Exploring the pharmacological mechanisms for alleviating OSA: Adenosine A2A receptor downregulation of the PI3K/Akt/HIF‑1 pathway (Review)
- Authors:
- Published online on: November 28, 2024 https://doi.org/10.3892/br.2024.1899
- Article Number: 21
-
Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
Over the past two decades, the rapid aging of the population has led to a 30% increase in the prevalence of OSA (1). Epidemiological research estimates that ~936 million adults aged 30-69 globally are affected by OSA, with a prevalence of ~22% in males and 17% in females (2). The COVID-19 pandemic has further exacerbated the incidence of OSA (3), solidifying its status as a common sleep-related breathing disorder in clinical settings (4). OSA is characterized by recurrent obstruction of the upper airway during sleep, leading to periodic hypoxemia, disturbances in sleep continuity and increased respiratory effort. OSA is characterized by intermittent hypoxia during sleep, which activates HIF-1. This activation leads to oxidative stress, damaging cellular components such as DNA, proteins and lipids. The repeated cycles of hypoxia and subsequent oxidative stress create an environment conducive to genetic alterations that may drive the malignant transformation of lung cells (5). OSA triggers a systemic inflammatory response. The intermittent hypoxia and sleep fragmentation associated with OSA activate the sympathetic nervous system, releasing pro-inflammatory cytokines such as IL-6, TNF-α and high-sensitivity C-reactive protein (hsCRP). Chronic inflammation disrupts normal cellular functions and tissue homeostasis, fostering an immunosuppressive microenvironment in the lungs that allows cancer cells to evade immune surveillance and elimination. In addition, inflammatory mediators can directly stimulate cell proliferation and angiogenesis, both crucial for tumor growth and metastasis (6). OSA is linked to significant metabolic changes, disrupting normal lipid metabolism and leading to abnormal lipid profiles, while also affecting glucose metabolism, resulting in insulin resistance and elevated blood glucose levels. These metabolic alterations create a favorable environment for cancer cells to thrive, as hyperglycemia provides an abundant energy source and can stimulate growth factor production (7,8). Notably, OSA has been emerged as a new risk factor for lung cancer (9) and is known to disrupt normal lipid metabolism (10,11).
A hallmark molecular feature of hypoxia in OSA is the upregulation and stabilization of HIF-1 (12-14), a critical genomic mediator of cellular adaptation to low oxygen levels. HIF-1 plays an essential role in regulating inflammatory pathways by modulating inflammatory responses, particularly preventing excessive reactions. In addition, it is associated with the regulation of genes such as MCAD and LCAD, both of which are involved in lipid metabolism. Recent research has highlighted the potential of A2A receptor (A2AR) blockade to disrupt the hypoxia/HIF-1-adenosine immunosuppressive axis, enhancing the effectiveness of cancer immunotherapy (15). Additionally, A2AR is indispensable for tissue protection under hypoxic conditions and for the regulation of lipid metabolism (16).
OSA is mainly mediated by intermittent hypoxia, which leads to low-grade inflammation (17). Adenosine receptors (ARs) have been shown to mitigate hypoxia-induced inflammation, particularly in the context of acute lung injury. During ischemic and hypoxic events, extracellular concentrations of adenosine, a crucial neurotransmitter involved in the immune response, can increase markedly, reaching levels ≤100 times higher than normal. The upregulation of adenosine and the induction of HIF-1 in response to hypoxia are directly linked, suggesting that HIF-1 activation through A2AR may contribute to anti-inflammatory responses and tissue protection. Although persistently elevated adenosine levels appear to trigger the release of inflammatory cytokines, A2AR is predominantly expressed in mature dendritic cells, leading to a reduction in pro-inflammatory cytokines (18). Previous studies have demonstrated that hypoxia activates adenosine signaling via A2AR (19-21). Additionally, the HIF-1/adenosine axis provides protective effects for the lungs during conditions such as acute respiratory distress syndrome, although it may contribute to inflammation and injury in chronic lung diseases (22).
Based on these findings, along with research showing that targeting the hypoxic-A2AR pathway can release anti-tumor T cells with immunosuppressive properties (23), A2AR has emerged as a critical receptor for mediating hypoxia-related tissue protection and regulating HIF-1 due to its anti-inflammatory effects (16). The molecular process by which A2AR regulates HIF-1 involves cytokine interactions and the activation of intracellular pathways such as Protein Kinase C (PKC), ATP-sensitive Potassium Channel (KATP), p38 MAPK and PI3K/Akt (24,25). These intricate networks present a promising therapeutic target for A2AR, particularly through the PI3K/Akt/HIF-1 pathway in the treatment of OSA (Fig. 1). The present study aimed to clarify the current prevalence of OSA and its associated hazards, including intermittent hypoxia, sleep disorders and its role as a risk factor for diseases such as lung cancer. It provided an in-depth exploration of the pharmacological mechanisms of A2AR in OSA, focusing on its effects on inflammation and lipid metabolism related to OSA through the PI3K/Akt/HIF-1 pathway.
2. Literature review
Interplay between OSA, inflammation and lipid metabolism
The oxidative stress caused by OSA has complex and multifaceted effects on lipid metabolism and cardiovascular health (26). In terms of lipid metabolism, the reactive oxygen species (ROS) produced by oxidative stress will attack the polyunsaturated fatty acids in the lipids, causing lipid peroxidation, such as the oxidation of low-density lipoprotein (LDL), which is more likely to be absorbed by the macrophages in the artery wall to form foam cells and start the formation of atherosclerotic plaque (27-29). Meanwhile, oxidative stress can alter the activity of enzymes involved in lipid metabolism, such as lipoprotein lipase (LpL), disrupting the balance between lipid synthesis and breakdown (30). It can also interfere with cholesterol transport and metabolism, affecting LDL receptors and related functions such as hydroxymethylglutaryl-CoA reductase, leading to lipid metabolism disorders (31). In terms of cardiovascular health, due to oxidative stress generated by OSA, LDL oxidation is the key to the development of atherosclerosis. Its accumulation in the arterial wall triggers inflammatory reaction, forms fat stripes and develops into plaques; and plaque rupture can cause thrombosis and cardiovascular events (32,33). In addition, oxidative stress damages endothelial cells, reducing their release of NO, leading to vasoconstriction, increased platelet aggregation and causing problems such as hypertension (34,35). In addition, oxidative stress activates inflammatory pathways and immune system, promotes the release of pro-inflammatory cytokines, intensifies the process of atherosclerosis and affects the function of immune cells (36-38). It also directly damages mitochondria in myocardial cells, affects energy production, leads to the decline of myocardial contractility and interferes with calcium ion regulation to damage myocardial function (39-41). It can be seen that oxidative stress caused by OSA has a significant effect on lipid metabolism and cardiovascular health.
The sleep disorder and intermittent hypoxia characteristic of OSA can trigger sympathetic excitation and inflammation, leading to vascular endothelial damage, altered coagulation function, abnormal lipid metabolism and disruptions in glucose homeostasis (42,43). The physiological consequences of OSA activate the sympathetic nervous system, induce oxidative stress and trigger systemic inflammatory responses (44). The systemic inflammatory cascade is thought to play a crucial role in both the onset and progression of OSA (45). Studies have shown that systemic inflammation associated with OSA is caused by the overflow of inflammatory cytokines from the upper airway mucosa to the bloodstream (46). Clinical studies have measured plasma concentrations of inflammatory markers in patients with OSA compared with control groups, revealing elevated levels of hsCRP, IL-6, TNF-α and pentraxin-3 among individuals with OSA (47). In addition, in a mouse experiment, the changes of atherosclerosis induced by intermittent hypoxia occurs with the increased expression of proinflammatory cytokines, chemokines and adhesion molecules, the increased migration of inflammatory cells and the expansion of the population of macrophages in the arterial wall (48,49). Other studies have also confirmed that the hypoxic state of OSA preferentially activated the pro-inflammatory factor NF-κB-mediated pathway, possibly caused by an inflammatory response to hypoxic exposure via adipocytes (50-52). This inflammatory shift is further associated with increased polarization of pro-inflammatory M1 macrophages, elevated expression of inducible nitric oxide synthase and worsening insulin resistance (53). This inflammatory response mechanism plays a crucial role in the pathogenesis of OSA-related cardiometabolic processes, ultimately leading to the development of cardiovascular disease and establishing a vicious cycle (54).
Lipid metabolism plays a pivotal role in the development of OSA (55,56). In patients with OSA, the concentrations of triglycerides, total cholesterol and low-density lipoprotein cholesterol increase, while the levels of high-density lipoproteins cholesterol decrease accordingly (57). A genome-wide association study confirms the genetic link between OSA and triglycerides levels (58). Notably, dyslipidemia and OSA share common genetic loci, such as peroxisome proliferator-activated receptor (PPAR)-γ (59) or apolipoprotein E (APOE) polymorphism (60). PPAR-γ is a major regulator of several genes related to lipid metabolism, including LpL, which is downregulated by hypoxia in a HIF-1-dependent manner (61). The e4 variant of APOE (APOE-e4) is independently associated with abnormal lipid metabolism and can exacerbate OSA progression and cognitive dysfunction through age-related lipid bioenergetics dysfunction (62-64). Additionally, abnormal blood lipids can lead to insulin resistance. The increase in free fatty acid (FFA) levels caused by enhanced fat breakdown reduces insulin-mediated glucose uptake in skeletal muscle by disrupting insulin signaling (65). Furthermore, FFAs activate the NF-κB pathway, leading to the production of pro-inflammatory cytokines such as TNF-α, IL1β and IL-6 in peripheral tissues. Animal studies have indicated that intermittent hypoxia leads to pancreatic and insulin resistance in insulin-sensitive organs and adipose tissue (66,67). In addition, the hypoxic stress associated with OSA activates the hypothalamic-pituitary-adrenal axis, resulting in elevated cortisol levels, which ultimately contribute to insulin resistance (68). This hypoxic stress mechanism also contributes to the development of other diseases that induce insulin resistance through oxidative stress and pancreatic islet cell apoptosis via the TRB3 and phosphorylated JNK pathways, thereby contributing to the onset of type 2 diabetes and hyperlipidemia in OSA populations (69).
