although baseline blood pressure was not altered in adulthood, IH increased susceptibility to angiotensin II hypertension only in males, supporting increased susceptibility of males exposed to IH to a second cardiovascular stressor.
Collapse
Affiliation(s)
- Lindsey A Ramirez
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Riyaz Mohamed
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Terri Marin
- Department of Nursing Science, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Elizabeth Snyder
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
7
|
Steppan J, Nandakumar K, Wang H, Jang R, Smith L, Kang S, Savage W, Bauer M, Choi R, Brady T, Wodu BP, Scafidi S, Scafidi J, Santhanam L. Neonatal exposure to hypoxia induces early arterial stiffening via activation of lysyl oxidases. Physiol Rep 2023; 11:e15656. [PMID: 37038896 PMCID: PMC10086679 DOI: 10.14814/phy2.15656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 04/12/2023] Open
Abstract
Hypoxia in the neonatal period is associated with early manifestations of adverse cardiovascular health in adulthood including higher risk of hypertension and atherosclerosis. We hypothesize that this occurs due to activation of lysyl oxidases (LOXs) and the remodeling of the large conduit vessels, leading to early arterial stiffening. Newborn C57Bl/6 mice were exposed to hypoxia (FiO2 = 11.5%) from postnatal day 1 (P1) to postnatal day 11 (P11), followed by resumption of normoxia. Controls were maintained in normoxia. Using in vivo (pulse wave velocity; PWV) and ex vivo (tensile testing) arterial stiffness indexes, we determined that mice exposed to neonatal hypoxia had significantly higher arterial stiffness compared with normoxia controls by young adulthood (P60), and it increased further by P120. Echocardiography performed at P60 showed that mice exposed to hypoxia displayed a compensated dilated cardiomyopathy. Western blotting revelated that neonatal hypoxia accelerated age-related increase in LOXL2 protein expression in the aorta and elevated LOXL2 expression in the PA at P11 with a delayed decay toward normoxic controls. In the heart and lung, gene and protein expression of LOX/LOXL2 were upregulated at P11, with a delayed decay when compared to normoxic controls. Neonatal hypoxia results in a significant increase in arterial stiffness in early adulthood due to aberrant LOX/LOXL2 expression. This suggests an acceleration in the mechanical decline of the cardiovascular system, that contributes to increased risk of hypertension in young adults exposed to neonatal hypoxia that may increase susceptibility to further insults.
Collapse
Affiliation(s)
- Jochen Steppan
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Kavitha Nandakumar
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Huilei Wang
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Rosie Jang
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Logan Smith
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Sara Kang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - William Savage
- Department of Chemical and Biomolecular EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Maria Bauer
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Rira Choi
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Travis Brady
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Bulouere Princess Wodu
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Joseph Scafidi
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of PediatricsJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Michael V. Johnston Center for Developmental NeuroscienceKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| |
Collapse
|
8
|
Lai CC, Tang CY, Fu SK, Tseng WC, Tseng KW. Effects of swimming training on myocardial protection in rats. Biomed Rep 2022; 16:19. [PMID: 35251606 PMCID: PMC8850963 DOI: 10.3892/br.2022.1502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/23/2021] [Indexed: 11/07/2022] Open
Abstract
Swimming is important for promoting and maintaining health, as it can increase the efficiency of the cardiovascular system and decrease the occurrence of cardiovascular diseases. The objective of the present study was to examine whether swimming training could decrease myocardial injury in rats caused by myocardial ischemia/reperfusion (I/R). Sprague-Dawley rats were randomized into four groups, namely the Sham, coronary artery occlusion, swimming training and ischemic preconditioning (IPC) groups. Myocardial I/R was induced in anesthetized male Sprague-Dawley rats by a 40-min occlusion followed by a 3-h reperfusion of the left anterior descending coronary artery. The rats were sacrificed after surgery and their hearts were examined. The results demonstrated that the number of TUNEL-positive nuclei and degree of caspase-3 activation were both significantly increased in the myocardium following myocardial I/R in rats, indicating increased cardiomyocyte apoptosis. On the other hand, swimming training decreased the serum levels of creatine phosphokinase, lactate dehydrogenase and cardiac troponin I, and was associated with reduced histological damage and myocardial infarct size. Furthermore, swimming training also reduced TNF-α levels, caspase-3 activation and enhanced Bcl-2 activation, which decreased the number of apoptotic cells in the myocardium. The findings of the present study showed that swimming training and IPC could similarly decrease myocardial injury following myocardial I/R, and may therefore be used as exercise training to effectively prevent myocardial injury.
