Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Int J Mol Sci ; 24(8)2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37108039

ABSTRACT

Obstructive sleep apnea (OSA) is an emerging risk factor for cancer occurrence and progression, mainly mediated by intermittent hypoxia (IH). Systemic IH, a main landmark of OSA, and local sustained hypoxia (SH), a classical feature at the core of tumors, may act separately or synergistically on tumor cells. Our aim was to compare the respective consequences of intermittent and sustained hypoxia on HIF-1, endothelin-1 and VEGF expression and on cell proliferation and migration in HepG2 liver tumor cells. Wound healing, spheroid expansion, proliferation and migration were evaluated in HepG2 cells following IH or SH exposure. The HIF-1α, endothelin-1 and VEGF protein levels and/or mRNA expression were assessed, as were the effects of HIF-1 (acriflavine), endothelin-1 (macitentan) and VEGF (pazopanib) inhibition. Both SH and IH stimulated wound healing, spheroid expansion and proliferation of HepG2 cells. HIF-1 and VEGF, but not endothelin-1, expression increased with IH exposure but not with SH exposure. Acriflavine prevented the effects of both IH and SH, and pazopanib blocked those of IH but not those of SH. Macitentan had no impact. Thus, IH and SH stimulate hepatic cancer cell proliferation via distinct signaling pathways that may act synergistically in OSA patients with cancer, leading to enhanced tumor progression.


Subject(s)
Sleep Apnea, Obstructive , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Hep G2 Cells , Acriflavine , Hypoxia/metabolism , Sleep Apnea, Obstructive/metabolism , Hypoxia-Inducible Factor 1 , Cell Proliferation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
3.
Antioxidants (Basel) ; 11(8)2022 Jul 27.
Article in English | MEDLINE | ID: mdl-36009181

ABSTRACT

RATIONALE: Intermittent hypoxia (IH) is one of the main features of sleep-disordered breathing (SDB). Recent findings indicate that hypoxia inducible factor-1 (HIF-1) promotes cardiomyocytes apoptosis during chronic IH, but the mechanisms involved remain to be elucidated. Here, we hypothesize that IH-induced ER stress is associated with mitochondria-associated ER membrane (MAM) alteration and mitochondrial dysfunction, through HIF-1 activation. METHODS: Right atrial appendage biopsies from patients with and without SDB were used to determine HIF-1α, Grp78 and CHOP expressions. Wild-type and HIF-1α+/- mice were exposed to normoxia (N) or IH (21-5% O2, 60 cycles/h, 8 h/day) for 21 days. Expressions of HIF-1α, Grp78 and CHOP, and apoptosis, were measured by Western blot and immunochemistry. In isolated cardiomyocytes, we examined structural integrity of MAM by proximity ligation assay and their function by measuring ER-to-mitochondria Ca2+ transfer by confocal microscopy. Finally, we measured mitochondrial respiration using oxygraphy and calcium retention capacity (CRC) by spectrofluorometry. MAM structure was also investigated in H9C2 cells incubated with 1 mM CoCl2, a potent HIF-1α inducer. RESULTS: In human atrial biopsies and mice, IH induced HIF-1 activation, ER stress and apoptosis. IH disrupted MAM, altered Ca2+ homeostasis, mitochondrial respiration and CRC. Importantly, IH had no effect in HIF-1α+/- mice. Similar to what observed under IH, HIF-1α overexpression was associated with MAM alteration in H9C2. CONCLUSION: IH-induced ER stress, MAM alterations and mitochondrial dysfunction were mediated by HIF-1; all these intermediate mechanisms ultimately inducing cardiomyocyte apoptosis. This suggests that HIF-1 modulation might limit the deleterious cardiac effects of SDB.

