Next Article in Journal
A Potential Probiotic Lactobacillus plantarum JBC5 Improves Longevity and Healthy Aging by Modulating Antioxidative, Innate Immunity and Serotonin-Signaling Pathways in Caenorhabditis elegans
Next Article in Special Issue
Effects of Ferroptosis on the Metabolome in Cardiac Cells: The Role of Glutaminolysis
Previous Article in Journal
Phenolic Content and Antioxidant Activity in Apples of the ‘Galaval’ Cultivar Grown on 17 Different Rootstocks
Previous Article in Special Issue
Jaboticaba (Myrciaria jaboticaba) Attenuates Ventricular Remodeling after Myocardial Infarction in Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidative Stress and Diseases Associated with High-Altitude Exposure

Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile
*
Author to whom correspondence should be addressed.
Submission received: 13 December 2021 / Revised: 11 January 2022 / Accepted: 13 January 2022 / Published: 28 January 2022
(This article belongs to the Special Issue Oxidative Stress in Cardiac Disease)

Abstract

:
Several diseases associated with high-altitude exposure affect unacclimated individuals. These diseases include acute mountain sickness (AMS), high-altitude cerebral edema (HACE), high-altitude pulmonary edema (HAPE), chronic mountain sickness (CMS), and, notably, high-altitude pulmonary hypertension (HAPH), which can eventually lead to right ventricle hypertrophy and heart failure. The development of these pathologies involves different molecules and molecular pathways that might be related to oxidative stress. Studies have shown that acute, intermittent, and chronic exposure to hypobaric hypoxia induce oxidative stress, causing alterations to molecular pathways and cellular components (lipids, proteins, and DNA). Therefore, the aim of this review is to discuss the oxidative molecules and pathways involved in the development of high-altitude diseases. In summary, all high-altitude pathologies are related to oxidative stress, as indicated by increases in the malondialdehyde (MDA) biomarker and decreases in superoxide dismutase (SOD) and glutathione peroxidase (GPx) antioxidant activity. In addition, in CMS, the levels of 8-iso-PGF2α and H2O2 are increased, and evidence strongly indicates an increase in Nox4 activity in HAPH. Therefore, antioxidant treatments seem to be a promising approach to mitigating high-altitude pathologies.

1. Introduction

In recent years, the number of people with acute, intermittent (shift work; days at high altitude and then rest days at sea level, for years), or permanent exposure to high altitudes has increased considerably, and it has been estimated that more than 81.6 million people live at altitudes ≥2500 m [1]. Exposure to high altitude is characterized as a hypobaric hypoxic condition due to the decrease in partial oxygen pressure, which reduces the bioavailability of oxygen in organisms [2]. Hypobaric hypoxia causes changes in the oxygen transport cascade from the air to the mitochondrial membrane [3], which trigger compensatory physiological mechanisms to help the individual tolerate and acclimatize to hypobaric hypoxia. However, in some cases, the individual is susceptible to diseases, of which acute mountain sickness (AMS) is the most common, followed by high-altitude pulmonary edema (HAPE), high-altitude cerebral edema (HACE) [4], high-altitude pulmonary hypertension (HAPH) [5,6], and chronic mountain sickness (CMS) [5,7,8].
Previous studies have shown that both acute and chronic exposure to hypobaric hypoxia increase the levels of oxidative stress biomarkers [9], highlighting that in certain tissues, such as the lung, oxidative stress is rapidly triggered under high-altitude exposure. For example, a study has shown that within 1.5 h of exposure to acute hypobaric hypoxia, the levels of reactive oxygen species (ROS) and the oxidative stress biomarker malondialdehyde (MDA), a product of lipid peroxidation, increase in the lungs of rats [10]. Notably, oxidative stress induced by hypobaric hypoxia causes structural alterations in lipids, proteins, and DNA, resulting in cellular damage [11]. Moreover, several studies have implicated oxidative stress in the development of pathologies induced by hypobaric hypoxia exposure, such as those mentioned above [8,10,12,13,14]. Therefore, the aim of this review is to discuss the oxidative molecules and pathways involved in the development of acute and chronic mountain sickness, pulmonary and cerebral high-altitude edema, and pulmonary hypertension, because this information is necessary to understand and potentially mitigate these pathologies, specifically by revealing pharmacological targets.

2. Oxidative Stress under Hypobaric Hypoxia

Reactive oxygen species are small and unstable molecules produced from oxygen (O2). These molecules can be generated through different pathways, such as the mitochondrial electron transport chain, uncoupled nitric oxide synthase (NOS), the NADPH oxidase complex, and xanthine oxidase [15]. The principal ROS molecules studied to date are superoxide (O2), hydroxyl radicals (•OH), peroxide radicals (ROO•), the alkoxy group (RO•), hydrogen peroxide (H2O2), hypochlorous acid (HOCl), ozone (O3), singlet oxygen (1O2), and peroxynitrite (ONOO) [16].
ROS have been related to several physiological processes involved in cellular regulation, such as cell differentiation, apoptosis, and growth, as well as in the immune system [17,18], emphasizing that ROS are now also recognized as important signaling mediators and second messengers that enhance the vasoreactivity of the pulmonary artery [19,20,21]. However, when the levels of ROS are increased through environmental and chemical stimuli and are not compensated by endogenous antioxidant systems, such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase/reductase (GSH-Px), peroxiredoxin/thioredoxin (PRX/Trx), or metalloproteases (MMPs), oxidative stress occurs [15,22]. Oxidative stress can alter several cellular structures (DNA, proteins, lipids, and organelles), leading to cell death in some cases [23].
Interestingly, oxidative stress induced by hypoxia exposure has been shown to cause endoplasmic reticulum dysfunction [24,25], which results in the accumulation of unfolded and/or misfolded proteins; this dysregulation contributes to the acceleration of several processes related to inflammatory pathways [26].
Several studies have shown a relationship between oxidative stress and hypobaric hypoxia exposure [8,10,14]. A recent study that exposed subjects to high altitude for 14 days showed an increase in capillary blood ROS production, which remained elevated from day 1 to day 14 [27]. In addition, oxidative biomarker analysis showed that the levels of protein carbonyls in the plasma increased from day 1 to day 7, those of urinary 8-hydroxy-2-deoxy guanosine (8-OH-dG) increased on the 2nd and 4th days, and the levels of urinary 8-isoprostane (8-iso-PGF2α) increased from the 1st day to the 4th day. In contrast, capillary antioxidant capacity decreased on the 1st, 2nd, and 4th days [27]. However, it is important to note that there are differences in oxidative and antioxidant responses among ethnic groups; for example, the Sherpa population in the Tibetan Plateau has a lower hemoglobin level and better oxygen saturation than lowlanders [28]. Additionally, a study by Horscroft et al. [29] showed that these individuals, compared to lowlanders, had a lower capacity for fatty acid oxidation and increased protection against oxidative stress in skeletal muscle. Moreover, a study by Julian et al. [30] found differences in antioxidant capacity (SOD and catalase) in Andean pregnancies compared to European pregnancies exposed to high-altitude conditions (3600–4100 m). Genetic differences in ethnic populations are intriguing and should be investigated in future studies.
Therefore, oxidative stress generated by increased ROS levels that is not compensated by antioxidant activity may contribute directly or indirectly to the development of pathologies associated with high-altitude exposure. Each of these high-altitude diseases will be separately discussed in the following sections.

3. Oxidative Stress and High-Altitude Diseases

3.1. Acute Mountain Sickness

Acute mountain sickness is the most common illness affecting unacclimatized people exposed to both acute [31] and intermittent [32] hypobaric hypoxia. The extent of this pathology mainly depends upon the individual’s speed of ascent and altitude [31]. The symptoms of AMS (headache, nausea/vomiting, fatigue and/or weakness, and dizziness/light headedness) can limit the activity and abilities of people experiencing this condition, especially during the first days of high-altitude exposure [33]. AMS can worsen, leading to HACE, which must be treated as a medical emergency [34]. However, HACE and AMS are currently considered different diseases even when patients present with both conditions at the same time [34].
Oxidative stress triggered by acute hypobaric hypoxia exposure damages several organs in the human body, especially the brain and heart [35]. Interestingly, a study demonstrated that AMS was correlated with an increase in the oxidative stress biomarker MDA, as measured in the breath [12]. Additionally, this study showed that oxidative stress can be a pathogenic factor for the development of AMS. This finding has been supported by several studies showing an increase in MDA concentration in several organs under hypobaric hypoxia conditions [12,14,36,37,38]. On the other hand, one study with subjects at 5100 m found no differences in the plasma MDA concentrations; however, subjects presenting with AMS showed increased lipid hydroperoxide levels and free-radical-induced vascular damage of the blood–brain barrier [39]. In addition, the use of lipid-soluble antioxidant vitamins was a potentially effective intervention that attenuated AMS [40]. Recently, a study with volunteers exposed to acute hypobaric hypoxia (4220 m; 36 h) found that all individuals presented with AMS after 8 h of exposure and showed increased MDA concentrations within 24 h of the start of the expedition [41].
However, in regard to antioxidant activity, a study by Tang et al. [38] (3500 m; 24 h) showed that 261 subjects had decreased SOD activity in plasma, suggesting that the decreased SOD activity was correlated with AMS severity. Additionally, a study by Irarrázaval et al. [41] showed glutathione peroxidase (GPx) activity decreased under hypobaric hypoxia conditions; this study emphasized the relationship between oxidative stress and AMS severity. Moreover, adults who had been born prematurely and were later exposed to hypobaric hypoxia (3840 m; 8 h) showed a decrease in antioxidant capacity and an increase in AMS severity [42].
In contrast, a proteomic analysis of plasma from subjects with AMS showed that the levels of oxidative-stress-protecting proteins, such as peroxiredoxin-6 (Prdx-6), GPx, and sulfhydryl oxidase 1 (QSOX1), were increased approximately twofold after 9 h of exposure to hypobaric hypoxia (4875 m), suggesting that the antioxidant activity of these proteins increases to regulate the high levels of ROS produced in response to hypobaric hypoxia [43]. These studies are summarized in Table 1. To clarify these contrasting results, more studies are necessary.
Regarding antioxidant treatment, a recent study with mice demonstrated that pretreatment with 5,6,7,8-tetrahydroxyflavone (5,6,7,8-THF), which has antioxidant properties, increased the activity levels of SOD, CAT, and GSH-Px and decreased the H2O2 and MDA contents in the brain and heart, indicating that the use of this flavone as an excellent antioxidant enhancer, lipid peroxidation inhibitor, and general anti-hypoxia agent for preventing AMS merits further exploration [35]. On the other hand, methazolamide, a lipophilic analog of acetazolamide with multiple advantages over acetazolamide that also functions as a carbonic anhydrase inhibitor to abolish AMS and HACE, has been considered an antioxidative stress drug [44] since a previous study showed that methazolamide can protect cultured neurocytes against H2O2-induced oxidative damage [45].
Additionally, because methazolamide can protect cerebral cortical neurons against oxidative stress and ameliorate cerebral-edema-inducing cases of AMS and HACE, researchers have proposed using methazolamide for the prevention and treatment of pathologies associated with high-altitude exposure [46]. These studies have shown that oxidative stress may be an important pathogenic factor in AMS physiopathology under acute hypobaric hypoxia, and that antioxidant treatment could be an important strategy to mitigate the severity of high-altitude sickness symptoms.

3.2. High-Altitude Cerebral Edema

High-altitude cerebral edema can develop from the progression of AMS, although these diseases are currently considered separate conditions [34]. HACE is a neurological disorder [47] characterized by signs and symptoms such as headache, loss of coordination, weakness, and impairments in consciousness [48,49]. Without appropriate treatment (such as reducing patient altitude), the individual may experience coma followed by death from brain herniation within 24 h [50].
Previous studies have reported that free radicals can damage endothelial cells of the blood–brain barrier, causing extracellular edema [51], which may contribute to the pathogenesis of both AMS and HACE [39,52]. To examine the roles of free radicals in these diseases, researchers have used sea buckthorn seed oil, which has antioxidant properties, to pretreat rats before exposure to acute hypobaric hypoxia (9144 m; 5 h) [13]. The results showed that this antioxidant pretreatment protected the brains of rats against transvascular leakage (altered cell membrane permeability) upon exposure to hypoxia by attenuating the generation of free radicals and MDA in the brain and increasing the antioxidant levels of GSH, GPx, and SOD [13]. This finding was later corroborated by a study showing that rats exposed to hypobaric hypoxia conditions (7620 m; 48 h) presented HACE with increased ROS and MDA levels and reduced antioxidant levels (SOD and GPx) in brain tissue [48]. The aforementioned increase in oxidative stress might upregulate the redox-sensitive transcription factor NF-κB, as indicated by studies showing an increase in NF-κB levels in the brains of rats exposed to acute hypobaric hypoxia [48,53]. In addition, NF-κB may contribute to transvascular leakage in the rat brain by upregulating the expression of pro-inflammatory cytokines (IL-6, IL-1, and TNF-α) and cell adhesion molecules (ICAM-1, VCAM-1, E-selectin, and P-selectin) [48].
Notably, treatment with ganglioside GM1, a neuroprotective sphingolipid, decreased the severity of HACE in rats exposed to acute hypobaric hypoxia (7600 m; 24 h), and the researchers of this study suggested that this neuroprotective effect was likely due to decreased ROS and MDA levels and increased SOD and GSH levels in rat brain tissue. Moreover, GM1 treatment decreased the expression of pro-inflammatory cytokines (IL-1β, TNF-α, and IL-6) in serum and brain tissue. In addition, GM1 activated the phosphoinositide 3-kinase (PI3K)/Akt-Nrf2 pathway, which decreases both oxidative stress and inflammation [54] (Table 1). Taken together, these studies indicate that oxidative stress induced by acute hypobaric hypoxia can contribute to HACE and that antioxidants are a promising treatment to mitigate HACE and AMS induced by hypoxia. However, to validate the specific antioxidant treatment that is most beneficial for this particular high-altitude pathology, more studies are needed.

3.3. High-Altitude Pulmonary Edema

High-altitude pulmonary edema is a non-cardiogenic, acute, and potentially lethal pulmonary disorder [10]. Rapid ascents to altitudes higher than 2450 m and exercise can lead to HAPE in unacclimatized individuals [47,55]. This illness usually manifests 2 to 3 days after acute exposure to high altitude and is characterized by increased pulmonary arterial pressure, hypoxic pulmonary vasoconstriction with elevated vascular permeability in alveolar capillaries, and hypoxemia [56,57].
Similar to that of HACE, the pathogenesis of HAPE in rats exposed to acute hypobaric hypoxia (7620 m; 24 h) involves increased ROS levels, trending toward levels causing oxidative stress and contributing to an increase in transvascular leakage; the same study showed that the expression of NF-κB, pro-inflammatory cytokines (IL-1, IL-6, and TNFα), and adhesion molecules (VCAM-1, ICAM1, P-selectin, and E-selectin) is increased, indicating that increases in ROS (H2O2), MDA, and pro-inflammatory molecule levels drive positive regulatory feedback [10]. Additionally, a recent study on HAPE induced by acute hypobaric hypoxia exposure (3250 m; 7 days) suggested that mutations in mitochondrial DNA complex I subunits may cause mitochondrial respiratory chain dysfunction, leading to oxidative stress, as determined by an increase in lipid peroxidation and protein carbonylation [58].
In addition, increased oxidative stress in HAPE patients compared to that in acclimatized controls may have been due to decreased SOD activity [58], although Sarada et al. [10] showed that after 6 h of exposure, the level of antioxidant enzymes, such as SOD, was increased in HAPE patients. This difference may be attributable to the duration of exposure to acute hypobaric hypoxia and the models used in the two studies mentioned in the paragraph above. Importantly, the levels of other antioxidants, such as GSH, did not differ, and those of GPx decreased [10] (Table 1). Therefore, we emphasize the importance of oxidative stress and the subsequent inflammatory response in HAPE pathology.
To attenuate oxidative stress, Purushothaman et al. [59] used sea buckthorn leaf extract as a pretreatment for HAPE in rats exposed to hypobaric hypoxia (9144 m; 5 h); this extract attenuated the generation of free radicals and MDA and increased GSH, glutathione reductase (GR), and GPx levels in the lung. Moreover, pretreatment with sea buckthorn leaf extract attenuated transvascular leakage into the lungs and decreased the expression of pro-inflammatory markers (TNF-α, IL-6, IL-10, and MCP-1) in bronchoalveolar lavage fluid [59]. Another study evaluating Rhodiola crenulata extract as a HAPE pretreatment—due to its antioxidant and ROS-scavenging properties—in rats exposed to hypobaric hypoxia (8000 m; 9 h) showed that this extract suppressed HAPE symptoms, possibly by reducing oxidative stress through decreased lipid peroxidation, H2O2, myeloperoxidase-H2O2-chloride system, and HIF-1α expression in the lung [60]. Additionally, this pretreatment reduced hypoxia-induced increases in plasma endothelin-1 (ET-1) levels and vascular endothelial growth factor (VEGF) levels in bronchoalveolar lavage fluid; both of these markers have been related to vascular leakage, and this increase was probably caused by inhibition of the oxidative stress-HIF-1 pathway [60]. Notably, studies in this section and throughout this review referring to the relationship between ROS (e.g., H2O2) and high-altitude sickness further support the role of oxidative stress.

3.4. Chronic Mountain Sickness

Chronic mountain sickness is a medical condition characterized by increased hematocrit levels and excessive polycythemia in patients living at high altitudes. This condition is also termed “Monge’s disease” because it was first described by Peruvian physician Monge [61]. CMS is typically characterized by at least three of the following symptoms: breathlessness, palpitations, sleep disturbance, cyanosis, vein dilatation, paresthesia, headache, or tinnitus, according to the Qinghai International Consensus scoring system [5]. This is a serious disease that is associated with morbidity and mortality [62]. In contrast to other pathologies associated with high altitude, CMS is found in residents living at high altitude (exposed to chronic hypobaric hypoxia), which is estimated to include 85 million people [63]; the prevalence of CMS (10–15%) can vary by altitude level, age, and genetic factors [5,64].
Interestingly, previous studies on residents at high altitude have shown that those with CMS present with higher oxidative stress markers (e.g., ascorbate radicals) and low antioxidant biomarkers (e.g., ascorbate) compared to residents at high altitude without CMS [65]. In addition, a recent CMS study by Bailey et al. [62] showed disequilibrium between free radical formation and antioxidant defenses in residents at high altitude, causing chronic systemic oxidative stress and inflammation. Moreover, this study showed that these oxidative and inflammatory responses are associated with accelerated cognitive decline and depression in residents at high altitude, particularly those with CMS [62].
In addition, 8-iso-PGF2α, an oxidative biomarker and a potent vasoconstrictor, is increased in people with CMS compared to people without CMS who live in the same high-altitude conditions. Furthermore, 8-iso-PGF2α is negatively associated with nocturnal saturation of oxygen and positively associated with hemoglobin concentration without alterations to SOD activity [66]. Moreover, a study on rats with CMS induced by chronic hypobaric hypoxia (5000 m; 30 days) demonstrated that in the CMS model group, the MDA level was significantly higher than that in the control group, and the concentrations of SOD and GPx were decreased [67]. Additionally, through a proteomic and metabolomics analysis, a recent study on rats with CMS induced by 28-day exposure to hypobaric hypoxia reported an increase in 8-hydroxyguanosine and trimethylamine N-oxide, which were associated with oxidative stress and pulmonary vasoconstriction processes, respectively [68] (Table 1).
Moreover, a study based on a proteomic analysis validated the potential plasma biomarkers in residents at high altitude with CMS, showing that hydrogen peroxide metabolic/catabolic, ROS metabolic, and acute inflammatory responses were significantly elevated. A protein–protein interaction analysis revealed that antioxidant activity, the hydrogen peroxide-induced catabolic process, and peroxidase and thioredoxin-1 activities constituted the key nodes in the molecular networks of CMS [8]. Therefore, it is important to consider increases in oxidative molecules (e.g., hydrogen peroxide) and oxidative biomarkers (e.g., MDA and 8-iso-PGF2α) and alterations to antioxidant activity (e.g., SOD and GSH-Px activity) as possible biomarkers for CMS in future studies.

3.5. High-Altitude Pulmonary Hypertension

One of the principal effects of hypobaric hypoxia exposure is hypoxic pulmonary artery vasoconstriction (HPV), which leads to the redistribution of blood to lung areas with the greatest ventilation [69]. Notably, when HPV is exacerbated and/or permanent, the pulmonary artery activates the vascular remodeling process, which increases vascular resistance and pulmonary artery pressure, leading to pulmonary hypertension—specifically HAPH [70,71,72].
Several studies have shown that increased ROS production induced by hypoxia is important to the development of HPV [73,74]. Interestingly, a recent study with rats under chronic hypobaric hypoxia (4200 m; 28 days) showed gradually decreased GPx and SOD activity on day 7 of high-altitude exposure; in contrast, the generation of both MDA and ROS increased in a time-dependent manner. Early high-altitude exposure first activates oxidative stress pathways; then, it gradually activates endoplasmic reticulum stress, which might promote apoptosis of alveolar epithelial cells.
The remodeling of pulmonary vessels has been correlated with the oxidative stress response during HAPH development [75]. This is supported by a study conducted by Bailey et al. [76] that exposed human subjects to acute hypobaric hypoxia (4559; 4–6 h) and found that the increases in pulmonary artery systolic pressure and vascular resistance observed at high altitude were associated with ROS molecules (ascorbate free radicals and lipid hydroperoxides), which altered endogenous vasodilator molecules, such as nitric oxide.
One of the principal consequences of HAPH is the development of right ventricular hypertrophy (RVH) due to excess pressure, which, in some cases, can trigger heart failure and ultimately lead to death [77]. Studies have also described the role of oxidative stress in the development of RVH under hypobaric hypoxia conditions, where it has been demonstrated that rats with RVH induced by exposure to chronic intermittent hypobaric hypoxia showed overexpression of NADPH oxidase complex-2 (Nox2) and p22phox and an increase in lipid peroxidation levels [37,78]. The link between RVH and oxidative stress is further supported by a study on rats under chronic hypobaric hypoxia conditions (5475 m; 28 days), which found that pulmonary artery vascular remodeling, RVH, and increased right ventricular systolic pressure occurred by the end of exposure; all of these effects were related to increased H2O2 and decreased SOD and GSH levels in the lung [79]. These results highlight the influence of oxidative stress on the pathological process of HAPH and the collateral cor pulmonale effects.

Pulmonary Artery Smooth Muscle Cell (PASMC) Remodeling

Several molecular alterations are involved in the development of pulmonary artery remodeling and subsequent HAPH, such as a decrease in the level of the endogenous vasodilator nitric oxide (NO) [6] and increases in asymmetric dimethyl arginine (ADMA) activity [80], intracellular calcium concentration ([Ca2+]i) [81], ET-1 expression [82], and IL-6 expression [83,84]; studies have demonstrated that all these molecules are related to oxidative stress, as discussed below.
The NADPH oxidase complex (specifically the Nox4 isoform) is considered to be a principal source of vascular pulmonary artery ROS production, which may be related to PASMC proliferation and pulmonary hypertension induced by hypobaric hypoxia [14]. NADPH is an electron donor complex that reduces molecular oxygen to superoxide (O2) or H2O2, depending on the isoform [85]; the predominant isoform in the pulmonary vascular system is Nox4, which has been shown to be overexpressed during pulmonary artery remodeling and in subsequent hypoxia-induced pulmonary artery hypertension (normobaric hypoxia; 10% O2; [86]). In particular, one study of chronic hypobaric hypoxia (4600 m; 30 days) reported an increase in the expression (mRNA and protein levels) of the Nox4 isoform and elevated levels of MDA in the lungs of rats with HAPH [14], findings corroborated by other studies [84,87]. A previous study on rats under chronic hypobaric hypoxia conditions (380 torr, four weeks) showed that the Nox2 isoform is necessary for HPV [88] (Table 1). Therefore, both Nox2 and Nox4 activity may directly or indirectly contribute to the development of HAPH induced by hypobaric hypoxia.
Additionally, given these findings, we hypothesize that the Nox2 isoform may be more closely related to the physiological HPV response and that Nox4 might contribute to the transition of HPV to PASMC remodeling and the development of subsequent pathology (HAPH) under hypobaric hypoxia. However, to confirm this hypothesis, further studies are needed.
NO bioavailability directly impacts PASMCs, inducing vasoconstriction and subsequent pulmonary hypertension [6]. The possible mechanism relating NO decrease to oxidative stress involves the reaction between superoxide and NO molecules that yields ONOO, which is an oxidative molecule that contributes to exacerbated oxidative stress [6,14,86].
As previously mentioned, hypoxia-induced pulmonary hypertension is characterized by sustained pulmonary artery vasoconstriction and vascular remodeling, which are both mediated by PASMC contraction and proliferation. It is important to note that [Ca2+]i is a major trigger for pulmonary vasoconstriction and an important stimulus for cell proliferation in PASMCs [89].
Studies have shown that oxidative stress or exacerbated levels of ROS (specifically superoxide and hydrogen peroxide) are necessary to increase [Ca2+]i under hypoxic conditions; this concentration increase is realized through voltage-dependent calcium channels (VDCCs) in PASMCs [89,90,91] and causes PASMC contraction and increases in pulmonary artery pressure.
As mentioned above, several studies have determined that under hypoxic conditions, ET-1 is produced by various cell types in the vascular system, such as endothelial cells, macrophages, fibroblasts, and smooth muscle cells (SMCs) [92,93,94]. Studies have demonstrated that the increase in ET-1 expression may be associated with oxidative stress, increasing the proliferation of SMCs through activation of the NADPH oxidase complex [95,96].
Therefore, oxidative molecules (O2 and H2O2) and activation of NADPH oxidase (specifically the Nox4 isoform) may be related to altered expressions or concentrations of several molecules (Ca2+, NO, and ET-1) involved in the development of PASMC remodeling and subsequent HAPH.
Finally, we want to highlight the paucity of studies relating oxidative stress and high-altitude diseases that prompted us to conduct this review.

4. Conclusions

Exposure to high altitude (acute, chronic, and intermittent) increases the level of ROS induced by hypobaric hypoxia and typically decreases the effects of the antioxidant system, resulting in oxidative damage to several tissues and contributing to the development of several high-altitude illnesses.
The high-altitude diseases discussed in this updated review, such as AMS, HACE, and HAPE, are related to increased MDA levels and decreased SOD and GPx antioxidant activity. Additionally, oxidative stress contributes to the development of HACE and HAPE pathologies through the activation of inflammatory pathways induced by oxidative stress. Studies have shown an increase in MDA, 8-iso-PGF2α, and hydrogen peroxide levels and a decrease in SOD and GPx activity in CMS, all of which seem to be important biomarkers of this disease. Finally, in HAPH, the increase in Nox4 activity and ROS levels may contribute to the pulmonary artery remodeling process. Therefore, more studies on oxidative stress in high-altitude illnesses are necessary since ROS appear to be key to the development of high-altitude diseases. Antioxidant treatments could help to mitigate these high-altitude pathologies; however, since ROS molecules are related to physiological processes, these treatments should be carefully studied, and we urge caution when drawing conclusions about these treatments or extrapolating them to humans.

Author Contributions

The conceptualization, investigation, resources, writing—original draft preparation, writing—review and editing, visualization, supervision, project administration, funding acquisition, were realized by all authors (E.P., S.E.A., P.S. and J.B.). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by GORE-FIC TARAPACA grant number BIP30477541-0.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMSAcute mountain sickness
CATCatalase
CMS Chronic mountain sickness
ET-1Endothelin-1
GPxGlutathione peroxidase
GRGlutathione reductase
GSHGlutathione
H2O2Hydrogen peroxide
HACEHigh-altitude cerebral edema
HAPEHigh-altitude pulmonary edema
HAPHHigh-altitude pulmonary hypertension
HIF-1αHypoxia-inducible factor 1-alpha
HPVHypoxic pulmonary vasoconstriction
IL-1βInterleukin-1β
IL-6Interleukin-6
MCP-1Monocyte chemoattractant protein-1
MDAMalondialdehyde
NF-κBNuclear factor kappa B
NONitric oxide
O2Superoxide
•OH Hydroxyl radical
ONOOPeroxynitrite
PASMCsPulmonary artery smooth muscle cells
PI3KPhosphoinositide 3-kinase
ROSReactive oxygen species
RVHRight ventricular hypertrophy
SMCsSmooth muscle cells
SODSuperoxide dismutase
TNF-αTumor necrosis factor-alpha
VEGFVascular endothelial growth factor

References

  1. Tremblay, J.C.; Ainslie, P.N. Global and country-level estimates of human population at high altitude. Proc. Natl. Acad. Sci. USA 2021, 118, e2102463118. [Google Scholar] [CrossRef] [PubMed]
  2. Heath, D.; Williams, D. The carotid bodies. In Man at High Altitude; Heath, D., Williams, D., Eds.; Churchill Livingstone: New York, NY, USA, 1977; pp. 60–82. [Google Scholar]
  3. Luks, A.M.; Swenson, E.R.; Bärtsch, P. Acute high-altitude sickness. Eur. Respir. Rev. 2017, 26, 160096. [Google Scholar] [CrossRef] [PubMed]
  4. Askew, E.W. Work at high altitude and oxidative stress: Antioxidant nutrients. Toxicology 2002, 180, 107–119. [Google Scholar] [CrossRef]
  5. León-Velarde, F.; Maggiorini, M.; Reeves, J.T.; Aldashev, A.; Asmus, I.; Bernardi, L.; Ge, R.-L.; Hackett, P.; Kobayashi, T.; Moore, L.G.; et al. Consensus statement on chronic and subacute high altitude diseases. High Alt. Med. Biol. 2005, 6, 147–157. [Google Scholar] [CrossRef] [PubMed]
  6. Siques, P.; de Pablo, A.L.L.; Brito, J.; Arribas, S.M.; Flores, K.; Arriaza, K.; Naveas, N.; González, M.C.; Hoorntje, A.; León-Velarde, F.; et al. Nitric oxide and superoxide anion balance in rats exposed to chronic and long term intermittent hypoxia. BioMed Res. Int. 2014, 2014, 610474. [Google Scholar] [CrossRef] [PubMed]
  7. Monge-C., C.; Arregui, A.; León-Velarde, F. Pathophysiology and epidemiology of chronic mountain-sickness. Int. J. Sports Med. 1992, 13, S79–S81. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, P.; Li, Z.; Yang, F.; Ji, L.; Yang, Y.; Liu, C.; Liu, H.; Ma, J.; Liu, J.; Dang, Z.; et al. Novel insights into plasma biomarker candidates in patients with chronic mountain sickness based on proteomics. Biosci. Rep. 2021, 41, BSR20202219. [Google Scholar] [CrossRef]
  9. Jefferson, J.A.; Simoni, J.; Escudero, E.; Hurtado, M.E.; Swenson, E.R.; Wesson, D.E.; Schreiner, G.F.; Schoene, R.B.; Johnson, R.J.; Hurtado, A. Increased oxidative stress following acute and chronic high altitude exposure. High Alt. Med. Biol. 2004, 5, 61–69. [Google Scholar] [CrossRef]
  10. Sarada, S.; Himadri, P.; Mishra, C.; Geetali, P.; Ram, M.S.; Ilavazhagan, G. Role of oxidative stress and NFkB in hypoxia-induced pulmonary edema. Exp. Biol. Med. 2008, 233, 1088–1098. [Google Scholar] [CrossRef]
  11. Dosek, A.; Ohno, H.; Acs, Z.; Taylor, A.W.; Radak, Z. High altitude and oxidative stress. Respir. Physiol. Neurobiol. 2007, 158, 128–131. [Google Scholar] [CrossRef]
  12. Araneda, O.F.; García, C.; Lagos, N.; Quiroga, G.; Cajigal, J.; Salazar, M.P.; Behn, C. Lung oxidative stress as related to exercise and altitude. Lipid peroxidation evidence in exhaled breath condensate: A possible predictor of acute mountain sickness. Eur. J. Appl. Physiol. 2005, 95, 383–390. [Google Scholar] [CrossRef]
  13. Purushothaman, J.; Suryakumar, G.; Shukla, D.; Malhotra, A.S.; Kasiganesan, H.; Kumar, R.; Sawhney, R.C.; Chami, A. Modulatory effects of seabuckthorn (Hippophae rhamnoides L.) in hypobaric hypoxia induced cerebral vascular injury. Brain Res. Bull. 2008, 77, 246–252. [Google Scholar] [CrossRef] [PubMed]
  14. Lüneburg, N.; Siques, P.; Brito, J.; Arriaza, K.; Pena, E.; Klose, H.; Leon-Velarde, F.; Böger, R.H. Long-term chronic intermittent hypobaric hypoxia in rats causes an imbalance in the asymmetric dimethylarginine/nitric oxide pathway and ROS activity: A possible synergistic mechanism for altitude pulmonary hypertension? Pulm. Med. 2016, 2016, 6578578. [Google Scholar] [CrossRef] [Green Version]
  15. Takimoto, E.; Kass, D.A. Role of oxidative stress in cardiac hypertrophy and remodeling. Hypertension 2007, 49, 241–248. [Google Scholar] [CrossRef]
  16. Li, R.; Jia, Z.; Trush, M.A. Defining ROS in biology and medicine. React. Oxyg. Species 2016, 1, 9–21. [Google Scholar] [CrossRef] [Green Version]
  17. Brieger, K.; Schiavone, S.; Miller, F.J.; Krause, K.H. Reactive oxygen species: From health to disease. Swiss Med. Wkly. 2012, 142, w13659. [Google Scholar] [CrossRef]
  18. Yan, S.; Resta, T.C.; Jernigan, N.L. Vasoconstrictor mechanisms in chronic hypoxia-induced pulmonary hypertension: Role of oxidant signaling. Antioxidants 2020, 9, 999. [Google Scholar] [CrossRef] [PubMed]
  19. Liu, J.Q.; Folz, R.J. Extracellular superoxide enhances 5-HT-induced murine pulmonary artery vasoconstriction. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 287, L111–L118. [Google Scholar] [CrossRef] [Green Version]
  20. Billaud, M.; Marthan, R.; Savineau, J.P.; Guibert, C. Vascular smooth muscle modulates endothelial control of vasoreactivity via reactive oxygen species production through myoendothelial communications. PLoS ONE 2009, 4, e6432. [Google Scholar] [CrossRef]
  21. Ward, J.P.T. Physiological redox signalling and regulation of ion channels: Implications for pulmonary hypertension. Exp. Physiol. 2017, 102, 1078–1082. [Google Scholar] [CrossRef]
  22. Peoples, J.N.; Saraf, A.; Ghazal, N.; Pham, T.T.; Kwong, J.Q. Mitochondrial dysfunction and oxidative stress in heart disease. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [PubMed]
  23. Richardson, A.G.; Schadt, E.E. The role of macromolecular damage in aging and age-related disease. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, S28–S32. [Google Scholar] [CrossRef] [PubMed]
  24. Pluquet, O.; Pourtier, A.; Abbadie, C. The unfolded protein response and cellular senescence. A review in the theme: Cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. Am. J. Physiol. Cell Physiol. 2015, 308, C415–C425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Amodio, G.; Moltedo, O.; Faraonio, R.; Remondelli, P. Targeting the endoplasmic reticulum unfolded protein response to counteract the oxidative stress-induced endothelial dysfunction. Oxid. Med. Cell. Longev. 2018, 2018, 4946289. [Google Scholar] [CrossRef] [PubMed]
  26. Marciniak, S.J.; Ron, D. Endoplasmic reticulum stress signaling in disease. Physiol. Rev. 2006, 86, 1133–1149. [Google Scholar] [CrossRef] [PubMed]
  27. Mrakic-Sposta, S.; Gussoni, M.; Dellanoce, C.; Marzorati, M.; Montorsi, M.; Rasica, L.; Pratali, L.; D’Angelo, G.; Martinelli, M.; Bastiani, L.; et al. Effects of acute and sub-acute hypobaric hypoxia on oxidative stress: A field study in the Alps. Eur. J. Appl. Physiol. 2021, 121, 297–306. [Google Scholar] [CrossRef] [PubMed]
  28. Beall, C.M. High-altitude adaptations. Lancet 2003, 362, s14–s15. [Google Scholar] [CrossRef]
  29. Horscroft, J.A.; Kotwica, A.O.; Laner, V.; West, J.A.; Hennis, P.J.; Levett, D.Z.H.; Howard, D.J.; Fernandez, B.O.; Burgess, S.L.; Ament, Z.; et al. Metabolic basis to Sherpa altitude adaptation. Proc. Natl. Acad. Sci. USA 2017, 114, 6382–6387. [Google Scholar] [CrossRef] [Green Version]
  30. Julian, C.G.; Vargas, E.; Browne, V.A.; Wilson, M.J.; Bigham, A.W.; Rodriguez, C.; McCord, J.M.; Moore, L.G. Potential role for elevated maternal enzymatic antioxidant status in Andean protection against altitude-associated SGA. J. Matern. Fetal Neonatal. Med. 2012, 25, 1233–1240. [Google Scholar] [CrossRef]
  31. Roach, R.C.; Hackett, P.H.; Oelz, O.; Bartsch, P.; Luks, A.M.; MacInnis, M.J.; Baillie, J.K.; Lake Louise AMS Score Consensus Committee. The 2018 Lake Louise acute mountain sickness score. High Alt. Med. Biol. 2018, 19, 4–6. [Google Scholar] [CrossRef]
  32. Brito, J.; Siqués, P.; León-Velarde, F.; De La Cruz, J.J.; López, V.; Herruzo, R. Chronic intermittent hypoxia at high altitude exposure for over 12 years: Assessment of hematological, cardiovascular, and renal effects. High Alt. Med. Biol. 2007, 8, 236–244. [Google Scholar] [CrossRef] [PubMed]
  33. Imray, C.; Wright, A.; Subudhi, A.; Roach, R. Acute mountain sickness: Pathophysiology, prevention, and treatment. Prog. Cardiovasc. Dis. 2010, 52, 467–484. [Google Scholar] [CrossRef] [PubMed]
  34. Luks, A.M.; Auerbach, P.S.; Freer, L.; Grissom, C.K.; Keyes, L.E.; McIntosh, S.E.; Rodway, G.W.; Schoene, R.B.; Zafren, K.; Hackett, P.H. Wilderness medical society clinical practice guidelines for the prevention and treatment of acute altitude illness: 2019 update. Wilderness Environ. Med. 2019, 30, S3–S18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Jing, L.; Shao, J.; Zhao, T.; He, L.; Ma, H. Protective effect of 5,6,7,8-trtrahydroxyflavone against acute hypobaric hypoxia induced-oxidative stress in mice. Pak. J. Pharm. Sci. 2021, 34, 513–519. [Google Scholar]
  36. Agrawal, A.; Rathor, R.; Suryakumar, G. Oxidative protein modification alters proteostasis under acute hypobaric hypoxia in skeletal muscles: A comprehensive in vivo study. Cell Stress Chaperones 2017, 22, 429–443. [Google Scholar] [CrossRef]
  37. Pena, E.; Siques, P.; Brito, J.; Arribas, S.M.; Böger, R.; Hannemann, J.; León-Velarde, F.; González, M.C.; López, M.R.; de Pablo, Á.L.L. Nox2 upregulation and p38α MAPK activation in right ventricular hypertrophy of rats exposed to long-term chronic intermittent hypobaric hypoxia. Int. J. Mol. Sci. 2020, 21, 8576. [Google Scholar] [CrossRef]
  38. Tang, X.G.; Wen, J.; Zhang, X.S.; Jiang, D.C. Association between decreased osteopontin and acute mountain sickness upon rapid ascent to 3500 m among young Chinese men. J. Travel Med. 2018, 25, tay075. [Google Scholar] [CrossRef] [Green Version]
  39. Bailey, D.M.; Davies, B.; Young, I.S.; Hullin, D.A.; Seddon, P.S. A potential role for free radical-mediated skeletal muscle soreness in the pathophysiology of acute mountain sickness. Aviat. Space Environ. Med. 2001, 72, 513–521. [Google Scholar]
  40. Bailey, D.M.; Davies, B. Acute mountain sickness; prophylactic benefits of antioxidant vitamin supplementation at high altitude. High Alt. Med. Biol. 2001, 2, 21–29. [Google Scholar] [CrossRef]
  41. Irarrázaval, S.; Allard, C.; Campodónico, J.; Pérez, D.; Strobel, P.; Vásquez, L.; Urquiaga, I.; Echeverría, G.; Leighton, F. Oxidative stress in acute hypobaric hypoxia. High Alt. Med. Biol. 2017, 18, 128–134. [Google Scholar] [CrossRef]
  42. Debevec, T.; Pialoux, V.; Poussel, M.; Willis, S.J.; Martin, A.; Osredkar, D.; Millet, G.P. Cardio-respiratory, oxidative stress and acute mountain sickness responses to normobaric and hypobaric hypoxia in prematurely born adults. Eur. J. Appl. Physiol. 2020, 120, 1341–1355. [Google Scholar] [CrossRef] [PubMed]
  43. Julian, C.G.; Subudhi, A.W.; Hill, R.C.; Wilson, M.J.; Dimmen, A.C.; Hansen, K.C.; Roach, R.C. Exploratory proteomic analysis of hypobaric hypoxia and acute mountain sickness in humans. J. Appl. Physiol. 2014, 116, 937–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Lu, H.; Zhang, H.; Jiang, Y. Methazolamide in high-altitude illnesses. Eur. J. Pharm. Sci. 2020, 148, 105326. [Google Scholar] [CrossRef] [PubMed]
  45. Sarang, S.S.; Yoshida, T.; Cadet, R.; Valeras, A.S.; Jensen, R.V.; Gullans, S.R. Discovery of molecular mechanisms of neuroprotection using cell-based bioassays and oligonucleotide arrays. Physiol. Genom. 2002, 11, 45–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Basnyat, B. High altitude cerebral and pulmonary edema. Travel Med. Infect. Dis. 2005, 3, 199–211. [Google Scholar] [CrossRef] [PubMed]
  47. Bartsch, P.; Bailey, D.M.; Berger, M.M.; Knauth, M.; Baumgartner, R.W. Acute mountain sickness: Controversies and advances. High Alt. Med. Biol. 2004, 5, 110–124. [Google Scholar] [CrossRef]
  48. Himadri, P.; Kumari, S.S.; Chitharanjan, M.; Dhananjay, S. Role of oxidative stress and inflammation in hypoxia-induced cerebral edema: A molecular approach. High Alt. Med. Biol. 2010, 11, 231–244. [Google Scholar] [CrossRef]
  49. Jensen, J.D.; Vincent, A.L. High Altitude Cerebral Edema; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  50. Bärtsch, P.; Swenson, E.R. Clinical practice: Acute high-altitude illnesses. N. Engl. J. Med. 2013, 368, 2294–2302. [Google Scholar] [CrossRef] [PubMed]
  51. Chan, P.H.; Schmidley, J.W.; Fishman, R.A.; Longar, S.M. Brain injury, edema, and vascular permeability changes induced by oxygen-derived free radicals. Neurology 1984, 34, 315–320. [Google Scholar] [CrossRef]
  52. Bakonyi, T.; Radak, Z. High altitude and free radicals. J. Sports Sci. Med. 2004, 3, 64–69. [Google Scholar]
  53. Sarada, S.K.S.; Titto, M.; Himadri, P.; Saumya, S.; Vijayalakshmi, V. Curcumin prophylaxis mitigates the incidence of hypobaric hypoxia-induced altered ion channels expression and impaired tight junction proteins integrity in rat brain. J. Neuroinflamm. 2015, 12, 113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gong, G.; Yin, L.; Yuan, L.; Sui, D.; Sun, Y.; Fu, H.; Chen, L.; Wang, X. Ganglioside GM1 protects against high altitude cerebral edema in rats by suppressing the oxidative stress and inflammatory response via the PI3K/AKT-Nrf2 pathway. Mol. Immunol. 2018, 95, 91–98. [Google Scholar] [CrossRef] [PubMed]
  55. Hopkins, S.R.; Garg, J.; Bolar, D.S.; Balouch, J.; Levin, D.L. Pulmonary blood flow heterogeneity during hypoxia and high-altitude pulmonary edema. Am. J. Respir. Crit. Care Med. 2005, 171, 83–87. [Google Scholar] [CrossRef] [PubMed]
  56. Maggiorini, M.; Brunner-La Rocca, H.P.; Peth, S.; Fischler, M.; Böhm, T.; Bernheim, A.; Kiencke, S.; Bloch, K.E.; Dehnert, C.; Naeije, R.; et al. Both tadalafil and dexamethasone may reduce the incidence of high-altitude pulmonary edema. Ann. Intern. Med. 2006, 145, 497–506. [Google Scholar] [CrossRef] [PubMed]
  57. Hackett, P.H.; Roach, R.C. High-altitude illness. N. Engl. J. Med. 2001, 345, 107–114. [Google Scholar] [CrossRef]
  58. Sharma, S.; Singh, Y.; Sandhir, R.; Singh, S.; Ganju, L.; Kumar, B.; Varshney, R. Mitochondrial DNA mutations contribute to high altitude pulmonary edema via increased oxidative stress and metabolic reprogramming during hypobaric hypoxia. Biochim. Biophys. Acta (BBA)—Bioenerg. 2021, 1862, 148431. [Google Scholar] [CrossRef]
  59. Purushothaman, J.; Suryakumar, G.; Shukla, D.; Jayamurthy, H.; Kasiganesan, H.; Kumar, R.; Sawhney, R.C. Modulation of hypoxia-induced pulmonary vascular leakage in rats by seabuckthorn (Hippophae rhamnoides L.). Evid.-Based Complement. Altern. Med. 2011, 2011, 574524. [Google Scholar] [CrossRef] [Green Version]
  60. Lee, S.Y.; Li, M.H.; Shi, L.S.; Chu, H.; Ho, C.W.; Chang, T.C. Rhodiola crenulata extract alleviates hypoxic pulmonary edema in rats. Evid.-Based Complement. Altern. Med. 2013, 2013, 718739. [Google Scholar] [CrossRef] [Green Version]
  61. Monge, C. High altitude disease. Arch. Intern. Med. 1937, 59, 32–40. [Google Scholar] [CrossRef]
  62. Bailey, D.M.; Brugniaux, J.V.; Filipponi, T.; Marley, C.J.; Stacey, B.; Soria, R.; Rimoldi, S.F.; Cerny, D.; Rexhaj, E.; Pratali, L.; et al. Exaggerated systemic oxidative-inflammatory-nitrosative stress in chronic mountain sickness is associated with cognitive decline and depression. J. Physiol. 2019, 597, 611–629. [Google Scholar] [CrossRef] [Green Version]
  63. Brito, J.; Siques, P.; Pena, E. Long-term chronic intermittent hypoxia: A particular form of chronic high-altitude pulmonary hypertension. Pulm. Circ. 2020, 10, 5–12. [Google Scholar] [CrossRef] [PubMed]
  64. Beall, C.M. Adaptation to high altitude: Phenotypes and genotypes. Annu. Rev. Anthropol. 2014, 43, 251–272. [Google Scholar] [CrossRef]
  65. Bailey, D.M.; Rimoldi, S.F.; Rexhaj, E.; Pratali, L.; Salmòn, C.S.; Villena, M.; McEneny, J.; Young, I.S.; Nicod, P.; Allemann, Y.; et al. Oxidative-nitrosative stress and systemic vascular function in highlanders with and without exaggerated hypoxemia. Chest 2013, 143, 444–451. [Google Scholar] [CrossRef] [PubMed]
  66. Julian, C.G.; Vargas, E.; Gonzales, M.; Dávila, R.D.; Ladenburger, A.; Reardon, L.; Schoo, C.; Powers, R.W.; Lee-Chiong, T.; Moore, L.G. Sleep-disordered breathing and oxidative stress in preclinical chronic mountain sickness (excessive erythrocytosis). Respir. Physiol. Neurobiol. 2013, 186, 188–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Maimaitiyiming, D.; Hu, G.; Aikemu, A.; Hui, S.W.; Zhang, X. The treatment of Uygur medicine Dracocephalum moldavica L on chronic mountain sickness rat model. Pharmacogn. Mag. 2014, 10, 477–482. [Google Scholar] [CrossRef] [Green Version]
  68. Gao, X.; Zhang, Z.; Li, X.; Li, C.; Hao, J.; Luo, Y.; Lei, M.; Li, J.; Liu, C.; He, K. Macitentan attenuates chronic mountain sickness in rats by regulating arginine and purine metabolism. J. Proteome Res. 2020, 19, 3302–3314. [Google Scholar] [CrossRef]
  69. Euler, U.S.V.; Liljestrand, G. Observations on the pulmonary arterial blood pressure in the cat. Acta Physiol. Scand. 1946, 12, 301–320. [Google Scholar] [CrossRef]
  70. Sommer, N.; Dietrich, A.; Schermuly, R.T.; Ghofrani, H.A.; Gudermann, T.; Schulz, R.; Seeger, W.; Grimminger, F.; Weissmann, N. Regulation of hypoxic pulmonary vasoconstriction: Basic mechanisms. Eur. Respir. J. 2008, 32, 1639–1651. [Google Scholar] [CrossRef] [Green Version]
  71. Neupane, M.; Swenson, E.R. High-altitude pulmonary vascular diseases. Adv. Pulm. Hypertens. 2017, 15, 149–157. [Google Scholar] [CrossRef] [Green Version]
  72. Ulloa, N.A.; Cook, J. Altitude Induced Pulmonary Hypertension; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  73. Weir, E.K.; Archer, S.L. The mechanism of acute hypoxic pulmonary vasoconstriction: The tale of two channels. FASEB J. 1995, 9, 183–189. [Google Scholar] [CrossRef]
  74. Desireddi, J.R.; Farrow, K.N.; Marks, J.D.; Waypa, G.B.; Schumacker, P.T. Hypoxia increases ROS signaling and cytosolic Ca2+ in pulmonary artery smooth muscle cells of mouse lungs slices. Antioxid. Redox Signal. 2010, 12, 595–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Pu, X.; Lin, X.; Duan, X.; Wang, J.; Shang, J.; Yun, H.; Chen, Z. Oxidative and endoplasmic reticulum stress responses to chronic high-altitude exposure during the development of high-altitude pulmonary hypertension. High Alt. Med. Biol. 2020, 21, 378–387. [Google Scholar] [CrossRef] [PubMed]
  76. Bailey, D.M.; Dehnert, C.; Luks, A.M.; Menold, E.; Castell, C.; Schendler, G.; Faoro, V.; Gutowski, M.; Evans, K.A.; Taudorf, S.; et al. High-altitude pulmonary hypertension is associated with a free radical-mediated reduction in pulmonary nitric oxide bioavailability. J. Physiol. 2010, 588, 4837–4847. [Google Scholar] [CrossRef] [PubMed]
  77. Penaloza, D.; Arias-Stella, J. The heart and pulmonary circulation at high altitudes: Healthy highlanders and chronic mountain sickness. Circulation 2007, 115, 1132–1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Pena, E.; Brito, J.; El Alam, S.; Siques, P. Oxidative stress, kinase activity and inflammatory implications in right ventricular hypertrophy and heart failure under hypobaric hypoxia. Int. J. Mol. Sci. 2020, 21, 6421. [Google Scholar] [CrossRef]
  79. Xu, D.; Li, Y.; Zhang, B.; Wang, Y.; Liu, Y.; Luo, Y.; Niu, W.; Dong, M.; Liu, M.; Dong, H.; et al. Resveratrol alleviate hypoxic pulmonary hypertension via anti-inflammation and anti-oxidant pathways in rats. Int. J. Med. Sci. 2016, 13, 942–954. [Google Scholar] [CrossRef] [Green Version]
  80. Siques, P.; Brito, J.; Schwedhelm, E.; Pena, E.; León-Velarde, F.; De La Cruz, J.J.; Böger, R.H.; Hannemann, J. Asymmetric dimethylarginine at sea level is a predictive marker of hypoxic pulmonary arterial hypertension at high altitude. Front. Physiol. 2019, 10, 651. [Google Scholar] [CrossRef] [PubMed]
  81. Sydykov, A.; Mamazhakypov, A.; Maripov, A.; Kosanovic, D.; Weissmann, N.; Ghofrani, H.A.; Sarybaev, A.S.; Schermuly, R.T. Pulmonary hypertension in acute and chronic high altitude maladaptation disorders. Int. J. Environ. Res. Public Health 2021, 18, 1692. [Google Scholar] [CrossRef]
  82. Berger, M.M.; Dehnert, C.; Bailey, D.M.; Luks, A.M.; Menold, E.; Castell, C.; Schendler, G.; Faoro, V.; Mairbäurl, H.; Bärtsch, P.; et al. Transpulmonary plasma ET-1 and nitrite differences in high altitude pulmonary hypertension. High Alt. Med. Biol. 2009, 10, 17–24. [Google Scholar] [CrossRef]
  83. Siques, P.; Brito, J.; Pena, E. Reactive oxygen species and pulmonary vasculature during hypobaric hypoxia. Front. Physiol. 2018, 9, 865. [Google Scholar] [CrossRef] [Green Version]
  84. Siques, P.; Pena, E.; Brito, J.; El Alam, S. Oxidative stress, kinase activation, and inflammatory pathways involved in effects on smooth muscle cells during pulmonary artery hypertension under hypobaric hypoxia exposure. Front. Physiol. 2021, 12, 690341. [Google Scholar] [CrossRef] [PubMed]
  85. Bache, R.J.; Chen, Y. NOX2-induced myocardial fibrosis and diastolic dysfunction. J. Am. Coll. Cardiol. 2014, 63, 2742–2744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Barman, S.A.; Chen, F.; Su, Y.; Dimitropoulou, C.; Wang, Y.; Catravas, J.D.; Han, W.; Orfi, L.; Szantai-Kis, C.; Keri, G.; et al. NADPH oxidase 4 is expressed in pulmonary artery adventitia and contributes to hypertensive vascular remodeling. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1704–1715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Klemm, D.J.; Majka, S.M.; Crossno, J.T.; Psilas, J.C.; Reusch, J.E.B.; Garat, C.V. Reduction of reactive oxygen species prevents hypoxia-induced CREB depletion in pulmonary artery smooth muscle cells. J. Cardiovasc. Pharmacol. 2011, 58, 181–191. [Google Scholar] [CrossRef] [Green Version]
  88. Norton, C.E.; Broughton, B.R.S.; Jernigan, N.L.; Walker, B.R.; Resta, T.C. Enhanced depolarization-induced pulmonary vasoconstriction following chronic hypoxia requires EGFR-dependent activation of NAD(P)H oxidase 2. Antioxid. Redox Signal. 2013, 18, 1777–1788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Wan, J.; Yamamura, A.; Zimnicka, A.M.; Voiriot, G.; Smith, K.A.; Tang, H.; Ayon, R.J.; Choudhury, M.S.; Ko, E.A.; Wang, J.; et al. Chronic hypoxia selectively enhances L- and T-type voltage-dependent Ca2+ channel activity in pulmonary artery by upregulating Cav1.2 and Cav3.2. Am. J. Physiol. Lung Cell. Mol. Physiol. 2013, 305, L154–L164. [Google Scholar] [CrossRef] [Green Version]
  90. Di Mise, A.; Wang, Y.X.; Zheng, Y.M. Role of transcription factors in pulmonary artery smooth muscle cells: An important link to hypoxic pulmonary hypertension. Adv. Exp. Med. Biol. 2017, 967, 13–32. [Google Scholar] [CrossRef]
  91. Wang, J.; Shimoda, L.A.; Sylvester, J.T. Capacitative calcium entry and TRPC channel proteins are expressed in rat distal pulmonary arterial smooth muscle. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 286, L848–L858. [Google Scholar] [CrossRef]
  92. Yanagisawa, M.; Kurihara, H.; Kimura, S.; Tomobe, Y.; Kobayashi, M.; Mitsui, Y.; Yazaki, Y.; Goto, K.; Masaki, T. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature 1988, 332, 411–415. [Google Scholar] [CrossRef] [Green Version]
  93. Miller, R.C.; Pelton, J.T.; Huggins, J.P. Endothelins—From receptors to medicine. Trends Pharmacol. Sci. 1993, 14, 54–60. [Google Scholar] [CrossRef]
  94. Kosacka, M.; Brzecka, A. Endothelin-1 and LOX-1 as markers of endothelial dysfunction in obstructive sleep apnea patients. Int. J. Environ. Res. Public Health 2021, 18, 1319. [Google Scholar] [CrossRef] [PubMed]
  95. Wedgwood, S.; Dettman, R.W.; Black, S.M. ET-1 stimulates pulmonary arterial smooth muscle cell proliferation via induction of reactive oxygen species. Am. J. Physiol. Lung Cell. Mol. Physiol. 2001, 281, L1058–L1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Wu, W.; Platoshyn, O.; Firth, A.L.; Yuan, J.X.J. Hypoxia divergently regulates production of reactive oxygen species in human pulmonary and coronary artery smooth muscle cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 293, L952–L959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Oxidative stress and antioxidants in high-altitude diseases.
Table 1. Oxidative stress and antioxidants in high-altitude diseases.
PathologySpeciesHypoxic
Condition
Oxidative Molecules and BiomarkersAntioxidant
Molecules
SampleReferences
AMSHumanAcute hypobaric hypoxia (3000–6125 m; 8 days)↑ MDA BreathAraneda et al., 2005 [12]
HumanAcute hypobaric hypoxia (5100 m; 20 days)≈ MDA
↑ Lipid hydroperoxide
Plasma
Serum
Bailey et al., 2001 [39]
HumanAcute hypobaric hypoxia (4220 m; 36 h)↑ MDA↓ GPxPlasmaIrarrázaval et al., 2017 [41]
HumanAcute hypobaric hypoxia (3840 m; 8 h)↑ MDA PlasmaDebevec et al., 2020 [42]
HumanAcute hypobaric hypoxia (4875 m; 9 h) ↑ Peroxiredoxin-6
↑ GPx
↑ Sulfhydryl oxidase 1
PlasmaJulian et al., 2014 [43]
HumanAcute hypobaric hypoxia (3500 m; 24 h)↑ MDA↓ SODPlasmaTang et al., 2018 [38]
RatAcute hypobaric hypoxia (7620 m; 24 h)↑ ROS
↑ MDA
↑ Protein carbonylation
↓ Sulfhydryl contentSkeletal muscleAgrawal et al., 2017 [36]
HACERatAcute hypobaric hypoxia (9144 m; 5 h)↑ MDA↓ GSH
↓ GPx
↓ SOD
BrainPurushothaman et al., 2008 [13]
RatAcute hypobaric hypoxia (7620 m; 48 h)↑ ROS
↑ MDA
↓ GPx
↓ SOD
BrainHimadri et al., 2010 [48]
RatAcute hypobaric hypoxia (7600 m; 24 h)↑ ROS
↑ MDA
↓ GSH
↓ SOD
BrainGong et al., 2018 [54]
HAPEHumanAcute hypobaric hypoxia (3250 m; 7 days)↑ MDA
↑ Protein carbonylation
↓ SOD
↓ GSH/GSSG
PlasmaSharma et al., 2021 [58]
RatAcute hypobaric hypoxia (7620 m; 24 h)↑ ROS
↑ MDA
≈ GSH
↓ GPx
↑ SOD
LungSarada et al., 2008 [10]
RatAcute hypobaric hypoxia (9144 m; 5 h)↑ MDA↓ GSH
↓ GPx
↓ GR
≈ SOD
LungPurushothaman et al., 2011 [59]
RatAcute hypobaric hypoxia (8000 m; 9 h)↑ ROS
↑ MDA
↑ Myeloperoxidase
LungLee et al., 2013 [60]
CMSHumanChronic hypobaric hypoxia (3600 m; high altitude residents)↑ ROS
↑ Ascorbate radical
↓ AscorbatePlasmaBailey et al., 2013 [65]
HumanChronic hypobaric hypoxia (3600 m; high altitude residents)↑ Ascorbate radical↓ GSH/GSSGPlasmaBailey et al., 2019 [62]
HumanChronic hypobaric hypoxia (4300 m; high altitude residents)↑ 8-Iso-PGF2α≈ SODPlasmaJulian et al., 2013 [66]
HumanChronic hypobaric hypoxia (4500 m; high altitude residents)↑ H2O2
↑ Peroxidase activity
↑ Thioredoxin-1PlasmaZhang et al., 2021 [8]
RatChronic hypobaric hypoxia (5000 m; 15 days)↑ MDA↓ GPx
↓ SOD
PlasmaMaimaitiyiming et al., 2014 [67]
RatChronic hypobaric hypoxia (5500 m; 4 weeks)↑ 8-Hydroxyguanosine
↑ Trimethylamine N-oxide
LungGao et al., 2020 [68]
HAPHHumanAcute hypobaric hypoxia (4559 m; 4–6 h)↑ Ascorbate free radical
↑ Lipid hydroperoxide
Plasma
Serum
Bailey et al., 2010 [76]
RatChronic hypobaric hypoxia (4200 m; 28 days)↑ ROS
↑ MDA
↓ GPx
↓ SOD
LungPu et al., 2020 [75]
RatIntermittent hypobaric hypoxia and chronic hypobaric hypoxia (4600 m; 30 days)↑ Nox4
↑ MDA
LungLüneburg et al., 2016 [14]
RatIntermittent hypobaric hypoxia and chronic hypobaric hypoxia (4600 m; 30 days)↑ Superoxide
↑ p22phox
Pulmonary arterySiques et al., 2014 [6]
RatIntermittent hypobaric hypoxia (4600 m; 30 days)↑ MDA
↑ Nox2
↑ p22phox
Right ventricle hypertrophyPena et al., 2020 [37]
AMS, acute mountain sickness; MDA, malondialdehyde; ROS, reactive oxygen species; GPx, glutathione peroxidase; SOD, superoxide dismutase; GSH, glutathione; CAT, catalase; HACE, high-altitude cerebral edema; HAPE, high-altitude pulmonary edema; GSSG, glutathione disulfide; GR, glutathione reductase; CMS, chronic mountain sickness; HAPH, high-altitude pulmonary hypertension; RVH, right ventricle hypertrophy; Nox2, NADPH oxidase-2; Nox4, NADPH oxidase-4; ↑, increased level; ↓, decreased level; ≈, no changes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pena, E.; El Alam, S.; Siques, P.; Brito, J. Oxidative Stress and Diseases Associated with High-Altitude Exposure. Antioxidants 2022, 11, 267. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox11020267

AMA Style

Pena E, El Alam S, Siques P, Brito J. Oxidative Stress and Diseases Associated with High-Altitude Exposure. Antioxidants. 2022; 11(2):267. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox11020267

Chicago/Turabian Style

Pena, Eduardo, Samia El Alam, Patricia Siques, and Julio Brito. 2022. "Oxidative Stress and Diseases Associated with High-Altitude Exposure" Antioxidants 11, no. 2: 267. https://0-doi-org.brum.beds.ac.uk/10.3390/antiox11020267

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop