Научная статья на тему 'NON-HEMATOPOIETIC ERYTHROPOIETIN-DERIVED PEPTIDES FOR ATHEROPROTECTION AND TREATMENT OF CARDIOVASCULAR DISEASES'

NON-HEMATOPOIETIC ERYTHROPOIETIN-DERIVED PEPTIDES FOR ATHEROPROTECTION AND TREATMENT OF CARDIOVASCULAR DISEASES Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
103
18
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
ATHEROSCLEROSIS / CYTOPROTECTION / EPOR/CD131 / ERYTHROPOIETIN

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Belyaeva Veronika S., Stepenko Yulia V., Lyubimov Igor I., Kulikov Alexandr L., Tietze Alesia A.

Relevance: Cardiovascular diseases continue to be the leading cause of premature adult death. Lipid profile and atherogenesis: Dislipidaemia leads to subsequent lipid accumulation and migration of immunocompetent cells into the vessel intima. Macrophages accumulate cholesterol forming foam cells - the morphological substrate of atherosclerosis in its initial stage. Inflammation and atherogenesis: Pro-inflammatory factors provoke oxidative stress, vascular wall damage and foam cells formation. Endothelial and mitochondrial dysfunction in the development of atherosclerosis: Endothelial mitochondria are some of the organelles most sensitive to oxidative stress. Damaged mitochondria produce excess superoxide and H2O2, which are the main factors of intracellular damage, further increasing endothelial dysfunction. Short non-hematopoietic erythropoietin-based peptides as innovative atheroprotectors: Research in recent decades has shown that erythropoietin has a high cytoprotective activity, which is mainly associated with exposure to the mitochondrial link and has been confirmed in various experimental models. There is also a short-chain derivative, the 11-amino acid pyroglutamate helix B surface peptide (PHBSP), which selectively binds to the erythropoietin heterodymic receptor and reproduces its cytoprotective properties. This indicates the promising use of short-chain derivatives of erythropoietin for the treatment and prevention of atherosclerotic vascular injury. In the future, it is planned to study the PHBSP derivatives, the modification of which consists in adding RGD and PGP tripeptides with antiaggregant properties to the original 11-member peptide. Copyright Author A et al. 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.

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 «NON-HEMATOPOIETIC ERYTHROPOIETIN-DERIVED PEPTIDES FOR ATHEROPROTECTION AND TREATMENT OF CARDIOVASCULAR DISEASES»

# Research Results in Pharmacology

Research Results in Pharmacology 6(3): 75-86

UDC: 615.27 DOI 10.3897/rrpharmacology. 6.58891

Research Article

Non-hematopoietic erythropoietin-derived peptides for atheroprotection and treatment of cardiovascular diseases

Veronika S. Belyaeva1, Yulia V. Stepenko1, Igor I. Lyubimov2, Alexandr L. Kulikov1, Alesia A. Tietze3, Indira S. Kochkarova1, Olga V. Martynova4, Olga N. Pokopeyko5, Liliya A. Krupen'kina1, Andrey S. Nagikh1 , Vladimir M. Pokrovskiy1 , Evgeniy A. Patrakhanov1 , Anastasia V. Belashova1, Petr R. Lebedev1, Anastasia V. Gureeva6

1 Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, 85 Pobedy St., Belgorod 308015 Russia

2 Gurus BioPharm LLC, 42/1 Bolshoi Blvd., Skolkovo, Moscow 143026Russia

3 University of Gothenburg, 9C Medicine Aregatan St., PO Box 462; Gothenburg, Sweden

4 Research Institute of Pharmacology of Living Systems, Belgorod State National University, 85 Pobedy St., Belgorod 308015 Russia

5 Sechenov University, 19c1, Bol'shayaPirogovskaya Str, Moscow 119146Russia

6 Kursk State Medical University, 3 Karl Marx St., Kursk 305041 Russia

Corresponding author: Veronika S. Belyaeva (nika.beliaeva@yandex.ru)

Academic editor: Tatyana Pokrovskaia ♦ Received 21 July 2020 ♦ Accepted 27 August 2020 ♦ Published 30 September 2020

Citation: Belyaeva VS, Stepenko YuV, Lyubimov II, Kulikov AL, Tietze AA, Kochkarova IS, Martynova OV, Pokopeyko ON, Krupen'kina LA, Nagikh AS, Pokrovskiy VM, Patrakhanov EA, Belashova AV, Lebedev PR, Gureeva AV (2020) Non-hematopoietic erythropoietin-derived peptides for atheroprotection and treatment of cardiovascular diseases. Research Results in Pharmacology 6(3): 75-86. https://doi.org/10.3897/rrpharmacology6.58891

Abstract

Relevance: Cardiovascular diseases continue to be the leading cause of premature adult death.

Lipid profile and atherogenesis: Dislipidaemia leads to subsequent lipid accumulation and migration of immunocompetent cells into the vessel intima. Macrophages accumulate cholesterol forming foam cells - the morphological substrate of atherosclerosis in its initial stage.

Inflammation and atherogenesis: Pro-inflammatory factors provoke oxidative stress, vascular wall damage and foam cells formation.

Endothelial and mitochondrial dysfunction in the development of atherosclerosis: Endothelial mitochondria are some of the organelles most sensitive to oxidative stress. Damaged mitochondria produce excess superoxide and H2O2, which are the main factors of intracellular damage, further increasing endothelial dysfunction.

Short non-hematopoietic erythropoietin-based peptides as innovative atheroprotectors: Research in recent decades has shown that erythropoietin has a high cytoprotective activity, which is mainly associated with exposure to the mitochondrial link and has been confirmed in various experimental models. There is also a short-chain derivative, the 11-amino acid pyroglutamate helix B surface peptide (PHBSP), which selectively binds to the erythropoietin hetero-dymic receptor and reproduces its cytoprotective properties. This indicates the promising use of short-chain derivatives of erythropoietin for the treatment and prevention of atherosclerotic vascular injury. In the future, it is planned to study the PHBSP derivatives, the modification of which consists in adding RGD and PGP tripeptides with antiaggregant properties to the original 11-member peptide.

Copyright Author A et al. 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.

Keywords

Atherosclerosis, cytoprotection, epor/cd131, erythropoietin.

Relevance

The high contribution of cardiovascular pathology to the overall structure of the causes of death and disablement in developed countries calls for in-depth study and improvement of correction methods. At the same time, the main task of research in this area is to find ways to prevent and treat atherosclerotic processes as the main cause of cardiovascular mortality.

Atherosclerotic vascular occlusion, as well as thrombosis and embolism following plaque rupture, lead to partial or complete ischemisation of the affected basin, the clinical consequences of which depend on the localisation and calibre of the vessel. The most significant from an epidemiological point of view is coronary and cerebral vascular lesions, leading to such nosologies as coronary heart disease (CHD) and ischemic stroke (Herrington et al. 2016).

Since the middle of the 20th century, most high-income countries have experienced a sharp decline in mortality due to CHD and brain stroke. For example, in the United Kingdom, cardiovascular mortality rates for middle-aged men (35-69 years old) fell from around 700 per 100,000 per year in 1950 to <200 by 2010, and for middle-aged women from -450 in 1950 to <100 by 2010. Most low-and middle-income countries have also reported a decline in stroke-related deaths over the past few decades. However, trends in CHD mortality in these regions have been less consistent: some countries have reported a decline, while others have reported an increase (especially in some Eastern European and Asian countries) in CHD mortality (Bennett et al. 2014; Moran et al. 2014).

The Global Burden of Disease 2010 Study showed that overal, global age-standardized death rates for both CHD and stroke decreased between 1990 and 2010, with a much higher rate in developed countries than in developing countries (GBD 2013 Mortality and Causes of Death Collaborators 2015). Nevertheless, despite relatively reassuring trends, CHD and ischaemic stroke remain the leading causes of premature adult mortality worldwide, indicating the highest importance of research into the underlying atherosclerotic process.

Lipid profile and atherogenesis

The introduction of effective methods for preventing and correcting the atherosclerotic process directly depends on a multi-level understanding of the pathophysiology of the vessel wall lesion. Molecular-biological, instrumental and other research methods provide consistent information on new aspects of the progression of atherosclerosis and

its clinical complications. These advances have not only deepened the understanding of the details of atherogenesis, but have also led to the discovery of many unexpected facts that have called into question many previously dominant concepts, and allowed the development of new therapeutic approaches to combat cardiovascular disease (Libby et al. 2016).