4.
Sci Rep ; 12(1): 12916, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35902610

ABSTRACT

The association between obstructive sleep apnea (OSA) and cancer is still debated and data are scarce regarding the link between OSA and breast cancer progression. Since conclusive epidemiological studies require large sample sizes and sufficient duration of exposure before incident cancer occurrence, basic science studies represent the most promising approach to appropriately address the topic. Here we assessed the impact of intermittent hypoxia (IH), the major hallmark of OSA, on the development of breast cancer and explored the specific involvement of the endothelin signaling pathway. Original in vitro and in vivo models were used where 3D-spheroids or cultures of murine 4T1 breast cancer cells were submitted to IH cycles, and nude NMRI mice, orthotopically implanted with 4T1 cells, were submitted to chronic IH exposure before and after implantation. The role of the endothelin-1 in promoting cancer cell development was investigated using the dual endothelin receptor antagonist, macitentan. In vitro exposure to IH significantly increased 4T1 cell proliferation and migration. Meta-analysis of 4 independent in vivo experiments showed that chronic IH exposure promoted tumor growth, assessed by caliper measurement (overall standardized mean difference: 1.00 [0.45-1.55], p < 0.001), bioluminescence imaging (1.65 [0.59-2.71]; p < 0.01) and tumor weight (0.86 [0.31-1.41], p < 0.01), and enhanced metastatic pulmonary expansion (0.77 [0.12-1.42]; p = 0.01). Both in vitro and in vivo tumor-promoting effects of IH were reversed by macitentan. Overall, these findings demonstrate that chronic intermittent hypoxia exposure promotes breast cancer growth and malignancy and that dual endothelin receptor blockade prevents intermittent hypoxia-induced tumor development.


Subject(s)
Neoplasms , Sleep Apnea, Obstructive , Animals , Endothelin-1/metabolism , Hypoxia/metabolism , Mice , Receptor, Endothelin A
5.
Front Med (Lausanne) ; 9: 829979, 2022.
Article in English | MEDLINE | ID: mdl-35252260

ABSTRACT

Sleep Apnea Syndrome (SAS) is one of the most common chronic diseases, affecting nearly one billion people worldwide. The repetitive occurrence of abnormal respiratory events generates cyclical desaturation-reoxygenation sequences known as intermittent hypoxia (IH). Among SAS metabolic sequelae, it has been established by experimental and clinical studies that SAS is an independent risk factor for the development and progression of non-alcoholic fatty liver disease (NAFLD). The principal goal of this study was to decrypt the molecular mechanisms at the onset of IH-mediated liver injury. To address this question, we used a unique mouse model of SAS exposed to IH, employed unbiased high-throughput transcriptomics and computed network analysis. This led us to examine hepatic mitochondrial ultrastructure and function using electron microscopy, high-resolution respirometry and flux analysis in isolated mitochondria. Transcriptomics and network analysis revealed that IH reprograms Nuclear Respiratory Factor- (NRF-) dependent gene expression and showed that mitochondria play a central role. We thus demonstrated that IH boosts the oxidative capacity from fatty acids of liver mitochondria. Lastly, the unbalance between oxidative stress and antioxidant defense is tied to an increase in hepatic ROS production and DNA damage during IH. We provide a comprehensive analysis of liver metabolism during IH and reveal the key role of the mitochondria at the origin of development of liver disease. These findings contribute to the understanding of the mechanisms underlying NAFLD development and progression during SAS and provide a rationale for novel therapeutic targets and biomarker discovery.

6.
J Am Heart Assoc ; 9(16): e016369, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32805159

ABSTRACT

BACKGROUND Sleep-disordered breathing is associated with a poor prognosis (mortality) in patients with ischemic cardiomyopathy. The understanding of mechanisms linking intermittent hypoxia (IH), the key feature of sleep-disordered breathing, to ischemic cardiomyopathy progression is crucial for identifying specific actionable therapeutic targets. The aims of the present study were (1) to evaluate the impact of IH on the time course evolution of cardiac remodeling and contractile dysfunction in a rat model of ischemic cardiomyopathy; and (2) to determine the impact of IH on sympathetic activity, hypoxia inducible factor-1 activation, and endoplasmic reticulum stress in the time course of ischemic cardiomyopathy progression. METHODS AND RESULTS Ischemic cardiomyopathy was induced by a permanent ligature of the left coronary artery in male Wistar rats (rats with myocardial infarction). Rats with myocardial infarction were then exposed to either IH or normoxia for up to 12 weeks. Cardiac remodeling and function were analyzed by Sirius red and wheat germ agglutinin staining, ultrasonography, and cardiac catheterization. Sympathetic activity was evaluated by spectral analysis of blood pressure variability. Hypoxia-inducible factor-1α activation and burden of endoplasmic reticulum stress were characterized by Western blots. Long-term IH exposure precipitated cardiac remodeling (hypertrophy and interstitial fibrosis) and contractile dysfunction during the time course evolution of ischemic cardiomyopathy in rodents. Among associated mechanisms, we identified the early occurrence and persistence of sympathetic activation, associated with sustained hypoxia-inducible factor-1α expression and a delayed pro-apoptotic endoplasmic reticulum stress. CONCLUSIONS Our data provide the demonstration of the deleterious impact of IH on post-myocardial infarction remodeling and contractile dysfunction. Further studies are needed to evaluate whether targeting sympathetic nervous system or HIF-1 overactivities could limit these effects and improve management of coexisting ischemic cardiomyopathy and sleep-disordered breathing.