Collapse
Affiliation(s)
- Chang-Chi Lai
- Department of Exercise and Health Sciences, University of Taipei, Taipei 11153, Taiwan, R.O.C
| | - Chia-Yu Tang
- Department of Physical Education, Chang Gung University, Taoyuan 33302, Taiwan, R.O.C
| | - Szu-Kai Fu
- Graduate Institute of Sports Training, University of Taipei, Taipei 11153, Taiwan, R.O.C
| | - Wei-Chin Tseng
- Department of Exercise and Health Sciences, University of Taipei, Taipei 11153, Taiwan, R.O.C
| | - Kuo-Wei Tseng
- Department of Exercise and Health Sciences, University of Taipei, Taipei 11153, Taiwan, R.O.C
| |
Collapse
|
9
|
Mixed-ligand manganese(II) complexes with 5-phenyltetrazole and polypyridine derivatives: Synthesis, crystal structures and biological activity. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
10
|
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
|
11
|
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
|
12
|
|
13
|
The impact of chronic intermittent hypoxia on hematopoiesis and the bone marrow microenvironment. Pflugers Arch 2016; 468:919-32. [PMID: 26856724 PMCID: PMC4842224 DOI: 10.1007/s00424-016-1797-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/21/2015] [Accepted: 01/26/2016] [Indexed: 12/18/2022]
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent sleep-related breathing disorder which is associated with patient morbidity and an elevated risk of developing hypertension and cardiovascular diseases. There is ample evidence for the involvement of bone marrow (BM) cells in the pathophysiology of cardiovascular diseases but a connection between OSA and modulation of the BM microenvironment had not been established. Here, we studied how chronic intermittent hypoxia (CIH) affected hematopoiesis and the BM microenvironment, in a rat model of OSA. We show that CIH followed by normoxia increases the bone marrow hypoxic area, increases the number of multipotent hematopoietic progenitors (CFU assay), promotes erythropoiesis, and increases monocyte counts. In the BM microenvironment of CIH-subjected animals, the number of VE-cadherin-expressing blood vessels, particularly sinusoids, increased, accompanied by increased smooth muscle cell coverage, while vWF-positive vessels decreased. Molecularly, we investigated the expression of endothelial cell-derived genes (angiocrine factors) that could explain the cellular phenotypes. Accordingly, we observed an increase in colony-stimulating factor 1, vascular endothelium growth factor, delta-like 4, and angiopoietin-1 expression. Our data shows that CIH induces vascular remodeling in the BM microenvironment, which modulates hematopoiesis, increasing erythropoiesis, and circulating monocytes. Our study reveals for the first time the effect of CIH in hematopoiesis and suggests that hematopoietic changes may occur in OSA patients.
Collapse
|
14
|
Abstract
In recent decades, with advances in neonatal intensive care, extremely premature infants are now surviving into adulthood. Epidemiologic data on the health of these ex-premature infants have begun to reveal a concerning motif-that is, prematurity, in and of itself, seems to be a risk factor for cardiovascular and metabolic disease in later adulthood. The mechanisms underlying this increased risk are unclear, but it is believed that both adverse fetal environment and postnatal exposures for a premature infant likely contribute to the developmental programming of disease by altering the normal trajectory of maturation and aging of multiple organ systems. This article specifically focuses on perinatal factors that may affect risk for cardiovascular disease.
