Skip to main content
Erschienen in: Respiratory Research 1/2018

Open Access 01.12.2018 | Research

Intermittent hypoxia promotes melanoma lung metastasis via oxidative stress and inflammation responses in a mouse model of obstructive sleep apnea

verfasst von: Lian Li, Fangyuan Ren, Chao Qi, Leiqian Xu, Yinshan Fang, Maoli Liang, Jing Feng, Baoyuan Chen, Wen Ning, Jie Cao

Erschienen in: Respiratory Research | Ausgabe 1/2018

Abstract

Background

Recently, increased tumor incidence and cancer-related mortality have been reported among patients with obstructive sleep apnea (OSA). Intermittent hypoxia (IH), the hallmark feature of OSA, contributes to the metastasis of tumors. However, the molecular mechanisms by which tumor metastasis is accelerated by OSA-like IH remain to be elucidated.

Methods

C57BL/6 J male mice were subjected to intravenous injection of B16F10 melanoma cells before receiving IH treatment. Then, the animals were randomly distributed into three groups (n = 8 each): normoxia (N) group, IH group, and antioxidant tempol group (IHT, exposed to IH after treatment with tempol). After the mice were sacrificed, the number and weight of lung metastatic colonies were assessed. The lung tissues with tumor metastasis were analyzed for markers of oxidative stress and inflammation and for HIF-1α using western blotting and real-time PCR (qRT-PCR). The level of reactive oxygen species (ROS) in B16F10 cell was also assessed after N, IH and IH with tempol treatments.

Results

Compared with normoxia, IH significantly increased the number and weight of mouse lung metastatic colonies. Treatment of B16F10 cells with IH significantly enhanced ROS generation. Lung tissues with tumor metastasis provided evidence of increased oxidative stress, as assessed by p22phox and SOD mRNA levels and the NRF2 protein level, as well as increased inflammation, as assessed by TNF-α and IL-6 mRNA levels and the NF-κB P65 protein level. HIF-1α protein levels were increased in response to IH treatment. Tempol, an important antioxidant, ameliorated IH-induced melanoma lung metastasis in mice and reduced oxidative stress and inflammation responses.

Conclusions

These results support the hypothesis that oxidative stress and inflammation responses play an important role in the pathogenesis of OSA-like IH-induced melanoma lung metastasis in mice. Antioxidant intervention provides a novel strategy for the prevention and treatment of cancer in OSA populations.

Background

Obstructive sleep apnea (OSA), a common sleep breathing disorder, is characterized by recurrent upper airway obstruction triggered by complete or partial upper airway collapse during sleep. It can lead to intermittent hypoxia (IH), sleep fragmentation and daytime sleepiness. OSA affects at least 4–10% of adults and has been reported as a critical predisposing factor for cardiovascular morbidity [14], metabolic disorder [5, 6], hypertension and cognitive dysfunction [7, 8]. Currently, several epidemiological studies have demonstrated that cancer progression and cancer-related mortality are accelerated in patients with OSA [914]. Therefore, IH is considered to be a major characteristic factor of OSA for promoting tumor invasion and metastasis [15, 16]. Animal studies also indicate that OSA-like IH enhances the growth, invasion and metastasis of tumors [1720]. However, the underlying mechanisms for this OSA-like IH-induced tumor metastasis are not completely understood.
Hypoxia is increasingly recognized as an important feature of the intra-tumoral microenvironment, and it can induce the growth and metastasis of tumors via the up-regulation of hypoxia-inducible factor-1α (HIF-1α) [21]. However, in contrast to continuous hypoxia, IH in patients with OSA is a unique physiological state with a phase of post-hypoxic re-oxygenation (ROX). It is characterized by higher frequency, more serious hypoxia and larger variation in blood oxygen saturation. The relationship between OSA-like IH and tumors has been explored in recent years. The hypothesis that OSA-like IH results in the development and progression of tumors has been proposed in several investigations [17, 2225]. OSA-like IH includes a phase of post-hypoxic ROX, which results in the production of reactive oxygen species (ROS) and increases oxidative stress and inflammation responses. These represent important pathological links between OSA and injuries of multiple organs including the myocardium [26, 27], carotid body [28, 29], adrenal gland [30] and brain [31, 32]. Recently, Gutsche et al. showed that IH increases pro-metastatic gene expression by activating NF-κB in inflammatory breast cancer cells [25]. Here, we hypothesized that oxidative stress and inflammation responses may play important roles in a mouse model of OSA-like IH-induced tumor metastasis.
4-Hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (tempol), a well-known antioxidant, promotes the clearance of ROS and protects mitochondria from oxidative damage. It has been reported that tempol can reduce the atherosclerosis associated with metabolic syndrome by reducing vascular inflammation and repressing NADPH-2 oxidase expression [33]. It also exerts neuroprotective effects by reducing superoxide anions and preventing peroxynitrite-associated inflammation [3436]. Recently, tempol has been confirmed to attenuate apoptotic signals in endothelial cells from IH-exposed rats by decreasing oxidative stress and inflammation injury [37]. Therefore, it is reasonable to hypothesize that tempol may attenuate IH-induced tumor metastasis by decreasing oxidative stress and inflammation responses. This study was designed to investigate the enhancement of OSA-like IH-induced melanoma lung metastasis by oxidative stress and inflammation responses, and to assess the interventional role of the antioxidant tempol.

Methods

Animals

C57BL/6 J male mice with the age of 8 weeks were purchased from Model Animal Research Center of Nanjing University. All mice were housed and cared for in standard cages and provided access to food and water freely at the General Hospital of Tianjin Medical University. The animals, after being injected with melanoma cells, were randomly distributed into three groups (n = 8): N group, IH group and IHT group, which was treated with tempol (Sigma-Aldrich, St. Louis, MO, USA). All animal experimental protocols were approved by the Animal Care and Use Committee at General Hospital of Tianjin Medical University and were performed in accordance with the guidelines outlined by the committee.

Melanoma cell culture

Murine melanoma B16F10 cells were obtained from the Laboratory of Lung Development and Diseases at Nankai University (Tianjin, China) with the American Type Culture Collection (ATCC; Manassas, VA, USA) as the original source. These cells were cultured in DMEM supplemented with 10% fetal bovine serum (Gibco, Carlsbad, CA, USA) and antibiotics in a 37 °C incubator with a humidified atmosphere containing 5% CO2.

Establishment of the induced melanoma lung metastasis model

Briefly, the mice were anaesthetized with chloral hydrate (Sangon Biotech, Shanghai, China) and then intravenously injected with 1 × 105 B16F10 cells diluted in 100 μL of PBS via tail vein. The number and weight of metastatic colonies per lung were assessed after 3 weeks.

Animal exposure to IH and tempol administration

The animal model of IH was established according to a previously described protocol [38]. The mice were exposed to IH for 6 h/day during the light period (10 AM–4 PM) for 21 consecutive days in a specialized plexiglas chamber (23 cm × 20 cm × 12 cm = 5520 cm3 ≈ 5.5 L) with 8 mice per cage. The flow of nitrogen (99.99% N2, hypoxia phase) or compressed air (air, ROX phase) was modulated by a gas control delivery system in the chamber to maintain a nadir oxygen concentration of 5% and an IH cycle. The alternating cycle lasted for 2 min, and consisted of hypoxia and ROX phases. Gas flow and O2 concentration were monitored continuously with an O2 analyzer (CY-12C, Meicheng, China). During the hypoxia phase, the O2 concentration in the chamber was decreased to 5% within 20 s by infusion of N2 and remained at that concentration for 30 s. During the ROX phase, the O2 concentration was rapidly increased to 21% within 10 s by flushing the chamber with compressed clean air, which was then sustained for 60 s. In general, 180 cycles were completed during the exposure period (10 AM–4 PM) every day. Mice exposed to the N condition were used as the control group. Mice in the IHT group received an intraperitoneal injection of 1 ml of 10% (w/v) tempol per kilogram body weight prior to IH exposure every day, throughout the whole exposure period of 3 weeks.

Metastasis assessment

Five lung lobes were carefully separated from the mice in the IH-induced melanoma metastasis model. The number of metastatic colonies per lung was counted after anterior and posterior image acquisition. Metastatic colonies were then separated from the lung lobes, and the weight of the metastases per lung was measured.

RNA isolation and qRT-PCR

Samples of mouse lung tumor tissue from the three groups (n = 8) were homogenized in Trizol. RNA isolation and qRT-PCR analysis were performed as previously described [39]. The expression of p22phox, SOD, TNF-α and IL-6 was determined using a SYBR Green Master Mix kit (Roche Diagnostics, Indianapolis, IN, USA). Mouse β-actin was used as an internal control. Gene expression was quantified relative to the mRNA levels from mouse lung tumor tissue, which was amplified in parallel. The sequences of specific primer pairs are listed in Table 1.
Table 1
Real-Time PCR primers used in this study
Genes
Forward primer 5′-3′
Reverse primer 5′-3′
p22 phox
AAGTACCTGACCGCTGTGG
AGGTAGATCACACTGGCAATG
SOD
TTATGATGGGCACTGCAAAGAA
ACTGCCTTTAGAGAACCAGCC
TNF-α
CCAAACCAGCCTGACAACTT
TCTAGCATGCTCCACCACTG
IL-6
GTGTAGCACAACTTCCAATTACGAA
GGAATTTCCGCCTCGAGTCT
β-actin
AGGCCAACCGTGAAAAGATG
AGAGCATAGCCCTCGTAGATGG

Western blot analysis

Proteins were extracted from mouse lung tumor tissue according to standard protocols as described previously [40]. Protein concentrations were measured using a BCA assay kit (Pierce, Rockford, IL, USA). The proteins were separated by SDS-PAGE and electroblotted onto PVDF membranes (Millipore, Bedford, MA, USA). These membranes were blocked with 5% skim milk for 1 h at room temperature and then incubated with primary antibodies at 4 °C overnight. The primary antibodies, including HIF-1α (1:200; Novus Biologicals, Littleton, CO, USA), NF-E2-related factor 2 (NRF2) (1:200; ProteinTech Group, Chicago, IL, USA), NF-κB P65 (1:5000; Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) and β-actin (1:5000; Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA), were used to probe the target proteins. Goat anti-mouse IgG-HRP or goat anti-rabbit IgG-HRP (1:5000; Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) were used as secondary antibodies and were incubated with the membranes for 2 h at room temperature. Bands were visualized with ECL reagents. The intensity of protein bands on the scanned images was analyzed with a Tanon Gis digital image analytical system. The accumulation of HIF-1α, NRF2 and NF-κB P65 was quantified as the ratio of the band intensities for the target protein and β-actin.

ROS detection in vitro

B16F10 cells were seeded in 60 mm culture dishes and incubated for 12 h with DMEM supplemented with 10% fetal bovine serum. After two washes with PBS, the culture medium was replaced with 2 ml DMEM supplemented with 2% fetal bovine serum. The cells were treated with N, IH (30 s 5% O2–90 s 21% O2) and IH with tempol for 8 h/d, for 2 d. ROS generation was determined using a Reactive Oxygen Species Assay Kit (Beyotime, China). The cells were washed twice with pre-warmed PBS and then incubated with DCFH-DA (10 mM) at 37 °C for 30 min. After two washes with PBS, the cells were collected with 1 ml lysis buffer. Then, the clear supernatant was transferred to a 96-well plate after centrifugation at 12000 rpm for 10 min. Relative fluorescence was read with a BioTek Cytation 5 System (BioTek, Winooski, VT) set at 488 nm excitation and 525 nm emission. All assays were repeated in triplicate.

Statistical analysis

SPSS version 16.0 (SPSS Inc., Chicago, IL) software package was used for statistical analysis. All data are presented as Mean ± SEM. Comparisons between several groups were analyzed using one-way ANOVA. Difference between experimental and control group was analyzed using Student’s t test. The statistically significant difference was considered at P < 0.05.

Results

OSA-like IH promotes melanoma lung metastasis

The number of lung metastatic melanomas was determined at day 21 after the exposure to N, IH and IHT conditions. The number of metastatic melanomas per lung in the IH group (12.8 ± 1.5) was significantly higher than that in the N group (4.8 ± 0.7) (Fig. 1a, b) (P < 0.01). However, the number of metastatic melanomas per lung in the IHT group (5.1 ± 0.8) was lower than that in the IH group (Fig. 1a, b) (P < 0.01).
In addition, compared to that of the N group, the weight of lung metastases in the IH group was significantly increased (Fig. 1c) (P < 0.05). The weight of lung metastases in the IHT group was decreased when compared with that of the IH group (Fig. 1c) (P < 0.05). These findings suggested that OSA-like IH increased melanoma metastasis in the lung, and this enhancement effect was prevented by pretreatment with the antioxidant tempol (Fig. 1a, b, c).