The role of Adenosine A2A receptor (A2AR) in inflammation and lipid metabolism
A growing body of evidence indicating that A2AR mediates potent anti-inflammatory responses in various cell types across multiple inflammation models (70,71) has spurred the development of A2AR agonists aimed at attenuating inflammation in disorders such as chronic obstructive pulmonary disease (COPD) and asthma (72). Research has shown that A2AR agonists can modulate immune responses by reducing the infiltration of pro-inflammatory T cells into the central nervous system, an essential mechanism for controlling flare-ups in multiple sclerosis (73). In a mouse model of carrageenan-induced pleurisy, the administration of the A2AR agonist CGS 21680 markedly reduced neutrophil infiltration, nitric oxide levels, cytokine production, NF-κB expression and PARP activation (74). By contrast, A2AR knockout mice exhibited increased inflammation characterized primarily by enhanced activity of macrophages and neutrophils, along with elevated mucin production in the bronchial airways and increased levels of the chemoattractant proteins chemokine (C-X-C motif) ligand and MCP-1 (75,76). These findings suggest that A2AR plays a protective role in pulmonary inflammation. Furthermore, the positive allosteric modulator AEA061 enhances inosine-mediated A2AR activation, leading to inhibition of pro-inflammatory cytokines and chemokine production by splenic monocytes (77). Studies have also demonstrated that activating A2AR can restore cAMP levels in myocardial tissue while inhibiting the NF-κB signaling pathway, markedly improving cardiac dysfunction associated with cirrhosis and exerting both anti-inflammatory and anti-apoptotic effects (78). In the context of psoriasis, A2AR activation inhibits M1 macrophage activation through the NF-κB-KRT16 pathway, which is crucial for initiating both innate and adaptive immunity (75). Additionally, inhalation of A2AR agonists has shown therapeutic potential for patients suffering from COVID-19-related inflammatory lung disease (79). These findings collectively highlight the therapeutic promise of A2AR modulation in various inflammatory conditions.
A2A receptors play a crucial role in regulating various physiological and pathological processes in adipocytes (80). Activation of A2AR can induce anti-inflammatory effects that are essential for the survival of beta cells, enhance insulin secretion, reduce food intake and promote thermogenesis and fat breakdown (81). For instance, studies have demonstrated that A2AR agonists, such as CGS21680 and PSB-0777, activate lipolysis in both humans and mice (82-84), improve glucose tolerance and protect C57Bl/6 mice from diet-induced obesity, highlighting the promising thermogenic effects of adenosine. Further investigations showed that the A2AR agonist CGS21680, when injected into Swiss strain mice fed a high-fat diet, produced similar effects on glucose homeostasis without significant changes in weight or obesity rates, while also decreasing certain inflammatory markers (85). Additionally, A2AR signaling has been linked to the regulation of CD8+ T cell responses through the coordination of glutathione metabolism (86). Notably, stimulation of A2AR expression on macrophages has been shown to release cholesterol, which inhibits the formation of foam cells (84). The structural dynamics of A2AR are influenced by its phospholipid environment and cholesterol, resulting in a propensity to bind lipid isoform modulators (87). In therapeutic applications, liposome treatments combined with adenosine or specific A2AR agonists have markedly improved joint scores in post-traumatic osteoarthritis rats and mice with high-fat diet-induced osteoarthritis (88). In addition, research indicates that the loss of A2AR in macrophages and liver cells leads to increased inflammation, elevated expression and transcriptional activity of SREBP1c and enhanced adipogenic events, exacerbating the severity of non-alcoholic fatty liver disease (89). Studies have also shown that deficiencies in C3a and C5a receptors can promote adipocyte browning and reduce diet-induced obesity by activating the inosine/A2AR pathway (90,91). Lastly, ongoing research and development efforts are focusing on dual-active adenosine A2A/A3 receptor ligands, such as LJ-4378, which have demonstrated anti-obesity effects and offer new treatment strategies for obesity and related metabolic diseases (92).
3. Mechanistic pathways
A2AR's Anti-inflammatory mechanism via PI3K/Akt/HIF-1 pathway
A2AR plays a significant role in exerting anti-inflammatory and tissue-protective effects, particularly in hypoxic environments, by modulating the PI3K/Akt/HIF-1 signaling pathway. OSA is associated with chronic inflammation and tissue remodeling, leading to progressive declines in pulmonary function (26). In OSA mouse models, a notable accumulation of proinflammatory M1-like macrophages, characterized by heightened CD36 expression, has been observed in the aorta, alongside elevated levels of inflammation-related transcription factors (93). During intermittent hypoxia, OSA triggers the release of proinflammatory factors, including TNF, CRP, IL-6 and IL-8, while increased NF-κB and TNF-α levels have been linked to OSA-related daytime sleepiness (94-96).
In this context, A2AR mitigates inflammation through the PI3K/Akt pathway. Notably, while serotonin receptor inhibition can limit the efficacy of intermittent hypercapnic-hypoxia therapies, blocking the A2AR pathway has demonstrated enhanced respiratory recovery (97). Additionally, A2AR influences the regulation of apnea, particularly in conditions such as apnea of prematurity, where its effects are mediated by caffeine inhibitors (98). The flavonoid compound Baicalin has also been shown to alleviate chronic hypoxia-induced pulmonary hypertension by activating the A2AR-induced Stromal Cell-Derived Factor 1(SDF-1)/C-X-C Chemokine Receptor Type 4 (CXCR4)/PI3K/Akt signaling pathway (95). In cases of liver ischemia/reperfusion injury, inhalation of high hydrogen concentrations has demonstrated ameliorative effects via the A2AR-mediated PI3K/Akt pathway (94).
Research has confirmed that A2AR inhibits the PI3K/Akt signaling pathway, which is essential for limiting inflammation and promoting anti-inflammatory responses in TLR-induced macrophages (99,100). This pathway negatively regulates TLR and NF-κB signaling in macrophages (101,102). The Bu-Shen-Fang-Chuan formula, commonly prescribed for COPD in China, has been shown to reduce TNF-α and IL-6 levels in bronchoalveolar fluid and serum, curbing cigarette smoke-induced inflammation, partially mediated through the PI3K/Akt pathway (103). A2AR also modulates HIF-1 expression; studies indicate that both Akt and HIF-1 protein levels increase in human mesenchymal stem cells in response to hypoxia, with Akt expression peaking earlier than HIF-1 (104,105). The use of PI3K inhibitors such as LY294002 and dual PI3K/mTOR inhibitors such asNVP-BEZ235 suppresses Akt activation, as well as HIF-1 and VEGF expression induced by hypoxia. Wortmannin, an Akt inhibitor, also inhibits HIF-1 expression at the protein level without affecting mRNA levels (104).
Activation of specific HIF-1 target genes can induce proinflammatory genes, notably IL-1β (106-108). However, in hypoxic macrophages, the activation of the PI3K/Akt pathway upregulates TLR4 expression through HIF-1 activation (109). The HIF-1 dimer then binds to the hypoxia response element in the promoter region, initiating the expression of over 100 genes involved in hypoxic adaptation, including those that promote VEGF and EPO, which promote angiogenesis and erythropoiesis (110,111). HIF-1 also regulates NF-κB, leading to increased levels of cytokines such as IL-8 (112,113), while IL-17 levels correlate with OSA severity (114). Therefore, A2AR inhibits the phosphorylation of PI3K and Akt, indirectly downregulating HIF-1(115). This mechanism drives the reduction of immune and inflammatory responses, thereby protecting tissues in hypoxic conditions (116).
A2AR's lipid metabolism mechanism via PI3K/Akt/HIF-1 pathway
A2AR plays a significant role in regulating lipid metabolism through the PI3K/Akt/HIF-1 pathway (115). Activation of A2AR inhibits the phosphorylation of PI3K and Akt, crucial steps in lipid metabolism regulation. Notably, HIF-1, activated downstream of this pathway, promotes lipid droplet accumulation and fatty acid reprogramming under hypoxic conditions.
Endogenous cannabinoids are naturally occurring lipids that bind to cannabinoid receptors and play a crucial role in regulating metabolism, particularly in energy balance, fat storage and glucose homeostasis (117). In the context of OSA, these cannabinoids can contribute to metabolic disorders and exacerbate conditions such as obesity, insulin resistance and type 2 diabetes. Elevated levels of endogenous cannabinoids, particularly anandamide and ethanolamine, have been observed in patients with OSA, along with increased levels of saturated fatty acids and n-3 fatty acids, which can enhance appetite and promote fat accumulation, both of which are linked to sleep quality (118,119). Additionally, patients with OSA often exhibit elevated levels of adenosine, adrenaline, norepinephrine and aldosterone, further complicating their metabolic profiles (120). In another animal study, intermittent hypoxia was demonstrated to mediate the expression of hypoxia-inducible factor 1αin pancreatic β-cells. This leads to increased reactive oxygen species and ultimately resulted in insulin resistance (12).
A2AR activation not only stimulates lipolysis and thermogenesis but also enhances the browning of adipose tissue. Specifically, A2AR activation enhances reverse cholesterol transport from peripheral tissues back to the liver, aided by macrophages, which helps prevent their transformation into foam cells (121). Furthermore, supplementation with adenosine has been shown to increase A2AR protein levels and enhance the expression of key lipolytic genes, such as ATGL and HSL, in adipose tissue, thereby providing a protective effect against diet-induced obesity. Additionally, elevated levels of inflammatory factors in the upper and lower airways of hypoxic mice, combined with the detrimental cycle between OSA and airway inflammation, contribute markedly to insulin resistance (122).
Metabolic changes in patients with OSA include increased lactic acid and specific fatty acids, such as arabinose and glyceraldehyde (123). Additionally, genes involved in cholesterol metabolism, such as malic enzyme and acetyl-CoA carboxylase, are impaired due to hypoxia associated with OSA (124,125). The role of HIF-1 in lipid uptake is crucial, as it induces the expression of fatty acid binding proteins (FABP3 and FABP7) and adipocyte differentiation-related protein (ADRP), necessary for lipid droplet formation. HIF-1 also promotes fatty acid synthase expression, enhancing fatty acid synthesis while inhibiting the oxygen-dependent stearoyl-CoA desaturase enzyme, which can affect cellular membrane integrity (126).
Furthermore, HIF-1 targets ATP citrate lyase (ACLY), which is upregulated in hypoxic tumor cells, influencing fatty acid biosynthesis and acetyl-CoA production. For example, in goose liver cells, insulin regulates lipid deposition through the PI3K/Akt/mTOR pathway, while HIF-2α upregulation under hypoxia activates lipid synthesis, promoting the progression of non-alcoholic fatty liver disease and hepatocellular carcinoma (127).
The synthesis of fatty acids leads to increased production of neutral triacylglycerols (TAGs), stored as lipid droplets for energy. HIF-1 induces key enzymes such asAGPAT2 and lipin-1, which facilitate LD accumulation and viability, also contributing to chemoresistance in hypoxic environments. The products of AGPAT2 can further be utilized for new membrane formation. Under hypoxia, unsaturated fatty acid oleate is preferentially released from TAGs into the phospholipid pool to balance saturated lipid accumulation (128). In addition, lipid signaling molecules such as sphingosine kinase 1 can stimulate HIF-1 activity. In summary, A2AR regulates dyslipidemia in OSA through the PI3K/Akt/HIF-1 pathway, highlighting its potential as a therapeutic target in managing lipid metabolism disorders.
