Endometrium

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Clinical Assessment and Management of the Endometrium in Recurrent Early Pregnancy Loss

Bansari G. Patel, M.D.,1 and Bruce A. Lessey, M.D., Ph.D.2

ABSTRACT

Unexplained and recurrent loss of pregnancies is a heartbreaking and frustrating condition. The routine diagnostic workup for pregnancy loss includes hormonal evaluation, screening for genetic or chromosomal defects, immunologic and thrombophilic testing, and evaluation of congenital or acquired Mu llerian defects. In cases of idiopathic pregnancy loss, defects in endometrial receptivity are increasingly being investigated. The role of the endometrium in pregnancy loss has historical roots but remains controversial. Exciting new directions based on microRNAs, proteomics, and epigenetics promises to keep this area of investigation both interesting and complex. With each new diagnostic and therapeutic biomarker identied comes a greater potential for diagnosis and treatment of women. The clinical assessment of the endometrium remains an important part of the investigation of couples with unexplained pregnancy loss.
KEYWORDS: Miscarriage, endometrium, endometriosis, luteal phase defect, hydrosalpinges, pregnancy loss

oss of a single pregnancy is a common event, occurring up to 35% of conceptions.1 Recurrent pregnancy loss (RPL), however, affects only 3 to 5% of women attempting conception.2,3 Factors that increase the risk for RPL include the loss of a chromosomally normal pregnancy, pregnancy loss after the rst trimester, and concomitant infertility. Only half of all miscarriages that occur in the general population can be assigned a cause, including chromosomal abnormalities, immunological, endocrine (luteal phase defect or diabetes), anatomical, thrombophilic, infectious, or iatrogenic causes (smoking and caffeine).3,4 This gap in our diagnostic accuracy for up to 50% of losses3,5 might be lled by an awareness of conditions such as endometriosis or
1 Department of Obstetrics and Gynecology; 2University Medical Group, Greenville Hospital System, Greenville, South Carolina. Address for correspondence and reprint requests: Bruce A. Lessey, M.D., Ph.D., Department of Obstetrics and Gynecology, University Medical Group, Greenville Hospital System, 890 W. Faris Rd., Suite 470, Greenville, SC 29605 (e-mail: blessey@ghs.org). Recurrent Early Pregnancy Loss; Guest Editor, Mary D.

tubal factor that appear to alter endometrial receptivity, leading to implantation failure.6 In fact, many of the factors that render women infertile are a likely cause of unexplained RPL. The efciency of implantation in the human is relatively low compared with other mammals. It is estimated that 10 million couples will seek medical assistance for infertility and/or RPL this year. Up to a third of in vitro fertilization (IVF) pregnancy losses are thought due to poor embryo quality, and it is estimated that up to 60% result from implantation defects. It is safe to say that most women with defects in endometrial receptivity go unrecognized because most couples with unexplained RPL do not have a thorough evaluation of
Stephenson, M.D., M.Sc. Semin Reprod Med 2011;29:491506. Copyright # 2011 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662. DOI: http://dx.doi.org/10.1055/s-0031-1293203. ISSN 1526-8004.

491

Downloaded by: World Health Organization ( WHO). Copyrighted material.

492

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

the endometrium. At the same time, despite the availability of specic biomarkers of endometrial receptivity, their use remains low due in part to a lack of randomized controlled clinical trials that validate their usefulness. In addition, a lack of a clear mechanism regarding how endometriosis causes pregnancy loss adds to this lack of consensus regarding the use of endometrial biomarkers. This article reviews the clinical assessment of the endometrium in the setting of RPL; however, much of the literature also extends to studies of unexplained infertility (UI) that we believe is a continuum with unexplained pregnancy loss.

Figure 1 Endometrial dating criteria of Noyes et al.19 This landmark study was published as the rst article in the inaugural issue of Fertility and Sterility. The histological patterns chosen represent an idealized representation of the observed changes in the endometrium throughout the secretory phase of the menstrual cycle.

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE ENDOMETRIUM The endometrium is a steroid target tissue7,8 that forms the inner mucosal lining of the uterus. It is composed of epithelial and stromal cells, and together with the myometrium is derived from the paramesonephric ducts that fuse during early embryogenesis. Like the kidney, the endometrium establishes its epithelial compartment from mesenchymal transformation, rather than preexisting embryonic epithelium.9 Adult endometrium consists of a basal and functional layer made up of columnar epithelial cells, supported by stromal and vascular elements. In lieu of pregnancy, the cycling endometrium is shed monthly and renews itself from both the exposed

stromal and glandular fragments. A 2009 study using hysteroscopic examination combined with scanning electron microscopy suggested that reestablishment of the luminal epithelium over the basal layer occurs through mesenchymal-epithelial transformation, reminiscent of its embryological origins.10 In addition to epithelial and stromal components, up to 50% of the endometrium is comprised of leukocytes. The immune system can be divided into innate immunity including monocytes, macrophages, dendritic cells, neutrophils, basophils, and mast cells, and uterine natural killer cells (uNKs) from the lymphoid lineage. The adaptive immune system includes the T and B cells, which require cells from innate immunity to establish an immunological memory.11 Bone marrow derived cells are known to trafc to the endometrium, through steroid-regulated chemokine production,12,13 that may have implications for pregnancy loss related to systemic inammation.1418 Given these dynamic cyclic changes along with easy access to the endometrium by ultrasound, hysteroscopy, or endometrial sampling, there are many ways to evaluate endometrial function. Histological changes of the endometrium were classied by the landmark studies of Noyes and colleagues, who established the original dating criteria for assess of endometrial development (Fig. 1).19 Simultaneously, Georgina Seegar Jones in

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

493

194920 developed the concept of what is now called luteal phase defect (LPD). The diagnosis of LPD remains one of the most studied areas of endometrial assessment and even today is of uncertain clinical importance.2124

Figure 2 (A) Schematic of the stages of implantation starting at ovulation and extending until the time of embryo apposition, adhesion, and invasion. (B) image from the Carnegie series showing stage 5a embryo that has completed its transition through the luminal epithelium and is becoming invested in the endometrial stroma (decidua).

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE IMPLANTATION WINDOW AND DEFECTS IN UTERINE RECEPTIVITY RPL, like UI, can be attributed to implantation failure.6,2527 The success of an early pregnancy requires synchronous interactions between the endometrium, corpus luteum (CL), and embryo; conversely, delayed implantation contributes to pregnancy loss.28,29 Hemochorial placentation in human and the higher primates occurs in stages (Fig. 2A) but requires apposition (initial interaction) between the embryo and the endometrial surface.30,31 The luminal surface epithelium is the primary barrier to embryo implantation throughout the menstrual cycle, except during a narrow window of implantation (WOI). Endometrial receptivity is acquired during the midsecretory phase in the

normal fertile women 5 to 7 days after ovulation, reecting the expression of a vast repertoire of genes that regulate multiple biochemical pathways facilitating embryouterine adhesion, embryonic survival, and subsequent invasion of the nascent embryo (Fig. 2B).3234 The concept of a receptor-mediated mechanism of implantation is now well established.3538 Based on numerous studies, multiple mechanisms for the initial embryoendometrial attachment phase have been envisioned (Fig. 3). Apposition is a brief event, followed quickly by intrusive probing by the embryonic trophoblast between the weakened epithelialepithelial lateral plane.39 Lateral adhesion allows cytotrophoblast invasion, digesting through the basement membrane into the underlying stroma (decidua).40,41 Once within stroma, vascular, immunological, and stromal contributions both stimulate and limit invasion. These complex interactions and communications provide a wealth of potential biomarkers for the evaluation of endometrial receptivity. Indeed, much of our current understanding of endometrial physiology is based on individual investigations of these many proteins that orchestrate implantation.

494

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

Figure 3 Models of embryoendometrial attachment (used with permission from Seminars in Reproductive Medicine, 2007).6 Adhesion molecules serve many functions throughout the body. Endometrialembryo interactions likely involve one or more of these mechanisms. As shown, many different combinations of ligands and receptors have been proposed, as outlined in the text. Interactions can involve integrins, extracellular matrix molecules such as bronectin, tastin, and trophinin, lectins, and selectins. Although not mutually exclusive, the primary receptor for the embryo is likely a loose attachment as proposed for L-selectin and its ligand that allows a transient attachment phase that precedes invasion. Other mechanisms as depicted may have other roles including immune modulation and cell signaling at the time of implantation. Used with permission from Thieme Medical Publishers, Inc.6

Based on numerous studies, it is estimated that the WOI extends from postovulatory days 6 to 10 (corresponding to cycle day 20 to 24). Wilcox and colleagues conrmed the timing of this window in normal fertile women and demonstrated that delayed implantation results in a higher risk of miscarriage.29 Based on a concept of asynchrony between the endometrium, ovary, and embryo, histology delay in the endometrium would logically move the WOI, providing a mechanism for pregnancy loss in women with LPD. The ovarian CL, like the endometrium, has an ephemeral window of receptivity, and delayed implantation likely occurs when the CL is less able to respond and protect the nascent embryo from menstruation. Endometriosis appears to play a role in unexplained RPL, as it does in UI.5,42 This disease of retrograde menstruation and inammation is an estrogen-dependent condition found in up to 70% of women with pain and 25 to 40% of infertile women.43,44 Its role in RPL has not been as well studied but could account for the bulk of idiopathic pregnancy loss. The presence of endometrial cells outside the uterine cavity results in

biochemical alterations in the eutopic endometrium leading to elevated estrogen sensitivity45 and progesterone resistance,46,47 resulting in absent or delayed expression of those proteins essential for establishment or maintenance of pregnancy.48,49 The remainder of this article reviews the established and new opportunities for the clinical assessment of the endometrium in women with RPL.

CLINICAL ASSESSMENT OF THE ENDOMETRIUM Endometrial Histology Noyes and colleagues rst described the criteria for endometrial dating in 1950. This landmark study was published in the rst issue of Fertility and Sterility, underscoring the perceived importance of endometrial receptivity early on.19,50 The study consisted of cataloging histological changes in endometrial biopsies from infertile women that were therefore abnormal by denition. After 60 years, this study was recently

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

495

repeated and validated using only fertile controls.23 The histological criteria established for endometrial dating provide a helpful idealization of endometrial progression during the secretory phase; however, they appear to lack the precision and reproducibility required for individual patients. Excessive variability in these criteria exists between both subjects and observers, rendering accurate prospective chronological assignment through the menstrual cycle a challenge.23 Despite their shortcomings, these criteria remain a primary method for the clinical assessment of the endometrium.23,24

Hormonal Assessment The state of endometrial receptivity in RPL could be assessed indirectly by measuring the endocrine milieu during the menstrual cycle. Serum levels of thyroidstimulating hormone and prolactin should always be considered, along with follicle-stimulating hormone and estradiol on day 3 of the cycle as a measure of ovarian reserve as well as ruling out thyroid disease and hyperprolactinemia. Patients with polycystic ovary syndrome (PCOS), who generally have a poor reproductive performance and increased risk of miscarriage,51 may require additional hormonal testing. Abnormalities in the endocrine milieu of PCOS can disrupt hypothalamic-pituitary-ovarian function leading to alterations in the endometrium contributing to implantation failure.52,53 Assessment of insulin and androgen levels is obligatory in these women because adverse effects of hyperinsulinemia or hyperandrogenemia may lead to a nonreceptive endometrium.51,5457 Furthermore, hyperandrogenemia and hyperinsulinemia have also been associated with LPD and alterations in reproductive function. Progesterone is the sentinel marker of the luteal phase and essential for pregnancy success.58 Serum measurements of progesterone are a clinical tool for assessment of ovulation and a surrogate marker for endometrial response.5961 The use of progesterone as a measure of reproductive competence is not without concerns. A low serum progesterone level may result from suboptimal follicular development leading to RPL or infertility.62 The activity of the CL depends on a tonic level of luteinizing hormone (LH) secretion from the pituitary. Strott and colleagues studied women with a short luteal phase (dened as <10 days in duration) and reported a relative deciency in gonadotropin levels, as well as LH surges of reduced magnitude.63 A gonadotropin deciency was created as a model for LPD, using antibodies to LH, noting a prompt decline in circulating progesterone levels following treatment.64 As expected, administration of gonadotropin-stimulating hormone antagonists produces a similar decrease in progesterone levels65 because progesterone is secreted in a pulsatile manner, corresponding to LH pulse frequency.66 Any

change in pulse generator could theoretically alter progesterone secretion, leading to LPD, but given the wide range of progesterone levels between its peak and trough can give a sixfold difference in measurements, raising questions about the usefulness of a single measurement. Thus the use of progesterone levels in the assessment of LPD would likely lead to more questions than answers.49,67 Progesterone is known to have a direct and indirect stimulatory action on key endometrial proteins, hence rendering it critical to the success of an early pregnancy.6881 Progesterone downregulates endometrial steroid receptors (estrogen receptors and progesterone receptors) during the midsecretory phase. The phenomena of inadequate progesterone production or secondary progesterone response (progesterone resistance),49 may be responsible for alterations in steroid receptor downregulation noted in the endometrium of some women with RPL. Persistent estrogen or progesterone receptors during the midluteal phase may alter that balance of estrogen and progesterone-mediated effects, and alter the paracrine dynamics that underlie the acquisition of endometrial receptivity.75,81 Thus both qualitative and quantitative study of steroid receptors, as well as the enzymes that metabolize these steroids, may serve as a useful tool for the assessment of endometrial function.45,75,82,83 Because progesterone is essential for the success of pregnancy, low progesterone action would logically lead to delayed endometrial maturation62 or differentiated function.49,84 The study by Usadi and colleagues in 2008, however, demonstrated signicant plasticity in the response to articially lowered progesterone levels during a mock luteal phase. In normal fertile women, they failed to demonstrate observable effects on endometrial histology.22 Functional studies on these samples using DNA microarray do, however, reveal dose-dependent differences in gene expression, suggesting that histology may be a blunt tool to study the true effect of progesterone on endometrial function.85

Endometrial Ultrastructure
PINOPODS

In 1972, during a series of scanning electron microscopic studies of receptive endometrium, morel-like apical projections from the rat uterine luminal surface were observed coincident with the implantation window. Enders and Nelson in 1973 determined that these projections from the surface cells were involved in the nonspecic uptake of uid from the uterine lumen. The term pinopods (translated from drinking foot; also known as pinopodes, uterodomes) described these bleb-like projections (Fig. 4AC).86 Pinopods have been extensively described in the human endometrium and suggested to be markers

Downloaded by: World Health Organization ( WHO). Copyrighted material.

496

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

Figure 4 Endometrial pinopods (A) are present in the secretory phase on the luminal surface and appear to be the same structures decorated with luminal biomarkers, such as (B) anb3 integrin and its presumed ligand (C) osteopontin (OPN).

Endometrial Imaging Imaging techniques are readily available and commonly used for the clinical evaluation of the uterus and endometrium.116119 Transvaginal ultrasound is now available to most practitioners and offers a convenient method to assess endometrial thickness, contour, and pelvic anatomy. Ultrasound is a sensitive method to identify abnormalities that could contribute to RPL including a uterine septum or acquired defects such as uterine broids or polyps. Compared with laparoscopy, ultrasound has been reported to have a sensitivity of 86%, a specicity of 98%, a positive predictive value of 99%, and a negative predictive value of 79% for the detection of pathology.119 During the proliferative phase the endometrium grows to a thickened strip with a trilaminar appearance. Some investigators found that a thin endometrium is associated with a poor outcome or reduced pregnancy rates,120124 but others found no predictive value of endometrial thickness.125127 A thicker endometrium was associated with positive pregnancy outcome in early

SONOHYSTEROGRAPHY

Two-dimensional (2D) and three-dimensional (3D) ultrasound are increasingly performed with uid instillation

Downloaded by: World Health Organization ( WHO). Copyrighted material.

of uterine receptivity8794 in both fertile and infertile women.93102 Although similar in structural appearance to those described in the rat, pinopods in the human endometrium do not appear to possess pinocytic activity.103 Pinopods are expressed in endometrial culture and might be a preferred site for embryo attachment.104,105 These expanded blebs of plasma membrane appear to be a site of various endometrial proteins including integrins,97 leukemia inhibitory factor (LIF), glycodelin, osteopontin and galectin-9 and galectin-3, L-selectin ligands, and decay accelerating factor.106112 Two prospective randomized studies did not conrm the expected pattern of pinopod expression in cycling women, drawing into question their signicance.113,114 Quinn et al found that pinopodes can be detected in the progesterone-exposed endometrium for an extended period of time, thus casting doubt on their role as markers for the implantation window in the human endometrium.115

IVF studies.128130 In cycles where conception occurred, endometrial thickness was reported to increase compared with nonconception cycles.131 Endometrial thickness is measured at the endometrialmyometrial interface through the sagittal plane of the uterine body, 1 cm from the fundus.132 Errors in endometrial thickness measurements can occur if an oblique section of the uterus is captured or if the uterus is not imaged along its entire length to achieve maximal fundal endometrial thickness. Despite the dependence on steroid hormones, studies suggested that endometrial growth does not always correlate well with hormone levels,124 and that neither thickness nor echogenic pattern correlated well with endometrial histology.133 Doppler ow studies are noninvasive ultrasound methods used to evaluate the blood ow to the uterus and endometrium. The extent to which blood ow and Doppler ndings predict the rate of implantation remains an active area of investigation. Blood ow changes during the menstrual cycle have been reported134,135 and correlate with serum progesterone levels.136 Uterine artery blood ow resistance has been reported to be predictive of implantation potential.137140 One report suggested that Doppler ndings also correlated with immunohistochemical biomarkers of uterine receptivity.141 Elevated uterine impedance was reported to be abnormal in RPL as well.136 A recent report found that women with an end-diastolic blood ow, an endometrial-subendometrial blood ow, and a multilayered endometrium were more likely to have a successful pregnancy than women without one or more of these signs.117 Schild, in contrast, found that blood ow in both the uterine and spiral arteries measured by power Doppler showed no correlation with implantation rates.142 There is presently no consensus about the predictive potential of measurements of ow or resistance in the endometrial blood vessels.119

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

497

into the uterine cavity (sonohysterography) to allow evaluation of the endometrium in women with RPL as well as those with infertility or abnormal bleeding.143147 These techniques are highly sensitive at detecting intrauterine pathology.116,144,147149 Compared with normal sonohysterograms, polyps or broids are easily detected when saline is introduced into the cavity and can be well displayed using 2D and 3D technology. A uterine septum may predispose to RPL and can be routinely detected by sonohysterography. Although patients with signicant pathology will ultimately undergo more invasive procedures such as hysteroscopy and/or endometrial dilation and curettage to diagnose and remove the abnormality, sonohysterogram is an essential screening tool to assess the uterine cavity and aid in the clinical assessment of the endometrium.

Biomarkers of Endometrial Receptivity A biomarker has been dened as a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention. Emerging evidence indicates that the endometrium can be biochemically defective without a histological delay.48,49,150 Derangements in the endometrium in otherwise normally developed endometrium have greatly expanded the possibilities for the use of alternative methods to assess endometrial receptivity as a predictor of future reproductive success.151 The extracellular matrix (ECM) along with its associated cell adhesion molecules (CAMs) maintain tissue integrity and hormonal responsiveness within tissues.152 Changes in the organization and composition of ECM in the human endometrium during the menstrual cycle and early pregnancy was assessed by immnunouorescence,153,154 showing cycle-specic changes in ECM components and CAMS that in turn suggested a role in embryoendometrial interactions.155,156 Integrins are a family of CAMs that function as both cell substratum and cellcell adhesion receptors. They interact with ECM ligands and other CAMs as well as matrix metalloproteinases to participate in a wide variety of physiological processes.157 Dynamic changes in integrin expression occur during the menstrual cycle and into pregnancy.67,154,158160 The three amino acid motif arggly-asp (RGD) was implicated in the process of implantation by several investigators.161163 RGD is present on many ECM ligands in the receptive endometrium, including osteopontin, tenascin, insulinlike growth factor-binding protein 1, and bronectin.164167 A functional blockade using RGD peptides and anti-RGD snake venom components was shown to effectively block implantation or attachment of embryos, suggesting a critical role of integrins and related ligands in the cascade of events leading to successful implantation.162,166,168,169

SELECTINS/CADHERINS

The search for a true receptor that mediates embryo attachment has been a goal for many implantation researchers35,36 and has led to numerous candidates,158,176179 including growth factor/receptor pairs, CAMS, ECM, and members of the cell adhesion families. Recent evidence supports L-selectin, a member of the selectin family, as a key mediator of the interaction between the uterus and embryo during the initial attachment.180 The presumed endometrial ligand for L-selectin appears to be a sialyl glycoprotein associated with the Lewis-X family that is recognized by the monoclonal antibody MECA-79. This factor has now been studied in normal cycling women during the menstrual cycle.181 Additionally, ligands of L-selectin have been suggested to be clinically useful markers of endometrial receptivity and may be absent in women with RPL.111
CYTOKINES

The expression of cytokines and growth factors along with their receptors during the menstrual cycle has been implicated in both endometrial development and implantation. LIF was one of the rst glycoproteins demonstrated to be critical for implantation. Null mutation of the LIF gene in female mice resulted in complete lack of implantation and exhibited decidualization failure.182184 Interestingly, this phenotype could be rescued by administration of exogenous LIF. Interleukin (IL)-11, another member of this IL-6 family, has also been implicated in decidualization. Both LIF and IL-11 signal through the

Downloaded by: World Health Organization ( WHO). Copyrighted material.

Integrins are perhaps the best characterized of the endometrial markers. In 1992, constitutive and cycledependent patterns of integrin expression were rst described.158,170 Certain integrin moieties appeared to be regulated within the cycling endometrium, and disruption of their expression was reported to have adverse effects on uterine receptivity and fertility. Three integrins were noted to be coexpressed and receptive to endometrium only during the putative window of implantation (Fig. 5).67 The anb3 integrin appears on apical endometrial epithelium at the opening of the window of implantation around cycle day 20 or 21 and is present into pregnancy. Daftary and colleagues also showed that b3-integrin subunit expression in the endometrium is regulated by HOXA10.171 This factor was more recently shown to be reduced in women with endometriosis, hydrosalpinges, and broids.172 Studies on integrins in RPL in 2006 found no difference in expression in women with unexplained RPL,173 but more recently, increased expression of a 1 and a 4 integrins was seen in some women with miscarriage.174 A polymorphism (A1/A2) in the b3integrin subunit was found to be signicantly associated with fetal loss before 10 weeks of gestation.175

498

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

Figure 5 Endometrial integrins throughout the menstrual cycle. Endometrial integrins undergo changes in expression throughout the menstrual cycle. The expression pattern of three integrins including a1b1, anb3, and a4b1 appear to frame the window of implantation that was later dened by Wilcox and colleagues.29 The anb3 integrin is located on the endometrial luminal surface appearing at the time of implantation and is absent in some women with implantation defects. This protein also predicts in vitro fertilization success and can be used to identify women with endometriosis who have otherwise unexplained infertility. Used with permission from the American Society for Reproductive Medicine and Elsevier.67

gp130 receptor. Numerous studies have imputed reductions in LIF and related proteins in women with implantation failure and infertility.185189 We reported decreased LIF expression in the endometrium of PCOS women190 that could account for their more frequent miscarriages compared with normal women. LIF remains one of the most validated biomarkers for endometrial receptivity.
DNA MICROARRAY

In the future, assessment of RPL will undoubted use panels of biomarkers that dene different and distinct types of endometrial defects. The process of biomarker discovery has been dramatically accelerated by highthroughput techniques such as DNA microarray, using both normal and pathological endometrium.34,80,191205 The advent of DNA microarrays has enabled expeditious identication of new pathways, signaling paradigms, and metabolic processes. The characterization of the endometrium throughout the normal menstrual cycle34 has set the stage for a much better understanding of the

factors involved in endometrial development and menstruation.192 This technique is not only enabling researchers to rediscover known biomarkers but also allows discovery of new pathways and unsuspected proteins.192,195,206 Development of these emerging techniques in conjunction with bioinformatics and proteomics will likely revolutionize our current understanding of RPL. Studies in infertility patients have already resulted in the new concept of progesterone resistance,49 with clear relevance to the study of RPL.46 MicroRNAs (miRNAs) are small nonprotein coding RNAs that regulate specic posttranscriptional messenger RNAs targeting them for degradation. Research on these molecules has recently exploded and will likely lead to the next class of biomarkers of endometrial dysfunction. Genes coding for miRNA represent 1 to 4% of the genes in higher organisms and are highly processed sequences derived from larger RNA transcripts. These small precursor miRNAs form a single stranded miRNA that binds to complementary sequences on target mRNA preventing their transcription.207209 Recent

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

499

characterization of miRNAs has added to our understanding of the regulation of gene expression in the normal and diseased endometrium.210214
IMMUNE CELLS OF THE ENDOMETRIUM

MANAGEMENT OF ENDOMETRIAL RECEPTIVITY DEFECTS IN RECURRENT PREGNANCY LOSS Only 50% of RPLs are explained. Many of these cases exhibit signs of mild ovulatory dysfunction and/or biochemical progesterone resistance including women with endometriosis and PCOS.46,190 After endocrine abnormalities (diabetes mellitus, hyperprolactinemia, and thyroid disease) are diagnosed and treated and mechanical causes involving the uterus (uterine septae, broids, polyps, Ashermans syndrome) are ruled out or corrected, one should consider other underlying causes of endometrial receptivity including PCOS, endometriosis, and subtle tubal disease.6 PCOS is a known risk factor for miscarriage.228230 Treatment of PCOS and associated hyperandrogenism and hyperinsulinemia has been the subject of several recent reviews.231234 Aromatase inhibitors in particular appear to be superior compared with clomiphene citrate in this condition,235237 although controlled trials for RPL are limited. In ovulatory women with PCOS, endometriosis can be a cofactor because many women with heavy irregular bleeding would be at risk for development of endometriosis. Elevated body mass index is also a risk factor for endometriosis.238

SUMMARY AND CONCLUSIONS Implantation failure and defects in endometrial receptivity are associated with a third to a half of all RPLs. Consensus for a systematic clinical assessment of the endometrium is lacking, although many opportunities exist but remain investigational. Endometrial histology appears to be the weakest index of function, given the variation seen between observers and between patients and the lack of correlation between histology and gene expression. The use of biomarkers for endometrial assessment is rapidly evolving, with the recent advancements using DNA microarray techniques. As new biomarkers become available as a test for endometriosis, the relationship between subtle endometriosis and RPL may become better established. The next frontier in the diagnosis and potentially treatment of RPL will likely be based on a study of the epigenetic changes in the cycling endometrium.244 Therapeutics directed at underlying dysfunction will likely provide better treatment options for women with this distressing condition.

REFERENCES
1. Gilchrist DM, Livingston JE, Hurlburt JA, Wilson RD. Recurrent spontaneous pregnancy loss. Investigation and reproductive follow-up. J Reprod Med 1991;36(3):184188 2. Katz VL, Kuller JA. Recurrent miscarriage. Am J Perinatol 1994;11(6):386397

Downloaded by: World Health Organization ( WHO). Copyrighted material.

Successful embryo implantation requires a complex and coordinated series of biochemical changes in the endometrium that allows for a controlled invasion of the fetal allograft at the interaction of maternalfetal compartments.35 Loss of the immunotolerant regulatory T cells and their associated interactions appear to account for both infertility and RPL observed in women with endometriosis or other inammatory diseases.215222 Altered T cells in the normal midsecretory phase could also affect the number of uNK cells. Studies report that the number of CD56bright NK cells are decreased in women with unexplained miscarriage223,224 and unexplained IVF failure.225 Further, monocytes can directly alter endometrial receptivity in the mouse model.226 Endometrial immune cells, their cytokines and chemokines, and the stromal paracrine signals such as IL-15 are all potential biomarkers that could be exploited to understand aberrant endometrium associated with unexplained recurrent pregnancy loss (uRPL).216,227 Many of these immunological cells and messages are potential biomarkers for the assessment of endometrial competence but will have to await further characterization in this rapidly expanding area of research.

Prior miscarriage is a risk factor for endometriosis,239 and endometriosis has been associated with RPL.5,42,240,241 Laparoscopy is the mainstay for diagnosis and treatment of endometriosis and should be considered for uRPL, especially when symptoms of dysmenorrhea are present. Medical management, at least for infertility associated with endometriosis, has not been proven efcacious.242 In our experience, women with mild endometriosis and RPL can be effectively treated without surgery. Because women with uRPL often conceive easily, the pregnancy losses likely occur as a result of disrupted endometrial/CL synchrony.28 Delayed implantation, due to a shift in the WOI, leads to inadequate CL response to rising human chorionic gonadotropin (hCG) levels. In a past systematic review, hCG was shown to be superior to luteal progesterone therapy for treatment of pregnancy loss, 243 perhaps because of this mechanism of loss. A single intramuscular injection of 5000 IU of hCG given 1 week after ovulation is an effective treatment for uRPL. We have achieved an overall 80% success rate using this simple method for women with otherwise unexplained RPL (unpublished results).

500

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

3. Stephenson M, Kutteh W. Evaluation and management of recurrent early pregnancy loss. Clin Obstet Gynecol 2007; 50(1):132145 4. Stephenson MD, Awartani KA, Robinson WP. Cytogenetic analysis of miscarriages from couples with recurrent miscarriage: a case-control study. Hum Reprod 2002; 17(2):446451 5. Vercammen EE, DHooghe TM. Endometriosis and recurrent pregnancy loss. Semin Reprod Med 2000; 18(4):363368 6. Donaghay M, Lessey BA. Uterine receptivity: alterations associated with benign gynecological disease. Semin Reprod Med 2007;25(6):461475 7. Brenner RM, West NB. Hormonal regulation of the reproductive tract in female mammals. Annu Rev Physiol 1975;37:273302 8. Strauss JF III, Lessey BA. The structure, function, and evaluation of the female reproductive tract. In: Strauss JF III, Barbieri RL, eds. Reproductive Endocrinology: Physiology, Pathophysiology, and Clinical Management. Philadelphia, PA: Saunders Elsevier; 2009:191233 9. Ekblom P, Ekblom M, Fecker L, et al. Role of mesenchymal nidogen for epithelial morphogenesis in vitro. Development 1994;120(7):20032014 10. Garry R, Hart R, Karthigasu KA, Burke C. A re-appraisal of the morphological changes within the endometrium during menstruation: a hysteroscopic, histological and scanning electron microscopic study. Hum Reprod 2009;24(6):1393 1401 11. Kamei M, Carman CV. New observations on the trafcking and diapedesis of monocytes. Curr Opin Hematol 2010; 17(1):4352 12. Taylor HS. Endometrial cells derived from donor stem cells in bone marrow transplant recipients. JAMA 2004;292(1): 8185 13. Du H, Taylor HS. Contribution of bone marrow-derived stem cells to endometrium and endometriosis. Stem Cells 2007;25(8):20822086 14. Halis G, Arici A. Endometriosis and inammation in infertility. Ann N Y Acad Sci 2004;1034:300315 15. Halme J. Role of peritoneal inammation in endometriosisassociated infertility. Ann N Y Acad Sci 1991;622:266274 16. Bukulmez O, Hardy DB, Carr BR, Word RA, Mendelson CR. Inammatory status inuences aromatase and steroid receptor expression in endometriosis. Endocrinology 2008; 149(3):11901204 17. Laird SM, Tuckerman EM, Cork BA, Linjawi S, Blakemore AI, Li TC. A review of immune cells and molecules in women with recurrent miscarriage. Hum Reprod Update 2003;9(2):163174 18. van Mourik MS, Macklon NS, Heijnen CJ. Embryonic implantation: cytokines, adhesion molecules, and immune cells in establishing an implantation environment. J Leukoc Biol 2009;85(1):419 19. Noyes RW, Hertig AI, Rock J. Dating the endometrial biopsy. Fertil Steril 1950;1:325 20. Jones GE. Some newer aspects of the management of infertility. J Am Med Assoc 1949;141(16):11231129, illust 21. Castelbaum AJ, Lessey BA. Insights into the evaluation of the luteal phase. In: Diamond MP, ed. Infertility and Reproductive Medicine. Philadelphia, PA: Saunders; 1995: 199213

22. Usadi RS, Groll JM, Lessey BA, et al. Endometrial development and function in experimentally induced luteal phase deciency. J Clin Endocrinol Metab 2008;93(10): 40584064 23. Murray MJ, Meyer WR, Zaino RJ, et al. A critical analysis of the accuracy, reproducibility, and clinical utility of histologic endometrial dating in fertile women. Fertil Steril 2004;81(5):13331343 24. Coutifaris C, Myers ER, Guzick DS, et al; NICHD National Cooperative Reproductive Medicine Network. Histological dating of timed endometrial biopsy tissue is not related to fertility status. Fertil Steril 2004;82(5):1264 1272 25. Dey SK, Lim H, Das SK, et al. Molecular cues to implantation. Endocr Rev 2004;25(3):341373 26. Tapia A, Gangi LM, Zegers-Hochschild F, et al. Differences in the endometrial transcript prole during the receptive period between women who were refractory to implantation and those who achieved pregnancy. Hum Reprod 2008;23(2):340351 n A, Albert C, Romero 27. de los Santos MJ, Mercader A, Gala JL, Pellicer A. Implantation rates after two, three, or ve days of embryo culture. Placenta 2003;24(Suppl B):S13S19 28. Pope WF. Uterine asynchrony: a cause of embryonic loss. Biol Reprod 1988;39(5):9991003 29. Wilcox AJ, Baird DD, Weinberg CR. Time of implantation of the conceptus and loss of pregnancy. N Engl J Med 1999;340(23):17961799 30. Enders AC. Current topic: structural responses of the primate endometrium to implantation. Placenta 1991;12(4): 309325 31. Enders AC. Implantation (Embryology). Encyclopedia of Human Biology. Vol 4. New York, NY: Chapman and Hall; 1991:423435 32. Aplin JD. Embryo implantation: the molecular mechanism remains elusive. Reprod Biomed Online 2006;13(6):833 839 33. Aplin JD, Kimber SJ. Trophoblast-uterine interactions at implantation. Reprod Biol Endocrinol 2004;2:48 34. Talbi S, Hamilton AE, Vo KC, et al. Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normoovulatory women. Endocrinology 2006;147(3):10971121 35. Carson DD, Bagchi I, Dey SK, et al. Embryo implantation. Dev Biol 2000;223(2):217237 36. Yoshinaga K, Yoshinaga K, Mori T. Receptor concept in implantation research. In: Yoshinaga K, Mori T, eds. Development of Preimplantation Embryos and Their Environment. New York, NY: Alan Liss; 1989:379387 37. Rogers PAW, Murphy CR, Yoshinaga K. Uterine receptivity for implantation: human studies. In: Yoshinaga K, ed. Blastocyst Implantation. Boston, MA: Serono Symposia USA; 1989:231238 38. Yoshinaga K. Uterine receptivity for blastocyst implantation. Ann N Y Acad Sci 1988;541:424431 39. Sutherland A. Mechanisms of implantation in the mouse: differentiation and functional importance of trophoblast giant cell behavior. Dev Biol 2003;258(2):241251 40. Murphy CR, Rogers PA, Hosie MJ, Leeton J, Beaton L. Tight junctions of human uterine epithelial cells change during the menstrual cycle: a morphometric study. Acta Anat (Basel) 1992;144(1):3638

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

501

41. Rogers PA, Murphy CR. Morphometric and freeze fracture studies of human endometrium during the peri-implantation period. Reprod Fertil Dev 1992;4(3):265269 42. Daya S. Endometriosis and spontaneous abortion. In: Ginsberg KA, ed. Infertility and Reproductive Medicine Clinics of North America. Philadelphia, PA: Saunders; 1996: 759773 43. Strathy JH, Molgaard CA, Coulam CB, Melton LJ III. Endometriosis and infertility: a laparoscopic study of endometriosis among fertile and infertile women. Fertil Steril 1982;38(6):667672 44. Verkauf BS. Incidence, symptoms, and signs of endometriosis in fertile and infertile women. J Fla Med Assoc 1987;74(9):671675 45. Lessey BA, Palomino WA, Apparao KB, Young SL, Lininger RA. Estrogen receptor-alpha (ER-alpha) and defects in uterine receptivity in women. Reprod Biol Endocrinol 2006;4(Suppl 1):S9 46. Young SL, Lessey BA. Progesterone function in human endometrium: clinical perspectives. Semin Reprod Med 2010;28(1):516 47. Jackson KS, Brudney A, Hastings JM, Mavrogianis PA, Kim JJ, Fazleabas AT. The altered distribution of the steroid hormone receptors and the chaperone immunophilin FKBP52 in a baboon model of endometriosis is associated with progesterone resistance during the window of uterine receptivity. Reprod Sci 2007;14(2):137150 48. Kao LC, Germeyer A, Tulac S, et al. Expression proling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology 2003;144(7):28702881 49. Burney RO, Talbi S, Hamilton AE, et al. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology 2007;148(8):38143826 50. Key JD, Kempers RD. Citation classics: most-cited articles from Fertility and Sterility. Fertil Steril 1987;47(6):910915 51. Dor J, Itzkowic DJ, Mashiach S, Lunenfeld B, Serr DM. Cumulative conception rates following gonadotropin therapy. Am J Obstet Gynecol 1980;136(1):102105 52. MacLaughlan SD, Palomino WA, Mo B, Lewis TD, Lininger RA, Lessey BA. Endometrial expression of Cyr61: a marker of estrogenic activity in normal and abnormal endometrium. Obstet Gynecol 2007;110(1):146154 53. Cermik D, Selam B, Taylor HS. Regulation of HOXA-10 expression by testosterone in vitro and in the endometrium of patients with polycystic ovary syndrome. J Clin Endocrinol Metab 2003;88(1):238243 54. Avellaira C, Villavicencio A, Bacallao K, et al. Expression of molecules associated with tissue homeostasis in secretory endometria from untreated women with polycystic ovary syndrome. Hum Reprod 2006;21(12):31163121 55. DuQuesnay R, Wright C, Aziz AA, et al. Infertile women with isolated polycystic ovaries are decient in endometrial expression of osteopontin but not alphavbeta3 integrin during the implantation window. Fertil Steril 2009;91(2): 489499 56. Apparao KB, Lovely LP, Gui Y, Lininger RA, Lessey BA. Elevated endometrial androgen receptor expression in women with polycystic ovarian syndrome. Biol Reprod 2002;66(2):297304 57. Gregory CW, Wilson EM, Apparao KB, et al. Steroid receptor coactivator expression throughout the menstrual

58. 59.

60.

61.

62.

63. 64.

65. 66.

67.

68.

69.

70.

71.

72. 73.

74.

75.

76.

cycle in normal and abnormal endometrium. J Clin Endocrinol Metab 2002;87(6):29602966 Baulieu EE. RU486: a compound that gets itself talked about. Hum Reprod 1994;9(Suppl 1):16 LH, Scott JZ, Williams KP. The Cumming DC, Honore late luteal phase in infertile women: comparison of simultaneous endometrial biopsy and progesterone levels. Fertil Steril 1985;43(5):715719 Daya S, Ward S, Burrows E. Progesterone proles in luteal phase defect cycles and outcome of progesterone treatment in patients with recurrent spontaneous abortion. Am J Obstet Gynecol 1988;158(2):225232 Hecht BR, Bardawil WA, Khan-Dawood FS, Dawood MY. Luteal insufciency: correlation between endometrial dating and integrated progesterone output in clomiphene citrateinduced cycles. Am J Obstet Gynecol 1990;163(6 Pt 1):19861991 Fritz MA, Lessey BA. Defective luteal function. In: Fraser IS, Jansen RPS, Lobo RA, Whitehead MI, eds. Estrogens and progestogens in clinical practice. London, UK: Churchill Livingstone; 1998:437453 Strott CA, Cargille CM, Ross GT, Lipsett MB. The short luteal phase. J Clin Endocrinol Metab 1970;30(2):246251 Groff TR, Raj HG, Talbert LM, Willis DL. Effects of neutralization of luteinizing hormone on corpus luteum function and cyclicity in Macaca fascicularis.. J Clin Endocrinol Metab 1984;59(6):10541057 Filicori M, Flamigni C. GnRH agonists and antagonists. Current clinical status. Drugs 1988;35(1):6382 Ellinwood WE, Norman RL, Spies HG. Changing frequency of pulsatile luteinizing hormone and progesterone secretion during the luteal phase of the menstrual cycle of rhesus monkeys. Biol Reprod 1984;31(4):714722 Lessey BA, Castelbaum AJ, Buck CA, Lei Y, Yowell CW, Sun J. Further characterization of endometrial integrins during the menstrual cycle and in pregnancy. Fertil Steril 1994;62(3):497506 Baulieu EE. Contragestion and other clinical applications of RU 486, an antiprogesterone at the receptor. Science 1989;245(4924):13511357 Good RG, Moyer DL. Estrogen-progesterone relationships in the development of secretory endometrium. Fertil Steril 1968;19(1):3749 Joshi SG, Ebert KM, Smith RA. Properties of the progestagen-dependent protein of the human endometrium. J Reprod Fertil 1980;59(2):287296 Strinden ST, Shapiro SS. Progesterone-altered secretory proteins from cultured human endometrium. Endocrinology 1983;112(3):862870 Navot D, Bergh P. Preparation of the human endometrium for implantation. Ann N Y Acad Sci 1991;622:212219 Ilesanmi AO, Hawkins DA, Lessey BA. Immunohistochemical markers of uterine receptivity in the human endometrium. Microsc Res Tech 1993;25(3):208222 Ding YQ, Zhu LJ, Bagchi MK, Bagchi IC. Progesterone stimulates calcitonin gene expression in the uterus during implantation. Endocrinology 1994;135(5):22652274 Lessey BA, Yeh I, Castelbaum AJ, et al. Endometrial progesterone receptors and markers of uterine receptivity in the window of implantation. Fertil Steril 1996;65(3):477483 Aplin JD, Jones CJP, McGinlay PB, Croxatto HB, Fazleabas AT. Progesterone regulates glycosylation in endometrium. Biochem Soc Trans 1997;25(4):11841187

Downloaded by: World Health Organization ( WHO). Copyrighted material.

502

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

77. Lalitkumar PGL, Sengupta J, Karande AA, Ghosh D. Placental protein 14 in endometrium during menstrual cycle and effect of early luteal phase mifepristone administration on its expression in implantation stage endometrium in the rhesus monkey. Hum Reprod 1998;13(12):34783486 78. Lessey BA. Endometrial integrins and the establishment of uterine receptivity. Hum Reprod 1998;13(Suppl 3):247 258; discussion 259261 79. Giudice LC. Genes associated with embryonic attachment and implantation and the role of progesterone. J Reprod Med 1999;44(2, Suppl):165171 nguez F, Remoh J, Pellicer A, Simo n C. Human 80. Dom endometrial receptivity: a genomic approach. Reprod Biomed Online 2003;6(3):332338 81. Lessey BA. Two pathways of progesterone action in the human endometrium: implications for implantation and contraception. Steroids 2003;68(1013):809815 82. Bulun SE, Cheng YH, Yin P, et al. Progesterone resistance in endometriosis: link to failure to metabolize estradiol. Mol Cell Endocrinol 2006;248(12):94103 83. Kitawaki J, Kusuki I, Koshiba H, Tsukamoto K, Fushiki S, Honjo H. Detection of aromatase cytochrome P-450 in endometrial biopsy specimens as a diagnostic test for endometriosis. Fertil Steril 1999;72(6):11001106 la M, Li TC, Warren 84. Klentzeris LD, Bulmer JN, Seppa MA, Cooke ID. Placental protein 14 in cycles with normal and retarded endometrial differentiation. Hum Reprod 1994;9(3):394398 85. Young SL, Lessey BA, Balthazar U, et al. Dening the relationship between progesterone dose, endometrial histology and gene expression using an in vivo luteal phase defect model. Reprod Sci 2011;18:272A 86. Enders AC, Nelson DM. Pinocytotic activity of the uterus of the rat. Am J Anat 1973;138(3):277299 87. Psychoyos A, Mandon P. [Study of the surface of the uterine epithelium by scanning electron microscope. Observations in the rat at the 4th and 5th day of pregnancy [in French]. CR Acad Sci Hebd Seances Acad Sci D 1971;272:27232725 88. Ciocca DR, Asch RH, Adams DJ, McGuire WL. Evidence for modulation of a 24K protein in human endometrium during the menstrual cycle. J Clin Endocrinol Metab 1983;57(3):496499 89. Psychoyos A, Martel D. Embryo-endometrial interactions at implantation. J Reprod Fertil 1985;41:195209 90. Martel D, Frydman R, Glissant M, Maggioni C, Roche D, Psychoyos A. Scanning electron microscopy of postovulatory human endometrium in spontaneous cycles and cycles stimulated by hormone treatment. J Endocrinol 1987; 114(2):319324 91. Martel D, Frydman R, Glissant M, Maggioni C, Roche D, Psychoyos A. Scanning electron microscopy of postovulatory human endometrium in spontaneous cycles and cycles stimulated by hormone treatment. J Endocrinol 1987; 114(2):319324 92. Martel D, Frydman R, Sarantis L, et al. Scanning electron microscopy of the uterine luminal epithelium as a marker of the implantation window. In: Yoshinaga K, ed. Blastocyst Implantation. Boston, MA: Adams Publishing Group; 1993:225 93. Psychoyos A, Nikas G. Uterine pinopodes as markers of uterine receptivity. Assist Reprod Rev 1994;4:2632 94. Nikas G, Drakakis P, Loutradis D, et al. Uterine pinopodes as markers of the nidation window in cycling women

95.

96.

97.

98. 99. 100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

111.

receiving exogenous oestradiol and progesterone. Hum Reprod 1995;10(5):12081213 Bentin-Ley U. Relevance of endometrial pinopodes for human blastocyst implantation. Hum Reprod 2000;15(Suppl 6):6773 Bulletti C, Flamigni C, de Ziegler D. Implantation markers and endometriosis. Reprod Biomed Online 2005;11(4):464 468 Nardo LG, Nikas G, Makrigiannakis A, Sinatra F, Nardo F. Synchronous expression of pinopodes and alpha v beta 3 and alpha 4 beta 1 integrins in the endometrial surface epithelium of normally menstruating women during the implantation window. J Reprod Med 2003;48(5):355361 Nikas G. Cell-surface morphological events relevant to human implantation. Hum Reprod 1999;14(Suppl 2):3744 Nikas G. Pinopodes as markers of endometrial receptivity in clinical practice. Hum Reprod 1999;14(Suppl 2):99106 Nikas G, Psychoyos A. Uterine pinopodes in periimplantation human endometrium. Clinical relevance. Ann N Y Acad Sci 1997;816:129142 Stavreus-Evers A, Nikas G, Sahlin L, Eriksson H, Landgren BM. Formation of pinopodes in human endometrium is associated with the concentrations of progesterone and progesterone receptors. Fertil Steril 2001;76(4):782791 Martel D, Monier MN, Roche D, Psychoyos A. Hormonal dependence of pinopode formation at the uterine luminal surface. Hum Reprod 1991;6(4):597603 Adams SM, Gayer N, Hosie MJ, Murphy CR. Human uterodomes (pinopods) do not display pinocytotic function. Hum Reprod 2002;17(8):19801986 Bentin-Ley U, Pedersen B, Lindenberg S, Larsen JF, Hamberger L, Horn T. Isolation and culture of human endometrial cells in a three-dimensional culture system. J Reprod Fertil 1994;101(2):327332 gren A, Nilsson L, Hamberger L, Larsen Bentin-Ley U, Sjo JF, Horn T. Presence of uterine pinopodes at the embryoendometrial interface during human implantation in vitro. Hum Reprod 1999;14(2):515520 von Wolff M, Strowitzki T, Becker V, Zepf C, Tabibzadeh S, Thaler CJ. Endometrial osteopontin, a ligand of b3-integrin, is maximally expressed around the time of the implantation window. Fertil Steril 2001;76(4):775781 Young SL, Lessey BA, Fritz MA, et al. In vivo and in vitro evidence suggest that HB-EGF regulates endometrial expression of human decay-accelerating factor. J Clin Endocrinol Metab 2002;87(3):13681375 Aghajanova L, Stavreus-Evers A, Nikas Y, Hovatta O, Landgren BM. Coexpression of pinopodes and leukemia inhibitory factor, as well as its receptor, in human endometrium. Fertil Steril 2003;79(Suppl 1):808814 Lai TH, Shih IeM, Vlahos N, Ho CL, Wallach E, Zhao Y. Differential expression of L-selectin ligand in the endometrium during the menstrual cycle. Fertil Steril 2005;83(Suppl 1):12971302 Stavreus-Evers A, Mandelin E, Koistinen R, Aghajanova L, la M. Glycodelin is present in pinopodes Hovatta O, Seppa of receptive-phase human endometrium and is associated with down-regulation of progesterone receptor B. Fertil Steril 2006;85(6):18031811 Foulk RA, Zdravkovic T, Genbacev O, Prakobphol A. Expression of L-selectin ligand MECA-79 as a predictive marker of human uterine receptivity. J Assist Reprod Genet 2007;24(7):316321

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

503

112. Shimizu Y, Kabir-Salmani M, Azadbakht M, Sugihara K, Sakai K, Iwashita M. Expression and localization of galectin9 in the human uterodome. Endocr J 2008;55(5):879887 113. Acosta AA, Elberger L, Borghi M, et al. Endometrial dating and determination of the window of implantation in healthy fertile women. Fertil Steril 2000;73(4):788798 114. Usadi RS, Murray MJ, Bagnell RC, et al. Temporal and morphologic characteristics of pinopod expression across the secretory phase of the endometrial cycle in normally cycling women with proven fertility. Fertil Steril 2003;79(4):970974 115. Quinn C, Ryan E, Claessens EA, et al. The presence of pinopodes in the human endometrium does not delineate the implantation window. Fertil Steril 2007;87(5):1015 1021 zar JL. Three-dimensional ultrasound assessment of 116. Alca endometrial receptivity: a review. Reprod Biol Endocrinol 2006;4:56 117. Dechaud H, Bessueille E, Bousquet PJ, Reyftmann L, Hamamah S, Hedon B. Optimal timing of ultrasonographic and Doppler evaluation of uterine receptivity to implantation. Reprod Biomed Online 2008;16(3):368375 118. Smith P. Evaluation of endometrium with transvaginal ultrasound. Acta Obstet Gynecol Scand 1993;72(8):686687 119. Van Voorhis BJ. Ultrasound assessment of the uterus and fallopian tube in infertile women. Semin Reprod Med 2008; 26(3):232240 120. Abdalla HI, Brooks AA, Johnson MR, Kirkland A, Thomas A, Studd JW. Endometrial thickness: a predictor of implantation in ovum recipients? Hum Reprod 1994;9(2): 363365 121. El-Toukhy T, Coomarasamy A, Khairy M, et al. The relationship between endometrial thickness and outcome of medicated frozen embryo replacement cycles. Fertil Steril 2008;89(4):832839 122. Isaacs JD Jr, Wells CS, Williams DB, Odem RR, Gast MJ, Strickler RC. Endometrial thickness is a valid monitoring parameter in cycles of ovulation induction with menotropins alone. Fertil Steril 1996;65(2):262266 123. Oliveira JBA, Baruf RLR, Mauri AL, Petersen CG, Borges MC, Franco JG Jr. Endometrial ultrasonography as a predictor of pregnancy in an in-vitro fertilization programme after ovarian stimulation and gonadotrophinreleasing hormone and gonadotrophins. Hum Reprod 1997; 12(11):25152518 124. Rabinowitz R, Laufer N, Lewin A, et al. The value of ultrasonographic endometrial measurement in the prediction of pregnancy following in vitro fertilization. Fertil Steril 1986;45(6):824828 125. Bassil S. Changes in endometrial thickness, width, length and pattern in predicting pregnancy outcome during ovarian stimulation in in vitro fertilization. Ultrasound Obstet Gynecol 2001;18(3):258263 126. Coulam CB, Bustillo M, Soenksen DM, Britten S. Ultrasonographic predictors of implantation after assisted reproduction. Fertil Steril 1994;62(5):10041010 127. De Geyter C, Schmitter M, De Geyter M, Nieschlag E, Holzgreve W, Schneider HP. Prospective evaluation of the ultrasound appearance of the endometrium in a cohort of 1,186 infertile women. Fertil Steril 2000;73(1):106113 128. Glissant A, de Mouzon J, Frydman R. Ultrasound study of the endometrium during in vitro fertilization cycles. Fertil Steril 1985;44(6):786790

129. Gonen Y, Casper RF, Jacobson W, Blankier J. Endometrial thickness and growth during ovarian stimulation: a possible predictor of implantation in in vitro fertilization. Fertil Steril 1989;52(3):446450 130. Noyes N, Liu HC, Sultan K, Schattman G, Rosenwaks Z. Endometrial thickness appears to be a signicant factor in embryo implantation in in-vitro fertilization. Hum Reprod 1995;10(4):919922 131. Imoedemhe DA, Shaw RW, Kirkland A, Chan R. Ultrasound measurement of endometrial thickness on different ovarian stimulation regimens during in-vitro fertilization. Hum Reprod 1987;2:545547 132. Friedler S, Schenker JG, Herman A, Lewin A. The role of ultrasonography in the evaluation of endometrial receptivity following assisted reproductive treatments: a critical review. Hum Reprod Update 1996;2(4):323335 133. Sterzik K, Grab D, Schneider V, Strehler EJ, Gagsteiger F, Rosenbusch BE. Lack of correlation between ultrasonography and histologic staging of the endometrium in in vitro fertilization (IVF) patients. Ultrasound Med Biol 1997; 23(2):165170 134. Scholtes MC, Wladimiroff JW, van Rijen HJ, Hop WC. Uterine and ovarian ow velocity waveforms in the normal menstrual cycle: a transvaginal Doppler study. Fertil Steril 1989;52(6):981985 135. Goswamy RK, Steptoe PC. Doppler ultrasound studies of the uterine artery in spontaneous ovarian cycles. Hum Reprod 1988;3(6):721726 136. Habara T, Nakatsuka M, Konishi H, Asagiri K, Noguchi S, Kudo T. Elevated blood ow resistance in uterine arteries of women with unexplained recurrent pregnancy loss. Hum Reprod 2002;17(1):190194 137. Sterzik K, Grab D, Sasse V, Hu tter W, Rosenbusch B, Terinde R. Doppler sonographic ndings and their correlation with implantation in an in vitro fertilization program. Fertil Steril 1989;52(5):825828 138. Steer CV, Tan SL, Mason BA, Campbell S. Midlutealphase vaginal color Doppler assessment of uterine artery impedance in a subfertile population. Fertil Steril 1994; 61(1):5358 139. Yaron Y, Botchan A, Amit A, Peyser MR, David MP, Lessing JB. Endometrial receptivity in the light of modern assisted reproductive technologies. Fertil Steril 1994;62(2): 225232 140. Cacciatore B, Simberg N, Fusaro P, Tiitinen A. Transvaginal Doppler study of uterine artery blood ow in in vitro fertilization-embryo transfer cycles. Fertil Steril 1996; 66(1):130134 141. Steer CV, Tan SL, Dillon D, Mason BA, Campbell S. Vaginal color Doppler assessment of uterine artery impedance correlates with immunohistochemical markers of endometrial receptivity required for the implantation of an embryo. Fertil Steril 1995;63(1):101108 142. Schild RL, Indefrei D, Eschweiler S, Van der Ven H, Fimmers R, Hansmann M. Three-dimensional endometrial volume calculation and pregnancy rate in an in-vitro fertilization programme. Hum Reprod 1999;14(5):1255 1258 143. Alatas C, Aksoy E, Akarsu C, Yakin K, Aksoy S, Hayran M. Evaluation of intrauterine abnormalities in infertile patients by sonohysterography. Hum Reprod 1997;12(3): 487490

Downloaded by: World Health Organization ( WHO). Copyrighted material.

504

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

144. Goldstein SR. Use of ultrasonohysterography for triage of perimenopausal patients with unexplained uterine bleeding. Am J Obstet Gynecol 1994;170(2):565570 145. Keltz MD, Olive DL, Kim AH, Arici A. Sonohysterography for screening in recurrent pregnancy loss. Fertil Steril 1997;67(4):670674 146. Parsons AK, Lense JJ. Sonohysterography for endometrial abnormalities: preliminary results. J Clin Ultrasound 1993; 21(2):8795 147. Burke C, Kelehan P, Wingeld M. Unsuspected endometrial pathology in the subfertile woman. Ir Med J 2007; 100(5):466469 148. Goldberg JM, Falcone T, Attaran M. Sonohysterographic evaluation of uterine abnormalities noted on hysterosalpingography. Hum Reprod 1997;12(10):21512153 149. Hamilton JA, Larson AJ, Lower AM, Hasnain S, Grudzinskas JG. Routine use of saline hysterosonography in 500 consecutive, unselected, infertile women. Hum Reprod 1998;13(9):24632473 150. Lessey BA, Castelbaum AJ, Sawin SW, et al. Aberrant integrin expression in the endometrium of women with endometriosis. J Clin Endocrinol Metab 1994;79(2):643649 151. Achache H, Revel A. Endometrial receptivity markers, the journey to successful embryo implantation. Hum Reprod Update 2006;12(6):731746 152. Getzenberg RH, Pienta KJ, Coffey DS. The tissue matrix: cell dynamics and hormone action. Endocr Rev 1990;11(3): 399417 153. Aplin JD, Charlton AK, Ayad S. An immunohistochemical study of human endometrial extracellular matrix during the menstrual cycle and rst trimester of pregnancy. Cell Tissue Res 1988;253(1):231240 154. Ruck P, Marzusch K, Kaiserling E, et al. Distribution of cell adhesion molecules in decidua of early human pregnancy. An immunohistochemical study. Lab Invest 1994;71(1):94101 155. Feinberg RF, Kliman HJ. Tropho-uteronectin (TUN): A unique oncofetal bronectin deposited in the extracellular matrix of the tropho-uterine junction and regulated in vitro by cultured human trophoblast cells. Trophblast Res 1993; 7:167173 156. Turpeenniemi-Hujanen T, Feinberg RF, Kauppila A, Puistola U. Extracellular matrix interactions in early human embryos: implications for normal implantation events. Fertil Steril 1995;64(1):132138 157. Albelda SM, Buck CA. Integrins and other cell adhesion molecules. FASEB J 1990;4(11):28682880 158. Lessey BA, Damjanovich L, Coutifaris C, Castelbaum A, Albelda SM, Buck CA. Integrin adhesion molecules in the human endometrium. Correlation with the normal and abnormal menstrual cycle. J Clin Invest 1992;90(1):188195 159. Tabibzadeh S. Patterns of expression of integrin molecules in human endometrium throughout the menstrual cycle. Hum Reprod 1992;7(6):876882 160. Damsky C, Sutherland A, Fisher S. Extracellular matrix 5: adhesive interactions in early mammalian embryogenesis, implantation, and placentation. FASEB J 1993;7(14):1320 1329 161. Armant DR, Kaplan HA, Mover H, Lennarz WJ. The effect of hexapeptides on attachment and outgrowth of mouse blastocysts cultured in vitro: evidence for the involvement of the cell recognition tripeptide Arg-GlyAsp. Proc Natl Acad Sci U S A 1986;83(18):6751 6755

162. Illera MJ, Cullinan E, Gui Y, Yuan L, Beyler SA, Lessey BA. Blockade of the alpha(v)beta(3) integrin adversely affects implantation in the mouse. Biol Reprod 2000;62(5):1285 1290 163. Illera MJ, Lorenzo PL, Gui YT, Beyler SA, Apparao KB, Lessey BA. A role for alphavbeta3 integrin during implantation in the rabbit model. Biol Reprod 2003;68:766771 164. Bronson RA, Fusi F. Evidence that an Arg-Gly-Asp adhesion sequence plays a role in mammalian fertilization. Biol Reprod 1990;43(6):10191025 165. Bronson RA, Fusi FM. Integrins and human reproduction. Mol Hum Reprod 1996;2(3):153168 166. Yelian FD, Yang Y, Hirata JD, Schultz JF, Armant DR. Molecular interactions between bronectin and integrins during mouse blastocyst outgrowth. Mol Reprod Dev 1995; 41(4):435448 167. Jones JI, Gockerman A, Busby WH Jr, Wright G, Clemmons DR. Insulin-like growth factor binding protein 1 stimulates cell migration and binds to the alpha 5 beta 1 integrin by means of its Arg-Gly-Asp sequence. Proc Natl Acad Sci U S A 1993;90(22):1055310557 168. Illera MJ, Lorenzo PL, Gui YT, Beyler SA, Apparao KB, Lessey BA. A role for alphavbeta3 integrin during implantation in the rabbit model. Biol Reprod 2003;68(3):766771 169. Sutherland AE, Calarco PG, Damsky CH. Developmental regulation of integrin expression at the time of implantation in the mouse embryo. Development 1993;119(4):11751186 170. Tabibzadeh S, Sun XZ. Cytokine expression in human endometrium throughout the menstrual cycle. Hum Reprod 1992;7(9):12141221 171. Daftary GS, Troy PJ, Bagot CN, Young SL, Taylor HS. Direct regulation of beta3-integrin subunit gene expression by HOXA10 in endometrial cells. Mol Endocrinol 2002; 16(3):571579 172. Matsuzaki S, Canis M, Darcha C, Pouly JL, Mage G. HOXA-10 expression in the mid-secretory endometrium of infertile patients with either endometriosis, uterine bromas or unexplained infertility. Hum Reprod 2009;24(12):3180 3187 173. Tuckerman EM, Laird SM, Prakash A, Li TC. Expression of integrins in the endometrium of women with recurrent miscarriage. Fertil Steril 2006;86(3):755757 174. Quenby S, Anim-Somuah M, Kalumbi C, Farquharson R, Aplin JD. Different types of recurrent miscarriage are associated with varying patterns of adhesion molecule expression in endometrium. Reprod Biomed Online 2007; 14(2):224234 175. Ivanov PD, Komsa-Penkova RS, Konova EI, et al. Polymorphism A1/A2 in the cell surface integrin subunit b3 and disturbance of implantation and placentation in women with recurrent pregnancy loss. Fertil Steril 2010; 94(7):28432845 176. Fukuda MN, Sato T, Nakayama J, et al. Trophinin and tastin, a novel cell adhesion molecule complex with potential involvement in embryo implantation. Genes Dev 1995; 9(10):11991210 177. Paria BC, Zhao XM, Das SK, Dey SK, Yoshinaga K. Zonula occludens-1 and E-cadherin are coordinately expressed in the mouse uterus with the initiation of implantation and decidualization. Dev Biol 1999;208(2):488501 178. Apparao KB, Murray MJ, Fritz MA, et al. Osteopontin and its receptor alphavbeta(3) integrin are coexpressed in the human endometrium during the menstrual cycle but

Downloaded by: World Health Organization ( WHO). Copyrighted material.

THE ENDOMETRIUM IN RECURRENT EARLY PREGNANCY LOSS/PATEL, LESSEY

505

179.

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

193.

regulated differentially. J Clin Endocrinol Metab 2001; 86(10):49915000 Chobotova K, Spyropoulou I, Carver J, et al. Heparinbinding epidermal growth factor and its receptor ErbB4 mediate implantation of the human blastocyst. Mech Dev 2002;119(2):137144 Genbacev OD, Prakobphol A, Foulk RA, et al. Trophoblast L-selectin-mediated adhesion at the maternal-fetal interface. Science 2003;299(5605):405408 Lai TH, Zhao Y, Shih IeM, Ho CL, Bankowski B, Vlahos N. Expression of L-selectin ligands in human endometrium during the implantation window after controlled ovarian stimulation for oocyte donation. Fertil Steril 2006;85(3): 761763 Bhatt H, Brunet LJ, Stewart CL. Uterine expression of leukemia inhibitory factor coincides with the onset of blastocyst implantation. Proc Natl Acad Sci U S A 1991; 88(24):1140811412 Stewart CL. Leukaemia inhibitory factor and the regulation of pre-implantation development of the mammalian embryo. Mol Reprod Dev 1994;39(2):233238 Stewart CL, Kaspar P, Brunet LJ, et al. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature 1992;359(6390):7679 Cullinan EB, Abbondanzo SJ, Anderson PS, Pollard JW, Lessey BA, Stewart CL. Leukemia inhibitory factor (LIF) and LIF receptor expression in human endometrium suggests a potential autocrine/paracrine function in regulating embryo implantation. Proc Natl Acad Sci U S A 1996; 93(7):31153120 Sherwin JR, Smith SK, Wilson A, Sharkey AM. Soluble gp130 is up-regulated in the implantation window and shows altered secretion in patients with primary unexplained infertility. J Clin Endocrinol Metab 2002;87(8):39533960 Mikolajczyk M, Wirstlein P, Skrzypczak J. Leukaemia inhibitory factor and interleukin 11 levels in uterine ushings of infertile patients with endometriosis. Hum Reprod 2006; 21(12):30543058 Dimitriadis E, Sharkey AM, Tan YL, Salamonsen LA, Sherwin JR. Immunolocalisation of phosphorylated STAT3, interleukin 11 and leukaemia inhibitory factor in endometrium of women with unexplained infertility during the implantation window. Reprod Biol Endocrinol 2007;5(1):44 e S, Bjuresten K, Hovatta O, Aghajanova L, Altma Landgren BM, Stavreus-Evers A. Disturbances in the LIF pathway in the endometrium among women with unexplained infertility. Fertil Steril 2009;91(6):26022610 Savaris RF, Groll JM, Young SL, et al. Progesterone resistance in PCOS endometrium: a microarray analysis in clomiphene citrate-treated and articial menstrual cycles. J Clin Endocrinol Metab 2011;96(6):17371746 Carson DD, Lagow E, Thathiah A, et al. Changes in gene expression during the early to mid-luteal (receptive phase) transition in human endometrium detected by high-density microarray screening. Mol Hum Reprod 2002;8(9): 871879 Catalano RD, Critchley HO, Heikinheimo O, et al. Mifepristone induced progesterone withdrawal reveals novel regulatory pathways in human endometrium. Mol Hum Reprod 2007;13(9):641654 Eyster KM, Boles AL, Brannian JD, Hansen KA. DNA microarray analysis of gene expression markers of endometriosis. Fertil Steril 2002;77(1):3842

194. Haouzi D, Mahmoud K, Fourar M, et al. Identication of new biomarkers of human endometrial receptivity in the natural cycle. Hum Reprod 2009;24(1):198 205 n C. Wide genomic analysis 195. Horcajadas JA, Pellicer A, Simo of human endometrial receptivity: new times, new opportunities. Hum Reprod Update 2007;13(1):7786 196. Kao LC, Yang J, Lessey BA, Giudice LC. Microarray expression proling reveals candidate genes for human uterine receptivity. Fertil Steril 2001;76:S59 197. Kao LC, Germeyer A, Tulac S, et al. Expression proling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology 2003;144:28702881 198. Kao LC, Tulac S, Lobo S, et al. Global gene proling in human endometrium during the window of implantation. Endocrinology 2002;143:21192138 199. Liu Y, Lee KF, Ng EH, Yeung WS, Ho PC. Gene expression proling of human peri-implantation endometria between natural and stimulated cycles. Fertil Steril 2008; 90(6):21522164 200. Matsuzaki S, Canis M, Pouly JL, Botchorishvili R, chelotte PJ, Mage G. Differential expression of genes De in eutopic and ectopic endometrium from patients with ovarian endometriosis. Fertil Steril 2006;86(3):548 553 ` re C, Boespug201. Matsuzaki S, Canis M, Vaurs-Barrie Tanguy O, Dastugue B, Mage G. DNA microarray analysis of gene expression in eutopic endometrium from patients with deep endometriosis using laser capture microdissection. Fertil Steril 2005;84(Suppl 2):11801190 202. Mettler L, Salmassi A, Schollmeyer T, Schmutzler AG, Pu ngel F, Jonat W. Comparison of c-DNA microarray analysis of gene expression between eutopic endometrium and ectopic endometrium (endometriosis). J Assist Reprod Genet 2007;24(6):249258 n J, van Os R, et al. Gene expression 203. Riesewijk A, Mart proling of human endometrial receptivity on days LH 2 versus LH 7 by microarray technology. Mol Hum Reprod 2003;9(5):253264 204. Schmidt A, Groth P, Haendler B, et al. Gene expression during the implantation window: microarray analysis of human endometrial samples. Ernst Schering Res Found Workshop 2005;(52):139157 205. Wu Y, Kajdacsy-Balla A, Strawn E, et al. Transcriptional characterizations of differences between eutopic and ectopic endometrium. Endocrinology 2006;147(1):232246 206. Aghajanova L, Hamilton AE, Giudice LC. Uterine receptivity to human embryonic implantation: histology, biomarkers, and transcriptomics. Semin Cell Dev Biol 2008; 19(2):204211 207. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116(2):281297 208. Cai X, Hagedorn CH, Cullen BR. Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs. RNA 2004;10(12):1957 1966 209. Cullen BR. Derivation and function of small interfering RNAs and microRNAs. Virus Res 2004;102(1):39 210. Burney RO, Hamilton AE, Aghajanova L, et al. MicroRNA expression proling of eutopic secretory endometrium in women with versus without endometriosis. Mol Hum Reprod 2009;15(10):625631

Downloaded by: World Health Organization ( WHO). Copyrighted material.

506

SEMINARS IN REPRODUCTIVE MEDICINE/VOLUME 29, NUMBER 6

2011

211. Toloubeydokhti T, Pan Q, Luo X, Bukulmez O, Chegini N. The expression and ovarian steroid regulation of endometrial micro-RNAs. Reprod Sci 2008;15(10):993 1001 212. Pan XY, Li X, Weng ZP, Wang B. Altered expression of claudin-3 and claudin-4 in ectopic endometrium of women with endometriosis. Fertil Steril 2009;91(5):16921699 213. Pan Q, Luo X, Toloubeydokhti T, Chegini N. The expression prole of micro-RNA in endometrium and endometriosis and the inuence of ovarian steroids on their expression. Mol Hum Reprod 2007;13(11):797806 214. Chung TK, Cheung TH, Huen NY, et al. Dysregulated microRNAs and their predicted targets associated with endometrioid endometrial adenocarcinoma in Hong Kong women. Int J Cancer 2009;124(6):13581365 215. Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol 2004; 5(3):266271 216. 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(5):517535 217. Jasper MJ, Tremellen KP, Robertson SA. Primary unexplained infertility is associated with reduced expression of the T-regulatory cell transcription factor Foxp3 in endometrial tissue. Mol Hum Reprod 2006;12(5): 301308 218. Robertson SA, Guerin LR, Moldenhauer LM, Hayball JD. Activating T regulatory cells for tolerance in early pregnancythe contribution of seminal uid. J Reprod Immunol 2009;83(1-2):109116 219. Saito S, Sasaki Y, Sakai M. CD4( )CD25high regulatory T cells in human pregnancy. J Reprod Immunol 2005;65(2): 111120 220. Zenclussen AC. CD4( )CD25 T regulatory cells in murine pregnancy. J Reprod Immunol 2005;65(2):101110 221. Quenby S, Farquharson R. Uterine natural killer cells, implantation failure and recurrent miscarriage. Reprod Biomed Online 2006;13(1):2428 222. Quenby S, Nik H, Innes B, et al. Uterine natural killer cells and angiogenesis in recurrent reproductive failure. Hum Reprod 2009;24(1):4554 223. Lachapelle MH, Miron P, Hemmings R, Roy DC. Endometrial T, B, and NK cells in patients with recurrent spontaneous abortion. Altered prole and pregnancy outcome. J Immunol 1996;156(10):40274034 224. Tuckerman E, Laird SM, Stewart R, Wells M, Li TC. Markers of endometrial function in women with unexplained recurrent pregnancy loss: a comparison between morphologically normal and retarded endometrium. Hum Reprod 2004;19(1):196205 225. Fukui A, Fujii S, Yamaguchi E, Kimura H, Sato S, Saito Y. Natural killer cell subpopulations and cytotoxicity for infertile patients undergoing in vitro fertilization. Am J Reprod Immunol 1999;41(6):413422 226. Jasper MJ, Care AS, Sullivan B, Ingman WV, Aplin JD, Robertson SA. Macrophage-derived LIF and IL1B regulate alpha(1,2)fucosyltransferase 2 (Fut2) expression in mouse uterine epithelial cells during early pregnancy. Biol Reprod 2011;84(1):179188 227. Quenby S, Tang AW. Uterine natural killer cells and reproduction. In: Farquharson RG, Stephenson MD, eds. Early Pregnancy. Cambridge, UK: Cambridge Press; 2010: 7584

228. Homburg R, Pap H, Brandes M, Huirne J, Hompes P, Lambalk CB. Endometrial biopsy during induction of ovulation with clomiphene citrate in polycystic ovary syndrome. Gynecol Endocrinol 2006;22(9):506510 229. Legro RS. Pregnancy considerations in women with polycystic ovary syndrome. Clin Obstet Gynecol 2007;50(1):295 304 230. Palomba S, Falbo A, Orio F Jr, Russo T, Tolino A, Zullo F. Metformin hydrochloride and recurrent miscarriage in a woman with polycystic ovary syndrome. Fertil Steril 2006; 85(5):1511, e3e5 231. Mitwally MF, Casper RF. Aromatase inhibition reduces the dose of gonadotropin required for controlled ovarian hyperstimulation. J Soc Gynecol Investig 2004;11(6):406 415 232. Bouchard P. Treatment of infertility in women with polycystic ovary syndrome. Ann Endocrinol (Paris) 2010; 71(3):225227 233. Ott J, Kurz C, Nouri K, et al. Pregnancy outcome in women with polycystic ovary syndrome comparing the effects of laparoscopic ovarian drilling and clomiphene citrate stimulation in women pre-treated with metformin: a retrospective study. Reprod Biol Endocrinol 2010;8:45 234. Tang T, Lord JM, Norman RJ, Yasmin E, Balen AH. Insulin-sensitising drugs (metformin, rosiglitazone, pioglitazone, D-chiro-inositol) for women with polycystic ovary syndrome, oligo amenorrhoea and subfertility. Cochrane Database Syst Rev 2010;(1):CD003053 235. Begum MR, Ferdous J, Begum A, Quadir E. Comparison of efcacy of aromatase inhibitor and clomiphene citrate in induction of ovulation in polycystic ovarian syndrome. Fertil Steril 2009;92(3):853857 236. Abu Hashim H, Mashaly AM, Badawy A. Letrozole versus laparoscopic ovarian diathermy for ovulation induction in clomiphene-resistant women with polycystic ovary syndrome: a randomized controlled trial. Arch Gynecol Obstet 2010;282(5):567571 237. Polyzos NP, Tzioras S, Badawy AM, Valachis A, Dritsas C, Mauri D. Aromatase inhibitors for female infertility: a systematic review of the literature. Reprod Biomed Online 2009;19(4):456471 238. Vitonis AF, Hankinson SE, Hornstein MD, Missmer SA. Adult physical activity and endometriosis risk. Epidemiology 2010;21(1):1623 239. Parazzini F, Ardovino I, Struzziero E, et al. Risk factors for pelvic endometriosis in women with pelvic pain or infertility. Gruppo Italiano per lo Studio dell endometriosi. Eur J Obstet Gynecol Reprod Biol 1999;83(2):195199 240. Tomassetti C, Meuleman C, Pexsters A, et al. Endometriosis, recurrent miscarriage and implantation failure: is there an immunological link? Reprod Biomed Online 2006;13(1):5864 241. Damewood MD. The association of endometriosis and repetitive (early) spontaneous abortions. Semin Reprod Endocrinol 1989;7(2):155160 242. Lessey BA. Medical management of endometriosis and infertility. Fertil Steril 2000;73(6):10891096 243. Daya S, Gunby J. Luteal phase support in assisted reproduction cycles. Cochrane Database Syst Rev 2004; (3):CD004830 244. Munro SK, Farquhar CM, Mitchell MD, Ponnampalam AP. Epigenetic regulation of endometrium during the menstrual cycle. Mol Hum Reprod 2010;16(5):297310

Downloaded by: World Health Organization ( WHO). Copyrighted material.

You might also like