Increased ROS generation in murine melanoma cells after exposure to OSA-like IH

OSA-like IH can be interrupted by posthypoxic reoxygenation, which results in the generation of O2 free radicals and oxidative stress. In current study, we first investigated the effect of OSA-like IH exposure on ROS levels in murine melanoma B16F10 cells. As shown in Fig. 2, we found that compared with normoxia, OSA-like IH markedly increased ROS levels (Fig. 2) (P < 0.05). However, the antioxidant tempol effectively diminishes this OSA-like IH-induced ROS production (Fig. 2).

Activated oxidative stress response in melanoma lung metastasis mouse model after exposure to OSA-like IH

ROS generation can activate oxidative stress and inflammation responses. Accordingly, we investigated the roles of oxidative stress and inflammation responses in the mouse model of OSA-like IH-induced tumor metastasis. To assess the activation of the oxidative stress response in the IH-induced lung metastasis mouse model, the mRNA expression levels of SOD and p22phox were evaluated by qRT-PCR. The transcription factor NF-E2-related factor 2 (NRF2), which is related to oxidative stress, was evaluated by western blotting. The mRNA expression level of SOD in the IH group was significantly decreased compared with that in the N group (Fig. 3A) (P < 0.05). The mRNA expression level of p22phox in the IH group was also significantly increased compared with that in the N group (Fig. 3B) (P <  0.05). The protein expression level of NRF2 was significantly increased in the IH group when compared with that in the N group (Fig. 3C a, b) (P < 0.01). These results indicated that OSA-like IH enhanced oxidative stress in the melanoma lung metastasis mouse model. As expected, oxidative stress in the IHT group was significantly reduced compared with that in the IH group (Fig. 3A, B, C).

Activated inflammation response in melanoma lung metastasis mouse model after exposure to OSA-like IH

Next, to examine the activation of the inflammation response in the OSA-like IH-induced lung metastasis mouse model, the mRNA expression levels of TNF-α and IL-6, as well as the protein level of the inflammation response transcription factor NF-κB P65, were evaluated by qRT-PCR and western blotting, respectively. The mRNA expression levels of TNF-α and IL-6 in IH group were significantly increased when compared with those in the N group (Fig. 4A, B) (P < 0.05). The protein expression level of NF-κB P65 in the IH group was significantly increased when compared with that in the N group (Fig. 4C a, b) (P < 0.05). These results suggested that OSA-like IH activated the inflammation response in this melanoma lung metastasis mouse model. Similarly, inflammation in the IHT group was significantly attenuated when compared with that in the IH group (Fig. 4A, B, C).

Increased HIF-1α expression in melanoma lung metastasis mouse model after exposure to OSA-like IH

HIF-1α is an important transcription factor for responding to hypoxia, and plays an important role in tumor metastasis. The protein expression level of HIF-1α in the IH group was significantly higher than that in the N group (P <  0.05). However, the protein expression level of HIF-1α in the IHT group was significantly lower than that in the IH group (Fig. 5a, b) (P <  0.05), suggesting that tempol treatment attenuates the OSA-like IH-induced expression of HIF-1α in this melanoma lung metastasis mouse model.

Discussion

Epidemiological studies have demonstrated that OSA can promote the development and mortality of cancers. Recently, the relationship between OSA-like IH and tumors has only been explored in animals. In the present study, OSA-like IH exposure increased melanoma metastasis in the lung, which is remarkably similar to previous reports [18], consistently with the observation that patients with OSA have a close relationship with high cancer incidence and cancer-related mortality [1014].
OSA-like IH may be interrupted by ROX. In the ROX phase, excessive ROS, primarily derived from activated NADPH oxidase, can lead to oxidant/antioxidant imbalance, and cause radical-induced oxidation and damage. Many studies have confirmed that oxidative stress is associated with the occurrence and progression of tumors [41]. Herein, we have reported that OSA-like IH enhanced ROS generation in B16F10 cells. The mRNA expression level of the enzymatic antioxidant SOD was significantly lower in the IH group than in the N group. In addition, the mRNA expression level of NADPH oxidase p22phox in the IH group was significantly increased when compared with that in the N group. NRF2, which can be activated by IH, is a key transcription factor for responding to oxidative stress. In the OSA-like IH-induced lung metastasis mouse model, the protein expression level of NRF2 in the IH group was significantly elevated compared with that in the N group. These results suggested that oxidative stress might be the mechanism underlying the melanoma lung metastasis induced by OSA-like IH.
Oxidative stress promotes inflammation responses, and in turn, inflammation enhances oxidative stress. NF-κB is another transcription factor activated by ROS, and it acts as a master regulator of the transcriptional responses to inflammation [42, 43]. Up-regulation of inflammatory molecules or cytokines is associated with tumor proliferation, invasiveness and metastasis [44, 45]. Gutsche et al. showed that in inflammatory breast cancer cells, IH induced the expression of multiple pro-metastatic genes via NF-κB [25]. In our study, we also verified that OSA-like IH significantly increased the protein level of NF-κB in a melanoma lung metastasis mouse model. The mRNA levels of inflammatory markers, including TNF-α and IL-6, were significantly induced by OSA-like IH exposure. Inflammatory responses activated by oxidative stress may be a potential mechanism underlying the melanoma lung metastasis induced by OSA-like IH.
Excessive ROS can be eliminated by exogenous antioxidants in several animal models [46, 47]. Tempol, a redox-cycling nitroxyl radical, can scavenge excessive ROS and increase SOD activity, and is thus considered to be a promising antioxidant for clinical and experimental application. It exerts neuroprotective effects by reducing superoxide anions and attenuating peroxynitrite-associated inflammation. Previous studies have demonstrated that tempol plays a protective role in cardiovascular disease, neurological disorders and cancer by reducing oxidative stress or inflammation. In our OSA-like IH-induced lung metastasis mouse model, tempol repressed the IH-induced oxidative stress and inflammation responses, as well as melanoma lung metastasis. In the future, antioxidant intervention may be an effective strategy for treating OSA-related cancer patients.
HIF-1α is an important transcription factor for the adaptive response to hypoxia. HIF-1α protein accumulates under hypoxic conditions, and is degraded after each reoxygenation [48]. In our study, the protein level of HIF-1α in the IH group was significantly higher than that in the N group, which was consistent with previous studies [24, 49, 50]. IH can induce ROS generation and cause oxidative stress responses. Tempol, known as an antioxidant, promotes the clearance of ROS and protects mitochondria from oxidative damage. Previous reports have shown that tempol or other antioxidants can prevent HIF-1α-mediated cancer progression [37, 5153]. Consistent with these reports, our findings suggest that antioxidant tempol attenuated melanoma lung metastasis and oxidative stress, as well as decreased HIF-1α expression. Therefore, our study indicates that increased ROS production and oxidative stress induced by OSA-like IH promote melanoma lung metastasis, which is in part regulated through up-regulation of HIF-1α expression. However, other studies have demonstrated that the stabilization of HIF-1α under hypoxia or IH is independent of oxidative stress [25, 54, 55]. These conflicting reports indicate that further study is necessary to confirm the relationship between HIF-1α and oxidative stress.

Conclusions

These results suggest that the oxidative stress and inflammation responses induced by IH exposure play important roles in the pathogenesis of tumor metastasis, and the use of antioxidants such as tempol may potentially serve as a unique strategy for OSA-related cancer patients.

Acknowledgements

None.

Funding

This study was supported by the grants from the National Natural Science Foundation of China (No. 81500070 and 81670084), and grants from Tianjin Medical University General Hospital for Young Scholars (ZYYFY2014011).

Availability of data and materials

All data generated and analyzed during the study are included in the published article and can be shared upon request.

Ethics approval

All animal experimental protocols were approved by the Animal Care and Use Committee at General Hospital of Tianjin Medical University and in accordance with the guidelines outlined by the committee.
Not applicable

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Bassetti C, Aldrich MS. Sleep apnea in acute cerebrovascular diseases: final report on 128 patients. Sleep. 1999;22:217–23.CrossRefPubMed Bassetti C, Aldrich MS. Sleep apnea in acute cerebrovascular diseases: final report on 128 patients. Sleep. 1999;22:217–23.CrossRefPubMed
2.
Zurück zum Zitat Drummond MJ, Glynn EL, Fry CS, Dhanani S, Volpi E, Rasmussen BB. Essential amino acids increase microRNA-499, −208b, and -23a and downregulate myostatin and myocyte enhancer factor 2C mRNA expression in human skeletal muscle. J Nutr. 2009;139:2279–84.CrossRefPubMedPubMedCentral Drummond MJ, Glynn EL, Fry CS, Dhanani S, Volpi E, Rasmussen BB. Essential amino acids increase microRNA-499, −208b, and -23a and downregulate myostatin and myocyte enhancer factor 2C mRNA expression in human skeletal muscle. J Nutr. 2009;139:2279–84.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Somers VK, White DP, Amin R, Abraham WT, Costa F, Culebras A, Daniels S, Floras JS, Hunt CE, Olson LJ, et al. Sleep apnea and cardiovascular disease: an American Heart Association/American College of Cardiology Foundation scientific statement from the American Heart Association Council for high blood pressure research professional education committee, council on clinical cardiology, stroke council, and council on cardiovascular nursing. J Am Coll Cardiol. 2008;52:686–717.CrossRefPubMed Somers VK, White DP, Amin R, Abraham WT, Costa F, Culebras A, Daniels S, Floras JS, Hunt CE, Olson LJ, et al. Sleep apnea and cardiovascular disease: an American Heart Association/American College of Cardiology Foundation scientific statement from the American Heart Association Council for high blood pressure research professional education committee, council on clinical cardiology, stroke council, and council on cardiovascular nursing. J Am Coll Cardiol. 2008;52:686–717.CrossRefPubMed
4.
Zurück zum Zitat Kokturk O, Ciftci TU, Mollarecep E, Ciftci B. Elevated C-reactive protein levels and increased cardiovascular risk in patients with obstructive sleep apnea syndrome. Int Heart J. 2005;46:801–9.CrossRefPubMed Kokturk O, Ciftci TU, Mollarecep E, Ciftci B. Elevated C-reactive protein levels and increased cardiovascular risk in patients with obstructive sleep apnea syndrome. Int Heart J. 2005;46:801–9.CrossRefPubMed
5.
Zurück zum Zitat Lurie A. Metabolic disorders associated with obstructive sleep apnea in adults. Adv Cardiol. 2011;46:67–138.CrossRefPubMed Lurie A. Metabolic disorders associated with obstructive sleep apnea in adults. Adv Cardiol. 2011;46:67–138.CrossRefPubMed
6.
Zurück zum Zitat Parish JM, Adam T, Facchiano L. Relationship of metabolic syndrome and obstructive sleep apnea. J Clin Sleep Med. 2007;3:467–72.PubMedPubMedCentral Parish JM, Adam T, Facchiano L. Relationship of metabolic syndrome and obstructive sleep apnea. J Clin Sleep Med. 2007;3:467–72.PubMedPubMedCentral
7.
Zurück zum Zitat Felmet KA, Petersen M. Obstructive sleep apnea and cognitive dysfunction. JAAPA. 2006;19:16–20.PubMed Felmet KA, Petersen M. Obstructive sleep apnea and cognitive dysfunction. JAAPA. 2006;19:16–20.PubMed
8.
Zurück zum Zitat Ferini-Strambi L, Baietto C, Di Gioia MR, Castaldi P, Castronovo C, Zucconi M, Cappa SF. Cognitive dysfunction in patients with obstructive sleep apnea (OSA): partial reversibility after continuous positive airway pressure (CPAP). Brain Res Bull. 2003;61:87–92.CrossRefPubMed Ferini-Strambi L, Baietto C, Di Gioia MR, Castaldi P, Castronovo C, Zucconi M, Cappa SF. Cognitive dysfunction in patients with obstructive sleep apnea (OSA): partial reversibility after continuous positive airway pressure (CPAP). Brain Res Bull. 2003;61:87–92.CrossRefPubMed
9.
Zurück zum Zitat Cao J, Feng J, Li L, Chen B. Obstructive sleep apnea promotes cancer development and progression: a concise review. Sleep Breath. 2015;19:453–7.CrossRefPubMed Cao J, Feng J, Li L, Chen B. Obstructive sleep apnea promotes cancer development and progression: a concise review. Sleep Breath. 2015;19:453–7.CrossRefPubMed
10.
Zurück zum Zitat Campos-Rodriguez F, Martinez-Garcia MA, Martinez M, Duran-Cantolla J, Pena Mde L, Masdeu MJ, Gonzalez M, Campo F, Gallego I, Marin JM, et al. Association between obstructive sleep apnea and cancer incidence in a large multicenter Spanish cohort. Am J Respir Crit Care Med. 2013;187:99–105.CrossRefPubMed Campos-Rodriguez F, Martinez-Garcia MA, Martinez M, Duran-Cantolla J, Pena Mde L, Masdeu MJ, Gonzalez M, Campo F, Gallego I, Marin JM, et al. Association between obstructive sleep apnea and cancer incidence in a large multicenter Spanish cohort. Am J Respir Crit Care Med. 2013;187:99–105.CrossRefPubMed
11.
Zurück zum Zitat Martinez-Garcia MA, Campos-Rodriguez F, Duran-Cantolla J, de la Pena M, Masdeu MJ, Gonzalez M, Del Campo F, Serra PC, Valero-Sanchez I, Ferrer MJ, et al. Obstructive sleep apnea is associated with cancer mortality in younger patients. Sleep Med. 2014;15:742–8.CrossRefPubMed Martinez-Garcia MA, Campos-Rodriguez F, Duran-Cantolla J, de la Pena M, Masdeu MJ, Gonzalez M, Del Campo F, Serra PC, Valero-Sanchez I, Ferrer MJ, et al. Obstructive sleep apnea is associated with cancer mortality in younger patients. Sleep Med. 2014;15:742–8.CrossRefPubMed
12.
Zurück zum Zitat Nieto FJ, Peppard PE, Young T, Finn L, Hla KM, Farre R. Sleep-disordered breathing and cancer mortality: results from the Wisconsin sleep cohort study. Am J Respir Crit Care Med. 2012;186:190–4.CrossRefPubMedPubMedCentral Nieto FJ, Peppard PE, Young T, Finn L, Hla KM, Farre R. Sleep-disordered breathing and cancer mortality: results from the Wisconsin sleep cohort study. Am J Respir Crit Care Med. 2012;186:190–4.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Zhang X, Giovannucci EL, Wu K, Gao X, Hu F, Ogino S, Schernhammer ES, Fuchs CS, Redline S, Willett WC, Ma J. Associations of self-reported sleep duration and snoring with colorectal cancer risk in men and women. Sleep. 2013;36:681–8.CrossRefPubMedPubMedCentral Zhang X, Giovannucci EL, Wu K, Gao X, Hu F, Ogino S, Schernhammer ES, Fuchs CS, Redline S, Willett WC, Ma J. Associations of self-reported sleep duration and snoring with colorectal cancer risk in men and women. Sleep. 2013;36:681–8.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Christensen AS, Clark A, Salo P, Nymann P, Lange P, Prescott E, Rod NH. Symptoms of sleep disordered breathing and risk of cancer: a prospective cohort study. Sleep. 2013;36:1429–35.CrossRefPubMedPubMedCentral Christensen AS, Clark A, Salo P, Nymann P, Lange P, Prescott E, Rod NH. Symptoms of sleep disordered breathing and risk of cancer: a prospective cohort study. Sleep. 2013;36:1429–35.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Rofstad EK, Gaustad JV, Egeland TA, Mathiesen B, Galappathi K. Tumors exposed to acute cyclic hypoxic stress show enhanced angiogenesis, perfusion and metastatic dissemination. Int J Cancer. 2010;127:1535–46.CrossRefPubMed Rofstad EK, Gaustad JV, Egeland TA, Mathiesen B, Galappathi K. Tumors exposed to acute cyclic hypoxic stress show enhanced angiogenesis, perfusion and metastatic dissemination. Int J Cancer. 2010;127:1535–46.CrossRefPubMed
16.
Zurück zum Zitat Lee SL, Rouhi P, Dahl Jensen L, Zhang D, Ji H, Hauptmann G, Ingham P, Cao Y. Hypoxia-induced pathological angiogenesis mediates tumor cell dissemination, invasion, and metastasis in a zebrafish tumor model. Proc Natl Acad Sci U S A. 2009;106:19485–90.CrossRefPubMedPubMedCentral Lee SL, Rouhi P, Dahl Jensen L, Zhang D, Ji H, Hauptmann G, Ingham P, Cao Y. Hypoxia-induced pathological angiogenesis mediates tumor cell dissemination, invasion, and metastasis in a zebrafish tumor model. Proc Natl Acad Sci U S A. 2009;106:19485–90.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Almendros I, Wang Y, Becker L, Lennon FE, Zheng J, Coats BR, Schoenfelt KS, Carreras A, Hakim F, Zhang SX, et al. Intermittent hypoxia-induced changes in tumor-associated macrophages and tumor malignancy in a mouse model of sleep apnea. Am J Respir Crit Care Med. 2014;189:593–601.CrossRefPubMedPubMedCentral Almendros I, Wang Y, Becker L, Lennon FE, Zheng J, Coats BR, Schoenfelt KS, Carreras A, Hakim F, Zhang SX, et al. Intermittent hypoxia-induced changes in tumor-associated macrophages and tumor malignancy in a mouse model of sleep apnea. Am J Respir Crit Care Med. 2014;189:593–601.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Almendros I, Montserrat JM, Torres M, Dalmases M, Cabanas ML, Campos-Rodriguez F, Navajas D, Farre R. Intermittent hypoxia increases melanoma metastasis to the lung in a mouse model of sleep apnea. Respir Physiol Neurobiol. 2013;186:303–7.CrossRefPubMed Almendros I, Montserrat JM, Torres M, Dalmases M, Cabanas ML, Campos-Rodriguez F, Navajas D, Farre R. Intermittent hypoxia increases melanoma metastasis to the lung in a mouse model of sleep apnea. Respir Physiol Neurobiol. 2013;186:303–7.CrossRefPubMed
19.
Zurück zum Zitat Almendros I, Montserrat JM, Torres M, Bonsignore MR, Chimenti L, Navajas D, Farre R. Obesity and intermittent hypoxia increase tumor growth in a mouse model of sleep apnea. Sleep Med. 2012;13:1254–60.CrossRefPubMed Almendros I, Montserrat JM, Torres M, Bonsignore MR, Chimenti L, Navajas D, Farre R. Obesity and intermittent hypoxia increase tumor growth in a mouse model of sleep apnea. Sleep Med. 2012;13:1254–60.CrossRefPubMed
20.
Zurück zum Zitat Almendros I, Montserrat JM, Ramirez J, Torres M, Duran-Cantolla J, Navajas D, Farre R. Intermittent hypoxia enhances cancer progression in a mouse model of sleep apnoea. Eur Respir J. 2012;39:215–7.CrossRefPubMed Almendros I, Montserrat JM, Ramirez J, Torres M, Duran-Cantolla J, Navajas D, Farre R. Intermittent hypoxia enhances cancer progression in a mouse model of sleep apnoea. Eur Respir J. 2012;39:215–7.CrossRefPubMed
21.
Zurück zum Zitat Pistollato F, Rampazzo E, Persano L, Abbadi S, Frasson C, Denaro L, D'Avella D, Panchision DM, Della Puppa A, Scienza R, Basso G. Interaction of hypoxia-inducible factor-1alpha and notch signaling regulates medulloblastoma precursor proliferation and fate. Stem Cells. 2010;28:1918–29.CrossRefPubMedPubMedCentral Pistollato F, Rampazzo E, Persano L, Abbadi S, Frasson C, Denaro L, D'Avella D, Panchision DM, Della Puppa A, Scienza R, Basso G. Interaction of hypoxia-inducible factor-1alpha and notch signaling regulates medulloblastoma precursor proliferation and fate. Stem Cells. 2010;28:1918–29.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Almendros I, Gileles-Hillel A, Khalyfa A, Wang Y, Zhang SX, Carreras A, Farre R, Gozal D. Adipose tissue macrophage polarization by intermittent hypoxia in a mouse model of OSA: effect of tumor microenvironment. Cancer Lett. 2015;361:233–9.CrossRefPubMed Almendros I, Gileles-Hillel A, Khalyfa A, Wang Y, Zhang SX, Carreras A, Farre R, Gozal D. Adipose tissue macrophage polarization by intermittent hypoxia in a mouse model of OSA: effect of tumor microenvironment. Cancer Lett. 2015;361:233–9.CrossRefPubMed
23.
Zurück zum Zitat Cortese R, Almendros I, Wang Y, Gozal D. Tumor circulating DNA profiling in xenografted mice exposed to intermittent hypoxia. Oncotarget. 2015;6:556–69.CrossRefPubMed Cortese R, Almendros I, Wang Y, Gozal D. Tumor circulating DNA profiling in xenografted mice exposed to intermittent hypoxia. Oncotarget. 2015;6:556–69.CrossRefPubMed
24.
Zurück zum Zitat Liu Y, Song X, Wang X, Wei L, Liu X, Yuan S, Lv L. Effect of chronic intermittent hypoxia on biological behavior and hypoxia-associated gene expression in lung cancer cells. J Cell Biochem. 2010;111:554–63.CrossRefPubMed Liu Y, Song X, Wang X, Wei L, Liu X, Yuan S, Lv L. Effect of chronic intermittent hypoxia on biological behavior and hypoxia-associated gene expression in lung cancer cells. J Cell Biochem. 2010;111:554–63.CrossRefPubMed
25.
Zurück zum Zitat Gutsche K, Randi EB, Blank V, Fink D, Wenger RH, Leo C, Scholz CC. Intermittent hypoxia confers pro-metastatic gene expression selectively through NF-kappaB in inflammatory breast cancer cells. Free Radic Biol Med. 2016;101:129–42.CrossRefPubMed Gutsche K, Randi EB, Blank V, Fink D, Wenger RH, Leo C, Scholz CC. Intermittent hypoxia confers pro-metastatic gene expression selectively through NF-kappaB in inflammatory breast cancer cells. Free Radic Biol Med. 2016;101:129–42.CrossRefPubMed
26.
Zurück zum Zitat Ding W, Zhang X, Huang H, Ding N, Zhang S, Hutchinson SZ, Zhang X. Adiponectin protects rat myocardium against chronic intermittent hypoxia-induced injury via inhibition of endoplasmic reticulum stress. PLoS One. 2014;9:e94545.CrossRefPubMedPubMedCentral Ding W, Zhang X, Huang H, Ding N, Zhang S, Hutchinson SZ, Zhang X. Adiponectin protects rat myocardium against chronic intermittent hypoxia-induced injury via inhibition of endoplasmic reticulum stress. PLoS One. 2014;9:e94545.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Almendros I, Farre R, Torres M, Bonsignore MR, Dalmases M, Ramirez J, Navajas D, Montserrat JM. Early and mid-term effects of obstructive apneas in myocardial injury and inflammation. Sleep Med. 2011;12:1037–40.CrossRefPubMed Almendros I, Farre R, Torres M, Bonsignore MR, Dalmases M, Ramirez J, Navajas D, Montserrat JM. Early and mid-term effects of obstructive apneas in myocardial injury and inflammation. Sleep Med. 2011;12:1037–40.CrossRefPubMed
28.
Zurück zum Zitat Del Rio R, Moya EA, Parga MJ, Madrid C, Iturriaga R. Carotid body inflammation and cardiorespiratory alterations in intermittent hypoxia. Eur Respir J. 2012;39:1492–500.CrossRefPubMed Del Rio R, Moya EA, Parga MJ, Madrid C, Iturriaga R. Carotid body inflammation and cardiorespiratory alterations in intermittent hypoxia. Eur Respir J. 2012;39:1492–500.CrossRefPubMed
29.
Zurück zum Zitat Lam SY, Liu Y, Ng KM, Lau CF, Liong EC, Tipoe GL, Fung ML. Chronic intermittent hypoxia induces local inflammation of the rat carotid body via functional upregulation of proinflammatory cytokine pathways. Histochem Cell Biol. 2012;137:303–17.CrossRefPubMed Lam SY, Liu Y, Ng KM, Lau CF, Liong EC, Tipoe GL, Fung ML. Chronic intermittent hypoxia induces local inflammation of the rat carotid body via functional upregulation of proinflammatory cytokine pathways. Histochem Cell Biol. 2012;137:303–17.CrossRefPubMed
30.
Zurück zum Zitat Kumar GK, Rai V, Sharma SD, Ramakrishnan DP, Peng YJ, Souvannakitti D, Prabhakar NR. Chronic intermittent hypoxia induces hypoxia-evoked catecholamine efflux in adult rat adrenal medulla via oxidative stress. J Physiol. 2006;575:229–39.CrossRefPubMedPubMedCentral Kumar GK, Rai V, Sharma SD, Ramakrishnan DP, Peng YJ, Souvannakitti D, Prabhakar NR. Chronic intermittent hypoxia induces hypoxia-evoked catecholamine efflux in adult rat adrenal medulla via oxidative stress. J Physiol. 2006;575:229–39.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Zhan G, Serrano F, Fenik P, Hsu R, Kong L, Pratico D, Klann E, Veasey SC. NADPH oxidase mediates hypersomnolence and brain oxidative injury in a murine model of sleep apnea. Am J Respir Crit Care Med. 2005;172:921–9.CrossRefPubMedPubMedCentral Zhan G, Serrano F, Fenik P, Hsu R, Kong L, Pratico D, Klann E, Veasey SC. NADPH oxidase mediates hypersomnolence and brain oxidative injury in a murine model of sleep apnea. Am J Respir Crit Care Med. 2005;172:921–9.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Xu W, Chi L, Row BW, Xu R, Ke Y, Xu B, Luo C, Kheirandish L, Gozal D, Liu R. Increased oxidative stress is associated with chronic intermittent hypoxia-mediated brain cortical neuronal cell apoptosis in a mouse model of sleep apnea. Neuroscience. 2004;126:313–23.CrossRefPubMed Xu W, Chi L, Row BW, Xu R, Ke Y, Xu B, Luo C, Kheirandish L, Gozal D, Liu R. Increased oxidative stress is associated with chronic intermittent hypoxia-mediated brain cortical neuronal cell apoptosis in a mouse model of sleep apnea. Neuroscience. 2004;126:313–23.CrossRefPubMed
33.
Zurück zum Zitat Cannizzo B, Quesada I, Militello R, Amaya C, Miatello R, Cruzado M, Castro C. Tempol attenuates atherosclerosis associated with metabolic syndrome via decreased vascular inflammation and NADPH-2 oxidase expression. Free Radic Res. 2014;48:526–33.CrossRefPubMed Cannizzo B, Quesada I, Militello R, Amaya C, Miatello R, Cruzado M, Castro C. Tempol attenuates atherosclerosis associated with metabolic syndrome via decreased vascular inflammation and NADPH-2 oxidase expression. Free Radic Res. 2014;48:526–33.CrossRefPubMed
34.
Zurück zum Zitat Hahn SM, Mitchell JB, Shacter E. Tempol inhibits neutrophil and hydrogen peroxide-mediated DNA damage. Free Radic Biol Med. 1997;23:879–84.CrossRefPubMed Hahn SM, Mitchell JB, Shacter E. Tempol inhibits neutrophil and hydrogen peroxide-mediated DNA damage. Free Radic Biol Med. 1997;23:879–84.CrossRefPubMed
35.
Zurück zum Zitat Lipman T, Tabakman R, Lazarovici P. Neuroprotective effects of the stable nitroxide compound Tempol on 1-methyl-4-phenylpyridinium ion-induced neurotoxicity in the nerve growth factor-differentiated model of pheochromocytoma PC12 cells. Eur J Pharmacol. 2006;549:50–7.CrossRefPubMed Lipman T, Tabakman R, Lazarovici P. Neuroprotective effects of the stable nitroxide compound Tempol on 1-methyl-4-phenylpyridinium ion-induced neurotoxicity in the nerve growth factor-differentiated model of pheochromocytoma PC12 cells. Eur J Pharmacol. 2006;549:50–7.CrossRefPubMed
36.
Zurück zum Zitat Rak R, Chao DL, Pluta RM, Mitchell JB, Oldfield EH, Watson JC. Neuroprotection by the stable nitroxide Tempol during reperfusion in a rat model of transient focal ischemia. J Neurosurg. 2000;92:646–51.CrossRefPubMed Rak R, Chao DL, Pluta RM, Mitchell JB, Oldfield EH, Watson JC. Neuroprotection by the stable nitroxide Tempol during reperfusion in a rat model of transient focal ischemia. J Neurosurg. 2000;92:646–51.CrossRefPubMed
37.
Zurück zum Zitat Guo H, Cao J, Li J, Yang X, Jiang J, Feng J, Li S, Zhang J, Chen B. Lymphocytes from intermittent hypoxia-exposed rats increase the apoptotic signals in endothelial cells via oxidative and inflammatory injury in vitro. Sleep Breath. 2015;19:969–76.CrossRefPubMed Guo H, Cao J, Li J, Yang X, Jiang J, Feng J, Li S, Zhang J, Chen B. Lymphocytes from intermittent hypoxia-exposed rats increase the apoptotic signals in endothelial cells via oxidative and inflammatory injury in vitro. Sleep Breath. 2015;19:969–76.CrossRefPubMed
38.
Zurück zum Zitat He Q, Yang QC, Zhou Q, Zhu H, Niu WY, Feng J, Wang Y, Cao J, Chen BY. Effects of varying degrees of intermittent hypoxia on proinflammatory cytokines and adipokines in rats and 3T3-L1 adipocytes. PLoS One. 2014;9:e86326.CrossRefPubMedPubMedCentral He Q, Yang QC, Zhou Q, Zhu H, Niu WY, Feng J, Wang Y, Cao J, Chen BY. Effects of varying degrees of intermittent hypoxia on proinflammatory cytokines and adipokines in rats and 3T3-L1 adipocytes. PLoS One. 2014;9:e86326.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Dong Y, Geng Y, Li L, Li X, Yan X, Fang Y, Li X, Dong S, Liu X, Li X, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary fibrosis in mice. J Exp Med. 2015;212:235–52.CrossRefPubMedPubMedCentral Dong Y, Geng Y, Li L, Li X, Yan X, Fang Y, Li X, Dong S, Liu X, Li X, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary fibrosis in mice. J Exp Med. 2015;212:235–52.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Geng Y, Dong Y, Yu M, Zhang L, Yan X, Sun J, Qiao L, Geng H, Nakajima M, Furuichi T, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP) 4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A. 2011;108:7058–63.CrossRefPubMedPubMedCentral Geng Y, Dong Y, Yu M, Zhang L, Yan X, Sun J, Qiao L, Geng H, Nakajima M, Furuichi T, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP) 4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A. 2011;108:7058–63.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49:1603–16.CrossRefPubMedPubMedCentral Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49:1603–16.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Scholz CC, Taylor CT. Hydroxylase-dependent regulation of the NF-kappaB pathway. Biol Chem. 2013;394:479–93.CrossRefPubMed Scholz CC, Taylor CT. Hydroxylase-dependent regulation of the NF-kappaB pathway. Biol Chem. 2013;394:479–93.CrossRefPubMed
43.
Zurück zum Zitat Muller-Edenborn K, Leger K, Glaus Garzon JF, Oertli C, Mirsaidi A, Richards PJ, Rehrauer H, Spielmann P, Hoogewijs D, Borsig L, et al. Hypoxia attenuates the proinflammatory response in colon cancer cells by regulating IkappaB. Oncotarget. 2015;6:20288–301.CrossRefPubMedPubMedCentral Muller-Edenborn K, Leger K, Glaus Garzon JF, Oertli C, Mirsaidi A, Richards PJ, Rehrauer H, Spielmann P, Hoogewijs D, Borsig L, et al. Hypoxia attenuates the proinflammatory response in colon cancer cells by regulating IkappaB. Oncotarget. 2015;6:20288–301.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Germano G, Allavena P, Mantovani A. Cytokines as a key component of cancer-related inflammation. Cytokine. 2008;43:374–9.CrossRefPubMed Germano G, Allavena P, Mantovani A. Cytokines as a key component of cancer-related inflammation. Cytokine. 2008;43:374–9.CrossRefPubMed
45.
Zurück zum Zitat Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44.CrossRefPubMed Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44.CrossRefPubMed
46.
Zurück zum Zitat Kita T, Nagano Y, Yokode M, Ishii K, Kume N, Ooshima A, Yoshida H, Kawai C. Probucol prevents the progression of atherosclerosis in Watanabe heritable hyperlipidemic rabbit, an animal model for familial hypercholesterolemia. Proc Natl Acad Sci U S A. 1987;84:5928–31.CrossRefPubMedPubMedCentral Kita T, Nagano Y, Yokode M, Ishii K, Kume N, Ooshima A, Yoshida H, Kawai C. Probucol prevents the progression of atherosclerosis in Watanabe heritable hyperlipidemic rabbit, an animal model for familial hypercholesterolemia. Proc Natl Acad Sci U S A. 1987;84:5928–31.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Williams RJ, Motteram JM, Sharp CH, Gallagher PJ. Dietary vitamin E and the attenuation of early lesion development in modified Watanabe rabbits. Atherosclerosis. 1992;94:153–9.CrossRefPubMed Williams RJ, Motteram JM, Sharp CH, Gallagher PJ. Dietary vitamin E and the attenuation of early lesion development in modified Watanabe rabbits. Atherosclerosis. 1992;94:153–9.CrossRefPubMed
48.
Zurück zum Zitat Toffoli S, Michiels C. Intermittent hypoxia is a key regulator of cancer cell and endothelial cell interplay in tumours. FEBS J. 2008;275:2991–3002.CrossRefPubMed Toffoli S, Michiels C. Intermittent hypoxia is a key regulator of cancer cell and endothelial cell interplay in tumours. FEBS J. 2008;275:2991–3002.CrossRefPubMed
49.
Zurück zum Zitat Louie E, Nik S, Chen JS, Schmidt M, Song B, Pacson C, Chen XF, Park S, Ju J, Chen EI. Identification of a stem-like cell population by exposing metastatic breast cancer cell lines to repetitive cycles of hypoxia and reoxygenation. Breast Cancer Res. 2010;12:R94.CrossRefPubMedPubMedCentral Louie E, Nik S, Chen JS, Schmidt M, Song B, Pacson C, Chen XF, Park S, Ju J, Chen EI. Identification of a stem-like cell population by exposing metastatic breast cancer cell lines to repetitive cycles of hypoxia and reoxygenation. Breast Cancer Res. 2010;12:R94.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Yuan G, Nanduri J, Khan S, Semenza GL, Prabhakar NR. Induction of HIF-1alpha expression by intermittent hypoxia: involvement of NADPH oxidase, Ca2+ signaling, prolyl hydroxylases, and mTOR. J Cell Physiol. 2008;217:674–85.CrossRefPubMedPubMedCentral Yuan G, Nanduri J, Khan S, Semenza GL, Prabhakar NR. Induction of HIF-1alpha expression by intermittent hypoxia: involvement of NADPH oxidase, Ca2+ signaling, prolyl hydroxylases, and mTOR. J Cell Physiol. 2008;217:674–85.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Ma J, Zhang Q, Chen S, Fang B, Yang Q, Chen C, Miele L, Sarkar FH, Xia J, Wang Z. Mitochondrial dysfunction promotes breast cancer cell migration and invasion through HIF1alpha accumulation via increased production of reactive oxygen species. PLoS One. 2013;8:e69485.CrossRefPubMedPubMedCentral Ma J, Zhang Q, Chen S, Fang B, Yang Q, Chen C, Miele L, Sarkar FH, Xia J, Wang Z. Mitochondrial dysfunction promotes breast cancer cell migration and invasion through HIF1alpha accumulation via increased production of reactive oxygen species. PLoS One. 2013;8:e69485.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Schroedl C, McClintock DS, Budinger GR, Chandel NS. Hypoxic but not anoxic stabilization of HIF-1alpha requires mitochondrial reactive oxygen species. Am J Physiol Lung Cell Mol Physiol. 2002;283:L922–31.CrossRefPubMed Schroedl C, McClintock DS, Budinger GR, Chandel NS. Hypoxic but not anoxic stabilization of HIF-1alpha requires mitochondrial reactive oxygen species. Am J Physiol Lung Cell Mol Physiol. 2002;283:L922–31.CrossRefPubMed
53.
Zurück zum Zitat Nazarewicz RR, Dikalova A, Bikineyeva A, Ivanov S, Kirilyuk IA, Grigor'ev IA, Dikalov SI. Does scavenging of mitochondrial superoxide attenuate cancer prosurvival signaling pathways? Antioxid Redox Signal. 2013;19:344–9.CrossRefPubMedPubMedCentral Nazarewicz RR, Dikalova A, Bikineyeva A, Ivanov S, Kirilyuk IA, Grigor'ev IA, Dikalov SI. Does scavenging of mitochondrial superoxide attenuate cancer prosurvival signaling pathways? Antioxid Redox Signal. 2013;19:344–9.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Chua YL, Dufour E, Dassa EP, Rustin P, Jacobs HT, Taylor CT, Hagen T. Stabilization of hypoxia-inducible factor-1alpha protein in hypoxia occurs independently of mitochondrial reactive oxygen species production. J Biol Chem. 2010;285:31277–84.CrossRefPubMedPubMedCentral Chua YL, Dufour E, Dassa EP, Rustin P, Jacobs HT, Taylor CT, Hagen T. Stabilization of hypoxia-inducible factor-1alpha protein in hypoxia occurs independently of mitochondrial reactive oxygen species production. J Biol Chem. 2010;285:31277–84.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Hagen T, Taylor CT, Lam F, Moncada S. Redistribution of intracellular oxygen in hypoxia by nitric oxide: effect on HIF1alpha. Science. 2003;302:1975–8.CrossRefPubMed Hagen T, Taylor CT, Lam F, Moncada S. Redistribution of intracellular oxygen in hypoxia by nitric oxide: effect on HIF1alpha. Science. 2003;302:1975–8.CrossRefPubMed
Metadaten
Titel
Intermittent hypoxia promotes melanoma lung metastasis via oxidative stress and inflammation responses in a mouse model of obstructive sleep apnea
verfasst von
Lian Li
Fangyuan Ren
Chao Qi
Leiqian Xu
Yinshan Fang
Maoli Liang
Jing Feng
Baoyuan Chen
Wen Ning
Jie Cao
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2018
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-018-0727-x

Weitere Artikel der Ausgabe 1/2018

Respiratory Research 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.