4. Discussion
Adenosine, deriving from ATP degradation, mediates its physiological effects through four distinct subtypes of G-protein-coupled receptors named A1R, A2AR, A2BR and A3R. Previous research has shown that hypoxia and inflammation can lead to the accumulation of extracellular ATP/ADP due to the cell membrane damage (129). Consequently, it is important to investigate the pharmacological effects of adenosine receptors in the context of OSA (129-135) (Table I). During sleep apnea, adenosine is released as a response to hypoxia, which can promote sleep and reduce apnea episodes through the activation of A1R/A3R (131) and the inhibition of A2AR (136). High doses of caffeine, a well-known adenosine receptor antagonist, may provide an improved means of apnea management (137). In addition, adenosine plays a role in sensitizing the carotid body during intermittent hypoxia (134). Caffeine has been shown to decrease both baseline and hypoxia-induced (5% O2) chemosensory activity in the carotid sinus nerve of rats with intermittent hypoxia (134). Therefore, blocking adenosine receptors and modulating adenosine metabolism in the carotid body could aid in the management of sleep apnea (135). Other research indicates that increased expression of A2AR during hypoxia may help protect cells from the damaging effects of low oxygen levels (138). Conversely, A2AR deficiency has been linked to airway inflammation and hyperresponsiveness (139). Mirtazapine, a prescription drug that acts as an A2AR antagonist, appears to be ineffective in markedly improving sleep apnea and may even contribute to weight gain, potentially worsening OSA (140). There is speculation that appropriate use of A2AR agonists could inhibit the PI3K/Akt pathway, indirectly reducing the expression of HIF-1 and pro-inflammatory cytokines while enhancing lipid metabolism. Notably, the inhibitory effect of A2AR activation on respiratory drive appears to vary with age. Thus, further research is needed to clarify the pharmacological effects of A2AR in the treatment of OSA and clinical trials should be conducted to explore its potential therapeutic applications.
Intermittent hypoxia in OSA can contribute to inflammation-related cardiac metabolic diseases, with vascular inflammation and remodeling linked to increased leukocyte-endothelial cell interactions and T cell activation. While HIF-1 is expressed in unstimulated cells, NECA (an A2AR agonist) does not enhance HIF-1 mRNA expression in the absence of LPS stimulation. This indicates that LPS-induced A2AR expression crucial for the LPS/NECA-mediated upregulation of HIF-1. The NF-κB pathway is influenced by HIF-1, acting as a potent inflammatory activator that drives the release of TNF, IL-6, IL-8 and C-C motif chemokine ligand 2/monocyte chemoattractant protein-1 (MCP-1). Although OSA is hypothesized to elevate ROS levels, more evidence is necessary to fully support this (141). HIF-1 regulates the expression of various factors, including EPO, VEGF, inducible nitric oxide synthase and heme oxygenase, along with molecules involved in glucose metabolism, mitochondrial function and cellular adaptation to intermittent hypoxia during oxidative stress. Inhibition of HIF-1 reduces the expression of VEGF and Bcl2 interacting protein 3, thereby protecting against delayed cell death. Notably, the protective effects mediated by the PI3K/Akt and HIF-1 pathways may be reversed in the hypoxic microenvironment of cancer, highlighting the regulatory role of A2AR on HIF-1 as a potential therapeutic target for OSA. Intermittent hypoxia can downregulate endothelial nitric oxide synthase and enhance endothelin-1 production through the Erk1/2 pathway, while also increasing phosphorylation via the PI3K/Akt pathway, leading to endothelial dysfunction (142). Beyond the influences of inflammation and lipid metabolism, further investigation into the pharmacological effects of the PI3K/Akt/HIF-1 pathway on glucose metabolism, mitochondrial function and cell apoptosis in relation to OSA is warranted.
In addition, repeated exposure to hypoxic conditions can lead to significant alterations in gene transcription and post-translational protein modifications, resulting in changes in plasma concentrations of lipids, proteins and other biological compounds (143). The advent of large metabolomic datasets has enabled the use of metabolites as biomarkers for disease progression (46). Research indicates that the pathogenesis of cardiovascular disease and metabolic complications associated with OSA may be markedly linked to specific metabolic changes. Mendelian randomization studies have identified associations between OSA and >10 metabolites, including the plasma metabolite 3-Dehydrocarnitine. The biosynthetic pathway of valine, leucine and isoleucine is implicated in OSA pathogenesis (144). However, there is a lack of exploration into how inflammation alters metabolic pathways in the development of OSA and its complications. Therefore, understanding the involvement of metabolites in inflammation is critical for unraveling OSA pathogenesis and identifying potential therapeutic targets (145).
In the clinical management of OSA, a variety of treatment approaches are currently available, each with its own distinct characteristics and implications. These treatment modalities primarily encompass continuous positive airway pressure (CPAP), oral appliance therapy, surgical interventions, lifestyle changes, drug treatment and hypoglossal nerve stimulation. CPAP is effective for moderate to severe OSA, improving symptoms and quality of life, but requires maintenance and can be uncomfortable (146,147). Oral appliances are portable and work well for mild OSA, though less effective for severe cases and may cause discomfort (148). Surgical options, such as uvulopalatopharyngoplasty, offer long-term benefits for patients with clear anatomical issues but carry risks and long recovery times (149,150). Lifestyle changes complement treatments with no major side effects, though their impact is gradual (151). Drug treatments may help mild OSA but are generally limited (152-154). Hypoglossal nerve stimulation is a less invasive alternative to CPAP or surgery, with good tolerance but concerns over cost and long-term efficacy (155). Consequently, given the diverse nature of these treatment options for OSA, it becomes of utmost importance to conduct a comprehensive comparison of different treatments for OSA (Table II) Such a comparison can provide valuable insights for healthcare providers and patients alike, enabling them to make more informed decisions regarding the most appropriate treatment approach based on individual circumstances (149).
The relationship between OSA and lipid profiles remains a complex area of study. While some clinical evidence indicates that CPAP devices can improve certain aspects of dyslipidemia by alleviating apnea-hypopnea, much of this data are derived from observational studies. CPAP may lead to reductions in inflammatory cytokines and inhibit lipid peroxidation, as evidenced by decreased levels of malondialdehyde and endothelial lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) (156). However, its effect on oxidized low-density lipoprotein (oxLDL) levels in patients with OSA with comorbidities appears negligible after one year of treatment (157). By contrast, randomized controlled trials assessing the effects of CPAP on lipid metabolism have produced inconclusive results, with two meta-analyses yielding conflicting evidence (57). Large population-based studies generally highlight a relationship between OSA and dyslipidemia; however, they often fail to adequately control for confounding factors such as diet, exercise and the use of lipid-lowering medications, introducing potential biases. Thus, there is a pressing need for new clinical studies with larger sample sizes that account for these variables. Such research should also facilitate the screening of comorbid conditions for more timely interventions, ultimately enhancing patient outcomes.
The use of A2AR agonists presents several limitations and challenges that must be carefully addressed. These agonists may exhibit non-specific effects on other adenosine receptors, potentially leading to side effects, so enhancing their specificity and selectivity is crucial. Additionally, the pharmacological efficacy and pharmacokinetics of A2AR agonists can impact their clinical application, making it essential to optimize drug structure to improve bioavailability, stability and half-life (158). This optimization could enhance efficacy and reduce dosing frequency, contributing to more convenient and effective treatment. Safety and tolerability are paramount, as long-term use of A2AR agonists may result in adverse reactions, particularly in the cardiovascular, digestive and nervous systems. Comprehensive evaluations of safety and tolerability are necessary to establish appropriate dosing and treatment plans (159). Furthermore, given the complex and multifactorial nature of inflammatory diseases, relying solely on A2AR agonists may yield limited effectiveness (160). Exploring combination therapies with other drugs represents a promising avenue for future research, aiming to enhance overall treatment efficacy while minimizing side effects. In addition, individual patient responses to A2AR agonists can vary markedly, making it vital to understand each patient's genetic background, disease subtype and immune status for developing personalized treatment plans, which could substantially improve treatment effectiveness and reduce adverse reactions (161).
The present study made significant contributions to our understanding of OSA and its associated aspects, proving valuable to the scientific community. It explored various elements related to OSA, examining traditional mechanisms such as intermittent hypoxia and sleep disruptions while focusing on adenosine receptors, particularly A2AR. By integrating the effects of A2AR on inflammation, lipid and glucose metabolism, mitochondrial function and cell apoptosis through the PI3K/Akt/HIF-1 pathway, the current review presented a comprehensive view of the complex interactions within OSA. This holistic understanding aids researchers in developing more targeted hypotheses and experimental designs, revealing previously overlooked connections among physiological processes. In contrast to earlier reviews that primarily addressed basic OSA pathophysiology and the effects of intermittent hypoxia on vascular endothelial damage and systemic inflammation (162), the present review offered a deeper analysis of the molecular mechanisms of A2AR and broader implications. It built on this foundation by exploring the regulatory role of A2AR in the PI3K/Akt/HIF-1 pathway and its consequences for inflammation and lipid metabolism, providing a more nuanced perspective on OSA. In addition, the present review emphasized the intricate interplay between inflammation and metabolic changes in OSA. It highlighted how specific metabolites contribute to OSA pathogenesis and how inflammation affects metabolic pathways, addressing an area less explored in prior research. This focused approach encourages further investigation into these interactions, potentially leading to the identification of novel biomarkers for early diagnosis and new therapeutic targets for more effective treatments. By contrast, another previous review centered on the relationship of OSA with cardiovascular diseases, examining how OSA-induced changes in blood pressure, lipid profiles and endothelial function contribute to cardiovascular morbidity (57). While this is an important area, it did not comprehensively address the role of metabolites or the interaction between inflammation and metabolism as our review does. By specifically focusing on these aspects, the present study underscored the significance of understanding metabolite involvement in inflammation, which could reveal new insights into the pathogenesis of OSA and therapeutic opportunities. Additionally, the present review discussed the potential clinical applications of A2AR agonists in treating inflammatory diseases beyond COPD and asthma, highlighting their limitations and challenges. By broadening the scope of the clinical utility of A2AR, it provided critical insights for researchers and clinicians, guiding future research and optimizing the use of A2AR agonists in various disease contexts. By contrast, a previous review primarily focused on traditional OSA treatment options, such as CPAP, oral appliances and surgery, without exploring the emerging potential of A2AR agonists (163). In conclusion, the present review stands out for its comprehensive integration of multiple mechanisms, its focus on the interplay between inflammation and metabolism and its exploration of clinical applications and limitations. Compared to earlier reviews, it offers a more detailed and multi-faceted perspective, advancing knowledge in the field of OSA and providing valuable resources for future research and clinical applications.
