Role of Hypoxia-Inducible Factor 1α in Adaptation to Hypoxia in the Pathogenesis of Novel Coronavirus Disease 2019

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

INTRODUCTION: A novel coronavirus (severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2)) emerged in December 2019 and rapidly spread over the world having provoked a pandemic of respiratory disease. This highly pathogenic virus can attack the lung tissue and derange gas exchange leading to acute respiratory distress syndrome and systemic hypoxia. Hypoxic conditions trigger activation of adaptation mechanisms including hypoxia-inducible factor-1á (HIF-1á) which is involved in the regulation of the key processes, e. g, proliferation and metabolism of cells and angiogenesis. Besides, the level of HIF-1á expression is associated with the intensity of the immune response of an organism including that of the innate immunity mediating inflammatory reaction. Therefore, understanding the peculiarities of the mechanisms underlying the pathogenesis of this disease is of great importance for effective therapy of coronavirus disease 2019 (COVID-19).

AIM: Analysis of the current data on HIF-1á and its effect on the pathogenesis and progression of COVID-19.

The analysis of the relevant domestic and international literature sources was performed in the following sections: HIF-1á as a key factor of adaptation to hypoxia, targets for HIF-1á in the aspect of the pathogenesis of COVID-19, disorders in HIF-1á-mediated adaptation to hypoxia as an element of the pathogenesis of hyperactivation of the immune cells.

CONCLUSION: HIF-1á prevents penetration of SARS-CoV-2 virus into a cell and primarily acts as the main regulator of the proinflammatory activity at the inflammation site surrounded by hypoxia. In the conditions of the deranged metabolic flexibility, a high level of HIF-1á evokes an excessive inflammatory response of the immune cells. A high HIF-1á level in cells of the inflammation focus is associated with enhanced production of the factors of angiogenesis mediating vascular permeability and capillary leakage process. This is accompanied by tissue damage and organ failure. At the same time, HIF-1á can mediate the anti-inflammatory effect through activation of adenosine receptor-dependent pathway, which is considered as a probable protection of cells and organs against damage by hyperactive immune cells.

Full Text

INTRODUCTION

In December 2019, an outburst of a deadly disease caused by a new coronavirus 2 associated with severe acute respiratory syndrome (SARS-CoV-2) was recorded in Wuhan (province Hubei, China) [1]. Subsequently, the disease was named Coronavirus Disease 2019 (COVID-19). SARS-CoV-2 is a highly contagious (index 1.4–2.5 [2]) and highly pathogenic infectious agent spread through airborne droplets in direct contact with infected patients and is potentially transmitted through fecal-oral way as well [3]. Infection with SARS-CoV-2 causes acute respiratory disease, with the common symptoms as fever, cough, shortness of breath and increased fatigue. The disease is associated with the threatening complications such as pneumonitis, acute respiratory distress-syndrome (ARDS), respiratory failure, shock, multiorgan failure [4].

An important pathological feature characteristic of patients with moderate, severe and extremely severe form of COVID-19 infection is a progressing systemic hypoxia [3].

Hypoxia is a stimulus that launches adaptation mechanisms associated with changes in cell metabolism, organ perfusion, and erythropoiesis and iron metabolism [5]. Hypoxia-inducible transcription factor 1á (HIF-1á) is considered nowadays to be a sensor that perceives hypoxia at the cell level [6]. HIF-1á permits cells to survive in severe hypoxic conditions, however, in case of extremely severe hypoxia as observed in a severe course of COVID-19 infection, HIF-1á can convert from a factor of adaptation into a factor of maladaptation: it is established that its excessive accumulation in cells of the innate immune system slows down their natural clearance and mediates the uncontrolled synthesis of proinflammatory cytokines [3, 7].

The activity of HIF-1á is known to mediate the activation of proangiogenic and proinflammatory factors, which is associated with diffuse damage into the alveolar-capillary barrier characteristic of ARDS in COVID-19 infection [8].

ARDS is an acute diffuse inflammatory lesion of the lung parenchyma that develops as a non-specific reaction to various damaging factors leading to acute respiratory failure due to damage to the lung tissue and reduction in the mass of airy lung tissue [9].

A subgroup of patients with COVID-19 and ARDS infection is characterized by overfilling of the pulmonary arteries with venous blood, and by severe hypoxemia, significant hemocoagulation disorders and rapid development of shock [10]. This atypical form of ARDS is characterized by a significant pulmonary shunt and abnormal respiratory mechanics with the underlying hypoxic pulmonary vasoconstriction and hypercoagulation associated with local microvascular thrombosis in the pulmonary circulation and with the addition of microvascular lesions of the kidneys, brain and other vital organs [11].

