Научная статья на тему 'Pleiotropic effects of erythropoietin. Influence of erythropoietin on processes of mesenchymal stem cells differentiation'

Pleiotropic effects of erythropoietin. Influence of erythropoietin on processes of mesenchymal stem cells differentiation Текст научной статьи по специальности «Биотехнологии в медицине»

CC BY
173
35
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
ANGIOGENESIS / ERYTHROPOIETIN / MESENCHYMAL STEM CELLS / OSTEOGENESIS

Аннотация научной статьи по биотехнологиям в медицине, автор научной работы — Zubareva Ekaterina V., Nadezhdin Sergey V., Burda Yuriy E., Nadezhdina Natalia A., Gashevskaya Anastasia S.

Structure and synthesis of Erythropoietin: Erythropoietin (EPO) is a glycoprotein hormone. Recombinant Erythropoietin (Epoetin): Human recombinant erythropoietin is characterised as a factor which stimulates differentiation and proliferation of erythroid precursor cells, and as a tissue protective factor. Anti-ischemic effects of recombinant Erythropoietin: Erythropoietin is one of the most perspective humoral agents which are involved in the preconditioning phenomenon. Erythropoietin receptors and signal transduction pathways: Erythropoietin effects on cells through their interconnection with erythropoietin receptors, which triggers complex intracellular signal cascades, such as JAK2/STAT signaling pathway, phosphatidylinositol 3-kinase (PI3K), protein kinase C, mitogen-activated protein kinase (MAPK), and nuclear factor (NF)-kB signaling pathways. Mechanisms of the effect of Erythropoietin on hematopoietic and non-hematopoietic cells and tissues: In addition to regulation of haemopoiesis, erythropoietin mediates bone formation as it has an effect on hematopoietic stem cells and osteoblastic niche, and this illustrates connection between the processes of haematopoiesis and osteopoiesis which take place in the red bone marrow. The effect of Erythropoietin on mesenchymal stem cells and process of bone tissue formation: Erythropoietin promotes mesenchymal stem cells proliferation, migration and differentiation in osteogenic direction. The evidence of which is expression of bone phenotype by cells under the influence of EPO, including activation of bone specific transcription factors Runx2, osteocalcin and bone sialoprotein. Conclusion: Erythropoietin has a pleiotropic effect on various types of cells and tissues. But the mechanisms which are involved in the process of bone tissue restoration via erythropoietin are still poorly understood.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «Pleiotropic effects of erythropoietin. Influence of erythropoietin on processes of mesenchymal stem cells differentiation»

# Research Results in Pharmacology

Research Results in Pharmacology 5(1): 53-66

UDC 615.275 DOI 10.3897/rrpharmacology. 5.33457

3 Review Article

Pleiotropic effects of Erythropoietin. Influence of Erythropoietin on processes of mesenchymal stem cells differentiation

Ekaterina V. Zubareva1, Sergey V. Nadezhdin1, Yuriy E. Burda2, Natalia A. Nadezhdina3, Anastasia S. Gashevskaya2

1 Belgorod State National Research University, 85 Pobedy St., Belgorod 308015, Russia

2 EFKO, 20 Ovchinnikovskaya Embankment, Bild. 1, Moscow 115035, Russia

3 Regional State Budgetary Healthcare Institution "Children's Regional Clinical Hospital" 44 Gubkin St., Belgorod 308036, Russia Corresponding author: Ekaterina V. Zubareva (zubareva@bsu.edu.ru)

Academic editor: Mikhail Korokin ♦ Received 29 December 2018 ♦ Accepted 21 February 2019 ♦ Published 28 March 2019

Citation: Zubareva EV, Nadezhdin SV, Burda YE, Nadezhdina NA, Gashevskaya AS (2019) Pleiotropic effects of Erythropoietin. Influence of Erythropoietin on processes of mesenchymal stem cells differentiation. Research Results in Pharmacology 5(1): 53-66. https://doi.org/10.3897/rrpharmacology.5.33457

Abstract

Structure and synthesis of Erythropoietin: Erythropoietin (EPO) is a glycoprotein hormone.

Recombinant Erythropoietin (Epoetin): Human recombinant erythropoietin is characterised as a factor which stimulates differentiation and proliferation of erythroid precursor cells, and as a tissue protective factor.

Anti-ischemic effects of recombinant Erythropoietin: Erythropoietin is one of the most perspective humoral agents which are involved in the preconditioning phenomenon.

Erythropoietin receptors and signal transduction pathways: Erythropoietin effects on cells through their interconnection with erythropoietin receptors, which triggers complex intracellular signal cascades, such as JAK2/STAT signaling pathway, phosphatidylinositol 3-kinase (PI3K), protein kinase C, mitogen-activated protein kinase (MAPK), and nuclear factor (NF)-kB signaling pathways.

Mechanisms of the effect of Erythropoietin on hematopoietic and non-hematopoietic cells and tissues: In addition to regulation of haemopoiesis, erythropoietin mediates bone formation as it has an effect on hematopoietic stem cells and osteoblastic niche, and this illustrates connection between the processes of haematopoiesis and osteopoiesis which take place in the red bone marrow.

The effect of Erythropoietin on mesenchymal stem cells and process of bone tissue formation: Erythropoietin promotes mesenchymal stem cells proliferation, migration and differentiation in osteogenic direction. The evidence of which is expression of bone phenotype by cells under the influence of EPO, including activation of bone specific transcription factors Runx2, osteocalcin and bone sialoprotein.

Conclusion: Erythropoietin has a pleiotropic effect on various types of cells and tissues. But the mechanisms which are involved in the process of bone tissue restoration via erythropoietin are still poorly understood.

Keywords

angiogenesis, erythropoietin, mesenchymal stem cells, osteogenesis.

Copyright Zubareva EVetal. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Structure and synthesis of Erythropoietin

Erythropoietin (EPO) is a glycoprotein hormone which belongs to type I Cytokine Superfamily (Coleman and Brines 2004). EPO is most similar to cytokines, which modulate growth and inflammation (Erbayraktar et al. 2003). EPO is made up of about 60% protein and 40% carbohydrate (Rolfing 2014). Molecular weight of the EPO is 30.4 kDa (Jelkmann 2013).

The feature of erythropoietin is its multifunctionality because, depending on conditions, this protein can act as cytokine, hormone and growth factor. Erythropoietin realizes its cytokine effect through its interaction with specific receptors located on the surface of the target cells, which leads to the signal transduction from external to internal medium of the target cells. Erythropoietin acts as a classic hormone because it is synthesized in one organ (interstitial cells of the peritubular capillary bed of the renal cortex in adults or perisinusoidal cells of the liver during the embryonic and perinatal period), then it is released into the blood flow and effects on remote structures, including the hematopoietic system (red blood cells progenitors and precursors found in human bone marrow). And, finally, as a growth factor, erythropoietin is essential for mature red blood cells formation from multipotent progenitor cells (Uversky and Redwan 2017).

The gene of human EPO codes the precursor protein which includes 193 aminoacidic residues (Lappin et al. 2002, Sasaki et al. 2000, Vazquez-Mellado et al. 2017). Removal of 27-aminoacid sequence results in formation of mature protein, which is subjected to the N-glycosyl-ation at three aspartic residues, O-glycosylation at serine residue; C-terminal arginine is removed to create final circulating form which includes 165 aminoacidic residues. Thus, mature EPO is heavily glycosylated (Jelkmann 2007, Ridley et al. 1994, Vazquez-Mellado et al. 2017), has three N-linked and one O-linked acidic oligosac-charide side chains (Jelkmann 2013, Ridley et al. 1994, Vazquez-Mellado et al. 2017). Four anti-parallel a-helix-es (aA, aB, aC and aD in order from N-terminal to C-ter-minal) with adjacent cycles determine spatial tertiary structure of EPO (Desai 2012, Lappin et al. 2002). Glycosylated chains maintain the biological activity of EPO and ensure stability of the structure (Maiese et al. 2008, Toyoda et al. 2000, Vazquez-Mellado et al. 2017). The EPO molecule is hydrophobic and requires presence of the disulphide bridges for its activity (Ridley et al. 1994).

EPO production depends on the transcription rate of the EPO gene (located in the 7th chromosome). The process of EPO gene transcription involves few transcription factors. The EPO gene promoter is inhibited by GATA-2 and nucleus factor kB (NF-kB), which probably are responsible for a decrease in EPO expression during inflammatory diseases (Bunn et al. 1998, La Ferla et al. 2002, Jelkmann 2013).

Kidneys are the primary site of EPO production. During hypoxia, the EPO gene expression increases in the

kidney peritubular fibroblasts via hypoxia-induced transcription factor, and EPO is subsequently released into the circulation (Rolfing 2014). Before the birth, the liver is a primary site of EPO production, in adults approximately 10% of EPO is synthesized by the liver (Dame et al. 1998, Jelkmann 2007, Rankin et al. 2007, Rolfing 2014, Zanjani et al. 1977). Predominant expression of EPO occurs in kidneys and liver, though transcripts can be also found in other tissues, such as brain, heart, and lungs (Elliott and Sinclair 2012).

Recombinant Erythropoietin (Epoetin)

Biotechnological achievements have made it possible to clone and produce a great amount of human recombinant erythropoietin (rhEPO) (Flaharty et al. 1990) and its analogues (erythropoiesis-stimulating agents (ESAs)) (Elliot and Sinclair 2012, Jacobs et al. 1985, Rolfing 2014). Endogenous erythropoietin and recombinant erythropoietin are identical, except for some minor differences in glyco-sylation (Koury and Bondurant 1992, Rolfing 2014). Human recombinant erythropoietin was first characterised as a hematopoietic factor (Ye et al. 2010). It was shown that this substance stimulated differentiation and proliferation of erythroid precursor cells (Broudy et al. 1991, Ye et al. 2010). Recombinant forms of erythropoietin are epoetin-alfa (human erythropoietin which is produced in cell culture using recombinant DNA technology), epoe-tin-beta (which is a synthetic recombinant form of EPO) and long-term acting analogue darbepoetin-alfa (which is also a synthetic form of erythropoietin) (Arcasoy 2008, Uversky and Redwan 2017). Use of recombinant forms of erythropoietin was characterised to treat anaemia during pregnancy (Sanchez-Gonzalez et al. 2016), chronic kidney disease (Togel et al. 2016) or chemotherapy-induced anaemia during oncological diseases (Arcasoy 2008, Elliot and Sinclair 2012, Suk et al. 2008).

Ubiquitous expression of erythropoietin receptors on non-erythroid cells is the evidence that the erythropoietin plays a lot of biologically important roles in non-hemato-poietic tissues (Arcasoy 2008, Arcasoy 2010).

Recombinant erythropoietin was characterized as a tissue protective factor (Elliot and Sinclair 2012, Rocha et al. 2015, Vinberg et al. 2015). Protective effect of eryth-ropoietin is dose-dependent and receptor-dependent. This circumstance has resulted in pharmacological synthesis of various types of erythropoietin: Asialoerythropoietin has half-life under 2 minutes (whereas for erythropoie-tin this index is approximately 5-10 hours); Carbam-yl-erythropoietin (CEPO) is characterised by presence of a few NH2 groups. It was shown that CEPO has bigger protective effect in such injuries, as spinal cord compression, diabetic neuropathy or experimental autoimmune encephalitis without the erythropoiesis stimulation effect (Buemi et al. 2009).

There were identified some properties of recombinant erythropoietin in regulation of genes expression and their function similar to vascular endothelial growth factor (VEGF), main angiogenic factor (Suk et al. 2008). It is known that erythropoietin is able to stimulate proliferation of endothelial cells in vitro and mobilize endothelial progenitor cells from bone marrow (Bahlmann et al. 2003, Bahlmann et al. 2004, Heeschen et al. 2003, Klopsch et al. 2009), increase adhesive and proliferative characteristics of circulating endothelial progenitor cells (George et al. 2005, Klopsch et al. 2009) and induce neovascularization of ischemic tissue in vivo (Ye et al. 2010). Under the influence of erythropoietin, embryonic stem cells can differentiate into cardiomyocytes; erythropoietin also facilitates proliferation of cardiomyocytes (Broudy et al. 1991, Sachinidis et al. 2002, Ye et al. 2010). Erythropoietin is able to influence differentiation of neuronal stem cells into neuronal cells (Chen et al. 2009, Kretz et al. 2005, Ye et al. 2010).

Anti-ischemic effects of recombinant Erythropoietin

In a pharmacological aspect, erythropoietin is one of the most perspective humoral agents which are involved in the preconditioning phenomenon. A number of studies showed that erythropoietin has a protective effect on ischemia-reperfusion in various organs and tissues (Da-nilenko 2013). The opportunity of using recombinant erythropoietin in suberythropoietic doses is justified for pharmacological preconditioning; influence of recombinant erythropoietin on ischemic tissues survival was revealed and it was concluded that this effect was similar to that from remote preconditioning (Kolesnik et al. 2010). Recombinant human erythropoietin is of increased interest in regard to pharmaceutical simulation of ischemic preconditioning because it proved its efficacy in protection of heart, brain and liver tissues from pathological influence of ischemia-reperfusion (Alehin et al. 2015).

Erythropoietin is highly effective in prevention and treatment of ischemic-reperfusion-induced acute kidney injury. Derivatives of erythropoietin (asialated erythro-poietin, carbamylated darbepoetin) have nephroprotec-tive effects (Elagin et al. 2018).

Recombinant erythropoietin was found to stimulate neovasculogenesis in ischemic muscle (Kolesnik et al. 2011, Pokrovskiy et al. 2012).

A possibility of pharmacological correction of ischemia and reperfusion injuries of small intestine and liver tissues with recombinant erythropoietin in a suberyth-rostimulating dose in rats was investigated. It was shown that injection of recombinant erythropoietin in a suberyth-rostimulating dose in rats had a significant effect on microcirculation recovery both in liver and small intestine tissues. Erythropoietin is considered as a preconditioning agent which realizes its effect via the early preconditioning mechanisms (Alekhin et al. 2011).

Using of recombinant erythropoietin Darbepoetin alpha is characterized by development of endothelium- and cardioprotective effect (Denisyuk et al. 2015, Denisyuk and Pokrovsky 2016, Korokina et al. 2009). In the experiments on animals' (rats') hearts, it was shown that application of erythropoietin led to improved coronary perfusion. This effect is caused by activation of endothelial NO synthase and Akt (Protein kinase B) activating phosphorylation, which leads to long NO-dependent vasodilatation. Some evidence was obtained that recombinant erythropoietin is characterized by a protective effect in L-NAME-induced endothelial disfunction and ischemia of limbs, abdominal organs and retina (Denisyuk et al. 2015).

An independent cardioprotective effect of erythropoi-etin was proven, as well as a decrease in the myocardial infarction zone and improvement of heart function in is-chemia-reperfusion, irrespective of changes in haemoglobin, red blood cell count and oxygen tension in blood. Erythropoietin is a trigger of ischemic preconditioning and may cause pharmacological preconditioning; realisation of natural mechanisms of protection from ischemia is carried out through the activation of ATP-dependent potassium channels and biosynthesis of nitric oxide, pharmacological mimetics of preconditioning which induce a reproducible and stable cardioprotective reply and which lack any serious side effects (Danilenko 2013). Some studies show that decreased apoptosis is one of the possible most important mechanism of erythropoietin's protective effect (Danilenko et al. 2012, Danilenko 2013, Denisyuk and Pokrovsky 2016, Denisyuk 2016).

Using recombinant erythropoietin leads to a significant increase in anti-inflammatory effect (Danilenko et al. 2011).

Erythropoietin receptors and signal transduction pathways

Erythropoietin is known to bind to membrane receptors (EPOR) localized on the surface of the target cells (Jelk-mann 2007). EPOR belongs to Class I of Cytokine super-family receptors, which are characterized by the presence of extracellular N-terminal sequence. Mature human EPOR is a glycoprotein which includes 484 aminoacidic residues and 1 N-glycan. Molecular weight of human EPOR is 52.6 kDa, and because of glycosylation and phosphorylation it increases to 60 kDa (Jelkmann 2007).

In hematopoietic cells, two molecules of erythropoie-tin receptor (EPOR) form a homodimer (EPOR/EPOR) with high affinity to its ligand erythropoietin (Rolfing et al. 2014). Binding with erythropoietin induces conformational changes and leads to tight conjunction of two monomer EPOR molecules. As a result, Janus kinases 2 (JAK2) molecules (Witthuhn et al. 1994), which contact with EPOR in the juxtamembrane domain, converge and subsequently transphosphorylate (Elliott and Sinclair 2012). Activation of JAK2 leads to phosphorylation of a few EPOR tyrosine residues, which serve as sites for

signal molecules binding, such as Src homology-2 (SH2) domain-containing proteins and signal transducer, and activator of transcription (STAT) 1, 3 and 5 (Carter-Su et al. 2016, Kirito et al. 2002, Klingmuller et al. 1996, Kwon et al. 2014, Li et al. 2015, Ma et al. 2016, Rafiee et al. 2005, Vazquez-Mellado et al. 2017, van der Kooij et al. 2008, Ye et al. 2010). Activated JAK2 also induces signaling pathways via nuclear factor kappa B (NF-kB) (Ma et al. 2016). Erythropoietin is known to be the main activating factor of JAK2/STAT3 signaling pathway, which is confirmed to be launched in blood diseases and cardiac cerebral vascular diseases (Chen et al. 2009, Piuhola et al. 2008, Ye et al. 2010). When STAT3 signaling transduction pathway is activated, the activated STAT3 transfers the signal directly into the nucleus and regulates multiple genes (Ye et al. 2010). STAT5 and NF-kB translocate into the nucleus and act as transcription factors for Bcl-2 (Correia et al. 2015, Ma et al. 2016) and Bcl-xL (Ma et al. 2016, Schwartz et al. 2015), which are antiapoptotic genes (Ma et al. 2016). In addition to activation of JAK/ STAT pathway, erythropoietin also stimulates other signaling pathways, including phosphatidylinositol-3 kinase/ Akt (PI3K/Akt) pathway, mitogen-activated protein ki-nase/extracellular signal-related kinase (MAPK/ ERK1/2) pathway and protein kinase C, ensuring the survival of the cell and its proliferation (Bao et al. 1999, Damen et al. 1995, Hiram-Bab et al. 2015, Hiram-Bab et al. 2017, Kwon et al. 2014, Vazquez-Mellado et al. 2017). The effect of erythropoietin is terminated by the influence of haematopoietic cell phosphatase (HCP) which catalyzes JAK2 dephosphorylation (Jelkmann 2007).

Unlike hematopoietic cells, in non-hematopoietic cells erythropoietin carries out its functions through the interaction with a heterodimeric receptor (EPOR/CD131) (Rolf-ing et al. 2014). In comparison with homodimeric EPOR, which is present in the hematopoietic system, the heterod-imeric receptor is characterised by lower affinity to its li-gand, erythropoietin. Due to this, for erythropoietin to realize its protective effect towards tissues and other pleiotropic effects, a bigger concentration of the hormone is required than during erythropoiesis stimulation (Rolfing 2014).

It is known that on the surfaces of the cells which are sensitive to erythropoietin, EPOR and a common receptor subunit p (PcR), also known as CD131, are co-expressed. The common receptor subunit p provides for an increase in the binding affinity of the ligand to the receptor complex and is also a signal-transducing component common to the granulocyte-macrophage colony stimulating factor (GM-CSF), IL-3 and IL-5 receptors. Furthermore, tissue protection is known to be realized through this heteroreceptor complex containing EPOR and PcR (Brines et al. 2004).

Non-hematopoietic intracellular signaling pathways include production of nitrogen oxide and signaling through JAK2, STAT3/5, PI3K/Akt, as well as MAPK pathways (Rolfing et al. 2014). Some functions are activated when specific pathways are activated, whereas for erythropoietin to exercise its full pleiotropic effect, all signalling intracel-lular pathways need to be activated (Rolfing et al. 2014).

Mechanisms of effect of Erythropoietin on hematopoietic and non-hematopoietic cells and tissues

The effect of Erythropoietin on hematopoietic tissues

Erythropoietin was first identified as a hormone whose main function was to stimulate differentiation of erythroid cell precursors in bone marrow into functional erythro-blasts (Coleman and Brines 2004, Debeljak et al. 2014, Flaharty et al. 1990, Ganz 2018, Perreault and Venters 2018, Ridley et al. 1994, Tojo et al. 2015), which makes it possible to regulate delivery of oxygen to the tissues (Semenza and Wang 1992). Erythropoietin increases the number of developing erythroid precursors and accelerates release of reticulocytes from bone marrow, but does not influence on the duration of the cell cycle or the number of meioses which take place during the cell differentiation (Ridley et al. 1994). Erythropoietin increases the pool of cells capable of differentiating into erythroid cells. It is an important hormone which determines differentiation of cells in the erythroid direction, interacts with early-stage (burst forming unit-erythroid, BFU-E) and late-stage (colony forming erythroid unit, CFU-E) erythroid precursors, cells committed to erythroid differentiation. Erythropoietin apparently stimulates proliferation of proerythroblasts and basophilic erythroblasts, which are the first cells to be morphologically recognized as erythroid elements; however, erythropoietin is not an obligatory differentiation growth factor in the erythroid direction at this stage (Ridley et al. 1994). Regulation of red blood cells production is mediated via presence of a specific cellular receptor for erythropoietin on the surface of cells (Arcasoy 2008).

The effect of Erythropoietin on non-hematopoietic tissues

For a long period of time, there was an opinion that ery-thropoietin influenced only on erythroid precursor cells (Lappin et al. 2002). At present, it is known that erythro-poietin is a pleiotropic growth factor and, besides its classic role in facilitating erythropoiesis, this hormone carries out a number of biological functions (Wang et al. 2018), including activity towards multiple cells and tissues (Coleman and Brines 2004, Debeljak et al. 2014, Holstein et al. 2011). This hormone is able to stimulate bone tissue recovery after damage (Holstein et al. 2007, Holstein et al. 2011, Garcia et al. 2011, Klontzas et al. 2016), wound healing due to erythropoietin-mediated stimulation of cell proliferation and angiogenesis (Gupta and Wish 2017) which is related to increased expression of vascular en-dothelial growth factor (VEGF), endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) (Holstein et al. 2011). Erythropoietin is known to have a neuroprotective effect (Bramlett et al. 2016, Juul

and Pet 2015, Liu et al. 2017, Marelli et al. 2016, Modrak et al. 2017, Ott et al. 2015) and to provide liver regeneration (Kedarisetty et al. 2015).

Expression of erythropoietin was found in multiple tissues and types of cells outside the kidney tissue, including astrocytes, neurones, female genital tracts, male genitals, mammary glands, placenta trophoblasts, bone marrow macrophages and erythroid precursors (Arcasoy 2010, Bernaudin et al. 1999, Conrad et al. 1996, Juul et al. 2000, Kobayashi et al. 2002, Magnanti et al. 2001, Marti et al. 1996, Masuda et al. 2000, Sato et al. 2000, Stopka et al. 1998, Vogt et al. 1989). mRNA of erythropoietin was also revealed in liver, spleen, lungs; besides, erythropoi-etin could be produced in small amounts in these organs (Jelkmann 2013). The research results showed that the receptors for erythropoietin are expressed on various cells of the fetus and the adult organism, including cells of central nervous system, digestive canal, kidney, muscle tissue (smooth, skeletal and cardiac), uterus, retina, pancreas, gonads and lungs (Ammarguellat et al. 1996, Anagnostou et al. 1994, Arcasoy 2010, Erbayraktar et al. 2003, Grimm et al. 2002, Liu et al. 1997, Ogilvie et al. 2000, Shiozawa et al. 2010, Westenfelder et al. 1999, Wu et al. 1999). Due to this, erythropoietin can influence non-hematopoietic cells and tissues (Debeljak et al. 2014).

Thus, it was revealed that erythropoietin, in addition to its basic role in the regulation of erythropoiesis in mammals, influences proliferation and apoptosis of various non-hematopoietic cells through the erythropoietin receptors (Arcasoy 2008). The characteristic of non-erythropoi-etic biological effects of erythropoietin and understanding the mechanisms of signal activation of the erythropoietin/ erythropoietin receptor systems in non-hematopoietic organs and types of cells are important for future development of new methods of applying erythropoietin and its derivatives (Arcasoy 2008).

The effect of Erythropoietin on mesenchymal stem cells and process of bone tissue formation

Mesenchymal stem cells (MSCs) are primary multipotent stem cells with an ability of self-renewal and differentiation into multiple cell lines. MSCs have fibroblast-li-ke morphology and express stromal markers, including CD73, CD105, CD29, CD44 and CD90, but not hematopoietic cell markers (such as CD34, CD44 and CD45) (Chen et al. 2013, Ye et al. 2010).

Bone-marrow-derived mesenchymal stem cells are suitable for transplantation and are widely used in tissue engineering and cellular therapy because they are characterised by simple selection and obtaining, multipotent potential, immunosuppression and stability during autologous transplantation (Li et al. 2017, Rennert et al. 2012, Ye et al. 2010).

However, the population of MSCs in bone marrow is not large (about 0.001-0.01% of all nuclear cells), thus it

is necessary to increase the number of MSCs in culture for subsequent use during scientific experiments or clinical application (Ye et al. 2010). Whereas some studies showed that MSCs tended to lose pluripotency and proliferative capacity when cultivated in vitro (Banfi et al. 2000, Digirolamo et al. 1999, Wagner et al. 2008, Ye et al. 2010), MSCs are also known to be capable of spontaneous differentiation during in vitro passaging in the absence of special inducing factors (Deng et al. 2006, Sa-chinidis et al. 2002, Ye et al. 2010). In this way, instability of cultured MSCs and their decreased sternness limit their wide application in clinic (Ye et al. 2010).

The indicator of undifferentiated state and multiple differentiation potential of MSCs is the mRNA expression level of embryonic transcription factors OCT4, Nanog, Sox2 and TERT (Boyer et al. 2005, Greco et al. 2007, Loh et al. 2006, Wang et al. 2006, Ye et al. 2010). It was shown that erythropoietin regulates the mRNA expression of TERT, OCT4, Nanog, Sox2 genes (Ye et al. 2010, Saei Arezoumand et al. 2017), which are linked with a cell differentiation delay. In this case, the effect of erythropoietin depends on duration of its action and concentration. It is proved that erythropoietin could maintain MSCs in undifferentiated state for 12 hours at a concentration of 5 U/ml (Ye et al. 2010).

Erythropoietin is also known to promote MSCs proliferation (Minguell et al. 2001, Ye et al. 2010, Zheng et al. 2009). Thus, influencing proliferation, erythropoietin is able to increase the number of MSCs while preserving their multiple differentiation potential, to improve MSCs expansion in cell culture and to maintain culture in vitro (Ye et al. 2010).

Despite the achievements in transplantation of bone-marrow-derived mesenchymal stem cells, only a tiny fraction of transplanted cells migrates to the target sites. This is why it is vital to develop a strategy of more effective delivery of MSCs to appropriate structures (Ye et al. 2010). It is known that erythropoietin activates a directed migration of bone-marrow-derived mesenchymal stem cells into the organs which were damaged, for example, kidneys and heart (Lin et al. 2008, Liu et al. 2013, Vazquez-Mellado et al. 2017). Erythropoietin promotes mobilization of bone-marrow-derived mesenchymal stem cells by increasing the stromal cell-derived factor-1 (SDF-1) chemokine levels (Satoh et al. 2006, Vazquez-Mellado et al. 2017).

In addition, the important problem of applying MSCs in tissue engineering is their low viability and functionality, as well as their potential capacity to differentiate after transplantation. The majority of MSCs undergo apoptosis after transplantation. This is why improving MSCs viability and functionality is necessary to enhance their potential application efficiency. A few strategies aiming at improving the ability of MSCs to form tissues have been studied, including those with applying growth factors, overexpression of regulatory genes by stem cells and with improving scaffolds biomaterials. All the approaches used were connected with activation of PI3K/Akt signaling pathway. This pathway plays key regulatory roles in MSCs survival, proliferation, migration, angiogenesis, cytokine production and differentiation processes (Chen et al. 2013).

The effect of Erythropoietin on osteogenic differentiation of MSCs

MSCs are able to differentiate into the osseous, cartilaginous and adipose tissue lineages after stimulation in certain conditions. When being cultivated in the presence of glucocorticoid dexamethasone, ascorbic acid and be-ta-glycerophosphate (factors of osteogenic differentiation), MSCs differentiate in osteogenic direction and form bone-like nodules with mineralized extracellular matrix containing hydroxyapatite. One of the potential pathways of signaling transduction which can participate in regulation of proliferation and differentiation of MSCs in os-teogenic direction is MAP kinase pathway. Members of MAP kinase family are extracellular signal-regulated ki-nases (ERK1 and ERK2), c-Jun N-terminal kinase (JNK), which is also known as stress-activated protein kinase, and p38-reactivating kinase (p38 RK or simply p38). Transforming growth factor-beta stimulates ERK and p38 and acts as a physiological regulator of osteoblasts differentiation and bone tissue remodelling by regulation and coordination of osteoblasts and osteoclasts activity. It was found out that differentiation of cells in osteoinductive culture medium activates ERK2, JNK2 and p38 in time-dependent manner. ERK2 is activated via phosphorylation during differentiation of cells (Jaiswal et al. 2000). Elevated activity of NF-kB (which belongs to the Rel transcription factor family) in human mesenchymal stem cells was found to stimulate differentiation of cells in osteogenic direction, whereas decreased NF-kB signaling does not prevent the osteogenic differentiation (Hess et al. 2009).

It was shown that microRNA (miRNA) promotes os-teogenic differentiation via gene regulation, for example miR-2861 maximizes the Runx2 activity (Meng et al. 2015). It is known that PI3K/Akt and P-catenin pathways are able to regulate bone development. In this case, increased levels of p-catenin lead to enhancement of Runx-2 expression and finally facilitated bone formation. There is evidence that miR-21 can regulate PI3K/Akt pathway, and also regulates p-catenin pathway (Meng et al. 2015).

BMP-2 and BMP-4 factors are known to stimulate MSCs differentiation in osteogenic direction. Moreover, MSCs, which produce BMP-2, improve bone tissue repair in the sites of defects (Levy et al. 2010).

Activation of STAT proteins, which are inactive cytoplas-mic transcription factors, occurs as a result of stimulation of a variety of receptor types, and is usually accompanied by STAT tyrosine and serine phosphorylation. Members of the JAK tyrosine kinase family usually mediate STAT tyrosine phosphorylation, which leads to activation of STAT, its dimerization and translocation into nucleus. Transcriptional activity of STAT is significantly regulated by serine phosphorylation, which usually leads to transcription of proliferation-related genes (Levy et al. 2010).

The role of JAK-STAT signaling in MSCs is not properly studied yet; perhaps JAK-STAT signaling turns on in response to the influence of BMP-2 and BMP-4 (Levy et al. 2010).

Based on pleiotropic effects of erythropoietin on bone tissue, this hormone is attractive for increasing efficiency of bone tissue repair (Omlor et al. 2016). However, there have been a few studies to investigate the influence of erythropoietin on bone tissue repair in vivo (Bozlar et al. 2006, Garcia et al. 2011, Holstein et al. 2007, Holstein et al. 2011, Omlor et al. 2016, Rölfing et al. 2012, Shio-zawa et al. 2010, Sun et al. 2012). At present, the effect of erythropoietin on bone tissue is not fully understood (Balaian et al. 2018).

A positive effect of erythropoietin on differentiation of mesenchymal stem cells in osteogenic direction attracts a significant interest (Wang et al. 2018). It is known that mesenchymal stem cells immediately after fracture migrate to site of injury and differentiate into osteoblasts preceding the bone regeneration (Nair et al. 2013).

Most scientific papers note that erythropoietin triggers differentiation of mesenchymal stem cells into osteoblasts in vitro (Balaian et al. 2018, Kim et al. 2011, Li et al. 2015, Nair et al. 2013, Shiozawa et al. 2010).

On the surface of human MSCs, there are EPOR and CD131 receptors, which form EPO heterodimer receptor (EPOR/CD131) (Brines et al. 2004, Rölfing et al. 2014, Ye et al. 2010, Zwezdaryk et al. 2007). According to data obtained by Rölfing et al. (2014), there is no exclusive intracellular pathway responsible for the osteogenic differentiation of erythropoietin-stimulated MSCs (Rölfing et al. 2014). It is known that osteogenic effect of erythro-poietin on the mesenchymal stem cells is realized through the mammalian target of rapamycin (mTOR), JAK2 and PI3K signaling pathways (Rölfing et al. 2014).

It was shown that PI3K/Akt pathway is important in the process of MSCs differentiation into different cell types. However, the role of the pathway is complicated because of the opposite effects. PI3K/Akt signaling pathway regulates proliferation of cells, apoptosis, differentiation and migration. The factors which can stimulate the pathway activation include various growth hormones, cytokines and other molecules (Chen et al. 2013). The key enzyme of the pathway is PI3K, which converts phosphatidylinositol 4,5-biphosphate into phosphatidylinositol 3,4,5-triphos-phate, which binds Akt and 3-phosphoinositide-dependent protein kinase 1 (PDK1), and allows PDK1 to phosphoryl-ate Akt. Class 1A PI3K consists of two subunits: p110 catalytic subunit and p85 regulatory subunit. There are four isoforms of p110 subunit: p110-alpha, -beta, -gamma and -sigma and three isoforms of p85 subunit: p85-alpha, -beta and -gamma. The main target protein of the PI3K pathway is Akt. There are three isoforms of Akt: Akt1, Akt2, Akt3. Activation of Akt triggers a cascade of responses which regulate the cell functions. For instance, Akt regulates migration of cells by means of Rac1 and rhoA, increases viability through bcl-2, elevates angiogenesis via VEGF, and enhances proliferation by activation of mammalian target of rapamycin (Chen et al. 2013).

Activation of PI3K/Akt signaling pathway in MSCs is applied both in cellular therapy and in tissue engineering (Chen et al. 2013).

It is proved that the ability of MSCs to secrete cytokines is regulated by means of activity of PI3K/Akt signaling pathway. In this case, MSCs are able to secrete a number of cytokines which can regulate other cells and contribute to tissue recovery. These cytokines include vascular endothelial growth factor, fibroblast growth factor (FGF), monocyte chemoattractant protein-1, hepatocyte growth factor (HGF), insulin-like growth factor 1 (IGF-1), stromal cell-derived factor 1 (SDF-1), and thrombopoietin. Overexpression of Akt in MSCs increases expression of genes VEGF, FGF-2, HGF and IGF-1 and thus enhances secretion of these cytokines. These cytokines influence MSCs, increasing their functionality. For example, MSCs are able to express receptor to HGF, c-Met (tyrosine-pro-tein kinase Met or hepatocyte growth factor receptor (HGFR)). HGF at a concentration of 20 ng/ml promotes osteogenic differentiation by activating c-Met, Akt pathways and enhanced expression of the cell-cycle inhibitor p27 and osteogenic transcription factors Runx2 and Os-terix, whereas at high concentrations (100 ng/ml), HGF promotes proliferation and suppresses osteogenic differentiation by activating the ERK1/2 pathway and inhibiting the Akt pathway (Chen et al. 2013).

IGF-1 is found to be expressed in MSCs to increase osteogenesis for further bone repair. The mechanism includes activation of insulin receptor substrate-1 and phosphatidylinositol-3-kinase signaling pathway (IRS1-PI3K). Also, IGF-1 can activate the MAPK pathway. Some studies show that inhibition of MAPK increases rather than decreases the rate of osteogenesis (Higuchi et al. 2002, Osyczka and Leboy 2005, Payne et al. 2010, Wang et al. 2012), proving that the PI3K/Akt pathway plays a key role in IGF-induced osteogenesis. It was shown that mechanically induced activation of Akt also increases the rate of osteogenesis. There are also opposite data about the role of the PI3K/Akt pathway in the process of osteogenesis, according to which, imatinib stimulates osteogenesis by inhibiting platelet-derived growth factor receptors/PI3K/Akt (PDGFR/PI3K/Akt) signaling in MSCs. However, the reason of this phenomenon is not completely understood yet. It is known that BMP-2 enhances osteogenesis by activating the PI3K/ Akt pathway. At present, a number of signaling pathways are known to be involved in the process of cell differentiation in certain direction (Chen et al. 2013, Higuchi et al. 2002, Osyczka and Leboy 2005, Payne et al. 2010, Wang et al. 2012).

Erythropoietin is known to stimulate bone tissue formation and to increase the number of osteoblasts in vivo (Balaian et al. 2018, Mihmanli et al. 2009, Rolfing et al. 2012, Sun et al. 2012), especially if the ephrinB2/EphB4 signaling pathway is activated (Balaian et al. 2018, Li et al. 2015,). Erythropoietin regulates expression of EphB4 receptor of bone marrow mesenchymal stem cells (Balaian et al. 2018). Interaction of ephrinB2 with EphB4 receptor influencing erythropoietin-activated MSCs, triggers differentiation of cells (Balaian et al. 2018). Erythropoietin enhances osteoblast activity through the EphB4 signaling

pathway, and also increases the number of ephrinB2-ex-pressed osteoclasts but decreases their resorption activity. The signals activated by erythropoietin through ephrinB2/ EphB4 result in bone formation (Balaian et al. 2018).

Erythropoietin was found to act in collaboration with ephrinB2 and to induce osteoblast differentiation, but it can also influence this process through other potential mechanisms (Balaian et al. 2018). In the mouse cell line RAW264.7, erythropoietin significantly increases expression of Nfatc1, which is an obligatory transcription factor for osteoclastogenesis and regulates differentiation and fusion of osteoclasts. Moreover, erythropoietin significantly decreases expression of Mmp9 and Ctsk, which are involved in osteoclast-mediated resorption of organic components of the bone (Balaian et al. 2018). Furthermore, erythropoietin enhances expression of ephrinB2 on the cell line RAW264.7, which can indicate that erythropoietin increases the number of osteoclasts which are less capable of bone tissue resorption, but express a higher level of ephrinB2 (Balaian et al. 2018).

Erythropoietin can stimulate bone tissue formation due to increased expression of vascular endothelial growth factor (Holstein et al. 2011).

It was revealed that erythropoietin activates hemato-poietic stem cells to produce BMP factors (BMP2, BMP6). In addition, it was discovered that erythropoietin influences directly on bone marrow mesenchymal stem cells and induces their differentiation in osteogenic direction (Kim et al. 2011, Shiozawa et al. 2010). It is important that in in vivo conditions erythropoietin mediates bone formation by influencing simultaneously on both hematopoietic stem cells and the osteoblast niche. Thus, erythropoietin regulates bone formation directly and indirectly, which illustrates a link between the hematopoiesis and osteopoiesis processes in bone marrow (McGee et al. 2012, Shiozawa et al. 2010).

The evidence of the fact that erythropoietin causes differentiation of bone marrow mesenchymal stem cells in osteogenic direction is bone phenotype expression by cells, including activation of the bone-specific transcription factor Runx2, osteocalcin (OCN) and bone sialoprotein (BSP) under the influence of erythropoietin. Moreover, erythropoietin mediates an increase in mineral accumulation by the cell cultures and enhanced expression of alkaline phosphatase (ALP) (Shiozawa et al. 2010).

For erythropoietin-mediated osteoblast differentiation of human mesenchymal stem cells, the mTOR signaling pathway needs to be activated (Kim et al. 2011). It was also found out that erythropoietin in a combination with receptor activator of nuclear factor kappa-B ligand (RANKL) increased the number of osteoclasts formed from mouse marrow mononuclear cells (mMMCs) and mouse macrophage cell line RAW264.7. However, erythropoietin inhibits osteoclasts activity by decreasing cathepsin K regulation in mTOR-independent manner (Kim et al. 2011). During osteoblastogenesis, mTOR signaling may influence proliferation and differentiation of osteoblasts (Kim et al. 2011).

The effect of Erythropoietin on angiogenesis

It is well known that osteogenesis and angiogenesis are interconnected processes which are both required for bone formation (Wu et al. 2014). Angiogenesis is a complex process of new blood vessels formation from pre-existing ones. The process of angiogenesis includes several various stages: production and release of angiogenic factors, binding with endothelial cell receptors and subsequent activation of proliferation, directed migration, extracellular matrix remodelling, preparation and stabilization of the vessels (Buemi et al. 2009).

Erythropoietin takes part in regulation of angiogene-sis processes including new blood vessels formation from pre-existing ones. Physiological angiogenesis occurs actively in developing embryo, in female reproductive organs to maintain cyclic tissue renewal, and in wound healing (Arcasoy 2008). The role of erythropoietin is described as that of a pro-angiogenic factor comparable with classical pro-angiogenic factors, such as vascular en-dothelial growth factor and basic fibroblast growth factor (bFGF) (Zwezdaryk et al. 2007).

Expression of erythropoietin receptors on the various types of vascular endothelial cells is connected with the ability of erythropoietin to stimulate migration and proliferation of endothelial cells (Arcasoy 2008). One of the supposed mechanisms of erythropoietin-mediated stimulation of tissue neo-vascularisation is mobilization of bone marrow endothelial precursor cells into the circu-

References

■ Alehin SA, Kolmykov DI, Pokrovsky MV (2015) Human recombinant erythropoietin gradient dosage influence on ischemic and reperfusion liver injury. Research result: pharmacology and clinical pharmacology 1(4): 9-14. https://doi.org/10.18413/2500-235X-2015-1-4-9-14

■ Alekhin SA, Pokrovskii MV, Pokrovskaya TG, Alekhina VV, Kolmykov DI, Ivanova LV, Gureev VV, Staroseltseva OA, Gudyrev OS, Filimonov VA (2011) Recombinant erythropoietin as a preconditioning agent in abdominal organs ischemic damage. Privolzhsky Scietific Bulletin [Privolzhskiy Nauchnyi Vestnik] 2: 68-73. [in Russian]

■ Ammarguellat F, Gogusev J, Drueke TB (1996) Direct effect of erythropoietin on rat vascular smooth-muscle cell via a putative

erythropoietin receptor. Nephrology, Dialysis, Transplantation 11(4): 687-692. [PubMed]

■ Anagnostou A, Liu Z, Steiner M, Chin K, Lee ES, Kessimian N, No-guchi CT (1994) Erythropoietin receptor mRNA expression in human endothelial cells. Proceedings of the National Academy of Sciences of the United States of America 91(9): 3974-3978. [PubMed] [PMC]

■ Arcasoy MO (2008) The non-haematopoietic biological effects of erythropoietin. British Journal of Haematology 141(1): 14-31. https://doi.org/10.1111/j.1365-2141.2008.07014.x [PubMed]

■ Arcasoy MO (2010) Non-erythroid effects of erythropoietin. Hae-matologica 95(11): 1803-1805. https://doi.org/10.3324/haema-tol.2010.030213 [PubMed] [PMC]

latory bed (Arcasoy 2008, Bahlmann et al. 2004, George et al. 2005, Heeschen et al. 2003, Satoh et al. 2006, Vazquez-Mellado et al. 2017).

Human mesenchymal stem cells derived from bone marrow are known to express erythropoietin receptors and can promote angiogenesis following an erythropoietin treatment. Thus, human MSCs promote angiogenesis not only by direct participating in the process like endothelial cells which form new vessels, but also by secretion and recruiting of the components which are necessary for this process (Liu et al. 2011, Liu et al. 2013, Zwezdaryk et al. 2007).

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

Conclusions

Erythropoietin has the pleiotropic effect on multiple number of cells and tissues; it also influences the processes of remodelling and recovery of bone tissue. It is known that the effects of erythropoietin on bone tissue are linked with increased osteogenesis, osteoclastogenesis and angioge-nesis. At the same moment, the mechanisms involved in the process of bone tissue build-up via erythropoietin are still not fully understood.

Acknowledgements

The research was funded under the grant - State assignment № 20.11677.2018/10.11.

■ Bahlmann FH, De Groot K, Spandau JM, Landry AL, Hertel B, Duckert T, Boehm SM, Menne J, Haller H, Fliser D (2004) Erythropoietin regulates endothelial progenitor cells. Blood 103(3): 921-926. https://doi.org/10.1182/blood-2003-04-1284 [PubMed]

■ Bahlmann FH, DeGroot, K, Duckert T, Niemczyk E, Bahlmann E, Boehm SM, Haller H, Fliser D (2003) Endothelial progenitor cell proliferation and differentiation is regulated by erythropoietin. Kidney International 64(5): 1648-1652. https://doi.org/10.1046/ j.1523-1755.2003.00279.x [PubMed]

■ Balaian E, Wobus M, Weidner H, Baschant U, Stiehler M, Eh-ninger G, Bornhäuser M, Hofbauer LC, Rauner M, and Platzbecker U (2018) Erythropoietin inhibits osteoblast function in myelodys-plastic syndromes via the canonical Wnt pathway. Haematolog-ica 103(1): 61-68. https://doi.org/10.3324/haematol.2017.172726 [PubMed] [PMC]

■ Banfi A, Muraglia A, Dozin B, Mastrogiacomo M, Cancedda R, Quarto R (2000) Proliferation kinetics and differentiation potential of ex vivo expanded human bone marrow stromal cells: Implications for their use in cell therapy. Experimental Hematology 28(6): 707-715. https://doi.org/10.1016/S0301-472X(00)00160-0 [PubMed]

■ Bao H, Jacobs-Helber SM, Lawson AE, Penta K, Wickrema A, Sawyer ST (1999) Protein kinase B (c-Akt), phosphatidylinosi-tol 3-kinase, and STAT5 are activated by erythropoietin (EPO) in HCD57 erythroid cells but are constitutively active in an EPO-in-

dependent, apoptosis-resistant subclone (HCD57-SREI cells). Blood 93(11): 3757-3773. [PubMed]

■ Bernaudin M, Marti HH, Roussel S, Divoux D, Nouvelot A, ■ MacKenzie ET, Petit E (1999) A potential role for erythropoietin in focal permanent cerebral ischemia in mice. Journal of Cerebral Blood Flow and Metabolism 19(6): 643-651. https://doi. org/10.1097/00004647-199906000-00007 [PubMed]

■ Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, Gifford DK, ■ Melton DA, Jaenisch R, Young RA (2005) Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 122(6): 947956. https://doi.org/10.1016/j.cell.2005.08.020 [PubMed] [PMC] ■

■ Bozlar M, Kalaci A, Aslan B, Baktiroglu L, Yanat AN, Tasci A (2006) Effects of erythropoietin on fracture healing in rats. Saudi Medical Journal 27(8): 1267-1269. [PubMed]

■ Bramlett HM, Dietrich WD, Dixon CE, Shear DA, Schmid KE, ■ Mondello S, Wang KK, Hayes RL, Povlishock JT, Tortella FC, Kochanek PM (2016) Erythropoietin treatment in traumatic brain injury: operation brain trauma therapy. Journal of Neurotrauma. 33(6): 538-552. https://doi.org/10.1089/neu.2015.4116 [PubMed] ■

■ Brines M, Grasso G, Fiordaliso F, Sfacteria A, Ghezzi P, Fratelli M, Latini R, Xie QW, Smart J, Su-Rick CJ, Pobre E, Diaz D, Gomez D, Hand C, Coleman T, Cerami A (2004) Erythropoietin mediates tissue protection through an erythropoietin and common be-ta-subunit heteroreceptor. Proceedings of the National Academy of Sciences of the United States of America 101(41): 14907-14912. ■ https://doi.org/10.1073/pnas.0406491101 [PubMed] [PMC]

■ Broudy VC, Lin N, Brice M, Nakamoto B, Papayannopoulou T (1991) Erythropoietin receptor characteristics on primary human erythroid cells. Blood 77(12): 2583-2590. [PubMed]

■ Buemi M, Lacquaniti A, Maricchiolo G, Bolignano D, Campo S, Cernaro V, Sturiale A, Grasso G, Buemi A, Allegra A, Donato V, Genovese L (2009) Regenerative medicine: does erythropoietin ■ have a role? Current Pharmaceutical Design 15(17): 2026-2036. https://doi.org/10.2174/138161209788453194 [PubMed]

■ Bunn HF, Gu J, Huang LE, Park JW, Zhu H (1998) Erythropoietin: a model system for studying oxygen-dependent gene regulation. The Jour- ■ nal of Experimental Biology 201(Pt 8): 1197-1201. [PubMed] [PMC]

■ Carter-Su C, Schwartz J, Argetsinger LS (2016) Growth hormone signaling pathways. Growth Hormone & IGF Research: Official Journal of the Growth Hormone Research Society and the International IGF Research Society 28: 11-15. https://doi.org/10.1016/). ■ ghir.2015.09.002 [PubMed]

■ Chen G, Zhang S, Shi J, Ai J, Hang C (2009) Effects of recombinant human erythropoietin (rhEPO) on JAK2/STAT3 pathway and endothelial apoptosis in the rabbit basilar artery after subarachnoid haemorrhage. Cytokine 45: 162-168. https://doi.org/10.1016/). ■ cyto.2008.11.015 [PubMed]

■ Chen J, Crawford R, Chen C, Xiao Y (2013) The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. Tissue Engineering Part B: Reviews 19(6): 516-528. https://doi. org/10.1089/ten.teb.2012.0672 [PubMed] ■

■ Coleman T, Brines M (2004) Science review: Recombinant human erythropoietin in critical illness: a role beyond anemia? Critical Care 8(5): 337-341. https://doi.org/10.1186/cc2897 [PubMed] [PMC]

■ Conrad KP, Benyo DF, Westerhausen-Larsen A, Miles TM (1996) Expression of erythropoietin by the human placenta. FASEB Jour-

nal: Official publication of the Federation of American Societies for Experimental Biology 10(7): 760-768. [PubMed] Correia C, Lee SH, Meng XW, Vincelette ND, Knorr KL, Ding H, Nowakowski GS, Dai H, Kaufmann SH (2015) Emerging understanding of Bcl-2 biology: Implications for neoplastic progression and treatment. Biochimica et Biophysica Acta 1853(7): 1658-1671. https://doi.org/10.1016/j.bbamcr.2015.03.012 [Pu-bMed] [PMC]

Dame C, Fahnenstich H, Freitag P, Hofmann D, Abdul-Nour T, Bartmann P, Fandrey J (1998) Erythropoietin mRNA expression in human fetal and neonatal tissue. Blood 92(9): 3218-3225. [PubMed] Damen JE, Wakao H, Miyajima A, Krosl J, Humphries RK, Cutler RL, Krystal G (1995). Tyrosine 343 in the erythropoietin receptor positively regulates erythropoietin-induced cell proliferation and Stat5 activation. The EMBO Journal 14(22): 5557-5568. [PubMed] [PMC] Danilenko LM (2013) Use of recombinant eritropoietin and resveratrol for pharmacological correction of myocardial ischemic injuries. Current Problems of Science and Education [Sovremen-nye problemy nauki i obrazovaniya] 6: 1019. [in Russian] Danilenko LM, Pokrovskiy MV, Novikov OO, Korokina LV, Gon-charova DB, Inderova AB, Svetlichnaya EN (2012) Trigger mechanism of antiischemic action of erythropoietin and resveratrol. Belgorod State University Scientific Bulletin. Medicine. Pharmacy [Nauchnye Vedomosti BelGU. Seriya Meditsina. Farmatsiya] 10(129, 18/2): 138-142. [in Russian]

Danilenko LM, Pokrovsky MV, Novikov OO, Kochkarov VI, Korolev AE, Korokina LV, Trifonov BV, Charitonova OV (2011) Antiinflammatory effects of distant ischaemic preconditioning in combination with recombinant erythropoietin. Belgorod State University Scientific Bulletin. Medicine. Pharmacy [Nauchnye Vedomosti BelGU. Seriya Meditsina. Farmatsiya] 4 (99, 13/2): 49-53. [in Russian]

Debeljak N, Solar P, Sytkowski AJ (2014) Erythropoietin and cancer: the unintended consequences of anemia correction. Frontiers in Immunology 5: 563. https://doi.org/10.3389/fimmu.2014.00563 [PubMed] [PMC]

Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell ED (2006) Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells 24(4): 1054-1064. https://doi.org/10.1634/stemcells.2005-0370 [PubMed]

Denisyuk TA (2016) Recombinant erythropoietin and correction of endotoksin-induced endothelial dysfunction by statin. Kursk Scientific and Practical Bulletin "Man and his Health" [Kurskii Nauchno-prakticheskii Vestnik "Chelovek i Ego Zdorov'e"] 2: 61-65. [in Russian]

Denisyuk TA, Pokrovskii MV (2016) Combined use of recombinant erythropoietin and statins at endotoxin-induced endothelial dysfunction. Allergology and Immunology. World Congress on Clinical Pathology and Rehabilitation in Medicine [Allergologiya i immunologiya. Vsemirnyy kongress po klinicheskoy patologii i reabilitatsii v meditsine] 17(1): 64-65. [in Russian] Denisyuk TA, Pokrovskii MV, Pokrovskaya TG, Korokin MV, Gudyrev OS, Reznikov KM, Povetkin SV, Stepchenko AA (2015) Correction of endotoxin-induced endothelial dysfunction by recombinant erythropoietin and inhibitors of HMG-Co-A-Reduc-tase. Kuban Scientific Medical Bulletin [Kubanskii Nauchnyi Meditsinskii Vestnik] 6 (155): 39-43. [in Russian]

■ Desai S (2012) Signal transduction effects induced by erythropoi-etin in a HNSCC model system. Richmond, Virginia: School of Medicine Virginia Commonwealth University. 113. https://scholar-scompass.vcu.edu/cgi/viewcontent.cgi?article=1411&context=etd

■ Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, Prock-op DJ (1999) Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. British Journal of Haematology 107(2): 275-281. https://doi. org/10.1046/j.1365-2141.1999.01715.x [PubMed]

■ Elagin VV, Kostina DA, Bratchikov OI, Pokrovskii MV, Pokrovskaya TG (2018) The study of renoprotective properties of erythropoietin derivatives on the kidney ischemia-reperfusion model. Kuban Scientific Medical Bulletin [Kubanskii Nauchnyi Meditsinskii Vestnik] 25(6): 73-77. [in Russian]

■ Elliott S, Sinclair AM (2012) The effect of erythropoietin on normal and neoplastic cells. Biologics 6: 163-189. https://doi. org/10.2147/BTT. S32281 [PubMed] [PMC]

■ Erbayraktar S, Yilmaz O, Gökmen N, Brines M (2003) Erythropoi-etin is a multifunctional tissue-protective cytokine. Current Hema-tology Reports 2(6): 465-470. [PubMed]

■ Flaharty KK, Caro J, Erslev A, Whalen JJ, Morris EM, Bjornsson TD, Vlasses PH (1990) Pharmacokinetics and erythropoietic response to human recombinant erythropoietin in healthy men. Clinical Pharmacology and Therapeutics 47(5): 557-564. https://doi. org/10.1038/clpt.1990.76 [PubMed]

■ Ganz T (2018) Erythropoietin and iron - a conflicted alliance? Kidney International 94: 851-853. https://doi.org/10.1016/j. kint.2018.07.027 [PubMed]

■ Garcia P, Speidel V, Scheuer C, Laschke MW, Holstein JH, Hist-ing T, Pohlemann T, Menger MD (2011) Low dose erythropoietin stimulates bone healing in mice. Journal of Orthopaedic Research: Official Publication of the Orthopaedic Research Society 29(2): 165-172. https://doi.org/10.1002/jor.21219 [PubMed]

■ George J, Goldstein E, Abashidze A, Wexler D, Hamed S, Shmi-lovich H, Deutsch V, Miller H, Keren G, Roth A (2005) Eryth-ropoietin promotes endothelial progenitor cell proliferative and adhesive properties in a PI 3-kinase-dependent manner. Cardiovascular Research 68(2): 299-306. https://doi.org/10.1016/j.cardi-ores.2005.06.022 [PubMed]

■ Greco SJ, Liu K, Rameshwar P (2007) Functional similarities among genes regulated by OCT4 in human mesenchymal and embryonic stem cells. Stem Cells 25(12): 3143-3154. https://doi. org/10.1634/stemcells.2007-0351 [PubMed]

■ Grimm C, Wenzel A, Groszer M, Mayser H, Seeliger M, Samardzi-ja M, Bauer C, Gassmann M, Reme CE (2002) HIF-1-induced erythropoietin in the hypoxic retina protects against light-induced retinal degeneration. Nature Medicine 8(7): 718-724. https://doi. org/10.1038/nm723 [PubMed]

■ Gupta N, Wish JB (2017) Erythropoietin and its cardiovascular effects. In: Rangaswami J, Lerma E, Ronco C (Eds) Cardio-Neph-rology. Springer, Cham, 119-128. https://doi.org/10.1007/978-3-319-56042-7_11

■ Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, Mildner-Rihm C, Martin H, Zeiher AM, Dimmeler S (2003) Erythropoietin is a potent physiologic stimulus for en-dothelial progenitor cell mobilization. Blood 102(4): 1340-1346. https://doi.org/10.1182/blood-2003-01-0223 [PubMed]

■ Hess K, Ushmorov A, Fiedler J, Brenner RE, Wirth T (2009) TNFal-pha promotes osteogenic differentiation of human mesenchymal stem cells by triggering the NF-kappaB signaling pathway. Bone 45(2): 367-376. https://doi.org/10.1016Zj.bone.2009.04.252 [PubMed]

■ Higuchi C, Myoui A, Hashimoto N, Kuriyama K, Yoshioka K, Yoshikawa H, Itoh K (2002) Continuous inhibition of MAPK signaling promotes the early osteoblastic differentiation and mineralization of the extracellular matrix. Journal of Bone and Mineral Research: the Official Journal of the American Society for Bone and Mineral Research 17(10): 1785-1794. https://doi.org/10.1359/ jbmr.2002.17.10.1785 [PubMed]

■ Hiram-Bab S, Liron T, Deshet-Unger N, Mittelman M, Gassmann M, Rauner M, Franke K, Wielockx B, Neumann D, Gabet Y (2015) Erythropoietin directly stimulates osteoclast precursors and induces bone loss. FASEB Journal: Official publication of the Federation of American Societies for Experimental Biology 29(5): 1890-1900. https://doi.org/10.1096/fj.14-259085 [PubMed]

■ Hiram-Bab S, Neumann D, Gabet Y (2017) Erythropoietin in bone - controversies and consensus. Cytokine 89: 155-159. https://doi. org/10.1016/j.cyto.2016.01.008 [PubMed]

■ Holstein JH, Menger MD, Scheuer C, Meier C, Culemann U, Wirbel RJ, Garcia P, Pohlemann T (2007) Erythropoietin (EPO): EPO-receptor signaling improves early endochondral ossification and mechanical strength in fracture healing. Life Sciences 80(10): 893-900. https://doi.org/10.1016/j.lfs.2006.11.023 [PubMed]

■ Holstein JH, Orth M, Scheuer C, Tami A, Becker SC, Garcia P, Histing T, Mörsdorf P, Klein M, Pohlemann T, Menger MD (2011) Erythropoietin stimulates bone formation, cell proliferation, and an-giogenesis in a femoral segmental defect model in mice. Bone 49(5): 1037-1045. https://doi.org/10.1016/j.bone.2011.08.004 [PubMed]

■ Jacobs K, Shoemaker C, Rudersdorf R, Neill SD, Kaufman RJ, Mufson A, Seehra J, Jones SS, Hewick R, Fritsch EF, Kawaki-ta M, Shimizu T, Miyake T (1985) Isolation and characterization of genomic and cDNA clones of human erythropoietin. Nature 313(6005): 806-810. [PubMed]

■ Jaiswal RK, Jaiswal N, Bruder SP, Mbalaviele G, Marshak DR, Pittenger MF (2000) Adult human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by mitogen-activated protein kinase. The Journal of Biological Chemistry 275(13): 9645-9652. [PubMed]

■ Jelkmann W (2007) Erythropoietin after a century of research: younger than ever. European Journal of Haematology 78(3): 183205. https://doi.org/10.1111/j.1600-0609.2007.00818.x [PubMed]

■ Jelkmann W (2013) Physiology and pharmacology of erythro-poietin. Transfusion Medicine and Hemotherapy 40(5): 302-309. https://doi.org/10.1159/000356193 [PubMed] [PMC]

■ Juul SE, Pet GC (2015) Erythropoietin and Neonatal Neuroprotection. Clinics in perinatology 42(3): 469-481. https://doi. org/10.1016/j.clp.2015.04.004 [PubMed] [PMC]

■ Juul SE, Zhao Y, Dame JB, Du Y, Hutson AD, Christensen RD (2000) Origin and fate of erythropoietin in human milk. Pediat-ric Research 48(5): 660-667. https://doi.org/10.1203/00006450-200011000-00018 [PubMed]

■ Kedarisetty CK, Anand L, Bhardwaj A, Bhadoria AS, Kumar G, Vyas AK, David P, Trehanpati N, Rastogi A, Bihari C, Maiwall R, Garg HK, Vashishtha C, Kumar M, Bhatia V, Sarin SK (2015) Combination of granulocyte colony-stimulating factor and erythropoietin improves outcomes of patients with decompensat-

ed cirrhosis. Gastroenterology 148(7): 1362-1370.e7. https://doi. org/10.1053/j.gastro.2015.02.054 [PubMed]

■ Kim J, Jung Y, Sun H, Joseph J, Mishra A, Shiozawa Y, Wang Y, Krebsbach PH, Taichman RS (2011) Erythropoietin mediated bone formation is regulated by mTOR signaling. Journal of Cellular Biochemistry 113(1): 220-228. https://doi.org/10.1002/ jcb.23347 [PubMed] [PMC]

■ Kirito K, Nakajima K, Watanabe T, Uchida M, Tanaka M, Ozawa K, Komatsu N (2002) Identification of the human erythropoietin receptor region required for Stat1 and Stat3 activation. Blood 99(1): 102-110. https://doi.org/10.1182/blood.V99.L102 [PubMed]

■ Klingmüller U, Bergelson S, Hsiao JG, Lodish HF (1996) Multiple tyrosine residues in the cytosolic domain of the erythropoietin receptor promote activation of STAT5. Proceedings of the National Academy of Sciences of the United States of America 93(16): 83248328. https://doi.org/10.1073/pnas.93.16.8324 [PubMed] [PMC]

■ Klontzas ME, Kenanidis EI, MacFarlane RJ, Michail T, Potoupnis ME, Heliotis M, Tsiridis E (2016) Investigational drugs for fracture healing: preclinical & clinical data. Expert Opinion on Inves-tigational Drugs 25(5): 585-596. https://doi.org/10.1517/1354378 4.2016.1161757 [PubMed]

■ Klopsch C, Furlani D, Gäbel R, Li W, Pittermann E, Ugurlucan M, Kundt G, Zingler C, Titze U, Wang W, Ong L-L, Wagner K, Li R-K, Ma N, Steinhoff G (2009) Intracardiac injection of erythropoietin induces stem cell recruitment and improves cardiac functions in a rat myocardial infarction model. Journal of Cellular and Molecular Medicine 13(4): 664-679. https://dx.doi.org/10.1111%2Fj.1582-4934.2008.00546.x [PubMed] [PMC]

■ Kobayashi T, Yanase H, Iwanaga T, Sasaki R, Nagao M (2002) Epididymis is a novel site of erythropoietin production in mouse reproductive organs. Biochemical and Biophysical Rresearch Communications 296(1): 145-151. https://doi.org/10.1016/ S0006-291X(02)00832-X [PubMed]

■ Kolesnik IM, Pokrovskii MV, Pokrovskaya TG, Gudyrev OS, Danilenko LM, Korokin MV, Alekhin SA, Grigorenko AP, Sta-roseltseva OA, Dolgikova IN, Bratchikov OI, Molchanova OV, Efremenkova DA, Polianskaia OS, Filimonov VA (2011) Pharmacological preconditioning by erythropoietin in limb ischemia. Biomedicine [Biomeditsina] 4: 90-92. [in Russian]

■ Kolesnik IM, Pokrovskii MV, Lasarenko VA (2010) Pharmacological preconditioning with erythropoietin - new possibilities for optimization of ischemic tissues survival potential. Kursk Scientific and Practical Bulletin "Man and his Health" [Kurskij nauchno-prak-ticheskij vestnik "Chelovek i ego zdorov'e"] 3: 32-36. [in Russian]

■ Korokina LV, Kolesnik IM, Pokrovskii MV, Korokin MV, Belous AS, Artjushkova EB, Pokrovskaya TG, Gudyrev OS, Korolev AE, Pavlova LA, Novikov OO (2009) Pharmacological correction of L-NAME induced nitric oxide deficiency with recombinant eryth-ropoetin. Kuban Scientific Medical Bulletin [Kubanskii Nauchnyi Meditsinskii Vestnik] 9 (114): 66-70. [in Russian]

■ Koury MJ, Bondurant MC (1992) The molecular mechanism of erythropoietin action. European Journal of Biochemistry 210(3): 649-663. https://doi.org/10.1111/j.1432-1033.1992.tb17466.x [PubMed]

■ Kretz A, Happold CJ, Marticke JK, Isenmann S (2005) Eryth-ropoietin promotes regeneration of adult CNS neurons via Jak2/ Stat3 and PI3K/AKT pathway activation. Molecular and Cellular Neurosciences 29(4): 569-579. https://doi.org/10.1016/). mcn.2005.04.009 [PubMed]

■ Kwon M-S, Kim M-H, Kim S-H, Park K-D, Yoo S-H, Oh I-U, Pak S, Seo Y-J (2014) Erythropoietin exerts cell protective effect by activating PI3K/Akt and MAPK pathways in C6 Cells. Neurological Research 36(3): 215-223. https://doi.org/10.1179/174313281 3Y.0000000284 [PubMed]

■ La Ferla K, Reimann C, Jelkmann W, HellwigBürgel T (2002) Inhibition of erythropoietin gene expression signaling involves the transcription factors GATA-2 and NF-kB. FASEB Journal: Official publication of the Federation of American Societies for Experimental Biology 16(13): 1811-1813. https://doi.org/10.1096/fj.02-0168fje [PubMed]

■ Lappin TR, Maxwell AP, Johnston PG (2002) EPO's Alter ego: erythropoietin has multiple actions. Stem Cells 20(6): 485-492. https://doi.org/10.1634/stemcells.20-6-485 [PubMed]

■ Levy O, Ruvinov E, Reem T, Granot Y, Cohen S (2010) Highly efficient osteogenic differentiation of human mesenchymal stem cells by eradication of STAT3 signaling. The International Journal of Biochemistry and Cell Biology 42(11): 1823-1830. https://doi. org/10.1016/j.biocel.2010.07.017 [PubMed]

■ Li B, Zhang G, Li R, Duan C (2015) Phosphoinositide 3-kinase/ Akt pathway mediates Fip1-like1-platelet-derived growth factor receptor a-induced cell infiltration and activation: possible molecular mechanism for the malignant phenotype of chronic eosino-philic leukemia. Cancer Translational Medicine 1: 31-34. http:// www.cancertm.com/text.asp?2015/1/1/31/151490

■ Li C, Shi C, Kim J, Chen Y, Ni S, Jiang L, Zheng C, Li D, Hou J, Taichman RS, Sun H (2015) Erythropoietin promotes bone formation through EphrinB2/EphB4 signaling. Journal of Dental Research 94(3): 455-463. https://doi.org/10.1177/0022034514566431 [Pu-bMed] [PMC]

■ Li J, Guo W, Xiong M, Zhang S, Han H, Chen J, Mao D, Yu H, Zeng Y (2017) Erythropoietin facilitates the recruitment of bone marrow mesenchymal stem cells to sites of spinal cord injury. Experimental and Therapeutic Medicine 13(5): 1806-1812. https:// doi.org/10.3892/etm.2017.4182 [PubMed] [PMC]

■ Lin JS, Chen YS, Chiang HS, Ma MC (2008) Hypoxic preconditioning protects rat hearts against ischaemia-reperfusion injury: Role of erythropoietin on progenitor cell mobilization. The Journal of Physiology 586(23): 5757-5769. https://doi.org/10.1113/jphys-iol.2008.160887 [PubMed] [PMC]

■ Liu C, Shen K, Liu Z, Noguchi CT (1997) Regulated human eryth-ropoietin receptor expression in mouse brain. The Journal of Biological Chemistry 272(51): 32395-32400. https://doi.org/10.1074/ jbc.272.51.32395 [PubMed]

■ Liu N, Tian J, Wang W, Cheng J, Hu D, Zhang J (2011) Effect and mechanism of erythropoietin on mesenchymal stem cell proliferation in vitro under the acute kidney injury microenvironment. Experimental Biology and Medicine (Maywood) 236(9): 1093-1099. https://doi.org/10.1258/ebm.2011.011001 [PubMed]

■ Liu N, Tian J, Wang WW, Han GF, Cheng J, Huang J, Zhang JY (2013) Effect of erythropoietin on the migration of bone marrow-derived mesenchymal stem cells to the acute kidney injury microenvironment. Experimental Cell Research 319(13): 20192027. https://doi.org/10.1016Zj.yexcr.2013.04.008 [PubMed]

■ Liu WC, Wen L, Xie T, Wang H, Gong JB, Yang XF (2017) Therapeutic effect of erythropoietin in patients with traumatic brain injury: a meta-analysis of randomized controlled trials. Journal of Neurosurgery 127(1): 8-15. https://doi.org/10.3171/2016.4. JNS152909 [PubMed]

■ Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J, Wong KY, Sung KW, Lee CW, Zhao XD, Chiu KP, Lipovich L, Kuznetsov VA, Robson P, Stanton LW, Wei CL, Ruan Y, Lim B, Ng HH (2006) The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nature Genetics 38(4): 431-440. https://doi.org/10.1038/ng1760 [PubMed]

■ Ma C, Cheng F, Wang X, Zhai C, Yue W, Lian Y, Wang Q (2016) Erythropoietin pathway: a potential target for the treatment of depression. International Journal of Molecular Sciences 17(5) pii: E677. https://doi.org/10.3390/ijms17050677 [PubMed] [PMC]

■ Magnanti M, Gandini O, Giuliani L, Gazzaniga P, Marti HH, Gradilone A, Frati L, Agliano AM, Gassmann M (2001) Eryth-ropoietin expression in primary rat Sertoli and peritubular myoid cells. Blood 98(9): 2872-2874. https://doi.org/10.1182/blood. V98.9.2872 [PubMed]

■ Maiese K, Chong ZZ, Hou J, Shang YC (2008) Erythropoietin and oxidative stress. Current Neurovascular Research 5(2): 125-142. https://doi.org/10.2174/156720208784310231 [PubMed] [PMC]

■ Marelli A, Czornyj L, Rocha L, Lazarowski A (2016) Erythropoietin as potential neuroprotective and antiepileptogenic agent in epilepsy and refractory epilepsy. In: Talevi A, Rocha L (Eds) Antiepileptic Drug Discovery. Methods in Pharmacology and Toxicology. Humana Press, New York, 147-161. https://doi.org/10.1007/978-1-4939-6355-3_8

■ Marti HH, Wenger RH, Rivas LA, Straumann U, Digicaylioglu M, Henn V, Yonekawa Y, Bauer C, Gassmann M (1996) Eryth-ropoietin gene expression in human, monkey and murine brain. The European Journal of Neuroscience 8(4): 666-676. https://doi. org/10.1111/j.1460-9568.1996.tb01252.x [PubMed]

■ Masuda S, Kobayashi T, Chikuma M, Nagao M, Sasaki R (2000) The oviduct produces erythropoietin in an estrogen- and oxygen-dependent manner. American Journal of Physiology. Endocrinology and Metabolism 278(6): E1038-E1044. https://doi. org/10.1152/ajpendo.2000.278.6.E1038 [PubMed]

■ McGee SJ, Havens AM, Shiozawa Y, Jung Y, Taichman RS (2012) Effects of erythropoietin on the bone microenvironment. Growth Factors 30(1): 22-28. https://doi.org/10.3109/08977194.2011.637 034 [PubMed] [PMC]

■ Meng YB, Li X, Li ZY, Zhao J, Yuan XB, Ren Y, Cui ZD, Liu YD, Yang XJ (2015) microRNA-21 promotes osteogenic differentiation of mesenchymal stem cells by the PI3K/ß-catenin pathway. Journal of Orthopaedic Research: Official Publication of the Orthopaedic Research Society 33(7): 957-964. https://doi. org/10.1002/jor.22884 [PubMed]

■ Mihmanli A, Dolanmaz D, Avunduk MC, Erdemli E (2009) Effects of recombinant human erythropoietin on mandibular distraction osteogenesis. Journal of Oral and Maxillofacial Surgery: Official Journal of the American Association of Oral and Maxil-lofacial Surgeons 67(11): 2337-2343. https://doi.org/10.1016/j. joms.2008.06.082 [PubMed]

■ Minguell JJ, Erices A, Conget P (2001) Mesenchymal stem cells. Experimental Biology and Medicine (Maywood) 226(6): 507520. [PubMed]

■ Modrak M, Sundem L, Elfar J (2017) Erythropoietin enhanced recovery after peripheral nerve injury. Neural Regeneration Research 12(8): 1268-1269. https://doi.org/10.4103/1673-5374.213544 [Pu-bMed] [PMC]

■ Nair AM, Tsai YT, Shah KM, Shen J, Weng H, Zhou J, Sun X, Saxena R, Borrelli J Jr, Tang L (2013) The effect of erythropoie-

tin on autologous stem cell-mediated bone regeneration. Biomaterials 34(30): 7364-7371. https://doi.org/10.1016/j.biomateri-als.2013.06.031 [PubMed]

■ Ogilvie M, Yu X, Nicolas-Metral V, Pulido SM, Liu C, Ruegg UT, Noguchi CT (2000) Erythropoietin stimulates proliferation and interferes with differentiation of myoblasts. The Journal of Biological Chemistry 275(50): 39754-39761. https://doi.org/10.1074/jbc. M004999200 [PubMed]

■ Omlor GW, Kleinschmidt K, Gantz S, Speicher A, Guehring T, Richter W (2016) Increased bone formation in a rabbit long-bone defect model after single local and single systemic application of erythropoietin. Acta Orthopaedica 87(4): 425-431. https://doi.org/ 10.1080/17453674.2016.1198200 [PubMed] [PMC]

■ Osyczka AM, Leboy PS (2005) Bone morphogenetic protein regulation of early osteoblast genes in human marrow stromal cells is mediated by extracellular signal-regulated kinase and phosphati-dylinositol 3-kinase signaling. Endocrinology 146(8): 3428-3437. https://doi.org/10.1210/en.2005-0303 [PubMed] [PMC]

■ Ott C, Martens H, Hassouna I, Oliveira B, Erck C, Zafeiriou MP, Peteri UK, Hesse D, Gerhart S, Altas B, Kolbow T, Stadler H, Ka-wabe H, Zimmermann WH, Nave KA, Schulz-Schaeffer W, Jahn O, Ehrenreich H (2015) Widespread expression of erythropoietin receptor in brain and its induction by injury. Molecular Medicine (Cambridge, Mass.) 21(1): 803-815. https://doi.org/10.2119/ molmed.2015.00192 [PubMed] [PMC]

■ Payne KA, Meszaros LB, Phillippi JA, Huard J (2010) Effect of phosphatidyl inositol 3-kinase, extracellular signal-regulated ki-nases 1/2, and p38 mitogen-activated protein kinase inhibition on osteogenic differentiation of muscle-derived stem cells. Tissue Engineering Part A 16(12): 3647-3655. https://doi.org/10.1089/ten. TEA.2009.0738 [PubMed] [PMC]

■ Perreault AA, Venters BJ (2018) Integrative view on how eryth-ropoietin signaling controls transcription patterns in erythroid cells. Current Opinion in Hematology 25(3): 189-195. https://doi. org/10.1097/MOH.0000000000000415 [PubMed]

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

■ Piuhola J, Kerkelä R, Keenan JI, Hampton MB, Richards AM, Pemberton CJ (2008) Direct cardiac actions of erythropoietin (EPO): effects on cardiac contractility, BNP secretion and ischae-mia/reperfusion injury. Clinical Science. 114(4): 293-304. https:// doi.org/10.1042/CS20070229 [PubMed]

■ Pokrovskii MV, Pokrovskaya TG, Arustamova AA, Kolesnik IM, Sakharov MS, Korobtsova IA, Myagchenko SV, Chetverikova AYu, Filimonov VA, Molchanova OV, Shmykova EA, Isaychenko NN, Khodov SV (2012) Stimulation of neovasculogenesis with recombinant erythropoietin. Innovations in Current Pharmacology: Proceedings of the IV Congress of Russian Pharmacologists, Kazan (Russia), September 2012. Moscow, 153. [Innovatsii v Sovremennoy Farmakologii: Materialy IV S"ezda Farmakologov Rossii, Kazan' (Rossiya), Sentyabr' 2012. Moskva, 153]. [in Russian]

■ Rafiee P, Shi Y, Su J, Pritchard KA, Jr, Tweddell JS, Baker JE (2005) Erythropoietin protects the infant heart against is-chemia-reperfusion injury by triggering multiple signaling pathways. Basic Research in Cardiology 100(3): 187-197. https://doi. org/10.1007/s00395-004-0508-1 [PubMed]

■ Rankin EB, Biju MP, Liu Q, Unger TL, Rha J, Johnson RS, Simon MC, Keith B, Haase VH (2007) Hypoxia-inducible factor-2 (HIF-2) regulates hepatic erythropoietin in vivo. The Journal of

Clinical Investigation 117(4): 1068-1077. https://doi.org/10.1172/ JCI30117 [PubMed] [PMC]

■ Rennert RC, Sorkin M, Garg RK, Gurtner GC (2012) Stem cell recruitment after injury: lessons for regenerative medicine. Regenerative Medicine 7(6): 833-850. https://doi.org/10.2217/rme.12.82. doi: 10.2217/rme.12.82 [PubMed] [PMC]

■ Ridley DM, Dawkins F, Perlin E (1994) Erythropoietin: a review. Journal of the National Medical Association 86(2): 129-135. [Pu-bMed] [PMC]

■ Rocha J, Eduardo-Figueira M, Barateiro A, Fernandes A, Brites D, Pinto R, Freitas M, Fernandes E, Mota-Filipe H, Sepodes B (2015) Erythropoietin reduces acute lung injury and multiple organ failure/dysfunction associated to a scald-burn inflammatory injury in the rat. Inflammation 38(1): 312-326. https://doi.org/10.1007/ s10753-014-0035-7 [PubMed]

■ Rölfing JHD (2014) The effect of erythropoietin on bone. Thesis Acta Orthopaedica Supplementum 353(85): 1-29. https://doi.org/ 10.3109/17453674.2013.869716

■ Rölfing JHD, Baatrup A, Stiehler M, Jensen J, Lysdahl H, Bünger C (2014) The osteogenic effect of erythropoietin on human mesen-chymal stromal cells is dose-dependent and involves non-hemato-poietic receptors and multiple intracellular signaling pathways. Stem Cell Reviews 10(1): 69-78. https://doi.org/10.1007/s12015-013-9476-x [PubMed]

■ Rölfing JHD, Bendtsen M, Jensen J, Stiehler M, Foldager CB, Hellfritzsch MB, Bünger C (2012) Erythropoietin augments bone formation in a rabbit posterolateral spinal fusion model. Journal of Orthopaedic Research: Official Publication of Orthopaedic Research Society 30(7): 1083-1088. https://doi.org/10.1002/ jor.22027 [PubMed]

■ Sachinidis A, Kolossov E, Fleischmann BK, Hescheler J (2002) Generation of cardiomyocytes from embryonic stem cells experimental studies. Herz 27(7): 589-597. https://doi.org/10.1007/ s00059-002-2423-2 [PubMed]

■ Saei Arezoumand K, Alizadeh E, Pilehvar-Soltanahmadi Y, Es-maeillou M, Zarghami N (2017) An overview on different strategies for the stemness maintenance of MSCs. Artificial Cells, Na-nomedicine, and Biotechnology 45(7): 1255-1271. https://doi.org /10.1080/21691401.2016.1246452 [PubMed]

■ Sanchez-Gonzalez LR, Castro-Melendez SE, Angeles-Torres AC, Castro-Cortina N, Escobar-Valencia A, Quiroga-Garza A (2016) Efficacy and safety of adjuvant recombinant human erythropoietin and ferrous sulfate as treatment for iron deficiency anemia during the third trimester of pregnancy. European Journal of Obstetrics, Gynecology, and Reproductive Biology 295: 32-36. https://doi. org/10.1016/j.ejogrb.2016.08.004 [PubMed]

■ Sasaki R, Masuda S, Nagao M (2000) Erythropoietin: Multiple physiological functions and regulation of biosynthesis. Bioscience, Biotechnology and Biochemistry 64(9): 1775-1793. https:// doi.org/10.1271/bbb.64.1775 [PubMed]

■ Sato T, Maekawa T, Watanabe S, Tsuji K, Nakahata T (2000) Eryth-roid progenitors differentiate and mature in response to endogenous erythropoietin. The Journal of Clinical Investigation 106(2): 263270. https://doi.org/10.1172/JCI9361 [PubMed] [PMC]

■ Satoh K, Kagaya Y, Nakano M, Ito Y, Ohta J, Tada H, Karibe A, Minegishi N, Suzuki N, Yamamoto M, Ono M, Watanabe J, Shi-rato K, Ishii N, Sugamura K, Shimokawa H (2006) Important role of endogenous erythropoietin system in recruitment of endothe-

lial progenitor cells in hypoxia-induced pulmonary hypertension in mice. Circulation 113(11): 1442-1450. https://doi.org/10.1161/ CIRCULATIONAHA.105.583732 [PubMed]

■ Schwartz C, Willebrand R, Huber S, Rupec RA, Wu D, Locksley R, Voehringer D (2015) Eosinophil-specific deletion of IKBa in mice reveals a critical role of NF-KB-induced Bcl-xL for inhibition of apoptosis. Blood 125(25): 3896-3904. https://doi.org/10.1182/ blood-2014-10-607788 [PubMed] [PMC]

■ Semenza GL, Wang GL (1992) A nuclear factor induced by hypox-ia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Molecular and Cellular Biology 12(12): 5447-5454. [PMC]

■ Shiozawa Y, Jung Y, Ziegler AM, Pedersen EA, Wang J, Wang Z, Song J, Wang J, Lee CH, Sud S, Pienta KJ, Krebsbach PH, Taichman RS (2010) Erythropoietin couples hematopoiesis with bone formation. PLoS One 5(5): e10853. https://doi.org/10.1371/ journal.pone.0010853 [PubMed] [PMC]

■ Stopka T, Zivny JH, Stopkova P, Prchal JF, Prchal JT (1998) Human hematopoietic progenitors express erythropoietin. Blood 91(10): 3766-3772. [PubMed]

■ Suk KK, Dunbar JA, Liu A, Daher NS, Leng CK, Leng JK, Lim P, Weller S, Fayard E (2008) Human recombinant erythropoietin and the incidence of retinopathy of prematurity: a multiple regression model. Journal AAPOS: the Official Publication of the American Association for Pediatric Ophthalmology and Strabismus 12(3): 233-238. https://doi.org/10.1016/jjaapos.2007.08.009 [PubMed]

■ Sun H, Jung Y, Shiozawa Y, Taichman R S, Krebsbach PH (2012) Erythropoietin modulates the structure of bone morphogenetic protein 2-engineered cranial bone. Tissue Engineering Part A 18(19-20): 2095-2105. https://doi.org/10.1089/ten.tea.2011.0742 [PMC]

■ Tögel FE, Ahlstrom JD, Yang Y, Hu Z, Zhang P, Westenfelder C

(2016) Carbamylated erythropoietin outperforms erythropoietin in the treatment of AKI-on-CKD and other AKI models. Journal of the American Society of Nephrology 27(11): 3394-3404. https:// doi.org/10.1681/asn.2015091059 [PMC]

■ Tojo Y, Sekine H, Hirano I, Pan X, Souma T, Tsujita T, Kawagu-chi S, Takeda N, Takeda K, Fong GH, Dan T, Ichinose M, Miyata T, Yamamoto M, Suzuki N (2015) Hypoxia signaling cascade for erythropoietin production in hepatocytes. Molecular and Cellular Biology. 35(15): 2658-2672. https://doi.org/10.1128/MCB.00161-15 [PubMed] [PMC]

■ Toyoda T, Itai T, Arakawa T, Aoki KH, Yamaguchi H (2000) Stabilization of human recombinant erythropoietin through interactions with the highly branched N-glycans. The Journal of Biochemistry 128(5): 731-737. [PubMed]

■ Uversky VN, Redwan EM (2017) Erythropoietin and co.: intrinsic structure and functional disorder. Molecular BioSystems 13: 56-72. https://doi.org/10.1039/C6MB00657D

■ van der Kooij MA, Groenendaal F, Kavelaars A, Heijnen CJ, van Bel F (2008) Neuroprotective properties and mechanisms of eryth-ropoietin in in vitro and in vivo experimental models for hypox-ia/ischemia. Brain Research Reviews 59(1): 22-33. https://doi. org/10.1016/j.brainresrev.2008.04.007 [PubMed]

■ Vazquez-Mellado MJ, Monjaras-Embriz V, Rocha-Zavaleta L

(2017) Erythropoietin, stem cell factor, and cancer cell migration. Vitamins and Hormones 105: 273-296. https://doi.org/10.1016/ bs.vh.2017.02.008 [PubMed]

■ Vinberg M, Miskowiak K, Hoejman P, Pedersen M, Kessing LV (2015) The effect of recombinant erythropoietin on plasma brain derived neurotrophic factor levels in patients with affective disorders: a randomised controlled study. PLoS One 10(5): e0127629. https://doi.org/10.1371/journal.pone.0127629 [PubMed]

■ Vogt C, Pentz S, Rich IN (1989) A role for the macrophage in normal hemopoiesis: III. In vitro and in vivo erythropoietin gene expression in macrophages detected by in situ hybridization. Experimental Hematology 17(5): 391-397. [PubMed]

■ Wagner W, Horn P, Castoldi M, Diehlmann A, Bork S, Saffrich R, Benes V, Blake J, Pfister S, Eckstein V, Ho AD (2008) Rep-licative senescence of mesenchymal stem cells: a continuous and organized process. PLoS One 3(5): e2213. https://doi.org/10.1371/ journal.pone.0002213 [PubMed] [PMC]

■ Wang J, Rao S, Chu J, Shen X, Levasseur DN, Theunissen TW, Orkin SH (2006) A protein interaction network for pluripotency of embryonic stem cells. Nature 444(7117): 364-368. https://doi. org/10.1038/nature05284 [PubMed]

■ Wang L, Wu F, Song Y, Duan Y, Jin Z (2018) Erythropoietin induces the osteogenesis of periodontal mesenchymal stem cells from healthy and periodontitis sources via activation of the p38 MAPK pathway. International Journal of Molecular Medicine 41(2): 829835. https://doi.org/10.3892/ijmm.2017.3294 [PubMed] [PMC]

■ Wang Y, Li J, Wang Y, Lei L, Jiang C, An S, Zhan Y, Cheng Q, Zhao Z, Wang J, Jiang L (2012) Effects of hypoxia on osteogenic differentiation of rat bone marrow mesenchymal stem cells. Molecular and Cellular Biochemistry 362(1-2): 25-33. https://doi. org/10.1007/s11010-011-1124-7 [PubMed] [PMC]

■ Westenfelder C, Biddle DL, Baranowski RL (1999) Human, rat, and mouse kidney cells express functional erythropoietin recep-

tors. Kidney International 55(3): 808-820. https://doi.org/10.1046/ j.1523-1755.1999.055003808.x [PubMed]

■ Witthuhn BA, Silvennoinen O, Miura O, Lai KS, Cwik C, Liu ET, Ihle JN (1994) Involvement of the Jak-3 Janus kinase in signalling by interleukins 2 and 4 in lymphoid and myeloid cells. Nature 370(6485): 153-157. https://doi.org/10.1038/370153a0 [PubMed]

■ Wu C, Giaccia AJ, Rankin EB (2014) Osteoblasts: a novel source of erythropoietin. Current Osteoporosis Reports 12(4): 428-432. https://doi.org/10.1007/s11914-014-0236-x [PubMed] [PMC]

■ Wu H, Lee SH, Gao J, Liu X, Iruela-Arispe ML (1999) Inactiva-tion of erythropoietin leads to defects in cardiac morphogenesis. Development 126(16): 3597-3605. [PubMed]

■ Ye L, Chen L, Yu Q, Cheng F (2010) Effect of recombinant human erythropoietin on the stemness of bone marrow-derived mes-enchymal stem cells in vitro. International Journal of Stem Cells 3(2): 175-182. [PubMed] [PMC]

■ Zanjani ED, Poster J, Burlington H, Mann LI, Wasserman LR (1977) Liver as the primary site of erythropoietin formation in the fetus. The Journal of Laboratory and Clinical Medicine 89(3): 640-644. [PubMed]

■ Zheng F, Cheng L, Yu Q, Liu Q, Cheng F (2009) The primary study of CD90(+)CD34(-) and Sca-1(+) stem cells mobilized by EPO plus G-CSF in mice. International Journal of Stem Cells 2(2): 129-134. [PMC]

■ Zwezdaryk KJ, Coffelt SB, Figueroa YG, Liua J, Phinney DG, LaMarca HL, Florez L, Morris CB, Hoyle GW, Scandurro AB (2007) Erythropoietin, a hypoxia-regulated factor, elicits a pro-an-giogenic program in human mesenchymal stem cells. Experimental Hematology 35(4): 640-652. https://doi.org/10.1016/j. exphem.2007.01.044 [PubMed]

Author contributions

■ Ekaterina V. Zubareva, PhD in Biology, Associate Professor, e-mail: zubareva@bsu.edu.ru, ORCID ID: 0000-0002-6480-7810.The author made substantial contributions to the conception of the article and participated in drafting the article.

■ Sergey V. Nadezhdin, PhD in Biology, Associate Professor, e-mail: nadezhdin@bsu.edu.ru, ORCID ID: 00000001-8444-6330. The author made substantial contributions to the conception of the article, participated in drafting the article and gave final approval of the version to be submitted.

■ Yuriy E. Burda, PhD in Medicine, Associate Professor, e-mail: yu.burda@brc.efko.ru, ORCID ID: 0000-00021183-4436. The author made substantial contributions to the conception of the article and participated in drafting the article.

■ Natalia A. Nadezhdina, otorhinolaryngologist, e-mail: nadezhdina.nat@yandex.ru, ORCID ID: 0000-00026425-3635. The author made substantial contributions to the conception of the article and participated in drafting the article.

■ Anastasia S. Gashevskaya, laboratory assistant, e-mail: gashevskayanastia@gmail.com. The author was engaged in the analysis of the research topic references, graphic material design, handling, care and observation of laboratory animals.

i Надоели баннеры? Вы всегда можете отключить рекламу.