Collapse
|
15
|
Efficacy of carvedilol in reversing hypertension induced by chronic intermittent hypoxia in rats. Eur J Pharmacol 2015; 765:58-67. [DOI: 10.1016/j.ejphar.2015.08.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/02/2015] [Accepted: 08/16/2015] [Indexed: 10/23/2022]
|
16
|
Chu A, Gozal D, Cortese R, Wang Y. Cardiovascular dysfunction in adult mice following postnatal intermittent hypoxia. Pediatr Res 2015; 77:425-33. [PMID: 25518007 DOI: 10.1038/pr.2014.197] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/16/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND Ex-premature infants are at higher risk for hypertension and cardiovascular disease as adults, although the mechanisms underlying such increased risks are unknown. We hypothesize that postnatal exposure to intermittent hypoxia (IH) leads to cardiovascular dysfunction in adulthood with alterations of the renin-angiotensin pathway. METHODS Neonatal mice were exposed to IH for 4 wk. At the age of 3 mo, various cardiovascular measurements were obtained. RESULTS IH-exposed mice exhibited higher systolic blood pressure, impaired baroreflex responses, and decreased heart rate variability. Furthermore, IH-exposed mice manifested evidence of endothelial dysfunction, as shown by reduced reperfusion indices after tail vessel occlusion and impaired vasodilatory responses to acetylcholine. CD31(+) endothelial cells isolated from mesenteric arteries of IH-exposed mice expressed higher levels of angiotensin-converting enzyme and reactive oxygen species; plasma angiotensin-II levels were also significantly higher in these animals. In addition, DNA methylation patterns of the Ace1 and the Agt genes in these cells were congruent with their expression patterns. CONCLUSION Our results suggest that exposures to postnatal IH alter the normal development of the renin-angiotensin system and promote the occurrence of cardiovascular dysfunction during adulthood in mice.
Collapse
Affiliation(s)
- Alison Chu
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - David Gozal
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Rene Cortese
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Yang Wang
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| |
Collapse
|
17
|
Diogo LN, Monteiro EC. The efficacy of antihypertensive drugs in chronic intermittent hypoxia conditions. Front Physiol 2014; 5:361. [PMID: 25295010 PMCID: PMC4170135 DOI: 10.3389/fphys.2014.00361] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022] Open
Abstract
Sleep apnea/hypopnea disorders include centrally originated diseases and obstructive sleep apnea (OSA). This last condition is renowned as a frequent secondary cause of hypertension (HT). The mechanisms involved in the pathogenesis of HT can be summarized in relation to two main pathways: sympathetic nervous system stimulation mediated mainly by activation of carotid body (CB) chemoreflexes and/or asphyxia, and, by no means the least important, the systemic effects of chronic intermittent hypoxia (CIH). The use of animal models has revealed that CIH is the critical stimulus underlying sympathetic activity and hypertension, and that this effect requires the presence of functional arterial chemoreceptors, which are hyperactive in CIH. These models of CIH mimic the HT observed in humans and allow the study of CIH independently without the mechanical obstruction component. The effect of continuous positive airway pressure (CPAP), the gold standard treatment for OSA patients, to reduce blood pressure seems to be modest and concomitant antihypertensive therapy is still required. We focus this review on the efficacy of pharmacological interventions to revert HT associated with CIH conditions in both animal models and humans. First, we explore the experimental animal models, developed to mimic HT related to CIH, which have been used to investigate the effect of antihypertensive drugs (AHDs). Second, we review what is known about drug efficacy to reverse HT induced by CIH in animals. Moreover, findings in humans with OSA are cited to demonstrate the lack of strong evidence for the establishment of a first-line antihypertensive regimen for these patients. Indeed, specific therapeutic guidelines for the pharmacological treatment of HT in these patients are still lacking. Finally, we discuss the future perspectives concerning the non-pharmacological and pharmacological management of this particular type of HT.
Collapse
Affiliation(s)
- Lucilia N Diogo
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa Lisboa, Portugal
| | - Emília C Monteiro
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa Lisboa, Portugal
| |
Collapse
|
18
|
Gozal D, Hakim F, Kheirandish-Gozal L. Chemoreceptors, baroreceptors, and autonomic deregulation in children with obstructive sleep apnea. Respir Physiol Neurobiol 2012; 185:177-85. [PMID: 22954503 DOI: 10.1016/j.resp.2012.08.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 08/21/2012] [Accepted: 08/23/2012] [Indexed: 11/17/2022]
Abstract
Obstructive sleep apnea (OSA) is highly prevalent sleep disorder of breathing in both adults and children that is fraught with substantial cardiovascular morbidities, the latter being attributable to a complex interplay between intermittent hypoxia (IH), episodic hypercapnia, recurrent large intra-thoracic pressure swings, and sleep disruption. Alterations in autonomic nervous system function could underlie the perturbations in cardiovascular, neurocognitive, immune, endocrine and metabolic functions that affect many of the patients suffering from OSA. Although these issues have received substantial attention in adults, the same has thus far failed to occur in children, creating a quasi misperception that children are protected. Here, we provide a critical overview of the evidence supporting the presence of autonomic nervous system (ANS) perturbations in children with OSA, draw some parallel assessments to known mechanisms in rodents and adult humans, particularly, peripheral and central chemoreceptor and baroreceptor pathways, and suggest future research directions.
Collapse
Affiliation(s)
- David Gozal
- Department of Pediatrics, Comer Children's Hospital, The University of Chicago, Chicago, IL, USA.
| | | | | |
Collapse
|
19
|
Hakim F, Gozal D, Kheirandish-Gozal L. Sympathetic and catecholaminergic alterations in sleep apnea with particular emphasis on children. Front Neurol 2012; 3:7. [PMID: 22319509 PMCID: PMC3268184 DOI: 10.3389/fneur.2012.00007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 01/10/2012] [Indexed: 01/04/2023] Open
Abstract
Sleep is involved in the regulation of major organ functions in the human body, and disruption of sleep potentially can elicit organ dysfunction. Obstructive sleep apnea (OSA) is the most prevalent sleep disorder of breathing in adults and children, and its manifestations reflect the interactions between intermittent hypoxia, intermittent hypercapnia, increased intra-thoracic pressure swings, and sleep fragmentation, as elicited by the episodic changes in upper airway resistance during sleep. The sympathetic nervous system is an important modulator of the cardiovascular, immune, endocrine and metabolic systems, and alterations in autonomic activity may lead to metabolic imbalance and organ dysfunction. Here we review how OSA and its constitutive components can lead to perturbation of the autonomic nervous system in general, and to altered regulation of catecholamines, both of which then playing an important role in some of the mechanisms underlying OSA-induced morbidities.
Collapse
Affiliation(s)
- Fahed Hakim
- Department of Pediatrics, Comer Children's Hospital, The University of Chicago Chicago, IL, USA
| | | | | |
Collapse
|
20
|
Abstract
The clinical syndrome of obstructive sleep apnea (OSAS) in children is a distinct, yet somewhat overlapping disorder with the condition that occurs in adults, such that the clinical manifestations, polysomnographic findings, diagnostic criteria and treatment approaches need to be considered in an age-specific manner. Childhood OSAS has now become widely recognized as a frequent disorder and as a major public health problem. Pediatric OSAS, particularly when obesity is concurrently present, is associated with substantial end-organ morbidities and increased healthcare utilization. Although adenotonsillectomy (T&A) remains the first line of treatment, evidence in recent years suggests that the outcomes of this surgical procedure may not be as favorable as expected, such that post-T&A polysomnographic evaluation may be needed, especially in high-risk patient groups. In addition, incorporation of nonsurgical approaches for milder forms of the disorder and for residual OSAS after T&A is now being investigated.
Collapse
Affiliation(s)
- Riva Tauman
- Sleep Disorders Center, Dana Children's Hospital, Tel Aviv Medical Center, Tel Aviv University, Tel Aviv 64239, Israel.
| | | |
Collapse
|
21
|
Andersen ML, Alvarenga TF, Mazaro-Costa R, Hachul HC, Tufik S. The association of testosterone, sleep, and sexual function in men and women. Brain Res 2011; 1416:80-104. [PMID: 21890115 DOI: 10.1016/j.brainres.2011.07.060] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 07/29/2011] [Accepted: 07/30/2011] [Indexed: 11/29/2022]
Abstract
Testosterone has been the focus of several investigations and review studies in males, but few have addressed its effects on sleep and sexual function, despite evidence of its androgenic effects on circadian activity in both sexes. Studies have been conducted to understand how sleeping increases (and how waking decreases) testosterone levels and how this rhythm can be related to sexual function. This review addresses the inter-relationships among testosterone, sexual function and sleep, including sleep-disordered breathing in both sexes, specifically its effects related to sleep deprivation. In addition, hormonal changes in testosterone that occur in the gonadal and adrenal axis with obstructive sleep apnea and other conditions of chronic sleep deprivation, and which consequently affect sexual life, have also been explored. Nevertheless, hormone-associated sleep disruptions occur across a lifetime, particularly in women. The association between endogenous testosterone and sex, sleep and sleep disturbances is discussed, including the results of clinical trials as well as animal model studies. Evidence of possible pathophysiological mechanisms underlying this relationship is also described. Unraveling the associations of sex steroid hormone concentrations with sleep and sexual function may have clinical implications, as sleep loss reduces testosterone levels in males, and low sex steroid hormone concentrations have been associated with sexual dysfunction.
Collapse
Affiliation(s)
- Monica L Andersen
- Departmento de Psicobiologia, Universidade Federal de São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
22
|
Shimoda LA, Polak J. Hypoxia. 4. Hypoxia and ion channel function. Am J Physiol Cell Physiol 2011; 300:C951-67. [PMID: 21178108 PMCID: PMC3093942 DOI: 10.1152/ajpcell.00512.2010] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 12/16/2010] [Indexed: 12/19/2022]
Abstract
The ability to sense and respond to oxygen deprivation is required for survival; thus, understanding the mechanisms by which changes in oxygen are linked to cell viability and function is of great importance. Ion channels play a critical role in regulating cell function in a wide variety of biological processes, including neuronal transmission, control of ventilation, cardiac contractility, and control of vasomotor tone. Since the 1988 discovery of oxygen-sensitive potassium channels in chemoreceptors, the effect of hypoxia on an assortment of ion channels has been studied in an array of cell types. In this review, we describe the effects of both acute and sustained hypoxia (continuous and intermittent) on mammalian ion channels in several tissues, the mode of action, and their contribution to diverse cellular processes.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Div. of Pulmonary and Critical Care Medicine, Johns Hopkins University, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA.
| | | |
Collapse
|
23
|
Li RC, Haribabu B, Mathis SP, Kim J, Gozal D. Leukotriene B4 receptor-1 mediates intermittent hypoxia-induced atherogenesis. Am J Respir Crit Care Med 2011; 184:124-31. [PMID: 21493735 DOI: 10.1164/rccm.201012-2039oc] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Obstructive sleep apnea, which is characterized by intermittent hypoxia (IH) during sleep, has emerged as an independent risk factor for cardiovascular disease, including atherosclerosis. Leukotriene B4 (LTB4) production is increased in patients with obstructive sleep apnea and negatively correlates to hypoxic levels during sleep, with continuous positive airway pressure therapy decreasing LTB4 production. OBJECTIVES Determine the potential role of LTB4 in IH-induced atherosclerosis in a monocyte cellular model and a murine model. METHODS THP-1 cells were exposed to IH for 3, 6, 24, and 48 hours. Macrophage transformation and foam cell formation were assessed after IH exposures. Apolipopotein E (ApoE)(-/-) or BLT1(-/-)/ApoE(-/-) mice were fed an atherogenic diet and exposed to IH (alternating 21% and 5.7% O(2) from 7 am to 7 PM each day) for 10 weeks. Atherosclerotic lesion formation in en face aorta was examined by oil red O staining. MEASUREMENTS AND MAIN RESULTS IH increased production of LTB4 and the expression of 5-lipoxygenase and leukotriene A4 hydrolase, the key enzymes for producing LTB4. IH was associated with transformation of monocytes to activated macrophages, as evidenced by increased expression of CD14 and CD68. In addition, IH exposures promoted increased cellular cholesterol accumulation and foam cell formation. The LTB4 receptor 1 (BLT1) antagonist U-75302 markedly attenuated IH-induced changes. Furthermore, IH promoted atherosclerotic lesion formation in ApoE(-/-) mice. IH-induced lesion formation was markedly attenuated in BLT1(-/-)/ApoE(-/-) mice. CONCLUSIONS BLT1-dependent pathways underlie IH-induced atherogenesis, and may become a potential novel therapeutic target for obstructive sleep apnea-associated cardiovascular disease.
Collapse
Affiliation(s)
- Richard C Li
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | |
Collapse
|
24
|
Chen TI, Lai CJ, Hsieh CJ, Tsai KL, Yang KT. Differences in left ventricular cardiomyocyte loss induced by chronic intermittent hypoxia between spontaneously hypertensive and Wistar-Kyoto rats. Sleep Breath 2010; 15:845-54. [PMID: 21136300 DOI: 10.1007/s11325-010-0448-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 11/12/2010] [Accepted: 11/18/2010] [Indexed: 02/07/2023]
Abstract
RATIONALE Chronic intermittent hypoxia (CIH) is thought to induce several cardiovascular effects in patients with obstructive sleep apnoea (OSA). However, the effects of CIH on patients with long-standing hypertension are unknown. PURPOSE This prospective study aimed to investigate the influence of combined OSA and hypertension on cardiomyocyte death. METHODS Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) were exposed to repetitive hypoxia-reoxygenation cycles (30 s of 5% O(2); 45 s of 21% O(2)) or room air for 6 h/day during the light phase (10 a.m.-4 p.m.) for 10, 20, or 30 days, and the levels of necrosis and apoptosis induced in their left ventricular cardiomyocyte were examined. RESULTS CIH increased the accumulation of reactive oxygen species, which induced cardiomyocyte necrosis in WKY and SHR (both p < 0.05). Cardiomyocyte oxidative stress levels by CIH were higher in SHR than in WKY (p < 0.05); therefore, cardiomyocyte necrosis was amplified (p < 0.05). Notably, if a superoxide-scavenging agent is injected beforehand, cardiomyocyte necrosis can be effectively inhibited (p < 0.05). When WKY and SHR are exposed to CIH, increases in mitochondria-released cytochrome c and activation of caspase-3 are found in the cytosolic fraction only in WKY. CONCLUSIONS CIH causes cardiomyocyte loss in SHR mainly through cardiomyocyte necrosis. In WKY however, CIH simultaneously induces apoptosis and necrosis of cardiomyocytes.
Collapse
Affiliation(s)
- Tsung-I Chen
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | | | | | | | | |
Collapse
|
25
|
Xiao D, Longo LD, Zhang L. Role of KATP and L-type Ca2+ channel activities in regulation of ovine uterine vascular contractility: effect of pregnancy and chronic hypoxia. Am J Obstet Gynecol 2010; 203:596.e6-12. [PMID: 20817142 PMCID: PMC2993850 DOI: 10.1016/j.ajog.2010.07.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 07/09/2010] [Accepted: 07/21/2010] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Our objective was to determine whether the pregnancy and high altitude long-term hypoxia-mediated changes in uterine artery contractility were regulated by K(ATP) and L-type Ca(2+) channel activities. STUDY DESIGN Uterine arteries were isolated from nonpregnant and near-term pregnant ewes that had been maintained at sea level (∼300 m) or exposed to high altitude (3801 m) for 110 days. Isometric tension was measured in a tissue bath. RESULTS Pregnancy increased diazoxide, but not verapamil-induced relaxations. Long-term hypoxia attenuated diazoxide-induced relaxations in near-term pregnant uterine arteries, but enhanced verapamil-induced relaxations in nonpregnant uterine arteries. Diazoxide decreased the maximal response (E(max)) of phenylephrine-induced contractions in near-term pregnant uterin arteries but not nonpregnant uterine arteries in normoxic sheep. In contrast, diazoxide had no effect on phenylephrine-induced E(max) in near-term pregnant uterine arteries but decreased it in nonpregnant uterine arteries in long-term hypoxia animals. Verapamil decreased the E(max) and pD(2) (-logEC(50)) of phenylephrine-induced contractions in both nonpregnant uterine arteries and near-term pregnant uterine arteries in normoxic and long-term hypoxia animals, except nonpregnant uterine arteries of normoxic animals in which verapamil showed no effect on the pD(2). CONCLUSION The results suggest that pregnancy selectively increases K(ATP), but not L-type Ca(2+) channel activity. Long-term hypoxia decreases the K(ATP) channel activity, which may contribute to the enhanced uterine vascular myogenic tone observed in pregnant sheep at high altitude hypoxia.
