hormonas sexuales y respuesta inmune

Upload: li-li-b-lula-greco

Post on 03-Apr-2018

221 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    1/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    The sexual dimorphism in immune responses in humans is well known; females have more

    vigorous cellular and humoral immune responses, they are more resistant to many infections,

    and they suffer a higher incidence of autoimmune diseases as compared with males [reviewed

    in Reference 1].

    Moreover, in women disease expression appears to be affected by their reproductive status.

    Patients with immune-based diseases, such as multiple sclerosis (MS), asthma or systemic

    lupus erythematosus (SLE), may have exacerbations during specific periods of the menstrualcycle or during pregnancy [2-4]. The differences in immune responses between the sexes and

    the reproductive phases in women are accompanied by variations in sex hormones. Therefore,

    these variations in levels of sex hormones have been suggested to cause the different immune

    responses. The actions of sex hormones, however, are extremely complex. In this overview, we

    will therefore discuss what is known about the effects of sex hormones on the function of the

    different immune cells in isolation. Moreover, we will also discuss briefly how sex hormones

    affect immune responses in complex situations like models of immune-mediated diseases

    related to specific and non-specific immune responses.

    1 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    2/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    Sex hormones

    Three classes of sex hormones exist: androgens (mainly testosterone), estrogens (which ismainly 17beta-estradiol in the ovarian cycle), and progesterone. In males, plasma testosterone

    concentrations are relatively stable throughout life, although testosterone production declines

    with age. In females, the production of sex hormones 17beta-estradiol and progesterone

    fluctuates during the menstrual cycle. In response to pituitary luteinizing hormone (LH) and

    follicle stimulating hormone (FSH), fluctuations in sex hormone concentrations in the menstrual

    cycle include increasing 17beta-estradiol, but low progesterone plasma concentrations in the

    follicular phase and high plasma 17beta-estradiol and progesterone concentrations in the luteal

    phase (see also Fig. 1).

    If pregnancy occurs, luteolysis is prevented and 17beta-estradiol and progesterone levels

    remain high. When the ovarian follicles are depleted later in female life (after menopause) sex

    hormone concentrations drop to low levels. Hormone replacement therapy (HRT) and the use of

    oral contraceptives (OCC) increase (synthetic) estrogen and progesterone concentrations.

    Immune system

    There are two arms of the immune system: the nonspecific (innate or natural) immune system

    and the specific (acquired or adaptive) immune system. The nonspecific immune response is

    the first line of defense against infections. It recognizes structures specific for microbes. The

    effector cells of the nonspecific immune response are monocytes, macrophages, granulocytes

    (neutrophils, eosinophils and basophils), dendritic cells and natural killer (NK) cells. These cellsattack microbes that have entered the body. They do so by phagocytosing the microbe

    2 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    3/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    (neutrophils, monocytes and macrophages), by lysis of infected cells (NK cells), or by producing

    cytokines to enhance nonspecific immune and specific immune responses (all cells). Dendritic

    cells are (together with monocytes and macrophages) the most important antigen presenting

    cells. They take up foreign antigens (including viruses or pathogens), process the antigens,

    present antigen peptides in the context of MHC II molecules on their surface and present themto the specific immune system mainly helper T lymphocytes.

    T lymphocytes and B lymphocytes are the cellular components of the specific immune

    response. Within the T lymphocyte population, cytotoxic T lymphocytes (Tc cells) can directly

    kill foreign or infected cells. The helper T lymphocytes (Th cells) provide help to other immune

    cells by producing cytokines. These Th cells can be divided into 5 subsets, i.e. the Th1 subset

    producing interferon (IFN) gamma to promote cellular immune responses, the Th2 subset

    producing mainly interleukin (IL)-4, IL-13 and IL-5 to provide optimal help for humoral immuneresponses, the Th17 subset producing IL-17, which plays a crucial role in autoimmunity and

    allergen-specific immune responses and the regulatory T cell (Treg) subset that is in the centre

    of immunoregulation and is capable of suppressing both Th1- and Th2-mediated specific

    immune responses. A Th9 subset has also been described. This population has only been

    studied in vitro and it is unclear whether this is a real subset or an adapted Th2 population [5].

    Receptors for sex hormones on immune cells

    Sex hormones are steroid hormones. Due to their lipophilic nature, steroid hormones can

    diffuse across the cell membrane; classical steroid hormone receptors can thus be found

    intracellularly and have direct genomic effects [6]. More recently, also non-genomic effects of

    steroid hormones have been described, which are most likely regulated via more recently

    described membrane receptors for steroid hormones [7,8]. An alternative explanation is that by

    their lipophilic nature, sex steroids can alter membrane properties by integrating into the

    membrane, thereby changing the function of the immune cells [9].

    Classical intracellular estrogen receptors are present in human monocytes [10-16], human

    neutrophils [17], human dendritic cells [18], murine peripheral NK cells [19], human Tc

    lymphocytes and B lymphocytes [20,21]. There is no evidence that classical intracellular

    progesterone receptors are expressed on resting lymphocytes [22-28], monocytes [24],

    neutrophils [29], NK cells, B lymphocytes, Treg cells and dendritic cells. Various studies,

    however, demonstrated that activated lymphocytes, for instance lymphocytes during pregnancy,

    can upregulate progesterone receptors [25-28,30,31]. More recently membrane-bound

    progesterone receptors have been shown to be present on resting lymphocytes [8,32].Androgen receptors have long been considered not to be present on T lymphocytes [20,33].

    3 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    4/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    More recent studies, however, have demonstrated testosterone receptors in Th lymphocytes in

    mice [34] and membrane-bound testosterone receptors on lymphocytes, which are different

    from the classical intracellular testosterone receptors [35]. B-lymphocytes do express

    intracellular androgen receptors [36]. Androgen receptor expression was also found in murine

    macrophages [37] and androgen receptor mRNA was found in human macrophages [38]. Thereare no reports about the presence of androgen receptors on human monocytes, neutrophils, NK

    cells, dendritic cells or Treg cells.

    Influence of estrogen, progesterone and testosterone on immune cells

    The most obvious effects of sex hormones on the immune response are the effects of thesehormones on the numbers of circulating immune cells. In the peripheral blood, about 65% of the

    leukocytes are granulocytes (90% neutrophils), 5-10% is monocytes, and 30% are lymphocytes

    (85-90% T lymphocytes and 10-15% B lymphocytes). Sex hormones have been shown to affect

    these cell numbers by affecting proliferation or apoptosis of the cells or by recruitment of new

    cells from the bone marrow [39,40]. Although total white blood cell counts did not differ between

    males and females, an increase in white blood cells counts was observed in the luteal phase

    (and during pregnancy) as compared with the follicular phase of the ovarian cycle [40-43]. This

    may be largely due to an increase in granulocyte numbers in these reproductive conditions

    [42,44-47]. This suggests a role for progesterone and/or estrogen in increasing the numbers of

    granulocytes. In addition, various studies point to a decrease of monocyte numbers in thepresence of estrogen, as shown by increased numbers of monocytes in males and menopausal

    women as compared with women in the follicular phase of the ovarian cycle [14,48]. However,

    the presence of estrogen together with progesterone may increase monocyte numbers as

    monocyte numbers are increased in the luteal phase and during pregnancy as compared with

    the follicular phase. Whether sex hormones also affect B and T lymphocyte numbers remains to

    be established. Conflicting results have been published [41,42,46,49-56]. Treg cell numbers

    may be modulated by sex hormones, since it has been shown that both estrogen [57,58] and

    testosterone [59] increase Treg cells numbers.

    Cells of the nonspecific immune system

    Monocytes

    One of the functions of monocytes is ingesting and killing micro-organisms by a process ofphagocytosis. A few studies have indicated that sex hormones may affect this function of

    4 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    5/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    monocytes. For instance, numbers of Fc receptors on monocytes, which are involved in the

    binding and phagocytosis of Ig coated particles, are increased on monocytes from females as

    compared with males [60]. In addition, it has been shown that androgens may decrease the

    number of Fc receptors on monocytes in certain patient groups [61], while animal studies have

    shown that estrogen is capable of enhancing clearance of IgG coated red blood cells [62].Another important function of monocytes is to direct immune responses by the production of

    cytokines. Cytokines mostly studied in this respect are: tumor necrosis factor (TNF)-alpha,

    IL-1beta, IL-12, and IL-6.

    TNF-alpha

    TNF-alpha has pleiotropic actions and has emerged as an especially important mediator in

    pro-inflammatory responses and activation of T cells [63]. Various observations suggest that

    sex hormones may influence monocyte TNF-alpha production: in males endotoxin-stimulated

    monocytes produce more TNF-alpha as compared to females [48,64,65]. Whether this is due to

    direct effects of high levels of testosterone in males remains uncertain since in vitro studies

    showed no effect of testosterone upon monocyte TNF-alpha production [66]. Furthermore,

    endotoxin-stimulated monocytes of women in the luteal phase produce more TNF-alpha as

    compared to monocytes of women in the follicular phase [7,42,67]. Although this suggests a

    role for the female sex hormones in increasing monocyte TNF-alpha production, hormone

    replacement therapy (HRT) in postmenopausal women and OCC use did not affect TNF-alphaproduction by monocytes [68,69]. These observations indicate that also other factors, apart from

    estrogen and progesterone, may affect monocyte TNF-alpha production.

    Various papers describe in vitro experiments in which stimulated monocytes were incubated

    with estrogen or progesterone. The results are conflicting and vary from a down regulation of

    endotoxin-induced TNF-alpha production by estrogen at both physiological and

    supraphysiological levels [64,70] to no effect of either estrogen or progesterone upon

    TNF-alpha production in stimulated monocytes [16,68,71-73].

