Volume 6, Issue 1 (Journal of Clinical and Basic Research (JCBR) 2022)                   jcbr 2022, 6(1): 11-27 | Back to browse issues page


XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

EmamiPari F, Kamali S, Nikzad G, Roosta Navi N, Soltani S, Kalani M R. A Review of Pleiotropic Potential of Erythropoietin as an Adjunctive Therapy for COVID-19. jcbr 2022; 6 (1) :11-27
URL: http://jcbr.goums.ac.ir/article-1-324-en.html
1- Department of Animal Sciences, College of Life Sciences, Kharazmi University of Tehran, Tehran, Iran
2- Department of Molecular Cell Biology - Molecular Cell Science, Faculty of Basic Sciences, Mohaghegh Ardabili University, Ardabil, Iran
3- Department of Molecular Cell Biology - Molecular Cell Science, Payame Noor University of Talesh, Gilan, Iran
4- Department of Biology, Shahr-e-Quds Branch, Islamic Azad University, Tehran, Iran
5- Molecular Medicine Department, Golestan University of Medical Sciences, Gorgan, Iran , kalani@goums.ac.ir
Full-Text [PDF 733 kb]   (820 Downloads)     |   Abstract (HTML)  (2154 Views)
Full-Text:   (426 Views)
INTRODUCTION
Coronaviruses (CoVs) which are a wide population of viruses, can infect human and some animals. The effects of the detected CoVs indicate that this group of viruses is responsible for the development of infectious diseases in  human respiratory system. Middle East respiratory syndrome (MERS), and also severe acute respiratory syndrome (SARS) could be mentioned in this group (1, 2). The new group of CoVs called the SARS-CoV-2 that has been discovered late 2019, is responsible for the recent global coronavirus outbreak  (1, 3). This virus was initially detected in Wuhan, China late 2019, however by end of January 2020, 92,262,621 new cases confirmed and about 1,995,000 deaths have been recorded. These statistics indicate the disease's rapid growth and spread (1, 4, 5).
  COVID-19 is related to poor lung function, respiratory compromise due to alveolar inflammation and cytokine storm, and fever (3). In other words, this disease affects the lungs and respiratory tract, causing the person to suffer from dry cough, which can sometimes progress into a severe form of pneumonia (1). It has been proposed that the angiotensin-converting enzyme 2 forms an infiltration pathway for SARS-CoV-2 in the epithelium of the airways and alveoli that leads to local cell death (6). The virus uses a subunit protein called S1 for binding to the host cell (7); the results of the virus' function in the body disrupts lung tissue and its function as well as the function of other essential organs, such as the heart, kidneys, arteries, nerves, skin, and the central nervous system (1, 6). The mortality rate of COVID-19 is 8.2% in men and 7.1% in women, which might be related to the presence of some genes on the X chromosome (4). Hypoxia seems to be one of the symptoms of COVID-19. An expected body reaction to hypoxia is increased production of erythropoietin; therefore, the use of EPO in the treatment of COVID-19 has been proposed (6, 8). This glycoprotein hormone/cytokine is primarily produced by a molecular signal known asthe hypoxia-inducible factor-2 (HIF2), which increases the count of the red-blood-cells (RBC) produced by the bone marrow by inhibiting the apoptosis of erythrocyte precursors. The main source of EPO is the liver during fetal development and kidneys in the adulthood (3, 5, 8, 9). Studies indicate that EPO expression occurs in various body tissues (10, 11). Erythropoietin therapy in COVID-19 improves respiration, reduces inflammation in effect of cytokine storm, and exerts neuroprotective effects on the brain and peripheral nervous system (10). In addition, with lowering the levels of IL-6 and hepcidin, EPO increases iron release from macrophages iron absorption by the bone marrow. Consequently, the virus's access to iron, which is required for its enzymatic activity, is reduced (3).
  It is also effective in reducing respiratory disorders through its anti-apoptotic effects, releasing leukocytes from the bone marrow, and affecting the distribution of iron so that it is not exposed to the coronavirus (3, 12). This article aimed to review the effect of recombinant human EPO on COVID-19 and related hypoxia as well as EPO counteraction with apoptosis.
MATERIALS AND METHODS
  The Science Direct, PubMed, and Google Scholar databases were systematically searched for published articles about COVID-19 and its treatment with recombinant human EPO. The World Health Organization was also used to obtain COVID-19 related statistics. In our research, relevant articles, were filtered using the terms "Review published during 2016 to 2021", and older articles were used to obtain some definitions of keywords such as EPO and cardiovascular therapies, induction of EPO hypoxemia in COVID-19, pathogenesis, and structure of COVID-19, etc. In general, the criteria for selecting articles included key information like a definition of the mechanism of injury in COVID-19, hypoxic effects and treatments in COVID-19, and therapeutic effects of the EPO. Furthermore, published research papers with high scientific impact based on their citation frequency were included in the study. Articles that had been focused on the psychological aspects of the disease were excluded. More than 100 articles were obtained, and only 96 articles were eligible to be enrolled in the study.
RESULTS
 Current Perspective, Structure, and Pathogenesis of SARS-CoV-2
  Coronaviruses are a large family of viruses that are very diverse in their phenotypes and genotypes (13). These viruses belong to the family Coronaviridea, the order Nidovirales, and the genus Coronavirus. There are four genera within the subfamily Ortho-Coronaviridea, namely α-CoV, β-CoV, γ-CoV, and δ-CoV (14). Furthermore, studies show that both the α- and β-CoV genera are known to infect mammals, while the δ- and γ-CoVs infect birds. SARS-CoV-2 belongs to the genus β-CoV due to its phylogenetic relationships and genomic structure. The Coronaviridae Study Group (CSG) of the International Committee on Taxonomy of Viruses named this virus SARS-CoV-2 because of the close resemblance between its sequences and the CoVs that are associated with SARS-CoV (13, 14, 15, 16). It is a medium-sized, single-stranded, RNA virus with nucleoprotein within a capsid consisting of protein matrix. The name CoV was chosen because of the crown-like ACE2 receptor on the virus's surface (13, 15).
  Coronaviruses include the largest length of genome (26.4 to 31.7 kb) among the known RNA viruses, with GC contents varying from 32% to 43% (13, 15).  The SARS-CoV-2 genome (average size 30 kb) is a large portion of the unstructured polyprotein (ORF1a/b), which is broken down into 15 or 16 separate proteins including four structural proteins and five lateral proteins (ORF8, ORF3a, ORF6, ORF7, and ORF9) (14). The four structural proteins include glycoproteins (present on the surface), spike protein (S), membrane protein (M), coating protein (E), and nucleocapsid protein (N); the latter is essential for the assembly and infection of SARS-CoV-2. Surface glycoproteins play a key role in host cell attachment and further division by N-terminal (S1 subunit) and C-terminal (S2 subunit) proteins and membrane-bound spike glycoprotein (13, 14). Understanding the structure and function of spike proteins can help produce monoclonal antibodies and vaccines.
