2Department of Digestive, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
3Department of Cardiology, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
Abstract
High-altitude pulmonary hypertension (HAPH) is a condition characterized by elevated pulmonary arterial pressure exceeding normal physiological values, resulting from a combination of high-altitude low-pressure, hypoxic environments, genetic susceptibility, immune dysfunction, and neurogenic disturbances. This condition predominantly manifests as right heart failure, severely impacting quality of life and life expectancy. Macrophages, as one of the most prevalent innate immune cells, have been increasingly recognized for their crucial role in the pathogenesis of HAPH. The low-pressure and hypoxic environment, along with other etiological factors, lead to metabolic abnormalities in tissue cells and the microenvironment. This results in increased secretion of chemokines, cytokines, and growth factors in the microenvironment, which promote the proliferation of tissue-resident macrophages and the differentiation of monocytes recruited from the blood into macrophages. This exacerbates the inflammatory cascade, further promoting cell proliferation, tissue repair, and inhibition of apoptosis. These processes contribute to the migration and proliferation of pulmonary arterial smooth muscle cells, endothelial cells, and fibroblasts, leading to vascular remodeling and ultimately the development of pulmonary arterial hypertension. This review examines the role of macrophage-mediated immune responses in high-altitude pulmonary arterial hypertension, with a focus on hypoxia as a key feature.
Highlights
- Key Role of Macrophages in High-Altitude Pulmonary Hypertension (HAPH): Research reveals that macrophages play a critical role in the development of HAPH through hypoxia-induced inflammatory responses and vascular remodeling.
- Hypoxia-Induced Hypoxia-Inducible Factors (HIFs) Signaling Pathway Regulation: In hypoxic environments, the stabilization of HIFs (HIF-1α and HIF-2α) promotes the proliferation of pulmonary artery smooth muscle and endothelial cells, further driving vascular remodeling.
- Macrophage Polarization and Pulmonary Vascular Remodeling: The dynamic polarization of M1 and M2 macrophages contributes to the pro-inflammatory and anti-inflammatory phases of HAPH, offering potential intervention targets for disease progression and treatment.
Introduction
High-altitude pulmonary hypertension (HAPH) is a condition affecting adults or children residing at altitudes above 2500 meters. It is caused by a combination of factors including decreased oxygen content due to higher altitudes, increased blood viscosity, enhanced sympathetic nervous activity, and genetic predispositions. These factors contribute to pathological and physiological responses such as proliferation of small pulmonary vessels, pulmonary vasoconstriction, and endothelial cell (EC) damage, leading to elevated PAP. Clinically, this condition is characterized by symptoms such as dyspnea, cough, cyanosis, sleep disturbances, irritability, and right heart failure. The condition is typically defined by a mean PAP (mPAP) ≥30 mm Hg or a systolic PAP ≥50 mm Hg measured at the residence location.1
According to the 2022 European Society of Cardiology and European Respiratory Society guidelines for the diagnosis and treatment of pulmonary hypertension (PH), PH is classified into 5 groups: Group 1—Pulmonary arterial hypertension; Group 2—PH due to left heart disease; Group 3—PH due to lung diseases and/or hypoxia; Group 4—PH due to chronic thromboembolic disease; and Group 5—PH with unclear and/or multifactorial etiology.2 This group has revised the hemodynamic criteria for PH, defining it as a mPAP >20 mm Hg at rest. Pulmonary hypertension associated with lung diseases and/or hypoxia includes PH caused by chronic lung diseases such as chronic obstructive pulmonary disease (COPD), interstitial lung disease, and bronchiectasis. It also encompasses PH resulting from chronic or intermittent hypoxia induced by high-altitude environments, chronic lung diseases, or sleep-disordered breathing.
Both chronic lung diseases and high-altitude hypoxic environments contribute to the development of PH through mechanisms involving hypoxia, inflammation, and pulmonary vascular remodeling, highlighting significant similarities and crosstalk between the 2 conditions. At the 7th World Symposium on HAPH, PH associated with lung diseases and/or hypoxia was a key topic of discussion.3 Chronic lung diseases, characterized by pathological changes such as alveolar destruction, impaired airway function, pulmonary fibrosis, and extracellular matrix (ECM) remodeling, lead to persistent ventilation/perfusion mismatches and hypoxemia.4 Prolonged hypoxia activates signaling pathways such as the hypoxia-inducible factors (HIFs) pathway, disrupting inflammatory response balance, promoting immune cell infiltration (e.g., macrophages), increasing cytokine secretion, and driving abnormal proliferation of pulmonary vascular smooth muscle cells, ECs, and fibroblasts, which results in vascular wall thickening, fibrosis, and pulmonary vascular remodeling.5 High-altitude hypoxia further exacerbates these processes and is a critical factor in the initiation and progression of PH, particularly in patients with chronic lung diseases. These patients already experience varying degrees of hypoxemia due to impaired lung function and ventilation/perfusion mismatch; high-altitude hypoxia further reduces arterial oxygen partial pressure, exacerbates hypoxic pulmonary vasoconstriction (HPV), and increases pulmonary vascular resistance (PVR).6,
In addition to PH caused by lung diseases, group 3 PH also includes hypoxia-associated PH. Among these, HAPH is a significant subtype of hypoxia-associated PH, with its pathogenesis primarily linked to prolonged exposure to hypoxic conditions in high-altitude environments. Hypoxia is one of the most distinctive and extensively studied factors contributing to HAPH. Hypoxia-induced cellular responses regulate various cellular changes, including signal transduction, transcription, translation, post-translational modifications, and alterations in metabolite patterns.9 As altitude increases, atmospheric pressure decreases, leading to a reduction in the partial pressure of oxygen in the atmosphere. According to the oxygen-hemoglobin dissociation curve, the oxygen content in arterial blood significantly decreases at altitudes above 2500 meters (
Hypoxia-Inducible Factors and High-Altitude Pulmonary Hypertension
One of the primary characteristics of HAPH is exposure to low-pressure, low-oxygen environments at high altitudes, leading to insufficient oxygen supply to cells and tissues. Both acute and chronic or intermittent hypoxia can trigger a series of cellular responses and biological processes at the molecular, cellular, and tissue levels. The pathological and physiological changes caused by hypoxia can lead to pulmonary vascular remodeling and contribute to the development of PH, with these processes beginning within the first few hours of hypoxic exposure.14
Hypoxia-inducible factors play a crucial role in regulating and adapting to hypoxic environments. Cellular responses to hypoxia involve 3 transcription factors known as HIF-1α, HIF-2α, and HIF-3α. These transcription factors dimerize with constitutively expressed β-subunits to form HIF-1, HIF-2, and HIF-3. Among the 3 HIF subtypes present in mammals, HIF-1 and HIF-2 are well-studied, while research on HIF-3 is limited and its role remains unclear.15 Typically, cells respond to hypoxia by stabilizing HIFs.16
Hypoxia-inducible factor activity is mainly regulated by α-subunit expression. Under normoxic conditions, HIF-1α and HIF-2α are hydroxylated by prolyl hydroxylase domain (PHD) proteins and subsequently ubiquitinated by von Hippel–Lindau (VHL) protein, leading to rapid degradation. During hypoxia, PHD activity decreases significantly, resulting in the stabilization and accumulation of HIF-1α/2α. These stabilized HIFs then dimerize with HIF-β and translocate to the nucleus to exert physiological effects.17 Activated HIFs regulate the transcription of multiple genes and modulate cellular responses to hypoxia by inducing or inhibiting a wide range of genes involved in regulating vascular tone, cell metabolism, proliferation, survival, and autophagy.18
The sustained low-pressure and hypoxic environment at high altitudes are key factor in the development of HAPH. Exposure to high-altitude conditions results in a reduction in oxygen partial pressure, which activates the HIF signaling pathway, triggering EC injury and smooth muscle cell proliferation in the pulmonary vasculature, ultimately leading to vascular wall thickening and remodeling. Studies using rats with HAPH induced by a simulated altitude of 5000 meters show that average mPAP, right ventricular systolic pressure (RVSP), PVR, and right ventricular hypertrophy gradually alleviate as the altitude decreases.19 The aforementioned studies indicate that PH at high altitudes is reversible, and early intervention may effectively improve patients’ symptoms. Research conducted on earthquake relief in Yushu, China, indicates that residents from low-altitude areas who rapidly ascended from 1500 meters to 3700 meters showed a positive correlation between average PAP, serum HIF-1α, and vascular endothelial growth factor (VEGF). However, these parameters normalized within 15 days of returning to lower altitudes.20 Daily intermittent, non-continuous, short-term reoxygenation partially prevented HAPH induced by 5000 meters hypoxia in rats.21 This further confirms the critical role of HIFs in the development of HAPH and highlights the importance of correcting HIF-related signaling pathways, such as through oxygen therapy, as a significant approach to treating HAPH.
In PH, pulmonary arterial remodeling occurs in the 3-layered structure of the vessel wall, including the intima, media, and adventitia. This remodeling primarily involves pathological changes in PA-ECs, pulmonary arterial smooth muscle cells (PA-SMCs), pulmonary arterial fibroblasts (PA-Fibs), and the ECM.22 Existing research confirms that HIF regulation of PH is complex and multifaceted, with abnormalities in HIF signaling present in PA-ECs, PA-SMCs, and PA-Fibs (
The Role of Hypoxia-Inducible Factor in Pulmonary Arterial Endothelial Cells
Endothelial cells (ECs) play a critical role in maintaining normal cardiovascular function. Endothelial cells exhibit different phenotypes, including apoptotic and proliferative forms, which contribute differently to the development of PH. In the early stages of PH, the presence of pathogenic factors damages and induces apoptosis in the cells within the pulmonary vascular wall. Endothelial cells that survive apoptosis undergo epigenetic reprogramming, entering an abnormal proliferation phase. Proliferative ECs gradually emerge with the progression of PH and become the predominant phenotype in later stages.23
For instance, Yamaji-Kegan et al24 induced PH in mice by injecting hypoxia-induced mitotic factor (HIMF, also known as FIZZ1 or RELMα). On the seventh day, they observed a significant increase in EC apoptosis in the pulmonary vasculature of the mice.24 Early EC apoptosis can directly lead to the loss of integrity in small distal pulmonary arteries, indirectly contributing to the formation of complex and obstructive arterial lesions.25 Endothelial dysfunction results in a deficiency of endothelial-derived vasodilators, such as prostacyclin and nitric oxide (NO), and an increase in endothelial-derived vasoconstrictors, such as endothelin, which promotes the development of PH.25,
Under hypoxic conditions, HIF plays a crucial role in the development of PH by promoting the production and release of cytokines from PA-ECs. In PH mouse models, knocking down the gene encoding PHD2 (
The Role of Hypoxia-Inducible Factors in Pulmonary Arterial Smooth Muscle Cells
In PH, vascular remodeling is primarily driven by interactions among molecules and cells in the intima, media, and adventitia of the pulmonary arterial wall, as well as the surrounding vascular space. Pulmonary arterial smooth muscle cells, which are a major component of the media, contribute to pulmonary vascular remodeling through proliferation, hypertrophy, and the secretion of chemokines and cytokines, as well as through the promotion of ECM production and degradation.31
During the progression of PH, various factors can induce senescence (not apoptosis) in PA-SMCs. Senescent PA-SMCs can promote their own proliferation by increasing the expression of paracrine cytokines such as interleukin (IL)-6.32 Enhancing PA-SMC apoptosis through pharmacological intervention in rats has been shown to mitigate induced PH.33
Hypoxia-inducible factor subunits are widely expressed in various tissues and cells, including PA-SMCs, which are among the cells with high expression of HIF subunits. In the PA-SMCs of PH patients, the stability and activity of HIF subunits are increased, promoting pulmonary artery narrowing and thickening of the pulmonary arterial wall through a series of signaling pathways. Correcting HIF signaling can improve the pathological progression of PH caused by PA-SMC proliferation. Studies have shown that primary cultures of rat PA-SMCs exposed to 4% O2 for 60 hours exhibit increased expression of the hypoxia-sensitive HIF-1α subunit, HIF target genes, and HIF-1α mRNA, while the mRNA and protein expression of PHD2, which is responsible for degrading HIF-2α, is reduced.34 The increased expression of HIF-1α under hypoxic conditions significantly enhances PA-SMC proliferation, while downregulation of HIF-related subunits can inhibit PA-SMC proliferation and migration.35,
The Role of Hypoxia-Inducible Factors in Pulmonary Fibroblasts
In HAPH and COPD, PA-Fibs are activated, exhibiting excessive proliferation, increased migration, and enhanced inflammatory activity.37,
Under hypoxic conditions, the expression of HIF and its related subunits is elevated in PH, and these factors contribute to the activation of PH-Fibs, thereby facilitating the development of PH. In PH-Fibs, the expression of HIF target genes, such as
The Role of Immune Responses in Pulmonary Arterial Hypertension
The immune system plays a crucial role in maintaining tissue homeostasis and responding to infections and injuries. Imbalances between immune response defects or enhancements can lead to disease development. Under normal physiological conditions, immune cells adapt their functional characteristics according to changes in their environment, ranging from adequate oxygen supply in the circulatory system to severely hypoxic pathological sites.9 However, under pathological conditions such as inflammation, injury, infection, ischemia, and cancer, severe hypoxia alters immune cells, leading to dysregulation of immune responses and ultimately resulting in tissue damage, cancer progression, and autoimmune diseases.9
It is well-established that during immune responses to damage or pathogen infiltration, early recruitment of immune cells is mediated by chemokines and inflammatory factors produced by the local microenvironment. These cells perform functions such as phagocytosis and pathogen clearance, releasing pro-inflammatory cytokines like interferon-γ (IFN-γ), IL-1β, and IL-6 during the initial stages of inflammation. In later stages of tissue repair, immune cells primarily secrete anti-inflammatory cytokines such as arginase 1 (Arg1), transforming growth factor-β (TGF-β), and IL-10, which stimulate cell proliferation, matrix synthesis, angiogenesis, and immune regulation, thereby promoting tissue repair and reconstruction.
Immune dysfunction plays a key role in the development of various diseases, including cancer, asthma, and autoimmune disorders. Similarly, in PH, the imbalance between pro-inflammatory and anti-inflammatory immune responses has been increasingly documented. During the early stages of PH, PA-Fibs exhibit a pro-inflammatory phenotype, with increased expression of inflammatory mediators driving the recruitment of innate immune cells, and elevated levels of inflammatory mediators in the pulmonary circulation.41 The accumulation of various immune cells such as macrophages, neutrophils, dendritic cells, mast cells, T lymphocytes, and B lymphocytes around the pulmonary vasculature in PH patients has been observed.41 Additionally, inflammatory infiltration around the pulmonary vessels typically occurs prior to pulmonary vascular remodeling, indicating that maladaptive immune responses play a critical role in this remodeling process.42
Beyond the role of pro-inflammatory immune responses in PH, anti-inflammatory immune responses also significantly impact the progression of PH. Anti-inflammatory immune cells, such as M2 macrophages and regulatory T cells, contribute to PH development by secreting anti-inflammatory cytokines, suppressing pro-inflammatory immune responses, promoting cell proliferation, inhibiting apoptosis, and facilitating ECM secretion and fibrosis, all of which contribute to pulmonary vascular remodeling and PH formation.43-
Interaction Between Hypoxia and Immune Response
The objective presence of hypoxia adds complexity to the pathophysiology of HAPH. Hypoxia-related signaling, particularly involving HIFs, plays a critical role in intercellular communication. Beyond direct interactions between hypoxia and PA-ECs, PA-SMCs, and PH-Fibs that facilitate pulmonary vascular remodeling, existing research confirms that crosstalk between perivascular immune cells and hypoxia is also crucial in the development of PH.
Hypoxia-induced tissue damage and the inflammatory environment resulting from changes in microenvironmental metabolism can recruit immune cells to infiltrate affected areas. Immune cells migrate from well-oxygenated vascular systems to hypoxic, inflamed regions, where there is a sharp increase in the energy demands for metabolism and involvement in pathological processes, such as inflammatory cytokines, enzymes, and inflammatory mediators.46 Concurrently, under hypoxic conditions, inhibition of mitochondrial oxidative phosphorylation and the electron transport chain further increase reactive oxygen species (ROS) production in macrophages, thereby activating and stabilizing HIF-1α.47 Hypoxia-inducible factors serves as a key regulatory factor in adapting to these conditions, influencing the migration, antigen presentation, cytokine and antimicrobial peptide production, phagocytosis, and metabolic reprogramming of various adaptive and innate immune cells.48
In diseases characterized by choroidal neovascularization, such as age-related macular degeneration, neutrophils infiltrating around blood vessels can activate Toll-like receptor 4 to promote HIF-1α expression. The HIF-1α, in turn, regulates the expression of matrix metalloproteinase 9 (MMP9) and IL-1β, further driving inflammation and vascular formation.49
In PH, HIF stabilization promotes the formation of PH by enhancing the migration and differentiation of immune cells. In hypoxic PH, stabilization of HIF-1α can activate downstream adenosine pathways.50 Adenosine, a signaling nucleoside produced under cell damage and stress conditions, activates signaling pathways by binding to specific adenosine receptors. Notably, the adenosine axis can promote M2 macrophage polarization. The M2 macrophages possess significant anti-inflammatory and repair capabilities. In PH, the accumulation of M2 macrophages around the pulmonary arteries plays a crucial role in promoting pulmonary vascular remodeling. In HAPH, the stabilizing effects of hypoxia-induced HIF on tissue cells through cytokine secretion and immune cell recruitment contribute to vascular remodeling.51 Furthermore, HIF enhances immune cell survival by providing necessary energy through inhibiting apoptosis pathways and modulating glycolysis pathways. Early studies indicated that HIF-1α could enhance the survival of hypoxia-induced centriolar cells through NF-κB signaling.52 Research by Sormendi et al53 found that constitutive loss of PHD-2, leading to activation of HIF-2α, increased neutrophil migration in highly confined environments. Other studies have confirmed that centriolar cells, through myeloperoxidase-catalyzed ROS production and secretion of proteolytic enzymes, contribute to adverse remodeling of pulmonary arteries and surrounding tissues through abnormal protein degradation.54,
The Role of Macrophages in Pulmonary Arterial Hypertension
Macrophages are innate immune cells present in nearly all tissues and organs. In the immune response, macrophages generally originate from 2 sources. One type, the tissue-resident macrophages, derives from embryonic progenitor cells that develop into self-maintaining populations through local proliferation during embryogenesis.56 These macrophages participate in inflammation and immune responses primarily by phagocytosing pathogens, necrotic cells, cellular debris, presenting antigens, and releasing cytokines. Additionally, macrophages contribute to tissue regeneration and repair through the release of growth factors, MMPs, and anti-inflammatory cytokines.57 These precursor cells enter specific tissues, mature, and localize there to form tissue-resident macrophages, such as Kupffer cells in the liver, microglia in the central nervous system, interstitial macrophages, and alveolar macrophages in the lungs.58,
In lung tissue, there are 2 types of macrophages: alveolar macrophages and interstitial macrophages. Alveolar macrophages, originating from the fetal liver, colonize the lungs during embryonic development and maintain their population through self-renewal. They clear microorganisms and other foreign substances entering the alveoli and serve as the first line of immune defense in the lungs, playing a crucial role in pathogen phagocytosis and antigen presentation.60 On the other hand, interstitial macrophages, which exist between blood monocytes and alveolar macrophage phenotypes, have origins from both embryonic yolk sac residency and blood monocyte recruitment. They play a vital role in regulating the local microenvironment, the secretion of cytokines, and immune responses to maintain lung stability and immune balance.61,
Macrophages respond to infections, pathological damage, and other stimuli through a polarization process that results in specific functional phenotypes and immune functions. Based on their functions and activation states, macrophages are primarily classified into two subtypes: classical M1 macrophages and alternatively activated M2 macrophages.67 The M1 macrophages are primarily pro-inflammatory. When the body is in states of infection, damage, or autoimmune diseases, pathogens and inflammatory cytokines in the local microenvironment, such as bacterial lipopolysaccharides, tumor necrosis factor-alpha (TNF-α), and granulocyte-macrophage colony-stimulating factor (GM-CSF), activate macrophages to polarize into the M1 phenotype. The M1 macrophages produce inflammatory cytokines like IL-1β and TNF-α, inhibiting surrounding cell proliferation, damaging adjacent tissues, and eliminating pathogens and abnormal immune responses.67,
Macrophages ensure appropriate inflammatory and repair responses during the immune response process through polarization. They play a crucial role in controlling infections, limiting lesion spread, and reconstructing damaged tissues. Although research on macrophage polarization in HAPH is limited, studies on other types of PH and hypoxic PH, which share many similarities with HAPH, indicate that in PH animal models and patients with PH, the predominant macrophage polarization phenotype in the perivascular regions is M2. Intermittent hypoxia-induced mice show an increase in M2 macrophages.72 Additionally, transgenic male mice depleted of CD68 (M0 macrophages) exhibit reduced expression of iNOS (M1 macrophages) and increased expression of CD206+ (M2 macrophages) during hypoxia-induced PH formation.73 These studies provide a theoretical basis for understanding macrophage polarization and the mechanisms of inflammatory factor secretion in HAPH. Interestingly, in an experiment where PH was induced in rats by colchicine, macrophage polarization exhibited dynamic changes over time. Initially, M1 macrophage polarization predominates, while later, M2 macrophage polarization becomes more prevalent. It is hypothesized that M1 macrophages participate in the initial inflammatory phase by accelerating EC apoptosis, while M2 macrophages dominate during the inflammation repair phase and subsequent abnormal tissue remodeling by promoting the proliferation of smooth muscle cells and ECs.74
The Role of Macrophage-Related Cytokines in Pulmonary Arterial Hypertension
The M2 macrophages exert their biological functions through anti-inflammatory responses, foreign body clearance, and tissue repair and regeneration. These functions are primarily driven by signaling molecules such as chemokines, cytokines, and growth factors. Therefore, exploring the molecular mechanisms of M2 macrophage recruitment and polarization, as well as their biological functions, provides a theoretical basis for treating PH. Previous research in fields such as breast cancer,75 ankylosing spondylitis,76 pulmonary fibrosis,77 and tissue repair78 has shown that IL-4 and IL-13 promote macrophage polarization towards the M2 phenotype mainly through the phosphorylation of downstream STAT6, which is a primary pathway for macrophage alternative activation. However, studies investigating IL-4 and STAT6 in macrophage polarization in PH are limited, and results vary depending on the type of PH. In vitro studies have demonstrated that IL-4 stimulation of human PA-ECs enhances the expression of CXCL-8 mRNA and protein, promoting neutrophil recruitment to the lung tissue, indicating that IL-4 aids in immune cell recruitment.79 Murine models of PH induced by Schistosoma mansoni and asthma, which exhibit elevated IL-4 and IL-13 levels, suggest a potential association with Type II hypersensitivity.80,
Chemokines are small molecular proteins with a conserved secondary structure, consisting of a flexible N-terminus, three anti-parallel β-folds, and a C-terminal α-helix.84 They play significant roles in regulating macrophage migration and polarization. Chemokines are categorized into 4 families based on their N-terminal cysteine residues: CXC (α-chemokines), CC (β-chemokines), XC (γ-chemokines), and CX3C (δ-chemokines).85 Dysregulation of chemokines and their receptors in HAPH has been increasingly studied, showing that M2 macrophage polarization is predominantly observed. Acute exposure to 3400 meters altitude leads to elevated levels of macrophage inflammatory protein-1α (MIP-1α), monocyte chemoattractant protein-1 (MCP-1), and IL-8 in the blood, with MIP-1α and MCP-1 further recruiting monocytes.86 In hypoxia-induced PH mouse models, the CCL2-CCR2, CCL5-CCR5, and CX3CL1/CX3CR1 pathways stimulate M2 macrophage increase and PA-SMCs proliferation, promoting PH formation; inhibition of these pathways can prevent or reverse PH.87 The CX3CR1 knockout mice and those treated with the CX3CR1 inhibitor F1 show reduced M2 macrophages, lower RVSP, reduced right ventricle-to-left ventricle plus septum (RV/LV+S) ratios, and alleviated pulmonary vascular remodeling, validating CX3CR1’s role in macrophage polarization and recruitment.66
Transforming growth factor-β, produced by tissue and immune cells, plays a critical role in tissue repair and healing. There is a close relationship between M2 macrophages and TGF-β in pulmonary tissue. The M2 macrophages secrete large amounts of TGF-β, contributing to pathological responses, while TGF-β drives macrophages towards the M2 phenotype.88,
Cytokines are secreted proteins that regulate cell proliferation, death, activation, or inhibition through autocrine, paracrine, and endocrine signaling.101 They play crucial roles in intercellular communication and cell function regulation. Numerous studies have confirmed that cytokines drive M2 macrophage polarization, enhancing the anti-inflammatory response and contributing to pulmonary arterial remodeling and PH progression. In hypoxia-induced PH mice, the IL-6/IL-21 signaling axis promotes PH through M2 macrophage polarization. Blocking IL-6 with the monoclonal antibody MR16-1 significantly reduces downstream IL-21 expression, and IL-21 treatment of alveolar macrophages from bronchoalveolar lavage fluid upregulates M2-related gene mRNA levels (such as Fizz1, Arg1, and CXCL12).102 Additionally, anti-inflammatory cytokines like IL-10 and IL-13 are elevated in PH patients and animal models.82,
Arginine metabolism plays a critical role in the development and progression of PH. Arginine can be hydrolyzed by arginase to produce ornithine or oxidized by NOS to produce NO.104 The NO is a vasodilator that counteracts vascular remodeling in PH by dilating blood vessels and inhibiting cell proliferation. Under pathological hypoxia, excessive arginase activity hydrolyzes arginine, leading to excessive ornithine production and insufficient NO production.105,
Prospect
In recent years, with the improvement in living standards and advancements in science and technology, there has been a notable increase in the number of people traveling to high-altitude areas for work, exploration, and tourism. The various challenges posed by acute and chronic mountain sickness have become increasingly apparent, significantly impeding these activities. Research into acute and chronic mountain sickness has also become more thorough and comprehensive. With a deeper understanding of the mechanisms underlying HAPH, and by drawing parallels with the pathophysiological mechanisms of other types of PH, we can more precisely develop prevention and treatment strategies for HAPH.
Given the critical role of macrophages in HAPH, designing specific drugs and employing gene technology to regulate abnormal macrophage polarization, mitigate inflammatory responses, and prevent pulmonary vascular remodeling represents significant breakthrough directions. Additionally, beyond the primary factors of high altitude, low pressure, and hypoxia, genetic susceptibility, immune abnormalities, and neurological dysfunctions also play crucial pathogenic roles. Conducting further genetic research and developing personalized treatment strategies through molecular biology and genomics can enhance our understanding of individual patient differences, offering more possibilities for personalized treatments.
Currently, to address the impact of high-altitude hypoxia on HAPH, advanced oxygen therapy technologies, bloodletting, and climate adjustment methods can be introduced to alleviate or prevent the onset of HAPH in high-altitude regions.
Footnotes
No datasets were generated or analyzed during the current study.
The authors would like to acknowledge the assistance of ChatGPT for its support in language refinement and linguistic clarification of the manuscript.
References
- León-Velarde F, Maggiorini M, Reeves JT. Consensus statement on chronic and subacute high altitude diseases. High Alt Med Biol. 2005;6(2):147-157.
- Humbert M, Kovacs G, Hoeper MM. 2022 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension. Eur Heart J. 2022;43(38):3618-3731.
- Humbert M, Galiè N, Rubin LJ, Simonneau G, McLaughlin VV. The Seventh World Symposium on Pulmonary Hypertension: our journey to Barcelona. Eur Respir J. 2024;64(4):2401222-.
- Burgess JK, Harmsen MC. Chronic lung diseases: entangled in extracellular matrix. Eur Respir Rev. 2022;31(163):210202-.
- Ogger PP, Byrne AJ. Macrophage metabolic reprogramming during chronic lung disease. Mucosal Immunol. 2021;14(2):282-295.
- Zhang Q, Liu H, Liu C. Tibetan mesenchymal stem cell-derived exosomes alleviate pulmonary vascular remodeling in hypoxic pulmonary hypertension rats. Stem Cells (Dayt Ohio). 2024;42(8):720-735.
- Zhang R, Su H, Ma X. MiRNA let-7b promotes the development of hypoxic pulmonary hypertension by targeting ACE2. Am J Physiol Lung Cell Mol Physiol. 2019;316(3):L547-L557.
- Liu H, Wang Y, Zhang Q. Macrophage-derived inflammation promotes pulmonary vascular remodeling in hypoxia-induced pulmonary arterial hypertension mice. Immunol Lett. 2023;263():113-122.
- Chen Y, Gaber T. Hypoxia/HIF modulates immune responses. Biomedicines. 2021;9(3):260-.
- Moore LG. Measuring high-altitude adaptation. J Appl Physiol (1985). 2017;123(5):1371-1385.
- Böger R, Hannemann J. Dual role of the L-arginine-ADMA-NO pathway in systemic hypoxic vasodilation and pulmonary hypoxic vasoconstriction. Pulm Circ. 2020;10(2):2045894020918850-.
- El Alam S, Pena E, Aguilera D, Siques P, Brito J. Inflammation in pulmonary hypertension and edema induced by hypobaric hypoxia exposure. Int J Mol Sci. 2022;23(20):12656-.
- Dunham-Snary KJ, Wu D, Sykes EA. Hypoxic pulmonary vasoconstriction: from molecular mechanisms to medicine. Chest. 2017;151(1):181-192.
- He S, Zhu T, Fang Z. The role and regulation of pulmonary artery smooth muscle cells in pulmonary hypertension. Int J Hypertens. 2020;2020():1478291-.
- Jaskiewicz M, Moszynska A, Serocki M. Hypoxia-inducible factor (HIF)-3a2 serves as an endothelial cell fate executor during chronic hypoxia. Excli J. 2022;21():454-469.
- Pressley M, Gallaher JA, Brown JS. Cycling hypoxia selects for constitutive HIF stabilization. Sci Rep. 2021;11(1):5777-.
- Shimoda LA. What’s HIF got to do with it? HIF-2 inhibition and pulmonary hypertension. Am J Respir Crit Care Med. 2018;198(11):1363-1365.
- Brahimi-Horn MC, Pouysségur J. HIF at a glance. J Cell Sci. 2009;122(Pt ):1055-1057.
- Shi H, Zhao Y, Li S, Wu H, Ma D, Wan C. TNF-α and IL-8 levels are positively correlated with hypobaric hypoxic pulmonary hypertension and pulmonary vascular remodeling in rats. Open Life Sci. 2023;18(1):20220650-.
- Yang SY, Feng EZ, Yan ZQ. The role of hypoxia inducible factor-1α and vascular endothelial growth factor in hypoxic pulmonary hypertension in patients with acute high altitude reaction of rescue workers in Yushu earthquake. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2011;23(9):539-542.
- Lyu Q, Bai Y, Cheng J. Intermittent short-duration reoxygenation protects against simulated high altitude-induced pulmonary hypertension in rats. FASEB J. 2021;35(2):e21212-.
- Tuder RM, Marecki JC, Richter A, Fijalkowska I, Flores S. Pathology of pulmonary hypertension. Clin Chest Med. 2007;28(1):23-42.
- Sakao S, Tatsumi K, Voelkel NF. Endothelial cells and pulmonary arterial hypertension: apoptosis, proliferation, interaction and transdifferentiation. Respir Res. 2009;10(1):95-.
- Yamaji-Kegan K, Takimoto E, Zhang A. Hypoxia-induced mitogenic factor (FIZZ1/RELMα) induces endothelial cell apoptosis and subsequent interleukin-4-dependent pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2014;306(12):L1090-L1103.
- Cober ND, VandenBroek MM, Ormiston ML, Stewart DJ. Evolving concepts in endothelial pathobiology of pulmonary arterial hypertension. Hypertension. 2022;79(8):1580-1590.
- Budhiraja R, Tuder RM, Hassoun PM. Endothelial dysfunction in pulmonary hypertension. Circulation. 2004;109(2):159-165.
- Dai Z, Li M, Wharton J, Zhu MM, Zhao YY. Prolyl-4 hydroxylase 2 (PHD2) deficiency in endothelial cells and hematopoietic cells induces obliterative vascular remodeling and severe pulmonary arterial hypertension in mice and humans through hypoxia-inducible Factor-2α. Circulation. 2016;133(24):2447-2458.
- Hu CJ, Poth JM, Zhang H. Suppression of HIF2 signalling attenuates the initiation of hypoxia-induced pulmonary hypertension. Eur Respir J. 2019;54(6):1900378-.
- Cowburn AS, Crosby A, Macias D. HIF2α-arginase axis is essential for the development of pulmonary hypertension. Proc Natl Acad Sci U S A. 2016;113(31):8801-8806.
- Dai Z, Zhu MM, Peng Y. Therapeutic targeting of vascular remodeling and right heart failure in pulmonary arterial hypertension with a HIF-2α inhibitor. Am J Respir Crit Care Med. 2018;198(11):1423-1434.
- Stenmark KR, Frid MG, Graham BB, Tuder RM. Dynamic and diverse changes in the functional properties of vascular smooth muscle cells in pulmonary hypertension. Cardiovasc Res. 2018;114(4):551-564.
- Wang AP, Yang F, Tian Y. Pulmonary artery smooth muscle cell senescence promotes the proliferation of PASMCs by paracrine IL-6 in Hypoxia-Induced Pulmonary Hypertension. Front Physiol. 2021;12():656139-.
- Zhu L, Li YL, Qian ZQ, Hua L, Yue Y, Yang DL. Osthole improves pulmonary artery hypertension by inducing apoptosis in pulmonary artery smooth muscle cells. J Pharm Pharmacol. 2021;73(8):1109-1117.
- Pisarcik S, Maylor J, Lu W. Activation of hypoxia-inducible factor-1 in pulmonary arterial smooth muscle cells by endothelin-1. Am J Physiol Lung Cell Mol Physiol. 2013;304(8):L549-L561.
- Chen M, Shen C, Zhang Y, Shu H. MicroRNA-150 attenuates hypoxia-induced excessive proliferation and migration of pulmonary arterial smooth muscle cells through reducing HIF-1α expression. Biomed Pharmacother. 2017;93():861-868.
- Deng J, Qin J, Cai Y, Zhong X, Zhang X, Yu S. Rutaecarpine suppresses proliferation and promotes apoptosis of human pulmonary artery smooth muscle cells in hypoxia possibly through HIF-1α-dependent pathways. J Cardiovasc Pharmacol. 2018;71(5):293-302.
- Luo Y, Dong HY, Zhang B. miR-29a-3p attenuates hypoxic pulmonary hypertension by inhibiting pulmonary adventitial fibroblast activation. Hypertension. 2015;65(2):414-420.
- Zhang S, Yin Z, Qin W. Pirfenidone inhibits hypoxic pulmonary hypertension through the NADPH/ROS/p38 pathway in adventitial fibroblasts in the pulmonary artery. Mediators Inflamm. 2020;2020():2604967-.
- Hu CJ, Laux A, Gandjeva A. The effect of hypoxia-inducible factor inhibition on the phenotype of fibroblasts in human and bovine pulmonary hypertension. Am J Respir Cell Mol Biol. 2023;69(1):73-86.
- Lv XM, Li MD, Cheng S. Neotuberostemonine inhibits the differentiation of lung fibroblasts into myofibroblasts in mice by regulating HIF-1α signaling. Acta Pharmacol Sin. 2018;39(9):1501-1512.
- Hu Y, Chi L, Kuebler WM, Goldenberg NM. Perivascular inflammation in pulmonary arterial hypertension. Cells. 2020;9(11):2338-.
- Perros F, Dorfmüller P, Souza R. Dendritic cell recruitment in lesions of human and experimental pulmonary hypertension. Eur Respir J. 2007;29(3):462-468.
- Qiu H, Zhang Y, Li Z. Donepezil ameliorates pulmonary arterial hypertension by inhibiting M2-macrophage activation. Front Cardiovasc Med. 2021;8():639541-.
- Fernandez-Gonzalez A, Mukhia A, Nadkarni J. Immunoregulatory macrophages modify local pulmonary immunity and ameliorate hypoxic-pulmonary. bioRxiv. 2023;():2023.07.31.551394-.
- Sada Y, Dohi Y, Uga S, Higashi A, Kinoshita H, Kihara Y. Non-suppressive regulatory T cell subset expansion in pulmonary arterial hypertension. Heart Vessels. 2016;31(8):1319-1326.
- Malkov MI, Lee CT, Taylor CT. Regulation of the hypoxia-inducible factor (HIF) by pro-inflammatory cytokines. Cells. 2021;10(9):2340-.
- Canton M, Sánchez-Rodríguez R, Spera I. Reactive oxygen species in macrophages: sources and targets. Front Immunol. 2021;12():734229-.
- Krzywinska E, Stockmann C. Hypoxia, metabolism and immune cell function. Biomedicines. 2018;6(2):56-.
- Zeng J, Wang Y, Zhu M. Neutrophil extracellular traps boost laser-induced mouse choroidal neovascularization through the activation of the choroidal endothelial cell TLR4/HIF-1α pathway. FEBS Journal. 2023;290(22):5395-5410.
- Garcia-Morales LJ, Chen NY, Weng T. Altered hypoxic-adenosine axis and metabolism in Group III pulmonary hypertension. Am J Respir Cell Mol Biol. 2016;54(4):574-583.
- Ye Y, Xu Q, Wuren T. Inflammation and immunity in the pathogenesis of hypoxic pulmonary hypertension. Front Immunol. 2023;14():1162556-.
- Walmsley SR, Print C, Farahi N. Hypoxia-induced neutrophil survival is mediated by HIF-1alpha-dependent NF-kappaB activity. J Exp Med. 2005;201(1):105-115.
- Sormendi S, Deygas M, Sinha A. HIF2α is a direct regulator of neutrophil motility. Blood. 2021;137(24):3416-3427.
- Klinke A, Berghausen E, Friedrichs K. Myeloperoxidase aggravates pulmonary arterial hypertension by activation of vascular Rho-kinase. JCI Insight. 2018;3(11):e97530-.
- Taylor S, Dirir O, Zamanian RT, Rabinovitch M, Thompson AAR. The role of neutrophils and neutrophil elastase in pulmonary arterial hypertension. Front Med (Lausanne). 2018;5():217-.
- Xu Y, Schrank PR, Williams JW. Macrophage fate mapping. Curr Protoc. 2022;2(6):e456-.
- Lampiasi N. Macrophage polarization: learning to manage it. Int J Mol Sci. 2022;23(13):7208-.
- Lazarov T, Juarez-Carreño S, Cox N, Geissmann F. Physiology and diseases of tissue-resident macrophages. Nature. 2023;618(7966):698-707.
- Zhao Y, Zou W, Du J, Zhao Y. The origins and homeostasis of monocytes and tissue-resident macrophages in physiological situation. J Cell Physiol. 2018;233(10):6425-6439.
- Joshi N, Walter JM, Misharin AV. Alveolar macrophages. Cell Immunol. 2018;330():86-90.
- Tan SYS, Krasnow MA. Developmental origin of lung macrophage diversity. Development. 2016;143(8):1318-1327.
- Schyns J, Bureau F, Marichal T. Lung interstitial macrophages: past, present, and future. J Immunol Res. 2018;2018():5160794-.
- Rabinovitch M, Guignabert C, Humbert M, Nicolls MR. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res. 2014;115(1):165-175.
- Kojima H, Tokunou T, Takahara Y. Hypoxia-inducible factor-1 α deletion in myeloid lineage attenuates hypoxia-induced pulmonary hypertension. Physiol Rep. 2019;7(7):e14025-.
- Pugliese SC, Poth JM, Fini MA, Olschewski A, El Kasmi KC, Stenmark KR. The role of inflammation in hypoxic pulmonary hypertension: from cellular mechanisms to clinical phenotypes. Am J Physiol Lung Cell Mol Physiol. 2015;308(3):L229-L252.
- Amsellem V, Abid S, Poupel L. Roles for the CX3CL1/CX3CR1 and CCL2/CCR2 chemokine systems in hypoxic pulmonary hypertension. Am J Respir Cell Mol Biol. 2017;56(5):597-608.
- Kadomoto S, Izumi K, Mizokami A. Macrophage polarity and disease control. Int J Mol Sci. 2021;23(1):144-.
- Mertens C, Marques O, Horvat NK, Simonetti M, Muckenthaler MU, Jung M. The macrophage iron signature in health and disease. Int J Mol Sci. 2021;22(16):8457-.
- Loegl J, Hiden U, Nussbaumer E. Hofbauer cells of M2a, M2b and M2c polarization may regulate feto-placental angiogenesis. Reprod (Camb Engl). 2016;152(5):447-455.
- Tang L, Zhang H, Wang C, Li H, Zhang Q, Bai J. M2A and M2C macrophage subsets ameliorate inflammation and fibroproliferation in acute lung injury through interleukin 10 pathway. Shock. 2017;48(1):119-129.
- Ahamada MM, Jia Y, Wu X. Macrophage polarization and plasticity in systemic lupus erythematosus. Front Immunol. 2021;12():734008-.
- Mao X, Li Y, Yang R. Single-cell RNA-sequencing reveals the active involvement of macrophage polarizations in pulmonary hypertension. Dis Markers. 2022;2022():5398157-.
- Zawia A, Arnold ND, West L. Altered macrophage polarization induces experimental pulmonary hypertension and is observed in patients with pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol. 2021;41(1):430-445.
- Fan Y, Hao Y, Gao D, Li G, Zhang Z. Phenotype and function of macrophage polarization in monocrotaline-induced pulmonary arterial hypertension rat model. Physiol Res. 2021;70(2):213-226.
- Rahal OM, Wolfe AR, Mandal PK. Blocking interleukin (IL)4- and IL13-mediated phosphorylation of STAT6 (Tyr641) decreases M2 polarization of macrophages and protects against macrophage-mediated radioresistance of inflammatory breast cancer. Int J Radiat Oncol Biol Phys. 2018;100(4):1034-1043.
- Lin S, Qiu M, Chen J. IL-4 modulates macrophage polarization in ankylosing spondylitis. Cell Physiol Biochem. 2015;35(6):2213-2222.
- Guo X, Li T, Xu Y. Increased levels of Gab1 and Gab2 adaptor proteins skew interleukin-4 (IL-4) signaling toward M2 macrophage-driven pulmonary fibrosis in mice. J Biol Chem. 2017;292(34):14003-14015.
- Chandran S, Schilke RM, Blackburn CMR. Lipin-1 contributes to IL-4 mediated macrophage polarization. Front Immunol. 2020;11():787-.
- Yang DF, Huang H, Guan S. Interleukin(IL)-4 promotion of CXCL-8 gene transcription is mediated by ERK1/2 pathway in human pulmonary artery endothelial cells. Mol Immunol. 2011;48(15-16):1784-1792.
- Kumar R, Mickael C, Chabon J. The causal role of IL-4 and IL-13 in Schistosoma mansoni pulmonary hypertension. Am J Respir Crit Care Med. 2015;192(8):998-1008.
- Li S, Ma X, Xie J, Yan X, Sun W. MicroRNA-206, IL-4, IL-13, and INF-γ levels in lung tissue and plasma are increased by the stimulation of particulate matter with a diameter of ≤2.5μm, and are associated with the poor prognosis of asthma induced pulmonary arterial hypertension patients. Clin Exp Hypertens. 2021;43(2):181-188.
- Tomaszewski M, Mertowska P, Janczewska M. In the search for biomarkers of pulmonary arterial hypertension, are cytokines IL-2, IL-4, IL-6, IL-10, and IFN-gamma the right indicators to use?. Int J Mol Sci. 2023;24(18):13694-.
- Harbaum L, Renk E, Yousef S. Acute effects of exercise on the inflammatory state in patients with idiopathic pulmonary arterial hypertension. BMC Pulm Med. 2016;16(1):145-.
- Luo H, Li L, Han S, Liu T. The role of monocyte/macrophage chemokines in pathogenesis of osteoarthritis: a review. Int J Immunogenet. 2024;51(3):130-142.
- Mamazhakypov A, Viswanathan G, Lawrie A, Schermuly RT, Rajagopal S. The role of chemokines and chemokine receptors in pulmonary arterial hypertension. Br J Pharmacol. 2021;178(1):72-89.
- Mishra KP, Sharma N, Soree P, Gupta RK, Ganju L, Singh SB. Hypoxia-induced inflammatory chemokines in subjects with a history of high-altitude pulmonary edema. Indian J Clin Biochem. 2016;31(1):81-86.
- Abid S, Marcos E, Parpaleix A. CCR2/CCR5-mediated macrophage-smooth muscle cell crosstalk in pulmonary hypertension. Eur Respir J. 2019;54(4):1802308-.
- Zhu L, Fu X, Chen X, Han X, Dong P. M2 macrophages induce EMT through the TGF-β/Smad2 signaling pathway. Cell Biol Int. 2017;41(9):960-968.
- Deng S, Jin P, Liu S. Recruitment of regulatory T cells with rCCL17 promotes M2 microglia/macrophage polarization through TGFβ/TGFβR/Smad2/3 pathway in a mouse model of intracerebral hemorrhage. Exp Neurol. 2023;367():114451-.
- Massagué J, Sheppard D. TGF-β signaling in health and disease. Cell. 2023;186(19):4007-4037.
- Andre P, Joshi SR, Briscoe SD, Alexander MJ, Li G, Kumar R. Therapeutic approaches for treating pulmonary arterial hypertension by correcting imbalanced TGF-beta superfamily signaling. Front Med (Lausanne). 2021;8():814222-.
- Calvier L, Chouvarine P, Legchenko E, Kokeny G, Mozes MM, Hansmann G. Chronic TGF-β1 signaling in pulmonary arterial hypertension induces sustained canonical Smad3 pathways in vascular smooth muscle cells. Am J Respir Cell Mol Biol. 2019;61(1):121-123.
- Yung LM, Yang P, Joshi S. ACTRIIA-Fc rebalances activin/GDF versus BMP signaling in pulmonary hypertension. Sci Transl Med. 2020;12(543):eaaz5660-.
- Wang J, Feng W, Li F. SphK1/S1P mediates TGF-β1-induced proliferation of pulmonary artery smooth muscle cells and its potential mechanisms. Pulm Circ. 2019;9(1):2045894018816977-.
- Sun X, Lu Q, Yegambaram M. TGF-β1 attenuates mitochondrial bioenergetics in pulmonary arterial endothelial cells via the disruption of carnitine homeostasis. Redox Biol. 2020;36():101593-.
- Li Z, Wang F, Zha S, Cao Q, Sheng J, Chen S. SIRT1 inhibits TGF-β-induced endothelial-mesenchymal transition in human endothelial cells with Smad4 deacetylation. J Cell Physiol. 2018;233(11):9007-9014.
- Gorelova A, Berman M, Al Ghouleh I. Endothelial-to-mesenchymal transition in pulmonary arterial hypertension. Antioxid Redox Signal. 2021;34(12):891-914.
- Erewele EO, Castellon M, Loya O. Hypoxia-induced pulmonary hypertension upregulates eNOS and TGF-β contributing to sex-linked differences in BMPR2 (+/R899X) mutant mice. Pulm Circ. 2022;12(4):e12163-.
- Yuan W, Liu W, Cai H. SB-431542, a specific inhibitor of the TGF-β type I receptor inhibits hypoxia-induced proliferation of pulmonary artery adventitial fibroblasts. Pharmazie. 2016;71(2):94-100.
- Zhang F, Wang H, Wang X. TGF-β induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget. 2016;7(32):52294-52306.
- Li AW, Lim WA. Engineering cytokines and cytokine circuits. Science. 2020;370(6520):1034-1035.
- Hashimoto-Kataoka T, Hosen N, Sonobe T. Interleukin-6/interleukin-21 signaling axis is critical in the pathogenesis of pulmonary arterial hypertension. Proc Natl Acad Sci U S A. 2015;112(20):E2677-E2686.
- Cho WK, Lee CM, Kang MJ. IL-13 receptor α2-arginase 2 pathway mediates IL-13-induced pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2013;304(2):L112-L124.
- Gao X, Zhang Z, Li X. Macitentan attenuates chronic mountain sickness in rats by regulating arginine and purine metabolism. J Proteome Res. 2020;19(8):3302-3314.
- Zheng HK, Zhao JH, Yan Y. Metabolic reprogramming of the urea cycle pathway in experimental pulmonary arterial hypertension rats induced by monocrotaline. Respir Res. 2018;19(1):94-.
- Chu Y, XiangLi X, Niu H. Arginase inhibitor attenuates pulmonary artery hypertension induced by hypoxia. Mol Cell Biochem. 2016;412(1-2):91-99.
- Zhang L, Liu X, Wei Q. Arginine attenuates chronic mountain sickness in rats via microRNA-144-5p. Mamm Genome. 2023;34(1):76-89.
- Ahmadipour B, Sharifi M, Khajali F. Pulmonary hypertensive response of broiler chickens to arginine and guanidinoacetic acid under high-altitude hypoxia. Acta Vet Hung. 2018;66(1):116-124.