Collapse
Affiliation(s)
- Daliao Xiao
- Center for Perinatal Biology, Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | | | | |
Collapse
|
26
|
Batinić-Haberle I, Rebouças JS, Spasojević I. Superoxide dismutase mimics: chemistry, pharmacology, and therapeutic potential. Antioxid Redox Signal 2010; 13:877-918. [PMID: 20095865 PMCID: PMC2935339 DOI: 10.1089/ars.2009.2876] [Citation(s) in RCA: 406] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oxidative stress has become widely viewed as an underlying condition in a number of diseases, such as ischemia-reperfusion disorders, central nervous system disorders, cardiovascular conditions, cancer, and diabetes. Thus, natural and synthetic antioxidants have been actively sought. Superoxide dismutase is a first line of defense against oxidative stress under physiological and pathological conditions. Therefore, the development of therapeutics aimed at mimicking superoxide dismutase was a natural maneuver. Metalloporphyrins, as well as Mn cyclic polyamines, Mn salen derivatives and nitroxides were all originally developed as SOD mimics. The same thermodynamic and electrostatic properties that make them potent SOD mimics may allow them to reduce other reactive species such as peroxynitrite, peroxynitrite-derived CO(3)(*-), peroxyl radical, and less efficiently H(2)O(2). By doing so SOD mimics can decrease both primary and secondary oxidative events, the latter arising from the inhibition of cellular transcriptional activity. To better judge the therapeutic potential and the advantage of one over the other type of compound, comparative studies of different classes of drugs in the same cellular and/or animal models are needed. We here provide a comprehensive overview of the chemical properties and some in vivo effects observed with various classes of compounds with a special emphasis on porphyrin-based compounds.
Collapse
Affiliation(s)
- Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
27
|
Bhattacharjee R, Gozal D. Cardiovascular disease and sleep disordered breathing: are children vulnerable? Sleep 2009; 32:1251-2. [PMID: 19848352 DOI: 10.1093/sleep/32.10.1251] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Rakesh Bhattacharjee
- Division of Sleep and Respiratory Medicine, Department ofPediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| | | |
Collapse
|
28
|
Abstract
Exposing rodents to brief episodes of hypoxia mimics the hypoxemia and the cardiovascular and metabolic effects observed in patients with obstructive sleep apnoea (OSA), a condition that affects between 5% and 20% of the population. Apart from daytime sleepiness, OSA is associated with a high incidence of systemic and pulmonary hypertension, peripheral vascular disease, stroke and sudden cardiac death. The development of animal models to study sleep apnoea has provided convincing evidence that recurrent exposure to intermittent hypoxia (IH) has significant vascular and haemodynamic impact that explain much of the cardiovascular morbidity and mortality observed in patients with sleep apnoea. However, the molecular and cellular mechanisms of how IH causes these changes is unclear and under investigation. This review focuses on the most recent findings addressing these mechanisms. It includes a discussion of the contribution of the nervous system, circulating and vascular factors, inflammatory mediators and transcription factors to IH-induced cardiovascular disease. It also highlights the importance of reactive oxygen species as a primary mediator of the systemic and pulmonary hypertension that develops in response to exposure to IH.
Collapse
Affiliation(s)
- Laura V González Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM, USA.
| | | | | | | |
Collapse
|
29
|
Abstract
Data from animal and human studies provide a biological plausibility to the notion that obstructive sleep apnea activates pathways that lead to insulin resistance, atherosclerosis and hypertension. Sleep apnea thus activates the same pathways as does obesity. That obstructive sleep apnea is a risk factor for cardiovascular disease is supported by epidemiological association studies. Longitudinal cohort studies also provide evidence that patients with untreated severe sleep apnea have an increased rate of cardiovascular events. But these studies, while highly suggestive, do not provide the evidence needed to convince the skeptic. This would only be obtained by randomized treatment trials with hard cardiovascular endpoints such as cardiac events and deaths. While such studies are in the planning stages, they will be challenging. There are issues about randomizing individuals with severe sleep apnea and excessive sleepiness into no therapy, since they are at known increased risk for car crashes. Thus, lack of therapy puts others on the road at risk as well as the subject with sleep apnea. There is, moreover, the concern that treating obstructive sleep apnea in very obese individuals will have little impact, since any effect of therapy for OSA will be overwhelmed by the effects of obesity itself. Data from randomized treatment trials for cardiovascular endpoints will likely not be available for many years. In the interim, physicians need to consider how to treat such patients. It is proposed that given that CPAP treatment for obstructive sleep apnea is highly effective and essentially totally safe, and that the evidence is suggestive that sleep apnea is a risk factor for cardiovascular disease, then we propose all patients with severe sleep apnea should be treated to reduce cardiovascular risk.
Collapse
|
30
|
Strategic plan for pediatric respiratory diseases research: an NHLBI working group report. Ann Am Thorac Soc 2009; 6:1-10. [PMID: 19131525 DOI: 10.1513/pats.200810-116cb] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Division of Lung Diseases of the National Heart, Lung, and Blood Institute (NHLBI) recently held a workshop to identify gaps in our understanding and treatment of childhood lung diseases and to define strategies to enhance translational research in this field. Leading experts with diverse experience in both laboratory and patient-oriented research reviewed selected areas of pediatric lung diseases, including perinatal programming and epigenetic influences; mechanisms of lung injury, repair, and regeneration; pulmonary vascular disease; sleep and control of breathing; and the application of novel translational methods to enhance personalized medicine. This report summarizes the proceedings of this workshop and provides recommendations for emphasis on targeted areas for future investigation. The priority areas identified for research in pediatric pulmonary diseases included: (1) epigenetic and environmental influences on lung development that program pediatric lung diseases; (2) injury, regeneration, and repair in the developing lung; (3) pulmonary vascular disease in children; (4) development and adaptation of ventilatory responses to postnatal life; (5) nonatopic wheezing: aberrant large airway development or injury?; (6) strategies to improve assessment, diagnosis, and treatment of pediatric respiratory diseases; and (7) predictive and personalized medicine for children.
Collapse
|
31
|
Abman S, Jobe A, Chernick V, Blaisdell C, Castro M, Ramirez MI, Gern JE, Cutting G, Redding G, Hagood JS, Whitsett J, Abman S, Raj JU, Barst R, Kato GJ, Gozal D, Haddad GG, Prabhakar NR, Gauda E, Martinez FD, Tepper R, Wood RE, Accurso F, Teague WG, Venegas J, Cole FS, Wright RJ, Gail D, Hamvas A, Kercsmar C, Kiley J, Weinmann G. Strategic plan for pediatric respiratory diseases research: an NHLBI working group report. Pediatr Pulmonol 2009; 44:2-13. [PMID: 19086051 PMCID: PMC2778243 DOI: 10.1002/ppul.20973] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Division of Lung Diseases of the National Heart, Lung and Blood Institute (NHLBI) recently held a workshop to identify gaps in our understanding and treatment of childhood lung diseases and to define strategies to enhance translational research in this field. Leading experts with diverse experience in both laboratory and patient-oriented research reviewed selected areas of pediatric lung diseases, including perinatal programming and epigenetic influences; mechanisms of lung injury, repair, and regeneration; pulmonary vascular disease (PVD); sleep and control of breathing; and the application of novel translational methods to enhance personalized medicine. This report summarizes the proceedings of this workshop and provides recommendations for emphasis on targeted areas for future investigation. The priority areas identified for research in pediatric pulmonary diseases included: (1) epigenetic and environmental influences on lung development that program pediatric lung diseases, (2) injury, regeneration, and repair in the developing lung, (3) PVD in children, (4) development and adaptation of ventilatory responses to postnatal life, (5) nonatopic wheezing: aberrant large airway development or injury? (6) strategies to improve assessment, diagnosis, and treatment of pediatric respiratory diseases, and (7) predictive and personalized medicine for children.
Collapse
Affiliation(s)
- Steve Abman
- Washington University School of Medicine, Medicine and Pediatrics, St. Louis, Missouri
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW To delineate some of the major morbid phenotypes that have emerged in pediatric obstructive sleep apnea (OSA), address new concepts in our understanding of OSA-associated morbidities, and elaborate on innovative therapeutic schemes that may improve outcomes for this condition. In addition, the conceptual framework whereby a childhood condition such as OSA can be linked to specific adult diseases will be presented. RECENT FINDINGS OSA in children is a frequent condition that affects up to 3% of nonobese, otherwise healthy children. In recent years, increased awareness of OSA and changes in obesity rates in children have contributed to significant changes in disease prevalence and clinical presentation, such that distinct morbidity-related phenotypes have become apparent. Furthermore, oxidative stress and systemic inflammatory pathways are mechanistically involved in the pathophysiology of OSA-associated morbidity. Adenotonsillectomy, the treatment of choice for pediatric OSA, may not be as efficacious as previously thought. Alternative nonsurgical therapies have started to emerge and may become an essential component of treatment. SUMMARY Pediatric OSA, particularly when obesity is concurrently present, is associated with substantial end-organ morbidities that primarily but not exclusively affect central nervous and cardiovascular systems. These morbidities are pathophysiologically mediated by inflammatory and free radical mediators. Although adenotonsillectomy remains the first line of treatment, more critical assessment of its role is needed, and incorporation of nonsurgical approaches to pediatric OSA seems warranted.
Collapse
|