It is now known that the initial event in the development of atherosclerosis is endothelial damage. Endothelial dysfunction (ED) causes infiltration and accumulation of low-density lipoprotein cholesterol (LDL) in the sub-endothelial space. In the pathological conditions, LDLs can interact with ROS to form oxidized LDL (oxLDLs) (Ketelhuth and Hansson 2011). Modified lipoprotein particles increase the expression of cell adhesion molecules (VCAM-1, P, and E-selectin) in endothelial cells, resulting in the recruitment of white blood cells (mainly monocytes and T-lymphocytes) into the subendothelial space. These inflammatory cells migrate into the intima during the interaction of protein-chemoattractants, such as of monocyte MCP-1, eotaxin, and interferon-y. Monocytes are differentiated into macrophages, express scavewnger receptors CD36, SRA, and LOX-1 and internalize modified lipoproteins. Due to their appearance, macrophages loaded with lipids are called foam cells, and their presence in the arterial wall is a sign of an early atherosclerotic lesion. T-lymphocytes and mast cells that migrate to the intima, along with the foam cells, secrete a wide profile of cytokines, which contribute to inflammation and the formation of reactive oxygen species (ROSs). The growth factors released by these cells and ROSs stimulate the migration of smooth muscle cells and collagen deposition, which leads to the development of atheromatous plaque (Kattoor et al. 2017).

In 1912, at a meeting of the Society of Russian Physicians in St. Petersburg, a major Russian scientist, N.N. Anitschkow, whose name is now used for the prize for the most outstanding research in the field of atherosclerosis, together with S.S. Khalatov, presented the first results of his revolutionary research to identify the relationship between nutritional factors, blood cholesterol levels, and atherosclerosis.

The main environmental factors determining blood cholesterol concentration are the consumption of saturated and polyunsaturated fats as well as cholesterol in food (Keys et al. 1957; Hegsted et al. 1993; Clarke et al. 1997), although metabolic, genetic and other factors also influence cholesterol levels (Dattilo and Kris-Etherton 1992). The geographical prevalence of atherosclerosis-associated diseases demonstrates their high dependence on national diets around the world. Atheroprotective diets associated with a lower cardiovascular risk have been identified

separately (Mozaffarian et al. 2011). Blood cholesterol (in particular, low-density lipoprotein cholesterol) is a widely recognised risk factor for CHD, as confirmed by a large number of prospective observational studies from around the world (Lewington et al. 2007; Baigent et al. 2010).

During the initial phase of atherosclerosis, the process of fat infiltration of the vascular wall (fatty streak stage) is initiated. Macrophages absorb apoB-containing lipoproteins, which break down in lysosomes, whereas excess free cholesterol enters the endoplasmic reticulum and is esterified by a cholesterol enzyme - cholesterol ac-yltransferase. The resulting cholesterol ether is packaged in cytoplasmic lipid droplets, which are characteristic of foam cells (Brown et al. 1979; Brown and Goldstein 1983; Rogacev et al. 2012). The chemical modification of ApoB lipoproteins in the form of oxidation and glycation enhances their absorption through a number of receptors (CD36, acceptor receptor A and a family of lectin-like receptors) that are not affected by negative feedback from excess cholesterol (Moore and Freeman 2006; Younis et al. 2008; Polonikov et al. 2017). Enzyme-mediated ApoE lipoprotein aggregation increases lipid absorption through phagocytosis (Torzewski et al. 2004; Torzewski and Lackner 2006), while native lipoprotein residues can stimulate the formation of foam cells through the ApoE receptor family (LRP1 and VLDLR) (Fujioka et al. 1998; Schwartz and Reaven 2012). Native LDL uptake through liquid-phase pinocytosis may also stimulate the formation of foam cells (Anzinger et al. 2012; Kruth 2013).

In addition to atherogenic lipoproteins, there are also atheroprotective ones, which are responsible for transporting cholesterol away from the vascular wall, thus reducing its lipid infiltration. The greatest importance was demonstrated for the ApoA-I and HDL proteins that transport cholesterol to the liver. In addition to ApoA-I and HDLs, ApoE production by macrophages is crucial in preventing the formation of atherosclerotic lesions. Most ApoE in plasma is produced by the liver, but about 5-10% is synthesized by macrophages (Linton et al. 1998). ApoE serves as a ligand for hepatic capture and clearance of all the lipoproteins in the ApoB group, except LDLs. Knocking out the ApoE gene in mice leads to hypercholesterolemia and spontaneous development of an atherosclerotic lesion (Zhang et al. 1992; Plump et al. 1992). Therefore, ApoE-deficient mice are some of the most routine models for studying the mechanisms of development of atherosclerotic lesions. In studying the role of macrophage ApoE in lipoprotein metabolism, its crucial role in lipoprotein metabolism between the vascular wall and blood was demonstrated, making ApoE one of the most important targets for atherosclerotic treatment. Interestingly, the atheroprotective effect of ApoE is implemented through several mechanisms. First, the expression of ApoE by hematopoietic stem cells reduces the proliferation and infiltration of monocytes into the intima (Murphy et al. 2011). In addition, ApoE reduces lysosomal accumulation of cholesterol by increasing lipase expression (Wu et al. 2007). It is important that

ApoE secretion by macrophages stimulates outflow in the absence and presence of exogenous acceptors, including HDLs and ApoA-I, which do not contain lipids (Mazzone and Reardon 1994; Langer et al. 2000; Huang and Mazzone 2002). Recent studies have shown that macrophatic ApoE facilitates the reverse transport of cholesterol in vivo (Zanotti et al. 2011). It also stimulates the outflow of phospholipids and cholesterol through ABCA1, ABCG1, and SR-BI (Chroni et al 2005; Yancey et al. 2007a; Kim et al. 2007; Lammers et al. 2009). In addition to cholesterol outflow, macrophage ApoE prevents inflammation (Yu et al. 2004; Ali et al. 2005; Jofre-Monseny et al. 2007; Li et al. 2015) and oxidative stress (Miyata and Smith 1996; Maor et al. 2000; Rosenblat et al. 2002; Rosen-blat and Aviram 2002). Local ApoE production is likely to be a critical atheroprotective mechanism, given that the content of atherosclerotic plaques has limited access to ApoA-1 and HDLs dissolved in plasma (Fazio et al. 1997; Fazio et al. 2002). Three common polymorphisms of ApoE have been identified in the human population, which are of predictorial value in assessing the risk of CHD regardless of plasma cholesterol levels (Davignon et al. 1988). ApoE3 is the most common isoform and is functionally similar to mouse ApoE. In contrast to iso-forms ApoE3 and ApoE2, ApoE4 isoform is incapable of either stimulating cholesterol outflow (Cullen et al. 1998; Gong et al. 2002; Okoro et al. 2012) or preventing inflammatory and oxidative activation (Rosenblat and Aviram 2002; Yu et al. 2004; Huebbe et al. 2010). The dysfunction of ApoE4 in the host leads to an increased risk of CHD compared to people expressing ApoE3 or ApoE2 (heterozygotes) (Utermann et al. 1984; Davignon et al. 1988; Song et al. 2004).

Thus, the experience gained from N.N. Anichkov's first work demonstrates that in the pathogenesis of atherosclerosis, correlated (environmental) factors must be considered inextricably linked with genetic predisposition (Zarate et al. 2016). This fact leads to a high proportion of research on cardiovascular diseases in the Genome-Wide Association Studies (GWAS) programme and the introduction of the found associations in the early genetic diagnosis panel. The emergence of GWAS and Mendelian randomization analysis opened a new era in the study of the pathobiology of atherosclerosis. Modern genetics has confirmed the causal role of LDLs in atherogenesis, but has also identified new targets that could revolutionize the treatment of the atherosclerotic process.

Inflammation and atherogenesis

Activation of pro-inflammatory cascades in macrophages and endothelium is an essential link in atherogenesis. Activated macrophages are prone to an increased generation of reactive oxygen species, increased cholesterol capture and cytokine/chemokine secretion, resulting in greater LDL oxidation, endothelial cell activation, mono-cyte recruitment, and foam cell formation (Brand et al.

1993; Wang et al. 2002; Bolick et al. 2009; Adamson and Leitinger 2011; Colin et al. 2014; Peled and Fisher 2014). Oxidative stress, modified lipoproteins and other factors (bioactive lipids, molecular patterns associated with damage, cytokines) stimulate inflammation through their own receptors (Adamson and Leitinger 2011; Chinetti-Gbagu-idi and Staels 2011; Colin et al. 2014). In addition, plasma membrane cholesterol in foam cells increases the transmission of signals through inflammatory receptors (Ye et al. 2011; Tall and Yvan-Charvet 2015). The atherogenic role of IL-ip- and IL-18-mediated inflammatory activation has recently been confirmed (Duewell et al. 2010; Lu and Kakkar 2014): the clinical study has shown that subjects receiving a monoclonal antibody to IL-1p, kanaki-numab, had a significantly lower frequency of repeated cardiovascular events, which did not depend on cholesterol reduction (Ridker et al. 2018).

The formation of macrophage foam cells and transmission of cholesterol-dependent inflammatory signals can be reduced by means of removing cholesterol by athero-protective HDLs and ApoA-Is using a number of mechanisms, including ABCA1-, ABCG1-, and SR-BI-mediat-ed elimination and water diffusion (Ye et al. 2011; Yancey et al. 2007b; Kellner-Weibel and de la Llera-Moya 2011). In addition, HDLs and ApoA-Is protect against atherosclerosis by reducing inflammation using mechanisms independent of cholesterol outflow (Tang et al. 2009; Kratzer et al. 2014). In addition, small non-coding RNAs have been found to influence the development of atherosclerosis by regulating inflammation and/or homeosta-sis of cholesterol in different types of cells in lesion foci (Michell and Vickers 2016; Donaldson et al. 2018). MiR-33a and MiR-33b contribute to atherosclerosis by disrupting cholesterol outflow and promoting inflammatory macrophage conversion (Rayner et al. 2011; Horie et al. 2012; Nishino et al. 2018). Other microRNAs, including MiR-223 and MiR-93, have an atheroprotective effect by increasing cholesterol outflow and facilitating the conversion of macrophage phenotype into anti-inflammatory M2 (Donaldson et al. 2018; Zhuang et al. 2012; Vickers et al. 2014; Ganta et al. 2017).

Endothelial and mitochondrial dysfunction in the development of

atherosclerosis

Currently, studying the pathobiological processes occurring in endothelial cells in atherosclerosis has led to the understanding that one of the most significant factors in atherogenesis is mitochondrial dysfunction. Mitochondria are a cell metabolic centre that synthesises the cell's main energy currency, adenosine 5'-triphosphate (ATP), through an oxidative phosphorylation reaction. However, the role of mitochondria in eukaryotic cells goes beyond their ability to act as metabolic mediators. These cellular organelles regulate various cellular processes, including

proliferation (Mitra et al. 2009), an immune response (Zhou et al. 2011a), apoptotic cell death (Kroemer et al. 2007), and also participate in the transmission of intracellular signals to the nucleus (Al-Mehdi et al. 2012). Cells in mitochondria vessels are involved in the regulation of a wide range of biological processes. Transgenic mice devoid of a number of mitochondrial proteins usually die during embryogenesis, which coincides with the development of the cardiovascular system, or are prone to the development of a cardiovascular pathology (Miller et al. 2010; Shenouda et al. 2011; Dong et al. 2013; Kroller-Schon et al. 2013).

Direct contact between endothelial cells and the bloodstream means that they are particularly vulnerable to damage from molecules circulating in the blood, on the one hand, and that they play an essential "protective" role, on the other hand. ED is associated with the development of almost all vascular diseases. Compared to cells of other types with higher energy needs, the content of mitochondria in endotheliocytes is relatively low. In rat endothelials, for example, mitochondria account for 2-6% of cell volume, whereas in hepatocytes and cardi-omyocytes - 28% and 32%, respectively (Dromparis and Michelakis 2013; Kluge et al. 2013). The low content of mitochondria in endothelium indicates that mitochondri-al-dependent oxidative phosphorylation does not contribute significantly to the energy supply of this tissue. In fact, endotheliocytes receive most of their energy from anaerobic glycolytic glucose metabolism. For example, in cultured endothelial cells, more than 75% of pig aorta ATPs is provided by glycolysis (Culic et al. 1997). Similar data indicate that mitochondria mainly serve as important signaling organelles in endothelial and vascular vessels (Quintero et al. 2006).

A large number of studies show a causal relationship between mitochondrial and endothelial dysfunctions. Phenotypic endotheliocyte dysfunction is highly correlated with mitochondrial biogenesis dysfunction, reduced mitochondrial mass and altered expression of electron transport chain (ETC) components (Ungvari et al. 2008; Dai et al. 2012). Such processes correspond to natural processes occurring in endotheliocytes during aging. Damaged mitochondria produce excess superoxide and H2O2, which are the main determinants of cellular aging (Passos et al. 2007).

Mitochondria are an important source of ROSs and at the same time have an antioxidant function. Although the majority of electrons passing through the redox gradient of the ETC eventually reach the V complex, 1-3% of electrons react prematurely with oxygen in complexes I and III to form superoxide and other types of ROSs known as mROSs (Quintero et al. 2006). Mitochondrial ROSs (mROSs) play a signal role in initiating cellular responses to stress. Changing transmembrane potential is an important factor that triggers excess mROS production under pathological conditions, including hypercholesterolemia, hyperglycemia, smoking, infections, and hypoxia. In ad-

dition to complexes I and III, other sources of mROSs have been identified in endotheliocytes. One of them is NADPH oxidase 4 (Nox4) (Lassegue et al. 2012).

Factors that modulate the production of endothelial mROSs have been proved to participate in atherosclerosis. One of such examples is a protein adapter, p66Shc. The fact that the expression of p66Shc may be relevant to the cardiovascular function is evident from the observation that mice knocked out by p66Shc are protected from ROS-dependent age-related ED (Francia et al. 2004), ED caused by hyperglycemia (Zhou et al. 2011b), and alimentary atherosclerosis (Napoli et al. 2003; Martin-Padura et al. 2008).

Thus, according to the modern understanding of the pathogenesis of cardiovascular diseases, mitochondrial dysfunction plays a significant role in the development of atherosclerosis and related complications (Marzetti et al. 2013; Chistiakov et al. 2018). Such changes in endotheli-ocytes and myocytes of the vessel wall lead to structural and functional disorders, manifested in the progression of atherosclerosis, hypertension and susceptibility to thrombosis (Peng et al. 2019). Dysfunction of mitochondria in other cells, including neurons and cardiomyocytes, leads to a decrease in their resistance to ischaemia, which is manifested in an increase in lethal outcomes against the background of brain stroke, coronary artery occlusion, infarction of kidneys and other organs. The emerging pathogenetic cascade is becoming a relevant target for a pharmacological intervention.

Short erythropoietin-based non-hematopoietic peptides as innovative atheroprotectors

Based on available information on the pathogenesis of atherosclerosis, one approach to treating atherogenesis is to use drugs with cytoprotective and mitochondrial activities. The current arsenal of drugs with a mitochon-drial-oriented mechanism of action includes metabolic enchancers (emoxypine and its derivatives, racetames), metabolic regulators (trimetazidine, mildronate), and an-tioxidants (Skulachev ions, SkQ). Preclinical studies in vitro and in vivo in recent decades have shown that a 34-kDa erythropoietin (EPO), an endogenous stimulant of erythrogenesis has a high cytoprotective activity, which is mostly associated with exposure to the mitochondrial link and has been confirmed in experimental models of ischemic and traumatic lesions of virtually all organs, including endothelium, myocardium, and the brain (Qin et al. 2014; Millet et al. 2016; Nekoui and Blaise 2017). These properties make it possible to consider EPO as an agent with a universal cytoprotective orientation. The remote (hematopoiesis) and paracrine action is associated with the ability of EPO to influence the cell cycle and to reduce the pro-apoptic orientation of cells, but the eryth-

ropoietic action is mediated by the homodymeric receptor complex of the erythropoietin receptor (EPOR/EPOR), while the cytoprotective, antioxidant and anti-inflammatory actions are mediated by the heterodymeric complex of EPOR/CD131 (Brines et al. 2004).

Despite the promising results of preclinical studies, information about the EPO antiapoptic activity received by hundreds of research teams is poorly translated into clinical reality. As a result, the search for molecules that selectively mimic the cytoprotective effects of EPO is one of the important tasks of modern pharmacology. To date, compounds with an amino acid sequence identical to that of EPO, but differing in carbohydrate fragment, have become very common. Such modifications improve the pharmacokinetics and reduce the ability to influence hemopoiesis, preserving the tissue-protective activity of the molecule. However, these drugs have not demonstrated high clinical efficacy as cytoprotectors either, possibly due to their low ability to penetrate his-togematic barriers. A principally novel approach is the search for low-molecular peptide derivatives of EPO, which have an antiapoptic activity and freely penetrate through biological barriers. The first of such derivatives is an 11-amino acid peptide PHBSP, which is an amino acid chain "Pyr-Glu-Gln-Leu-Glu-Arg-Ala-Leu-Asn-Ser-Ser" and has a selective affinity for the heterody-meric complex EPOR/CD131 (Brines et al. 2004). In developing this peptide, the authors relied on the ami-no acid sequence of alpha-spiral B erythropoietin, since this is the only structural part of the molecule that does not participate in the spatial interaction with the homo-dymeric complex EPOR/EPOR. Several studies have shown that PHBSP has more pronounced antiapoptic properties than the basic molecule, exhibits an analgesic activity and does not have a hemopoietic effect intrinsic to EPO (Brines et al. 2004; Swartjes et al. 2014; Zhang et al. 2017; Yang et al. 2019). However, a significant disadvantage of PHBSP is its very short half-life, approaching several minutes. In addition, studies carried out in our laboratory in vivo and in vitro have revealed a pro-thrombotic activity in this peptide, which is to some extent characteristic of all erythropoietin molecules.

Conclusion

In our view, a modification of PHBSP to improve the pharmacokinetic and pharmacodynamic parameters may be a further promising development of a short-chain-pep-tide-based CVD pharmacotherapy. To solve this problem, our team, together with the scientific team of A. Titze (University of Gothenburg), has worked on searching for the ways of modifying the base molecule. To eliminate the pro-thrombotic activity, it was decided to add the amino

acid sequence RGD (Arg-Gly-Asp) to PHBSP. It is known that this motif has pronounced antiaggregant properties (Pytela et al. 1986, Sheu et al. 1995, Hung et al. 2017). In parallel, the PGP motive (Pro-Gly-Pro) was added to a number of compounds to improve their pharmacokinetic characteristics. This amino acid sequence stabilizes the molecule in biological media by inhibiting the activity of proteolytic enzymes (Shevchenko et al. 2019). Moreover, PGP also has the ability to block the angiotensin-conver-ting enzyme (Wang et al. 2011), one of the most important proatherogenic factors that catalyzes the reaction of angiotensin II formation and promotes vascular wall remodeling (Montezano et al. 2014). A number of Russian studies have also identified the antithrombotic and antiaggregant properties of PGP (Pastorova et al. 2001; Lyapina et al. 2007; Liapina et al. 2010), making its adherence to PHBSP even more promising. The key question remains the nature of embedding RGD and PGP motifs in the base molecule. A bioinformatic analysis has made it possible

References

■ Adamson S, Leitinger N (2011) Phenotypic modulation of macrophages in response to plaque lipids. Current Opinion in Lipidology 22(5): 335-342. https://doi.org/10.1097/MOL.0b013e32834a97e4 [PubMed] [PMC]

■ Ali K, Middleton M, Puré E, Rader DJ (2005) Apolipoprotein E suppresses the type I inflammatory response in vivo. Circulation Research 97(9): 922-927. https://doi.org/10.1161/01. RES.0000187467.67684.43 [PubMed]

■ Al-Mehdi AB, Pastukh VM, Swiger BM, Reed DJ, Patel MR, Bardwell GC (2012) Perinuclear mitochondrial clustering creates an oxidant-rich nuclear domain required for hypoxia-induced transcription. Science Signaling 5(231): ra47. https://doi.org/10.1126/ scisignal.2002712 [PubMed] [PMC]

■ Anzinger JJ, Chang J, Xu Q, Barthwal MK, Bohnacker T, Wymann MP, Kruth HS (2012) Murine bone marrow-derived macrophages differentiated with GM-CSF become foam cells by PI3Ky-dependent fluid-phase pinocytosis of native LDL. Journal of Lipid Research 53(1): 34-42. https://doi.org/10.1194/jlr.M018887 [PubMed] [PMC]

■ Bennett DA, Krishnamurthi RV, Barker-Collo S, Forouzanfar MH, Naghavi M, Connor M, Lawes CM, Moran AE, Anderson LM, Roth GA, Mensah GA, Ezzati M, Murray CJ, Feigin VL (2014) Global burden of diseases, injuries, and risk factors 2010 Study Stroke Expert Group. The global burden of ischemic stroke: findings of the GBD 2010 study. Global Heart 9: 107-112. https://doi.org/10.1016/j. gheart.2014.01.001_[PubMed]

■ Bolick DT, Skaflen MD, Johnson LE, Kwon SC, Howatt D, Daugherty A, Ravichandran KS, Hedrick CC (2009) G2A deficiency in mice promotes macrophage activation and atherosclerosis. Circulation Research 104(3): 318-327. https://doi.org/10.1161/CIRCRE-SAHA.108.181131 [PubMed] [PMC]

■ Brand K, Mackman N, Curtiss LK (1993) Interferon-gamma inhibits macrophage apolipoprotein E production by posttranslational mechanisms. The Journal of Clinical Investigation 91(5): 2031-2039. https://doi.org/10.1172/JCI116425 [PubMed] [PMC]

to determine the most optimal localizations for joining tri-peptides, which do not affect the pharmacophores of interest, preserving both cytoprotective (Korokin et al. 2019; Korokin et al. 2020) and antiaggregant activities. Further in vitro and in vivo studies will open up the possibility of screening the most promising compounds.

Acknowledgements

The reported work was funded by the Ministry of Science and Higher Education of the Russian Federation (Agreement No. 05.605.21.0191, unique Agreement id -RFMEFI60519X0191).

Conflict of interest

The authors have no conflict of interest to declare.

■ 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 beta-subu-nit 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]

■ Brown MS, Goldstein JL (1983) Lipoprotein metabolism in the macrophage: implications for cholesterol deposition in atherosclerosis. Annual Review of Biochemistry 52: 223-261. https://doi. org/10.1146/annurev.bi.52.070183.001255 [PubMed]

■ Brown MS, Goldstein JL, Krieger M, Ho YK, Anderson RG (1979) Reversible accumulation of cholesteryl esters in macrophages incubated with acetylated lipoproteins. The Journal of Cell Biology 82(3): 597-613. https://doi.org/10.1083/jcb.823.597 [PubMed] [PMC]

■ Chinetti-Gbaguidi G, Staels B (2011) Macrophage polarization in metabolic disorders: functions and regulation. Current Opinion in Lipidology 22(5): 365-372. https://doi.org/10.1097/MOL.0b013e-32834a77b4 [PubMed] [PMC]

■ Chistiakov DA, Shkurat TP, Melnichenko AA, Grechko AV, Orek-hov AN (2018) The role of mitochondrial dysfunction in cardiovascular disease: A brief review. Annals of Medicine 50(2): 121-127. https://doi.org/10.1080/07853890.2017.1417631 [PubMed]

■ Cholesterol Treatment Trialists' (CTT) Collaboration, Baigent C, Blackwell L, Emberson J, Holland L. E, Reith C, Bhala N, Peto R, Barnes EH, Keech A, Simes J, Collins R (2010) Efficacy and safety of more intensive lowering of LDL cholesterol: a me-ta-analysis of data from 170,000 participants in 26 randomised trials. Lancet 376(9753): 1670-1681. https://doi.org/10.1016/S0140-6736(10)61350-5 [PubMed] [PMC]

■ Chroni A, Nieland TJ, Kypreos KE, Krieger M, Zannis VI (2005) SR-BI mediates cholesterol efflux via its interactions with li-pid-bound ApoE. Structural mutations in SR-BI diminish cholesterol

efflux. Biochemistry 44(39): 13132-13143. https://doi.org/10.1021/ bi051029o [PubMed]

■ Clarke R, Frost C, Collins R, Appleby P, Peto R (1997) Dietary lipids and blood cholesterol: quantitative meta-analysis of metabolic ward studies. BMJ 314(7074): 112-117. https://doi.org/10.1136/ bmj.314.7074.112 [PubMed] [PMC]

■ Colin S, Chinetti-Gbaguidi G, Staels B (2014) Macrophage pheno-types in atherosclerosis. Immunological Reviews 262(1): 153-166. https://doi.org/10.1111/imr.12218

■ Culic O, Gruwel ML, Schrader J (1997) Energy turnover of vascular endothelial cells. American Journal of. Physiology-Cell Physiology 273(1): 205-213. https://doi.org/10.1152/ajpcell.1997.273.LC205 [PubMed]

■ Cullen P, Cignarella A, Brennhausen B, Mohr S, Assmann G, von Eckardstein A (1998) Phenotype-dependent differences in apoli-poprotein E metabolism and in cholesterol homeostasis in human monocyte-derived macrophages. The Journal of Clinical Investigation 101(8): 1670-1677. https://doi.org/10.1172/JCI119887 [Pu-bMed] [PMC]

■ Dai DF, Rabinovitch PS, Ungvari Z (2012) Mitochondria and cardiovascular aging. Circulation Research 110(8): 1109-1124. htpps:// doi.org/10.1161/CIRCRESAHA.111.246140 [PubMed] [PMC]

■ Dattilo AM, Kris-Etherton PM (1992) Effects of weight reduction on blood lipids and lipoproteins: a meta-analysis. The American Journal of Clinical Nutrition 56(2): 320-328. https://doi.org/10.1093/ ajcn/56.2.320 [PubMed]

■ Davignon J, Gregg RE, Sing CF (1988) Apolipoprotein E polymorphism and atherosclerosis. Arteriosclerosis 8(1): 1-21. https://doi. org/10.1161/01.atv.8.1.1

■ Donaldson CJ, Lao KH, Zeng L (2018) The salient role of microR-NAs in atherogenesis. Journal of Molecular and Cellular Cardiology 122: 98-113. https://doi.org/10.1016/j.yjmcc.2018.08.004 [PubMed]

■ Dong M, Yang X, Lim S, Cao Z, Honek J, Lu H (2013) Cold exposure promotes atherosclerotic plaque growth and instability via UCP1-dependent lipolysis. Cell Metabolism 18(1): 118-129. https:// doi.org/10.1016/j.cmet.2013.06.003 [PubMed] [PMC]

■ Dromparis P, Michelakis ED (2013) Mitochondria in vascular health and disease. Annual Review of Physiology 75: 95-126. https://doi. org/10.1146/annurev-physiol-030212-183804 [PubMed]

■ Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nunez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung V, Latz E (2010) NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464(7293): 1357-1361. https://doi.org/10.1038/nature08938 [PubMed] [PMC]

■ Fazio S, Babaev VR, Burleigh ME, Major AS, Hasty AH, Linton MF (2002) Physiological expression of macrophage apoE in the artery wall reduces atherosclerosis in severely hyperlipidemic mice. Journal of Lipid Research 43(10): 1602-1609. https://doi.org/10.1194/ jlr.m200108-jlr200 [PubMed]

■ Fazio S, Babaev VR, Murray AB, Hasty AH, Carter KJ, Gleaves LA, Atkinson JB, Linton MF (1997) Increased atherosclerosis in mice reconstituted with apolipoprotein E null macrophages. Proceedings of the National Academy of Sciences of the United States of America 94(9): 4647-4652. https://doi.org/10.1073/pnas.94.9.4647 [Pu-bMed] [PMC]

■ Francia P, Delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C, Migliaccio E (2004) Deletion of p66shc gene protects against age-re-

lated endothelial dysfunction. Circulation 110(18): 2889-2895. https://doi.org/10.1161/01.CIR.0000147731.24444.4D [PubMed]

■ Fujioka Y, Cooper AD, Fong LG (1998) Multiple processes are involved in the uptake of chylomicron remnants by mouse peritoneal macrophages. Journal of Lipid Research 39(12): 2339-2349. [PubMed]

■ Ganta VC, Choi MH, Kutateladze A, Fox TE, Farber CR, Annex BH (2017) A microRNA93-interferon regulatory factor-9-immuno-responsive hene-1-itaconic acid pathway modulates M2-like macrophage polarization to revascularize ischemic muscle. Circulation 135(24): 2403-2425. https://doi.org/10.1161/CIRCULATIONA-HA.116.025490 [PubMed] [PMC]

■ GBD (2013) Mortality and Causes of Death Collaborators (2015) Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 385(9963): 117-171. https://doi.org/10.1016/S0140-6736(14)61682-2 [PubMed] [PMC]

■ Gong JS, Kobayashi M, Hayashi H, Zou K, Sawamura N, Fujita SC, Yanagisawa K, Michikawa M (2002) Apolipoprotein E (ApoE) isoform-dependent lipid release from astrocytes prepared from human ApoE3 and ApoE4 knock-in mice. The Journal of Biological Chemistry 277(33): 29919-29926. https://doi.org/10.1074/jbc. M203934200 [PubMed]

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

■ Hegsted DM, Ausman LM, Johnson JA, Dallal GE (1993) Dietary fat and serum lipids: an evaluation of the experimental data. The American Journal of Clinical Nutrition 57(6): 875-883. https://doi. org/10.1093/ajcn/57.6.875 [PubMed]

■ Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S (2016) Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease. Circulation Research 118: 535-546. https://doi.org/10.1161/CIRCRESAHA.115.307611 [PubMed]

■ Horie T, Baba O, Kuwabara Y, Chujo Y, Watanabe S, Kinoshita M, Horiguchi M, Nakamura T, Chonabayashi K, Hishizawa M, Haseg-awa K, Kume N, Yokode M, Kita T, Kimura T, Ono K (2012) Mi-croRNA-33 deficiency reduces the progression of atherosclerotic plaque in ApoE-/- mice. Journal of the American Heart Association 1(6): e003376. https://doi.org/10.1161/JAHA.112.003376 [PubMed] [PMC]

■ Huang ZH, Mazzone T (2002) ApoE-dependent sterol efflux from macrophages is modulated by scavenger receptor class B type I expression. Journal of Lipid Research 43(3): 375-382. [PubMed]

■ Huebbe P, Lodge JK, Rimbach G (2010) Implications of apolipoprotein E genotype on inflammation and vitamin E status. Molecular Nutrition & Food Research 54(5): 623-630. https://doi.org/10.1002/ mnfr.200900398 [PubMed]

■ Hung YC, Kuo YJ, Huang SS, Huang TF (2017) Trimucrin, an Arg-Gly-Asp containing disintegrin, attenuates myocardial ischemia-rep-erfusion injury in murine by inhibiting platelet function. European Journal of Pharmacology 813: 24-32. https://doi.org/10.1016/j. ejphar.2017.07.039 [PubMed]

■ Jofre-Monseny L, Loboda A, Wagner AE, Huebbe P, Boesch-Saa-datmandi C, Jozkowicz A, Minihane AM, Dulak J, Rimbach G (2007) Effects of apoE genotype on macrophage inflammation and heme oxygenase-1 expression. Biochemical and Biophysical Research Communications 357(1): 319-324. https://doi.org/10.1016/j. bbrc.2007.03.150 [PubMed] [PMC]

■ Kattoor AJ, Pothineni N, Palagiri D, Mehta JL (2017) Oxidative stress in atherosclerosis. Current Atherosclerosis Reports 19(11): 42. https://doi.org/10.1007/s11883-017-0678-6 [PubMed]

■ Kellner-Weibel G, de la Llera-Moya M (2011) Update on HDL receptors and cellular cholesterol transport. Current Atherosclerosis Reports 13(3): 233-241. https://doi.org/10.1007/s11883-011-0169-0 [PubMed]

■ Ketelhuth DF, Hansson GK (2011) Cellular immunity, low-density lipoprotein and atherosclerosis: break of tolerance in the artery wall. Thrombosis and Haemostasis 106(5): 779-786. https://doi. org/10.1160/TH11-05-0321 [PubMed]

■ Keys A, Anderson JT, Grande F (1957) Prediction of serum-cholesterol responses of man to changes in fats in the diet. Lancet 273(7003): 959-966. https://doi.org/10.1016/s0140-6736(57)91998-0 [PubMed]

■ Kim WS, Rahmanto AS, Kamili A, Rye KA, Guillemin GJ, Gelissen IC, Jessup W, Hill AF, Garner B (2007) Role of ABCG1 and ABCA1 in regulation of neuronal cholesterol efflux to apolipoprotein E discs and suppression of amyloid-beta peptide generation. The Journal of Biological Chemistry 282(5): 2851-2861. https://doi.org/10.1074/ jbc.M607831200 [PubMed]

■ Kluge MA, Fetterman JL, Vita JA (2013) Mitochondria and endothelial function. Circulation Research 112(8): 1171-1188. https://doi. org/10.1161/CIRCRESAHA.111.300233_[PubMed] [PMC]

■ Korokin M, Gureev V, Gudyrev O, Golubev I, Korokina L, Pere-sypkina A, Pokrovskaia T, Lazareva G, Soldatov V, Zatolokina M, Pobeda A, Avdeeva E, Beskhmelnitsyna E, Denisyuk T, Avdeeva N, Bushueva O, Pokrovskii M (2020) Erythropoietin mimetic peptide (pHBSP) corrects endothelial dysfunction in a rat model of preec-lampsia. International Journal of Molecular Sciences 21(18): E6759. https://doi.org/10.3390/ijms21186759 [PubMed]

■ Korokin M, Soldatov V, Tietze A, Golubev M, Belykh A, Kubekina M, Puchenkova O, Denisyuk T, Gureyev V, Pokrovskaya T, Gudyrev O, Zhuchenko M, Zatolokina M, Pokrovskiy M (2019) 11-amino acid peptide imitating the structure of erythropoietin a-helix b improves endothelial function, but stimulates thrombosis in rats. Pharmacy & Pharmacology 7(6): 312-320. https://doi.org/10.19163/2307-9266-2019-7-6-312-320

■ Kratzer A, Giral H, Landmesser U (2014) High-density lipoproteins as modulators of endothelial cell functions: alterations in patients with coronary artery disease. Cardiovascular Research 103(3): 350361. https://doi.org/10.1093/cvr/cvu139 [PubMed]

■ Kroemer G, Galluzzi L, Brenner C (2007) Mitochondrial membrane permeabilization in cell death. Physiological Reviews 87(1): 99163. https://doi.org/10.1152/physrev.00013.2006 [PubMed]

■ Kröller-Schön S, Jansen T, Schüler A, Oelze M, Wenzel P, Hausding M (2013) Proliferator coactivator 1a deletion induces angiotensin II - associated vascular dysfunction by increasing mitochondrial ox-idative stress and vascular inflammation. Arteriosclerosis, Thrombosis, and Vascular Biology 33(8): 1928-1935. https://doi.org/10.1161/ ATVBAHA.113.301717 [PubMed]

■ Kruth HS (2013) Fluid-phase pinocytosis of LDL by macrophages: a novel target to reduce macrophage cholesterol accumulation in atherosclerotic lesions. Current Pharmaceutical Design 19(33): 5865-5872. https://doi.org/10.2174/1381612811319330005 [PubMed] [PMC]

■ Lammers B, Out R, Hildebrand RB, Quinn CM, Williamson D, Hoekstra M, Meurs I, Van Berkel TJ, Jessup W, Van Eck M (2009) Independent protective roles for macrophage Abcg1 and Apoe in the

atherosclerotic lesion development. Atherosclerosis 205(2): 420426. https://doi.org/10.1016/j.atherosclerosis.2009.01.017 [PubMed]

■ Langer C, Huang Y, Cullen P, Wiesenhutter B, Mahley RW, Assmann G, von Eckardstein A (2000) Endogenous apolipoprotein E modulates cholesterol efflux and cholesteryl ester hydrolysis mediated by high-density lipoprotein-3 and lipid-free apolipoproteins in mouse peritoneal macrophages. Journal of Molecular Medicine 78(4): 217227. https://doi.org/10.1007/s001090000096 [PubMed]

■ Lassegue B, Martin AS, Griendling KK (2012) Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular. Circulation Research 110(10): 1364-1390. https://doi. org/10.1161/CIRCRESAHA.111.243972 [PubMed] [PMC]

■ Li K, Ching D, Luk FS, Raffai RL (2015) Apolipoprotein E enhances microRNA-146a in monocytes and macrophages to suppress nuclear factor-KB-driven inflammation and atherosclerosis. Circulation Research 117(1): e1-e11. https://doi.org/10.1161/CIRCRESA-HA.117.305844 [PubMed] [PMC]

■ Liapina LA, Grigor'eva ME, Andreeva LA, Miasoedov NF (2010) Protective antithrombotic effects of proline-containing peptides in the animal body subjected to stress. Biology Bulletin of the Russian Academy of Sciences 37(4): 462-467. https://doi.org/10.1134/ S1062359010040096 [PubMed]

■ Libby P, Bornfeldt KE, Tall AR (2016) Atherosclerosis: successes, surprises, and future challenges. Circulation Research 118(4): 531534. https://doi.org/10.1161/CIRCRESAHA.116.308334_ [PubMed] [PMC]

■ Linton MF, Hasty AH, Babaev VR, Fazio S (1998) Hepatic apo E expression is required for remnant lipoprotein clearance in the absence of the low density lipoprotein receptor. The Journal of Clinical Investigation 101(8): 1726-1736. https://doi.org/10.1172/JCI2181 [PubMed] [PMC]

■ Lu X, Kakkar V (2014) Inflammasome and atherogenesis. Current Pharmaceutical Design 20(1): 108-124. https://doi.org/10.2174/138 16128113199990586 [PubMed]

■ Lyapina LA, Pastorova VE, Obergan TY (2007) Changes in he-mostatic parameters after intranasal administration of peptide Pro-Gly-Pro. Bulleting of Experimental Biolody and Medicine 144(4): 491-493. https://doi.org/10.1007/s10517-007-0358-6

■ Maor I, Kaplan M, Hayek T, Vaya J, Hoffman A, Aviram M (2000) Oxidized monocyte-derived macrophages in aortic atherosclerotic lesion from apolipoprotein E-deficient mice and from human carotid artery contain lipid peroxides and oxysterols. Biochemical and Biophysical Research Communications 269(3): 775-780. https://doi. org/10.1006/bbrc.2000.2359 [PubMed]

■ Martin-Padura I, Nigris F, Migliaccio E, Mansueto G, Minardi S, Rienzo M (2008) Deletion confers vascular protection in advanced atherosclerosis in hypercholesterolemic apolipoprotein E knockout mice. Endothelium 15(5-6): 276-287. https://doi. org/10.1080/10623320802487791 [PubMed]

■ Marzetti E, Csiszar A, Dutta D, Balagopal G, Calvani R, Leeu-wenburgh C (2013) Role of mitochondrial dysfunction and altered autophagy in cardiovascular aging and disease: from mechanisms to therapeutics. American Journal of Physiology - Heart Circulatory Physiology 305(4): 459-476. https://doi.org/10.1152/ ajpheart.00936.2012_[PubMed] [PMC]

■ Mazzone T, Reardon C (1994) Expression of heterologous human apolipoprotein E by J774 macrophages enhances cholesterol efflux to HDL3. Journal of Lipid Research 35(8): 1345-1353. [PubMed]

■ Michell DL, Vickers KC (2016) Lipoprotein carriers of microRNAs. Biochimica et Biophysica Acta 1861(12 Pt B): 2069-2074. https:// doi.org/10.1016/j.bbalip.2016.01.011 [PubMed] [PMC]

■ Miller JD, Peotta VA, Chu Y, Weiss RM, Zimmerman K, Brooks RM (2010) MnSOD protects against COX1-mediated endothelial dysfunction in chronic heart failure. American Journal of Physiology-Heart Circulatory Physiology 298(5): 1600-1607. https://doi. org/10.1152/ajpheart.01108.2009 [PubMed] [PMC]

■ Millet A, Bouzat P, Trouve-Buisson T, Batandier C, Pernet-Gallay K, Gaide-Chevronnay L (2016) Erythropoietin and its derivates modulate mitochondrial dysfunction after diffuse traumatic brain injury. Journal of Neurotrauma 33(17): 1625-1633. https://doi.org/10.1089/ neu.2015.4160_ [PubMed]

■ Mitra K, Wunder C, Roysam B, Lin G, Lippincott-Schwartz J (2009) A hyperfused mitochondrial state achieved at G1-S regulates cyclin E buildup and entry into S phase. Proceedings of the National Academy of Sciences of the United States of America 106(29): 1196011965. https://doi.org/10.1073/pnas.0904875106 [PubMed] [PMC]

■ Miyata M, Smith JD (1996) Apolipoprotein E allele-specific antioxidant activity and effects on cytotoxicity by oxidative insults and beta-amyloid peptides. Nature Genetics 14(1): 55-61. https://doi. org/10.1038/ng0996-55 [PubMed]

■ Montezano AC, Nguyen Dinh Cat A, Rios FJ, Touyz RM (2014) Angiotensin II and vascular injury. Current Hypertension Reports 16(6): 431. https://doi.org/10.1007/s11906-014-0431-2 [PubMed]

■ Moore KJ, Freeman MW (2006) Scavenger receptors in atherosclerosis: beyond lipid uptake. Arteriosclerosis, Thrombosis, and Vascular Biology 26(8): 1702-1711 https://doi.org/10.1161/01. ATV.0000229218.97976.43 [PubMed]

■ Moran AE, Forouzanfar MH, Roth GA, Mensah GA, Ezzati M, Murray CJ, Naghavi M (2014) Temporal trends in ischemic heart disease mortality in 21 world regions, 1980 to 2010: the Global Burden of Disease 2010 study. Circulation 129: 1483-1492. https://doi. org/10.1161/CIRCULATIONAHA.113.004042_[PubMed] [PMC]

■ Mozaffarian D, Appel LJ, Van Horn L (2011) Components of a cardioprotective diet: new insights. Circulation 123(24): 2870-2891. https://doi.org/10.1161/CIRCULATI0NAHA.110.968735 [Pu-bMed] [PMC]

■ Murphy AJ, Akhtari M, Tolani S, Pagler T, Bijl N, Kuo CL, Wang M, Sanson M, Abramowicz S, Welch C, Bochem AE, Kuivenhoven JA, Yvan-Charvet L, Tall AR (2011) ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. The Journal of Clinical Investigation 121(10): 4138-4149. https://doi.org/10.1172/JCI57559 [PubMed] [PMC]

■ Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, Somma P (2003) Deletion of the p66Shc longevity gene reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early atherogenesis in mice fed a high-fat diet. Proceedings of the National Academy of Sciences of the United States of America 100(4): 21122116. https://doi.org/10.1073/pnas.0336359100JPubMed] [PMC]

■ Nekoui A, Blaise G (2017) Erythropoietin and nonhematopoietic effects. The American Journal of the Medical Sciences 353(1):76-81. https://doi.org/10.1016/j.amjms.2016.10.009 [PubMed]

■ Nishino T, Horie T, Baba O, Sowa N, Hanada R, Kuwabara Y, Nakao T, Nishiga M, Nishi H, Nakashima Y, Nakazeki F, Ide Y, Koyama S, Kimura M, Nagata M, Yoshida K, Takagi Y, Nakamura T, Hasegawa K, Miyamoto S, Kimura T, Ono K (2018) SREBF1/MicroRNA-33b Axis Exhibits Potent Effect on Unstable Atherosclerotic Plaque For-

mation In Vivo. Arteriosclerosis, Thrombosis, and Vascular Biology 38(10): 2460-2473. https://doi.org/10.1161/ATVBAHA.118.311409 [PubMed]

■ Okoro EU, Zhao Y, Guo Z, Zhou L, Lin X, Yang H (2012) Apolipoprotein E4 is deficient in inducing macrophage ABCA1 expression and stimulating the Sp1 signaling pathway. PloS one 7(9): e44430. https://doi.org/10.1371/journal.pone.0044430 [PubMed] [PMC]

■ Passos JF, Saretzki G, Ahmed S, Nelson G, Richter T, Peters H (2007) Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biology 5(5): 2889-2895e110. https://doi.org/10.1371/journal.pbio.0050110 [Pu-bMed] [PMC]

■ Pastorova VE, Liapina LA, Alshmarin IP, Ostrovskaia PU, Gudashe-va TA, Lugovskoi EV (2001) Fibrin-depolymerization activity and the antiplatelet effect of small cyclic and linear proline-containing peptides. Biology Bulletin of the Russian Academy of Sciences 28(5): 593-596. https://doi.org/10.1023/A:1016700412205 [PubMed]

■ Peled M, Fisher EA (2014) Dynamic Aspects of Macrophage Polarization during Atherosclerosis Progression and Regression. Frontiers in Immunology 5: 579. https://doi.org/10.3389/fimmu.2014.00579 [PubMed] [PMC]

■ Peng W, Cai G, Xia Y, Chen J, Wu P, Wang Z, Li G, Wei D (2019) Mitochondrial dysfunction in atherosclerosis. DNA and Cell Biology 38(7): 597-606. https://doi.org/10.1089/dna.2018.4552 [PubMed]

■ Plump AS, Smith JD, Hayek T, Aalto-Setälä K, Walsh A, Verstuyft JG, Rubin EM, Breslow JL (1992) Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell 71(2): 343-353. https://doi. org/10.1016/0092-8674(92)90362-g [PubMed]

■ Polonikov A, Bykanova M, Ponomarenko I, Sirotina S, Bocharova A, Vagaytseva K, Stepanov V, Churnosov M, Bushueva O, Solodilo-va M, Shvetsov Y, Ivanov V (2017) The contribution of CYP2C gene subfamily involved in epoxygenase pathway of arachidonic acids metabolism to hypertension susceptibility in Russian population. Clinical and Experimental Hypertension 39(4): 306-311. https://doi. org/10.1080/10641963.2016.1246562 [PubMed]

■ Prospective Studies Collaboration, Lewington S, Whitlock G, Clarke R, Sherliker P, Emberson J, Halsey J, Qizilbash N, Peto R, Collins R (2007) Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths. Lancet 370(9602): 18291839. https://doi.org/10.1016/S0140-6736(07)61778-4 [PubMed]

■ Pytela R, Pierschbacher MD, Ginsberg MH, Plow EF, Ruoslahti E (1986) Platelet membrane glycoprotein IIb/IIIa: member of a family of Arg-Gly-Asp-specific adhesion receptors. Science 231(4745): 1559-1562. https://doi.org/10.1126/science.2420006 [PubMed]

■ Qin C, Zhou S, Xiao Y, Chen L (2014) Erythropoietin enhances mi-tochondrial biogenesis in cardiomyocytes exposed to chronic hypox-ia through Akt/eNOS signalling pathway. Cell Biology International 38(3): 335-342. https://doi.org/10.1002/cbin.10205 [PubMed]

■ Quintero M, Colombo SL, Godfrey A, Moncada S (2006) Mitochondria as signaling organelles in the vascular endothelium. Proceeding of the. National Academy of Sciences of the United States of America 103(14): 5379-5384. https://doi.org/10.1073/pnas.0601026103 [PubMed] [PMC]

■ Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, Parathath S, van Gils JM, Rayner AJ, Chang AN, Suarez Y, Fernandez-Hernando C, Fisher EA, Moore KJ (2011) Antagonism of miR-33 in

mice promotes reverse cholesterol transport and regression of atherosclerosis. The Journal of Clinical Investigation 121(7): 2921-2931. https://doi.org/10.1172/JCI57275JPubMed] [PMC]

■ Ridker PM, MacFadyen JG, Everett BM, Libby P, Thuren T, Glynn RJ, CANTOS Trial Group (2018) Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet 391(10118): 319-328. https://doi. org/10.1016/S0140-6736(17)32814-3 [PubMed]

■ Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Große-Dunker G, Heisel I, Hornof F, Jeken J, Rebling NM, Ulrich C, Scheller B, Böhm M, Fliser D, Heine GH (2012) CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. Journal of the American College of Cardiology 60(16): 1512-1520. https://doi.org/10.1016/jjacc.2012.07.019JPubMed]

■ Rosenblat M, Aviram M (2002) Oxysterol-induced activation of macrophage NADPH-oxidase enhances cell-mediated oxidation of LDL in the atherosclerotic apolipoprotein E deficient mouse: inhibitory role for vitamin E. Atherosclerosis 160(1): 69-80. https://doi. org/10.1016/s0021-9150(01)00563-9 [PubMed]

■ Rosenblat M, Coleman R, Aviram M (2002) Increased macrophage glutathione content reduces cell-mediated oxidation of LDL and atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis 163(1): 17-28. https://doi.org/10.1016/s0021-9150(01)00744-4 [PubMed]

■ Schwartz EA, Reaven PD (2012) Lipolysis of triglyceride-rich lipoproteins, vascular inflammation, and atherosclerosis. Biochimica et Biophysica Acta 1821(5): 858-866. https://doi.org/10.1016Zj.bba-lip.2011.09.021 [PubMed]

■ Shenouda SM, Widlansky ME, Chen K, Xu G, Holbrook M, Tabit CE (2011) Altered mitochondrial dynamics contributes to endothelial dysfunction in diabetes mellitus. Circulation 124(4): 444—453. https://doi.org/10.1161/CIRCULATIONAHA.110.014506 [PubMed] [PMC]

■ Sheu JR, Yen MH, Peng IC, Chang MC, Huang TF (1995) Triflavin, an Arg-Gly-Asp-containing peptide, prevents platelet plug formation in vivo by blocking glycoprotein IIb/IIIa complex. European Journal of Pharmacology 294(1): 231-238. https://doi.org/10.1016/0014-2999(95)00530-7 [PubMed]

■ Shevchenko KV, Nagaev IY, Andreeva LA, Shevchenko VP, My-asoedov NF (2019) Stability of prolin-containing peptides in biological media. Biochemistry (Moscow), Supplement Series B: Biomedical Chemistry 65(3): 180-201. https://doi.org/10.18097/ PBMC20196503180 [PubMed]

■ Song Y, Stampfer MJ, Liu S (2004) Meta-analysis: apolipoprotein E genotypes and risk for coronary heart disease. Annals of Internal Medicine 141(2): 137-147. https://doi.org/10.7326/0003-4819-141-2-200407200-00013 [PubMed]

■ Swartjes M, Velzen M, Niesters M (2014) ARA 290, a peptide derived from the tertiary structure of erythropoietin, produces long-term relief of neuropathic pain coupled with suppression of the spinal microglia response. Molecular Pain 10: 1744-1769. https://doi. org/10.1186/2F174480691013 [PubMed] [PMC]

■ Tall AR, Yvan-Charvet L (2015) Cholesterol, inflammation and innate immunity. Nature Reviews. Immunology 15(2): 104-116. https://doi.org/10.1038/nri3793 [PubMed] [PMC]

■ Tang C, Liu Y, Kessler PS, Vaughan AM, Oram JF (2009) The macrophage cholesterol exporter ABCA1 functions as an antiinflamma-

tory receptor. The Journal of Biological Chemistry 284(47): 3233632343. https://doi.org/10.1074/jbc.M109.047472 [PubMed] [PMC]

■ Torzewski M, Lackner KJ (2006) Initiation and progression of atherosclerosis - enzymatic or oxidative modification of low-density lipoprotein?. Clinical Chemistry and Laboratory Medicine 44(12): 1389-1394. https://doi.org/10.1515/CCLM.2006.259 [PubMed]

■ Torzewski M, Suriyaphol P, Paprotka K, Spath L, Ochsenhirt V, Schmitt A, Han SR, Husmann M, Gerl VB, Bhakdi S, Lackner KJ (2004) Enzymatic modification of low-density lipoprotein in the arterial wall: a new role for plasmin and matrix metallopro-teinases in atherogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology 24(11): 2130-2136. https://doi.org/10.1161/01. ATV.0000144016.85221.66 [PubMed]

■ Ungvari Z, Labinskyy N, Gupte S, Chander PN, Edwards JG, Csiszar A (2008) Dysregulation of mitochondrial biogenesis in vascular endothelial and smooth muscle cells of aged rats. American Journal of Physiology-Heart Circulatory Physiology 294(5): 2121-2128. https://doi.org/10.1152/ajpheart.00012.2008 [PubMed]

■ Utermann G, Hardewig A, Zimmer F (1984) Apolipoprotein E phe-notypes in patients with myocardial infarction. Human Genetics 65(3): 237-241. https://doi.org/10.1007/BF00286509 [PubMed]

■ Vickers KC, Landstreet SR, Levin MG, Shoucri BM, Toth CL, Taylor RC, Palmisano BT, Tabet F, Cui HL, Rye KA, Sethupathy P, Re-maley AT (2014) MicroRNA-223 coordinates cholesterol homeosta-sis. Proceedings of the National Academy of Sciences of the United States of America 111: 14518-14523. [PubMed] [PMC]

■ Wang XQ, Panousis CG, Alfaro ML, Evans GF, Zuckerman SH (2002) Interferon-gamma-mediated downregulation of cholesterol efflux and ABC1 expression is by the Stat1 pathway. Arteriosclerosis, Thrombosis, and Vascular Biology 22(5): e5-e9. https://doi. org/10.1161/01.atv.0000018287.03856.dd_[PubMed]

■ Wang Z, Zhang S, Jin H, Wang W, Huo J, Zhou L, Wang Y, Feng F, Zhang L (2011) Angiotensin-I-converting enzyme inhibitory pep-tides: Chemical feature based pharmacophore generation. European Journal of Mediccinal Chemistry 46(8): 3428-3433. https://doi. org/10.1016/j.ejmech.2011.05.007 [PubMed]

■ Wu D, Sharan C, Yang H, Goodwin JS, Zhou L, Grabowski GA, Du H, Guo Z (2007) Apolipoprotein E-deficient lipoproteins induce foam cell formation by downregulation of lysosomal hydrolases in macrophages. Journal of Lipid Research 48(12): 2571-2578. https:// doi.org/10.1194/jlr.M700217-JLR200 [PubMed]

■ Yancey PG, Jerome WG, Yu H, Griffin EE, Cox BE, Babaev VR, Fazio S, Linton MF (2007a) Severely altered cholesterol home-ostasis in macrophages lacking apoE and SR-BI. Journal of Lipid Research 48(5): 1140-1149. https://doi.org/10.1194/jlr.M600539-JLR200 [PubMed]

■ Yancey PG, Yu H, Linton MF, Fazio S (2007b) A pathway-dependent on apoE, ApoAI, and ABCA1 determines formation of buoyant high-density lipoprotein by macrophage foam cells. Arteriosclerosis, Thrombosis, and Vascular biology 27(5): 1123-1131. https://doi. org/10.1161/ATVBAHA.107.139592 [PubMed]

■ Yang C, Zhang C, Jia J, Wang L, Zhang W, Li J, Xu M, Rong R, Zhu T (2019) Cyclic helix B peptide ameliorates acute myocardial infarction in mice by inhibiting apoptosis and inflammatory responses. Cell Death Discovery 5(1): 1-10. https://doi.org/10.1038/s41420-019-0161-y [PubMed] [PMC]

■ Ye D, Lammers B, Zhao Y, Meurs I, Van Berkel TJ, Van Eck M (2011) ATP-binding cassette transporters A1 and G1, HDL metabo-

lism, cholesterol efflux, and inflammation: important targets for the treatment of atherosclerosis. Current Drug Targets 12(5): 647-660. https://doi.org/10.2174/138945011795378522 [PubMed]

■ Younis N, Sharma R, Soran H, Charlton-Menys V, Elseweidy M, Durrington PN (2008) Glycation as an atherogenic modification of LDL. Current Opinion in Lipidology 19(4): 378-384. https://doi. org/10.1097/M0L.0b013e328306a057_[PubMed]

■ Yu S, Duan RS, Chen Z, Quezada HC, Bao L, Nennesmo I, Zhu SW, Winblad B, Ljunggren HG, Zhu J (2004) Increased susceptibility to experimental autoimmune neuritis after upregulation of the autoreactive T cell response to peripheral myelin antigen in apolipopro-tein E-deficient mice. Journal of Neuropathology and Experimental Neurology 63(2): 120-128. https://doi.org/10.1093/jnen/63.2.120 [PubMed]

■ Zanotti I, Pedrelli M, Poti F, Stomeo G, Gomaraschi M, Calabresi L, Bernini F (2011) Macrophage, but not systemic, apolipoprotein E is necessary for macrophage reverse cholesterol transport in vivo. Arteriosclerosis, Thrombosis, and Vascular Biology 31(1): 74-80. https://doi.org/10.1161/ATVBAHA.110.213892 [PubMed]

■ Zárate A, Manuel-Apolinar L, Basurto L, De la Chesnaye E, Saldívar I (2016) Colesterol y aterosclerosis. Consideraciones históricas y tratamiento [Cholesterol and atherosclerosis. Historical consider-

ations and treatment]. Archivos de Cardiologia de Mexico 86(2): 163-169. https://doi.org/10.1016/j.acmx.2015.12.002 [PubMed]

■ Zhang C, Yang C, Zhu T (2017) From erythropoietin to its peptide derivatives: smaller but stronger. Current Protein and Peptide Science 18(12): 1191-1194. https://doi.org/10.2174/138920371766616 0909130006 [PubMed]

■ Zhang SH, Reddick RL, Piedrahita JA, Maeda N (1992) Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science 258(5081): 468-471. https://doi.org/10.1126/ science.1411543 [PubMed] [Full Text]

■ Zhou R, Yazdi AS, Menu P, Tschopp J (2011a) A role for mitochondria in NLRP3 inflammasome activation. Nature 469(7329): 221225. https://doi.org/10.1038/nature09663 [PubMed]

■ Zhou S, Chen HZ, Wan YZ, Zhang QJ, Wei YS, Huang S (2011b) Repression of P66Shc expression by SIRT1 contributes to the prevention of hyperglycemia-induced endothelial dysfunction. Circulation Research 109(6): 639-648. https://doi/org/10.1161/CIRCRE-SAHA.111.243592 [PubMed]

■ Zhuang G, Meng C, Guo X, Cheruku PS, Shi L, Xu H, Li H, Wang G, Evans AR, Safe S, Wu C, Zhou B (2012) A novel regulator of macrophage activation: miR-223 in obesity-associated adipose tissue inflammation. Circulation 125: 2892-2903. [PubMed]

Author contributions

All authors contributed equally to the writing of the submitted review.

■ Veronika S. Belyaeva, Postgraduate student, e-mail: nika.beliaeva@yandex.ru, ORCID ID 0000-0003-29410241. The author participated in the article writing.

■ Yulia V. Stepenko, Postgraduate student, e-mail: julia.v.stepenko@gmail.com, ORCID ID 0000-0002-7414-7326. The author participated in the article writing.

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

■ Igor I. Lyubimov, Candidate of Biological Sciences, General Manager, e-mail: lyubimov@gurus.bio, ORCID ID 0000-0002-0057-8537. The author consulted on the idea of the review, analysis of literature data and conclusions.

■ Alexandr L. Kulikov, Researcher, e-mail: alex-3031@yandex.ru, ORCID ID 0000-0001-6422-0377. The author was engaged in writing the article and formalization of the references.

■ Alesia A. Tietze, Senior Lecturer (Assistant Professor) in Medicinal Chemistry with focus on total synthesis of bioactive peptides, e-mail: a.tietze@tietze-lab.com, ORCID ID 0000-0002-9281-548X. The author consulted on the idea of the review and analyzed the literature data.

■ Indira S. Kochkarova, Junior researcher, Research Institute of Pharmacology of Living Systems, e-mail: koch-karova@bsu.edu.ru. The author analyzed the literature data.

■ Olga V. Martynova, Researcher, e-mail: m.olga91@mail.ru, ORCID ID 0000-0003-4104-4993. The author analyzed the literature data.

■ Olga N. Pokopeyko, Undergraduate student, Moscow, Russia, e-mail: pokopejko@yandex.ru. The author analyzed the literature data.

■ Liliya A. Krupen'kina, Assistant Professor, e-mail: crupenkina@lyandex.ru, ORCID ID 0000-0003-3299-5877. The author analyzed the literature data.

■ Andrey S. Nagikh, Postgraduate student, e-mail: nagikhandrew@gmail.com, ORCID ID 0000-0002-2505-4422. The author analyzed the literature data.

■ Vladimir M. Pokrovskiy, 5-year student, e-mail: vmpokrovsky@yandex.ru, ORCID ID 0000-0003-3138-2075. The author analyzed the literature data.

■ Evgeniy A. Patrakhanov, 5-year student, e-mail: pateval7@gmail.com, ORCID ID 0000-0002-8415-4562. The author analyzed the literature data.

■ Anastasia V. Belashova, 5-year student, e-mail: belashova_av@mail.ru, ORCID ID 0000-0001-9737-6378. The author participated in the article writing.

■ Petr R. Lebedev, 5-year student, e-mail: Artkelt98@yandex.ru, ORCID ID 0000-0001-9102-3360. The author analyzed the literature data.

■ Anastasia V. Gureeva, 3-year student, e-mail: nastasyi.207@gmail.com, ORCID ID 0000-0003-1719-7316. The author participated in the article writing.

i Надоели баннеры? Вы всегда можете отключить рекламу.