Научная статья на тему 'The role of subpopulations of cd8+ t lymphocytes in the development of pregnancy'

The role of subpopulations of cd8+ t lymphocytes in the development of pregnancy Текст научной статьи по специальности «Фундаментальная медицина»

CC BY
157
21
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
T-ЛИМФОЦИТЫ / БЕРЕМЕННОСТЬ / ЦИТОТОКСИЧНОСТЬ / ДЕЦИДУАЛЬНАЯ ОБОЛОЧКА / T LYMPHOCYTES / PREGNANCY / CYTOTOXIC / DECIDUA

Аннотация научной статьи по фундаментальной медицине, автор научной работы — Stepanova O.I., Bazhenov D.O., Khokhlova E.V., Kogan I.Yu., Sokolov D.I.

At the present time, a broad spectrum of CD8+ T lymphocyte subsets is revealed, including naïve cells, memory cells and regulatory subpopulations. Along with cells with high cytolytic activity, some subsets with marked regulatory activity were found there. Each subpopulation is characterized by a set of produced mediators, surface and intracellular markers allowing to suggest their differential in vivo functional activity. The present review article proposes a classification of CD8+ Т cells which takes into account their morphological and functional features. According to conventional view, the CD8+ Т lymphocytes is a cell population exhibiting high cytotoxic ability which is of critical significance in pregnancy, under the conditions of semi-allogenic fetal cell invasion into the endometrium. The fraction of CD8+ T cells is rather high in decidual structures. The review discusses the known mechanisms of differentiation regulation, selective migration and activity of CD8+ T cells in decidual membrane and placenta in the course of pregnancy. Perforine and granzyme are the main cytotoxicity factors of CD8+ Т cells. IL-2, IL-5, IL-13, IFNγ, IL-17, TGF-β and IL-10 cytokines are considered regulatory mediators of CD8+ cells. To induce the effector properties of CD8+ T cells, an antigenic stimulation is required, which is provided by interactions between the CD8+ Т cells and activated CD4+ Т cells or dendritic cells, cytokine effects. Specific differentiation of the CD8+ T cells is determined by differences in microenvironvent. In the course of pregnancy, accumulation of CD8+ Т cells is observed in decidual membrane, but their phenotype and functional properties differ from CD8+ Т cells in peripheral blood. At present time, the mechanisms of selective CD8+ T cell migration to decidual membrane are studied. These events are suggested to be mediated by means of CXCR3 and CCR5 chemokine receptors, IL-6 and IL-15 cytokines. The features of CD8+ Т cell activities, and production of some cytokines, e.g., CSF2, IFNγ, IL-1β, IL-2, IL-6, IL-8,IL-10, IL-12 and TNFα in decidual membrane and is of critical significance for effective invasion of trophoblast cells. In turn, the trophoblast and placental cells promote development of regulatory CD8+ Т lymphocytes in decidual membrane, being able to induce CD8+ T cell apoptosis in decidual membrane. Hence, interaction between the maternal CD8+ T cells and trophoblast in the area of uterine-placental contact is an important link during development of immunological tolerance in the maternal/fetal system.

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.

Роль различных субпопуляций CD8+Т-лимфоцитов при беременности

В настоящее время выявлено существование широкого спектра субпопуляций CD8+Т-лимфоцитов, среди которых выделяют субпопуляции наивных клеток, клеток памяти, регуляторных. Кроме клеток с высоким уровнем цитотоксической активности, выявлены субпопуляции, обладающие выраженной регуляторной активностью. Каждая субпопуляция характеризуется совокупностью продуцируемых медиаторов, поверхностных и внутриклеточных маркеров, позволяющих предположить их различную функциональную активность в условиях in vivo. В настоящем обзоре описана классификация CD8+Т-лимфоцитов, учитывающая их морфофункциональные признаки. Традиционно считается, что CD8+Т-лимфоциты являются популяцией лимфоцитов, обладающей высокой цитотоксической активностью, что имеет чрезвычайное значение в условиях инвазии полуалогенных плодовых клеток в эндометрий при беременности. Доля CD8+Т-лимфоцитов в децидуальной оболочке довольно велика. В обзоре обсуждаются известные на сегодняшний день механизмы регуляции дифференцировки, избирательной миграции и функциональной активности CD8+Т-лимфоцитов в децидуальной оболочке и плаценте при беременности. Основными факторами цитотоксического действия CD8+Т-лимфоцитов являются перфорин и гранзим. К регуляторным медиаторам CD8+Т-лимфоцитов относят цитокины IL-2, IL-5, IL-13, IFNγ, IL-17, TGF-β и IL-10. Для развития эффекторных свойств CD8+Т-лимфоцитов необходима антигенная стимуляция, которую обеспечивает взаимодействие CD8+Т-лимфоцитов с активированными CD4+Т-лимфоцитами или дендритными клетками, воздействие цитокинов. Условия специфической дифференцировки CD8+Т-лимфоцитов формируются за счет различного характера микроокружения. В децидуальной оболочке при беременности наблюдается концентрация CD8+Т-лимфоцитов, но их фенотип и функциональнальная активность отличаются от CD8+Т-лимфоцитов периферической крови. В настоящее время продолжается изучение механизмов избирательной миграции CD8+Т-лимфоцитов с регуляторными свойствами в децидуальную оболочку. Полагают, что это обеспечивается при участии хемокиновых рецепторов CXCR3 и CCR5, цитокинов IL-6 и IL-15. Характер активности CD8+Т-лимфоцитов и продукция ими цитокинов CSF2, IFNγ, IL-1β, IL-2, IL-6, IL-8,IL-10, IL-12 и TNFα в децидуальной оболочке имеют решающее значение для успешной инвазии клеток трофобласта. В свою очередь, клетки трофобласта и плаценты способствуют формированию пула регуляторных CD8+Т-лимфоцитов в децидуальной оболочке, способны индуцировать апоптоз CD8+Т-лимфоцитов. Таким образом, взаимодействие CD8+Т-лимфоцитов матери и трофобласта в зоне маточно-плацентарного контакта является важным звеном в формировании иммунологической толлерантности в системе мать-плод.

Текст научной работы на тему «The role of subpopulations of cd8+ t lymphocytes in the development of pregnancy»

Medical Immunology (Russia)/ Медицинская иммунология ОбЗОРЫ Meditsinskaya Immunologiya

2018, Т. 20, № 5, стр. 621-638 D * 2018, Vol. 20, No 5, pp. 621-638

© 2018, СПб РО РААКИ KeVWWS © 2018, SPb RAACI

РОЛЬ РАЗЛИЧНЫХ СУБПОПУЛЯЦИЙ CD8+Т-ЛИМФОЦИTOВ ПРИ БЕРЕМЕННОСТИ

Степанова О.И.1, Баженов Д.О.1, Хохлова Е.В.1, Коган И.Ю.1, Соколов Д.И.1, 2, Сельков С.А.1

1ФГБНУ«Научно-исследовательский институт акушерства, гинекологии ирепродуктологии имени Д.О. Отта», Санкт-Петербург, Россия

2 ГБОУ ВПО «Первый Санкт-Петербургский государственный медицинский университет имени академика И.П. Павлова» Министерства здравоохранения РФ, Санкт-Петербург, Россия

Резюме. В настоящее время выявлено существование широкого спектра субпопуляций CDS+Т-лимфоцитов, среди которых выделяют субпопуляции наивных клеток, клеток памяти, регуляторных. Кроме клеток с высоким уровнем цитотоксической активности, выявлены субпопуляции, обладающие выраженной регуляторной активностью. Каждая субпопуляция характеризуется совокупностью продуцируемых медиаторов, поверхностных и внутриклеточных маркеров, позволяющих предположить их различную функциональную активность в условиях in vivo. В настоящем обзоре описана классификация CDS+Т-лимфоцитов, учитывающая их морфофункциональные признаки. Традиционно считается, что CDS+Т-лимфоциты являются популяцией лимфоцитов, обладающей высокой цитотоксической активностью, что имеет чрезвычайное значение в условиях инвазии полуалогенных плодовых клеток в эндометрий при беременности. Доля CDS+Т-лимфоцитов в децидуальной оболочке довольно велика. В обзоре обсуждаются известные на сегодняшний день механизмы регуляции дифференцировки, избирательной миграции и функциональной активности CDS+Т-лимфоцитов в децидуальной оболочке и плаценте при беременности. Основными факторами цитотоксического действия CDS+Т-лимфоцитов являются перфорин и гранзим. К регуляторным медиаторам CDS+Т-лимфоцитов относят цитокины IL-2, IL-5, IL-13, IFNy, IL-17, TGF-p и IL-10. Для развития эффек-торных свойств CDS+Т-лимфоцитов необходима антигенная стимуляция, которую обеспечивает взаимодействие CDS+Т-лимфоцитов с активированными CD4+Т-лимфоцитами или дендритными клетками, воздействие цитокинов. Условия специфической дифференцировки CDS+Т-лимфоцитов формируются за счет различного характера микроокружения. В децидуальной оболочке при беременности наблюдается концентрация CDS+Т-лимфоцитов, но их фенотип и функциональнальная активность отличаются от CDS+Т-лимфоцитов периферической крови. В настоящее время продолжается изучение механизмов избирательной миграции CDS+Т-лимфоцитов с регуляторными свойствами в децидуальную оболочку. Полагают, что это обеспечивается при участии хемокиновых рецепторов CXCR3 и CCR5, цитокинов IL-6 и IL-15. Характер активности CDS+Т-лимфоцитов и продукция ими цитокинов CSF2, IFNy, IL-1 в, IL-2, IL-6, IL-8,IL-10, IL-12 и TNFa в децидуальной оболочке имеют решающее значение для успешной инвазии клеток трофобласта. В свою очередь, клетки трофобла-

Адрес для переписки:

Соколов Дмитрий Игоревич

ФГБНУ «Научно-исследовательский институт

акушерства, гинекологии и репродуктологии

имени Д.О. Отта»

199034, Россия, Санкт-Петербург,

Менделеевская линия, 3.

Тел.: 8(812) 323-75-45, 328-98-50.

Факс: 8(812) 323-75-45.

E-mail: falcojuggen@yandex.ru

Образец цитирования:

О.И. Степанова, Д.О. Баженов, Е.В. Хохлова, Коган И.Ю., Д.И. Соколов, С.А. Сельков «Рольразличньк субпопуляций CD^T-лимфоцитов при беременности» // Медицинская иммунология, 2018. Т. 20, № 5. С. 621-638. doi: 10.15789/1563-0625-2018-5-621-638 © Степанова О.И. и соавт., 2018

Address for correspondence:

Sokolov Dmitry I.

D. Ott Research Institute of Obstetrics, Ginecology and Reproductology

199034, Russian Federation, St. Petersburg,

Mendeleevskaya line, 3.

Phone: 7(812) 323-75-45, 328-98-50.

Fax: 7(812) 323-75-45.

E-mail: falcojuggen@yandex.ru

For citation:

O.I. Stepanova, D.O. Bazhenov, E.V. Khokhlova, KoganI.Yu., D.I. Sokolov, S.A. Selkov "The role of subpopulations of CD8+ T lymphocytes in the development of pregnancy", Medical Immunology (Russia)/Meditsinskaya Immunologiya, 2018, Vol. 20, no. 5, pp. 621-638.

doi: 10.15789/1563-0625-2018-5-621-638 DOI: 10.15789/1563-0625-2018-5-621-638

Степанова О.И. и др. Медицинская Иммунология

Stepanova O.I. et al. Medical Immunology (Russia)/Meditsinskaya Immunologiya

ста и плаценты способствуют формированию пула регуляторных CDS+Т-лимфоцитов в децидуаль-ной оболочке, способны индуцировать апоптоз CDS+Т-лимфоцитов. Таким образом, взаимодействие CDS+Т-лимфоцитов матери и трофобласта в зоне маточно-плацентарного контакта является важным звеном в формировании иммунологической толлерантности в системе мать-плод.

Ключевые слова: T-лимфоциты, беременность, цитотоксичность, децидуальная оболочка

THE ROLE OF SUBPOPULATIONS OF CD8+ T LYMPHOCYTES IN THE DEVELOPMENT OF PREGNANCY

Stepanova O.I.a, Bazhenov D.O.a, Khokhlova E.V.a, Kogan I.Yu.a, Sokolov D.I.0' b, Selkov S.A.a

a D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation b First St. Petersburg State I. Pavlov Medical University, St. Petersburg, Russian Federation

Abstract. At the present time, a broad spectrum of CD8+ T lymphocyte subsets is revealed, including naive cells, memory cells and regulatory subpopulations. Along with cells with high cytolytic activity, some subsets with marked regulatory activity were found there. Each subpopulation is characterized by a set of produced mediators, surface and intracellular markers allowing to suggest their differential in vivo functional activity. The present review article proposes a classification of CD8+ Т cells which takes into account their morphological and functional features. According to conventional view, the CD8+ Т lymphocytes is a cell population exhibiting high cytotoxic ability which is of critical significance in pregnancy, under the conditions of semi-allogenic fetal cell invasion into the endometrium. The fraction of CD8+ T cells is rather high in decidual structures. The review discusses the known mechanisms of differentiation regulation, selective migration and activity of CD8+ T cells in decidual membrane and placenta in the course of pregnancy. Perforine and granzyme are the main cytotoxicity factors of CD8+ Т cells. IL-2, IL-5, IL-13, IFNy, IL-17, TGF-p and IL-10 cytokines are considered regulatory mediators of CD8+ cells. To induce the effector properties of CD8+ T cells, an antigenic stimulation is required, which is provided by interactions between the CD8+ Т cells and activated CD4+ Т cells or dendritic cells, cytokine effects. Specific differentiation of the CD8+ T cells is determined by differences in microenvironvent. In the course of pregnancy, accumulation of CD8+ Т cells is observed in decidual membrane, but their phenotype and functional properties differ from CD8+ Т cells in peripheral blood. At present time, the mechanisms of selective CD8+ T cell migration to decidual membrane are studied. These events are suggested to be mediated by means of CXCR3 and CCR5 chemokine receptors, IL-6 and IL-15 cytokines. The features of CD8+ Т cell activities, and production of some cytokines, e.g., CSF2, IFNy, IL-1p, IL-2, IL-6, IL-8,IL-10, IL-12 and TNFa in decidual membrane and is of critical significance for effective invasion of trophoblast cells. In turn, the trophoblast and placental cells promote development of regulatory CD8+ Т lymphocytes in decidual membrane, being able to induce CD8+ T cell apoptosis in decidual membrane. Hence, interaction between the maternal CD8+ T cells and trophoblast in the area of uterine-placental contact is an important link during development of immunological tolerance in the maternal/fetal system.

Keywords: T lymphocytes, pregnancy, cytotoxic, decidua

The present work was supported by the Russian Foundation for Basic Research grant No. 17-0400679 and the Russian Science Foundation grant No. 17-15-01230.

Introduction

Cytotoxic lymphocytes are one of the key cellular elements, whose action is aimed at viral clearance. However, the spectrum of their function is much wider. It is noted that the change in metabolic pathways and function of CD8+ T lymphocytes is

associated with aging of the organism [46]. In connection with the possibility of recognition of MHC I, CD8+ T lymphocytes are an important part of the antitumor immune response. In particular, activated CD8+ T lymphocytes control cell growth in solid tumours [23]. CD8+ T lymphocytes are involved in many pathological processes, for example, in nerve fibre damage in multiple sclerosis [133], cerebral vasculature in cerebral malaria [126], bronchial asthma [95], rheumatoid arthritis [16] and immunopathology in skin leishmaniasis [94]. In the host graft system,

the effect of CD8+ T lymphocytes is also crucial for engraftment of the graft [5, 112, 151].

Pregnancy is a unique example of the coexistence of genetically diverse tissues. In this case, the placenta is not an absolute barrier to the cells of the immune system of the mother: there is a mutual penetration of the cells of the mother and foetus — the phenomenon of microchimerism, which can both positively and negatively affect the outcome of pregnancy [101, 108]. T lymphocytes are of great importance for the recognition and elimination of foreign genetic material in the body, however, in the case of physiological pregnancy, there is no attack from the mother's immune system — explained by the immunological tolerance for the foetus. At present, the mechanisms of the development of this tolerance are intensively studied. In the endometrium and decidua, a change in the CD8+ T lymphocyte content is observed when pregnancy occurs compared with non-pregnant women and obstetric pathologies compared to the physiological course of pregnancy, but the role of these cells has not been sufficiently studied. It has been established that along with CD8+ T lymphocytes possessing cytotoxic action, there are CD8+ T lymphocytes with regulatory properties, as well as several subpopulations of memory cells. CD8+ T lymphocytes probably play one of the most important roles in the preservation or rejection of the foetus. At the present time, however, the whole spectrum of CD8+ T lymphocyte subpopulations is insufficiently characterized and the role of these cells in maintaining a physiological pregnancy is not fully understood.

Phenotype and functional activity of CD8+ T lymphocytes

For a long time, CD8+ T lymphocytes have been considered a homogeneous population. At present, it has become clear that the diversity of CD8+ T lymphocyte subpopulations is determined by their differences both in phenotype, and in functional purpose, and in the characteristics of intercellular interactions. The vast majority of CD8+ T lymphocytes express the transcription factors T-bet and STAT4 and secrete the cytokines IFNy and TNFa [93]. The main factors of the cytotoxic effect of CD8+ T lymphocytes are perforin and granzymes [93]. In addition, CD8+ T lymphocytes express a rather wide spectrum of receptors characteristic for NK cells: KIR - CD158a/h, CD158bj, CD94, NKG2A, NKG2C [132].

Several different subpopulations of CD8+ T lymphocytes are described in the public sources. The earliest classification of these lymphocytes is based on the spectrum of cytokines secreted by them. According to this classification, CD8+ T lymphocytes can be

divided into 3 subpopulations: Tc1 — producing IFNy and not producing IL-4; Tc2 — not producing IFNy, producing IL-4; and Tc0 — producing IFNy and IL-4 [90, 138]. The majority of CD8+ T lymphocytes of human peripheral blood (80%) belongs to Tc1 [138]. Tc1 secrete IL-2, IFNy, small amounts of IL-5, IL-13, and express the transcription factors T-bet and Hlx. Tc2 and Tc0 secrete IL-4, IL-5, IL-10, IL-13 [138]. Tc2 express the transcription factors GATA3 and Hlx [83]. Tc0 and Tc2 are characterized by high expression of CD30, CD40L, CD28, whereas Tc1 have a low expression of these molecules [138]. Tc2 are identified by the expression of the CRTH2 marker. The cytotoxic activity of the three subpopulations of CD8+ T lymphocytes is not different [138]. At the functional level, Tc1 accumulates faster in the lymph nodes and promotes a faster development of the immune response, and they are more apoptotic after activation (the mechanism does not involve changes in Fas and FasL expression) than Tc2 [128, 138]. Cytokine IL-4 stimulates the development of IL-4-producing subpopulations, suppressing production of IFNy [138]. The effect of IL-4 results in a loss of ability of Tc1 to secrete IL-2, and hence a loss of ability to spontaneously proliferate [111]. IL-12 inhibits the production of IL-5, IL-4 and IL-10, but stimulates the production of IFNy, stimulates the development of the Tc1 subpopulation [138]. Subpopulations of Tc0 and Tc2 are not affected by IL-12 or IL-4 [138]. Prostaglandin E2 promotes a shift in the equilibrium toward Tc2 [10]. Due to the secretion of various cytokines, Tc can change the direction of differentiation of CD4+ T lymphocytes. Currently, there are still studies using this classification.

In the most frequently used classification, CD8+ T lymphocytes are divided depending on the expressed markers on naïve (CD45RA+ CCR7+), effector (CD45RA+CCR7-), effector memory cells (CD45RA-CCR7-) and memory cells (CD45RA-CCR7+) [130]. Naïve CD8+ T lymphocytes have the phenotype CD44low; when activated, the expression of this marker increases [22]. Depending on the expression of CD28 and CD27, effector cells and memory effector cells are divided into the following subpopulations: EM1 (CD28+CD27+), EM2 (CD28-CD27+), EM3 (CD28"CD27"), and EM4 (CD28+CD27-) [130].

During the immune response, CD8+ T lymphocytes form memory cells, which are divided into three types: resident, effector and central [70]. Resident memory CD8+ T lymphocytes are localized in non-lymphoid tissues (mucous membranes and reproductive tract), differ in the inability to leave tissues in circulation and are characterized by the

phenotype CD8+CD103+CD69+ [70, 124]. Effective CD8+ T memory lymphocytes can migrate between tissues and secondary lymphoid organs, they do not express homing molecules that determine migration to the lymph nodes [124], are characterized by the CD8+CD45RO+CD62L-CCR7- [70] phenotype, high expression of cytolytic enzymes [130], high expression of transcription factor GATA3, high level of expression of IL-6Ra, IL-7Ra and increased level of proliferation [74]. Under the in vitro conditions, it was shown that after activation (CD3/CD28), these cells also produce cytokines IL-2, IL-5, IL-13 and IFNy [74]. In the experiment, it was shown that the proliferation of effector CD8+ T lymphocytes is stimulated by IFNy, with the synergistic action of IL-6 and IL-15 [74]. Also, the proliferation of effector CD8+ T lymphocytes can support IL-2 (autocrine). Cytokines secreted by effector memory CD8+ T lymphocytes are involved in antiviral and antibacterial defence of the body, in addition, there is an increase in the content of these cells in autoimmune processes (bronchial asthma) [74]. Central memory CD8+ T lymphocytes are localized in secondary lymphoid organs, they express homing molecules that determine migration to the lymph nodes, have a CD8+CD45RO+CD62L+CCR7+ phenotype and a high proliferative potential upon re-encounter with antigens [70, 124].

By analogy with CD4+ lymphocytes, Tc17 producing IL-17 are isolated. They are characterized by the phenotype CD27-CD28+CD45RA-CCR5+CCR6+, secretion of cytokines IL-17, IFNy [68]. A subpopulation of CD8+ non-cytotoxic IL-17-producing T lymphocytes (Tcn17) is also formed, which is formed in the presence of TGF-p and IL-6 [83]. This subpopulation is characterized by the expression of the transcription factor Th17 RORyt, reduced expression of the transcription factors GATA3, T-bet, Hlx, the lack of production of IFNy, granzyme B, IL-10 and the absence of cytolytic activity [83].

CD8+ regulatory T lymphocytes (CD8+ Treg) are a population of cells newly identified and are currently intensively studied. Along with CD4+ Treg, CD8+ Treg are important for the formation of immunological tolerance [84]. Their presence in immunologically privileged organs, participation in many pathologies of immune genesis, as well as their key role in the host-transplant reaction have been described. For CD8+ Treg, the expression of CTLA-4, CD25, HLA-DR, CD45RA, CCR7, CD62L, CD28, CD101, CD103, CD122, TcRa/p, ICOS, FOXO1 and HELIOS, IL-2ra, CCR4, GARP, IL-10 and TGF-p [2, 7, 37, 55, 75, 84, 119]. Expression of these molecules is important both for maintaining the CD8+ Treg population and

for manifesting their functional activity against other cells [55]. According to some data, CD8+ Treg does not express CTLA-4, FasL [119]. CD4+ T lymphocytes requires the expression of FoxP3 for the implementation of regulatory properties [39, 121, 148]. Depending on the expression of FoxP3 CD8+ Treg can be divided into CD8+FoxP3+ and CD8+FoxP3- subpopulations. For CD8+FoxP3+Treg, CD62L expression is also characteristic. Phenotype CD8+ FoxP3- Treg is characterized by the expression of CD103, which is crucial for the formation and implementation of suppressive properties of CD8+ Treg lymphocytes, especially for the subpopulation of CD8+ FoxP3 cells [75, 84]. There are data on intracellular expression of FoxP3, characteristic of CD8+ Treg of lymphocytes [7, 55]. Expression of surface molecules is largely specifically regulated by various microRNAs [55]. Probably, the formation of two subpopulations (CD8+FoxP3+ and CD8+FoxP3- Treg) is associated with the action of TGF-p (in conjunction with the activation of the T cell receptor) and IL-10 [37, 84]. Both subpopulations have a low expression of perforin and granzymes, intracellular expression and secretion of IL-10, and their suppressive effect is not due to cytotoxic effects, but to the production of TGF-p and IL-10 and contact interactions involving the CTLA-4 molecule [55, 84, 119].

Thus, the population of CD8+ T lymphocytes is a collection of cells quite diverse in phenotype and function. The conditions for differentiation of CD8+ T lymphocytes are also different.

Differentiation of CD8+ T lymphocytes

Recent thymic emigrants that have just emerged from the bone marrow are localized in the blood and spleen, and differ from mature naïve cells: they have a decreased secretion of immunoregulatory cytokines, reduced expression of CD62L, CXCR4, granzyme B, reduced cytotoxicity, the ability to rapidly proliferate with an immune response and provide antiviral protection [27]. It is believed that decidual CD8+ T lymphocytes refer to such recent thymic emigrants at the periphery, but this issue has not been studied enough.

The antigen-independent differentiation of CD8+ T lymphocytes occurs in the thymus, from where the naïve cells exit into the periphery. In addition, naïve CD8+ T lymphocytes undergo antigen-dependent differentiation in secondary lymphoid organs: in the lymph nodes [15, 135] and in the spleen [11], naïve CD8+ T lymphocytes interact with dendritic cells (DC) via the XCR1 + molecule in the presence of secreted IL-15, and also in the presence of CD4+ T lymphocytes secreting IL-2 [70]. Naïve CD8+ T lymphocytes are located in the paracortical area of the lymph node [63].

In secondary lymphoid organs, CD8+ T lymphocytes, along with DC, contribute to the activation of CD4+ T lymphocytes [11]. The interaction of CD8+ T lymphocytes with CD4+ T helper cells is not necessarily accompanied by detachment from DC [11]. Further, the aggregate migrates into the white pulp of the spleen [11].

After activation, CD8+ T lymphocytes can differentiate into short-living effector cells that attack infected cells and die as a result of apoptosis after performing their function, or differentiate into circulating central memory cells [9]. About 5-10% of activated CD8+ T lymphocytes are transformed into memory cells [9]. Central memory cells are localized mainly in the interfollicular region of the lymph nodes [63]. CD8+ T lymphocytes are stored in the lymph nodes due to the expression of the homing receptors CCR7 and CD62L (L-selectin) [131].

Developing the effector properties of CD8+ T lymphocytes requires a triple signal: antigen stimulation through TcR, a costimulatory signal from CD28, microenvironment — exposure to cytokines IL-12, IFNa, IFNp [24, 88]. IL-12 and IFN type I cytokines play an important role both for the formation of a pool of effector cells and for the formation of memory cells [147]. These cytokines promote differentiation mainly into the Tc1 subpopulation [90]. The effect of cytokines causes a change in the expression of hundreds of genes responsible for proliferation, cytotoxic lymphocyte effector activity, survivability, migration, a wide range of transcription factors (including T-bet and Blimp-1) [24]. Secreted on the first day after activation of naïve CD8+T lymphocytes, IFNy via autocrine action activates and differentiates CD8+ T lymphocytes into cytotoxic T lymphocytes, stimulates the expression of the transcription factor T-bet, granzyme B production and plays an important role in early activation [22]. The effect of IFNy is synergistically enhanced by the action of IFNa and IFNp, but it does not matter for the differentiation of naïve CD8+ T lymphocytes in the presence of IL-12 [22].

The interaction of CD8+ T lymphocytes with activated CD4+ T lymphocytes, DC, is important for the formation of a pool of memory CD8+ T lymphocytes, as well as the effect of cytokines. The interaction of CD8+ T lymphocytes with DC is exercised through the CD70 molecule on the DC and CD27 on the surface of CD8+ T lymphocytes, and the interaction of CD40L on activated CD4+ T lymphocytes with CD40 on DC is simultaneously observed [50]. The expansion of memory CD8+ T lymphocytes depends on the interaction with CD4 T helper cells via CD40/CD40L and minor histocompatibility molecules [109]. The kinetics of cellular interaction remains a debating point [50]. It is believed that there

is also a reserve pathway for maintaining memory cell differentiation, which is relevant in the absence of IL-12 and IFN type I, but it is not yet identified [147].

In the absence of CD4+ T lymphocytes, naïve CD8+ T lymphocytes cannot activate into effector cells, whereas memory CD8+ T lymphocytes can proliferate and differentiate into effector cells [76].

The IL7R, IL2R, IL15R, microRNA (miR)-155 receptors play an important role in determining the direction of differentiation of CD8+ T lymphocytes in the direction of the effector or memory cells, as well as the nature of the activation of TcR, in particular, the impact through the molecules of diacylglycerol (DAG), DAG kinase (DGK) a and Ç [8, 60, 113, 131, 150]. Deletion of DGK a and Ç leads to a decrease in the number of memory cells. This effect can be detected by observing a decrease in CD8+ expression by T lymphocytes of CD127, CD62L expression, as well as disturbance of the homeostasis of a subpopulation of memory cells and their viability [150]. Also, the deletion of DGK a and Ç causes a decrease in CD8+ T lymphocyte expression of the chemokine receptors CCR4, CCR5 and CXCR3, responsible for migration to the lymph nodes during the immune response [150].

CD8+ T regulatory lymphocytes can differentiate from naïve or effector CD8+ T lymphocytes in both the thymus and periphery [2]. As in the case of CD4+ Treg cells, TGF-p is important in the formation of the CD8+ Treg subpopulation: it suppresses the cytotoxic activity of CD8+ lymphocytes [127] by inducing microRNAs, leading to suppression of IFNy production [17, 84]. The effect of TGF-p was also manifested in the increased expression of CTLA-4, ICOS by the CD8+ T lymphocytes, and the CD62L on the FoxP3+ subpopulation of CD8+ T lymphocytes, but molecule CD103 on the subpopulation of CD8+FoxP3" T lymphocytes [75, 84]. TGF-p-induced CD8+FoxP3+ and FoxP3- cells reduce production of IFNy and do not proliferate in response to IL-2 [84]. The effect of monoclonal anti-CD3 antibodies with in vitro conditions leads to the induction of differentiation and expansion of CD8+CD25+FoxP3+ Treg lymphocytes [32]. Mesenchymal stem cells (MSC) of the placenta promote the differentiation of the CD8+ Treg lymphocytes (CD8+IL-10+) with the participation of the PDL2 molecule (anti-programmed death ligand-2) [77]. IFNy and TNFa stimulate the expression of PDL2 on placental MSC and differentiation of CD8+IL-10+ lymphocytes [77].

Thus, CD8+ T lymphocytes are capable of differentiation both in the thymus and in the periphery. The direction of differentiation depends on the nature of the intercellular interactions within the antigen presentation process in conjunction with the action

of soluble factors. The mechanisms of regulation of differentiation of CD8+ T lymphocytes, in particular at the periphery, have not been sufficiently studied. After differentiation, CD8+ T lymphocytes acquire the ability to specifically migrate to further their function.

Migration of CD8+ T lymphocytes to the decidua

CD8+ T lymphocytes are present in peripheral blood, endometrium of non-pregnant women and in the decidua during pregnancy. There is little data on migration mechanisms of CD8+ T lymphocytes. However, several specific molecules can be isolated, whose expression promotes selective migration of cytotoxic lymphocytes into the decidua. Migration of CD8+ T lymphocytes to the decidua is facilitated by an increase in the expression of CXCR3 [21] and CCR5 [63, 93]. CXCR3 ligands produce decidual stromal cells: CXCL9 (MIG), CXCL10 (IP-10), CXCL11 (I-TAC) [30]. CXCL9 is expressed in the placenta and in the chorioamniotic membrane, is found in the bloodstream, and the increased production of this molecule is associated with premature delivery and low foetal weight [29, 62, 65, 105, 115]. The increased CXCL11 content in amniotic fluid in the second trimester of pregnancy is associated with premature delivery [30] and may be associated with the involvement of CD8+ T lymphocytes by enhancing their migration. CCR5 ligands are CCL3 (MIP-1a), CCL4 (MIP-1p), RANTES. CCL4 is produced by decidual stromal cells [30], and CCL3 is expressed in the placenta by trophoblast, endothelial cells [1]. RANTES is produced by the decidua [28] and the placenta [47]. Trophoblast produces MIP-1a and RANTES, CCL3 [54, 99].

The decidua is observed to be the producer of IL-15 [96]. Migration of CD8+ memory cells largely depends on the binding of P- and E-selectins, the expression of which increases in the presence of IL-15 [66]. CD8+ T lymphocytes are characterized by expression of the CXCR6 receptor, which, through interaction with the chemokine CXCL16 secreted by trophoblast, attracts T lymphocytes to the decidua [52]. CD103 expression can be associated with the migration of CD8+ Treg into the inflammation zone [56, 84]. In the placenta, the CD103 ligand -E-cadherin, is expressed by the villous trophoblast and is important for the functioning of the trophoblast itself [14, 78, 81], as well as the decidua [33, 154]. Expression by villous and extravillous trophoblast, endometrial cells of prostaglandin D2 can cause specific migration to the decidua (to the implantation site of the foetal egg) of Tc2 lymphocytes due to their expression of CRTH2 [89].

When endometrial cells are differentiated into decidual cells, the expression of CXCL9 (MIG), CXCL10 (IP10), CXCL11 (ITAC) and CCL5 (RANTES) is suppressed, which leads to a decrease

in Tc1 involvement in the decidua compared to the endometrium of non-pregnant women [35]. This can be one of the mechanisms of formation of immunological tolerance in the mother-foetus system.

If there is an infection, the migration of CD8+ T lymphocytes to the decidua increases, activation of CD8+ T lymphocytes occurs and im-munological aggression against the foetus without elimination of the pathogen develops due to the action of CD8+ T lymphocytes [21]. Expression of E-cadhe-rin with syncytiotrophoblast during preeclampsia is enhanced in comparison with physiological pregnancy [53, 81], which may contribute to excessive involvement of CD8+ T lymphocytes to the placenta.

Cytokine IL-6 plays an important role in the formation and development of pregnancy, is found in significant amounts in the stroma of the decidua, trophoblast and endothelium of the spiral arteries. It has been established that IL-6 promotes the proliferation and differentiation of CD8+CD25+FoxP3+ Treg, an increase of their share in the blood stream and, as a result, an increase in their migration to the decidua [42, 56, 92], which can significantly contribute to the formation of immunological tolerance of mother's immune system in relation to the foetus and the development of the placenta [102], as well as in the process of rejection of the foetal tissues during the delivery [97].

In the endometrium of non-pregnant women, the proportion of IFNy+CD8+ T lymphocytes reaches 25% of all CD8+ T lymphocytes; in spontaneous abortions, there is a significant decrease in the content of IFNy+CD8+ T lymphocytes in the decidua compared with the endometrium of non-pregnant women (less than 11% of CD8+ T lymphocytes) [31], which indicates the important role of CD8+ T lymphocytes in implantation and the establishment of pregnancy.

Currently, there is no consensus on the persistence of the population of cytotoxic lymphocytes in the decidua during pregnancy. Some authors claim that during the pregnancy, the amount of CD8+ T lymphocytes in the decidua of women does not change [145] and is 45-75% of the T lymphocytes of the decidua (T lymphocytes constitute 10-20% of decidual leukocytes) [34, 52, 93]. Other authors point out a different content of cytotoxic lymphocytes in the decidua, depending on the period of pregnancy. In the first trimester of pregnancy, the proportion of CD8+ T lymphocytes in the decidua is insignificant and is 2-7% of the CD45+ cells, while in the third trimester of pregnancy it is up to 30% [21]. Such a difference in the data obtained by the authors on the content of T lymphocytes in the decidua can be associated with the absence of standardized methods for isolating cells from the decidua.

The ratio of T cell populations in the systemic blood stream and decidua is different. In the decidua, the CD8+ T lymphocyte count is 62%, and CD4+ is 33% [91] of the total number of T lymphocytes, whereas in the peripheral blood, the proportion of CD8+ lymphocytes is 21%, and 75% of CD4+ lymphocytes [91]. The number of CD8+ memory T lymphocytes in the decidua considerably exceeds their number in peripheral blood [134], and the proportion of naïve CD8+ T lymphocytes is much lower than in peripheral blood [130], which indicates the presence of predominantly differentiated cells. Decidual CD8+ T lymphocytes have a lower degree of expression of perforin and granzyme B compared to CD8+ T lymphocytes of peripheral blood, and a small number of CD8+ T lymphocytes expressing granzyme B [130] (Table 1).

With pathologies of pregnancy, there is a change in the content of CD8+ T lymphocytes in the blood and decidua. Thus, with preeclampsia, a decrease in CD8+FoxP3+ T lymphocytes (CD8+ T regulatory cells) in peripheral blood of women has been

observed [152]. Spontaneous abortion shows an increased CD8+ lymphocyte count in the decidua [6]. The development of chronic chorioamnionitis in pregnant women is accompanied by an increase in the cytotoxic activity of CD8+ T lymphocytes, characterized by the phenotype CD300a+CD8+, in the systemic blood flow compared to women with a physiological pregnancy [64].

Thus, CD8+ T lymphocytes play one of the decisive roles in the physiological and pathological development of pregnancy. Not only the existence of specific migration mechanisms of various subpopulations of CD8+ T lymphocytes in the endometrium and decidua is observed, but their concentration in these tissues in comparison with peripheral blood. At the same time, the ratio of various subpopulations of CD8+ T lymphocytes in the decidua at various stages of development of pregnancy is substantive.

Phenotypic and functional features of CD8+ T lymphocytes of the decidua

The presence of CD8+ T lymphocytes in the endo-metrium is important for the implantation of the

TABLE 1. CHARACTERISTICS OF SUBPOPULATIONS OF DECIDUAL CD8+ T LYMPHOCYTES

Subpopulation of decidual CD8+ lymphocytes Surface markers Intracellular markers Secreted cytokines Share in the decidua

naïve CD8+, CD45RA+, CCR7+, CD62L+, CD45RO- [87], CD27+, CD28+ [131], CD44low [22] T-bet and STAT4 [93] IFNy, TNFa [93] 3% [131]

Resident memory cells CD8+CD45R0+CD103+CD69+ [70, 136]

Memory cells Central memory cells CD8+,CD45RO+, CCR7+, CD62L+, CD45RA-, CD27+, CD28+ [70] Eomeslow [87] 2% [131]

Memory cells (memory effector cells) CD8+, CD45RO+, CD62L-, CCR7- [70], CD45RA- [131], IL-6Ra+, IL-7Ra+ [41, 74], CD103+IC0S+, CD69+, HLA-DR+ [91, 129, 130, 134] GATA3 [74], Tbet+, Eomes+ [87] Contradictory data on the production of cytolytic enzymes (increased [130], reduced expression of granzyme B and perforin [134]) IL-2, IL-5, IL-13, IFNy [74] 65% [131]

Effector T lymphocytes CD8+, CD45RA+ [131], CD45RO- [87], CD62L-, CCR7-[41] T-bet+ [87] Reduced expression of granzyme B and perforin 13% [131]

CD8-suppressor (regulatory) cells CD8+CD28-, CD69+, HLA-DR+ [129], CD103+/-, CD62L+/-, CD25+, HLA-DR+, CD45RA+, CCR7+, CD28+, CD101+, CD122+, TcRa/ß+, ICOS+, FOXO1+, HELIOS+, IL-2RA+, CCR4+, GARP+ [2, 7, 37, 55, 75, 84, 119] FoxP3+/- [41] Vbeta 9 [119] CCR4, IL-10 and TGF-p [2, 7, 37, 55, 75, 84, 119] IL-10, low expression of perforin and granzyme

Tc17 CD27-, CD28+, CD45RA-, CCR5+, CCR6+ [18] IL-17, IFNy [68]

ovum. Cells of the Tc2 subpopulation predominate in the implantation site. They express CRTH2, which is an additional receptor of prostaglandin D2 and is characteristic of Th2 [89]. The cytolytic activity of CD8+ T lymphocytes is hormone-dependent: it is present in the proliferative phase of the menstrual cycle and is absent in the secretory phase [142].

Decidual CD8+ T lymphocytes differ in phenotype and functional characteristics from peripheral blood lymphocytes (Table 1). In the bloodstream, naive CD8+T lymphocytes predominate [131]. Decidual and placental CD8+ T lymphocytes in the vast majority are activated effector cells or effector memory Т cells [131] (Table 1). Activated effector cells are characterized by the phenotype CD45RA+, CD45RO-, CD62L-, CCR7-, expression of Tbet transcription factor [41, 87, 131]. Effector memory cells are characterized by the phenotype CD8+CD45RA-CCR7-CD28-CD45RO+CD103+ICOS+, increased expression of CD69 and HLA-DR and lack of perforin. Whereas in the peripheral blood, naive CD8 T cells (CD8+CD45RA+CD45RO-CCR7+) predominate [91, 129, 130, 134]. Also for effector memory T cells, expression of NK cell receptors (KIR — CD158a/h, CD158bj, CD94, NKG2A, NKG2C) is characteristic [69, 132]. NK receptor expression is controlled by the cytokines IL-2, IL-4, IL-23, TGF-p, IL-15, IL-6, IL-10, IL-21, and also depends on the nature of the antigen (due to the interaction with CD28) [132]. Expression of one ofthe NK receptors — CD158a/h, is increased in CD8+ T lymphocytes of the decidua compared to CD8+ T lymphocytes of peripheral blood; the expression of the remaining NK receptors on CD8+ T lymphocytes of the decidua and peripheral blood is the same [132]. At the same time, decidual CD8+ T lymphocytes are characterized by reduced cytotoxic activity compared with peripheral CD8+ T lymphocytes. One of the proposed mechanisms for this phenomenon may be the reduced expression of FasL, as well as perforin and granzyme B. At the same time, high expression of mRNA perforin and granzyme B was established, which indicates a post-translational mechanism of blocking the production of these proteins [130, 134]. Also, reduced cytotoxicity may be associated with the effect of the locus molecules HLA-G and HLA-C expressed by the trophoblast. The functions of NK receptors on CD8+ T lymphocytes have not been adequately studied at present.

The decidual CD8+ T lymphocytes of the first trimester of pregnancy secrete in vitro CSF2, IFNy, IL-1p, IL-2, IL-6, IL-8, IL-10, IL-12, and TNF (especially IFNy and IL-8), that stimulates trophoblast invasion [91, 114]. A high level of TGF-p provides a high threshold level of cytolytic activity of

CD8+ T lymphocytes in the decidua [21], which also contributes to the successful invasion of trophoblast. This effect can be associated with suppression of CD8 molecule expression by cytotoxic T lymphocytes in the presence of TGF-p [98].

An ICOS molecule is expressed on 96% of decidual CD8+ T lymphocytes [91], which belongs to the family of CD28-B7 costimulatory molecules. Expression of ICOS is inducible. ICOS binds to ICOSL, expressed on professional APC (B cells, macrophages, DC), epithelial and endothelial cells [143], including on the trophoblast [91]. It was noted that as a result of ICOS binding to ligand, decidual CD8+ T lymphocytes secrete more IFNy and IL-10 than CD8+ T lymphocytes of peripheral blood [91]. Such stimulation of secretory activity of CD8+ T lymphocytes upon contact with trophoblast through ICOS can be directed to stimulation of trophoblast invasion. It was shown that the ICOS molecule is important for the realization of the immunological response by T helper cells (cell proliferation, activation), T regulatory cells (differentiation, proliferation) [146] and Th17 lymphocytes [61, 91, 143]. The important role of the ICOS molecule expressed by CD8+ T lymphocytes in the antigen-dependent response in tissue transplantation has been shown. In particular, in the "graft versus host" system, the deletion of ICOS on CD8+ T lymphocytes leads to a fatal outcome, caused by excessive expansion and secretory activity of CD8+ T lymphocytes [153]. In peripheral CD8+ T lymphocytes, early expression of ICOS results in migration to non-lymphoid organs, increased cytolytic activity and decreased ability of CD8+ T lymphocytes to participate in the development of a secondary immune response [82]. Probably, decidual CD8+ T lymphocytes thus contribute to the formation of a favourable cytokine microenvironment and a decrease in the cytotoxicity of the cells of the immune system of the mother during the trophoblast implantation. On the other hand, decidual CD8+ T lymphocytes of the first trimester of pregnancy in vitro have the ability for extracellular cytolysis [114]. When decidual CD8+ T lymphocytes are exposed to proinflammatory cytokines and activation of their TcR, their expression of perforin and granzyme B, as well as their degranulation of cytotoxic granules, is enhanced, which distinguishes these cells from decidual NK cells characterized by greatly reduced cytolytic activity [21]. Simultaneously, decidual CD8+ T lymphocytes produce more granulin as compared to a similar subpopulation of peripheral blood, which can provide local protection against intracellular and extracellular pathogens [21].

The proteins Tim-3 (T cell immunoglobulin mucin-3) and PD-1 (programmed cell death-1)

are important signalling molecules of CD8+ T lymphocytes. Their functions have not been sufficiently studied. The high level of expression of PD-1 and Tim-3 CD8+ T lymphocytes correlates with reduced antiviral activity of these cells, reduced proliferative activity of CD8+ T lymphocytes and reduced secretion of pro-inflammatory cytokines CD8+ T lymphocytes, while their blockade leads to restoration of proliferation and synthetic activity cells [57, 100]. There is also evidence that the interaction of PD-1 and B7-H1 determines the cytotoxic pathway of differentiation of CD8+ T lymphocytes [40]. Signals from PD-1 and Tim-3 synergistically reinforce each other [57, 140]. It has been shown that Tim+PD-1+CD8+ T lymphocytes are concentrated in the decidua in a physiological pregnancy and participate in the establishment of pregnancy in the early stages [57, 140, 149]. Tim+PD-1+CD8+ T lymphocytes show an increased level of proliferative activity in comparison with Tim-3-PD-1-CD8+ lymphocytes, have greater expression of activation markers CD69 and HLA-DR and greater secretion of Th2 cytokines (in particular, IL-4 and IL-10) [140]. In the case of spontaneous abortions, both the amount of Tim+PD-1+CD8+ T lymphocytes in the decidua, and the production of anti-inflammatory cytokines are reduced. At the same time, increased production of proinflammatory cytokines IFNy and TNFa is observed [149]. The mouse model shows that the blockade of Tim-3 and PD-1 leads to a significant decrease in the weight of the foetus and the number of offspring, which indicates the important role of these molecules in providing foetal protection and normal pregnancy [140]. In women with spontaneous abortions, an association of a decreased number of decidual CD8+ T lymphocytes with the PD-1+CD8+ phenotype and a decreased proliferative activity has been observed [140]. PD-1L is expressed on syncytiotrophoblast cells directly in contact with the parent cells [137], which is one of the mechanisms of formation of immunological tolerance for trophoblast cells. Expression of PD-1L with trophoblast is stimulated by EGF and IFNy [48]. It is likely that increased production of IFNy trophoblast in early pregnancy [3, 4] and its autocrine effect may be one of the mechanisms of survival of a semi-allogenic foetus as a result of the interaction of PD-1 on T lymphocytes and PD-1L on the trophoblast. From the first to the second trimester, the expression of PD-1L in the placenta is increased, the regulation of expression is probably carried out depending on oxygenation (insufficient oxygenation in the uteroplacental blood flow zone causes a decrease in expression of PD-1L) [49]. Decreased expression of PD-1L in the decidua is associated with habitual miscarriage [77].

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

Thus, Tim and PD-1 are important in establishing the tolerogenic nature of CD8+ T lymphocyte differentiation in pregnancy.

Factors secreted by the placenta and decidua, significantly affect the formation of immunological tolerance in pregnancy, in particular, affect the apoptosis of the mother's cells. It was noted that the level of apoptosis of CD3+ T lymphocytes in peripheral blood is higher in pregnancy than in non-pregnant women [25]. At the same time, the level of apoptosis of CD3+CD8+ T lymphocytes at the onset of pregnancy is lower than in non-pregnant women, and the maximum level of apoptosis of this subpopulation of T lymphocytes is observed in the first trimester of pregnancy [25], which can be aimed at ensuring foetal survival in early pregnancy and elimination of alloreactive cells. The decidua produces significant amounts of glycodelins A (GdA), which inhibits the cytotoxic activity of activated CD8+ T lymphocytes and their proliferation [123] using CD7 as a receptor [125]. GdA suppresses the synthesis of IL-2R, which interferes with the action of IL-2 on T lymphocytes and the formation of functionally mature cytotoxic cells [122]. The placenta secrets the preimplantation factor (PIF), which binds to the proteins of the CD8+ T lymphocytes cytoskeleton and suppresses the immune response of the mother [12]. Interaction with soluble HLA-C, whose concentration in the peripheral blood rises in pregnancy, causes apoptosis of CD8+ T lymphocytes by enhancing its production of FasL [19, 104].

CD8+ T lymphocytes active against paternal antigens, including anti-minor histocompatibility antigens (mHag-peptides derived from polymorphic proteins, recognized as heterogenous) have been described in the literature, but they do not exert any influence on the outcome of pregnancy [131]. This may be due to the formation of subpopulations of regulatory CD4 and CD8 lymphocytes, levelling their influence [131].

In pregnancy, virus-specific CD8+ T lymphocytes accumulate in the decidua [134]. But since decidual CD8+ T lymphocytes acquire more regulatory properties than cytotoxic ones, while retaining the ability to recognize the antigen present in the HLA locus molecule, including the viral antigen — there is no complete elimination of the infected cells [21, 134]. Most likely, the activity of CD8+ T lymphocytes can be aimed at resisting a viral infection in the context of maintaining pregnancy.

Thus, decidual CD8+ T lymphocytes differ in phenotype, secretory and cytolytic activity from CD8+ T lymphocytes of peripheral blood, which is aimed at supporting the invasion of trophoblast in the early stages of development of pregnancy, as well as the

formation of immunological tolerance for the half-allogenic foetus. The disturbance of the homeostasis of decidual CD8+ T lymphocytes has serious consequences that affect the outcome of pregnancy. The mechanisms of maintaining the balance of the functioning of CD8+ T lymphocytes within the framework of maintaining the physiological course of pregnancy and at the same time effective protection against infectious agents remain insufficiently studied. The diverse interaction of CD8+ T lymphocytes with microenvironment cells in the utero-placental blood flow zone has a significant effect on the nature of the functional activity of all interacting cell types.

Intercellular interactions of CD8+ T lymphocytes

In pregnancy, trophoblast cells play a central role in the development of immunological tolerance. In the regulation of the cytotoxicity of immune system cells from the trophoblast side, the expression of the locus molecules of the main histocompatibility complex HLA-G and HLA-C is important. Receptors for the molecules of the HLA-G locus on CD8+ T lymphocytes are the molecules CD8, LILRB1 and CD160 [72]. HLA-G locus molecules are expressed on the trophoblast in combination with endoplasmic reticulum aminopeptidase 1 (ERAP1), which generates peptides capable of forming a stable complex with HLA-G [106]. This contributes to the successful invasion of the trophoblast. Expression of this complex on trophoblast cells is enhanced by exposure to LIF and VEGF [106]. The extravillous cytotrophoblast produces both a membrane and a soluble form of HLA-G [59, 67]. The soluble form is found not only in the amniotic fluid, but also in the mother's bloodstream [13, 45, 120]. It was shown that the soluble form of the HLA-G locus molecule promotes immunological tolerance, causing apoptosis of CD8+ T lymphocytes [73], including through interaction with the CD8 molecule [72]. HLA-G locus molecules can also inhibit the proliferation of CD4+ lymphocytes, thereby reducing the risk of developing a mixed lymphocyte response from both the mother and the foetus. The low level of production of HLA-G by the trophoblast is associated with complications, including the development of preeclampsia [45, 85]. At the same time, some of the latest studies have revealed an increased level of HLA-G mRNA in the decidua in preeclampsia [103].

Prostaglandin E2 (PGE2), indolamine 2,3-dioxy-genase (IDO) and TGF-p also participate in the suppression of proliferation of CD8+ lymphocytes [80].

According to the latest data described in the literature, several stages can be distinguished in the interaction of trophoblast and CD8+ T lymphocytes. First, direct recognition of HLA-C by the T cell receptor of CD8+ T lymphocytes. Further, a peptide

(viral or allogenic) is presented in the MHC I complex, activation of CD8 lymphocytes. The mouse model shows that the presentation of foetal antigens occurs exclusively in an indirect way, due to maternal APC, and antigen presentation occurs only in the late stages of pregnancy, which is associated with the late onset of the expression of MHC I molecules on foetal cells [18, 21, 36]. At the same time, it has been repeatedly shown that macrophages and DC of the placenta and decidua are of a tolerogenic nature [26, 38, 79, 116, 118, 139, 144]. The literature also describes the presentation of minor and major histocompatibility antigens in the maternal APC after the capture of trophoblast debris by CD4+ T lymphocytes, which in turn secrete cytokines that affect the differentiation of CD8+ T lymphocytes.

Antigen-presenting cells and trophoblast produce significant amounts of IL-27 [20, 43, 107]. In naïve CD8+ T cells (CD8+CD45RA+CCR7+), IL-27 enhances the expression of the T-bet transcription factor, which indicates the polarization of CD8+ T lymphocytes in the Tc1 direction, stimulates proliferation, IFNy secretion, granzyme B and cytotoxicity of these cells [117]. However, the mechanisms of influence of IL-27 on the development of pregnancy remain unclear. The IL-27 receptor is expressed on CD8+ T lymphocytes to a greater extent than on CD4+ lymphocytes, mainly on CD8+CD45RO+ lymphocytes (i.e., memory cells) [117]. Intracellular expression of the IL-27 receptor (TCCR/gp130) is characteristic of naïve cells, with activation the surface expression of the receptor is enhanced [117]. The mouse model shows that the production of IL-27 by the decidua is enhanced before implantation [86]. In women with habitual miscarriages, much lower expression of IL-27 in the decidua is observed compared to women with spontaneous abortion and the physiological course of pregnancy [141].

The extravillous cytotrophoblast expresses the B7H2 molecule (ICOS ligand). The specific interaction of ICOS/B7H2 contributes to the secretion of T cells by cytokines IFNy, IL-10 and IL-2 [91]. According to other data, foetal formation causes strong proliferation of CD8+ T lymphocytes in the uterine lymph nodes during pregnancy, but they are simultaneously susceptible to clonal deletion [36]. The remaining activated CD8+ T lymphocytes are characterized by reduced secretion of IFNy, IL-2 and IL-4 and inability to cytolytic activity [36]. Previously, it was also noted that IL-4, secreted by Th2, leads to a decrease in the production of IFNy, TNFa and IL-10 by Tc1 cells [110].

It has been established that trophoblast stimulates the formation of a subpopulation of CD8+

T regulatory cells [77, 119]. Since activation of CD8+ T lymphocytes involves CD4+ T lymphocytes, they will have a significant effect on the functioning of cytotoxic T lymphocytes. CD4+ Treg cells by secretion of IL-10 contribute to the formation of CD8+ cell memory in viral infection [71]. CD4+ Treg lymphocytes suppress CD8+ T cell response: CD4+ Treg cause apoptosis of CD8+ cytotoxic T lymphocytes and reduce their expression of granzymes A and B [44]. CD4+ Th1 lymphocytes are involved in the induction and maintenance of viability of CD8+ lymphocytes (Tc1), both effector and memory cells, through IL-2 secretion, activation of DC by CD40/ CD40L interactions, and also using pMHC I [51]. CD8+ Treg stimulate the differentiation of CD4+ Treg [75]. Subpopulations of CD8 Tc1 and Tc2 lymphocytes differently influence the differentiation of CD4+ T lymphocytes: in the presence of Tc1, Th1-lymphocytes predominate, and in the presence of Tc2, Th2-lymphocytes predominate [138]. It has been shown that CD8+ T lymphocytes stimulate the expansion of cells of a specific subset of CD4+ Treg expressing the TCR Vp5 chain whose proliferation regulation mechanism differs significantly from other Tregs and does not depend on IL-2 [58]. The enhancement of the surface expression of TNFa, observed with the activation of CD8+ lymphocytes, promotes the expansion of Vp5+ Treg lymphocytes [58]. Simultaneously, the expression of TNFR2 on Vp5+ Treg plays a leading role in inducing the proliferation of these cells [58]. The action of Treg is aimed at limiting the cytotoxic activity of CD8+ T lymphocytes, therefore the revealed regulation mechanism is a system of mutual influence of T cell subpopulations during the antiviral response. The results were obtained on a mouse model, but there is reason to assume that such a mechanism exists in humans [58].

The secretory products of decidual CD8+ lymphocytes of the first trimester of pregnancy stimulate trophoblast invasion [114], which can be mediated by the action of IFNy. Secreted by CD8+ lymphocytes, IL-8 can play an important role in vascular remodelling in the first trimester of pregnancy by stimulating MMP production. Thus, CD8+ lymphocytes contribute to the formation of the placenta in the early stages of pregnancy.

The trophoblast cells (including due to the production of the locus molecules HLA-C and HLA-G), T helper lymphocytes, APC of the placenta and decidua, have a significant effect on the formation of the subpopulation composition of CD8+ T lymphocytes of the decidua. In turn, decidual CD8+ T lymphocytes affect trophoblast cells and CD4+ T lymphocytes; however, the molecular mechanisms of these cell-cell interactions have not been sufficiently studied.

Conclusion

The population of CD8+ T lymphocytes is a collection of cells quite diverse in phenotype and function. Great importance is attributed to subpopulations of memory CD8+ T lymphocytes and CD8+ Treg, identified relatively recently. Differentiation of CD8+ T lymphocytes depends on the cytokine environment and antigen presentation involving surface molecules. Not only differentiation is possible in the primary lymphoid organs, but also in the secondary ones; however, the molecular mechanisms for the regulation of differentiation of CD8+ T lymphocytes have not been adequately studied. Migration of CD8+ T lymphocytes to the decidua is caused both by changes in expression of surface molecules of CD8+ T lymphocytes under the action of soluble products in the utero-placental contact zone, and by changes in the production of soluble and surface molecules by cells of the decidua. Molecular mechanisms of specific migration of CD8+ T lymphocytes in pregnancy are poorly understood. An important role of the concentration of CD8+T lymphocytes in the endometrium and decidua during the whole pregnancy is distinguished. Violation of the quantitative content or functional activity of CD8+T lymphocytes in the decidua can lead to serious complications of pregnancy. CD8+ T lymphocytes in the utero-placental contact zone interact with a wide range of cells. Their functioning in these tissues is aimed at stimulating the invasion of trophoblast and the formation of immunological tolerance, accompanied by a decrease in its own cytotoxic activity and an increase in regulatory function. However, the molecular mechanisms of intercellular interactions are subject to further detailed study.

Список литературы / References

1. Aandahl E.M., Torgersen K.M.,Tasken K. CD8+ regulatory T cells - а distinct T-cell lineage or a transient T-cell phenotype? Hum. Immunol., 2008, Vol. 69, no. 11, pp. 696-699.

2. Aboagye-Mathiesen G., Toth F.D., Petersen P.M., Gildberg A., Norskov-Lauritsen N., Zachar V., Ebbesen P. Differential interferon production in human first and third trimester trophoblast cultures stimulated with viruses. Placenta, 1993, Vol. 14, no. 2, pp. 225-234.

3. Aboagye-Mathiesen G., Toth F.D., Zdravkovic M., Ebbesen P. Production of interferons in human placental trophoblast subpopulations and their possible roles in pregnancy. Clin. Diagn. Lab. Immunol., 1994, Vol. 1, no. 6, pp. 650-659.

4. Akoglu B., Lafferton B., Kalb S., Yosuf S.E., Herrmann E., Zeuzem S., Gossmann J., Kachel H.G., Scheuermann E.H., Faust D. Rejection quantity in kidney transplant recipients is associated with increasing intracellular interleukin-2 in CD8+ T-cells. Transpl. Immunol., 2014, Vol. 31, no. 1, pp. 17-21.

5. Arck P.C. Stress and pregnancy loss: role of immune mediators, hormones and neurotransmitters. Am. J. Reprod. Immunol., 2001, Vol. 46, no. 2, pp. 117-123.

6. Arruvito L., Payaslian F., Baz P., Podhorzer A., Billordo A., Pandolfi J., Semeniuk G., Arribalzaga E., Fainboim L. Identification and clinical relevance of naturally occurring human CD8+HLA-DR+ regulatory T cells. J. Immunol., 2014, Vol. 193, no. 9, pp. 4469-4476.

7. Bachmann M.F., Wolint P., Walton S., Schwarz K., Oxenius A. Differential role of IL-2R signaling for CD8+ T cell responses in acute and chronic viral infections. Eur. J. Immunol., 2007, Vol. 37, no. 6, pp. 1502-1512.

8. Badovinac V.P., Porter B.B., Harty J.T. Programmed contraction of CD8(+) T cells after infection. Nat. Immunol., 2002, Vol. 3, no. 7, pp. 619-626

9. Bao Y.S., Wang M., Zhang P., Zhou Z., Zhai W.J., Wang H., Jiang E.L., Huang Y., Feng S.Z., Han M.Z. Regulation of immunological balance between TH1/TH2 and Tc1/Tc2 lymphocytes by prostaglandin E2. Zhongguo Shi Yan Xue Ye Xue Za Zhi, 2010, Vol. 18, no. 2, pp. 431-435.

10. Barinov A., Galgano A., Krenn G., Tanchot C., Vasseur F., Rocha B. CD4/CD8/Dendritic cell complexes in the spleen: CD8+ T cells can directly bind CD4+ T cells and modulate their response. PLoS ONE, 2017, Vol. 12, no. 7, e0180644. doi: 10.1371/journal.pone.0180644.

11. Barnea E.R., Hayrabedyan S., Todorova K., Almogi-Hazan O., Or R., Guingab J., McElhinney J., Fernandez N., Barder T. Prelmplantation factor (PIF*) regulates systemic immunity and targets protective regulatory and cytoskeleton proteins. Immunobiology, 2016, Vol. 221, no. 7, pp. 778-793.

12. Beneventi F., Locatelli E., de Amici M., Martinetti M., Spinillo A. Soluble HLA-G concentrations in obese women during pregnancy and in cord blood. J. Reprod. Immunol., 2017, Vol. 119, pp. 31-37.

13. Blechschmidt K., Mylonas I., Mayr D., Schiessl B., Schulze S., Becker K.F., Jeschke U. Expression of E-cadherin and its repressor snail in placental tissue of normal, preeclamptic and HELLP pregnancies. Virchows Arch., 2007, Vol. 450, no. 2, pp. 195-202.

14. Capece T., Kim M. The role of lymphatic niches in T cell differentiation. Mol. Cells, 2016, Vol. 39, no. 7, pp. 515-523.

15. Carvalheiro H., Duarte C., Silva-Cardoso S., da Silva J.A., Souto-Carneiro M.M. CD8+ T cell profiles in patients with rheumatoid arthritis and their relationship to disease activity. Arthritis Rheumatol., 2015, Vol. 67, no. 2, pp. 363-371.

16. Chandran P.A., Keller A., Weinmann L., Seida A.A., Braun M., Andreev K., Fischer B., Horn E., Schwinn S., Junker M., Houben R., Dombrowski Y., Dietl J., Finotto S., Wolfl M., Meister G., Wischhusen J. The TGF-beta-inducible miR-23a cluster attenuates IFN-gamma levels and antigen-specific cytotoxicity in human CD8(+) T cells. J. Leukoc. Biol., 2014, Vol. 96, no. 4, pp. 633-645.

17. Clark D.A., Chaouat G., Wong K., Gorczynski R.M., Kinsky R. Tolerance mechanisms in pregnancy: a reappraisal of the role of class I paternal MHC antigens. Am. J. Reprod. Immunol., 2010, Vol. 63, no. 2, pp. 93-103.

18. Contini P., Ghio M., Merlo A., Poggi A., Indiveri F., Puppo F. Apoptosis of antigen-specific T lymphocytes upon the engagement of CD8 by soluble HLA class I molecules is Fas ligand/Fas mediated: evidence for the involvement of p56lck, calcium calmodulin kinase II, and Calcium-independent protein kinase C signaling pathways and for NF-kappaB and NF-AT nuclear translocation. J. Immunol., 2005, Vol. 175, no. 11, pp. 7244-7254.

19. Coulomb-L'Hermine A., Larousserie F., Pflanz S., Bardel E., Kastelein R.A., Devergne O. Expression of interleukin-27 by human trophoblas T cells. Placenta, 2007, Vol. 28, no. 11-12, pp. 1133-1140.

20. Crespo A.C., van der Zwan A., Ramalho-Santos J., Strominger J.L., Tilburgs T. Cytotoxic potential of decidual NK cells and CD8+ T cells awakened by infections. J. Reprod. Immunol., 2017, Vol. 119, pp. 85-90.

21. Curtsinger J.M., Agarwal P., Lins D.C., Mescher M.F. Autocrine IFN-gamma promotes naive CD8 T cell differentiation and synergizes with IFN-alpha to stimulate strong function. J. Immunol., 2012, Vol. 189, no. 2, pp. 659-668.

22. Curtsinger J.M., Gerner M.Y., Lins D.C., Mescher M.F. Signal 3 availability limits the CD8 T cell response to a solid tumor. J. Immunol., 2007, Vol. 178, no. 11, pp. 6752-6760.

23. Curtsinger J.M., Mescher M.F. Inflammatory cytokines as a third signal for T cell activation. Curr. Opin. Immunol., 2010, Vol. 22, no. 3, pp. 333-340.

24. Darmochwal-Kolarz D., Sobczak E., Pozarowski P., Kolarz B., Rolinski J., Oleszczuk J. T CD3+CD8+ lymphocytes are more susceptible for apoptosis in the first trimester of normal human pregnancy. J. Immunol. Res., 2014, Vol. 2014, 6705246 9p. doi: 10.1155/2014/670524.

25. Dauven D., Ehrentraut S., Langwisch S., Zenclussen A.C., Schumacher A. Immune modulatory effects of human chorionic gonadotropin on dendritic cells supporting fetal survival in murine pregnancy. Front. Endocrinol. (Lausanne), 2016, Vol. 7, p. 146.

26. Deets K.A., Berkley A.M., Bergsbaken T., Fink P.J. Cutting edge: Enhanced clonal burst size corrects an otherwise defective memory response by CD8+ recent thymic emigrants. J. Immunol., 2016, Vol. 196, no. 6, pp. 2450-2455.

27. Denison F.C., Kelly R.W., Calder A.A., Riley S.C. Cytokine secretion by human fetal membranes, decidua and placenta at term. Hum. Reprod., 1998, Vol. 13, no. 12, pp. 3560-3565.

28. Dong S., Kurtis J.D., Pond-Tor S., Kabyemela E., Duffy P.E., Fried M. CXC ligand 9 response to malaria during pregnancy is associated with low-birth-weight deliveries. Infect. Immun., 2012, Vol. 80, no. 9, pp. 3034-3038.

29. Du M.R., Wang S.C., Li D.J. The integrative roles of chemokines at the maternal-fetal interface in early pregnancy. Cell. Mol. Immunol., 2014, Vol. 11, no. 5, pp. 438-448.

30. Ebina Y., Shimada S., Deguchi M., Maesawa Y., Iijima N.,Yamada H. Divergence of helper, cytotoxic, and regulatory T cells in the decidua from miscarriage. Am. J. Reprod. Immunol., 2016, Vol. 76, no. 3, pp. 199-204.

31. Ellis S.D., McGovern J.L., van Maurik A., Howe D., Ehrenstein M.R., Notley C.A. Induced CD8+FoxP3+ Treg cells in rheumatoid arthritis are modulated by p38 phosphorylation and monocytes expressing membrane tumor necrosis factor alpha and CD86. Arthritis Rheumatol., 2014, Vol. 66, no. 10, pp. 2694-2705.

32. Erden O., Imir A., Guvenal T., Muslehiddinoglu A., Arici S., Cetin M., Cetin A. Investigation of the effects of heparin and low molecular weight heparin on E-cadherin and laminin expression in rat pregnancy by immunohistochemistry. Hum. Reprod., 2006, Vol. 21, no. 11, pp. 3014-3018.

33. Erlebacher A. Immunology of the maternal-fetal interface. Annu. Rev. Immunol., 2013, Vol. 31, pp. 387-411.

34. Erlebacher A. Mechanisms of T cell tolerance towards the allogeneic fetus. Nat. Rev. Immunol., 2013, Vol. 13, no. 1, pp. 23-33.

35. Erlebacher A., Vencato D., Price K.A., Zhang D., Glimcher L.H. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J. Clin. Invest., 2007, Vol. 117, no. 5, pp. 1399-1411.

36. Eusebio M., Kuna P., Kraszula L., Kupczyk M., Pietruczuk M. Allergy-related changes in levels of CD8+CD25+FoxP3(bright) Treg cells and FoxP3 mRNA expression in peripheral blood: the role of IL-10 or TGF-beta. J. Biol. Regul. Homeost. Agents, 2014, Vol. 28, no. 3, pp. 461-470.

37. Fang W.N., Shi M., Meng C.Y., Li D.D., Peng J.P. The Balance between conventional DCs and plasmacytoid DCs is pivotal for immunological tolerance during pregnancy in the mouse. Sci. Rep., 2016, Vol. 6, 26984. doi: 10.1038/srep26984.

38. Gavin M.A., Torgerson T.R., Houston E., DeRoos P., Ho W.Y., Stray-Pedersen A., Ocheltree E.L., Greenberg P.D., Ochs H.D., Rudensky A.Y. Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc. Natl. Acad. Sci. USA, 2006, Vol. 103, no. 17, pp. 6659-6664.

39. Goldberg M.V., Maris C.H., Hipkiss E.L., Flies A.S., Zhen L., Tuder R.M., Grosso J.F., Harris T.J., Getnet D., Whartenby K.A., Brockstedt D.G., Dubensky T.W., Jr., Chen L., Pardoll D.M., Drake C.G. Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells. Blood, 2007, Vol. 110, no. 1, pp. 186-192.

40. Golubovskaya V., Wu L. Different subsets of T cells, memory, effector functions, and CAR-T immuno-therapy. Cancers (Basel), 2016, Vol. 8, no. 3.

41. Gomez-Lopez N., Olson D.M., Robertson S.A. Interleukin-6 controls uterine Th9 cells and CD8(+) T regulatory cells to accelerate parturition in mice. Immunol. Cell Biol., 2016, Vol. 94, no. 1, pp. 79-89.

42. Gringhuis S.I., Kaptein T.M., Wevers B.A., van der Vlist M., Klaver E.J., van Die I., Vriend L.E., de Jong M.A., Geijtenbeek T.B. Fucose-based PAMPs prime dendritic cells for follicular T helper cell polarization via DC-SIGN-dependent IL-27 production. Nat. Commun., 2014, Vol. 5, p. 5074.

43. Gu J., Lu L., Chen M., Xu L., Lan Q., Li Q., Liu Z., Chen G., Wang P., Wang X., Brand D., Olsen N., Zheng S.G. TGF-beta-induced CD4+Foxp3+ T cells attenuate acute graft-versus-host disease by suppressing expansion and killing of effector CD8+ cells. J. Immunol., 2014, Vol. 193, no. 7, pp. 3388-3397.

44. He Y., Chen S., Huang H., Chen Q. Association between decreased plasma levels of soluble human leukocyte antigen-G and severe pre-eclampsia. J. Perinat. Med., 2016, Vol. 44, no. 3, pp. 283-290.

45. Henson S.M., Lanna A., Riddell N.E., Franzese O., Macaulay R., Griffiths S.J., Puleston D.J., Watson A.S., Simon A.K., Tooze S.A., Akbar A.N. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8(+) T cells. J. Clin. Invest, 2014, Vol. 124, no. 9, pp. 4004-4016.

46. Hentschke M.R., Krauspenhar B., Guwzinski A., Caruso F.B., Silveira I.D., Antonello I.C., Gadonski G., Poli-de-Figueiredo C.E., da Costa B.E. PP040. Expression of RANTES (CCL5) in maternal plasma, fetal plasma and placenta in pre-eclampsia and normotensive controls. Pregnancy Hypertens, 2012, Vol. 2, no. 3, p. 263.

47. Holets L.M., Carletti M.Z., Kshirsagar S.K., Christenson L.K., Petroff M.G. Differentiation-induced post-transcriptional control of B7-H1 in human trophoblas T cells. Placenta, 2009, Vol. 30, no. 1, pp. 48-55.

48. Holets L.M., Hunt J.S., Petroff M.G. Trophoblast CD274 (B7-H1) is differentially expressed across gestation: influence of oxygen concentration. Biol. Reprod., 2006, Vol. 74, no. 2, pp. 352-358.

49. Hoyer S., Prommersberger S., Pfeiffer I.A., Schuler-Thurner B., Schuler G., Dorrie J., Schaft N. Concurrent interaction of DCs with CD4(+) and CD8(+) T cells improves secondary CTL expansion: It takes three to tango. Eur. J. Immunol., 2014, Vol. 44, no. 12, pp. 3543-3559.

50. Huang H., Hao S., Li F., Ye Z., Yang J., Xiang J. CD4+ Th1 cells promote CD8+ Tc1 cell survival, memory response, tumor localization and therapy by targeted delivery of interleukin 2 via acquired pMHC I complexes. Immunology, 2007, Vol. 120, no. 2, pp. 148-159.

51. Huang Y., Zhu X.Y., Du M.R., Li D.J. Human trophoblasts recruited T lymphocytes and monocytes into decidua by secretion of chemokine CXCL16 and interaction with CXCR6 in the first-trimester pregnancy. J. Immunol., 2008, Vol. 180, no. 4, pp. 2367-2375.

52. Irtegun S., Tekin M.A., Alpayci R. Increased expression of E-cadherin, endothelin-1, and CD68 in preeclamptic placentas. Erciyes Medical Journal, 2016, Vol. 38, no. 4, pp. 149-152.

53. Ishii M., Hayakawa S., Suzuki M.K., Yoshino N., Honda M., Nishinarita S., Chishima F., Nagaishi M., Satoh K. Expression of functional chemokine receptors of human placental cells. Am. J. Reprod. Immunol., 2000, Vol. 44, no. 6, pp. 365-373.

54. Jebbawi F., Fayyad-Kazan H., Merimi M., Lewalle P., Verougstraete J.C., Leo O., Romero P., Burny A., Badran B., Martiat P., Rouas R. A microRNA profile of human CD8(+) regulatory T cells and characterization of the effects of microRNAs on Treg cell-associated genes. J. Transl. Med., 2014, Vol. 12, p. 218.

55. Jenkinson S.E., Whawell S.A., Swales B.M., Corps E.M., Kilshaw P.J., Farthing P.M. The alphaE(CD103)beta7 integrin interacts with oral and skin keratinocytes in an E-cadherin-independent manner*. Immunology, 2011, Vol. 132, no. 2, pp. 188-196.

56. Jin H.T., Anderson A.C., Tan W.G., West E.E., Ha S.J., Araki K., Freeman G.J., Kuchroo V.K., Ahmed R. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc. Natl. Acad. Sci. USA, 2010, Vol. 107, no. 33, pp. 14733-14738.

57. Joedicke J.J., Myers L., Carmody A.B., Messer R.J., Wajant H., Lang K.S., Lang P.A., Mak T.W., Hasenkrug K.J., Dittmer U. Activated CD8+ T cells induce expansion of Vbeta5+ regulatory T cells via TNFR2 signaling. J. Immunol., 2014, Vol. 193, no. 6, pp. 2952-2960.

58. Juch H., Blaschitz A., Dohr G., Hutter H. HLA class I expression in the human placenta. Wien. Med. Wochenschr., 2012, Vol. 162, no. 9-10, pp. 196-200.

59. Kaech S.M., Tan J.T., Wherry E.J., Konieczny B.T., Surh C.D., Ahmed R. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol., 2003, Vol. 4, no. 12, pp. 1191-1198.

60. Kalble F., Mai C., Wagner M., Schober L., Schaier M., Zeier M., Spratte J., Fluhr H., Steinborn A. Aberrant ICOS+ T cell differentiation in women with spontaneous preterm labor. Am. J. Reprod. Immunol., 2016, Vol. 76, no. 5, pp. 415-425.

61. Karjalainen M.K., Ojaniemi M., Haapalainen A.M., Mahlman M., Salminen A., Huusko J.M., Maatta T.A., Kaukola T., Anttonen J., Ulvila J., Haataja R., Teramo K., Kingsmore S.F., Palotie A., Muglia L.J., Ramet M., Hallman M. CXCR3 polymorphism and expression associate with spontaneous preterm birth. J. Immunol., 2015, Vol. 195, no. 5, pp. 2187-2198.

62. Kastenmuller W., Brandes M., Wang Z., Herz J., Egen J.G., Germain R.N. Peripheral prepositioning and local CXCL9 chemokine-mediated guidance orchestrate rapid memory CD8+ T cell responses in the lymph node. Immunity, 2013, Vol. 38, no. 3, pp. 502-513.

63. Kim C.J., Romero R., Chaemsaithong P., Kim J.S. Chronic inflammation of the placenta: definition, classification, pathogenesis, and clinical significance. Am. J. Obstet. Gynecol., 2015, Vol. 213, no. 4, pp. S53-69.

64. Kim C.J., Romero R., Kusanovic J.P., Yoo W., Dong Z., Topping V., Gotsch F., Yoon B.H., Chi J.G., Kim J.S. The frequency, clinical significance, and pathological features of chronic chorioamnionitis: a lesion associated with spontaneous preterm birth. Mod. Pathol., 2010, Vol. 23, no. 7, pp. 1000-1011.

65. Kim M.T., Harty J.T. Impact of inflammatory cytokines on effector and memory CD8+ T cells. Front. Immunol., 2014, Vol. 5, p. 295.

66. Klitkou L., Dahl M., Hviid T.V., Djurisic S., Piosik Z.M., Skovbo P., Moller A.M., Steffensen R., Christiansen O.B. Human leukocyte antigen (HLA)-G during pregnancy part I: correlations between maternal soluble HLA-G at midterm, at term, and umbilical cord blood soluble HLA-G at term. Hum. Immunol., 2015, Vol. 76, no. 4, pp. 254-259.

67. Kondo T., Takata H., Matsuki F., Takiguchi M. Cutting edge: Phenotypic characterization and differentiation of human CD8+ T cells producing IL-17. J. Immunol., 2009, Vol. 182, no. 4, pp. 1794-1798.

68. Kuttruff S., Koch S., Kelp A., Pawelec G., Rammensee H.G., Steinle A. NKp80 defines and stimulates a reactive subset of CD8 T cells. Blood, 2009, Vol. 113, no. 2, pp. 358-369.

69. Laidlaw B.J., Craft J.E., Kaech S.M. The multifaceted role of CD4(+) T cells in CD8(+) T cell memory. Nat. Rev. Immunol., 2016, Vol. 16, no. 2, pp. 102-111.

70. Laidlaw B.J., Cui W., Amezquita R.A., Gray S.M., Guan T., Lu Y., Kobayashi Y., Flavell R.A., Kleinstein S.H., Craft J., Kaech S.M. Production of IL-10 by CD4(+) regulatory T cells during the resolution of infection promotes the maturation of memory CD8(+) T cells. Nat. Immunol., 2015, Vol. 16, no. 8, pp. 871-879.

71. le Bouteiller P. HLAG in human early pregnancy: Control of uterine immune cell activation and likely vascular remodeling. Biomed. J., 2015, no. 38, p. 3238.

72. le Bouteiller P., Pizzato N., Barakonyi A., Solier C. HLA-G, pre-eclampsia, immunity and vascular events. J. Reprod. Immunol., 2003, Vol. 59, no. 2, pp. 219-234.

73. Lee N., You S., Shin M.S., Lee W.W., Kang K.S., Kim S.H., Kim W.U., Homer R.J., Kang M.J., Montgomery R.R., Dela Cruz C.S., Shaw A.C., Lee P.J., Chupp G.L., Hwang D., Kang I. IL-6 receptor alpha defines effector memory CD8+ T cells producing Th2 cytokines and expanding in asthma. Am. J. Respir. Crit. Care Med., 2014, Vol. 190, no. 12, pp. 1383-1394.

74. Lerret N.M., Houlihan J.L., Kheradmand T., Pothoven K.L., Zhang Z.J., Luo X. Donor-specific CD8+ Foxp3+ T cells protect skin allografts and facilitate induction of conventional CD4+ Foxp3+ regulatory T cells. Am. J. Transplant., 2012, Vol. 12, no. 9, pp. 2335-2347.

75. Li C., Lin Y., Gao C., Qi Z. Function of CD4(+) T cells in CD8(+) T cell mediated rejection. Zhonghua Yi Xue Za Zhi, 2014, Vol. 94, no. 34, pp. 2690-2694.

76. Li H., Wang W., Wang G., Hou Y., Xu F., Liu R., Wang F., Xue J., Hu T., Luan X. Interferon-gamma and tumor necrosis factor-alpha promote the ability of human placenta-derived mesenchymal stromal cells to express programmed death ligand-2 and induce the differentiation of CD4(+)interleukin-10(+) and CD8(+) interleukin-10(+)Treg subsets. Cytotherapy, 2015, Vol. 17, no. 11, pp. 1560-1571.

77. Li H.W., Cheung A.N., Tsao S.W., Cheung A.L., O W.S. Expression of e-cadherin and beta-catenin in trophoblastic tissue in normal and pathological pregnancies. Int. J. Gynecol. Pathol., 2003, Vol. 22, no. 1, pp. 63-70.

78. Li M., Piao L., Chen C.P., Wu X., Yeh C.C., Masch R., Chang C.C., Huang S.J. Modulation of decidual macrophage polarization by macrophage colony-stimulating factor derived from first-trimester decidual cells: implication in preeclampsia. Am. J. Pathol., 2016, Vol. 186, no. 5, pp. 1258-1266.

79. Li M., Sun X., Kuang X., Liao Y., Li H., Luo D. Mesenchymal stem cells suppress CD8+ T cell-mediated activation by suppressing natural killer group 2, member D protein receptor expression and secretion of prostaglandin E2, indoleamine 2,3-dioxygenase and transforming growth factor-beta. Clin. Exp. Immunol., 2014, Vol. 178, no. 3, pp. 516-524.

80. Li X.L., Dong X., Xue Y., Li C.F., Gou W.L., Chen Q. Increased expression levels of E-cadherin, cytokeratin 18 and 19 observed in preeclampsia were not correlated with disease severity. Placenta, 2014, Vol. 35, no. 8, pp. 625-631.

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

81. Liu D., Burd E.M., Coopersmith C.M., Ford M.L. Retrogenic ICOS expression increases differentiation of KLRG-1hiCD127loCD8+ T cells during listeria infection and diminishes recall responses. J. Immunol., 2016, Vol. 196, no. 3, pp. 1000-1012.

82. Liu S.J., Tsai J.P., Shen C.R., Sher Y.P., Hsieh C.L., Yeh Y.C., Chou A.H., Chang S.R., Hsiao K.N., Yu F.W., Chen H.W. Induction of a distinct CD8 Tnc17 subset by transforming growth factor-beta and interleukin-6. J. Leukoc. Biol., 2007, Vol. 82, no. 2, pp. 354-360.

83. Liu Y., Lan Q., Lu L., Chen M., Xia Z., Ma J., Wang J., Fan H., Shen Y., Ryffel B., Brand D., Quismorio F., Liu Z., Horwitz D.A., Xu A., Zheng S.G. Phenotypic and functional characteristic of a newly identified CD8+ Foxp3-CD103+ regulatory T cells. J. Mol. Cell. Biol., 2014, Vol. 6, no. 1, pp. 81-92.

84. Marozio L., Garofalo A., Berchialla P., Tavella A.M., Salton L., Cavallo F., Benedetto C. Low expression of soluble human leukocyte antigen G in early gestation and subsequent placenta-mediated complications of pregnancy. J. Obstet. Gynaecol. Res., 2017, Vol. 43, no. 9, pp. 1391-1396.

85. Mas A.E., Petitbarat M., Dubanchet S., Fay S., Ledee N., Chaouat G. Immune regulation at the interface during early steps of murine implantation: involvement of two new cytokines of the IL-12 family (IL-23 and IL-27) and of TWEAK. Am. J. Reprod. Immunol., 2008, Vol. 59, no. 4, pp. 323-338.

86. McLane L.M., Banerjee P.P., Cosma G.L., Makedonas G., Wherry E.J., Orange J.S., Betts M.R. Differential localization of T-bet and Eomes in CD8 T cell memory populations. J. Immunol., 2013, Vol. 190, no. 7, pp. 3207-3215.

87. Mescher M.F., Curtsinger J.M., Agarwal P., Casey K.A., Gerner M., Hammerbeck C.D., Popescu F., Xiao Z. Signals required for programming effector and memory development by CD8+ T cells. Immunol. Rev., 2006, Vol. 211, pp. 81-92.

88. Michimata T., Tsuda H., Sakai M., Fujimura M., Nagata K., Nakamura M., Saito S. Accumulation of CRTH2-positive T-helper 2 and T-cytotoxic 2 cells at implantation sites of human decidua in a prostaglandin D(2)-mediated manner. Mol. Hum. Reprod., 2002, Vol. 8, no. 2, pp. 181-187.

89. Mosmann T.R., Li L., Sad S. Functions of CD8 T-cell subsets secreting different cytokine patterns. Semin. Immunol., 1997, Vol. 9, no. 2, pp. 87-92.

90. Nagamatsu T., Barrier B.F., Schust D.J. The regulation of T-cell cytokine production by ICOS-B7H2 interactions at the human fetomaternal interface. Immunol. Cell Biol., 2011, Vol. 89, no. 3, pp. 417-425.

91. Nakagawa T., Tsuruoka M., Ogura H., Okuyama Y., Arima Y., Hirano T., Murakami M. IL-6 positively regulates Foxp3+CD8+ T cells in vivo. Int. Immunol., 2010, Vol. 22, no. 2, pp. 129-139.

92. Nancy P., Erlebacher A. T cell behavior at the maternal-fetal interface. Int. J. Dev. Biol., 2014, Vol. 58, no. 2-4, pp. 189-198.

93. Novais F.O., Carvalho A.M., Clark M.L., Carvalho L.P., Beiting D.P., Brodsky I.E., Carvalho E.M., Scott P. CD8+ T cell cytotoxicity mediates pathology in the skin by inflammasome activation and IL-1beta production. PLoS Pathog., 2017, Vol. 13, no. 2, e1006196. doi: 10.1371/journal.ppat.1006196.

94. O'Sullivan S., Cormican L., Faul J.L., Ichinohe S., Johnston S.L., Burke C.M., Poulter L.W. Activated, cytotoxic CD8(+) T lymphocytes contribute to the pathology of asthma death. Am. J. Respir. Crit. Care Med., 2001, Vol. 164, no. 4, pp. 560-564.

95. Okada S., Okada H., Sanezumi M., Nakajima T., Yasuda K., Kanzaki H. Expression of interleukin-15 in human endometrium and decidua. Mol. Hum. Reprod., 2000, Vol. 6, no. 1, pp. 75-80.

96. Osman I., Young A., Ledingham M.A., Thomson A.J., Jordan F., Greer I.A., Norman J.E. Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol. Hum. Reprod., 2003, Vol. 9, no. 1, pp. 41-45.

97. Ouellette M.J., St-Jacques S., Lambert R.D. CD8 membrane expression is down-regulated by transforming growth factor (TGF)-beta 1, TGF-beta 2, and prostaglandin E2. Am. J. Reprod. Immunol., 1999, Vol. 41, no. 3, pp. 183-191.

98. Paparini D., Gori S., Grasso E., Scordo W., Calo G., Perez Leiros C., Ramhorst R., Salamone G. Acetylcholine contributes to control the physiological inflammatory response during the peri-implantation period. Acta Physiol. (Oxf.), 2015, Vol. 214, no. 2, pp. 237-247.

99. Penaloza-MacMaster P. CD8 T-cell regulation by T regulatory cells and the programmed cell death protein 1 pathway. Immunology, 2017, Vol. 151, no. 2, pp. 146-153.

100. Peterson S.E., Nelson J.L., Gadi V.K., Gammill H.S. Fetal cellular microchimerism in miscarriage and pregnancy termination. Chimerism, 2013, Vol. 4, no. 4, pp. 136-138.

101. Pitman H., Innes B.A., Robson S.C., Bulmer J.N., Lash G.E. Altered expression of interleukin-6, interleukin-8 and their receptors in decidua of women with sporadic miscarriage. Hum. Reprod., 2013, Vol. 28, no. 8, pp. 2075-2086.

102. Prins J.R., van der Hoorn M.L., Keijser R., Ris-Stalpers C., van Beelen E., Afink G.B., Claas F.H., van der Post J.A., Scherjon S.A. Higher decidual EBI3 and HLA-G mRNA expression in preeclampsia: Cause or consequence of preeclampsia. Hum. Immunol., 2016, Vol. 77, no. 1, pp. 68-70.

103. Puppo F., Contini P., Ghio M., Brenci S., Scudeletti M., Filaci G., Ferrone S., Indiveri F. Soluble human MHC class I molecules induce soluble Fas ligand secretion and trigger apoptosis in activated CD8(+) Fas (CD95)(+) T lymphocytes. Int. Immunol., 2000, Vol. 12, no. 2, pp. 195-203.

104. Raman K., Wang H., Troncone M.J., Khan W.I., Pare G., Terry J. Overlap chronic placental inflammation is associated with a unique gene expression pattern. PLoS ONE, 2015, Vol. 10, no. 7, e0133738. doi: 10.1371/journal. pone.0133738.

105. Reeves E., James E. Tumour and placenta establishment: The importance of antigen processing and presentation. Placenta, 2017, Vol. 56, pp. 34-39.

106. Remoli M.E., Gafa V., Giacomini E., Severa M., Lande R., Coccia E.M. IFN-beta modulates the response to TLR stimulation in human DC: involvement of IFN regulatory factor-1 (IRF-1) in IL-27 gene expression. Eur. J. Immunol., 2007, Vol. 37, no. 12, pp. 3499-3508.

107. Rijnink E.C., Penning M.E., Wolterbeek R., Wilhelmus S., Zandbergen M., van Duinen S.G., Schutte J., Bruijn J.A., Bajema I.M. Tissue microchimerism is increased during pregnancy: a human autopsy study. Mol. Hum. Reprod., 2015, Vol. 21, no. 11, pp. 857-864.

108. Ryu S.J., Jung K.M., Yoo H.S., Kim T.W., Kim S., Chang J., Choi E.Y. Cognate CD4 help is essential for the reactivation and expansion of CD8 memory T cells directed against the hematopoietic cell-specific dominant minor histocompatibility antigen, H60. Blood, 2009, Vol. 113, no. 18, pp. 4273-4280.

109. Sad S., Li L., Mosmann T.R. Cytokine-deficient CD8+ Tc1 cells induced by IL-4: retained inflammation and perforin and Fas cytotoxicity but compromised long term killing of tumor cells. J. Immunol., 1997, Vol. 159, no. 2, pp. 606-613.

110. Sad S., Mosmann T.R. Interleukin (IL) 4, in the absence of antigen stimulation, induces an anergy-like state in differentiated CD8+ TC1 cells: loss of IL-2 synthesis and autonomous proliferation but retention of cytotoxicity and synthesis of other cytokines. J. Exp. Med., 1995, Vol. 182, no. 5, pp. 1505-1515.

111. San Segundo D., Ballesteros M.A., Naranjo S., Zurbano F., Minambres E., Lopez-Hoyos M. Increased numbers of circulating CD8 effector memory T cells before transplantation enhance the risk of acute rejection in lung transplant recipients. PLoS ONE, 2013, Vol. 8, no. 11, e80601. doi: 10.1371/journal.pone.0080601.

112. Sandau M.M., Kohlmeier J.E., Woodland D.L., Jameson S.C. IL-15 regulates both quantitative and qualitative features of the memory CD8 T cell pool. J. Immunol., 2010, Vol. 184, no. 1, pp. 35-44.

113. Scaife P.J., Bulmer J.N., Robson S.C., Innes B.A., Searle R.F. Effector activity of decidual CD8+ T lymphocytes in early human pregnancy. Biol. Reprod., 2006, Vol. 75, no. 4, pp. 562-567.

114. Schettini J.A.C., Gomes T.V., Santos Barreto A.K., da Silva Junior C.D., da Matta M., Coutinho I.C.N., de Oliveira M., Torres L.C. High Levels of CXCL8 and low levels of CXCL9 and CXCL10 in women with maternal RhD alloimmunization. Front. Immunol., 2017, Vol. 8, p. 700.

115. Schliefsteiner C., Peinhaupt M., Kopp S., Logl J., Lang-Olip I., Hiden U., Heinemann A., Desoye G., Wadsack C. Human placental hofbauer cells maintain an anti-inflammatory M2 phenotype despite the presence of gestational diabetes mellitus. Front. Immunol., 2017, Vol. 8, p. 888.

116. Schneider R., Yaneva T., Beauseigle D., El-Khoury L., Arbour N. IL-27 increases the proliferation and effector functions of human naïve CD8+ T lymphocytes and promotes their development into Tc1 cells. Eur. J. Immunol., 2011, Vol. 41, no. 1, pp. 47-59.

117. Schumacher A., Costa S.D., Zenclussen A.C. Endocrine factors modulating immune responses in pregnancy. Front. Immunol., 2014, Vol. 5, p. 196.

118. Shao L., Jacobs A.R., Johnson V.V., Mayer L. Activation of CD8+ regulatory T cells by human placental trophoblasts. J. Immunol., 2005, Vol. 174, no. 12, pp. 7539-7547.

119. Shobeiri S.S., Abediankenari S., Lashtoo-Aghaee B., Rahmani Z., Esmaeili-Gorji B. Evaluation of soluble human leukocyte antigen-G in peripheral blood of pregnant women with gestational diabetes mellitus. Caspian J. Intern. Med., 2016, Vol. 7, no. 3, pp. 178-182.

120. Sokolov D.I., Stepanova O.I., Selkov S.I. The role of the different subpopulations of CD4+T lymphocytes during pregnancy. Medical Immunology (Russia), 2016, Vol. 18, no. 6, pp. 521-536.

121. Soni C., Karande A.A. Glycodelin-A interferes with IL-2/IL-2R signalling to induce cell growth arrest, loss of effector functions and apoptosis in T-lymphocytes. Hum. Reprod., 2012, Vol. 27, no. 4, pp. 1005-1015.

122. Soni C., Karande A.A. Glycodelin A suppresses the cytolytic activity of CD8+ T lymphocytes. Mol. Immunol., 2010, Vol. 47, no. 15, pp. 2458-2466.

123. Steinert E.M., Schenkel J.M., Fraser K.A., Beura L.K., Manlove L.S., Igyarto B.Z., Southern P.J., Masopust D. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell, 2015, Vol. 161, no. 4, pp. 737-749

124. Sundar Raj S., Soni C., Karande A.A. Glycodelin A triggers T cell apoptosis through a novel calcium-independent galactose-binding lectin activity. Mol. Immunol., 2009, Vol. 46, no. 16, pp. 3411-3419.

125. Swanson P.A. 2nd, Hart G.T., Russo M.V., Nayak D., Yazew T., Pena M., Khan S.M., Janse C.J., Pierce S.K., McGavern D.B. CD8+ T cells induce fatal brainstem pathology during cerebral malaria via luminal antigen-specific engagement of brain vasculature. PLoS Pathog., 2016, Vol. 12, no. 12, e1006022. doi: 10.1371/journal.ppat.1006022.

126. Thomas D.A., Massague J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell, 2005, Vol. 8, no. 5, pp. 369-380.

127. Thompson A.W., Lotze M.T. The cytokine handbook. Gulf Professional Publishing, 2003. 1572 p.

128. Tilburgs T., Scherjon S.A., Roelen D.L., Claas F.H. Decidual CD8+CD28- T cells express CD103 but not perforin. Hum. Immunol., 2009, Vol. 70, no. 2, pp. 96-100.

129. Tilburgs T., Schonkeren D., Eikmans M., Nagtzaam N.M., Datema G., Swings G.M., Prins F., van Lith J.M., van der Mast B.J., Roelen D.L., Scherjon S.A., Claas F.H. Human decidual tissue contains differentiated CD8+ effector-memory T cells with unique properties. J. Immunol., 2010, Vol. 185, no. 7, pp. 4470-4477.

130. Tilburgs T., Strominger J.L. CD8+ effector T cells at the fetal-maternal interface, balancing fetal tolerance and antiviral immunity. Am. J. Reprod. Immunol., 2013, Vol. 69, no. 4, pp. 395-407.

131. Tilburgs T., van der Mast B.J., Nagtzaam N.M., Roelen D.L., Scherjon S.A., Claas F.H. Expression of NK cell receptors on decidual T cells in human pregnancy. J. Reprod. Immunol., 2009, Vol. 80, no. 1-2, pp. 22-32.

132. Tsunoda I., Kuang L.Q., Kobayashi-Warren M., Fujinami R.S. Central nervous system pathology caused by autoreactive CD8+ T-cell clones following virus infection. J. Virol., 2005, Vol. 79, no. 23, pp. 14640-14646.

133. van Egmond A., van der Keur C., Swings G.M., Scherjon S.A., Claas F.H. The possible role of virus-specific CD8(+) memory T cells in decidual tissue. J. Reprod. Immunol., 2016, Vol. 113, pp. 1-8.

134. van Panhuys N. Studying dendritic cell - T cell interactions under in vivo conditions. Methods Mol. Biol., 2017, Vol. 1584, pp. 569-583.

135. Vasquez J.C., Huttner A., Zhang L., Marks A., Chan A., Baehring J.M., Kahle K.T., Dhodapkar K.M. SOX2 immunity and tissue resident memory in children and young adults with glioma. J. Neurooncol., 2017, Vol. 134, no. 1, pp. 41-53.

136. Veras E., Kurman R.J., Wang T.L., Shih I.M. PD-L1 Expression in human placentas and gestational trophoblastic diseases. Int. J. Gynecol. Pathol., 2017, Vol. 36, no. 2, pp. 146-153.

137. Vukmanovic- Stejic M., Vyas B., Gorak-Stolinska P., Noble A., Kemeny D.M. Human Tc1 and Tc2/Tc0 CD8 T-cell clones display distincT cell surface and functional phenotypes. Blood, 2000, Vol. 95, no. 1, pp. 231-240.

138. Wang H., He M., Hou Y., Chen S., Zhang X., Zhang M., Ji X. Role of decidual CD14(+) macrophages in the homeostasis of maternal-fetal interface and the differentiation capacity of the cells during pregnancy and parturition. Placenta, 2016, Vol. 38, pp. 76-83.

139. Wang S.C., Li Y.H., Piao H.L., Hong X.W., Zhang D., Xu Y.Y., Tao Y., Wang Y., Yuan M.M., Li D.J., Du M.R. PD-1 and Tim-3 pathways are associated with regulatory CD8+ T-cell function in decidua and maintenance of normal pregnancy. Cell Death Dis., 2015, Vol. 6, e1738. doi: 10.1038/cddis.2015.112.

140. Wang W.J., Liu F.J., Qu H.M., Hao C.F., Qu Q.L., Xiong W., Bao H.C., Wang X.R. Regulation of the expression of Th17 cells and regulatory T cells by IL-27 in patients with unexplained early recurrent miscarriage. J. Reprod. Immunol., 2013, Vol. 99, no. 1-2, pp. 39-45.

141. White H.D., Crassi K.M., Givan A.L., Stern J.E., Gonzalez J.L., Memoli V.A., Green W.R., Wira C.R. CD3+ CD8+ CTL activity within the human female reproductive tract: influence of stage of the menstrual cycle and menopause. J. Immunol., 1997, Vol. 158, no. 6, pp. 3017-3027.

142. Wikenheiser D.J., Stumhofer J.S. ICOS co-stimulation: friend or foe? Front. Immunol., 2016, Vol. 7, p. 304.

143. Wilczynski J.R., Kalinka J., Radwan M. The role of T-regulatory cells in pregnancy and cancer. Front. Biosci., 2008, Vol. 13, pp. 2275-2289.

144. Williams P.J., Searle R.F., Robson S.C., Innes B.A., Bulmer J.N. Decidual leucocyte populations in early to late gestation normal human pregnancy. J. Reprod. Immunol., 2009, Vol. 82, no. 1, pp. 24-31.

145. Wu Q., Gardiner G.J., Berry E., Wagner S.R., Lu T., Clay B.S., Moore T.V., Ferreira C.M., Williams J.W., Luster A.D., Medoff B.D., Cannon J.L., Sperling A.I., Shilling R.A. ICOS-expressing lymphocytes promote

resolution of CD8-mediated lung injury in a mouse model of lung rejection. PLoS ONE, 2013, Vol. 8, no. 8, e72955. doi: 10.1371/journal.pone.0072955.

146. Xiao Z., Casey K.A., Jameson S.C., Curtsinger J.M., Mescher M.F. Programming for CD8 T cell memory development requires IL-12 or type I IFN. J. Immunol., 2009, Vol. 182, no. 5, pp. 2786-2794.

147. Xin L., Ertelt J.M., Rowe J.H., Jiang T.T., Kinder J.M., Chaturvedi V., Elahi S., Way S.S. Cutting edge: committed Th1 CD4+ T cell differentiation blocks pregnancy-induced Foxp3 expression with antigen-specific fetal loss. J. Immunol., 2014, Vol. 192, no. 7, pp. 2970-2974.

148. Xu Y.Y., Wang S.C., Lin Y.K., Li D.J., Du M.R. Tim-3 and PD-1 regulate CD8+ T cell function to maintain early pregnancy in mice. J. Reprod. Dev., 2017, Vol. 63, no. 3, pp. 289-294.

149. Yang J., Zhang P., Krishna S., Wang J., Lin X., Huang H., Xie D., Gorentla B., Huang R., Gao J., Li Q.J., Zhong X.P. Unexpected positive control of NFkappaB and miR-155 by DGKalpha and zeta ensures effector and memory CD8+ T cell differentiation. Oncotarget, 2016, Vol. 7, no. 23, pp. 33744-33764.

150. Yap M., Boeffard F., Clave E., Pallier A., Danger R., Giral M., Dantal J., Foucher Y., Guillot-Gueguen C., Toubert A., Soulillou J.P., Brouard S., Degauque N. Expansion of highly differentiated cytotoxic terminally differentiated effector memory CD8+ T cells in a subset of clinically stable kidney transplant recipients: a potential marker for late graft dysfunction. J. Am. Soc. Nephrol., 2014, Vol. 25, no. 8, pp. 1856-1868.

151. Yu J., Qian L., Wu F., Li M., Chen W., Wang H. Decreased frequency of peripheral blood CD8+CD25+FoxP3+regulatory T cells correlates with IL-33 levels in pre-eclampsia. Hypertens Pregnancy, 2017, Vol. 36, no. 2, pp. 217-225.

152. Yu X.Z., Liang Y., Nurieva R.I., Guo F., Anasetti C., Dong C. Opposing effects of ICOS on graft-versus-host disease mediated by CD4 and CD8 T cells. J. Immunol, 2006, Vol. 176, no. 12, pp. 7394-7401.

153. Zhang X.H., Liang X., Liang X.H., Wang T.S., Qi Q.R., Deng W.B., Sha A.G., Yang Z.M. The mesenchymal-epithelial transition during in vitro decidualization. Reprod. Sci., 2013, Vol. 20, no. 4, pp. 354-360.

Авторы:

Степанова О.И. — к.б.н., старший научный сотрудник лаборатории межклеточных взаимодействий, отдел иммунологии и межклеточных взаимодействий ФГБНУ«Научно-исследовательский институт акушерства, гинекологии и репродуктологии имени Д.О. Отта», Санкт-Петербург, Россия Баженов Д.О. — младший научный сотрудник лаборатории межклеточных взаимодействий, отдел иммунологии и межклеточных взаимодействий ФГБНУ «Научно-исследовательский институт акушерства, гинекологии и репродуктологии имени Д.О. Отта», Санкт-Петербург, Россия Хохлова Е.В. — лаборант-исследователь лаборатории межклеточных взаимодействий, отдел иммунологии и межклеточных взаимодействий ФГБНУ «Научно-исследовательский институт акушерства, гинекологии и репродуктологии имени Д.О. Отта», Санкт-Петербург, Россия

Коган И.Ю. — д.м.н., член-корр. РАН, профессор, врио директора ФГБНУ «Научно-исследовательский институт акушерства, гинекологии и репродуктологии имени Д.О. Отта», Санкт-Петербург, Россия Соколов Д.И. — д.б.н., заведующий лабораторией межклеточных взаимодействий, отдел иммунологии и межклеточных взаимодействий ФГБНУ «Научно-исследовательский институт акушерства, гинекологии и репродуктологии имени Д.О. Отта»; ГБОУВПО «Первый Санкт-Петербургский государственный медицинский университет имени академика И.П. Павлова» Министерства здравоохранения РФ, кафедра иммунологии, Санкт-Петербург, Россия Сельков С.А. — д.м.н., профессор, руководитель отдела иммунологии и межклеточных взаимодействий ФГБНУ «Научно-исследовательский институт акушерства, гинекологии и репродуктологии имени Д.О. Отта», Санкт-Петербург, Россия

Поступила 30.11.2017 Принята к печати 04.12.2017

Authors:

Stepanova O.I., PhD (Biology), Senior Research Associate, Cell Interactions Laboratory, Department of Immunology and Cell Interactions, D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation

Bazhenov D.O., Junior Research Associate, Cell Interactions Laboratory, Department of Immunology and Cell Interactions, D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation

Khokhlova E.V., Research Assistant, Cell Interactions Laboratory, Department of Immunology and Cell Interactions, D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation

Kogan I.Yu., PhD, MD (Medicine), Corresponding Member, Russian Academy of Sciences, Professor, Acting Director, D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation Sokolov D.I., PhD, MD (Biology), Head, Cell Interactions Laboratory, Department of Immunology and Cell Interactions, D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation; First St. Petersburg State I. Pavlov Medical University, Department of Immunology, St. Petersburg, Russian Federation

Selkov S.A., PhD, MD (Medicine), Professor, Head, Department of Immunology and Cell Interactions, D. Ott Research Institute of Obstetrics, Ginecology and Reproductology, St. Petersburg, Russian Federation

Received 30.11.2017 Accepted 04.12.2017

i Надоели баннеры? Вы всегда можете отключить рекламу.