    IL-1beta

    Production of IL-1beta, which mediates a wide variety of immune responses, also shows

    differences in different reproductive stages. In the luteal phase an increased IL-1beta plasma

    concentration and an increased frequency of IL-1beta-producing monocytes after endotoxinstimulation was demonstrated as compared to the follicular phase [42,74,75]. This suggests a

    5 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    6/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    potentiating effect of estrogen and/or progesterone on monocyte IL-1beta production. However,

    also other mechanisms may be present since in males a higher frequency of IL-1beta-producing

    monocytes after endotoxin stimulation was demonstrated as compared with females in the

    follicular phase [48]. The effect of testosterone upon monocyte IL-1beta production in vitro was

    also studied by us and others. Although we showed that incubation of whole blood withphysiological concentrations of testosterone increased monocyte IL-1beta production [66],

    Morishita et al [76] did not find this.

    IL-12

    IL-12 is mainly produced by monocytes and macrophages and plays an important role in theinduction of cell-mediated immunity; i.e. together with IFN gamma it is a major inducer of Th1

    differentiation [77]. IL-12 is thus an important cytokine that links the nonspecific immune system

    to the specific immune system. We have shown no differences in IL-12 production after

    LPS-stimulation when comparing monocytes from the luteal phase with those from the follicular

    phase in women [42]. In men, however, the IL-12 production by monocytes after LPS

    stimulation was increased as compared with women [42,48]. This may suggest that

    testosterone stimulates monocyte IL-12 production. Indeed, physiological levels of testosterone

    increased IL-12 production by LPS-stimulated monocytes in vitro [66], while no effect of

    estrogen on LPS-stimulated IL-12 production was found [43]. However, others found a

    decreasing effect [78] or an increasing [71] effect of estrogen on stimulated IL-12 production;progesterone did not affect the production of IL-12 in vitro [43,78].

    IL-6

    IL-6 stimulates B lymphocyte and T lymphocyte differentiation and activates macrophages and

    NK cells, while it also possesses anti-inflammatory properties. It is generally thought that IL-6production is decreased by estrogen. Indeed, plasma IL-6 levels are increased after menopause

    [79-82] and decreased by HRT treatment [18,81,83]. However, no variations in plasma IL-6

    levels or spontaneous leukocyte IL-6 production during the menstrual cycle [7,67,84-88] were

    found. Whether the production of IL-6 after LPS is stimulation is also influenced by estrogen is

    unclear. No difference [86,89], an increase [88] or a decrease [65] in IL-6 production was found

    between the follicular and luteal phase when whole blood was stimulated with LPS. Moreover,

    stimulated IL-6 production was either decreased [90] or not affected [69,91] by the estrogenic

    compound in HRT.

    6 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    7/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    The effects of gender and reproductive condition upon monocyte functions are obvious. The

    most consistent effects are: plasma IL-6 levels appear to be decreased by estrogens;

    LPS-stimulated TNF-alpha and IL-1beta productions are increased in males as compared to

    females, and these are also increased in the luteal phase as compared to the follicular phase of

    the ovarian cycle; LPS-stimulated IL-12 production is only increased in males and not affectedby the ovarian cycle. These differences in monocyte function may play a role in the differences

    in immune responses between sex and reproductive condition. Whether these differences are

    due to direct effects of sex hormones remains uncertain, since in vitro experiments in which

    monocytes were incubated with sex hormones revealed conflicting results upon monocyte

    cytokine production. Such conflicting results may be due to differences in experimental setup.

    For instance incubations with sex hormones are performed using whole blood, peripheral blood

    mononuclear cells or isolated monocytes.

    Macrophages

    Macrophages are tissue-residing cells derived from monocytes [92]. In these tissues they stand

    guard against foreign invaders and alert the adaptive immune system with signals and through

    antigen presentation. In order to maximize their effector functions, macrophages need to be

    activated and the type of activation determines its effector functions [93]. Presently, two major

    macrophage activation states have been described: the classically activated state and the

    alternatively activated state [93]. The first one is associated with increased generation of oxygenradicals and cell killing to remove invading microorganisms and the second one with increased

    expression of phagocytic receptors to remove cell debri and increased tissue remodeling to

    reconstruct the tissue during wound healing. In addition, macrophages with a nonactivated,

    anti-inflammatory phenotype have been described, these are called the immunosuppressive

    macrophages [93]. These producers of IL-10 are important in controlling inflammation and

    collateral damage associated with excessive inflammation [93]. Sex hormones have been found

    to affect these activation states and effector functions in macrophages and their effects are

    reviewed below.

    Classically activated macrophages: Macrophages become classically activated after exposure

    to IFN gamma or microbial products like LPS and they upregulate mechanisms to destroy

    microorganisms [93]. As the classically activated phenotype has been known for the longest

    time, most studies investigating effects of sex hormones on macrophage activation have studied

    this phenotype. Many studies have described upregulation of inducible nitric oxide (NO)

    synthase (iNOS) and NO production, increased production of proinflammatory cytokines and

    increased cell surface expression of Toll-like receptor (TLR)4 in macrophages [94-99] after

    treatment with estrogens. This may lead to enhanced killing of microorganisms and thus to

    increased resistance to extracellular bacteria and infections in the presence of estrogen [100].Progesterone on the other hand was found to inhibit NO production, TLR4 expression and

    7 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    8/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    proinflammatory cytokine production [101-104], counteracting the effects of estrogens. The net

    effect on classical activation of macrophages therefore seems to depend on the ratio of these

    two hormones, though this has not been studied yet to the best of our knowledge.

    Like progesterone, androgens have also been found to reduce classical activation by inhibiting

    TLR4 expression and oxygen radical formation [105-107], and this may explain the increasedsusceptibility of men for infections [108-110]

    Alternatively activated macrophages: Macrophages become alternatively activated after

    exposure to IL-4 and/or IL-13 and they adopt a phenotype intent on taking up apoptotic cells

    and cell debri and reconstructing damaged tissues. This phenotype has been discovered

    relatively recently and the effects of sex hormones have not been studied much in relation to

    alternative activation. The few studies published that did investigate sex hormones and

    alternative activation showed increased alternative activation of macrophages after treatmentwith either progesterone or estrogen and decreased alternative activation after treatment with

    androgens [111,112].

    Immunosuppressive macrophages: This subset is the least-known of the subsets and little to no

    data are available about the effects sex hormones have on this phenotype. One study did

    investigate IL-10 production by macrophages from hemorrhaged mice treated with estrogens or

    androgens and found reduced production by estrogen and increased production by androgen

    [113]. These findings are again in line with the anti-inflammatory effects usually found for

    androgens.

    Dendritic cells

    Dendritic cells are also tissue-residing cells derived from monocytes [92]. Immature dendritic

    cells reside in the tissues. They may be activated by microbial products or inflammatory

    cytokines after which they differentiate into mature dendritic cells [114], while migrating to lymph

    nodes to present antigen to naive T cells. In human dendritic cells, derived from monocytes in

    vitro, both estrogen and progesterone upregulated IL-10 production by mature and immaturedendritic cells [115,116]. These hormones also increased apoptosis of dendritic cells [116]. On

    the other hand, neither progesterone, nor estrogen affected the phenotype of mature and

    immature dendritic cells [115] or the T cell stimulatory capacity [116]. Although analysis of

    subpopulations of dendritic cells is still difficult, it has been shown recently that progesterone

    inhibited IFN gamma production by plasmacoid dendritic cells [117]. Since IFN gamma is

    necessary to fight viruses and also primes antiviral responses of the specific immune system,

    progesterone may therefore inhibit antiviral responses [117].

    Neutrophils

    8 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    9/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    Neutrophils play a key role in the first-line defense against invading pathogens by phagocytosis,

    the release of anti-microbials and the generation of neutrophil extracellular traps [118].

    Therefore, they need to be able to respond to chemotactic stimuli to get to the place of actionand they need to be able to produce those anti-microbial factors. Studies have shown that

    progesterone enhanced chemotactic activity of neutrophils, while estrogens decreased this

    activity [119]. Various groups have also investigated the effects of progesterone and estrogen

    on free radical production by neutrophils. This has been shown to be increased [120],

    decreased [121] or not affected [122] by estrogen or progesterone incubations in vitro. Sex

    hormones also affect NO production via NOS activity. NO production by neutrophils was found

    to have anti-inflammatory effects since it prevented neutrophil adhesion to the endothelium

    [123]. NO-synthase in neutrophils varies with reproductive condition: increased NO synthase

    expression was observed in vivo in the luteal phase as compared with the follicular phase and

    in postmenopausal women on estrogen therapy as compared with untreated postmenopausalwomen [124]. This is in line with in vitro results showing that estrogen upregulated NOS

    expression in neutrophils in vitro [124,125].

    Taken together, it appears that both sex and reproductive condition affect neutrophil function. In

    general, estrogen has anti-inflammatory effects on neutrophils, while progesterone has

    pro-inflammatory effects on these cells. Therefore, sex hormones can affect nonspecific

    immune responses in vivo by modulating neutrophil function.

    NK cells

    There are various reports on the effects of the reproductive condition and gender sex on the

    main function of NK cells, i.e. their ability to lyse other cells. Various studies have shown a

    negative association of the lysing activity of NK cells with estrogen or progesterone levels: an

    increased potency to lyse other cells was found in postmenopausal women and in males ascompared to females with a regular menstrual cycle and women on OCC [126,127]. In addition,

    exposure to OCC caused a reduction in NK-activity as compared to non-users [126,128,129],

    while NK cell activity was decreased in postmenopausal women using HRT [130]. Indeed, in

    vitro studies have directly demonstrated that estrogen decreased the potency of NK cells to lyse

    other cells [131,132], while no effect of progesterone on NK cell activity was demonstrated

    [132,133]. Unfortunately, the suppressive effect of estrogen on NK cell activity is not always

    observed during the menstrual cycle, since results vary from no difference between follicular

    and luteal phase, as well as highest NK cell activity in follicular phase, periovulatory phase or

    luteal phase [126,127,134-136]. Such different results may be due to different time points in the

    ovarian cycle of measuring the NK cell activity.

    9 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    10/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    Another important function of NK cells is cytokine production. The cytokine repertoire of

    peripheral NK cells is mainly type 1 cytokines (IFN gamma) [137,138]. Although there are many

    studies into cytokine production of uterine NK cells during pregnancy, little is known aboutcytokine production by peripheral NK cells in relation to reproductive condition or separate

    effects of sex hormones on peripheral NK cells. During pregnancy, induced IFN gamma

    production of peripheral NK cells was found to be decreased, however, no effect of the

    menstrual cycle upon IFN gamma production of NK cells was found [139]. This suggests that

    during pregnancy other mechanisms rather than sex hormones affect IFN gamma production by

    NK cells.

    Effect of sex hormones on the nonspecific immune response in vivo

    Acute immune actions, which are part of the innate immune system, appear to work favorably

    for women as compared to men. In general, men are more susceptible to viral, bacterial, fungal

    and parasitic infections (with the exception of sexually transmitted diseases) then women

    [140,141]. They also have a higher risk of developing more severe disease [142]. This may be

    due to the effects of androgens and estrogens on the innate immune cells. As described above,

    in general androgens have been shown to suppress innate immune cell function, resulting in

    suppression of the resistance to infections [105], whereas estrogens were found to stimulateinnate immune cells, which can promote resistance to infections [143]. The effects of androgen

    and estrogen on non-specific immune responses are also obvious from the fact that male sex is

    a significant risk factor for the development of sepsis. This is supported by many animal models

    of endotoxemia and bacterial challenge in which females demonstrate better survival when

    subjected to severe sepsis than males [142]. In males infections can more easily progress to

    damaging inflammation, hence the increased susceptibility for sepsis. More on the mechanisms

    can be read in some excellent reviews on the subject [144-146].

    Wound healing also differs between males and females [147]. Recent studies have suggested

    that the male genotype is a strong risk factor for impaired wound healing in the elderly

    [148-150]. These sex effects are mediated by estrogens and androgens, since wound healing

    increases after estrogen treatment [151] and after deprivation of androgens after castration of

    male mice [152]. The striking acceleration of wound regeneration after androgen deprivation

    was associated with a reduced inflammatory response [152]. Although exact mechanisms by

    which sex hormones affect wound healing are not clear yet, it seems likely that the effects are,

    at least partly, mediated by the effects of sex hormones on the nonspecific immune response.

    For instance, the effects of sex hormones on the macrophage subsets may be important in

    wound healing, since a subset switching of macrophages is essential in wound healing[153,154]. The types of macrophages present in wounds determine whether chronic

    10 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    11/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    inflammation will occur or whether proper healing is started [111]; classically activated

    macrophages are needed in the first stage, whereas alternatively activated macrophages have

    been found important in the wound healing stage. Both estrogen and progesterone have been

    found to promote wound healing by enhancing alternative activation of macrophages and failure

    to switch was associated with chronic inflammation of wounds [111].

    Cells of the specific immune system

    Cytotoxic T lymphocytes (Tc)

    Sex hormones have been described to affect Tc lymphocyte function (reviewed by Grossman et

    al., Reference 155). Many studies (vitro and in vivo) have shown that both estrogen and

    progesterone suppress cell-mediated immune responses [155]. Although the exact mechanisms

    are unclear, estrogen and progesterone inhibited proliferation of Tc lymphocytes (or Tc

    lymphocyte cell lines) after stimulation with mitogenic substances [156-159]. In addition, both

    hormones have also been shown to increase apoptosis in activated Tc lymphocytes [160]. Only

    a few studies have evaluated the role of sex hormones on Tc lymphocyte cytolytic activity. One

    study suggested that cytolytic activity of Tc lymphocytes in the uterine endometrium is

    depressed by estrogen and progesterone [161] and another study suggested that progesteronedecreased cytolytic activity of these lymphocytes from the decidua [162]. Also, cytotoxicity in

    peripheral Tc lymphocytes was decreased by estrogen [162].

    Helper T lymphocytes (Th)

    The main function of Th lymphocytes is cytokine production. As indicated above, Thlymphocytes can be subdivided in Th1, Th2, Th9, Th17 and Treg. The effects of sex hormones

    on these subtypes will be discussed below.

    Th1 subtype: The most important cytokine produced by Th1 cells is IFN gamma. Although

    various studies have evaluated the effects of sex hormones on IFN gamma production, it

    remains elusive whether there are direct effects of sex hormones on lymphocyte IFN gamma

    production. Increased IFN gamma production [163] as well as similar IFN gamma production

    [48] by stimulated male lymphocytes as compared to female lymphocytes was found.Furthermore, no effect of the menstrual cycle upon induced IFN gamma production was found

    11 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    12/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    [46], while induced IFN gamma was decreased in HRT-treated postmenopausal women as

    compared to untreated postmenopausal women [130]. Others, however, found no effect of

    synthetic hormones on lymphocyte IFN gamma production [90,164]. These in vivo results are in

    line with in vitro experiments in which neither progesterone, estrogen nor testosterone altered

    IFN gamma production [66,163,165].

    Th2 subtype: As estrogen has been shown to enhance humoral immune responses [166] it

    seems likely that it would also affect Th2 cytokine production. IL-4 is released predominantly by

    Th2 lymphocytes. In males and after menopause IL-4 production is similar to fertile women

    [163,167,168], suggesting no effect of sex hormones on the production of IL-4. In contrast to

    this suggestion the production of IL-4 was significantly increased in Th cells in the luteal phase

    as compared with the follicular phase of the ovarian cycle [46]. This may be due to increased

    progesterone concentrations in the luteal phase, since one study demonstrated an increase inIL-4 production by Th lymphocytes after incubation with progesterone [165]. Synthetic

    hormones, such as in OCC or HRT, have not been found to affect IL-4 production by

    lymphocytes [90,163,168].

    Interleukin-10 (IL-10), another Th2 type cytokine was not produced differently by lymphocytes of

    both males or postmenopausal women and premenopausal women [48,163]. In addition, during

    the menstrual cycle IL-10 production by lymphocytes after stimulation is stable [46,169],

    suggesting no effect of sex hormones on IL-10 production. Results with oral contraceptives,which do not influence IL-10 production [164] and in vitro experiments, which showed no effect

    of estrogen, progesterone or testosterone on IL-10 production [66,163,165] after polyclonal

    stimulation of whole T lymphocyte populations, corroborate this suggestion.

    Th17 and Th9 subtype: Unfortunately, at this time no data exist showing an effect of sex

    hormones on the Th17 or Th9 subtype. Apart from pregnancy, in which the number of Th17

    cells does not differ from non-pregnant individuals, also no data are available on the effect of

    the reproductive condition on Th17 or Th9 cells [170].

    Treg subtype: Numbers of Treg cells appear to be influenced by estrogen [57,58] and

    testosterone [59], but not much is known whether Treg cells function is also under control of sex

    hormones. Human data are scarce at this moment. One study showed that estrogen increases

    suppressive function of Treg cells [57]. This potentiating of suppressive function by estrogen

    has been confirmed in mice studies. Estrogen increased Foxp3 expression in mice Treg cells in

    vitro [58,171,172]. Foxp3 is a transcription factor present in Treg cells that controls the

    development and function of Treg cells [171,172]. An increase in this factor by estrogen thus

    suggests that estrogen increases Treg cell function. In vivo studies in mice corroborated this

    suggestion: estrogen treatment upregulated Foxp3 expression together with Treg cell function in

    mice [58,173].

    12 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    13/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    B lymphocytes

    The main function of B lymphocytes is the production of antibodies. Effects of sex on B cell

    function can therefore be derived from differences in plasma levels of antibodies. Females have

    higher serum levels of total IgM and IgG as compared to males [174-178] with no changes

    during the menstrual cycle [52,179]. During OCC use immunoglobulin levels and

    immunoglobulin production are unaltered as compared to females not taking OCC [180,181],

    but others found immunoglobulin levels to be decreased [182] or even increased [183] in

    females using OCC as compared to females not using OCC. The higher serum levels of

    immunoglobulin in females may suggest a stimulating effect of female sex hormones or an

    inhibiting effect of testosterone upon this parameter or both. Indeed, in vitro studies, haveshown that estrogen induced polyclonal activation of human B cells and increased IgG and IgM

    production by PBMCs [184,185]. Testosterone inhibited immunoglobulin IgG and IgM

    productions [186]. It has also been shown that estrogen increased and testosterone decreased

    autoantibody production by PBMC in patients with SLE [187,188]. Animal data have shown that

    estrogen also induced a switch in antibody isotype. In mice treated with estrogen, autoantibody

    production was increased and the antibodies were mainly of the IgG isotype [189,190]. Present

    evidence to date thus points towards an important role for estrogen and testosterone in antibody

    production.

    In conclusion, there are obvious effects of sex hormones on lymphocytes. As described above,

    both female sex hormones seem to inhibit Tc cytotoxicity, while estrogen and testosterone also

    affect humoral immunity: estrogen upregulates (auto)antibody production and testosterone

    inhibits (auto)antibody production. Whether sex hormones also affect lymphocyte cytokine

    production remains to be established. More recently, it has been shown that sex hormones also

    affect Treg: estrogen both increased Treg cell numbers and Treg cell function.

    Effects of sex hormones on specific immune responses in vivo

    The effects of sex hormones on the immune response in vivo can be studied using immune

    disease models and study the effects of gender, reproductive condition and sex hormone

    treatment on the incidence and course of the disease. The ability to mount more vigorous

    cell-mediated and humoral immune responses in women increases their chances of developing

    unwanted Th1, Th2, Th9, and Th17-mediated responses to self-antigens or otherwise

    innocuous compounds such as allergens. The result is increased incidences of autoimmunediseases like multiple sclerosis (MS) and allergic diseases like asthma in women. Until recently

    13 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    14/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    these self-reactive and allergen-reactive processes were primarily thought to be associated with

    Th1 cells and Th2 cells, but with the discovery of Th17 and Th9 cells, the disease mechanisms

    appear to be far more complex [191]. Unfortunately, little to no data is available about the

    effects of sex hormones on these new subsets in the context of autoimmune diseases and

    asthma. Therefore, we will shortly discuss the effects of sex hormones on the development andcourse of MS as an example of a Th1-mediated disease and asthma as a Th2-mediated

    disease and keep Th17 and Th9 out of this context for the moment.

    Multiple sclerosis (MS)

    MS is an autoimmune disease characterized by a Th1 and Th17-mediated chronic inflammatorydemyelinating process of axons in the central nervous system and is more common in women

    than in men [191,192]. In 80% of the cases, MS starts with a relapsing/remitting phenotype and

    may become progressive after several years [193]. Changes in MS symptoms have been

    related to reproductive status. About 40% of women with relapsing/remitting MS have reported

    an increase in symptoms in the time preceding menstruation when both estrogen and

    progesterone levels have dropped [194,195]. In contrast, in the third trimester of pregnancy

    when estrogen and progesterone levels are highest, the rate of relapse significantly reduces,

    only to increase again post-partum when hormone levels have dropped [196,197]. Little data

    exists on the effects of OCC and HRT use. Two studies have reported an improvement of MS

    symptoms in women using OCC [194,195] and one study reported an improvement in mostwomen using HRT during menopause [198].

    The effects of sex hormones on MS development have been tested quite extensively in a model

    of MS called experimental autoimmune encephalitis (EAE), as reviewed by Van den Broek et

    al., recently [199]. The role of estrogen in MS and EAE is complex and seems to be

    dichotomous: on the one hand it appears to increase the risk of developing the disease, while

    on the other hand high concentrations were found to be beneficial on the clinical manifestations

    of EAE. Most of the studies have focused on the protective effects of sex hormones in thecourse of the disease. There may be different mechanisms by which estrogen affects MS. The

    protective effect of high concentrations of estrogen may be due to an increase of the

    anti-inflammatory IL-10 production of specific T cell clones directed against proteins of the

    myelin sheath [200]. Also the homing of destructive Th1 and Th17 cells into the central nervous

    system was limited by estrogen [201]. Furthermore, Treg cell function and numbers were found

    increased by high levels of estrogen and these can limit the expansion of Th1 effector cells

    [57,58,202,203]. A regulatory B cell mediated protection has also been shown [204]. Finally, the

    effect of estrogen may also be mediated through decreased numbers of macrophages and

    dendritic cells in the central nervous system [205].

    Progesterone also has protective effects on experimental MS [206-209] as it attenuated diseaseseverity and reduced the inflammatory response. The high progesterone and estrogen during

    14 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    15/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    pregnancy may thus explain the improvement of MS signs during pregnancy [210]. It was

    speculated that the Th2-promoting effects of progesterone [199] may be responsible for tipping

    the Th1/Th2 balance in a protective direction, but no formal evidence for this hypothesis exists.

    The neuroprotective effects of progesterone were accompanied by increased IL-10 production

    and increased infiltration of B cells, possibly regulatory B cells, and cytotoxic T cells into thespinal cord [207].

    Administration of androgen significantly delayed onset and progression of EAE and its

    protective effects are postulated to be responsible for the reduced susceptibility of men for MS

    [211-213]. Both castration of male mice and treatment of female mice with androgens showed

    similar results. Androgen was found to inhibit infiltration of Th1 cells into the spinal cord [214] as

    well as reduce the expression of Th1 cytokines [212] and increase the production of

    anti-inflammatory cytokine IL-10 [199].The effects of female sex hormones on Th1 inflammation in the context of EAE all point at

    protection against disease. This may explain why women remit during pregnancy, but it fails to

    explain why women are at risk of developing MS in the first place. This particular question

    needs more research. The effects of androgens on EAE development are consistent with what

    is found in men: androgens inhibit EAE development, which is why men are less susceptible to

    the disease.

    Asthma

    Like autoimmune diseases, asthma is also more prevalent among women in the reproductive

    stages of life than men [215,216]. Asthma is a common chronic inflammation of the airways,

    which is characterized by a Th2 polarization with an overabundance of eosinophils, mast cells,

    and activated Th2 lymphocytes in the lungs and increased levels of IgE in serum.

    There is substantial support for a role of female sex hormones in the development of asthma,

    starting with the fact that the prevalence of asthma and other atopic conditions is higher in boys

    than girls before puberty and reverses to a higher prevalence in girls and women after puberty

    when female sex hormone levels have increased [217].

    Also other observations suggest a link between female sex hormones and the development of

    asthma since an earlier menarche increases the risk of developing asthma in females [218,219].

    As described above for MS, the effects of estrogen on asthma also appear to be dichotomous.Although it may increase the incidence of asthma, estrogen also appears to be beneficial, since

    15 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    16/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    30% to 40% of menstruating female asthmatics experience perimenstrual asthma worsening

    with increased symptoms and a greater likelihood of hospitalization [220,221]. Since estrogen

    levels are low perimenstrually, this suggests a protective effect of estrogen on asthma. Indeed,

    most of the limited number of case reports and studies investigating OCC use suggest a small

    beneficial effect on perimenstrual asthma and mild asthma in general among women[218,220,222]. With the Th2 dominance during pregnancy, one would expect an increase in

    asthma severity in pregnant women with asthma. However, this does not seem to be the case in

    women with mild asthma: in an equal number of women symptoms worsen, stay the same, or

    decrease with pregnancy. Only in women with severe asthma, symptoms may increase with

    pregnancy [223]. After menopause asthma incidence declines in women as compared to men

    and younger women, but not in women who receive HRT to treat symptoms of menopause

    [224-228].

    A small number of investigations in animal models have tried to elucidate how female sex

    hormones affect asthma. The few studies published have focused little on mechanisms but

    rather on inflammatory and allergic endpoints like airway hyperresponsiveness, IgE levels and

    eosinophils. Yet these studies failed to come up with conclusive proof of the effects of female

    sex hormones on asthma development because they reported both inhibition and aggravation of

    airway inflammation by estrogen and progesterone [229-234]. The effects of testosterone on the

    other hand are clear: testosterone has been shown to suppress Th2 inflammation in the lungs

    [235,236]. In addition, in an interesting study by Okuyama et al. it was shown that sex

    differences in airway inflammation are not only caused by the hormonal environment during

    inflammation, but also by intrinsic differences between male and female immune cells [237].These intrinsic differences between males and females are probably partly due to local

    regulation of airway inflammation by macrophages and not by T cells [238,239].

    Results from these animal models show that the influence of female sex hormones on Th2-type

    inflammations is most likely a complex interplay of estrogens, progesterone and their receptors.

    To be able to tease out their effects we need to understand which parts of the Th2 immune

    response cascade are different between males and females to identify the most likely targets for

    the modulation by sex hormones. The effects of androgens are clearer showing inhibition ofTh2-type inflammation, which explains the reduced susceptibility of men.

    Conclusions

    The aim of this review was to describe the effects of sex hormones, both female (estrogen and

    progesterone) as well as male sex hormones (testosterone), on immune responses in humans.Available evidence from animal studies suggests that sex hormones regulate immune

    16 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    17/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    responses in vivo (as reviewed in Reference 1). Since clinical data on human immune

    responses are largely lacking, we focused on the effects of sex hormones on isolated human

    immune cells and on human data of effects of sex and reproductive condition on two immune

    mediated diseases (MS and asthma) as well as on experiments studying the role of sex

    hormones in animal models for these data.

    In the past, the effects of gender and the reproductive condition upon the specific immune

    response have gained much more attention then the effects on the nonspecific immune

    response. It is now much clearer that there are also important effects of sex hormones on cells

    of the nonspecific immune response. This should not be surprising, since many reproductive

    processes, such as ovulation and menstruation, are regulated by nonspecific immune

    responses. Ovaries therefore have an interest in regulating the nonspecific immune response.

    Sex hormones regulate nonspecific immune responses, by affecting monocyte, macrophage,granulocyte and NK cell numbers, but also by affecting the function of these cells.

    Many studies have been performed on the effects of sex hormones on the specific immune

    response. At present, evidence points towards an important role for estrogens and testosterone

    in the humoral response i.e. (auto)antibody production; estrogen increases, while testosterone

    decreases antibody production. Sex hormones also have prominent effects on the cellular

    immune response, by affecting T lymphocytes. Numbers of T lymphocytes are decreased in

    males and female sex hormones appear to inhibit T cell proliferation and cytotoxicity of Tclymphocytes. Th cells are also affected by female sex hormones: progesterone induces Th2

    development and estrogen seems to increase the number and function of regulatory T cells.

    Unfortunately, in vivo studies on the effects of sex hormones on immune responses in humans

    are largely lacking. Although the female preponderance in autoimmune diseases has been

    known for many years, the mechanisms for this are not clear. Effects of the reproductive

    condition and sex hormone treatment on symptoms of autoimmune diseases and asthma are

    not consistent. This may amongst others be due to a lack of standardization how and whensymptoms are scored and what sex hormone treatment is given (combination of

    progesterone/estrogen treatment, duration of treatment, amount of hormones given). Such a

    lack of standardization is also seen in animal models. Therefore in order to get more insight into

    the effects of sex hormones on immune responses and immune based diseases, it is important

    to standardize experiments and not only study effects of sex hormones separately but also in

    combination and in different concentrations and for different time periods.

    It is obvious from this review that we have decades worth of research, which have alreadytaught us a lot about how sex hormones influence the immune system. However, with every

    17 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    18/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    new discovery more questions emerge. For instance, the recent discovery of Th9, Th17 and

    Treg subsets make us wonder how sex hormones affect these subsets. Future research should

    focus on these subsets, but also on how these hormones work together in more complicated

    set-ups than isolated cells. This is not an easy task, but with the ever-expanding research tools

    and the increased interest in sex differences in disease development and management weshould be able to move forward in this challenging field.

    Author(s) Affiliation

    MM Faas - Immunoendocrinology, Division of Medical Biology, Department of Pathology and

    Medical Biology, University Medical Centre Groningen and University of Groningen

    P de Vos - Immunoendocrinology, Division of Medical Biology, Department of Pathology and

    Medical Biology, University Medical Centre Groningen and University of Groningen

    BN Melgert - Department of Pharmacokinetics, Toxicology and Targeting, University Center forPharmacy, University of Groningen, Groningen, The Netherlands

    References

    1. Ansar AS, Penhale WJ, Talal N. Sex hormones, immune responses, and autoimmune

    diseases. Mechanisms of sex hormone action. Am J Pathol 1985; 121(3): 531-51.

    2. Case AM, Reid RL. Effects of the menstrual cycle on medical disorders. Arch Intern Med1998; 158(13): 1405-12.

    3. Skobeloff EM, Spivey WH, Silverman R, Eskin BA, Harchelroad F, Alessi TV. The effect

    of the menstrual cycle on asthma presentations in the emergency department. Arch Intern Med

    1996; 156(16): 1837-40.

    4. Whitacre CC. Sex differences in autoimmune disease. Nat Immunol 2001; 2(9): 777-80.

    5. Murphy KM, Stockinger B. Effector T cell plasticity: flexibility in the face of changing

    circumstances. Nat Immunol 2010; 11(8): 674-80.

    6. Beato M, Sanchez-Pacheco A. Interaction of steroid hormone receptors with the

    transcription initiation complex. Endocr Rev 1996; 17(6): 587-609.

    7. Marino M, Galluzzo P, Ascenzi P. Estrogen signaling multiple pathways to impact genetranscription. Curr Genomics 2006; 7(8): 497-508.

    8. Dosiou C, Hamilton AE, Pang Y, Overgaard MT, Tulac S, Dong J et al. Expression of

    membrane progesterone receptors on human T lymphocytes and Jurkat cells and activation of

    G-proteins by progesterone. J Endocrinol 2008; 196(1): 67-77.

    9. Lamche HR, Silberstein PT, Knabe AC, Thomas DD, Jacob HS, Hammerschmidt DE.

    Steroids decrease granulocyte membrane fluidity, while phorbol ester increases membrane

    fluidity. Studies using electron paramagnetic resonance. Inflammation 1990; 14(1): 61-70.

    10. Suenaga R, Evans MJ, Mitamura K, Rider V, Abdou NI. Peripheral blood T cells and

    monocytes and B cell lines derived from patients with lupus express estrogen receptortranscripts similar to those of normal cells. J Rheumatol 1998; 25(7): 1305-12.

    18 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    19/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    11. Suenaga R, Mitamura K, Evans MJ, Abdou NI. Binding affinity and quantity of estrogen

    receptor in peripheral blood monocytes of patients with systemic lupus erythematosus. Lupus

    1996; 5(3): 227-31.

    12. White MM, Zamudio S, Stevens T, Tyler R, Lindenfeld J, Leslie K, Moore LG. Estrogen,

    progesterone, and vascular reactivity: potential cellular mechanisms. Endocr Rev 1995; 16(6):739-51.

    13. Weusten JJ, Blankenstein MA, Gmelig-Meyling FH, Schuurman HJ, Kater L, Thijssen

    JH. Presence of oestrogen receptors in human blood mononuclear cells and thymocytes. Acta

    Endocrinol (Copenh) 1986; 112(3): 409-14.

    14. Ben Hur H, Mor G, Insler V, Blickstein I, Amir-Zaltsman Y, Sharp A et al. Menopause is

    associated with a significant increase in blood monocyte number and a relative decrease in the

    expression of estrogen receptors in human peripheral monocytes. Am J Reprod Immunol 1995;

    34(6): 363-9.

    15. Wada K, Itoh T, Nakagawa M, Misao R, Mori H, Tamaya T. Estrogen binding sites in

    peripheral blood monocytes and effects of danazol on their sites in vitro. Gen Pharmacol 1992;23(4): 693-700.

    16. Phiel KL, Henderson RA, Adelman SJ, Elloso MM. Differential estrogen receptor gene

    expression in human peripheral blood mononuclear cell populations. Immunol Lett 2005; 97(1):

    107-13.

    17. Molero L, Garcia-Duran M, Diaz-Recasens J, Rico L, Casado S, Lopez-Farre A.

    Expression of estrogen receptor subtypes and neuronal nitric oxide synthase in neutrophils from

    women and men: regulation by estrogen. Cardiovasc Res 2002; 56(1): 43-51.

    18. Komi J, Lassila O. Nonsteroidal anti-estrogens inhibit the functional differentiation of

    human monocyte-derived dendritic cells. Blood 2000; 95(9): 2875-82.

    19. Curran EM, Berghaus LJ, Vernetti NJ, Saporita AJ, Lubahn DB, Estes DM. Natural killercells express estrogen receptor-alpha and estrogen receptor-beta and can respond to estrogen

    via a non-estrogen receptor-alpha-mediated pathway. Cell Immunol 2001; 214(1): 12-20.

    20. Cohen JH, Danel L, Cordier G, Saez S, Revillard JP. Sex steroid receptors in peripheral

    T cells: absence of androgen receptors and restriction of estrogen receptors to OKT8-positive

    cells. J Immunol 1983; 131(6): 2767-71.

    21. Stimson WH. Oestrogen and human T lymphocytes: presence of specific receptors in

    the T-suppressor/cytotoxic subset. Scand J Immunol 1988; 28(3): 345-50.

    22. Mansour I, Reznikoff-Etievant MF, Netter A. No evidence for the expression of the

    progesterone receptor on peripheral blood lymphocytes during pregnancy. Hum Reprod 1994;9(8): 1546-9.

    23. Vegeto E, Pollio G, Pellicciari C, Maggi A. Estrogen and progesterone induction of

    survival of monoblastoid cells undergoing TNF-alpha-induced apoptosis. FASEB J 1999; 13(8):

    793-803.

    24. Schust DJ, Anderson DJ, Hill JA. Progesterone-induced immunosuppression is not

    mediated through the progesterone receptor. Hum Reprod 1996; 11(5): 980-5.

    25. Szekeres-Bartho J. Progesterone receptors on lymphocytes. Hum Reprod 1995; 10(3):

    695-6.

    26. Szekeres-Bartho J, Reznikoff-Etievant MF, Varga P, Pichon MF, Varga Z, Chaouat G.

    Lymphocytic progesterone receptors in normal and pathological human pregnancy. J ReprodImmunol 1989; 16(3): 239-47.

    19 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    20/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    27. Szekeres-Bartho J, Weill BJ, Mike G, Houssin D, Chaouat G. Progesterone receptors in

    lymphocytes of liver-transplanted and transfused patients. Immunol Lett 1989; 22(4): 259-61.

    28. Szekeres-Bartho J. Immunological relationship between the mother and the fetus. Int

    Rev Immunol 2002; 21(6): 471-95.29. Aerts JL, Christiaens MR, Vandekerckhove P. Evaluation of progesterone receptor

    expression in eosinophils using real-time quantitative PCR. Biochim Biophys Acta 2002;

    1571(3): 167-72.

    30. Barakonyi A, Polgar B, Szekeres-Bartho J. The role of gamma/delta T-cell

    receptor-positive cells in pregnancy: part II. Am J Reprod Immunol 1999; 42(2): 83-7.

    31. Polgar B, Barakonyi A, Xynos I, Szekeres-Bartho J. The role of gamma/delta T cell

    receptor positive cells in pregnancy. Am J Reprod Immunol 1999; 41(4): 239-44.

    32. Szekeres-Bartho J, Philibert D, Chaouat G. Progesterone suppression of pregnancy

    lymphocytes is not mediated by glucocorticoid effect. Am J Reprod Immunol 1990; 23(2): 42-3.

    33. Rife SU, Marquez MG, Escalante A, Velich T. The effect of testosterone on the immune

    response. 1. Mechanism of action on antibody-forming cells. Immunol Invest 1990; 19(3):

    259-70.

    34. Chien EJ, Chang CP, Lee WF, Su TH, Wu CH. Non-genomic immunosuppressive

    actions of progesterone inhibits PHA-induced alkalinization and activation in T cells. J Cell

    Biochem 2006; 99(1): 292-304.

    35. Benten WP, Lieberherr M, Giese G, Wrehlke C, Stamm O, Sekeris CE et al. Functional

    testosterone receptors in plasma membranes of T cells. FASEB J 1999; 13(1): 123-33.

    36. Benten WP, Stephan C, Wunderlich F. B cells express intracellular but not surface

    receptors for testosterone and estradiol. Steroids 2002; 67(7): 647-54.37. Bebo BF, Jr., Schuster JC, Vandenbark AA, Offner H. Androgens alter the cytokine

    profile and reduce encephalitogenicity of myelin-reactive T cells. J Immunol 1999; 162(1):

    35-40.

    38. Grimaldi CM. Sex and systemic lupus erythematosus: the role of the sex hormones

    estrogen and prolactin on the regulation of autoreactive B cells. Curr Opin Rheumatol 2006;

    18(5): 456-61.

    39. Thongngarm T, Jenkins JK, Ndebele K, McMurray RW. Estrogen and progesterone

    modulate monocyte cell cycle progression and apoptosis. Am J Reprod Immunol 2003; 49(3):

    129-38.

    40. Bain BJ, England JM. Variations in leucocyte count during menstrual cycle. Br Med J1975; 2(5969): 473-5.

    41. Mathur S, Mathur RS, Goust JM, Williamson HO, Fudenberg HH. Cyclic variations in

    white cell subpopulations in the human menstrual cycle: correlations with progesterone and

    estradiol. Clin Immunol Immunopathol 1979; 13(3): 246-53.

    42. Bouman A, Moes H, Heineman MJ, de Leij LF, Faas MM. The immune response during

    the luteal phase of the ovarian cycle: increasing sensitivity of human monocytes to endotoxin.

    Fertil Steril 2001; 76(3): 555-9.

    43. Elenkov IJ, Wilder RL, Bakalov VK, Link AA, Dimitrov MA, Fisher S et al. IL-12,

    TNF-alpha, and hormonal changes during late pregnancy and early postpartum: implications for

    autoimmune disease activity during these times. J Clin Endocrinol Metab 2001; 86(10): 4933-8.

    20 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    21/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    44. Apseloff G, Bao X, LaBoy-Goral L, Friedman H, Shah A. Practical considerations

    regarding the influence of the menstrual cycle on leukocyte parameters in clinical trials. Am J

    Ther 2000; 7(5): 297-302.

    45. Northern AL, Rutter SM, Peterson CM. Cyclic changes in the concentrations of

    peripheral blood immune cells during the normal menstrual cycle. Proc Soc Exp Biol Med 1994;207(1): 81-8.

    46. Faas M, Bouman A, Moes H, Heineman MJ, de Leij L, Schuiling G. The immune

    response during the luteal phase of the ovarian cycle: a Th2-type response? Fertil Steril 2000;

    74(5): 1008-13.

    47. Veenstra van Nieuwenhoven AL, Bouman A, Moes H, Heineman MJ, de Leij LF,

    Santema J, Faas MM. Cytokine production in natural killer cells and lymphocytes in pregnant

    women compared with women in the follicular phase of the ovarian cycle. Fertil Steril 2002;

    77(5): 1032-7.

    48. Bouman A, Schipper M, Heineman MJ, Faas MM. Gender difference in the non-specific

    and specific immune response in humans. Am J Reprod Immunol 2004; 52(1): 19-26.49. Kumru S, Godekmerdan A, Yilmaz B. Immune effects of surgical menopause and

    estrogen replacement therapy in peri-menopausal women. J Reprod Immunol 2004; 63(1):

    31-8.

    50. Burleson MH, Malarkey WB, Cacioppo JT, Poehlmann KM, Kiecolt-Glaser JK, Berntson

    GG, Glaser R. Postmenopausal hormone replacement: effects on autonomic, neuroendocrine,

    and immune reactivity to brief psychological stressors. Psychosom Med 1998; 60(1): 17-25.

    51. Auerbach L, Hafner T, Huber JC, Panzer S. Influence of low-dose oral contraception on

    peripheral blood lymphocyte subsets at particular phases of the hormonal cycle. Fertil Steril

    2002; 78(1): 83-9.52. Lopez-Karpovitchs X, Larrea F, Cardenas R, Valencia X, Piedras J, Diaz-Sanchez V,

    Alarcon-Segovia D. Peripheral blood lymphocyte subsets and serum immunoglobulins in

    Sheehan's syndrome and in normal women during the menstrual cycle. Rev Invest Clin 1993;

    45(3): 247-53.

    53. Yang JH, Chen CD, Wu MY, Chao KH, Yang YS, Ho HN. Hormone replacement therapy

    reverses the decrease in natural killer cytotoxicity but does not reverse the decreases in the

    T-cell subpopulation or interferon-gamma production in postmenopausal women. Fertil Steril

    2000; 74(2): 261-7.

    54. Giglio T, Imro MA, Filaci G, Scudeletti M, Puppo F, De Cecco L et al. Immune cell

    circulating subsets are affected by gonadal function. Life Sci 1994; 54(18): 1305-12.55. Gronroos M, Eskola J. In vitro functions of lymphocytes during high-dose

    medroxyprogesterone acetate (MPA) treatment. Cancer Immunol Immunother 1984; 17(3):

    218-20.

    56. Porter VR, Greendale GA, Schocken M, Zhu X, Effros RB. Immune effects of hormone

    replacement therapy in post-menopausal women. Exp Gerontol 2001; 36(2): 311-26.

    57. Prieto GA, Rosenstein Y. Oestradiol potentiates the suppressive function of human CD4

    CD25 regulatory T cells by promoting their proliferation. Immunology 2006; 118(1): 58-65.

    58. Tai P, Wang J, Jin H, Song X, Yan J, Kang Y et al. Induction of regulatory T cells by

    physiological level estrogen. J Cell Physiol 2008; 214(2): 456-64.59. Page ST, Plymate SR, Bremner WJ, Matsumoto AM, Hess DL, Lin DW et al. Effect of

    21 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    22/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    medical castration on CD4+ CD25+ T cells, CD8+ T cell IFN-gamma expression, and NK cells:

    a physiological role for testosterone and/or its metabolites. Am J Physiol Endocrinol Metab

    2006; 290(5): E856-E863.

    60. Fries LF, Brickman CM, Frank MM. Monocyte receptors for the Fc portion of IgG

    increase in number in autoimmune hemolytic anemia and other hemolytic states and aredecreased by glucocorticoid therapy. J Immunol 1983; 131(3): 1240-5.

    61. Schreiber AD, Chien P, Tomaski A, Cines DB. Effect of danazol in immune

    thrombocytopenic purpura. N Engl J Med 1987; 316(9): 503-8.

    62. Friedman D, Netti F, Schreiber AD. Effect of estradiol and steroid analogues on the

    clearance of immunoglobulin G-coated erythrocytes. J Clin Invest 1985; 75(1): 162-7.

    63. Beutler BA, Milsark IW, Cerami A. Cachectin/tumor necrosis factor: production,

    distribution, and metabolic fate in vivo. J Immunol 1985; 135(6): 3972-7.

    64. Asai K, Hiki N, Mimura Y, Ogawa T, Unou K, Kaminishi M. Gender differences in

    cytokine secretion by human peripheral blood mononuclear cells: role of estrogen in modulating

    LPS-induced cytokine secretion in an ex vivo septic model. Shock 2001; 16(5): 340-3.65. Schwarz E, Schafer C, Bode JC, Bode C. Influence of the menstrual cycle on the

    LPS-induced cytokine response of monocytes. Cytokine 2000; 12(4): 413-6.

    66. Posma E, Moes H, Heineman MJ, Faas M. The effect of testosterone on cytokine

    production in the specific and non-specific immune response. Am J Reprod Immunol 2004; 52:

    237-43.

    67. Brannstrom M, Friden BE, Jasper M, Norman RJ. Variations in peripheral blood levels of

    immunoreactive tumor necrosis factor alpha (TNFalpha) throughout the menstrual cycle and

    secretion of TNFalpha from the human corpus luteum. Eur J Obstet Gynecol Reprod Biol 1999;

    83(2): 213-7.

    68. Bouman A, Schipper M, Heineman MJ, Faas M. 17b-estradiol and progesterone do notinfluence the production of cytokine from lipopolysaccharide-stimulated monocytes in humans.

    Fertil Steril 2004; 82 (suppl 3): 1212-9.

    69. Rogers A, Eastell R. Effects of estrogen therapy of postmenopausal women on

    cytokines measured in peripheral blood. J Bone Miner Res 1998; 13(10): 1577-86.

    70. Denison FC, Kelly RW, Calder AA. Differential secretion of chemokines from peripheral

    blood in pregnant compared with non-pregnant women. J Reprod Immunol 1997; 34(3): 225-40.

    71. Parker MG. Transcriptional activation by oestrogen receptors. Biochem Soc Symp 1998;

    63: 45-50.

    72. Rogers A, Eastell R. The effect of 17beta-estradiol on production of cytokines in culturesof peripheral blood. Bone 2001; 29(1): 30-4.

    73. Ralston SH, Russell RG, Gowen M. Estrogen inhibits release of tumor necrosis factor

    from peripheral blood mononuclear cells in postmenopausal women. J Bone Miner Res 1990;

    5(9): 983-8.

    74. Cannon JG, Dinarello CA. Increased plasma interleukin-1 activity in women after

    ovulation. Science 1985; 227(4691): 1247-9.

    75. Polan ML, Loukides JA, Honig J. Interleukin-1 in human ovarian cells and in peripheral

    blood monocytes increases during the luteal phase: evidence for a midcycle surge in the

    human. Am J Obstet Gynecol 1994; 170(4): 1000-6.

    76. Morishita M, Miyagi M, Iwamoto Y. Effects of sex hormones on production ofinterleukin-1 by human peripheral monocytes. J Periodontol 1999; 70(7): 757-60.

    22 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    23/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    77. Trinchieri G. Interleukin-12: a proinflammatory cytokine with immunoregulatory functions

    that bridge innate resistance and antigen-specific adaptive immunity. Annu Rev Immunol 1995;

    13: 251-76.

    78. Matalka KZ. The effect of estradiol, but not progesterone, on the production of cytokines

    in stimulated whole blood, is concentration-dependent. Neuro Endocrinol Lett 2003; 24(3-4):185-91.

    79. McKane WR, Khosla S, Peterson JM, Egan K, Riggs BL. Circulating levels of cytokines

    that modulate bone resorption: effects of age and menopause in women. J Bone Miner Res

    1994; 9(8): 1313-8.

    80. Kania DM, Binkley N, Checovich M, Havighurst T, Schilling M, Ershler WB. Elevated

    plasma levels of interleukin-6 in postmenopausal women do not correlate with bone density. J

    Am Geriatr Soc 1995; 43(3): 236-9.

    81. Rachon D, Mysliwska J, Suchecka-Rachon K, Wieckiewicz J, Mysliwski A. Effects of

    oestrogen deprivation on interleukin-6 production by peripheral blood mononuclear cells of

    postmenopausal women. J Endocrinol 2002; 172(2): 387-95.82. Cioffi M, Esposito K, Vietri MT, Gazzerro P, D'Auria A, Ardovino I et al. Cytokine pattern

    in postmenopause. Maturitas 2002; 41(3): 187-92.

    83. Straub RH, Hense HW, Andus T, Scholmerich J, Riegger GA, Schunkert H. Hormone

    replacement therapy and interrelation between serum interleukin-6 and body mass index in

    postmenopausal women: a population-based study. J Clin Endocrinol Metab 2000; 85(3):

    1340-4.

    84. Jilma B, Dirnberger E, Loscher I, Rumplmayr A, Hildebrandt J, Eichler HG et al.

    Menstrual cycle-associated changes in blood levels of interleukin-6, alpha1 acid glycoprotein,

    and C-reactive protein. J Lab Clin Med 1997; 130(1): 69-75.

    85. Al Harthi L, Wright DJ, Anderson D, Cohen M, Matity Ahu D, Cohn J et al. The impact ofthe ovulatory cycle on cytokine production: evaluation of systemic, cervicovaginal, and salivary

    compartments. J Interferon Cytokine Res 2000; 20(8): 719-24.

    86. Abrahamsen B, Stilgren LS, Rettmer E, Bonnevie-Nielsen V, Beck-Nielsen H. Effects of

    the natural and artificial menstrual cycle on the production of osteoprotegerin and the bone

    resorptive cytokines IL-1beta and IL-6. Calcif Tissue Int 2003; 72(1): 18-23.

    87. Verthelyi D, Klinman DM. Sex hormone levels correlate with the activity of

    cytokine-secreting cells in vivo. Immunology 2000; 100(3): 384-90.

    88. Konecna L, Yan MS, Miller LE, Scholmerich J, Falk W, Straub RH. Modulation of IL-6

    production during the menstrual cycle in vivo and in vitro. Brain Behav Immun 2000; 14(1):

    49-61.89. Angstwurm MW, Gartner R, Ziegler-Heitbrock HW. Cyclic plasma IL-6 levels during

    normal menstrual cycle. Cytokine 1997; 9(5): 370-4.

    90. Berg G, Ekerfelt C, Hammar M, Lindgren R, Matthiesen L, Ernerudh J. Cytokine

    changes in postmenopausal women treated with estrogens: a placebo-controlled study. Am J

    Reprod Immunol 2002; 48(2): 63-9.

    91. Brooks-Asplund EM, Tupper CE, Daun JM, Kenney WL, Cannon JG. Hormonal

    modulation of interleukin-6, tumor necrosis factor and associated receptor secretion in

    postmenopausal women. Cytokine 2002; 19(4): 193-200.

    92. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol

    2005; 5(12): 953-64.93. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization.

    23 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    24/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    Front Biosci 2008; 13: 453-61.

    94. Rettew JA, Huet YM, Marriott I. Estrogens augment cell surface TLR4 expression on

    murine macrophages and regulate sepsis susceptibility in vivo. Endocrinology 2009; 150(8):

    3877-84.

    95. Lezama-Davila CM, Isaac-Marquez AP, Barbi J, Cummings HE, Lu B, Satoskar AR.Role of phosphatidylinositol-3-kinase-gamma (PI3Kgamma)-mediated pathway in

    17beta-estradiol-induced killing of L. mexicana in macrophages from C57BL/6 mice. Immunol

    Cell Biol 2008; 86(6): 539-43.

    96. Calippe B, Douin-Echinard V, Laffargue M, Laurell H, Rana-Poussine V, Pipy B et al.

    Chronic estradiol administration in vivo promotes the proinflammatory response of

    macrophages to TLR4 activation: involvement of the phosphatidylinositol 3-kinase pathway. J

    Immunol 2008; 180(12): 7980-8.

    97. Lezama-Davila CM, Isaac-Marquez AP, Barbi J, Oghumu S, Satoskar AR.

    17Beta-estradiol increases Leishmania mexicana killing in macrophages from DBA/2 mice by

    enhancing production of nitric oxide but not pro-inflammatory cytokines. Am J Trop Med Hyg2007; 76(6): 1125-7.

    98. Azenabor AA, Yang S, Job G, Adedokun OO. Expression of iNOS gene in macrophages

    stimulated with 17beta-estradiol is regulated by free intracellular Ca2+. Biochem Cell Biol 2004;

    82(3): 381-90.

    99. Hu SK, Mitcho YL, Rath NC. Effect of estradiol on interleukin 1 synthesis by

    macrophages. Int J Immunopharmacol 1988; 10(3): 247-52.

    100. Salem ML. Estrogen, a double-edged sword: modulation of TH1- and TH2-mediated

    inflammations by differential regulation of TH1/TH2 cytokine production. Curr Drug Targets

    Inflamm Allergy 2004; 3(1): 97-104.

    101. Miller L, Alley EW, Murphy WJ, Russell SW, Hunt JS. Progesterone inhibits induciblenitric oxide synthase gene expression and nitric oxide production in murine macrophages. J

    Leukoc Biol 1996; 59(3): 442-50.

    102. Su L, Sun Y, Ma F, Lu P, Huang H, Zhou J. Progesterone inhibits Toll-like receptor

    4-mediated innate immune response in macrophages by suppressing NF-kappaB activation and

    enhancing SOCS1 expression. Immunol Lett 2009; 125(2): 151-5.

    103. Jones LA, Anthony JP, Henriquez FL, Lyons RE, Nickdel MB, Carter KC et al. Toll-like

    receptor-4-mediated macrophage activation is differentially regulated by progesterone via the

    glucocorticoid and progesterone receptors. Immunology 2008; 125(1): 59-69.

    104. Miller L, Hunt JS. Regulation of TNF-alpha production in activated mouse

    macrophages by progesterone. J Immunol 1998; 160(10): 5098-104.105. Rettew JA, Huet-Hudson YM, Marriott I. Testosterone reduces macrophage expression

    in the mouse of toll-like receptor 4, a trigger for inflammation and innate immunity. Biol Reprod

    2008; 78(3): 432-7.

    106. Mohan PF, Jacobson MS. Inhibition of macrophage superoxide generation by

    dehydroepiandrosterone. Am J Med Sci 1993; 306(1): 10-5.

    107. Rom WN, Harkin T. Dehydroepiandrosterone inhibits the spontaneous release of

    superoxide radical by alveolar macrophages in vitro in asbestosis. Environ Res 1991; 55(2):

    145-56.

    108. Falagas ME, Mourtzoukou EG, Vardakas KZ. Sex differences in the incidence and

    severity of respiratory tract infections. Respir Med 2007; 101(9): 1845-63.109. Klein SL. Hormonal and immunological mechanisms mediating sex differences in

    24 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    25/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    parasite infection. Parasite Immunol 2004; 26(6-7): 247-64.

    110. Beery TA. Sex differences in infection and sepsis. Crit Care Nurs Clin North Am 2003;

    15(1): 55-62.

    111. Routley CE, Ashcroft GS. Effect of estrogen and progesterone on macrophage

    activation during wound healing. Wound Repair Regen 2009; 17(1): 42-50.112. Frisancho-Kiss S, Coronado MJ, Frisancho JA, Lau VM, Rose NR, Klein SL,

    Fairweather D. Gonadectomy of male BALB/c mice increases Tim-3(+) alternatively activated

    M2 macrophages, Tim-3(+) T cells, Th2 cells and Treg in the heart during acute

    coxsackievirus-induced myocarditis. Brain Behav Immun 2009; 23(5): 649-57.

    113. Angele MK, Knoferl MW, Schwacha MG, Ayala A, Cioffi WG, Bland KI, Chaudry IH.

    Sex steroids regulate pro- and anti-inflammatory cytokine release by macrophages after

    trauma-hemorrhage. Am J Physiol 1999; 277(1 Pt 1): C35-C42.

    114. Jacobs B, Wuttke M, Papewalis C, Seissler J, Schott M. Dendritic cell subtypes and in

    vitro generation of dendritic cells. Horm Metab Res 2008; 40(2): 99-107.

    115. Huck B, Steck T, Habersack M, Dietl J, Kammerer U. Pregnancy associated hormonesmodulate the cytokine production but not the phenotype of PBMC-derived human dendritic cells.

    Eur J Obstet Gynecol Reprod Biol 2005; 122(1): 85-94.

    116. Kyurkchiev D, Ivanova-Todorova E, Hayrabedyan S, Altankova I, Kyurkchiev S.

    Female sex steroid hormones modify some regulatory properties of monocyte-derived dendritic

    cells. Am J Reprod Immunol 2007; 58(5): 425-33.

    117. Hughes GC, Thomas S, Li C, Kaja MK, Clark EA. Cutting edge: progesterone regulates

    IFN-alpha production by plasmacytoid dendritic cells. J Immunol 2008; 180(4): 2029-33.

    118. Hickey MJ, Kubes P. Intravascular immunity: the host-pathogen encounter in blood

    vessels. Nat Rev Immunol 2009; 9(5): 364-75.119. Miyagi M, Aoyama H, Morishita M, Iwamoto Y. Effects of sex hormones on chemotaxis

    of human peripheral polymorphonuclear leukocytes and monocytes. J Periodontol 1992; 63(1):

    28-32.

    120. Molloy EJ, O'Neill AJ, Grantham JJ, Sheridan-Pereira M, Fitzpatrick JM, Webb DW,

    Watson RW. Sex-specific alterations in neutrophil apoptosis: the role of estradiol and

    progesterone. Blood 2003; 102(7): 2653-9.

    121. Bekesi G, Kakucs R, Varbiro S, Racz K, Sprintz D, Feher J, Szekacs B. In vitro effects

    of different steroid hormones on superoxide anion production of human neutrophil granulocytes.

    Steroids 2000; 65(12): 889-94.

    122. Cassidy RA. Influence of steroids on oxidant generation in activated humangranulocytes and mononuclear leukocytes. Shock 2003; 20(1): 85-90.

    123. Kubes P, Suzuki M, Granger DN. Nitric oxide: an endogenous modulator of leukocyte

    adhesion. Proc Natl Acad Sci U S A 1991; 88(11): 4651-5.

    124. Garcia-Duran M, de Frutos T, Diaz-Recasens J, Garcia-Galvez G, Jimenez A, Monton

    M et al. Estrogen stimulates neuronal nitric oxide synthase protein expression in human

    neutrophils. Circ Res 1999; 85(11): 1020-6.

    125. Stefano GB, Prevot V, Beauvillain JC, Fimiani C, Welters I, Cadet P et al. Estradiol

    coupling to human monocyte nitric oxide release is dependent on intracellular calcium

    transients: evidence for an estrogen surface receptor. J Immunol 1999; 163(7): 3758-63.

    126. Yovel G, Shakhar K, Ben Eliyahu S. The effects of sex, menstrual cycle, and oral

    25 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    26/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    contraceptives on the number and activity of natural killer cells. Gynecol Oncol 2001; 81(2):

    254-62.

    127. Souza SS, Castro FA, Mendonca HC, Palma PV, Morais FR, Ferriani RA, Voltarelli JC.

    Influence of menstrual cycle on NK activity. J Reprod Immunol 2001; 50(2): 151-9.

    128. Baker DA, Hameed C, Tejani N, Milch P, Thomas J, Monheit AG, Dattwyler RJ.Lymphocyte subsets in women on low dose oral contraceptives. Contraception 1985; 32(4):

    377-82.

    129. Scanlan JM, Werner JJ, Legg RL, Laudenslager ML. Natural killer cell activity is

    reduced in association with oral contraceptive use. Psychoneuroendocrinology 1995; 20(3):

    281-7.

    130. Stopinska-Gluszak U, Waligora J, Grzela T, Gluszak M, Jozwiak J, Radomski D et al.

    Effect of estrogen/progesterone hormone replacement therapy on natural killer cell cytotoxicity

    and immunoregulatory cytokine release by peripheral blood mononuclear cells of

    postmenopausal women. J Reprod Immunol 2006; 69(1): 65-75.

    131. Ferguson MM, McDonald FG. Oestrogen as an inhibitor of human NK cell cytolysis.FEBS Lett 1985; 191(1): 145-8.

    132. Sulke AN, Jones DB, Wood PJ. Hormonal modulation of human natural killer cell

    activity in vitro. J Reprod Immunol 1985; 7(2): 105-10.

    133. Uksila J. Human NK cell activity is not inhibited by pregnancy and cord serum factors

    and female steroid hormones in vitro. J Reprod Immunol 1985; 7(2): 111-20.

    134. Sulke AN, Jones DB, Wood PJ. Variation in natural killer activity in peripheral blood

    during the menstrual cycle. Br Med J (Clin Res Ed) 1985; 290(6472): 884-6.

    135. Thyss A, Caldani C, Bourcier C, Benita G, Schneider M. Comparison of natural killer

    activity during the first and second halves of the menstrual cycle in women. Br J Cancer 1984;

    50(1): 127-8.136. White D, Jones DB, Cooke T, Kirkham N. Natural killer (NK) activity in peripheral blood

    lymphocytes of patients with benign and malignant breast disease. Br J Cancer 1982; 46(4):

    611-6.

    137. Mendes R, Bromelow KV, Westby M, Galea-Lauri J, Smith IE, O'Brien ME,

    Souberbielle BE. Flow cytometric visualisation of cytokine production by CD3-CD56+ NK cells

    and CD3+CD56+ NK-T cells in whole blood. Cytometry 2000; 39(1): 72-8.

    138. Biassoni R, Ferrini S, Prigione I, Pelak VS, Sekaly RP, Long EO. Activated CD3-

    CD16+ natural killer cells express a subset of the lymphokine genes induced in activated alpha

    beta + and gamma delta + T cells. Scand J Immunol 1991; 33(3): 247-52.

    139. Bouman A, Moes H, Heineman MJ, de Leij LF, Faas MM. Cytokine production bynatural killer lymphocytes in follicular and luteal phase of the ovarian cycle in humans. Am J

    Reprod Immunol 2001; 45(3): 130-4.

    140. Klein SL. The effects of hormones on sex differences in infection: from genes to

    behavior. Neurosci Biobehav Rev 2000; 24(6): 627-38.

    141. Roberts CW, Walker W, Alexander J. Sex-associated hormones and immunity to

    protozoan parasites. Clin Microbiol Rev 2001; 14(3): 476-88.

    142. Marriott I, Huet-Hudson YM. Sexual dimorphism in innate immune responses to

    infectious organisms. Immunol Res 2006; 34(3): 177-92.

    143. NICOL T, BILBEY DL, CHARLES LM, CORDINGLEY JL, VERNON-ROBERTS B.

    OESTROGEN: THE NATURAL STIMULANT OF BODY DEFENCE. J Endocrinol 1964; 30:277-91.

    26 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    27/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    144. Marriott I, Huet-Hudson YM. Sexual dimorphism in innate immune responses to

    infectious organisms. Immunol Res 2006; 34(3): 177-92.

    145. Beagley KW, Gockel CM. Regulation of innate and adaptive immunity by the female

    sex hormones oestradiol and progesterone. FEMS Immunol Med Microbiol 2003; 38(1): 13-22.

    146. Choudhry MA, Bland KI, Chaudry IH. Trauma and immune response--effect of gender

    differences. Injury 2007; 38(12): 1382-91.

    147. Gilliver SC, Ruckshanthi JP, Hardman MJ, Nakayama T, Ashcroft GS. Sex dimorphism

    in wound healing: the roles of sex steroids and macrophage migration inhibitory factor.

    Endocrinology 2008; 149(11): 5747-57.

    148. Herrick S, Ashcroft G, Ireland G, Horan M, McCollum C, Ferguson M. Up-regulation of

    elastase in acute wounds of healthy aged humans and chronic venous leg ulcers are associated

    with matrix degradation. Lab Invest 1997; 77(3): 281-8.

    149. Ashcroft GS, Horan MA, Ferguson MW. Aging alters the inflammatory and endothelial

    cell adhesion molecule profiles during human cutaneous wound healing. Lab Invest 1998; 78(1):47-58.

    150. Ashcroft GS, Greenwell-Wild T, Horan MA, Wahl SM, Ferguson MW. Topical estrogen

    accelerates cutaneous wound healing in aged humans associated with an altered inflammatory

    response. Am J Pathol 1999; 155(4): 1137-46.

    151. Ashcroft GS, Horan MA, Ferguson MW. Aging alters the inflammatory and endothelial

    cell adhesion molecule profiles during human cutaneous wound healing. Lab Invest 1998; 78(1):

    47-58.

    152. Gilliver SC, Wu F, Ashcroft GS. Regulatory roles of androgens in cutaneous wound

    healing. Thromb Haemost 2003; 90(6): 978-85.

    153. Deonarine K, Panelli MC, Stashower ME, Jin P, Smith K, Slade HB et al. Geneexpression profiling of cutaneous wound healing. J Transl Med 2007; 5: 11.

    154. Leibovich SJ, Ross R. The role of the macrophage in wound repair. A study with

    hydrocortisone and antimacrophage serum. Am J Pathol 1975; 78(1): 71-100.

    155. Grossman CJ. Regulation of the immune system by sex steroids. Endocr Rev 1984;

    5(3): 435-55.

    156. Mendelsohn J, Multer MM, Bernheim JL. Inhibition of human lymphocyte stimulation by

    steroid hormones: cytokinetic mechanisms. Clin Exp Immunol 1977; 27(1): 127-34.

    157. Wyle FA, Kent JR. Immunosuppression by sex steroid hormones. The effect upon

    PHA- and PPD-stimulated lymphocytes. Clin Exp Immunol 1977; 27(3): 407-15.

    158. Mori T, Kobayashi H, Nishimura T, Mori TS, Fujii G, Inou T. Inhibitory effect ofprogesterone on the phytohaemagglutinin- induced transformation of human peripheral

    lymphocytes. Immunol Commun 1975; 4(6): 519-27.

    159. Van Voorhis BJ, Anderson DJ, Hill JA. The effects of RU 486 on immune function and

    steroid-induced immunosuppression in vitro. J Clin Endocrinol Metab 1989; 69(6): 1195-9.

    160. McMurray RW, Suwannaroj S, Ndebele K, Jenkins JK. Differential effects of sex

    steroids on T and B cells: modulation of cell cycle phase distribution, apoptosis and bcl-2

    protein levels. Pathobiology 2001; 69(1): 44-58.

    161. White HD, Crassi KM, Givan AL, Stern JE, Gonzalez JL, Memoli VA et al. CD3+ CD8+

    CTL activity within the human female reproductive tract: influence of stage of the menstrualcycle and menopause. J Immunol 1997; 158(6): 3017-27.

    27 / 32

  • 7/28/2019 Hormonas Sexuales y Respuesta Inmune

    28/32

    Sex Hormones and Immunoregulation

    Written by MM Faas, P de Vos & BN Melgert

    Tuesday, 12 July 2011 00:00

    162. Takao T, Kumagai C, Hisakawa N, Matsumoto R, Hashimoto K. Effect of

    17beta-estradiol on tumor necrosis factor-alpha-induced cytotoxicity in the human peripheral T

    lymphocytes. J Endocrinol 2005; 184(1): 191-7.

    163. Giron-Gonzalez JA, Moral FJ, Elvira J, Garcia-Gil D, Guerrero F, Gavilan I, Escobar L.

    Consistent production of a higher TH1:TH2 cytokine ratio by stimulated T cells in mencompared with women. Eur J Endocrinol 2000; 143(1): 31-6.

    164. Agarwal SK, Marshall GD, Jr. Perimenstrual alterations in type-1/type-2 cytokine

    balance of normal women. Ann Allergy Asthma Immunol 1999; 83(3): 222-8.

    165. Piccinni MP, Giudizi MG, Biagiotti R, Beloni L, Giannarini L, Sampognaro S et al.

    Progesterone favors the development of human T helper cells producing Th2-type cytokines

    and promotes both IL-4 production and membrane CD30 expression in established Th1 cell

    clones. J Immunol 1995; 155(1): 128-33.

    166. Cutolo M, Sulli A, Capellino S, Villaggio B, Montagna P, Seriolo B, Straub RH. Sex

    hormones influence on the immune system: basic and clinical aspects in autoimmunity. Lupus

    2004; 13(9): 635-8.167. Cioffi M, Esposito K, Vietri MT, Gazzerro P, D'Auria A, Ardovino I et al. Cytokine

    pattern in postmenopause. Maturitas 2002; 41(3): 187-92.

    168. Kamada M, Irahara M, Maegawa M, Ohmoto Y, Murata K, Yasui T et al. Transient

    increase in the levels of T-helper 1 cytokines in postmenopausal women and the effects of

    hormone replacement therapy. Gynecol Obstet Invest 2001; 52(2): 82-8.

    169. Maskill JK, Laird SM, Okon M, Li TC, Blakemore AI. Stability of serum interleukin-10

    levels during the menstrual cycle. Am J Reprod Immunol 1997; 38(5): 339-42.

    170. Nakashima A, Ito M, Yoneda S, Shiozaki A, Hidaka T, Saito S. Circulating and decidual

    Th17 cell levels in healthy pregnancy. Am J Reprod Immunol 2010; 63(2): 104-9.

    171. Tai P, Wang J, Jin H, Song X, Yan J, Kang Y et al. Induction of regulatory T cells byphysiological level estrogen. J Cell Physiol 2008; 214(2): 456-64.

    172. Polanczyk MJ, Carson BD, Subra