Subject(s)
Cardiomyopathies/physiopathology , Hypoxia/physiopathology , Myocardial Contraction/physiology , Sleep Apnea Syndromes/physiopathology , Ventricular Remodeling/physiology , Animals , Blood Pressure/physiology , Cardiomyopathies/complications , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum Stress/physiology , Heart , Hypoxia/complications , Hypoxia-Inducible Factor 1/metabolism , Lung/pathology , Male , Myocardial Infarction/pathology , Myocardial Ischemia/etiology , Myocardium/pathology , Rats , Rats, Wistar , Sleep Apnea Syndromes/complications , Time Factors
7.
Ther Adv Chronic Dis ; 11: 2040622320922104, 2020.
Article in English | MEDLINE | ID: mdl-32637058

ABSTRACT

BACKGROUND: Chronic intermittent hypoxia (IH), the hallmark feature of obstructive sleep apnoea syndrome, contributes to infarct size enhancement after myocardial ischemia-reperfusion (I/R). Curcumin (Curc), the natural pigment of Curcuma longa, has been demonstrated to be beneficial in the context of myocardial injury. In this study, we assessed the effects of Curc on the maladaptive cardiac response to IH, and particularly on IH-induced hypoxia inducible factor-1 (HIF-1) expression, oxidative stress, inflammation, endoplasmic reticulum (ER) stress and apoptosis. METHODS: Swiss/SV129 mice were exposed to normoxia or IH (21-5% FiO2, 60 s cycles, 8 h per day, for 21 days) and treated orally with Curc (100 mg kg-1 day-1, oral gavage) or its vehicle. Mice were then either euthanised for heart sampling in order to perform biochemical and histological analysis, or subjected to an in vivo ischemia-reperfusion protocol in order to measure infarct size. RESULTS: IH increased nuclear HIF-1α expression and superoxide anion (O2 .-) production as well as nuclear factor kappa B (NF-kB) p65, glucose-regulated protein (Grp78) and C/EBP homologous protein (CHOP) expression. IH also induced apoptosis and increased infarct size after I/R . The IH-induced HIF-1 activation, oxidative stress, inflammation, ER stress and apoptosis were abolished by chronic Curc treatment. Curc also significantly decreased infarct size only in mice exposed to IH. CONCLUSION: Curc prevents IH-induced myocardial cell death signalling. Curc might be used as a combined therapy with continuous positive airway pressure in sleep apnoea patients with high cardiovascular risk.

8.
Can J Cardiol ; 36(6): 936-940, 2020 06.
Article in English | MEDLINE | ID: mdl-32387037

ABSTRACT

Chronic intermittent hypoxia (CIH) occurring during sleep apnea amplifies infarct size owing to ischemia-reperfusion. CIH activates hypoxia-inducible factor 1 (HIF-1) and activating transcription factor 4 (ATF4). However, whether HIF-1 and ATF4 interact to promote cardiomyocyte death remains unexplored. For the first time, we observed that in myocardium from apneic patients, CCAAT enhancer-binding protein homologous protein (CHOP) expression is increased and HIF-1α expression is correlated with sleep apnea severity. In mice, single-allele deletion of HIF-1α prevents CIH increase in CHOP expression and infarct size. We uncovered a physical interaction between HIF-1α and ATF4 in CIH that may represent a novel cardiomyocyte death complex.


Subject(s)
Activating Transcription Factor 4/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia , Myocardial Infarction , Sleep Apnea Syndromes , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Death , Humans , Hypoxia/complications , Hypoxia/etiology , Hypoxia/metabolism , Mice , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/metabolism
9.
Mater Sci Eng C Mater Biol Appl ; 106: 110281, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31753335

ABSTRACT

Artemisinin and its derivatives are currently recommended by World Health Organization for the treatment of malaria. Severe malaria requires a parenteral administration of artemisinin-based formulations. However, the effective use of artemisinin is limited by the pharmacokinetic characteristics of the drug (low water solubility, poor bioavailability and short half-life). To overcome some of these drawbacks, artemisinin-loaded surface-decorated nanoparticles were prepared by co-nanoprecipitation of γ-cyclodextrin bioesterified with C10 alkyl chains and polyethylene glycol (PEG) derivatives (polysorbate 80 and DMPE-mPEG2000). Using a single dose (1.5 mg kg-1 or 2 mg kg-1) by intravenous administration, we investigated the in vivo pharmacokinetic properties in healthy rats of two types of artemisinin-loaded nanoparticle formulations, namely, nanosphere and nanoreservoir systems versus an ethanolic-aqueous solution of artemisinin as reference. Significantly enhanced pharmacokinetic parameters were obtained with artemisinin-loaded nanoparticles. In comparison to reference formulation, the geometric mean exposures in plasma (AUC0-t) exhibited 2.35 and 3.26-fold increases when artemisinin was loaded in nanoreservoir and nanosphere systems, respectively. Its plasma half-life increased 4.00 and 6.25-fold and its clearance decreased up to 2.5 and 4.72-fold. Artemisinin was successfully administered intravenously by means of surface-decorated amphiphilic γ-cyclodextrin nanostructures and showed a longer elimination half-life with respect to an artemisinin solution in ethanol. Therefore, these systems are likely to provide significant advantages for the intravenous treatment of severe malaria.


Subject(s)
Antimalarials/pharmacokinetics , Artemisinins/chemistry , Nanoparticles/chemistry , gamma-Cyclodextrins/chemistry , Administration, Intravenous , Animals , Antimalarials/blood , Antimalarials/chemistry , Artemisinins/blood , Artemisinins/pharmacokinetics , Drug Carriers/chemistry , Half-Life , Male , Particle Size , Polyethylene Glycols/chemistry , Rats , Rats, Wistar , Surface Properties
10.
Toxins (Basel) ; 11(9)2019 09 10.
Article in English | MEDLINE | ID: mdl-31510060

ABSTRACT

Myocardial infarction (MI) followed by left ventricular (LV) remodeling is the most frequent cause of heart failure. Lebetin 2 (L2), a snake venom-derived natriuretic peptide, exerts cardioprotection during acute myocardial ischemia-reperfusion (IR) ex vivo. However, its effects on delayed consequences of IR injury, including post-MI inflammation and fibrosis have not been defined. Here, we determined whether a single L2 injection exerts cardioprotection in IR murine models in vivo, and whether inflammatory response to ischemic injury plays a role in L2-induced effects. We quantified infarct size (IS), fibrosis, inflammation, and both endothelial cell and cardiomyocyte densities in injured myocardium and compared these values with those induced by B-type natriuretic peptide (BNP). Both L2 and BNP reduced IS, fibrosis, and inflammatory response after IR, as evidenced by decreased leukocyte and proinflammatory M1 macrophage infiltrations in the infarcted area compared to untreated animals. However, only L2 increased anti-inflammatory M2-like macrophages. L2 also induced a higher density of endothelial cells and cardiomyocytes. Our data show that L2 has strong, acute, prolonged cardioprotective effects in post-MI that are mediated, at least in part, by the modulation of the post-ischemic inflammatory response and especially, by the enhancement of M2-like macrophages, thus reducing IR-induced necrotic and fibrotic effects.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Natriuretic Peptide, Brain/therapeutic use , Viper Venoms/therapeutic use , Animals , Blood Pressure/drug effects , Fibrosis , Heart Rate/drug effects , Male , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Rats, Wistar
11.
Am J Physiol Cell Physiol ; 317(2): C270-C276, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31116583

ABSTRACT

Zinc is involved in the expression and function of various transcription factors, including the hypoxia-inducible factor-1 (HIF-1). HIF-1 and its target gene endothelin-1 (ET-1) are activated by intermittent hypoxia (IH), one of the main consequences of obstructive sleep apnea (OSA), and both play a key role in the cardiovascular consequences of IH. Because OSA and IH are associated with zinc deficiency, we investigated the effect of zinc deficiency caused by chelation on the HIF-1/ET-1 pathway and its functional consequences in endothelial cells. Primary human microvascular endothelial cells (HMVEC) were incubated with submicromolar doses of the zinc-specific membrane-permeable chelator N,N,N',N'-tetrakis(2-pyridylmethyl)-ethylene diamine (TPEN, 0.5 µM) or ET-1 (0.01 µM) with or without bosentan, a dual ET-1-receptor antagonist. HIF-1α expression was silenced by transfection with specific siRNA. Nuclear HIF-1 content was assessed by immunofluorescence microscopy and Western blot. Migratory capacity of HMVEC was evaluated with a wound-healing scratch assay. Zinc chelation by TPEN exposure induced the translocation of the cytosolic HIF-1α subunit of HIF-1 to the nucleus as well as an HIF-1-mediated ET-1 secretion by HMVEC. Incubation with either TPEN or ET-1 increased endothelial wound-healing capacity. Both HIF-1α silencing or bosentan abolished this effect. Altogether, these results suggest that zinc deficiency upregulates ET-1 signaling through HIF-1 activation and stimulates endothelial cell migration, suggesting an important role of zinc in the vascular consequences of IH and OSA mediated by HIF-1-ET- signaling.


Subject(s)
Cell Movement/drug effects , Chelating Agents/pharmacology , Endothelial Cells/drug effects , Endothelin-1/metabolism , Ethylenediamines/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Zinc/deficiency , Active Transport, Cell Nucleus , Bosentan/pharmacology , Cells, Cultured , Endothelial Cells/metabolism , Endothelin Receptor Antagonists/pharmacology , Endothelin-1/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Secretory Pathway , Signal Transduction
12.
Sci Rep ; 9(1): 7252, 2019 05 10.
Article in English | MEDLINE | ID: mdl-31076597

ABSTRACT

Muscle wasting reduces functional capacity and increases cardiometabolic risk in chronic disease. Neuromuscular electrical stimulation (NMES) of the lower limb has been shown to reverse muscle wasting in these patients but its effect on cardiometabolic health is unclear. We investigated a mouse model of in-vivo non-invasive chronic NMES on muscle mass, insulin sensitivity and arterial blood pressure (BP). Twenty-three C57BL6 mice underwent unilateral NMES or sham training over 2.5 weeks while anesthetized by isoflurane. Lower limb muscle mass and the stimulated limb to non-stimulated limb muscle mass ratio were compared between groups (NMES vs. sham). Insulin sensitivity was assessed 48 h after training using an intraperitoneal insulin tolerance test (ITT) and BP was assessed before and after training using the tail-cuff technique. After training, muscle mass increased in NMES vs. sham (416 ± 6 vs. 397 ± 6 mg, p = 0.04) along with the ratio of muscle mass (+3 ± 1% vs. -1 ± 1% p = 0.04). Moreover, insulin sensitivity improved in NMES vs. sham (average blood glucose during ITT: 139.6 ± 8.5 vs. 161.9 ± 9.0 mg/dl blood, p = 0.01). BP was decreased in both groups, although it is likely that the effect of NMES on BP was dampened by repetitive anesthesia. The metabolic benefit of NMES training could be of great utility in patients with chronic disease. Moreover, the clinical-like mouse model of NMES is an effective tool to investigate the systemic effects of local muscle strengthening.


Subject(s)
Insulin Resistance/physiology , Muscle, Skeletal/physiopathology , Animals , Electric Stimulation/methods , Electric Stimulation Therapy/methods , Lower Extremity/physiology , Mice , Mice, Inbred C57BL , Muscle Strength/physiology , Muscular Atrophy/physiopathology
14.
Invest Ophthalmol Vis Sci ; 59(12): 5256-5265, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30383197

ABSTRACT

Purpose: Obstructive sleep apnea recently has been associated with a higher frequency of ischemic optic neuropathies. Intermittent hypoxia (IH) has been proposed as a major component of obstructive sleep apnea cardiovascular consequences. However, there currently are no pathophysiologic data regarding the effect of IH on the ocular vascular system. Thus, we assessed the impact of chronic IH exposure on the morphology and vascular reactivity of the rat ophthalmic artery (OA). Methods: Rats were exposed to 14 days of IH or normoxia (NX). Ophthalmic artery reactivity was studied using wire myography in rats treated or not with tempol (1 mM/day). Expression of endothelin-1 (ET-1) and its receptors, and of the three nitric oxide synthase (NOS) isoform genes was quantified using quantitative polymerase chain reaction (qPCR) in the retina and optic nerve. Structural alterations (optical and electron microscopy) and superoxide anion production were studied in OA sections. Results: Superoxide ion expression in the OA wall was increased by 23% after IH exposure. Ophthalmic artery contractile response to 3.10-8 M ET-1 was increased by 18.6% and nitric oxide-mediated relaxation was significantly delayed in IH compared to NX rats. In the absence of nitric oxide, cytochrome P450 blockade increased relaxation to acetylcholine in IH rats and delayed it in NX rats. Tempol treatment abolished the IH-induced changes in OA reactivity. Conclusions: These results strongly suggest that chronic IH induces oxidative stress in the rat OA, associated with endothelial dysfunction through alterations of nitric oxide and endothelium-derived hyperpolarising factors (EDHF) pathways.


Subject(s)
Endothelin-1/metabolism , Hypoxia/physiopathology , Ophthalmic Artery/physiopathology , Oxidative Stress , Receptor, Endothelin A/metabolism , Animals , Chronic Disease , Cyclic N-Oxides/pharmacology , Hypoxia/metabolism , Male , Muscle, Smooth, Vascular/physiology , Myography , Nitric Oxide Synthase/genetics , Ophthalmic Artery/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Spin Labels , Superoxides/metabolism
17.
Sci Rep ; 8(1): 2997, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445096

ABSTRACT

We investigated the effects of intermittent hypoxia (IH), such as that encountered in severe obstructive sleep apnea (OSA) patients, on the development and severity of myocardial ischemia-related ventricular arrhythmias. Rats were exposed to 14 days of IH (30 s at 5%O2 and 30 s at 21%O2, 8 h·day-1) or normoxia (N, similar air-air cycles) and submitted to a 30-min coronary ligature. Arterial blood pressure (BP) and ECG were recorded for power spectral analysis, ECG interval measurement and arrhythmia quantification. Left ventricular monophasic action potential duration (APD) and expression of L-type calcium (LTCC) and transient receptor potential (TRPC) channels were assessed in adjacent epicardial and endocardial sites. Chronic IH enhanced the incidence of ischemic arrhythmias, in particular ventricular fibrillation (66.7% vs. 33.3% in N rats, p < 0.05). IH also increased BP and plasma norepinephine levels along with increased low-frequency (LF), decreased high-frequency (HF) and increased LF/HF ratio of heart rate and BP variability. IH prolonged QTc and Tpeak-to-Tend intervals, increased the ventricular APD gradient and upregulated endocardial but not epicardial LTCC, TRPC1 and TRPC6 (p < 0.05). Chronic IH, is a major risk factor for sudden cardiac death upon myocardial ischemia through sympathoactivation and alterations in ventricular repolarization, transmural APD gradient and endocardial calcium channel expression.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Hypoxia/physiopathology , Myocardial Ischemia/physiopathology , Sleep Apnea, Obstructive/physiopathology , Ventricular Function/physiology , Animals , Cells, Cultured , Chronic Disease , Coronary Vessels/surgery , Death, Sudden, Cardiac , Disease Models, Animal , Gene Expression Regulation , Humans , Male , Rats , Rats, Wistar , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism
18.
Sleep Med Rev ; 38: 113-130, 2018 04.
Article in English | MEDLINE | ID: mdl-29107469

ABSTRACT

Many associations between ocular disorders and obstructive sleep apnea (OSA) have been studied, such as nonarteritic anterior ischemic optic neuropathy, glaucoma, papilledema, retinal vein occlusion, eyelid hyperlaxity, lower-eyelid ectropion and recurrent corneal erosions. The objective of this review is to synthetize the possible vascular disorders of the retina and the optic nerve associated with sleep apnea patients and to discuss the underlying pathophysiological hypotheses. Main mechanisms involved in the ocular complications of OSA are related to intermittent hypoxia, sympathetic system activation, oxidant stress, and deleterious effects of endothelin 1. The main evidence-based medicine data suggest that OSA should be screened in patients with ischemic optic neuropathy and diabetic retinopathy. The effect of OSA treatment and emerging therapies are discussed.


Subject(s)
Diabetic Retinopathy/physiopathology , Glaucoma/physiopathology , Optic Neuropathy, Ischemic/physiopathology , Sleep Apnea, Obstructive/physiopathology , Humans , Intraocular Pressure/physiology , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/therapy
19.
Diabetes ; 66(12): 2942-2951, 2017 12.
Article in English | MEDLINE | ID: mdl-28882901

ABSTRACT

Obstructive sleep apnea syndrome is a highly prevalent disease resulting in transient respiratory arrest and chronic intermittent hypoxia (cIH). cIH is associated with insulin resistance and impaired metabolic homeostasis in rodents and humans, but the exact underlying mechanisms remain unclear. In the current study, we investigated the effects of 2 weeks of cIH (1-min cycle, fraction of inspired oxygen 21-5%, 8 h/day) on whole-body insulin sensitivity and glucose tolerance in lean mice. Although food intake and body weight were reduced compared with normoxia, cIH induced systemic insulin resistance in a hypoxia-inducible factor 1-independent manner and impaired insulin signaling in liver, white adipose tissue, and skeletal muscle. Unexpectedly, cIH improved whole-body glucose tolerance independently of changes in body weight and glucose-induced insulin response. This effect was associated with elevated phosphorylation of Thr172-AMPK and Ser237-TBC1 domain family member 1 (TBC1D1) in skeletal muscle, suggesting a tissue-specific AMPK-dependent increase in TBC1D1-driven glucose uptake. Remarkably, although food intake, body weight, and systemic insulin sensitivity were still affected, the improvement in glucose tolerance by cIH was abolished in muscle-specific AMPKα1α2-deficient mice. We conclude that cIH impairs insulin sensitivity while improving whole-body glucose tolerance by promoting specific activation of the skeletal muscle AMPK pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hypoxia/metabolism , Insulin Resistance , Muscle, Skeletal/enzymology , Animals , Chronic Disease , Enzyme Activation , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Mice , Mice, Inbred C57BL
20.
Am J Physiol Cell Physiol ; 313(4): C460-C468, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28747336

ABSTRACT

Performing hypoxia-reoxygenation cycles in cell culture with a cycle duration accurately reflecting what occurs in obstructive sleep apnea (OSA) patients is a difficult but crucial technical challenge. Our goal was to develop a novel device to expose multiple cell culture dishes to intermittent hypoxia (IH) cycles relevant to OSA with limited gas consumption. With gas flows as low as 200 ml/min, our combination of plate holders with gas-permeable cultureware generates rapid normoxia-hypoxia cycles. Cycles alternating 1 min at 20% O2 followed by 1 min at 2% O2 resulted in Po2 values ranging from 124 to 44 mmHg. Extending hypoxic and normoxic phases to 10 min allowed Po2 variations from 120 to 25 mmHg. The volume of culture medium or the presence of cells only modestly affected the Po2 variations. In contrast, the nadir of the hypoxia phase increased when measured at different heights above the membrane. We validated the physiological relevance of this model by showing that hypoxia inducible factor-1α expression was significantly increased by IH exposure in human aortic endothelial cells, murine breast carcinoma (4T1) cells as well as in a blood-brain barrier model (2.5-, 1.5-, and 6-fold increases, respectively). In conclusion, we have established a new device to perform rapid intermittent hypoxia cycles in cell cultures, with minimal gas consumption and the possibility to expose several culture dishes simultaneously. This device will allow functional studies of the consequences of IH and deciphering of the molecular biology of IH at the cellular level using oxygen cycles that are clinically relevant to OSA.


Subject(s)
Blood-Brain Barrier/metabolism , Breast Neoplasms/metabolism , Cell Culture Techniques , Endothelial Cells/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Sleep Apnea, Obstructive/metabolism , Animals , Cell Culture Techniques/instrumentation , Cell Hypoxia , Cell Line, Tumor , Culture Media/metabolism , Equipment Design , Female , Gases , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Time Factors , Tumor Hypoxia
SELECTION OF CITATIONS
SEARCH DETAIL
...