  As mentioned earlier, CoVs are RNA viruses that can infect the respiratory, circulatory, gastrointestinal, and central nervous systems in various vertebrates such as bats, birds, penguins, snakes, mice, and humans. According to statistics, SARS-CoV and MERS-CoV are highly contagious and have resulted in death of thousands of people in the last two decades (17, 18). Since late December 2019, a new coronavirus called SARS-CoV-2 has caused a global outbreak of a respiratory disease known as COVID-19 (19, 20, 21, 22). Infection with the virus was first discovered in Wuhan, China; this serious contagious infection spreads throughout China and to other countries (17, 22). According to studies, SARS-CoV-2 is thought to have a common human-animal origin due to the similarity of the RaTG13 sequence to the CoV strains found in bats and pangolins. In the next stage, the virus acquires the ability to spread from human to human. Human-to-human transmission of COVID-19 is possible through respiratory particles or direct contact with patients (13, 18). The most common early symptoms of this disease are fever, dry cough, fatigue, diarrhea, etc. (17, 22). Redness and burning of the eyes and loss of smell have also been reported in some patients with COVID-19 (23). In general, the symptoms of COVID-19 range from mild respiratory tract infection to acute complications such as severe progressive pneumonia, multi-organ failure, which could be fatal, especially in elderly patients, people with respiratory or cardiovascular diseases, and immunocompromised individuals (19, 20, 21, 21). Studies show that acute respiratory distress syndrome (ARDS) in patients with COVID-19 leads to decreased blood oxygen levels. Generally, when a person inhales through the nose, air travels down the pharynx, larynx, and trachea into the alveoli, where oxygen is transferred from the inhaled air into the blood and then is transported to all body parts. During ARDS, the capillaries inside the lungs leak more fluid into the lungs than normal conditions, preventing oxygen transport from the lungs to other body organs; consequently, the body organs are weakened or paralyzed (24, 25). The probable reason for CoVs infection in the respiratory tract is the presence of dipeptidyl peptidase 4 and ACE2 in the lower respiratory tract, which are the most important human receptors for MERS-CoV and SARS-CoV (14). Similar to SARS-CoV, SARS-CoV-2 uses the ACE2 receptor to enter human cells, which might be due to the high similarity rate (70%) of the genetic sequences between the two viruses (26, 27). However, the spike protein differs between the two viruses. The spike protein in SARS-CoV-2 has more overlap with the ACE2 receptor, thereby facilitating human-to-human transmission (14, 26, 27). With the entry of alveolar epithelial cells and the rapid proliferation of the virus in those cells, a severe immune response coupled with cytokine hyper-secretion syndrome and lung tissue damage occurs (14). Furthermore, the number of T cells, CD4 receptor, and CD8 T cells are reduced, resulting in immune suppression. The resulting secondary infection causes respiratory dysfunction (28).
Recombinant EPO and its structure
  Human EPO (EPO) is a 30.4 kDa glycoprotein hormone composed of 165 amino acids (29, 30). By binding to its receptor on the cell surface, EPO receptor (EPOR), EPO acts as a hematopoietic growth factor and stimulates the proliferation, differentiation, and maturation of erythroid progenitor cells during the process of erythropoiesis (8, 31, 32, 33). It induces hypoxia in the liver, renal interstitial cells, neurons and astrocytes, lungs, spleen, and bone marrow (34, 35) and acts as a cytokine with pleiotropic protective effects. The EPOR is a 52 kDa peptide with a single carbohydrate chain (33, 36) and belongs to the class 1 cytokine receptor superfamily. It consists of a WSXWS motif in the extracellular domain, a single global domain, and a cytoplasmic domain without tyrosine kinase activity; the latter is associated with Janus kinase (JAK) and forms homodimer, heterodimer, and heterotrimer complexes (37, 38). Studies indicate the significance of the presence of EPOR on the surface of erythroid progenitor cells to make red blood cells (39, 40, 41 42). However, EPOR expression is not restricted to erythroid tissue. This receptor is also expressed in non-erythroid cells, including neurons, endothelial cells, bone marrow stromal cells, and skeletal muscle myoblasts (41, 43). Using several signaling pathways, such as JAK2, EPO can exert its protective and anti-apoptotic effects on various cells and tissues in the body, including the lungs, kidneys, heart muscles, the nervous system, retina, pancreas, and endothelial cells (12, 30, 44). Erythropoietin is held adjacent to cytoplasmic JAK2 kinases by binding to its homodimer receptor complex, thereby providing the possibility of JAK transphosphorylation, receptor phosphorylation, and activation of signal transducers and activators of transcription (STAT) and other downstream signaling pathways, including alpha serine/threonine-protein kinase and Extracellular signal-regulated kinases (45, 46, 47, 48).
  Recombinant human EPO was first discovered in 1985 using recombinant DNA technology (49). It is a glycoprotein with 165 amino acids arranged inside the molecule in a single chain by two disulfide bonds. Moreover, it has an N-glycosylation site in three specific asparagine amino acids (Asn24, Asn38, Asn83) and an O-glycosylation site in Ser126 (50, 51). Types of recombinant human EPO available today include alpha epoetin (Procrit, Epogen, Epogen, and Eprex), which is the first recombinant human EPO treatment product (52), beta epoetin (NeoRecormon, and Recormon), gamma epoetin (Dynepo), and omega epoetin (Epomax) (53). Recombinant human EPO is an erythrocyte-stimulating factor with protective effects on the body (44, 53, 54). It is used to treat chronic inflammatory diseases, infants' prematurity, immune system failure, and chronic renal failure-related anemia also known as end-stage renal disease (30, 50, 53, 55, 56). The common β-receptor (βCR), CD131 (also known as the hemopoietin receptor) (57), is a suggested alternative to EPOR (58, 59). It is expressed in non-hematopoietic tissues, including central and peripheral nerve tissue, heart, kidneys, retina, endothelium, and muscles (60), and shares the WSXWS motif in the extracellular part with the EPO (57). Studies on an in-vitro model of colitis in mice indicated the role of heterocomplexes βCR–EPOR in developing the EPO protective response (54), although there is still no clear understanding of its signaling pathway (60, 61). Some studies have shown that EPO derivatives can enhance its protective effects by binding to alternative EPO receptors such as EPOR/βc heterodimer receptor in non-hematopoietic tissues without stimulation of erythrocyte production (58, 61). These EPO derivatives include asialo-EPO, carbamylated EPO, recombinant helix B peptide, and EV-3, an EPO transcript characterized by deletion of exon 3 (62, 63),
Therapeutic effects of EPO on COVID-19
In general, production of EPO at high altitudes can help adapt to hypoxic conditions, which can provide therapeutic goals for the prevention and treatment of COVID-19 (6, 8, 10, 64). Depending on the severity of the disease as well as other factors such as age, underlying conditions, etc., symptoms of COVID-19 may vary and can include inflammatory endothelial damage, shortness of breath, vasoconstriction and pneumonia, neuro-edema, inflammation of the heart, heart thrombosis, stroke, hemolysis, and potential carotid dysfunction. Increasing EPO levels may have a significant therapeutic effect on most of the above pathological features. Recombinant human EPO may effectively suppress silent hypoxemia and the loss of red blood cells in severe cases of COVID-19 (6, 10, 22, 64, 65, 66, 67, 68). The effects of EPO on various organs of the body are discussed below.
Effects of EPO on inflammation and cytokine storm caused by COVID-19
  Pneumonia, lymphopenia, lymphocyte depletion markers, and cytokine storm have been described as indicators of severe COVID-19 (3, 4, 10). In this case, the amount of C-reactive protein (CRP), D-dimer, and pro-inflammatory cytokines is significantly increased, which are involved in tissue damage (69, 70, 71). On the other hand, in these patients, the amount of T cells, CD4+ or CD8+ T cells cells, or natural killer cells as well as B cells, basophils, monocytes, and even eosinophils, are significantly reduced. Subsequently, SARS-CoV-2 infection could potentially induce T-cell apoptosis. Nucleotide-binding _domain_leucine rich (NLR), whisch is a family of host immune-dependent pattern-recognition molecules, is resistant to several pathogen viruses. Several experiments performed with double-stranded RNA (poly-I: C) and single-stranded RNA (ssRNA40) analogs show an NLRP3-mediated serious response can be activated by a 'virus-mimeti’ RNA strains. Inflammation of NLRP3 is an essential host action to fight infection through sensitivity to viral RNA, which could otherwise be very destructive. In general, this excessive inflammatory response is a major factor in the developing an ARDS with NLRP3 inflammation. In animal studies, EPO could effectively reduce lung damage caused by viral infection by suppressing NLRP3 inflammation, which depends on the activation of EPOR/JAK2/STAT3 and inhibition of the NF-κB pathway (70, 71). Subsequently, the inflammation is significantly reduced by caspase-1 cleavage, leading to the development and progression of COVID-19.
Suppression of pro-inflammatory cytokines by EPO protects cells against apoptosis and repairs tissue lesions. In addition, EPO-R which is expressed on several immune cells, thus enabling EPO to regulate their differentiation and activation, and even function directly. Respiratory burst of phagocytes activates macrophage EPO signaling, which causes severe inflammation. Modulation of the immune system and the fight against inflammation by EPO promises another beneficial effect in severe COVID-19 infection. Therefore, EPO, as an anti-inflammatory, immunosuppressive, and antiviral drug, is beneficial for patients with COVID-19. Moreover, known adverse effects of antiviral medications such as anemia, observed during SARS, may be resolved with EPO (10). Recent observations suggest that SARS-CoV-2 infections may produces even kidney or heart failure due to harmful inflammation (during the cytokine storm) which target vascular beds. Erythropoietin is able to significantly protect inflammation caused tissue damage (6).
Effects of EPO on the treatment of blood disorders caused by COVID-19
  Erythropoietin has been shown to stimulate the production of red blood cells in hypoxia by binding to REPO on erythroid progenitor cells (25). Recombinant EPO enables development of therapeutic strategies that stimulate erythropoiesis by erythrocyte stimulating factor (ESA) in diseases where the production of normal erythrocytes is impaired (25, 70). Although iron is essential for the normal functioning of various proteins and enzymes, this element can be toxic in the Fenton reaction by producing reactive oxygen species (ROS). Therefore, its availability, absorption from the intestine, transport in the body, storage, and metabolism must be fully regulated. Most of the iron in the body is used to make hemoglobin and is stored intracellularly through chelation with proteins, such as ferritin. Hepcidin is the main regulator of iron homeostasis, and its serum production is highly expanded during infection and severe inflammatory conditions. Studies show that overexpression of hepcidin occurs through the JAK-STAT3 pathway and pattern recognition receptor signaling in hepatocytes and myeloid leukocytes, reducing iron availability to red blood cells. There is growing evidence that emphasizes the possible role of iron, ROS, and iron-induced damage-associated molecular patterns (DAMPs) in activation of the NLRP3 and NF-κB signaling pathways, respectively. At all stages of SARS-COV-2 infection, the host body must limit the amount of free iron available to pathogens in order to prevent increased infectivity and pathogenicity (56, 68, 70, 71, 72, 73). In general, hepcidin overexpression due to SARS-CoV-2-related infection and inflammation causes iron deposition in mononuclear phagocytes, which leads to the growth of pathogenic agents that are highly sensitive to the amount of iron. In this condition, macrophages begin to express high levels of pro-inflammatory cytokines. Generally, EPO may also affect iron storage by transferring it to bone marrow, reducing available free iron. The role of EPO is to redistribute iron to produce enough hemoglobin (68, 70, 71, 73).
  There is some evidence that exogenous EPO administration to healthy volunteers can significantly reduce serum hepcidin without affecting serum iron levels (68). Moreover, EPO effectively inhibits activation of the NF-κB and JAK/STAT3 pathways, which may be induced by DAMP, iron, and iron-induced ROS (68, 70, 71). Experiments show that most COVID-19 patients shows seroious anemia; this probably can be partly due to the changes in hepcidin and irregular iron metabolism. On the other hand, as mentioned in the previous sections, patients infected with SARS-CoV-2 usually have a severe inflammatory response. Studies show that in conditions like inflammation, erythrocyte production stimulants have very limited effects on red blood cell production; therefore, in patients with healthy or compromised kidneys where anemia due to inflammation is unlikely, an effective response to erythrocyte production is impossible. However, it can be hypothesized that in patients with COVID-19, there may be a link between the elevated cytokinin and the concomitant effects of hepcidin levels and irregular iron metabolism. Effective agents in stabilizing hypoxia-causing factors are oral drugs that increase EPO production and increase iron availability. Preliminary results indicate that these drugs can treat anemia more effectively in the inflammatory environment than erythrocyte production (70, 73).
Effects of EPO in the treatment of COVID-19-related cardiovascular disorders
  Many studies in the United States, China, and Italy indicate that the new CoV also attacks the heart. To prove the link between heart diseases and COVID-19, researchers have pointed out the followings:
  • High mortality rates among patients with the new CoV have a history of cardiovascular disease and hypertension.
  • Elevated blood troponin level is a biomarker of cardiomyocyte damage and has been observed in patients with acute disease.
  • Infection of the heart muscle in patients with no history of the disease.
The new CoV has many similarities to the pathogens MERS and SARS in terms of cardiovascular symptoms (2). The SARS-CoV-2 infection can develop several cardiovascular abnormalities, including bou not limited to cardiomyopathy, myocardial injury and myocarditis or even pericarditis. Other abnormalities like and cardiac arrest following arrhythmia, heart failure, and coagulation abnormalities are reported too. Many patients have suffered from cardiac symptoms like palpitations, chest stiffness, or acute cardiovascular injury (65). Patients with cardiovascular diseases and hypertension have a higher risk of developing severe COVID-19. Myocardial damage, which may diagnosed by high levels of cardiac disease biomarkers, is due to ischemic and/or non-ischemic reason such as myocarditis (74, 75, 76 77). Numerous studies suggest an acute myocardial injury in patients with COVID-19 (65, 77). Limited cases of COVID-19 analysis indicate a significant interstitial influence on pro-inflammatory mononuclear cells in cardiac tissue, which confirms the myocardial injuries (78, 79). Recently, Tavazzi have reported the first case of SARS-CoV-2 pathogens in the heart. They reported that the heart could be directly infected with SARS-CoV-2 (77).  Hu et al. also reported a 53-year-old patient with COVID-19 admitted to the intensive care unit due to systolic dysfunction and viral myocarditis (65).