5. Future directions
Although the significance of inflammation and metabolic changes in OSA is recognized, there are still several areas that require further research. First, the mechanism of A2AR in OSA needs deeper exploration. This includes understanding its effect on different cell types and signaling pathways. Additionally, the effects of A2AR on different age groups and its influence on respiratory drive demand further investigation. Exploring the potential value of A2AR agonists in treating OSA and determining the optimal dosage and treatment duration is crucial. Large-scale randomized controlled trials should be conducted to evaluate the effectiveness of CPAP and other treatment methods on various aspects of patients with OSA. Currently, the effect of CPAP on lipid metabolism is controversial and most existing studies are observational, lacking large-scale randomized controlled trials to determine its exact efficacy. Finally, by employing modern technologies such as metabolomics and genomics, further research can be carried out on the detailed interaction mechanisms between inflammation and metabolism in OSA. Studying OSA-related metabolites and biomarkers can improve the accuracy of early diagnosis, enable timely intervention, reduce the occurrence of complications and help discover new therapeutic targets. However, the role of specific metabolites in the pathogenesis of OSA still needs further clarification. In summary, further research is needed in multiple aspects of OSA, including the mechanism of A2AR, the effectiveness of treatment methods such asCPAP and the role of metabolites in pathogenesis. These efforts will enhance our understanding of OSA and lead to improved diagnosis and treatment strategies.
6. Conclusion
OSA is intricately linked to inflammation and dyslipidemia, conditions often exacerbated by OSA-related comorbidities. The present study delved into the molecular mechanisms underlying OSA, emphasizing the role of A2AR in modulating these processes. It found that A2AR exerts an inhibitory effect on the PI3K/Akt/HIF-1 pathway, markedly influencing inflammation and lipid metabolism associated with OSA. This supports the hypothesis that adenosine receptors are the main molecular process drivers of OSA onset. Furthermore, A2AR activation appears to stimulate key factors such as Akt and HIF-1, which are known to play roles in the regulation of intermittent hypoxia. However, the complexity of interactions among these factors suggests that further investigation is warranted. The potential of A2AR as an anti-inflammatory agent and a regulator of lipid metabolism, particularly through its influence on the PI3K-Akt-HIF-1 pathway under hypoxic conditions, is promising. Additionally, non-selective adenosine receptor antagonists, such as caffeine, have been shown to improve sleep apnea symptoms, indicating that A2AR may serve as a viable target for pharmacological interventions aimed at alleviating inflammation and dyslipidemia associated with OSA. Future research should explore the implications of OSA-related comorbidities, particularly the pathways involved in OSA-associated dyslipidemia. These insights could illuminate the cellular processes driving OSA and aid in identifying potential therapeutic targets for prevention and treatment.
Acknowledgements
Not applicable.
Funding
Funding: The present study was supported by National Natural Science Foundation of China (grant nos. 82074576 and 81704190), Sichuan Provincial Administration of Traditional Chinese Medicine (grant no. 2021MS444) and Key Laboratory of Sports Medicine of Sichuan Province (grant no. 2023-A037).
Availability of data and materials
Not applicable.
Authors' contributions
NM wrote the first draft of the manuscript. PL prepared the review tables and figures. NL, YH, NM and LK were responsible for critical revisions of the article. YH contributed to the acquisition of funds. Data authentication is not applicable. All authors read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Gottlieb DJ and Punjabi NM: Diagnosis and Management of Obstructive Sleep Apnea: A review. JAMA. 323:1389–1400. 2020.PubMed/NCBI View Article : Google Scholar | |
Benjafield AV, Ayas NT, Eastwood PR, Heinzer R, Ip MSM, Morrell MJ, Nunez CM, Patel SR, Penzel T, Pépin JL, et al: Estimation of the global prevalence and burden of obstructive sleep apnoea: A literature-based analysis. Lancet Respir Med. 7:687–698. 2019.PubMed/NCBI View Article : Google Scholar | |
Mohamadian M, Chiti H, Shoghli A, Biglari S, Parsamanesh N and Esmaeilzadeh A: COVID-19: Virology, biology and novel laboratory diagnosis. J Gene Med. 23(e3303)2021.PubMed/NCBI View Article : Google Scholar | |
Franklin KA and Lindberg E: Obstructive sleep apnea is a common disorder in the population-a review on the epidemiology of sleep apnea. J Thorac Dis. 7:1311–1322. 2015.PubMed/NCBI View Article : Google Scholar | |
Gu X, Zhang J, Shi Y, Shen H, Li Y, Chen Y and Liang L: ESM1/HIF-1α pathway modulates chronic intermittent hypoxia-induced non-small-cell lung cancer proliferation, stemness and epithelial-mesenchymal transition. Oncol Rep. 45:1226–1234. 2021.PubMed/NCBI View Article : Google Scholar | |
Hirsch Allen AJ, Kendzerska T, Bhatti P, Jen R, Myers R, Hajipour M, van Eeden SF and Ayas N: Obstructive sleep apnea severity, circulating biomarkers, and cancer risk. J Clin Sleep Med. 20:1415–1422. 2024.PubMed/NCBI View Article : Google Scholar | |
Ni W, Niu Y, Cao S, Fan C, Fan J, Zhu L and Wang X: Intermittent hypoxia exacerbates anxiety in high-fat diet-induced diabetic mice by inhibiting TREM2-regulated IFNAR1 signaling. J Neuroinflammation. 21(166)2024.PubMed/NCBI View Article : Google Scholar | |
Yang M, Cai W, Lin Z, Tuohuti A and Chen X: Intermittent Hypoxia Promotes TAM-Induced Glycolysis in Laryngeal Cancer Cells via Regulation of HK1 Expression through Activation of ZBTB10. Int J Mol Sci. 24(14808)2023.PubMed/NCBI View Article : Google Scholar | |
Cheong AJY, Tan BKJ, Teo YH, Tan NKW, Yap DWT, Sia CH, Ong TH, Leow LC, See A and Toh ST: Obstructive Sleep Apnea and Lung Cancer: A Systematic Review and Meta-Analysis. Ann Am Thorac Soc. 19:469–475. 2022.PubMed/NCBI View Article : Google Scholar | |
Tang SS, Liang CH, Liu YL, Wei W, Deng XR, Shi XY, Wang LM, Zhang LJ and Yuan HJ: Intermittent hypoxia is involved in gut microbial dysbiosis in type 2 diabetes mellitus and obstructive sleep apnea-hypopnea syndrome. World J Gastroenterol. 28:2320–2333. 2022.PubMed/NCBI View Article : Google Scholar | |
Almendros I, Martinez-Garcia MA, Farré R and Gozal D: Obesity, sleep apnea, and cancer. Int J Obes (Lond). 44:1653–1667. 2020.PubMed/NCBI View Article : Google Scholar | |
Prabhakar NR, Peng YJ and Nanduri J: Hypoxia-inducible factors and obstructive sleep apnea. J Clin Invest. 130:5042–5051. 2020.PubMed/NCBI View Article : Google Scholar | |
Moriondo G, Soccio P, Minoves M, Scioscia G, Tondo P, Foschino Barbaro MP, Pépin JL, Briançon-Marjollet A and Lacedonia D: Intermittent Hypoxia Mediates Cancer Development and Progression Through HIF-1 and miRNA Regulation. Arch Bronconeumol. 59:629–637. 2023.PubMed/NCBI View Article : Google Scholar : (In English, Spanish). | |
Wang N, Su X, Sams D, Prabhakar NR and Nanduri J: P300/CBP Regulates HIF-1-Dependent Sympathetic Activation and Hypertension by Intermittent Hypoxia. Am J Respir Cell Mol Biol. 70:110–118. 2024.PubMed/NCBI View Article : Google Scholar | |
Halpin-Veszeleiova K and Hatfield SM: Oxygenation and A2AR blockade to eliminate hypoxia/HIF-1α-adenosinergic immunosuppressive axis and improve cancer immunotherapy. Curr Opin Pharmacol. 53:84–90. 2020.PubMed/NCBI View Article : Google Scholar | |
Liu Z, Yan S, Wang J, Xu Y, Wang Y, Zhang S, Xu X, Yang Q, Zeng X, Zhou Y, et al: Endothelial adenosine A2a receptor-mediated glycolysis is essential for pathological retinal angiogenesis. Nat Commun. 8(584)2017.PubMed/NCBI View Article : Google Scholar | |
Belaidi E, Morand J, Gras E, Pépin JL and Godin-Ribuot D: Targeting the ROS-HIF-1-endothelin axis as a therapeutic approach for the treatment of obstructive sleep apnea-related cardiovascular complications. Pharmacol Ther. 168:1–11. 2016.PubMed/NCBI View Article : Google Scholar | |
Guan S, Suman S, Amann JM, Wu R, Carbone DP, Wang J and Dikov MM: Metabolic reprogramming by adenosine antagonism and implications in non-small cell lung cancer therapy. Neoplasia. 32(100824)2022.PubMed/NCBI View Article : Google Scholar | |
Bruzzese L, Fromonot J, By Y, Durand-Gorde JM, Condo J, Kipson N, Guieu R, Fenouillet E and Ruf J: NF-κB enhances hypoxia-driven T-cell immunosuppression via upregulation of adenosine A(2A) receptors. Cell Signal. 26:1060–1067. 2014.PubMed/NCBI View Article : Google Scholar | |
Sitkovsky MV, Kjaergaard VJ, Lukashev D and Ohta A: Hypoxia-adenosinergic immunosuppression: Tumor protection by T regulatory cells and cancerous tissue hypoxia. Clin Cancer Res. 14:5947–5952. 2008.PubMed/NCBI View Article : Google Scholar | |
