VIEWS AND REVIEWS 1 2 3 4 5 6 7 8 9 10 a b Q5 Kassie J. Hyde, M.D. and Danny J. Schust, M.D. 11 b Department of Obstetrics, Gynecology and Women's Health; a University of Missouri School of Medicine, Columbia, 12 Missouri 13 14 15 16 Characterization of the implanting human fetus as an allograft prompted a field of research in reproductive immunology that continues 17 to fascinate and perplex scientists. Paternal- or partner-derived alloantigens are present in the maternal host at multiple times during 18 the reproductive process. They begin with exposure to semen, continue through implantation and placentation, and may persist for 19 decades in the form of fetal microchimerism. Changes in maternal immune responses that allow allogenic fertilization and survival of semiallogenic concepti to delivery must be balanced with a continued need to respond appropriately to pathogenic invaders, com20 mensals, cell or tissue damage, and any tendency toward malignant transformation. This complex and sophisticated balancing act is 21 essential for survival of mother, fetus, and the species itself. We will discuss concepts of alloimmune recognition, tolerance, and igno22 rance as they pertain to mammalian reproduction with a focus on human reproduction, maternal immune modulation, and the very 23 earliest events in the reproductive process, fertilization and implantation. (Fertil SterilÒ 2016;-:-–-. Ó2016 by American Society 24 for Reproductive Medicine.) 25 Key Words: Reproductive immunology, implantation, placenta, fertilization, allograft 26 Discuss: You can discuss this article with its authors and with other ASRM members at 27 28 29 30 31 32 regnancy poses perhaps one of derived (fetal) antigens on the leadnanoparticles and microparticles, 33 the most unique and chaling edge syncytiotrophoblast may and exosomes into the maternal cir34 lenging tasks for the immune be accessible during initial embryo culation, [b] soluble forms of the 35 system. The female reproductive tract implantation; [3] at the surface of nonclassic major histocompatibility 36 must balance protection from pathothe syncytiotrophoblast layer of the complex (MHC) class I molecule 37 genic infections and inflammatory villous placenta, which is bathed in HLA-G are secreted into the maternal 38 insults with acceptance of specific maternal blood; [4] within the circulation, and [c] fetal cells traffic 39 foreign antigens to support reprodecidua of the more mature, after iminto the maternal circulatory system 40 ductive function. There are several plantation, placenta when invading where they can remain in tissues 41 sites and times at which the maternal extravillous trophoblast cells interact for decades (fetal microchimerism). 42 immune system may be challenged with decidual immune cells as they In the absence of an appropriate 43 with fetal/paternal/partner alloantitraverse this tissue to reach the inner response in female immunoregula44 gens during the course of successful third of the myometrium (in tory and immunosuppressive activ45 reproduction. These include: [1] humans), remodel, and replace the ities to these non-self antigens, the 46 within the female genital tract when supporting cells of the maternal spisurvival of the human species would 47 partner/paternal antigens are introral arteries; and [5] more systemibe at risk. Given the enormity of this 48 duced by semen and sperm to allow cally within the mother when [a] responsibility, it comes as no surprise 49 fertilization; [2] within the early syncytiotrophoblast turnover at the that the mechanisms involved are 50 maternal decidua when paternally villous interface sheds microvesicles, complex and redundant (1). Like 51 other mucosal surfaces, the epithelial 52 components and subepithelial im53 Received May 6, 2016; revised and accepted July 13, 2016. mune cells of the female genital tract K.J.H. has nothing to disclose. D.J.S. has nothing to disclose. 54 serve as the initital barrier to and Reprint requests: Danny J. Schust, M.D., Department of Obstetrics, Gynecology and Women's Health, 55 University of Missouri School of Medicine, 500 North Keene Street, Suite 203, Columbia, Missurveyor of all incoming antigens. 56 souri 65201 (E-mail:
[email protected]). Immune responses at these sites 57 Fertility and Sterility® Vol. -, No. -, - 2016 0015-0282/$36.00 must reliably target pathogen58 Copyright ©2016 American Society for Reproductive Medicine, Published by Elsevier Inc. derived antigens for destruction, http://dx.doi.org/10.1016/j.fertnstert.2016.07.1073 59
The immunologic challenges of human reproduction: an evolving story
P
VOL. - NO. - / - 2016
1
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118
VIEWS AND REVIEWS 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177
yet, simultaneously tolerating antigens of commensal organisms, semen, and the fetal allograft (2). Endocrine and immunologic mechanisms work to alter the cellular, cytokine, and chemokine environments at mucosal interfaces within the female reproductive tract to promote tolerance of specific non-self antigens. Specific glycosylation patterns expressed on non-self cells, such as placental trophoblast cells or human gametes, may also play a specific role in immune regulation (1). These and several other mechanisms will be discussed in this article. At present, researchers in the field of reproductive immunology have concentrated the lion's share of their energies on immunologic changes surrounding acceptance of the fetal allograft. This focus was largely driven by the classic articles of Sir Peter Medawar and colleagues (3) in the early 1950s that proposed the immunologic conundrum of the fetal allograft in humans. For several years, studies in this area focused on immunologic ignorance as central to this process. It was hypothesized that the mother simply did not recognize her fetus as foreign and therefore did not mount an alloimmune response. Reports on the presence of circulating antipaternal antibodies in maternal sera suggested that mammalian females, including humans, do indeed recognize paternally derived antigens on their allogeneic concepti, at least during later stages of pregnancy when the implanting fetus has invaded into the maternal decidua (4–8). The overarching hypothesis for most of the pregnancy therefore changed from ignorance to tolerance of the fetus. The latter requires substantial maternal immune deviations during pregnancy. These alterations are not simply reductions in immune responsiveness, but are much more nuanced. They may also differ before and after implantation. In fact, although pregnancy has generally been considered a time of at least local immune suppression, several lines of evidence indicate that the local environment at the implantation site in humans is proinflammatory (9). These observations have been used to explain the beneficial effects of endometrial injury on implantation and pregnancy success in patients undergoing assisted reproduction (9, 10). Likewise, human pregnancy, particularly in the third trimester, has been characterized as a period of maternal systemic inflammation (11). This state appears to be to be magnified in women experiencing preeclampsia (11, 12) and may represent one aspect of preparation for labor in all women (13). The best characterization of the immune state in successful pregnancy, however, is neither inflammatory nor immunosuppressed but rather one of tightly controlled and complex immune modulation. The characteristics of this modulation likely differ during the course of pregnancy. Some may be quite pronounced locally but less so systemically. Some of these changes appear to proceed the time of implantation, with its resultant exposure of fetal antigens to the maternal immune system. Some can even be detected even before conception. Finally, characterization of pregnancy as a state of solely immune tolerance or immune ignorance of paternal alloantigen is almost certainly an oversimplification. Both are likely involved, in time- and location-specific manners.
IMMUNOLOGIC INTERACTIONS IN THE FEMALE REPRODUCTIVE TRACT: A FOCUS ON TOLERANCE The immune system can be divided into two discrete compartments: peripheral and mucosal. Most of the general immunologic principles have been best investigated and described for the peripheral immune system. During the past few decades, our understanding of the mucosal immune compartment has grown rapidly. Charged with the responsibility for defense against blood-borne pathogens, the peripheral immune system primarily consists of the spleen, lymph nodes, and peripheral blood. In contrast, as part of the innate defense system, the mucosal immune system serves to provide protection against pathogens that gain exposure to the body through the mucosal surfaces of the respiratory and gastrointestinal tracts, lacrimal ducts, mammary ducts, and genitourinary tract (14). This combined mucosal surface area of several hundred square meters protects from antigens in ingested food, inhaled particles, and resident microorganisms using a combination of T cells, B cells (including IgA-secreting plasma cells), natural killer (NK) cells, macrophages, and dendritic cells. Humoral factors, including lysozyme, lactoferrin, peroxidase, mucins, defensins, and digestive enzymes, also play key roles in defending the mucosal compartment (14, 15). Although the female reproductive tract is a component of the mucosal immune system, immunologic interactions at the surface of the female genital tract can differ from those manifested in the peripheral compartment and at other mucosal immune sites (16). Particularly notable in these cycling tissues is the strong hormonal control that dictates the transport of immunoglobulins, levels of cytokines, quality of antigen presentation, and distribution of certain immune cell populations. Hormonal contributions to immune regulation are particularly evident during pregnancy when systemic and local levels of pregnancy-related steroid and protein hormones are dramatically altered by the presence of the fetalplacental unit. Still, while supporting physiologic events, such as fertilization, implantation, pregnancy, and parturition, the mucosal immune system of the female genital tract must continue to protect against infectious pathogens and tolerate commensals (14). The female immune system undergoes constant transformation and adaptation beginning with each menstrual cycle and continuing through gestation in the event of fertilization. For example, compared with the mucosa of the gut or lung, the mucosal surface of the female reproductive tract requires more extreme MHC-related tolerance mechanisms that enable it to endure prolonged exposure to HLA-mismatched developing embryos (2). Data from animal and human studies indicate that immune cells at the maternal-to-fetal interface are selected and maintained in ways that differ from immune cells in the periphery as well as in other mucosal locations (17, 18). Furthermore, investigations have shown the existence of particular cellular homing mechanisms to recruit these specific cell populations to the reproductive tract (19, 20). Changes in immune cell populations and secretory profiles in the decidua that promote allograft
VOL. - NO. - / - 2016
2
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236
Fertility and Sterility® acceptance are not the only sites where one can document immune changes during the before and peri-implantation stages of pregnancy. The oviducts and uterus provide cytokines and soluble factors to promote blastocyst development, modulate embryo metabolic function and gene expression, and increase implantation success (21, 22). Among the many contributors to this localized immune regulation, some of the most important mechanisms include the presence of unique immune cell populations, alterations in T-cell profiles, and the immune effects of gestationassociated hormones.
Immune Cells Populating the Female Reproductive Tract The female genital tract contains at least five notable and unusual immune cell types that deserve specific mention: decidual granular lymphocytes, NKT cells, T-cell receptor (TCR)gdþ cells, decidual macrophages, and innate lymphoid cells (Fig. 1). During the menstrual cycle, resident cell populations of the human endometrium undergo dramatic changes. One remarkable immune cell type, called decidual granular lymphocytes, comprises most of the total endometrial lymphocyte population in the late luteal phase and in early pregnancy (18, 23–25). Often referred to as decidual NK
cells, decidual granular lymphocytes have been shown to exhibit a specific and distinct surface receptor repertoire. These allow regulation by MHC class I molecules and nonMHC molecules that differ from those recognized by NK cells in the periphery (26, 27). Furthermore, rather than having a primarily cytotoxic function typical of peripheral NK cells, these NK-like cells of the decidua largely serve to support trophoblast invasion, spiral artery remodeling, and vascular growth in the decidua through direct contact with fetally derived placental cells (28, 29). Decidual granular lymphocytes likely derive from endometrial NK cells rather than peripheral NK cells. Based on this understanding, it is unlikely that assessments of peripheral NK cells would lend direct information concerning the function or competency of their decidual counterparts (30). Immune cells that simultaneously exhibit characteristics of NK cells and T cells have been described in the peripheral immune system and within the endometrium and decidua of humans (31, 32). These NKT cells carry a phenotype that suggests innate immune function. Investigations focused on NKT cells at the implantation sites of murine pregnancies have demonstrated that their presence and expansion at this site is mediated by interactions with fetally expressed MHC class I or class I-like products derived from the paternal genome (33). Therefore, the large increase in decidual NKT
FIGURE 1 •Decidual granular lymphocytes •NKT cells •TCRγδ+ cells •Decidual macrophages •Innate lymphoid cells
PopulaƟon Trends
MHC Tolerance Mechanisms
•Th1 cells: Secre on of INF-y, IL-12, IL-2, TNF- β IL•Th •Th2: An body produc on and sec on of IL-10, IL-4, IL-5 secre •Tr •Treg: Secre on of IL-10 •Th •Th17: Secre on of IL-17
Unique Immune Cell PopulaƟons
Cytokine Secretory Profiles
T cell RegulaƟon
GestaƟonassociated Hormonal Control
LigaƟon of Specific Co-sƟmulatory Signals
Estrogen
Progesterone
Shi to Th2 environment
Increase in Th2-type responses, Th1-type cytokine produc on, and leukemia inhibitory factor
Decidual T cells
print & web 4C=FPO
237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295
Increased Treg and Th17 ac vity during ovula on and fer liza on
Shi to Th2- dominant environment during luteal phase of menstrual cycle
• Expression of CD28, CDL4, PD1, and CTLA-4
MHC class I HLA-G, HLA-E, and HLA-C expression by extravillous trophoblast cells
Surrounding pregnancyrelated cells • Expression of B7-1 and B7-2 on decidual dendri c cells • Expression of B7-H1 and B7H2 by trophoblast cells • Reversed B7-1/2 signaling by decidual stromal cells
Recruitment, enhanced func on and prolifera on of Treg cells
Decrease in CD8+ prolifera on and cytokine secre on Sustaining and expanding Treg cells
Human Chorionic Gonadotrophin
Decrease in IL-17
Increase in IL-27 and IL10
Chemoa ractant for Treg cells
Immune adaptations in the human deciduas. Hyde. Immunologic challenges in reproduction. Fertil Steril 2016.
VOL. - NO. - / - 2016
3
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354
VIEWS AND REVIEWS 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413
cells that occurs during pregnancy may represent a mechanism through which the fetus modulates the maternal immune response to induce tolerance. In addition, because these NKT cells have been shown to secrete much interleukin (IL)-4 (see later section on Cytokine secretion profiles), they have been also implicated in the formation of the Th2-dominant environment necessary for successful gestation (34). The peripheral immune compartment and the human reproductive tract contain T cells bearing a TCR comprised of a ab heterodimer (TCRabþ). However, unlike the peripheral compartment, the female genital tract also contains a subset of T cells with a distinctive TCR comprised of a gd heterodimer (TCRgdþ cells), whose population in the decidua increases during early pregnancy (35–37). Unlike their TCRabþ counterparts, TCRgdþ cells may serve in the recognition of non-MHC-restricted antigens as well as support trophoblast invasion and survival (38, 39). Decidual macrophages are responsible for an array of functions at the maternal-to-fetal interface. However, unlike macrophages present in other tissues, these immune cells exhibit a predominately immunosuppressive phenotype, designated M2 polarization, that is characterized by increased IL-10 production and indoleamine 2, 3-dioxygenase activity. Indoleamine 2, 3-dioxygenase catabolizes tryptophan, an amino acid necessary for T-cell activation and proliferation. Before embryo attachment, these decidual macrophages produce leukocyte inhibitory factor and IL-1b, which may help the endometrium become receptive to implantation by regulation of surface glycan structures on epithelial cells (40). In addition, these macrophages play a role in uterine vasculature transformation and fetal immune tolerance (41). One recently described group of immune cells, called innate lymphoid cells (ILCs) (42, 43) may soon be recognized as particularly relevant for our understanding of reproductive immunology. Like other innate immune cells, these cytokine-secreting cells are antigen-independent and have been subclassified based on their cytokine secretion patterns. Group 1 ILCs produce type 1 cytokines, group 2 ILCs produce type 2 cytokines, and group 3 ILCs produce Th17associated cytokines. In the periphery, group 1 ILCs include NK cells. The presence and function of ILCs in the decidua is still unclear but there is a growing body of literature that aims to better define these cells (44). The B lymphocytes can present antigen and secrete cytokines, although they are most typically defined by their ability to produce antibodies as a central component of humoral immunity. There are very few B cells present in the human decidua (14) and much less is known about their role at the site of implantation when compared with T cells, dendritic cells, or macrophages. Data from murine studies have demonstrated a complex and nuanced pregnancy-specific, partial deletion of maternal B cells directed against paternally expressed antigens, which may aid in tolerance of the fetal allograft without simultaneous decrement in pathogen surveillance (45, 46). Similar, but much less marked, numerical changes have been documented in B lymphocyte in the latter parts of human pregnancy, although these may not translate into functional changes (47, 48). Still, the
importance of humoral immunity in human pregnancy maintenance should not be dismissed, as one of the only widely accepted causes of early recurrent pregnancy loss in humans is the autoimmune disease named for the autoantibodies that define its diagnosis: the antiphospholipid syndrome (14). Although there are relatively few B cells present in the human decidua, there are significantly more of these immunoglobulin-producing cells found in the lower female genital tract (endocervix > ectocervix > vagina) and interestingly, in the fallopian tubes (14, 49, 50). Distribution patterns in the lower genital tract can be easily explained by the likelihood of pathogen exposure but that in the fallopian tubes remains unclear. Could nonpathogen directed humoral immunity be particularly important at this site of fertilization and early embryo development? The concept is enticing and identifies an area for future investigation.
T-cell Regulation The TCR on CD4þ T cells recognizes antigen presented by MHC class II molecules on antigen-presenting cells. The MHC class II molecules are particularly important in defense against exogenous pathogens. Helper and regulatory T cells are CD4þ. The typical TCR on CD8þ T cells recognizes endogenous antigens presented on MHC class I molecules at the surface of antigen-presenting cells. These endogenous antigens may be derived from viruses, intracellular bacteria, or result from malignant transformation. In general, gametes and most placental cells are MHC class I and class II negative (51). The major exception is the extravillous trophoblast cell, which, although MHC class II negative, does express the nonpolymorphic nonclassic MHC class I molecules HLA-G and HLA-E, and the more polymorphic classic MHC class I molecule HLA-C (51). This unique MHC expression pattern likely helps to limit classic allorecognition reactions, yet promoting local uterine NK cell-mediated cytokine modulation and maternal vascular remodeling. Antigen presentation has additional layers of complexity. The CD4þ and CD8þ T-cell responses to cognate antigens involve the presence of costimulatory signals in addition to recognition of MHC-complexed antigen by the TCR (52). These costimulatory signals require interaction between a molecule called CD28 on the T cell and B7 molecules expressed on the antigen-presenting cell. Important, if the second signal is not present, the T cell will adapt to tolerate the cognate antigen (53, 54). If the second signal is present, the T cell will proliferate and begin to secrete cytokines. Adding one more layer to the complexity of this interaction is the presence of inhibitors of second signal interactions. Cytotoxic T lymphocyte antigen 4 is expressed on T cells and its expression increases when T cells are activated. This likely serves to control excessive response to antigen. Finally, other B7 family members act as ligands for a molecule expressed on activated T and B cells called programmed death 1 (PD-1) (55). Ligation of PD-1 with its receptor suppresses T-cell activation and promotes T-cell apoptosis or anergy. Ligation can also alter the cytokine secretory profiles of allogenic CD4þ T cells in response to VOL. - NO. - / - 2016
4
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472
Fertility and Sterility® 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531
B7 family member expression on human decidual stromal cells (53, 56). As early as insemination, the maternal immune system comes into contact with partner-derived alloantigens. Subsequent trophoblast shedding and continuous trafficking of fetal cells into the mother creates a supply of the same or related alloantigens that could stimulate tolerance reactions by maternal T cells (53). Both decidual stromal cells and trophoblast cells have been shown to display members of the B7 family of ligands; CD28, CDL4, and PD-1 are present on decidual T cells. The B7 family members B7-1 and B7-2 on decidual dendritic cells have been shown to promote the development of a Th2 phenotype (see later section Cytokine secretion profiles) upon ligation with responding T cells. A different B7 family member, B7-H1, has been detected on trophoblast cells. The B7-H1 receptor ligation also drives the development of a Th2 phenotype upon ligation to PD-1 on decidual CD4þ, CD8þ, or T regulatory (Treg) cells (53, 54). Other decidual stromal cells have been shown to increase indoleamine 2, 3 dioxygenase production through reversed B7-1/2 signaling after ligation with cytotoxic T lymphocyte antigen 4 on Treg cells (53). Finally, B7-H2 expressed on trophoblast cells interacts with Treg cells to cause an increase in interferon gamma (INF-g) and IL-10 production (54). Both cytokines are necessary for successful gestation. To control abnormally high levels of INF-g that might lead to pregnancy loss or preeclampsia, ligation of either B7-H1 or B7-DC to PD-1 has been shown to decrease additional cytokine secretion and lymphocyte proliferation (55, 56). In summary, these findings demonstrate the great importance of costimulatory interactions of decidual T cells with surrounding pregnancy-related cells in modulating maternal immune responses.
Cytokine Secretion Profiles Subclassification of mature T-helper cell phenotypes is largely dependent on the cytokine microenvironment in which naïve T helper (Th0) cells mature and differentiate in addition to their resultant cytokine secretion profiles. Th0 cells exposed to INF-g become Th1 cells and Th0 cells exposed to IL-4 become Th2-type cells (57, 58). The Th1 responses are largely inflammatory in nature and are associated with the release of IFN-g, IL-12, IL-2, and tumor necrosis factor (TNF)-b. In contrast, Th2 responses are characterized by antibody production and the secretion of the cytokines IL-10, IL-4, and IL-5 (59–61). Other T-helper responses include Treg cell responses, defined by the secretion of IL-10, and Th17 responses, defined by the secretion of IL-17 (62). During the menstrual cycle, there is a distinct increase in Treg and proinflammatory Th17 cell activity at the time of ovulation and possibly also at the time of fertilization (63, 64). The localized inflammatory-type reactions that are required for implantation (9, 65) must be tightly controlled and simultaneous increases in tolerogenic immune responses occur. When placed in mixed culture with paternal or unrelated peripheral blood mononuclear cells (PBMCs), Treg cells purified from pregnant women have been shown to suppress secretion of the inflammatory cytokine INF-g and
are potent suppressors of the inflammatory type 1 (cellmediated) immune responses (66). These cells also suppress IL-4 secretion against paternal antigens during pregnancy (67). The Th1-to-Th2 balance shifts to favor Th2 cells during the luteal phase of the menstrual cycle. At the same time, uterine NK cells dramatically increase in number, becoming the major immune cell population in the decidua (68–70). The number of uterine NK cells continues to increase until the end of the first trimester. Together, these changes illustrate the general bias in the female immune system toward an anti-inflammatory profile and away from proinflammatory Th1 responses that are typical of graft rejection (71). These specific changes in the female immune system that occur with each cycle could serve to condition the reproductive tract for contact with the foreign paternal antigens present in semen. They could also initiate a tolerizing response toward these antigens that are subsequently present on fetal tissue at the time of implantation (72). The clinical significance of these immune players has been well established in several disorders of pregnancy. As arguably one of the most influential components in establishing tolerance during gestation, insufficient or defective regulatory T cells in the human decidua appear to play a role in the pathophysiology of preterm births, recurrent pregnancy loss, and preeclampsia (73–75). Some investigators suggest that Th17 cells also have important roles in preterm labor, spontaneous abortions, and preeclampsia (76–79). The clinical impact of abnormalities in the activity of decidual NK cells has also been well documented. In cases of spontaneous abortion and recurrent pregnancy loss, decidual NK cells are more cytotoxic than corresponding cells from matched normal pregnancies (78, 80). With regard to the Th1-to-Th2 paradigm, Th1-type cytokines have been shown to be harmful to implanting embryos. Abnormal Th1 immune responses to trophoblast antigens have been documented in women with otherwise unexplained recurrent spontaneous abortion (81–85). Although certainly suggestive, investigations on the roles of T cells and their cytokine secretions in pregnancy loss are blurred by the possibility that some of the changes measured may be a reflection of the maternal immune system response to the abnormal and/or failing pregnancy rather than a part of the etiology of the loss (86).
Reproductive Hormones Estrogen (E), P, and hCG are three of the many hormones that play an important role in immune modulation within the female genital tract and the maintenance of the semiallogeneic implanting fetus (87, 88). Compared with the periphery, the concentration of these hormones is significantly increased at the maternal-to-fetal interface, which likely contributes to their corresponding effects (89). At high concentrations, Es foster the development of Th2 immune responses in vitro (90–92). In addition, the function and proliferation of Treg cells has been shown to be enhanced by E exposure. Estrogen further enhances Treg cell recruitment to the uterus by stimulating uterine expression of the chemokines CCL3, CCL4, and CCL5 (93). Similar to E, dramatic increases
VOL. - NO. - / - 2016
5
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590
VIEWS AND REVIEWS 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649
in P are observed during early pregnancy and maintained throughout gestation (94, 95). The immune effects of P are complex. This steroid hormone has been associated with Th2-type T-cell responses, increased Th1-type cytokine production, and increases in leukemia inhibitory factor (96–98). In addition, in vitro studies have provided evidence for the suppressive immune effects of this hormone by demonstrating P-related decreases in CD8þ T-cell proliferation and cytokine secretion (99). Progesterone has been shown to be important in sustaining and expanding Treg cells at the time of embryo implantation after their initial E-stimulated proliferation (66). Pregnancy associated protein hormones, such as hCG, also regulate local and systemic maternal immunity. There is a decrease in the expression of proinflammatory IL-17 concomitant with an increase in anti-inflammatory IL-27 and IL-10 in women supplemented with hCG during IVF (100). The hCG also acts as a chemoattractant that sequesters Treg cells at the fetal-tomaternal interface (101).
FETAL IMMUNOLOGIC CONTRIBUTIONS Remarkably, the developing fetus contributes to the type and extent of immune responses in the female genital tract toward itself as well as toward microbial invaders. During fertilization, sperm must bind to a specialized extracellular glycoprotein matrix, called the zona pellucida (ZP), and then travel through this specialized matrix to fuse with the egg to form a zygote (102). The lectin-like and protein-to-protein interactions involved in human sperm binding to the ZP have been hypothesized to be important for later immune cell recognition of the embryo before blastocyst formation (103). During the pre-embryo stage, there are equal contributions from both maternal and foreign paternal genomes, but shortly after formation, the pre-embryo begins to express paternal MHC molecules (104). The ZP has been shown to protect mouse pre-embryos bearing paternal MHC antigens from destruction by cytotoxic T cells by serving as a physical blockade against rejection of the pre-embryo (102, 104). There is also evidence that by providing a source of paternally derived antigens, fetal microchimerism could induce a donor-specific type of immune tolerance at the maternal-to-fetal interface (105). Studies confirm that the female immune system recognizes and responds to paternal trophoblast antigens by producing antibodies and T cells in the peripheral blood and decidua that are reactive to fetal human leukocyte antigens (5, 106, 107). The fetus is not a static entity in the immune interactions occurring at the maternal-to-fetal interface nor is it immunologically incompetent (108). As the pre-embryo develops into a fetus, multiple and diverse mechanisms in the intra-amniotic cavity and the neonatal circulation modulate proinflammatory immune responses. Fetal immune cells do recognize foreign pathogen and non-self antigens, although during much of pregnancy, naïve fetal T cells have a propensity to differentiate into Treg cells with antigen exposure; a propensity thought to promote tolerance (109). When this response is directed against microbial invaders, it may allow the developing baby to tolerate at least a low level of this immune activation (110). This ability to tolerate local
infection may decrease the likelihood of premature delivery due to infection-controlling immune activation. It could also reduce fetal exposure to toxic immune products necessary for definitive microbe elimination without promoting the creation of new resistance mechanisms by the offending organism (110). As the pregnancy continues, fetal immune cells will begin to transition toward more adult-type immune cells characterized by more protective immune responses in preparation for delivery, although some fetal-like protolerogenic cells survive in offspring for many years (111, 112).
THE ROLE OF SEMEN AND SPERM Changes in female genital tract immunity that enable allogeneic pregnancy are initiated long before implantation. The ability of both the cellular and soluble components of human semen to alter immune responses within the female genital tract may be critical for the success of human fertilization and reproduction (113–115). Upon entering the female genital tract, human semen and male germ cells face a relatively hostile environment (116). In efforts to ensure safe passage, multiple gene expression pathways are initiated in female epithelial cells upon contact with seminal fluid (117, 118). Of the >700 genes found to be differentially expressed in ectocervical tissue after intercourse, most are involved in the presentation of proinflammatory antigens and the expression of cytokines and chemokines that recruit and activate immune cells such as T cells, dendritic cells, and macrophages (117, 118). These semen-induced changes in gene expression and resultant cellular response are known as the leukocyte reaction (117, 119–122). The absence of paternal MHC class I or class II molecules on the human sperm surface serves to bypass the stimulation of specific histocompatibility-based adaptive responses within the female genital tract (123), yet the environment remains potentially hostile as nonantigenspecific innate immune mediators have also been activated. As with implantation, the initial inflammatory immune response to introduction of male germ cells that need to traverse the genital tract is seemingly counterintuitive and must therefore be nuanced and tightly regulated. The evidence for male-to-female signaling by seminal fluid is vast with studies in porcine, equine, bovine, ovine, canine, and human species. Despite the induction of what at first blush would appear to be an ominous immune response for male germ cell invaders, these studies show that exposure to seminal fluid results in higher fertilization rates, increased sperm survival, and improved embryo implantation and development (124). As the leukocyte reaction in the female genital tract commences, the destruction of incompetent or senescent nonfertilizing spermatozoa occurs through processes of apoptosis and phagocytosis that have been termed silent (125, 126). They differ from related processes typical of somatic cells because they occur in the absence of significant release of reactive oxygen species or proinflammatory cytokines in the local environment. If an infectious agent is present, however, the seminal fluid provides protection for the remaining competent cells from reactive oxygen species that may be generated (125). VOL. - NO. - / - 2016
6
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708
Fertility and Sterility® 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767
Although sperm are MHC negative (123), the deposition of seminal fluid at coitus provides the initial contact of the female genital tract immune system with paternal antigens present on the immune cells, paternal epithelial cells, and cellular debris that are present within seminal fluid. The soluble immune components of seminal fluid then modulate the maternal response to these foreign antigens in such a way that pregnancy can progress when these same antigens are present on the conceptus in a resulting or future pregnancy (127).
induction of an anti-inflammatory state in the female genital tract and the recruitment of Treg cell populations to the genital tract. Together, these responses not only serve to modulate responses to paternally derived antigens in semen, but also help prevent rejection of the implanting embryo (129, 139, 140). Investigations focused on the role of PGE2 have uncovered proinflammatory and immunosuppressive functions, although the dominant effects appear to be the promotion of Th2-cell responses concomitant with the inhibition of Th1-cell responses (1).
Seminal Fluid and Regulatory T Cells
Seminal Fluid and Dendritic Cells
Perhaps the most significant pathway sequence initiated by seminal fluid is the activation and expansion of maternal Treg cells. As discussed previously, elevations in local and circulating E levels at ovulation cause an initial and nonspecific increase in circulating Treg cells. In conjunction, seminal fluid ‘‘primes’’ this reaction before conceptus antigen encounter by providing the means to expand specific, paternal antigen-reactive Treg cells through TCR ligation. Only through this mechanism is the suppressive function of Treg cells fully realized (128). First demonstrated in mice, proliferation of Treg cells in para-aortic lymph nodes and responding lymphocytes is markedly increased several days after coitus when females are mated with intact males. This effect is not observed when mating is performed with male mice whose seminal vesicles have been excised (129). Other investigators (130) have provided evidence for the induction of female immune tolerance of seminal fluid by demonstrating a decrease in cell-mediated immunity, a process significantly impacted by Treg cells, in similarly designed murine experiments. Demonstrated increases in Treg cell proliferation are not limited to the lymph nodes. Uterine Treg cell populations are also dependent on exposure to seminal plasma (129). In addition, seminal fluid induces uterine expression of messenger RNA encoding the Treg chemokine CCL19, which further increases recruitment of these cells to the uterus before embryo implantation (131, 132). Seminal fluid has effects on the upper reproductive tract that may further affect the quantity and activity of uterine Treg cells. This complex secretion has been shown to increase macrophage populations in the corpus lutea (CL) that, in turn, increase circulating plasma P levels (66, 133). Progesterone is important in both Treg cell differentiation and stability. The CL macrophages are also vital for the remodeling events that must take place within the ovary to maintain the steroidogenic function that supports early pregnancy (66, 134).
The effects of seminal fluid are not limited to those observed in T cells. For the female immune system to mount its robust response to antigens contained in seminal fluid, many immune cells must be present in endometrial tissues during and after ovulation during the period of fertilization and the temporal window of implantation (141, 142). In murine studies, seminal plasma has been shown to induce several dendritic cell chemokines (chemokine [CC] motif ligand [CCL]2, CCL3, CCL5, CCL20, CCL22, and CXCL10) that aid in the after coital recruitment and maturation of additional dendritic cells to endometrial tissues (141, 143). Important, these dendritic cells express markers that identify them as tolerogenic (142). Human in vitro studies have shown that this specific phenotype appears particularly adept at differential induction of Treg cells that produce and secrete more levels of IL-10 and TGF-b compared with dendritic cells with a nontolerogenic phenotype (144, 145). Such dendritic cells are also important in the clonal expansion of the subset of Treg cells that express TCRs specific for seminal fluid antigens (145). Interestingly, recent studies in mice have provided evidence that seminal fluid may influence dendritic and Treg cells by increasing the levels of specific microRNAs in the female reproductive tract (146). A novel mechanism has also been proposed in which direct delivery of sperm microRNA in microvesicles to the oocyte can impact embryo development and the inheritance of paternal traits (147–149). Last, HLA-G, an unusual MHC class I molecule typically expressed only by extravillous cytotrophoblast cells, has been identified in human seminal plasma where it has been proposed to induce tolerogenic dendritic cells, aid in the induction of Treg cells, and modulate NK cell immune responses (150–152).
Role of Transforming Growth Factor b and Prostaglandin E2 Transforming growth factor (TGF) b and prostaglandin E2 (PGE2) are arguably the two most important immune modulators in seminal fluid. Both of these key signaling agents act by binding to receptors on cervical and possibly endometrial cells. They have also been shown to play roles in inducing Treg cells (135–138). Transforming growth factor-b is integral to the
Clinical Relevance Given the evidence supporting an important role for seminal fluid in inducing maternal immune changes that support fertilization and implantation, it comes as a bit of a surprise that IVF and artificial insemination procedures are so successful as each separates sperm cells from the seminal plasma and discards the latter. This shows that seminal fluid is not absolutely required for a successful fertilization event. Rather, the exposure of the female genital tract to seminal plasma appears to most directly impact the ‘‘likelihood’’ of further development of a pregnancy and its overall health. To this point, exposure to seminal fluid has been associated with a
VOL. - NO. - / - 2016
7
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826
VIEWS AND REVIEWS 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885
reduction in certain disease states in pregnant women and improved fetal growth and health (122). Decreased populations or impaired functionality of Treg cells have been described in women with preeclampsia, miscarriage, and primary unexplained infertility (74, 153–156). Women who have had prolonged vaginal or oral exposure to seminal plasma have been shown to have a lower risk of developing preeclampsia than women with significantly less exposure (157, 158). Male offspring of seminal plasma-deficient fathers have higher rates of obesity and metabolic syndrome (159). Finally, a recent meta-analysis (160) of seven randomized controlled trials involving patients undergoing IVF demonstrated a statistically significant improvement in clinical pregnancy rates (PRs) in those women exposed to seminal plasma at or near the time of oocyte pickup or ET when compared with those patients without exposure to seminal fluid.
FERTILIZATION AND EARLY IMPLANTATION: A ‘‘NEW’’ VIEWPOINT Given the complexity, redundancy, finesse, and time and location dependency of the immune interactions that support pregnancy maintenance, it should come as no great surprise that theorists have come full circle in their descriptions of these interactions. Fairly recent investigations in painstakingly designed murine models addressing maternal immune responses to paternal antigens have again introduced the concept of immunologic ignorance, at least as it applies to before and peri-implantation events in mice (161, 162). In the setting of an allograft, T-cell antigen recognition can occur in two ways: direct and indirect (163). For direct recognition, T cells in the host directly recognize allogenic MHC/peptide presented on donor cells. This type of recognition appears to be the major contributor to most allograft rejection (164). Indirect recognition involves host T cell recognition of graft-derived antigens displayed by host antigen presenting cells expressing host MHC. Erlebacher et al. (162) have demonstrated that, in mice, allorecognition of paternal antigens only occurred through indirect recognition pathways, that allorecognition through this pathway resulted in specific deletion of potentially alloreactive T cells, and that allorecognition occurred only after about the middle of mouse gestation. Other factors involved in this delayed allorecognition might include poor MHC expression on gametes and the early embryo, lack of damage to epithelial barriers as sperm and embryo transit the female reproductive tract (limiting danger signals) (8, 165), alloantigen masking (1), and the relatively small amounts of potential alloantigen exposure that are present during early implantation before maternal spiral artery remodeling. Tolerance mechanisms may support and sustain ignorance. Delayed alloantigen recognition explained by immunologic ignorance was supported by another study published at about the same time (161) demonstrating that mice were ignorant of paternal antigen when exposed through intravaginal insemination. This state of specific ignorance to paternal alloantigen persisted from the time of semen exposure through estrous and early implantation. It was not
present if the paternal antigen was administered SC. The demonstrated importance of E to this process reinforces the concept that timing and location of antigen exposure may be critical. Again, several other factors may have been involved, including alloantigen masking (1, 166), poor MHC expression on gametes, silent sperm phagocytosis and limited danger signals, and the multiple immunosuppressive properties of human seminal fluid. It is worth mentioning that some investigators have attempted to explain maternal tolerance of the fetal allograft using paradigm-shifting descriptions of the mechanisms underlying immune responses. For example, proponents of the danger model (165, 167) posit that the initial event signaling an immune response is not the discrimination of self versus non-self but rather the recognition of tissue damage or cellular stress that releases a danger signal. This adds a third, albeit initiating, signal to the two signal theories of immune recognition relying on a combination of self non-self recognition and a second signal from T helper cells. In the two signal models, the helper T cells themselves require antigen recognition and a costimulatory signal or they will die. In the danger model, T-cell recognition of antigens in the absence of danger signals will die. Only antigen recognition in the local environment of tissue damage signaling danger will be fully activated. Cells undergoing programmed cell death (apoptosis) do not release danger signals. Like more classic models of immune response, the danger model can be used to explain many, but not all, of the alloimmune challenges of human fertilization and implantation. For instance, although millions of sperm enter the female genital tract after intercourse, nonfertilizing sperm die by apoptosis rather than necrosis. Therefore, in the absence of tissue damage within the female mucosal tract, no danger signals are generated and no immune response against alloantigens in semen occurs. The existence of alternate and well-reasoned models for immune responses speak directly to the fact that experimental data surrounding the immunology of eutherian pregnancy is so complex that a single simplified model presently appears insufficient. In conclusions, discussions of the immune interactions surrounding the maintenance of human pregnancy invoke multiple, complex, overlapping, and redundant mechanisms. There remain inconsistencies within the large volume of available literature that can frustrate the reproductive immunologist. These may arise from several sources. The impact of certain mechanisms may be time and location specific. Most must be balanced by the necessity to retain appropriate immune responses to pathogen and nonpathogen related local immune insults. Some pregnancy-specific immune changes may be initiated to address differing levels of antigen exposure, such as those occurring during implantation of a relatively small embryo versus those encountered throughout pregnancy during placental syncytiotrophoblast turnover. Some may be particularly difficult to study in humans, including those interactions occurring at the site of implantation and during the first steps of placental development when an unusual form of syncytiotrophoblast acts as the invading edge of fetally derived tissues. Because the latter interactions are particularly difficult to study in humans for logistical and ethical reasons, some of our VOL. - NO. - / - 2016
8
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944
Fertility and Sterility® 945 946 947 948 949 950 951 952 953 954 955 956 Q4 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003
insights may derive only from animal studies, many of which may have limitations due to the high degree of placental invasion noted in humans when compared with most model species. The complexity of general immune responses may presently defy explanation using a single model. For these and many other reasons, our understanding of the intricacies of these interactions continues to be regrettably incomplete. The challenge is great. Our response to this challenge must remain focused and robust.
20.
21. 22. 23. 24. 25.
REFERENCES 1. 2.
3.
4. 5. 6.
7. 8.
9. 10.
11.
12.
13.
14.
15.
16. 17.
18. 19.
26.
Clark GF, Schust DJ. Manifestations of immune tolerance in the human female reproductive tract. Front Immunol 2013;4:26. Rametse CL, Olivier AJ, Masson L, Barnabas S, McKinnon LR. Role of semen in altering the balance between inflammation and tolerance in the female genital tract: does it contribute to HIV risk? Viral Immunol 2014;27:200–6. Medawar P. Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Symp Soc Exp Biol 1953;7: 320–38. Payne R, Rolfs MR. Fetomaternal leukocyte incompatibility. J Clin Invest 1958;37:1756–63. Van Rood JJ, Eernisse JG, van Leeuwen A. Leucocyte antibodies in sera from pregnant women. Nature 1958;181:1735–6. Bartel G, Walch K, Wahrmann M, Pils S, Kussel L, Polterauer S, et al. Prevalence and qualitative properties of circulating anti-human leukocyte antigen alloantibodies after pregnancy: no association with unexplained recurrent miscarriage. Hum Immunol 2011;72:187–92. Tafuri A, Alferink J, Moller P, Hammerling GJ, Arnold B. T cell awareness of paternal alloantigens during pregnancy. Science 1995;270:630–3. Norton MT, Fortner KA, Oppenheimer KH, Bonney EA. Evidence that CD8 T-cell homeostasis and function remain intact during murine pregnancy. Immunology 2010;131:426–37. Dekel N, Gnainsky Y, Granot I, Racicot K, Mor G. The role of inflammation for a successful implantation. Am J Reprod Immunol 2014;72:141–7. Gnainsky Y, Granot I, Aldo P, Barash A, Or Y, Mor G, et al. Biopsy-induced inflammatory conditions improve endometrial receptivity: the mechanism of action. Reproduction 2015;149:75–85. Redman CW, Sargent IL. Pre-eclampsia, the placenta and the maternal systemic inflammatory response—a review. Placenta 2003;24(Suppl A): S21–7. Redman CW, Tannetta DS, Dragovic RA, Gardiner C, Southcombe JH, Collett GP, et al. Review: Does size matter? Placental debris and the pathophysiology of pre-eclampsia. Placenta 2012;33(Suppl):S48–54. Shynlova O, Lee YH, Srikhajon K, Lye SJ. Physiologic uterine inflammation and labor onset: integration of endocrine and mechanical signals. Reprod Sci 2013;20:154–67. Kutteh WH, Stovall DW, Schust DJ. Immunology and reproduction. In: Strauss JF III, Barbieri RL, editors. Yen and Jaffe's reproductive endocrinology: physiology, pathophysiology, and clinical management. Philadelphia: Elsevier Saunders; 2014:287–307. Kutteh WH, Modoveanu Z, Mestecky J. Mucosal immunity in the female reproductive tract: correlation of immunoglobulins, cytokines, and reproductive hormones in human cervical mucus around the time of ovulation. AIDS Res Hum Retroviruses 1998;14(Suppl 1):S51–5. Kutteh WH, Mestecky J. Secretory immunity in the female reproductive tract. Am J Reprod Immunol 1994;31:40–6. Gould DS, Ploegh HL, Schust DJ. Murine female reproductive tract intraepithelial lymphocytes display selection characteristics distinct from both peripheral and other mucosal T cells. J Reprod Immunol 2001;52:85. Loke YW, King A. Immunological aspects of human implantation. J Reprod Fertil Steril 2000;55:83–90. Schon MP, Arya A, Murphy EA, Adams CM, Strauch UG, Agace WW, et al. Mucosal T lymphocyte numbers are selectively reduced in integrin alpha E (CD103)-deficient mice. J Immunol 1999;162:6641–9.
27. 28.
29.
30.
31. 32.
33. 34. 35.
36.
37.
38.
39.
40.
41. 42. 43. 44.
45.
Perry LL, Feilzer K, Portis JL, Caldwell HD. Distinct homing pathways direct T lymphocytes to the genital and intestinal mucosae in Chlamydia-infected mice. J Immunol 1998;160:2905–14. Hardy K, Spanos S. Growth factor expression and function in the human and mouse preimplantation embryo. J Endocrinol 2002;172:221–36. O'Neill C. The potential roles for embryotrophic ligands in preimplantation embryo development. Hum Reprod Update 2008;14:275–88. Johnson PM, Christmas PE, Vince GS. Immunological aspects of implantation and implantation failure. Hum Reprod 1999;14(Suppl 2):26–36. Vince G, Johnson P. Leukocyte populations and cytokine regulation in human uteroplacental tissues. Biochem Soc Trans 2000;28:191–5. Loke YW, King A. Decidual natural-killer-cell interaction with trophoblast: cytolysis or cytokine production? Biochem Soc Trans 2000;28:196–8. Male V, Trundley A, Gardner L, Northfield J, Chang C, Apps R, et al. Natural killer cells in human pregnancy. Methods Mol Biol 2010;612:447–63. Chazara O, Xiong S, Moffett A. Maternal KIR and fetal HLA-C: a fine balance. J Leukoc Biol 2011;90:703–16. Kopcow HD, Allan DS, Chen X, Rybalov B, Andzelm MM, Ge B, et al. Human decidual NK cells form immature activating synapses and are not cytotoxic. Proc Natl Acad Sci U S A 2005;102:15563–8. Hanna J, Goldman-Wohl D, Hamani Y, Avraham I, Greenfield C, NatansonYaron S, et al. Decidual NK cells regulate key developmental processes at the human fetal-maternal interface. Nat Med 2006;12:1065–74. Manaster I, Mizrahi S, Goldman-Wohl D, Sela HY, Stern-Ginossar N, Lankry D, et al. Endometrial NK cells are special immature cells that await pregnancy. J Immunol 2008;181:1869–76. Godfrey D, Hammond KJ, Poulton LD, Smyth MJ, Baxter AG. NKT cells: facts, functions, and fallacies. Immunol Today 2000;21:573–83. Boyson JE, Rybalov B, Koopman LA, Exley M, Balk SP, Racke FK, et al. CD1d and invariant NKT cells at the human maternal-fetal interface. Proc Natl Acad Sci U S A 2002;99:13741–6. Dang Y, Heyborne K. Cutting edge: regulation of uterine NKT cells by a fetal class I molecule other than CD1. J Immunol 2001;166:3641–4. Dang Y, Beckers J, Wang CR, Heyborne K. Natural killer 1.1þ ab T cells in the periimplantation uterus. Immunology 2000;101:484. Mincheva-Nilsson L, Baranov V, Yeung MM, Hammarstrom S, Hammarstrom ML. Immunomorphologic studies of human deciduaassociated lymphoid cells in normal early pregnancy. Adv Exp Med Biol 1995;371A:367–71. Christmas SE, Brew R, Thornton SM, Deniz G, Flanagan BF. Extensive TCR junctional diversity of V gamma 9/V delta 2 clones from human female reproductive tissues. J Immunol 1995;155:2453–8. Vassiliadou N, Bulmer J. Characterization of endometrial T lymphocyte subpopulations in spontaneous early pregnancy loss. Hum Reprod 1998;13: 44–7. Hayday AC. [gamma][delta] cells: a right time and a right place for a conserved third way of protection. Annu Rev Immunol 2000;18:975– 1026. Fan DX, Duan J, Li MQ, Xu B, Li DJ, Jin LP. The decidual gamma-delta T cell upregulates the biological functions of trophoblasts via IL-10 secretion in early human pregnancy. Clin Immunol 2011;141:284–92. Nakamura H, Jasper MJ, Hull ML, Aplin JD, Robertson SA. Macrophages regulate expression of a 1,2-fucosyltransferase genes in human endometrial epithelial cells. Mol Hum Reprod 2012;18:204–15. Nagamatsu T, Schust DJ. The immunomodulatory roles of macrophages at the maternal-fetal interface. Reprod Sci 2010;17:209–18. Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells—how did we miss them? Nat Rev Immunol 2013;13:75–87. Kumar V. Innate lymphoid cells: new paradigm in immunology of inflammation. Immunol Lett 2014;157:23–37. Doisne JM, Balmas E, Boulenouar S, Gaynor LM, Kieckbusch J, Gardner L, et al. Composition, development, and function of uterine innate lymphoid cells. J Immunol 2015;195:3937–45. Aït-Azzouzene D, Gendron MC, Houdayer M, Langkopf A, B€ urki K, Nemazee D, et al. Maternal B lymphocytes specific for paternal histocompatibility antigens are partially deleted during pregnancy. J Immunol 1998; 161:2677–83.
VOL. - NO. - / - 2016
9
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062
VIEWS AND REVIEWS 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 Q1 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121
46.
47. 48.
49. 50.
51. 52. 53.
54.
55.
56.
57. 58.
59. 60.
61.
62. 63. 64.
65.
66.
67.
68.
69.
Muzzio DO, Ziegler KB, Ehrhardt J, Zygmunt M, Jensen F. Marginal zone B cells emerge as a critical component of pregnancy well-being. Reproduction 2016;151:29–37. Dauby N. B cell responses in pregnancy and vaccine efficacy. Hum Reprod Update 2015;21:407–8. Kraus TA, Engel SM, Sperling RS, Kellerman L, Lo Y, Wallenstein S, et al. Characterizing the pregnancy immune phenotype: results of the viral immunity and pregnancy (VIP) study. J Clin Immunol 2012;32:300–11. Mestecky J, Fultz PN. Mucosal immune system of the human genital tract. J Infect Dis 1999;179(Suppl 3):S470–4. Givan AL, White HD, Stern JE, Colby E, Gosselin EJ, Guyre PM, et al. Flow cytometric analysis of leukocytes in the human female reproductive tract: comparison of fallopian tube, uterus, cervix, and vagina. Am J Reprod Immunol 1997;38:350–9. Hyde KJ, Andelin CO, Schust DJ. Immunology of isolated and recurrent spontaneous pregnancy loss. Glob Libr Women's Med 2014. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013;13:227–42. Petroff MG, Perchellet A. B7 family molecules as regulators of the maternal immune system in pregnancy. Am J Reprod Immunol 2010; 63:506–19. Nagamatsu T, Barrier BF, Schust DJ. The regulation of T-cell cytokine production by ICOS-B7H2 interactions at the human fetomaternal interface. Immuno Cell Biol 2011;89:417–25. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, et al. Engagement of the PD-1 immunoinhibitoy receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 2000;192:1027–34. Nagamatsu T, Schust DJ, Sugimoto J, Barrier BF. Human decidual stromal cells suppress cytokine secretion by allogenic CD4þ T cells via PD-1 ligand interactions. Hum Reprod 2009;24:3160–71. Asnagli H, Murphy KM. Stability and commitment in T helper cell development. Curr Opin Immunol 2001;13:242–7. Palmer EM, van Seventer GA. Human T helper cell differentiation is regulated by the combined action of cytokines and accessory cell-dependent costimulatory signals. J Immunol 1997;158:2654–62. O'Garra A, Arai N. The molecular basis of T helper 1 and T helper 2 cell differentiation. Trends Cell Biol 2000;10:542–50. Kurt-Jones E, Hamberg S, Ohara J, Paul WE, Abbas AK. Heterogeneity of helper/inducer T lymphocytes. I. Lymphokine production and lymphokine responsiveness. J Exp Med 1987;166:1774–87. Romagnani S. Human TH1 and TH2 subsets: regulation of differentiation and role in protection and immunopathology. Int Arch Allergy Immunol 1992;98:279–85. Mucida D, Cheroutre H. The many face-lifts of CD4 T helper cells. Adv Immunol 2010;107:139–52. Fish EN. The X-files in immunity: sex-based differences predispose immune responses. Nat Rev Immunol 2008;8:737–44. 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;63:104–9. Gnainsky Y, Granot I, Aldo PB, Barash A, Or Y, Schechtman E, et al. Local injury of the endometrium induces an inflammatory response that promotes successful implantation. Fertil Steril 2010;94:2030–6. Mao G, Wang J, Kang Y, Tai P, Wen J, Zou Q, et al. Progesterone increases systemic and local uterine proportions of CD4þCD25þ Treg cells during midterm pregnancy in mice. Endocrinology 2010;151:5477–88. Mjosberg J, Berg G, Ernerudh J, Ekerfelt C. CD4þ CD25þ regulatory T cells in human pregnancy: development of a Treg-MLC-ELISPOT suppression assay and indications of paternal specific Tregs. Immunology 2007;120: 456–66. 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;74:1008–13. Bulmer JN, Morrison L, Longfellow M, Ritson A, Pace D. Granulated lymphocytes in human endometrium: histochemical and immunohistochemical studies. Hum Reprod 1991;6:791–8.
70. 71. 72. 73.
74.
75.
76.
77.
78.
79.
80.
81. 82.
83.
84.
85.
86. 87.
88.
89.
90.
Lee JY, Lee M, Lee SK. Role of endometrial immune cells in implantation. Clin Exp Reprod Med 2011;38:119–25. Zenclussen AC. Adaptive immune responses during pregnancy. Am J Reprod Immunol 2013;69:291–303. Schumacher A, Zenclussen AC. The paternal contribution to fetal tolerance. Adv Exp Med Biol 2015;868:211–25. Mei S, Tan J, Chen H, Chen Y, Zhang J. Changes of CD4þCD25 high regulatory T cells and FOXP3 expression in unexplained recurrent spontaneous abortion patients. Fertil Steril 2010;94:2244–7. Sasaki Y, Sakai M, Miyazaki S, Higuma S, Shiozaki A, Saito S. Decidual and peripheral blood CD4þCD25þ regulatory T cells in early pregnancy subjects and spontaneous abortion cases. Mol Hum Reprod 2004;10:347–53. Galazka K, Wicherek L, Pitynski K, Kijowski J, Zajac K, Bednarek W, et al. Changes in the subpopulation of CD25þ CD4þ and FOXP3þ regulatory T cells in decidua with respect to the progression of labor at term and the lack of analogical changes in the subpopulation of suppressive B7-H4 macrophages—a preliminary report. Am J Reprod Immunol 2009; 61:136–46. Lee SK, Kim JY, Hur SE, Kim CJ, Na BJ, Lee M, et al. An imbalance in interleukin-17-producing T and Foxp3þ regulatory T cells in women with idiopathic recurrent pregnancy loss. Hum Reprod 2011;26:2964–71. Toldi G, Rigo J Jr, Stenczer B, Vasarhelyi B, Molvarec A. Increased prevalence of IL-17-producing peripheral blood lymphocytes in pre-eclampsia. Am J Reprod Immunol 2011;66:223–9. Wang WJ, Hao CF, Yi L, Yin GJ, Bao SH, Qiu LH, et al. Increased prevalence of T helper 17 (Th17) cells in peripheral blood and decidua in unexplained recurrent spontaneous abortion patients. J Reprod Immunol 2010;84: 164–70. Q2 Ito M, Nakashima A, Hidaka T, Okabe M, Bac ND, Ina S, et al. A role for IL-17 in induction of an inflammation at the fetomaternal interface in preterm labour. J Reprod Immunol 2010;84:75–85. Chao KH, Yang YS, Ho HN, Chen SU, Chen HF, Dai HJ, et al. Decidual natural killer cytotoxicity decreased in normal pregnancy but not in an embryonic pregnancy and recurrent spontaneous abortion. Am J Reprod Immunol 1995;34:274–80. Hill J, Polgar K, Anderson D. T-helper 1-type immunity to trophoblast in women with recurrent spontaneous abortion. JAMA 1995;273:1933–6. Hill JA, Polgar K, Harlow BL, Anderson DJ. Evidence of embryo- and trophoblast-toxic cellular immune response(s) in women with recurrent spontaneous abortion. Am J Obstet Gynecol 1992;166:1044–52. Berkowitz R, Hill JA, Kurtz CB, Anderson DJ. Effects of products of activated leukocytes (lymphokines and monokines) on the growth of malignant trophoblast cells in vitro. Am J Obstet Gynecol 1988;158:199–203. Hunt J, Soares MJ, Lei MG, Smith RN, Wheaton D, Atherton RA, et al. Products of lipopolysaccharide-activated macrophages (tumor necrosis factoralpha, transforming growth factor-beta) but not lipopolysaccharide modify DNA synthesis by rat trophoblast cells exhibiting the 80-kDa lipopolysaccharide-binding protein. J Immunol 1989;143:1606–13. Hill JA, Haimovici F, Anderson DJ. Products of activated lymphocytes and macrophages inhibit mouse embryo development in vitro. J Immunol 1987;139:2250–4. Moffett A, Shreeve N. First do no harm: uterine natural killer (NK) cells in assisted reproduction. Hum Reprod 2015;30:1519–25. Siiteri P, Febres F, Clemens LE, Chang RJ, Gondos B, Stites D. Progesterone and maintenance of pregnancy: is progesterone nature's immunosuppressant? Ann N Y Acad Sci 1977;286:384–97. Wira CR, Fahey JV, Rodriguez-Garcia M, Shen Z, Patel MV. Regulation of mucosal immunity in the female reproductive tract: the role of sex hormones in immune protection against sexually transmitted pathogens. Am J Reprod Immunol 2014;72:236–58. Runnebaum B, Stober I, Zander J. Progesterone, 20 alphadihydroprogesterone and 20 beta-dihydroprogesterone in mother and child at birth. Acta Endocrinol (Copenh) 1975;80:569–76. Salem M, Matsuzaki G, Kishihara K, Madkour GA, Nomoto K. beta-estradiol suppresses T cell-mediated delayed-type hypersensitivity through suppression of antigen-presenting cell function and Th1 induction. Int Arch Allergy Immunol 2000;121:161–9.
VOL. - NO. - / - 2016
10
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180
Fertility and Sterility® 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239
91.
92.
93.
94.
95.
96.
97.
98. 99.
100.
101.
102.
103. 104.
105.
106. 107.
108.
109.
110.
111.
Correale J, Arias M, Gilmore W. Steroid hormone regulation of cytokine secretion by proteolipid protein-specific CD4þ T cell clones isolated from multiple sclerosis patients and normal control subjects. J Immunol 1998; 161:3365–74. Cutolo M, Wilder R. Different roles for androgens and estrogens in the susceptibility to autoimmune rheumatic diseases. Rheum Dis Clin North Am 2000;26:825–39. Kallikourdis M, Betz AG. Periodic accumulation of regulatory T cells in the uterus: preparation for the implantation of a semiallogeneic fetus? PLoS One 2007;2:e382. Jackson DL, Schust DJ. The role of the placenta in autoimmune disease and early pregnancy loss. In: Kay HH, Nelson DM, Wang Y, editors. The placenta: from development to disease. Hoboken, NJ: Wiley-Blackwell; 2011:215–21. Prieto GA, Rosenstein Y. Estradiol potentiates the suppressive function of human CD4 CD25 regulatory T cells by promoting their proliferation. J Immunol 2006;118:58–65. Piccinni MP, Beloni L, Livi C, Maggi E, Scarselli G, Romagnani S. Defective production of both leukemia inhibitory factor and type 2 T-helper cytokines by decidual T cells in unexplained recurrent abortions. Nat Med 1998;4: 1020–4. Hunt JS, Miller L, Roby KF, Huang J, Platt JS, DeBrot BL. Female steroid hormones regulate production of pro-inflammatory molecules in uterine leukocytes. J Reprod Immunol 1997;35:87–99. Szekeres-Bartho J, Polgar B. PIBF: the double edged sword. Pregnancy and tumor. Am J Reprod Immunol 2010;64:77–86. Vassiliadou N, Tucker L, Anderson D. Progesterone-induced inhibition of chemokine receptor expression on peripheral blood mononuclear cells correlates with reduced HIV-1 infectability in vitro. J Immunol 1999;162: 7510–8. Koldehoff M, Katzorke T, Wisbrun NC, Propping D, Wohlers S, Bielfeld P, et al. Modulating impact of human chorionic gonadotropin hormone on the maturation and function of hematopoietic cells. J Leukoc Biol 2011; 90:1017–26. Schumacher A, Brachwitz N, Sohr S, Engeland K, Langwisch S, Dolaptchieva M, et al. Human chorionic gonadotropin attracts regulatory T cells into the fetal-maternal interface during early human pregnancy. J Immunol 2009;182:5488–97. Clark GF. The mammalian zona pellucida: a matrix that mediates both gamete binding and immune recognition? Syst Biol Reprod Med 2010; 56:349–64. Clark GF. The role of carbohydrate recognition during human sperm-egg binding. Hum Reprod 2013;28:566–77. Ewoldsen MA, Ostlie NS, Warner CM. Killing of mouse blastocyst stage embryos by cytotoxic T lymphocytes directed to major histocompatibility complex antigens. J Immunol 1987;138:2764–70. Scherjon S, Lashley L, van der Hoorn ML, Claas F. Fetus specific T cell modulation during fertilization, implantation and pregnancy. Placenta 2011; 32:S291–7. Hunt JS, Pace JL, Morales PJ, Ober C. Immunogenicity of the soluble isoforms of HLA-G. Mol Hum Reprod 2003;9:729–35. Tiburgs T, Scherjon SA, Vandermast BJ, Haasnoot GW, Versteeg VDV, VerSteeg MM, et al. Fetal-maternal HLA-C mismatch is associated with decidual T cell activation and induction of functional T regulatory cells. J Reprod Immunol 2009;82:148–57. Kutteh WH, Rainey WE, Beutler B, Carr BR. Tumor necrosis factor-alpha and interleukin-1 beta production by human fetal Kupffer cells. Am J Obstet Gynecol 1991;165:112–20. Mold JE, Venkatasubrahmanyam S, Burt TD, Micha€elsson J, Rivera JM, Galkina SA, et al. Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans. Science 2010;330:1695–9. Witkin S, Linhares I, Bongiovanni A, Herway C, Skupski D. Unique alterations in infection-induced immune activation during pregnancy. BJOG 2011;118:145–53. Mold JE, Micha€ elsson J, Burt TD, Meunch MO, Beckerman KP, Busch MP, et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science 2008;322:1562–5.
112.
113. 114.
115.
116. 117.
118.
119. 120. 121.
122. 123. 124.
125.
126.
127. 128.
129.
130.
131.
132. 133.
134.
Burt TD. Fetal regulatory T cells and peripheral immune tolerance in utero: implications for development and disease. Am J Reprod Immunol 2013;69: 346–58. Kelly RW, Critchley HO. Immunomodulation of human seminal plasma: a benefit for spermatozoan and pathogen? Hum Reprod 1997;12:2200–7. Politch JA, Tucker L, Bowman FP, Anderson DJ. Concentrations and significance of cytokines and other immunologic factors in semen of healthy fertile men. Hum Reprod 2007;22:2928–35. Robertson SA, Guerin LR, Moldenhauer LM, Hayball JD. Activating T regulatory cells for tolerance in early pregnancy—the contribution of seminal fluid. J Reprod Immunol 2009;83:109–16. Drobnis EZ, Overstreet JW. Natural history of mammalian spermatozoa in the female reproductive tract. Oxf Rev Reprod Biol 1992;14:1–45. Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA. Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol 2012;188: 2445–54. Sharkey DJ, Macpherson AM, Tremellen KP, Robertson SA. Seminal plasma differentially regulates inflammatory cytokine gene expression in human cervical and vaginal epithelial cells. Mol Hum Reprod 2007;13:491–501. Pandya IJ, Cohen J. The leukocytic reaction of the human uterine cervix to spermatozoa. Fertil Steril 1985;43:417–21. Thompson LA, Barratt CL, Bolton AE, Cooke ID. The leukocytic reaction of the human uterine cervix. Am J Reprod Immunol 1992;28:85–9. O'Leary S, Jasper MJ, Warnes GM, Armstrong DT, Robertson SA. Seminal plasma regulates endometrial cytokine expression, leukocyte recruitment and embryo development in the pig. Reprod 2004;128:237–47. Robertson SA. Seminal fluid signaling in the female reproductive tract: lessons from rodents and pigs. J Anim Sci 2007;85:E36–44. Hutter H, Dohr G. HLA expression on immature and mature human germ cells. J Reprod Immunol 1998;38:101–22. Schjenken JE, Robertson SA. Seminal fluid and immune adaptation for pregnancy—comparative biology in mammalian species. Reprod Domest Anim 2014;49:27–36. Aitken RJ, Baker MA. Oxidative stress, spermatozoa and leukocytic infiltration: relationships forged by the opposing forces of microbial invasion and the search for perfection. J Reprod Immunol 2013;100:11–9. Koppers AJ, Mitchell LA, Wang P, Lin M, Aitken RJ. Phosphoinositide 3kinase signalling pathway involvement in a truncated apoptotic cascade associated with motility loss and oxidative DNA damage in human spermatozoa. Biochem J 2011;436:687–98. Robertson SA, Sharkey DJ. The role of semen in induction of maternal immune tolerance to pregnancy. Semin Immunol 2001;13:243–54. Robertson SA, Prins JR, Sharkey DJ, Moldenhauer LM. Seminal fluid and the generation of regulatory T cells for embryo implantation. Am J Rep Immuno 2013;69:315–30. Guerin LR, Moldenhauer LM, Prins JR, Bromfield JJ, Hayball JD, Robertson SA. Seminal fluid regulates accumulation of FOXP3þ regulatory T Cells in the preimplantation mouse uterus through expanding the FOXP3þ cell pool and CCL19-mediated recruitment. Biol Reprod 2011;85:397–408. Robertson SA, Guerin LR, Bromfield JJ, Branson KM, Ahlstrom AC, Care AS. Seminal fluid drives expansion of the CD4þCD25þ T regulatory cell pool and induces tolerance to paternal alloantigens in mice. Biol Reprod 2009;80:1036–45. Alt C, Laschinger M, Engelhardt B. Functional expression of the lymphoid chemokines CCL19 (ELC) and CCL 21 (SLC) at the blood-brain barrier suggests their involvement in G-proteindependent lymphocyte recruitment into the central nervous system during experimental autoimmune encephalomyelitis. Eur J Immunol 2002;32:2133–44. Engelhardt B. Molecular mechanisms involved in T cell migration across the blood-brain barrier. J Neural Transm 2006;113:477–85. O'Leary S, Jasper MJ, Robertson SA, Armstrong DT. Seminal plasma regulates ovarian progesterone production, leukocyte recruitment and follicular cell responses in the pig. Reproduction 2006;25:1–12. Gangnuss S, Sutton-McDowall ML, Robertson SA, Armstrong DT. Seminal plasma regulates corpora lutea macrophage populations during early pregnancy in mice. Biol Reprod 2004;71:1135–41.
VOL. - NO. - / - 2016
11
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298
VIEWS AND REVIEWS 135. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of 1299 peripheral CD4þCD25- naive T cells to CD4þCD25þ regulatory T cells 1300 by TGF-beta induction of transcription factor Foxp3. J Exp Med 2003; 1301 198:1875–86. 1302 136. Baratelli F, Lin Y, Zhu L, Yang S-C, Heuze-Vourc'h N, Zeng G, et al. Prosta1303 glandin E2 induces FOXP3 gene expression and T regulatory cell function in 1304 human CD4þ T Cells. J Immunol 2005;175:1483–90. 1305 137. Sharkey DJ, Macpherson AM, Tremellen KP, Mottershead DG, Gilchrist RB, Robertson SA. TGF-beta mediates proinflammatory seminal fluid signaling 1306 in human cervical epithelial cells. J Immunol 2012;189:1024–35. 1307 138. Gutsche S, vonWolff M, Strowitzki T, Thaler CJ. Seminal plasma induces 1308 mRNA expression of IL-1beta, IL-6 and LIF in endometrial epithelial cells 1309 in vitro. Mol Hum Reprod 2003;9:785–91. 1310 139. Robertson SA, Ingman WV, O'Leary S, Sharkey DJ, Tremellen KP. Trans1311 forming growth factor beta—a mediator of immune deviation in seminal plasma. J Reprod Immunol 2002;57:109–28. 1312 140. Clark DA, Fernandes J, Banwatt D. Prevention of spontaneous abortion in 1313 the CBA x DBA/2 mouse model by intravaginal TGFbeta and local recruit1314 ment of CD4þ8þ FOXP3þ cells. Am J Reprod Immunol 2008;59:525–34. 1315 141. Robertson SA, Mau VJ, Tremellen KP, Seamark RF. Role of high molecular 1316 weight seminal vesicle proteins in eliciting the uterine inflammatory 1317 response to semen in mice. J Reprod Fertil 1996;107:265–77. 1318 142. Blois SM, Kammerer U, Alba Soto C, Tometten MC, Shaikly V, Barrientos G, et al. Dendritic cells: key to fetal tolerance? Biol Reprod 2007;77:590–8. 1319 143. Robertson SA, Allanson M, Mau VJ. Molecular regulation of uterine leuko1320 cyte recruitment during early pregnancy in the mouse. Troph Res 1998;11: 1321 101–20. 1322 144. Amodio G, Mugione A, Sanchez AM, Vigano P, Candiani M, Somigliana E, 1323 et al. HLA-G expressing DC-10 and CD4(þ) T cells accumulate in human 1324 decidua during pregnancy. Hum Immunol 2013;74:406–11. 1325 145. Remes LF, Rodriguez RC, Sabatte J, Cabrini M, Jancic C, Ostrowski M, et al. Semen promotes the differentiation of tolerogenic dendritic cells. J Immu1326 nol 2012;189:4777–86. 1327 146. Schjenken JE, Robertion SA. Induction of endogenous miR223 expression 1328 by sperm in the female reproductive tract following mating in mice. Am J 1329 Reprod Immunol 2013;70:18–9. 1330 147. Tompkins AJ, Chatterjee D, Maddox M, Wang J, Arciero E, Camussi G, 1331 et al. The emergence of extracellular vesicles in urology: fertility, cancer, biomarkers and targeted pharmacotherapy. J Extracell Vesicles 2015;4: 1332 23815. 1333 Q3 148. Rullston T, Ohlsson Teague EM, Palmer NO, DeBlasio MJ, Mitchell M, 1334 Corbett M, et al. Paternal obesity initiates metabolic disturbances in two 1335 generations of mice with incomplete penetrance to the F2 generation 1336 and alters the transcriptional profile of testis and sperm microRNA content. 1337 FASEB J 2013;27:4226–43. 1338 149. Liu WM, Pang RT, Chiu PC, Wong BP, Lao K, Lee KF, et al. Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proc 1339 Natl Acad Sci U S A 2012;109:490–4. 1340 150. Larsen MH, Bzorek M, Pass MB, Larsen LG, Nielsen MW, Svendsen SG, 1341 et al. Human leukocyte antigen-G in the male reproductive system and 1342 in seminal plasma. Mol Hum Reprod 2011;17:727–38. 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357
151. 152.
153.
154.
155.
156.
157.
158.
159.
160.
161.
162.
163.
164.
165. 166. 167.
Ristich V, Liang S, Zhang W, Wu J, Horuzsko A. Tolerization of dendritic cells by HLA-G. Eur J Immunol 2005;35:1133–42. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4þCD25highFOXP3þ regulatory T cells. Stem Cells 2008;26:212–22. Guerin LR, Prins JR, Robertson SA. Regulatory T-cells and immune tolerance in pregnancy: a new target for infertility treatment? Hum Reprod Update 2009;15:517–35. Quinn KH, Lacoursiere DY, Cui L, Bui J, Parast MM. The unique pathophysiology of early-onset severe preeclampsia: role of decidual T regulatory cells. J Reprod Immunol 2011;91:76–82. Arruvito L, Sanz M, Banham AH, Fainboim L. Expansion of CD4þCD25þand FOXP3þ regulatory T cells during the follicular phase of the menstrual cycle: implications for human reproduction. J Immunol 2007;178:2572–8. Jin LP, Chen QY, Zhang T, Guo PF, Li DJ. The CD4þCD25 bright regulatory T cells and CTLA-4 expression in peripheral and decidual lymphocytes are down-regulated in human miscarriage. Clin Immunol 2009;133:402–10. Basso O, Christensen K, Olsen J. Higher risk of pre-eclampsia after change of partner. An effect of longer interpregnancy intervals? Epidemiology 2001;12:624–9. Kho EM, McCowan LM, North RA, Roberts CT, Chan E, Black MA. Duration of sexual relationship and its effect on preeclampsia and small for gestational age perinatal outcome. J Reprod Immunol 2009;82:66–73. Bromfield JJ, Schjenken JE, Chin PY, Care AS, Jasper MJ, Robertson SA. Maternal tract factors contribute to paternal seminal fluid impact on metabolic phenotype in offspring. Proc Natl Acad Sci U S A 2014;111:2200–5. Crawford G, Ray A, Gudi A, Shah A, Homburg R. The role of seminal plasma for improved outcomes during in vitro fertilization treatment: review of the literature and meta-analysis. Hum Reprod Update 2015;21: 275–84. Seavey MM, Mosmann TR. Paternal antigen-bearing cells transferred during insemination do not stimulate anti-paternal CD8þ T cells: role of estradiol in locally inhibiting CD8þ T cell responses. J Immunol 2006;177: 7567–78. Erlebacher A, Vencato D, Price KA, Zhang D, Glimcher LH. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest 2007;117:1399–411. Faustman DL, Steinman RM, Gebel HM, Hauptfeld V, Davie JM, Lacy PE. Prevention of rejection of murine islet allografts by pretreatment with anti-dendritic cell antibody. Proc Natl Acad Sci U S A 1984;81:3864–8. Suchin EJ, Langmuir PB, Palmer E, Sayegh MH, Wells AD, Turka LA. Quantifying the frequency of alloreactive T cells in vivo: new answers to an old question. J Immunol 2001;166:973–81. Bonney EA. Preeclampsia: a view through the danger model. J Reprod Immunol 2007;76:68–74. Tollner TL, Bevins CL, Cherr GN. Multifunctional glycoprotein DEFB126—a curious story of defensing-clad spermatozoa. Nat Rev Urol 2012;9:365–75. Matzinger P. The danger model: a renewed sense of self. Science 2002; 296:301–5.
VOL. - NO. - / - 2016
12
REV 5.4.0 DTD FNS30413_proof 28 July 2016 1:08 pm ce DPH
1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416