Furthermore, cardiac arrest leading to sudden death is frequently seen in COVID-19 (74, 80). Among 85 cases of death due to COVID-19, the leading cause of death with seven cases was the cardiac causes (81). Chances of survival of COVID-19 patients with acute symptoms of cardiac arrest are relatively poor (82). Despite all the available interpretations, there is still no credible and direct evidence to confirm cardiac arrest as a complication of COVID-19 (83). However, EPO has beneficial, anti-ischemic,  regeneration-promoting, and anti-apoptotic effects on a variety of tissues, including the lung, kidney, heart muscle, nervous system, retina, pancreas, and endothelial cells (12, 44), which are exerted through its specific receptor EPOR/βCR (84).
  Furthermore, EPO stabilizes vascular integrity, increases the number of endothelial cells, and protects these cells against ischemia and apoptosis. Due to its molecular mechanism of action of EPO, it induces Ca 2+ in the EC aorta by activating the phospholipase C-γ1 (PLC-γ1) signaling pathway, which leads to the activation of a potential transient receptor vanilloid 1 (5). After the autopsy of patients with COVID-19, researchers in Switzerland found that in some patients, the whole-cell layer is inflamed in the inner layer of blood vessels and lymph of various organs. The researchers concluded that the CoV, through ACE-2, causes inflammation in the intravascular layer. This inflammation can lead to minor circulatory disorders, which damage the heart and can lead to pulmonary embolism and blockage of arteries in the brain and gastrointestinal tract (66). Moreover, EPO treatment in rats show that they protect the heart by reducing the myocardial inflammatory responses and mitochondrial membrane potential and reducing myocardial cell apoptosis through the mitochondrial pathway by reducing NF-Κb p65 expression (24).
Therapeutic effects of EPO on pulmonary arteries
  Studies show that most COVID-19 patients suffer from acute lung damage. Due to the prevalence of vascular damage in the different tissues, the main roles of EPO is to protect the pulmonary endothelium and prevent pulmonary edema. Experiments on mice show that EPO plays an important role in suppressing pulmonary edema and reducing the swelling of alveolar epithelial cells in acute pulmonary damage due to ischemia-reperfusion injury. Another study by Heitrich et al. on mouse models of acute sepsis-induced lung injury and acute kidney injury showed that EPO, through the expression of EPO-R and vascular endothelial growth factor/ vascular endothelial growth factor 2, had protective effects on the lungs and kidneys (79). This may be related to improved oxygen supply for tissues, which will lead to the reduction of inflammation stimuli (10, 66). Moreover, the anti-inflammatory effects of erythropoietin in protection of lung damage have been reported in animal models too. The critical mechanism for this level of the pulmonary edema is vasoconstriction due to hypoxia in the pulmonary arteries and capillaries. Redistribution of blood flow from the basal regions to the apical surface of the lungs also is effective. In this case, EPO counteracts the constriction of the pulmonary arteries by increasing the endothelial capacity to produce nitric oxide (NO), which causes vasodilation. Studies in transgenic (Tg6) mice injected with large amounts of human EPO revealed that despite the upper hematocrit level of 80% roughly, the blood pressure, heart rate, and output were in the normal range, which may be due to step by step cardiovascular adaptation by consequently NO-induced vasodilation (10). Compatible mechanisms with erythrocytosis in animals also include extra-activity of endothelial NO synthase molecules (eNOS). Moreover, increased NO synthesis by eNOS in Tg6 mice induced dilation of the peripheral arteries. When this experiment was replicated on Tibetans and Andeans, the increase in NO metabolism in lungs of these people acted as an alternate mechanism that counteracts the lack of oxygen at high altitudes. In conclusion, EPO can be an effective selection in therapeutic strategy to prevent acute lung injury and edema caused by SARS-CoV-2 by protecting the pulmonary vascular endothelium and causing vasoconstriction (10).
The effects of EPO in the treatment of neurological disorders caused by COVID- 19
  Studies have shown that coronaviruses can attack the central nervous system and induce neurological diseases (10). Moreover, studies on the neuro-therapeutic potential of EPO after ischemic injury of the central nervous system in patients with COVID-19 produced acceptable results. Due to improved tissue oxygenation, EPO can act as a neuroprotective, anti-apoptotic, antioxidant, angiogenetic, and nootropic agent. In 2005, Brines and Cerami  showed that EPO receptor cells in the central nervous system (astrocytes, neurons, oligodendrocytes, and nerve progenitor cells) could act as a line of defense against damage caused by ischemic events and prevent apoptosis (50). On the other hand, aging is one of the most important factors in cerebral ischemia. Studies show that older mice with ischemic stroke have significant differences in neuroinflammation, increased permeability of the blood-brain barrier, high rate of severe apoptosis, myocardial infarction, expression of genes associated with reduced cell apoptosis, regulation of inflammatory mediators, and central nervous system dysfunction compared with young and healthy mice. These factors can indirectly affect the rate of EPO secretion and its positive effects in patients with COVID-19 (65, 85, 86).
Effects of EPO on COVID-19-induced hypoxia induction
  As mentioned in the previous sections, if the body is experiencing a reduction in red blood cells and a lack of oxygen, EPO produced under hypoxic conditions in COVID-19 can compensate for the erythrocyte deficiency by stimulating erythropoiesis (87, 88). Moreover, the kidneys are the main source of EPO production, targeting red blood cell progenitors in the bone marrow which can stimulate the blood cells propagations (9, 87, 88). Therefore, EPO treatment for SARS-COV-2 may help to save hemoglobin levels and consequently improve oxygen delivery to tissues.  Lechuga et al. suggested that SARS-COV-2 itself attacks the hemoglobin beta chains, but the EPO can effectively treat red cell deficiency in patients with COVID-19 during hypoxemia (87). In addition, EPO could be a good treatment option for short-term medical emergencies of silent hypoxemia. It has been reported that EPO can exert neuroprotective effects in ischemic stroke and brain injury and prevents cardiorespiratory dysfunction that occurs following intermittent hypoxemia (6). Thus, EPO can reduce the severe neurological symptoms caused by COVID-19 by acting on both the central and peripheral systems.  According to Khoo et al., SARS-CoV-2 may specifically affect brainstem function, which can be neutralized with EPO therapy (89). In conclusion, increasing the level of EPO is one of the most promising ways to deal with the adverse effects of hypoxemia in COVID-19 (6).