Ke RH, Xiong J, Liu Y and Ye ZR: Adenosine A2a receptor induced gliosis via Akt/NF-kappaB pathway in vitro. Neurosci Res. 65:280–285. 2009.PubMed/NCBI View Article : Google Scholar | |
Eckle T, Kewley EM, Brodsky KS, Tak E, Bonney S, Gobel M, Anderson D, Glover LE, Riegel AK, Colgan SP and Eltzschig HK: Identification of hypoxia-inducible factor HIF-1A as transcriptional regulator of the A2B adenosine receptor during acute lung injury. J Immunol. 192:1249–1256. 2014.PubMed/NCBI View Article : Google Scholar | |
Steingold JM and Hatfield SM: Targeting Hypoxia-A2A Adenosinergic Immunosuppression of Antitumor T Cells During Cancer Immunotherapy. Front Immunol. 11(570041)2020.PubMed/NCBI View Article : Google Scholar | |
Castillo CA, León D, Ruiz MA, Albasanz JL and Martín M: Modulation of adenosine A1 and A2A receptors in C6 glioma cells during hypoxia: Involvement of endogenous adenosine. J Neurochem. 105:2315–2329. 2008.PubMed/NCBI View Article : Google Scholar | |
Pang L, Ng KT, Liu J, Yeung WO, Zhu J, Chiu TS, Liu H, Chen Z, Lo CM and Man K: Plasmacytoid dendritic cells recruited by HIF-1α/eADO/ADORA1 signaling induce immunosuppression in hepatocellular carcinoma. Cancer Lett. 522:80–92. 2021.PubMed/NCBI View Article : Google Scholar | |
Orrù G, Storari M, Scano A, Piras V, Taibi R and Viscuso D: Obstructive Sleep Apnea, oxidative stress, inflammation and endothelial dysfunction-An overview of predictive laboratory biomarkers. Eur Rev Med Pharmacol Sci. 24:6939–6948. 2020.PubMed/NCBI View Article : Google Scholar | |
Fang S, Sun S, Cai H, Zou X, Wang S, Hao X, Wan X, Tian J, Li Z, He Z, et al: IRGM/Irgm1 facilitates macrophage apoptosis through ROS generation and MAPK signal transduction: Irgm1(+/-) mice display increases atherosclerotic plaque stability. Theranostics. 11:9358–9375. 2021.PubMed/NCBI View Article : Google Scholar | |
Lin YF, Li MH, Huang RH, Zhang SZ, Xu XF, Zhou HM, Liu MH, Liao XX, Liao LZ, Guo Y and Zhuang XD: GP73 enhances the ox-LDL-induced inflammatory response in THP-1 derived macrophages via affecting NLRP3 inflammasome signaling. Int J Cardiol. 387(131109)2023.PubMed/NCBI View Article : Google Scholar | |
Balzan S and Lubrano V: LOX-1 receptor: A potential link in atherosclerosis and cancer. Life Sci. 198:79–86. 2018.PubMed/NCBI View Article : Google Scholar | |
Sylvers-Davie KL and Davies BSJ: Regulation of lipoprotein metabolism by ANGPTL3, ANGPTL4, and ANGPTL8. Am J Physiol Endocrinol Metab. 321:E493–E508. 2021.PubMed/NCBI View Article : Google Scholar | |
Li W and Wang W: Causal modulation of lipid metabolism may shape the inflammatory microenvironment and potentially augment immunotherapy: A comprehensive genetic landscape revealed by Mendelian randomization analysis. Int Immunol. 36:291–302. 2024.PubMed/NCBI View Article : Google Scholar | |
Yuan T, Yang T, Chen H, Fu D, Hu Y, Wang J, Yuan Q, Yu H, Xu W and Xie X: New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol. 20:247–260. 2019.PubMed/NCBI View Article : Google Scholar | |
Lara-Guzmán OJ, Gil-Izquierdo A, Medina S, Osorio E, Álvarez-Quintero R, Zuluaga N, Oger C, Galano JM, Durand T and Muñoz-Durango K: Oxidized LDL triggers changes in oxidative stress and inflammatory biomarkers in human macrophages. Redox Biol. 15:1–11. 2018.PubMed/NCBI View Article : Google Scholar | |
Incalza MA, D'Oria R, Natalicchio A, Perrini S, Laviola L and Giorgino F: Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul Pharmacol. 100:1–19. 2018.PubMed/NCBI View Article : Google Scholar | |
Xu D, Hu YH, Gou X, Li FY, Yang XY, Li YM and Chen F: Oxidative Stress and Antioxidative Therapy in Pulmonary Arterial Hypertension. Molecules. 27(3724)2022.PubMed/NCBI View Article : Google Scholar | |
Li M, Xin S, Gu R, Zheng L, Hu J, Zhang R and Dong H: Novel Diagnostic Biomarkers Related to Oxidative Stress and Macrophage Ferroptosis in Atherosclerosis. Oxid Med Cell Longev. 2022(8917947)2022.PubMed/NCBI View Article : Google Scholar | |
Kattoor AJ, Pothineni NVK, Palagiri D and Mehta JL: Oxidative Stress in Atherosclerosis. Curr Atheroscler Rep. 19(42)2017.PubMed/NCBI View Article : Google Scholar | |
Batty M, Bennett MR and Yu E: The Role of Oxidative Stress in Atherosclerosis. Cells. 11(3843)2022.PubMed/NCBI View Article : Google Scholar | |
Ranjbarvaziri S, Kooiker KB, Ellenberger M, Fajardo G, Zhao M, Vander Roest AS, Woldeyes RA, Koyano TT, Fong R, Ma N, et al: Altered Cardiac Energetics and Mitochondrial Dysfunction in Hypertrophic Cardiomyopathy. Circulation. 144:1714–1731. 2021.PubMed/NCBI View Article : Google Scholar | |
Tian L, Cao W, Yue R, Yuan Y, Guo X, Qin D, Xing J and Wang X: Pretreatment with Tilianin improves mitochondrial energy metabolism and oxidative stress in rats with myocardial ischemia/reperfusion injury via AMPK/SIRT1/PGC-1 alpha signaling pathway. J Pharmacol Sci. 139:352–360. 2019.PubMed/NCBI View Article : Google Scholar | |
Dikalova AE, Pandey A, Xiao L, Arslanbaeva L, Sidorova T, Lopez MG, Billings FT 4th, Verdin E, Auwerx J, Harrison DG and Dikalov SI: Mitochondrial Deacetylase Sirt3 Reduces Vascular Dysfunction and Hypertension While Sirt3 Depletion in Essential Hypertension Is Linked to Vascular Inflammation and Oxidative Stress. Circ Res. 126:439–452. 2020.PubMed/NCBI View Article : Google Scholar | |
Kent BD, Grote L, Ryan S, Pépin JL, Bonsignore MR, Tkacova R, Saaresranta T, Verbraecken J, Lévy P, Hedner J and McNicholas WT: Diabetes mellitus prevalence and control in sleep-disordered breathing: The European Sleep Apnea Cohort (ESADA) study. Chest. 146:982–990. 2014.PubMed/NCBI View Article : Google Scholar | |
Giampá SQC, Lorenzi-Filho G and Drager LF: Obstructive sleep apnea and metabolic syndrome. Obesity (Silver Spring). 31:900–911. 2023.PubMed/NCBI View Article : Google Scholar | |
Lévy P, Kohler M, McNicholas WT, Barbé F, McEvoy RD, Somers VK, Lavie L and Pépin JL: Obstructive sleep apnoea syndrome. Nat Rev Dis Primers. 1(15015)2015.PubMed/NCBI View Article : Google Scholar | |
Unnikrishnan D, Jun J and Polotsky V: Inflammation in sleep apnea: An update. Rev Endocr Metab Disord. 16:25–34. 2015.PubMed/NCBI View Article : Google Scholar | |
Alterki A, Abu-Farha M, Al Shawaf E, Al-Mulla F and Abubaker J: Investigating the Relationship between Obstructive Sleep Apnoea, Inflammation and Cardio-Metabolic Diseases. Int J Mol Sci. 24(6807)2023.PubMed/NCBI View Article : Google Scholar | |
Chen Z, Zeng J, Pei X, Zhao J, Zhao F, Zhang G, Liang K, Li J and Zhao X: Causal Relationships Between Circulating Inflammatory Proteins and Obstructive Sleep Apnea: A Bidirectional Mendelian Randomization Study. Nat Sci Sleep. 16:787–800. 2024.PubMed/NCBI View Article : Google Scholar | |
Malicki M, Karuga FF, Szmyd B, Sochal M and Gabryelska A: Obstructive Sleep Apnea, Circadian Clock Disruption, and Metabolic Consequences. Metabolites. 13(60)2022.PubMed/NCBI View Article : Google Scholar | |
Gras E, Belaidi E, Briançon-Marjollet A, Pépin JL, Arnaud C and Godin-Ribuot D: Endothelin-1 mediates intermittent hypoxia-induced inflammatory vascular remodeling through HIF-1 activation. J Appl Physiol (1985). 120:437–443. 2016.PubMed/NCBI View Article : Google Scholar | |
Taylor CT, Kent BD, Crinion SJ, McNicholas WT and Ryan S: Human adipocytes are highly sensitive to intermittent hypoxia induced NF-kappaB activity and subsequent inflammatory gene expression. Biochem Biophys Res Commun. 447:660–665. 2014.PubMed/NCBI View Article : Google Scholar | |
Imamura T, Poulsen O and Haddad GG: Intermittent hypoxia induces murine macrophage foam cell formation by IKK-β-dependent NF-κB pathway activation. J Appl Physiol (1985). 121:670–677. 2016.PubMed/NCBI View Article : Google Scholar | |
Lv R, Zhao Y, Wang X, He Y, Dong N, Min X, Liu X, Yu Q, Yuan K, Yue H and Yin Q: GLP-1 analogue liraglutide attenuates CIH-induced cognitive deficits by inhibiting oxidative stress, neuroinflammation, and apoptosis via the Nrf2/HO-1 and MAPK/NF-κB signaling pathways. Int Immunopharmacol. 142(113222)2024.PubMed/NCBI View Article : Google Scholar | |
Murphy AM, Thomas A, Crinion SJ, Kent BD, Tambuwala MM, Fabre A, Pepin JL, Roche HM, Arnaud C and Ryan S: Intermittent hypoxia in obstructive sleep apnoea mediates insulin resistance through adipose tissue inflammation. Eur Respir J. 49(1601731)2017.PubMed/NCBI View Article : Google Scholar | |
Arnaud C, Poulain L, Lévy P and Dematteis M: Inflammation contributes to the atherogenic role of intermittent hypoxia in apolipoprotein-E knock out mice. Atherosclerosis. 219:425–431. 2011.PubMed/NCBI View Article : Google Scholar | |
Huang T, Sands SA, Stampfer MJ, Tworoger SS, Hu FB and Redline S: Insulin Resistance, Hyperglycemia, and Risk of Developing Obstructive Sleep Apnea in Men and Women in the United States. Ann Am Thorac Soc. 19:1740–1749. 2022.PubMed/NCBI View Article : Google Scholar | |
Meng X, Wen H and Lian L: Association between triglyceride glucose-body mass index and obstructive sleep apnea: A study from NHANES 2015-2018. Front Nutr. 11(1424881)2024.PubMed/NCBI View Article : Google Scholar | |
Barros D and García-Río F: Obstructive sleep apnea and dyslipidemia: From animal models to clinical evidence. Sleep. 42(zsy236)2019.PubMed/NCBI View Article : Google Scholar | |
Tang H, Zhou Q, Zheng F, Wu T, Tang YD and Jiang J: The Causal Effects of Lipid Profiles on Sleep Apnea. Front Nutr. 9(910690)2022.PubMed/NCBI View Article : Google Scholar | |
Sun J, Hu J, Tu C, Zhong A and Xu H: Obstructive Sleep Apnea Susceptibility Genes in Chinese Population: A Field Synopsis and Meta-Analysis of Genetic Association Studies. PLoS One. 10(e0135942)2015.PubMed/NCBI View Article : Google Scholar | |
Uyrum E, Balbay O, Annakkaya AN, Gulec Balbay E, Silan F and Arbak P: The relationship between obstructive sleep apnea syndrome and apolipoprotein E genetic variants. Respiration. 89:195–200. 2015.PubMed/NCBI View Article : Google Scholar | |
Meszaros M and Bikov A: Obstructive Sleep Apnoea and Lipid Metabolism: The Summary of Evidence and Future Perspectives in the Pathophysiology of OSA-Associated Dyslipidaemia. Biomedicines. 10(2754)2022.PubMed/NCBI View Article : Google Scholar | |
Burke SL, Hu T, Spadola CE, Li T, Naseh M, Burgess A and Cadet T: Mild cognitive impairment: associations with sleep disturbance, apolipoprotein e4, and sleep medications. Sleep Med. 52:168–176. 2018.PubMed/NCBI View Article : Google Scholar | |
Huguenard CJC, Cseresznye A, Darcey T, Nkiliza A, Evans JE, Hazen SL, Mullan M, Crawford F and Abdullah L: Age and APOE affect L-carnitine system metabolites in the brain in the APOE-TR model. Front Aging Neurosci. 14(1059017)2023.PubMed/NCBI View Article : Google Scholar | |
Turner AD, Locklear CE, Oruru D, Briggs AQ, Bubu OM and Seixas A: Exploring the combined effects of sleep apnea and APOE-e4 on biomarkers of Alzheimer's disease. Front Aging Neurosci. 14(1017521)2023.PubMed/NCBI View Article : Google Scholar | |
Kim JK, Fillmore JJ, Chen Y, Yu C, Moore IK, Pypaert M, Lutz EP, Kako Y, Velez-Carrasco W, Goldberg IJ, et al: Tissue-specific overexpression of lipoprotein lipase causes tissue-specific insulin resistance. Proc Natl Acad Sci USA. 98:7522–7527. 2001.PubMed/NCBI View Article : Google Scholar | |
Wang N, Shi XF, Khan SA, Wang B, Semenza GL, Prabhakar NR and Nanduri J: Hypoxia-inducible factor-1 mediates pancreatic β-cell dysfunction by intermittent hypoxia. Am J Physiol Cell Physiol. 319:C922–C932. 2020.PubMed/NCBI View Article : Google Scholar | |
Fang Y, Zhang Q, Tan J, Li L, An X and Lei P: Intermittent hypoxia-induced rat pancreatic β-cell apoptosis and protective effects of antioxidant intervention. Nutr Diabetes. 4(e131)2014.PubMed/NCBI View Article : Google Scholar | |
Dempsey JA, Veasey SC, Morgan BJ and O'Donnell CP: Pathophysiology of sleep apnea. Physiol Rev. 90:47–112. 2010.PubMed/NCBI View Article : Google Scholar | |
Zeng S, Wang Y, Ai L, Huang L, Liu Z, He C, Bai Q and Li Y: Chronic intermittent hypoxia-induced oxidative stress activates TRB3 and phosphorylated JNK to mediate insulin resistance and cell apoptosis in the pancreas. Clin Exp Pharmacol Physiol. 51(e13843)2024.PubMed/NCBI View Article : Google Scholar | |
Nascimento DC, Viacava PR, Ferreira RG, Damaceno MA, Piñeros AR, Melo PH, Donate PB, Toller-Kawahisa JE, Zoppi D, Veras FP, et al: Sepsis expands a CD39(+) plasmablast population that promotes immunosuppression via adenosine-mediated inhibition of macrophage antimicrobial activity. Immunity. 54:2024–2041.e8. 2021.PubMed/NCBI View Article : Google Scholar | |
Pasquini S, Contri C, Borea PA, Vincenzi F and Varani K: Adenosine and Inflammation: Here, There and Everywhere. Int J Mol Sci. 22(7685)2021.PubMed/NCBI View Article : Google Scholar | |
Haskó G, Linden J, Cronstein B and Pacher P: Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 7:759–770. 2008.PubMed/NCBI View Article : Google Scholar | |
Junqueira SC, Dos Santos Coelho I, Lieberknecht V, Cunha MP, Calixto JB, Rodrigues ALS, Santos ARS and Dutra RC: Inosine, an Endogenous Purine Nucleoside, Suppresses Immune Responses and Protects Mice from Experimental Autoimmune Encephalomyelitis: A Role for A2A Adenosine Receptor. Mol Neurobiol. 54:3271–3285. 2017.PubMed/NCBI View Article : Google Scholar | |
Impellizzeri D, Di Paola R, Esposito E, Mazzon E, Paterniti I, Melani A, Bramanti P, Pedata F and Cuzzocrea S: CGS 21680, an agonist of the adenosine (A2A) receptor, decreases acute lung inflammation. Eur J Pharmacol. 668:305–316. 2011.PubMed/NCBI View Article : Google Scholar | |
Lu Y, Zhu W, Zhang GX, Chen JC, Wang QL, Mao MY, Deng SC, Jin LP, Liu H and Kuang YH: Adenosine A2A receptor activation regulates the M1 macrophages activation to initiate innate and adaptive immunity in psoriasis. Clin Immunol. 266(110309)2024.PubMed/NCBI View Article : Google Scholar | |
Mohsenin A, Mi T, Xia Y, Kellems RE, Chen JF and Blackburn MR: Genetic removal of the A2A adenosine receptor enhances pulmonary inflammation, mucin production, and angiogenesis in adenosine deaminase-deficient mice. Am J Physiol Lung Cell Mol Physiol. 293:L753–L761. 2007.PubMed/NCBI View Article : Google Scholar | |
Welihinda AA, Kaur M, Raveendran KS and Amento EP: Enhancement of inosine-mediated A(2A)R signaling through positive allosteric modulation. Cell Signal. 42:227–235. 2018.PubMed/NCBI View Article : Google Scholar | |
Zhao N, Shao Z, Xia G, Liu H, Zhang L, Zhao X, Dang S, Qian L, Xu W, Yu Z and Wang R: Protective role of the CD73-A2AR axis in cirrhotic cardiomyopathy through negative feedback regulation of the NF-κB pathway. Front Immunol. 15(1428551)2024.PubMed/NCBI View Article : Google Scholar | |
Correale P, Caracciolo M, Bilotta F, Conte M, Cuzzola M, Falcone C, Mangano C, Falzea AC, Iuliano E, Morabito A, et al: Therapeutic effects of adenosine in high flow 21% oxygen aereosol in patients with Covid19-pneumonia. PLoS One. 15(e0239692)2020.PubMed/NCBI View Article : Google Scholar | |
Tozzi M and Novak I: Purinergic Receptors in Adipose Tissue As Potential Targets in Metabolic Disorders. Front Pharmacol. 8(878)2017.PubMed/NCBI View Article : Google Scholar | |
Kotańska M, Dziubina A, Szafarz M, Mika K, Bednarski M, Nicosia N, Temirak A, Müller CE and Kieć-Kononowicz K: Preliminary Evidence of the Potent and Selective Adenosine A2B Receptor Antagonist PSB-603 in Reducing Obesity and Some of Its Associated Metabolic Disorders in Mice. Int J Mol Sci. 23(13439)2022.PubMed/NCBI View Article : Google Scholar | |
Zhao W, Ma L, Cai C and Gong X: Caffeine Inhibits NLRP3 Inflammasome Activation by Suppressing MAPK/NF-κB and A2aR Signaling in LPS-Induced THP-1 Macrophages. Int J Biol Sci. 15:1571–1581. 2019.PubMed/NCBI View Article : Google Scholar | |
Zohair B, Chraa D, Rezouki I, Benthami H, Razzouki I, Elkarroumi M, Olive D, Karkouri M and Badou A: The immune checkpoint adenosine 2A receptor is associated with aggressive clinical outcomes and reflects an immunosuppressive tumor microenvironment in human breast cancer. Front Immunol. 14(1201632)2023.PubMed/NCBI View Article : Google Scholar | |
Leiva A, Guzmán-Gutiérrez E, Contreras-Duarte S, Fuenzalida B, Cantin C, Carvajal L, Salsoso R, Gutiérrez J, Pardo F and Sobrevia L: Adenosine receptors: Modulators of lipid availability that are controlled by lipid levels. Mol Aspects Med. 55:26–44. 2017.PubMed/NCBI View Article : Google Scholar | |
DeOliveira CC, Paiva Caria CR, Ferreira Gotardo EM, Ribeiro ML and Gambero A: Role of A(1) and A(2A) adenosine receptor agonists in adipose tissue inflammation induced by obesity in mice. Eur J Pharmacol. 799:154–159. 2017.PubMed/NCBI View Article : Google Scholar | |
Chen S, Fan J, Xie P, Ahn J, Fernandez M, Billingham LK, Miska J, Wu JD, Wainwright DA, Fang D, et al: CD8+ T cells sustain antitumor response by mediating crosstalk between adenosine A2A receptor and glutathione/GPX4. J Clin Invest. 134(e170071)2024.PubMed/NCBI View Article : Google Scholar | |
Bruzzese A, Dalton JAR and Giraldo J: Insights into adenosine A2A receptor activation through cooperative modulation of agonist and allosteric lipid interactions. PLoS Comput Biol. 16(e1007818)2020.PubMed/NCBI View Article : Google Scholar | |
Friedman B, Corciulo C, Castro CM and Cronstein BN: Adenosine A2A receptor signaling promotes FoxO associated autophagy in chondrocytes. Sci Rep. 11(968)2021.PubMed/NCBI View Article : Google Scholar | |
Cai Y, Li H, Liu M, Pei Y, Zheng J, Zhou J, Luo X, Huang W, Ma L, Yang Q, et al: Disruption of adenosine 2A receptor exacerbates NAFLD through increasing inflammatory responses and SREBP1c activity. Hepatology. 68:48–61. 2018.PubMed/NCBI View Article : Google Scholar | |
Kong LR, Chen XH, Sun Q, Zhang KY, Xu L, Ding L, Zhou YP, Zhang ZB, Lin JR and Gao PJ: Loss of C3a and C5a receptors promotes adipocyte browning and attenuates diet-induced obesity via activating inosine/A2aR pathway. Cell Rep. 42(112078)2023.PubMed/NCBI View Article : Google Scholar | |
Lim J, Iyer A, Suen JY, Seow V, Reid RC, Brown L and Fairlie DP: C5aR and C3aR antagonists each inhibit diet-induced obesity, metabolic dysfunction, and adipocyte and macrophage signaling. FASEB J. 27:822–831. 2013.PubMed/NCBI View Article : Google Scholar | |
Kim K, Im H, Son Y, Kim M, Tripathi SK, Jeong LS and Lee YH: Anti-obesity effects of the dual-active adenosine A(2A)/A(3) receptor-ligand LJ-4378. Int J Obes (Lond). 46:2128–2136. 2022.PubMed/NCBI View Article : Google Scholar | |
Shamsuzzaman A, Amin RS, Calvin AD, Davison D and Somers VK: Severity of obstructive sleep apnea is associated with elevated plasma fibrinogen in otherwise healthy patients. Sleep Breath. 18:761–766. 2014.PubMed/NCBI View Article : Google Scholar | |
Li H, Chen O, Ye Z, Zhang R, Hu H, Zhang N, Huang J, Liu W and Sun X: Inhalation of high concentrations of hydrogen ameliorates liver ischemia/reperfusion injury through A(2A) receptor mediated PI3K-Akt pathway. Biochem Pharmacol. 130:83–92. 2017.PubMed/NCBI View Article : Google Scholar | |
Huang X, Wu P, Huang F, Xu M, Chen M, Huang K, Li GP, Xu M, Yao D and Wang L: Baicalin attenuates chronic hypoxia-induced pulmonary hypertension via adenosine A(2A) receptor-induced SDF-1/CXCR4/PI3K/AKT signaling. J Biomed Sci. 24(52)2017.PubMed/NCBI View Article : Google Scholar | |
Dal-Cim T, Poluceno GG, Lanznaster D, de Oliveira KA, Nedel CB and Tasca CI: Guanosine prevents oxidative damage and glutamate uptake impairment induced by oxygen/glucose deprivation in cortical astrocyte cultures: involvement of A(1) and A(2A) adenosine receptors and PI3K, MEK, and PKC pathways. Purinergic Signal. 15:465–476. 2019.PubMed/NCBI View Article : Google Scholar | |
Wen MH, Wu MJ, Vinit S and Lee KZ: Modulation of Serotonin and Adenosine 2A Receptors on Intermittent Hypoxia-Induced Respiratory Recovery following Mid-Cervical Contusion in the Rat. J Neurotrauma. 36:2991–3004. 2019.PubMed/NCBI View Article : Google Scholar | |
Mathew OP: Apnea of prematurity: Pathogenesis and management strategies. J Perinatol. 31:302–310. 2011.PubMed/NCBI View Article : Google Scholar | |
Lv B, Yang L, Gao Y and Li G: Epoxyeicosatrienoic Acids Attenuate LPS-Induced NIH/3T3 Cell Fibrosis through the A(2A)R and PI3K/Akt Signaling Pathways. Bull Exp Biol Med. 177:185–189. 2024.PubMed/NCBI View Article : Google Scholar | |
Mori Y, Higuchi M, Masuyama N and Gotoh Y: Adenosine A2A receptor facilitates calcium-dependent protein secretion through the activation of protein kinase A and phosphatidylinositol-3 kinase in PC12 cells. Cell Struct Funct. 29:101–110. 2004.PubMed/NCBI View Article : Google Scholar | |
Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K and Tsatsanis C: Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J Immunol. 198:1006–1014. 2017.PubMed/NCBI View Article : Google Scholar | |
Troutman TD, Bazan JF and Pasare C: Toll-like receptors, signaling adapters and regulation of the pro-inflammatory response by PI3K. Cell Cycle. 11:3559–3567. 2012.PubMed/NCBI View Article : Google Scholar | |
Li Q, Wang G, Xiong SH, Cao Y, Liu B, Sun J, Li L, Mohammadtursun N, Yu H, Dong J and Wu J: Bu-Shen-Fang-Chuan formula attenuates cigarette smoke-induced inflammation by modulating the PI3K/Akt-Nrf2 and NF-κB signalling pathways. J Ethnopharmacol. 261(113095)2020.PubMed/NCBI View Article : Google Scholar | |
Li L, Qu Y, Mao M, Xiong Y and Mu D: The involvement of phosphoinositid 3-kinase/Akt pathway in the activation of hypoxia-inducible factor-1alpha in the developing rat brain after hypoxia-ischemia. Brain Res. 1197:152–158. 2008.PubMed/NCBI View Article : Google Scholar | |
Li L, Yin X, Ma N, Lin F, Kong X, Chi J and Feng Z: Desferrioxamine regulates HIF-1 alpha expression in neonatal rat brain after hypoxia-ischemia. Am J Transl Res. 6:377–383. 2014.PubMed/NCBI | |
Hutami IR, Izawa T, Khurel-Ochir T, Sakamaki T, Iwasa A and Tanaka E: Macrophage Motility in Wound Healing Is Regulated by HIF-1α via S1P Signaling. Int J Mol Sci. 22(8992)2021.PubMed/NCBI View Article : Google Scholar | |
Cramer T, Yamanishi Y, Clausen BE, Förster I, Pawlinski R, Mackman N, Haase VH, Jaenisch R, Corr M, Nizet V, et al: HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell. 112:645–657. 2003.PubMed/NCBI View Article : Google Scholar | |
Peyssonnaux C, Datta V, Cramer T, Doedens A, Theodorakis EA, Gallo RL, Hurtado-Ziola N, Nizet V and Johnson RS: HIF-1alpha expression regulates the bactericidal capacity of phagocytes. J Clin Invest. 115:1806–1815. 2005.PubMed/NCBI View Article : Google Scholar | |
Kim SY, Jeong E, Joung SM and Lee JY: PI3K/Akt contributes to increased expression of Toll-like receptor 4 in macrophages exposed to hypoxic stress. Biochem Biophys Res Commun. 419:466–471. 2012.PubMed/NCBI View Article : Google Scholar | |
Kunze R, Zhou W, Veltkamp R, Wielockx B, Breier G and Marti HH: Neuron-specific prolyl-4-hydroxylase domain 2 knockout reduces brain injury after transient cerebral ischemia. Stroke. 43:2748–2756. 2012.PubMed/NCBI View Article : Google Scholar | |
Majmundar AJ, Wong WJ and Simon MC: Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell. 40:294–309. 2010.PubMed/NCBI View Article : Google Scholar | |
Vgontzas AN, Papanicolaou DA, Bixler EO, Kales A, Tyson K and Chrousos GP: Elevation of plasma cytokines in disorders of excessive daytime sleepiness: Role of sleep disturbance and obesity. J Clin Endocrinol Metab. 82:1313–1316. 1997.PubMed/NCBI View Article : Google Scholar | |
Ryan S: Adipose tissue inflammation by intermittent hypoxia: Mechanistic link between obstructive sleep apnoea and metabolic dysfunction. J Physiol. 595:2423–2430. 2017.PubMed/NCBI View Article : Google Scholar | |
Toujani S, Kaabachi W, Mjid M, Hamzaoui K, Cherif J and Beji M: Vitamin D deficiency and interleukin-17 relationship in severe obstructive sleep apnea-hypopnea syndrome. Ann Thorac Med. 12:107–113. 2017.PubMed/NCBI View Article : Google Scholar | |
Gao C, Koko MYF, Ding M, Hong W, Li J, Dong N and Hui M: Intestinal alkaline phosphatase (IAP, IAP Enhancer) attenuates intestinal inflammation and alleviates insulin resistance. Front Immunol. 13(927272)2022.PubMed/NCBI View Article : Google Scholar | |
Xie Y, Shi X, Sheng K, Han G, Li W, Zhao Q, Jiang B, Feng J, Li J and Gu Y: PI3K/Akt signaling transduction pathway, erythropoiesis and glycolysis in hypoxia (Review). Mol Med Rep. 19:783–791. 2019.PubMed/NCBI View Article : Google Scholar | |
Rahman SMK, Uyama T, Hussain Z and Ueda N: Roles of Endocannabinoids and Endocannabinoid-Like Molecules in Energy Homeostasis and Metabolic Regulation: A Nutritional Perspective. Annu Rev Nutr. 41:177–202. 2021.PubMed/NCBI View Article : Google Scholar | |
Murillo-Rodríguez E: The Endocannabinoid System as Prognostic Biomarker of the Obstructive Sleep Apnea Morbidity in COVID-19-Recovered Individuals. Sleep Vigil. 5:205–211. 2021.PubMed/NCBI View Article : Google Scholar | |
Wu YQ, Wang B, Song L, Wang Q, Chen XY and Liu ZL: A study on the endogenous cannabinoid system synthetic and catabolic enzyme levels in patients with obstructive sleep apnea. Zhonghua Jie He He Hu Xi Za Zhi. 34:359–361. 2011.PubMed/NCBI(In Chinese). | |
Gonzaga CC, Gaddam KK, Ahmed MI, Pimenta E, Thomas SJ, Harding SM, Oparil S, Cofield SS and Calhoun DA: Severity of obstructive sleep apnea is related to aldosterone status in subjects with resistant hypertension. J Clin Sleep Med. 6:363–368. 2010.PubMed/NCBI | |
Reiss AB, Carsons SE, Anwar K, Rao S, Edelman SD, Zhang H, Fernandez P, Cronstein BN and Chan ES: Atheroprotective effects of methotrexate on reverse cholesterol transport proteins and foam cell transformation in human THP-1 monocyte/macrophages. Arthritis Rheum. 58:3675–3683. 2008.PubMed/NCBI View Article : Google Scholar | |
Lee EJ, Heo W, Kim JY, Kim H, Kang MJ, Kim BR, Kim JH, Park DY, Kim CH, Yoon JH and Cho HJ: Alteration of inflammatory mediators in the upper and lower airways under chronic intermittent hypoxia: Preliminary Animal Study. Mediators Inflamm. 2017(4327237)2017.PubMed/NCBI View Article : Google Scholar | |
Papandreou C: Independent associations between fatty acids and sleep quality among obese patients with obstructive sleep apnoea syndrome. J Sleep Res. 22:569–572. 2013.PubMed/NCBI View Article : Google Scholar | |
Lebkuchen A, Carvalho VM, Venturini G, Salgueiro JS, Freitas LS, Dellavance A, Martins FC, Lorenzi-Filho G, Cardozo KHM and Drager LF: Metabolomic and lipidomic profile in men with obstructive sleep apnoea: Implications for diagnosis and biomarkers of cardiovascular risk. Sci Rep. 8(11270)2018.PubMed/NCBI View Article : Google Scholar | |
Xu H, Zheng X, Qian Y, Guan J, Yi H, Zou J, Wang Y, Meng L, Zhao A, Yin S and Jia W: Metabolomics Profiling for Obstructive Sleep Apnea and Simple Snorers. Sci Rep. 6(30958)2016.PubMed/NCBI View Article : Google Scholar | |
Kamphorst JJ, Cross JR, Fan J, de Stanchina E, Mathew R, White EP, Thompson CB and Rabinowitz JD: Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc Natl Acad Sci USA. 110:8882–8887. 2013.PubMed/NCBI View Article : Google Scholar | |
Chen J, Chen J, Huang J, Li Z, Gong Y, Zou B, Liu X, Ding L, Li P, Zhu Z, et al: HIF-2α upregulation mediated by hypoxia promotes NAFLD-HCC progression by activating lipid synthesis via the PI3K-AKT-mTOR pathway. Aging (Albany NY). 11:10839–10860. 2019.PubMed/NCBI View Article : Google Scholar | |
Ackerman D, Tumanov S, Qiu B, Michalopoulou E, Spata M, Azzam A, Xie H, Simon MC and Kamphorst JJ: Triglycerides Promote Lipid Homeostasis during Hypoxic Stress by Balancing Fatty Acid Saturation. Cell Rep. 24:2596–2605.e5. 2018.PubMed/NCBI View Article : Google Scholar | |
Le TT, Berg NK, Harting MT, Li X, Eltzschig HK and Yuan X: Purinergic Signaling in Pulmonary Inflammation. Front Immunol. 10(1633)2019.PubMed/NCBI View Article : Google Scholar | |
Hanidziar D and Robson SC: Synapomorphic features of hepatic and pulmonary vasculatures include comparable purinergic signaling responses in host defense and modulation of inflammation. Am J Physiol Gastrointest Liver Physiol. 321:G200–G212. 2021.PubMed/NCBI View Article : Google Scholar | |
Guieu R, Deharo JC, Maille B, Crotti L, Torresani E, Brignole M and Parati G: Adenosine and the Cardiovascular System: The Good and the Bad. J Clin Med. 9(1366)2020.PubMed/NCBI View Article : Google Scholar | |
Vecchio EA, White PJ and May LT: The adenosine A(2B) G protein-coupled receptor: Recent advances and therapeutic implications. Pharmacol Ther. 198:20–33. 2019.PubMed/NCBI View Article : Google Scholar | |
Bahreyni A, Avan A, Shabani M, Ryzhikov M, Fiuji H, Soleimanpour S, Khazaei M and Hassanian SM: Therapeutic potential of A2 adenosine receptor pharmacological regulators in the treatment of cardiovascular diseases, recent progress, and prospective. J Cell Physiol. 234:1295–1299. 2019.PubMed/NCBI View Article : Google Scholar | |
Sacramento JF, Gonzalez C, Gonzalez-Martin MC and Conde SV: Adenosine Receptor Blockade by Caffeine Inhibits Carotid Sinus Nerve Chemosensory Activity in Chronic Intermittent Hypoxic Animals. Adv Exp Med Biol. 860:133–137. 2015.PubMed/NCBI View Article : Google Scholar | |
Conde SV, Monteiro EC and Sacramento JF: Purines and Carotid Body: New Roles in Pathological Conditions. Front Pharmacol. 8(913)2017.PubMed/NCBI View Article : Google Scholar | |
Li XC, Hong FF, Tu YJ, Li YA, Ma CY, Yu CY, Fang L, Chen JY, Li ZL, Bao SJ, et al: Blockade of adenosine A(2A) receptor alleviates cognitive dysfunction after chronic exposure to intermittent hypoxia in mice. Exp Neurol. 350(113929)2022.PubMed/NCBI View Article : Google Scholar | |
Aranda JV and Beharry KD: Pharmacokinetics, pharmacodynamics and metabolism of caffeine in newborns. Semin Fetal Neonatal Med. 25(101183)2020.PubMed/NCBI View Article : Google Scholar | |
Kobayashi S and Millhorn DE: Stimulation of expression for the adenosine A2A receptor gene by hypoxia in PC12 cells. A potential role in cell protection. J Biol Chem. 274:20358–20365. 1999.PubMed/NCBI View Article : Google Scholar | |
Nadeem A, Fan M, Ansari HR, Ledent C and Jamal Mustafa S: Enhanced airway reactivity and inflammation in A2A adenosine receptor-deficient allergic mice. Am J Physiol Lung Cell Mol Physiol. 292:L1335–L1344. 2007.PubMed/NCBI View Article : Google Scholar | |
Marshall NS, Yee BJ, Desai AV, Buchanan PR, Wong KK, Crompton R, Melehan KL, Zack N, Rao SG, Gendreau RM, et al: Two randomized placebo-controlled trials to evaluate the efficacy and tolerability of mirtazapine for the treatment of obstructive sleep apnea. Sleep. 31:824–831. 2008.PubMed/NCBI View Article : Google Scholar | |
Kent BD, Ryan S and McNicholas WT: Obstructive sleep apnea and inflammation: relationship to cardiovascular co-morbidity. Respir Physiol Neurobiol. 178:475–481. 2011.PubMed/NCBI View Article : Google Scholar | |
Peng L, Li Y, Li X, Du Y, Li L, Hu C, Zhang J, Qin Y, Wei Y and Zhang H: Extracellular vesicles derived from intermittent hypoxia-treated red blood cells impair endothelial function through regulating eNOS phosphorylation and ET-1 expression. Cardiovasc Drugs Ther. 35:901–913. 2021.PubMed/NCBI View Article : Google Scholar | |
Lv R, Liu X, Zhang Y, Dong N, Wang X, He Y, Yue H and Yin Q: Pathophysiological mechanisms and therapeutic approaches in obstructive sleep apnea syndrome. Signal Transduct Target Ther. 8(218)2023.PubMed/NCBI View Article : Google Scholar | |
Popadic V, Brajkovic M, Klasnja S, Milic N, Rajovic N, Lisulov DP, Divac A, Ivankovic T, Manojlovic A, Nikolic N, et al: Correlation of dyslipidemia and inflammation with obstructive sleep apnea severity. Front Pharmacol. 13(897279)2022.PubMed/NCBI View Article : Google Scholar | |
Sun X, Wang C, He Y, Chen K and Miao Y: Effect of inflammatory cytokines and plasma metabolome on OSA: A bidirectional two-sample Mendelian randomization study and mediation analysis. Front Immunol. 15(1416870)2024.PubMed/NCBI View Article : Google Scholar | |
Andrade RGS, Viana FM, Nascimento JA, Drager LF, Moffa A, Brunoni AR, Genta PR and Lorenzi-Filho G: Nasal vs Oronasal CPAP for OSA Treatment: A Meta-Analysis. Chest. 153:665–674. 2018.PubMed/NCBI View Article : Google Scholar | |
Shen C, Ou Y, Ouyang R and Zong D: Prevalence and characteristics of pain in moderate-to-severe obstructive sleep apnea patients and effect of CPAP treatment. Sci Rep. 13(15758)2023.PubMed/NCBI View Article : Google Scholar | |
Lorenzi-Filho G, Almeida FR and Strollo PJ: Treating OSA: Current and emerging therapies beyond CPAP. Respirology. 22:1500–1507. 2017.PubMed/NCBI View Article : Google Scholar | |
Chang JL, Goldberg AN, Alt JA, Mohammed A, Ashbrook L, Auckley D, Ayappa I, Bakhtiar H, Barrera JE, Bartley BL, et al: International Consensus Statement on Obstructive Sleep Apnea. Int Forum Allergy Rhinol. 13:1061–1482. 2023.PubMed/NCBI View Article : Google Scholar | |
MacKay S, Carney AS, Catcheside PG, Chai-Coetzer CL, Chia M, Cistulli PA, Hodge JC, Jones A, Kaambwa B, Lewis R, et al: Effect of multilevel upper airway surgery vs medical management on the apnea-hypopnea index and patient-reported daytime sleepiness among patients with moderate or severe obstructive sleep apnea: The SAMS Randomized Clinical Trial. JAMA. 324:1168–1179. 2020.PubMed/NCBI View Article : Google Scholar | |
Sánchez AI, Martínez P, Miró E, Bardwell WA and Buela-Casal G: CPAP and behavioral therapies in patients with obstructive sleep apnea: Effects on daytime sleepiness, mood, and cognitive function. Sleep Med Rev. 13:223–233. 2009.PubMed/NCBI View Article : Google Scholar | |
Schmickl CN, Landry SA, Orr JE, Chin K, Murase K, Verbraecken J, Javaheri S, Edwards BA, Owens RL and Malhotra A: Acetazolamide for OSA and Central Sleep Apnea: A Comprehensive Systematic Review and Meta-Analysis. Chest. 158:2632–2645. 2020.PubMed/NCBI View Article : Google Scholar | |
Ulrich S, Nussbaumer-Ochsner Y, Vasic I, Hasler E, Latshang TD, Kohler M, Muehlemann T, Wolf M and Bloch KE: Cerebral oxygenation in patients with OSA: effects of hypoxia at altitude and impact of acetazolamide. Chest. 146:299–308. 2014.PubMed/NCBI View Article : Google Scholar | |
Chapman JL, Vakulin A, Hedner J, Yee BJ and Marshall NS: Modafinil/armodafinil in obstructive sleep apnoea: A systematic review and meta-analysis. Eur Respir J. 47:1420–1428. 2016.PubMed/NCBI View Article : Google Scholar | |
Mashaqi S, Patel SI, Combs D, Estep L, Helmick S, Machamer J and Parthasarathy S: The hypoglossal nerve stimulation as a novel therapy for treating obstructive sleep apnea-a literature review. Int J Environ Res Public Health. 18(1642)2021.PubMed/NCBI View Article : Google Scholar | |
Akinnusi ME, Laporta R and El-Solh AA: Lectin-like oxidized low-density lipoprotein receptor-1 modulates endothelial apoptosis in obstructive sleep apnea. Chest. 140:1503–1510. 2011.PubMed/NCBI View Article : Google Scholar | |
Feres MC, Fonseca FA, Cintra FD, Mello-Fujita L, de Souza AL, De Martino MC, Tufik S and Poyares D: An assessment of oxidized LDL in the lipid profiles of patients with obstructive sleep apnea and its association with both hypertension and dyslipidemia, and the impact of treatment with CPAP. Atherosclerosis. 241:342–349. 2015.PubMed/NCBI View Article : Google Scholar | |
Trevethick MA, Mantell SJ, Stuart EF, Barnard A, Wright KN and Yeadon M: Treating lung inflammation with agonists of the adenosine A2A receptor: Promises, problems and potential solutions. Br J Pharmacol. 155:463–474. 2008.PubMed/NCBI View Article : Google Scholar | |
Guo D, Mulder-Krieger T, IJzerman AP and Heitman LH: Functional efficacy of adenosine A2A receptor agonists is positively correlated to their receptor residence time. Br J Pharmacol. 166:1846–1859. 2012.PubMed/NCBI View Article : Google Scholar | |
de Lera Ruiz M, Lim YH and Zheng J: Adenosine A2A receptor as a drug discovery target. J Med Chem. 57:3623–3650. 2014.PubMed/NCBI View Article : Google Scholar | |
Al-Attraqchi OHA, Attimarad M, Venugopala KN, Nair A and Al-Attraqchi NHA: Adenosine A2A receptor as a potential drug target-current status and future perspectives. Curr Pharm Des. 25:2716–2740. 2019.PubMed/NCBI View Article : Google Scholar | |
Kohler M and Stradling JR: Mechanisms of vascular damage in obstructive sleep apnea. Nat Rev Cardiol. 7:677–685. 2010.PubMed/NCBI View Article : Google Scholar | |
Gambino F, Zammuto MM, Virzì A, Conti G and Bonsignore MR: Treatment options in obstructive sleep apnea. Intern Emerg Med. 17:971–978. 2022.PubMed/NCBI View Article : Google Scholar |