Despite the steps undertaken to treat respiratory failure, more than 80% of patients with severe COVID-19 infection who received ventilation die by day 28 from the onset of the disease [12]. The high mortality rate of patients and the atypical course of ARDS in patients with COVID-19 infection provide the ground to suggest that COVID-19 infection creates preconditions for dysfunction of the body's adaptation mechanisms to hypoxic conditions [13].

The aim of this study to analyze the current data on the hypoxia-inducible factor 1á and its effect on the pathogenesis and progression of coronavirus disease 2019.

HIF-1á as Key Factor in Adaptation to Hypoxia

HIF-1á is a conservative heterodimeric transcription factor that allows the body to adapt to changes in the concentration and availability of oxygen in the blood [6]. The HIF-1 heterodimer comprises an oxygen-dependent HIF-1á subunit and a constitutively expressed HIF-1â subunit [14]. The HIF-1â subunit is continuously expressed in the cell, regardless of the oxygen level in the cell. The expression of HIF-1á gradually increases under hypoxic conditions. There exist 3 isoforms of HIF-á possessing specific properties: HIF-1á, HIF-2á and HIF-3á. Isoforms of HIF-á subunits regulate 5 groups of genes (Table. 1) [15, 16].

 

Таблица 1. Изоформы фактора, индуцируемого гипоксией, 1á [15, 16]

Isoform

Group of Genes

HIF-1α

Genes of glycolysis enzymes — phosphofructokinase, phosphoglycerate kinase and lactate dehydrogenase

HIF-2α

Genes of TGF-á and erythropoietin

HIF-3α

sqrdl, zp3v2 genes, whose products are involved in the regulation of metabolism and embryonic development

HIF-1α and HIF-2α

VEGF, IL-6 and GLUT1 genes

HIF-1α and HIF-3α

REDD1 gene which product is involved in the stimulation of autophagy in hypoxia

Notes: IL — interleukin, GLUT — glucose transporter (unidirectional glucose transporter protein), REDD — gene regulated during development and reaction to DNA damage, TGF — transforming growth factor, VEGF — vascular endothelial growth factor

 

The expression of α-subunits of the transcription factor enhances in response to both acute and chronic hypoxia [16]. HIF-1á is most active in conditions of acute, short-term (2–24 hours) and severe hypoxia or anoxia. HIF-2á isoform is responsible for adaptation to moderate chronic hypoxia [14]. HIF-3á is assumed to be a negative regulator of HIF-1á and HIF-2á activity due to competition for binding to HIF-1â subunits [15]. At the same time, the biological role of HIF-3á remains poorly understood and continues to be actively investigated. Thus, the ability of HIF-3á subunit to stabilize upon selective binding to oleoylethanolamide has recently been demonstrated, which, according to researchers, points to the function of HIF-3á as a sensor of endogenous lipids [17].

Under normoxic conditions, HIF-1á interacts with and binds to the von Hippel–Lindau (VHL) protein, which activates the ubiquitin ligase system and leads to proteasomal degradation of HIF-1á. Hydroxylation of prolin residues in HIF-1á is vital for VHL protein binding and depends on á-ketoglutarate-dependent dioxygenases, prolyl hydroxylases (PHD) and asparaginyl hydroxylase, a factor inhibiting HIF (FIH) [14]. Under hypoxic conditions, prolyl hydroxylases are inhibited, which leads to stabilization of HIF-1á and dimerization with HIF-1â. After dimerization, HIF moves to the nucleus to bind to hypoxia response elements (HREs) in the promoter region with the participation of co-activators of CREB-binding protein (cyclic AMP response element binding protein, CBP). At the same time, HIF activates genes that control cell oxygen homeostasis, including genes involved in oxygen consumption, erythrocyte production, angiogenesis, iron metabolism and mitochondrial metabolism [18]. It has been established that activation of these genes is associated with changes in the metabolism of a wide range of cells and their proliferation, vasomotor control and immune regulation [18, 19] (Figure 1).

 

Fig. 1. Regulation of the hypoxia-inducible factor, 1α [18, 19]. Notes: ROS — reactive oxygen species, TAC– tricarbonic acid cycle, RNA — ribonucleic acid, CBP — cyclic AMP response element binding
protein (co-activators of CREB-binding protein), FIH — factor inhibiting HIF; GLUT — glucose transporter, HIF — hypoxia-inducible factor, HK — hexokinase, HREs — hypoxia response elements, LDHA — lactate dehydrogenase A, PDK — pyruvate dehydrogenase kinase, VEGF — vascular endothelial growth factor, VHL — von Hippel–Lindau suppressor protein.

 

Prolyl hydroxylation of HIF-1á is associated with the utilization of á-ketoglutarate (a NAD-dependent substrate of the tricarbonic acid cycle (TAC)), while another TAC substrate — succinate, is an allosteric inhibitor of this process [20]. In normoxia, succinate is converted to fumarate with the participation of succinate dehydrogenase, which is involved in both the TAC and the electron transfer chain in mitochondria. The loss of succinate dehydrogenase activity creates preconditions for the accumulation of succinate in cells. Therefore, elevated levels of cellular succinate are linked with dysfunction of the electron transport chain complex II associated with increased production of reactive oxygen species (ROS) [21], dysregulation of proliferation and migration genes [22], posttranslational modification of proteins by the succinylation type [23].

On the other hand, a number of researchers have demonstrated an important role of succinate in the mechanisms of adaptation to hypoxia [24]. There are two phases of the formation of adaptation mechanisms: the phase of induction of adaptation and the phase of formation of genome-dependent reactions of long-term adaptation [24]. The phase of rapid adaptation to hypoxia is associated with the active use of succinate as an energy substrate, while the phase of long-term adaptation to hypoxia is characterized by expression of a number of enzymes, membrane transporters, regulators of the mitochondrial life cycle and other biomolecules under the influence of transcription factors (HIF-á family) [20].

The level of intracellular lactate is another important aspect participating in the mechanisms of adaptation to hypoxia. Intracellular lactate has been found to regulate the hypoxic response of the cell by stabilizing HIF-1á, HIF-2á and N-myc downstream regulated gene (NDRG) 3 through inhibiting PHD [25]. It is noteworthy that the stabilization of HIF-1á can occur under normoxic conditions, irrespective of oxygen concentration, which is commonly referred to as the pseudohypoxic state of the cell. In this case, the stabilization of HIF-1á occurs due to a disorder of PHD, deficit of their substrate á-ketoglutarate or cofactors — Fe2+ and ascorbate [23]. Finally, increased deubiquitinase activity may also contribute to the stabilization of HIF-1á in normoxia [26]. In hypoxia or pseudohypoxia, the concomitant increase in HIF-1á activity activates pyruvate dehydrogenase kinase (PDK) 1 and lactate dehydrogenase A (LDHA), additionally directing pyruvate metabolism towards lactate formation [14].

Among other TAC metabolites capable of mediating the stabilization of HIF-1á, the role of 2-hydroxylglutarate, fumarate and pyruvate is also noted [23]. In this regard, it seems promising to study the role of TAC metabolites and enzymes involved in their metabolism to evaluate them as possible biomarkers and therapeutic targets in the mechanisms of adaptation to hypoxia for clinical practice.

Targets for HIF-1á in the Aspect of COVID-19 Pathogenesis

Angiotensin converting enzyme (ACE) 2 is expressed in various human cells, including vascular epithelium, lungs, kidneys and intestine, and plays a significant role in the mechanisms of vasodilation [27]. The participation of ACE-2 as a receptor for the penetration of the SARS-CoV-2 virus into the cell has been established [8]. For the virus to penetrate into a host cell, the viral particle binds to ACE-2 receptor and membrane-bound serine protease (TMPRSS) 2 through a receptor-binding domain. ACE-2 acts as a receptor for SARS-CoV-2 by binding to the spike protein of the viral capsid, while TMPRSS2 triggers spike protein priming [28]. Therefore, SARS-CoV-2 is most tropic to cells that express both ACE-2 and TMPRSS2 on the surface of the cell membrane. The activity of ACE-2 can be regulated through the expression of mRNA or protein, as well as through the cleavage of the cell membrane, which is mediated by a disintegrin and metalloproteinase domain (ADAM) 17 [29].

To date, a number of studies have been conducted to determine the role of HIF-1á in the penetration of the SARS-CoV-2 virus into human cells [30]. In a model of hypoxic pulmonary hypertension, the authors demonstrated that HIF-1á can produce an inhibitory effect on the penetration of the virus through ACE-2 through let-7b microRNA [31]. In a study on the mechanism of prostate cancer cell resistance to therapy, it was found that HIF-1á suppresses TMPRSS2 [32]. It has been demonstrated that HIF-1á can also increase the expression of ADAM17, which cleaves ACE-2 off the surface of alveocytes thereby preventing the penetration of SARS-CoV-2 into the cell [29]. Thus, a change in the level of HIF-1á can significantly affect susceptibility of the body to infection with the SARS-CoV-2 virus (Figure 2).

 

Fig. 2. Activation of hypoxia-inducible factor 1α signal pathway associated with reduction of the expression of ACE-2 and TMPRSS2 and an increase in the activity of ADAM17 metalloproteinase. Notes: SARS-CoV-2 — severe acute respiratory syndrome-related coronavirus 2, HIF — hypoxia-inducible factor, ACE — angiotensin-converting enzyme, TMPRSS — serine transmembrane protease, ADAM — a disintegrin and metalloproteinase domain, mRNA — matrix ribonucleic acid.

 

A study on cell lines demonstrated that hypoxia and the prolyl hydroxylase inhibitor HIF-1á (roxadustate) reduce the expression of ACE-2 and inhibit the penetration and replication of SARS-CoV-2 in lung epithelial cells through the HIF-1á-dependent pathway, which is considered a potential approach for the treatment of COVID-19 by the authors [33]. Therefore, HIF-1á is of great importance in the pathogenesis of COVID-19 infection and requires further research as a potential application point for the development of therapeutic strategies.

Disorder in HIF-1á-Mediated Adaptation to Hypoxia as an Element of the Pathogenesis of Hyperactivation of Immune Cells

Metabolic flexibility, known as metabolic adapta-tion, depends on both nutrient availability and oxygen levels [34]. Hypoxic conditions are associated with production of ROS, which leads to damage and dysfunction of mitochondria [35]. Many studies confirm the idea of dysregulation of mitochondrial functions underlying a decrease in metabolic flexibility [34]. Examples of diseases where this occurs may be type 2 diabetes mellitus, obesity, metabolic syndrome, systemic inflammation, malignant tumors, which, in turn, is associated with metabolic reprogramming of cells of the immune system [36].

Metabolic flexibility coordinates metabolic reprogramming, which is characterized by a shift in energy production from mitochondrial oxidative phosphorylation to aerobic glycolysis while main- taining a normal level of partial tension of oxygen in the blood; this aerobic glycolysis is called the Warburg effect [34–37].

It is known that the shift to the glycolytic pheno- type in tumor cells is explained by the action of HIF-1á and its effect on glucose metabolism [38]. In immune cells, such as neutrophils, HIF-1á signaling also regulates glucose uptake and oxygen consumption [39]. According to some authors, the progression of COVID-19 infection is similar to the progression of a malignant tumor, and an important role here is played by HIF-1á [40].

Metabolic reprogramming of immune cells in hypoxia may occur in different ways. Thus, S Rajasundaram, et al. (2018) suggest that the realization of HIF-1á-mediated mechanism of adaptation to hypoxia can produce an anti-inflammatory effect through activation of the adenosine receptor-dependent pathway [41]. Hypoxic conditions stimulate increased release of adenosine with activation of the adenosine signaling system. Thus, adenosine receptors are involved in the release of IL-10 from monocytes/macrophages, and are also involved in inhibiting the proliferation and secretion of tumor necrosis factor (TNF) á, interferon-γ, IL-2, IL-4 and IL-5. The authors suggest that this pathway may be life-saving, as it can protect inflamed tissues of vital organs from damage by hyperactive immune cells [41].

On the other hand, S Galván–Peña, et al. (2014) note that in the progression of inflammation, the effect of hypoxic conditions becomes more noticeable [42]. HIF-1á is important for the formation of the proinflammatory phenotype of M1 macrophages, since its transcriptional activity conditions the metabolic switching of macrophages to glycolysis through the induction of expression of glycolytic genes and glucose transporters [42].

Macrophages, like neutrophils, are involved in inflammatory processes being the earliest immune cells penetrating inflammatory tissues [42]. Staying in the inflammatory microenvironment, macrophages can act depending on the predominant local stimuli either as pro-inflammatory (M1) or anti-inflammatory (M2) agents. After exposure to local stimuli, M1 and M2 macrophages release a wide range of immune mediators.

M1 macrophages possess a proinflammatory cytotoxic effect and produce ROS, cytokines (TNF, IL-1, IL-6, IL-12 and IL-23) and chemokines (chemokine ligand 5, CCL-5; chemokine (C-X-C motif) ligand 9, CXCL9; chemokine (C-X-C motif) ligand 10, CXCL10; chemokine (C-X-C motif) ligand 5, CXCL5) [43]. On the contrary, M2 macrophages mediate anti-inflammatory functions (wound healing, tissue remodeling, angiogenesis) and produce VEGF, epidermal growth factor, transforming growth factor-â, IL-4, IL-13 and IL-10 [43].

Activation of the HIF-1á signaling pathway results in the formation of a proinflammatory pheno-type of M1 macrophages, which in severe cases of COVID-19 infection leads to excessive production of proinflammatory cytokines the phenomenon of a cytokine storm [40].

To date, it has been established that the cytokine storm determines the severity of the disease in patients with COVID-19 infection to a greater extent than the degree of lung damage [44]. Cytokine storm is a disorder in the adequate cytokine production by cells of both innate and adaptive immunity. Dysregulation of the immune system leads to over-intensive activation of more leukocytes, which release inflam- matory cytokines including IL-1â and IL-6, which eventually leads to multiple organ failure [44]. The cytokine profile, directly related to the severity of COVID-19 infection, is characterized by an increase in IL-2, IL-7, granulocyte colony stimulating factor, interferon-ã-inducible protein 10, monocyte chemo-attractant protein 1, macrophage inflammatory protein 1á and TNF [44].

Besides, the state of hypoxia and stabilization of HIF-1á can provoke or intensify a cytokine storm, since the level of VEGF secretion increases under the action of HIF-1á [20]. VEGF and VEGF receptors are directly involved in the migration of circulating inflammatory cells into tissues and in increase in the vascular permeability, which is associated with the progression of the severity of the condition in patients with COVID-19 infection [45].

It should also be added that systemic hypoxia in COVID-19 infection impairs neutrophil turnover [3]. Hypoxia contributes to the reduction of neutrophil apoptosis through HIF-1á, nuclear factor kappaB and macrophage inflammatory protein 1á [46]. It was established that instead of natural clearance, the excessive intravascular activation of neutrophils occurs with subsequent NETosis (neutrophil extra-cellular traps) [47]. The result is a launch of blood clotting and damage to the vascular wall with the development of vasculitis [47].

Thrombosis in the microcirculatory system (small arteries, small veins and capillaries) can cause damage to vital organs, which aggravates the condition of patients with COVID-19 [5, 48]. In addition, hypoxia is one of the important factors contributing to coagulo-pathy [5]. Thus, genes associated with HIF-1á-mediated adaptation to hypoxia induce production of prothrombotic factors such as plasminogen activator inhibitor-1 and tissue factors, and at the same time activation of antithrombotic factors takes place such as thrombomodulin, protein S and tissue factor pathway inhibitor [49]. The study of the effect of HIF-1á signaling pathway, as a biological marker of adaptation to hypoxia, for the activation of various prothrombotic and antithrombotic factors in patients with COVID-19 infection requires further study [5].

CONCLUSION

Systemic hypoxia, which accompanies all stages of coronavirus disease 2019, plays a significant role in its pathogenesis and outcome of the disease. Hypoxia-inducible factor 1á allows cells to adapt to adverse hypoxic conditions, however, being over-stimulated, it can turn from a factor of adaptation into a factor of maladaptation, since its excessive accumulation in cells of the innate immune system disrupts apoptosis of neutrophils in acute respiratory distress syndrome and leads to a cytokine storm.

Summarizing the data, it can be assumed that hypoxia-inducible factor 1á prevents the penetration of the SARS-CoV-2 virus into cells. It also functions as the main regulator of proinflammatory activity at the site of inflammation surrounded by hypoxia. However, in conditions of impaired metabolic flexibility, as, for example, in diabetes mellitus, metabolic syndrome, a high level of hypoxia-inducible factor 1á induces an excessive inflammatory response from the cells of the immune system. A high level of hypoxia-inducible factor 1α in cells in the inflammation focus is associated with an increased production of angio-genesis factors mediating vascular permeability and the capillary leakage process. This is accompanied by tissue damage and organ failure. At the same time, it was found that hypoxia-inducible factor 1α can mediate the anti-inflammatory effect through activation of the adenosine receptor-dependent pathway, which is considered to be a possible protection of cells and organs from damage by hyperactive immune cells.

Further study of the contribution of hypoxia- induced factor 1á to adaptation of the body to hypoxic conditions and its role in the formation of an inflammatory response in the pathogenesis of coronavirus disease 2019 is of particular value. The prospects of correcting the systemic inflammatory response in patients with coronavirus disease 2019 are associated with the determination of ways to control the level of hypoxia-inducible factor 1.

ADDITIONALLY

Funding. This article was not supported by any external sources of funding.

Conflict of interests. The authors declare no conflicts of interests.

Contribution of the authors: R. E. Kalinin, I. A. Suchkov — editing; S. N. Raitsev — study concept and design, data collection and analysis, writing the text; V. I. Zvyagina — study concept and design, editing; E. S. Bel’skikh — data collection and analysis, editing. The authors confirm the correspondence of their authorship to the ICMJE International Criteria. All authors made a substantial contribution to the conception of the work, acquisition, analysis, interpretation of data for the work, drafting and revising the work, final approval of the version to be published and agree to be accountable for all aspects of the work.

Финансирование. Авторы заявляют об отсутствии внешнего финансирования при проведении исследования.

Конфликт интересов. Авторы заявляют об отсутствии конфликта интересов.

Вклад авторов: Калинин Р. Е., Сучков И. А. — редактирование; Райцев С. Н. ― концепция и дизайн исследования, сбор и обработка материала, написание текста; Звягина В. И. ― концепция и дизайн исследования, редактирование; Бельских Э. С. ― сбор и обработка материала, редактирование. Авторы подтверждают соответствие своего авторства международным критериям ICMJE (все авторы внесли существенный вклад в разработку концепции, подготовку статьи, прочли и одобрили финальную версию перед публикацией).

×

About the authors

Roman E. Kalinin

Ryazan State Medical University

Email: kalinin-re@yandex.ru
ORCID iD: 0000-0002-0817-9573

MD, Dr. Sci. (Med.), Professor

Russian Federation, Ryazan

Igor' A. Suchkov

Ryazan State Medical University

Email: suchkov_med@mail.ru
ORCID iD: 0000-0002-1292-5452

MD, Dr. Sci. (Med.), Professor

Russian Federation, Ryazan

Sergey N. Raitsev

Ryazan State Medical University

Author for correspondence.
Email: raitsevsergei@yandex.ru
ORCID iD: 0000-0002-6892-1768
Russian Federation, Ryazan

Valentina I. Zvyagina

Ryazan State Medical University

Email: vizvyagina@yandex.ru
ORCID iD: 0000-0003-2800-5789

Cand. Sci. (Biol.), Associate Professor

Russian Federation, Ryazan

Eduard S. Bel'skikh

Ryazan State Medical University

Email: ed.bels@yandex.ru
ORCID iD: 0000-0003-1803-0542

Cand. Sci. (Med.)

 
Russian Federation, Ryazan

References

  1. WHO. Novel Coronavirus (2019-nCoV). Situation Report — 1. 21 January 2020 [Internet]. Available at: https://www.who.int/docs/default-source/coronaviruse/situation-reports/20200121-sitrep-1-2019-ncov.pdf?sfvrsn=20a99c10_4. Accessed: 2023 January 31.
  2. Phan LT, Nguyen TV, Luong QC, et al. Importation and Human-to-Human Transmission of a Novel Coronavirus in Vietnam. N Engl J Med. 2020;382(9):872–4. doi: 10.1056/NEJMc2001272
  3. Serebrovska ZO, Chong EY, Serebrovska TV, et al. Hypoxia, HIF-1α, and COVID-19: from pathogenic factors to potential therapeutic targets. Acta Pharmacol Sin. 2020;41(12):1539–46. doi: 10.1038/s41401-020-00554-8
  4. Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: Lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol. 2020;38(1):1–9. doi: 10.12932/AP-200220-0772
  5. Hirota K. Hypoxia-dependent signaling in perioperative and critical care medicine. J Anesth. 2021;35(5):741–56. doi: 10.1007/s00540-021-02940-w
  6. Semenza GL. HIF-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol. 2001;13(2):167–71. doi: 10.1016/s0955-0674(00)00194-0
  7. Levchenkova OS, Novikov VE. Inducers of the regulatory factor to hypoxia adaptation. I. P. Pavlov Russian Medical Biological Herald. 2014;(2):133–43. (In Russ).
  8. Batah SS, Fabro AT. Pulmonary pathology of ARDS in COVID-19: A pathological review for clinicians. Respir Med. 2021;176:106239. doi: 10.1016/j.rmed.2020.106239
  9. Matthay MA, Zemans RL, Zimmerman GA, et al. Acute respiratory distress syndrome. Nat Rev. 2019;5(18):1–22. doi: 10.1038/s41572-019-0069-0
  10. Marini JJ, Gattinoni L. Management of COVID-19 Respiratory Distress. JAMA. 2020;323(22):2329–30. doi: 10.1001/jama.2020.6825
  11. Radovanovic D, Rizzi M, Pini S, et al. Helmet CPAP to Treat Acute Hypoxemic Respiratory Failure in Patients with COVID-19: A Management Strategy Proposal. J Clin Med. 2020;9(4):1191. doi: 10.3390/jcm9041191
  12. Yang X, Yu Y, Xu J, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;8(5):475–81. doi: 10.1016/S2213-2600(20)30079-5
  13. Guarnotta V, Ferrigno R, Martino M, et al. Glucocorticoid excess and COVID-19 disease. Rev Endocr Metab Disord. 2021;22(4):703–14. doi: 10.1007/s11154-020-09598-x
  14. Prabhakar NR, Semenza GL. Adaptive and maladaptive cardio-respiratory responses to continuous and intermittent hypoxia mediated by hypoxia-inducible factors 1 and 2. Physiol Rev. 2012;92(3):967–1003. doi: 10.1152/physrev.00030.2011
  15. Ravenna L, Salvatori L, Russo MA. HIF3α: the little we know. FEBS J. 2016;283(6):993–1003. doi: 10.1111/febs.13572
  16. Zhukova AG, Kazitskaya AS, Sazontova TG, et al. Hypoxia-inducible factor (HIF): structure, function, and genetic polymorphism. Hygiene and Sanitation, Russian Journal. 2019;98(7):723–8. (In Russ). doi: 10.18821/0016-9900-2019-98-7-723-728
  17. Diao X, Ye F, Zhang M, et al. Identification of oleoylethanolamide as an endogenous ligand for HIF-3α. Nat Commun. 2022;13(1):2529. doi: 10.1038/s41467-022-30338-z
  18. Zeidan EM, Hossain MA, El-Daly M, et al. Mitochondrial Regulation of the Hypoxia-Inducible Factor in the Development of Pulmonary Hypertension. J Clin Med. 2022;11(17):5219. doi: 10.3390/jcm11175219
  19. Urrutia AA, Aragonés J. HIF Oxygen Sensing Pathways in Lung Biology. Biomedicines. 2018;6(2):68. doi: 10.3390/biomedicines6020068
  20. Luk’yanova LD. Signal’nyye mekhanizmy gipoksii. Moscow; 2019. (In Russ).
  21. Tretter L, Patocs A, Chinopoulos C. Succinate, an intermediate in metabolism, signal transduction, ROS, hypoxia, and tumorigenesis. Biochim Biophys Acta. 2016;1857(8):1086–101. doi: 10.1016/j.bbabio. 2016.03.012
  22. Xiao M, Yang H, Xu W, et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 2012; 26(12):1326–38. doi: 10.1101/gad.191056.112
  23. Paredes F, Williams HC, San Martin A. Metabolic adaptation in hypoxia and cancer. Cancer Lett. 2021;502:133–42. doi: 10.1016/j.canlet. 2020.12.020
  24. Prikhodko VA, Selizarova NO, Okovityi SV. Molecular mechanisms for hypoxia development and adaptation to it. Part I. Arkhiv Patologii. 2021;83(2):52–61. (In Russ). doi: 10.17116/patol20218302152
  25. Baltazar F, Afonso J, Costa M, et al. Lactate Beyond a Waste Metabolite: Metabolic Affairs and Signaling in Malignancy. Front Oncol. 2020;10:231. doi: 10.3389/fonc.2020.00231
  26. Schober AS, Berra E. DUBs, New Members in the Hypoxia Signaling club. Front Oncol. 2016;6:53. doi: 10.3389/fonc.2016.00053
  27. Paz Ocaranza M, Riquelme JA, García L, et al. Counter-regulatory renin-angiotensin system in cardiovascular disease. Nat Rev Cardiol. 2020;17(2):116–29. doi: 10.1038/s41569-019-0244-8
  28. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020;181(2):271–80.e8. doi: 10.1016/j.cell.2020.02.052
  29. Lian G, Li X, Zhang L, et al. Macrophage metabolic reprogramming aggravates aortic dissection through the HIF1α-ADAM17 pathway*. EBioMedicine. 2019;49:291–304. doi: 10.1016/j.ebiom.2019.09.041
  30. Zhang H, Penninger JM, Li Y, et al. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020;46(4):586–90. doi: 10.1007/s00134-020-05985-9
  31. Zhang R, Su H, Ma X, et al. MiRNA let-7b promotes the development of hypoxic pulmonary hypertension by targeting ACE2. Am J Physiol Lung Cell Mol Physiol. 2019;316(3):L547–57. doi: 10.1152/ajplung.00387.2018
  32. Fernandez EV, Reece KM, Ley AM, et al. Dual targeting of the androgen receptor and hypoxia-inducible factor 1α pathways synergistically inhibits castration-resistant prostate cancer cells. Mol Pharmacol. 2015;87(6):1006–12. doi: 10.1124/mol.114.097477
  33. Wing PAC, Keeley TP, Zhuang X, et al. Hypoxic and pharmacological activation of HIF inhibits SARS-CoV-2 infection of lung epithelial cells. Cell Rep. 2021;35(3):109020. doi: 10.1016/j.celrep.2021.109020
  34. Olson KA, Schell JC, Rutter J, et al. Pyruvate and Metabolic Flexibility: Illuminating a Path Toward Selective Cancer Therapies. Trends Biochem Sci. 2016;41(3):219–30. doi: 10.1016/j.tibs.2016.01.002
  35. Sulkshane P, Ram J, Thakur A, et al. Ubiquitination and receptor-mediated mitophagy converge to eliminate oxidation-damaged mitochondria during hypoxia. Redox Biol. 2021;45:102047. doi: 10.1016/j.redox.2021.102047
  36. Smith RL, Soeters MR, Wüst RCI, et al. Metabolic Flexibility as an Adaptation to Energy Resources and Requirements in Health and Disease. Endocr Rev. 2018;39(4):489–517. doi: 10.1210/er.2017-00211
  37. Vaupel P, Schmidberger H, Mayer A. The Warburg effect: essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol. 2019;95(7):912–9. doi: 10.1080/09553002.2019.1589653
  38. Semenza GL. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin Cancer Biol. 2009;19(1):12–6. doi: 10.1016/j.semcancer.2008.11.009
  39. Walmsley SR, Chilvers ER, Thompson AA, et al. Prolyl hydroxylase 3 (PHD3) is essential for hypoxic regulation of neutrophilic inflammation in humans and mice. J Clin Invest. 2011;121(3):1053–63. doi: 10.1172/JCI43273
  40. Jahani M, Dokaneheifard S, Mansouri K. Hypoxia: A key feature of COVID-19 launching activation of HIF-1 and cytokine storm. J Inflamm (Lond). 2020;17:33. doi: 10.1186/s12950-020-00263-3
  41. Rajasundaram S. Adenosine A2A Receptor Signaling in the Immunopathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol. 2018;9:402. doi: 10.3389/fimmu.2018.00402
  42. Galván–Peña S, O'Neill LAJ. Metabolic reprograming in macrophage polarization. Front Immunol. 2014;5:420. doi: 10.3389/fimmu. 2014.00420
  43. Shapouri–Moghaddam A, Mohammadian S, Vazini H, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233(9):6425–40. doi: 10.1002/jcp.26429
  44. Ramasamy S, Subbian S. Critical Determinants of Cytokine Storm and Type I Interferon Response in COVID-19 Pathogenesis. Clin Microbiol Rev. 2021;34(3):e00299-20. doi: 10.1128/CMR.00299-20
  45. Shibuya M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis: A Crucial Target for Anti- and Pro-Angiogenic Therapies. Genes Cancer. 2011;2(12):1097–105. doi: 10.1177/1947601911423031
  46. Walmsley SR, Print C, Farahi N, et al. Hypoxia-induced neutrophil survival is mediated by HIF-1alpha-dependent NF-kappaB activity. J Exp Med. 2005;201(1):105–15. doi: 10.1084/jem.20040624
  47. Juss JK, House D, Amour A, et al. Acute Respiratory Distress Syndrome Neutrophils Have a Distinct Phenotype and Are Resistant to Phosphoinositide 3-Kinase Inhibition. Am J Respir Crit Care Med. 2016;194(8):961–73. doi: 10.1164/rccm.201509-1818OC
  48. Lyubavin AV, Kotlyarov SN. Peculiarities of the Course of Acute Coronary Syndrome in Patients with New Coronavirus Infection. Nauka Molodykh (Eruditio Juvenium). 2022;10(1):101–12. (In Russ). doi: 10.23888/HMJ2022101101-112
  49. Chauhan A, Kaur R, Chakrbarti P, et al. ‘Silent Hypoxemia’ Leads to Vicious Cycle of Infection, Coagulopathy and Cytokine Storm in COVID-19: Can Prophylactic Oxygen Therapy Prevent It? Indian J Clin Biochem. 2021;36(4):468–72. doi: 10.1007/s12291-021-00967-0

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Fig. 1. Regulation of the hypoxia-inducible factor, 1α [18, 19]. Notes: ROS — reactive oxygen species, TAC– tricarbonic acid cycle, RNA — ribonucleic acid, CBP — cyclic AMP response element binding protein (co-activators of CREB-binding protein), FIH — factor inhibiting HIF; GLUT — glucose transporter, HIF — hypoxia-inducible factor, HK — hexokinase, HREs — hypoxia response elements, LDHA — lactate dehydrogenase A, PDK — pyruvate dehydrogenase kinase, VEGF — vascular endothelial growth factor, VHL — von Hippel–Lindau suppressor protein.

Download (88KB)
3. Fig. 2. Activation of hypoxia-inducible factor 1α signal pathway associated with reduction of the expression of ACE-2 and TMPRSS2 and an increase in the activity of ADAM17 metalloproteinase. Notes: SARS-CoV-2 — severe acute respiratory syndrome-related coronavirus 2, HIF — hypoxia-inducible factor, ACE — angiotensin-converting enzyme, TMPRSS — serine transmembrane protease, ADAM — a disintegrin and metalloproteinase domain, mRNA — matrix ribonucleic acid.

Download (32KB)

Copyright (c) 2024 Eco-Vector

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Media Registry Entry of the Federal Service for Supervision of Communications, Information Technology and Mass Communications (Roskomnadzor) PI No. FS77-76803 dated September 24, 2019.



This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies