Научная статья на тему 'Апоптоз в сосудистой патологии: настоящее и будущее'

Апоптоз в сосудистой патологии: настоящее и будущее Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
222
39
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
АПОПТОЗ / ЭНДОТЕЛИАЛЬНЫЕ КЛЕТКИ / РЕСТЕНОЗ / АТЕРОСКЛЕРОТИЧЕСКАЯ БЛЯШКА / ГЛАДКОМЫШЕЧНЫЕ КЛЕТКИ / APOPTOSIS / ENDOTHELIAL CELLS / RESTENOSIS / ATHEROSCLEROTIC PLAQUE / SMOOTH MUSCLE CELLS

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Калинин Роман Евгеньевич, Сучков Игорь Александрович, Климентова Эмма Анатольевна, Егоров Андрей Александрович, Поваров Владислав Олегович

В настоящее время роль апоптоза признана при ряде сосудистых заболеваний. Он представляет собой запрограммированную клеточную гибель, которая находится под контролем генетических механизмов и необходима для нормального существования организма. Главной его задачей является уничтожение дефектных или мутантных клеток. Частицы погибших клеток поглощаются макрофагами без развития воспалительной реакции. Апоптоз принимает активное участие в эмбриогенезе, клеточном гемостазе, уничтожении опухолевых клеток. Данный процесс можно разделить на три фазы: сигнальная, эффекторная и деградационная. Его основным компонентом являются цитоплазматические протеазы каспазы. Каспазы находятся в цитоплазме в неактивном состоянии в виде прокаспаз. При активации они расщепляются на субъединицы. Белки семейства Bcl-2 являются активными участниками митохондриального пути апоптоза. Они влияют на проницаемость наружной мембраны митохондрий. Нарушения в механизмах апоптоза лежат в основе многих заболеваний, включая ишемические повреждения, аутоиммунные расстройства, злокачественные новообразования. Способность влиять на выживаемость или смерть клетки известна своим огромным терапевтическим потенциалом. В настоящее время активно развиваются исследования, направленные на изучение сигнальных путей, которые контролируют клеточный цикл и апоптоз. В статье обобщены механизмы, участвующие в гибели эндотелия сосудистой стенки и гладкомышечных клеток, а также рассмотрена потенциальная роль апоптоза при атеросклерозе.

i Надоели баннеры? Вы всегда можете отключить рекламу.

Похожие темы научных работ по фундаментальной медицине , автор научной работы — Калинин Роман Евгеньевич, Сучков Игорь Александрович, Климентова Эмма Анатольевна, Егоров Андрей Александрович, Поваров Владислав Олегович

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

Apoptosis in vascular pathology: present and future

Apoptosis is recognized as a programmed cell death controlled by genetic mechanisms and required for normal existence of an organism. Its main task is elimination of defective or mutant cells. The particles of dead cells are engulfed by macrophages with no development of inflammatory reaction. Apoptosis actively participates in embryogenesis, cellular homeostasis, elimination of tumor cells, and may be divided to three phases: signal, effector, and degradation. Its main components are cytoplasmic proteases caspases. Caspases exist in the cytoplasm in inactive condition -in the form pf procaspases. Being activated, they break down to subunits. Proteins of Bcl-2 family are active participants of the mitochondrial pathway of apoptosis. They influence permeability of the outer membrane of mitochondria. Disorders in the mechanisms of apoptosis underlie many diseases including ischemic lesions, autoimmune disorders, malignant neoplasms. The ability to influence survival or death of cell is known to possess enormous therapeutic potential. At present, active research is under way to study signal pathways that control cell cycle and apoptosis. The article discusses the mechanisms participating in death of vascular endothelium and smooth muscle cells, potential role of apoptosis in atherosclerosis is also described.

Текст научной работы на тему «Апоптоз в сосудистой патологии: настоящее и будущее»

REVIEW

АПОПТОЗ В СОСУДИСТОЙ ПАТОЛОГИИ: НАСТОЯЩЕЕ И БУДУЩЕЕ

©Р.Е. Калинин, И.А. Сучков, Э.А. Климентова, А.А. Егоров, В.О. Поваров

ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова Минздрава России, Рязань, Россия

В настоящее время роль апоптоза признана при ряде сосудистых заболеваний. Он представляет собой запрограммированную клеточную гибель, которая находится под контролем генетических механизмов и необходима для нормального существования организма. Главной его задачей является уничтожение дефектных или мутантных клеток. Частицы погибших клеток поглощаются макрофагами без развития воспалительной реакции. Апоптоз принимает активное участие в эмбриогенезе, клеточном гемостазе, уничтожении опухолевых клеток. Данный процесс можно разделить на три фазы: сигнальная, эффекторная и деградационная. Его основным компонентом являются цитоплазматические протеазы - каспазы. Каспазы находятся в цитоплазме в неактивном состоянии - в виде прокаспаз. При активации они расщепляются на субъединицы. Белки семейства Bcl-2 являются активными участниками митохон-дриального пути апоптоза. Они влияют на проницаемость наружной мембраны митохондрий. Нарушения в механизмах апоптоза лежат в основе многих заболеваний, включая ишемиче-ские повреждения, аутоиммунные расстройства, злокачественные новообразования. Способность влиять на выживаемость или смерть клетки известна своим огромным терапевтическим потенциалом. В настоящее время активно развиваются исследования, направленные на изучение сигнальных путей, которые контролируют клеточный цикл и апоптоз. В статье обобщены механизмы, участвующие в гибели эндотелия сосудистой стенки и гладкомышечных клеток, а также рассмотрена потенциальная роль апоптоза при атеросклерозе.

Ключевые слова: апоптоз; эндотелиальные клетки; рестеноз; атеросклеротическая бляшка; гладкомышечные клетки.

Apoptosis is recognized as a programmed cell death controlled by genetic mechanisms and required for normal existence of an organism. Its main task is elimination of defective or mutant cells. The particles of dead cells are engulfed by macrophages with no development of inflammatory reaction. Apoptosis actively participates in embryogenesis, cellular homeostasis, elimination of tumor cells, and may be divided to three phases: signal, effector, and degradation. Its main components are cytoplasmic proteases - caspases. Caspases exist in the cytoplasm in inactive condition -in the form pf procaspases. Being activated, they break down to subunits. Proteins of Bcl-2 family are active participants of the mitochondrial pathway of apoptosis. They influence permeability of the outer membrane of mitochondria. Disorders in the mechanisms of apoptosis underlie many diseases including ischemic lesions, autoimmune disorders, malignant neoplasms. The ability to influence survival or death of cell is known to possess enormous therapeutic potential. At present, active

APOPTOSIS IN VASCULAR PATHOLOGY: PRESENT AND FUTURE

R.E. Kalinin, I.A. Suchkov, E.A. Klimentova, A.A. Egorov, V.O. Povarov

Ryazan State Medical University, Ryazan, Russia

research is under way to study signal pathways that control cell cycle and apoptosis. The article discusses the mechanisms participating in death of vascular endothelium and smooth muscle cells, potential role of apoptosis in atherosclerosis is also described.

Keywords: apoptosis; endothelial cells; restenosis; atherosclerotic plaque; smooth muscle

cells.

The scientists have long observed the death of cells of the vascular wall in the absence of evident necrosis [1]. In 1972 J.F. Kerr et al. described a new form of cell death different from necrosis that they called 'apoptosis' (translated from ancient Greek as 'leaf fall') [2]. This is a programmed cell death controlled by genetic mechanisms and necessary for normal existence of an organism. Its main role is maintenance of constant cell composition and elimination of defective cells [3]. Disorders in the mechanisms of cell death form the basis for many diseases including ischemic lesions, autoimmune disorders, malignant neoplasms.

Molecular Mechanisms of Apoptosis

Apoptosis may be activated through a system of internal signals. Cells trigger the internal program of self-destruction in response to alteration of hemodynamic parameters or to loss of contact with neighboring cells. On the other hand, apoptosis may be initiated through the external stimuli, such as cytokines, hormones, oxidized lipids, ionizing or viral agents in which case the process takes a more acute and massive character. Some pathways of cell death are associated with multiple cells (for example, those associated with radiation lesion of DNA), while others occur only in particular cells ensuring their effective removal. The proportion between proapoptotic and antiapoptotic molecules determines the destiny of a cell at a particular moment of time [4].

There are several main functional stages in a programmed cascade of cell death.

In the phase of initiation, or signaling, cells receive inducing signals via attachment of certain molecules ((such as tumor necrosis factor a (TNF-a), Fas-ligand)) to the death receptors (TNFR1, Fas) on the surface of cell with subsequent assembly of death domain-

associated proteins (for example, FADD) with resultant activation of caspase-8 [5].

In the control, or effector phase, activation of caspases takes place with loss of mitochondrial membrane potential. Caspases are a family of cysteine proteases that participate in transduction and realization of the apoptotic program. They exist as inactive pro-enzymes activated by proteolytic cleavage. In central transitions of the apoptosis pathway, caspases 3, 8 and 9 are located.

Effector phase is controlled by Bcl-2 family of proteins which inhibit liberation of cytochrome C or of the apoptosis-inducing factor from mitochondria. Bcl-2 protein family includes both inhibitors (Bcl-2, Bcl-xL and others), and inductors (Bcl-xS, Bax, Bid, Bad, Bak and others) of death [6]. The proportion between antiapoptotic and proapoptotic proteins determines whether the cell will undergo death or not. After receiving a respective signal, Bax or Bak undergo conformational changes and move to the mitochondrial membrane where they induce release of cyto-chrome C to the cytosol [7].

Phase of degradation of genomic DNA leads to irreversible loss of viability of the cell.

In the phase of recognition the dead cell are removed from tissue as a result of phagocytosis by the neighboring cells using different mechanisms (for example, through phosphatidyl serine, PS, or receptor of vitronectin) [8].

Apoptosis of Endothelial Cells

Endothelium of the vessel wall participates in different physiological processes; therefore its death may present the initial stage of pathological conditions. Cell-to-cell contacts are necessary for maintenance and preservation of the endothelial cells, and their loss leads to activation of death program.

Extracellular matrix may generate signals that suppress p53-controlled pathway of apoptosis. For example, interaction with proteins of the extracellular matrix through avP3 integrin inhibits the activity of p53, decreases the level of Bax and activates NF-kB pathway, that promotes survival of cells. Another mechanism consists in activation of the antiapoptotic pathway through stimulation of the activity of proteins of Bcl-2 family. Growth factors (VEGF) inhibit death of cells in response to different stimuli, such as TNF-a, and destruction of the extracellular matrix occurs. This is due to stimulation of the signal pathway of protein kinase C or PI3K-Akt, or due to activation of Bcl-2 antiapoptotic proteins [9].

Fibroblast growth factor (FGF-2) is one of the factors associated with protection of endothelial cells. The study of А. Karsan et al. (1997) showed that FGF-2 specifically induces Bcl-2, but no other proteins of Bcl family. However, other non-Bcl-2 dependent mechanisms, such as phosphorylation of tyro-sine, may also inhibit death of endothelium with the help of FGF-2 [10].

Cells of vascular endothelium are permanently exposed to certain hemodynamic forces, whichhave a great effect on their cellular structure and function [11]. NO metabolites released in response to shear stress inhibit activation of caspase-3 and prevent cell death [12,13]. Both suppression of apoptosis and activation of caspase-3 by physiological levels of shear stress may be prevented by pharmacological inhibition of NO synthase [14].

Incubation of endothelial cells with TNF-a noticeably enhances apoptosis through activation of caspase-3. This process can be completely reversed by caspase inhibitors [15]. On the other hand, TNF-a is capable of inducing proteins of Bcl-2 family, and also of activating NF-kB pathway. So, TNF-a can induce both proapoptotic and antiapoptotic pathways [16]. Bcl-2 and Bcl-XL proteins can suppress activation of endothelial cells through specific inhibition of NF-kB pathway, whichshows that these proteins have cytoprotective effects that counteract inflammatory stimuli. Interestingly, interleukin-10

REVIEW

(IL-10) possesses antiapoptotic effect, therefore the balance between survival and death of cells may depend on the balance between pro-and antiinflammatory cytokines.

Apoptosis of Smooth Muscle Cells

After implementation of numerous research works, growth of smooth muscle cells (SMCs) is considered to be the result of opposite effects of cell proliferation and apopto-sis. Many growth factors associated with SMCs act as mitogens partially preventing apoptosis and maintaining secretion of Bcl-2 proteins. For example, IGF-I stimulates enhanced expression of inactive phosphorylated form of Bad through P13-kinase dependent pathway. Growth factors and their respective receptors are synthesized in many vascular diseases and may prevent apoptosis of SMCs within the arterial wall [17].

Mechanical factors. M.J. Pollman, et al. (1999) showed that percutaneous transluminal balloon angioplasty (PTA) induces a rapid death of SMCs within the normal arterial wall [18]. Apoptosis of SMCs is associated with activation of stress-activated protein kinase, and introduction of antioxidants causes its inhibition. This signal pathway cannot be activated in neointimal SMCs due to activation of Bcl-xl in them [19]. Oxidized low density lipoproteins (LDLP) promote apoptosis of SMCs partially by suppression of Bcl-2 and activation of caspase-3.

Vasoactive Substances. Local production of high levels of NO in the arterial wall inhibits growth of cells and formation of neointima after PTA [19]. In its turn, angio-tensin II (AT II) in the vessel wall facilitates growth of SMCs and thickening of the wall [20]. NO-induced apoptosis is prevented by inhibition of cGMP-dependent protein kinase Ia, and also by addition of AT II. Proapoptotic effect of NO manifests itself at the supraphysiological (pathological or inflammatory) level, and its antiapoptotic effect is observed with physiological (endothe-lial) concentrations [21]. Similarly, AT II prevents death of SMCs through stimulation of AT1 receptor, but promotes apoptosis through stimulation of AT2 receptor [22].

REVIEW

Oncogenes and suppressor genes of tumors. C-myc oncogene is expressed in SMCs of atherosclerotic plaques, and these cells demonstrate a lower growth rate as compare to SMCs obtained from the wall of normal arteries. T. Jacob, et al. (2012) proved in their work that SMCs of ASPs are hypersensitive to p53-mediated apoptosis. Activation of p53 temporarily enhances surface expression of Fas and sensitizes the cell to death [23].

Apoptosis in Atherosclerosis

Development of atherosclerotic plaques is associated both with death and proliferation of cells. At present, evidence is obtained of existence of apoptosis in all cells of a plaque: SMCs, macrophages, lymphocytes and endothelial cells. Apoptosis is most common in macrophages, which suggests participation of these cells in its induction. Programmed cell death is almost not detected in normal arteries, is hardly detected in fat streaks and is more common in diffused atherosclerotic lesions [24]. SMCs die in atherosclerotic plaques despite the presence of growth factors. This is associated with expression of c-myc onco-gene in the absence of Bcl-2 proteins and with enhanced sensitivity to p53 [25]. Recent data showed enhanced activity of proapoptotic Bax protein in human fat and in atherosclerotic plaques. Bax was not detected in normal arteries with predominating expression of Bcl-xl protein. The balance between pro- and antiapoptotic proteins in atherosclerosis favors the former and implies programming of SMCs to death in the presence of additional proapoptotic stimuli [26].

Apoptosis plays a double role in development of atherosclerosis. Death of SMCs may weaken the fibrous coating of the plaque through reduction of synthesis of the extracellular matrix, which leads to its rupture. On the other hand, the death of macrophages weakens the inflammatory response, reduces synthesis of metalloproteinases with subsequent destruction of the extracellular matrix, which ultimately promotes stabilization of the plaque. The level of cell death in the plaque is 2-10%, and the level of proliferation is less than 1%. Apoptosis is associated with natural

progression of atherosclerotic lesions through development of a lipid 'necrotic' nucleus. Death of macrophages is often identified along the edges of the lipid nucleus, which suggests that it may actively facilitate its formation. Elimination of apoptotic cells from an ASP may be ineffective since they compete with LDLP for being eliminated by macrophages. Non-eliminated cells undergo secondary necrosis, which evokes inflammatory response.

One of the main roles of apoptosis in atherosclerosis is associated with its procoagulant potential. Exposure of the cell surface to phosphatidyl serine significantly enhances the activity of tissue factor, which determines thrombogenicity of atherosclerotic plaques and provokes acute ischemic events [27].

Therapeutic Modulation of Apoptosis in Atherosclerosis

M.J. Pollman, et al. (1998) reported regression of a plaque in rabbits after inhibition of Bcl-xl expression through anti-sense oligonucleotide with subsequent induction of the apoptosis of neointima [28].

NO is a potent antiapoptotic molecule for endothelial cells. B Y. Wang, et al. (1999) also watched regression of atheromatous lesions in rabbits after introduction of L-arginine for induction of apoptosis of macrophages through NO synthesis [29]. On the other hand, it may be dangerous to induce mass death of cells of atherosclerotic lesions without anticoagulant therapy, taking into account their prothrom-bogenic potential. Dying endothelial cells are mostly localized in the poststenotic area with low shear stress. Consequently, delivery of NO with addition of L-arginine or delivery of endothelial growth factors may restrict death, thrombosis of cells and change the progress of atherosclerosis [30].

Antagonists of AT II receptors and blockers of calcium channels are strong inductors of apoptosis. Antioxidants such as trimetazidine, probucol, C and E vitamins inhibit apoptosis of SMCs after surgical intervention [31,32]. Application of this group of drugs is not sufficiently present in the literature, and the obtained results are contradictory.

s2

Finally, initiation of cell death in atherosclerotic plaques strongly depends on inflammatory balance. Introduction of IL-10 may be a reasonable strategy to reduce the inflammatory reaction, apoptotic cell death and, consequently, stabilization of the plaque. Apoptosis and Restenosis In different atherosclerotic and restenotic lesions 2-50% of cells undergo apoptosis. The models of formation of neointima after PTA in different animals have shown that in the process of recovery, apop-tosis occurs at different moments of time and at different levels.

H. Perlman, et al. (1997) showed that death of SMCs is observed as early as 30 minutes after PTBA and is linked with reduction of Bcl-x protein. After that the second wave of apoptosis was observed associated with enhanced proliferation of SMCs [33]. Death of cells in later stages, in the absence of proliferation, participates in regulation of the thickening of intima.

J.M. Isner, et al. (1995) showed a higher rate of apoptosis in restenotic lesions as compared to the primary atherosclerosis of the coronary arteries [34]. However, this conclusion was argued by G. Bauriedel, et al. (1998), who observed reduction of the level of apoptosis in

REVIEW

the zone of restenosis. A high cell density is a key finding in late restenosis in the region of the surgery and agrees with the paradigm of the lower parameters of apoptosis leading to hyperplasia. This corresponds with detection of decreased activity of p53 in the restenotic material of coronary arteries [35].

R. Shibata, et al. (2003) noted that PTBA led to insignificant increase in Bax protein and to significant increase in the level of Bcl-xl in 14 days after surgery. The use of Rho-kinase inhibitor in this study induced neointimal apoptosis of SMCs due to activation of Bax, which led to reduction of neointima formation [36].

P. Krishnan, et al. (2019) in their work studied restenotic lesions after the use of a drug-coated balloon during PTA. Lesions in this case were characterized by a significantly reduced number of cells in neointima, SMCs, fibroblast density, by increased activity of caspase-3 and by detection of deposits of type III collagen, in comparison with lesions in case of using the balloons without coating [37].

Conclusion

Further study of death and proliferation of cells is required to develop interventions that could prevent initiation of different diseases.

Литература

1. Егорова И.Э., Бахтаирова В.И., Суслова А.И. Молекулярные механизмы апоптоза, вовлеченные в развитие различных патологических процессов. В сб.: Инновационные технологии в фармации. Иркутск: ИГМУ; 2019. Вып. 6. С. 107-114.

2. Kerr J.F., Wyllie A.H., Currie A.R. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics // British Journal of Cancer. 1972. Vol. 26, №4. Р. 239-257. doi: 10.1038/bjc.1972.33

3. Майборода A.A. Апоптоз - гены и белки // Сибирский медицинский журнал (Иркутск). 2013. Т. 118, №3. С. 130-135.

4. Варга О.Ю., Рябков В.А. Апоптоз: понятие, механизмы реализации, значение // Экология человека. 2006. №7. С. 28-32.

5. Яровая Г.А., Нешкова Е.А., Мартынова Е.А., и др. Роль протеолитических ферментов в контроле различных стадий апоптоза // Лабораторная медицина. 2011. №11. С. 39-52.

6. Zhu Z.R., He Q., Wu W.B., et al. MiR-140-3p is Involved in In-Stent Restenosis by Targeting C-Myb and BCL-2 in Peripheral Artery Disease // Journal of Atherosclerosis and Thrombosis. 2018. Vol. 25, №11. P. 1168-1181. doi:10.5551/jat.44024

7. Цыган В.Н. Роль апоптоза в регуляции иммунного ответа // Обзоры по клинической фармакологии и лекарственной терапии. 2004. Т. 3, №2. С. 62-77.

8. Geng Y.J. Molecular signal transduction in vascular cell apoptosis // Cell Research. 2001. Vol. 11, №4. P. 253-264. doi: 10.1038/sj .cr.7290094

9. Mallat Z., Tedgui A. Apoptosis in the vasculature: mechanisms and functional importance // British Journal of Pharmacology. 2000. Vol. 130, №5. P. 947-962. doi: 10.1038/sj .bjp.0703407

10. Karsan A., Yee E., Poirier G.G., et al. Fibroblast growth factor-2 inhibits endothelial cell apoptosis by Bcl-2 dependent and independent mechanisms // The American Journal of Pathology. 1997. Vol. 151, №6. P. 1775-1784.

REVIEW

11. Fitzgerald T.N., Shepherd B.R., Asada H., et al. Laminar shear stress stimulates vascular smooth muscle cell apoptosis via the Akt pathway // Journal of Cellular Physiology. 2008. Vol. 216, №2. P. 389-395. doi:10.1002/jcp.21404

12. Jagadeeshaa D.K., Miller F.J. jr., Bhalla R.K. Inhibition of Apoptotic Signaling and Neointimal Hyperplasia by Tempol and Nitric Oxide Synthase following Vascular Injury // Journal of Vascular Research. 2009. Vol. 46, №2. P. 109-118. doi:10. 1159/000151444

13. Калинин Р.Е., Сучков И.А., Мжаванадзе Н.Д., и др. Дисфункция эндотелия у пациентов с имплантируемыми сердечно-сосудистыми электронными устройствами (обзор литературы) // Наука молодых (Eruditio Juvenium). 2016. №3. С. 84-92.

14. Duran X., Vilahur G., Badimon L. Exogenous in vivo NO- donor treatment preserves p53 levels and protects vascular cells from apoptosis // Atherosclerosis. 2009. Vol. 205, №1. P. 101-106. doi:10.1016/ j.atherosclerosis.2008.11.016

15. Beohar N., Flaherty J.D., Davidson C.J., et al. Anti-restenotic effects of a locally delivered caspases inhibitor in a balloon injury model // Circulation. 2004. Vol. 109, №1. P. 108-113. doi:10.1161/01. CIR.0000105724.30980.CD

16. Жураковский И.П., Битхаева М.В., Архипов С.А., и др. Изменения экспрессии белков семейства bcl-2 в печени крыс и уровень цитокинов в сыворотке крови при персистенции бактериальной инфекции // Кубанский научный медицинский вестник. 2012. №2(131). С. 84-87.

17. Kutuk O., Basaga Н. Bcl-2 protein family: Implications in vascular apoptosis and atherosclerosis // Apoptosis. 2006. Vol. 11, №10. P. 1661-1675. doi:10.1007/s10495-006-9402-7

18. Pollman M.J., Hall J.L., Gibbons G.H. Determinants of vascular smooth muscle cell apoptosis after balloon angioplasty injury: influence of redox state and cell phenotype // Circulation Research. 1999. Vol. 84, №1. P. 113-121. doi:10.1161/01.res.84.1.113

19. Пожилова Е.В., Новиков В.Е. Синтаза оксида азота и эндогенный оксид азота в физиологии и патологии клетки // Вестник Смоленской государственной медицинской академии. 2015. Т. 14, №4. С. 35-41.

20. Du J., Leng J., Zhang L., et al. Angiotensin II-Induced Apoptosis of Human Umbilical Vein Endothelial Cells was Inhibited by Blueberry Antho-cyanin Through Bax- and Caspase 3-Dependent Pathways // Medical Science Monitor. 2016. Vol. 22. P. 3223-3228. doi:10.12659/msm.896916

21. Новицкий В.В., Рязанцева Н.В., Старикова Е.Г., и др. Регуляция апоптоза клеток с использованием газовых трансмиттеров (оксид азота, монооксид углерода и сульфид водорода) // Вестник науки Сибири. 2011. №1(1). С. 635-640.

22. Yang H.Y. Bian Y.F., Zhang H.P., et al. Angioten-sin-(1-7) treatment ameliorates angiotensin II-indu-

ced apoptosis of human umbilical vein endothelial cells // Clinical and Experimental Pharmacology & Physiology. 2012. Vol. 39, №12. P. 1004-1010. doi:10.1111/1440-1681.12016

23. Jacob T., Hingorani A., Ascher E. p53 gene therapy modulates signal transduction in the apoptotic and cell cycle pathways downregulating neointimal hyperplasia // Vascular and Endovascular Surgery.

2012. Vol. 46, №1. P. 45-53. doi:10.1177/153857 4411422277

24. Владимирская Т.Э., Швед И.А., Демидчик Ю.Е. Соотношение эксперессии белков BCL-2 b Bax в стенке коронарных артерий, пораженных атеросклерозом // Известия национальной академии наук Беларусии. Серия медицинских наук. 2015. №4. С. 51-55.

25. Parkes J.L., Cardell R.R., Hubbard F.C. Jr., et al. Cultured human atherosclerotic plaque smooth muscle cells retain transforming potential and display enhanced expression of the myc protoonco-gene // The American Journal of Pathology. 1991. Vol. 138, №3. P. 765-775.

26. Tian X., Shi Y., Liu N., et al Upregulation of DAPK contributes to homocysteine-induced endothelial apoptosis via the modulation of Bcl2/Bax and activation of caspase 3 // Molecular Medicine Reports. 2016. Vol. 14, №5. P. 4173-4179. doi:10. 3892/mmr.2016.5733

27. Хаспекова С.Г., Антонова О.А., Шустова О.Н., и др. Активность тканевого фактора в микрочастицах, продуцируемых in vitro эндотелиаль-ными клетками, моноцитами, гранулоцитами и тромбоцитами // Биохмимия. 2016. Т. 81, №2. С. 206-214.

28. Pollman M.J., Hall J.L., Mann M.J., et al. Inhibition of neointimal cell bcl-x expression induces apoptosis and regression of vascular disease // Nature Medicine. 1998. Vol. 4. P. 222-227. doi:10.1038/ nm0298-222

29. Wang B.Y., Ho H.K., Lin P.S., et al. Regression of atherosclerosis: role of nitric oxide and apoptosis // Circulation. 1999. Vol. 99, №9. P. 1236-1241. doi:10.1161/01.cir.99.9.1236

30. Калинин Р.Е., Сучков И.А., Пшенников А.С., и др. Эффективность L-аргинина в лечении атеросклероза артерий нижних конечностей и профилактике рестеноза зоны реконструкции // Вестник Ивановской Медицинской Академии.

2013. Т. 18, №2. С. 18-21.

31. de Araújo Júnior R.F., Leitao Oliveira A.L., de Melo Silveira R.F., et al. Telmisartan induces apoptosis and regulates Bcl-2 in human renal cancer cells // Experimental Biology and Medicine (May-wood N.J.). 2015. Vol. 240, №1. P. 34-44. doi:10. 1177/1535370214546267

32. Пшенников А.С., Деев Р.В. Морфологическая иллюстрация изменений артериального эндотелия на фоне ишемического и реперфузионного повреждений // Российский медико-биологичес-

кий вестник имени академика И.П. Павлова. 2018. Т. 26, №2. С. 184-194. doi:10.23888/PAV-LOVJ2018262184-194

33. Perlman H., Maillard L., Krasinski K., et al. Evidence for the rapid onset of apoptosis in medial smooth muscle cells after balloon injury // Circulation. 1997. Vol. 95, №4. P. 981-987. doi:10.1161/ 01.cir.95.4.981

34. Isner J.M., Kearney M., Bortman S., et al. Apoptosis in human atherosclerosis and restenosis // Circulation. 1995. Vol. 91, №11. P. 2703-2711. doi:10. 1161/01.cir.91.11.2703

35. Bauriedel G., Schluckebier S., Hutter R., et al. Apoptosis in restenosis versus stable-angina atherosclerosis. Implications for the pathogenesis of restenosis // Arteriosclerosis, Thrombosis, and Vascular Biology. 1998. Vol. 18, №7. P. 1132-1139. doi:10.1161/01.atv.18.7.1132

36. Shibata R., Kai H., Seki Y., et al. Rho-kinase Inhibition Reduces Neointima Formation After Vascular Injury by Enhancing Bax Expression and Apoptosis // Journal of Cardiovascular Pharmacology. 2003. Vol. 42, suppl 1. Р. S43-S47. doi:10.1097/ 00005344-200312001-00011

37. Krishnan P., Purushothaman K.R., Purushothaman M., et al. Histological features of restenosis associated with paclitaxel drug-coated balloon: implications for therapy // Cardiovascular Pathology. 2019. Vol. 43. P. 107139. doi:10.1016/j.carpath.2019. 06.003

References

1. Egorova IE, Bakhtairova VI, Suslova AI. Molecular mechanisms of apoptosis involved in the development of different pathological processes. Innovat-sionnye Tekhnologii v Farmatsii. Irkutsk: ISMU; 2019;6:107-14. (In Russ).

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

2. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. British Journal of Cancer. 1972;26(4):239-57. doi: 10.1038/bjc.1972.33

3. Majboroda AA. Apoptosis - genes and proteins. SibirskijMedicinskij Zurnal (Irkutsk). 2013;118(3): 130-5. (In Russ).

4. Varga OYu, Ryabkov VA. Apoptosis: concept, mechanisms of realization, significance. Ekologiya Cheloveka. 2006;(7):28-32. (In Russ).

5. Yarovaya GA, Neshkova EA, Martynova EA, et al. Rol' proteoliticheskikh fermentov v kontrole raz-lichnykh stadiy apoptoza. Laboratornaya Medi-tsina. 2011;(11):39-51. (In Russ).

6. Zhu ZR, He Q, Wu WB, et al. MiR-140-3p is Involved in In-Stent Restenosis by Targeting C-Myb and BCL-2 in Peripheral Artery Disease. Journal of Atherosclerosis and Thrombosis. 2018;25(11): 116881. doi:10.5551/jat.44024

7. Tsygan VN. Rol' apoptoza v regulyatsii immunnogo otveta. Obzory po Klinicheskoy Farmakologii i Lekarstvennoy Terapii. 2004;3(2):62-77. (In Russ).

REVIEW

8. Geng YJ. Molecular signal transduction in vascular cell apoptosis. Cell Research. 2001;11(4):253-64. doi: 10.1038/sj .cr.7290094

9. Mallat Z, Tedgui A. Apoptosis in the vasculature: mechanisms and functional importance. British Journal of Pharmacology. 2000;130(5):947-62. doi:10.1038/sj .bjp.0703407

10. Karsan A, Yee E, Poirier GG, et al. Fibroblast growth factor-2 inhibits endothelial cell apoptosis by Bcl-2 dependent and independent mechanisms. The American Journal of Pathology. 1997;151(6): 1775-84.

11. Fitzgerald TN, Shepherd BR, Asada H. Laminar shear stress stimulates vascular smooth muscle cell apoptosis via the Akt pathway. Journal of Cellular Physiology. 2008;216(2):389-95. doi:10.1002/jcp.21404

12. Jagadeeshaa DK, Miller FJ jr, Bhalla RK. Inhibition of Apoptotic Signaling and Neointimal Hyperplasia by Tempol and Nitric Oxide Synthase following Vascular Injury. Journal of Vascular Research. 2009;46(2):109-18. doi:10.1159/000151444

13. Kalinin RE, Suchkov IA, Mzhavanadze ND, et al. Endothelial Dysfunction In Patients With Cardiac Implantable Electronic Devices (Literature Review). Nauka Molodykh (Eruditio Juvenium). 2016; (3):84-92. (In Russ).

14. Duran X, Vilahur G, Badimon L. Exogenous in vivo NO- donor treatment preserves p53 levels and protects vascular cells from apoptosis. Atherosclerosis. 2009;205(1):101-6. doi:10.1016/j.atheroscle-rosis.2008.11.016

15. Beohar N, Flaherty JD, Davidson CJ, et al. Anti-restenotic effects of a locally delivered caspases inhibitor in a balloon injury model. Circulation. 2004;109(1):108-13. doi:10.1161/01.CIR.00001057 24.30980.CD

16. Zhurakovsky IP, Bitkhaeva MV, Arkhipov S A, et al. Changes in Bcl-2 protein family expression in the liver and serum cytokines level at persisting bacterial infection. Kubanskii Nauchnyi Meditsinskii Vestnik. 2012;(2(131)):84-7. (In Russ).

17. Kutuk O, Basaga Н. Bcl-2 protein family: Implications in vascular apoptosis and atherosclerosis. Apoptosis. 2006;11(10):1661-75. doi:10.1007/s104 95-006-9402-7

18. Pollman MJ, Hall JL, Gibbons G.H. Determinants of vascular smooth muscle cell apoptosis after balloon angioplasty injury: influence of redox state and cell phenotype. Circulation Research. 1999; 84(1):113-21. doi:10.1161/01.res.84.1.113

19. Pozhilova EV, Novikov VE. Physiological and pathological value of cellular synthase of nitrogen oxide and endogenous nitrogen oxide. Vestnik Smo-lenkskoy Gosudarstvennoy Medicinskoy Akademii. 2015;14(4): 35-41. (In Russ).

20. Du J, Leng J, Zhang L, et al. Angiotensin II-Induced Apoptosis of Human Umbilical Vein Endothelial Cells was Inhibited by Blueberry Antho-

REVIEW

cyanin Through Bax- and Caspase 3-Dependent Pathways. Medical Science Monitor. 2016;22:3223-8. doi: 10.12659/msm.896916

21. Novitskiy VV, Ryazantseva NV, Starikova EG, et al. Regulyatsiya apoptoza kletok s ispol'zovaniyem gazovykh transmitterov (oksid azota, monooksid ugleroda i sul'fid vodoroda). Vestnik Nauki Sibiri. 2011;1(1):635-40. (In Russ).

22. Yang HY, Bian YF, Zhang HP, et al. Angiotensin-(1-7) treatment ameliorates angiotensin II-induced apoptosis of human umbilical vein endothelial cells. Clinical and Experimental Pharmacology & Physiology. 2012;39(12):1004-10. doi:10.1111/1440-1681. 12016

23. Jacob T, Hingorani A, Ascher E. p53 gene therapy modulates signal transduction in the apoptotic and cell cycle pathways downregulating neointimal hy-perplasia. Vascular and Endovascular Surgery. 2012;46(1):45-53. doi:10.1177/1538574411422277

24. Vladimirskaya TE, Shved IA, Demidchik YuE. Ratio of expression of the Bcl-2 and Bax proteins in the atherosclerotic coronary artery wall. Proceedings of the National Academy of Sciences of Belarus. Medical Series. 2015;(4):51-5. (In Russ).

25. Parkes JL, Cardell RR, Hubbard FC Jr, et al. Cultured human atherosclerotic plaque smooth muscle cells retain transforming potential and display enhanced expression of the myc protooncogene. The American Journal of Pathology. 1991;138(3):765-75.

26. Tian X, Shi Y, Liu N, et al. Upregulation of DAPK contributes to homocysteine-induced endothelial apoptosis via the modulation of Bcl2/Bax and activation of caspase 3. Molecular Medicine Reports. 2016;14(5):4173-79. doi:10.3892/mmr.2016.5733

27. Khaspekova SG, Antonova OA, Shustova ON, et al. Tissue factor activity in microparticles produced in vitro by endothelial cells, monocytes, granulocytes, and platelets. Biochemistry. 2016;81(2):206-14. (In Russ).

28. Pollman MJ, Hall JL, Mann MJ, et al. Inhibition of neointimal cell bcl-x expression induces apoptosis and regression of vascular disease. Nature Medi-

cine. i998;4:222-7. doi:i0.i038/nm0298-222

29. Wang BY, Ho HK, Lin PS, et al. Regression of atherosclerosis: role of nitric oxide and apoptosis. Circulation. 1999;99(9):1236-41. doi:i0.ii6i/0i. cir.99.9.1236

30. Kalinin RE, Suchkov IA Pshennikov AS, et al. L-arginine efficiency in treatment for lower limbs arteries atherosclerosis and in prevention of reconstruction zone restenosis. Vestnik Ivanovskoy MeditsinskoyAkademii. 2013;18(2):18-21. (In Russ).

31. de Araújo Júnior RF, Leitao Oliveira AL, de Melo Silveira RF, et al. Telmisartan induces apoptosis and regulates Bcl-2 in human renal cancer cells. Experimental Biology and Medicine (Maywood NJ). 2015;240(1):34-44. doi:10.1177/1535370214546267

32. Pshennikov AS, Deev RV. Morphological illustration of alterations in the arterial endothelium in ischemic and reperfusion injuries. I.P. Pavlov Russian Medical Biological Herald. 2018;26(2):184-94. doi:10.23888/ PAVLOVJ2018262184-194 (In Russ).

33. Perlman H, Maillard L, Krasinski K, et al. Evidence for the rapid onset of apoptosis in medial smooth muscle cells after balloon injury. Circulation. 1997;95(4):981-7. doi:10.1161/01.cir.95.4.981

34. Isner JM, Kearney M, Bortman S, et al. Apoptosis in human atherosclerosis and restenosis. Circulation. 1995;91(11):2703-11. doi:10.1161/01.cir.91.11.2703

35. Bauriedel G, Schluckebier S, Hutter R, et al. Apoptosis in restenosis versus stable-angina atherosclerosis. Implications for the pathogenesis of restenosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 1998;18(7):1132-9. doi:10.1161/01.atv.l8.7.1132

36. Shibata R., Kai H., Seki Y., et al. Rho-kinase Inhibition Reduces Neointima Formation After Vascular Injury by Enhancing Bax Expression and Apoptosis. Journal of Cardiovascular Pharmacology. 2003; 42(l):S43-7. doi:l0.l097/00005344-2003l200l-000ll

37. Krishnan P, Purushothaman KR, Purushothaman M, et al. Histological features of restenosis associated with paclitaxel drug-coated balloon: implications for therapy. Cardiovasc Pathol. 20l9;43: 107139. doi:l0.l0l6/j.carpath.20l9.06.003

Дополнительная информация [Additional Info]

Источник финансирования. Бюджет ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова Минздрава России. [Financing of stady. Budget of Ryazan State Medical University.]

Конфликт интересов. Авторы декларируют отсутствие явных и потенциальных конфликтов интересов, о которых необходимо сообщить, в связи с публикацией данной статьи. [^пИМ of interests. The authors declare no actual and potential conflict of interests which should be stated in connection with publication of the article.]

Участие авторов. Калинин Р.Е. - концепция литературного обзора, редактирование, Сучков И.А., Егоров А.А., Поваров В.О. - редактирование, Климентова Э.А., - сбор, перевод и анализ материала, написание текста. [Participation of authors. R.E. Kalinin - concept of the review, editing, I.A. Suchkov, A.A. Egorov, V.O. Povarov - editing, E.A. Klimentova - collection, translation and analysis of material, writing the text.]

REVIEW

Информация об авторах [Authors Info]

Калинин Роман Евгеньевич - д.м.н., проф., зав. кафедрой сердечно-сосудистой, рентгенэндоваскулярной, оперативной хирургии и топографической анатомии, ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова Минздрава России, Рязань, Россия. [Roman E. Kalinin - MD, PhD, Professor, Head of the Department of Cardiovascular, X-ray Endovascular, Operative Surgery and Topographic Anatomy, Ryazan State Medical University, Ryazan, Russia.] SPIN: 5009-2318, ORCID ID: 0000-0002-0817-9573, Researcher ID: M-1554-2016.

Сучков Игорь Александрович - д.м.н., проф., проф. кафедры сердечно-сосудистой, рентгенэндоваскулярной, оперативной хирургии и топографической анатомии, ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова Минздрава России, Рязань, Россия. [Igor'A. Suchkov - MD, PhD, Professor, Professor of the Department of Cardiovascular, X-ray Endovascular, Operative Surgery and Topographic Anatomy, Ryazan State Medical University, Ryazan, Russia.] SPIN: 6473- 8662, ORCID ID: 0000-0002-1292-5452, Researcher ID: M-1180-2016.

*Климентова Эмма Анатольевна - к.м.н., аспирант кафедры сердечно-сосудистой, рентгенэндоваскулярной, оперативной хирургии и топографической анатомии, ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова, Рязань, Россия. [Emma A. Klimentova - MD, PhD, PhD-Student of the Department of Cardiovascular, X-Ray Endovascular, Operative Surgery and Topographic Anatomy, Ryazan State Medical University, Ryazan, Russia.]

SPIN: 5629-9835, ORCID ID: 0000-0003-4855-9068, Researcher ID: P-1670-2017. E- mail: klimentowa.emma@yandex.ru

Егоров Андрей Александрович - к.м.н., докторант кафедры сердечно-сосудистой, рентгенэндоваскулярной, оперативной хирургии и топографической анатомии, ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова, Рязань, Россия. [Andrey A. Egorov - MD, PhD, PhD-Student of the Department of Cardiovascular, X-Ray Endovascular, Operative Surgery and Topographic Anatomy, Ryazan State Medical University, Ryazan, Russia.] SPIN: 2408-4176, ORCID ID: 0000-0003-0768-7602.

Поваров Владислав Олегович - аспирант кафедры сердечно-сосудистой, рентгенэндоваскулярной, оперативной хирургии и топографической анатомии, ФГБОУ ВО Рязанский государственный медицинский университет им. акад. И.П. Павлова, Рязань, Россия. [Vladislav O. Povarov - PhD-Student of the Department of Cardiovascular, X-Ray Endovascular, Operative Surgery and Topographic Anatomy, Ryazan State Medical University, Ryazan, Russia.]

SPIN: 2873-1391, ORCID ID: 0000-0001-8810-9518, Researcher ID: P-2383-2017.

Цитировать: Калинин Р.Е., Сучков И.А., Климентова Э.А., Егоров А.А., Поваров В.О. Апоптоз в сосудистой патологии: настоящее и будущее // Российский медико-биологический вестник имени академика И.П. Павлова. 2019. Т. 27, №4. С. 79-87. doi:10.23888/ PAVLOVJ202028179-87

To cite this article: Kalinin RE, Suchkov IA, Klimentova EA, Egorov AA, Povarov VO. Apoptosis in vascular pathology: present and future.

I.P. Pavlov Russian Medical Biological Herald. 2020;28(1):79-87. doi:10.23888/PAVL0VJ202028179-87

Поступила/Received: 11.11.2019 Принята в печать/Accepted: 31.03.2020

i Надоели баннеры? Вы всегда можете отключить рекламу.