EPO restriction
  The amount of D-dimer in patients with COVID-19 increases significantly, followed by many distinct blood disorders caused by the deposition of coagulation fibrin filaments in small blood vessels, lymphatic capillaries, accompanied with the deposition of these fibers outside the cell (67). Moreover, studies show that patients with severe COVID-19 often have prolonged prothrombin, increased D-dimer levels, and decreased fibrinogen and disseminated intravascular coagulation (67, 73). Inflammatory cytokine storm is a hallmark of COVID-19. In patients with severe infections, shortness of breath and lymphopenia are accompanied with elevated levels of IL-2R, IL-6, IL-10, and TNF-α. The severe secondary inflammatory condition leads to a reduction in homeostasis and a marked change in coagulation parameters.  The deterioration of coagulation parameters during COVID-19 could lead to various life-threatening complications (90).
  Despite its numerous benefits, EPO can aggravate blood coagulation disorders and deterioration of various organs by forming microthrombosis (91). Other side effects of recombinant human EPO administration include high blood pressure, increased tumor growth, thromboembolism, anaplasia of pure red blood cells, etc. (67, 73). Experimental results also indicated  arterial hypertension as another side effect of recombinant human EPO, which increases the risk of cardiovascular disease (67, 68, 70), resulting in vasoconstriction (90) as well as dysregulation of the renin-aldosterone system (reduced excretion of sodium by the kidneys) (25, 71). However, the hypertensive effects of recombinant human EPO are independent of its erythropoietic activity (68, 70). In addition, current reported research claim that infection with SARS-CoV-2 is likely to predispose its victims to thrombosis (92).
DISCUSSION
  Erythropoietin is a hematopoietic factor that stimulates erythrocyte production. The gene encoding EPO is located on chromosome 7 and encodes a polypeptide that consists of four α-strands (8, 31, 68). As the atmospheric oxygen pressure decreases at high altitudes and certain tissues in the body may become hypoxic, the need to produce erythrocytes to carry more oxygen to the tissues increases. This results in increased expression of EPO. On the other hand, the expression of EPO stimulates erythroid progenitor cells in the bone marrow to increase erythrocyte production (10, 34, 35, 93, 94).  The receptors of EPO are expressed on erythroid cells, nerve cells, stromal cells of bone marrow, lung, liver, heart, and brain cells (68, 95, 96). In addition to producing erythrocytes and reducing the possible risks of hypoxia, EPO has anti-apoptotic properties that have been observed in kidney cells, lungs, etc. (30, 44). Further research led to the production of recombinant human EPO, which is made by recombinant DNA technology (49, 53). One of the functions of recombinant human EPO is to stimulate the production of erythrocytes in the body (44, 53, 54). The results indicate that EPO derivatives, which bind to a specific receptor, have protective effects on non-hematopoietic tissues independent of erythrocyte production (62, 65). In acute hypoxemia of nerve tissue following COVID-19 in the central nervous system, EPO prevents nerve cell apoptosis (65). According to Sahebnasagh et al., EPO effectively reduces COVID-19-related lung damage by suppressing NLRP3 inflammation (68), which acts as a host defense mechanism against viral RNA infection by suppressing pro-inflammatory cytokines (6, 10). Furthermore, EPO increases iron storage and transports iron to the bone marrow, decreasing hepcidin, the main regulator of iron homeostasis (68, 70, 71, 73).  The presence of EPOR-βCR on the lungs, kidneys, and endothelial muscles, with anti-ischemic, regenerative and anti-apoptotic effects, stabilizes vascular integrity and protects against ischemia and apoptosis (5, 44, 84, 96). In some patients with COVID-19, the entire intracellular layer of blood and lymphatic vessels of various organs become inflamed, which is caused by ACE2 that disrupts blood flow and causes heart damage. This can eventually lead to pulmonary embolism and blockage of arteries in the brain and gastrointestinal tract. Erythropoetin reduces the myocardial inflammatory response and mitochondrial membrane potential by protecting the heart and reducing myocardial cell apoptosis through the mitochondrial pathway by decreasing the NFKB p65 expression  (5, 44, 84). Studies show that injection of human EPO into mice increases NO metabolism by stimulating the activity of eNO, an adaptive mechanism that protects the pulmonary vascular endothelium and prevents vascular adaptation (10). In general, EPO therapy has numerous benefits COVID-19, but the limitations such as risk of microthrombosis and blood failure in some organs should be taken into account (91). Recombinant human EPO increases arterial blood pressure and causes cardiovascular disease, directly affecting vasoconstriction and disrupting the renin-aldosterone system (25, 67, 68, 70, 71, 90). In conclusion, to prevent coagulation and anemia in patients with COVID-19 by EPO, it is advised to co-administer anticoagulants or antithrombotic agents, such as heparin (6).
CONCLUSION        
  Erythropoietin can act as a neuroprotective, anti-apoptotic, antioxidant, angiogenic, and erythrocyte precursor stimulant against hypoxia-induced damages. The function of EPO and recombinant human EPO can reduce or eliminate the adverse effects of COVID-19. However, EPO should be used with caution for treatment of COVID-19. A real gap in the current knowledge of applying EPO for treatment of COVID-19 patients is the dosage regarding the increased velocity and risk of thrombosis. Logically, this dosage gap needs more animal research to be determined. However, based on our experience, some of the animal experiments on SARS-CoV-2 may not extend to human cases. Therefore, prescribing lower and safe doses in critical patients may be a promising approach.
ACKNOWLEDGMENTS
  The authors express their appreciation and gratitude to the :union: of Iranian Biologists for providing the necessary basis for writing the article.
DECLARATIONS
Funding
The authors received no financial support for this article's research, authorship, and/or publication.
Ethics approvals and consent to participate
Not applicable.
Conflict of interest
The authors declare that there is no conflict of interest regarding the publication of this article.
Article Type: Review | Subject: Cellular and Molecular Biology

References
1. World Health Organization [Internet]. 2021.
2. Alhogbani T. Acute myocarditis associated with novel Middle east respiratory syndrome coronavirus. Ann Saudi Med. 2016;36(1):78-80. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
3. Hadadi A, Mortezazadeh M, Kolahdouzan K, Alavian G. Does recombinant human erythropoietin administration in critically ill COVID-19 patients have miraculous therapeutic effects? J Med Virol. 2020;92(7):915-8. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
4. Tay MZ, Poh CM, Rénia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nature Reviews Immunology. 2020;20(6):363-74. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
5. Kimáková P, Solár P, Solárová Z, Komel R, Debeljak N. Erythropoietin and Its Angiogenic Activity. Int J Mol Sci. 2017;18(7). [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
6. Soliz J, Schneider-Gasser EM, Arias-Reyes C, Aliaga-Raduan F, Poma-Machicao L, Zubieta-Calleja G, et al. Coping with hypoxemia: Could erythropoietin (EPO) be an adjuvant treatment of COVID-19? Respir Physiol Neurobiol. 2020;279:103476-. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
7. Baig AM, Khaleeq A, Ali U, Syeda H. Evidence of the COVID-19 Virus Targeting the CNS: Tissue Distribution, Host-Virus Interaction, and Proposed Neurotropic Mechanisms. ACS Chem Neurosci. 2020;11(7):995-8. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
8. Lacombe C, Mayeux P. Biology of erythropoietin. Haematologica. 1998;83(8):724-32. [View at Publisher] [Google Scholar]
9. Kobayashi H, Liu J, Urrutia AA, Burmakin M, Ishii K, Rajan M, et al. Hypoxia-inducible factor prolyl-4-hydroxylation in FOXD1 lineage cells is essential for normal kidney development. Kidney Int. 2017;92(6):1370-83. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
10. Ehrenreich H, Weissenborn K, Begemann M, Busch M, Vieta E, Miskowiak KW. Erythropoietin as candidate for supportive treatment of severe COVID-19. Mol Med. 2020;26(1):58. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
11. Dame C, Fahnenstich H, Freitag P, Hofmann D, Abdul-Nour T, Bartmann P, et al. Erythropoietin mRNA expression in human fetal and neonatal tissue Blood. 1998;92(9):3218-25. [View at Publisher] [DOI] [PMID] [Google Scholar]
12. Ghezzi P, Brines M. Erythropoietin as an anti-apoptotic, tissue-protective cytokine. Cell Death & Differentiation. 2004;11(1):S37-S44. [View at Publisher] [DOI] [PMID] [Google Scholar]
13. Shanmugaraj B, Siriwattananon K, Wangkanont K, Phoolcharoen W. Perspectives on monoclonal antibody therapy as potential therapeutic intervention for Coronavirus disease-19 (COVID-19). Asian Pac J Allergy Immunol. 2020;38(1):10-8. [Google Scholar]
14. Li H, Liu SM, Yu XH, Tang SL, Tang CK. Coronavirus disease 2019 (COVID-19): current status and future perspectives. Int J Antimicrob Agents. 2020;55(5):105951. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
15. Mousavizadeh L, Ghasemi S. Genotype and phenotype of COVID-19: Their roles in pathogenesis. J Microbiol Immunol Infect. 2021;54(2):159-63. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
16. Vellingiri B, Jayaramayya K, Iyer M, Narayanasamy A, Govindasamy V, Giridharan B, et al. COVID-19: A promising cure for the global panic. Sci Total Environ. 2020;725:138277. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
17. Zhang R, Wang X, Ni L, Di X, Ma B, Niu S, et al. COVID-19: Melatonin as a potential adjuvant treatment. Life Sci. 2020;250:117583. [View at Publisher] [DOI:10.1016/j.lfs.2020.117583] [PMID] [PMCID] [Google Scholar]
18. Asselah T, Durantel D, Pasmant E, Lau G, Schinazi RF. COVID-19: Discovery, diagnostics and drug development. J Hepatol. 2021;74(1):168-84. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
19. Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, et al. A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19. N Engl J Med. 2020;382(19):1787-99. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
20. Dai M, Liu D, Liu M, Zhou F, Li G, Chen Z, et al. Patients with Cancer Appear More Vulnerable to SARS-CoV-2: A Multicenter Study during the COVID-19 Outbreak. Cancer Discov. 2020;10(6):783-91. https://doi.org/10.2139/ssrn.3558017 [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
21. Zhu FC, Li YH, Guan XH, Hou LH, Wang WJ, Li JX, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet. 2020;395(10240):1845-54. [View at Publisher] [DOI] [Google Scholar]
22. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497-506. [View at Publisher] [DOI] [Google Scholar]
23. Hu K, Patel J, Swiston C, Patel BC. Ophthalmic Manifestations Of Coronavirus (COVID-19). StatPearls. Treasure Island (FL): StatPearls Publishing [Google Scholar]
24. Brier ME, Bunke CM, Lathon PV, Aronoff GR. Erythropoietin-induced antinatriuresis mediated by angiotensin II in perfused kidneys. J Am Soc Nephrol. 1993;3(9):1583-90. [View at Publisher] [DOI] [PMID] [Google Scholar]
25. Geier MR, Geier DA. Respiratory conditions in coronavirus disease 2019 (COVID-19): Important considerations regarding novel treatment strategies to reduce mortality. Med Hypotheses. 2020;140:109760. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
26. Hui DS, E IA, Madani TA, Ntoumi F, Kock R, Dar O, et al. The continuing 2019-nCoV epidemic threat of novel coronaviruses to global health - The latest 2019 novel coronavirus outbreak in Wuhan, China. Int J Infect Dis. 2020;91:264-6. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
27. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, et al. Addendum: A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;588(7836):E6. [View at Publisher] [DOI] [PMID] [Google Scholar]
28. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front Immunol. 2020;11:827. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
29. Broxmeyer HE. Erythropoietin: multiple targets, actions, and modifying influences for biological and clinical consideration. J Exp Med. 2013;210(2):205-8. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
30. Genc S, Koroglu TF, Genc K. Erythropoietin and the nervous system. Brain Res. 2004;1000(1-2):19-31. [View at Publisher] [DOI] [PMID] [Google Scholar]
31. Sun Y, Zhou C, Polk P, Nanda A, Zhang JH. Mechanisms of erythropoietin-induced brain protection in neonatal hypoxia-ischemia rat model. J Cereb Blood Flow Metab. 2004;24(2):259-70. [View at Publisher] [DOI] [PMID] [Google Scholar]
32. Kuhrt D, Wojchowski DM. Emerging EPO and EPO receptor regulators and signal transducers. Blood. 2015;125(23):3536-41. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
33. Krantz SB. Erythropoietin. Blood. 1991;77(3):419-34. https://doi.org/10.1182/blood.V77.3.419.419 [View at Publisher] [DOI] [PMID] [Google Scholar]
34. Marti HH, Wenger RH, Rivas LA, Straumann U, Digicaylioglu M, Henn V, et al. Erythropoietin gene expression in human, monkey and murine brain. Eur J Neurosci. 1996;8(4):666-76. [View at Publisher] [DOI] [PMID] [Google Scholar]
35. Jelkmann W. Physiology and pharmacology of erythropoietin. Transfus Med Hemother. 2013;40(5):302-9. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
36. Wu H, Liu X, Jaenisch R, Lodish HF. Generation of committed erythroid BFU-E and CFU-E progenitors does not require erythropoietin or the erythropoietin receptor. Cell. 1995;83(1):59-67. [View at Publisher] [DOI] [Google Scholar]
37. Lin CS, Lim SK, D'Agati V, Costantini F. Differential effects of an erythropoietin receptor gene disruption on primitive and definitive erythropoiesis. Genes Dev. 1996;10(2):154-64. [View at Publisher] [DOI] [PMID] [Google Scholar]
38. Suresh S, Rajvanshi PK, Noguchi CT. The Many Facets of Erythropoietin Physiologic and Metabolic Response. Front Physiol. 2019;10:1534. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
39. Liu L, Damen JE, Cutler RL, Krystal G. Multiple cytokines stimulate the binding of a common 145-kilodalton protein to Shc at the Grb2 recognition site of Shc. Mol Cell Biol. 1994;14(10):6926-35. [View at Publisher] [DOI] [PMID] [PMCID]
40. Witthuhn BA, Quelle FW, Silvennoinen O, Yi T, Tang B, Miura O, et al. JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin. Cell. 1993;74(2):227-36. [View at Publisher] [DOI] [Google Scholar]
41. Juul SE, Yachnis AT, Christensen RD. Tissue distribution of erythropoietin and erythropoietin receptor in the developing human fetus. Early Hum Dev. 1998;52(3):235-49. [View at Publisher] [DOI] [Google Scholar]
42. Livnah O, Stura EA, Middleton SA, Johnson DL, Jolliffe LK, Wilson IA. Crystallographic evidence for preformed dimers of erythropoietin receptor before ligand activation. Science. 1999;283(5404):987-90. [View at Publisher] [DOI] [PMID] [Google Scholar]
43. Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, et al. SHED: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A. 2003;100(10):5807-12. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
44. Watowich SS. The erythropoietin receptor: molecular structure and hematopoietic signaling pathways. J Investig Med. 2011;59(7):1067-72. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
45. Jacobs K, Shoemaker C, Rudersdorf R, Neill SD, Kaufman RJ, Mufson A, et al. Isolation and characterization of genomic and cDNA clones of human erythropoietin. Nature. 1985;313(6005):806-10. [View at Publisher] [DOI] [PMID] [Google Scholar]
46. Erythropoietins, Erythropoietic Factors, and Erythropoiesis. 2 ed. Steven G. Elliott MF, Graham Molineux, editor. Birkhäuser Basel 2009: Birkhäuser Basel; 2009. XVIII, 330 p.
47. Egrie JC, Browne J, Lai P, Lin FK. Characterization of recombinant monkey and human erythropoietin. Prog Clin Biol Res. 1985;191:339-50. [View at Publisher] [Google Scholar]
48. Wei Y, Zhou J, Yu H, Jin X. AKT phosphorylation sites of Ser473 and Thr308 regulate AKT degradation. Biosci Biotechnol Biochem. 2019;83(3):429-35. [View at Publisher] [DOI] [PMID] [Google Scholar]
49. Thomas R. Gelzleichter. Chapter 7 - Early Characterization of Biosimilar Therapeutics. M. Plitnick DJH, editor. In : Lisa Nonclinical Development of Novel Biologics, Biosimilars, Vaccines and Specialty Biologics, Academic Press, ; 2013. [View at Publisher] [DOI] [Google Scholar]
50. Brines M, Cerami A. Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci. 2005;6(6):484-94. [View at Publisher] [DOI] [PMID] [Google Scholar]
51. Arcasoy MO. The non-haematopoietic biological effects of erythropoietin. Br J Haematol. 2008;141(1):14-31. [View at Publisher] [DOI] [PMID] [Google Scholar]
52. Alley W, Tao L, Shion H, Yu YQ, Rao C, Chen W. UPLC-MS assessment on the structural similarity of recombinant human erythropoietin (rhEPO) analogues from manufacturers in China for attribute monitoring. Talanta. 2020;220:121335. [View at Publisher] [DOI] [PMID] [Google Scholar]
53. Fisher JW. Landmark advances in the development of erythropoietin. Experimental Biology and Medicine. 2010 Dec;235(12):1398-411. [View at Publisher] [DOI] [PMID] [Google Scholar]
54. Jubinsky PT, Krijanovski OI, Nathan DG, Tavernier J, Sieff CA. The beta chain of the interleukin-3 receptor functionally associates with the erythropoietin receptor. Blood. 1997;90(5):1867-73. [View at Publisher] [DOI] [PMID] [Google Scholar]
55. Brines M, Cerami A. The receptor that tames the innate immune response. Mol Med. 2012;18(1):486-96. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
56. Anusornvongchai T, Nangaku M, Jao TM, Wu CH, Ishimoto Y, Maekawa H, et al. Palmitate deranges erythropoietin production via transcription factor ATF4 activation of unfolded protein response. Kidney Int. 2018;94(3):536-50. [View at Publisher] [DOI] [PMID] [Google Scholar]
57. Nairz M, Haschka D, Dichtl S, Sonnweber T, Schroll A, Aßhoff M, et al. Cibinetide dampens innate immune cell functions thus ameliorating the course of experimental colitis. Scientific Reports. 2017;7(1):13012. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
58. Erbayraktar S, Grasso G, Sfacteria A, Xie QW, Coleman T, Kreilgaard M, et al. Asialoerythropoietin is a nonerythropoietic cytokine with broad neuroprotective activity in vivo. Proc Natl Acad Sci U S A. 2003;100(11):6741-6. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
59. Fiordaliso F, Chimenti S, Staszewsky L, Bai A, Carlo E, Cuccovillo I, et al. A nonerythropoietic derivative of erythropoietin protects the myocardium from ischemia-reperfusion injury. Proc Natl Acad Sci U S A. 2005;102(6):2046-51. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
60. Bonnas C, Wüstefeld L, Winkler D, Kronstein-Wiedemann R, Dere E, Specht K, et al. EV-3, an endogenous human erythropoietin isoform with distinct functional relevance. Sci Rep. 2017;7(1):3684. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
61. Robertson CS, Garcia R, Gaddam SSK, Grill RJ, Cerami Hand C, Tian TS, et al. Treatment of mild traumatic brain injury with an erythropoietin-mimetic peptide. J Neurotrauma. 2013;30(9):765-74. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
62. Simon F, Floros N, Ibing W, Schelzig H, Knapsis A. Neurotherapeutic potential of erythropoietin after ischemic injury of the central nervous system. Neural Regen Res. 2019;14(8):1309-12. [DOI] [PMID] [PMCID] [Google Scholar]
63. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020;30(3):269-71. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
64. MacRedmond R, Singhera GK, Dorscheid DR. Erythropoietin inhibits respiratory epithelial cell apoptosis in a model of acute lung injury. Eur Respir J. 2009;33(6):1403-14. [View at Publisher] [DOI] [PMID] [Google Scholar]
65. Hu H, Ma F, Wei X, Fang Y. Coronavirus fulminant myocarditis treated with glucocorticoid and human immunoglobulin. Eur Heart J. 2021;42(2):206. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
66. Olsen NV, Aachmann-Andersen N-J, Oturai P, Munch-Andersen T, Bornø A, Hulston C, et al. Erythropoietin down-regulates proximal renal tubular reabsorption and causes a fall in glomerular filtration rate in humans. J Physiol. 2011;589(Pt 6):1273-81. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
67. Al-Samkari H, Karp Leaf RS, Dzik WH, Carlson JCT, Fogerty AE, Waheed A, et al. COVID-19 and coagulation: bleeding and thrombotic manifestations of SARS-CoV-2 infection. Blood. 2020;136(4):489-500. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
68. Sahebnasagh A, Mojtahedzadeh M, Najmeddin F, Najafi A, Safdari M, Rezai Ghaleno H, et al. A Perspective on Erythropoietin as a Potential Adjuvant Therapy for Acute Lung Injury/Acute Respiratory Distress Syndrome in Patients with COVID-19. Arch Med Res. 2020;51(7):631-5. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
69. Shimaoka M, Park EJ. Advances in understanding sepsis. Eur J Anaesthesiol Suppl. 2008;42:146-53. [DOI] [PMID] [PMCID]
70. Fishbane S, Hirsch JS. Erythropoiesis-Stimulating Agent Treatment in Patients With COVID-19. Am J Kidney Dis. 2020;76(3):303-5. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
71. Leventhal J, Angeletti A, Cravedi P. EPO in Patients With COVID-19: More Than an Erythropoietic Hormone. Am J Kidney Dis. 2020;76(3):441. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
72. Farmer S, Horváth-Puhó E, Vestergaard H, Hermann AP, Frederiksen H. Chronic myeloproliferative neoplasms and risk of osteoporotic fractures; a nationwide population-based cohort study. Br J Haematol. 2013;163(5):603-10. [View at Publisher] [DOI] [PMID] [Google Scholar]
73. Sukhomlin T. Could an acute respiratory distress syndrome in COVID-19 infected patients be calmed down simply by iron withdrawal from lung tissues? J Med Virol. 2021;93(2):577-8. [View at Publisher] [DOI] [PMID] [Google Scholar]
74. Guo T, Fan Y, Chen M, Wu X, Zhang L, He T, et al. Cardiovascular Implications of Fatal Outcomes of Patients With Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020;5(7):811-8. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
75. Tang YD, Rinder HM, Katz SD. Effects of recombinant human erythropoietin on antiplatelet action of aspirin and clopidogrel in healthy subjects: results of a double-blind, placebo-controlled randomized trial. Am Heart J. 2007;154(3):494.e1-7. [View at Publisher] [DOI] [PMID] [Google Scholar]
76. Sarkisian L, Saaby L, Poulsen TS, Gerke O, Jangaard N, Hosbond S, et al. Clinical Characteristics and Outcomes of Patients with Myocardial Infarction, Myocardial Injury, and Nonelevated Troponins. Am J Med. 2016;129(4):446.e5-.e21. [View at Publisher] [DOI] [PMID] [Google Scholar]
77. Tavazzi G, Pellegrini C, Maurelli M, Belliato M, Sciutti F, Bottazzi A, et al. Myocardial localization of coronavirus in COVID-19 cardiogenic shock. Eur J Heart Fail. 2020;22(5):911-5. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
78. Puntmann VO, Carerj ML, Wieters I, Fahim M, Arendt C, Hoffmann J, et al. Outcomes of Cardiovascular Magnetic Resonance Imaging in Patients Recently Recovered From Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020;5(11):1265-73. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
79. Heitrich M, García DM, Stoyanoff TR, Rodríguez JP, Todaro JS, Aguirre MV. Erythropoietin attenuates renal and pulmonary injury in polymicrobial induced-sepsis through EPO-R, VEGF and VEGF-R2 modulation. Biomed Pharmacother. 2016;82:606-13. [View at Publisher] [DOI] [PMID] [Google Scholar]
80. Du Y, Tu L, Zhu P, Mu M, Wang R, Yang P, et al. Clinical Features of 85 Fatal Cases of COVID-19 from Wuhan. A Retrospective Observational Study. Am J Respir Crit Care Med. 2020;201(11):1372-9. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
81. Bikdeli B, Madhavan MV, Jimenez D, Chuich T, Dreyfus I, Driggin E, et al. COVID-19 and Thrombotic or Thromboembolic Disease: Implications for Prevention, Antithrombotic Therapy, and Follow-Up: JACC State-of-the-Art Review. J Am Coll Cardiol. 2020;75(23):2950-73. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
82. Kakavas S, Demestiha T, Vasileiou P, Xanthos T. Erythropoetin as a novel agent with pleiotropic effects against acute lung injury. Eur J Clin Pharmacol. 2011;67(1):1-9. [View at Publisher] [DOI] [PMID] [Google Scholar]
83. Lundby C, Olsen NV. Effects of recombinant human erythropoietin in normal humans. J Physiol. 2011;589(Pt 6):1265-71. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
84. Uversky VN, Redwan EM. Erythropoietin and co.: intrinsic structure and functional disorder. Mol Biosyst. 2016;13(1):56-72. [View at Publisher] [DOI] [PMID] [Google Scholar]
85. Masuda S, Okano M, Yamagishi K, Nagao M, Ueda M, Sasaki R. A novel site of erythropoietin production. Oxygen-dependent production in cultured rat astrocytes. J Biol Chem. 1994;269(30):19488-93. [View at Publisher] [DOI] [Google Scholar]
86. Zhang X, Dong S, Qin Y, Bian X. Protective effect of erythropoietin against myocardial injury in rats with sepsis and its underlying mechanisms. Mol Med Rep. 2015;11(5):3317-29. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
87. Lechuga GC, Souza-Silva F, Sacramento CQ, Trugilho MRO, Valente RH, Napoleão-Pêgo P, et al. SARS-CoV-2 Proteins Bind to Hemoglobin and Its Metabolites. Int J Mol Sci. 2021;22(16):9035. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
88. Pugh CW, Ratcliffe PJ. New horizons in hypoxia signaling pathways. Exp Cell Res. 2017;356(2):116-21. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
89. Khoo A, McLoughlin B, Cheema S, Weil RS, Lambert C, Manji H, et al. Postinfectious brainstem encephalitis associated with SARS-CoV-2. J Neurol Neurosurg Psychiatry. 2020;91(9):1013-4. [View at Publisher] [DOI] [PMID] [Google Scholar]
90. Luo H-C, You C-Y, Lu S-W, Fu Y-Q. Characteristics of coagulation alteration in patients with COVID-19. Ann Hematol. 2021;100(1):45-52. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
91. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46(6):1089-98. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
92. Elliott S, Pham E, Macdougall IC. Erythropoietins: a common mechanism of action. Exp Hematol. 2008;36(12):1573-84. [View at Publisher] [DOI] [PMID] [Google Scholar]
93. Liongue C, Sertori R, Ward AC. Evolution of Cytokine Receptor Signaling. J Immunol. 2016;197(1):11-8. [View at Publisher] [DOI] [PMID] [Google Scholar]
94. Bazan JF. Structural design and molecular evolution of a cytokine receptor superfamily. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(18):6934-8. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]
95. Miura Y, Miura O, Ihle JN, Aoki N. Activation of the mitogen-activated protein kinase pathway by the erythropoietin receptor. J Biol Chem. 1994;269(47):29962-9. [View at Publisher] [DOI] [Google Scholar]
96. Brines M, Grasso G, Fiordaliso F, Sfacteria A, Ghezzi P, Fratelli M, et al. Erythropoietin mediates tissue protection through an erythropoietin and common beta-subunit heteroreceptor. Proc Natl Acad Sci U S A. 2004;101(41):14907-12. [View at Publisher] [DOI] [PMID] [PMCID] [Google Scholar]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 4.0 | Journal of Clinical and Basic Research

Designed & Developed by : Yektaweb

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0).